Antagonist Anti-notch3 Antibodies And Their Use In The Prevention And Treatment Of Notch3-related Diseases

Fung; Sek Chung ;   et al.

Patent Application Summary

U.S. patent application number 13/669216 was filed with the patent office on 2013-04-04 for antagonist anti-notch3 antibodies and their use in the prevention and treatment of notch3-related diseases. This patent application is currently assigned to GENENTECH, INC.. The applicant listed for this patent is Genentech, Inc.. Invention is credited to Sek Chung Fung, Kang Li, Yucheng Li, Sanjaya Singh, Bin-Bing Stephen Zhou.

Application Number20130084288 13/669216
Document ID /
Family ID39367005
Filed Date2013-04-04

United States Patent Application 20130084288
Kind Code A1
Fung; Sek Chung ;   et al. April 4, 2013

ANTAGONIST ANTI-NOTCH3 ANTIBODIES AND THEIR USE IN THE PREVENTION AND TREATMENT OF NOTCH3-RELATED DISEASES

Abstract

The present invention relates to antagonist antibodies that specifically bind to Notch 3 and inhibit its activation. The present invention includes antibodies binding to a conformational epitope comprising the first Lin12 domain and the second dimerization domain. The present invention also includes uses of these antibodies to treat or prevent Notch 3 related diseases or disorders.


Inventors: Fung; Sek Chung; (Gaithersburg, MD) ; Li; Kang; (San Diego, CA) ; Li; Yucheng; (San Diego, CA) ; Singh; Sanjaya; (Sandy Hook, CT) ; Zhou; Bin-Bing Stephen; (Ho Hokus, NJ)
Applicant:
Name City State Country Type

Genentech, Inc.;

South San Francisco

CA

US
Assignee: GENENTECH, INC.
South San Francisco
CA

Family ID: 39367005
Appl. No.: 13/669216
Filed: November 5, 2012

Related U.S. Patent Documents

Application Number Filing Date Patent Number
13353173 Jan 18, 2012 8329868
13669216
13023128 Feb 8, 2011 8148106
13353173
11958099 Dec 17, 2007 7935791
13023128
60875597 Dec 18, 2006
60879218 Jan 6, 2007

Current U.S. Class: 424/133.1 ; 424/139.1; 435/375; 435/6.13; 530/387.9
Current CPC Class: A61P 35/00 20180101; A61P 19/02 20180101; A61K 2039/505 20130101; A61P 3/06 20180101; C07K 14/705 20130101; C07K 16/28 20130101; C07K 2319/30 20130101; A61P 3/04 20180101; A61P 37/00 20180101; A61P 1/16 20180101; C07K 2317/76 20130101; A61P 29/00 20180101; C07K 16/18 20130101; A61P 3/00 20180101; A61P 35/02 20180101; C07K 2317/56 20130101; A61P 3/10 20180101
Class at Publication: 424/133.1 ; 530/387.9; 424/139.1; 435/375; 435/6.13
International Class: C07K 16/28 20060101 C07K016/28

Claims



1. An isolated polypeptide comprising the amino acid sequence of the variable light ("VL") chain region of a monoclonal antibody that binds to Notch3, wherein the antibody binds to a conformational epitope of a Notch3 fragment comprising amino acids 1378-1640 of SEQ ID NO:1, and wherein the antibody inhibits Notch3 signaling.

2. The polypeptide of claim 1, wherein the antibody binds to amino acid residues in the LIN12 domain (SEQ ID NO:9) and the dimerization domain (SEQ ID NO:18).

3. The polypeptide of claim 1, wherein the VL chain region comprises CDR-L1 of SEQ ID NO:35, CDR-L2 of SEQ ID NO:36, and CDR-L3 of SEQ ID NO:37.

4. The polypeptide of claim 3, wherein the VL chain region comprises SEQ ID NO:3.

5. The polypeptide of claim 1, wherein the VL chain region comprises CDR-L1 of SEQ ID NO:41, CDR-L2 of SEQ ID NO:42, and CDR-L3 of SEQ ID NO:43.

6. The polypeptide of claim 5, wherein the VL chain region comprises SEQ ID NO:5.

7. The polypeptide of claim 1, wherein the antibody inhibits Notch3 signaling by about 50%.

8. A method of treating a Notch 3 related disease or disorder comprising administering to a mammal a monoclonal antibody that specifically binds to Notch3, wherein the antibody specifically binds to a conformational epitope of a Notch3 fragment consisting of amino acids 1378-1640 of SEQ ID NO:1, and wherein the antibody inhibits Notch3 signaling.

9. The method of claim 8, wherein the antibody binds to amino acid residues in the LIN12 domain (SEQ ID NO:9) and the dimerization domain (SEQ ID NO:18).

10. The method of claim 8, wherein the antibody comprises a VH chain region comprising CDR-H1 of SEQ ID NO:32, CDR-H2 of SEQ ID NO:33, and CDR-H3 of SEQ ID NO:34, and a VL chain region comprising CDR-L1 of SEQ ID NO:35, CDR-L2 of SEQ ID NO:36, and CDR-L3 of SEQ ID NO:37.

11. The method of claim 10, wherein the VH chain region comprises SEQ ID NO:2, and the VL chain region comprises SEQ ID NO:3.

12. The method of claim 8, wherein the antibody is a humanized form of a monoclonal antibody comprising the VH chain region of SEQ ID NO:2 and the VL chain region of SEQ ID NO:3.

13. The method of claim 8, wherein the antibody comprises a VH chain region comprising CDR-H1 of SEQ ID NO:38, CDR-H2 of SEQ ID NO:39, and CDR-H3 of SEQ ID NO:40, and a VL chain region comprising CDR-L1 of SEQ ID NO:41, CDR-L2 of SEQ ID NO:42, and CDR-L3 of SEQ ID NO:43.

14. The method of claim 13, wherein the VH chain region comprises SEQ ID NO:4, and the VL chain region comprises SEQ ID NO:5.

15. The method of claim 8, wherein the antibody is a humanized form of a monoclonal antibody comprising the VH chain region of SEQ ID NO:4 and the VL chain region of SEQ ID NO:5.

16. The method of claim 8, wherein the disease or disorder is a cancer.

17. The method of claim 16, wherein the cancer is selected from the group consisting of non-small cell lung cancer, ovarian cancer, T-cell acute lymphoblastic leukemia, pancreatic cancer, prostate cancer, plasma cell neoplasms, neuroblastoma and extramedullary plasmacytoma.

18. The method of claim 8, wherein the disease or disorder is selected from the group consisting of liver disease involving aberrant vascularization, diabetes, diseases or disorders involving vascular cell fate, and rheumatoid arthritis.

19. A method of inhibiting Notch3 signaling in a cell comprising the step of contacting the cell with a monoclonal antibody that binds to a conformational epitope of a Notch3 fragment consisting of amino acids 1378-1640 of SEQ ID NO:1.

20. The method of claim 19, wherein the antibody inhibits ligand-mediated Notch3 signaling.

21. The method of claim 20, wherein the ligand is Jagged1, Jagged2, or Delta-like 4.

22. The method of claim 20, wherein the antibody reduces ligand binding to Notch3.

23. The method of claim 19, wherein a reduction in Notch3 signaling is determined by measuring Notch3-mediated reporter gene expression in vitro.

24. The method of claim 23, wherein the reporter gene encodes a firefly luciferase protein.

25. The method of claim 19, wherein Notch3 signaling is inhibited by about 50%.

26. The method of claim 19, wherein the cell is a cancer cell.
Description



RELATED APPLICATIONS

[0001] This application is a divisional application of U.S. patent application Ser. No. 13/353,173, now U.S. Pat. No. 8,329,868, filed Jan. 18, 2012, which is a divisional application of U.S. patent application Ser. No. 13/023,128, now U.S. Pat. No. 8,148,106, filed Feb. 8, 2011, which is a divisional application of U.S. patent application Ser. No. 11/958,099, now U.S. Pat. No. 7,935,791, filed Dec. 17, 2007, which claims the benefit of U.S. Provisional Patent Application No. 60/875,597, filed Dec. 18, 2006, and U.S. Provisional Patent Application No. 60/879,218, filed Jan. 6, 2007. The disclosures of the foregoing applications are incorporated herein by reference in their entirety.

FIELD OF THE INVENTION

[0002] The present invention relates to antagonist anti-Notch3 antibodies and their use in the amelioration, treatment, or prevention of a Notch3-related disease or disorder.

BACKGROUND OF THE INVENTION

[0003] The Notch gene was first described in 1917 when a strain of the fruit fly Drosophila melanogaster was found to have notched wing blades (Morgan, Am Nat 51:513 (1917)). The gene was cloned almost seventy years later and was determined to be a cell surface receptor playing a key role in the development of many different cell types and tissues in Drosophila (Wharton et al., Cell 43:567 (1985)). The Notch signaling pathway was soon found to be a signaling mechanism mediated by cell-cell contact and has been evolutionarily conserved from Drosophila to human. Notch receptors have been found to be involved in many cellular processes, such as differentiation, cell fate decisions, maintenance of stem cells, cell motility, proliferation, and apoptosis in various cell types during development and tissue homeostasis (For review, see Artavanis-Tsakonas, et al., Science 268:225 (1995)).

[0004] Mammals possess four Notch receptor proteins (designated Notch1 to Notch4) and five corresponding ligands (designated Delta-1 (DLL-1), Delta-3 (DLL-3), Delta-4 (DLL-4), Jagged-1 and Jagged-2). The mammalian Notch receptor genes encode .about.300 kD proteins that are cleaved during their transport to the cell surface and exist as heterodimers. The extracellular portion of the Notch receptor has thirty-four epidermal growth factor (EGF)-like repeats and three cysteine-rich Notch/LIN12 repeats. The association of two cleaved subunits is mediated by sequences lying immediately N-terminal and C-terminal of the cleavage site, and these two subunits constitute the Notch heterodimerization (HD) domains (Wharton, et al., Cell 43:567 (1985); Kidd, et al., Mol Cell Biol 6:3431 (1986); Kopczynski, et al., Genes Dev 2:1723 (1988); Yochem, et al., Nature 335:547 (1988)).

[0005] At present, it is still not clear how Notch signaling is regulated by different receptors or how the five ligands differ in their signaling or regulation. The differences in signaling and/or regulation may be controlled by their expression patterns in different tissues or by different environmental cues. It has been documented that Notch ligand proteins, including Jagged/Serrate and Delta/Delta-like, specifically bind to the EGF repeat region and induce receptor-mediated Notch signaling (reviewed by Bray, Nature Rev Mol Cell Biol. 7:678 (2006), and by Kadesch, Exp Cell Res. 260:1 (2000)). Among the EGF repeats, the 10th to 12th repeats are required for ligand binding to the Notch receptor, and the other EGF repeats may enhance receptor-ligand interaction (Xu, et al., J Biol. Chem. 280:30158 (2005); Shimizu, et al., Biochem Biophys Res Comm. 276:385 (2000)). Although the LIN12 repeats and the dimerization domain are not directly involved in ligand binding, they play important roles in maintaining the heterodimeric protein complex, preventing ligand-independent protease cleavage and receptor activation (Sanche-Irizarry, et al., Mol Cell Biol. 24:9265 (2004); Vardar et al., Biochem. 42:7061 (2003)).

[0006] The expression of mutant forms of Notch receptors in developing Xenopus embryos interferes profoundly with normal development (Coffman, et al., Cell 73: 659 (1993)). A Notch1 knockout was found to be embryonic lethal in mice (Swiatek, et al., Genes & Dev 8:707 (1994)). In humans, there have been several genetic diseases, including cancer, linked to different Notch receptor mutations (Artavanis-Tsakonas, et al., Science 284:770 (1999)). For instance, aberrant activation of Notch1 receptor caused by translocation can lead to T cell lymphoblastic leukemia (Ellisen, et al., Cell 66:649 (1991)). Certain mutations in the HD domains of Notch1 receptor enhance signaling without ligand binding (Malecki, et al., Mol Cell Biol 26:4642 (2006)), further implicating their roles in Notch receptor activation. The signal induced by ligand binding is transmitted to the nucleus by a process involving two proteolytic cleavages of the receptor followed by nuclear translocation of the intracellular domain (Notch-IC). Although LIN12 repeats and HD domains were thought to prevent signaling in the absence of ligands, it is still unclear how ligand binding facilitates proteolytic cleavage events.

[0007] Notch receptors have been linked to a wide range of diseases including cancer, neurological disorders, and immune diseases, as evidenced by reports of the over-expression of Notch receptors in various human disease tissues and cell lines as compared to normal or nonmalignant cells (Joutel, et al. Cell & Dev Biol 9:619 (1998); Nam, et al., Curr Opin Chem Biol 6:501 (2002)). The Notch3 receptor is over-expressed in various solid tumors, including non-small cell lung cancer (NSCLC) and ovarian cancer (Haruki, et al., Cancer Res 65:3555 (2005); Park, et al., Cancer Res 66:6312 (2006); Lu, et al., Clin Cancer Res 10:3291 (2004)), suggesting the significance of Notch3 receptor expression in solid tumors. Furthermore, Notch3 receptor expression is upregulated in plasma cell neoplasms, including multiple myeloma, plasma cell leukemia, and extramedullary plasmacytoma (Hedvat, et al., Br J Haematol 122:728 (2003); pancreatic cancer (Buchler, et al., Ann Surg 242:791 (2005)); and T cell acute lymphoblastic leukemias (T-ALL) (Bellavia, et al., Proc Natl Acad Sci USA 99:3788 (2002); Screpanti, et al., Trends Mol Med 9:30 (2003)). Notch3 receptor is also expressed in a subset of neuroblastoma cell lines and serves as a marker for this type of tumor that has constitutional or tumor-specific mutations in the homeobox gene Phox2B (van Limpt, et al., Cancer Lett 228:59 (2005)). Other indications and diseases that have been linked to Notch3 receptor expression include neurological disorders (Joutel, et al., Nature 383:707 (1996)), diabetes (Anastasi, et al., J Immunol 171:4504 (2003), rheumatoid arthritis (Yabe, et al., J Orthop Sci 10:589 (2005)), vascular related diseases (Sweeney, et al., FASEB J 18:1421 (2004)), and Alagille syndrome (Flynn, et al., J Pathol 204:55 (2004)).

[0008] Although Notch3 receptor over-expression (including gene amplification) has been observed in various cancers, no activating mutations have yet been reported. It is plausible that an increased level of Notch3 receptors in tumors can be activated by different ligands in stromal cells or tumor cells and lead to enhanced Notch3 signaling. Particularly, Notch ligands have been localized to the vascular endothelium during both development and tumorigenesis (Mailhos, et al., Differentiation 69:135 (2001); Taichman, et al., Dev Dyn 225: 166 (2002)), suggesting endothelial cells could provide the ligands for Notch3 receptor activation in tumors. Similar tumor-stroma cross-talk mediated by Notch ligand and receptor have been demonstrated in different type of cancers (Houde, et al., Blood 104: 3697 (2004); Jundt, et al., Blood 103: 3511 (2004); Zeng, et al., Cancer Cell 8: 13 (2005)). Increased Notch3 signaling caused by over-expression of intracellular Notch3 (Notch3-IC) can lead to tumorigenesis in T-ALL and breast cancer animal models (Vacca, et al., The EMBO J 25: 1000 (2006); Hu, et al., Am J Pathol 168: 973 (2006)).

[0009] Notch signaling and its role in cell self-renewal have been implicated in cancer stem cells, which are a minority population in tumors and can initiate tumor formation (Reya, et al., Nature 414:105 (2001)). Normal stem cells from many tissues, including intestinal and neuronal stem cells, depend on Notch signaling for self-renewal and fate determination (Fre, et al., Nature, 435: 964 (2005); van Es, et al., Nature, 435:959 (2005); Androutsellis-Theotokis, et al., Nature, 442: 823 (2006)). Similar mechanisms could exist in cancer stem cells, and inhibition of Notch signaling by .gamma.-secretase inhibitors was shown to deplete cancer stem cells and block engraftment in embryonal brain tumors (Fan, et al., Cancer Res 66:7445 (2006)).

[0010] Inhibition of Notch signaling by .gamma.-secretase inhibitor has striking antineoplastic effects in Notch-expressing transformed cells in vitro and in xenograft models (Weijzen, et al., Nat Medicine 8: 879 (2002); Bocchetta, et al., Oncogene 22:81 (2003); Weng, et al., Science, 306:269 (2004)). More recently, a .gamma.-secretase inhibitor has been shown to efficaciously kill colon adenomas in Apc (min+) mice (van Es, et al., Nature, 435: 959 (2005)), although the therapeutic window, due to its effect on normal stem cells and the inhibition of multiple Notch pathways, is very narrow. Different from Notch1, a Notch3 gene knockout in mice was not embryonically lethal and had few defects (Domenga, et al., Genes & Dev 18: 2730 (2004)), suggesting that Notch 3 provides a potentially better therapeutic target than Notch 1.

[0011] Tournier-Lasserve et al. (U.S. Application 2003/0186290) teach the association of Notch3 receptor and CADASIL. The application discloses various mutations in the Notch3 gene and their possible association with the disease CADASIL. The application suggests the use of diagnostic antibodies to detect such mutations. The application also suggests therapeutic antibodies to treat CADASIL, i.e. agonistic antibodies, but no specific antibodies are disclosed nor how to make such antibodies.

[0012] In view of the large number of human diseases associated with the Notch3 signaling pathway, it is important that new ways of preventing and treating these diseases be identified. The current invention provides novel anti-Notch3 antibodies useful for this unmet medical need.

SUMMARY OF THE INVENTION

[0013] The present invention provides novel antibodies and fragments thereof that specifically bind to a conformational epitope of the human Notch3 receptor, the epitope comprising the LIN12 domain and the heterodimerization domain. Another aspect of the invention includes the epitope binding site and antibodies that bind this same epitope as the antibodies of the present invention. The antibodies of the present invention inhibit ligand-induced signaling through the Notch3 receptor.

[0014] The invention includes the amino acid sequences of the variable heavy and light chain of the antibodies and their corresponding nucleic acid sequences. Another embodiment of the invention includes the CDR sequences of these antibodies. Another embodiment includes humanized forms of these antibodies.

[0015] Another embodiment of the present invention includes the cell lines and vectors harboring the antibody sequences of the present invention.

[0016] The present invention also includes the conformational epitope recognized by the antagonist antibodies of the invention. The present invention also includes antibodies that bind this conformational epitope. The embodiments include a Notch 3 conformational epitope comprising the LIN12 domain having at least 80%, 85%, 90%, or 95% sequence identity with SEQ ID NO. 9 and the dimerization domain 2 having at least 80%, 85%, 90%, or 95% sequence identity with SEQ ID NO. 18. More particularly, the Notch 3 conformational epitope comprising amino acid residues 1395-1396, 1402-1404 and 1420-1422 of the L1 LIN12 domain and amino acid residues 1576-1578 and 1626-1628 of the D2 dimerization domain. The present invention includes antibodies that bind this conformational epitope.

[0017] Another embodiment of the preset invention is the use of any of these antibodies for the preparation of a medicament or composition for the treatment of diseases and disorders associated with Notch 3 receptor activation.

[0018] Another embodiment of the preset invention is the use of any of these antibodies in the treatment of disorders associated with Notch 3 activation comprising the inhibition of said activation by, e.g., inhibiting Notch 3 signaling, or neutralization of the receptor by blocking ligand binding. Notch 3 related disorders may include, but are not limited to, T-cell acute lymphoblastic leukemia, lymphoma, liver disease involving aberrant vascularization, diabetes, ovarian cancer, diseases involving vascular cell fate, rheumatoid arthritis, pancreatic cancer, non-small cell lung cancer, plasma cell neoplasms (such as multiple myeloma, plasma cell leukemia, and extramedullary plasmacytoma), and neuroblastoma.

BRIEF DESCRIPTION OF THE FIGURES

[0019] FIG. 1 depicts the amino acid sequence of Notch3. The EGF repeat region extends from amino acid residue 43 to 1383; the LIN12 domain extends from amino acid residue 1384 to 1503; and the dimerization domain extends from amino acid residue 1504 to 1640.

[0020] FIG. 2 (A-H) depicts the amino acid sequence comparison between human Notch 1 (SEQ ID NO:44), Notch 2 (SEQ ID NO:45), Notch 3 (SEQ ID NO:1), and Notch 4 (SEQ ID NO:46).

[0021] FIG. 3 depicts the percent identity of Notch 1, Notch 2, Notch 3, and Notch 4.

[0022] FIGS. 4A and 4B depict the heavy and light chain variable region sequences of anti-Notch3 monoclonal antibody MAb 256A-4 (SEQ ID NO:2 and SEQ ID NO:3, respectively), with CDR regions underlined.

[0023] FIGS. 5A and 5B depict the heavy and light chain variable region sequences of anti-Notch3 monoclonal antibody MAb 256A-8 (SEQ ID NO: 4 and SEQ ID NO:5, respectively), with CDR regions underlined.

[0024] FIG. 6 depicts a luciferase reporter assay of Example 5 showing inhibitory effects by anti-Notch3 MAbs on the Notch3 ligand Jagged 1.

[0025] FIG. 7 depicts the luciferase reporter assay showing inhibitory effects by anti-Notch3 MAbs on the Notch3 ligand Jagged 2.

[0026] FIG. 8 depicts the luciferase reporter assay showing inhibitory effects by anti-Notch3 MAbs on the Notch3 ligand DLL4.

[0027] FIGS. 9A and B depict the luciferase reporter assay showing inhibitory effects to native Notch3 in ovarian cancer cells by anti-Notch3 MAbs. (9A) Human ovarian cancer cell line, OV/CAR3 and (9B) Human ovarian cancer cell line, A2780.

[0028] FIG. 10 depicts the apoptosis assay of Example 6 showing that cell survival effect induced by Jagged1 was inhibited by anti-Notch3 MAbs.

[0029] FIGS. 11A and B depict the inhibitory effect of anti-Notch3 MAbs on cell migration (11A) and invasion (11B) of Example 7.

[0030] FIG. 12 depicts a schematic diagram of the Notch1-Notch3 domain-swap protein expressed as a fusion protein with human IgG/Fc linked to C-terminus.

[0031] FIG. 13A depicts an ELISA using anti-human Fc control antibody as the detection antibody showing that the proteins of FIG. 12 were expressed in conditioned medium. FIG. 13B depicts an ELISA using 256A-4 as the detection antibody. FIG. 13C depicts an ELISA using 256A-8 as the detection antibody. FIG. 13D depicts an ELISA using a positive control antibody 256A-13 as the detection antibody.

[0032] FIGS. 14A and B depict the comparison of the engineered Notch3 leader peptide coding sequence (SEQ ID NO:47) to the native Notch3 leader peptide coding sequence (SEQ ID NO:48) (NCBI GENBANK.RTM. Accession No. NM.sub.--000435) showing the changes of nucleotides (14A) and the translated amino acid sequence of the engineered Notch leader peptide sequence (SEQ ID NO:6) (14B).

[0033] FIG. 15 depicts the generation of domain swap construct by PCR-SOE method. Arrow bars represent PCR primers. Open bar, Notch3 sequence. Filled bar, Notch1 sequence.

[0034] FIG. 16 depicts the amino acid sequences used in the Notch3 LIN12 domain epitope mapping of the MAb 256A-4 and 256A-8.

[0035] FIG. 17 depicts the amino acid sequences used in the Notch3 dimerization domain epitope mapping of the MAb 256A-4 and 256A-8.

[0036] FIG. 18 depicts a schematic of the epitope binding site for MAb 256A-4 and 256A-8.

DETAILED DESCRIPTION

[0037] This invention is not limited to the particular methodology, protocols, cell lines, vectors, or reagents described herein because they may vary. Further, the terminology used herein is for the purpose of describing particular embodiments only and is not intended to limit the scope of the present invention. As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise, e.g., reference to "a host cell" includes a plurality of such host cells. Unless defined otherwise, all technical and scientific terms and any acronyms used herein have the same meanings as commonly understood by one of ordinary skill in the art in the field of the invention. Although any methods and materials similar or equivalent to those described herein can be used in the practice of the present invention, the exemplary methods, devices, and materials are described herein.

[0038] All patents and publications mentioned herein are incorporated herein by reference to the extent allowed by law for the purpose of describing and disclosing the proteins, enzymes, vectors, host cells, and methodologies reported therein that might be used with the present invention. However, nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.

DEFINITIONS

[0039] Terms used throughout this application are to be construed with ordinary and typical meaning to those of ordinary skill in the art. However, Applicants desire that the following terms be given the particular definitions as defined below.

[0040] The phrase "substantially identical" with respect to an antibody chain polypeptide sequence may be construed as an antibody chain exhibiting at least 70%, or 80%, or 90%, or 95% sequence identity to the reference polypeptide sequence. The term with respect to a nucleic acid sequence may be construed as a sequence of nucleotides exhibiting at least about 85%, or 90%, or 95%, or 97% sequence identity to the reference nucleic acid sequence.

[0041] The term "identity" or "homology" shall be construed to mean the percentage of amino acid residues in the candidate sequence that are identical with the residue of a corresponding sequence to which it is compared, after aligning the sequences and introducing gaps, if necessary to achieve the maximum percent identity for the entire sequence, and not considering any conservative substitutions as part of the sequence identity. Neither N- nor C-terminal extensions nor insertions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are well known in the art. Sequence identity may be measured using sequence analysis software.

[0042] The term "antibody" is used in the broadest sense, and specifically covers monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, and multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired biological activity. Antibodies (Abs) and immunoglobulins (Igs) are glycoproteins having the same structural characteristics. While antibodies exhibit binding specificity to a specific target, immunoglobulins include both antibodies and other antibody-like molecules which lack target specificity. The antibodies of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass. Native antibodies and immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each heavy chain has at one end a variable domain (V.sub.H) followed by a number of constant domains. Each light chain has a variable domain at one end (V.sub.L) and a constant domain at its other end.

[0043] As used herein, "anti-Notch3 antibody" means an antibody which binds specifically to human Notch3 in such a manner so as to inhibit or substantially reduce the binding of Notch3 to its ligands or to inhibit Notch 3 signaling.

[0044] The term "variable" in the context of variable domain of antibodies, refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular target. However, the variability is not evenly distributed through the variable domains of antibodies. It is concentrated in three segments called complementarity determining regions (CDRs; i.e., CDR1, CDR2, and CDR3) also known as hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework (FR). The variable domains of native heavy and light chains each comprise four FR regions, largely a adopting a .beta.-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the .beta.-sheet structure. The CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the target binding site of antibodies (see Kabat, et al. Sequences of Proteins of Immunological Interest, National Institute of Health, Bethesda, Md. (1987)). As used herein, numbering of immunoglobulin amino acid residues is done according to the immunoglobulin amino acid residue numbering system of Kabat, et al., unless otherwise indicated.

[0045] The term "antibody fragment" refers to a portion of a full-length antibody, generally the target binding or variable region. Examples of antibody fragments include F(ab), F(ab'), F(ab').sub.2 and Fv fragments. The phrase "functional fragment or analog" of an antibody is a compound having qualitative biological activity in common with a full-length antibody. For example, a functional fragment or analog of an anti-Notch3 antibody is one which can bind to a Notch3 receptor in such a manner so as to prevent or substantially reduce the ability of the receptor to bind to its ligands or initiate signaling. As used herein, "functional fragment" with respect to antibodies, refers to Fv, F(ab) and F(ab').sub.2 fragments. An "Fv" fragment consists of a dimer of one heavy and one light chain variable domain in a tight, non-covalent association (V.sub.H-V.sub.L dimer). It is in this configuration that the three CDRs of each variable domain interact to define a target binding site on the surface of the V.sub.H-V.sub.L dimer. Collectively, the six CDRs confer target binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for a target) has the ability to recognize and bind target, although at a lower affinity than the entire binding site.

[0046] "Single-chain Fv" or "sFv" antibody fragments comprise the V.sub.H and V.sub.L domains of an antibody, wherein these domains are present in a single polypeptide chain. Generally, the Fv polypeptide further comprises a polypeptide linker between the V.sub.H and V.sub.L domains which enables the sFv to form the desired structure for target binding.

[0047] The term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy chain variable domain (V.sub.H) connected to a light chain variable domain (V.sub.L) in the same polypeptide chain. By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another changing and create two antigen-binding sites.

[0048] The F(ab) fragment contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain. F(ab') fragments differ from F(ab) fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region. F(ab') fragments are produced by cleavage of the disulfide bond at the hinge cysteines of the F(ab').sub.2 pepsin digestion product. Additional chemical couplings of antibody fragments are known to those of ordinary skill in the art.

