WO2023284733A1 - GITR/TGF-β DUAL-TARGETED FUSION PROTEIN AND USE THEREOF - Google Patents

GITR/TGF-β DUAL-TARGETED FUSION PROTEIN AND USE THEREOF Download PDF

Info

Publication number
WO2023284733A1
WO2023284733A1 PCT/CN2022/105175 CN2022105175W WO2023284733A1 WO 2023284733 A1 WO2023284733 A1 WO 2023284733A1 CN 2022105175 W CN2022105175 W CN 2022105175W WO 2023284733 A1 WO2023284733 A1 WO 2023284733A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
terminus
amino acid
fusion protein
cancer
Prior art date
Application number
PCT/CN2022/105175
Other languages
French (fr)
Chinese (zh)
Inventor
周雅琼
高章照
吴振华
聂磊
胡锋
齐健
王海彬
Original Assignee
海正生物制药有限公司
浙江博锐生物制药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 海正生物制药有限公司, 浙江博锐生物制药有限公司 filed Critical 海正生物制药有限公司
Publication of WO2023284733A1 publication Critical patent/WO2023284733A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the invention relates to the field of biomedicine. Specifically, the present invention discloses a GITR/TGF- ⁇ dual-targeting fusion protein, a conjugate and a pharmaceutical composition comprising the same, and the use of the fusion protein or the conjugate in preparing medicines.
  • Immunotherapy treatment options are broad and include a variety of approaches.
  • One of the most successful treatment options to date has been treatment with immune checkpoint inhibitors (such as anti-PD-L1 antibodies and anti-PD-1 antibodies), which have demonstrated highly durable responses and significantly prolonged survival in a variety of advanced tumors. overall lifetime.
  • immune checkpoint inhibitors such as anti-PD-L1 antibodies and anti-PD-1 antibodies
  • TGF-beta Transforming growth factor-beta
  • TGF-beta is a multifunctional cytokine belonging to the transforming factor superfamily.
  • the TGF- ⁇ signaling pathway is mediated by the TGF- ⁇ receptor complex composed of TGF- ⁇ type I receptor (TGF- ⁇ RI) and TGF- ⁇ type II receptor (TGF- ⁇ RII), and regulates cell growth, proliferation, differentiation, apoptosis Important cellular processes such as death and migration.
  • TGF- ⁇ signaling pathway is closely related to tumor occurrence and development.
  • TGF- ⁇ contributes to epithelial-mesenchymal transition (EMT) of tumor cells, promoting tumor cell invasion and metastasis.
  • EMT epithelial-mesenchymal transition
  • TGF- ⁇ inhibits the proliferation of T cells and B cells and inhibits the production of immune factors by B lymphocytes. It can also weaken the activity of T cells by inhibiting the function of antigen-presenting cells (such as dendritic cells). This mechanism suppresses the antitumor response of the immune system. In addition, TGF- ⁇ can promote the growth and metastasis of tumor cells by stimulating angiogenesis.
  • TGF- ⁇ inhibitors have been developed in the prior art, such as TGF- ⁇ Trap (dimeric extracellular domain of TGF- ⁇ RII, see for example WO2015118175A2 and WO2018205985A1) and anti-TGF- ⁇ monoclonal antibody fresolimumab (see for example US7723486B2 ).
  • TGF- ⁇ Trap dimeric extracellular domain of TGF- ⁇ RII, see for example WO2015118175A2 and WO2018205985A1
  • anti-TGF- ⁇ monoclonal antibody fresolimumab see for example US7723486B2 .
  • the combination therapy of TGF- ⁇ inhibitors and anti-PD-L1 antibodies or anti-PD-1 antibodies can relieve the suppression of the immune system, thereby enhancing the effect of anti-tumor therapy.
  • WO2018205985A1 and WO2015118175A2 disclose bifunctional molecules of anti-PD-L1 antibody and TGF- ⁇ Trap, which can simultaneously block
  • GITR Glucocorticoid-induced tumor necrosis factor receptor
  • TNFRSF tumor necrosis factor receptor superfamily
  • GITR is highly expressed on the surface of regulatory T cells (Treg), and has lower expression on the surface of naive T cells and memory T cells.
  • Teff effector T cells
  • GITR signaling plays an important role in immune regulation.
  • the activation of GITR on the surface of Teff can promote the proliferation and survival of Teff cells, thereby enhancing immune function.
  • the activation of GITR on the surface of Treg promotes the failure of Treg cells, thereby indirectly activating the function of Teff.
  • the present invention provides a dual targeting fusion protein targeting GITR and TGF- ⁇ , which comprises from N-terminus to C-terminus:
  • the first polypeptide comprises an extracellular domain of TGF- ⁇ RII (TGFBR2-ECD);
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain
  • Lk 1 and Lk 2 are each independently a linker or absent;
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL;
  • Ln 1 and Ln 2 are each independently a linker or absent.
  • the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6.
  • the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
  • said third polypeptide comprises the amino acid sequence of SEQ ID NO:3.
  • the dimerization domain comprises from N-terminus to C-terminus the CH2 and CH3 domains of an immunoglobulin, preferably the CH2 and CH3 domains of human IgG1.
  • the second polypeptide has the following structure from the N-terminus to the C-terminus:
  • Dd is a dimerization domain comprising from N-terminus to C-terminus the hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin, preferably the Fc fragment of human IgG1; preferably, the hinge region or part thereof comprises The amino acid sequence of SEQ ID NO:9, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8;
  • Lk 1 and Lk 2 are each independently a linker.
  • the fusion protein comprises the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
  • the invention provides a polynucleotide encoding a fusion protein of the invention.
  • the present invention also provides an expression vector or host cell comprising a polynucleotide encoding the fusion protein of the present invention.
  • the invention also relates to a conjugate comprising a fusion protein of the invention conjugated to at least one therapeutic agent.
  • the therapeutic agent is selected from detectable markers, chemotherapeutic agents, cytotoxins, radionuclides, immune checkpoint inhibitors, cytokines and enzymes.
  • the present invention provides a pharmaceutical composition comprising:
  • the present invention also relates to the use of said fusion protein, polynucleotide, conjugate or pharmaceutical composition.
  • FIG. 1 Analysis of fusion proteins C15 and C2 by reducing protein gel electrophoresis (SDS-PAGE). Molecular weights (kDa) indicated using protein standards are shown on the left. Lane 1: C15; Lane 2: C2.
  • Figure 2 Binding activity of B22, C15, C12 and C2 to TGF- ⁇ analyzed by ELISA.
  • Figure 3 In vitro binding affinity and kinetics of C12, 336B11 and 36E5 to hGITR analyzed by Fortebio.
  • Figure 4 In vitro binding activity of C12 and C15 to CHOS-hGITR cells analyzed by FACS.
  • Figure 5 In vitro activation of GITR by C12, C15 and 36E5 analyzed by GITR Blockade Bioassay. IgG Isotype was used as negative control.
  • FIG. 6 In vitro blocking of TGF- ⁇ activity by C12 and anti-TGF- ⁇ antibody BMK-R2 analyzed by TGF- ⁇ receptor reporter assay. Human IgG1 was used as a negative control.
  • Figure 7 The activities of C12, 336B11 and 36E5 to activate T cells in vitro analyzed by ELISA.
  • Protein samples 336B11, C12, IgG Isotype (IgG isotype, negative control) and 36E5.
  • the protein concentration is from left to right: 10, 1, 0.1 ⁇ g/ml.
  • medium culture medium was used as a blank control.
  • Figure 8 The activities of C12, C15 and 36E5 to activate T cells in vitro.
  • Protein samples C12, C15, 36E5 and IgG Isotype (IgG isotype, negative control).
  • IgG Isotype IgG isotype, negative control.
  • protein concentration from left to right 50, 10, 2, 0.4, 0.08 ⁇ g/ml.
  • Medium culture medium was used as a blank control.
  • Figure 9 Growth curves of tumor volume in mice treated with control and fusion protein C12 in the murine colon cancer MC38-OVA model.
  • Tumor volumes are expressed as "mean ⁇ standard error (SEM)".
  • At least one (species) or “one (species) or more (species)” may mean 1, 2, 3, 4, 5, 6, 7, 8 (species) or more ( kind).
  • the terms "about” and “approximately” when used in conjunction with a numerical variable generally mean that the value of the variable and all values of the variable are within experimental error (e.g., within a 95% confidence interval for the mean) or within Within ⁇ 10% of the specified value, or within a wider range.
  • the expression “comprises” or its synonymous similar expressions “comprising”, “containing” and “having”, etc. are open-ended and do not exclude additional unrecited elements, steps or components.
  • the expression “consisting essentially of” means limiting the scope to the specified elements, steps or ingredients, plus the optional presence of elements, steps or ingredients that do not materially affect the basic and novel characteristics of the claimed subject matter. It should be understood that the expression “comprising” encompasses the expressions “consisting essentially of” and “consisting of”.
  • fusion protein optionally comprising a linker encompasses the inclusion or absence of a linker.
  • polypeptide refers to a polymer comprising at least two amino acids or derivatives thereof linked by peptide bonds.
  • polypeptide and “protein” are often used interchangeably herein.
  • proteins can be formed from one or more polypeptides covalently or non-covalently, eg, dimeric proteins.
  • the n-th amino acid refers to the n-th amino acid counted from the 1st amino acid at the amino terminal (N-terminal) of the polypeptide.
  • the amino terminus (N terminus) and the carboxyl terminus (C terminus) may indicate the relative positions of two or more amino acid sequences in a polypeptide, and do not mean that the two or more amino acid sequences are in close proximity, for example, in the present invention
  • the first polypeptide can be located at the N-terminus of the second polypeptide
  • the second polypeptide can be located at the N-terminus of the third polypeptide
  • the first polypeptide and the second polypeptide and/or the second Additional components eg, amino acid sequences, eg, linkers
  • the amino terminus (N terminus) and the carboxyl terminus (C terminus) can also indicate the position of the specified amino acid or amino acid sequence in the polypeptide in which it is located, for example, the first polypeptide of the present invention can be located at the N terminus of the fusion protein of the present invention It can also be located at the C-terminus of the fusion protein of the present invention.
  • fusion protein or "fusion polypeptide” refers to a protein comprising at least two polypeptides covalently linked that are not normally covalently linked in nature. Fusion proteins can be formed by covalently linking at least two polypeptides by chemical, enzymatic or recombinant DNA techniques.
  • the term "mutation" refers to a substitution, deletion or addition comprising one or more amino acids in a polypeptide.
  • the substitutions may be conservative or non-conservative.
  • suitable conservative amino acid substitutions are known to those skilled in the art, and conservative amino acid substitutions can generally be made without altering the biological activity of the resulting molecule.
  • those skilled in the art recognize that single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter the desired biological activity (see, e.g., Watson et al., Molecular Biology of the Gene, 4th Edition, 1987, The Benjamin/Cummings Pub.co., p.224).
  • the Fc fragment may contain mutations such as deletion of the C-terminal lysine in the CH3 domain.
  • a wild-type polypeptide or protein refers to a naturally occurring polypeptide or protein that has not been artificially modified.
  • a “mutant” or “mutant” of a polypeptide or protein carries a mutation relative to a "wild-type” polypeptide or protein.
  • the wild-type TGF- ⁇ RII extracellular domain comprises the sequence of SEQ ID NO:4.
  • TGF- ⁇ RII ectodomain mutants carry mutations relative to SEQ ID NO:4.
  • polynucleotide or “nucleic acid” refers to an oligomer or polymer comprising at least two linked nucleotides or nucleotide derivatives, including deoxygenated nucleotides, usually linked together by phosphodiester bonds.
  • a fusion protein or polynucleotide of the invention may be "isolated".
  • isolated means that the polypeptide or polynucleotide may, for example, be artificially engineered, and/or separated from other substances in its naturally occurring environment.
  • An "isolated" polynucleotide, such as a cDNA molecule may be substantially free of other cellular material or culture medium when prepared by recombinant techniques, or substantially free of chemical precursors or other chemical components when chemically synthesized.
  • a "vector” is a vehicle for introducing exogenous nucleic acid into a host cell, and when the vector is transformed into an appropriate host cell, the exogenous nucleic acid is amplified or expressed.
  • Vectors include those into which nucleic acids encoding polypeptides or fragments thereof can be introduced, typically by restriction digestion and ligation.
  • Vectors also include those comprising nucleic acid encoding a polypeptide.
  • Vectors usually remain episomal, but can be designed to allow integration of a gene, or part thereof, into the chromosome of the genome.
  • vectors for artificial chromosomes such as yeast artificial vectors and mammalian artificial chromosomes.
  • vector encompasses plasmids, linearized plasmids, viral vectors, cosmids, phage vectors, phagemids, artificial chromosomes (eg, yeast artificial chromosomes and mammalian artificial chromosomes), and the like.
  • a vector can be expressed or replicated in a host cell.
  • expression refers to the process by which a polypeptide is produced by the transcription and translation of a polynucleotide.
  • An "expression vector” includes a vector capable of expressing a polypeptide comprising a polynucleotide sequence encoding a polypeptide of interest.
  • the polynucleotide sequence encoding the polypeptide of interest is operably linked to regulatory sequences capable of affecting its expression.
  • regulatory sequences may include promoter and terminator sequences, and optionally may include one or more origins of replication, one or more selectable markers, enhancers, polyadenylation signals, and the like.
  • Expression vectors are generally derived from plasmid or viral DNA, or may contain elements of both.
  • an expression vector may refer to a recombinant DNA or RNA construct, such as a plasmid, phage vector, recombinant virus or other vector, which, when introduced into an appropriate host cell, results in the expression of cloned DNA or RNA.
  • Appropriate expression vectors are well known to those skilled in the art and include expression vectors replicable in eukaryotic cells and/or prokaryotic cells as well as expression vectors that remain episomal or integrate into the host cell genome.
  • a "host cell” is a cell used to receive, maintain, replicate or amplify a vector. Host cells can also be used to express nucleic acids or polypeptides encoded by vectors. Host cells can be eukaryotic or prokaryotic. Suitable host cells include, but are not limited to, CHO cells, COS cells, HeLa cells, and HEK cells (eg, HEK293 cells).
  • affinity or "binding affinity” is used to measure the strength of the binding between a molecule and its ligand through non-covalent forces, such as the binding strength between the fusion protein of the present invention and its target, such as GITR and Binding strength between extracellular domains of GITRL or between TGFBR2-ECD and TGF- ⁇ .
  • KD can be easily determined using conventional techniques, for example, methods that can be used include but are not limited to: equilibrium dialysis; biofilm layer interferometry, for example by Octet RED96 detection system; enzyme-linked immunosorbent assay (ELISA); surface plasmon Resonance (SPR) method; or by other methods known to the skilled person.
  • methods that can be used include but are not limited to: equilibrium dialysis; biofilm layer interferometry, for example by Octet RED96 detection system; enzyme-linked immunosorbent assay (ELISA); surface plasmon Resonance (SPR) method; or by other methods known to the skilled person.
  • Specific binding means that two molecules bind to each other with a relatively high affinity.
  • the K D value between two molecules that specifically binds can be 10 -6 , 10 -7 , 10 -8 to 10 -9 M or lower.
  • the K D value between GITR and the extracellular domain of GITRL can be 10 -6 to 10 -9 M or lower
  • the K D value between TGFBR2-ECD and TGF- ⁇ can be 10 -6 to 10 -9 A KD value of M or lower binds specifically.
  • treating means preventing, curing, ameliorating, alleviating, arresting or partially arresting the symptoms of a disease or disorder.
  • subjects in need of treatment include, but are not limited to: those with a disease or condition; those at risk of having a disease; and those in whom the condition is to be prevented.
  • a subject is successfully "treated” according to the methods of the invention if the subject exhibits one or more of the following: increased immune response, increased anti-tumor response, lysis of immune cells Increased cellular activity, increased killing of tumor cells by immune cells, decreased number or absence of cancer cells; reduced tumor size; suppressed or absent cancer cell infiltration into surrounding organs (including spread of cancer cells into soft tissue and bone); tumor or inhibition of cancer cell metastasis or absence; inhibition or absence of cancer growth; relief of one or more symptoms associated with a particular cancer; reduction in morbidity and mortality; improved quality of life; reduction in tumorigenicity; A decrease in number or frequency; or some combination of effects.
  • a therapeutically effective amount refers to the amount of a substance, compound, material or composition which is at least sufficient to produce a therapeutic effect after administration to a subject. Thus, it is the amount necessary to prevent, cure, ameliorate, arrest or partially arrest the symptoms of a disease or disorder.
  • a therapeutically effective amount of a fusion protein or pharmaceutical composition of the invention preferably inhibits cell growth or tumor growth by at least about 10%, at least about 20%, at least About 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, preferably at least about 80%.
  • the effect of inhibiting tumor growth can be evaluated in animal model systems predictive of therapeutic effect on human tumors; alternatively, it can also be evaluated by examining the ability to inhibit cell growth, which can be determined in vitro by assays well known to those skilled in the art.
  • a therapeutically effective amount of the fusion protein or pharmaceutical composition of the present invention can reduce tumor size, or alleviate symptoms of a subject in other ways such as preventing and/or treating metastasis or recurrence.
  • a therapeutically effective amount will be influenced by factors such as the subject's weight, severity of symptoms and the particular composition or route of administration chosen.
  • a therapeutically effective amount can be administered in one or more administrations.
  • the invention provides a kind of fusion protein, it comprises from N-terminus to C-terminus:
  • the first polypeptide comprises a TGF- ⁇ RII extracellular domain
  • the second polypeptide comprises a dimerization domain
  • the third polypeptide comprises three GITRL extracellular domains.
  • the fusion protein of the present invention is a GITR/TGF- ⁇ double-targeting fusion protein, which specifically binds TGF- ⁇ and GITR at the same time.
  • the fusion protein of the present invention inhibits the TGF- ⁇ signaling pathway by binding to TGF- ⁇ , and acts as a TGF- ⁇ inhibitor; meanwhile, the fusion protein of the present invention activates the GITR signaling pathway by binding to GITR, and acts as a GITR agonist.
  • the fusion protein of the present invention can be used as an immunotherapeutic agent to induce, promote, enhance, activate or prolong immune response, especially induce, promote, enhance, activate or prolong immune response against tumor cells.
  • the first polypeptide comprises a TGF- ⁇ RII extracellular domain (TGFBR2-ECD).
  • TGF- ⁇ RII extracellular domain refers to the extracellular region of the TGF- ⁇ type II (TGF- ⁇ RII) receptor, which has TGF- ⁇ binding activity. Dimerized TGFBR2-ECD can act as a TGF- ⁇ Trap to bind and capture TGF- ⁇ dimers, thereby inhibiting TGF- ⁇ signaling pathway and eliminating TGF- ⁇ signaling pathway-related immunosuppression. Inhibition of the TGF- ⁇ signaling pathway can be determined using various detection methods known in the art, such as detecting the expression of downstream genes controlled by the TGF- ⁇ signaling pathway.
  • TGFBR2-ECD contained in the first polypeptide may be wild type or a mutant thereof.
  • wild-type TGFBR2-ECD see, for example, WO2015118175A2.
  • the amino acid sequence of an exemplary wild-type TGFBR2-ECD is shown in SEQ ID NO:4.
  • the fusion protein comprising the TGFBR2-ECD mutant is less likely to be degraded during the production process and has better druggability.
  • the TGFBR2-ECD mutant comprises the amino acid sequence of SEQ ID NO:5 or SEQ ID NO:6.
  • the first polypeptide comprises wild-type TGFBR2-ECD.
  • the wild-type TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4.
  • the first polypeptide comprises a TGFBR2-ECD mutant.
  • the TGFBR2-ECD mutant comprises the amino acid sequence of SEQ ID NO:5 or SEQ ID NO:6.
  • the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:4. In yet another specific embodiment, the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:5. In yet another specific embodiment, the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:6.
  • dimerization domain refers to a domain capable of facilitating the combination of two or more polypeptides into dimers or multimers (e.g., three, four, five, six, seven) through covalent or non-covalent interactions. , octa or nine-mer) polypeptides. It is believed that the dimerized TGF- ⁇ RII extracellular domain can act as a TGF- ⁇ Trap (see eg WO2015118175A2 and WO2018205985A1 ) to bind TGF- ⁇ dimers and inhibit TGF- ⁇ signaling pathway. The dimerization domain promotes the fusion protein of the present invention to form dimers or multimers, so that the fusion protein of the present invention can function as TGF- ⁇ Trap.
  • Non-limiting examples of dimerization domains may include the heavy chain constant region 3 (CH3) of immunoglobulins IgG, IgA, IgD, and the heavy chain constant region 4 (CH4) of IgM and IgE.
  • the dimerization domain may further comprise other domains of a particular type of immunoglobulin, such as the hinge region or part thereof and/or the heavy chain constant region 2 (CH2) of IgG1, IgG2, IgG3 or IgG4, to obtain a protein having the desired Specific fusion proteins, such as enhancing antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular endocytosis (ADCP), complement-dependent cytotoxicity (CDC), and binding activity to FcRn to prolong the half-life of fusion proteins Wait.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cellular endocytosis
  • CDC complement-dependent cytotoxicity
  • the dimerization domain comprises the CH2 and CH3 domains of an immunoglobulin, preferably the CH2 and CH3 domains of a human IgGl.
  • the dimerization domain further comprises an immunoglobulin hinge region or portion thereof.
  • the hinge region is the hinge region of human IgG1.
  • the dimerization domain comprises the Fc fragment of an immunoglobulin, preferably the Fc fragment of human IgGl.
  • immunoglobulin Fc fragment refers to the hinge region or part thereof and two or more domains in the heavy chain constant region CH2, CH3, CH4 of an immunoglobulin.
  • the Fc fragment of IgG may comprise the hinge region or part thereof and the CH2 and CH3 domains of an immunoglobulin
  • the Fc fragments of IgM and IgE may comprise the hinge region or part thereof, CH2, CH3 and CH4 domains of an immunoglobulin .
  • the Fc fragment can be derived from any immunoglobulin, e.g., any class or subclass of IgG, IgM, IgD, IgE, IgA, and IgY, e.g., IgGl, IgG2, IgG3, IgG4, IgAl, IgA2, IgG2a, and IgG2b.
  • Portions in the Fc fragment eg, hinge region, CH2 and CH3 may be derived from the same or different immunoglobulins.
  • each portion in the Fc fragment is derived from the same immunoglobulin, preferably human IgG, eg human IgGl, IgG2, IgG3 or IgG4.
  • Fc fragments can be antibody derivatives produced by enzymatic treatment of full-length antibodies (such as non-antigen-binding parts after papain cleavage), and derivatives produced by chemical synthesis or genetic engineering techniques (such as DNA recombinant technology).
  • one or more amino acids may be added, substituted or deleted in the hinge region, CH2 and/or CH3 domains, for example modifying CH2 to alter the glycosylation modification of the Fc fragment, or additions or deletions in the hinge region for formation of molecules
  • One or more cysteines between disulfide bonds may be added, substituted or deleted in the hinge region, CH2 and/or CH3 domains, for example modifying CH2 to alter the glycosylation modification of the Fc fragment, or additions or deletions in the hinge region for formation of molecules
  • One or more cysteines between disulfide bonds may be added, substituted or deleted in the hinge region, CH2 and/or CH3 domains, for example modifying CH2 to alter the glycosylation modification of the Fc fragment, or additions or deletions in the hinge region for formation of molecules
  • One or more cysteines between disulfide bonds may be added, substituted or deleted in the hinge region, CH2 and/or CH3 domains, for example modifying CH2 to alter the glycosylation
  • the second polypeptide further comprises a linker fused to the N-terminus and/or C-terminus of said dimerization domain.
  • the linker is as described herein.
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain
  • Lk 1 and Lk 2 are each independently a linker or absent.
  • the dimerization domain comprises a CH2, CH3, CH4 domain, or a combination thereof. In some embodiments, the dimerization domain comprises at least one CH3 domain. In some embodiments, the dimerization domain comprises CH2 and CH3 domains from N-terminus to C-terminus, preferably the CH2 and CH3 domains of human IgGl. In some embodiments, the CH2 and CH3 domains are derived from human IgGl. In some embodiments, the dimerization domain comprises, from N-terminus to C-terminus, CH2 and CH3 domains derived from human IgGl. In one embodiment, the C-terminal lysine of the CH3 domain is deleted. In one embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In one embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:8.
  • the dimerization domain comprises the hinge region of an immunoglobulin or a portion thereof and a CH2, CH3, CH4 domain or a combination thereof. In some embodiments, the dimerization domain comprises from N-terminus to C-terminus the hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin, preferably the Fc fragment of human IgGl. The CH2 and CH3 domains are described above.
  • the hinge region is derived from human IgGl. In some embodiments, the hinge region does not comprise one or more cysteines. In one embodiment, the hinge region comprises the amino acid sequence of SEQ ID NO:9.
  • the dimerization domain comprises, from N-terminus to C-terminus, the CH2 and CH3 domains of an immunoglobulin, wherein said CH2 and CH3 domains comprise SEQ ID NO: 7 or SEQ ID NO: 8 amino acid sequence.
  • the dimerization domain comprises from N-terminus to C-terminus the hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin, wherein said hinge region or part thereof comprises the sequence of SEQ ID NO:9 Amino acid sequence, the CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In a preferred embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:8.
  • the linker comprises 5-25 amino acids each independently selected from glycine, serine, and alanine.
  • the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10.
  • the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
  • the linker comprises the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 11.
  • neither Lk 1 nor Lk 2 is present, and the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain, said dimerization domain being as previously defined.
  • Lk 1 and Lk 2 are each independently a linker, and the second polypeptide has the following structure from the N-terminus to the C-terminus:
  • Dd is a dimerization domain, and the dimerization domain is as defined above;
  • Lk 1 and Lk 2 are each independently a linker, the linker being as defined above.
  • Lk 1 is a linker
  • Lk 2 is absent
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Lk 1 does not exist
  • Lk 2 is a linker
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain, and the dimerization domain is as defined above;
  • Each of Lk 1 and Lk 2 is a linker, the linker being as defined above.
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminal to the C-terminus, wherein the hinge region comprises the amino acid sequence of SEQ ID NO:9, and the CH2 and CH3 domains comprise SEQ ID The amino acid sequence of NO:7 or SEQ ID NO:8;
  • Lk 1 and Lk 2 are each independently a linker, the linker being as defined above.
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminal to the C-terminus, wherein the hinge region comprises the amino acid sequence of SEQ ID NO:9, and the CH2 and CH3 domains comprise SEQ ID The amino acid sequence of NO:7 or SEQ ID NO:8;
  • Lk 1 and Lk 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminal to the C-terminus, wherein the hinge region comprises the amino acid sequence of SEQ ID NO:9, and the CH2 and CH3 domains comprise SEQ ID NO: the amino acid sequence of 8;
  • Lk 1 and Lk 2 are each independently a linker, wherein Lk 1 comprises the amino acid sequence of SEQ ID NO:10, and Lk 2 comprises the amino acid sequence of SEQ ID NO:11.
  • the third polypeptide comprises 3 GITRL extracellular domains.
  • a single-chain polypeptide comprising 3 GITRL ectodomains (single-chain GITRL ectodomain trimer) compared to a single GITRL ectodomain (monomeric GITRL ectodomain) Trimers similar to the full-length GITRL protein are formed on the cell surface, and such trimers have higher GITR activation activity as GITR agonists.
  • GITR Glucocorticoid-induced tumor necrosis factor receptor
  • TNFRSF tumor necrosis factor receptor superfamily
  • activation (or agonism) of GITR signaling can activate effector T cells and suppress the immunosuppressive activity of regulatory T cells.
  • GITR agonist refers to a substance capable of inducing, promoting, enhancing, activating or prolonging GITR signaling. Activation of GITR signaling can be determined using various assays known in the art, such as by the assays described herein.
  • GITR agonists include, but are not limited to, GITRL, GITRL extracellular domain, GITR agonist antibodies.
  • the fusion protein or conjugate of the present invention can act as a GITR agonist to induce, promote, enhance, activate or prolong an immune response, such as inducing, promoting, enhancing, activating or prolonging an immune response against a tumor or tumor cells.
  • GITRL glucocorticoid-induced TNF-related ligand
  • GITR glucocorticoid-induced TNF receptor
  • GITRL refers to human GITRL protein.
  • the human GITRL (hGITRL) protein is encoded by the TNFSF18 gene, and its amino acid sequence is shown in GenBank accession number NP_005083.2 (or Uniprot ID: Q9Y5U5).
  • GITRL extracellular domain refers to the extracellular region of the human GITRL protein, which specifically binds and agonizes GITR. Those skilled in the art will understand that the exact amino acid sequence that the extracellular domain of GITRL can comprise can vary without affecting its ability to agonize GITR. Typically, the GITRL extracellular domain may comprise about amino acids 71-199 of NP_005083.2, such as amino acids 72-199 (SEQ ID NO: 1) or amino acids 75-199 (SEQ ID NO ) of NP_005083.2 :2). In some embodiments, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the third polypeptide may further comprise a linker between said GITRL extracellular domains.
  • the linker is as described herein.
  • the third polypeptide has the following structure from N-terminus to C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL;
  • Ln 1 and Ln 2 are each independently a linker or absent.
  • the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2. In one embodiment, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:2.
  • the linker comprises 5-25 amino acids each independently selected from glycine, serine, and alanine.
  • the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10.
  • the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
  • said linker comprises the amino acid sequence of SEQ ID NO:12.
  • neither Ln 1 nor Ln 2 is present, and the third polypeptide has the following structure from N-terminus to C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, and the extracellular domain of GITRL is as defined above.
  • Ln 1 and Ln 2 are each independently a linker
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain, and the GITRL extracellular domain is as defined above;
  • Ln 1 and Ln 2 are each independently a linker, the linker being as defined above.
  • Ln 1 is a linker
  • Ln 2 is absent
  • the third polypeptide has the following structure from N-terminus to C-terminus:
  • Ln 1 does not exist
  • Ln 2 is a linker
  • the third polypeptide has the following structure from N-terminus to C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain, and the GITRL extracellular domain is as defined above;
  • Each of Ln 1 and Ln 2 is a linker, the linker being as defined above.
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
  • Ln 1 and Ln 2 are each independently a linker, the linker being as defined above.
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
  • Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:10, SEQ ID NO:11 or SEQ ID NO:12.
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
  • Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:12.
  • the third polypeptide comprises the amino acid sequence of SEQ ID NO:3.
  • the linker may comprise 1-50 amino acids, preferably 1-25, 3-25, 5-25, 4-10 amino acids, for example 4, 5, 6, 7, 8, 9 or 10 amino acids.
  • Amino acids are generally unhindered amino acids such as glycine (G), alanine (A) and serine (S).
  • Suitable linkers are well known to those skilled in the art, for example linkers may comprise glycine, alanine, serine or combinations thereof.
  • Exemplary linkers may include, but are not limited to: polyglycine, polyalanine, GGA, GS, GGSG, GGGS, GGGSGGG, and GGGGSSGS.
  • the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10. In a preferred embodiment, n is an integer selected from 2-5. In one embodiment, n is 4 (SEQ ID NO: 10). In another embodiment, n is 2 (SEQ ID NO: 11).
  • the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
  • Fusion proteins of the invention optionally include a signal peptide.
  • Signal peptides also known as signal sequences, transit peptides, leader sequences, or leader peptides
  • the preferred signal peptide improves the expression level of the fusion protein and ensures the correct folding of the fusion protein, and helps to secrete the fusion protein out of cells.
  • Secretory signal peptides are usually cleaved during protein secretion, allowing the protein to be released outside the cell as a mature protein.
  • the fusion protein of the present invention comprising a secretion signal peptide can also be called a fusion protein precursor, which is also included in the scope of the fusion protein of the present invention.
  • the signal peptide can be located anywhere in the fusion protein. Preferably, the signal peptide is located at the N-terminus of the fusion protein.
  • the signal peptide may be any signal peptide known in the art, preferably a secretory signal peptide commonly used in expression in mammalian cells (eg, CHO cells).
  • the signal peptide sequence comprises the amino acid sequence of SEQ ID NO: 13 or SEQ ID NO: 14.
  • the fusion protein of the present invention comprises from N-terminus to C-terminus:
  • the first polypeptide comprises an extracellular domain of TGF- ⁇ RII (TGFBR2-ECD);
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain
  • Lk 1 and Lk 2 are each independently a linker or absent;
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain
  • Ln 1 and Ln 2 are each independently a linker or absent.
  • the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6. In a preferred embodiment, the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:5.
  • the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2. In a preferred embodiment, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:2.
  • the dimerization domain comprises CH2 and CH3 domains. In a preferred embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In some embodiments, the dimerization domain comprises a hinge region, CH2 and CH3 domains. In one embodiment, the hinge region comprises the amino acid sequence of SEQ ID NO:9. In one embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In a preferred embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:8. In some embodiments, the dimerization domain comprises a hinge region, CH2 and CH3 domains, wherein the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO: 8 amino acid sequence.
  • the linker comprises 5-25 amino acids each independently selected from glycine, serine, and alanine.
  • the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10.
  • the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain comprising the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2;
  • Ln 1 and Ln 2 are each independently a linker.
  • the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:2, and each of Ln 1 and Ln 2 is independently a linker comprising the amino acid sequence of SEQ ID NO:12.
  • said third polypeptide comprises the amino acid sequence of SEQ ID NO:3.
  • the second polypeptide has the following structure from the N-terminus to the C-terminus:
  • Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO :7 or the amino acid sequence of SEQ ID NO:8;
  • Lk 1 and Lk 2 are each independently a linker.
  • the dimerization domain comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, the CH2 and CH3 domains Comprising the amino acid sequence of SEQ ID NO: 8, Lk 1 and Lk 2 are each independently a linker, wherein Lk 1 contains the amino acid sequence of SEQ ID NO: 10, and Lk 2 contains the amino acid sequence of SEQ ID NO: 11.
  • the fusion protein of the present invention comprises from N-terminus to C-terminus:
  • the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6;
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO :7 or the amino acid sequence of SEQ ID NO:8;
  • Lk 1 and Lk 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12;
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2;
  • Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:10, SEQ ID NO:11 or SEQ ID NO:12.
  • the fusion protein of the present invention comprises from the N-terminus to the C-terminus:
  • the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6;
  • the second polypeptide has the following structure from N-terminus to C-terminus:
  • Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO : the amino acid sequence of 8;
  • Lk 1 and Lk 2 are each independently a linker, wherein Lk 1 comprises the amino acid sequence of SEQ ID NO: 10, and Lk 2 comprises the amino acid sequence of SEQ ID NO: 11; and
  • the third polypeptide has the following structure from the N-terminus to the C-terminus:
  • Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
  • Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:12.
  • the fusion protein comprises the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
  • any fusion protein as described above further comprises a signal peptide.
  • the signal peptide is located at the N-terminus of the fusion protein.
  • the signal peptide comprises the amino acid sequence of SEQ ID NO: 13 or SEQ ID NO: 14.
  • any fusion protein as described above comprises a signal peptide at the N-terminus, wherein the signal peptide comprises the amino acid sequence of SEQ ID NO: 13 or SEQ ID NO: 14.
  • the fusion protein forms dimers or multimers.
  • the invention provides isolated polynucleotides encoding fusion proteins of the invention.
  • the polynucleotide encodes a precursor of a fusion protein of the invention.
  • the polynucleotides can be obtained by methods known in the art, for example, recombinant DNA techniques, chemical synthesis, and the like. Polynucleotides can generally be codon-optimized for the host cell used for expression.
  • the present invention also provides an expression vector comprising the polynucleotide of the present invention.
  • Methods for constructing suitable expression vectors using the polynucleotides of the present invention are well known to those skilled in the art, including but not limited to in vitro recombinant DNA, chemical synthesis, and in vivo recombination (genetic recombination).
  • the present invention also provides a host cell comprising the polynucleotide or expression vector of the present invention.
  • Suitable host cells include, but are not limited to, prokaryotic cells (eg, E. coli) and eukaryotic cells (eg, yeast cells, insect cells, and mammalian cells).
  • the host cells are simian COS cells, Chinese Hamster Ovary (CHO) cells, HEK293F cells or immune effector cells (eg effector T cells).
  • the host cell is a yeast cell, an insect cell or a mammalian cell.
  • the host cell is an effector T cell.
  • Another aspect of the present invention relates to a method for producing the fusion protein of the present invention, the method comprising:
  • polynucleotide or expression vector of the present invention can be used to introduce the polynucleotide or expression vector of the present invention (especially the polynucleotide or expression vector encoding the expression precursor of the fusion protein of the present invention) into suitable host cells.
  • Such methods include, but are not limited to, lipofection, electroporation, stable transfection, calcium phosphate transfection, and the like.
  • to further purify the fusion protein of the present invention conventional affinity chromatography, ion exchange chromatography, hydrophobic interaction chromatography, reverse phase chromatography, gel filtration or a combination thereof can be used.
  • the invention further provides a conjugate comprising a fusion protein of the invention conjugated to at least one therapeutic agent.
  • conjugate refers to the linking of two or more moieties to each other by covalent or non-covalent interactions.
  • fusion proteins of the invention and at least one therapeutic agent are covalently conjugated.
  • the therapeutic agent is selected from detectable markers, chemotherapeutic agents, cytotoxins, radionuclides, immune checkpoint inhibitors, cytokines, and enzymes.
  • the therapeutic agent is a detectable label, such as a fluorescent or luminescent label, a radioactive label, an MRI (magnetic resonance imaging) or CT (computed tomography) contrast agent.
  • a detectable label such as a fluorescent or luminescent label, a radioactive label, an MRI (magnetic resonance imaging) or CT (computed tomography) contrast agent.
  • the therapeutic agent is a cytotoxin.
  • cytotoxins include, but are not limited to, paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, epipodophyllotoxin glucopyranoside, epipodophyllotoxin thienenoside, Vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxyanthraxendione, mitoxantrone, mitoxantrone, actinomycin D, 1-dehydrotestosterone, sugar Corticosteroids, procaine, tetracaine, lidocaine, carcinomacin, calicheamicin, maytansin, alistatin, propranolol and puromycin and their analogs or homologues.
  • the therapeutic agent is a chemotherapeutic agent.
  • chemotherapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil, carbamide), alkylating agents (e.g., nitrogen mustard, chlorambucil, phenylalanine mustard, carmustine (BSNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, Mitomycin C and cis-dichlorodiamine platinum(II) (DDP) cisplatin), anthraninomycins (eg, daunorubicin and doxorubicin), antibiotics (eg, actinomycin D , bleomycin, mithromycin, and antramycin (AMC)) and antimitotic agents (eg, vincristine and vinblastine).
  • the therapeutic agent is a biologically active protein, such as a toxin with enzymatic activity or an active fragment thereof, such as abrin, ricin A, Pseudomonas exotoxin, or diphtheria toxin.
  • a biologically active protein such as a toxin with enzymatic activity or an active fragment thereof, such as abrin, ricin A, Pseudomonas exotoxin, or diphtheria toxin.
  • the therapeutic agent is a cytokine.
  • cytokines include, but are not limited to, tumor necrosis factors (such as TNF ⁇ and TNF ⁇ ), interferons (such as INF ⁇ , INF ⁇ , and INF ⁇ ), interleukins (such as IL-1, IL-2, IL-4, IL-5, IL- 6.
  • colony-stimulating factors such as macrophage colony stimulatory factors, granulocyte colony stimulating factor, granulocyte and macrophage colony stimulating factor, multiple colony stimulating factor, stem cell factor
  • growth factors such as VEGF, HGF, FGF, FGF-2, PDGF, IGF, TGF, NGF, EPO and EGF
  • chemokines such as VEGF, HGF, FGF, FGF-2, PDGF, IGF, TGF, NGF, EPO and EGF
  • the therapeutic agent is an immune checkpoint inhibitor.
  • immune checkpoint inhibitors include antibodies or antigen-binding fragments thereof against immune checkpoint molecules such as PD-L1, PD-1, CTLA4, LAG-3, BTLA, TIM-2, LAIR1, HVEM, and TIM-4 .
  • the antibody or antigen-binding fragment thereof may be selected from synthetic antibodies, recombinantly produced antibodies, multispecific antibodies, bispecific antibodies, human antibodies, non-human antibodies, humanized antibodies, chimeric antibodies, intrabodies, single domain antibodies , Fab fragment, Fab' fragment, F(ab') 2 fragment, Fv fragment, dsFv fragment, Fd fragment, Fd' fragment, scFv, scFab, diabody.
  • the therapeutic agent is a radionuclide.
  • radionuclides include, but are not limited to, iodine-125, iodine-131, indium-111, indium-131, yttrium-90, rhodium-105, bismuth-212, and lutetium-177.
  • the therapeutic agent can also be an enzyme capable of producing a detectable product (e.g., horseradish peroxidase), gold nanoparticles/nanorods, virosomes, liposomes, nanomagnetic particles, prodrug-activating enzymes (e.g., DT-cardiac diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)) and nanoparticles in any form, etc.
  • a detectable product e.g., horseradish peroxidase
  • gold nanoparticles/nanorods e.g., gold nanoparticles/nanorods, virosomes, liposomes, nanomagnetic particles, prodrug-activating enzymes (e.g., DT-cardiac diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)) and nanoparticles in any form, etc.
  • DTD DT-cardiac diaphorase
  • conjugates are known to those skilled in the art.
  • the therapeutic agents described above can be conjugated to fusion proteins of the invention using linker technology used in the art.
  • linker types that have been used for cytotoxin conjugation include, but are not limited to, hydrazone, thioether, ester, disulfide, and peptide linkers.
  • polynucleotides encoding protein therapeutics such as cytokines, antibodies or antigen-binding fragments thereof, biologically active proteins or enzymes as described above
  • polynucleotides encoding fusion proteins of the present invention can be combined using recombinant DNA technology. Operably linked and expressed in a suitable host cell.
  • the present invention further provides a pharmaceutical composition, which comprises: (i) the fusion protein or conjugate of the present invention; and (ii) a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all substances that are physiologically compatible.
  • pharmaceutically acceptable carriers include: solvents (such as water or oils), dispersants, suspending agents, coatings, diluents, adjuvants, antioxidants (such as ascorbic acid, ascorbyl palmitate, sodium sulfite, , butylated hydroxyanisole (BHA), propyl gallate, sodium metabisulfite, ⁇ -tocopherol, etc.), excipients, preservatives (such as antibacterial and antifungal agents), surfactants (such as polysorbates , poloxamer, triton, sodium octyl glycoside, sodium lauryl sulfate, sodium lauryl sulfate, lauryl-sulfobetaine, polyethylene glycol, polypropylene glycol, etc.), wetting agent, emulsifier, filler , carbohydrates (such as fructose, sucrose, trehalose), sorbitol, etc.,
  • the fusion protein, conjugate or pharmaceutical composition of the present invention can be administered through one or more administration routes using one or more methods known in the art. Those skilled in the art will appreciate that the route and/or manner of administration will vary depending on the desired result. Preferred routes of administration include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration such as injection or infusion.
  • parenteral administration refers to modes of administration other than enteral and topical administration, usually injection, including but not limited to intravenous, intramuscular, intraarterial, intrathecal, intrasaccular, intraorbital, Intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcutaneous, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, and intrasternal injection and infusion.
  • the fusion protein, conjugate or pharmaceutical composition of the present invention may also be administered by non-parenteral routes, such as topical, epidermal or mucosal routes, for example, intranasally, orally, vaginally, rectally, sublingually or partial.
  • the pharmaceutical composition can be prepared in various dosage forms suitable for administration, such as solutions, suspensions, microemulsions, emulsions, liposomes, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the pharmaceutical composition is prepared as injection, powder or liposome.
  • the fusion protein, polynucleotide, conjugate or pharmaceutical composition provided by the present invention is used for treating and/or preventing cancer.
  • the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to activate GITR signaling and inhibit TGF- ⁇ signaling.
  • the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to induce, promote, enhance, activate or prolong an immune response in a subject in need thereof.
  • the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to induce, promote, enhance, activate or prolong an immune response against a tumor or tumor cells in a subject in need thereof.
  • the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to activate effector T cells, such as CD8 + effector T cells.
  • the present invention also relates to the use of the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention in the preparation of medicines.
  • the medicament is for the treatment and/or prevention of cancer.
  • the medicament is for activating GITR signaling and inhibiting TGF-beta signaling.
  • the medicament is for inducing, promoting, enhancing, activating or prolonging an immune response in a subject in need thereof.
  • the medicament is for inducing, promoting, enhancing, activating or prolonging an immune response against a tumor or tumor cells in a subject in need thereof.
  • the drug is used to activate effector T cells.
  • the T cells are CD8 + effector T cells.
  • the present invention further provides a method for treating and/or preventing cancer, which comprises administering a therapeutically effective amount of the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention to a subject in need.
  • the method induces, promotes, enhances, activates or prolongs an immune response against a tumor or tumor cells in a subject in need thereof.
  • the present invention also provides a method for inducing, promoting, enhancing, activating or prolonging an immune response, which comprises administering the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention.
  • the present invention also provides a method for activating GITR signal transduction and inhibiting TGF- ⁇ signal transduction, and/or a method for activating effector T cells, which comprises administering the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention .
  • cancer refers to or describes the physiological condition in mammals that is often characterized by unregulated cell growth.
  • Non-limiting examples of cancer include, but are not limited to, adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, and leukemia.
  • cancer More specific examples include: squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, Hodgkin and non-Hodgkin lymphoma, pancreatic cancer, glioblastoma, glioma, Cervical cancer, ovarian cancer, liver cancer such as hepatocellular carcinoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma (such as multiple myeloma), salivary gland cancer, kidney cancer such as renal cell carcinoma and Kleman's tumor, basal cell carcinoma, melanoma, prostate cancer, vulvar cancer, thyroid cancer, testicular cancer, esophageal cancer and various types of head and neck cancer.
  • liver cancer such as hepatocellular carcinoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma (such as multiple myeloma), salivary gland cancer, kidney cancer such as renal cell carcinoma and Kleman's
  • the cancer is selected from adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, leukemia, squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, Hodgkin's and non-small cell lung cancer.
  • Hodgkin lymphoma pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, hepatocellular carcinoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, bone marrow cancer, multiple myeloma, salivary gland cancer, kidney cancer, renal cell carcinoma, Wellman's tumor, basal cell carcinoma, melanoma, prostate cancer, vulvar cancer, thyroid cancer, testicular cancer, esophageal cancer, and head and neck cancer.
  • the term "subject” refers to mammals, including, but not limited to, goats, sheep, pigs, rats, mice, rabbits, guinea pigs, cows, horses, dogs, cats, non-human Long animals (such as gorillas, baboons and chimpanzees) and humans. In some embodiments, the subject is a human.
  • the fusion proteins, conjugates or pharmaceutical compositions of the invention can be used in combination with other therapeutic agents.
  • Other therapeutic agents include, for example, chemotherapeutic agents, immune checkpoint inhibitors, or antibodies targeting tumor-associated antigens.
  • the other therapeutic agent is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is an anti-PD-L1 antibody, an anti-PD-1 antibody, an anti-CTLA4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, or a combination thereof.
  • the fusion protein, conjugate or pharmaceutical composition of the invention and the other therapeutic agent can be administered all at once or separately. When administered separately (in the case of mutually different administration regimens), they may be administered continuously without interruption or at predetermined intervals.
  • the fusion protein of the present invention or the pharmaceutical composition of the present invention may also be used in combination with radiotherapy, for example comprising administering ionizing radiation to the patient earlier than, during and/or after the fusion protein of the present invention or pharmaceutical composition Application process.
  • kits which includes the fusion protein of the present invention, its encoding polynucleotide or expression vector or a combination thereof, and instructions for use.
  • Kits generally include a label indicating the intended use and/or method of use of the kit contents.
  • label includes any written or recorded material provided on or with the kit or otherwise provided with the kit.
  • the fusion protein of the present invention may comprise a TGFBR2-ECD mutant.
  • a fusion protein comprising a TGFBR2-ECD mutant is less likely to be broken and degraded during production, showing Greater stability.
  • the fusion protein of the present invention targets GITR and TGF- ⁇ dual targets, and can suppress TGF- ⁇ while activating GITR.
  • the fusion protein of the present invention has a higher activity of activating effector T cells. The present invention proposes this dual-target fusion protein product for the first time, which has great application potential in clinic.
  • the wild-type TGFBR2-ECD (SEQ ID NO: 4) and the single-chain GITRL extracellular domain trimer (SEQ ID NO: 3, which contains 3 GITRL extracellular domains connected by a linker) are connected through the Fc fragment, Construct and produce GITR/TGF- ⁇ dual targeting fusion protein C2 (SEQ ID NO: 17) (see Table 2).
  • the fusion protein C2 (as shown in Example 2) with wild-type TGFBR2-ECD is prone to degradation, resulting in a large fragment comprising a single-chain GITRL extracellular domain trimer and an Fc fragment and a small fragment containing TGFBR2-ECD ( see picture 1). It was found by mass spectrometry that the degradation site of the fusion protein was mainly located in three regions of the N-terminus of wild-type TGFBR2-ECD, namely the first region Q-K-S and the second region N-NDM-I (where "-" indicates the break point), And the third area "DNNG” (see Table 1).
  • TGFBR2-ECD mutants for the preparation of fusion proteins.
  • hydrophilic glycine (G) and/or hydrophilic serine (S) that can undergo potential O-glycosylation modification are introduced into the three regions of wild-type TGFBR2-ECD, and the obtained TGFBR2-
  • S hydrophilic serine
  • SEQ ID NO:5 The amino acid sequence of the ECD mutant is shown in SEQ ID NO:5; a wide range of hydrophilic amino acids are introduced into the above three regions of wild-type TGFBR2-ECD and their upstream and downstream regions, especially serine, which can undergo potential O-sugar modification (S) and threonine (T), the amino acid sequence of the obtained TGFBR2-ECD mutant is shown in SEQ ID NO: 6 (see Table 1).
  • TGFBR2-ECD sequence structure SEQ ID NO: Wild type IPPHV QKS V NNDMI VT DNNG AVKF...NIIFSEEYNTSNPD 4 mutant IPPHV GGS V GGSG IVT GGSG AVKF...NIIFSEEYNTSNPD 5 mutant TPPHT QTS T NNSMI TT GTSG ATKY...NIIFSEEYNTSNPD 6
  • GITR/TGF- ⁇ dual-targeting fusion protein C12 (the TGFBR2-ECD mutant is located at the N-terminus of the fusion protein, formula (1), SEQ ID NO: 15), C15 (the TGFBR2-ECD mutant is located at the C-terminal of the fusion protein). terminal, formula (2), SEQ ID NO:16) and C2 (wild-type TGFBR2-ECD is located at the C-terminal of the fusion protein, formula (2), SEQ ID NO:17) (see Table 2).
  • 336B11 Simultaneously construct and produce single-targeting protein 336B11 (see 336B11 in CN107406878A for amino acid sequence) and B22 as contrasting proteins, wherein 336B11 is GITR single-targeting protein (single-chain GITRL extracellular domain trimer fused to the N-terminal of Fc fragment ), B22 is a TGF- ⁇ single-targeting protein (wild-type TGFBR2-ECD fused to the N-terminus of the Fc fragment) (see Table 2).
  • 336B11 is GITR single-targeting protein (single-chain GITRL extracellular domain trimer fused to the N-terminal of Fc fragment )
  • B22 is a TGF- ⁇ single-targeting protein (wild-type TGFBR2-ECD fused to the N-terminus of the Fc fragment) (see Table 2).
  • Reducing protein gel electrophoresis take a sample and add an appropriate amount of sample buffer, add DTT to one of the samples for reduction, heat the reduced sample in a 70°C water bath for 10 minutes, load 2 ⁇ g of the sample, and separate by 10% protein gel electrophoresis.
  • the experimental results are shown in FIG. 1 , lane 1 is C15 (containing TGFBR2-ECD mutant), and lane 2 is C2 (containing wild-type TGFBR2-ECD).
  • a major protein band is shown in swimming lane 1, which is consistent with the expected molecular weight of the target protein C15 (SEQ ID NO: 15).
  • Lane 2 shows three main protein bands, the band with the largest molecular weight is consistent with the expected molecular weight of the target protein C2 (SEQ ID NO: 16), and the other two protein bands are C2 in the N-terminus of TGFBR2-ECD.
  • the degradation products produced by the fragmentation correspond to the large fragment (degradation band 1) containing single-chain GITRL ectodomain trimer and Fc fragment (degradation band 1) and the small fragment (degradation band 2) containing TGFBR2-ECD, respectively. It can be seen that C2 is significantly degraded compared to C15. Degradation of the GITR/TGF- ⁇ dual targeting fusion protein was effectively controlled by inclusion of the TGFBR2-ECD mutant.
  • Example 3 The activity of GITR/TGF- ⁇ dual-targeting fusion protein binding to TGF- ⁇ in vitro
  • Test purpose To detect the binding activity of single-target and dual-target fusion proteins to TGF- ⁇ by ELISA, to compare the pros and cons of dual-target fusion protein samples with different structural designs for TGF- ⁇ binding in vitro, and B22 is used as a positive control.
  • Test samples B22, C15, C12 and C2 prepared as in Example 1.
  • Human TGF- ⁇ (purchased from Acro) at a concentration of 0.5 ⁇ g/ml was directly coated onto a 96-well plate, 100 ⁇ l/well, overnight at 4°C.
  • Example 4 Affinity and kinetics of GITR/TGF- ⁇ dual-targeting fusion protein binding to hGITR in vitro
  • Detection purpose pass Octet RED96 measures the affinity of GITR/TGF- ⁇ dual targeting fusion protein to hGITR.
  • Test samples C12 and 336B11 prepared as in Example 1 and anti-GITR agonist antibody 36E5 (see CN103951753A for the amino acid sequence).
  • Coupled ligand This method uses HIS1K (Anti-Penta-HIS biosensor, ForteBio) chip, soak the chip with SD buffer for 10-15 minutes in advance, and load the ligand (hGITR) at a concentration of 50 ⁇ g/ml for a preset time For 300 seconds, the ligand loading preset value is 1nm, and the chip is infiltrated with running buffer (SD buffer) until the baseline is stable to remove free ligand.
  • HIS1K Anti-Penta-HIS biosensor, ForteBio
  • Example 5 The activity of GITR/TGF- ⁇ dual-targeting fusion protein binding to CHOS-hGITR cells in vitro
  • Test samples C12 and C15 prepared as in Example 1.
  • CHOS cells were transfected with the recombinant vector of the full sequence of membrane protein hGITR (Uniprot ID: Q9Y5U5), pressurized selection after 24 hours of transfection, and selected medium (CD FortiCHO TM medium + 15 ⁇ mol/L MSX + 10 ⁇ g/ml purine Mycin), until the cell viability increased.
  • the monoclonal cell line was screened by the limiting dilution method, and the monoclonal cell line was incubated with 336B11, and then the supernatant was removed, and anti-human IgG-488 (Invitrogen, 1:2000) was added as a detection antibody, and was screened by flow cytometry. Cell lines with higher mean fluorescence intensity (MFI) and better peak shape are used as stable transfection CHOS-hGITR cell lines.
  • MFI mean fluorescence intensity
  • Test results the results are shown in Figure 4 and Table 5. From the results of EC 50 (nM), it can be seen that the binding activity of C12 to CHOS-hGITR cells is slightly better than that of C15.
  • GITR/TGF- ⁇ dual targeting fusion protein activates GITR activity in vitro
  • GITR Blockade Bioassay is a cell-dependent bioluminescent detection method. Its detection principle is: when GITR agonists are added to the genetically engineered cell line GS-H3/GITR cells expressing GITR (provided by GenScript), GITR activation and Expression of the luciferase gene under the control of NFAT, which can subsequently be detected by adding a luciferase detection reagent (Bio-Glo, Promega; containing the luciferase substrate luciferin and reaction buffer) The luminescence signal generated by the catalytic substrate can be used to evaluate the activity of GITR agonists to activate GITR.
  • a luciferase detection reagent Bio-Glo, Promega; containing the luciferase substrate luciferin and reaction buffer
  • Test samples C12 and C15 prepared as in Example 1, anti-GITR agonist antibody 36E5 (positive control) and IgG isotype (negative control).
  • the test results are shown in Figure 5 and Table 6.
  • the anti-GITR agonist antibody 36E5 can effectively bind to GITR and activate downstream pathways to generate signals.
  • the negative control IgG isotype cannot bind to GITR and cannot activate downstream pathways to generate signals.
  • Both C12 and C15 can activate downstream signaling pathways with similar activities, and the dose-response curve EC 50 and dose-response maximum of the samples are significantly better than 36E5.
  • Example 7 In vitro inhibition of TGF- ⁇ activity by GITR/TGF- ⁇ dual targeting fusion protein
  • TGF- ⁇ receptor reporter gene assay (TGF- ⁇ receptor Reporter Gene Bioassay) is a bioluminescent detection method that relies on genetically engineered cell lines.
  • TGF- ⁇ protein binds to the TGF- ⁇ receptor on the cell surface
  • the receptor-mediated signal transduction pathway is activated to induce the expression of luciferase, and then by adding the luciferase detection reagent (Bio-Glo , Promega) to read the luminescent signal generated by the luciferase-catalyzed substrate.
  • the addition of anti-TGF- ⁇ antibodies or inhibitors blocks the binding of TGF- ⁇ to its receptor, resulting in a decrease in the luminescent signal.
  • A549/SBE cells (provided by GenScript) were used as a functional cell line, and the antibody TGF- ⁇ antibody BMK-R2 (GenScript) was used as a positive control to evaluate the GITR/TGF- ⁇ dual-targeting fusion protein Block the activity of TGF- ⁇ in vitro.
  • Detection samples C12 prepared as in Example 1 and anti-TGF- ⁇ antibody BMK-R2.
  • Example 8 GITR/TGF- ⁇ dual-targeting fusion protein activates the activity of effector T cells in vitro
  • Test samples C12, C15, 336B11 and anti-GITR agonist antibody 36E5 prepared as in Example 1.
  • the Raji cell cross-linking method was used to detect the activity of the fusion protein to activate T cells.
  • the detection index was IFN- ⁇ , and the steps were as follows:
  • sample proteins C12, C15, 36E5 and 336B11 the highest concentration is 50 ⁇ g/ml or 10 ⁇ g/ml, 5-fold gradient dilution, 4 concentration gradients for each sample, and 3 replicate wells for each concentration;
  • Example 9 In vivo tumor suppressive activity of GITR/TGF- ⁇ dual targeting fusion protein
  • the anti-tumor effect of the fusion protein of the present invention was tested in the mouse model of colon cancer MC38-OVA cell line subcutaneously implanted with C57BL/6-hGITR female transgenic mice.
  • mice Collect MC38-OVA cells in the exponential growth phase (Crown Biotechnology Co., Ltd.), and subcutaneously inoculate C57BL/6-hGITR on the right back on Day 0 at a dose of 1 ⁇ 10 6 cells/mouse
  • Day 3 tumor cell inoculation
  • the intraperitoneal injection was administered on the 3rd, 6th, 9th, and 12th day after the inoculation of tumor cells, and the administration was administered four times in total.
  • the dosage of the test group was C12 30mg/kg, and the control group was an equal volume of vehicle (PBS solution). From the 6th day (Day 6), the body weight and tumor size of the mice were measured twice a week.
  • the tumor growth of the treatment group and the control group is shown in FIG. 9 .
  • the average tumor volume of mice in the vehicle control group was 897.16 mm 3 on the 8th day (Day 20) after administration.
  • the average tumor volume of mice in the vehicle control group was 2025.13 mm 3 on day 12 (Day 24) after administration.
  • the average tumor volume of the fusion protein C12 (30 mg/kg) treatment group on Day 24 was 1039.94 mm 3 , and the relative tumor inhibition rate TGI (%) was 49%. It shows that the dual targeting fusion protein C12 of the present invention has significant tumor suppressing activity.
  • TGI (%) (1-T/C) ⁇ 100%, where T and C are the tumor volume (TV) of the treatment group and the control group at a specific time point, respectively.

Abstract

The present invention relates to the field of biomedicine, and provides a GITR/TGF-β dual-targeted fusion protein, a conjugate and pharmaceutical composition comprising same, and a use of the fusion protein or conjugate in the preparation of a drug.

Description

GITR/TGF-β双靶向融合蛋白及其用途GITR/TGF-β dual targeting fusion protein and its application 技术领域technical field
本发明涉及生物医药领域。具体而言,本发明公开了GITR/TGF-β双靶向融合蛋白、包含其的缀合物和药物组合物,以及所述融合蛋白或缀合物在制备药物中的用途。The invention relates to the field of biomedicine. Specifically, the present invention discloses a GITR/TGF-β dual-targeting fusion protein, a conjugate and a pharmaceutical composition comprising the same, and the use of the fusion protein or the conjugate in preparing medicines.
背景技术Background technique
免疫疗法治疗选择广泛,包括多种方法。迄今为止,最成功的治疗方案之一是免疫检查点抑制剂(例如抗PD-L1抗体和抗PD-1抗体)治疗,其可在多种晚期肿瘤中表现出高度持久的反应和显著延长的整体生存期。尽管检查点抑制剂在抗癌治疗中取得了进展,但绝大多数患者对检查点抑制剂仍无响应。Immunotherapy treatment options are broad and include a variety of approaches. One of the most successful treatment options to date has been treatment with immune checkpoint inhibitors (such as anti-PD-L1 antibodies and anti-PD-1 antibodies), which have demonstrated highly durable responses and significantly prolonged survival in a variety of advanced tumors. overall lifetime. Despite the progress of checkpoint inhibitors in anticancer therapy, the vast majority of patients remain unresponsive to checkpoint inhibitors.
转化生长因子-β(TGF-β)是属于转化因子超家族的多功能细胞因子。TGF-β信号通路由TGF-βⅠ型受体(TGF-βRⅠ)和TGF-βⅡ型受体(TGF-βRⅡ)组成的TGF-β受体复合物介导,调控细胞生长、增殖、分化、凋亡、迁移等重要细胞过程。TGF-β信号通路与肿瘤发生和发展密切相关。在一方面,TGF-β有助于肿瘤细胞上皮-间充质转化(EMT),促进肿瘤细胞浸润和转移。在另一方面,TGF-β抑制T细胞和B细胞的增殖并抑制B淋巴细胞产生免疫因子,也可以通过抑制抗原提呈细胞(如树突状细胞)的功能减弱T细胞的活性,通过多种机制抑制免疫系统的抗肿瘤反应。此外,TGF-β可以通过刺激血管生成促进肿瘤细胞的生长和转移。Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine belonging to the transforming factor superfamily. The TGF-β signaling pathway is mediated by the TGF-β receptor complex composed of TGF-β type I receptor (TGF-βRⅠ) and TGF-β type II receptor (TGF-βRⅡ), and regulates cell growth, proliferation, differentiation, apoptosis Important cellular processes such as death and migration. TGF-β signaling pathway is closely related to tumor occurrence and development. In one aspect, TGF-β contributes to epithelial-mesenchymal transition (EMT) of tumor cells, promoting tumor cell invasion and metastasis. On the other hand, TGF-β inhibits the proliferation of T cells and B cells and inhibits the production of immune factors by B lymphocytes. It can also weaken the activity of T cells by inhibiting the function of antigen-presenting cells (such as dendritic cells). This mechanism suppresses the antitumor response of the immune system. In addition, TGF-β can promote the growth and metastasis of tumor cells by stimulating angiogenesis.
现有技术中已开发了一些TGF-β抑制剂,例如TGF-βTrap(二聚的TGF-βRⅡ的胞外结构域,参见例如WO2015118175A2和WO2018205985A1)和抗TGF-β单克隆抗体fresolimumab(参见例如US7723486B2)。临床上使用TGF-β抑制剂与抗PD-L1抗体或抗PD-1抗体联合疗法解除免疫系统的抑制,从而增强抗肿瘤治疗效果。例如,WO2018205985A1和WO2015118175A2公开了的抗PD-L1抗体和TGF-βTrap的双功能分子,可以同时阻断TGF-β信号通路并抑制免疫检查点,用于癌症治疗。Some TGF-β inhibitors have been developed in the prior art, such as TGF-β Trap (dimeric extracellular domain of TGF-βRII, see for example WO2015118175A2 and WO2018205985A1) and anti-TGF-β monoclonal antibody fresolimumab (see for example US7723486B2 ). Clinically, the combination therapy of TGF-β inhibitors and anti-PD-L1 antibodies or anti-PD-1 antibodies can relieve the suppression of the immune system, thereby enhancing the effect of anti-tumor therapy. For example, WO2018205985A1 and WO2015118175A2 disclose bifunctional molecules of anti-PD-L1 antibody and TGF-βTrap, which can simultaneously block TGF-β signaling pathway and inhibit immune checkpoint for cancer treatment.
糖皮质激素诱导的肿瘤坏死因子受体(Glucocorticoid-induced tumor necrosis factor receptor,GITR或AITR)是肿瘤坏死因子受体超家族(TNFRSF)中的一员。GITR在调节性T细胞(regulatory T cells,Treg)表面高表达,在幼稚T细胞和记忆T细胞表面有较低的表达。当效应T细胞(effector T cells,Teff)被激活后,GITR的表达水平会在短时间内快速升高。GITR信号传导在免疫调节中起重要作用。一方面,Teff表面GITR的激活可以促进Teff细胞的增殖和存活,从而增强免疫功能。在另一方面,Treg表面 GITR的激活促使Treg细胞衰竭,从而间接的起到激活Teff功能的作用。Glucocorticoid-induced tumor necrosis factor receptor (GITR or AITR) is a member of the tumor necrosis factor receptor superfamily (TNFRSF). GITR is highly expressed on the surface of regulatory T cells (Treg), and has lower expression on the surface of naive T cells and memory T cells. When effector T cells (Teff) are activated, the expression level of GITR will increase rapidly in a short time. GITR signaling plays an important role in immune regulation. On the one hand, the activation of GITR on the surface of Teff can promote the proliferation and survival of Teff cells, thereby enhancing immune function. On the other hand, the activation of GITR on the surface of Treg promotes the failure of Treg cells, thereby indirectly activating the function of Teff.
发明内容Contents of the invention
在一方面,本发明提供一种靶向GITR和TGF-β的双靶向融合蛋白,其从N端至C端包含:In one aspect, the present invention provides a dual targeting fusion protein targeting GITR and TGF-β, which comprises from N-terminus to C-terminus:
第一多肽–第二多肽–第三多肽    式(1);The first polypeptide-the second polypeptide-the third polypeptide Formula (1);
或者or
第三多肽–第二多肽–第一多肽    式(2);The third polypeptide-the second polypeptide-the first polypeptide formula (2);
其中in
所述第一多肽包含TGF-βRⅡ的胞外结构域(TGFBR2-ECD);The first polypeptide comprises an extracellular domain of TGF-βRII (TGFBR2-ECD);
所述第二多肽从N端至C端具有如下结构:The second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 –Dd–Lk 2 ,
其中in
Dd为二聚化结构域;Dd is a dimerization domain;
Lk 1和Lk 2各自独立地为接头或不存在;并且 Lk 1 and Lk 2 are each independently a linker or absent; and
所述第三多肽从N端至C端具有如下结构:The third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ,
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL;
Ln 1和Ln 2各自独立地为接头或不存在。 Ln 1 and Ln 2 are each independently a linker or absent.
在一实施方案中,所述TGFBR2-ECD包含SEQ ID NO:4、SEQ ID NO:5或SEQ ID NO:6的氨基酸序列。In one embodiment, the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6.
在一实施方案中,所述GITRL胞外结构域包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列。在一具体实施方案中,所述第三多肽包含SEQ ID NO:3的氨基酸序列。In one embodiment, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2. In a specific embodiment, said third polypeptide comprises the amino acid sequence of SEQ ID NO:3.
在一实施方案中,所述二聚化结构域从N端至C端包含免疫球蛋白的CH2和CH3结构域,优选人IgG1的CH2和CH3结构域。In one embodiment, the dimerization domain comprises from N-terminus to C-terminus the CH2 and CH3 domains of an immunoglobulin, preferably the CH2 and CH3 domains of human IgG1.
在一实施方案中,所述第二多肽从N端至C端具有如下结构:In one embodiment, the second polypeptide has the following structure from the N-terminus to the C-terminus:
Lk 1–Dd–Lk 2Lk 1 -Dd -Lk 2 ;
其中in
Dd为二聚化结构域,其从N端至C端包含免疫球蛋白的铰链区或其部分、CH2和CH3结构域,优选人IgG1的Fc片段;优选地,所述铰链区或其部分包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain comprising from N-terminus to C-terminus the hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin, preferably the Fc fragment of human IgG1; preferably, the hinge region or part thereof comprises The amino acid sequence of SEQ ID NO:9, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8;
Lk 1和Lk 2各自独立地为接头。 Lk 1 and Lk 2 are each independently a linker.
在一具体实施方案中,所述融合蛋白包含SEQ ID NO:15或SEQ ID NO:16的氨基酸序列。In a specific embodiment, the fusion protein comprises the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
在另一方面,本发明提供一种多核苷酸,其编码本发明的融合蛋白。本发明还提供一种表达载体或宿主细胞,其包含编码本发明的融合蛋白的多核苷酸。In another aspect, the invention provides a polynucleotide encoding a fusion protein of the invention. The present invention also provides an expression vector or host cell comprising a polynucleotide encoding the fusion protein of the present invention.
本发明还涉及一种缀合物,其包含与至少一种治疗剂缀合的本发明的融合蛋白。优选地,所述治疗剂选自可检测标记物、化疗剂、细胞毒素、放射性核素、免疫检查点抑制剂、细胞因子和酶。The invention also relates to a conjugate comprising a fusion protein of the invention conjugated to at least one therapeutic agent. Preferably, the therapeutic agent is selected from detectable markers, chemotherapeutic agents, cytotoxins, radionuclides, immune checkpoint inhibitors, cytokines and enzymes.
在又一方面,本发明提供一种药物组合物,其包含:In yet another aspect, the present invention provides a pharmaceutical composition comprising:
(ⅰ)本发明的融合蛋白,以及(i) fusion protein of the present invention, and
(ⅱ)药学上可接受的载剂。(ii) A pharmaceutically acceptable carrier.
在另一方面,本发明还涉及所述融合蛋白、多核苷酸、缀合物或者药物组合物的用途。In another aspect, the present invention also relates to the use of said fusion protein, polynucleotide, conjugate or pharmaceutical composition.
附图说明Description of drawings
图1:通过还原性蛋白凝胶电泳(SDS-PAGE)分析融合蛋白C15和C2。使用蛋白标准品指示的分子量(kDa)在左侧示出。泳道1:C15;泳道2:C2。Figure 1: Analysis of fusion proteins C15 and C2 by reducing protein gel electrophoresis (SDS-PAGE). Molecular weights (kDa) indicated using protein standards are shown on the left. Lane 1: C15; Lane 2: C2.
图2:通过ELISA分析的B22、C15、C12和C2对TGF-β的结合活性。Figure 2: Binding activity of B22, C15, C12 and C2 to TGF-β analyzed by ELISA.
图3:通过Fortebio分析的C12、336B11和36E5对hGITR的体外结合亲和力和动力学。Figure 3: In vitro binding affinity and kinetics of C12, 336B11 and 36E5 to hGITR analyzed by Fortebio.
图4:通过FACS分析的C12和C15体外结合CHOS-hGITR细胞的活性。Figure 4: In vitro binding activity of C12 and C15 to CHOS-hGITR cells analyzed by FACS.
图5:通过GITR Blockade Bioassay分析的C12、C15和36E5体外激活GITR的活性。IgG同种型(IgG Isotype)用作阴性对照。Figure 5: In vitro activation of GITR by C12, C15 and 36E5 analyzed by GITR Blockade Bioassay. IgG Isotype was used as negative control.
图6:通过TGF-β受体报告基因实验分析的C12和抗TGF-β抗体BMK-R2体外阻断TGF-β的活性。人IgG1用作阴性对照。Figure 6: In vitro blocking of TGF-β activity by C12 and anti-TGF-β antibody BMK-R2 analyzed by TGF-β receptor reporter assay. Human IgG1 was used as a negative control.
图7:通过ELISA分析的C12、336B11以及36E5体外激活T细胞的活性。蛋白样品:336B11、C12、IgG Isotype(IgG同种型,阴性对照)和36E5。对于每组样品,蛋白浓度从左至右:10、1、0.1μg/ml。medium(培养基)作为空白对照。Figure 7: The activities of C12, 336B11 and 36E5 to activate T cells in vitro analyzed by ELISA. Protein samples: 336B11, C12, IgG Isotype (IgG isotype, negative control) and 36E5. For each group of samples, the protein concentration is from left to right: 10, 1, 0.1 μg/ml. medium (culture medium) was used as a blank control.
图8:C12、C15以及36E5体外激活T细胞的活性。蛋白样品:C12、C15、36E5和IgG Isotype(IgG同种型,阴性对照)。对于每组样品,蛋白浓度从左至右:50、10、2、0.4、0.08μg/ml。Medium(培养基)作为空白对照。Figure 8: The activities of C12, C15 and 36E5 to activate T cells in vitro. Protein samples: C12, C15, 36E5 and IgG Isotype (IgG isotype, negative control). For each group of samples, protein concentration from left to right: 50, 10, 2, 0.4, 0.08 μg/ml. Medium (culture medium) was used as a blank control.
图9:在鼠源结肠癌MC38-OVA模型中,经对照和融合蛋白C12处理的小鼠肿瘤体积的生长曲线。注:肿瘤体积表示为“平均值±标准误差(SEM)”。Figure 9: Growth curves of tumor volume in mice treated with control and fusion protein C12 in the murine colon cancer MC38-OVA model. NOTE: Tumor volumes are expressed as "mean ± standard error (SEM)".
具体实施方式detailed description
定义definition
在本发明中,除非另有说明,否则本文中使用的科学和技术名词具有本领域技术人员所通常理解的含义。并且,本文中所用的蛋白质和核酸化学、分子生物学、细胞和组织培养、微生物学、免疫学相关术语和实验室操作步骤均为相应领域内广泛使用的术语和常规步骤。同时,为了更好地理解本发明,下面提供相关术语的定义和解释。In the present invention, unless otherwise specified, the scientific and technical terms used herein have the meanings commonly understood by those skilled in the art. Moreover, the terms related to protein and nucleic acid chemistry, molecular biology, cell and tissue culture, microbiology, immunology and laboratory operation steps used herein are all terms and routine procedures widely used in the corresponding fields. Meanwhile, in order to better understand the present invention, definitions and explanations of related terms are provided below.
如本文所用,“至少一个(种)”或“一个(种)或多个(种)”可以表示1、2、3、4、5、6、7、8个(种)或更多个(种)。As used herein, "at least one (species)" or "one (species) or more (species)" may mean 1, 2, 3, 4, 5, 6, 7, 8 (species) or more ( kind).
如本文所用,术语“约”、“大约”当与数值变量联合使用时,通常指该变量的数值和该变量的所有数值在实验误差内(例如对于平均值95%的置信区间内)或在指定数值的±10%内,或更宽范围内。As used herein, the terms "about" and "approximately" when used in conjunction with a numerical variable generally mean that the value of the variable and all values of the variable are within experimental error (e.g., within a 95% confidence interval for the mean) or within Within ±10% of the specified value, or within a wider range.
如本文所用,表述“包含”或与其同义的类似表述“包括”、“含有”和“具有”等是开放性的,不排除额外的未列举的元素、步骤或成分。表述“由…组成”排除未指明的任何元素、步骤或成分。表述“基本上由…组成”指范围限制在指定的元素、步骤或成分,加上任选存在的不会实质上影响所要求保护的主题的基本和新的特征的元素、步骤或成分。应当理解,表述“包含”涵盖表述“基本上由…组成”和“由…组成”。As used herein, the expression "comprises" or its synonymous similar expressions "comprising", "containing" and "having", etc. are open-ended and do not exclude additional unrecited elements, steps or components. The expression "consisting of" excludes any element, step or ingredient not specified. The expression "consisting essentially of" means limiting the scope to the specified elements, steps or ingredients, plus the optional presence of elements, steps or ingredients that do not materially affect the basic and novel characteristics of the claimed subject matter. It should be understood that the expression "comprising" encompasses the expressions "consisting essentially of" and "consisting of".
如本文所用,术语“任选”或“任选地”是指随后描述的事件或情况可能发生或可能不发生,该描述包括发生所述事件或情况和不发生所述事件或情况。例如,融合蛋白任选地包含接头涵盖包含或者不包含接头的情况。As used herein, the term "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that description includes that said event or circumstance occurs and that it does not. For example, a fusion protein optionally comprising a linker encompasses the inclusion or absence of a linker.
如本文所用,术语“多肽”指包含至少两个通过肽键连接的氨基酸或其衍生物的聚合物。在本文中,术语“多肽”和“蛋白”通常可以互换使用。在一些实施方案中,蛋白可以由一条或更多条多肽以共价或非共价方式形成,例如二聚体蛋白。As used herein, the term "polypeptide" refers to a polymer comprising at least two amino acids or derivatives thereof linked by peptide bonds. The terms "polypeptide" and "protein" are often used interchangeably herein. In some embodiments, proteins can be formed from one or more polypeptides covalently or non-covalently, eg, dimeric proteins.
在本文中,当指定氨基酸在多肽中的位置时,第n位氨基酸指从多肽的氨基端(N端)的第1个氨基酸开始数的第n个氨基酸。Herein, when specifying the position of an amino acid in a polypeptide, the n-th amino acid refers to the n-th amino acid counted from the 1st amino acid at the amino terminal (N-terminal) of the polypeptide.
在本文中,氨基端(N端)和羧基端(C端)可以表示多肽中两个或多个氨基酸序列的相对位置,并不表示所述两个或多个氨基酸序列紧邻,例如在本发明的融合蛋白中,第一多肽可以位于第二多肽的N端,第二多肽可以位于第三多肽的N端,并且第一多肽与第二多肽之间和/或第二多肽与第三多肽之间可以包含其他的组分(例如氨基酸序列,例如接头)。或者,氨基端(N端)和羧基端(C端)也可以表示指定氨基酸或氨基酸序列在其所处于的多肽中的位置,例如本发明的第一多肽可以位于本发明融合蛋白的N端也可以位于本发明融合蛋白的C端。Herein, the amino terminus (N terminus) and the carboxyl terminus (C terminus) may indicate the relative positions of two or more amino acid sequences in a polypeptide, and do not mean that the two or more amino acid sequences are in close proximity, for example, in the present invention In the fusion protein, the first polypeptide can be located at the N-terminus of the second polypeptide, the second polypeptide can be located at the N-terminus of the third polypeptide, and between the first polypeptide and the second polypeptide and/or the second Additional components (eg, amino acid sequences, eg, linkers) may be included between the polypeptide and the third polypeptide. Alternatively, the amino terminus (N terminus) and the carboxyl terminus (C terminus) can also indicate the position of the specified amino acid or amino acid sequence in the polypeptide in which it is located, for example, the first polypeptide of the present invention can be located at the N terminus of the fusion protein of the present invention It can also be located at the C-terminus of the fusion protein of the present invention.
如本文所用,术语“融合蛋白”或“融合多肽”指包含共价连接的至少两个多肽的蛋白,所述至少两个多肽在自然环境下通常不共价连接。可以通过化学、酶促或重组DNA技 术使至少两个多肽共价连接形成融合蛋白。As used herein, the term "fusion protein" or "fusion polypeptide" refers to a protein comprising at least two polypeptides covalently linked that are not normally covalently linked in nature. Fusion proteins can be formed by covalently linking at least two polypeptides by chemical, enzymatic or recombinant DNA techniques.
如本文所用,术语“突变”是指多肽中包含一个或多个氨基酸的取代、缺失或添加。所述取代可以是保守或非保守取代。在多肽或蛋白中,合适的保守氨基酸取代是本领域技术人员已知的,并且一般可以进行保守氨基酸取代而不改变所得分子的生物活性。通常,本领域技术人员认识到多肽的非必需区中的单个氨基酸取代基本上不改变期望的生物活性(参见,例如Watson et al.,Molecular Biology of the Gene,4th Edition,1987,The Benjamin/Cummings Pub.co.,p.224)。例如,Fc片段可以包含突变,例如删除CH3结构域中C端的赖氨酸。As used herein, the term "mutation" refers to a substitution, deletion or addition comprising one or more amino acids in a polypeptide. The substitutions may be conservative or non-conservative. In polypeptides or proteins, suitable conservative amino acid substitutions are known to those skilled in the art, and conservative amino acid substitutions can generally be made without altering the biological activity of the resulting molecule. Generally, those skilled in the art recognize that single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter the desired biological activity (see, e.g., Watson et al., Molecular Biology of the Gene, 4th Edition, 1987, The Benjamin/Cummings Pub.co., p.224). For example, the Fc fragment may contain mutations such as deletion of the C-terminal lysine in the CH3 domain.
如本文所用,“野生型”多肽或蛋白是指天然存在的,未经人为改造的多肽或蛋白。多肽或蛋白的“突变型”或“突变体”相对于“野生型”的多肽或蛋白带有突变。例如,野生型TGF-βRⅡ胞外结构域包含SEQ ID NO:4的序列。TGF-βRⅡ胞外结构域突变体相对于SEQ ID NO:4带有突变。As used herein, a "wild-type" polypeptide or protein refers to a naturally occurring polypeptide or protein that has not been artificially modified. A "mutant" or "mutant" of a polypeptide or protein carries a mutation relative to a "wild-type" polypeptide or protein. For example, the wild-type TGF-βRII extracellular domain comprises the sequence of SEQ ID NO:4. TGF-βRII ectodomain mutants carry mutations relative to SEQ ID NO:4.
如本文所用,术语“多核苷酸”或“核酸”指包含至少两个连接的核苷酸或核苷酸衍生物的寡聚体或聚合物,包括通常通过磷酸二酯键连接在一起的脱氧核糖核酸(DNA)和核糖核酸(RNA)。As used herein, the term "polynucleotide" or "nucleic acid" refers to an oligomer or polymer comprising at least two linked nucleotides or nucleotide derivatives, including deoxygenated nucleotides, usually linked together by phosphodiester bonds. Ribonucleic acid (DNA) and ribonucleic acid (RNA).
本发明的融合蛋白或多核苷酸可以是“分离的”。表述“分离的”指多肽或多核苷酸可以例如是经过人工改造的,和/或与其天然存在的环境中的其他物质分离的。诸如cDNA分子的“分离的”多核苷酸可以在通过重组技术制备时基本上不含其他细胞物质或培养基,或者在化学合成时基本上不含化学前体或其他化学成分。A fusion protein or polynucleotide of the invention may be "isolated". The expression "isolated" means that the polypeptide or polynucleotide may, for example, be artificially engineered, and/or separated from other substances in its naturally occurring environment. An "isolated" polynucleotide, such as a cDNA molecule, may be substantially free of other cellular material or culture medium when prepared by recombinant techniques, or substantially free of chemical precursors or other chemical components when chemically synthesized.
如本文所用,“载体”是用于将外源核酸导入宿主细胞的媒介,当载体转化入适当的宿主细胞时,外源核酸得以扩增或表达。载体包括那些通常通过限制酶切消化和连接可以将编码多肽或其片段的核酸引入其中的载体。载体还包括那些包含编码多肽的核酸的载体。载体通常保持游离,但是可以设计为使基因或其部分整合入基因组的染色体。还考虑人工染色体的载体,例如酵母人工载体和哺乳动物人工染色体。如本文所用,载体的定义涵盖质粒、线性化质粒、病毒载体、粘粒、噬菌体载体、噬菌粒、人工染色体(例如,酵母人工染色体和哺乳动物人工染色体)等。如本文所用,载体可以在宿主细胞中表达或复制。As used herein, a "vector" is a vehicle for introducing exogenous nucleic acid into a host cell, and when the vector is transformed into an appropriate host cell, the exogenous nucleic acid is amplified or expressed. Vectors include those into which nucleic acids encoding polypeptides or fragments thereof can be introduced, typically by restriction digestion and ligation. Vectors also include those comprising nucleic acid encoding a polypeptide. Vectors usually remain episomal, but can be designed to allow integration of a gene, or part thereof, into the chromosome of the genome. Also contemplated are vectors for artificial chromosomes, such as yeast artificial vectors and mammalian artificial chromosomes. As used herein, the definition of vector encompasses plasmids, linearized plasmids, viral vectors, cosmids, phage vectors, phagemids, artificial chromosomes (eg, yeast artificial chromosomes and mammalian artificial chromosomes), and the like. As used herein, a vector can be expressed or replicated in a host cell.
如本文所用,“表达”指通过多核苷酸的转录和翻译产生多肽的过程。“表达载体”包括能够表达多肽的载体,其包含编码目标多肽的多核苷酸序列。编码目标多肽的多核苷酸序列与能够影响其表达的调控序列可操作地连接。这类调控序列可以包括启动子和终止子序列,并且任选地可以包括一个或多个复制起点、一个或多个选择标记、增强子、多腺苷酸化信号等。表达载体一般来源于质粒或病毒DNA,或者可以包含这两者的元件。因此,表达载体可以指重组DNA或RNA构建体,例如质粒、噬菌体载体、 重组病毒或其他载体,当引入适当的宿主细胞时,导致克隆DNA或RNA的表达。适当的表达载体是本领域技术人员公知的,并且包括在真核细胞和/或原核细胞中可复制的表达载体以及保持游离的表达载体或者整合入宿主细胞基因组的表达载体。As used herein, "expression" refers to the process by which a polypeptide is produced by the transcription and translation of a polynucleotide. An "expression vector" includes a vector capable of expressing a polypeptide comprising a polynucleotide sequence encoding a polypeptide of interest. The polynucleotide sequence encoding the polypeptide of interest is operably linked to regulatory sequences capable of affecting its expression. Such regulatory sequences may include promoter and terminator sequences, and optionally may include one or more origins of replication, one or more selectable markers, enhancers, polyadenylation signals, and the like. Expression vectors are generally derived from plasmid or viral DNA, or may contain elements of both. Thus, an expression vector may refer to a recombinant DNA or RNA construct, such as a plasmid, phage vector, recombinant virus or other vector, which, when introduced into an appropriate host cell, results in the expression of cloned DNA or RNA. Appropriate expression vectors are well known to those skilled in the art and include expression vectors replicable in eukaryotic cells and/or prokaryotic cells as well as expression vectors that remain episomal or integrate into the host cell genome.
如本文所用,“宿主细胞”是用于接受、保持、复制或扩增载体的细胞。宿主细胞还可以用来表达核酸或载体所编码的多肽。宿主细胞可以是真核细胞或原核细胞。合适的宿主细胞包括但不限于CHO细胞、COS细胞、HeLa细胞和HEK细胞(例如HEK293细胞)。As used herein, a "host cell" is a cell used to receive, maintain, replicate or amplify a vector. Host cells can also be used to express nucleic acids or polypeptides encoded by vectors. Host cells can be eukaryotic or prokaryotic. Suitable host cells include, but are not limited to, CHO cells, COS cells, HeLa cells, and HEK cells (eg, HEK293 cells).
如本文所用,“亲和力”或“结合亲和力”用来衡量分子与其配体之间通过非共价作用力相互结合的强度,例如本发明的融合蛋白与其靶点之间的结合强度,例如GITR与GITRL胞外结构域之间或者TGFBR2-ECD与TGF-β之间的结合强度。亲和力的大小通常报告为平衡解离常数K D,常通过测量结合速率常数(k on)和解离速率常数(k dis)并计算k dis除以k on的商来确定(K D=k dis/k on)。K D可以利用常规技术容易地测定,例如可以使用的方法包括但不限于:平衡透析法;生物膜层干涉技术,例如通过Octet RED96检测系统;酶联免疫吸附测定法(ELISA);表面等离子体共振(SPR)法;或者通过技术人员已知的其他方法。 As used herein, "affinity" or "binding affinity" is used to measure the strength of the binding between a molecule and its ligand through non-covalent forces, such as the binding strength between the fusion protein of the present invention and its target, such as GITR and Binding strength between extracellular domains of GITRL or between TGFBR2-ECD and TGF-β. The magnitude of affinity is usually reported as the equilibrium dissociation constant, K D , which is often determined by measuring the association rate constant (k on ) and the dissociation rate constant (k dis ) and calculating the quotient of k dis divided by k on (K D = k dis / k on ). KD can be easily determined using conventional techniques, for example, methods that can be used include but are not limited to: equilibrium dialysis; biofilm layer interferometry, for example by Octet RED96 detection system; enzyme-linked immunosorbent assay (ELISA); surface plasmon Resonance (SPR) method; or by other methods known to the skilled person.
