CN110785434A - Methods of treatment using IL-13R antibodies - Google Patents

Methods of treatment using IL-13R antibodies Download PDF

Info

Publication number
CN110785434A
CN110785434A CN201880042193.3A CN201880042193A CN110785434A CN 110785434 A CN110785434 A CN 110785434A CN 201880042193 A CN201880042193 A CN 201880042193A CN 110785434 A CN110785434 A CN 110785434A
Authority
CN
China
Prior art keywords
ser
antibody
val
gly
thr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201880042193.3A
Other languages
Chinese (zh)
Inventor
B·林德马克
A·G·L·黄
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Csl Co
Ya Shi Kang Private Co Ltd
Original Assignee
Csl Co
Ya Shi Kang Private Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Csl Co, Ya Shi Kang Private Co Ltd filed Critical Csl Co
Priority claimed from PCT/SG2018/050319 external-priority patent/WO2019004943A1/en
Publication of CN110785434A publication Critical patent/CN110785434A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides a method of treating cutaneous T-cell lymphoma (CTCL), in particular mycosis fungoides and/or sezary syndrome, comprising administering to a patient in need thereof a therapeutically effective amount of an antagonist antibody or binding fragment thereof specific for an IL-13 receptor.

Description

Methods of treatment using IL-13R antibodies
The present disclosure relates to a therapy for the treatment of cutaneous T-cell lymphoma, in particular mycosis fungoides (mycosis fungoides) and/or sezary syndrome (sezary syndrome).
Background
Cutaneous T-cell lymphoma (CTCL) is a sub-class of non-Hodgkin lymphoma (non-Hodgkin lymphoma). Unlike most non-hodgkin lymphomas (which are generally B-cell associated), CTCL is caused by mutations in T cells. Cancerous T cells in the body initially migrate to the skin, causing various lesions to appear. These lesions change shape as the disease progresses, typically beginning with symptoms that appear as a rash (which may be extremely itchy), and eventually forming spots and tumors before spreading to other parts of the body.
CTCL can be subdivided into mycosis fungoides and sezary syndrome. Sezary syndrome is a rare, aggressive CTCL subtype that is distinct from, but closely related to, Mycosis Fungoides (MF) forms. Sezary syndrome is characterized by exfoliative erythroderma and a large number of circulating malignant T cells (sezary cells). Sezary syndrome may also involve lymph nodes and internal organs in some patients.
Mycosis fungoides, also known as Alibert-Bazin syndrome (Alibert-Bazin syndrome) or mycosis fungoides (granuloma fungoides), is the most common form of cutaneous T-cell lymphoma. It generally affects the skin, but may develop internally over time. Symptoms include rashes, tumors, skin lesions, and itchy skin.
Although the etiology remains unclear, most cases are not hereditary. Most cases occur in people over the age of 20 years and are more common in men than women.
Mycosis fungoides can be treated in a variety of ways. Common treatments include simple sunlight, ultraviolet light, topical steroids, topical and systemic chemotherapy, topical superficial radiotherapy, the histone deacetylase inhibitor vorinostat (vorinostat), total skin electron beam radiation (total skin electron beam radiation), photodegradation (photopheresis), and systemic therapy (e.g., interferons, retinoids) or biologic therapy. The treatments are usually combined.
If the treatment is successful, the disease can enter a remission phase and the remission phase can continue indefinitely. Treatment can stop disease progression, and this is referred to as stable disease. Stable disease can also persist indefinitely, but this is a less satisfactory situation.
Disadvantageously, even with treatment, the disease may progress to involve nodules, blood and internal organs, or to convert to higher grade lymphomas. The disease is not curable, but many patients experience long-term disease control. Quality of life and the time period to maximize remission or stabilize the disease, while minimizing therapy and toxicity, are two major goals in clinical care.
In 2010, the U.S. food and drug administration awarded an orphan drug qualification (orphan drug designation) to the naloxone lotion, a topical opioid receptor competitive antagonist for the treatment of itch in cutaneous T-cell lymphomas.
There remains a need for alternative treatments for cutaneous T-cell lymphomas, such as mycosis fungoides and sezary syndrome. The inventors have data indicating that an antagonist antibody or binding fragment thereof specific for the IL-13 receptor would be a useful therapy.
Disclosure of Invention
The present disclosure will be summarized in the following paragraphs. Thus, there is provided:
1. a method of treating cutaneous T cell lymphoma comprising administering to a patient in need thereof a therapeutically effective amount of an antagonist antibody or binding fragment thereof specific for an IL-13 receptor.
2. The method according to paragraph 1, wherein the antibody or binding fragment thereof is specific for IL-13R α 1(IL-13R alpha1) or anti-IL-13R α 2(IL-13R alpha 2).
3. The method according to paragraph 2, wherein the antibody or binding fragment thereof is an anti-IL-13R α 1 antibody.
4. A method according to paragraph 2 or 3, wherein the antibody or binding fragment thereof binds to the epitope FFYQ.
5. The method according to any of paragraphs 1 to 4, wherein the antibody or binding fragment thereof has a heavy chain variable domain comprising CDR H1, CDR H2 and CDR H3 having the sequences shown in SEQ ID NOs 1,2 and 3 or 4 (or any of sequences 12 to 38), respectively, or a variable domain wherein one, two or three amino acids in the CDRs are added, substituted or deleted.
6. The method according to any of paragraphs 1 to 5, wherein the antibody or binding fragment thereof has a light chain variable domain comprising CDR L1, CDR L2 and CDR L3 having the sequences set forth in SEQ ID NOs 5,6 and 7 (or any of sequences 39 to 52), respectively, or a variable domain wherein one, two or three amino acids in the CDRs are substituted with additions or deletions.
7. A method according to any of paragraphs 1 to 6, wherein the antibody or binding fragment thereof has a light chain variable domain comprising the sequence set forth in SEQ ID NO 9 or a sequence at least 95% identical thereto (which remains specific for an IL-13 receptor, specifically IL-13R α 1).
8. A method according to any of paragraphs 1 to 6, wherein the antibody or binding fragment thereof has a light chain variable domain comprising the sequence of SEQ ID NO 9.
9. A method according to any of paragraphs 1 to 8, wherein the antibody or binding fragment thereof has a heavy chain variable domain comprising the sequence set forth in SEQ ID NO:8 or a sequence at least 95% percent identical thereto (which remains specific for an IL-13 receptor, specifically IL-13R α 1).
10. A method according to any of paragraphs 1 to 9, wherein the antibody or binding fragment thereof has a heavy chain variable domain comprising the sequence set forth in SEQ ID NO 8.
11. A method according to any of paragraphs 1 to 10, wherein the antibody or binding fragment thereof is human or humanized.
12. A method according to any of paragraphs 1 to 11, wherein the antibody-binding fragment is selected from the group consisting of: fv, dsFv, scFv, Fab 'or F (ab') 2And (3) fragment.
13. A method according to any one of paragraphs 1 to 11, wherein the antibody is a full length antibody.
14. A method according to paragraph 13, wherein the antibody heavy chain has the sequence shown in SEQ ID NO. 10 or a sequence at least 95% identical thereto, in particular SEQ ID NO. 10.
15. A method according to paragraph 13 or 14, wherein the antibody light chain has the sequence shown in SEQ ID NO. 11 or a sequence at least 95% identical thereto, in particular SEQ ID NO. 11.
16. A method according to any of paragraphs 1 to 15, wherein the antibody or binding fragment thereof inhibits IL-13 signaling through the IL-13 receptor complex.
17. The method according to any of paragraphs 1 to 16, wherein the CTCL is refractory to first line therapy, for example.
18. The method according to any one of paragraphs 1 to 17, wherein the lymphoma is mycosis fungoides.
19. The method according to any one of paragraphs 1 to 18, wherein the lymphoma is sezary syndrome.
20. The method according to any of paragraphs 1 to 19, wherein the antibody or binding fragment thereof is administered in a pharmaceutical formulation, e.g. a parenteral formulation.
21. A method according to any of paragraphs 1 to 20, wherein the anti-IL-13 receptor antibody is administered at a dose in the range of 1ng to 1000 μ g.
22. A method according to any of paragraphs 1 to 21, wherein the antibody or antibody binding fragment is used as a monotherapy.
23. A method according to any of paragraphs 1 to 21, wherein the antibody or binding fragment is used as part of a combination therapy comprising another therapeutic agent.
24. The method according to paragraph 23, wherein the other therapeutic agent is an anti-cancer agent, e.g. a chemotherapeutic agent or a combination of chemotherapeutic agents, such as selected from the group comprising: platins (e.g., cisplatin or oxaliplatin), gemcitabine (gemcitabine), capecitabine (capecitabine), 5-FU, FOLFOX, FOLFIRI, flofirnox, and combinations of two or more thereof.
25. A method according to paragraph 23 or 24, wherein the therapeutic agent is an immunomodulatory agent, such as a cytokine, for example selected from the group comprising: IL-13, IL-12 and IL-2.
26. The method according to any one of paragraphs 1 to 25, wherein the antibody is bound to a payload.
27. A method according to paragraph 26, wherein the payload is a toxin or a drug.
28. Antagonist antibodies or binding fragments thereof specific for the IL-13 receptor are useful for treating cutaneous T cell lymphoma.
29. An antagonist antibody or binding fragment for use according to paragraph 28, wherein the antibody or binding fragment thereof is specific for IL-13R α 1(IL-13R alpha1) or anti-IL-13R α 2(IL-13R alpha 2).
30. An antagonist antibody or binding fragment for use according to paragraph 29, wherein the antibody is an anti-IL-13R α 1 antibody.
31. An antagonist antibody or binding fragment for use according to paragraph 29 or 30, wherein the antibody binds epitope FFYQ.
32. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 31, wherein the antibody has a heavy chain variable domain comprising CDR H1, CDR H2 and CDR H3 having the sequences shown in SEQ ID NOs 1,2,3 or 4 (or any one of sequences 12 to 38), respectively, or a variable domain in which one, two or three amino acids in the CDRs are independently substituted or deleted by addition.
33. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 32, wherein the antibody has a light chain variable domain comprising CDR L1, CDR L2 and CDR L3 having the sequences shown in SEQ ID NOs 5,6 and 7 (or any one of sequences 39 to 52), respectively, or a variable domain in which one, two or three amino acids in the CDRs are independently substituted or deleted by addition.
34. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 33, wherein the antibody has a light chain variable domain comprising the sequence set forth in SEQ ID NO. 9 or a sequence at least 95% percent identical thereto (which remains specific for an IL-13 receptor, specifically IL-13R α 1).
35. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 34, wherein the antibody has a light chain variable domain comprising the sequence set forth in SEQ ID No. 9.
36. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 35, wherein the antibody has a heavy chain variable domain comprising the sequence set forth in SEQ ID NO:8 or a sequence at least 95% percent identical thereto (which remains specific for an IL-13 receptor, specifically IL-13R α 1).
37. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 36, wherein the antibody has a heavy chain variable domain comprising the sequence shown in SEQ ID No. 8.
38. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 37, wherein the antibody is human or humanized.
39. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 38, wherein the antibody binding fragment is selected from the group consisting of Fv, dsFv, scFv, Fab 'or F (ab') 2A group of fragments.
40. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 38, wherein the antibody is a full length antibody.
41. The antagonist antibody or binding fragment for use according to paragraph 40, wherein the antibody heavy chain has the sequence shown in SEQ ID NO. 10 or a sequence at least 95% identical thereto, as set forth in SEQ ID NO. 10.
42. An antagonist antibody or binding fragment for use according to paragraph 40 or 41, wherein the antibody light chain has the sequence shown in SEQ ID NO. 11 or a sequence at least 95% identical thereto, as set forth in SEQ ID NO. 11.
43. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 42, wherein the antibody or binding fragment thereof inhibits IL-13 signaling through the IL-13 receptor complex.
44. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 43, wherein the CTCL is refractory to, for example, first line therapy.
45. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 44, wherein the lymphoma is mycosis fungoides.
46. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 45, wherein the lymphoma is sezary syndrome.
47. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 46, wherein the antibody or binding fragment is administered in a pharmaceutical formulation, for example a parenteral formulation.
48. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 47, wherein the anti-IL-13 receptor antibody or binding fragment is administered at a dose in the range of 1ng to 1000 μ g.
49. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 48, wherein the antibody or antibody binding fragment is for use as a monotherapy.
50. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 48, wherein the antibody or binding fragment is for use as part of a combination therapy comprising another therapeutic agent.
51. An antagonist antibody or binding fragment for use according to paragraph 50, wherein the other therapeutic agent is an anti-cancer agent, e.g. a chemotherapeutic agent or a combination of chemotherapeutic agents, such as selected from the group comprising: platinum (such as cisplatin or oxaliplatin), gemcitabine, capecitabine, 5-FU, FOLFOX, FOLFIRI, flofirnox, and combinations of two or more thereof.
52. An antagonist antibody or binding fragment for use according to paragraph 50 or 51, wherein the therapeutic agent is an immunomodulatory agent, such as a cytokine, e.g. selected from the group comprising: IL-13, IL-12 and IL-2.
53. An antagonist antibody or binding fragment for use according to any one of paragraphs 28 to 52, wherein the antibody or binding fragment is bound to a payload.
54. An antagonist antibody or binding fragment for use according to paragraph 53, wherein the payload is a toxin or a drug.
55. Use of an antagonist antibody or binding fragment thereof specific for an IL-13 receptor for the manufacture of a medicament for the treatment of cutaneous T cell lymphoma.