[0049] The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison, et al., Proc Natl Acad Sci USA 81:6851 (1984)). Monoclonal antibodies are highly specific, being directed against a single target site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the target. In addition to their specificity, monoclonal antibodies are advantageous in that they may be synthesized by the hybridoma culture, uncontaminated by other immunoglobulins. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies for use with the present invention may be isolated from phage antibody libraries using well known techniques. The parent monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler, et al., Nature 256:495 (1975), or may be made by recombinant methods.

[0050] "Humanized" forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab').sub.2 or other target-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin template sequence. The humanized antibody may also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin template chosen.

[0051] The terms "cell," "cell line," and "cell culture" include progeny. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or biological property, as screened for in the originally transformed cell, are included. The "host cells" used in the present invention generally are prokaryotic or eukaryotic hosts.

[0052] "Transformation" of a cellular organism, cell, or cell line with DNA means introducing DNA into the target cell so that the DNA is replicable, either as an extrachromosomal element or by chromosomal integration. "Transfection" of a cell or organism with DNA refers to the taking up of DNA, e.g., an expression vector, by the cell or organism whether or not any coding sequences are in fact expressed. The terms "transfected host cell" and "transformed" refer to a cell in which DNA was introduced. The cell is termed "host cell" and it may be either prokaryotic or eukaryotic. Typical prokaryotic host cells include various strains of E. coli. Typical eukaryotic host cells are mammalian, such as Chinese hamster ovary or cells of human origin. The introduced DNA sequence may be from the same species as the host cell or a different species from the host cell, or it may be a hybrid DNA sequence, containing some foreign and some homologous DNA.

[0053] The term "vector" means a DNA construct containing a DNA sequence which is operably linked to a suitable control sequence capable of effecting the expression of the DNA sequence in a suitable host. Such control sequences include a promoter to effect transcription, an optional operator sequence to control such transcription, a sequence encoding suitable mRNA ribosome binding sites, and sequences which control the termination of transcription and translation. The vector may be a plasmid, a phage particle, or simply a potential genomic insert. Once transformed into a suitable host, the vector may replicate and function independently of the host genome, or may in some instances, integrate into the genome itself. In the present specification, "plasmid" and "vector" are sometimes used interchangeably, as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of vectors which serve equivalent function as and which are, or become, known in the art.

[0054] "Mammal" for purposes of treatment refers to any animal classified as a mammal, including human, domestic and farm animals, nonhuman primates, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, etc.

[0055] The word "label" when used herein refers to a detectable compound or composition which can be conjugated directly or indirectly to a molecule or protein, e.g., an antibody. The label may itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable.

[0056] As used herein, "solid phase" means a non-aqueous matrix to which the antibody of the present invention can adhere. Examples of solid phases encompassed herein include those formed partially or entirely of glass (e.g., controlled pore glass), polysaccharides (e.g., agarose), polyacrylamides, polystyrene, polyvinyl alcohol, and silicones. In certain embodiments, depending on the context, the solid phase can comprise the well of an assay plate; in others it is a purification column (e.g., an affinity chromatography column).

[0057] As used herein, the term "Notch3-mediated disorder" means a condition or disease which is characterized by the overexpression and/or hypersensitivity of the Notch3 receptor. Specifically it would be construed to include conditions associated with cancers such as non-small cell lung cancer, ovarian cancer, and T-cell acute lymphoblastic leukemia. Other cancers including pancreatic, prostate cancer, plasma cell neoplasms (e.g., multiple myeloma, plasma cell leukemia and extramedullary plasmacytoma), neuroblastoma and extramedullary plasmacytoma are also encompassed under the scope of this term. Other types of diseases include lymphoma, Alagille syndrome, liver disease involving aberrant vascularization, neurologic diseases, diabetes, diseases involving vascular cell fate, and rheumatoid arthritis.

Notch 3 Receptor Immunogen for Generating Antibodies

[0058] Soluble targets or fragments thereof can be used as immunogens for generating antibodies. The antibody is directed against the target of interest. Preferably, the target is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can result in a therapeutic benefit in that mammal. Whole cells may be used as the immunogen for making antibodies. The immunogen may be produced recombinantly or made using synthetic methods. The immunogen may also be isolated from a natural source.

[0059] For transmembrane molecules, such as receptors, fragments of these (e.g., the extracellular domain of a receptor) can be used as the immunogen. Alternatively, cells expressing the transmembrane molecule can be used as the immunogen. Such cells can be derived from a natural source (e.g., cancer cell lines) or may be cells which have been transformed by recombinant techniques to over-express the transmembrane molecule. Other forms of the immunogen useful for preparing antibodies will be apparent to those in the art.

[0060] Alternatively, a gene or a cDNA encoding human Notch3 receptor may be cloned into a plasmid or other expression vector and expressed in any of a number of expression systems according to methods well known to those of skill in the art. Methods of cloning and expressing Notch3 receptor and the nucleic acid sequence for human Notch3 receptor are known (see, for example, U.S. Pat. Nos. 5,821,332 and 5,759,546). Because of the degeneracy of the genetic code, a multitude of nucleotide sequences encoding Notch3 receptor protein or polypeptides may be used. One may vary the nucleotide sequence by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the nucleotide sequence that codes for naturally occurring Notch3 receptor and all such variations may be considered. Any one of these polypeptides may be used in the immunization of an animal to generate antibodies that bind to human Notch3 receptor.

[0061] Recombinant Notch3 proteins from other species may also be used as immunogen to generate antibodies because of the high degree of conservation of the amino acid sequence of Notch3. A comparison between human and mouse Notch3 showed over 90% amino acid sequence identity between the two species.

[0062] The immunogen Notch3 receptor may, when beneficial, be expressed as a fusion protein that has the Notch3 receptor attached to a fusion segment. The fusion segment often aids in protein purification, e.g., by permitting the fusion protein to be isolated and purified by affinity chromatography, but can also be used to increase immunogenicity. Fusion proteins can be produced by culturing a recombinant cell transformed with a fusion nucleic acid sequence that encodes a protein including the fusion segment attached to either the carboxyl and/or amino terminal end of the protein. Fusion segments may include, but are not limited to, immunoglobulin Fc regions, glutathione-S-transferase, .beta.-galactosidase, a poly-histidine segment capable of binding to a divalent metal ion, and maltose binding protein.

[0063] Recombinant Notch3 receptor protein as described in Example 1 was used to immunize mice to generate the hybridomas that produce the monoclonal antibodies of the present invention. Exemplary polypeptides comprise all or a portion of SEQ ID NO. 1 or variants thereof.

Antibody Generation

[0064] The antibodies of the present invention may be generated by any suitable method known in the art. The antibodies of the present invention may comprise polyclonal antibodies. Methods of preparing polyclonal antibodies are known to the skilled artisan (Harlow, et al., Antibodies: a Laboratory Manual, Cold spring Harbor Laboratory Press, 2nd ed. (1988), which is hereby incorporated herein by reference in its entirety).

[0065] For example, an immunogen as described in Example 1 may be administered to various host animals including, but not limited to, rabbits, mice, rats, etc., to induce the production of sera containing polyclonal antibodies specific for the antigen. The administration of the immunogen may entail one or more injections of an immunizing agent and, if desired, an adjuvant. Various adjuvants may be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum. Additional examples of adjuvants which may be employed include the MPL-TDM adjuvant (monophosphoryl lipid A, synthetic trehalose dicorynomycolate). Immunization protocols are well known in the art and may be performed by any method that elicits an immune response in the animal host chosen. Adjuvants are also well known in the art.

[0066] Typically, the immunogen (with or without adjuvant) is injected into the mammal by multiple subcutaneous or intraperitoneal injections, or intramuscularly or through IV. The immunogen may include a Notch3 polypeptide, a fusion protein, or variants thereof. Depending upon the nature of the polypeptides (i.e., percent hydrophobicity, percent hydrophilicity, stability, net charge, isoelectric point etc.), it may be useful to conjugate the immunogen to a protein known to be immunogenic in the mammal being immunized. Such conjugation includes either chemical conjugation by derivatizing active chemical functional groups to both the immunogen and the immunogenic protein to be conjugated such that a covalent bond is formed, or through fusion-protein based methodology, or other methods known to the skilled artisan. Examples of such immunogenic proteins include, but are not limited to, keyhole limpet hemocyanin, ovalbumin, serum albumin, bovine thyroglobulin, soybean trypsin inhibitor, and promiscuous T helper peptides. Various adjuvants may be used to increase the immunological response as described above.

[0067] The antibodies of the present invention comprise monoclonal antibodies. Monoclonal antibodies are antibodies which recognize a single antigenic site. Their uniform specificity makes monoclonal antibodies much more useful than polyclonal antibodies, which usually contain antibodies that recognize a variety of different antigenic sites. Monoclonal antibodies may be prepared using hybridoma technology, such as those described by Kohler, et al., Nature 256:495 (1975); U.S. Pat. No. 4,376,110; Harlow, et al., Antibodies: A Laboratory Manual, Cold spring Harbor Laboratory Press, 2nd ed. (1988) and Hammerling, et al., Monoclonal Antibodies and T-Cell Hybridomas, Elsevier (1981), recombinant DNA methods, or other methods known to the artisan. Other examples of methods which may be employed for producing monoclonal antibodies include, but are not limited to, the human B-cell hybridoma technique (Kosbor, et al., Immunology Today 4:72 (1983); Cole, et al., Proc Natl Acad Sci USA 80:2026 (1983)), and the EBV-hybridoma technique (Cole, et al., Monoclonal Antibodies and Cancer Therapy, pp. 77-96, Alan R. Liss (1985)). Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof. The hybridoma producing the mAb of this invention may be cultivated in vitro or in vivo.

[0068] In the hybridoma model, a host such as a mouse, a humanized mouse, a mouse with a human immune system, hamster, rabbit, camel, or any other appropriate host animal, is immunized to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, pp. 59-103 (1986)).

[0069] Generally, in making antibody-producing hybridomas, either peripheral blood lymphocytes ("PBLs") are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, pp. 59-103 (1986)). Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine or human origin. Typically, a rat or mouse myeloma cell line is employed. The hybridoma cells may be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), substances that prevent the growth of HGPRT-deficient cells.

[0070] Preferred immortalized cell lines are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. Among these myeloma cell lines are murine myeloma lines, such as those derived from the MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif., and SP2/0 or X63-Ag8-653 cells available from the American Type Culture Collection (ATCC), Manassas, Va., USA. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J Immunol 133:3001 (1984); Brodeur, et al., Monoclonal Antibody Production Techniques and Applications, Marcel Dekker, Inc, pp. 51-63 (1987)). The mouse myeloma cell line NSO may also be used (European Collection of Cell Cultures, Salisbury, Wilshire, UK).

[0071] The culture medium in which hybridoma cells are grown is assayed for production of monoclonal antibodies directed against Notch3. The binding specificity of monoclonal antibodies produced by hybridoma cells may be determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques are known in the art and within the skill of the artisan. The binding affinity of the monoclonal antibody to Notch3 can, for example, be determined by a Scatchard analysis (Munson, et al., Anal Biochem 107:220 (1980)).

[0072] After hybridoma cells are identified that produce antibodies of the desired specificity, affinity, and/or activity, the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, pp. 59-103 (1986)). Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium (D-MEM) or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal.

[0073] The monoclonal antibodies secreted by the subclones are suitably separated or isolated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-SEPHAROSE.RTM. affinity media, hydroxylaptite chromatography, gel exclusion chromatography, gel electrophoresis, dialysis, or affinity chromatography.

[0074] A variety of methods exist in the art for the production of monoclonal antibodies and thus, the invention is not limited to their sole production in hybridomas. For example, the monoclonal antibodies may be made by recombinant DNA methods, such as those described in U.S. Pat. No. 4,816,567. In this context, the term "monoclonal antibody" refers to an antibody derived from a single eukaryotic, phage, or prokaryotic clone. DNA encoding the monoclonal antibodies of the invention is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies, or such chains from human, humanized, or other sources) (Innis, et al. In PCR Protocols. A Guide to Methods and Applications, Academic (1990), Sanger, et al., Proc Natl Acad Sci 74:5463 (1977)). The hybridoma cells serve as a source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, NS0 cells, Simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also may be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc Natl Acad Sci USA 81:6851 (1984)) or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.

[0075] The antibodies may be monovalent antibodies. Methods for preparing monovalent antibodies are well known in the art. For example, one method involves recombinant expression of immunoglobulin light chain and modified heavy chain. The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy chain cross-linking. Alternatively, the relevant cysteine residues are substituted with another amino acid residue or are deleted so as to prevent cross-linking.

[0076] Antibody fragments which recognize specific epitopes may be generated by known techniques. Traditionally, these fragments were derived via proteolytic digestion of intact antibodies (see, e.g., Morimoto, et al., J Biochem Biophys Methods 24:107 (1992); Brennan, et al., Science 229:81 (1985)). For example, Fab and F(ab').sub.2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab').sub.2 fragments). F(ab').sub.2 fragments contain the variable region, the light chain constant region and the CH1 domain of the heavy chain. However, these fragments can now be produced directly by recombinant host ells. For example, the antibody fragments can be isolated from an antibody phage library. Alternatively, F(ab').sub.2--SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab').sub.2 fragments (Carter, et al., Bio/Technology 10:163 (1992). According to another approach, F(ab').sub.2 fragments can be isolated directly from recombinant host cell culture. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner. In other embodiments, the antibody of choice is a single chain Fv fragment (Fv) (PCT patent application WO 93/16185).

[0077] For some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use chimeric, humanized, or human antibodies. A chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi, et al., BioTechniques 4:214 (1986); Gillies, et al., J Immunol Methods 125:191 (1989); U.S. Pat. Nos. 5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entirety.

[0078] A humanized antibody is designed to have greater homology to a human immunoglobulin than animal-derived monoclonal antibodies. Humanization is a technique for making a chimeric antibody wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. Humanized antibodies are antibody molecules generated in a non-human species that bind the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework (FR) regions from a human immunoglobulin molecule. Often, framework residues in the human framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. See, e.g., U.S. Pat. No. 5,585,089; Riechmann, et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties. Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Pat. Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28:489 (1991); Studnicka, et al., Protein Engineering 7:805 (1994); Roguska, et al., Proc Natl Acad Sci USA 91:969 (1994)), and chain shuffling (U.S. Pat. No. 5,565,332).

[0079] Generally, a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. Humanization can be essentially performed following the methods of Winter and co-workers (Jones, et al., Nature 321:522 (1986); Riechmann, et al., Nature 332:323 (1988); Verhoeyen, et al., Science 239:1534 (1988)), by substituting non-human CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possible some FR residues are substituted from analogous sites in rodent antibodies.

[0080] It is further important that humanized antibodies retain high affinity for the antigen and other favorable biological properties. To achieve this goal, according to a preferred method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of certain residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin sequences, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is maximized, although it is the CDR residues that directly and most substantially influence antigen binding.

[0081] The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is important to reduce antigenicity. According to an exemplary method, the so-called "best-fit" method, the sequence of the variable domain of a non-human antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of that of the non-human parent antibody is then accepted as the human FR for the humanized antibody (Sims, et al., J Immunol 151:2296 (1993); Chothia, et al., J Mol Biol 196:901 (1987)). Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter, et al., Proc Natl Acad Sci USA 89:4285 (1992); Presta, et al., J Immunol 151:2623 (1993)).

[0082] Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also, U.S. Pat. Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety. The techniques of Cole, et al. and Boerder, et al. are also available for the preparation of human monoclonal antibodies (Cole, et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Riss (1985); and Boerner, et al., J Immunol 147:86 (1991)).

[0083] Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. For example, the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells. Alternatively, the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes. The mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production. The modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring which express human antibodies. See, e.g., Jakobovits, et al., Proc Natl Acad Sci USA 90:2551 (1993); Jakobovits, et al., Nature 362:255 (1993); Bruggermann, et al., Year in Immunol 7:33 (1993); Duchosal, et al., Nature 355:258 (1992)). The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention. Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies. For an overview of this technology for producing human antibodies, see Lonberg, et al., Int Rev Immunol 13:65-93 (1995). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., PCT publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0 598 877; U.S. Pat. Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825; 5,661,016; 5,545,806; 5,814,318; 5,885,793; 5,916,771; and 5,939,598, which are incorporated by reference herein in their entirety. In addition, companies such as Abgenix, Inc. (Freemont, Calif.), Genpharm (San Jose, Calif.), and Medarex, Inc. (Princeton, N.J.) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.

[0084] Also human mAbs could be made by immunizing mice transplanted with human peripheral blood leukocytes, splenocytes or bone marrows (e.g., Trioma techniques of XTL). Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection." In this approach a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope (Jespers, et al., Bio/technology 12:899 (1988)).

[0085] Further, antibodies to the polypeptides of the invention can, in turn, be utilized to generate anti-idiotype antibodies that "mimic" polypeptides of the invention using techniques well known to those skilled in the art (See, e.g., Greenspan, et al., FASEB J7:437 (1989); Nissinoff, J Immunol 147:2429 (1991)). For example, antibodies which bind to and competitively inhibit polypeptide multimerization and/or binding of a polypeptide of the invention to a ligand can be used to generate anti-idiotypes that "mimic" the polypeptide multimerization and/or binding domain and, as a consequence, bind to and neutralize polypeptide and/or its ligand. Such neutralizing anti-idiotypes or Fab fragments of such anti-idiotypes can be used in therapeutic regimens to neutralize polypeptide ligand. For example, such anti-idiotypic antibodies can be used to bind a polypeptide of the invention and/or to bind its ligands/receptors, and thereby block its biological activity.

[0086] The antibodies of the present invention may be bispecific antibodies. Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present invention, one of the binding specificities may be directed towards Notch3, the other may be for any other antigen, and preferably for a cell-surface protein, receptor, receptor subunit, tissue-specific antigen, virally derived protein, virally encoded envelope protein, bacterially derived protein, or bacterial surface protein, etc.

[0087] Methods for making bispecific antibodies are well known. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein, et al., Nature 305:537 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829 and in Traunecker, et al., EMBO J 10:3655 (1991).

[0088] Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It may have the first heavy-chain constant region (CH1) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transformed into a suitable host organism. For further details of generating bispecific antibodies see, for example Suresh, et al., Meth In Enzym 121:210 (1986).

[0089] Heteroconjugate antibodies are also contemplated by the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980). It is contemplated that the antibodies may be prepared in vitro using known methods in synthetic protein chemistry, including those involving cross-linking agents. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioester bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Pat. No. 4,676,980.

[0090] In addition, one can generate single-domain antibodies to Notch3. Examples of this technology have been described in WO94/25591 for antibodies derived from Camelidae heavy chain Ig, as well in US2003/0130496 describing the isolation of single domain fully human antibodies from phage libraries.

[0091] One can also create a single peptide chain binding molecules in which the heavy and light chain Fv regions are connected. Single chain antibodies ("scFv") and the method of their construction are described in U.S. Pat. No. 4,946,778. Alternatively, Fab can be constructed and expressed by similar means. All of the wholly and partially human antibodies are less immunogenic than wholly murine mAbs, and the fragments and single chain antibodies are also less immunogenic.

[0092] Antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty, et al., Nature 348:552 (1990). Clarkson, et al., Nature 352:624 (1991) and Marks, et al., J Mol Biol 222:581 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks, et al., Bio/Technology 10:779 (1992)), as well as combinatorial infection and in vivo recombination as a strategy for constructing very large phage libraries (Waterhouse, et al., Nuc Acids Res 21:2265 (1993)). Thus, these techniques are viable alternatives to traditional monoclonal antibody hybridoma techniques for isolation of monoclonal antibodies.

[0093] The DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc Natl Acad Sci USA 81:6851 (1984)).

[0094] Another alternative is to use electrical fusion rather than chemical fusion to form hybridomas. This technique is well established. Instead of fusion, one can also transform a B cell to make it immortal using, for example, an Epstein Barr Virus, or a transforming gene. See, e.g., "Continuously Proliferating Human Cell Lines Synthesizing Antibody of Predetermined Specificity," Zurawaki, et al., in Monoclonal Antibodies, ed. by Kennett, et al., Plenum Press, pp. 19-33. (1980)). Anti-Notch3 mAbs can be raised by immunizing rodents (e.g., mice, rats, hamsters, and guinea pigs) with Notch3 protein, fusion protein, or its fragments expressed by either eukaryotic or prokaryotic systems. Other animals can be used for immunization, e.g., non-human primates, transgenic mice expressing immunoglobulins, and severe combined immunodeficient (SCID) mice transplanted with human B lymphocytes. Hybridomas can be generated by conventional procedures by fusing B lymphocytes from the immunized animals with myeloma cells (e.g., Sp2/0 and NSO), as described earlier (Kohler, et al., Nature 256:495 (1975)). In addition, anti-Notch3 antibodies can be generated by screening of recombinant single-chain Fv or Fab libraries from human B lymphocytes in phage-display systems. The specificity of the mAbs to Notch3 can be tested by ELISA, Western immunoblotting, or other immunochemical techniques. The inhibitory activity of the antibodies on complement activation can be assessed by hemolytic assays, using sensitized chicken or sheep RBCs for the classical complement pathway. The hybridomas in the positive wells are cloned by limiting dilution. The antibodies are purified for characterization for specificity to human Notch3 by the assays described above.

Identification of Anti-Notch-3 Antibodies

[0095] The present invention provides antagonist monoclonal antibodies that inhibit and neutralize the action of Notch3. In particular, the antibodies of the present invention bind to and inhibit the activation of Notch3. The antibodies of the present invention include the antibodies designated 256A-4 and 256A-8, which are disclosed herein. The present invention also includes antibodies that bind to the same epitope as one of these antibodies.

[0096] Candidate anti-Notch3 antibodies were tested by enzyme linked immunosorbent assay (ELISA), Western immunoblotting, or other immunochemical techniques. Assays performed to characterize the individual antibodies are described in the Examples.

[0097] Antibodies of the invention include, but are not limited to, polyclonal, monoclonal, monovalent, bispecific, heteroconjugate, multispecific, human, humanized or chimeric antibodies, single chain antibodies, single-domain antibodies, Fab fragments, F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), and epitope-binding fragments of any of the above.

[0098] The antibodies may be human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab' and F(ab').sub.2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and single-domain antibodies comprising either a VL or VH domain. Antigen-binding antibody fragments, including single-chain antibodies, may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CH1, CH2, and CH3 domains. Also included in the invention are antigen-binding fragments comprising any combination of variable region(s) with a hinge region, CH1, CH2, and CH3 domains. The antibodies of the invention may be from any animal origin including birds and mammals. Preferably, the antibodies are from human, non-human primates, rodents (e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken.

[0099] As used herein, "human" antibodies" include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins, as described infra and, for example in, U.S. Pat. No. 5,939,598 by Kucherlapati, et al.

[0100] The antibodies of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopes of Notch3 or may be specific for both Notch3 as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J Immunol 147:60 (1991); U.S. Pat. Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920; 5,601,819; Kostelny, et al., J Immunol 148:1547 (1992).

[0101] Antibodies of the present invention may be described or specified in terms of the epitope(s) or portion(s) of Notch3 which they recognize or specifically bind. The epitope(s) or polypeptide portion(s) may be specified as described herein, e.g., by N-terminal and C-terminal positions, by size in contiguous amino acid residues, or listed in the Tables and Figures.

[0102] Antibodies of the present invention may also be described or specified in terms of their cross-reactivity. Antibodies that bind Notch3 polypeptides, which have at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using methods known in the art and described herein) to Notch3 are also included in the present invention. Anti-Notch3 antibodies may also bind with a K.sub.D of less than about 10.sup.-7 M, less than about 10.sup.-6 M, or less than about 10.sup.-5 M to other proteins, such as anti-Notch3 antibodies from species other than that against which the anti-Notch3 antibody is directed.

[0103] In specific embodiments, antibodies of the present invention cross-react with monkey homologues of human Notch3 and the corresponding epitopes thereof. In a specific embodiment, the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of the specific antigenic and/or immunogenic polypeptides disclosed herein.

[0104] Further included in the present invention are antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide encoding Notch3 under stringent hybridization conditions. Antibodies of the present invention may also be described or specified in terms of their binding affinity to a polypeptide of the invention. Preferred binding affinities include those with an equilibrium dissociation constant or K.sub.D from 10.sup.-8 to 10.sup.-15 M, 10.sup.-8 to 10.sup.-12 M, 10.sup.-8 to 10.sup.-10 M, or 10.sup.-10 to 10.sup.-12 M. The invention also provides antibodies that competitively inhibit binding of an antibody to an epitope of the invention as determined by any method known in the art for determining competitive binding, for example, the immunoassays described herein. In preferred embodiments, the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.

Vectors and Host Cells

[0105] In another aspect, the present invention provides isolated nucleic acid sequences encoding an antibody as disclosed herein, vector constructs comprising a nucleotide sequence encoding the antibodies of the present invention, host cells comprising such a vector, and recombinant techniques for the production of the antibody.

[0106] For recombinant production of an antibody, the nucleic acid encoding it is isolated and inserted into a replicable vector for further cloning (amplification of the DNA) or for expression. DNA encoding the antibody is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody). Standard techniques for cloning and transformation may be used in the preparation of cell lines expressing the antibodies of the present invention.

[0107] Vectors

[0108] Many vectors are available. The vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence. Recombinant expression vectors containing a nucleotide sequence encoding the antibodies of the present invention can be prepared using well known techniques. The expression vectors include a nucleotide sequence operably linked to suitable transcriptional or translational regulatory nucleotide sequences such as those derived from mammalian, microbial, viral, or insect genes. Examples of regulatory sequences include transcriptional promoters, operators, enhancers, mRNA ribosomal binding sites, and/or other appropriate sequences which control transcription and translation initiation and termination. Nucleotide sequences are "operably linked" when the regulatory sequence functionally relates to the nucleotide sequence for the appropriate polypeptide. Thus, a promoter nucleotide sequence is operably linked to, e.g., the antibody heavy chain sequence if the promoter nucleotide sequence controls the transcription of the appropriate nucleotide sequence.

[0109] In addition, sequences encoding appropriate signal peptides that are not naturally associated with antibody heavy and/or light chain sequences can be incorporated into expression vectors. For example, a nucleotide sequence for a signal peptide (secretory leader) may be fused in-frame to the polypeptide sequence so that the antibody is secreted to the periplasmic space or into the medium. A signal peptide that is functional in the intended host cells enhances extracellular secretion of the appropriate antibody. The signal peptide may be cleaved from the polypeptide upon secretion of antibody from the cell. Examples of such secretory signals are well known and include, e.g., those described in U.S. Pat. Nos. 5,698,435; 5,698,417; and 6,204,023.

[0110] The vector may be a plasmid vector, a single or double-stranded phage vector, or a single or double-stranded RNA or DNA viral vector. Such vectors may be introduced into cells as polynucleotides by well known techniques for introducing DNA and RNA into cells. The vectors, in the case of phage and viral vectors also may be introduced into cells as packaged or encapsulated virus by well known techniques for infection and transduction. Viral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells. Cell-free translation systems may also be employed to produce the protein using RNAs derived from the present DNA constructs. Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publications WO 86/05807 and WO 89/01036; and U.S. Pat. No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.

[0111] Host Cells

[0112] The antibodies of the present invention can be expressed from any suitable host cell. Examples of host cells useful in the present invention include prokaryotic, yeast, or higher eukaryotic cells and include but are not limited to microorganisms such as bacteria (e.g., E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., Baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).

[0113] Prokaryotes useful as host cells in the present invention include gram negative or gram positive organisms such as E. coli, B. subtilis, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, Serratia, and Shigella, as well as Bacilli, Pseudomonas, and Streptomyces. One preferred E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.

[0114] Expression vectors for use in prokaryotic host cells generally comprise one or more phenotypic selectable marker genes. A phenotypic selectable marker gene is, for example, a gene encoding a protein that confers antibiotic resistance or that supplies an autotrophic requirement. Examples of useful expression vectors for prokaryotic host cells include those derived from commercially available plasmids such as the pKK223-3 vector (Pharmacia Fine Chemicals, Uppsala, Sweden), PGEM.RTM.1 vector (Promega Biotec, Madison, Wis., USA), and the pET (Novagen, Madison, Wis., USA) and pRSET (Invitrogen, Carlsbad, Calif.) series of vectors (Studier, J Mol Biol 219:37 (1991); Schoepfer, Gene 124:83 (1993)). Promoter sequences commonly used for recombinant prokaryotic host cell expression vectors include T7, (Rosenberg, et al., Gene 56:125 (1987)), .beta.-lactamase (penicillinase), lactose promoter system (Chang, et al., Nature 275:615 (1978); Goeddel, et al., Nature 281:544 (1979)), tryptophan (trp) promoter system (Goeddel, et al., Nucl Acids Res 8:4057 (1980)), and tac promoter (Sambrook, et al., Molecular Cloning, A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory (1990)).

[0115] Yeasts or filamentous fungi useful in the present invention include those from the genus Saccharomyces, Pichia, Actinomycetes, Kluyveromyces, Schizosaccharomyces, Candida, Trichoderma, Neurospora, and filamentous fungi such as Neurospora, Penicillium, Tolypocladium, and Aspergillus. Yeast vectors will often contain an origin of replication sequence from a 2.mu. yeast plasmid, an autonomously replicating sequence (ARS), a promoter region, sequences for polyadenylation, sequences for transcription termination, and a selectable marker gene. Suitable promoter sequences for yeast vectors include, among others, promoters for metallothionein, 3-phosphoglycerate kinase (Hitzeman, et al., J Biol Chem 255:2073 (1980)) or other glycolytic enzymes (Holland, et al., Biochem 17:4900 (1978)) such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. Other suitable vectors and promoters for use in yeast expression are further described in Fleer, et al., Gene 107:285 (1991). Other suitable promoters and vectors for yeast and yeast transformation protocols are well known in the art. Yeast transformation protocols are well known. One such protocol is described by Hinnen, et al., Proc Natl Acad Sci 75:1929 (1978). The Hinnen protocol selects for Trp.sup.+ transformants in a selective medium.

[0116] Mammalian or insect host cell culture systems may also be employed to express recombinant antibodies. In principle, any higher eukaryotic cell culture is workable, whether from vertebrate or invertebrate culture. Examples of invertebrate cells include plant and insect cells (Luckow, et al., Bio/Technology 6:47 (1988); Miller, et al., Genetics Engineering, Setlow, et al., eds. Vol. 8, pp. 277-9, Plenam Publishing (1986); Mseda, et al., Nature 315:592 (1985)). For example, Baculovirus systems may be used for production of heterologous proteins. In an insect system,--Autographa californica nuclear polyhedrosis virus (AcNPV) may be used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter). Other hosts that have been identified include Aedes, Drosophila melanogaster, and Bombyx mori. A variety of viral strains for transfection are publicly available, e.g., the L-1 variant of AcNPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells. Moreover, plant cells cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco and also be utilized as hosts.

[0117] Vertebrate cells, and propagation of vertebrate cells, in culture (tissue culture) has become a routine procedure. See Tissue Culture, Kruse, et al., eds., Academic Press (1973). Examples of useful mammalian host cell lines are monkey kidney; human embryonic kidney line; baby hamster kidney cells; Chinese hamster ovary cells/-DHFR (CHO, Urlaub, et al., Proc Natl Acad Sci USA 77:4216 (1980)); mouse sertoli cells; human cervical carcinoma cells (HELA); canine kidney cells; human lung cells; human liver cells; mouse mammary tumor; and NS0 cells.

[0118] Host cells are transformed with the above-described vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, transcriptional and translational control sequences, selecting transformants, or amplifying the genes encoding the desired sequences. Commonly used promoter sequences and enhancer sequences are derived from polyoma virus, Adenovirus 2, Simian virus 40 (SV40), and human cytomegalovirus (CMV). DNA sequences derived from the SV40 viral genome may be used to provide other genetic elements for expression of a structural gene sequence in a mammalian host cell, e.g., SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites. Viral early and late promoters are particularly useful because both are easily obtained from a viral genome as a fragment which may also contain a viral origin of replication. Exemplary expression vectors for use in mammalian host cells are commercially available.

[0119] The host cells used to produce an antibody of this invention may be cultured in a variety of media. Commercially available media such as Ham's F10 (Sigma, St Louis, Mo.), Minimal Essential Medium (MEM, Sigma, St Louis, Mo.), RPMI-1640 (Sigma, St Louis, Mo.), and Dulbecco's Modified Eagle's Medium (DMEM, Sigma, St Louis, Mo.) are suitable for culturing host cells. In addition, any of the media described in Ham, et al., Meth Enzymol 58:44 (1979), Barnes, et al., Anal Biochem 102:255 (1980), and U.S. Pat. No. 4,767,704; 4,657,866; 4,560,655; 5,122,469; 5,712,163; or 6,048,728 may be used as culture media for the host cells. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as X-chlorides, where X is sodium, calcium, magnesium; and phosphates), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as gentamicin), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.

Polynucleotides Encoding Antibodies

[0120] The invention further provides polynucleotides or nucleic acids, e.g., DNA, comprising a nucleotide sequence encoding an antibody of the invention and fragments thereof. Exemplary polynucleotides include those encoding antibody chains comprising one or more of the amino acid sequences described herein. The invention also encompasses polynucleotides that hybridize under stringent or lower stringency hybridization conditions to polynucleotides that encode an antibody of the present invention.

[0121] The polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. For example, if the nucleotide sequence of the antibody is known, a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier, et al., Bio/Techniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.

[0122] Alternatively, a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A.sup.+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody of the invention) by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art.

[0123] Once the nucleotide sequence and corresponding amino acid sequence of the antibody is determined, the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook, et al., Molecular Cloning, A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory (1990); Ausubel, et al., eds., Current Protocols in Molecular Biology, John Wiley & Sons (1998), which are both incorporated by reference herein in their entireties), to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, and/or insertions.

[0124] In a specific embodiment, the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of the CDRs by well known methods, e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine the regions of sequence hypervariability. Using routine recombinant DNA techniques, one or more of the CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody, as described supra. The framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions (see, e.g., Chothia, et al., J Mol Biol 278: 457 (1998) for a listing of human framework regions). Preferably, the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention. Preferably, as discussed supra, one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibody to its antigen. Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable region cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds. Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art.

[0125] In addition, techniques developed for the production of "chimeric antibodies" (Morrison, et al., Proc Natl Acad Sci 81:851 (1984); Neuberger, et al., Nature 312:604 (1984); Takeda, et al., Nature 314:452 (1985)) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. As described supra, a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region, e.g., humanized antibodies.

[0126] Alternatively, techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778; Bird, Science 242:423 (1988); Huston, et al., Proc Natl Acad Sci USA 85:5879 (1988); and Ward, et al., Nature 334:544 (1989)) can be adapted to produce single chain antibodies. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E. coli may also be used (Skerra, et al., Science 242:1038 (1988)).

Methods of Producing Anti-Notch3 Antibodies

[0127] The antibodies of the invention can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques.

[0128] Recombinant expression of an antibody of the invention, or fragment, derivative, or analog thereof, (e.g., a heavy or light chain of an antibody of the invention or a single chain antibody of the invention), requires construction of an expression vector containing a polynucleotide that encodes the antibody or a fragment of the antibody. Once a polynucleotide encoding an antibody molecule has been obtained, the vector for the production of the antibody may be produced by recombinant DNA technology. An expression vector is constructed containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.

[0129] The expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody of the invention. In one aspect of the invention, vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.

[0130] A variety of host-expression vector systems may be utilized to express the antibody molecules of the invention as described above. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ. Bacterial cells such as E. coli, and eukaryotic cells are commonly used for the expression of a recombinant antibody molecule, especially for the expression of whole recombinant antibody molecule. For example, mammalian cells such as CHO, in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus, are an effective expression system for antibodies (Foecking, et al., Gene 45:101 (1986); Cockett, et al., Bio/Technology 8:2 (1990)).

[0131] In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include, but are not limited to, CHO, COS, 293, 3T3, or myeloma cells.

[0132] For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the antibody molecule may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for one to two days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the antibody molecule. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.

[0133] A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler, et al., Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska, et al., Proc Natl Acad Sci USA 48:202 (1992)), and adenine phosphoribosyltransferase (Lowy, et al., Cell 22:817 (1980)) genes can be employed in tk, hgprt or aprt-cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler, et al., Proc Natl Acad Sci USA 77:357 (1980); O'Hare, et al., Proc Natl Acad Sci USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan, et al., Proc Natl Acad Sci USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside G-418 (Wu, et al., Biotherapy 3:87 (1991)); and hygro, which confers resistance to hygromycin (Santerre, et al., Gene 30:147 (1984)). Methods commonly known in the art of recombinant DNA technology may be routinely applied to select the desired recombinant clone, and such methods are described, for example, in Ausubel, et al., eds., Current Protocols in Molecular Biology, John Wiley & Sons (1993); Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press (1990); and in Chapters 12 and 13, Dracopoli, et al., eds, Current Protocols in Human Genetics, John Wiley & Sons (1994); Colberre-Garapin, et al., J Mol Biol 150:1 (1981), which are incorporated by reference herein in their entireties.

[0134] The expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington, et al., "The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells," DNA Cloning, Vol. 3. Academic Press (1987)). When a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse, et al., Mol Cell Biol 3:257 (1983)).

[0135] The host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides. Alternatively, a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc Natl Acad Sci USA 77:2197 (1980)). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.

[0136] Once an antibody molecule of the invention has been produced by an animal, chemically synthesized, or recombinantly expressed, it may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and size-exclusion chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. In addition, the antibodies of the present invention or fragments thereof can be fused to heterologous polypeptide sequences described herein or otherwise known in the art, to facilitate purification.

[0137] The present invention encompasses antibodies recombinantly fused or chemically conjugated (including both covalently and non-covalently conjugations) to a polypeptide. Fused or conjugated antibodies of the present invention may be used for ease in purification. See e.g., PCT publication WO 93/21232; EP 439,095; Naramura, et al., Immunol Lett 39:91 (1994); U.S. Pat. No. 5,474,981; Gillies, et al., Proc Natl Acad Sci USA 89:1428 (1992); Fell, et al., J Immunol 146:2446 (1991), which are incorporated by reference in their entireties.

[0138] Moreover, the antibodies or fragments thereof of the present invention can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., Valencia, Calif.), among others, many of which are commercially available. As described in Gentz, et al., Proc Natl Acad Sci USA 86:821 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, et al., Cell 37:767 (1984)) and the "flag" tag.

Antibody Purification

[0139] When using recombinant techniques, an antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. If the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed fragments, may be removed, for example, by centrifugation or ultrafiltration. Carter, et al., Bio/Technology 10:163 (1992) describe a procedure for isolating antibodies which are secreted to the periplasmic space of E. coli. Briefly, cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonylfluoride (PMSF) over about 30 minutes. Cell debris can be removed by centrifugation. Where the antibody is secreted into the medium, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. A protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.

[0140] The antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel elecrophoresis, dialysis, and affinity chromatography, with affinity chromatography being the preferred purification technique. The suitability of protein A as an affinity ligand depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody. Protein A can be used to purify antibodies that are based on human IgG1, IgG2 or IgG4 heavy chains (Lindmark, et al., J Immunol Meth 62:1 (1983)). Protein G is recommended for all mouse isotypes and for human IgG3 (Guss, et al., EMBO J 5:1567 (1986)). The matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the antibody comprises a CH3 domain, the Bakerbond ABX.TM. resin (J. T. Baker; Phillipsburg, N.J.) is useful for purification. Other techniques for protein purification such as fractionation on an ion-exchange column, ethanol precipitation, Reverse Phase HPLC, chromatography on silica, chromatography on heparin SEPHAROSE.RTM. chromatography on an anion or cation exchange resin (such as a polyaspartic acid column), chromatofocusing, SDS-PAGE, and ammonium sulfate precipitation are also available depending on the antibody to be recovered.

[0141] Following any preliminary purification step(s), the mixture comprising the antibody of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, preferably performed at low salt concentrations (e.g., from about 0-0.25M salt).

Pharmaceutical Formulation

[0142] Therapeutic formulations of the polypeptide or antibody may be prepared for storage as lyophilized formulations or aqueous solutions by mixing the polypeptide having the desired degree of purity with optional "pharmaceutically-acceptable" carriers, excipients or stabilizers typically employed in the art (all of which are termed "excipients"), i.e., buffering agents, stabilizing agents, preservatives, isotonifiers, non-ionic detergents, antioxidants, and other miscellaneous additives. See Remington's Pharmaceutical Sciences, 16th edition, Osol, Ed. (1980). Such additives must be nontoxic to the recipients at the dosages and concentrations employed.

[0143] Buffering agents help to maintain the pH in the range which approximates physiological conditions. They are preferably present at concentration ranging from about 2 mM to about 50 mM. Suitable buffering agents for use with the present invention include both organic and inorganic acids and salts thereof such as citrate buffers (e.g., monosodium citrate-disodium citrate mixture, citric acid-trisodium citrate mixture, citric acid-monosodium citrate mixture, etc.), succinate buffers (e.g., succinic acid-monosodium succinate mixture, succinic acid-sodium hydroxide mixture, succinic acid-disodium succinate mixture, etc.), tartrate buffers (e.g., tartaric acid-sodium tartrate mixture, tartaric acid-potassium tartrate mixture, tartaric acid-sodium hydroxide mixture, etc.), fumarate buffers (e.g., fumaric acid-monosodium fumarate mixture, fumaric acid-disodium fumarate mixture, monosodium fumarate-disodium fumarate mixture, etc.), gluconate buffers (e.g., gluconic acid-sodium glyconate mixture, gluconic acid-sodium hydroxide mixture, gluconic acid-potassium glyconate mixture, etc.), oxalate buffer (e.g., oxalic acid-sodium oxalate mixture, oxalic acid-sodium hydroxide mixture, oxalic acid-potassium oxalate mixture, etc.), lactate buffers (e.g., lactic acid-sodium lactate mixture, lactic acid-sodium hydroxide mixture, lactic acid-potassium lactate mixture, etc.) and acetate buffers (e.g., acetic acid-sodium acetate mixture, acetic acid-sodium hydroxide mixture, etc.). Additionally, there may be mentioned phosphate buffers, histidine buffers and trimethylamine salts such as Tris.

[0144] Preservatives may be added to retard microbial growth, and may be added in amounts ranging from 0.2%-1% (w/v). Suitable preservatives for use with the present invention include phenol, benzyl alcohol, meta-cresol, methyl paraben, propyl paraben, octadecyldimethylbenzyl ammonium chloride, benzalconium halides (e.g., chloride, bromide, iodide), hexamethonium chloride, and alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, and 3-pentanol.

[0145] Isotonicifiers sometimes known as "stabilizers" may be added to ensure isotonicity of liquid compositions of the present invention and include polhydric sugar alcohols, preferably trihydric or higher sugar alcohols, such as glycerin, erythritol, arabitol, xylitol, sorbitol and mannitol.

[0146] Stabilizers refer to a broad category of excipients which can range in function from a bulking agent to an additive which solubilizes the therapeutic agent or helps to prevent denaturation or adherence to the container wall. Typical stabilizers can be polyhydric sugar alcohols (enumerated above); amino acids such as arginine, lysine, glycine, glutamine, asparagine, histidine, alanine, ornithine, L-leucine, 2-phenylalanine, glutamic acid, threonine, etc.; organic sugars or sugar alcohols, such as lactose, trehalose, stachyose, mannitol, sorbitol, xylitol, ribitol, myoinisitol, galactitol, glycerol and the like, including cyclitols such as inositol; polyethylene glycol; amino acid polymers; sulfur containing reducing agents, such as urea, glutathione, thioctic acid, sodium thioglycolate, thioglycerol, alpha.-monothioglycerol and sodium thio sulfate; low molecular weight polypeptides (i.e. <10 residues); proteins such as human serum albumin, bovine serum albumin, gelatin or immunoglobulins; hydrophylic polymers, such as polyvinylpyrrolidone monosaccharides, such as xylose, mannose, fructose, glucose; disaccharides such as lactose, maltose, sucrose and trisaccacharides such as raffinose; and polysaccharides such as dextran. Stabilizers may be present in the range from 0.1 to 10,000 weights per part of weight active protein.

[0147] Non-ionic surfactants or detergents (also known as "wetting agents") may be added to help solubilize the therapeutic agent as well as to protect the therapeutic protein against agitation-induced aggregation, which also permits the formulation to be exposed to shear surface stressed without causing denaturation of the protein. Suitable non-ionic surfactants include polysorbates (20, 80, etc.), polyoxamers (184, 188 etc.), PLURONIC.RTM. polyols, polyoxyethylene sorbitan monoethers (TWEEN-20.RTM., TWEEN-80.RTM., etc.). Non-ionic surfactants may be present in a range of about 0.05 mg/ml to about 1.0 mg/ml, preferably about 0.07 mg/ml to about 0.2 mg/ml.

[0148] Additional miscellaneous excipients include bulking agents, (e.g., starch), chelating agents (e.g., EDTA), antioxidants (e.g., ascorbic acid, methionine, vitamin E), and cosolvents. The formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. For example, it may be desirable to further provide an immunosuppressive agent. Such molecules are suitably present in combination in amounts that are effective for the purpose intended. The active ingredients may also be entrapped in microcapsule prepared, for example, by coascervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin micropheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences, 16th edition, Osal, Ed. (1980).

[0149] The formulations to be used for in vivo administration should be sterile. This is readily accomplished, for example, by filtration through sterile filtration membranes. Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT.TM. (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37.degree. C. resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S--S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.

[0150] The amount of therapeutic polypeptide, antibody, or fragment thereof which will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques. Where possible, it is desirable to determine the dose-response curve and the pharmaceutical compositions of the invention first in vitro, and then in useful animal model systems prior to testing in humans.

[0151] In a preferred embodiment, an aqueous solution of therapeutic polypeptide, antibody or fragment thereof is administered by subcutaneous injection. Each dose may range from about 0.5 .mu.g to about 50 .mu.g per kilogram of body weight, or more preferably, from about 3 .mu.g to about 30 .mu.g per kilogram body weight.

[0152] The dosing schedule for subcutaneous administration may vary from once a month to daily depending on a number of clinical factors, including the type of disease, severity of disease, and the subject's sensitivity to the therapeutic agent.

Therapeutic Uses of Anti-Notch-3 Antibodies

[0153] It is contemplated that the antibodies of the present invention may be used to treat a mammal. In one embodiment, the antibody is administered to a nonhuman mammal for the purposes of obtaining preclinical data, for example. Exemplary nonhuman mammals to be treated include nonhuman primates, dogs, cats, rodents and other mammals in which preclinical studies are performed. Such mammals may be established animal models for a disease to be treated with the antibody or may be used to study toxicity of the antibody of interest. In each of these embodiments, dose escalation studies may be performed on the mammal.

[0154] An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) can be used as a therapeutic. The present invention is directed to antibody-based therapies which involve administering antibodies of the invention to an animal, a mammal, or a human, for treating a Notch3-mediated disease, disorder, or condition. The animal or subject may be a mammal in need of a particular treatment, such as a mammal having been diagnosed with a particular disorder, e.g., one relating to Notch3. Antibodies directed against Notch3 are useful against cancer and other Notch3-associated diseases including neurological disorders, diabetes, rheumatoid arthritis, vascular related diseases, and Alagille symdrome in mammals, including but not limited to cows, pigs, horses, chickens, cats, dogs, non-human primates etc., as well as humans. For example, by administering a therapeutically acceptable dose of an anti-Notch3 antibody, or antibodies, of the present invention, or a cocktail of the present antibodies, or in combination with other antibodies of varying sources, disease symptoms may be ameliorated or prevented in the treated mammal, particularly humans.

[0155] Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (including fragments, analogs and derivatives thereof as described herein) and nucleic acids encoding antibodies of the invention as described below (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein). The antibodies of the invention can be used to treat, inhibit, or prevent diseases, disorders, or conditions associated with aberrant expression and/or activity of Notch3, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein. The treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of Notch3 includes, but is not limited to, alleviating at least one symptom associated with those diseases, disorders, or conditions. Antibodies of the invention may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.

[0156] Anti-Notch3 antibodies of the present invention may be used therapeutically in a variety of diseases. The present invention provides a method for preventing or treating Notch3-mediated diseases in a mammal. The method comprises administering a disease preventing or treating amount of anti-Notch3 antibody to the mammal. The anti-Notch3 antibody binds to Notch3 and antagonizes its function. Notch3 signaling has been linked to various diseases such as various cancers (Haruki, et al., Cancer Res 65:3555 (2005); Park, et al., Cancer Res 66:6312 (2006); Lu, et al., Clin Cancer Res 10:3291 (2004)); Hedvat, et al., Br J Haematol 122:728 (2003); Buchler, et al., Ann Surg 242:791 (2005)); Bellavia, et al., Proc Natl Acad Sci USA 99:3788 (2002); Screpanti, et al., Trends Mol Med 9:30 (2003)); van Limpt, et al., Cancer Lett 228:59 (2005)), neurological disorders (Joutel, et al., Nature 383:707 (1996)), diabetes (Anastasi, et al., J Immunol 171:4504 (2003), rheumatoid arthritis (Yabe, et al., J Orthop Sci 10:589 (2005)), vascular related diseases (Sweeney, et al., FASEB J 18:1421 (2004)), and Alagille syndrome (Flynn, et al., J Pathol 204:55 (2004)). Anti-Notch3 antibodies will also be effective to prevent the above mentioned diseases.

[0157] The amount of the antibody which will be effective in the treatment, inhibition, and prevention of a disease or disorder associated with aberrant expression and/or activity of Notch3 can be determined by standard clinical techniques. The dosage will depend on the type of disease to be treated, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The antibody can be administered in treatment regimes consistent with the disease, e.g., a single or a few doses over one to several days to ameliorate a disease state or periodic doses over an extended time to inhibit disease progression and prevent disease recurrence. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.

[0158] For antibodies, the dosage administered to a patient is typically 0.1 mg/kg to 150 mg/kg of the patient's body weight. Preferably, the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body weight. Generally, human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible. Further, the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.

[0159] The antibody composition will be formulated, dosed and administered in a manner consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The "therapeutically effective amount" of the antibody to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat a disease or disorder. The antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as used hereinbefore or about from 1 to 99% of the heretofore employed dosages.

[0160] The antibodies of the invention may be administered alone or in combination with other types of cancer treatments including conventional chemotherapeutic agents (paclitaxel, carboplatin, cisplatin and doxorbicin), anti-EGFR agents (gefitinib, erlotinib and cetuximab), anti-angiogenesis agents (bevacizumab and sunitinib), as well as immuno-modulating agents such as interferon-.alpha. and thalidomide.

[0161] In a preferred aspect, the antibody is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).

[0162] Various delivery systems are known and can be used to administer an antibody of the present invention, including injection, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu, et al., J Biol Chem 262:4429 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc.

[0163] The anti-Notch3 antibody can be administered to the mammal in any acceptable manner. Methods of introduction include but are not limited to parenteral, subcutaneous, intraperitoneal, intrapulmonary, intranasal, epidural, inhalation, and oral routes, and if desired for immunosuppressive treatment, intralesional administration. Parenteral infusions include intramuscular, intradermal, intravenous, intraarterial, or intraperitoneal administration. The antibodies or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, it may be desirable to introduce the therapeutic antibodies or compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. In addition, the antibody is suitably administered by pulse infusion, particularly with declining doses of the antibody. Preferably the dosing is given by injections, most preferably intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.

[0164] Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. The antibody may also be administered into the lungs of a patient in the form of a dry powder composition (See e.g., U.S. Pat. No. 6,514,496).

[0165] In a specific embodiment, it may be desirable to administer the therapeutic antibodies or compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, topical application, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering an antibody of the invention, care must be taken to use materials to which the protein does not absorb.

[0166] In another embodiment, the antibody can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527 (1990); Treat, et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein, et al., eds., pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-27; see generally ibid.).

[0167] In yet another embodiment, the antibody can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, Science 249:1527 (1990); Sefton, CRC Crit. Ref Biomed Eng 14:201 (1987); Buchwald, et al., Surgery 88:507 (1980); Saudek, et al., N Engl J Med 321:574 (1989)). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer, et al., eds., CRC Press (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen, et al., eds., Wiley (1984); Ranger, et al., J Macromol Sci Rev Macromol Chem 23:61 (1983); see also Levy, et al., Science 228:190 (1985); During, et al., Ann Neurol 25:351 (1989); Howard, et al., J Neurosurg 71:105 (1989)). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target.

[0168] The present invention also provides pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of the antibody and a physiologically acceptable carrier. In a specific embodiment, the term "physiologically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such physiological carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin. Such compositions will contain an effective amount of the antibody, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.

[0169] In one embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.

[0170] The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.

[0171] In addition, the antibodies of the present invention may be conjugated to various effector molecules such as heterologous polypeptides, drugs, radionucleotides, or toxins. See, e.g., PCT publications WO 92/08495; WO 91/14438; WO 89/12624; U.S. Pat. No. 5,314,995; and EP 396,387. An antibody or fragment thereof may be conjugated to a therapeutic moiety such as a cytotoxin (e.g., a cytostatic or cytocidal agent), a therapeutic agent, or a radioactive metal ion (e.g., alpha-emitters such as, for example, 213Bi). A cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologues thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).

[0172] Techniques for conjugating such therapeutic moieties to antibodies are well known, see, e.g., Amon, et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies and Cancer Therapy, Reisfeld, et al. (eds.), pp. 243-56 Alan R. Liss (1985); Hellstrom, et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery, 2nd ed., Robinson, et al., eds., pp. 623-53, Marcel Dekker (1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review," in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera, et al., eds., pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy," in Monoclonal Antibodies For Cancer Detection and Therapy, Baldwin, et al., eds., pp. 303-16, Academic Press (1985); and Thorpe, et al., Immunol Rev 62:119 (1982). Alternatively, an antibody can be conjugated to a second antibody to form an antibody heteroconjugate. See, e.g., U.S. Pat. No. 4,676,980.

[0173] The conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, .alpha.-interferon, .beta.-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-.alpha., TNF-.beta., AIM I (See, International Publication No. WO 97/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi, et al., Int Immunol, 6:1567 (1994)), VEGI (See, International Publication No. WO 99/23105); a thrombotic agent; an anti-angiogenic agent, e.g., angiostatin or endostatin; or biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-1"), interleukin-2 ("IL-2"), interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growth factors.

Articles of Manufacture

[0174] In another embodiment of the invention, an article of manufacture containing materials useful for the treatment of the disorders described above is provided. The article of manufacture comprises a container and a label. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is effective for preventing or treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The active agent in the composition is the antibody. The label on, or associated with, the container indicates that the composition is used for treating the condition of choice. The article of manufacture may further comprise a second container comprising a pharmaceutically acceptable buffer, such as phosphate-buffered saline, Ringer's solution, and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.

Antibody-Based Gene Therapy

[0175] In a another aspect of the invention, nucleic acids comprising sequences encoding antibodies or functional derivatives thereof, are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of Notch3, by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid. In this embodiment of the invention, the nucleic acids produce their encoded protein that mediates a therapeutic effect. Any of the methods for gene therapy available can be used according to the present invention. Exemplary methods are described below.

[0176] For general reviews of the methods of gene therapy, see Goldspiel, et al., Clinical Pharmacy 12:488 (1993); Wu, et al., Biotherapy 3:87 (1991); Tolstoshev, Ann Rev Pharmacol Toxicol 32:573 (1993); Mulligan, Science 260:926 (1993); Morgan, et al., Ann Rev Biochem 62:191 (1993); May, TIBTECH 11:155 (1993).

[0177] In a one aspect, the compound comprises nucleic acid sequences encoding an antibody, said nucleic acid sequences being part of expression vectors that express the antibody or fragments or chimeric proteins or heavy or light chains thereof in a suitable host. In particular, such nucleic acid sequences have promoters operably linked to the antibody coding region, said promoter being inducible or constitutive, and, optionally, tissue-specific.

[0178] In another particular embodiment, nucleic acid molecules are used in which the antibody coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the antibody encoding nucleic acids (Koller, et al., Proc Natl Acad Sci USA 86:8932 (1989); Zijlstra, et al., Nature 342:435 (1989)). In specific embodiments, the expressed antibody molecule is a single chain antibody; alternatively, the nucleic acid sequences include sequences encoding both the heavy and light chains, or fragments thereof, of the antibody.

[0179] Delivery of the nucleic acids into a patient may be either direct, in which case the patient is directly exposed to the nucleic acid or nucleic acid-carrying vectors, or indirect, in which case, cells are first transformed with the nucleic acids in vitro, then transplanted into the patient. These two approaches are known, respectively, as in vivo or ex vivo gene therapy.

