Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATMENT OF GASTROINTESTINAL BLEEDING IN PATIENTS WITH SEVERE VON WILLEBRAND DISEASE BY ADMINISTRATION OF RECOMBINANT VWF
Document Type and Number:
WIPO Patent Application WO/2019/010497
Kind Code:
A1
Abstract:
The present invention relates to a method for treating gastrointestinal bleeding in a subject with severe von Willebrand Disease comprising administering to the subject at least one dose of recombinant von Willebrand Factor (rVWF) ranging from about 40 IU/kg to about 100 IU/kg, wherein the first dose further comprises recombinant Factor VIII (rFVIII).

Inventors:
CHAPMAN MIRANDA (US)
EWENSTEIN BRUCE (US)
PLODER BETTINA (US)
Application Number:
PCT/US2018/041320
Publication Date:
January 10, 2019
Filing Date:
July 09, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BAXALTA INC (US)
BAXALTA GMBH (CH)
CHAPMAN MIRANDA (US)
EWENSTEIN BRUCE (US)
PLODER BETTINA (US)
International Classes:
A61K38/36; A61K38/37; A61P7/04
Domestic Patent References:
WO2012171031A12012-12-13
WO1986006096A11986-10-23
WO1986006096A11986-10-23
WO2004039337A22004-05-13
Foreign References:
US55950990A1990-07-23
US4640835A1987-02-03
US4496689A1985-01-29
US4301144A1981-11-17
US4670417A1987-06-02
US4791192A1988-12-13
US4179337A1979-12-18
US20080009040A12008-01-10
US20070212770A12007-09-13
US20060094104A12006-05-04
US8852888B22014-10-07
US9409971B22016-08-09
US8597910B12013-12-03
US20160129090A12016-05-12
US3941763A1976-03-02
US20060160948A12006-07-20
US7005502B12006-02-28
Other References:
JOAN C. GILL ET AL: "Hemostatic efficacy, safety, and pharmacokinetics of a recombinant von Willebrand factor in severe von Willebrand disease", BLOOD, 22 October 2015 (2015-10-22), pages 2038 - 2046, XP055510258, Retrieved from the Internet [retrieved on 20180926]
PIER MANNUCCIO MANNUCCI ET AL: "Pharmacokinetics and safety of a novel recombinant human von willlebrand factor manufactured with a plasma-free method: a prospective clinical trial", BLOOD, 18 June 2013 (2013-06-18), XP055510217, Retrieved from the Internet [retrieved on 20180926]
M. SINGAL ET AL: "Recombinant von Willebrand Factor: a first-of-its kind product for von Willebrand disease", DRUGS OF TODAY, 1 December 2016 (2016-12-01), pages 653 - 664, XP055510191, Retrieved from the Internet [retrieved on 20180926]
MASSIMO FRANCHINI ET AL: "Gastrointestinal angiodysplasia and bleeding in von Willebrand disease", THROMBOSIS AND HAEMOSTASIS, vol. 112, no. 09, 1 January 2014 (2014-01-01), DE, pages 427 - 431, XP055509883, ISSN: 0340-6245, DOI: 10.1160/TH13-11-0952
MANNUCCI PM; FRANCHINI M., HAEMOPHILIA, vol. 23, no. 2, 2017, pages 182 - 187
TURECEK PL ET AL., HAMOSTASEOLOGIE, vol. 29, no. 1, 2009, pages S32 - 38
MANNUCCI PM ET AL., BLOOD, vol. 122, no. 5, 2013, pages 648 - 657
GILL JC ET AL., BLOOD, vol. 126, no. 17, 2015, pages 2038 - 2046
TURECEK ET AL., SEMIN. THROMB. HEMOST., vol. 36, no. 5, 2010, pages 510 - 521
ANDERSSON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 83, 1986, pages 2979 - 2983
LIEBERMAN, PHARMACEUTICAL DOSAGE FORMS, vol. 1-3, 1992
LLOYD, THE ART, SCIENCE AND TECHNOLOGY OF PHARMACEUTICAL COMPOUNDING, 1999
PICKAR: "Dosage Calculations", 1999
GENNARO,: "Remington: The Science and Practice of Pharmacy, 20th Edition,", 2003, LIPPINCOTT, WILLIAMS & WILKINS
TURECEK ET AL., SEMIN. THROMB. HEMOST., vol. 28, 2002, pages 149 - 160
WEISS ET AL., J. CLIN. INVEST., vol. 52, 1973, pages 2708 - 2716
MACFARLANE ET AL., THROMB. DIATH. HAEMORRH., vol. 34, 1975, pages 306 - 308
BROWN; BOSAK, THROMB. RES., vol. 43, 1986, pages 303 - 311
FAVALORO, THROMB. HAEMOST., vol. 83, 2000, pages 127 - 135
CUMMING ET AL., J CLIN PATHOL., vol. 46, no. 5, May 1993 (1993-05-01), pages 470 - 473
WEN ET AL., J. CLIN. LAB. ANAL., vol. 7, 1993, pages 317 - 323
FAVALORO ET AL., PATHOLOGY, vol. 29, no. 4, 1997, pages 341 - 456
ABUCHOWSKI; DAVIS: "Enzymes as Drugs", 1981, JOHN WILEY AND SONS, pages: 367 383
HERMANSON G.: "Bioconjugate Techniques 2nd Ed.,", 2008, ACADEMIC PRESS, INC.
JENNINGS; LUGOWSKI, J. IMMUNOL., vol. 127, 1981, pages 1011 - 8
FEMANDES; GREGONRADIS, BIOCHIM BIOPHYS ACTA, vol. 1341, 1997, pages 26 - 34
VAN WEZEL, A. L., NATURE, vol. 216, 1967, pages 64 - 5
BUTLER, SPIER & GRIFFITHS, ANIMAL CELL BIOTECHNOLOGY, vol. 3, 1988, pages 283 - 303
TANG ET AL., PHARM RES., vol. 21, 2004, pages 191 - 200
CHANG ET AL., PHARM RES., vol. 13, 1996, pages 243 - 9
CHEN, DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, vol. 18, 1992, pages 1311 - 1354
LANKHOF ET AL., THROMB. HAEMOST., vol. 77, 1997, pages 1008 - 1013
PIETU ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 164, 1989, pages 1339 - 1347
MERRIFIELD: "Chem. Polypeptides", 1973, pages: 335 - 61
MERRIFIELD, J. AM. CHEM. SOC., vol. 85, 1963, pages 2149
DAVIS ET AL., BIOCHEM. INTL., vol. 10, 1985, pages 394 - 414
FINN ET AL.: "The Proteins (3rd ed.)", vol. 2, 1976, pages: 105 - 253
ERICKSON ET AL.: "The Proteins (3rd ed.)", vol. 2, 1976, pages: 257 - 527
FERNANDES; GREGORIADIS, BIOCHIM. BIOPHYS. ACTA, vol. 1341, 1997, pages 26 - 34
SAENKO ET AL., HAEMOPHILIA, vol. 12, 2006, pages 42 - 51
MIGNEAULT ET AL., BIOTECHNIQUES, vol. 37, 2004, pages 790 - 796
SAMBROOK ET AL.: "Molecular Cloning", 1989, COLD SPRING HARBOR LABORATORY PRESS, pages: 9.47 - 9.51
SADLER, JE, ANNU REV BIOCHEM, vol. 67, 1998, pages 395 - 424
TURECEK ET AL., SEMIN THROMB HEMOST, vol. 36, 2010, pages 510 - 521
TURECEK, P., HIJMOSTASEOLOGIE, vol. 37, no. 1, 2017, pages S15 - S25
"Remington's Pharmaceutical Sciences, 18th Ed.,", vol. 18042, 1990, MACK PUBLISHING CO., pages: 1435 - 1712
STRYER, L.: "Biochemistry (4th Ed.)", 1995, FREEMAN
GAIT: "Oligonucleotide Synthesis: A Practical Approach", 1984, IRL PRESS
NELSON; COX: "Lehninger, Principles of Biochemistry 3rd Ed.,", 2000, W. H. FREEMAN PUB.
BERG ET AL.: "Biochemistry, 5th Ed.,", 2002, W. H. FREEMAN PUB.
CHANG ET AL., PHARMRES., vol. 13, 1996, pages 243 - 9
A. P. MACKENZIE, PHIL TRANS R SOC LONDON, SER B, BIOL, vol. 278, 1977, pages 167
POWELL ET AL.: "Compendium of Excipients fir Parenteral Formulations", PDA J. PHARM. SCI. TECHNOLOGY, vol. 52, 1998, pages 238 - 311, XP009119027
REMMELE R.L. JR. ET AL., BIOCHEMISTRY, vol. 38, no. 16, 1999, pages 5241 - 7
LAURSEN T ET AL., BASIC CLIN PHARMACOL TOXICOL., vol. 98, no. 2, 2006, pages 218 - 21
HOLLANDER-RODRIGUEZ JC ET AL., AM. FAM. PHYSICIAN., vol. 73, no. 2, 2006, pages 283 - 90
CARPENTER ET AL., DEVELOP. BIOL. STANDARD, vol. 74, 1991, pages 225
CHANG, B, J. PHARM. SCI., vol. 85, 1996, pages 1325
CHEN B ET AL., PHARM RES., vol. 20, no. 12, 2003, pages 1952 - 60
TOMITA M ET AL., BIOCHEMISTRY, vol. 8, no. 12, 1969, pages 5149 - 60
LAM XM ET AL., J PHARM SCL, vol. 86, no. 11, 1997, pages 1250 - 5
AKERS MJ; DEFELIPPIS MR: "Peptides and Proteins as Parenteral Solutions. In: Pharmaceutical Formulation Development of Peptides and Proteins", 1999, PHARMACEUTICAL SCIENCE. TAYLOR AND FRANCIS
FRANSSON J.R., PHARM. SCI., vol. 86, no. 9, 1997, pages 4046 - 1050
YIN J ET AL., PHARM RES., vol. 21, no. 12, 2004, pages 2377 - 83
LAM XM; YANG JY ET AL., J PHARM SCI., vol. 86, no. 11, 1997, pages 1250 - 5
CHEN B ET AL., PHARM SCI., vol. 88, no. 4, 1999, pages 477 - 82
FATOUROS, A. ET AL., INT. J. PHARM., vol. 155, 1997, pages 121 - 131
DERRICK TS ET AL., PHARM. SCI., vol. 93, no. 10, 2004, pages 2549 - 57
ROY S ET AL., J PHARM SCL, vol. 94, no. 2, 2005, pages 382 - 96
REMMELE RL JR. ET AL., PHARM RES., vol. 15, no. 2, 1998, pages 200 - 8
KAPPELGAARD A.M., HORM RES., vol. 62, no. 3, 2004, pages 98 - 103
MINOGUE SC; SUN DA., ANESTHANALG., vol. 100, no. 3, 2005, pages 683 - 6
WELLS G; COYLE D; CAMERON C ET AL.: "Safety, Effectiveness, and Cost-Effectiveness of New Oral Anticoagulants Compared with Warfarin in Preventing Stroke and Other Cardiovascular Events in Patients with Atrial Fibrillation [Internet", CANADIAN AGENCY FOR DRUGS AND TECHNOLOGIES IN HEALTH, 9 April 2012 (2012-04-09), pages 3
SOBIERAJ DM; COLEMAN CI; TONGBRAM V ET AL.: "Venous Thromboembolism Prophylaxis in Orthopedic Surgery [Internet", AGENCY FOR HEALTHCARE RESEARCH AND QUALITY (US, March 2012 (2012-03-01)
WELLS G; COYLE D; CAMERON C ET AL.: "Safety, Effectiveness, and Cost-Effectiveness of New Oral Anticoagulants Compared with Warfarin in Preventing Stroke and Other Cardiovascular Events in Patients with Atrial Fibrillation [Intemet", CANADIAN AGENCY FOR DRUGS AND TECHNOLOGIES IN HEALTH, 9 April 2012 (2012-04-09), pages 3
"Remington's Pharmaceutical Sciences, 18th Ed.,", 1990, MACK PUBLISHING CO., pages: 1435 - 1712
RANDI AM., THROMB RES., vol. 141, no. 2, 2016, pages 55 - 58
RANDI AM; LAFFAN MA, J THROMB HAEMOST., vol. 15, no. 1, 2017, pages 13 - 20
FRANCHINI M; MANNUCCI PM, THROMB HAEMOST., vol. 112, no. 3, 2014, pages 427 - 431
FRANCHINI M; MANNUCCI PM., BR JHAEMATOL., vol. 161, no. 2, 2013, pages 177 - 182
BERNTORP E ET AL., HAEMOPHILIA, vol. 15, no. 1, 2009, pages 122 - 130
TURECEK PL ET AL., HAMOSTASEOLOGIE, vol. 29, no. 1, 2009, pages S32 - 3 8
SELVAM S; JAMES P, SEMIN THROMB HEMOST., 2017
FRANCHINI M; MANNUCCI PM, EXPERT REV HEMATOL., vol. 9, no. 9, 2016, pages 825 - 830
Attorney, Agent or Firm:
MACDOUGALL, Christina, A. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method for treating gastrointestinal bleeding in a subject with severe von Willebrand Disease (VWD) comprising administering to the subject at least one dose of recombinant von Willebrand Factor (rVWF) ranging from about 40 IU/kg to about 100 IU/kg, wherein the first dose further comprises recombinant Factor VIII (rFVIII).

2. The method of treatment of claim 1, wherein said rFVIII is administered at a dose of about 20 IU/kg to about 50 IU/kg.

3. The method of treatment of claim 1, wherein said method further comprises administering to the subject a second dose of recombinant von Willebrand Factor (rVWF) ranging from about 40 IU/kg to about 100 IU/kg, wherein the second dose does not comprise recombinant Factor VIII (rFVIII).

4. The method of treatment of claim 1, wherein said rVWF to FVIII ratio is about 1.5:0.8.

5. The method of treatment of claim 1, wherein said rVWF to FVIII ratio is about 1.3: 1.

6. The method of treatment of claim 1, wherein said rVWF to FVIII ratio is about 1.1 :0.8.

7. The method of treatment of claim 1, wherein said rVWF to FVIII ratio is about 1.5: 1.

8. The method of treatment of claim 1, wherein said rVWF to FVIII ratio is about 1.1 : 1.2.

9. The method of treatment of claim 1, wherein said rVWF is administered every 8 to 12 hours. Atl

10. The method of treatment of any one of claims 1 to 9, wherein 40-60 IU/kg rVWF of said rVWF is administered and wherein said gastrointestinal bleeding is minor or moderate gastrointestinal bleeding.

11. The method of treatment of any one of claims 1 to 9, wherein 40-80 IU/kg rVWF of said rVWF and wherein said gastrointestinal bleeding is major or severe gastrointestinal bleeding.

12. The method of treatment of any one of claims 1 to 9, wherein said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days.

13. The method of treatment of any one of claims 1 to 9, wherein 40-60 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, wherein said gastrointestinal bleeding is minor or moderate gastrointestinal bleeding.

14. The method of treatment of any one of claims 1 to 9, wherein 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, wherein said gastrointestinal bleeding is major or severe gastrointestinal bleeding.

15. The method of treatment of any one of claims 1 to 14, wherein said subject has Type 3 VWD.

16. The method of treatment of any one of claims 1 to 14, wherein said subject has severe type 1 VWD.

17. The method of treatment of any one of claims 1 to 14, wherein said subject has severe type 2 VWD.

18. The method of any one of claims 1 to 17, wherein the subject had been treated for at least 1 bleeding event within the previous 12 months.

19. The method of any one of claims 1 to 17, wherein the subject had been treated for more than 1 bleeding event within the previous 12 months.

Description:
TREATMENT OF GASTROINTESTINAL BLEEDING IN PATIENTS WITH SEVERE VON WILLEBRAND DISEASE BY ADMINISTRATION OF

RECOMBINANT VWF

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority to U.S. Provisional Patent Application No.

62/530,027, filed on July 7, 2017, which is hereby incorporated by reference in its entirety.

BACKGROUND OF THE INVENTION

[0002] Coagulation diseases, such as von Willebrand Disease (VWD) generally result from a deficiency in the coagulation cascade, von Willebrand Disease (VWD) refers to the group of diseases caused by a deficiency of von Willebrand factor. Von Willebrand factor helps blood platelets clump together and stick to the blood vessel wall, which is necessary for normal blood clotting.

[0003] von Willebrand disease (VWD) is the most common inherited bleeding disorder, with an estimated prevalence rate of 1% (Veyradier A, et al, Medicine (Baltimore). 2016, 95(l l):e3038). However, excluding milder forms of the disease, only about 1/10,000 patients actually require treatment. Current treatment for these coagulopathies includes a replacement therapy using pharmaceutical preparations comprising the normal coagulation factor.

[0004] VWF is a glycoprotein circulating in plasma as a series of multimers ranging in size from about 500 to 20,000 kD. The full length of cDNA of VWF has been cloned; the propolypeptide corresponds to amino acid residues 23 to 764 of the full length prepro-VWF (Eikenboom et al (1995) Haemophilia 1, 77 90). Multimeric forms of VWF are composed of 250 kD polypeptide subunits linked together by disulfide bonds. VWF mediates the initial platelet adhesion to the sub-endothelium of the damaged vessel wall, with the larger multimers exhibiting enhanced hemostatic activity. Multimerized VWF binds to the platelet surface glycoprotein Gplba, through an interaction in the Al domain of VWF, facilitating platelet adhesion. Other sites on VWF mediate binding to the blood vessel wall. Thus, VWF forms a bridge between the platelet and the vessel wall that is essential to platelet adhesion and primary hemostasis under conditions of high shear stress. Normally, endothelial cells secrete large polymeric forms of VWF and those forms of VWF that have a lower molecular weight arise from proteolytic cleavage. The multimers of exceptionally large molecular masses are stored in the Weibel-Pallade bodies of the endothelial cells and liberated upon stimulation by agonists such as thrombin and histamine.

[0005] For patients with VWD, it is recommended that they be treated with von

Willebrand factor (VWF) replacement given the need for prolonged hemostasis, particularly in major surgery (Mannucci PM and Franchini M., Haemophilia, 2017, 23(2): 182-187;

National Institutes of Health. National Heart, Lung, and Blood Institute. The Diagnosis, Evaluation, and Management of von Willebrand Disease NIH Publication No. 08-5832; December, 2007). Plasma-derived VWF therapies contain factor VIII (FVIII) and have the potential for FVIII accumulation with repeated dosing. VONVENDI ® (von Willebrand factor [recombinant], Shire, Westlake Village, CA) is the first and only recombinant VWF (rVWF) concentrate (Turecek PL, et al. Hamostaseologie. 2009;29(suppl l):S32-38; Mannucci PM, et al. Blood, 2013;122(5):648-657; Gill JC, et al. Blood, 2015;126(17):2038-2046).

[0006] Gastrointestinal (GI) bleeding events occur in up to 20% of patients with von Willebrand disease (VWD) and have been observed in association with angiodysplastic lesions in 2%-4% of patients with VWD. GI bleeds are closely associated with the absence of higher molecular weight and ultra-large multimers (ULMs) of von Willebrand factor (VWF), which are most often seen in patients with type 2A and type 3 VWD. Higher doses and longer durations of therapy with plasma-derived VWF replacement concentrates are usually needed to resolve GI bleeds compared with bleeds at other sites, and treatment may still be unsuccessful.

BRIEF SUMMARY OF THE INVENTION

[0007] The present invention provides a method for treating gastrointestinal bleeding in a patient with severe von Willebrand Disease (VWD). The method comprises administering to the subject at least one dose of recombinant von Willebrand Factor (rVWF) ranging from about 40 IU/kg to about 100 IU/kg, wherein the first dose further comprises recombinant Factor VIII (rFVIII).

[0008] In some embodiments, the rFVIII is administered at a dose of about 20 IU/kg to about 50 IU/kg. [0009] In some embodiments, the method further comprises administering to the subject a second dose of recombinant von Willebrand Factor (rVWF) ranging from about 40 IU/kg to about 100 IU/kg, wherein the second dose does not comprise recombinant Factor VIII (rFVIII).

[0010] In some embodiments, the rVWF to FVIII ratio is about 1.5:0.8. In some embodiments, the rVWF to FVIII ratio is about 1.3: 1. In some embodiments, the rVWF to FVIII ratio is about 1.1 :0.8. In some embodiments, the rVWF to FVIII ratio is about 1.5: 1. In some embodiments, the rVWF to FVIII ratio is about 1.1 : 1.2.

[0011] In some embodiments, the rVWF is administered every 8 to 12 hours.

[0012] In some embodiments, the 40-60 IU/kg rVWF of said rVWF is administered and wherein said gastrointestinal bleeding is minor or moderate gastrointestinal bleeding.

[0013] In some embodiments, the 40-80 IU/kg rVWF of said rVWF and wherein said gastrointestinal bleeding is major or severe gastrointestinal bleeding.

[0014] In some embodiments, the rVWF is administered every 8 to 12 hours for about 3 days to about 7 days.

[0015] In some embodiments, the 40-60 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, wherein said gastrointestinal bleeding is minor or moderate gastrointestinal bleeding.

[0016] In some embodiments, the 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, wherein said gastrointestinal bleeding is major or severe gastrointestinal bleeding.

[0017] In some embodiments, the subject has Type 3 VWD. In some embodiments, the subject has severe type 1 VWD. In some embodiments, the subject has severe type 2 VWD.

[0018] In some embodiments, the e subject had been treated for at least 1 bleeding event within the previous 12 months. In some embodiments, the subject had been treated for more than 1 bleeding event within the previous 12 months.

[0019] Other objects, advantages and embodiments of the invention will be apparent from the detailed description following. BRIEF DESCRIPTION OF THE DRAWINGS

[0020] FIG. 1 shows hemostatic efficiency of rVWF for treatment in GI bleeds.

BE=bleeding event; GI = gastrointestinal. 4 patients experienced a total of 6 GI bleeds. Minor and moderate BEs were rated as "Good" if 1-2 infusions more than estimated were required to control that bleeing episode and no additional VWF-containing product was required. Major BEs were rated as "Good" if < 1.5 times more infusions than estimated were required to control that bleeding episode and no additional VWF-containing product was required. Minor, moderate, and major BEs were rated as "Excellent" if the actual number of infusions was less than or equal to the estimated number required to treat the BE, and no additional VWF-containing product was required.

[0021] FIG. 2 shows VWF nucleic acid and amino acid sequences.

DETAILED DESCRIPTION OF THE INVENTION

Introduction

[0022] The present invention provides methods for treating gastrointestinal bleeding in a patient with severe von Willebrand Disease (VWD) comprising administering to the subject at least one dose of recombinant von Willebrand Factor (rVWF) ranging from about 40 IU/kg to about 100 IU/kg, wherein the first dose further comprises recombinant Factor VIII (rFVIII).

[0023] The disclosure of PCT Application Publication No. WO2012/171031 is herein incorporated by reference in its entirety for all purposes.

Definitions

[0024] As used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an antibody" includes a plurality of such antibodies and reference to "a host cell" includes reference to one or more host cells and equivalents thereof known to those skilled in the art, and so forth. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as "solely," "only" and the like in connection with the recitation of claim elements, or use of a "negative" limitation. [0025] Before the invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, 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.

[0026] As used herein. "rVWF" refers to recombinant VWF.

[0027] As used herein, "rFVIII" refers to recombinant FVIII.

[0028] The term "recombinant" when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all.

[0029] As used herein, "recombinant VWF" includes VWF obtained via recombinant DNA technology. In certain embodiments, VWF proteins of the invention can comprise a construct, for example, prepared as in WO 1986/06096 published on Oct. 23, 1986 and U.S. patent application Ser. No. 07/559,509, filed on Jul. 23, 1990, in the name of Ginsburg et al, which is incorporated herein by reference with respect to the methods of producing recombinant VWF. The VWF in the present invention can include all potential forms, including the monomeric and multimeric forms. It should also be understood that the present invention encompasses different forms of VWF to be used in combination. For example, the VWF of the present invention may include different multimers, different derivatives and both biologically active derivatives and derivatives not biologically active.

[0030] In the context of the present invention, the recombinant VWF embraces any member of the VWF family from, for example, a mammal such as a primate, human, monkey, rabbit, pig, rodent, mouse, rat, hamster, gerbil, canine, feline, and biologically active derivatives thereof. Mutant and variant VWF proteins having activity are also embraced, as are functional fragments and fusion proteins of the VWF proteins. Furthermore, the VWF of the invention may further comprise tags that facilitate purification, detection, or both. The VWF described herein may further be modified with a therapeutic moiety or a moiety suitable imaging in vitro or in vivo. [0031] As used herein, "plasma-derived VWF (pdVWF)" includes all forms of the protein found in blood including the mature VWF obtained from a mammal having the property of in vivo-stabilizing, e.g. binding, of at least one FVIII molecule.

[0032] The term "highly multimeric VWF" or "high molecular weight VWF" refers to VWF comprising at least 10 subunits, or 12, 14, or 16 subunits, to about 20, 22, 24 or 26 subunits or more. The term "subunit" refers to a monomer of VWF. As is known in the art, it is generally dimers of VWF that polymerize to form the larger order multimers (see Turecek et al, Semin. Thromb. Hemost. 2010, 36(5): 510-521 which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings regarding multimer analysis of VWF).

[0033] As used herein, the term "factor VIII" or "FVIII" refers to any form of factor VIII molecule with the typical characteristics of blood coagulation factor VIII, whether endogenous to a patient, derived from blood plasma, or produced through the use of recombinant DNA techniques, and including all modified forms of factor VIII. Factor VIII (FVIII) exists naturally and in therapeutic preparations as a heterogeneous distribution of polypeptides arising from a single gene product (see, e.g., Andersson et al, Proc. Natl. Acad. Sci. USA, 83:2979-2983 (1986)). Commercially available examples of therapeutic preparations containing Factor VIII include those sold under the trade names of HEMOFIL M, ADVATE, and RECOMBINATE (available from Baxter Healthcare Corporation, Deerfield, III, U.S.A.).

[0034] As used herein, "plasma FVIII activity" and "in vivo FVIII activity" are used interchangeably. The in vivo FVIII activity measured using standard assays may be endogenous FVIII activity, the activity of a therapeutically administered FVIII (recombinant or plasma derived), or both endogenous and administered FVIII activity. Similarly, "plasma FVIII" refers to endogenous FVIII or administered recombinant or plasma derived FVIII.

[0035] As used herein "von Willebrand Disease" refers to the group of diseases caused by a deficiency of von Willebrand factor. Von Willebrand factor helps blood platelets clump together and stick to the blood vessel wall, which is necessary for normal blood clotting. As described in further detail herein, there are several types of Von Willebrand disease including type l, 2A, 2B, 2M and 3.

[0036] The terms "isolated," "purified," or "biologically pure" refer to material that is substantially or essentially free from components that normally accompany it as found in its native state. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. VWF is the predominant species present in a preparation is substantially purified. The term "purified" in some embodiments denotes that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. In other embodiments, it means that the nucleic acid or protein is at least 50% pure, more preferably at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more pure. "Purify" or "purification" in other embodiments means removing at least one contaminant from the composition to be purified. In this sense, purification does not require that the purified compound be homogenous, e.g., 100% pure.

[0037] As used herein, "administering" (and all grammatical equivalents) includes intravenous administration, intramuscular administration, subcutaneous administration, oral administration, administration as a suppository, topical contact, intraperitoneal, intralesional, or intranasal administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject. Administration is by any route including parenteral, and transmucosal (e.g., oral, nasal, vaginal, rectal, or transdermal). Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial. Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.

[0038] The terms "therapeutically effective amount or dose" or "therapeutically sufficient amount or dose" or "effective or sufficient amount or dose" refer to a dose that produces therapeutic effects for which it is administered. For example, a therapeutically effective amount of a drug useful for treating hemophilia can be the amount that is capable of preventing or relieving one or more symptoms associated with hemophilia. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols. 1-3, 1992); Lloyd, The Art, Science and Technology of Pharmaceutical Compounding (1999); Pickar, Dosage Calculations (1999); and Remington: The Science and Practice of Pharmacy, 20th Edition, 2003, Gennaro, Ed., Lippincott, Williams & Wilkins).

[0039] As used herein, the terms "patient" and "subject" are used interchangeably and refer to a mammal (preferably human) that has a disease or has the potential of contracting a disease. [0040] As used herein, the term "about" denotes an approximate range of plus or minus 10% from a specified value. For instance, the language "about 20%" encompasses a range of 18-22%.

[0041] As used herein, the term "half-life" refers to the period of time it takes for the amount of a substance undergoing decay (or clearance from a sample or from a patient) to decrease by half.

I. Recombinant von Willebrand Factor (rVWF)

[0042] The present invention utilizes compositions comprising von Willebrand Factor (rVWF) for pretreatment of subject with severe VWD who are undergoing a surgical procedure, such as, but not limited to, major surgery, minor surgery, or oral surgery.

[0043] In certain embodiments, VWF proteins of the invention may comprise a construct, for example, prepared as in WO 1986/06096 published on Oct. 23, 1986 and U.S. patent application Ser. No. 07/559,509, filed on Jul. 23, 1990, in the name of Ginsburg et al., which is incorporated herein by reference with respect to the methods of producing recombinant VWF. The VWF useful for the present invention includes all potential forms, including the monomeric and multimeric forms. One particularly useful form of VWF are homo-multimers of at least two VWFs. The VWF proteins may be either a biologically active derivative, or when to be used solely as a stabilizer for FVIII the VWF may be of a form not biologically active. It should also be understood that the present invention encompasses different forms of VWF to be used in combination. For example, a composition useful for the present invention may include different multimers, different derivatives and both biologically active derivatives and derivatives not biologically active.

[0044] In primary hemostasis VWF serves as a bridge between platelets and specific components of the extracellular matrix, such as collagen. The biological activity of VWF in this process can be measured by different in vitro assays (Turecek et al, Semin. Thromb. Hemost. 28: 149-160, 2002). The ristocetin cofactor assay is based on the agglutination of fresh or formalin-fixed platelets induced by the antibiotic ristocetin in the presence of VWF.

[0045] The degree of platelet agglutination depends on the VWF concentration and can be measured by the turbidimetric method, e.g. by use of an aggregometer (Weiss et al, J. Clin. Invest. 52: 2708-2716, 1973; Macfarlane et al, Thromb. Diath. Haemorrh. 34: 306-308, 1975). The second method is the collagen binding assay, which is based on ELISA technology (Brown et Bosak, Thromb. Res. 43: 303-311, 1986; Favaloro, Thromb. Haemost. 83: 127-135, 2000). A microliter plate is coated with type I or III collagen. Then the VWF is bound to the collagen surface and subsequently detected with an enzyme-labeled polyclonal antibody. The last step is the substrate reaction, which can be photometrically monitored with an ELISA reader. As provided herein, the specific Ristocetin Cofactor activity of the VWF (VWF:RCo) of the present invention is generally described in terms of mU^g of VWF, as measured using in vitro assays.

[0046] An advantage of the rVWF compositions of the present invention over pdVWF is that rVWF exhibits a higher specific activity than pdVWF. In some embodiments, the rVWF of the invention has a specific activity of at least about 20, 22.5, 25, 27.5, 30, 32.5, 35, 37.5, 40, 42.5, 45, 47.5, 50, 52.5, 55, 57.5, 60, 62.5, 65, 67.5, 70, 72.5, 75, 77.5, 80, 82.5, 85, 87.5, 90, 92.5, 95, 97.5, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150 or more mU/ g.

[0047] The rVWF of the present invention is highly multimeric comprising about 10 to about 40 subunits. In further embodiments, the multimeric rVWF produced using methods of the present invention comprise about 10-30, 12-28, 14-26, 16-24, 18-22, 20-21 subunits. In further embodiments, the rVWF is present in multimers varying in size from dimers to multimers of over 40 subunits (>10 million Daltons). The largest multimers provide multiple binding sites that can interact with both platelet receptors and subendothelial matrix sites of injury, and are the most hemostatically active form of VWF. Application of ADAMTS13 will cleave the ultra-large rVWF multimers over time, but during production (generally through expression in cell culture), rVWF compositions of the present invention are generally not exposed to ADAMTS13 and retain their highly multimeric structure.

[0048] In one embodiment, a rVWF composition used in the methods described herein has a distribution of rVWF oligomers characterized in that 95% of the oligomers have between 6 subunits and 20 subunits. In other embodiments, the a rVWF composition has a distribution of rVWF oligomers characterized in that 95% of the oligomers have a range of subunits selected from variations 458 to 641 found in Table 2 of WO 2012/171031, which is herein incorporated by reference in its entirety for all purposes.

[0049] In one embodiment, a rVWF composition can be characterized according to the percentage of rVWF molecules that are present in a particular higher order rVWF multimer or larger multimer. For example, in one embodiment, at least 20% of rVWF molecules in a rVWF composition used in the methods described herein are present in an oligomeric complex of at least 10 subunits. In another embodiment, at least 20% of rVWF molecules in a rVWF composition used in the methods described herein are present in an oligomeric complex of at least 12 subunits. In yet other embodiments, a rVWF composition used in the methods provided herein has a minimal percentage (e.g., has at least X %) of rVWF molecules present in a particular higher-order rVWF multimer or larger multimer (e.g., a multimer of at least Y subunits) according to any one of variations 134 to 457 found in Table 3 to Table 5, which is herein incorporated by reference in its entirety for all purposes.

[0050] In accordance with the above, the rVWF composition administered to the subject (with or without FVIII) generally comprises a significant percentage of high molecular weight (HMW) rVWF multimers. In further embodiments, the HMW rVWF multimer composition comprises at least 10%-80% rVWF decamers or higher order multimers. In further embodiments, the composition comprises about 10-95%, 20-90%, 30-85%, 40-80%, 50-75%, 60-70% decamers or higher order multimers. In further embodiments, the HMW rVWF multimer composition comprises at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% decamers or higher order multimers.

[0051] Assessment of the number and percentage of rVWF multimers can be conducted using methods known in the art, including without limitation methods using electrophoresis and size exclusion chromatography methods to separate VWF multimers by size, for example as discussed by Cumming et al, (J Clin Pathol. 1993 May; 46(5): 470-473, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assessment of VWF multimers). Such techniques may further include

immunoblotting techniques (such as Western Blot), in which the gel is immunoblotted with a radiolabeled antibody against VWF followed by chemiluminescent detection (see for example Wen et al., (1993), J. Clin. Lab. Anal, 7: 317-323, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assessment of VWF multimers). Further assays for VWF include VWF:Antigen (VWF:Ag), VWF:Ristocetin Cofactor (VWF:RCof), and VWF:Collagen Binding Activity assay

(VWF:CBA), which are often used for diagnosis and classification of Von Willebrand Disease, (see for example Favaloro et al, Pathology, 1997, 29(4): 341-456, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assays for VWF).

[0052] In further embodiments, higher order rVWF multimers of the invention are stable for about 1 to about 90 hours post-administration. In still further embodiments, the higher order rVWF multimers are stable for about 5-80, 10-70, 15-60, 20-50, 25-40, 30-35 hours post-administration. In yet further embodiments, the higher order rVWF multimers are stable for at least 3, 6, 12, 18, 24, 36, 48, 72 hours post-administration. In certain embodiments the stability of the rVWF multimers is assessed in vitro.

[0053] In one embodiment, higher order rVWF multimers used in the compositions and methods provided herein have a half-life of at least 12 hour post administration. In another embodiment, the higher order rVWF multimers have a half-life of at least 24 hour post administration. In yet other embodiments, the higher order rVWF multimers have a half-life selected from variations 642 to 1045 found in Table 6 of WO 2012/171031, which is herein incorporated by reference in its entirety for all purposes.

[0054] In specific aspects, the rVWF (recombinant or plasma derived) used in accordance with the present invention are not modified with any conjugation, post-translation or covalent modifications. In particular embodiments, the rVWF of the present invention is not modified with a water soluble polymer, including without limitation, a polyethylene glycol (PEG), a polypropylene glycol, a polyoxyalkylene, a polysialic acid, hydroxyl ethyl starch, a poly- carbohydrate moiety, and the like.

[0055] In other aspects, the rVWF (recombinant or plasma derived) used in accordance with the present invention is modified through conjugation, post-translation modification, or covalent modification, including modifications of the N- or C-terminal residues as well as modifications of selected side chains, for example, at free sulfhydryl-groups, primary amines, and hydroxyl-groups. In one embodiment, a water soluble polymer is linked to the protein (directly or via a linker) by a lysine group or other primary amine. In one embodiment, the rVWF proteins of the present invention may be modified by conjugation of a water soluble polymer, including without limitation, a polyethylene glycol (PEG), a polypropylene glycol, a polyoxyalkylene, a polysialic acid, hydroxyl ethyl starch, a poly-carbohydrate moiety, and the like.

[0056] Water soluble polymers that may be used to modify the rVWF and/or FVIII include linear and branched structures. The conjugated polymers may be attached directly to the coagulation proteins of the invention, or alternatively may be attached through a linking moiety. Non-limiting examples of protein conjugation with water soluble polymers can be found in U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192, and

4,179,337, as well as in Abuchowski and Davis "Enzymes as Drugs," Holcenberg and Roberts, Eds., pp. 367 383, John Wiley and Sons, New York (1981), and Hermanson G., Bioconjugate Techniques 2nd Ed., Academic Press, Inc. 2008.

[0057] Protein conjugation may be performed by a number of well-known techniques in the art, for example, see Hermanson G., Bioconjugate Techniques 2nd Ed., Academic Press, Inc. 2008. Examples include linkage through the peptide bond between a carboxyl group on one of either the coagulation protein or water-soluble polymer moiety and an amine group of the other, or an ester linkage between a carboxyl group of one and a hydroxyl group of the other. Another linkage by which a coagulation protein of the invention could be conjugated to a water-soluble polymer compound is via a Schiff base, between a free amino group on the polymer moiety being reacted with an aldehyde group formed at the non-reducing end of the polymer by periodate oxidation (Jennings and Lugowski, J. Immunol. 1981 ; 127: 1011-8; Femandes and Gregonradis, Biochim Biophys Acta. 1997; 1341; 26-34). The generated Schiff Base can be stabilized by specific reduction with NaCNBEb to form a secondary amine. An alternative approach is the generation of terminal free amino groups on the polymer by reductive animation with NH4CI after prior oxidation. Bifunctional reagents can be used for linking two amino or two hydroxyl groups. For example a polymer containing an amino group can be coupled to an amino group of the coagulation protein with reagents like BS3 (Bis(sulfosuccinimidyl)suberate/Pierce, Rockford, 111.). In addition heterobifunctional cross linking reagents like Sulfo-EMCS (N-.epsilon.-Maleimidocaproyloxy) sulfosuccinimide ester/Pierce) can be used for instance to link amine and thiol groups. In other embodiments, an aldehyde reactive group, such as PEG alkoxide plus diethyl acetal of bromoacetaldehyde; PEG plus DMSO and acetic anhydride, and PEG chloride plus the phenoxide of 4- hydroxybenzaldehyde, succinimidyl active esters, activated dithiocarbonate PEG, 2,4,5- trichlorophenylcloroformate and P-nitrophenylcloroformate activated PEG, may be used in the conjugation of a coagulation protein.

[0058] In some aspects, the rVWF used in methods of the present invention has been matured in vitro with furin. In further embodiments, the furin is recombinant furin.

[0059] In further aspects, the rVWF used in the methods of the present invention are produced by expression in a mammalian cell culture using methods known in the art. In particular embodiments, the mammalian culture comprises CHO cells. In an exemplary embodiment, the rVWF of the invention comprises rVWF protein isolated from a CHO cell expression system. In a further embodiment, the propeptide removal is mediated in vitro through exposure of the pro-VWF to furin~in a still further embodiment, the Furin used for propeptide removal is recombinant furin. In as yet further embodiment, fully glycosylated/ ABO blood group glycans are absent.

[0060] In yet further embodiments, the rVWF used in methods and compositions of the present invention by expression in a suitable eukaryotic host system. Examples of eukaryotic cells include, without limitation, mammalian cells, such as CHO, COS, HEK 293, BHK, SK- Hep, and HepG2; insect cells, e.g., SF9 cells, SF21 cells, S2 cells, and High Five cells; and yeast cells, e.g., Saccharomyces or Schizosaccharomyces cells. In one embodiment, the VWF can be expressed in yeast cells, insect cells, avian cells, mammalian cells, and the like. For example, in a human cell line, a hamster cell line, or a murine cell line. In one particular embodiment, the cell line is a CHO, BHK, or HEK cell line. Typically, mammalian cells, e.g., CHO cell from a continuous cell line, can be used to express the VWF of the present invention.

[0061] In certain embodiments, the nucleic acid sequence comprising a sequence coding for VWF can be a vector. The vector can be delivered by a virus or can be a plasmid. The nucleic acid sequence coding for the protein can be a specific gene or a biologically functional part thereof. In one embodiment, the protein is at least a biologically active part of VWF. A wide variety of vectors can be used for the expression of the VWF and can be selected from eukaryotic expression vectors. Examples of vectors for eukaryotic expression include: (i) for expression in yeast, vectors such as pAO, pPIC, pYES, pMET, using promoters such as AOX1, GAP, GAL1, AUG1, etc; (ii) for expression in insect cells, vectors such as pMT, pAc5, pIB, pMIB, pBAC, etc., using promoters such as PH, plO, MT, Ac5, OpIE2, gp64, polh, etc., and (iii) for expression in mammalian cells, vectors such as pSVL, pCMV, pRc/RSV, pcDNA3, pBPV, etc., and vectors derived from viral systems such as vaccinia virus, adeno-associated viruses, herpes viruses, retroviruses, etc., using promoters such as CMV, SV40, EF-1, UbC, RSV, ADV, BPV, and β-actin.

[0062] In some embodiments of the present invention, the nucleic acid sequence further comprises other sequences suitable for a controlled expression of a protein such as promoter sequences, enhancers, TATA boxes, transcription initiation sites, polylinkers, restriction sites, poly-A-sequences, protein processing sequences, selection markers, and the like which are generally known to a person of ordinary skill in the art.

[0063] In certain embodiments, the cell-culture methods of the invention may comprise the use of a microcarrier. In some embodiments, the cell-cultures of the embodiments can be performed in large bioreactors under conditions suitable for providing high volume-specific culture surface areas to achieve high cell densities and protein expression. One means for providing such growth conditions is to use microcarriers for cell-culture in stirred tank bioreactors. The concept of cell-growth on microcarriers was first described by van Wezel (van Wezel, A. L., Nature 216:64-5 (1967)) and allows for cell attachment on the surface of small solid particles suspended in the growth medium. These methods provide for high surface-to-volume ratios and thus allow for efficient nutrient utilization. Furthermore, for expression of secreted proteins in eukaryotic cell lines, the increased surface-to-volume ratio allows for higher levels of secretion and thus higher protein yields in the supernatant of the culture. Finally, these methods allow for the easy scale-up of eukaryotic expression cultures.

[0064] The cells expressing VWF can be bound to a spherical or a porous microcarrier during cell culture growth. The microcarrier can be a microcarrier selected from the group of microcarriers based on dextran, collagen, plastic, gelatine and cellulose and others as described in Butler (1988. In: Spier & Griffiths, Animal Cell Biotechnology 3:283-303). It is also possible to grow the cells to a biomass on spherical microcarriers and subculture the cells when they have reached final fermenter biomass and prior to production of the expressed protein on a porous microcarrier or vice versa. Suitable spherical microcarriers can include smooth surface microcarriers, such as Cytodex™ 1, Cytodex™ 2, and Cytode™ 3 (GE Healthcare) and macroporous microcarriers such as Cytopore™. 1, Cytopore™ 2, Cytoline™ 1 , and Cytoline™ 2 (GE Healthcare).

[0065] In certain embodiments, rVWF is expressed in cells cultured in cell culture media that produces high molecular weight rVWF. The terms "cell culture solution," "cell culture medium or media," and "cell culture supernatant" refer to aspects of cell culture processes generally well known in the art. In the context of the present invention, a cell culture solution can include cell culture media and cell culture supernatant. The cell culture media are externally added to the cell culture solution, optionally together with supplements, to provide nutrients and other components for culturing the cells expressing VWF. The cell culture supernatant refers to a cell culture solution comprising the nutrients and other components from the cell culture medium as well as products released, metabolized, and/or excreted from the cells during culture. In further embodiments, the media can be animal protein-free and chemically defined. Methods of preparing animal protein-free and chemically defined culture media are known in the art, for example in US 2008/0009040 and US 2007/0212770, which are both incorporated herein for all purposes and in particular for all teachings related to cell culture media. "Protein free" and related terms refers to protein that is from a source exogenous to or other than the cells in the culture, which naturally shed proteins during growth. In another embodiment, the culture medium is polypeptide free. In another embodiment, the culture medium is serum free. In another embodiment the culture medium is animal protein free. In another embodiment the culture medium is animal component free. In another embodiment, the culture medium contains protein, e.g., animal protein from serum such as fetal calf serum. In another embodiment, the culture has recombinant proteins exogenously added. In another embodiment, the proteins are from a certified pathogen free animal. The term "chemically defined" as used herein shall mean, that the medium does not comprise any undefined supplements, such as, for example, extracts of animal components, organs, glands, plants, or yeast. Accordingly, each component of a chemically defined medium is accurately defined. In a preferred embodiment, the media are animal-component free and protein free.

[0066] In further embodiments, subsequent to purification from a mammalian cell culture, rFVIII is reconstituted prior to administration. In still further embodiments, the rVWF is treated with furin prior to or subsequent to reconstitution. In further embodiments, the Furin is recombinant furin. In still further embodiments, the rVWF of the invention is not exposed to ADAMTS 13, with the result that ultra large (i.e., comprising 10 or more subunits) are present in rVWF compositions of the invention.

[0067] In specific aspects, the rVWF used in methods of the present invention is contained in a formulation containing a buffer, a sugar and/or a sugar alcohol (including without limitation trehalose and mannitol), a stabilizer (such as glycine), and a surfactant (such as polysorbate 80). In further embodiments, for formulations containing rFVIII, the formulation may further include sodium, histidine, calcium, and glutathione.

[0068] In one aspect, the formulations comprising rVWF is lyophilized prior to administration. Lyophilization is carried out using techniques common in the art and should be optimized for the composition being developed [Tang et al, Pharm Res. 21 : 191 -200. (2004) and Chang et al, Pharm Res. 13:243-9 (1996)] .

[0069] Methods of preparing pharmaceutical formulations can include one or more of the following steps: adding a stabilizing agent as described herein to said mixture prior to lyophilizing, adding at least one agent selected from a bulking agent, an osmolality regulating agent, and a surfactant, each of which as described herein, to said mixture prior to lyophilization. A lyophilized formulation is, in one aspect, at least comprised of one or more of a buffer, a bulking agent, and a stabilizer. In this aspect, the utility of a surfactant is evaluated and selected in cases where aggregation during the lyophilization step or during reconstitution becomes an issue. An appropriate buffering agent is included to maintain the formulation within stable zones of pH during lyophilization.

[0070] The standard reconstitution practice for lyophilized material is to add back a volume of pure water or sterile water for injection (WFI) (typically equivalent to the volume removed during lyophilization), although dilute solutions of antibacterial agents are sometimes used in the production of pharmaceuticals for parenteral administration [Chen, Drug Development and Industrial Pharmacy, 18: 131 1 -1354 (1992)] . Accordingly, methods are provided for preparation of reconstituted recombinant VWF compositions comprising the step of adding a diluent to a lyophilized recombinant VWF composition of the invention.

[0071] The lyophilized material may be reconstituted as an aqueous solution. A variety of aqueous carriers, e.g., sterile water for injection, water with preservatives for multi dose use, or water with appropriate amounts of surfactants (for example, an aqueous suspension that contains the active compound in admixture with excipients suitable for the manufacture of aqueous suspensions). In various aspects, such excipients are suspending agents, for example and without limitation, sodium carboxymethylcellulose, methylcellulose,

hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents are a naturally-occurring phosphatide, for example and without limitation, lecithin, or condensation products of an alkylene oxide with fatty acids, for example and without limitation, poly oxy ethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example and without limitation, heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example and without limitation, polyethylene sorbitan monooleate. In various aspects, the aqueous suspensions also contain one or more preservatives, for example and without limitation, ethyl, or n-propyl, p-hydroxybenzoate.

[0072] In certain embodiments, compositions of the present invention are liquid formulations for administration with the use of a syringe or other storage vessel. In further embodiments, these liquid formulations are produced from lyophilized material described herein reconstituted as an aqueous solution. [0073] In a further aspect, the compositions of the invention further comprise one or more pharmaceutically acceptable carriers. The phrases "pharmaceutically" or "pharmacologically" acceptable refer to molecular entities and compositions that are stable, inhibit protein degradation such as aggregation and cleavage products, and in addition do not produce allergic, or other adverse reactions when administered using routes well-known in the art, as described below. "Pharmaceutically acceptable carriers" include any and all clinically useful solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like, including those agents disclosed above.

II. Production of Recombinant VWF [0074] The free mature recombinant von Willebrand Factor (rVWF) of the present invention can be produced recombinantly. One skilled in the art recognizes useful methods for expressing a recombinant protein in a host cell. In some instances, the method includes expressing a nucleic acid sequence encoding rVWF in a host cell such as a CHO cell and culturing the resulting host cell under certain conditions to produce rVWF, prepro-VWF, pro- VWF, and the like.

[0075] In certain embodiments, the nucleic acid sequence comprising a sequence coding for VWF can be an expression vector. The vector can be delivered by a virus or can be a plasmid. The nucleic acid sequence coding for the protein can be a specific gene or a biologically functional part thereof. In one embodiment, the protein is at least a biologically active part of VWF. The nucleic acid sequence can further comprise other sequences suitable for a controlled expression of a protein such as promoter sequences, enhancers, TATA boxes, transcription initiation sites, polylinkers, restriction sites, poly-A-sequences, protein processing sequences, selection markers, and the like which are generally known to a person of ordinary skill in the art.

[0076] A wide variety of vectors can be used for the expression of the VWF and can be selected from eukaryotic expression vectors. Examples of vectors for eukaryotic expression include: (i) for expression in yeast, vectors such as pAO, pPIC, pYES, pMET, using promoters such as AOXl, GAP, GALl, AUGl, etc; (ii) for expression in insect cells, vectors such as pMT, pAc5, pIB, pMIB, pBAC, etc., using promoters such as PH, plO, MT, Ac5, OpIE2, gp64, polh, etc., and (iii) for expression in mammalian cells, vectors such as pSVL, pCMV, pRc/RSV, pcDNA3, pBPV, etc., and vectors derived from viral systems such as vaccinia virus, adeno-associated viruses, herpes viruses, retroviruses, etc., using promoters such as CMV, SV40, EF-1, UbC, RSV, ADV, BPV, and β -actin.

[0077] In some aspects, the rVWF used in the methods of the present invention is produced by expression in a mammalian cell culture using methods known in the art. In particular embodiments, the mammalian culture comprises CHO cells. In further embodiments, the rVWF is co-expressed with recombinant Factor VIII (rFVIII) in the same culture. In such embodiments, the rVWF and the rFVIII are purified together (co-purified) or separately using methods known in the art. In other embodiments, the rVWF is expressed in a culture that does not contain rFVIII.

[0078] In some embodiments, rVWF is expressed and isolated from a suitable eukaryotic host system. Examples of eukaryotic cells include, without limitation, mammalian cells, such as CHO, COS, HEK 293, BHK, SK-Hep, and HepG2; insect cells, e.g. , SF9 cells, SF21 cells, S2 cells, and High Five cells; and yeast cells, e.g. , Saccharomyces or Schizosaccharomyces cells. In one embodiment, the VWF can be expressed in yeast cells, insect cells, avian cells, mammalian cells, and the like. For example, in a human cell line, a hamster cell line, or a murine cell line. In one particular embodiment, the cell line is a CHO, BHK, or HEK cell line. Typically, mammalian cells, e.g., CHO cell from a continuous cell line, can be used to express the VWF of the present invention. In certain instances, VWF protein is expressed and isolated from a CHO cell expression system.

[0079] VWF can be produced in a cell culture system or according to any cell culture method recognized by those in the art. In some embodiments, the cell cultures can be performed in large bioreactors under conditions suitable for providing high volume-specific culture surface areas to achieve high cell densities and protein expression. One means for providing such growth conditions is to use microcarriers for cell-culture in stirred tank bioreactors. The concept of cell-growth on microcarriers was first described by van Wezel (van Wezel, A. L., Nature , 1967, 216:64-5) and allows for cell attachment on the surface of small solid particles suspended in the growth medium. These methods provide for high surface-to-volume ratios and thus allow for efficient nutrient utilization. Furthermore, for expression of secreted proteins in eukaryotic cell lines, the increased surface-to-volume ratio allows for higher levels of secretion and thus higher protein yields in the supernatant of the culture. Finally, these methods allow for the easy scale-up of eukaryotic expression cultures. [0080] The cells expressing VWF can be bound to a spherical or a porous microcarrier during cell culture growth. The microcarrier can be a microcarrier selected from the group of microcarriers based on dextran, collagen, plastic, gelatine and cellulose and others as described in Butler (1988. In: Spier & Griffiths, Animal Cell Biotechnology 3:283-303). 1 1 is also possible to grow the cells to a biomass on spherical microcarriers and subculture the cells when they have reached final fermenter biomass and prior to production of the expressed protein on a porous microcarrier or vice versa. Suitable spherical microcarriers can include smooth surface microcarriers, such as Cytodex™ 1, Cytodex™ 2, and Cytodex™ 3 (GE Healthcare) and macroporous microcarriers such as Cytopore™ 1, Cytopore™ 2, Cytoline™ 1 , and Cytoline™ 2 (GE Healthcare).

[0081] In a further embodiment, the VWF propeptide is cleaved from the non-mature VWF in vitro through exposure of the pro-VWF to furin. In some embodiments, the furin used for propeptide cleavage is recombinant furin.

[0082] In certain embodiments, rVWF is expressed in cells cultured in cell culture media that produces high molecular weight rVWF. The terms "cell culture solution," "cell culture medium or media," and "cell culture supernatant" refer to aspects of cell culture processes generally well known in the art. In the context of the present invention, a cell culture solution can include cell culture media and cell culture supernatant. The cell culture media are externally added to the cell culture solution, optionally together with supplements, to provide nutrients and other components for culturing the cells expressing VWF. The cell culture supernatant refers to a cell culture solution comprising the nutrients and other components from the cell culture medium as well as products released, metabolized, and/or excreted from the cells during culture. In further embodiments, the media can be animal protein-free and chemically defined. Methods of preparing animal protein-free and chemically defined culture media are known in the art, for example in US 2006/0094104, US 2007/0212770, and US 2008/0009040, which are both incorporated herein for all purposes and in particular for all teachings related to cell culture media. "Protein free" and related terms refers to protein that is from a source exogenous to or other than the cells in the culture, which naturally shed proteins during growth. In another embodiment, the culture medium is polypeptide free. In another embodiment, the culture medium is serum free. In another embodiment the culture medium is animal protein free. In another embodiment the culture medium is animal component free. In another embodiment, the culture medium contains protein, e.g. , animal protein from serum such as fetal calf serum. In another embodiment, the culture has recombinant proteins exogenously added. In another embodiment, the proteins are from a certified pathogen free animal. The term "chemically defined" as used herein shall mean, that the medium does not comprise any undefined supplements, such as, for example, extracts of animal components, organs, glands, plants, or yeast. Accordingly, each component of a chemically defined medium is accurately defined. In a preferred embodiment, the media are animal-component free and protein free.

[0083] In certain embodiments, the culture of cells expressing VWF can be maintained for at least about 7 days, or at least about 14 days, 21 days, 28 days, or at least about 5 weeks, 6 weeks, 7 weeks, or at least about 2 months, or 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18 months or longer. The cell density at which a cell-culture is maintained at for production of a recombinant VWF protein will depend upon the culture-conditions and medium used for protein expression. One of skill in the art will readily be able to determine the optimal cell density for a cell-culture producing an VWF. In one embodiment, the culture is maintained at a cell density of between about 0.5xl0 6 and 4xl0 7 cells/ml for an extended period of time. In other embodiments, the cell density is maintained at a concentration of between about l .Oxl O 6 and about l .Oxl O 7 cells/ml for an extended period of time. In other embodiments, the cell density is maintained at a concentration of between about l .OxlO 6 and about 4.0xl 0 6 cells/ml for an extended period of time. In other embodiments, the cell density is maintained at a concentration of between about l .Oxl O 6 and about 4.0xl0 6 cells/ml for an extended period of time. In yet other embodiments, the cell density may be maintained at a concentration between about 2.0xl0 6 and about 4.0xl0 6 , or between about l .OxlO 6 and about 2.5xl 0 6 , or between about 1.5xl0 6 and about 3.5xl 0 6 , or any other similar range, for an extended period of time. After an appropriate time in cell culture, the rVWF can be isolated from the expression system using methods known in the art.

[0084] In a specific embodiment, the cell density of the continuous cell culture for production of rVWF is maintained at a concentration of no more than 2.5xl0 6 cells/mL for an extended period. In other specific embodiments, the cell density is maintained at no more than 2.0xl0 6 cells/mL, 1.5xl 0 6 cells/mL, l .Oxl O 6 cells/mL, 0.5xl 0 6 cells/mL, or less. In one embodiment, the cell density is maintained at between 1.5xl0 6 cells/mL and 2.5xl0 6 cells/mL.

[0085] In one embodiment of the cell cultures described above, the cell culture solution comprises a medium supplement comprising copper. Such cell culture solutions are described, for example, in U. S. Patent No. 8,852,888 and U. S. Patent No. 9,409,971, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to cell culture methods and compositions for producing recombinant VWF.

[0086] The polynucleotide and amino acid sequences of prepro-VWF are set out in SEQ ID NO: 1 and SEQ ID NO:2, respectively, and are available at GenBank Accession Nos. NM_000552 (Homo sapiens von Willebrand factor (VWF) mRNA) and NP_000543, respectively. The amino acid sequence corresponding to the mature VWF protein is set out in SEQ ID NO: 3 (corresponding to amino acids 764-2813 of the full length prepro-VWF amino acid sequence). In some embodiments, the VWF exhibits at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 100% identity to the sequence of SEQ ID NO:3. In some embodiments, the rVWF of the invention exhibits at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 100% identity to the sequence of SEQ ID NO:3. See, for example, U.S. Patent No. 8,597,910, U.S. Patent Publication No. 2016/0129090, as well as FIG. 6.

[0087] One form of useful rVWF has at least the property of in vivo-stabilizing, e.g. binding, of at least one Factor VIII (FVIII) molecule and having optionally a glycosylation pattern which is pharmacologically acceptable. Specific examples thereof include VWF without the A2 domain thus resistant to proteolysis (Lankhof et al. , Thromb. Haemost. 77: 1008-1013, 1997), and a VWF fragment from Val 449 to Asn 730 including the glycoprotein lb-binding domain and binding sites for collagen and heparin (Pietu et al. , Biochem.

Biophys. Res. Commun. 164: 1339-1347, 1989). The determination of the ability of a VWF to stabilize at least one FVIII molecule is, in one aspect, carried out in VWF-deficient mammals according to methods known in the state in the art.

[0088] The rVWF of the present invention can be produced by any method known in the art. One specific example is disclosed in WO86/06096 published on Oct. 23, 1986 and U.S. Patent Application No. 07/559,509, filed on Jul. 23, 1990, which is incorporated herein by reference with respect to the methods of producing recombinant VWF. Thus, methods are known in the art for (i) the production of recombinant DNA by genetic engineering, e.g. via reverse transcription of RNA and/or amplification of DNA, (ii) introducing recombinant DNA into prokaryotic or eukaryotic cells by transfection, e.g. via electroporation or microinjection, (iii) cultivating the transformed cells, e.g. in a continuous or batchwise manner, (iv) expressing VWF, e.g. constitutively or upon induction, and (v) isolating the VWF, e.g. from the culture medium or by harvesting the transformed cells, in order to (vi) obtain purified rVWF, e.g. via anion exchange chromatography or affinity chromatography. A recombinant VWF is, in one aspect, made in transformed host cells using recombinant DNA techniques well known in the art. For instance, sequences coding for the polypeptide could be excised from DNA using suitable restriction enzymes. Alternatively, the DNA molecule is, in another aspect, synthesized using chemical synthesis techniques, such as the phosphoramidate method. Also, in still another aspect, a combination of these techniques is used.

[0089] The invention also provides vectors encoding polypeptides of the invention in an appropriate host. The vector comprises the polynucleotide that encodes the polypeptide operatively linked to appropriate expression control sequences. Methods of effecting this operative linking, either before or after the polynucleotide is inserted into the vector, are well known. Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation. The resulting vector having the polynucleotide therein is used to transform an appropriate host. This

transformation may be performed using methods well known in the art.

[0090] Any of a large number of available and well-known host cells are used in the practice of this invention. The selection of a particular host is dependent upon a number of factors recognized by the art, including, for example, compatibility with the chosen expression vector, toxicity of the peptides encoded by the DNA molecule, rate of transformation, ease of recovery of the peptides, expression characteristics, bio-safety and costs. A balance of these factors must be struck with the understanding that not all host cells are equally effective for the expression of a particular DNA sequence. Within these general guidelines, useful microbial host cells include, without limitation, bacteria, yeast and other fungi, insects, plants, mammalian (including human) cells in culture, or other hosts known in the art.

[0091] Transformed host cells are cultured under conventional fermentation conditions so that the desired compounds are expressed. Such fermentation conditions are well known in the art. Finally, the polypeptides are purified from culture media or the host cells themselves by methods well known in the art.

[0092] Depending on the host cell utilized to express a compound of the invention, carbohydrate (oligosaccharide) groups are optionally attached to sites that are known to be glycosylation sites in proteins. Generally, O-linked oligosaccharides are attached to serine (Ser) or threonine (Thr) residues while N-linked oligosaccharides are attached to asparagine (Asn) residues when they are part of the sequence Asn-X-Ser/Thr, where X can be any amino acid except proline. X is preferably one of the 19 naturally occurring amino acids not counting proline. The structures of N-linked and O-linked oligosaccharides and the sugar residues found in each type are different. One type of sugar that is commonly found on both N-linked and O-linked oligosaccharides is N-acetylneuraminic acid (referred to as sialic acid). Sialic acid is usually the terminal residue of both N-linked and O-linked

oligosaccharides and, by virtue of its negative charge, in one aspect, confers acidic properties to the glycosylated compound. Such site(s) may be incorporated in the linker of the compounds of this invention and are preferably glycosylated by a cell during recombinant production of the polypeptide compounds (e.g. , in mammalian cells such as CHO, BHK, COS). In other aspects, such sites are glycosylated by synthetic or semi- synthetic procedures known in the art.

[0093] In some embodiments, sialysation (also referred to as sialylation), can be performed on the column as part of the purification procedures described herein (including the anion exchange, cation exchange, size exclusion, and/or immunoaffinity methods). In some embodiments, the sialylation results in increased stability of the rVWF as compared to rVWF that has not undergone sialylation. In some embodiments, the sialylation results in increased stability of the rVWF in blood circulation (for example, after administration to a subject) as compared to rVWF that has not undergone sialylation. In some embodiments, the increased stability of salivated rVWF results in an increase of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more as compared rVWF that has not undergone sialylation. In some embodiments, the sialylation results in increased half-life for the rVWF as compared to rVWF that has not undergone sialylation. In some embodiments, the sialylation results in increased half-life for the rVWF in blood circulation (for example, after administration to a subject) as compared to rVWF that has not undergone sialylation. In some embodiments, the increased half-life of sialylated rVWF results in an increase of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more as compared rVWF that has not undergone sialylation. In some embodiments, the increased half-life of sialylated rVWF results in rVWF that is stable for 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 12 hours, 24 hours or more in blood circulation (for example, after administration to a subject) as compared to rVWF that has not undergone sialylation. In some embodiments, sialylation increases the number of 2,3 sialylation and/or 2,6 sialylation. In some embodiments, sialylation is increased by the addition of 2,3 sialyltransferase and/or 2,6 sialyltransferase and CMP -NANA (Cytidine-5'-monophospho-N- acetylneuraminic acid sodium salt) as an additional buffer step. In some embodiments, sialylation is increased by the addition of 2,3 sialyltransferase and CMP -NANA (Cytidine-5'- monophospho-N-acetylneuraminic acid sodium salt) as an additional buffer step. In some embodiments, 2,3 sialylation is increased by the addition of 2,3 sialyltransferase and CMP- NANA (Cytidine-5'-monophospho-N-acet lneuraminic acid sodium salt) as an additional buffer step.

[0094] In some embodiments, 2,6 sialylation is increased by the addition of 2,6 sialyltransferase and CMP -NANA (Cytidine-5 '-monophospho-N-acetylneuraminic acid sodium salt) as an additional buffer step. In some embodiments, 2,3 sialylation and/or 2,6 sialylation are increased by the addition of 2,3 sialyltransferase and/or 2,6 sialyltransferase and CMP -NANA (Cytidine-5 '-monophospho-N-acetylneuraminic acid sodium salt) as an additional buffer step. In some embodiments, CMP -NANA is chemically or enzymatic modified to transfer modified sialic acid to potential free position. In some embodiments, sialylation is performed by loading rVWF onto the resin, washing with one or more buffers as described herein to deplete unwanted impurities, apply one or more buffers containing sialyltransferase and CMP -NANA at conditions that allow additional sialylation, and washing with one or more buffers to deplete excess of the sialylation reagents, and eluting with one or more buffers the enhanced rVWF (e.g., the rVWF with increased sialylation). In some embodiments, the sialylation process is performed as part of a cation exchange method, an anion exchange method, a size exclusion method, or an immunoaffinity purification method, as described herein.

[0095] Alternatively, the compounds are made by synthetic methods using, for example, solid phase synthesis techniques. Suitable techniques are well known in the art, and include those described in Merrifield (1973), Chem. Polypeptides, pp. 335-61 (Katsoyannis and

Panayotis eds.); Merrifield (1963), J. Am. Chem. Soc. 85: 2149; Davis et al. (1985),

Biochem. Intl. 10: 394-414; Stewart and Young (1969), Solid Phase Peptide Synthesis; U.S.

Pat. No. 3,941,763; Finn et al. (1976), The Proteins (3rd ed.) 2: 105-253; and Erickson et al. (1976), The Proteins (3rd ed.) 2: 257-527 '. Solid phase synthesis is the preferred technique of making individual peptides since it is the most cost-effective method of making small peptides [0096] Fragments, variants and analogs of VWF can be produced according to methods that are well-known in the art. Fragments of a polypeptide can be prepared using, without limitation, enzymatic cleavage (e.g. , trypsin, chymotrypsin) and also using recombinant means to generate a polypeptide fragments having a specific amino acid sequence.

Polypeptide fragments may be generated comprising a region of the protein having a particular activity, such as a multimerization domain or any other identifiable VWF domain known in the art.

[0097] Methods of making polypeptide analogs are also well-known. Amino acid sequence analogs of a polypeptide can be substitutional, insertional, addition or deletion analogs. Deletion analogs, including fragments of a polypeptide, lack one or more residues of the native protein which are not essential for function or immunogenic activity. Insertional analogs involve the addition of, e.g. , amino acid(s) at a non-terminal point in the polypeptide. This analog may include, for example and without limitation, insertion of an immunoreactive epitope or simply a single residue. Addition analogs, including fragments of a polypeptide, include the addition of one or more amino acids at either or both termini of a protein and include, for example, fusion proteins. Combinations of the aforementioned analogs are also contemplated.

[0098] Substitutional analogs typically exchange one amino acid of the wild-type for another at one or more sites within the protein, and may be designed to modulate one or more properties of the polypeptide without the complete loss of other functions or properties. In one aspect, substitutions are conservative substitutions. "Conservative amino acid substitution" is substitution of an amino acid with an amino acid having a side chain or a similar chemical character. Similar amino acids for making conservative substitutions include those having an acidic side chain (glutamic acid, aspartic acid); a basic side chain (arginine, lysine, histidine); a polar amide side chain (glutamine, asparagine); a hydrophobic, aliphatic side chain (leucine, isoleucine, valine, alanine, glycine); an aromatic side chain (phenylalanine, tryptophan, tyrosine); a small side chain (glycine, alanine, serine, threonine, methionine); or an aliphatic hydroxyl side chain (serine, threonine).

[0099] In one aspect, analogs are substantially homologous or substantially identical to the recombinant VWF from which they are derived. Analogs include those which retain at least some of the biological activity of the wild-type polypeptide, e.g. blood clotting activity. [00100] Polypeptide variants contemplated include, without limitation, polypeptides chemically modified by such techniques as ubiquitination, glycosylation, including polysialation (or polysialylation), conjugation to therapeutic or diagnostic agents, labeling, covalent polymer attachment such as pegylation (derivatization with polyethylene glycol), introduction of non-hydrolyzable bonds, and insertion or substitution by chemical synthesis of amino acids such as ornithine, which do not normally occur in human proteins. Variants retain the same or essentially the same binding properties of non-modified molecules of the invention. Such chemical modification may include direct or indirect (e.g., via a linker) attachment of an agent to the VWF polypeptide. In the case of indirect attachment, it is contemplated that the linker may be hydrolyzable or non-hydrolyzable.

[00101] Preparing pegylated polypeptide analogs will in one aspect comprise the steps of (a) reacting the polypeptide with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the binding construct polypeptide becomes attached to one or more PEG groups, and (b) obtaining the reaction product(s). In general, the optimal reaction conditions for the acylation reactions are determined based on known parameters and the desired result. For example, the larger the ratio of PEG: protein, the greater the percentage of poly -pegylated product. In some embodiments, the binding construct has a single PEG moiety at the N-terminus. Polyethylene glycol (PEG) may be attached to the blood clotting factor to, for example, provide a longer half-life in vivo. The PEG group may be of any convenient molecular weight and is linear or branched. The average molecular weight of the PEG ranges from about 2 kiloDalton ("kD") to about 100 kDa, from about 5 kDa to about 50 kDa, or from about 5 kDa to about 10 kDa. In certain aspects, the PEG groups are attached to the blood clotting factor via acylation or reductive alkylation through a natural or engineered reactive group on the PEG moiety (e.g. , an aldehyde, amino, thiol, or ester group) to a reactive group on the blood clotting factor (e.g., an aldehyde, amino, or ester group) or by any other technique known in the art.

[00102] Methods for preparing polysialylated polypeptide are described in United States Patent Publication 20060160948, Fernandes et Gregoriadis; Biochim. Biophys. Acta 1341 : 26-34, 1997, and Saenko et al , Haemophilia 12:42-51, 2006. Briefly, a solution of colominic acid (CA) containing 0.1 M NaI04 is stirred in the dark at room temperature to oxidize the

CA. The activated CA solution is dialyzed against, e.g., 0.05 M sodium phosphate buffer, pH 7.2 in the dark and this solution was added to a rVWF solution and incubated for 18 h at room temperature in the dark under gentle shaking. Free reagents are optionally be separated from the rVWF-polysialic acid conjugate by, for example, ultrafiltration/diafiltration.

Conjugation of rVWF with polysialic acid is achieved using glutaraldehyde as cross-linking reagent (Migneault et al , Biotechniques 37: 790-796, 2004).

[00103] It is also contemplated in another aspect that prepro-VWF and pro-VWF polypeptides will provide a therapeutic benefit in the formulations of the present invention. For example, US Patent No. 7,005,502 describes a pharmaceutical preparation comprising substantial amounts of pro-VWF that induces thrombin generation in vitro. In addition to recombinant, biologically active fragments, variants, or other analogs of the naturally- occurring mature VWF, the present invention contemplates the use of recombinant biologically active fragments, variants, or analogs of the prepro-VWF (set out in SEQ ID NO:2) or pro-VWF polypeptides (amino acid residues 23 to 764 of SEQ ID NO: 2) in the formulations described herein.

[00104] Polynucleotides encoding fragments, variants and analogs may be readily generated by a worker of skill to encode biologically active fragments, variants, or analogs of the naturally-occurring molecule that possess the same or similar biological activity to the naturally-occurring molecule. In various aspects, these polynucleotides are prepared using PCR techniques, digestion/ligation of DNA encoding molecule, and the like. Thus, one of skill in the art will be able to generate single base changes in the DNA strand to result in an altered codon and a missense mutation, using any method known in the art, including, but not limited to site- specific mutagenesis. As used herein, the phrase "moderately stringent hybridization conditions" means, for example, hybridization at 42 0 C in 50% formamide and washing at 60°C in 0.1 x SSC, 0.1% SDS. It is understood by those of skill in the art that variation in these conditions occurs based on the length and GC nucleotide base content of the sequences to be hybridized. Formulas standard in the art are appropriate for determining exact hybridization conditions. See Sambrook et al , 9.47-9.51 in Molecular Cloning, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989).

A. VWF Multimers

[00105] Assessment of the number and percentage of rVWF multimers can be conducted using methods known in the art, including without limitation methods using electrophoresis and size exclusion chromatography methods to separate VWF multimers by size, for example as discussed by Cumming et al, (J Clin Pathol, 1993 May; 46(5): 470-473, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assessment of VWF multimers). Such techniques may further include

immunoblotting techniques (such as Western Blot), in which the gel is immunoblotted with a radiolabelled antibody against VWF followed by chemiluminescent detection (see, for example, Wen et al, J. Clin. Lab. Anal., 1993, 7: 317-323, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assessment of VWF multimers). Further assays for VWF include VWF:Antigen (VWF:Ag), VWF:Ristocetin Cofactor (VWF:RCof), and VWF:Collagen Binding Activity assay

(VWF:CBA), which are often used for diagnosis and classification of Von Willebrand Disease (see, for example, Favaloro et al , Pathology, 1997, 29(4): 341-456; Sadler, JE, Annu Rev Biochem, 1998, 67:395-424; and Turecek et al , Semin Thromb Hemost, 2010, 36:510- 521, which are hereby incorporated by reference in their entirety for all purposes and in particular for all teachings related to assays for VWF). In some embodiments, the rVWF obtained using the present methods includes any multimer pattern present in the loading sample of the rVWF. In some embodiments, the rVWF obtained using the present methods includes physiological occurring multimer patters as well as ultralarge VWF-multimer patterns.

b. VWF Assays

[00106] In primary hemostasis VWF serves as a bridge between platelets and specific components of the extracellular matrix, such as collagen. The biological activity of VWF in this process can be measured by different in vitro assays (Turecek et al, Semin Thromb Hemost, 2010, 36: 510-521).

[00107] The VWF: Ristocetin Cofactor (VWF:RCof) assay is based on the agglutination of fresh or formalin-fixed platelets induced by the antibiotic ristocetin in the presence of VWF. The degree of platelet agglutination depends on the VWF concentration and can be measured by the turbidimetric method, e.g. , by use of an aggregometer (Weiss et al , J. Clin. Invest, 1973, 52: 2708-2716; Macfarlane et al , Thromb. Diath. Haemorrh., 1975, 34: 306-308). As provided herein, the specific ristocetin cofactor activity of the VWF (VWF:RCo) of the present invention is generally described in terms of mU^g of VWF, as measured using in vitro assays.

[00108] In some embodiments, the rVWF purified according to the methods of the present invention has a specific activity of at least about 20, 22.5, 25, 27,5, 30, 32.5, 35, 37.5, 40, 42.5, 45, 47.5, 50, 52.5, 55, 57.5, 60, 62.5, 65, 67.5, 70, 72.5, 75, 77.5, 80, 82.5, 85, 87.5, 90, 92.5, 95, 97.5, 100, 105, 110, 1 15, 120, 125, 130, 135, 140, 145, 150 or more mU^g. In some embodiments, rVWF used in the methods described herein has a specific activity of from 20 mU^g to 150 mU In some embodiments, the rVWF has a specific activity of from 30 mU^g to 120 mU^g. In some embodiments, the rVWF has a specific activity from 40 mU^g to 90 mU^g. In some embodiments, the rVWF has a specific activity selected from variations 1 to 133 found in Table 3, below.

Table 3. Exemplary embodiments for the specific activity of rVWF found in the

com ositions and used in the methods rovided herein.

Var. = Variation [00109] The rVWF of the present invention is highly multimeric comprising about 10 to about 40 subunits. In further embodiments, the multimeric rVWF produced using methods of the present invention comprise about 10-30, 12-28, 14-26, 16-24, 18-22, 20-21 subunits. In some embodiments, the rVWF is present in multimers varying in size from dimers to multimers of over 40 subunits (> 10 million Daltons). The largest multimers provide multiple binding sites that can interact with both platelet receptors and subendothelial matrix sites of injury, and are the most hemostatically active form of VWF. In some embodiments, the rVWF of the present invention comprises ultralarge multimers (ULMs). Generally, high and ultralarge multimers are considered to be hemostatically most effective (see, for example, Turecek, P., Hamostaseologie, (Vol. 37): Supplement 1, pages S15-S25 (2017)). In some embodiments, the rVWF is between 500 kDa and 20,000 kDa. In some embodiments, any desired multimer pattern can be obtained using the methods described. In some

embodiments, when anion exchange and/or cation exchanger methods are employed, the pH, conductivity, and/or counterion concentration of the buffers in the one or more wash step(s) or the gradient buffers can be manipulated to obtain the desired multimer pattern. In some embodiments, then size exclusion chromatography methods are employed, the collection criteria can be employed to obtain the desired multimer pattern. In some embodiments, the described multimer partem comprises ultralarge multimers. In some embodiments, the ultralarge multimers are at least 10,000 kDa, at least 11,000 kDa, at least 12,000 kDa, at least 13,000 kDa, at least 14,000 kDa, at least 15,000 kDa, at least 16,000 kDa, at least 17,000 kDa, at least 18,000 kDa, at least 19,000 kDa, at least 20,000 kDa. In some embodiments, the ultralarge multimers are between about 10,000 kDa and 20,000 kDa. In some

embodiments, the ultralarge multimers are between about 11,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 12,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 13,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 14,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 15,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 16,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 17,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 18,000 kDa and 20,000 kDa. In some embodiments, the ultralarge multimers are between about 19,000 kDa and 20,000 kDa. In some embodiments, the rVWF obtained using the present methods includes any multimer pattern present in the loading sample of the rVWF. In some embodiments, the rVWF obtained using the present methods includes physiolocical occurring multimer patters as well as ultra large VWF-multimer patterns.

[00110] In some embodiments, the rVWF composition prepared by the purification method described herein has a distribution of rVWF oligomers characterized in that 95% of the oligomers have between 6 subunits and 20 subunits. In some embodiments, the rVWF composition has a distribution of rVWF oligomers characterized in that 95% of the oligomers have a range of subunits selected from variations 458 to 641 found in 4.

Table 4. Exemplary embodiments for the distribution of rVWF oligomers found in the compositions and used in the methods provided herein.

4-10 Var. 490 10-18 Var. 536 18-34 Var. 582 28-32 Var. 628

4-8 Var. 491 10-16 Var. 537 18-32 Var. 583 28-30 Var. 629

6-40 Var. 492 10-14 Var. 538 18-30 Var. 584 30-40 Var. 630

6-38 Var. 493 10-12 Var. 539 18-28 Var. 585 30-38 Var. 631

6-36 Var. 494 12-40 Var. 540 18-26 Var. 586 30-36 Var. 632

6-34 Var. 495 12-38 Var. 541 18-24 Var. 587 30-34 Var. 633

6-32 Var. 496 12-36 Var. 542 18-22 Var. 588 30-32 Var. 634

6-30 Var. 497 12-34 Var. 543 18-20 Var. 589 32-40 Var. 635

6-28 Var. 498 12-32 Var. 544 20-40 Var. 590 32-38 Var. 636

6-26 Var. 499 12-30 Var. 545 20-38 Var. 591 32-36 Var. 637

6-24 Var. 500 12-28 Var. 546 20-36 Var. 592 32-34 Var. 638

6-22 Var. 501 12-26 Var. 547 20-34 Var. 593 34-40 Var. 639

6-20 Var. 502 12-24 Var. 548 20-32 Var. 594 36-38 Var. 640

6-18 Var. 503 12-22 Var. 549 20-30 Var. 595 38-40 Var. 641

Var. = Variation

[00111] In some embodiments, the rVWF composition prepared by the methods provided herein can be characterized according to the percentage of rVWF molecules that are present in a particular higher order rVWF multimer or larger multimer. For example, in one embodiment, at least 20% of rVWF molecules in a rVWF composition used in the methods described herein are present in an oligomeric complex of at least 10 subunits. In another embodiment, at least 20% of rVWF molecules in a rVWF composition used in the methods described herein are present in an oligomeric complex of at least 12 subunits. In yet other embodiments, a rVWF composition used in the methods provided herein has a minimal percentage (e.g. , has at least X%) of rVWF molecules present in a particular higher-order rVWF multimer or larger multimer (e.g. , a multimer of at least Y subunits) according to any one of variations 134 to 457 found in Table 5 to Table 7.

Table 5. Exemplary embodiments for the percentage of rVWF molecules that are present in a particular higher order rVWF multimer or larger multimer found in the compositions and used in the methods provided herein.

55% Var. 143 Var. 161 Var. 179 Var. 197 Var. 215 Var. 233

60% Var. 144 Var. 162 Var. 180 Var. 198 Var. 216 Var. 234

65% Var. 145 Var. 163 Var. 181 Var. 199 Var. 217 Var. 235

70% Var. 146 Var. 164 Var. 182 Var. 200 Var. 218 Var. 236

75% Var. 147 Var. 165 Var. 183 Var. 201 Var. 219 Var. 237

80% Var. 148 Var. 166 Var. 184 Var. 202 Var. 220 Var. 238

85% Var. 149 Var. 167 Var. 185 Var. 203 Var. 221 Var. 239

90% Var. 150 Var. 168 Var. 186 Var. 204 Var. 222 Var. 240

95% Var. 151 Var. 169 Var. 187 Var. 205 Var. 223 Var. 241

Var. = Variation

Table 6. Exemplary embodiments for the percentage of rVWF molecules that are present in a particular higher order rVWF multimer or larger multimer found in the compositions and used in the methods provided herein.

Var. = Variation

Table 7. Exemplary embodiments for the percentage of rVWF molecules that are present in a particular higher order rVWF multimer or larger multimer found in the compositions and used in the methods provided herein.

30% Var. 354 Var. 372 Var. 390 Var. 408 Var. 426 Var. 444

35% Var. 355 Var. 373 Var. 391 Var. 409 Var. 427 Var. 445

40% Var. 356 Var. 374 Var. 392 Var. 410 Var. 428 Var. 446

45% Var. 357 Var. 375 Var. 393 Var. 411 Var. 429 Var. 447

50% Var. 358 Var. 376 Var. 394 Var. 412 Var. 430 Var. 448

55% Var. 359 Var. 377 Var. 395 Var. 413 Var. 431 Var. 449

60% Var. 360 Var. 378 Var. 396 Var. 414 Var. 432 Var. 450

65% Var. 361 Var. 379 Var. 397 Var. 415 Var. 433 Var. 451

70% Var. 362 Var. 380 Var. 398 Var. 416 Var. 434 Var. 452

75% Var. 363 Var. 381 Var. 399 Var. 417 Var. 435 Var. 453

80% Var. 364 Var. 382 Var. 400 Var. 418 Var. 436 Var. 454

85% Var. 365 Var. 383 Var. 401 Var. 419 Var. 437 Var. 455

90% Var. 366 Var. 384 Var. 402 Var. 420 Var. 438 Var. 456

95% Var. 367 Var. 385 Var. 403 Var. 421 Var. 439 Var. 457

Var. = Variation

[00112] In accordance with the above, the rVWF comprises a significant percentage of high molecular weight (HMW) rVWF multimers. In further embodiments, the HMW rVWF multimer composition comprises at least 10% - 80% rVWF decamers or higher order multimers. In further embodiments, the composition comprises about 10-95%, 20-90%, 30- 85%, 40-80%, 50-75%, 60-70% decamers or higher order multimers. In further

embodiments, the HMW rVWF multimer composition comprises at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% decamers or higher order multimers.

[00113] Assessment of the number and percentage of rVWF multimers can be conducted using methods known in the art, including without limitation methods using electrophoresis and size exclusion chromatography methods to separate rVWF multimers by size, for example as discussed by Cumming et al, (J Clin Pathol. 1993 May; 46(5): 470-473, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assessment of rVWF multimers). Such techniques may further include immunoblotting techniques (such as Western Blot), in which the gel is immunoblotted with a radiolabelled antibody against VWF followed by chemiluminescent detection (see for example Wen et al, (1993), J. Clin. Lab. Anal, 7: 317-323, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assessment of rVWF multimers). Further assays for VWF include VWF: Antigen (VWF: Ag), VWF Ristocetin Cofactor (VWF:RCof), and VWF:Collagen Binding Activity assay

(VWF:CBA), which are often used for diagnosis and classification of Von Willebrand Disease, (see for example Favaloro et al, Pathology, 1997, 29(4): 341-456, which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to assays for VWF).

[00114] In some embodiments, the ratio of rFVIII procoagulant activity (IU rFVIILC) to rVWF Ristocetin cofactor activity (IU rVWF:RCo) for the rVWF prepared according to the methods of the present invention is between 3:1 and 1 :5. In further embodiments, the ratio is between 2:1 and 1:4. In still further embodiments, the ratio is between 5:2 and 1:4. In further embodiments, the ratio is between 3:2 and 1:3. In still further embodiments, the ratio is about 1:1, 1:2, 1:3, 1:4, 1:5, 2:1, 2:3, 2:4, 2:5, 3:1, 3:2, 3:4, or 3:5. In further embodiments, the ratio is between 1 : 1 and 1 :2. In yet further embodiments, the ratio is 1.1:1, 1.2:1, 1.3:1, 1.4:1, 1.5:1, 1.6:1, 1.7:1, 1.8:1, 1.9:1, or 2:1. In certain embodiments, the ratio of rFVIII procoagulant activity (IU rFVIII: C) to rVWF Ristocetin cofactor activity (IU rVWF:RCo) in a composition useful for a method described herein is selected from variations 1988 to 2140 found in Table 8.

Table 8. Exemplary embodiments for the ratio of rFVIII procoagulant activity (IU rFVIILC) to rVWF Ristocetin cofactor activity (IU rVWF:RCo) in compositions and used in methods provided herein.

(IU rFVIII: C) to (IU rFVIII: C) to (IU rFVIII: C) to (IU rFVIII: C) to (IU rVWF:RCo) (IU rVWF:RCo) (IU rVWF:RCo) (IU rVWF:RCo)

Var. Var. Var. Var.

4:1 3:1-3:5 4:3-1:4 4:5-2:3

1988 2027 2065 2103

Var. Var. Var. Var.

3:1 3:1-2:3 4:3-1:3 4:5-3:4

1989 2028 2066 2104

Var. Var. Var. Var.

2:1 3:1-3:4 4:3-2:5 3:4-1:6

1990 2029 2067 2105

Var. Var. Var. Var.

3:2 3:1-4:5 4:3-1:2 3:4-1:5

1991 2030 2068 2106

Var. Var. Var. Var.

4:3 3:1-5:6 4:3-3:5 3:4-1:4

1992 2031 2069 2107

Var. Var. Var. Var.

1:1 3:1-1:1 4:3-2:3 3:4-1:3

1993 2032 2070 2108

Var. Var. Var. Var.

5:6 3:1-4:3 4:3-3:4 3:4-2:5

1994 2033 2071 2109

Var. Var. Var. Var.

4:5 3:1-3:2 4:3-4:5 3:4-1:2

1995 2034 2072 2110

Var. Var. Var. Var.

3:4 3:1-2:1 4:3-5:6 3:4-3:5

1996 2035 2073 2111

Var. Var. Var. Var.

2:3 2:1-1:6 4:3-1:1 3:4-2:3

1997 2036 2074 2112

Var. Var. Var. Var.

3:5 2:1-1:5 1:1-1:6 2:3-1:6

1998 2037 2075 2113

Var. Var. Var. Var.

1:2 2:1-1:4 1:1-1:5 2:3-1:5

1999 2038 2076 2114

Var. Var. Var. Var.

2:5 2:1-1:3 1:1-1:4 2:3-1:4

2000 2039 2077 2115

Var. Var. Var. Var.

1:3 2:1-2:5 1:1-1:3 2:3-1:3

2001 2040 2078 2116 Var. Var. Var. Var.

1:4 2:1-1:2 1:1-2:5 2:3-2:5

2002 2041 2079 2117

Var. Var. Var. Var.

1:5 2:1-3:5 1:1-1:2 2:3-1:2

2003 2042 2080 2118

Var. Var. Var. Var.

1:6 2:1-2:3 1:1-3:5 2:3-3:5

2004 2043 2081 2119

Var. Var. Var. Var.

4:1-1:6 2:1-3:4 1:1-2:3 3:5-1:6

2005 2044 2082 2120

Var. Var. Var. Var.

4:1-1:5 2:1-4:5 1:1-3:4 3:5-1:5

2006 2045 2083 2121

Var. Var. Var. Var.

4:1-1:4 2:1-5:6 1:1-4:5 3:5-1:4

2007 2046 2084 2122

Var. Var. Var. Var.

4:1-1:3 2:1-1:1 1:1-5:6 3:5-1:3

2008 2047 2085 2123

Var. Var. Var. Var.

4:1-2:5 2:1-4:3 5:6-1:6 3:5-2:5

2009 2048 2086 2124

Var. Var. Var. Var.

4:1-1:2 2:1-3:2 5:6-1:5 3:5-1:2

2010 2049 2087 2125

Var. Var. Var. Var.

4:1-3:5 3:2-1:6 5:6-1:4 1:2-1:6

2011 2050 2088 2126

Var. Var. Var. Var.

4:1-2:3 3:2-1:5 5:6-1:3 1:2-1:5

2012 2051 2089 2127

Var. Var. Var. Var.

4:1-3:4 3:2-1:4 5:6-2:5 1:2-1:4

2013 2052 2090 2128

Var. Var. Var. Var.

4:1-4:5 3:2-1:3 5:6-1:2 1:2-1:3

2014 2053 2091 2129

Var. Var. Var. Var.

4:1-5:6 3:2-2:5 5:6-3:5 1:2-2:5

2015 2054 2092 2130

Var. Var. Var. Var.

4:1-1:1 3:2-1:2 5:6-2:3 2:5-1:6

2016 2055 2093 2131

Var. Var. Var. Var.

4:1-4:3 3:2-3:5 5:6-3:4 2:5-1:5

2017 2056 2094 2132

Var. Var. Var. Var.

4:1-3:2 3:2-2:3 5:6-4:5 2:5-1:4

2018 2057 2095 2133

Var. Var. Var. Var.

4:1-2:1 3:2-3:4 4:5-1:6 2:5-1:3

2019 2058 2096 2134

Var. Var. Var. Var.

4:1-3:1 3:2-4:5 4:5-1:5 1:3-1:6

2020 2059 2097 2135

Var. Var. Var. Var.

3:1-1:6 3:2-5:6 4:5-1:4 1:3-1:5

2021 2060 2098 2136

Var. Var. Var. Var.

3:1-1:5 3:2-1:1 4:5-1:3 1:3-1:4

2022 2061 2099 2137

Var. Var. Var. Var.

3:1-1:4 3:2-4:3 4:5-2:5 1:4-1:6

2023 2062 2100 2138

Var. Var. Var. Var.

3:1-1:3 4:3-1:6 4:5-1:2 1:4-1:5

2024 2063 2101 2139

Var. Var. Var. Var.

3:1-2:5 4:3-1:5 4:5-3:5 1:5-1:6

2025 2064 2102 2140

Var.

3:1-1:2

2026

Var. = Variation

[00115] In further embodiments, higher order rVWF multimers of the invention are stable for about 1 to about 90 hours post-administration. In still further embodiments, the higher order rVWF multimers are stable for about 5-80, 10-70, 15-60, 20-50, 25-40, 30-35 hours post-administration. In yet further embodiments, the higher order rVWF multimers are stable for at least 3, 6, 12, 18, 24, 36, 48, 72 hours post-administration. In certain embodiments the stability of the rVWF multimers is assessed in vitro.

[00116] In one embodiment, higher order rVWF multimers used in the compositions and methods provided herein have a half-life of at least 12 hour post administration. In another embodiment, the higher order rVWF multimers have a half-life of at least 24 hour post administration. In yet other embodiments, the higher order rVWF multimers have a half-life selected from variations 642 to 1045 found in Table 9.

Table 9. Exemplary embodiments for the half-life of higher order rVWF multimers found in the compositions prepared by the methods provided herein.

at least 90 Var. 674 6-8 Var. 775 16-27 Var. 876 33-42 Var. 977

2-90 Var. 675 8-90 Var. 776 16-24 Var. 877 33-29 Var. 978

2-84 Var. 676 8-84 Var. 777 16-22 Var. 878 33-36 Var. 979

2-78 Var. 677 8-78 Var. 778 16-20 Var. 879 36-90 Var. 980

2-72 Var. 678 8-72 Var. 779 16-18 Var. 880 36-84 Var. 981

2-66 Var. 679 8-66 Var. 780 18-90 Var. 881 36-78 Var. 982

2-60 Var. 680 8-60 Var. 781 18-84 Var. 882 36-72 Var. 983

2-54 Var. 681 8-54 Var. 782 18-78 Var. 883 36-66 Var. 984

2-48 Var. 682 8-48 Var. 783 18-72 Var. 884 36-60 Var. 985

2-45 Var. 683 8-45 Var. 784 18-66 Var. 885 36-54 Var. 986

2-42 Var. 684 8-42 Var. 785 18-60 Var. 886 36-48 Var. 987

2-39 Var. 685 8-39 Var. 786 18-54 Var. 887 36-45 Var. 988

2-36 Var. 686 8-36 Var. 787 18-48 Var. 888 36-42 Var. 989

2-33 Var. 687 8-33 Var. 788 18-45 Var. 889 36-39 Var. 990

2-30 Var. 688 8-30 Var. 789 18-42 Var. 890 39-90 Var. 991

2-27 Var. 689 8-27 Var. 790 18-39 Var. 891 39-84 Var. 992

2-24 Var. 690 8-24 Var. 791 18-36 Var. 892 39-78 Var. 993

2-22 Var. 691 8-22 Var. 792 18-33 Var. 893 39-72 Var. 994

2-20 Var. 692 8-20 Var. 793 18-30 Var. 894 39-66 Var. 995

2-18 Var. 693 8-18 Var. 794 18-27 Var. 895 39-60 Var. 996

2-16 Var. 694 8-16 Var. 795 18-24 Var. 896 39-54 Var. 997

2-14 Var. 695 8-14 Var. 796 18-22 Var. 897 39-48 Var. 998

2-12 Var. 696 8-12 Var. 797 18-20 Var. 898 39-45 Var. 999

2-10 Var. 697 8-10 Var. 798 20-90 Var. 899 39-42 Var. 1000

2-8 Var. 698 10-90 Var. 799 20-84 Var. 900 42-90 Var. 1001

2-6 Var. 699 10-84 Var. 800 20-78 Var. 901 42-84 Var. 1002

2-4 Var. 700 10-78 Var. 801 20-72 Var. 902 42-78 Var. 1003

3-90 Var. 701 10-72 Var. 802 20-66 Var. 903 42-72 Var. 1004

3-84 Var. 702 10-66 Var. 803 20-60 Var. 904 42-66 Var. 1005

3-78 Var. 703 10-60 Var. 804 20-54 Var. 905 42-60 Var. 1006

3-72 Var. 704 10-54 Var. 805 20-48 Var. 906 42-54 Var. 1007

3-66 Var. 705 10-48 Var. 806 20-45 Var. 907 42-48 Var. 1008

3-60 Var. 706 10-45 Var. 807 20-42 Var. 908 42-45 Var. 1009

3-54 Var. 707 10-42 Var. 808 20-39 Var. 909 45-90 Var. 1010

3-48 Var. 708 10-39 Var. 809 20-36 Var. 910 45-84 Var. 1011

3-45 Var. 709 10-36 Var. 810 20-33 Var. 911 45-78 Var. 1012

3-42 Var. 710 10-33 Var. 811 20-30 Var. 912 45-72 Var. 1013

3-39 Var. 711 10-30 Var. 812 20-27 Var. 913 45-66 Var. 1014

3-36 Var. 712 10-27 Var. 813 20-24 Var. 914 45-60 Var. 1015

3-33 Var. 713 10-24 Var. 814 20-22 Var. 915 45-54 Var. 1016

3-30 Var. 714 10-22 Var. 815 22-90 Var. 916 45-48 Var. 1017

3-27 Var. 715 10-20 Var. 816 22-84 Var. 917 48-90 Var. 1018

3-24 Var. 716 10-18 Var. 817 22-78 Var. 918 48-84 Var. 1019

3-22 Var. 717 10-16 Var. 818 22-72 Var. 919 48-78 Var. 1020

3-20 Var. 718 10-14 Var. 819 22-66 Var. 920 48-72 Var. 1021

3-18 Var. 719 10-12 Var. 820 22-60 Var. 921 48-66 Var. 1022

3-16 Var. 720 12-90 Var. 821 22-54 Var. 922 48-60 Var. 1023 3-14 Var. 721 12-84 Var. 822 22-48 Var. 923 48-54 Var. 1024

3-12 Var. 722 12-78 Var. 823 22-45 Var. 924 54-90 Var. 1025

3-10 Var. 723 12-72 Var. 824 22-42 Var. 925 54-84 Var. 1026

3-8 Var. 724 12-66 Var. 825 22-39 Var. 926 54-78 Var. 1027

3-6 Var. 725 12-60 Var. 826 22-36 Var. 927 54-72 Var. 1028

3-4 Var. 726 12-54 Var. 827 22-33 Var. 928 54-66 Var. 1029

4-90 Var. 727 12-48 Var. 828 22-30 Var. 929 54-60 Var. 1030

4-84 Var. 728 12-45 Var. 829 22-27 Var. 930 60-90 Var. 1031

4-78 Var. 729 12-42 Var. 830 22-24 Var. 931 60-84 Var. 1032

4-72 Var. 730 12-39 Var. 831 24-90 Var. 932 60-78 Var. 1033

4-66 Var. 731 12-36 Var. 832 24-84 Var. 933 60-72 Var. 1034

4-60 Var. 732 12-33 Var. 833 24-78 Var. 934 60-66 Var. 1035

4-54 Var. 733 12-30 Var. 834 24-72 Var. 935 66-90 Var. 1036

4-48 Var. 734 12-27 Var. 835 24-66 Var. 936 66-84 Var. 1037

4-45 Var. 735 12-24 Var. 836 24-60 Var. 937 66-78 Var. 1038

4-42 Var. 736 12-22 Var. 837 24-54 Var. 938 66-72 Var. 1039

4-39 Var. 737 12-20 Var. 838 24-48 Var. 939 72-90 Var. 1040

4-36 Var. 738 12-18 Var. 839 24-45 Var. 940 72-84 Var. 1041

4-33 Var. 739 12-16 Var. 840 24-42 Var. 941 72-78 Var. 1042

4-30 Var. 740 12-14 Var. 841 24-39 Var. 942 78-90 Var. 1043

4-27 Var. 741 14-90 Var. 842 24-36 Var. 943 78-84 Var. 1044

4-24 Var. 742 14-84 Var. 843 24-33 Var. 944 84-90 Var. 1045

Var. = Variation

[00117] In some embodiments, the pro-VWF and/or purified rVWF purified in accordance with the present invention is not modified with any conjugation, post-translation or covalent modifications. In particular embodiments, the pro-VWF and/or purified rVWF of the present invention is not modified with a water soluble polymer, including without limitation, a polyethylene glycol (PEG), a polypropylene glycol, a polyoxyalkylene, a polysialic acid, hydroxyl ethyl starch, a poly-carbohydrate moiety, and the like.

[00118] In some embodiments, the pro-VWF and/or purified rVWF purified in accordance with the present invention is modified through conjugation, post-translation modification, or covalent modification, including modifications of the N- or C- terminal residues as well as modifications of selected side chains, for example, at free sulfhydryl-groups, primary amines, and hydroxyl-groups. In one embodiment, a water soluble polymer is linked to the protein (directly or via a linker) by a lysine group or other primary amine. In some embodiments, the pro-VWF and/or purified rVWF of the present invention may be modified by conjugation of a water soluble polymer, including without limitation, a polyethylene glycol (PEG), a polypropylene glycol, a polyoxyalkylene, a polysialic acid, hydroxyl ethyl starch, a poly- carbohydrate moiety, and the like. [00119] Water soluble polymers that may be used to modify the pro-VWF and/or purified rVWF include linear and branched structures. The conjugated polymers may be attached directly to the coagulation proteins of the invention, or alternatively may be attached through a linking moiety. Non-limiting examples of protein conjugation with water soluble polymers can be found in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192, and 4,179,337, as well as in Abuchowski and Davis "Enzymes as Drugs," Holcenberg and Roberts, Eds., pp. 367 383, John Wiley and Sons, New York (1981), and Hermanson G., Bioconjugate Techniques 2nd Ed., Academic Press, Inc. 2008.

[00120] Protein conjugation may be performed by a number of well-known techniques in the art, for example, see Hermanson G., Bioconjugate Techniques 2nd Ed., Academic Press, Inc. 2008. Examples include linkage through the peptide bond between a carboxyl group on one of either the coagulation protein or water-soluble polymer moiety and an amine group of the other, or an ester linkage between a carboxyl group of one and a hydroxyl group of the other. Another linkage by which a coagulation protein of the invention could be conjugated to a water-soluble polymer compound is via a Schiff base, between a free amino group on the polymer moiety being reacted with an aldehyde group formed at the non-reducing end of the polymer by periodate oxidation (Jennings and Lugowski, J. Immunol. 1981; 127: 1011-8; Femandes and Gregonradis, Biochim Biophys Acta. 1997; 1341; 26-34). The generated Schiff Base can be stabilized by specific reduction with NaCNBFb to form a secondary amine. An alternative approach is the generation of terminal free amino groups on the polymer by reductive animation with NH4CI after prior oxidation. Bifunctional reagents can be used for linking two amino or two hydroxyl groups. For example, a polymer containing an amino group can be coupled to an amino group of the coagulation protein with reagents like BS3 (Bis(sulfosuccinimidyl)suberate/Pierce, Rockford, 111.). In addition,

heterobifunctional cross linking reagents like Sulfo-EMCS (Ν-ε-Maleimidocaproyloxy) sulfosuccinimide ester/Pierce) can be used for instance to link amine and thiol groups. In other embodiments, an aldehyde reactive group, such as PEG alkoxide plus diethyl acetal of bromoacetaldehyde; PEG plus DMSO and acetic anhydride, and PEG chloride plus the phenoxide of 4-hydroxybenzaldehyde, succinimidyl active esters, activated dithiocarbonate PEG, 2,4,5-trichlorophenylcloroformate and P-nitrophenylcloroformate activated PEG, may be used in the conjugation of a coagulation protein.

[00121] Another method for measuring the biological activity of VWF is the collagen binding assay, which is based on ELISA technology (Brown and Bosak, Thromb. Res., 1986, 43:303-311 ; Favaloro, Thromb. Haemost, 2000, 83 127-135). A microtiter plate is coated with type I or III collagen. Then the VWF is bound to the collagen surface and subsequently detected with an enzyme-labeled polyclonal antibody. The last step is a substrate reaction, which can be photometrically monitored with an ELISA reader.

[00122] Immunological assays of von Willebrand factors (VWF:Ag) are immunoassays that measure the concentration of the VWF protein in plasma. They give no indication as to VWF function. A number of methods exist for measuring VWF:Ag and these include both enzyme-linked immunosorbent assay (ELISA) or automated latex immunoassays (LIA.) Many laboratories now use a fully automated latex immunoassay. Historically laboratories used a variety of techniques including Laurell electroimmunoassay 'Laurell Rockets' but these are rarely used in most labs today.

III. Kits

[00123] As an additional aspect, the invention includes kits which comprise one or more lyophilized compositions packaged in a manner which facilitates their use for administration to subjects. In one embodiment, such a kit includes pharmaceutical formulation described herein (e.g. , a composition comprising a therapeutic protein or peptide), packaged in a container such as a sealed bottle or vessel, with a label affixed to the container or included in the package that describes use of the compound or composition in practicing the method. In one embodiment, the pharmaceutical formulation is packaged in the container such that the amount of headspace in the container (e.g. , the amount of air between the liquid formulation and the top of the container) is very small. Preferably, the amount of headspace is negligible (e.g., almost none). In one embodiment, the kit contains a first container having a therapeutic protein or peptide composition and a second container having a physiologically acceptable reconstitution solution for the composition. In one aspect, the pharmaceutical formulation is packaged in a unit dosage form. The kit may further include a device suitable for administering the pharmaceutical formulation according to a specific route of administration. Preferably, the kit contains a label that describes use of the pharmaceutical formulations.

IV. rVWF for Methods of Treating GI Bleeding in Patient with Severe VWD

[00124] One of the advantages of administering rVWF to subjects with severe VWD to pretreat for surgery is that the higher specific activity of rVWF as compared to pdVWF allows flexibility in the amount of rVWF administered and the number of times the subject is re-dosed. As will be appreciated and as is discussed in further detail herein, the coadministered FVIII may be recombinant or plasma derived

[00125] Single or multiple administrations of rVWF are carried out with the dose levels and pattern being selected by the treating physician. For the prevention or treatment of disease, the appropriate dosage depends on the type of disease to be treated (e.g., von Willebrand disease), the severity and course of the disease, whether drug is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the drug, and the discretion of the attending physician.

[00126] In some aspects, rVWF is administered prior to a surgical procedure to a subject at a range from 20-60 IU/kg, e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 20-60, 35-70, 20-40, 35-60, 45-60, 45-55, 45-50, 50-60, 55-60, or 50-55 IU/kg. In some embodiments, rVWF is administered between 12 hours and 24 hours, e.g., 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, 24 hours, 12 hours and 24 hours, 14 hours and 24 hours, 16 and 24 hours, 18 hours and 24 hours, or 20 hours and 24 hours prior to the surgical procedure. In some aspects, Factor VIII (FVIII) is not administered with the rVWF prior to the surgical procedure.

[00127] In some embodiments, rVWF is administered to the subject at a range of 5-90 IU/kg, e.g., 5-90, 5-50, 10-90, 15-90, 20-90, 30-90, 40-90, 50-90, 60-90, 70-90, 80-90, 5-80, 10-70, 20-60, 30-50, 35-60, 5-50, 5-40, 5-30. 5-20, 10-90, 10-50, or 20-40 IU/kg 1 hour prior to surgery. In other embodiments, rVWF is administered at a dose of 70-200 IU/kg, e.g., 70- 200, 80-200-, 90-200, 100-200, 110-200, 120-200, 130-200, 130-200, 140-200, 150-200, 160-200, 170-200, 180-200, 190-200, 70-170, 80-180, 60-160, 50-150, 40-140, 30, 130, 20- 120, 10-110, 70-100, or 70-90 IU/kg after the surgery. In some cases, the surgical procedure is selected from a group consisting of major surgery, minor surgery, and oral surgery.

[00128] In some embodiments, the subject is administered 35-60 IU/kg rVWF between 12 hours and 24 hours prior to major surgery. In other embodiments, the subject is administered 15-90 IU/kg rVWF 1 hour prior to major surgery. In another embodiment, the subject is administered 150-220 IU/kg rVWF after major surgery. In some instances, the subject undergoing major surgery is administered a total dosage of 220-320 IU/kg. [00129] In some embodiments, the subject is administered 50-60 IU/kg rVWF between 12 hours and 24 hours prior to minor surgery. In other embodiments, the subject is administered 5-50 IU/kg rVWF 1 hour prior to minor surgery. In another embodiment, the subject is administered 70-150 IU/kg rVWF after minor surgery. In some instances, the subject undergoing minor surgery is administered a total dosage of 100-220 IU/kg.

[00130] In some embodiments, the subject is administered 20-40 IU/kg rVWF between 12 hours and 24 hours prior to oral surgery. In other embodiments, the subject is administered 20-50 IU/kg rVWF 1 hour prior to oral surgery. In another embodiment, the subject is administered 10-50 IU/kg rVWF during oral surgery. In another embodiment, the subject is administered 20-50 IU/kg rVWF after oral surgery. In some instances, the subject undergoing oral surgery is administered a total dosage of 70-190 IU/kg.

[00131] Compositions of rVWF can be contained in pharmaceutical formulations, as described herein. Such formulations can be administered orally, topically, transdermally, parenterally, by inhalation spray, vaginally, rectally, or by intracranial injection. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques. Administration by intravenous, intradermal, intramuscular, intramammary, intraperitoneal, intrathecal, retrobulbar, intrapulmonary injection and or surgical implantation at a particular site is contemplated as well. Generally, compositions are essentially free of pyrogens, as well as other impurities that could be harmful to the recipient.

[00132] In one aspect, formulations of the invention are administered by an initial bolus followed by a continuous infusion to maintain therapeutic circulating levels of drug product. As another example, the inventive compound is administered as a one-time dose. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient. The route of administration can be, but is not limited to, by intravenous, intraperitoneal, subcutaneous, or intramuscular administration. The frequency of dosing depends on the pharmacokinetic parameters of the agents and the route of administration. The optimal pharmaceutical formulation is determined by one skilled in the art depending upon the route of administration and desired dosage. See for example, Remington's Pharmaceutical

Sciences, 18th Ed., 1990, Mack Publishing Co., Easton, Pa. 18042 pages 1435-1712, the disclosure of which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to formulations, routes of administration and dosages for pharmaceutical products. Such formulations influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agents. Depending on the route of administration, a suitable dose is calculated according to body weight, body surface area or organ size. Appropriate dosages may be ascertained through use of established assays for determining blood level dosages in conjunction with appropriate dose-response data. The final dosage regimen is determined by the attending physician, considering various factors which modify the action of drugs, e.g. the drug's specific activity, the severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. By way of example, a typical dose of a recombinant VWF of the present invention is approximately 50 IU/kg, equal to 500 μg/kg. As studies are conducted, further information will emerge regarding the appropriate dosage levels and duration of treatment for various diseases and conditions.

[00133] The practice of the present invention may employ, unless otherwise indicated, conventional techniques and descriptions of organic chemistry, polymer technology, molecular biology (including recombinant techniques), cell biology, biochemistry, and immunology, which are within the skill of the art. Such conventional techniques include polymer array synthesis, hybridization, ligation, and detection of hybridization using a label. Specific illustrations of suitable techniques can be had by reference to the example herein below. However, other equivalent conventional procedures can, of course, also be used. Such conventional techniques and descriptions can be found in standard laboratory manuals such as Genome Analysis: A Laboratory Manual Series (Vols. I-IV), Using Antibodies: A

Laboratory Manual, Cells: A Laboratory Manual, PCR Primer: A Laboratory Manual, and Molecular Cloning: A Laboratory Manual (all from Cold Spring Harbor Laboratory Press), Stryer, L. (1995) Biochemistry (4th Ed.) Freeman, Highly stabilized York, Gait,

"Oligonucleotide Synthesis: A Practical Approach" 1984, IRL Press, London, Nelson and Cox (2000), Lehninger, Principles of Biochemistry 3rd Ed., W. H. Freeman Pub., Highly stabilized York, N.Y. and Berg et al. (2002) Biochemistry, 5th Ed., W. H. Freeman Pub., Highly stabilized York, N.Y., all of which are herein incorporated in their entirety by reference for all purposes.

[00134] Note that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a polymerase" refers to one agent or mixtures of such agents, and reference to "the method" includes reference to equivalent steps and methods known to those skilled in the art, and so forth.

[00135] Note that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a polymerase" refers to one agent or mixtures of such agents, and reference to "the method" includes reference to equivalent steps and methods known to those skilled in the art, and so forth.

[00136] 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. All publications mentioned herein are incorporated herein by reference for the purpose of describing and disclosing devices, compositions, formulations and methodologies which are described in the publication and which might be used in connection with the presently described invention.

[00137] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention.

[00138] In the above description, numerous specific details are set forth to provide a more thorough understanding of the present invention. However, it will be apparent to one of skill in the art that the present invention may be practiced without one or more of these specific details. In other instances, well-known features and procedures well known to those skilled in the art have not been described in order to avoid obscuring the invention.

[00139] Although the present invention is described primarily with reference to specific embodiments, it is also envisioned that other embodiments will become apparent to those skilled in the art upon reading the present disclosure, and it is intended that such

embodiments be contained within the present inventive method. Lyophilized VWF Formulations

[00140] The present method also provides formulations of rVWF for use in the treatment methods provided herein. In some embodiments, the rVWF composition is used for the production of a pharmaceutical composition. In some embodiments, the rVWF can be formulated into a lyophilized formulation.

[00141] In some embodiments, the formulations comprising a VWF polypeptide of the invention are lyophilized after purification and prior to administration to a subject.

Lyophilization is carried out using techniques common in the art and should be optimized for the composition being developed (Tang et al , Pharm Res. 21 : 191-200, (2004) and Chang et al , Pharm Res. 13:243-9 (1996)).

[00142] A lyophilization cycle is, in one aspect, composed of three steps: freezing, primary drying, and secondary drying (A. P. Mackenzie, Phil Trans R Soc London, Ser B, Biol 278: 167 (1977)). In the freezing step, the solution is cooled to initiate ice formation.

Furthermore, this step induces the crystallization of the bulking agent. The ice sublimes in the primary drying stage, which is conducted by reducing chamber pressure below the vapor pressure of the ice, using a vacuum and introducing heat to promote sublimation. Finally, adsorbed or bound water is removed at the secondary drying stage under reduced chamber pressure and at an elevated shelf temperature. The process produces a material known as a lyophilized cake. Thereafter the cake can be reconstituted with either sterile water or suitable diluent for injection.

[00143] The lyophilization cycle not only determines the final physical state of excipients but also affects other parameters such as reconstitution time, appearance, stability and final moisture content. The composition structure in the frozen state proceeds through several transitions (e.g., glass transitions, wettings, and crystallizations) that occur at specific temperatures and the structure may be used to understand and optimize the lyophilization process. The glass transition temperature (Tg and/or Tg') can provide information about the physical state of a solute and can be determined by differential scanning calorimetry (DSC). Tg and Tg' are an important parameter that must be taken into account when designing the lyophilization cycle. For example, Tg' is important for primary drying. Furthermore, in the dried state, the glass transition temperature provides information on the storage temperature of the final product. Ιλ Pharmaceutical Formulations and Excipients in General

[00144] Excipients are additives that either impart or enhance the stability and delivery of a drug product (e.g. , protein). Regardless of the reason for their inclusion, excipients are an integral component of a formulation and therefore need to be safe and well tolerated by patients. For protein drugs, the choice of excipients is particularly important because they can affect both efficacy and immunogenicity of the drug. Hence, protein formulations need to be developed with appropriate selection of excipients that afford suitable stability, safety, and marketability.

[00145] A lyophilized formulation is, in one aspect, at least comprised of one or more of a buffer, a bulking agent, and a stabilizer. In this aspect, the utility of a surfactant is evaluated and selected in cases where aggregation during the lyophilization step or during

reconstitution becomes an issue. An appropriate buffering agent is included to maintain the formulation within stable zones of pH during lyophilization. A comparison of the excipient components contemplated for liquid and lyophilized protein formulations is provided in Table 10.

Table 1 : Excipient components of lyophilized protein formulations

[00146] The principal challenge in developing formulations for proteins is stabilizing the product against the stresses of manufacturing, shipping and storage. The role of formulation excipients is to provide stabilization against these stresses. Excipients are also be employed to reduce viscosity of high concentration protein formulations in order to enable their delivery and enhance patient convenience. In general, excipients can be classified on the basis of the mechanisms by which they stabilize proteins against various chemical and physical stresses. Some excipients are used to alleviate the effects of a specific stress or to regulate a particular susceptibility of a specific protein. Other excipients have more general effects on the physical and covalent stabilities of proteins. The excipients described herein are organized either by their chemical type or their functional role in formulations. Brief descriptions of the modes of stabilization are provided when discussing each excipient type.

[00147] Given the teachings and guidance provided herein, those skilled in the art will know what amount or range of excipient can be included in any particular formulation to achieve a biopharmaceutical formulation of the invention that promotes retention in stability of the biopharmaceutical (e.g. , a protein). For example, the amount and type of a salt to be included in a biopharmaceutical formulation of the invention is selected based on the desired osmolality (e.g. , isotonic, hypotonic or hypertonic) of the final solution as well as the amounts and osmolality of other components to be included in the formulation.

[00148] By way of example, inclusion of about 5% sorbitol can achieve isotonicity while about 9% of a sucrose excipient is needed to achieve isotonicity. Selection of the amount or range of concentrations of one or more excipients that can be included within a

biopharmaceutical formulation of the invention has been exemplified above by reference to salts, polyols and sugars. However, those skilled in the art will understand that the considerations described herein and further exemplified by reference to specific excipients are equally applicable to all types and combinations of excipients including, for example, salts, amino acids, other tonicity agents, surfactants, stabilizers, bulking agents,

cryoprotectants, lyoprotectants, anti-oxidants, metal ions, chelating agents and/or preservatives.

[00149] Further, where a particular excipient is reported in molar concentration, those skilled in the art will recognize that the equivalent percent (%) w/v (e.g. , (grams of substance in a solution sample/mL of solution) X 100%) of solution is also contemplated.

[00150] Of course, a person having ordinary skill in the art would recognize that the concentrations of the excipients described herein share an interdependency within a particular formulation. By way of example, the concentration of a bulking agent may be lowered where, e.g. , there is a high protein concentration or where, e.g. , there is a high stabilizing agent concentration. In addition, a person having ordinary skill in the art would recognize that, in order to maintain the isotonicity of a particular formulation in which there is no bulking agent, the concentration of a stabilizing agent would be adjusted accordingly (e.g. , a "tonicifying" amount of stabilizer would be used). Common excipients are known in the art and can be found in Powell et al , Compendium of Excipients fir Parenteral Formulations (1998), PDA J. Pharm. Sci. Technology, 52:238-311. c Pharmaceutical Buffers and Buffering Agents

[00151] The stability of a pharmacologically active protein formulation is usually observed to be maximal in a narrow pH range. This pH range of optimal stability needs to be identified early during pre-formulation studies. Several approaches, such as accelerated stability studies and calorimetric screening studies, are useful in this endeavor (Remmele R.L. Jr., et al, Biochemistry, 38(16): 5241-7 (1999)). Once a formulation is finalized, the protein must be manufactured and maintained throughout its shelf-life. Hence, buffering agents are almost always employed to control pH in the formulation.

[00152] The buffer capacity of the buffering species is maximal at a pH equal to the pKa and decreases as pH increases or decreases away from this value. Ninety percent of the buffering capacity exists within one pH unit of its pKa. Buffer capacity also increases proportionally with increasing buffer concentration.

[00153] Several factors need to be considered when choosing a buffer. First and foremost, the buffer species and its concentration need to be defined based on its pKa and the desired formulation pH. Equally important is to ensure that the buffer is compatible with the protein and other formulation excipients, and does not catalyze any degradation reactions. A third important aspect to be considered is the sensation of stinging and irritation the buffer may induce upon administration. For example, citrate is known to cause stinging upon injection (Laursen T, et al , Basic Clin Pharmacol Toxicol, 98(2): 218-21 (2006)). The potential for stinging and irritation is greater for drugs that are administered via the subcutaneous (SC) or intramuscular (IM) routes, where the drug solution remains at the site for a relatively longer period of time than when administered by the IV route where the formulation gets diluted rapidly into the blood upon administration. For formulations that are administered by direct IV infusion, the total amount of buffer (and any other formulation component) needs to be monitored. One has to be particularly careful about potassium ions administered in the form of the potassium phosphate buffer, which can induce cardiovascular effects in a patient (Hollander-Rodriguez JC, et al, Am. Fam. Physician., 73(2): 283-90 (2006)).

[00154] Buffers for lyophilized formulations need additional consideration. Some buffers like sodium phosphate can crystallize out of the protein amorphous phase during freezing resulting in shifts in pH. Other common buffers such as acetate and imidazole may sublime or evaporate during the lyophilization process, thereby shifting the pH of formulation during lyophilization or after reconstitution. [00155] The buffer system present in the compositions is selected to be physiologically compatible and to maintain a desired pH of the pharmaceutical formulation. In one embodiment, the pH of the solution is between pH 2.0 and pH 12.0. For example, the pH of the solution may be 2.0, 2.3, 2.5, 2.7, 3.0, 3.3, 3.5, 3.7, 4.0, 4.3, 4.5, 4.7, 5.0, 5.3, 5.5, 5.7, 6.0, 6.3, 6.5, 6.7, 7.0, 7.3, 7.5, 7.7, 8.0, 8.3, 8.5, 8.7, 9.0, 9.3, 9.5, 9.7, 10.0, 10.3, 10.5, 10.7, 11.0, 11.3, 11.5, 11.7, or 12.0.

[00156] The pH buffering compound may be present in any amount suitable to maintain the pH of the formulation at a predetermined level. In one embodiment, the pH buffering concentration is between 0.1 mM and 500 mM (1 M). For example, it is contemplated that the pH buffering agent is at least 0.1, 0.5, 0.7, 0.8 0.9, 1.0, 1.2, 1.5, 1.7, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, 60, 70, 80, 90, 100, 200, or 500 mM.

[00157] Exemplary pH buffering agents used to buffer the formulation as set out herein include, but are not limited to organic acids, glycine, histidine, glutamate, succinate, phosphate, acetate, citrate, Tris, HEPES, and amino acids or mixtures of amino acids, including, but not limited to aspartate, histidine, and glycine. In one embodiment of the present invention, the buffering agent is citrate. cL Pharmaceutical Stabilizers and Bulking Agents

[00158] In one aspect of the present pharmaceutical formulations, a stabilizer (or a combination of stabilizers) is added to prevent or reduce storage-induced aggregation and chemical degradation. A hazy or turbid solution upon reconstitution indicates that the protein has precipitated or at least aggregated. The term "stabilizer" means an excipient capable of preventing aggregation or physical degradation, including chemical degradation (for example, autolysis, deamidation, oxidation, etc.) in an aqueous state. Stabilizers

contemplated include, but are not limited to, sucrose, trehalose, mannose, maltose, lactose, glucose, raffinose, cellobiose, gentiobiose, isomaltose, arabinose, glucosamine, fructose, mannitol, sorbitol, glycine, arginine HCL, poly-hydroxy compounds, including

polysaccharides such as dextran, starch, hydroxyethyl starch, cyclodextrins, N-methyl pyrollidene, cellulose and hyaluronic acid, sodium chloride, (Carpenter et al , Develop. Biol. Standard 74:225, (1991)). In the present formulations, the stabilizer is incorporated in a concentration of about 0.1, 0.5, 0.7, 0.8 0.9, 1.0, 1.2, 1.5, 1.7, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 700, 900, or 1000 mM. In one embodiment of the present invention, mannitol and trehalose are used as stabilizing agents.

[00159] If desired, the formulations also include appropriate amounts of bulking and osmolality regulating agents. Bulking agents include, for example and without limitation, mannitol, glycine, sucrose, polymers such as dextran, polyvinylpyrolidone,

carboxymethylcellulose, lactose, sorbitol, trehalose, or xylitol. In one embodiment, the bulking agent is mannitol. The bulking agent is incorporated in a concentration of about 0.1, 0.5, 0.7, 0.8 0.9, 1.0, 1.2, 1.5, 1.7, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 500, 700, 900, or 1000 mM. a Pharmaceutical Surfactants

[00160] Proteins have a high propensity to interact with surfaces making them susceptible to adsorption and denaturation at air-liquid, vial-liquid, and liquid-liquid (silicone oil) interfaces. This degradation pathway has been observed to be inversely dependent on protein concentration and results in either the formation of soluble and insoluble protein aggregates or the loss of protein from solution via adsorption to surfaces. In addition to container surface adsorption, surface-induced degradation is exacerbated with physical agitation, as would be experienced during shipping and handling of the product.

[00161] Surfactants are commonly used in protein formulations to prevent surface-induced degradation. Surfactants are amphipathic molecules with the capability of out-competing proteins for interfacial positions. Hydrophobic portions of the surfactant molecules occupy interfacial positions (e.g. , air/liquid), while hydrophilic portions of the molecules remain oriented towards the bulk solvent. At sufficient concentrations (typically around the detergent's critical micellar concentration), a surface layer of surfactant molecules serves to prevent protein molecules from adsorbing at the interface. Thereby, surface-induced degradation is minimized. Surfactants contemplated herein include, without limitation, fatty acid esters of sorbitan polyethoxylates, e.g., polysorbate 20 and polysorbate 80. The two differ only in the length of the aliphatic chain that imparts hydrophobic character to the molecules, C-12 and C-18, respectively. Accordingly, polysorbate-80 is more surface- active and has a lower critical micellar concentration than polysorbate-20.

[00162] Detergents can also affect the thermodynamic conformational stability of proteins. Here again, the effects of a given detergent excipient will be protein specific. For example, polysorbates have been shown to reduce the stability of some proteins and increase the stability of others. Detergent destabilization of proteins can be rationalized in terms of the hydrophobic tails of the detergent molecules that can engage in specific binding with partially or wholly unfolded protein states. These types of interactions could cause a shift in the conformational equilibrium towards the more expanded protein states (e.g. increasing the exposure of hydrophobic portions of the protein molecule in complement to binding polysorbate). Alternatively, if the protein native state exhibits some hydrophobic surfaces, detergent binding to the native state may stabilize that conformation.

[00163] Another aspect of polysorbates is that they are inherently susceptible to oxidative degradation. Often, as raw materials, they contain sufficient quantities of peroxides to cause oxidation of protein residue side-chains, especially methionine. The potential for oxidative damage arising from the addition of stabilizer emphasizes the point that the lowest effective concentrations of excipients should be used in formulations. For surfactants, the effective concentration for a given protein will depend on the mechanism of stabilization.

[00164] Surfactants are also added in appropriate amounts to prevent surface related aggregation phenomenon during freezing and drying (Chang, B, J. Pharm. Sci. 85: 1325,

(1996)). Thus, exemplary surfactants include, without limitation, anionic, cationic, nonionic, zwitterionic, and amphoteric surfactants including surfactants derived from naturally- occurring amino acids. Anionic surfactants include, but are not limited to, sodium lauryl sulfate, dioctyl sodium sulfo succinate and dioctyl sodium sulfonate, chenodeoxycholic acid, N-lauroylsarcosine sodium salt, lithium dodecyl sulfate, 1-octanesulfonic acid sodium salt, sodium cholate hydrate, sodium deoxycholate, and glycodeoxycholic acid sodium salt.

Cationic surfactants include, but are not limited to, benzalkonium chloride or benzethonium chloride, cetylpyridinium chloride monohydrate, and hexadecyltrimethylammonium bromide. Zwitterionic surfactants include, but are not limited to, CHAPS, CHAPSO, SB3-10, and SB3- 12. Non-ionic surfactants include, but are not limited to, digitonin, Triton X-100, Triton X- 114, TWEEN-20, and TWEEN-80. Surfactants also include, but are not limited to lauromacrogol 400, polyoxyl 40 stearate, poly oxy ethylene hydrogenated castor oil 10, 40, 50 and 60, glycerol monostearate, polysorbate 40, 60, 65 and 80, soy lecithin and other phospholipids such as dioleyl phosphatidyl choline (DOPC), dimyristoylphosphatidyl glycerol (DMPG), dimyristoylphosphatidyl choline (DMPC), and (dioleyl phosphatidyl glycerol) DOPG; sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose. Compositions comprising these surfactants, either individually or as a mixture in different ratios, are therefore further provided. In one embodiment of the present invention, the surfactant is TWEEN-80. In the present formulations, the surfactant is incorporated in a concentration of about 0.01 to about 0.5 g/L. In formulations provided, the surfactant concentration is 0.005, 0.01, 0.02, 0.03, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9 or 1.0 g/L. f Pharmaceutical Salts

[00165] Salts are often added to increase the ionic strength of the formulation, which can be important for protein solubility, physical stability, and isotonicity. Salts can affect the physical stability of proteins in a variety of ways. Ions can stabilize the native state of proteins by binding to charged residues on the protein's surface. Alternatively, salts can stabilize the denatured state by binding to peptide groups along the protein backbone (- CONH-). Salts can also stabilize the protein native conformation by shielding repulsive electrostatic interactions between residues within a protein molecule. Salts in protein formulations can also shield attractive electrostatic interactions between protein molecules that can lead to protein aggregation and insolubility. In formulations provided, the salt concentration is between 0.1, 1, 10, 20, 30, 40, 50, 80, 100, 120, 150, 200, 300, and 500 mM. g ^ Other Common Excipient Components: Pharmaceutical Amino Acids

[00166] Amino acids have found versatile use in protein formulations as buffers, bulking agents, stabilizers and antioxidants. Thus, in one aspect histidine and glutamic acid are employed to buffer protein formulations in the pH range of 5.5 - 6.5 and 4.0 - 5.5

respectively. The imidazole group of histidine has a pKa = 6.0 and the carboxyl group of glutamic acid side chain has a pKa of 4.3 which makes these amino acids suitable for buffering in their respective pH ranges. Glutamic acid is particularly useful in such cases. Histidine is commonly found in marketed protein formulations, and this amino acid provides an alternative to citrate, a buffer known to sting upon injection. Interestingly, histidine has also been reported to have a stabilizing effect, with respect to aggregation when used at high concentrations in both liquid and lyophilized presentations (Chen B, et al , Pharm Res., 20(12): 1952-60 (2003)). Histidine was also observed by others to reduce the viscosity of a high protein concentration formulation. However, in the same study, the authors observed increased aggregation and discoloration in histidine containing formulations during freeze- thaw studies of the antibody in stainless steel containers. Another note of caution with histidine is that it undergoes photo-oxidation in the presence of metal ions (Tomita M, et al, Biochemistry, 8(12): 5149-60 (1969)). The use of methionine as an antioxidant in formulations appears promising; it has been observed to be effective against a number of oxidative stresses (Lam XM, et al , J Pharm ScL, 86(11): 1250-5 (1997)).

[00167] In various aspects, formulations are provided which include one or more of the amino acids glycine, proline, serine, arginine and alanine have been shown to stabilize proteins by the mechanism of preferential exclusion. Glycine is also a commonly used bulking agent in lyophilized formulations. Arginine has been shown to be an effective agent in inhibiting aggregation and has been used in both liquid and lyophilized formulations.

In formulations provided, the amino acid concentration is between 0.1, 1, 10, 20, 30, 40, 50, 80, 100, 120, 150, 200, 300, and 500 mM. In one embodiment of the present invention, the amino acid is glycine.

L Other Common Excipient Components: Pharmaceutical Antioxidants

[00168] Oxidation of protein residues arises from a number of different sources. Beyond the addition of specific antioxidants, the prevention of oxidative protein damage involves the careful control of a number of factors throughout the manufacturing process and storage of the product such as atmospheric oxygen, temperature, light exposure, and chemical contamination. The invention therefore contemplates the use of the pharmaceutical antioxidants including, without limitation, reducing agents, oxygen/free-radical scavengers, or chelating agents. Antioxidants in therapeutic protein formulations are, in one aspect, water-soluble and remain active throughout the product shelf -life. Reducing agents and oxygen/free-radical scavengers work by ablating active oxygen species in solution. Chelating agents such as EDTA are effective by binding trace metal contaminants that promote free- radical formation. For example, EDTA was utilized in the liquid formulation of acidic fibroblast growth factor to inhibit the metal ion catalyzed oxidation of cysteine residues.

[00169] In addition to the effectiveness of various excipients to prevent protein oxidation, the potential for the antioxidants themselves to induce other covalent or physical changes to the protein is of concern. For example, reducing agents can cause disruption of

intramolecular disulfide linkages, which can lead to disulfide shuffling. In the presence of transition metal ions, ascorbic acid and EDTA have been shown to promote methionine oxidation in a number of proteins and peptides (Akers MJ, and Defelippis MR. Peptides and Proteins as Parenteral Solutions. In: Pharmaceutical Formulation Development of Peptides and Proteins. Sven Frokjaer, Lars Hovgaard, editors. Pharmaceutical Science. Taylor and Francis, UK (1999)); Fransson J.R., /. Pharm. Sci. 86(9): 4046-1050 (1997); Yin J, et al, Pharm Res., 21(12): 2377-83 (2004)). Sodium thiosulfate has been reported to reduce the levels of light and temperature induced methionine-oxidation in rhuMab HER2; however, the formation of a thiosulfate-protein adduct was also reported in this study (Lam XM, Yang JY, et al , J Pharm Sci. 86(11): 1250-5 (1997)). Selection of an appropriate antioxidant is made according to the specific stresses and sensitivities of the protein. Antioxidants contemplated in certain aspects include, without limitation, reducing agents and oxygen/free-radical scavengers, EDTA, and sodium thiosulfate.

L Other Common Excipient Components: Pharmaceutical Metal Ions

[00170] In general, transition metal ions are undesired in protein formulations because they can catalyze physical and chemical degradation reactions in proteins. However, specific metal ions are included in formulations when they are co-factors to proteins and in suspension formulations of proteins where they form coordination complexes (e.g. , zinc suspension of insulin). Recently, the use of magnesium ions (10 -120 mM) has been proposed to inhibit the isomerization of aspartic acid to isoaspartic acid (WO 2004039337).

[00171] Two examples where metal ions confer stability or increased activity in proteins are human deoxyribonuclease (rhDNase, Pulmozyme®), and Factor VIII. In the case of rhDNase, Ca +2 ions (up to 100 mM) increased the stability of the enzyme through a specific binding site (Chen B, et al , / Pharm Sci., 88(4): 477-82 (1999)). In fact, removal of calcium ions from the solution with EGTA caused an increase in deamidation and aggregation.

However, this effect was observed only with Ca +2 ions; other divalent cations Mg +2 , Mn +2 and Zn +2 were observed to destabilize rhDNase. Similar effects were observed in Factor VIII. Ca +2 and Sr +2 ions stabilized the protein while others like Mg +2 , Mn +2 and Zn +2 , Cu +2 and Fe +2 destabilized the enzyme (Fatouros, A., et al , Int. J. Pharm., 155, 121-131 (1997). In a separate study with Factor VIII, a significant increase in aggregation rate was observed in the presence of A1 +3 ions (Derrick TS, et al , I. Pharm. Sci., 93(10): 2549-57 (2004)). The authors note that other excipients like buffer salts are often contaminated with Al +3 ions and illustrate the need to use excipients of appropriate quality in formulated products.

Other Common Excipient Components: Pharmaceutical Preservatives

[00172] Preservatives are necessary when developing multi-use parenteral formulations that involve more than one extraction from the same container. Their primary function is to inhibit microbial growth and ensure product sterility throughout the shelf- life or term of use of the drug product. Commonly used preservatives include, without limitation, benzyl alcohol, phenol and m-cresol. Although preservatives have a long history of use, the development of protein formulations that includes preservatives can be challenging.

Preservatives almost always have a destabilizing effect (aggregation) on proteins, and this has become a major factor in limiting their use in multi-dose protein formulations (Roy S, et al. , J Pharm ScL, 94(2): 382-96 (2005)).

[00173] To date, most protein drugs have been formulated for single-use only. However, when multi-dose formulations are possible, they have the added advantage of enabling patient convenience, and increased marketability. A good example is that of human growth hormone (hGH) where the development of preserved formulations has led to commercialization of more convenient, multi-use injection pen presentations. At least four such pen devices containing preserved formulations of hGH are currently available on the market.

Norditropin® (liquid, Novo Nordisk), Nutropin AQ® (liquid, Genentech) & Genotropin (lyophilized - dual chamber cartridge, Pharmacia & Upjohn) contain phenol while

Somatrope® (Eli Lilly) is formulated with m-cresol.

[00174] Several aspects need to be considered during the formulation development of preserved dosage forms. The effective preservative concentration in the drug product must be optimized. This requires testing a given preservative in the dosage form with

concentration ranges that confer anti-microbial effectiveness without compromising protein stability. For example, three preservatives were successfully screened in the development of a liquid formulation for interleukin-1 receptor (Type I), using differential scanning calorimetry (DSC). The preservatives were rank ordered based on their impact on stability at concentrations commonly used in marketed products (Remmele RL Jr., et al, Pharm Res., 15(2): 200-8 (1998)).

[00175] Development of liquid formulations containing preservatives are more challenging than lyophilized formulations. Freeze-dried products can be lyophilized without the preservative and reconstituted with a preservative containing diluent at the time of use. This shortens the time for which a preservative is in contact with the protein significantly minimizing the associated stability risks. With liquid formulations, preservative effectiveness and stability have to be maintained over the entire product shelf-life (- 18 -24 months). An important point to note is that preservative effectiveness has to be demonstrated in the final formulation containing the active drug and all excipient components. [00176] Some preservatives can cause injection site reactions, which is another factor that needs consideration when choosing a preservative. In clinical trials that focused on the evaluation of preservatives and buffers in Norditropin, pain perception was observed to be lower in formulations containing phenol and benzyl alcohol as compared to a formulation containing m-cresol (Kappelgaard A.M., Horm Res. 62 Suppl 3:98-103 (2004)).

Interestingly, among the commonly used preservative, benzyl alcohol possesses anesthetic properties (Minogue SC, and Sun DA., AnesthAnalg., 100(3): 683-6 (2005)). In various aspects the use of preservatives provide a benefit that outweighs any side effects.

L Methods of Preparation of Pharmaceutical Formulations [00177] The present invention further contemplates methods for the preparation of pharmaceutical formulations.

[00178] The present methods further comprise one or more of the following steps: adding a stabilizing agent as described herein to said mixture prior to lyophilizing, adding at least one agent selected from a bulking agent, an osmolality regulating agent, and a surfactant, each of which as described herein, to said mixture prior to lyophilization.

[00179] The standard reconstitution practice for lyophilized material is to add back a volume of pure water or sterile water for injection (WFI) (typically equivalent to the volume removed during lyophilization), although dilute solutions of antibacterial agents are sometimes used in the production of pharmaceuticals for parenteral administration (Chen, Drug Development and Industrial Pharmacy, 18: 1311-1354 (1992)). Accordingly, methods are provided for preparation of reconstituted rVWF compositions comprising the step of adding a diluent to a lyophilized rVWF composition of the invention.

[00180] The lyophilized material may be reconstituted as an aqueous solution. A variety of aqueous carriers, e.g., sterile water for injection, water with preservatives for multi dose use, or water with appropriate amounts of surfactants (for example, an aqueous suspension that contains the active compound in admixture with excipients suitable for the manufacture of aqueous suspensions). In various aspects, such excipients are suspending agents, for example and without limitation, sodium carboxymethylcellulose, methylcellulose,

hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents are a naturally-occurring phosphatide, for example and without limitation, lecithin, or condensation products of an alkylene oxide with fatty acids, for example and without limitation, polyoxy ethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example and without limitation, heptadecaethyl-eneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example and without limitation, polyethylene sorbitan monooleate. In various aspects, the aqueous suspensions also contain one or more preservatives, for example and without limitation, ethyl, or n-propyl, p-hydroxybenzoate.

L Exemplary rVWF Formulation for Administration

[00181] In some embodiments, the present methods provide for an enhanced formulation that allows a final product with high potency (high rVWF concentration and enhanced long term stability) in order to reduce the volume for the treatment (100 IU/ml to 10000 IU/ml). In some embodiments, the rVWF concentration in the formulation for administration is about 100 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 500 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 1000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 2000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 3000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 4000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 5000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 6000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 7000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 8000 IU/ml to 10000 IU/ml. In some embodiments, the rVWF concentration in the formulation for administration is about 9000 IU/ml to 10000 IU/ml.

[00182] In some embodiments, the formulation for administration comprises one or more zwitterionic compounds, including for example, amino acids like Histidine, Glycine, Arginine. In some embodiments, the formulation for administration comprises a component with amphipathic characteristic having a minimum of one hydrophobic and one hydrophilic group, including for example polysorbate 80, octylpyranosid, dipeptides, and/or amphipathic peptides. In some embodiments, the formulation for administration comprises a non reducing sugar or sugar alcohol or disaccharides, including for example, sorbitol, mannitol, sucrose, or trehalose. In some embodiments, the formulation for administration comprises a nontoxic water soluble salt, including for example, sodium chloride, that results in a physiological osmolality. In some embodiments, the formulation for administration comprises a pH in a range from 6.0 to 8.0. In some embodiments, the formulation for administration comprises a pH of about 6.0, about 6.5, about 7, about 7.5 or about 8.0. In some embodiments, the formulation for administration comprises one or more bivalent cations that stabilize rVWF, including for example, Ca2+, Mg2+, Zn2+, Mn2+ and/or combinations thereof. In some embodiments, the formulation for administration comprises about 1 mM to about 50 mM Glycine, about 1 mM to about 50 mM Histidine, about zero to about 300 mM sodium chloride (e.g., less than 300 mM sodium), about 0.01 % to about 0.05% polysorbate 20 (or polysorbate 80), and about 0.5 % to about 20% (w/w) sucrose with a pH of about 7.0 and having a physiological osmolarity at the time point of administration.

[00183] In some embodiments, the formulation for administration can be freeze dried. In some embodiments, the formulation for administration is stable and can be stored in liquid state at about 2°C to about 8°C, as well as at about 18°C to about 25°C. In some

embodiments, the formulation for administration is stable and can be stored in liquid state at about 2°C to about 8°C. In some embodiments, the formulation for administration is stable and can be stored in liquid state at about 18°C to about 25°C.

V. Administration of rVWF for Methods of Treating GI Bleeding in Patient with Severe V A I)

[00184] One of the advantages of administering rVWF to subjects with severe VWD to treat GI bleeding episodes is that the higher specific activity of rVWF as compared to pdVWF allows flexibility in the amount of rVWF administered and the number of times the subject is re-dosed. As will be appreciated and as is discussed in further detail herein, the coadministered FVIII may be recombinant or plasma derived.

[00185] To administer compositions to human or test animals, in one aspect, the compositions comprises one or more pharmaceutically acceptable carriers. The phrases "pharmaceutically" or "pharmacologically" acceptable refer to molecular entities and compositions that are stable, inhibit protein degradation such as aggregation and cleavage products, and in addition do not produce allergic, or other adverse reactions when administered using routes well-known in the art, as described below. "Pharmaceutically acceptable carriers" include any and all clinically useful solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like, including those agents disclosed above.

[00186] The pharmaceutical formulations are administered orally, topically, transdermally, parenterally, by inhalation spray, vaginally, rectally, or by intracranial injection. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques. Administration by intravenous, intradermal, intramusclar, intramammary, intraperitoneal, intrathecal, retrobulbar, and/or intrapulmonary injection at a particular site is contemplated as well. Generally, compositions are essentially free of pyrogens, as well as other impurities that could be harmful to the recipient.

[00187] Single or multiple administrations of rVWF are carried out with the dose levels and pattern being selected by the treating physician. For the prevention or treatment of disease, the appropriate dosage depends on the type of disease to be treated (e.g., von Willebrand disease), the severity and course of the disease, whether drug is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the drug, and the discretion of the attending physician.

[00188] In some aspects, rVWF is administered to a subject at a range from 40-100 IU/kg, e.g., 40, 45, 50, 55, 60, 65, 70, 75, 80, 90, 100, 40-100, 40-80, 50-80, 60-80, 70-80, 40-50, 40-60, 40-70, 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 IU/kg. In some embodiments, rVWF is administered at least once during a GI bleeding episode. In other embodiments, rVWF is administered two or more times, e.g., 2, 3, 4, 5, or more times, during a GI bleeding episode. In some instances, the subject is administered one or more infusions of rVWF. Each infusion can include a range from about 40-80 IU/kg rVWF, e.g., 40, 45, 50, 55, 60, 65, 70, 75, 80, 40-80, 50-80, 60-80, 70-80, 40-50, 40-60, 40-70, 40-50, 50-60, or 60-70 IU/kg rVWF. In some embodiments, the infusions can be substantially equal in amount. For instance, a first infusion and a second infusion can be substantially equal in amount. In some embodiments, the total dose of rVWF administered to the subject per bleeding episode is about 40-150 IU/kg, e.g., 40-150, 40-125, 40-100, 40-90, 40-75, 50-150, 50-100, 75-150, or 100-150 IU/kg.

[00189] In some embodiments, for minor and moderate bleeing events only 1-2 infusions more than estimated were required to control that bleeing episode and no additional VWF- containing product was required. In some embodiments, for major bleeding events <1.5 times more infusions than estimated were required to control that bleeding episode and no additional VWF-containing product was required. In some embodiments, minor, moderate, and major bleeding events the actual number of infusions was less than or equal to the estimated number required to treat the bleeding event, and no additional VWF-containing product was required.

[00190] In some embodiments, rVWF is administered at least once a day, at least twice a day, every 8-12 hours, and the like. In some instances, rVWF is administered for a total of 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, and the like. In some embodiments, the rVWF is administered every 8 hours, every 9 hours, every 10 hours, every 11 hours, or every 12 hours. In some embodiments, rVWF is administered every 8 to 12 hours for about 3 days to about 7 days.

[00191] In some embodiments, recombinant Factor VIII (rFVIII) is also administered to the subject with severe VWD to treat a GI bleeding episode. In some cases, the treatment administered comprises rVWF and rFVIII. In other cases, the treatment administered does not include rFVIII. In some embodiments, rFVIII is administered to the subject at a range of about 10-70 IU/kg, e.g., 10-70, 10-60, 10-50, 10-40, 10-30, 10-20, 20-30, 30-40, 40-50, 50- 60, or 60-70 IU/kg. In some instances, rFVIII is administered in the initial (first) dose or initial (first) infusion. In some cases, rFVIII is not administered as part of a second dose or second infusion. In some embodiments, a subject with VWD who is experiencing a GI bleeding episode is administered a single infusion of rVWF and rFVIII. In some

embodiments, the second administration of rVWF is not administerd with FVIII.

[00192] In some embodiments, of the method, when rVWF and FVIII are administered together, the rVWF to FVIII ratio is about 1.5:0.8. In some embodiments, of the method, when rVWF and FVIII are administered together, the rVWF to FVIII ratio is about 1.3: 1. In some embodiments, of the method, when rVWF and FVIII are administered together, the rVWF to FVIII ratio is about 1.1 :0.8. In some embodiments, of the method, when rVWF and FVIII are administered together, the rVWF to FVIII ratio is about 1.5: 1. In some embodiments, of the method, when rVWF and FVIII are administered together, the rVWF to FVIII ratio is about 1.1 : 1.2.

[00193] In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 40 IU/kg rVWF of the rVWF is administered when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 45 IU/kg rVWF of the rVWF is administered when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 50 IU/kg rVWF of the rVWF is administered when the gastrointestinal bleeding is minor or moderate

gastrointestinal bleeding. In some embodiments, about 55 IU/kg rVWF of the rVWF is administered when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 60 IU/kg rVWF of the rVWF is administered when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, wherein the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 40 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 45 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 50 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 55 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 60 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 8 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate

gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 9 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 10 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 11 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 12 hours for about 3 days to about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 3 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 4 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 5 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some

embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 6 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, 40-60 IU/kg rVWF of the rVWF is administered every 8 to 12 hours for about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding. In some embodiments, about 40, about 45, about 50, about 55, or about 60 IU/kg rVWF of the rVWF is administered every about 8 hours, about 9 hours, about 10 hours, about 11 hours, or about 12 hours for about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days, when the gastrointestinal bleeding is minor or moderate gastrointestinal bleeding.

[00194] In some embodiments, about 40 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 45 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 50 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 55 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 60 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 65 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 70 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 75 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 80 IU/kg rVWF of said rVWF and when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 40 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 45 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 50 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 55 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 60 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 65 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 70 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 75 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 8 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 9 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 10 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 11 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 12 hours for about 3 days to about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 3 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 4 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 5 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 6 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, 40-80 IU/kg rVWF of said rVWF is administered every 8 to 12 hours for about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding. In some embodiments, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, or about 80 IU/kg rVWF of said rVWF is administered every about 8 hours, about 9 hours, about 10 hours, about 11 hours, or about 12 hours for about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days when the gastrointestinal bleeding is major or severe gastrointestinal bleeding.

[00195] Generally, Type 1 VWD is indicated by <30 IU/dL VWF:RCo, <30 IU/dL

VWF:Ag, low or normal FVIII, and >0.5-0.7 IU/dLVWF:RCo/VWF:Ag Ratio. Type 2A VWD is indicated by <30 IU/dL VWF:RCo, <30-200 IU/dL VWF: Ag, low or normal FVIII, and O.5-0.7 IU/dL VWF:RCo/VWF:Ag Ratio. Type 2B VWD is indicated by <30-200 IU/dL VWF:RCo, <30 IU/dL VWF:Ag, low or normal FVIII, and usually <0.5-0.7

IU/dLVWF:RCo/VWF:Ag Ratio. Type 2M VWD is indicated by <30 IU/dL VWF:RCo, <30-200 IU/dL VWF:Ag, low or normal FVIII, and O.5-0.7 IU/dL VWF:RCo/VWF:Ag Ratio. Type 2N VWD is indicated by 30-2000 IU/dL VWF:RCo, 30-200 IU/dL VWF:Ag, very low FVIII, and >0.5-0.7 IU/dLVWF:RCo/VWF:Ag Ratio. Type 3 VWD is indicated by <3 IU/dL VWF:RCo, <3 IU/dL VWF:Ag, extremely low (<10 IU/dL) FVIII, and the VWF:RCo/VWF:Ag Ratio is not applicable. Normal is indicated by 50-200 IU/dL

VWF:RCo, 50-200 IU/dL VWF:Ag, normal FVIII, and >0.5-0.7 IU/dLVWF:RCo/VWF:Ag Ratio. In some embodiments, the subject has Type 3 VWD. In some embodiments, the subject has severe type 1 VWD. In some embodiments, the subject has severe type 2 VWD.

[00196] In some embodiments, the subject had been treated for at least 1 bleeding event within the previous 12 months. In some embodiments, the subject had been treated for more than 1 bleeding event within the previous 12 months.

[00197] Generally, minor bleeding is characterized by Acute or subacute clinically overt bleeding that did not satisfy the criteria for major bleeding and led to hospital admission for bleeding, physician-guided medical or surgical treatment for bleeding, or a change in antithrombotic therapy (including study drugs) for bleeding (Aristotle clinical definition); All other bleeding (except major and ICH) (RE-LY clinical definition); Overt bleeding not meeting the criteria for major bleeding but requiring medical intervention, unscheduled contact (visit or telephone) with a physician, temporary interruption of study drug (i.e., delayed dosing), pain, or impairment of daily activities) Rocket-AF clinical definition);

Clinically relevant bleeding was defined as skin hematoma > 25 cm 2 , spontaneous nosebleed of > 5 minutes duration, macroscopic hematuria, spontaneous rectal bleeding, gingival bleeding for >5 minutes, any bleeding leading to hospitalization, any bleeding leading to transfusion < 2 U, or any other bleeding considered relevant by the investigator (Petro clinical definition); and/or CRNM (clinically relevant non- major bleeding) defined as acute or subacute, clinically overt, not major, and leading to hospital admission for bleeding, physician- guided medical or surgical treatment for bleeding, or a change in antithrombotic therapy as well as minor bleeding events defined as acute clinically overt events not meeting the criteria for either major or CRNM bleeding (Aristotle-J clinical definition). See, for example, Wells G, Coyle D, Cameron C, et al. Safety, Effectiveness, and Cost-Effectiveness of New Oral Anticoagulants Compared with Warfarin in Preventing Stroke and Other Cardiovascular Events in Patients with Atrial Fibrillation [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2012 Apr 9. 3, CLINICAL REVIEW.

Available on the World Wide Web at www.ncbi.nlm.nih.gov/books/NBK169813/. Minor bleeding can include events were defined as those not fulfilling the criteria of major or clinically significant bleeding; minor bleeding from a wound (bleeding at the injection site, epistaxis, or wound hematoma not requiring operative decompression); overt bleeding that did not meet the criteria for major hemorrhage and associated with >1 of the following: epistaxis lasting more than 5min or requiring intervention, ecchymosis or hematoma >5cm at its greatest dimension, hematuria not associated with urinary catheter related trauma, GI hemorrhage not related to intubation or placement of a NG tube, wound hematoma or complications, subconjunctival hemorrhage necessitating cessation of medication; minor bleeding in the GI or urinary tract and hematoma at the site of an injection; and/or overt bleeding not meeting the criteria for major hemorrhage. See, for example, Sobieraj DM, Coleman CI, Tongbram V, et al. Venous Thromboembolism Prophylaxis in Orthopedic

Surgery [Internet]. Rockville (MD): Agency for Healthcare Research and Quality (US); 2012 Mar. (Comparative Effectiveness Reviews, No. 49.) Appendix F, Additional Evidence Tables. Available from the World Wide Web at www.ncbi.nlm.nih.gov/books/NBK92309/. [00198] Generally major bleeding is characterized by International Society on Thrombosis and Haemostasis (ISTH) standards, and includes, any life threatening and/or fatal bleeding; symptomatic bleeding into a critical area or organ and major bleeding was separated into intracranial (intracerebral, subdural) and extracranial (GI, non-GI) bleeding (RE-LY clinical definition); symptomatic bleeding into a critical anatomic site (Rocket-AF clinical definition); Life-threatening retroperitoneal, intracranial, intraocular, or intraspinal bleeding; or bleeding requiring surgery (Artistotle-J clinical definition). Major bleeing events can include those where there is fall in hemoglobin at least 20g/L or transfusion of > 2 units of whole blood (packed cells mentioned in life-threatening bleed definition; RE-LY definition of life-threatening bleeding: > 1 of the following criteria: (1) fatal, symptomatic intracranial bleed; (2) reduction in hemoglobin level of at least 5.0 g/L; (3) transfusion of at least 4 U of blood or packed cells; (4) associated with hypotension requiring the use of intravenous inotropic agents; or (5) necessitated surgical intervention); fall in hemoglobin > 2g/dL or transfusion of > 2 units of whole blood/red cells (ISTH or Rocket-AF clinical definition); and/or bleeding requiring surgery or transfusion of >2 U or associated with a decrease in hemoglobin of > 2.0 g/L episodes. See, for example, Wells G, Coyle D, Cameron C, et al. Safety, Effectiveness, and Cost-Effectiveness of New Oral Anticoagulants Compared with Warfarin in Preventing Stroke and Other Cardiovascular Events in Patients with Atrial Fibrillation [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2012 Apr 9. 3, CLINICAL REVIEW. Available on the World Wide Web at

www.ncbi.nlm.nih.gov/books/NBK169813/. Major bleeding can include clinically overt bleeding associated with > 20 g/L fall in Hb; clinically overt leading to transfusion of >2U packed cells or whole blood; fatal, retroperitoneal, intracranial, intraocular or intraspinal bleeding; bleeding warranting treatment cessation or leading to reoperation; fatal, retroperitoneal, intracranial, or intraspinal bleeding; bleeding that involved any other critical organ; bleeding leading to reoperation; overt bleeding with a bleeding index > 2; major bleeding from a wound (wound hematoma requiring operative decompression), or major bleeding not related to a wound (gastrointestinal or intracerebral hemorrhage); clinically overt bleeding associated with either a decrease in Hb >2 g/dL or a need for a transfusion of >2U RBC; intracranial or retroperitoneal (resulted in the permanent discontinuation of anticoagulation). See, for example, Sobieraj DM, Coleman CI, Tongbram V, et al. Venous Thromboembolism Prophylaxis in Orthopedic Surgery [Internet]. Rockville (MD): Agency for Healthcare Research and Quality (US); 2012 Mar. (Comparative Effectiveness Reviews, No. 49.) Appendix F, Additional Evidence Tables. Available from: the World Wide Web at www.ncbi.nlm.nih.gov/books/NBK92309/.

[00199] Compositions of rVWF can be contained in pharmaceutical formulations, as described herein. Such formulations can be administered orally, topically, transdermally, parenterally, by inhalation spray, vaginally, rectally, or by intracranial injection. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques. Administration by intravenous, intradermal, intramuscular, intramammary, intraperitoneal, intrathecal, retrobulbar, intrapulmonary injection and or surgical implantation at a particular site is contemplated as well. Generally, compositions are essentially free of pyrogens, as well as other impurities that could be harmful to the recipient.

[00200] In one aspect, formulations of the invention are administered by an initial bolus followed by a continuous infusion to maintain therapeutic circulating levels of drug product. As another example, the inventive compound is administered as a one-time dose. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient. The route of administration can be, but is not limited to, by intravenous, intraperitoneal, subcutaneous, or intramuscular administration. The frequency of dosing depends on the pharmacokinetic parameters of the agents and the route of administration. The optimal pharmaceutical formulation is determined by one skilled in the art depending upon the route of administration and desired dosage. See for example, Remington's Pharmaceutical

Sciences, 18th Ed., 1990, Mack Publishing Co., Easton, Pa. 18042 pages 1435-1712, the disclosure of which is hereby incorporated by reference in its entirety for all purposes and in particular for all teachings related to formulations, routes of administration and dosages for pharmaceutical products. Such formulations influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agents. Depending on the route of administration, a suitable dose is calculated according to body weight, body surface area or organ size. Appropriate dosages may be ascertained through use of established assays for determining blood level dosages in conjunction with appropriate dose-response data. The final dosage regimen is determined by the attending physician, considering various factors which modify the action of drugs, e.g. the drug's specific activity, the severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. By way of example, a typical dose of a recombinant VWF of the present invention is approximately 50 IU/kg, equal to 500 μg/kg. As studies are conducted, further information will emerge regarding the appropriate dosage levels and duration of treatment for various diseases and conditions.

[00201] The practice of the present invention may employ, unless otherwise indicated, conventional techniques and descriptions of organic chemistry, polymer technology, molecular biology (including recombinant techniques), cell biology, biochemistry, and immunology, which are within the skill of the art. Such conventional techniques include polymer array synthesis, hybridization, ligation, and detection of hybridization using a label. Specific illustrations of suitable techniques can be had by reference to the example herein below. However, other equivalent conventional procedures can, of course, also be used. Such conventional techniques and descriptions can be found in standard laboratory manuals such as Genome Analysis: A Laboratory Manual Series (Vols. I-IV), Using Antibodies: A Laboratory Manual, Cells: A Laboratory Manual, PCR Primer: A Laboratory Manual, and Molecular Cloning: A Laboratory Manual (all from Cold Spring Harbor Laboratory Press), Stryer, L. (1995) Biochemistry (4th Ed.) Freeman, Highly stabilized York, Gait,

"Oligonucleotide Synthesis: A Practical Approach" 1984, IRL Press, London, Nelson and Cox (2000), Lehninger, Principles of Biochemistry 3rd Ed., W. H. Freeman Pub., Highly stabilized York, N.Y. and Berg et al. (2002) Biochemistry, 5th Ed., W. H. Freeman Pub., Highly stabilized York, N.Y., all of which are herein incorporated in their entirety by reference for all purposes.

[00202] Note that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a polymerase" refers to one agent or mixtures of such agents, and reference to "the method" includes reference to equivalent steps and methods known to those skilled in the art, and so forth.

[00203] Note that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a polymerase" refers to one agent or mixtures of such agents, and reference to "the method" includes reference to equivalent steps and methods known to those skilled in the art, and so forth. [00204] 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. All publications mentioned herein are incorporated herein by reference for the purpose of describing and disclosing devices, compositions, formulations and methodologies which are described in the publication and which might be used in connection with the presently described invention.

[00205] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges is also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention.

[00206] In the above description, numerous specific details are set forth to provide a more thorough understanding of the present invention. However, it will be apparent to one of skill in the art that the present invention may be practiced without one or more of these specific details. In other instances, well-known features and procedures well known to those skilled in the art have not been described in order to avoid obscuring the invention.

[00207] Although the present invention is described primarily with reference to specific embodiments, it is also envisioned that other embodiments will become apparent to those skilled in the art upon reading the present disclosure, and it is intended that such

embodiments be contained within the present inventive method.

EXAMPLES EXAMPLE 1 : TREATMENT OF GI BLEEDING EPISODES WITH RECOMBINANT VON WILLEBRAND FACTOR IN PATIENTS WITH SEVERE VON WILLEBRAND DISEASE: SUBANALYSIS FROM PIVOTAL PHASE 3 ON-DEMAND STUDY

INTRODUCTION

[00208] Gastrointestinal (GI) bleeding events occur in up to 20% of patients with von Willebrand disease (VWD) and have been observed in association with angiodysplastic lesions in 2%-4% of patients with VWD (1-3). GI bleeds are closely associated with the absence of higher molecular weight and ultra-large multimers (ULMs) of von Willebrand factor (VWF), which are most often seen in patients with type 2A and type 3 VWD (4). Higher doses and longer durations of therapy with plasma-derived VWF replacement concentrates are usually needed to resolve GI bleeds compared with bleeds at other sites, and treatment may still be unsuccessful (5). VONVENDI (von Willebrand factor [recombinant], Baxalta US Inc., Westlake Village, CA) is a recombinant VWF (rVWF) concentrate in which ULMs, the most hemostatically effective VWF multimers, are preserved because they are not exposed to ADAMTS13 during manufacturing (6).

OBJECTIVES

[00209] The pivotal phase 3 clinical trial of rVWF evaluated its efficacy and safety with and without recombinant factor VIII (rFVIII) (ADVATE [antihemophilic factor

(recombinant)], Baxalta US Inc., Westlake Village, CA) for the treatment of bleeds in patients with severe VWD (7). This subanalysis was performed using data from patients who experienced GI bleeding events during participation in the pivotal clinical trial. METHODS

[00210] Phase 3, prospective, randomized clinical trial (NCT01410227) to assess patient demographics, GI bleed characteristics, hemostatic efficacy, timing of treatment and bleeding resolution, and dosages of rVWF ± rFVIII. The study population included men and women aged 18-65 y, who had type 3 or severe type 1 or 2 VWD and had been treated for >1 bleeding event within 12 mo before enrollment. On-demand treatment of bleeds:

Minor/moderate bleeds: 40-60 IU/kg rVWF; Major/severe bleeds: up to 80 IU/kg rVWF every 8-12 h for 3-7 d.

[00211] Initial dose of rVWF was coadministered with rFVIII at a ratio of 1.3: 1 ± 0.2 rVWF:rFVIII. rVWF was administered alone thereafter provided hemostatic FVIILC levels were achieved.

[00212] Hemostatic efficacy was rated on a 4-point scale (none = 4, moderate = 3, good = 2, excellent = 1).

[00213] Adverse events were monitored throughout the study. RESULTS

[00214] A total of 192 bleeding events were treated with rVWF and assessed for hemostatic efficacy during the study; hemostatic efficacy was rated as either excellent (96.9%) or good (3.1%) in each case. 4 patients with type 3 VWD and a median age of 32.5 y experienced a total of 6 GI bleeding events (Table 1).

Table 1. Patient Demographics in GI Bleed Subgroup

Patient Age, y Weight, kg Sex VWD Type GI Bleeds

During

Study, n

1 26 72 Male 3 1

2 42 85 Male 3 2

3 37 85 Female 3 2

4 28 77 Female 3 1

GI=gastrointestinal; VWD=von Willebrand disease.

Table 2. Bleed Characteristics and Efficacy in GI Bleed Subgroup

Patient Bleeds Severity of Days to Clinical Infusions Time to

Treated GI Bleeds Treatment* Efficacy to Resolution†, During n Rating Resolution, h

Study, n n

1 4 Major/Severe 0 Excellent 1 Unknown

2 6 Moderate 3 Excellent 1 1.8

2 6 Moderate 7 Excellent 1 2.7

3 2 Minor 3 Excellent 1 18.6

3 2 Minor 0 Good 2 Unknown

4 1 Major/Severe 3 Excellent 2 14.0

GI=gastrointestinal .

*Days from bleeding onset to first infusion.

†Time from first infusion of rVWF to resolution of bleeding episode

Table 3. rVWF and rFVIII Use in GI Bleed Subgroup Patient Severity of GI Infusion 1 Infusion Hemostatic Duration Bleeds 2 Efficacy Between

Rating rVWF

Infusions*, rVWF, rFVIII, rVWF, h

lU/kg lU/kg lU/kg

1 Major/Severe 57.5 41.5 Excellent N/A

2 Moderate 60.1 49.4 - Excellent N/A

2 Moderate 59.9 46.0 - Excellent N/A

3 Minor 53.6 19.4 - Excellent N/A

3 Minor 53.5 19.3 53.5 Good 50.6

4 Major/Severe 60.5 25.0 60.5 Excellent 22.1

GI=gastrointestinal; rFVIII=recombinant factor VIII; rVWF=recombinant von Willebrand factor.

" ime from end of rVWF infusion 1 to start of rVWF infusion 2. Table 4. Adverse Events in GI Bleed Subgroup

Adverse Events Patients, n Events, n

Total 4 28

Nonserious 4 26

Not related 4 23

Possibly related 1 3*

Serious 2 2

Not related 2 2

Possibly related 0 0

GI=gastrointestinal .

Tachycardia, dysgeusia, and infusion site paresthesia.

[00215] Of the 6 GI bleeds, 2 each were reported as minor, moderate, and major/severe (Table 2). 67% of GI bleeds (4/6) required only 1 infusion of rVWF to successfully treat the bleed; 33% of GI bleeds (2/6) required 2 infusions to achieve hemostasis. Median time to resolution, which was known for 4/6 bleeds, was 8.3 h (range, 1.8-18.6 h). 100% of GI bleeds treated with rVWF had a hemostatic efficacy rating of excellent (83% [5/6]) or good (17% [1/6]; Figure 1).

[00216] The 4 patients with GI bleeds experienced a total of 28 adverse events (Table 4). 3 possibly related nonserious adverse events occurred in 1 patient (tachycardia, dysgeusia, and infusion site paresthesia). Serious adverse events included GI hemorrhage and constipation in 1 patient each, and neither event was considered related to study drug.

[00217] The GI bleed resulted from 2 chronic ulcers with evidence of a recent hemorrhage and, per protocol, was considered a serious adverse event because the investigator thought it would have also occurred in a healthy individual under the same circumstances. rVWF and rFVIII use are shown in Table 3. The median dose per infusion was 58.7 IU/kg (range, 53.5- 60.5 IU/kg) for rVWF and 33.3 IU/kg (range, 19.3-49.4 IU/kg) for rFVIII. The median total dose per bleed was 60.0 IU/kg (range, 53.6-121.0 IU/kg) for rVWF and 33.3 IU/kg (range, 19.3-49.4 IU/kg) for rFVIII.

CONCLUSIONS

[00218] In this subanalysis of the pivotal phase 3 clinical trial, rVWF was safe and effective for the on-demand treatment of GI bleeds in patients with severe VWD.

[00219] Of 6 GI bleeds (2 minor, 2 moderate, 2 major/severe), hemostatic efficacy was rated as excellent for 5 (83%) and good for 1 (17%). A single infusion of rVWF was successful in treating 4 (67%) of the GI bleeds (1 minor, 2 moderate, 1 major/severe). Time to resolution of the GI bleeds was available for 4 patients and ranged from 1.8-18.6 h (median, 8.3 h).

[00220] These findings from a small cohort of patients warrant further assessment of the role of rVWF in the treatment of GI bleeding and angiodysplasia in a larger population of patients with VWD.

[00221] The emerging association between angiodysplasia and a lack of higher molecular weight VWF multimers suggests that rVWF, with its higher ULM content, may be of particular benefit in this patient population (8,9).

REFERENCES

1. Randi AM. Thromb Res. 2016;141 Suppl 2: S55-58.

2. Randi AM and Laffan MA. J Thromb Haemost. 2017;15(1): 13-20.

3. Franchini M and Mannucci PM. Thromb Haemost. 2014; 112(3): 427-431.

4. Franchini M and Mannucci PM. Br J Haematol. 2013;161(2): 177-182.

5. Berntorp E, et al. Haemophilia. 2009;15(1): 122-130.

6. Turecek PL, et al. Hamostaseologie. 2009;29(suppl l): S32-38.

7. Gill JC, et al. Blood. 2015;126(17):2038-2046.

8. Selvam S and James P. Semin Thromb Hemost. 2017.

9. Franchini M and Mannucci PM. Expert Rev Hematol. 2016;9(9): 825-830.

EXAMPLE 2: Pharmacokinetics, Safety, and efficacy of recombinant von Willebrand Factor (rVWF) in the Treatment of Bleeding BRIEF SUMMARY:

[00222] The purpose of this Phase 3 study is to assess the pharmacokinetics of rVWF:rFVIII and rVWF, and to assess the safety and efficacy of rVWF:rFVIII and rVWF in the treatment of bleeding events in subjects with severe hereditary von Willebrand disease (VWD).

Experimental: PK 50 Biological: Recombinant von Willebrand factor (rVWF)

Only Arm (minimum of 7 Intravenous administration

subjects with type 3 Other Names:

VWD)

PK assessment (50 IU/kg . BAX 111

rVWF) only, no treatment . rVWF

of bleeding episodes

Drug: Placebo

Syringe supplied with physiologic saline solution for infusion

Other Names:

• saline

• physiologic saline

Biological: Recombinant factor Villi (rFVIII)

Intravenous administration

Other Names:

. rFVIII

. ADVATE

Experimental: Treatment Biological: Recombinant von Willebrand factor (rVWF)

Only (up to 7 subjects Intravenous administration

independent of VWD Other Names:

subtype)

Treatment of bleeding . BAX 111

episodes for a total of 12 . rVWF

months •

Biological: Recombinant factor Villi (rFVIII)

Intravenous administration

Other Names:

. rFVIII

. ADVATE

PRIMARY OUTCOME MEASURES

[00223] Primary Outcome Measures: Primary Outcome #1

[00224] Percentage of Participants With Treatment Success for Treated Bleeding Episodes [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF] [00225] Treatment success was defined as the extent of control of bleeding episodes (BEs) using a mean efficacy rating score of <2.5 for a participant's BEs treated with study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII

[rFVIII]) during the study period. Scores used: Excellent = 1 - actual infusions < estimated number of infusions required to treat BE; no additional VWF required (all BEs); Good = 2 - >l-2 infusions (minor/moderate BEs) or < 1.5 infusions (major BEs) greater than estimated required to control BE; no additional VWF required (all BEs); Moderate = 3 > 3 infusions (minor/moderate BEs) or > 1.5 infusions (major BEs) greater than estimated required to control BE; no additional VWF required (all BEs); None = 4 - severe uncontrolled bleeding or intensity of bleeding not changed; additional VWF required. Included participants with available primary efficacy rating (prospective-excluding gastrointestinal bleeds) in the Full Analysis Set.

SECONDARY OUTCOME MEASURES

Secondary Outcome Measures: Secondary Outcome #1

[00226] Percentage of Treated Bleeding Episodes With an Efficacy Rating of "Excellent" or "Good" [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

[00227] Efficacy ratings "excellent" or "good" for the control of bleeding episodes (BEs) with study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) are defined as follows: Excellent - actual infusions < estimated number of infusions required to treat BE; no additional von Willebrand Factor (VWF) required (all BEs); Good - >l-2 infusions (minor/moderate BEs) or <1.5 infusions (major BEs) greater than estimated required to control BE; no additional VWF required (all BEs). The data set included prospectively estimated BEs treated with study product with an available efficacy rating from participants in the Full Analysis Set

Secondary Outcome Measures: Secondary Outcome #2

[00228] Percentage of Treated Bleeding Episodes With an Efficacy Rating of "Excellent" or "Good", Excluding Gastrointestinal Bleeds [Time Frame: For 12 months after first infusion of rVWF: rFVIII or rVWF]

[00229] Efficacy ratings of "excellent" or "good" for the control of bleeding episodes (BEs) with study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) are defined as follows: Excellent - actual infusions < estimated number of infusions required to treat BE; no additional von Willebrand Factor (VWF) required (all BEs); Good - >l-2 infusions (minor/moderate BEs) or <1.5 infusions (major BEs) greater than estimated required to control BE; no additional VWF required (all BEs). The data set included prospectively estimated BEs excluding gastrointestinal (GI) bleeds treated with study product with an available efficacy rating from participants in the Full Analysis Set.

Secondary Outcome Measures: Secondary Outcome #3

[00230] Number of Infusions of rVWF:rFVIII and/or rVWF Per Bleeding Episode

[Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

[00231] The actual number of infusions of recombinant von Willebrand factor: recombinant factor VIII (rVWF: rFVIII) and/or rVWF required to treat a bleeding episode (BE). BEs were to be initially treated with an infusion of rVWF: rFVIII and subsequently with rVWF with or without rFVIII, based on FVIII levels, if available. In cases, where no FVIII levels were available, the individual participant's PK data was used to determine the rFVIII dose. The data set included prospectively estimated BEs treated with study product with an available efficacy rating from participants in the Full Analysis Set.

Secondary Outcome Measures: Secondary Outcome #4

[00232] Number of Units of rVWF: rFVIII and/or rVWF Per Bleeding Episode

[Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

[00233] The number of units is provided as the actual dose [IU/kg] of recombinant von Willebrand factor: recombinant factor VIII (rVWF:rFVIII) and/or rVWF required to treat a bleeding episode (BE). BEs were to be initially treated with an infusion of rVWF:rFVIII and subsequently with rVWF with or without rFVIII, based on FVIII levels, if available. In cases, where no FVIII levels were available, the individual participant's PK data was used to determine the rFVIII dose. The data set included prospectively estimated BEs treated with study product of known lot number with an available efficacy rating from participants in the Full Analysis Set.

Secondary Outcome Measures: Secondary Outcome #5

[00234] Percentage of Participants Who Develop Inhibitory Antibodies to FVIII

[Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF].

[00235] Development of neutralizing antibodies (inhibitors) to factor VIII (FVIII) was assessed by the Nijmegen modification of the Bethesda assay. Positive FVIII inhibitor tests were defined as > 0.4 Bethesda units/mL (BU/mL) by the Nijmegen- modified Bethesda assay that is confirmed by a second test performed on an independent sample obtained 2-4 weeks following the first test. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #6

[00236] Percentage of Participants Who Develop Inhibitory Antibodies to VWF

[Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF].

[00237] Neutralizing antibodies (inhibitors) to Von Willebrand Factor Ristocetin cofactor (VWF:RCo), VWF collagen binding (VWF:CB) and VWF Factor VIII binding

(VWF:FVIIIB) activities were measured using Nijmegen modification of the Bethesda assay.

One Bethesda Unit (BU) is thereby defined as the amount of inhibitor that decreased the measured activity in the assays to 50% of that of the negative control samples. The assays were validated using human plasma samples from two type 3 VWD patients with low (1-2 BU/mL) and high (-10 BU/mL) titer inhibitors and plasma samples from non-human primates immunized with human rVWF (>100 BU/mL). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #7

[00238] Percentage of Participants Who Develop Binding Antibodies to VWF

[Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

[00239] The presence of total binding anti-VWF antibodies was determined by an enzyme- linked immunosorbent assay (ELISA) employing polyclonal anti-human immunoglobulin (Ig) antibodies (IgG, IgM and IgA). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #8

[00240] Percentage of Participants Who Develop Binding Antibodies to CHO

[Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF].

[00241] The presence of total binding anti-CHO antibodies was determined by measuring total immunoglobulin (Ig) antibodies (IgG, IgA, IgM) against Chinese Hamster Ovary (CHO) protein using an enzyme-linked immunosorbent assay (ELISA). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #9

[00242] Percentage of Participants Who Develop Binding Antibodies to rFurin

[Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF].

[00243] The presence of total binding anti-rFurin antibodies was determined by measuring total immunoglobulin (Ig) antibodies (IgG, IgA, IgM) against rFurin protein using an enzyme-linked immunosorbent assay (ELISA). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #10

[00244] Percentage of Participants Who Develop Binding Antibodies to Mouse

Immunoglobulin [Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF].

[00245] The presence of total binding anti-Murine immunoglobulin (IgG) antibodies was determined using an enzyme-linked immunosorbent assay (ELISA). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #11

[00246] Percentage of Participants Who Had an Occurrence of Thrombotic Events

[Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

Secondary Outcome Measures: Secondary Outcome #12

[00247] Number of Adverse Events Related to Study Product Including Clinically

Significant Changes in Laboratory Parameters and Vital Signs [Time Frame: For 12 months after first infusion of rVWF : rF VIII or rVWF] .

[00248] Adverse Events (AEs) related to study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) are described. Only laboratory parameters (hematology and clinical chemistry) and vital signs (physical examination, ECG) with clinically significant findings that are recorded as AEs are included. Categories presented as Severity-System Organ Class-Preferred Term Seriousness: serious adverse event (SAE); non serious adverse event (nsAE) System Organ Class: Cardiac disorders (CARD); General disorders and administration site conditions (GEN);

Investigations (INV); Nervous system disorders (NERV); Skin and subcutaneous tissue disorders (SKN); Vascular disorders (VAS). Category title includes number of AEs [N] for the category.

Secondary Outcome Measures: Secondary Outcome #13

[00249] Number of Participants With Adverse Events Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs [Time Frame: For 12 months after first infusion of rVWF: rFVIII or rVWF].

[00250] Number of participants with Adverse Events (AEs) related to study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII

[rFVIII]) are described. Only laboratory parameters (hematology and clinical chemistry) and vital signs (physical examination, ECG) with clinically significant findings that are recorded as AEs are included. Categories presented as Severity-System Organ Class-Preferred Term Seriousness: serious adverse event (SAE); non serious adverse event (nsAE) System Organ Class: Cardiac disorders (CARD); General disorders and administration site conditions (GEN); Investigations (INV); Nervous system disorders (NERV); Skin and subcutaneous tissue disorders (SKN); Vascular disorders (VAS).

Secondary Outcome Measures: Secondary Outcome #14

[00251] Number of Adverse Events by Infusion Related to Study Product Including

Clinically Significant Changes in Laboratory Parameters and Vital Signs [Time Frame: For 12 months after first infusion of rVWF: rFVIII or rVWF].

[00252] Adverse Events (AEs) by infusion related to study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) are described. Only laboratory parameters (hematology and clinical chemistry) and vital signs (physical examination, ECG) with clinically significant findings that are recorded as AEs are included. Categories presented as Severity-System Organ Class-Preferred Term Seriousness: serious adverse event (SAE); non serious adverse event (nsAE) System Organ Class: Cardiac disorders (CARD); General disorders and administration site conditions (GEN);

Investigations (INV); Nervous system disorders (NERV); Skin and subcutaneous tissue disorders (SKN); Vascular disorders (VAS).

Secondary Outcome Measures: Secondary Outcome #15 [00253] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00254] Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for subjects in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #16

[00255] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00256] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #17

[00257] PK50 - Mean Residence Time of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00258] Mean Residence Time (MRT) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #18

[00259] PK50 - Clearance of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00260] Clearance (CL) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #19

[00261] PK50 - Incremental Recovery of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00262] Incremental Recovery (IR) at the maximum plasma concentration of von

Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #20

[00263] PK50 - Elimination Phase Half-Life of VWF:Co [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00264] Elimination Phase Half-Life (Tl/2) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant FVIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #21

[00265] PK50 - Volume of Distribution at Steady State of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00266] Volume of Distribution at Steady State (Vss) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #22

[00267] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00268] Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von

Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #23

[00269] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout].

[00270] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von

Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #24

[00271] PK50 - Mean Residence Time of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00272] Mean Residence Time (MRT) of von Willebrand Factor Antigen (VWF: Ag) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII] or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #25

[00273] PK50 - Clearance of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] [00274] Clearance (CL) of von Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #26

[00275] PK50 - Incremental Recovery of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00276] Incremental Recovery (IR) at the maximum plasma concentration of von

Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von

Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #27

[00277] PK50 - Elimination Phase Half-Life of VWF: Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00278] Elimination Phase Half-Life (Tl/2) of von Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor

Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant FVIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category

Secondary Outcome Measures: Secondary Outcome #28 [00279] PK50 - Volume of Distribution at Steady State of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00280] Volume of Distribution at Steady State (Vss) of von Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #29

[00281] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00282] Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #30

[00283] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00284] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factor:von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #31

[00285] PK50 - Mean Residence Time of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00286] Mean Residence Time (MRT) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #32

[00287] PK50 - Clearance of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00288] Clearance (CL) of von Willebrand Factor Collagen Binding (VWF: CB) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #33

[00289] PK50 - Incremental Recovery of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00290] Incremental Recovery (IR) at the maximum plasma concentration of von

Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #34

[00291] PK50 - Elimination Phase Half-Life of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00292] Elimination Phase Half-Life (Tl/2) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant FVIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #35

[00293] PK50 - Volume of Distribution at Steady State of VWF: CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00294] Volume of Distribution at Steady State (Vss) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg

VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #36

[00295] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of FVIILC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00296] Area under the plasma concentration curve (AUC) from time 0 to infinity of Factor VIII activity (FVIIFC) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #37

[00297] PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00298] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of Factor VIII activity (FVIIFC) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #38

[00299] PK50 - Mean Residence Time of FVIIFC [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00300] Mean Residence Time (MRT) of Factor VIII activity (FVIIFC) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF : RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII] for participants in the PK50 arms (Arm 1 and Arm 2).

Secondary Outcome Measures: Secondary Outcome #39

[00301] PK50 - Clearance of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00302] Clearance (CL) of Factor VIII activity (FVIIFC) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII] for participants in the PK50 arms (Arm 1 and Arm 2).

Secondary Outcome Measures: Secondary Outcome #40

[00303] PK50 - Incremental Recovery of FVIIFC [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00304] Incremental Recovery (IR) at the maximum plasma concentration of Factor VIII activity (FVIIFC) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII] for participants in the PK50 arms (Arm 1 and Arm 2).

Secondary Outcome Measures: Secondary Outcome #41

[00305] PK50 - Elimination Phase Half-Life of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] [00306] Elimination Phase Half-Life (Tl/2) of Factor VIII activity (FVIIFC) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant FVIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII] for participants in the PK50 arms (Arm 1 and Arm 2).

Secondary Outcome Measures: Secondary Outcome #42

[00307] PK50 - Volume of Distribution at Steady State of FVIII: C [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00308] Volume of Distribution at Steady State (Vss) of Factor VIII activity (FVIII: C) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII] for participants in the PK50 arms (Arm 1 and Arm 2).

Secondary Outcome Measures: Secondary Outcome #43

[00309] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00310] Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF)

[rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #44

[00311] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96

Hours (AUC0-96h/Dose) of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00312] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF)

[rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #45

[00313] PK80 - Mean Residence Time of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00314] Mean Residence Time (MRT) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #46

[00315] PK80 - Clearance of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00316] Clearance (CL) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #47

[00317] PK80 - Incremental Recovery of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00318] Incremental Recovery (IR) at the maximum plasma concentration Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor

Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1 ] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #48

[00319] PK80 - Elimination Phase Half-Life of VWF:Co [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00320] Elimination Phase Half-Life (Tl/2) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category

Secondary Outcome Measures: Secondary Outcome #49 [00321] PK80 - Volume of Distribution at Steady State of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00322] Volume of Distribution at Steady State (Vss) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study. PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #50

[00323] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00324] Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Antigen (VWF:Ag) after infusion of 80 IU/kg recombinant von

Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #51

[00325] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00326] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Antigen (VWF:Ag) after infusion of 80 IU/kg recombinant von

Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #52

[00327] PK80 - Mean Residence Time of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00328] Mean Residence Time (MRT) of von Willebrand Factor Antigen (VWF: Ag) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #53

[00329] PK80 - Clearance of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00330] Clearance (CL) of von Willebrand Factor Antigen (VWF: Ag) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #54

[00331] PK80 - Incremental Recovery of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00332] Incremental Recovery (IR) at the maximum plasma concentration Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Antigen (VWF:Ag) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #55

[00333] PK80 - Elimination Phase Half-Life of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00334] Elimination Phase Half-Life (Tl/2) of von Willebrand Factor Antigen (VWF:Ag) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #56

[00335] PK80 - Volume of Distribution at Steady State of VWF: Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00336] Volume of Distribution at Steady State (Vss) of von Willebrand Factor Antigen (VWF:Ag) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm

(participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #57

[00337] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00338] Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #58

[00339] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00340] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participatns from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #59

[00341] PK80 - Mean Residence Time of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00342] Mean Residence Time (MRT) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #60

[00343] PK80 - Clearance of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00344] Clearance (CL) of von Willebrand Factor Collagen Binding (VWF: CB) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #61 [00345] PK80 - Incremental Recovery of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00346] Incremental Recovery (IR) at the maximum plasma concentration Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #62

[00347] PK80 - Elimination Phase Half-Life of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00348] Elimination Phase Half-Life (Tl/2) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #63

[00349] PK80 - Volume of Distribution at Steady State of VWF: CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] [00350] Volume of Distribution at Steady State (Vss) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #64

[00351] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00352] Area under the plasma concentration curve (AUC) from time 0 to infinity of Factor VIII activity (FVIIFC) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #65

[00353] PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00354] Area under the plasma concentration curve (AUC) from time 0 to 96 hours of Factor VIII activity (FVIIFC) after infusion of 80 IU/kg recombinant von Willebrand

Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

Secondary Outcome Measures: Secondary Outcome #66

[00355] PK80- Ratio of Intra-participant PK of VWF:RCo, VWF:Ag and VWF:CB at Baseline and After 6 Months [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

[00356] Area under the plasma concentration curve (AUC) from time 0 to infinity per dose (AUC0-∞/dose) for von Willebrand Factor Ristocetin cofactor (VWF:RCo), von Willebrand Factor Antigen (VWF:Ag) and von Willebrand Factor Collagen Binding (VWF:CB). Each parameter was compared between the two PK assessments after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. 13 participants had data available for this endpoint i.e. data for PK1 and PK2.

Eligibility Criteria

[00357] 18 Years to 65 Years (Adult, Older Adult); All sexes.

Inclusion Criteria:

[00358] Participant has been diagnosed with:

· Type 1 (Von Willebrand factor: Ristocetin cofactor activity (VWF:RCo) < 20 IU/dL) or,

• Type 2A (VWF:RCo < 20 IU/dL), Type 2B (as diagnosed by genotype), Type 2N (Factor VIII activity (FVIILC) <10% and historically documented genetics), Type 2M or,

· Type 3 (Von Willebrand factor antigen (VWF:Ag) < 3 IU/dL) or,

• Severe Von Willebrand disease (VWD) with a history of requiring substitution

therapy with von Willebrand factor concentrate to control bleeding [00359] Participant, who participates in the treatment for bleeding episodes, has had a minimum of 1 documented bleed (medical history) requiring VWF coagulation factor replacement therapy during the previous 12 months prior to enrollment.

[00360] Participant has a Karnofsky score > 60%

[00361] Participant is at least 18 and not older than 65 years of age at enrollment

[00362] If female of childbearing potential, participant presents with a negative pregnancy test

[00363] Participant agrees to employ adequate birth control measures for the duration of the study

[00364] Participant is willing and able to comply with the requirements of the protocol Exclusion Criteria:

[00365] Participant has been diagnosed with pseudo VWD or another hereditary or acquired coagulation disorder other than VWD (eg qualitative and quantitative platelet disorders or elevated PT/international normalized ratio [INR] >1.4).

[00366] Participant has a documented history of a VWF:RCo half-life of <6 hours.

[00367] Participant has a history or presence of a VWF inhibitor at screening.

[00368] Participant has a history or presence of a factor VIII (FVIII) inhibitor with a titer >0.4 BU (by Nijmegen assay) or > 0.6 BU (by Bethesda assay).

[00369] Participant has a known hypersensitivity to any of the components of the study drugs, such as mouse or hamster proteins.

[00370] Participant has a medical history of immunological disorders, excluding seasonal allergic rhinitis/conjunctivitis, mild asthma, food allergies or animal allergies.

[00371] Participant has a medical history of a thromboembolic event.

[00372] Participant is HIV positive with an absolute CD4 count <200/mm3.

[00373] Participant has been diagnosed with cardiovascular disease (New York Heart Association [NYHA] classes 1-4.

[00374] Participant has an acute illness (eg, influenza, flu-like syndrome, allergic rhinitis/conjunctivitis, non-seasonal asthma) at screening. [00375] Participant has been diagnosed with significant liver disease as evidenced by any of the following: serum alanine aminotransferase (ALT) 5 times the upper limit of normal; hypoalbuminemia; portal vein hypertension (eg, presence of otherwise unexplained splenomegaly, history of esophageal varices).

[00376] Participant has been diagnosed with renal disease, with a serum creatinine level >2 mg/dL.

[00377] In the judgment of the investigator, the participant has another clinically significant concomitant disease (eg, uncontrolled hypertension) that may pose additional risks for the participant.

[00378] Participant has been treated with an immunomodulatory drug, excluding topical treatment (e.g., ointments, nasal sprays), within 30 days prior to enrollment

[00379] Participant is pregnant or lactating at the time of enrollment.

[00380] Participant has participated in another clinical study involving an IP or

investigational device within 30 days prior to enrollment or is scheduled to participate in another clinical study involving an investigational product or investigational device during the course of this study.

[00381] Participant has a history of drug or alcohol abuse within the 2 years prior to enrollment.

[00382] Participant has a progressive fatal disease and/or life expectancy of less than 3 months.

[00383] Participant is identified by the investigator as being unable or unwilling to cooperate with study procedures.

[00384] Participant suffers from a mental condition rendering him/her unable to understand the nature, scope and possible consequences of the study and/or evidence of an uncooperative attitude.

[00385] Participant is in prison or compulsory detention by regulatory and/or juridical order Pre-Assignment Details

[00386] 49 participants provided informed consent and were screened for the study, of which 37 were exposed to study product. Reasons for discontinuation were 6 screen failures, consent withdrawn by 3 participants, 1 physician decision, 1 participant received high doses of rFVIII for oral procedure and arm for which 1 participant was eligible was closed.

Reporting Groups

I Description

Arm 1: PK50 + Treatment In Part A, (pharmacokinetic [PK] assessment followed by on- demand treatment for bleeding episodes [BEs] for 6 months) participants were initially infused either with 50 IU/kg recombinant von Willebrand Factorvon Willebrand Ristocetin cofactor (VWF:RCo rVWF) [rVWF] administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII)

[rVWF: rFVIII] or 50 IU/kg rVWF administered together with saline. Participants then crossed over to the alternate infusion after washout (PK). For on-demand treatment, participants received study product [VWF: rFVIII or rVWF], where BEs were initially treated with rVWF: rFVIII and subsequently with rVWF with or without rFVIII, based on FVIII levels (dose based on previous FVIII levels or if not available from the individual participant's PK data at discretion of investigator). In part, B participants continued to receive on- demand treatment for BEs with study product [VWF: rFVIII or rVWF] for a further 6 months.

In Part A, (pharmacokinetic [PK] assessment followed by on- demand treatment for bleeding episodes [BEs] for 6 months) participants were initially infused either with 50 IU/kg recombinant von Willebrand Factorvon Willebrand Ristocetin cofactor (VWF:RCo rVWF) [rVWF] administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII)

[rVWF: rFVIII] or 50 IU/kg rVWF administered together with saline. Participants then crossed over to the alternate infusion after washout (PK). For on-demand treatment, participants received study product [VWF: rFVIII or rVWF], where BEs were initially treated with rVWF: rFVIII and subsequently with rVWF with or without rFVIII, based on FVIII levels (dose based on previous FVIII levels or if not available from the individual participant's PK data at discretion of investigator). Participants then exited the study or could opt to sign informed consent to move to Arm 1 receive treatment for bleeding episodes with study product.

Arm 3: PK80 + Treatment In Part A, participants initially underwent a first PK

assessment of an infusion of 80 IU/kg recombinant von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF]. After the first PK assessment participants received on demand treatment for bleeding episodes (BEs) with study product [VWF: rFVIII or rVWF], where BEs were initially treated with rVWF:rFVIII and subsequently with rWVF with or without rFVIII, based on FVIII levels. If FVIII levels not available, the individual participant's PK data was used to determine rFVIII dose at discretion of investigator.

Participants received on-demand treatment for 6 months after the first study product infusion. After 6 months participants underwent a second PK assessment of an infusion of 80 IU/kg rVWF. In part B, participants continued to receive on-demand treatment for BEs with study product [VWF: rFVIII or rVWF] for a further 6 months.

Arm 4: Treatment Only In Part A, participants received on-demand treatment for bleeding episodes (BEs) with study product (recombinant von Willebrand Factor [rVWF] administered together with recombinant Factor VIII [rFVIII] (rVWF: rFVIII) or rVWF alone), where BEs were initially treated with rVWF: rFVIII and subsequently with rVWF with or without rFVIII, based on FVIII levels. If not available, the individual participant's PK data was used to determine rFVIII dose at discretion of investigator. Participants received on-demand treatment for 6 months after the first study product infusion. In part, B participants continued to receive on-demand treatment for BEs with study product [VWF: rFVIII or rVWF] for a further 6 months. No pharmacokinetic (PK) assessments were conducted in this arm.

Participant Flow: Overall Study

Arm 4:

Arm 1: PK50 + Arm 2: Arm 3: PK80 +

Treatment Treatment PK50 Only Treatment

Only

STARTED 8 8 15 6

COMPLETED 4 8 13 5

NOT COMPLETED 4 0 2 1

Adverse Event 1 0 0 0

Withdrawal by

3 0 1 0 Subject

Pregnancy 0 0 0 1

Met Excl. Criteria

0 0 1 0 After Starting Stud

Baseline Characteristics [00387] Baseline consists of all participants in study [N=37] so is a total of the four arms described in Participant Flow (Arm 1 : PK50 + Treatment [N=8]; Arm 2: PK50 Only [N=8]; Arm 3: PK80 + Treatment [N=15]; Arm 4: Treatment Only [N=6]). RESULTS;

Outcome Measures

Primary Outcome Measures: Primary Outcome #1

[00388] 1. Primary: Percentage of Participants With Treatment Success for Treated Bleeding Episodes [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF].

Measure Type Primary

Percentage of Participants With Treatment Success for Treated

Measure Title

Bleeding Episodes

Measure Description Treatment success was defined as the extent of control of bleeding episodes (BEs) using a mean efficacy rating score of <2.5 for a participant's BEs treated with study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) during the study period. Scores used: Excellent = 1 - actual infusions < estimated number of infusions required to treat BE; no additional VWF required (all BEs); Good = 2 - >l-2 infusions (minor/moderate BEs) or < 1.5 infusions (major BEs) greater than estimated required to control BE; no additional VWF required (all BEs); Moderate = 3 > 3 infusions (minor/moderate BEs) or > 1.5 infusions (major BEs) greater than estimated required to control BE; no additional VWF required (all BEs); None = 4 - severe uncontrolled bleeding or intensity of bleeding not changed;

additional VWF required. Included participants with available primary efficacy rating (prospective-excluding gastrointestinal bleeds) in the Full Analysis Set.

Time Frame For 12 months after first infusion of rVWF:rFVIII or rVWF

Reporting Groups

Description

Full Analysis Set Comprises of participants treated with study product (recombinant von

Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) for whom at least one efficacy rating scale was available. Measured Values

Full Analysis Set

Participants Analyzed 18

Percentage of Participants With Treatment Success for Treated

Bleeding Episodes 100.0

:[Units: Percent of participants] (84.7 to 100.0)

Number (90% Confidence Interval) Statistical Analysis 1 for Percentage of Participants With Treatment Success for Treated Bleedin Episodes

Secondary Outcome Measures: Secondary Outcome #1

[00389] 2. Secondary: Percentage of Treated Bleeding Episodes With an Efficacy Rating of "Excellent" or "Good" [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF].

Table 10: Secondary Outcome #1

Table 11: Re ortin Grou s

Tabel 12: Measured Values

I Full Analysis Set

Participants Analyzed 22 lUnits Analyzed (Bleeding episodes) 130

Percentage of Treated Bleeding Episodes With an Efficacy Rating of

"Excellent" or "Good" 100.0

{Units: Percent of bleeding episodes] (97.7 to 100.0)

Number (90% Confidence Interval)

No statistical analysis provided for Percentage of Treated Bleeding Episodes With an Efficacy Rating of "Excellent" or "Good" Secondary Outcome Measures: Secondary Outcome #2

[00390] 3. Secondary: Percentage of Treated Bleeding Episodes With an Efficacy Rating of "Excellent" or "Good", Excluding Gastrointestinal Bleeds [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 13: Secondary Outcome #2

Measure Type Secondary

Percentage of Treated Bleeding Episodes With an Efficacy Rating of

Measure Title

"Excellent" or "Good", Excluding Gastrointestinal Bleeds

Measure Description Efficacy ratings of "excellent" or "good" for the control of bleeding episodes (BEs) with study product (recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) are defined as follows: Excellent - actual infusions < estimated number of infusions required to treat BE; no additional von Willebrand Factor (VWF) required (all BEs); Good - >l-2 infusions

(minor/moderate BEs) or < 1.5 infusions (major BEs) greater than estimated required to control BE; no additional VWF required (all BEs). The data set included prospectively estimated BEs excluding gastrointestinal (GI) bleeds treated with study product with an available efficacy rating from participants in the Full Analysis Set.

Time Frame For 12 months after first infusion of rVWF:rFVIII or rVWF

Table 14: Reporting Groups

Description

Full Analysis Set Comprised of participants treated with study product (recombinant von

Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) for whom at least one efficacy rating scale was available.

Table 15: Measured Values

Participants Analyzed

lUnits Analyzed (Bleeding episodes)

Percentage of Treated Bleeding Episodes With an Efficacy Rating

;"Excellent" or "Good", Excluding Gastrointestinal Bleeds

![Units: Percent of bleeding episodes]

iGeometric Mean (90% Confidence Interval) No statistical analysis provided for Percentage of Treated Bleeding Episodes With an Efficacy Rating of "Excellent" or "Good", Excluding Gastrointestinal Bleeds

Secondary Outcome Measures: Secondary Outcome #3

[00391] 4. Secondary: Number of Infusions of rVWF:rFVIII and/or rVWF Per Bleeding Episode [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 16: Secondary Outcome #3

Measure Type Secondary

Number of Infusions of rVWF:rFVIII and/or rVWF Per Bleeding

Measure Title

Episode

Table 18: Measured Values

Full Analysis Set jParticipants Analyzed 22 iUnits Analyzed (Bleeding episodes) 192

Number of Infusions of rVWF:rFVIII and/or rVWF Per Bleeding

!Episode 1.0 j[Units: Number of infusions] (1.0 to 1.0) Median (90% Confidence Interval) No statistical analysis provided for Number of Infusions of rVWF:rFVIII and/or rVWF Per Bleeding Episode

Secondary Outcome Measures: Secondary Outcome #4

[00392] 5. Secondary: Number of Units of rVWF:rFVIII and/or rVWF Per Bleeding Episode [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 19: Secondary Outcome #4

Table 20: Re ortin Grou s

Table 21: Measured Values

Full Analysis Set jParticipants Analyzed 22 iUnits Analyzed (Bleeding episodes) 174

Number of Units of rVWF: rFVIII and/or rVWF Per Bleeding

!Episode 48.2

!| Units: IU/kg] (43.9 to 50.2)

!Median (90% Confidence Interval) No statistical analysis provided for Number of Units of rVWF:rFVIII and/or rVWF Per Bleeding Episode

Secondary Outcome Measures: Secondary Outcome #5

[00393] 6. Secondary: Percentage of Participants Who Develop Inhibitory Antibodies to FVIII [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 22: Secondary Outcome #5

Table 23: Reporting Groups

I Description

Safety Analysis Set j Comprised of participants who were treated with study product

i (recombinant von Willebrand Factor [rVWF] with or without

I recombinant factor VIII [rFVIII]) at least once during the study.

Table 24: Measured Values

Safety Analysis Set

Participants Analyzed 37

Percentage of Participants Who Develop Inhibitory Antibodies to

FVIII

j[Units: Percent of participants]

Before 1st treatment with study product [N=37] 0 During 1st treatment until study end [N=27] 0 At final study visit | N=24| 0 No statistical analysis provided for Percentage of Participants Who Develop Inhibitory Antibodies to FVIII

Secondary Outcome Measures: Secondary Outcome #6

[00394] 7. Secondary: Percentage of Participants Who Develop Inhibitory Antibodies to VWF [Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 25: Secondary Outcome #6

Table 26: Reporting Groups

Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study.

Table 27: Measured Values

Safety Analysis Set

Participants Analyzed 37

Percentage of Participants Who Develop Inhibitory Antibodies to

VWF [Units: Percent of participants]

Before 1st treatment with study product [N=37] 0

During 1st treatment until study end [N=27] 0

At final study visit [N=24] 0

No statistical analysis provided for Percentage of Participants Who Develop Inhibitory Antibodies to VWF

Secondary Outcome Measures: Secondary Outcome #7

[00395] 8. Secondary: Percentage of Participants Who Develop Binding Antibodies to VWF [Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 28: Secondar Outcome #7

After signing informed consent until 12 months after first infusion

Time Frame

of rVWF:rFVIII or rVWF

Table 29: Reporting Groups

I Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study. Table30: Measured Values

Safety Analysis Set iParticipants Analyzed 37 Percentage of Participants Who Develop Binding Antibodies to

VWF

[Units: Percent of participants]

Before 1st treatment with study product [N=37] 0

During 1st treatment until study end [N=28] 0

At final study visit [N=24] 0

No statistical analysis provided for Percentage of Participants Who Develop Binding Antibodies to VWF

Secondary Outcome Measures: Secondary Outcome #8

[00396] 9. Secondary: Percentage of Participants Who Develop Binding Antibodies to CHO [Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 30: Secondary Outcome #9

Table 31: Reporting Groups

Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study.

Table 32: Measured Values

Safety Analysis Set

Participants Analyzed 37 Percentage of Participants Who Develop Binding Antibodies to

CHO

{Units: Percent of participants]

Before 1st treatment with study product [N=37]

During 1st treatment until study end [N=28]

At final study visit [N=24]

No statistical analysis provided for Percentage of Participants Who Develop Binding Antibodies to CHO

Secondary Outcome Measures: Secondary Outcome #9

[00397] 10. Secondary: Percentage of Participants Who Develop Binding Antibodies to rFurin [Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 33: Secondar Outcome #9

Table 34: Reporting Groups

Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study.

Table 35: Measured Values

Safety Analysis Set

Participants Analyzed 37 Percentage of Participants Who Develop Binding Antibodies to

!rFurin

{Units: Percent of participants]

Before 1st treatment with study product [N=37]

During 1st treatment until study end [N=28]

At final study visit [N=24]

No statistical analysis provided for Percentage of Participants Who Develop Binding Antibodies to rFurin

Secondary Outcome Measures: Secondary Outcome #10

[00398] 11. Secondary: Percentage of Participants Who Develop Binding Antibodies to Mouse Immunoglobulin [Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 36: Secondar Outcome #10

Table 37: Reporting Groups

Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study. Table 38: Measured Values

j Safety Analysis Set

Participants Analyzed 37 Percentage of Participants Who Develop Binding Antibodies to Mouse Immunoglobulin

{Units: Percent of participants]

Before 1st treatment with study product [N=37]

During 1st treatment until study end [N=28]

At final study visit [N=24]

No statistical analysis provided for Percentage of Participants Who Develop Binding Antibodies to Mouse Immunoglobulin

Secondary Outcome Measures: Secondary Outcome #1 1

[00399] 12. Secondary: Percentage of Participants Who Had an Occurrence of Thrombotic Events [Time Frame: After signing informed consent until 12 months after first infusion of rVWF:rFVIII or rVWF]

After signing informed consent until 12 months after first infusion

Time Frame

of rVWF:rFVIII or rVWF

Table 40: Reporting Groups

I Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study.

Table 41: Measured Values

Safety Analysis Set

Participants Analyzed

Percentage of Participants Who Had an Occurrence of Thrombotic

!Events

j| Units: Percent of participants] No statistical analysis provided for Percentage of Participants Who Had an Occurrence of Thrombotic Events

Secondary Outcome Measures: Secondary Outcome #12

[00400] 13. Secondary: Number of Adverse Events Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 42: Secondary Outcome #12

! Measure Type ; Secondary

I Number of Adverse Events Related to Study Product Including

I Measure Title i Clinically Significant Changes in Laboratory Parameters and Vital

I Signs

I Measure Description i Adverse Events (AEs) related to study product (recombinant von

; Willebrand Factor [rVWF] with or without recombinant factor VIII i [rFVIII]) are described. Only laboratory parameters (hematology

! and clinical chemistry) and vital signs (physical examination, ECG)

; with clinically significant findings that are recorded as AEs are i included. Categories presented as Severity-System Organ Class-

; Preferred Term Seriousness: serious adverse event (SAE); non

! serious adverse event (nsAE) System Organ Class: Cardiac i disorders (CARD); General disorders and administration site

I conditions (GEN); Investigations (INV); Nervous system disorders

; (NERV); Skin and subcutaneous tissue disorders (SKN); Vascular

! disorders (VAS). Category title includes number of AEs [N] for the i category.

; Time Frame i For 12 months after first infusion of rVWF:rFVIII or rVWF

Table 43: Reporting Groups

I Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study.

Table 44: Measured Values

Safety Analysis Set

Participants Analyzed 37 iUnits Analyzed (Adverse Events) 125 jNumber of Adverse Events Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs

{Units: Number of Adverse Events]

SAE-GEN-Chest discomfort [N=l]

SAE-INV-Heart rate increased [N=l]

nsAE-CARD-Tachycardia [N=l]

nsAE-GEN-Infusion site paraesthesia [N=l]

nsAE-INV-ECG T wave inversion [N=l]

nsAE-NERV-Dysgeusia [N=l]

nsAE-SKN-Pruritus generalized [N=l]

nsAE-VAS-Hot flush [N=l]

No statistical analysis provided for Number of Adverse Events Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs

Secondary Outcome Measures: Secondary Outcome #13

[00401] 14. Secondary: Number of Participants With Adverse Events Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF]

Table 45: Secondary Outcome #13

Time Frame For 12 months after first infusion of rVWF:rFVIII or rVWF

Table 46: Reportin Groups

Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study.

Table 47: Measured Values

Safety Analysis Set

Participants Analyzed 37

Number of Participants With Adverse Events Related to Study Product

Including Clinically Significant Changes in Laboratory Parameters and

jVital Signs

j[Units: Number of participants]

SAE-GEN-Chest discomfort 1

SAE-INV-Heart rate increased 1 nsAE-CARD-Tachycardia 1 nsAE-GEN-Infusion site paraesthesia 1 nsAE-INV-ECG T wave inversion 1 nsAE-NERV-Dysgeusia 1 nsAE-SKN-Pruritus generalized 1 nsAE-VAS-Hot flush 1

No statistical analysis provided for Number of Participants With Adverse Events Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs

Secondary Outcome Measures: Secondary Outcome #14

[00402] 15. Secondary: Number of Adverse Events by Infusion Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs [Time Frame: For 12 months after first infusion of rVWF:rFVIII or rVWF] Table 48: Secondary Outcome #14

! Measure Type Secondary

I Measure Title Number of Adverse Events by Infusion Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs

Measure Description Adverse Events (AEs) by infusion related to study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) are described. Only laboratory parameters (hematology and clinical chemistry) and vital signs (physical examination, ECG) with clinically significant findings that are recorded as AEs are included. Categories presented as Severity- System Organ Class-Preferred Term Seriousness: serious adverse event (SAE); non serious adverse event (nsAE) System Organ Class: Cardiac disorders (CARD); General disorders and administration site conditions (GEN); Investigations (INV); Nervous system disorders (NERV); Skin and subcutaneous tissue disorders (SKN); Vascular disorders (VAS).

Time Frame For 12 months after first infusion of rVWF:rFVIII or rVWF

Table 49 : Reporting Groups

Description

Safety Analysis Set Comprised of participants who were treated with study product

(recombinant von Willebrand Factor [rVWF] with or without recombinant factor VIII [rFVIII]) at least once during the study. Table 50: Measured Values

Safety

Analysis Set

Participants Analyzed 37

Units Analyzed (Infusions) 318

Number of Adverse Events by Infusion Related to Study Product Including

Clinically Significant Changes in Laboratory Parameters and Vital Signs

[Units: Number of Adverse Events]

SAE-GEN-Chest discomfort 1

SAE-INV-Heart rate increased 1 nsAE-CARD-Tachycardia 1 nsAE-GEN-Infusion site paraesthesia 1 nsAE-INV-ECG T wave inversion 1 nsAE-NERV-Dysgeusia 1 nsAE-SKN-Pruritus generalized 1 nsAE-VAS-Hot flush

1 No statistical analysis provided for Number of Adverse Events by Infusion Related to Study Product Including Clinically Significant Changes in Laboratory Parameters and Vital Signs

Secondary Outcome Measures: Secondary Outcome #15

[00403] 16. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 51: Secondary Outcome #15

Measure Type Secondary

PK50 - Area Under the Plasma Concentration/Time Curve From

Measure Title

Time 0 to Infinity (AUC0-∞/Dose) of VWF:RCo

Measure Description Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for subjects in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 52: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total of participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 53: Measured Values

PK50 Arms

Participants Analyzed

PK50 - Area Under the Plasma Concentration/Time Curve From Time 0

to Infinity (AUC0-∞/Dose) of VWF:RCo

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

32.4 rVWF:rFVIII [N=16]

(27.5 to 40.1)

32.7 rVWF [N=14]

(29.0 to 47.8)

No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO-oo/Dose) of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #16

[00404] 17. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 54: Secondar Outcome #16

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 55: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 56: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Area Under the Plasma Concentration/Time Curve From Time 0

to 96 Hours (AUC0-96h/Dose) of VWF:RCo

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

31.6 rVWF: rFVIII [N=16]

(27.3 to 37.3)

31.3 rVWF [N=14]

(28.4 to 43.7)

No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #17

[00405] 18. Secondary: PK50 - Mean Residence Time of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 57: Secondary Outcome #17

Measure Type Secondary Measure Title PK50 - Mean Residence Time of VWF:RCo

Measure Description Mean Residence Time (MRT) of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2)

[rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 59: Measured Values

No statistical analysis provided for PK50 - Mean Residence Time of WVF:RCo

Secondary Outcome Measures: Secondary Outcome #18 [00406] 19. Secondary: PK50 - Clearance of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 60: Secondar Outcome #18

Table 61: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms. Table 62: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Clearance of VWF:RCo

[Units: dL/kg/hours]

Median (95% Confidence Interval)

0.031

rVWF: rFVIII [N=16]

(0.025 to 0.041) 0.031

rVWF [N=14]

(0.021 to 0.035)

No statistical analysis provided for PK50 - Clearance of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #19

[00407] 20. Secondary: PK50 - Incremental Recovery of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 63: Secondar Outcome # 19

Measure Type Secondary

Measure Title PK50 - Incremental Recovery of VWF:RCo

Measure Description Incremental Recovery (IR) at the maximum plasma concentration of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 64: Reporting Groups

Description

Comprised of participants who underwent PK analysis of study product (50 IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms. Table 65: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Incremental Recovery of VWF:RCo

[Units: (U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

1.8

rVWF:rFVIII [N=16]

(1.6 to 2.4)

1.8

rVWF [N=14]

(1.5 to 2.2)

No statistical analysis provided for PK50 - Incremental Recovery of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #20

[00408] 21. Secondary: PK50 - Elimination Phase Half-Life of VWF:Co [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 66: Secondar Outcome #20

Table 67: Reporting Groups Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 68: Measured Values

No statistical analysis provided for PK50 - Elimination Phase Half-Life of VWF:Co

Secondary Outcome Measures: Secondary Outcome #21

[00409] 22. Secondary: PK50 - Volume of Distribution at Steady State of

VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 69: Secondary Outcome #21

together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 70: Reporting Groups

Table 71: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Volume of Distribution at Steady State of VWF:RCo

[Units: dL/kg]

Median (95% Confidence Interval)

0.70

rVWF: rFVIII [N=16]

(0.66 to 0.93)

0.83

rVWF [N=14]

(0.70 to 0.97)

No statistical analysis provided for PK50 - Volume of Distribution at Steady State of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #22

[00410] 23. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 72: Secondar Outcome #23

Table 74: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Area Under the Plasma Concentration/Time Curve From Time 0

to Infinity (AUC0-∞/Dose) of VWF:Ag

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

67.8 rVWF: rFVIII [N=16]

(55.1 to 81.7)

67.1 rVW I [N=14]

(55.6 to 80.5) No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO-oo/Dose) of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #23

[00411] 24. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout]

Table 75: Secondar Outcome #24

Table 76: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms. Table 77: Measured Values PK50 Arms

Participants Analyzed 16

PK50 - Area Under the Plasma Concentration/Time Curve From Time 0

to 96 Hours (AUC0-96h/Dose) of VWF:Ag

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

62.1 rVWF:rFVIII [N=16]

(52.8 to 74.9)

62.2 rVWF [N=14]

(54.7 to 74.5)

No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #24

[00412] 25. Secondary: PK50 - Mean Residence Time of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 78: Secondary Outcome #24

Table 79: Reporting Groups Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 80: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Mean Residence Time of V F:Ag

[Units: Hours]

Median (95% Confidence Interval)

32.1

rVWF: rFVIII [N=16]

(29.8 to 41.1)

34.3

rVWF [N=14]

(30.4 to 41.4)

No statistical analysis provided for PK50 - Mean Residence Time of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #25

[00413] 26. Secondary: PK50 - Clearance of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 81: Secondary Outcome #25

Measure Type Secondary

Measure Title PK50 - Clearance of VWF:Ag

Measure Description Clearance (CL) of von Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the

category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 82: Reporting Groups

i Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 83: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Clearance of VWF:Ag

[Units: dL/kg/hours]

Median (95% Confidence Interval)

0.015

rVWF: rFVIII [N=16]

(0.013 to 0.018)

0.015

rVWF [N=14]

(0.013 to 0.018)

No statistical analysis provided for PK50 - Clearance of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #26

[00414] 27. Secondary: PK50 - Incremental Recovery of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 84: Secondary Outcome #26 Measure Type Secondary

Measure Title PK50 - Incremental Recovery of VWF:Ag

Measure Description Incremental Recovery (IR) at the maximum plasma concentration of von Willebrand Factor Antigen (VWF:Ag) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3 : 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 85: Reportin . Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 86: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Incremental Recovery of WVF:Ag

[Units: (U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

2.3

rVWF: rFVIII [N=16]

(2.0 to 2.5)

2.2

rVW I [N=14]

(1.9 to 2.5)

No statistical analysis provided for PK50 - Incremental Recovery of WVF:Ag Secondary Outcome Measures: Secondary Outcome #27

[00415] 28. Secondary: PK50 - Elimination Phase Half-Life of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 87: Secondar Outcome #27

Table 88: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 89: Measured Values

PK50 Arms

jParticipants Analyzed 16

PK50 - Elimination Phase Half-Life of VWF:Ag

I [Units: Hours]

Median (95% Confidence Interval)

rVWF: rFVIII [N=16] 21.8

No statistical analysis provided for PK50 - Elimination Phase Half-Life of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #28

[00416] 29. Secondary: PK50 - Volume of Distribution at Steady State of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 90: Secondary Outcome #28

Table 91: Reporting Groups

Table 92: Measured Values

P 50 Arms

Participants Analyzed 16

PK50 - Volume of Distribution at Steady State of VWF:Ag

[Units: dL/kg]

Median (95% Confidence Interval)

0.50

rVWF:rFVIII [N=16]

(0.45 to 0.56)

0.49

rVWF [N=14]

(0.45 to 0.58)

No statistical analysis provided for PK50 - Volume of Distribution at Steady State of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #29

[00417] 30. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 93: Secondar Outcome #30

PK evaluation for both infusions and washout.

Table 94: Reporting . Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 95: Measured Values

PK50 Arms

Participants Analyzed 16

:PK50 - Area Under the Plasma Concentration/Time Curve From Time 0

to Infinity (AUC0-∞/Dose) of VWF:CB

|| Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

80.1 rVWF: rFVIII [N=16]

(68.4 to 95.0)

81.3 rVWF [N=14]

(71.2 to 99.8)

No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO /Dose) of VWF:CB

Secondary Outcome Measures: Secondary Outcome #30

[00418] 31. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 96: Secondary Outcome #30

Measure Type Secondary PK50 - Area Under the Plasma Concentration/Time Curve From

Measure Title

Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:CB

Measure Description Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 97: Reporting . Groups

Description

Comprised of participants who underwent PK analysis of study product (50 IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 98: Measured Values

PK50 Arms

Participants Analyzed 16

:PK50 - Area Under the Plasma Concentration/Time Curve From Time 0

to 96 Hours (AUC0-96h/Dose) of VWF:CB

|| Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

78.7 rVWF: rFVIII [N=16]

(66.5 to 90.5)

75.1 rVW I [N=14]

(69.2 to 97.0)

No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of V F:CB Secondary Outcome Measures: Secondary Outcome #31

[00419] 32. Secondary: PK50 - Mean Residence Time of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 99: Secondary Outcome #31

Table 100: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 101: Measured Values

\ PK50 Arms

Participants Analyzed 16

K50 - Mean Residence Time of V F:CB

I [Units: Hours] Median (95% Confidence Interval)

27.5

rVWF:rFVIII [N=16]

(22.7 to 32.1)

26.1

rVWF [N=14]

(25.1 to 33.2)

No statistical analysis provided for PK50 - Mean Residence Time of VWF:CB

Secondary Outcome Measures: Secondary Outcome #32

[00420] 33. Secondary: PK50 - Clearance of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 102: Secondary Outcome #32

arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 104: Measured Values

Participants Analyzed

PK50 - Clearance of VWF:CB

[Units: dL/kg/hours]

Median (95% Confidence Interval)

0.012

rVWF:rFVIII [N=16]

(0.011 to 0.015)

0.012

rVWF [N=14]

(0.011 to 0.015)

No statistical analysis provided for PK50 - Clearance of VWF:CB

Secondary Outcome Measures: Secondary Outcome #33

[00421] 34. Secondary: PK50 - Incremental Recovery of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 105: Secondary Outcome #33

Measure Type Secondary

Measure Title PK50 - Incremental Recovery of VWF:CB

Measure Description Incremental Recovery (IR) at the maximum plasma concentration of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of

1.3: 1±0.2) [rVWF: rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6,

Time Frame

12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 106: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 107: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Incremental Recovery of V F:CB

[Units: (U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

3.4

rVWF: rFVIII [N=16]

(3.0 to 3.7)

3.2

rVWF [N=14]

(2.8 to 3.7)

No statistical analysis provided for PK50 - Incremental Recovery of VWF:CB

Secondary Outcome Measures: Secondary Outcome #34

[00422] 35. Secondary: PK50 - Elimination Phase Half-Life of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 108 : Secondary Outcome #34

Measure Type Secondary

Measure Title j PK50 - Elimination Phase Half-Life of VWF :CB

Measure Description Elimination Phase Half-Life (Tl/2) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 50 IU/kg

recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant FVIII (rFVIII) (ratio of 1.3 : 1±0.2)

[rVWF:rFVIII], or 50 IU/kg VWF:RCo rVWF administered together with saline (placebo) [rVWF] for participants in the PK50 arms (Arm 1 and Arm 2). Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 109: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 110: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Elimination Phase Half-Life of VWF:CB

[Units: Hours]

Median (95% Confidence Interval)

19.3

rVWF: rFVIII [N=16]

(14.9 to 23.4)

18.3

rVW I [N=14]

(17.4 to 24.8)

No statistical analysis provided for PK50 - Elimination Phase Half-Life of VWF:CB

Secondary Outcome Measures: Secondary Outcome #35 [00423] 36. Secondary: PK50 - Volume of Distribution at Steady State of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 111: Secondar Outcome #35

Table 112: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms. Table 113: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Volume of Distribution at Steady State of VWF:CB

[Units: dL/kg]

Median (95% Confidence Interval)

0.35

rVWF: rFVIII [N=16]

(0.31 to 0.40) 0.36

rVWF [N=14]

(0.28 to 0.42)

No statistical analysis provided for PK50 - Volume of Distribution at Steady State of VWF:CB

Secondary Outcome Measures: Secondary Outcome #36

[00424] 37. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of FVIILC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 114: Secondary Outcome #36

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF: rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Tabel 116: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Area Under the Plasma Concentration/Time Curve From Time

0 to Infinity (AUC0-∞/Dose) of FVIII:C

[Units: hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

145.4 rVWF:rFVIII [N=16]

(118.8 to 189.5)

113.0 rVWF [N=14]

(93.0 to 167.4)

No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO-oo/Dose) of FVIII:C

Secondary Outcome Measures: Secondary Outcome #37

[00425] 38. Secondary: PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of FVIII:C [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 116: Secondar Outcome #37

participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Tabel 117: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 118: Measured Values

PK50 Arms

Participants Analyzed 16

IPK50 - Area Under the Plasma Concentration/Time Curve From Time

!O to 96 Hours (AUC0-96h/Dose) of FVIII:C

!| Units: hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

127.8 rVWF: rFVIII [N=16]

(112.3 to 145.1)

101.8 rVWF [N=14]

(74.4 to 124.4)

No statistical analysis provided for PK50 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of FVIII:C

Secondary Outcome Measures: Secondary Outcome #38

[00426] 39. Secondary: PK50 - Mean Residence Time of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 119: Secondary Outcome #38 Measure Type Secondary

Measure Title PK50 - Mean Residence Time of FVIII:C

Measure Description Mean Residence Time (MRT) of Factor VIII activity (FVIII:C) after infusion of 50 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF:rFVIII] for participants in the PK50 arms (Arm 1 and Arm 2).

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 120: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF: rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 121: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Mean Residence Time of FVIII:C

44.0

[Units: Hours]

(38.0 to 75.0)

Median (95 % Confidence Interval)

No statistical analysis provided for PK50 - Mean Residence Time of FVIII:C

Secondary Outcome Measures: Secondary Outcome #39

[00427] 40. Secondary: PK50 - Clearance of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 122: Secondary Outcome #39

Table 123: Reporting Groups

i Description

PK50 Arms i Comprised of participants who underwent PK analysis of study product (50

i IU/kg recombinant von Willebrand Factor (rVWF) administered together with j 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg

\ rVWF administered together with saline [rVWF]) i.e. a total participants from i Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 i arms have received at least one PK infusion and have provided data suitable i for PK analysis. Only PK data included from the PK50 arms.

Table 124: Measured Values

PK50 Arms

Participants Analyzed

PK50 - Clearance of FVIII:C

0.007

[Units: dL/kg/hours]

(0.006 to 0.009)

Median (95% Confidence Interval)

No statistical analysis provided for PK50 - Clearance of FVIII:C

Secondary Outcome Measures: Secondary Outcome #40

[00428] 41. Secondary: PK50 - Incremental Recovery of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 125: Secondary Outcome #40

Table 126: Reporting Groups

Description

Overall Study Arm ! No text entered. Table 127: Measured Values

Overall Study Arm

jParticipants Analyzed 16

IPK50 - Incremental Recovery of FVIII:C

2.3

!| Units: (U/dL)/(U VWF: RCo/kg)]

(1.9 to 2.7)

Median (95% Confidence Interval)

No statistical analysis provided for PK50 - Incremental Recovery of FVIII:C

Secondary Outcome Measures: Secondary Outcome #41

[00429] 42. Secondary: PK50 - Elimination Phase Half-Life of FVIILC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 128: Secondary Outcome #41 Measure Type Secondary

Measure Title PK50 - Elimination Phase Half-Life of FVIII:C

Measure Description Elimination Phase Half-Life (Tl/2) of Factor VIII activity (FVIIFC) after infusion of 50 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) administered together with 38.5 IU/kg recombinant FVIII (rFVIII) (ratio of 1.3: 1±0.2) [rVWF: rFVIII] for participants in the PK50 arms (Arm 1 and Arm 2).

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 129: Reporting Groups

Description

PK50 Arms Comprised of participants who underwent PK analysis of study product (50

IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF: rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

No statistical analysis provided for PK50 - Elimination Phase Half-Life of FVIII:C

Secondary Outcome Measures: Secondary Outcome #42

[00430] 43. Secondary: PK50 - Volume of Distribution at Steady State of FVIIFC [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 131: Secondary Outcome #42

Description

Comprised of participants who underwent PK analysis of study product (50 IU/kg recombinant von Willebrand Factor (rVWF) administered together with 38.5 IU/kg recombinant Factor VIII (rFVIII) [rVWF:rFVIII] or 50 IU/kg rVWF administered together with saline [rVWF]) i.e. a total participants from Arm 1 [PK50+Treatment] and Arm 2 [PK50 only]. Participants in the PK50 arms have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included from the PK50 arms.

Table 133: Measured Values

PK50 Arms

Participants Analyzed 16

PK50 - Volume of Distribution at Steady State of FVIII:C

0.32

|| Units: dL/kg]

(0.29 to 0.44)

Median (95% Confidence Interval)

No statistical analysis provided for PK50 - Volume of Distribution at Steady State of FVIII:C

Secondary Outcome Measures: Secondary Outcome #43

[00431] 44. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 136: Measured Values

P 80 Arm

Participants Analyzed 15

PK80 - Area Under the Plasma Concentration/Time Curve From Time 0

to Infinity (AUC0-∞/Dose) of VWF:RCo

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

36.9

PK1 of rVWF [N=15]

(29.2 to 41.7)

PK2 of rVWF [N=13] 38.9 (28.1 to 43.3)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO-oo/Dose) of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #44

[00432] 45. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:RCo [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 137: Secondar Outcome #44

Description

Comprised of participants who underwent PK analysis of study product (80 IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm. Table 137: Measured Values

PK80 Arm

Participants Analyzed 15

IPK80 - Area Under the Plasma Concentration/Time Curve From Time 0

to 96 Hours (AUC0-96h/Dose) of VWF:RCo

!| Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

35.6

PK1 of rVWF [N=15]

(28.9 to 41.2)

37.9

PK2 of rVWF [N=13]

(25.9 to 41.8)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #45

[00433] 46. Secondary: PK80 - Mean Residence Time of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 138: Secondar Outcome #45

PK evaluation for both infusions and washout.

Table 139 : Reporting Groups

Description

Comprised of participants who underwent PK analysis of study product (80 IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment] . Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 140: Measured Values

No statistical analysis provided for PK80 - Mean Residence Time of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #46

[00434] 47. Secondary: PK80 - Clearance of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 141: Secondar Outcome #47

; Measure Type ; Secondary

j Measure Title j PK80 - Clearance of VWF:RCo

; Measure Description ; Clearance (CL) of von Willebrand Factor Ristocetin cofactor

! (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand i Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo

I rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 142 : Reportin Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 143: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Clearance of VWF:RCo

[Units: dL/kg/hours]

Median (95% Confidence Interval)

0.027

PK1 of rVWF [N=15]

(0.024 to 0.034)

0.026

PK2 of rVWF [N=13]

(0.023 to 0.036)

No statistical analysis provided for PK80 - Clearance of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #47

[00435] 48. Secondary: PK80 - Incremental Recovery of VWF:RCo [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 144: Secondary Outcome #47

Measure Type Secondary

Measure Title PK80 - Incremental Recovery of VWF:RCo

Measure Description Incremental Recovery (IR) at the maximum plasma concentration

Area under the plasma concentration curve (AUC) from time 0 to infinity of von Willebrand Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 145: Reporting Groups

I Description

PK80 Arm j Comprised of participants who underwent PK analysis of study product (80 i IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm I 3 [PK80+Treatment]. Participants in this arm have received at least one PK j infusion and have provided data suitable for PK analysis. Only PK data I included in this arm.

Table 146: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Incremental Recovery of VWF:RCo

[Units: (U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

1.8

PK1 of rVWF [N=15]

(1.7 to 2.2)

1.8

PK2 of rVWF [N=13]

( 1.6 1ο 2.0)

No statistical analysis provided for PK80 - Incremental Recovery of VWF:RCo Secondary Outcome Measures: Secondary Outcome #48

[00436] 49. Secondary: PK80 - Elimination Phase Half-Life of VWF:Co [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 147: Secondary Outcome #48

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout. Tabel 148: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 149: Measured Values

No statistical analysis provided for PK80 - Elimination Phase Half-Life of VWF:Co

Secondary Outcome Measures: Secondary Outcome #49

[00437] 50. Secondary: PK80 - Volume of Distribution at Steady State of VWF:RCo

[Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 150: Secondar Outcome #50

Measure Type Secondary

Measure Title PK80 - Volume of Distribution at Steady State of VWF:RCo

Measure Description Volume of Distribution at Steady State (Vss) of von Willebrand

Factor Ristocetin cofactor (VWF:RCo) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor

Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study. PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 151: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 152: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Volume of Distribution at Steady State of VWF:RCo

[Units: dL/kg]

Median (95% Confidence Interval)

0.78

PK1 of rVWF [N=15]

(0.58 to 0.86)

0.75

PK2 of rVWF [N=13]

(0.58 to 1.01)

No statistical analysis provided for PK80 - Volume of Distribution at Steady State of VWF:RCo

Secondary Outcome Measures: Secondary Outcome #50

[00438] 51. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 153: Secondary Outcome #50

Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 154: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 155: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Area Under the Plasma Concentration/Time Curve From Time 0

to Infinity (AUC0-∞/Dose) of VWF:Ag

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

66.6

PK1 of rVWF [N=15]

(50.4 to 89.4)

86.9

PK2 of rVWF [N=13]

(54.9 to 100.5)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO-oo/Dose) of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #51

[00439] 52. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:Ag [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 156: Secondary Outcome #51

Measure Type Secondary

PK80 - Area Under the Plasma Concentration/Time Curve From

Measure Title

Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:Ag

Measure Description Area under the plasma concentration curve (AUC) from time 0 to 96 hours of von Willebrand Factor Antigen (VWF:Ag) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2] . Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 158: Measured Values

PK80 Arm

Participants Analyzed 15

:PK80 - Area Under the Plasma Concentration/Time Curve From Time 0

!to 96 Hours (AUC0-96h/Dose) of VWF:Ag

!| Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

61.3

PK1 of rVWF [N=15]

(48.8 to 73.7)

77.4

PK2 of rVWF [N=13]

(53.0 to 87.6)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:Ag Secondary Outcome Measures: Secondary Outcome #52

[00440] 53. Secondary: PK80 - Mean Residence Time of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 159: Secondary Outcome #52

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout. Table 160: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 161: Measured Values

\ P 80 Arm

Participants Analyzed 15

K80 - Mean Residence Time of VWF:Ag

j [Units: Hours]

Median (95% Confidence Interval) 38.4

PK1 of rVWF [N=15]

(31.9 to 48.1)

36.9

PK2 of rVWF [N=13]

(30.0 to 50.8)

No statistical analysis provided for PK80 - Mean Residence Time of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #53

[00441] 54. Secondary: PK80 - Clearance of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 162: Secondar Outcome #54

Table 164: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Clearance of VWF:Ag

[Units: dL/kg/hours]

Median (95% Confidence Interval)

0.015

PK1 of rVWF [N=15]

(0.011 to 0.020)

0.012

PK2 of rVWF [N=13]

(0.010 to 0.018)

No statistical analysis provided for PK80 - Clearance of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #54

[00442] 55. Secondary: PK80 - Incremental Recovery of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 166: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 167: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Incremental Recovery of VWF:Ag

[Units: (U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

2.2

PK1 of rVWF [N=15]

(1.9 to 2.6)

2.4

PK2 of rVWF [N=13]

(2.0 to 2.9)

No statistical analysis provided for PK80 - Incremental Recovery of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #55

[00443] 56. Secondary: PK80 - Elimination Phase Half-Life of VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 168: Secondary Outcome #56

(participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 169 : Reportin Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 170: Measured Values

No statistical analysis provided for PK80 - Elimination Phase Half-Life of VWF:Ag

Secondary Outcome Measures: Secondary Outcome #56

[00444] 57. Secondary: PK80 - Volume of Distribution at Steady State of

VWF:Ag [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 171: Secondary Outcome #57

Measure Type Secondary

Measure Title PK80 - Volume of Distribution at Steady State of VWF:Ag

Measure Description Volume of Distribution at Steady State (Vss) of von Willebrand

Factor Antigen (VWF: Ag) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 172: Reporting Groups

Description

Comprised of participants who underwent PK analysis of study product (80 IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 173: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Volume of Distribution at Steady State of VWF:Ag

[Units: dL/kg]

Median (95% Confidence Interval)

0.55

PK1 of rVWF [N=15]

(0.46 to 0.61)

0.50

PK2 of rVWF [N=13]

(0.41 to 0.57)

No statistical analysis provided for PK80 - Volume of Distribution at Steady State of VWF:Ag Secondary Outcome Measures: Secondary Outcome #57

[00445] 58. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Description

Comprised of participants who underwent PK analysis of study product (80 IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment] . Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 176: Measured Values

i PKSO Arm Participants Analyzed 15

PK80 - Area Under the Plasma Concentration/Time Curve From Time 0

to Infinity (AUC0-∞/Dose) of VWF:CB

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

73.9

PK1 of rVWF [N=15]

(57.3 to 96.2)

90.8

PK2 of rVWF [N=13]

(66.0 to 105.2)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO-oo/Dose) of VWF:CB

Secondary Outcome Measures: Secondary Outcome #58

[00446] 59. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:CB [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 177: Secondary Outcome #58

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 178: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment] . Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 188: Measured Values

P 80 Arm

Participants Analyzed 15

PK80 - Area Under the Plasma Concentration/Time Curve From Time 0

to 96 Hours (AUC0-96h/Dose) of VWF:CB

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

71.9

PK1 of rVWF [N=15]

(57.0 to 89.8)

88.1

PK2 of rVWF [N=13]

(63.8 to 96.3)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of VWF:CB

Secondary Outcome Measures: Secondary Outcome #59

[00447] 60. Secondary: PK80 - Mean Residence Time of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.] Table 189: Secondary Outcome #59

I Measure Type I Secondary

I Measure Title | PK80 - Mean Residence Time of VWF: CB

i Measure Description i Mean Residence Time (MRT) of von Willebrand Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 190: Reporting Groups

I Description

PK80 Arm I Comprised of participants who underwent PK analysis of study product (80

i IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm j 3 [PK80+Treatment] . Participants in this arm have received at least one PK I infusion and have provided data suitable for PK analysis. Only PK data

I included in this arm.

Table 191: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Mean Residence Time of VWF:CB

[Units: Hours]

Median (95% Confidence Interval)

30.9

PK1 of rVWF [N=15]

(24.3 to 35.0)

28 7

PK2 of rVWF [N=13]

(25.6 to 37.2)

No statistical analysis provided for PK80 - Mean Residence Time of VWF:CB

Secondary Outcome Measures: Secondary Outcome #60

[00448] 61. Secondary: PK80 - Clearance of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 194: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Clearance of VWF:CB

[Units: dL/kg/hours]

Median (95% Confidence Interval)

0.014

PK1 of rVWF [N=15]

(0.010 to 0.017)

0.011

PK2 of rVWF [N=13]

(0.010 to 0.015) No statistical analysis provided for PK80 - Clearance of VWF:CB

Secondary Outcome Measures: Secondary Outcome #61

[00449] 62. Secondary: PK80 - Incremental Recovery of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 195: Secondar Outcome #61

Table 196: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 197: Measured Values

PK80 Arm

jParticipants Analyzed 15

No statistical analysis provided for PK80 - Incremental Recovery of VWF:CB

Secondary Outcome Measures: Secondary Outcome #62

[00450] 63. Secondary: PK80 - Elimination Phase Half-Life of VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 198: Secondary Outcome #62

Table 199: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80 IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

No statistical analysis provided for PK80 - Elimination Phase Half-Life of VWF:CB

Secondary Outcome Measures: Secondary Outcome #63

[00451] 64. Secondary: PK80 - Volume of Distribution at Steady State of

VWF:CB [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 201: Secondary Outcome #63

Measure Type Secondary

Measure Title PK80 - Volume of Distribution at Steady State of VWF:CB

Measure Description Volume of Distribution at Steady State (Vss) of von Willebrand

Factor Collagen Binding (VWF:CB) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor

Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category. PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 202: Reporting Groups

Description

Comprised of participants who underwent PK analysis of study product (80 IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment] . Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm. Table 203: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Volume of Distribution at Steady State of VWF:CB

[Units: dL/kg]

Median (95% Confidence Interval)

0.39

PK1 of rVWF [N=15]

(0.34 to 0.46)

0.36

PK2 of rVWF [N=13]

(0.33 to 0.40)

No statistical analysis provided for PK80 - Volume of Distribution at Steady State of VWF:CB

Secondary Outcome Measures: Secondary Outcome #64

[00452] 65. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUC0-∞/Dose) of FVIIF C [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 204: Secondary Outcome #65

i Measure Type ! Secondary PK80 - Area Under the Plasma Concentration/Time Curve From

Measure Title

Time 0 to Infinity (AUC0-∞/Dose) of FVIIFC

Measure Description Area under the plasma concentration curve (AUC) from time 0 to infinity of Factor VIII activity (FVIIFC) after infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. Category title includes number of participants [N] who provided data for the category.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.

Table 205 : Reporting Groups

Description

Comprised of participants who underwent PK analysis of study product (80 IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 206: Measured Values

PK80 Arm

Participants Analyzed 15

PK80 - Area Under the Plasma Concentration/Time Curve From Time 0

to Infinity (AUC0-∞/Dose) of FVIII:C

[Units: (hours * U/dL)/(U VWF: RCo/kg)]

Median (95% Confidence Interval)

96.8

PK1 of rVWF [N=15]

(64.0 to 126.5)

94.8

PK2 of rV F [N=13]

(60.4 to 106.5)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to Infinity (AUCO-oo/Dose) of FVIII:C Secondary Outcome Measures: Secondary Outcome #65

[00453] 66. Secondary: PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of FVIIFC [Time Frame: PK evaluations at pre- infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 208: Reporting Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm.

Table 209: Measured Values

PK80 Arm

Participants Analyzed 15

IPK80 - Area Under the Plasma Concentration/Time Curve From Time 0

!to 96 Hours (AUC0-96h/Dose) of FVIII:C

{Units: (hours * U/dL)/(U VWF: RCo/kg)] Median (95% Confidence Interval)

81 7

PK1 of rVWF [N=15]

(54.7 to 104.3) 71 8

PK2 of rVWF [N=13]

(49.6 to 89.2)

No statistical analysis provided for PK80 - Area Under the Plasma Concentration/Time Curve From Time 0 to 96 Hours (AUC0-96h/Dose) of FVIII:C

Secondary Outcome Measures: Secondary Outcome #66

[00454] 67. Secondary: PK80- Ratio of Intra-participant PK of VWF:RCo, VWF:Ag and VWF:CB at Baseline and After 6 Months [Time Frame: PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout.]

Table 210: Secondary Outcome #66

Measure Type Secondary

PK80- Ratio of Intra-participant PK of VWF:RCo, VWF:Ag and

Measure Title

VWF:CB at Baseline and After 6 Months

Measure Description Area under the plasma concentration curve (AUC) from time 0 to infinity per dose (AUC0-∞/dose) for von Willebrand Factor Ristocetin cofactor (VWF:RCo), von Willebrand Factor Antigen (VWF:Ag) and von Willebrand Factor Collagen Binding

(VWF:CB). Each parameter was compared between the two PK assessments after infusion of 80 IU/kg recombinant von Willebrand Factorvon Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] for participants in the PK80 arm (participants from Arm 3 with PK80 data only). PK assessment conducted at first infusion of 80 IU/kg rWVF [PK1] and the second infusion of 80 IU/kg rVWF after participants were treated on demand for bleeding episodes for at least 6 months since their first infusion of study product [PK2]. 13 participants had data available for this endpoint i.e. data for PK1 and PK2.

PK evaluations at pre-infusion, then at 15, 30 and 60 mins, and 3, 6, 12, 24, 30, 48, 72 and 96 hrs post-infusion. PK evaluation timeframe

Time Frame

for 28 ± 3 days after first infusion of study product which includes PK evaluation for both infusions and washout. Table 211: Population Description

Participants from the PK80 Arm who had pharmacokinetic (PK) data available after both the first infusion of 80 IU/kg recombinant von Willebrand Factor: von Willebrand Factor Ristocetin cofactor (VWF:RCo rVWF) [rVWF] [PK1] and the second infusion of 80 IU/kg rVWF [PK2]. l abel 212: Reportin Groups

Description

PK80 Arm Comprised of participants who underwent PK analysis of study product (80

IU/kg recombinant von Willebrand Factor [rVWF]) i.e. participants from Arm 3 [PK80+Treatment]. Participants in this arm have received at least one PK infusion and have provided data suitable for PK analysis. Only PK data included in this arm. Tabel 213: Measured Values

PK80 Arm

Participants Analyzed 13

PK80- Ratio of Intra-participant PK of VWF:RCo, VWF:Ag and

VWF:CB at Baseline and After 6 Months

[Units: Ratio of AUC0-∞/dose]

Geometric Mean (90% Confidence Interval)

0.9587

AUC0-oo/dose - VWF:RCo

(0.8466 to 1.0857)

1.0914

AUC0-oo/dose - VWF:Ag

(1.0132 to 1.1757)

1.0666

AUC0-oo/dose - VWF:CB

(1.0004 to 1.1372)

No statistical analysis provided for PK80- Ratio of Intra-participant PK of VWF:RCo, VWF:Ag and VWF:CB at Baseline and After 6 Months

[00455] The examples set forth above are provided to give those of ordinary skill in the art a complete disclosure and description of how to make and use the embodiments of the compositions, systems and methods of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Modifications of the above-described modes for carrying out the invention that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All patents and publications mentioned in the specification are indicative of the levels of skill of those skilled in the art to which the invention pertains. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually.

[00456] All headings and section designations are used for clarity and reference purposes only and are not to be considered limiting in any way. For example, those of skill in the art will appreciate the usefulness of combining various aspects from different headings and sections as appropriate according to the spirit and scope of the invention described herein.

[00457] All references cited herein are hereby incorporated by reference herein in their entireties and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.

[00458] Many modifications and variations of this application can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments and examples described herein are offered by way of example only, and the application is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which the claims are entitled.