Combination Treatment For Non-hematologic Malignancies

Gualberto; Antonio ;   et al.

Patent Application Summary

U.S. patent application number 12/567040 was filed with the patent office on 2011-01-20 for combination treatment for non-hematologic malignancies. This patent application is currently assigned to PFIZER INC.. Invention is credited to Bruce D. Cohen, Antonio Gualberto, Carrie L. Melvin, M. Luisa Roberts.

Application Number20110014207 12/567040
Document ID /
Family ID35058695
Filed Date2011-01-20

United States Patent Application 20110014207
Kind Code A1
Gualberto; Antonio ;   et al. January 20, 2011

COMBINATION TREATMENT FOR NON-HEMATOLOGIC MALIGNANCIES

Abstract

The present invention relates to a therapeutic method for the treatment of non-hematologic malignancies comprising administering anti-IGF-1R antibodies, particularly human anti-IGF-1R antibodies, to a patient, in conjunction with the administration of at least one other therapeutic agent. The invention further relates to pharmaceutical compositions comprising these antibodies and methods of using such compositions thereof for treatment.


Inventors: Gualberto; Antonio; (East Greenwich, RI) ; Cohen; Bruce D.; (East Lyme, CT) ; Melvin; Carrie L.; (Wayland, MA) ; Roberts; M. Luisa; (Noank, CT)
Correspondence Address:
    PFIZER INC;Mary J Hosley
    150 EAST 42ND STREET, MS: 150/02/E112
    NEW YORK
    NY
    10017-5612
    US
Assignee: PFIZER INC.

Family ID: 35058695
Appl. No.: 12/567040
Filed: September 25, 2009

Related U.S. Patent Documents

Application Number Filing Date Patent Number
11182343 Jul 15, 2005 7618626
12567040
60588721 Jul 16, 2004

Current U.S. Class: 424/158.1
Current CPC Class: A61K 31/28 20130101; A61K 31/28 20130101; A61K 31/337 20130101; A61K 45/06 20130101; A61K 39/39541 20130101; A61K 31/573 20130101; A61K 31/337 20130101; A61K 39/39541 20130101; A61P 35/00 20180101; A61K 2300/00 20130101; A61K 31/573 20130101; A61K 2300/00 20130101; A61K 2300/00 20130101; A61K 2300/00 20130101
Class at Publication: 424/158.1
International Class: A61K 39/395 20060101 A61K039/395; A61P 35/00 20060101 A61P035/00

Claims



1. A method for the treatment of lung cancer in a patient in need of such treatment comprising the step of administering to the patient a therapeutically effective amount of an antibody that specifically binds to IGF-1R in combination with a therapeutically effective amount of a taxane and a therapeutically effective amount of carboplatin, wherein the antibody is selected from the group consisting of 2.12.1, 2.13.2, 2.14.3, 4.9.2, 4.17.3, and 6.1.1.

2. (canceled)

3. The method of claim 1, wherein the taxane is docetaxel.

4. The method of claim 1, wherein the taxane is paclitaxel.

5-16. (canceled)

17. The method according to claim 1, where in the amount of the antibody is in the range of 0.05-20 mg/kg.

18. The method according to claim 1, where in the amount of the antibody is in the range of 0.5-10 mg/kg.

19. The method according to claim 4, wherein the antibody is 2.13.2.

20. The method according to claim 4, wherein the cancer is non-small cell lung cancer.

21. A method of treatment for non-small cell lung cancer in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of antibody 2.13.2 in combination of carboplatin and a therapeutically effective amount of paclitaxel.

22. The method according to claim 21, wherein the amount of the antibody is in the range of 0.05-20 mg/kg.

23. The method according to claim 21, wherein the antibody is administered once every two weeks, once every three weeks, or once every month.
Description



BACKGROUND OF THE INVENTION

[0001] The present invention relates to a method of treatment for non-hematologic malignancies comprising the administration of anti-insulin-like growth factor I receptor (IGF-1R) antibodies, in conjunction with other therapeutic agents such as chemotherapeutic agents and hormonal therapy.

[0002] The insulin-like growth factor (IGF) signaling system plays an important role in the growth and development of many tissues and regulates overall growth. Insulin-like growth factor (IGF-1) is a 7.5-kD polypeptide that circulates in plasma in high concentrations and is detectable in most tissues. IGF-1 stimulates cell differentiation and cell proliferation, and is required by most mammalian cell types for sustained proliferation. These cell types include, among others, human diploid fibroblasts, epithelial cells, smooth muscle cells, T lymphocytes, neural cells, myeloid cells, chondrocytes, osteoblasts and bone marrow stem cells.

[0003] The first step in the transduction pathway leading to IGF-1-stimulated cellular proliferation or differentiation is binding of IGF-1 or IGF-2 (or insulin at supraphysiological concentrations) to the IGF-1 receptor. The IGF-1 receptor (IGF-1R) is composed of two types of subunits: an alpha subunit (a 130-135 kD protein that is entirely extracellular and functions in ligand binding) and a beta subunit (a 95-kD transmembrane protein, with transmembrane and cytoplasmic domains). IGF binding proteins (IGFBPs) have growth inhibiting effects by, at least in part, competitively binding IGFs and preventing their association with IGF-1F. The interactions between IGF-1, IGF-2, IGF1R, and IGFBPs affect many physiological and pathological processes such as development, growth and metabolic regulation.

[0004] The IGF-1R is initially synthesized as a single chain proreceptor polypeptide that is processed by glycosylation, proteolytic cleavage, and covalent bonding to assemble into a mature 460-kD heterotetramer comprising two alpha-subunits and two beta-subunits. The beta subunit(s) possesses ligand-activated tyrosine kinase activity. This activity is implicated in the signaling pathways mediating ligand action which involve autophosphorylation of the beta-subunit and phosphorylation of IGF-1R substrates.

[0005] There is considerable evidence for a role for IGF-1 and/or IGF-1R in the maintenance of tumor cells in vitro and in vivo. IGF-1R levels are elevated in tumors of lung (Kaiser et al., J. Cancer Res. Clin. Oncol. 119: 665-668, 1993; Moody et al., Life Sciences 52: 1161-1173, 1993; Macauley et al., Cancer Res., 50: 2511-2517, 1990), breast (Pollack et al., Cancer Lett. 38: 223-230, 1987; Foekens et al., Cancer Res. 49: 7002-7009, 1989; Cullen et al., Cancer Res. 49: 7002-7009, 1990; Arteaga et al., J. Clin. Invest. 84: 1418-1423, 1989), prostate and colon (Remaole-Bennet et al., J. Clin. Endocrinol. Metab. 75: 609-616, 1992; Guo et al., Gastroenterol. 102: 1101-1108, 1992). In addition, IGF-1 appears to be an autocrine stimulator of human gliomas (Sandberg-Nordqvist et al., Cancer Res. 53: 2475-2478, 1993), while IGF-1 stimulated the growth of fibrosarcomas that overexpressed IGF-1R (Butler et al., Cancer Res. 58: 3021-27, 1998). In addition, individuals with "high normal" levels of IGF-1 have an increased risk of common cancers compared to individuals with IGF-1 levels in the "low normal" range (Rosen et al., Trends Endocrinol. Metab. 10: 136-41, 1999). For a review of the role IGF-1/IGF-1 receptor interaction plays in the growth of a variety of human tumors, see Macaulay, Br. J. Cancer, 65: 311-320, 1992.

[0006] Numerous classes of antineoplastic agents are currently in use. Docetaxel, one of a group of drugs called "taxanes," which are derived from the bark and needles of yew trees, is the first anticancer agent to show a significantly higher response rate than doxorubicin, a very active agent and widely used chemotherapy in the first-line treatment of metastatic breast cancer. Docetaxel also is the first chemotherapy drug as a single agent to demonstrate increased survival among patients with advanced breast cancer compared to the combination of mitomycin C and vinblastine, a commonly used regimen in this patient population. Median time to progression and time to treatment failure were significantly longer for docetaxel than for mitomycin C in combination with vinblastine, and the one-year survival rate significantly greater. Promising results have also been recorded for docetaxel in other human malignancies, such as ovarian, lung, head and neck, gastric and pancreatic cancers.

[0007] Paclitaxel, also a taxane, binds to microtubules and prevents their molecular disassembly, thereby inhibiting mitosis (cell division). With the spindle still in place the cell cannot divide into daughter cells. Paclitaxel is most effective against ovarian carcinomas and advanced breast carcinomas.

[0008] Hormonal therapy can be very effective in lowering the risk of recurrence for women with hormone-receptor-positive breast cancer. Tamoxifen is the hormonal therapy that has been around the longest--nearly 30 years. It blocks the effect of estrogen on breast cancer cells, keeping the cells from growing. Tamoxifen can reduce recurrence by 40-50% in post-menopausal women, and by 30-50% in pre-menopausal women. It also lowers the risk of a new breast cancer developing in the unaffected breast, and can slow down the progression of advanced disease.

[0009] In recent years, aromatase inhibitors have been used as hormonal therapy. This type of therapy is recommended only for postmenopausal women with hormone-receptor-positive breast cancer. It works by blocking the production of estrogen in muscle and fat tissue, which is the main source of estrogen in women beyond menopause, after which the ovaries stop making significant levels of estrogen.

[0010] Prostate cancer is the most common cancer and the second cause of cancer death in men in the United States. About 10% of the initial cases of prostate cancer present with metastatic disease. However, in the rest, metastases will develop despite treatment with surgery, radiation or medical therapy, and those metastases will eventually become refractory to hormonal treatment. The use of chemotherapy in hormone refractory (androgen independent) progressive prostate cancer (HRPC) has been characterized historically by poor efficacy and high toxicity. Newer regimens containing docetaxel have shown a survival benefit over previous palliative regimens. Despite this positive trend, the median survival of HRPC patients treated with docetaxel and prednisone is only 18.9 months; clearly, more effective regimens are required for the treatment of HRPC patients.

[0011] Although some currently available anti-cancer treatments have been successful, complete responses to these treatments are infrequently observed, and the patient population refractory to these treatments is still large. Thus, development of new therapeutic regimens, particularly those capable of augmenting or potentiating the anti-tumor activity of other anti-neoplastic agents, is necessary.

[0012] In view of the roles that IGF-1 and IGF-1R have in such disorders as cancer and other proliferative disorders when IGF-1 and/or IGF-1R are overexpressed, antibodies to IGF-1R have been produced that block binding of IGF-1 or IGF-2 to IGF-1R. Such antibodies are described, for example, in International Patent Application No. WO 02/053596, published Jul. 11, 2002; International Patent Application Nos. WO 05/016967 and WO 05/016970, both published Feb. 24, 2005; International Patent Application No. WO 03/106621, published Dec. 24, 2003; International Patent Application No. WO 04/083248, published Sep. 30, 2004; International Patent Application No. WO 03/100008, published Dec. 4, 2003; International Patent Publication WO 04/087756, published Oct. 14, 2004; and International Patent Application No WO 05/005635, published Jan. 26, 2005. Because of their ability to block a tumor cell survival pathway, it is desirable to use such anti-IGF-1R antibodies to treat cancer, particularly non-hematological malignancies, in patients to obtain an improved clinical benefit relative to standard cancer treatment regimes alone.

SUMMARY OF THE INVENTION

[0013] The present invention is directed to a method for the treatment of an advanced non-hematologic malignancy in a patient in need of such treatment comprising the step of administering to the patient a therapeutically effective amount of an anti-IGF-1R antibody.

[0014] More particularly, the present invention is directed to a method comprising the step of administering to the patient an antibody that specifically binds to IGF-1R in combination with a therapeutically effective amount of at least one agent selected from the group consisting of an alkylating agent, a folate antagonist, a pyrimidine antagonist, a cytotoxic antibiotic, a platinum compound, a taxane, a vinca alkaloid, a topoisomerase inhibitor, an EGFR inhibitor, and a hormonal therapy agent. Preferably the antibody is one that specifically binds to human IGF-1R.

[0015] In a preferred embodiment of the present invention, the anti-IGF-1R antibody has the following properties: (a) a binding affinity for human IGF-1R of K.sub.d of 8.times.10.sup.-9 or less, and (b) inhibition of binding between human IGF-1R and IGF-1 with an IC.sub.50 of less than 100 nM.

[0016] In another preferred embodiment of the present invention, the anti-IGF-1R antibody comprises (a) a heavy chain comprising the amino acid sequences of CDR-1, CDR-2, and CDR-3 of an antibody selected from the group consisting of 2.12.1, 2.13.2, 2.14.3, 4.9.2, 4.17.3, and 6.1.1, and (b) a light chain comprising the amino acid sequences of CDR-1, CDR-2, and CDR-3 of an antibody selected from the group consisting of 2.12.1, 2.13.2, 2.14.3, 4.9.2, 4.17.3, and 6.1.1, or (c) sequences having changes from the CDR sequences of an antibody selected from the group consisting of 2.12.1, 2.13.2, 2.14.3, 4.9.2, 4.17.3, and 6.1.1, said sequences being selected from the group consisting of conservative changes, wherein the conservative changes are selected from the group consisting of replacement of nonpolar residues by other nonpolar residues, replacement of polar charged residues by other polar uncharged residues, replacement of polar charged residues by other polar charged residues, and substitution of structurally similar residues; and non-conservative substitutions, wherein the non-conservative substitutions are selected from the group consisting of substitution of polar charged residue for polar uncharged residues and substitution of nonpolar residues for polar residues, additions and deletions.

[0017] The present invention is also directed to a pharmaceutical composition for the treatment of a non-hematologic malignancy comprising (a) a therapeutically effective amount of an antibody that specifically binds IGF-1R, (b) a therapeutically effective amount of at least one agent selected from the group consisting of an alkylating agent, a folate antagonist, a pyrimidine antagonist, a cytotoxic antibiotic, a platinum compound, a taxane, a vinca alkaloid, a topoisomerase inhibitor, an EGFR inhibitor, and a hormonal therapy agent; and (c) a pharmaceutically acceptable carrier.