[0180] In a specific embodiment, the nucleic acid sequences are directly administered in vivo, where it is expressed to produce the encoded product. This can be accomplished by any of numerous methods known in the art, e.g., by constructing them as part of an appropriate nucleic acid expression vector and administering it so that they become intracellular, e.g., by infection using defective or attenuated retrovirals or other viral vectors (see U.S. Pat. No. 4,980,286), or by direct injection of naked DNA, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, encapsulation in liposomes, microparticles, or microcapsules, or by administering them in linkage to a peptide which is known to enter the nucleus, by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu, et al., J Biol Chem 262:4429 (1987)) (which can be used to target cell types specifically expressing the receptors), etc. In another embodiment, nucleic acid-ligand complexes can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation. In yet another embodiment, the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180; WO 92/22635; WO92/20316; WO93/14188, WO 93/20221). Alternatively, the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination (Koller, et al., Proc Natl Acad Sci USA 86:8932 (1989); Zijlstra, et al., Nature 342:435 (1989)).

[0181] In a specific embodiment, viral vectors that contain nucleic acid sequences encoding an antibody of the invention are used. For example, a retroviral vector can be used (see Miller, et al., Meth Enzymol 217:581 (1993)). These retroviral vectors contain the components necessary for the correct packaging of the viral genome and integration into the host cell DNA. The nucleic acid sequences encoding the antibody to be used in gene therapy are cloned into one or more vectors, which facilitate the delivery of the gene into a patient. More detail about retroviral vectors can be found in Boesen, et al., Biotherapy 6:291 (1994), which describes the use of a retroviral vector to deliver the mdr1 gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy. Other references illustrating the use of retroviral vectors in gene therapy are: Clowes, et al., Clin Invest 93:644 (1994); Kiem, et al., Blood 83:1467 (1994); Salmons, et al., Human Gene Therapy 4:129 (1993); and Grossman, et al., Curr Opin Gen and Dev 3:110 (1993).

[0182] Adenoviruses may also be used in the present invention. Adenoviruses are especially attractive vehicles in the present invention for delivering antibodies to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky, et al., Curr Opin Gen Dev 3:499 (1993) present a review of adenovirus-based gene therapy. Bout, et al., Human Gene Therapy 5:3 (1994) demonstrated the use of adenovirus vectors to transfer genes to the respiratory epithelia of rhesus monkeys. Other instances of the use of adenoviruses in gene therapy can be found in Rosenfeld, et al., Science 252:431 (1991); Rosenfeld, et al., Cel168:143 (1992); Mastrangeli, et al., J Clin Invest 91:225 (1993); PCT Publication WO94/12649; Wang, et al., Gene Therapy 2:775 (1995). Adeno-associated virus (AAV) has also been proposed for use in gene therapy (Walsh, et al., Proc Soc Exp Biol Med 204:289 (1993); U.S. Pat. Nos. 5,436,146; 6,632,670; and 6,642,051).

[0183] Another approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection. Usually, the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a patient.

[0184] In this embodiment, the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell. Such introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion, etc. Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Loeffler, et al., Meth Enzymol 217:599 (1993); Cohen, et al., Meth Enzymol 217:618 (1993); Cline, Pharmac Ther 29:69 (1985)) and may be used in accordance with the present invention, provided that the necessary developmental and physiological functions of the recipient cells are not disrupted. The technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.

[0185] The resulting recombinant cells can be delivered to a patient by various methods known in the art. Recombinant blood cells (e.g., hematopoietic stem or progenitor cells) are preferably administered intravenously. The amount of cells envisioned for use depends on the desired effect, patient state, etc., and can be determined by one skilled in the art.

[0186] Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood, fetal liver, etc.

[0187] In one embodiment, the cell used for gene therapy is autologous to the patient. Nucleic acid sequences encoding an antibody of the present invention are introduced into the cells such that they are expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect. In a specific embodiment, stem or progenitor cells are used. Any stem and/or progenitor cells which can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention (see e.g. PCT Publication WO 94/08598; Stemple, et al., Cell 71:973 (1992); Rheinwald, Meth Cell Bio 21A:229 (1980); Pittelkow, et al., Mayo Clinic Proc 61:771 (1986)).

EXAMPLES

Example 1

Generation of Immunogen: Notch3 Extracellular Domain-Fc Fusion Protein

[0188] Anti-Notch3 monoclonal antibodies that specifically bind to the LIN12/dimerization domain (herein after "LD") of human Notch3 were generated using a recombinant Notch3-Fc fusion protein as immunogen comprising Notch3 LD fused to a gamma 1 Fc region at the carboxy terminal end. Specifically, the immunogen comprised amino acid residues 1378 to 1640 of Notch3 LD (See FIG. 1) and human .gamma.1Fc fusion protein (Notch3 LD/Fc). A control antibody was generated comprising the Notch3 EGF repeat region from amino acid residues 43 to 1377 (designated 255A-79).

[0189] Notch3 protein sequence was analyzed using an internet-based research software and service (Motif Search). Human liver and pancreatic RNAs (Ambion, Inc. Austin, Tex.) were used as templates to synthesize the first strand of cDNA using a standard commercially available cDNA synthesis kit. The cDNAs encoding the Notch3 LD and the EGF repeat region were PCR-amplified in the presence of Betaine (1-2M) and DMSO (5%). The PCR-synthesized Notch3-LD DNA fragment (.about.0.8 kb) and Notch3-EGF repeat DNA fragment (.about.4 kb) were cloned into expression vectors comprising a His-.gamma.1Fc in the commercially available vector pSec or in the commercially available vector pCD3.1, each bearing a different antibiotic marker. This cloning resulted in two expression plasmids, one expressing a Notch3-LD/Fc fusion protein and the other expressing a Notch3-EGF/Fc fusion protein.

[0190] To facilitate the plasmid construction and to enhance the expression of the various Notch 3 recombinant proteins, oligonucleotides corresponding to the leader peptide sequence comprising the first 135 base pairs of the Notch3 nucleic acid coding sequence were generated. These oligonucleotides contained some changes in the wobble coding positions to lower the GC content. All nucleotide sequence changes were silent, i.e., no amino acid sequence changes (FIG. 14A). After annealing the oligonucleotides together, the engineered leader peptide coding sequence was linked to the rest of the coding sequence by PCR-SOE (Ho, et al., Gene 77:51 (1989); Horton, et al., Bio Techniques 8:528 (1990)) (See FIG. 15). This leader peptide coding sequence was used in Notch3-LD/Fc and Notch3 expression constructs. Therefore, both of the Fc fusion proteins comprise a signal peptide linked to the N-terminus, and a human .gamma.1Fc sequence fused to the C-terminus. The amino acid sequence of Notch3-LD, including the leader peptide, is shown in FIG. 14 and SEQ ID NO:6.

[0191] Expression of Notch3-EGF/Fc and Notch3-LD/Fc fusion proteins was verified by transient transfection of the Notch3 expression plasmids into 293T (ATCC Number CRL-11268, Manassas, Va.) and CHO cells (Invitrogen, Carlsbad, Calif.), respectively. Prior to transfection, cells were cultured in DMEM (Invitrogen, Carlsbad, Calif.) growth medium containing 10% fetal calf serum (FCS), 2 mM of glutamine, and 1.times. essential amino acid solution followed by seeding about 3-5.times.10.sup.5 cells per well in 6-well plate and growing for approximately 24 hours. Three micrograms each of the Notch3 fusion protein expression plasmids were transfected into cells in each well using a LIPOFECTAMINE.TM. 2000 transfection system (Invitrogen, Carlsbad, Calif.) following the manufacturer's protocol. After transfection, the cells were cultured in fresh growth medium and cultured in a CO.sub.2 incubator for approximately 40-48 hours before subjecting to Notch3 fusion protein expression analysis. Alternatively, after transfection, the cells were cultured in growth medium for 3-4 hours, then switched to DMEM medium containing 2% FCS and cultured for approximately 60-66 hours before drawing conditioned medium for secreted protein analysis.

[0192] Stable cell lines were generated for both Notch3-LD/Fc (His-Fc.gamma./pSec vector) and Notch3-EGF/Fc (His-Fc.gamma./pSec vector). Each plasmid was transfected into CHO cells. After transfection, the cells were cultured in DMEM growth medium overnight, then switched to growth medium with 800 .mu.g/ml hygromycin and cultured at least two weeks until the cells not carrying Notch3 expression plasmid were eliminated by the antibiotics. Conditioned media from the stable cell lines were subjected to Western blot analysis.

[0193] Stable or transient transfected cells were assayed for expression and secretion of Notch3-LD/Fc or Notch3-EGF/Fc fusion protein. Transfected cells harvested from culture dishes were washed once with phosphate buffered saline (PBS) and resuspended in deionized water, mixed with an equal volume of 2.times. protein sample loading buffer (BioRad, Hercules, Calif.) and then heated at about 100.degree. C. for 10 minutes. Secreted protein was analyzed using conditioned medium mixed with an equal volume of 2.times. protein sample loading buffer and heated at 100.degree. C. for 10 minutes. The samples were separated using 4-15% gradient SDS-PAGE. The proteins were transferred from the gel to a PVDF membrane (BioRad, Hercules, Calif.), which was blocked in 5% non-fat dry milk in PBST (PBS with 0.05% TWEEN-20.RTM.) for at least one hour prior to transfer of protein.

[0194] Notch3-EGF/Fc and Notch3-LD/Fc fusion proteins were detected by incubating with yFc-specific, HRP-conjugated antibody (Sigma, St Louis, Mo.) in blocking buffer for one hour at room temperature. The membrane was washed three times in PBST and developed with a chemiluminescent substrate.

[0195] For Notch3 domain/Fc fusion protein purification, CHO stable cell lines as described above were cultured in DMEM with 2% FCS for up to 5 days. One liter of conditioned medium was collected and subjected to protein-A bead-packed column chromatography for affinity binding. The column was washed with PBS, and the bound proteins were eluted in 50 mM citrate buffer (pH 2.8), and the pH was brought to neutral by adding 1 M Tris-HCl buffer (pH 8). Purity of the protein was assessed by protein gel analysis using 4-15% gradient SDS-PAGE. Protein concentration was assayed using Coomassie blue reagent following the manufacturer's protocol (Pierce, Rockford, Ill.). Through this procedure, milligram quantities of Notch3-LD/Fc and Notch3-EGF/Fc protein were purified for immunization and ELISA binding assays.

Example 2

Generation of Anti-Notch3 MAbs

[0196] Male A/J mice (Harlan, Houston, Tex.), 8-12 weeks old, were injected subcutaneously with 25 .mu.g of Notch3-EGF/Fc or Notch3-LD/Fc in complete Freund's adjuvant (Difco Laboratories, Detroit, Mich.) in 200 .mu.l of PBS. Two weeks after the injections and three days prior to sacrifice, the mice were again injected intraperitoneally with 25 .mu.g of the same antigen in PBS. For each fusion, single cell suspensions were prepared from spleen of an immunized mouse and used for fusion with Sp2/0 myeloma cells; 5.times.10.sup.8 of Sp2/0 and 5.times.10.sup.8 of spleen cells were fused in a medium containing 50% polyethylene glycol (M.W. 1450) (Kodak, Rochester, N.Y.) and 5% dimethylsulfoxide (Sigma, St. Louis, Mo.). The cells were then adjusted to a concentration of 1.5.times.10.sup.5 spleen cells per 200 .mu.l of the suspension in Iscove medium (Invitrogen, Carlsbad, Calif.), supplemented with 10% fetal bovine serum, 100 units/ml of penicillin, 100 .mu.g/ml of streptomycin, 0.1 .mu.M hypoxanthine, 0.4 .mu.M aminopterin, and 16 .mu.M thymidine. Two hundred microliters of the cell suspension were added to each well of about sixty 96-well plates. After around ten days, culture supernatants were withdrawn for screening their antibody-binding activity using ELISA.

[0197] The 96-well flat bottom IMMULON.RTM. II microtest plates (Dynatech Laboratories, Chantilly, Va.) were coated using 100 .mu.l of Notch3-EGF/Fc or Notch3-LD/Fc (0.1 .mu.g/ml) in (PBS) containing 1.times. Phenol Red and 3-4 drops pHix/liter (Pierce, Rockford, Ill.) and incubated overnight at room temperature. After the coating solution was removed by flicking of the plate, 200 .mu.l of blocking buffer containing 2% BSA in PBST containing 0.1% merthiolate was added to each well for one hour to block non-specific binding. The wells were then washed with PBST. Fifty microliters of culture supernatant from each fusion well were collected and mixed with 50 .mu.l of blocking buffer and then added to the individual wells of the microtiter plates. After one hour of incubation, the wells were washed with PBST. The bound murine antibodies were then detected by reaction with horseradish peroxidase (HRP)-conjugated, Fc-specific goat anti-mouse IgG (Jackson ImmunoResearch Laboratories, West Grove, Pa.). HRP substrate solution containing 0.1% 3,3,5,5-tetramethyl benzidine and 0.0003% hydrogen peroxide was added to the wells for color development for 30 minutes. The reaction was terminated by the addition of 50 ml of 2 M H.sub.2SO.sub.4/well. The OD at 450 nm was read with an ELISA plate reader (Molecular Devices, Sunnyvale, Calif.).

[0198] Among 185 hybridomas isolated and analyzed, two hybridoma clones from mice immunized with Notch3-LD/Fc generated Notch3 antagonizing antibodies, which were further characterized. The ELISA using supernatant from the two hybridoma clones producing MAbs 256A-4 and 256A-8 showed strong binding activity to the purified Notch3 LD/FC fusion protein to which it was generated and did not bind to human Notch1-LD/Fc (LIN/dimerization domain fused to Fc region at the carboxyl terminus) or a control human Fc protein (data not shown) (Table 1). Later studies using functional assays also demonstrated that MAbs 256A-4 and 256A-8 specifically antagonize Notch3 relative to Notch1 and Notch2 (data not shown).

TABLE-US-00001 TABLE 1 ELISA OD readings of anti-Notch3 Mabs using hybridoma supernatant Notch3-LD/Fc Notch1-LD/Fc Mean S.D. Mean S.D. 256A-4 4.000 0.000 0.106 0.004 256A-8 4.000 0.000 0.115 0.014 Control IgG1* 0.064 0.006 0.066 0.006 *Control IgG was an irrelevant IgG1 monoclonal antibody.

[0199] The positive hybridoma clones from this primary ELISA screening were further isolated by single colony-picking and a second ELISA assay as described above was done to verify specific binding to the chosen immunogen. The confirmed hybridoma clones were expanded in larger scale cultures. The monoclonal antibodies (MAbs) were purified from the medium of these large scale cultures using a protein A affinity column. The anti-Notch3 MAbs were then characterized using cell-based binding assays, microscopy, Western blot, and FACS analysis.

Example 3

Cell-Based Binding Assays for Anti-Notch3 MAbs

[0200] The cell-based binding assays used to characterize the anti-Notch3 MAbs required cloning a full-length human Notch3 open reading frame into a vector, in this case PcDNA.TM. 3.1/Hygro (Invitrogen, Carlsbad, Calif.). The Notch3-coding region was synthesized by RT-PCR using human liver tumor RNA (Ambion, Inc., Austin, Tex.) as a template. The final plasmid construct, Notch3/Hygro, expressed a full-length Notch3 protein as depicted in FIG. 1. A stable cell line expressing Notch3 was generated by transfection of Notch3/Hygro plasmid construct into 293T cells (ATCC No. CRL-11268) using a LIPOFECTAMINE.TM. 2000 kit following the same procedure as described in Example 1. After transfection, the cells were cultured in DMEM growth medium overnight, then reseeded in growth medium with 200 .mu.g/ml hygromycin and cultured for 12-14 days. Well-isolated single colonies were picked and grown in separate wells until enough clonal cells were amplified. Stable 293T clones that were resistant to hygromycin selection and expressed high levels of Notch3 protein were identified by Western blot analysis, and by fluorescent electromicroscopy using polyclonal anti-Notch3 antibodies (R&D Systems, Minneapolis, Minn.).

[0201] A partial Notch3 expression plasmid containing only the Notch LIN12/dimerization (LD) domain and the transmembrane (TM) domain was also constructed by PCR and subcloning into PCDNA.TM. 3.1 vector (Invitrogen, Carlsbad, Calif.). This plasmid construct also contains a V5 tag at its C-terminus and was termed Notch3-LDTM/V5. A stable cell line expressing this plasmid, Notch3-LDTM/V5, was generated according to the procedure described in Example 1.

[0202] Human Sup-T1 cell line (ATCC No. CRL-1942) naturally expressing Notch3 was also confirmed by Western blot. Sup-T1 cells were grown in RPMI1640 media containing 10% fetal calf serum, 2 mM of glutamine and 1.times. essential amino acid solution.

[0203] Cell-based antibody-binding was assessed using FMAT.TM. (fluorescence macro-confocal high-throughput screening) 8100 HTS System (Applied Biosystems, Foster City, Calif.) following the protocol provided by the manufacturer. Cell lines naturally expressing Notch3 or stably transfected with Notch3 expression constructs were seeded in 96-well plates. Alternatively, transiently transfected 293T or CHO cells were seeded in the 96-well plate. The cells were seeded at a density of 30,000-50,000 cells per well. After 20-24 hours, anti-Notch3 MAbs and 1.times.PBS reaction buffer were added to the wells and incubated for one hour at 37.degree. C. Cy-5-conjugated anti-mouse IgG antibody was added in the wells after removal of primary antibodies.

[0204] Cell-based antibody-binding was also assessed by fluorescence-activated cell sorter (FACS) using an internally generated 293T/Notch3-stable cell line and two cancer lines, human Sup-T1 and A2780 cell lines (UK ECACC No. Cat. No. 93112519), which both naturally express Notch3 (data not shown). Cells were first incubated with anti-Notch3 MAbs in 1.times.PBS. After three washes, the cells were incubated with fluorescent molecule-conjugated secondary antibody. The cells were resuspended, fixed in 1.times.PBS with 0.1% paraformaldehyde, and analyzed by FACS (BD Sciences, Palo Alto, Calif.). The results indicated that both MAbs bind to Notch3 receptor expressed either from recombinant plasmid constructs or as native protein in cultured cells (Table 2). However, Western blot showed that when the Notch3 receptor or the Notch3-LD/Fc fusion protein are denatured in SDS-PAGE and transferred to nylon blot membrane, the anti-Notch3 MAbs no longer bind, suggesting a conformational epitope. Transiently transfected 293T cells containing a Notch3/Hygro plasmid were also stained with immunofluorescence as described above and observed by fluorescent microscopy.

TABLE-US-00002 TABLE 2 Binding activity of anti-Notch3 MAbs in cell-based FACS analysis shown as mean fluorescent intensity Monoclonal Antibody 293T/Notch3-stable cell line Sup-T1 256A-4 195 43 256A-8 189 45 negative control* 21 23 positive control** 198 74

[0205] The cell-based FMAT.TM. and FACS analyses confirmed that both MAbs 256A-4 and 256A-8 indeed bind to the Notch3 receptor expressed either from recombinant plasmid constructs or as native protein in cultured cells (Table 2 and Table 3).

TABLE-US-00003 TABLE 3 Summary of anti-Notch3 MAbs binding activity in cell-based FMAT .TM. mAb 256A-4 mAb 256A-8 mAb G3 Notch3 (full-length)/ + + - 293T

[0206] G3 is a negative control human IgG1 Mab. A positive binding signal was determined based on the FMAT.TM. signal read-out that was significantly higher than G3 and other negative hybridoma clones (p>0.01). The negative signal of G3 FMAT.TM. binding read-out was considered background. Transiently transfected 293T cells with Notch3/Hygro plasmid were also stained with immunofluorescence as described above and observed by fluorescent microscopy.

Example 4

Western Blot Analysis of Anti-Notch3 MAb Binding Activity

[0207] Western blot was performed to assess the anti-Notch3 MAbs' binding activity to Notch3 under denaturing conditions, as well as expression levels of Notch3 and other Notch-related proteins in human cell lines. Purified Notch3-LD/Fc fusion protein was combined with protein loading buffer. Protein samples were also prepared from the transiently or stably transfected cells described in Example 1, which were harvested from culture dishes, washed once with PBS, resuspended in total cellular protein extract buffer (Pierce, Rockford, Ill.), and heated at 100.degree. C. for 10 minutes after adding equal volume of 2.times. protein sample loading buffer. All samples were separated by electrophoresis in a 4-15% gradient SDS-PAGE. The proteins were transferred from gel to PVDF membrane and anti-Notch3 MAbs were applied to the Western blot membrane as the primary detection antibody. An HRP-conjugated secondary antibody was used for detection and the signal generated using a chemiluminescent substrate as described above. Positive control antibodies against human Fc, V5 tag, Notch3 and Notch1 were purchased from Invitrogen, R&D Systems, Santa Cruz Biotechnologies, and Orbigen.

[0208] Western blot analysis showed that MAbs 256A-4 and 256A-8 do not bind to Notch3-LD/Fc under denaturing conditions, which is in distinct contrast to the results observed in ELISA and FACS analyses where Notch3 LIN12/heterodimerization domains are maintained in native molecular conformation. Therefore, it is concluded that MAbs 256A-4 and 256A-8 bind to multiple epitopes in Notch3-LD that have to be maintained in their native conformation. This conclusion was confirmed by the results from epitope mapping discussed in Example 8 below.

Example 5

Assessing Functionality of Anti-Notch3 MAbs by Luciferase Reporter Assay

[0209] A. Plasmid constructs

[0210] The full length Notch3 expression construct described in Example 3 above was confirmed by sequencing, and is identical to the published sequence depicted in FIG. 1. Human Jagged1 plasmid was obtained from OriGene (Rockville, Md.), and verified by sequencing as identical to NM.sub.--000214 (NCBI/GENBANK.RTM. accession number). Because the OriGene Jagged1 plasmid did not have an antibiotic selection marker, the Not I fragment containing Jagged1 coding sequence was transferred into PCDNA.TM. 3.1/Hygromycin. A 3.7 Kb subclone of human Jagged2 cDNA was generated by first strand cDNA synthesis from human T-cell leukemia cell line, HH (ATCC No. CRL-2105) and PCR-amplified. The Jagged2 cDNA was subsequently subcloned. The expression of Notch3, Jagged1, and Jagged2 was verified by transient transfection and Western blot as described in Example 4.

[0211] To generate a luciferase reporter plasmid for Notch signaling, two complementary oligonucleotide primers containing tandem repeats of CBF1 binding motif were synthesized having the following sequences:

TABLE-US-00004 (SEQ ID NO 7) 5'GCTCGAGCTCGTGGGAAAATACCGTGGGAAAATGAACCGTGGGAAA ATCTCGTGG (SEQ ID NO 8) 5'GCTCGAGATTTTCCCACGAGATTTTCCCACGGTTC

[0212] These two oligoprimers were annealed at 65.degree. C. in 100 mM of NaCl with each oligo at a concentration of 4 mM. After annealing to each other, the primers were extended by PCR. The PCR product was cloned into a commercially available vector. The insert was verified by sequencing, which contains four tandem repeats of CBF1 binding motif and two flanking Xho I sites. The insert was excised using Xho I and ligated downstream of the firefly luciferase reporter coding sequence. After luciferase reporter assay and sequencing analysis, plasmid clones with eight repeats of CBF1 binding motifs were selected and designated CBF1-Luc.

[0213] B. Stable Cell Line Generation

[0214] Two stable cell lines were generated for functional assays using human embryonic kidney cell lines (HEK293). One cell line contained the Notch3-expressing plasmid and CBF1-Luc reporter plasmid integrated into the nuclear genome. This cell line was generated by cotransfecting Notch3/hygromycin and CBF1-Luc plasmids into 293T cells using LIPOFECTAMINE.TM. 2000 transfection system according to the manufacturer's protocol. Stable transfection cell clones were selected against 200 .mu.g/ml hygromycin in DMEM growth medium, and screened by luciferase reporter assay and Western blot. A cell line with relatively high level of Notch3 expression (based on Western blot) and luciferase activity was selected for use in functional assay, and designated NC85.

[0215] The second stable cell line contained a Notch ligand expression construct, such as Jagged1 or Jagged2, or PCDNA.TM. 3.1 as negative control. Stable cell lines expressing human Jagged1 or harboring PCDNA.TM. 3.1 were generated by transfection into 293T cells and selection against hygromycin as described above. Jagged2 was subcloned, transfected into a 293T cell line and expected to be integrated into a specific locus in the genome. Hygromycin-resistant cells were selected as above.

[0216] C. Luciferase Reporter Assay Under Coculture Conditions

[0217] NC85 cells were mixed and cocultured with another 293T cell line stably expressing human Jagged1 (Jagged1/293T), Jagged2/293F, or PCDNA.TM. 3.1/293T, respectively, for 24 to 48 hours. At the end of the co-culture, the media was removed by aspiration, cells were lysed in 1.times. Passive Lysis Buffer (E1501, Promega, Madison, Wis.) and luciferase activities were assayed using the Luciferase Assay System following manufacturer's protocol (E1501, Promega, Madison, Wis.) in TD-20/20 luminometer (Turner Designs Instrument, Sunnyvale, Calif.). As illustrated in FIG. 6 and FIG. 7, when NC85 cells were cocultured with Jagged1/293T or with Jagged2/293F, the luciferase activity was increased 2-4 fold as compared to that of coculturing with PCDNA.TM. 3.1/293T cells. To assess the inhibitory effect of anti-Notch3 MAbs, the antibodies were added to the cell culture at beginning of seeding and mixing of cocultured cells. (256-A, 256A-8 and an EGF-Repeat Domain control 255A-79).

[0218] D. Luciferase Reporter Assay by Culturing Cells on Notch Ligand-Coated Plates

[0219] Regular 96-well tissue culture plates from Becton Dickinson Labware (#18779, Palo Alto, Calif.) were coated with rat Jagged1/Fc, human DLL-4 (R&D Systems, Minneapolis, Minn.) or Human Fc (Jackson ImmunoResearch, West Grove, Pa.), bovine serum albumin (Sigma, St Louis, Mo.). One hundred microliters of each protein (3 .mu.g/ml in PBS) was distributed in a well and maintained at room temperature or 4.degree. C. for at least 8 hours until the coating solution was removed before use. NC85 cells or cancer cells were seeded at 3-5.times.10.sup.4 cells per well and allowed to grow for 28-48 hours. The luciferase reporter assay and antibody inhibition assay were performed as described in Section C above. The luciferase reporter assay demonstrated the two MAbs 256A-4 and 256A-8 binding to LIN12/dimerization domain almost completely blocked Jagged1 and Jagged2-induced luciferase reporter activity (FIGS. 6 and 7). In contrast, a MAb specifically binding to Notch3-EGF domain (255A-79), as a control, only inhibited Jagged1-induced luciferase reporter activity (about 60% inhibition, FIG. 6), but not Jagged2-induced luciferase reporter activity (FIG. 7). The ability of MAbs 256A-4 and 256A-8 to block DLL-4-induced luciferase reporter activity is shown in FIG. 8.

[0220] Additional functional assays demonstrated that MAbs 256A-4 and 256A-8 inhibited ligand-induced up-regulation of Notch target genes. 293T cells expressing recombinant Notch3 were cultured on Jagged-1-coated plates. In the presence of MAbs 256A-4 and 256A-8, up-regulation of HESS and HEY2, two Notch target genes, was inhibited, as measured by quantitative RT-PCR (data not shown).

[0221] To verify whether the anti-Notch3 MAbs can bind to native Notch3 expressed in human cancer cells and block the receptor signaling, a reporter assay was performed using two ovarian cancer cell lines, OV/CAR3 and A2780. Both 256A-4 and 256A-8 significantly blocked Jagged1-induced Notch signaling mediated by native Notch3 in OV/CAR3 cells (FIG. 9a). Similarly, both MAbs inhibited about 50% of luciferase activity induced by D114 coated on the plate (FIG. 9b). The latter result is consistent with the fact that both Notch1 and Notch3 are expressed in A2780 cells. These results suggest that the anti-Notch3 MAbs can inhibit native Notch3-mediated signaling in cancer cells.

Example 6

Apoptosis Assay

[0222] Annexin V is an early apoptotic marker on the cell surface, and the apoptotic cell population can be marked by fluorophore-labeled anti-Annexin V antibody and quantified by FACS analysis. NC85 cells were seeded at 5-6.times.10.sup.4 cells per well in Fc- or Jagged1/Fc-coated 96-well plate as described above and maintained in serum-free DMEM medium for 24 hours. Apoptotic cells were stained by FITC-labeled anti-Annexin V antibody (BD Biosciences, Palo Alto, Calif.) and analyzed by FACS. Cells cultured on Jagged1/Fc-coated surface had significantly lower apoptotic cell population comparing to those cultured on Fc-coated plate (FIG. 10). To study the antibody's functional effect, anti-Notch3 MAbs were added in cell culture at the beginning of the study. As shown in FIG. 10, anti-Notch3 MAbs 256A-4 and 256A-8 blocked about 50-65% of the cell survival effect induced by Jagged1.