56. Use of an antagonist antibody or binding fragment according to paragraph 55, wherein the antibody or binding fragment thereof is specific for IL-13R α 1(IL-13R alpha1) or anti-IL-13R α 2(IL-13R alpha 2).
57. The use of an antagonist antibody or binding fragment according to paragraph 56, wherein the antibody is an anti-IL-13R α 1 antibody.
58. Use of an antagonist antibody or binding fragment according to paragraphs 56 or 57, wherein the antibody binds to the epitope FFYQ.
59. The use of an antagonist antibody or a binding fragment according to any one of paragraphs 55 to 58, wherein the antibody or binding fragment has a heavy chain variable domain comprising CDR H1, CDR H2 and CDR H3 having the sequences shown in SEQ ID NOs: 1,2 and 3 or 4 (or any one of sequences 12 to 38), respectively, or a variable domain in which one, two or three amino acids of the CDRs are independently added, deleted or substituted.
60. The use of an antagonist antibody or a binding fragment according to any one of paragraphs 55 to 59, wherein the antibody has a heavy chain variable domain comprising CDR L1, CDR L2 and CDR L3 having the sequences shown in SEQ ID NOs: 5,6 and 7 (or any one of sequences 39 to 52), respectively, or a sequence in which one, two or three amino acids in the CDRs have been independently added, deleted or substituted.
61. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 60, wherein the antibody has a light chain variable domain comprising the sequence set forth in SEQ ID NO. 9 or a sequence at least 95% percent identical thereto (which remains specific for an IL-13 receptor, specifically IL-13R α 1).
62. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 61, wherein the antibody has a light chain variable domain comprising the sequence shown in SEQ ID NO 9.
63. The use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 62, wherein the antibody has a heavy chain variable domain comprising the sequence set forth in SEQ ID NO. 8 or a sequence at least 95% percent identical thereto (which remains specific for an IL-13 receptor, specifically IL-13R α 1).
64. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 63, wherein the antibody has a heavy chain variable domain comprising the sequence shown in SEQ ID NO. 8.
65. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 64, wherein the antibody is human or humanized.
66. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 65, wherein the antibody binding fragment is selected from the group consisting of Fv, dsFv, scFv, Fab 'or F (ab') 2A group of fragments.
67. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 65, wherein the antibody is a full length antibody.
68. The use of an antagonist antibody or binding fragment according to paragraph 67, wherein the antibody heavy chain has the sequence shown in SEQ ID NO. 10 or a sequence at least 95% identical thereto, as set forth in SEQ ID NO. 10.
69. Use of an antagonist antibody or binding fragment according to paragraphs 67 or 68, wherein the antibody light chain has the sequence shown in SEQ ID NO. 11 or a sequence at least 95% identical thereto, as set forth in SEQ ID NO. 11.
70. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 69, wherein the antibody or binding fragment thereof inhibits IL-13 signaling through the IL-13 receptor complex.
71. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 70, wherein the CTCL is refractory, for example, to first line therapy.
72. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 71, wherein the lymphoma is mycosis fungoides.
73. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 72, wherein the lymphoma is sezary syndrome.
74. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 73, wherein the antibody or binding fragment thereof is administered in a pharmaceutical formulation, e.g. a parenteral formulation.
75. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 74, wherein the anti-IL-13 receptor antibody or binding fragment thereof is administered at a dose in the range of 1ng to 1000 μ g.
76. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 75, wherein the antibody or antibody binding fragment is for use as a monotherapy.
77. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 76, wherein the antibody or binding fragment is used as part of a combination therapy comprising another therapeutic agent.
78. The use of an antagonist antibody or binding fragment according to paragraph 77, wherein the other therapeutic agent is an anti-cancer agent, e.g., a chemotherapeutic agent or a combination of chemotherapeutic agents, such as selected from the group comprising: platinum (such as cisplatin or oxaliplatin), gemcitabine, capecitabine, 5-FU, FOLFOX, FOLFIRI, flofirnox, and combinations of two or more thereof.
79. Use of an antagonist antibody or binding fragment according to paragraphs 77 or 78, wherein the therapeutic agent is an immunomodulatory agent, such as a cytokine, for example selected from the group comprising: IL-13, IL-12 and IL-2.
80. Use of an antagonist antibody or binding fragment according to any one of paragraphs 55 to 79, wherein the antibody or binding fragment is bound to a payload.
81. The use of an antagonist antibody or binding fragment according to paragraph 80, wherein the payload is a toxin or a drug.
In one embodiment, the IL13-R1 α 1 antibody or binding fragment employed in the present disclosure includes CDRH3 independently selected from the sequences including SEQ ID NOs 13 to 38.
In one embodiment, the anti-IL 13R antibody or binding fragment employed in the present disclosure includes a VH CDR1 comprising the amino acid sequence set forth in SEQ ID No. 1, a VHCDR2 comprising the amino acid sequence set forth in SEQ ID No. 2, and a VH CDR3 comprising the amino acid sequence set forth in SEQ ID No. 3 or 12.
In one embodiment, the anti-IL 13R antibodies or binding fragments employed in the present disclosure include CDRH1 comprising the amino acid sequence set forth in SEQ ID No. 1, CDRH2 comprising the amino acid sequence set forth in SEQ ID No. 2, and CDRH3 comprising the amino acid sequence set forth in SEQ ID No. 4, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, or 38.
In one embodiment, the anti-IL 13R antibody or binding fragment employed in the present disclosure includes CDRH1 comprising the amino acid sequence set forth in SEQ ID No. 1, CDRH2 comprising the amino acid sequence set forth in SEQ ID No. 2, and CDRH3 comprising the amino acid sequence set forth in SEQ ID No. 4.
In one embodiment, CDRL1 is an amino acid sequence comprising SEQ ID NO 5.
In one embodiment, CDRL2 is an amino acid sequence comprising SEQ ID NO 6.
In one embodiment, CDL3 comprises SEQ ID NO 39.
In one embodiment, the IL-13R α 1 antibody employed in the formulations of the present disclosure includes a CDRL3 independently selected from the sequences including SEQ ID NOs 40 to 52:
in one embodiment, an anti-IL-13R α antibody or binding fragment employed in the present disclosure includes CDRL1 comprising the amino acid sequence of SEQ ID No. 5, CDRL2 comprising the amino acid sequence of SEQ ID No. 6, and CDRL3 comprising the amino acid sequence set forth in SEQ ID No. 39.
In one embodiment, an anti-IL-13R α antibody of the disclosure includes a VL CDRL1 including amino acid sequence SEQ ID No. 5, a CDRL2 including amino acid sequence SEQ ID No. 6, and a CDRL3 including amino acid sequence set forth in SEQ ID nos. 7, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, or 52.
In one embodiment, an anti-IL-13R α antibody of the present disclosure includes a CDRL1 including the amino acid sequence of SEQ ID No. 31, a CDRL2 including the amino acid sequence of SEQ ID No. 6, and a CDRL3 including the amino acid sequence set forth in SEQ ID No. 7.
In one embodiment, the anti-IL 13R antibodies of the present disclosure include a CDRH1 comprising the amino acid sequence set forth in SEQ ID No. 1, a CDRH2 comprising the amino acid sequence set forth in SEQ ID No. 2, and a CDRH3 comprising the amino acid sequence set forth in SEQ ID No. 3 or 12, a CDRL1 comprising the amino acid sequence SEQ ID No. 5, a CDRL2 comprising the amino acid sequence set forth in SEQ ID No. 6, and a CDRL3 comprising the amino acid sequence set forth in SEQ ID No. 39.
In one embodiment, an anti-IL 13R antibody of the present disclosure includes CDRH1 including the amino acid sequence set forth in SEQ ID No. 1, CDRH2 including the amino acid sequence set forth in SEQ ID No. 2, and CDRH3 including the amino acid sequence set forth in SEQ ID No. 3, 4 or 12, CDRL1 including the amino acid sequence SEQ ID No. 5, CDRL2 including the amino acid sequence SEQ ID No. 6, and CDRL3 including the amino acid sequence set forth in SEQ ID No. 7, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51 or 52.
In one embodiment, the anti-IL 13R antibodies of the present disclosure include a CDRH1 comprising the amino acid sequence set forth in SEQ ID No. 1, a CDRH2 comprising the amino acid sequence set forth in SEQ ID No. 2, and a CDRH3 comprising the amino acid sequence set forth in SEQ ID No. 3, 4 or 12, a CDRL1 comprising the amino acid sequence SEQ ID No. 5, a CDRL2 comprising the amino acid sequence set forth in SEQ ID No. 6, and a CDRL3 comprising the amino acid sequence set forth in SEQ ID No. 7.
In one embodiment, an anti-IL 13R antibody of the present disclosure includes CDRH1 comprising the amino acid sequence set forth in SEQ ID No. 1, CDRH2 comprising the amino acid sequence set forth in SEQ ID No. 2, and CDRH3 comprising the amino acid sequence set forth in SEQ ID No. 4, CDRL1 comprising the amino acid sequence SEQ ID No. 5, CDRL2 comprising the amino acid sequence set forth in SEQ ID No. 6, and CDRL3 comprising the amino acid sequence set forth in SEQ ID No. 7.
In one embodiment, the VH regions are independently selected from the group consisting of sequences comprising SEQ ID NO 8, 53, 54 or 55.
In one embodiment, the VL is independently selected from the group consisting of SEQ ID NO 56, 57 or 58.
In one embodiment, the VH sequence is SEQ ID NO:53 (or a sequence at least 95% identical thereto) and the VL sequence is SEQ ID NO:56, SEQ ID NO:9 or 57, SEQ ID NO:58, or SEQ ID NO:55 (or a sequence at least 95% identical thereto).
In one embodiment, the VH sequence is SEQ ID NO:54 (or a sequence at least 95% identical thereto) and the VL sequence is SEQ ID NO: 9; 56, 57 or 58 (or a sequence at least 95% identical to any one thereof).
In one embodiment, the VH sequence is SEQ ID NO:55 (or a sequence at least 95% identical thereto) and the VL sequence is SEQ ID NO: 9; 56, 57 or 58 (or a sequence at least 95% identical to any one thereof).
In one embodiment, the VH sequence is SEQ ID NO:8 (or a sequence at least 95% identical thereto) and the VL sequence is SEQ ID NO:9, SEQ ID NO:56, SEQ ID NO:57, or SEQ ID NO:58 (or a sequence at least 95% identical thereto).
In one embodiment, the VL sequence is SEQ ID NO:56 (or a sequence at least 95% identical thereto) and the VH sequence is SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, or SEQ ID NO: 8. (or a sequence at least 95% identical to any one thereof)
In one embodiment, the VL sequence is SEQ ID NO:9 or 57 (or a sequence at least 95% identical thereto) and the VH sequence is SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, or SEQ ID NO:8 (or a sequence at least 95% identical to any one thereof).
In one embodiment, the VL sequence is SEQ ID NO:58 (or a sequence at least 95% identical thereto) and the VH sequence is SEQ ID NO:53, SEQ ID NO:54, SEQ ID NO:55, or SEQ ID NO:8 (or a sequence at least 95% identical to any one thereof).
In one embodiment, the VH sequence is SEQ ID NO:8 (or a sequence at least 95% identical thereto) and the VL sequence is SEQ ID NO:9 or 57 ((or a sequence at least 95% identical thereto).
Variable regions as used herein refers to regions in an antibody chain that include CDRs and appropriate frameworks.
In one embodiment, the heavy chain comprises a sequence independently selected from the group consisting of seq id no:10, 59, 60, 61, 62, 63, 64 or a sequence at least 95% identical to any one thereof.
In one embodiment, the light chains are independently selected from SEQ ID NO 11, SEQ ID NO 65, SEQ ID NO 66, or a sequence at least 95% identical to any one thereof.
In one embodiment, the heavy chains are independently selected from SEQ ID NOs 10, 59, 60, 61, 62, 63, and 64 (or sequences at least 95% identical thereto), and the light chains are independently selected from SEQ ID NOs 11, 65, and 66 (or sequences at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:10 (or a sequence at least 95% identical thereto) and the light chain is independently selected from SEQ ID NOS: 11, 65, and 66 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:59 (or a sequence at least 95% identical thereto) and the light chain is independently selected from SEQ ID NOS: 11, 65, and 66 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:60 (or a sequence at least 95% identical thereto) and the light chain is independently selected from SEQ ID NOS: 11, 65, and 66 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO 61 (or a sequence at least 95% identical thereto) and the light chain is independently selected from SEQ ID NOS 11, 65, and 66 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:62 (or a sequence at least 95% identical thereto) and the light chain is independently selected from SEQ ID NOS: 11, 65, and 66 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:63 (or a sequence at least 95% identical thereto) and the light chain is independently selected from SEQ ID NOS: 11, 65, and 66 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:64 (or a sequence at least 95% identical thereto) and the light chain is independently selected from SEQ ID NOS: 11, 65, and 66 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:62 or 64 (or a sequence at least 95% identical to either thereof) and the light chain has the sequence set forth in SEQ ID NO:11 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:62 (or a sequence at least 95% identical to any one thereof) and the light chain has the sequence set forth in SEQ ID NO:11 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:64 (or a sequence at least 95% identical to any one thereof) and the light chain has the sequence set forth in SEQ ID NO:11 (or a sequence at least 95% identical thereto).
In one embodiment, the heavy chain is SEQ ID NO:10 (or a sequence at least 95% identical to any one thereof) and the light chain has the sequence set forth in SEQ ID NO:11 (or a sequence at least 95% identical thereto).
Drawings
Figure 1 anti-tumor activity of asan 004 in samples from patients with primary CTCL.
FIG. 2 shows expression of IL-13R α 1 as determined by flow cytometry in Hut-78 cells
FIG. 3 shows ASLAN004 antitumor Activity in HUT-78 cells
Sequence of
Sequences 1,2, 4 to 11 and 13 to 66 are contained in the sequence listing only
SEQ ID NO:3 CDRH3
Met Pro Asn Trp Gly Ser Xaa Asp Xbb
Xaa is: phe or Leu
Xbb is: his, Tyr, Thr or Ser
SEQ ID NO:12 CDRH3
X 1Pro Asn Trp Gly X 6X 7Asp X 9
X1 represents Phe, Met, Gln, Leu or Val
X6 represents Ser or Ala
X7 represents Phe, Leu, Ala or Met
X9 represents Tyr, Gln, Lys, Arg, Trp, His, Ala, Thr, Ser, Asn or Gly
SEQ ID NO:39 CDRL3 Gln X 2X 3X 4X 5
X2 represents Gln, Arg, Met, Ser, Thr or Val.
X3 represents Tyr or Val.
X4 represents Glu, Ala, Gly or Ser.
X5 represents Thr, Ala or Ser.
Detailed Description
Cutaneous T cell lymphoma as used herein refers to the rare type of non-hodgkin's lymphoma that affects the skin. It is caused by leukocytes growing in an uncontrolled manner, called T-cell lymphocytes, for example causing raised, rash-like or itching skin patches, bumps on the skin and/or swollen lymph nodes. As mentioned above, it can be classified into mycosis fungoides, sezary syndrome, CD30+ cutaneous T-cell lymphoma and extranodal NK/T-cell lymphoma (nasal type).
Sezary syndrome is a rare and aggressive type of cutaneous T-cell lymphoma that is closely associated with mycosis fungoides. As mentioned above, patients have erythroderma exfoliativa and a large number of circulating malignant T cells (sezary cells). The condition may also involve lymph nodes and internal organs.
Sezary syndrome may develop from mycosis fungoides, or the complete symptoms may develop de novo.
Mycosis fungoides, also known as acalypha-barr syndrome or mycosis fungoides, is the most common form of cutaneous T-cell lymphoma. It generally affects the skin, but may develop internally over time. Symptoms include rashes, tumors, skin lesions, and itchy skin.
Paget's disease reticulocytosis (also known as "acromatic mycosis" (acromatic mycosis), "localized epidermophilic reticulosis" (palmoplantar mycosis), "unilesion mycosis" (unifocal mycosis) and "wonto-mycosis" (Woringer-Kolopp disease)) can be considered a form of mycosis fungoides.
The disease is abnormal expression of CD 4T cells. These T cells are skin-associated, meaning that they are biochemically and biologically most relevant to the skin in a dynamic manner. Diagnosis is sometimes difficult because the early stages of the disease often resemble eczema or even psoriasis. Diagnosis is generally achieved by skin biopsy. Several biopsies are recommended to make the diagnosis more definitive. The diagnosis is made by a combination of clinical pictures and examinations and confirmed by biopsy.
Pre-fungal infection (premycotic) is a mycosis stage in which a patient has a red, scaly, itchy skin area on an area of the body that is not normally exposed to sunlight. This early stage mycosis fungoides is difficult to diagnose. The pre-fungal infection phase may last from months to decades.
Secondary cutaneous CD30+ large cell lymphoma is a skin condition that may arise in the case of mycosis fungoides, and in patients with lymphomatoid papulosis (lymphomatoid papulosis).
For staging mycosis fungoides, various tests can be used to assess nodules, blood and internal organs, but most patients presenting with disease are clearly limited to the skin, in the form of plaques (flat spots) and plaques (slightly raised or 'wrinkled' spots).
Extranodal NK/T cell lymphoma of the nasal type, which is known in the REAL classification as angiocentric lymphoma (angiocentric lymphoma), and also known as nasal type NK lymphoma, NK/T cell lymphoma, and pleomorphic/malignant midline reticuloid cell proliferative disorder (polymorphic/malignant midline lymphoma), is a skin condition reported in korea as the most common form of cutaneous lymphoma following mycosis fungoides.
Staging of mycosis fungoides and sezary syndrome
Mycosis Fungoides (MF) and Sezary Syndrome (SS) are staged based on 4 factors:
t describes how much skin is affected by lymphoma (tumor).
N describes the extent of lymphoma in the lymph nodes.
M is related to the spread of lymphoma to other organs (cancer metastasis).
B is about lymphoma cells in blood.
Class T
T1: the skin lesion may be a small plaque (flat lesion), a pimple (small bump) and/or a spot (convex or concave, flat lesion), but the lesion covers less than 10% of the skin surface.
T2: the plaques, papules, and/or plaques cover 10% or more of the skin surface.
T3: at least one of the skin lesions is a tumor that is at least 1 centimeter (cm) in diameter (1cm slightly less than 1/2 inches).
T4: skin lesions have spread, grown larger, and grow together to cover at least 80% of the skin surface.
Class N
N0: the lymph nodes were not enlarged and no lymph node biopsy was required.
N1: the lymph nodes are swollen, but under the microscope, the morphology of the cells appears normal or near normal.
N2: lymph nodes swell and the morphology of the cells appears more abnormal under the microscope.
N3: lymph nodes swell and the morphology of the cells appears microscopically extremely abnormal.
NX: the lymph nodes are enlarged but have not been excised (biopsy) for observation under a microscope.
Class M
M0: lymphoma cells have not spread to the skin or outside lymph nodes.
M1: lymphoma cells have spread to other organs or tissues, such as the liver or spleen.
Class B
B0: less than 5% of the lymphocytes in the blood are sezary (lymphoma) cells.
B1: the number of sertraline cells in the blood was smaller (greater than in B0 but less than in B2).
B2: the number of sertraline cells in the blood is high.
Grouping of phases
Once the values of T, N, M and B are known, they are combined to determine the overall stage of lymphoma. This process is called phase grouping.
And stage IA: t1, N0, M0, B0 or B1.
Skin lesions were present but no tumors. Skin lesions cover less than 10% of the skin surface (T1), lymph nodes are not swollen (N0), lymphoma cells do not spread to other organs or tissues (M0), and the number of sezary cells in the blood is low (B0 or B1).
And (3) stage IB: t2, N0, M0, B0 or B1.
Skin lesions were present but no tumors. Skin lesions cover at least 10% of the skin surface (T2), lymph nodes are not swollen (N0), lymphoma cells do not spread to other organs or tissues (M0), and the number of sezary cells in the blood is low (B0 or B1).
Stage IIA: t1 or T2, N1 or N2, M0, B0 or B1.
Skin lesions were present but no tumors. Skin lesions may cover up to 80% of the skin surface (T1 or T2). Lymph nodes swollen, but under the microscope, the morphology of the cells did not appear particularly abnormal (N1 or N2). Lymphoma cells did not spread to other organs or tissues (M0), and the number of sertraline cells in the blood was small (B0 or B1).
Stage IIB: t3, N0 to N2, M0, B0 or B1.
At least one of the skin lesions is a tumor having a diameter of 1cm or greater (T3). Lymph nodes were normal (N0) or swollen, but under the microscope, the morphology of the cells did not appear particularly abnormal (N1 or N2). Lymphoma cells did not spread to other organs or tissues (M0), and the number of sertraline cells in the blood was small (B0 or B1).
Stage IIIA: t4, N0 to N2, M0, B0.
The skin lesion covers at least 80% of the skin surface (T4). Lymph nodes were normal (N0) or swollen, but under the microscope, the morphology of the cells did not appear particularly abnormal (N1 or N2). Lymphoma cells did not spread to other organs or tissues (M0), and there were few (or no) sertraline cells present in the blood (B0).
Stage IIIB: t4, N0 to N2, M0, B1
The skin lesion covers at least 80% of the skin surface (T4). Lymph nodes were normal (N0) or swollen, but under the microscope, the morphology of the cells did not appear particularly abnormal (N1 or N2). Lymphoma cells did not spread to other organs or tissues (M0) and the number of sertraline cells in the blood was low (B1).
Stage IVA 1: any of T, N0 to N2, M0, B2.
The skin lesion may cover any amount of the skin surface (any T). Lymph nodes were normal (N0) or swollen, but under the microscope, the morphology of the cells did not appear particularly abnormal (N1 or N2). Lymphoma cells did not spread to other organs or tissues (M0) and the number of sertraline cells in the blood was high (B2).
Stage IVA 2: any T, N3, M0, any B.
The skin lesion may cover any amount of the skin surface (any T). Some lymph nodes were swollen and the morphology of the cells appeared extremely abnormal under the microscope (N3). Lymphoma cells do not spread to other organs or tissues (M0). The sertraline cell may or may not be in the blood (any B).
And (3) IVB stage: any T, any N, M1, any B
The skin lesion may cover any amount of the skin surface (any T). The lymph node may be normal or abnormal (any N), and the tuckley cells may or may not be in the blood (any B). Lymphoma cells have spread to other organs or tissues, such as the liver or spleen (M1).
Staging of other cutaneous lymphomas
The staging system for cutaneous lymphoma types other than mycosis fungoides and sezary syndrome remains fairly new and physicians still attempt to determine their degree of applicability. The system is based on 3 factors:
t describes how much skin is affected by lymphoma (tumor).
N describes the extent of lymphoma in the lymph nodes.
M is related to the spread of lymphoma to other organs (cancer metastasis).
For these lymphomas, only class T was used in the diagnosis. If sites other than the skin (e.g., lymph nodes) are involved in diagnosis, these lymphomas are no longer considered cutaneous lymphomas and are staged like conventional non-hodgkin lymphomas. Classes N and M are used only in cases where lymphoma progresses during treatment (continues to grow) or recurs after treatment.
Class T
T1: there is only a single skin lesion.
T1 a: the skin lesion is less than 5cm (about 2 inches) in diameter.
T1 b: the diameter of the skin lesion is larger than 5 cm.
T2: there are 2 or more lesions on the skin. These may be in a single body zone or in 2 body zones adjacent to each other.
T2 a: all skin lesions can be placed within a circle of 15cm (about 6 inches) in diameter.
T2 b: the circle required to encompass all skin lesions is greater than 15cm in diameter but less than 30cm (about 1 foot) in diameter.
T2 c: the diameter of the circle required to enclose all skin lesions is greater than 30 cm.
T3: skin lesions are present in body regions that are not adjacent to each other, or at least 3 different body regions.
T3 a: there are many lesions that involve 2 body regions that are not adjacent to each other.
T3 b: there are many lesions that involve 3 or more body regions.
Class N
N0: no lymph node enlargement or containing lymphoma cells.
N1: there are lymphoma cells in the lymph nodes that drain (drain) areas at the skin containing the lymphoma.
N2: one of the following is true:
at least 2 groups of lymph nodes from different regions contain lymphoma cells.
There are lymphoma cells in the lymph nodes that do not drain the area at the skin containing the lymphoma.
N3: lymph nodes deep within the chest or abdomen contain lymphoma cells.
Class M
M0: there were no signs of lymphoma outside the skin or lymph nodes.
M1: lymphoma has spread to other organs or tissues.
This system does not assign an overall stage to lymphoma as does the mycosis fungoides/sezary syndrome system. Because this system is still quite new, it is not clear to what extent it can help predict the prognosis (perspective) of an individual.
As used herein, the IL-13 receptor is a type I cytokine receptor with two subunits, IL-13R α 1(Uniprot No. P78552) and IL4R α (Q9H 186). The formation of these is a dimer with IL-13. the signal is generated via activation of the JAK/Signal Transducer and Activator of Transcription (STAT) pathways.
IL-13 receptors may also stimulate IL-4 signaling.
Antagonists as used herein refers to decreasing or inhibiting biological activity or function, such as physiological antagonists, in particular blocking or reducing the effects of JAK/signal transducers and transcriptional activator pathways.
As used herein, unless the context indicates otherwise, an "antibody" encompasses substantially intact antibody molecules, as well as chimeric antibodies, humanized antibodies, human antibodies (in which at least one amino acid mutation relative to a naturally occurring human antibody), single chain antibodies (e.g., antibody heavy chain, antibody light chain), multispecific antibodies (e.g., bispecific antibodies), homodimers and heterodimers of antibody heavy and/or light chains, and may also include antigen-binding fragments and derivatives thereof.
In one embodiment, the antibody or binding fragment thereof is monoclonal.
An "antigen-binding fragment" as used herein refers to a functional fragment of an antibody that is capable of binding to its specific antigen.
Unless the context indicates otherwise, antibody-binding fragments, antigen-binding fragments, and binding fragments are used interchangeably herein.
Examples of antibody binding fragments include Fab, modified Fab, Fab ', modified Fab ', F (ab ') 2Fv, Fab-dsFv, single domain antibody (e.g., VH or VL or VHH), scFv, diabody, triabody or tetrabody, diabody, triabody, tetrabody, and epitope-binding fragment of any of the foregoing (see, e.g., holly)Gell (Holliger) and Hedelson (Hudson), 2005, Nature Biotech 23(9) 1126-1136; adal (Adair) and Lawson (Lawson), 2005, Drug Design review-Online (Drug Design Reviews-Online) 2(3), 209-217). Methods for producing and making such antibody fragments are well known in the art (see, e.g., Verma et al, 1998, Journal of Immunological Methods, 216, 165-181). Other antibody fragments for use in the present invention include Fab and Fab' fragments as described in International patent applications WO2005/003169, WO2005/003170 and WO 2005/003171.
Examples of multispecific antibodies, including full-length antibodies, include DVD-Ig, IgG-scFv, scFv-IgG, and IgG-V.
IgG-scFv as used herein is a full length antibody with scFv at the C-terminus of each heavy chain or each light chain.
scFv-IgG as used herein is a full length antibody with scFv on the N-terminus of each heavy chain or each light chain.
V-IgG as used herein is a full length antibody with a variable domain on the N-terminus of each heavy chain or each light chain.
IgG-V As used herein is a full length antibody with a variable domain at the C-terminus of each heavy chain or each light chain
DVD-Ig (also known as double V-domain IgG) is a full length antibody with 4 additional variable domains, one on the N-terminus of each heavy chain and each light chain.
In one embodiment, the antibody binding fragment is or includes a Fab or Fab' fragment.
Antibody binding fragments including Fab or Fab 'fragments include Fabdab, Fab' dab, Fabfv, Fab 'Fv, Fabdsfv, Fab-scFv, Fab' -scFv, Fab- (scFv)2, Fab '- (scFv)2, DiFab'.
Fabdab as used herein refers to a Fab fragment having a domain antibody attached (optionally via a linker) to its heavy or light chain.
Fab 'dab as used herein refers to a Fab' fragment having a domain antibody attached (optionally via a linker) to its heavy or light chain.
FabFv as used herein refers to Fab fragments with additional variable regions attached to the C-terminus of each of: CH1 for the heavy chain and CL for the light chain, see e.g. WO 2009/040562. The forms may be provided in their pegylated forms, see for example WO 2011/061492.
The Fab 'Fv as used herein is similar to the Fabfv in that the Fab part is replaced by Fab'. The form may be provided in its pegylated form.
FabdsFv as used herein refers to a FabFv in which an internal Fv disulfide bond stabilizes the attached C-terminal variable region, see, e.g., WO 2010/035012. Can be provided in its pegylated form
A Fab-scFv (also referred to as diabody) as employed herein is a Fab molecule having an scFv attached (optionally via a linker) to the C-terminus of either the light or heavy chain.
A Fab '-scFv as employed herein is a Fab' molecule having an scFv attached (optionally via a linker) to the C-terminus of either the light chain or the heavy chain.
DiFab as used herein refers to two Fab molecules linked via the C-terminus of their heavy chains.
DiFab 'as used herein refers to two Fab' molecules linked via one or more disulfide bonds in their hinge region.
DiFab and DiFab' molecules contain their chemically bound forms.
Antibodies and binding fragments according to the present disclosure in which one, two, or three amino acids in the CDRs are altered refer to modified antibodies that retain specificity for an antigen (target antigen), i.e., modified antibodies that are specific for the IL-13 receptor, specifically the IL-13R α 1 receptor.
By "one, two, three amino acids in a CDR are altered" is meant that the total of the altered amino acids in the CDRs of the variable domain is no more than three. It does not refer to up to three amino acid changes in each of the CDRs of a given variable domain.
Modified variable domains having at least 95% (e.g., 96, 97, 98, 99%) identity to the sequences specifically disclosed herein form an aspect of the present disclosure.
Having specificity for a target antigen as used herein refers to the fact that: the antibody or binding fragment binds only to its specific antigen or binds to its specific antigen with an affinity that is 2-fold, 3-fold, 4-fold, 5-fold greater than the affinity of the substance or antigen to which it does not have specificity.
In one embodiment, the binding affinity (K) of an antibody or binding fragment of the disclosure D) 1nM or less, such as 0.5nM, 0.3nM, or 0.2 nM. In one embodiment, the binding affinity is about 254 pM.
Antibodies and binding fragments may be referred to herein as "actives".
As used herein, 'pharmaceutical formulation' refers to a therapeutically effective formulation comprising an antibody or binding fragment thereof and a pharmaceutically acceptable diluent, excipient and/or carrier.
As used herein, a 'therapeutically effective amount' or 'therapeutically effective' refers to an amount that provides a therapeutic effect for a given condition and dosing regimen. This is a predetermined amount of active material calculated to produce the desired therapeutic effect in combination with the required additives and diluents, i.e., carriers or delivery vehicles. Furthermore, it is intended to mean an amount sufficient to reduce and/or prevent clinically significant deficits in the activity, function and response of the host/patient. Alternatively, the therapeutically effective amount is sufficient to cause an improvement in a clinically significant condition in the host/patient. As will be appreciated by those of skill in the art, the amount of an active agent (e.g., an antibody or binding fragment according to the present disclosure) can vary depending on its specific activity. Suitable dosages may contain a predetermined amount of the active composition calculated to produce the desired therapeutic effect in association with the diluent required. In the methods of manufacture and use of the compositions of the present invention, a therapeutically effective amount of the active ingredient is provided, for example, as a unit dose.
The therapeutically effective amount can be determined by the skilled medical practitioner or veterinarian based on the patient characteristics (e.g., age, weight, sex, condition, complications, other diseases, etc.), as is well known in the art.
In one embodiment, the patient is a human.
As used herein, the term payload includes, for example, biologically active proteins (such as enzymes), other antibodies or antibody binding fragments, synthetic or naturally occurring polymers, nucleic acids and fragments thereof (e.g., DNA, RNA, and fragments thereof), radionuclides (in particular radioiodine), radioisotopes, chelated metals, nanoparticles, and reporter groups (such as fluorescent compounds or compounds detectable by NMR or ESR spectroscopy).
In one embodiment, the payload is a toxin, such as a toxin selected from the group consisting of calicheamicin (calicheamicin), apridin (aplidine), anastrozole (anastrozole), azacytidine, bortezomib (bortezomib), bryodin-1 (bryostatin-1), busulfan, rivastigmine (combestin), carmustine (carmustine), dolastatin (dolastatin), epothilone (epothilone), staurosporine (staurosporin), maytansinoid (maytansinoid), spongistatin (spongistatin), rhizomycin (rhizoxin), halichondrosine (halichondrosin), fisetin (roridin), hemiasterlin (hemistrilin), paclitaxel (taxol), cytochalin B (cytochalasin B), gracillin D (brevicidin D), vincristine (vincristine), vincristine (vinc, Daunorubicin (daunorubicin), dihydroxyanthralin dione (dihydroanthracin dione), mitoxantrone (mitoxantrone), mithramycin (mithramycin), actinomycin D (actinomycin D), 1-dehydrotestosterone, glucocorticoids, procaine (procaine), tetracaine (tetracaine), lidocaine (lidocaine), propranolol (propranolol), and puromycin (puromycin), and analogs or homologs thereof.
In one embodiment, the payload is a drug selected from nitrogen mustard, ethylenimine derivatives, alkyl sulfonates, nitrosoureas, gemcitabine, triazines, folic acid analogs, anthracyclines (anthracyclines), taxanes (taxanes), COX-2 inhibitors, pyrimidine analogs, purine analogs, antibiotics, enzyme inhibitors, epidophyllotoxins (epidophyllotoxin), platinum coordination complexes, vinca alkaloids (vinca alkaloids), substituted ureas, methylhydrazine derivatives, adrenocortical inhibitors, hormone antagonists, endostatin (endostatin), paclitaxel (captothecin), rhodorubicin (capreomycin), berrubicin, vincristine, daunorubicin (vincristine), vincristine (vincristine), epirubicin (oxypetacin), epirubicin), mitomycin (oxypetacin), mitomycin (oxypetamycin), oxypetasone (oxypetasone), oxypetasone (oxypetasone), doxycycline, oxypetasone), doxycycline (methotrexate), doxine, doxycycline (methotrexate), doxycycline, dox
Other payloads may comprise chelating radionuclides such as 111In and 90Y, Lu177, Bismuth213, Californium252, Iridium192, and Tungsten188/Rhenium 188; or drugs such as, but not limited to, alkylphosphocholines, topoisomerase I inhibitors, taxoids, and suramin (suramin).
Related proteins, polypeptides and peptides include, but are not limited to, immunoglobulins, toxins such as abrin (abrin), ricin a (ricin a), pseudomonas exotoxin (pseudomonas exotoxin) or diphtheria toxin, proteins such as insulin, tumor necrosis factor, α -interferon, β -interferon, nerve growth factor, platelet-derived growth factor or tissue plasminogen activator, thrombotic or anti-angiogenic agents such as angiostatin or endostatin, or biological response modifiers such as lymphokines, interleukin-1 (IL-1), interleukin-2 (IL-2), granulocyte macrophage-stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), Nerve Growth Factor (NGF) or other growth factors, and immunoglobulins.
Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radionuclides, positron emitting metals (for positron emission tomography), and nonradioactive paramagnetic metal ions see generally U.S. Pat. No. 4,741,900 which can be combined with antibodies for use as diagnostic agents suitable enzymes include horseradish peroxidase, alkaline phosphatase, β -galactosidase, or acetylcholinesterase, suitable prosthetic groups include streptavidin, avidin, and biotin, suitable fluorescent materials include umbelliferone, fluorescein isothiocyanate, rhodamine (rhodamine), dichlorotriazinylamine fluorescein, dansyl chloride, and phycoerythrin, suitable luminescent materials include luminol (luminol), suitable bioluminescent materials include luciferase, fluorescein, and aequorin, and suitable radionuclides include 125I, 131I, 111In, and 99 Tc.
In another example, the payload can increase the in vivo half-life of the antibody and/or decrease the immunogenicity of the antibody and/or enhance delivery of the antibody across the epithelial barrier to the immune system. Examples of suitable effector molecules of this type comprise polymers, albumin binding proteins or albumin binding compounds, such as those described in WO 05/117984.
Where the effector molecule is a polymer, it may, in general, be a synthetic or naturally occurring polymer, for example an optionally substituted straight or branched chain polyalkylene, polyalkenylene or polyoxyalkylene polymer or a branched or unbranched polysaccharide, for example a homologous or heterologous polysaccharide.
Certain optional substituents that may be present on the above-described synthetic polymers comprise one or more hydroxyl, methyl, or methoxy groups.
Specific examples of synthetic polymers comprise optionally substituted linear or branched poly (ethylene glycol), poly (propylene glycol) poly (vinyl alcohol) or derivatives thereof, in particular optionally substituted poly (ethylene glycol) such as methoxy poly (ethylene glycol) or derivatives thereof.
Particular naturally occurring polymers include lactose, amylose, dextran, glycogen or derivatives thereof.
As used herein, "derivative" is intended to refer to a modified molecule (e.g., a polymer) that retains the essential characteristics of the original molecule, comprising a reactive derivative, e.g., a thiol-selective reactive group, such as maleimide and the like. The reactive group may be attached to the polymer directly or through a linker segment. It will be appreciated that the residues of the groups will in some cases form part of the product as a linking group between the antibody fragment and the polymer.
The size of the polymer may vary as desired, but will generally be in the average molecular weight range 500Da to 50000Da, for example 5000 to 40000Da, such as 20000 to 40000 Da. In particular, the polymer size may be selected based on the intended use of the product, for example, localization to certain tissues such as tumors, or the ability to extend the circulating half-life (for review, see Chapman, 2002, Advanced Drug Delivery Reviews, 54, 531-545).
Thus, for example, when the product is intended to leave the circulation and penetrate tissue, e.g. for the treatment of tumours, it may be advantageous to use a small molecular weight polymer, e.g. with a molecular weight of about 5000 Da. For applications where the product remains in the recycle, it may be advantageous to use higher molecular weight polymers, for example having a molecular weight in the range of 20000Da to 40000 Da.
Suitable polymers comprise polyalkylene polymers such as poly (ethylene glycol) or especially methoxy poly (ethylene glycol) or derivatives thereof, and especially have a molecular weight in the range of from about 15000Da to about 40000 Da.
In one example, an antibody for use in the invention is linked to a poly (ethylene glycol) (PEG) moiety. In one particular example, the antibody is an antibody fragment, and the PEG molecule may be linked by any available amino acid side chain or terminal amino acid functional group located in the antibody fragment, such as any free amino, imino, thiol, hydroxyl, or carboxyl group. The amino acids may be naturally occurring in antibody fragments or may be engineered into fragments using recombinant DNA methods (see, e.g., US5,219,996; US5,667,425; WO 98/25971; WO 2008/038024). In one example, the antibody molecule of the invention is a modified Fab fragment, wherein the modification is the addition of one or more amino acids at the C-terminus of its heavy chain to allow for the attachment of effector molecules. Suitably, the additional amino acids form a modified hinge region containing one or more cysteine residues that can be linked to an effector molecule. Multiple sites can be used to attach two or more PEG molecules.
Suitably, the PEG molecule is covalently linked via a thiol group of at least one cysteine residue located in the antibody fragment each polymer molecule attached to the modified antibody fragment may be covalently linked to a sulphur atom of the cysteine residue located in the fragment the covalent linkage is typically a disulphide bond or, more precisely, a sulphur-carbon bond when a thiol group is used as the point of attachment, an appropriately activated effector molecule may be used, for example a thiol selective derivative such as maleimide and cysteine derivatives the activated polymer may be used as a starting material in the preparation of the polymer modified antibody fragments as described above the activated polymer may be any polymer containing a thiol reactive group such as α -halocarboxylic acid or ester, for example iodoacetamide, an imide such as maleimide, vinylsulphone or a disulphide, which starting material may be obtained commercially (e.g. from Nektar, formerly sierwater Polymers), USA javelvet viille (hunter, hunter USA) or may be prepared from conventional chemical commercial starting materials including special PEG-20 g. branhamel-r, formerly branhamel-r.
In one embodiment, the antibody is a pegylated modified Fab fragment or di Fab, i.e., having PEG (poly (ethylene glycol)) covalently attached thereto, e.g., according to the methods disclosed in EP 0948544 or EP1090037 [ see also poly (ethylene glycol) Chemistry, biotechnology and Biomedical Applications (poly (ethylene glycol) Chemistry, biotechnology and Biomedical Applications), 1992, j.milton harris (j.milton harris) (eds.), plena Press, new york; poly (ethylene glycol) Chemistry and Biological Applications (poly (ethylene glycol) Chemistry and Biological Applications), 1997, j. milton harris and s. sellipsky (s. zalipsky) (eds.), american society of Chemistry, colombia, washington; and bioconjugate Protein Coupling technology of Biomedical science (Bioconjugation Protein Coupling technology for the Biomedical Sciences), 1998, M. Allam (M.Aslam) and A. dtex (A.Dent), Grove Publishers, New York; chapman, A.2002, advanced drug delivery review 2002,54: 531-. In one example, PEG is linked to a cysteine in the hinge region. In one example, the PEG-modified Fab fragment has a maleimide group covalently linked to a single thiol group in the modified hinge region. The lysine residue may be covalently linked to a maleimide group, and each amine group on the lysine residue may be linked to a methoxy poly (ethylene glycol) polymer having a molecular weight of about 20,000 Da. Thus, the total molecular weight of the PEG attached to the Fab fragment may be about 40,000 Da.
A particular PEG molecule comprises a N, N' -bis (methoxypoly (ethylene glycol) MW 20,000) modified 2- [3- (N-maleimido) propionamido ] ethylamide of lysine, also known as PEG2MAL40K (available from nyctam, formerly hilwatt).
Alternative sources of PEG linkers include NOFs that supply GL2-400MA2 (where m in the following structure is 5) and GL2-400MA (where m is 2) and n is about 450:
Figure BDA0002332624410000301
m is 2 or 5.
That is, each PEG is about 20,000 Da. Other alternative PEG effector molecules of the following types:
in one embodiment, antibodies pegylated (e.g., with PEG as described herein) are provided, linked through a cysteine amino acid residue at or near amino acid 226 in the chain, e.g., amino acid 226 of the heavy chain (according to consecutive numbering).
In one embodiment, the antibody or binding fragment is provided in a pharmaceutical formulation comprising one or more excipients, diluents and/or carriers. Accordingly, there is provided a pharmaceutical composition comprising an antibody or binding fragment as described above.
It will be appreciated by those skilled in The art that The antibodies of The invention, or antigen-binding fragments, derivatives or variants thereof, will generally be administered in admixture with a suitable pharmaceutical excipient diluent or carrier selected with respect to The intended route of administration and standard pharmaceutical Practice (see, for example, Remington: The Science and Practice of Pharmacy, 19 th edition, 1995, eds., Arfangso Naro (Alfonso Gennaro), Mark Press (Mack Publishing Company, Pa., USA).
For example, the antibodies of the invention or antigen binding fragments, derivatives or variants thereof may be administered orally, buccally or sublingually in the form of tablets, capsules, beads, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate, delayed or controlled release applications.
The tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, Hydroxypropylmethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
Solid compositions of a similar type may also be used as fillers in gelatin capsules. In this aspect, suitable excipients include lactose, starch, cellulose, lactose, or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the compounds of the invention may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof. Alternatively, the capsule may be filled with a liquid formulation.
The antibody of the invention, or antigen-binding fragment, derivative or variant thereof, may also be administered parenterally by intracavernosal injection, e.g., intravenously, intraarticularly, intraarterially, intraperitoneally, intrathecally, intracerebroventricularly, intrasternally, intracranially, intramuscularly or subcutaneously, or it may be administered by infusion techniques. They are best used in the form of sterile aqueous solutions, which may contain other substances, for example, enough salts or glucose to render the solution isotonic with blood. The aqueous solution should be suitably buffered (preferably to a pH of 3 to 9) if desired. Preparation of suitable parenteral formulations under sterile conditions can be readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain antioxidants, buffers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may contain suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Ready-to-use injection solutions and suspensions can be prepared from sterile powders, granules and tablets.
An exemplary method: 1) addition of excipients, such as buffers and detergents (usually Tween) to inhibit aggregation in aqueous formulations; 2) freeze-drying with suitable excipients provides bulk, stability and aesthetic benefits to the cake; 3) compounds such as trehalose are used to form glass sugars.
For oral and parenteral administration, or other routes of administration, to human patients, the daily dosage level of the antibody, or antigen-binding fragment, derivative or variant thereof, of the present invention will generally be from 1 μ g to 1000mg (i.e., about 0.015 to 15mg/kg) per adult human, administered in single or divided doses.
For example, the dosage level may be from about 0.5mg/kg to about 10mg/kg, the dosage regimen may be two or three times per week, and the administration may be, for example, intravenous or subcutaneous. In another example, the dosing regimen may range from once weekly to once monthly, either intravenously or by subcutaneous injection.
The antibodies of the invention, or antigen-binding fragments, derivatives or variants thereof, may also be administered intranasally or by inhalation and are conveniently delivered, for example, in the form of a dry powder inhaler, pump, nebulizer or atomizer, from a pressurized container by presenting an aerosol spray using a suitable propellant, such as dichlorodifluoromethane, trichlorofluoro-methane, dichlorotetrafluoro-ethane, hydrofluoroalkane (such as 1,1,1, 2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3, 3-heptafluoropropane (HFA 227EA3)), carbon dioxide or other suitable gas, in the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. If a mixture of ethanol and propellant is used as solvent, it may additionally contain a lubricant, such as sorbitan trioleate. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated containing a powder mix of an antibody or binding fragment of the invention and a suitable powder base such as lactose or starch.
The aerosol or dry powder formulation is suitably arranged such that each dose (or metered dose or 'emitted dose') contains at least 1 μ g of an antibody or antigen-binding fragment, derivative or variant thereof of the invention for delivery to a patient. It will be appreciated that the total daily dose of the aerosol will vary from patient to patient and may be administered in a single dose or, more usually, in divided doses throughout the day.
Alternatively, the antibody of the invention, or antigen-binding fragment, derivative or variant thereof, may be administered in the form of a suppository or pessary, or it may be administered topically in the form of an emulsion, solution, cream, ointment or powder. The compounds of the invention may also be administered transdermally, for example, by the use of a skin patch. It can also be administered by the ocular route.
For ophthalmic use, the antibody or antigen-binding fragment, derivative or variant thereof of the present invention may be formulated as a suspension in isotonic, pH adjusted sterile saline, or, where appropriate, as a solution in isotonic, pH adjusted sterile saline, optionally in combination with a preservative such as benzalkonium chloride. Alternatively, it may be formulated as an ointment, such as petrolatum.
For topical application to the skin, the antibodies of the invention or antigen-binding fragments, derivatives or variants thereof may be formulated as a suitable ointment, suspended or dissolved in, for example, a mixture having one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, the antibody or binding fragment may be formulated as a suitable emulsion or cream, suspended or dissolved in a mixture of, for example, one or more of the following: mineral oil, sorbitan monostearate, polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
Formulations suitable for topical administration in the mouth include buccal tablets comprising the active ingredient in a flavored base, usually sucrose and acacia or tragacanth; lozenges comprising the active ingredient in an inert base (e.g. gelatin and glycerin or sucrose and acacia); and mouthwashes comprising the active ingredient in a suitable liquid carrier.
In one embodiment, a sustained release drug delivery system, such as microspheres, is employed. Which are specifically designed for reducing the frequency of injections. An example of such a system is Nutropin Depot, which encapsulates recombinant human growth hormone (rhGH) in biodegradable microspheres that, once injected, slowly release rhGH over a sustained period of time.
Alternatively, the antibodies of the invention, or antigen binding fragments, derivatives or variants thereof, may be administered by a surgically implanted device that, for example, releases the active directly to the desired site.
Electroporation therapy (EPT) systems may also be used for the administration of antibodies or antigen-binding fragments, derivatives or variants thereof. The means for delivering a pulsed electric field to the cell increases the permeability of the cell membrane to the drug, resulting in a significant enhancement of intracellular drug delivery.
The antibody or antigen binding fragment, derivative or variant thereof may also be delivered by Electron Incorporation (EI). EI occurs when small particles on the surface of the skin, up to 30 microns in diameter, are subjected to electrical pulses consistent with or similar to those used in electroporation. In EI, these particles are driven through the stratum corneum and into the deeper layers of the skin. The particles may be loaded or coated with a drug or gene, or may simply act as "bullets" that create pores in the skin through which the drug may enter.
An alternative method of delivery of the antibody or antigen-binding fragment, derivative or variant thereof is the thermosensitive ReGel injectable method. Below body temperature, ReGel is an injectable liquid, while at body temperature it immediately forms a gel reservoir that slowly erodes and dissolves into a known safe biodegradable polymer. The active drug is delivered over time as the biopolymer dissolves.
The antibody or antigen binding fragment, derivative or variant thereof or the pharmaceutical product may also be delivered orally. One such system employs the natural process of oral intake of vitamin B12 in vivo to co-deliver proteins and polypeptides. By using the vitamin B12 uptake system, proteins or polypeptides can move through the intestinal wall. A complex is created between a vitamin B12 analog and a drug that retains both significant affinity for Intrinsic Factor (IF) in the vitamin B12 portion of the complex and significant biological activity of the "antibody" portion of the complex.
In the context of this specification, "comprising" should be interpreted as "including". Aspects of the present disclosure including certain elements are also intended to extend to alternative embodiments "consisting of or" consisting essentially of the relevant elements. The embodiments positively recited herein may be employed as the basis for disclaimer statements. All references mentioned herein are specifically incorporated by reference. Embodiments of the invention may be combined as technically appropriate. The headings herein are for segmenting documents and are not intended to be used to interpret the meaning of the disclosure provided herein.
Priority is claimed in this application. The priority document can be used as a basis for corrections, in particular sequences. The invention will now be described with reference to the following examples, which are illustrative only and should not be construed as limiting the scope of the disclosure in any way.
Examples of the invention
Example 1
Monoclonal antibody ASLAN004 (anti-IL-13R α 1 antibody) was tested on leukemia cells from patients with Sezary syndrome in vitro, the antibody concentration employed was 0.01 μ g/ml the results are shown in FIG. 1, which shows that the antibody has anti-tumor activity in the absence and also in the presence of IL-13.
Example 2
HUT-78 has a surface expression of IL-13R α 1 measurable by flow cytometry expression of IL-13R α 1 (dark grey histogram) was determined by flow cytometry in Hut-78 cells treated with/without IL-13(100 ng/ml). We used 2 different abs; anti-IL-13R α 1 from R & D (A) or from Sigma (B.) as a control group (grey histogram), we used the same type in (A) and only the secondary abs in (B).
The results show HUT-78 cells express IL13R α 1 on their cell surface and IL-13R α 1 expression does not change with or without the addition of IL-13 finally, the results indicate that the anti-IL 13R α 1 antibody from sigma (B) has a larger dynamic range relative to the antibody from R & d (a).
Example 3
Cells were preincubated with inhibitors (a1 ═ ASLAN 004; a2 ═ anti-IL-13 Ra2 Ab (boledend (Biolegend)); STAT-6 inhibitor ═ AS 1517499 (Axon)) at 37 ℃ for 1 hour, followed by addition of either medium (a) or IL-13(B) alone, by ANOVA statistics, followed by the dukay's post-hoc test (Tukey's post-hoc test) results are shown in fig. 3.
Sequence listing
<110> Asia-lion private company Limited
<120> therapeutic methods using IL-13R antibodies
<130>P000223_WO
<150>SG10201705435W
<151>2017-06-30
<160>66
<170>PatentIn version 3.5
<210>1
<211>10
<212>PRT
<213> Artificial sequence
<220>
<223>CDRH1
<400>1
Gly Tyr Ser Phe Thr Ser Tyr Trp Ile Gly
1 5 10
<210>2
<211>10
<212>PRT
<213> Artificial sequence
<220>
<223>CDRH2
<400>2
Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg
1 5 10
<210>3
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223>CDRH3
<220>
<221>MISC_FEATURE
<222>(7)..(7)
<223> Xaa is Phe or Leu
<220>
<221>MISC_FEATURE
<222>(9)..(9)
<223> Xaa is His, Tyr, Thr or Ser
<400>3
Met Pro Asn Trp Gly Ser Xaa Asp Xaa
1 5
<210>4
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223>CDRH3
<400>4
Met Pro Asn Trp Gly Ser Leu Asp His
1 5
<210>5
<211>12
<212>PRT
<213> Artificial sequence
<220>
<223>CDRL1
<400>5
Arg Ala Ser Gln Ser Ile Ser Ser Ser Tyr Leu Ala
1 5 10
<210>6
<211>7
<212>PRT
<213> Artificial sequence
<220>
<223>CDRL2
<400>6
Gly Ala Ser Ser Arg Ala Thr
1 5
<210>7
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223>CDRL3
<400>7
Gln Gln Tyr Ala Ser
1 5
<210>8
<211>118
<212>PRT
<213> Artificial sequence
<220>
<223> VH region
<400>8
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210>9
<211>104
<212>PRT
<213> Artificial sequence
<220>
<223> VL region
<400>9
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Ala Ser Phe Gly
85 90 95
Gln Gly Thr Lys Val Glu Ile Lys
100
<210>10
<211>445
<212>PRT
<213> Artificial sequence
<220>
<223> IgG4 heavy chain
<400>10
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190
Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys
210 215 220
Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu
245 250 255
Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln
260 265 270
Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys
275 280 285
Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu
290 295 300
Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys
305 310 315 320
Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys
325 330 335
Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser
340 345 350
Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys
355 360 365
Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln
370 375 380
Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln
405 410 415
Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn
420 425 430
His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
435 440 445
<210>11
<211>211
<212>PRT
<213> Artificial sequence
<220>
<223> kappa light chain
<400>11
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Ala Ser Phe Gly
85 90 95
Gln Gly Thr Lys Val Glu Ile Lys Arg Thr Val Ala Ala Pro Ser Val
100 105 110
Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu Lys Ser Gly Thr Ala Ser
115 120 125
Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala Lys Val Gln
130 135 140
Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser Gln Glu Ser Val
145 150 155 160
Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser Ser Thr Leu
165 170 175
Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr Ala Cys Glu
180 185 190
Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys Ser Phe Asn Arg
195 200 205
Gly Glu Cys
210
<210>12
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223>CDRH3
<220>
<221>MISC_FEATURE
<222>(1)..(1)
<223> Xaa represents Phe, Met, Gln, Leu or Val
<220>
<221>MISC_FEATURE
<222>(6)..(6)
<223> Xaa represents Ser or Ala
<220>
<221>MISC_FEATURE
<222>(7)..(7)
<223> Xaa represents Phe, Leu, Ala or Met
<220>
<221>MISC_FEATURE
<222>(9)..(9)
<223> Xaa represents Tyr, Gln, Lys, Arg, Trp, His, Ala, Thr, Ser, Asn or Gly
<400>12
Xaa Pro Asn Trp Gly Xaa Xaa Asp Xaa
1 5
<210>13
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>13
Phe Pro Asn Trp Gly Ala Leu Asp Gln
1 5
<210>14
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>14
Val Pro Asn Met Gly Ser Leu Asp Thr
1 5
<210>15
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>15
Phe Pro Asn Trp Gly Ser Met Asp Ala
1 5
<210>16
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>16
Phe Pro Asn Trp Gly Ser Leu Asp His
1 5
<210>17
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>17
Met Pro Asn Trp Gly Ser Phe Asp Tyr
1 5
<210>18
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>18
Met Pro Asn Trp Gly Ser Phe Asp Thr
1 5
<210>19
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>19
Met Pro Asn Trp Gly Ser Phe Asp Ser
1 5
<210>20
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>20
Met Pro Asn Trp Gly Ser Leu Asp Thr
1 5
<210>21
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>21
Met Pro Asn Trp Gly Ser Leu Asp Ala
1 5
<210>22
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>22
Met Pro Asn Trp Gly Ser Leu Asp Asn
1 5
<210>23
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>23
Met Pro Asn Trp Gly Ala Leu Asp Ser
1 5
<210>24
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>24
Met Pro Asn Trp Gly Ser Leu Asp Asn
1 5
<210>25
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>25
Met Pro Asn Trp Gly Ser Leu Asp Tyr
1 5
<210>26
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>26
Met Pro Asn Trp Gly Ser Phe Asp His
1 5
<210>27
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>27
Met Pro Asn Trp Gly Ser Leu Asp Ser
1 5
<210>28
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>28
Met Pro Asn Trp Gly Ser Leu Asp Gly
1 5
<210>29
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>29
Val Pro Asn Trp Gly Ser Leu Asp Gly
1 5
<210>30
<211>28
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>30
Cys Ala Arg Phe Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly
1 5 10 15
Thr Leu Val Thr Val Ser Ser Ala Ser Ile Lys Gly
20 25
<210>31
<211>28
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>31
Cys Ala Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly
1 5 10 15
Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly
20 25
<210>32
<211>28
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>32
Cys Ala Arg Met Pro Asn Trp Gly Ser Phe Asp Tyr Trp Gly Gln Gly
1 5 10 15
Thr Leu Val Thr Val Ser Ser Ala Ser Ile Lys Gly
20 25
<210>33
<211>12
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>33
Val Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp
1 5 10
<210>34
<211>27
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>34
Val Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
1 5 10 15
Leu Val Thr Val Ser Ser Ala Asp Ile Lys Gly
20 25
<210>35
<211>27
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>35
Ala Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
1 5 10 15
Leu Val Thr Val Ser Ser Ala Ser Ile Lys Gly
20 25
<210>36
<211>25
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>36
Phe Pro Asn Trp Gly Ser Phe Asp Tyr Trp Gly Gln Gly Thr Leu Val
1 5 10 15
Thr Val Ser Ser Ala Ser Ile Lys Gly
20 25
<210>37
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>37
Val Pro Asn Trp Gly Ser Leu Asp Ala
1 5
<210>38
<211>9
<212>PRT
<213> Artificial sequence
<220>
<223> CDR H3 sequence
<400>38
Phe Pro Asn Trp Gly Ser Phe Asp Tyr
1 5
<210>39
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223>CDL3
<220>
<221>MISC_FEATURE
<222>(2)..(2)
<223> Xaa represents Gln, Arg, Met, Ser, Thr or Val
<220>
<221>MISC_FEATURE
<222>(3)..(3)
<223> Xaa represents Tyr or Val
<220>
<221>MISC_FEATURE
<222>(4)..(4)
<223> Xaa represents Glu, Ala, Gly or Ser
<220>
<221>MISC_FEATURE
<222>(5)..(5)
<223> Xaa represents Thr, Ala or Ser
<400>39
Gln Xaa Xaa Xaa Xaa
1 5
<210>40
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>40
Gln Arg Tyr Ala Thr
1 5
<210>41
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>41
Gln Arg Tyr Ser Thr
1 5
<210>42
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>42
Gln Met Tyr Ser Thr
1 5
<210>43
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>43
Gln Gln Val Gly Thr
1 5
<210>44
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>44
Gln Gln Val Ser Thr
1 5
<210>45
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>45
Gln Gln Tyr Ser Thr
1 5
<210>46
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>46
Gln Ser Tyr Ser Thr
1 5
<210>47
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>47
Gln Gln Tyr Ala Thr
1 5
<210>48
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>48
Gln Gln Tyr Ser Ser
1 5
<210>49
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>49
Gln Thr Tyr Ser Thr
1 5
<210>50
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>50
Gln Gln Tyr Gly Ser
1 5
<210>51
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>51
Gln Gln Tyr Glu Ala
1 5
<210>52
<211>5
<212>PRT
<213> Artificial sequence
<220>
<223> CDR L3 sequence
<400>52
Gln Gln Tyr Glu Thr
1 5
<210>53
<211>118
<212>PRT
<213> Artificial sequence
<220>
<223> VH region
<400>53
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu LysIle Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Phe Pro Asn Trp Gly Ser Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210>54
<211>118
<212>PRT
<213> Artificial sequence
<220>
<223> VH region
<400>54
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Met Pro Asn Trp Gly Ser Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210>55
<211>118
<212>PRT
<213> Artificial sequence
<220>
<223> VH region
<400>55
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Val Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210>56
<211>103
<212>PRT
<213> Artificial sequence
<220>
<223> VL region
<400>56
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Glu Thr Phe Gly
85 90 95
Gln Gly Thr Lys Val Glu Ile
100
<210>57
<211>103
<212>PRT
<213> Artificial sequence
<220>
<223> VL region
<400>57
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Ala Ser Phe Gly
85 90 95
Gln Gly Thr Lys Val Glu Ile
100
<210>58
<211>103
<212>PRT
<213> Artificial sequence
<220>
<223> VL region
<400>58
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Glu Ala Phe Gly
85 90 95
Gln Gly Thr Lys Val Glu Ile
100
<210>59
<211>444
<212>PRT
<213> Artificial sequence
<220>
<223> heavy chain
<400>59
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Met Pro Asn Trp Gly Ser Phe Asp Tyr Trp Gly Gln Gly Thr
100105 110
Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190
Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys
210 215 220
Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu
245 250 255
Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln
260265 270
Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys
275 280 285
Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu
290 295 300
Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys
305 310 315 320
Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys
325 330 335
Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser
340 345 350
Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys
355 360 365
Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln
370 375 380
Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln
405 410 415
Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn
420 425430
His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly
435 440
<210>60
<211>444
<212>PRT
<213> Artificial sequence
<220>
<223> heavy chain
<400>60
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Val Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190
Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys
210 215 220
Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu
245 250 255
Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln
260 265 270
Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys
275 280 285
Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu
290 295 300
Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys
305 310 315 320
Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys
325 330 335
Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser
340 345 350
Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys
355 360 365
Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln
370 375 380
Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln
405 410 415
Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn
420 425 430
His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly
435 440
<210>61
<211>444
<212>PRT
<213> Artificial sequence
<220>
<223> heavy chain
<400>61
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Val Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Ile Lys Gly ProSer Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190
Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys
210 215 220
Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu
245 250 255
Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln
260 265 270
Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala LysThr Lys
275 280 285
Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu
290 295 300
Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys
305 310 315 320
Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys
325 330 335
Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser
340 345 350
Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys
355 360 365
Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln
370 375 380
Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln
405 410 415
Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn
420 425 430
His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly
435 440
<210>62
<211>444
<212>PRT
<213> Artificial sequence
<220>
<223> heavy chain
<400>62
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190
Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys
210 215 220
Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu
245 250 255
Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln
260 265 270
Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys
275280 285
Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu
290 295 300
Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys
305 310 315 320
Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys
325 330 335
Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser
340 345 350
Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys
355 360 365
Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln
370 375 380
Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln
405 410 415
Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn
420 425 430
His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly
435 440
<210>63
<211>444
<212>PRT
<213> Artificial sequence
<220>
<223> heavy chain
<400>63
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Ile Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser
180 185 190
Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly Pro Pro Cys
210 215 220
Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val Phe Leu
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu
245 250 255
Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln
260 265 270
Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys
275 280 285
Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser Val Leu
290 295 300
Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys
305 310 315 320
Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys
325 330 335
Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser
340 345 350
Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys
355 360 365
Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln
370 375 380
Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg Trp Gln
405 410 415
Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn
420 425 430
His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly
435 440
<210>64
<211>443
<212>PRT
<213> Artificial sequence
<220>
<223> heavy chain
<400>64
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Val Ile Tyr Pro Gly Asp Ser Tyr Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly Gln Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Met Pro Asn Trp Gly Ser Leu Asp His Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro
115 120 125
Leu Ala Pro Cys Ser Arg Ser ThrSer Glu Ser Thr Ala Ala Leu Gly
130 135 140
Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn
145 150 155 160
Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln
165 170 175
Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Thr Ser Ser
180 185 190
Asn Phe Gly Thr Gln Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser
195 200 205
Asn Thr Lys Val Asp Lys Thr Val Glu Arg Lys Cys Cys Val Glu Cys
210 215 220
Pro Pro Cys Pro Ala Pro Pro Val Ala Gly Pro Ser Val Phe Leu Phe
225 230 235 240
Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val
245 250 255
Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu Val Gln Phe
260 265 270
Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro
275 280 285
Arg Glu Glu Gln Phe Asn Ser Thr Phe ArgVal Val Ser Val Leu Thr
290 295 300
Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val
305 310 315 320
Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys Thr
325 330 335
Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg
340 345 350
Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly
355 360 365
Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro
370 375 380
Glu Asn Asn Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp Gly Ser
385 390 395 400
Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln
405 410 415
Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His
420 425 430
Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440
<210>65
<211>211
<212>PRT
<213> Artificial sequence
<220>
<223> light chain
<400>65
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Glu Ala Phe Gly
85 90 95
Gln Gly Thr Lys Val Glu Ile Lys Arg Thr Val Ala Ala Pro Ser Val
100 105 110
Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu Lys Ser Gly Thr Ala Ser
115 120 125
Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala Lys Val Gln
130 135 140
Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser Gln Glu Ser Val
145 150 155 160
Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser Ser Thr Leu
165 170 175
Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr Ala Cys Glu
180 185 190
Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys Ser Phe Asn Arg
195 200 205
Gly Glu Cys
210
<210>66
<211>211
<212>PRT
<213> Artificial sequence
<220>
<223> light chain
<400>66
Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Ile Ser Ser Ser
20 25 30
Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu
35 40 45
Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser
50 55 60
Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu
65 70 75 80
Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Glu Thr Phe Gly
85 90 95
Gln Gly Thr Lys Val Glu Ile Lys Arg Thr Val Ala Ala Pro Ser Val
100 105 110
Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu Lys Ser Gly Thr Ala Ser
115 120 125
Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala Lys Val Gln
130 135 140
Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser Gln Glu Ser Val
145 150 155 160
Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser Ser Thr Leu
165 170 175
Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr Ala Cys Glu
180 185 190
Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys Ser Phe Asn Arg
195 200 205
Gly Glu Cys
210

Claims (29)

1. A method of treating Cutaneous T Cell Lymphoma (CTCL) comprising administering to a patient in need thereof a therapeutically effective amount of an antagonist antibody or binding fragment thereof specific for an IL-13 receptor.
2. The method of claim 1, wherein said antibody thereof is specific for IL-13R α 1(IL-13R alpha1) or anti-IL-13R α 2(IL13R alpha 2).
3. The method of claim 2, wherein the antibody or binding fragment thereof is an anti-IL-13R α 1 antibody or binding fragment.
4. The method of claim 2 or 3, wherein the antibody binds epitope FFYQ.
5. The method of any one of claims 1 to 4, wherein the antibody or binding fragment thereof has a heavy chain variable domain comprising CDR H1 shown in SEQ ID NO 1, CDR H2 shown in SEQ ID NO 2, and CDR H3 having a sequence independently selected from SEQ ID NO 3, 4, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, and 38, or a variable domain wherein one, two, or three amino acids in the CDRs are substituted with additions or deletions.
6. The method of any one of claims 1 to 5, wherein the antibody or binding fragment thereof has a light chain variable domain comprising CDR L1 shown in SEQ ID NO 5, CDR L2 shown in SEQ ID NO 6, and CDR L3 independently selected from SEQ ID NO 7, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, and 52, or a variable domain wherein one, two, or three amino acids of the CDRs are substituted with additions or deletions.
7. The method of any one of claims 1 to 6, wherein the antibody or binding fragment thereof has a light chain variable domain comprising the sequence set forth in SEQ ID NOs 9, 56, 57, 58 or a sequence at least 95% percent identical thereto.
8. The method of any one of claims 1 to 7, wherein the antibody or binding fragment thereof has a light chain variable domain comprising the sequence set forth in SEQ ID NO 9.
9. The method of any one of claims 1 to 8, wherein the antibody or binding fragment thereof has a heavy chain variable domain comprising the sequence set forth in SEQ ID NOs 8, 53, 54, 55 or a sequence at least 95% percent identical thereto.
10. The method of any one of claims 1 to 9, wherein the antibody or binding fragment thereof has a heavy chain variable domain comprising the sequence set forth in seq id No. 8.
11. The method of any one of claims 1 to 10, wherein the antibody or binding fragment thereof is a human or humanized antibody.
12. The method of any one of claims 1 to 11, wherein the antibody binds to a fragment, such as a fragment selected from the group consisting of: fv, dsFv, scFv, Fab 'or F (ab') 2And (3) fragment.
13. The method of any one of claims 1-11, wherein the antibody is a full-length antibody.
14. The method according to claim 13, wherein the antibody heavy chain has the sequence shown in SEQ ID No. 10, 59, 60, 61, 62, 63 or 64, in particular SEQ ID No. 10.
15. The method according to claim 13 or 14, wherein the antibody light chain has the sequence shown in SEQ ID No. 11, 65 or 66, in particular SEQ ID No. 11.
16. The method of any one of claims 1-15, wherein the antibody or binding fragment thereof inhibits IL-13 signaling through an IL-13 receptor complex.
17. The method of any one of claims 1-16, wherein the CTCL is refractory to first-line therapy.
18. The method of any one of claims 1-17, wherein the lymphoma is mycosis fungoides.
19. The method of any one of claims 1-18, wherein the lymphoma is sezary syndrome.
20. The method of any one of claims 1 to 19, wherein the antibody or binding fragment thereof is administered in a pharmaceutical formulation, such as a parenteral formulation.
21. The method of any one of claims 1 to 20, wherein the anti-IL 13 receptor antibody is administered at a dose in the range of 1ng to 1000 μ g.
22. The method of any one of claims 1-21, wherein the antibody or antibody binding fragment is used as a monotherapy.
23. The method of any one of claims 1-21, wherein the antibody or binding fragment is used as part of a combination therapy comprising another therapeutic agent.
24. The method of claim 23, wherein the other therapeutic agent is an anti-cancer agent, e.g., a chemotherapeutic agent or a combination of chemotherapeutic agents, such as selected from the group comprising: platinum (such as cisplatin or oxaliplatin), gemcitabine, capecitabine, 5-FU, FOLFOX, FOLFIRI, flofirnox, and combinations of two or more thereof.
25. The method of claim 23 or 24, wherein the therapeutic agent is an immunomodulatory agent, such as a cytokine, for example selected from the group comprising: IL-13, IL-12 and IL-2.
26. The method of any one of claims 1-25, wherein the antibody or binding fragment is bound to a payload.
27. The method of claim 26, wherein the payload is a toxin or a drug molecule.
28. An anti-IL-13 receptor antibody or antibody fragment thereof, such as an anti-IL-13R α 1 antibody, for use in the treatment of cutaneous T-cell lymphoma (CTCL), such as sezary syndrome.
29. Use of an anti-IL-13 receptor antibody or antibody fragment thereof, such as an anti-IL-13R α 1 antibody, for the manufacture of a medicament for the treatment of cutaneous T-cell lymphoma (CTCL), such as sezary syndrome.
CN201880042193.3A 2017-06-30 2018-06-29 Methods of treatment using IL-13R antibodies Pending CN110785434A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
SG10201705435W 2017-06-30
SG10201705435W 2017-06-30
PCT/SG2018/050319 WO2019004943A1 (en) 2017-06-30 2018-06-29 Method of treatment using il-13r antibody

Publications (1)

Publication Number Publication Date
CN110785434A true CN110785434A (en) 2020-02-11

Family

ID=69383110

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201880042193.3A Pending CN110785434A (en) 2017-06-30 2018-06-29 Methods of treatment using IL-13R antibodies

Country Status (3)

Country Link
EP (1) EP3645566A1 (en)
JP (1) JP2020525500A (en)
CN (1) CN110785434A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024041650A1 (en) * 2022-08-25 2024-02-29 Nanjing Legend Biotech Co., Ltd. Chimeric antigen receptors targeting interleukin 13 receptor subunit alpha 2 and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016123142A1 (en) * 2015-01-26 2016-08-04 The University Of Chicago IL13RAα2 BINDING AGENTS AND USE THEREOF IN CANCER TREATMENT

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003080675A2 (en) * 2002-03-22 2003-10-02 Amrad Operations Pty Ltd MONOCLONAL ANTIBODY AGAINST INTERLEUKIN-13 RECEPTOR ALPHA 1 (IL-13Rα1)
JP2007031414A (en) * 2005-06-24 2007-02-08 International Medical Center Of Japan Therapeutic agent of mucous membrane damage of digestive tract by using il-13 signaling inhibition as mechanism, and method for screening medicament
PL2829551T3 (en) * 2006-10-19 2018-04-30 Csl Limited High affinity antibody antagonists of interleukin-13 receptor alpha 1

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016123142A1 (en) * 2015-01-26 2016-08-04 The University Of Chicago IL13RAα2 BINDING AGENTS AND USE THEREOF IN CANCER TREATMENT

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LARISA J. GESKIN等: "Interleukin-13 is overexpressed in cutaneous T-cell lymphoma cells and regulates their proliferation", 《BLOOD》 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024041650A1 (en) * 2022-08-25 2024-02-29 Nanjing Legend Biotech Co., Ltd. Chimeric antigen receptors targeting interleukin 13 receptor subunit alpha 2 and methods of use thereof

Also Published As

Publication number Publication date
EP3645566A1 (en) 2020-05-06
JP2020525500A (en) 2020-08-27

Similar Documents

Publication Publication Date Title
JP7458981B2 (en) Combination of antibody-drug conjugate and tubulin inhibitor
WO2020130125A1 (en) Combination of antibody-drug conjugate and kinase inhibitor
JP6224268B2 (en) Duocarmycin ADC for use in the treatment of endometrial cancer
EP3347054B1 (en) Dosing regimens for anti-tf-antibody drug-conjugates
TW200838875A (en) Combination therapy with angiogenesis inhibitors
JP2017513901A (en) Method of treating early stage breast cancer with trastuzumab-MCC-DM1 and pertuzumab
JP7473474B2 (en) Treatment of metastatic brain tumors by administration of antibody-drug conjugates
TW202034959A (en) Combination of antibody-drug conjugate and parp inhibitor
JP2018516968A (en) Pharmaceutical combinations and uses thereof
AU2018285523A1 (en) Antibody drug conjugates that bind LGR5
CN112367999A (en) Combination therapy
CN112105380A (en) Netin-1 binding antibodies and uses thereof
CN110785434A (en) Methods of treatment using IL-13R antibodies
US20200165347A1 (en) Method of treatment using il-13r antibody
US20230034186A1 (en) Methods of treating cancer using multi-specific binding proteins that bind nkg2d, cd16 and a tumor-associated antigen
WO2022109302A9 (en) Anti-galectin-9 antibodies and therapeutic uses thereof
CN115666724A (en) Method for treating cervical cancer by administering the PD-1 inhibitor antibody, cimirapril mab
CN116194142A (en) Methods of treating cancer pain by administering PD-1 inhibitors
JP2024095789A (en) Treatment of metastatic brain tumors by administration of antibody-drug conjugates
CN116490209A (en) Combination therapy and methods of use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20200211

WD01 Invention patent application deemed withdrawn after publication