WO2022203552A1 - Monoclonal antibody that specifically binds to gd2 - Google Patents

Monoclonal antibody that specifically binds to gd2 Download PDF

Info

Publication number
WO2022203552A1
WO2022203552A1 PCT/RU2022/050096 RU2022050096W WO2022203552A1 WO 2022203552 A1 WO2022203552 A1 WO 2022203552A1 RU 2022050096 W RU2022050096 W RU 2022050096W WO 2022203552 A1 WO2022203552 A1 WO 2022203552A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
ser
amino acid
val
acid sequence
Prior art date
Application number
PCT/RU2022/050096
Other languages
English (en)
French (fr)
Inventor
Sergei Andreevich AGEEV
Yulia Sergeevna CHERNYKH
Diana Aleksandrovna KONDINSKAIA
Valeriia Evgenevna SHIGINA
Dina Khaidarovna SAKHAROVA
Mariia Anatolevna GREFENSHTEIN
Alina Konstantinovna STOLYAROVA
Valery Vladimirovich SOLOVYEV
Pavel Andreevich IAKOVLEV
Dmitry Valentinovich MOROZOV
Original Assignee
Joint Stock Company "Biocad"
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
Priority claimed from RU2021107773A external-priority patent/RU2796937C2/ru
Application filed by Joint Stock Company "Biocad" filed Critical Joint Stock Company "Biocad"
Priority to CR20230456A priority Critical patent/CR20230456A/es
Priority to CN202280024459.8A priority patent/CN117677695A/zh
Publication of WO2022203552A1 publication Critical patent/WO2022203552A1/en
Priority to CONC2023/0012549A priority patent/CO2023012549A2/es

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • C07K16/3084Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties against tumour-associated gangliosides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to the field of biotechnology and medicine, in particular to a monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 (ganglioside GD2).
  • the invention further relates to nucleic acids encoding said antibody, expression vectors, host cells and methods for producing same, methods for producing the antibodies according to the invention, pharmaceutical compositions comprising the antibody according to the invention, pharmaceutical compositions comprising the antibody according to the invention and other therapeutically active compounds, methods for treating diseases or disorders mediated by GD2, uses of the antibodies or pharmaceutical compositions thereof for treating diseases or disorders mediated by GD2, and uses of the antibodies and other therapeutically active compounds for treating diseases or disorders mediated by GD2.
  • GD2 is the first ganglioside proven to be an effective target antigen for cancer immunotherapy.
  • Gangliosides are composed of glycosphingolipids and sialic acids (N-acetylneuraminic acid, Neu5Ac or NANA), which are nine-carbon monosaccharides. Ganglioside nomenclature is based on the number and position of the NANA residues. Monosaccharides are first added to ceramide to form lactosylceramide, and then NANA residues are added to form gangliosides. Each sugar is bound by specific glycosyltransferases.
  • GD2 has two NANA (a-2,8 sialic acid and a-2,3 sialic acid), and is derived from precursor GD3 by adding Gal-NAc through the enzyme GM2/GD2 synthase (bl,4-N-acetylgalactosaminyltransferase).
  • GM2/GD2 synthase bl,4-N-acetylgalactosaminyltransferase
  • the end-terminal penta-oligosaccharide constitutes the specific epitope of GD2 to which the most specific antibodies are directed.
  • This critical enzyme GM2/GD2 synthase responsible for making GD2 has been successfully exploited as a molecular marker of minimal residual neuroblastoma in the bone marrow, with major prognostic impact on patient survival.
  • GD3 and GDlb are the most common cross-reactive gangliosides recognized by anti-GD2 antibodies.
  • a GD2-derivative with a 9-O-acetyl modification on the terminal sialic acid is called O-acetyl -GD2. While most anti-GD2 antibodies cross-react with 0-GD2, some do not. Anti-0-GD2 antibodies with no cross-reactivity with GD2 had less cross-reactivity with normal neurons.
  • Gangliosides are found on the cell surface of the nervous system in vertebrates.
  • GD2 is expressed on neural stem cells, mesenchymal stem cells (MSCs) and peripheral sympathoadrenergic progenitors, and it is involved in neural differentiation and proliferation. While the role of polysialic acid in neuronal development has been extensively studied, the precise functions of gangliosides, and specifically of GD2, remain unknown. After birth, GD2 expression is restricted to the CNS, predominantly in neuronal cell bodies, and MSCs, as well as peripheral nerves and skin melanocytes at low levels. GD2 is thought to play a role in the maintenance and repair of nervous tissues, which undergo continually progressive degenerative changes through the regulation of complement activation and subsequent inflammation, although the exact immunologic mechanism remains obscure.
  • MSCs mesenchymal stem cells
  • peripheral sympathoadrenergic progenitors peripheral sympathoadrenergic progenitors
  • GD2 (+) MSCs have the potentials to differentiate into multiple clones, including neurons (MAYA SUZUKI ET AL., Disialoganglioside GD2 as a therapeutic target for human diseases, Expert Opinion on Therapeutic Targets, 2015, v. 15, pages 349-362, PMID: 25604432, DOI: 10.1517/14728222.2014.986459).
  • GD2 is hyperexpressed in a variety of embryonal cancers (neuroblastoma, brain tumors, retinoblastoma, Ewing’s sarcoma, rhabdomyosarcoma), bone tumors (osteosarcoma, Ewing’s sarcoma), soft tissue sarcomas (leiomyosarcoma, liposarcoma, fibrosarcoma), lung cancer, melanoma, and breast cancer.
  • Tumor monoclonal antibodies have demonstrated clinical efficacy, thus becoming an important method for cancer immunotherapy. Due to its limited expression in normal tissue, the disialogangloside GD2 expressed on neuroblastoma cells is an excellent candidate for mAh therapy.
  • the present invention relates to an isolated monoclonal antibody or antigenbinding fragment thereof that specifically binds to GD2 (ganglioside GD2), wherein the antibody or antigen -binding fragment thereof includes:
  • CDR1 with an amino acid sequence selected from the group: GHNMN (SEQ ID NO: 1) or GKNMN (SEQ ID NO: 2),
  • CDR3 with an amino acid sequence selected from the group: GMIY (SEQ ID NO: 4), GMFY (SEQ ID NO: 5), GMYY (SEQ ID NO: 6) or GMLY (SEQ ID NO: 7); and
  • CDR1 with an amino acid sequence selected from the group: RSSRSLVHRNGNTYLH (SEQ ID NO: 8) or RSSQNLVHRNGNTYLH (SEQ ID NO: 9),
  • CDR2 with an amino acid sequence selected from the group: KVSNRFG (SEQ ID NO: 10) or KVNNRFS (SEQ ID NO: 11),
  • CDR3 with an amino acid sequence selected from the group: GQSTHVPPLT (SEQ ID NO: 12) or SQSTHVPPLS (SEQ ID NO: 13).
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises:
  • RVTLTVDKSISTAYMELSRLRSDDTAVYYCVS (SEQ ID NO: 44)
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises:
  • FR2 with an amino acid sequence selected from the group: WYLQKPGQSPKLLIH (SEQ ID NO: 47) or W YLQKPGQ SPQLLIH (SEQ ID NO: 48),
  • GVPDRFSGSGSGTDFTLKISRVEAEDVGVYFC SEQ ID NO: 49
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes: a) a heavy chain variable domain that comprises:
  • RVTLTVDKSISTAYMELSRLRSDDTAVYYCVS (SEQ ID NO: 44)
  • FR2 with an amino acid sequence selected from the group: WYLQKPGQSPKLLIH (SEQ ID NO: 47) or WYLQKPGQ SPQLLIH (SEQ ID NO: 48),
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises an amino acid sequence that has at least 98 % identity to the amino acid sequence of SEQ ID NO: 17.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16 or SEQ ID NO: 17.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises an amino acid sequence that has at least 96 % identity to the amino acid sequence of SEQ ID NO: 21.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20 or SEQ ID NO: 21.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes:
  • a heavy chain variable domain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16 or SEQ ID NO: 17;
  • a light chain variable domain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20 or SEQ ID NO: 21.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes: (a) a heavy chain variable domain that comprises the amino acid sequence of SEQ ID NO:
  • the isolated monoclonal antibody that specifically binds to GD2 is a full-length IgG antibody.
  • the isolated monoclonal antibody is a full-length IgG antibody that is of human IgGl, IgG2, IgG3 or IgG4 isotype.
  • the isolated monoclonal antibody is a full-length IgG antibody that is of human IgGl isotype.
  • the isolated monoclonal antibody comprises YTE mutations (M252Y, S254T, T256E) and/or K322A in the Fc fragment as compared to the naturally-occurring sequence of the Fc fragment.
  • the isolated monoclonal antibody includes a heavy chain comprising an amino acid sequence that is selected from the group: SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36 or SEQ ID NO: 37.
  • the isolated monoclonal antibody includes a light chain comprising an amino acid sequence that is selected from the group: SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40 or SEQ ID NO: 41.
  • the isolated monoclonal antibody includes:
  • a heavy chain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36 or SEQ ID NO: 37, and
  • the isolated monoclonal antibody includes:
  • the isolated monoclonal antibody includes:
  • the present invention relates to an isolated nucleic acid that encodes any above antibody or antigen-binding fragment thereof.
  • the isolated nucleic acid is DNA.
  • the present invention relates to an expression vector that comprises any of the above nucleic acids.
  • the present invention relates to a method for producing a host cell to produce any above antibody or antigen-binding fragment thereof, and comprises transformation of the cell with the above vector.
  • the present invention relates to a host cell for producing any above antibody or antigen-binding fragment thereof, the host cell comprising any of the above nucleic acids.
  • the present invention relates to a method for producing any above antibody or antigen-binding fragment thereof, which comprises culturing said host cell in a growth medium under conditions sufficient to produce said antibody, if necessary, followed by isolation and purification of the resulting antibody.
  • the present invention relates to a pharmaceutical composition used for treating a disease or disorder mediated by GD2, which comprises any above antibody or antigenbinding fragment thereof in a therapeutically effective amount in combination with one or more pharmaceutically acceptable excipients.
  • the disease or disorder mediated by GD2 is selected from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • the present invention relates to a pharmaceutical composition for treating a disease or disorder mediated by GD2, the pharmaceutical combination comprising any above antibody or antigen-binding fragment thereof and at least one other therapeutically active compound.
  • the disease or disorder mediated by GD2 is selected from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • the other therapeutically active compound is an antibody, chemotherapeutic agent, or hormone therapy agent. In some embodiments of the pharmaceutical composition, the other therapeutically active compound is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is selected from a PD-1 inhibitor, PD-L1 inhibitor, or CTLA-4 inhibitor.
  • the PD-1 inhibitor is an antibody that specifically binds to PD-1.
  • the antibody that specifically binds to PD-1 is selected from the group: prolgolimab, pembrolizumab, nivolumab.
  • the CTLA-4 inhibitor is an antibody that specifically binds to CTLA-4.
  • the antibody that specifically binds to CTLA-4 is ipilimumab or nurulimab.
  • the PD-L1 inhibitor is an antibody that specifically binds to PD-L1.
  • the antibody that specifically binds to PD-L1 is selected from the group: durvalumab, avelumab, atezolizumab, manelimab.
  • the other therapeutically active compound is selected from the group: IL-2, GM-CSF, isotretinoin, one or more other cytokines, or any combination of therapeutically active compounds from this group.
  • the present invention relates to a method for inhibiting the biological activity of GD2 in a subject in need of such inhibition, comprising administering an effective amount of any above antibody or antigen-binding fragment thereof.
  • the present invention relates to a method for treatment of a disease or disorder mediated by GD2, which comprises administering in a subject in need of such treatment any above antibody or antigen-binding fragment thereof or said pharmaceutical composition, in a therapeutically effective amount.
  • the present invention relates to a method for treating a disease or disorder mediated by GD2, comprising administering in a subject in need of such treatment any above antibody or antigen-binding fragment thereof, and selected from the group: a) administration of at least one other therapeutically active compound, b) radiotherapy, c) hematopoietic stem cell transplantation, d) surgical treatment and, if necessary, adjuvant therapy, or e) any combination of the above a) to d).
  • the disease or disorder mediated by GD2 is selected from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • other therapeutically active compound is an antibody, chemotherapeutic agent, or hormone therapy agent.
  • the other therapeutically active compound is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is selected from a PD-1 inhibitor, PD-L1 inhibitor, or CTLA-4 inhibitor.
  • the PD-1 inhibitor is an antibody that specifically binds to PD-1.
  • the antibody that specifically binds to PD-1 is selected from the group comprising prolgolimab, pembrolizumab, nivolumab.
  • the CTLA-4 inhibitor is an antibody that specifically binds to CTLA-4.
  • the antibody that specifically binds to CTLA-4 is ipilimumab or nurulimab.
  • the PD-L1 inhibitor is an antibody that specifically binds to PD-L1.
  • the antibody that specifically binds to PD-L1 is selected from the group: durvalumab, avelumab, atezolizumab, manelimab.
  • the other therapeutically active compound is selected from the group: IL-2, GM-CSF, isotretinoin, one or more other cytokines, or any combination of therapeutically active compounds from this group.
  • the present invention relates to the use of the above antibody or antigenbinding fragment thereof or the above pharmaceutical composition for treating in a subject in need of such treatment a disease or disorder mediated by GD2.
  • the present invention relates to the use of any of the above antibody or antigen-binding fragment thereof and at least one of the group: a) other therapeutically active compound, b) radiotherapy, c) hematopoietic stem cell transplantation or d) surgical treatment and, if necessary, adjuvant therapy, for treating a disease or disorder mediated by GD2.
  • the disease or disorder mediated by GD2 is selected from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • the other therapeutically active compound is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is selected from a PD- 1 inhibitor, PD-L1 inhibitor, or CTLA-4 inhibitor.
  • the PD-1 inhibitor is an antibody that specifically binds to PD-1.
  • the antibody that specifically binds to PD-1 is selected from the group: prolgolimab, pembrolizumab, nivolumab.
  • the CTLA-4 inhibitor is an antibody that specifically binds to CTLA-4.
  • the antibody that specifically binds to CTLA-4 is ipilimumab or nurulimab.
  • the PD-L1 inhibitor is an antibody that specifically binds to PD-L1.
  • the antibody that specifically binds to PD-L1 is selected from the group: durvalumab, avelumab, atezolizumab, manelimab.
  • the other therapeutically active compound is selected from the group: IL-2, GM-CSF, isotretinoin, one or more other cytokines, or any combination of therapeutically active compounds from this group.
  • Figure 1 is the structure of the Fab fragment in complex with the GD2 target.
  • Figure 2 is a map of vector bearing the heavy chain genetic sequence of anti-GD2 antibody.
  • Figure 3 is a map of vector bearing the light chain genetic sequence of anti-GD2 antibody.
  • Figure 4 is an electrophoregram of 10-008 candidates under reducing and non-reducing conditions, by gradient gel 4-20% SDS-PAGE.
  • Figure 5 is a graph showing the presence of antibody-dependent cellular cytotoxicity of antibody 10-008 in an assay using SK-N-BE(2) target cells. Points free of antibody were used as a negative control.
  • Figure 6 is a graph showing the complement-dependent cytotoxicity of antibodies to GD2 according to the invention in an assay using SK-N-BE(2) target cells.
  • the graph shows the fluorescence level of vital dye (used to show the number of living cells) vs. the concentration of antibodies upon addition of human serum. EC so values were obtained in the SigmaPlot 14.0 software, based on the logistic model.
  • Ka as used herein is intended to refer to the association rate of a particular antibody-antigen interaction
  • KD or “Kd” is intended to refer to the dissociation rate of a particular antibody-antigen interaction.
  • Binding affinity generally refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g. an antibody) and its binding partner (e.g. an antigen). Unless indicated otherwise, "binding affinity” refers to intrinsic (characteristic, true) binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g. antibody and antigen).
  • the affinity of a molecule X for its binding partner Y can generally be represented by the dissociation constant (Kd).
  • the preferred Kd value is about 200 nM, 150 nM, 100 nM, 60 nM, 50 nM, 40 nM, 30 nM, 20 nM, 10 nM, 8 nM, 6 nM, 4 nM, 2 nM, 1 nM, or less.
  • Affinity can be measured by common methods known in the art, including those described in the present description. Low-affinity antibodies generally bind an antigen slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind an antigen faster and tend to remain bound longer. A variety of methods of measuring binding affinity are known in the art, any of which can be used for the purposes of the present invention.
  • koff ' or "kdis” refers to the off rate constant of a particular interaction between a binding molecule and antigen.
  • the off rate constant koff can be measured using bio-layer interferometry, for example, using OctetTM system.
  • off-rate screening refers to screening in which candidates are examined only based on koff values.
  • R 2 refers to the coefficient of determination.
  • Response refers to the antibody-antigen binding signal.
  • in vitro refers to a biological entity, a biological process, or a biological reaction outside the body under artificial conditions.
  • a cell grown in vitro is to be understood as a cell grown in an environment outside the body, e.g. in a test tube, a culture vial, or a microtiter plate.
  • IC50 inhibitor concentration 50%
  • concentrations of a formulation at which a measurable activity or response, for example, growth/proliferation of cells such as tumor cells, is inhibited by 50%.
  • IC50 value can be calculated using appropriate dose-response curves, using special statistical software for curve fitting.
  • ED50 EC50 (50% effective dose/concentration) refers to concentrations of a formulation producing 50% biological effect (which may include cytoxicity).
  • the present invention relates to a monoclonal antibody or or antigen-binding fragment thereof that specifically binds to GD2 (ganglioside GD2).
  • mAb refers to an antibody that is synthesized and isolated by a separate clonal population of cells.
  • the antibody of the invention is a recombinant antibody.
  • the term "recombinant antibody” refers to an antibody that is expressed in a cell or cell line comprising nucleotide sequence(s) encoding antibodies, wherein said nucleotide sequence(s) is (are) not associated with the cell in nature.
  • the present invention relates to an isolated monoclonal antibody or antigenbinding fragment thereof that specifically binds to GD2 (ganglioside GD2), wherein the antibody or antigen -binding fragment thereof includes:
  • CDR1 with an amino acid sequence selected from the group: GHNMN (SEQ ID NO: 1) or GKNMN (SEQ ID NO: 2),
  • CDR3 with an amino acid sequence selected from the group: GMIY (SEQ ID NO: 4), GMFY (SEQ ID NO: 5), GMYY (SEQ ID NO: 6) or GMLY (SEQ ID NO: 7); and
  • CDR1 with an amino acid sequence selected from the group: RSSRSLVHRNGNTYLH (SEQ ID NO: 8) or RSSQNLVHRNGNTYLH (SEQ ID NO: 9),
  • CDR2 with an amino acid sequence selected from the group: KVSNRFG (SEQ ID NO: 10) or KVNNRFS (SEQ ID NO: 11),
  • CDR3 with an amino acid sequence selected from the group: GQSTHVPPLT (SEQ ID NO: 12) or SQSTHVPPLS (SEQ ID NO: 13).
  • isolated used to describe various antibodies in this description refers to an antibody which has been identified and separated and/or regenerated from a cell or cell culture, in which the antibody is expressed.
  • Impurities contaminant components
  • the isolated polypeptide is typically prepared by at least one purification step.
  • Amplification of the GD2 gene and/or overexpression of protein thereof have been observed in many cancers, for example, in any of the diseases from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • antibody or “immunoglobulin” (Ig), as used in the present description, includes whole antibodies.
  • the term “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds, or an antigen-binding portion. Each heavy chain comprises a heavy chain variable region (abbreviated referred to in the present description as VH) and a heavy chain constant region.
  • VH heavy chain variable region
  • the type of a heavy chain present defines the class of an antibody; these chains are found in IgA, IgD, IgE, IgG, and IgM antibodies, respectively.
  • Distinct heavy chains differ in size and composition; a and g contain approximately 450 amino acids, while m and e have approximately 550 amino acids.
  • the constant region is identical in all antibodies of the same isotype, but differs in antibodies of different isotypes.
  • Heavy chains g, a and d have a constant region composed of three constant domains CHI, CH2 and CH3 (in a line), and a hinge region for added flexibility (Woof J., Burton D., Nat Rev Immunol 4, 2004, cc.89-99); heavy chains m and e have a constant region composed of four constant domains CHI, CH2, CH3 and CH4 (Janeway C.A., Jr. et al, Immunobiology, 5th ed., publ. by Garland Publishing, 2001). In mammals, known are only two types of light chains denoted by lambda (l) and kappa (K).
  • Each light chain consists of a light chain variable region (abbreviated referred to in the present description as VL) and light chain constant region.
  • the approximate length of a light chain is 211 to 217 amino acids.
  • the light chain is a kappa (K) light chain
  • the constant domain CL is preferably C kappa (K).
  • Antibodies can be of any class (e.g., IgA, IgD, IgE, IgG, and IgM, preferably IgG), or subclass (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2, preferably IgGl).
  • class e.g., IgA, IgD, IgE, IgG, and IgM, preferably IgG
  • subclass e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2, preferably IgGl.
  • VL and VH regions can be further subdivided into hyper-variability regions called complementarity determining regions (CDRs), interspersed between regions that are more conserved, termed framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy -terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of antibodies may mediate the binding of immunoglobulin to host tissues or factors, including various cells of the immune system (e.g. effector cells) and the first component (Clq) of the classical complement system.
  • antigen-binding portion of an antibody or "antigen -binding fragment” refers to one or more fragments of an antibody that retain the capability of specific binding to an antigen. It has been shown that the antigen-binding function of antibody can be performed by fragments of a full- length antibody.
  • binding fragments which are included within the term "antigenbinding portion" of an antibody include (i) Fab-fragment, monovalent fragment, consisting of VL, VH, CL and CHI domains; (ii) F(ab')2 fragment, a bivalent fragment comprising two Fab- fragments linked by a disulfide bridge at the hinge region; (iii) Fd-fragment consisting of VH and CHI domains; (iv) Fv-fragment consisting of VL and VH domains of a single arm of an antibody; (v) dAb-fragment (Ward et al., (1989) Nature 341 :544-546), which consists of a VH/VHH domain.
  • VL and VH two regions of the Fv-fragment, VL and VH, are encoded by different genes, they can be joined using recombinant methods using a synthetic linker that enables to receive them as a single protein chain in which the VL and VH regions are paired to form monovalent molecules (known as a single-chain Fv (scFv); see e.g. Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). It is assumed that such single-stranded molecules are also included within the term "antigen-binding portion" of antibody. Such antibody fragments are produced using conventional techniques known to those skilled in the art, and these fragments are screened in the same manner as intact antibodies are.
  • variable domain refers to the fact that certain portions of the variable domains greatly differ in sequence among antibodies.
  • the V domain mediates antigen binding and determines specificity of each particular antibody for its particular antigen.
  • variability is not evenly distributed across the 110-amino acid span of the variable domains.
  • the V regions consist of invariant fragments termed framework regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability termed “hypervariable regions” or CDRs.
  • FRs framework regions
  • hypervariable regions or CDRs.
  • the variable domains of native heavy and light chains each comprise four FRs, largely adopting a beta-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the beta-sheet structure.
  • the hypervariable regions in each chain are held together in close proximity by FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Rabat et al., Sequences of Proteins of Immunological Interest. 5 th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)).
  • the constant domains are not involved directly in binding of antibody to antigen, but exhibit various effector functions, such as participation of antibody in antibody-dependent cellular cytotoxicity (ADCC).
  • hypervariable region refers to the amino acid residues of antibody which are responsible for antigen binding.
  • the hypervariable region typically comprises amino acid residues from a “complementarity determining region” or "CDR" and/or those residues from a “hypervariable loop”.
  • Rabat numbering scheme or “numbering according to Rabat” as used in this application refers to the system for numbering of amino acid residues that are more variable (i.e. hypervariable) than other amino acid residues in variable regions of heavy and light chains of the antibody (Rabat et al. Ann. N.Y. Acad. Sci., 190:382-93 (1971); Rabat et al. Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242 (1991)).
  • the antibody of the present invention "which binds" a target antigen refers to an antibody that binds the antigen with sufficient affinity such that the antibody can be used as a diagnostic and/or therapeutic agent targeting a protein or cell or tissue expressing the antigen, and slightly cross-reacts with other proteins.
  • analytical methods fluorescence-activated cell sorting (FACS), radioimmunoassay (RIA) or ELISA, in such embodiments, the degree of antibody binding to a non-target protein is less than 10 % of antibody binding to a specific target protein.
  • the term “specific binding” or “specifically binds to” or “is specific for” a particular polypeptide or an epitope on a particular target polypeptide means binding that is significantly (measurably) different from a non-specific interaction.
  • Specific binding may be measured, for example, by determining binding of a molecule as compared to binding of a control molecule. For example, specific binding may be determined by competition with another molecule that is similar to the target, for example, an excess of non- labeled target. In this case, specific binding is indicated if the binding of the labeled target to a probe is competitively inhibited by the excess of unlabeled target.
  • the term “specific binding” or phrases “specifically binds to” or " is specific for” a particular polypeptide or an epitope on a particular target polypeptide may be described by example of a molecule having a Kd for the target of at least about 200 nM, or at least about 150 nM, or at least about 100 nM, or at least about 60 nM, or at least about 50 nM, or at least about 40 nM, or at least about 30 nM, or at least about 20 nM, or at least about 10 nM, or at least about 8 nM, or at least about 6 nM, or at least about 4 nM, or at least about 2 nM, or at least about 1 nM, or greater.
  • the term “specific binding” refers to binding where a molecule binds to a particular polypeptide or epitope on a particular polypeptide without substantially binding to any other polypeptide or epitope on a polypeptide.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises:
  • RVTLTVDKSISTAYMELSRLRSDDTAVYYCVS (SEQ ID NO: 44)
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises: (i) FR1 with the amino acid sequence DIVMTQTPLSLSVTPGERASLSC (SEQ ID NO:
  • FR2 with an amino acid sequence selected from the group: WYLQKPGQSPKLLIH (SEQ ID NO: 47) or WYLQKPGQ SPQLLIH (SEQ ID NO: 48),
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes: a) a heavy chain variable domain that comprises:
  • RVTLTVDKSISTAYMELSRLRSDDTAVYYCVS (SEQ ID NO: 44)
  • FR2 with an amino acid sequence selected from the group: WYLQKPGQSPKLLIH (SEQ ID NO: 47) or WYLQKPGQ SPQLLIH (SEQ ID NO: 48),
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises an amino acid sequence that has at least 98 % identity to the amino acid sequence of SEQ ID NO: 17.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a heavy chain variable domain that comprises an amino acid sequence that is selected from the group:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises an amino acid sequence that has at least 96 % identity to the amino acid sequence of SEQ ID NO: 21.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes a light chain variable domain that comprises an amino acid sequence that is selected from the group: DIVMTQTPLSLSVTPGERASLSCRSSRSLVHRNGNTYLHWYLQKPGQSPKLLIHK
  • V SNRF GGVPDRFSGSGSGTDFTLKISRVEAED VGVYF CGQSTHVPPLTF GQGTKLELK (SEQ ID NO: 18),
  • V SNRF GGVPDRFSGSGSGTDFTLKISRVEAED VGVYF CGQSTHVPPLTF GQGTKLELK (SEQ ID NO: 19)
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes:
  • a heavy chain variable domain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16 or SEQ ID NO: 17;
  • a light chain variable domain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO: 20 or SEQ ID NO: 21.
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes:
  • the isolated monoclonal antibody or antigenbinding fragment thereof includes:
  • the isolated monoclonal antibody that specifically binds to GD2 is a full-length IgG antibody.
  • the isolated monoclonal antibody is a full-length IgG antibody that is of human IgGl, IgG2, IgG3 or IgG4 isotype.
  • the isolated monoclonal antibody is a full-length IgG antibody that is of human IgGl isotype.
  • the isolated monoclonal antibody comprises YTE mutations (M252Y, S254T, T256E) and/or K322A in the Fc fragment as compared to the naturally-occurring sequence of the Fc fragment.
  • the above mutations in the Fc fragment are numbered according to EU numbering for amino acid chains of antibodies(Edelman, G.M., et al., Proc. Natl. Acad. Sci. USA 63 (1969), pp. 78-85; Kabat, E.A., et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD, (1991).
  • the isolated monoclonal antibody includes a heavy chain comprising an amino acid sequence that is selected from the group:
  • the isolated monoclonal antibody includes a light chain comprising an amino acid sequence that is selected from the group:
  • the isolated monoclonal antibody includes:
  • a heavy chain that comprises an amino acid sequence that is selected from the group: SEQ ID NO: 22, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 31, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 36 or SEQ ID NO: 37, and
  • the isolated monoclonal antibody includes:
  • the isolated monoclonal antibody includes:
  • the isolated monoclonal antibody that specifically binds to GD2 is antibody 07-006.
  • the antibody 07-006 includes:
  • the antibody 07-006 includes:
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO: 14;
  • the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 18.
  • the antibody 07-006 includes:
  • the antibody 07-006 includes:
  • the antibody 07-006 includes:
  • the isolated monoclonal antibody that specifically binds to GD2 is antibody 07-015.
  • the antibody 07-015 includes:
  • the antibody 07-015 includes:
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO:
  • the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 19.
  • the antibody 07-015 includes:
  • the antibody 07-015 includes:
  • the antibody 07-015 includes:
  • the isolated monoclonal antibody that specifically binds to GD2 is antibody 07-016.
  • the antibody 07-016 includes:
  • the antibody 07-016 includes:
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO:
  • the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 18.
  • the antibody 07-016 includes:
  • the antibody 07-016 includes:
  • the antibody 07-016 includes:
  • the isolated monoclonal antibody that specifically binds to GD2 is antibody 07-028.
  • the antibody 07-028 includes:
  • the antibody 07-028 includes:
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO:
  • the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 20.
  • the antibody 07-028 includes:
  • the antibody 07-028 includes:
  • the antibody 07-028 includes:
  • a heavy chain variable domain comprising: (i) CDR1 (IMGT) with the amino acid sequence of SEQ ID NO: 65,
  • the isolated monoclonal antibody that specifically binds to GD2 is antibody 07-031.
  • the antibody 07-031 includes:
  • the antibody 07-031 includes:
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO:
  • the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 19.
  • the antibody 07-031 includes:
  • the antibody 07-031 includes:
  • the antibody 07-031 includes: (a) a heavy chain variable domain comprising:
  • the isolated monoclonal antibody that specifically binds to GD2 is antibody 07-041.
  • the antibody 07-041 includes:
  • the antibody 07-041 includes:
  • the heavy chain variable domain comprises the amino acid sequence of SEQ ID NO:
  • the light chain variable domain comprises the amino acid sequence of SEQ ID NO: 21.
  • the antibody 07-041 includes:
  • the antibody 07-041 includes:
  • the antibody 07-041 includes:
  • Modification(s) of amino acid sequences of antibodies is (are) provided. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of antibody are prepared by introducing appropriate nucleotide changes into the nucleic acid encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions, and/or insertions and/or substitutions of residues within the amino acid sequences of antibody. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • the amino acid changes also may alter post- translational processes in the antibody, such as changing the number or position of glycosylation sites.
  • Variant of modification of amino acid sequences of antibodies using amino acid substitutions is substitution of at least one amino acid residue in the antibody molecule with a different residue.
  • the sites of greatest interest for substitutional mutagenesis include hypervariable regions or CDRs, but FR or Fc alterations are also contemplated.
  • Conservative substitutions are shown in Table A under the heading "preferred substitutions”. If such substitutions cause alteration of the biological activity, further substantial changes can be made, which are denoted as "exemplary substitutions" set forth in Table A, or alterations described in more detail below when describing amino acid classes, and also product screening may be performed.
  • the antibodies 07-006, 07-015, 07-016, 07-028, 07- 031, 07-041 include an Fc fragment that comprises YTE mutations (M252Y, S254T, T256E) and/or K322A as compared to the naturally-occurring sequence of the Fc fragment.
  • the antibodies 07-006, 07-015, 07-016, 07-028, 07- 031, 07-041 include an Fc fragment that comprises YTE mutations (M252Y, S254T, T256E) and K322A as compared to the naturally-occurring sequence of the Fc fragment.
  • the antibodies 07-006, 07-015, 07-016, 07-028, 07- 031, 07-041 include an Fc fragment that comprises YTE mutations (M252Y, S254T, T256E) as compared to the naturally-occurring sequence of the Fc fragment.
  • the antibodies 07-006, 07-015, 07-016, 07-028, 07- 031, 07-041 include an Fc fragment that comprises the K322A mutation as compared to the naturally-occurring sequence of the Fc fragment.
  • the antibody 07-006 with YTE and K322A mutations includes:
  • the antibody 07-015 with YTE and K322A mutations includes:
  • the antibody 07-016 with YTE and K322A mutations includes:
  • the antibody 07-028 with YTE and K322A mutations includes:
  • the antibody 07-031 with YTE and K322A mutations includes:
  • the antibody 07-041 with YTE and K322A mutations includes:
  • the antibody 07-006 with YTE mutations includes:
  • the antibody 07-015 with YTE mutations includes:
  • the antibody 07-016 with YTE mutations includes:
  • the antibody 07-028 with YTE mutations includes:
  • the antibody 07-031 with YTE mutations includes:
  • the antibody 07-041 with YTE mutations includes:
  • the antibody 07-006 with the K322A mutation includes:
  • the antibody 07-015 with the K322A mutation includes:
  • the antibody 07-016 with the K322A mutation includes:
  • the antibody 07-028 with the K322A mutation includes:
  • the antibody 07-031 with the K322A mutation includes:
  • the antibody 07-041 with the K322A mutation includes:
  • the antibodies according to the invention may be afucosylated antibodies.
  • the antibodies according to the invention may be fucosylated antibodies.
  • the presence or absence of antibody fucosylation will depend on the cell culture that is used to produce antibodies according to the invention.
  • antibody fragments rather than whole antibodies.
  • the small sizes of the fragments contributes to rapid clearance thereof and may contribute to better penetration into dense tumors.
  • F(ab')2 fragments can be isolated directly from recombinant host cell culture Fab and F(ab')2 fragment with increased in vivo half-life retaining epitope binding receptor residues are described in US 5869046. Other techniques for the production of antibody fragments will be apparent to those skilled in the art.
  • the antibody of choice is a single chain Fv fragment (scFv) (see WO 93/16185; US 5571894 and US 5587458).
  • Fv and scFv are the only species with intact binding sites that are devoid of constant regions; as a result, they are suitable for reduced nonspecific binding during in vivo use.
  • scFv fusion proteins may be constructed to yield fusion of an effector protein at either N- or C-terminus of an scFv (see Antibody Engineering, ed. Borrebaeck, above).
  • the antibody fragment may also be a "linear antibody", e.g. as described in U.S. 5641870.
  • the present invention relates to an isolated nucleic acid that encodes any above antibody or antigen-binding fragment thereof that specifically binds to GD2.
  • the nucleic acid molecules may be isolated.
  • nucleic acid means a precise sequence of nucleotides, modified or not, determining a fragment or a region of a nucleic acid, containing unnatural nucleotides or not, and being either a double-strand DNA or RNA, a single-strand DNA or RNA, or transcription products of said DNAs.
  • the present invention does not relate to nucleotide sequences in their natural chromosomal environment, i.e. in a natural state.
  • the sequences of the present invention have been isolated and/or purified, i.e., they were sampled directly or indirectly, for example by copying, their environment having been at least partially modified.
  • isolated nucleic acids obtained by recombinant genetics, by means, for example, of host cells, or obtained by chemical synthesis should also be mentioned here.
  • a reference to a nucleotide sequence encompasses the complement thereof unless otherwise specified.
  • a reference to a nucleic acid having a particular sequence should be understood as one which encompasses the complementary strand thereof with the complementary sequence thereof.
  • An "isolated" nucleic acid molecule is one which is identified and separated from at least one nucleic acid molecule-impurity, which the former is bound to in the natural source of antibody nucleic acid.
  • An isolated nucleic acid molecule is different from the form or set in which it is found under natural conditions.
  • an isolated nucleic acid molecule is different from a nucleic acid molecule that exists in cells under natural conditions.
  • the present invention relates to a nucleic acid molecule comprising a nucleotide sequence encoding an amino acid sequence selected from SEQ ID NOs: 1-76.
  • a nucleic acid molecule can also comprise any combination of said nucleotide sequences.
  • the isolated nucleic acid is DNA.
  • the nucleic acid molecule of the invention may be isolated from any source that produces the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2. In certain embodiments, the nucleic acid molecule of the invention may be synthesized, rather than isolated.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain variable domain of the antibody 07-006, and includes a nucleotide sequence with SEQ ID NO: 77.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain variable domain of the antibody 07-006, and includes a nucleotide sequence with SEQ ID NO: 78.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain variable domain of the antibody 07-015, and includes a nucleotide sequence with SEQ ID NO: 79.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain variable domain of the antibody 07-015, and includes a nucleotide sequence with SEQ ID NO: 80.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain variable domain of the antibody 07-016, and includes a nucleotide sequence with SEQ ID NO: 81.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain variable domain of the antibody 07-016, and includes a nucleotide sequence with SEQ ID NO: 82.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain variable domain of the antibody 07-028, and includes a nucleotide sequence with SEQ ID NO: 83. In some embodiments of the invention, the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain variable domain of the antibody 07-028, and includes a nucleotide sequence with SEQ ID NO: 84.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain variable domain of the antibody 07-031, and includes a nucleotide sequence with SEQ ID NO: 85.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain variable domain of the antibody 07-031, and includes a nucleotide sequence with SEQ ID NO: 86.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain variable domain of the antibody 07-041, and includes a nucleotide sequence with SEQ ID NO: 87.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain variable domain of the antibody 07-041, and includes a nucleotide sequence with SEQ ID NO: 88.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain of the antibody 10-001, and includes a nucleotide sequence with SEQ ID NO: 89.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain of the antibody 10-001, and includes a nucleotide sequence with SEQ ID NO: 90.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain of the antibody 10-002, and includes a nucleotide sequence with SEQ ID NO: 91.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain of the antibody 10-002, and includes a nucleotide sequence with SEQ ID NO: 92.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain of the antibody 10-003, and includes a nucleotide sequence with SEQ ID NO: 93.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain of the antibody 10-003, and includes a nucleotide sequence with SEQ ID NO: 94. In some embodiments of the invention, the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain of the antibody 10-007, and includes a nucleotide sequence with SEQ ID NO: 95.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain of the antibody 10-007, and includes a nucleotide sequence with SEQ ID NO: 96.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain of the antibody 10-008, and includes a nucleotide sequence with SEQ ID NO: 97.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain of the antibody 10-008, and includes a nucleotide sequence with SEQ ID NO: 98.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the heavy chain of the antibody 10-009, and includes a nucleotide sequence with SEQ ID NO: 99.
  • the nucleic acid is a nucleic acid that encodes the amino acid sequence of the light chain of the antibody 10-009, and includes a nucleotide sequence with SEQ ID NO: 100.
  • the nucleic acid molecules may be used to express the monoclonal antibody or antigenbinding fragment thereof that specifically binds to GD2.
  • the present invention relates to an expression vector comprising the above isolated nucleic acid.
  • the present invention relates to a vector suitable for the expression of any of nucleotide sequences described herein.
  • vector means a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • the vector is a plasmid, i.e., a circular double stranded piece of DNA into which additional DNA segments may be ligated.
  • the vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • vectors are capable of autonomous replication in a host cell into which they are introduced (e.g. bacterial vectors having a bacterial origin site of replication and episomal mammalian vectors).
  • vectors e.g.
  • non-episomal mammalian vectors may be integrated into the genome of a host cell upon introduction into a host cell, and thereby are replicated along with the host gene.
  • certain vectors are capable of directing the expression of genes to which they are operably linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply, “expression vectors”).
  • the present invention relates to vectors comprising nucleic acid molecules that encode any of the amino acid sequences of a monoclonal antibody that specifically binds to GD2 or portions thereof (e.g. heavy chain and/or light chain binding domain sequences), as described herein.
  • the invention further relates to vectors comprising nucleic acid molecules encoding the antibodies or fragments thereof.
  • Expression vectors include plasmids, retroviruses, adenoviruses, adeno-associated viruses (AAVs), plant viruses, such as cauliflower mosaic virus, tobacco mosaic virus, cosmids, YACs, EBV derived episomes, and the like.
  • DNA molecules may be ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the DNA.
  • An expression vector and expression control sequences may be chosen to be compatible with the expression host cell used.
  • DNA molecules partially or fully encoding the sequences of first and second binding domains (for example, heavy and light chain sequences where a binding domain comprises a heavy and light chain sequence) can be introduced into individual vectors.
  • any combination of said DNA molecules is introduced into the same expression vector.
  • DNA molecules may be introduced into an expression vector by standard methods (e.g. ligation of complementary restriction sites on an antibody gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • a suitable vector is one that includes restriction sites such that any VH or VL sequence can easily be inserted and expressed, as described above. Polyadenylation and transcription termination may occur at a native chromosomal site downstream of coding regions.
  • a recombinant expression vector can also encode a signal peptide that facilitates secretion of an antibody chain from a host cell.
  • An antibody chain gene may be cloned into a vector such that the signal peptide is linked in-frame to the amino terminus of an immunoglobulin chain.
  • the signal peptide may be an immunoglobulin signal peptide or a heterologous signal peptide (i.e. a signal peptide from a non-immunoglobulin protein).
  • the recombinant vector expression of the invention can carry regulatory sequences that control the expression of antibody chain genes in a host cell. It will be understood by those skilled in the art that the design of an expression vector, including the selection of regulatory sequences, may depend on such factors as the choice of a host cell to be transformed, the level of expression of a desired protein, and so forth.
  • Preferred control sequences for an expression host cell in mammals include viral elements that ensure high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from a retroviral LTR, cytomegalovirus (CMV) (such as a CMV promoter/enhancer), simian virus 40 (SV40) (such as a SV40 promoter/enhancer), adenovirus, (e.g. the major late promoter adenovirus (AdMLP)), polyomavirus and strong mammalian promoters such as native immunoglobulin promoter or actin promoter.
  • CMV cytomegalovirus
  • SV40 simian virus 40
  • AdMLP major late promoter adenovirus
  • polyomavirus e.g. the major late promoter adenovirus (AdMLP)
  • AdMLP major late promoter adenovirus
  • the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of a vector in host cells (e.g. origins of replication) and selectable marker genes.
  • the selectable marker gene facilitates the selection of host cells into which a vector has been introduced.
  • the vector may include an expression control sequence.
  • expression control sequence refers to polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein secretion.
  • control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include the promoter of ribosome binding site, and transcription termination sequences; in eukaryotes, typically, such control sequences include promoters and transcription termination sequences.
  • control sequences includes at least all components, the presence of which is essential for expression and processing, and can also include additional components, the presence of which is advantageous, for example, leader sequences and fusion partner sequences.
  • the present invention relates to a method for producing a host cell to produce any above antibody or antigen-binding fragment thereof that specifically binds to GD2, and comprises transformation of the cell with the above vector.
  • the present invention relates to a host cell for producing any above antibody or antigen-binding fragment thereof that specifically binds to GD2, the host cell comprising any of the above nucleic acids.
  • recombinant host cell (or simply “host cell”) as used herein refers to a cell into which a recombinant expression vector has been introduced.
  • host cells which may include, for example, a vector according to the invention described above.
  • the present invention also relates to host cells that comprise, for example, a nucleotide sequence encoding a heavy chain or antigen-binding portions thereof, a light chain-encoding nucleotide sequence or antigen-binding portions thereof, or both, of the binding domain of the binding molecule of the invention.
  • host cells that comprise, for example, a nucleotide sequence encoding a heavy chain or antigen-binding portions thereof, a light chain-encoding nucleotide sequence or antigen-binding portions thereof, or both, of the binding domain of the binding molecule of the invention.
  • Nucleic acid molecules encoding the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention and vectors comprising these nucleic acid molecules may be used for transfection of a suitable mammalian or cell thereof, plant or cell thereof, bacterial or yeast host cell. Transformation may be carried out by any known technique of introducing polynucleotides into a host cell.
  • nucleic acid molecules may be introduced into mammalian cells by viral vectors.
  • Mammalian cell lines used as hosts for transformation are well known in the art and include a plurality of immortalized cell lines available. These include, e.g., Chinese hamster ovary (CHO) cells, NSO cells, SP2 cells, HEK-293T cells, FreeStyle 293 cells (Invitrogen), NIH-3T3 cells, HeLa cells, baby hamster kidney (BHK) cells, African green monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells, and a number of other cell lines. Cell lines are selected by determining which cell lines have high expression levels and provide for necessary characteristics of the protein produced. Other cell lines that may be used are insect cell lines, such as Sf9 or Sf21 cells.
  • the antibodies or fragments thereof are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibodies or fragments thereof in host cells or, more preferably, secretion of the antibodies or fragments thereof into the culture medium in which the host cells are grown.
  • the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 may be isolated from culture medium using standard protein purification techniques.
  • Plant host cells include e.g. Nicotiana, Arabidopsis, duckweed, corn, wheat, potato, etc.
  • Bacterial host cells include Escherichia and Streptomyces species.
  • Yeast host cells include Schizosaccharomyces pombe, Saccharomyces cerevisiae and Pichia pastoris.
  • level of production of the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 from a production cell line may be enhanced using a number of known techniques.
  • the glutamine synthetase gene expression system (the GS system) is a common approach for enhancing expression under certain conditions.
  • the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 from various cell lines will have a different glycosylation profile as compared to each other.
  • the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 encoded by nucleic acid molecules described herein, or comprising amino acid sequences provided herein are part of the present invention, regardless of the glycosylation of the binding molecules, and, in general, regardless of the presence or absence of post-translational modifications.
  • the above host cell does not refer to a host cell produced using human embryos.
  • the above host cell does not refer to a host cell produced by modifying the genetic integrity of human germline cells.
  • the present invention relates to a method for obtaining an antibody or antigen-binding fragment thereof that specifically binds to GD2, comprising culturing the above host cell in a growth medium under conditions sufficient to produce said antibody or fragment thereof, if necessary, followed by isolation and purification of the resulting antibody or fragment thereof.
  • the present invention relates to methods for obtaining the monoclonal antibody or antigenbinding fragment thereof that specifically binds to GD2 according to this invention.
  • One embodiment of the invention relates to a method for producing monoclonal antibody or antigenbinding fragment thereof that specifically binds to GD2, as defined herein, which comprises the production of a recombinant host cell capable of expressing the monoclonal antibody or antigenbinding fragment thereof that specifically binds to GD2, culturing of said host cell under conditions suitable for expression/production of the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2, and isolation of the resulting monoclonal antibody or fragment thereof that specifically binds to GD2.
  • the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2, produced by such expression in such recombinant host cells is referred to herein as "recombinant monoclonal antibody that specifically binds to GD2" or "an antigen-binding fragment of the recombinant monoclonal antibody that specifically binds to GD2" .
  • the invention also relates to the progeny from such host cells.
  • Another aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 as an active ingredient (or as the only active ingredient).
  • the present invention relates to a pharmaceutical composition used for treating a disease or disorder mediated by GD2, which comprises any of the above antibodies or antigen-binding fragments thereof in a therapeutically effective amount in combination with one or more pharmaceutically acceptable excipients.
  • “Pharmaceutical composition” refers to a composition comprising an antibody of the present invention and at least one of components selected from the group comprising pharmaceutically acceptable and pharmacologically compatible fillers, solvents, diluents, carriers, auxiliary, distributing and sensing agents, delivery agents, such as preservatives, stabilizers, filler, disintegrators, moisteners, emulsifiers, suspending agents, thickeners, sweeteners, flavouring agents, aromatizing agents, antibacterial agents, fungicides, lubricants, and prolonged delivery controllers, the choice and suitable proportions of which depend on the type and way of administration and dosage.
  • pharmaceutically acceptable and pharmacologically compatible fillers such as preservatives, stabilizers, filler, disintegrators, moisteners, emulsifiers, suspending agents, thickeners, sweeteners, flavouring agents, aromatizing agents, antibacterial agents, fungicides, lubricants, and prolonged delivery controllers, the choice and suitable proportions of which depend
  • suspending agents examples include ethoxylated isostearyl alcohol, polyoxyethene, sorbitol and sorbitol ether, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacant and their mixtures as well. Protection against action of microorganisms can be provided by various antibacterial and antifungal agents, such as, for example, parabens, chlorobutanole, sorbic acid, and similar compounds.
  • the composition may also contain isotonic agents, such as, for example, sugars, polyols, sodium chloride, and the like. Prolonged action of the composition may be achieved by agents slowing down absorption of active ingredient, for example, aluminum monostearate and gelatine.
  • suitable carriers, solvents, diluents and delivery agents are water, ethanol, polyalcohols and their mixtures, natural oils (such as olive oil) and organic esters (such as ethyl oleate) for injections.
  • suitable carriers are lactose, milk sugar, sodium citrate, calcium carbonate, calcium phosphate, and the like.
  • disintegrators and distributors are starch, alginic acid and its salts, silicates and the like.
  • lubricants are magnesium stearate, sodium lauryl sulfate, talc, and polyethylene glycol of high molecular weight as well.
  • the pharmaceutical composition for peroral, sublingual, transdermal, intraocular, intramuscular, intravenous, subcutaneous, local or rectal administration of active ingredient, alone or in combination with another active compound may be administered to human and animals in a standard administration form, in a mixture with traditional pharmaceutical carriers.
  • suitable standard administration forms include peroral forms such as tablets, gelatin capsules, pills, powders, granules, chewing-gums and peroral solutions or suspensions; sublingual and transbuccal administration forms; aerosols; implants; local, transdermal, subcutaneous, intramuscular, intravenous, intranasal or intraocular administration forms and rectal administration forms.
  • excipient or "auxiliary substance” is used herein to describe any ingredient other than the antibody of the present invention.
  • excipient or "auxiliary substance” is used herein to describe any ingredient other than the antibody of the present invention.
  • auxiliary substance are substances of inorganic or organic nature which are used in the pharmaceutical production/manufacturing in order to give drug products the necessary physicochemical properties.
  • compositions are intended to improve, prevent, or treat disorders that may be associated with GD2.
  • disease or disorder mediated by GD2 refers to any disease or disorder that is either directly, or indirectly associated with GD2, including etiology, development, progression, persistence or pathology of a disease or disorder.
  • Treat”, “treating” and “treatment” refer to a method of alleviating or abrogating a biological disorder and/or at least one of its attendant symptoms.
  • to “alleviate” a disease, disorder or condition means reducing the severity and/or occurrence frequency of the symptoms of the disease, disorder, or condition.
  • references herein to “treatment” include references to curative, palliative and prophylactic treatment.
  • the subject of treatment, or patient is a mammal, preferably a human subject.
  • Said subject may be either male or female, of any age.
  • disorder means any condition that would benefit from treatment with the compound of the present invention. This includes chronic and acute disorders or diseases including those pathological conditions that predispose the mammal to the disorder in question.
  • “Therapeutically effective amount” refers to that amount of the therapeutic agent being administered during treatment which will relieve to some extent one or more of the symptoms of the disease being treated.
  • the disease or disorder mediated by GD2 is selected from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • the pharmaceutical compositions of the present invention and methods of preparation thereof will be undoubtedly apparent to those skilled in the art.
  • the pharmaceutical compositions should preferably be manufactured in compliance with the GMP (Good Manufacturing Practice) requirements.
  • the composition may comprise a buffer composition, tonicity agents, stabilizers and solubilizers. Prolonged action of composition may be achieved by agents slowing down absorption of active pharmaceutical ingredient, for example, aluminum monostearate and gelatine.
  • suitable carriers, solvents, diluents and delivery agents include water, ethanol, polyalcohols and their mixtures, oils, and organic esters for injections.
  • Any method for administering peptides, proteins or antibodies which is accepted in the art may be suitably employed for the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention.
  • pharmaceutically acceptable refers to one or more compatible liquid or solid components that are suitable for administration in a mammal, preferably a human.
  • buffer refers to a solution, which is capable of resisting changes in pH by the action of its acid-base conjugate components, and which allows the product of antibody that specifically binds to GD2 to resist changes in pH.
  • the pharmaceutical composition preferably has a pH in the range from 4.0 to 8.0.
  • buffers used include, but are not limited to, acetate, phosphate, citrate, histidine, succinate, etc. buffer solutions.
  • tonic agent refers to an excipient that can increase the osmotic pressure of a liquid antibody formulation.
  • "Isotonic” drug is a drug that has an osmotic pressure equivalent to that of human blood. Isotonic drugs typically have an osmotic pressure from about 250 to 350 mOsm/kg. Isotonic agents used include, but are not limited to, polyols, saccharides and sucrose, amino acids, metal salts, for example, sodium chloride, etc.
  • Stabilizer refers to an excipient or a mixture of two or more excipients that provide the physical and/or chemical stability of the active agent.
  • Stabilizers may be amino acids, for example, but not limited to, arginine, histidine, glycine, lysine, glutamine, proline; surfactants, for example, but not limited to, polysorbate 20 (trade name: Tween 20), polysorbate 80 (trade name: Tween 80), polyethylene-polypropylene glycol and copolymers thereof (trade names: Poloxamer, Pluronic, sodium dodecyl sulfate (SDS); antioxidants, for example, but not limited to, methionine, acetylcysteine, ascorbic acid, monothioglycerol, sulfurous acid salts, etc.; chelating agents, for example, but not limited to, ethylenediaminetetraacetic acid (EDTA), diethylenetriaminepentaacetic acid (DTP A), sodium
  • the pharmaceutical composition according to the invention is a stable composition.
  • the pharmaceutical composition is "stable" if the active agent retains physical stability and/or chemical stability and/or biological activity thereof during the specified shelf life at storage temperature, for example, of 2-8 °C.
  • the active agent retains both physical and chemical stability, as well as biological activity. Storage period is adjusted based on the results of stability test in accelerated or natural aging conditions.
  • a pharmaceutical composition according to the invention may be manufactured, packaged, or widely sold in the form of a single unit dose or a plurality of single unit doses in the form of a ready formulation.
  • single unit dose refers to discrete quantity of a pharmaceutical composition containing a predetermined quantity of an active ingredient.
  • the quantity of the active ingredient typically equals the dose of the active ingredient to be administered in a subject, or a convenient portion of such dose, for example, half or a third of such dose.
  • compositions according to the present invention are typically suitable for parenteral administration as sterile formulations intended for administration in a human body through the breach in skin or mucosal barriers, bypassing the gastrointestinal tract by virtue of injection, infusion and implantation.
  • parenteral administration includes, inter alia, subcutaneous, intraperitoneal, intramuscular, intravenous, intraarterial, intrathecal, intraventricular, intraurethral, intracranial, intrasynovial, transdermal injection or infusion, and kidney dialytic infusion techniques.
  • Intra-tumor delivery for example, intra-tumor injection, may also be employed. Regional perfusion is also contemplated.
  • Preferred embodiments include intravenous and subcutaneous routes. Any method for administering peptides or proteins, which is accepted in the art may be suitably employed for the antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention.
  • Injectable formulations may be prepared, packaged, or sold, without limitation, in unit dosage form, such as in ampoules, vials, in plastic containers, pre-filled syringes, autoinjection devices.
  • Formulations for parenteral administration include, inter alia, suspensions, solutions, emulsions in oily or aqueous bases, pastes, and the like.
  • the invention provides a composition for parenteral administration comprising a pharmaceutical composition which is provided in dry (i.e. powder or granular) form for reconstitution with a suitable base (e.g. sterile pyrogen-free water) prior to administration.
  • a suitable base e.g. sterile pyrogen-free water
  • Such medicinal formulation may be prepared by, for example, lyophilization, i.e. a process, which is known in the art as freeze drying, and which involves freezing a product followed by removal of solvent from frozen material.
  • the antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention may also be administered intranasally or by inhalation, either alone, as a mixture with a suitable pharmaceutically acceptable excipient from an inhaler, such as a pressurised aerosol container, pump, spray, atomiser, or nebuliser, where a suitable propellant is used or not used, or as nasal drops, or spray.
  • an inhaler such as a pressurised aerosol container, pump, spray, atomiser, or nebuliser, where a suitable propellant is used or not used, or as nasal drops, or spray.
  • Medicinal formulations for parenteral administration may be formulated to be immediate or modified release.
  • Modified release medicinal formulations include delayed-, sustained-, pulsed- , controlled-, targeted and programmed release.
  • the present invention relates to a pharmaceutical composition for treating a disease or disorder mediated by GD2, the pharmaceutical combination comprising any above antibody or antigen-binding fragment thereof and at least one other therapeutically active compound.
  • the other therapeutically active compound is an antibody, chemotherapeutic agent, or hormone therapy agent.
  • the other therapeutically active compound is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is selected from a PD-1 inhibitor, PD-L1 inhibitor, or CTLA-4 inhibitor.
  • the PD-1 inhibitor is an antibody that specifically binds to PD-1.
  • the antibody that specifically binds to PD-1 is selected from the group: prolgolimab, pembrolizumab, nivolumab.
  • the CTLA-4 inhibitor is an antibody that specifically binds to CTLA-4.
  • the antibody that specifically binds to CTLA-4 is ipilimumab or nurulimab.
  • the PD-L1 inhibitor is an antibody that specifically binds to PD-L1.
  • the antibody that specifically binds to PD-L1 is selected from the group: durvalumab, avelumab, atezolizumab, manelimab.
  • the other therapeutically active compound is selected from the group: IL-2, GM-CSF, isotretinoin, one or more other cytokines, or any combination of therapeutically active compounds from this group.
  • the antibody or antigen-binding fragment thereof that specifically binds to GD2 is used in the treatment of disorders mediated by GD2 activity.
  • the subject of treatment, or patient is a mammal, preferably a human subject.
  • Said subject may be either male or female, of any age.
  • the therapeutically effective amount of an antibody or fragment thereof may reduce the number of cancer cells; reduce the initial tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit to some extent tumor growth; and/or relieve to some extent one or more of the symptoms associated with the disorder.
  • the antibody or fragment thereof may to some extent prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • in vivo efficacy can, for example, be measured by assessing survival, time to tumor progression (TTP), tumor response rate to treatment (RR), duration of response and/or quality of life.
  • the antibody or antigen-binding fragment thereof that specifically binds to GD2 may be administered without further therapeutic treatment, i.e. as an independent therapy.
  • the present invention relates to a method for inhibiting the biological activity of GD2 in a subject in need of such inhibition, comprising administering an effective amount of any above antibody or antigen-binding fragment thereof.
  • the present invention relates to a method for treatment of a disease or disorder mediated by GD2, which comprises administering in a subject in need of such treatment any above antibody or antigen-binding fragment thereof or said pharmaceutical composition, in a therapeutically effective amount.
  • the present invention relates to a method for treating a disease or disorder mediated by GD2, that comprises administering in a subject in need of such treatment any of the above antibodies or antigen-binding fragments thereof, and selected from the group: a) administration of at least one other therapeutically active compound, b) radiotherapy, c) hematopoietic stem cell transplantation, d) surgical treatment and, if necessary, adjuvant therapy, or e) any combination of the above a) to d).
  • the disease or disorder mediated by GD2 is selected from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • other therapeutically active compound is an antibody, chemotherapeutic agent, or hormone therapy agent.
  • chemotherapeutic agent is a chemical compound useful in the treatment of a malignant neoplasm.
  • chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylmelamine; acetogenins (e.g.
  • bullatacin and bullatacinone delta-9-tetrahydrocannabinol (dronabinol MARINOL®); beta-lapachone; lapachol; colchicines; betulinic acid; camptothecin (including the synthetic analogue topotecan (HYCAMTIN®), CPT-11 (irinotecan, CAMPTOSAR®), acetylcamptothecin, scopolectin, and 9- aminocamptothecin); bryostatin; cally statin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); podophyllotoxin; podophyllinic acid; teniposide; cryptophycins (e.g.
  • cryptophycin 1 and cryptophycin 8 dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g.
  • calicheamicin e.g. calicheamicin gamma II and calicheamicin omega II (see, e.g. Agnew, Chem. Inti. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including ADRIAMYCIN®, morpholino-doxorubicin, cyanomorpholin
  • paclitaxel TAXOL®
  • albumin- engineered nanoparticle formulation of paclitaxel ABRAXANE®
  • docetaxel TXOTERE®
  • chlorambucil 6-thioguanine
  • mercaptopurine methotrexate
  • platinum agents such as cisplatin, oxaliplatin, and carboplatin
  • vinca alkaloids which prevent tubulin polymerization from forming microtubules, including vinblastine (VELBAN®), vincristine (ONCOVIN®), vindesine (ELDISINE®), FILDESIN®), and vinorelbine (NAVELBINE®)
  • etoposide VP- 16
  • ifosfamide mitoxantrone; leucovorin; novantrone; edatrexate; daunomycin; aminopterin; ibandronate; topoisomerase inhibitor RFS 2000; difluorometlhylornithine (DMFO
  • ELOXATINTM oxaliplatin
  • Hormonal agents are agents that act to regulate or inhibit hormone action on tumors.
  • anti-estrogens with mixed agonist/antagonist profile including, tamoxifen (NOLVADEX®), 4-hydroxytamoxifen, toremifene (FARESTON®), idoxifene, droloxifene, raloxifene (EVTSTA®), trioxifene, keoxifene, and selective estrogen receptor modulators (SERMs), such as SERM3; pure anti-estrogens without agonist properties, such as fulvestrant (FASLODEX®), and EM800 (such agents may block estrogen receptor (ER) dimerization, inhibit DNA binding, increase ER turnover, and/or suppress ER levels); aromatase inhibitors, including steroidal aromatase inhibitors, such as formestane and exemestane (AROMASESl®), and nonsteroidal aromatase inhibitors, such as anastrazole (AREVIIDEX®
  • the other therapeutically active compound is an immune checkpoint inhibitor.
  • immune checkpoint inhibitor refers to compounds that inhibit the activity of immune checkpoints. Inhibition includes reduction of function and full blockade.
  • inhibitory checkpoint molecules include B7-H3, B7-H4, BTLA, CTLA-4, KIR, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, TIGIT, and VISTA.
  • the immune checkpoint inhibitor is an antibody that specifically recognizes an immune checkpoint protein.
  • a number of immune checkpoint inhibitors are known and in analogy of these known immune checkpoint protein inhibitors, alternative immune checkpoint inhibitors may be developed in the near future.
  • the immune checkpoint inhibitors include, but are not limited to, peptides, antibodies, nucleic acid molecules, and low molecular weight compounds.
  • the immune checkpoint inhibitor is selected from a PD-1 inhibitor, PD-L1 inhibitor, or CTLA-4 inhibitor.
  • the PD-1 inhibitor is an antibody that specifically binds to PD-1.
  • the PD-1 inhibitor is an antibody that specifically binds to PD-1.
  • antibodies that specifically bind to PD-1 include pembrolizumab, nivolumab, prolgolimab, toripalimab, cemiplimab, sintilimab and others. The most preferred ones are prolgolimab, pembrolizumab, nivolumab.
  • the antibody that specifically binds to PD-1 is selected from the group comprising prolgolimab, pembrolizumab, nivolumab.
  • the CTLA-4 inhibitor is an antibody that specifically binds to CTLA-4. In some embodiments of the invention, the CTLA-4 inhibitor is an antibody that specifically binds to CTLA-4. Examples of antibodies that specifically bind to CTLA4 include ipilimumab, tremelimumab, zalifrelimab, nurulimab and others. The most preferred ones are ipilimumab or nurulimab.
  • the antibody that specifically binds to CTLA-4 is ipilimumab or nurulimab.
  • the PD-L1 inhibitor is an antibody that specifically binds to PD-L1.
  • the antibody that specifically binds to PD-L1 is selected from the group: durvalumab, avelumab, atezolizumab, manelimab.
  • the other therapeutically active compound is selected from the group: IL-2, GM-CSF, isotretinoin, one or more other cytokines, or any combination of therapeutically active compounds from this group.
  • the present invention relates to the use of the above antibody or antigenbinding fragment thereof or the above pharmaceutical composition for treating in a subject in need of such treatment a disease or disorder mediated by GD2.
  • the present invention relates to the use of any of the above antibody or antigen-binding fragment thereof and at least one of the group: a) other therapeutically active compound, b) radiotherapy, c) hematopoietic stem cell transplantation or d) surgical treatment and, if necessary, adjuvant therapy, for treating a disease or disorder mediated by GD2.
  • the disease or disorder mediated by GD2 is selected from the group: brain tumor, neuroblastoma, glioblastoma, medulloblastoma, retinoblastoma, astrocytoma, melanoma, B-cell lymphoma, small cell lung cancer, renal carcinoma, desmoplastic small round cell fibroma, osteosarcoma, Ewing's sarcoma, breast cancer, rhabdomyosarcoma, leiomyosarcoma, liposarcoma, fibrosarcoma, or soft tissue sarcoma.
  • the other therapeutically active compound is an immune checkpoint inhibitor.
  • the immune checkpoint inhibitor is selected from a PD- 1 inhibitor, PD-L1 inhibitor, or CTLA-4 inhibitor.
  • the PD-1 inhibitor is an antibody that specifically binds to PD-1.
  • the antibody that specifically binds to PD-1 is selected from the group: prolgolimab, pembrolizumab, nivolumab.
  • the CTLA-4 inhibitor is an antibody that specifically binds to CTLA-4.
  • the antibody that specifically binds to CTLA-4 is ipilimumab or nurulimab.
  • the PD-L1 inhibitor is an antibody that specifically binds to PD-L1.
  • the antibody that specifically binds to PD-L1 is selected from the group: durvalumab, avelumab, atezolizumab, manelimab.
  • the other therapeutically active compound is selected from the group: IL-2, GM-CSF, isotretinoin, one or more other cytokines, or any combination of therapeutically active compounds from this group.
  • the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention will be administered in an amount that is effective in treatment of the condition in question, i.e. in doses and during the periods of time required to achieve the desired result.
  • a therapeutically effective amount may vary according to factors such as the particular condition being treated, the age, sex and weight of the patient, and whether the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 is being administered as a stand-alone treatment or in combination with one or more additional drugs or treatments.
  • Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in a unit dosage form for ease of administration and uniformity of dosage.
  • a unit dosage form as used herein refers to physically discrete units suited as unitary dosages for patients/subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the desired pharmaceutical carrier.
  • the unit dosage forms of the invention is typically dictated by and directly dependent on (a) the unique characteristics of a therapeutic agent and particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in subjects.
  • the doses and dosage regimen are adjusted in accordance with methods well-known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a detectable therapeutic effect to a patient may also be determined, as can the temporal requirements for administering each agent to provide a detectable therapeutic effect to a patient.
  • certain doses and administration regimens are exemplified herein, these examples in no way limit the doses and administration regimens that may be provided to a patient in practicing the embodiments of the invention.
  • dosage values may vary with the type and severity of the condition to be alleviated, and may include single or multiple doses.
  • specific dosage regimens should be adjusted over time according to the individual need and the judgment of a medical professional administering or supervising the administration of the compositions, and that dosage ranges set forth in the present description are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
  • the dosage regimen with the compositions of this invention may be based on a variety of factors, including the type of disease, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of administration, and the particular monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 employed.
  • the dosage regimen may vary widely, but can be determined routinely using standard methods. For example, doses may be adjusted based on pharmacokinetic and pharmacodynamic parameters, which may include clinical effects such as toxic effects or laboratory values.
  • the present invention encompasses intra-patient dose-escalation as determined by one skilled in the art. Methods for determining appropriate dosage and regimen are well-known in the art and would be understood by a skilled artisan once provided the ideas disclosed herein.
  • a suitable dose of a monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention will be in the range of 0.1-200 mg/kg, preferably 0.1-100 mg/kg, including about 0.5-50 mg/kg, for example about 1-20 mg/kg.
  • the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 may be administered, e.g. in a dose of at least 0.25 mg/kg, such as at least 0.5 mg/kg, including at least 1 mg/kg, e.g. at least 1.5 mg/kg, such as at least 2 mg/kg, e.g. at least 3 mg/kg, including at least 4 mg/kg, e.g.
  • the administration will typically be repeated in appropriate time intervals, such as once a week, once every two weeks, once every three weeks or once every four weeks, and for as long as deemed appropriate by a responsible physician, who may, in some cases, increase or reduce the dose if necessary.
  • the monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention are also used in diagnostic purposes (e.g. in vitro, ex vivo).
  • the present monoclonal antibody or antigen-binding fragment thereof that specifically binds to GD2 according to the invention may be used for detecting or measuring the level of GD2 in samples obtained from a patient (e.g. tissue sample or a sample of body fluid, such as an inflammatory exudate, blood, serum, intestinal fluid, saliva or urine).
  • Suitable methods for detection and measurement include immunoassays, such as flow cytometry, enzyme-linked immunosorbent assay (ELISA), chemiluminescent assay, radioimmunoassay, and immunohi stol ogy .
  • immunoassays such as flow cytometry, enzyme-linked immunosorbent assay (ELISA), chemiluminescent assay, radioimmunoassay, and immunohi stol ogy .
  • Desired gene segments were prepared from oligonucleotides made by chemical synthesis.
  • the gene segments of 300-1400 bp long, which were flanked by singular restriction sites, were assembled by annealing and ligation of oligonucleotides including PCR amplification and subsequently cloned via the restriction sites.
  • the DNA sequences of the subcloned gene fragments were confirmed by DNA sequencing.
  • DNA sequences were determined by Sanger sequencing.
  • the Unipro's UGENE suite version 1.29 and SnapGene Viewer were used for sequence creation, mapping, analysis, annotation and illustration.
  • variants of expression plasmids intended for expression of antibodies in prokaryotic cells E.coli
  • transient expression in eukaryotic cells e.g., in CHO cells
  • the vectors contained: an origin of replication which allows replication of said plasmid in E. coli, genes which confer resistance in E. coli to various antibiotics (e.g. to ampicillin, kanamycin).
  • the fusion genes comprising the described antibody chains as described below were generated by PCR and/or gene synthesis and assembled with known recombinant methods and techniques by connection of the according nucleic acid segments, e.g. using unique restriction sites in the corresponding vectors. The subcloned nucleic acid sequences were verified by DNA sequencing. For transient transfections, larger quantities of the plasmids were prepared by plasmid preparation from transformed E. coli cultures.
  • Anti-GD2 antibody molecules were created using structural data obtained in silico. In silico scaffolding was performed using the internal algorithm of JSC "Biocad”. The PrepWizard instrument from Schrodinger Suite 2017-2 was employed to prepare the structures. It was followed by folding using the Prime instrument from Schrodinger Suite 2017-2.
  • the antibodies were optimized in silico to thereby produce antibody candidates for further study, the antibody candidates are indicated in Table 1.
  • leader antibodies were selected from Table 1 as follows: 07-006, 07-015, 07-016, 07- 028, 07-031, 07-041; these antibodies surprisingly showed the best parameters (see examples below).
  • Table 2 shows the identity analysis for the heavy chain variable fragments of anti-GD2 antibodies 07-006, 07-015, 07-016, 07-028, 07-031 or 07-041.
  • the heavy chain variable fragments of anti-GD2s according to the invention have at least 98 % identity to each other.
  • Table 3 shows the humanization analysis for the heavy chain variable fragments of anti- GD2 antibodies 07-006, 07-015, 07-016, 07-028, 07-031 or 07-041.
  • the heavy chain variable fragments of anti-GD2 antibodies according to the invention have a degree of humanization of more than 80 %.
  • Table 4 shows the identity analysis for the light chain variable fragments of anti-GD2 antibodies 07-006, 07-015, 07-016, 07-028, 07-031 or 07-041 Table 4. % identity in antibody VLs
  • the light chain variable fragments of anti-GD2 antibodies according to the invention have at least 96 % identity to each other.
  • Table 5 shows the humanization analysis for the light chain variable fragments of anti- GD2 antibodies 07-006, 07-015, 07-016, 07-028, 07-031 or 07-041.
  • the light variable fragments of candidate anti-GD2s according to the invention have a degree of humanization of more than 80 %.
  • genes of the heavy /light chain variable domains of the antibody to GD2 according to the invention that is selected from the group: 07-006, 07-015, 07-016, 07-028, 07-031 or 07-041, were synthesized de novo. To this end, we synthesized oligonucleotides of 55-60 bp each forming a completely overlapping gene sequence. Each gene was assembled using two-round PCR, which resulted in production of fragments of 339 bp each.
  • Fusion of the heavy chain variable domain gene and the Fc fragment of human IgGl, the light chain variable domain and CK were performed using PCR and/or gene synthesis and assembly using known recombination methods and processes by connecting the appropriate nucleic acid segments, for example, using SOE-PCR (Splicing by overlap extension).
  • the heavy and light chain genes of the antibody to GD2 according to the invention selected from the group: 07-006, 07-015, 07-016, 07-028, 07-031 or 07-041 were cloned into PEE plasmids for producing protein in the IgGl format in mammalian cells.
  • the cloned nucleic acid sequences were verified by DNA sequencing.
  • the desired quantities of the resulting plasmids ( Figures 2 and 3) were produced in E.coli cells and purified using a commercial plasmid DNA isolation kit from Qiagen.
  • the resulting gene constructs were transferred for transient production of proteins in CHO cell line.
  • the Fc heavy chain constant domain was modified by introducing point mutations M252Y, S254T, T256 (YTE) and/or K322A.
  • the set of YTE mutations makes it possible to achieve prolonged pharmacokinetics, whereas the introduction of the K322A mutation reduces the complement-dependent cytotoxicity of the resulting antibodies.
  • the Fc portion of the antibody was afucosylated, thus leading to increased antibody-dependent cellular cytotoxicity.
  • Table 6 The resulting antibodies are shown in Table 6.
  • Assembly of genetic constructs included the fusion of the heavy chain variable domain gene and the Fc of human IgGl, into which point mutations were pre-introduced.
  • Heavy chain genes with substitutions were cloned into pEE plasmids for producing protein together with the already produced light chain constructs, in the IgGl format in the CHO-lg6-Fut8 cell line.
  • the cloned nucleic acid sequences were verified by DNA sequencing.
  • the required quantities of the resulting plasmids ( Figures 2, 3) were cultured in E.coli cells and purified using Qiagen kit.
  • the resulting gene constructs were transferred for transient production of proteins in CHO- Ig6-Fut8 cell line.
  • Full-length antibodies were produced in the CHO cell growth medium, afucosylated forms of full-length antibodies were produced in the CHO-lg6-Fut8 cell growth medium. Following transfection of cells with expression vectors, orbital feed-batch cultivation was performed in serum-free medium for 7 days. Secretion of the antibodies in question was monitored using the Pall ForteBio's Octet RED96 system for molecular interactions analysis on protein A biosensors.
  • test anti-GD2 antibodies specifically bind to the ganglioside GD2 (Table 7) with high affinity.
  • Antibodies were heated in PBS pH 7.4 using an amplifier in plastic test tubes at 50°C for 48 hours, followed by a shift to +4°C. After the end of the program, the samples were analyzed before and following heating using analytical gel chromatography on a TSK Gel G3000 SWxl column. The areas of the target peaks of the samples before and following heating were compared. A less than 5% change in the area of the monomer peak following heating for 48 hours indicates the stability of the product and the possibility of long-term storage (Table 8).
  • Jurkat-NFAT-Luc-CD16 cells were cultured at 37°C with 5% CO2 onRPMI-1640 medium (10%FBS, 10 ⁇ g/ml gentamicin, 2mM L-glutamine, 0.3 ⁇ g/ml puromycin and 200 ⁇ g/ml hygromycin); SK-N-BE(2) was cultured under the same conditions in DMEM/F12 medium (10% FBS, 10 mcg/ml gentamicin and 2mM L-glutamine).
  • Figure 5 shows that antibody 10-008 has antibody-dependent cellular cytotoxicity in an assay using the reporter Jurkat-NFAT-Luc-CD16 cell line.
  • the SK-N-BE (2) (human neuroblastoma) cell line was used as target cells for the analysis.
  • the cells were grown in DMEM medium supplemented with 10% bovine serum.
  • bovine serum albumin 0.1% bovine serum albumin at a concentration of 1x10 6 cells/ml, and prepared a number of dilutions of the test antibody candidates.
  • test antibodies 07-041 dFuc (afucosylated variant of 07-041)
  • 10-007, 10-008, 10-009 cause the death of target cells in the presence of human serum.
  • Antibodies 10-008 and 10-009 have a decreased effect as compared to that of other antibodies (see Figure 6).
  • amino acid sequence CDR2 (Kabat) of the heavy chain variable domain of antibodies 07-006, 07-015, 07-016, 07-028, 07-031, 07-041, 10-001, 10-002, 10-003, 10-007, 10-008, 10-009 ⁇ 400> 3
  • Gly Ala lie Asp Pro Phe Tyr Gly Gly Thr Ser Tyr Asn Gin Lys Phe 50 55 60
  • Gly Ala lie Asp Pro Phe Tyr Gly Gly Thr Ser Tyr Asn Gin Lys Phe 50 55 60
  • Gly Ala lie Asp Pro Phe Tyr Gly Gly Thr Ser Tyr Asn Gin Lys Phe 50 55 60
  • Val Ser Gly Met lie Tyr Trp Gly Gin Gly Thr Leu Val Thr Val Ser 100 105 110
  • Gly Ala lie Asp Pro Phe Tyr Gly Gly Thr Ser Tyr Asn Gin Lys Phe 50 55 60
  • Gly Ala lie Asp Pro Phe Tyr Gly Gly Thr Ser Tyr Asn Gin Lys Phe 50 55 60
  • Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro Glu Val Thr
  • Gly Ala lie Asp Pro Phe Tyr Gly Gly Thr Ser Tyr Asn Gin Lys Phe 50 55 60

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
PCT/RU2022/050096 2021-03-24 2022-03-24 Monoclonal antibody that specifically binds to gd2 WO2022203552A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CR20230456A CR20230456A (es) 2021-03-24 2022-03-24 Anticuerpo monoclonal o fragmento de unión al antígeno del mismo que se une específicamente al gd2 (gangliósido gd2), y uso del mismo
CN202280024459.8A CN117677695A (zh) 2021-03-24 2022-03-24 与gd2特异性结合的单克隆抗体
CONC2023/0012549A CO2023012549A2 (es) 2021-03-24 2023-09-22 Anticuerpo monoclonal o fragmento de unión al antígeno del mismo que se une específicamente al gd2 (gangliósido gd2), y uso del mismo

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
RU2021107773A RU2796937C2 (ru) 2021-03-24 Моноклональное антитело или его антигенсвязывающий фрагмент, которое специфически связывается с gd2 (ганглиозид gd2), и его применение
RU2021107773 2021-03-24

Publications (1)

Publication Number Publication Date
WO2022203552A1 true WO2022203552A1 (en) 2022-09-29

Family

ID=83395963

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/RU2022/050096 WO2022203552A1 (en) 2021-03-24 2022-03-24 Monoclonal antibody that specifically binds to gd2

Country Status (9)

Country Link
CN (1) CN117677695A (es)
AR (1) AR125219A1 (es)
CL (1) CL2023002843A1 (es)
CO (1) CO2023012549A2 (es)
CR (1) CR20230456A (es)
EC (1) ECSP23072412A (es)
TW (1) TW202304989A (es)
UY (1) UY39687A (es)
WO (1) WO2022203552A1 (es)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005070967A2 (en) * 2004-01-22 2005-08-04 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
WO2011160119A2 (en) * 2010-06-19 2011-12-22 Memorial Sloan-Kettering Cancer Center Anti-gd2 antibodies
RU2680267C2 (ru) * 2013-03-15 2019-02-19 Мемориал Слоан Кеттеринг Кэнсер Сентер Высокоаффинные антитела к gd2

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005070967A2 (en) * 2004-01-22 2005-08-04 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
WO2011160119A2 (en) * 2010-06-19 2011-12-22 Memorial Sloan-Kettering Cancer Center Anti-gd2 antibodies
RU2680267C2 (ru) * 2013-03-15 2019-02-19 Мемориал Слоан Кеттеринг Кэнсер Сентер Высокоаффинные антитела к gd2

Also Published As

Publication number Publication date
CL2023002843A1 (es) 2024-03-22
UY39687A (es) 2022-08-31
ECSP23072412A (es) 2023-10-31
CO2023012549A2 (es) 2023-10-09
TW202304989A (zh) 2023-02-01
CN117677695A (zh) 2024-03-08
CR20230456A (es) 2023-12-13
AR125219A1 (es) 2023-06-28

Similar Documents

Publication Publication Date Title
US11840567B2 (en) Bispecific antibodies with specific binding to CD47 and PD-L1
US11236167B2 (en) Monoclonal antibody to PD-L1
CN107921131A (zh) Pd‑l1(“程序性死亡配体1”)抗体
RU2734432C1 (ru) Моноклональное антитело, которое специфически связывается с GITR
US20230011234A1 (en) Immunocytokine comprising heterodymeric protein complex based on il-15/il-15ra
WO2022203552A1 (en) Monoclonal antibody that specifically binds to gd2
IL309115A (en) Anti-CTLA-4 antibodies
RU2796937C2 (ru) Моноклональное антитело или его антигенсвязывающий фрагмент, которое специфически связывается с gd2 (ганглиозид gd2), и его применение
EP3875486A1 (en) Monoclonal antibody that specifically binds to cd20
RU2815823C2 (ru) Выделенное биспецифическое антитело, которое специфически связывается с CD47 и PD-L1
RU2779652C2 (ru) АНТИТЕЛА, СПЕЦИФИЧНЫЕ К CD47 и PD-L1
WO2022271053A1 (en) Isolated bispecific antibody that specifically binds to cd47 and pd-l1
EA044786B1 (ru) Моноклональное антитело, которое специфически связывается с gitr
EA043937B1 (ru) ИММУНОЦИТОКИН, ВКЛЮЧАЮЩИЙ ГЕТЕРОДИМЕРНЫЙ БЕЛКОВЫЙ КОМПЛЕКС НА ОСНОВЕ IL-15 И IL-15Rα, И ЕГО ПРИМЕНЕНИЕ
EA044757B1 (ru) Моноклональное антитело, которое специфически связывается с cd20
EA041915B1 (ru) Моноклональное антитело к pd-l1
OA20189A (en) Monoclonal antibody that specifically binds to CD20.

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: 22776218

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: MX/A/2023/011246

Country of ref document: MX

Ref document number: 2301006077

Country of ref document: TH

Ref document number: 002713-2023

Country of ref document: PE

WWE Wipo information: entry into national phase

Ref document number: 202280024459.8

Country of ref document: CN

Ref document number: P6002425/2023

Country of ref document: AE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112023019502

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 202392653

Country of ref document: EA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 11202307212V

Country of ref document: SG

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112023019502

Country of ref document: BR

Free format text: COM BASE NA PORTARIA 48 DE 20/06/2022, SOLICITA-SE QUE SEJA APRESENTADO, EM ATE 60 (SESSENTA) DIAS, NOVO CONTEUDO DE LISTAGEM DE SEQUENCIA POIS O CONTEUDO APRESENTADO NA PETICAO NO 870230101315, DE 17/11/2023 NAO ESTA EM LINGUA VERNACULA.

WWE Wipo information: entry into national phase

Ref document number: 523450814

Country of ref document: SA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112023019502

Country of ref document: BR

Free format text: REAPRESENTE O CONTEUDO DA LISTAGEM DE SEQUENCIAS COM OS CAMPOS 140 / 141 , POIS JA SE CONHECE O NUMERO DO PROCESSO NO BRASIL.

122 Ep: pct application non-entry in european phase

Ref document number: 22776218

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 112023019502

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20230922