WO2020048454A1 - 融合蛋白及其在制备用于治疗肿瘤和/或病毒感染的药物中的应用 - Google Patents
融合蛋白及其在制备用于治疗肿瘤和/或病毒感染的药物中的应用 Download PDFInfo
- Publication number
- WO2020048454A1 WO2020048454A1 PCT/CN2019/104217 CN2019104217W WO2020048454A1 WO 2020048454 A1 WO2020048454 A1 WO 2020048454A1 CN 2019104217 W CN2019104217 W CN 2019104217W WO 2020048454 A1 WO2020048454 A1 WO 2020048454A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibody
- fusion protein
- umy02
- seq
- human
- Prior art date
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2878—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/555—Interferons [IFN]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/21—Interferons [IFN]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/555—Interferons [IFN]
- C07K14/56—IFN-alpha
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70578—NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/62—DNA sequences coding for fusion proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/33—Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/565—Complementarity determining region [CDR]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/30—Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/70—Fusion polypeptide containing domain for protein-protein interaction
- C07K2319/74—Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
Definitions
- the invention belongs to the field of biomedicine. Specifically, the present invention relates to a fusion protein and use thereof. More specifically, the present invention relates to a fusion protein containing an anti-OX40 antibody and human interferon and its use in the preparation of a medicament for treating tumors and / or viral infections.
- Human OX40 is mainly expressed on activated T cells, including CD4, CD8, Th and Treg cells, etc. [1]. in The expression of OX40 on T cells is very low, but its expression level is up-regulated and reaches its peak within 12 hours to 5-6 days after antigen-induced stimulation. Similarly, the expression of OX40L is also affected by the state of cell activation [1]. APC cells can detect the expression of OX40L 1-3 days after antigen stimulation. Interestingly, in addition to immune cells, muscle cells also express OX40L under the stimulation of inflammatory factors [2,3], suggesting that the OX40L-OX40 signaling pathway may widely affect the body's inflammatory response.
- OX40L / OX40 co-stimulatory molecules The activation of antigen-dependent OX40L / OX40 co-stimulatory molecules is coupled to multiple signaling pathways in T cells. Crystal structure studies have shown that the combination of OX40L and OX40 can induce the trimerization of OX40-OX40L complex [4], thereby forming a binding site with receptor-associated factor (TRAF) in the cell. The latter (TRAF2, 5) can further activate the NF- ⁇ B signaling pathway and inhibit T cell apoptosis [2,5,6].
- OX40 activation can lead to high expression of Bcl-2 and Bcl-xL [7], suggesting that OX40 may induce the expression of anti-apoptotic proteins through the NF- ⁇ B signaling pathway and achieve its function of inhibiting T cell apoptosis.
- PKB / PI3K is another important signaling pathway downstream of OX40.
- the co-stimulatory signal of OX40 on T cells is necessary for maintaining PKB activation
- constitutively activated PKB can antagonize the down-regulation of anti-apoptotic proteins in T cells caused by OX40 deficiency.
- the OX40 co-stimulatory signal can maintain Survivin protein expression through the PKB / PI3K signaling pathway [8].
- TCR and OX40 on T cells can also cause the activation of calcium flux and NFAT signaling pathway, and regulate the expression of cytokines including IL-2, IL-4, IL-5 and IFN- ⁇ [9] .
- the above studies indicate that the activation of OX40 can regulate the proliferation, apoptosis and cytokine secretion of T cells through the NF- ⁇ B signaling pathway, PKB / PI3K signaling pathway and NFAT signaling pathway, thereby achieving the effect of enhancing the vitality of the immune system.
- OX40 has become an important target for immunotherapy. Numerous pre-clinical and clinical studies have suggested that OX40 can be an important target for tumor immunotherapy. Interestingly, recent studies have also found a role for the OX40 signaling pathway in suppressing hepatitis B virus infection [10], suggesting that OX40 agonists may be a potential means of treating antiviral infections, such as patients with hepatitis B.
- Interferon is a class of highly active and versatile glycoproteins. on the one hand. Interferon can exert a strong anti-tumor effect by regulating tumor cell proliferation, inhibiting tumor metastasis and angiogenesis, and activating anti-tumor immune response; on the other hand, by regulating the human immune system, interferon has important clinical application value in anti-virus For example, interferon has become one of the important methods for clinical treatment of hepatitis B virus infection.
- OX40 agonist and interferon have important application value or potential in antitumor and antiviral aspects, but the existing evidence shows that both have insufficient patient response rates and efficacy. Therefore, there is currently a need for OX40 agonists and interferons that have better therapeutic effects.
- the object of the present invention is to provide a fusion protein comprising an antibody or an antigen-binding fragment thereof that specifically binds human OX40 and human interferon.
- the invention also provides the use of the fusion protein for treating tumors and / or viral infections.
- the present invention adopts the following technical solutions:
- the invention provides a fusion protein comprising:
- human interferon is connected to the carboxyl or amino terminus of the light or heavy chain of the antibody directly or through a peptide linker.
- the fusion protein according to the present invention wherein the antibody or antigen-binding fragment thereof that specifically binds human OX40 comprises:
- Antibody heavy chain variable region comprising VHCDR1 having the amino acid sequence of SEQ ID NO: 1, VHCDR2 having the amino acid sequence of SEQ ID NO: 2, and amino acid sequence having SEQ ID NO: 3 VHCDR3;
- Antibody light chain variable region comprising VL CDR1 having the amino acid sequence of SEQ ID NO: 4, VL2 having the amino acid sequence of SEQ ID NO: 5, CDR2, and the amino acid sequence having SEQ ID NO: 6 VL CDR3.
- the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO: 7
- the light chain variable region comprises the amino acid sequence shown in SEQ ID NO: 8.
- the fusion protein according to the present invention wherein the antibody or antigen-binding fragment thereof that specifically binds human OX40 is a camelized single domain antibody, scFv, scFv dimer, BsFv, dsFv, dsFv2, dsFv-dsFv ', Fv Fragment, Fab, Fab ', F (ab') 2, ds bifunctional antibody, Nanobody, domain antibody or bivalent domain antibody.
- the fusion protein according to the present invention wherein the antibody further comprises an immunoglobulin constant region, such as a constant region of human IgG1, IgG2, or IgG4.
- an immunoglobulin constant region such as a constant region of human IgG1, IgG2, or IgG4.
- the human interferon is selected from the group consisting of type I human interferon, type II human interferon, and type III human interferon.
- the human interferon is IFN ⁇ 2a, IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ 2b; more preferably, the human interferon is IFN ⁇ 2b, and its amino acid sequence is shown in SEQ ID NO: 9.
- the human interferon is a mutant of IFN ⁇ 2b, which has one or more mutations selected from the following amino acid sequences shown in SEQ ID NO: 9: T106A, R149A, A145G, A145D, R120A , L117A;
- the mutant of IFN ⁇ 2b has a double mutation selected from the following in the amino acid sequence shown in SEQ ID NO: 9:
- T106A / A145D T106A / R149A, T106A / A145G, T106A / L117A, T106A / R120A.
- the peptide linker is selected from (G) n, KESGSVSSEQLAQFRSLD, EGKSSGSGSESKST, GSAGSAAGSGEF, (GGGGS) n, (GGSGG) n; preferably, the peptide linker is (GGGGS) n, Wherein n is an integer between 0-5; preferably, n is an integer between 1-3.
- fusion protein according to the present invention, wherein the fusion protein is selected from:
- UMY02-L1 which contains the amino acid sequence shown in SEQ ID NO: 10 and the amino acid sequence shown in SEQ ID NO: 11, wherein the heavy chain, peptide linker and human of the antibody whose amino acid sequence shown in SEQ ID NO: 10 is OX40 Interferon, the light chain of an antibody whose amino acid sequence shown in SEQ ID NO: 11 is OX40;
- UMY02-L2 which contains the amino acid sequence shown in SEQ ID NO: 12 and the amino acid sequence shown in SEQ ID NO: 13,
- the heavy chain of the antibody whose amino acid sequence is shown in SEQ ID NO: 12 is OX40
- the light chain, peptide linker and human interferon of the antibody whose amino acid sequence is shown in SEQ ID NO: 13 is OX40;
- UMY02-L3 which contains the amino acid sequence shown in SEQ ID NO: 14 and the amino acid sequence shown in SEQ ID NO: 13, wherein the amino acid sequence shown in SEQ ID NO: 14 is the heavy chain of the antibody of OX40, SEQ ID The light chain, peptide linker and human interferon of the antibody whose amino acid sequence is NO: 13 is OX40;
- UMY02-L4 where the heavy chain is shown in SEQ ID NO: 14, the light chain and human interferon are shown in SEQ ID ID NO: 13, and the human interferon has no peptide linker;
- UMY02-L5 where the heavy chain is shown as SEQ ID NO: 14, the light chain and human interferon are shown as light chain and human interferon in SEQ ID NO: 13, and the peptide linker is GGGGS;
- UMY02-L6 where the heavy chain is shown as SEQ ID NO: 14, the light chain and human interferon are shown as light chain and human interferon in SEQ ID NO: 13, and the peptide linker is (GGGGS) 2;
- UMY02-L7 where the heavy chain is shown in SEQ ID NO: 15, the light chain and human interferon are shown in SEQ ID ID: 13, and the peptide linker is GGGGS;
- UMY02-L8 wherein the heavy chain is shown in SEQ ID NO: 15, the light chain and human interferon are shown in SEQ ID ID: 13 and the human interferon, and the peptide linker is (GGGGS) 2;
- UMY02-L13 where the heavy chain is shown as SEQ ID NO: 15, the light chain and human interferon are shown as light chain and human interferon in SEQ ID NO: 13, the peptide linker is (GGGGS) 2, and the interferon is in For example, it is mutated to T106A / A145D based on SEQ ID NO: 9;
- UMY02-L14 where the heavy chain is shown as SEQ ID NO: 15, the light chain and human interferon are shown as light chain and human interferon in SEQ ID NO: 13, the peptide linker is (GGGGS) 2, and the interferon is in For example, it is mutated to T106A / R149A based on SEQ ID NO: 9;
- UMY02-L15 where the heavy chain is shown as SEQ ID NO: 15, the light chain and human interferon are shown as the light chain and human interferon in SEQ ID NO: 13, the peptide linker is (GGGGS) 2, and the interferon is in For example, it is mutated into T106A / R120A based on SEQ ID NO: 9;
- UMY02-L16 where the heavy chain is shown as SEQ ID NO: 15, the light chain and human interferon are shown as the light chain and human interferon in SEQ ID NO: 13, the peptide linker is (GGGGS) 2, and the interferon is
- the basic mutation of SEQ ID NO: 9 is T106A / A145G;
- UMY02-L17 where the heavy chain is shown as SEQ ID NO: 15, the light chain and human interferon are shown as light chain and human interferon in SEQ ID NO: 13, the peptide linker is (GGGGS) 3, and the interferon is
- the basic mutation of SEQ ID NO: 9 is T106A / R149A;
- UMY02-L18 where the heavy chain is shown as SEQ ID NO: 15, the light chain and human interferon are shown as light chain and human interferon in SEQ ID NO: 13, the peptide linker is (GGGGS) 2, and the interferon is
- the basic mutation of SEQ ID NO: 9 is T106A / L117A;
- Fusion proteins OX40IFN- ⁇ 2a, OX40-IFN ⁇ , OX40-IFN ⁇ , OX40-IFN ⁇ 3, among which OX40IFN- ⁇ 2a, OX40-IFN ⁇ , OX40-IFN ⁇ , OX40-IFN ⁇ 3 heavy chain is shown as SEQ ID ID NO: 15;
- the peptide linker and interferon are shown in SEQ ID NO: 16;
- the OX40IFN- ⁇ light chain, the peptide linker and interferon are shown in SEQ ID NO: 17;
- the OX40IFN- ⁇ light chain, the peptide linker and interferon are shown in SEQ ID NO : 18;
- OX40IFN- ⁇ 3 light chain, peptide linker and interferon are shown in SEQ ID NO: 19.
- the present invention provides an isolated polynucleotide encoding the fusion protein.
- the invention provides a vector comprising the isolated polynucleotide.
- the present invention provides a host cell including the vector.
- the present invention also provides a method for expressing the fusion protein, which comprises culturing the host cell under conditions capable of expressing an isolated polynucleotide.
- the invention also provides a kit comprising the fusion protein.
- the invention also provides a pharmaceutical composition comprising the fusion protein and a pharmaceutically acceptable carrier.
- the invention also provides the use of the fusion protein in the preparation of a medicament for the treatment of conditions that can benefit from enhanced immune response and / or exposure to interferon.
- the condition is a cancer or a viral infection, such as a hepatitis B virus infection.
- the present invention provides a method of treating a condition that can benefit by enhancing an immune response and / or by being exposed to interferon, said method comprising administering to a subject in need thereof a therapeutically effective amount of said fusion protein; preferably Where the condition is a cancer or viral infection, such as a hepatitis B virus infection.
- the invention provides a fusion protein that treats a condition that can benefit by enhancing an immune response and / or that can benefit from exposure to interferon; preferably, wherein the condition is a cancer or a viral infection, such as a hepatitis B virus infection.
- the present invention provides a fusion protein of OX40 agonistic antibody and interferon, which simultaneously combines the different mechanisms of action of the two, and forms a synergistically enhanced effect: on the one hand, when the interferon portion of the fusion protein binds to a highly expressed interferon receptor,
- the surface of somatic cells may enhance the activity of OX40-activating antibodies in a receptor-mediated mode, thereby better enhancing the activity of the immune system;
- the fusion protein of the present invention is more disturbing The half-life of the hormone molecule is greatly prolonged, and it can be used at a lower frequency in the clinic. Compared with the current interferon which requires daily injection in the clinic, it has a great advantage.
- the fusion protein of the present invention may show unique advantages in terms of efficacy and compliance in antitumor and antiviral treatment.
- Figure 1 shows a schematic diagram of the structure of a fusion protein according to the present invention.
- Figure 2 shows the results of an ELISA binding experiment of a fusion protein according to the present invention with human OX40 protein.
- FIG. 3 FACS test results show that MT01-L1 antibody binds to both human and cynomolgus monkey OX40, but not to mouse OX40.
- Fig. 4 shows the proliferation inhibitory effect of the fusion protein according to the present invention on Daudi cells.
- Figure 5 shows that the fusion protein according to the present invention activates NF-kB signaling pathway activity in Jurkat cells.
- FIG. 6 shows that the fusion protein according to the present invention can promote the OX40 signaling pathway activating activity of MT01-C1 antibody.
- the OX40 monoclonal antibody (MT01-C1 or MT01-C1 (G2)) alone or the interferon molecule IFN ⁇ 2b has no or only weak activation of the OX40 signaling pathway in Jurkat cells in this experimental system (6a).
- the fusion proteins (UMY02-L1, UMY02-L2, and UMY02-L3) have significant activation activity on the OX40 signaling pathway under the same conditions (6b).
- Figure 7 shows the pharmacokinetic curves of UMY02-L1 and UMY02-L3 in mice (7a).
- Figure 7b shows the pharmacokinetic curves of MT01-C1 in mice.
- Fig. 8 shows the killing effect of the fusion protein according to the present invention on tumor cells.
- antibody in the present invention includes any immunoglobulin, monoclonal antibody, polyclonal antibody, multispecific antibody, or bispecific (bivalent) antibody that can bind to a specific antigen.
- a natural intact antibody contains two heavy chains and two light chains. Each heavy chain is composed of a variable region and first, second, and third constant regions; each light chain is composed of a variable region and a constant region. Mammalian heavy chains can be divided into ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , and mammalian light chains can be divided into ⁇ or ⁇ .
- the antibody is "Y" -shaped, and the neck of the Y-type structure consists of the second and third constant regions of the two heavy chains, which are bound by disulfide bonds.
- Each arm of the "Y" structure includes the variable region of one of the heavy chains and a first constant region that binds to the variable and constant regions of a light chain.
- the variable regions of the light and heavy chains determine antigen binding.
- the variable region of each chain contains three hypervariable regions, called complementarity determining regions (CDR).
- the CDRs of the light chain (L) include LCDR1, LCDR2, and LCDR3, and the CDRs of the heavy chain (H) include HCDR1, HCDR2, and HCDR3.
- the CDR boundaries of the antibodies and antigen-binding fragments disclosed in the present invention can be named or identified by Kabat, Chothia or Al-Lazikani nomenclature.
- the three CDRs are separated by a side continuous portion called a framework region (FR), which is more highly conserved than the CDRs and forms a scaffold to support the hypervariable loop.
- FR framework region
- the constant regions of the heavy and light chains have nothing to do with antigen binding, but have multiple effector functions.
- Antibodies can be divided into several classes based on the amino acid sequence of the constant region of the heavy chain. Depending on whether they contain ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ heavy chains, antibodies can be divided into five major classifications or isomers: IgA, IgD, IgE, IgG, and IgM.
- IgG1 ⁇ 1 heavy chain
- IgG2 ⁇ 2 heavy chain
- IgG3 ⁇ 3 heavy chain
- IgG4 ⁇ 4 heavy chain
- IgA1 ⁇ 1 heavy chain
- IgA2 ⁇ 2 heavy chain
- antigen-binding fragment refers to an antibody fragment formed from an antibody portion containing one or more CDRs or any other antibody fragment that binds an antigen but does not have a complete antibody structure.
- antigen-binding fragments include, but are not limited to, such as bifunctional antibodies (diabody), Fab, Fab ', F (ab') 2, Fv fragments, disulfide-stabilized Fv fragments (dsFv), (dsFv) 2, Bispecific dsFv (dsFv-dsFv '), disulfide-stabilized bifunctional antibody (dsdiabody), single chain antibody molecule (scFv), scFv dimer (bivalent bifunctional antibody), bivalent single chain antibody (BsFv), multispecific antibodies, camelized single domain antibodies, Nanobodies, domain antibodies, and bivalent domain antibodies.
- the antigen-binding fragment can bind the same antigen as the parent antibody.
- an antigen-binding fragment can bind the same anti
- the "Fab" fragment of an antibody refers to the portion of the antibody molecule that is composed of a light chain (including a variable region and a constant region) and a variable region and a portion of the constant region of a heavy chain that are bound by disulfide bonds.
- Fab ' refers to a Fab fragment that contains a partial hinge region.
- F (ab ') 2 refers to a dimer of Fab.
- the Fc segment of an antibody is responsible for many different effector functions such as ADCC and CDC, but does not participate in antigen binding.
- the "Fv” segment of an antibody refers to the smallest antibody fragment that contains a complete antigen-binding site.
- the Fv fragment consists of a variable region of a light chain and a variable region of a heavy chain.
- a “fusion protein” refers to a recombinant protein that connects a cDNA encoding a protein of interest with a cDNA encoding an antibody or an antibody fragment at the gene level and expresses it in a eukaryotic or prokaryotic expression system.
- Linker refers to a peptide chain or a derivative thereof consisting of 1 to 50 amino acids that have formed a peptide bond, and the N-terminus and C-terminus of the peptide chain or a derivative thereof are covalently formed with any one of an anti-OX40 antibody or an interferon Bond, thereby binding the anti-OX40 antibody to interferon.
- the anti-OX40 antibody and interferon can be integrated by binding the N-terminus or C-terminus of the interferon through a linker sequence or directly using a peptide bond on the C- or N-terminal side of the heavy or light chain of the anti-OX40 antibody, respectively. .
- fusion protein of interferon and anti-OX40 antibody a fusion protein obtained by binding the C-terminus of the heavy or light chain of the anti-OX40 antibody to the N-terminus of the interferon via a linker sequence; or, A fusion protein in which the N-terminus of the heavy or light chain of an anti-OX40 antibody is bound to the C-terminus of an interferon via a linker sequence.
- Single-chain Fv antibody or “scFv” refers to an engineered antibody in which the light chain variable region and the heavy chain variable region are directly connected or connected through a peptide chain (Huston, JS, etc., Proc Natl Acad Sci USA, 85: 5879 (1988)).
- Single-chain antibody Fv-Fc or “scFv-Fc” refers to an engineered antibody composed of scFv linked to the Fc segment of an antibody.
- HCAb Heavy-chain-only antibodies, HCAb
- heavy chain antibodies were originally derived from the camel family (camel, dromedary, and llama).
- camelized antibodies have the full function of confirmed antigen binding (Hamers, Casterman, C. et al., Nature 363 (6428): 446-8 (1993); Nguyen, VK., Et al., "Heavy-chain antibodies” in Camelidae: a case of evolutionary innovation, Immunogenetics. 54 (1): 39-47 (2002); Nguyen VK. Et al., Immunology. 109 (1): 93-101 (2003)).
- Variable regions of heavy chain antibodies ( VH domain) is the smallest known antigen-binding unit for adaptive immunity (Koch-Nolte F. et al., FASEB J. 21 (13): 3490-8. Epub (2007)).
- Nanobody refers to an antibody fragment that consists of a VH domain from a heavy chain antibody and two constant regions, CH2 and CH3.
- a “diabody” includes a small antibody fragment with two antigen-binding sites, where the fragment contains a VH domain and a VL domain connected on the same polypeptide chain (see Holliger P. et al., Proc Natl Acad. Sci. A. 90 (14): 6444-8 (1993); EP404097; W093 / 11161).
- the adapter between the two domains is very short, so that the two domains on the same chain cannot pair with each other, thus forcing the two domains to pair with the complementary domains of the other two chains to form two antibody binding sites.
- These two antibody binding sites can be targeted to bind the same or different antigens (or epitopes).
- a “domain antibody” refers to an antibody fragment that contains only one heavy chain variable region or one light chain variable region.
- two or more VH domains are covalently bound by a polypeptide adapter and form a bivalent domain antibody.
- the two VH domains of a bivalent domain antibody can target the same or different antigens.
- "(dsFv) 2" contains three peptide chains: two VH genes are connected by a polypeptide adapter and are bound to two VL groups via a disulfide bond.
- a "bispecific ds bifunctional antibody” contains VL1-VH2 (connected by two polypeptide adapters) and VH1-VL2 (also connected by two polypeptide adapters), both of which are between VH1 and VL1 Binding via disulfide bonds.
- a "scFv dimer” is a bivalent bifunctional antibody or a bivalent single chain antibody (BsFv) that contains two VH-VL (linked by a polypeptide adapter) group that is dimerized, wherein The VH of the two groups cooperates with the VL of the other group to form two binding sites, which can be targeted to bind the same antigen (or epitope) or different antigens (or epitopes).
- the "scFv dimer” is a bispecific bifunctional antibody that contains V L1- V H2 (connected by a peptide adapter) and V H1- V L2 (connected by a peptide adapter) interconnected Among them, V H1 and V L1 cooperate, V H2 and V L2 cooperate, and each cooperative pair has different antigen specificity.
- whole-human origin used in this application when used in an antibody or an antigen-binding fragment means that the antibody or antigen-binding fragment has a certain amino acid sequence or consists of the amino acid sequence, and the amino acid sequence corresponds to a human Or an amino acid sequence of an antibody produced by a human immune cell or derived from a non-human source such as a transgenic non-human animal using a human antibody library, or another sequence encoding a human-derived antibody.
- fully human antibodies do not contain amino acid residues (especially antigen-binding residues) derived from non-human antibodies.
- humanized when used in antibodies or antigen-binding fragments, is meant to include CDRs derived from non-human animals, FR regions derived from humans, and constant regions derived from humans (when applicable) ) Antibodies or antigen-binding fragments. Because humanized antibodies or antigen-binding fragments have reduced immunogenicity, they can be used as human therapeutics in certain embodiments.
- the non-human animal is a mammal such as a mouse, rat, rabbit, goat, sheep, guinea pig or hamster.
- the humanized antibody or antigen-binding fragment consists essentially of human-derived sequences, except that the CDR sequences are of non-human origin.
- the human-derived FR region may include the same amino acid sequence as the human-derived antibody from which it is derived, or it may include some amino acid changes, for example, no more than 10, 9, 8, 7, 6, 5, , 4, 3, 2 or 1 amino acid change.
- the amino acid change may be present only in the heavy chain FR region, only in the light chain FR region, or in both chains.
- the humanized antibodies include human FRl-3 and human JH and JK.
- chimeric refers to having a portion of the heavy and / or light chain derived from one species, and the rest of the heavy and / or light chain is derived from antibodies or antigens from different species. Fragment.
- a chimeric antibody may include a constant region derived from a human and a variable region derived from a non-human animal, such as a mouse.
- OX40 refers to a receptor that binds to OX40L. It is a type I membrane protein belonging to the TNF receptor family. Other names are ACT-4, OX40L receptor, CD134 antigen, ACT35 antigen, TNFRSF4. It has a molecular weight of 50 kDa and is stored in SwissProt under the accession number P43489.
- human interferon used in this application refers to a class of highly active, multifunctional secreted glycoproteins, which have antiviral, immunomodulatory and antitumor effects. According to its gene sequence and receptor specificity, it can be divided into type I, type II and type III IFNs.
- Human type I interferons include 13 subtypes of IFN ⁇ and IFN ⁇ , IFN ⁇ , IFN ⁇ , and IFN ⁇ .
- Type I IFNs have a common cell surface receptor, IFNAR, consisting of two subunits, IFNAR1 and IFNAR2.
- the cell surface receptor of IFN ⁇ is IFNGR, which consists of two subunits, IFNGR1 and IFNGR2.
- Type III IFNs include IFN ⁇ 1, IFN ⁇ 2, IFN ⁇ 3, and IFN ⁇ 4. Interferons in the fusion proteins of the present application also include interferon variants known in the art.
- telomere binding refers to a non-random binding reaction between two molecules, such as a reaction between an antibody and an antigen.
- the antibodies or antigen-binding fragments thereof of the present application specifically bind to human and / or monkey OX40, and their binding affinity (K D ) is ⁇ 10 ⁇ 6 M.
- the K D in the present application refers to the ratio (k off / k on ) of the dissociation speed to the binding speed, which can be measured by a surface plasmon resonance method, for example, using an instrument such as Biacore.
- UY02-L1 refers to a heavy chain, a peptide linker, and a human interferon as shown in SEQ ID NO: 10, and a light chain as shown in SEQ ID NO: 11, in which the heavy chain carboxyl terminus A fusion protein of human interferon IFN ⁇ 2b as shown in SEQ ID NO: 9 was linked via a peptide linker.
- UY02-L2 used in this application refers to a heavy chain as shown in SEQ ID NO: 12, a light chain as shown in SEQ ID NO: 13, a peptide linker, and human interferon, in which the light chain carboxyl terminus A fusion protein of human interferon IFN ⁇ 2b as shown in SEQ ID NO: 9 was linked via a peptide linker.
- UY02-L3 refers to a heavy chain as shown in SEQ ID NO: 14, a light chain as shown in SEQ ID NO: 13, a peptide linker, and human interferon, in which the light chain carboxyl terminus A fusion protein of human interferon IFN ⁇ 2b as shown in SEQ ID NO: 9 was linked via a peptide linker.
- MMT01-C1 used in this application means that it has the same VH (SEQ ID NO: 7) and VL (SEQ ID NO: 8) sequences as UMY02-L1, UMY02-L2, and UMY02-L3, and the heavy and light chains are constant. Regions are monoclonal antibodies against human IgG1 and kappa chains, respectively.
- MT01-C1 (G2) used in this application means that it has the same VH (SEQ ID NO: 7) and VL (SEQ ID ID NO: 8) sequences as UMY02-L1, UMY02-L2, and UMY02-L3, and repeats
- VH SEQ ID NO: 7
- VL SEQ ID NO: 8
- the light chain constant regions are monoclonal antibodies to human IgG2 and kappa chains, respectively.
- MMT01-L1 used in this application means that it has the same heavy and light chain CDR sequences (SEQ ID NO: 1-6) as UMY02-L1, UMY02-L2, and UMY02-L3, and the heavy and light chain constant regions are human Human-mouse chimeric antibodies to IgG1 and kappa chains.
- the "MT01-C1", “MT01-C1 (G2)”, and “MT01-L1” are all derived from the Chinese patent 201711476160.3, and the specific sequences are shown in Table 1 and the sequence table, which are incorporated by reference in their entirety. This article.
- conservative substitution when “conservative substitution” is used in an amino acid sequence, it means that one amino acid residue is replaced with another amino acid residue in a side chain having similar physicochemical properties. For example, between hydrophobic side chain amino acid residues (such as Met, Ala, VaL, Leu, and Ile), neutral hydrophilic side chain residues (such as Cys, Ser, Thr, Asn, and Gln), acidic side chain residues Conservative substitutions are made between bases (e.g. Asp, Glu), basic side chain amino acids (e.g. His, Lys and Arg) or aromatic side chain residues (e.g. Trp, Tyr and Phe). It is known in the art that conservative substitutions generally do not cause significant changes in the conformational structure of the protein and are therefore capable of retaining the biological activity of the protein.
- bases e.g. Asp, Glu
- basic side chain amino acids e.g. His, Lys and Arg
- aromatic side chain residues e.g. Trp, Tyr and Phe
- percent sequence identity when “percent sequence identity” is used for an amino acid sequence (or nucleic acid sequence), it means that after the sequence alignment is performed, and if necessary, a gap is introduced to maximize the number of identical amino acids (or nucleic acids), in the candidate sequence, the The percentage of amino acid (or nucleic acid) residues with the same sequence as the candidate sequence. Conservative substitutions of the amino acid residues may or may not be considered the same residues. Sequences can be aligned by tools disclosed in the art to determine the percent sequence identity of amino acid (or nucleic acid) sequences. Those skilled in the art can use the default parameters of the tool or adjust the parameters appropriately according to the needs of the comparison, for example, by selecting an appropriate algorithm.
- T cells used in the present application include CD4 + T cells, CD8 + T cells, T helper type 1 T cells, T helper type 2 T cells, T helper type 17 T cells, and suppressor T cells.
- Effective function refers to the biological activity of an antibody's Fc region that binds to its effectors, such as the C1 complex and Fc receptor.
- exemplary effector functions include complement-dependent cytotoxicity (CDC) induced by the interaction of antibodies with C1q on the C1 complex, antibody-dependent cell-mediated cells induced by binding of the Fc region of the antibody to Fc receptors on effector cells Toxicity (ADCC) and phagocytosis.
- cancer or “cancer condition” refers to any medical condition that is mediated by the growth, proliferation, or metastasis of tumors or malignant cells and causes solid tumors and non-solid tumors such as leukemia.
- tumor in the present invention refers to a solid substance of a tumor and / or a malignant cell.
- viral infection refers to a pathogenic process in which a virus invades a human body through various pathways and proliferates in human cells, causing damage to the body, including chronic viral infections, such as hepatitis B, hepatitis C, and herpes viruses , EB (Epstein-Barr) virus, HIV, cytomegalovirus, herpes simplex virus type I, herpes simplex virus type 2, human papilloma virus, adenovirus virus infection, Kaposi's sarcoma-associated herpes virus epidemic , Torquetenovirus, JC virus or BK virus.
- chronic viral infections such as hepatitis B, hepatitis C, and herpes viruses , EB (Epstein-Barr) virus, HIV, cytomegalovirus, herpes simplex virus type I, herpes simplex virus type 2, human papilloma virus, adenovirus virus infection, Kaposi's sarcoma-associated herpes virus epidemic
- Treatment or “therapy” of a condition includes preventing or reducing a condition, reducing the rate at which a condition arises or developing, reducing the risk of developing a condition, preventing or delaying the development of symptoms associated with a condition , Reduce or terminate the symptoms associated with a condition, produce a complete or partial reversal of a condition, cure a condition, or a combination of the above.
- treatment or “therapy” can refer to inhibiting or slowing the growth, reproduction, or metastasis of tumors or malignant cells, or some combination of the above.
- treatment or “therapy” includes clearing all or part of a tumor, inhibiting or slowing tumor growth and metastasis, preventing or delaying tumor development, or some combination of the above.
- isolated matter has been artificially altered from its natural state. If some "isolated” substance or component appears in nature, it has been altered or removed from its original state, or both.
- a naturally occurring polynucleotide or polypeptide in a living animal is not isolated, but if these polynucleotides or polypeptides are sufficiently separated from substances that coexist in their natural state and exist in a sufficiently pure state, they can We think that it is "separated”.
- the purity of the antibody and antigen-binding fragment is at least 90%, 93%, 95%, 96%, 97%, 98%, 99%, which is determined by electrophoresis methods (such as SDS-PAGE, isoelectric focusing) , Capillary electrophoresis), or chromatography (such as ion exchange chromatography or reversed-phase HPLC).
- electrophoresis methods such as SDS-PAGE, isoelectric focusing
- Capillary electrophoresis Capillary electrophoresis
- chromatography such as ion exchange chromatography or reversed-phase HPLC.
- a "vector” refers to a vehicle that can be operatively inserted into a polynucleotide encoding a protein and obtain expression of the protein.
- Vectors can be used to transform, transduce, or transfect host cells so that the genetic material elements they carry are expressed within the host cells.
- vectors include: plasmids, phagemids, cosmids, artificial chromosomes such as yeast artificial chromosomes (YAC), bacterial artificial chromosomes (BAC) or P1-derived artificial chromosomes (PAC), phages such as lambda phage or M13 phage, and animal viruses.
- vectors The types of animal viruses used as vectors are retroviruses (including lentivirus, adenovirus, adeno-associated virus, herpes virus (such as herpes simplex virus), pox virus, baculovirus, papilloma virus, papilloma vacuolar virus (such as SV40)).
- Vectors can contain a variety of elements that control expression, including promoter sequences, transcription initiation sequences, enhancer sequences, selection elements, and reporter genes.
- the vector may contain a replication initiation site.
- a vector may also include components that assist it in entering cells, including, but not limited to, viral particles, liposomes, or protein shells.
- a "host cell” in the present invention is a cell that is directed into an exogenous polynucleotide and / or vector.
- the "therapeutically effective amount” or “effective dose” in the present invention refers to the dose or concentration of a certain drug to effectively treat a disease.
- the therapeutically effective amount is that at the dose or concentration, the antibody or antigen-conjugate can clear all or part of the tumor, inhibit or slow tumor growth, and inhibit Tumor cell metastases, alleviate any symptoms or markers associated with a tumor or cancer condition, prevent or delay the development of a tumor or cancer condition, inhibit or clear a virus or virus-infected cells, or some combination of the above.
- “Pharmaceutically acceptable” means the carrier, vehicle, diluent, excipient, and / or salt referred to, and is generally chemically and / or physically compatible with the other ingredients in the formulation, and Physiologically compatible with the recipient.
- the present application provides exemplary fusion proteins UMY02-L1, UMY02-L2, and / or UMY02-L3.
- CDR sequences can be modified to include substitutions of one or more amino acids, thereby resulting in improved biological activity such as improved binding affinity to human OX40.
- phage display technology can be used to produce and express a library of antibody variants (eg, Fab or FcFv variants), and then screen for antibodies that have an affinity for human OX40.
- computer software can be used to simulate the binding of the antibody to human OX40 and identify the amino acid residues on the antibody that form the binding interface. Substitutions of these residues can be avoided to prevent reduced binding affinity, or these residues can be targeted for substitution to form stronger binding.
- at least one (or all) substitutions in a CDR sequence are conservative substitutions.
- the fusion protein and antigen-binding fragment include one or more CDR sequences, which have the same fusion proteins as the exemplary fusion proteins UMY02-L1, UMY02-L2, and / or UMY02-L3 provided in the present application.
- Sequence identity of at least 80% e.g., at least 85%, 88%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%
- the parent antibody has substantially the same sequence, but its corresponding CDR sequence is 100% sequence identical to the listed sequence Sex.
- the fusion protein described herein is capable of specifically binding to human OX40 with a binding affinity (K D ) of ⁇ 10 ⁇ 7 M, which is measured by surface plasmon resonance method.
- the binding affinity value can be expressed as a K D value, which is calculated by the ratio of the dissociation rate to the binding rate (k off / k on ) when the binding of the antigen and the antigen-binding molecule reaches equilibrium.
- the antigen binding affinity e.g., K D
- K D may be suitably determined by suitable methods known in the art, for example, including the use of instruments such as the Biacore plasmon resonance binding assay.
- the fusion protein described herein binds to human OX40 at an EC50 (ie, half binding concentration) of 10 ng / mL to 10 ⁇ g / mL.
- the binding of the antibody or fusion protein to human OX40 can be determined by methods known in the art such as sandwich methods such as ELISA, Western blot, FACS or other binding assays.
- the test antibody ie, the primary antibody
- the unbound antibody is washed away, and a labeled secondary antibody is introduced, which can bind to the primary antibody.
- Able to detect bound secondary antibodies The detection can be performed on a microplate reader when using immobilized OX40, or can be performed using FACS analysis when using cells expressing human OX40.
- the fusion protein described herein binds human OX40 at an EC50 (ie, an effective concentration of 50%) of 0.1 ⁇ g / mL to 10 ⁇ g / mL (determined using FACS analysis).
- the fusion proteins described herein can activate the human OX40 signaling pathway in an FcR-mediated or interferon receptor-mediated manner, and thus provide methods including, for example, inducing activated T cells to produce cytokines (such as CD4 + T cells and CD8 + T cells), induces the proliferation of activated T cells (such as CD4 + T cells and CD8 + T cells), and reverses the biological activity of inhibitory functions of regulatory Tregs.
- cytokines such as CD4 + T cells and CD8 + T cells
- activated T cells such as CD4 + T cells and CD8 + T cells
- the fusion protein is specific for human OX40.
- the fusion protein does not bind to murine OX40, but binds to monkey OX40 with a binding affinity similar to human OX40.
- the binding of a monoclonal antibody MT01-L1 having the same CDR sequence as the fusion protein of the present invention to murine OX40 cannot be detected by conventional binding assay methods such as FACS analysis, and FACS detects that both MT01-L1 and monkey OX40 and human OX40 There are combinations.
- the fusion protein has a constant region of the IgG2 isotype, which has a reduced or eliminated effector function.
- Effector functions such as ADCC and CDC can cause cytotoxicity to cells expressing OX40.
- Some normal cells are capable of expressing OX40.
- certain embodiments of the antibodies of the invention have reduced or even eliminated effector functions.
- Many tests are known to estimate ADCC or CDC activity, such as Fc receptor binding assay, complement Clq binding assay, and cell lysis method, which can be easily selected by those skilled in the art. Not wishing to be bound by theory, but it is believed that antibodies with reduced or eliminated effector functions such as ADCC and CDC will not cause or minimize cytotoxicity to cells expressing OX40, such as those normal cells, Therefore, unwanted side effects are avoided.
- the fusion protein described in the present application has a prolonged action time in the organism than the interferon molecule. This is due to the longer half-life and drug retention time of the fusion protein in the animal. This property is beneficial to reduce the number of medications for patients and improve the efficacy of the drugs.
- the fusion proteins described herein have reduced side effects.
- the anti-OX40 antibody and its antigen-binding fragment may have a fully human IgG sequence, and therefore its immunogenicity is lower than that of a humanized antibody.
- the fusion protein and its antigen-binding fragment may have the form of IgG2 or IgG4 to eliminate ADCC and CDC.
- the fusion protein described herein has the advantage that it can be used in combination with substances having immunogenicity, such as tumor cells, purified tumor antigens, and cells transfected with encoded immunostimulatory factors, tumor vaccines.
- the anti-OX40 antibody and its antigen-binding fragment may be included in a combination therapy, including standard chemotherapy and radiation therapy, target-based small molecule therapy, and other emerging immune checkpoint modulator therapies.
- the antibody and its antigen-binding fragment can be used as a base molecule for an antibody-drug conjugate, a bispecific, or a multivalent antibody.
- the fusion protein and the antigen-binding fragment thereof described herein are a camelized single domain antibody, a bifunctional antibody (diabody), scFv, scFv dimer, BsFv, dsFv, (dsFv) 2, dsFv-dsFv ', Fv fragment, Fab, Fab', F (ab ') 2, ds diabody, Nanobody, domain antibody or bivalent domain antibody.
- the fusion proteins described herein include an immunoglobulin constant region.
- the immunoglobulin constant regions include heavy and / or light chain constant regions.
- the heavy chain constant region includes a CH1, CH1-CH2, or CH1-CH3 region.
- the immunoglobulin constant region may further include one or more modifications to obtain the desired properties.
- the constant region can be modified to reduce or eliminate one or more effector functions to enhance FcRn receptor binding or introduce one or more cysteine residues.
- the antibodies and antigen-binding fragments thereof further comprise a conjugate.
- the antibodies or antigen-binding fragments thereof of the present invention may be linked to various conjugates (see, for example, "Conjugate Vaccines", Contributions, Microbiology, Immunology, JMCruse, and RELewis, Jr. (eds.), Carger Press New York (1989)).
- conjugates can be linked to the antibody or antigen conjugate by other means such as covalent binding, affinity binding, intercalation, coordinate binding, complexation, binding, mixing, or addition.
- the antibodies and antigen-binding fragments disclosed herein can be engineered to contain specific sites other than the epitope-binding portion, and these sites can be used to bind one or more conjugates.
- a site may contain one or more reactive amino acid residues, such as cysteine and histidine residues, to assist in covalent attachment to the conjugate.
- the antibody may be attached to the conjugate indirectly, or through another conjugate.
- the antibody or antigen-binding fragment thereof can bind biotin and then indirectly bind a second conjugate, which is linked to avidin.
- the conjugate may be a detectable label, a pharmacokinetic modification moiety, a purification moiety, or a cytotoxic moiety.
- detectable labels may include fluorescent labels (e.g. fluorescein, rhodamine, dansyl, phycoerythrin or Texas red), enzyme substrate labels (e.g. horseradish peroxidase, alkaline phosphatase , Luciferase, glucoamylase, lysozyme, sugar oxidase or ⁇ -D galactosidase), stable isotope or radioisotope, chromophore moiety, digoxin, biotin / avidin, DNA molecule Or gold for testing.
- the conjugate may be a pharmacokinetic modification moiety such as PEG, which helps extend the half-life of the antibody.
- conjugate may be a purified portion such as a magnetic bead.
- a "cytotoxic moiety" can be any agent that is harmful to the cell or that may damage or kill the cell.
- cytotoxic fractions include, but are not limited to, paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, turpentine, mitomycin, etopoxil, tenipogan, vincristine, Vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithromycin, actinomycin D, l-dehydrotestosterone, glucocorticoids, Procaine, tetracaine, lidocaine, prochalorol, puromycin and its analogs, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-mercaptopurine, Acacia Cytidine, 5fluorouracil dacarbaba), alkylating agents (e.g.
- cyclophosphamide Busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (DDP) cisplatin), anthracycline antibiotics (e.g. daunorubicin (formerly Daunorubicin ) And doxorubicin), antibiotics (such as dactinomycin (formerly known as actinomycin), bleomycin, mithromycin, and ampicillin (AMC)), and antimitotic agents (such as vincristine Alkali and Changchun ).
- DDP cis-dichlorodiamine platinum
- the amino acid sequence in the fusion protein of the present application can be converted into the corresponding DNA coding sequence. Due to the degeneracy of the genetic code, the transformed DNA sequences can be completely identical, while the encoded protein sequence remains unchanged.
- a vector including a polynucleotide encoding the fusion protein can be introduced into a host cell for cloning (amplifying DNA) or gene expression.
- the fusion protein can be prepared by a method known in the art for homologous recombination.
- vectors are available.
- Vector components typically include, but are not limited to, two or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancement sequence, a promoter (for example: SV40, CMV, EF-1a), and Transcription termination sequence.
- the vector system includes mammals, bacteria, yeast systems, etc., and will include plasmids such as, but not limited to, pALTER, pBAD, pcDNA, pCal, pL, pELpGEMEX, pGEX, pCLpCMV, pEGFP, pEGFT, pSV2, pFUSE, pVITRO, pVIVO, pMAL, pMONO, pSELECT, pUNO, pDUO, Psg5L, pBABE, pWPXL, pBI, p15TV-L, pPro18, pTD, pRS420, pLexA, pACT2 and other vectors are available from laboratories or are commercially available.
- Suitable vectors may include plasmid or viral vectors (eg, replication defective retroviruses, adenoviruses and adeno-associated viruses).
- a vector including a polynucleotide encoding the fusion protein can be introduced into a host cell for cloning or gene expression.
- the host cell suitable for cloning or expressing the DNA in the vector in the present invention is a prokaryotic cell, a yeast, or the above-mentioned higher eukaryotic cell.
- Prokaryotic cells suitable for use in the present invention include Eubacteria such as Gram-negative or Gram-positive bacteria, for example, Enterobacteriaceae, such as E.
- eukaryotic microorganisms such as filamentous fungi or yeast can also be used as host cells to clone or express vectors encoding fusion proteins.
- Saccharomyces cerevisiae, or baker's yeast, are the most commonly used lower eukaryotic host microorganisms.
- Kluyveromyces hosts such as Kluyveromyces lactis, Kluyveromyces fragilis (ATCC 12, 424), Kluyveromyces bulgaricus (ATCC 16,045), Kluyveromyces welchii (ATCC 24,178), Kluyveromyces kluyveri (ATCC 56,500), Kluyveromyces drosophila (ATCC 36,906) Kluyveromyces thermolyticus and Kluyveromyces marxianus: Yarrowia lipolytica (EP 402,226); Pichia pastoris (EP 183,070); Candida: Trichoderma reesei (EP 244,234); Alternaria Mildew; Western Schwann yeast, such as: Western Schwann yeast; and filamentous fungi, such as: Neurospora, Penicillium, Curvularia and Asper
- the host cells provided in the present invention and suitable for expressing glycosylated antibodies or antigen-binding fragments thereof are derived from multicellular organisms.
- invertebrate cells include plant and insect cells.
- a variety of baculoviral strains and their variants, as well as corresponding permissive insect host cells, have been discovered from hosts such as: Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito) ), Aedes albopictus (mosquitoes), Drosophila melanogaster (drosophila) and silkworm.
- a number of virus strains for transfection are publicly available, such as the Bm-5 variants of Spodoptera litura nuclear polyhedrosis virus and Bombyx mori nuclear polyhedrosis virus, all of which can be used in the present invention, especially For transfection of Spodoptera frugiperda cells.
- Plant cell cultures of cotton, corn, potatoes, soybeans, petunias, tomatoes and tobacco can also be used as hosts.
- spinal cells are of most interest, and the culture (tissue culture) of spinal cells has become a routine operation.
- Examples of usable mammalian host cells are SV40 transformed monkey kidney cell CV1 line (COS-7, ATCC CRL 1651); human embryonic kidney cell line (293 or subculture 293 cell subclones, Graham et al.,]. Gen.Virol. 36:59 (1977)); young hamster kidney cells (B blood, ATCC CCL 10); Chinese hamster ovary cells / -DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77 : 4216 (1980)); mouse testis support cells (TM4, Mather, Biol.Reprod.
- monkey kidney cells (CV1ATCC CCL70); African green monkey kidney cells (VERO-76 ATCC CRL-1587); human cervical cancer cells (HELA, ATCC CCL2); canine kidney cells (MDCK, ATCC CCL34); Buffalo rat liver cells (BRL3A, ATCC CRL1442); human lung cells ( W138, ATCC (CCL75); human hepatocytes (Hep G2, HB 8065); mouse breast tumors (MMT 060562, ATCC CCL51); TRI cells (Matherther et al. Annals NYAcad. Sci. 383: 44-68 (1982) ); MRC 5 cells; FS4 cells; and human liver cancer cell line (HepG2).
- the host cell is a 293F cell.
- the host cell is transformed with the expression or cloning vector that can produce the fusion protein, and cultured in a conventional nutrient medium, which is modified to be suitable for inducing a promoter, selecting transformed cells, or expanding.
- a gene encoding a sequence of interest is modified to be suitable for inducing a promoter, selecting transformed cells, or expanding.
- the host cells used to produce the fusion protein in the present invention can be cultured in a variety of media known in the art.
- the medium may also contain any other necessary additives at appropriate concentrations known in the art.
- the conditions of the medium such as temperature, pH, and the like, are the conditions previously used to select host cells for expression, and are well known to those of ordinary skill.
- the antibodies can be produced intracellularly, in the parietal space, or secreted directly into the culture medium. If the antibody is produced intracellularly, the host cell or particle debris of the lysed fragment is first removed, for example, by centrifugation or ultrasound. Carter et al., Bio / Technology 10: 163-167 (1992) describes a method for isolating antibodies secreted into the membrane space of E. coli. Briefly, the cell paste is opened in the presence of uranium acetate (pH 3.5), EDTA, and benzylsulfonium fluoride (PMSF) for about 30 minutes or more. Centrifuge to remove cell debris.
- uranium acetate pH 3.5
- EDTA EDTA
- PMSF benzylsulfonium fluoride
- a commercially available protein concentration filter such as Amicon or Millipore Pellicon ultrafiltration unit is usually first used to concentrate the supernatant of the expression system.
- Protease inhibitors such as PMSF can be added in any of the foregoing steps to inhibit protein degradation and antibiotics to prevent the growth of accidental contaminants.
- Antibodies produced from the cells can be purified using purification methods such as hydroxyapatite chromatography, gel electrophoresis, dialysis, DEAE-cellulose ion exchange chromatography columns, ammonium sulfate precipitation, salting out, and affinity chromatography, where Affinity chromatography is the preferred purification technique.
- the type of antibody and the presence of any immunoglobulin Fc domain in the antibody determine whether protein A is suitable as an affinity ligand.
- Protein A can be used to purify antibodies based on human ⁇ 1, ⁇ 2 or ⁇ 4 heavy chains (Lindmark et al., J. lmmunol. Meth. 62: Bu 13 (1983)).
- Protein G is applicable to all murine isomers and human gamma 3 (Guss et al., EMBO J. 5: 1567 1575 (1986)).
- Agarose is the most commonly used affinity ligand attachment matrix, but other matrices are also available.
- Mechanically stable matrices such as controlled-porosity glass or poly (styrene) benzene can achieve faster flow rates and shorter processing times than with agarose. If the antibody contains a CH3 domain, it can be purified with Bakerbond ABX.TM resin (J.T. Baker, Phillipsburg, N.J.).
- proteins purification can also be determined based on the antibodies obtained, such as fractionation in ion exchange columns, ethanol precipitation, reversed-phase HPLC, silica gel chromatography, anion or cation exchange resin-based heparin agarose gel chromatography (such as polyaspart Lysine column), chromatography focus, SDS-PAGE, and ammonium sulfate precipitation.
- low pH hydrophobic interaction chromatography can be used to treat a mixture containing antibodies and impurities of interest, using an elution buffer at a pH of about 2.5-4.5, preferably at a low salt concentration (e.g., (From about 0 to 0.25M salt concentration).
- the application provides a kit comprising the fusion protein.
- the kit is used to detect the presence or level of OX40 in a biological sample.
- the biological sample may include cells or tissues.
- the kit includes a fusion protein conjugated to a detectable label.
- the kit includes an unlabeled fusion protein, and further includes a secondary antibody capable of binding to the unlabeled fusion protein.
- the kit may further include instructions for use and a package that separates each component in the kit.
- the fusion protein is linked to a substrate or instrument for a sandwich assay such as an ELISA or immunochromatographic assay.
- a substrate or instrument for a sandwich assay such as an ELISA or immunochromatographic assay.
- Suitable substrates or instruments can be, for example, microplates and test strips.
- the application further provides a pharmaceutical composition comprising the fusion protein and one or more pharmaceutically acceptable carriers.
- the pharmaceutically acceptable carriers used in the pharmaceutical compositions disclosed herein may include, for example, pharmaceutically acceptable liquid, gel or solid carriers, aqueous media, non-aqueous media, antimicrobial substances, etc. Osmotic substances, buffers, antioxidants, anesthetics, suspending agents / dispersing agents, integrating agents, diluents, adjuvants, excipients or non-toxic auxiliary substances, other components known in the art or a combination of the above.
- Suitable components may include, for example, antioxidants, fillers, binders, disintegrants, buffers, preservatives, lubricants, flavoring agents, thickeners, colorants, emulsifiers or stabilizers such as Sugar and cyclodextrin.
- Suitable antioxidants may include, for example, methionine, ascorbic acid, EDTA, sodium thiosulfate, platinum, catalase, citric acid, cysteine, mercaptoglycerol, mercaptoacetic acid, mercaptosorbitol, butylmethyl Anisole, butylated hydroxytoluene and / or propyl gallate.
- Including one or more antioxidants such as methionine in a composition containing the fusion protein disclosed in the present invention can reduce the oxidation of the fusion protein.
- a reduction in oxidation prevents or reduces a decrease in binding affinity, thereby increasing antibody stability and extending shelf life.
- a pharmaceutically acceptable carrier may include, for example, an aqueous medium such as sodium chloride injection, Ringer's solution injection, isotonic glucose injection solution, sterile water injection solution, or glucose and lactic acid forest.
- non-aqueous media such as: non-volatile oils of plant origin, cottonseed oil, corn oil, sesame oil, or peanut oil, antibacterial substances at bacterial or fungal inhibitory concentrations, isotonic agents such as sodium chloride or glucose Buffers such as phosphate or citrate buffers, antioxidants such as sodium bisulfate, local anesthetics such as procaine hydrochloride, suspending and dispersing agents such as sodium carboxymethyl cellulose, hydroxypropyl Methylcellulose or polyvinylpyrrolidone, emulsifiers such as polysorbate 80 (Tween-80), integration agents such as EDTA (ethylenediaminetetraacetic acid) or EGTA (ethylene glycol bis (2-aminoaminoe
- Antibacterial agents as carriers can be added to pharmaceutical compositions in multiple-dose containers, including phenols or cresols, mercury preparations, benzyl alcohol, chlorobutanol, methyl and propyl parabens, Thimerosa, ammonium chlorobenzyl and chlorophenethyl ammonium.
- Suitable excipients may include, for example, water, salt, glucose, glycerol or ethanol.
- Suitable non-toxic auxiliary substances may include, for example, emulsifiers, pH buffers, stabilizers, solubilizers, or the like, such as sodium acetate, sorbitan laurate, triethanolamine oleate, or cyclodextrin. substance.
- the pharmaceutical composition may be a liquid solution, suspension, emulsion, pill, capsule, tablet, sustained release preparation or powder.
- Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, polyvinylpyrrolidone, sodium saccharin, cellulose, magnesium carbonate, and the like.
- the pharmaceutical composition is formulated as an injectable composition.
- injectable pharmaceutical compositions can be prepared in any conventional form, for example, liquid solvents, suspensions, emulsifiers or solid forms suitable for producing liquid solvents, suspensions or emulsifiers.
- Injectable preparations may include ready-to-use sterile and / or pyrogen-free solutions, sterile dry solubles that are combined with solvents before use, such as lyophilized powder, including subcutaneous tablets, sterile suspensions for injection, Sterile dry insoluble products that are combined with the medium before use, and sterile and / or pyrogen-free emulsions.
- the solvent may be an aqueous phase or a non-aqueous phase.
- the unit-dose injectable preparation is packaged in an ampoule, a tube, or a syringe with a needle. It is known in the art that all preparations for injection should be sterile and pyrogen-free.
- sterile lyophilized powders can be prepared by dissolving the antibodies or antigen-binding fragments thereof disclosed herein in a suitable solvent.
- the solvent may contain a powder or a reconstituted solution prepared from the powder, or other pharmacological components to improve the stability. Suitable excipients include, but are not limited to, water, glucose, sorbitol, fructose, corn syrup, xylitol, glycerol, glucose, brown sugar, or other suitable substances.
- the solvent may contain a buffer, such as a citrate buffer, a sodium or potassium phosphate buffer, or other buffers known to those skilled in the art. In one embodiment, the pH of the buffer is neutral.
- each vial can contain a single dose or multiple doses of the anti-OX40 antibody or antigen-binding fragment thereof or a combination thereof.
- the amount of filling in each vial can be slightly higher than that required for each dose or multiple doses (e.g., a 10% excess), thereby ensuring accurate sampling and precise administration.
- the lyophilized powder can be stored under appropriate conditions, such as in the range of about 4 ° C to room temperature.
- the lyophilized powder was reconstituted with water for injection to obtain a preparation for injection administration.
- the lyophilized powder can be redissolved in sterile pyrogen-free water or other suitable liquid carriers. The exact amount is determined by the chosen therapy and can be determined based on experience.
- Methods of treatment comprising administering a therapeutically effective amount of a fusion protein described herein to a subject in need thereof.
- the therapeutically effective dose of the fusion protein provided in this application depends on a variety of factors known in the art, such as weight, age, past medical history, current treatment, subject's health and potential for cross-infection, allergies, hypersensitivity, and side effects, and Route of administration and extent of tumor development. Those skilled in the art, such as a doctor or veterinarian, can proportionally reduce or increase the dose according to these or other conditions or requirements.
- the fusion proteins provided herein can be administered at a therapeutically effective dose of between about 0.01 mg / kg and about 100 mg / kg. In certain embodiments, the fusion protein is administered at a dose of about 50 mg / kg or less, and in certain embodiments, the dosage is 10 mg / kg or less, 5 mg / kg or less, 1 mg / kg or less, 0.5 mg / kg or less or 0.1 mg / kg or less.
- a particular dose can be administered at multiple intervals, such as once a day, twice a day or more, twice a month or more, once a week, once every two weeks, once every three weeks, once a month, or every two Once a month or more.
- the dosage administered may vary with the course of treatment.
- the initial administered dose may be higher than the subsequent administered dose.
- the administered dose is adjusted during the course of treatment based on the response of the subject to be administered.
- the dosing regimen can be adjusted to achieve an optimal response (eg, a therapeutic response). For example, it may be administered in a single dose or in divided doses over two periods.
- the fusion protein disclosed in the present invention can be administered by means known in the art, such as injection (e.g., subcutaneous injection, intraperitoneal injection, intravenous injection, including intravenous drip, intramuscular or intradermal injection) or non-injection Administration (eg, oral, nasal, sublingual, rectal, or topical).
- injection e.g., subcutaneous injection, intraperitoneal injection, intravenous injection, including intravenous drip, intramuscular or intradermal injection
- non-injection Administration eg, oral, nasal, sublingual, rectal, or topical.
- the fusion protein can be used to treat disorders related to its molecular mechanism, including tumors and cancers, such as non-small cell lung cancer, small cell lung cancer, renal cell carcinoma, colorectal cancer, ovarian cancer, breast cancer, pancreas Dirty cancer, gastric cancer, bladder cancer, esophageal cancer, mesothelioma, melanoma, head and neck cancer, thyroid cancer, sarcoma, prostate cancer, glioblastoma, cervical cancer, thymic cancer, leukemia, lymphoma, bone marrow Neoplasms, mycoses fungoids, Merkel cell carcinoma and other malignant hematological diseases such as classic Hodgkin lymphoma (CHL), primary mediastinal large B-cell lymphoma, T cells / histiocytic cells Of B-cell-rich lymphomas, EBV-positive and negative PTLD and EBV-related diffuse large B-cell lymphoma (DLBCL), plasmablastic lympho
- CHL
- the conditions treated by the fusion protein include chronic viral infections, such as hepatitis B, hepatitis C, herpes virus, Epstein-Barr virus, HIV, cytomegalovirus, herpes simplex virus type I, simplex Herpesvirus type 2, human papilloma virus, adenovirus viral infection, Kaposi's sarcoma-associated herpes virus epidemic, Torquetenovirus, JC virus, or BK virus, etc.
- chronic viral infections such as hepatitis B, hepatitis C, herpes virus, Epstein-Barr virus, HIV, cytomegalovirus, herpes simplex virus type I, simplex Herpesvirus type 2, human papilloma virus, adenovirus viral infection, Kaposi's sarcoma-associated herpes virus epidemic, Torquetenovirus, JC virus, or BK virus, etc.
- the application further provides methods for using the fusion proteins.
- the application provides a method of treating a condition or disorder associated with the fusion protein mechanism in an individual, comprising administering a therapeutically effective amount of the fusion protein described herein.
- the fusion protein disclosed in the present invention can be administered alone or in combination with one or more other treatment means or substances.
- the fusion protein disclosed in the present invention can be used in combination with chemotherapy, radiation therapy, cancer treatment surgery (such as tumor resection), antiviral drugs, one or more antiemetic drugs or other chemotherapy-induced complications, or any other application.
- chemotherapy radiation therapy
- cancer treatment surgery such as tumor resection
- antiviral drugs one or more antiemetic drugs or other chemotherapy-induced complications, or any other application.
- the fusion protein disclosed in the present invention when used in combination with one or more therapeutic substances, it may be administered simultaneously with the one or more therapeutic substances.
- the fusion protein can be administered simultaneously as part of the same pharmaceutical composition.
- fusion proteins that are "combined" with other therapeutic substances need not be administered at the same time or in the same composition as the therapeutic substance.
- a fusion protein administered before or after another therapeutic substance is also considered to be “combined” with the therapeutic substance, even if the fusion protein is administered differently from the second substance.
- Drug administration can be used in accordance with the methods of the product description of the other therapeutic substance, or refer to the surgeon's desk reference book 2003 (Physicians' Desk Reference, 57th Ed Medical; Economics Company; ISBN: 1563634457; 57th Edition (November 2002)), or refer to other methods known in the art.
- the therapeutic substance is capable of inducing or enhancing an immune response against cancer.
- tumor vaccines can be used to induce an immune response to certain tumors or cancers.
- Cytokine therapy can be used to improve the presentation of tumor antigens to the immune system.
- cytokine therapy examples include, but are not limited to, interferons such as interferon ⁇ , ⁇ , and ⁇ , colony stimulating factors such as macrophage CSF, granulocyte macrophage CSF, and granulocyte CSF, and interleukins such as 1L-L, 1L-1a , 1L-2, 1L3, 1L-4, 1L-5, 1L-6, 1L-7, 1L-8, 1L-9, 1L-10, 1L-ll and 1L-12, tumor necrosis factors such as TNF- ⁇ and TNF- ⁇ .
- interferons such as interferon ⁇ , ⁇ , and ⁇
- colony stimulating factors such as macrophage CSF, granulocyte macrophage CSF, and granulocyte CSF
- interleukins such as 1L-L, 1L-1a , 1L-2, 1L3, 1L-4, 1L-5, 1L-6, 1L-7, 1L-8, 1L-9, 1L-10, 1L-ll and 1
- agents that inactivate immunosuppressive targets such as PD-L1 / PD-1 antibodies, TGF- ⁇ inhibitors, IL-10 inhibitors, and Fas ligand inhibitors can also be used.
- Another group of agents includes those that activate an immune response against tumors or cancer cells, such as those that increase T cell activation (such as T cell co-stimulatory agonists such as CTLA-4, ICOS), and improve dendritic cell function And antigen-presenting ones.
- Example 1 Preparation of the fusion protein of the present invention
- This example exemplifies the design and expression of several anti-OX40 antibody-human interferon fusion proteins.
- the heavy and light chain sequences of the anti-human OX40 activating antibody are derived from MT01-C1 or MT01-C1 (G2) in Chinese patent 201711476160.3, where "MT01-C1" refers to UMY02-L1, UMY02-L2 and UMY02-L3 has the same VH (SEQ ID NO: 7) and VL (SEQ ID NO: 8) sequences, and the heavy and light chain constant regions are monoclonal antibodies of human IgG1 and ⁇ chains, respectively.
- MT01-C1 (G2) means that it has the same VH (SEQ ID ID NO: 7) and VL (SEQ ID ID NO: 8) sequences as UMY02-L1, UMY02-L2, and UMY02-L3, and the heavy and light chain constant regions, respectively Monoclonal antibodies to human IgG2 and kappa chains.
- the interferon IFN ⁇ 2b sequence is taken from human interferon IFN ⁇ 2b (NP_000596.2), and its amino acid sequence is shown in SEQ ID NO: 9.
- UY02-L1 refers to a heavy chain, a peptide linker, and a human interferon as shown in SEQ ID NO: 10, and a light chain as shown in SEQ ID NO: 11 in which a peptide linker is passed through the carboxyl end of the heavy chain.
- a fusion protein of human interferon IFN ⁇ 2b as shown in SEQ ID NO: 9 was linked.
- UY02-L2 refers to a heavy chain as shown in SEQ ID NO: 12, a light chain as shown in SEQ ID NO: 13, a peptide linker, and human interferon, in which the carboxyl end of the light chain is connected by a peptide linker
- UY02-L3 refers to a heavy chain as shown in SEQ ID NO: 14, a light chain as shown in SEQ ID NO: 13, a peptide linker and human interferon, in which the carboxyl end of the light chain is connected by a peptide linker
- the fusion protein of human interferon IFN ⁇ 2b as shown in SEQ ID NO: 9.
- the cDNA sequences encoding the antibody heavy and light chains of the fusion protein were cloned into mammalian cell expression vector pcDNA3.4, respectively.
- the heavy chain expression plasmid and light chain expression plasmid were transfected into HEK293 cells with Lipofectamine 2000 transfection reagent (Invitrogen) at a molar ratio of 2: 1, and cultured at 37 ° C and 5% carbon dioxide for 7 days. The culture supernatant was collected, and the antibody in the supernatant was purified by Protein A affinity chromatography. The purified antibody was dialyzed against PBS solution, freeze-dried and concentrated, and then stored at -20 ° C.
- a 96-well high affinity plate was coated with a human OX40 protein solution at a concentration of 1 ⁇ g / mL at 100 ⁇ L / well, and shaken overnight at 4 ° C. The next day was washed three times with 300 ⁇ L of PBST (Tween 20: 0.5 ⁇ ), and then blocked with 100 ⁇ L / well of 5% BSA / PBS for 2 hours and shaken at room temperature. 300 ⁇ L PBST was washed 3 times. A gradient dilution solution of the fusion protein sample was prepared in PBS. Add 100 ⁇ L / well to a 96-well plate and shake at room temperature for 1 hour. 300 ⁇ L PBST was washed 3 times.
- a secondary antibody goat anti-human IgG HRP solution was prepared, added to a 96-well plate at 100 ⁇ L / well, and shaken at room temperature for 1 hour. 300 ⁇ L PBST was washed 4 times. Add 100 ⁇ L / well TMB and develop color for 20 min. 100 ⁇ L / well of 0.6 N H 2 SO 4 was added to terminate the color development, and the OD 450 nm was detected.
- Example 3 Binding to human, cynomolgus monkey and mouse OX40 (FACS)
- CHO cells were transfected with an expression plasmid encoding human, cynomolgus monkey or mouse OX40 protein and cultured for 48 hours.
- the OX40 antibody MT01-L1 concentration gradient solution was prepared with PBS to prepare a final concentration of 10 ⁇ working solution.
- CHO-hOX40 cells were collected, washed and counted in PBS, diluted to 2 * 10 6 / ml cell suspension; 10 ⁇ L of OX40 antibody MT01-L1 working solution was added to 100 ⁇ L of cell suspension, and incubated at 4 ° C in the dark for 30 min; washed with PBS After 2 times, add the secondary antibody, incubate at 4 ° C in the dark for 30 min, wash once with PBS, and then suspend it in 400 ⁇ L FACS buffer, and then test on the machine. As shown in FIG. 3, the results show that MT01-L1 is bound to human OX40 (hsOX40), and its EC50 is 0.66 ⁇ g / mL.
- the inventors tested the binding of the antibody to CHO cells expressing mouse OX40, and found that MT01-L1 did not bind to mouse OX40 (msOX40).
- the inventors of the present application tested the binding of the antibody to CHO cells expressing cynomolgus monkey OX40, and found that MT01-L1 significantly binds to cynomolgus monkey OX40 (cyOX40).
- Interferon receptors are highly expressed on Daudi cells (ATCC), so interferons have biological activity on them.
- Daudi cells were plated at 20,000 cells / 90 ⁇ L / well in a 96-well plate.
- the sample to be tested was prepared into 10 ⁇ working solution with a concentration gradient dilution, and added to the 96-well plate at 10 ⁇ L / well, and placed in a 37 ° C incubator.
- CCK8 detects OD450 and calculates the proliferation inhibition rate of cells in each well. This inhibition rate reflects the activity of the interferon in the sample.
- the experimental results showed that the fusion proteins UMY02-L1, UMY02-L2, and UMY02-L3 have inhibitory activity (IC50) on Daudi cells of 9.549, 9.152, and 27.44 pM ( Figure 4).
- the inventors of the present application have constructed a cell experimental system for detecting OX40 activator. Specifically, the inventors of the present application constructed a "Jurkat-OX40-NF ⁇ B-luciferase reporter gene" stable transfected cell line, and after mixing the OX40 activating antibody with the stable transduced cell line and HEK293 cells overexpressing FcR Can activate the expression of the NF ⁇ B-luciferase reporter gene.
- the fusion protein concentration gradient solution was prepared with PBS to prepare a final concentration of 2 ⁇ working solution, which was operated on ice.
- Jurkat-NFkB-luc-OX40 cells and HEK293 cells overexpressing FcR were collected, resuspended in culture medium after centrifugation, and plated into 384-well plates.
- Add the fusion protein working solution and an appropriate amount of cell suspension After incubation for 5 hours, add the One-Glo (Promega) detection reagent. After mixing, use the Pherastar autofocus fluorescence luminometer microplate reader to detect the fluorescence signal.
- the OX40 antibody MT01-C1 As shown in Figure 5, after detection, the OX40 antibody MT01-C1, the fusion proteins UMY02-L1, UMY02-L2, and UMY02-L3 activated the NF ⁇ B-luciferase reporter genes in the above experimental system with EC50 of 0.04523, 0.02437, and 0.02837, respectively. And 0.02907ng / mL.
- the results showed that FcR-mediated OX40 activation activities of UMY02-L1, UMY02-L2, and UMY02-L3 were stronger than MT01-C1.
- the "Jurkat-OX40-NF ⁇ B-luciferase reporter gene" stably transformed cell line was plated at 10,000 cells / well into a 384-well plate.
- PBS was used to configure the fusion protein concentration gradient solution or the control solution to prepare a final concentration of 2 ⁇ working solution, which was operated on ice.
- a 384-well plate add the fusion protein or interferon working solution and an appropriate amount of cell suspension. After 5 hours of incubation, add the One-Glo (Promega) detection reagent. After mixing, use the Pherastar autofocus fluorescence luminometer and microplate reader to detect the fluorescence. signal.
- the final concentration of all antibodies or interferons in the experiment was 10 nM.
- the OX40 antibody MT01-C1 or MT01-C1 (G2) has no activation effect on the OX40 signaling pathway in Jurkat cells, and the addition of interferon IFN ⁇ 2b alone or the simultaneous addition of OX40 antibodies MT01-C1 and IFN ⁇ 2b is only less than 40% compared with the control group. Activation effect.
- the fusion proteins UMY02-L1, UMY02-L2, or UMY02-L3 were added, the OX40 signaling pathway of Jurkat cells was significantly activated, and the degree of activation was significantly greater than that of the interferon group alone ( Figure 6b).
- mice Eighteen female C57BL / 6 mice, 6-8 weeks old, were divided into 3 groups of 6 mice each. They were given intravenous injection of UMY02-L1, UMY02-L3, and MT01-C1. The dose was 5 mg / kg.
- UMY02-L1 and UMY02-L3 collect the peripheral venous blood of animals before and after administration, and 1, 2, 6, 24, 48, 72, 96, 174, 220, and 288 hours after administration;
- MT01-C1 collect Peripheral venous blood of the animals before, and 1, 2, 6, 24, 48, 72, 96, 192, and 312 hours after administration; serum was collected by centrifugation. Three animal serums were collected at each time point, and six animals in each group were collected blood samples at different time points.
- a human OX40 protein solution at a concentration of 1 ⁇ g / mL was coated with a 96-well high affinity plate at 100 ⁇ L / well, and shaken at 4 ° C. overnight. The next day was washed three times with 300 ⁇ L of PBST (Tween 20: 0.5 ⁇ ), and then blocked with 100 ⁇ L / well of 5% BSA / PBS for 1 hour and shaken at room temperature. 300 ⁇ L PBST was washed 4 times. PBS was used to prepare 100-fold dilutions of the serum samples to be tested and the serum solutions of different concentrations of reference substance. Add 100 ⁇ L / well to a 96-well plate and shake at room temperature for 1.5 hours.
- 300 ⁇ L PBST was washed 4 times.
- a secondary antibody donkey anti-human IgG HRP Jackson ImmunoResearch, article number 709-035-149
- 300 ⁇ L PBST was washed 4 times.
- 100 ⁇ L / well of 0.6 N H 2 SO 4 was added to stop the color development, and the OD 450 nm was detected.
- a 0.5 ⁇ g / mL rabbit anti-human IFN (abcam, article number ab222552) solution was prepared, added to a 96-well plate at 100 ⁇ L / well, and shaken at room temperature for 1.5 hours. 300 ⁇ L PBST was washed 4 times.
- a secondary antibody goat anti-rabbit IgG HRP (Kingsui Biotechnology, Cat. No. A00098) solution was prepared, added to a 96-well plate at 100 ⁇ L / well, and shaken at room temperature for 1 hour. 300 ⁇ L PBST was washed 4 times. Add 100 ⁇ L / well TMB and develop color for 20 min.
- mice were intravenously injected with 5 mg / kg of UMY02-L3 and UMY02-L1. After 7 days of administration, the serum drug concentration of UMY02-L3 was still above 10 ⁇ g / mL. After 12 days of administration, UMY02- L1 serum drug concentration was still above 10 ⁇ g / mL. It is shown that the fusion proteins UMY02-L3 and UMY02-L1 have similar pharmacokinetic properties in mice as the antibody drug MT01-C1, which greatly prolongs the half-life of 2-3 hours compared to IFN ⁇ 2b (see the product manual of Merck's product Intron® A ).
- Example 8 Effect of antibody subtype and peptide linker length on antibody fusion protein activity
- the heavy chains of the fusion proteins UMY02-L4, UMY02-L5 and UMY02-L6 are shown in SEQ ID NO: 14, and the light chain and human interferon are shown in SEQ ID ID NO: 13 and the light chain and human interferon, The only difference is the number of peptide linkers, as shown in Table 1 below.
- the heavy chains of the fusion proteins UMY02-L7 and UMY02-L8 are shown in SEQ ID NO: 15, and the light chain and human interferon are shown in SEQ ID ID NO: 13, and the difference is only in the peptide
- the number of connectors is shown in Table 1 below.
- Example 4 According to the method of Example 4, the effects of these antibody fusion proteins on the proliferation of Daudi cells were detected. As a result, it was found that different peptide linker lengths had a significant effect on the interferon activity of antibody fusion proteins. The shorter the peptide linker length, the lower the interferon activity. (See Table 1).
- OX40 antibody MT01-C1 and each antibody fusion protein can effectively activate the OX40 signaling pathway of Jurkat-OX40 cells.
- OX40 activating antibodies can also be fused to interferon mutants. Compared with wild-type interferon, these mutant interferons have lower specific activity, so they can work together with OX40 antibody at the same drug concentration, while avoiding toxic side effects caused by excessive interferon activity.
- the heavy chains of fusion proteins UMY02-L13, UMY02-L14, UMY02-L15, UMY02-L16, UMY02-L17 and UMY02-L18 are shown in SEQ ID NO: 15, and the light chain and human interferon are shown in SEQ ID ID NO:
- the light chain and human interferon shown in 13 are only different:
- the peptide linkers of UMY02-L13, UMY02-L14, UMY02-L15, UMY02-L16, and UMY02-L18 are (GGGGS) 2, and the peptide linkers of UMY02-L17 are (GGGGS) 3;
- Interferons of the fusion proteins UMY02-L13, UMY02-L14, UMY02-L15, UMY02-L16, UMY02-L17 and UMY02-L18 are mutated as shown in Table 2 on the basis of SEQ ID ID NO: 9.
- Example 4 According to the method of Example 4, the effects of these antibody fusion proteins on the proliferation of Daudi cells were detected. As a result, it was found that the interferon activity of the mutant antibody fusion protein was significantly lower than that of wild-type IFN ⁇ 2b (Table 2). At the same time, the activation effect of these antibody fusion proteins on the OX40 signaling pathway of Jurkat cells was detected according to the method of Example 5, and the results showed that the OX40 activation activity of these mutant antibody fusion proteins was basically unchanged (Table 2).
- the amino acid position number of the mutation is indicated by referring to the amino acid sequence SEQ ID NO: 9 of the wild-type human interferon IFN ⁇ 2b.
- IFN ⁇ 2b The activity of IFN ⁇ 2b is defined as 1.
- the inventors fused the heavy chain of the OX40 antibody with different types of interferons to prepare different fusion proteins, among which the heavy chains of OX40IFN- ⁇ 2a, OX40-IFN ⁇ , OX40-IFN ⁇ , and OX40-IFN ⁇ 3 are as shown in SEQ ID ID NO: 15; OX40IFN- ⁇ 2a light chain, peptide linker and interferon are shown in SEQ ID NO: 16; OX40IFN- ⁇ light chain, peptide linker and interferon are shown in SEQ ID NO: 17; OX40IFN- ⁇ light chain, The peptide linker and interferon are shown in SEQ ID NO: 18; the OX40IFN- ⁇ 3 light chain, peptide linker and interferon are shown in SEQ ID NO: 19.
- the killing effect of these antibody fusion proteins on tumor cells was tested.
- the inventors mixed and co-cultured human-derived PBMCs and ovarian cancer SKOV3 cells at a ratio of 20: 1, and administered PBS (control) or 0.1nM OX40 mAb MT01-C1 and different antibody fusion proteins (OX40IFN - ⁇ 2a, OX40-IFN ⁇ , OX40-IFN ⁇ , OX40-IFN ⁇ 3) stimulation.
- the CCK8 kit was used to detect the cytotoxic effect of PBMC on SKOV3 cells.
- FIG. 8 shows that PBMC has no inhibitory effect on SKOV3 cell proliferation without stimulation; OX40 monoclonal antibody stimulates SKOV3 inhibition rate of about 28% by PBMC; and cytotoxicity of PBMC is strong under stimulation of fusion protein Stimulated by OX40 monoclonal antibody, OX40 monoclonal antibody MT01-C1-IFN ⁇ 2a and OX40 monoclonal antibody MT01-C1-FN ⁇ were significantly enhanced, with inhibition rates of 60% and 63%, respectively.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Immunology (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Zoology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Chemical & Material Sciences (AREA)
- Gastroenterology & Hepatology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Epidemiology (AREA)
- Biotechnology (AREA)
- Biomedical Technology (AREA)
- Virology (AREA)
- Toxicology (AREA)
- Microbiology (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Mycology (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Cell Biology (AREA)
- Peptides Or Proteins (AREA)
- Preparation Of Compounds By Using Micro-Organisms (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
Claims (16)
- 一种融合蛋白,其包含:a)特异性结合人OX40的抗体或其抗原结合片段;和b)人干扰素;其中所述人干扰素直接或通过肽接头连接至所述抗体的轻链或重链的羧基端或氨基端。
- 根据权利要求1所述的融合蛋白,其中所述特异性结合人OX40的抗体或其抗原结合片段包含:抗体重链可变区,该重链可变区包含具有SEQ ID NO:1的氨基酸序列的VH CDR1、具有SEQ ID NO:2的氨基酸序列的VH CDR2、和具有SEQ ID NO:3的氨基酸序列的VH CDR3;和抗体轻链可变区,该轻链可变区包含具有SEQ ID NO:4的氨基酸序列的VL CDR1、具有SEQ ID NO:5的氨基酸序列的VL CDR2、和具有SEQ ID NO:6的氨基酸序列的VL CDR3。
- 根据权利要求2所述的融合蛋白,其中所述重链可变区包含SEQ ID NO:7所示的氨基酸序列,所述轻链可变区包含SEQ ID NO:8所示的氨基酸序列。
- 根据权利要求1-3任一项所述的融合蛋白,其中所述特异性结合人OX40的抗体或其抗原结合片段是骆驼化单域抗体、scFv、scFv二聚体、BsFv、dsFv、dsFv2、dsFv-dsFv'、Fv片段、Fab、Fab'、F(ab')2、ds双功能抗体、纳米抗体、域抗体或双价域抗体。
- 根据权利要求1-4任一项所述的融合蛋白,其中所述抗体进一步包含免疫球蛋白的恒定区;优选地,所述恒定区为人IgG1、IgG2或IgG4的恒定区。
- 根据权利要求1-5任一项所述的融合蛋白,其中所述人干扰素选自I型人干扰素、II型人干扰素和III型人干扰素;优选地,所述人干扰素为IFNα2a、IFNβ、IFNγ、IFNλ3、IFNα2b;更优选地,所述人干扰素为IFNα2b,其氨基酸序列如SEQ ID NO:9所示;进一步优选地,所述人干扰素为IFNα2b的突变体,其在SEQ ID NO:9所示的氨基酸序列上,具有选自以下的一种或多种突变:T106A、R149A、A145G、A145D、R120A、L117A;更进一步优选地,所述IFNα2b的突变体在SEQ ID NO:9所示的氨基酸序列上具有选自以下的双突变:T106A/A145D、T106A/R149A、T106A/A145G、T106A/R120A、T106A/L117A。
- 根据权利要求1-7任一项所述的融合蛋白,其中所述肽接头选自(G)n、KESGSVSSEQLAQFRSLD、EGKSSGSGSESKST、GSAGSAAGSGEF、(GGGGS)n、(GGSGG)n;优选地,所述肽接头为(GGGGS)n,其中n为0-5之间的整数;优选地,n为1-3之间的整数。
- 根据权利要求1-7任一项所述的融合蛋白,其中所述所述融合蛋白选自:UMY02-L1、UMY02-L2、UMY02-L3、UMY02-L4、UMY02-L5、UMY02-L6、UMY02-L7、UMY02-L8、UMY02-L13、UMY02-L14、UMY02-L15、UMY02-L16、UMY02-L17、UMY02-L18、OX40IFN-α2a、OX40-IFNβ、OX40-IFNγ、OX40-IFNλ3中的一种或多种。
- 一种分离的多核苷酸,其编码根据权利要求1-8中任一项所述的融合蛋白。
- 一种载体,其包括根据权利要求9所述的分离的多核苷酸。
- 一种宿主细胞,其包括根据权利要求10所述的载体。
- 一种表达根据权利要求1-8任一项所述的融合蛋白的方法,其包括在能够表达根据权利要求9所述的分离的多核苷酸的条件下培养根据权利要求11所述的宿主细胞。
- 一种试剂盒,其包括根据权利要求1-8中任一项所述的融合蛋白。
- 一种药物组合物,其包括根据权利要求1-8中任一项所述的融合蛋白以及药学上可接受的载体。
- 根据权利要求1-8中任一项所述的融合蛋白在制备药物中的用途,所述药物用于治疗能通过增强免疫应答和/或能通过暴露于干扰素而受益的病况;优选地,其中所述病况是癌症或病毒感染;更优选地,所述病毒感染为乙肝病毒感染。
- 一种治疗能通过增强免疫应答和/或能通过暴露于干扰素而受益的病况的方法,所述方法包括给予有需要的受试者治疗有效量的如权利要求1-8中任一项所述的融合蛋白或如权利要求14所述的药物组合物;优选地,其中所述病况是癌症或病毒感染;更优选地,其中所述病毒感染为乙肝病毒感染。
Priority Applications (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP19857108.5A EP3848397A4 (en) | 2018-09-04 | 2019-09-03 | FUSION PROTEIN AND ITS APPLICATION IN THE PREPARATION OF A MEDICINE FOR THE TREATMENT OF A TUMOR AND/OR A VIRAL INFECTION |
JP2021510232A JP7404343B2 (ja) | 2018-09-04 | 2019-09-03 | 融合タンパク質、並びに腫瘍及び/又はウイルス感染の治療薬を製造するためのその使用 |
CN201980054796.XA CN112585169A (zh) | 2018-09-04 | 2019-09-03 | 融合蛋白及其在制备用于治疗肿瘤和/或病毒感染的药物中的应用 |
KR1020217006850A KR20210056344A (ko) | 2018-09-04 | 2019-09-03 | 융합 단백질 및 그가 종양 및/또는 바이러스 감염을 치료하기 위한 약물 제조에서의 응용 |
AU2019336426A AU2019336426A1 (en) | 2018-09-04 | 2019-09-03 | Fusion protein and its application in preparing medicine for treating tumor and/or viral infection |
US17/192,399 US20210198375A1 (en) | 2018-09-04 | 2021-03-04 | Fusion protein and its applicaton in preparing medicine for treating tumor and/or viral infection |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201811024783.1 | 2018-09-04 | ||
CN201811024783 | 2018-09-04 |
Related Child Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US17/192,399 Continuation-In-Part US20210198375A1 (en) | 2018-09-04 | 2021-03-04 | Fusion protein and its applicaton in preparing medicine for treating tumor and/or viral infection |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020048454A1 true WO2020048454A1 (zh) | 2020-03-12 |
Family
ID=69721450
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2019/104217 WO2020048454A1 (zh) | 2018-09-04 | 2019-09-03 | 融合蛋白及其在制备用于治疗肿瘤和/或病毒感染的药物中的应用 |
Country Status (6)
Country | Link |
---|---|
EP (1) | EP3848397A4 (zh) |
JP (1) | JP7404343B2 (zh) |
KR (1) | KR20210056344A (zh) |
CN (1) | CN112585169A (zh) |
AU (1) | AU2019336426A1 (zh) |
WO (1) | WO2020048454A1 (zh) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN114053403A (zh) * | 2020-07-31 | 2022-02-18 | 南京优迈生物科技有限公司 | 融合蛋白在制备疫苗佐剂或预防和治疗病毒感染的药物中的用途 |
WO2024067785A1 (zh) * | 2022-09-29 | 2024-04-04 | 中国科学院生物物理研究所 | 基于干扰素协同免疫检查点阻断抗体治疗慢性乙型肝炎 |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN114966061B (zh) * | 2022-07-28 | 2022-10-21 | 中国食品药品检定研究院 | 一种抗ox40抗体的生物活性检测方法 |
Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0404097A2 (de) | 1989-06-22 | 1990-12-27 | BEHRINGWERKE Aktiengesellschaft | Bispezifische und oligospezifische, mono- und oligovalente Rezeptoren, ihre Herstellung und Verwendung |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
WO1994004678A1 (en) | 1992-08-21 | 1994-03-03 | Casterman Cecile | Immunoglobulins devoid of light chains |
WO1994025591A1 (en) | 1993-04-29 | 1994-11-10 | Unilever N.V. | PRODUCTION OF ANTIBODIES OR (FUNCTIONALIZED) FRAGMENTS THEREOF DERIVED FROM HEAVY CHAIN IMMUNOGLOBULINS OF $i(CAMELIDAE) |
US6005079A (en) | 1992-08-21 | 1999-12-21 | Vrije Universiteit Brussels | Immunoglobulins devoid of light chains |
WO2014028502A1 (en) * | 2012-08-13 | 2014-02-20 | ImmunGene Inc. | Engineered antibody-interferon fusion molecules for treatment of autoimmune diseases |
CN107106656A (zh) * | 2014-10-29 | 2017-08-29 | 梯瓦制药澳大利亚股份有限公司 | 干扰素α2b变体 |
CN108218990A (zh) * | 2017-12-29 | 2018-06-29 | 南京优迈生物科技有限公司 | 分离的抗体或其抗原结合片段及其在肿瘤治疗中的应用 |
Family Cites Families (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DK3411398T3 (da) | 2016-02-05 | 2024-06-24 | Orionis Biosciences BV | Målrettede terapeutiske midler og anvendelse heraf |
US20210198375A1 (en) * | 2018-09-04 | 2021-07-01 | Nanjing Umab-Biopharma Co., Ltd. | Fusion protein and its applicaton in preparing medicine for treating tumor and/or viral infection |
-
2019
- 2019-09-03 WO PCT/CN2019/104217 patent/WO2020048454A1/zh unknown
- 2019-09-03 KR KR1020217006850A patent/KR20210056344A/ko unknown
- 2019-09-03 EP EP19857108.5A patent/EP3848397A4/en active Pending
- 2019-09-03 JP JP2021510232A patent/JP7404343B2/ja active Active
- 2019-09-03 CN CN201980054796.XA patent/CN112585169A/zh active Pending
- 2019-09-03 AU AU2019336426A patent/AU2019336426A1/en active Pending
Patent Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0404097A2 (de) | 1989-06-22 | 1990-12-27 | BEHRINGWERKE Aktiengesellschaft | Bispezifische und oligospezifische, mono- und oligovalente Rezeptoren, ihre Herstellung und Verwendung |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
WO1994004678A1 (en) | 1992-08-21 | 1994-03-03 | Casterman Cecile | Immunoglobulins devoid of light chains |
US6005079A (en) | 1992-08-21 | 1999-12-21 | Vrije Universiteit Brussels | Immunoglobulins devoid of light chains |
WO1994025591A1 (en) | 1993-04-29 | 1994-11-10 | Unilever N.V. | PRODUCTION OF ANTIBODIES OR (FUNCTIONALIZED) FRAGMENTS THEREOF DERIVED FROM HEAVY CHAIN IMMUNOGLOBULINS OF $i(CAMELIDAE) |
WO2014028502A1 (en) * | 2012-08-13 | 2014-02-20 | ImmunGene Inc. | Engineered antibody-interferon fusion molecules for treatment of autoimmune diseases |
CN107106656A (zh) * | 2014-10-29 | 2017-08-29 | 梯瓦制药澳大利亚股份有限公司 | 干扰素α2b变体 |
CN108218990A (zh) * | 2017-12-29 | 2018-06-29 | 南京优迈生物科技有限公司 | 分离的抗体或其抗原结合片段及其在肿瘤治疗中的应用 |
Non-Patent Citations (31)
Title |
---|
"Contributions to Microbiology and Immunology", 1989, CARGER PRESS, article "Conjugate Vaccines" |
"Physicians' Desk Reference", November 2002, MEDICAL ECONOMICS COMPANY |
AI-LAZIKANI, B.CHOTHIA, C.LESK, A.M., J. MOL. BIOL., vol. 273, no. 4, 1997, pages 927 |
BURGESS JKCARLIN SPACK RAARNDT GMAU WWJOHNSON PRBLACK JLHUNT NH: "Detection and characterization of OX40 ligand expression in human airway smooth muscle cells: a possible role in asthma?", J ALLERGY CLIN IMMUNOL, vol. 113, 2004, pages 683 - 9 |
CARTER ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 163 - 167 |
CHOTHIA, C., J. MOL. BIOL., vol. 186, no. 3, 1985, pages 651 - 63 |
CHOTHIA, C., NATURE, vol. 342, no. 6252, 1989, pages 877 - 83 |
CHOTHIA, C.LESK, A.M., J. MOL. BIOL., vol. 196, 1987, pages 901 |
COMPAAN DMHYMOWITZ SG: "The crystal structure of the costimulatory OX40-OX40L complex", STRUCTURE, vol. 14, 2006, pages 1321 - 30, XP025134045, DOI: 10.1016/j.str.2006.06.015 |
CROFT M: "Control of immunity by the TNFR-related molecule OX40 (CD134", ANNU REV IMMUNOL, vol. 28, 2010, pages 57 - 78, XP055039398, DOI: 10.1146/annurev-immunol-030409-101243 |
GUSS ET AL., EMBO J., vol. 5, 1986, pages 1567 - 1575 |
HAMERS CASTERMAN C., NATURE, vol. 363, no. 6428, 1993, pages 446 - 8 |
HOLLIGER P. ET AL., PROC NATL ACAD SCI USA., vol. 90, no. 14, 1993, pages 6444 - 8 |
HUSTON JS ET AL., PROC NATL ACAD SCI USA, vol. 85, 1988, pages 5879 |
IMURA AHORI TIMADA KISHIKAWA TTANAKA YMAEDA MIMAMURA SUCHIYAMA T: "The human OX40/gp34 system directly mediates adhesion of activated T cells to vascular endothelial cells", J EXP MED., vol. 183, 1996, pages 2185 - 95, XP002915335, DOI: 10.1084/jem.183.5.2185 |
KOCH-NOLTE F. ET AL., FASEB J., vol. 21, no. 13, 2007, pages 3490 - 8 |
LINDMARK ET AL., J. LMMUNOL. METH., vol. 62, 1983, pages 1 - 13 |
MATHER ET AL., ANNALS N. Y. ACAD. SCI., vol. 383, 1982, pages 44 - 68 |
MATHER, BIOL. REPROD., vol. 23, 1980, pages 243 - 251 |
MUYLDERMANS S., J BIOTECHNOL, vol. 74, no. 4, 2001, pages 277 - 302 |
NGUYEN VK, IMMUNOLOGY, vol. 109, no. 1, 2003, pages 93 - 101 |
NGUYEN VK: "Heavy-chain antibodies in Camelidae: a case of evolutionary innovation", IMMUNOGENETICS, vol. 54, no. 1, 2002, pages 39 - 47 |
PUBLICOVER JGAGGAR AJESPERSEN JMHALAC UJOHNSON AJGOODSELL AAVANESYAN LNISHIMURA SLHOLDORF MMANSFIELD KG: "An OX40/OX40L interaction directs successful immunity to hepatitis B virus", SCI TRANSL MED, 2018 |
RIECHMANN L.MUYLDERMANS S., J IMMUNOL METHODS, vol. 231, no. 1-2, 1999, pages 25 - 38 |
ROGERS PRSONG JGRAMAGLIA IKILLEEN NCROFT M: "OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells", IMMUNITY, vol. 15, 2001, pages 445 - 55, XP002269749, DOI: 10.1016/S1074-7613(01)00191-1 |
See also references of EP3848397A4 |
SO TSONG JSUGIE KALTMAN ACROFT M: "Signals from OX40 regulate nuclear factor of activated T cells cl and T cell helper 2 lineage commitment", PROC NATL ACAD SCI USA., vol. 103, 2006, pages 3740 - 5 |
SONG JSO TCHENG MTANG XCROFT M: "Sustained survivin expression from OX40 costimulatory signals drives T cell clonal expansion", IMMUNITY, vol. 22, 2005, pages 621 - 31 |
SONG JSO TCROFT M: "Activation of NF-kappaBl by OX40 contributes to antigen-driven T cell expansion and survival", J IMMUNOL., vol. 180, 2008, pages 7240 - 8 |
URLAUB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216 |
WILLOUGHBY JGRIFFITHS JTEWS ICRAGG MS: "OX40: Structure and function - What questions remain?", MOL IMMUNOL, vol. 83, 2017, pages 13 - 22, XP029925307, DOI: 10.1016/j.molimm.2017.01.006 |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN114053403A (zh) * | 2020-07-31 | 2022-02-18 | 南京优迈生物科技有限公司 | 融合蛋白在制备疫苗佐剂或预防和治疗病毒感染的药物中的用途 |
WO2024067785A1 (zh) * | 2022-09-29 | 2024-04-04 | 中国科学院生物物理研究所 | 基于干扰素协同免疫检查点阻断抗体治疗慢性乙型肝炎 |
Also Published As
Publication number | Publication date |
---|---|
EP3848397A4 (en) | 2022-07-20 |
AU2019336426A1 (en) | 2021-04-29 |
JP2021536231A (ja) | 2021-12-27 |
JP7404343B2 (ja) | 2023-12-25 |
KR20210056344A (ko) | 2021-05-18 |
CN112585169A (zh) | 2021-03-30 |
EP3848397A1 (en) | 2021-07-14 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20210269528A1 (en) | Novel anti-pd-l1 antibodies | |
US10696745B2 (en) | Anti-PD-L1 antibodies | |
JP2022161997A (ja) | 新規抗pd-1抗体 | |
JP7317858B2 (ja) | 単離された抗体またはその抗原結合断片、および腫瘍治療におけるその使用 | |
CN106432501B (zh) | 新型抗pd-l1抗体 | |
US20210198375A1 (en) | Fusion protein and its applicaton in preparing medicine for treating tumor and/or viral infection | |
CN106243225B (zh) | 新型抗-pd-l1抗体 | |
CN111484555B (zh) | 新型双特异性cd3/cd20多肽复合物 | |
TWI844684B (zh) | 一種抗ceacam5的單殖株抗體及其製備方法和用途 | |
WO2020048454A1 (zh) | 融合蛋白及其在制备用于治疗肿瘤和/或病毒感染的药物中的应用 | |
TW202030206A (zh) | 新型雙特異性cd3/cd20多肽複合物 | |
CN113444180B (zh) | 靶向axl蛋白的抗体及其抗原结合片段、其制备方法和应用 | |
WO2022261846A1 (zh) | 靶向axl蛋白的抗体及其抗原结合片段、其制备方法和应用 | |
CN114829385A (zh) | 包含il-7变体的双功能分子 | |
WO2022116079A1 (zh) | 一种抗ceacam5的人源化抗体及其制备方法和用途 | |
CN116323671A (zh) | 具有增加的选择性的多靶向性双特异性抗原结合分子 | |
WO2021197393A1 (zh) | 一种抗人cd47抗体及其抗原结合片段、制备方法和应用 | |
CN114634577A (zh) | 融合蛋白及其制备用于治疗肿瘤和病毒感染的药物的应用 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 19857108 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2021510232 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2019857108 Country of ref document: EP Effective date: 20210406 |
|
ENP | Entry into the national phase |
Ref document number: 2019336426 Country of ref document: AU Date of ref document: 20190903 Kind code of ref document: A |