US20220073625A1 - Humanized anti-axl antibodies - Google Patents

Humanized anti-axl antibodies Download PDF

Info

Publication number
US20220073625A1
US20220073625A1 US17/372,961 US202117372961A US2022073625A1 US 20220073625 A1 US20220073625 A1 US 20220073625A1 US 202117372961 A US202117372961 A US 202117372961A US 2022073625 A1 US2022073625 A1 US 2022073625A1
Authority
US
United States
Prior art keywords
seq
amino acid
antibody
acid sequence
chain variable
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US17/372,961
Inventor
Patricius Hendrikus Cornelis Van Berkel
David G. Williams
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
BerGenBio ASA
Original Assignee
BerGenBio ASA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by BerGenBio ASA filed Critical BerGenBio ASA
Priority to US17/372,961 priority Critical patent/US20220073625A1/en
Publication of US20220073625A1 publication Critical patent/US20220073625A1/en
Assigned to BERGENBIO ASA reassignment BERGENBIO ASA CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: BERGENBIO AS
Assigned to BERGENBIO ASA reassignment BERGENBIO ASA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VAN BERKEL, Patricius Hendrikus Cornelis
Assigned to BERGENBIO AS reassignment BERGENBIO AS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WILLIAMS, DAVID GARETH
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C07K2317/70Immunoglobulins 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present disclosure relates to humanized anti-Axl antibodies.
  • Axl is a member of the TAM (Tyro3-Axl-Mer) receptor tyrosine kinases (RTK) that share the vitamin K-dependent ligand Gas6 (growth arrest-specific 6).
  • TAM family RTKs regulate a diverse range of cellular responses including cell survival, proliferation, autophagy, migration, angiogenesis, platelet aggregation, and natural killer cell differentiation.
  • Axl is expressed in many embryonic tissues and is thought to be involved in mesenchymal and neural development, with expression in adult tissues largely restricted to smooth muscle cells (MGI Gene Expression Database; www.informatics.jax.org).
  • Axl activation is linked to several signal transduction pathways, including Aid, MAP kinases, NF- ⁇ B, STAT, and others. Originally identified as a transforming gene from a patient with chronic myelogenous leukaemia, Axl has since been associated with various high-grade cancers and correlated with poor prognosis.
  • Axl receptor overexpression has been detected in a wide range of solid tumours and myeloid leukaemia (Linger et al, Adv Cancer Res. 100: 35, 2008; Linger et al, Expert Opin Ther Targets. 14:1073, 2010).
  • Axl expression correlates with malignant progression and is an independent predictor of poor patient overall survival in several malignancies including pancreatic (Song et al, Cancer. 117:734, 2011), prostate (Paccez et al, Oncogene. 32:698, 2013), lung (Ishikawa et al. Ann Surg Oncol. 2012; Zhang et al, Nat Genet. 44:852, 2012), breast (Gjerdrum, Proc natl Acad Sci USA 107:1124, 2010), colon cancer (Yuen et al, PLoS One, 8:e54211, 2013) and acute myeloid leukaemia (AML) (Ben-Batalla et al, Blood 122:2443, 2013).
  • Ax signal transduction is activated by a protein ligand (Gas6) secreted by tumour associated macrophages (Loges et al, Blood. 115:2264, 2010) or autocrine mechanisms (Gjerdrum, Proc natl Acad Sci USA 107:1124, 2010), that drives receptor dimerization, autophosphorylation and downstream signalling, such as via PI3 kinase (PI3K)-AKT, particularly AKT and mitogen-activated protein kinase (MAPK) pathways (Korshunov, Clinical Science. 122:361, 2012).
  • PI3 kinase PI3 kinase
  • MAPK mitogen-activated protein kinase
  • Heterodimerization with other tyrosine kinase receptors e.g. epidermal growth factor receptor (EGFR) is also reported to occur (Linger et al, Expert Opin Ther Targets. 14:1073, 2010; Meyer et al Science
  • Axl-targeting agents block tumour formation, metastasis and reverse drug resistance (e.g. to erlotinib) by reversing EMT/CSC characteristics in several experimental cancer models, including triple negative breast cancer, hormone resistant prostate cancer and adenocarcinoma of the lung (Holland et al Cancer Res 70:1544, 2010; Gjerdrum, Proc natl Acad Sci USA 107:1124, 2010; Zhang et al. Nat Genet. 44: 852, 2012; Paccez et al, Oncogene. 32:698, 2013).
  • EP2267454A2 [Diagnosis and prevention of cancer cell invasion measuring . . . Axl—Max Planck]; WO2009063965 [anti Axl—Chugai Pharmaceutical]; WO2011159980A1 [anti-Axl—Genentech], WO2011014457A1 [combination treatments Axl and VEGF antagonists—Genentech]; WO2012-175691A1 [Anti Axl 20G7-D9—INSERM], WO2012-175692A1 [Anti Axl 3E3E8—INSERM]; WO2009/062690A1 [anti Axl—U3 Pharma] and WO2010/130751A1 [humanised anti Axl—U3 Pharma].
  • GB1410826.0 discloses the murine anti-Axl antibody designated herein as “mouse 1H12”.
  • mouse 1H12 the murine anti-Axl antibody designated herein as “mouse 1H12”.
  • the present disclosure concerns such antibodies, along with antibody-drug conjugates comprising the humanised 1H12 antibodies and PBD drug-moieties.
  • the present disclosure provides humanized anti-AXL antibodies derived from the ‘mouse 1H12’ antibody.
  • the present inventors have generated a number of humanised heavy chain variable regions (SEQ ID NOs: 2 and 3) and humanised light chain variable regions (SEQ ID NOs:5 to 8) with a view to creating antibodies that have lower immunogenicity in a human individual than the ‘mouse 1H12’ antibody or ‘chimeric 1H12’ antibody whilst retaining antigen-binding potency.
  • these humanised antibodies have also been found to have other advantageous properties, such as increased charge at physiological pH and improved affinity for some Axl ligands.
  • the present disclosure comprises an isolated humanized antibody that binds to AXL, wherein the isolated humanized antibody comprises a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1, 2, or 3.
  • the antibody further comprises a light chain variable region having the amino acid sequence of SEQ ID NO: 4, 5, 6, 7, or 8 and, optionally, further comprises a constant region derived from one or more human antibodies.
  • the isolated humanized antibody that binds to AXL comprises; a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 or 3; a light chain variable region having the amino acid sequence of SEQ ID NO: 5, 6, 7, or 8; and, optionally, comprises a constant region derived from one or more human antibodies.
  • the humanized antibody does not comprise a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4.
  • AXL is human AXL.
  • sequences of the antibody heavy chain variable regions and/or the light chain variable regions disclosed herein may be modified by, for example, insertions, substitutions and/or deletions to the extent that the humanized antibody maintains the ability to bind to AXL.
  • the skilled person can ascertain the maintenance of this activity by performing the functional assays described herein, or known in the art.
  • the heavy chain variable region comprises no more than 20 insertions, substitutions and/or deletions, such as no more than 15, no more than 10, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 insertion, substitution and/or deletion.
  • the light chain variable region comprises no more than 20 insertions, substitutions and/or deletions, such as no more than 15, no more than 10, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 insertion, substitution and/or deletion.
  • humanized antibodies of the disclosure include antibodies comprising V H and V L domains with amino acid sequences that are identical to the sequences described herein.
  • the antibody of the conjugates described herein is an antibody (Ab) which binds AXL. That is, the conjugates described herein are conjugates comprising antibodies which specifically bind to AXL.
  • AXL refers to the Axl member of the TAM family of receptor tyrosine kinases.
  • Human Axl refers to the Axl member of the human TAM family of receptor tyrosine kinases.
  • the human Axl polypeptide corresponds to Genbank accession no. AAH32229, version no. AAH32229.1 G1:21619004, record update date: Mar. 6, 2012 01:18 PM (SEQ ID NO.9).
  • the nucleic acid encoding the human Axl polypeptide corresponds to Genbank accession no. M76125, version no. M76125.1 G1:292869, record update date: Jun. 23, 2010 08:53 AM.
  • Murine Axl refers to the Axl member of the murine TAM family of receptor tyrosine kinases.
  • the murine Axl polypeptide corresponds to Genbank accession no. AAH46618, version no. AAH46618.1 G1:55777082, record update date: Mar. 6, 2012 01:36 PM (SEQ ID NO.10).
  • the nucleic acid encoding the murine Axl polypeptide corresponds to Genbank accession no. NM_009465, version no. NM_009465.4 GI:300794836, record update date: Mar. 12, 2014 03:52 PM.
  • the humanized antibody binds human AXL with a dissociation constant (K D ) of at least 10 ⁇ 6 M, such as at least 5 ⁇ 10 ⁇ 7 M, at least 10 ⁇ 7 M, at least 5 ⁇ 10 ⁇ 8 M, at least 10 ⁇ 9 M, such as at least 5 ⁇ 10 ⁇ 10 M, at least 10 ⁇ 10 M, at least 5 ⁇ 10 ⁇ 11 M, at least 10 ⁇ 11 M, at least 5 ⁇ 10 ⁇ 12 M, at least 10 ⁇ 12 M, at least 5 ⁇ 10 ⁇ 13 M, at least 10 ⁇ 13 M, at least 5 ⁇ 10 ⁇ 14 M, at least 10 ⁇ 14 M, at least 5 ⁇ 10 ⁇ 15 M, or at least 10 ⁇ 15 M.
  • K D dissociation constant
  • the humanized antibody competitively inhibits the in vivo and/or in vitro binding to human AXL of an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4.
  • the humanized antibody competitively inhibits the in vivo and/or in vitro binding to human-AXL of the ‘mouse 1H12’ antibody.
  • an equimolar dose of the humanised antibody competitively inhibits at least 20% of the binding by the ‘mouse 1H12’ antibody, such as at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of the binding. Percentage binding may be measured by, for example, a competitive ELISA assay where % inhibition of binding is calculated as [(1 ⁇ absorbance of test sample)/(absorbance of negative control)].
  • the humanized antibody has a higher affinity for an Axl antigen (for example the Axl-Strep-His antigen described in Protocol 4) than an antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies (for example, Abi described herein).
  • the KD of the humanized antibody with the Axl antigen (for example the Axl-Strep-His antigen described in Protocol 4) will be no more than 0.9 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO.
  • a constant region derived from one or more human antibodies for example no more than 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.05, 0.01, or 0.001 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies.
  • the humanized antibody has a higher affinity for an Axl antigen (for example the Axl-Fc antigen described in Protocol 4) than an antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies (for example, Abi described herein).
  • the KD of the humanized antibody with the Axl antigen (for example the Axl-Fc antigen described in Protocol 4) will be no more than 0.9 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO.
  • a constant region derived from one or more human antibodies for example no more than 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.05, 0.01, or 0.001 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies.
  • a molecule carries no net charge when the pH of its surrounding equal the molecules pI.
  • the net charge of a molecule affects the solubility of the molecule, with biological molecules such as proteins typically having minimum solubility in water or salt solutions at the pH that corresponds to their pI.
  • proteins whose pI is 7.35-7.45 are at their minimum solubility in human blood, whose pH is typically in the range 7.35-7.45.
  • the humanized antibody of the disclosure has a pI greater than an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4. In some embodiments the humanized antibody of the disclosure has a pI greater than the mouse 1H12 antibody. In some embodiments the humanized antibody of the disclosure has a pI of at least 8.00, such as at least 8.05, at least 8.10, at least 8.15, at least 8.20, at least 8.30, at least 8.40, at least 8.50, at least 9, at least 9.5, at least 10, at least 10.5, or at least 11.
  • the humanized antibody of the disclosure has a pI less than an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4. In some embodiments the humanized antibody of the disclosure has a pI less than the mouse 1H12 antibody. In some embodiments the humanized antibody of the disclosure has a pI of no more than 7.0, such as no more than 6.5, no more than 6.0, no more than 5.5, no more than 5.0, no more than 4.5, or no more than 4.0.
  • the humanized antibody of the disclosure has reduced immunogenicity in a human subject as compared to a non-humanized antibody of the same specificity (for example, a mouse antibody precursor prior to humanization.
  • the humanized antibody has immunogenicity in a human subject lower than an otherwise identical antibody or antibody fragment comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4.
  • the humanized antibody has immunogenicity in a human subject lower than the ‘mouse 1H12’ antibody.
  • Low or reduced immunogenicity can be characterized by the ability to treat patients for extended periods with measurable alleviation of symptoms and low and/or acceptable toxicity.
  • Low or acceptable immunogenicity and/or high affinity, as well as other suitable properties, can contribute to the therapeutic results achieved.
  • “Reduced immunogenicity” is defined herein as raising significant HAHA, HACA or HAMA responses in less 90%, such as less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10% of the proportion of patients who show a significant HAHA, HACA or HAMA response when treated with the mouse 1H12 antibody.
  • the disclosure also provided the means produce the antibodies of the disclosure.
  • the disclosure provides nucleic acid molecules encoding the humanised antibodies, along with nucleic acid molecules complementary to nucleic acid molecules encoding the humanised antibodies.
  • the disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising an antibody pf the disclosure, optionally further comprising a pharmaceutically acceptable carrier or excipient.
  • the disclosure provides a vector, such as an expression vector, comprising a nucleic acid of the disclosure.
  • the disclosure provides host cells transfected with a vector of the disclosure.
  • the host cells may be prokaryotic or eukaryotic.
  • the cells may be bacterial, fungal, insect, or mammalian (such as mouse, primate or human).
  • the disclosure provides a method of making the antibodies by culturing the host cells of the disclosure.
  • the disclosure provides methods relating to the identification of subjects particularly suitable for treatment with the antibodies or pharmaceutical composition of the disclosure. Also provided are methods for determining the optimum timing and dosage of administration of the antibodies of the disclosure to a subject.
  • the subject has a proliferative disease, such as cancer.
  • the subject has an autoimmune disease.
  • administration of the treatment inhibits or reduces one or more aspects of the disease, for example reduces tumour volume, or reduces the level of one or more biomarkers of tumour progression, such as AXL, Akt3, or GAS6.
  • the level of the biomarker is reduced to no more than 90% of the level immediately before treatment, such as no more than 80%, no more than 70%, no more than 60%, no more than 50%, no more than 40%, no more than 30%, no more than 20%, no more than 10%, or no more than 5% of the level immediately before treatment.
  • the disclosure provides a method of selecting a subject for treatment with the antibody or pharmaceutical composition of the disclosure, the method comprising assessing the level of one or more biomarkers associated with disease pathology, wherein subjects having the one or more biomarker, or subjects having a level of the one or more biomarkers which exceeds a threshold level, are selected for treatment.
  • the biomarker is AXL, Akt3, or GAS8.
  • the threshold is at least 10% higher than the upper boundary of the normal clinical range, such as at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 100% higher, or at least 200% higher.
  • the disclosure provides a method of timing the administration of treatment of a subject with the antibody or pharmaceutical composition of the disclosure, the method comprising assessing the level of one or more biomarkers associated with disease pathology, wherein the treatment is administered when the subject has the one or more biomarker, or the subject has a level of one or more biomarkers which exceeds a threshold level.
  • the biomarker is AXL, Akt3, or GAS6.
  • the threshold is at least 10% higher than the upper boundary of the normal clinical range, such as at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 100% higher, or at least 200% higher.
  • the disclosure provides a method of determining the optimum dosage of the antibody or pharmaceutical composition of the disclosure for administration to a subject, the method comprising assessing the level of one or more biomarkers associated with disease pathology, wherein subjects having the one or more biomarker, or subjects having a level of the one or more biomarkers which exceeds the threshold level, are selected for a particular dosage level.
  • the biomarker is AXL, Akt3, or GAS6.
  • the threshold is at least 10% higher than the upper boundary of the normal clinical range, such as at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 100% higher, or at least 200% higher.
  • the level of one or more biomarkers is assessed in a sample of blood, urine, other body fluid, or tissue.
  • Level of one or more biomarkers samples can be assessed by immunoassay, proteomic assay, nucleic acid hybridization or amplification assays, immunohistochemistry, or in situ hybridization assays.
  • antibody encompasses any molecule comprising an antibody antigen-binding site (as, for example, formed by a paired VH domain and a VL domain).
  • antibody encompasses monoclonal antibodies (including intact monoclonal antibodies), polyclonal antibodies, multispecific antibodies formed from at least two different epitope binding fragments (e.g., bispecific antibodies), human antibodies, humanized antibodies, camelised antibodies, chimeric antibodies, single-chain antibodies (such as scFv fusions with CH3), antibody fragments that exhibit the desired biological activity (e.g.
  • the antigen binding portion for example minibodies
  • anti-idiotypic (anti-Id) antibodies intrabodies, and epitope-binding fragments of any of the above, so long as they exhibit the desired biological activity, for example, the ability to bind the cognate antigen.
  • Antibodies may be murine, human, humanized, chimeric, or derived from other species.
  • the antibody is a single-chain Fv antibody fused to a CH3 domain (scFv-CH3).
  • the antibody is a single-chain Fv antibody fused to a Fc region (scFv-Fc).
  • the antibody is a minibody.
  • An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen. (Janeway, C., Travers, P., Walport, M., Shlomchik (2001) Immuno Biology, 5th Ed., Garland Publishing, New York).
  • a target antigen generally has numerous binding sites, also called epitopes, recognized by CDRs on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody.
  • An antibody includes an intact immunoglobulin molecule or an immunologically active portion of a intact immunoglobulin molecule, i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease.
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain at least one antigen binding site.
  • the antibody can be of any isotype (e.g. IgG, IgE, IgM, IgD, and IgA), class (e.g. IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass, or allotype (e.g.
  • human G1m1, G1m2, G1m3, non-G1m1 [that, is any allotype other than G1m1], G1m17, G2m23, G3m21, G3m28, G3m11, G3m5, G3m13, G3m14, G3m10, G3m15, G3m16, G3m6, G3m24, G3m26, G3m27, A2m1, A2m2, Km1, Km2 and Km3) of antibody molecule.
  • the immunoglobulins can be derived from any species, including human, murine, or rabbit origin.
  • an “intact antibody” herein is one comprising VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1, CH2 and CH3.
  • the constant domains may be native sequence constant domains (e.g. human native sequence constant domains) or amino acid sequence variant thereof.
  • the intact antibody may have one or more “effector functions” which refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.
  • the antibody is an intact IgG antibody. That is an antibody comprising two light chains, each having a variable and constant domain, and two heavy chains, each having one variable domain and three constant domains.
  • humanized antibodies include any combination of the herein described Anti-AXL antibodies.
  • the mouse framework residues from the murine 1H12 antibody have been largely replaced with the corresponding residues from human immunoglobulins.
  • critical human residues may be modified as necessary to support the antigen binding site formed by the CDRs and recapitulate the antigen binding potency of the original mouse antibody.
  • Such changes or variations optionally and preferably retain or reduce the immunogenicity in humans or other primate species relative to non-modified antibodies.
  • a humanized antibody can be produced by a non-human animal or prokaryotic or eukaryotic cell that is capable of expressing functionally rearranged human immunoglobulin (e.g., heavy chain and/or light chain) genes.
  • the antibody when it is a single chain antibody, it can comprise a linker peptide that is not found in native human antibodies.
  • an Fv can comprise a linker peptide, such as two to about eight glycine or other amino acid residues, which connects the variable region of the heavy chain and the variable region of the light chain.
  • linker peptides are considered to be of human origin.
  • the humanised antibody of the disclosure are produced by a method comprising he step of grafting the CDRs of the mouse 1H12 antibody into human FW regions such as AB021508, AB063892, AF233253, and AJ399878.
  • the method of producing the humanised antibodies of the invention further comprises the step of back-mutating mismatches at vernier and 5 ⁇ CDR envelope residues.
  • the method of producing the humanised antibodies of the invention further comprises the step of back-mutating mismatched vernier residues only.
  • the human constant region of the humanized antibody can be of any class (IgG, IgA, IgM, IgE, IgD, etc.) or isotype and can comprise a kappa or lambda light chain.
  • the human constant region comprises an IgG heavy chain or defined fragment, for example, at least one of isotypes, IgG1, IgG2, IgG3 or IgG4.
  • the humanized antibody comprises an IgG1 heavy chain and a IgG1 K light chain.
  • the isolated humanized antibodies described herein comprise antibody amino acid sequences disclosed herein encoded by any suitable polynucleotide.
  • sequences of the antibody heavy chain variable regions and/or the light chain variable regions disclosed herein may be modified by substitution, insertion or deletion.
  • Amino acid sequences that are substantially the same as the sequences described herein include sequences comprising conservative amino acid substitutions, as well as amino acid deletions and/or insertions.
  • a conservative amino acid substitution refers to the replacement of a first amino acid by a second amino acid that has chemical and/or physical properties (e.g., charge, structure, polarity, hydrophobicity/hydrophilicity) that are similar to those of the first amino acid.
  • Preferred conservative substitutions are those wherein one amino acid is substituted for another within the groups of amino acids indicated herein below:
  • Particular preferred conservative amino acids substitution groups are: Val-Leu-Ile, Phe-Tyr, Lys-Arg, Ala-Val, and Asn-Gin.
  • the antibody of the conjugates described herein comprises a heavy chain having an amino acid sequence with 80% or more amino acid sequence identity (for example, about 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more sequence identity) to a heavy chain described herein.
  • amino acid sequence identity for example, about 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more sequence identity
  • the antibody of the conjugates described herein comprises a light chain having an amino acid sequence with 80% or more amino acid sequence identity (for example, about 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more sequence identity) to a light chain described herein.
  • amino acid sequence identity for example, about 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more sequence identity
  • the antibody of the conjugates described herein comprises a heavy chain having an amino acid sequence identical to the amino acid sequence of a heavy chain described herein, except that it includes 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid modifications (e.g., substitutions, insertions and/or deletions) relative to the amino acid sequence of the heavy chain described herein.
  • the antibody of the conjugates described herein comprises a light chain having an amino acid sequence identical to the amino acid sequence of a light chain described herein, except that it includes 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid modifications (e.g., substitutions, insertions and/or deletions) relative to the amino acid sequence of the light chain described herein.
  • Humanized antibodies, fragments and regions can be produced by cloning DNA segments encoding the H and L chain antigen-binding regions of the anti-AXL antibody, and joining these DNA segments to DNA segments including CH and CL regions, respectively, to produce full length immunoglobulin-encoding genes.
  • the immunoglobulin cDNAs can be obtained from mRNA of hybridoma cell lines. Antibody heavy and light chains are cloned in a mammalian expression vector system. Assembly is documented with DNA sequence analysis.
  • the antibody construct can be expressed in human or other mammalian host cell lines. The construct can be validated by transient transfection assays and immunoassay of the expressed antibody. Stable cell lines with the highest productivity can be isolated and screened using rapid assay methods.
  • the cytotoxic or cytostatic activity of an antibody is measured by: exposing mammalian cells having receptor proteins to the antibody in a cell culture medium; culturing the cells for a period from about 6 hours to about 5 to 7 days; and measuring cell viability.
  • Cell-based in vitro assays are used to measure viability (proliferation), cytotoxicity, and induction of apoptosis (caspase activation) of an antibody of the disclosure.
  • the in vitro potency of antibodies can be measured by a cell proliferation assay.
  • the CellTiter-Glo® Luminescent Cell Viability Assay is a commercially available (Promega Corp., Madison, Wis.), homogeneous assay method based on the recombinant expression of Coleoptera luciferase (U.S. Pat. Nos. 5,583,024; 5,674,713 and 5,700,670).
  • This cell proliferation assay determines the number of viable cells in culture based on quantitation of the ATP present, an indicator of metabolically active cells (Crouch et al (1993) J. Immunol. Meth. 160:81-88; U.S. Pat. No. 6,602,677).
  • the CellTiter-Glo® Assay is conducted in 96 well format, making it amenable to automated high-throughput screening (HTS) (Cree et al (1995) AntiCancer Drugs 6:398-404).
  • the homogeneous assay procedure involves adding the single reagent (CellTiter-Glo® Reagent) directly to cells cultured in serum-supplemented medium. Cell washing, removal of medium and multiple pipetting steps are not required.
  • the system detects as few as 15 cells/well in a 384-well format in 10 minutes after adding reagent and mixing.
  • the cells may be treated continuously with antibody, or they may be treated and separated from antibody. Generally, cells treated briefly, i.e. 3 hours, showed the same potency effects as continuously treated cells.
  • the homogeneous “add-mix-measure” format results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present.
  • the amount of ATP is directly proportional to the number of cells present in culture.
  • the CellTiter-Glo® Assay generates a “glow-type” luminescent signal, produced by the luciferase reaction, which has a half-life generally greater than five hours, depending on cell type and medium used. Viable cells are reflected in relative luminescence units (RLU).
  • the substrate, Beetle Luciferin is oxidatively decarboxylated by recombinant firefly luciferase with concomitant conversion of ATP to AMP and generation of photons.
  • the in vitro potency of antibody-drug conjugates can also be measured by a cytotoxicity assay.
  • Cultured adherent cells are washed with PBS, detached with trypsin, diluted in complete medium, containing 10% FCS, centrifuged, re-suspended in fresh medium and counted with a haemocytometer. Suspension cultures are counted directly. Monodisperse cell suspensions suitable for counting may require agitation of the suspension by repeated aspiration to break up cell clumps.
  • the cell suspension is diluted to the desired seeding density and dispensed (100 ⁇ l per well) into black 96 well plates. Plates of adherent cell lines are incubated overnight to allow adherence. Suspension cell cultures can be used on the day of seeding.
  • a stock solution (1 ml) of antibody (20 ⁇ g/ml) is made in the appropriate cell culture medium.
  • Serial 10-fold dilutions of stock antibody are made in 15 ml centrifuge tubes by serially transferring 100 ⁇ l to 900 ⁇ l of cell culture medium.
  • AlamarBlue (Invitrogen) is dispensed over the whole plate (20 ⁇ l per well) and incubated for 4 hours. Alamar blue fluorescence is measured at excitation 570 nm, emission 585 nm on the Varioskan flash plate reader. Percentage cell survival is calculated from the mean fluorescence in the antibody treated wells compared to the mean fluorescence in the control wells.
  • the antibody of the disclosure may be used to target a target location.
  • the target location is preferably a proliferative cell population.
  • the antibody is an antibody for an antigen present on a proliferative cell population.
  • the antigen is absent or present at a reduced level in a non-proliferative cell population compared to the amount of antigen present in the proliferative cell population, for example a tumour cell population.
  • the target location may be in vitro, in vivo or ex vivo.
  • the antibodies of the disclosure include those with utility for anticancer activity.
  • the present disclosure provides an antibody as described herein for use in therapy.
  • an antibody as described herein for use in the treatment of a proliferative disease.
  • a second aspect of the present disclosure provides the use of an antibody in the manufacture of a medicament for treating a proliferative disease.
  • proliferative disease pertains to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or hyperplastic growth, whether in vitro or in vivo.
  • proliferative conditions include, but are not limited to, benign, pre-malignant, and malignant cellular proliferation, including but not limited to, neoplasms and tumours (e.g. histocytoma, glioma, astrocyoma, osteoma), cancers (e.g.
  • lung cancer small cell lung cancer, gastrointestinal cancer, bowel cancer, colon cancer, breast carinoma, ovarian carcinoma, prostate cancer, testicular cancer, liver cancer, kidney cancer, bladder cancer, pancreas cancer, brain cancer, sarcoma, osteosarcoma, Kaposi's sarcoma, melanoma), lymphomas, leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g. of connective tissues), and atherosclerosis.
  • Cancers of particular interest include, but are not limited to, metastatic cancer cells, such as circulating tumour cells, which may be found circulating in body fluids such as blood or lymph, lymphomas (e.g., non-Hodgkin's lymphoma, NHL), leukemia (particularly acute myeloid leukemia, AML) and ovarian cancers.
  • lymphomas e.g., non-Hodgkin's lymphoma, NHL
  • leukemia particularly acute myeloid leukemia, AML
  • ovarian cancers e.g., ovarian cancers.
  • Any type of cell may be treated, including but not limited to, lung, gastrointestinal (including, e.g. bowel, colon), breast (mammary), ovarian, prostate, liver (hepatic), kidney (renal), bladder, pancreas, brain, and skin.
  • gastrointestinal including, e.g. bowel, colon
  • breast mammary
  • ovarian prostate
  • liver hepatic
  • kidney renal
  • bladder pancreas
  • brain and skin.
  • the antibody of the present disclosure may be used to treat various diseases or disorders, e.g. characterized by the overexpression of a tumour antigen.
  • exemplary conditions or hyperproliferative disorders include benign or malignant tumours; leukemias, haematological, and lymphoid malignancies.
  • Others include neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic and immunologic, including autoimmune, disorders.
  • the disease or disorder to be treated is a hyperproliferative disease such as cancer.
  • cancer to be treated herein include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemias or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer,
  • Autoimmune diseases for which the antibody may be used in treatment include rheumatologic disorders (such as, for example, rheumatoid arthritis, Sjögren's syndrome, scleroderma, lupus such as SLE and lupus nephritis, polymyositis/dermatomyositis, cryoglobulinemia, anti-phospholipid antibody syndrome, and psoriatic arthritis), osteoarthritis, autoimmune gastrointestinal and liver disorders (such as, for example, inflammatory bowel diseases (e.g.
  • autoimmune gastritis and pernicious anemia such as, for example, ANCA-associated vasculitis, including Churg-Strauss vasculitis, Wegener's granulomatosis, and polyarteriitis
  • vasculitis such as, for example, ANCA-associated vasculitis, including Churg-Strauss vasculitis, Wegener's granulomatosis, and polyarteriitis
  • autoimmune neurological disorders such as, for example, multiple sclerosis, opsocionus myocionus syndrome, myasthenia gravis, neuromyelitis optica, Parkinson's disease, Alzheimer's disease, and autoimmune polyneuropathies
  • renal disorders such as, for example, glomerulonephritis, Goodpasture's syndrome, and Berger's disease
  • autoimmune dermatologic disorders such as, for example, psoriasis, urticaria, hives, pemphigus vulgaris, bullous pemphigoid, and
  • Graves' disease and thyroiditis More preferred such diseases include, for example, rheumatoid arthritis, ulcerative colitis, ANCA-associated vasculitis, lupus, multiple sclerosis, Sjögren's syndrome, Graves' disease, IDDM, pernicious anemia, thyroiditis, and glomerulonephritis.
  • the antibody of the present disclosure may be used in a method of therapy. Also provided is a method of treatment, comprising administering to a subject in need of treatment a therapeutically-effective amount of an antibody of the disclosure.
  • a therapeutically-effective amount is an amount sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom.
  • the actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage, is within the responsibility of general practitioners and other medical doctors.
  • An antibody may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
  • treatments and therapies include, but are not limited to, chemotherapy (the administration of active agents, including, e.g. drugs, such as chemotherapeutics); surgery; and radiation therapy.
  • A“chemotherapeutic agent” is a chemical compound useful in the treatment of cancer, regardless of mechanism of action.
  • Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, antibodies, photosensitizers, and kinase inhibitors.
  • Chemotherapeutic agents include compounds used in “targeted therapy” and conventional chemotherapy.
  • chemotherapeutic agents include: erlotinib (TARCEVA®, Genentech/OSI Pharm.), docetaxel (TAXOTERE®, Sanofi-Aventis), 5-FU (fluorouracil, 5-fluorouracil, CAS No. 51-21-8), gemcitabine (GEMZAR®, Lilly), PD-0325901 (CAS No. 391210-10-9, Pfizer), cisplatin (cis-diamine, dichloroplatinum(II), CAS No. 15663-27-1), carboplatin (CAS No.
  • paclitaxel TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.J.
  • trastuzumab HERCEPTIN®, Genentech
  • temozolomide 4-methyl-5-oxo-2,3,4,6,8-pentazabicyclo [4.3.0] nona-2,7,9-triene-9-carboxamide, CAS No.
  • tamoxifen (Z)-2-[4-(1,2-diphenylbut-1-enyl)phenoxy]-N,N-dimethylethanamine, NOLVADEX®, ISTUBAL®, VALODEX®), and doxorubicin (ADRIAMYCIN®), Akti-1/2, HPPD, and rapamycin.
  • chemotherapeutic agents include: oxaliplatin (ELOXATIN®, Sanofi), bortezomib (VELCADE®, Millennium Pharm.), sutent (SUNITINIB®, SU11248, Pfizer), letrozole (FEMARA®, Novartis), imatinib mesylate (GLEEVEC®, Novartis), XL-518 (Mek inhibitor, Exelixis, WO 2007/044515), ARRY-886 (Mek inhibitor, AZD6244, Array BioPharma, Astra Zeneca), SF-1126 (PI3K inhibitor, Semafore Pharmaceuticals), BEZ-235 (PI3K inhibitor, Novartis), XL-147 (PI3K inhibitor, Exelixis), PTK787/ZK 222584 (Novartis), fulvestrant (FASLODEX®, AstraZeneca), leucovorin (folinic acid), rapamycin (siroli
  • calicheamicin calicheamicin gamma1I, calicheamicin omegal1 ( Angew Chem. Intl. Ed. Engl . (1994) 33:183-186); dynemicin, dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-nordeucine, morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-dox
  • chemotherapeutic agent include: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumours such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA® (letrozole),
  • SERMs
  • chemotherapeutic agent therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen Idec), ofatumumab (ARZERRA®, GSK), pertuzumab (PERJETATM, OMNITARGTM, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
  • therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panit
  • Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the conjugates of the disclosure include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab,
  • compositions according to the present disclosure may comprise, in addition to the active ingredient, i.e. an antibody, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • a pharmaceutically acceptable excipient e.g. an antibody
  • carrier e.g. a pharmaceutically acceptable sulfate, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art.
  • Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, e.g. cutaneous, subcutaneous, or intravenous.
  • compositions for oral administration may be in tablet, capsule, powder or liquid form.
  • a tablet may comprise a solid carrier or an adjuvant.
  • Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • a capsule may comprise a solid carrier such a gelatin.
  • the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringers Injection.
  • Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
  • the antibody While it is possible for the antibody to be used (e.g., administered) alone, it is often preferable to present it as a composition or formulation.
  • the composition is a pharmaceutical composition (e.g., formulation, preparation, medicament) comprising an antibody, as described herein, and a pharmaceutically acceptable carrier, diluent, or excipient.
  • a pharmaceutical composition e.g., formulation, preparation, medicament
  • a pharmaceutically acceptable carrier e.g., diluent, or excipient.
  • the composition is a pharmaceutical composition comprising at least one antibody, as described herein, together with one or more other pharmaceutically acceptable ingredients well known to those skilled in the art, including, but not limited to, pharmaceutically acceptable carriers, diluents, excipients, adjuvants, fillers, buffers, preservatives, anti-oxidants, lubricants, stabilisers, solubilisers, surfactants (e.g., wetting agents), masking agents, colouring agents, flavouring agents, and sweetening agents.
  • pharmaceutically acceptable carriers diluents, excipients, adjuvants, fillers, buffers, preservatives, anti-oxidants, lubricants, stabilisers, solubilisers, surfactants (e.g., wetting agents), masking agents, colouring agents, flavouring agents, and sweetening agents.
  • the composition further comprises other active agents, for example, other therapeutic or prophylactic agents.
  • Suitable carriers, diluents, excipients, etc. can be found in standard pharmaceutical texts. See, for example, Handbook of Pharmaceutical Additives, 2nd Edition (eds. M. Ash and I. Ash), 2001 (Synapse Information Resources, Inc., Endicott, N.Y., USA), Remington's Pharmaceutical Sciences, 20th edition, pub. Lippincott, Williams & Wilkins, 2000; and Handbook of Pharmaceutical Excipients, 2nd edition, 1994.
  • Another aspect of the present disclosure pertains to methods of making a pharmaceutical composition
  • a pharmaceutical composition comprising admixing at least one [ 11 C]-radiolabelled antibody or antibody-like molecule, as defined herein, together with one or more other pharmaceutically acceptable ingredients well known to those skilled in the art, e.g., carriers, diluents, excipients, etc. If formulated as discrete units (e.g., tablets, etc.), each unit contains a predetermined amount (dosage) of the active compound.
  • pharmaceutically acceptable pertains to compounds, ingredients, materials, compositions, dosage forms, etc., which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of the subject in question (e.g., human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • Each carrier, diluent, excipient, etc. must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
  • the formulations may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with a carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active compound with carriers (e.g., liquid carriers, finely divided solid carrier, etc.), and then shaping the product, if necessary.
  • carriers e.g., liquid carriers, finely divided solid carrier, etc.
  • the formulation may be prepared to provide for rapid or slow release; immediate, delayed, timed, or sustained release; or a combination thereof.
  • Formulations suitable for parenteral administration include aqueous or non-aqueous, isotonic, pyrogen-free, sterile liquids (e.g., solutions, suspensions), in which the active ingredient is dissolved, suspended, or otherwise provided (e.g., in a liposome or other microparticulate).
  • Such liquids may additional contain other pharmaceutically acceptable ingredients, such as anti-oxidants, buffers, preservatives, stabilisers, bacteriostats, suspending agents, thickening agents, and solutes which render the formulation isotonic with the blood (or other relevant bodily fluid) of the intended recipient.
  • excipients include, for example, water, alcohols, polyols, glycerol, vegetable oils, and the like.
  • suitable isotonic carriers for use in such formulations include Sodium Chloride Injection, Ringers Solution, or Lactated Ringers Injection.
  • the concentration of the active ingredient in the liquid is from about 1 ng/ml to about 10 ⁇ g/ml, for example from about 10 ng/ml to about 1 ⁇ g/ml.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.
  • appropriate dosages of the antibody, and compositions comprising the antibody can vary from patient to patient. Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects.
  • the selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds, and/or materials used in combination, the severity of the condition, and the species, sex, age, weight, condition, general health, and prior medical history of the patient.
  • the amount of compound and route of administration will ultimately be at the discretion of the physician, veterinarian, or clinician, although generally the dosage will be selected to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects.
  • Administration can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell(s) being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician, veterinarian, or clinician.
  • a suitable dose of the antibody is in the range of about 100 ng to about 25 mg (more typically about 1 ⁇ g to about 10 mg) per kilogram body weight of the subject per day.
  • the active compound is a salt, an ester, an amide, a prodrug, or the like
  • the amount administered is calculated on the basis of the parent compound and so the actual weight to be used is increased proportionately.
  • the antibody is administered to a human patient according to the following dosage regime: about 100 mg, 3 times daily.
  • the antibody is administered to a human patient according to the following dosage regime: about 150 mg, 2 times daily.
  • the antibody is administered to a human patient according to the following dosage regime: about 200 mg, 2 times daily.
  • the antibody is administered to a human patient according to the following dosage regime: about 50 or about 75 mg, 3 or 4 times daily.
  • the antibody is administered to a human patient according to the following dosage regime: about 100 or about 125 mg, 2 times daily.
  • an antibody of the disclosure will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the molecule is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the molecule is suitably administered to the patient at one time or over a series of treatments.
  • about 1 ⁇ g/kg to 15 mg/kg (e.g. 0.1-20 mg/kg) of molecule is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • a typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • An exemplary dosage of antibody to be administered to a patient is in the range of about 0.1 to about 10 mg/kg of patient weight.
  • An exemplary dosing regimen comprises a course of administering an initial loading dose of about 4 mg/kg, followed by additional doses every week, two weeks, or three weeks of an antibody. Other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • treatment pertains generally to treatment and therapy, whether of a human or an animal (e.g., in veterinary applications), in which some desired therapeutic effect is achieved, for example, the inhibition of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, regression of the condition, amelioration of the condition, and cure of the condition.
  • Treatment as a prophylactic measure i.e., prophylaxis, prevention is also included.
  • terapéuticaally-effective amount pertains to that amount of an antibody, or a material, composition or dosage from comprising an antibody, which is effective for producing some desired therapeutic effect, commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.
  • prophylactically-effective amount refers to that amount of an antibody, or a material, composition or dosage from comprising an antibody, which is effective for producing some desired prophylactic effect, commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.
  • the subject/patient may be an animal, mammal, a placental mammal, a marsupial (e.g., kangaroo, wombat), a monotreme (e.g., duckbilled platypus), a rodent (e.g., a guinea pig, a hamster, a rat, a mouse), murine (e.g., a mouse), a lagomorph (e.g., a rabbit), avian (e.g., a bird), canine (e.g., a dog), feline (e.g., a cat), equine (e.g., a horse), porcine (e.g., a pig), ovine (e.g., a sheep), bovine (e.g., a cow), a primate, simian (e.g., a monkey or ape), a monkey (e.g., marmoset, baboon), an
  • the subject/patient may be any of its forms of development, for example, a foetus.
  • the subject/patient is a human.
  • FIG. 1 AXL binding ELISA of fully humanised constructs. (2A) to Human AXL. (2B) to Cynomolgus monkey AXL.
  • FIG. 2 Accelerated stability analysis
  • Protocol 1 Production of DNA Plasmids for Expression
  • Transformed bacteria were spread on LB-agar Zeocin/Blasticidin plates, as required, incubated overnight at 37 C, then colonies were picked from each plate.
  • VH or VK colonies/glycerol stocks were inoculated into 3 ml LB containing Zeocin 25 ⁇ g/ml or Blasticidin 50 ⁇ g/ml, respectively.
  • p21, p27, pAdvantage (Promega) and pSVLT were inoculated in LB-Ampicillin and shaken overnight.
  • check cells are at least 80% confluent and replace the medium (2 ml/well).
  • Protocol 3 IgG Quantitation by ELISA
  • Protocol 4 AXL Binding ELISA
  • Protocol 4A SPR Measurement of Antibody Affinity
  • Protocol 5 Capillary Isoelectric Focusing
  • Protocol 6 Protein Thermal Shift Protocol
  • Protein thermal shift dye (2.5 ⁇ L 1:1000 dilution) was added to sample proteins (17.5 ⁇ L of 0.5 mg/mL in PBS) in a 96 well optical plate and mixed thoroughly. Every sample was done in quadruplicate. The plate was sealed with a optical adhesive film and bubbles in the wells were removed by centrifugation 1 min at 500 g, then placed on ice. The sealed plate was introduced in the 7500 Real-Time PCR System and subsequently the experiment was set up as follows:
  • Time (minutes) % B 0 22.5 1 22.5 11 50 11.5 90 13.5 90 14.5 22.5 16 22.5
  • a stability assay was performed during which the antibodies were heated in sterile PBS at 40° C. for 60 hr and analysed for aggregation by SEC and for binding activity by human AXL ELISA. No increase in aggregation or loss in AXL binding activity was detected for any of the antibodies (see FIG. 2 ).
  • An isolated humanized antibody that binds to AXL wherein the isolated humanized antibody comprises a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3.
  • the isolated humanized antibody according to statement 1 wherein the isolated humanized antibody further comprises a light chain variable region having the amino acid sequence of SEQ ID NO: 4, 5, 6, 7, or 8; and, optionally, comprises a constant region derived from one or more human antibodies.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present disclosure relates to humanized anti-Axl antibodies.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 15/566,635, filed Oct. 13, 2017, which is a national phase application under 35 U.S.C. § 371 of PCT International Application No. PCT/EP2016/058368, filed Apr. 15, 2016, which claims priority to Great Britain Application No. 1506411.6, filed Apr. 15, 2015, which are hereby incorporated by reference in its entirety.
  • INCORPORATION-BY-REFERENCE OF MATERIAL SUBMITTED ELECTRONICALLY
  • Incorporated by reference in its entirety herein is a computer-readable nucleotide/amino acid sequence listing submitted concurrently herewith and identified as follows: One 53,106 Byte ASCII (Text) file named “35710-302-SQL_ST25.TXT,” created on Nov. 24, 2021.
  • The present disclosure relates to humanized anti-Axl antibodies.
  • BACKGROUND
  • Axl
  • Axl is a member of the TAM (Tyro3-Axl-Mer) receptor tyrosine kinases (RTK) that share the vitamin K-dependent ligand Gas6 (growth arrest-specific 6). TAM family RTKs regulate a diverse range of cellular responses including cell survival, proliferation, autophagy, migration, angiogenesis, platelet aggregation, and natural killer cell differentiation. Axl is expressed in many embryonic tissues and is thought to be involved in mesenchymal and neural development, with expression in adult tissues largely restricted to smooth muscle cells (MGI Gene Expression Database; www.informatics.jax.org). Axl activation is linked to several signal transduction pathways, including Aid, MAP kinases, NF-κB, STAT, and others. Originally identified as a transforming gene from a patient with chronic myelogenous leukaemia, Axl has since been associated with various high-grade cancers and correlated with poor prognosis.
  • Axl receptor overexpression has been detected in a wide range of solid tumours and myeloid leukaemia (Linger et al, Adv Cancer Res. 100: 35, 2008; Linger et al, Expert Opin Ther Targets. 14:1073, 2010).
  • Axl expression correlates with malignant progression and is an independent predictor of poor patient overall survival in several malignancies including pancreatic (Song et al, Cancer. 117:734, 2011), prostate (Paccez et al, Oncogene. 32:698, 2013), lung (Ishikawa et al. Ann Surg Oncol. 2012; Zhang et al, Nat Genet. 44:852, 2012), breast (Gjerdrum, Proc natl Acad Sci USA 107:1124, 2010), colon cancer (Yuen et al, PLoS One, 8:e54211, 2013) and acute myeloid leukaemia (AML) (Ben-Batalla et al, Blood 122:2443, 2013).
  • Ax signal transduction is activated by a protein ligand (Gas6) secreted by tumour associated macrophages (Loges et al, Blood. 115:2264, 2010) or autocrine mechanisms (Gjerdrum, Proc natl Acad Sci USA 107:1124, 2010), that drives receptor dimerization, autophosphorylation and downstream signalling, such as via PI3 kinase (PI3K)-AKT, particularly AKT and mitogen-activated protein kinase (MAPK) pathways (Korshunov, Clinical Science. 122:361, 2012). Heterodimerization with other tyrosine kinase receptors, e.g. epidermal growth factor receptor (EGFR), is also reported to occur (Linger et al, Expert Opin Ther Targets. 14:1073, 2010; Meyer et al Science Signalling 6:ra66, 2013).
  • Aberrant activation of Axl in tumour cells is widely associated with acquired drug resistance to targeted therapeutics in vitro and in vivo (Zhang et al. Nat Genet. 44: 852, 2012; Byers et al. Clin Cancer Res. 19: 279, 2013). Axl-targeting agents block tumour formation, metastasis and reverse drug resistance (e.g. to erlotinib) by reversing EMT/CSC characteristics in several experimental cancer models, including triple negative breast cancer, hormone resistant prostate cancer and adenocarcinoma of the lung (Holland et al Cancer Res 70:1544, 2010; Gjerdrum, Proc natl Acad Sci USA 107:1124, 2010; Zhang et al. Nat Genet. 44: 852, 2012; Paccez et al, Oncogene. 32:698, 2013).
  • Anti-Axl Antibodies
  • Applications relating to Ax and anti-Axl antibodies include EP2267454A2 [Diagnosis and prevention of cancer cell invasion measuring . . . Axl—Max Planck]; WO2009063965 [anti Axl—Chugai Pharmaceutical]; WO2011159980A1 [anti-Axl—Genentech], WO2011014457A1 [combination treatments Axl and VEGF antagonists—Genentech]; WO2012-175691A1 [Anti Axl 20G7-D9—INSERM], WO2012-175692A1 [Anti Axl 3E3E8—INSERM]; WO2009/062690A1 [anti Axl—U3 Pharma] and WO2010/130751A1 [humanised anti Axl—U3 Pharma].
  • GB1410826.0 discloses the murine anti-Axl antibody designated herein as “mouse 1H12”. In view of the advantageous properties of this antibody and its potential clinical applications in humans, it is desirable to identify humanised versions of the murine antibody which have reduced immunogenicity to humans. The present disclosure concerns such antibodies, along with antibody-drug conjugates comprising the humanised 1H12 antibodies and PBD drug-moieties.
  • SUMMARY
  • The present disclosure provides humanized anti-AXL antibodies derived from the ‘mouse 1H12’ antibody.
  • The present inventors have generated a number of humanised heavy chain variable regions (SEQ ID NOs: 2 and 3) and humanised light chain variable regions (SEQ ID NOs:5 to 8) with a view to creating antibodies that have lower immunogenicity in a human individual than the ‘mouse 1H12’ antibody or ‘chimeric 1H12’ antibody whilst retaining antigen-binding potency. Surprisingly, these humanised antibodies have also been found to have other advantageous properties, such as increased charge at physiological pH and improved affinity for some Axl ligands.
  • Accordingly, in one aspect the present disclosure comprises an isolated humanized antibody that binds to AXL, wherein the isolated humanized antibody comprises a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1, 2, or 3. In some embodiments the antibody further comprises a light chain variable region having the amino acid sequence of SEQ ID NO: 4, 5, 6, 7, or 8 and, optionally, further comprises a constant region derived from one or more human antibodies.
  • In some embodiments the isolated humanized antibody that binds to AXL comprises; a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 or 3; a light chain variable region having the amino acid sequence of SEQ ID NO: 5, 6, 7, or 8; and, optionally, comprises a constant region derived from one or more human antibodies.
  • In some embodiments, the humanized antibody does not comprise a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4.
  • In some embodiments the isolated humanized antibody that binds to AXL comprises:
  • (i) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
    (ii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
    (iii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
    (iv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7;
    (v) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8;
    (vi) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
    (vii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
    (viii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
    (ix) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7;
    (x) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8;
    (xi) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
    (xii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
    (xiii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
    (xiv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7; or
    (xv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8.
  • In some embodiments AXL is human AXL.
  • The sequences of the antibody heavy chain variable regions and/or the light chain variable regions disclosed herein may be modified by, for example, insertions, substitutions and/or deletions to the extent that the humanized antibody maintains the ability to bind to AXL. The skilled person can ascertain the maintenance of this activity by performing the functional assays described herein, or known in the art.
  • Accordingly, in some embodiments the heavy chain variable region comprises no more than 20 insertions, substitutions and/or deletions, such as no more than 15, no more than 10, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 insertion, substitution and/or deletion. In some embodiments the light chain variable region comprises no more than 20 insertions, substitutions and/or deletions, such as no more than 15, no more than 10, no more than 9, no more than 8, no more than 7, no more than 6, no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 insertion, substitution and/or deletion.
  • In some embodiments the humanized antibodies of the disclosure include antibodies comprising VH and VL domains with amino acid sequences that are identical to the sequences described herein.
  • DETAILED DISCLOSURE
  • Antibody Properties
  • Antigen Binding
  • The antibody of the conjugates described herein is an antibody (Ab) which binds AXL. That is, the conjugates described herein are conjugates comprising antibodies which specifically bind to AXL.
  • As used herein, AXL refers to the Axl member of the TAM family of receptor tyrosine kinases. ‘Human Axl’ refers to the Axl member of the human TAM family of receptor tyrosine kinases. In some embodiments, the human Axl polypeptide corresponds to Genbank accession no. AAH32229, version no. AAH32229.1 G1:21619004, record update date: Mar. 6, 2012 01:18 PM (SEQ ID NO.9). In one embodiment, the nucleic acid encoding the human Axl polypeptide corresponds to Genbank accession no. M76125, version no. M76125.1 G1:292869, record update date: Jun. 23, 2010 08:53 AM. ‘Murine Axl’ refers to the Axl member of the murine TAM family of receptor tyrosine kinases. In some embodiments, the murine Axl polypeptide corresponds to Genbank accession no. AAH46618, version no. AAH46618.1 G1:55777082, record update date: Mar. 6, 2012 01:36 PM (SEQ ID NO.10). In one embodiment, the nucleic acid encoding the murine Axl polypeptide corresponds to Genbank accession no. NM_009465, version no. NM_009465.4 GI:300794836, record update date: Mar. 12, 2014 03:52 PM.
  • Antibody Affinity
  • In some embodiments the humanized antibody binds human AXL with a dissociation constant (KD) of at least 10−6 M, such as at least 5×10−7 M, at least 10−7 M, at least 5×10−8 M, at least 10−9 M, such as at least 5×10−10 M, at least 10−10 M, at least 5×10−11 M, at least 10−11 M, at least 5×10−12 M, at least 10−12 M, at least 5×10−13 M, at least 10−13 M, at least 5×10−14 M, at least 10−14 M, at least 5×10−15 M, or at least 10−15 M.
  • In one embodiment the humanized antibody competitively inhibits the in vivo and/or in vitro binding to human AXL of an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4. In one embodiment the humanized antibody competitively inhibits the in vivo and/or in vitro binding to human-AXL of the ‘mouse 1H12’ antibody. In some embodiments an equimolar dose of the humanised antibody competitively inhibits at least 20% of the binding by the ‘mouse 1H12’ antibody, such as at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of the binding. Percentage binding may be measured by, for example, a competitive ELISA assay where % inhibition of binding is calculated as [(1−absorbance of test sample)/(absorbance of negative control)].
  • In some embodiments the humanized antibody has a higher affinity for an Axl antigen (for example the Axl-Strep-His antigen described in Protocol 4) than an antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies (for example, Abi described herein). In some embodiments the KD of the humanized antibody with the Axl antigen (for example the Axl-Strep-His antigen described in Protocol 4) will be no more than 0.9 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies, for example no more than 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.05, 0.01, or 0.001 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies.
  • In some embodiments the humanized antibody has a higher affinity for an Axl antigen (for example the Axl-Fc antigen described in Protocol 4) than an antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies (for example, Abi described herein). In some embodiments the KD of the humanized antibody with the Axl antigen (for example the Axl-Fc antigen described in Protocol 4) will be no more than 0.9 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies, for example no more than 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.05, 0.01, or 0.001 of the KD of the antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies.
  • Antibody Isoelectric Point (pI)
  • A molecule carries no net charge when the pH of its surrounding equal the molecules pI. The net charge of a molecule affects the solubility of the molecule, with biological molecules such as proteins typically having minimum solubility in water or salt solutions at the pH that corresponds to their pI. Thus, proteins whose pI is 7.35-7.45 are at their minimum solubility in human blood, whose pH is typically in the range 7.35-7.45.
  • In some embodiments the humanized antibody of the disclosure has a pI greater than an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4. In some embodiments the humanized antibody of the disclosure has a pI greater than the mouse 1H12 antibody. In some embodiments the humanized antibody of the disclosure has a pI of at least 8.00, such as at least 8.05, at least 8.10, at least 8.15, at least 8.20, at least 8.30, at least 8.40, at least 8.50, at least 9, at least 9.5, at least 10, at least 10.5, or at least 11.
  • In some embodiments the humanized antibody of the disclosure has a pI less than an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4. In some embodiments the humanized antibody of the disclosure has a pI less than the mouse 1H12 antibody. In some embodiments the humanized antibody of the disclosure has a pI of no more than 7.0, such as no more than 6.5, no more than 6.0, no more than 5.5, no more than 5.0, no more than 4.5, or no more than 4.0.
  • Antibody Immunogenicity
  • Preferably the humanized antibody of the disclosure has reduced immunogenicity in a human subject as compared to a non-humanized antibody of the same specificity (for example, a mouse antibody precursor prior to humanization. In one embodiment the humanized antibody has immunogenicity in a human subject lower than an otherwise identical antibody or antibody fragment comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4. In one embodiment the humanized antibody has immunogenicity in a human subject lower than the ‘mouse 1H12’ antibody.
  • Low or reduced immunogenicity can be characterized by the ability to treat patients for extended periods with measurable alleviation of symptoms and low and/or acceptable toxicity. Low or acceptable immunogenicity and/or high affinity, as well as other suitable properties, can contribute to the therapeutic results achieved. “Reduced immunogenicity” is defined herein as raising significant HAHA, HACA or HAMA responses in less 90%, such as less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10% of the proportion of patients who show a significant HAHA, HACA or HAMA response when treated with the mouse 1H12 antibody.
  • The disclosure also provided the means produce the antibodies of the disclosure.
  • Accordingly, in another aspect the disclosure provides nucleic acid molecules encoding the humanised antibodies, along with nucleic acid molecules complementary to nucleic acid molecules encoding the humanised antibodies.
  • In another aspect, the disclosure provides a pharmaceutical composition comprising an antibody pf the disclosure, optionally further comprising a pharmaceutically acceptable carrier or excipient.
  • In another aspect the disclosure provides a vector, such as an expression vector, comprising a nucleic acid of the disclosure.
  • In another aspect, the disclosure provides host cells transfected with a vector of the disclosure. The host cells may be prokaryotic or eukaryotic. For example, the cells may be bacterial, fungal, insect, or mammalian (such as mouse, primate or human).
  • In another aspect the disclosure provides a method of making the antibodies by culturing the host cells of the disclosure.
  • The disclosure provides methods relating to the identification of subjects particularly suitable for treatment with the antibodies or pharmaceutical composition of the disclosure. Also provided are methods for determining the optimum timing and dosage of administration of the antibodies of the disclosure to a subject. In some embodiments the subject has a proliferative disease, such as cancer. In some embodiments the subject has an autoimmune disease. Preferably, administration of the treatment inhibits or reduces one or more aspects of the disease, for example reduces tumour volume, or reduces the level of one or more biomarkers of tumour progression, such as AXL, Akt3, or GAS6. In some embodiments the level of the biomarker is reduced to no more than 90% of the level immediately before treatment, such as no more than 80%, no more than 70%, no more than 60%, no more than 50%, no more than 40%, no more than 30%, no more than 20%, no more than 10%, or no more than 5% of the level immediately before treatment.
  • In one aspect the disclosure provides a method of selecting a subject for treatment with the antibody or pharmaceutical composition of the disclosure, the method comprising assessing the level of one or more biomarkers associated with disease pathology, wherein subjects having the one or more biomarker, or subjects having a level of the one or more biomarkers which exceeds a threshold level, are selected for treatment. In some embodiments the biomarker is AXL, Akt3, or GAS8. In some embodiments the threshold is at least 10% higher than the upper boundary of the normal clinical range, such as at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 100% higher, or at least 200% higher.
  • In another aspect the disclosure provides a method of timing the administration of treatment of a subject with the antibody or pharmaceutical composition of the disclosure, the method comprising assessing the level of one or more biomarkers associated with disease pathology, wherein the treatment is administered when the subject has the one or more biomarker, or the subject has a level of one or more biomarkers which exceeds a threshold level. In some embodiments the biomarker is AXL, Akt3, or GAS6. In some embodiments the threshold is at least 10% higher than the upper boundary of the normal clinical range, such as at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 100% higher, or at least 200% higher.
  • In another aspect the disclosure provides a method of determining the optimum dosage of the antibody or pharmaceutical composition of the disclosure for administration to a subject, the method comprising assessing the level of one or more biomarkers associated with disease pathology, wherein subjects having the one or more biomarker, or subjects having a level of the one or more biomarkers which exceeds the threshold level, are selected for a particular dosage level. In some embodiments the biomarker is AXL, Akt3, or GAS6. In some embodiments the threshold is at least 10% higher than the upper boundary of the normal clinical range, such as at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 100% higher, or at least 200% higher.
  • In some embodiments the level of one or more biomarkers is assessed in a sample of blood, urine, other body fluid, or tissue. Level of one or more biomarkers samples can be assessed by immunoassay, proteomic assay, nucleic acid hybridization or amplification assays, immunohistochemistry, or in situ hybridization assays.
  • Definitions
  • Antibody
  • The term “antibody” as used encompasses any molecule comprising an antibody antigen-binding site (as, for example, formed by a paired VH domain and a VL domain). Thus, for example, the term “antibody” encompasses monoclonal antibodies (including intact monoclonal antibodies), polyclonal antibodies, multispecific antibodies formed from at least two different epitope binding fragments (e.g., bispecific antibodies), human antibodies, humanized antibodies, camelised antibodies, chimeric antibodies, single-chain antibodies (such as scFv fusions with CH3), antibody fragments that exhibit the desired biological activity (e.g. the antigen binding portion; for example minibodies), and anti-idiotypic (anti-Id) antibodies, intrabodies, and epitope-binding fragments of any of the above, so long as they exhibit the desired biological activity, for example, the ability to bind the cognate antigen. Antibodies may be murine, human, humanized, chimeric, or derived from other species. In one embodiment the antibody is a single-chain Fv antibody fused to a CH3 domain (scFv-CH3). In one embodiment the antibody is a single-chain Fv antibody fused to a Fc region (scFv-Fc). In one embodiment the antibody is a minibody.
  • An antibody is a protein generated by the immune system that is capable of recognizing and binding to a specific antigen. (Janeway, C., Travers, P., Walport, M., Shlomchik (2001) Immuno Biology, 5th Ed., Garland Publishing, New York). A target antigen generally has numerous binding sites, also called epitopes, recognized by CDRs on multiple antibodies. Each antibody that specifically binds to a different epitope has a different structure. Thus, one antigen may have more than one corresponding antibody. An antibody includes an intact immunoglobulin molecule or an immunologically active portion of a intact immunoglobulin molecule, i.e., a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof, such targets including but not limited to, cancer cell or cells that produce autoimmune antibodies associated with an autoimmune disease.
  • In particular, antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain at least one antigen binding site. The antibody can be of any isotype (e.g. IgG, IgE, IgM, IgD, and IgA), class (e.g. IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass, or allotype (e.g. human G1m1, G1m2, G1m3, non-G1m1 [that, is any allotype other than G1m1], G1m17, G2m23, G3m21, G3m28, G3m11, G3m5, G3m13, G3m14, G3m10, G3m15, G3m16, G3m6, G3m24, G3m26, G3m27, A2m1, A2m2, Km1, Km2 and Km3) of antibody molecule. The immunoglobulins can be derived from any species, including human, murine, or rabbit origin.
  • An “intact antibody” herein is one comprising VL and VH domains, as well as a light chain constant domain (CL) and heavy chain constant domains, CH1, CH2 and CH3. The constant domains may be native sequence constant domains (e.g. human native sequence constant domains) or amino acid sequence variant thereof. The intact antibody may have one or more “effector functions” which refer to those biological activities attributable to the Fc region (a native sequence Fc region or amino acid sequence variant Fc region) of an antibody. Examples of antibody effector functions include C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell surface receptors such as B cell receptor and BCR.
  • In preferred embodiments the antibody is an intact IgG antibody. That is an antibody comprising two light chains, each having a variable and constant domain, and two heavy chains, each having one variable domain and three constant domains.
  • Humanized
  • As used herein “humanized” antibodies include any combination of the herein described Anti-AXL antibodies. In these antibodies the mouse framework residues from the murine 1H12 antibody have been largely replaced with the corresponding residues from human immunoglobulins. As many of the human amino acid residues as possible are retained, but critical human residues may be modified as necessary to support the antigen binding site formed by the CDRs and recapitulate the antigen binding potency of the original mouse antibody. Such changes or variations optionally and preferably retain or reduce the immunogenicity in humans or other primate species relative to non-modified antibodies.
  • It is pointed out that a humanized antibody can be produced by a non-human animal or prokaryotic or eukaryotic cell that is capable of expressing functionally rearranged human immunoglobulin (e.g., heavy chain and/or light chain) genes. Further, when the antibody is a single chain antibody, it can comprise a linker peptide that is not found in native human antibodies. For example, an Fv can comprise a linker peptide, such as two to about eight glycine or other amino acid residues, which connects the variable region of the heavy chain and the variable region of the light chain. Such linker peptides are considered to be of human origin.
  • For example, in some embodiments the humanised antibody of the disclosure are produced by a method comprising he step of grafting the CDRs of the mouse 1H12 antibody into human FW regions such as AB021508, AB063892, AF233253, and AJ399878. In some embodiments the method of producing the humanised antibodies of the invention further comprises the step of back-mutating mismatches at vernier and 5 Å CDR envelope residues. In other embodiments the method of producing the humanised antibodies of the invention further comprises the step of back-mutating mismatched vernier residues only.
  • The human constant region of the humanized antibody can be of any class (IgG, IgA, IgM, IgE, IgD, etc.) or isotype and can comprise a kappa or lambda light chain. In one embodiment, the human constant region comprises an IgG heavy chain or defined fragment, for example, at least one of isotypes, IgG1, IgG2, IgG3 or IgG4. In another embodiment, the humanized antibody comprises an IgG1 heavy chain and a IgG1 K light chain. The isolated humanized antibodies described herein comprise antibody amino acid sequences disclosed herein encoded by any suitable polynucleotide.
  • Sequence Modifications
  • The sequences of the antibody heavy chain variable regions and/or the light chain variable regions disclosed herein may be modified by substitution, insertion or deletion. Amino acid sequences that are substantially the same as the sequences described herein include sequences comprising conservative amino acid substitutions, as well as amino acid deletions and/or insertions. A conservative amino acid substitution refers to the replacement of a first amino acid by a second amino acid that has chemical and/or physical properties (e.g., charge, structure, polarity, hydrophobicity/hydrophilicity) that are similar to those of the first amino acid. Preferred conservative substitutions are those wherein one amino acid is substituted for another within the groups of amino acids indicated herein below:
      • Amino acids having polar side chains (Asp, Glu, Lys, Arg, His, Asn, Gin, Ser, Thr, Tyr, and Cys)
      • Amino acids having non-polar side chains (Gly, Ala, Val, Leu, lie, Phe, Trp, Pro, and Met)
      • Amino acids having aliphatic side chains (Gly, Ala Val, Leu, Ile)
      • Amino acids having cyclic side chains (Phe, Tyr, Trp, His, Pro)
      • Amino acids having aromatic side chains (Phe, Tyr, Trp)
      • Amino acids having acidic side chains (Asp, Glu)
      • Amino acids having basic side chains (Lys, Arg, His)
      • Amino acids having amide side chains (Asn, Gin)
      • Amino acids having hydroxy side chains (Ser, Thr)
      • Amino acids having sulphur-containing side chains (Cys, Met),
      • Neutral, weakly hydrophobic amino acids (Pro, Ala, Gly, Ser, Thr)
      • Hydrophilic, acidic amino acids (Gin, Asn, Glu, Asp), and
      • Hydrophobic amino acids (Leu, Ile, Val)
  • Particular preferred conservative amino acids substitution groups are: Val-Leu-Ile, Phe-Tyr, Lys-Arg, Ala-Val, and Asn-Gin.
  • In some embodiments, the antibody of the conjugates described herein comprises a heavy chain having an amino acid sequence with 80% or more amino acid sequence identity (for example, about 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more sequence identity) to a heavy chain described herein. In some embodiments, the antibody of the conjugates described herein comprises a light chain having an amino acid sequence with 80% or more amino acid sequence identity (for example, about 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91% or more, 92% or more, 93% or more, 94% or more, 95% or more, 96% or more, 97% or more, 98% or more, 99% or more sequence identity) to a light chain described herein.
  • In some embodiments, the antibody of the conjugates described herein comprises a heavy chain having an amino acid sequence identical to the amino acid sequence of a heavy chain described herein, except that it includes 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid modifications (e.g., substitutions, insertions and/or deletions) relative to the amino acid sequence of the heavy chain described herein. In some embodiments, the antibody of the conjugates described herein comprises a light chain having an amino acid sequence identical to the amino acid sequence of a light chain described herein, except that it includes 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acid modifications (e.g., substitutions, insertions and/or deletions) relative to the amino acid sequence of the light chain described herein.
  • Antibody Production
  • Humanized antibodies, fragments and regions can be produced by cloning DNA segments encoding the H and L chain antigen-binding regions of the anti-AXL antibody, and joining these DNA segments to DNA segments including CH and CL regions, respectively, to produce full length immunoglobulin-encoding genes.
  • For full-length antibody molecules, the immunoglobulin cDNAs can be obtained from mRNA of hybridoma cell lines. Antibody heavy and light chains are cloned in a mammalian expression vector system. Assembly is documented with DNA sequence analysis. The antibody construct can be expressed in human or other mammalian host cell lines. The construct can be validated by transient transfection assays and immunoassay of the expressed antibody. Stable cell lines with the highest productivity can be isolated and screened using rapid assay methods.
  • Biological Activity
  • In Vitro Cell Proliferation Assays
  • Generally, the cytotoxic or cytostatic activity of an antibody is measured by: exposing mammalian cells having receptor proteins to the antibody in a cell culture medium; culturing the cells for a period from about 6 hours to about 5 to 7 days; and measuring cell viability. Cell-based in vitro assays are used to measure viability (proliferation), cytotoxicity, and induction of apoptosis (caspase activation) of an antibody of the disclosure.
  • The in vitro potency of antibodies can be measured by a cell proliferation assay. The CellTiter-Glo® Luminescent Cell Viability Assay is a commercially available (Promega Corp., Madison, Wis.), homogeneous assay method based on the recombinant expression of Coleoptera luciferase (U.S. Pat. Nos. 5,583,024; 5,674,713 and 5,700,670). This cell proliferation assay determines the number of viable cells in culture based on quantitation of the ATP present, an indicator of metabolically active cells (Crouch et al (1993) J. Immunol. Meth. 160:81-88; U.S. Pat. No. 6,602,677). The CellTiter-Glo® Assay is conducted in 96 well format, making it amenable to automated high-throughput screening (HTS) (Cree et al (1995) AntiCancer Drugs 6:398-404). The homogeneous assay procedure involves adding the single reagent (CellTiter-Glo® Reagent) directly to cells cultured in serum-supplemented medium. Cell washing, removal of medium and multiple pipetting steps are not required. The system detects as few as 15 cells/well in a 384-well format in 10 minutes after adding reagent and mixing. The cells may be treated continuously with antibody, or they may be treated and separated from antibody. Generally, cells treated briefly, i.e. 3 hours, showed the same potency effects as continuously treated cells.
  • The homogeneous “add-mix-measure” format results in cell lysis and generation of a luminescent signal proportional to the amount of ATP present. The amount of ATP is directly proportional to the number of cells present in culture. The CellTiter-Glo® Assay generates a “glow-type” luminescent signal, produced by the luciferase reaction, which has a half-life generally greater than five hours, depending on cell type and medium used. Viable cells are reflected in relative luminescence units (RLU). The substrate, Beetle Luciferin, is oxidatively decarboxylated by recombinant firefly luciferase with concomitant conversion of ATP to AMP and generation of photons.
  • The in vitro potency of antibody-drug conjugates can also be measured by a cytotoxicity assay. Cultured adherent cells are washed with PBS, detached with trypsin, diluted in complete medium, containing 10% FCS, centrifuged, re-suspended in fresh medium and counted with a haemocytometer. Suspension cultures are counted directly. Monodisperse cell suspensions suitable for counting may require agitation of the suspension by repeated aspiration to break up cell clumps.
  • The cell suspension is diluted to the desired seeding density and dispensed (100 μl per well) into black 96 well plates. Plates of adherent cell lines are incubated overnight to allow adherence. Suspension cell cultures can be used on the day of seeding.
  • A stock solution (1 ml) of antibody (20 μg/ml) is made in the appropriate cell culture medium.
  • Serial 10-fold dilutions of stock antibody are made in 15 ml centrifuge tubes by serially transferring 100 μl to 900 μl of cell culture medium.
  • Four replicate wells of each antibody dilution (100 μl) are dispensed in 96-well black plates, previously plated with cell suspension (100 μl), resulting in a final volume of 200 μl. Control wells receive cell culture medium (100 μl).
  • If the doubling time of the cell line is greater than 30 hours, antibody incubation is for 5 days, otherwise a four day incubation is done.
  • At the end of the incubation period, cell viability is assessed with the Alamar blue assay.
  • AlamarBlue (Invitrogen) is dispensed over the whole plate (20 μl per well) and incubated for 4 hours. Alamar blue fluorescence is measured at excitation 570 nm, emission 585 nm on the Varioskan flash plate reader. Percentage cell survival is calculated from the mean fluorescence in the antibody treated wells compared to the mean fluorescence in the control wells.
  • Use
  • The antibody of the disclosure may be used to target a target location.
  • The target location is preferably a proliferative cell population. The antibody is an antibody for an antigen present on a proliferative cell population.
  • In one embodiment the antigen is absent or present at a reduced level in a non-proliferative cell population compared to the amount of antigen present in the proliferative cell population, for example a tumour cell population.
  • The target location may be in vitro, in vivo or ex vivo.
  • The antibodies of the disclosure include those with utility for anticancer activity. Thus, in one aspect, the present disclosure provides an antibody as described herein for use in therapy.
  • In a further aspect there is also provides an antibody as described herein for use in the treatment of a proliferative disease. A second aspect of the present disclosure provides the use of an antibody in the manufacture of a medicament for treating a proliferative disease.
  • One of ordinary skill in the art is readily able to determine whether or not a candidate conjugate treats a proliferative condition for any particular cell type. For example, assays which may conveniently be used to assess the activity offered by a particular compound are described in the examples below.
  • The term “proliferative disease” pertains to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or hyperplastic growth, whether in vitro or in vivo.
  • Examples of proliferative conditions include, but are not limited to, benign, pre-malignant, and malignant cellular proliferation, including but not limited to, neoplasms and tumours (e.g. histocytoma, glioma, astrocyoma, osteoma), cancers (e.g. lung cancer, small cell lung cancer, gastrointestinal cancer, bowel cancer, colon cancer, breast carinoma, ovarian carcinoma, prostate cancer, testicular cancer, liver cancer, kidney cancer, bladder cancer, pancreas cancer, brain cancer, sarcoma, osteosarcoma, Kaposi's sarcoma, melanoma), lymphomas, leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g. of connective tissues), and atherosclerosis. Cancers of particular interest include, but are not limited to, metastatic cancer cells, such as circulating tumour cells, which may be found circulating in body fluids such as blood or lymph, lymphomas (e.g., non-Hodgkin's lymphoma, NHL), leukemia (particularly acute myeloid leukemia, AML) and ovarian cancers.
  • Any type of cell may be treated, including but not limited to, lung, gastrointestinal (including, e.g. bowel, colon), breast (mammary), ovarian, prostate, liver (hepatic), kidney (renal), bladder, pancreas, brain, and skin.
  • It is contemplated that the antibody of the present disclosure may be used to treat various diseases or disorders, e.g. characterized by the overexpression of a tumour antigen. Exemplary conditions or hyperproliferative disorders include benign or malignant tumours; leukemias, haematological, and lymphoid malignancies. Others include neuronal, glial, astrocytal, hypothalamic, glandular, macrophagal, epithelial, stromal, blastocoelic, inflammatory, angiogenic and immunologic, including autoimmune, disorders.
  • Generally, the disease or disorder to be treated is a hyperproliferative disease such as cancer. Examples of cancer to be treated herein include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemias or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g. epithelial squamous cell cancer), lung cancer including small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • Autoimmune diseases for which the antibody may be used in treatment include rheumatologic disorders (such as, for example, rheumatoid arthritis, Sjögren's syndrome, scleroderma, lupus such as SLE and lupus nephritis, polymyositis/dermatomyositis, cryoglobulinemia, anti-phospholipid antibody syndrome, and psoriatic arthritis), osteoarthritis, autoimmune gastrointestinal and liver disorders (such as, for example, inflammatory bowel diseases (e.g. ulcerative colitis and Crohn's disease), autoimmune gastritis and pernicious anemia, autoimmune hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, and celiac disease), vasculitis (such as, for example, ANCA-associated vasculitis, including Churg-Strauss vasculitis, Wegener's granulomatosis, and polyarteriitis), autoimmune neurological disorders (such as, for example, multiple sclerosis, opsocionus myocionus syndrome, myasthenia gravis, neuromyelitis optica, Parkinson's disease, Alzheimer's disease, and autoimmune polyneuropathies), renal disorders (such as, for example, glomerulonephritis, Goodpasture's syndrome, and Berger's disease), autoimmune dermatologic disorders (such as, for example, psoriasis, urticaria, hives, pemphigus vulgaris, bullous pemphigoid, and cutaneous lupus erythematosus), hematologic disorders (such as, for example, thrombocytopenic purpura, thrombotic thrombocytopenic purpura, post-transfusion purpura, and autoimmune hemolytic anemia), atherosclerosis, uveitis, autoimmune hearing diseases (such as, for example, inner ear disease and hearing loss), Behcet's disease, Raynaud's syndrome, organ transplant, and autoimmune endocrine disorders (such as, for example, diabetic-related autoimmune diseases such as insulin-dependent diabetes mellitus (IDDM), Addison's disease, and autoimmune thyroid disease (e.g. Graves' disease and thyroiditis)). More preferred such diseases include, for example, rheumatoid arthritis, ulcerative colitis, ANCA-associated vasculitis, lupus, multiple sclerosis, Sjögren's syndrome, Graves' disease, IDDM, pernicious anemia, thyroiditis, and glomerulonephritis.
  • Methods of Treatment
  • The antibody of the present disclosure may be used in a method of therapy. Also provided is a method of treatment, comprising administering to a subject in need of treatment a therapeutically-effective amount of an antibody of the disclosure. The term “therapeutically effective amount” is an amount sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage, is within the responsibility of general practitioners and other medical doctors.
  • An antibody may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. Examples of treatments and therapies include, but are not limited to, chemotherapy (the administration of active agents, including, e.g. drugs, such as chemotherapeutics); surgery; and radiation therapy.
  • A“chemotherapeutic agent” is a chemical compound useful in the treatment of cancer, regardless of mechanism of action. Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, antibodies, photosensitizers, and kinase inhibitors. Chemotherapeutic agents include compounds used in “targeted therapy” and conventional chemotherapy.
  • Examples of chemotherapeutic agents include: erlotinib (TARCEVA®, Genentech/OSI Pharm.), docetaxel (TAXOTERE®, Sanofi-Aventis), 5-FU (fluorouracil, 5-fluorouracil, CAS No. 51-21-8), gemcitabine (GEMZAR®, Lilly), PD-0325901 (CAS No. 391210-10-9, Pfizer), cisplatin (cis-diamine, dichloroplatinum(II), CAS No. 15663-27-1), carboplatin (CAS No. 41575-94-4), paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.J.), trastuzumab (HERCEPTIN®, Genentech), temozolomide (4-methyl-5-oxo-2,3,4,6,8-pentazabicyclo [4.3.0] nona-2,7,9-triene-9-carboxamide, CAS No. 85622-93-1, TEMODAR®, TEMODAL®, Schering Plough), tamoxifen ((Z)-2-[4-(1,2-diphenylbut-1-enyl)phenoxy]-N,N-dimethylethanamine, NOLVADEX®, ISTUBAL®, VALODEX®), and doxorubicin (ADRIAMYCIN®), Akti-1/2, HPPD, and rapamycin.
  • More examples of chemotherapeutic agents include: oxaliplatin (ELOXATIN®, Sanofi), bortezomib (VELCADE®, Millennium Pharm.), sutent (SUNITINIB®, SU11248, Pfizer), letrozole (FEMARA®, Novartis), imatinib mesylate (GLEEVEC®, Novartis), XL-518 (Mek inhibitor, Exelixis, WO 2007/044515), ARRY-886 (Mek inhibitor, AZD6244, Array BioPharma, Astra Zeneca), SF-1126 (PI3K inhibitor, Semafore Pharmaceuticals), BEZ-235 (PI3K inhibitor, Novartis), XL-147 (PI3K inhibitor, Exelixis), PTK787/ZK 222584 (Novartis), fulvestrant (FASLODEX®, AstraZeneca), leucovorin (folinic acid), rapamycin (sirolimus, RAPAMUNE®, Wyeth), lapatinib (TYKERB®, GSK572016, Glaxo Smith Kline), lonafamib (SARASAR™, SCH 66336, Schering Plough), sorafenib (NEXAVAR®, BAY43-9006, Bayer Labs), gefitinib (IRESSA®, AstraZeneca), irinotecan (CAMPTOSAR®, CPT-11, Pfizer), tipifamib (ZARNESTRA™, Johnson & Johnson), ABRAXANE™ (Cremophor-free), albumin-engineered nanoparticle formulations of paclitaxel (American Pharmaceutical Partners, Schaumberg, II), vandetanib (rINN, ZD6474, ZACTIMA®, AstraZeneca), chloranmbucil, AG1478, AG1571 (SU 5271; Sugen), temsirolimus (TORISEL®, Wyeth), pazopanib (GlaxoSmithKline), canfosfamide (TELCYTA®, Telik), thiotepa and cyclosphosphamide (CYTOXAN®, NEOSAR®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analog topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogs); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlomaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g. calicheamicin, calicheamicin gamma1I, calicheamicin omegal1 (Angew Chem. Intl. Ed. Engl. (1994) 33:183-186); dynemicin, dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-nordeucine, morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, nemorubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, rordin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; vinorelbine (NAVELBINE®); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XELODA®, Roche); ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.
  • Also included in the definition of “chemotherapeutic agent” are: (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumours such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole), FEMARA® (letrozole; Novartis), and ARIMIDEX® (anastrozole; AstraZeneca); (iii) anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); (iv) protein kinase inhibitors such as MEK inhibitors (WO 2007/044515); (v) lipid kinase inhibitors; (vi) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, for example, PKC-alpha, Raf and H-Ras, such as oblimersen (GENASENSE®, Genta Inc.); (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME®) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN®, LEUVECTIN®, and VAXID®; PROLEUKIN® rIL-2; topoisomerase 1 inhibitors such as LURTOTECAN®; ABARELIX® rmRH; (ix) anti-angiogenic agents such as bevacizumab (AVASTIN®, Genentech); and pharmaceutically acceptable salts, acids and derivatives of any of the above.
  • Also included in the definition of “chemotherapeutic agent” are therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen Idec), ofatumumab (ARZERRA®, GSK), pertuzumab (PERJETA™, OMNITARG™, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
  • Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the conjugates of the disclosure include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizumab, numavizumab, ocrelizumab, omalizumab, palivizumab, pascolizumab, pecfusituzumab, pectuzumab, pertuzumab, pexelizumab, ralivizumab, ranibizumab, reslivizumab, reslizumab, resyvizumab, rovelizumab, ruplizumab, sibrotuzumab, siplizumab, sontuzumab, tacatuzumab tetraxetan, tadocizumab, talizumab, tefibazumab, tocilizumab, toralizumab, trastuzumab, tucotuzumab celmoleukin, tucusituzumab, umavizumab, urtoxazumab, and visilizumab.
  • Pharmaceutical compositions according to the present disclosure, and for use in accordance with the present disclosure, may comprise, in addition to the active ingredient, i.e. an antibody, a pharmaceutically acceptable excipient, carrier, buffer, stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, e.g. cutaneous, subcutaneous, or intravenous.
  • Pharmaceutical compositions for oral administration may be in tablet, capsule, powder or liquid form. A tablet may comprise a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. A capsule may comprise a solid carrier such a gelatin.
  • For intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringers Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required.
  • Formulations
  • While it is possible for the antibody to be used (e.g., administered) alone, it is often preferable to present it as a composition or formulation.
  • In one embodiment, the composition is a pharmaceutical composition (e.g., formulation, preparation, medicament) comprising an antibody, as described herein, and a pharmaceutically acceptable carrier, diluent, or excipient.
  • In one embodiment, the composition is a pharmaceutical composition comprising at least one antibody, as described herein, together with one or more other pharmaceutically acceptable ingredients well known to those skilled in the art, including, but not limited to, pharmaceutically acceptable carriers, diluents, excipients, adjuvants, fillers, buffers, preservatives, anti-oxidants, lubricants, stabilisers, solubilisers, surfactants (e.g., wetting agents), masking agents, colouring agents, flavouring agents, and sweetening agents.
  • In one embodiment, the composition further comprises other active agents, for example, other therapeutic or prophylactic agents.
  • Suitable carriers, diluents, excipients, etc. can be found in standard pharmaceutical texts. See, for example, Handbook of Pharmaceutical Additives, 2nd Edition (eds. M. Ash and I. Ash), 2001 (Synapse Information Resources, Inc., Endicott, N.Y., USA), Remington's Pharmaceutical Sciences, 20th edition, pub. Lippincott, Williams & Wilkins, 2000; and Handbook of Pharmaceutical Excipients, 2nd edition, 1994.
  • Another aspect of the present disclosure pertains to methods of making a pharmaceutical composition comprising admixing at least one [11C]-radiolabelled antibody or antibody-like molecule, as defined herein, together with one or more other pharmaceutically acceptable ingredients well known to those skilled in the art, e.g., carriers, diluents, excipients, etc. If formulated as discrete units (e.g., tablets, etc.), each unit contains a predetermined amount (dosage) of the active compound.
  • The term “pharmaceutically acceptable,” as used herein, pertains to compounds, ingredients, materials, compositions, dosage forms, etc., which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of the subject in question (e.g., human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each carrier, diluent, excipient, etc. must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
  • The formulations may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with a carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active compound with carriers (e.g., liquid carriers, finely divided solid carrier, etc.), and then shaping the product, if necessary.
  • The formulation may be prepared to provide for rapid or slow release; immediate, delayed, timed, or sustained release; or a combination thereof.
  • Formulations suitable for parenteral administration (e.g., by injection), include aqueous or non-aqueous, isotonic, pyrogen-free, sterile liquids (e.g., solutions, suspensions), in which the active ingredient is dissolved, suspended, or otherwise provided (e.g., in a liposome or other microparticulate). Such liquids may additional contain other pharmaceutically acceptable ingredients, such as anti-oxidants, buffers, preservatives, stabilisers, bacteriostats, suspending agents, thickening agents, and solutes which render the formulation isotonic with the blood (or other relevant bodily fluid) of the intended recipient. Examples of excipients include, for example, water, alcohols, polyols, glycerol, vegetable oils, and the like. Examples of suitable isotonic carriers for use in such formulations include Sodium Chloride Injection, Ringers Solution, or Lactated Ringers Injection. Typically, the concentration of the active ingredient in the liquid is from about 1 ng/ml to about 10 μg/ml, for example from about 10 ng/ml to about 1 μg/ml. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.
  • Dosage
  • It will be appreciated by one of skill in the art that appropriate dosages of the antibody, and compositions comprising the antibody, can vary from patient to patient. Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects. The selected dosage level will depend on a variety of factors including, but not limited to, the activity of the particular compound, the route of administration, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds, and/or materials used in combination, the severity of the condition, and the species, sex, age, weight, condition, general health, and prior medical history of the patient. The amount of compound and route of administration will ultimately be at the discretion of the physician, veterinarian, or clinician, although generally the dosage will be selected to achieve local concentrations at the site of action which achieve the desired effect without causing substantial harmful or deleterious side-effects.
  • Administration can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell(s) being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician, veterinarian, or clinician.
  • In general, a suitable dose of the antibody is in the range of about 100 ng to about 25 mg (more typically about 1 μg to about 10 mg) per kilogram body weight of the subject per day. Where the active compound is a salt, an ester, an amide, a prodrug, or the like, the amount administered is calculated on the basis of the parent compound and so the actual weight to be used is increased proportionately.
  • In one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 100 mg, 3 times daily.
  • In one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 150 mg, 2 times daily.
  • In one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 200 mg, 2 times daily.
  • However in one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 50 or about 75 mg, 3 or 4 times daily.
  • In one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 100 or about 125 mg, 2 times daily.
  • For the prevention or treatment of disease, the appropriate dosage of an antibody of the disclosure will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the molecule is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The molecule is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.1-20 mg/kg) of molecule is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. A typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. An exemplary dosage of antibody to be administered to a patient is in the range of about 0.1 to about 10 mg/kg of patient weight. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. An exemplary dosing regimen comprises a course of administering an initial loading dose of about 4 mg/kg, followed by additional doses every week, two weeks, or three weeks of an antibody. Other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • Treatment
  • The term “treatment,” as used herein in the context of treating a condition, pertains generally to treatment and therapy, whether of a human or an animal (e.g., in veterinary applications), in which some desired therapeutic effect is achieved, for example, the inhibition of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, regression of the condition, amelioration of the condition, and cure of the condition. Treatment as a prophylactic measure (i.e., prophylaxis, prevention) is also included.
  • The term “therapeutically-effective amount,” as used herein, pertains to that amount of an antibody, or a material, composition or dosage from comprising an antibody, which is effective for producing some desired therapeutic effect, commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.
  • Similarly, the term “prophylactically-effective amount,” as used herein, pertains to that amount of an antibody, or a material, composition or dosage from comprising an antibody, which is effective for producing some desired prophylactic effect, commensurate with a reasonable benefit/risk ratio, when administered in accordance with a desired treatment regimen.
  • The Subject/Patient
  • The subject/patient may be an animal, mammal, a placental mammal, a marsupial (e.g., kangaroo, wombat), a monotreme (e.g., duckbilled platypus), a rodent (e.g., a guinea pig, a hamster, a rat, a mouse), murine (e.g., a mouse), a lagomorph (e.g., a rabbit), avian (e.g., a bird), canine (e.g., a dog), feline (e.g., a cat), equine (e.g., a horse), porcine (e.g., a pig), ovine (e.g., a sheep), bovine (e.g., a cow), a primate, simian (e.g., a monkey or ape), a monkey (e.g., marmoset, baboon), an ape (e.g., gorilla, chimpanzee, orangutang, gibbon), or a human.
  • Furthermore, the subject/patient may be any of its forms of development, for example, a foetus. In one preferred embodiment, the subject/patient is a human.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1: AXL binding ELISA of fully humanised constructs. (2A) to Human AXL. (2B) to Cynomolgus monkey AXL.
  • FIG. 2: Accelerated stability analysis
  • Materials and Methods
  • Protocol 1: Production of DNA Plasmids for Expression
  • Materials
      • For heavy chain construct selection, Zeocin 25 μg/ml (Invivogen) was used.
      • For light chain construct selection, Blasticidin 100 μg/ml (Life Technologies) was used.
  • Method
  • Transformed bacteria were spread on LB-agar Zeocin/Blasticidin plates, as required, incubated overnight at 37 C, then colonies were picked from each plate.
  • VH or VK colonies/glycerol stocks were inoculated into 3 ml LB containing Zeocin 25 μg/ml or Blasticidin 50 μg/ml, respectively.
  • p21, p27, pAdvantage (Promega) and pSVLT (generous gift from Tom Vink) were inoculated in LB-Ampicillin and shaken overnight.
  • 3 ml overnight colony was seeded into 200 ml of LB-antibiotic and shaken overnight. DNA plasmids were isolated from each culture using the Promega PureYield™ Plasmid Maxiprep Kit following the manufacturer's instructions.
  • Protocol 2. Transient Transfection of HEK293T Cells with Expression Constructs
  • Materials
      • Cells: HEK293T cells
      • Culture medium: DMEM high glucose 4.5 g/L (PAA) with 10% v/v FCS, penicillin and streptomycin
      • Fugene HD transfection reagent (Promega #E2311)
      • Opti-MEM (Life Technologies #11058-021) or
      • FreeStyle 293 Expression Medium (Life Technologies #12338-018)
  • Method
  • Grow HEK293T cells in a T75 or T175 flask in a CO2-gassed cell culture incubator. Split cultures 1:3 every 2 days or 1:4 to 1:5 every 3-4 days. The cells adhere weakly to the flasks and only a light trypsinisation is necessary to detach cells during passaging.
  • The day before transfection:
      • 1. Trypsinise the cells, wash 1× in DMEM/10% FCS and count the cells.
      • 2. Seed cells in a 6 well plate in 2 ml per well containing 2×105 cells.
  • Next day, check cells are at least 80% confluent and replace the medium (2 ml/well).
      • 1. 1.2 μg of total DNA (0.6 ug of each high and light chain DNA) is needed for each transfection and better results are obtained if the DNA concentration is at or above 90 ng/μl.
      • 2. Add 0.6 ug of VH and 0.6 ug VK expression plasmid DNAs into of Fugene HD (4.5 μl) and OptiMEM/Freestyle medium, in a total volume of 60 ul, avoiding touching the sides of the tube with the Fugene HD.
      • 3. Mix and leave at RT for 15 minutes.
      • 4. Add Fugene mixture drop-wise around the well of HEK293T cells.
      • 5. Return the 6-well plate to the CO2-gassed cell culture incubator for 4 days.
      • 6. Harvest each conditioned medium, centrifuge, and store at 4° C.
  • Protocol 3: IgG Quantitation by ELISA
  • Materials
      • Nunc-Immuno Plate MaxiSorp (Life Technologies, 43945A)
      • Goat Anti-Human IgG(Fc)—AffiniPure: Stratech Scientific, 109-005-098-JIR; 1 mg: 1.3 mg/ml
      • Human IgG1/kappa antibody (Sigma, 1-3889-1 mg: 1 mg/ml)
      • Goat anti-human kappa light chain peroxidase conjugate (Sigma, A-7164-1 ml)
      • 1-Step Turbo TMB-ELISA, 250 mL (Thermo Scientific: #34022)
      • Acid stop=0.1M HCL
      • Sample enzyme conjugate (SEC) buffer Tween 20 (0.02% v/v), BSA 0.2% (w/v) in PBS
      • Washing buffer 1×PBS, Tween 20 (0.1% v/v)
  • Method
      • 1. Coat each well of a 96-well immunoplate with 100 μl aliquots of 0.4 μg/ml (dilute stock×3000=10 ul in 30 ml: coat 5 ml per plate) goat anti-human IgG antibody, diluted in PBS, incubate overnight at 4° C. (or 37 C 1 hr). (Plates may be stored for 1 month at this stage). Also coat another blank plate with BSA/PBS blocking solution.
      • 2. wash plate 3× with 200 μl/well of washing buffer.
      • 3. Block coated plate: add 200 ul 3% BSA in PBS: incubate 37 C 1 hr
      • 4. Into blank plate, dispense 200 μl SEC buffer into all wells except row B, cols 2-11 (blue, below).
      • 5. Prepare 1 ug/ml solution of the human IgG1/kappa antibody in SEC buffer (×1000 diln)
  • 1 2 3 4 5 6 7 8 9 10 11 12
    A STD unk4
    B STD unk4
    C unk1 unk5
    D unk1 unk5
    E unk2 unk6
    F unk2 unk6
    G unk3 unk7
    H unk3 unk7
    200 μl diluent in every well
    +50 μl sample +50 μl sample
    Transfer 100 μl
    Figure US20220073625A1-20220310-P00001
    Transfer 100 μl
    Figure US20220073625A1-20220310-P00001
    200 66.67 22.22 7.41 2.47 0.82 200 66.67 22.22 7.41 2.47 0.82 ng/ml
    Stock std = 1 μg/ml
        • BLOCKED UNCOATED PLATE:
      • 6. Pipette 50 μl/well of stds/unknowns into rows A-H, cols 1 and 7 (makes a ×5 dilution).
      • 7. Serially transfer 100 μl across plate to achieve serial ×3 dilution series.
      • 8. Transfer 100 μl from each well to the corresponding well of the BLOCKED anti-IgG-COATED plate.
        • BLOCKED anti-IgG COATED PLATE:
      • 9. Incubate at 37° C. for 1 hr. Rinse all the wells 3× with washing buffer (200 μl).
      • 10. Dilute the goat anti-human kappa light chain peroxidase conjugate 5000-fold in SEC buffer and add 100 01 to each well. Repeat the incubation and washing steps (step 9).
      • 11. Add 100 μl of TMB Turbo substrate to each well, incubate in the dark at room temperature for 10 min.
      • 12. Stop the reaction by adding 50 μl of acid (0.1M HCl) to each well.
      • 13. Read the optical density at 450 nm.
  • Protocol 4: AXL Binding ELISA
  • Materials
      • Human AXL-Strep-His was produced by Evitria AG in transiently transfected CHO cells and purified on Ni Sepharose High Performance (GE Healthcare 17-5268-01) following manufacturers instructions and stored in aliquots at −20 C.
      • Goat anti-human kappa light chain peroxidase conjugate (Sigma, A-7164-1 ml)
      • Nunc-Immuno Plate MaxiSorp (Life Technologies, 43945A)
      • Plate washer Biotek LS405
      • 3% BSA: BSA 3% w/v in PBS
      • PBS Tween: Tween 20 0.05% v/v in PBS
      • PBS/Tween/BSA: BSA 0.5% w/v in PBS/Tween
      • 1-Step Turbo TMB-ELISA (Thermo Scientific #3402)
  • Protocol
      • 1. Dispense 50 μl/well of human AXL-strep-His (1 ug/ml in PBS)
      • 2. Cover with adhesive plate sealer and incubate at 4 C overnight.
      • 3. Block: Dispense 50 μl/well of 3% BSA and incubate for 1 hr 37 C,
      • 4. Wash plate with PBS/Tween
      • 5. Serially 3-fold dilute 5E5 antibodies (2 ml HEK293T culture supernatants) on non-binding polypropylene plate in PBS/Tween/BSA: serially transfer 50 ul onto 100 ul.
      • 6. Transfer 50 ul from antibody dilution plate onto washed, blocked AXL-coated plate
      • 7. Incubate 37 C 1 hr
      • 8. Wash plate with PBS/Tween
      • 9. Dispense anti-human IgG-HRP conjugate, diluted 1:1000 in PBS/Tween/BSA
      • 10. Incubate 37 C 1 hr
      • 11. Wash plate with PBS/Tween 3
      • 12. Wash plate with PBS
      • 13. Dispense 100 ul/well 1-Step Turbo TMB-ELISA substrate solution
      • 14. Incubate 30 min at room temperature (or less if reaction is rapid)
      • 15. Dispense 100 ul/well 0.6M HCl to stop the substrate reaction
      • 16. Measure optical density at 450 nm
  • Protocol 4A: SPR Measurement of Antibody Affinity
  • Materials
      • 1. Sensor Chip CM5 Biacore; Cat. #BR-1000-14
      • 2. Amine Coupling Kit (EDC, NHS, ethanolamine-HCl) Biacore; Cat. #BR-1000-50
      • 3. Immobilization buffer (10 mM Na acetate, pH 4.0) Biacore; Cat. #BR-1003-49
      • 4. 50 mM NaOH Biacore; Cat. #BR-1003-58
      • 5. Running buffer (PBS/Tween20 0.05% v/v)
      • 6. Biacore T200 GE Healthcare
      • 7. Regeneration solution: 10 mM HCl, 1 M NaCl
  • Coupling Method
      • Activate flow cell 2 with NHS-EDC 420s at Sp/min, then inject human Axl-Strep-His (Evitria) (10 μg/mL in 10 mM sodium acetate, pH 4.0) to achieve a coupling of 10-20 RU. Block with ethanolamine for 420s. Repeat the process for flow cell 1 but with no antigen to create a reference flow cell. 12RU AXL fusion protein was coated.
      • For the Fc fusion, Human Axl-Fc chimera (R&D Systems #154-AL) (5 μg/mL in 10 mM sodium acetate, pH 4.5) was used with the above protocol. 16RU of AXL-Fc was coated.
  • Protocol with AXL-Strep-his Antigen
      • Serial 10× dilutions of antibody, from 3000 nM, were made in PBS/Tween20. 2× regeneration cycles were used with 10 mM HCl, 1 M NaCl for 30s at 30 μl/min for both cycles. Start-up solution was PBS/Tween20 and set to 3 cycles.
      • Sample injection parameters were 120s at 30 μl/min, with 600s dissociation time.
      • Prime and normalise detector were run before sample application with experimental conditions 25 C and sample storage at 4 C.
      • Kinetic analysis used BIAevaluation software with bivalent ligand binding model. For chimeric 1H12 with AXL-Strep-His, “heterogeneous ligand” binding model was used due to poor fit with bivalent or monovalent algorithms.
      • Each antibody dilution was injected twice.
  • Protocol with AXL-Fc Chimera Antigen
      • The above protocol was used except that the running buffer was HBSEP+, serial 10× dilutions of antibody were from 500 nM in HBSEP+ running buffer and were injected for 180 sec.
      • Analysis used BIAevaluation software with the bivalent binding model.
  • Protocol 5: Capillary Isoelectric Focusing
  • Materials
      • PA 800 plus (AB SCIEX)
      • Anolyte Solution: 200 mM Phosphoric Acid (Sigma-Aldrich #345 245).
      • 4.3M Urea (Sigma-Aldrich #U0631) in water.
      • Catholyte Solution: 300 mM Sodium Hydroxide.
      • 3M Urea (Sigma-Aldrich #U0631) in cIEF Gel (Beckman Coulter #477497).
      • Chemical Mobiliser 350 mM Acetic Acid (Sigma-Aldrich #537 020).
      • Pharmalyte 3-10 (GE Healthcare 17-0456-01).
      • Cathodic Stabiliser 500 mM Arginine (Sigma-Aldrich #A5006).
      • cIEF Peptide markers PI 4-10 (Beckman Coulter #A58481).
      • Anodic Stabiliser: 200 mM Iminodiacetic Acid (Sigma-Aldrich #220 000).
      • Neutral Capillary, 50 μm i.d.×45 cm, (Beckman Coulter #477 441)
  • Method
      • Turn on the PA800+ machine and UV lamp, allowing it to warm up 30 mins before use.
      • Clean the system electrodes and interface block with a damp Kimwipe.
      • Prepare buffer trays as shown, with 1.5 mL reagent per vial, 1 mL of water in the waste
      • 1. DDI water
      • 2. Anolyte
      • 3. Urea Solution
      • 4. 3M urea/cIEF Gel
      • 5. Chemical Mobilizer
      • 6. Waste
      • 7. Catholyte
      • 8. Chemical Mobilizer
      • 9. BI (Inlet Buffer Tray)
      • 10. BO (Outlet Buffer Tray)
      • NOTE. Each set of buffer vials is good for 6 consecutive runs or for 24 hours inside the instrument.
        • Insert the capillary cartridge into the system and close the front panel.
      • NOTE. Do not expose the neutral-coated capillary ends to air for more than fifteen min. When the capillary is not in use, submerge the capillary ends in vials filled with DDI water.
        • Prepare cIEF master mix using the table below. Dispense each reagent into a centrifuge tube, vortex 1 min, invert every 15-20 sec to ensure complete mixing and store at 2° C. to 8° C. for up to 1 day.
  • Volume per
    Reagent sample (μl)
    3M Urea-cIEF Gel 100
    Pharmalyte 3-10 12
    Cathodic Stabiliser 20
    Anodic Stabiliser 2
    pI marker 10 2
    pI marker 9.5 2
    pI marker 7 2
    pI marker 5.5 2
    pI marker 4.1 2
        • Desalt and concentrate each antibody (to about 5 mg/ml) in 2M urea using Amicon Ultra 0.5 mL centrifugal filters (Sigma Z677108).
        • Mix 200 μL of master mix with 10 μL of protein, vortex the cIEF sample for 1 min, inverting the tube every 15-20 sec, then centrifuge at high speed to remove any air bubbles. Transfer 100 μL of sample into a micro vial. Place the micro vial into a universal plastic vial and cap it with a blue cap. Then place the sample vial in the inlet sample tray.
        • Run the “cIEF Conditioning—PA 800 plus.met” method to condition the column.
          • Rinse for 5 min at 50 psi with Chemical Mobilizer. See FIG. 2.10.
          • 2 Rinse for 2 min at 50 psi with DDI water.
          • 3 Rinse for 5 min at 50 psi with cIEF gel.
          • 4 Submerge both of the capillary ends in vials filled with DDI water.
        • Use the “cIEF Separation—PA 800 plus.met” method to create a sequence containing all protein samples and a blank.
          • Rinse for 3 min at 50 psi with Urea Solution. See FIG. 2.11.
          • Rinse for 2 min at 50 psi with DDI water.
          • Inject sample for 99.0 sec at 25 psi.
          • Water dip by submerging both capillary ends in DDI water.
          • Focusing step, 15 min at 25 kV under normal polarity (Time=0).
          • Chemical mobilization, 30 min at 30 kV under normal polarity (Time=15 min).
          • Stop data collection (Time=45 min).
          • Rinse for 2 min at 50 psi with DDI water (Time=45.10 min).
          • Submerged both of the capillary ends in DDI water (Time=47.20 min).
          • End the method (Time=47.30 min).
        • Put the “cIEF Shutdown—PA 800 plus.met” method at the end of the sequence to rinse the capillary and turn off the UV.
          • Rinse for 2 min at 50 psi with DDI water. See FIG. 2.12.
          • Rinse for 5 min at 50 psi with cIEF gel.
          • Turn off the UV lamp.
          • Submerge both of the capillary ends in vials filled with DDI water.
        • For short term storage (1 to 3 days), leave the capillary on the instrument. For long term storage (over 3 days), place the capillary cartridge in the storage box with both ends submerged in water tubes and store upright at 4 C.
  • Protocol 6: Protein Thermal Shift Protocol
  • Materials
      • 96 well optical plate semi-skirted (Stariab cat. L1402-9700).
      • Protein thermal shift dye kit (Life Technologies cat. 446148).
      • Microamp optical adhesive film (Applied Biosystems)
      • 7500 Real-Time PCR System (Life Technologies)
      • Test items: Antibodies from Evitria and Spirogen
      • Protein thermal shift v1.2 software (Life Technologies)
  • Method
  • Protein thermal shift dye (2.5 μL 1:1000 dilution) was added to sample proteins (17.5 μL of 0.5 mg/mL in PBS) in a 96 well optical plate and mixed thoroughly. Every sample was done in quadruplicate. The plate was sealed with a optical adhesive film and bubbles in the wells were removed by centrifugation 1 min at 500 g, then placed on ice. The sealed plate was introduced in the 7500 Real-Time PCR System and subsequently the experiment was set up as follows:
  • Experiment Name Name (using up to
    100 letters/numbers)
    Instrument type 7500 Fast (96 Wells)
    or 7500 (96 Wells)
    Experiment type Melt Curve
    Reagent type Other
    Ramp speed Standard
    Reporter ROX
    Quencher None
    Passive Reference None
  • Temperature cycling on the ABI 7500 set up:
  • Reaction vol 20 μl
    Ramp mode continuous
    Step Ramp rate Temp ° C. Time (mm:ss)
    1 100% 25.0 02:00
    2  1% 99.0 02:00
  • Data analysis and derivation of Tm data were done using the software following the manufacturers instructions.
  • Protocol 7: HPLC Size Exclusion Chromatography
  • Materials
      • Shimadzu HPLC system, or equivalent, consisting of the following, or equivalent:
      • 2× DGU-20ASR Prominence Degassing units
      • 2× LC-20ADXR Nexera Pumps
      • SIL-20ACXR Nexera Autosampler
      • CTO-20AC Prominence Column oven
      • SPD-M30A Prominence DAD detector
      • Computer with LabSolutions software.
      • TSKgeI Super SW mAb HTP 4 um 4.6 mm×15 cm
      • 200 mM Potassium Phosphate, 250 mM Potassium Chloride, 10% v/v i-Propanol, pH 6.95.
  • Sample Preparation
    • Mobile Phase: 200 mM Potassium Phosphate, 250 mM Potassium Chloride, 10% v/v i-Propanol, pH 6.95.
    • Analytical Sample: Inject 2-20 μL of neat ADC sample (at least 1 mg/ml for best results).
  • Typically 10 μL of 5 mg/ml gives good results.
  • HPLC Parameters
    • Method File name: MSOP-018
    • HPLC Column: TSKgeI Super SW mAb HTP 4 um 4.6 mm×15 cm
    • Flow Rate: 0.5 ml/min
    • Injection volume: 2-20 μL
    • Detection, UV: 280 nm
      • 330 nm (for information only)
    • Column Temp: ambient temperature
    • Autosampler Temp: 15° C.
    • Gradient: Isocratic
  • Method
    • Intact ADCs typically elute at ˜16-18 minutes.
    • Aggregates typically elute at ˜11-14 minutes.
    • Low molecular weight species typically elute at ˜>20 minutes.
  • Protocol 8: Hydrophobic Interaction Chromatograph
  • Materials
      • HPLC system, or equivalent, consisting of the following, or equivalent:
      • SRD-3600 SOLVENT RACK, 6 DEGASS. LINES
      • HPG-3400RS PUMP (Thermo Scientific)
      • HPG-3200RS PUMP (Thermo Scientific)
      • WPS-3000TFC ANALYTICAL AUTOSAMPLER (Thermo Scientific)
      • TCC-3000RS COLUMN THERMOSTAT (Thermo Scientific)
      • DAD-3000RS DETECTOR (Thermo Scientific)
      • Computer with Chromeleon software (Thermo Scientific)
      • Proteomix HIC Butyl-NP5, 5 um, non-porous, 4.6×35 mm (Sepax) column
      • Ammonium sulfate ((NH4)2SO4)
      • Sodium acetate (NaOAc)
      • i-Propanol
      • Water, HPLC grade
    • Mobile Phase A: 1.25 M (NH4)2SO4, 25 mM NaOAc (pH 5.50)
    • Mobile Phase B: 75% 25 mM NaOAc (pH 5.50), 25% i-Propanol
    • Blank Solution: Water
  • Sample Preparation
    • Analytical Sample: ≤10 μL of neat ADC sample at a concentration of 1-5 mg/mL.
  • HPLC Parameters
    • Method File name: HIC_Gradient_1_AB
    • HPLC Column: Proteomix HIC Butyl-NP5, Sum, non-porous, 4.6×35 mm (Sepax)
    • Flow Rate: 0.8 ml/min
    • Injection volume: 5 10 μL
    • Detection, UV: 214 nm
      • 330 nm (for information only)
    • Column Temp: 25° C.
    • Autosampler Temp: 10° C.
  • Gradient
  • Time (min) % B
    0 0
    1 0
    13 100
    14 100
    14.1 0
    18 0
  • Method
  • Flush the flow-path of the HPLC and column with water. Set up the HPLC under the operating conditions outlined above and equilibrate the system for a minimum of 10 minutes.
  • Protocol 9: Reverse Phase Chromatograph
  • Materials
      • HPLC system, or equivalent, consisting of the following, or equivalent:
      • SRD-3600 SOLVENT RACK, 6 DEGASS. LINES
      • HPG-3400RS PUMP (Thermo Scientific)
      • HPG-3200RS PUMP (Thermo Scientific)
      • WPS-3000TFC ANALYTICAL AUTOSAMPLER (Thermo Scientific)
      • TCC-3000RS COLUMN THERMOSTAT (Thermo Scientific)
      • DAD-3000RS DETECTOR (Thermo Scientific)
      • Computer with Chromeleon software (Thermo Scientific)
      • Aeris Widepore XB-C18, 200 Å, 3.6 μm, 2.1×150 mm (Phenomenex, 00F-4482-AN)
      • Acetonitrile, HPLC grade.
      • Water, HPLC grade.
      • Trifluoroacetic acid (TFA), HPLC grade.
      • 100 mM NaBorate, pH 8.4
      • 500 mM DTT
      • 49:49:2 Acetonitrile/Water/Formic acid
    • Mobile Phase A: Water+0.1% v/v TFA.
    • Mobile Phase B: Acetonitrile+0.1% v/v TFA.
    • Blank Solution: 1:1 v/v Acetonitrile/Water.
  • Sample Preparation
  • To 40 μL of sample (5 mg/ml) add water, 30 μl, NaBorate, 20 μl, and DTT (500 mM), 10 μL. Incubate at 37° C., 30 min, then add 100 μl of 49:49:2 Acetonitrle/Water/Formic acid.
  • HPLC Parameters
    • Method File name: RP_Aeris_Column6
    • HPLC Column: Aeris Widepore XB-C18, 200 Å, 3.6 μm, 2.1×150 mm (Phenomenex, 00F-4482-AN)
    • Flow Rate: 1.0 ml/min
    • Injection volume: 10 μl (or full loop)
    • Detection, UV: 214 nm
      • 330 nm (for information only)
    • Column Temp: 80° C.
    • Autosampler Temp: 15° C.
  • Gradient
  • Time (minutes) % B
    0 22.5
    1 22.5
    11 50
    11.5 90
    13.5 90
    14.5 22.5
    16 22.5
  • Method
  • Set up the HPLC under the operating conditions outlined above and equilibrate the system for a minimum of 10 minutes. Inject a blank sample, followed by the sample for analysis.
  • Example 1: Characterization of Humanized Antibodies
  • The five antibodies described below were produced, expressed, and quantified according to Protocols 1-3 described herein. The expression levels recorded are shown below in Table 1.
      • Ab1 is an anti-AXL antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies.
      • Ab2 is an anti-AXL antibody comprising a VH domain having the sequence according to SEQ ID NO. 2, a VL domain having the sequence according to SEQ ID NO. 5, and a constant region derived from one or more human antibodies.
      • Ab3 is an anti-AXL antibody comprising a VH domain having the sequence according to SEQ ID NO. 2, a VL domain having the sequence according to SEQ ID NO. 7, and a constant region derived from one or more human antibodies.
      • Ab4 is an anti-AXL antibody comprising a VH domain having the sequence according to SEQ ID NO. 3, a VL domain having the sequence according to SEQ ID NO. 5, and a constant region derived from one or more human antibodies.
      • Ab5 is an anti-AXL antibody comprising a VH domain having the sequence according to SEQ ID NO. 3, a VL domain having the sequence according to SEQ ID NO. 7, and a constant region derived from one or more human antibodies.
  • A stability assay was performed during which the antibodies were heated in sterile PBS at 40° C. for 60 hr and analysed for aggregation by SEC and for binding activity by human AXL ELISA. No increase in aggregation or loss in AXL binding activity was detected for any of the antibodies (see FIG. 2).
  • The antibodies were further characterised using Protocols 5-9 as described herein. The results are shown below in Table 1 (all assays were performed on the HEK293F expression product unless otherwise stated; “F”=HEK293F, “T”=HEK293T).
  • TABLE 1
    RP HPLC
    SEC % LC2 peak
    Expression (μg/mL) monomer HIC retention % protein
    in . . . 280 nm time min Tm 280 nm
    Antibody T F CHO CHO F F CHO pl (° C.) F CHO
    Ab1 33.7, 7.16 366 97.9 87% 4.5 4.5 8.05 69.52 0 0
    40.1
    Ab2 18.2 2.55 265 97.6 86% 5.2 5.2 8.06 59.71 6.4% 5.9%
    Ab3 15.2 2.24 287 98.3 88% 4.8 4.7 8.15 62.05 0 0
    Ab4 17.8 1.91 306 96.9 96% 5.6 5.6 7.56 60.32 8.1% 5.9%
    Ab5 16.5 1.95 298 97.7 98% 5.0 4.9 7.54 63.06 0 0
  • Binding of the antibodies to AXL antigens indicated that binding was unusually sensitive to both antigen preparation and presentation and antibody geometry.
  • Initial measurements by ELISA using Axl-Strep-His antigen, as disclosed in Protocol 4 suggested that the binding of antibodies comprising humanised 1H12 heavy and light chains (Ab2-Ab5) were broadly similar to the antibody comprising the murine VH and VL domains (Abi) (see FIG. 1).
  • SPR measurements of antibody affinity using Axl-Strep-His antigen indicated that Ab2, Ab3, and Ab5 had higher affinity for Axl-Strep-His than Abi (see Table 2).
  • SPR measurements of antibody affinity using Axl-Fc antigen indicated that Ab2 and Ab4 had higher affinity for Axl-Fc than Abi (see Table 2).
  • TABLE 2
    AXL-Strep-His antigen AXL-Fc antigen
    Antibody ka (1/Ms) kd (1/s) KD1 (nM) ka (1/Ms) kd (1/s) KD (nM)
    Ab1 1.31E+04 5.33E−04 40.7** 3.09E+04 1.42E−04 4.6
    Ab2 1.32E+04 4.41E−04 33.4  8.37E+04 1.54E−04 1.84
    Ab3 1.92E+04 6.36E−04 33.1  2.45E+04 2.00E−04 8.5
    Ab4 9759 5.90E−04 60.4  2.90E+05 3.96E−04 1.37
    Ab5 1.89E+04 4.35E−04 23    2.06E+04 1.59E−04 7.69
    **The observed binding data for 1H12 chimeric antibody did not fit monovalent or bivalent algorithms well, so in this one case, a “heterogeneous ligand” model was used. For all other binding data, the bivalent ligand model was used.
  • Abbreviations
    • 5 Å+ set The FW residues in the 5 Å CDR envelope, defined by the homology model, together with the canonical, vernier and VH/VK interface residues
    • 1H12 The anti-AXL mouse monoclonal antibody
    • 1H12 VK VK of mouse 1H12 antibody
    • 1H12RKA1 Humanised version, A1, of 1H12 VK
    • 1H12RHA Humanised version, A, of 1H12 VH
    • 1H12RHA x IgG1k antibody comprising the VH and VK constructs 1H12RHA and
    • 1H12RKA 1H12RKA respectively, functionally contiguous with the constant regions of human IgG1 and Ig-kappa heavy and light chains respectively
    • A Adenine
    • Å Angstrom
    • Ac acetyl
    • Acm acetamidomethyl
    • Alloc allyloxycarbonyl
    • B7 The anti-LPA antibody product of mouse hybridoma clone B7
    • Boc di-tert-butyl dicarbonate
    • bp base pairs
    • Bzl benzyl, where Bzl-OMe is methoxybenzyl and Bzl-Me is methylbenzene
    • C Cytosine
    • Cbz or Z benzyloxy-carbonyl, where Z—Cl and Z—Br are chloro- and bromobenzyloxy carbonyl respectively
    • CDR Complementarity determining region in the immunoglobulin variable regions, defined using the Kabat numbering system
    • CHO Chinese hamster ovary cell line
    • D-gene Diversity gene
    • DMF N,N-dimethylformamide
    • DNA Deoxyribonucleic acid
    • Dnp dinitrophenyl
    • DTT dithiothreitol
    • Fmoc 9H-fluoren-9-ylmethoxycarbonyl
    • FW Framework region: the immunoglobulin variable regions excluding the CDR regions
    • G Guanine
    • IgG Immunoglobulin G
    • imp N-10 imine protecting group: 3-(2-methoxyethoxy)propanoate-Val-Ala-PAB
    • MC-OSu maleimidocaproyl-O—N-succinimide
    • J-gene Joining gene
    • Kabat an immunoglobulin alignment and numbering system pioneered by Elvin A Kabat
    • mAb monoclonal antibody
    • Moc methoxycarbonyl
    • MP maleimidopropanamide
    • Mtr 4-methoxy-2,3,6-trimethtylbenzenesulfonyl
    • PAB para-aminobenzyloxycarbonyl
    • PEG ethyleneoxy
    • PNZ p-nitrobenzyl carbamate
    • Psec 2-(phenylsulfonyl)ethoxycarbonyl
    • T Thymine
    • TBDMS tert-butyldimethylsilyl
    • TBDPS tert-butyldiphenylsilyl
    • t-Bu tert-butyl
    • Teoc 2-(trimethylsilyl)ethoxycarbonyl
    • Tos tosyl
    • Troc 2,2,2-trichlorethoxycarbonyl chloride
    • Trt trityl
    • V region The segment of IgG chains which is variable in sequence between different antibodies. It extends to Kabat residue 109 in the light chain and 113 in the heavy chain.
    • VCI Framework residue classified as vernier or canonical or VH-VL interface
    • V-gene The gene segment that is rearranged, together with a J (and D for VH) gene, to generate a complete VK (or VH)
    • VH Immunoglobulin heavy chain variable region
    • VK Immunoglobulin kappa light chain variable region
    • Xan xanthyl
    REFERENCES
    • [1] C. Chothia, et al., “Domain association in immunoglobulin molecules. The packing of variable domains,” J Mol. Biol. 186(3), 651 (1985).
    • [2] J. Foote and G. Winter, “Antibody framework residues affecting the conformation of the hypervariable loops,” J Mol. Biol. 224(2), 487 (1992).
    • [3] E. A Kabat, et al., sequences of proteins of immunological interest, 5 ed. (NIH National Technical Information Service, 1991).
    • [4] V. Morea, A. M. Lesk, and A. Tramontano, “Antibody modeling: implications for engineering and design,” Methods 20(3), 267 (2000).
    STATEMENTS OF DISCLOSURE
  • 1. An isolated humanized antibody that binds to AXL, wherein the isolated humanized antibody comprises a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 2 or SEQ ID NO: 3.
    2. The isolated humanized antibody according to statement 1, wherein the isolated humanized antibody further comprises a light chain variable region having the amino acid sequence of SEQ ID NO: 4, 5, 6, 7, or 8; and, optionally, comprises a constant region derived from one or more human antibodies.
    3. The isolated humanized antibody according to either one of statements 1 or 2, wherein the isolated humanized antibody comprises:
    • (i) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
    • (ii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
    • (iii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
    • (iv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7;
    • (v) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8;
    • (vi) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
    • (vii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
    • (viii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
    • (ix) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7;
    • (x) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8;
    • (xi) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
    • (xii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
    • (xiii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
    • (xiv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7; or
    • (xv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8.
      4. The humanized antibody according to any one of statements 1 to 3, wherein said antibody binds human AXL with an affinity (Kd) of at least 10−6 M.
      5. The humanized antibody according to statement 3, wherein said antibody binds human AXL with an affinity (Kd) of at least 10−9 M.
      6. The humanized antibody according to any one of statements 1 to 5, wherein said antibody competitively inhibits the binding to human AXL of an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 1 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4.
      7. The humanized antibody according to any one of statements 1 to 6, wherein said antibody binds the Axl-Strep-His antigen with an affinity (Kd) of no more 0.6 of the Kd of an antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies.
      8. The humanized antibody according to any one of statements 1 to 7, wherein said antibody binds the Axl-Fc antigen with an affinity (Kd) of no more 0.5 of the Kd of an antibody comprising a VH domain having the sequence according to SEQ ID NO. 1, a VL domain having the sequence according to SEQ ID NO. 4, and a constant region derived from one or more human antibodies.
      9. The humanized antibody according to any one of statements 1 to 8, wherein said antibody competitively inhibits the binding to human AXL of the mouse 1H12 antibody.
      10. The humanized antibody according to any one of statements 1 to 9, wherein said antibody has a pI of at least 8.00.
      11. The humanized antibody according to statement 10 wherein the antibody has a pI of at least 8.15.
      12. The humanized antibody according to any one of statements 1 to 11, wherein said antibody or antibody fragment has a constant region of either isotype IgG1, IgG2, IgG3 or IgG4, or a mutated IgG constant region, and optionally a light chain constant region of isotype kappa or lambda.
      13. The humanized antibody according to any one of statements 1 to 12, wherein the humanized antibody fragment is a scFv, Fab or F(ab′)2.
      14. The antibody according to any one of statements 1 to 13, for use in therapy.
      15. The antibody according to any one of statements 1 to 14, for use in the treatment of a proliferative disease in a subject.
      16. The antibody according to any one of statements 1 to 15, for use in the treatment of a proliferative disease in a subject, wherein the subject has raised levels of AXL, Akt3, or GAS6 and wherein the method comprises identifying that the subject has raised levels of AXL, Akt3, or GAS6 and administering the antibody or conjugate to the patient.
      17. The antibody or drug-conjugate according to any one of statements 1 to 16, for use in the treatment of a proliferative disease in a subject, wherein the proliferative disease is associated with raised levels of AXL, Akt3, or GAS6, the method comprising administering the conjugate to the patient.
      18. The drug-conjugate according to any one of statements 15 to 17, wherein the disease is cancer.
      19. A pharmaceutical composition comprising the antibody of any one of statements 1 to 13 and a pharmaceutically acceptable diluent, carrier or excipient.
      20. The pharmaceutical composition of statement 19 further comprising a therapeutically effective amount of a chemotherapeutic agent.
      21. Use of an antibody according to any one of statements 1 to 13 in the preparation of a medicament for use in the treatment of a proliferative disease in a subject.
      22. A method of treating cancer comprising administering to a patient the pharmaceutical composition according to either one of statements 20 or 21.
      23. The method of statement 22 wherein the patient is administered a chemotherapeutic agent, in combination with the composition.
      24. A polynucleotide encoding a humanized antibody according to any one of statements 1 to 13.
      25. A vector comprising the polynucleotide of statement 24.
      26. The vector of statement 25 wherein the vector is an expression vector.
      27. A host cell comprising a vector according to either one of statements 25 or 26.
      28. The host cell according to statement 27 wherein the host cell is prokaryotic, eukaryotic, or mammalian.
      29. A method of selecting an individual for treatment with the according to any one of statements 1 to 13, or with the pharmaceutical composition of either one of statements 20 or 21, which method comprises assessing the level of AXL;
      • wherein individuals having raised levels of AXL are selected for treatment.
        30. A method of timing the application of treatment of an individual with the antibody or drug-conjugate according to any one of statements 1 to 13, or with the pharmaceutical composition of either one of statements 20 or 21, which method comprises assessing the level of AXL;
      • wherein the treatment is applied if the individual has raised levels of AXL.
        31. The method according to either one of statements 29 or 30, wherein the individual has cancer and treatment reduces tumour volume.
  • SEQUENCES
    [1H12VH]
    SEQ ID NO: 1 
    MGFKMESQFQVFVFVFLWLSGVDGEVQLVESGGDLVKPGGSLKLSCAASG
    FTFSSYGMSWVRQTPDKRLEWVATISSGGSYTYYPDSVKGRFTISRDNAK
    NTLYLQMSSLKSEDTAMYYCARHPIYYTYDDTMDYWGQGTSVTVSS
    [1H12RHA]
    SEQ ID NO: 2
    MGFKMESQFQVFVFVFLWLSGVDGQVQLVESGGGVVQPGRSLRLSCAASG
    FTFSSYGMSWVRQAPGKGLEWVATISSGGSYTYYPDSVKGRFTISRDNSK
    NTLYLQMNSLRAEDTAVYYCARHPIYYTYDDTMDYWGQGTTVTVSS
    [1H12RHB]
    SEQ ID NO: 3
    MGFKMESQFQVFVFVFLWLSGVDGEVQLVESGGGLVQPGGSLRLSCAASG
    FTFSSYGMSWVRQAPGKGLEWVATISSGGSYTYYPDSVKGRFTISRDNAK
    NSLYLQMNSLRAEDTAVYYCARHPIYYTYDDTMDYWGQGTLVTVSS
    [1H12VK]
    SEQ ID NO: 4
    MGFKMESQFQVFVFVFLWLSGVDGENVLTQSPAIMAASPGEKVTMTCSAS
    SSVSSGNFHWYQQKPGTSPKLWIYRTSNLASGVPARFSGSGSGTSYSLTI
    SSMEAEDAATYYCQQWSGYPWTFGGGTKLEIK
    [1H12RKA]
    SEQ ID NO: 5
    MGFKMESQFQVFVFVFLWLSGVDGEIVLTQSPATLSLSPGERATLSCSAS
    SSVSSGNFHWYQQKPGLAPRLLIYRTSNLASGIPDRFSGSGSGTDFTLTI
    SRLEPEDFAVYYCQQWSGYPWTFGPGTKVDIK
    [1H12RKA1]
    SEQ ID NO: 6
    MGFKMESQFQVFVFVFLWLSGVDGENVLTQSPATLSLSPGERATLSCSAS
    SSVSSGNFHWYQQKPGLAPRLWIYRTSNLASGIPDRFSGSGSGTDYTLTI
    SRLEPEDFAVYYCQQWSGYPWTFGPGTKVDIK
    [1H12RKB]
    SEQ ID NO: 7
    MGFKMESQFQVFVFVFLWLSGVDGEIVLTQSPGTLSLSPGERATLSCSAS
    SSVSSGNFHWYQQKPGLAPRLLIYRTSNLASGIPARFSGSGSGTDFTLTI
    SSLEPEDFAVYYCQQWSGYPWTFGGGTKLEIK
    [1H12RKB1]
    SEQ ID NO: 8
    MGFKMESQFQVFVFVFLWLSGVDGENVLTQSPGTLSLSPGERATLSCSAS
    SSVSSGNFHWYQQKPGLAPRLWIYRTSNLASGIPARFSGSGSGTDYTLTI
    SSLEPEDFAVYYCQQWSGYPWTFGGGTKLEIK
    [Human Axl]
    SEQ ID NO: 9
    MAWRCPRMGRVPLAWCLALCGWACMAPRGTQAEESPFVGNPGNITGARGL
    TGTLRCQLQVQGEPPEVHWLRDGQILELADSTQTQVPLGEDEQDDWIVVS
    QLRITSLQLSDTGQYQCLVFLGHQTFVSQPGYVGLEGLPYFLEEPEDRTV
    AANTPFNLSCQAQGPPEPVDLLWLQDAVPLATAPGHGPQRSLHVPGLNKT
    SSFSCEAHNAKGVTTSRTATITVLPQQPRNLHLVSRQPTELEVAWTPGLS
    GIYPLTHCTLQAVLSDDGMGIQAGEPDPPEEPLTSQASVPPHQLRLGSLH
    PHTPYHIRVACTSSQGPSSWTHWLPVETPEGVPLGPPENISATRNGSQAF
    VHWQEPRAPLQGTLLGYRLAYQGQDTPEVLMDIGLRQEVTLELQGDGSVS
    NLTVCVAAYTAAGDGPWSLPVPLEAWRPGQAQPVHQLVKEPSTPAFSWPW
    WYVLLGAVVAAACVLILALFLVHRRKKETRYGEVFEPTVERGELVVRYRV
    RKSYSRRTTEATLNSLGISEELKEKLRDVMVDRHKVALGKTLGEGEFGAV
    MEGQLNQDDSILKVAVKTMKIAICTRSELEDFLSEAVCMKEFDHPNVMRL
    IGVCFQGSERESFPAPVVILPFMKHGDLHSFLLYSRLGDQPVYLPTQMLV
    KFMADIASGMEYLSTKRFIHRDLAARNCMLNENMSVCVADFGLSKKIYNG
    DYYRQGRIAKMPVKWIAIESLADRVYTSKSDVWSFGVTMWEIATRGQTPY
    PGVENSEIYDYLRRGNRLKQPADCLDGLYALMSRCWELNPQDRPSFTELR
    EDLENTLKALPPAQEPDEILYVNMDEGGGYPEPPGAAGGADPPTQPDPKD
    SCSCLTAAEVHPAGRYVLCPSTTPSPAQPADRGSPAAPGQEDGA
    [Murine Axl]
    SEQ ID NO: 10
    MGRVPLAWWLALCCWGCAAHKDTQTEAGSPFVGNPGNITGARGLTGTLRC
    ELQVQGEPPEVVWLRDGQILELADNTQTQVPLGEDWQDEWKVVSQLRISA
    LQLSDAGEYQCMVHLEGRTFVSQPGFVGLEGLPYFLEEPEDKAVPANTPF
    NLSCQAQGPPEPVTLLWLQDAVPLAPVTGHSSQHSLQTPGLNKTSSFSCE
    AHNAKGVTTSRTATITVLPQRPHHLHVVSRQPTELEVAWTPGLSGIYPLT
    HCNLQAVLSDDGVGIWLGKSDPPEDPLTLQVSVPPHQLRLEKLLPHTPYH
    IRISCSSSQGPSPWTHWLPVETTEGVPLGPPENVSAMRNGSQVLVRWQEP
    RVPLQGTLLGYRLAYRGQDTPEVLMDIGLTREVTLELRGDRPVANLTVSV
    TAYTSAGDGPWSLPVPLEPWRPGQGQPLHHLVSEPPPRAFSWPWWYVLLG
    ALVAAACVLILALFLVHRRKKETRYGEVFEPTVERGELVVRYRVRKSYSR
    RTTEATLNSLGISEELKEKLRDVMVDRHKVALGKTLGEGEFGAVMEGQLN
    QDDSILKVAVKTMKIAICTRSELEDFLSEAVCMKEFDHPNVMRLIGVCFQ
    GSDREGFPEPVVILPFMKHGDLHSFLLYSRLGDQPVFLPTQMLVKFMADI
    ASGMEYLSTKRFIHRDLAARNCMLNENMSVCVADFGLSKKIYNGDYYRQG
    RIAKMPVKWIAIESLADRVYTSKSDVWSFGVTMWEIATRGQTPYPGVENS
    EIYDYLRQGNRLKQPVDCLDGLYALMSRCWELNPRDRPSFAELREDLENT
    LKALPPAQEPDEILYVNMDEGGSHLEPRGAAGGADPPTQPDPKDSCSCLT
    AADVHSAGRYVLCPSTAPGPTLSADRGCPAPPGQEDGA

Claims (15)

1.-31. (canceled)
32. An isolated humanized antibody that binds to AXL, wherein the isolated humanized antibody comprises:
a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 or SEQ ID NO: 3; and
a light chain variable region having the amino acid sequence of SEQ ID NO: 4, 5, 6, 7, or 8.
33. The isolated humanized antibody according to claim 32, wherein the isolated humanized antibody comprises:
(i) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
(ii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
(iii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
(iv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7;
(v) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 2 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8;
(vi) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 4;
(vii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 5;
(viii) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 6;
(viv) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 7; or
(x) a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3 and a light chain variable region having the amino acid sequence of SEQ ID NO: 8.
34. The humanized antibody according to claim 32, wherein said antibody or antibody fragment has a constant region of isotype IgG1, IgG2, IgG3 or IgG4, or a mutated IgG constant region.
35. The humanized antibody according to claim 32, wherein said antibody or antibody fragment has a light chain constant region of isotype kappa or lambda.
36. The humanized antibody according to claim 32, wherein the humanized antibody fragment is a scFv, Fab or F(ab)2.
37. A method of treating a proliferative disease in a subject, comprising administering to the subject an antibody according to claim 32.
38. A polynucleotide encoding a humanized antibody according to claim 32.
39. A vector comprising the polynucleotide of claim 38.
40. The vector of claim 39 wherein the vector is an expression vector.
41. A host cell comprising a vector according to claim 39.
42. The host cell according to claim 41 wherein the host cell is prokaryotic, eukaryotic, or mammalian.
43. A method of making a humanized antibody that binds to AXL, the method comprising culturing the host cell according to claim 41 under conditions for production of said antibody.
44. The method according to claim 43, further comprising isolating said antibody.
45. The method according to claim 44, further comprising formulating the antibody into a composition including at least one additional component.
US17/372,961 2015-04-15 2021-07-12 Humanized anti-axl antibodies Pending US20220073625A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/372,961 US20220073625A1 (en) 2015-04-15 2021-07-12 Humanized anti-axl antibodies

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GB1506411.6 2015-04-15
GBGB1506411.6A GB201506411D0 (en) 2015-04-15 2015-04-15 Humanized anti-axl antibodies
PCT/EP2016/058368 WO2016166296A2 (en) 2015-04-15 2016-04-15 Humanized anti-axl antibodies
US201715566635A 2017-10-13 2017-10-13
US17/372,961 US20220073625A1 (en) 2015-04-15 2021-07-12 Humanized anti-axl antibodies

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US15/566,635 Continuation US11059893B2 (en) 2015-04-15 2016-04-15 Humanized anti-AXL antibodies
PCT/EP2016/058368 Continuation WO2016166296A2 (en) 2015-04-15 2016-04-15 Humanized anti-axl antibodies

Publications (1)

Publication Number Publication Date
US20220073625A1 true US20220073625A1 (en) 2022-03-10

Family

ID=53333841

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/566,635 Active 2036-10-27 US11059893B2 (en) 2015-04-15 2016-04-15 Humanized anti-AXL antibodies
US17/372,961 Pending US20220073625A1 (en) 2015-04-15 2021-07-12 Humanized anti-axl antibodies

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/566,635 Active 2036-10-27 US11059893B2 (en) 2015-04-15 2016-04-15 Humanized anti-AXL antibodies

Country Status (4)

Country Link
US (2) US11059893B2 (en)
EP (1) EP3283525A2 (en)
GB (1) GB201506411D0 (en)
WO (1) WO2016166296A2 (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201410826D0 (en) * 2014-06-18 2014-07-30 Bergenbio As Anti-axl antibodies
US20210215697A1 (en) 2018-05-14 2021-07-15 Bergenbio Asa Serum Biomarkers
CN110483639A (en) 2018-05-15 2019-11-22 复旦大学 Target antibody and the antibody-drug conjugates and its preparation method and application of AXL
WO2021013746A1 (en) 2019-07-19 2021-01-28 Genmab A/S Axl antibody-drug conjugates for use in treating cancer
GB201912059D0 (en) 2019-08-22 2019-10-09 Bergenbio As Combaination therapy of a patient subgroup
GB202004189D0 (en) 2020-03-23 2020-05-06 Bergenbio As Combination therapy
AU2021252094A1 (en) 2020-04-08 2022-11-10 Bergenbio Asa AXL inhibitors for antiviral therapy
GB202006072D0 (en) 2020-04-24 2020-06-10 Bergenbio Asa Method of selecting patients for treatment with cmbination therapy
GB202104037D0 (en) 2021-03-23 2021-05-05 Bergenbio Asa Combination therapy
WO2022269605A1 (en) 2021-06-24 2022-12-29 Yeda Research And Development Co. Ltd. Combination therapy for the treatment of cancer comprising an anti-egfr antibody and an axl-inhibitor

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9975954B2 (en) * 2014-06-18 2018-05-22 Bergenbio Asa Anti-Axl antibodies

Family Cites Families (423)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3361742A (en) 1964-12-07 1968-01-02 Hoffmann La Roche 5-oxo-1h-pyrrolo-[2, 1-c][1, 4]-benzodiazepin-2-crylamides
US3523941A (en) 1967-03-06 1970-08-11 Hoffmann La Roche Benzodiazepine compounds and process for their preparation
US3524849A (en) 1967-10-27 1970-08-18 Hoffmann La Roche Process for the preparation of pyrrolo-benzodiazepine acrylamides and intermediates useful therein
JPS4843755B1 (en) 1969-06-26 1973-12-20
IL33558A (en) 1968-12-30 1973-10-25 Fujisawa Pharmaceutical Co Antibiotic pyrrolo-benzodiazepine compound,its derivatives and processes for their production
FR2027356A1 (en) 1968-12-30 1970-09-25 Fujisawa Pharmaceutical Co Benzodiazepinone antibiotics
GB1410826A (en) 1972-10-30 1975-10-22 Nakao K Method of and apparatus for disposing of broken pieces of fragile material
JPS585916B2 (en) 1977-12-27 1983-02-02 株式会社ミドリ十字 New benzodiazepine compounds
JPS5615289A (en) 1979-07-17 1981-02-14 Green Cross Corp:The Novel benzodiazepinnbased compound 3
JPS57131791A (en) 1980-12-31 1982-08-14 Fujisawa Pharmaceut Co Ltd Benzodiazepine derivative and its preparation
CA1185602A (en) 1981-02-27 1985-04-16 Emilio Kyburz Imidazodiazepines
CA1184175A (en) 1981-02-27 1985-03-19 Walter Hunkeler Imidazodiazepines
CA1173441A (en) 1981-02-27 1984-08-28 Hoffmann-La Roche Limited Imidazodiazepines
JPS58180487A (en) 1982-04-16 1983-10-21 Kyowa Hakko Kogyo Co Ltd Antibiotic dc-81 and its preparation
US4427588A (en) 1982-11-08 1984-01-24 Bristol-Myers Company Process for conversion of oxotomaymycin to tomaymycin
US4427587A (en) 1982-11-10 1984-01-24 Bristol-Myers Company Total synthesis of antitumor antibiotics BBM-2040A and BBM-2040B
JPS59152329A (en) 1983-02-17 1984-08-31 Green Cross Corp:The Local disorder inhibitor
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
FR2586683B1 (en) 1985-08-29 1988-07-01 Centre Nat Rech Scient NOVEL NEOTHRAMYCIN DERIVATIVES, THEIR PREPARATION PROCESS AND THEIR APPLICATION AS MEDICAMENTS
US5583024A (en) 1985-12-02 1996-12-10 The Regents Of The University Of California Recombinant expression of Coleoptera luciferase
DE3783588T2 (en) 1986-04-17 1993-06-09 Kyowa Hakko Kogyo Kk NEW DC-88A AND DC-89A1 CONNECTIONS AND THEIR PRODUCTION METHOD.
JP2660201B2 (en) 1988-08-05 1997-10-08 塩野義製薬株式会社 Novel pyrrolo [1,4] benzodiazepine derivatives and senile dementia drugs
WO1991002536A1 (en) 1989-08-23 1991-03-07 Scripps Clinic And Research Foundation Compositions and methods for detection and treatment of epstein-barr virus infection and immune disorders
JPH053790A (en) 1990-04-19 1993-01-14 Fujisawa Pharmaceut Co Ltd Dehydropeptidase-i
US5256643A (en) 1990-05-29 1993-10-26 The Government Of The United States Human cripto protein
AU9016591A (en) 1990-10-25 1992-05-26 Tanox Biosystems, Inc. Glycoproteins associated with membrane-bound immunoglobulins as antibody targets on B cells
ATE240740T1 (en) 1991-03-15 2003-06-15 Amgen Inc PEGYLATION OF POLYPEPTIDES
US5440021A (en) 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
US5543503A (en) 1991-03-29 1996-08-06 Genentech Inc. Antibodies to human IL-8 type A receptor
JP4156662B2 (en) 1991-03-29 2008-09-24 ジェネンテック・インコーポレーテッド Human PF4A receptor and use thereof
FR2676230B1 (en) 1991-05-07 1993-08-27 Centre Nat Rech Scient NOVEL PYRROLO [1,4] -BENZODIAZEPINES DERIVATIVES, PROCESS FOR THEIR PREPARATION AND MEDICAMENTS CONTAINING THEM.
US5468634A (en) 1991-06-24 1995-11-21 The University Of North Carolina At Chapel Hill Axl oncogene
JP3050424B2 (en) 1991-07-12 2000-06-12 塩野義製薬株式会社 Human endothelin receptor
US5264557A (en) 1991-08-23 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Polypeptide of a human cripto-related gene, CR-3
US5362852A (en) 1991-09-27 1994-11-08 Pfizer Inc. Modified peptide derivatives conjugated at 2-hydroxyethylamine moieties
US6011146A (en) 1991-11-15 2000-01-04 Institut Pasteur Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
US6153408A (en) 1991-11-15 2000-11-28 Institut Pasteur And Institut National De La Sante Et De La Recherche Medicale Altered major histocompatibility complex (MHC) determinant and methods of using the determinant
GB9205051D0 (en) 1992-03-09 1992-04-22 Cancer Res Campaign Tech Pyrrolobenzodiazepine derivatives,their preparation,and compositions containing them
FR2696176B1 (en) 1992-09-28 1994-11-10 Synthelabo Piperidine derivatives, their preparation and their therapeutic application.
IL107366A (en) 1992-10-23 2003-03-12 Chugai Pharmaceutical Co Ltd Genes coding for megakaryocyte potentiator
US5644033A (en) 1992-12-22 1997-07-01 Health Research, Inc. Monoclonal antibodies that define a unique antigen of human B cell antigen receptor complex and methods of using same for diagnosis and treatment
US5869445A (en) 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
US5801005A (en) 1993-03-17 1998-09-01 University Of Washington Immune reactivity to HER-2/neu protein for diagnosis of malignancies in which the HER-2/neu oncogene is associated
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
GB9316162D0 (en) 1993-08-04 1993-09-22 Zeneca Ltd Fungicides
US5773223A (en) 1993-09-02 1998-06-30 Chiron Corporation Endothelin B1, (ETB1) receptor polypeptide and its encoding nucleic acid methods, and uses thereof
EP0647450A1 (en) 1993-09-09 1995-04-12 BEHRINGWERKE Aktiengesellschaft Improved prodrugs for enzyme mediated activation
JPH09506250A (en) 1993-11-23 1997-06-24 ジェネンテク,インコーポレイテッド Protein tyrosine kinase named Rse
EP0739488A1 (en) 1994-01-14 1996-10-30 Genentech, Inc. Antagonists to insulin receptor tyrosine kinase inhibitor
US5538861A (en) 1994-07-29 1996-07-23 Amgen Inc. DNA encoding a stimulating factor for the axl receptor
US5750370A (en) 1995-06-06 1998-05-12 Human Genome Sciences, Inc. Nucleic acid encoding human endothlein-bombesin receptor and method of producing the receptor
US6211142B1 (en) 1995-03-10 2001-04-03 Genentech, Inc. Compositions comprising gas6 polypeptides and articles of manufacture comprising the same
JPH08336393A (en) 1995-04-13 1996-12-24 Mitsubishi Chem Corp Production of optically active gamma-substituted-beta-hydroxybutyric ester
US5707829A (en) 1995-08-11 1998-01-13 Genetics Institute, Inc. DNA sequences and secreted proteins encoded thereby
US20020193567A1 (en) 1995-08-11 2002-12-19 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
JP3646191B2 (en) 1996-03-19 2005-05-11 大塚製薬株式会社 Human gene
US6218519B1 (en) 1996-04-12 2001-04-17 Pro-Neuron, Inc. Compounds and methods for the selective treatment of cancer and bacterial infections
CA2254843A1 (en) 1996-05-17 1997-11-27 Schering Corporation Human b-cell antigens, related reagents
EP0961619A4 (en) 1996-09-27 2001-09-26 Bristol Myers Squibb Co Hydrolyzable prodrugs for delivery of anticancer drugs to metastatic cells
US6759509B1 (en) 1996-11-05 2004-07-06 Bristol-Myers Squibb Company Branched peptide linkers
US5945511A (en) 1997-02-20 1999-08-31 Zymogenetics, Inc. Class II cytokine receptor
US7033827B2 (en) 1997-02-25 2006-04-25 Corixa Corporation Prostate-specific polynucleotide compositions
US20030185830A1 (en) 1997-02-25 2003-10-02 Corixa Corporation Compositions and methods for the therapy and diagnosis of prostate cancer
DE69824287T2 (en) 1997-03-10 2005-06-16 The Regents Of The University Of California, Oakland PROSTATE STEM CELL ANTIGEN (PSCA)
US6541212B2 (en) 1997-03-10 2003-04-01 The Regents Of The University Of California Methods for detecting prostate stem cell antigen protein
US6261791B1 (en) 1997-03-10 2001-07-17 The Regents Of The University Of California Method for diagnosing cancer using specific PSCA antibodies
US6555339B1 (en) 1997-04-14 2003-04-29 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human protein-coupled receptors
US6319688B1 (en) 1997-04-28 2001-11-20 Smithkline Beecham Corporation Polynucleotide encoding human sodium dependent phosphate transporter (IPT-1)
US6890749B2 (en) 1997-05-15 2005-05-10 Abbott Laboratories Reagents and methods useful for detecting diseases of the prostate
WO1998051824A1 (en) 1997-05-15 1998-11-19 Abbott Laboratories Reagents and methods useful for detecting disease of the urinary tract
US6235769B1 (en) 1997-07-03 2001-05-22 Sugen, Inc. Methods of preventing and treating neurological disorders with compounds that modulate the function of the C-RET receptor protein tyrosine kinase
US6602677B1 (en) 1997-09-19 2003-08-05 Promega Corporation Thermostable luciferases and methods of production
US20030060612A1 (en) 1997-10-28 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20020034749A1 (en) 1997-11-18 2002-03-21 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
US6110695A (en) 1997-12-02 2000-08-29 The Regents Of The University Of California Modulating the interaction of the chemokine, B Lymphocyte Hemoattractant, and its Receptor, BLR1
AU762991B2 (en) 1998-03-13 2003-07-10 Burnham Institute, The Molecules that home to various selected organs or tissues
AU3375199A (en) 1998-04-01 1999-10-18 Genentech Inc. Antagonists to growth arrest specific gene 6 to treat insulin-resistant disorders
JP2002520000A (en) 1998-05-13 2002-07-09 エピミューン, インコーポレイテッド Expression vector and method of using the vector to stimulate an immune response
US20030064397A1 (en) 1998-05-22 2003-04-03 Incyte Genomics, Inc. Transmembrane protein differentially expressed in prostate and lung tumors
US6835807B1 (en) 1998-05-22 2004-12-28 Daiichi Pharmaceuticals Co., Ltd. Drug complex and drug delivery system
US20020187472A1 (en) 2001-03-09 2002-12-12 Preeti Lal Steap-related protein
AU5094699A (en) 1998-07-08 2000-02-01 E-Ink Corporation Methods for achieving improved color in microencapsulated electrophoretic devices
EP1413582B1 (en) 1998-08-27 2006-03-15 Spirogen Limited Dimeric pyrrolobenzodiazepines
GB9818732D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Collection of compounds
GB9818730D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Collections of compounds
GB9818731D0 (en) 1998-08-27 1998-10-21 Univ Portsmouth Compounds
WO2000012130A1 (en) 1998-08-27 2000-03-09 Smithkline Beecham Corporation Rp105 agonists and antagonists
JP4689781B2 (en) 1998-09-03 2011-05-25 独立行政法人科学技術振興機構 Amino acid transport protein and its gene
WO2000020579A1 (en) 1998-10-02 2000-04-13 Mcmaster University Spliced form of erbb-2/neu oncogene
WO2001057188A2 (en) 2000-02-03 2001-08-09 Hyseq, Inc. Novel nucleic acids and polypeptides
US20020119158A1 (en) 1998-12-17 2002-08-29 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US6468546B1 (en) 1998-12-17 2002-10-22 Corixa Corporation Compositions and methods for therapy and diagnosis of ovarian cancer
US6962980B2 (en) 1999-09-24 2005-11-08 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US6858710B2 (en) 1998-12-17 2005-02-22 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030091580A1 (en) 2001-06-18 2003-05-15 Mitcham Jennifer L. Compositions and methods for the therapy and diagnosis of ovarian cancer
EP1141017B1 (en) 1998-12-30 2008-09-10 Beth Israel Deaconess Medical Center, Inc. Characterization of the soc/crac calcium channel protein family
US20030190669A1 (en) 1998-12-30 2003-10-09 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
CN1201004C (en) 1999-01-29 2005-05-11 考丽克萨有限公司 HER-2/neu fusion proteins
GB9905124D0 (en) 1999-03-05 1999-04-28 Smithkline Beecham Biolog Novel compounds
AU3395900A (en) 1999-03-12 2000-10-04 Human Genome Sciences, Inc. Human lung cancer associated gene sequences and polypeptides
US7312303B2 (en) 1999-05-11 2007-12-25 Genentech, Inc. Anti-PRO4980 antibodies
US6268488B1 (en) 1999-05-25 2001-07-31 Barbas, Iii Carlos F. Prodrug activation using catalytic antibodies
AU4952600A (en) 1999-06-03 2000-12-28 Takeda Chemical Industries Ltd. Screening method with the use of cd100
EP1185635A2 (en) 1999-06-10 2002-03-13 D. Collen Research Foundation vzw Non-human transgenic animals deficient in gas6 function and their use
EP2283867B1 (en) 1999-06-25 2014-05-21 ImmunoGen, Inc. Methods of treatment using anti-ERBB antibody-maytansinoid conjugates
US6302318B1 (en) 1999-06-29 2001-10-16 General Electric Company Method of providing wear-resistant coatings, and related articles
US20030119113A1 (en) 1999-07-20 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7297770B2 (en) 1999-08-10 2007-11-20 Genentech, Inc. PRO6496 polypeptides
US7294696B2 (en) 1999-08-17 2007-11-13 Genentech Inc. PRO7168 polypeptides
US6909006B1 (en) 1999-08-27 2005-06-21 Spirogen Limited Cyclopropylindole derivatives
CA2382812A1 (en) 1999-08-30 2001-03-08 Joseph Schlessinger Crystal structures of domains of receptor protein tyrosine kinases and their ligands
AU7573000A (en) 1999-09-01 2001-03-26 Genentech Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030206918A1 (en) 1999-09-10 2003-11-06 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030129192A1 (en) 1999-09-10 2003-07-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US20030232056A1 (en) 1999-09-10 2003-12-18 Corixa Corporation Compositions and methods for the therapy and diagnosis of ovarian cancer
US6750054B2 (en) 2000-05-18 2004-06-15 Lexicon Genetics Incorporated Human semaphorin homologs and polynucleotides encoding the same
EP1226177B1 (en) 1999-10-29 2008-07-09 Genentech, Inc. Anti-prostate stem cell antigen (psca) antibody compositions and methods of use
WO2001032926A2 (en) 1999-11-01 2001-05-10 Curagen Corporation Differentially expressed genes involved in angiogenesis, the polypeptides encoded thereby, and methods of using the same
EP2404927B1 (en) 1999-11-29 2016-05-11 The Trustees of Columbia University in the City of New York Isolation of five novel genes coding for new fc receptors-type melanoma involved in the pathogenesis of lymphoma/melanoma
EP1248800A2 (en) 1999-11-30 2002-10-16 Corixa Corporation Compositions and methods for therapy and diagnosis of breast cancer
CA2393738A1 (en) 1999-12-10 2001-06-14 Epimmune Inc. Inducing cellular immune responses to her2/neu using peptide and nucleic acid compositions
EP2295078A3 (en) 1999-12-23 2011-03-23 ZymoGenetics, L.L.C. Method for treating inflammation
DK1246846T3 (en) 1999-12-23 2008-12-08 Zymogenetics Inc Soluble interleukin-20 receptor
US6610286B2 (en) 1999-12-23 2003-08-26 Zymogenetics, Inc. Method for treating inflammation using soluble receptors to interleukin-20
NZ502058A (en) 1999-12-23 2003-11-28 Ovita Ltd Isolated mutated nucleic acid molecule for regulation of ovulation rate
US20040001827A1 (en) 2002-06-28 2004-01-01 Dennis Mark S. Serum albumin binding peptides for tumor targeting
WO2001045746A2 (en) 1999-12-24 2001-06-28 Genentech, Inc. Methods and compositions for prolonging elimination half-times of bioactive compounds
US7294695B2 (en) 2000-01-20 2007-11-13 Genentech, Inc. PRO10268 polypeptides
WO2001053463A2 (en) 2000-01-21 2001-07-26 Corixa Corporation COMPOUNDS AND METHODS FOR PREVENTION AND TREATMENT OF HER-2/neu ASSOCIATED MALIGNANCIES
US20030224379A1 (en) 2000-01-21 2003-12-04 Tang Y. Tom Novel nucleic acids and polypeptides
AU2001238596A1 (en) 2000-02-22 2001-09-03 Millennium Pharmaceuticals, Inc. 18607, a novel human calcium channel
US20030219806A1 (en) 2000-02-22 2003-11-27 Millennium Pharmaceuticals, Inc. Novel 18607, 15603, 69318, 12303, 48000, 52920, 5433, 38554, 57301, 58324, 55063, 52991, 59914, 59921 and 33751 molecules and uses therefor
US20040052793A1 (en) 2001-02-22 2004-03-18 Carter Paul J. Caspase activivated prodrugs therapy
US20040002068A1 (en) 2000-03-01 2004-01-01 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US20040005561A1 (en) 2000-03-01 2004-01-08 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
AU2001245280A1 (en) 2000-03-07 2001-09-17 Hyseq, Inc. Novel nucleic acids and polypeptides
AU2001249411B2 (en) 2000-03-24 2007-02-15 Fahri Saatcioglu Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods
AU2001250412A1 (en) 2000-03-31 2001-10-08 Ipf Pharmaceuticals Gmbh Diagnostic and medicament for analysing the cell surface proteome of tumour and inflammatory cells and for treating tumorous and inflammatory diseases, preferably using specific chemokine receptor analysis and the chemokine receptor-ligand interaction
US7279294B2 (en) 2000-04-03 2007-10-09 The United States Of America As Represented By The Secretary, Dept. Of Health And Human Services, Nih Tumor markers in ovarian cancer
CA2402392A1 (en) 2000-04-07 2001-10-18 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated known g protein-coupled receptors
US20030119115A1 (en) 2000-05-17 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO2001090304A2 (en) 2000-05-19 2001-11-29 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
US20020051990A1 (en) 2000-06-09 2002-05-02 Eric Ople Novel gene targets and ligands that bind thereto for treatment and diagnosis of ovarian carcinomas
WO2001098351A2 (en) 2000-06-16 2001-12-27 Incyte Genomics, Inc. G-protein coupled receptors
JP2004502414A (en) 2000-06-30 2004-01-29 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド B7-like polynucleotides, polypeptides and antibodies
EP1294885A2 (en) 2000-06-30 2003-03-26 Amgen, Inc. B7-like molecules and uses thereof
WO2002002634A2 (en) 2000-06-30 2002-01-10 Incyte Genomics, Inc. Human extracellular matrix and cell adhesion polypeptides
WO2002006339A2 (en) 2000-07-03 2002-01-24 Curagen Corporation Proteins and nucleic acids encoding same
US20040044179A1 (en) 2000-07-25 2004-03-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6891030B2 (en) 2000-07-27 2005-05-10 Mayo Foundation For Medical Education And Research T-cell immunoregulatory molecule
US7205108B2 (en) 2000-07-28 2007-04-17 Ulrich Wissenbach Trp8, Trp9 and Trp10, novel markers for cancer
US7229623B1 (en) 2000-08-03 2007-06-12 Corixa Corporation Her-2/neu fusion proteins
AU2001283360A1 (en) 2000-08-14 2002-02-25 Corixa Corporation Methods for diagnosis and therapy of hematological and virus-associated malignancies
PL365789A1 (en) 2000-08-14 2005-01-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of her-2/neu-associated malignancies
GB0020953D0 (en) 2000-08-24 2000-10-11 Smithkline Beecham Biolog Vaccine
EP1445317A3 (en) 2000-08-24 2004-12-15 Genentech Inc. Compositions and methods for the diagnosis and treatment of tumor
WO2002022660A2 (en) 2000-09-11 2002-03-21 Hyseq, Inc. Novel nucleic acids and polypeptides
US6613567B1 (en) 2000-09-15 2003-09-02 Isis Pharmaceuticals, Inc. Antisense inhibition of Her-2 expression
ATE333888T1 (en) 2000-09-15 2006-08-15 Zymogenetics Inc USE OF A POLYPEPTIDE CONTAINING THE EXTRACELLULAR DOMAIN OF IL-20RA AND IL-20RB FOR THE TREATMENT OF INFLAMMATION
US20030119121A1 (en) 2000-09-15 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
UA83458C2 (en) 2000-09-18 2008-07-25 Байоджен Айдек Ма Інк. The isolated polypeptide baff-r (the receptor of the factor of activation of b-cells of the family tnf)
CA2422814A1 (en) 2000-09-18 2002-03-21 Biogen, Inc. Cripto mutant and uses thereof
ES2254492T3 (en) 2000-09-19 2006-06-16 Moses Lee COMPOSITIONS AND PROCEDURES FOR THE USE OF AQUIRAL ANALOGS OF CC-1065 AND DUOCARMYCINES.
EP1474528A4 (en) 2000-10-13 2006-06-14 Protein Design Labs Inc Methods of diagnosis of prostate cancer, compositions and methods of screening for modulators of prostate cancer
AU2002230659A1 (en) 2000-11-07 2002-05-21 Zymogenetics Inc. Human tumor necrosis factor receptor
US20020150573A1 (en) 2000-11-10 2002-10-17 The Rockefeller University Anti-Igalpha-Igbeta antibody for lymphoma therapy
US20040018194A1 (en) 2000-11-28 2004-01-29 Francisco Joseph A. Recombinant anti-CD30 antibodies and uses thereof
WO2002061087A2 (en) 2000-12-19 2002-08-08 Lifespan Biosciences, Inc. Antigenic peptides, such as for g protein-coupled receptors (gpcrs), antibodies thereto, and systems for identifying such antigenic peptides
WO2002054940A2 (en) 2001-01-12 2002-07-18 University Of Medicine & Dentistry Of New Jersey Bone morphogenetic protein-2 in the treatment and diagnosis of cancer
US20030119119A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030119126A1 (en) 2001-01-16 2003-06-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
WO2002059377A2 (en) 2001-01-24 2002-08-01 Protein Design Labs Methods of diagnosis of breast cancer, compositions and methods of screening for modulators of breast cancer
AU2002251841A1 (en) 2001-01-30 2002-08-12 Corixa Corporation Compositions and methods for the therapy and diagnosis of pancreatic cancer
WO2002064775A1 (en) 2001-02-12 2002-08-22 Bionomics Limited Identification of genes involved in the tumourigenic process
AU2002258518A1 (en) 2001-03-14 2002-09-24 Millennium Pharmaceuticals, Inc. Nucleic acid molecules and proteins for the identification, assessment, prevention, and therapy of ovarian cancer
EP1243276A1 (en) 2001-03-23 2002-09-25 Franciscus Marinus Hendrikus De Groot Elongated and multiple spacers containing activatible prodrugs
US20040236091A1 (en) 2001-03-28 2004-11-25 Chicz Roman M. Translational profiling
US6362331B1 (en) 2001-03-30 2002-03-26 Council Of Scientific And Industrial Research Process for the preparation of antitumor agents
WO2003008537A2 (en) 2001-04-06 2003-01-30 Mannkind Corporation Epitope sequences
US6820011B2 (en) 2001-04-11 2004-11-16 The Regents Of The University Of Colorado Three-dimensional structure of complement receptor type 2 and uses thereof
JP5232350B2 (en) 2001-04-17 2013-07-10 ザ ボード オブ トラスティーズ オブ ザ ユニバーシティ オブ アーカンソー Repeat sequence of CA125 gene and its use for diagnostic and therapeutic intervention
CA2444691A1 (en) 2001-04-18 2002-10-31 Protein Design Labs, Inc. Methods of diagnosis of lung cancer, compositions and methods of screening for modulators of lung cancer
DE60230868D1 (en) 2001-04-26 2009-03-05 Biogen Inc CRIPTOBLOCKING ANTIBODIES AND THEIR USE
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
JP2005504513A (en) 2001-05-09 2005-02-17 コリクサ コーポレイション Compositions and methods for treatment and diagnosis of prostate cancer
WO2002092836A2 (en) 2001-05-11 2002-11-21 Sloan-Kettering Institute For Cancer Research Nucleic acid sequence encoding ovarian antigen, ca125, and uses thereof
EA007275B1 (en) 2001-05-24 2006-08-25 Займодженетикс, Инк. Taci-immunoglobulin fusion proteins
EP2277542B1 (en) 2001-06-01 2014-04-16 Cornell Research Foundation Inc. Modified antibodies to prostrate-specific membrane antigen and uses thereof
WO2002098358A2 (en) 2001-06-04 2002-12-12 Eos Biotechnology, Inc. Methods of diagnosis and treatment of androgen-dependent prostate cancer, prostate cancer undergoing androgen-withdrawal, and androgen-independent prostate cancer
JP2005518185A (en) 2001-06-04 2005-06-23 キュラジェン コーポレイション Novel protein and nucleic acid encoding it
WO2002099060A2 (en) 2001-06-05 2002-12-12 Exelixis, Inc. Dgks as modifiers of the p53 pathway and methods of use
ATE483976T1 (en) 2001-06-05 2010-10-15 Exelixis Inc GFATS AS P53 PATHWAY MODIFIERS AND METHODS OF USE
US7235358B2 (en) 2001-06-08 2007-06-26 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
WO2002101075A2 (en) 2001-06-13 2002-12-19 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of cervical cancer
AU2002347428A1 (en) 2001-06-18 2003-01-02 Eos Biotechnology Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
US7189507B2 (en) 2001-06-18 2007-03-13 Pdl Biopharma, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
US20030148408A1 (en) 2001-09-18 2003-08-07 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7705120B2 (en) 2001-06-21 2010-04-27 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
WO2003002717A2 (en) 2001-06-28 2003-01-09 Schering Corporation Biological activity of ak155
US20030120040A1 (en) 2001-06-29 2003-06-26 Genentech, Inc. Secreted and Transmembrane polypeptides and nucleic acids encoding the same
AU2002314433A1 (en) 2001-07-02 2003-01-21 Licentia Ltd. Ephrin-tie receptor materials and methods
US20040076955A1 (en) 2001-07-03 2004-04-22 Eos Biotechnology, Inc. Methods of diagnosis of bladder cancer, compositions and methods of screening for modulators of bladder cancer
WO2003003984A2 (en) 2001-07-05 2003-01-16 Curagen Corporation Novel proteins and nucleic acids encoding same
US7446185B2 (en) 2001-07-18 2008-11-04 The Regents Of The University Of California Her2/neu target antigen and use of same to stimulate an immune response
WO2003009814A2 (en) 2001-07-25 2003-02-06 Millennium Pharmaceuticals, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of prostate cancer
HUP0500992A3 (en) 2001-08-03 2007-11-28 Genentech Inc Tacis and br3 polypeptides and uses thereof
CA2457819A1 (en) 2001-08-14 2003-02-27 The General Hospital Corporation Nucleic acid and amino acid sequences involved in pain
US20030092013A1 (en) 2001-08-16 2003-05-15 Vitivity, Inc. Diagnosis and treatment of vascular disease
WO2003018621A2 (en) 2001-08-23 2003-03-06 Oxford Biomedica (Uk) Limited Genes
US6902930B2 (en) 2001-08-29 2005-06-07 Vanderbilt University Human Mob-5 (IL-24) receptors and uses thereof
US20030124579A1 (en) 2001-09-05 2003-07-03 Eos Biotechnology, Inc. Methods of diagnosis of ovarian cancer, compositions and methods of screening for modulators of ovarian cancer
JP2005505271A (en) 2001-09-06 2005-02-24 アジェンシス, インコーポレイテッド Nucleic acids and corresponding proteins given the name STEAP-1 useful in the treatment and detection of cancer
WO2003025138A2 (en) 2001-09-17 2003-03-27 Protein Design Labs, Inc. Methods of diagnosis of cancer compositions and methods of screening for modulators of cancer
US20050004017A1 (en) 2001-09-18 2005-01-06 Yuval Reiss Methods and compositions for treating hcap associated diseases
CA2460621A1 (en) 2001-09-19 2003-03-27 Nuvelo, Inc. Novel nucleic acids and polypeptides
US7091186B2 (en) 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
WO2003026577A2 (en) 2001-09-24 2003-04-03 Seattle Genetics, Inc. P-amidobenzylethers in drug delivery agents
US20030077644A1 (en) 2001-09-28 2003-04-24 Bing Yang Diagnosis and treatment of diseases caused by mutations in CD72
US20040249144A1 (en) 2001-10-03 2004-12-09 Zairen Sun Regulated breast cancer genes
WO2003029277A2 (en) 2001-10-03 2003-04-10 Rigel Pharmaceuticals, Inc. Modulators of lymphocyte activation and migration
US20050123925A1 (en) 2002-11-15 2005-06-09 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1578385A4 (en) 2001-10-19 2011-11-09 Genentech Inc Compositions and methods for the diagnosis and treatment of inflammatory bowel disorders
WO2003035846A2 (en) 2001-10-24 2003-05-01 National Jewish Medical And Research Center Structure of tall-1 and its cognate receptor
JP4255382B2 (en) 2001-10-31 2009-04-15 アルコン,インコーポレイテッド Bone morphogenetic protein (BMP), BMP receptor and BMP binding protein and their use in the diagnosis and treatment of glaucoma
WO2003042661A2 (en) 2001-11-13 2003-05-22 Protein Design Labs, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
US20030232350A1 (en) 2001-11-13 2003-12-18 Eos Biotechnology, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
WO2003043583A2 (en) 2001-11-20 2003-05-30 Seattle Genetics, Inc. Treatment of immunological disorders using anti-cd30 antibodies
US7344843B2 (en) 2001-11-29 2008-03-18 Serono Genetics Institute S.A. Agonists and antagonists of prolixin for the treatment of metabolic disorders
SI1461428T1 (en) 2001-12-03 2012-07-31 Alexion Pharma Inc Method for producing hybrid antibodies
WO2003048202A2 (en) 2001-12-03 2003-06-12 Asahi Kasei Pharma Corporation Nf-kappab activating genes
AU2002366951A1 (en) 2001-12-10 2003-07-09 Nuvelo,Inc. Novel nucleic acids and polypeptides
US20030134790A1 (en) 2002-01-11 2003-07-17 University Of Medicine And Dentistry Of New Jersey Bone Morphogenetic Protein-2 And Bone Morphogenetic Protein-4 In The Treatment And Diagnosis Of Cancer
CA2473549C (en) 2002-01-16 2011-02-15 Japan Science And Technology Agency Moving-image holographic reproducing device and color moving-image holographic reproducing device
US7452675B2 (en) 2002-01-25 2008-11-18 The Queen's Medical Center Methods of screening for TRPM4b modulators
US20030157573A1 (en) 2002-02-12 2003-08-21 Orna Mor Use of Axl receptor for diagnosis and treatment of renal disease
AU2003215365A1 (en) 2002-02-21 2003-09-09 Duke University Treatment methods using anti-cd22 antibodies
EP2388265A1 (en) 2002-02-22 2011-11-23 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO2003083047A2 (en) 2002-03-01 2003-10-09 Exelixis, Inc. MP53s AS MODIFIERS OF THE p53 PATHWAY AND METHODS OF USE
US20050220798A1 (en) 2002-06-04 2005-10-06 Reinhard Ebner Cancer-linked gene as target for chemotherapy
WO2003104399A2 (en) 2002-06-07 2003-12-18 Avalon Pharmaceuticals, Inc Cancer-linked gene as target for chemotherapy
US6660856B2 (en) 2002-03-08 2003-12-09 Kaohsiung Medical University Synthesis of pyrrolo[2,1-c][1,4]benzodiazepine analogues
EP2258712A3 (en) 2002-03-15 2011-05-04 Multicell Immunotherapeutics, Inc. Compositions and Methods to Initiate or Enhance Antibody and Major-histocompatibility Class I or Class II-restricted T Cell Responses by Using Immunomodulatory, Non-coding RNA Motifs
CA2486490A1 (en) 2002-03-19 2003-12-31 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA2478981A1 (en) 2002-03-21 2003-10-02 Sunesis Pharmaceuticals, Inc. Identification of kinase inhibitors
US7193069B2 (en) 2002-03-22 2007-03-20 Research Association For Biotechnology Full-length cDNA
JP2005520566A (en) 2002-03-22 2005-07-14 バイオジェン・アイデック・エムエイ・インコーポレイテッド Cripto specific antibodies
US7317087B2 (en) 2002-03-25 2008-01-08 The Uab Research Foundation Members of the FC receptor homolog gene family (FCRH1-3, 6), related reagents, and uses thereof
WO2003083074A2 (en) 2002-03-28 2003-10-09 Idec Pharmaceuticals Corporation Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
US20030194704A1 (en) 2002-04-03 2003-10-16 Penn Sharron Gaynor Human genome-derived single exon nucleic acid probes useful for gene expression analysis two
BR0308953A (en) 2002-04-05 2006-03-14 Agensys Inc compositions, protein, polynucleotide, method of generating an immune response, detection method, pharmaceutical composition, antibody or fragment thereof, transgenic animal, hybridoma, method of providing a cytotoxic agent or diagnostic agent and method of inhibiting cell growth cancerous
US20040101874A1 (en) 2002-04-12 2004-05-27 Mitokor Inc. Targets for therapeutic intervention identified in the mitochondrial proteome
MXPA04010092A (en) 2002-04-16 2004-12-13 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
AU2003239158A1 (en) 2002-04-17 2003-11-03 Baylor College Of Medicine Aib1 as a prognostic marker and predictor of resistance to encocrine therapy
WO2003093444A2 (en) 2002-05-03 2003-11-13 Incyte Corporation Transporters and ion channels
CA2485983A1 (en) 2002-05-15 2003-11-27 Avalon Pharmaceuticals Cancer-linked gene as target for chemotherapy
AU2003232453A1 (en) 2002-05-30 2003-12-19 David K. Bol Human solute carrier family 7 member 11 (hslc7a11)
AU2003240495A1 (en) 2002-06-04 2003-12-19 Incyte Corporation Diagnostics markers for lung cancer
DK1513934T3 (en) 2002-06-06 2011-05-02 Oncotherapy Science Inc Genes and polypeptides related to human colon cancer diseases
WO2003104270A2 (en) 2002-06-06 2003-12-18 Ingenium Pharmaceuticals Ag Dudulin 2 genes, expression products, non-human animal model: uses in human hematological disease
WO2003105758A2 (en) 2002-06-12 2003-12-24 Avalon Pharmaceuticals, Inc. Cancer-linked gene as target for chemotherapy
US20040249130A1 (en) 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
EP1552002A4 (en) 2002-06-18 2006-02-08 Archemix Corp Aptamer-toxin molecules and methods for using same
WO2004000221A2 (en) 2002-06-20 2003-12-31 The Regents Of The University Of California Compositions and methods for modulating lymphocyte activity
EP1534331B1 (en) 2002-06-21 2014-10-29 Johns Hopkins University School of Medicine Membrane associated tumor endothelium markers
DE10229834A1 (en) 2002-07-03 2004-01-29 Zinser Textilmaschinen Gmbh Drafting system for spinning machines with a downstream compacting device
EP1382969A1 (en) 2002-07-17 2004-01-21 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Diagnosis and prevention of cancer cell invasion
WO2004009622A2 (en) 2002-07-19 2004-01-29 Cellzome Ag Protein complexes of cellular networks underlying the development of cancer and other diseases
CN1692127A (en) 2002-07-25 2005-11-02 健泰科生物技术公司 TACI antibodies and uses thereof
ES2556641T3 (en) 2002-07-31 2016-01-19 Seattle Genetics, Inc. Drug conjugates and their use to treat cancer, an autoimmune disease or an infectious disease
CA2494104A1 (en) 2002-07-31 2004-04-22 Seattle Genetics, Inc. Anti-cd20 antibody-drug conjugates for the treatment of cancer and immune disorders
JP2004121218A (en) 2002-08-06 2004-04-22 Jenokkusu Soyaku Kenkyusho:Kk Method for testing bronchial asthma or chronic obstructive pulmonary disease
AU2003251471A1 (en) 2002-08-06 2004-02-25 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with human cxc chemokine receptor 5(cxcr5)
MXPA05001933A (en) 2002-08-19 2005-04-28 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
AU2003278725A1 (en) 2002-08-27 2004-03-19 Bristol-Myers Squibb Company Polynucleotide predictor set for identifying protein tyrosine kinase modulators
WO2004020595A2 (en) 2002-08-29 2004-03-11 Five Prime Therapeutics, Inc. Novel human polypeptides encoded by polynucleotides
WO2004019993A1 (en) 2002-08-30 2004-03-11 Ramot At Tel Aviv University Ltd. Self-immolative dendrimers releasing many active moieties upon a single activating event
AU2002951346A0 (en) 2002-09-05 2002-09-26 Garvan Institute Of Medical Research Diagnosis of ovarian cancer
MXPA05002455A (en) 2002-09-06 2005-06-03 Mannkind Corp Epitope sequences.
EP1581648A2 (en) 2002-09-09 2005-10-05 Nura, Inc. G protein coupled receptors and uses thereof
JP2004113151A (en) 2002-09-27 2004-04-15 Sankyo Co Ltd Oncogene and its application
EP1554309A2 (en) 2002-10-03 2005-07-20 McGILL UNIVERSITY Antibodies and cyclic peptides which bind cea (carcinoembryonic antigen) and their use as cancer therapeutics
US20060183120A1 (en) 2002-10-04 2006-08-17 Teh Bin T Molecular sub-classification of kidney tumors and the discovery of new diagnostic markers
US20040138269A1 (en) 2002-10-11 2004-07-15 Sugen, Inc. Substituted pyrroles as kinase inhibitors
EP1581169A4 (en) 2002-11-08 2008-09-17 Genentech Inc Compositions and methods for the treatment of natural killer cell related diseases
EP1578940A4 (en) 2002-11-13 2007-12-12 Genentech Inc Methods and compositions for diagnosing dysplasia
GB0226593D0 (en) 2002-11-14 2002-12-24 Consultants Ltd Compounds
EP1560599A1 (en) 2002-11-14 2005-08-10 Syntarga B.V. Prodrugs built as multiple self-elimination-release spacers
JP2006516189A (en) 2002-11-15 2006-06-29 ザ ボード オブ トラスティーズ オブ ザ ユニバーシティ オブ アーカンソー CA125 gene and its use for diagnosis and therapy
JP4915980B2 (en) 2002-11-15 2012-04-11 エムユーエスシー ファウンデーション フォー リサーチ デベロップメント Complement receptor 2 targeted complement regulator
WO2004046342A2 (en) 2002-11-20 2004-06-03 Biogen Idec Inc. Novel gene targets and ligands that bind thereto for treatment and diagnosis of carcinomas
US7557092B2 (en) 2002-11-21 2009-07-07 University Of Utah Research Foundation Purinergic modulation of smell
AU2003298786A1 (en) 2002-11-26 2004-06-18 Protein Design Labs, Inc. Methods of detecting soft tissue sarcoma, compositions and methods of screening for soft tissue sarcoma modulators
US20070154886A1 (en) 2002-12-06 2007-07-05 Macina Roberto A Composition, splice variants and methods relating to ovarian specific genes and proteins
JP2004198419A (en) 2002-12-13 2004-07-15 Bayer Healthcare Llc Detection method using timp1
WO2004058171A2 (en) 2002-12-20 2004-07-15 Protein Design Labs, Inc. Antibodies against gpr64 and uses thereof
AU2003299819A1 (en) 2002-12-23 2004-07-22 Human Genome Sciences, Inc. Neutrokine-alpha conjugate, neutrokine-alpha complex, and uses thereof
WO2004063709A2 (en) 2003-01-08 2004-07-29 Bristol-Myers Squibb Company Biomarkers and methods for determining sensitivity to epidermal growth factor receptor modulators
US20050227301A1 (en) 2003-01-10 2005-10-13 Polgen Cell cycle progression proteins
WO2004063355A2 (en) 2003-01-10 2004-07-29 Protein Design Labs, Inc. Novel methods of diagnosis of metastatic cancer, compositions and methods of screening for modulators of matastatic cancer
WO2004065577A2 (en) 2003-01-14 2004-08-05 Bristol-Myers Squibb Company Polynucleotides and polypeptides associated with the nf-kb pathway
EP1583821A4 (en) 2003-01-15 2007-07-18 Millennium Pharm Inc Methods and compositions for treating urological disorders using 44390, 54181, 211, 5687, 884, 1405, 636, 4421, 5410, 30905, 2045, 16405, 18560, 2047, 33751, 52872, 14063, 20739, 32544, 43239, 44373, 51164, 53010, 16852, 1587, 2207, 22245, 2387, 52908, 69112, 14990, 18547, 115, 579, 15985, 15625, 76
CA2516128A1 (en) 2003-02-14 2004-09-02 Sagres Discovery, Inc. Therapeutic targets in cancer
US20030224411A1 (en) 2003-03-13 2003-12-04 Stanton Lawrence W. Genes that are up- or down-regulated during differentiation of human embryonic stem cells
US6884799B2 (en) 2003-03-31 2005-04-26 Council Of Scientific And Industrial Research Non-cross-linking pyrrolo[2,1-c][1,4]benzodiazepines and process thereof
GB0321295D0 (en) 2003-09-11 2003-10-15 Spirogen Ltd Synthesis of protected pyrrolobenzodiazepines
GB0416511D0 (en) 2003-10-22 2004-08-25 Spirogen Ltd Pyrrolobenzodiazepines
AU2004284075A1 (en) 2003-10-22 2005-05-06 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Pyrrolobenzodiazepine derivatives, compositions comprising the same and methods related thereto
SG195524A1 (en) 2003-11-06 2013-12-30 Seattle Genetics Inc Monomethylvaline compounds capable of conjugation to ligands
EP1720908A2 (en) 2004-02-17 2006-11-15 Absalus, Inc. Super-humanized antibodies against respiratory syncytial virus
WO2005082023A2 (en) 2004-02-23 2005-09-09 Genentech, Inc. Heterocyclic self-immolative linkers and conjugates
GB0404578D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Pyrrolobenzodiazepines
GB0404574D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Amino acids
GB0404577D0 (en) 2004-03-01 2004-04-07 Spirogen Ltd Pyrrolobenzodiazepines
CA2558195C (en) 2004-03-01 2012-11-06 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4]benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
DE102004010943A1 (en) 2004-03-03 2005-09-29 Degussa Ag Process for the preparation of N-protected 4-ketproline derivatives
EP1723152B1 (en) 2004-03-09 2015-02-11 Spirogen Sàrl Pyrrolobenzodiazepines
FR2869231B1 (en) 2004-04-27 2008-03-14 Sod Conseils Rech Applic THERAPEUTIC COMPOSITION CONTAINING AT LEAST ONE PYRROLOBENZODIAZEPINE DERIVATIVE AND FLUDARABINE
GB0410725D0 (en) 2004-05-13 2004-06-16 Spirogen Ltd Pyrrolobenzodiazepine therapeutic agents
AU2005286607B2 (en) 2004-09-23 2011-01-27 Genentech, Inc. Cysteine engineered antibodies and conjugates
JP2008521828A (en) 2004-11-29 2008-06-26 シアトル ジェネティックス, インコーポレイテッド Engineered antibodies and immunoconjugates
US7849909B2 (en) 2004-12-24 2010-12-14 Showa Denko K.K. Production method of thermoelectric semiconductor alloy, thermoelectric conversion module and thermoelectric power generating device
AU2006210724A1 (en) 2005-02-03 2006-08-10 Antitope Limited Human antibodies and proteins
ES2657443T3 (en) 2005-03-25 2018-03-05 Gitr, Inc. Anti-GITR antibodies and uses thereof
CA2605507C (en) 2005-04-19 2016-06-28 Seattle Genetics, Inc. Humanized anti-cd70 binding agents and uses thereof
JP5061096B2 (en) 2005-04-21 2012-10-31 スピロゲン リミティッド Pyrrolobenzodiazepine
CN101248089A (en) 2005-07-01 2008-08-20 米德列斯公司 Human monoclonal antibodies to programmed death ligand 1(PD-L1)
DE602006006199D1 (en) 2005-10-05 2009-05-20 Spirogen Ltd 4-Ä4- (5-OXO-2,3,5,11A-TETRAHYDRO-5H-PYRROLOA2, 1-CÜÄ1,4ÜBENZODIAZEPIN-8-YLOXY) -BUTYRYLAMINOÜ-1H-PYRROLO-2-CARBOXYL ACKYLEEDER DERIVATIVES AND RELATED COMPOUND FOR TREATING ONE PROLIFERATIVE DISEASE
BRPI0617165B1 (en) 2005-10-07 2023-10-03 Exelixis Inc MEK INHIBITOR COMPOUNDS, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM AND METHODS OF USE THEREOF
ES2374964T3 (en) 2006-01-25 2012-02-23 Sanofi CYTOTOXIC AGENTS UNDERSTANDING NEW TOMAIMYCIN DERIVATIVES.
JP5622390B2 (en) 2006-07-18 2014-11-12 サノフイ Anti-EPHA2 antagonist antibody for cancer treatment
US20080112961A1 (en) 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
EP1914242A1 (en) 2006-10-19 2008-04-23 Sanofi-Aventis Novel anti-CD38 antibodies for the treatment of cancer
US8455622B2 (en) 2006-12-01 2013-06-04 Seattle Genetics, Inc. Variant target binding agents and uses thereof
CL2008001334A1 (en) 2007-05-08 2008-09-22 Genentech Inc ANTI-MUC16 ANTIBODY DESIGNED WITH CISTEINE; CONJUGADO THAT UNDERSTANDS IT; METHOD OF PRODUCTION; PHARMACEUTICAL FORMULATION THAT UNDERSTANDS IT; AND ITS USE TO TREAT CANCER.
EP3124046B1 (en) 2007-07-12 2019-12-25 GITR, Inc. Combination therapies employing gitr binding molecules
ES2435779T3 (en) 2007-07-19 2013-12-23 Sanofi Cytotoxic agents comprising new tomaimycin derivatives and their therapeutic use
ES2450755T3 (en) 2007-10-19 2014-03-25 Genentech, Inc. Anti-TENB2 antibodies engineered with cysteine, and antibody and drug conjugates
CA2705164A1 (en) 2007-11-12 2009-05-22 U3 Pharma Gmbh Axl antibodies
RU2559530C2 (en) 2007-11-15 2015-08-10 Чугаи Сейяку Кабусики Кайся Monoclonal antibodies capable of binding with axl protein and their application
US8609105B2 (en) 2008-03-18 2013-12-17 Seattle Genetics, Inc. Auristatin drug linker conjugates
GB0813432D0 (en) 2008-07-22 2008-08-27 Spirogen Ltd Pyrrolobenzodiazepines
PE20110435A1 (en) 2008-08-25 2011-07-20 Amplimmune Inc ANTAGONIST COMPOSITIONS OF PD-1
GB0819097D0 (en) 2008-10-17 2008-11-26 Spirogen Ltd Pyrrolobenzodiazepines
GB0819095D0 (en) 2008-10-17 2008-11-26 Spirogen Ltd Pyrrolobenzodiazepines
SG10201706294VA (en) 2009-02-05 2017-09-28 Immunogen Inc Novel benzodiazepine derivatives
TWI526223B (en) 2009-05-11 2016-03-21 U3製藥有限責任公司 Humanized axl antibodies
CN102459344A (en) 2009-05-15 2012-05-16 中外制药株式会社 Anti-axl antibody
WO2011005481A1 (en) 2009-06-22 2011-01-13 Medimmune, Llc ENGINEERED Fc REGIONS FOR SITE-SPECIFIC CONJUGATION
TW201106972A (en) 2009-07-27 2011-03-01 Genentech Inc Combination treatments
JP5382792B2 (en) 2009-08-14 2014-01-08 独立行政法人理化学研究所 Method for simultaneously measuring primary and secondary photosensitivity anisotropy in optical second-order nonlinear thin film, apparatus for executing the method, and program for causing computer to execute the method
FR2949469A1 (en) 2009-08-25 2011-03-04 Sanofi Aventis ANTICANCER DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
EP2473524A4 (en) 2009-09-01 2013-05-22 Abbott Lab Dual variable domain immunoglobulins and uses thereof
EP2473531A4 (en) 2009-09-03 2013-05-01 Merck Sharp & Dohme Anti-gitr antibodies
US20110070227A1 (en) 2009-09-18 2011-03-24 Anna-Marie Novotney-Barry Treatment of Autoimmune and Inflammatory Diseases
CA2775350A1 (en) 2009-09-24 2011-03-31 Seattle Genetics, Inc. Dr5 ligand drug conjugates
US9040526B2 (en) 2010-02-09 2015-05-26 Bristol-Myers Squibb Company Benzylpyrrolidinone derivatives as modulators of chemokine receptor activity
GB201006340D0 (en) 2010-04-15 2010-06-02 Spirogen Ltd Synthesis method and intermediates
CN103068405A (en) 2010-04-15 2013-04-24 西雅图基因公司 Targeted pyrrolobenzodiazapine conjugates
CN102971329B (en) 2010-04-15 2016-06-29 麦迪穆有限责任公司 Pyrrolobenzodiazepines for treating proliferative disease is tall and erect
DK2528625T3 (en) 2010-04-15 2013-10-14 Spirogen Sarl Pyrrolobenzodiazepines and their conjugates
WO2011130615A2 (en) 2010-04-15 2011-10-20 Dr. Reddy's Laboratories Ltd. Preparation of lacosamide
WO2011159980A1 (en) * 2010-06-18 2011-12-22 Genentech, Inc. Anti-axl antibodies and methods of use
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2012064733A2 (en) 2010-11-09 2012-05-18 Medimmune, Llc Antibody scaffold for homogenous conjugation
KR20140010067A (en) 2011-02-15 2014-01-23 이뮤노젠 아이엔씨 Methods of preparation of conjugates
DK2699264T3 (en) 2011-04-20 2018-06-25 Medimmune Llc ANTIBODIES AND OTHER MOLECULES BINDING B7-H1 AND PD-1
US9409988B2 (en) 2011-06-22 2016-08-09 Inserm (Institut National De La Sante Et De La Recherche Medicale) Anti-Axl antibodies and uses thereof
US9249228B2 (en) 2011-06-22 2016-02-02 Oribase Pharma Anti-Axl antibodies and uses thereof
SG11201400770SA (en) 2011-09-20 2014-04-28 Spirogen Sarl Pyrrolobenzodiazepines as unsymmetrical dimeric pbd compounds for inclusion in targeted conjugates
EA036202B1 (en) 2011-10-14 2020-10-14 Сиэтл Дженетикс, Инк. Pyrrolobenzodiazepines and targeted conjugates
ES2945932T3 (en) 2011-10-14 2023-07-10 Seagen Inc Pyrrolobenzodiazepines and targeted conjugates
CN103987407B (en) 2011-10-14 2016-08-24 麦迪穆有限责任公司 Pyrrolobenzodiazepines Zhuo and conjugate thereof
BR112014009070B1 (en) 2011-10-14 2021-11-23 Medimmune Limited SYNTHESIS METHOD AND INTERMEDIATES USEFUL IN THE PREPARATION OF PYROLOBENZO-DIAZEPINES
US9399073B2 (en) 2011-10-14 2016-07-26 Seattle Genetics, Inc. Pyrrolobenzodiazepines
EP2751110B1 (en) 2011-10-14 2017-04-19 MedImmune Limited Asymmetrical bis-(5H-Pyrrolo[2,1-c][1,4]benzodiazepin-5-one) derivatives for the treatment of proliferative and autoimmune diseases
EP2589609A1 (en) 2011-11-03 2013-05-08 Pierre Fabre Medicament Antigen binding protein and its use as addressing product for the treatment of cancer
US11147852B2 (en) 2011-12-23 2021-10-19 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
BR112014027143B1 (en) 2012-04-30 2020-06-09 Medimmune Ltd pyrrolobenzodiazepines
MX2014013144A (en) 2012-04-30 2015-05-11 Ucl Business Plc Pyrrolobenzodiazepines.
WO2013177481A1 (en) 2012-05-25 2013-11-28 Immunogen, Inc. Benzodiazepines and conjugates thereof
JP2015527318A (en) 2012-07-09 2015-09-17 ジェネンテック, インコーポレイテッド Immune complex comprising anti-CD22
JP2015528818A (en) 2012-08-02 2015-10-01 ジェネンテック, インコーポレイテッド Anti-ETBR antibodies and immunoconjugates
SI2904011T1 (en) 2012-10-02 2017-10-30 Bristol-Myers Squibb Company Combination of anti-kir antibodies and anti-pd-1 antibodies to treat cancer
WO2014057118A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sarl Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
CA2885340C (en) 2012-10-12 2016-11-08 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
AU2013328673B2 (en) 2012-10-12 2017-07-13 Medimmune Limited Synthesis and intermediates of pyrrolobenzodiazepine derivatives for conjugation
CN104955485B (en) 2012-10-12 2018-01-30 Adc疗法责任有限公司 Pyrrolobenzodiazepines Zhuo Anti-HER 2 conjugate
JP6392763B2 (en) 2012-10-12 2018-09-19 エイディーシー・セラピューティクス・エス・アーAdc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugate
HUE039329T2 (en) 2012-10-12 2018-12-28 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
RS57694B1 (en) 2012-10-12 2018-11-30 Adc Therapeutics Sa Pyrrolobenzodiazepine - anti-psma antibody conjugates
ES2660029T3 (en) 2012-10-12 2018-03-20 Medimmune Limited Antibody-pyrrolobenzodiazepine conjugates
MX364327B (en) 2012-10-12 2019-04-23 Medimmune Ltd Pyrrolobenzodiazepine-anti-cd22 antibody conjugates.
ES2680153T3 (en) 2012-10-12 2018-09-04 Adc Therapeutics Sa Anti-PSMA-pyrrolobenzodiazepine antibody conjugates
BR112015008251B1 (en) 2012-10-12 2023-09-26 Medimmune Limited PYRROLOBENZODIAZEPINES, CONJUGATES THEREOF, COMPOSITION AND PHARMACEUTICAL COMPOSITION COMPRISING THE CONJUGATES, USE OF THE CONJUGATES FOR THE TREATMENT OF A PROLIFERATIVE DISEASE AND METHOD FOR PREPARING THE CONJUGATES
NZ710745A (en) 2013-03-13 2019-03-29 Genentech Inc Pyrrolobenzodiazepines and conjugates thereof
JP2016518382A (en) 2013-04-26 2016-06-23 ピエール、ファーブル、メディカマン Axl antibody drug conjugate and use thereof for the treatment of cancer
MX2016002547A (en) 2013-08-28 2016-06-17 Stemcentrx Inc Site-specific antibody conjugation methods and compositions.
EP3038659A4 (en) 2013-08-28 2017-07-26 AbbVie Stemcentrx LLC Engineered anti-dll3 conjugates and methods of use
PL3702373T3 (en) 2013-09-13 2022-12-05 Beigene Switzerland Gmbh Anti-pd1 antibodies and their use as therapeutics and diagnostics
HUE062317T2 (en) 2013-09-20 2023-10-28 Bristol Myers Squibb Co Combination of anti-lag-3 antibodies and anti-pd-1 antibodies to treat tumors
CN106102774A (en) 2013-12-17 2016-11-09 豪夫迈·罗氏有限公司 Comprise OX40 and combine agonist and PD 1 axle combines the combination treatment of antagonist
MX2016012779A (en) 2014-03-31 2017-04-27 Genentech Inc Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists.
GB201410825D0 (en) 2014-06-18 2014-07-30 Bergenbio As Anti-axl antibodies
KR102130600B1 (en) 2014-07-03 2020-07-08 베이진 엘티디 Anti-PD-L1 Antibodies and Their Use as Therapeutics and Diagnostics
RU2715038C2 (en) 2014-07-11 2020-02-21 Дженентек, Инк. Anti-pd-l1 antibodies and methods for their diagnostic use
SG11201700074YA (en) 2014-07-15 2017-02-27 Genentech Inc Compositions for treating cancer using pd-1 axis binding antagonists and mek inhibitors
CR20170099A (en) 2014-09-17 2017-07-19 Genentech Inc PIRROLOBENZODIAZEPINAS AND CONJUGADOS DE ANTIBERPOS-DISULFURO DE LAS SISAS
WO2016053107A1 (en) 2014-10-03 2016-04-07 Synaffix B.V. Sulfamide linker, conjugates thereof, and methods of preparation
TW201619200A (en) 2014-10-10 2016-06-01 麥迪紐有限責任公司 Humanized anti-OX40 antibodies and uses thereof
MX2017005751A (en) 2014-11-03 2018-04-10 Genentech Inc Method and biomarkers for predicting efficacy and evaluation of an ox40 agonist treatment.
MX2017006320A (en) 2014-11-17 2017-08-10 Genentech Inc Combination therapy comprising ox40 binding agonists and pd-1 axis binding antagonists.
EP3233119A2 (en) 2014-12-18 2017-10-25 Bergen Teknologioverforing AS Anti-axl antagonistic antibodies
WO2016127052A1 (en) 2015-02-05 2016-08-11 Bristol-Myers Squibb Company Cxcl11 and smica as predictive biomarkers for efficacy of anti-ctla4 immunotherapy
GB201506407D0 (en) 2015-04-15 2015-05-27 Berkel Patricius H C Van And Howard Philip W Site-specific antibody-drug conjugates
GB201506409D0 (en) 2015-04-15 2015-05-27 Williams David G And Berkel Patricius H C Van And Howard Philip W Humanized anti-axl antibodies and their conjugates
CN113577264A (en) 2015-04-17 2021-11-02 百时美施贵宝公司 Compositions comprising a combination of an anti-PD-1 antibody and an additional antibody
AU2016256911B2 (en) 2015-05-07 2022-03-31 Agenus Inc. Anti-OX40 antibodies and methods of use thereof
US20160347848A1 (en) 2015-05-28 2016-12-01 Medimmune Limited Therapeutic combinations and methods for treating neoplasia
CA2988115A1 (en) 2015-06-03 2016-12-08 Bristol-Myers Squibb Company Anti-gitr antibodies for cancer diagnostics
JP2018526977A (en) 2015-06-29 2018-09-20 ザ ロックフェラー ユニヴァーシティ Antibody against CD40 with enhanced agonist activity
BR112018014016A2 (en) 2016-01-25 2019-02-05 Pfizer combination of an ox40 agonist and a 4-1bb monoclonal antibody agonist for cancer treatment
PT3544636T (en) 2017-02-08 2021-05-04 Medimmune Ltd Pyrrolobenzodiazepine-antibody conjugates

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9975954B2 (en) * 2014-06-18 2018-05-22 Bergenbio Asa Anti-Axl antibodies

Also Published As

Publication number Publication date
WO2016166296A3 (en) 2016-11-24
US11059893B2 (en) 2021-07-13
EP3283525A2 (en) 2018-02-21
WO2016166296A2 (en) 2016-10-20
US20190352407A1 (en) 2019-11-21
GB201506411D0 (en) 2015-05-27

Similar Documents

Publication Publication Date Title
US20220073625A1 (en) Humanized anti-axl antibodies
US20210009705A1 (en) Anti-ox40 antibodies and methods of use
EP3126394B1 (en) Anti-ox40 antibodies and methods of use
US11318211B2 (en) Dosage regimes for the administration of an anti-CD19 ADC
US20220347309A1 (en) Pyrrolobenzodiazepine resistance
US11596696B2 (en) Combination therapy with an anti-CD25 antibody-drug conjugate
US20180142025A1 (en) Diagnostic test involving anti-cd25-adc
BR112019026498A2 (en) dosing regimens for administration of an anti-cd25 adc
US20210322564A1 (en) Combination therapy
US20230132256A1 (en) Combination therapy
US11976122B2 (en) Anti-IL13Rα2 antibodies
US10214584B2 (en) Therapeutic pharmaceutical composition employing anti-SLC6A6 antibody
US20190127461A1 (en) Cancer treatment pharmaceutical composition using anti-mct5 antibody
AU2022302769A1 (en) Combination therapy using antibody-drug conjugates
CA3220935A1 (en) Combination therapy using antibody-drug conjugates
WO2022074033A1 (en) Combination therapy

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: BERGENBIO ASA, NORWAY

Free format text: CHANGE OF NAME;ASSIGNOR:BERGENBIO AS;REEL/FRAME:064069/0604

Effective date: 20170322

Owner name: BERGENBIO ASA, NORWAY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:VAN BERKEL, PATRICIUS HENDRIKUS CORNELIS;REEL/FRAME:064032/0573

Effective date: 20160831

Owner name: BERGENBIO AS, NORWAY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WILLIAMS, DAVID GARETH;REEL/FRAME:064032/0558

Effective date: 20160810

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED