Humanized Anti-axl Antibodies

VAN BERKEL; PATRICIUS HENDRIKUS CORNELIS ;   et al.

Patent Application Summary

U.S. patent application number 17/372961 was filed with the patent office on 2022-03-10 for humanized anti-axl antibodies. The applicant listed for this patent is BERGENBIO ASA. Invention is credited to PATRICIUS HENDRIKUS CORNELIS VAN BERKEL, DAVID G. WILLIAMS.

Application Number20220073625 17/372961
Document ID /
Family ID
Filed Date2022-03-10

United States Patent Application 20220073625
Kind Code A1
VAN BERKEL; PATRICIUS HENDRIKUS CORNELIS ;   et al. March 10, 2022

HUMANIZED ANTI-AXL ANTIBODIES

Abstract

The present disclosure relates to humanized anti-Axl antibodies.


Inventors: VAN BERKEL; PATRICIUS HENDRIKUS CORNELIS; (Lausanne, CH) ; WILLIAMS; DAVID G.; (Epson, GB)
Applicant:
Name City State Country Type

BERGENBIO ASA

Bergen

NO
Appl. No.: 17/372961
Filed: July 12, 2021

Related U.S. Patent Documents

Application Number Filing Date Patent Number
15566635 Oct 13, 2017 11059893
PCT/EP2016/058368 Apr 15, 2016
17372961

International Class: C07K 16/28 20060101 C07K016/28

Foreign Application Data

Date Code Application Number
Apr 15, 2015 GB 1506411.6

Claims



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).sub.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.
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] 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. .sctn. 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

[0002] 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.

[0003] The present disclosure relates to humanized anti-Axl antibodies.

BACKGROUND

[0004] Axl

[0005] 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-.kappa.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.

[0006] 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).

[0007] 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).

[0008] 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).

[0009] 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).

[0010] Anti-Axl Antibodies

[0011] 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].

[0012] 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

[0013] The present disclosure provides humanized anti-AXL antibodies derived from the `mouse 1H12` antibody.

[0014] 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.

[0015] 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.

[0016] 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.

[0017] 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.

[0018] 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.

[0019] In some embodiments AXL is human AXL.

[0020] 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.

[0021] 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.

[0022] In some embodiments the humanized antibodies of the disclosure include antibodies comprising V.sub.H and V.sub.L domains with amino acid sequences that are identical to the sequences described herein.

DETAILED DISCLOSURE

[0023] Antibody Properties

[0024] Antigen Binding

[0025] 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.

[0026] 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.

[0027] Antibody Affinity

[0028] In some embodiments the humanized antibody binds human AXL with a dissociation constant (K.sub.D) of at least 10.sup.-6 M, such as at least 5.times.10.sup.-7 M, at least 10.sup.-7 M, at least 5.times.10.sup.-8 M, at least 10.sup.-9 M, such as at least 5.times.10.sup.-10 M, at least 10.sup.-10 M, at least 5.times.10.sup.-11 M, at least 10.sup.-11 M, at least 5.times.10.sup.-12 M, at least 10.sup.-12 M, at least 5.times.10.sup.-13 M, at least 10.sup.-13 M, at least 5.times.10.sup.-14 M, at least 10.sup.-14 M, at least 5.times.10.sup.-15 M, or at least 10.sup.-15 M.

[0029] 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)].

[0030] 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.

[0031] 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.

[0032] Antibody Isoelectric Point (pI)

[0033] 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.

[0034] 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.

[0035] 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.

[0036] Antibody Immunogenicity

[0037] 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.

[0038] 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.

[0039] The disclosure also provided the means produce the antibodies of the disclosure.

[0040] 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.

[0041] In another aspect, the disclosure provides a pharmaceutical composition comprising an antibody pf the disclosure, optionally further comprising a pharmaceutically acceptable carrier or excipient.

[0042] In another aspect the disclosure provides a vector, such as an expression vector, comprising a nucleic acid of the disclosure.

[0043] 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).

[0044] In another aspect the disclosure provides a method of making the antibodies by culturing the host cells of the disclosure.

[0045] 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.

[0046] 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.

[0047] 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.

[0048] 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.

[0049] 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

[0050] Antibody

[0051] 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.

[0052] 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.

[0053] 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.

[0054] 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.

[0055] 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.

[0056] Humanized

[0057] 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.

[0058] 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.

[0059] 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 .ANG. 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.

[0060] 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.

[0061] Sequence Modifications

[0062] 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: [0063] Amino acids having polar side chains (Asp, Glu, Lys, Arg, His, Asn, Gin, Ser, Thr, Tyr, and Cys) [0064] Amino acids having non-polar side chains (Gly, Ala, Val, Leu, lie, Phe, Trp, Pro, and Met) [0065] Amino acids having aliphatic side chains (Gly, Ala Val, Leu, Ile) [0066] Amino acids having cyclic side chains (Phe, Tyr, Trp, His, Pro) [0067] Amino acids having aromatic side chains (Phe, Tyr, Trp) [0068] Amino acids having acidic side chains (Asp, Glu) [0069] Amino acids having basic side chains (Lys, Arg, His) [0070] Amino acids having amide side chains (Asn, Gin) [0071] Amino acids having hydroxy side chains (Ser, Thr) [0072] Amino acids having sulphur-containing side chains (Cys, Met), [0073] Neutral, weakly hydrophobic amino acids (Pro, Ala, Gly, Ser, Thr) [0074] Hydrophilic, acidic amino acids (Gin, Asn, Glu, Asp), and [0075] Hydrophobic amino acids (Leu, Ile, Val)

[0076] Particular preferred conservative amino acids substitution groups are: Val-Leu-Ile, Phe-Tyr, Lys-Arg, Ala-Val, and Asn-Gin.

[0077] 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.

[0078] 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.

[0079] Antibody Production

[0080] 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.

[0081] 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.

[0082] Biological Activity

[0083] In Vitro Cell Proliferation Assays

[0084] 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.

[0085] The in vitro potency of antibodies can be measured by a cell proliferation assay. The CellTiter-Glo.RTM. 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.RTM. 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.RTM. 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.

[0086] 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.RTM. 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.

[0087] 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.

[0088] The cell suspension is diluted to the desired seeding density and dispensed (100 .mu.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.

[0089] A stock solution (1 ml) of antibody (20 .mu.g/ml) is made in the appropriate cell culture medium.

[0090] Serial 10-fold dilutions of stock antibody are made in 15 ml centrifuge tubes by serially transferring 100 .mu.l to 900 .mu.l of cell culture medium.

[0091] Four replicate wells of each antibody dilution (100 .mu.l) are dispensed in 96-well black plates, previously plated with cell suspension (100 .mu.l), resulting in a final volume of 200 .mu.l. Control wells receive cell culture medium (100 .mu.l).

[0092] 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.

[0093] At the end of the incubation period, cell viability is assessed with the Alamar blue assay.

[0094] AlamarBlue (Invitrogen) is dispensed over the whole plate (20 .mu.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.

[0095] Use

[0096] The antibody of the disclosure may be used to target a target location.

[0097] The target location is preferably a proliferative cell population. The antibody is an antibody for an antigen present on a proliferative cell population.

[0098] 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.

[0099] The target location may be in vitro, in vivo or ex vivo.

[0100] 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.

[0101] 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.

[0102] 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.

[0103] 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.

[0104] 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.

[0105] 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.

[0106] 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.

[0107] 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.

[0108] Autoimmune diseases for which the antibody may be used in treatment include rheumatologic disorders (such as, for example, rheumatoid arthritis, Sjogren'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, Sjogren's syndrome, Graves' disease, IDDM, pernicious anemia, thyroiditis, and glomerulonephritis.

[0109] Methods of Treatment

[0110] 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.

[0111] 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.

[0112] 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.

[0113] Examples of chemotherapeutic agents include: erlotinib (TARCEVA.RTM., Genentech/OSI Pharm.), docetaxel (TAXOTERE.RTM., Sanofi-Aventis), 5-FU (fluorouracil, 5-fluorouracil, CAS No. 51-21-8), gemcitabine (GEMZAR.RTM., 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.RTM., Bristol-Myers Squibb Oncology, Princeton, N.J.), trastuzumab (HERCEPTIN.RTM., 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.RTM., TEMODAL.RTM., Schering Plough), tamoxifen ((Z)-2-[4-(1,2-diphenylbut-1-enyl)phenoxy]-N,N-dimethylethanamine, NOLVADEX.RTM., ISTUBAL.RTM., VALODEX.RTM.), and doxorubicin (ADRIAMYCIN.RTM.), Akti-1/2, HPPD, and rapamycin.

[0114] More examples of chemotherapeutic agents include: oxaliplatin (ELOXATIN.RTM., Sanofi), bortezomib (VELCADE.RTM., Millennium Pharm.), sutent (SUNITINIB.RTM., SU11248, Pfizer), letrozole (FEMARA.RTM., Novartis), imatinib mesylate (GLEEVEC.RTM., 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.RTM., AstraZeneca), leucovorin (folinic acid), rapamycin (sirolimus, RAPAMUNE.RTM., Wyeth), lapatinib (TYKERB.RTM., GSK572016, Glaxo Smith Kline), lonafamib (SARASAR.TM., SCH 66336, Schering Plough), sorafenib (NEXAVAR.RTM., BAY43-9006, Bayer Labs), gefitinib (IRESSA.RTM., AstraZeneca), irinotecan (CAMPTOSAR.RTM., CPT-11, Pfizer), tipifamib (ZARNESTRA.TM., Johnson & Johnson), ABRAXANE.TM. (Cremophor-free), albumin-engineered nanoparticle formulations of paclitaxel (American Pharmaceutical Partners, Schaumberg, II), vandetanib (rINN, ZD6474, ZACTIMA.RTM., AstraZeneca), chloranmbucil, AG1478, AG1571 (SU 5271; Sugen), temsirolimus (TORISEL.RTM., Wyeth), pazopanib (GlaxoSmithKline), canfosfamide (TELCYTA.RTM., Telik), thiotepa and cyclosphosphamide (CYTOXAN.RTM., NEOSAR.RTM.); 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.RTM. 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.RTM.); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XELODA.RTM., 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.

[0115] 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.RTM.; tamoxifen citrate), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON.RTM. (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.RTM. (megestrol acetate), AROMASIN.RTM. (exemestane; Pfizer), formestanie, fadrozole, RIVISOR.RTM. (vorozole), FEMARA.RTM. (letrozole; Novartis), and ARIMIDEX.RTM. (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.RTM., Genta Inc.); (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME.RTM.) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN.RTM., LEUVECTIN.RTM., and VAXID.RTM.; PROLEUKIN.RTM. rIL-2; topoisomerase 1 inhibitors such as LURTOTECAN.RTM.; ABARELIX.RTM. rmRH; (ix) anti-angiogenic agents such as bevacizumab (AVASTIN.RTM., Genentech); and pharmaceutically acceptable salts, acids and derivatives of any of the above.

[0116] Also included in the definition of "chemotherapeutic agent" are therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN.RTM., Genentech); cetuximab (ERBITUX.RTM., Imclone); panitumumab (VECTIBIX.RTM., Amgen), rituximab (RITUXAN.RTM., Genentech/Biogen Idec), ofatumumab (ARZERRA.RTM., GSK), pertuzumab (PERJETA.TM., OMNITARG.TM., 2C4, Genentech), trastuzumab (HERCEPTIN.RTM., Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG.RTM., Wyeth).

[0117] 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.

[0118] 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.

[0119] 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.

[0120] 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.

[0121] Formulations

[0122] 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.

[0123] 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.

[0124] 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.

[0125] In one embodiment, the composition further comprises other active agents, for example, other therapeutic or prophylactic agents.

[0126] 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.

[0127] Another aspect of the present disclosure pertains to methods of making a pharmaceutical composition comprising admixing at least one [.sup.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.

[0128] 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.

[0129] 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.

[0130] The formulation may be prepared to provide for rapid or slow release; immediate, delayed, timed, or sustained release; or a combination thereof.

[0131] 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 .mu.g/ml, for example from about 10 ng/ml to about 1 .mu.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.

[0132] Dosage

[0133] 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.

[0134] 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.

[0135] In general, a suitable dose of the antibody is in the range of about 100 ng to about 25 mg (more typically about 1 .mu.g 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.

[0136] In one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 100 mg, 3 times daily.

[0137] In one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 150 mg, 2 times daily.

[0138] In one embodiment, the antibody is administered to a human patient according to the following dosage regime: about 200 mg, 2 times daily.

[0139] 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.

[0140] 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.

[0141] 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 .mu.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 .mu.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. 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.

[0142] Treatment

[0143] 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.

[0144] 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.

[0145] 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.

[0146] The Subject/Patient

[0147] 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.

[0148] 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

[0149] FIG. 1: AXL binding ELISA of fully humanised constructs. (2A) to Human AXL. (2B) to Cynomolgus monkey AXL.

[0150] FIG. 2: Accelerated stability analysis

[0151] Materials and Methods

[0152] Protocol 1: Production of DNA Plasmids for Expression

[0153] Materials [0154] For heavy chain construct selection, Zeocin 25 .mu.g/ml (Invivogen) was used. [0155] For light chain construct selection, Blasticidin 100 .mu.g/ml (Life Technologies) was used.

[0156] Method

[0157] Transformed bacteria were spread on LB-agar Zeocin/Blasticidin plates, as required, incubated overnight at 37 C, then colonies were picked from each plate.

[0158] VH or VK colonies/glycerol stocks were inoculated into 3 ml LB containing Zeocin 25 .mu.g/ml or Blasticidin 50 .mu.g/ml, respectively.

[0159] p21, p27, pAdvantage (Promega) and pSVLT (generous gift from Tom Vink) were inoculated in LB-Ampicillin and shaken overnight.

[0160] 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.TM. Plasmid Maxiprep Kit following the manufacturer's instructions.

[0161] Protocol 2. Transient Transfection of HEK293T Cells with Expression Constructs

[0162] Materials [0163] Cells: HEK293T cells [0164] Culture medium: DMEM high glucose 4.5 g/L (PAA) with 10% v/v FCS, penicillin and streptomycin [0165] Fugene HD transfection reagent (Promega #E2311) [0166] Opti-MEM (Life Technologies #11058-021) or [0167] FreeStyle 293 Expression Medium (Life Technologies #12338-018)

[0168] Method

[0169] Grow HEK293T cells in a T75 or T175 flask in a CO.sub.2-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.

[0170] The day before transfection: [0171] 1. Trypsinise the cells, wash 1.times. in DMEM/10% FCS and count the cells. [0172] 2. Seed cells in a 6 well plate in 2 ml per well containing 2.times.10.sup.5 cells.

[0173] Next day, check cells are at least 80% confluent and replace the medium (2 ml/well). [0174] 1. 1.2 .mu.g 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/.mu.l. [0175] 2. Add 0.6 ug of VH and 0.6 ug VK expression plasmid DNAs into of Fugene HD (4.5 .mu.l) and OptiMEM/Freestyle medium, in a total volume of 60 ul, avoiding touching the sides of the tube with the Fugene HD. [0176] 3. Mix and leave at RT for 15 minutes. [0177] 4. Add Fugene mixture drop-wise around the well of HEK293T cells. [0178] 5. Return the 6-well plate to the CO.sub.2-gassed cell culture incubator for 4 days. [0179] 6. Harvest each conditioned medium, centrifuge, and store at 4.degree. C.