Example 7

Cell Migration Assays, Invasion Assays, and Morphology Assays

[0223] In vitro cell migration and invasion assays are frequently used to assess metastasis potential of cancer cells. These assays were performed to assay the inhibitory effect exerted by the anti-Notch3 MAbs on the tumorgenic 293T/Notch3-stable cell line (NC85). The invasion assay was performed using COSTAR.RTM. 48-well insert plate (Sigma-Aldrich, St. Louis, Mo.). The insert divides the well into upper and lower chambers which are separated by a porous membrane (pore diameter=8 .mu.m) at the bottom of the insert. Notch ligands, Jagged1/Fc, DLL-4, or human Fc, were immobilized on the membrane surface as describe in above sections. NC85 cells were seeded at 100,000 cells per well and maintained in serum-free DMEM in the upper chamber and 10% FCS/DMEM in the lower chamber. After 10-24 hours, cells that remained on the top surface of the insert membrane were removed, and the cells that passed the membrane adhering on the bottom of the insert membrane were stained by 0.05% crystal velvet in PBS. The dye was extracted from the cells by 30% acetic acid and absorption readings at 590 nm were recorded. The anti-Notch3 MAbs were added to cell culture 24 hours before seeding NC85 cells in the COSTAR.RTM. assay plate and all MAbs were added to the cell culture 24 hours before seeding NC85 cells in the COSTAR.RTM. assay plate. Fresh MAbs were added to maintain the same concentration in the migration assay plate. Experimental results are shown in FIG. 11A.

[0224] The invasion assay was performed using Becton Dickinson 48-well MATRIGEL.TM. plate (BD Labware, Palo Alto, Calif.). The cell culture well was divided by an insert well into upper and lower chambers, which are separated by a porous membrane (pore diameter=8 .mu.m) at the bottom of the insert well. An optimized density of MATRIGEL.TM. matrix was coated on the membrane top surface and fibronectin was coated on the membrane bottom surface by the manufacturer. NC85, Jagged1/293T, and pcDNA3.1/293T cells were mixed pair-wise such as indicated in FIG. 11B. A total of 6-10.times.10.sup.4 cells were seeded in each well in the 48-well MATRIGEL.TM. plate and cultured in growth medium for 24 hours. The cells that remained on top of the insert membrane in the upper chamber were removed and the cells that passed the membrane adhering on the bottom of the insert membrane were stained by 0.05% crystal velvet in PBS. The dye was extracted and absorption measurements were as described in the previous section. MAbs were added at the beginning of the mixed cell culture. The results are shown in FIG. 11B.

[0225] The cell migration assay results showed that when NC85 cells were cultured on Jagged1-coated membrane, the activation of Notch3 signaling significantly increased cell migration, and MAbs 256A-4 and 256A-8 clearly inhibited the migration (FIG. 11A). The invasion experiment showed a similar trend (FIG. 11B).

[0226] Additionally, the effect of MAbs 256A-4 and 256A-8 on Jagged-1-induced formation of cell "spheres" was examined. When 293T cells over-expressing Notch3 were cultured on Jagged-1-coated plates, the cells formed loosely attached "cell balls" or "spheres." In the presence of MAbs 256A-4 and 256A-8, however, formation of these cell spheres was inhibited (data not shown).

Example 8

Mapping the Binding Epitope of Anti-Notch3 MAbs

[0227] A. Domain Swap Strategy and Rationale

[0228] First, the antagonist Notch3 MAbs bind to Notch3 LIN12/dimerization domain (LD), but not to the homologous human Notch1 LIN12/dimerization domain (See FIGS. 12 and 13). Second, the anti-Notch3 MAbs do not bind to denatured Notch3 protein in Western blot as discussed in Example 4, indicating the MAbs bind to conformational epitopes. Third, Notch3 and Notch1 share approximately 55% amino acid sequence homology in the LIN12/dimerization domain, and therefore it was concluded that a domain swap between Notch3 and Notch1 within this region would not disrupt the protein conformation.

[0229] B. Generating Domain Swap Fusion Protein Constructs

[0230] Sequence analysis indicated that Notch3 has three LIN12 repeats and its dimerization domain is divided into two segments. Therefore, five domain swap protein constructs were generated with each of the three LIN12 repeats and the two dimerization segments replaced by the corresponding domains of Notch1. The domain swap constructs were generated using PCR-SOE (Ho, et al., Gene 77:51 (1989); Horton, et al., BioTechniques 8:528 (1990)) as illustrated in FIG. 12. PCR and PCR-SOE reactions were performed using PCR with 1M Betaine and 5% DMSO added to the reaction. PCR thermocycling was almost same for PCR and PCR-SOE except that the annealing step of each PCR cycle was extended one minute in PCR-SOE. The final PCR-SOE product was subcloned and verified by sequencing. The plasmid clone with the correct insert sequence was cleaved with Nhe I and Xho I to excise the insert, which was gel-purified and subcloned. The five Notch3/Notch1 domain swap constructs are illustrated in FIG. 12. To facilitate the epitope mapping, the human IgG kappa chain signaling peptide was used as leader peptide in the domain swap constructs. The amino acid sequences are shown in FIGS. 16 and 17.

[0231] Notch1-LD cDNA was PCR-amplified using PCR and methods described in the above section. The first strand cDNA template was synthesized from PA-1 cell total RNA (ATCC No. CRL-1572). The human IgG kappa chain leader peptide coding sequence was PCR-amplified, used as leader peptide to link to the 5' of Notch1-LD by PCR-SOE and subcloned in His-ylFc/pSec.

[0232] Based on ELISA analysis results, target domains L1, D1 and D2 were further divided into subdomains. ELISA binding analysis using the subdomain expression constructs showed that only L1 and D2 were required for the Notch3 MAb binding. The D1 domain was not required. Therefore, L1 and D2 domains were divided into clusters of amino acid mutations for further analysis of the specific binding site. Constructs containing L1 and D2 subdomain swap or clusters of amino acid mutations as shown in FIG. 16 and FIG. 17 were generated.

[0233] C. Expression of Notch3/Notch1 Domain Swap Fusion Protein

[0234] Notch3/Notch1-LD domain swap plasmids were transiently transfected in CHO cells using LIPOFECTAMINE.TM. 2000 transfection system. CHO cells were seeded in DMEM growth medium with 10% FCS at 0.8.about.1.times.10.sup.6 cells per well in E-well plate, maintained in CO.sub.2 incubator overnight before transfection. The cells were recovered after transfection in the growth medium for about 3 hours, then switched to DMEM with 2% FCS, and cultured for three days. The conditioned media were harvested and centrifuged at 3500 rpm for 10 minutes. The supernatant containing Notch3-LD domain swap protein secreted from CHO was collected and prepared for Western blot and ELISA binding analyses. ELISA showed that all the domain-swap fusion proteins were expressed and secreted in conditioned medium (Table 4), which was further confirmed by Western blot analysis (data not shown).

[0235] The ELISA readings used anti-human Fc antibody as detection antibody showing all the proteins were expressed in conditioned medium. Human IgG/Fc was used as a control. The starting point of human IgG/Fc coated in each well is 100 ng.

TABLE-US-00005 TABLE 4 ELISA Readings Dilution N1-LD N3-LD L1-swap L2-swap L3-swap D1-swap D2-swap hlgG-Fc Statistics: mean 1 3.2000 3.3445 3.4380 3.0970 3.2910 3.2870 3.4110 3.5510 0.250000 3.1305 2.7625 2.9890 2.7390 2.9050 3.0225 2.9570 3.4995 0.062500 2.3785 1.3870 2.8145 1.2835 2.6855 2.2575 2.3240 3.5805 0.015625 1.0085 0.3960 1.5245 0.3865 1.7350 0.9110 0.8800 3.2355 0.003906 0.3300 0.1075 0.4755 0.1220 0.5970 0.3450 0.2130 1.8585 0.000977 0.2095 0.0400 0.1640 0.1105 0.1780 0.1635 0.0615 0.5865 0.000244 0.1340 0.0225 0.0500 0.0595 0.0575 0.1045 0.0275 0.1445 6.104E-05 0.1000 0.0135 0.0405 0.0505 0.0230 0.0575 0.0305 0.0315 1.526E-05 0.0975 0.0165 0.0205 0.0430 0.0180 0.0400 0.0155 0.0220 3.815E-06 0.0580 0.0140 0.0135 0.0300 0.0150 0.0425 0.0235 0.0230 9.537E-07 0.0540 0.0125 0.0155 0.0245 0.0215 0.0480 0.0145 0.0165 2.384E-07 0.0415 0.0125 0.0145 0.0305 0.0155 0.0370 0.0150 0.0190 Statistics: S.D. 1 0.0778 0.0290 0.0679 0.0255 0.0933 0.1018 0.0283 0.0071 0.250000 0.0191 0.0304 0.0354 0.0396 0.0693 0.1619 0.1202 0.0148 0.062500 0.0898 0.0919 0.0007 0.1096 0.0318 0.0021 0.0071 0.0290 0.015625 0.0474 0.0354 0.0106 0.0417 0.1075 0.0071 0.0325 0.1450 0.003906 0.0523 0.0177 0.0460 0.0113 0.0453 0.0339 0.0057 0.0573 0.000977 0.0092 0.0057 0.0042 0.0191 0.0156 0.0205 0.0007 0.0955 0.000244 0.0226 0.0092 0.0014 0.0106 0.0064 0.0035 0.0049 0.0276 6.104E-05 0.0113 0.0007 0.0064 0.0035 0.0057 0.0134 0.0064 0.0064 1.526E-05 0.0021 0.0035 0.0049 0.0042 0.0000 0.0028 0.0007 0.0028 3.815E-06 0.0113 0.0028 0.0021 0.0000 0.0042 0.0064 0.0007 0.0057 9.537E-07 0.0014 0.0007 0.0007 0.0007 0.0064 0.0057 0.0021 0.0078 2.384E-07 0.0120 0.0035 0.0049 0.0021 0.0007 0.0113 0.0014 0.0127

[0236] Abbreviations for proteins used in the ELISA binding assays of Table 4 include: N1-LD, Notch1-LD/Fc. N3-LD, Notch3-LD/Fc. L1-swap: 1st LIN12 domain swap. L2-swap: 2nd LIN12 domain swap. L3-swap: 3rd LIN12 domain swap. D1-swap: 1st dimerization domain swap. D2-swap: 2nd dimerization domain swap. hIgG-Fc, human IgG Fc.

[0237] D. Epitope Binding Analysis Using ELISA

[0238] The 96-well flat bottom IMMULON II microtest plates (Dynatech Laboratories, Chantilly, Va.) were coated with anti-human Fc antibody (Jackson ImmunoResearch) by adding 100 .mu.l of the antibody (0.1 .mu.g/ml) in phosphate buffered saline (PBS) containing 1.times. Phenol Red and 3-4 drops pHix/liter (Pierce, Rockford, Ill.), and incubated overnight at room temperature. After the coating solution was removed by flicking of the plate, 200 .mu.l of blocking buffer containing 2% BSA in PBST and 0.1% merthiolate was added to each well for one hour to block non-specific binding. The wells were then washed with PBST. Fifty microliters of the above conditioned medium from each transfection of Notch3/Notch1 domain swap construct were collected, mixed with 50 .mu.l of blocking buffer, and added to the individual wells of the microtiter plates. After one hour of incubation, the Notch3/Notch1-LD domain swap protein was captured by the coated anti-Fc antibody, and the wells were washed with PBST. Anti-Notch3 MAbs and isotype-matched control MAbs were serially diluted in blocking buffer as above, and 50 .mu.l of the diluted MAbs were added in each well to assess binding to the bound Notch3/Notch1 domain swap protein. Horseradish peroxidase (HRP)-conjugated, Fc-specific goat anti-mouse IgG was used for detection. HRP substrate solution containing 0.1% 3,3,5,5-tetramethyl benzidine and 0.0003% hydrogen peroxide was added to the wells for color development for 30 minutes. The reaction was terminated by addition of 50 ml of 2 M H2SO.sub.4/well. The OD at 450 nm was read with an ELISA reader. Subdomain swap constructs and clusters of mutations were similarly examined by ELISA analysis above.

[0239] ELISA binding experiments using MAbs 256A-4 and 256A-8 against the domain-swap proteins showed that the swap of the 1st LIN12 domain (L1) and 2nd dimerization domain (D2) completely abolished all the three MAbs binding, while the swap of 1st dimerization domain (D1) abolished binding of MAbs 256A-4 and 256A-8 (FIGS. 13 B&C). Swap of the 3rd LIN12 domain (L3) significantly weakened the binding. Nevertheless, both MAbs were still able to bind to the fusion protein. The swap of the 2nd LIN12 domain had no interference with the binding of the MAbs (FIGS. 13B and C). A positive control antibody, which was previously mapped to bind to the 1st LIN12 domain, bound to all domain swap fusion protein except L1 (FIG. 13D). In contrast, isotype control negative antibody, G3, does not bind to any of the domain swap fusion proteins in the ELISA assay (data not shown). It was concluded from the above experiments that the 1st LIN12 domain and 2nd dimerization domain were required for MAbs 256A-4 and 256A-8 binding.

[0240] To further map the epitopes in the 1st LIN12 domain (L1) to which anti-Notch3 MAbs bind, the L1 domain was further divided into three subdomains, L1-sub1, L1-sub2 and L1-sub3, and swapped with the corresponding sequences in Notch1 (FIG. 16). An ELISA binding assay showed that L1-sub1 swap has no inhibitory effects on binding activity, and L1-sub2 and L1-sub3 swap abolished binding (FIG. 16). In L1-sub2 and L1-sub3 regions, there are five clusters of amino acid residues that differ between Notch3 and Notch1. Therefore, swap fusion protein constructs were generated within these five clusters of amino acids (FIG. 16). ELISA analysis demonstrated that L1-cluster4 swap had no inhibition on all three MAbs binding. The remaining four clusters of swap partially or completely abolished the anti-Notch MAbs binding. Thus, those four clusters of amino acid residues represented four different epitopes to which the MAbs bind. L1-cluster3 (amino acids: DRE) and L1-cluster5 (amino acids: SVG) are required. L1-cluster1 (amino acids: AKR) and cluster2 (amino acids: DQR) also played a role in anti-Notch3 MAb binding, whose mutations significantly weakened the MAb binding.

[0241] To map the epitopes in the 2.sup.nd dimerization (D2) domain of Notch3 to which anti-Notch3 MAbs bind, the D2 domain was further divided into five subdomains, D2-sub1, D2-sub2, D2-sub3, D2-sub4 and D2-sub5. The sequences in those subdomains were swapped with the corresponding sequences in Notch1 (FIG. 17). An ELISA binding assay showed that MAbs 256A-4 and 256A-8 have strong binding to D1-sub2 and D2-sub3 swap, but not to D2-sub1 and D2-sub4 swap. Both MAbs showed weak binding to D2-sub5 (FIG. 17). Therefore, the data suggested that D2-sub1 and D2-sub4 are required for the anti-Notch3 MAb binding and D2-sub5 may help the binding activity.

[0242] Both MAbs 256A-4 and 256A-8 are antagonistic antibodies binding to the conformational epitope comprising L1 and D2, while another antibody 256A-13 that binds only to L1 is agonistic (See co-pending U.S. application Ser. No. 11/874,682, filed Oct. 18, 2007). Furthermore, agonistic 256A-13 competes with antagonistic 256A-4 for an epitope within L1, and the epitope mapping studies suggest that they bind to an overlapping epitope on L1. The major difference is that the antagonistic antibodies also bind to D2, while the agonistic antibody does not. To test the hypothesis that simultaneous binding to L1 and D2 is responsible for the antagonistic activity, an antibody, 256A-2 binding to a similar epitope in D2 as 256A-4 was analyzed. MAb 256A-2 is neither antagonistic nor agonistic (data not shown). Studies showed that 256A-2 does not compete with 256A-13 and can bind to Notch3 simultaneously. Furthermore, 256A-2 and 256A-13 individually can partially compete with 256A-4, however, in combination these two antibodies completely block binding of 256A-4 to Notch3 (data not shown). Studies also showed that separate binding of two antibodies to the epitopes in L1 and D2 does not lead to the inhibition of ligand-dependent Notch3 activation, suggesting that the antagonistic antibodies form a bridge, possibly locking and stabilizing the L1 and D2 interaction, and preventing the ligand induced conformational changes. (See FIG. 18)

Example 9

Sequencing of Anti-Notch3 MAbs

[0243] Because antibody binding properties are dependent on the variable regions of both heavy chain and light chain, the variable sequences of 256A-4 and 256A-8 were subtyped and sequenced. The antibody IgG subtype was determined using an ISOSTRIP.TM. mouse monoclonal antibody isotyping kit (Roche Diagnostics, Indianapolis, Ind.). The results showed that both MAbs, 256A-4 and 256A-8 have an IgG1 heavy chain and a kappa light chain.

[0244] The variable region sequences of heavy chain and light chain were decoded through RT-PCR and cDNA cloning. Total RNAs from hybridoma clones 256A-4 and 256A-8 were isolated using an RNeasy Mini kit following the manufacturer's protocol (QIAGEN, Valencia, Calif.). The first strand cDNA was synthesized using the RNA template and SUPERSCRIPT.RTM. III reverse transcriptase kit. The variable region of light chain and heavy chain cDNAs were PCR-amplified from the first strand cDNA using degenerative forward primers covering the 5'-end of mouse kappa chain coding region and a reverse primer matching the constant region at the juncture to the 3'-end of the variable region, or using degenerative forward primers covering the 5'-end of mouse heavy chain coding region and a constant region reverse primer in mouse heavy chain. The PCR product was cloned into a commercially available vector and sequenced by Lone Star Lab (Houston, Tex.). The nucleotide sequences were analyzed utilizing the DNASTAR.RTM. computer software program (DNASTAR, Inc., Madison, Wis.). Each anti-Notch3 MAb sequence was determined by sequences from multiple PCR clones derived from the same hybridoma clone.

[0245] MAb 256A-4 contains 123 and 116 amino acid residues, respectively, in its variable region of heavy chain and light chain (FIGS. 4A and 4B). MAb 256A-8 consists of 122 and 123 amino acid residues in heavy chain and light chain variable regions, respectively (FIGS. 5A and 5B).

Example 10

Impact of Notch3 Antagonistic Antibodies on Metalloprotease Cleavage of Notch3

[0246] Notch receptor activation involves ligand induced metalloprotease cleavage at juxtamembrane site (S2) generating an extracellular subunit. This cleavage is an essential prerequisite to S3 cleavage to release the activated Notch intracellular region. Both 256A-4 and 256A-8 were found to require the presence of at least a portion of the Notch3 L1 and D2 domains for their bindings. These two domains are not located in close proximity in the linear sequence, but rather are on two separate polypeptides, suggesting these antibodies may stabilize an inactive, autoinhibited Notch configuration. To test whether the antagonizing antibodies can inhibit sequential Notch activation events, including two proteolytic cleavages, 293T cells stably expressing a recombinant Notch3 receptor (NC85 cells) are treated with either immobilized recombinant Jagged-1 or cocultured with 293T cells expressing Jagged-1. The soluble extracellular subunits generated by proteolytic cleavage in the culture medium are detected by an ELISA assay using an antibody bound to a solid surface that recognizes the Notch3 cleavage product. Notch3 antagonistic MAbs are expected to decrease the generation of soluble Notch3 extracellular subunits in the conditioned medium, whereas non-functional Notch3 binding antibodies would not.

[0247] To directly detect the S2 cleavage fragment, an 7.5% SDS PAGE electrophoresis and Western blot with Notch3 C-terminal antibody are performed. The S2 fragment is 57 amino acids residues smaller and migrates slightly faster than the non-cleaved Notch3 small subunit (transmembrane subunit).

[0248] To examine whether Notch3 antagonistic MAbs inhibit ligand-induced metalloprotease cleavage of Notch3 at S2, 293T cells expressing recombinant Notch3 were treated with the .gamma.secretase inhibitor compound E (1 .mu.M) for 4 hours, which stabilizes the product of cleavage at site S2, allowing it to accumulate. In the presence of MAbs 256A-4 and 256A-8, Jagged-1-induced metalloprotease cleavage of Notch3 at S2 was inhibited (data not shown).

Example 11

Efficacy Study Using Human Cancer Models in Xenograft Mice

[0249] A. Human Cancer Cells and Tumorigenic Cells

[0250] Human cancer cell lines with Notch3 expression such as HCC2429, HCC95 may be obtained from Academic Institutes, or from the ATCC. The 293T/PCDNA.TM. 3.1, and 293T/Notch3 (NC85) cells are generated by transfecting 293T with related genes and selecting with hygromycin as describe in previous sections. All cells are cultured in DMEM or RPMI 1640 medium with 10% fetal bovine serum, sodium pyruvate, nonessential amino acids, L-glutamine, vitamin solution, and penicillin-streptomycin (Flow Laboratories, Rockville, Md.). Cell lines are incubated in a mixture of 5% CO.sub.2 and 95% air at 37.degree. C. in an incubator. Cultures are maintained for no longer than 3 weeks after recovery from frozen stocks. Logarithmically growing single-cell suspensions cells with .gtoreq.90% viability are used for tumor cells injection after washing with PBS.

[0251] B. Animals

[0252] Mice are obtained from, for example, the Animal Production Area of the National Cancer Institute at Frederick Cancer Research and Development Center, Frederick, Md. The animals are purpose-bred and are experimentally naive at the outset of the study. Mice selected for use in the studies are chosen to be as uniform in age and weight as possible. They are 6-8 weeks of age and their body weights at initiation of weight range from approximately 18 to 25 grams. Records of the dates of birth for the animals used in this study are retained in the study raw data, and the weight range at the time of group assignment is specified in the report. Each animal is identified by a numbered ear tag. The animals are group housed by treatment group (4 mice/cage) in polystyrene disposable shoe-box cages containing cellulose bedding, meeting or exceeding NIH guidelines. During the course of the study, the environmental conditions in the animal room is monitored and maintained within a temperature range of 18-26.degree. C., and the relative humidity is recorded daily. A 12-hour light/dark illumination cycle is maintained throughout the study. Animals have irradiated food. No contaminants are known to be present in the food at levels that would interfere with the results of this study. Autoclaved water is available to each animal via water bottles. No contaminants are known to be present in the water at levels that would interfere with the results of this study. Prior to assignment to the study, all study animals are acclimatized to their designated housing for at least 7 days prior to the first day of dosing.

[0253] C. Tumor Models and Efficacy Studies

[0254] Mice are anesthetized using sodium pentobarbital (50 mg/kg body weight) and placed in the right lateral decubitus position. Cancer cells, such as non-small cell lung cancer (NSCLC) cell lines, HCC2429 (Haruki, et al. Cancer Res. 65:3555 (2005)), HCC95 (From Dr. John Mina), and H2122 (ATCC No. CRL5985), in 50 .mu.l Hank's containing 10% MATRIGEL.TM. matrix are injected into the left lobe of the lungs. After the tumor-cell injection, the mice are turned to the left lateral decubitus position and observed for 45-60 min until they recover fully. Records of tumor cell injections are maintained in the raw study data.

[0255] All animals are observed within their cages at least once daily during study and clinical findings recorded in the study raw data. Animals that show pronounced detrimental effects may be removed from the study should it be deemed necessary. Body weight is measured once each week during the treatment. Cancer tissues from each mouse, where available, are harvested and stored for potential future biological characterization.

Example 12

Assay for Notch3 Related Diseases

[0256] To identify other Notch3 related diseases, one can sequence the Notch3 gene from patient samples, perform FISH (fluorescence in situ hybridization) and CGH (comparative genomic hybridization) analysis to look for translocation and gene amplification using patient cells, or perform immunohistochemistry to check for the over-expression of Notch3 receptor using patient tissue or tumor sections. In addition, one can isolate and culture cells from a patient suspected of having a Notch3 associated disease and study the impact of an antagonistic antibody of the present invention on cell migration, invasion, survival and proliferation. Protocols for cell migration and invasion assay are described in Example 7 and the protocol for an apoptosis assay is described in Example 6. For the cell proliferation assay, cells cultured from patient samples are be seeded in 96-well plate coated with and without Notch ligands. Antagonistic antibodies are added at the beginning of the culture. Cell numbers are counted at specific time points using trypan blue staining. Notch3 FISH and CGH analysis may be performed using the published protocols of Park, et al. (Cancer Res, 66: 12 (2006)).

[0257] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Sequence CWU 1

1

4812321PRTHomo sapiens 1Met Gly Pro Gly Ala Arg Gly Arg Arg Arg Arg Arg Arg Pro Met Ser 1 5 10 15 Pro Pro Pro Pro Pro Pro Pro Val Arg Ala Leu Pro Leu Leu Leu Leu 20 25 30 Leu Ala Gly Pro Gly Ala Ala Ala Pro Pro Cys Leu Asp Gly Ser Pro 35 40 45 Cys Ala Asn Gly Gly Arg Cys Thr Gln Leu Pro Ser Arg Glu Ala Ala 50 55 60 Cys Leu Cys Pro Pro Gly Trp Val Gly Glu Arg Cys Gln Leu Glu Asp 65 70 75 80 Pro Cys His Ser Gly Pro Cys Ala Gly Arg Gly Val Cys Gln Ser Ser 85 90 95 Val Val Ala Gly Thr Ala Arg Phe Ser Cys Arg Cys Pro Arg Gly Phe 100 105 110 Arg Gly Pro Asp Cys Ser Leu Pro Asp Pro Cys Leu Ser Ser Pro Cys 115 120 125 Ala His Gly Ala Arg Cys Ser Val Gly Pro Asp Gly Arg Phe Leu Cys 130 135 140 Ser Cys Pro Pro Gly Tyr Gln Gly Arg Ser Cys Arg Ser Asp Val Asp 145 150 155 160 Glu Cys Arg Val Gly Glu Pro Cys Arg His Gly Gly Thr Cys Leu Asn 165 170 175 Thr Pro Gly Ser Phe Arg Cys Gln Cys Pro Ala Gly Tyr Thr Gly Pro 180 185 190 Leu Cys Glu Asn Pro Ala Val Pro Cys Ala Pro Ser Pro Cys Arg Asn 195 200 205 Gly Gly Thr Cys Arg Gln Ser Gly Asp Leu Thr Tyr Asp Cys Ala Cys 210 215 220 Leu Pro Gly Phe Glu Gly Gln Asn Cys Glu Val Asn Val Asp Asp Cys 225 230 235 240 Pro Gly His Arg Cys Leu Asn Gly Gly Thr Cys Val Asp Gly Val Asn 245 250 255 Thr Tyr Asn Cys Gln Cys Pro Pro Glu Trp Thr Gly Gln Phe Cys Thr 260 265 270 Glu Asp Val Asp Glu Cys Gln Leu Gln Pro Asn Ala Cys His Asn Gly 275 280 285 Gly Thr Cys Phe Asn Thr Leu Gly Gly His Ser Cys Val Cys Val Asn 290 295 300 Gly Trp Thr Gly Glu Ser Cys Ser Gln Asn Ile Asp Asp Cys Ala Thr 305 310 315 320 Ala Val Cys Phe His Gly Ala Thr Cys His Asp Arg Val Ala Ser Phe 325 330 335 Tyr Cys Ala Cys Pro Met Gly Lys Thr Gly Leu Leu Cys His Leu Asp 340 345 350 Asp Ala Cys Val Ser Asn Pro Cys His Glu Asp Ala Ile Cys Asp Thr 355 360 365 Asn Pro Val Asn Gly Arg Ala Ile Cys Thr Cys Pro Pro Gly Phe Thr 370 375 380 Gly Gly Ala Cys Asp Gln Asp Val Asp Glu Cys Ser Ile Gly Ala Asn 385 390 395 400 Pro Cys Glu His Leu Gly Arg Cys Val Asn Thr Gln Gly Ser Phe Leu 405 410 415 Cys Gln Cys Gly Arg Gly Tyr Thr Gly Pro Arg Cys Glu Thr Asp Val 420 425 430 Asn Glu Cys Leu Ser Gly Pro Cys Arg Asn Gln Ala Thr Cys Leu Asp 435 440 445 Arg Ile Gly Gln Phe Thr Cys Ile Cys Met Ala Gly Phe Thr Gly Thr 450 455 460 Tyr Cys Glu Val Asp Ile Asp Glu Cys Gln Ser Ser Pro Cys Val Asn 465 470 475 480 Gly Gly Val Cys Lys Asp Arg Val Asn Gly Phe Ser Cys Thr Cys Pro 485 490 495 Ser Gly Phe Ser Gly Ser Thr Cys Gln Leu Asp Val Asp Glu Cys Ala 500 505 510 Ser Thr Pro Cys Arg Asn Gly Ala Lys Cys Val Asp Gln Pro Asp Gly 515 520 525 Tyr Glu Cys Arg Cys Ala Glu Gly Phe Glu Gly Thr Leu Cys Asp Arg 530 535 540 Asn Val Asp Asp Cys Ser Pro Asp Pro Cys His His Gly Arg Cys Val 545 550 555 560 Asp Gly Ile Ala Ser Phe Ser Cys Ala Cys Ala Pro Gly Tyr Thr Gly 565 570 575 Thr Arg Cys Glu Ser Gln Val Asp Glu Cys Arg Ser Gln Pro Cys Arg 580 585 590 His Gly Gly Lys Cys Leu Asp Leu Val Asp Lys Tyr Leu Cys Arg Cys 595 600 605 Pro Ser Gly Thr Thr Gly Val Asn Cys Glu Val Asn Ile Asp Asp Cys 610 615 620 Ala Ser Asn Pro Cys Thr Phe Gly Val Cys Arg Asp Gly Ile Asn Arg 625 630 635 640 Tyr Asp Cys Val Cys Gln Pro Gly Phe Thr Gly Pro Leu Cys Asn Val 645 650 655 Glu Ile Asn Glu Cys Ala Ser Ser Pro Cys Gly Glu Gly Gly Ser Cys 660 665 670 Val Asp Gly Glu Asn Gly Phe Arg Cys Leu Cys Pro Pro Gly Ser Leu 675 680 685 Pro Pro Leu Cys Leu Pro Pro Ser His Pro Cys Ala His Glu Pro Cys 690 695 700 Ser His Gly Ile Cys Tyr Asp Ala Pro Gly Gly Phe Arg Cys Val Cys 705 710 715 720 Glu Pro Gly Trp Ser Gly Pro Arg Cys Ser Gln Ser Leu Ala Arg Asp 725 730 735 Ala Cys Glu Ser Gln Pro Cys Arg Ala Gly Gly Thr Cys Ser Ser Asp 740 745 750 Gly Met Gly Phe His Cys Thr Cys Pro Pro Gly Val Gln Gly Arg Gln 755 760 765 Cys Glu Leu Leu Ser Pro Cys Thr Pro Asn Pro Cys Glu His Gly Gly 770 775 780 Arg Cys Glu Ser Ala Pro Gly Gln Leu Pro Val Cys Ser Cys Pro Gln 785 790 795 800 Gly Trp Gln Gly Pro Arg Cys Gln Gln Asp Val Asp Glu Cys Ala Gly 805 810 815 Pro Ala Pro Cys Gly Pro His Gly Ile Cys Thr Asn Leu Ala Gly Ser 820 825 830 Phe Ser Cys Thr Cys His Gly Gly Tyr Thr Gly Pro Ser Cys Asp Gln 835 840 845 Asp Ile Asn Asp Cys Asp Pro Asn Pro Cys Leu Asn Gly Gly Ser Cys 850 855 860 Gln Asp Gly Val Gly Ser Phe Ser Cys Ser Cys Leu Pro Gly Phe Ala 865 870 875 880 Gly Pro Arg Cys Ala Arg Asp Val Asp Glu Cys Leu Ser Asn Pro Cys 885 890 895 Gly Pro Gly Thr Cys Thr Asp His Val Ala Ser Phe Thr Cys Thr Cys 900 905 910 Pro Pro Gly Tyr Gly Gly Phe His Cys Glu Gln Asp Leu Pro Asp Cys 915 920 925 Ser Pro Ser Ser Cys Phe Asn Gly Gly Thr Cys Val Asp Gly Val Asn 930 935 940 Ser Phe Ser Cys Leu Cys Arg Pro Gly Tyr Thr Gly Ala His Cys Gln 945 950 955 960 His Glu Ala Asp Pro Cys Leu Ser Arg Pro Cys Leu His Gly Gly Val 965 970 975 Cys Ser Ala Ala His Pro Gly Phe Arg Cys Thr Cys Leu Glu Ser Phe 980 985 990 Thr Gly Pro Gln Cys Gln Thr Leu Val Asp Trp Cys Ser Arg Gln Pro 995 1000 1005 Cys Gln Asn Gly Gly Arg Cys Val Gln Thr Gly Ala Tyr Cys Leu 1010 1015 1020 Cys Pro Pro Gly Trp Ser Gly Arg Leu Cys Asp Ile Arg Ser Leu 1025 1030 1035 Pro Cys Arg Glu Ala Ala Ala Gln Ile Gly Val Arg Leu Glu Gln 1040 1045 1050 Leu Cys Gln Ala Gly Gly Gln Cys Val Asp Glu Asp Ser Ser His 1055 1060 1065 Tyr Cys Val Cys Pro Glu Gly Arg Thr Gly Ser His Cys Glu Gln 1070 1075 1080 Glu Val Asp Pro Cys Leu Ala Gln Pro Cys Gln His Gly Gly Thr 1085 1090 1095 Cys Arg Gly Tyr Met Gly Gly Tyr Met Cys Glu Cys Leu Pro Gly 1100 1105 1110 Tyr Asn Gly Asp Asn Cys Glu Asp Asp Val Asp Glu Cys Ala Ser 1115 1120 1125 Gln Pro Cys Gln His Gly Gly Ser Cys Ile Asp Leu Val Ala Arg 1130 1135 1140 Tyr Leu Cys Ser Cys Pro Pro Gly Thr Leu Gly Val Leu Cys Glu 1145 1150 1155 Ile Asn Glu Asp Asp Cys Gly Pro Gly Pro Pro Leu Asp Ser Gly 1160 1165 1170 Pro Arg Cys Leu His Asn Gly Thr Cys Val Asp Leu Val Gly Gly 1175 1180 1185 Phe Arg Cys Thr Cys Pro Pro Gly Tyr Thr Gly Leu Arg Cys Glu 1190 1195 1200 Ala Asp Ile Asn Glu Cys Arg Ser Gly Ala Cys His Ala Ala His 1205 1210 1215 Thr Arg Asp Cys Leu Gln Asp Pro Gly Gly Gly Phe Arg Cys Leu 1220 1225 1230 Cys His Ala Gly Phe Ser Gly Pro Arg Cys Gln Thr Val Leu Ser 1235 1240 1245 Pro Cys Glu Ser Gln Pro Cys Gln His Gly Gly Gln Cys Arg Pro 1250 1255 1260 Ser Pro Gly Pro Gly Gly Gly Leu Thr Phe Thr Cys His Cys Ala 1265 1270 1275 Gln Pro Phe Trp Gly Pro Arg Cys Glu Arg Val Ala Arg Ser Cys 1280 1285 1290 Arg Glu Leu Gln Cys Pro Val Gly Val Pro Cys Gln Gln Thr Pro 1295 1300 1305 Arg Gly Pro Arg Cys Ala Cys Pro Pro Gly Leu Ser Gly Pro Ser 1310 1315 1320 Cys Arg Ser Phe Pro Gly Ser Pro Pro Gly Ala Ser Asn Ala Ser 1325 1330 1335 Cys Ala Ala Ala Pro Cys Leu His Gly Gly Ser Cys Arg Pro Ala 1340 1345 1350 Pro Leu Ala Pro Phe Phe Arg Cys Ala Cys Ala Gln Gly Trp Thr 1355 1360 1365 Gly Pro Arg Cys Glu Ala Pro Ala Ala Ala Pro Glu Val Ser Glu 1370 1375 1380 Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Ala Lys Arg Gly Asp 1385 1390 1395 Gln Arg Cys Asp Arg Glu Cys Asn Ser Pro Gly Cys Gly Trp Asp 1400 1405 1410 Gly Gly Asp Cys Ser Leu Ser Val Gly Asp Pro Trp Arg Gln Cys 1415 1420 1425 Glu Ala Leu Gln Cys Trp Arg Leu Phe Asn Asn Ser Arg Cys Asp 1430 1435 1440 Pro Ala Cys Ser Ser Pro Ala Cys Leu Tyr Asp Asn Phe Asp Cys 1445 1450 1455 His Ala Gly Gly Arg Glu Arg Thr Cys Asn Pro Val Tyr Glu Lys 1460 1465 1470 Tyr Cys Ala Asp His Phe Ala Asp Gly Arg Cys Asp Gln Gly Cys 1475 1480 1485 Asn Thr Glu Glu Cys Gly Trp Asp Gly Leu Asp Cys Ala Ser Glu 1490 1495 1500 Val Pro Ala Leu Leu Ala Arg Gly Val Leu Val Leu Thr Val Leu 1505 1510 1515 Leu Pro Pro Glu Glu Leu Leu Arg Ser Ser Ala Asp Phe Leu Gln 1520 1525 1530 Arg Leu Ser Ala Ile Leu Arg Thr Ser Leu Arg Phe Arg Leu Asp 1535 1540 1545 Ala His Gly Gln Ala Met Val Phe Pro Tyr His Arg Pro Ser Pro 1550 1555 1560 Gly Ser Glu Pro Arg Ala Arg Arg Glu Leu Ala Pro Glu Val Ile 1565 1570 1575 Gly Ser Val Val Met Leu Glu Ile Asp Asn Arg Leu Cys Leu Gln 1580 1585 1590 Ser Pro Glu Asn Asp His Cys Phe Pro Asp Ala Gln Ser Ala Ala 1595 1600 1605 Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg Leu Asp Phe Pro 1610 1615 1620 Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu Pro Pro Glu 1625 1630 1635 Pro Ser Val Pro Leu Leu Pro Leu Leu Val Ala Gly Ala Val Leu 1640 1645 1650 Leu Leu Val Ile Leu Val Leu Gly Val Met Val Ala Arg Arg Lys 1655 1660 1665 Arg Glu His Ser Thr Leu Trp Phe Pro Glu Gly Phe Ser Leu His 1670 1675 1680 Lys Asp Val Ala Ser Gly His Lys Gly Arg Arg Glu Pro Val Gly 1685 1690 1695 Gln Asp Ala Leu Gly Met Lys Asn Met Ala Lys Gly Glu Ser Leu 1700 1705 1710 Met Gly Glu Val Ala Thr Asp Trp Met Asp Thr Glu Cys Pro Glu 1715 1720 1725 Ala Lys Arg Leu Lys Val Glu Glu Pro Gly Met Gly Ala Glu Glu 1730 1735 1740 Ala Val Asp Cys Arg Gln Trp Thr Gln His His Leu Val Ala Ala 1745 1750 1755 Asp Ile Arg Val Ala Pro Ala Met Ala Leu Thr Pro Pro Gln Gly 1760 1765 1770 Asp Ala Asp Ala Asp Gly Met Asp Val Asn Val Arg Gly Pro Asp 1775 1780 1785 Gly Phe Thr Pro Leu Met Leu Ala Ser Phe Cys Gly Gly Ala Leu 1790 1795 1800 Glu Pro Met Pro Thr Glu Glu Asp Glu Ala Asp Asp Thr Ser Ala 1805 1810 1815 Ser Ile Ile Ser Asp Leu Ile Cys Gln Gly Ala Gln Leu Gly Ala 1820 1825 1830 Arg Thr Asp Arg Thr Gly Glu Thr Ala Leu His Leu Ala Ala Arg 1835 1840 1845 Tyr Ala Arg Ala Asp Ala Ala Lys Arg Leu Leu Asp Ala Gly Ala 1850 1855 1860 Asp Thr Asn Ala Gln Asp His Ser Gly Arg Thr Pro Leu His Thr 1865 1870 1875 Ala Val Thr Ala Asp Ala Gln Gly Val Phe Gln Ile Leu Ile Arg 1880 1885 1890 Asn Arg Ser Thr Asp Leu Asp Ala Arg Met Ala Asp Gly Ser Thr 1895 1900 1905 Ala Leu Ile Leu Ala Ala Arg Leu Ala Val Glu Gly Met Val Glu 1910 1915 1920 Glu Leu Ile Ala Ser His Ala Asp Val Asn Ala Val Asp Glu Leu 1925 1930 1935 Gly Lys Ser Ala Leu His Trp Ala Ala Ala Val Asn Asn Val Glu 1940 1945 1950 Ala Thr Leu Ala Leu Leu Lys Asn Gly Ala Asn Lys Asp Met Gln 1955 1960 1965 Asp Ser Lys Glu Glu Thr Pro Leu Phe Leu Ala Ala Arg Glu Gly 1970 1975 1980 Ser Tyr Glu Ala Ala Lys Leu Leu Leu Asp His Phe Ala Asn Arg 1985 1990 1995 Glu Ile Thr Asp His Leu Asp Arg Leu Pro Arg Asp Val Ala Gln 2000 2005 2010 Glu Arg Leu His Gln Asp Ile Val Arg Leu Leu Asp Gln Pro Ser 2015 2020 2025 Gly Pro Arg Ser Pro Pro Gly Pro His Gly Leu Gly Pro Leu Leu 2030 2035 2040 Cys Pro Pro Gly Ala Phe Leu Pro Gly Leu Lys Ala Ala Gln Ser 2045 2050 2055 Gly Ser Lys Lys Ser Arg Arg Pro Pro Gly Lys Ala Gly Leu Gly 2060 2065 2070 Pro Gln Gly Pro Arg Gly Arg Gly Lys Lys Leu Thr Leu Ala Cys 2075 2080 2085 Pro Gly Pro Leu Ala Asp Ser Ser Val Thr Leu Ser Pro Val Asp 2090 2095 2100 Ser Leu Asp Ser Pro Arg Pro Phe Gly Gly Pro Pro Ala Ser Pro 2105 2110 2115 Gly Gly Phe Pro Leu Glu Gly Pro Tyr Ala Ala Ala Thr Ala Thr 2120 2125 2130 Ala Val Ser Leu Ala Gln Leu Gly Gly Pro Gly Arg Ala Gly Leu 2135 2140 2145 Gly Arg Gln Pro Pro Gly Gly Cys Val Leu Ser Leu Gly Leu Leu 2150 2155 2160 Asn Pro Val Ala Val Pro Leu Asp Trp Ala Arg Leu Pro Pro Pro 2165 2170 2175 Ala Pro Pro Gly Pro Ser Phe Leu Leu Pro Leu Ala Pro Gly Pro 2180 2185 2190 Gln Leu Leu Asn Pro Gly Thr Pro Val Ser Pro Gln Glu Arg Pro 2195 2200 2205 Pro Pro Tyr Leu Ala Val Pro Gly His Gly Glu Glu Tyr Pro Val 2210 2215 2220 Ala Gly Ala His Ser Ser Pro Pro Lys Ala Arg Phe Leu Arg Val 2225 2230 2235 Pro Ser Glu His Pro Tyr Leu

Thr Pro Ser Pro Glu Ser Pro Glu 2240 2245 2250 His Trp Ala Ser Pro Ser Pro Pro Ser Leu Ser Asp Trp Ser Glu 2255 2260 2265 Ser Thr Pro Ser Pro Ala Thr Ala Thr Gly Ala Met Ala Thr Thr 2270 2275 2280 Thr Gly Ala Leu Pro Ala Gln Pro Leu Pro Leu Ser Val Pro Ser 2285 2290 2295 Ser Leu Ala Gln Ala Gln Thr Gln Leu Gly Pro Gln Pro Glu Val 2300 2305 2310 Thr Pro Lys Arg Gln Val Leu Ala 2315 2320 2122PRTArtificial sequenceHEAVY CHAIN VARIABLE REGION OF MAB 256A-4 2Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly 1 5 10 15 Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser His Tyr 20 25 30 Tyr Met Ser Trp Val Arg Gln Thr Pro Glu Lys Arg Leu Glu Trp Val 35 40 45 Ala Tyr Ile Ser Asn Gly Gly Gly Arg Thr Asp Tyr Pro Asp Ser Val 50 55 60 Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Leu His 65 70 75 80 Leu Gln Met Ser Ser Leu Lys Ser Glu Asp Thr Ala Met Tyr Tyr Cys 85 90 95 Thr Arg Leu Asp Tyr Phe Gly Gly Ser Pro Tyr Phe Asp Tyr Trp Gly 100 105 110 Gln Gly Thr Thr Leu Thr Val Ser Ser Ala 115 120 3114PRTArtificial sequenceLIGHT CHAIN VARIABLE REGION OF MAB 256 A-4 3Glu Ile Val Leu Thr Gln Ser Pro Ala Ile Thr Ala Ala Ser Leu Gly 1 5 10 15 Gln Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met 20 25 30 His Trp Tyr Gln Gln Lys Ser Gly Thr Ser Pro Lys Pro Trp Ile Tyr 35 40 45 Glu Ile Ser Lys Leu Ala Ser Gly Val Pro Pro Arg Phe Ser Gly Ser 50 55 60 Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Ser Met Glu Ala Glu 65 70 75 80 Asp Ala Ala Ile Tyr Tyr Cys Gln Gln Trp Asn Tyr Pro Leu Ile Thr 85 90 95 Phe Gly Ser Gly Thr Lys Leu Glu Ile Lys Arg Ala Asp Ala Ala Pro 100 105 110 Thr Val 4122PRTArtificial sequenceHEAVY CHAIN VARIABLE REGION OF MAB 256 A-8 4Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly 1 5 10 15 Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser His Tyr 20 25 30 Tyr Met Ser Trp Val Arg Gln Thr Pro Glu Lys Arg Leu Glu Trp Val 35 40 45 Ala Tyr Ile Asn Ser Gly Gly Gly Arg Thr Asp Tyr Pro Asp Ser Val 50 55 60 Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Ala Lys Asn Thr Leu His 65 70 75 80 Leu Gln Met Ser Ser Leu Lys Ser Glu Asp Thr Ala Met Tyr Tyr Cys 85 90 95 Ala Arg Leu Asp Tyr Tyr Gly Gly Ser Pro Tyr Phe Asp Tyr Trp Gly 100 105 110 Gln Gly Thr Thr Leu Thr Val Ser Ser Ala 115 120 5114PRTArtificial sequenceLIGHT CHAIN VARIABLE REGION OF MAB 256 A-8 5Glu Ile Val Leu Thr Gln Ser Pro Ala Ile Thr Ala Ala Ser Leu Gly 1 5 10 15 Gln Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met 20 25 30 His Trp Tyr Gln Gln Lys Ser Gly Thr Ser Pro Lys Pro Trp Ile Tyr 35 40 45 Glu Ile Ser Lys Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser 50 55 60 Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Ser Met Glu Ala Glu 65 70 75 80 Asp Ala Ala Ile Tyr Tyr Cys Gln Gln Trp Asn Tyr Pro Leu Ile Thr 85 90 95 Phe Gly Ser Gly Thr Lys Leu Glu Ile Lys Arg Ala Asp Ala Ala Pro 100 105 110 Thr Val 645PRTArtificial sequenceMODIFIED NOTCH 3 LEADER SEQUENCE 6Met Gly Pro Gly Ala Arg Gly Arg Arg Arg Arg Arg Arg Pro Met Ser 1 5 10 15 Pro Pro Pro Pro Pro Pro Pro Val Arg Ala Leu Pro Leu Leu Leu Leu 20 25 30 Leu Ala Gly Pro Gly Ala Ala Ala Pro Pro Cys Leu Asp 35 40 45 755DNAArtificial sequenceOLIGONUCLEOTIDE PRIMER 7gctcgagctc gtgggaaaat accgtgggaa aatgaaccgt gggaaaatct cgtgg 55835DNAArtificial sequenceOLIGONUCLEOTIDE PRIMER 8gctcgagatt ttcccacgag attttcccac ggttc 35939PRTArtificial sequenceNOTCH 3 LIN 12 DOMAIN 9Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Ala Lys Arg Gly Asp Gln 1 5 10 15 Arg Cys Asp Arg Glu Cys Asn Ser Pro Gly Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Ser Val Gly 35 1039PRTArtificial sequenceNOTCH 3/ NOTCH1 LIN 12 DOMAIN SWAP (FIG 16 L1-SUB1) 10Glu Glu Ala Cys Glu Leu Pro Glu Cys Gln Ala Lys Arg Gly Asp Gln 1 5 10 15 Arg Cys Asp Arg Glu Cys Asn Ser Pro Gly Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Ser Val Gly 35 1139PRTArtificial sequenceNOTCH 3 / NOTCH 1 LIN 12 DOMAIN SWAP (FIG. 16 L1-SUB2) 11Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Glu Asp Ala Gly Asn Lys 1 5 10 15 Val Cys Ser Arg Glu Cys Asn Ser Pro Gly Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Ser Val Gly 35 1239PRTArtificial sequenceNOTCH 3 / NOTCH 1 LIN 12 DOMAIN SWAP (FIG. 16 L1-SUB3) 12Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Ala Lys Arg Gly Asp Gln 1 5 10 15 Arg Cys Asp Leu Gln Cys Asn Asn His Ala Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Asn Phe Asn 35 1339PRTArtificial sequenceNOTCH 3/ NOTCH 1 LIN12 AMINO ACID SWAP (FIG 16 L1-AA SWAP 1) 13Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Glu Asp Ala Gly Asp Gln 1 5 10 15 Arg Cys Asp Arg Glu Cys Asn Ser Pro Gly Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Ser Val Gly 35 1439PRTArtificial sequenceNOTCH 3/ NOTCH 1 LIN12 AMINO ACID SWAP (FIG 16 L1-AA SWAP 2) 14Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Ala Lys Arg Gly Asn Lys 1 5 10 15 Val Cys Asp Arg Glu Cys Asn Ser Pro Gly Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Ser Val Gly 35 1539PRTArtificial sequenceNOTCH 3/ NOTCH 1 LIN12 AMINO ACID SWAP (FIG 16 L1-AA SWAP 3) 15Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Ala Lys Arg Gly Asp Gln 1 5 10 15 Arg Cys Ser Leu Gln Cys Asn Ser Pro Gly Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Ser Val Gly 35 1639PRTArtificial sequenceNOTCH 3/ NOTCH 1 LIN12 AMINO ACID SWAP (FIG 16 L1-AA SWAP 4) 16Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Ala Lys Arg Gly Asp Gln 1 5 10 15 Arg Cys Asp Arg Glu Cys Asn Asn His Ala Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Ser Val Gly 35 1739PRTArtificial sequenceNOTCH 3/ NOTCH 1 LIN12 AMINO ACID SWAP (FIG 16 L1-AA SWAP 5) 17Glu Pro Arg Cys Pro Arg Ala Ala Cys Gln Ala Lys Arg Gly Asp Gln 1 5 10 15 Arg Cys Asp Arg Glu Cys Asn Ser Pro Gly Cys Gly Trp Asp Gly Gly 20 25 30 Asp Cys Ser Leu Asn Phe Asn 35 1869PRTArtificial sequenceNOTCH 3 DIMERIZATION DOMAIN 2 (FIG 17 D2) 18Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 1969PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN SWAP (FIG 17 D2-SUB1) 19Glu Leu Ala Pro Asp Val Arg Gly Ser Ile Val Tyr Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2069PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN SWAP (FIG 17 D2-SUB2) 20Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Gln Cys Val Gln Ala Ala Ala Ser Ser Gln Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2169PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN SWAP (FIG 17 D2-SUB3) 21Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Gln Ser 20 25 30 Ala Thr Asp Ala Ala Ala Phe Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2269PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN SWAP (FIG 17 D2-SUB4) 22Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ala Ser Leu Gly Ser 35 40 45 Leu Asn Ile Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2369PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN SWAP (FIG 17 D2-SUB5) 23Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Lys Ile Glu Ala Val Gln Ser Glu Thr Val Glu 50 55 60 Pro Pro Ala Pro Ser 65 2469PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP1) 24Glu Leu Ala Pro Asp Val Arg Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2569PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP2) 25Glu Leu Ala Pro Glu Val Ile Gly Ser Ile Val Tyr Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2669PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP3) 26Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ala Ser Leu Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2769PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP4) 27Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Gly Ser 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2869PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP5) 28Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asn Ile Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 2969PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP6) 29Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Lys Ile Glu Asp Val Arg Gly Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 3069PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP7) 30Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Ala Val Gln Ser Glu Pro Leu Glu 50 55 60 Pro Pro Glu Pro Ser 65 3169PRTArtificial sequenceNOTCH 3/ NOTCH 1 DIMERIZATION DOMAIN AMINO ACID SWAP (FIG 17 D2-AA SWAP8) 31Glu Leu Ala Pro Glu Val Ile Gly Ser Val Val Met Leu Glu Ile Asp 1 5 10 15 Asn Arg Leu Cys Leu Gln Ser Pro Glu Asn Asp His Cys Phe Pro Asp 20 25 30 Ala Gln Ser Ala Ala Asp Tyr Leu Gly Ala Leu Ser Ala Val Glu Arg 35 40 45 Leu Asp Phe Pro Tyr Pro Leu Arg Asp Val Arg Gly Glu Thr Val Glu 50 55 60 Pro Pro Ala Pro Ser 65 3210PRTArtificial sequenceCDR-H1 OF MAB 256A-4 32Gly Phe Thr Phe Ser His Tyr Tyr Met Ser 1 5 10 3312PRTArtificial sequenceCDR-H2 OF MAB 256A-4 33Ile Ser Asn Gly Gly Gly Arg Thr Asp Tyr

Pro Asp 1 5 10 3413PRTArtificial sequenceCDR-H3 OF MAB 256A-4 34Arg Leu Asp Tyr Phe Gly Gly Ser Pro Tyr Phe Asp Tyr 1 5 10 3510PRTArtificial sequenceCDR-L1 OF MAB 256A-4 35Ser Ala Ser Ser Ser Val Ser Tyr Met His 1 5 10 367PRTArtificial sequenceCDR-L2 OF MAB 256A-4 36Glu Ile Ser Lys Leu Ala Ser 1 5 379PRTArtificial sequenceCDR-L3 OF MAB 256A-4 37Gln Gln Trp Asn Tyr Pro Leu Ile Thr 1 5 3810PRTArtificial sequenceCDR-H1 OF MAB 256A-8 38Gly Phe Thr Phe Ser His Tyr Tyr Met Ser 1 5 10 3912PRTArtificial sequenceCDR-H2 OF MAB 256A-8 39Tyr Ile Asn Ser Gly Gly Gly Arg Thr Asp Tyr Pro 1 5 10 4012PRTArtificial sequenceCDR-H3 OF MAB 256A-8 40Leu Asp Tyr Tyr Gly Gly Ser Pro Tyr Phe Asp Tyr 1 5 10 4110PRTArtificial sequenceCDR-L1 OF MAB 256A-8 41Ser Ala Ser Ser Ser Val Ser Tyr Met His 1 5 10 428PRTArtificial sequenceCDR-L2 OF MAB 256A-8 42Tyr Glu Ile Ser Lys Leu Ala Ser 1 5 439PRTArtificial sequenceCDR-L3 OF MAB 256A-8 43Gln Gln Trp Asn Tyr Pro Leu Ile Thr 1 5 442556PRTHomo sapienshuman Notch1 44Met Pro Pro Leu Leu Ala Pro Leu Leu Cys Leu Ala Leu Leu Pro1 5 10 15Ala Leu Ala Ala Arg Gly Pro Arg Cys Ser Gln Pro Gly Glu Thr 20 25 30Cys Leu Asn Gly Gly Lys Cys Glu Ala Ala Asn Gly Thr Glu Ala 35 40 45Cys Val Cys Gly Gly Ala Phe Val Gly Pro Arg Cys Gln Asp Pro 50 55 60Asn Pro Cys Leu Ser Thr Pro Cys Lys Asn Ala Gly Thr Cys His 65 70 75Val Val Asp Arg Arg Gly Val Ala Asp Tyr Ala Cys Ser Cys Ala 80 85 90Leu Gly Phe Ser Gly Pro Leu Cys Leu Thr Pro Leu Asp Asn Ala 95 100 105Cys Leu Thr Asn Pro Cys Arg Asn Gly Gly Thr Cys Asp Leu Leu 110 115 120Thr Leu Thr Glu Tyr Lys Cys Arg Cys Pro Pro Gly Trp Ser Gly 125 130 135Lys Ser Cys Gln Gln Ala Asp Pro Cys Ala Ser Asn Pro Cys Ala 140 145 150Asn Gly Gly Gln Cys Leu Pro Phe Glu Ala Ser Tyr Ile Cys His 155 160 165Cys Pro Pro Ser Phe His Gly Pro Thr Cys Arg Gln Asp Val Asn 170 175 180Glu Cys Gly Gln Lys Pro Gly Leu Cys Arg His Gly Gly Thr Cys 185 190 195His Asn Glu Val Gly Ser Tyr Arg Cys Val Cys Arg Ala Thr His 200 205 210Thr Gly Pro Asn Cys Glu Arg Pro Tyr Val Pro Cys Ser Pro Ser 215 220 225Pro Cys Gln Asn Gly Gly Thr Cys Arg Pro Thr Gly Asp Val Thr 230 235 240His Glu Cys Ala Cys Leu Pro Gly Phe Thr Gly Gln Asn Cys Glu 245 250 255Glu Asn Ile Asp Asp Cys Pro Gly Asn Asn Cys Lys Asn Gly Gly 260 265 270Ala Cys Val Asp Gly Val Asn Thr Tyr Asn Cys Arg Cys Pro Pro 275 280 285Glu Trp Thr Gly Gln Tyr Cys Thr Glu Asp Val Asp Glu Cys Gln 290 295 300Leu Met Pro Asn Ala Cys Gln Asn Gly Gly Thr Cys His Asn Thr 305 310 315His Gly Gly Tyr Asn Cys Val Cys Val Asn Gly Trp Thr Gly Glu 320 325 330Asp Cys Ser Glu Asn Ile Asp Asp Cys Ala Ser Ala Ala Cys Phe 335 340 345His Gly Ala Thr Cys His Asp Arg Val Ala Ser Phe Tyr Cys Glu 350 355 360Cys Pro His Gly Arg Thr Gly Leu Leu Cys His Leu Asn Asp Ala 365 370 375Cys Ile Ser Asn Pro Cys Asn Glu Gly Ser Asn Cys Asp Thr Asn 380 385 390Pro Val Asn Gly Lys Ala Ile Cys Thr Cys Pro Ser Gly Tyr Thr 395 400 405Gly Pro Ala Cys Ser Gln Asp Val Asp Glu Cys Ser Leu Gly Ala 410 415 420Asn Pro Cys Glu His Ala Gly Lys Cys Ile Asn Thr Leu Gly Ser 425 430 435Phe Glu Cys Gln Cys Leu Gln Gly Tyr Thr Gly Pro Arg Cys Glu 440 445 450Ile Asp Val Asn Glu Cys Val Ser Asn Pro Cys Gln Asn Asp Ala 455 460 465Thr Cys Leu Asp Gln Ile Gly Glu Phe Gln Cys Ile Cys Met Pro 470 475 480Gly Tyr Glu Gly Val His Cys Glu Val Asn Thr Asp Glu Cys Ala 485 490 495Ser Ser Pro Cys Leu His Asn Gly Arg Cys Leu Asp Lys Ile Asn 500 505 510Glu Phe Gln Cys Glu Cys Pro Thr Gly Phe Thr Gly His Leu Cys 515 520 525Gln Tyr Asp Val Asp Glu Cys Ala Ser Thr Pro Cys Lys Asn Gly 530 535 540Ala Lys Cys Leu Asp Gly Pro Asn Thr Tyr Thr Cys Val Cys Thr 545 550 555Glu Gly Tyr Thr Gly Thr His Cys Glu Val Asp Ile Asp Glu Cys 560 565 570Asp Pro Asp Pro Cys His Tyr Gly Ser Cys Lys Asp Gly Val Ala 575 580 585Thr Phe Thr Cys Leu Cys Arg Pro Gly Tyr Thr Gly His His Cys 590 595 600Glu Thr Asn Ile Asn Glu Cys Ser Ser Gln Pro Cys Arg His Gly 605 610 615Gly Thr Cys Gln Asp Arg Asp Asn Ala Tyr Leu Cys Phe Cys Leu 620 625 630Lys Gly Thr Thr Gly Pro Asn Cys Glu Ile Asn Leu Asp Asp Cys 635 640 645Ala Ser Ser Pro Cys Asp Ser Gly Thr Cys Leu Asp Lys Ile Asp 650 655 660Gly Tyr Glu Cys Ala Cys Glu Pro Gly Tyr Thr Gly Ser Met Cys 665 670 675Asn Ile Asn Ile Asp Glu Cys Ala Gly Asn Pro Cys His Asn Gly 680 685 690Gly Thr Cys Glu Asp Gly Ile Asn Gly Phe Thr Cys Arg Cys Pro 695 700 705Glu Gly Tyr His Asp Pro Thr Cys Leu Ser Glu Val Asn Glu Cys 710 715 720Asn Ser Asn Pro Cys Val His Gly Ala Cys Arg Asp Ser Leu Asn 725 730 735Gly Tyr Lys Cys Asp Cys Asp Pro Gly Trp Ser Gly Thr Asn Cys 740 745 750Asp Ile Asn Asn Asn Glu Cys Glu Ser Asn Pro Cys Val Asn Gly 755 760 765Gly Thr Cys Lys Asp Met Thr Ser Gly Tyr Val Cys Thr Cys Arg 770 775 780Glu Gly Phe Ser Gly Pro Asn Cys Gln Thr Asn Ile Asn Glu Cys 785 790 795Ala Ser Asn Pro Cys Leu Asn Gln Gly Thr Cys Ile Asp Asp Val 800 805 810Ala Gly Tyr Lys Cys Asn Cys Leu Leu Pro Tyr Thr Gly Ala Thr 815 820 825Cys Glu Val Val Leu Ala Pro Cys Ala Pro Ser Pro Cys Arg Asn 830 835 840Gly Gly Glu Cys Arg Gln Ser Glu Asp Tyr Glu Ser Phe Ser Cys 845 850 855Val Cys Pro Thr Gly Trp Gln Ala Gly Gln Thr Cys Glu Val Asp 860 865 870Ile Asn Glu Cys Val Leu Ser Pro Cys Arg His Gly Ala Ser Cys 875 880 885Gln Asn Thr His Gly Gly Tyr Arg Cys His Cys Gln Ala Gly Tyr 890 895 900Ser Gly Arg Asn Cys Glu Thr Asp Ile Asp Asp Cys Arg Pro Asn 905 910 915Pro Cys His Asn Gly Gly Ser Cys Thr Asp Gly Ile Asn Thr Ala 920 925 930Phe Cys Asp Cys Leu Pro Gly Phe Arg Gly Thr Phe Cys Glu Glu 935 940 945Asp Ile Asn Glu Cys Ala Ser Asp Pro Cys Arg Asn Gly Ala Asn 950 955 960Cys Thr Asp Cys Val Asp Ser Tyr Thr Cys Thr Cys Pro Ala Gly 965 970 975Phe Ser Gly Ile His Cys Glu Asn Asn Thr Pro Asp Cys Thr Glu 980 985 990Ser Ser Cys Phe Asn Gly Gly Thr Cys Val Asp Gly Ile Asn Ser 995 1000 1005Phe Thr Cys Leu Cys Pro Pro Gly Phe Thr Gly Ser Tyr Cys Gln 1010 1015 1020His Asp Val Asn Glu Cys Asp Ser Gln Pro Cys Leu His Gly Gly 1025 1030 1035Thr Cys Gln Asp Gly Cys Gly Ser Tyr Arg Cys Thr Cys Pro Gln 1040 1045 1050Gly Tyr Thr Gly Pro Asn Cys Gln Asn Leu Val His Trp Cys Asp 1055 1060 1065Ser Ser Pro Cys Lys Asn Gly Gly Lys Cys Trp Gln Thr His Thr 1070 1075 1080Gln Tyr Arg Cys Glu Cys Pro Ser Gly Trp Thr Gly Leu Tyr Cys 1085 1090 1095Asp Val Pro Ser Val Ser Cys Glu Val Ala Ala Gln Arg Gln Gly 1100 1105 1110Val Asp Val Ala Arg Leu Cys Gln His Gly Gly Leu Cys Val Asp 1115 1120 1125Ala Gly Asn Thr His His Cys Arg Cys Gln Ala Gly Tyr Thr Gly 1130 1135 1140Ser Tyr Cys Glu Asp Leu Val Asp Glu Cys Ser Pro Ser Pro Cys 1145 1150 1155Gln Asn Gly Ala Thr Cys Thr Asp Tyr Leu Gly Gly Tyr Ser Cys 1160 1165 1170Lys Cys Val Ala Gly Tyr His Gly Val Asn Cys Ser Glu Glu Ile 1175 1180 1185Asp Glu Cys Leu Ser His Pro Cys Gln Asn Gly Gly Thr Cys Leu 1190 1195 1200Asp Leu Pro Asn Thr Tyr Lys Cys Ser Cys Pro Arg Gly Thr Gln 1205 1210 1215Gly Val His Cys Glu Ile Asn Val Asp Asp Cys Asn Pro Pro Val 1220 1225 1230Asp Pro Val Ser Arg Ser Pro Lys Cys Phe Asn Asn Gly Thr Cys 1235 1240 1245Val Asp Gln Val Gly Gly Tyr Ser Cys Thr Cys Pro Pro Gly Phe 1250 1255 1260Val Gly Glu Arg Cys Glu Gly Asp Val Asn Glu Cys Leu Ser Asn 1265 1270 1275Pro Cys Asp Ala Arg Gly Thr Gln Asn Cys Val Gln Arg Val Asn 1280 1285 1290Asp Phe His Cys Glu Cys Arg Ala Gly His Thr Gly Arg Arg Cys 1295 1300 1305Glu Ser Val Ile Asn Gly Cys Lys Gly Lys Pro Cys Lys Asn Gly 1310 1315 1320Gly Thr Cys Ala Val Ala Ser Asn Thr Ala Arg Gly Phe Ile Cys 1325 1330 1335Lys Cys Pro Ala Gly Phe Glu Gly Ala Thr Cys Glu Asn Asp Ala 1340 1345 1350Arg Thr Cys Gly Ser Leu Arg Cys Leu Asn Gly Gly Thr Cys Ile 1355 1360 1365Ser Gly Pro Arg Ser Pro Thr Cys Leu Cys Leu Gly Pro Phe Thr 1370 1375 1380Gly Pro Glu Cys Gln Phe Pro Ala Ser Ser Pro Cys Leu Gly Gly 1385 1390 1395Asn Pro Cys Tyr Asn Gln Gly Thr Cys Glu Pro Thr Ser Glu Ser 1400 1405 1410Pro Phe Tyr Arg Cys Leu Cys Pro Ala Lys Phe Asn Gly Leu Leu 1415 1420 1425Cys His Ile Leu Asp Tyr Ser Phe Gly Gly Gly Ala Gly Arg Asp 1430 1435 1440Ile Pro Pro Pro Leu Ile Glu Glu Ala Cys Glu Leu Pro Glu Cys 1445 1450 1455Gln Glu Asp Ala Gly Asn Lys Val Cys Ser Leu Gln Cys Asn Asn 1460 1465 1470His Ala Cys Gly Trp Asp Gly Gly Asp Cys Ser Leu Asn Phe Asn 1475 1480 1485Asp Pro Trp Lys Asn Cys Thr Gln Ser Leu Gln Cys Trp Lys Tyr 1490 1495 1500Phe Ser Asp Gly His Cys Asp Ser Gln Cys Asn Ser Ala Gly Cys 1505 1510 1515Leu Phe Asp Gly Phe Asp Cys Gln Arg Ala Glu Gly Gln Cys Asn 1520 1525 1530Pro Leu Tyr Asp Gln Tyr Cys Lys Asp His Phe Ser Asp Gly His 1535 1540 1545Cys Asp Gln Gly Cys Asn Ser Ala Glu Cys Glu Trp Asp Gly Leu 1550 1555 1560Asp Cys Ala Glu His Val Pro Glu Arg Leu Ala Ala Gly Thr Leu 1565 1570 1575Val Val Val Val Leu Met Pro Pro Glu Gln Leu Arg Asn Ser Ser 1580 1585 1590Phe His Phe Leu Arg Glu Leu Ser Arg Val Leu His Thr Asn Val 1595 1600 1605Val Phe Lys Arg Asp Ala His Gly Gln Gln Met Ile Phe Pro Tyr 1610 1615 1620Tyr Gly Arg Glu Glu Glu Leu Arg Lys His Pro Ile Lys Arg Ala 1625 1630 1635Ala Glu Gly Trp Ala Ala Pro Asp Ala Leu Leu Gly Gln Val Lys 1640 1645 1650Ala Ser Leu Leu Pro Gly Gly Ser Glu Gly Gly Arg Arg Arg Arg 1655 1660 1665Glu Leu Asp Pro Met Asp Val Arg Gly Ser Ile Val Tyr Leu Glu 1670 1675 1680Ile Asp Asn Arg Gln Cys Val Gln Ala Ser Ser Gln Cys Phe Gln 1685 1690 1695Ser Ala Thr Asp Val Ala Ala Phe Leu Gly Ala Leu Ala Ser Leu 1700 1705 1710Gly Ser Leu Asn Ile Pro Tyr Lys Ile Glu Ala Val Gln Ser Glu 1715 1720 1725Thr Val Glu Pro Pro Pro Pro Ala Gln Leu His Phe Met Tyr Val 1730 1735 1740Ala Ala Ala Ala Phe Val Leu Leu Phe Phe Val Gly Cys Gly Val 1745 1750 1755Leu Leu Ser Arg Lys Arg Arg Arg Gln His Gly Gln Leu Trp Phe 1760 1765 1770Pro Glu Gly Phe Lys Val Ser Glu Ala Ser Lys Lys Lys Arg Arg 1775 1780 1785Glu Pro Leu Gly Glu Asp Ser Val Gly Leu Lys Pro Leu Lys Asn 1790 1795 1800Ala Ser Asp Gly Ala Leu Met Asp Asp Asn Gln Asn Glu Trp Gly 1805 1810 1815Asp Glu Asp Leu Glu Thr Lys Lys Phe Arg Phe Glu Glu Pro Val 1820 1825 1830Val Leu Pro Asp Leu Asp Asp Gln Thr Asp His Arg Gln Trp Thr 1835 1840 1845Gln Gln His Leu Asp Ala Ala Asp Leu Arg Met Ser Ala Met Ala 1850 1855 1860Pro Thr Pro Pro Gln Gly Glu Val Asp Ala Asp Cys Met Asp Val 1865 1870 1875Asn Val Arg Gly Pro Asp Gly Phe Thr Pro Leu Met Ile Ala Ser 1880 1885 1890Cys Ser Gly Gly Gly Leu Glu Thr Gly Asn Ser Glu Glu Glu Glu 1895 1900 1905Asp Ala Pro Ala Val Ile Ser Asp Phe Ile Tyr Gln Gly Ala Ser 1910 1915 1920Leu His Asn Gln Thr Asp Arg Thr Gly Glu Thr Ala Leu His Leu 1925 1930 1935Ala Ala Arg Tyr Ser Arg Ser Asp Ala Ala Lys Arg Leu Leu Glu 1940 1945 1950Ala Ser Ala Asp Ala Asn Ile Gln Asp Asn Met Gly Arg Thr Pro 1955 1960 1965Leu His Ala Ala Val Ser Ala Asp Ala Gln Gly Val Phe Gln Ile 1970 1975 1980Leu Ile Arg Asn Arg Ala Thr Asp Leu Asp Ala Arg Met His Asp 1985 1990 1995Gly Thr Thr Pro Leu Ile Leu Ala Ala Arg Leu Ala Val Glu Gly 2000 2005 2010Met Leu Glu Asp Leu Ile Asn Ser His Ala Asp Val Asn Ala Val 2015 2020 2025Asp Asp Leu Gly Lys Ser Ala Leu His Trp Ala Ala Ala Val Asn 2030 2035 2040Asn Val Asp Ala Ala Val Val Leu Leu Lys Asn Gly Ala Asn Lys 2045 2050 2055Asp Met Gln Asn Asn Arg Glu Glu Thr Pro Leu Phe Leu Ala Ala 2060 2065 2070Arg Glu Gly Ser Tyr Glu Thr Ala Lys Val Leu Leu Asp His Phe 2075 2080 2085Ala Asn Arg Asp Ile Thr Asp His Met Asp Arg Leu Pro Arg Asp 2090 2095 2100Ile Ala Gln Glu Arg Met His His Asp Ile Val Arg Leu Leu Asp 2105

2110 2115Glu Tyr Asn Leu Val Arg Ser Pro Gln Leu His Gly Ala Pro Leu 2120 2125 2130Gly Gly Thr Pro Thr Leu Ser Pro Pro Leu Cys Ser Pro Asn Gly 2135 2140 2145Tyr Leu Gly Ser Leu Lys Pro Gly Val Gln Gly Lys Lys Val Arg 2150 2155 2160Lys Pro Ser Ser Lys Gly Leu Ala Cys Gly Ser Lys Glu Ala Lys 2165 2170 2175Asp Leu Lys Ala Arg Arg Lys Lys Ser Gln Asp Gly Lys Gly Cys 2180 2185 2190Leu Leu Asp Ser Ser Gly Met Leu Ser Pro Val Asp Ser Leu Glu 2195 2200 2205Ser Pro His Gly Tyr Leu Ser Asp Val Ala Ser Pro Pro Leu Leu 2210 2215 2220Pro Ser Pro Phe Gln Gln Ser Pro Ser Val Pro Leu Asn His Leu 2225 2230 2235Pro Gly Met Pro Asp Thr His Leu Gly Ile Gly His Leu Asn Val 2240 2245 2250Ala Ala Lys Pro Glu Met Ala Ala Leu Gly Gly Gly Gly Arg Leu 2255 2260 2265Ala Phe Glu Thr Gly Pro Pro Arg Leu Ser His Leu Pro Val Ala 2270 2275 2280Ser Gly Thr Ser Thr Val Leu Gly Ser Ser Ser Gly Gly Ala Leu 2285 2290 2295Asn Phe Thr Val Gly Gly Ser Thr Ser Leu Asn Gly Gln Cys Glu 2300 2305 2310Trp Leu Ser Arg Leu Gln Ser Gly Met Val Pro Asn Gln Tyr Asn 2315 2320 2325Pro Leu Arg Gly Ser Val Ala Pro Gly Pro Leu Ser Thr Gln Ala 2330 2335 2340Pro Ser Leu Gln His Gly Met Val Gly Pro Leu His Ser Ser Leu 2345 2350 2355Ala Ala Ser Ala Leu Ser Gln Met Met Ser Tyr Gln Gly Leu Pro 2360 2365 2370Ser Thr Arg Leu Ala Thr Gln Pro His Leu Val Gln Thr Gln Gln 2375 2380 2385Val Gln Pro Gln Asn Leu Gln Met Gln Gln Gln Asn Leu Gln Pro 2390 2395 2400Ala Asn Ile Gln Gln Gln Gln Ser Leu Gln Pro Pro Pro Pro Pro 2405 2410 2415Pro Gln Pro His Leu Gly Val Ser Ser Ala Ala Ser Gly His Leu 2420 2425 2430Gly Arg Ser Phe Leu Ser Gly Glu Pro Ser Gln Ala Asp Val Gln 2435 2440 2445Pro Leu Gly Pro Ser Ser Leu Ala Val His Thr Ile Leu Pro Gln 2450 2455 2460Glu Ser Pro Ala Leu Pro Thr Ser Leu Pro Ser Ser Leu Val Pro 2465 2470 2475Pro Val Thr Ala Ala Gln Phe Leu Thr Pro Pro Ser Gln His Ser 2480 2485 2490Tyr Ser Ser Pro Val Asp Asn Thr Pro Ser His Gln Leu Gln Val 2495 2500 2505Pro Glu His Pro Phe Leu Thr Pro Ser Pro Glu Ser Pro Asp Gln 2510 2515 2520Trp Ser Ser Ser Ser Pro His Ser Asn Val Ser Asp Trp Ser Glu 2525 2530 2535Gly Val Ser Ser Pro Pro Thr Ser Met Gln Ser Gln Ile Ala Arg 2540 2545 2550Ile Pro Glu Ala Phe Lys 2555 452471PRTHomo sapienshuman Notch2 45Met Pro Ala Leu Arg Pro Ala Leu Leu Trp Ala Leu Leu Ala Leu1 5 10 15Trp Leu Cys Cys Ala Ala Pro Ala His Ala Leu Gln Cys Arg Asp 20 25 30Gly Tyr Glu Pro Cys Val Asn Glu Gly Met Cys Val Thr Tyr His 35 40 45Asn Gly Thr Gly Tyr Cys Lys Cys Pro Glu Gly Phe Leu Gly Glu 50 55 60Tyr Cys Gln His Arg Asp Pro Cys Glu Lys Asn Arg Cys Gln Asn 65 70 75Gly Gly Thr Cys Val Ala Gln Ala Met Leu Gly Lys Ala Thr Cys 80 85 90Arg Cys Ala Ser Gly Phe Thr Gly Glu Asp Cys Gln Tyr Ser Thr 95 100 105Ser His Pro Cys Phe Val Ser Arg Pro Cys Leu Asn Gly Gly Thr 110 115 120Cys His Met Leu Ser Arg Asp Thr Tyr Glu Cys Thr Cys Gln Val 125 130 135Gly Phe Thr Gly Lys Glu Cys Gln Trp Thr Asp Ala Cys Leu Ser 140 145 150His Pro Cys Ala Asn Gly Ser Thr Cys Thr Thr Val Ala Asn Gln 155 160 165Phe Ser Cys Lys Cys Leu Thr Gly Phe Thr Gly Gln Lys Cys Glu 170 175 180Thr Asp Val Asn Glu Cys Asp Ile Pro Gly His Cys Gln His Gly 185 190 195Gly Thr Cys Leu Asn Leu Pro Gly Ser Tyr Gln Cys Gln Cys Pro 200 205 210Gln Gly Phe Thr Gly Gln Tyr Cys Asp Ser Leu Tyr Val Pro Cys 215 220 225Ala Pro Ser Pro Cys Val Asn Gly Gly Thr Cys Arg Gln Thr Gly 230 235 240Asp Phe Thr Phe Glu Cys Asn Cys Leu Pro Gly Phe Glu Gly Ser 245 250 255Thr Cys Glu Arg Asn Ile Asp Asp Cys Pro Asn His Arg Cys Gln 260 265 270Asn Gly Gly Val Cys Val Asp Gly Val Asn Thr Tyr Asn Cys Arg 275 280 285Cys Pro Pro Gln Trp Thr Gly Gln Phe Cys Thr Glu Asp Val Asp 290 295 300Glu Cys Leu Leu Gln Pro Asn Ala Cys Gln Asn Gly Gly Thr Cys 305 310 315Ala Asn Arg Asn Gly Gly Tyr Gly Cys Val Cys Val Asn Gly Trp 320 325 330Ser Gly Asp Asp Cys Ser Glu Asn Ile Asp Asp Cys Ala Phe Ala 335 340 345Ser Cys Thr Pro Gly Ser Thr Cys Ile Asp Arg Val Ala Ser Phe 350 355 360Ser Cys Met Cys Pro Glu Gly Lys Ala Gly Leu Leu Cys His Leu 365 370 375Asp Asp Ala Cys Ile Ser Asn Pro Cys His Lys Gly Ala Leu Cys 380 385 390Asp Thr Asn Pro Leu Asn Gly Gln Tyr Ile Cys Thr Cys Pro Gln 395 400 405Gly Tyr Lys Gly Ala Asp Cys Thr Glu Asp Val Asp Glu Cys Ala 410 415 420Met Ala Asn Ser Asn Pro Cys Glu His Ala Gly Lys Cys Val Asn 425 430 435Thr Asp Gly Ala Phe His Cys Glu Cys Leu Lys Gly Tyr Ala Gly 440 445 450Pro Arg Cys Glu Met Asp Ile Asn Glu Cys His Ser Asp Pro Cys 455 460 465Gln Asn Asp Ala Thr Cys Leu Asp Lys Ile Gly Gly Phe Thr Cys 470 475 480Leu Cys Met Pro Gly Phe Lys Gly Val His Cys Glu Leu Glu Ile 485 490 495Asn Glu Cys Gln Ser Asn Pro Cys Val Asn Asn Gly Gln Cys Val 500 505 510Asp Lys Val Asn Arg Phe Gln Cys Leu Cys Pro Pro Gly Phe Thr 515 520 525Gly Pro Val Cys Gln Ile Asp Ile Asp Asp Cys Ser Ser Thr Pro 530 535 540Cys Leu Asn Gly Ala Lys Cys Ile Asp His Pro Asn Gly Tyr Glu 545 550 555Cys Gln Cys Ala Thr Gly Phe Thr Gly Val Leu Cys Glu Glu Asn 560 565 570Ile Asp Asn Cys Asp Pro Asp Pro Cys His His Gly Gln Cys Gln 575 580 585Asp Gly Ile Asp Ser Tyr Thr Cys Ile Cys Asn Pro Gly Tyr Met 590 595 600Gly Ala Ile Cys Ser Asp Gln Ile Asp Glu Cys Tyr Ser Ser Pro 605 610 615Cys Leu Asn Asp Gly Arg Cys Ile Asp Leu Val Asn Gly Tyr Gln 620 625 630Cys Asn Cys Gln Pro Gly Thr Ser Gly Val Asn Cys Glu Ile Asn 635 640 645Phe Asp Asp Cys Ala Ser Asn Pro Cys Ile His Gly Ile Cys Met 650 655 660Asp Gly Ile Asn Arg Tyr Ser Cys Val Cys Ser Pro Gly Phe Thr 665 670 675Gly Gln Arg Cys Asn Ile Asp Ile Asp Glu Cys Ala Ser Asn Pro 680 685 690Cys Arg Lys Gly Ala Thr Cys Ile Asn Gly Val Asn Gly Phe Arg 695 700 705Cys Ile Cys Pro Glu Gly Pro His His Pro Ser Cys Tyr Ser Gln 710 715 720Val Asn Glu Cys Leu Ser Asn Pro Cys Ile His Gly Asn Cys Thr 725 730 735Gly Gly Leu Ser Gly Tyr Lys Cys Leu Cys Asp Ala Gly Trp Val 740 745 750Gly Ile Asn Cys Glu Val Asp Lys Asn Glu Cys Leu Ser Asn Pro 755 760 765Cys Gln Asn Gly Gly Thr Cys Asp Asn Leu Val Asn Gly Tyr Arg 770 775 780Cys Thr Cys Lys Lys Gly Phe Lys Gly Tyr Asn Cys Gln Val Asn 785 790 795Ile Asp Glu Cys Ala Ser Asn Pro Cys Leu Asn Gln Gly Thr Cys 800 805 810Phe Asp Asp Ile Ser Gly Tyr Thr Cys His Cys Val Leu Pro Tyr 815 820 825Thr Gly Lys Asn Cys Gln Thr Val Leu Ala Pro Cys Ser Pro Asn 830 835 840Pro Cys Glu Asn Ala Ala Val Cys Lys Glu Ser Pro Asn Phe Glu 845 850 855Ser Tyr Thr Cys Leu Cys Ala Pro Gly Trp Gln Gly Gln Arg Cys 860 865 870Thr Ile Asp Ile Asp Glu Cys Ile Ser Lys Pro Cys Met Asn His 875 880 885Gly Leu Cys His Asn Thr Gln Gly Ser Tyr Met Cys Glu Cys Pro 890 895 900Pro Gly Phe Ser Gly Met Asp Cys Glu Glu Asp Ile Asp Asp Cys 905 910 915Leu Ala Asn Pro Cys Gln Asn Gly Gly Ser Cys Met Asp Gly Val 920 925 930Asn Thr Phe Ser Cys Leu Cys Leu Pro Gly Phe Thr Gly Asp Lys 935 940 945Cys Gln Thr Asp Met Asn Glu Cys Leu Ser Glu Pro Cys Lys Asn 950 955 960Gly Gly Thr Cys Ser Asp Tyr Val Asn Ser Tyr Thr Cys Lys Cys 965 970 975Gln Ala Gly Phe Asp Gly Val His Cys Glu Asn Asn Ile Asn Glu 980 985 990Cys Thr Glu Ser Ser Cys Phe Asn Gly Gly Thr Cys Val Asp Gly 995 1000 1005Ile Asn Ser Phe Ser Cys Leu Cys Pro Val Gly Phe Thr Gly Ser 1010 1015 1020Phe Cys Leu His Glu Ile Asn Glu Cys Ser Ser His Pro Cys Leu 1025 1030 1035Asn Glu Gly Thr Cys Val Asp Gly Leu Gly Thr Tyr Arg Cys Ser 1040 1045 1050Cys Pro Leu Gly Tyr Thr Gly Lys Asn Cys Gln Thr Leu Val Asn 1055 1060 1065Leu Cys Ser Arg Ser Pro Cys Lys Asn Lys Gly Thr Cys Val Gln 1070 1075 1080Lys Lys Ala Glu Ser Gln Cys Leu Cys Pro Ser Gly Trp Ala Gly 1085 1090 1095Ala Tyr Cys Asp Val Pro Asn Val Ser Cys Asp Ile Ala Ala Ser 1100 1105 1110Arg Arg Gly Val Leu Val Glu His Leu Cys Gln His Ser Gly Val 1115 1120 1125Cys Ile Asn Ala Gly Asn Thr His Tyr Cys Gln Cys Pro Leu Gly 1130 1135 1140Tyr Thr Gly Ser Tyr Cys Glu Glu Gln Leu Asp Glu Cys Ala Ser 1145 1150 1155Asn Pro Cys Gln His Gly Ala Thr Cys Ser Asp Phe Ile Gly Gly 1160 1165 1170Tyr Arg Cys Glu Cys Val Pro Gly Tyr Gln Gly Val Asn Cys Glu 1175 1180 1185Tyr Glu Val Asp Glu Cys Gln Asn Gln Pro Cys Gln Asn Gly Gly 1190 1195 1200Thr Cys Ile Asp Leu Val Asn His Phe Lys Cys Ser Cys Pro Pro 1205 1210 1215Gly Thr Arg Gly Leu Leu Cys Glu Glu Asn Ile Asp Asp Cys Ala 1220 1225 1230Arg Gly Pro His Cys Leu Asn Gly Gly Gln Cys Met Asp Arg Ile 1235 1240 1245Gly Gly Tyr Ser Cys Arg Cys Leu Pro Gly Phe Ala Gly Glu Arg 1250 1255 1260Cys Glu Gly Asp Ile Asn Glu Cys Leu Ser Asn Pro Cys Ser Ser 1265 1270 1275Glu Gly Ser Leu Asp Cys Ile Gln Leu Thr Asn Asp Tyr Leu Cys 1280 1285 1290Val Cys Arg Ser Ala Phe Thr Gly Arg His Cys Glu Thr Phe Val 1295 1300 1305Asp Val Cys Pro Gln Met Pro Cys Leu Asn Gly Gly Thr Cys Ala 1310 1315 1320Val Ala Ser Asn Met Pro Asp Gly Phe Ile Cys Arg Cys Pro Pro 1325 1330 1335Gly Phe Ser Gly Ala Arg Cys Gln Ser Ser Cys Gly Gln Val Lys 1340 1345 1350Cys Arg Lys Gly Glu Gln Cys Val His Thr Ala Ser Gly Pro Arg 1355 1360 1365Cys Phe Cys Pro Ser Pro Arg Asp Cys Glu Ser Gly Cys Ala Ser 1370 1375 1380Ser Pro Cys Gln His Gly Gly Ser Cys His Pro Gln Arg Gln Pro 1385 1390 1395Pro Tyr Tyr Ser Cys Gln Cys Ala Pro Pro Phe Ser Gly Ser Arg 1400 1405 1410Cys Glu Leu Tyr Thr Ala Pro Pro Ser Thr Pro Pro Ala Thr Cys 1415 1420 1425Leu Ser Gln Tyr Cys Ala Asp Lys Ala Arg Asp Gly Val Cys Asp 1430 1435 1440Glu Ala Cys Asn Ser His Ala Cys Gln Trp Asp Gly Gly Asp Cys 1445 1450 1455Ser Leu Thr Met Glu Asn Pro Trp Ala Asn Cys Ser Ser Pro Leu 1460 1465 1470Pro Cys Trp Asp Tyr Ile Asn Asn Gln Cys Asp Glu Leu Cys Asn 1475 1480 1485Thr Val Glu Cys Leu Phe Asp Asn Phe Glu Cys Gln Gly Asn Ser 1490 1495 1500Lys Thr Cys Lys Tyr Asp Lys Tyr Cys Ala Asp His Phe Lys Asp 1505 1510 1515Asn His Cys Asp Gln Gly Cys Asn Ser Glu Glu Cys Gly Trp Asp 1520 1525 1530Gly Leu Asp Cys Ala Ala Asp Gln Pro Glu Asn Leu Ala Glu Gly 1535 1540 1545Thr Leu Val Ile Val Val Leu Met Pro Pro Glu Gln Leu Leu Gln 1550 1555 1560Asp Ala Arg Ser Phe Leu Arg Ala Leu Gly Thr Leu Leu His Thr 1565 1570 1575Asn Leu Arg Ile Lys Arg Asp Ser Gln Gly Glu Leu Met Val Tyr 1580 1585 1590Pro Tyr Tyr Gly Glu Lys Ser Ala Ala Met Lys Lys Gln Arg Met 1595 1600 1605Thr Arg Arg Ser Leu Pro Gly Glu Gln Glu Gln Glu Val Ala Gly 1610 1615 1620Ser Lys Val Phe Leu Glu Ile Asp Asn Arg Gln Cys Val Gln Asp 1625 1630 1635Ser Asp His Cys Phe Lys Asn Thr Asp Ala Ala Ala Ala Leu Leu 1640 1645 1650Ala Ser His Ala Ile Gln Gly Thr Leu Ser Tyr Pro Leu Val Ser 1655 1660 1665Val Val Ser Glu Ser Leu Thr Pro Glu Arg Thr Gln Leu Leu Tyr 1670 1675 1680Leu Leu Ala Val Ala Val Val Ile Ile Leu Phe Ile Ile Leu Leu 1685 1690 1695Gly Val Ile Met Ala Lys Arg Lys Arg Lys His Gly Ser Leu Trp 1700 1705 1710Leu Pro Glu Gly Phe Thr Leu Arg Arg Asp Ala Ser Asn His Lys 1715 1720 1725Arg Arg Glu Pro Val Gly Gln Asp Ala Val Gly Leu Lys Asn Leu 1730 1735 1740Ser Val Gln Val Ser Glu Ala Asn Leu Ile Gly Thr Gly Thr Ser 1745 1750 1755Glu His Trp Val Asp Asp Glu Gly Pro Gln Pro Lys Lys Val Lys 1760 1765 1770Ala Glu Asp Glu Ala Leu Leu Ser Glu Glu Asp Asp Pro Ile Asp 1775 1780 1785Arg Arg Pro Trp Thr Gln Gln His Leu Glu Ala Ala Asp Ile Arg 1790 1795 1800Arg Thr Pro Ser Leu Ala Leu Thr Pro Pro Gln Ala Glu Gln Glu 1805 1810 1815Val Asp Val Leu Asp Val Asn Val Arg Gly Pro Asp Gly Cys Thr 1820 1825 1830Pro Leu Met Leu Ala Ser Leu Arg Gly Gly Ser Ser Asp Leu Ser

1835 1840 1845Asp Glu Asp Glu Asp Ala Glu Asp Ser Ser Ala Asn Ile Ile Thr 1850 1855 1860Asp Leu Val Tyr Gln Gly Ala Ser Leu Gln Ala Gln Thr Asp Arg 1865 1870 1875Thr Gly Glu Met Ala Leu His Leu Ala Ala Arg Tyr Ser Arg Ala 1880 1885 1890Asp Ala Ala Lys Arg Leu Leu Asp Ala Gly Ala Asp Ala Asn Ala 1895 1900 1905Gln Asp Asn Met Gly Arg Cys Pro Leu His Ala Ala Val Ala Ala 1910 1915 1920Asp Ala Gln Gly Val Phe Gln Ile Leu Ile Arg Asn Arg Val Thr 1925 1930 1935Asp Leu Asp Ala Arg Met Asn Asp Gly Thr Thr Pro Leu Ile Leu 1940 1945 1950Ala Ala Arg Leu Ala Val Glu Gly Met Val Ala Glu Leu Ile Asn 1955 1960 1965Cys Gln Ala Asp Val Asn Ala Val Asp Asp His Gly Lys Ser Ala 1970 1975 1980Leu His Trp Ala Ala Ala Val Asn Asn Val Glu Ala Thr Leu Leu 1985 1990 1995Leu Leu Lys Asn Gly Ala Asn Arg Asp Met Gln Asp Asn Lys Glu 2000 2005 2010Glu Thr Pro Leu Phe Leu Ala Ala Arg Glu Gly Ser Tyr Glu Ala 2015 2020 2025Ala Lys Ile Leu Leu Asp His Phe Ala Asn Arg Asp Ile Thr Asp 2030 2035 2040His Met Asp Arg Leu Pro Arg Asp Val Ala Arg Asp Arg Met His 2045 2050 2055His Asp Ile Val Arg Leu Leu Asp Glu Tyr Asn Val Thr Pro Ser 2060 2065 2070Pro Pro Gly Thr Val Leu Thr Ser Ala Leu Ser Pro Val Ile Cys 2075 2080 2085Gly Pro Asn Arg Ser Phe Leu Ser Leu Lys His Thr Pro Met Gly 2090 2095 2100Lys Lys Ser Arg Arg Pro Ser Ala Lys Ser Thr Met Pro Thr Ser 2105 2110 2115Leu Pro Asn Leu Ala Lys Glu Ala Lys Asp Ala Lys Gly Ser Arg 2120 2125 2130Arg Lys Lys Ser Leu Ser Glu Lys Val Gln Leu Ser Glu Ser Ser 2135 2140 2145Val Thr Leu Ser Pro Val Asp Ser Leu Glu Ser Pro His Thr Tyr 2150 2155 2160Val Ser Asp Thr Thr Ser Ser Pro Met Ile Thr Ser Pro Gly Ile 2165 2170 2175Leu Gln Ala Ser Pro Asn Pro Met Leu Ala Thr Ala Ala Pro Pro 2180 2185 2190Ala Pro Val His Ala Gln His Ala Leu Ser Phe Ser Asn Leu His 2195 2200 2205Glu Met Gln Pro Leu Ala His Gly Ala Ser Thr Val Leu Pro Ser 2210 2215 2220Val Ser Gln Leu Leu Ser His His His Ile Val Ser Pro Gly Ser 2225 2230 2235Gly Ser Ala Gly Ser Leu Ser Arg Leu His Pro Val Pro Val Pro 2240 2245 2250Ala Asp Trp Met Asn Arg Met Glu Val Asn Glu Thr Gln Tyr Asn 2255 2260 2265Glu Met Phe Gly Met Val Leu Ala Pro Ala Glu Gly Thr His Pro 2270 2275 2280Gly Ile Ala Pro Gln Ser Arg Pro Pro Glu Gly Lys His Ile Thr 2285 2290 2295Thr Pro Arg Glu Pro Leu Pro Pro Ile Val Thr Phe Gln Leu Ile 2300 2305 2310Pro Lys Gly Ser Ile Ala Gln Pro Ala Gly Ala Pro Gln Pro Gln 2315 2320 2325Ser Thr Cys Pro Pro Ala Val Ala Gly Pro Leu Pro Thr Met Tyr 2330 2335 2340Gln Ile Pro Glu Met Ala Arg Leu Pro Ser Val Ala Phe Pro Thr 2345 2350 2355Ala Met Met Pro Gln Gln Asp Gly Gln Val Ala Gln Thr Ile Leu 2360 2365 2370Pro Ala Tyr His Pro Phe Pro Ala Ser Val Gly Lys Tyr Pro Thr 2375 2380 2385Pro Pro Ser Gln His Ser Tyr Ala Ser Ser Asn Ala Ala Glu Arg 2390 2395 2400Thr Pro Ser His Ser Gly His Leu Gln Gly Glu His Pro Tyr Leu 2405 2410 2415Thr Pro Ser Pro Glu Ser Pro Asp Gln Trp Ser Ser Ser Ser Pro 2420 2425 2430His Ser Ala Ser Asp Trp Ser Asp Val Thr Thr Ser Pro Thr Pro 2435 2440 2445Gly Gly Ala Gly Gly Gly Gln Arg Gly Pro Gly Thr His Met Ser 2450 2455 2460Glu Pro Pro His Asn Asn Met Gln Val Tyr Ala 2465 2470 462002PRTHomo sapienshuman Notch4 46Met Gln Pro Pro Ser Leu Leu Leu Leu Leu Leu Leu Leu Leu Leu1 5 10 15Cys Val Ser Val Val Arg Pro Arg Gly Leu Leu Cys Gly Ser Phe 20 25 30Pro Glu Pro Cys Ala Asn Gly Gly Thr Cys Leu Ser Leu Ser Leu 35 40 45Gly Gln Gly Thr Cys Gln Cys Ala Pro Gly Phe Leu Gly Glu Thr 50 55 60Cys Gln Phe Pro Asp Pro Cys Gln Asn Ala Gln Leu Cys Gln Asn 65 70 75Gly Gly Ser Cys Gln Ala Leu Leu Pro Ala Pro Leu Gly Leu Pro 80 85 90Ser Ser Pro Ser Pro Leu Thr Pro Ser Phe Leu Cys Thr Cys Leu 95 100 105Pro Gly Phe Thr Gly Glu Arg Cys Gln Ala Lys Leu Glu Asp Pro 110 115 120Cys Pro Pro Ser Phe Cys Ser Lys Arg Gly Arg Cys His Ile Gln 125 130 135Ala Ser Gly Arg Pro Gln Cys Ser Cys Met Pro Gly Trp Thr Gly 140 145 150Glu Gln Cys Gln Leu Arg Asp Phe Cys Ser Ala Asn Pro Cys Val 155 160 165Asn Gly Gly Val Cys Leu Ala Thr Tyr Pro Gln Ile Gln Cys His 170 175 180Cys Pro Pro Gly Phe Glu Gly His Ala Cys Glu Arg Asp Val Asn 185 190 195Glu Cys Phe Gln Asp Pro Gly Pro Cys Pro Lys Gly Thr Ser Cys 200 205 210His Asn Thr Leu Gly Ser Phe Gln Cys Leu Cys Pro Val Gly Gln 215 220 225Glu Gly Pro Arg Cys Glu Leu Arg Ala Gly Pro Cys Pro Pro Arg 230 235 240Gly Cys Ser Asn Gly Gly Thr Cys Gln Leu Met Pro Glu Lys Asp 245 250 255Ser Thr Phe His Leu Cys Leu Cys Pro Pro Gly Phe Ile Gly Pro 260 265 270Gly Cys Glu Val Asn Pro Asp Asn Cys Val Ser His Gln Cys Gln 275 280 285Asn Gly Gly Thr Cys Gln Asp Gly Leu Asp Thr Tyr Thr Cys Leu 290 295 300Cys Pro Glu Thr Trp Thr Gly Trp Asp Cys Ser Glu Asp Val Asp 305 310 315Glu Cys Glu Ala Gln Gly Pro Pro His Cys Arg Asn Gly Gly Thr 320 325 330Cys Gln Asn Ser Ala Gly Ser Phe His Cys Val Cys Val Ser Gly 335 340 345Trp Gly Gly Thr Ser Cys Glu Glu Asn Leu Asp Asp Cys Ile Ala 350 355 360Ala Thr Cys Ala Pro Gly Ser Thr Cys Ile Asp Arg Val Gly Ser 365 370 375Phe Ser Cys Leu Cys Pro Pro Gly Arg Thr Gly Leu Leu Cys His 380 385 390Leu Glu Asp Met Cys Leu Ser Gln Pro Cys His Gly Asp Ala Gln 395 400 405Cys Ser Thr Asn Pro Leu Thr Gly Ser Thr Leu Cys Leu Cys Gln 410 415 420Pro Gly Tyr Ser Gly Pro Thr Cys His Gln Asp Leu Asp Glu Cys 425 430 435Leu Met Ala Gln Gln Gly Pro Ser Pro Cys Glu His Gly Gly Ser 440 445 450Cys Leu Asn Thr Pro Gly Ser Phe Asn Cys Leu Cys Pro Pro Gly 455 460 465Tyr Thr Gly Ser Arg Cys Glu Ala Asp His Asn Glu Cys Leu Ser 470 475 480Gln Pro Cys His Pro Gly Ser Thr Cys Leu Asp Leu Leu Ala Thr 485 490 495Phe His Cys Leu Cys Pro Pro Gly Leu Glu Gly Gln Leu Cys Glu 500 505 510Val Glu Thr Asn Glu Cys Ala Ser Ala Pro Cys Leu Asn His Ala 515 520 525Asp Cys His Asp Leu Leu Asn Gly Phe Gln Cys Ile Cys Leu Pro 530 535 540Gly Phe Ser Gly Thr Arg Cys Glu Glu Asp Ile Asp Glu Cys Arg 545 550 555Ser Ser Pro Cys Ala Asn Gly Gly Gln Cys Gln Asp Gln Pro Gly 560 565 570Ala Phe His Cys Lys Cys Leu Pro Gly Phe Glu Gly Pro Arg Cys 575 580 585Gln Thr Glu Val Asp Glu Cys Leu Ser Asp Pro Cys Pro Val Gly 590 595 600Ala Ser Cys Leu Asp Leu Pro Gly Ala Phe Phe Cys Leu Cys Pro 605 610 615Ser Gly Phe Thr Gly Gln Leu Cys Glu Val Pro Leu Cys Ala Pro 620 625 630Asn Leu Cys Gln Pro Lys Gln Ile Cys Lys Asp Gln Lys Asp Lys 635 640 645Ala Asn Cys Leu Cys Pro Asp Gly Ser Pro Gly Cys Ala Pro Pro 650 655 660Glu Asp Asn Cys Thr Cys His His Gly His Cys Gln Arg Ser Ser 665 670 675Cys Val Cys Asp Val Gly Trp Thr Gly Pro Glu Cys Glu Ala Glu 680 685 690Leu Gly Gly Cys Ile Ser Ala Pro Cys Ala His Gly Gly Thr Cys 695 700 705Tyr Pro Gln Pro Ser Gly Tyr Asn Cys Thr Cys Pro Thr Gly Tyr 710 715 720Thr Gly Pro Thr Cys Ser Glu Glu Met Thr Ala Cys His Ser Gly 725 730 735Pro Cys Leu Asn Gly Gly Ser Cys Asn Pro Ser Pro Gly Gly Tyr 740 745 750Tyr Cys Thr Cys Pro Pro Ser His Thr Gly Pro Gln Cys Gln Thr 755 760 765Ser Thr Asp Tyr Cys Val Ser Ala Pro Cys Phe Asn Gly Gly Thr 770 775 780Cys Val Asn Arg Pro Gly Thr Phe Ser Cys Leu Cys Ala Met Gly 785 790 795Phe Gln Gly Pro Arg Cys Glu Gly Lys Leu Arg Pro Ser Cys Ala 800 805 810Asp Ser Pro Cys Arg Asn Arg Ala Thr Cys Gln Asp Ser Pro Gln 815 820 825Gly Pro Arg Cys Leu Cys Pro Thr Gly Tyr Thr Gly Gly Ser Cys 830 835 840Gln Thr Leu Met Asp Leu Cys Ala Gln Lys Pro Cys Pro Arg Asn 845 850 855Ser His Cys Leu Gln Thr Gly Pro Ser Phe His Cys Leu Cys Leu 860 865 870Gln Gly Trp Thr Gly Pro Leu Cys Asn Leu Pro Leu Ser Ser Cys 875 880 885Gln Lys Ala Ala Leu Ser Gln Gly Ile Asp Val Ser Ser Leu Cys 890 895 900His Asn Gly Gly Leu Cys Val Asp Ser Gly Pro Ser Tyr Phe Cys 905 910 915His Cys Pro Pro Gly Phe Gln Gly Ser Leu Cys Gln Asp His Val 920 925 930Asn Pro Cys Glu Ser Arg Pro Cys Gln Asn Gly Ala Thr Cys Met 935 940 945Ala Gln Pro Ser Gly Tyr Leu Cys Gln Cys Ala Pro Gly Tyr Asp 950 955 960Gly Gln Asn Cys Ser Lys Glu Leu Asp Ala Cys Gln Ser Gln Pro 965 970 975Cys His Asn His Gly Thr Cys Thr Pro Lys Pro Gly Gly Phe His 980 985 990Cys Ala Cys Pro Pro Gly Phe Val Gly Leu Arg Cys Glu Gly Asp 995 1000 1005Val Asp Glu Cys Leu Asp Gln Pro Cys His Pro Thr Gly Thr Ala 1010 1015 1020Ala Cys His Ser Leu Ala Asn Ala Phe Tyr Cys Gln Cys Leu Pro 1025 1030 1035Gly His Thr Gly Gln Trp Cys Glu Val Glu Ile Asp Pro Cys His 1040 1045 1050Ser Gln Pro Cys Phe His Gly Gly Thr Cys Glu Ala Thr Ala Gly 1055 1060 1065Ser Pro Leu Gly Phe Ile Cys His Cys Pro Lys Gly Phe Glu Gly 1070 1075 1080Pro Thr Cys Ser His Arg Ala Pro Ser Cys Gly Phe His His Cys 1085 1090 1095His His Gly Gly Leu Cys Leu Pro Ser Pro Lys Pro Gly Phe Pro 1100 1105 1110Pro Arg Cys Ala Cys Leu Ser Gly Tyr Gly Gly Pro Asp Cys Leu 1115 1120 1125Thr Pro Pro Ala Pro Lys Gly Cys Gly Pro Pro Ser Pro Cys Leu 1130 1135 1140Tyr Asn Gly Ser Cys Ser Glu Thr Thr Gly Leu Gly Gly Pro Gly 1145 1150 1155Phe Arg Cys Ser Cys Pro His Ser Ser Pro Gly Pro Arg Cys Gln 1160 1165 1170Lys Pro Gly Ala Lys Gly Cys Glu Gly Arg Ser Gly Asp Gly Ala 1175 1180 1185Cys Asp Ala Gly Cys Ser Gly Pro Gly Gly Asn Trp Asp Gly Gly 1190 1195 1200Asp Cys Ser Leu Gly Val Pro Asp Pro Trp Lys Gly Cys Pro Ser 1205 1210 1215His Ser Arg Cys Trp Leu Leu Phe Arg Asp Gly Gln Cys His Pro 1220 1225 1230Gln Cys Asp Ser Glu Glu Cys Leu Phe Asp Gly Tyr Asp Cys Glu 1235 1240 1245Thr Pro Pro Ala Cys Thr Pro Ala Tyr Asp Gln Tyr Cys His Asp 1250 1255 1260His Phe His Asn Gly His Cys Glu Lys Gly Cys Asn Thr Ala Glu 1265 1270 1275Cys Gly Trp Asp Gly Gly Asp Cys Arg Pro Glu Asp Gly Asp Pro 1280 1285 1290Glu Trp Gly Pro Ser Leu Ala Leu Leu Val Val Leu Ser Pro Pro 1295 1300 1305Ala Leu Asp Gln Gln Leu Phe Ala Leu Ala Arg Val Leu Ser Leu 1310 1315 1320Thr Leu Arg Val Gly Leu Trp Val Arg Lys Asp Arg Asp Gly Arg 1325 1330 1335Asp Met Val Tyr Pro Tyr Pro Gly Ala Arg Ala Glu Glu Lys Leu 1340 1345 1350Gly Gly Thr Arg Asp Pro Thr Tyr Gln Glu Arg Ala Ala Pro Gln 1355 1360 1365Thr Gln Pro Leu Gly Lys Glu Thr Asp Ser Leu Ser Ala Gly Phe 1370 1375 1380Val Val Val Met Gly Val Asp Leu Ser Arg Cys Gly Pro Asp His 1385 1390 1395Pro Ala Ser Arg Cys Pro Trp Asp Pro Gly Leu Leu Leu Arg Phe 1400 1405 1410Leu Ala Ala Met Ala Ala Val Gly Ala Leu Glu Pro Leu Leu Pro 1415 1420 1425Gly Pro Leu Leu Ala Val His Pro His Ala Gly Thr Ala Pro Pro 1430 1435 1440Ala Asn Gln Leu Pro Trp Pro Val Leu Cys Ser Pro Val Ala Gly 1445 1450 1455Val Ile Leu Leu Ala Leu Gly Ala Leu Leu Val Leu Gln Leu Ile 1460 1465 1470Arg Arg Arg Arg Arg Glu His Gly Ala Leu Trp Leu Pro Pro Gly 1475 1480 1485Phe Thr Arg Arg Pro Arg Thr Gln Ser Ala Pro His Arg Arg Arg 1490 1495 1500Pro Pro Leu Gly Glu Asp Ser Ile Gly Leu Lys Ala Leu Lys Pro 1505 1510 1515Lys Ala Glu Val Asp Glu Asp Gly Val Val Met Cys Ser Gly Pro 1520 1525 1530Glu Glu Gly Glu Glu Val Gly Gln Ala Glu Glu Thr Gly Pro Pro 1535 1540 1545Ser Thr Cys Gln Leu Trp Ser Leu Ser Gly Gly Cys Gly Ala Leu 1550 1555 1560Pro Gln Ala Ala Met Leu Thr Pro Pro Gln Glu Ser Glu Met Glu 1565 1570 1575Ala Pro Asp Leu Asp Thr Arg Gly Pro Asp Gly Val Thr Pro Leu 1580 1585 1590Met Ser Ala Val Cys Cys Gly Glu Val Gln Ser Gly Thr Phe Gln 1595 1600 1605Gly Ala Trp Leu Gly Cys Pro Glu Pro Trp Glu Pro Leu Leu Asp 1610 1615 1620Gly Gly Ala Cys Pro Gln Ala His Thr Val Gly Thr Gly Glu Thr 1625 1630 1635Pro Leu His Leu Ala Ala Arg Phe Ser Arg Pro Thr Ala Ala Arg 1640 1645

1650Arg Leu Leu Glu Ala Gly Ala Asn Pro Asn Gln Pro Asp Arg Ala 1655 1660 1665Gly Arg Thr Pro Leu His Ala Ala Val Ala Ala Asp Ala Arg Glu 1670 1675 1680Val Cys Gln Leu Leu Leu Arg Ser Arg Gln Thr Ala Val Asp Ala 1685 1690 1695Arg Thr Glu Asp Gly Thr Thr Pro Leu Met Leu Ala Ala Arg Leu 1700 1705 1710Ala Val Glu Asp Leu Val Glu Glu Leu Ile Ala Ala Gln Ala Asp 1715 1720 1725Val Gly Ala Arg Asp Lys Trp Gly Lys Thr Ala Leu His Trp Ala 1730 1735 1740Ala Ala Val Asn Asn Ala Arg Ala Ala Arg Ser Leu Leu Gln Ala 1745 1750 1755Gly Ala Asp Lys Asp Ala Gln Asp Asn Arg Glu Gln Thr Pro Leu 1760 1765 1770Phe Leu Ala Ala Arg Glu Gly Ala Val Glu Val Ala Gln Leu Leu 1775 1780 1785Leu Gly Leu Gly Ala Ala Arg Glu Leu Arg Asp Gln Ala Gly Leu 1790 1795 1800Ala Pro Ala Asp Val Ala His Gln Arg Asn His Trp Asp Leu Leu 1805 1810 1815Thr Leu Leu Glu Gly Ala Gly Pro Pro Glu Ala Arg His Lys Ala 1820 1825 1830Thr Pro Gly Arg Glu Ala Gly Pro Phe Pro Arg Ala Arg Thr Val 1835 1840 1845Ser Val Ser Val Pro Pro His Gly Gly Gly Ala Leu Pro Arg Cys 1850 1855 1860Arg Thr Leu Ser Ala Gly Ala Gly Pro Arg Gly Gly Gly Ala Cys 1865 1870 1875Leu Gln Ala Arg Thr Trp Ser Val Asp Leu Ala Ala Arg Gly Gly 1880 1885 1890Gly Ala Tyr Ser His Cys Arg Ser Leu Ser Gly Val Gly Ala Gly 1895 1900 1905Gly Gly Pro Thr Pro Arg Gly Arg Arg Phe Ser Ala Gly Met Arg 1910 1915 1920Gly Pro Arg Pro Asn Pro Ala Ile Met Arg Gly Arg Tyr Gly Val 1925 1930 1935Ala Ala Gly Arg Gly Gly Arg Val Ser Thr Asp Asp Trp Pro Cys 1940 1945 1950Asp Trp Val Ala Leu Gly Ala Cys Gly Ser Ala Ser Asn Ile Pro 1955 1960 1965Ile Pro Pro Pro Cys Leu Thr Pro Ser Pro Glu Arg Gly Ser Pro 1970 1975 1980Gln Leu Asp Cys Gly Pro Pro Ala Leu Gln Glu Met Pro Ile Asn 1985 1990 1995Gln Gly Gly Glu Gly Lys Lys 2000 47135DNAArtificial sequencesequence is synthesized 47atgggtccag gtgcaagagg tagaaggcgt agaaggagac caatgagccc 50acctcctccg ccacctccag tgagagcact gcctttgctg ttgctgctgg 100ctggacctgg tgcagcagct cctccttgcc tggac 13548135DNAArtificial sequenceSequence is synthesized 48atggggccgg gggcccgtgg ccgccgccgc cgccgtcgcc cgatgtcgcc 50gccaccgcca ccgccacccg tgcgggcgct gcccctgctg ctgctgctag 100cggggccggg ggctgcagcc cccccttgcc tggac 135

* * * * *


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