DETAILED DESCRIPTION OF THE DRAWINGS

[0018] FIGS. 1A-10 show alignments of the nucleotide sequences of the light chain variable regions from six human anti-IGF-1R antibodies to each other and to germline sequences. FIG. 1A shows the alignment of the nucleotide sequences of the variable region of the light chain (VL) of antibodies 2.12.1 (SEQ ID NO: 1) 2.13.2 (SEQ ID NO: 5), 2.1-4.3 (SEQ ID NO: 9) and 4.9.2 (SEQ ID NO: 13) to each other and to the germline V.kappa. A30 sequence (SEQ ID NO: 39). FIG. 1B shows the alignment of the nucleotide sequence of VL of antibody 4.17.3 (SEQ ID NO: 17) to the germline V.kappa. O12 sequence (SEQ ID NO: 41). FIG. 10 shows the alignment of the nucleotide sequence of VL of antibody 6.1.1 (SEQ ID NO: 21) to the germline V.kappa. A27 sequence (SEQ ID NO: 37). The alignments also show the CDR regions of the VL from each antibody. The consensus sequences for FIGS. 1A-10 are shown in SEQ ID NOS: 53-55, respectively.

[0019] FIGS. 2A-2D show alignments of the nucleotide sequences of the heavy chain variable regions from six human anti-IGF-1R antibodies to each other and to germline sequences. FIG. 2A shows the alignment of the nucleotide sequence of the VH of antibody 2.12.1 (SEQ ID NO: 3) to the germline VH DP-35 sequence (SEQ ID NO: 29). FIG. 2B shows the alignment of the nucleotide sequence of the VH of antibody 2.14.3 (SEQ ID NO: 11) to the germline VIV-4/4.35 sequence (SEQ ID NO: 43). FIGS. 2C-1 and 2C-2 show the alignments of the nucleotide sequences of the VH of antibodies 2.13.2 (SEQ ID NO: 7), 4.9.2 (SEQ ID NO: 15) and 6.1.1 (SEQ ID NO: 23) to each other and to the germline VH DP-47 sequence (SEQ ID NO: 31). FIG. 2D shows the alignment of the nucleotide sequence of the VH of antibody 4.17.3 (SEQ ID NO: 19) to the germline VH DP-71 sequence (SEQ ID NO: 35). The alignment also shows the CDR regions of the antibodies. The consensus sequences for FIGS. 2A-2D are shown in SEQ ID NOS: 56-59, respectively.

[0020] FIG. 3A shows the number of mutations in different regions of the heavy and light chains of 2.13.2 and 2.12.1 compared to the germline sequences. FIGS. 3A-D show alignments of the amino acid sequences from the heavy and light chains of antibodies 2.13.2 and 2.12.1 with the germline sequences from which they are derived. FIG. 3B shows an alignment of the amino acid sequence of the heavy chain of antibody 2.13.2 (SEQ ID NO: 45) with that of germline sequence DP-47(3-23)/D6-19/JH6 (SEQ ID NO: 46). FIG. 3C shows an alignment of the amino acid sequence of the light chain of antibody 2.13.2 (SEQ ID NO: 47) with that of germline sequence A30/Jk2 (SEQ ID NO: 48). FIG. 3D shows an alignment of the amino acid sequence of the heavy chain of antibody 2.12.1 (SEQ ID NO: 49) with that of germline sequence DP-35(3-11)/D3-3/JH6 (SEQ ID NO: 50). FIG. 3E shows an alignment of the amino acid sequence of the light chain of antibody 2.12.1 (SEQ ID NO: 51) with that of germline sequence A30/Jk1 (SEQ ID NO: 52). For FIGS. 3B-E, the signal sequences are in italic, the CDRs are underlined, the constant domains are bold, the framework (FR) mutations are highlighted with a plus sign ("+") above the amino acid residue and CDR mutations are highlighted with an asterisk above the amino acid residue.

[0021] FIG. 4 shows that anti-IGF-1R antibodies 2.13.2 and 4.9.2 reduce IGF-1R phosphotyrosine signal in 3T3-IGF-1R tumors.

[0022] FIG. 5 shows that anti-IGF-1R antibody 2.13.2 inhibits 3T3-IGF-1R tumor growth in vivo.

DETAILED DESCRIPTION OF THE INVENTION

[0023] The present invention are directed to the treatment of non-hematologic malignancies, including breast, lung, brain, skin, ovarian, prostate, head and neck, colorectal, gastric, bladder, renal, esophageal, and pancreatic cancers, as well as solid tumors of childhood. Treatment of both early stage and advanced (metastatic) cancers are within the scope of the present invention. In preferred embodiments, the method of the present invention is used in the treatment of breast cancer, prostate cancer, and non-small cell lung cancer (NSCLC).

[0024] There are many classes of chemotherapeutic drugs currently in use for the treatment of non-hematological malignancies that are suitable for use in the combination therapy of the present invention. For example, alkylating agents are a class of drugs that alkylate DNA, restricting uncoiling and replication of strands. Alkylating agents include cyclophosphamide (CYTOXAN), ifosfamide (IFEX), mechlorethamine hydrochloride (MUSTARGEN), thiotepa (THIOPLEX), streptozotocin (ZANOSAR), carmustine (BICNU, GLIADEL WAFER), lomustine (CEENU), and dacarbazine (DTIC-DOME). A preferred alkylating agent for use in the methods of the present invention is cyclophosphamide.

[0025] Folate antagonists bind to dihydrofolate reductase (DHFR) and interfere with pyrimidine (thymidine) synthesis. Methotrexate (MATREX, FOLEX, TREXALL), trimetrexate (NEUTREXIN) and pemetrexed (ARIMTA) are folate antagonists suitable for use in the methods of the present invention. In addition to DHFR, pemetrexed also inhibits thymidylate synthase and glycinamide ribonucleotide formyl transferase, two other folate-dependent enzymes involved in thymidine synthesis.

[0026] Pyrimidine antagonists inhibit enzymes involved in pyrimidine synthesis. As pyrimidine analogs, they also interfere with DNA production by competing with normal nucleotides for incorporation into the DNA molecule. Pyrimidine antagonists suitable for use in the methods of the present invention include 5-fluorouracil (5-FU); capecitabine (XELODA), a prodrug of 5'-deoxy-5-fluorouridine (5'-FDUR), which is enzymatically converted to 5-FU in vivo; raltitrexed (TOMUDEX); tegafur-uracil (UFTORAL); and gemcitabine (GEMZAR).

[0027] Anthracycline antibiotics exert a cytotoxic effect by inhibiting the uncoiling of DNA by intercalation between DNA strands. Anthracyclines and anthracyclines derivatives include doxorubicin hydrochloride (ADRIAMYCIN, RUBEX, DOXIL), epirubicin hydrochloride (ELLENCE, PHARMORUBICIN), daunorubicin (CERUBIDINE, DAUNOXOME), nemorubicin, idarubicin hydrochloride (IDAMYCIN PFS, ZAVEDOS) and mitoxantrone (DHAD, NOVANTRONE). Preferred anthracyclines for use with the present invention include doxorubicin and epirubicin.

[0028] Other cytotoxic antibiotics are useful as cancer chemotherapeutic agents and suitable for use in the present invention. These include dactinomycin (actinomycin D, COSMEGEN), plicamycin (MITHRACIN), mitomycin (MUTAMYCIN), and bleomycin (BLENOXANE). Dactinomycin is particularly preferred.

[0029] Platinum compounds exert their anti-neoplastic effect by intercalation and intracalation between DNA strands, which inhibits uncoiling of the DNA. Platinum compounds useful in the methods of the present invention include cisplatin (PLATINOL) and carboplatin (PARAPLATIN).

[0030] Taxanes promote assembly of microtubules while inhibiting their disassembly into tubulin, thereby blocking a cell's ability to break down the mitotic spindle during mitosis. They have demonstrated significant activity against many solid tumors as single agent therapy and in combination with other chemotherapy agents. One embodiment of the combination therapy of the present invention includes the use of one or more taxanes in combination with the IGF-1R antibody. Suitable taxanes for use in combination with the IGF-1R antibody include docetaxel (TAXOTERE) and paclitaxel (TAXOL).

[0031] Vinca alkaloids, like taxanes, are "spindle poisons," acting on the microtubules that form the mitotic spindle. They inhibit mitosis by interfering with microtubule assembly, keeping the spindle from being formed. Vinca alkaloids include vindesine (ELDISINE), vinblastine sulfate (VELBAN), vincristine sulfate (ONCOVIN) and vinorelbine tartrate (NAVELBINE). A preferred vinca alkaloid for use in the methods of the present invention is vinorelbine.

[0032] The camptothecin analogs act through inhibition of topoisomerase I, an enzyme critical for DNA replication and packaging. Levels of topoisomerase I are higher in tumor cells than in normal tissue. Camptothecin analogs useful in the methods of the present invention include irinotecan (CAMPTOSAR) and topotecan (HYCAMTIN). Irinotecan is particularly preferred.

[0033] Inhibitors of topoisomerase II interfere with the normal DNA breakage resealing process (as do inhibitors of topoisomerase I), and they also interfere with the separation of newly replicated chromosomes, resulting in clastogenic mutation and potential cell death. The anthracyline antibiotics discussed above exhibit topoisomerase II inhibitory activity. Derivatives of podophyllotoxin, an extract of the mayapple that is an antimitotic glucoside) are also topoisomerase II inhibitors. Podophyllotoxin derivatives suitable for use in the present invention include etoposide (VEPESID), etoposide phosphate (ETOPOPHOS), and teniposide (VUMON). Etoposide is particularly preferred.

[0034] Compounds directed at inhibition of epidermal growth factor receptor (EGFR) tyrosine kinase (TK) represent a relatively new class of antineoplastic drugs that are useful in the method of the present invention. Many human cancers express members of the EGFR family on the cell surface. When a ligand binds to EGFR, it sets off a cascade of cellular reactions that result in increased cell division and influence other aspects of cancer development and progression, including angiogenesis, metastatic spread, and inhibition of apoptosis. EGFR-TK inhibitors may selectively target one of the members of the EGFR family (EGFR (also known as HER1 or ErbB-1), HER2/neu (also known as ErbB-2), HER3 (also known as ErbB-3), or HER4 (also known as ErbB-4)), or may target two or more of them. EGFR-TK inhibitors suitable for use in the present invention include gefitinib (IRESSA), erlotinib (TARCEVA), trastuzumab (HERCEPTIN), panitumumab (ABX-EGF; Abgenix/Amgen), lapatinib (GlaxoSmithKline), CI-1033 (Pfizer), GW2016 (GlaxoSmithKline), EKB-569 (Wyeth), PKI-166 (Novartis), CP-724,714 (Pfizer), and BIBX-1382 (Boeringer-Ingelheim). Additional EGFR-TK inhibitors are described in United States Patent Publication No. US 2002-0169165A1, published Nov. 14, 2002.

[0035] Another embodiment of the combination therapy of the present invention includes the use of hormonal therapy in combination with the IGF-1R antibody, particularly anti-estrogens in the treatment of breast cancer. Some hormonal treatments compete with estrogen for binding sites in breast tissue. These include tamoxifen citrate (NOLVADEX) and fulvestrant (FASLODEX). Similarly, anti-androgens block testosterone receptors and therefore are useful in the treatment of androgen-dependent prostate cancer.

[0036] Other hormone treatments include aromatase inhibitors. This class of hormonal agents inactivate aromatase, the enzyme which converts androgens to estrogens. Examples of aromatase inhibitors suitable for use in combination with the IGF-1R antibody include anastrozole (ARIMIDEX), letrozole (FEMARA), exemestane (AROMASIN), and fadrozole hydrochloride. Exemestane is a particularly preferred aromatase inhibitor for use in the methods of the present invention.

[0037] Co-administration of the antibody with an additional therapeutic agent (combination therapy) encompasses administering a pharmaceutical composition comprising both the anti-IGF-1R antibody and one or more additional therapeutic agents, and administering two or more separate pharmaceutical compositions, one comprising the anti-IGF-1R antibody and the other(s) comprising the additional therapeutic agent(s). Further, although co-administration or combination (conjoint) therapy generally mean that the antibody and additional therapeutic agents are administered at the same time as one another, it also encompasses simultaneous, sequential or separate dosing of the individual components of the treatment.

[0038] The present invention also encompasses the administration of other therapeutic agents in addition to the first and second components, either concurrently with one or more of those components, or sequentially. Such therapeutic agents include analgesics, cancer vaccines, anti-vascular agents, anti-proliferative agents, and anti-emetic agents. Preferred anti-emetic agents include aprepitant, ondansetron hydrochloride, granisetron hydrochloride, and metoclopramide.

[0039] Each administration may vary in its duration from a rapid administration to a continuous perfusion. As a result, for the purposes of the present invention, the combinations are not exclusively limited to those that are obtained by physical association of the constituents, but also to those that permit a separate administration, which can be simultaneous or spaced out over a period of time. The compositions according to the invention are preferably compositions which can be administered parentally. However, these compositions may be administered orally or intraperitoneally in the case of localized regional therapies.

[0040] As will be appreciated by one of skill in the art, the choice of therapeutic agents to be used in combination with IGF-1R antibodies, and the timing of their use, will be determined in part by the type and stage of the cancer that is being treated. For example, in early breast cancer (where the cancer has not spread outside the breast), surgery and radiation are generally followed by adjuvant chemotherapy or adjuvant hormonal therapy, either of which may be combined with IGF-1R antibodies in the methods of the present invention. Typical adjuvant chemotherapy for early breast cancer includes cyclophosphamide, methotrexate and 5-FU ("CMF"); 5-FU, doxorubicin, and cyclophosphamide ("FAC"); docetaxel, doxorubicin, and cyclophosphamide ("TAC"); doxorubicin and cyclophosphamide ("AC"); doxorubicin and cyclophosphamide followed by paclitaxel ("AC and T"); and 5-FU, epirubicin, and cyclophosphamide ("FEC"). Tamoxifen is a preferred hormonal treatment at this stage.

[0041] In locally advanced breast cancer, wherein the cancer has spread only to nearby tissues or lymph nodes, the patient is often given chemotherapy prior to surgery and radiation, which are then followed by adjuvant hormonal therapy. Alternatively, surgery/radiation is followed by adjuvant chemotherapy, then adjuvant hormonal therapy. IGF-1R antibodies may be administered in conjunction with the chemotherapeutic or hormonal therapy agents whether they are used either before or after surgery/radiation. Typical chemotherapy regimes for locally advanced breast cancer include FAC, AC, FEC, and doxorubicin plus docetaxel ("AT").

[0042] Metastatic breast cancer has spread to other parts of the body from the breast in which it started. Chemotherapy optionally may be preceded by hormonal therapy. First line hormonal therapy currently includes tamoxifen and anastrozole. First line chemotherapy regimens currently include FAC, TAC, docetaxel plus epirubicin, docetaxel, paclitaxel, capecitabine, vinorelbine, and trastuzumab. Second line chemotherapy treatments include docetaxel, alone or in combination with capecitabine. The methods of the present invention are suitable for use both as first line therapy and second line therapy.

[0043] In the United States, the combination of paclitaxel and carboplatin has become accepted as the standard of care for first line treatment of inoperable Stage IIIB (i.e. cancer has spread to structures near the lung, to lymph nodes in the mediastinum, or to lymph nodes on the other side of the chest or in the lower neck) and Stage IV (i.e. cancer has spread to other parts of the body or to another lobe of the lungs) non-small cell lung cancer (NSCLC). But the overall response rate is only approximately 28% for patients with performance status 0-1 in efficacy studies with a predominantly Stage IV population. In Europe, first line treatment for NSCLC is gemcitabine and cisplatin. Other treatment regimens for NSCLC include paclitaxel alone or with cisplatin or gemcitabine; docetaxel alone or with cisplatin or gemcitabine; vinorelbine alone or with gemcitabine; irinotecan alone or with gemcitabine; pemetrexed; and gefitinib.

[0044] It is known that signaling through IGF-1R is required for the tumorgenicity of cell lines and has been shown to decrease the cytotoxicity of chemotherapy, and that blocking IGF-1R activity enhances the effectiveness of current therapies and prevents tumor progression in animal models. It was therefore expected that an inhibitor of IGF-1R such as the antibodies of the present invention would reduce tumor cell survival and enhance the efficacy of chemotherapy when given in combination.

[0045] When incubated with cells, fully human monoclonal antibodies that are highly specific and potent inhibitors of IGF-1-induced receptor autophosphorylation induced down-regulation of IGF-1R by receptor internalization. The doses that down-regulated IGF-1R in solid tumor ex vivo models (31.25-125 .mu.g) corresponded to antibody concentrations of 8-40 .mu.g/ml at Day 1 and 2-20 .mu.g/ml at Day 9. Intraperitoneal administration of the anti-IGF-1R antibodies to athymic mice bearing tumors of the transfectant IGF-1R over-expressing NIH-3T3 cell line resulted in a dose dependent inhibition of tumor growth. The serum concentration of anti-IGF-1R antibodies that led to 50% growth inhibition was 20 .mu.g/ml at Day 1, and 13 .mu.g/ml at Day 9. Similar anti-tumor studies were extended to human tumor xenograft models. As a single agent, anti-IGF-1R antibodies inhibited the growth of several xenograft models including breast, lung and colorectal carcinomas.

[0046] The combination of anti-IGF-1R antibodies with paclitaxel or carboplatin was tested in the H460 and EBC-1 human NSCLC tumor xenograft models. Combination of anti-IGF-1R antibodies with those agents increased their tumor growth inhibition compared to each agent alone.

[0047] Unless otherwise defined herein, scientific, technical, and medical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, nomenclatures used in connection with, and techniques of, cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry described herein are those well known and commonly used in the art.

[0048] The following terms, unless otherwise indicated, shall be understood to have the following meanings:

[0049] An "antibody" refers to an intact immunoglobulin or to an antigen-binding portion thereof that competes with the intact antibody for specific binding. Antigen-binding portions may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies. Antigen-binding portions include, inter alia, Fab, Fab', F(ab').sub.2, Fv, dAb, and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), chimeric antibodies, diabodies and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.

[0050] Immunoglobulin chains exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. The CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N-terminus to C-terminus, both light and heavy chains comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987); Chothia et al., Nature 342:878-883 (1989).

[0051] An "isolated antibody" is an antibody that (1) is not associated with naturally-associated components, including other naturally-associated antibodies, that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, or (4) does not occur in nature. Examples of isolated antibodies include an anti-IGF-1R antibody that has been affinity purified using IGF-1R is an isolated antibody, an anti-IGF-1R antibody that has been synthesized by a hybridoma or other cell line in vitro, and a human anti-IGF-1R antibody derived from a transgenic mouse.

[0052] The term "chimeric antibody" refers to an antibody that contains one or more regions from one antibody and one or more regions from one or more other antibodies. In a preferred embodiment, one or more of the CDRs are derived from a human anti-IGF-1R antibody. In a more preferred embodiment, all of the CDRs are derived from a human anti-IGF-1R antibody. In another preferred embodiment, the CDRs from more than one human anti-IGF-1R antibodies are mixed and matched in a chimeric antibody. Further, the framework regions may be derived from one of the same anti-IGF-1R antibodies, from one or more different antibodies, such as a human antibody, or from a humanized antibody.

[0053] The term "epitope" includes any protein determinant capable of specific binding to an immunoglobulin or T-cell receptor. Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar sides chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. An antibody is said to specifically bind an antigen when the dissociation constant is preferably .ltoreq.100 nM and most preferably .ltoreq.10 nM.

[0054] As applied to polypeptides, the term "substantial identity" means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 75% or 80% sequence identity, preferably at least 90% or 95% sequence identity, even more preferably at least 98% or 99% sequence identity. Preferably, residue positions that are not identical differ by conservative amino acid substitutions. A "conservative amino acid substitution" is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein. In cases where two or more amino acid sequences differ from each other by conservative substitutions, the percent sequence identity or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well-known to those of skill in the art. See, e.g., Pearson, Methods Mol. Biol. 24: 307-31 (1994). Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; and 6) sulfur-containing side chains are cysteine and methionine. Conservative amino acids substitution groups include: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.

[0055] Preferred amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and (4) confer or modify other physicochemical or functional properties of such analogs. Analogs can include various mutations of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally-occurring sequence (preferably in the portion of the polypeptide outside the domain(s) forming intermolecular contacts. A conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence).

[0056] The phrase "in combination with" encompasses simultaneous, sequential or separate dosing of the individual components of the treatment. For example, the antibody may be administered once every three days, while the additional therapeutic agent is administered once daily. The antibody may be administered prior to or subsequent to treatment of the disorder with the additional therapeutic agent. Similarly, the anti-IGF-1R antibody may be administered prior to or subsequent to other therapy, such as radiotherapy, chemotherapy, photodynamic therapy, surgery or other immunotherapy.

[0057] The terms "concurrently" and "simultaneously" are used interchangeably and mean the compounds of the combination therapy of the present invention are administered (1) simultaneously in time, or (2) at different times during the course of a common treatment schedule. The term "sequentially" as used herein means administration of the a first component, followed by administration of a second component. Anti-IGF-1R antibodies may be the first component or the second component. After administration of one component, the second component can be administered substantially immediately after the first component, or the second component can be administered an effective time period after the first component; the effective time period is the amount of time given for realization of maximum benefit from the administration of the first component.

[0058] The term "patient" includes mammals. In a preferred embodiment, the mammal is a human.

[0059] The term "treating," as used herein, unless otherwise indicated, means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term "treatment," as used herein, unless otherwise indicated, refers to the act of treating as "treating" is defined immediately above.

[0060] Human antibodies avoid certain of the problems associated with antibodies that possess mouse or rat variable and/or constant regions. More preferred are fully human anti-human IGF-1R antibodies. Fully human anti-IGF-1R antibodies are expected to minimize the immunogenic and allergic responses intrinsic to mouse or mouse-derivatized monoclonal antibodies (Mabs) and thus to increase the efficacy and safety of the administered antibodies. The use of fully human antibodies can be expected to provide a substantial advantage in the treatment of chronic and recurring human diseases, such as inflammation and cancer, which may require repeated antibody administrations. In another embodiment, the invention provides an anti-IGF-1R antibody that does not bind complement.

[0061] In another aspect of the invention, the anti-IGF-1R antibodies bind to IGF-1R with high affinity. In one embodiment, the anti-IGF-1R antibody binds to IGF-1R with a K.sub.d of 1.times.10.sup.-8 M or less. In a more preferred embodiment, the antibody binds to IGF-1R with a K.sub.d or 1.times.10.sup.-9 M or less. In an even more preferred embodiment, the antibody binds to IGF-1R with a K.sub.d or 5.times.10.sup.-10 M or less. In another preferred embodiment, the antibody binds to IGF-1R with a K.sub.d or 1.times.10.sup.-10 M or less. In another preferred embodiment, the antibody binds to IGF-1R with substantially the same K.sub.d as an antibody selected from 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another preferred embodiment, the antibody binds to IGF-1R with substantially the same K.sub.d as an antibody that comprises one or more CDRs from an antibody selected from 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1.

[0062] The invention also employs an anti-IGF-1R antibody that binds the same antigen or epitope as a human anti-IGF-1R antibody. The invention may also employ an anti-IGF-1R antibody that cross-competes with a human anti-IGF-1R antibody. In a preferred embodiment, the human anti-IGF-1R antibody is 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another preferred embodiment, the human anti-IGF-1R comprises one or more CDRs from an antibody selected from 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1

[0063] The invention can also be practiced using an anti-IGF-1R antibody that comprises variable sequences encoded by a human .kappa. gene. In a preferred embodiment, the variable sequences are encoded by either the V.kappa. A27, A30 or O12 gene family. In a preferred embodiment, the variable sequences are encoded by a human V.kappa. A30 gene family. In a more preferred embodiment, the light chain comprises no more than ten amino acid substitutions from the germline V.kappa. A27, A30 or O12, preferably no more than six amino acid substitutions, and more preferably no more than three amino acid substitutions. In a preferred embodiment, the amino acid substitutions are conservative substitutions.

[0064] In a preferred embodiment, the VL of the anti-IGF-1R antibody contains the same amino acid substitutions, relative to the germline amino acid sequence, as any one or more of the VL of antibodies 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1.

[0065] In another preferred embodiment, the light chain comprises an amino acid sequence that is the same as the amino acid sequence of the VL of 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another highly preferred embodiment, the light chain comprises amino acid sequences that are the same as the CDR regions of the light chain of 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another preferred embodiment, the light chain comprises an amino acid sequence from at least one CDR region of the light chain of 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1.

[0066] The present invention can also be carried out using an anti-IGF-1R antibody or portion thereof comprising a human heavy chain or a sequence derived from a human heavy chain. In one embodiment, the heavy chain amino acid sequence is derived from a human V.sub.H DP-35, DP-47, DP-70, DP-71 or VIV-4/4.35 gene family. In a preferred embodiment, the heavy chain amino acid sequence is derived from a human V.sub.H DP-47 gene family. In a more preferred embodiment, the heavy chain comprises no more than eight amino acid changes from germline V.sub.H DP-35, DP-47, DP-70, DP-71 or VIV-4/4.35, more preferably no more than six amino acid changes, and even more preferably no more than three amino acid changes.

[0067] In a preferred embodiment, the VH of the anti-IGF-1R antibody contains the same amino acid substitutions, relative to the germline amino acid sequence, as any one or more of the VH of antibodies 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another embodiment, the amino acid substitutions are made in the same position as those found in any one or more of the VH of antibodies 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.17.3., 4.9.2 or 6.1.1, but conservative amino acid substitutions are made rather than using the same amino acid.

[0068] In another preferred embodiment, the heavy chain comprises an amino acid sequence that is the same as the amino acid sequence of the VH of 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another highly preferred embodiment, the heavy chain comprises amino acid sequences that are the same as the CDR regions of the heavy chain of 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another preferred embodiment, the heavy chain comprises an amino acid sequence from at least one CDR region of the heavy chain of 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1. In another preferred embodiment, the heavy chain comprises amino acid sequences from CDRs from different heavy chains. In a more preferred embodiment, the CDRs from different heavy chains are obtained from 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 or 6.1.1.

[0069] In another embodiment, the invention employs an anti-IGF-1R antibody that inhibits the binding of IGF-1 to IGF-1R or the binding of IGF-2 to IGF-1R. In a preferred embodiment, the IGF-1R is human. In another preferred embodiment, the anti-IGF-1R antibody is a human antibody. In another embodiment, the antibody or portion thereof inhibits binding between IGF-1R and IGF-1 with an IC.sub.50 of no more than 100 nM. In a preferred embodiment, the IC.sub.50 is no more than 10 nM. In a more preferred embodiment, the IC.sub.50 is no more than 5 nM. The IC.sub.50 can be measured by any method known in the art. Typically, an IC.sub.50 can be measured by ELISA or RIA. In a preferred embodiment, the IC.sub.50 is measured by RIA.

[0070] In another embodiment, the invention employs an anti-IGF-1R antibody that prevents activation of the IGF-1R in the presence of IGF-i. In another aspect of the invention, the antibody causes the downregulation of IGF-1R from a cell treated with the antibody. In a preferred embodiment, the antibody is selected 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, or 6.1.1, or comprises a heavy chain, light chain or antigen-binding region thereof.

[0071] Human antibodies can be produced by immunizing a non-human animal comprising of some or all of the human immunoglobulin locus with an IGF-1R antigen. In a preferred embodiment, the non-human animal is a XENOMOUSE.TM., which is an engineered mouse strain that comprises large fragments of the human immunoglobulin loci and is deficient in mouse antibody production. See, e.g., Green et al. Nature Genetics 7:13-21 (1994) and U.S. Pat. Nos. 5,916,771, 5,939,598, 5,985,615, 5,998,209, 6,075,181, 6,091,001, 6,114,598, and 6,130,364. See also International Patent Application Nos. WO 91/10741, published Jul. 25, 1991; WO 94/02602, published Feb. 3, 1994; WO 96/34096 and WO 96/33735, both published Oct. 31, 1996; WO 98/16654, published Apr. 23, 1998; WO 98/24893, published Jun. 11, 1998; WO 98/50433, published Nov. 12, 1998; WO 99/45031, published Sep. 10, 1999; WO 99/53049, published Oct. 21, 1999; WO 00/09560, published Feb. 24, 2000; and WO 00/037504, published Jun. 29, 2000. The XENOMOUSE.TM. produces an adult-like human repertoire of fully human antibodies, and generates antigen-specific human monoclonal antibodies. A second generation XENOMOUSE.TM. contains approximately 80% of the human antibody repertoire through introduction of megabase sized, germline configuration YAC fragments of the human heavy chain loci and .kappa. light chain loci. See Mendez et al. Nature Genetics 15:146-156 (1997), Green and Jakobovits J. Exp. Med. 188:483-495 (1998).

[0072] The IGF-1R antigen can be administered with an adjuvant to stimulate the immune response. Such adjuvants include complete or incomplete Freund's adjuvant, RIBI (muramyl dipeptides) or ISCOM (immunostimulating complexes). Such adjuvants may protect the polypeptide from rapid dispersal by sequestering it in a local deposit, or they may contain substances that stimulate the host to secrete factors that are chemotactic for macrophages and other components of the immune system.

[0073] The nucleic acid molecule encoding the variable region of the light chain may be derived from the A30, A27 or O12 V.kappa. gene. In a preferred embodiment, the light chain is derived from the A30 V.kappa. gene. In an even more preferred embodiment, the nucleic acid molecule encoding the light chain contains no more than ten amino acid changes from the germline A30 V.kappa. gene, preferably no more than six amino acid changes, and even more preferably no more than three amino acid changes.

[0074] In one embodiment, the antibody contains no greater than ten amino acid changes in either the VH or VL regions of the mutated anti-IGF-1R antibody compared to the anti-IGF-1R antibody prior to mutation. In a more preferred embodiment, there are no more than five amino acid changes in either the VH or VL regions of the mutated anti-IGF-1R antibody, more preferably no more than three amino acid changes. In another embodiment, there are no more than fifteen amino acid changes in the constant domains, more preferably, no more than ten amino acid changes, even more preferably, no more than five amino acid changes.

[0075] SEQ ID NOS: 2, 6, 10, 14, 18 and 22 provide the amino acid sequences of the variable regions of six anti-IGF-1R K light chains. SEQ ID NOS: 4, 8, 12, 16, 20 and 24 provide the amino acid sequences of the variable regions of six anti-IGF-1R heavy chains. SEQ ID NO: 26 depicts the amino acid sequence and SEQ ID NO: 25 depicts the nucleic acid sequence encoding the constant region of the light chain of the anti-IGF-1R antibodies 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 and 6.1.1. SEQ ID NO: 28 depicts the amino acid sequence and SEQ ID NO: 27 depicts the nucleic acid sequence encoding the constant region of the heavy chain of the anti-IGF-1R antibodies 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, 4.17.3 and 6.1.1. SEQ ID NOS: 30, 32, 34, 36 and 44 provide the amino acid sequences of the germline heavy chains DP-35, DP-47, DP-70, DP-71 and VIV-4, respectively. SEQ ID NO: 33 provides the nucleotide sequence of the germline heavy chain DP-70. SEQ ID NOS: 38, 40 and 42 provide the amino acid sequences of the three germline K light chains from which the six anti-IGF-1R K light chains are derived.

[0076] The anti-IGF-1R antibodies can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the pharmaceutical composition comprises an antibody and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion, may also be included.

[0077] The pharmaceutical compositions may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The preferred form depends on the intended mode of administration and therapeutic application. Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. A preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular, or infusion). In a preferred embodiment, the antibody is administered by intravenous infusion or injection. In another preferred embodiment, the antibody is administered by intramuscular or subcutaneous injection. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.

[0078] Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating the anti-IGF-1R antibody in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.

[0079] In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.

[0080] The pharmaceutical composition may include a "therapeutically effective amount" or a "prophylactically effective amount" of an antibody or antibody portion of the invention. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.

[0081] Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. Pharmaceutical composition comprising the antibody or comprising a combination therapy comprising the antibody and one or more additional therapeutic agents may be formulated for single or multiple doses. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. A particularly useful formulation is 5 mg/ml anti-IGF-1R antibody in a buffer of 20 mM sodium citrate, pH 5.5, 140 mM NaCl, and 0.2 mg/ml polysorbate 80.

[0082] The antibody, with or without an additional agent, may be administered once, or more than once for at least the period of time until the condition is treated, palliated or cured. The antibody generally will be administered for as long as the tumor is present provided that the antibody causes the tumor or cancer to stop growing or to decrease in weight or volume. The antibody will generally be administered as part of a pharmaceutical composition as described supra. The dosage of antibody will generally be in the range of 0.025-100 mg/kg, more preferably 0.05-50 mg/kg, more preferably 0.05-20 mg/kg, and even more preferably 0.1-10 mg/kg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.

[0083] The antibody may be administered from three times daily to once every six months. The administration may be on a schedule such as three times daily, twice daily, once daily, once every two days, once every three days, once weekly, once every two weeks, once every month, once every two months, once every three months and once every six months. The antibody may be administered via an oral, mucosal, buccal, intranasal, inhalable, intravenous, subcutaneous, intramuscular, parenteral, intratumor or topical route.

[0084] The antibody may be administered at a site distant from the site of the tumor. The antibody may also be administered continuously via a minipump.

[0085] In certain embodiments, the antibody may be administered in an aerosol or inhalable form. Dry aerosol in the form of finely divided solid particles that are not dissolved or suspended in a liquid are also useful in the practice of the present invention. The pharmaceutical formulations of the present invention may be administered in the form of an aerosol spray using for example, a nebulizer such as those described in U.S. Pat. Nos. 4,624,251; 3,703,173; 3,561,444; and 4,635,627.

[0086] The serum concentration of the antibody may be measured by any method known in the art. The antibody may also be administered prophylactically in order to prevent a cancer or tumor from occurring. This may be especially useful in patients that have a "high normal" level of IGF-1 because these patients have been shown to have a higher risk of developing common cancers. See Rosen et al., supra.

[0087] The antibody employed in the method of the invention can be labeled. This can be done by incorporation of a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). In certain situations, the label or marker can also be therapeutic. Various methods of labeling polypeptides and glycoproteins are known in the art and may be used. Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., .sup.3H, .sup.14C, .sup.15N, .sup.35S, .sup.90Y, .sup.99Tc, .sup.111In, .sup.125I, .sup.131I), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, .beta.-galactosidase, luciferase, alkaline phosphatase), chemiluminescent, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags). In some embodiments, labels are attached by spacer arms of various lengths to reduce potential steric hindrance.

[0088] The antibodies employed in the present invention are preferably derived from cells that express human immunoglobulin genes. Use of transgenic mice is known in the art to produce such "human" antibodies. One such method is described in U.S. Patent Application Ser. No. 08/759,620, filed Dec. 3, 1996. See also Mendez et al. Nature Genetics 15:146-156 (1997); Green and Jakobovits J. Exp. Med. 188:483-495 (1998); European Patent No. EP 0 463 151 (grant published Jun. 12, 1996); and International Patent Application Nos. WO 94/02602, published Feb. 3, 1994; WO 96/34096, published Oct. 31, 1996; and WO 98/24893, published Jun. 11, 1998.

[0089] As noted above, the invention encompasses use of antibody fragments. Antibody fragments, such as Fv, F(ab').sub.2 and Fab may be prepared by cleavage of the intact protein, e.g. by protease or chemical cleavage. Alternatively, a truncated gene is designed. For example, a chimeric gene encoding a portion of the F(ab').sub.2 fragment would include DNA sequences encoding the CH1 domain and hinge region of the H chain, followed by a translational stop codon to yield the truncated molecule.

[0090] In one approach, consensus sequences encoding the heavy and light chain J regions may be used to design oligonucleotides for use as primers to introduce useful restriction sites into the J region for subsequent linkage of V region segments to human C region segments. C region cDNA can be modified by site directed mutagenesis to place a restriction site at the analogous position in the human sequence.

[0091] Expression vectors for use in obtaining the antibodies employed in the invention include plasmids, retroviruses, cosmids, YACs, EBV derived episomes, and the like. A convenient vector is normally one that encodes a functionally complete human CH or CL immunoglobulin sequence, with appropriate restriction sites engineered so that any VH or VL sequence can be easily inserted and expressed. In such vectors, splicing usually occurs between the splice donor site in the inserted J region and the splice acceptor site preceding the human C region, and also at the splice regions that occur within the human CH exons. Polyadenylation and transcription termination occur at native chromosomal sites downstream of the coding regions. The resulting chimeric antibody may be joined to any strong promoter, including retroviral LTRs, e.g. SV-40 early promoter (Okayama et al. Mol. Cell. Bio. 3:280 (1983)), Rous sarcoma virus LTR (Gorman et al. Proc. Natl. Acad. Sci. 79:6777 (1982)), and moloney murine leukemia virus LTR (Grosschedl et al. Cell 41:885 (1985)); native Ig promoters, etc.

[0092] Antibodies that are generated for use in the invention need not initially possess a particular desired isotype. Rather, the antibody as generated can possess any isotype and can be isotype switched thereafter using conventional techniques. These include direct recombinant techniques (see e.g., U.S. Pat. No. 4,816,397), and cell-cell fusion techniques (see e.g., U.S. Pat. No. 5,916,771).

[0093] As noted above, the effector function of the antibodies of the invention may be changed by isotype switching to an IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM for various therapeutic uses. Furthermore, dependence on complement for cell killing can be avoided through the use of bispecifics, immunotoxins, or radiolabels, for example.

[0094] Bispecific antibodies can be generated that comprise (i) two antibodies: one with a specificity for IGF-1R and the other for a second molecule (ii) a single antibody that has one chain specific for IGF-1R and a second chain specific for a second molecule, or (iii) a single chain antibody that has specificity for IGF-1R and the other molecule. Such bispecific antibodies can be generated using well known techniques, e.g., Fanger et al. Immunol. Methods 4:72-81 (1994); Wright and Harris, supra; and Traunecker et al. Int. J. Cancer (Suppl.) 7:51-52 (1992).

[0095] Antibodies for use in the invention also include "kappabodies" (Ill et al. Protein Eng. 10:949-57 (1997)), "minibodies" (Martin et al. EMBO J. 13:5303-9 (1994)), "diabodies" (Holliger et al. Proc. Natl. Acad. Sci. (USA) 90:6444-6448 (1993)), and "janusins" (Traunecker et al. EMBO J. 10:3655-3659 (1991) and Traunecker et al. Int. J. Cancer Suppl. 7:51-52 (1992)) may also be prepared.

[0096] The antibodies employed can be modified to act as immunotoxins by conventional techniques. See e.g., Vitetta Immunol. Today 14:252 (1993). See also U.S. Pat. No. 5,194,594. Radiolabeled antibodies can also be prepared using well-known techniques. See e.g., Junghans et al. in Cancer Chemotherapy and Biotherapy 655-686 (2d edition, Chafner and Longo, eds., Lippincott Raven (1996)). See also U.S. Pat. Nos. 4,681,581, 4,735,210, 5,101,827, 5,102,990 (Re. 35,500), 5,648,471, and 5,697,902.

[0097] Particular antibodies useful in practice of the invention include those described in International Patent Application No. WO 02/053596, which further describes antibodies 2.12.1, 2.13.2., 2.14.3, 3.1.1, 4.9.2, and 4.17.3. As disclosed in that published application, hybridomas producing these antibodies were deposited in the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, Va. 20110-2209, on Dec. 12, 2000 with the following deposit numbers:

TABLE-US-00001 Hybridoma Deposit No. 2.12.1 PTA-2792 2.13.2 PTA-2788 2.14.3 PTA-2790 3.1.1 PTA-2791 4.9.2 PTA-2789 4.17.3 PTA-2793

[0098] These antibodies are either fully human IgG2 or IgG4 heavy chains with human kappa light chains. In particular the invention concerns use of antibodies having amino acid sequences of these antibodies.

[0099] Antibodies employed in the invention preferably possess very high affinities, typically possessing Kds of from about 10.sup.-9 through about 10.sup.-11 M, when measured by either solid phase or solution phase.

[0100] Antibodies used in the present invention can be expressed in cell lines other than hybridoma cell lines. Sequences encoding the cDNAs or genomic clones for the particular antibodies can be used for transformation of suitable mammalian or nonmammalian host cells. Transformation can be by any known method for introducing polynucleotides into a host cell, including, for example packaging the polynucleotide in a virus (or into a viral vector) and transducing a host cell with the virus (or vector) or by transfection procedures known in the art, as exemplified by U.S. Pat. Nos. 4,399,216, 4,912,040, 4,740,461, and 4,959,455. Methods for introduction of heterologous polynucleotides into mammalian cells are well known in the art and include, but are not limited to, dextran-mediated transfection, calcium phosphate precipitation, polybrene mediated transfection, protoplast fusion, electroporation, particle bombardment, encapsulation of the polynucleotide(s) in liposomes, peptide conjugates, dendrimers, and direct microinjection of the DNA into nuclei.

[0101] Mammalian cell lines available as hosts for expression are well known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC), including but not limited to Chinese hamster ovary (CHO) cells, NSO.sub.0, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), and human hepatocellular carcinoma cells (e.g., Hep G2). Non-mammalian cells can also be employed, including bacterial, yeast, insect, and plant cells. Site directed mutagenesis of the antibody CH2 domain to eliminate glycosylation may be preferred in order to prevent changes in either the immunogenicity, pharmacokinetic, and/or effector functions resulting from non-human glycosylation. The glutamine synthase system of expression is discussed in whole or part in connection with European Patent Nos. 0 216 846, 0 256 055, and 0 323 997, and European Patent Application No. 89303964.4.

[0102] Antibodies for use in the invention can also be produced transgenically through the generation of a mammal or plant that is transgenic for the immunoglobulin heavy and light chain sequences of interest and production of the antibody in a recoverable form therefrom. Transgenic antibodies can be produced in, and recovered from, the milk of goats, cows, or other mammals. See, e.g., U.S. Pat. Nos. 5,827,690, 5,756,687, 5,750,172, and 5,741,957.

[0103] The advantages of the present invention can be further appreciated by reference to the following examples. These examples serve intended to illustrate preferred embodiments of the invention and are by no means intended to limit the effective scope of the claims.

Example I

Anti-IGF-1R Antibodies in Combination with Docetaxel in the Treatment of Advanced Non-Hematologic Malignancies

[0104] Patients with advanced-stage non-hematologic malignancies (measurable disease defined by at least one lesion that can be accurately measured and whose size is .gtoreq.2 cm.times.1 cm by conventional computed tomography (CT) scan or .gtoreq.1 cm.times.1 cm by spiral CT scan) received a standard dose of docetaxel (TAXOTERE) (up to 75 mg/m.sup.2, using actual body weight to calculate body surface area (BSA)) by intravenous (IV) infusion over 1 hour on Day 1 only of each cycle. After the docetaxel infusion was completed, anti-IGF-1R antibodies as described herein were administered intravenously in a 5 mg/ml liquid formulation at a dose between 0.1 mg/kg and 10 mg/kg. The treatment regimen was repeated after 21 days, with escalation of the anti-IGF-1R antibody dose, and every 21 days thereafter until disease progression or unacceptable toxicity develops for a minimum of 2 cycles and a maximum of 17 cycles. The pre-medication regimen for docetaxel included oral dexamethasone 8 mg twice daily for three days starting one day prior to docetaxel administration. Prophylactic anti-emetics were provided as appropriate.

[0105] Dose escalation used an accelerated titration design utilizing a dose-doubling schema with 3-6 subjects per dose level (cohort). Within each new cohort there was no required waiting period between subjects. Subsequent cohorts were not opened until the first subject at the current dose level had been observed for 21 days and subsequent subjects had been observed for 14 days.

[0106] The following endpoints were measured: safety, tolerability, pharmacokinetic (PK) parameters of the anti-IGF-1R antibody; human anti-human antibody response (HAHA); response rate and time to progression; and number of circulating tumor cells (CTC) and circulating soluble IGF-1R.

Example II

Anti-IGF-1R Antibodies in Combination with Paclitaxel and Carboplatin in the Treatment of Advanced Non-Small Cell Lung Cancer

[0107] In Part 1 of the study, patients with Stage IIIB or Stage IV or recurrent (after surgery/radiation), measurable, non-small cell lung cancer (NSCLC) who have received no prior chemotherapy received paclitaxel (TAXOL) at a standard dose of 200 mg/m.sup.2 by IV infusion over 3 hours. Prior to receiving paclitaxel, all patients received prophylactic anti-allergic/emetic medicines. Carboplatin (PARAPLATIN) was administered by IV infusion over 15-30 minutes; the dose was calculated based on the Calvert formula with a target area under the curve (AUC) of 6 mg/ml.times.min. After the carboplatin infusion was completed, anti-IGF-1R antibodies as described herein were administered intravenously in a 5 mg/ml formulation at a dose between 0.05 mg/kg and 10 mg/kg. The treatment regimen was repeated after 21 days, with escalation of the anti-IGF-1R antibody dose, and every 21 days thereafter until disease progression or unacceptable toxicity develops, for a minimum of 1 cycle and a maximum of 6 cycles.

[0108] Doses were escalated using an accelerated titration design utilizing a dose-doubling schema with 3-6 subjects per cohort. Within each new cohort there was no required waiting period between subjects. Subsequent cohorts were not opened until the first subject at the current dose level has been observed for 21 days and subsequent subjects have been observed for 14 days.

[0109] Once at least six patients have been observed for 21 days (i.e., completed one cycle), the randomized second portion of the study will begin.

[0110] Part 2 of the study is a two-arm randomized, non-comparative study of anti-IGF-1R antibody in combination with paclitaxel and carboplatin (Arm A) and of paclitaxel and carboplatin alone (Arm B). On Day 1 of Part 2, the patients in both arms are given the same dosages of paclitaxel and carboplatin, over the same time periods, as in the first part. After administration of carboplatin, patients in Arm A are also given the same anti-IGF-1R antibody dose they were given in Part 1. The dose is determined in view of the safety and tolerability demonstrated in Part 1. The treatment is repeated after 21 days, and every 21 days thereafter, until progression or unacceptable toxicity occurs for a minimum of 2 cycles and a maximum of 6.

[0111] The following endpoints are measured: PK parameters of the anti-IGF-1R antibody, HAHA, response rate and time to progression, CTC, circulating IGF-1, IGFBPs, and soluble circulating IGF-1R.

Example III

Anti-IGF-1R in Combination with Docetaxel and Epirubicin in Metastatic Breast Cancer

[0112] Patients having metastatic breast cancer with at least one lesion that can be accurately measured in two dimensions and whose size is 2 cm.times.1 cm by conventional CT scan or 1 cm.times.1 cm by spiral CT scan are given epirubicin 75 mg/m.sup.2 as a single 15 minute infusion. After a one hour pause, docetaxel (TAXOTERE) 75 mg/m.sup.2 is administered as a single IV infusion, followed by IV infusion of anti-IGF-1R antibodies as described herein at a dose between 0.05 mg/kg and 10 mg/kg. Prophylactic anti-emetics are given as appropriate. The treatment is repeated after 21 days with escalation of the anti-IGF-1R antibody dose, and every 21 days thereafter until disease progression or unacceptable toxicity develops for a minimum of 2 cycles and a maximum of 6.

[0113] Doses are escalated using an accelerated titration design utilizing a dose-doubling schema with 3-6 subjects per cohort. Within each new cohort there is no required waiting period between subjects. Subsequent cohorts may not be opened until the first subject at the current dose level has been observed for 21 days and subsequent subjects have been observed for 14 days.

[0114] The following endpoints are measured: PK parameters, HAHA, response rate and time to progression. Time to progression and overall survival are calculated using the Kaplan-Meier product limit method.

Example IV

Anti-IGF-1R in Combination with Docetaxel and Prednisone in Hormone-Refractory Prostate Cancer

[0115] Subjects are patients with metastatic adenocarcinoma of the prostate who, after at least one hormonal treatment (orchiectomy, estrogens, LHRH therapy, etc.), have testosterone levels less than 50 ng/dL, prostate-specific antigen (PSA) above 20 ng/mL, and an increase in PSA >50% over nadir value on hormonal therapy measured on 3 successive occasions at least 1 week apart. A pre-medication regimen for docetaxel includes oral dexamethasone 8 mg twice a day given for 3 days starting one day prior to docetaxel administration. A 75 mg/m.sup.2 dose of docetaxel (TAXOTERE) (using actual body weight to calculate BSA) is administered by IV infusion over 1 hour on Day 1 only of each cycle. After the docetaxel infusion is completed, anti-IGF-1R antibodies as described herein are administered intravenously in a 5 mg/ml liquid formulation. Prednisone is given daily in two oral 5 mg doses per day, starting on Day 1. Prophylactic anti-emetics may be given as appropriate. The treatment regimen is repeated every 21 days (.+-.3 days) until disease progression or unacceptable toxicity develops, for a maximum of 10 cycles.

[0116] The following endpoints are measured: PSA response, population PK parameters of the anti-IGF-1R antibody, HAHA, total number of CTCs and CTCs expressing IGF-1R.

[0117] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Sequence CWU 1

1

601291DNAHomo sapiens 1tgcatctgta ggagacagag tcaccttcac ttgccgggca agtcaggaca ttagacgtga 60tttaggctgg tatcagcaga aaccagggaa agctcctaag cgcctgatct atgctgcatc 120ccgtttacaa agtggggtcc catcaaggtt cagcggcagt ggatctggga cagaattcac 180tctcacaatc agcagcctgc agcctgaaga ttttgcaact tattactgtc tacagcataa 240taattatcct cggacgttcg gccaagggac cgaggtggaa atcatacgaa c 2912136PRTHomo sapiens 2Ala Ser Val Gly Asp Arg Val Thr Phe Thr Cys Arg Ala Ser Gln Asp1 5 10 15Ile Arg Arg Asp Leu Gly Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro 20 25 30Lys Arg Leu Ile Tyr Ala Ala Ser Arg Leu Gln Ser Gly Val Pro Ser 35 40 45Arg Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser 50 55 60Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln His Asn65 70 75 80Asn Tyr Pro Arg Thr Phe Gly Gln Gly Thr Glu Val Glu Ile Ile Arg 85 90 95Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln 100 105 110Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr 115 120 125Pro Arg Glu Ala Lys Val Gln Trp 130 1353352DNAHomo sapiens 3gggaggcttg gtcaagcctg gaggtccctg agactctcct gtgcagcctc tggattcact 60ttcagtgact actatatgag ctggatccgc caggctccag ggaaggggct ggaatgggtt 120tcatacatta gtagtagtgg tagtaccaga gactacgcag actctgtgaa gggccgattc 180accatctcca gggacaacgc caagaactca ctgtatctgc aaatgaacag cctgagagcc 240gaggacacgg ccgtgtatta ctgtgtgaga gatggagtgg aaactacttt ttactactac 300tactacggta tggacgtctg gggccaaggg accacggtca ccgtctcctc ag 3524174PRTHomo sapiens 4Gly Arg Leu Gly Gln Ala Trp Arg Ser Leu Arg Leu Ser Cys Ala Ala1 5 10 15Ser Gly Phe Thr Phe Ser Asp Tyr Tyr Met Ser Trp Ile Arg Gln Ala 20 25 30Pro Gly Lys Gly Leu Glu Trp Val Ser Tyr Ile Ser Ser Ser Gly Ser 35 40 45Thr Arg Asp Tyr Ala Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg 50 55 60Asp Asn Ala Lys Asn Ser Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala65 70 75 80Glu Asp Thr Ala Val Tyr Tyr Cys Val Arg Asp Gly Val Glu Thr Thr 85 90 95Phe Tyr Tyr Tyr Tyr Tyr Gly Met Asp Val Trp Gly Gln Gly Thr Thr 100 105 110Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu 115 120 125Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys 130 135 140Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser145 150 155 160Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ser Cys Ala 165 1705322DNAHomo sapiens 5gacatccaga tgacccagtt tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60atcacttgcc gggcaagtca gggcattaga aatgatttag gctggtatca gcagaaacca 120gggaaagccc ctaagcgcct gatctatgct gcatcccgtt tgcacagagg ggtcccatca 180aggttcagcg gcagtggatc tgggacagaa ttcactctca caatcagcag cctgcagcct 240gaagattttg caacttatta ctgtttacaa cataatagtt acccgtgcag ttttggccag 300gggaccaagc tggagatcaa ac 3226107PRTHomo sapiens 6Asp Ile Gln Met Thr Gln Phe Pro Ser Ser Leu Ser Ala Ser Val Gly1 5 10 15Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Gly Ile Arg Asn Asp 20 25 30Leu Gly Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Arg Leu Ile 35 40 45Tyr Ala Ala Ser Arg Leu His Arg Gly Val Pro Ser Arg Phe Ser Gly 50 55 60Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro65 70 75 80Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln His Asn Ser Tyr Pro Cys 85 90 95Ser Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys 100 1057375DNAHomo sapiens 7aggtgcagct gttggagtct gggggaggct tggtacagcc tggggggtcc ctgagactct 60cctgtacagc ctctggattc acctttagca gctatgccat gaactgggtc cgccaggctc 120cagggaaggg gctggagtgg gtctcagcta ttagtggtag tggtggtacc acattctacg 180cagactccgt gaagggccgg ttcaccatct ccagagacaa ttccaggacc acgctgtatc 240tgcaaatgaa cagcctgaga gccgaggaca cggccgtata ttactgtgcg aaagatcttg 300gctggtccga ctcttactac tactactacg gtatggacgt ctggggccaa gggaccacgg 360tcaccgtctc ctcag 3758124PRTHomo sapiens 8Val Gln Leu Leu Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly Ser1 5 10 15Leu Arg Leu Ser Cys Thr Ala Ser Gly Phe Thr Phe Ser Ser Tyr Ala 20 25 30Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val Ser 35 40 45Ala Ile Ser Gly Ser Gly Gly Thr Thr Phe Tyr Ala Asp Ser Val Lys 50 55 60Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Arg Thr Thr Leu Tyr Leu65 70 75 80Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys Ala 85 90 95Lys Asp Leu Gly Trp Ser Asp Ser Tyr Tyr Tyr Tyr Tyr Gly Met Asp 100 105 110Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser 115 1209302DNAHomo sapiens 9tcctccctgt ctgcatctgt aggagacaga gtcaccttca cttgccgggc aagtcaggac 60attagacgtg atttaggctg gtatcagcag aaaccaggga aagctcctaa gcgcctgatc 120tatgctgcat cccgtttaca aagtggggtc ccatcaaggt tcagcggcag tggatctggg 180acagaattca ctctcacaat cagcagcctg cagcctgaag attttgcaac ttattactgt 240ctacagcata ataattatcc tcggacgttc ggccaaggga ccgaggtgga aatcatacga 300ac 30210100PRTHomo sapiens 10Ser Ser Leu Ser Ala Ser Val Gly Asp Arg Val Thr Phe Thr Cys Arg1 5 10 15Ala Ser Gln Asp Ile Arg Arg Asp Leu Gly Trp Tyr Gln Gln Lys Pro 20 25 30Gly Lys Ala Pro Lys Arg Leu Ile Tyr Ala Ala Ser Arg Leu Gln Ser 35 40 45Gly Val Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr 50 55 60Leu Thr Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys65 70 75 80Leu Gln His Asn Asn Tyr Pro Arg Thr Phe Gly Gln Gly Thr Glu Val 85 90 95Glu Ile Ile Arg 10011338DNAHomo sapiens 11gggcccagga ctggtgaagc cttcggagac cctgtccctc acctgcactg tctctggtgg 60ctccatcagt aattactact ggagctggat ccggcagccc gccgggaagg gactggagtg 120gattgggcgt atctatacca gtgggagccc caactacaac ccctccctca agagtcgagt 180caccatgtca gtagacacgt ccaagaacca gttctccctg aagctgaact ctgtgaccgc 240cgcggacacg gccgtgtatt actgtgcggt aacgattttt ggagtggtta ttatctttga 300ctactggggc cagggaaccc tggtcaccgt ctcctcag 33812112PRTHomo sapiens 12Gly Pro Gly Leu Val Lys Pro Ser Glu Thr Leu Ser Leu Thr Cys Thr1 5 10 15Val Ser Gly Gly Ser Ile Ser Asn Tyr Tyr Trp Ser Trp Ile Arg Gln 20 25 30Pro Ala Gly Lys Gly Leu Glu Trp Ile Gly Arg Ile Tyr Thr Ser Gly 35 40 45Ser Pro Asn Tyr Asn Pro Ser Leu Lys Ser Arg Val Thr Met Ser Val 50 55 60Asp Thr Ser Lys Asn Gln Phe Ser Leu Lys Leu Asn Ser Val Thr Ala65 70 75 80Ala Asp Thr Ala Val Tyr Tyr Cys Ala Val Thr Ile Phe Gly Val Val 85 90 95Ile Ile Phe Asp Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ser 100 105 11013322DNAHomo sapiens 13gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60atcacttgcc gggcaagtca gggcattaga agtgatttag gctggtttca gcagaaacca 120gggaaagccc ctaagcgcct gatctatgct gcatccaaat tacaccgtgg ggtcccatca 180aggttcagcg gcagtggatc tgggacagaa ttcactctca caatcagccg cctgcagcct 240gaagattttg caacttatta ctgtctacag cataatagtt accctctcac tttcggcgga 300gggaccaagg tggagatcaa ac 32214107PRTHomo sapiens 14Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly1 5 10 15Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Gly Ile Arg Ser Asp 20 25 30Leu Gly Trp Phe Gln Gln Lys Pro Gly Lys Ala Pro Lys Arg Leu Ile 35 40 45Tyr Ala Ala Ser Lys Leu His Arg Gly Val Pro Ser Arg Phe Ser Gly 50 55 60Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Arg Leu Gln Pro65 70 75 80Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln His Asn Ser Tyr Pro Leu 85 90 95Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys 100 10515376DNAHomo sapiens 15gaggtgcagc tgttggagtc tgggggaggc ttggtacagc ctggggggtc cctgagactc 60tcctgtgcag cctctggatt cacctttagc agctatgcca tgagctgggt ccgccaggct 120ccagggaagg ggctggagtg ggtctcagct attagtggta gtggtggtat cacatactac 180gcagactccg tgaagggccg gttcaccatc tccagagaca attccaagaa cacgctgtat 240ctgcaaatga acagcctgag agccgaggac acggccgtat attactgtgc gaaagatctg 300ggctacggtg acttttacta ctactactac ggtatggacg tctggggcca agggaccacg 360gtcaccgtct cctcag 37616125PRTHomo sapiens 16Glu Val Gln Leu Leu Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly1 5 10 15Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr 20 25 30Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val 35 40 45Ser Ala Ile Ser Gly Ser Gly Gly Ile Thr Tyr Tyr Ala Asp Ser Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr65 70 75 80Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95Ala Lys Asp Leu Gly Tyr Gly Asp Phe Tyr Tyr Tyr Tyr Tyr Gly Met 100 105 110Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser 115 120 12517279DNAHomo sapiens 17caggagacag agtcaccatc acttgccggg caagtcagag cattagtacc tttttaaatt 60ggtatcagca gaaaccaggg aaagccccta aactcctgat ccatgttgca tccagtttac 120aaggtggggt cccatcaagg ttcagtggca gtggatctgg gacagatttc actctcacca 180tcagcagtct gcaacctgaa gattttgcaa cttactactg tcaacagagt tacaatgccc 240cactcacttt cggcggaggg accaaggtgg agatcaaac 2791892PRTHomo sapiens 18Gly Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Ser Thr1 5 10 15Phe Leu Asn Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu 20 25 30Ile His Val Ala Ser Ser Leu Gln Gly Gly Val Pro Ser Arg Phe Ser 35 40 45Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln 50 55 60Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Ser Tyr Asn Ala Pro65 70 75 80Leu Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys 85 9019341DNAHomo sapiens 19cccaggactg gtgaagcctt cggagaccct gtccctcacc tgcactgtct ctggtggctc 60catcagtagt tactactgga gttggatccg gcagccccca gggaagggac tggagtggat 120tgggtatatc tattacagtg ggagcaccaa ctacaacccc tccctcaaga gtcgagtcac 180catatcagta gacacgtcca agaaccagtt ctccctgaag ctgagttctg tgaccgctgc 240ggacacggcc gtgtattact gtgccaggac gtatagcagt tcgttctact actacggtat 300ggacgtctgg ggccaaggga ccacggtcac cgtctcctca g 34120113PRTHomo sapiens 20Pro Gly Leu Val Lys Pro Ser Glu Thr Leu Ser Leu Thr Cys Thr Val1 5 10 15Ser Gly Gly Ser Ile Ser Ser Tyr Tyr Trp Ser Trp Ile Arg Gln Pro 20 25 30Pro Gly Lys Gly Leu Glu Trp Ile Gly Tyr Ile Tyr Tyr Ser Gly Ser 35 40 45Thr Asn Tyr Asn Pro Ser Leu Lys Ser Arg Val Thr Ile Ser Val Asp 50 55 60Thr Ser Lys Asn Gln Phe Ser Leu Lys Leu Ser Ser Val Thr Ala Ala65 70 75 80Asp Thr Ala Val Tyr Tyr Cys Ala Arg Thr Tyr Ser Ser Ser Phe Tyr 85 90 95Tyr Tyr Gly Met Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser 100 105 110Ser21274PRTHomo sapiens 21Ala Gly Ala Gly Cys Cys Ala Cys Cys Cys Thr Cys Thr Cys Cys Thr1 5 10 15Gly Thr Ala Gly Gly Gly Cys Cys Ala Gly Thr Cys Ala Gly Ala Gly 20 25 30Thr Gly Thr Thr Cys Gly Cys Gly Gly Cys Ala Gly Gly Thr Ala Cys 35 40 45Thr Thr Ala Gly Cys Cys Thr Gly Gly Thr Ala Cys Cys Ala Gly Cys 50 55 60Ala Gly Ala Ala Ala Cys Cys Thr Gly Gly Cys Cys Ala Gly Gly Cys65 70 75 80Thr Cys Cys Cys Ala Gly Gly Cys Thr Cys Cys Thr Cys Ala Thr Cys 85 90 95Thr Ala Thr Gly Gly Thr Gly Cys Ala Thr Cys Cys Ala Gly Cys Ala 100 105 110Gly Gly Gly Cys Cys Ala Cys Thr Gly Gly Cys Ala Thr Cys Cys Cys 115 120 125Ala Gly Ala Cys Ala Gly Gly Thr Thr Cys Ala Gly Thr Gly Gly Cys 130 135 140Ala Gly Thr Gly Gly Gly Thr Cys Thr Gly Gly Gly Ala Cys Ala Gly145 150 155 160Ala Cys Thr Thr Cys Ala Cys Thr Cys Thr Cys Ala Cys Cys Ala Thr 165 170 175Cys Ala Gly Cys Ala Gly Ala Cys Thr Gly Gly Ala Gly Cys Cys Thr 180 185 190Gly Ala Ala Gly Ala Thr Thr Thr Thr Gly Cys Ala Gly Thr Gly Thr 195 200 205Thr Thr Thr Ala Cys Thr Gly Thr Cys Ala Gly Cys Ala Gly Thr Ala 210 215 220Thr Gly Gly Thr Ala Gly Thr Thr Cys Ala Cys Cys Thr Cys Gly Asn225 230 235 240Ala Cys Gly Thr Thr Cys Gly Gly Cys Cys Ala Ala Gly Gly Gly Ala 245 250 255Cys Cys Ala Ala Gly Gly Thr Gly Gly Ala Ala Ala Thr Cys Ala Ala 260 265 270Ala Cys2291PRTHomo sapiens 22Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val Arg Gly Arg Tyr1 5 10 15Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile 20 25 30Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser Gly 35 40 45Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu Pro 50 55 60Glu Asp Phe Ala Val Phe Tyr Cys Gln Gln Tyr Gly Ser Ser Pro Arg65 70 75 80Thr Phe Gly Gln Gly Thr Lys Val Glu Ile Lys 85 9023367DNAHomo sapiens 23gaggtgcagc tgttggagtc tgggggaggc ttggtacagc ctggggggtc cctgagactc 60tcctgtgcag cctctggatt cacctttagc agctatgcca tgagctgggt ccgccaggct 120ccagggaagg ggctggagtg ggtctcaggt attactggga gtggtggtag tacatactac 180gcagactccg tgaagggccg gttcaccatc tccagagaca attccaagaa cacgctgtat 240ctgcaaatga acagcctgag agccgaggac acggccgtat attactgtgc gaaagatcca 300gggactacgg tgattatgag ttggttcgac ccctggggcc agggaaccct ggtcaccgtc 360tcctcag 36724122PRTHomo sapiens 24Glu Val Gln Leu Leu Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly1 5 10 15Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr 20 25 30Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val 35 40 45Ser Gly Ile Thr Gly Ser Gly Gly Ser Thr Tyr Tyr Ala Asp Ser Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr65 70 75 80Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95Ala Lys Asp Pro Gly Thr Thr Val Ile Met Ser Trp Phe Asp Pro Trp 100 105 110Gly Gln Gly Thr Leu Val Thr Val Ser Ser 115 12025320DNAHomo sapiens 25gaactgtggc tgcaccatct gtcttcatct tcccgccatc tgatgagcag ttgaaatctg 60gaactgcctc tgttgtgtgc ctgctgaata acttctatcc cagagaggcc aaagtacagt 120ggaaggtgga taacgccctc caatcgggta actcccagga gagtgtcaca gagcaggaca 180gcaaggacag cacctacagc ctcagcagca ccctgacgct gagcaaagca gactacgaga 240aacacaaagt ctacgcctgc gaagtcaccc atcagggcct gagctcgccc gtcacaaaga 300gcttcaacag gggagagtgt 32026106PRTHomo sapiens 26Thr Val Ala Ala Pro Ser Val Phe Ile

Phe Pro Pro Ser Asp Glu Gln1 5 10 15Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr 20 25 30Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser 35 40 45Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr 50 55 60Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys65 70 75 80His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro 85 90 95Val Thr Lys Ser Phe Asn Arg Gly Glu Cys 100 10527978DNAHomo sapiens 27gcctccacca agggcccatc ggtcttcccc ctggcgccct gctccaggag cacctccgag 60agcacagcgg ccctgggctg cctggtcaag gactacttcc ccgaaccggt gacggtgtcg 120tggaactcag gcgctctgac cagcggcgtg cacaccttcc cagctgtcct acagtcctca 180ggactctact ccctcagcag cgtggtgacc gtgccctcca gcaacttcgg cacccagacc 240tacacctgca acgtagatca caagcccagc aacaccaagg tggacaagac agttgagcgc 300aaatgttgtg tcgagtgccc accgtgccca gcaccacctg tggcaggacc gtcagtcttc 360ctcttccccc caaaacccaa ggacaccctc atgatctccc ggacccctga ggtcacgtgc 420gtggtggtgg acgtgagcca cgaagacccc gaggtccagt tcaactggta cgtggacggc 480gtggaggtgc ataatgccaa gacaaagcca cgggaggagc agttcaacag cacgttccgt 540gtggtcagcg tcctcaccgt tgtgcaccag gactggctga acggcaagga gtacaagtgc 600aaggtctcca acaaaggcct cccagccccc atcgagaaaa ccatctccaa aaccaaaggg 660cagccccgag aaccacaggt gtacaccctg cccccatccc gggaggagat gaccaagaac 720caggtcagcc tgacctgcct ggtcaaaggc ttctacccca gcgacatcgc cgtggagtgg 780gagagcaatg ggcagccgga gaacaactac aagaccacac ctcccatgct ggactccgac 840ggctccttct tcctctacag caagctcacc gtggacaaga gcaggtggca gcaggggaac 900gtcttctcat gctccgtgat gcatgaggct ctgcacaacc actacacgca gaagagcctc 960tccctgtctc cgggtaaa 97828326PRTHomo sapiens 28Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys Ser Arg1 5 10 15Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr 20 25 30Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser 35 40 45Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser 50 55 60Leu Ser Ser Val Val Thr Val Pro Ser Ser Asn Phe Gly Thr Gln Thr65 70 75 80Tyr Thr Cys Asn Val Asp His Lys Pro Ser Asn Thr Lys Val Asp Lys 85 90 95Thr Val Glu Arg Lys Cys Cys Val Glu Cys Pro Pro Cys Pro Ala Pro 100 105 110Pro Val Ala Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp 115 120 125Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp 130 135 140Val Ser His Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly145 150 155 160Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn 165 170 175Ser Thr Phe Arg Val Val Ser Val Leu Thr Val Val His Gln Asp Trp 180 185 190Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro 195 200 205Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr Lys Gly Gln Pro Arg Glu 210 215 220Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn225 230 235 240Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile 245 250 255Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr 260 265 270Thr Pro Pro Met Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys 275 280 285Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys 290 295 300Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu305 310 315 320Ser Leu Ser Pro Gly Lys 32529296DNAHomo sapiens 29caggtgcagc tggtggagtc tgggggaggc ttggtcaagc ctggagggtc cctgagactc 60tcctgtgcag cctctggatt caccttcagt gactactaca tgagctggat ccgccaggct 120ccagggaagg ggctggagtg ggtttcatac attagtagta gtggtagtac catatactac 180gcagactctg tgaagggccg attcaccatc tccagggaca acgccaagaa ctcactgtat 240ctgcaaatga acagcctgag agccgaggac acggccgtgt attactgtgc gagaga 2963098PRTHomo sapiens 30Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Lys Pro Gly Gly1 5 10 15Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Asp Tyr 20 25 30Tyr Met Ser Trp Ile Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val 35 40 45Ser Tyr Ile Ser Ser Ser Gly Ser Thr Ile Tyr Tyr Ala Asp Ser Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Ser Leu Tyr65 70 75 80Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95Ala Arg31296PRTHomo sapiens 31Gly Ala Gly Gly Thr Gly Cys Ala Gly Cys Thr Gly Thr Thr Gly Gly1 5 10 15Ala Gly Thr Cys Thr Gly Gly Gly Gly Gly Ala Gly Gly Cys Thr Thr 20 25 30Gly Gly Thr Ala Cys Ala Gly Cys Cys Thr Gly Gly Gly Gly Gly Gly 35 40 45Thr Cys Cys Cys Thr Gly Ala Gly Ala Cys Thr Cys Thr Cys Cys Thr 50 55 60Gly Thr Gly Cys Ala Gly Cys Cys Thr Cys Thr Gly Gly Ala Thr Thr65 70 75 80Cys Ala Cys Cys Thr Thr Thr Ala Gly Cys Ala Gly Cys Thr Ala Thr 85 90 95Gly Cys Cys Ala Thr Gly Ala Gly Cys Thr Gly Gly Gly Thr Cys Cys 100 105 110Gly Cys Cys Ala Gly Gly Cys Thr Cys Cys Ala Gly Gly Gly Ala Ala 115 120 125Gly Gly Gly Gly Cys Thr Gly Gly Ala Gly Thr Gly Gly Gly Thr Cys 130 135 140Thr Cys Ala Gly Cys Thr Ala Thr Thr Ala Gly Thr Gly Gly Thr Ala145 150 155 160Gly Thr Gly Gly Thr Gly Gly Thr Ala Gly Cys Ala Cys Ala Thr Ala 165 170 175Cys Thr Ala Cys Gly Cys Ala Gly Ala Cys Thr Cys Cys Gly Thr Gly 180 185 190Ala Ala Gly Gly Gly Cys Cys Gly Gly Thr Thr Cys Ala Cys Cys Ala 195 200 205Thr Cys Thr Cys Cys Ala Gly Ala Gly Ala Cys Ala Ala Thr Thr Cys 210 215 220Cys Ala Ala Gly Ala Ala Cys Ala Cys Gly Cys Thr Gly Thr Ala Thr225 230 235 240Cys Thr Gly Cys Ala Ala Ala Thr Gly Ala Ala Cys Ala Gly Cys Cys 245 250 255Thr Gly Ala Gly Ala Gly Cys Cys Gly Ala Gly Gly Ala Cys Ala Cys 260 265 270Gly Gly Cys Cys Gly Thr Ala Thr Ala Thr Thr Ala Cys Thr Gly Thr 275 280 285Gly Cys Gly Ala Ala Ala Gly Ala 290 2953298PRTHomo sapiens 32Glu Val Gln Leu Leu Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly1 5 10 15Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Tyr 20 25 30Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val 35 40 45Ser Ala Ile Ser Gly Ser Gly Gly Ser Thr Tyr Tyr Ala Asp Ser Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr65 70 75 80Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95Ala Lys33296DNAHomo sapiens 33caggtgcagc tgcaggagtc gggcccagga ctggtgaagc cttcggggac cctgtccctc 60acctgcgctg tctctggtgg ctccatcagc agtagtaact ggtggagttg ggtccgccag 120cccccaggga aggggctgga gtggattggg gaaatctatc atagtgggag caccaactac 180aacccgtccc tcaagagtcg agtcaccata tcagtagaca agtccaagaa ccagttctcc 240ctgaagctga gctctgtgac cgccgcggac acggccgtgt attactgtgc gagaga 2963498PRTHomo sapiens 34Gln Val Gln Leu Gln Glu Ser Gly Pro Gly Leu Val Lys Pro Ser Gly1 5 10 15Thr Leu Ser Leu Thr Cys Ala Val Ser Gly Gly Ser Ile Ser Ser Ser 20 25 30Asn Trp Trp Ser Trp Val Arg Gln Pro Pro Gly Lys Gly Leu Glu Trp 35 40 45Ile Gly Glu Ile Tyr His Ser Gly Ser Thr Asn Tyr Asn Pro Ser Leu 50 55 60Lys Ser Arg Val Thr Ile Ser Val Asp Lys Ser Lys Asn Gln Phe Ser65 70 75 80Leu Lys Leu Ser Ser Val Thr Ala Ala Asp Thr Ala Val Tyr Tyr Cys 85 90 95Ala Arg35293DNAHomo sapiens 35caggtgcagc tgcaggagtc gggcccagga ctggtgaagc cttcggagac cctgtccctc 60acctgcactg tctctggtgg ctccatcagt agttactact ggagctggat ccggcagccc 120ccagggaagg gactggagtg gattgggtat atctattaca gtgggagcac caactacaac 180ccctccctca agagtcgagt caccatatca gtagacacgt ccaagaacca gttctccctg 240aagctgagct ctgtgaccgc tgcggacacg gccgtgtatt actgtgcgag aga 2933697PRTHomo sapiens 36Gln Val Gln Leu Gln Glu Ser Gly Pro Gly Leu Val Lys Pro Ser Glu1 5 10 15Thr Leu Ser Leu Thr Cys Thr Val Ser Gly Gly Ser Ile Ser Ser Tyr 20 25 30Tyr Trp Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Leu Glu Trp Ile 35 40 45Gly Tyr Ile Tyr Tyr Ser Gly Ser Thr Asn Tyr Asn Pro Ser Leu Lys 50 55 60Ser Arg Val Thr Ile Ser Val Asp Thr Ser Lys Asn Gln Phe Ser Leu65 70 75 80Lys Leu Ser Ser Val Thr Ala Ala Asp Thr Ala Val Tyr Tyr Cys Ala 85 90 95Arg37290PRTHomo sapiens 37Gly Ala Ala Ala Thr Thr Gly Thr Gly Thr Thr Gly Ala Cys Gly Cys1 5 10 15Ala Gly Thr Cys Thr Cys Cys Ala Gly Gly Cys Ala Cys Cys Cys Thr 20 25 30Gly Thr Cys Thr Thr Thr Gly Thr Cys Thr Cys Cys Ala Gly Gly Gly 35 40 45Gly Ala Ala Ala Gly Ala Gly Cys Cys Ala Cys Cys Cys Thr Cys Thr 50 55 60Cys Cys Thr Gly Cys Ala Gly Gly Gly Cys Cys Ala Gly Thr Cys Ala65 70 75 80Gly Ala Gly Thr Gly Thr Thr Ala Gly Cys Ala Gly Cys Ala Gly Cys 85 90 95Thr Ala Cys Thr Thr Ala Gly Cys Cys Thr Gly Gly Thr Ala Cys Cys 100 105 110Ala Gly Cys Ala Gly Ala Ala Ala Cys Cys Thr Gly Gly Cys Cys Ala 115 120 125Gly Gly Cys Thr Cys Cys Cys Ala Gly Gly Cys Thr Cys Cys Thr Cys 130 135 140Ala Thr Cys Thr Ala Thr Gly Gly Thr Gly Cys Ala Thr Cys Cys Ala145 150 155 160Gly Cys Ala Gly Gly Gly Cys Cys Ala Cys Thr Gly Gly Cys Ala Thr 165 170 175Cys Cys Cys Ala Gly Ala Cys Ala Gly Gly Thr Thr Cys Ala Gly Thr 180 185 190Gly Gly Cys Ala Gly Thr Gly Gly Gly Thr Cys Thr Gly Gly Gly Ala 195 200 205Cys Ala Gly Ala Cys Thr Thr Cys Ala Cys Thr Cys Thr Cys Ala Cys 210 215 220Cys Ala Thr Cys Ala Gly Cys Ala Gly Ala Cys Thr Gly Gly Ala Gly225 230 235 240Cys Cys Thr Gly Ala Ala Gly Ala Thr Thr Thr Thr Gly Cys Ala Gly 245 250 255Thr Gly Thr Ala Thr Thr Ala Cys Thr Gly Thr Cys Ala Gly Cys Ala 260 265 270Gly Thr Ala Thr Gly Gly Thr Ala Gly Cys Thr Cys Ala Cys Cys Thr 275 280 285Cys Cys 2903896PRTHomo sapiens 38Glu Ile Val Leu Thr Gln Ser Pro Gly Thr Leu Ser Leu Ser Pro Gly1 5 10 15Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Gln Ser Val Ser Ser Ser 20 25 30Tyr Leu Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu 35 40 45Ile Tyr Gly Ala Ser Ser Arg Ala Thr Gly Ile Pro Asp Arg Phe Ser 50 55 60Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Arg Leu Glu65 70 75 80Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln Tyr Gly Ser Ser Pro 85 90 9539288DNAHomo sapiensmisc_feature(288)..(288)n is a, c, g, or t 39gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60atcacttgcc gggcaagtca gggcattaga aatgatttag gctggtatca gcagaaacca 120gggaaagccc ctaagcgcct gatctatgct gcatccagtt tgcaaagtgg ggtcccatca 180aggttcagcg gcagtggatc tgggacagaa ttcactctca caatcagcag cctgcagcct 240gaagattttg caacttatta ctgtctacag cataatagtt accctccn 2884096PRTHomo sapiens 40Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly1 5 10 15Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Gly Ile Arg Asn Asp 20 25 30Leu Gly Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Arg Leu Ile 35 40 45Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro65 70 75 80Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln His Asn Ser Tyr Pro Pro 85 90 9541288DNAHomo sapiens 41gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60atcacttgcc gggcaagtca gagcattagc agctatttaa attggtatca gcagaaacca 120gggaaagccc ctaagctcct gatctatgct gcatccagtt tgcaaagtgg ggtcccatca 180aggttcagtg gcagtggatc tgggacagat ttcactctca ccatcagcag tctgcaacct 240gaagattttg caacttacta ctgtcaacag agttacagta cccctcch 2884296PRTHomo sapiens 42Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly1 5 10 15Asp Arg Val Thr Ile Thr Cys Arg Ala Ser Gln Ser Ile Ser Ser Tyr 20 25 30Leu Asn Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile 35 40 45Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro65 70 75 80Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln Ser Tyr Ser Thr Pro Pro 85 90 9543293DNAHomo sapiens 43caggtgcagc tgcaggagtc gggcccagga ctggtgaagc cttcggagac cctgtccctc 60acctgcactg tctctggtgg ctccatcagt agttactact ggagctggat ccggcagccc 120gccgggaagg gactggagtg gattgggcgt atctatacca gtgggagcac caactacaac 180ccctccctca agagtcgagt caccatgtca gtagacacgt ccaagaacca gttctccctg 240aagctgagct ctgtgaccgc cgcggacacg gccgtgtatt actgtgcgag aga 2934497PRTHomo sapiens 44Gln Val Gln Leu Gln Glu Ser Gly Pro Gly Leu Val Lys Pro Ser Glu1 5 10 15Thr Leu Ser Leu Thr Cys Thr Val Ser Gly Gly Ser Ile Ser Ser Tyr 20 25 30Tyr Trp Ser Trp Ile Arg Gln Pro Ala Gly Lys Gly Leu Glu Trp Ile 35 40 45Gly Arg Ile Tyr Thr Ser Gly Ser Thr Asn Tyr Asn Pro Ser Leu Lys 50 55 60Ser Arg Val Thr Met Ser Val Asp Thr Ser Lys Asn Gln Phe Ser Leu65 70 75 80Lys Leu Ser Ser Val Thr Ala Ala Asp Thr Ala Val Tyr Tyr Cys Ala 85 90 95Arg45470PRTHomo sapiens 45Met Glu Phe Gly Leu Ser Trp Leu Phe Leu Val Ala Ile Leu Lys Gly1 5 10 15Val Gln Cys Glu Val Gln Leu Leu Glu Ser Gly Gly Gly Leu Val Gln 20 25 30Pro Gly Gly Ser Leu Arg Leu Ser Cys Thr Ala Ser Gly Phe Thr Phe 35 40 45Ser Ser Tyr Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu 50 55 60Glu Trp Val Ser Ala Ile Ser Gly Ser Gly Gly Thr Thr Phe Tyr Ala65 70 75 80Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Arg Thr 85 90 95Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val 100 105 110Tyr Tyr Cys Ala Lys Asp Leu Gly Trp Ser Asp Ser Tyr Tyr Tyr Tyr 115 120 125Tyr Gly Met Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser 130 135 140Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys Ser Arg145 150 155 160Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr

165 170 175Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser 180 185 190Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser 195 200 205Leu Ser Ser Val Val Thr Val Pro Ser Ser Asn Phe Gly Thr Gln Thr 210 215 220Tyr Thr Cys Asn Val Asp His Lys Pro Ser Asn Thr Lys Val Asp Lys225 230 235 240Thr Val Glu Arg Lys Cys Cys Val Glu Cys Pro Pro Cys Pro Ala Pro 245 250 255Pro Val Ala Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp 260 265 270Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp 275 280 285Val Ser His Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly 290 295 300Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn305 310 315 320Ser Thr Phe Arg Val Val Ser Val Leu Thr Val Val His Gln Asp Trp 325 330 335Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro 340 345 350Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr Lys Gly Gln Pro Arg Glu 355 360 365Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn 370 375 380Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile385 390 395 400Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr 405 410 415Thr Pro Pro Met Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys 420 425 430Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys 435 440 445Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu 450 455 460Ser Leu Ser Pro Gly Lys465 47046470PRTHomo sapiens 46Met Glu Phe Gly Leu Ser Trp Leu Phe Leu Val Ala Ile Leu Lys Gly1 5 10 15Val Gln Cys Glu Val Gln Leu Leu Glu Ser Gly Gly Gly Leu Val Gln 20 25 30Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe 35 40 45Ser Ser Tyr Ala Met Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu 50 55 60Glu Trp Val Ser Ala Ile Ser Gly Ser Gly Gly Ser Thr Tyr Tyr Ala65 70 75 80Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ser Lys Asn 85 90 95Thr Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val 100 105 110Tyr Tyr Cys Ala Lys Gly Tyr Ser Ser Gly Trp Tyr Tyr Tyr Tyr Tyr 115 120 125Tyr Gly Met Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser 130 135 140Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys Ser Arg145 150 155 160Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr 165 170 175Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser 180 185 190Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser 195 200 205Leu Ser Ser Val Val Thr Val Pro Ser Ser Asn Phe Gly Thr Gln Thr 210 215 220Tyr Thr Cys Asn Val Asp His Lys Pro Ser Asn Thr Lys Val Asp Lys225 230 235 240Thr Val Glu Arg Lys Cys Cys Val Glu Cys Pro Pro Cys Pro Ala Pro 245 250 255Pro Val Ala Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp 260 265 270Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp 275 280 285Val Ser His Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly 290 295 300Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn305 310 315 320Ser Thr Phe Arg Val Val Ser Val Leu Thr Val Val His Gln Asp Trp 325 330 335Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro 340 345 350Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr Lys Gly Gln Pro Arg Glu 355 360 365Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn 370 375 380Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile385 390 395 400Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr 405 410 415Thr Pro Pro Met Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys 420 425 430Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys 435 440 445Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu 450 455 460Ser Leu Ser Pro Gly Lys465 47047236PRTHomo sapiens 47Met Asp Met Arg Val Pro Ala Gln Leu Leu Gly Leu Leu Leu Leu Trp1 5 10 15Phe Pro Gly Ala Arg Cys Asp Ile Gln Met Thr Gln Phe Pro Ser Ser 20 25 30Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Arg Ala Ser 35 40 45Gln Gly Ile Arg Asn Asp Leu Gly Trp Tyr Gln Gln Lys Pro Gly Lys 50 55 60Ala Pro Lys Arg Leu Ile Tyr Ala Ala Ser Arg Leu His Arg Gly Val65 70 75 80Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr 85 90 95Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln 100 105 110His Asn Ser Tyr Pro Cys Ser Phe Gly Gln Gly Thr Lys Leu Glu Ile 115 120 125Lys Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp 130 135 140Glu Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn145 150 155 160Phe Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu 165 170 175Gln Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp 180 185 190Ser Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr 195 200 205Glu Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser 210 215 220Ser Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys225 230 23548236PRTHomo sapiens 48Met Asp Met Arg Val Pro Ala Gln Leu Leu Gly Leu Leu Leu Leu Trp1 5 10 15Phe Pro Gly Ala Arg Cys Asp Ile Gln Met Thr Gln Ser Pro Ser Ser 20 25 30Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Arg Ala Ser 35 40 45Gln Gly Ile Arg Asn Asp Leu Gly Trp Tyr Gln Gln Lys Pro Gly Lys 50 55 60Ala Pro Lys Arg Leu Ile Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val65 70 75 80Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr 85 90 95Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln 100 105 110His Asn Ser Tyr Pro Tyr Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile 115 120 125Lys Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp 130 135 140Glu Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn145 150 155 160Phe Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu 165 170 175Gln Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp 180 185 190Ser Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr 195 200 205Glu Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser 210 215 220Ser Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys225 230 23549470PRTHomo sapiens 49Met Glu Phe Gly Leu Ser Trp Val Phe Leu Val Ala Ile Ile Lys Gly1 5 10 15Val Gln Cys Gln Ala Gln Leu Val Glu Ser Gly Gly Gly Leu Val Lys 20 25 30Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe 35 40 45Ser Asp Tyr Tyr Met Ser Trp Ile Arg Gln Ala Pro Gly Lys Gly Leu 50 55 60Glu Trp Val Ser Tyr Ile Ser Ser Ser Gly Ser Thr Arg Asp Tyr Ala65 70 75 80Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn 85 90 95Ser Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val 100 105 110Tyr Tyr Cys Val Arg Asp Gly Val Glu Thr Thr Phe Tyr Tyr Tyr Tyr 115 120 125Tyr Gly Met Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser 130 135 140Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys Ser Arg145 150 155 160Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr 165 170 175Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser 180 185 190Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser 195 200 205Leu Ser Ser Val Val Thr Val Pro Ser Ser Asn Phe Gly Thr Gln Thr 210 215 220Tyr Thr Cys Asn Val Asp His Lys Pro Ser Asn Thr Lys Val Asp Lys225 230 235 240Thr Val Glu Arg Lys Cys Cys Val Glu Cys Pro Pro Cys Pro Ala Pro 245 250 255Pro Val Ala Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp 260 265 270Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp 275 280 285Val Ser His Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly 290 295 300Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn305 310 315 320Ser Thr Phe Arg Val Val Ser Val Leu Thr Val Val His Gln Asp Trp 325 330 335Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro 340 345 350Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr Lys Gly Gln Pro Arg Glu 355 360 365Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn 370 375 380Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile385 390 395 400Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr 405 410 415Thr Pro Pro Met Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys 420 425 430Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys 435 440 445Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu 450 455 460Ser Leu Ser Pro Gly Lys465 47050473PRTHomo sapiens 50Met Glu Phe Gly Leu Ser Trp Val Phe Leu Val Ala Ile Ile Lys Gly1 5 10 15Val Gln Cys Gln Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Lys 20 25 30Pro Gly Gly Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe 35 40 45Ser Asp Tyr Tyr Met Ser Trp Ile Arg Gln Ala Pro Gly Lys Gly Leu 50 55 60Glu Trp Val Ser Tyr Ile Ser Ser Ser Gly Ser Thr Ile Tyr Tyr Ala65 70 75 80Asp Ser Val Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn 85 90 95Ser Leu Tyr Leu Gln Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val 100 105 110Tyr Tyr Cys Ala Arg Val Leu Arg Phe Leu Glu Trp Leu Leu Tyr Tyr 115 120 125Tyr Tyr Tyr Tyr Gly Met Asp Val Trp Gly Gln Gly Thr Thr Val Thr 130 135 140Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro145 150 155 160Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala Ala Leu Gly Cys Leu Val 165 170 175Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala 180 185 190Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly 195 200 205Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Asn Phe Gly 210 215 220Thr Gln Thr Tyr Thr Cys Asn Val Asp His Lys Pro Ser Asn Thr Lys225 230 235 240Val Asp Lys Thr Val Glu Arg Lys Cys Cys Val Glu Cys Pro Pro Cys 245 250 255Pro Ala Pro Pro Val Ala Gly Pro Ser Val Phe Leu Phe Pro Pro Lys 260 265 270Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val 275 280 285Val Val Asp Val Ser His Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr 290 295 300Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu305 310 315 320Gln Phe Asn Ser Thr Phe Arg Val Val Ser Val Leu Thr Val Val His 325 330 335Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys 340 345 350Gly Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr Lys Gly Gln 355 360 365Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met 370 375 380Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro385 390 395 400Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn 405 410 415Tyr Lys Thr Thr Pro Pro Met Leu Asp Ser Asp Gly Ser Phe Phe Leu 420 425 430Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val 435 440 445Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln 450 455 460Lys Ser Leu Ser Leu Ser Pro Gly Lys465 47051236PRTHomo sapiens 51Met Asp Met Arg Val Pro Ala Gln Leu Leu Gly Leu Leu Leu Leu Trp1 5 10 15Phe Pro Gly Ala Arg Cys Asp Ile Gln Met Thr Gln Ser Pro Ser Ser 20 25 30Leu Ser Ala Ser Val Gly Asp Arg Val Thr Phe Thr Cys Arg Ala Ser 35 40 45Gln Asp Ile Arg Arg Asp Leu Gly Trp Tyr Gln Gln Lys Pro Gly Lys 50 55 60Ala Pro Lys Arg Leu Ile Tyr Ala Ala Ser Arg Leu Gln Ser Gly Val65 70 75 80Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr 85 90 95Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln 100 105 110His Asn Asn Tyr Pro Arg Thr Phe Gly Gln Gly Thr Glu Val Glu Ile 115 120 125Ile Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp 130 135 140Glu Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn145 150 155 160Phe Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu 165 170 175Gln Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp 180 185 190Ser Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr 195 200 205Glu Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser 210 215 220Ser Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys225 230 23552236PRTHomo sapiens 52Met Asp Met Arg Val Pro Ala Gln Leu Leu Gly Leu Leu Leu Leu Trp1 5 10 15Phe Pro Gly Ala Arg Cys Asp Ile Gln Met Thr Gln Ser Pro Ser Ser 20 25 30Leu Ser Ala Ser Val Gly Asp Arg Val Thr Ile Thr Cys Arg Ala Ser 35 40

45Gln Gly Ile Arg Asn Asp Leu Gly Trp Tyr Gln Gln Lys Pro Gly Lys 50 55 60Ala Pro Lys Arg Leu Ile Tyr Ala Ala Ser Ser Leu Gln Ser Gly Val65 70 75 80Pro Ser Arg Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr 85 90 95Ile Ser Ser Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gln 100 105 110His Asn Ser Tyr Pro Trp Thr Phe Gly Gln Gly Thr Lys Val Glu Ile 115 120 125Lys Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp 130 135 140Glu Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn145 150 155 160Phe Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu 165 170 175Gln Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp 180 185 190Ser Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr 195 200 205Glu Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser 210 215 220Ser Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys225 230 23553326DNAArtificial SequenceDescription of Artificial Sequence Consensus Sequence 53gacatccaga tgacccagty tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60wtcacttgcc gggcaagtca ggrcattaga mrtgatttag gctggtwtca gcagaaacca 120gggaaagcyc ctaagcgcct gatctatgct gcatccmrwt trcammgwgg ggtcccatca 180aggttcagcg gcagtggatc tgggacagaa ttcactctca caatcagcmg cctgcagcct 240gaagattttg caacttatta ctgtytacar cataatartt aycckybsns kttyggcsrr 300gggaccrags tggaratcaw acgaac 32654322DNAArtificial SequenceDescription of Artificial Sequence Consensus Sequence 54gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgyaggaga cagagtcacc 60atcacttgcc gggcaagtca gagcattagy asctwtttaa attggtatca gcagaaacca 120gggaaagccc ctaarctcct gatcyatgyt gcatccagtt trcaargtgg ggtcccatca 180aggttcagtg gcagtggatc tgggacagat ttcactctca ccatcagcag tctgcaacct 240gaagattttg caacttacta ctgtcaacag agttacartr ccccayychc tttcggcgga 300gggaccaagg tggagatcaa ac 32255325DNAArtificial SequenceDescription of Artificial Sequence Consensus Sequence 55gaaattgtgt tgacgcagtc tccaggcacc ctgtctttgt ctccagggga aagagccacc 60ctctcctgya gggccagtca gagtgttmgc rgcagstact tagcctggta ccagcagaaa 120cctggccagg ctcccaggct cctcatctat ggtgcatcca gcagggccac tggcatccca 180gacaggttca gtggcagtgg gtctgggaca gacttcactc tcaccatcag cagactggag 240cctgaagatt ttgcagtgtw ttactgtcag cagtatggta gytcacctcs nacgttcggc 300caagggacca aggtggaaat caaac 32556376DNAArtificial SequenceDescription of Artificial Sequence Consensus Sequence 56caggtgcagc tggtggagtc tgggggaggc ttggtcaagc ctggagggtc cctgagactc 60tcctgtgcag cctctggatt cacyttcagt gactactaya tgagctggat ccgccaggct 120ccagggaagg ggctggartg ggtttcatac attagtagta gtggtagtac cakakactac 180gcagactctg tgaagggccc attcaccatc tccagggaca acgccaagaa ctcactgtat 240ctgcaaatga acagcctgag agccgaggac acggccgtgt attactgtgy gagagatgga 300gtggaaacta ctttttacta ctactactac ggtatggacg tctggggcca agggaccacg 360gtcaccgtct cctcag 37657358DNAArtificial SequenceDescription of Artificial Seqence Consensus Sequence 57caggtgcagc tgcaggagtc gggcccagga ctggtgaagc cttcggagac cctgtccctc 60acctgcactg tctctggtgg ctccatcagt arttactact ggagctggat ccggcagccc 120gccgggaagg gactggagtg gattgggcgt atctatacca gtgggagcmc caactacaac 180ccctccctca agagtcgagt caccatgtca gtagacacgt ccaagaacca gttctccctg 240aagctgarct ctgtgaccgc cgcggacacg gccgtgtatt actgtgcggt aacgattttt 300ggagtggtta ttatctttga ctactggggc cagrganccc tggtcaccgt ctcctcag 35858418DNAArtificial SequenceDescription of Artificial Sequence Consensus Sequence 58caggtgcagc tgttggagtc tgggggaggc ttggtacagc ctggggggtc cctgagactc 60tcctgtrcag cctctggatt cacctttagc agctatgcca tgarctgggt ccgccaggct 120ccagggaagg ggctggagtg ggtctcagst attastggka gtggtggtab yacatwctac 180gcagactccg tgaagggccc gttcaccatc tccagagaca attccargam cacgctgtat 240ctgcaaatga acagcctgag agccgaggac acggccgtat attactgtgc gaaagatctk 300ggctrsksyg actyttacta ctactactac ggtatggacg tctggggcca agggacyacg 360gtgattatga gttggttcga cccctggggc cagggaaccc tggtcaccgt ctcctcag 41859364DNAArtificial SequenceDescription of Artificial Sequence Consensus Sequence 59caggtgcagc tgcaggagtc gggcccagga ctggtgaagc cttcggagac cctgtccctc 60acctgcactg tctctggtgg ctccatcagt agttactact ggagytggat ccggcagccc 120ccagggaagg gactggagtg gattgggtat atctattaca gtgggagcac caactacaac 180ccctccctca agagtcgact caccatatca gtagacacgt ccaagaacca gttctccctg 240aagctgagyt ctgtgaccgc tgcggacacg gccgtgtatt actgtgccag gacgtatagc 300agttcgttct actactacgg tatggacgtc tggggccaag ggaccacggt caccgtctcc 360tcag 3646015PRTArtificial SequenceDescription of Artificial Sequence Consensus Sequence 60Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser1 5 10 15

* * * * *


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

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

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

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