“特异性结合”是指两个分子之间以较高的亲和力相互结合,通常,特异性结合的两个分子之间的K D值可以为10 -6、10 -7、10 -8到10 -9M或更低。例如,GITR与GITRL胞外结构域之间可以以10 -6到10 -9M或更低的K D值特异性结合,TGFBR2-ECD与TGF-β之间可以以10 -6到10 -9M或更低的K D值特异性结合。 "Specific binding" means that two molecules bind to each other with a relatively high affinity. Usually, the K D value between two molecules that specifically binds can be 10 -6 , 10 -7 , 10 -8 to 10 -9 M or lower. For example, the K D value between GITR and the extracellular domain of GITRL can be 10 -6 to 10 -9 M or lower, and the K D value between TGFBR2-ECD and TGF-β can be 10 -6 to 10 -9 A KD value of M or lower binds specifically.
术语“治疗”是指防止、治愈、改善、减缓、阻滞或部分阻滞疾病或病症的症状。因此,需要治疗的受试者包括但不限于:患有疾病或病症者;有患病风险者;以及欲预防体内的病症者。在癌症或肿瘤的情况下,若受试者显示出下列的一或多项,则受试者成功地被根据本发明的方法“治疗”:免疫反应增加、抗肿瘤反应增加、免疫细胞的溶细胞活性增加、免疫细胞杀死肿瘤细胞增加、癌细胞数目减少或完全不存在;肿瘤尺寸缩小;癌细胞浸润到周围器官中(包括癌细胞扩散入软组织和骨骼中)受抑制或不存在;肿瘤或癌细胞转移受抑制或不存在;癌症生长受抑制或不存在;与特定癌症相关的一种或多种症状缓解;发病率和死亡率降低;生活质量提升;致瘤性降低;癌干细胞的数目或频率减少;或某些效果的组合。The term "treating" means preventing, curing, ameliorating, alleviating, arresting or partially arresting the symptoms of a disease or disorder. Thus, subjects in need of treatment include, but are not limited to: those with a disease or condition; those at risk of having a disease; and those in whom the condition is to be prevented. In the case of cancer or tumors, a subject is successfully "treated" according to the methods of the invention if the subject exhibits one or more of the following: increased immune response, increased anti-tumor response, lysis of immune cells Increased cellular activity, increased killing of tumor cells by immune cells, decreased number or absence of cancer cells; reduced tumor size; suppressed or absent cancer cell infiltration into surrounding organs (including spread of cancer cells into soft tissue and bone); tumor or inhibition of cancer cell metastasis or absence; inhibition or absence of cancer growth; relief of one or more symptoms associated with a particular cancer; reduction in morbidity and mortality; improved quality of life; reduction in tumorigenicity; A decrease in number or frequency; or some combination of effects.
如本文所用,“治疗有效量”或“治疗有效剂量”指施用于受试者之后至少足以产生疗效的物质、化合物、材料或组合物的量。因此,其为防止、治愈、改善、阻滞或部分阻滞疾病或病症的症状所必需的量。例如,对于肿瘤的治疗,相对于未接受治疗的受试者,治疗有效量的本发明的融合蛋白或药物组合物优选地将细胞生长或肿瘤生长抑制至少约10%、至少约20%、至少约30%、至少约40%、至少约50%、至少约 60%、至少约70%,优选至少约80%。抑制肿瘤生长的作用可以在预测对人类肿瘤的疗效的动物模型系统中评价;或者,也可以通过检查抑制细胞生长的能力来评价,这种抑制可以通过本领域技术人员公知的试验在体外测定。治疗有效量的本发明的融合蛋白或药物组合物能够减小肿瘤大小,或者以其他方式缓解受试者的症状如预防和/或治疗转移或复发。本领域技术人员理解,治疗有效量受多种因素影响,例如受试者的体重、症状的严重性和选择的特定组合物或给药途径。可以在一次或多次施用中给予治疗有效量。As used herein, "therapeutically effective amount" or "therapeutically effective dose" refers to the amount of a substance, compound, material or composition which is at least sufficient to produce a therapeutic effect after administration to a subject. Thus, it is the amount necessary to prevent, cure, ameliorate, arrest or partially arrest the symptoms of a disease or disorder. For example, for the treatment of tumors, a therapeutically effective amount of a fusion protein or pharmaceutical composition of the invention preferably inhibits cell growth or tumor growth by at least about 10%, at least about 20%, at least About 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, preferably at least about 80%. The effect of inhibiting tumor growth can be evaluated in animal model systems predictive of therapeutic effect on human tumors; alternatively, it can also be evaluated by examining the ability to inhibit cell growth, which can be determined in vitro by assays well known to those skilled in the art. A therapeutically effective amount of the fusion protein or pharmaceutical composition of the present invention can reduce tumor size, or alleviate symptoms of a subject in other ways such as preventing and/or treating metastasis or recurrence. Those skilled in the art understand that a therapeutically effective amount will be influenced by factors such as the subject's weight, severity of symptoms and the particular composition or route of administration chosen. A therapeutically effective amount can be administered in one or more administrations.
融合蛋白fusion protein
在一方面,本发明提供一种融合蛋白,其从N端至C端包含:In one aspect, the invention provides a kind of fusion protein, it comprises from N-terminus to C-terminus:
第一多肽–第二多肽–第三多肽    式(1);The first polypeptide-the second polypeptide-the third polypeptide Formula (1);
或者or
第三多肽–第二多肽–第一多肽   式(2);The third polypeptide-the second polypeptide-the first polypeptide formula (2);
其中所述第一多肽包含TGF-βRⅡ胞外结构域,所述第二多肽包含二聚化结构域,并且所述第三多肽包含3个GITRL胞外结构域。Wherein the first polypeptide comprises a TGF-βRII extracellular domain, the second polypeptide comprises a dimerization domain, and the third polypeptide comprises three GITRL extracellular domains.
本发明的融合蛋白是GITR/TGF-β双靶向融合蛋白,其同时特异性结合TGF-β和GITR。本发明的融合蛋白通过结合TGF-β抑制TGF-β信号通路,起到TGF-β抑制剂的作用;同时本发明的融合蛋白通过结合GITR激活GITR信号通路,起到GITR激动剂的作用。本发明的融合蛋白可以作为免疫治疗剂来诱导、促进、增强、激活或延长免疫反应,特别是诱导、促进、增强、激活或延长针对肿瘤细胞的免疫反应。The fusion protein of the present invention is a GITR/TGF-β double-targeting fusion protein, which specifically binds TGF-β and GITR at the same time. The fusion protein of the present invention inhibits the TGF-β signaling pathway by binding to TGF-β, and acts as a TGF-β inhibitor; meanwhile, the fusion protein of the present invention activates the GITR signaling pathway by binding to GITR, and acts as a GITR agonist. The fusion protein of the present invention can be used as an immunotherapeutic agent to induce, promote, enhance, activate or prolong immune response, especially induce, promote, enhance, activate or prolong immune response against tumor cells.
第一多肽first polypeptide
在一些实施方案中,第一多肽包含TGF-βRⅡ胞外结构域(TGFBR2-ECD)。In some embodiments, the first polypeptide comprises a TGF-βRII extracellular domain (TGFBR2-ECD).
如本文所用,术语“TGF-βRⅡ胞外结构域”或“TGFBR2-ECD”是指TGF-βⅡ型(TGF-βRⅡ)受体的胞外区,其具有结合TGF-β的活性。二聚化TGFBR2-ECD可以作为TGF-βTrap结合和捕获TGF-β二聚体,从而抑制TGF-β信号通路以及消除TGF-β信号通路相关的免疫抑制。可以使用本领域已知的各种检测方法确定TGF-β信号通路的抑制,例如检测TGF-β信号通路控制的下游基因的表达。As used herein, the term "TGF-βRII extracellular domain" or "TGFBR2-ECD" refers to the extracellular region of the TGF-β type II (TGF-βRII) receptor, which has TGF-β binding activity. Dimerized TGFBR2-ECD can act as a TGF-βTrap to bind and capture TGF-β dimers, thereby inhibiting TGF-β signaling pathway and eliminating TGF-β signaling pathway-related immunosuppression. Inhibition of the TGF-β signaling pathway can be determined using various detection methods known in the art, such as detecting the expression of downstream genes controlled by the TGF-β signaling pathway.
第一多肽中包含的TGFBR2-ECD可以是野生型或其突变体。关于野生型TGFBR2-ECD描述可以参见例如WO2015118175A2。示例性野生型TGFBR2-ECD的氨基酸序列如SEQ ID NO:4所示。TGFBR2-ECD contained in the first polypeptide may be wild type or a mutant thereof. For the description of wild-type TGFBR2-ECD, see, for example, WO2015118175A2. The amino acid sequence of an exemplary wild-type TGFBR2-ECD is shown in SEQ ID NO:4.
发明人发现,野生型TGFBR2-ECD容易在N端第1-24位氨基酸中发生断裂,不利于后续的成药性开发。为解决这一问题,发明人开发了TGFBR2-ECD突变体,其为相对于野生型TGFBR2-ECD的改进型多肽。相比于包含野生型TGFBR2-ECD的融合 蛋白,包含TGFBR2-ECD突变体的融合蛋白在生产过程中不易产生降解,具有更好的成药性。The inventors found that the wild-type TGFBR2-ECD is prone to breakage in amino acids 1-24 of the N-terminus, which is not conducive to the subsequent development of druggability. To solve this problem, the inventors developed TGFBR2-ECD mutants, which are improved polypeptides relative to wild-type TGFBR2-ECD. Compared with the fusion protein comprising the wild-type TGFBR2-ECD, the fusion protein comprising the TGFBR2-ECD mutant is less likely to be degraded during the production process and has better druggability.
在一实施方案中,所述TGFBR2-ECD突变体包含SEQ ID NO:5或SEQ ID NO:6的氨基酸序列。In one embodiment, the TGFBR2-ECD mutant comprises the amino acid sequence of SEQ ID NO:5 or SEQ ID NO:6.
在一些实施方案中,第一多肽包含野生型TGFBR2-ECD。在一具体实施方案中,所述野生型TGFBR2-ECD包含SEQ ID NO:4的氨基酸序列。In some embodiments, the first polypeptide comprises wild-type TGFBR2-ECD. In a specific embodiment, the wild-type TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4.
在一些实施方案中,第一多肽包含TGFBR2-ECD突变体。在一具体实施方案中,所述TGFBR2-ECD突变体包含SEQ ID NO:5或SEQ ID NO:6的氨基酸序列。In some embodiments, the first polypeptide comprises a TGFBR2-ECD mutant. In a specific embodiment, the TGFBR2-ECD mutant comprises the amino acid sequence of SEQ ID NO:5 or SEQ ID NO:6.
在一具体实施方案中,第一多肽包含TGFBR2-ECD,其包含SEQ ID NO:4的氨基酸序列。在又一具体实施方案中,第一多肽包含TGFBR2-ECD,其包含SEQ ID NO:5的氨基酸序列。在又一具体实施方案中,第一多肽包含TGFBR2-ECD,其包含SEQ ID NO:6的氨基酸序列。In a specific embodiment, the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:4. In yet another specific embodiment, the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:5. In yet another specific embodiment, the first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:6.
第二多肽second polypeptide
如本文所用,“二聚化结构域”是指能够促进两条或更多条多肽通过共价或非共价作用组合成二聚体或多聚体(例如三、四、五、六、七、八或九聚体)的多肽。据信,二聚化的TGF-βRⅡ胞外结构域可以作为TGF-βTrap(参见例如WO2015118175A2和WO2018205985A1)结合TGF-β二聚体并抑制TGF-β信号通路。二聚化结构域促进本发明的融合蛋白形成二聚体或多聚体,从而本发明的融合蛋白可以作为TGF-βTrap发挥作用。As used herein, "dimerization domain" refers to a domain capable of facilitating the combination of two or more polypeptides into dimers or multimers (e.g., three, four, five, six, seven) through covalent or non-covalent interactions. , octa or nine-mer) polypeptides. It is believed that the dimerized TGF-βRII extracellular domain can act as a TGF-β Trap (see eg WO2015118175A2 and WO2018205985A1 ) to bind TGF-β dimers and inhibit TGF-β signaling pathway. The dimerization domain promotes the fusion protein of the present invention to form dimers or multimers, so that the fusion protein of the present invention can function as TGF-βTrap.
二聚化结构域的非限制性实例可以包括免疫球蛋白IgG、IgA、IgD的重链恒定区3(CH3)以及IgM和IgE的重链恒定区4(CH4)。二聚化结构域可以进一步包含特定类型的免疫球蛋白的其他结构域,例如IgG1、IgG2、IgG3或IgG4的铰链区或其部分和/或重链恒定区2(CH2),以获得具有期望的特性的融合蛋白,例如增强抗体依赖的细胞毒作用(ADCC)、抗体依赖的细胞胞吞作用(ADCP)、补体依赖的细胞毒性作用(CDC)以及和FcRn结合的结合活性以延长融合蛋白的半衰期等。Non-limiting examples of dimerization domains may include the heavy chain constant region 3 (CH3) of immunoglobulins IgG, IgA, IgD, and the heavy chain constant region 4 (CH4) of IgM and IgE. The dimerization domain may further comprise other domains of a particular type of immunoglobulin, such as the hinge region or part thereof and/or the heavy chain constant region 2 (CH2) of IgG1, IgG2, IgG3 or IgG4, to obtain a protein having the desired Specific fusion proteins, such as enhancing antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular endocytosis (ADCP), complement-dependent cytotoxicity (CDC), and binding activity to FcRn to prolong the half-life of fusion proteins Wait.
在一实施方案中,二聚化结构域包含免疫球蛋白的CH2和CH3结构域,优选人IgG1的CH2和CH3结构域。In one embodiment, the dimerization domain comprises the CH2 and CH3 domains of an immunoglobulin, preferably the CH2 and CH3 domains of a human IgGl.
在优选的实施方案中,二聚化结构域进一步包含免疫球蛋白的铰链区或其部分。优选地,所述铰链区为人IgG1的铰链区。在一具体实施方案中,二聚化结构域包含免疫球蛋白的Fc片段,优选人IgG1的Fc片段。In preferred embodiments, the dimerization domain further comprises an immunoglobulin hinge region or portion thereof. Preferably, the hinge region is the hinge region of human IgG1. In a specific embodiment, the dimerization domain comprises the Fc fragment of an immunoglobulin, preferably the Fc fragment of human IgGl.
如本文所用,“免疫球蛋白Fc片段”或“Fc片段”是指免疫球蛋白的铰链区或其部分以及重链恒定区CH2、CH3、CH4中的两个或更多结构域。例如,IgG的Fc片段可以包含免疫球蛋白的铰链区或其部分以及CH2和CH3结构域,而IgM和IgE的Fc片段 可以包含免疫球蛋白的铰链区或其部分、CH2、CH3和CH4结构域。Fc片段可以衍生自任何免疫球蛋白,例如,IgG、IgM、IgD、IgE、IgA和IgY中的任何类别或亚类,例如IgG1、IgG2、IgG3、IgG4、IgA1、IgA2、IgG2a和IgG2b。Fc片段中的各部分(例如铰链区、CH2和CH3)可以衍生自相同或不同的免疫球蛋白。优选地,Fc片段中的各部分衍生自相同的免疫球蛋白,优选人IgG,例如人IgG1、IgG2、IgG3或IgG4。Fc片段可以是通过酶促处理全长抗体所产生的抗体衍生物(例如木瓜蛋白酶切割后的非抗原结合部分),以及通过化学合成或基因工程技术(例如DNA重组技术)产生的衍生物。As used herein, "immunoglobulin Fc fragment" or "Fc fragment" refers to the hinge region or part thereof and two or more domains in the heavy chain constant region CH2, CH3, CH4 of an immunoglobulin. For example, the Fc fragment of IgG may comprise the hinge region or part thereof and the CH2 and CH3 domains of an immunoglobulin, while the Fc fragments of IgM and IgE may comprise the hinge region or part thereof, CH2, CH3 and CH4 domains of an immunoglobulin . The Fc fragment can be derived from any immunoglobulin, e.g., any class or subclass of IgG, IgM, IgD, IgE, IgA, and IgY, e.g., IgGl, IgG2, IgG3, IgG4, IgAl, IgA2, IgG2a, and IgG2b. Portions in the Fc fragment (eg, hinge region, CH2 and CH3) may be derived from the same or different immunoglobulins. Preferably, each portion in the Fc fragment is derived from the same immunoglobulin, preferably human IgG, eg human IgGl, IgG2, IgG3 or IgG4. Fc fragments can be antibody derivatives produced by enzymatic treatment of full-length antibodies (such as non-antigen-binding parts after papain cleavage), and derivatives produced by chemical synthesis or genetic engineering techniques (such as DNA recombinant technology).
本领域技术人员可以根据已知的算法和软件判断CH2、CH3、CH4和铰链区在免疫蛋白中的位置和氨基酸序列,可以应用的算法和软件的描述可以参见例如William R.Strohl,Lila M.Strohl,(2012),Antibody structure–function relationships,In Woodhead Publishing Series in Biomedicine,Therapeutic Antibody Engineering,Woodhead Publishing,pp.37-56。可以通过本领域技术人员熟知的方法获得各种类型免疫球蛋白Fc片段的氨基酸序列,例如使用公共数据库(例如Uniprot或NCBI)获得免疫球蛋白重链恒定区各个部分的氨基酸序列。并且,本领域技术人员知晓,可以根据需要对Fc片段进行修饰。例如可以在铰链区、CH2和/或CH3结构域中添加、取代或缺失一个或多个氨基酸,例如修饰CH2以改变Fc片段的糖基化修饰,或者在铰链区中增加或缺失用于形成分子间二硫键的一个或多个半胱氨酸。Those skilled in the art can judge the position and amino acid sequence of CH2, CH3, CH4 and hinge regions in the immune protein according to known algorithms and software, and the description of applicable algorithms and software can be found in, for example, William R. Strohl, Lila M. Strohl, (2012), Antibody structure–function relationships, In Woodhead Publishing Series in Biomedicine, Therapeutic Antibody Engineering, Woodhead Publishing, pp.37-56. The amino acid sequences of various types of immunoglobulin Fc fragments can be obtained by methods well known to those skilled in the art, for example, using public databases (eg Uniprot or NCBI) to obtain the amino acid sequences of various parts of the immunoglobulin heavy chain constant region. Moreover, those skilled in the art know that the Fc fragment can be modified as needed. For example, one or more amino acids may be added, substituted or deleted in the hinge region, CH2 and/or CH3 domains, for example modifying CH2 to alter the glycosylation modification of the Fc fragment, or additions or deletions in the hinge region for formation of molecules One or more cysteines between disulfide bonds.
在其他实施方案中,第二多肽进一步包含融合至所述二聚化结构域N端和/或C端的接头。所述接头如本文所述。In other embodiments, the second polypeptide further comprises a linker fused to the N-terminus and/or C-terminus of said dimerization domain. The linker is as described herein.
在一些实施方案中,第二多肽从N端至C端具有如下结构:In some embodiments, the second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 –Dd–Lk 2 ,
其中in
Dd为二聚化结构域;Dd is a dimerization domain;
Lk 1和Lk 2各自独立地为接头或不存在。 Lk 1 and Lk 2 are each independently a linker or absent.
在一些实施方案中,二聚化结构域包含CH2、CH3、CH4结构域或其组合。在一些实施方案中,二聚化结构域包含至少一个CH3结构域。在一些实施方案中,二聚化结构域从N端至C端包含CH2和CH3结构域,优选人IgG1的CH2和CH3结构域。在一些实施方案中,所述CH2和CH3结构域衍生自人IgG1。在一些实施方案中,二聚化结构域从N端至C端包含衍生自人IgG1的CH2和CH3结构域。在一实施方案中,所述CH3结构域的C端赖氨酸缺失。在一实施方案中,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列。在一实施方案中,所述CH2和CH3结构域包含SEQ ID NO:8的氨基酸序列。In some embodiments, the dimerization domain comprises a CH2, CH3, CH4 domain, or a combination thereof. In some embodiments, the dimerization domain comprises at least one CH3 domain. In some embodiments, the dimerization domain comprises CH2 and CH3 domains from N-terminus to C-terminus, preferably the CH2 and CH3 domains of human IgGl. In some embodiments, the CH2 and CH3 domains are derived from human IgGl. In some embodiments, the dimerization domain comprises, from N-terminus to C-terminus, CH2 and CH3 domains derived from human IgGl. In one embodiment, the C-terminal lysine of the CH3 domain is deleted. In one embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In one embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:8.
在一些实施方案中,二聚化结构域包含免疫球蛋白的铰链区或其部分以及CH2、 CH3、CH4结构域或其组合。在一些实施方案中,二聚化结构域从N端至C端包含免疫球蛋白的铰链区或其部分、CH2和CH3结构域,优选人IgG1的Fc片段。所述CH2和CH3结构域如上所述。在一些实施方案中,所述铰链区衍生自人IgG1。在一些实施方案中,所述铰链区不包含一个或多个半胱甘酸。在一实施方案中,所述铰链区包含SEQ ID NO:9的氨基酸序列。In some embodiments, the dimerization domain comprises the hinge region of an immunoglobulin or a portion thereof and a CH2, CH3, CH4 domain or a combination thereof. In some embodiments, the dimerization domain comprises from N-terminus to C-terminus the hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin, preferably the Fc fragment of human IgGl. The CH2 and CH3 domains are described above. In some embodiments, the hinge region is derived from human IgGl. In some embodiments, the hinge region does not comprise one or more cysteines. In one embodiment, the hinge region comprises the amino acid sequence of SEQ ID NO:9.
在一具体实施方案中,二聚化结构域从N端至C端包含免疫球蛋白的CH2和CH3结构域,其中所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列。In a specific embodiment, the dimerization domain comprises, from N-terminus to C-terminus, the CH2 and CH3 domains of an immunoglobulin, wherein said CH2 and CH3 domains comprise SEQ ID NO: 7 or SEQ ID NO: 8 amino acid sequence.
在一具体实施方案中,二聚化结构域从N端至C端包含免疫球蛋白的铰链区或其部分、CH2和CH3结构域,其中所述铰链区或其部分包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列。在一优选实施方案中,所述CH2和CH3结构域包含SEQ ID NO:8的氨基酸序列。In a specific embodiment, the dimerization domain comprises from N-terminus to C-terminus the hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin, wherein said hinge region or part thereof comprises the sequence of SEQ ID NO:9 Amino acid sequence, the CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In a preferred embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:8.
在一些实施方案中,所述接头包含5-25个氨基酸,所述氨基酸各自独立地选自甘氨酸、丝氨酸和丙氨酸。在一具体实施方案中,所述接头包含(GGGGS) n,其中n选自1-10的整数。在一具体实施方案中,所述接头包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。在一具体实施方案中,所述接头包含SEQ ID NO:10或SEQ ID NO:11的氨基酸序列。 In some embodiments, the linker comprises 5-25 amino acids each independently selected from glycine, serine, and alanine. In a specific embodiment, the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10. In a specific embodiment, the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12. In a specific embodiment, the linker comprises the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 11.
在一些实施方案中,Lk 1和Lk 2都不存在,第二多肽从N端至C端具有如下结构: In some embodiments, neither Lk 1 nor Lk 2 is present, and the second polypeptide has the following structure from N-terminus to C-terminus:
Dd;Dd;
其中in
Dd为二聚化结构域,所述二聚化结构域如前定义。Dd is a dimerization domain, said dimerization domain being as previously defined.
在一些实施方案中,Lk 1和Lk 2各自独立地为接头,第二多肽从N端至C端具有如下结构: In some embodiments, Lk 1 and Lk 2 are each independently a linker, and the second polypeptide has the following structure from the N-terminus to the C-terminus:
Lk 1–Dd–Lk 2Lk 1 -Dd -Lk 2 ;
其中in
Dd为二聚化结构域,所述二聚化结构域如前定义;Dd is a dimerization domain, and the dimerization domain is as defined above;
Lk 1和Lk 2各自独立地为接头,所述接头如前定义。 Lk 1 and Lk 2 are each independently a linker, the linker being as defined above.
在一些实施方案中,Lk 1为接头,Lk 2不存在,第二多肽从N端至C端具有如下结构: In some embodiments, Lk 1 is a linker, Lk 2 is absent, and the second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd; Lk 1 -Dd;
或者or
Lk 1不存在,Lk 2为接头,第二多肽从N端至C端具有如下结构: Lk 1 does not exist, Lk 2 is a linker, and the second polypeptide has the following structure from N-terminus to C-terminus:
Dd–Lk 2Dd–Lk 2 ;
其中in
Dd为二聚化结构域,所述二聚化结构域如前定义;Dd is a dimerization domain, and the dimerization domain is as defined above;
Lk 1和Lk 2各自为接头,所述接头如前定义。 Each of Lk 1 and Lk 2 is a linker, the linker being as defined above.
在一具体实施方案中,第二多肽从N端至C端具有如下结构:In a specific embodiment, the second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 -Dd -Lk 2 ;
其中in
Dd为二聚化结构域,其从N端至C端包含铰链区、CH2和CH3结构域,其中所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminal to the C-terminus, wherein the hinge region comprises the amino acid sequence of SEQ ID NO:9, and the CH2 and CH3 domains comprise SEQ ID The amino acid sequence of NO:7 or SEQ ID NO:8;
Lk 1和Lk 2各自独立地为接头,所述接头如前定义。 Lk 1 and Lk 2 are each independently a linker, the linker being as defined above.
在一具体实施方案中,第二多肽从N端至C端具有如下结构:In a specific embodiment, the second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 -Dd -Lk 2 ;
其中in
Dd为二聚化结构域,其从N端至C端包含铰链区、CH2和CH3结构域,其中所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminal to the C-terminus, wherein the hinge region comprises the amino acid sequence of SEQ ID NO:9, and the CH2 and CH3 domains comprise SEQ ID The amino acid sequence of NO:7 or SEQ ID NO:8;
Lk 1和Lk 2各自独立地为接头,其包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。 Lk 1 and Lk 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
在一具体实施方案中,第二多肽从N端至C端具有如下结构:In a specific embodiment, the second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 -Dd -Lk 2 ;
其中in
Dd为二聚化结构域,其从N端至C端包含铰链区、CH2和CH3结构域,其中所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminal to the C-terminus, wherein the hinge region comprises the amino acid sequence of SEQ ID NO:9, and the CH2 and CH3 domains comprise SEQ ID NO: the amino acid sequence of 8;
Lk 1和Lk 2各自独立地为接头,其中Lk 1包含SEQ ID NO:10的氨基酸序列,Lk 2包含SEQ ID NO:11的氨基酸序列。 Lk 1 and Lk 2 are each independently a linker, wherein Lk 1 comprises the amino acid sequence of SEQ ID NO:10, and Lk 2 comprises the amino acid sequence of SEQ ID NO:11.
第三多肽third polypeptide
在本发明的实施方案中,第三多肽包含3个GITRL胞外结构域。不受任何理论所束缚,相比于单个GITRL胞外结构域(单体GITRL胞外结构域),包含3个GITRL胞外结构域的单链多肽(单链GITRL胞外结构域三聚体)在细胞表面形成与全长GITRL蛋白类似的三聚体,这样的三聚体作为GITR激动剂具有更高的GITR激活活性。In an embodiment of the invention, the third polypeptide comprises 3 GITRL extracellular domains. Without being bound by any theory, a single-chain polypeptide comprising 3 GITRL ectodomains (single-chain GITRL ectodomain trimer) compared to a single GITRL ectodomain (monomeric GITRL ectodomain) Trimers similar to the full-length GITRL protein are formed on the cell surface, and such trimers have higher GITR activation activity as GITR agonists.
糖皮质激素诱导的肿瘤坏死因子受体(Glucocorticoid-induced tumor necrosis factor  receptor,GITR)是肿瘤坏死因子受体超家族(TNFRSF)中的一员。不受任何理论束缚,据信GITR信号传导的激活(或激动)可以激活效应T细胞,并抑制调节性T细胞的免疫抑制活性。如本文所用,“GITR激动剂”是指能够诱导、促进、增强、激活或延长GITR信号传导的物质。可以使用本领域已知的各种检测方法确定GITR信号传导的激活,例如通过本文描述的检测方法。GITR激动剂包括但不限于GITRL、GITRL胞外结构域、GITR激动剂抗体。本发明的融合蛋白或缀合物可以作为GITR激动剂来诱导、促进、增强、激活或延长免疫反应,例如诱导、促进、增强、激活或延长针对肿瘤或肿瘤细胞的免疫反应。Glucocorticoid-induced tumor necrosis factor receptor (GITR) is a member of the tumor necrosis factor receptor superfamily (TNFRSF). Without being bound by any theory, it is believed that activation (or agonism) of GITR signaling can activate effector T cells and suppress the immunosuppressive activity of regulatory T cells. As used herein, "GITR agonist" refers to a substance capable of inducing, promoting, enhancing, activating or prolonging GITR signaling. Activation of GITR signaling can be determined using various assays known in the art, such as by the assays described herein. GITR agonists include, but are not limited to, GITRL, GITRL extracellular domain, GITR agonist antibodies. The fusion protein or conjugate of the present invention can act as a GITR agonist to induce, promote, enhance, activate or prolong an immune response, such as inducing, promoting, enhancing, activating or prolonging an immune response against a tumor or tumor cells.
如本文所用,“GITR配体(Glucocorticoid-induced TNF-related ligand,GITRL)”或“糖皮质激素诱导的肿瘤坏死因子受体配体”是指糖皮质激素诱导的肿瘤坏死因子受体(Glucocorticoid-induced tumor necrosis factor receptor,GITR)蛋白的结合配体。在一些实施方案中,GITRL是指人GITRL蛋白。人GITRL(hGITRL)蛋白由TNFSF18基因编码,其氨基酸序列如GenBank登录号NP_005083.2(或Uniprot ID:Q9Y5U5)所示。如本文所用,“GITRL胞外结构域”是指人GITRL蛋白的胞外区,其特异性结合并激动GITR。本领域技术人员会理解,GITRL胞外结构域可以包含的确切氨基酸序列可以变化而不影响其激动GITR的功能。通常,GITRL胞外结构域可以包含NP_005083.2的约第71-199位氨基酸,例如NP_005083.2的第72-199位氨基酸(SEQ ID NO:1)或第75-199位氨基酸(SEQ ID NO:2)。在一些实施方案中,GITRL胞外结构域包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列。As used herein, "GITR ligand (Glucocorticoid-induced TNF-related ligand, GITRL)" or "glucocorticoid-induced TNF receptor ligand" refers to glucocorticoid-induced tumor necrosis factor receptor (Glucocorticoid- induced tumor necrosis factor receptor (GITR) protein binding ligand. In some embodiments, GITRL refers to human GITRL protein. The human GITRL (hGITRL) protein is encoded by the TNFSF18 gene, and its amino acid sequence is shown in GenBank accession number NP_005083.2 (or Uniprot ID: Q9Y5U5). As used herein, "GITRL extracellular domain" refers to the extracellular region of the human GITRL protein, which specifically binds and agonizes GITR. Those skilled in the art will understand that the exact amino acid sequence that the extracellular domain of GITRL can comprise can vary without affecting its ability to agonize GITR. Typically, the GITRL extracellular domain may comprise about amino acids 71-199 of NP_005083.2, such as amino acids 72-199 (SEQ ID NO: 1) or amino acids 75-199 (SEQ ID NO ) of NP_005083.2 :2). In some embodiments, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
第三多肽还可以进一步包含位于所述GITRL胞外结构域之间的接头。所述接头如本文所述。The third polypeptide may further comprise a linker between said GITRL extracellular domains. The linker is as described herein.
在一些实施方案中,第三多肽从N端至C端具有如下结构:In some embodiments, the third polypeptide has the following structure from N-terminus to C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ;
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL;
Ln 1和Ln 2各自独立地为接头或不存在。 Ln 1 and Ln 2 are each independently a linker or absent.
在一些实施方案中,所述GITRL胞外结构域包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列。在一实施方案中,所述GITRL胞外结构域包含SEQ ID NO:2的氨基酸序列。In some embodiments, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2. In one embodiment, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:2.
在一些实施方案中,所述接头包含5-25个氨基酸,所述氨基酸各自独立地选自甘氨酸、丝氨酸和丙氨酸。在一具体实施方案中,所述接头包含(GGGGS) n,其中n选自1-10的整数。在一具体实施方案中,所述接头包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。在一具体实施方案中,所述接头包含SEQ ID NO:12的氨基酸序列。 In some embodiments, the linker comprises 5-25 amino acids each independently selected from glycine, serine, and alanine. In a specific embodiment, the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10. In a specific embodiment, the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12. In a specific embodiment, said linker comprises the amino acid sequence of SEQ ID NO:12.
在一些实施方案中,Ln 1和Ln 2都不存在,第三多肽从N端至C端具有如下结构: In some embodiments, neither Ln 1 nor Ln 2 is present, and the third polypeptide has the following structure from N-terminus to C-terminus:
Sd 1–Sd 2–Sd 3Sd 1 -Sd 2 -Sd 3 ;
其中Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,所述GITRL胞外结构域如前定义。 Wherein Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, and the extracellular domain of GITRL is as defined above.
在另一些实施方案中,Ln 1和Ln 2各自独立地为接头,第三多肽从N端至C端具有如下结构: In other embodiments, Ln 1 and Ln 2 are each independently a linker, and the third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ;
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,所述GITRL胞外结构域如前定义; Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain, and the GITRL extracellular domain is as defined above;
Ln 1和Ln 2各自独立地为接头,所述接头如前定义。 Ln 1 and Ln 2 are each independently a linker, the linker being as defined above.
在一些实施方案中,Ln 1为接头,Ln 2不存在,第三多肽从N端至C端具有如下结构: In some embodiments, Ln 1 is a linker, Ln 2 is absent, and the third polypeptide has the following structure from N-terminus to C-terminus:
Sd 1–Ln 1–Sd 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Sd 3 ;
或者or
Ln 1不存在,Ln 2为接头,第三多肽从N端至C端具有如下结构: Ln 1 does not exist, Ln 2 is a linker, and the third polypeptide has the following structure from N-terminus to C-terminus:
Sd 1–Sd 2–Ln 2–Sd 3Sd 1 -Sd 2 -Ln 2 -Sd 3 ;
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,所述GITRL胞外结构域如前定义; Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain, and the GITRL extracellular domain is as defined above;
Ln 1和Ln 2各自为接头,所述接头如前定义。 Each of Ln 1 and Ln 2 is a linker, the linker being as defined above.
在一具体实施方案中,第三多肽从N端至C端具有如下结构:In a specific embodiment, the third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ;
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,其包含SEQ ID NO:2的氨基酸序列; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
Ln 1和Ln 2各自独立地为接头,所述接头如前定义。 Ln 1 and Ln 2 are each independently a linker, the linker being as defined above.
在又一具体实施方案中,第三多肽从N端至C端具有如下结构:In yet another specific embodiment, the third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ;
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,其包含SEQ ID NO:2的氨基酸序列; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
Ln 1和Ln 2各自独立地为接头,其包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。 Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:10, SEQ ID NO:11 or SEQ ID NO:12.
在另一具体实施方案中,第三多肽从N端至C端具有如下结构:In another specific embodiment, the third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ;
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,其包含SEQ ID NO:2的氨基酸序列; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
Ln 1和Ln 2各自独立地为接头,其包含SEQ ID NO:12的氨基酸序列。 Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:12.
在一具体实施方案中,第三多肽包含SEQ ID NO:3的氨基酸序列。In a specific embodiment, the third polypeptide comprises the amino acid sequence of SEQ ID NO:3.
接头connector
融合蛋白的各组分之间(例如第一和第二多肽之间,第二和第三多肽之间,第三多肽中的第一、第二和第三GITRL胞外结构域之间)任选地通过接头连接。优选地,接头可以包含1-50个氨基酸,优选1-25个、3-25个、5-25个、4-10个氨基酸,例如4个、5个、6个、7个、8个、9个或10个氨基酸。氨基酸通常为无空间位阻的氨基酸,例如甘氨酸(G)、丙氨酸(A)和丝氨酸(S)。合适的接头为本领域技术人员熟知的,例如接头可以包含甘氨酸、丙氨酸、丝氨酸或其组合。示例性的接头可以包括但不限于:多聚甘氨酸、多聚丙氨酸、GGA、GS、GGSG、GGGS、GGGSGGG和GGGGSSGS。Between the components of the fusion protein (for example between the first and second polypeptide, between the second and the third polypeptide, between the first, second and third GITRL extracellular domains in the third polypeptide Between) are optionally connected by a linker. Preferably, the linker may comprise 1-50 amino acids, preferably 1-25, 3-25, 5-25, 4-10 amino acids, for example 4, 5, 6, 7, 8, 9 or 10 amino acids. Amino acids are generally unhindered amino acids such as glycine (G), alanine (A) and serine (S). Suitable linkers are well known to those skilled in the art, for example linkers may comprise glycine, alanine, serine or combinations thereof. Exemplary linkers may include, but are not limited to: polyglycine, polyalanine, GGA, GS, GGSG, GGGS, GGGSGGG, and GGGGSSGS.
在一实施方案中,接头包含(GGGGS) n,其中n为选自1-10的整数。在优选的实施方案中,n为选自2-5的整数。在一实施方案中,n为4(SEQ ID NO:10)。在另一实施方案中,n为2(SEQ ID NO:11)。 In one embodiment, the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10. In a preferred embodiment, n is an integer selected from 2-5. In one embodiment, n is 4 (SEQ ID NO: 10). In another embodiment, n is 2 (SEQ ID NO: 11).
在又一具体实施方案中,接头包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。In yet another specific embodiment, the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
信号肽signal peptide
本发明的融合蛋白任选地包含信号肽。信号肽(又称为信号序列、转运肽、前导序列或前导肽)是引导新生蛋白定位、转移和/或分泌的短肽(通常包含5-30个氨基酸)。在本发明中,优选的信号肽改善融合蛋白的表达水平和确保融合蛋白的正确折叠,并且帮助将融合蛋白分泌至细胞外。分泌信号肽通常在蛋白的分泌过程中被切割,使得蛋白作为成熟蛋白释放到细胞外。包含分泌信号肽的本发明的融合蛋白又可称为融合蛋白前体,其同样包括在本发明的融合蛋白范围之内。Fusion proteins of the invention optionally include a signal peptide. Signal peptides (also known as signal sequences, transit peptides, leader sequences, or leader peptides) are short peptides (typically comprising 5-30 amino acids) that direct the localization, transfer, and/or secretion of nascent proteins. In the present invention, the preferred signal peptide improves the expression level of the fusion protein and ensures the correct folding of the fusion protein, and helps to secrete the fusion protein out of cells. Secretory signal peptides are usually cleaved during protein secretion, allowing the protein to be released outside the cell as a mature protein. The fusion protein of the present invention comprising a secretion signal peptide can also be called a fusion protein precursor, which is also included in the scope of the fusion protein of the present invention.
信号肽可以位于融合蛋白的任何位置。优选地,信号肽位于融合蛋白的N端。The signal peptide can be located anywhere in the fusion protein. Preferably, the signal peptide is located at the N-terminus of the fusion protein.
信号肽可以是本领域已知的任何信号肽,优选通常在哺乳动物细胞(例如CHO细胞)表达中使用的分泌信号肽。在一具体实施方案中,所述信号肽序列包含SEQ ID NO:13或SEQ ID NO:14的氨基酸序列。The signal peptide may be any signal peptide known in the art, preferably a secretory signal peptide commonly used in expression in mammalian cells (eg, CHO cells). In a specific embodiment, the signal peptide sequence comprises the amino acid sequence of SEQ ID NO: 13 or SEQ ID NO: 14.
融合蛋白的具体实施方案Specific embodiments of fusion proteins
在一些实施方案中,本发明的融合蛋白从N端至C端包含:In some embodiments, the fusion protein of the present invention comprises from N-terminus to C-terminus:
第一多肽–第二多肽–第三多肽    式(1);The first polypeptide-the second polypeptide-the third polypeptide Formula (1);
或者or
第三多肽–第二多肽–第一多肽    式(2);The third polypeptide-the second polypeptide-the first polypeptide formula (2);
其中in
所述第一多肽包含TGF-βRⅡ的胞外结构域(TGFBR2-ECD);The first polypeptide comprises an extracellular domain of TGF-βRII (TGFBR2-ECD);
所述第二多肽从N端至C端具有如下结构:The second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 –Dd–Lk 2 ,
其中in
Dd为二聚化结构域;Dd is a dimerization domain;
Lk 1和Lk 2各自独立地为接头或不存在;并且 Lk 1 and Lk 2 are each independently a linker or absent; and
所述第三多肽从N端至C端具有如下结构:The third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ,
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域;并且 Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain; and
Ln 1和Ln 2各自独立地为接头或不存在。 Ln 1 and Ln 2 are each independently a linker or absent.
在一些实施方案中,所述TGFBR2-ECD包含SEQ ID NO:4、SEQ ID NO:5或SEQ ID NO:6的氨基酸序列。在优选的实施方案中,所述TGFBR2-ECD包含SEQ ID NO:5的氨基酸序列。In some embodiments, the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6. In a preferred embodiment, the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:5.
在一些实施方案中,所述GITRL胞外结构域包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列。在优选的实施方案中,所述GITRL胞外结构域包含SEQ ID NO:2的氨基酸序列。In some embodiments, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2. In a preferred embodiment, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:2.
在一些实施方案中,所述二聚化结构域包含CH2和CH3结构域。在优选的实施方案中,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列。在一些实施方案中,所述二聚化结构域包含铰链区、CH2和CH3结构域。在一实施方案中,所述铰链区包含SEQ ID NO:9的氨基酸序列。在一实施方案中,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列。在一优选实施方案中,所述CH2和CH3结构域包含SEQ ID NO:8的氨基酸序列。在一些实施方案中,所述二聚化结构域包含铰链区、CH2和CH3结构域,其中所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:8的氨基酸序列。In some embodiments, the dimerization domain comprises CH2 and CH3 domains. In a preferred embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In some embodiments, the dimerization domain comprises a hinge region, CH2 and CH3 domains. In one embodiment, the hinge region comprises the amino acid sequence of SEQ ID NO:9. In one embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8. In a preferred embodiment, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:8. In some embodiments, the dimerization domain comprises a hinge region, CH2 and CH3 domains, wherein the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO: 8 amino acid sequence.
在一些实施方案中,所述接头包含5-25个氨基酸,所述氨基酸各自独立地选自甘氨酸、丝氨酸和丙氨酸。在优选的实施方案中,所述接头包含(GGGGS) n,其中n选自1-10的整数。在一实施方案中,所述接头包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。 In some embodiments, the linker comprises 5-25 amino acids each independently selected from glycine, serine, and alanine. In a preferred embodiment, the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10. In one embodiment, the linker comprises the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12.
在一实施方案中,所述第三多肽从N端至C端具有如下结构:In one embodiment, the third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ;
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,其包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列;并且 Sd 1 , Sd 2 and Sd 3 are each independently a GITRL extracellular domain comprising the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2; and
Ln 1和Ln 2各自独立地为接头。 Ln 1 and Ln 2 are each independently a linker.
在一些实施方案中,所述GITRL胞外结构域包含SEQ ID NO:2的氨基酸序列,Ln 1和Ln 2各自独立地为接头,其包含SEQ ID NO:12的氨基酸序列。在一具体的实施方案中,所述第三多肽包含SEQ ID NO:3的氨基酸序列。 In some embodiments, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:2, and each of Ln 1 and Ln 2 is independently a linker comprising the amino acid sequence of SEQ ID NO:12. In a specific embodiment, said third polypeptide comprises the amino acid sequence of SEQ ID NO:3.
在一实施方案中,所述第二多肽从N端至C端具有如下结构:In one embodiment, the second polypeptide has the following structure from the N-terminus to the C-terminus:
Lk 1–Dd–Lk 2Lk 1 -Dd -Lk 2 ;
其中in
Dd为二聚化结构域,其从N端至C端包含铰链区、CH2和CH3结构域,所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO :7 or the amino acid sequence of SEQ ID NO:8;
Lk 1和Lk 2各自独立地为接头。 Lk 1 and Lk 2 are each independently a linker.
在一些实施方案中,所述二聚化结构域从N端至C端包含铰链区、CH2和CH3结构域,所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:8的氨基酸序列,Lk 1和Lk 2各自独立地为接头,其中Lk 1包含SEQ ID NO:10的氨基酸序列,Lk 2包含SEQ ID NO:11的氨基酸序列。 In some embodiments, the dimerization domain comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, the CH2 and CH3 domains Comprising the amino acid sequence of SEQ ID NO: 8, Lk 1 and Lk 2 are each independently a linker, wherein Lk 1 contains the amino acid sequence of SEQ ID NO: 10, and Lk 2 contains the amino acid sequence of SEQ ID NO: 11.
在一些实施方案中,本发明的融合蛋白从N端至C端包含:In some embodiments, the fusion protein of the present invention comprises from N-terminus to C-terminus:
第一多肽–第二多肽–第三多肽    式(1);The first polypeptide-the second polypeptide-the third polypeptide Formula (1);
或者or
第三多肽–第二多肽–第一多肽    式(2);The third polypeptide-the second polypeptide-the first polypeptide formula (2);
其中in
所述第一多肽包含TGFBR2-ECD,其包含SEQ ID NO:4、SEQ ID NO:5或SEQ ID NO:6的氨基酸序列;The first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6;
所述第二多肽从N端至C端具有如下结构:The second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 –Dd–Lk 2 ,
其中in
Dd为二聚化结构域,其从N端至C端包含铰链区、CH2和CH3结构域,所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO :7 or the amino acid sequence of SEQ ID NO:8;
Lk 1和Lk 2各自独立地为接头,其包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列;并且 Lk 1 and Lk 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO: 10, SEQ ID NO: 11 or SEQ ID NO: 12; and
所述第三多肽从N端至C端具有如下结构:The third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ,
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,其包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2;
Ln 1和Ln 2各自独立地为接头,其包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。 Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:10, SEQ ID NO:11 or SEQ ID NO:12.
在一具体实施方案中,本发明的融合蛋白从N端至C端包含:In a specific embodiment, the fusion protein of the present invention comprises from the N-terminus to the C-terminus:
第一多肽–第二多肽–第三多肽    式(1);The first polypeptide-the second polypeptide-the third polypeptide Formula (1);
其中in
所述第一多肽包含TGFBR2-ECD,其包含SEQ ID NO:4、SEQ ID NO:5或SEQ ID NO:6的氨基酸序列;The first polypeptide comprises TGFBR2-ECD comprising the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6;
所述第二多肽从N端至C端具有如下结构:The second polypeptide has the following structure from N-terminus to C-terminus:
Lk 1–Dd–Lk 2Lk 1 –Dd–Lk 2 ,
其中in
Dd为二聚化结构域,其从N端至C端包含铰链区、CH2和CH3结构域,所述铰链区包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain, which comprises a hinge region, CH2 and CH3 domains from the N-terminus to the C-terminus, the hinge region comprises the amino acid sequence of SEQ ID NO: 9, and the CH2 and CH3 domains comprise SEQ ID NO : the amino acid sequence of 8;
Lk 1和Lk 2各自独立地为接头,其中Lk 1包含SEQ ID NO:10的氨基酸序列,Lk 2包含SEQ ID NO:11的氨基酸序列;并且 Lk 1 and Lk 2 are each independently a linker, wherein Lk 1 comprises the amino acid sequence of SEQ ID NO: 10, and Lk 2 comprises the amino acid sequence of SEQ ID NO: 11; and
所述第三多肽从N端至C端具有如下结构:The third polypeptide has the following structure from the N-terminus to the C-terminus:
Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ,
其中in
Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域,其包含SEQ ID NO:2的氨基酸序列; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL, which comprises the amino acid sequence of SEQ ID NO:2;
Ln 1和Ln 2各自独立地为接头,其包含SEQ ID NO:12的氨基酸序列。 Ln 1 and Ln 2 are each independently a linker comprising the amino acid sequence of SEQ ID NO:12.
在一具体实施方案中,所述融合蛋白包含SEQ ID NO:15或SEQ ID NO:16的氨基酸序列。In a specific embodiment, the fusion protein comprises the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
在其他实施方案中,如前所述的任一融合蛋白还包含信号肽。优选地,信号肽位于融合蛋白的N端。在一具体实施方案中,所述信号肽包含SEQ ID NO:13或SEQ ID NO:14的氨基酸序列。In other embodiments, any fusion protein as described above further comprises a signal peptide. Preferably, the signal peptide is located at the N-terminus of the fusion protein. In a specific embodiment, the signal peptide comprises the amino acid sequence of SEQ ID NO: 13 or SEQ ID NO: 14.
在一实施方案中,如前所述的任一融合蛋白在N端包含信号肽,其中所述信号肽包含SEQ ID NO:13或SEQ ID NO:14的氨基酸序列。In one embodiment, any fusion protein as described above comprises a signal peptide at the N-terminus, wherein the signal peptide comprises the amino acid sequence of SEQ ID NO: 13 or SEQ ID NO: 14.
在一些实施方案中,所述融合蛋白形成二聚体或多聚体。In some embodiments, the fusion protein forms dimers or multimers.
核酸、载体和宿主细胞Nucleic acids, vectors and host cells
在另一方面,本发明提供分离的多核苷酸,其编码本发明的融合蛋白。在一些优选实施方式中,所述多核苷酸编码本发明的融合蛋白的前体。可以通过本领域公知的方法获得所述多核苷酸,例如,重组DNA技术、化学合成等。通常可以针对用于表达的宿主细胞对多核苷酸进行密码子优化。In another aspect, the invention provides isolated polynucleotides encoding fusion proteins of the invention. In some preferred embodiments, the polynucleotide encodes a precursor of a fusion protein of the invention. The polynucleotides can be obtained by methods known in the art, for example, recombinant DNA techniques, chemical synthesis, and the like. Polynucleotides can generally be codon-optimized for the host cell used for expression.
本发明还提供一种表达载体,其包含本发明的多核苷酸。利用本发明的多核苷酸构建合适的表达载体的方法是本领域技术人员所公知的,包括但不限于体外重组DNA、化学合成和体内重组(遗传重组)等。The present invention also provides an expression vector comprising the polynucleotide of the present invention. Methods for constructing suitable expression vectors using the polynucleotides of the present invention are well known to those skilled in the art, including but not limited to in vitro recombinant DNA, chemical synthesis, and in vivo recombination (genetic recombination).
本发明还提供一种宿主细胞,其包含本发明的多核苷酸或表达载体。合适的宿主细胞包括但不限于原核细胞(例如,大肠杆菌)和真核细胞(例如,酵母细胞、昆虫细胞和哺乳动物细胞)。在优选的实施方案中,宿主细胞为猿猴COS细胞、中国仓鼠卵巢(CHO)细胞、HEK293F细胞或免疫效应细胞(例如效应T细胞)。在一实施方案中,宿主细胞为酵母细胞、昆虫细胞或哺乳动物细胞。在一实施方案中,宿主细胞为效应T细胞。The present invention also provides a host cell comprising the polynucleotide or expression vector of the present invention. Suitable host cells include, but are not limited to, prokaryotic cells (eg, E. coli) and eukaryotic cells (eg, yeast cells, insect cells, and mammalian cells). In preferred embodiments, the host cells are simian COS cells, Chinese Hamster Ovary (CHO) cells, HEK293F cells or immune effector cells (eg effector T cells). In one embodiment, the host cell is a yeast cell, an insect cell or a mammalian cell. In one embodiment, the host cell is an effector T cell.
融合蛋白的生产方法Production method of fusion protein
本发明的另一方面涉及一种方法,其用于生产本发明的融合蛋白,所述方法包括:Another aspect of the present invention relates to a method for producing the fusion protein of the present invention, the method comprising:
(i)在适合多核苷酸或表达载体表达的情况下培养本发明的宿主细胞,和(i) culturing the host cell of the invention under conditions suitable for expression of the polynucleotide or expression vector, and
(ⅱ)从所述宿主细胞或其培养物回收本发明的融合蛋白。(ii) recovering the fusion protein of the present invention from said host cell or its culture.
可以采用本领域已知的各种方法将本发明的多核苷酸或表达载体(特别是本发明的编码融合蛋白表达前体的多核苷酸或表达载体)导入合适的宿主细胞中。这类方法包括但不限于脂质体转染、电穿孔、稳定转染、磷酸钙转染等。在一些实施方案中,为了进一步纯化本发明的融合蛋白,可以使用常规的亲和层析、离子交换层析、疏水性相互作用层析、反相层析、凝胶过滤或其组合。Various methods known in the art can be used to introduce the polynucleotide or expression vector of the present invention (especially the polynucleotide or expression vector encoding the expression precursor of the fusion protein of the present invention) into suitable host cells. Such methods include, but are not limited to, lipofection, electroporation, stable transfection, calcium phosphate transfection, and the like. In some embodiments, to further purify the fusion protein of the present invention, conventional affinity chromatography, ion exchange chromatography, hydrophobic interaction chromatography, reverse phase chromatography, gel filtration or a combination thereof can be used.
缀合物Conjugate
本发明进一步提供一种缀合物,其包含与至少一种治疗剂缀合的本发明的融合蛋白。The invention further provides a conjugate comprising a fusion protein of the invention conjugated to at least one therapeutic agent.
如本文所用,“缀合”是指两个或多个部分通过共价或非共价作用相互连接。优选地,本发明的融合蛋白和至少一种治疗剂共价缀合。As used herein, "conjugate" refers to the linking of two or more moieties to each other by covalent or non-covalent interactions. Preferably, fusion proteins of the invention and at least one therapeutic agent are covalently conjugated.
在一些实施方案中,所述治疗剂选自可检测标记物、化疗剂、细胞毒素、放射性 核素、免疫检查点抑制剂、细胞因子和酶。In some embodiments, the therapeutic agent is selected from detectable markers, chemotherapeutic agents, cytotoxins, radionuclides, immune checkpoint inhibitors, cytokines, and enzymes.
在一些实施方案中,治疗剂为可检测标记物,例如荧光或发光标记物、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。In some embodiments, the therapeutic agent is a detectable label, such as a fluorescent or luminescent label, a radioactive label, an MRI (magnetic resonance imaging) or CT (computed tomography) contrast agent.
在一些实施方案中,治疗剂为细胞毒素。细胞毒素的实例包括但不限于紫杉醇、细胞松弛素B、短杆菌肽D、溴化乙啶、吐根碱、丝裂霉素、表鬼臼毒吡喃葡糖苷、表鬼臼毒噻吩糖苷、长春新碱、长春碱、秋水仙素、阿霉素、柔红霉素、二羟基炭疽菌素二酮、米托蒽醌、光辉霉素、放线菌素D、1-脱氢睾酮、糖皮质激素、普鲁卡因、丁卡因、利多卡因、癌霉素、刺孢霉素、美坦生、阿里他汀、普萘洛尔和嘌呤霉素和它们的类似物或同系物。In some embodiments, the therapeutic agent is a cytotoxin. Examples of cytotoxins include, but are not limited to, paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, epipodophyllotoxin glucopyranoside, epipodophyllotoxin thienenoside, Vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxyanthraxendione, mitoxantrone, mitoxantrone, actinomycin D, 1-dehydrotestosterone, sugar Corticosteroids, procaine, tetracaine, lidocaine, carcinomacin, calicheamicin, maytansin, alistatin, propranolol and puromycin and their analogs or homologues.
在一些实施方案中,治疗剂为化疗剂。化疗剂的实例包括但不限于抗代谢物(例如,氨甲喋呤、6-巯基嘌呤、6-硫代鸟嘌呤、阿糖胞苷、5-氟尿嘧啶、氨烯咪胺)、烷化剂(例如,氮芥、苯丁酸氮芥、苯丙氨酸氮芥、卡莫司汀(BSNU)和洛莫司汀(CCNU)、环磷酰胺、白消安、二溴甘露糖醇、链唑霉素、丝裂霉素C和顺-二氯二胺合铂(Ⅱ)(DDP)顺铂)、氨茴霉素类(例如,柔红菌素和阿霉素)、抗生素(例如,放线菌素D、博来霉素、光辉霉素和安曲霉素(AMC))和抗有丝分裂剂(例如,长春新碱和长春碱)。在一些实施方案中,治疗剂为生物活性蛋白,例如具有酶活性的毒素或其活性片段,如相思豆毒蛋白、蓖麻毒蛋白A、假单胞菌外毒素或白喉毒素。In some embodiments, the therapeutic agent is a chemotherapeutic agent. Examples of chemotherapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil, carbamide), alkylating agents (e.g., nitrogen mustard, chlorambucil, phenylalanine mustard, carmustine (BSNU) and lomustine (CCNU), cyclophosphamide, busulfan, dibromomannitol, streptozotocin, Mitomycin C and cis-dichlorodiamine platinum(II) (DDP) cisplatin), anthraninomycins (eg, daunorubicin and doxorubicin), antibiotics (eg, actinomycin D , bleomycin, mithromycin, and antramycin (AMC)) and antimitotic agents (eg, vincristine and vinblastine). In some embodiments, the therapeutic agent is a biologically active protein, such as a toxin with enzymatic activity or an active fragment thereof, such as abrin, ricin A, Pseudomonas exotoxin, or diphtheria toxin.
在一些实施方案中,治疗剂为细胞因子。细胞因子的实例包括但不限于肿瘤坏死因子(例如TNFα和TNFβ)、干扰素(例如INFα、INFβ和INFγ)、白介素(例如IL-1、IL-2、IL-4、IL-5、IL-6、IL-7、IL-8、IL-10、IL-12、IL-13、IL-15、IL-17、IL-18、IL21和IL-23)、集落刺激因子(例如巨噬细胞集落刺激因子、粒细胞集落刺激因子、粒细胞和巨噬细胞集落刺激因子、多重集落刺激因子、干细胞因子)、生长因子(例如VEGF、HGF、FGF、FGF-2、PDGF、IGF、TGF、NGF、EPO和EGF)和趋化因子。In some embodiments, the therapeutic agent is a cytokine. Examples of cytokines include, but are not limited to, tumor necrosis factors (such as TNFα and TNFβ), interferons (such as INFα, INFβ, and INFγ), interleukins (such as IL-1, IL-2, IL-4, IL-5, IL- 6. IL-7, IL-8, IL-10, IL-12, IL-13, IL-15, IL-17, IL-18, IL21 and IL-23), colony-stimulating factors (such as macrophage colony stimulatory factors, granulocyte colony stimulating factor, granulocyte and macrophage colony stimulating factor, multiple colony stimulating factor, stem cell factor), growth factors (such as VEGF, HGF, FGF, FGF-2, PDGF, IGF, TGF, NGF, EPO and EGF) and chemokines.
在一些实施方案中,治疗剂为免疫检查点抑制剂。免疫检查点抑制剂的实例包括针对免疫检查点分子(例如PD-L1、PD-1、CTLA4、LAG-3、BTLA、TIM-2、LAIR1、HVEM和TIM-4)的抗体或其抗原结合片段。抗体或其抗原结合片段可以选自合成抗体、重组产生的抗体、多特异性抗体、双特异性抗体、人抗体、非人抗体、人源化抗体、嵌合抗体、胞内抗体、单域抗体、Fab片段、Fab’片段、F(ab’) 2片段、Fv片段、dsFv片段、Fd片段、Fd’片段、scFv、scFab、双抗体。 In some embodiments, the therapeutic agent is an immune checkpoint inhibitor. Examples of immune checkpoint inhibitors include antibodies or antigen-binding fragments thereof against immune checkpoint molecules such as PD-L1, PD-1, CTLA4, LAG-3, BTLA, TIM-2, LAIR1, HVEM, and TIM-4 . The antibody or antigen-binding fragment thereof may be selected from synthetic antibodies, recombinantly produced antibodies, multispecific antibodies, bispecific antibodies, human antibodies, non-human antibodies, humanized antibodies, chimeric antibodies, intrabodies, single domain antibodies , Fab fragment, Fab' fragment, F(ab') 2 fragment, Fv fragment, dsFv fragment, Fd fragment, Fd' fragment, scFv, scFab, diabody.
在一些实施方案中,治疗剂为放射性核素。放射性核素的例子包括但不限于碘125、碘131、铟111、铟131、钇90、铑105、铋212和镥177。In some embodiments, the therapeutic agent is a radionuclide. Examples of radionuclides include, but are not limited to, iodine-125, iodine-131, indium-111, indium-131, yttrium-90, rhodium-105, bismuth-212, and lutetium-177.
治疗剂还可以是能够产生可检测产物的酶(例如辣根过氧化物酶)、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))和任何形式的纳米颗粒等。The therapeutic agent can also be an enzyme capable of producing a detectable product (e.g., horseradish peroxidase), gold nanoparticles/nanorods, virosomes, liposomes, nanomagnetic particles, prodrug-activating enzymes (e.g., DT-cardiac diaphorase (DTD) or biphenyl hydrolase-like protein (BPHL)) and nanoparticles in any form, etc.
制备缀合物的方法是本领域技术人员已知的。例如,可以利用本领域使用的接头技术将上述治疗剂与本发明的融合蛋白缀合。已经用于细胞毒素缀合的接头类型的实例包括但不限于腙、硫醚、酯、二硫化物和肽接头。或者,可以利用重组DNA技术将编码蛋白治疗剂(例如如上所述的细胞因子、抗体或其抗原结合片段、生物活性蛋白或者酶)的多核苷酸与编码本发明的融合蛋白的多核苷酸可操作地连接并在合适的宿主细胞中表达。Methods of preparing conjugates are known to those skilled in the art. For example, the therapeutic agents described above can be conjugated to fusion proteins of the invention using linker technology used in the art. Examples of linker types that have been used for cytotoxin conjugation include, but are not limited to, hydrazone, thioether, ester, disulfide, and peptide linkers. Alternatively, polynucleotides encoding protein therapeutics (such as cytokines, antibodies or antigen-binding fragments thereof, biologically active proteins or enzymes as described above) and polynucleotides encoding fusion proteins of the present invention can be combined using recombinant DNA technology. Operably linked and expressed in a suitable host cell.
药物组合物pharmaceutical composition
本发明进一步提供一种药物组合物,其包含:(ⅰ)本发明的融合蛋白或缀合物;以及(ⅱ)药学上可接受的载剂。The present invention further provides a pharmaceutical composition, which comprises: (i) the fusion protein or conjugate of the present invention; and (ii) a pharmaceutically acceptable carrier.
在本文中,“药学上可接受的载剂”包括生理学相容的任何和所有的物质。药学上可接受的载剂的非限制性实例包括:溶剂(例如水或油)、分散剂、助悬剂、包衣、稀释剂、佐剂、抗氧化剂(例如抗坏血酸、棕榈酸抗坏血酸酯、亚硫酸钠、丁羟茴醚(BHA)、没食子酸丙酯、焦亚硫酸钠、α-生育酚等)、赋形剂、防腐剂(例如抗细菌剂和抗真菌剂)、表面活性剂(例如聚山梨酯类、泊洛沙姆、triton、辛基糖苷钠、十二烷基硫酸钠、月桂硫酸钠、月桂基-磺基甜菜碱、聚乙二醇、聚丙二醇等)、湿润剂、乳化剂、填充剂、碳水化合物(例如果糖、蔗糖、海藻糖、甘露糖、葡聚糖、环糊精等)、氨基酸(例如丙氨酸、精氨酸、天冬酰胺、天冬氨酸、半胱氨酸、谷酰胺、谷氨酸、甘氨酸、组氨酸、异亮氨酸、亮氨酸、赖氨酸、蛋氨酸、苯丙氨酸、脯氨酸、丝氨酸、苏氨酸、色氨酸、酪氨酸、缬氨酸)、缓冲剂(例如醋酸、盐酸、硫酸、组氨酸、柠檬酸盐、磷酸盐、琥珀酸盐)、稳定剂、螯合剂(例如EDTA)、等渗剂和延迟吸收剂(例如单硬脂酸盐和明胶)等。优选地,载剂适合于静脉内、肌内、皮下、肠胃外、脊柱或表皮施用(如通过注射或输注)。As used herein, "pharmaceutically acceptable carrier" includes any and all substances that are physiologically compatible. Non-limiting examples of pharmaceutically acceptable carriers include: solvents (such as water or oils), dispersants, suspending agents, coatings, diluents, adjuvants, antioxidants (such as ascorbic acid, ascorbyl palmitate, sodium sulfite, , butylated hydroxyanisole (BHA), propyl gallate, sodium metabisulfite, α-tocopherol, etc.), excipients, preservatives (such as antibacterial and antifungal agents), surfactants (such as polysorbates , poloxamer, triton, sodium octyl glycoside, sodium lauryl sulfate, sodium lauryl sulfate, lauryl-sulfobetaine, polyethylene glycol, polypropylene glycol, etc.), wetting agent, emulsifier, filler , carbohydrates (such as fructose, sucrose, trehalose, mannose, dextran, cyclodextrin, etc.), amino acids (such as alanine, arginine, asparagine, aspartic acid, cysteine, Glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine , valine), buffers (such as acetic acid, hydrochloric acid, sulfuric acid, histidine, citrate, phosphate, succinate), stabilizers, chelating agents (such as EDTA), isotonic and absorption delaying agents ( Such as monostearate and gelatin), etc. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (eg, by injection or infusion).
本发明的融合蛋白、缀合物或药物组合物可以利用本领域公知的一种或多种方法通过一种或多种给药途径给药。本领域技术人员应当理解,给药途径和/或方式根据期望的结果而不同。优选给药途径包括静脉内、肌肉内、皮内、腹膜内、皮下、脊柱或其他肠胃外给药途径,例如注射或输注。本文使用的短语“肠胃外给药”是指除肠和局部给药以外的给药模式,通常是注射,包括但不限于静脉内、肌内、动脉内、鞘内、囊内、眶内、心内、皮内、腹膜内、经气管、皮下、表皮下、关节内、囊下、蛛网膜下、脊柱内、硬膜外和胸骨内注射和输注。或者,本发明的融合蛋白、缀合物或药物组合物也可以通过非肠胃外途径给药,如局部、表皮或粘膜途径给药,例如,鼻内、经口、阴道、直肠、舌下或局部。The fusion protein, conjugate or pharmaceutical composition of the present invention can be administered through one or more administration routes using one or more methods known in the art. Those skilled in the art will appreciate that the route and/or manner of administration will vary depending on the desired result. Preferred routes of administration include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration such as injection or infusion. As used herein, the phrase "parenteral administration" refers to modes of administration other than enteral and topical administration, usually injection, including but not limited to intravenous, intramuscular, intraarterial, intrathecal, intrasaccular, intraorbital, Intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcutaneous, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, and intrasternal injection and infusion. Alternatively, the fusion protein, conjugate or pharmaceutical composition of the present invention may also be administered by non-parenteral routes, such as topical, epidermal or mucosal routes, for example, intranasally, orally, vaginally, rectally, sublingually or partial.
可以将药物组合物制备成各种适于给药的剂型,例如溶液、悬液、微乳状液、乳剂、脂质体、片剂、丸剂、胶囊剂、粉剂、缓释剂等形式。在一些实施方案中,将所述药物组合物制备为注射液、粉剂或脂质体。The pharmaceutical composition can be prepared in various dosage forms suitable for administration, such as solutions, suspensions, microemulsions, emulsions, liposomes, tablets, pills, capsules, powders, sustained-release formulations and the like. In some embodiments, the pharmaceutical composition is prepared as injection, powder or liposome.
治疗treat
本发明提供的融合蛋白、多核苷酸、缀合物或药物组合物用于治疗和/或预防癌症。在一实施方案中,所述融合蛋白、多核苷酸、缀合物或药物组合物用于激活GITR信号传导和抑制TGF-β信号传导。在一实施方案中,所述融合蛋白、多核苷酸、缀合物或药物组合物用于在有需要的受试者中诱导、促进、增强、激活或延长免疫反应。在一实施方案中,所述融合蛋白、多核苷酸、缀合物或药物组合物用于诱导、促进、增强、激活或延长有需要的受试者中针对肿瘤或肿瘤细胞的免疫反应。在一实施方案中,所述融合蛋白、多核苷酸、缀合物或药物组合物用于激活效应T细胞,例如CD8 +效应T细胞。 The fusion protein, polynucleotide, conjugate or pharmaceutical composition provided by the present invention is used for treating and/or preventing cancer. In one embodiment, the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to activate GITR signaling and inhibit TGF-β signaling. In one embodiment, the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to induce, promote, enhance, activate or prolong an immune response in a subject in need thereof. In one embodiment, the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to induce, promote, enhance, activate or prolong an immune response against a tumor or tumor cells in a subject in need thereof. In one embodiment, the fusion protein, polynucleotide, conjugate or pharmaceutical composition is used to activate effector T cells, such as CD8 + effector T cells.
本发明还涉及本发明的融合蛋白、多核苷酸、缀合物或药物组合物在制备药物中的用途。在一实施方案中,所述药物用于治疗和/或预防癌症。在一实施方案中,所述药物用于激活GITR信号传导和抑制TGF-β信号传导。在一实施方案中,所述药物用于在有需要的受试者中诱导、促进、增强、激活或延长免疫反应。在一实施方案中,所述药物用于诱导、促进、增强、激活或延长有需要的受试者中针对肿瘤或肿瘤细胞的免疫反应。在一实施方案中,所述药物用于激活效应T细胞。在一实施方案中,所述T细胞为CD8 +效应T细胞。 The present invention also relates to the use of the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention in the preparation of medicines. In one embodiment, the medicament is for the treatment and/or prevention of cancer. In one embodiment, the medicament is for activating GITR signaling and inhibiting TGF-beta signaling. In one embodiment, the medicament is for inducing, promoting, enhancing, activating or prolonging an immune response in a subject in need thereof. In one embodiment, the medicament is for inducing, promoting, enhancing, activating or prolonging an immune response against a tumor or tumor cells in a subject in need thereof. In one embodiment, the drug is used to activate effector T cells. In one embodiment, the T cells are CD8 + effector T cells.
本发明进一步提供一种治疗和/或预防癌症的方法,其包括向有需要的受试者给药治疗有效量的本发明的融合蛋白、多核苷酸、缀合物或药物组合物。在一实施方案中,所述方法诱导、促进、增强、激活或延长有需要的受试者中针对肿瘤或肿瘤细胞的免疫反应。The present invention further provides a method for treating and/or preventing cancer, which comprises administering a therapeutically effective amount of the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention to a subject in need. In one embodiment, the method induces, promotes, enhances, activates or prolongs an immune response against a tumor or tumor cells in a subject in need thereof.
本发明还提供一种诱导、促进、增强、激活或延长免疫反应的方法,其包括施用本发明的融合蛋白、多核苷酸、缀合物或药物组合物。The present invention also provides a method for inducing, promoting, enhancing, activating or prolonging an immune response, which comprises administering the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention.
本发明还提供一种激活GITR信号传导和抑制TGF-β信号传导的方法,和/或激活效应T细胞的方法,其包括施用本发明的融合蛋白、多核苷酸、缀合物或药物组合物。The present invention also provides a method for activating GITR signal transduction and inhibiting TGF-β signal transduction, and/or a method for activating effector T cells, which comprises administering the fusion protein, polynucleotide, conjugate or pharmaceutical composition of the present invention .
在本文中,术语“癌症”或“肿瘤”是指或描述哺乳动物中特征通常为细胞生长不受调节的生理状况。癌症的非限制性的实例包括但不限于腺癌、淋巴瘤、母细胞瘤、黑素瘤、肉瘤和白血病。癌症的更具体实例包括:鳞状细胞癌、小细胞肺癌、非小细胞肺癌、胃肠癌、霍奇金和非霍奇金淋巴瘤、胰腺癌、胶质母细胞瘤、神经胶质瘤、宫颈癌、卵巢癌、肝癌诸如肝细胞癌、膀胱癌、乳癌、结肠癌、结肠直肠癌、子宫内膜癌、 骨髓瘤(诸如多发性骨髓瘤)、唾液腺癌、肾癌诸如肾细胞癌和威尔曼瘤、基底细胞癌、黑素瘤、前列腺癌、外阴癌、甲状腺癌、睾丸癌、食管癌及各种类型的头颈癌。As used herein, the term "cancer" or "tumor" refers to or describes the physiological condition in mammals that is often characterized by unregulated cell growth. Non-limiting examples of cancer include, but are not limited to, adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, and leukemia. More specific examples of cancer include: squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, Hodgkin and non-Hodgkin lymphoma, pancreatic cancer, glioblastoma, glioma, Cervical cancer, ovarian cancer, liver cancer such as hepatocellular carcinoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma (such as multiple myeloma), salivary gland cancer, kidney cancer such as renal cell carcinoma and Kleman's tumor, basal cell carcinoma, melanoma, prostate cancer, vulvar cancer, thyroid cancer, testicular cancer, esophageal cancer and various types of head and neck cancer.
在一实施方案中,癌症选自腺癌、淋巴瘤、母细胞瘤、黑素瘤、肉瘤、白血病、鳞状细胞癌、小细胞肺癌、非小细胞肺癌、胃肠癌、霍奇金和非霍奇金淋巴瘤、胰腺癌、胶质母细胞瘤、神经胶质瘤、宫颈癌、卵巢癌、肝癌、肝细胞癌、膀胱癌、乳癌、结肠癌、结肠直肠癌、子宫内膜癌、骨髓瘤、多发性骨髓瘤、唾液腺癌、肾癌、肾细胞癌、威尔曼瘤、基底细胞癌、黑素瘤、前列腺癌、外阴癌、甲状腺癌、睾丸癌、食管癌和头颈癌。In one embodiment, the cancer is selected from adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, leukemia, squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, Hodgkin's and non-small cell lung cancer. Hodgkin lymphoma, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, hepatocellular carcinoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, bone marrow cancer, multiple myeloma, salivary gland cancer, kidney cancer, renal cell carcinoma, Wellman's tumor, basal cell carcinoma, melanoma, prostate cancer, vulvar cancer, thyroid cancer, testicular cancer, esophageal cancer, and head and neck cancer.
如本文中所使用的,术语“受试者”是指哺乳动物,包括但不限于,山羊、绵羊、猪、大鼠、小鼠、兔、豚鼠、牛、马、狗、猫、非人灵长类动物(例如大猩猩、狒狒和黑猩猩)和人。在一些实施方案中,所述受试者是人。As used herein, the term "subject" refers to mammals, including, but not limited to, goats, sheep, pigs, rats, mice, rabbits, guinea pigs, cows, horses, dogs, cats, non-human Long animals (such as gorillas, baboons and chimpanzees) and humans. In some embodiments, the subject is a human.
联合治疗combination therapy
本发明的融合蛋白、缀合物或药物组合物可以与其他治疗剂联用。其他治疗剂包括例如化疗剂、免疫检查点抑制剂或靶向肿瘤相关抗原的抗体。在一实施方案中,其他治疗剂为免疫检查点抑制剂。在一些实施方案中,所述免疫检查点抑制剂为抗PD-L1抗体、抗PD-1抗体、抗CTLA4抗体、抗LAG-3抗体、抗TIM-3抗体或其组合。The fusion proteins, conjugates or pharmaceutical compositions of the invention can be used in combination with other therapeutic agents. Other therapeutic agents include, for example, chemotherapeutic agents, immune checkpoint inhibitors, or antibodies targeting tumor-associated antigens. In one embodiment, the other therapeutic agent is an immune checkpoint inhibitor. In some embodiments, the immune checkpoint inhibitor is an anti-PD-L1 antibody, an anti-PD-1 antibody, an anti-CTLA4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, or a combination thereof.
本发明的融合蛋白、缀合物或药物组合物和其他治疗剂可以全部在一次施用或分开施用。当分开施用时(采用互相不同的施用方案的情况下),它们可以连续施用而不中断或以预定的间隔施用。本发明的融合蛋白或本发明的药物组合物还可以与放疗联合使用,例如包括向患者施用电离辐射,其早于、在其过程中和/或晚于本发明的融合蛋白或药物组合物的施用过程。The fusion protein, conjugate or pharmaceutical composition of the invention and the other therapeutic agent can be administered all at once or separately. When administered separately (in the case of mutually different administration regimens), they may be administered continuously without interruption or at predetermined intervals. The fusion protein of the present invention or the pharmaceutical composition of the present invention may also be used in combination with radiotherapy, for example comprising administering ionizing radiation to the patient earlier than, during and/or after the fusion protein of the present invention or pharmaceutical composition Application process.
试剂盒Reagent test kit
本发明还涉及一种试剂盒,其包括本发明的融合蛋白、编码其的多核苷酸或表达载体或者其组合,以及使用说明。试剂盒一般包括表明试剂盒内容物的预期用途和/或使用方法的标签。术语标签包括在试剂盒上或与试剂盒一起提供的或以其他方式随试剂盒提供的任何书面的或记录的材料。The present invention also relates to a kit, which includes the fusion protein of the present invention, its encoding polynucleotide or expression vector or a combination thereof, and instructions for use. Kits generally include a label indicating the intended use and/or method of use of the kit contents. The term label includes any written or recorded material provided on or with the kit or otherwise provided with the kit.
有益效果Beneficial effect
本发明的融合蛋白实现以下有益效果:The fusion protein of the present invention realizes the following beneficial effects:
1)以较高的亲和活性结合TGF-β和GITR;1) Bind TGF-β and GITR with higher affinity activity;
2)优异的激活GITR信号传导和抑制TGF-β信号传导的活性;2) Excellent activity of activating GITR signal transduction and inhibiting TGF-β signal transduction;
3)优异的T细胞激活活性;以及3) excellent T cell activation activity; and
4)优异的肿瘤抑制活性。4) Excellent tumor suppressive activity.
进一步的,本发明的融合蛋白可以包含TGFBR2-ECD突变体,与包含野生型TGFBR2-ECD的融合蛋白相比,包含TGFBR2-ECD突变体的融合蛋白在生产过程中不易发生断裂和降解,表现出更高的稳定性。Further, the fusion protein of the present invention may comprise a TGFBR2-ECD mutant. Compared with a fusion protein comprising a wild-type TGFBR2-ECD, a fusion protein comprising a TGFBR2-ECD mutant is less likely to be broken and degraded during production, showing Greater stability.
此外,本发明的融合蛋白靶向GITR和TGF-β双靶点,能够在激活GITR的同时阻遏TGF-β。相比于单靶向GITR的GITRL胞外结构域三聚体蛋白336B11和抗GITR激动剂抗体36E5,本发明的融合蛋白激活效应T细胞的活性更高。本发明首次提出了这种双靶向融合蛋白产品,在临床上有较大的应用潜力。In addition, the fusion protein of the present invention targets GITR and TGF-β dual targets, and can suppress TGF-β while activating GITR. Compared with the GITRL extracellular domain trimer protein 336B11 and the anti-GITR agonist antibody 36E5 that single-target GITR, the fusion protein of the present invention has a higher activity of activating effector T cells. The present invention proposes this dual-target fusion protein product for the first time, which has great application potential in clinic.
实施例Example
通过参考在此给出的一些具体实施例可获得对本发明的进一步的理解,这些实施例仅用于说明本发明,其无意于对本发明的范围做出任何限制。显然,可以对本发明作出多种改动和变化而不脱离本发明的实质,因此,这些改动和变化同样在本申请要求保护的范围内。A further understanding of the invention may be obtained by reference to certain specific examples which are given herein for illustration only and are not intended to limit the scope of the invention in any way. Apparently, various modifications and changes can be made to the present invention without departing from the essence of the present invention. Therefore, these modifications and changes are also within the protection scope of the present application.
除非另外指明,实施例部分中所用的试剂和仪器都是可商购的。Reagents and equipment used in the Examples section are commercially available unless otherwise indicated.
实施例1 GITR/TGF-β双靶向融合蛋白的构建与生产Example 1 Construction and production of GITR/TGF-β dual targeting fusion protein
将野生型TGFBR2-ECD(SEQ ID NO:4)和单链GITRL胞外结构域三聚体(SEQ ID NO:3,其包含由接头连接的3个GITRL胞外结构域)通过Fc片段连接,构建并产生GITR/TGF-β双靶向融合蛋白C2(SEQ ID NO:17)(见表2)。The wild-type TGFBR2-ECD (SEQ ID NO: 4) and the single-chain GITRL extracellular domain trimer (SEQ ID NO: 3, which contains 3 GITRL extracellular domains connected by a linker) are connected through the Fc fragment, Construct and produce GITR/TGF-β dual targeting fusion protein C2 (SEQ ID NO: 17) (see Table 2).
带有野生型TGFBR2-ECD的融合蛋白C2(如实施例2所示)容易发生降解,产生包含单链GITRL胞外结构域三聚体和Fc片段的大片段和含有TGFBR2-ECD的小片段(见图1)。经质谱检测发现,融合蛋白发生降解的位点主要位于野生型TGFBR2-ECD的N端的三个区域,即第一区域Q-K-S和第二区域N-NDM-I(其中“-”表示断裂点),以及第三区域“DNNG”(见表1)。此外,由于断裂产生的主要降解物中Fc部分结构完整,以常规的Protein A介质无法有效去除降解产物,对下游纯化工艺的开发带来巨大挑战,严重影响分子成药性开发。因此在采用TGFBR2-ECD作为融合蛋白一部分时,不管在融合蛋白中的顺序如何,都需要针对三个区域或三个区域及其上下游序列同时进行针对性的有效突变,从而获得高质量的融合蛋白。The fusion protein C2 (as shown in Example 2) with wild-type TGFBR2-ECD is prone to degradation, resulting in a large fragment comprising a single-chain GITRL extracellular domain trimer and an Fc fragment and a small fragment containing TGFBR2-ECD ( see picture 1). It was found by mass spectrometry that the degradation site of the fusion protein was mainly located in three regions of the N-terminus of wild-type TGFBR2-ECD, namely the first region Q-K-S and the second region N-NDM-I (where "-" indicates the break point), And the third area "DNNG" (see Table 1). In addition, due to the complete structure of the Fc part of the main degradation products produced by the fragmentation, conventional Protein A media cannot effectively remove the degradation products, which poses a huge challenge to the development of downstream purification processes and seriously affects the development of druggability of molecules. Therefore, when using TGFBR2-ECD as a part of the fusion protein, regardless of the order in the fusion protein, it is necessary to simultaneously carry out targeted and effective mutations for the three regions or the three regions and their upstream and downstream sequences, so as to obtain high-quality fusion protein.
基于野生型TGFBR2-ECD的序列(SEQ ID NO:4),针对上述三个区域及其上下游区域进行突变改造,获得TGFBR2-ECD突变体用于融合蛋白的制备。具体地,向野生型TGFBR2-ECD的三个区域的中引入亲水性的甘氨酸(G)和/或亲水性且可发生潜在O- 糖基化修饰的丝氨酸(S),获得的TGFBR2-ECD突变体的氨基酸序列如SEQ ID NO:5所示;向野生型TGFBR2-ECD的上述三个区域及其上下游区域引入广泛的亲水性氨基酸,特别是可发生潜在O-糖修饰的丝氨酸(S)和苏氨酸(T),获得的TGFBR2-ECD突变体的氨基酸序列如SEQ ID NO:6所示(见表1)。Based on the sequence of wild-type TGFBR2-ECD (SEQ ID NO: 4), the above three regions and their upstream and downstream regions were mutated and transformed to obtain TGFBR2-ECD mutants for the preparation of fusion proteins. Specifically, hydrophilic glycine (G) and/or hydrophilic serine (S) that can undergo potential O-glycosylation modification are introduced into the three regions of wild-type TGFBR2-ECD, and the obtained TGFBR2- The amino acid sequence of the ECD mutant is shown in SEQ ID NO:5; a wide range of hydrophilic amino acids are introduced into the above three regions of wild-type TGFBR2-ECD and their upstream and downstream regions, especially serine, which can undergo potential O-sugar modification (S) and threonine (T), the amino acid sequence of the obtained TGFBR2-ECD mutant is shown in SEQ ID NO: 6 (see Table 1).
表1Table 1
TGFBR2-ECDTGFBR2-ECD 序列结构sequence structure SEQ ID NO:SEQ ID NO:
野生型Wild type IPPHV QKSV NNDMIVT DNNGAVKF……NIIFSEEYNTSNPD IPPHV QKS V NNDMI VT DNNG AVKF…NIIFSEEYNTSNPD 44
突变体mutant IPPHV GGSV GGSGIVT GGSGAVKF……NIIFSEEYNTSNPD IPPHV GGS V GGSG IVT GGSG AVKF…NIIFSEEYNTSNPD 55
突变体mutant TPPHT QTST NNSMITT GTSGATKY……NIIFSEEYNTSNPD TPPHT QTS T NNSMI TT GTSG ATKY…NIIFSEEYNTSNPD 66
注:三个区域用下划线示出;TGFBR2-ECD突变体中包含的氨基酸突变以加粗字体示出。Note: Three regions are underlined; amino acid mutations contained in TGFBR2-ECD mutants are shown in bold.
最终构建GITR/TGF-β双靶向融合蛋白C12(TGFBR2-ECD突变体位于融合蛋白的N端,式(1),SEQ ID NO:15)、C15(TGFBR2-ECD突变体位于融合蛋白的C端,式(2),SEQ ID NO:16)和C2(野生型TGFBR2-ECD位于融合蛋白的C端,式(2),SEQ ID NO:17)(见表2)。Finally construct GITR/TGF-β dual-targeting fusion protein C12 (the TGFBR2-ECD mutant is located at the N-terminus of the fusion protein, formula (1), SEQ ID NO: 15), C15 (the TGFBR2-ECD mutant is located at the C-terminal of the fusion protein). terminal, formula (2), SEQ ID NO:16) and C2 (wild-type TGFBR2-ECD is located at the C-terminal of the fusion protein, formula (2), SEQ ID NO:17) (see Table 2).
同时构建并产生单靶向蛋白336B11(氨基酸序列参见CN107406878A中的336B11)和B22作为对比蛋白,其中336B11为GITR单靶向蛋白(单链GITRL胞外结构域三聚体融合至Fc片段的N端),B22为TGF-β单靶向蛋白(野生型TGFBR2-ECD融合至Fc片段的N端)(见表2)。Simultaneously construct and produce single-targeting protein 336B11 (see 336B11 in CN107406878A for amino acid sequence) and B22 as contrasting proteins, wherein 336B11 is GITR single-targeting protein (single-chain GITRL extracellular domain trimer fused to the N-terminal of Fc fragment ), B22 is a TGF-β single-targeting protein (wild-type TGFBR2-ECD fused to the N-terminus of the Fc fragment) (see Table 2).
利用基因全合成及分子克隆技术将编码GITR/TGF-β双靶向融合蛋白或者对比蛋白(均在N端包含分泌信号肽,信号肽的示例性序列示于SEQ ID NO:13和SEQ ID NO:14)的多核苷酸序列克隆至表达载体pCDNA3.4(购自Thermo Fisher)中,将获得的真核表达载体瞬转ExpiCHOS进行常规表达,经过10天左右的无血清细胞培养,收集培养上清用Protein A介质柱层析进行纯化,得到的本发明融合蛋白和对比蛋白的结构示于表2。Utilize gene total synthesis and molecular cloning technology to encode GITR/TGF-β dual-targeting fusion protein or contrast protein (both contain secretion signal peptide at N-terminus, the exemplary sequence of signal peptide is shown in SEQ ID NO: 13 and SEQ ID NO The polynucleotide sequence of :14) was cloned into the expression vector pCDNA3.4 (purchased from Thermo Fisher), and the obtained eukaryotic expression vector was transiently transformed into ExpiCHOS for routine expression. After about 10 days of serum-free cell culture, the collected cultured Purified by Protein A medium column chromatography, the structures of the fusion protein of the present invention and the comparison protein obtained are shown in Table 2.
表2Table 2
Figure PCTCN2022105175-appb-000001
Figure PCTCN2022105175-appb-000001
实施例2 SDS-PAGE检测GITR/TGF-β双靶向融合蛋白的纯度情况Example 2 SDS-PAGE detection of the purity of GITR/TGF-β dual targeting fusion protein
如实施例1所述的融合蛋白C15和C2经Protein A介质一步纯化后,通过还原性蛋白凝胶电泳(SDS-PAGE)进行蛋白纯度检测。After the fusion proteins C15 and C2 as described in Example 1 were purified in one step by Protein A medium, the protein purity was detected by reducing protein gel electrophoresis (SDS-PAGE).
还原性蛋白凝胶电泳:取样品加入适量上样缓冲液,其中一份样品加入DTT进行还原,还原样品70℃水浴加热10分钟后,上样2μg,经10%蛋白凝胶电泳分离。实验结果如图1所示,泳道1为C15(含有TGFBR2-ECD突变体),泳道2为C2(含有野生型TGFBR2-ECD)。Reducing protein gel electrophoresis: take a sample and add an appropriate amount of sample buffer, add DTT to one of the samples for reduction, heat the reduced sample in a 70°C water bath for 10 minutes, load 2 μg of the sample, and separate by 10% protein gel electrophoresis. The experimental results are shown in FIG. 1 , lane 1 is C15 (containing TGFBR2-ECD mutant), and lane 2 is C2 (containing wild-type TGFBR2-ECD).
由图1可知,泳道1中显示一条主要蛋白条带,其与目的蛋白C15(SEQ ID NO:15)的预期分子量一致。泳道2中显示三条主要蛋白条带,其中分子量最大的条带与目的蛋白C2(SEQ ID NO:16)的预期分子量一致,另外两条蛋白条带为C2在TGFBR2-ECD的N端发生分子间断裂而产生的降解产物,分别对应于包含单链GITRL胞外结构域三聚体和Fc片段的大片段(降解条带1)和含有TGFBR2-ECD的小片段(降解条带2)。由此可见,C2相比于C15发生显著降解。通过包含TGFBR2-ECD突变体,GITR/TGF-β双靶向融合蛋白的降解得到有效控制。As can be seen from Figure 1, a major protein band is shown in swimming lane 1, which is consistent with the expected molecular weight of the target protein C15 (SEQ ID NO: 15). Lane 2 shows three main protein bands, the band with the largest molecular weight is consistent with the expected molecular weight of the target protein C2 (SEQ ID NO: 16), and the other two protein bands are C2 in the N-terminus of TGFBR2-ECD. The degradation products produced by the fragmentation correspond to the large fragment (degradation band 1) containing single-chain GITRL ectodomain trimer and Fc fragment (degradation band 1) and the small fragment (degradation band 2) containing TGFBR2-ECD, respectively. It can be seen that C2 is significantly degraded compared to C15. Degradation of the GITR/TGF-β dual targeting fusion protein was effectively controlled by inclusion of the TGFBR2-ECD mutant.
实施例3 GITR/TGF-β双靶向融合蛋白体外结合TGF-β的活性Example 3 The activity of GITR/TGF-β dual-targeting fusion protein binding to TGF-β in vitro
测试目的:通过ELISA检测单靶向和双靶向融合蛋白对TGF-β的结合活性,来比较不同结构设计的双靶向融合蛋白样品体外对TGF-β结合的优劣,其中B22用作阳性对照。Test purpose: To detect the binding activity of single-target and dual-target fusion proteins to TGF-β by ELISA, to compare the pros and cons of dual-target fusion protein samples with different structural designs for TGF-β binding in vitro, and B22 is used as a positive control.
测试样品:如实施例1制备的B22、C15、C12和C2。Test samples: B22, C15, C12 and C2 prepared as in Example 1.
测试步骤:Test steps:
a.将0.5μg/ml浓度的人源TGF-β(购自Acro)直接包被至96孔板,100μl/孔,4℃过夜。a. Human TGF-β (purchased from Acro) at a concentration of 0.5 μg/ml was directly coated onto a 96-well plate, 100 μl/well, overnight at 4°C.
b.250μl/孔1×PBS洗涤3次,加入250μl/孔2%BSA-PBS,37℃封闭2小时。b. Wash 3 times with 250 μl/well 1×PBS, add 250 μl/well 2% BSA-PBS, and block at 37°C for 2 hours.
c.250μl/孔1×PBS洗涤3次,加入梯度稀释的样品B22、C15、C12和C2,100μl/孔,37℃孵育1小时。c. Wash 3 times with 250 μl/well 1×PBS, add serially diluted samples B22, C15, C12 and C2, 100 μl/well, and incubate at 37°C for 1 hour.
d.250μl/孔1×PBST洗涤3次,加入100μl/孔anti-human Fc antibody-HRP(1:5000),37℃孵育1小时。d. Wash 3 times with 250 μl/well 1×PBST, add 100 μl/well anti-human Fc antibody-HRP (1:5000), and incubate at 37°C for 1 hour.
e.250μl/孔1×PBST洗涤3次,加入100μl/孔TMB,37℃孵育10分钟,2M盐酸终止反应。e. Wash 3 times with 250 μl/well 1×PBST, add 100 μl/well TMB, incubate at 37°C for 10 minutes, and stop the reaction with 2M hydrochloric acid.
f.酶标仪上检测450nm吸收值,Graphpad Prism5分析数据。f. Detect the absorbance at 450nm on a microplate reader, and analyze the data with Graphpad Prism5.
结果:融合蛋白体外结合TGF-β的ELISA结果如图2和表3所示,结果显示C2、C12和C15均具有TGF-β结合活性。从EC 50(nM)来看,C12和C15的TGF-β结合活性相当,并且与B22相当,表明双靶向蛋白C12和C15对TGF-β的结合活性与野生型TGFBR2-ECD相当。此外,C12和C15对TGF-β的结合活性均优于C2,其中C15组合顺序与C2一致,但C12、C15均采用了TGFBR2-ECD的突变体,而C2采用的野生型TGFBR2-ECD。如实施例2所示,包含野生型TGFBR2-ECD的双靶向融合蛋白C2容易发生降解,这可能是导致其TGF-β结合活性较低的原因之一。 Results: The ELISA results of the fusion protein binding to TGF-β in vitro are shown in Figure 2 and Table 3, and the results showed that C2, C12 and C15 all had TGF-β binding activity. In terms of EC50 (nM), the TGF-β binding activities of C12 and C15 were comparable and comparable to B22, indicating that the binding activity of dual targeting proteins C12 and C15 to TGF-β was comparable to that of wild-type TGFBR2-ECD. In addition, the binding activity of C12 and C15 to TGF-β was better than that of C2, and the combination sequence of C15 was consistent with that of C2, but both C12 and C15 used the mutant TGFBR2-ECD, while C2 used the wild-type TGFBR2-ECD. As shown in Example 2, the dual-targeting fusion protein C2 containing wild-type TGFBR2-ECD is prone to degradation, which may be one of the reasons for its low TGF-β binding activity.
表3table 3
Figure PCTCN2022105175-appb-000002
Figure PCTCN2022105175-appb-000002
实施例4 GITR/TGF-β双靶向融合蛋白体外结合hGITR的亲和力和动力学Example 4 Affinity and kinetics of GITR/TGF-β dual-targeting fusion protein binding to hGITR in vitro
检测目的:通过
Figure PCTCN2022105175-appb-000003
Octet RED96测定GITR/TGF-β双靶向融合蛋白对hGITR的亲和力。
Detection purpose: pass
Figure PCTCN2022105175-appb-000003
Octet RED96 measures the affinity of GITR/TGF-β dual targeting fusion protein to hGITR.
检测样品:如实施例1制备的C12和336B11以及抗GITR激动剂抗体36E5(氨基酸序列参见CN103951753A)。Test samples: C12 and 336B11 prepared as in Example 1 and anti-GITR agonist antibody 36E5 (see CN103951753A for the amino acid sequence).
检测流程:Detection process:
a.偶联配体:本方法采用HIS1K(Anti-Penta-HIS biosensor,ForteBio)芯片,提前用SD缓冲液浸泡芯片10-15分钟,以50μg/ml浓度加载配体(hGITR),预设时长300秒,配体Loading预设值1nm,用运行缓冲液(SD缓冲液)浸润芯片至基线平稳,去除游离配体。a. Coupled ligand: This method uses HIS1K (Anti-Penta-HIS biosensor, ForteBio) chip, soak the chip with SD buffer for 10-15 minutes in advance, and load the ligand (hGITR) at a concentration of 50 μg/ml for a preset time For 300 seconds, the ligand loading preset value is 1nm, and the chip is infiltrated with running buffer (SD buffer) until the baseline is stable to remove free ligand.
b.动力学实验:将C12、336B11和36E5用SD缓冲液各自稀释至50nM。以样品为分析物,hGITR为配体,进行亲和力检测实验。配体与分析物结合时间(Association)200秒,分析物解离时间(Dissociation)300秒。b. Kinetic experiment: C12, 336B11 and 36E5 were each diluted to 50 nM with SD buffer. Using the sample as the analyte and hGITR as the ligand, the affinity detection experiment was carried out. The ligand and analyte binding time (Association) is 200 seconds, and the analyte dissociation time (Dissociation) is 300 seconds.
c.以ForteBio Data Analysis 9.0采用静态拟合模型进行分析。c. Analyze with ForteBio Data Analysis 9.0 using static fitting model.
检测结果:动力学分析过程如图3所示,亲和力结果如表4所示。从表4可知,C12对hGITR的亲和力显著优于36E5,并且与336B11的亲和力相当。Detection results: the kinetic analysis process is shown in Figure 3, and the affinity results are shown in Table 4. It can be seen from Table 4 that the affinity of C12 to hGITR is significantly better than that of 36E5 and comparable to that of 336B11.
表4Table 4
样品sample K D(M) K D (M) k on(1/Ms) k on (1/Ms) k dis(1/s) k dis (1/s)
C12C12 5.17E-105.17E-10 6.34E+056.34E+05 3.28E-043.28E-04
336B11336B11 5.24E-105.24E-10 5.39E+055.39E+05 2.82E-042.82E-04
36E536E5 3.56E-093.56E-09 2.07E+052.07E+05 7.38E-047.38E-04
实施例5 GITR/TGF-β双靶向融合蛋白体外结合CHOS-hGITR细胞的活性Example 5 The activity of GITR/TGF-β dual-targeting fusion protein binding to CHOS-hGITR cells in vitro
检测样品:如实施例1制备的C12和C15。Test samples: C12 and C15 prepared as in Example 1.
检测流程:Detection process:
a.以膜蛋白hGITR全序列(Uniprot ID:Q9Y5U5)重组载体转染CHOS细胞,转染24小时后加压筛选,采用选择培养基(CD FortiCHO TM培养基+15μmol/L MSX+10μg/ml嘌呤霉素),直至细胞活率上升。采用有限稀释法筛选单克隆细胞株,将单克隆细胞株与336B11共孵育,随后去除上清,加入anti-human IgG-488(Invitrogen,1:2000)作为检测抗体,使用流式细胞仪筛选出平均荧光强度(MFI)较高且峰形较好的细胞株,作为稳定转染CHOS-hGITR细胞株待用。 a. CHOS cells were transfected with the recombinant vector of the full sequence of membrane protein hGITR (Uniprot ID: Q9Y5U5), pressurized selection after 24 hours of transfection, and selected medium (CD FortiCHO TM medium + 15 μmol/L MSX + 10 μg/ml purine Mycin), until the cell viability increased. The monoclonal cell line was screened by the limiting dilution method, and the monoclonal cell line was incubated with 336B11, and then the supernatant was removed, and anti-human IgG-488 (Invitrogen, 1:2000) was added as a detection antibody, and was screened by flow cytometry. Cell lines with higher mean fluorescence intensity (MFI) and better peak shape are used as stable transfection CHOS-hGITR cell lines.
b.将样品C12和C15按照浓度梯度与CHOS-hGITR单克隆细胞株在室温下孵育1小时;用PBS冲洗2次,加入anti-human IgG-488(Invitrogen,1:2000)与细胞在室温下孵育45分钟,再用PBS冲洗2次;然后用流式细胞仪检测荧光信号并计算EC 50b. Incubate samples C12 and C15 with CHOS-hGITR monoclonal cell line according to the concentration gradient for 1 hour at room temperature; rinse twice with PBS, add anti-human IgG-488 (Invitrogen, 1:2000) and mix with cells at room temperature Incubate for 45 minutes, and then wash twice with PBS; then detect the fluorescent signal by flow cytometry and calculate the EC 50 .
检测结果:结果如图4和表5所示,从EC 50(nM)的结果可以看出C12对CHOS-hGITR细胞的结合活性略优于C15。 Test results: the results are shown in Figure 4 and Table 5. From the results of EC 50 (nM), it can be seen that the binding activity of C12 to CHOS-hGITR cells is slightly better than that of C15.
表5table 5
Figure PCTCN2022105175-appb-000004
Figure PCTCN2022105175-appb-000004
实施例6 GITR/TGF-β双靶向融合蛋白体外激活GITR的活性Example 6 GITR/TGF-β dual targeting fusion protein activates GITR activity in vitro
GITR Blockade Bioassay是依赖细胞的生物发光检测方法,其检测原理是:当向表达GITR的遗传工程化的细胞系GS-H3/GITR细胞(金斯瑞提供)加入GITR激动剂时,引起GITR激活和NFAT控制下的萤光素酶基因的表达,随后可以通过加入萤光素酶 检测试剂(Bio-Glo,Promega;含有萤光素酶底物萤光素和反应缓冲液)来检测萤光素酶催化底物产生的发光(luminescence)信号,从而评价GITR激动剂激活GITR的活性。GITR Blockade Bioassay is a cell-dependent bioluminescent detection method. Its detection principle is: when GITR agonists are added to the genetically engineered cell line GS-H3/GITR cells expressing GITR (provided by GenScript), GITR activation and Expression of the luciferase gene under the control of NFAT, which can subsequently be detected by adding a luciferase detection reagent (Bio-Glo, Promega; containing the luciferase substrate luciferin and reaction buffer) The luminescence signal generated by the catalytic substrate can be used to evaluate the activity of GITR agonists to activate GITR.
检测样品:如实施例1制备的C12和C15,抗GITR激动剂抗体36E5(阳性对照)以及IgG同种型(阴性对照)。Test samples: C12 and C15 prepared as in Example 1, anti-GITR agonist antibody 36E5 (positive control) and IgG isotype (negative control).
GITR激动剂功能活性检测实验步骤如下:The experimental steps for detecting the functional activity of GITR agonists are as follows:
a.离心收集靶细胞,使用含有10%FBS的EMEM Nutrient Mixture培养基重悬细胞并调整细胞密度,接种于384孔实验板中。a. Collect the target cells by centrifugation, resuspend the cells in EMEM Nutrient Mixture medium containing 10% FBS and adjust the cell density, and inoculate them in 384-well experimental plates.
b.将实验板转移至细胞培养箱(37℃/5%CO 2)中孵育16小时。 b. Transfer the experimental plate to a cell culture incubator (37° C./5% CO 2 ) and incubate for 16 hours.
c.使用测定缓冲液配置2×浓度的对照品和供试品。c. Use the assay buffer to configure 2×concentrations of the reference substance and the test substance.
d.离心收集效应细胞,使用测定缓冲液重悬细胞并调整细胞密度。d. Collect the effector cells by centrifugation, resuspend the cells in assay buffer and adjust the cell density.
e.将完成孵育的384孔实验板从细胞培养箱中取出并移除培养基,依次转移对照品和供试品工作液及效应细胞悬液至384孔实验板中。e. Take out the incubated 384-well experimental plate from the cell culture incubator, remove the medium, and transfer the control substance, the test product working solution and the effector cell suspension to the 384-well experimental plate in turn.
f.将实验板转移至细胞培养箱(37℃/5%CO 2)中孵育约6小时。 f. Transfer the experimental plate to a cell culture incubator (37° C./5% CO 2 ) and incubate for about 6 hours.
g.配制荧光素酶底物工作液。g. Prepare luciferase substrate working solution.
h.将完成孵育的384孔实验板从细胞培养箱中取出,转移荧光素酶底物工作液至384孔实验板中。h. Take out the incubated 384-well experimental plate from the cell culture incubator, and transfer the luciferase substrate working solution to the 384-well experimental plate.
i.在PHERA Star FSX上读取化学发光值,记录数据。i. Read the chemiluminescence value on PHERA Star FSX and record the data.
j.数据分析:依据相对化学发光信号值(Relative Luminescence Unit)和最终检测浓度的对应关系建立相应的量效曲线图。j. Data analysis: According to the corresponding relationship between the relative chemiluminescence signal value (Relative Luminescence Unit) and the final detection concentration, the corresponding dose-effect curve is established.
检测结果如图5和表6所示,抗GITR激动剂抗体36E5可有效结合GITR,并激活下游通路产生信号,阴性对照IgG同种型无法和GITR结合,无法激活下游通路产生信号。C12和C15均可以激活下游信号通路,活性相当,并且样品的量效曲线EC 50以及量效最大值都明显优于36E5。 The test results are shown in Figure 5 and Table 6. The anti-GITR agonist antibody 36E5 can effectively bind to GITR and activate downstream pathways to generate signals. The negative control IgG isotype cannot bind to GITR and cannot activate downstream pathways to generate signals. Both C12 and C15 can activate downstream signaling pathways with similar activities, and the dose-response curve EC 50 and dose-response maximum of the samples are significantly better than 36E5.
表6Table 6
Figure PCTCN2022105175-appb-000005
Figure PCTCN2022105175-appb-000005
实施例7 GITR/TGF-β双靶向融合蛋白的体外抑制TGF-β活性Example 7 In vitro inhibition of TGF-β activity by GITR/TGF-β dual targeting fusion protein
TGF-β受体报告基因实验(TGF-β receptor Reporter Gene Bioassay)是依赖基因工程化的细胞系的生物发光检测方法。当TGF-β蛋白与细胞表面的TGF-β受体结合之后,受体介导的信号转导通路被激活从而诱导萤光素酶的表达,然后通过加入萤光素酶检 测试剂(Bio-Glo,Promega)来读取萤光素酶催化底物产生的发光信号。加入抗TGF-β抗体或抑制剂之后阻断TGF-β与其受体结合,导致发光信号降低。本实验采用A549/SBE细胞(金斯瑞提供)作为功能性细胞系,以抗体TGF-β抗体BMK-R2(金斯瑞)作为阳性对照,用以评估GITR/TGF-β双靶向融合蛋白体外阻断TGF-β的活性。TGF-β receptor reporter gene assay (TGF-β receptor Reporter Gene Bioassay) is a bioluminescent detection method that relies on genetically engineered cell lines. When the TGF-β protein binds to the TGF-β receptor on the cell surface, the receptor-mediated signal transduction pathway is activated to induce the expression of luciferase, and then by adding the luciferase detection reagent (Bio-Glo , Promega) to read the luminescent signal generated by the luciferase-catalyzed substrate. The addition of anti-TGF-β antibodies or inhibitors blocks the binding of TGF-β to its receptor, resulting in a decrease in the luminescent signal. In this experiment, A549/SBE cells (provided by GenScript) were used as a functional cell line, and the antibody TGF-β antibody BMK-R2 (GenScript) was used as a positive control to evaluate the GITR/TGF-β dual-targeting fusion protein Block the activity of TGF-β in vitro.
检测样品:如实施例1制备的C12和抗TGF-β抗体BMK-R2。Detection samples: C12 prepared as in Example 1 and anti-TGF-β antibody BMK-R2.
实验流程描述如下:The experimental procedure is described as follows:
a.离心收集靶细胞,使用含有10%FBS的F12K Nutrient Mixture培养基重悬细胞并调整细胞密度,接种于96孔实验板中。a. Collect the target cells by centrifugation, resuspend the cells in F12K Nutrient Mixture medium containing 10% FBS and adjust the cell density, and inoculate them in 96-well experimental plates.
b.将实验板转移至细胞培养箱(37℃/5%CO 2)中孵育16小时。 b. Transfer the experimental plate to a cell culture incubator (37° C./5% CO 2 ) and incubate for 16 hours.
c.使用测定缓冲液配置7×浓度的EC100对照品和供试品。c. Use the assay buffer to configure the EC100 reference substance and the test substance at a concentration of 7×.
d.将完成孵育的96孔实验板从细胞培养箱中取出,依次转移TGF-β1(EC100),对照品和供试品工作液至96孔实验板中。d. Take out the incubated 96-well experimental plate from the cell culture incubator, and transfer TGF-β1 (EC100), the control substance and the working solution of the test product to the 96-well experimental plate in turn.
e.将实验板转移至细胞培养箱(37℃/5%CO 2)中孵育约24小时。 e. Transfer the experimental plate to a cell culture incubator (37° C./5% CO 2 ) and incubate for about 24 hours.
f.配制荧光素酶底物工作液。f. Prepare luciferase substrate working solution.
g.将完成孵育的96孔实验板从细胞培养箱中取出,转移荧光素酶底物工作液至96孔实验板中。g. Take out the incubated 96-well experimental plate from the cell culture incubator, and transfer the luciferase substrate working solution to the 96-well experimental plate.
h.在PHERA Star FSX上读取化学发光值,记录数据。h. Read the chemiluminescence value on the PHERA Star FSX and record the data.
i.数据分析,依据相对化学发光信号值(Relative Luminescence Unit)和最终检测浓度的对应关系建立相应的量效曲线图。i. Data analysis, based on the corresponding relationship between the relative chemiluminescence signal value (Relative Luminescence Unit) and the final detection concentration to establish a corresponding dose-effect curve.
实验结果示于图6和表7。由图6和表7可知:阳性对照(BMK-R2)可有效和TGF-β结合,并抑制下游通路产生信号,其量效曲线的IC 50为0.226nM,阴性对照(Human IgG1)无法和TGF-β结合,无法抑制下游通路产生信号。样品C12可有效和TGF-β结合,并抑制下游通路产生信号,其量效曲线的IC 50为0.29nM,与BMK-R2活性相当。 The experimental results are shown in Figure 6 and Table 7. From Figure 6 and Table 7, it can be seen that the positive control (BMK-R2) can effectively bind to TGF-β, and inhibit the downstream pathway to generate signals, and the IC50 of the dose-effect curve is 0.226nM, and the negative control (Human IgG1) cannot bind to TGF-β. -β binding, unable to inhibit signaling from downstream pathways. Sample C12 can effectively bind to TGF-β, and inhibit downstream pathways to generate signals. The IC 50 of the dose-effect curve is 0.29nM, which is comparable to the activity of BMK-R2.
表7Table 7
Figure PCTCN2022105175-appb-000006
Figure PCTCN2022105175-appb-000006
实施例8 GITR/TGF-β双靶向融合蛋白体外活化效应T细胞的活性Example 8 GITR/TGF-β dual-targeting fusion protein activates the activity of effector T cells in vitro
检测样品:如实施例1制备的C12、C15、336B11以及抗GITR激动剂抗体36E5。Test samples: C12, C15, 336B11 and anti-GITR agonist antibody 36E5 prepared as in Example 1.
实验采用Raji细胞交联方式检测融合蛋白活化T细胞的活性,检测指标为IFN-γ,步骤如下:In the experiment, the Raji cell cross-linking method was used to detect the activity of the fusion protein to activate T cells. The detection index was IFN-γ, and the steps were as follows:
1.包被0.2μg/ml OKT3至96孔板上;1. Coat 0.2μg/ml OKT3 onto a 96-well plate;
2.加入分离的T细胞(购自睿智化学),培养4天;2. Add isolated T cells (purchased from Wisdom Chemicals) and culture for 4 days;
3.取出T细胞,停止预刺激,放置2天;3. Take out T cells, stop pre-stimulation, and leave for 2 days;
4.以1×10 5细胞/孔将Raji细胞加入预先结合2μg/ml OKT3的96孔中; 4. Add Raji cells at 1×10 5 cells/well to 96 wells pre-bound with 2 μg/ml OKT3;
5.加入样品蛋白C12、C15、36E5和336B11,最高浓度为50μg/ml或者10μg/ml,5倍梯度稀释,每个样品4个浓度梯度,每个浓度3个复孔;5. Add sample proteins C12, C15, 36E5 and 336B11, the highest concentration is 50 μg/ml or 10 μg/ml, 5-fold gradient dilution, 4 concentration gradients for each sample, and 3 replicate wells for each concentration;
6.混合T细胞,按照1E5/孔的浓度加入96孔板,培养4天;6. Mix T cells, add to 96-well plate at a concentration of 1E5/well, and culture for 4 days;
7.收集上清,用ELISA检测IFN-γ含量。7. Collect the supernatant and detect the IFN-γ content by ELISA.
结果如图7和图8所示:根据T细胞被激活后释放的细胞因子IFN-γ含量,可以得知C12活化效应T细胞的活性远高于阳性对照抗体36E5和单靶向蛋白336B11,并且高于C15。由此可见,本发明的双靶向融合蛋白C12活化效应T细胞的活性优于单靶向GITR的GITRL胞外结构域三聚体蛋白336B11和抗GITR激动剂抗体36E5。另外C12的效果优于C15,推测C12的结构对双靶向融合蛋白最为有利,C15可能在功能发挥上存在空间位阻,这与实施例5和实施例6结果一致。The results are shown in Figure 7 and Figure 8: According to the content of the cytokine IFN-γ released after T cells are activated, it can be known that the activity of C12 activated effector T cells is much higher than that of the positive control antibody 36E5 and the single targeting protein 336B11, and higher than C15. It can be seen that the dual-targeting fusion protein C12 of the present invention has better activity in activating effector T cells than the GITRL extracellular domain trimer protein 336B11 of single-targeting GITR and anti-GITR agonist antibody 36E5. In addition, the effect of C12 is better than that of C15. It is speculated that the structure of C12 is the most favorable for the dual-targeting fusion protein, and C15 may have steric hindrance in its function, which is consistent with the results of Examples 5 and 6.
实施例9 GITR/TGF-β双靶向融合蛋白的体内肿瘤抑制活性Example 9 In vivo tumor suppressive activity of GITR/TGF-β dual targeting fusion protein
在鼠源结肠癌MC38-OVA细胞株皮下移植C57BL/6-hGITR雌性转基因小鼠动物模型中测试本发明的融合蛋白的抗肿瘤作用。The anti-tumor effect of the fusion protein of the present invention was tested in the mouse model of colon cancer MC38-OVA cell line subcutaneously implanted with C57BL/6-hGITR female transgenic mice.
收集指数生长期的MC38-OVA细胞(中美冠科生物技术有限公司),以1×10 6细胞/小鼠的剂量于第0天(Day 0)在右侧背部皮下接种C57BL/6-hGITR小鼠,接种肿瘤细胞后第3天(Day 3),称量所有动物的体重。根据小鼠的体重对其分组,以保证不同组别间的体重相似,每组9只,共2组。腹腔注射给药,分别于接种肿瘤细胞后第3、6、9、12天给药,共给药四次。给药剂量试验组为C12 30mg/kg,对照组为等体积溶媒(PBS溶液),从第6天(Day 6)开始,每周测量两次小鼠的体重和肿瘤的大小。 Collect MC38-OVA cells in the exponential growth phase (Crown Biotechnology Co., Ltd.), and subcutaneously inoculate C57BL/6-hGITR on the right back on Day 0 at a dose of 1×10 6 cells/mouse For mice, on Day 3 after tumor cell inoculation (Day 3), the body weight of all animals was weighed. The mice were grouped according to their body weight to ensure that the body weights of different groups were similar, with 9 mice in each group, 2 groups in total. The intraperitoneal injection was administered on the 3rd, 6th, 9th, and 12th day after the inoculation of tumor cells, and the administration was administered four times in total. The dosage of the test group was C12 30mg/kg, and the control group was an equal volume of vehicle (PBS solution). From the 6th day (Day 6), the body weight and tumor size of the mice were measured twice a week.
结果显示,实验过程中,动物健康状态良好,小鼠对各治疗抗体耐受性良好。治疗组和对照组的肿瘤生长情况见图9。溶媒对照组小鼠在给药结束后第8天(Day 20)的平均肿瘤体积为897.16mm 3。融合蛋白C12(30mg/kg)治疗组在Day 20的平均肿瘤体积为363.98mm 3,相对肿瘤抑制率TGI(%)为59%,相较对照组统计学上有显著性差异(p=0.0498)。溶媒对照组小鼠在给药结束后第12天(Day 24)的平均肿瘤体积为2025.13mm 3。融合蛋白C12(30mg/kg)治疗组在Day 24的平均肿瘤体积为1039.94mm 3,相对肿瘤抑制率TGI(%)为49%。表明本发明的双靶向融合蛋白C12有显著的肿瘤抑制活性。 The results showed that during the experiment, the animals were in good health and the mice tolerated each treatment antibody well. The tumor growth of the treatment group and the control group is shown in FIG. 9 . The average tumor volume of mice in the vehicle control group was 897.16 mm 3 on the 8th day (Day 20) after administration. The average tumor volume of the fusion protein C12 (30mg/kg) treatment group on Day 20 was 363.98mm 3 , and the relative tumor inhibition rate TGI (%) was 59%, which was statistically significantly different from that of the control group (p=0.0498) . The average tumor volume of mice in the vehicle control group was 2025.13 mm 3 on day 12 (Day 24) after administration. The average tumor volume of the fusion protein C12 (30 mg/kg) treatment group on Day 24 was 1039.94 mm 3 , and the relative tumor inhibition rate TGI (%) was 49%. It shows that the dual targeting fusion protein C12 of the present invention has significant tumor suppressing activity.
相对肿瘤抑制率TGI(%)的计算公式如下:The calculation formula of relative tumor inhibition rate TGI (%) is as follows:
TGI(%)=(1-T/C)×100%,其中T和C分别为治疗组和对照组在某一特定时间点的肿瘤体积(TV)。TGI (%)=(1-T/C)×100%, where T and C are the tumor volume (TV) of the treatment group and the control group at a specific time point, respectively.
序列表sequence listing
Figure PCTCN2022105175-appb-000007
Figure PCTCN2022105175-appb-000007
Figure PCTCN2022105175-appb-000008
Figure PCTCN2022105175-appb-000008
Figure PCTCN2022105175-appb-000009
Figure PCTCN2022105175-appb-000009

Claims (15)

  1. 一种融合蛋白,其从N端至C端包含:A fusion protein comprising from N-terminus to C-terminus:
    第一多肽–第二多肽–第三多肽  式(1);The first polypeptide-the second polypeptide-the third polypeptide Formula (1);
    或者or
    第三多肽–第二多肽–第一多肽  式(2);The third polypeptide-the second polypeptide-the first polypeptide Formula (2);
    其中in
    所述第一多肽包含TGF-βRⅡ的胞外结构域(TGFBR2-ECD);The first polypeptide comprises an extracellular domain of TGF-βRII (TGFBR2-ECD);
    所述第二多肽从N端至C端具有如下结构:The second polypeptide has the following structure from N-terminus to C-terminus:
    Lk 1–Dd–Lk 2Lk 1 –Dd–Lk 2 ,
    其中in
    Dd为二聚化结构域;Dd is a dimerization domain;
    Lk 1和Lk 2各自独立地为接头或不存在;并且 Lk 1 and Lk 2 are each independently a linker or absent; and
    所述第三多肽从N端至C端具有如下结构:The third polypeptide has the following structure from the N-terminus to the C-terminus:
    Sd 1–Ln 1–Sd 2–Ln 2–Sd 3Sd 1 -Ln 1 -Sd 2 -Ln 2 -Sd 3 ,
    其中in
    Sd 1、Sd 2和Sd 3各自独立地为GITRL胞外结构域; Sd 1 , Sd 2 and Sd 3 are each independently an extracellular domain of GITRL;
    Ln 1和Ln 2各自独立地为接头或不存在。 Ln 1 and Ln 2 are each independently a linker or absent.
  2. 权利要求1的融合蛋白,其中所述TGFBR2-ECD包含SEQ ID NO:4、SEQ ID NO:5或SEQ ID NO:6的氨基酸序列;优选地,所述TGFBR2-ECD包含SEQ ID NO:5的氨基酸序列。The fusion protein of claim 1, wherein the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6; preferably, the TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:5 amino acid sequence.
  3. 权利要求1或2的融合蛋白,其中所述GITRL胞外结构域包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列;优选地,所述GITRL胞外结构域包含SEQ ID NO:2的氨基酸序列。The fusion protein of claim 1 or 2, wherein the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:1 or SEQ ID NO:2; preferably, the GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO:2 amino acid sequence.
  4. 权利要求1-3中任一项的融合蛋白,其中所述第三多肽包含SEQ ID NO:3的氨基酸序列。The fusion protein according to any one of claims 1-3, wherein the third polypeptide comprises the amino acid sequence of SEQ ID NO:3.
  5. 权利要求1-4中任一项的融合蛋白,其中所述二聚化结构域从N端至C端包含免疫球蛋白的CH2和CH3结构域,优选人IgG1的CH2和CH3结构域;更优选地,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列。The fusion protein according to any one of claims 1-4, wherein the dimerization domain comprises the CH2 and CH3 domains of an immunoglobulin from the N-terminus to the C-terminus, preferably the CH2 and CH3 domains of human IgG1; more preferably Preferably, the CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8.
  6. 权利要求1-5中任一项的融合蛋白,其中所述第二多肽从N端至C端具有如下结构:The fusion protein according to any one of claims 1-5, wherein the second polypeptide has the following structure from the N-terminus to the C-terminus:
    Lk 1–Dd–Lk 2Lk 1 -Dd -Lk 2 ;
    其中in
    Dd为二聚化结构域,其从N端至C端包含免疫球蛋白的铰链区或其部分、CH2和CH3结构域,优选人IgG1的Fc片段;优选地,所述铰链区或其部分包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列;Dd is a dimerization domain comprising from N-terminus to C-terminus the hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin, preferably the Fc fragment of human IgG1; preferably, the hinge region or part thereof comprises The amino acid sequence of SEQ ID NO:9, said CH2 and CH3 domains comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8;
    Lk 1和Lk 2各自独立地为接头。 Lk 1 and Lk 2 are each independently a linker.
  7. 权利要求1-6中任一项的融合蛋白,其中The fusion protein of any one of claims 1-6, wherein
    所述TGFBR2-ECD包含SEQ ID NO:4、SEQ ID NO:5或SEQ ID NO:6的氨基酸序列;The TGFBR2-ECD comprises the amino acid sequence of SEQ ID NO:4, SEQ ID NO:5 or SEQ ID NO:6;
    所述GITRL胞外结构域包含SEQ ID NO:1或SEQ ID NO:2的氨基酸序列;并且The GITRL extracellular domain comprises the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2; and
    所述二聚化结构域从N端至C端包含免疫球蛋白的铰链区或其部分、CH2和CH3结构域,所述铰链区或其部分包含SEQ ID NO:9的氨基酸序列,所述CH2和CH3结构域包含SEQ ID NO:7或SEQ ID NO:8的氨基酸序列。The dimerization domain comprises a hinge region or part thereof, CH2 and CH3 domains of an immunoglobulin from the N-terminus to the C-terminus, the hinge region or part thereof comprises the amino acid sequence of SEQ ID NO: 9, the CH2 and the CH3 domain comprise the amino acid sequence of SEQ ID NO:7 or SEQ ID NO:8.
  8. 权利要求1-7中任一项的融合蛋白,其中所述接头包含5-25个氨基酸,所述氨基酸各自独立地选自甘氨酸、丝氨酸和丙氨酸;优选地,所述接头包含(GGGGS) n,其中n选自1-10的整数;更优选地,所述接头包含SEQ ID NO:10、SEQ ID NO:11或SEQ ID NO:12的氨基酸序列。 The fusion protein according to any one of claims 1-7, wherein the linker comprises 5-25 amino acids, each of which is independently selected from glycine, serine and alanine; preferably, the linker comprises (GGGGS) n , wherein n is an integer selected from 1-10; more preferably, the linker comprises the amino acid sequence of SEQ ID NO:10, SEQ ID NO:11 or SEQ ID NO:12.
  9. 权利要求1的融合蛋白,其包含SEQ ID NO:15或SEQ ID NO:16的氨基酸序列。The fusion protein of claim 1, which comprises the amino acid sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
  10. 一种多核苷酸,其编码权利要求1-9中任一项的融合蛋白。A polynucleotide encoding the fusion protein of any one of claims 1-9.
  11. 一种表达载体,其包含权利要求10的多核苷酸。An expression vector comprising the polynucleotide of claim 10.
  12. 一种宿主细胞,其包含权利要求10的多核苷酸或权利要求11的表达载体;A host cell comprising the polynucleotide of claim 10 or the expression vector of claim 11;
    优选地,所述宿主细胞为真核细胞;更优选地,所述宿主细胞为酵母细胞、哺乳动物细胞或免疫效应细胞;进一步优选地,所述宿主细胞为效应T细胞。Preferably, the host cell is a eukaryotic cell; more preferably, the host cell is a yeast cell, a mammalian cell or an immune effector cell; further preferably, the host cell is an effector T cell.
  13. 一种缀合物,其包含与至少一种治疗剂缀合的权利要求1-9中任一项的融合蛋白;优选地,所述治疗剂选自可检测标记物、化疗剂、细胞毒素、放射性核素、免疫检查点抑制剂、细胞因子和酶。A conjugate comprising the fusion protein of any one of claims 1-9 conjugated to at least one therapeutic agent; preferably, the therapeutic agent is selected from the group consisting of detectable markers, chemotherapeutic agents, cytotoxins, Radionuclides, immune checkpoint inhibitors, cytokines and enzymes.
  14. 一种药物组合物,其包含:A pharmaceutical composition comprising:
    (ⅰ)权利要求1-9中任一项的融合蛋白,以及(i) the fusion protein of any one of claims 1-9, and
    (ⅱ)药学上可接受的载剂;(ii) a pharmaceutically acceptable carrier;
    优选的,所述药物组合物制备为注射液。Preferably, the pharmaceutical composition is prepared as an injection.
  15. 权利要求1-9中任一项的融合蛋白、权利要求10的多核苷酸、权利要求13的缀合物或者权利要求14的药物组合物在制备药物中的用途;Use of the fusion protein according to any one of claims 1-9, the polynucleotide according to claim 10, the conjugate according to claim 13 or the pharmaceutical composition according to claim 14 in the preparation of medicines;
    优选地,所述药物用于治疗和/或预防癌症;Preferably, the drug is used to treat and/or prevent cancer;
    或者or
    所述药物用于激活GITR信号传导和抑制TGF-β信号传导;或者The drug is used to activate GITR signaling and inhibit TGF-beta signaling; or
    所述药物用于在有需要的受试者中诱导、促进、增强、激活或延长免疫反应;或者The medicament is used to induce, promote, enhance, activate or prolong an immune response in a subject in need thereof; or
    所述药物用于诱导、促进、增强、激活或延长有需要的受试者中针对肿瘤或肿瘤细胞的免疫反应;The medicament is for inducing, promoting, enhancing, activating or prolonging an immune response against a tumor or tumor cells in a subject in need thereof;
    优选地,所述癌症选自腺癌、淋巴瘤、母细胞瘤、黑素瘤、肉瘤、白血病、鳞状细胞癌、小细胞肺癌、非小细胞肺癌、胃肠癌、霍奇金和非霍奇金淋巴瘤、胰腺癌、胶质母细胞瘤、神经胶质瘤、宫颈癌、卵巢癌、肝癌、肝细胞癌、膀胱癌、乳癌、结肠癌、结肠直肠癌、子宫内膜癌、骨髓瘤、多发性骨髓瘤、唾液腺癌、肾癌、肾细胞癌、威尔曼瘤、基底细胞癌、黑素瘤、前列腺癌、外阴癌、甲状腺癌、睾丸癌、食管癌和头颈癌。Preferably, the cancer is selected from adenocarcinoma, lymphoma, blastoma, melanoma, sarcoma, leukemia, squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, Hodgkin's and non-Hodgkin's Chickkin's lymphoma, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, hepatocellular carcinoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma , multiple myeloma, salivary gland cancer, kidney cancer, renal cell carcinoma, Wellman's tumor, basal cell carcinoma, melanoma, prostate cancer, vulvar cancer, thyroid cancer, testicular cancer, esophageal cancer, and head and neck cancer.
PCT/CN2022/105175 2021-07-14 2022-07-12 GITR/TGF-β DUAL-TARGETED FUSION PROTEIN AND USE THEREOF WO2023284733A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110796450.6 2021-07-14
CN202110796450.6A CN113429488B (en) 2021-07-14 2021-07-14 GITR/TGF-beta dual-targeting fusion protein and application thereof

Publications (1)

Publication Number Publication Date
WO2023284733A1 true WO2023284733A1 (en) 2023-01-19

Family

ID=77760371

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/105175 WO2023284733A1 (en) 2021-07-14 2022-07-12 GITR/TGF-β DUAL-TARGETED FUSION PROTEIN AND USE THEREOF

Country Status (2)

Country Link
CN (1) CN113429488B (en)
WO (1) WO2023284733A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113429488B (en) * 2021-07-14 2022-08-30 海正生物制药有限公司 GITR/TGF-beta dual-targeting fusion protein and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102203258A (en) * 2008-07-02 2011-09-28 新兴产品开发西雅图有限公司 TGF-b antagonist multi-target binding proteins
CN108026158A (en) * 2015-08-12 2018-05-11 免疫医疗有限公司 GITRL fusion proteins and application thereof
WO2020033715A1 (en) * 2018-08-08 2020-02-13 Cedars-Sinai Medical Center Compositions and methods for treating cancer and autoimmune diseases
WO2021093760A1 (en) * 2019-11-12 2021-05-20 江苏恒瑞医药股份有限公司 TGF-β RECEPTOR-CONTAINING FUSION PROTEIN AND PHARMACEUTICAL USE THEREOF
CN113429488A (en) * 2021-07-14 2021-09-24 海正生物制药有限公司 GITR/TGF-beta dual-targeting fusion protein and application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2432091T5 (en) * 2005-03-25 2022-03-18 Gitr Inc GITR binding molecules and uses thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102203258A (en) * 2008-07-02 2011-09-28 新兴产品开发西雅图有限公司 TGF-b antagonist multi-target binding proteins
CN108026158A (en) * 2015-08-12 2018-05-11 免疫医疗有限公司 GITRL fusion proteins and application thereof
WO2020033715A1 (en) * 2018-08-08 2020-02-13 Cedars-Sinai Medical Center Compositions and methods for treating cancer and autoimmune diseases
WO2021093760A1 (en) * 2019-11-12 2021-05-20 江苏恒瑞医药股份有限公司 TGF-β RECEPTOR-CONTAINING FUSION PROTEIN AND PHARMACEUTICAL USE THEREOF
CN113429488A (en) * 2021-07-14 2021-09-24 海正生物制药有限公司 GITR/TGF-beta dual-targeting fusion protein and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LI SHENG QING , LI HUAN ZHANG , YANG QIAO XIN , NI DIAN TAO: "Purification and characterization of the fusion protein TGF-βRⅡ/Fc", CHINESE JOURNAL OF CELLULAR AND MOLECULAR IMMUNOLOGY, vol. 19, no. 4, 30 August 2003 (2003-08-30), pages 400 - 402+405, XP093023988, ISSN: 1007-8738 *
YOO CHANGHOON, OH DO-YOUN, CHOI HYE JIN, KUDO MASATOSHI, UENO MAKOTO, KONDO SHUNSUKE, CHEN LI-TZONG, OSADA MOTONOBU, HELWIG CHRIST: "Phase I study of bintrafusp alfa, a bifunctional fusion protein targeting TGF-β and PD-L1, in patients with pretreated biliary tract cancer", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 8, no. 1, 1 January 2020 (2020-01-01), pages 1 - 9, XP093023991, DOI: 10.1136/jitc-2020-000564 *

Also Published As

Publication number Publication date
CN113429488B (en) 2022-08-30
CN113429488A (en) 2021-09-24

Similar Documents

Publication Publication Date Title
US11970537B2 (en) Fusion protein dimer using antibody Fc region as backbone and use thereof
US20210260163A1 (en) Novel cytokine prodrugs
CN110799528A (en) Multimeric IL-15-based molecules
US11673931B2 (en) Immunocytokines for the treatment of cancer
WO2013147153A1 (en) Anti-lamp5 antibody and utilization thereof
KR20170105077A (en) Tumor necrosis factor (TNF) superfamily receptor-binding molecules and uses thereof
KR20140138129A (en) Targeted mutant alpha-helical bundle cytokines
KR20160007604A (en) Bispecific constructs and their use in the treatment of various diseases
WO2021219127A1 (en) Bispecific antibody targeting her2 and pd-1 and application thereof
CN112739379A (en) Fc-binding fragment comprising OX40 antigen binding site
CN114040926A (en) Polypeptide binding to CD123 and uses thereof
US20230174603A1 (en) A protein complex comprising an immunocytokine
CN114040927A (en) Polypeptide binding to CD33 and application thereof
WO2023284733A1 (en) GITR/TGF-β DUAL-TARGETED FUSION PROTEIN AND USE THEREOF
JP2023520587A (en) Antibodies against NKp46 and constructs thereof for the treatment of cancer and infectious diseases
KR20240046224A (en) Bispecific antibodies and their uses
TW202104262A (en) Antibody Binding to PD-1
CN115776990A (en) Canine PD-1 binding polypeptides and uses thereof
TWI815184B (en) TGFBR2-ECD mutants and fusion proteins containing them and their applications
WO2023020459A1 (en) MONOCLONAL ANTIBODY TARGETING SIRPα AND USE THEREOF
KR20230042524A (en) Anti-HER2 / Anti-4-1BB Bispecific Antibodies and Uses Thereof
WO2024083226A1 (en) Antibody fusion protein and preparation and use thereof
CN111448214A (en) Miniaturized antibodies against glucocorticoid-induced tumor necrosis factor receptor (GITR), polymers thereof, and uses thereof
EP3613766B1 (en) Polypeptide improved in protein purity and affinity for antigen, conjugate thereof with antibody or antigen-binding fragment, and preparation method therefor
WO2022057821A1 (en) Pd-l1 antibodies, fusion proteins, and uses thereof

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: 22841368

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE