Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
USE OF ANGIOPOIETINS IN PROMOTING BLOOD COAGULATION AND IN THE TREATMENT OF BLOOD COAGULATION DISORDERS
Document Type and Number:
WIPO Patent Application WO/2017/079556
Kind Code:
A1
Abstract:
The present invention provides methods for promoting blood coagulation and/or treating blood coagulation disorders in a subject in need thereof. The methods of the present invention comprise administering to a subject in need thereof a therapeutically effective amount of a pharmaceutical composition comprising an angiopoietin protein or a modified angiopoietin protein molecule such as AngF1 -Fc-F1 or AngF2-Fc-F2.

Inventors:
DALY CHRISTOPHER (US)
DAVIS SAMUEL (US)
Application Number:
PCT/US2016/060527
Publication Date:
May 11, 2017
Filing Date:
November 04, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
REGENERON PHARMA (US)
International Classes:
A61P7/04; A61K38/18
Domestic Patent References:
WO2015066426A22015-05-07
WO2001005825A22001-01-25
Foreign References:
US7691366B22010-04-06
US7008781B12006-03-07
US7008781B12006-03-07
US6265564B12001-07-24
US6441137B12002-08-27
US6825008B22004-11-30
Other References:
BENJAMIN WU ET AL: "Pharmacokinetics of Peptide-Fc Fusion Proteins", JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 103, no. 1, 27 January 2014 (2014-01-27), pages 53 - 64, XP055128807, ISSN: 0022-3549, DOI: 10.1002/jps.23783
DAVIS S ET AL: "ANGIOPOIETINS HAVE DISTINCT MODULAR DOMAINS ESSENTIAL FOR RECEPTOR BINDING, DIMERIZATION AND SUPERCLUSTERING", NATURE STRUCTURAL BIOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 10, no. 1, 1 January 2003 (2003-01-01), pages 38 - 44, XP008052277, ISSN: 1072-8368, DOI: 10.1038/NSB880
ANASTASIOU GEORGIA ET AL: "Thrombomodulin as a regulator of the anticoagulant pathway: implication in the development of thrombosis", BLOOD COAGULATION & FIBRINOLYSIS, vol. 23, no. 1, January 2012 (2012-01-01), pages 1 - 10, XP055342516
ESMON, C.T. ET AL., J. BIOL. CHEM., vol. 257, 1982, pages 7944 - 7947
OWEN, W.G. ET AL., J. BIOL. CHEM., vol. 256, 1981, pages 5532 - 5535
VAN DE WOUWER, M. ET AL., ARTERIOSCLEROSIS, THROMBOSIS, AND VASCULAR BIOLOGY, vol. 24, 2004, pages 1374 - 1383
WEILER-GUETTLER, H. ET AL., THE JOURNAL OF CLINICAL INVESTIGATION, vol. 101, 1998, pages 1983 - 1991
ESMON, C.T., PROC. NATL. ACAD. SCI. U.S.A., vol. 78, 1981, pages 2249 - 2252
THURSTON ET AL., NAT. MED., 2000
DAVIS, S. ET AL., NAT. STRUCT. BIOL., vol. 10, 2003, pages 38 - 44
POWELL ET AL.: "Compendium of excipients for parenteral formulations", J PHARM SCI TECHNOL, vol. 52, 1998, pages 238 - 311
WU ET AL., J. BIOL. CHEM., vol. 262, 1987, pages 4429 - 4432
LANGER AND WISE: "Medical Applications of Controlled Release", 1984, CRC PRESS
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
DAVIS, S. ET AL., SCIENCE, vol. 266, 1994, pages 816 - 819
BARTON, W.A. ET AL., STRUCTURE, vol. 13, 2005, pages 825 - 832
BARTON, W.A., NAT. STRUCT. MOL. BIOL., vol. 13, 2006, pages 524 - 532
PETERS, K.G. ET AL., BRITISH JOURNAL OF CANCER, vol. 77, 1998, pages 51 - 56
HACKENG, T.M. ET AL., THE BIOCHEMICAL JOURNAL, vol. 319, 1996, pages 399 - 405
HOCKIN, M.F. ET AL., ARTERIOSCLEROSIS, THROMBOSIS, AND VASCULAR BIOLOGY, vol. 17, 1997, pages 2765 - 2775
SLUNGAARD, A ET AL., BLOOD, vol. 102, 2003, pages 146 - 151
LASZIK Z. ET AL., CIRCULATION, vol. 96, 1997, pages 3633 - 3640
DALY, C. ET AL., GENES & DEVELOPMENT., vol. 18, 2004, pages 1060 - 1071
LI, J.J. ET AL., THROMB. HAEMOST., vol. 85, 2001, pages 204 - 206
PETERSON J.E. ET AL., AMERICAN JOURNAL OF HEMATOLOGY, vol. 85, 2010, pages 487 - 493
FIEDLER, U. ET AL., BLOOD, vol. 103, 2004, pages 4150 - 4156
GOEDE, V. ET AL., BRITISH JOURNAL OF CANCER, vol. 103, 2010, pages 1407 - 1414
WANG, X. ET AL., TRANSLATIONAL ONCOLOGY, vol. 7, 2014, pages 188 - 195
Attorney, Agent or Firm:
TERMES, Lance et al. (US)
Download PDF:
Claims:
What is claimed is:

1 . A method of treating, preventing or ameliorating at least one symptom, indication or complication of a blood coagulation disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of an angiopoietin protein and a pharmaceutically acceptable carrier.

2. The method of claim 1 , wherein the blood coagulation disorder is selected from the group consisting of inborn platelet pathologies such as thrombasthenia of Glanzmann and Naegeli (Glanzmann thrombasthenia), Bernard-Soulier syndrome (abnormal glycoprotein Ib-IX-V complex), gray platelet syndrome (deficient alpha granules), delta storage pool deficiency (deficient dense granules), and Von Willebrand disease; platelet disorders caused by insufficient production including myelodysplastic syndrome or other bone marrow disorders or by destruction by the immune system including immune thrombocytopenic purpura/ITP;

coagulation factor disorders such as hemophilias including hemophilia A (factor VIII deficiency), hemophilia B (factor IX deficiency or "Christmas disease") and hemophilia C (factor XI deficiency, mild bleeding tendency); deficiency of Vitamin K, Factor XII mutations,

thrombocytopenia, uremia, and congenital afibrinogenemia; liver failure including acute, chronic, early and end-stage; and disorders associated with warfarin treatment, aspirin treatment, surgery, and injury.

3. The method of claim 1 , wherein the angiopoietin protein binds purified soluble extracellular domain of thrombomodulin with an EC50 value of less than 100 nM.

4. The method of claim 1 , wherein the angiopoietin protein is administered intravenously.

5. The method of claim 1 , wherein the angiopoietin protein is administered subcutaneously.

6. The method of any one of claims 1 -5, wherein the angiopoietin protein is hAngl .

7. The method of any one of claims 1 -5, wherein the angiopoietin protein is hAng2.

8. The method of any one of claim 1 -5, wherein the angiopoietin protein is a modified angiopoietin protein.

9. The method of claim 8, wherein the modified angiopoietin protein comprises a fusion protein comprising angiopoietin-1 or a fragment thereof fused to a multimerizing domain.

10. The method of claim 9, wherein the multimerizing domain is an

immunoglobulin Fc fragment.

1 1 . The method of claim 10, wherein the fusion protein comprises at least one fibrinogen-like domain of angiopoietin-1 fused to an Fc fragment.

12. The method of claim 1 1 , wherein the fusion protein comprises a first fibrinogen-like domain of angiopoietin-1 fused at its C-terminal end to the N-terminal end of an Fc fragment and the Fc fragment fused at its C-terminal end to the N-terminal end of a second fibrinogen-like domain of angiopoietin-1 .

13. The method of claim 8, wherein the modified angiopoietin is AngF1 -Fc-F1 .

14. The method of claim 8, wherein the modified angiopoietin comprises the amino acid sequence of SEQ ID NO: 2.

15. The method of claim 8, wherein the modified angiopoietin comprises a fusion protein comprising angiopoietin-2 or a fragment thereof fused to a multimerizing domain.

16. The method of claim 15, wherein the multimerizing domain is an immunoglobulin Fc fragment.

17. The method of claim 16 wherein the fusion protein comprises at least one fibrinogen-like domain of angiopoietin-2 fused to an Fc fragment.

18. The method of claim 17, wherein the fusion protein comprises a first fibrinogen-like domain of angiopoietin-2 fused at its C-terminal end to the N-terminal end of an Fc fragment and the Fc fragment fused at its C-terminal end to the N-terminal end of a second fibrinogen-like domain of angiopoietin-2.

19. The method of claim 8, wherein the modified angiopoietin is AngF2-Fc-F2.

20. The method of claim 8, wherein the modified angiopoietin comprises the amino acid sequence of SEQ ID NO:4.

21 . A method of stimulating blood coagulation, comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of an angiopoietin protein and a pharmaceutically acceptable carrier.

22. The method of claim 21 , wherein the subject is undergoing surgery.

23. The method of claim 21 , wherein the subject is bleeding due to injury.

24. The method of claim 21 , wherein the angiopoietin protein binds purified soluble extracellular domain of thrombomodulin with an EC50 value of less than 100 nM.

25. The method of claim 21 , wherein the angiopoietin protein is administered intravenously.

26. The method of claim 21 , wherein the angiopoietin protein is administered subcutaneously.

27. The method of claim 21 , wherein the angiopoietin protein is administered at a site of injury or a site associated with a surgical procedure.

28. The method of any one of claims 21 -27, wherein the angiopoietin protein is hAngl .

29. The method of any one of claims 21 -27, wherein the angiopoietin protein is hAng2.

30. The method of any one of claims 21 -27, wherein the angiopoietin protein is a modified angiopoietin protein.

31 . The method of claim 30, wherein the modified angiopoietin comprises a fusion protein comprising angiopoietin-1 or a fragment thereof fused to a multimerizing domain.

32. The method of claim 31 , wherein the multimerizing domain is an immunoglobulin Fc fragment.

33. The method of claim 32, wherein the fusion protein comprises at least one fibrinogen-like domain of angiopoietin-1 fused to an Fc fragment.

34. The method of claim 33, wherein the fusion protein comprises a first fibrinogen-like domain of angiopoietin-1 fused at its C-terminal end to the N-terminal end of an Fc fragment and the Fc fragment fused at its C-terminal end to the N-terminal end of a second fibrinogen-like domain of angiopoietin-1 .

35. The method of claim 30, wherein the modified angiopoietin is AngF1 -Fc-F1 .

36. The method of claim 30, wherein the modified angiopoietin comprises the amino acid sequence of SEQ ID NO: 2.

37. The method of claim 30, wherein the modified angiopoietin comprises a fusion protein comprising angiopoietin-2 or a fragment thereof fused to a multimerizing domain.

38. The method of claim 37, wherein the multimerizing domain is an immunoglobulin Fc fragment.

39. The method of claim 38, wherein the fusion protein comprises at least one fibrinogen-like domain of angiopoietin-2 fused to an Fc fragment.

40. The method of claim 39, wherein the fusion protein comprises a first fibrinogen-like domain of angiopoietin-2 fused at its C-terminal end to the N-terminal end of an Fc fragment and the Fc fragment fused at its C-terminal end to the N-terminal end of a second fibrinogen-like domain of angiopoietin-2.

41 . The method of claim 30, wherein the modified angiopoietin is AngF2-Fc-F2.

42. The method of claim 30 wherein the modified angiopoietin comprises the amino acid sequence of SEQ ID NO:4.

Description:
USE OF ANGIOPOIETINS IN PROMOTING BLOOD COAGULATION AND IN THE

TREATMENT OF BLOOD COAGULATION DISORDERS

REFERENCE TO A SEQUENCE LISTING

[0001 ] This application incorporates by reference the Sequence Listing submitted in

Computer Readable Form as file 10194WO01 -Sequence.txt, created on October 31 , 2016, and containing 36,151 bytes.

FIELD OF THE INVENTION

[0002] The present invention resides in the field of medicine, and relates to the use of angiopoietin molecules or variants thereof to promote blood coagulation and/or treat blood coagulation disorders in a subject in need thereof.

BACKGROUND

[0003] Excessive bleeding or slowed blood coagulation leads to death or debilitation for patients. Blood coagulation disorders in humans are caused by inborn genetic conditions or may be a side effect of drug treatment for other conditions. Blood loss may occur during surgery or after injury. Injury is the second leading cause of death for people aged five to 45 years, and over four million people worldwide die of injuries every year, often because of extensive blood loss. Current treatment options include adsorbent chemicals (such as zeolites, e.g., QuikClot), other hemostatic agents (such as microfibrillar collagen hemostat (MCH), chitosan hemostat, and styptics), thrombin, fibrin glue, desmopressin, coagulation factor concentrates, prothrombin complex concentrate, cryoprecipitate, fresh frozen plasma, recombinant Factor VII, and antifibrinolytic drugs such as tranexamic acid (TXA), epsilon-aminocaproic acid and

aminomethylbenzoic acid. Mechanical devices, such as a hemostatic clamp, are also used. There remains a need for effective therapeutic and preventive approaches to promote blood coagulation or treat blood coagulation disorders.

BRIEF SUMMARY OF THE INVENTION

[0004] According to one aspect of the present invention, methods are provided for stimulating blood coagulation, or treating, preventing or ameliorating at least one symptom, indication or complication of a blood coagulation disorder in a subject in need thereof. The present invention is predicated, in part, on the discovery that both angiopoietin-1 (Ang1 ) and angiopoietin-2 (Ang2) bind to and modulate the function of thrombomodulin (TM), an endothelial cell surface molecule that plays an essential role as a coagulation regulator. TM functions as a cofactor in the thrombin-mediated activation of protein C, an anticoagulant protein, and of thrombin- activatable fibrinolysis inhibitor (TAFI), an inhibitor of blood clot dissolution. As detailed in the Examples and the discussion below, both Ang1 and Ang2 inhibited activation of protein C and TAFI in cultured endothelial cells, and inhibited the binding of thrombin to TM in vitro. These results suggest a previously undescribed role for angiopoietins in the regulation of hemostasis.

[0005] In one aspect, the present invention provides a method of treating, preventing or ameliorating at least one symptom, indication or complication of a blood coagulation disorder comprising administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective amount of an angiopoietin protein and a pharmaceutically acceptable carrier. In another aspect, the present invention provides for the use of a therapeutically effective amount of an angiopoietin protein (i) for treating, preventing or ameliorating at least one symptom, indication or complication of a blood coagulation disorder, or (ii) in the

manufacture of a medicament for treating, preventing or ameliorating at least one symptom, indication or complication of a blood coagulation disorder.

[0006] In one embodiment, the blood coagulation disorder is selected from the group consisting of inborn platelet pathologies such as thrombasthenia of Glanzmann and Naegeli (Glanzmann thrombasthenia), Bernard-Soulier syndrome (abnormal glycoprotein Ib-IX-V complex), gray platelet syndrome (deficient alpha granules), delta storage pool deficiency (deficient dense granules), and Von Willebrand disease; platelet disorders caused by insufficient production including myelodysplastic syndrome or other bone marrow disorders or by destruction by the immune system including immune thrombocytopenic purpura/ITP;

coagulation factor disorders such as hemophilias including hemophilia A (factor VIII deficiency), hemophilia B (factor IX deficiency or "Christmas disease") and hemophilia C (factor XI deficiency, mild bleeding tendency); deficiency of Vitamin K, Factor XII mutations,

thrombocytopenia, uremia, and congenital afibrinogenemia; liver failure including acute, chronic, early and end-stage; and disorders associated with warfarin treatment, aspirin treatment, surgery, and injury.

[0007] In another aspect, the present invention provides a method of stimulating blood coagulation, comprising administering to a subject in need thereof a pharmaceutical

composition comprising a therapeutically effective amount of an angiopoietin protein and a pharmaceutically acceptable carrier. In one embodiment, the subject is undergoing surgery. In one embodiment, the subject is bleeding due to injury. In yet another aspect, the present invention provides for the use of a therapeutically effective amount of an angiopoietin protein (i) for stimulating blood coagulation, or (ii) in the manufacture of a medicament for stimulating blood coagulation.

[0008] In some cases, the angiopoietin protein is administered at a site of injury or a site associated with a surgical procedure.

[0009] In various embodiments of the methods and uses discussed above, the angiopoietin protein of the pharmaceutical composition may be specified binding properties and/or comprise a native or modified angiopoietin protein.

[0010] In an embodiment, the angiopoietin protein binds a purified soluble extracellular domain of thrombomodulin with an EC50 value of less than 100 nM.

[0011 ] In some cases, the angiopoietin protein is hAngl . In other cases, the angiopoietin protein is hAng2. In an embodiment, the angiopoietin protein is a modified angiopoietin protein.

[0012] In some embodiments, the modified angiopoietin protein comprises a fusion protein comprising angiopoietin-1 or a fragment thereof fused to a multimerizing domain. In some cases, the multimerizing domain is an immunoglobulin Fc fragment. In an embodiment, the fusion protein comprises at least one fibrinogen-like domain of angiopoietin-1 fused to an Fc fragment. For example, the fusion protein may comprise a first fibrinogen-like domain of angiopoietin-1 fused at its C-terminal end to the N-terminal end of an Fc fragment and the Fc fragment fused at its C-terminal end to the N-terminal end of a second fibrinogen-like domain of angiopoietin-1 . In some cases, the modified angiopoietin is AngF1 -Fc-F1 . In one embodiment, the modified angiopoietin comprises an amino acid sequence of SEQ ID NO: 2.

[0013] In some embodiments, the modified angiopoietin protein comprises a fusion protein comprising angiopoietin-2 or a fragment thereof fused to a multimerizing domain. In some cases, the multimerizing domain is an immunoglobulin Fc fragment. In an embodiment, the fusion protein comprises at least one fibrinogen-like domain of angiopoietin-2 fused to an Fc fragment. For example, the fusion protein may comprise a first fibrinogen-like domain of angiopoietin-2 fused at its C-terminal end to the N-terminal end of an Fc fragment and the Fc fragment fused at its C-terminal end to the N-terminal end of a second fibrinogen-like domain of angiopoietin-2. In some cases, the modified angiopoietin is AngF2-Fc-F2. In one embodiment, the modified angiopoietin comprises an amino acid sequence of SEQ ID NO:4.

[0014] In some cases, the angiopoietin protein is administered intravenously. In some cases, the angiopoietin protein is administered subcutaneously. [0015] In certain embodiments, the angiopoietin is administered in combination with a second therapeutic agent or therapy.

[0016] Other embodiments of the present invention will become apparent from a review of the ensuing detailed description.

BRIEF DESCRIPTION OF THE FIGURES

[0017] Figures 1 A, 1 B and 1 C show that Ang1 and Ang2 interact with TM via their fibrinogen-like domains: Fig. 1 A shows that Ang1 -FD-Fc and Ang2-FD-Fc bind saturably to COS cells overexpressing TM; Fig. 1 B shows that BowAngl and BowAng2 bind the soluble extracellular domains of either TM or Tie2; Fig. 1 C shows that soluble Tie2 extracellular domain competes with the soluble extracellular domain of TM for binding to BowAngl or BowAng2.

[0018] Figures 2A, 2B, 2C, 2D and 2E show that Ang1 and Ang2 inhibit TM-dependent formation of APC and aTAFI: Fig. 2A shows that native Ang1 and Ang2 inhibit TM-dependent formation of APC in cultured HUVECs; Fig. 2B shows that native Ang1 inhibits TM-dependent formation of APC, while PF4 increases TM-dependent formation of APC, in cultured HUVECs; Fig. 2C shows that native Ang1 and Ang2 inhibit TM-dependent formation of aTAFI in cultured HUVECs; Fig. 2D shows that native Ang1 and Ang2 inhibit TM-dependent formation of APC in COS cells overexpressing TM but lacking Tie2 expression; Fig. 2E shows that native Ang1 and Ang2 inhibit TM-dependent formation of aTAFI in COS cells overexpressing TM but lacking Tie2 expression.

[0019] Figures 3A and 3B show that inhibition of APC formation by Ang1 is independent of Tie2: Fig. 3A shows inhibition of Tie2 expression in HUVECs infected with adenovirus encoding a Tie2 specific shRNA; Fig. 3B shows that native Ang1 inhibits TM-dependent formation of APC in HUVECs infected with adenovirus encoding a Tie2 specific shRNA.

[0020] Figures 4A and 4B show that Ang1 and Ang2 inhibit binding of thrombin to TM: Fig. 4A shows that native Ang1 inhibits association of thrombin to soluble extracellular domain of TM; Fig. 4B shows that native Ang2 inhibits association of thrombin to soluble extracellular domain of TM.

[0021 ] Figures 5A, 5B, 5C and 5D show that Ang1 is present in platelets and is released upon activation: Fig. 5A shows that Ang1 is present in mouse platelets; Fig. 5B shows that Ang1 levels are higher in mouse serum than in mouse plasma; Fig. 5C shows that Ang1 levels are higher in plasma prepared from mice treated with thrombin compared to plasma prepared from untreated mice; Fig. 5D shows that Ang1 levels are higher in plasma prepared from blood collected at the site of tail nicks in mice compared to plasma prepared blood collected by cardiac puncture in mice (systemic).

[0022] Figures 6A and 6B show that intravenous administration of BowAngl and BowAng2 to mice promotes a rapid increase in circulating thrombin/antithrombin level and fibrin deposition in lung : Fig. 6A shows that BowAng l and BowAng2 promote an increase in fibrin deposition in lung tissue in mice; Fig. 6B shows that BowAng l and BowAng2 promote an increase in circulating thrombin/antithrombin level in mice.

DETAILED DESCRIPTION

[0023] Before the present invention is described, it is to be understood that this invention is not limited to particular methods and experimental conditions described, as such methods and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.

[0024] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. As used herein, the term "about," when used in reference to a particular recited numerical value, means that the value may vary from the recited value by no more than 1 %. For example, as used herein, the expression "about 1 00" includes 99 and 101 and all values in between (e.g., 99.1 , 99.2, 99.3, 99.4, etc.).

[0025] Although any methods and materials similar or equivalent to those described herein can be used in the practice of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference in their entirety.

Definitions

[0026] As used herein, an "angiopoietin" includes Angiopoietin-1 (Ang1 ) or Angiopoietin-2 (Ang2), unless otherwise specified. The term "hAngl " refers to human Ang1 , and the term "hAng2" refers to human Ang2.

[0027] The term "EC 5 o" or "EC50", as used herein, refers to the half maximal effective concentration, which includes the concentration of a ligand that induces a response, for example a cellular response, halfway between the baseline and maximum after a specified exposure time. The EC 5 o essentially represents the concentration of a ligand where 50% of its maximal effect is observed.

[0028] The term "IC 5 o" or "IC50", as used herein, refers to the half maximal inhibitory concentration of a cellular response. In other words, the measure of the effectiveness of a particular moiety {e.g. protein, compound, or molecule) in inhibiting biological or biochemical function, wherein an assay quantitates the amount of such moiety needed to inhibit a given biological process.

[0029] The terms "modified angiopoietin" or "modified angiopoietin protein" refer to

recombinant angiopoietins {e.g., angiopoietin expressed in an adenoviral vector; Thurston et al 2000, Nat. Med.), mutant and chimeric forms of angiopoietins, and fusion proteins comprising angiopoietin or a fragment thereof that specifically bind thrombomodulin (TM).

[0030] The terms "native Ang1 " and "native Ang2," as used herein, refer to a full-length Ang1 or Ang2, respectively, without N-terminal signal peptide.

[0031 ] The term "preventing," as used herein refers to preventing development of a symptom, indication or complication of a blood coagulation disorder.

[0032] As used herein, the term "subject" refers to an animal, preferably a mammal, that exhibits one or more symptoms, indications or complications of a blood coagulation disorder, and/or who has been diagnosed with a blood coagulation disorder and/or is in need of amelioration, prevention and/or treatment of a blood coagulation disorder. The term "a subject in need thereof" may also include, e.g., subjects who, prior to treatment, exhibit (or have exhibited) one or more symptoms or indications of a blood coagulation disorder including but not limited to excessive bleeding and slow blood coagulation time.

[0033] As used herein, the terms "treat", "treating", or the like, mean to alleviate a symptom or a complication, eliminate the causation of a symptom or a complication either on a temporary or permanent basis, or to prevent or slow the appearance of a symptom or complication of a blood coagulation disorder in the subject. In the context of the present invention, the terms "treat", "treating", or the like, refer to e.g., reducing bleeding or reducing time needed to coagulate blood in a patient in need thereof.

[0034] By the phrase "therapeutically effective amount" is meant an amount that produces the desired effect for which it is administered. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding). Methods for Treating, Preventing or Ameliorating Blood Coagulation Disorders or Promoting Blood Coagulation.

[0035] The angiopoietins were identified as ligands for the endothelial-specific receptor tyrosine kinase, Tie2. Angiopoietin/Tie2 signaling profoundly influences angiogenic processes, both during development and in the adult, for example in tumor vasculature. Angiopoietins have not previously been implicated in any processes other than the regulation of blood vessel formation and function via modulation of Tie2 activity.

[0036] As shown herein, both Angiopoietin-1 (Ang1 ) and Angiopoietin-2 (Ang2) bind to and modulate the function of thrombomodulin (TM), an endothelial cell surface molecule that plays an essential role as a coagulation regulator. TM functions as a cofactor in the thrombin- mediated activation of protein C, an anticoagulant protein (Esmon, C.T., et al., 1982, J. Biol. Chem.257:7944-7947; Owen, W.G., et al., 1981 , J. Biol. Chem. 256:5532-5535; Van de

Wouwer, M., et al., 2004, Arteriosclerosis, thrombosis, and vascular biology, 24:1374-1383; Weiler-Guettler, H., et al., 1998, The Journal of clinical investigation, 101 :1983-1991 ; Esmon, C.T., 1981 , Proc. Natl. Acad. Sci. U.S.A., 78:2249-2252), and of thrombin-activatable fibrinolysis inhibitor (TAFI), an inhibitor of blood clot dissolution.

[0037] As demonstrated in the examples, Ang1 and Ang2 inhibited both activation of protein C and TAFI in cultured endothelial cells, and inhibited the binding of thrombin to TM in vitro. Administration of thrombin to mice rapidly increased plasma Ang1 levels, and Ang1 levels were significantly elevated in plasma prepared from wound blood. Further, administration of a modified angiopoietin promoted a rapid increase in circulating thrombin/antithrombin level and fibrin deposition in the lungs of mice. These results indicate a previously undescribed role for angiopoietins in the regulation of hemostasis.

[0038] Accordingly, the present invention provides methods for treating, preventing or ameliorating at least one symptom, indication or complication of a blood coagulation disorder, or stimulating blood coagulation, in a subject. The methods according to these aspects of the invention comprise administering a therapeutically effective amount of a pharmaceutical composition comprising an angiopoietin protein or a modified angiopoietin protein to a subject in need thereof.

[0039] In various embodiments, the blood coagulation disorder may include, but is not limited to, inborn platelet pathologies such as thrombasthenia of Glanzmann and Naegeli (Glanzmann thrombasthenia), Bernard-Soulier syndrome (abnormal glycoprotein Ib-IX-V complex), gray platelet syndrome (deficient alpha granules), delta storage pool deficiency (deficient dense granules), and Von Willebrand disease. Blood coagulation disorders also include but are not limited to, platelet disorders caused by insufficient production (e.g., in myelodysplastic syndrome or other bone marrow disorders) or destruction by the immune system (immune thrombocytopenic purpura/ITP). Blood coagulation disorders also include, but are not limited to, coagulation factor disorders such as hemophilias including hemophilia A (factor VIII deficiency), hemophilia B (factor IX deficiency or "Christmas disease") and hemophilia C (factor XI deficiency, mild bleeding tendency). Blood coagulation disorders also include, but are not limited to, deficiency of Vitamin K, Factor XII mutations, thrombocytopenia, uremia, and congenital afibrinogenemia. Deficiencies in blood coagulation may also be associated with liver failure (acute and chronic forms, early liver failure, end-stage liver failure).

[0040] Bleeding or excessive bleeding may also be associated with treatment of a patient with warfarin or aspirin, or may occur during surgery or after injury. In particular embodiments, promotion of blood coagulation at a site of injury or surgery is contemplated.

[0041 ] The present invention includes methods for treating, preventing or reducing the severity of a blood coagulation disorder, or stimulating blood coagulation, comprising

administering a therapeutically effective amount of a pharmaceutical composition comprising an angiopoietin protein or a modified angiopoietin protein to a subject in need thereof, wherein the pharmaceutical composition is administered to the subject in a single dose, or in multiple doses, e.g., as part of a specific therapeutic dosing regimen. For example, the therapeutic dosing regimen may comprise administering multiple doses of the pharmaceutical composition to the subject at a frequency of about once a day, once every two days, once every three days, once every four days, once every five days, once every six days, once a week, once every two weeks, once every three weeks, once every four weeks, once a month, once every two months, once every three months, once every four months, or less frequently. In some embodiments, the therapeutic dosing regimen comprises administering multiple doses of the pharmaceutical composition to the subject at a frequency of about once a day, about 2 times a day, about 3 times a day or more than 4 times a day.

[0042] In certain embodiments the angiopoietin protein or modified angiopoietin protein is administered subcutaneously, intravenously, intracranial^, intraventricularly, or delivered systemically in an adenoviral vector to a subject in need thereof. In certain embodiments the angiopoietin protein or modified angiopoietin protein is administered at a site of injury or a site associated with a surgical procedure. In one embodiment, administration at a site of injury or surgery is by injection. In some cases, the site-specific administration of the angiopoietin protein or modified angiopoietin protein results in a high relative concentration of angiopoietin at the site to promote blood coagulation. In one embodiment, the "high relative concentration" is measured against plasma levels of angiopoietin, and may be, for example, greater than 10 nM, greater than 50 nM or greater than 100 nM.

Modified Angiopoietins

[0043] The methods of the present invention comprise administering to a subject in need thereof a therapeutic composition comprising an angiopoietin or a variant thereof. In some cases, the variant angiopoietin is a modified angiopoietin.

[0044] Non-limiting examples of categories of modified angiopoietins include recombinant angiopoietins {e.g., angiopoietin expressed in an adenoviral vector; Thurston et al 2000, Nat. Med.), mutant and chimeric forms of angiopoietins, and fusion proteins comprising angiopoietin or a fragment thereof that specifically bind thrombomodulin (TM).

[0045] According to certain exemplary embodiments of the present invention, the modified angiopoietin is a fusion protein comprising one or more domains of the angiopoietin molecule fused to a multimerizing domain. In general terms, the multimerizing domain(s) of the present invention function to connect the various components of the angiopoietin molecule {e.g., the fibrinogen-like domains) with one another. As used herein, a "multimerizing domain" is any macromolecule that has the ability to associate (covalently or non-covalently) with a second macromolecule of the same or similar structure or constitution. For example, a multimerizing domain may be a polypeptide comprising an immunoglobulin CH3 domain. A non-limiting example of a multimerizing domain is an Fc portion of an immunoglobulin, e.g., an Fc domain of an IgG selected from the isotypes lgG1 , lgG2, lgG3, and lgG4, as well as any allotype within each isotype group. In certain embodiments, the multimerizing domain is an Fc fragment or an amino acid sequence of 1 to about 200 amino acids in length containing at least one cysteine residues. In other embodiments, the multimerizing domain is a cysteine residue or a short cysteine-containing peptide. Other multimerizing domains include peptides or polypeptides comprising or consisting of a leucine zipper, a helix-loop motif, or a coiled-coil motif.

[0046] In certain embodiments, the modified angiopoietin is a fusion protein comprising one or more fibrinogen-like domains of the angiopoietin-1 molecule fused to the Fc fragment of an immunoglobulin comprising any of the amino acid sequences, as set forth in US Patent No. 7,008,781 . In certain exemplary embodiments, the fusion protein that can be used in the context of the methods of the present invention comprises a first fibrinogen-like domain of angiopoietin fused at its C-terminal end to the N-terminal end of an IgG Fc fragment and the C- terminal of the Fc fragment fused to the N-terminal end of a second fibrinogen-like domain of angiopoietin (Davis, S., et al, 2003; Nat. Struct. Biol. 10:38-44), wherein the angiopoietin may be Ang1 or Ang2. According to certain exemplary embodiments, the methods of the present invention comprise the use of the modified angiopoietin referred to and known in the art as AngF1 -Fc-F1 . In certain embodiments, the modified angiopoietin is a dimer comprising two AngF1 -Fc-F1 s that associate through intramolecular association of the Fc fragments (also referred to as BowAngI , as disclosed in Davis, 2003, supra. According to certain embodiments, the fusion protein comprises the amino acid sequence of SEQ ID NO: 2. According to certain exemplary embodiments, the methods of the present invention comprise the use of the modified angiopoietin referred to and known in the art as AngF2-Fc-F2. In certain embodiments, the modified angiopoietin is a dimer comprising two AngF2-Fc-F2s that associate through intramolecular association of the Fc fragments (also referred to as BowAng2, as disclosed in Davis, 2003, supra). In some embodiments, the fusion protein comprises the amino acid sequence of SEQ ID NO: 4. BowAngI and BowAng2 contain the Ang1 or Ang2 fibrinogen domain followed by human Fc and another fibrinogen domain, so that following Fc-mediated dimerization, the proteins contain 4 angiopoietin fibrinogen domains (Davis, 2003, supra).

[0047] Other modified angiopoietins that can be used in the context of the methods of the present invention include any of the modified angiopoietin molecules as set forth in US Patent Nos. 6265564, 6441 137, and 6825008.

[0048] The present invention includes modified angiopoietins that bind purified soluble extracellular domain of TM with an EC 5 o value of less than about 100 nM, as measured by an in vitro assay, e.g., using the assay format as defined in Example 3 herein (e.g., assessing the ability of modified angiopoietins to bind purified soluble extracellular domain of TM), or a substantially similar assay. In certain embodiments, the modified angiopoietins of the present invention bind purified soluble extracellular domain of TM), with an EC 5 o value of less than about 90 nM, 80 nM, 70 nM, 60 nM, 50 nM, 40 nM, 30 nM, 20 nM or 10 nM as measured by an in vitro assay, e.g., using the assay format as defined in Example 3 herein (e.g., assessing the ability of modified angiopoietins to bind purified soluble extracellular domain of TM), or a substantially similar assay.

[0049] The present invention includes modified angiopoietins that inhibit APC formation in cultured HUVECs with an IC 5 o of less than about 100 nM as measured by an cell-based assay, e.g., using the assay format as defined in Example 5 herein (e.g., assessing the ability of modified angiopoietins to inhibit APC formation in cultured HUVECs), or a substantially similar assay. In certain embodiments, the modified angiopoietins of the present invention inhibit APC formation in a cultured HUVECs with an IC 5 o of less than about 90 nM, 80 nM, 70 nM, 60 nM, 50 nM, 40 nM, 30 nM, 20 nM or 10 nM as measured by an cell-based assay, e.g., using the assay format as defined in Example 5 herein (e.g., assessing the ability of modified angiopoietins to inhibit APC formation in cultured HUVECs), or a substantially similar assay.

Pharmaceutical Compositions

[0050] The present invention includes methods which comprise administering an angiopoietin protein or a modified angiopoietin protein to a subject wherein the angiopoietin protein or modified angiopoietin protein is contained within a pharmaceutical composition. The

pharmaceutical compositions of the invention may be formulated with suitable carriers {e.g., pharmaceutically acceptably carriers), excipients, and other agents that provide suitable transfer, delivery, tolerance, and the like. A multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, PA. These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTIN™), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. "Compendium of excipients for parenteral formulations" PDA (1998) J Pharm Sci Technol 52:238-31 1 .

[0051 ] Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the mutant viruses, receptor mediated endocytosis (see, e.g., Wu et al., 1987, J. Biol. Chem. 262: 4429-4432). Methods of administration include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The composition 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.

[0052] A pharmaceutical composition of the present invention can be delivered

subcutaneously or intravenously with a standard needle and syringe. In addition, with respect to subcutaneous delivery, a pen delivery device readily has applications in delivering a

pharmaceutical composition of the present invention. Such a pen delivery device can be reusable or disposable. A reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused. In a disposable pen delivery device, there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.

[0053] In certain situations, the pharmaceutical composition can be delivered in a controlled release system. In one embodiment, a pump may be used. In another embodiment, polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), 1984, CRC Press, Boca Raton, Florida. In yet another embodiment, a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, 1984, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138). Other controlled release systems are discussed in the review by Langer, 1990, Science 249:1527-1533.

[0054] The injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by known methods. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the angiopoietin protein or modified angiopoietin protein described above in a sterile aqueous medium or an oily medium

conventionally used for injections. As the aqueous medium for injections, there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol {e.g., ethanol), a polyalcohol {e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc. As the oily medium, there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc. The injection thus prepared is preferably filled in an appropriate ampoule.

[0055] Advantageously, the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients. Such dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc. Dosage

[0056] The amount of the angiopoietin protein or modified angiopoietin protein (e.g., AngF1 - Fc-F1 or AngF2-Fc-F2) administered to a subject according to the methods of the present invention is, generally, a therapeutically effective amount. As used herein, the phrase

"therapeutically effective amount" means an amount of the angiopoietin protein or modified angiopoietin protein that results in reduction in the severity or duration of a symptom, indication or complication of a blood coagulation disorder, or promotes blood coagulation.

[0057] In the case of an angiopoietin protein or a modified angiopoietin protein, a

therapeutically effective amount can be from about 0.05 mg to about 600 mg, e.g., about 0.05 mg, about 0.1 mg, about 1 .0 mg, about 1 .5 mg, about 2.0 mg, about 10 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, about 80 mg, about 90 mg, about 100 mg, about 1 10 mg, about 120 mg, about 130 mg, about 140 mg, about 150 mg, about 160 mg, about 170 mg, about 180 mg, about 190 mg, about 200 mg, about 210 mg, about 220 mg, about 230 mg, about 240 mg, about 250 mg, about 260 mg, about 270 mg, about 280 mg, about 290 mg, about 300 mg, about 310 mg, about 320 mg, about 330 mg, about 340 mg, about 350 mg, about 360 mg, about 370 mg, about 380 mg, about 390 mg, about 400 mg, about 410 mg, about 420 mg, about 430 mg, about 440 mg, about 450 mg, about 460 mg, about 470 mg, about 480 mg, about 490 mg, about 500 mg, about 510 mg, about 520 mg, about 530 mg, about 540 mg, about 550 mg, about 560 mg, about 570 mg, about 580 mg, about 590 mg, or about 600 mg, of the angiopoietin protein or modified angiopoietin protein.

[0058] The amount of the angiopoietin protein or modified angiopoietin protein contained within the individual doses may be expressed in terms of milligrams of protein per kilogram of the subject's body weight {i.e., mg/kg). For example, the modified angiopoietin may be administered to a subject at a dose of about 0.0001 to about 100 mg/kg of patient body weight. In certain embodiments, the angiopoietin protein or modified angiopoietin protein is

administered to a subject in need thereof at a dose of about 5 - 25 mg/kg of the subject's body weight.

EXAMPLES

[0059] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the methods and

compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric.

Example 1. Thrombomodulin binds Ang1 -FD-Fc in expression cloning study

[0060] cDNA expression libraries were constructed in the expression vector pJFE with mRNA isolated from human umbilical vein endothelial cells (HUVECs) and from the endothelial-like cell line EA.hy926, using a kit (Invitrogen). The libraries were transfected into dishes of COS cells and screened as in Davis, S., et al., (1994; Science. 266:816-819) with the chimeric molecule Ang1 -FD-Fc, consisting of the Tie2-binding fibrinogen-like domain (FD) of Ang1 fused to a human Fc tag (Davis, 2003, supra, SEQ ID NO:5), at a concentration of 1 μg/ml. Stained cells were rescued, and plasmid DNA extracted from them was progressively enriched until isolated clones were obtained. Out of eight independent clones, one of the associated cDNAs coded for Tie2, and the other seven clones, differing only in the length of untranslated regions, contained cDNAs encoding thrombomodulin (TM), a cell surface protein highly expressed in endothelial cells (Esmon, 1981 , supra). Only rare cells were stained on dishes of library-transfected COS cells, indicating that Ang1 -FD-Fc binding to Tie2 and TM is highly specific.

Example 2. Ang1 -FD-Fc and Ang2-FD-Fc bind saturably to COS cells overexpressing TM

[0061 ] In this Example, the ability of Ang1 -FD-Fc and Ang2-FD-Fc to bind to COS cells overexpressing TM was assessed. COS cells in 48-well plates were transfected with an expression vector containing TM, or with empty vector as a control. Two days after transfection, cells were incubated with 0, 0.16, 0.4, 1 , 2.56, 6.4, 16, 40 or 100 nM of Ang1 -FD-Fc (SEQ ID NO:5; residues 1 -15 correspond to the signal peptide) or Ang2-FD-Fc (SEQ ID NO:6; residues 1 -15 correspond to the signal peptide) in 200 μΙ binding buffer (PBS/10% calf serum) for 30 minutes at room temperature. After two PBS washes, cells were fixed in cold MeOH, followed by incubation with binding buffer containing alkaline phosphatase-conjugated secondary antibody (a 1 :1000 dilution of alkaline phosphatase-conjugated goat anti-human Fc (Promega)) for 30 min. After two more PBS washes, 200 μΙ PNPP substrate (Sigma) was added.

Reactions were stopped after 5 min by addition of 50 μΙ 3N NaOH, and the OD405 was taken as a measure of binding. Measurements were in quadruplicate. There was significant variability in these measurements, probably a consequence of avidity effects resulting from variable transfection efficiencies. Some nonspecific binding of Ang1 -FD-Fc to the empty vector- transfected cells was observed at high concentrations, but was less than 20% of binding to cells expressing TM.

[0062] Specific binding, given in OD405 units, is defined as the difference between binding to TM-transfected COS cells and binding to COS cells transfected with empty vector. Both Ang1 - FD-Fc and Ang2-FD-Fc, but not a control Fc protein, bound saturably to COS cells

overexpressing TM (Fig. 1 A), with apparent binding affinities of ~88nM for Ang1 -FD-Fc and ~8.3nM for Ang2-FD-Fc.

[0063] The ability of an Ang2-FD-Fc triple mutant that does not bind to Tie2 (Barton, W.A. et al., 2005, Structure, 13:825-832; SEQ ID NO:7; residues 1 -15 correspond to the signal peptide) to bind to COS cells overexpressing TM was assessed. In the Ang2-FD-Fc triple mutant, the mutated residues (F468A/Y474A/Y475A) are located at the Ang2/Tie2 interface (Barton, W.A., 2006, Nat. Struct. Mol. Biol. 13:524-532). In staining experiments (as described in Example 1 ), binding of the Ang2-FD-Fc triple mutant to COS cells overexpressing TM was not observed. This observation provides further support for the notion that residues at the angiopoietin/Tie2 binding interface are also required for binding to TM.

Example 3. BowAngl and BowAng2 bind soluble purified extracellular domain of TM

[0064] In this Example, the ability of the chimeric angiopoietins BowAngl (SEQ ID NO:2) and BowAng2 (SEQ ID NO:4) to associate with the purified soluble extracellular domain of TM was assessed.

[0065] The soluble extracellular domain of human TM (R&D Systems) or the soluble extracellular domain of human Tie2 (produced at Regeneron; SEQ ID NO:9) at 100nM were incubated with BowAngl or BowAng2 at 100, 250 or 500nM or with 500nM human Fc control protein in binding buffer (0.15M NaCI, 20mM Tris pH 7.5, 2mM CaCI 2 , 0.1 % Triton X-100, 1 mg/ml BSA) on ice for 60 minutes. Following incubation, BowAngl , BowAng2 or human Fc were pulled down by incubation with 25 μΙ protein A/G beads (Santa Cruz Biotechnology) for 60 minutes. The beads were then washed with binding buffer and proteins were eluted by heating in SDS sample buffer and then resolved on an SDS gel. Co-precipitation of TM and Tie2 was assessed by western blot with an anti-TM monoclonal antibody (Santa Cruz Biotechnology (D- 3), used at 1 :500 dilution) or a monoclonal anti-Tie2 antibody (clone 33.1 , Peters, K.G. et al., 1998, British Journal of Cancer, 77:51 -56).

[0066] As a positive control, binding of BowAngl and BowAng2 to the soluble extracellular domain of Tie2 was assessed. Both BowAngl and BowAng2, but not a control protein (human Fc), associated with TM, with estimated EC50 values in the hundreds of nM (Fig. 1 B).

Consistent with the cell surface binding assay, BowAng2 bound to TM significantly more tightly than did BowAngI (Fig. 1 B). Since soluble TM is monomeric, this assay provides a measure of monomer-monomer interactions between TM and one fibrinogen-like domain of BowAngI or BowAng2. Thus it is free of avidity effects inherent in the binding of dimeric forms of angiopoietins to cell surface TM (Fig. 1 A), so it is not unexpected that lower apparent affinities are observed. The relatively low apparent affinity of angiopoietins for monomeric Tie2 observed here (estimated EC50 values in the hundreds of nM) is consistent with the previously reported avidity effects that are characteristic of angiopoietin-Tie2 binding (Davis, 2003, supra).

[0067] Co-precipitation experiments failed to provide evidence for the existence of the formation of complexes on the surface of cells that include both Tie2 and TM (data not shown).

Example 4. Soluble Tie2 extracellular domain competes with TM for binding to BowAngI and BowAng2

[0068] In this Example, the ability of soluble Tie2 extracellular domain to compete with soluble TM extracellular domain for binding to BowAngI and BowAng2 was assessed.

[0069] 10OnM BowAngI or BowAng2 was incubated with soluble extracellular domain of human TM (R&D Systems; 100nM) in the absence or presence of excess (2μΜ) soluble extracellular domain of human Tie2 (produced at Regeneron) in binding buffer (0.15M NaCI, 20mM Tris pH 7.5, 2mM CaCI 2 , 0.1 % Triton X-100, 1 mg/ml BSA) on ice for 60 minutes.

Following incubation, BowAngI or BowAng2 were pulled down by incubation with 25 μΙ protein A/G beads (Santa Cruz Biotechnology) for 60 minutes. The beads were then washed with binding buffer and proteins were eluted by heating in SDS sample buffer and then resolved on an SDS gel. Co-precipitation of TM and Tie2 was assessed by western blot with an anti-TM monoclonal antibody (Santa Cruz Biotechnology (D-3), used at 1 :500 dilution) or a monoclonal anti-Tie2 antibody (clone 33.1 (Peters, supra).

[0070] As shown in Fig. 1 C, Tie2 competed with TM for binding to BowAngI and BowAng2, suggesting that the binding sites on the angiopoietins for TM and Tie2 do overlap. However, Tie2 was less effective at competing with TM for binding to BowAng2, consistent with the observation that BowAng2 appears to bind TM more tightly than does BowAngI , while binding less tightly than BowAngI to Tie2 in this solution binding assay (Fig. 1 B).

Example 5. Native Ang1 and Ang2 inhibit APC and TAFI formation in cultured HUVECs [0071 ] In this Example, the ability of angiopoietins to modulate the thrombin/TM dependent activation of protein C and TAFI was assessed. Addition of protein C or TAFI to cultured endothelial cells in the presence of thrombin results in cleavage of these substrates in a TM- dependent fashion (Hackeng, T.M. et al., 1996, The Biochemical journal. 319 (Pt 2):399-405; Hockin, M.F., et al., 1997, Arteriosclerosis, thrombosis, and vascular biology. 1997;17:2765- 2775; Slungaard, A. et al., 2003, Blood. 102:146-151 ). Several previous reports have used cultured endothelial cells to assess the effects of various proteins (e.g., the platelet a-granule protein platelet factor 4 (PF4)) on the activity of the thrombin/TM complex (Slungaard, supra).

[0072] APC formation on HUVEC monolayers was assessed using a modified version of a previously described method (Slungaard, supra). 50,000 HUVECs (VEC Technologies) were plated in 48-well plates in 200 μΙ of MCDB131 complete medium. The next day the cells were washed three times with reaction buffer (HBSS + 10mM HEPES pH 7.4 + 1 mg/ml BSA) and then incubated for 60 min at 37°C with 100 μΙ of reaction buffer containing 0.2μΜ protein C (Enzyme Research Labs), 0.1 U/ml thrombin (Sigma) and native human Ang1 (R&D Systems), native human Ang2 (R&D Systems), human PF4 (R&D Systems) or a control Fc-containing protein at 1 , 10, 50, 100, 500 or 1000 nM. Following incubation, cell supernatants (90 μΙ of buffer from each well) were transferred to a 96 well plate and incubated with the thrombin inhibitor hirudin (Sigma) at 25 U/ml for 10 minutes at 37°C. The chromogenic APC substrate S- 2366 (Chromogenix) was then added at 0.2mM and cleavage of S-2366 was followed by monitoring the change in OD 405 nm. The OD 405 nm at -10 minutes after S-2366 addition (the OD 405 nm was still increasing linearly at this time point) was used to assess the relative levels of TM-dependent APC formation in each sample (after subtracting the value observed when the procedure was carried out with no HUVECs in the wells).

[0073] The graph in Fig. 2A depicts the relative amounts of APC formation (compared to APC formation in the presence of the control protein) as a function of angiopoietin concentration. The error bars represent the SD, n = 3. Both Ang1 and Ang2 substantially inhibited APC formation (relative to the amount generated in the presence of 1 μΜ of a control protein), with IC 5 o values in the hundreds of nM (Fig. 2A). Consistent with its higher affinity for TM, Ang2 was a more potent inhibitor of APC production than was Ang1 (Fig. 2A).

[0074] As an additional control for the specificity of the assay, the effect of PF4 on APC formation in HUVECs was assessed. HUVECs were incubated with protein C and thrombin plus either a control protein (0.5μΜ), native Ang1 (0.5μΜ) or PF4 (10 μg/ml, ~1 .3μΜ) for 60 minutes and cell supernatants were then assayed for APC levels. The graph in Fig. 2B depicts the relative amounts of APC generated (the amount of APC generated in the presence of the control protein was assigned a value of 1 .0). The error bars represent the SD, n = 3. Relative levels of APC in the Ang1 and the PF4 treatment groups were significantly different from the control group ( *** , P < 0.001 , one-way ANOVA with Tukey's multiple comparisons test). In contrast to angiopoietins, addition of PF4 to the HUVECs modestly increased APC formation (Fig. 2B), as demonstrated previously (Slungaard, supra).

[0075] The ability of angiopoietins to affect aTAFI formation in HUVECs was assessed. 50,000 HUVECs (VEC Technologies) were plated in 48-well plates in 200 μΙ of MCDB131 complete medium. The next day the cells were washed three times with reaction buffer (HBSS + 10mM HEPES pH 7.4 + 1 mg/ml BSA) and then incubated at 37°C for 60 minutes with 100 μΙ of reaction buffer containing 0.2μΜ TAFI (Enzyme Research Labs), 1 U/ml thrombin, and native human Ang1 (1 nM or 100 nM), native human Ang2 (1 nM or 100 nM) or a control Fc-containing protein (1 μΜ). Following incubation, cell supernatants were used to measure aTAFI activity using the fluorogenic ACTIFLUOR TAFIa Activity Assay (American Diagnostica).

[0076] The graph in Fig. 2C depicts the relative amounts of aTAFI generated (the amount of aTAFI generated in the presence of the control protein was assigned a value of 1 .0). The error bars represent the SD, n = 3. Relative levels of aTAFI in the Ang1 10OnM and the Ang2 10OnM treatment groups were significantly different from the control group ( *** , P < 0.001 , one-way ANOVA with Tukey's multiple comparisons test). Similar to their effect on formation of APC, native Ang1 and Ang2 at 100 nM significantly inhibited aTAFI formation in HUVECs (Fig. 2C).

[0077] These experiments indicate that in addition to binding TM, native Ang1 and Ang2 inhibit TM function in endothelial cells.

Example 6. Ang1 and Ang2 inhibit both APC and aTAFI formation in COS cells overexpressing TM but lacking Tie2 expression (inhibition does not require Tie2)

[0078] In this Example, the ability of Ang1 and Ang2 to inhibit APC and aTAFI formation in COS cells overexpressing TM but lacking Tie2 expression was assessed.

[0079] COS7 cells transfected with expression vectors for TM or control empty vector were seeded, one day after transfection, into 48 well plates (~80,000/well). The next day, cells were washed three times with reaction buffer (HBSS + 10 mM HEPES + 1 mg/ml BSA) and then incubated with 100 μΙ of reaction buffer containing 1 , 3, 9, 27, 81 , 243, or 729 nM of native human Ang1 or Ang2 for 30 minutes at 4°C. 0.2 μΜ protein C and 0.1 U/ml thrombin were then added and incubated for 60 minutes at 37°C. Following incubation, the reaction was stopped with hirudin and APC levels were determined, as above.

[0080] Following incubation, 90 μΙ of buffer from each well was transferred to a 96 well plate and incubated with the thrombin inhibitor hirudin (Sigma) at 25 U/ml for 10 minutes at 37°C. The chromogenic APC substrate S-2366 (Chromogenix) was then added at 0.2mM and cleavage of S-2366 was followed by monitoring the change in OD 405 nm. The OD 405 nm at -10 minutes after S-2366 addition (the OD 405 nm was still increasing linearly at this time point) was used to assess the relative levels of TM-dependent APC formation in each sample (after subtracting the value observed when the procedure was carried out with no HUVECs in the wells). The graph in Fig. 2D depicts relative APC formation as a function of angiopoietin concentration (the amount formed in the absence of angiopoietin was assigned a value of 1 .0)

[0081 ] The ability of angiopoietins to affect aTAFI formation in COS cells was assessed. COS cells transfected with expression vectors for TM or control empty vector in 10cm dishes were rinsed in PBS, collected using a spatula, disaggregated by trituration, and pelleted. Cells were resuspended in reaction buffer (HBSS + 10mM HEPES pH7.4 + 1 mg/ml BSA, -80,000 cells/reaction) and incubated with 72 μΙ of reaction buffer containing various concentrations of native human Ang1 or Ang2 (1 , 3, 9, 27, 81 , 243, or 729 nM) for 30 minutes at 4°C. Following this, 0.2 μΜ TAFI and 1 U/ml thrombin were added in a final volume of 80 μΙ and incubated for 30 minutes at 22°C. Cells were then pelleted and 75 μΙ of the supernatants were transferred to a 96 well plate. 25 μΙ of 2mM 5,5'-dithiobis-(2-nitrobenzoic acid) (Sigma) and 50 μΙ of a thiol- releasing TAFI substrate (R2 reagent, Pefakit TAFI, Pentapharm) were then added, and the OD 405 nm at 10 minutes was used to determine TAFI activity. The graph in Fig. 2E depicts aTAFI formation relative to the amount generated in the absence of angiopoietin.

[0082] In COS cells overexpressing TM, but lacking Tie2 expression, angiopoietins significantly inhibited both APC and aTAFI formation, indicating that the inhibition does not require Tie2 (Fig. 2D and 2E). In contrast to their effect in HUVECs, angiopoietin-mediated inhibition in COS cells was observed even at low nM concentrations, presumably reflecting enhanced, avidity-driven angiopoietin binding to overexpressed TM. A possible explanation for this observation is that in HUVECs, the endothelial Protein C receptor (EPCR) makes blockade of APC formation more difficult than in COS cells. EPCR is expressed at high levels in HUVECs and binds Protein C with high affinity (40), promoting delivery of protein C to the thrombin/TM complex, thus amplifying its activity (41 ). In this context, it is interesting to note that, in contrast to HUVECs, EPCR is expressed at low levels in microvasculature (Laszik Z., et al., 1997, Circulation. 96:3633-3640).

Example 7. Inhibition of APC formation by Ang1 is independent of Tie 2

[0083] In this Example, the effect of knocking down Tie2 expression on Ang1 -dependent inhibition of APC-formation in HUVECs was assessed. HUVECs were infected with

adenoviruses (~ 30 pfu/cell) encoding either a non-silencing control shRNA or a Tie2 specific shRNA as described (Daly, C. et al., 2004, Genes & Development. 18:1060-1071 .

[0084] The sequence of the Tie2 shRNA was: 5'- TG AAGTACCTG ATATTCTA-3 ' (SEQ ID NO:8) (nucleotides 946-964 in the human Tie2 (TEK) cDNA, NM_000459). At 3 days after infection, cells were either lysed for Western blot to demonstrate Tie2 knockdown (Fig. 3A) or used to test the effect of native Ang1 on APC formation in the presence of protein C and thrombin (Fig. 3B). The APC assay was done as described in Example 5 above (except that thrombin was present at 1 U/ml), in the presence of 1 μΜ control protein or Ang1 . Western blot for Tie2 was performed with the monoclonal antibody clone 33.1 (Peters, supra).

[0085] The graph in Fig. 3B depicts the relative amounts of APC generated in the presence of 1 μΜ control protein or native Ang1 (the amount of APC generated in the presence of the control protein was assigned a value of 1 .0). The error bars represent the SD, n = 3. Ang1 significantly inhibited APC formation in cells treated with control shRNA or Tie2 shRNA ( *** , P < 0.001 , oneway ANOVA with Tukey's multiple comparisons test).

[0086] Despite almost complete inhibition of Tie2 expression by the shRNA (Fig. 3A), Ang1 was still capable of inhibiting APC formation (Fig. 3B), indicating that Tie2 is not required for this effect.

Example 8. Ang1 and Ang2 inhibit binding of thrombin to TM

[0087] In this Example, the ability of Ang1 and Ang2 to inhibit binding of thrombin to TM was assessed in vitro. Soluble TM extracellular domain (with a 6-His tag) at 100nM was incubated with 5nM thrombin (Sigma) in the presence of 1 μΜ of an Fc-containing control protein or 0.25, 0.5 or 1 .ΟμΜ native Ang1 (Fig. 4A) or native Ang2 (Fig. 4B) in binding buffer (0.15M NaCI, 20mM Tris pH 7.5, 0.1 % Triton X-100, 2mM CaCI 2 , 1 mg/ml BSA) for 60 minutes on ice. TM was then pulled down by incubation for 60 minutes with 20 μΙ Ni-NTA beads (Thermo Scientific). Beads were then washed with binding buffer and bound proteins were eluted by heating in SDS sample buffer. Western blots were then performed to assess pull-down of TM (monoclonal antibody D-3 from Santa Cruz Biotechnology) and co-precipitation of thrombin (goat polyclonal antibody from R&D Systems).

[0088] Increasing concentrations of native Ang1 or Ang2 progressively inhibited the association of thrombin with soluble TM in vitro, with Ang2 having a more potent effect (Fig. 4A, 4B). This result suggests that angiopoietins can inhibit formation of a stable complex of thrombin with TM, and that this is one mechanism by which angiopoietins interfere with generation of APC and aTAFI.

Example 9. Ang1 is present in mouse platelets

[0089] In this Example, the presence of Ang1 in mouse platelets was assessed. It has been reported that human platelets contain Ang1 and release it upon stimulation with thrombin in vitro (Li, J.J. et al., 2001 , Thromb. Haemost. 85:204-206).

[0090] All mouse experiments were approved by Regeneron's Institutional Animal Care and Use Committee. Blood was collected from C57BI/6 mice by cardiac puncture and expelled into tubes containing EDTA and immediately mixed. Blood was then centrifuged at 300 x g for 10 minutes. The supernatant (platelet-rich plasma) was collected and spun at 2,000 x g for 10 minutes and the pellet was then resuspended directly in SDS sample buffer and heated.

Aliquots of platelet lysate representing ~1 .6 x 10 6 platelets were run on SDS gels along with the indicated amounts (ng) of mouse PF4, human Ang1 or human Ang2 to allow estimation of the levels of these proteins in the platelet lysate. Ang1 was detected with a rabbit polyclonal antibody that recognizes both mouse and human Ang1 (mouse and human Ang1 proteins are 97% identical). Ang2 was detected with a goat polyclonal antibody raised against a peptide that is identical in the human and mouse proteins.

[0091 ] Aliquots of platelet lysate were then used in western blots, probing for PF4 (goat polyclonal from R&D Systems raised against mouse PF4, used at 1 :5000 dilution), Ang1 (rabbit polyclonal generated at QCB for Regeneron, used at 1 μg/ml) or Ang2 (goat polyclonal (C-19) from Santa Cruz Biotechnology, used at 1 :200 dilution). The Ang1 and Ang2 antibodies recognize both the mouse and human proteins.

[0092] As shown in Fig. 5A, Ang1 protein was detected in mouse platelets at levels

(~30ng/10 6 platelets) similar to those reported in human platelets (~55ng/10 6 platelets) (46). As a positive control, the presence of PF4 was detected in platelet lysate at ~3ng/10 6 platelets, similar to the estimated level of PF4 in human platelets (~12ng/10 6 platelets) (Peterson J.E., et al., 2010, American journal of hematology, 85:487-493). In contrast, Ang2 was not detected in mouse platelets (Fig. 5A). The polyclonal antibody used to detect Ang2 was raised against a peptide that is identical in human/mouse Ang2 and this antibody readily detects Ang2 in mouse tissue lysates by western blot. While Ang2 is not observed in platelets, it is present in endothelial cell Weibel-Palade bodies (WPB) and is released upon stimulation with thrombin in vitro (Fiedler, U. et al, 2004, Blood, 103:4150-4156), suggesting the possibility that Ang2 is released from activated endothelial cells at sites of vessel injury. Ang1 is present in mouse platelets (Fig. 5A) but Ang2 is not detected in mouse platelets (Ang2 has been shown in art to be found in endothelial cell Weibel-Palade bodies).

Example 10. Ang1 is 100X higher in serum than in plasma

[0093] In this Example, the level of Ang1 was measured in mouse serum and mouse plasma.

[0094] For serum preparation, blood was collected by cardiac puncture and expelled into serum separator tubes (BD Biosciences). Following a 30 minute incubation to allow for blood clotting, samples were centrifuged at 2,000 x g for 10 minutes and the serum layer was collected and stored at -80 e C.

[0095] For plasma preparation, blood was collected by cardiac puncture, expelled into EDTA tubes (BD Biosciences), mixed and then centrifuged at 2,000 x g at 4 e C for 10 minutes. The supernatant (platelet-poor plasma) was collected and centrifuged again at 2,000 x g for 10 minutes. The plasma supernatant was collected and either used immediately or stored in aliquots at -80 e C.

[0096] Blood was collected from C57BL/6 mice by cardiac puncture and used to prepare either plasma or serum. Ang1 and PF4 levels were then determined by ELISA.

[0097] Ang1 ELISA: 96 well ELISA plates (Corning) were coated overnight at 4°C with 10 μg/ml of His-tagged Tie2 extracellular domain. Wells were washed 3 times with PBS + 0.05% Tween 20 (PBST) and blocked for 2 hours at RT with PBS + 3% BSA. Wells were washed again and incubated for 1 hour with 100 μΙ of protein standard (recombinant human Ang1 from R&D Systems) or plasma/serum samples diluted in PBST + 3% BSA. Plasma samples were diluted 1 :5, serum samples were diluted 1 :20. Wells were then washed and incubated at RT for 1 hour with 100 μΙ of biotinylated goat anti-Angl antibody (R&D Systems, catalog number BAF923) at 0.2 μg/ml. Wells were then washed and incubated in 100 μΙ of streptavidin-HRP (R&D Systems) at 1 :200 dilution for 20 minutes at RT. Wells were then washed and incubated for 20 minutes at RT with 100 μΙ of substrate solution (R&D Systems, catalog number DY999) protected from light. 50 μΙ of 1 M sulfuric acid was added to stop the reactions and OD values were read at 450 nm with subtraction at 540 nm.

[0098] PF4 ELISA was performed according to the protocol outlined in the mouse

CXCL4/PF4 DuoSet (R&D Systems, catalog number DY595). Plasma samples were diluted 1 :10 or 1 :50 and serum samples were diluted 1 :2000.

[0099] Ang1 levels are given as apparent concentrations since the standard curve used to estimate Ang1 concentration was generated with human Ang1 protein. Bars represent the mean and SD, n = 4. Both Ang1 and PF4 levels were significantly higher in serum ( ** , P < 0.01 ;

P < 0.001 , f-test). Consistent with Ang1 release from platelets upon degranulation ex vivo, Ang1 levels were ~100-fold higher in serum than in plasma (Fig. 5B). Similarly, PF4 levels were hundreds of fold higher in serum, providing a positive control (Fig. 5B). Due to the lack of availability of a reliable mouse Ang2 ELISA, we were not able to compare Ang2 levels in mouse serum versus plasma. However, consistent with our data showing the absence of Ang2 in platelets, Ang2 levels in plasma and serum from healthy volunteers are similar (< 5 ng/ml) (Goede, V., et al., 2010 British Journal of Cancer, 103:1407-1414; Wang, X., et al., 2014, Translational Oncology, 7:188-195).

Example 11 : Ang1 is released from activated platelets in vivo

[0100] To determine whether Ang1 is released from activated platelets in vivo, plasma levels of Ang1 (and PF4 as a positive control) were measured 10 minutes after intravenous

administration of thrombin to mice.

[0101 ] C57BL/6 mice were injected into the tail vein with PBS or 10 units of thrombin (Sigma) in a volume of 100 μΙ. At 10 minutes after injection, blood was drawn by cardiac puncture and plasma was prepared for use in Ang1 and PF4 ELISAs as in Example 10. Plasma levels of Ang1 and PF4 were determined by ELISA. Bars represent the mean and SD (n = 9 for PBS, n = 8 for thrombin). The levels of both Ang1 and PF4 were significantly higher in the thrombin treated samples ( ** , P < 0.01 ; P < 0.001 , f-test). Any bloods that were difficult to draw were discarded and plasma samples in which the Ang1 level was > 30 ng/ml were not used in the analysis, as these samples appeared to be outliers and likely had some degree of platelet activation and Ang1 release ex vivo.

[0102] As shown in Fig. 5C, plasma levels of both Ang1 and PF4 were significantly higher following acute thrombin treatment, consistent with release of both of these factors from activated platelets. Ang1 plasma level is higher following acute thrombin treatment (consistent with Ang1 being released from activated platelets) (Fig. 5C).

Example 12: Ang1 is released from platelets in response to blood vessel injury

[0103] To assess whether Ang1 is released from platelets in response to blood vessel injury, Ang1 levels were measured in plasma prepared from blood collected at the site of tail nicks in mice.

[0104] Blood was collected from C57BL/6 mice either by cardiac puncture (systemic) (see Example 9) or by tail nick. For tail nick experiments to assess the levels of Ang1 in wound blood, C57BL/6 mice were placed under a heating lamp for approximately 5 min. Mice were then placed in a restrainer and the distal 2 to 4 mm of tail was severed with a single diagonal cut. Blood was collected into heparinized capillary tubes for an initial period of 30 seconds and discarded. Blood was then collected into a second heparinized capillary tube until the tube was full (this took 2-5 minutes). A disposable Pasteur pipette was used to expel the blood into a microfuge tube containing EDTA. Plasma was then prepared as described above. For this experiment, the samples used to assess systemic plasma Ang1 levels were collected by cardiac puncture of na ' ive animals. To mimic the procedure for preparation of plasma from wound blood, the blood was stored in heparinized capillary tubes for 4-5 min before being expelled into a microfuge tube containing EDTA. To ensure that the higher levels of Ang1 in the tail nick samples did not simply reflect differences in the plasma preparation procedures, the cardiac puncture blood samples were incubated in heparinized capillary tubes for 4-5 minutes before proceeding with plasma isolation to mimic the procedure for the tail nick samples. Plasma was prepared and Ang1 and PF4 levels were determined by ELISA. Plasma was prepared and Ang1 and PF4 levels were determined by ELISA as in Example 10. Bars represent the mean and SD (n = 9 for systemic samples, n = 10 for tail nick samples). The levels of both Ang1 and PF4 were significantly higher in the tail nick samples ( *** , P < 0.001 , f-test).

[0105] Both Ang1 and PF4 levels were significantly higher in plasma prepared from tail nick blood than in plasma prepared from systemic blood (collected via cardiac puncture) (Fig. 5D), suggesting release of Ang1 from platelets at the site of injury. Ang1 level was higher in plasma prepared from tail nick blood than in plasma prepared from systemic blood, suggesting release of Ang1 from platelets at the site of injury (Fig. 5D).

Example 13. Intravenous administration of BowAngl and BowAng2 to mice promotes a rapid increase in circulating thrombin/antithrombin level and fibrin deposition in lung [0106] In this Example, the effect of BowAngl and BowAng2 on circulating

thrombin/antithrombin level and on fibrin deposition in lung was assessed.

[0107] At 20 minutes following intravenous injection of 6 mg/kg of Fc control, BowAngl or BowAng2, lungs were harvested. Lungs were homogenized in ice-cold PBS containing complete™ protease inhibitor cocktail (with EDTA) plus 10 units/ml heparin. Following overnight incubation at 4°C, lysates were spun for 10 minutes in a microcentrifuge. The pellets were resuspended in 3M urea and incubated at 37°C for 2 hours to extract cross-linked fibrin.

Samples were then vortexed and spun for 10 minutes in a microcentrifuge. Pellets were resuspended in SDS-PAGE sample buffer and incubated at 65°C for 90 minutes. The amount of fibrin in the samples was determined by Western blot using a monoclonal anti-fibrin antibody from American Diagnostica (Fig. 6A).

[0108] At 20 minutes following intravenous injection of Fc control, BowAngl (7.5 mg/kg) or BowAng2 (4 mg/kg), the levels of thrombin/antithrombin III complex in plasma were measured using a sandwich enzyme immunoassay kit (Enzygnost® TAT micro, Dade Behring) (Fig. 6B).

[0109] The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims.