WO2019201301A1 - Anti-gitr antibody and use thereof - Google Patents

Anti-gitr antibody and use thereof Download PDF

Info

Publication number
WO2019201301A1
WO2019201301A1 PCT/CN2019/083241 CN2019083241W WO2019201301A1 WO 2019201301 A1 WO2019201301 A1 WO 2019201301A1 CN 2019083241 W CN2019083241 W CN 2019083241W WO 2019201301 A1 WO2019201301 A1 WO 2019201301A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
amino acid
acid sequence
gitr
seq
Prior art date
Application number
PCT/CN2019/083241
Other languages
French (fr)
Chinese (zh)
Inventor
翟天航
付凤根
黄威峰
刘军建
Original Assignee
信达生物制药(苏州)有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 信达生物制药(苏州)有限公司 filed Critical 信达生物制药(苏州)有限公司
Priority to CN201980022627.8A priority Critical patent/CN111918878B/en
Publication of WO2019201301A1 publication Critical patent/WO2019201301A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to novel antibodies and antibody fragments that specifically bind to GITR and compositions containing the antibodies or antibody fragments. Furthermore, the invention relates to nucleic acids encoding the antibodies or antibody fragments thereof, and host cells comprising the same, and related uses. Furthermore, the invention relates to the therapeutic and diagnostic use of these antibodies and antibody fragments.
  • GITR Glucocorticoid-induced tumor necrosis factor receptor
  • TNFR tumor necrosis factor receptor
  • GITRL GITR ligand
  • GITR is expressed at low levels in resting T cells; after T cell activation, expression is significantly upregulated. GITR is highly expressed constitutively in regulatory T cells (Treg), and when these cells are activated, expression is further upregulated (Nocentini and Riccardo, E.J. Immunol. 2005, 35: 1016-1022).
  • GITRL GITR ligands
  • APCs antigen presenting cells
  • GITRL binding of GITRL on APC to GITR on response T cells triggers GITR signaling, stimulates effector T cell activation and inhibits Treg cell activity.
  • GITR has several effects on effector T cells and regulatory T cells, including: costimulation and activation of effector T cells, and reduction of Treg cell suppression of effector T cells (Nocentini, Eur. J. Immunol. 2007, 37: 1165-1169).
  • GITR signaling pathway activation of the GITR signaling pathway can result in an increase in the immune response. Therefore, a substance capable of activating the GITR signaling pathway can activate an immune response, thereby increasing the body's ability to resist tumors and infections, and the GITR molecule is highly expressed in Treg cells in the tumor microenvironment, thereby utilizing the ADCC activity of the GITR antibody to clear the GITR molecule.
  • the expressed Treg cells can further enhance tumor killing activity.
  • GITR A number of antibodies to GITR have been studied in the prior art (see CN103951753A, CN105829343A, CN106459203A, WO2016196792A1, WO2017068186A9, etc.).
  • Such antibodies are agonists of GITR (i.e., are activating antibodies) that induce or enhance GITR signaling and are effective in treating a variety of GITR-related diseases or conditions that require an enhanced immune response. Since such an anti-GITR antibody is an activated antibody, its activation activity is a more critical indicator of its activity (for example, immunopotentiating activity) relative to its binding affinity to an antigen.
  • the invention thus provides a novel antibody that binds to GITR, as well as antigen-binding fragments thereof.
  • an anti-GITR antibody of the invention has one or more of the following characteristics:
  • T cells eg, CD4 T cells
  • Combination with an anti-PD-1 antibody can better inhibit tumor activity, for example, to reduce tumor volume in a subject without affecting the subject's body weight.
  • the invention provides an antibody or antigen-binding fragment thereof that binds to GITR, comprising three heavy chain CDRs (HCDRs) of the sequence set forth in SEQ ID NO: 17, 18, 19 or 20, and/or 3 light chain CDRs (LCDR) of the sequence set forth in SEQ ID NO: 21, 22, 23 or 24.
  • HCDRs heavy chain CDRs
  • LCDR light chain CDRs
  • the invention provides a nucleic acid encoding an antibody or fragment thereof of the invention, a vector comprising the nucleic acid, a host cell comprising the vector.
  • the invention provides methods of making an antibody or fragment thereof of the invention.
  • the invention provides immunoconjugates, pharmaceutical compositions, and combination products comprising an antibody of the invention.
  • the invention also provides methods of mediating ADCC or activating the NF-kappaB signaling pathway in a subject using an antibody of the invention, and methods of preventing or treating cancer or infection.
  • the invention also relates to a method of detecting GITR in a sample.
  • Figure 1 shows the ability of a chimeric antibody to bind to a CHO-S cell line expressing human GITR (CHO-hGITR) by flow cytometry.
  • Figure 2 shows the ability of humanized antibodies to bind to a human GITR-expressing CHO-S cell line (CHO-hGITR) by flow cytometry.
  • Figure 3 shows the ability of humanized antibodies to bind to CHO-S cell line (CHO-cynoGITR) expressing cynomolgus GITR by flow cytometry.
  • Figure 4 shows the MOA assay for the ability of humanized antibodies to activate Hela-GITR-NF-Kappa B luciferace cell lines.
  • Figure 5 shows the ability of humanized antibodies to activate IL-4 release from activated CD4 T cells.
  • Figure 6 shows the ability of humanized antibodies to activate CD4 T cells to release IFN-[gamma].
  • Figure 7 shows the MOA assay for the determination of humanized antibody ADCC activity.
  • Figure 8 shows the in vivo pharmacodynamic assay of HZ37G5.
  • Figure 9 shows the in vivo efficacy test of HZ22F4 molecule.
  • Figure 10 shows mouse body weight detection.
  • GITR refers to "glucocorticoid-induced TNF-related genes and/or polypeptides", also referred to in the art as TNF receptor superfamily 18 (TNFRSF18), and refers to any vertebrate source, including Any natural GITR of mammals such as primates (eg, humans, cynomolgus monkeys) and rodents (eg, mice and rats), unless otherwise indicated.
  • the term encompasses "full length", unprocessed GITR, and any form of GITR due to processing in cells.
  • the term also encompasses naturally occurring variants of GITR, such as splice variants or allelic variants.
  • the human GITR amino acid sequence can be found in GenBank Accession No.
  • the amino acid sequence of a particular human GITR polypeptide can be found in SEQ ID NO:41.
  • the amino acid sequence of a specific cynomolgus monkey GITR polypeptide is set forth in SEQ ID NO:42.
  • anti-GITR antibody refers to an antibody capable of binding (human or cynomolgus) GITR with sufficient affinity.
  • the protein or fragment thereof such that the antibody can be used as a diagnostic and/or therapeutic agent in a targeted (human or cynomolgus) GITR.
  • the anti-GITR antibody binds to a non- (human or cynomolgus) GITR protein to a lesser extent than about 10%, about 20%, about 30% of the antibody binds to (human or cynomolgus) GITR.
  • RIA radioimmunoassay
  • Antibody fragment refers to a molecule that is distinct from an intact antibody that comprises a portion of an intact antibody and binds to an antigen to which the intact antibody binds.
  • antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single chain antibodies (eg, scFv); single domain antibodies; Specific antibodies or fragments thereof; camelid antibodies; and bispecific antibodies or multispecific antibodies formed from antibody fragments.
  • epitope refers to a portion of an antigen (eg, GITR) that specifically interacts with an antibody molecule.
  • an antigen eg, GITR
  • An antibody that "binds to the same or overlapping epitope" as a reference antibody refers to an antibody that blocks more than 50% of the binding of the reference antibody to its antigen in a competition assay, in other words, the reference antibody is blocked in the competition assay. More than 50% of the antibody binds to its antigen.
  • An antibody that competes with a reference antibody for binding to its antigen refers to an antibody that blocks 50%, 60%, 70%, 80%, 90% or more of the binding of the reference antibody to its antigen in a competition assay.
  • the reference antibody blocks 50%, 60%, 70%, 80%, 90% or more of the binding of the antibody to its antigen in a competition assay.
  • Numerous types of competitive binding assays can be used to determine whether an antibody competes with another assay such as solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition. Assay (see for example Stahli et al, 1983, Methods in Enzymology 9: 242-253).
  • An antibody that inhibits (eg, competitively inhibits) binding of a reference antibody to its antigen refers to an antibody that inhibits binding of 50%, 60%, 70%, 80%, 90%, or 95% of the reference antibody to its antigen. . Conversely, the reference antibody inhibits binding of the antibody to its antigen by 50%, 60%, 70%, 80%, 90% or more.
  • the binding of an antibody to its antigen can be measured by affinity (eg, equilibrium dissociation constant). Methods for determining affinity are known in the art.
  • An antibody that exhibits the same or similar binding affinity and/or specificity as a reference antibody refers to an antibody that is capable of binding at least 50%, 60%, 70%, 80%, 90% or 95% of the reference antibody. Affinity and / or specificity. This can be determined by any method known in the art for determining binding affinity and/or specificity.
  • a “complementarity determining region” or “CDR region” or “CDR” is a sequence that is hypervariable in an antibody variable domain and that forms a structurally defined loop ("hypervariable loop") and/or contains an antigen contact residue ( The area of the "antigen contact point”).
  • the CDR is primarily responsible for binding to an epitope.
  • the CDRs of the heavy and light chains are commonly referred to as CDR1, CDR2 and CDR3, numbered sequentially from the N-terminus.
  • the CDRs located within the antibody heavy chain variable domain are referred to as HCDR1, HCDR2 and HCDR3, while the CDRs located within the antibody light chain variable domain are referred to as LCDR1, LCDR2 and LCDR3.
  • each CDR can be determined using any one or combination of a number of well-known antibody CDR assignment systems, including For example: Chothia based on the three-dimensional structure of the antibody and the topology of the CDR loop (Chothia et al.
  • the residues of each CDR are as follows.
  • the CDRs can also be determined based on having the same Kabat numbering position as the reference CDR sequence (e.g., any of the exemplary CDRs of the invention).
  • the CDRs of the antibodies of the invention are bordered by AbM rules, for example as shown in Table 1.
  • the boundaries of the CDRs of the variable regions of the same antibody obtained based on different assignment systems may vary. That is, the CDR sequences of the same antibody variable region defined under different assignment systems are different.
  • the scope of the antibody also encompasses an antibody whose variable region sequence comprises the particular CDR sequence, but due to the application of a different protocol (eg Different assignment system rules or combinations result in different claimed CDR boundaries than the specific CDR boundaries defined by the present invention.
  • Antibodies with different specificities have different CDRs.
  • the CDRs differ between antibodies and antibodies, only a limited number of amino acid positions within the CDRs are directly involved in antigen binding.
  • the minimum binding unit can be a sub-portion of the CDR.
  • residues of the remainder of the CDR sequences can be determined by the structure of the antibody and protein folding. Accordingly, the invention also contemplates variants of any of the CDRs presented herein. For example, in a variant of a CDR, the amino acid residues of the smallest binding unit may remain unchanged, while the remaining CDR residues defined by Kabat or Chothia may be replaced by conservative amino acid residues.
  • PD-1 axis binding antagonist refers to a molecule that inhibits the interaction of a PD-1 axis binding partner with one or more of its binding partners, thereby removing signals originating from the PD-1 signaling axis. Conducted T cell dysfunction - one result is the restoration or enhancement of T cell function (eg proliferation, cytokine production, target cell killing).
  • PD-1 axis binding antagonists include PD-1 binding antagonists (eg, anti-PD-1 antibodies), PD-L1 binding antagonists (eg, anti-PD-L1 antibodies), and PD-L2 binding antagonists. (eg anti-PD-L2 antibody).
  • PD-1 binding antagonist refers to a molecule that reduces, blocks, inhibits, eliminates or interferes with the binding of PD-1 to one or more of its binding partners (such as PD-L1, PD-L2). Interacting signal transduction.
  • the PD-1 binding antagonist is a molecule that inhibits PD-1 binding to one or more of its binding partners.
  • the PD-1 binding antagonist inhibits PD-1 binding to PD-L1 and/or PD-L2.
  • a PD-1 binding antagonist includes an anti-PD-1 antibody that reduces, blocks, inhibits, abolishes or interferes with signal transduction derived from PD-1 interaction with PD-L1 and/or PD-L2, antigen binding Fragments, immunoadhesins, fusion proteins, oligopeptides and other molecules.
  • the PD-1 binding antagonist reduces a cell surface protein-mediated negative costimulatory signal (via PD-1 mediated signaling) expressed on or via T lymphocytes, thereby enabling dysfunctional T cells Less dysfunctional (eg, enhancing effector response to antigen recognition).
  • the PD-1 binding antagonist is an anti-PD-1 antibody.
  • the PD-1 binding antagonist is MDX-1106 (nivolumab), MK-3475 (pembrolizumab), CT-011 (pidilizumab) or AMP-224 as disclosed in WO2015/095423.
  • the anti-PD-1 antibody is "Antibody C” (WO 2017/133540).
  • PD-L1 binding antagonist refers to a molecule that reduces, blocks, inhibits, eliminates or interferes with the binding of PD-L1 from one or more of its binding partners (such as PD-1, B7-1). Interacting signal transduction.
  • the PD-L1 binding antagonist is a molecule that inhibits PD-L1 binding to its binding partner.
  • the PD-L1 binding antagonist inhibits PD-L1 binding to PD-1 and/or B7-1.
  • the PD-L1 binding antagonist comprises reducing, blocking, inhibiting, eliminating or interfering with the interaction of PD-L1 from one or more of its binding partners (such as PD-1, B7-1) Signal transduction of anti-PD-L1 antibodies, antigen-binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules.
  • the PD-L1 binding antagonist reduces a cell surface protein-mediated negative costimulatory signal (via PD-L1 mediated signaling) expressed on or via T lymphocytes, thereby enabling dysfunctional T cells Less dysfunctional (eg, enhancing effector response to antigen recognition).
  • the PD-L1 binding antagonist is an anti-PD-L1 antibody.
  • the anti-PD-L1 antibody is YW243.55.S70, MDX-1105, MPDL3280A or MEDI4736 as disclosed in WO2015/095423.
  • PD-L2 binding antagonist refers to a molecule that reduces, blocks, inhibits, eliminates or interferes with signals derived from the interaction of PD-L2 with one or more of its binding partners, such as PD-1. divert.
  • the PD-L2 binding antagonist is a molecule that inhibits PD-L2 binding to one or more of its binding partners.
  • the PD-L2 binding antagonist inhibits PD-L2 binding to PD-1.
  • the PD-L2 antagonist comprises reducing, blocking, inhibiting, eliminating or interfering with signal transduction derived from the interaction of PD-L2 with one or more of its binding partners, such as PD-1.
  • Anti-PD-L2 antibodies, antigen-binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules are included in the PD-L2 binding antagonist.
  • the PD-L2 binding antagonist reduces a cell surface protein-mediated negative costimulatory signal (via PD-L2 mediated signaling) expressed on or via T lymphocytes, thereby enabling dysfunctional T cells Less dysfunctional (eg, enhancing effector response to antigen recognition).
  • the PD-L2 binding antagonist is an immunoadhesin.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcR Fc receptor
  • cytotoxic cells eg, NK cells, neutrophils, and macrophages.
  • Secretory immunoglobulins enable these cytotoxic effector cells to specifically bind to target cells carrying the antigen, followed by cytotoxicity to kill the cytotoxic form of the target cells.
  • the primary cell NK cells that mediate ADCC express only FcyRIII, whereas monocytes express FcyRI, FcyRII and FcyRIII.
  • Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-92 (1991) summarizes FcR expression on hematopoietic cells.
  • an in vitro ADCC assay can be performed, such as that described in U.S. Patent No. 5,500,362 or 5,821,337 or U.S. Patent No. 6,737,056 (Presta). Effector cells that can be used in such assays include PBMC and NK cells. Alternatively, or additionally, the ADCC activity of the molecule of interest can be assessed in vivo, for example, in animal models such as those disclosed in Clynes et al, PNAS (USA) 95:652-656 (1998). An exemplary assay for assessing ADCC activity is provided in the Examples herein.
  • cytotoxic agent refers to a substance which inhibits or prevents cell function and/or causes cell death or destruction.
  • “Chemotherapeutic agents” include chemical compounds that are useful in the treatment of cancer.
  • chemotherapeutic agents see those disclosed in WO 2015/031667, including but not limited to antineoplastic agents, including alkylating agents; antimetabolites; natural products; antibiotics; enzymes; heterogeneous agents; hormones and antagonists; ; antiandrogens; and non-steroidal antiandrogens.
  • small molecule drug refers to a low molecular weight organic compound capable of modulating a biological process.
  • a “functional Fc region” possesses an “effector function” of the native sequence Fc region.
  • effector functions include C1q binding; CDC; Fc receptor binding; ADCC; phagocytosis; downregulation of cell surface receptors (eg, B cell receptor; BCR), and the like.
  • Such effector functions generally require that the Fc region be associated with a binding domain (eg, an antibody variable domain) and can be assessed using a variety of assays, such as those disclosed herein.
  • Fc region is used herein to define a C-terminal region of an immunoglobulin heavy chain that comprises at least a portion of a constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • the human IgG heavy chain Fc region extends from Cys226 or Pro230 to the carbonyl terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • the numbering of amino acid residues in the Fc region or constant region is based on the EU numbering system, which is also referred to as the EU index, as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National. Institutes of Health, Bethesda, MD, 1991.
  • Antibody in IgG form refers to the IgG form to which the heavy chain constant region of an antibody belongs.
  • the heavy chain constant regions of all antibodies of the same type are identical, and the heavy chain constant regions differ between different types of antibodies.
  • an antibody in the IgGl form refers to an Ig domain whose heavy chain constant region Ig domain is IgG1.
  • therapeutic agent encompasses any substance that is effective in preventing or treating a tumor (eg, cancer) and an infection (eg, a chronic infection), including chemotherapeutic agents, cytotoxic agents, vaccines, other antibodies, anti-infective active agents, Small molecule drugs or immunomodulators.
  • an effective amount refers to an amount or dose of an antibody or fragment or conjugate or composition of the invention that, when administered to a patient in single or multiple doses, produces the desired effect in a patient in need of treatment or prevention.
  • An effective amount can be readily determined by the attending physician as a person skilled in the art by considering various factors such as the species of the mammal; its size, age and general health; the particular disease involved; the extent or severity of the disease; Response of an individual patient; specific antibody administered; mode of administration; bioavailability characteristics of the administered formulation; selected dosing regimen; and use of any concomitant therapy.
  • Therapeutically effective amount means an amount effective to achieve the desired therapeutic result at the desired dosage and for the period of time required.
  • the therapeutically effective amount of an antibody or antibody fragment or conjugate or composition thereof can vary depending on a variety of factors, such as the disease state, the age, sex and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody fragment or conjugate or composition thereof are less than a therapeutically beneficial effect.
  • a "therapeutically effective amount” preferably inhibits measurable parameters (eg, tumor growth rate, tumor volume, etc.) of at least about 20%, more preferably at least about 40%, even more preferably at least about 50%, relative to an untreated subject, 60% or 70% and still more preferably at least about 80% or 90%.
  • measurable parameters eg, tumor growth rate, tumor volume, etc.
  • the ability of a compound to inhibit measurable parameters can be evaluated in an animal model system that predicts efficacy in human tumors.
  • prophylactically effective amount is meant an amount effective to achieve the desired prophylactic result at the desired dosage and for the period of time required. Generally, a prophylactically effective amount will be less than a therapeutically effective amount since the prophylactic dose is administered to a subject prior to the earlier stage of the disease or at an earlier stage of the disease.
  • host cell refers to a cell into which an exogenous nucleic acid is introduced, including progeny of such a cell.
  • Host cells include "transformants" and "transformed cells" which include primary transformed cells and progeny derived therefrom, regardless of the number of passages.
  • Progeny may not be identical in nucleic acid content to the parental cell, but may contain mutations. Mutant progeny of the same function or biological activity selected or selected in the originally transformed cells are included herein.
  • Human antibody refers to an antibody having an amino acid sequence corresponding to the amino acid sequence of an antibody produced by a human or human cell or derived from a non-human source, which utilizes a human antibody library or other human Antibody coding sequence. This definition of a human antibody specifically excludes a humanized antibody comprising a non-human antigen-binding residue.
  • a “humanized” antibody refers to a chimeric antibody comprising an amino acid residue from a non-human CDR and an amino acid residue from a human FR.
  • a humanized antibody will comprise substantially all of at least one, typically two variable domains, wherein all or substantially all of the CDRs (eg, CDRs) correspond to those of a non-human antibody, and all Or substantially all of the FRs correspond to those of human antibodies.
  • the humanized antibody optionally can comprise at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized form" of an antibody (eg, a non-human antibody) refers to an antibody that has been humanized.
  • cancer and “cancerous” refer to or describe a physiological condition in a mammal that is typically characterized by unregulated cell growth.
  • tumor refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre
  • infectious disease refers to a disease caused by a pathogen, including, for example, a viral infection, a bacterial infection, a fungal infection, or a protozoan such as a parasitic infection.
  • chronic infection refers to an infection in which an infectious agent (eg, a pathogen such as a virus, a bacterium, a protozoan such as a parasite, a fungus, or the like) has induced an immune response in an infected host, but has not yet been as acutely infected. It is also removed or eliminated from the host as in the process.
  • an infectious agent eg, a pathogen such as a virus, a bacterium, a protozoan such as a parasite, a fungus, or the like
  • Chronic infections can be persistent, latent or slow.
  • acute infections such as influenza
  • persistent infections can persist for months, years, decades, or lifetime (eg, hepatitis B) at relatively low levels.
  • latent infections are characterized by long-term asymptomatic activity, interrupted by high levels of rapid infection and elevated pathogen levels over time (eg, herpes simplex).
  • slow infection is characterized by a gradual and continuous increase in disease symptoms, such as a long-term incubation period, followed by an onset of prolonged and progressive clinical processes following the onset of clinical symptoms.
  • an “immunoconjugate” is an antibody that is conjugated to one or more other substances, including but not limited to cytotoxic agents or labels.
  • label refers to a compound or composition that is directly or indirectly conjugated or fused to an agent, such as a polynucleotide probe or antibody, and that facilitates detection of the agent to which it is conjugated or fused.
  • the label itself may be detectable (eg, a radioisotope label or a fluorescent label) or, in the case of an enzymatic label, may catalyze a chemical change in a substrate compound or composition that is detectable.
  • the term is intended to encompass the direct labeling of a probe or antibody by coupling (ie, physically linking) a detectable substance to a probe or antibody and indirectly labeling the probe or antibody by reaction with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end labeling of a biotinylated DNA probe such that it can be detected with a fluorescently labeled streptavidin protein.
  • mammals include, but are not limited to, domesticated animals (eg, cows, sheep, cats, dogs, and horses), primates (eg, humans and non-human primates such as monkeys), rabbits, and rodents (eg, , mice and rats).
  • domesticated animals eg, cows, sheep, cats, dogs, and horses
  • primates eg, humans and non-human primates such as monkeys
  • rabbits eg, mice and rats
  • rodents eg, mice and rats.
  • the individual or subject is a human.
  • an “isolated” antibody is one which has been separated from components of its natural environment.
  • the antibody is purified to greater than 95% or 99% purity, such as by, for example, electrophoresis (eg, SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (eg, ion exchange or reversed phase) Determined by HPLC).
  • electrophoresis eg, SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatography eg, ion exchange or reversed phase
  • isolated nucleic acid encoding an anti-GITR antibody or fragment thereof refers to one or more nucleic acid molecules encoding an antibody heavy or light chain (or a fragment thereof), including such nucleic acid molecules in a single vector or separate vectors. And such nucleic acid molecules present at one or more positions in the host cell.
  • the sequences are aligned for optimal comparison purposes (eg, for optimal alignment, in the first and second amino acid sequences or nucleic acid sequences) Vacancies are introduced in one or both or non-homologous sequences can be discarded for comparison purposes.
  • the length of the aligned reference sequences is at least 30%, preferably at least 40%, more preferably at least 50%, 60% and even more preferably at least 70%, 80% for comparison purposes. , 90%, 100% of the reference sequence length.
  • the amino acid residues or nucleotides at the corresponding amino acid position or nucleotide position are then compared. When the position in the first sequence is occupied by the same amino acid residue or nucleotide at the corresponding position in the second sequence, then the molecule is identical at this position.
  • Mathematical algorithms can be used to achieve sequence comparisons and percent identity calculations between two sequences.
  • the Needlema and Wunsch ((1970) J. Mol. Biol. 48: 444-453) algorithm in the GAP program that has been integrated into the GCG software package is used (at http://www.gcg.com) Obtained), using a Blossum 62 matrix or PAM250 matrix and vacancy weights 16, 14, 12, 10, 8, 6 or 4 and length weights 1, 2, 3, 4, 5 or 6, to determine between the two amino acid sequences Percent identity.
  • the GAP program in the GCG software package (available at http://www.gcg.com) is used, using the NWSgapdna.CMP matrix and the vacancy weights of 40, 50, 60, 70 or 80 and The length weights 1, 2, 3, 4, 5 or 6, determine the percent identity between the two nucleotide sequences.
  • a particularly preferred set of parameters (and a set of parameters that should be used unless otherwise stated) is a Blossum 62 scoring matrix with a gap penalty of 12, a gap extension penalty of 4, and a frameshift gap penalty of 5.
  • nucleic acid sequences and protein sequences described herein can be further used as "query sequences" to perform searches against public databases to, for example, identify other family member sequences or related sequences.
  • pharmaceutically acceptable adjuvant refers to a diluent, adjuvant (eg, Freund's adjuvant (complete and incomplete)), excipient, carrier or stabilizer, and the like, which are administered with the active substance.
  • composition refers to a composition that is present in a form that permits the biological activity of the active ingredient contained therein to be effective, and does not include additional toxicity to the subject to whom the composition is administered. Ingredients.
  • combination product refers to a kit of fixed or non-fixed combinations in the form of a dosage unit or a portion for administration in combination, wherein two or more therapeutic agents can be independently at the same time or within a time interval Separate administration, especially when these time intervals allow the combined partner to exhibit collaboration, for example, synergistic effects.
  • fixed combination refers to the simultaneous administration of an antibody of the invention and a combination partner (eg, other therapeutic agents, eg, an anti-PD1 antibody, an anti-PDL1 antibody, or an anti-PDL2 antibody) to a patient in a single entity or dosage form.
  • non-fixed combination means that the antibody of the invention and a combination partner (eg, other therapeutic agents, eg, an anti-PD1 antibody, an anti-PDL1 antibody, or an anti-PDL2 antibody) are administered to the patient simultaneously, in parallel, or sequentially as separate entities, without a specific time. Limitations, wherein such administration provides a therapeutically effective level of both compounds in a patient. The latter also applies to cocktail therapy, for example the administration of three or more therapeutic agents.
  • the pharmaceutical combination is a non-fixed combination.
  • combination therapy refers to the administration of two or more therapeutic agents to treat a cancer or infection as described in the present disclosure.
  • administration involves co-administering these therapeutic agents in a substantially simultaneous manner, for example, as a single capsule having a fixed ratio of active ingredient.
  • administration includes co-administration of the individual active ingredients in multiple or separate containers (e.g., tablets, capsules, powders, and liquids).
  • the powder and/or liquid can be reconstituted or diluted to the desired dose prior to administration.
  • such administration also includes the use of each type of therapeutic agent in a sequential manner at substantially the same time or at different times. In either case, the treatment regimen will provide a beneficial effect of the combination of drugs in treating the condition or condition described herein.
  • treating refers to slowing, interrupting, arresting, ameliorating, stopping, reducing, or reversing the progression or severity of an existing symptom, disorder, condition, or disease.
  • prevention includes inhibition of the occurrence or progression of a disease or condition or a symptom of a particular disease or condition.
  • a subject with a family history of cancer is a candidate for a prophylactic regimen.
  • prevention refers to the administration of a drug prior to the onset of a symptom or symptom of a cancer, particularly in a subject at risk for cancer.
  • anti-infective active agent includes any molecule that specifically inhibits or eliminates the growth of microorganisms at the concentration of administration and the interval of administration, but is not lethal to the host, such as viruses, bacteria, fungi or protozoa, such as parasites.
  • anti-infective active agent includes antibiotics, antibacterial agents, antiviral agents, antifungal agents, and antiprotozoal agents.
  • the anti-infective active agent is non-toxic to the host at the concentration of administration and the interval of administration.
  • Antibacterial anti-infective active agents or antibacterial agents can be broadly classified as either bactericidal (i.e., directly killed) or bacteriostatic (i.e., preventing division). Antibacterial anti-infective active agents can be further reclassified as narrow-spectrum antibacterial agents (i.e., affecting only subtypes of bacterial subtypes, e.g., Gram-negative, etc.) or broad-spectrum antibacterial agents (i.e., affecting a wide variety of species).
  • antiviral agent includes any substance that inhibits or eliminates the growth, pathogenesis, and/or survival of a virus.
  • antifungal agent includes any substance that inhibits or eliminates the growth, pathogenesis and/or survival of fungi.
  • antigenic animal agent includes any substance that inhibits or eliminates the growth, morbidity and/or survival of a protozoan organism (eg, a parasite).
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes a vector that is a self-replicating nucleic acid structure and a vector that binds to the genome of a host cell into which it has been introduced. Some vectors are capable of directing the expression of a nucleic acid to which they are operably linked. Such vectors are referred to herein as "expression vectors.”
  • Subject/patient sample refers to a collection of cells or fluids obtained from a patient or subject.
  • the source of the tissue or cell sample may be a solid tissue, such as from a fresh, frozen and/or preserved organ or tissue sample or a biopsy sample or a puncture sample; blood or any blood component; body fluids such as cerebrospinal fluid, amniotic fluid (amniotic fluid) ), peritoneal fluid (ascites), or interstitial fluid; cells from the subject's pregnancy or development at any time.
  • Tissue samples may contain compounds that are naturally not intermixed with tissue in nature, such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, and the like.
  • tumor samples include, but are not limited to, tumor biopsy, fine needle aspirate, bronchial lavage fluid, pleural fluid (thoracic fluid), sputum, urine, surgical specimens, circulating tumor cells, serum, plasma, circulation Plasma protein, ascites, primary cell cultures or cell lines derived from tumors or exhibiting tumor-like properties, and preserved tumor samples, such as formalin-fixed, paraffin-embedded tumor samples or frozen tumors sample.
  • the anti-GITR antibody or fragment of the invention binds with high affinity GITR (GITR e.g. human or cynomolgus GITR), for example, in the following equilibrium dissociation constant (K D) in combination with GITR, the K D is less than about 10 nM, preferably, less than or equal to about 7 nM, more preferably less than or equal to about 6 nM, more preferably less than or equal to about 5 nM, 4.9 nM, 4.8 nM, 4.7 nM, 4.6 nM, 4.5 nM, 4 nM, or of 3 nM, and most preferably, the K D of less than or equal to about 2.5nM, 2.4nM, 2.3nM, 2.2nM, 2.1nM.
  • K D equilibrium dissociation constant
  • the anti-GITR antibody of the present invention to 1nM-6nM, preferably 1.5nM-5nM, 1.7nM-5nM, 1.8nM-5nM, K D 1.9nM-5nM GITR binding.
  • the GITR is a human GITR.
  • the antibody binding affinity is determined using a bio-optical interference assay (eg, Fortebio affinity measurement).
  • an antibody or fragment thereof of the invention binds to a cell expressing GITR, eg, at less than or equal to about 2 nM, 1.9 nM, 1.5 nM, 1 nM, 0.9 nM, 0.8 nM, 0.7 nM, 0.6 nM, 0.5 nM. , EC50 of 0.4nM.
  • the binding is determined by flow cytometry (eg, FACS).
  • the GITR expressing cell is a CHO cell (eg, CHO-S cell) that expresses GITR.
  • the GITR is a human GITR or a cynomolgus monkey GITR.
  • an antibody or fragment thereof of the invention activates the NF-kappaB signaling pathway downstream of the GITR by binding to a GITR molecule on the cell surface.
  • an antibody of the invention or a fragment thereof has a higher ability to activate than a known anti-GITR antibody, such as the GITR antibody reported in patent application US20130183321A1 (GITR, INC. (Cambridge, MA, US)).
  • the light and heavy chain sequences of known anti-GITR antibody molecules are SEQ ID NO: 44 and SEQ ID NO: 54, respectively, in US20130183321A1.
  • a GITR antibody molecule of the invention is capable of significantly and efficiently activating the NF kappa B signaling pathway compared to a control antibody molecule.
  • the activation is obtained by luciferase assay.
  • an antibody of the invention or a fragment thereof enhances effector T cell function, such as activating effector T cells.
  • an antibody or fragment thereof of the invention is capable of enhancing proliferation of effector T cells.
  • an antibody or fragment thereof of the invention increases the secretion/expression of interferon (eg, IFN-[gamma]).
  • an antibody or fragment thereof of the invention increases secretion/expression of interleukin (eg, IL-2).
  • the T cell is a CD4 T cell.
  • a GITR antibody or fragment thereof of the invention is capable of activating a GITR signaling pathway in a regulatory T cell (eg, a Treg cell), thereby eliminating cells by mediating the ADCC effect.
  • a regulatory T cell eg, a Treg cell
  • the inhibitory signal of the effector T cell itself is released, the activity of the CD4 T cell and/or the CD8 T cell is enhanced, and the other aspect is to eliminate the inhibitory effect of the T cell on the effector T cell, thereby maximally activating the effector T cell and enhancing An immune response to tumor cells.
  • the ADCC activity of the antibody is detected by detecting NF-AT signaling activation.
  • activation of the NF-AT signal is manifested by expression of a fluorescent reporter gene.
  • the GITR antibody molecules of the invention are capable of significantly and efficiently activating the NF-AT signaling pathway compared to known anti-GITR antibody molecules.
  • the GITR antibody molecules of the invention are capable of significantly and efficiently mediating ADCC effects compared to known anti-GITR antibody molecules.
  • a GITR antibody molecule of the invention is capable of significantly inhibiting or eliminating regulatory T cells compared to known anti-GITR antibody molecules.
  • the effector T cells of the invention are CD4 T cells.
  • a known anti-GITR antibody molecule is an anti-GITR antibody as reported in, for example, patent application US20130183321A1 (GITR, INC. (Cambridge, MA, US)).
  • the light and heavy chain sequences of known anti-GITR antibody molecules are SEQ ID NO: 44 and SEQ ID NO: 54, respectively, in US20130183321A1.
  • an antibody or fragment thereof of the invention (optionally in combination with a therapeutic modality and/or other therapeutic agent, such as a PD-1 axis binding antagonist) is capable of preventing or treating a tumor.
  • an antibody or fragment thereof of the invention can be used to inhibit or reduce tumor growth (eg, reduce tumor volume).
  • an antibody or fragment thereof of the invention can also be used to maintain body weight in a tumor patient.
  • an antibody or fragment thereof of the invention is capable of inhibiting or reducing tumor growth (eg, reducing tumor volume) in combination with a therapeutic modality and/or other therapeutic agent, such as a PD1-axis binding antagonist.
  • an antibody or fragment thereof of the invention is capable of maintaining the weight of a tumor patient in combination with a therapeutic modality and/or other therapeutic agent, such as a PD1-axis binding antagonist.
  • a therapeutic modality and/or other therapeutic agent such as a PD1-axis binding antagonist.
  • an antibody of the invention or a fragment thereof is capable of inhibiting or reducing tumor growth (eg, reducing tumor volume) ), while not affecting the weight of cancer patients.
  • the tumor is a gastrointestinal tumor.
  • the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like.
  • an antibody of the invention or a fragment thereof is capable of preventing or treating an infection, such as a chronic infection, including a bacterial infection, a fungal infection, a viral infection, a protozoal infection. Wait.
  • an infection such as a chronic infection, including a bacterial infection, a fungal infection, a viral infection, a protozoal infection. Wait.
  • an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region (VH), wherein said VH comprises
  • an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a light chain variable region (VL), wherein the VL comprises:
  • an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region VH and a light chain variable region VL, wherein
  • VL contains:
  • the VH comprises or consists of an amino acid sequence selected from the group consisting of SEQ ID NO: 17, 18, 19 or 20.
  • VL comprises or consists of an amino acid sequence selected from the group consisting of SEQ ID NO: 21, 22, 23 or 24.
  • the anti-GITR antibody or antigen-binding fragment thereof of the invention comprises
  • an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region (VH) and/or a light chain variable region (VL), wherein
  • said VH comprises a complementarity determining region (CDR) HCDR1, HCDR2 and HCDR3, wherein HCDR1 comprises or consists of the amino acid sequence selected from SEQ ID NO: 1, 2, 3 or 4, or HCDR1 comprises An amino acid sequence having one, two or three alterations (preferably amino acid substitutions, preferably conservative substitutions) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 1, 2, 3 or 4; HCDR2 comprising an SEQ ID NO: 5 Or the amino acid sequence of 6 or consists of the amino acid sequence, or HCDR2 comprises one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) compared to the amino acid sequence selected from SEQ ID NO: 5 or 6.
  • CDR complementarity determining region
  • HCDR3 comprises or consists of the amino acid sequence selected from SEQ ID NO: 7 or 8, or HCDR3 comprises one, two or more than the amino acid sequence selected from SEQ ID NO: 7 or 8.
  • An amino acid sequence of three alterations preferably amino acid substitutions, preferably conservative substitutions;
  • VL comprises a complementarity determining region (CDR) LCDR1, LCDR2 and LCDR3, wherein LCDR1 comprises or consists of the amino acid sequence selected from SEQ ID NO: 9 or 10, or LCDR1 comprises and is selected from the group consisting of ID NO: amino acid sequence of 9 or 10 having an amino acid sequence of one, two or three changes (preferably amino acid substitution, preferably conservative substitution); LCDR2 comprising an amino acid selected from the group consisting of SEQ ID NO: 11, 12, 13 or 14. The sequence consists either of said amino acid sequence, or LCDR2 comprises one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) compared to an amino acid sequence selected from SEQ ID NO: 11, 12, 13 or 14.
  • CDR complementarity determining region
  • LCDR3 comprises or consists of the amino acid sequence selected from SEQ ID NO: 15 or 16, or LCDR3 comprises one, two or three compared to the amino acid sequence selected from SEQ ID NO: 15 or 16. Amino acid sequence that changes (preferably amino acid substitutions, preferably conservative substitutions).
  • the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain variable region (VH) and/or a light chain variable region (VL), wherein
  • the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain variable region (VH) and/or a light chain variable region (VL), wherein said VH comprises a complementarity determining region ( CDR) HCDR1, HCDR2 and HCDR3 comprising (CDR) LCDR1, LCDR2 and LCDR3, wherein the combination of HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3 comprised by the antibody or antigen-binding fragment thereof is as follows (Table A):
  • Table A Exemplary combinations of HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 in an antibody or antigen-binding fragment thereof of the present invention
  • an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein
  • amino acid sequence of an amino acid change (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, said amino acid change does not occur in the CDR region;
  • amino acid sequence of an amino acid change (preferably an amino acid substitution, more preferably an amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR regions.
  • the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain variable region (VH) and a light chain variable region (VL), wherein the antibody or antigen-binding fragment thereof comprises
  • VH heavy chain variable region
  • VL light chain variable region
  • Table B Exemplary combinations of heavy chain variable region VH and light chain variable region VL in an antibody or antigen-binding fragment thereof of the invention
  • an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain and/or a light chain, wherein
  • amino acid sequence of amino acid change (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR region of the heavy chain, and more preferably, the amino acid change does not occur in Heavy chain variable region;
  • amino acid change comprising one or more (preferably no more than 20 or 10, more preferably no more than 5, 4, 3, 2) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 29, 30, 31 or 32
  • Amino acid sequence of amino acid change preferably amino acid substitution, more preferably amino acid conservative substitution
  • the amino acid change does not occur in the CDR region of the light chain, and more preferably, the amino acid change does not occur in Light chain variable region.
  • the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain and a light chain, wherein the combination of the heavy chain and the light chain comprised by the antibody or antigen-binding fragment thereof is as follows (Table C):
  • Table C Exemplary combinations of heavy and light chains in an antibody or antigen-binding fragment thereof of the invention
  • the heavy and/or light chain of an anti-GITR antibody or fragment thereof of the invention further comprises a signal peptide sequence, such as METDTLLLWVLLLWVPGSTG (SEQ ID NO: 43).
  • the amino acid changes described herein include substitutions, insertions or deletions of amino acids.
  • the amino acid changes described herein are amino acid substitutions, preferably conservative substitutions.
  • the amino acid changes described herein occur in regions outside the CDRs (e.g., in FR). More preferably, the amino acid changes described herein occur in regions outside the heavy chain variable region and/or outside the light chain variable region.
  • the substitution is a conservative substitution.
  • a conservative substitution is one in which one amino acid is replaced by another amino acid in the same class, for example, one acidic amino acid is replaced by another acidic amino acid, one basic amino acid is replaced by another basic amino acid, or one neutral amino acid is passed through another neutral amino acid.
  • Replacement An exemplary substitution is shown in Table D below:
  • the substitution occurs in the CDR regions of the antibody.
  • the variant obtained has modifications (e. g., improved) relative to the parent antibody in certain biological properties (e.g., increased affinity) and/or will have certain biological properties that are substantially retained by the parent antibody.
  • An exemplary substitution variant is an affinity matured antibody.
  • the antibodies provided herein are altered to increase or decrease the extent of glycosylation of the antibody.
  • Addition or deletion of a glycosylation site to an antibody can be conveniently accomplished by altering the amino acid sequence to create or remove one or more glycosylation sites.
  • the antibody comprises an Fc region
  • the saccharide attached thereto can be altered.
  • modifications that remove unwanted glycosylation sites may be useful, such as removal of fucose modules to enhance antibody-dependent cellular cytotoxicity (ADCC) function (see Shield et al. (2002) JBC 277:26733 ).
  • ADCC antibody-dependent cellular cytotoxicity
  • galactosidation modification can be performed to modify complement dependent cytotoxicity (CDC).
  • one or more amino acid modifications can be introduced into the Fc region of an antibody provided herein to produce an Fc region variant to enhance the effectiveness of, for example, an antibody in treating cancer or a cell proliferative disorder.
  • An Fc region variant can include a human Fc region sequence (eg, a human IgGl, IgG2, IgG3, or IgG4 Fc region) comprising an amino acid modification (eg, a substitution) at one or more amino acid positions.
  • a human Fc region sequence eg, a human IgGl, IgG2, IgG3, or IgG4 Fc region
  • amino acid modification eg, a substitution
  • cysteine engineered antibody such as a "thiocarba" wherein one or more residues of the antibody are replaced with a cysteine residue.
  • Cysteine engineered antibodies can be generated as described, for example, in U.S. Patent No. 7,521,541.
  • the antibodies provided herein can be further modified to contain other non-protein portions known in the art and readily available.
  • Portions suitable for antibody derivatization include, but are not limited to, water soluble polymers.
  • water soluble polymers include, but are not limited to, polyethylene glycol (PEG), ethylene glycol/propylene glycol copolymers, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly -1,3-dioxane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyamino acid (homopolymer or random copolymer), and dextran or poly(n-ethylene) Pyrrolidone) polyethylene glycol, propylene glycol homopolymer, polypropylene oxide/ethylene oxide copolymer, polyoxyethylated polyol (such as glycerin), polyvinyl alcohol, and mixtures thereof.
  • PEG polyethylene glycol
  • an anti-GITR antibody or antigen-binding fragment thereof of the invention has one or more of the following characteristics:
  • an anti-GITR antibody of the invention is an antibody in the IgGl form or an antibody in the IgG2 form or an antibody in the IgG4 form.
  • the anti-GITR antibody is a monoclonal antibody.
  • the anti-GITR antibody is humanized. Different methods for humanizing antibodies are known to the skilled person, as reviewed by Almagro & Fransson, the contents of which are incorporated herein by reference in its entirety (Almagro JC and Fransson J (2008) Frontiers in Bioscience 13: 1619-1633).
  • the anti-GITR antibody is a human antibody.
  • Human antibodies can be prepared using a variety of techniques known in the art. Human antibodies are generally described in van Dijk and van de Winkel, Curr. Opin. Pharmacol 5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol 20: 450-459 (2008).
  • the anti-GITR antibody is a chimeric antibody.
  • an anti-GITR antibody of the invention further encompasses an antibody fragment thereof, preferably an antibody fragment selected from the group consisting of Fab, Fab', Fab'-SH, Fv, single chain antibody (eg, scFv) or (Fab' 2 ) Single domain antibody, double antibody (dAb) or linear antibody.
  • the anti-GITR antibody molecule is in the form of a bispecific or multispecific antibody molecule.
  • the bispecific antibody molecule has a first binding specificity for GITR and a second binding specificity for PD-1 or PD-L1 or PD-L2.
  • the bispecific antibody molecule binds to GITR and PD-1.
  • the bispecific antibody molecule binds to GITR and PD-L1.
  • the bispecific antibody molecule binds to GITR and PD-L2.
  • the multispecific antibody molecule can have any combination of binding specificities for the aforementioned molecules.
  • the multispecific antibody molecule can be, for example, a trispecific antibody molecule comprising a first binding specificity for GITR and a second and third binding specificity for a molecule of one or more of the following: PD-1, PD- L1 or PD-L2.
  • the invention provides a nucleic acid encoding any of the above anti-GITR antibodies or fragments thereof.
  • a vector comprising the nucleic acid is provided.
  • the vector is an expression vector.
  • a host cell comprising the nucleic acid or the vector is provided.
  • the host cell is eukaryotic.
  • the host cell is selected from the group consisting of a yeast cell, a mammalian cell (eg, a CHO cell or a 293 cell), or other cell suitable for use in the preparation of an antibody or antigen-binding fragment thereof.
  • the host cell is prokaryotic.
  • the invention provides a nucleic acid encoding any of the above anti-GITR antibodies or fragments thereof.
  • the nucleic acid may comprise a nucleic acid encoding an amino acid sequence of a light chain variable region and/or a heavy chain variable region of an antibody, or a nucleic acid comprising an amino acid sequence encoding a light chain and/or a heavy chain of an antibody.
  • An exemplary nucleic acid sequence encoding an antibody heavy chain variable region comprising at least 80%, 85%, 90%, 91%, 92%, 93% of a nucleic acid sequence selected from the group consisting of SEQ ID NO: 33, 34, 35 or 36 a nucleic acid sequence of 94%, 95%, 96%, 97%, 98% or 99% identity, or a nucleic acid sequence selected from the group consisting of SEQ ID NO: 33, 34, 35 or 36.
  • An exemplary nucleic acid sequence encoding an antibody light chain variable region comprises at least 80%, 85%, 90%, 91%, 92%, 93% of a nucleic acid sequence selected from the group consisting of SEQ ID NO: 37, 38, 39 or 40 A nucleic acid sequence of 94%, 95%, 96%, 97%, 98% or 99% identity, or a nucleic acid sequence selected from the group consisting of SEQ ID NO: 37, 38, 39 or 40.
  • one or more vectors comprising the nucleic acid are provided.
  • the vector is an expression vector, such as a eukaryotic expression vector.
  • Vectors include, but are not limited to, viruses, plasmids, cosmids, lambda phage, or yeast artificial chromosomes (YAC).
  • YAC yeast artificial chromosomes
  • the vector is a pTT5 vector.
  • a host cell comprising the vector.
  • Suitable host cells for cloning or expressing a vector encoding the antibody include prokaryotic or eukaryotic cells as described herein.
  • antibodies can be produced in bacteria, particularly when glycosylation and Fc effector functions are not required.
  • the antibody can be isolated from the bacterial cell paste in the soluble fraction and can be further purified.
  • the host cell is eukaryotic.
  • the host cell is selected from the group consisting of a yeast cell, a mammalian cell, or other cell suitable for use in the preparation of an antibody or antigen-binding fragment thereof.
  • a eukaryotic microorganism such as a filamentous fungus or yeast is a suitable cloning or expression host for a vector encoding an antibody.
  • fungal and yeast strains that have been "humanized" by the glycosylation pathway result in the production of antibodies with partial or complete human glycosylation patterns. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004), and Li et al, Nat. Biotech. 24: 210-215 (2006).
  • Host cells suitable for expression of glycosylated antibodies are also derived from multicellular organisms (invertebrates and vertebrates). Vertebrate cells can also be used as hosts.
  • mammalian cell lines engineered to be suitable for suspension growth can be used.
  • useful mammalian host cell lines are the monkey kidney CV1 line (COS-7) transformed with SV40; human embryonic kidney line (293 HEK or 293F or 293 cells such as, for example, Graham et al, J. Gen Virol. 36:59 ( (described in 1977) and so on.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR-CHO cells (Urlaub et al, Proc. Natl.
  • Tumor cell lines such as Y0, NSO and Sp2/0.
  • Tumor cell lines such as Y0, NSO and Sp2/0.
  • the invention provides a method of making an anti-GITR antibody or fragment thereof (preferably an antigen-binding fragment), wherein the method comprises suitably expressing an antibody or fragment thereof (preferably an antigen-binding fragment) encoding the antibody
  • the host cell is cultured under conditions of a nucleic acid, and the antibody or fragment thereof (preferably an antigen-binding fragment) is optionally isolated.
  • the method further comprises recovering an anti-GITR antibody or fragment thereof (preferably an antigen-binding fragment) from the host cell.
  • a method of making an anti-GITR antibody comprising culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, and optionally, under conditions suitable for expression of the antibody
  • the antibody is recovered from the host cell (or host cell culture medium).
  • a nucleic acid encoding an antibody (such as the antibodies described above) is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acids are readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of specifically binding to genes encoding the heavy and light chains of the antibody).
  • the anti-GITR antibodies provided herein can be identified, screened, or characterized for their physical/chemical properties and/or biological activity by a variety of assays known in the art.
  • the antibody of the present invention is tested for its antigen binding activity, for example, by known methods such as ELISA, Western blot, and the like. Binding to GITR can be determined using methods known in the art, and exemplary methods are disclosed herein. In some embodiments, biophotonic interferometry (eg, Fortebio affinity measurement) or MSD assay is used.
  • a competition assay can be used to identify antibodies that compete with any of the anti-GITR antibodies disclosed herein for binding to GITR.
  • a competitive antibody binds to an epitope (eg, a linear or conformational epitope) that is identical or overlapping with an epitope to which any of the anti-GITR antibodies disclosed herein bind.
  • an epitope eg, a linear or conformational epitope
  • a detailed exemplary method for locating epitopes bound by antibodies is found in Morris (1996) "Epitope Mapping Protocols", Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ).
  • the invention also provides assays for identifying biologically active anti-GITR antibodies.
  • Biological activity can include, for example, binding to GITR (eg, binding to human and/or cynomolgus GITR), increasing GITR-mediated signal transduction (eg, increasing NFkappa-B signaling pathway), and attenuating cells expressing GITR by ADCC (eg, Treg cells) Enhancing T effector cell function (eg, CD4 effector T cells) (eg, by increasing cytokine production of effector T cells (eg, interferons such as IFN-[gamma] or interleukins such as IL2)).
  • ADCC eg, Treg cells
  • Enhancing T effector cell function eg, CD4 effector T cells
  • effector T cells eg, interferons such as IFN-[gamma] or interleukins such as IL2
  • Antibodies having such biological activity in vivo and/or in vitro are also provided.
  • the antibodies of the invention are tested for such biological activity.
  • T cell activation can be assayed using methods known in the art. For example, it is determined by the level of a cytokine released after T cell activation, such as interferon (such as IFN- ⁇ ) or interleukin (such as IL2).
  • GITR signaling e.g., NF-kappaB signaling pathway
  • a transgenic cell expressing a human GITR and a reporter gene comprising a NF-kappa B promoter fused to a reporter gene (e.g., beta luciferase) is generated. Addition of anti-GITR antibodies to cells results in increased NF-kappa B transcription, which is detected using an assay for the reporter gene (eg, a luciferase reporter assay).
  • the ADCC effect of an antibody can be determined using methods known in the art. For example, activation by NF-AT signal.
  • a transgenic cell comprising an ADCC effector cell comprising an NF-AT promoter fused to a reporter gene (eg, beta luciferase) is obtained.
  • a reporter gene eg, beta luciferase
  • Co-culture of effector cells with cells that highly express GITR, while the addition of anti-GITR antibodies results in increased NF-AT transcription of effector cells, which is detected using assays for reporter genes (eg, luciferase reporter assay).
  • Cells for use in any of the above in vitro assays include naturally expressing GITR or engineered to express a GITR cell line. Such cells include T cells that naturally express GITR, Treg cells, and activated memory T cells. Such cells also include cell lines encoding GITR and GITR-transfected cells that are not normally expressing GITR.
  • any of the above assays can be performed by replacing or supplementing the anti-GITR antibody with an immunoconjugate of the invention.
  • any of the above assays can be performed using an anti-GITR antibody and another active agent.
  • the invention provides an immunoconjugate comprising any of the anti-GITR antibodies and other materials provided herein, such as a cytotoxic agent.
  • a cytotoxic agent such as a cytotoxic agent.
  • other substances such as therapeutic agents, such as cytotoxic or immunosuppressive agents or chemotherapeutic agents.
  • Cytotoxic agents include any agent that is harmful to cells. Examples of cytotoxic agents (e.g., chemotherapeutic agents) suitable for forming immunoconjugates are known in the art.
  • cytotoxic agents include, but are not limited to, radioisotopes; growth inhibitors; enzymes and fragments thereof such as nucleases; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including Fragments and/or variants; and various anti-tumor or anti-cancer agents known.
  • the immunoconjugate is for use in preventing or treating a tumor, such as a gastrointestinal tumor.
  • the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like.
  • the immunoconjugate is used to prevent or treat an infection, such as a chronic infection, such as a bacterial infection, a viral infection, a fungal infection, a protozoal infection, and the like.
  • the invention provides compositions comprising any of the anti-GITR antibodies or fragments thereof, preferably antigen-binding fragments thereof, or immunoconjugates thereof, preferably a composition, which is a pharmaceutical composition.
  • the composition further comprises a pharmaceutical excipient.
  • a composition eg, a pharmaceutical composition, comprises an anti-GITR antibody or fragment thereof of the invention, or an immunoconjugate thereof, and one or more additional therapeutic agents (eg, chemotherapeutic agents, cytotoxic agents, A combination of a vaccine, other antibody, anti-infective active agent, small molecule drug or immunomodulatory agent, preferably a PD-1 axis binding antagonist, such as an anti-PD-1 antibody, an anti-PD-L1 antibody or an anti-PD-L2 antibody.
  • additional therapeutic agents eg, chemotherapeutic agents, cytotoxic agents, A combination of a vaccine, other antibody, anti-infective active agent, small molecule drug or immunomodulatory agent, preferably a PD-1 axis binding antagonist, such as an anti-PD-1 antibody, an anti-PD-L1 antibody or an anti-PD-L2 antibody.
  • the composition is for use in preventing or treating a tumor, such as a gastrointestinal tumor.
  • the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like.
  • the composition is for preventing or treating an infection, such as a chronic infection, such as a bacterial infection, a viral infection, a fungal infection, a protozoal infection, and the like.
  • the present invention also encompasses a composition (including a pharmaceutical composition or a pharmaceutical preparation) comprising an anti-GITR antibody or an immunoconjugate thereof, and a composition (including a pharmaceutical composition or a pharmaceutical preparation) comprising a polynucleotide encoding an anti-GITR antibody.
  • the compositions comprise one or more antibodies or fragments thereof that bind to GITR or one or more polynucleotides that encode one or more antibodies or fragments thereof that bind to GITR.
  • These compositions may also contain suitable pharmaceutical excipients such as pharmaceutically acceptable carriers, pharmaceutically acceptable excipients, including buffering agents known in the art.
  • Pharmaceutically acceptable carriers suitable for use in the present invention may be sterile liquids such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be used as liquid carriers, especially for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, dried skim milk , glycerin, propylene, glycol, water, ethanol, etc.
  • compositions may also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • these compositions may take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained release formulations and the like.
  • Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, saccharin.
  • Preparation can be made by mixing an anti-GITR antibody of the invention having the desired purity with one or more optional pharmaceutical excipients (Remington's Pharmaceutical Sciences, 16th Ed., Osol, A. Ed. (1980)).
  • the pharmaceutical preparations of the anti-GITR antibodies described herein are preferably in the form of a lyophilized formulation or an aqueous solution.
  • Exemplary lyophilized antibody formulations are described in U.S. Patent No. 6,267,958.
  • Aqueous antibody preparations include those described in U.S. Patent No. 6,171,586 and WO2006/044908, the latter including a histidine-acetate buffer.
  • compositions or formulations of the present invention may further comprise one or more additional active ingredients which are required for the particular indication being treated, preferably those which do not adversely affect each other's complementary activities.
  • additional active ingredients such as chemotherapeutic agents, cytotoxic agents, vaccines, other antibodies, anti-infective active agents, small molecule drugs or immunomodulators such as PD-1 axis binding antagonists (eg, anti- PD-1 antibody or anti-PD-L1 antibody or anti-PD-L2 antibody) and the like.
  • PD-1 axis binding antagonists eg, anti- PD-1 antibody or anti-PD-L1 antibody or anti-PD-L2 antibody
  • the active ingredient is suitably present in combination in an amount effective for the intended use.
  • sustained release formulations can be prepared. Suitable examples of sustained release formulations include semipermeable matrices of solid hydrophobic polymers containing antibodies in the form of shaped articles such as films or microcapsules.
  • the invention also provides a combination product comprising an antibody of the invention, or an antigen binding fragment thereof, or an immunoconjugate thereof, and one or more additional therapeutic agents (eg, chemotherapeutic agents, other antibodies, Cytotoxic agents, vaccines, anti-infective active agents, small molecule drugs or immunomodulators, etc.).
  • additional therapeutic agents eg, chemotherapeutic agents, other antibodies, Cytotoxic agents, vaccines, anti-infective active agents, small molecule drugs or immunomodulators, etc.
  • other antibodies are, for example, anti-PD-1 antibodies or anti-PD-L1 antibodies or anti-PD-L2 antibodies.
  • the combination product is for use in preventing or treating a tumor, such as a gastrointestinal tumor.
  • the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like.
  • the combination product is for use in preventing or treating an infection, such as a chronic infection, such as a bacterial infection, a viral infection, a fungal infection, a protozoal infection, and the like.
  • the invention provides a method of activating a NF-KappaB signaling pathway in a subject, comprising administering to the subject an effective amount of an anti-GITR antibody of the invention, or an antigen-binding fragment thereof, an immunoconjugate, a pharmaceutical composition Or a combination of products.
  • a further aspect of the invention provides a method of increasing effector T cell function, such as activating effector T cells, in a subject, comprising administering to the subject an effective amount of an anti-GITR antibody or antigen-binding fragment thereof, immunologically conjugated to the subject Compound, pharmaceutical composition or combination product.
  • the function is an increase in effector T cell proliferation.
  • the activated T cell is characterized by increased secretion/expression of interferon or interleukin.
  • the interleukin is IL2.
  • the interferon is IFN-[gamma].
  • the present invention provides a method of mediating ADCC in a subject to eliminate regulatory T cells, comprising administering to the subject an effective amount of an anti-GITR antibody or antigen-binding fragment thereof, immunoconjugate of the present invention. , a pharmaceutical composition or a combination product.
  • the regulatory T cell is a Treg cell.
  • the invention provides a method of preventing or treating a tumor in a subject, the method comprising administering to the subject an effective amount of any of the anti-GITR antibodies or fragments thereof, immunoconjugates, Pharmaceutical composition or combination product.
  • the invention provides a method of preventing or treating an infection in a subject, the method comprising administering to the subject an effective amount of any of the anti-GITR antibodies or fragments thereof, immunoconjugates, Pharmaceutical composition or combination product.
  • the invention also provides a method of preventing or treating other conditions or conditions associated with Treg proliferation in a subject, the method comprising administering to the subject an effective amount of any of the anti-GITR antibodies or fragments thereof described herein , immunoconjugate, pharmaceutical composition or combination product.
  • the subject can be a mammal, for example, a primate, preferably a higher primate, for example, a human (eg, a patient having the disease described herein or at risk of having the disease described herein).
  • the subject has or is at risk of having a disease described herein (eg, a tumor or infectious disease as described herein).
  • the subject has received or has received other treatments, such as chemotherapy treatment and/or radiation therapy.
  • the subject is immunocompromised due to infection or has a risk of being immunocompromised by the infection.
  • the tumors described herein include, but are not limited to, solid tumors, hematological cancers, soft tissue tumors, and metastatic lesions.
  • the cancers described herein for treatment include but breast cancer, lung cancer, ovarian cancer, prostate cancer, colon cancer, rectal cancer, colorectal cancer, cervical cancer, brain cancer, skin cancer, liver cancer, pancreas Solid tumors such as cancer or stomach cancer, as well as blood cancers such as leukemia and lymphoma.
  • the cancer is a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like.
  • metastatic cancer e.g., metastatic cancer expressing GITR or PDl
  • Treatment of metastatic cancer can be achieved using the antibody molecules described herein.
  • the tumor is a cancer that expresses elevated levels of GITR and/or PDl.
  • the cancer described herein is colon cancer and metastatic cancer thereof.
  • the infection is acute or chronic.
  • the chronic infection is a persistent infection, a latent infection, or a slow infection.
  • the chronic infection is caused by a pathogen selected from the group consisting of bacteria, viruses, fungi, and protozoa.
  • the invention provides the use of an anti-GITR antibody or fragment thereof for the manufacture or preparation of a medicament for the treatment of a related disease or condition as referred to herein.
  • an antibody or antibody fragment or immunoconjugate or composition or product of the invention delays the onset of a condition and/or a condition associated with the condition.
  • the prophylactic or therapeutic methods described herein further comprise administering to the subject or individual a combination of an antibody molecule or pharmaceutical composition or immunoconjugate disclosed herein, and one or more additional therapies, For example, treatment modalities and/or other therapeutic agents.
  • the treatment modality includes surgery (eg, tumor resection); radiation therapy (eg, external particle beam therapy, which involves three-dimensional conformal radiation therapy in which the illumination region is designed), local illumination (eg, pointing to a pre-selected target) Or irradiation of organs) or focused illumination).
  • surgery eg, tumor resection
  • radiation therapy eg, external particle beam therapy, which involves three-dimensional conformal radiation therapy in which the illumination region is designed
  • local illumination eg, pointing to a pre-selected target
  • irradiation of organs e.g, a pre-selected target
  • focused illumination e.g, a pre-selected target
  • the therapeutic agent is selected from the group consisting of a chemotherapeutic agent, a cytotoxic agent, a vaccine, other antibodies, an anti-infective active agent, a small molecule drug, or an immunomodulatory agent.
  • Exemplary vaccines include, but are not limited to, cancer vaccines.
  • the vaccine can be a DNA based vaccine, an RNA based vaccine or a virus transduced vaccine.
  • Cancer vaccines can be prophylactic or therapeutic.
  • anti-infective active agents include, but are not limited to, antiviral, antifungal, antiprotozoal, antibacterial agents such as the nucleoside analog zidovudine (AST), ganciclovir, foscarnet or cidovir As described above.
  • Exemplary additional antibodies include, but are not limited to, anti-PD-1 antibodies, anti-PDL1 antibodies, or anti-PDL2 antibodies.
  • anti-PD-1 antibodies, anti-PDL1 antibodies or anti-PDL2 antibodies are known in the art, see for example WO2007/005874, WO2009/101611, WO2009/114335, WO2010/027827 and WO2011/066342, and the like.
  • the antibody combinations described herein can be administered separately, eg, as separate antibodies, or when ligated (eg, as a bispecific or trispecific antibody molecule).
  • Such combination therapies encompasses combined administration (eg, two or more therapeutic agents are included in the same formulation or separate formulations), and administered separately, in which case other therapeutic agents and/or agents may be administered.
  • Administration of the antibodies of the invention occurs previously, simultaneously, and/or after.
  • administration of the anti-GITR antibody and administration of the additional therapeutic agent are within about one month, or within about one, two or three weeks, or about 1, 2, 3, 4, 5, or 6 days of each other. occur.
  • the therapeutically effective amount of the pharmaceutical composition comprising the anti-GITR antibody to be employed will depend, for example, on the therapeutic background and objectives. Those skilled in the art will appreciate that appropriate dosage levels for treatment will vary, depending in part on the molecule being delivered, the indication being used, the route of administration, and the weight, body surface area or organ size and/or condition of the patient (age and general health status). In certain embodiments, the clinician can titrate the dose and alter the route of administration to achieve optimal therapeutic effect.
  • the frequency of administration will depend on the pharmacokinetic parameters of the particular anti-GITR antibody in the formulation used. Typically, the clinician will administer the composition until the dose that achieves the desired effect is achieved.
  • the antibodies of the invention may therefore be administered in a single dose, or in two or more doses (which may comprise the same or different amounts of the desired molecule) over a period of time, or by continuous infusion via an implant device or catheter.
  • the appropriate dose can be determined by using appropriate dose response data.
  • the antibody can be administered to the patient over an extended period of time. In certain embodiments, the antibody is administered every two weeks, every month, every two months, every three months, every four months, every five months, or every six months.
  • the administration route of the pharmaceutical composition is according to known methods, for example, orally, by intravenous injection, intraperitoneal, intracerebral (intrinsic), intraventricular, intramuscular, intraocular, intraarterial, intraportal or intralesional. Route; through a sustained release system or through an implanted device.
  • the composition can be administered by bolus injection or by continuous infusion or by an implant device.
  • composition may also be topically applied via an implanted membrane, sponge or another suitable material on which the desired molecule is absorbed or encapsulated.
  • the device when an implant device is used, the device can be implanted into any suitable tissue or organ and the desired molecule can be delivered via diffusion, timed release bolus, or continuous administration.
  • any treatment can be performed by replacing or supplementing the anti-GITR antibody with the immunoconjugate of the invention.
  • any of the anti-GITR antibodies or antigen-binding fragments thereof provided herein can be used to detect the presence of GITR in a biological sample.
  • detection includes quantitative or qualitative detection, and exemplary detection methods may involve immunohistochemistry, immunocytochemistry, flow cytometry (eg, FACS), magnetic binding of antibody molecules, ELISA assays. Method, PCR-technology (for example, RT-PCR).
  • the biological sample is a blood, serum or other liquid sample of biological origin.
  • the biological sample comprises cells or tissues.
  • the biological sample is from a hyperproliferative or cancerous lesion.
  • an anti-GITR antibody for use in a diagnostic or detection method.
  • a method of detecting the presence of a GITR in a biological sample comprises detecting the presence of a GITR protein in a biological sample.
  • the GITR is a human GITR.
  • the method comprises contacting a biological sample with an anti-GITR antibody as described herein under conditions that permit binding of the anti-GITR antibody to GITR, and detecting whether a complex is formed between the anti-GITR antibody and the GITR . The formation of the complex indicates the presence of GITR.
  • the method can be an in vitro or in vivo method.
  • an anti-GITR antibody is used to select a subject suitable for treatment with an anti-GITR antibody, for example wherein GITR is a biomarker for selecting the subject.
  • a cancer or tumor can be diagnosed using an antibody of the invention, for example, to assess (eg, monitor) the treatment or progression, diagnosis, and/or staging of a disease (eg, cancer or tumor) described herein in a subject.
  • a labeled anti-GITR antibody is provided.
  • Labels include, but are not limited to, directly detected labels or moieties (such as fluorescent labels, chromophore labels, electron dense labels, chemiluminescent labels, and radioactive labels), as well as indirectly detected portions, such as enzymes or ligands, such as By enzymatic reaction or molecular interaction.
  • Exemplary labels include, but are not limited to, radioisotopes 32 P, 14 C, 125 I, 3 H, and 131 I, iridium groups such as rare earth chelates or ruthenium and its derivatives, rhodamine and its derivatives, Dan dansyl, umbelliferone, Luceriferase, for example, firefly luciferase and bacterial luciferase (U.S. Patent No.
  • luciferin 2,3-dihydrogen Pyridazinone, horseradish peroxidase (HR), alkaline phosphatase, beta-galactosidase, glucoamylase, lytic enzyme, carbohydrate oxidase, for example, glucose oxidase, galactose oxidase And glucose-6-phosphate dehydrogenase, heterocyclic oxidases such as uricase and xanthine oxidase, and enzymes utilizing hydrogen peroxide dye precursors such as HR, lactoperoxidase, or microperoxide Microperoxidase, biotin/avidin, spin labeling, phage labeling, stable free radicals, and more.
  • HR horseradish peroxidase
  • alkaline phosphatase beta-galactosidase
  • glucoamylase lytic enzyme
  • carbohydrate oxidase for example, glucose oxidase, galactose oxida
  • the sample is obtained prior to treatment with an anti-GITR antibody. In some embodiments, the sample is obtained prior to treatment with a cancer drug. In some embodiments, the sample is obtained after the cancer has metastasized. In some embodiments, the sample is formalin fixed, paraffin coated (FFPE). In some embodiments, the sample is a biopsy (eg, a core biopsy), a surgical specimen (eg, from a surgically resected specimen), or a fine needle aspirate.
  • FFPE formalin fixed, paraffin coated
  • the GITR is detected prior to treatment, for example, prior to initiation of treatment or prior to treatment after the treatment interval.
  • a method of treating a tumor or infection comprising: examining a subject (eg, a sample) (eg, a sample of a subject comprising cancer cells) for detecting the presence of a GITR, thereby determining A GITR value, the GITR value is compared to a control value, and if the GITR value is greater than the control value, the subject is administered a therapeutically effective amount of an anti-GITR antibody, optionally in combination with one or more other therapies (eg, as described herein) Anti-GITR antibodies), thus treating tumors or infections.
  • a subject eg, a sample
  • Hybridoma technology is achieved by fusing two cells while maintaining the main features of both. These two cells are antigen-immunized mouse spleen cells and mouse myeloma cells, respectively.
  • the main feature of mouse spleen cells (B lymphocytes) immunized by specific antigens is its antibody secretion function, but it cannot be continuously cultured in vitro, and mouse myeloma cells can divide and proliferate indefinitely under culture conditions, that is, The so-called immortality.
  • the selective medium only the hybrid cells in which the B cells are fused with the myeloma cells can have the ability to continue to culture, and form a cell clone having both the secretory function of the antibody and the immortality of the cells.
  • mice were immunized with human GITR protein, and then spleen cells of the mouse were fused with myeloma cells to obtain hybridoma cells capable of expressing positive antibodies.
  • mice were sacrificed by cervical dislocation, and the body surface was sterilized with 75% alcohol for 5 min, and then placed on the anatomical plate of the mouse in the ultra-clean table, and the left side was placed, and the limbs were fixed with a 7-gauge needle.
  • the spleen was removed by aseptic opening of the abdominal cavity, washed with a basal medium (configuration method as follows), and the connective tissue attached thereto was carefully removed.
  • the spleen was then transferred to another dish containing the basal medium. Press the spleen with a elbow needle, insert a small needle into the spleen, and squeeze with the scorpion to fully release the spleen cells to make a spleen cell suspension.
  • the cell suspension was filtered through a 70 ⁇ M cell sieve, washed once with 30 ml of basal medium, and centrifuged at 1200 rpm for 6 min.
  • Lysis of red blood cells The supernatant was removed and the cells were resuspended in 10 ml RBC lysis buffer (GIBCO). Then add 20 ml of RBC lysis buffer. The suspension was allowed to stand for 5 min and centrifuged at 1100 rpm for 6 min. After removing the supernatant, the cells were resuspended in 10 ml of basal medium, and then 30 ml of basal medium was added, and centrifuged at 1100 rpm for 6 min. After removing the supernatant, the cells were resuspended in 20 ml of basal medium and counted.
  • Electrofusion Mouse myeloma cells SP2/0 cells (ATCC) were resuspended in 20 ml of basal medium and counted. SP2/0 and spleen cells were mixed at a ratio of 1:2 to 1:1, and centrifuged at 1000 rpm for 6 min. After removing the supernatant, the mixed cells were resuspended in 10 ml of fusion buffer (BTXpress). Further, 15 ml of the fusion buffer was added, and the mixture was centrifuged at 1000 rpm for 5 minutes, and the supernatant was removed. After repeating the above steps, the cells were re-selected with an appropriate amount of fusion buffer, and the mixed cell density was adjusted to 1 ⁇ 10 7 cells/ml.
  • the parameters of the electrofusion amplifier are set as follows. 2 ml of the cell suspension was added to each electrofusion dish for electrofusion.
  • the cells were allowed to stand in an electric fusion dish for 5 min at room temperature. The cells were transferred to a centrifuge tube, and the cells were diluted to 1 to 2 ⁇ 10 4 cells/ml using a screening medium (the configuration method is as follows). 100 ⁇ l of the cell suspension was added to each well of a 96-well plate. The screening medium was changed on the 7th day after the fusion. Screening was performed 10 days after culture (or longer, depending on the state of cell growth). Hybridoma cells expressing a specific anti-GITR antibody were screened by FACS (C6 (BD Biosciences)).
  • Subcloning step A 96-well plate was prepared, and 200 ⁇ l of the basal medium as described above was added to each of the 2nd to 8th columns. The cells of the positive wells selected by the above fusion were made into cell suspensions and added to the first column. 100 ⁇ l of the cell suspension of column 1 was added to the second column, and after thorough mixing, 100 ⁇ l was added to the next column. The above procedure was repeated until the last column volume became 300 ⁇ l; the 96-well plate was allowed to stand for 15 min, and the count was observed under a microscope. The corresponding volume of 100 cells was added to 20 ml of the basal medium as described above, and the plates were mixed and mixed at 200 ⁇ l per well. After one week, the microscope was observed under the microscope, and the monoclonal wells were judged and labeled, and the positive wells were picked out.
  • Cryopreservation of cells Observe the state of the cells, and wait for the cells to grow well. When the viability is >90%, centrifuge at 1000 rpm for 5 min to remove the supernatant. Resuspend the cells to 1 ⁇ 10 7 cells/ml with cryopreservation solution (45.5% FBS, 44.5% RPMI-1640, 10% DMSO), dispense into a cryotube, place in a programmed cooling box, and freeze at -80 °C. Save.
  • cryopreservation solution 45.5% FBS, 44.5% RPMI-1640, 10% DMSO
  • the present invention utilizes molecular biological techniques to obtain antibody sequences in anti-GITR-positive hybridoma cells, and uses the same to construct a human-mouse chimeric antibody.
  • RNA extraction fresh cells, centrifuged at 300 g for 5 min, the supernatant was removed, and 500 ⁇ l of LY buffer (Biomiga) was added to the pellet (20 ⁇ l of ⁇ -mercaptoethanol was added per 1 ml before use) and mixed until clear. It was added to a DNA removal tube, centrifuged at 13,000 rpm for 2 min, and the flow-through was collected. 100% ethanol was added to the flow through solution at a ratio of 1/2, and mixed 5 times to clarify.
  • LY buffer Biomiga
  • RNA concentration was determined.
  • cDNA was obtained by reverse transcription using PrimeScript II 1st Strand cDNA Synthesis Kit (Takara):
  • the reaction system I is configured as follows:
  • Reaction System I After incubation at 65 ° C for 5 min, it was quickly cooled on ice. The following reverse transcription system was added to Reaction System I in a total amount of 20 ⁇ l:
  • reverse transcription translation was carried out under the following conditions: 42 ° C for 60 min ⁇ 95 ° C for 5 min, and then cooled on ice to obtain cDNA.
  • the heavy and light chain variable regions were amplified by PCR, and the PCR reaction system was as follows:
  • the PCR reaction conditions are as follows:
  • PCR product obtained by the above PCR reaction was added, 0.5 ⁇ l of pMD20-T vector (Clontech), 5 ⁇ l of Ligation Mighty Mix (Takara), gently mixed, and reacted at 37 ° C for 2 hours to obtain a ligation product.
  • VH Heavy chain variable region primers for mouse anti-GITR antibodies
  • VL Light chain variable region
  • TOP10 competent cells (Tiangen Biochemical Technology (Beijing) Co., Ltd.) were taken at -80 °C, and thawed on ice. 5 ⁇ l of the ligation product obtained above was added to the thawed TOP10 competent cells, mixed, and incubated on ice for 30 min. After heat shock at 42 °C for 90 s, it was rapidly cooled on ice for 2 min, and 900 ⁇ l of LB medium (Biotech (Shanghai) Co., Ltd.) was added to the EP tube, and cultured at 37 ° C for 1 h at 220 rpm shaker.
  • LB medium Biotech (Shanghai) Co., Ltd.
  • the PCR system is as follows:
  • the PCR amplification product was recovered by gel cutting.
  • the homology recombination system is as follows:
  • the reaction was carried out at 37 ° C for 30 min to obtain a recombinant product.
  • the recombinant product was transformed into TOP10 competent cells, and monoclonal sequencing was picked.
  • the clone containing the plasmid with the correct insertion direction was selected as a positive clone, and the positive clone was preserved.
  • a plasmid containing an anti-GITR antibody was extracted from the positive clone obtained above.
  • 293F cells (Invitrogen) were passaged according to the desired transfection volume, and the cell density was adjusted to 1.5 ⁇ 10 6 cells/ml one day before transfection. The cell density on the day of transfection was approximately 3 x 10 6 cells/ml.
  • a final volume of 1/10 of F17 medium (Gibco, A13835-01) was used as a transfection buffer, and an appropriate plasmid was added and mixed.
  • Add appropriate polyethyleneimine (PEI) Polysciences, 23966) to the plasmid (the ratio of plasmid to PEI is 1:3 in 293F cells), mix and incubate for 10 min at room temperature to obtain a DNA/PEI mixture.
  • PEI polyethyleneimine
  • the gravity column used for purification was treated with 0.5 M NaOH overnight, and the glass bottle and the like were washed with distilled water and then dry-baked at 180 ° C for 4 hours to obtain a purification column.
  • the collected medium was centrifuged at 4500 rpm for 30 min before purification, and the cells were discarded.
  • the supernatant was then filtered using a 0.22 ⁇ l filter.
  • Each tube was filled with 1 ml of Protein A and equilibrated with 10 ml of binding buffer (sodium phosphate 20 mM. NaCl 150 mM, pH 7.0). The filtered supernatant was added to the purification column and re-equilibrated with 15 ml of binding buffer.
  • elution buffer citric acid + sodium citrate 0.1 M, pH 3.5
  • eluate was collected
  • 80 ⁇ l of Tris-HCl was added per 1 ml of the eluate.
  • the collected antibodies were concentrated by ultrafiltration into PBS (Gibco, 70011-044), and the concentration was measured.
  • control antibody used in the present invention is a GITR antibody reported in the patent application US20130183321A1 (GITR, INC. (Cambridge, MA, US)), the light and heavy chain sequences thereof are SEQ ID NO: 44 and SEQ ID NO in US20130183321A1, respectively. :54, which will be referred to as TRX518 in the following.
  • Example 3 Biofilm thin layer interference technique for determining the binding kinetics of the chimeric antibody of the present invention to an antigen
  • the equilibrium dissociation constant (KD) of the antibody of the present invention in combination with human GITR was determined by biofilm thin layer interferometry (ForteBio).
  • the ForteBio affinity assay was performed according to the existing method (Estep, P et al, High throughput solution Based measurement of antibody-antigen affinity and epitope binning. MAbs, 2013.5(2): p. 270-8).
  • cDNA was cloned into pCHO1.0 vector (Invitrogen), a plasmid obtained was transfected into CHO-S cells (Invitrogen, ExpiCHO TM Expression System Kit , NO: A29133), generated CHO-S cells (CHO-hGITR) overexpressing human GITR.
  • CHO-hGITR cells were counted and diluted to 2 x 10 6 cells/ml, and 100 ⁇ l/well was added to a U-bottom 96-well plate. Centrifuge for 5 min at 400 g to remove the cell culture medium. Samples (chimeric antibodies CH22F4, CH37G5, and TRX518, respectively) (antibody dilution method: maximum antibody concentration of 400 nM, three-fold dilution in PBS, a total of 12 concentrations tested) were added to the U-plate and resuspended cells, 100 ⁇ l / Hole, allowed to stand on ice for 30 min. The supernatant was removed by centrifugation at 400 g for 5 min, and the cells were washed 1 time with PBS.
  • PBS was removed by centrifugation at 400 g for 5 min, and 100 ⁇ l of anti-human Fc-labeled secondary antibody (Souther Biotech; 2040-09) (diluted 1:200 in PBS) was added to each well, and incubated on ice for 30 min in the dark. The supernatant was removed by centrifugation at 400 g for 5 min, and the cells were washed 1 time with PBS. The cells were resuspended in 80 ⁇ l of 1 ⁇ PBS and detected by FACS.
  • the antibodies CH22F4 and CH37G5 all bind to human GITR molecules overexpressed on CHO-S cells with EC50 of 0.4568 nM and 0.9069 nM, respectively, and the binding ability is similar to that of TRX518.
  • the chimeric antibody obtained in Example 2 was humanized. And through the following steps for humanization:
  • the CDRs, light chain variable regions and heavy chain variable regions of the two humanized antibodies (HZ22F4 and HZ37G5) obtained in the present invention, and the amino acid sequences of the light and heavy chains are described in Tables 1-3 as described above.
  • the present invention is measured using ForteBio assay humanized antibody binds to human GITR solution equilibrium dissociation constant (K D).
  • the ForteBio affinity assay was the same as in Example 3 with the exception that the antibodies used were the humanized antibodies HZ22F4 and HZ37G5.
  • the affinities of the antibodies HZ22F4 and HZ37G5 are shown in Table 9:
  • the humanized antibodies HZ22F4 and HZ37G5 bind to human GITR overexpressed on CHO-S cells with EC50 of 0.5492 nM and 1.974 nM, respectively, and have similar or better EC50 values than TRX518, ie similar or Better combination ability.
  • cDNA was cloned into pCHO1.0 vector (Invitrogen), the plasmid was transfected into CHO-S cells (Invitrogen, ExpiCHO TM Expression System Kit , NO: A29133), generated CHO-S cells (CHO-cynoGITR) overexpressing cynomolgus GITR.
  • the rest of the test methods were the same as in Example 4 except that the antibodies used were humanized antibodies HZ22F4 and HZ37G5, and the binding conditions are shown in FIG.
  • the humanized antibodies HZ22F4 and HZ37G5 bind to the cynomolgus monkey GITR overexpressed on CHO-S cells with EC50 of 0.3533 nM and 0.9931 nM, respectively, and have similar or superior binding ability compared to TRX518.
  • the activating antibody against GITR can bind to the cell surface GITR molecule to activate the downstream NF-kappa B signaling pathway.
  • Hela-GITR Hela-GITR-NF kappa B luciferase
  • the cDNA encoding human GITR (sequence see SEQ ID NO: 41) was cloned into pCHO1.0 vector (Invitrogen), and the obtained plasmid and NF-kappaB luciferace reporter plasmid (Promega) were co-transfected into Hela cells (ATCC, Cat. No.: CCL-2TM), Hela cells (Hela-GITR) that overexpress the human GITR with the NF-kappaB luciferace reporter gene system.
  • HeLa-GITR cells in logarithmic growth phase were taken, the culture supernatant was discarded, and the cells were washed once with PBS (Gibco).
  • PBS Gibco
  • Add appropriate amount of Trypsin (Gibco) and digest at 37 ° C, 5% CO 2 for 2 min. 4 times Trypsin volume of DMEM medium (ATCC) containing 10% FBS was added, and the cells were transferred to a 50 ml centrifuge tube and counted, 400 g, and centrifuged for 5 min.
  • DMEM medium (ATCC) was added and the cells were resuspended to 1 x 10 5 cells/mL. The cells were added to a 96-well white cell culture plate (Nunclon) at 50 ⁇ l/well.
  • sample humanized antibody HZ22F4, HZ37G5 prepared by the present invention
  • positive control TRX5 positive control TRX528
  • the antibody dilution method was: the highest antibody concentration was 80 nM, and the dilution was three times in the assay buffer (2% FBS DMEM culture). In the base), a total of 10 concentrations were tested). Incubate for 6 hours in a 37 ° C / 5% CO 2 incubator.
  • Bio-GloTM buffer promega
  • Bio-GloTM substrate promega
  • Bio-GloTM reagent 100 ⁇ l/well, was added to the cells cultured for 6 hours as described above. Read immediately.
  • the antibodies HZ22F4 and HZ37G5 can effectively activate the NF kappa B signaling pathway with EC50 of 0.3394nM and 0.3208nM, respectively, which is significantly better than TRX518 (1.139nM). Activation ability.
  • CD4T cells were incubated together to detect the relative expression of IL-2 and IFN- ⁇ in the system, thereby reflecting the activation of CD4 T cells by different antibodies.
  • PBMC separation Take 50 ml of fresh blood from the donor, add 2.5 times PBS, gently add to FiColl (Thermo), divide into 4 tubes, centrifuge at 400 g for 30 min, increase the speed to 7, and decelerate to 0. At the end of centrifugation, the tube was gently removed, and the middle white cloud-like cell population was aspirated into PBS and washed twice with PBS.
  • FiColl Thermo
  • CD4 + T cell isolation PBMC cells isolated as described above were isolated and enriched for CD4 + T cells according to the EasySep Human CD4 + T Cell Enrichment Kit (stem cell) instructions and used in T cell culture medium (recipe is shown in the table below). Hanging.
  • CD4 + T cell activation taking the above-described CD4 + T cells isolated and cell density was adjusted by a T cell culture medium of 1 * 10 6 / ml, according cells: beads (1: 1) added Dynabeads Human T-Activator CD3 / CD28 (Invitrogen), cultured in a 5% CO 2 incubator at 37 ° C for one week.
  • T cell activation assay Nunc 96 well flat bottom plate (Nunclon) was taken, 100 ⁇ l/well was added to PBS diluted 0.25 ⁇ g/ml of Purified NA/LE Mouse anti-human CD3Clone UCHT1 (BD Biosciences), and coated at 37 ° C for 2 hours to remove the package. Being liquid.
  • the above-mentioned activated CD4 + T cells were removed for one week, washed twice with T cell culture medium, and the cell density was adjusted to 1*10 6 /ml, and 100 ⁇ l of the cell suspension per well was added to the 96-well flat bottom plate.
  • the antibody of the present invention HZ22F4, HZ37G5
  • TRX518 100 ⁇ l were simultaneously added (the antibody dilution method was: the highest antibody concentration was 400 nM, three-fold dilution in T cell medium, a total of 10 concentrations were tested) and Purified NA/LE Mouse antibody Human CD28Clone CD28 (BD Biosciences) (final concentration 2 ⁇ g/ml) was cultured for 3 and 5 days, and IL-2 and IFN- ⁇ were detected with Human IL-2 Kit 1000 Test and Human IFN gamma 1000 test kit (both purchased from cisbio). Relative expression level (in DeltaF%).
  • the experimental results are shown in Tables 10 and 11 and Figures 5 and 6.
  • the antibodies of the invention all efficiently activate CD4 + T cells in vitro.
  • the data unit in the table is the DeltaF% average.
  • the IgG1 subtype antibody against GITR can bind to the GITR molecule with high expression on the surface of Treg, which in turn mediates the ADCC effect to clear Treg.
  • Promega's Jurkat-ADCCNF-AT luciferase effector cell line (hereinafter referred to as ADCC effector cell) was used as a test cell strain and CHO-hGITR cells (as described above) as target cells, and the antibody of the present invention was incubated, and the test was carried out.
  • the expression of the light reporter gene reflects the activation of the NF-AT signal, thereby detecting the ADCC activity of the antibody.
  • ADCC effector cells in logarithmic growth phase (cultured according to Promega instructions), centrifuged to remove supernatant, washed twice with PBS, and resuspended in assay medium (5% low IgG serum in 1640 medium (Gibco)) to adjust cell density
  • assay medium 5% low IgG serum in 1640 medium (Gibco)
  • the CHO-hGITR target cells prepared and incubated as described above were centrifuged, and the supernatant was centrifuged, and the cell density was adjusted to 2*10 6 /ml by resuspending in the assay medium, and the cells were subjected to 1: 1 ratio was mixed and added to a 96-well white cell culture plate (Nunclon) at 50 ⁇ l/well.
  • sample humanized antibody HZ22F4, HZ37G5 prepared by the present invention
  • positive control antibody TRX528
  • the antibody dilution method was: the maximum antibody concentration was 66.66 nM, and the dilution was three times in the detection medium. A total of 12 concentrations were tested). Incubate for 12 hours in a 37 ° C / 5% CO 2 incubator.
  • Bio-GloTM buffer promega
  • Bio-GloTM substrate promega
  • Bio-GloTM reagent 100 ⁇ l/well, was added to the cells cultured for 12 hours as described above. Read immediately.
  • the antibodies HZ22F4 and HZ37G5 can effectively activate the NF-AT signaling pathway downstream of ADCC.
  • the EC50 is 0.02958nM and 0.04056nM, respectively. Compared with TRX518, it has a significantly better EC50. Value.
  • MC38 cells were used to inoculate hGITR transgenic mice to determine the anti-tumor effect of the GITR antibody of the present invention.
  • mice Female C57B1/6 background human GITR transgenic mice (approximately 5 weeks old) were purchased from Biotech Test Animal Technology Co., Ltd. The mice were domesticated for 7 days after arrival and the study was started.
  • Mouse MC38 cells were purchased from Nanjing Yinhe Biomedical Co., Ltd. and routinely subcultured for subsequent in vivo experiments in strict accordance with the instructions. The cells were collected by centrifugation, resuspended in sterile PBS and adjusted to a cell density of 5 x 10 6 /ml.
  • a MC38-hGITR tumor-bearing mouse model was established by subcutaneous inoculation of 0.2 ml of cell suspension on day 0 into the right abdomen region of human GITR transgenic mice.
  • mice with tumor volume ranging from 87.4 mm 3 to 228.4 mm 3 were selected and grouped according to the average tumor volume group (5 mice per group).
  • Antibody C is a monoclonal antibody "Antibody C" against anti-PD-1 (WO2017/133540).
  • the mice were administered on the 6th, 10th, 13th and 17th day after the inoculation, and the tumor volume and body weight of each group were monitored during the administration period. The monitoring frequency was 2 times/week and the monitoring was continued for 5 weeks.
  • TGI% 100%* (h-IgG control tumor volume – treatment group tumor volume) / (h-IgG control tumor volume - tumor volume before administration of h-IgG control group), wherein the average tumor volume before administration of the h-IgG control group was 106 mm 3 .
  • Tumor volume determination: The maximum long axis (L) and the largest broad axis (W) of the tumor were measured using a vernier caliper, and the tumor volume was calculated as follows: V L x W 2 /2. The weight was measured using an electronic balance.
  • ⁇ h-IgG was purchased from Equitech-Bio, article number: SLH56-0001.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a novel antibody and antibody fragment that specifically bind to GITR and a composition comprising the antibody or the antibody fragment. Moreover, the present invention relates to nucleic acids encoding the antibody or the antibody fragment thereof, a host cell comprising same, and a related use. Furthermore, the present invention relates to a therapeutic and diagnostic use for the antibody and antibody fragment.

Description

抗GITR抗体及其用途anti-GITR antibody and use thereof
本发明涉及特异性结合GITR的新型抗体和抗体片段以及含有所述抗体或抗体片段的组合物。此外,本发明涉及编码所述抗体或其抗体片段的核酸及包含其的宿主细胞,以及相关用途。此外,本发明涉及这些抗体和抗体片段的治疗和诊断用途。The present invention relates to novel antibodies and antibody fragments that specifically bind to GITR and compositions containing the antibodies or antibody fragments. Furthermore, the invention relates to nucleic acids encoding the antibodies or antibody fragments thereof, and host cells comprising the same, and related uses. Furthermore, the invention relates to the therapeutic and diagnostic use of these antibodies and antibody fragments.
背景技术Background technique
糖皮质激素诱导的肿瘤坏死因子受体(glucocorticoid induced tumor necrosis factor receptor,GITR),亦称作CD357及GITR-D,最早是在多柔比星处理的小鼠T细胞中被发现的(Nocentin,PNAS.1997;94:6216-6221),是肿瘤坏死因子受体(tumor necrosis factor receptor,TNFR)超家族的第18个成员(TNFRSF18),其他成员包括CD40,CD27,4-1BB,OX40等。GITR由其同源配体,GITR配体(GITRL)活化,可以激活下游的NF-kappa B信号通路。Glucocorticoid-induced tumor necrosis factor receptor (GITR), also known as CD357 and GITR-D, was first discovered in dopamine-treated mouse T cells (Nocentin, PNAS.1997;94:6216-6221), is the 18th member of the tumor necrosis factor receptor (TNFR) superfamily (TNFRSF18), and other members include CD40, CD27, 4-1BB, OX40 and the like. GITR is activated by its cognate ligand, the GITR ligand (GITRL), which activates the downstream NF-kappa B signaling pathway.
GITR在静息T细胞中以低水平表达;在T细胞活化后,表达显著上调。GITR在调节性T细胞(Treg)中高水平组成型表达,并且当这些细胞被活化时,表达进一步上调(Nocentini和Riccardi,E.J.Immunol.2005,35:1016-1022)。GITR is expressed at low levels in resting T cells; after T cell activation, expression is significantly upregulated. GITR is highly expressed constitutively in regulatory T cells (Treg), and when these cells are activated, expression is further upregulated (Nocentini and Riccardo, E.J. Immunol. 2005, 35: 1016-1022).
GITR配体(GITRL)主要在抗原递呈细胞(APC,包括巨噬细胞、B细胞、树突状细胞及内皮细胞)上表达。APC上的GITRL与应答T细胞上GITR的结合触发GITR信号传导,刺激效应性T细胞激活并抑制Treg细胞的活性。由此,GITR对效应T细胞和调节性T细胞具有若干作用,包括:共刺激和活化效应T细胞,降低Treg细胞对效应T细胞的抑制等(Nocentini,Eur.J.Immunol.2007,37:1165-1169)。GITR ligands (GITRL) are primarily expressed on antigen presenting cells (APCs, including macrophages, B cells, dendritic cells, and endothelial cells). Binding of GITRL on APC to GITR on response T cells triggers GITR signaling, stimulates effector T cell activation and inhibits Treg cell activity. Thus, GITR has several effects on effector T cells and regulatory T cells, including: costimulation and activation of effector T cells, and reduction of Treg cell suppression of effector T cells (Nocentini, Eur. J. Immunol. 2007, 37: 1165-1169).
这些作用意味着GITR信号通路的活化可以导致免疫应答的增强。因此,能够活化GITR信号通路的物质可以激活机体免疫应答,进而增加机体抗肿瘤和感染的能力等,同时GITR分子在肿瘤微环境中Treg细胞中高表达,因此利用GITR抗体的ADCC活性清除GITR分子高表达的Treg细胞可以进一步增强肿瘤杀伤活性。These effects imply that activation of the GITR signaling pathway can result in an increase in the immune response. Therefore, a substance capable of activating the GITR signaling pathway can activate an immune response, thereby increasing the body's ability to resist tumors and infections, and the GITR molecule is highly expressed in Treg cells in the tumor microenvironment, thereby utilizing the ADCC activity of the GITR antibody to clear the GITR molecule. The expressed Treg cells can further enhance tumor killing activity.
现有技术研究了多种GITR的抗体(参见CN103951753A、CN105829343A、CN106459203A、WO2016196792A1、WO2017068186A9等)。这样的抗体是GITR的激动剂(即,是激活型抗体),可诱导或增强GITR信号传导,在治疗需要增强免疫应答的多种GITR相关疾病或病症中是有效的。由于这样的抗GITR抗体是激活型抗体,故相对于其与抗原的结合亲和力,其激活活性的高低是其发挥活性(例如。免疫增强活性)更为关键的指标。A number of antibodies to GITR have been studied in the prior art (see CN103951753A, CN105829343A, CN106459203A, WO2016196792A1, WO2017068186A9, etc.). Such antibodies are agonists of GITR (i.e., are activating antibodies) that induce or enhance GITR signaling and are effective in treating a variety of GITR-related diseases or conditions that require an enhanced immune response. Since such an anti-GITR antibody is an activated antibody, its activation activity is a more critical indicator of its activity (for example, immunopotentiating activity) relative to its binding affinity to an antigen.
因此,需要开发具有更好的激活活性,更好ADCC效应,以及具有更好的疗效,例如抗肿瘤作用的抗体。Therefore, there is a need to develop antibodies that have better activation activity, better ADCC effects, and have better therapeutic effects, such as anti-tumor effects.
发明内容Summary of the invention
本发明因此提供了一种新的结合GITR的抗体,以及其抗原结合片段。The invention thus provides a novel antibody that binds to GITR, as well as antigen-binding fragments thereof.
在一些实施方案中,本发明的抗GITR抗体具有以下一个或多个特性:In some embodiments, an anti-GITR antibody of the invention has one or more of the following characteristics:
(i)以高亲和力与GITR结合;(i) combined with GITR with high affinity;
(ii)与细胞表面的GITR有效结合;(ii) effective binding to the GITR on the cell surface;
(iii)具有激动剂活性,例如能够有效激活NF-kappaB信号通路;(iii) having agonist activity, for example, capable of efficiently activating the NF-kappaB signaling pathway;
(iv)有效激活T细胞(例如CD4T细胞);(iv) effective activation of T cells (eg, CD4 T cells);
(v)能够有效介导ADCC效应;(v) can effectively mediate the ADCC effect;
(vi)具有更好的抗肿瘤活性,例如能够降低受试者中的肿瘤体积,同时不影响受试者的体重;(vi) having better anti-tumor activity, for example, capable of reducing tumor volume in a subject without affecting the weight of the subject;
(vii)与抗PD-1抗体组合能够更好地抑制肿瘤活性,例如能够降低受试者中的肿瘤体积,同时不影响受试者体重。(vii) Combination with an anti-PD-1 antibody can better inhibit tumor activity, for example, to reduce tumor volume in a subject without affecting the subject's body weight.
在一些实施方案中,本发明提供了结合GITR的抗体或其抗原结合片段,包含如SEQ ID NO:17、18、19或20所示的序列的3个重链CDR(HCDR),和/或如SEQ ID NO:21、22、23或24所示的序列 的3个轻链CDR(LCDR)。In some embodiments, the invention provides an antibody or antigen-binding fragment thereof that binds to GITR, comprising three heavy chain CDRs (HCDRs) of the sequence set forth in SEQ ID NO: 17, 18, 19 or 20, and/or 3 light chain CDRs (LCDR) of the sequence set forth in SEQ ID NO: 21, 22, 23 or 24.
在一些实施方案中,本发明提供了编码本发明抗体或其片段的核酸,包含所述核酸的载体,包含所述载体的宿主细胞。In some embodiments, the invention provides a nucleic acid encoding an antibody or fragment thereof of the invention, a vector comprising the nucleic acid, a host cell comprising the vector.
在一些实施方案中,本发明提供了制备本发明抗体或其片段的方法。In some embodiments, the invention provides methods of making an antibody or fragment thereof of the invention.
在一些实施方案中,本发明提供了包含本发明抗体的免疫缀合物、药物组合物和组合产品。In some embodiments, the invention provides immunoconjugates, pharmaceutical compositions, and combination products comprising an antibody of the invention.
本发明还提供了利用本发明抗体在受试者中介导ADCC或激活NF-kappaB信号通路的方法,以及预防或治疗癌症或感染的方法。The invention also provides methods of mediating ADCC or activating the NF-kappaB signaling pathway in a subject using an antibody of the invention, and methods of preventing or treating cancer or infection.
本发明还涉及在样品中检测GITR的方法。The invention also relates to a method of detecting GITR in a sample.
在下面的附图和具体实施方案中进一步说明本发明。然而,这些附图和具体实施方案不应被认为限制本发明的范围,并且本领域技术人员容易想到的改变将包括在本发明的精神和所附权利要求的保护范围内。The invention is further illustrated in the following figures and detailed description. However, the drawings and the specific embodiments are not to be construed as limiting the scope of the present invention, and the modifications which are obvious to those skilled in the art are included in the spirit of the invention and the scope of the appended claims.
附图说明DRAWINGS
图1显示了流式细胞术测定嵌合抗体与表达人GITR的CHO-S细胞株(CHO-hGITR)的结合能力。Figure 1 shows the ability of a chimeric antibody to bind to a CHO-S cell line expressing human GITR (CHO-hGITR) by flow cytometry.
图2显示了流式细胞术测定人源化抗体与表达人GITR的CHO-S细胞株(CHO-hGITR)的结合能力。Figure 2 shows the ability of humanized antibodies to bind to a human GITR-expressing CHO-S cell line (CHO-hGITR) by flow cytometry.
图3显示了流式细胞术测定人源化抗体与表达食蟹猴GITR的CHO-S细胞株(CHO-cynoGITR)的结合能力。Figure 3 shows the ability of humanized antibodies to bind to CHO-S cell line (CHO-cynoGITR) expressing cynomolgus GITR by flow cytometry.
图4显示了MOA法测定人源化抗体激活Hela-GITR-NF-Kappa B luciferace细胞株能力。Figure 4 shows the MOA assay for the ability of humanized antibodies to activate Hela-GITR-NF-Kappa B luciferace cell lines.
图5显示了人源化抗体激活CD4T细胞释放IL-2能力检测。Figure 5 shows the ability of humanized antibodies to activate IL-4 release from activated CD4 T cells.
图6显示了人源化抗体激活CD4T细胞释放IFN-γ能力检测。Figure 6 shows the ability of humanized antibodies to activate CD4 T cells to release IFN-[gamma].
图7显示了MOA法测定人源化抗体ADCC活性。Figure 7 shows the MOA assay for the determination of humanized antibody ADCC activity.
图8显示了HZ37G5分子体内药效检测。Figure 8 shows the in vivo pharmacodynamic assay of HZ37G5.
图9显示了HZ22F4分子体内药效检测。Figure 9 shows the in vivo efficacy test of HZ22F4 molecule.
图10显示了小鼠体重检测。Figure 10 shows mouse body weight detection.
发明详述Detailed description of the invention
I.定义I. Definition
在下文详细描述本发明前,应理解本发明不限于本文中描述的特定方法学、方案和试剂,因为这些可以变化。还应理解本文中使用的术语仅为了描述具体实施方案,而并不意图限制本发明的范围,其仅会由所附权利要求书限制。除非另外定义,本文中使用的所有技术和科学术语与本发明所属领域中普通技术人员通常的理解具有相同的含义。Before the present invention is described in detail below, it is to be understood that the invention is not limited to the particular methodology, aspects, and reagents described herein, as these may vary. It is also understood that the terminology used herein is for the purpose of describing the particular embodiment All technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs, unless otherwise defined.
为了解释本说明书,将使用以下定义,并且只要适当,以单数形式使用的术语也可以包括复数,并且反之亦然。要理解,本文所用的术语仅是为了描述具体的实施方案,并且不意欲是限制性的。For the purpose of interpreting the specification, the following definitions will be used, and the terms used in the singular may also include the plural, and vice versa, as appropriate. It is understood that the terminology used herein is for the purpose of describing particular embodiments and is not intended to be limiting.
术语“约”在与数字数值联合使用时意为涵盖具有比指定数字数值小5%的下限和比指定数字数值大5%的上限的范围内的数字数值。The term "about" when used in connection with a numerical value is meant to encompass a numerical value within the range of the lower limit of 5% less than the specified numerical value and the upper limit of 5% greater than the specified numerical value.
如本文所用,术语“和/或”意指可选项中的任一项或可选项的两项。As used herein, the term "and/or" means either of the alternatives or two of the alternatives.
如本文所用,术语“包含”或“包括”意指包括所述的要素、整数或步骤,但是不排除任意其他要素、整数或步骤。在本文中,当使用术语“包含”或“包括”时,除非另有指明,否则也涵盖由所述及的要素、整数或步骤组合的情形。例如,当提及“包含”某个具体序列的抗体可变区时,也旨在涵盖由该具体序列组成的抗体可变区。The term "comprising" or "including", as used herein, is meant to include the recited elements, integers or steps, but does not exclude any other elements, integers or steps. As used herein, when the term "comprises" or "comprising" is used, it is intended to encompass a combination of elements, integers or steps. For example, when referring to an antibody variable region that "comprises" a particular sequence, it is also intended to encompass an antibody variable region consisting of that particular sequence.
如本文所用,术语“GITR”是指“糖皮质激素诱导的TNF相关基因和/或多肽”,在本领域中也称为TNF受体超家族18(TNFRSF18),指来自任何脊椎动物来源,包括哺乳动物诸如灵长类(例如人、 食蟹猴)和啮齿类(例如小鼠和大鼠)的任何天然GITR,除非另有说明。该术语涵盖“全长”,未加工的GITR以及因细胞中的加工所致的任何形式的GITR。该术语还涵盖GITR的天然发生变体,例如剪接变体或等位变体。人GITR氨基酸序列可参见GenBank登记号Q9Y5U5。一个具体的人GITR多肽的氨基酸序列可见于SEQ ID NO:41中。一个具体的食蟹猴GITR多肽的氨基酸序列见SEQ ID NO:42。As used herein, the term "GITR" refers to "glucocorticoid-induced TNF-related genes and/or polypeptides", also referred to in the art as TNF receptor superfamily 18 (TNFRSF18), and refers to any vertebrate source, including Any natural GITR of mammals such as primates (eg, humans, cynomolgus monkeys) and rodents (eg, mice and rats), unless otherwise indicated. The term encompasses "full length", unprocessed GITR, and any form of GITR due to processing in cells. The term also encompasses naturally occurring variants of GITR, such as splice variants or allelic variants. The human GITR amino acid sequence can be found in GenBank Accession No. Q9Y5U5. The amino acid sequence of a particular human GITR polypeptide can be found in SEQ ID NO:41. The amino acid sequence of a specific cynomolgus monkey GITR polypeptide is set forth in SEQ ID NO:42.
本文所用的术语“抗GITR抗体”、“抗GITR”、“GITR抗体”或“结合GITR的抗体”是指这样的抗体,所述抗体能够以足够的亲合力结合(人或食蟹猴)GITR蛋白或其片段以致所述抗体可以用作靶向(人或食蟹猴)GITR中的诊断剂和/或治疗剂。在一个实施方案中,抗GITR抗体与非(人或食蟹猴)GITR蛋白结合的程度低于所述抗体与(人或食蟹猴)GITR结合的约10%、约20%、约30%、约40%、约50%、约60%、约70%、约80%或约90%或以上,如例如通过放射性免疫测定(RIA)或生物光干涉测定法或MSD测定法测量的。The term "anti-GITR antibody", "anti-GITR", "GITR antibody" or "antibody that binds GITR" as used herein refers to an antibody capable of binding (human or cynomolgus) GITR with sufficient affinity. The protein or fragment thereof such that the antibody can be used as a diagnostic and/or therapeutic agent in a targeted (human or cynomolgus) GITR. In one embodiment, the anti-GITR antibody binds to a non- (human or cynomolgus) GITR protein to a lesser extent than about 10%, about 20%, about 30% of the antibody binds to (human or cynomolgus) GITR. About 40%, about 50%, about 60%, about 70%, about 80% or about 90% or more, as measured, for example, by radioimmunoassay (RIA) or biooptical interferometry or MSD assay.
“抗体片段”指与完整抗体不同的分子,其包含完整抗体的一部分且结合完整抗体所结合的抗原。抗体片段的例子包括但不限于Fv,Fab,Fab’,Fab’-SH,F(ab’)2;双抗体;线性抗体;单链抗体(例如scFv);单结构域抗体;双价或双特异性抗体或其片段;骆驼科抗体;和由抗体片段形成的双特异性抗体或多特异性抗体。"Antibody fragment" refers to a molecule that is distinct from an intact antibody that comprises a portion of an intact antibody and binds to an antigen to which the intact antibody binds. Examples of antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single chain antibodies (eg, scFv); single domain antibodies; Specific antibodies or fragments thereof; camelid antibodies; and bispecific antibodies or multispecific antibodies formed from antibody fragments.
如本文所用,术语“表位”指抗原(例如,GITR)中与抗体分子特异性相互作用的部分。As used herein, the term "epitope" refers to a portion of an antigen (eg, GITR) that specifically interacts with an antibody molecule.
与参照抗体“结合相同或重叠表位的抗体”是指这样的抗体,其在竞争测定中阻断50%以上的所述参照抗体与其抗原的结合,反言之,参照抗体在竞争测定中阻断50%以上的该抗体与其抗原的结合。An antibody that "binds to the same or overlapping epitope" as a reference antibody refers to an antibody that blocks more than 50% of the binding of the reference antibody to its antigen in a competition assay, in other words, the reference antibody is blocked in the competition assay. More than 50% of the antibody binds to its antigen.
与参照抗体竞争结合其抗原的抗体是指这样的抗体,其在竞争测定中阻断50%、60%、70%、80%、90%或95%以上的所述参照抗体与其抗原的结合。反言之,参照抗体在竞争测定中阻断50%、60%、70%、80%、90%或95%以上的该抗体与其抗原的结合。众多类型的竞争性结合测定可用于确定一种抗体是否与另一种竞争,这些测定例如:固相直接或间接放射免疫测定(RIA)、固相直接或间接酶免疫测定(EIA)、夹心竞争测定(参见例如Stahli等,1983,Methods in Enzymology 9:242-253)。An antibody that competes with a reference antibody for binding to its antigen refers to an antibody that blocks 50%, 60%, 70%, 80%, 90% or more of the binding of the reference antibody to its antigen in a competition assay. In other words, the reference antibody blocks 50%, 60%, 70%, 80%, 90% or more of the binding of the antibody to its antigen in a competition assay. Numerous types of competitive binding assays can be used to determine whether an antibody competes with another assay such as solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition. Assay (see for example Stahli et al, 1983, Methods in Enzymology 9: 242-253).
抑制(例如竞争性抑制)参照抗体与其抗原的结合的抗体是指这样的抗体,其抑制50%、60%、70%、80%、90%或95%以上的所述参照抗体与其抗原的结合。反言之,参照抗体抑制50%、60%、70%、80%、90%或95%以上的该抗体与其抗原的结合。抗体与其抗原的结合可以亲和力(例如平衡解离常数)衡量。测定亲和力的方法是本领域已知的。An antibody that inhibits (eg, competitively inhibits) binding of a reference antibody to its antigen refers to an antibody that inhibits binding of 50%, 60%, 70%, 80%, 90%, or 95% of the reference antibody to its antigen. . Conversely, the reference antibody inhibits binding of the antibody to its antigen by 50%, 60%, 70%, 80%, 90% or more. The binding of an antibody to its antigen can be measured by affinity (eg, equilibrium dissociation constant). Methods for determining affinity are known in the art.
与参照抗体显示相同或相似的结合亲和力和/或特异性的抗体是指这样的抗体,其能够具有参照抗体的至少50%、60%、70%、80%、90%或95%以上的结合亲和力和/或特异性。这可以通过本领域已知的任何测定结合亲和力和/或特异性的方法进行测定。An antibody that exhibits the same or similar binding affinity and/or specificity as a reference antibody refers to an antibody that is capable of binding at least 50%, 60%, 70%, 80%, 90% or 95% of the reference antibody. Affinity and / or specificity. This can be determined by any method known in the art for determining binding affinity and/or specificity.
“互补决定区”或“CDR区”或“CDR”是抗体可变结构域中在序列上高变并且形成在结构上确定的环(“超变环”)和/或含有抗原接触残基(“抗原接触点”)的区域。CDR主要负责与抗原表位结合。重链和轻链的CDR通常被称作CDR1、CDR2和CDR3,从N-端开始顺序编号。位于抗体重链可变结构域内的CDR被称作HCDR1、HCDR2和HCDR3,而位于抗体轻链可变结构域内的CDR被称作LCDR1、LCDR2和LCDR3。在一个给定的轻链可变区或重链可变区氨基酸序列中,各CDR的精确氨基酸序列边界可以使用许多公知的抗体CDR指派系统的任一种或其组合确定,所述指派系统包括例如:基于抗体的三维结构和CDR环的拓扑学的Chothia(Chothia等人.(1989)Nature 342:877-883,Al-Lazikani等人,“Standard conformations for the canonical structures of immunoglobulins”,Journal of Molecular Biology,273,927-948(1997)),基于抗体序列可变性的Kabat(Kabat等人, Sequences of Proteins of Immunological Interest,第4版,U.S.Department of Health and Human Services,National Institutes of Health(1987)),AbM(University of Bath),Contact(University College London),国际ImMunoGeneTics database(IMGT)(在万维网上imgt.cines.fr/上),以及基于利用大量晶体结构的近邻传播聚类(affinity propagation clustering)的North CDR定义。A "complementarity determining region" or "CDR region" or "CDR" is a sequence that is hypervariable in an antibody variable domain and that forms a structurally defined loop ("hypervariable loop") and/or contains an antigen contact residue ( The area of the "antigen contact point"). The CDR is primarily responsible for binding to an epitope. The CDRs of the heavy and light chains are commonly referred to as CDR1, CDR2 and CDR3, numbered sequentially from the N-terminus. The CDRs located within the antibody heavy chain variable domain are referred to as HCDR1, HCDR2 and HCDR3, while the CDRs located within the antibody light chain variable domain are referred to as LCDR1, LCDR2 and LCDR3. In a given light chain variable region or heavy chain variable region amino acid sequence, the exact amino acid sequence boundaries of each CDR can be determined using any one or combination of a number of well-known antibody CDR assignment systems, including For example: Chothia based on the three-dimensional structure of the antibody and the topology of the CDR loop (Chothia et al. (1989) Nature 342: 877-883, Al-Lazikani et al., "Standard conformations for the canonical structures of immunoglobulins", Journal of Molecular Biology, 273, 927-948 (1997)), Kabat based on antibody sequence variability (Kabat et al, Sequences of Proteins of Immunological Interest, 4th edition, US Department of Health and Human Services, National Institutes of Health (1987)), AbM (University of Bath), Contact (University College London), International ImMunoGeneTics database (IMGT) (imgt.cines.fr/ on the World Wide Web), and based on affinity propagation clustering using a large number of crystal structures North CDR definition.
例如,根据不同的CDR确定方案,每一个CDR的残基如下所述。For example, depending on the CDR determination protocol, the residues of each CDR are as follows.
Figure PCTCN2019083241-appb-000001
Figure PCTCN2019083241-appb-000001
CDR也可以基于与参考CDR序列(例如本发明示例性CDR之任一)具有相同的Kabat编号位置而确定。The CDRs can also be determined based on having the same Kabat numbering position as the reference CDR sequence (e.g., any of the exemplary CDRs of the invention).
除非另有说明,否则在本发明中,术语“CDR”或“CDR序列”涵盖以上述任一种方式确定的CDR序列。Unless otherwise indicated, in the present invention, the term "CDR" or "CDR sequence" encompasses a CDR sequence determined in any of the above manners.
除非另有说明,否则在本发明中,当提及抗体可变区中的残基位置(包括重链可变区残基和轻链可变区残基)时,是指根据Kabat编号系统(Kabat等人,Sequences of Proteins of Immunological Interest,5th Ed.Public Health Service,National Institutes of Health,Bethesda,Md.(1991))的编号位置。Unless otherwise indicated, in the present invention, when referring to the position of a residue in the variable region of an antibody (including a heavy chain variable region residue and a light chain variable region residue), it is meant according to the Kabat numbering system ( The numbered positions of Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991).
在一个实施方案中,本发明抗体的CDR通过AbM规则确定边界,例如如表1所示。In one embodiment, the CDRs of the antibodies of the invention are bordered by AbM rules, for example as shown in Table 1.
然而,应该注意,基于不同的指派系统获得的同一抗体的可变区的CDR的边界可能有所差异。即不同指派系统下定义的同一抗体可变区的CDR序列有所不同。因此,在涉及用本发明定义的具体CDR序列限定抗体时,所述抗体的范围还涵盖了这样的抗体,其可变区序列包含所述的具体CDR序列,但是由于应用了不同的方案(例如不同的指派系统规则或组合)而导致其所声称的CDR边界与本发明所定义的具体CDR边界不同。However, it should be noted that the boundaries of the CDRs of the variable regions of the same antibody obtained based on different assignment systems may vary. That is, the CDR sequences of the same antibody variable region defined under different assignment systems are different. Thus, where an antibody is defined by a particular CDR sequence as defined by the present invention, the scope of the antibody also encompasses an antibody whose variable region sequence comprises the particular CDR sequence, but due to the application of a different protocol (eg Different assignment system rules or combinations result in different claimed CDR boundaries than the specific CDR boundaries defined by the present invention.
具有不同特异性(即,针对不同抗原的不同结合位点)的抗体具有不同的CDR。然而,尽管CDR在抗体与抗体之间是不同的,但是CDR内只有有限数量的氨基酸位置直接参与抗原结合。使用Kabat,Chothia,AbM、Contact和North方法中的至少两种,可以确定最小重叠区域,从而提供用于抗原结合的“最小结合单位”。最小结合单位可以是CDR的一个子部分。正如本领域技术人员明了,通过抗体的结构和蛋白折叠,可以确定CDR序列其余部分的残基。因此,本发明也考虑本文所给出的任何CDR的变体。例如,在一个CDR的变体中,最小结合单位的氨基酸残基可以保持不变,而根据Kabat或 Chothia定义的其余CDR残基可以被保守氨基酸残基替代。Antibodies with different specificities (ie, different binding sites for different antigens) have different CDRs. However, although the CDRs differ between antibodies and antibodies, only a limited number of amino acid positions within the CDRs are directly involved in antigen binding. Using at least two of the Kabat, Chothia, AbM, Contact, and North methods, the minimal overlap region can be determined to provide a "minimum binding unit" for antigen binding. The minimum binding unit can be a sub-portion of the CDR. As will be apparent to those skilled in the art, residues of the remainder of the CDR sequences can be determined by the structure of the antibody and protein folding. Accordingly, the invention also contemplates variants of any of the CDRs presented herein. For example, in a variant of a CDR, the amino acid residues of the smallest binding unit may remain unchanged, while the remaining CDR residues defined by Kabat or Chothia may be replaced by conservative amino acid residues.
术语“PD-1轴结合拮抗剂”是指如下的分子,其抑制PD-1轴结合配偶与一种或多种它的结合配偶相互作用,从而去除源自PD-1信号传导轴上的信号传导的T细胞功能障碍-一项结果是恢复或增强T细胞功能(例如增殖,细胞因子生成,靶细胞杀伤)。如本文中使用的,PD-1轴结合拮抗剂包括PD-1结合拮抗剂(例如抗PD-1抗体),PD-L1结合拮抗剂(例如抗PD-L1抗体)和PD-L2结合拮抗剂(例如抗PD-L2抗体)。The term "PD-1 axis binding antagonist" refers to a molecule that inhibits the interaction of a PD-1 axis binding partner with one or more of its binding partners, thereby removing signals originating from the PD-1 signaling axis. Conducted T cell dysfunction - one result is the restoration or enhancement of T cell function (eg proliferation, cytokine production, target cell killing). As used herein, PD-1 axis binding antagonists include PD-1 binding antagonists (eg, anti-PD-1 antibodies), PD-L1 binding antagonists (eg, anti-PD-L1 antibodies), and PD-L2 binding antagonists. (eg anti-PD-L2 antibody).
术语“PD-1结合拮抗剂”指如下的分子,其降低,阻断,抑制,消除或干扰源自PD-1与一种或多种它的结合配偶(诸如PD-L1,PD-L2)相互作用的信号转导。在一些实施方案中,PD-1结合拮抗剂是抑制PD-1结合一种或多种它的结合配偶的分子。在一个特定方面,PD-1结合拮抗剂抑制PD-1结合PD-L1和/或PD-L2。例如,PD-1结合拮抗剂包括降低,阻断,抑制,消除或干扰源自PD-1与PD-L1和/或PD-L2相互作用的信号转导的抗PD-1抗体,其抗原结合片段,免疫粘附素,融合蛋白,寡肽和其它分子。在一个实施方案中,PD-1结合拮抗剂降低由或经由T淋巴细胞上表达的细胞表面蛋白质介导的负面共刺激信号(经由PD-1介导信号传导),从而使得功能障碍性T细胞不太功能障碍性(例如增强对抗原识别的效应器应答)。在一些实施方案中,PD-1结合拮抗剂是抗PD-1抗体。在一个具体实施方案中,PD-1结合拮抗剂是WO2015/095423中公开的MDX-1106(nivolumab)、MK-3475(pembrolizumab)、CT-011(pidilizumab)或AMP-224。在一个实施方案中,抗PD-1抗体是“Antibody C”(WO2017/133540)。The term "PD-1 binding antagonist" refers to a molecule that reduces, blocks, inhibits, eliminates or interferes with the binding of PD-1 to one or more of its binding partners (such as PD-L1, PD-L2). Interacting signal transduction. In some embodiments, the PD-1 binding antagonist is a molecule that inhibits PD-1 binding to one or more of its binding partners. In a specific aspect, the PD-1 binding antagonist inhibits PD-1 binding to PD-L1 and/or PD-L2. For example, a PD-1 binding antagonist includes an anti-PD-1 antibody that reduces, blocks, inhibits, abolishes or interferes with signal transduction derived from PD-1 interaction with PD-L1 and/or PD-L2, antigen binding Fragments, immunoadhesins, fusion proteins, oligopeptides and other molecules. In one embodiment, the PD-1 binding antagonist reduces a cell surface protein-mediated negative costimulatory signal (via PD-1 mediated signaling) expressed on or via T lymphocytes, thereby enabling dysfunctional T cells Less dysfunctional (eg, enhancing effector response to antigen recognition). In some embodiments, the PD-1 binding antagonist is an anti-PD-1 antibody. In a specific embodiment, the PD-1 binding antagonist is MDX-1106 (nivolumab), MK-3475 (pembrolizumab), CT-011 (pidilizumab) or AMP-224 as disclosed in WO2015/095423. In one embodiment, the anti-PD-1 antibody is "Antibody C" (WO 2017/133540).
术语“PD-L1结合拮抗剂”指如下的分子,其降低,阻断,抑制,消除或干扰源自PD-L1与一种或多种它的结合配偶(诸如PD-1,B7-1)相互作用的信号转导。在一些实施方案中,PD-L1结合拮抗剂是抑制PD-L1结合它的结合配偶的分子。在一个特定方面,PD-L1结合拮抗剂抑制PD-L1结合PD-1和/或B7-1。在一些实施方案中,PD-L1结合拮抗剂包括降低,阻断,抑制,消除或干扰源自PD-L1与一种或多种它的结合配偶(诸如PD-1,B7-1)相互作用的信号转导的抗PD-L1抗体,其抗原结合片段,免疫粘附素,融合蛋白,寡肽和其它分子。在一个实施方案中,PD-L1结合拮抗剂降低由或经由T淋巴细胞上表达的细胞表面蛋白质介导的负面共刺激信号(经由PD-L1介导信号传导),从而使得功能障碍性T细胞不太功能障碍性(例如增强对抗原识别的效应器应答)。在一些实施方案中,PD-L1结合拮抗剂是抗PD-L1抗体。在一个具体方面,抗PD-L1抗体是WO2015/095423中公开的YW243.55.S70、MDX-1105、MPDL3280A或MEDI4736。The term "PD-L1 binding antagonist" refers to a molecule that reduces, blocks, inhibits, eliminates or interferes with the binding of PD-L1 from one or more of its binding partners (such as PD-1, B7-1). Interacting signal transduction. In some embodiments, the PD-L1 binding antagonist is a molecule that inhibits PD-L1 binding to its binding partner. In a specific aspect, the PD-L1 binding antagonist inhibits PD-L1 binding to PD-1 and/or B7-1. In some embodiments, the PD-L1 binding antagonist comprises reducing, blocking, inhibiting, eliminating or interfering with the interaction of PD-L1 from one or more of its binding partners (such as PD-1, B7-1) Signal transduction of anti-PD-L1 antibodies, antigen-binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules. In one embodiment, the PD-L1 binding antagonist reduces a cell surface protein-mediated negative costimulatory signal (via PD-L1 mediated signaling) expressed on or via T lymphocytes, thereby enabling dysfunctional T cells Less dysfunctional (eg, enhancing effector response to antigen recognition). In some embodiments, the PD-L1 binding antagonist is an anti-PD-L1 antibody. In a specific aspect, the anti-PD-L1 antibody is YW243.55.S70, MDX-1105, MPDL3280A or MEDI4736 as disclosed in WO2015/095423.
术语“PD-L2结合拮抗剂”指如下的分子,其降低,阻断,抑制,消除或干扰源自PD-L2与一种或多种它的结合配偶(诸如PD-1)相互作用的信号转导。在一些实施方案中,PD-L2结合拮抗剂是抑制PD-L2结合一种或多种它的结合配偶的分子。在一个特定方面,PD-L2结合拮抗剂抑制PD-L2结合PD-1。在一些实施方案中,PD-L2拮抗剂包括降低,阻断,抑制,消除或干扰源自PD-L2与一种或多种它的结合配偶(诸如PD-1)相互作用的信号转导的抗PD-L2抗体,其抗原结合片段,免疫粘附素,融合蛋白,寡肽和其它分子。在一个实施方案中,PD-L2结合拮抗剂降低由或经由T淋巴细胞上表达的细胞表面蛋白质介导的负面共刺激信号(经由PD-L2介导信号传导),从而使得功能障碍性T细胞不太功能障碍性(例如增强对抗原识别的效应器应答)。在一些实施方案中,PD-L2结合拮抗剂是免疫粘附素。The term "PD-L2 binding antagonist" refers to a molecule that reduces, blocks, inhibits, eliminates or interferes with signals derived from the interaction of PD-L2 with one or more of its binding partners, such as PD-1. divert. In some embodiments, the PD-L2 binding antagonist is a molecule that inhibits PD-L2 binding to one or more of its binding partners. In a specific aspect, the PD-L2 binding antagonist inhibits PD-L2 binding to PD-1. In some embodiments, the PD-L2 antagonist comprises reducing, blocking, inhibiting, eliminating or interfering with signal transduction derived from the interaction of PD-L2 with one or more of its binding partners, such as PD-1. Anti-PD-L2 antibodies, antigen-binding fragments thereof, immunoadhesins, fusion proteins, oligopeptides and other molecules. In one embodiment, the PD-L2 binding antagonist reduces a cell surface protein-mediated negative costimulatory signal (via PD-L2 mediated signaling) expressed on or via T lymphocytes, thereby enabling dysfunctional T cells Less dysfunctional (eg, enhancing effector response to antigen recognition). In some embodiments, the PD-L2 binding antagonist is an immunoadhesin.
“抗体依赖性细胞介导的细胞毒性”或“ADCC”指其中结合到某些细胞毒性细胞(例如NK细胞,嗜中性粒细胞和巨噬细胞)上存在的Fc受体(FcR)上的分泌型免疫球蛋白使得这些细胞毒性效应细胞能够特异性结合携带抗原的靶细胞,随后用细胞毒素杀死靶细胞的细胞毒性形式。介导ADCC的主要 细胞NK细胞只表达FcγRIII,而单核细胞表达FcγRI,FcγRII和FcγRIII。Ravetch和Kinet,Annu.Rev.Immunol.9:457-92(1991)第464页表3总结了造血细胞上的FcR表达。为了评估目的分子的ADCC活性,可进行体外ADCC测定法,诸如美国专利No.5,500,362或5,821,337或美国专利No.6,737,056(Presta)中所记载的。可用于此类测定法的效应细胞包括PBMC和NK细胞。或者/另外,可在体内评估目的分子的ADCC活性,例如在动物模型中,诸如Clynes等人,PNAS(USA)95:652-656(1998)中所披露的。本文实施例中提供了用于评估ADCC活性的一种例示性测定法。"Antibody-dependent cell-mediated cytotoxicity" or "ADCC" refers to an Fc receptor (FcR) that is present on certain cytotoxic cells (eg, NK cells, neutrophils, and macrophages). Secretory immunoglobulins enable these cytotoxic effector cells to specifically bind to target cells carrying the antigen, followed by cytotoxicity to kill the cytotoxic form of the target cells. The primary cell NK cells that mediate ADCC express only FcyRIII, whereas monocytes express FcyRI, FcyRII and FcyRIII. Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-92 (1991) summarizes FcR expression on hematopoietic cells. In order to assess the ADCC activity of the molecule of interest, an in vitro ADCC assay can be performed, such as that described in U.S. Patent No. 5,500,362 or 5,821,337 or U.S. Patent No. 6,737,056 (Presta). Effector cells that can be used in such assays include PBMC and NK cells. Alternatively, or additionally, the ADCC activity of the molecule of interest can be assessed in vivo, for example, in animal models such as those disclosed in Clynes et al, PNAS (USA) 95:652-656 (1998). An exemplary assay for assessing ADCC activity is provided in the Examples herein.
术语“细胞毒性剂”用在本发明中指抑制或防止细胞功能和/或引起细胞死亡或破坏的物质。The term "cytotoxic agent" as used in the present invention refers to a substance which inhibits or prevents cell function and/or causes cell death or destruction.
“化疗剂”包括在治疗癌症中有用的化学化合物。化疗剂的例子参见WO2015/031667中所公开的那些,包括但不限于抗肿瘤剂,包括烷化剂;抗代谢物;天然产物;抗生素;酶;杂类试剂;激素和拮抗剂;抗雌激素;抗雄激素;以及非类固醇抗雄激素等。"Chemotherapeutic agents" include chemical compounds that are useful in the treatment of cancer. For examples of chemotherapeutic agents, see those disclosed in WO 2015/031667, including but not limited to antineoplastic agents, including alkylating agents; antimetabolites; natural products; antibiotics; enzymes; heterogeneous agents; hormones and antagonists; ; antiandrogens; and non-steroidal antiandrogens.
术语“小分子药物”是指低分子量的能够调节生物过程的有机化合物。The term "small molecule drug" refers to a low molecular weight organic compound capable of modulating a biological process.
“功能性Fc区”拥有天然序列Fc区的“效应器功能”。例示性的“效应器功能”包括C1q结合;CDC;Fc受体结合;ADCC;吞噬作用;细胞表面受体(例如B细胞受体;BCR)下调等。此类效应器功能一般要求Fc区与结合结构域(例如抗体可变域)联合,而且可以使用多种测定法来评估,例如本文所公开的那些。A "functional Fc region" possesses an "effector function" of the native sequence Fc region. Exemplary "effector functions" include C1q binding; CDC; Fc receptor binding; ADCC; phagocytosis; downregulation of cell surface receptors (eg, B cell receptor; BCR), and the like. Such effector functions generally require that the Fc region be associated with a binding domain (eg, an antibody variable domain) and can be assessed using a variety of assays, such as those disclosed herein.
术语“Fc区”在本文中用于定义免疫球蛋白重链的C端区域,所述区域包含至少一部分的恒定区。该术语包括天然序列Fc区和变体Fc区。在某些实施方案中,人IgG重链Fc区从Cys226或Pro230延伸至重链的羰基端。然而,Fc区的C端赖氨酸(Lys447)可以存在或者可以不存在。除非另外说明,Fc区或恒定区中的氨基酸残基的编号是根据EU编号系统,其也被称为EU索引,如在Kabat等,Sequences of Proteins of Immunological Interest,5th Ed.Public Health Service,National Institutes of Health,Bethesda,MD,1991中所述。The term "Fc region" is used herein to define a C-terminal region of an immunoglobulin heavy chain that comprises at least a portion of a constant region. The term includes native sequence Fc regions and variant Fc regions. In certain embodiments, the human IgG heavy chain Fc region extends from Cys226 or Pro230 to the carbonyl terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise indicated, the numbering of amino acid residues in the Fc region or constant region is based on the EU numbering system, which is also referred to as the EU index, as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National. Institutes of Health, Bethesda, MD, 1991.
“IgG形式的抗体”是指抗体的重链恒定区所属于的IgG形式。所有同一型的抗体的重链恒定区都是相同的,不同型的抗体之间的重链恒定区不同。例如,IgG1形式的抗体是指其重链恒定区Ig结构域为IgG1的Ig结构域。"Antibody in IgG form" refers to the IgG form to which the heavy chain constant region of an antibody belongs. The heavy chain constant regions of all antibodies of the same type are identical, and the heavy chain constant regions differ between different types of antibodies. For example, an antibody in the IgGl form refers to an Ig domain whose heavy chain constant region Ig domain is IgG1.
本文所述的术语“治疗剂”涵盖在预防或治疗肿瘤(例如癌症)和感染(例如慢性感染)中有效的任何物质,包括化疗剂、细胞毒性剂、疫苗、其它抗体、抗感染活性剂、小分子药物或免疫调节剂。The term "therapeutic agent" as used herein encompasses any substance that is effective in preventing or treating a tumor (eg, cancer) and an infection (eg, a chronic infection), including chemotherapeutic agents, cytotoxic agents, vaccines, other antibodies, anti-infective active agents, Small molecule drugs or immunomodulators.
术语“有效量”指本发明的抗体或片段或缀合物或组合物的这样的量或剂量,其以单一或多次剂量施用患者后,在需要治疗或预防的患者中产生预期效果。有效量可以由作为本领域技术人员的主治医师通过考虑以下多种因素来容易地确定:诸如哺乳动物的物种;它的大小、年龄和一般健康;涉及的具体疾病;疾病的程度或严重性;个体患者的应答;施用的具体抗体;施用模式;施用制剂的生物利用率特征;选择的给药方案;和任何伴随疗法的使用。The term "effective amount" refers to an amount or dose of an antibody or fragment or conjugate or composition of the invention that, when administered to a patient in single or multiple doses, produces the desired effect in a patient in need of treatment or prevention. An effective amount can be readily determined by the attending physician as a person skilled in the art by considering various factors such as the species of the mammal; its size, age and general health; the particular disease involved; the extent or severity of the disease; Response of an individual patient; specific antibody administered; mode of administration; bioavailability characteristics of the administered formulation; selected dosing regimen; and use of any concomitant therapy.
“治疗有效量”指以需要的剂量并持续需要的时间段,有效实现所需治疗结果的量。抗体或抗体片段或其缀合物或组合物的治疗有效量可以根据多种因素如疾病状态、个体的年龄、性别和重量和抗体或抗体部分在个体中激发所需反应的能力而变动。治疗有效量也是这样的一个量,其中抗体或抗体片段或其缀合物或组合物的任何有毒或有害作用不及治疗有益作用。相对于未治疗的对象,“治疗有效量”优选地抑制可度量参数(例如肿瘤生长率,肿瘤体积等)至少约20%、更优选地至少约40%、甚至更优选地至少约50%、60%或70%和仍更优选地至少约80%或90%。可以在预示人肿瘤中的功效的动物模型系统中评价化合物抑制可度量参数(例如,癌症)的能力。"Therapeutically effective amount" means an amount effective to achieve the desired therapeutic result at the desired dosage and for the period of time required. The therapeutically effective amount of an antibody or antibody fragment or conjugate or composition thereof can vary depending on a variety of factors, such as the disease state, the age, sex and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody fragment or conjugate or composition thereof are less than a therapeutically beneficial effect. A "therapeutically effective amount" preferably inhibits measurable parameters (eg, tumor growth rate, tumor volume, etc.) of at least about 20%, more preferably at least about 40%, even more preferably at least about 50%, relative to an untreated subject, 60% or 70% and still more preferably at least about 80% or 90%. The ability of a compound to inhibit measurable parameters (eg, cancer) can be evaluated in an animal model system that predicts efficacy in human tumors.
“预防有效量”指以需要的剂量并持续需要的时间段,有效实现所需预防结果的量。通常,由于预防性剂量在对象中在疾病较早阶段之前或在疾病较早阶段使用,故预防有效量将小于治疗有效量。By "prophylactically effective amount" is meant an amount effective to achieve the desired prophylactic result at the desired dosage and for the period of time required. Generally, a prophylactically effective amount will be less than a therapeutically effective amount since the prophylactic dose is administered to a subject prior to the earlier stage of the disease or at an earlier stage of the disease.
术语“宿主细胞”、“宿主细胞系”和“宿主细胞培养物”可交换地使用且是指其中引入外源核酸的细胞,包括这种细胞的后代。宿主细胞包括“转化体”和“转化的细胞”,其包括初级转化的细胞 和来源于其的后代,而不考虑传代的数目。后代在核酸内容上可能与亲本细胞不完全相同,而是可以包含突变。本文中包括在最初转化的细胞中筛选或选择的具有相同功能或生物学活性的突变体后代。The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to a cell into which an exogenous nucleic acid is introduced, including progeny of such a cell. Host cells include "transformants" and "transformed cells" which include primary transformed cells and progeny derived therefrom, regardless of the number of passages. Progeny may not be identical in nucleic acid content to the parental cell, but may contain mutations. Mutant progeny of the same function or biological activity selected or selected in the originally transformed cells are included herein.
“人抗体”指具有这样的氨基酸序列的抗体,所述氨基酸序列对应于下述抗体的氨基酸序列,所述抗体由人或人细胞生成或来源于非人来源,其利用人抗体库或其它人抗体编码序列。人抗体的这种定义明确排除包含非人抗原结合残基的人源化抗体。"Human antibody" refers to an antibody having an amino acid sequence corresponding to the amino acid sequence of an antibody produced by a human or human cell or derived from a non-human source, which utilizes a human antibody library or other human Antibody coding sequence. This definition of a human antibody specifically excludes a humanized antibody comprising a non-human antigen-binding residue.
“人源化”抗体是指包含来自非人CDR的氨基酸残基和来自人FR的氨基酸残基的嵌合抗体。在一些实施方案中,人源化抗体将包含基本上所有的至少一个、通常两个可变结构域,其中所有或基本上所有的CDR(例如,CDR)对应于非人抗体的那些,并且所有或基本上所有的FR对应于人抗体的那些。人源化抗体任选可以包含至少一部分的来源于人抗体的抗体恒定区。抗体(例如非人抗体)的“人源化形式”是指已经进行了人源化的抗体。A "humanized" antibody refers to a chimeric antibody comprising an amino acid residue from a non-human CDR and an amino acid residue from a human FR. In some embodiments, a humanized antibody will comprise substantially all of at least one, typically two variable domains, wherein all or substantially all of the CDRs (eg, CDRs) correspond to those of a non-human antibody, and all Or substantially all of the FRs correspond to those of human antibodies. The humanized antibody optionally can comprise at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody (eg, a non-human antibody) refers to an antibody that has been humanized.
术语“癌症”和“癌性”指向或描述哺乳动物中特征通常为细胞生长不受调节的生理疾患。The terms "cancer" and "cancerous" refer to or describe a physiological condition in a mammal that is typically characterized by unregulated cell growth.
术语“肿瘤”指所有赘生性(neoplastic)细胞生长和增殖,无论是恶性的还是良性的,及所有癌前(pre-cancerous)和癌性细胞和组织。术语“癌症”,“癌性”,“细胞增殖性病症”,“增殖性病症”和“肿瘤”在本文中提到时并不互相排斥。The term "tumor" refers to all neoplastic cell growth and proliferation, whether malignant or benign, and all pre-cancerous and cancerous cells and tissues. The terms "cancer", "cancerous", "cell proliferative disorder", "proliferative disorder" and "tumor" are not mutually exclusive when referred to herein.
术语“感染性疾病”是指病原体引发的疾病,包括例如病毒感染、细菌感染、真菌感染或者原生动物例如寄生虫感染。The term "infectious disease" refers to a disease caused by a pathogen, including, for example, a viral infection, a bacterial infection, a fungal infection, or a protozoan such as a parasitic infection.
术语“慢性感染”是指这样的感染,其中传染原(例如,病原体如病毒、细菌、原生动物例如寄生虫、真菌或诸如此类)已经在感染的宿主中诱导了免疫应答,但尚未如在急性感染过程中一样被从宿主中清除或消除。慢性感染可以是持续性的、潜伏性的或缓慢的。尽管急性感染(如流感)通常被免疫系统在数天或数周内解决,持续性的感染可以相对低的水平持续数月、数年、数十年或一生(例如,乙型肝炎)。相比之下,潜伏性的感染的特征是长期的无症状活动,被一段时间的迅速增加的高度感染和升高的病原体水平不时打断(例如单纯疱疹)。最后,缓慢感染的特征是疾病症状的逐渐和连续增加,诸如长期的潜伏期,随后在临床症状出现后是延长的和进展的临床过程开始。The term "chronic infection" refers to an infection in which an infectious agent (eg, a pathogen such as a virus, a bacterium, a protozoan such as a parasite, a fungus, or the like) has induced an immune response in an infected host, but has not yet been as acutely infected. It is also removed or eliminated from the host as in the process. Chronic infections can be persistent, latent or slow. Although acute infections (such as influenza) are usually resolved by the immune system in days or weeks, persistent infections can persist for months, years, decades, or lifetime (eg, hepatitis B) at relatively low levels. In contrast, latent infections are characterized by long-term asymptomatic activity, interrupted by high levels of rapid infection and elevated pathogen levels over time (eg, herpes simplex). Finally, slow infection is characterized by a gradual and continuous increase in disease symptoms, such as a long-term incubation period, followed by an onset of prolonged and progressive clinical processes following the onset of clinical symptoms.
“免疫缀合物”是与一个或多个其它物质(包括但不限于细胞毒性剂或标记)缀合的抗体。An "immunoconjugate" is an antibody that is conjugated to one or more other substances, including but not limited to cytotoxic agents or labels.
本文所使用的术语“标记”是指被直接或间接缀合或融合至试剂(诸如多核苷酸探针或抗体)并且促进其所缀合或融合的试剂的检测的化合物或组合物。标记本身可以是可检测的(例如,放射性同位素标记或萦光标记)或在酶促标记的情况下可以催化可检测的底物化合物或组合物的化学改变。术语旨在涵盖通过将可检测物质偶联(即,物理连接)至探针或抗体来直接标记探针或抗体以及通过与直接标记的另一种试剂反应来间接标记探针或抗体。间接标记的实例包括使用萦光标记的二级抗体进行的一级抗体的检测和具有生物素的DNA探针的末端标记,使得其可以用萦光标记的链霉抗生素蛋白来检测。The term "label" as used herein, refers to a compound or composition that is directly or indirectly conjugated or fused to an agent, such as a polynucleotide probe or antibody, and that facilitates detection of the agent to which it is conjugated or fused. The label itself may be detectable (eg, a radioisotope label or a fluorescent label) or, in the case of an enzymatic label, may catalyze a chemical change in a substrate compound or composition that is detectable. The term is intended to encompass the direct labeling of a probe or antibody by coupling (ie, physically linking) a detectable substance to a probe or antibody and indirectly labeling the probe or antibody by reaction with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end labeling of a biotinylated DNA probe such that it can be detected with a fluorescently labeled streptavidin protein.
“个体”或“受试者”包括哺乳动物。哺乳动物包括但不限于,家养动物(例如,牛,羊,猫,狗和马),灵长类动物(例如,人和非人灵长类动物如猴),兔,以及啮齿类动物(例如,小鼠和大鼠)。在一些实施方案中,个体或受试者是人。An "individual" or "subject" includes a mammal. Mammals include, but are not limited to, domesticated animals (eg, cows, sheep, cats, dogs, and horses), primates (eg, humans and non-human primates such as monkeys), rabbits, and rodents (eg, , mice and rats). In some embodiments, the individual or subject is a human.
“分离的”抗体是这样的抗体,其已经与其天然环境的组分分离。在一些实施方案中,将抗体纯化至超过95%或99%纯度,如通过例如电泳(例如,SDS-PAGE,等电聚焦(IEF),毛细管电泳)或层析(例如,离子交换或反相HPLC)确定的。对于用于评估抗体纯度的方法的综述,参见,例如,Flatman等,J.Chromatogr.B848:79-87(2007)。An "isolated" antibody is one which has been separated from components of its natural environment. In some embodiments, the antibody is purified to greater than 95% or 99% purity, such as by, for example, electrophoresis (eg, SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatography (eg, ion exchange or reversed phase) Determined by HPLC). For a review of methods for assessing antibody purity, see, for example, Flatman et al, J. Chromatogr. B848: 79-87 (2007).
“分离的编码抗GITR抗体或其片段的核酸”是指一个或多个核酸分子,其编码抗体重链或轻链(或其片段),包括在单一载体或分开的载体中的这样的核酸分子,以及存在于宿主细胞中的一个或多个位置处的这样的核酸分子。"Isolated nucleic acid encoding an anti-GITR antibody or fragment thereof" refers to one or more nucleic acid molecules encoding an antibody heavy or light chain (or a fragment thereof), including such nucleic acid molecules in a single vector or separate vectors. And such nucleic acid molecules present at one or more positions in the host cell.
如下进行序列之间序列同一性的计算。The calculation of sequence identity between sequences is performed as follows.
为确定两个氨基酸序列或两个核酸序列的同一性百分数,将所述序列出于最佳比较目的比对(例如,可以为了最佳比对而在第一和第二氨基酸序列或核酸序列之一或二者中引入空位或可以为比较目的而抛弃非同源序列)。在一个优选实施方案中,为比较目的,所比对的参考序列的长度是至少30%、 优选地至少40%、更优选地至少50%、60%和甚至更优选地至少70%、80%、90%、100%的参考序列长度。随后比较在对应氨基酸位置或核苷酸位置处的氨基酸残基或核苷酸。当第一序列中的位置由第二序列中对应位置处的相同氨基酸残基或核苷酸占据时,则所述分子在这个位置处是相同的。To determine the percent identity of two amino acid sequences or two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (eg, for optimal alignment, in the first and second amino acid sequences or nucleic acid sequences) Vacancies are introduced in one or both or non-homologous sequences can be discarded for comparison purposes. In a preferred embodiment, the length of the aligned reference sequences is at least 30%, preferably at least 40%, more preferably at least 50%, 60% and even more preferably at least 70%, 80% for comparison purposes. , 90%, 100% of the reference sequence length. The amino acid residues or nucleotides at the corresponding amino acid position or nucleotide position are then compared. When the position in the first sequence is occupied by the same amino acid residue or nucleotide at the corresponding position in the second sequence, then the molecule is identical at this position.
可以利用数学算法实现两个序列间的序列比较和同一性百分数的计算。在一个优选实施方案中,使用已经集成至GCG软件包的GAP程序中的Needlema和Wunsch((1970)J.Mol.Biol.48:444-453)算法(在http://www.gcg.com可获得),使用Blossum 62矩阵或PAM250矩阵和空位权重16、14、12、10、8、6或4和长度权重1、2、3、4、5或6,确定两个氨基酸序列之间的同一性百分数。在又一个优选的实施方案中,使用GCG软件包中的GAP程序(在http://www.gcg.com可获得),使用NWSgapdna.CMP矩阵和空位权重40、50、60、70或80和长度权重1、2、3、4、5或6,确定两个核苷酸序列之间的同一性百分数。特别优选的参数集合(和除非另外说明否则应当使用的一个参数集合)是采用空位罚分12、空位延伸罚分4和移码空位罚分5的Blossum 62评分矩阵。Mathematical algorithms can be used to achieve sequence comparisons and percent identity calculations between two sequences. In a preferred embodiment, the Needlema and Wunsch ((1970) J. Mol. Biol. 48: 444-453) algorithm in the GAP program that has been integrated into the GCG software package is used (at http://www.gcg.com) Obtained), using a Blossum 62 matrix or PAM250 matrix and vacancy weights 16, 14, 12, 10, 8, 6 or 4 and length weights 1, 2, 3, 4, 5 or 6, to determine between the two amino acid sequences Percent identity. In yet another preferred embodiment, the GAP program in the GCG software package (available at http://www.gcg.com) is used, using the NWSgapdna.CMP matrix and the vacancy weights of 40, 50, 60, 70 or 80 and The length weights 1, 2, 3, 4, 5 or 6, determine the percent identity between the two nucleotide sequences. A particularly preferred set of parameters (and a set of parameters that should be used unless otherwise stated) is a Blossum 62 scoring matrix with a gap penalty of 12, a gap extension penalty of 4, and a frameshift gap penalty of 5.
还可以使用PAM120加权余数表、空位长度罚分12,空位罚分4),利用已经并入ALIGN程序(2.0版)的E.Meyers和W.Miller算法,((1989)CABIOS,4:11-17)确定两个氨基酸序列或核苷酸序列之间的同一性百分数。You can also use the PAM120 weighted remainder table, the gap length penalty of 12, and the gap penalty 4), using the E. Meyers and W. Miller algorithms that have been incorporated into the ALIGN program (version 2.0), ((1989) CABIOS, 4:11- 17) Determine the percent identity between two amino acid sequences or nucleotide sequences.
额外地或备选地,可以进一步使用本文所述的核酸序列和蛋白质序列作为“查询序列”以针对公共数据库执行检索,以例如鉴定其他家族成员序列或相关序列。Additionally or alternatively, the nucleic acid sequences and protein sequences described herein can be further used as "query sequences" to perform searches against public databases to, for example, identify other family member sequences or related sequences.
术语“药用辅料”指与活性物质一起施用的稀释剂、佐剂(例如弗氏佐剂(完全和不完全的))、赋形剂、载体或稳定剂等。The term "pharmaceutically acceptable adjuvant" refers to a diluent, adjuvant (eg, Freund's adjuvant (complete and incomplete)), excipient, carrier or stabilizer, and the like, which are administered with the active substance.
术语“药物组合物”指这样的组合物,其以允许包含在其中的活性成分的生物学活性有效的形式存在,并且不包含对施用所述组合物的受试者具有不可接受的毒性的另外的成分。The term "pharmaceutical composition" refers to a composition that is present in a form that permits the biological activity of the active ingredient contained therein to be effective, and does not include additional toxicity to the subject to whom the composition is administered. Ingredients.
术语“组合产品”是指一种剂量单位形式的固定组合或非固定组合或用于组合施用的部分的试剂盒,其中两种或更多种治疗剂可以独立地在同一时间或在时间间隔内分开施用,尤其是在这些时间间隔允许组合伴侣展示协作,例如,协同效应时。术语“固定组合”是指本发明抗体和组合伴侣(例如其他治疗剂,例如抗PD1抗体、抗PDL1抗体或抗PDL2抗体)以单一实体或剂量的形式同时施用于患者。术语“非固定组合”意指本发明抗体和组合伴侣(例如其他治疗剂,例如抗PD1抗体、抗PDL1抗体或抗PDL2抗体)作为分开的实体同时、并行或依次施用于患者,没有特定的时间限制,其中这样的施用提供了患者体内两种化合物的治疗有效水平。后者也适用于鸡尾酒疗法,例如施用三种或更多种治疗剂。在一个优选的实施方案中,药物组合是非固定组合。The term "combination product" refers to a kit of fixed or non-fixed combinations in the form of a dosage unit or a portion for administration in combination, wherein two or more therapeutic agents can be independently at the same time or within a time interval Separate administration, especially when these time intervals allow the combined partner to exhibit collaboration, for example, synergistic effects. The term "fixed combination" refers to the simultaneous administration of an antibody of the invention and a combination partner (eg, other therapeutic agents, eg, an anti-PD1 antibody, an anti-PDL1 antibody, or an anti-PDL2 antibody) to a patient in a single entity or dosage form. The term "non-fixed combination" means that the antibody of the invention and a combination partner (eg, other therapeutic agents, eg, an anti-PD1 antibody, an anti-PDL1 antibody, or an anti-PDL2 antibody) are administered to the patient simultaneously, in parallel, or sequentially as separate entities, without a specific time. Limitations, wherein such administration provides a therapeutically effective level of both compounds in a patient. The latter also applies to cocktail therapy, for example the administration of three or more therapeutic agents. In a preferred embodiment, the pharmaceutical combination is a non-fixed combination.
术语“组合疗法”是指施用两种或更多种治疗剂以治疗如本公开所述的癌症或感染。这种施用包括以基本上同时的方式共同施用这些治疗剂,例如以具有固定比例的活性成分的单一胶囊。或者,这种施用包括对于各个活性成分在多种或在分开的容器(例如片剂、胶囊、粉末和液体)中的共同施用。粉末和/或液体可以在施用前重构或稀释至所需剂量。此外,这种施用还包括以大致相同的时间或在不同的时间以顺序的方式使用每种类型的治疗剂。在任一情况下,治疗方案将提供药物组合在治疗本文所述的病症或病状中的有益作用。The term "combination therapy" refers to the administration of two or more therapeutic agents to treat a cancer or infection as described in the present disclosure. Such administration involves co-administering these therapeutic agents in a substantially simultaneous manner, for example, as a single capsule having a fixed ratio of active ingredient. Alternatively, such administration includes co-administration of the individual active ingredients in multiple or separate containers (e.g., tablets, capsules, powders, and liquids). The powder and/or liquid can be reconstituted or diluted to the desired dose prior to administration. Moreover, such administration also includes the use of each type of therapeutic agent in a sequential manner at substantially the same time or at different times. In either case, the treatment regimen will provide a beneficial effect of the combination of drugs in treating the condition or condition described herein.
用于本文时,“治疗”指减缓、中断、阻滞、缓解、停止、降低、或逆转已存在的症状、病症、病况或疾病的进展或严重性。As used herein, "treating" refers to slowing, interrupting, arresting, ameliorating, stopping, reducing, or reversing the progression or severity of an existing symptom, disorder, condition, or disease.
用于本文时,“预防”包括对疾病或病症或特定疾病或病症的症状的发生或发展的抑制。在一些实施方式中,具有癌症家族病史的受试者是预防性方案的候选。通常,在癌症的背景中,术语“预防”是指在癌症的病征或症状发生前,特别是在具有癌症风险的受试者中发生前的药物施用。As used herein, "prevention" includes inhibition of the occurrence or progression of a disease or condition or a symptom of a particular disease or condition. In some embodiments, a subject with a family history of cancer is a candidate for a prophylactic regimen. Generally, in the context of cancer, the term "prevention" refers to the administration of a drug prior to the onset of a symptom or symptom of a cancer, particularly in a subject at risk for cancer.
术语“抗感染活性剂”包括在施用浓度和给药间隔下特异性抑制或消除微生物生长但对宿主不致命的任何分子,所述微生物诸如病毒、细菌、真菌或原生动物,例如寄生虫。用于本文时,术语抗感染活性剂包括抗生素、抗菌剂、抗病毒剂、抗真菌剂和抗原生动物剂。在一个具体方面中,抗感染活性剂在施用浓度和给药间隔对宿主是无毒的。The term "anti-infective active agent" includes any molecule that specifically inhibits or eliminates the growth of microorganisms at the concentration of administration and the interval of administration, but is not lethal to the host, such as viruses, bacteria, fungi or protozoa, such as parasites. As used herein, the term anti-infective active agent includes antibiotics, antibacterial agents, antiviral agents, antifungal agents, and antiprotozoal agents. In a specific aspect, the anti-infective active agent is non-toxic to the host at the concentration of administration and the interval of administration.
抗细菌的抗感染活性剂或抗菌剂可广泛的分类为杀菌的(即,直接杀死)或抑菌的(即,阻止分裂)。抗菌的抗感染活性剂可进一步再分类为窄谱抗菌剂(即,仅影响小类细菌亚型,例如,革兰氏 阴性等)或广谱抗菌剂(即,影响广泛种类)。Antibacterial anti-infective active agents or antibacterial agents can be broadly classified as either bactericidal (i.e., directly killed) or bacteriostatic (i.e., preventing division). Antibacterial anti-infective active agents can be further reclassified as narrow-spectrum antibacterial agents (i.e., affecting only subtypes of bacterial subtypes, e.g., Gram-negative, etc.) or broad-spectrum antibacterial agents (i.e., affecting a wide variety of species).
术语“抗病毒剂”包括抑制或消除病毒生长、致病和/或存活的任何物质。The term "antiviral agent" includes any substance that inhibits or eliminates the growth, pathogenesis, and/or survival of a virus.
术语“抗真菌剂”包括抑制或消除真菌生长、致病和/或存活的任何物质。The term "antifungal agent" includes any substance that inhibits or eliminates the growth, pathogenesis and/or survival of fungi.
术语“抗原生动物剂”包括抑制或消除原生动物生物体(例如寄生虫)生长、发病和/或存活的任何物质。The term "antigenic animal agent" includes any substance that inhibits or eliminates the growth, morbidity and/or survival of a protozoan organism (eg, a parasite).
术语“载体”当在本文中使用时是指能够增殖与其相连的另一个核酸的核酸分子。该术语包括作为自我复制核酸结构的载体以及结合到已经引入其的宿主细胞的基因组中的载体。一些载体能够指导与其可操作相连的核酸的表达。这样的载体在本文中被称为“表达载体”。The term "vector," as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes a vector that is a self-replicating nucleic acid structure and a vector that binds to the genome of a host cell into which it has been introduced. Some vectors are capable of directing the expression of a nucleic acid to which they are operably linked. Such vectors are referred to herein as "expression vectors."
“受试者/患者样品”指从患者或受试者得到的细胞或流体的集合。组织或细胞样品的来源可以是实体组织,像来自新鲜的、冷冻的和/或保存的器官或组织样品或活检样品或穿刺样品;血液或任何血液组分;体液,诸如脑脊液、羊膜液(羊水)、腹膜液(腹水)、或间隙液;来自受试者的妊娠或发育任何时间的细胞。组织样品可能包含在自然界中天然不与组织混杂的化合物,诸如防腐剂、抗凝剂、缓冲剂、固定剂、营养物、抗生素、等等。肿瘤样品的例子在本文中包括但不限于肿瘤活检、细针吸出物、支气管灌洗液、胸膜液(胸水)、痰液、尿液、手术标本、循环中的肿瘤细胞、血清、血浆、循环中的血浆蛋白质、腹水、衍生自肿瘤或展现出肿瘤样特性的原代细胞培养物或细胞系,以及保存的肿瘤样品,诸如福尔马林固定的、石蜡包埋的肿瘤样品或冷冻的肿瘤样品。"Subject/patient sample" refers to a collection of cells or fluids obtained from a patient or subject. The source of the tissue or cell sample may be a solid tissue, such as from a fresh, frozen and/or preserved organ or tissue sample or a biopsy sample or a puncture sample; blood or any blood component; body fluids such as cerebrospinal fluid, amniotic fluid (amniotic fluid) ), peritoneal fluid (ascites), or interstitial fluid; cells from the subject's pregnancy or development at any time. Tissue samples may contain compounds that are naturally not intermixed with tissue in nature, such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, and the like. Examples of tumor samples include, but are not limited to, tumor biopsy, fine needle aspirate, bronchial lavage fluid, pleural fluid (thoracic fluid), sputum, urine, surgical specimens, circulating tumor cells, serum, plasma, circulation Plasma protein, ascites, primary cell cultures or cell lines derived from tumors or exhibiting tumor-like properties, and preserved tumor samples, such as formalin-fixed, paraffin-embedded tumor samples or frozen tumors sample.
II.抗体II. Antibody
在一些实施方案中,本发明的抗GITR抗体或其片段以高亲和力结合GITR(例如人GITR或食蟹猴GITR),例如,以以下平衡解离常数(K D)与GITR结合,所述K D小于大约10nM,优选地,小于或等于大约7nM,更优选地小于或等于大约6nM,更优选地小于或等于大约5nM、4.9nM、4.8nM、4.7nM、4.6nM、4.5nM、4nM、或3nM,最优选地,所述K D小于或等于大约2.5nM、2.4nM、2.3nM、2.2nM、2.1nM。在一些实施方案中,本发明的抗GITR抗体以1nM-6nM,优选地1.5nM-5nM、1.7nM-5nM、1.8nM-5nM、1.9nM-5nM的K D结合GITR。在一些实施方案中,GITR为人GITR。在一些实施方案中,抗体结合亲和力是使用生物光干涉测定法(例如Fortebio亲和测量)测定的。 In some embodiments, the anti-GITR antibody or fragment of the invention binds with high affinity GITR (GITR e.g. human or cynomolgus GITR), for example, in the following equilibrium dissociation constant (K D) in combination with GITR, the K D is less than about 10 nM, preferably, less than or equal to about 7 nM, more preferably less than or equal to about 6 nM, more preferably less than or equal to about 5 nM, 4.9 nM, 4.8 nM, 4.7 nM, 4.6 nM, 4.5 nM, 4 nM, or of 3 nM, and most preferably, the K D of less than or equal to about 2.5nM, 2.4nM, 2.3nM, 2.2nM, 2.1nM. In some embodiments, the anti-GITR antibody of the present invention to 1nM-6nM, preferably 1.5nM-5nM, 1.7nM-5nM, 1.8nM-5nM, K D 1.9nM-5nM GITR binding. In some embodiments, the GITR is a human GITR. In some embodiments, the antibody binding affinity is determined using a bio-optical interference assay (eg, Fortebio affinity measurement).
在一些实施方案中,本发明的抗体或其片段结合表达GITR的细胞,例如,以小于或等于大约2nM、1.9nM、1.5nM、1nM、0.9nM、0.8nM、0.7nM、0.6nM、0.5nM、0.4nM的EC50。在一些实施方案中,所述结合用流式细胞术(例如FACS)测定。在一些实施方案中,表达GITR的细胞为表达GITR的CHO细胞(例如CHO-S细胞)。在一些实施方案中,GITR为人GITR或食蟹猴GITR。In some embodiments, an antibody or fragment thereof of the invention binds to a cell expressing GITR, eg, at less than or equal to about 2 nM, 1.9 nM, 1.5 nM, 1 nM, 0.9 nM, 0.8 nM, 0.7 nM, 0.6 nM, 0.5 nM. , EC50 of 0.4nM. In some embodiments, the binding is determined by flow cytometry (eg, FACS). In some embodiments, the GITR expressing cell is a CHO cell (eg, CHO-S cell) that expresses GITR. In some embodiments, the GITR is a human GITR or a cynomolgus monkey GITR.
在一些实施方案中,本发明的抗体或其片段通过与细胞表面的GITR分子结合而激活GITR下游的NF-kappaB信号通路。在一些实施方案中,本发明的抗体或其片段激活能力优于已知的抗GITR抗体,例如专利申请US20130183321A1(GITR,INC.(Cambridge,MA,US))中报道的GITR抗体。在一个实施方案中,已知的抗GITR抗体分子的轻链和重链序列在US20130183321A1中分别为SEQ ID NO:44和SEQ ID NO:54。在一些实施方案中,本发明的GITR抗体分子相比对照抗体分子,能够显著有效激活NF kappa B信号通路。在一些实施方案中,所述激活是通过萦光素酶检测获得的。In some embodiments, an antibody or fragment thereof of the invention activates the NF-kappaB signaling pathway downstream of the GITR by binding to a GITR molecule on the cell surface. In some embodiments, an antibody of the invention or a fragment thereof has a higher ability to activate than a known anti-GITR antibody, such as the GITR antibody reported in patent application US20130183321A1 (GITR, INC. (Cambridge, MA, US)). In one embodiment, the light and heavy chain sequences of known anti-GITR antibody molecules are SEQ ID NO: 44 and SEQ ID NO: 54, respectively, in US20130183321A1. In some embodiments, a GITR antibody molecule of the invention is capable of significantly and efficiently activating the NF kappa B signaling pathway compared to a control antibody molecule. In some embodiments, the activation is obtained by luciferase assay.
在一些实施方案中,本发明抗体或其片段提高效应T细胞功能,例如激活效应T细胞。在一些实施方案中,本发明的抗体或其片段能够增强效应T细胞的增殖。在一些实施方案中,本发明的抗体或其片段提高干扰素(例如IFN-γ)分泌/表达。在一些实施方案中,本发明的抗体或其片段提高白细胞介素(例如IL-2)分泌/表达。在一些实施方案中,T细胞是CD4T细胞。In some embodiments, an antibody of the invention or a fragment thereof enhances effector T cell function, such as activating effector T cells. In some embodiments, an antibody or fragment thereof of the invention is capable of enhancing proliferation of effector T cells. In some embodiments, an antibody or fragment thereof of the invention increases the secretion/expression of interferon (eg, IFN-[gamma]). In some embodiments, an antibody or fragment thereof of the invention increases secretion/expression of interleukin (eg, IL-2). In some embodiments, the T cell is a CD4 T cell.
在一些实施方案中,本发明的GITR抗体或其片段能够激活调节T细胞(例如Treg细胞)中的GITR信号通路,进而通过介导ADCC效应消除细胞。由此,一方面解除效应T细胞自身的抑制信号,增强CD4T细胞和/或CD8T细胞的活性,另一个方面消除调节T细胞对效应T细胞的抑制作用,从而最大程度地激活效应T细胞,增强针对肿瘤细胞的免疫应答反应。在一些实施方案中,通过检测NF-AT信号激活来检测抗体的ADCC活性。在一些实施方案中,所述NF-AT信号的激活由萦光报告 基因的表达体现。在一些实施方案中,本发明的GITR抗体分子相比已知的抗GITR抗体分子,能够显著有效激活NF-AT信号通路。在一些实施方案中,本发明的GITR抗体分子相比已知的抗GITR抗体分子,能够显著有效介导ADCC效应。在一些实施方案中,本发明的GITR抗体分子相比已知的抗GITR抗体分子,能够显著有效抑制或消除调节T细胞。在一些实施方案中,本发明的效应T细胞是CD4T细胞。在一些实施方案中,已知的抗GITR抗体分子是例如专利申请US20130183321A1(GITR,INC.(Cambridge,MA,US))中报道的抗GITR抗体。在一个实施方案中,已知的抗GITR抗体分子的轻链和重链序列在US20130183321A1中分别为SEQ ID NO:44和SEQ ID NO:54。In some embodiments, a GITR antibody or fragment thereof of the invention is capable of activating a GITR signaling pathway in a regulatory T cell (eg, a Treg cell), thereby eliminating cells by mediating the ADCC effect. Thus, on the one hand, the inhibitory signal of the effector T cell itself is released, the activity of the CD4 T cell and/or the CD8 T cell is enhanced, and the other aspect is to eliminate the inhibitory effect of the T cell on the effector T cell, thereby maximally activating the effector T cell and enhancing An immune response to tumor cells. In some embodiments, the ADCC activity of the antibody is detected by detecting NF-AT signaling activation. In some embodiments, activation of the NF-AT signal is manifested by expression of a fluorescent reporter gene. In some embodiments, the GITR antibody molecules of the invention are capable of significantly and efficiently activating the NF-AT signaling pathway compared to known anti-GITR antibody molecules. In some embodiments, the GITR antibody molecules of the invention are capable of significantly and efficiently mediating ADCC effects compared to known anti-GITR antibody molecules. In some embodiments, a GITR antibody molecule of the invention is capable of significantly inhibiting or eliminating regulatory T cells compared to known anti-GITR antibody molecules. In some embodiments, the effector T cells of the invention are CD4 T cells. In some embodiments, a known anti-GITR antibody molecule is an anti-GITR antibody as reported in, for example, patent application US20130183321A1 (GITR, INC. (Cambridge, MA, US)). In one embodiment, the light and heavy chain sequences of known anti-GITR antibody molecules are SEQ ID NO: 44 and SEQ ID NO: 54, respectively, in US20130183321A1.
在一些实施方案中,本发明的抗体或其片段(任选地与治疗方式和/或其它治疗剂,例如PD-1轴结合拮抗剂组合)能够预防或治疗肿瘤。在一些实施方案中,本发明的抗体或其片段能够用于抑制或减小肿瘤生长(例如减小肿瘤体积)。在一些实施方案中,本发明的抗体或其片段还能够用于维持肿瘤患者体重。在一些实施方案中,本发明的抗体或其片段能够与治疗方式和/或其它治疗剂,例如PD1-轴结合拮抗剂组合抑制或减小肿瘤生长(例如减小肿瘤体积)。在一些实施方案中,本发明的抗体或其片段能够与治疗方式和/或其它治疗剂,例如PD1-轴结合拮抗剂组合维持肿瘤患者体重。在一些实施方案中,本发明的抗体或其片段(任选地与治疗方式和/或其它治疗剂,例如PD-1轴结合拮抗剂组合)能够抑制或减小肿瘤生长(例如减小肿瘤体积),同时不影响肿瘤患者体重。优选地,肿瘤为胃肠道肿瘤。优选地,肿瘤为癌症,例如胃肠道癌症,例如胃癌、直肠癌、结肠癌、结肠直肠癌等。在一些实施方案中,本发明的抗体或其片段(任选地与PD-1轴结合拮抗剂组合)能够预防或治疗感染,例如慢性感染,包括细菌感染、真菌感染、病毒感染、原生动物感染等。In some embodiments, an antibody or fragment thereof of the invention (optionally in combination with a therapeutic modality and/or other therapeutic agent, such as a PD-1 axis binding antagonist) is capable of preventing or treating a tumor. In some embodiments, an antibody or fragment thereof of the invention can be used to inhibit or reduce tumor growth (eg, reduce tumor volume). In some embodiments, an antibody or fragment thereof of the invention can also be used to maintain body weight in a tumor patient. In some embodiments, an antibody or fragment thereof of the invention is capable of inhibiting or reducing tumor growth (eg, reducing tumor volume) in combination with a therapeutic modality and/or other therapeutic agent, such as a PD1-axis binding antagonist. In some embodiments, an antibody or fragment thereof of the invention is capable of maintaining the weight of a tumor patient in combination with a therapeutic modality and/or other therapeutic agent, such as a PD1-axis binding antagonist. In some embodiments, an antibody of the invention or a fragment thereof (optionally in combination with a therapeutic modality and/or other therapeutic agent, such as a PD-1 axis binding antagonist) is capable of inhibiting or reducing tumor growth (eg, reducing tumor volume) ), while not affecting the weight of cancer patients. Preferably, the tumor is a gastrointestinal tumor. Preferably, the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like. In some embodiments, an antibody of the invention or a fragment thereof (optionally in combination with a PD-1 axis binding antagonist) is capable of preventing or treating an infection, such as a chronic infection, including a bacterial infection, a fungal infection, a viral infection, a protozoal infection. Wait.
在一些实施方案中,本发明的抗GITR抗体或其抗原结合片段包含重链可变区(VH),其中所述VH包含In some embodiments, an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region (VH), wherein said VH comprises
(i)表B所列任一抗体的VH中所含的三个互补决定区域(CDR),或(i) three complementarity determining regions (CDRs) contained in the VH of any of the antibodies listed in Table B, or
(ii)相对于(i)的序列,在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换)的序列。(ii) a sequence comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions) on the three CDR regions relative to the sequence of (i).
在一些实施方案中,本发明的抗GITR抗体或其抗原结合片段包含轻链可变区(VL),其中所述VL包含:In some embodiments, an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a light chain variable region (VL), wherein the VL comprises:
(i)表B所列任一抗体的VL中所含的三个互补决定区域(CDR);或(i) three complementarity determining regions (CDRs) contained in the VL of any of the antibodies listed in Table B;
(ii)相对于(i)的序列,在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换)的序列。(ii) a sequence comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions) on the three CDR regions relative to the sequence of (i).
在一些实施方案中,本发明的抗GITR抗体或其抗原结合片段包含重链可变区VH和轻链可变区VL,其中In some embodiments, an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region VH and a light chain variable region VL, wherein
(a)所述VH包含(a) The VH contains
(i)表B所列任一抗体的VH中所含的三个互补决定区域(CDR),或(i) three complementarity determining regions (CDRs) contained in the VH of any of the antibodies listed in Table B, or
(ii)相对于(i)的序列,在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换)的序列;和/或(ii) a sequence comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions) on said three CDR regions relative to the sequence of (i); and / or
(b)所述VL包含:(b) The VL contains:
(i)表B所列任一抗体的VL中所含的三个互补决定区域(CDR);或(i) three complementarity determining regions (CDRs) contained in the VL of any of the antibodies listed in Table B;
(ii)相对于(i)的序列,在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换)的序列。(ii) a sequence comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably conservative substitutions) on the three CDR regions relative to the sequence of (i).
在优选的实施方案中,VH包含选自SEQ ID NO:17、18、19或20所示的氨基酸序列,或由所述氨基酸序列组成。In a preferred embodiment, the VH comprises or consists of an amino acid sequence selected from the group consisting of SEQ ID NO: 17, 18, 19 or 20.
在优选的实施方案中,VL包含选自SEQ ID NO:21、22、23或24所示的氨基酸序列,或由所述氨基酸序列组成。In a preferred embodiment, VL comprises or consists of an amino acid sequence selected from the group consisting of SEQ ID NO: 21, 22, 23 or 24.
在优选的实施方案中,本发明抗GITR抗体或其抗原结合片段包含In a preferred embodiment, the anti-GITR antibody or antigen-binding fragment thereof of the invention comprises
(i)如SEQ ID NO:17或19所示的重链可变区的3个互补决定区HCDR,以及如SEQ ID NO:21或23所示的轻链可变区的3个互补决定区LCDR,或者(i) three complementarity determining region HCDRs of the heavy chain variable region set forth in SEQ ID NO: 17 or 19, and three complementarity determining regions of the light chain variable region set forth in SEQ ID NO: 21 or LCDR, or
(ii)如SEQ ID NO:18或20所示的重链可变区的3个互补决定区HCDR,以及如SEQ ID NO:22或24所示的轻链可变区的3个互补决定区LCDR。(ii) three complementarity determining region HCDRs of the heavy chain variable region set forth in SEQ ID NO: 18 or 20, and three complementarity determining regions of the light chain variable region set forth in SEQ ID NO: 22 or LCDR.
在一些实施方案中,本发明的抗GITR抗体或其抗原结合片段包含重链可变区(VH)和/或轻链可变区(VL),其中In some embodiments, an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region (VH) and/or a light chain variable region (VL), wherein
(i)所述VH包含互补决定区域(CDR)HCDR1、HCDR2和HCDR3,其中HCDR1包含选自SEQ ID NO:1、2、3或4的氨基酸序列,或由所述氨基酸序列组成,或者HCDR1包含与选自SEQ ID NO:1、2、3或4的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;HCDR2包含选自SEQ ID NO:5或6的氨基酸序列,或由所述氨基酸序列组成,或者HCDR2包含与选自SEQ ID NO:5或6的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;HCDR3包含选自SEQ ID NO:7或8的氨基酸序列或由所述氨基酸序列组成,或者HCDR3包含与选自SEQ ID NO:7或8的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;(i) said VH comprises a complementarity determining region (CDR) HCDR1, HCDR2 and HCDR3, wherein HCDR1 comprises or consists of the amino acid sequence selected from SEQ ID NO: 1, 2, 3 or 4, or HCDR1 comprises An amino acid sequence having one, two or three alterations (preferably amino acid substitutions, preferably conservative substitutions) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 1, 2, 3 or 4; HCDR2 comprising an SEQ ID NO: 5 Or the amino acid sequence of 6 or consists of the amino acid sequence, or HCDR2 comprises one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) compared to the amino acid sequence selected from SEQ ID NO: 5 or 6. Amino acid sequence; HCDR3 comprises or consists of the amino acid sequence selected from SEQ ID NO: 7 or 8, or HCDR3 comprises one, two or more than the amino acid sequence selected from SEQ ID NO: 7 or 8. An amino acid sequence of three alterations (preferably amino acid substitutions, preferably conservative substitutions);
和/或and / or
(ii)其中所述VL包含互补决定区域(CDR)LCDR1、LCDR2和LCDR3,其中LCDR1包含选自SEQ ID NO:9或10的氨基酸序列或由所述氨基酸序列组成,或者LCDR1包含与选自SEQ ID NO:9或10的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;LCDR2包含选自SEQ ID NO:11、12、13或14的氨基酸序列或由所述氨基酸序列组成,或者LCDR2包含与选自SEQ ID NO:11、12、13或14的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;LCDR3包含选自SEQ ID NO:15或16的氨基酸序列或由所述氨基酸序列组成,或者LCDR3包含与选自SEQ ID NO:15或16的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列。(ii) wherein said VL comprises a complementarity determining region (CDR) LCDR1, LCDR2 and LCDR3, wherein LCDR1 comprises or consists of the amino acid sequence selected from SEQ ID NO: 9 or 10, or LCDR1 comprises and is selected from the group consisting of ID NO: amino acid sequence of 9 or 10 having an amino acid sequence of one, two or three changes (preferably amino acid substitution, preferably conservative substitution); LCDR2 comprising an amino acid selected from the group consisting of SEQ ID NO: 11, 12, 13 or 14. The sequence consists either of said amino acid sequence, or LCDR2 comprises one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) compared to an amino acid sequence selected from SEQ ID NO: 11, 12, 13 or 14. Amino acid sequence; LCDR3 comprises or consists of the amino acid sequence selected from SEQ ID NO: 15 or 16, or LCDR3 comprises one, two or three compared to the amino acid sequence selected from SEQ ID NO: 15 or 16. Amino acid sequence that changes (preferably amino acid substitutions, preferably conservative substitutions).
在优选的实施方案中,本发明提供抗GITR抗体或其抗原结合片段,其包含重链可变区(VH)和/或轻链可变区(VL),其中In a preferred embodiment, the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain variable region (VH) and/or a light chain variable region (VL), wherein
(a)所述VH包含(a) The VH contains
(i)表A所示的HCDR1、HCDR2和HCDR3的组合;或(i) a combination of HCDR1, HCDR2 and HCDR3 as shown in Table A; or
(ii)(i)的HCDR组合的变体,所述变体在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换);(ii) a variant of the HCDR combination of (i) comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes (preferably amino acid substitutions, preferably) on said three CDR regions Conservative substitution);
和/或and / or
(ii)所述VL包含(ii) the VL contains
(i)表A所示的LCDR1、LCDR2和LCDR3的组合;或者(i) a combination of LCDR1, LCDR2 and LCDR3 as shown in Table A; or
(ii)(i)的LCDR组合的变体,所述变体在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换)。(ii) a variant of the LCDR combination of (i), said variant comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes on said three CDR regions (preferably amino acid substitution, preferably Conservative substitution).
在优选的实施方案中,本发明提供抗GITR抗体或其抗原结合片段,其包含重链可变区(VH)和/或轻链可变区(VL),其中所述VH包含互补决定区域(CDR)HCDR1、HCDR2和HCDR3,所述VL包含(CDR)LCDR1、LCDR2和LCDR3,其中所述抗体或其抗原结合片段所包含的HCDR1、HCDR2、HCDR3、LCDR1、LCDR2和LCDR3的组合如下表(表A)所示:In a preferred embodiment, the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain variable region (VH) and/or a light chain variable region (VL), wherein said VH comprises a complementarity determining region ( CDR) HCDR1, HCDR2 and HCDR3 comprising (CDR) LCDR1, LCDR2 and LCDR3, wherein the combination of HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3 comprised by the antibody or antigen-binding fragment thereof is as follows (Table A):
表A:本发明抗体或其抗原结合片段中HCDR1、HCDR2、HCDR3、LCDR1、LCDR2和LCDR3的示例性组合Table A: Exemplary combinations of HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 in an antibody or antigen-binding fragment thereof of the present invention
Figure PCTCN2019083241-appb-000002
Figure PCTCN2019083241-appb-000002
Figure PCTCN2019083241-appb-000003
Figure PCTCN2019083241-appb-000003
在一些实施方案中,本发明的抗GITR抗体或其抗原结合片段包含重链可变区VH和/或轻链可变区VL,其中,In some embodiments, an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain variable region VH and/or a light chain variable region VL, wherein
(a)重链可变区VH(a) Heavy chain variable region VH
(i)包含与选自SEQ ID NO:17、18、19或20的氨基酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;或者(i) comprising at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or an amino acid sequence selected from the group consisting of SEQ ID NO: 17, 18, 19 or 20. 99% identical amino acid sequence or consists of; or
(ii)包含选自SEQ ID NO:17、18、19或20的氨基酸序列或由其组成;或者(ii) comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NO: 17, 18, 19 or 20;
(iii)包含与选自SEQ ID NO:17、18、19或20的氨基酸序列相比具有1个或多个(优选不超过10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在CDR区中;(iii) comprising one or more (preferably no more than 10, more preferably no more than 5, 4, 3, 2, 1) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 17, 18, 19 or 20. Amino acid sequence of an amino acid change (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, said amino acid change does not occur in the CDR region;
和/或and / or
(b)轻链可变区VL(b) Light chain variable region VL
(i)包含与选自SEQ ID NO:21、22、23或24的氨基酸序列具有至少90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;(i) comprising at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or an amino acid sequence selected from the group consisting of SEQ ID NO: 21, 22, 23 or 99% identical amino acid sequence or consists of;
(ii)包含选自SEQ ID NO:21、22、23或24的氨基酸序列或由其组成;或者(ii) comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NO: 21, 22, 23 or 24;
(iii)包含与选自SEQ ID NO:21、22、23或24的氨基酸序列相比具有1个或多个(优选不超过10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在CDR区中。(iii) comprising one or more (preferably no more than 10, more preferably no more than 5, 4, 3, 2, 1) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 21, 22, 23 or 24. The amino acid sequence of an amino acid change (preferably an amino acid substitution, more preferably an amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR regions.
在优选的实施方案中,本发明提供抗GITR抗体或其抗原结合片段,其包含重链可变区(VH)和轻链可变区(VL),其中所述抗体或其抗原结合片段所包含的重链可变区VH和轻链可变区VL的组合如下表(表B)所示:In a preferred embodiment, the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain variable region (VH) and a light chain variable region (VL), wherein the antibody or antigen-binding fragment thereof comprises The combination of the heavy chain variable region VH and the light chain variable region VL is shown in the following table (Table B):
表B:本发明抗体或其抗原结合片段中重链可变区VH和轻链可变区VL的示例性组合Table B: Exemplary combinations of heavy chain variable region VH and light chain variable region VL in an antibody or antigen-binding fragment thereof of the invention
Figure PCTCN2019083241-appb-000004
Figure PCTCN2019083241-appb-000004
在一些实施方案中,本发明的抗GITR抗体或其抗原结合片段包含重链和/或轻链,其中In some embodiments, an anti-GITR antibody or antigen-binding fragment thereof of the invention comprises a heavy chain and/or a light chain, wherein
(a)重链(a) heavy chain
(i)包含与选自SEQ ID NO:25、26、27或28的氨基酸序列具有至少85%、90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;(i) comprising at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, and an amino acid sequence selected from the group consisting of SEQ ID NO: 25, 26, 27 or a 98% or 99% identity amino acid sequence or consists of;
(ii)包含选自SEQ ID NO:25、26、27或28的氨基酸序列或由其组成;或者(ii) comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NO: 25, 26, 27 or 28;
(iii)包含与选自SEQ ID NO:25、26、27或28的氨基酸序列相比具有1个或多个(优选不超过20个或10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在重链的CDR区中,更优选地,所述氨基酸改变不发生在重链可变区中;(iii) comprising one or more (preferably no more than 20 or 10, more preferably no more than 5, 4, 3, 2) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 25, 26, 27 or 28. Amino acid sequence of amino acid change (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR region of the heavy chain, and more preferably, the amino acid change does not occur in Heavy chain variable region;
和/或and / or
(b)轻链(b) Light chain
(i)包含与选自SEQ ID NO:29、30、31或32的氨基酸序列具有至少85%、90%、91%、92%、93%、94%、95%、96%、97%、98%或99%同一性的氨基酸序列或由其组成;(i) comprising at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, and an amino acid sequence selected from the group consisting of SEQ ID NO: 29, 30, 31 or 32, a 98% or 99% identity amino acid sequence or consists of;
(ii)包含选自SEQ ID NO:29、30、31或32的氨基酸序列或由其组成;或者(ii) comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NO: 29, 30, 31 or 32;
(iii)包含与选自SEQ ID NO:29、30、31或32的氨基酸序列相比具有1个或多个(优选不超过 20个或10个,更优选不超过5、4、3、2、1个)的氨基酸改变(优选氨基酸置换,更优选氨基酸保守置换)的氨基酸序列,优选地,所述氨基酸改变不发生在轻链的CDR区中,更优选地,所述氨基酸改变不发生在轻链可变区中。(iii) comprising one or more (preferably no more than 20 or 10, more preferably no more than 5, 4, 3, 2) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 29, 30, 31 or 32 Amino acid sequence of amino acid change (preferably amino acid substitution, more preferably amino acid conservative substitution), preferably, the amino acid change does not occur in the CDR region of the light chain, and more preferably, the amino acid change does not occur in Light chain variable region.
在优选的实施方案中,本发明提供抗GITR抗体或其抗原结合片段,其包含重链和轻链,其中所述抗体或其抗原结合片段所包含的重链和轻链的组合如下表(表C)所示:In a preferred embodiment, the invention provides an anti-GITR antibody or antigen-binding fragment thereof comprising a heavy chain and a light chain, wherein the combination of the heavy chain and the light chain comprised by the antibody or antigen-binding fragment thereof is as follows (Table C):
表C:本发明抗体或其抗原结合片段中重链和轻链的示例性组合Table C: Exemplary combinations of heavy and light chains in an antibody or antigen-binding fragment thereof of the invention
Figure PCTCN2019083241-appb-000005
Figure PCTCN2019083241-appb-000005
在一些实施方案中,本发明抗GITR抗体或其片段的重链和/或轻链还包含信号肽序列,例如METDTLLLWVLLLWVPGSTG(SEQ ID NO:43)。In some embodiments, the heavy and/or light chain of an anti-GITR antibody or fragment thereof of the invention further comprises a signal peptide sequence, such as METDTLLLWVLLLWVPGSTG (SEQ ID NO: 43).
在本发明的一个实施方案中,本文所述的氨基酸改变包括氨基酸的置换、插入或缺失。优选的,本文所述的氨基酸改变为氨基酸置换,优选地保守置换。In one embodiment of the invention, the amino acid changes described herein include substitutions, insertions or deletions of amino acids. Preferably, the amino acid changes described herein are amino acid substitutions, preferably conservative substitutions.
在优选的实施方案中,本发明所述的氨基酸改变发生在CDR外的区域(例如在FR中)。更优选地,本发明所述的氨基酸改变发生在重链可变区外和/或轻链可变区外的区域。In a preferred embodiment, the amino acid changes described herein occur in regions outside the CDRs (e.g., in FR). More preferably, the amino acid changes described herein occur in regions outside the heavy chain variable region and/or outside the light chain variable region.
在一些实施方案中,置换为保守性置换。保守置换是指一个氨基酸经相同类别内的另一氨基酸置换,例如一个酸性氨基酸经另一酸性氨基酸置换,一个碱性氨基酸经另一碱性氨基酸置换,或一个中性氨基酸经另一中性氨基酸置换。示例性的置换如下表D所示:In some embodiments, the substitution is a conservative substitution. A conservative substitution is one in which one amino acid is replaced by another amino acid in the same class, for example, one acidic amino acid is replaced by another acidic amino acid, one basic amino acid is replaced by another basic amino acid, or one neutral amino acid is passed through another neutral amino acid. Replacement. An exemplary substitution is shown in Table D below:
表DTable D
原始残基Primitive residue 示例性置换Exemplary replacement 优选的保守氨基酸置换Preferred conservative amino acid substitution
Ala(A)Ala(A) Val、Leu、IleVal, Leu, Ile ValVal
Arg(R)Arg(R) Lys、Gln、AsnLys, Gln, Asn LysLys
Asn(N)Asn(N) Gln、His、Asp、Lys、ArgGln, His, Asp, Lys, Arg GlnGln
Asp(D)Asp(D) Glu、AsnGlu, Asn GluGlu
Cys(C)Cys(C) Ser、AlaSer, Ala SerSer
Gln(Q)Gln(Q) Asn、GluAsn, Glu AsnAsn
Glu(E)Glu(E) Asp、GlnAsp, Gln AspAsp
Gly(G)Gly(G) AlaAla AlaAla
His(H)His(H) Asn、Gln、Lys、ArgAsn, Gln, Lys, Arg ArgArg
Ile(I)Ile(I) Leu、Val、Met、Ala、Phe、正亮氨酸Leu, Val, Met, Ala, Phe, norleucine LeuLeu
Leu(L)Leu(L) 正亮氨酸、Ile、Val、Met、Ala、PheNorleucine, Ile, Val, Met, Ala, Phe IleIle
Lys(K)Lys(K) Arg、Gln、AsnArg, Gln, Asn ArgArg
Met(M)Met(M) Leu、Phe、IleLeu, Phe, Ile LeuLeu
Phe(F)Phe(F) Trp、Leu、Val、Ile、Ala、TyrTrp, Leu, Val, Ile, Ala, Tyr TyrTyr
Pro(P)Pro(P) AlaAla AlaAla
Ser(S)Ser(S) ThrThr ThrThr
Thr(T)Thr(T) Val、SerVal, Ser SerSer
Trp(W)Trp(W) Tyr、PheTyr, Phe TyrTyr
Tyr(Y)Tyr(Y) Trp、Phe、Thr、SerTrp, Phe, Thr, Ser PhePhe
Val(V)Val(V) Ile、Leu、Met、Phe、Ala、正亮氨酸Ile, Leu, Met, Phe, Ala, norleucine LeuLeu
在某些实施方案中,置换发生在抗体的CDR区。通常,获得的变体相对于亲本抗体在某些生物学特性方面(例如,增加的亲和力)具有修饰(例如,改善)和/或将具有亲本抗体的基本上保留的某些生 物学特性。示例性置换变体是亲和力成熟抗体。In certain embodiments, the substitution occurs in the CDR regions of the antibody. Generally, the variant obtained has modifications (e. g., improved) relative to the parent antibody in certain biological properties (e.g., increased affinity) and/or will have certain biological properties that are substantially retained by the parent antibody. An exemplary substitution variant is an affinity matured antibody.
在某些实施方案中,本文中所提供的抗体经改变以增加或降低抗体经糖基化的程度。对抗体的糖基化位点的添加或缺失可通过改变氨基酸序列以便产生或移除一或多个糖基化位点而方便地实现。当抗体包含Fc区时,可以改变附着于其的糖类。在一些应用中,除去不想要的糖基化位点的修饰可以是有用的,例如除去岩藻糖模块以提高抗体依赖性细胞性细胞毒性(ADCC)功能(参见Shield等(2002)JBC277:26733)。在其它应用中,可以进行半乳糖苷化修饰以修饰补体依赖性细胞毒性(CDC)。In certain embodiments, the antibodies provided herein are altered to increase or decrease the extent of glycosylation of the antibody. Addition or deletion of a glycosylation site to an antibody can be conveniently accomplished by altering the amino acid sequence to create or remove one or more glycosylation sites. When the antibody comprises an Fc region, the saccharide attached thereto can be altered. In some applications, modifications that remove unwanted glycosylation sites may be useful, such as removal of fucose modules to enhance antibody-dependent cellular cytotoxicity (ADCC) function (see Shield et al. (2002) JBC 277:26733 ). In other applications, galactosidation modification can be performed to modify complement dependent cytotoxicity (CDC).
在某些实施方案中,可在本文中所提供抗体的Fc区中引入一个或多个氨基酸修饰,以此产生Fc区变体,以便增强例如抗体治疗癌症或细胞增殖性疾病的有效性。Fc区变体可包括在一或多个氨基酸位置处包含氨基酸修饰(例如置换)的人Fc区序列(例如人IgGl、IgG2、IgG3或IgG4Fc区)。关于Fc变体的实例参见美国专利号7,332,581,美国专利号6,737,056,美国专利号6,737,056;WO2004/056312和Shields等人,J.Biol.Chem.9(2):6591-6604(2001),美国专利号6,194,551、WO 99/51642和Idusogie等人J.Immunol.164:4178-4184(2000),美国专利号7,371,826,Duncan&Winter,Nature322:738-40(1988);美国专利号5,648,260;美国专利号5,624,821;和WO 94/29351。In certain embodiments, one or more amino acid modifications can be introduced into the Fc region of an antibody provided herein to produce an Fc region variant to enhance the effectiveness of, for example, an antibody in treating cancer or a cell proliferative disorder. An Fc region variant can include a human Fc region sequence (eg, a human IgGl, IgG2, IgG3, or IgG4 Fc region) comprising an amino acid modification (eg, a substitution) at one or more amino acid positions. For examples of Fc variants, see U.S. Patent No. 7,332,581, U.S. Patent No. 6,737,056, U.S. Patent No. 6,737,056, WO2004/056312, and Shields et al., J. Biol. Chem. 9(2): 6591-6604 (2001), U.S. Patent No. 6,194,551, WO 99/51642 and Idusogie et al. J. Immunol. 164: 4178-4184 (2000), U.S. Patent No. 7,371,826, Duncan & Winter, Nature 322: 738-40 (1988); U.S. Patent No. 5,648,260; U.S. Patent No. 5,624,821; And WO 94/29351.
在某些实施方案中,可能需要产生经半胱氨酸工程改造的抗体,例如“硫代MAb”,其中抗体的一或多个残基经半胱氨酸残基置换。可以如,例如美国专利号7,521,541中所述地生成半胱氨酸改造的抗体。In certain embodiments, it may be desirable to produce a cysteine engineered antibody, such as a "thiocarba", wherein one or more residues of the antibody are replaced with a cysteine residue. Cysteine engineered antibodies can be generated as described, for example, in U.S. Patent No. 7,521,541.
在某些实施方案中,本文中所提供的抗体可进一步经修饰为含有本领域中已知且轻易获得的其他非蛋白质部分。适合抗体衍生作用的部分包括,但不限于,水溶性聚合物。水溶性聚合物的非限制性实例包括,但不限于,聚乙二醇(PEG)、乙二醇/丙二醇共聚物、羧甲基纤维素、葡聚糖、聚乙烯醇、聚乙烯吡咯烷酮、聚-1,3-二烷、聚-1,3,6-三烷、乙烯/马来酸酐共聚物、聚氨基酸(均聚物或无规共聚物)、及葡聚糖或聚(n-乙烯基吡咯烷酮)聚乙二醇、丙二醇均聚物、聚环氧丙烷/氧化乙烯共聚物、聚氧乙基化多元醇(例如甘油)、聚乙烯醇、及其混合物。In certain embodiments, the antibodies provided herein can be further modified to contain other non-protein portions known in the art and readily available. Portions suitable for antibody derivatization include, but are not limited to, water soluble polymers. Non-limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), ethylene glycol/propylene glycol copolymers, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly -1,3-dioxane, poly-1,3,6-trioxane, ethylene/maleic anhydride copolymer, polyamino acid (homopolymer or random copolymer), and dextran or poly(n-ethylene) Pyrrolidone) polyethylene glycol, propylene glycol homopolymer, polypropylene oxide/ethylene oxide copolymer, polyoxyethylated polyol (such as glycerin), polyvinyl alcohol, and mixtures thereof.
在一些实施方案中,本发明的抗GITR抗体或其抗原结合片段具有以下一个或多个特性:In some embodiments, an anti-GITR antibody or antigen-binding fragment thereof of the invention has one or more of the following characteristics:
(i)显示与表3所列的任一抗体对GITR相同或相似的结合亲和力和/或特异性;(i) showing the same or similar binding affinity and/or specificity to any of the antibodies listed in Table 3 for GITR;
(ii)抑制(例如,竞争性抑制)表3所列的任一抗体与GITR的结合;(ii) inhibiting (eg, competitive inhibition) the binding of any of the antibodies listed in Table 3 to GITR;
(iii)与表3所示的任一抗体结合相同或重叠的表位;(iii) an epitope that binds to the same or overlaps with any of the antibodies shown in Table 3;
(iv)与表3所示的任一抗体竞争结合GITR;(iv) competing with any of the antibodies shown in Table 3 to bind GITR;
(v)具有表3所列的任一抗体分子的一个或多个生物学特性。(v) having one or more of the biological properties of any of the antibody molecules listed in Table 3.
在一些实施方案中,本发明的抗GITR抗体是IgG1形式的抗体或IgG2形式的抗体或IgG4形式的抗体。In some embodiments, an anti-GITR antibody of the invention is an antibody in the IgGl form or an antibody in the IgG2 form or an antibody in the IgG4 form.
在一些实施方案中,抗GITR抗体是单克隆抗体。In some embodiments, the anti-GITR antibody is a monoclonal antibody.
在一些实施方案中,抗GITR抗体是人源化的。用于使抗体人源化的不同方法是技术人员已知的,如由Almagro&Fransson综述的,其内容通过提述完整并入本文(Almagro JC和Fransson J(2008)Frontiers inBioscience13:1619-1633)。In some embodiments, the anti-GITR antibody is humanized. Different methods for humanizing antibodies are known to the skilled person, as reviewed by Almagro & Fransson, the contents of which are incorporated herein by reference in its entirety (Almagro JC and Fransson J (2008) Frontiers in Bioscience 13: 1619-1633).
在一些实施方案中,抗GITR抗体是人抗体。可使用本领域中已知的各种技术来制备人抗体。人抗体一般描述于van Dijk和van de Winkel,Curr.Opin.Pharmacol 5:368-74(2001)以及Lonberg,Curr.Opin.Immunol 20:450-459(2008)。In some embodiments, the anti-GITR antibody is a human antibody. Human antibodies can be prepared using a variety of techniques known in the art. Human antibodies are generally described in van Dijk and van de Winkel, Curr. Opin. Pharmacol 5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol 20: 450-459 (2008).
在一些实施方案中,抗GITR抗体是嵌合抗体。In some embodiments, the anti-GITR antibody is a chimeric antibody.
在一些实施方案中,至少部分的抗GITR抗体的框架序列是人共有框架序列。在一个实施方案中,本发明的抗GITR抗体还涵盖其抗体片段,优选地选自以下的抗体片段:Fab、Fab’、Fab’-SH、Fv、单链抗体(例如scFv)或(Fab’) 2、单结构域抗体、双抗体(dAb)或线性抗体。 In some embodiments, at least a portion of the framework sequence of the anti-GITR antibody is a human consensus framework sequence. In one embodiment, an anti-GITR antibody of the invention further encompasses an antibody fragment thereof, preferably an antibody fragment selected from the group consisting of Fab, Fab', Fab'-SH, Fv, single chain antibody (eg, scFv) or (Fab' 2 ) Single domain antibody, double antibody (dAb) or linear antibody.
在某些实施方案中,抗GITR抗体分子处于双特异性或多特异性抗体分子形式。在一个实施方案中,双特异性抗体分子具有针对GITR的第一结合特异性和针对PD-1或PD-L1或PD-L2的第二结合特异性。在一个实施方案中,双特异性抗体分子与GITR和PD-1结合。在另一个实施方案中,双特异性抗体分子与GITR和PD-L1结合。在又一个实施方案中,双特异性抗体分子与GITR和PD-L2结合。多特异性抗体分子可以具有任何针对前述分子的结合特异性的组合。多特异性抗体分子例如可以是三特异性抗体分子,其包含针对GITR的第一结合特异性和针对以下一种或多种的分子的第二及第三结合特异性:PD-1、PD-L1或PD-L2。In certain embodiments, the anti-GITR antibody molecule is in the form of a bispecific or multispecific antibody molecule. In one embodiment, the bispecific antibody molecule has a first binding specificity for GITR and a second binding specificity for PD-1 or PD-L1 or PD-L2. In one embodiment, the bispecific antibody molecule binds to GITR and PD-1. In another embodiment, the bispecific antibody molecule binds to GITR and PD-L1. In yet another embodiment, the bispecific antibody molecule binds to GITR and PD-L2. The multispecific antibody molecule can have any combination of binding specificities for the aforementioned molecules. The multispecific antibody molecule can be, for example, a trispecific antibody molecule comprising a first binding specificity for GITR and a second and third binding specificity for a molecule of one or more of the following: PD-1, PD- L1 or PD-L2.
II.本发明的核酸以及包含其的宿主细胞II. Nucleic acids of the invention and host cells comprising the same
在一方面,本发明提供了编码以上任何抗GITR抗体或其片段的核酸。在一个实施方案中,提供包含所述核酸的载体。在一个实施方案中,载体是表达载体。在一个实施方案中,提供包含所述核酸或所述载体的宿主细胞。在一个实施方案中,宿主细胞是真核的。在另一个实施方案中,宿主细胞选自酵母细胞、哺乳动物细胞(例如CHO细胞或293细胞)或适用于制备抗体或其抗原结合片段的其它细胞。在另一个实施方案中,宿主细胞是原核的。In one aspect, the invention provides a nucleic acid encoding any of the above anti-GITR antibodies or fragments thereof. In one embodiment, a vector comprising the nucleic acid is provided. In one embodiment, the vector is an expression vector. In one embodiment, a host cell comprising the nucleic acid or the vector is provided. In one embodiment, the host cell is eukaryotic. In another embodiment, the host cell is selected from the group consisting of a yeast cell, a mammalian cell (eg, a CHO cell or a 293 cell), or other cell suitable for use in the preparation of an antibody or antigen-binding fragment thereof. In another embodiment, the host cell is prokaryotic.
在一方面,本发明提供了编码以上任何抗GITR抗体或其片段的核酸。所述核酸可以包含编码抗体的轻链可变区和/或重链可变区的氨基酸序列的核酸,或包含编码抗体的轻链和/或重链的氨基酸序列的核酸。示例性的编码抗体重链可变区的核酸序列包含与选自SEQ ID NO:33、34、35或36的核酸序列具有至少80%、85%、90%、91%、92%、93%、94%、95%、96%、97%、98%或99%的同一性的核酸序列,或者包含选自SEQ ID NO:33、34、35或36的核酸序列。示例性的编码抗体轻链可变区的核酸序列包含与选自SEQ ID NO:37、38、39或40的核酸序列具有至少80%、85%、90%、91%、92%、93%、94%、95%、96%、97%、98%或99%的同一性的核酸序列,或者包含选自SEQ ID NO:37、38、39或40的核酸序列。In one aspect, the invention provides a nucleic acid encoding any of the above anti-GITR antibodies or fragments thereof. The nucleic acid may comprise a nucleic acid encoding an amino acid sequence of a light chain variable region and/or a heavy chain variable region of an antibody, or a nucleic acid comprising an amino acid sequence encoding a light chain and/or a heavy chain of an antibody. An exemplary nucleic acid sequence encoding an antibody heavy chain variable region comprising at least 80%, 85%, 90%, 91%, 92%, 93% of a nucleic acid sequence selected from the group consisting of SEQ ID NO: 33, 34, 35 or 36 a nucleic acid sequence of 94%, 95%, 96%, 97%, 98% or 99% identity, or a nucleic acid sequence selected from the group consisting of SEQ ID NO: 33, 34, 35 or 36. An exemplary nucleic acid sequence encoding an antibody light chain variable region comprises at least 80%, 85%, 90%, 91%, 92%, 93% of a nucleic acid sequence selected from the group consisting of SEQ ID NO: 37, 38, 39 or 40 A nucleic acid sequence of 94%, 95%, 96%, 97%, 98% or 99% identity, or a nucleic acid sequence selected from the group consisting of SEQ ID NO: 37, 38, 39 or 40.
在一个实施方案中,提供包含所述核酸的一个或多个载体。在一个实施方案中,载体是表达载体,例如真核表达载体。载体包括但不限于病毒、质粒、粘粒、λ噬菌体或酵母人工染色体(YAC)。在一个实施方案中,载体是pTT5载体。In one embodiment, one or more vectors comprising the nucleic acid are provided. In one embodiment, the vector is an expression vector, such as a eukaryotic expression vector. Vectors include, but are not limited to, viruses, plasmids, cosmids, lambda phage, or yeast artificial chromosomes (YAC). In one embodiment, the vector is a pTT5 vector.
在一个实施方案中,提供包含所述载体的宿主细胞。用于克隆或表达编码抗体的载体的适当宿主细胞包括本文描述的原核或真核细胞。例如,抗体可在细菌中产生,特别当不需要糖基化和Fc效应子功能时。对于抗体片段和多肽在细菌中的表达,见,例如,美国专利号5,648,237,5,789,199和5,840,523,还见Charlton,Methods in Molecular Biology,卷248(B.K.C.Lo,编辑,Humana Press,Totowa,NJ,2003),第245-254页,其描述抗体片段在大肠杆菌中的表达)。在表达后,抗体可以从可溶级分中的细菌细胞糊状物分离,并且可以进一步纯化。In one embodiment, a host cell comprising the vector is provided. Suitable host cells for cloning or expressing a vector encoding the antibody include prokaryotic or eukaryotic cells as described herein. For example, antibodies can be produced in bacteria, particularly when glycosylation and Fc effector functions are not required. For expression of antibody fragments and polypeptides in bacteria, see, for example, U.S. Patent Nos. 5,648,237, 5,789,199 and 5,840,523, also to Charlton, Methods in Molecular Biology, Vol. 248 (BKCLo, ed., Humana Press, Totowa, NJ, 2003). , pp. 245-254, which describes the expression of antibody fragments in E. coli). After expression, the antibody can be isolated from the bacterial cell paste in the soluble fraction and can be further purified.
在一个实施方案中,宿主细胞是真核的。在另一个实施方案中,宿主细胞选自酵母细胞、哺乳动物细胞或适用于制备抗体或其抗原结合片段的其它细胞。例如,真核微生物诸如丝状真菌或酵母是关于编码抗体的载体的合适克隆或表达宿主。例如,糖基化途径已经进行“人源化”的真菌和酵母菌株导致产生具有部分或完全人糖基化模式的抗体。参见Gerngross,Nat.Biotech.22:1409-1414(2004),和 Li等,Nat.Biotech.24:210-215(2006)。适于表达糖基化抗体的宿主细胞也衍生自多细胞生物体(无脊椎动物和脊椎动物)。也可以将脊椎动物细胞用作宿主。例如,可以使用被改造以适合于悬浮生长的哺乳动物细胞系。有用哺乳动物宿主细胞系的其它实例是用SV40转化的猴肾CV1系(COS-7);人胚肾系(293HEK或293F或293细胞,如例如Graham等,J.Gen Virol.36:59(1977)中所描述的)等。其它有用的哺乳动物宿主细胞系包括中国仓鼠卵巢(CHO)细胞,包括DHFR-CHO细胞(Urlaub等,Proc.Natl.Acad.Sci.USA 77:216(1980)、CHO-S细胞等;以及骨髓瘤细胞系如Y0,NS0和Sp2/0。关于适合产生抗体的某些哺乳动物宿主细胞系的综述见例如Yazaki和Wu,Methods in Molecular Biology,卷248(B.K.C.Lo,ed.,Humana Press,Totowa,NJ),第255-268页(2003)。In one embodiment, the host cell is eukaryotic. In another embodiment, the host cell is selected from the group consisting of a yeast cell, a mammalian cell, or other cell suitable for use in the preparation of an antibody or antigen-binding fragment thereof. For example, a eukaryotic microorganism such as a filamentous fungus or yeast is a suitable cloning or expression host for a vector encoding an antibody. For example, fungal and yeast strains that have been "humanized" by the glycosylation pathway result in the production of antibodies with partial or complete human glycosylation patterns. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004), and Li et al, Nat. Biotech. 24: 210-215 (2006). Host cells suitable for expression of glycosylated antibodies are also derived from multicellular organisms (invertebrates and vertebrates). Vertebrate cells can also be used as hosts. For example, mammalian cell lines engineered to be suitable for suspension growth can be used. Other examples of useful mammalian host cell lines are the monkey kidney CV1 line (COS-7) transformed with SV40; human embryonic kidney line (293 HEK or 293F or 293 cells such as, for example, Graham et al, J. Gen Virol. 36:59 ( (described in 1977) and so on. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR-CHO cells (Urlaub et al, Proc. Natl. Acad. Sci. USA 77: 216 (1980), CHO-S cells, etc.; Tumor cell lines such as Y0, NSO and Sp2/0. For a review of certain mammalian host cell lines suitable for antibody production, see, for example, Yazaki and Wu, Methods in Molecular Biology, Vol. 248 (BKCLo, ed., Humana Press, Totowa). , NJ), pp. 255-268 (2003).
在一个实施方案中,本发明提供制备抗GITR抗体或其片段(优选的抗原结合片段)的方法,其中所述方法包括在适于表达编码所述抗体或其片段(优选的抗原结合片段)的核酸的条件下培养所述宿主细胞,以及任选地分离所述抗体或其片段(优选地抗原结合片段)。在某个实施方案中,所述方法还包括从宿主细胞回收抗GITR抗体或其片段(优选地抗原结合片段)。In one embodiment, the invention provides a method of making an anti-GITR antibody or fragment thereof (preferably an antigen-binding fragment), wherein the method comprises suitably expressing an antibody or fragment thereof (preferably an antigen-binding fragment) encoding the antibody The host cell is cultured under conditions of a nucleic acid, and the antibody or fragment thereof (preferably an antigen-binding fragment) is optionally isolated. In a certain embodiment, the method further comprises recovering an anti-GITR antibody or fragment thereof (preferably an antigen-binding fragment) from the host cell.
在一个实施方案中,提供了制备抗GITR抗体的方法,其中所述方法包括,在适合抗体表达的条件下,培养包含编码所述抗体的核酸的宿主细胞,如上文所提供的,和任选地从所述宿主细胞(或宿主细胞培养基)回收所述抗体。为了重组产生抗GITR抗体,分离编码抗体(例如上文所描述的抗体)的核酸,并将其插入一个或多个载体,用于在宿主细胞中进一步克隆和/或表达。此类核酸易于使用常规规程分离和测序(例如通过使用能够与编码抗体重链和轻链的基因特异性结合的寡核苷酸探针进行)。In one embodiment, a method of making an anti-GITR antibody, wherein the method comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, and optionally, under conditions suitable for expression of the antibody The antibody is recovered from the host cell (or host cell culture medium). For recombinant production of an anti-GITR antibody, a nucleic acid encoding an antibody (such as the antibodies described above) is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acids are readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of specifically binding to genes encoding the heavy and light chains of the antibody).
III.测定法III. Assay
可以通过本领域中已知的多种测定法对本文中提供的抗GITR抗体鉴定,筛选,或表征其物理/化学特性和/或生物学活性。一方面,对本发明的抗体测试其抗原结合活性,例如通过已知的方法诸如ELISA,Western印迹,等来进行。可使用本领域已知方法来测定对GITR的结合,本文中公开了例示性方法。在一些实施方案中,使用生物光干涉测定法(例如Fortebio亲和测量)或MSD测定法。The anti-GITR antibodies provided herein can be identified, screened, or characterized for their physical/chemical properties and/or biological activity by a variety of assays known in the art. In one aspect, the antibody of the present invention is tested for its antigen binding activity, for example, by known methods such as ELISA, Western blot, and the like. Binding to GITR can be determined using methods known in the art, and exemplary methods are disclosed herein. In some embodiments, biophotonic interferometry (eg, Fortebio affinity measurement) or MSD assay is used.
另一方面,可使用竞争测定法来鉴定与本文中公开的任何抗GITR抗体竞争对GITR的结合的抗体。在某些实施方案中,此类竞争性抗体结合与本文中公开的任何抗GITR抗体所结合表位相同或重叠的表位(例如线性或构象表位)。用于定位抗体所结合表位的详细例示性方法见Morris(1996)“Epitope Mapping Protocols”,Methods in Molecular Biology vol.66(Humana Press,Totowa,NJ)。In another aspect, a competition assay can be used to identify antibodies that compete with any of the anti-GITR antibodies disclosed herein for binding to GITR. In certain embodiments, such a competitive antibody binds to an epitope (eg, a linear or conformational epitope) that is identical or overlapping with an epitope to which any of the anti-GITR antibodies disclosed herein bind. A detailed exemplary method for locating epitopes bound by antibodies is found in Morris (1996) "Epitope Mapping Protocols", Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ).
本发明还提供了用于鉴定具有生物学活性的抗GITR抗体的测定法。生物学活性可以包括例如结合GITR(例如结合人和/或食蟹猴GITR),提高GITR介导的信号转导(例如提高NFkappa-B信号通路),通过ADCC消减表达GITR的细胞(例如Treg细胞),增强T效应细胞功能(例如CD4效应T细胞)(例如通过提高效应T细胞的细胞因子生成(例如干扰素如IFN-γ或白细胞介素如IL2))。还提供在体内和/或在体外具有此类生物学活性的抗体。The invention also provides assays for identifying biologically active anti-GITR antibodies. Biological activity can include, for example, binding to GITR (eg, binding to human and/or cynomolgus GITR), increasing GITR-mediated signal transduction (eg, increasing NFkappa-B signaling pathway), and attenuating cells expressing GITR by ADCC (eg, Treg cells) Enhancing T effector cell function (eg, CD4 effector T cells) (eg, by increasing cytokine production of effector T cells (eg, interferons such as IFN-[gamma] or interleukins such as IL2)). Antibodies having such biological activity in vivo and/or in vitro are also provided.
在某些实施方案中,对本发明的抗体测试此类生物学活性。In certain embodiments, the antibodies of the invention are tested for such biological activity.
可以使用本领域已知的方法来测定T细胞活化。例如,通过T细胞活化后释放的细胞因子,例如干扰素(如IFN-γ)或白细胞介素(例如IL2)的水平来测定。还可以使用本领域公知的方法来测定GITR信号传导(例如NF-kappaB信号通路),从而测定T细胞的活化。在一个实施方案中,生成表 达人GITR和报告基因(包含融合至报告基因(例如β萦光素酶)的NF-kappa B启动子)的转基因细胞。对细胞添加抗GITR抗体导致NF-kappa B转录升高,这使用针对报告基因的测定法(例如萦光素酶报告基因测定法)来检测。T cell activation can be assayed using methods known in the art. For example, it is determined by the level of a cytokine released after T cell activation, such as interferon (such as IFN-γ) or interleukin (such as IL2). GITR signaling (e.g., NF-kappaB signaling pathway) can also be assayed using methods well known in the art to determine T cell activation. In one embodiment, a transgenic cell expressing a human GITR and a reporter gene comprising a NF-kappa B promoter fused to a reporter gene (e.g., beta luciferase) is generated. Addition of anti-GITR antibodies to cells results in increased NF-kappa B transcription, which is detected using an assay for the reporter gene (eg, a luciferase reporter assay).
可以使用本领域已知的方法来测定抗体的ADCC效应。例如通过NF-AT信号激活。在一个实施方案中,获得ADCC效应细胞(包含融合至报告基因(例如β萦光素酶)的NF-AT启动子)的转基因细胞。对效应细胞与高表达GITR的细胞进行共培养,同时添加抗GITR抗体导致效应细胞NF-AT转录升高,这使用针对报告基因的测定法(例如萦光素酶报告基因测定法)来检测。The ADCC effect of an antibody can be determined using methods known in the art. For example, activation by NF-AT signal. In one embodiment, a transgenic cell comprising an ADCC effector cell comprising an NF-AT promoter fused to a reporter gene (eg, beta luciferase) is obtained. Co-culture of effector cells with cells that highly express GITR, while the addition of anti-GITR antibodies results in increased NF-AT transcription of effector cells, which is detected using assays for reporter genes (eg, luciferase reporter assay).
供任何上述体外测定法使用的细胞包括天然表达GITR或经改造而表达GITR细胞系。此类细胞包括天然表达GITR的T细胞,Treg细胞和活化后的记忆T细胞。此类细胞还包括表达GITR和并非正常情况下表达GITR的编码GITR DNA转染的细胞系。Cells for use in any of the above in vitro assays include naturally expressing GITR or engineered to express a GITR cell line. Such cells include T cells that naturally express GITR, Treg cells, and activated memory T cells. Such cells also include cell lines encoding GITR and GITR-transfected cells that are not normally expressing GITR.
可以理解的是,能够使用本发明的免疫缀合物替换或补充抗GITR抗体来进行任何上述测定法。It will be appreciated that any of the above assays can be performed by replacing or supplementing the anti-GITR antibody with an immunoconjugate of the invention.
可以理解的是,能够使用抗GITR抗体和别的活性剂来进行任何上述测定法。It will be appreciated that any of the above assays can be performed using an anti-GITR antibody and another active agent.
IV.免疫缀合物IV. Immunoconjugate
在一些实施方案中,本发明提供了免疫缀合物,其包含本文中提供的任何抗GITR抗体和其它物质,例如细胞毒性剂。在一些实施方案中,其它物质例如治疗剂,如细胞毒性剂或免疫抑制剂或化疗剂。细胞毒性剂包括任何对细胞有害的药剂。适合于形成免疫缀合物的细胞毒性剂(例如化疗剂)的例子是本领域中已知的。例如,细胞毒性剂包括但不限于:放射性同位素;生长抑制剂;酶及其片段如核酸水解酶;抗生素;毒素如小分子毒素或细菌、真菌、植物或动物起源的酶促活性毒素,包括其片段和/或变体;和已知的各种抗肿瘤或抗癌剂。In some embodiments, the invention provides an immunoconjugate comprising any of the anti-GITR antibodies and other materials provided herein, such as a cytotoxic agent. In some embodiments, other substances such as therapeutic agents, such as cytotoxic or immunosuppressive agents or chemotherapeutic agents. Cytotoxic agents include any agent that is harmful to cells. Examples of cytotoxic agents (e.g., chemotherapeutic agents) suitable for forming immunoconjugates are known in the art. For example, cytotoxic agents include, but are not limited to, radioisotopes; growth inhibitors; enzymes and fragments thereof such as nucleases; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including Fragments and/or variants; and various anti-tumor or anti-cancer agents known.
在一些实施方案中,所述免疫缀合物用于预防或治疗肿瘤,例如胃肠道肿瘤。在一些实施方案中,肿瘤为癌症,例如胃肠道癌症,例如胃癌、直肠癌、结肠癌、结肠直肠癌等。在一些实施方案中,所述免疫缀合物用于预防或治疗感染,例如慢性感染,例如细菌感染、病毒感染、真菌感染、原生动物感染等。In some embodiments, the immunoconjugate is for use in preventing or treating a tumor, such as a gastrointestinal tumor. In some embodiments, the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like. In some embodiments, the immunoconjugate is used to prevent or treat an infection, such as a chronic infection, such as a bacterial infection, a viral infection, a fungal infection, a protozoal infection, and the like.
V.药物组合物和药物制剂V. Pharmaceutical compositions and pharmaceutical preparations
在一些实施方案中,本发明提供包含本文所述的任何抗GITR抗体或其片段(优选地其抗原结合片段)或其免疫缀合物的组合物,优选地组合物为药物组合物。在一个实施方案中,所述组合物还包含药用辅料。在一个实施方案中,组合物,例如,药物组合物,包含本发明的抗GITR抗体或其片段或其免疫缀合物,以及一种或多种其它治疗剂(例如化疗剂、细胞毒性剂、疫苗、其它抗体、抗感染活性剂、小分子药物或免疫调节剂,优选地PD-1轴结合拮抗剂,例如抗PD-1抗体、抗PD-L1抗体或抗PD-L2抗体)的组合。In some embodiments, the invention provides compositions comprising any of the anti-GITR antibodies or fragments thereof, preferably antigen-binding fragments thereof, or immunoconjugates thereof, preferably a composition, which is a pharmaceutical composition. In one embodiment, the composition further comprises a pharmaceutical excipient. In one embodiment, a composition, eg, a pharmaceutical composition, comprises an anti-GITR antibody or fragment thereof of the invention, or an immunoconjugate thereof, and one or more additional therapeutic agents (eg, chemotherapeutic agents, cytotoxic agents, A combination of a vaccine, other antibody, anti-infective active agent, small molecule drug or immunomodulatory agent, preferably a PD-1 axis binding antagonist, such as an anti-PD-1 antibody, an anti-PD-L1 antibody or an anti-PD-L2 antibody.
在一些实施方案中,所述组合物用于预防或治疗肿瘤,例如胃肠道肿瘤。在一些实施方案中,肿瘤为癌症,例如胃肠道癌症,例如胃癌、直肠癌、结肠癌、结肠直肠癌等。在一些实施方案中,所述组合物用于预防或治疗感染,例如慢性感染,例如细菌感染、病毒感染、真菌感染、原生动物感染等。In some embodiments, the composition is for use in preventing or treating a tumor, such as a gastrointestinal tumor. In some embodiments, the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like. In some embodiments, the composition is for preventing or treating an infection, such as a chronic infection, such as a bacterial infection, a viral infection, a fungal infection, a protozoal infection, and the like.
本发明还包括包含抗GITR抗体或其免疫缀合物的组合物(包括药物组合物或药物制剂)和包含编 码抗GITR抗体的多核苷酸的组合物(包括药物组合物或药物制剂)。在某些实施方案中,组合物包含一种或多种结合GITR的抗体或其片段或一种或多种编码一种或多种结合GITR的抗体或其片段的多核苷酸。这些组合物还可以包含合适的药用辅料,如本领域中已知的药用载体、药用赋形剂,包括缓冲剂。The present invention also encompasses a composition (including a pharmaceutical composition or a pharmaceutical preparation) comprising an anti-GITR antibody or an immunoconjugate thereof, and a composition (including a pharmaceutical composition or a pharmaceutical preparation) comprising a polynucleotide encoding an anti-GITR antibody. In certain embodiments, the compositions comprise one or more antibodies or fragments thereof that bind to GITR or one or more polynucleotides that encode one or more antibodies or fragments thereof that bind to GITR. These compositions may also contain suitable pharmaceutical excipients such as pharmaceutically acceptable carriers, pharmaceutically acceptable excipients, including buffering agents known in the art.
适用于本发明的药用载体可以是无菌液体,如水和油,包括那些具有石油、动物、植物或合成起源的,如花生油、大豆油、矿物油、芝麻油等。当静脉内施用药物组合物时,水是优选的载体。还可以将盐水溶液和水性右旋糖以及甘油溶液用作液体载体,特别是用于可注射溶液。合适的药用赋形剂包括淀粉、葡萄糖、乳糖、蔗糖、明胶、麦芽、米、面粉、白垩、硅胶、硬脂酸钠、甘油单硬脂酸酯、滑石、氯化钠、干燥的脱脂乳、甘油、丙烯、二醇、水、乙醇等。对于赋形剂的使用及其用途,亦参见“Handbook of PharmaceuticalExcipients”,第五版,R.C.Rowe,P.J.Seskey和S.C.Owen,PharmaceuticalPress,London,Chicago。若期望的话,所述组合物还可以含有少量的润湿剂或乳化剂,或pH缓冲剂。这些组合物可以采用溶液、悬浮液、乳剂、片剂、丸剂、胶囊剂、粉末、持续释放配制剂等的形式。口服配制剂可以包含标准载体,如药用级甘露醇、乳糖、淀粉、硬脂酸镁、糖精。Pharmaceutically acceptable carriers suitable for use in the present invention may be sterile liquids such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be used as liquid carriers, especially for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, dried skim milk , glycerin, propylene, glycol, water, ethanol, etc. For the use of excipients and their use, see also "Handbook of Pharmaceutical Excipients", Fifth Edition, R. C. Rowe, P. J. Seskey and S. C. Owen, Pharmaceutical Press, London, Chicago. If desired, the compositions may also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions may take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained release formulations and the like. Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, saccharin.
可以通过将具有所需纯度的本发明的抗GITR抗体与一种或多种任选的药用辅料(Remington′s Pharmaceutical Sciences,第16版,Osol,A.编(1980))混合来制备包含本文所述的抗GITR抗体的药物制剂,优选地以冻干制剂或水溶液的形式。Preparation can be made by mixing an anti-GITR antibody of the invention having the desired purity with one or more optional pharmaceutical excipients (Remington's Pharmaceutical Sciences, 16th Ed., Osol, A. Ed. (1980)). The pharmaceutical preparations of the anti-GITR antibodies described herein are preferably in the form of a lyophilized formulation or an aqueous solution.
示例性的冻干抗体制剂描述于美国专利号6,267,958。水性抗体制剂包括美国专利号6,171,586和WO2006/044908中所述的那些,后一种制剂包括组氨酸-乙酸盐缓冲剂。Exemplary lyophilized antibody formulations are described in U.S. Patent No. 6,267,958. Aqueous antibody preparations include those described in U.S. Patent No. 6,171,586 and WO2006/044908, the latter including a histidine-acetate buffer.
本发明的药物组合物或制剂还可以包含一种或多种其它活性成分,所述活性成分是被治疗的特定适应证所需的,优选具有不会不利地影响彼此的互补活性的那些活性成分。例如,理想的是还提供其它抗癌活性成分,例如化疗剂、细胞毒性剂、疫苗、其它抗体、抗感染活性剂、小分子药物或免疫调节剂,例如PD-1轴结合拮抗剂(例如抗PD-1抗体或抗PD-L1抗体或抗PD-L2抗体)等。所述活性成分以对于目的用途有效的量合适地组合存在。The pharmaceutical compositions or formulations of the present invention may further comprise one or more additional active ingredients which are required for the particular indication being treated, preferably those which do not adversely affect each other's complementary activities. . For example, it is desirable to also provide other anti-cancer active ingredients such as chemotherapeutic agents, cytotoxic agents, vaccines, other antibodies, anti-infective active agents, small molecule drugs or immunomodulators such as PD-1 axis binding antagonists (eg, anti- PD-1 antibody or anti-PD-L1 antibody or anti-PD-L2 antibody) and the like. The active ingredient is suitably present in combination in an amount effective for the intended use.
可制备持续释放制剂。持续释放制剂的合适实例包括含有抗体的固体疏水聚合物的半渗透基质,所述基质呈成形物品,例如薄膜或微囊形式。Sustained release formulations can be prepared. Suitable examples of sustained release formulations include semipermeable matrices of solid hydrophobic polymers containing antibodies in the form of shaped articles such as films or microcapsules.
关于包含本发明抗体的药物制剂/药物组合物的其它组分,还可以参见WO2015/031667或WO2015/187835等中公开的那些。With regard to other components of the pharmaceutical preparations/pharmaceutical compositions comprising the antibodies of the invention, reference is also made to those disclosed in WO 2015/031667 or WO 2015/187835 and the like.
VI.组合产品VI. Combination products
在一些实施方案中,本发明还提供了组合产品,其包含本发明的抗体或其抗原结合片段,或其免疫缀合物,以及一种或多种其它治疗剂(例如化疗剂、其他抗体、细胞毒性剂、疫苗、抗感染活性剂、小分子药物或免疫调节剂等)。在一些实施方案中,其它抗体例如抗PD-1抗体或抗PD-L1抗体或抗PD-L2抗体。In some embodiments, the invention also provides a combination product comprising an antibody of the invention, or an antigen binding fragment thereof, or an immunoconjugate thereof, and one or more additional therapeutic agents (eg, chemotherapeutic agents, other antibodies, Cytotoxic agents, vaccines, anti-infective active agents, small molecule drugs or immunomodulators, etc.). In some embodiments, other antibodies are, for example, anti-PD-1 antibodies or anti-PD-L1 antibodies or anti-PD-L2 antibodies.
在一些实施方案中,所述组合产品用于预防或治疗肿瘤,例如胃肠道肿瘤。在一些实施方案中,肿瘤为癌症,例如胃肠道癌症,例如胃癌、直肠癌、结肠癌、结肠直肠癌等。在一些实施方案中,所述组合产品用于预防或治疗感染,例如慢性感染,例如细菌感染、病毒感染、真菌感染、原生动物感 染等。In some embodiments, the combination product is for use in preventing or treating a tumor, such as a gastrointestinal tumor. In some embodiments, the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like. In some embodiments, the combination product is for use in preventing or treating an infection, such as a chronic infection, such as a bacterial infection, a viral infection, a fungal infection, a protozoal infection, and the like.
VII.用途VII. Use
本发明一方面提供了在受试者中激活NF-KappaB信号通路的方法,包括向受试者施用有效量的本发明的抗GITR的抗体或其抗原结合片段、免疫缀合物、药物组合物或组合产品。In one aspect, the invention provides a method of activating a NF-KappaB signaling pathway in a subject, comprising administering to the subject an effective amount of an anti-GITR antibody of the invention, or an antigen-binding fragment thereof, an immunoconjugate, a pharmaceutical composition Or a combination of products.
本发明还一方面提供了在受试者中提高效应T细胞功能,例如激活效应T细胞的方法,包括向受试者施用有效量的本发明的抗GITR的抗体或其抗原结合片段、免疫缀合物、药物组合物或组合产品。在一些实施方案中,所述功能是效应T细胞增殖增强。在一些实施方案中,所述激活T细胞表现为干扰素或白细胞介素分泌/表达增加。在一些实施方案中,白细胞介素是IL2。在一些实施方案中,干扰素是IFN-γ。A further aspect of the invention provides a method of increasing effector T cell function, such as activating effector T cells, in a subject, comprising administering to the subject an effective amount of an anti-GITR antibody or antigen-binding fragment thereof, immunologically conjugated to the subject Compound, pharmaceutical composition or combination product. In some embodiments, the function is an increase in effector T cell proliferation. In some embodiments, the activated T cell is characterized by increased secretion/expression of interferon or interleukin. In some embodiments, the interleukin is IL2. In some embodiments, the interferon is IFN-[gamma].
本发明还一方面提供给了在受试者中介导ADCC而消除调节T细胞的方法,包括向受试者施用有效量的本发明的抗GITR的抗体或其抗原结合片段、免疫缀合物、药物组合物或组合产品。在一些实施方案中,所述调节T细胞是Treg细胞。In a further aspect, the present invention provides a method of mediating ADCC in a subject to eliminate regulatory T cells, comprising administering to the subject an effective amount of an anti-GITR antibody or antigen-binding fragment thereof, immunoconjugate of the present invention. , a pharmaceutical composition or a combination product. In some embodiments, the regulatory T cell is a Treg cell.
本发明一方面还提供了在受试者中预防或治疗肿瘤的方法,所述方法包括向所述受试者施用有效量的本文所述的任何抗GITR抗体或其片段、免疫缀合物、药物组合物或组合产品。In an aspect, the invention provides a method of preventing or treating a tumor in a subject, the method comprising administering to the subject an effective amount of any of the anti-GITR antibodies or fragments thereof, immunoconjugates, Pharmaceutical composition or combination product.
本发明一方面还提供了在受试者中预防或治疗感染的方法,所述方法包括向所述受试者施用有效量的本文所述的任何抗GITR抗体或其片段、免疫缀合物、药物组合物或组合产品。In an aspect, the invention provides a method of preventing or treating an infection in a subject, the method comprising administering to the subject an effective amount of any of the anti-GITR antibodies or fragments thereof, immunoconjugates, Pharmaceutical composition or combination product.
本发明还提供了在受试者中预防或治疗与Treg增殖有关的其它病状或病况的方法,所述方法包括向所述受试者施用有效量的本文所述的任何抗GITR抗体或其片段、免疫缀合物、药物组合物或组合产品。The invention also provides a method of preventing or treating other conditions or conditions associated with Treg proliferation in a subject, the method comprising administering to the subject an effective amount of any of the anti-GITR antibodies or fragments thereof described herein , immunoconjugate, pharmaceutical composition or combination product.
受试者可以是哺乳动物,例如,灵长类,优选地,高级灵长类,例如,人类(例如,患有本文所述疾病或具有患有本文所述疾病的风险的患者)。在一个实施方案中,受试者患有本文所述疾病(例如,如本文所述的肿瘤或感染性疾病)或具有患有本文所述疾病的风险。在某些实施方案中,受试者接受或已经接受过其它治疗,例如化疗治疗和/或放射疗法。备选地或组合下,受试者因感染而免疫受损或具有因感染而免疫受损的风险。The subject can be a mammal, for example, a primate, preferably a higher primate, for example, a human (eg, a patient having the disease described herein or at risk of having the disease described herein). In one embodiment, the subject has or is at risk of having a disease described herein (eg, a tumor or infectious disease as described herein). In certain embodiments, the subject has received or has received other treatments, such as chemotherapy treatment and/or radiation therapy. Alternatively or in combination, the subject is immunocompromised due to infection or has a risk of being immunocompromised by the infection.
在一些实施方案中,本文所述的肿瘤,例如癌症,包括但不限于实体瘤、血液学癌、软组织肿瘤和转移性病灶。在一些实施方案中,本文所述的用于治疗的癌症包括但乳腺癌、肺癌、卵巢癌、前列腺癌、结肠癌、直肠癌、结肠直肠癌、宫颈癌、脑癌、皮肤癌、肝癌、胰腺癌或胃癌等实体瘤,以及血癌例如白血病和淋巴瘤。在一些实施方案中,癌症为胃肠道癌症,例如胃癌、直肠癌、结肠癌、结肠直肠癌等。In some embodiments, the tumors described herein, eg, cancer, include, but are not limited to, solid tumors, hematological cancers, soft tissue tumors, and metastatic lesions. In some embodiments, the cancers described herein for treatment include but breast cancer, lung cancer, ovarian cancer, prostate cancer, colon cancer, rectal cancer, colorectal cancer, cervical cancer, brain cancer, skin cancer, liver cancer, pancreas Solid tumors such as cancer or stomach cancer, as well as blood cancers such as leukemia and lymphoma. In some embodiments, the cancer is a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer, and the like.
可以使用本文所述的抗体分子实现对转移性癌(例如,表达GITR或PD1的转移性癌)的治疗。Treatment of metastatic cancer (e.g., metastatic cancer expressing GITR or PDl) can be achieved using the antibody molecules described herein.
在一个实施方案中,肿瘤是表达升高水平的GITR和/或PD1的癌症。In one embodiment, the tumor is a cancer that expresses elevated levels of GITR and/or PDl.
在一些实施方案中,本文所述的癌症是结肠癌及其转移性癌症。In some embodiments, the cancer described herein is colon cancer and metastatic cancer thereof.
在一些实施方案中,所述感染是急性的或慢性的。在一些实施方案中,所述慢性感染是持续性的感染、潜伏的感染或缓慢感染。在一些实施方案中,所述慢性感染是由选自细菌、病毒、真菌和原生 动物的病原体导致的。In some embodiments, the infection is acute or chronic. In some embodiments, the chronic infection is a persistent infection, a latent infection, or a slow infection. In some embodiments, the chronic infection is caused by a pathogen selected from the group consisting of bacteria, viruses, fungi, and protozoa.
在其他方面,本发明提供抗GITR抗体或其片段在生产或制备药物中的用途,所述药物用于治疗本文提及的相关疾病或病症。In other aspects, the invention provides the use of an anti-GITR antibody or fragment thereof for the manufacture or preparation of a medicament for the treatment of a related disease or condition as referred to herein.
在一些实施方案中,本发明的抗体或抗体片段或免疫缀合物或组合物或产品会延迟病症和/或与病症相关的症状的发作。In some embodiments, an antibody or antibody fragment or immunoconjugate or composition or product of the invention delays the onset of a condition and/or a condition associated with the condition.
在一些实施方案中,本文所述的预防或治疗方法还包括向所述受试者或个体组合施用本文公开的抗体分子或药物组合物或免疫缀合物,以及一种或多种其它疗法,例如治疗方式和/或其它治疗剂。In some embodiments, the prophylactic or therapeutic methods described herein further comprise administering to the subject or individual a combination of an antibody molecule or pharmaceutical composition or immunoconjugate disclosed herein, and one or more additional therapies, For example, treatment modalities and/or other therapeutic agents.
在一些实施方案中,治疗方式包括外科手术(例如肿瘤切除术);放射疗法(例如,外粒子束疗法,它涉及其中设计照射区域的三维适形放射疗法)、局部照射(例如,指向预选靶或器官的照射)或聚焦照射)等。In some embodiments, the treatment modality includes surgery (eg, tumor resection); radiation therapy (eg, external particle beam therapy, which involves three-dimensional conformal radiation therapy in which the illumination region is designed), local illumination (eg, pointing to a pre-selected target) Or irradiation of organs) or focused illumination).
在一些实施方案中,治疗剂选自化疗剂、细胞毒性剂、疫苗、其它抗体、抗感染活性剂、小分子药物或免疫调节剂。In some embodiments, the therapeutic agent is selected from the group consisting of a chemotherapeutic agent, a cytotoxic agent, a vaccine, other antibodies, an anti-infective active agent, a small molecule drug, or an immunomodulatory agent.
示例性的疫苗包括但不限于癌症疫苗。疫苗可以是基于DNA的疫苗、基于RNA的疫苗或基于病毒转导的疫苗。癌症疫苗可以是预防性的或治疗性的。Exemplary vaccines include, but are not limited to, cancer vaccines. The vaccine can be a DNA based vaccine, an RNA based vaccine or a virus transduced vaccine. Cancer vaccines can be prophylactic or therapeutic.
示例性的抗感染活性剂包括但不限于,抗病毒剂、抗真菌剂、抗原生动物剂、抗菌剂,例如核苷类似物齐多夫定(AST)、更昔洛韦、膦甲酸或cidovir,如上文所述。Exemplary anti-infective active agents include, but are not limited to, antiviral, antifungal, antiprotozoal, antibacterial agents such as the nucleoside analog zidovudine (AST), ganciclovir, foscarnet or cidovir As described above.
示例性的其它抗体包括但不限于抗PD-1抗体、抗PDL1抗体或抗PDL2抗体。本领域已知多种抗PD-1抗体、抗PDL1抗体或抗PDL2抗体,参见例如WO2007/005874、WO2009/101611、WO2009/114335、WO2010/027827和WO2011/066342等。Exemplary additional antibodies include, but are not limited to, anti-PD-1 antibodies, anti-PDL1 antibodies, or anti-PDL2 antibodies. A variety of anti-PD-1 antibodies, anti-PDL1 antibodies or anti-PDL2 antibodies are known in the art, see for example WO2007/005874, WO2009/101611, WO2009/114335, WO2010/027827 and WO2011/066342, and the like.
在一些实施方案中,本文中描述的抗体组合可以分别施用,例如,作为单独的抗体分别施用,或连接时(例如作为双特异性或三特异性抗体分子)施用。In some embodiments, the antibody combinations described herein can be administered separately, eg, as separate antibodies, or when ligated (eg, as a bispecific or trispecific antibody molecule).
更多的可以与抗GITR抗体或其片段组合的疗法或治疗剂可以参见WO2015/031667。Further therapeutic or therapeutic agents which can be combined with anti-GITR antibodies or fragments thereof can be found in WO 2015/031667.
此类组合疗法涵盖组合施用(例如两种或更多种治疗剂包含在同一配制剂或分开的配制剂中),和分开施用,在该情况中,可以在施用别的治疗剂和/或药剂之前,同时,和/或之后发生本发明的抗体的施用。在一个实施方案中,抗GITR抗体的施用和别的治疗剂的施用彼此在约一个月内,或约一,两或三周内,或约1,2,3,4,5,或6天内发生。Such combination therapies encompasses combined administration (eg, two or more therapeutic agents are included in the same formulation or separate formulations), and administered separately, in which case other therapeutic agents and/or agents may be administered. Administration of the antibodies of the invention occurs previously, simultaneously, and/or after. In one embodiment, administration of the anti-GITR antibody and administration of the additional therapeutic agent are within about one month, or within about one, two or three weeks, or about 1, 2, 3, 4, 5, or 6 days of each other. occur.
待采用的含抗GITR抗体的药物组合物的治疗有效量将例如取决于治疗背景和目标。本领域技术人员将理解用于治疗的适当剂量水平将部分地根据以下因素而变化:递送的分子、所针对使用的适应症、施用途径、以及患者的体重、身体表面积或器官大小和/或情况(年龄和一般健康状态)。在某些实施方案中,临床医师可滴定剂量并且更改施用途径以获得最佳治疗效果。The therapeutically effective amount of the pharmaceutical composition comprising the anti-GITR antibody to be employed will depend, for example, on the therapeutic background and objectives. Those skilled in the art will appreciate that appropriate dosage levels for treatment will vary, depending in part on the molecule being delivered, the indication being used, the route of administration, and the weight, body surface area or organ size and/or condition of the patient (age and general health status). In certain embodiments, the clinician can titrate the dose and alter the route of administration to achieve optimal therapeutic effect.
给药频率将取决于在所用制剂中的具体抗GITR抗体的药代动力学参数。通常,临床医师施用组合物直到达到获得预期效果的剂量。本发明的抗体因此可以单一剂量施用,或者在一定时间内以两次或更多次剂量(其可包含相同或不同量的所需分子)施用,或者通过植入装置或导管连续输注施用。适当的剂量可通过使用适当的剂量反应数据确定。在某些实施方案中,可在延长的时间期限内向患者施用抗体。在某些实施方案中,抗体是每两周、每月、每两个月、每三个月、每四个月、每五个月或每 六个月给药。The frequency of administration will depend on the pharmacokinetic parameters of the particular anti-GITR antibody in the formulation used. Typically, the clinician will administer the composition until the dose that achieves the desired effect is achieved. The antibodies of the invention may therefore be administered in a single dose, or in two or more doses (which may comprise the same or different amounts of the desired molecule) over a period of time, or by continuous infusion via an implant device or catheter. The appropriate dose can be determined by using appropriate dose response data. In certain embodiments, the antibody can be administered to the patient over an extended period of time. In certain embodiments, the antibody is administered every two weeks, every month, every two months, every three months, every four months, every five months, or every six months.
药物组合物的施用途径是根据已知方法,例如,经口、通过静脉内注射、腹膜内、脑内(实质内)、脑室内、肌内、眼内、动脉内、门脉内或病灶内途径;通过持续释放系统或通过植入装置。在某些实施方案中,组合物可通过弹丸注射或通过连续输注或通过植入装置施用。The administration route of the pharmaceutical composition is according to known methods, for example, orally, by intravenous injection, intraperitoneal, intracerebral (intrinsic), intraventricular, intramuscular, intraocular, intraarterial, intraportal or intralesional. Route; through a sustained release system or through an implanted device. In certain embodiments, the composition can be administered by bolus injection or by continuous infusion or by an implant device.
组合物还可经由植入膜、海绵或其上吸收或胶囊包封所需分子的另一种适当的材料被局部施用。在某些实施方案中,当使用植入装置时,所述装置可被植入到任何合适的组织或器官中,并且可经由扩散、定时释放的大丸剂、或连续施用递送所需分子。The composition may also be topically applied via an implanted membrane, sponge or another suitable material on which the desired molecule is absorbed or encapsulated. In certain embodiments, when an implant device is used, the device can be implanted into any suitable tissue or organ and the desired molecule can be delivered via diffusion, timed release bolus, or continuous administration.
可以理解的是,能够使用本发明的免疫缀合物替换或补充抗GITR抗体来进行任何治疗。It will be appreciated that any treatment can be performed by replacing or supplementing the anti-GITR antibody with the immunoconjugate of the invention.
VIII.用于诊断和检测的方法和组合物VIII. Methods and compositions for diagnosis and detection
在某些实施方案中,本文中提供的任何抗GITR抗体或其抗原结合片段可以用于检测GITR在生物样品中的存在。术语“检测”用于本文中时,包括定量或定性检测,示例性的检测方法可以涉及免疫组织化学、免疫细胞化学、流式细胞术(例如,FACS)、抗体分子复合的磁珠、ELISA测定法、PCR-技术(例如,RT-PCR)。在某些实施方案中,生物样品是血、血清或生物来源的其他液体样品。在某些实施方案中,生物样品包含细胞或组织。在一些实施方案中,生物样品来自过度增生性或癌性病灶。In certain embodiments, any of the anti-GITR antibodies or antigen-binding fragments thereof provided herein can be used to detect the presence of GITR in a biological sample. The term "detection" as used herein, includes quantitative or qualitative detection, and exemplary detection methods may involve immunohistochemistry, immunocytochemistry, flow cytometry (eg, FACS), magnetic binding of antibody molecules, ELISA assays. Method, PCR-technology (for example, RT-PCR). In certain embodiments, the biological sample is a blood, serum or other liquid sample of biological origin. In certain embodiments, the biological sample comprises cells or tissues. In some embodiments, the biological sample is from a hyperproliferative or cancerous lesion.
在一个实施方案中,提供用于诊断或检测方法的抗GITR抗体。在另一个方面中,提供检测GITR在生物样品中的存在的方法。在某些实施方案中,方法包含检测GITR蛋白在生物样品中的存在。在某些实施方案中,GITR是人GITR。在某些实施方案中,所述方法包括将生物样品与如本文所述的抗GITR抗体在允许抗GITR抗体与GITR结合的条件下接触,并检测在抗GITR抗体和GITR之间是否形成复合物。复合物的形成表示存在GITR。该方法可以是体外或体内方法。在一个实施方案中,抗GITR抗体被用于选择适合利用抗GITR抗体的治疗的受试者,例如其中GITR是用于选择所述受试者的生物标记物。In one embodiment, an anti-GITR antibody for use in a diagnostic or detection method is provided. In another aspect, a method of detecting the presence of a GITR in a biological sample is provided. In certain embodiments, the methods comprise detecting the presence of a GITR protein in a biological sample. In certain embodiments, the GITR is a human GITR. In certain embodiments, the method comprises contacting a biological sample with an anti-GITR antibody as described herein under conditions that permit binding of the anti-GITR antibody to GITR, and detecting whether a complex is formed between the anti-GITR antibody and the GITR . The formation of the complex indicates the presence of GITR. The method can be an in vitro or in vivo method. In one embodiment, an anti-GITR antibody is used to select a subject suitable for treatment with an anti-GITR antibody, for example wherein GITR is a biomarker for selecting the subject.
在一个实施方案中,可以使用本发明抗体诊断癌症或肿瘤,例如评价(例如,监测)对象中本文所述疾病(例如,癌症或肿瘤)的治疗或进展、其诊断和/或分期。在某些实施方案中,提供标记的抗GITR抗体。标记包括但不限于,被直接检测的标记或部分(如萦光标记、发色团标记、电子致密标记、化学发光标记和放射性标记),以及被间接检测的部分,如酶或配体,例如,通过酶促反应或分子相互作用。示例性标记包括但不限于,放射性同位素 32P、 14C、 125I、 3H和 131I,萦光团如稀土螯合物或萦光素及其衍生物,罗丹明及其衍生物,丹酰(dansyl),伞形酮(umbelliferone),萦光素酶(luceriferase),例如,萤火虫萦光素酶和细菌萦光素酶(美国专利号4,737,456),萦光素,2,3-二氢酞嗪二酮,辣根过氧化物酶(HR),碱性磷酸酶,β-半乳糖苷酶,葡糖淀粉酶,溶解酶,糖类氧化酶,例如,葡萄糖氧化酶,半乳糖氧化酶,和葡萄糖-6-磷酸脱氢酶,杂环氧化酶如尿酸酶和黄嘌呤氧化酶,以及利用过氧化氢氧化染料前体的酶如HR,乳过氧化物酶,或微过氧化物酶(microperoxidase),生物素/亲和素,自旋标记,噬菌体标记,稳定的自由基,等等。 In one embodiment, a cancer or tumor can be diagnosed using an antibody of the invention, for example, to assess (eg, monitor) the treatment or progression, diagnosis, and/or staging of a disease (eg, cancer or tumor) described herein in a subject. In certain embodiments, a labeled anti-GITR antibody is provided. Labels include, but are not limited to, directly detected labels or moieties (such as fluorescent labels, chromophore labels, electron dense labels, chemiluminescent labels, and radioactive labels), as well as indirectly detected portions, such as enzymes or ligands, such as By enzymatic reaction or molecular interaction. Exemplary labels include, but are not limited to, radioisotopes 32 P, 14 C, 125 I, 3 H, and 131 I, iridium groups such as rare earth chelates or ruthenium and its derivatives, rhodamine and its derivatives, Dan Dansyl, umbelliferone, Luceriferase, for example, firefly luciferase and bacterial luciferase (U.S. Patent No. 4,737,456), luciferin, 2,3-dihydrogen Pyridazinone, horseradish peroxidase (HR), alkaline phosphatase, beta-galactosidase, glucoamylase, lytic enzyme, carbohydrate oxidase, for example, glucose oxidase, galactose oxidase And glucose-6-phosphate dehydrogenase, heterocyclic oxidases such as uricase and xanthine oxidase, and enzymes utilizing hydrogen peroxide dye precursors such as HR, lactoperoxidase, or microperoxide Microperoxidase, biotin/avidin, spin labeling, phage labeling, stable free radicals, and more.
在本文中提供的任何发明的一些实施方案中,样品是在用抗GITR抗体治疗之前获得的。在一些实施方案中,样品是在用癌症药物治疗之前获得的。在一些实施方案中,样品是在癌症已经转移之后获得的。在一些实施方案中,样品是福尔马林固定、石蜡包膜(FFPE)的。在一些实施方案中,样品是活检(例如芯活检),手术标本(例如来自手术切除的标本),或细针吸出物。In some embodiments of any of the inventions provided herein, the sample is obtained prior to treatment with an anti-GITR antibody. In some embodiments, the sample is obtained prior to treatment with a cancer drug. In some embodiments, the sample is obtained after the cancer has metastasized. In some embodiments, the sample is formalin fixed, paraffin coated (FFPE). In some embodiments, the sample is a biopsy (eg, a core biopsy), a surgical specimen (eg, from a surgically resected specimen), or a fine needle aspirate.
在一些实施方案中,在治疗之前,例如,在起始治疗之前或在治疗间隔后的某次治疗之前检测GITR。In some embodiments, the GITR is detected prior to treatment, for example, prior to initiation of treatment or prior to treatment after the treatment interval.
在一些实施方案中,提供了一种治疗肿瘤或感染的方法,所述方法包括:对受试者(例如,样品)(例如,包含癌细胞的受试者样品)检验GITR的存在,因而确定GITR值,将GITR值与对照值比较,并且如果GITR值大于对照值,则向受试者施用治疗有效量的任选与一种或多种其他疗法组合的抗GITR抗体(例如,本文所述的抗GITR抗体),因而治疗肿瘤或感染。In some embodiments, a method of treating a tumor or infection is provided, the method comprising: examining a subject (eg, a sample) (eg, a sample of a subject comprising cancer cells) for detecting the presence of a GITR, thereby determining A GITR value, the GITR value is compared to a control value, and if the GITR value is greater than the control value, the subject is administered a therapeutically effective amount of an anti-GITR antibody, optionally in combination with one or more other therapies (eg, as described herein) Anti-GITR antibodies), thus treating tumors or infections.
IX本发明的示例性抗GITR抗体IX Exemplary Anti-GITR Antibodies of the Invention
Figure PCTCN2019083241-appb-000006
Figure PCTCN2019083241-appb-000006
Figure PCTCN2019083241-appb-000007
Figure PCTCN2019083241-appb-000007
本发明的这些以及其它方面和实施方案在附图(附图简述紧随其后)和以下的发明详述中得到描述并且示例于以下实施例中。上文以及整个本申请中所论述的任何或所有特征可以在本发明的各种实施方案中组合。以下实施例进一步说明本发明,然而,应理解实施例以说明而非限定的方式来描述,并且本领域技术人员可以进行多种修改。These and other aspects and embodiments of the present invention are described in the drawings (hereinafter briefly described below) and the following detailed description of the invention and are illustrated in the following examples. Any or all of the features discussed above and throughout this application may be combined in various embodiments of the invention. The invention is further illustrated by the following examples, which are to be understood by way of illustration and not limitation
实施例Example
实施例1.杂交瘤细胞的制备Example 1. Preparation of hybridoma cells
杂交瘤技术是通过融合两种细胞而同时保持两者的主要特征。这两种细胞分别是经抗原免疫的小鼠脾细胞和小鼠骨髓瘤细胞。被特异性抗原免疫的小鼠脾细胞(B淋巴细胞)的主要特征是它的抗体分泌功能,但不能在体外连续培养,小鼠骨髓瘤细胞则可在培养条件下无限分裂、增殖,即具有所谓永生性。在选择培养基的作用下,只有B细胞与骨髓瘤细胞融合的杂交细胞才能具有持续培养的能力,形成同时具备抗体分泌功能和保持细胞永生性两种特征的细胞克隆。本实验通过人GITR蛋白免疫小鼠,再获取小鼠的脾细胞和骨髓瘤细胞融合,获得能够表达阳性抗体的杂交瘤细胞。Hybridoma technology is achieved by fusing two cells while maintaining the main features of both. These two cells are antigen-immunized mouse spleen cells and mouse myeloma cells, respectively. The main feature of mouse spleen cells (B lymphocytes) immunized by specific antigens is its antibody secretion function, but it cannot be continuously cultured in vitro, and mouse myeloma cells can divide and proliferate indefinitely under culture conditions, that is, The so-called immortality. Under the action of the selective medium, only the hybrid cells in which the B cells are fused with the myeloma cells can have the ability to continue to culture, and form a cell clone having both the secretory function of the antibody and the immortality of the cells. In this experiment, mice were immunized with human GITR protein, and then spleen cells of the mouse were fused with myeloma cells to obtain hybridoma cells capable of expressing positive antibodies.
杂交瘤融合Hybridoma fusion
实验动物及免疫信息Laboratory animals and immune information
Figure PCTCN2019083241-appb-000008
Figure PCTCN2019083241-appb-000008
电融合皿准备:用70%乙醇彻底浸泡电融合皿,并于超净台中吹干备用。Prepare the electrofusion dish: Thoroughly soak the electrofusion dish with 70% ethanol and blow dry in a clean bench.
分离脾细胞:颈脱位将小鼠处死,用75%酒精消毒体表5min,随即放入超净台内小鼠解剖板上,左侧卧位,用7号针头固定四肢。无菌打开腹腔取出脾脏,用基础培养基(配置方法如下表)洗涤,并仔细去掉周围附着的结缔组织。随后将脾脏转移到另一个盛有基础培养基的平皿中。以弯头针头压住脾脏,用小针头在脾脏上插孔,并用镊子挤压,使脾细胞充分释放,制成脾细胞悬液。细胞悬液经70μM细胞筛网过滤后用30ml基础培养基洗一遍,1200rpm离心6min。Isolation of spleen cells: The mice were sacrificed by cervical dislocation, and the body surface was sterilized with 75% alcohol for 5 min, and then placed on the anatomical plate of the mouse in the ultra-clean table, and the left side was placed, and the limbs were fixed with a 7-gauge needle. The spleen was removed by aseptic opening of the abdominal cavity, washed with a basal medium (configuration method as follows), and the connective tissue attached thereto was carefully removed. The spleen was then transferred to another dish containing the basal medium. Press the spleen with a elbow needle, insert a small needle into the spleen, and squeeze with the scorpion to fully release the spleen cells to make a spleen cell suspension. The cell suspension was filtered through a 70 μM cell sieve, washed once with 30 ml of basal medium, and centrifuged at 1200 rpm for 6 min.
Figure PCTCN2019083241-appb-000009
Figure PCTCN2019083241-appb-000009
裂解红细胞:去除上清,用10ml RBC裂解缓冲液(GIBCO)重悬细胞。然后再加入20ml RBC裂解缓冲液。悬液静置5min后1100rpm离心6min。去上清后用10ml基础培养基重悬细胞,然后再加入30ml基础培养基,1100rpm离心6min。去除上清后,细胞重悬于20ml基础培养基中并计数。Lysis of red blood cells: The supernatant was removed and the cells were resuspended in 10 ml RBC lysis buffer (GIBCO). Then add 20 ml of RBC lysis buffer. The suspension was allowed to stand for 5 min and centrifuged at 1100 rpm for 6 min. After removing the supernatant, the cells were resuspended in 10 ml of basal medium, and then 30 ml of basal medium was added, and centrifuged at 1100 rpm for 6 min. After removing the supernatant, the cells were resuspended in 20 ml of basal medium and counted.
电融合:用20ml基础培养基重悬小鼠骨髓瘤细胞SP2/0细胞(ATCC)并计数。将SP2/0 和脾细胞以1:2~1:1的比例混合,1000rpm离心6min。去除上清后将混合的细胞重悬于10ml融合缓冲液(BTXpress)中。再加入15ml融合缓冲液,1000rpm离心5min,去除上清。重复上述步骤一遍后,用适量融合缓冲液重选细胞,调整混合细胞密度至1×10 7个细胞/ml。电融合仪的参数设置如下。每个电融合皿中加入2ml细胞悬液进行电融合。 Electrofusion: Mouse myeloma cells SP2/0 cells (ATCC) were resuspended in 20 ml of basal medium and counted. SP2/0 and spleen cells were mixed at a ratio of 1:2 to 1:1, and centrifuged at 1000 rpm for 6 min. After removing the supernatant, the mixed cells were resuspended in 10 ml of fusion buffer (BTXpress). Further, 15 ml of the fusion buffer was added, and the mixture was centrifuged at 1000 rpm for 5 minutes, and the supernatant was removed. After repeating the above steps, the cells were re-selected with an appropriate amount of fusion buffer, and the mixed cell density was adjusted to 1 × 10 7 cells/ml. The parameters of the electrofusion amplifier are set as follows. 2 ml of the cell suspension was added to each electrofusion dish for electrofusion.
ConditionCondition Mouse(SP2/0-ECF-F)Mouse(SP2/0-ECF-F)
Alignment:Alignment: 60v,30sec60v, 30sec
Membrane breaking:Membrane breaking: 1500V,30μs,3X1500V, 30μs, 3X
Post-fusion pulse:Post-fusion pulse: 60V,3sec60V, 3sec
电融合后铺板:细胞于电融合皿中室温静置5min。将细胞转移入离心管中,用筛选培养基(配置方法如下表)稀释细胞至1~2×10 4个细胞/ml。96孔板中每孔加入100μl细胞悬液。融合后第7天更换筛选培养基。培养第10天(或更久,根据细胞生长状态)后进行筛选。通过FACS(C6(BD Biosciences))检测筛选出表达特异性抗GITR抗体的杂交瘤细胞。 Plate after electrofusion: The cells were allowed to stand in an electric fusion dish for 5 min at room temperature. The cells were transferred to a centrifuge tube, and the cells were diluted to 1 to 2 × 10 4 cells/ml using a screening medium (the configuration method is as follows). 100 μl of the cell suspension was added to each well of a 96-well plate. The screening medium was changed on the 7th day after the fusion. Screening was performed 10 days after culture (or longer, depending on the state of cell growth). Hybridoma cells expressing a specific anti-GITR antibody were screened by FACS (C6 (BD Biosciences)).
Figure PCTCN2019083241-appb-000010
Figure PCTCN2019083241-appb-000010
阳性杂交瘤细胞亚克隆Positive hybridoma cell subcloning
亚克隆步骤:准备一块96孔板,第2至第8列每孔加入200μl如上所述的基础培养基。将上述融合筛选出的阳性孔的细胞制成细胞悬液并加入第1列。将第1列细胞悬液取100μl加入第2列,充分混匀后取100μl加入下一列。重复上述步骤,直至最后一列体积变为300μl;静置96孔板15min,显微镜下观察计数。取100个细胞对应的体积加入20ml如上所述的基础培养基中,并混匀铺板,每孔200μl。一周后显微镜下观察,判断并标记出单克隆孔,待测挑出阳性孔。Subcloning step: A 96-well plate was prepared, and 200 μl of the basal medium as described above was added to each of the 2nd to 8th columns. The cells of the positive wells selected by the above fusion were made into cell suspensions and added to the first column. 100 μl of the cell suspension of column 1 was added to the second column, and after thorough mixing, 100 μl was added to the next column. The above procedure was repeated until the last column volume became 300 μl; the 96-well plate was allowed to stand for 15 min, and the count was observed under a microscope. The corresponding volume of 100 cells was added to 20 ml of the basal medium as described above, and the plates were mixed and mixed at 200 μl per well. After one week, the microscope was observed under the microscope, and the monoclonal wells were judged and labeled, and the positive wells were picked out.
细胞冻存:观察细胞状态,等细胞生长良好,活力>90%时,1000rpm离心5min,去除上清。用冻存液(45.5%FBS,44.5%RPMI-1640,10%DMSO)重悬细胞至1×10 7个细胞/ml,分装至冻存管,放入程序降温盒中,-80℃冻存。 Cryopreservation of cells: Observe the state of the cells, and wait for the cells to grow well. When the viability is >90%, centrifuge at 1000 rpm for 5 min to remove the supernatant. Resuspend the cells to 1×10 7 cells/ml with cryopreservation solution (45.5% FBS, 44.5% RPMI-1640, 10% DMSO), dispense into a cryotube, place in a programmed cooling box, and freeze at -80 °C. Save.
实施例2.嵌合抗体的生产和纯化Example 2. Production and purification of chimeric antibodies
本发明利用分子生物学技术,获得抗GITR阳性杂交瘤细胞中的抗体序列,并利用其构建人鼠嵌合抗体。The present invention utilizes molecular biological techniques to obtain antibody sequences in anti-GITR-positive hybridoma cells, and uses the same to construct a human-mouse chimeric antibody.
杂交瘤测序Hybridoma sequencing
RNA抽提:新鲜细胞,300g离心5min,去除上清,沉淀中加入500μl LY缓冲液(Biomiga)(在使用前每1ml加入20μlβ巯基乙醇),混匀至澄清。加入到DNA去除管中,13000rpm离心2min,收集流穿液。按1/2的比例向流穿液中加入100%乙醇,混匀5次至澄清。将澄清的溶液加入到RNA收集管中,13000rpm离心1min去除液体,加入500μl RB(Recovery Buffer,回收缓冲液)(Takara),13000rpm离心30s,再加入500μl RNA洗涤缓冲液(Biomiga)(用之前加入乙醇),离心30s,重复一遍上述过程后,离心彻底挥发去除乙醇后,向收集柱中加入30μl DEPC水,12000g离心2min,收集洗脱液。测定RNA浓度。RNA extraction: fresh cells, centrifuged at 300 g for 5 min, the supernatant was removed, and 500 μl of LY buffer (Biomiga) was added to the pellet (20 μl of β-mercaptoethanol was added per 1 ml before use) and mixed until clear. It was added to a DNA removal tube, centrifuged at 13,000 rpm for 2 min, and the flow-through was collected. 100% ethanol was added to the flow through solution at a ratio of 1/2, and mixed 5 times to clarify. Add the clarified solution to the RNA collection tube, centrifuge at 13000 rpm for 1 min to remove the liquid, add 500 μl RB (Recovery Buffer, Recovery Buffer) (Takara), centrifuge at 13000 rpm for 30 s, and add 500 μl RNA Wash Buffer (Biomiga). Ethanol), centrifugation for 30 s, repeat the above process, centrifuge to completely evaporate the ethanol, then add 30 μl of DEPC water to the collection column, centrifuge at 12000 g for 2 min, and collect the eluate. The RNA concentration was determined.
利用PrimeScript II 1st Strand cDNA Synthesis Kit(Takara)反转录获得cDNA:cDNA was obtained by reverse transcription using PrimeScript II 1st Strand cDNA Synthesis Kit (Takara):
配置反应体系I如下:The reaction system I is configured as follows:
Figure PCTCN2019083241-appb-000011
Figure PCTCN2019083241-appb-000011
65℃温育5min后,迅速置冰上冷却。向反应体系I中加入下列反转录体系,总量为20μl:After incubation at 65 ° C for 5 min, it was quickly cooled on ice. The following reverse transcription system was added to Reaction System I in a total amount of 20 μl:
Figure PCTCN2019083241-appb-000012
Figure PCTCN2019083241-appb-000012
缓慢混匀后按下列条件进行反转录翻译:42℃60min→95℃5min,然后放冰上冷却,获得cDNA。After slowly mixing, reverse transcription translation was carried out under the following conditions: 42 ° C for 60 min → 95 ° C for 5 min, and then cooled on ice to obtain cDNA.
将cDNA连接T载体:Link the cDNA to the T vector:
PCR分别扩增重链和轻链可变区,PCR反应体系如下:The heavy and light chain variable regions were amplified by PCR, and the PCR reaction system was as follows:
Figure PCTCN2019083241-appb-000013
Figure PCTCN2019083241-appb-000013
PCR反应条件如下:The PCR reaction conditions are as follows:
Figure PCTCN2019083241-appb-000014
Figure PCTCN2019083241-appb-000014
取4.5μl上述PCR反应获得的PCR产物,加入0.5μl pMD20-T载体(Clontech),5μl Ligation Mighty Mix(Takara),轻轻混匀,于37℃反应2h,获得连接产物。4.5 μl of the PCR product obtained by the above PCR reaction was added, 0.5 μl of pMD20-T vector (Clontech), 5 μl of Ligation Mighty Mix (Takara), gently mixed, and reacted at 37 ° C for 2 hours to obtain a ligation product.
表6.小鼠抗GITR抗体的重链可变区(VH)引物(Primer Mix 1)Table 6. Heavy chain variable region (VH) primers for mouse anti-GITR antibodies (Primer Mix 1)
Figure PCTCN2019083241-appb-000015
Figure PCTCN2019083241-appb-000015
Figure PCTCN2019083241-appb-000016
Figure PCTCN2019083241-appb-000016
表7.小鼠抗GITR抗体的轻链可变区(VL)引物(Primer Mix 2):Table 7. Light chain variable region (VL) primers for mouse anti-GITR antibodies (Primer Mix 2):
Figure PCTCN2019083241-appb-000017
Figure PCTCN2019083241-appb-000017
转化细胞:Transformed cells:
-80℃取出TOP10感受态细胞(天根生化科技(北京)有限公司),冰上融化,取上述获得的连接产物5μl加入到融化的TOP10感受态细胞中,混匀后冰上孵育30min。42℃热激90s后迅速冰上冷却2min,向EP管中补加900μl LB培养基(生工生物工程(上海)股份有限公司),37℃,220rpm摇床培养1h。3000g离心2min,吸除800μl上清,用剩余的培养基将菌体重悬并涂布在氨苄青霉素抗性的平板上。于37℃培养过夜,挑克隆测序。TOP10 competent cells (Tiangen Biochemical Technology (Beijing) Co., Ltd.) were taken at -80 °C, and thawed on ice. 5 μl of the ligation product obtained above was added to the thawed TOP10 competent cells, mixed, and incubated on ice for 30 min. After heat shock at 42 °C for 90 s, it was rapidly cooled on ice for 2 min, and 900 μl of LB medium (Biotech (Shanghai) Co., Ltd.) was added to the EP tube, and cultured at 37 ° C for 1 h at 220 rpm shaker. After centrifugation at 3000 g for 2 min, 800 μl of the supernatant was aspirated, and the bacteria were suspended and coated on the ampicillin-resistant plate with the remaining medium. Incubate overnight at 37 ° C and pick and clone.
构建嵌合抗体Construction of chimeric antibodies
PCR扩增已经测序的实施例1中杂交瘤产生的鼠抗GITR抗体VH及VL区PCR amplification of the VH and VL regions of the mouse anti-GITR antibody produced by the hybridoma of Example 1 which has been sequenced
PCR体系如下:The PCR system is as follows:
Figure PCTCN2019083241-appb-000018
Figure PCTCN2019083241-appb-000018
Figure PCTCN2019083241-appb-000019
Figure PCTCN2019083241-appb-000019
*对于VH扩增,应用Primer Mix 1,对于VL扩增,应用Primer Mix 2;* For VH amplification, use Primer Mix 1, for VL amplification, apply Primer Mix 2;
^为上述测序正确的pMD20-T质粒^Sequencing the correct pMD20-T plasmid for the above sequencing
切胶回收PCR扩增产物。The PCR amplification product was recovered by gel cutting.
同源重组反应:Homologous recombination reaction:
同源重组体系如下:The homology recombination system is as follows:
Figure PCTCN2019083241-appb-000020
Figure PCTCN2019083241-appb-000020
37℃反应30min,获得重组产物。重组产物转化TOP10感受态细胞,并挑取单克隆测序,选择包含插入方向正确的质粒的克隆作为阳性克隆,保存阳性克隆。The reaction was carried out at 37 ° C for 30 min to obtain a recombinant product. The recombinant product was transformed into TOP10 competent cells, and monoclonal sequencing was picked. The clone containing the plasmid with the correct insertion direction was selected as a positive clone, and the positive clone was preserved.
嵌合抗体的表达和纯化Expression and purification of chimeric antibodies
从上文获得的阳性克隆中提取包含抗GITR抗体的质粒。A plasmid containing an anti-GITR antibody was extracted from the positive clone obtained above.
根据所需转染体积传代293F细胞(Invitrogen),转染前一天将细胞密度调整至1.5×10 6个细胞/ml。转染当天细胞密度约为3×10 6个细胞/ml。取终体积1/10的F17培养基(Gibco,A13835-01)作为转染缓冲液,加入适当的质粒,混匀。加合适的聚乙烯亚胺(PEI)(Polysciences,23966)到质粒中(质粒与PEI的比例在293F细胞中为1:3),混匀后室温孵育10min,获得DNA/PEI混合物。用DNA/PEI混合物重悬细胞后,36.5℃,8%的CO 2。24h后补加转染体积2%的FEED(Sigma),于36.5℃,120rpm,8%的CO 2条件下培养。连续培养至第6天或者细胞活力≤60%时,收集细胞上清进行纯化。 293F cells (Invitrogen) were passaged according to the desired transfection volume, and the cell density was adjusted to 1.5 × 10 6 cells/ml one day before transfection. The cell density on the day of transfection was approximately 3 x 10 6 cells/ml. A final volume of 1/10 of F17 medium (Gibco, A13835-01) was used as a transfection buffer, and an appropriate plasmid was added and mixed. Add appropriate polyethyleneimine (PEI) (Polysciences, 23966) to the plasmid (the ratio of plasmid to PEI is 1:3 in 293F cells), mix and incubate for 10 min at room temperature to obtain a DNA/PEI mixture. After resuspending the cells with the DNA/PEI mixture, 36.5 ° C, 8% CO 2 . After 24 h, a transfection volume of 2% FEED (Sigma) was added and cultured at 36.5 ° C, 120 rpm, 8% CO 2 . When the culture was continued until day 6 or when the cell viability was ≤ 60%, the cell supernatant was collected for purification.
将纯化使用的重力柱使用0.5M NaOH过夜处理,玻璃瓶等用蒸馏水洗净后在180℃干烤4h,获得纯化柱。纯化前将收集的培养基4500rpm离心30min,弃掉细胞。再将上清使用0.22μl的滤器过滤。每管装填1ml Protein A,并使用10ml结合缓冲液(磷酸钠20mM.NaCl150mM,PH7.0)平衡。将过滤后的上清加入纯化柱后使用15ml结合缓冲液再平衡。加5ml洗脱缓冲液(柠檬酸+柠檬酸钠0.1M,PH3.5),收集洗脱液,每1ml的洗脱液加入80μl Tris-HCl。将收集的抗体超滤浓缩交换到PBS(Gibco,70011-044)中,并检测浓度。The gravity column used for purification was treated with 0.5 M NaOH overnight, and the glass bottle and the like were washed with distilled water and then dry-baked at 180 ° C for 4 hours to obtain a purification column. The collected medium was centrifuged at 4500 rpm for 30 min before purification, and the cells were discarded. The supernatant was then filtered using a 0.22 μl filter. Each tube was filled with 1 ml of Protein A and equilibrated with 10 ml of binding buffer (sodium phosphate 20 mM. NaCl 150 mM, pH 7.0). The filtered supernatant was added to the purification column and re-equilibrated with 15 ml of binding buffer. 5 ml of elution buffer (citric acid + sodium citrate 0.1 M, pH 3.5) was added, and the eluate was collected, and 80 μl of Tris-HCl was added per 1 ml of the eluate. The collected antibodies were concentrated by ultrafiltration into PBS (Gibco, 70011-044), and the concentration was measured.
本发明获得的2个嵌合抗体(CH22F4和CH37G5)的CDR、轻链可变区和重链可变区,轻链和重链的氨基酸序列,以及序列编号请参见上表1-3。The CDRs, the light chain variable region and the heavy chain variable region, the amino acid sequences of the light chain and the heavy chain of the two chimeric antibodies (CH22F4 and CH37G5) obtained by the present invention, and the sequence numbers are shown in Table 1-3 above.
本发明所用的对照抗体为专利申请US20130183321A1(GITR,INC.(Cambridge,MA,US))中报道的GITR抗体,其轻链和重链序列在US20130183321A1中分别为SEQ ID NO:44和SEQ ID NO:54,下文中将其简称为TRX518。The control antibody used in the present invention is a GITR antibody reported in the patent application US20130183321A1 (GITR, INC. (Cambridge, MA, US)), the light and heavy chain sequences thereof are SEQ ID NO: 44 and SEQ ID NO in US20130183321A1, respectively. :54, which will be referred to as TRX518 in the following.
实施例3 生物膜薄层干涉技术测定本发明的嵌合抗体与抗原的结合动力学Example 3 Biofilm thin layer interference technique for determining the binding kinetics of the chimeric antibody of the present invention to an antigen
采用生物膜薄层干涉测定技术(ForteBio)测定本发明抗体结合人GITR的平衡解离常数(KD)。ForteBio亲和力测定按照现有的方法(Estep,P等人,High throughput solution Based measurement of antibody-antigen affinity and epitope binning.MAbs,2013.5(2):第270-8页)进 行。The equilibrium dissociation constant (KD) of the antibody of the present invention in combination with human GITR was determined by biofilm thin layer interferometry (ForteBio). The ForteBio affinity assay was performed according to the existing method (Estep, P et al, High throughput solution Based measurement of antibody-antigen affinity and epitope binning. MAbs, 2013.5(2): p. 270-8).
实验开始前半个小时,根据样品数量,取合适数量的AMQ(Pall,1506091)(用于样品检测)或AHQ(Pall,1502051)(用于阳性对照检测)传感器浸泡于SD buffer(PBS 1×,BSA0.1%,Tween-20 0.05%)中。Half an hour before the start of the experiment, according to the number of samples, take the appropriate amount of AMQ (Pall, 1506091) (for sample detection) or AHQ (Pall, 1502051) (for positive control detection) sensor soaked in SD buffer (PBS 1 ×, BSA 0.1%, Tween-20 0.05%).
取100μl的SD缓冲液、抗体(CH22F4、CH37G5和TRX518)、抗原(包括人GITR、及食蟹猴GITR,均自Acrobiosystems购买)分别加入到96孔黑色聚苯乙烯半量微孔板(Greiner,675076)中。根据样品位置布板,选择传感器位置。仪器设置参数如下:运行步骤:Baseline、Loading~1nm、Baseline、Association和Dissociation;各个步骤运行时间取决于样品结合和解离速度,转速为400rpm,温度为30℃。使用ForteBio分析软件分析K D值。 100 μl of SD buffer, antibodies (CH22F4, CH37G5 and TRX518), antigens (including human GITR, and cynomolgus GITR, purchased from Acrobiosystems) were added to 96-well black polystyrene half-well microplates (Greiner, 675076). )in. Select the sensor location based on the sample position layout. The instrument setting parameters are as follows: running steps: Baseline, Loading ~ 1 nm, Baseline, Association, and Dissociation; the running time of each step depends on the sample binding and dissociation speed, the rotation speed is 400 rpm, and the temperature is 30 °C. Analysis of K D values using ForteBio analysis software.
在以上测定法所述的实验中,抗体CH22F4、CH37G5的亲和力如表8所示:In the experiments described in the above assays, the affinities of the antibodies CH22F4 and CH37G5 are shown in Table 8:
表8.ForteBio检测抗原抗体结合的亲和力常数(平衡解离常数)Table 8. Affinity constants (equilibrium dissociation constants) for ForteBio detection of antigen-antibody binding
Figure PCTCN2019083241-appb-000021
Figure PCTCN2019083241-appb-000021
在以上试验中,嵌合抗体CH22F4、CH37G5的K D值分别为1.96E-09M、2.04E-09M,与对照组的TRX518相比,本研究中的抗体具有更优的K D值。 In the above test, a chimeric antibody CH22F4, K D values were CH37G5 1.96E-09M, 2.04E-09M, TRX518 compared with the control group, the present study the antibody has a K D value is better.
实施例4 嵌合抗体和过表达人GITR的CHO-S细胞的结合实验Example 4 Binding experiment of chimeric antibody and CHO-S cells overexpressing human GITR
本研究利用流式细胞仪检测了梯度稀释的本发明的嵌合抗体与表面过表达人GITR的CHO-S稳定细胞株的结合情况。In this study, the binding of a gradient-diluted chimeric antibody of the present invention to a CHO-S stable cell strain overexpressing human GITR was detected by flow cytometry.
将编码人GITR(SEQ ID NO:41)的cDNA克隆到pCHO1.0载体(Invitrogen)中,将获得的质粒转染到CHO-S细胞(Invitrogen,ExpiCHO TM Expression System Kit,货号:A29133),产生过表达人GITR的CHO-S细胞(CHO-hGITR)。 Encoding human GITR (SEQ ID NO: 41) cDNA was cloned into pCHO1.0 vector (Invitrogen), a plasmid obtained was transfected into CHO-S cells (Invitrogen, ExpiCHO TM Expression System Kit , NO: A29133), generated CHO-S cells (CHO-hGITR) overexpressing human GITR.
将CHO-hGITR细胞计数,并稀释至2×10 6个细胞/ml,向U型底96孔板中加入100μl/孔。400g离心5min,去除细胞培养基。将样品(分别是嵌合抗体CH22F4、CH37G5,以及TRX518)(抗体稀释方法为:最高抗体浓度为400nM,三倍稀释在PBS中,总共测试了12个浓度)加入U型板并重悬细胞,100μl/孔,冰上静置30min。400g离心5min去除上清,PBS洗细胞1遍。400g离心5min去除PBS,每孔加入100μl抗人Fc的PE标记的二抗(SoutherBiotech;2040-09)(1:200稀释于PBS中),在冰上避光孵育30min。400g离心5min去除上清,PBS洗细胞1遍。用80μl 1×PBS重悬细胞,FACS检测。 CHO-hGITR cells were counted and diluted to 2 x 10 6 cells/ml, and 100 μl/well was added to a U-bottom 96-well plate. Centrifuge for 5 min at 400 g to remove the cell culture medium. Samples (chimeric antibodies CH22F4, CH37G5, and TRX518, respectively) (antibody dilution method: maximum antibody concentration of 400 nM, three-fold dilution in PBS, a total of 12 concentrations tested) were added to the U-plate and resuspended cells, 100 μl / Hole, allowed to stand on ice for 30 min. The supernatant was removed by centrifugation at 400 g for 5 min, and the cells were washed 1 time with PBS. PBS was removed by centrifugation at 400 g for 5 min, and 100 μl of anti-human Fc-labeled secondary antibody (Souther Biotech; 2040-09) (diluted 1:200 in PBS) was added to each well, and incubated on ice for 30 min in the dark. The supernatant was removed by centrifugation at 400 g for 5 min, and the cells were washed 1 time with PBS. The cells were resuspended in 80 μl of 1×PBS and detected by FACS.
在以上测定法所述的实验中,抗体CH22F4、CH37G5和CHO-hGITR细胞的结合情况如图1所示。In the experiments described in the above assay, the binding of antibodies CH22F4, CH37G5 and CHO-hGITR cells is shown in Figure 1.
在以上试验中,抗体CH22F4、CH37G5均结合CHO-S细胞上过表达的人GITR分子,EC50分别为0.4568nM、0.9069nM,与TRX518相比,结合能力相似。In the above experiments, the antibodies CH22F4 and CH37G5 all bind to human GITR molecules overexpressed on CHO-S cells with EC50 of 0.4568 nM and 0.9069 nM, respectively, and the binding ability is similar to that of TRX518.
实施例5 嵌合抗体的人源化Example 5 Humanization of chimeric antibodies
将实施例2得到的嵌合抗体进行人源化。并经过以下步骤进行人源化:The chimeric antibody obtained in Example 2 was humanized. And through the following steps for humanization:
①确定CDR环结构;1 determining the structure of the CDR ring;
②在人种系序列数据库为重链和轻链的每个V/J区域找到最接近的同源序列;2 Find the closest homologous sequence in each V/J region of the heavy and light chains in the human germline sequence database;
③筛选与重链轻链最匹配的人种系以及最低量的回复突变;3 screening the human germline that best matches the heavy chain light chain and the lowest amount of back mutation;
④将嵌合抗体的CDR区构建至人的骨架区上;4 constructing the CDR regions of the chimeric antibody onto the human framework region;
⑤使用序列和结构特征,确定骨架区中起到维持CDR功能的氨基酸位置;5 using sequence and structural features to determine the position of the amino acid in the framework region that serves to maintain CDR function;
⑥在确定为重要的序列位置进行回复突变(返回到输入氨基酸类型);6 performing a back mutation at the sequence position determined to be important (returning to the input amino acid type);
⑦优化风险位点的氨基酸。7 Optimize the amino acids at the risk site.
本发明获得的2个人源化抗体(HZ22F4和HZ37G5)的CDR、轻链可变区和重链可变区,轻链和重链的氨基酸序列请参见如上文所述的表1-3。The CDRs, light chain variable regions and heavy chain variable regions of the two humanized antibodies (HZ22F4 and HZ37G5) obtained in the present invention, and the amino acid sequences of the light and heavy chains are described in Tables 1-3 as described above.
实施例6 ForteBio测定人源化抗体与抗原的结合动力学Example 6 Determination of Binding Kinetics of Humanized Antibody to Antigen by ForteBio
采用ForteBio测定法测定本发明的人源化抗体结合人GITR的平衡解离常数(K D)。ForteBio亲和力测定方法同实施例3,例外是使用的抗体是人源化抗体HZ22F4和HZ37G5。在以上测定法所述的实验中,抗体HZ22F4和HZ37G5的亲和力如表9所示: The present invention is measured using ForteBio assay humanized antibody binds to human GITR solution equilibrium dissociation constant (K D). The ForteBio affinity assay was the same as in Example 3 with the exception that the antibodies used were the humanized antibodies HZ22F4 and HZ37G5. In the experiments described in the above assays, the affinities of the antibodies HZ22F4 and HZ37G5 are shown in Table 9:
表9.ForteBio检测抗原抗体结合的亲和力常数Table 9. Affinity constants for antigen-antibody binding by ForteBio
Figure PCTCN2019083241-appb-000022
Figure PCTCN2019083241-appb-000022
在以上试验中,本文所述的人源化抗体HZ22F4、HZ37G5的K D值分别为4.58E-09M、4.83E-09M,与对照抗体相比,本研究中的人源化抗体具有更优的K D值。 In the above test, described herein, humanized antibodies HZ22F4, K D values were HZ37G5 4.58E-09M, 4.83E-09M, compared to the control antibodies, the present study of humanized antibodies have better K D value.
实施例7 人源化抗体和过表达人GITR的CHO-S细胞的结合实验Example 7 Binding experiment of humanized antibody and CHO-S cells overexpressing human GITR
本研究利用流式细胞仪检测了梯度稀释的本发明的人源化抗体与CHO-hGITR细胞的结合情况。试验方法同实施例4,例外是使用的抗体是人源化抗体HZ22F4和HZ37G5。结合情况如图2所示。This study examined the binding of a gradient-diluted humanized antibody of the present invention to CHO-hGITR cells using flow cytometry. The test method was the same as in Example 4 except that the antibodies used were humanized antibodies HZ22F4 and HZ37G5. The combination is shown in Figure 2.
在以上试验中,人源化抗体HZ22F4、HZ37G5结合CHO-S细胞上过表达的人GITR,EC50分别为0.5492nM、1.974nM,与TRX518相比,具有相似或更优的EC50数值,即相似或更优的结合能力。In the above experiments, the humanized antibodies HZ22F4 and HZ37G5 bind to human GITR overexpressed on CHO-S cells with EC50 of 0.5492 nM and 1.974 nM, respectively, and have similar or better EC50 values than TRX518, ie similar or Better combination ability.
实施例8 人源化抗体和过表达食蟹猴GITR的细胞的结合实验Example 8 Binding experiment of humanized antibody and cells overexpressing cynomolgus monkey GITR
本研究利用流式细胞仪检测了梯度稀释的本发明的人源化抗体与表面过表达食蟹猴GITR的CHO-S稳定细胞株的结合情况。In this study, the binding of a gradient-diluted humanized antibody of the present invention to a CHO-S stable cell line overexpressing cynomolgus monkey GITR was detected by flow cytometry.
将编码食蟹猴GITR(SEQ ID NO:42)的cDNA克隆到pCHO1.0载体(Invitrogen)上,将质粒转染到CHO-S细胞(Invitrogen,ExpiCHO TM Expression System Kit,货号:A29133),产生过表达食蟹猴GITR的CHO-S细胞(CHO-cynoGITR)。其余部分试验方法同实施例4,例外是使用的抗体是人源化抗体HZ22F4和HZ37G5,结合情况如图3所示。 Encoding cynomolgus monkey GITR (SEQ ID NO: 42) cDNA was cloned into pCHO1.0 vector (Invitrogen), the plasmid was transfected into CHO-S cells (Invitrogen, ExpiCHO TM Expression System Kit , NO: A29133), generated CHO-S cells (CHO-cynoGITR) overexpressing cynomolgus GITR. The rest of the test methods were the same as in Example 4 except that the antibodies used were humanized antibodies HZ22F4 and HZ37G5, and the binding conditions are shown in FIG.
在以上试验中,人源化抗体HZ22F4、HZ37G5结合CHO-S细胞上过表达的食蟹猴GITR,EC50分别为0.3533nM、0.9931nM,与TRX518相比,具有相似或更优的结合能力。In the above experiments, the humanized antibodies HZ22F4 and HZ37G5 bind to the cynomolgus monkey GITR overexpressed on CHO-S cells with EC50 of 0.3533 nM and 0.9931 nM, respectively, and have similar or superior binding ability compared to TRX518.
实施例9 MOA方法检测抗体的生物学活性Example 9 Detection of Biological Activity of Antibodies by MOA Method
针对GITR的激活性抗体可以与细胞表面GITR分子结合激活下游NF-kappa B信号通路。本研究使用内部构建的Hela-GITR-NF kappa B luciferase(以下简称Hela-GITR)的检测细胞株,通过检测萦光报告基因的表达反应出NF-kappa B信号的激活情况,从而检测本发明抗体的激活作用。The activating antibody against GITR can bind to the cell surface GITR molecule to activate the downstream NF-kappa B signaling pathway. In this study, an internally constructed Hela-GITR-NF kappa B luciferase (hereinafter referred to as Hela-GITR) cell line was detected, and the activation of NF-kappa B signal was detected by detecting the expression of the fluorescent reporter gene, thereby detecting the antibody of the present invention. Activation.
Hela-GITR-NF kappa B luciferase细胞株的构建Construction of Hela-GITR-NF kappa B luciferase cell line
将编码人GITR(序列见SEQ ID NO:41)的cDNA克隆到pCHO1.0载体(Invitrogen)上,将获得的质粒和NF-kappaB luciferace报告基因质粒(Promega)共转染到Hela细胞(ATCC,货号:CCL-2TM),产生过表达人GITR同时带有NF-kappaB luciferace报告基因系统的Hela细胞(Hela-GITR)。The cDNA encoding human GITR (sequence see SEQ ID NO: 41) was cloned into pCHO1.0 vector (Invitrogen), and the obtained plasmid and NF-kappaB luciferace reporter plasmid (Promega) were co-transfected into Hela cells (ATCC, Cat. No.: CCL-2TM), Hela cells (Hela-GITR) that overexpress the human GITR with the NF-kappaB luciferace reporter gene system.
取对数生长期的Hela-GITR细胞,弃培养上清,PBS(Gibco)洗一遍细胞。加入适量Trypsin(Gibco)于37℃、5%CO 2消化2min。加入4倍Trypsin体积的含10%FBS的DMEM培养基(ATCC),转移细胞至50ml离心管并计数,400g,离心5min。加入DMEM培养基(ATCC),重悬细胞至1×10 5个细胞/mL。将细胞加入96孔白色细胞培养板(Nunclon),50μl/孔。同时每孔加入50μl样品(本发明制备的人源化抗体HZ22F4、HZ37G5)及阳性对照TRX518),(抗体稀释方法为:最高抗体浓度为80nM,三倍稀释在测定缓冲液(2%FBS DMEM培养基)中,总共测试了10个浓度)。于37℃/5%CO 2培养箱中培养6小时。 HeLa-GITR cells in logarithmic growth phase were taken, the culture supernatant was discarded, and the cells were washed once with PBS (Gibco). Add appropriate amount of Trypsin (Gibco) and digest at 37 ° C, 5% CO 2 for 2 min. 4 times Trypsin volume of DMEM medium (ATCC) containing 10% FBS was added, and the cells were transferred to a 50 ml centrifuge tube and counted, 400 g, and centrifuged for 5 min. DMEM medium (ATCC) was added and the cells were resuspended to 1 x 10 5 cells/mL. The cells were added to a 96-well white cell culture plate (Nunclon) at 50 μl/well. At the same time, 50 μl of sample (humanized antibody HZ22F4, HZ37G5 prepared by the present invention) and positive control TRX518) were added to each well. (The antibody dilution method was: the highest antibody concentration was 80 nM, and the dilution was three times in the assay buffer (2% FBS DMEM culture). In the base), a total of 10 concentrations were tested). Incubate for 6 hours in a 37 ° C / 5% CO 2 incubator.
检测:提前将Bio-GloTM缓冲液(promega)融化,加入Bio-GloTM底物(promega),混匀,获得Bio-GloTM试剂。向如上所述培养了6个小时的细胞中加入Bio-GloTM试剂,100μl/孔。立即读数。Detection: Bio-GloTM buffer (promega) was thawed in advance, Bio-GloTM substrate (promega) was added, and mixed to obtain Bio-GloTM reagent. Bio-GloTM reagent, 100 μl/well, was added to the cells cultured for 6 hours as described above. Read immediately.
在以上试验中,实验结果如图4所示,抗体HZ22F4、HZ37G5均可以有效激活NF kappa B信号通路,EC50分别为0.3394nM、0.3208nM、与TRX518(1.139nM)相比,具有显著更优的激活能力。In the above experiments, the experimental results are shown in Figure 4. The antibodies HZ22F4 and HZ37G5 can effectively activate the NF kappa B signaling pathway with EC50 of 0.3394nM and 0.3208nM, respectively, which is significantly better than TRX518 (1.139nM). Activation ability.
实施例10 CD4T激活细胞实验Example 10 CD4T Activated Cell Experiment
本研究将抗体和激活过的CD4T细胞共同孵育,通过检测体系中IL-2和IFN-γ的相对表达量,从而反应出不同抗体对CD4T细胞的激活作用。In this study, antibodies and activated CD4T cells were incubated together to detect the relative expression of IL-2 and IFN-γ in the system, thereby reflecting the activation of CD4 T cells by different antibodies.
PBMC分离:取捐赠者新鲜血液50ml,添加2.5倍PBS,轻轻加入到FiColl(Thermo),分4管,400g离心30min,升速度为7,减速度为0。离心结束,轻轻取出离心管,吸取中间白色云雾状细胞群至PBS中,PBS洗2次。PBMC separation: Take 50 ml of fresh blood from the donor, add 2.5 times PBS, gently add to FiColl (Thermo), divide into 4 tubes, centrifuge at 400 g for 30 min, increase the speed to 7, and decelerate to 0. At the end of centrifugation, the tube was gently removed, and the middle white cloud-like cell population was aspirated into PBS and washed twice with PBS.
CD4 +T细胞分离:取如上所述分离的PBMC细胞,按照EasySep Human CD4 +T Cell Enrichment Kit(stem cell)说明书分离富集CD4 +T细胞,并使用T细胞培养基(配方见下表)重悬。 CD4 + T cell isolation: PBMC cells isolated as described above were isolated and enriched for CD4 + T cells according to the EasySep Human CD4 + T Cell Enrichment Kit (stem cell) instructions and used in T cell culture medium (recipe is shown in the table below). Hanging.
Figure PCTCN2019083241-appb-000023
Figure PCTCN2019083241-appb-000023
CD4 +T细胞激活:取上述分离获得的CD4 +T细胞,并用T细胞培养基调整细胞密度为1*10 6/ml,按照细胞:beads(1:1)加入Dynabeads Human T-Activator CD3/CD28(Invitrogen),于37℃,5%CO 2培养箱中培养一周。 CD4 + T cell activation: taking the above-described CD4 + T cells isolated and cell density was adjusted by a T cell culture medium of 1 * 10 6 / ml, according cells: beads (1: 1) added Dynabeads Human T-Activator CD3 / CD28 (Invitrogen), cultured in a 5% CO 2 incubator at 37 ° C for one week.
T细胞激活实验:取Nunc 96孔平底板(Nunclon),100μl/孔加入PBS稀释的0.25μg/ml的Purified NA/LE Mouse抗人CD3Clone UCHT1(BD Biosciences),37℃包被2小时,去除包被液。将上述激活一周的CD4 +T细胞去除beads,用T细胞培养基清洗两次,并将细胞密度调整为1*10 6/ml,每孔体积100μl细胞悬液加入上述96孔平底板中,,同时加入本发明的抗体(HZ22F4,HZ37G5)以及TRX518 100μl(抗体稀释方法为:最高抗体浓度为400nM,三倍稀释在T细胞培养基中,总共测试了10个浓度)以及Purified NA/LE Mouse抗人CD28Clone CD28(BD Biosciences)(终浓度为2μg/ml),培养3和5天,分别用Human IL-2Kit 1000Test和Human IFN gamma 1000test试剂盒(均购自cisbio)检测IL-2和IFN-γ的相对表达量(以DeltaF%计)。 T cell activation assay: Nunc 96 well flat bottom plate (Nunclon) was taken, 100 μl/well was added to PBS diluted 0.25 μg/ml of Purified NA/LE Mouse anti-human CD3Clone UCHT1 (BD Biosciences), and coated at 37 ° C for 2 hours to remove the package. Being liquid. The above-mentioned activated CD4 + T cells were removed for one week, washed twice with T cell culture medium, and the cell density was adjusted to 1*10 6 /ml, and 100 μl of the cell suspension per well was added to the 96-well flat bottom plate. The antibody of the present invention (HZ22F4, HZ37G5) and TRX518 100 μl were simultaneously added (the antibody dilution method was: the highest antibody concentration was 400 nM, three-fold dilution in T cell medium, a total of 10 concentrations were tested) and Purified NA/LE Mouse antibody Human CD28Clone CD28 (BD Biosciences) (final concentration 2 μg/ml) was cultured for 3 and 5 days, and IL-2 and IFN-γ were detected with Human IL-2 Kit 1000 Test and Human IFN gamma 1000 test kit (both purchased from cisbio). Relative expression level (in DeltaF%).
实验结果如表10和11及图5、6所示。本发明的抗体均可以在体外有效激活CD4 +T细胞。其中表中的数据单位为DeltaF%平均值。 The experimental results are shown in Tables 10 and 11 and Figures 5 and 6. The antibodies of the invention all efficiently activate CD4 + T cells in vitro. The data unit in the table is the DeltaF% average.
表10.IL2的相对表达量(DeltaF%平均值)Table 10. Relative expression levels of IL2 (DeltaF% average)
Figure PCTCN2019083241-appb-000024
Figure PCTCN2019083241-appb-000024
表11.IFN-γ的相对表达量(DeltaF%平均值)Table 11. Relative expression levels of IFN-γ (DeltaF% average)
Figure PCTCN2019083241-appb-000025
Figure PCTCN2019083241-appb-000025
实施例11 ADCC效应实验Example 11 ADCC effect experiment
针对GITR的IgG1亚型抗体可以与Treg表面高表达的GITR分子结合,进而介导ADCC效应清除Treg。本研究使用Promega的Jurkat-ADCCNF-AT luciferase效应细胞株(以下简称ADCC效应细胞)作为检测细胞株与作为靶细胞的CHO-hGITR细胞(如上所述)和本发明的抗体共孵育,通过检测萦光报告基因的表达反应出NF-AT信号的激活情况,从而检测抗体的ADCC活性。The IgG1 subtype antibody against GITR can bind to the GITR molecule with high expression on the surface of Treg, which in turn mediates the ADCC effect to clear Treg. In this study, Promega's Jurkat-ADCCNF-AT luciferase effector cell line (hereinafter referred to as ADCC effector cell) was used as a test cell strain and CHO-hGITR cells (as described above) as target cells, and the antibody of the present invention was incubated, and the test was carried out. The expression of the light reporter gene reflects the activation of the NF-AT signal, thereby detecting the ADCC activity of the antibody.
取对数生长期的ADCC效应细胞(根据Promega公司说明培养),离心去上清,PBS洗2遍,用检测培养基(5%low IgG血清的1640培养基(Gibco))重悬调整细胞密度为2*10 7/ml,取如上所述制备并孵育的CHO-hGITR靶细胞,离心去上清,用检测培养基重悬调整细胞密度为2*10 6/ml,将上述细胞按1:1比例混合,并加入到96孔白色细胞培养板(Nunclon),50μl/孔。同时每孔加入50μl样品(本发明制备的人源化抗体HZ22F4、HZ37G5)及阳性对照抗体(TRX518),(抗体稀释方法为:最高抗体终浓度为66.66nM,三倍稀释在检测培养基中,总共测试了12个浓度)。于37℃/5%CO 2培养箱中培养12小时。 ADCC effector cells in logarithmic growth phase (cultured according to Promega instructions), centrifuged to remove supernatant, washed twice with PBS, and resuspended in assay medium (5% low IgG serum in 1640 medium (Gibco)) to adjust cell density For 2*10 7 /ml, the CHO-hGITR target cells prepared and incubated as described above were centrifuged, and the supernatant was centrifuged, and the cell density was adjusted to 2*10 6 /ml by resuspending in the assay medium, and the cells were subjected to 1: 1 ratio was mixed and added to a 96-well white cell culture plate (Nunclon) at 50 μl/well. At the same time, 50 μl of sample (humanized antibody HZ22F4, HZ37G5 prepared by the present invention) and positive control antibody (TRX518) were added to each well. (The antibody dilution method was: the maximum antibody concentration was 66.66 nM, and the dilution was three times in the detection medium. A total of 12 concentrations were tested). Incubate for 12 hours in a 37 ° C / 5% CO 2 incubator.
检测:提前将Bio-GloTM缓冲液(promega)融化,加入Bio-GloTM底物(promega),混匀,得到Bio-GloTM试剂。向如上所述培养了12个小时的细胞中加入Bio-GloTM试剂,100μl/孔。立即读数。Detection: Bio-GloTM buffer (promega) was thawed in advance, Bio-GloTM substrate (promega) was added, and mixed to obtain Bio-GloTM reagent. Bio-GloTM reagent, 100 μl/well, was added to the cells cultured for 12 hours as described above. Read immediately.
在以上试验中,实验结果如图7所示,抗体HZ22F4、HZ37G5均可以有效激活ADCC下游的NF-AT信号通路,EC50分别为0.02958nM、0.04056nM、与TRX518相比,具有显著更优的EC50数值。In the above experiments, the experimental results are shown in Figure 7. The antibodies HZ22F4 and HZ37G5 can effectively activate the NF-AT signaling pathway downstream of ADCC. The EC50 is 0.02958nM and 0.04056nM, respectively. Compared with TRX518, it has a significantly better EC50. Value.
实施例12 抗肿瘤药效试验Example 12 Antitumor Efficacy Test
本实验采用MC38细胞接种hGITR转基因小鼠测定本发明的GITR抗体的抗肿瘤作用。In this experiment, MC38 cells were used to inoculate hGITR transgenic mice to determine the anti-tumor effect of the GITR antibody of the present invention.
人GITR转基因小鼠:Human GITR transgenic mice:
雌性C57Bl/6背景的人GITR转基因小鼠(约5周大)购自百奥赛图实验动物技术有限公司。小鼠在到达后驯化7天,随后开始研究。Female C57B1/6 background human GITR transgenic mice (approximately 5 weeks old) were purchased from Biotech Test Animal Technology Co., Ltd. The mice were domesticated for 7 days after arrival and the study was started.
细胞:cell:
小鼠MC38细胞购自南京银河生物医药有限公司,并严格按照说明书进行常规传代培养用于后续体内实验。离心收集细胞,在无菌PBS中重悬细胞并调整细胞密度为5×10 6个/ml。在第0天取0.2ml细胞悬液皮下接种至人GITR转基因小鼠右侧腹部区域中来建立MC38-hGITR荷瘤小鼠模型。 Mouse MC38 cells were purchased from Nanjing Yinhe Biomedical Co., Ltd. and routinely subcultured for subsequent in vivo experiments in strict accordance with the instructions. The cells were collected by centrifugation, resuspended in sterile PBS and adjusted to a cell density of 5 x 10 6 /ml. A MC38-hGITR tumor-bearing mouse model was established by subcutaneous inoculation of 0.2 ml of cell suspension on day 0 into the right abdomen region of human GITR transgenic mice.
给药:Dosing:
肿瘤细胞接种6天后检测各只小鼠瘤体积,挑选出瘤体积在87.4mm 3~228.4mm 3范围 内的小鼠按瘤体积平均分组(每组5只小鼠)分组,给药剂量和方式如表12所示,其中Antibody C为抗PD-1的单克隆抗体“Antibody C”(WO2017/133540)。分别在接种后第6、10、13、17天给药,在给药期间监测各组小鼠瘤体积和体重变化,监测频率均为2次/周,连续监测5周。在每次给药前测定体重和肿瘤体积,接种后第24天计算相对肿瘤抑制率(TGI%),计算公式如下:TGI%=100%*(h-IgG对照组肿瘤体积–治疗组肿瘤体积)/(h-IgG对照组肿瘤体积–h-IgG对照组给药前肿瘤体积),其中h-IgG对照组给药前肿瘤体积平均值为106mm 3。肿瘤体积测定:采用游标卡尺测定肿瘤的最大长轴(L)和最大宽轴(W),肿瘤体积按如下公式计算:V=L×W 2/2。采用电子天平测定体重。 Six days after tumor cell inoculation, the tumor volume of each mouse was measured, and mice with tumor volume ranging from 87.4 mm 3 to 228.4 mm 3 were selected and grouped according to the average tumor volume group (5 mice per group). As shown in Table 12, Antibody C is a monoclonal antibody "Antibody C" against anti-PD-1 (WO2017/133540). The mice were administered on the 6th, 10th, 13th and 17th day after the inoculation, and the tumor volume and body weight of each group were monitored during the administration period. The monitoring frequency was 2 times/week and the monitoring was continued for 5 weeks. Body weight and tumor volume were measured before each administration, and the relative tumor inhibition rate (TGI%) was calculated on the 24th day after inoculation, and the formula was calculated as follows: TGI%=100%* (h-IgG control tumor volume – treatment group tumor volume) / (h-IgG control tumor volume - tumor volume before administration of h-IgG control group), wherein the average tumor volume before administration of the h-IgG control group was 106 mm 3 . Tumor volume determination: The maximum long axis (L) and the largest broad axis (W) of the tumor were measured using a vernier caliper, and the tumor volume was calculated as follows: V = L x W 2 /2. The weight was measured using an electronic balance.
表12.实验设计表Table 12. Experimental Design Table
Figure PCTCN2019083241-appb-000026
Figure PCTCN2019083241-appb-000026
*每隔3或4天给药,共4次。* Dosing every 3 or 4 days for a total of 4 times.
^h-IgG购自Equitech-Bio,货号:SLH56-0001。^h-IgG was purchased from Equitech-Bio, article number: SLH56-0001.
肿瘤抑制率结果如图8,9和表13所示:在接种后第24天,HZ37G5和HZ22F4单药均显示很好的肿瘤抑制率,分别为:53%和50%,优于TRX518组的36%。HZ37G5和HZ22F4与抗PD-1抗体Antibody C联合用药,肿瘤抑制效果均明显优于Antibody C,HZ37G5,HZ22F4,以及TRX518单药组。其中HZ37G5和Antibody C联合用药组,以及HZ22F4和Antibody C联合用药组5只小鼠均有4只肿瘤完全消失。同时我们对小鼠体重进行检测,结果如图10所示小鼠体重无显著差异。因此,本发明的针对GITR分子的抗体对肿瘤有明显的抑制效果,且当所述抗体与PD-1抗体联合用药对肿瘤的抑制效果更加显著。The results of tumor inhibition rate are shown in Figures 8, 9 and Table 13. On the 24th day after inoculation, HZ37G5 and HZ22F4 showed good tumor inhibition rates, 53% and 50%, respectively, which was better than TRX518 group. 36%. HZ37G5 and HZ22F4 combined with anti-PD-1 antibody Antibody C, the tumor inhibition effect was significantly better than Antibody C, HZ37G5, HZ22F4, and TRX518 single drug group. Among them, 4 mice in the combination of HZ37G5 and Antibody C, and 5 mice in the combination of HZ22F4 and Antibody C completely disappeared. At the same time, we tested the body weight of the mice, and the results showed no significant difference in the body weight of the mice as shown in FIG. Therefore, the antibody against the GITR molecule of the present invention has a significant inhibitory effect on tumors, and when the antibody is combined with the PD-1 antibody, the inhibitory effect on the tumor is more remarkable.
表13.第24天肿瘤抑制率Table 13. Tumor inhibition rate on day 24
Figure PCTCN2019083241-appb-000027
Figure PCTCN2019083241-appb-000027

Claims (23)

  1. 结合GITR的抗体或其抗原结合片段,其包含An antibody or antigen-binding fragment thereof that binds to GITR, which comprises
    (i)如SEQ ID NO:17或19所示的重链可变区的3个互补决定区HCDR,以及如SEQ ID NO:21或23所示的轻链可变区的3个互补决定区LCDR,或者(i) three complementarity determining region HCDRs of the heavy chain variable region set forth in SEQ ID NO: 17 or 19, and three complementarity determining regions of the light chain variable region set forth in SEQ ID NO: 21 or LCDR, or
    (ii)如SEQ ID NO:18或20所示的重链可变区的3个互补决定区HCDR,以及如SEQ ID NO:22或24所示的轻链可变区的3个互补决定区LCDR。(ii) three complementarity determining region HCDRs of the heavy chain variable region set forth in SEQ ID NO: 18 or 20, and three complementarity determining regions of the light chain variable region set forth in SEQ ID NO: 22 or LCDR.
  2. 结合GITR的抗体或其抗原结合片段,其包含重链可变区的3个互补决定区HCDR,以及轻链可变区的3个互补决定区LCDR,其中An antibody or antigen-binding fragment thereof that binds to GITR, comprising three complementarity determining region HCDRs of the heavy chain variable region, and three complementarity determining regions LCDR of the light chain variable region, wherein
    (i)HCDR1包含SEQ ID NO:1或3或4所示的氨基酸序列,HCDR2包含SEQ ID NO:5所示的氨基酸序列,HCDR3包含SEQ ID NO:7所示的氨基酸序列,LCDR1包含SEQ ID NO:9所示的氨基酸序列,LCDR2包含SEQ ID NO:11、13或14所示的氨基酸序列,且LCDR3包含SEQ ID NO:15所示的氨基酸序列;或者(i) HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 1 or 3 or 4, HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 5, HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 7, and LCDR1 comprises SEQ ID NO: the amino acid sequence shown in 9, LCDR2 comprises the amino acid sequence shown in SEQ ID NO: 11, 13 or 14, and LCDR3 comprises the amino acid sequence shown in SEQ ID NO: 15;
    (ii)HCDR1包含SEQ ID NO:2所示的氨基酸序列,HCDR2包含SEQ ID NO:6所示的氨基酸序列,HCDR3包含SEQ ID NO:8所示的氨基酸序列,LCDR1包含SEQ ID NO:10所示的氨基酸序列,LCDR2包含SEQ ID NO:12所示的氨基酸序列,且LCDR3包含SEQ ID NO:16所示的氨基酸序列。(ii) HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 2, HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 6, HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 8, and LCDR1 comprises SEQ ID NO: The amino acid sequence shown, LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 12, and LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 16.
  3. 结合GITR的抗体或其抗原结合片段,其包含重链可变区和/或轻链可变区,其中An antibody or antigen-binding fragment thereof that binds to GITR, comprising a heavy chain variable region and/or a light chain variable region, wherein
    所述重链可变区包含:The heavy chain variable region comprises:
    (i)表B所列任一抗体的VH中所含的三个互补决定区域(HCDR);(i) three complementarity determining regions (HCDRs) contained in the VH of any of the antibodies listed in Table B;
    (ii)表A所示的HCDR1、HCDR2和HCDR3的组合;(ii) a combination of HCDR1, HCDR2 and HCDR3 shown in Table A;
    (iii)(i)或(ii)的HCDR组合的变体,所述变体在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换);或(iii) a variant of the HCDR combination of (i) or (ii) comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes in said three CDR regions (preferably Amino acid substitution, preferably conservative substitution); or
    (iv)HCDR1、HCDR2和HCDR3,其中HCDR1包含选自SEQ ID NO:1、2、3或4的氨基酸序列,或由所述氨基酸序列组成,或者HCDR1包含与选自SEQ ID NO:1、2、3或4的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;HCDR2包含选自SEQ ID NO:5或6的氨基酸序列,或由所述氨基酸序列组成,或者HCDR2包含与选自SEQ ID NO:5或6的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;HCDR3包含选自SEQ ID NO:7或8的氨基酸序列或由所述氨基酸序列组成,或者HCDR3包含与选自SEQ ID NO:7或8的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;(iv) HCDR1, HCDR2 and HCDR3, wherein HCDR1 comprises or consists of the amino acid sequence selected from SEQ ID NO: 1, 2, 3 or 4, or HCDR1 comprises and is selected from the group consisting of SEQ ID NO: 1, 2 An amino acid sequence having one, two or three alterations (preferably amino acid substitutions, preferably conservative substitutions) compared to the amino acid sequence of 3 or 4; HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 5 or 6, or Amino acid sequence composition, or HCDR2 comprises an amino acid sequence having one, two or three alterations (preferably amino acid substitutions, preferably conservative substitutions) compared to an amino acid sequence selected from SEQ ID NO: 5 or 6; HCDR3 comprises a mutation selected from the group consisting of SEQ ID NO: The amino acid sequence of 7 or 8 consists of or consists of the amino acid sequence, or HCDR3 comprises one, two or three changes (preferably amino acid substitution, preferably conservative) compared to the amino acid sequence selected from SEQ ID NO: 7 or 8. Alternate amino acid sequence;
    和/或and / or
    所述轻链可变区包含:The light chain variable region comprises:
    (i)表B所列任一抗体的VL中所含的三个互补决定区域(LCDR);(i) three complementarity determining regions (LCDR) contained in the VL of any of the antibodies listed in Table B;
    (ii)表A所示的LCDR1、LCDR2和LCDR3的组合;(ii) a combination of LCDR1, LCDR2 and LCDR3 shown in Table A;
    (iii)(i)或(ii)的LCDR组合的变体,所述变体在所述三个CDR区上共包含至少一个且不超过5、4、3、2或1个氨基酸改变(优选氨基酸置换,优选保守置换);或(iii) a variant of the LCDR combination of (i) or (ii), said variant comprising at least one and no more than 5, 4, 3, 2 or 1 amino acid changes on said three CDR regions (preferred Amino acid substitution, preferably conservative substitution); or
    (iv)LCDR1、LCDR2和LCDR3,其中LCDR1包含选自SEQ ID NO:9或10的氨基酸序列或由所述氨基酸序列组成,或者LCDR1包含与选自SEQ ID NO:9或10的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;LCDR2包含选自SEQ ID NO:11、12、13或14的氨基酸序列或由所述氨基酸序列组成,或者LCDR2包含与选自SEQ ID NO:11、12、13或14的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列;LCDR3包含选自SEQ ID NO:15或16的氨基酸序列或由所述氨基酸序列组成,或者LCDR3包含与选自SEQ ID NO:15或16的氨基酸序列相比具有一个、两个或三个改变(优选氨基酸置换,优选保守置换)的氨基酸序列。(iv) LCDR1, LCDR2 and LCDR3, wherein LCDR1 comprises or consists of the amino acid sequence selected from SEQ ID NO: 9 or 10, or LCDR1 comprises an amino acid sequence selected from SEQ ID NO: 9 or 10. An amino acid sequence having one, two or three alterations (preferably amino acid substitutions, preferably conservative substitutions); LCDR2 comprises or consists of an amino acid sequence selected from SEQ ID NO: 11, 12, 13 or 14 or LCDR2 An amino acid sequence comprising one, two or three alterations (preferably amino acid substitutions, preferably conservative substitutions) compared to an amino acid sequence selected from the group consisting of SEQ ID NO: 11, 12, 13 or 14; LCDR3 comprising a selected from the group consisting of SEQ ID NO: The amino acid sequence of 15 or 16 consists of or consists of the amino acid sequence, or LCDR3 comprises one, two or three changes (preferably amino acid substitutions, preferably conservative substitutions) compared to the amino acid sequence selected from SEQ ID NO: 15 or 16. Amino acid sequence.
  4. 权利要求1至3中任一项的抗体或其抗原结合片段,其包含The antibody or antigen-binding fragment thereof according to any one of claims 1 to 3, comprising
    (i)包含与SEQ ID NO:17或19所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的重链可变区,和/或包含与SEQ ID NO:21或23所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的轻链可变区,或者(i) a heavy chain variable region comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 17 or 19, and/or comprising an amino acid represented by SEQ ID NO: 21 or a light chain variable region of an amino acid sequence having at least 90% sequence identity, or
    (ii)包含与SEQ ID NO:18或20所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的重链可变区,和/或包含与SEQ ID NO:22或24所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的轻链可变区,或者(ii) a heavy chain variable region comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 18 or 20, and/or comprising the amino acid set forth in SEQ ID NO: 22 or 24. a light chain variable region of an amino acid sequence having at least 90% sequence identity, or
    (iii)包含SEQ ID NO:17或19所示的氨基酸序列的重链可变区,和/或包含SEQ ID NO:21或23所示的氨基酸序列的轻链可变区,或者(iii) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 17 or 19, and/or a light chain variable region comprising the amino acid sequence of SEQ ID NO: 21 or 23, or
    (iv)包含SEQ ID NO:18或20所示的氨基酸序列的重链可变区,和/或包含SEQ ID NO:22或24所示的氨基酸序列的轻链可变区。(iv) a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 18 or 20, and/or a light chain variable region comprising the amino acid sequence of SEQ ID NO: 22 or 24.
  5. 权利要求1至4中任一项的抗体或其抗原结合片段,其包含The antibody or antigen-binding fragment thereof according to any one of claims 1 to 4, comprising
    (i)包含与SEQ ID NO:25或27所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的重链,和/或包含与SEQ ID NO:29或31所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的轻链,或者(i) a heavy chain comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 25 or 27, and/or comprising at least the amino acid sequence set forth in SEQ ID NO: 29 or 31 a light chain of 90% sequence identity amino acid sequence, or
    (ii)包含与SEQ ID NO:26或28所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的重链,和/或包含与SEQ ID NO:30或32所示的氨基酸序列具有至少90%序列同一性的氨基酸序列的轻链,或者(ii) a heavy chain comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 26 or 28, and/or comprising at least the amino acid sequence set forth in SEQ ID NO: 30 or 32 a light chain of 90% sequence identity amino acid sequence, or
    (iii)包含SEQ ID NO:25或27所示的氨基酸序列的重链,和/或包含SEQ ID NO:29或31所示的氨基酸序列的轻链,或者(iii) a heavy chain comprising the amino acid sequence of SEQ ID NO: 25 or 27, and/or a light chain comprising the amino acid sequence of SEQ ID NO: 29 or 31, or
    (iv)包含SEQ ID NO:26或28所示的氨基酸序列的重链,和/或包含SEQ ID NO:30或32所示的氨基酸序列的轻链。(iv) a heavy chain comprising the amino acid sequence of SEQ ID NO: 26 or 28, and/or a light chain comprising the amino acid sequence of SEQ ID NO: 30 or 32.
  6. 权利要求1至5中任一项的结合GITR的抗体或其抗原结合片段,其具有以下一个或多个特性:The GITR-binding antibody or antigen-binding fragment thereof according to any one of claims 1 to 5, which has one or more of the following characteristics:
    (i)显示与表3所列的任一抗体对GITR相同或相似的结合亲和力和/或特异性;(i) showing the same or similar binding affinity and/or specificity to any of the antibodies listed in Table 3 for GITR;
    (ii)抑制(例如,竞争性抑制)表3所列的任一抗体与GITR的结合;(ii) inhibiting (eg, competitive inhibition) the binding of any of the antibodies listed in Table 3 to GITR;
    (iii)与表3所示的任一抗体结合相同或重叠的表位;(iii) an epitope that binds to the same or overlaps with any of the antibodies shown in Table 3;
    (iv)与表3所示的任一抗体竞争结合GITR;(iv) competing with any of the antibodies shown in Table 3 to bind GITR;
    (v)具有表3所列的任一抗体分子的一个或多个生物学特性。(v) having one or more of the biological properties of any of the antibody molecules listed in Table 3.
  7. 权利要求1至6中任一项的结合GITR的抗体或其抗原结合片段,其具有以下一个或多个特性:The GITR-binding antibody or antigen-binding fragment thereof according to any one of claims 1 to 6, which has one or more of the following characteristics:
    (i)以高亲和力结合GITR,例如人GITR;(i) combining GITR with high affinity, such as human GITR;
    (ii)具有激动剂活性,例如能够有效激活NF-kappaB信号通路;(ii) having agonist activity, for example, capable of efficiently activating the NF-kappaB signaling pathway;
    (iii)能够有效介导ADCC效应;(iii) capable of effectively mediating the ADCC effect;
    (iv)能够激活T细胞,例如CD4T细胞;(iv) being capable of activating T cells, such as CD4 T cells;
    (v)具有更好的抗肿瘤活性,例如能够降低受试者中的肿瘤体积,同时不影响受试者的体重;(v) having better anti-tumor activity, for example, capable of reducing tumor volume in a subject without affecting the weight of the subject;
    (vi)与抗PD-1抗体组合能够更好地抑制肿瘤活性,例如能够降低受试者中的肿瘤体积,同时不影响受试者体重。(vi) Combination with an anti-PD-1 antibody can better inhibit tumor activity, for example, to reduce tumor volume in a subject without affecting the subject's body weight.
  8. 权利要求1至7中任一项的结合GITR的抗体或其抗原结合片段,其中所述抗体是IgG1形式或IgG2形式或IgG4形式的抗体或抗原结合片段。The GITR-binding antibody or antigen-binding fragment thereof according to any one of claims 1 to 7, wherein the antibody is an antibody or antigen-binding fragment in the form of IgG1 or IgG2 or IgG4.
  9. 权利要求1至8中任一项的结合GITR的抗体或其抗原结合片段,其中所述抗体是单克隆抗体。The GITR-binding antibody or antigen-binding fragment thereof according to any one of claims 1 to 8, wherein the antibody is a monoclonal antibody.
  10. 权利要求1至9中任一项的结合GITR的抗体或其抗原结合片段,其中所述抗体是人源化的抗体或人抗体或嵌合抗体。The GITR-binding antibody or antigen-binding fragment thereof according to any one of claims 1 to 9, wherein the antibody is a humanized antibody or a human antibody or a chimeric antibody.
  11. 权利要求1至10中任一项的抗体或其抗原结合片段,其中所述抗原结合片段是选 自以下的抗体片段:Fab、Fab’、Fab’-SH、Fv、单链抗体(例如scFv)或(Fab’) 2、单结构域抗体、双抗体(dAb)或线性抗体。 The antibody or antigen-binding fragment thereof according to any one of claims 1 to 10, wherein the antigen-binding fragment is an antibody fragment selected from the group consisting of Fab, Fab', Fab'-SH, Fv, single-chain antibody (e.g., scFv) Or (Fab') 2 , a single domain antibody, a diabody (dAb) or a linear antibody.
  12. 权利要求1至11中任一项的抗体或其抗原结合片段,其中所述抗体为双特异性或多特异性抗体分子,优选地,双特异性抗体分子与GITR和PD-1、PD-L1或PD-L2结合。The antibody or antigen-binding fragment thereof according to any one of claims 1 to 11, wherein the antibody is a bispecific or multispecific antibody molecule, preferably a bispecific antibody molecule with GITR and PD-1, PD-L1 Or PD-L2 binding.
  13. 分离的核酸,其编码权利要求1至12中任一项的结合GITR的抗体或其任意一条或多条链或其片段。An isolated nucleic acid encoding the GITR-binding antibody of any one of claims 1 to 12, or any one or more of its strands or fragments thereof.
  14. 包含权利要求13的核酸的载体,优选地所述载体是表达载体。A vector comprising the nucleic acid of claim 13, preferably the vector is an expression vector.
  15. 包含权利要求13的核酸或权利要求14的载体的宿主细胞,优选地,所述宿主细胞是原核的或真核的,更优选的选自酵母细胞、哺乳动物细胞(例如293细胞或CHO细胞)或适用于制备抗体或其抗原结合片段的其它细胞。A host cell comprising the nucleic acid of claim 13 or the vector of claim 14, preferably the host cell is prokaryotic or eukaryotic, more preferably selected from the group consisting of a yeast cell, a mammalian cell (e.g., a 293 cell or a CHO cell). Or suitable for the preparation of antibodies or other antigen-binding fragments thereof.
  16. 制备结合GITR的抗体或其抗原结合片段的方法,所述方法包括在适于表达编码权利要求1至12中任一项的结合GITR的抗体或其抗原结合片段的核酸或权利要求13所述的核酸的条件下培养权利要求15的宿主细胞,任选地分离所述抗体或其抗原结合片段,任选地所述方法还包括从所述宿主细胞回收所述结合GITR的抗体或其抗原结合片段。A method of producing an antibody or antigen-binding fragment thereof that binds to GITR, the method comprising a nucleic acid suitable for expression of the antibody or antigen-binding fragment thereof encoding the GITR according to any one of claims 1 to 12, or the method of claim 13. Culturing the host cell of claim 15 under conditions of a nucleic acid, optionally isolating the antibody or antigen-binding fragment thereof, optionally the method further comprising recovering the GITR-binding antibody or antigen-binding fragment thereof from the host cell .
  17. 免疫缀合物,其包含权利要求1至12中任一项的结合GITR的抗体或其抗原结合片段和其它物质,例如细胞毒性剂或标记。An immunoconjugate comprising the GITR-binding antibody or antigen-binding fragment thereof of any one of claims 1 to 12 and other substances, such as a cytotoxic agent or a label.
  18. 药物组合物,其包含权利要求1至12中任一项的结合GITR的抗体或其抗原结合片段或权利要求17的免疫缀合物,以及任选地一种或多种其它治疗剂,例如抗PD-1抗体、抗PD-L1抗体或抗PD-L2抗体,以及任选地药用辅料。A pharmaceutical composition comprising the GITR-binding antibody or antigen-binding fragment thereof of any one of claims 1 to 12 or the immunoconjugate of claim 17, and optionally one or more additional therapeutic agents, such as an anti- PD-1 antibody, anti-PD-L1 antibody or anti-PD-L2 antibody, and optionally a pharmaceutical excipient.
  19. 组合产品,其包含权利要求1至12中任一项的结合GITR的抗体或其抗原结合片段或权利要求17的免疫缀合物,以及一种或多种其它治疗剂,例如化疗剂、细胞毒性剂、疫苗、其它抗体、抗感染活性剂、小分子药物或免疫调节剂,优选地PD-1轴结合拮抗剂,例如抗PD-1抗体、抗PD-L1抗体或抗PD-L2抗体。A combination product comprising the GITR-binding antibody or antigen-binding fragment thereof of any one of claims 1 to 12 or the immunoconjugate of claim 17, and one or more other therapeutic agents, such as a chemotherapeutic agent, cytotoxicity Agents, vaccines, other antibodies, anti-infective agents, small molecule drugs or immunomodulators, preferably PD-1 axis binding antagonists, such as anti-PD-1 antibodies, anti-PD-L1 antibodies or anti-PD-L2 antibodies.
  20. 在受试者中介导ADCC或激活效应T细胞或激活NF-kappaB信号通路或消除调节T细胞(例如Treg细胞)的方法,所述方法包括向所述受试者施用有效量的权利要求1至12中任一项的结合GITR的抗体或其抗原结合片段、或权利要求17的免疫缀合物、或权利要求18的药物组合物,或权利要求19的组合产品。A method of mediating ADCC or activating an effector T cell or activating a NF-kappaB signaling pathway or eliminating a regulatory T cell (eg, a Treg cell) in a subject, the method comprising administering to the subject an effective amount of claim 1 The GITR-binding antibody or antigen-binding fragment thereof, or the immunoconjugate of claim 17, or the pharmaceutical composition of claim 18, or the combination of claim 19.
  21. 预防或治疗受试者肿瘤或感染的方法,所述方法包括向所述受试者施用有效量的权利要求1至12中任一项的结合GITR的抗体或其抗原结合片段、或权利要求17的免疫缀合物、或权利要求18的药物组合物、或权利要求19的组合产品,优选地,所述肿瘤为癌症,例如胃肠道癌症,例如胃癌、直肠癌、结肠癌、结肠直肠癌等;优选地,所述感染为例如细菌感染、病毒感染、真菌感染或原生动物感染,优选地所述感染为慢性感染。A method of preventing or treating a tumor or infection in a subject, the method comprising administering to the subject an effective amount of the GITR-binding antibody or antigen-binding fragment thereof according to any one of claims 1 to 12, or claim 17 The immunoconjugate, or the pharmaceutical composition of claim 18, or the combination of claim 19, preferably, the tumor is a cancer, such as a gastrointestinal cancer, such as gastric cancer, rectal cancer, colon cancer, colorectal cancer Preferably, the infection is, for example, a bacterial infection, a viral infection, a fungal infection or a protozoal infection, preferably the infection is a chronic infection.
  22. 权利要求21所述的方法,其还包括向所述受试者联合施用一种或多种疗法,例如治疗方式和/或其它治疗剂,优选地,治疗方式包括手术治疗和/或放射疗法,和/或其它治疗剂选自化疗剂、细胞毒性剂、疫苗、其它抗体、抗感染活性剂、小分子药物或免疫调节剂,优选地PD-1轴结合拮抗剂,例如抗PD-1抗体或抗PD-L1抗体或抗PD-L2抗体。The method of claim 21, further comprising administering to the subject one or more therapies, such as a treatment modality and/or other therapeutic agent, preferably, the remedy comprising surgery and/or radiation therapy, And/or other therapeutic agents are selected from the group consisting of chemotherapeutic agents, cytotoxic agents, vaccines, other antibodies, anti-infective active agents, small molecule drugs or immunomodulators, preferably PD-1 axis binding antagonists, such as anti-PD-1 antibodies or Anti-PD-L1 antibody or anti-PD-L2 antibody.
  23. 检测样品中GITR的方法,所述方法包括A method of detecting GITR in a sample, the method comprising
    (a)将样品与权利要求1至12中任一项的任何结合GITR的抗体或其抗原结合片段接触;和(a) contacting the sample with any of the GITR-binding antibodies or antigen-binding fragments thereof according to any one of claims 1 to 12;
    (b)检测结合GITR的抗体或其抗原结合片段与GITR间的复合物的形成;任选地,结合GITR的抗体是被可检测地标记的。(b) detecting the formation of a complex between the antibody or antigen-binding fragment thereof that binds to GITR and GITR; optionally, the antibody that binds to the GITR is detectably labeled.
PCT/CN2019/083241 2018-04-20 2019-04-18 Anti-gitr antibody and use thereof WO2019201301A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201980022627.8A CN111918878B (en) 2018-04-20 2019-04-18 anti-GITR antibodies and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810364111.9 2018-04-20
CN201810364111.9A CN110386981A (en) 2018-04-20 2018-04-20 Anti- GITR antibody and application thereof

Publications (1)

Publication Number Publication Date
WO2019201301A1 true WO2019201301A1 (en) 2019-10-24

Family

ID=68239925

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/083241 WO2019201301A1 (en) 2018-04-20 2019-04-18 Anti-gitr antibody and use thereof

Country Status (3)

Country Link
CN (2) CN110386981A (en)
TW (1) TWI743469B (en)
WO (1) WO2019201301A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022150728A1 (en) * 2021-01-08 2022-07-14 Lanier Biotherapeutics Neutralizing antibodies to il-17a, fusion proteins thereof, and uses thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111732658B (en) * 2020-06-28 2022-04-26 英诺湖医药(杭州)有限公司 GITR monoclonal antibodies and medical application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102574924A (en) * 2009-09-03 2012-07-11 先灵公司 Anti-gitr antibodies

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201605896A (en) * 2013-08-30 2016-02-16 安美基股份有限公司 GITR antigen binding proteins
MA41044A (en) * 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102574924A (en) * 2009-09-03 2012-07-11 先灵公司 Anti-gitr antibodies

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022150728A1 (en) * 2021-01-08 2022-07-14 Lanier Biotherapeutics Neutralizing antibodies to il-17a, fusion proteins thereof, and uses thereof

Also Published As

Publication number Publication date
CN111918878B (en) 2022-08-23
TWI743469B (en) 2021-10-21
CN111918878A (en) 2020-11-10
TW201946658A (en) 2019-12-16
CN110386981A (en) 2019-10-29

Similar Documents

Publication Publication Date Title
CN111511765B (en) Anti-galectin-9 antibodies and uses thereof
KR102384845B1 (en) Anti-PD-L1 antibodies and uses thereof
CN110536903B (en) anti-OX 40 antibodies and uses thereof
CA2597717C (en) Antibodies against cxcr4 and methods of use thereof
KR20190105024A (en) Anti-CD47 Antibody and Uses thereof
JP2023538782A (en) CCR8 antibody and its uses
KR101869413B1 (en) S100a4 antibodies and therapeutic uses thereof
CA3101670C (en) Anti-il-23p19 antibody and use thereof
TWI756621B (en) Novel bispecific antibody molecules and bispecific antibodies that simultaneously bind pd-l1 and lag-3
WO2021259199A1 (en) Anti-cd73 antibody and use thereof
TWI793395B (en) Bispecific antibodies that bind to pd-l1 and ox40
WO2022068810A1 (en) Anti-claudin18.2 and cd3 bispecific antibody and use thereof
WO2021254481A1 (en) Anti-claudin18.2 antibody and use thereof
KR20150013935A (en) Antibody against transporter and use thereof
WO2019201301A1 (en) Anti-gitr antibody and use thereof
CN112996815B (en) Human PD-L1 antibodies
CN115087671A (en) anti-IL-27 antibodies and uses thereof
WO2021175191A1 (en) Anti-tim-3 antibody and use thereof
WO2021249542A1 (en) Antibodies binding tnfr2 and uses thereof
CN115368458A (en) anti-CD 73 antibodies and uses thereof

Legal Events

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

Ref document number: 19788393

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19788393

Country of ref document: EP

Kind code of ref document: A1