[0180] Protocol 3: IgG Quantitation by ELISA

[0181] Materials [0182] Nunc-Immuno Plate MaxiSorp (Life Technologies, 43945A) [0183] Goat Anti-Human IgG(Fc)--AffiniPure: Stratech Scientific, 109-005-098-JIR; 1 mg: 1.3 mg/ml [0184] Human IgG1/kappa antibody (Sigma, 1-3889-1 mg: 1 mg/ml) [0185] Goat anti-human kappa light chain peroxidase conjugate (Sigma, A-7164-1 ml) [0186] 1-Step Turbo TMB-ELISA, 250 mL (Thermo Scientific: #34022) [0187] Acid stop=0.1M HCL [0188] Sample enzyme conjugate (SEC) buffer Tween 20 (0.02% v/v), BSA 0.2% (w/v) in PBS [0189] Washing buffer 1.times.PBS, Tween 20 (0.1% v/v)

[0190] Method [0191] 1. Coat each well of a 96-well immunoplate with 100 .mu.l aliquots of 0.4 .mu.g/ml (dilute stock.times.3000=10 ul in 30 ml: coat 5 ml per plate) goat anti-human IgG antibody, diluted in PBS, incubate overnight at 4.degree. 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. [0192] 2. wash plate 3.times. with 200 .mu.l/well of washing buffer. [0193] 3. Block coated plate: add 200 ul 3% BSA in PBS: incubate 37 C 1 hr [0194] 4. Into blank plate, dispense 200 .mu.l SEC buffer into all wells except row B, cols 2-11 (blue, below). [0195] 5. Prepare 1 ug/ml solution of the human IgG1/kappa antibody in SEC buffer (.times.1000 diln)

TABLE-US-00001 [0195] 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 .mu.l diluent in every well +50 .mu.l sample +50 .mu.l sample Transfer 100 .mu.l Transfer 100 .mu.l 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 .mu.g/ml

[0196] BLOCKED UNCOATED PLATE: [0197] 6. Pipette 50 .mu.l/well of stds/unknowns into rows A-H, cols 1 and 7 (makes a .times.5 dilution). [0198] 7. Serially transfer 100 .mu.l across plate to achieve serial .times.3 dilution series. [0199] 8. Transfer 100 .mu.l from each well to the corresponding well of the BLOCKED anti-IgG-COATED plate. [0200] BLOCKED anti-IgG COATED PLATE: [0201] 9. Incubate at 37.degree. C. for 1 hr. Rinse all the wells 3.times. with washing buffer (200 .mu.l). [0202] 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). [0203] 11. Add 100 .mu.l of TMB Turbo substrate to each well, incubate in the dark at room temperature for 10 min. [0204] 12. Stop the reaction by adding 50 .mu.l of acid (0.1M HCl) to each well. [0205] 13. Read the optical density at 450 nm.

[0206] Protocol 4: AXL Binding ELISA

[0207] Materials [0208] 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. [0209] Goat anti-human kappa light chain peroxidase conjugate (Sigma, A-7164-1 ml) [0210] Nunc-Immuno Plate MaxiSorp (Life Technologies, 43945A) [0211] Plate washer Biotek LS405 [0212] 3% BSA: BSA 3% w/v in PBS [0213] PBS Tween: Tween 20 0.05% v/v in PBS [0214] PBS/Tween/BSA: BSA 0.5% w/v in PBS/Tween [0215] 1-Step Turbo TMB-ELISA (Thermo Scientific #3402)

[0216] Protocol [0217] 1. Dispense 50 .mu.l/well of human AXL-strep-His (1 ug/ml in PBS) [0218] 2. Cover with adhesive plate sealer and incubate at 4 C overnight. [0219] 3. Block: Dispense 50 .mu.l/well of 3% BSA and incubate for 1 hr 37 C, [0220] 4. Wash plate with PBS/Tween 3.times. [0221] 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. [0222] 6. Transfer 50 ul from antibody dilution plate onto washed, blocked AXL-coated plate [0223] 7. Incubate 37 C 1 hr [0224] 8. Wash plate with PBS/Tween 3.times. [0225] 9. Dispense anti-human IgG-HRP conjugate, diluted 1:1000 in PBS/Tween/BSA [0226] 10. Incubate 37 C 1 hr [0227] 11. Wash plate with PBS/Tween 3 [0228] 12. Wash plate with PBS 3.times. [0229] 13. Dispense 100 ul/well 1-Step Turbo TMB-ELISA substrate solution [0230] 14. Incubate 30 min at room temperature (or less if reaction is rapid) [0231] 15. Dispense 100 ul/well 0.6M HCl to stop the substrate reaction [0232] 16. Measure optical density at 450 nm

[0233] Protocol 4A: SPR Measurement of Antibody Affinity

[0234] Materials [0235] 1. Sensor Chip CM5 Biacore; Cat. #BR-1000-14 [0236] 2. Amine Coupling Kit (EDC, NHS, ethanolamine-HCl) Biacore; Cat. #BR-1000-50 [0237] 3. Immobilization buffer (10 mM Na acetate, pH 4.0) Biacore; Cat. #BR-1003-49 [0238] 4. 50 mM NaOH Biacore; Cat. #BR-1003-58 [0239] 5. Running buffer (PBS/Tween20 0.05% v/v) [0240] 6. Biacore T200 GE Healthcare [0241] 7. Regeneration solution: 10 mM HCl, 1 M NaCl

[0242] Coupling Method [0243] Activate flow cell 2 with NHS-EDC 420s at Sp/min, then inject human Axl-Strep-His (Evitria) (10 .mu.g/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. [0244] For the Fc fusion, Human Axl-Fc chimera (R&D Systems #154-AL) (5 .mu.g/mL in 10 mM sodium acetate, pH 4.5) was used with the above protocol. 16RU of AXL-Fc was coated.

[0245] Protocol with AXL-Strep-his Antigen [0246] Serial 10.times. dilutions of antibody, from 3000 nM, were made in PBS/Tween20. 2.times. regeneration cycles were used with 10 mM HCl, 1 M NaCl for 30s at 30 .mu.l/min for both cycles. Start-up solution was PBS/Tween20 and set to 3 cycles. [0247] Sample injection parameters were 120s at 30 .mu.l/min, with 600s dissociation time. [0248] Prime and normalise detector were run before sample application with experimental conditions 25 C and sample storage at 4 C. [0249] 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. [0250] Each antibody dilution was injected twice.

[0251] Protocol with AXL-Fc Chimera Antigen [0252] The above protocol was used except that the running buffer was HBSEP+, serial 10.times. dilutions of antibody were from 500 nM in HBSEP+ running buffer and were injected for 180 sec. [0253] Analysis used BIAevaluation software with the bivalent binding model.

[0254] Protocol 5: Capillary Isoelectric Focusing

[0255] Materials [0256] PA 800 plus (AB SCIEX) [0257] Anolyte Solution: 200 mM Phosphoric Acid (Sigma-Aldrich #345 245). [0258] 4.3M Urea (Sigma-Aldrich #U0631) in water. [0259] Catholyte Solution: 300 mM Sodium Hydroxide. [0260] 3M Urea (Sigma-Aldrich #U0631) in cIEF Gel (Beckman Coulter #477497). [0261] Chemical Mobiliser 350 mM Acetic Acid (Sigma-Aldrich #537 020). [0262] Pharmalyte 3-10 (GE Healthcare 17-0456-01). [0263] Cathodic Stabiliser 500 mM Arginine (Sigma-Aldrich #A5006). [0264] cIEF Peptide markers PI 4-10 (Beckman Coulter #A58481). [0265] Anodic Stabiliser: 200 mM Iminodiacetic Acid (Sigma-Aldrich #220 000). [0266] Neutral Capillary, 50 .mu.m i.d..times.45 cm, (Beckman Coulter #477 441)

[0267] Method [0268] Turn on the PA800+ machine and UV lamp, allowing it to warm up 30 mins before use. [0269] Clean the system electrodes and interface block with a damp Kimwipe. [0270] Prepare buffer trays as shown, with 1.5 mL reagent per vial, 1 mL of water in the waste [0271] 1. DDI water [0272] 2. Anolyte [0273] 3. Urea Solution [0274] 4. 3M urea/cIEF Gel [0275] 5. Chemical Mobilizer [0276] 6. Waste [0277] 7. Catholyte [0278] 8. Chemical Mobilizer [0279] 9. BI (Inlet Buffer Tray) [0280] 10. BO (Outlet Buffer Tray) [0281] NOTE. Each set of buffer vials is good for 6 consecutive runs or for 24 hours inside the instrument. [0282] Insert the capillary cartridge into the system and close the front panel. [0283] 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. [0284] 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.degree. C. to 8.degree. C. for up to 1 day.

TABLE-US-00002 [0284] Volume per Reagent sample (.mu.l) 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

[0285] Desalt and concentrate each antibody (to about 5 mg/ml) in 2M urea using Amicon Ultra 0.5 mL centrifugal filters (Sigma Z677108). [0286] Mix 200 .mu.L of master mix with 10 .mu.L 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 .mu.L 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. [0287] Run the "cIEF Conditioning--PA 800 plus.met" method to condition the column. [0288] Rinse for 5 min at 50 psi with Chemical Mobilizer. See FIG. 2.10. [0289] 2 Rinse for 2 min at 50 psi with DDI water. [0290] 3 Rinse for 5 min at 50 psi with cIEF gel. [0291] 4 Submerge both of the capillary ends in vials filled with DDI water. [0292] Use the "cIEF Separation--PA 800 plus.met" method to create a sequence containing all protein samples and a blank. [0293] Rinse for 3 min at 50 psi with Urea Solution. See FIG. 2.11. [0294] Rinse for 2 min at 50 psi with DDI water. [0295] Inject sample for 99.0 sec at 25 psi. [0296] Water dip by submerging both capillary ends in DDI water. [0297] Focusing step, 15 min at 25 kV under normal polarity (Time=0). [0298] Chemical mobilization, 30 min at 30 kV under normal polarity (Time=15 min). [0299] Stop data collection (Time=45 min). [0300] Rinse for 2 min at 50 psi with DDI water (Time=45.10 min). [0301] Submerged both of the capillary ends in DDI water (Time=47.20 min). [0302] End the method (Time=47.30 min). [0303] Put the "cIEF Shutdown--PA 800 plus.met" method at the end of the sequence to rinse the capillary and turn off the UV. [0304] Rinse for 2 min at 50 psi with DDI water. See FIG. 2.12. [0305] Rinse for 5 min at 50 psi with cIEF gel. [0306] Turn off the UV lamp. [0307] Submerge both of the capillary ends in vials filled with DDI water. [0308] 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.

[0309] Protocol 6: Protein Thermal Shift Protocol

[0310] Materials [0311] 96 well optical plate semi-skirted (Stariab cat. L1402-9700). [0312] Protein thermal shift dye kit (Life Technologies cat. 446148). [0313] Microamp optical adhesive film (Applied Biosystems) [0314] 7500 Real-Time PCR System (Life Technologies) [0315] Test items: Antibodies from Evitria and Spirogen [0316] Protein thermal shift v1.2 software (Life Technologies)

[0317] Method

[0318] Protein thermal shift dye (2.5 .mu.L 1:1000 dilution) was added to sample proteins (17.5 .mu.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:

TABLE-US-00003 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

[0319] Temperature cycling on the ABI 7500 set up:

TABLE-US-00004 Reaction vol 20 .mu.l Ramp mode continuous Step Ramp rate Temp .degree. C. Time (mm:ss) 1 100% 25.0 02:00 2 1% 99.0 02:00

[0320] Data analysis and derivation of Tm data were done using the software following the manufacturers instructions.

[0321] Protocol 7: HPLC Size Exclusion Chromatography

[0322] Materials [0323] Shimadzu HPLC system, or equivalent, consisting of the following, or equivalent: [0324] 2.times. DGU-20A.sub.SR Prominence Degassing units [0325] 2.times. LC-20AD.sub.XR Nexera Pumps [0326] SIL-20AC.sub.XR Nexera Autosampler [0327] CTO-20AC Prominence Column oven [0328] SPD-M30A Prominence DAD detector [0329] Computer with LabSolutions software. [0330] TSKgeI Super SW mAb HTP 4 um 4.6 mm.times.15 cm [0331] 200 mM Potassium Phosphate, 250 mM Potassium Chloride, 10% v/v i-Propanol, pH 6.95.

[0332] Sample Preparation [0333] Mobile Phase: 200 mM Potassium Phosphate, 250 mM Potassium Chloride, 10% v/v i-Propanol, pH 6.95. [0334] Analytical Sample: Inject 2-20 .mu.L of neat ADC sample (at least 1 mg/ml for best results).

[0335] Typically 10 .mu.L of 5 mg/ml gives good results.

[0336] HPLC Parameters [0337] Method File name: MSOP-018 [0338] HPLC Column: TSKgeI Super SW mAb HTP 4 um 4.6 mm.times.15 cm [0339] Flow Rate: 0.5 ml/min [0340] Injection volume: 2-20 .mu.L [0341] Detection, UV: 280 nm [0342] 330 nm (for information only) [0343] Column Temp: ambient temperature [0344] Autosampler Temp: 15.degree. C. [0345] Gradient: Isocratic

[0346] Method [0347] Intact ADCs typically elute at .about.16-18 minutes. [0348] Aggregates typically elute at .about.11-14 minutes. [0349] Low molecular weight species typically elute at .about.>20 minutes.

[0350] Protocol 8: Hydrophobic Interaction Chromatograph

[0351] Materials [0352] HPLC system, or equivalent, consisting of the following, or equivalent: [0353] SRD-3600 SOLVENT RACK, 6 DEGASS. LINES [0354] HPG-3400RS PUMP (Thermo Scientific) [0355] HPG-3200RS PUMP (Thermo Scientific) [0356] WPS-3000TFC ANALYTICAL AUTOSAMPLER (Thermo Scientific) [0357] TCC-3000RS COLUMN THERMOSTAT (Thermo Scientific) [0358] DAD-3000RS DETECTOR (Thermo Scientific) [0359] Computer with Chromeleon software (Thermo Scientific) [0360] Proteomix HIC Butyl-NP5, 5 um, non-porous, 4.6.times.35 mm (Sepax) column [0361] Ammonium sulfate ((NH.sub.4).sub.2SO.sub.4) [0362] Sodium acetate (NaOAc) [0363] i-Propanol [0364] Water, HPLC grade [0365] Mobile Phase A: 1.25 M (NH.sub.4).sub.2SO.sub.4, 25 mM NaOAc (pH 5.50) [0366] Mobile Phase B: 75% 25 mM NaOAc (pH 5.50), 25% i-Propanol [0367] Blank Solution: Water

[0368] Sample Preparation [0369] Analytical Sample: .ltoreq.10 .mu.L of neat ADC sample at a concentration of 1-5 mg/mL.

[0370] HPLC Parameters [0371] Method File name: HIC_Gradient_1_AB [0372] HPLC Column: Proteomix HIC Butyl-NP5, Sum, non-porous, 4.6.times.35 mm (Sepax) [0373] Flow Rate: 0.8 ml/min [0374] Injection volume: 5 10 .mu.L [0375] Detection, UV: 214 nm [0376] 330 nm (for information only) [0377] Column Temp: 25.degree. C. [0378] Autosampler Temp: 10.degree. C.

[0379] Gradient

TABLE-US-00005 Time (min) % B 0 0 1 0 13 100 14 100 14.1 0 18 0

[0380] Method

[0381] 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.

[0382] Protocol 9: Reverse Phase Chromatograph

[0383] Materials [0384] HPLC system, or equivalent, consisting of the following, or equivalent: [0385] SRD-3600 SOLVENT RACK, 6 DEGASS. LINES [0386] HPG-3400RS PUMP (Thermo Scientific) [0387] HPG-3200RS PUMP (Thermo Scientific) [0388] WPS-3000TFC ANALYTICAL AUTOSAMPLER (Thermo Scientific) [0389] TCC-3000RS COLUMN THERMOSTAT (Thermo Scientific) [0390] DAD-3000RS DETECTOR (Thermo Scientific) [0391] Computer with Chromeleon software (Thermo Scientific) [0392] Aeris Widepore XB-C18, 200 .ANG., 3.6 .mu.m, 2.1.times.150 mm (Phenomenex, 00F-4482-AN) [0393] Acetonitrile, HPLC grade. [0394] Water, HPLC grade. [0395] Trifluoroacetic acid (TFA), HPLC grade. [0396] 100 mM NaBorate, pH 8.4 [0397] 500 mM DTT [0398] 49:49:2 Acetonitrile/Water/Formic acid [0399] Mobile Phase A: Water+0.1% v/v TFA. [0400] Mobile Phase B: Acetonitrile+0.1% v/v TFA. [0401] Blank Solution: 1:1 v/v Acetonitrile/Water.

[0402] Sample Preparation

[0403] To 40 .mu.L of sample (5 mg/ml) add water, 30 .mu.l, NaBorate, 20 .mu.l, and DTT (500 mM), 10 .mu.L. Incubate at 37.degree. C., 30 min, then add 100 .mu.l of 49:49:2 Acetonitrle/Water/Formic acid.

[0404] HPLC Parameters [0405] Method File name: RP_Aeris_Column6 [0406] HPLC Column: Aeris Widepore XB-C18, 200 .ANG., 3.6 .mu.m, 2.1.times.150 mm (Phenomenex, 00F-4482-AN) [0407] Flow Rate: 1.0 ml/min [0408] Injection volume: 10 .mu.l (or full loop) [0409] Detection, UV: 214 nm [0410] 330 nm (for information only) [0411] Column Temp: 80.degree. C. [0412] Autosampler Temp: 15.degree. C.

[0413] Gradient

TABLE-US-00006 Time (minutes) % B 0 22.5 1 22.5 11 50 11.5 90 13.5 90 14.5 22.5 16 22.5

[0414] Method

[0415] 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

[0416] 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. [0417] 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. [0418] 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. [0419] 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. [0420] 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. [0421] 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.

[0422] A stability assay was performed during which the antibodies were heated in sterile PBS at 40.degree. 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).

[0423] 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-US-00007 TABLE 1 RP HPLC SEC % LC2 peak Expression (.mu.g/mL) monomer HIC retention % protein in . . . 280 nm time min Tm 280 nm Antibody T F CHO CHO F F CHO pl (.degree. 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

[0424] Binding of the antibodies to AXL antigens indicated that binding was unusually sensitive to both antigen preparation and presentation and antibody geometry.

[0425] 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).

[0426] 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).

[0427] 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-US-00008 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

[0428] 5 .ANG.+ set The FW residues in the 5 .ANG. CDR envelope, defined by the homology model, together with the canonical, vernier and VH/VK interface residues [0429] 1H12 The anti-AXL mouse monoclonal antibody [0430] 1H12 VK VK of mouse 1H12 antibody [0431] 1H12RKA1 Humanised version, A1, of 1H12 VK [0432] 1H12RHA Humanised version, A, of 1H12 VH [0433] 1H12RHA x IgG1k antibody comprising the VH and VK constructs 1H12RHA and [0434] 1H12RKA 1H12RKA respectively, functionally contiguous with the constant regions of human IgG1 and Ig-kappa heavy and light chains respectively [0435] A Adenine [0436] .ANG. Angstrom [0437] Ac acetyl [0438] Acm acetamidomethyl [0439] Alloc allyloxycarbonyl [0440] B7 The anti-LPA antibody product of mouse hybridoma clone B7 [0441] Boc di-tert-butyl dicarbonate [0442] bp base pairs [0443] Bzl benzyl, where Bzl-OMe is methoxybenzyl and Bzl-Me is methylbenzene [0444] C Cytosine [0445] Cbz or Z benzyloxy-carbonyl, where Z--Cl and Z--Br are chloro- and bromobenzyloxy carbonyl respectively [0446] CDR Complementarity determining region in the immunoglobulin variable regions, defined using the Kabat numbering system [0447] CHO Chinese hamster ovary cell line [0448] D-gene Diversity gene [0449] DMF N,N-dimethylformamide [0450] DNA Deoxyribonucleic acid [0451] Dnp dinitrophenyl [0452] DTT dithiothreitol [0453] Fmoc 9H-fluoren-9-ylmethoxycarbonyl [0454] FW Framework region: the immunoglobulin variable regions excluding the CDR regions [0455] G Guanine [0456] IgG Immunoglobulin G [0457] imp N-10 imine protecting group: 3-(2-methoxyethoxy)propanoate-Val-Ala-PAB [0458] MC-OSu maleimidocaproyl-O--N-succinimide [0459] J-gene Joining gene [0460] Kabat an immunoglobulin alignment and numbering system pioneered by Elvin A Kabat [0461] mAb monoclonal antibody [0462] Moc methoxycarbonyl [0463] MP maleimidopropanamide [0464] Mtr 4-methoxy-2,3,6-trimethtylbenzenesulfonyl [0465] PAB para-aminobenzyloxycarbonyl [0466] PEG ethyleneoxy [0467] PNZ p-nitrobenzyl carbamate [0468] Psec 2-(phenylsulfonyl)ethoxycarbonyl [0469] T Thymine [0470] TBDMS tert-butyldimethylsilyl [0471] TBDPS tert-butyldiphenylsilyl [0472] t-Bu tert-butyl [0473] Teoc 2-(trimethylsilyl)ethoxycarbonyl [0474] Tos tosyl [0475] Troc 2,2,2-trichlorethoxycarbonyl chloride [0476] Trt trityl [0477] 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. [0478] VCI Framework residue classified as vernier or canonical or VH-VL interface [0479] V-gene The gene segment that is rearranged, together with a J (and D for VH) gene, to generate a complete VK (or VH) [0480] VH Immunoglobulin heavy chain variable region [0481] VK Immunoglobulin kappa light chain variable region [0482] Xan xanthyl

REFERENCES

[0482] [0483] [1] C. Chothia, et al., "Domain association in immunoglobulin molecules. The packing of variable domains," J Mol. Biol. 186(3), 651 (1985). [0484] [2] J. Foote and G. Winter, "Antibody framework residues affecting the conformation of the hypervariable loops," J Mol. Biol. 224(2), 487 (1992). [0485] [3] E. A Kabat, et al., sequences of proteins of immunological interest, 5 ed. (NIH National Technical Information Service, 1991). [0486] [4] V. Morea, A. M. Lesk, and A. Tramontano, "Antibody modeling: implications for engineering and design," Methods 20(3), 267 (2000).

STATEMENTS OF DISCLOSURE

[0487] 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: [0488] (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; [0489] (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; [0490] (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; [0491] (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; [0492] (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; [0493] (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; [0494] (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; [0495] (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; [0496] (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; [0497] (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; [0498] (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; [0499] (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; [0500] (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; [0501] (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 [0502] (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.sup.-6 M. 5. The humanized antibody according to statement 3, wherein said antibody binds human AXL with an affinity (Kd) of at least 10.sup.-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').sub.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; [0503] 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; [0504] 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.

TABLE-US-00009 [0504] 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

Sequence CWU 1

1

101146PRTArtificial sequenceSynthetic heavy chain variable region 1H12VH 1Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Glu Val Gln Leu Val Glu Ser Gly 20 25 30Gly Asp Leu Val Lys Pro Gly Gly Ser Leu Lys Leu Ser Cys Ala Ala 35 40 45Ser Gly Phe Thr Phe Ser Ser Tyr Gly Met Ser Trp Val Arg Gln Thr 50 55 60Pro Asp Lys Arg Leu Glu Trp Val Ala Thr Ile Ser Ser Gly Gly Ser65 70 75 80Tyr Thr Tyr Tyr Pro Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg 85 90 95Asp Asn Ala Lys Asn Thr Leu Tyr Leu Gln Met Ser Ser Leu Lys Ser 100 105 110Glu Asp Thr Ala Met Tyr Tyr Cys Ala Arg His Pro Ile Tyr Tyr Thr 115 120 125Tyr Asp Asp Thr Met Asp Tyr Trp Gly Gln Gly Thr Ser Val Thr Val 130 135 140Ser Ser1452146PRTArtificial sequenceSynthetic heavy chain variable region 1H12RHA 2Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Gln Val Gln Leu Val Glu Ser Gly 20 25 30Gly Gly Val Val Gln Pro Gly Arg Ser Leu Arg Leu Ser Cys Ala Ala 35 40 45Ser Gly Phe Thr Phe Ser Ser Tyr Gly Met Ser Trp Val Arg Gln Ala 50 55 60Pro Gly Lys Gly Leu Glu Trp Val Ala Thr Ile Ser Ser Gly Gly Ser65 70 75 80Tyr Thr Tyr Tyr Pro Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg 85 90 95Asp Asn Ser Lys Asn Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala 100 105 110Glu Asp Thr Ala Val Tyr Tyr Cys Ala Arg His Pro Ile Tyr Tyr Thr 115 120 125Tyr Asp Asp Thr Met Asp Tyr Trp Gly Gln Gly Thr Thr Val Thr Val 130 135 140Ser Ser1453146PRTArtificial sequenceSynthetic heavy chain variable region 1H12RHB 3Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Glu Val Gln Leu Val Glu Ser Gly 20 25 30Gly Gly Leu Val Gln Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala Ala 35 40 45Ser Gly Phe Thr Phe Ser Ser Tyr Gly Met Ser Trp Val Arg Gln Ala 50 55 60Pro Gly Lys Gly Leu Glu Trp Val Ala Thr Ile Ser Ser Gly Gly Ser65 70 75 80Tyr Thr Tyr Tyr Pro Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg 85 90 95Asp Asn Ala Lys Asn Ser Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala 100 105 110Glu Asp Thr Ala Val Tyr Tyr Cys Ala Arg His Pro Ile Tyr Tyr Thr 115 120 125Tyr Asp Asp Thr Met Asp Tyr Trp Gly Gln Gly Thr Leu Val Thr Val 130 135 140Ser Ser1454132PRTArtificial sequenceSynthetic light chain variable region 1H12VK 4Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Glu Asn Val Leu Thr Gln Ser Pro 20 25 30Ala Ile Met Ala Ala Ser Pro Gly Glu Lys Val Thr Met Thr Cys Ser 35 40 45Ala Ser Ser Ser Val Ser Ser Gly Asn Phe His Trp Tyr Gln Gln Lys 50 55 60Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr Arg Thr Ser Asn Leu Ala65 70 75 80Ser Gly Val Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Ser Tyr 85 90 95Ser Leu Thr Ile Ser Ser Met Glu Ala Glu Asp Ala Ala Thr Tyr Tyr 100 105 110Cys Gln Gln Trp Ser Gly Tyr Pro Trp Thr Phe Gly Gly Gly Thr Lys 115 120 125Leu Glu Ile Lys 1305132PRTArtificial sequenceSynthetic light chain variable region 1H12RKA 5Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Glu Ile Val Leu Thr Gln Ser Pro 20 25 30Ala Thr Leu Ser Leu Ser Pro Gly Glu Arg Ala Thr Leu Ser Cys Ser 35 40 45Ala Ser Ser Ser Val Ser Ser Gly Asn Phe His Trp Tyr Gln Gln Lys 50 55 60Pro Gly Leu Ala Pro Arg Leu Leu Ile Tyr Arg Thr Ser Asn Leu Ala65 70 75 80Ser Gly Ile Pro Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe 85 90 95Thr Leu Thr Ile Ser Arg Leu Glu Pro Glu Asp Phe Ala Val Tyr Tyr 100 105 110Cys Gln Gln Trp Ser Gly Tyr Pro Trp Thr Phe Gly Pro Gly Thr Lys 115 120 125Val Asp Ile Lys 1306132PRTArtificial sequenceSynthetic light chain variable region 1H12RKA1 6Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Glu Asn Val Leu Thr Gln Ser Pro 20 25 30Ala Thr Leu Ser Leu Ser Pro Gly Glu Arg Ala Thr Leu Ser Cys Ser 35 40 45Ala Ser Ser Ser Val Ser Ser Gly Asn Phe His Trp Tyr Gln Gln Lys 50 55 60Pro Gly Leu Ala Pro Arg Leu Trp Ile Tyr Arg Thr Ser Asn Leu Ala65 70 75 80Ser Gly Ile Pro Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Tyr 85 90 95Thr Leu Thr Ile Ser Arg Leu Glu Pro Glu Asp Phe Ala Val Tyr Tyr 100 105 110Cys Gln Gln Trp Ser Gly Tyr Pro Trp Thr Phe Gly Pro Gly Thr Lys 115 120 125Val Asp Ile Lys 1307132PRTArtificial sequenceSynthetic light chain variable region 1H12RKB 7Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Glu Ile Val Leu Thr Gln Ser Pro 20 25 30Gly Thr Leu Ser Leu Ser Pro Gly Glu Arg Ala Thr Leu Ser Cys Ser 35 40 45Ala Ser Ser Ser Val Ser Ser Gly Asn Phe His Trp Tyr Gln Gln Lys 50 55 60Pro Gly Leu Ala Pro Arg Leu Leu Ile Tyr Arg Thr Ser Asn Leu Ala65 70 75 80Ser Gly Ile Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe 85 90 95Thr Leu Thr Ile Ser Ser Leu Glu Pro Glu Asp Phe Ala Val Tyr Tyr 100 105 110Cys Gln Gln Trp Ser Gly Tyr Pro Trp Thr Phe Gly Gly Gly Thr Lys 115 120 125Leu Glu Ile Lys 1308132PRTArtificial sequenceSynthetic light chain variable region 1H12RKB1 8Met Gly Phe Lys Met Glu Ser Gln Phe Gln Val Phe Val Phe Val Phe1 5 10 15Leu Trp Leu Ser Gly Val Asp Gly Glu Asn Val Leu Thr Gln Ser Pro 20 25 30Gly Thr Leu Ser Leu Ser Pro Gly Glu Arg Ala Thr Leu Ser Cys Ser 35 40 45Ala Ser Ser Ser Val Ser Ser Gly Asn Phe His Trp Tyr Gln Gln Lys 50 55 60Pro Gly Leu Ala Pro Arg Leu Trp Ile Tyr Arg Thr Ser Asn Leu Ala65 70 75 80Ser Gly Ile Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Tyr 85 90 95Thr Leu Thr Ile Ser Ser Leu Glu Pro Glu Asp Phe Ala Val Tyr Tyr 100 105 110Cys Gln Gln Trp Ser Gly Tyr Pro Trp Thr Phe Gly Gly Gly Thr Lys 115 120 125Leu Glu Ile Lys 1309894PRTHomo sapiens 9Met Ala Trp Arg Cys Pro Arg Met Gly Arg Val Pro Leu Ala Trp Cys1 5 10 15Leu Ala Leu Cys Gly Trp Ala Cys Met Ala Pro Arg Gly Thr Gln Ala 20 25 30Glu Glu Ser Pro Phe Val Gly Asn Pro Gly Asn Ile Thr Gly Ala Arg 35 40 45Gly Leu Thr Gly Thr Leu Arg Cys Gln Leu Gln Val Gln Gly Glu Pro 50 55 60Pro Glu Val His Trp Leu Arg Asp Gly Gln Ile Leu Glu Leu Ala Asp65 70 75 80Ser Thr Gln Thr Gln Val Pro Leu Gly Glu Asp Glu Gln Asp Asp Trp 85 90 95Ile Val Val Ser Gln Leu Arg Ile Thr Ser Leu Gln Leu Ser Asp Thr 100 105 110Gly Gln Tyr Gln Cys Leu Val Phe Leu Gly His Gln Thr Phe Val Ser 115 120 125Gln Pro Gly Tyr Val Gly Leu Glu Gly Leu Pro Tyr Phe Leu Glu Glu 130 135 140Pro Glu Asp Arg Thr Val Ala Ala Asn Thr Pro Phe Asn Leu Ser Cys145 150 155 160Gln Ala Gln Gly Pro Pro Glu Pro Val Asp Leu Leu Trp Leu Gln Asp 165 170 175Ala Val Pro Leu Ala Thr Ala Pro Gly His Gly Pro Gln Arg Ser Leu 180 185 190His Val Pro Gly Leu Asn Lys Thr Ser Ser Phe Ser Cys Glu Ala His 195 200 205Asn Ala Lys Gly Val Thr Thr Ser Arg Thr Ala Thr Ile Thr Val Leu 210 215 220Pro Gln Gln Pro Arg Asn Leu His Leu Val Ser Arg Gln Pro Thr Glu225 230 235 240Leu Glu Val Ala Trp Thr Pro Gly Leu Ser Gly Ile Tyr Pro Leu Thr 245 250 255His Cys Thr Leu Gln Ala Val Leu Ser Asp Asp Gly Met Gly Ile Gln 260 265 270Ala Gly Glu Pro Asp Pro Pro Glu Glu Pro Leu Thr Ser Gln Ala Ser 275 280 285Val Pro Pro His Gln Leu Arg Leu Gly Ser Leu His Pro His Thr Pro 290 295 300Tyr His Ile Arg Val Ala Cys Thr Ser Ser Gln Gly Pro Ser Ser Trp305 310 315 320Thr His Trp Leu Pro Val Glu Thr Pro Glu Gly Val Pro Leu Gly Pro 325 330 335Pro Glu Asn Ile Ser Ala Thr Arg Asn Gly Ser Gln Ala Phe Val His 340 345 350Trp Gln Glu Pro Arg Ala Pro Leu Gln Gly Thr Leu Leu Gly Tyr Arg 355 360 365Leu Ala Tyr Gln Gly Gln Asp Thr Pro Glu Val Leu Met Asp Ile Gly 370 375 380Leu Arg Gln Glu Val Thr Leu Glu Leu Gln Gly Asp Gly Ser Val Ser385 390 395 400Asn Leu Thr Val Cys Val Ala Ala Tyr Thr Ala Ala Gly Asp Gly Pro 405 410 415Trp Ser Leu Pro Val Pro Leu Glu Ala Trp Arg Pro Gly Gln Ala Gln 420 425 430Pro Val His Gln Leu Val Lys Glu Pro Ser Thr Pro Ala Phe Ser Trp 435 440 445Pro Trp Trp Tyr Val Leu Leu Gly Ala Val Val Ala Ala Ala Cys Val 450 455 460Leu Ile Leu Ala Leu Phe Leu Val His Arg Arg Lys Lys Glu Thr Arg465 470 475 480Tyr Gly Glu Val Phe Glu Pro Thr Val Glu Arg Gly Glu Leu Val Val 485 490 495Arg Tyr Arg Val Arg Lys Ser Tyr Ser Arg Arg Thr Thr Glu Ala Thr 500 505 510Leu Asn Ser Leu Gly Ile Ser Glu Glu Leu Lys Glu Lys Leu Arg Asp 515 520 525Val Met Val Asp Arg His Lys Val Ala Leu Gly Lys Thr Leu Gly Glu 530 535 540Gly Glu Phe Gly Ala Val Met Glu Gly Gln Leu Asn Gln Asp Asp Ser545 550 555 560Ile Leu Lys Val Ala Val Lys Thr Met Lys Ile Ala Ile Cys Thr Arg 565 570 575Ser Glu Leu Glu Asp Phe Leu Ser Glu Ala Val Cys Met Lys Glu Phe 580 585 590Asp His Pro Asn Val Met Arg Leu Ile Gly Val Cys Phe Gln Gly Ser 595 600 605Glu Arg Glu Ser Phe Pro Ala Pro Val Val Ile Leu Pro Phe Met Lys 610 615 620His Gly Asp Leu His Ser Phe Leu Leu Tyr Ser Arg Leu Gly Asp Gln625 630 635 640Pro Val Tyr Leu Pro Thr Gln Met Leu Val Lys Phe Met Ala Asp Ile 645 650 655Ala Ser Gly Met Glu Tyr Leu Ser Thr Lys Arg Phe Ile His Arg Asp 660 665 670Leu Ala Ala Arg Asn Cys Met Leu Asn Glu Asn Met Ser Val Cys Val 675 680 685Ala Asp Phe Gly Leu Ser Lys Lys Ile Tyr Asn Gly Asp Tyr Tyr Arg 690 695 700Gln Gly Arg Ile Ala Lys Met Pro Val Lys Trp Ile Ala Ile Glu Ser705 710 715 720Leu Ala Asp Arg Val Tyr Thr Ser Lys Ser Asp Val Trp Ser Phe Gly 725 730 735Val Thr Met Trp Glu Ile Ala Thr Arg Gly Gln Thr Pro Tyr Pro Gly 740 745 750Val Glu Asn Ser Glu Ile Tyr Asp Tyr Leu Arg Arg Gly Asn Arg Leu 755 760 765Lys Gln Pro Ala Asp Cys Leu Asp Gly Leu Tyr Ala Leu Met Ser Arg 770 775 780Cys Trp Glu Leu Asn Pro Gln Asp Arg Pro Ser Phe Thr Glu Leu Arg785 790 795 800Glu Asp Leu Glu Asn Thr Leu Lys Ala Leu Pro Pro Ala Gln Glu Pro 805 810 815Asp Glu Ile Leu Tyr Val Asn Met Asp Glu Gly Gly Gly Tyr Pro Glu 820 825 830Pro Pro Gly Ala Ala Gly Gly Ala Asp Pro Pro Thr Gln Pro Asp Pro 835 840 845Lys Asp Ser Cys Ser Cys Leu Thr Ala Ala Glu Val His Pro Ala Gly 850 855 860Arg Tyr Val Leu Cys Pro Ser Thr Thr Pro Ser Pro Ala Gln Pro Ala865 870 875 880Asp Arg Gly Ser Pro Ala Ala Pro Gly Gln Glu Asp Gly Ala 885 89010888PRTMus musculus 10Met Gly Arg Val Pro Leu Ala Trp Trp Leu Ala Leu Cys Cys Trp Gly1 5 10 15Cys Ala Ala His Lys Asp Thr Gln Thr Glu Ala Gly Ser Pro Phe Val 20 25 30Gly Asn Pro Gly Asn Ile Thr Gly Ala Arg Gly Leu Thr Gly Thr Leu 35 40 45Arg Cys Glu Leu Gln Val Gln Gly Glu Pro Pro Glu Val Val Trp Leu 50 55 60Arg Asp Gly Gln Ile Leu Glu Leu Ala Asp Asn Thr Gln Thr Gln Val65 70 75 80Pro Leu Gly Glu Asp Trp Gln Asp Glu Trp Lys Val Val Ser Gln Leu 85 90 95Arg Ile Ser Ala Leu Gln Leu Ser Asp Ala Gly Glu Tyr Gln Cys Met 100 105 110Val His Leu Glu Gly Arg Thr Phe Val Ser Gln Pro Gly Phe Val Gly 115 120 125Leu Glu Gly Leu Pro Tyr Phe Leu Glu Glu Pro Glu Asp Lys Ala Val 130 135 140Pro Ala Asn Thr Pro Phe Asn Leu Ser Cys Gln Ala Gln Gly Pro Pro145 150 155 160Glu Pro Val Thr Leu Leu Trp Leu Gln Asp Ala Val Pro Leu Ala Pro 165 170 175Val Thr Gly His Ser Ser Gln His Ser Leu Gln Thr Pro Gly Leu Asn 180 185 190Lys Thr Ser Ser Phe Ser Cys Glu Ala His Asn Ala Lys Gly Val Thr 195 200 205Thr Ser Arg Thr Ala Thr Ile Thr Val Leu Pro Gln Arg Pro His His 210 215 220Leu His Val Val Ser Arg Gln Pro Thr Glu Leu Glu Val Ala Trp Thr225 230 235 240Pro Gly Leu Ser Gly Ile Tyr Pro Leu Thr His Cys Asn Leu Gln Ala 245 250 255Val Leu Ser Asp Asp Gly Val Gly Ile Trp Leu Gly Lys Ser Asp Pro 260 265 270Pro Glu Asp Pro Leu Thr Leu Gln Val Ser Val Pro Pro His Gln Leu 275 280 285Arg Leu Glu Lys Leu Leu Pro His Thr Pro Tyr His Ile Arg Ile Ser 290 295 300Cys Ser Ser Ser Gln Gly Pro Ser Pro Trp Thr His Trp Leu Pro Val305 310 315 320Glu Thr Thr Glu Gly Val Pro Leu Gly Pro Pro Glu Asn Val Ser Ala 325 330 335Met Arg Asn Gly Ser Gln Val Leu Val Arg Trp Gln Glu Pro Arg Val 340 345 350Pro Leu Gln Gly Thr Leu Leu Gly Tyr Arg Leu Ala Tyr Arg Gly Gln 355 360 365Asp Thr Pro Glu Val Leu Met Asp Ile Gly Leu Thr Arg Glu Val Thr 370 375 380Leu Glu Leu Arg Gly Asp Arg Pro Val Ala Asn Leu Thr Val Ser Val385 390 395 400Thr Ala Tyr Thr Ser Ala Gly Asp Gly Pro Trp Ser Leu Pro Val Pro

405 410 415Leu Glu Pro Trp Arg Pro Gly Gln Gly Gln Pro Leu His His Leu Val 420 425 430Ser Glu Pro Pro Pro Arg Ala Phe Ser Trp Pro Trp Trp Tyr Val Leu 435 440 445Leu Gly Ala Leu Val Ala Ala Ala Cys Val Leu Ile Leu Ala Leu Phe 450 455 460Leu Val His Arg Arg Lys Lys Glu Thr Arg Tyr Gly Glu Val Phe Glu465 470 475 480Pro Thr Val Glu Arg Gly Glu Leu Val Val Arg Tyr Arg Val Arg Lys 485 490 495Ser Tyr Ser Arg Arg Thr Thr Glu Ala Thr Leu Asn Ser Leu Gly Ile 500 505 510Ser Glu Glu Leu Lys Glu Lys Leu Arg Asp Val Met Val Asp Arg His 515 520 525Lys Val Ala Leu Gly Lys Thr Leu Gly Glu Gly Glu Phe Gly Ala Val 530 535 540Met Glu Gly Gln Leu Asn Gln Asp Asp Ser Ile Leu Lys Val Ala Val545 550 555 560Lys Thr Met Lys Ile Ala Ile Cys Thr Arg Ser Glu Leu Glu Asp Phe 565 570 575Leu Ser Glu Ala Val Cys Met Lys Glu Phe Asp His Pro Asn Val Met 580 585 590Arg Leu Ile Gly Val Cys Phe Gln Gly Ser Asp Arg Glu Gly Phe Pro 595 600 605Glu Pro Val Val Ile Leu Pro Phe Met Lys His Gly Asp Leu His Ser 610 615 620Phe Leu Leu Tyr Ser Arg Leu Gly Asp Gln Pro Val Phe Leu Pro Thr625 630 635 640Gln Met Leu Val Lys Phe Met Ala Asp Ile Ala Ser Gly Met Glu Tyr 645 650 655Leu Ser Thr Lys Arg Phe Ile His Arg Asp Leu Ala Ala Arg Asn Cys 660 665 670Met Leu Asn Glu Asn Met Ser Val Cys Val Ala Asp Phe Gly Leu Ser 675 680 685Lys Lys Ile Tyr Asn Gly Asp Tyr Tyr Arg Gln Gly Arg Ile Ala Lys 690 695 700Met Pro Val Lys Trp Ile Ala Ile Glu Ser Leu Ala Asp Arg Val Tyr705 710 715 720Thr Ser Lys Ser Asp Val Trp Ser Phe Gly Val Thr Met Trp Glu Ile 725 730 735Ala Thr Arg Gly Gln Thr Pro Tyr Pro Gly Val Glu Asn Ser Glu Ile 740 745 750Tyr Asp Tyr Leu Arg Gln Gly Asn Arg Leu Lys Gln Pro Val Asp Cys 755 760 765Leu Asp Gly Leu Tyr Ala Leu Met Ser Arg Cys Trp Glu Leu Asn Pro 770 775 780Arg Asp Arg Pro Ser Phe Ala Glu Leu Arg Glu Asp Leu Glu Asn Thr785 790 795 800Leu Lys Ala Leu Pro Pro Ala Gln Glu Pro Asp Glu Ile Leu Tyr Val 805 810 815Asn Met Asp Glu Gly Gly Ser His Leu Glu Pro Arg Gly Ala Ala Gly 820 825 830Gly Ala Asp Pro Pro Thr Gln Pro Asp Pro Lys Asp Ser Cys Ser Cys 835 840 845Leu Thr Ala Ala Asp Val His Ser Ala Gly Arg Tyr Val Leu Cys Pro 850 855 860Ser Thr Ala Pro Gly Pro Thr Leu Ser Ala Asp Arg Gly Cys Pro Ala865 870 875 880Pro Pro Gly Gln Glu Asp Gly Ala 885

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed