STABILIZATION OF IMMUNOGLOBULINS AND OTHER PROTEINS THROUGH AQUEOUS FORMULATIONS WITH SODIUM CHLORIDE AT WEAK ACIDIC TO NEUTRAL ph

- Baxter Healthcare S.A.

The present invention provides, among other aspects, storage stabile aqueous formulations of labile proteins at a mildly acidic to neutral pH. The present invention also provides methods for stabilizing a labile therapeutic protein composition at a mildly acidic to neutral pH. Advantageously, the methods and formulations provided herein allow stabile aqueous compositions of labile proteins at mildly acidic to neutral pH useful for parenteral administration.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCES TO RELATED APPLICATIONS

The present application claims the benefit of U.S. Provisional Application No. 61/384,209, filed Sep. 17, 2010, the content of which is expressly incorporated herein by reference in its entirety for all purposes.

BACKGROUND OF THE INVENTION

Biologics are medicinal products created by biological processes, including preparations isolated from natural sources (e.g., human plasma) and recombinant DNA technologies. Within the healthcare and pharmaceutical industries, biologics are becoming increasingly important for patient treatment and overall revenue growth (Goodman M. Nat Rev Drug Discov. (2009) November; 8(11):837). One important class of biologic drugs is therapeutic proteins, both isolated from natural sources and recombinantly produced. For example, plasma proteins are manufactured for therapeutic administration by isolation from pooled human plasma (e.g., GAMMAGARD LIQUID® [IVIG, Immune Globulin Intravenous (Human) 10%]; Baxter International, Deerfield, Ill.) and recombinant means (e.g., ADVATE® [Antihemophilic Factor (Recombinant), Plasma/Albumin-Free Method]; Baxter International, Deerfield, Ill.).

The administration of therapeutic proteins are primarily performed by intravenous (IV), subcutaneous (SQ), and intramuscular administration, although other routes of administration may be used depending upon the therapeutic protein and condition being treated. Most of the immunoglobulins are administered intravenously as larger volumes can be delivered rapidly by the intravenous route to provide the physiologic levels of IgG needed for the effective treatment of various diseases, such as primary immune deficiencies (PID), immune (idiopathic) thrombocytopenic purpura (ITP) and the Kawasaki syndrome. Due to the nature of IV administration, therapy via this route is a slow and timely process, leading to problems with patient compliance.

Subcutaneous (SQ) administration of therapeutic proteins is a convenient alternative to intravenous administration. Compared to IV infusions, SQ administration has several advantages. For example, it can reduce the incidence of systemic reactions, it does not require sometimes-difficult IV access, and gives patients more independence.

In order to improve patient compliance, it would be convenient to provide the protein in a liquid ready to use formulation. However, many human or humanized therapeutic proteins are highly unstable when formulated at or near neutral pH. A variety of degradation pathways exist for proteins especially in liquid formulations, implicating both chemical and physical instability. Chemical instability includes deamination, aggregation, clipping of the peptide backbone, and oxidation of methionine residues. Physical instability encompasses many phenomena, including, for example, aggregation. Protein instability is particularly problematic for labile proteins that are unstable at mildly acidic to neutral pH. To combat these issues, intravenously administrable immunoglobulins have been formulated at acidic pH, effectively increasing their stability in the formulation (products that are formulated at acidic pH are, e.g., Gamunex (Talecris), Gammagard Liquid (Baxter) or Privigen (CSL).

To combat these issues, therapeutic protein compositions are often formulated at acidic pH, effectively increasing their stability in the formulation. Unfortunately, scientific publications have reported that, for example, intramuscular administration of acidic aqueous preparations can cause pain, and potentially could result in tissue damage (Steen et al., 2001; Sluka et al., 2000, the disclosures of which are incorporated by reference herein in their entireties for all purposes). In other cases, where aqueous formulations have been found not to adequately stabilize the therapeutic proteins, lyophilized formulations are used which must be reconstituted prior to administration. In both cases, these factors can cause a less satisfactory drug administration experience and/or inconvenience for the patient, resulting in reduced patient compliance.

As such, there is a need in the art for formulations and methods of formulation that stabilize these labile therapeutic proteins in compositions (e.g., aqueous compositions) at mildly acidic to neutral pH. The present invention satisfies these and other needs by, among other aspects, providing immunoglobulin compositions formulated with histidine at mild acidic to neutral pH that stabilize labile therapeutic proteins.

BRIEF SUMMARY OF INVENTION

The present invention is based in part by the surprising finding that the addition of alkali metal salts to aqueous formulations at mildly acidic to neutral pH stabilizes both plasma-derived and recombinant labile therapeutic proteins. As shown by the present studies, immunoglobulins and coagulation factors formulated at mildly acidic to neutral pH with less than 75 mM of an alkali metal chloride salt are highly unstable. However, the addition of greater than 75 mM of an alkali metal chloride salt (e.g., 100 mM or 150 mM sodium chloride) unexpectedly stabilizes these formulations. This finding is contrary to the result of adding alkali metal chloride salts to formulations at acidic pH values, which is shown herein to destabilize these compositions.

Advantageously, the ability to stably formulate labile proteins (e.g., immunoglobulins, coagulation factors, etc.) at mildly acidic and neutral pH allows for the production of therapeutic formulations that are simpler to self-administer. Furthermore, the ability to stably formulate labile therapeutic proteins at mildly acidic to neutral pH allows for the manufacture of pharmaceutical compositions that may be administered subcutaneously (SQ) or intramuscularly (IM) without the pain and potential for tissue damage that is associated with the SQ and IM administration of compositions formulated at acidic pH.

In one aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.5.

In a specific embodiment of the compositions provided above, the composition comprises from 100 mM to 200 mM of an alkali metal chloride salt.

In a specific embodiment of the compositions provided above, the composition comprises from 125 mM to 175 mM of an alkali metal chloride salt.

In a specific embodiment of the compositions provided above, the composition comprises 150±15 mM an alkali metal chloride salt.

In a specific embodiment of the compositions provided above, the alkali metal chloride salt is sodium chloride.

In a specific embodiment of the compositions provided above, the alkali metal chloride salt is potassium chloride.

In a specific embodiment of the compositions provided above, the amino acid is selected from the group consisting of glycine, proline, and histidine.

In a specific embodiment of the compositions provided above, the amino acid is glycine.

In a specific embodiment of the compositions provided above, the amino acid is proline.

In a specific embodiment of the compositions provided above, the amino acid is histidine.

In a specific embodiment of the compositions provided above, the concentration of the amino acid is from 50 mM to 500 mM.

In a specific embodiment of the compositions provided above, the concentration of the amino acid is from 100 mM to 400 mM.

In a specific embodiment of the compositions provided above, the concentration of the amino acid is from 150 mM to 350 mM.

In a specific embodiment of the compositions provided above, the concentration of the amino acid is from 200 mM to 300 mM.

In a specific embodiment of the compositions provided above, the concentration of the amino acid is from 225 mM to 275 mM.

In a specific embodiment of the compositions provided above, the concentration of the amino acid is 250±10 mM.

In a specific embodiment of the compositions provided above, the pH of the composition is from 5.5 to 7.0.

In a specific embodiment of the compositions provided above, the pH of the composition is from 5.5 to 6.5.

In a specific embodiment of the compositions provided above, the pH of the composition is from 5.5 to 6.0.

In a specific embodiment of the compositions provided above, the pH of the composition is from 6.0 to 7.5.

In a specific embodiment of the compositions provided above, the pH of the composition is from 6.0 to 7.0.

In a specific embodiment of the compositions provided above, the pH of the composition is from 6.0 to 6.5.

In a specific embodiment of the compositions provided above, the pH of the composition is from 6.5 to 7.5.

In a specific embodiment of the compositions provided above, the pH of the composition is from 6.5 to 7.0.

In a specific embodiment of the compositions provided above, the pH of the composition is from 7.0 to 7.5.

In a specific embodiment of the compositions provided above, the labile therapeutic protein is a human or humanized protein.

In a specific embodiment of the compositions provided above, the labile therapeutic protein is an immunoglobulin.

In a specific embodiment of the compositions provided above, the immunoglobulin is an IgG immunoglobulin.

In a specific embodiment of the compositions provided above, the immunoglobulin is a polyclonal immunoglobulin.

In a specific embodiment of the compositions provided above, the immunoglobulin is a monoclonal immunoglobulin.

In a specific embodiment of the compositions provided above, the immunoglobulin is a recombinant immunoglobulin.

In a specific embodiment of the compositions provided above, the immunoglobulin is enriched from pooled human plasma.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is 50±5 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is less than 50 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is at least 50 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is from 50 g/L to 150 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is 100±10 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is at least 100 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is 150±15 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is from 150 g/L to 250 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is 200±20 g/L.

In a specific embodiment of the compositions provided above, the concentration of the immunoglobulin is at least 200 g/L.

In a specific embodiment of the compositions provided above, at least 95% of the protein in the composition is immunoglobulin.

In a specific embodiment of the compositions provided above, at least 95% of the protein in the composition is IgG immunoglobulin.

In a specific embodiment of the compositions provided above, at least 98% of the protein in the composition is IgG immunoglobulin.

In a specific embodiment of the compositions provided above, the composition is stable for at least 6 months when stored at between about 28° C. and about 32° C.

In a specific embodiment of the compositions provided above, the composition is stable for at least 1 year when stored at between about 28° C. and about 32° C.

In a specific embodiment of the compositions provided above, the composition is stable for at least 2 years when stored at between about 28° C. and about 32° C.

In a specific embodiment of the compositions provided above, the composition is stable for at least 1 month when stored at between about 38° C. and about 42° C.

In a specific embodiment of the compositions provided above, the composition is stable for at least 3 months when stored at between about 38° C. and about 42° C.

In a specific embodiment of the compositions provided above, the composition is stable for at least 6 months when stored at between about 38° C. and about 42° C.

In a specific embodiment of the compositions provided above, the composition is stable for at least 1 year when stored at between about 28° C. and about 32° C.

In a specific embodiment of the compositions provided above, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is between 0% and 5%.

In a specific embodiment of the compositions provided above, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is between 0% and 2%.

In a specific embodiment of the compositions provided above, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is from 0% to 5% and the percentage of immunoglobulin in the monomeric state is from 80% to 100%.

In a specific embodiment of the compositions provided above, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is from 0% to 2% and the percentage of immunoglobulin in the monomeric state is from 85% to 100%.

In a specific embodiment of the compositions provided above, the labile therapeutic protein is a coagulation factor.

In a specific embodiment of the compositions provided above, the coagulation factor is selected from the group consisting of Factor VII, Factor VIII, Factor IX, and von Willebrand Factor (vWF).

In a specific embodiment of the compositions provided above, the coagulation factor is Factor VIII.

In a specific embodiment of the compositions provided above, the pH of the composition is between about 6.0 and about 7.0.

In a specific embodiment of the compositions provided above, the pH of the composition is 6.5±0.2.

In a specific embodiment of the compositions provided above, the composition retains at least 80% of its Factor VIII activity when stored at a temperature between about 2° C. and about 8° C. for at least 1 month.

In a specific embodiment of the compositions provided above, the composition retains at least 80% of its Factor VIII activity when stored at a temperature between about 2° C. and about 8° C. for at least 2 months.

In a specific embodiment of the compositions provided above, the composition retains at least 80% of its Factor VIII activity when stored at a temperature between about 2° C. and about 8° C. for at least 3 months.

In a specific embodiment of the compositions provided above, the coagulation factor is Factor VII.

In a specific embodiment of the compositions provided above, the coagulation factor is Factor IX.

In a specific embodiment of the compositions provided above, the coagulation factor is von Willebrand Factor (vWF).

In a specific embodiment of the compositions provided above, the coagulation factor is a protein K-dependent coagulation complex.

In a specific embodiment of the compositions provided above, the protein K-dependent coagulation complex comprises the coagulation factors Factor II, Factor IX, and Factor X.

In a specific embodiment of the compositions provided above, the protein K-dependent coagulation complex further comprises Factor VII.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 3 months in an aqueous formulation containing: from 0 mM to 50 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 2 months in an aqueous formulation containing: from 0 mM to 50 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 1 month in an aqueous formulation containing: from 0 mM to 50 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 2 weeks in an aqueous formulation containing: (a) from 0 mM to 50 mM of an alkali metal chloride salt; (b) an amino acid; and (c) a pH of from 5.5 to 7.

In a specific embodiment of the compositions provided above, the composition is formulated for subcutaneous and/or intramuscular administration.

In another aspect, the present invention provides a method for stabilizing an aqueous composition of a labile therapeutic protein, the method comprising formulating the composition at a pH between 5.5 and 7.0, wherein the formulated composition comprises: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; and an amino acid.

In a specific embodiment of the methods provided above, the formulated composition comprises from 100 mM to 200 mM of an alkali metal chloride salt.

In a specific embodiment of the methods provided above, the formulated composition comprises from 125 mM to 175 mM of an alkali metal chloride salt.

In a specific embodiment of the methods provided above, the formulated composition comprises 150±15 mM an alkali metal chloride salt.

In a specific embodiment of the methods provided above, the alkali metal chloride salt is sodium chloride.

In a specific embodiment of the methods provided above, the alkali metal chloride salt is potassium chloride.

In a specific embodiment of the methods provided above, the amino acid is selected from the group consisting of glycine, proline, and histidine.

In a specific embodiment of the methods provided above, the amino acid is glycine.

In a specific embodiment of the methods provided above, the amino acid is proline.

In a specific embodiment of the methods provided above, the amino acid is histidine.

In a specific embodiment of the methods provided above, the formulated composition comprises from 50 mM to 500 mM of the amino acid.

In a specific embodiment of the methods provided above, the formulated composition comprises from 100 mM to 400 mM of the amino acid.

In a specific embodiment of the methods provided above, the formulated composition comprises from 150 mM to 350 mM of the amino acid.

In a specific embodiment of the methods provided above, the formulated composition comprises from 200 mM to 300 mM of the amino acid.

In a specific embodiment of the methods provided above, the formulated composition comprises from 225 mM to 275 mM of the amino acid.

In a specific embodiment of the methods provided above, the formulated composition comprises from 250±10 mM of the amino acid.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 5.5 to 7.0.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 5.5 to 6.5.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 5.5 to 6.0.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 6.0 to 7.5.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 6.0 to 7.0.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 6.0 to 6.5.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 6.5 to 7.5.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 6.5 to 7.0.

In a specific embodiment of the methods provided above, the pH of the formulated composition is from 7.0 to 7.5.

In a specific embodiment of the methods provided above, the labile therapeutic protein is a human or humanized protein.

In a specific embodiment of the methods provided above, the labile therapeutic protein is an immunoglobulin.

In a specific embodiment of the methods provided above, the immunoglobulin is an IgG immunoglobulin.

In a specific embodiment of the methods provided above, the immunoglobulin is a polyclonal immunoglobulin.

In a specific embodiment of the methods provided above, the immunoglobulin is a monoclonal immunoglobulin.

In a specific embodiment of the methods provided above, the immunoglobulin is a recombinant immunoglobulin.

In a specific embodiment of the methods provided above, the immunoglobulin is enriched from pooled human plasma.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is 50±5 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is less than 50 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is at least 50 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is from 50 g/L to 150 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is 100±10 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is at least 100 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is 150±15 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is from 150 g/L to 250 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is 200±20 g/L.

In a specific embodiment of the methods provided above, the concentration of the immunoglobulin is at least 200 g/L.

In a specific embodiment of the methods provided above, at least 95% of the protein in the formulated composition is immunoglobulin.

In a specific embodiment of the methods provided above, at least 95% of the protein in the formulated composition is IgG immunoglobulin.

In a specific embodiment of the methods provided above, at least 98% of the protein in the formulated composition is IgG immunoglobulin.

In a specific embodiment of the methods provided above, the formulated composition is stable for at least 6 months when stored at between about 28° C. and about 32° C.

In a specific embodiment of the methods provided above, the formulated composition is stable for at least 1 year when stored at between about 28° C. and about 32° C.

In a specific embodiment of the methods provided above, the formulated composition is stable for at least 2 years when stored at between about 28° C. and about 32° C.

In a specific embodiment of the methods provided above, the formulated composition is stable for at least 1 month when stored at between about 38° C. and about 42° C.

In a specific embodiment of the methods provided above, the formulate composition is stable for at least 3 months when stored at between about 38° C. and about 42° C.

In a specific embodiment of the methods provided above, the formulated composition is stable for at least 6 months when stored at between about 38° C. and about 42° C.

In a specific embodiment of the methods provided above, the formulated composition is stable for at least 1 year when stored at between about 28° C. and about 32° C.

In a specific embodiment of the methods provided above, the formulated composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is between 0% and 5%.

In a specific embodiment of the methods provided above, the formulated composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is between 0% and 2%.

In a specific embodiment of the methods provided above, the formulated composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is from 0% to 5% and the percentage of immunoglobulin in the monomeric state is from 80% to 100%.

In a specific embodiment of the methods provided above, the formulated composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is from 0% to 2% and the percentage of immunoglobulin in the monomeric state is from 85% to 100%.

In a specific embodiment of the methods provided above, the labile therapeutic protein is a coagulation factor.

In a specific embodiment of the methods provided above, the coagulation factor is selected from the group consisting of Factor VII, Factor VIII, Factor IX, and von Willebrand Factor (vWF).

In a specific embodiment of the methods provided above, the coagulation factor is Factor VIII.

In a specific embodiment of the methods provided above, the pH of the composition is between about 6.0 and about 7.0.

In a specific embodiment of the methods provided above, the pH of the composition is 6.5±0.2.

In a specific embodiment of the methods provided above, the formulated composition retains at least 80% of its Factor VIII activity when stored at a temperature between about 2° C. and about 8° C. for at least 1 month.

In a specific embodiment of the methods provided above, the formulated composition retains at least 80% of its Factor VIII activity when stored at a temperature between about 2° C. and about 8° C. for at least 2 months.

In a specific embodiment of the methods provided above, the formulated composition retains at least 80% of its Factor VIII activity when stored at a temperature between about 2° C. and about 8° C. for at least 3 months.

In a specific embodiment of the methods provided above, the coagulation factor is Factor VII.

In a specific embodiment of the methods provided above, the coagulation factor is Factor IX.

In a specific embodiment of the methods provided above, the coagulation factor is von Willebrand Factor (vWF).

In a specific embodiment of the methods provided above, the coagulation factor is a protein K-dependent coagulation complex.

In a specific embodiment of the methods provided above, the protein K-dependent coagulation complex comprises the coagulation factors Factor II, Factor IX, and Factor X.

In a specific embodiment of the methods provided above, the protein K-dependent coagulation complex further comprises Factor VII.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 3 months in an aqueous formulation containing: from 0 mM to 50 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 2 months in an aqueous formulation containing: from 0 mM to 50 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 1 month in an aqueous formulation containing: from 0 mM to 50 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 2 weeks in an aqueous formulation containing: from 0 mM to 50 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.

In a specific embodiment of the methods provided above, the composition is formulated for subcutaneous and/or intramuscular administration.

BRIEF DESCRIPTION OF DRAWINGS

FIG. 1. Aggregation of 20% IgG formulations after 6 months storage at 38 to 42° C.

FIG. 2. Aggregate formation in Tetabulin NG formulated at neutral pH in the presence of 150 mM sodium chloride during storage at 28 to 32° C.

FIG. 3. Aggregate formation in Partobulin NG formulated at neutral pH in the presence of 150 mM sodium chloride during storage at 28 to 32° C.

FIG. 4. Differences in Tetanus Anti-toxin potency of Tetabulin NG formulated with 150 mM sodium chloride in a pH range of 5.5 to 7.5 during storage at 28 to 32° C.

FIG. 5. Differences in Anti-D potency of Partobulin NG formulated with 150 mM sodium chloride in a pH range of 5.5 to 7.5 during storage at 28° C. to 32° C.

FIG. 6. Anti-MIF titer after three months storage at 38° C. to 42° C.

FIG. 7. The Factor VIII activity during 12 weeks storage of the aqueous formulation at 2 to 8° C. without sodium chloride.

FIG. 8. The Factor VIII activity during 12 weeks storage of the aqueous formulation at 2 to 8° C. with 150 mM sodium chloride.

DETAILED DESCRIPTION OF INVENTION I. Introduction

Therapeutic proteins are often times formulated at acidic pH or as lyophilized compositions due to their labile nature in aqueous solution at or near neutral pH. As discussed above, these formulations are less convenient, may cause pain and/or tissue damage upon administration, and likely reduce patient compliance. Advantageously, the present invention provides means for stably formulating these labile proteins in aqueous solution at or near neutral pH. In one aspect, the present invention provides labile therapeutic protein compositions stabilized by the addition of moderate levels of alkali metal chloride salts (e.g., 75 mM to 200 mM, preferably 100 mM to 200 mM) to formulations at mildly acidic to neutral pH. The present invention is based in part on the surprising discovery that labile therapeutic proteins are significantly stabilized at mildly acidic to neutral pH by the addition of an alkali metal chloride salt at a final concentration of between about 75 mM and about 200 mM.

Our new studies provided herein demonstrate that purified plasma-derived immunoglobulin preparations formulated in 0.25 M glycine could be stabilized by the addition of sodium chloride in a pH dependent manner. Examples 1 and 2 shows that these immunoglobulin preparations, having a final concentration of between about 90 g/L and about 220 g/L, were stabilized for at least 24 months when stored at a temperature of 28° C. to 32° C., and for at least 6 months when stored at a temperature of 38° C. to 42° C. Maximum stability was observed with addition of 150 mM sodium chloride.

Under these conditions, the addition of sodium chloride to formulations at pH values at and above 7.0 resulted in considerably higher aggregation and fragmentation rates, compared to samples formulated at a pH between 5.5 and 7.0 (Table 5 and Table 6). Similarly, it was previously observed that sodium chloride significantly destabilized immunoglobulin formulations with acidic pH values (under 5.0).

Similar stabilizing effects were demonstrated with the addition of 150 mM sodium chloride to two hyper-immune immunoglobulin preparations, Partobulin® NG and Tetabulin® NG. Example 3 demonstrates that addition of 150 mM sodium chloride to these plasma-derived immunoglobulin preparations results in reduced protein aggregation at pH values between 5.5 and 6.5 and reduced loss of anti-D titer for Partobulin® NG between pH 5.5 and pH 6.5, while stabilizing the Tetanus anti-toxin titer across the entire range of pH investigated.

Next, it was demonstrated that sodium chloride would also stabilize recombinant antibody preparations formulated at mildly acidic to neutral pH. Example 4 shows that a recombinant anti-MIF antibody formulated at pH 5.6 to 6.5 with 0.25 M glycine and 150 mM sodium chloride Was stabilized upon storage at elevated temperatures (38° C. to 42° C.) for six months. Consistent with the previous observations, sodium chloride enhanced degradation when the antibody was formulated at low pH (4.5) and increased aggregation when formulated at a higher pH (pH 7.3).

Finally, to investigate whether or not the stabilizing effect of sodium chloride at mildly acidic to neutral pH can be applied to non-immunoglobulin labile therapeutic proteins, formulations of recombinant Factor VIII (rFVIII) were prepared. As the foundation for the rFVIII formulation, the protein was formulated as in the reconstituted ADVATE® (Baxter International; Deerfield Ill.) product. Despite the presence of several traditional stabilizing agents, including mannitol, trehalose, histidine, calcium chloride, polysorbate-80 and glutathione, rFVIII is extremely unstable, even at 2° C. to 8° C., in aqueous formulation. As such, ADVATE® is marketed as a lyophilized formulation that is reconstituted immediately prior to administration.

Remarkably, as shown in Example 5, the inclusion of 150 mM sodium chloride almost completely stabilizes rFVIII activity at pH 6.0 to 7.0, when stored in aqueous formulation at 2° C. to 8° C. for at least 12 weeks. Accordingly, it has now been demonstrated that intermediate levels of an alkali metal chloride salt can stabilize a wide range of labile therapeutic proteins when formulated at a mildly acidic to neutral pH.

II. Definitions

As used herein, a “labile therapeutic protein” refers to a class of therapeutically useful proteins that are unstable when formulated at mildly acid to neutral pH in the absence of an alkali metal chloride salt. Generally, protein stability can be measured by several different metrics, including aggregation, loss of enzymatic activity, loss of antigenic titer, or degradation. Labile therapeutic proteins will display one or more of these unwanted characteristics when stored at mildly acidic to neutral pH in the absence of an alkali metal chloride salt. The absolute time for which a labile protein is stable will be dependent upon the individual characteristics of the protein, which can be readily determined by the skilled artisan. For example, certain blood coagulations proteins (e.g., Factor VIII) are stable for less than two months under refrigeration when formulated a mildly acidic to neutral pH in the absence of suitable levels of an alkali metal chloride salt. In other cases, for example plasma derived immunoglobulin preparations, a labile therapeutic protein may be stable at room temperature for less than six months when formulated a mildly acidic to neutral pH in the absence of suitable levels of an alkali metal chloride salt, as compared to more than two years in the presence of moderate levels of an alkali metal chloride salt.

In the context of the present invention, a labile protein will show a marked increase in stability upon the addition of an alkali metal chloride salt to a formulation at mildly acid to neutral pH. Inclusion of an alkali metal chloride salt may, for example, reduce aggregation of the labile protein by at least about 20%; maintain at least about 20% more enzymatic activity; maintain at least about 20% more antigenic titer; and/or reduce degradation by at least about 20%, when stored for a given period of time. For the purposes of the present invention, a labile therapeutic protein may be isolated from a natural source (e.g., plasma-derived) or recombinantly produced. For example, in certain embodiments, labile plasma derived blood proteins, such as immunoglobulins, and blood coagulation factors (e.g., Factor VIII) are particularly well suited for formulation as described herein. Non-limiting examples of coagulation proteins include, Factor II (prothrombin), Factor III (platelet tissue factor), Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor XI, Factor XII, Factor XIII, von Willebrand Factor (vWF). Likewise, labile recombinant proteins, such as antibodies and blood coagulation factors may be stabilized according to the formulations and methods provided herein.

As used herein, a “storage stable” aqueous composition refers to a protein solution that has been formulated to increase the stability of the protein in solution, for example by at least 20%, over a given storage time. In the context of the present invention, a labile protein solution formulated at a mildly acidic to neutral pH can be made “storage stable” by the addition of a moderate level (about 75 mM to about 200 mM, preferably about 100 mM to about 200 mM) of an alkali metal chloride salt. The stability of the protein in any given formulation can be measured, for example, by monitoring the formation of aggregates, loss of bulk enzymatic activity, loss of antigenic titer or formation of degradation products, over a period of time. The absolute stability of a formulation, and the stabilizing effects of the alkali metal chloride salt, will vary dependent upon the labile protein being stabilized.

As used herein, the term “time of stability” refers to the length of time a composition is considered stable. For example, the time of stability for a composition may refer to the length of time for which the level of protein aggregation and/or degradation in the composition remains below a certain threshold (e.g., 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, etc.), the length of time a composition maintains an enzymatic activity above a certain threshold (e.g., 100%, 95%, 90, 85, 80, 75, 70, 65, 60, 55, 50, etc. of the amount of activity present in the composition at the start of the storage period), or the length of time a composition maintains an antigenic titer (e.g., 100%, 95%, 90, 85, 80, 75, 70, 65, 60, 55, 50, etc. of the antigenic titer present in the composition at the start of the storage period). In the context of the present invention, a storage stable aqueous composition of a labile therapeutic protein formulated at mildly acidic to neutral pH with a moderate level of an alkali metal chloride salt will have a longer time of stability than a composition of the same labile therapeutic protein formulated at mildly acidic to neutral pH without a moderate level of an alkali metal chloride salt. A storage stable aqueous composition of a labile therapeutic protein, as provided herein, will have a time of stability that is, for example, at least 20% greater than the time of stability for the same composition formulated in the absence of a moderate level of an alkali metal chloride salt, or at least 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190% greater, or at least 2 times greater, or at least 2.5, 3.0 times, 3.5 times, 4.0 times, 4.5 times, 5.0 times, 5.5 times, 6.0 times, 6.5 times, 7.0 times, 7.5 times, 8.0 times, 8.5 times, 9.0 times, 9.5 times, 10 times, or more times greater than the time of stability for the same composition formulated in the absence of a moderate level of an alkali metal chloride salt.

As used herein, the term “stable” refers to a state of a protein composition (e.g., an immunoglobulin solution) suitable for pharmaceutical administration. In the context of the present invention, an immunoglobulin solution is generally considered to be stable when the level of immunoglobulin aggregation and/or degradation in the composition remains below a certain threshold (e.g., below 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, etc.) or the when the antigenic titer remains above a certain threshold (e.g., above 100%, 95%, 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, 50%, etc. of the antigenic titer present in the composition at the start of a storage period).

The European Pharmacopoeia (Ph. Eur.) standard for normal human immunoglobulins is that the composition has: (i) a monomer and dimer content equal to or greater than 85% of the total area of a standard chromatogram; and (ii) a polymer and aggregate sum content of not more than 10% of the total area of the chromatogram. For IGIV the sum of the peak areas of the monomer and dimer represents not less than 95 percent and the maximum amount of polymers and aggregates is no more than 2%. Accordingly, in one embodiment, a storage-stable immunoglobulin composition provided herein is considered to be stable when at least 85% of the immunoglobulin content is monomeric and no more than 5%, preferably no more than 2%, of the immunoglobulin content is aggregated.

As used herein, “storage” means that a formulation is not immediately administered to a subject once prepared, but is kept for a period of time under particular conditions (e.g. at a particular temperature, under a particular atmosphere, protected from light, etc.) prior to use. For example, a liquid formulation can be kept for days, weeks, months or years, prior to administration to a subject under varied temperatures such as refrigerated (0° to 10° C.) or room temperature (e.g., between about 20° C. and 25° C.).

For the purposes of the present invention, when referring to a concentration of an individual component of a composition, the phrases “no more than X” and “from 0 to X” are equivalent and refer to any concentration between and including 0 and X. For example, the phrases “a concentration of no more than 2%” and “a concentration of from 0% to 2%” are equivalent and include 0%, 1%, and 2%.

For the purposes of the present invention, when referring to a concentration of an individual component of a composition, the phrases “no less than X” refers to any concentration X or higher. For example, the phrase “a concentration of no less than 98%” includes 98%, 99%, and 100%.

For the purposes of the present invention, when referring to a concentration of an individual component of a composition, the phrases “between X and Y” and “from X to X” are equivalent and refer to any concentration between and including X and Y. For example, the phrases “a concentration of between 49% and 51%” and “a concentration of from 49% to 51%” are equivalent and include 49%, 50%, and 51%.

As used herein, an “alkali metal chloride salt” refers to an inorganic salt of an alkali metal and chlorine. For the purposes of the present invention, the alkali metal chloride salt will be a pharmaceutically acceptable salt, most commonly sodium or potassium chloride. In a preferred embodiment, the salt is sodium chloride.

Likewise, an “alkali metal cation” will most commonly refer to a sodium cation (Na+) or potassium cation (K+) and can be contributed by an alkali metal chloride salt or other source. In the context of the present invention, a hydrogen ion is not considered an alkali metal cation, and thus the inclusion of hydrochloric acid will not contribute to the alkali metal cation content of the formulation.

As used herein, a “coagulation factor” refers to a protein involved in the intrinsic (contact activation) or extrinsic (tissue factor) pathway of the coagulation cascade. Non-limiting examples of coagulation proteins include, Factor II (prothrombin), Factor III (platelet tissue factor), Factor. V, Factor VII, Factor VIII, Factor IX, Factor X, Factor XI, Factor XII, Factor XIII, von Willebrand Factor (vWF), and the like. Coagulation proteins stabilized by the formulations and methods provided herein may be plasma-derived or recombinantly produced.

As used herein, the term “core coagulation factor” refers to any one of Factor VII, Factor VIII, Factor IX, and von Willebrand Factor (vWF), as well as conservative or natural variants, biologically active fragments, and natural isoforms thereof.

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 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., Anderson 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, Ill., U.S.A.).

As used herein, an “antibody” refers to a polypeptide substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof, which specifically bind and recognize an analyte (antigen). The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD, and IgE, respectively.

An exemplary immunoglobulin (antibody) structural unit is composed of two pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively. In a particular exemplary embodiment, the immunoglobulin will consist of an immunoglobulin preparation isolated from pooled plasma (preferably human plasma) comprising IgG immunoglobulins.

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%. As used herein, about also includes the exact amount. Hence “about 20%” means “about 20%” and also “20%.”

By “therapeutically effective amount or dose” or “sufficient/effective amount or dose,” it is meant a dose that produces effects for which it is administered. 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).

As used herein, a “stabilizing agent” refers to a chemical, other than an alkali metal chloride salt, which aids in the stabilization of a labile therapeutic agent in an aqueous formulation under mildly acid to neutral pH. Examples of suitable stabilizing agents for the formulations and methods provided herein include, without limitation, buffering agents (e.g., TRIS, HEPES, potassium or sodium phosphate, amino acids, etc.), osmolytes (e.g., sugars, sugar alcohols, etc.), bulking agents (e.g., amino acids, etc.), divalent salts, surfactants, and the like.

As used herein, “amino acids” refers to any natural or non-natural pharmaceutically acceptable amino acid. Non-limiting examples of amino acids include, isoleucine, alanine, leucine, asparagine, lysine, aspartic acid, methionine, cysteine, phenylalanine, glutamic acid, threonine, glutamine, tryptophan, glycine, valine, proline, selenocysteine, serine, tyrosine, arginine, histidine, ornithine, taurine, and the like.

Any sugar such as mono-, di-, or polysaccharides, or water-soluble glucans, including for example fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, dextran, trehalose, pullulan, dextrin, cyclodextrin, soluble starch, hydroxyethyl starch, and carboxymethylcellulose may be used.

As used herein, a “sugar alcohol” refers to a hydrocarbon having between about 4 and about 8 carbon atoms and at least one hydroxyl group. Non-limiting examples of sugar alcohols that may be used in formulations provided herein include, mannitol, sorbitol, inositol, galactitol, dulcitol, xylitol, and arabitol.

As used herein, the term “activity” refers to a functional activity or activities of a polypeptide or portion thereof associated with a full-length (complete) protein. Functional activities include, but are not limited to, biological activity, catalytic or enzymatic activity, antigenicity (i.e., the ability to bind or compete with a polypeptide for binding to an anti-polypeptide antibody), immunogenicity, ability to form multimers, and the ability to specifically bind to a receptor or ligand for the polypeptide.

III. Formulations

Among other aspects, the present invention provides stabilized formulations of labile proteins for therapeutic administration. The following embodiments are based in part on the unexpected discovery that the addition of moderate levels of an alkali metal chloride salt (i.e., about 75 mM to about 200 mM, preferably about 100 mM to about 200 mM) to formulations at mildly acidic to neutral pH stabilize various plasma-derived and recombinant proteins that are otherwise labile at these pH values.

The labile therapeutic protein compositions provided by the present invention take advantage of the increased stability afforded when these proteins are formulated at mildly acidic to neutral pH. Generally, this includes pH values between about 5.5 and about 7.5. In a preferred embodiment, the pH value is between about 5.5 and about 7.0. However, the range of pH values at which any individual labile therapeutic protein is stabilized by the addition of a moderate level (i.e., between about 75 mM and about 200 mM, preferably between about 100 mM and about 200 mM) of an alkali metal chloride salt may vary slightly, dependent upon the properties of the individual protein. For example, in one embodiment, a storage stable formulation will have a pH between about 5.5 and about 7.0. In another embodiment, a storage stable formulation will have a pH between about 5.5 and about 6.5. In other embodiments, the pH of the stabilizing formulation will be between about 6.0 and about 7.0. In another embodiment, the pH of the stabilizing formulation will be between about 5.5 and about 6.0. In one embodiment, the pH of the stabilizing formulation will be between about 6.0 and about 6.5. In another embodiment, the pH of the stabilizing formulation will be between about 6.5 and about 7.0. In another embodiment, a storage stable formulation will have a pH between about 6.0 and about 7.5. In another embodiment, a storage stable formulation will have a pH between about 6.5 and about 7.5. In another embodiment, a storage stable formulation will have a pH between about 7.0 and about 7.5. In other embodiments, the pH of the stabilizing formulation is 5.5±0.2, 5.6±0.2, 5.7±0.2, 5.8±0.2, 5.9±0.2, 6.0±0.2, 6.1±0.2, 6.2±0.2, 6.3±0.2, 6.4±0.2, 6.5±0.2, 6.6±0.2, 6.7±0.2, 6.8±0.2, 6.9±0.2, 7.0±0.2, 7.1±0.2, 7.2±0.2, 7.3±0.2, 7.4±0.2, or 7.5±0.2. In other embodiments, the pH of the stabilizing formulation is 5.5±0.1, 5.6±0.1, 5.7±0.1, 5.8±0.1, 5.9±0.1, 6.0±0.1, 6.1±0.1, 6.2±0.1, 6.3±0.1, 6.4±0.1, 6.5±0.1, 6.6±0.1, 6.7±0.1, 6.8±0.1, 6.9±0.1, 7.0±0.1, 7.1±0.1, 7.2±0.1, 7.3±0.1, 7.4±0.1, or 7.5±0.1. In yet other embodiments, the pH of the stabilizing formulation is 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, or 7.5.

In addition to the alkali metal chloride salt, the pharmaceutical compositions provided herein include one or more stabilizing agents. In a specific embodiment, the stabilizing agent is a buffering agent suitable for intravenous, intravitreal, subcutaneous, and/or intramuscular administration. Non-limiting examples of buffering agents suitable for formulating the storage stable compositions provided herein include glycine, histidine, proline, or other amino acids, salts like citrate, phosphate, acetate, glutamate, tartrate, benzoate, lactate, gluconate, malate, succinate, formate, propionate, carbonate, or any combination thereof adjusted to an appropriate pH. Generally, the buffering agent will be sufficient to maintain a suitable pH in the formulation for an extended period of time.

A. Labile Proteins

Among other aspects, the present invention provides stabilized formulations of labile therapeutic proteins for therapeutic administration. The following embodiments are based in part on the unexpected discovery that the formulation of labile therapeutic proteins, including but not limited to immunoglobulin and coagulation factors, with a moderate amount of an alkali metal chloride salt at mildly acidic to neutral pH stabilizes proteins that are otherwise labile at these pH values and/or labile when formulated with an alkali metal chloride salt an acidic pH.

As will be recognized by one of skill in the art, the formulation of a protein composition at a particular pH may introduce residual counter ions contributed from one or more pH modifying agents. For example, the storage stable compositions provided herein may contain chloride anions contributed from hydrochloric acid, acetate anions contributed from glacial acetic acid, sodium cations contributed from sodium hydroxide, and the like. In the context of the present invention, a storage stable labile therapeutic protein composition consisting of or consisting essentially of: a labile therapeutic protein, a moderate concentration of an alkali metal chloride salt, and a stabilizing agent may further comprise one or more counter ion, as necessitated by the formulation process at the desired pH.

In one embodiment, a labile therapeutic protein that may benefit from a formulation or method provided herein is stable for less than 3 months at a temperature between 28° C. and 32° C. when in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 5.5 and 7.0. In another embodiment, the labile therapeutic protein is stable for less than 2 months at a temperature between 28° C. and 32° C. in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 5.5 and 7.0. In another embodiment, the labile therapeutic protein is stable for less than 1 month at a temperature between 28° C. and 32° C. in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 5.5 and 7.0. In yet another embodiment, the labile therapeutic protein is stable for less than 2 weeks at a temperature between 28° C. and 32° C. in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 5.5 and 7.0. In a preferred embodiment, the labile therapeutic protein is an immunoglobulin.

In a related embodiment, a labile therapeutic protein that may benefit from a formulation or method provided herein is stable for less than 3 months at a temperature between 2° C. and 8° C. when in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 6.0 and 7.5. In another embodiment, the labile therapeutic protein is stable for less than 2 months at a temperature between 2° C. and 8° C. in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 6.0 and 7.5. In another embodiment, the labile therapeutic protein is stable for less than 1 month at a temperature between 2° C. and 8° C. in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 6.0 and 7.5. In yet another embodiment, the labile therapeutic protein is stable for less than 2 weeks at a temperature between 2° C. and 8° C. in an aqueous formulation containing less than 50 mM of an alkali metal chloride salt at a pH between 5.5 and 7.5, preferably between 6.0 and 7.5. In a specific embodiment, the labile therapeutic protein is a coagulation factor. In a preferred embodiment, the labile therapeutic protein is Factor VIII.

In preferred embodiments of the invention, the labile therapeutic protein will be a human protein, a chimeric protein (e.g., a mouse/human chimera or rat/human chimera), or a humanized protein. For example, in one preferred embodiment, the labile therapeutic protein will be a recombinant chimeric or humanized monoclonal antibody. In other preferred embodiment, the labile therapeutic protein will be a plasma protein, either recombinant or plasma-derived, e.g., a protein composition isolated from pooled human plasma. Non-limiting examples of plasma-derived labile proteins that may be formulated according to the present invention include: Factor II (prothrombin), Factor III (platelet tissue factor), Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor XI, Factor XII, Factor XIII, and von Willebrand Factor (vWF).

The final concentration of the labile therapeutic protein in the formulations provided herein will be dependent upon many factors, including without limitation, the potency and specific activity of the protein, the disease or condition being treated, the route of administration, and other factors that will be well understood by the skilled practitioner. In one embodiment, the labile therapeutic protein will be formulated at a low protein concentration, for example, between 0.05 mg/mL and 10 mg/mL, or between 0.1 mg/mL and 20 mg/mL, or between 0.5 mg/mL and 10 mg/mL, or between 0.1 mg/mL and 0.5 mg/mL. In other embodiments, the labile therapeutic protein will be formulated at a moderate protein concentration, between 20 mg/mL and 80 mg/mL. For example, between 20 mg/mL and 40 mg/mL. Or between 40 mg/mL and 60 mg/mL. Or between 60 mg/mL and 80 mg/mL. In yet other embodiments, the labile therapeutic protein will be formulated at a high protein concentration, between 80 mg/mL and 250 mg/mL. For example, in one embodiment, the protein concentration will be between 80 mg/mL and 120 mg/mL. In another embodiment, the protein concentration will be between 120 mg/mL and 180 mg/mL. In yet another embodiment, the protein concentration will be between 180 mg/mL and 250 mg/mL.

In certain embodiments, the final protein concentration may be between 0.5% and 25%. In another embodiment, the final protein concentration may be between 0.5% and 20%. In another embodiment, the final protein concentration may be between 0.5% and 15%. In another embodiment, the final protein concentration may be between 0.5% and 10%. In another embodiment, the final protein concentration may be between 0.5% and 5%. In one embodiment, a composition with a final protein concentration as described above will be formulated for intravenous administration.

In certain embodiments, the final protein concentration may be between 5% and 25%. In another embodiment, the final protein concentration may be between 10% and 25%. In another embodiment, the final protein concentration may be between 15% and 25%. In another embodiment, the final protein concentration may be between 20% and 25%. In one embodiment, a composition with a final protein concentration as described above will be formulated for subcutaneous or intramuscular administration.

In certain embodiments, the labile therapeutic protein will be formulated at a final concentration of from 0.05 g/L to 250 g/L. In certain embodiments, the labile protein is formulated at a final concentration of 0.05±0.01 g/L, 0.06±0.01 g/L, 0.07±0.01 g/L, 0.08±0.01 g/L, 0.09±0.01 g/L, 0.1±0.01 g/L, 0.2±0.02 g/L, 0.3±0.03 g/L, 0.4±0.04 g/L, 0.5±0.05 g/L, 0.6±0.06 g/L, 0.7±0.07 g/L, 0.8±0.08 g/L, 0.9±0.09 g/L, 1±0.1 g/L, 210.2 g/L, 3±0.3 g/L, 4±0.4 g/L, 5±0.5 g/L, 6±0.6 g/L, 7±0.7 g/L, 8±0.8 g/L, 9±0.9 g/L, 10±1 g/L, 11±1.1 g/L, 12±1.2 g/L, 13±1.3 g/L, 14±1.4 g/L, 15±1.5 g/L, 16±1.6 g/L, 17±1.7 g/L, 18±1.8 g/L, 19±1.9 g/L, 20±2 g/L, 21±2.1 g/L, 22±2.2 g/L, 23±2.3 g/L, 24±2.4 g/L, 25±2.5 g/L, 26±2.6 g/L, 27±2.7 g/L, 28±2.8 g/L, 29±2.9 g/L, 30±3 g/L, 35±3.5 g/L, 40±4 g/L, 45±4.5 g/L, 50±5 g/L, 55±5.5 g/L, 60±6 g/L, 65±6.5 g/L, 70±7 g/L, 75±7.5 g/L, 80±8 g/L, 85±8.5 g/L, 90±9 g/L, 95±9.5 g/L, 100±10 g/L, 110±11 g/L, 120±12 g/L, 130±13 g/L, 140±14 g/L, 150±15 g/L, 160±16 g/L, 170±17 g/L, 180±18 g/L, 190±19 g/L, 200±20 g/L, 210±21 g/L, 220±22 g/L, 230±23 g/L, 240±24 g/L, 250±25 g/L, or higher concentrations, depending upon the characteristics of the protein being formulated, the intended therapeutic use of the protein, and the preferred method of administration. In yet other embodiments, the final concentration of the labile protein in the formulation is 0.05 g/L, 0.06 g/L, 0.07 g/L, 0.08 g/L, 0.09 g/L, 0.1 g/L, 0.2 g/L, 0.3 g/L, 0.4 g/L, 0.5 g/L, 0.6 g/L, 0.7 g/L, 0.8 g/L, 0.9 g/L, 1 g/L, 2 g/L, 3 g/L, 4 g/L, 5 g/L, 6 g/L, 7 g/L, 8 g/L, 9 g/L, 10 g/L, 11 g/L, 12 g/L, 13 g/L, 14 g/L, 15 g/L, 16 g/L, 17 g/L, 18 g/L, 19 g/L, 20 g/L, 21 g/L, 22 g/L, 23 g/L, 24 g/L, 25 g/L, 26 g/L, 27 g/L, 28 g/L, 29 g/L, 30 g/L, 35 g/L, 40 g/L, 45 g/L, 50 g/L, 55 g/L, 60 g/L, 65 g/L, 70 g/L, 75 g/L, 80 g/L, 85 g/L, 90 g/L, 95 g/L, 100 g/L, 110 g/L, 120 g/L, 130 g/L, 140 g/L, 150 g/L, 160 g/L, 170 g/L, 180 g/L, 190 g/L, 200 g/L, 210 g/L, 220 g/L, 230 g/L, 240 g/L, 250 g/L, or higher

In certain embodiments, the formulations provided herein will stabilize a labile therapeutic protein composition when stored at a temperature between 2° C. and 42° C. In one embodiments, a labile therapeutic protein will be stabilized by the formulations provided herein when stored under refrigeration, i.e., stored at a temperature between 2° C. and 8° C. In another embodiment, a labile therapeutic protein will be stabilized by the formulations provided herein when stored at room temperature, i.e., stored at a temperature between 20° C. and 25° C. In other embodiments, the protein may be stabilized when stored at a temperature between 28° C. and 32° C. In yet another embodiment, the protein may be stabilized when stored at a temperature between 38° C. and 42° C. The temperatures at which a labile therapeutic protein will be stabilized by the formulations provided herein will be dependent upon the characteristics of the individual protein, which can readily be determined by one of skill in the art.

Likewise, the extent of time for which a labile protein is stabilized by the formulations provided herein will depend upon the individual protein. For example, in one embodiment, a storage stable, aqueous composition provided herein will be stable for at least 2 months. In another embodiment, the composition will be stable for at least 3 months. In yet other embodiment, the composition will be stable for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or more months. In a preferred embodiment, the composition will be stable for at least 6 months. In a more preferred embodiment, the composition will be stable for at least 1 year. In another preferred embodiment, the composition will be stable for at least 2 years.

The extent to which a labile therapeutic protein is stabilized by the formulations provided herein may also be expressed as a percentage increase in the time the composition is stable under standard storage conditions. For example, in one embodiment, a labile therapeutic protein composition may be stable under storage conditions for at least 25% longer when formulated with an alkali metal chloride salt at a mildly acidic to neutral pH, as provided herein, as compared to the stability of the same protein under mildly acidic to neutral pH in the absence of the alkali metal chloride salt. In other embodiments, the composition may be stable for at least 50% longer when formulated according to the present invention, or at least 75%, 100%, 150%, 200%, 250%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000%, or longer when formulated according to the present invention. For example, in one embodiment, the labile therapeutic protein composition may be stable under storage conditions for from 25% to 1000% longer when formulated with an alkali metal chloride salt at a mildly acidic to neutral pH, as provided herein, as compared to the stability of the same protein under mildly acidic to neutral pH in the absence of the alkali metal chloride salt. In other embodiments, the composition may be stable for from 50% to 1000%, 100% to 1000%, 200% to 1000%, 300% to 1000%, 400% to 1000%, 500% to 1000%, 600% to 1000%, or 700% to 1000%, when formulated according to the present invention.

Protein stability may be measured using various metrics, including but not limited to, the extent or rate of protein aggregation, the loss of enzymatic activity, the loss of anti-antigen titer, and/or the extent or rate of protein degradation. One of skill in the art will recognize that certain metrics will be more or less relevant to individual proteins. For example, the stability of an enzyme may be determined by monitoring the loss of enzymatic activity over time, but not by monitoring the loss of an anti-antigen titer. Conversely, the stability of an antibody may be measured by monitoring the loss of anti-antigen titer, but not by enzymatic activity.

Accordingly, in one embodiment, a labile immunoglobulin composition is an immunoglobulin composition that losses from 20% to 100% of its anti-antigen titer when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 6 months. In another embodiment, a labile immunoglobulin composition is an immunoglobulin composition that losses from 30% to 100% of its anti-antigen titer when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 6 months. In another embodiment, a labile immunoglobulin composition is an immunoglobulin composition that losses from 40% to 100% of its anti-antigen titer when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 6 months. In another embodiment, a labile immunoglobulin composition is an immunoglobulin composition that losses from 50% to 100% of its anti-antigen titer when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 6 months.

In one embodiment, the stability of a labile protein composition may be determined by monitoring the extent or rate of protein aggregation within the formulation. Protein aggregation may be determined for example, by size exclusion chromatography (SEC), high performance size exclusion chromatography (HP-SEC), dynamic light scattering, non-denaturing gel electrophoresis and the like. Although the absolute aggregation level at which a labile protein composition will be considered unstable will vary from protein to protein, the level of aggregation that results in a significant loss of the therapeutic value of the composition will generally be regarded as unstable.

In another embodiment, wherein the labile therapeutic protein is an enzyme, the stability of the composition may be determined by monitoring the loss of bulk enzymatic activity in the preparation. For example, in one embodiment, a 20% loss of enzymatic activity will correspond to an unstable composition. In other embodiments, a 10% loss of enzymatic activity, or a 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, or higher loss of enzymatic activity will correspond to an unstable composition.

In one embodiment, a labile protein composition is an enzyme composition that losses from 10% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.5 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 1 month. In another embodiment, a labile protein composition is an enzyme composition that losses from 20% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 1 month. In one embodiment, a labile protein composition is an enzyme composition that losses from 30% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 1 month. In one embodiment, a labile protein composition is an enzyme composition that losses from 40% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 1 month. In one embodiment, a labile protein composition is an enzyme composition that losses from 50% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 1 month.

In one embodiment, a labile protein composition is an enzyme composition that losses from 10% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.5 and stored at a particular temperature (e.g.; from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 2 months. In another embodiment, a labile protein composition is an enzyme composition that losses from 20% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 2 months. In one embodiment, a labile protein composition is an enzyme composition that losses from 30% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 2 months. In one embodiment, a labile protein composition is an enzyme composition that losses from 40% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 2 months. In one embodiment, a labile protein composition is an enzyme composition that losses from 50% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 2 months.

In one embodiment, a labile protein composition is an enzyme composition that losses from 10% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.5 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 3 months. In another embodiment, a labile protein composition is an enzyme composition that losses from 20% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 3 months. In one embodiment, a labile protein composition is an enzyme composition that losses from 30% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 3 months. In one embodiment, a labile protein composition is an enzyme composition that losses from 40% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 3 months. In one embodiment, a labile protein composition is an enzyme composition that losses from 50% to 100% of enzymatic activity when formulated with less than 75 mM (i.e., between 0 mM and 75 mM) of an alkali metal chloride salt at a pH from 5.5 to 7.0 and stored at a particular temperature (e.g., from 2° C. to 8° C., from 20° C. to 25° C., from 28° C. to 32° C., or from 38° C. to 42° C.) for between 1 day and 3 months.

In another embodiment, the stability of a labile protein composition may be determined by monitoring the extent or rate of protein degradation within the formulation. Protein degradation may be determined for example, by size exclusion chromatography (SEC), high performance size exclusion chromatography (HP-SEC), dynamic, light scattering, non-denaturing gel electrophoresis and the like. Although the absolute degradation level at which a labile protein composition will be considered unstable will vary from protein to protein, the level of degradation that results in a significant loss of the therapeutic value of the composition will generally be regarded as unstable.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein, the composition comprising between 75 mM and 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between 5.5 and 7.5. In a specific embodiment, the stabilizing agent is a buffer. In a more specific embodiment, the stabilizing agent is an amino acid. In a yet more specific embodiment, the amino acid is glycine, histidine, or proline. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein, the composition consisting essentially of: between 75 mM and 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between 5.5 and 7.5. In a specific embodiment, the stabilizing agent is a buffer. In a more specific embodiment, the stabilizing agent is an amino acid. In a yet more specific embodiment, the amino acid is glycine, histidine, or proline. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein, the composition consisting of: between 75 mM and 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between 5.5 and 7.5. In a specific embodiment, the stabilizing agent is a buffer. In a more specific embodiment, the stabilizing agent is an amino acid. In a yet more specific embodiment, the amino acid is glycine, histidine, or proline. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a closely related embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein, the composition comprising: between about 75 mM and about 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between about 5.5 and about 7.5. In a specific embodiment, the stabilizing agent is a buffer. In a more specific embodiment, the stabilizing agent is an amino acid. In a yet more specific embodiment, the amino acid is glycine, histidine, or proline. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein, the composition consisting essentially of: between about 75 mM and about 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between about 5.5 and about 7.5. In a specific embodiment, the stabilizing agent is a buffer. In a more specific embodiment, the stabilizing agent is an amino acid. In a yet more specific embodiment, the amino acid is glycine, histidine, or proline. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein, the composition consisting of: between about 75 mM and about 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between about 5.5 and about 7.5. In a specific embodiment, the stabilizing agent is a buffer. In a more specific embodiment, the stabilizing agent is an amino acid. In a yet more specific embodiment, the amino acid is glycine, histidine, or proline. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

1. Alkali Metal Chloride Salts

Surprisingly, it was found that the addition of a moderate level of an alkali metal chloride salt to an aqueous composition of a labile therapeutic protein formulated at a mildly acidic to neutral pH significantly stabilized the labile protein, regardless of the identity of the protein. In one embodiment, a stabilizing formulation provided herein contains between 75 mM and 200 mM of an alkali metal chloride salt. In a preferred embodiment, the formulation contains between 100 mM and 200 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 150 mM and 200 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 75 mM and 175 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 75 mM and 150 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 75 mM and 125 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 100 mM and 175 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 100 mM and 150 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 100 mM and 125 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 125 mM and 200 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 125 mM and 175 mM of an alkali metal chloride salt. In another embodiment, the formulation contains between 125 mM and 150 mM of an alkali metal chloride salt. In yet other embodiments, the formulation contains 70±7 mM, 75±7.5 mM, 80±8 mM, 90±9 mM, 100 mM±10, 110±11 mM, 120 mM±12, 125±12.5 mM, 130±13 mM, 140±14 mM, 150±15 mM, 160±16 mM, 170±17 mM, 175±17.5 mM, 180±18 mM, 190±19 mM, 200±20 mM, 210±21 mM, or 220±22 mM of an alkali metal chloride salt. In yet other more specific embodiments, the formulation contains 70 mM, 75 mM, 80 mM, 85 mM, 90 mM, 95 mM, 100 mM, 105 mM, 110 mM, 115 mM, 120 mM, 125 mM, 130 mM, 135 mM, 140 mM, 145 mM, 150 mM, 155 mM, 160 mM, 165 mM, 170 mM, 175 mM, 180 mM, 185 mM, 190 mM, 195 mM, 200 mM, 205 mM, 210 mM, 215 mM, or 220 mM of an alkali metal chloride salt. In one preferred embodiment, the alkali metal chloride salt is sodium chloride. In another preferred embodiment, the alkali metal chloride salt is potassium chloride.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; 150±15 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; 150±15 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; 150±15 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride.

TABLE 1 Particular combinations of alkali metal chloride salt concentration and pH useful for the aqueous formulation of labile therapeutic proteins. Alkali Metal Chloride Salt (mM) pH 75-250 75-225 75-200 75-175 75-150 75-125 100-250 100-225 5.5-7.5 Var. 1 Var. 73 Var. 145 Var. 217 Var. 289 Var. 361 Var. 433 Var. 505 5.5-7.0 Var. 2 Var. 74 Var. 146 Var. 218 Var. 290 Var. 362 Var. 434 Var. 506 5.5-6.5 Var. 3 Var. 75 Var. 147 Var. 219 Var. 291 Var. 363 Var. 435 Var. 507 5.5-6.0 Var. 4 Var. 76 Var. 148 Var. 220 Var. 292 Var. 364 Var. 436 Var. 508 6.0-7.5 Var. 5 Var. 77 Var. 149 Var. 221 Var. 293 Var. 365 Var. 437 Var. 509 6.0-7.0 Var. 6 Var. 78 Var. 150 Var. 222 Var. 294 Var. 366 Var. 438 Var. 510 6.0-6.5 Var. 7 Var. 79 Var. 151 Var. 223 Var. 295 Var. 367 Var. 439 Var. 511 6.5-7.5 Var. 8 Var. 80 Var. 152 Var. 224 Var. 296 Var. 368 Var. 440 Var. 512 6.5-7.0 Var. 9 Var. 81 Var. 153 Var. 225 Var. 297 Var. 369 Var. 441 Var. 513 5.5 ± 0.2 Var. 10 Var. 82 Var. 154 Var. 226 Var. 298 Var. 370 Var. 442 Var. 514 5.6 ± 0.2 Var. 11 Var. 83 Var. 155 Var. 227 Var. 299 Var. 371 Var. 443 Var. 515 5.7 ± 0.2 Var. 12 Var. 84 Var. 156 Var. 228 Var. 300 Var. 372 Var. 444 Var. 516 5.8 ± 0.2 Var. 13 Var. 85 Var. 157 Var. 229 Var. 301 Var. 373 Var. 445 Var. 517 5.9 ± 0.2 Var. 14 Var. 86 Var. 158 Var. 230 Var. 302 Var. 374 Var. 446 Var. 518 6.0 ± 0.2 Var. 15 Var. 87 Var. 159 Var. 231 Var. 303 Var. 375 Var. 447 Var. 519 6.1 ± 0.2 Var. 16 Var. 88 Var. 160 Var. 232 Var. 304 Var. 376 Var. 448 Var. 520 6.2 ± 0.2 Var. 17 Var. 89 Var. 161 Var. 233 Var. 305 Var. 377 Var. 449 Var. 521 6.3 ± 0.2 Var. 18 Var. 90 Var. 162 Var. 234 Var. 306 Var. 378 Var. 450 Var. 522 6.4 ± 0.2 Var. 19 Var. 91 Var. 163 Var. 235 Var. 307 Var. 379 Var. 451 Var. 523 6.5 ± 0.2 Var. 20 Var. 92 Var. 164 Var. 236 Var. 308 Var. 380 Var. 452 Var. 524 6.6 ± 0.2 Var. 21 Var. 93 Var. 165 Var. 237 Var. 309 Var. 381 Var. 453 Var. 525 6.7 ± 0.2 Var. 22 Var. 94 Var. 166 Var. 238 Var. 310 Var. 382 Var. 454 Var. 526 6.8 ± 0.2 Var. 23 Var. 95 Var. 167 Var. 239 Var. 311 Var. 383 Var. 455 Var. 527 6.9 ± 0.2 Var. 24 Var. 96 Var. 168 Var. 240 Var. 312 Var. 384 Var. 456 Var. 528 7.0 ± 0.2 Var. 25 Var. 97 Var. 169 Var. 241 Var. 313 Var. 385 Var. 457 Var. 529 7.1 ± 0.2 Var. 26 Var. 98 Var. 170 Var. 242 Var. 314 Var. 386 Var. 458 Var. 530 7.2 ± 0.2 Var. 27 Var. 99 Var. 171 Var. 243 Var. 315 Var. 387 Var. 459 Var. 531 7.3 ± 0.2 Var. 28 Var. 100 Var. 172 Var. 244 Var. 316 Var. 388 Var. 460 Var. 532 7.4 ± 0.2 Var. 29 Var. 101 Var. 173 Var. 245 Var. 317 Var. 389 Var. 461 Var. 533 7.5 ± 0.2 Var. 30 Var. 102 Var. 174 Var. 246 Var. 318 Var. 390 Var. 462 Var. 534 5.5 ± 0.1 Var. 31 Var. 103 Var. 175 Var. 247 Var. 319 Var. 391 Var. 463 Var. 535 5.6 ± 0.1 Var. 32 Var. 104 Var. 176 Var. 248 Var. 320 Var. 392 Var. 464 Var. 536 5.7 ± 0.1 Var. 33 Var. 105 Var. 177 Var. 249 Var. 321 Var. 393 Var. 465 Var. 537 5.8 ± 0.1 Var. 34 Var. 106 Var. 178 Var. 250 Var. 322 Var. 394 Var. 466 Var. 538 5.9 ± 0.1 Var. 35 Var. 107 Var. 179 Var. 251 Var. 323 Var. 395 Var. 467 Var. 539 6.0 ± 0.1 Var. 36 Var. 108 Var. 180 Var. 252 Var. 324 Var. 396 Var. 468 Var. 540 6.1 ± 0.1 Var. 37 Var. 109 Var. 181 Var. 253 Var. 325 Var. 397 Var. 469 Var. 541 6.2 ± 0.1 Var. 38 Var. 110 Var. 182 Var. 254 Var. 326 Var. 398 Var. 470 Var. 542 6.3 ± 0.1 Var. 39 Var. 111 Var. 183 Var. 255 Var. 327 Var. 399 Var. 471 Var. 543 6.4 ± 0.1 Var. 40 Var. 112 Var. 184 Var. 256 Var. 328 Var. 400 Var. 472 Var. 544 6.5 ± 0.1 Var. 41 Var. 113 Var. 185 Var. 257 Var. 329 Var. 401 Var. 473 Var. 545 6.6 ± 0.1 Var. 42 Var. 114 Var. 186 Var. 258 Var. 330 Var. 402 Var. 474 Var. 546 6.7 ± 0.1 Var. 43 Var. 115 Var. 187 Var. 259 Var. 331 Var. 403 Var. 475 Var. 547 6.8 ± 0.1 Var. 44 Var. 116 Var. 188 Var. 260 Var. 332 Var. 404 Var. 476 Var. 548 6.9 ± 0.1 Var. 45 Var. 117 Var. 189 Var. 261 Var. 333 Var. 405 Var. 477 Var. 549 7.0 ± 0.1 Var. 46 Var. 118 Var. 190 Var. 262 Var. 334 Var. 406 Var. 478 Var. 550 7.1 ± 0.1 Var. 47 Var. 119 Var. 191 Var. 263 Var. 335 Var. 407 Var. 479 Var. 551 7.2 ± 0.1 Var. 48 Var. 120 Var. 192 Var. 264 Var. 336 Var. 408 Var. 480 Var. 552 7.3 ± 0.1 Var. 49 Var. 121 Var. 193 Var. 265 Var. 337 Var. 409 Var. 481 Var. 553 7.4 ± 0.1 Var. 50 Var. 122 Var. 194 Var. 266 Var. 338 Var. 410 Var. 482 Var. 554 7.5 ± 0.1 Var. 51 Var. 123 Var. 195 Var. 267 Var. 339 Var. 411 Var. 483 Var. 555 5.5 Var. 52 Var. 124 Var. 196 Var. 268 Var. 340 Var. 412 Var. 484 Var. 556 5.6 Var. 53 Var. 125 Var. 197 Var. 269 Var. 341 Var. 413 Var. 485 Var. 557 5.7 Var. 54 Var. 126 Var. 198 Var. 270 Var. 342 Var. 414 Var. 486 Var. 558 5.8 Var. 55 Var. 127 Var. 199 Var. 271 Var. 343 Var. 415 Var. 487 Var. 559 5.9 Var. 56 Var. 128 Var. 200 Var. 272 Var. 344 Var. 416 Var. 488 Var. 560 6   Var. 57 Var. 129 Var. 201 Var. 273 Var. 345 Var. 417 Var. 489 Var. 561 6.1 Var. 58 Var. 130 Var. 202 Var. 274 Var. 346 Var. 418 Var. 490 Var. 562 6.2 Var. 59 Var. 131 Var. 203 Var. 275 Var. 347 Var. 419 Var. 491 Var. 563 6.3 Var. 60 Var. 132 Var. 204 Var. 276 Var. 348 Var. 420 Var. 492 Var. 564 6.4 Var. 61 Var. 133 Var. 205 Var. 277 Var. 349 Var. 421 Var. 493 Var. 565 6.5 Var. 62 Var. 134 Var. 206 Var. 278 Var. 350 Var. 422 Var. 494 Var. 566 6.6 Var. 63 Var. 135 Var. 207 Var. 279 Var. 351 Var. 423 Var. 495 Var. 567 6.7 Var. 64 Var. 136 Var. 208 Var. 280 Var. 352 Var. 424 Var. 496 Var. 568 6.8 Var. 65 Var. 137 Var. 209 Var. 281 Var. 353 Var. 425 Var. 497 Var. 569 6.9 Var. 66 Var. 138 Var. 210 Var. 282 Var. 354 Var. 426 Var. 498 Var. 570 7   Var. 67 Var. 139 Var. 211 Var. 283 Var. 355 Var. 427 Var. 499 Var. 571 7.1 Var. 68 Var. 140 Var. 212 Var. 284 Var. 356 Var. 428 Var. 500 Var. 572 7.2 Var. 69 Var. 141 Var. 213 Var. 285 Var. 357 Var. 429 Var. 501 Var. 573 7.3 Var. 70 Var. 142 Var. 214 Var. 286 Var. 358 Var. 430 Var. 502 Var. 574 7.4 Var. 71 Var. 143 Var. 215 Var. 287 Var. 359 Var. 431 Var. 503 Var. 575 7.5 Var. 72 Var. 144 Var. 216 Var. 288 Var. 360 Var. 432 Var. 504 Var. 576

TABLE 2 Particular combinations of histidine concentration and pH useful for the formulation of immunoglobulins. Alkali Metal Chloride Salt (mM) pH 100-200 100-175 100-150 100-125 125-250 125-225 125-200 125-175 5.5-7.5 Var. 577 Var. 649 Var. 721 Var. 793 Var. 865 Var. 937 Var. 1009 Var. 1081 5.5-7.0 Var. 578 Var. 650 Var. 722 Var. 794 Var. 866 Var. 938 Var. 1010 Var. 1082 5.5-6.5 Var. 579 Var. 651 Var. 723 Var. 795 Var. 867 Var. 939 Var. 1011 Var. 1083 5.5-6.0 Var. 580 Var. 652 Var. 724 Var. 796 Var. 868 Var. 940 Var. 1012 Var. 1084 6.0-7.5 Var. 581 Var. 653 Var. 725 Var. 797 Var. 869 Var. 941 Var. 1013 Var. 1085 6.0-7.0 Var. 582 Var. 654 Var. 726 Var. 798 Var. 870 Var. 942 Var. 1014 Var. 1086 6.0-6.5 Var. 583 Var. 655 Var. 727 Var. 799 Var. 871 Var. 943 Var. 1015 Var. 1087 6.5-7.5 Var. 584 Var. 656 Var. 728 Var. 800 Var. 872 Var. 944 Var. 1016 Var. 1088 6.5-7.0 Var. 585 Var. 657 Var. 729 Var. 801 Var. 873 Var. 945 Var. 1017 Var. 1089 5.5 ± 0.2 Var. 586 Var. 658 Var. 730 Var. 802 Var. 874 Var. 946 Var. 1018 Var. 1090 5.6 ± 0.2 Var. 587 Var. 659 Var. 731 Var. 803 Var. 875 Var. 947 Var. 1019 Var. 1091 5.7 ± 0.2 Var. 588 Var. 660 Var. 732 Var. 804 Var. 876 Var. 948 Var. 1020 Var. 1092 5.8 ± 0.2 Var. 589 Var. 661 Var. 733 Var. 805 Var. 877 Var. 949 Var. 1021 Var. 1093 5.9 ± 0.2 Var. 590 Var. 662 Var. 734 Var. 806 Var. 878 Var. 950 Var. 1022 Var. 1094 6.0 ± 0.2 Var. 591 Var. 663 Var. 735 Var. 807 Var. 879 Var. 951 Var. 1023 Var. 1095 6.1 ± 0.2 Var. 592 Var. 664 Var. 736 Var. 808 Var. 880 Var. 952 Var. 1024 Var. 1096 6.2 ± 0.2 Var. 593 Var. 665 Var. 737 Var. 809 Var. 881 Var. 953 Var. 1025 Var. 1097 6.3 ± 0.2 Var. 594 Var. 666 Var. 738 Var. 810 Var. 882 Var. 954 Var. 1026 Var. 1098 6.4 ± 0.2 Var. 595 Var. 667 Var. 739 Var. 811 Var. 883 Var. 955 Var. 1027 Var. 1099 6.5 ± 0.2 Var. 596 Var. 668 Var. 740 Var. 812 Var. 884 Var. 956 Var. 1028 Var. 1100 6.6 ± 0.2 Var. 597 Var. 669 Var. 741 Var. 813 Var. 885 Var. 957 Var. 1029 Var. 1101 6.7 ± 0.2 Var. 598 Var. 670 Var. 742 Var. 814 Var. 886 Var. 958 Var. 1030 Var. 1102 6.8 ± 0.2 Var. 599 Var. 671 Var. 743 Var. 815 Var. 887 Var. 959 Var. 1031 Var. 1103 6.9 ± 0.2 Var. 600 Var. 672 Var. 744 Var. 816 Var. 888 Var. 960 Var. 1032 Var. 1104 7.0 ± 0.2 Var. 601 Var. 673 Var. 745 Var. 817 Var. 889 Var. 961 Var. 1033 Var. 1105 7.1 ± 0.2 Var. 602 Var. 674 Var. 746 Var. 818 Var. 890 Var. 962 Var. 1034 Var. 1106 7.2 ± 0.2 Var. 603 Var. 675 Var. 747 Var. 819 Var. 891 Var. 963 Var. 1035 Var. 1107 7.3 ± 0.2 Var. 604 Var. 676 Var. 748 Var. 820 Var. 892 Var. 964 Var. 1036 Var. 1108 7.4 ± 0.2 Var. 605 Var. 677 Var. 749 Var. 821 Var. 893 Var. 965 Var. 1037 Var. 1109 7.5 ± 0.2 Var. 606 Var. 678 Var. 750 Var. 822 Var. 894 Var. 966 Var. 1038 Var. 1110 5.5 ± 0.1 Var. 607 Var. 679 Var. 751 Var. 823 Var. 895 Var. 967 Var. 1039 Var. 1111 5.6 ± 0.1 Var. 608 Var. 680 Var. 752 Var. 824 Var. 896 Var. 968 Var. 1040 Var. 1112 5.7 ± 0.1 Var. 609 Var. 681 Var. 753 Var. 825 Var. 897 Var. 969 Var. 1041 Var. 1113 5.8 ± 0.1 Var. 610 Var. 682 Var. 754 Var. 826 Var. 898 Var. 970 Var. 1042 Var. 1114 5.9 ± 0.1 Var. 611 Var. 683 Var. 755 Var. 827 Var. 899 Var. 971 Var. 1043 Var. 1115 6.0 ± 0.1 Var. 612 Var. 684 Var. 756 Var. 828 Var. 900 Var. 972 Var. 1044 Var. 1116 6.1 ± 0.1 Var. 613 Var. 685 Var. 757 Var. 829 Var. 901 Var. 973 Var. 1045 Var. 1117 6.2 ± 0.1 Var. 614 Var. 686 Var. 758 Var. 830 Var. 902 Var. 974 Var. 1046 Var. 1118 6.3 ± 0.1 Var. 615 Var. 687 Var. 759 Var. 831 Var. 903 Var. 975 Var. 1047 Var. 1119 6.4 ± 0.1 Var. 616 Var. 688 Var. 760 Var. 832 Var. 904 Var. 976 Var. 1048 Var. 1120 6.5 ± 0.1 Var. 617 Var. 689 Var. 761 Var. 833 Var. 905 Var. 977 Var. 1049 Var. 1121 6.6 ± 0.1 Var. 618 Var. 690 Var. 762 Var. 834 Var. 906 Var. 978 Var. 1050 Var. 1122 6.7 ± 0.1 Var. 619 Var. 691 Var. 763 Var. 835 Var. 907 Var. 979 Var. 1051 Var. 1123 6.8 ± 0.1 Var. 620 Var. 692 Var. 764 Var. 836 Var. 908 Var. 980 Var. 1052 Var. 1124 6.9 ± 0.1 Var. 621 Var. 693 Var. 765 Var. 837 Var. 909 Var. 981 Var. 1053 Var. 1125 7.0 ± 0.1 Var. 622 Var. 694 Var. 766 Var. 838 Var. 910 Var. 982 Var. 1054 Var. 1126 7.1 ± 0.1 Var. 623 Var. 695 Var. 767 Var. 839 Var. 911 Var. 983 Var. 1055 Var. 1127 7.2 ± 0.1 Var. 624 Var. 696 Var. 768 Var. 840 Var. 912 Var. 984 Var. 1056 Var. 1128 7.3 ± 0.1 Var. 625 Var. 697 Var. 769 Var. 841 Var. 913 Var. 985 Var. 1057 Var. 1129 7.4 ± 0.1 Var. 626 Var. 698 Var. 770 Var. 842 Var. 914 Var. 986 Var. 1058 Var. 1130 7.5 ± 0.1 Var. 627 Var. 699 Var. 771 Var. 843 Var. 915 Var. 987 Var. 1059 Var. 1131 5.5 Var. 628 Var. 700 Var. 772 Var. 844 Var. 916 Var. 988 Var. 1060 Var. 1132 5.6 Var. 629 Var. 701 Var. 773 Var. 845 Var. 917 Var. 989 Var. 1061 Var. 1133 5.7 Var. 630 Var. 702 Var. 774 Var. 846 Var. 918 Var. 990 Var. 1062 Var. 1134 5.8 Var. 631 Var. 703 Var. 775 Var. 847 Var. 919 Var. 991 Var. 1063 Var. 1135 5.9 Var. 632 Var. 704 Var. 776 Var. 848 Var. 920 Var. 992 Var. 1064 Var. 1136 6   Var. 633 Var. 705 Var. 777 Var. 849 Var. 921 Var. 993 Var. 1065 Var. 1137 6.1 Var. 634 Var. 706 Var. 778 Var. 850 Var. 922 Var. 994 Var. 1066 Var. 1138 6.2 Var. 635 Var. 707 Var. 779 Var. 851 Var. 923 Var. 995 Var. 1067 Var. 1139 6.3 Var. 636 Var. 708 Var. 780 Var. 852 Var. 924 Var. 996 Var. 1068 Var. 1140 6.4 Var. 637 Var. 709 Var. 781 Var. 853 Var. 925 Var. 997 Var. 1069 Var. 1141 6.5 Var. 638 Var. 710 Var. 782 Var. 854 Var. 926 Var. 998 Var. 1070 Var. 1142 6.6 Var. 639 Var. 711 Var. 783 Var. 855 Var. 927 Var. 999 Var. 1071 Var. 1143 6.7 Var. 640 Var. 712 Var. 784 Var. 856 Var. 928 Var. 1000 Var. 1072 Var. 1144 6.8 Var. 641 Var. 713 Var. 785 Var. 857 Var. 929 Var. 1001 Var. 1073 Var. 1145 6.9 Var. 642 Var. 714 Var. 786 Var. 858 Var. 930 Var. 1002 Var. 1074 Var. 1146 7   Var. 643 Var. 715 Var. 787 Var. 859 Var. 931 Var. 1003 Var. 1075 Var. 1147 7.1 Var. 644 Var. 716 Var. 788 Var. 860 Var. 932 Var. 1004 Var. 1076 Var. 1148 7.2 Var. 645 Var. 717 Var. 789 Var. 861 Var. 933 Var. 1005 Var. 1077 Var. 1149 7.3 Var. 646 Var. 718 Var. 790 Var. 862 Var. 934 Var. 1006 Var. 1078 Var. 1150 7.4 Var. 647 Var. 719 Var. 791 Var. 863 Var. 935 Var. 1007 Var. 1079 Var. 1151 7.5 Var. 648 Var. 720 Var. 792 Var. 864 Var. 936 Var. 1008 Var. 1080 Var. 1152

TABLE 3 Particular combinations of histidine concentration and pH useful for the formulation of immunoglobulins. Alkali Metal Chloride Salt (mM) pH 125-150 150-250 150-225 150-200 150-175 75 ± 7.5 100 ± 10 125 ± 12.5 5.5-7.5 Var. 1153 Var. 1225 Var. 1297 Var. 1369 Var. 1441 Var. 1513 Var. 1585 Var. 1657 5.5-7.0 Var. 1154 Var. 1226 Var. 1298 Var. 1370 Var. 1442 Var. 1514 Var. 1586 Var. 1658 5.5-6.5 Var. 1155 Var. 1227 Var. 1299 Var. 1371 Var. 1443 Var. 1515 Var. 1587 Var. 1659 5.5-6.0 Var. 1156 Var. 1228 Var. 1300 Var. 1372 Var. 1444 Var. 1516 Var. 1588 Var. 1660 6.0-7.5 Var. 1157 Var. 1229 Var. 1301 Var. 1373 Var. 1445 Var. 1517 Var. 1589 Var. 1661 6.0-7.0 Var. 1158 Var. 1230 Var. 1302 Var. 1374 Var. 1446 Var. 1518 Var. 1590 Var. 1662 6.0-6.5 Var. 1159 Var. 1231 Var. 1303 Var. 1375 Var. 1447 Var. 1519 Var. 1591 Var. 1663 6.5-7.5 Var. 1160 Var. 1232 Var. 1304 Var. 1376 Var. 1448 Var. 1520 Var. 1592 Var. 1664 6.5-7.0 Var. 1161 Var. 1233 Var. 1305 Var. 1377 Var. 1449 Var. 1521 Var. 1593 Var. 1665 5.5 ± 0.2 Var. 1162 Var. 1234 Var. 1306 Var. 1378 Var. 1450 Var. 1522 Var. 1594 Var. 1666 5.6 ± 0.2 Var. 1163 Var. 1235 Var. 1307 Var. 1379 Var. 1451 Var. 1523 Var. 1595 Var. 1667 5.7 ± 0.2 Var. 1164 Var. 1236 Var. 1308 Var. 1380 Var. 1452 Var. 1524 Var. 1596 Var. 1668 5.8 ± 0.2 Var. 1165 Var. 1237 Var. 1309 Var. 1381 Var. 1453 Var. 1525 Var. 1597 Var. 1669 5.9 ± 0.2 Var. 1166 Var. 1238 Var. 1310 Var. 1382 Var. 1454 Var. 1526 Var. 1598 Var. 1670 6.0 ± 0.2 Var. 1167 Var. 1239 Var. 1311 Var. 1383 Var. 1455 Var. 1527 Var. 1599 Var. 1671 6.1 ± 0.2 Var. 1168 Var. 1240 Var. 1312 Var. 1384 Var. 1456 Var. 1528 Var. 1600 Var. 1672 6.2 ± 0.2 Var. 1169 Var. 1241 Var. 1313 Var. 1385 Var. 1457 Var. 1529 Var. 1601 Var. 1673 6.3 ± 0.2 Var. 1170 Var. 1242 Var. 1314 Var. 1386 Var. 1458 Var. 1530 Var. 1602 Var. 1674 6.4 ± 0.2 Var. 1171 Var. 1243 Var. 1315 Var. 1387 Var. 1459 Var. 1531 Var. 1603 Var. 1675 6.5 ± 0.2 Var. 1172 Var. 1244 Var. 1316 Var. 1388 Var. 1460 Var. 1532 Var. 1604 Var. 1676 6.6 ± 0.2 Var. 1173 Var. 1245 Var. 1317 Var. 1389 Var. 1461 Var. 1533 Var. 1605 Var. 1677 6.7 ± 0.2 Var. 1174 Var. 1246 Var. 1318 Var. 1390 Var. 1462 Var. 1534 Var. 1606 Var. 1678 6.8 ± 0.2 Var. 1175 Var. 1247 Var. 1319 Var. 1391 Var. 1463 Var. 1535 Var. 1607 Var. 1679 6.9 ± 0.2 Var. 1176 Var. 1248 Var. 1320 Var. 1392 Var. 1464 Var. 1536 Var. 1608 Var. 1680 7.0 ± 0.2 Var. 1177 Var. 1249 Var. 1321 Var. 1393 Var. 1465 Var. 1537 Var. 1609 Var. 1681 7.1 ± 0.2 Var. 1178 Var. 1250 Var. 1322 Var. 1394 Var. 1466 Var. 1538 Var. 1610 Var. 1682 7.2 ± 0.2 Var. 1179 Var. 1251 Var. 1323 Var. 1395 Var. 1467 Var. 1539 Var. 1611 Var. 1683 7.3 ± 0.2 Var. 1180 Var. 1252 Var. 1324 Var. 1396 Var. 1468 Var. 1540 Var. 1612 Var. 1684 7.4 ± 0.2 Var. 1181 Var. 1253 Var. 1325 Var. 1397 Var. 1469 Var. 1541 Var. 1613 Var. 1685 7.5 ± 0.2 Var. 1182 Var. 1254 Var. 1326 Var. 1398 Var. 1470 Var. 1542 Var. 1614 Var. 1686 5.5 ± 0.1 Var. 1183 Var. 1255 Var. 1327 Var. 1399 Var. 1471 Var. 1543 Var. 1615 Var. 1687 5.6 ± 0.1 Var. 1184 Var. 1256 Var. 1328 Var. 1400 Var. 1472 Var. 1544 Var. 1616 Var. 1688 5.7 ± 0.1 Var. 1185 Var. 1257 Var. 1329 Var. 1401 Var. 1473 Var. 1545 Var. 1617 Var. 1689 5.8 ± 0.1 Var. 1186 Var. 1258 Var. 1330 Var. 1402 Var. 1474 Var. 1546 Var. 1618 Var. 1690 5.9 ± 0.1 Var. 1187 Var. 1259 Var. 1331 Var. 1403 Var. 1475 Var. 1547 Var. 1619 Var. 1691 6.0 ± 0.1 Var. 1188 Var. 1260 Var. 1332 Var. 1404 Var. 1476 Var. 1548 Var. 1620 Var. 1692 6.1 ± 0.1 Var. 1189 Var. 1261 Var. 1333 Var. 1405 Var. 1477 Var. 1549 Var. 1621 Var. 1693 6.2 ± 0.1 Var. 1190 Var. 1262 Var. 1334 Var. 1406 Var. 1478 Var. 1550 Var. 1622 Var. 1694 6.3 ± 0.1 Var. 1191 Var. 1263 Var. 1335 Var. 1407 Var. 1479 Var. 1551 Var. 1623 Var. 1695 6.4 ± 0.1 Var. 1192 Var. 1264 Var. 1336 Var. 1408 Var. 1480 Var. 1552 Var. 1624 Var. 1696 6.5 ± 0.1 Var. 1193 Var. 1265 Var. 1337 Var. 1409 Var. 1481 Var. 1553 Var. 1625 Var. 1697 6.6 ± 0.1 Var. 1194 Var. 1266 Var. 1338 Var. 1410 Var. 1482 Var. 1554 Var. 1626 Var. 1698 6.7 ± 0.1 Var. 1195 Var. 1267 Var. 1339 Var. 1411 Var. 1483 Var. 1555 Var. 1627 Var. 1699 6.8 ± 0.1 Var. 1196 Var. 1268 Var. 1340 Var. 1412 Var. 1484 Var. 1556 Var. 1628 Var. 1700 6.9 ± 0.1 Var. 1197 Var. 1269 Var. 1341 Var. 1413 Var. 1485 Var. 1557 Var. 1629 Var. 1701 7.0 ± 0.1 Var. 1198 Var. 1270 Var. 1342 Var. 1414 Var. 1486 Var. 1558 Var. 1630 Var. 1702 7.1 ± 0.1 Var. 1199 Var. 1271 Var. 1343 Var. 1415 Var. 1487 Var. 1559 Var. 1631 Var. 1703 7.2 ± 0.1 Var. 1200 Var. 1272 Var. 1344 Var. 1416 Var. 1488 Var. 1560 Var. 1632 Var. 1704 7.3 ± 0.1 Var. 1201 Var. 1273 Var. 1345 Var. 1417 Var. 1489 Var. 1561 Var. 1633 Var. 1705 7.4 ± 0.1 Var. 1202 Var. 1274 Var. 1346 Var. 1418 Var. 1490 Var. 1562 Var. 1634 Var. 1706 7.5 ± 0.1 Var. 1203 Var. 1275 Var. 1347 Var. 1419 Var. 1491 Var. 1563 Var. 1635 Var. 1707 5.5 Var. 1204 Var. 1276 Var. 1348 Var. 1420 Var. 1492 Var. 1564 Var. 1636 Var. 1708 5.6 Var. 1205 Var. 1277 Var. 1349 Var. 1421 Var. 1493 Var. 1565 Var. 1637 Var. 1709 5.7 Var. 1206 Var. 1278 Var. 1350 Var. 1422 Var. 1494 Var. 1566 Var. 1638 Var. 1710 5.8 Var. 1207 Var. 1279 Var. 1351 Var. 1423 Var. 1495 Var. 1567 Var. 1639 Var. 1711 5.9 Var. 1208 Var. 1280 Var. 1352 Var. 1424 Var. 1496 Var. 1568 Var. 1640 Var. 1712 6   Var. 1209 Var. 1281 Var. 1353 Var. 1425 Var. 1497 Var. 1569 Var. 1641 Var. 1713 6.1 Var. 1210 Var. 1282 Var. 1354 Var. 1426 Var. 1498 Var. 1570 Var. 1642 Var. 1714 6.2 Var. 1211 Var. 1283 Var. 1355 Var. 1427 Var. 1499 Var. 1571 Var. 1643 Var. 1715 6.3 Var. 1212 Var. 1284 Var. 1356 Var. 1428 Var. 1500 Var. 1572 Var. 1644 Var. 1716 6.4 Var. 1213 Var. 1285 Var. 1357 Var. 1429 Var. 1501 Var. 1573 Var. 1645 Var. 1717 6.5 Var. 1214 Var. 1286 Var. 1358 Var. 1430 Var. 1502 Var. 1574 Var. 1646 Var. 1718 6.6 Var. 1215 Var. 1287 Var. 1359 Var. 1431 Var. 1503 Var. 1575 Var. 1647 Var. 1719 6.7 Var. 1216 Var. 1288 Var. 1360 Var. 1432 Var. 1504 Var. 1576 Var. 1648 Var. 1720 6.8 Var. 1217 Var. 1289 Var. 1361 Var. 1433 Var. 1505 Var. 1577 Var. 1649 Var. 1721 6.9 Var. 1218 Var. 1290 Var. 1362 Var. 1434 Var. 1506 Var. 1578 Var. 1650 Var. 1722 7   Var. 1219 Var. 1291 Var. 1363 Var. 1435 Var. 1507 Var. 1579 Var. 1651 Var. 1723 7.1 Var. 1220 Var. 1292 Var. 1364 Var. 1436 Var. 1508 Var. 1580 Var. 1652 Var. 1724 7.2 Var. 1221 Var. 1293 Var. 1365 Var. 1437 Var. 1509 Var. 1581 Var. 1653 Var. 1725 7.3 Var. 1222 Var. 1294 Var. 1366 Var. 1438 Var. 1510 Var. 1582 Var. 1654 Var. 1726 7.4 Var. 1223 Var. 1295 Var. 1367 Var. 1439 Var. 1511 Var. 1583 Var. 1655 Var. 1727 7.5 Var. 1224 Var. 1296 Var. 1368 Var. 1440 Var. 1512 Var. 1584 Var. 1656 Var. 1728

TABLE 4 Particular combinations of histidine concentration and pH useful for the formulation of immunoglobulins. Alkali Metal Chloride Salt (mM) 175 ± pH 150 ± 15 17.5 200 ± 20 225 ± 22.5 250 ± 25 5.5-7.5 Var. 1729 Var. 1801 Var. 1873 Var. 1945 Var. 2017 5.5-7.0 Var. 1730 Var. 1802 Var. 1874 Var. 1946 Var. 2018 5.5-6.5 Var. 1731 Var. 1803 Var. 1875 Var. 1947 Var. 2019 5.5-6.0 Var. 1732 Var. 1804 Var. 1876 Var. 1948 Var. 2020 6.0-7.5 Var. 1733 Var. 1805 Var. 1877 Var. 1949 Var. 2021 6.0-7.0 Var. 1734 Var. 1806 Var. 1878 Var. 1950 Var. 2022 6.0-6.5 Var. 1735 Var. 1807 Var. 1879 Var. 1951 Var. 2023 6.5-7.5 Var. 1736 Var. 1808 Var. 1880 Var. 1952 Var. 2024 6.5-7.0 Var. 1737 Var. 1809 Var. 1881 Var. 1953 Var. 2025 5.5 ± 0.2 Var. 1738 Var. 1810 Var. 1882 Var. 1954 Var. 2026 5.6 ± 0.2 Var. 1739 Var. 1811 Var. 1883 Var. 1955 Var. 2027 5.7 ± 0.2 Var. 1740 Var. 1812 Var. 1884 Var. 1956 Var. 2028 5.8 ± 0.2 Var. 1741 Var. 1813 Var. 1885 Var. 1957 Var. 2029 5.9 ± 0.2 Var. 1742 Var. 1814 Var. 1886 Var. 1958 Var. 2030 6.0 ± 0.2 Var. 1743 Var. 1815 Var. 1887 Var. 1959 Var. 2031 6.1 ± 0.2 Var. 1744 Var. 1816 Var. 1888 Var. 1960 Var. 2032 6.2 ± 0.2 Var. 1745 Var. 1817 Var. 1889 Var. 1961 Var. 2033 6.3 ± 0.2 Var. 1746 Var. 1818 Var. 1890 Var. 1962 Var. 2034 6.4 ± 0.2 Var. 1747 Var. 1819 Var. 1891 Var. 1963 Var. 2035 6.5 ± 0.2 Var. 1748 Var. 1820 Var. 1892 Var. 1964 Var. 2036 6.6 ± 0.2 Var. 1749 Var. 1821 Var. 1893 Var. 1965 Var. 2037 6.7 ± 0.2 Var. 1750 Var. 1822 Var. 1894 Var. 1966 Var. 2038 6.8 ± 0.2 Var. 1751 Var. 1823 Var. 1895 Var. 1967 Var. 2039 6.9 ± 0.2 Var. 1752 Var. 1824 Var. 1896 Var. 1968 Var. 2040 7.0 ± 0.2 Var. 1753 Var. 1825 Var. 1897 Var. 1969 Var. 2041 7.1 ± 0.2 Var. 1754 Var. 1826 Var. 1898 Var. 1970 Var. 2042 7.2 ± 0.2 Var. 1755 Var. 1827 Var. 1899 Var. 1971 Var. 2043 7.3 ± 0.2 Var. 1756 Var. 1828 Var. 1900 Var. 1972 Var. 2044 7.4 ± 0.2 Var. 1757 Var. 1829 Var. 1901 Var. 1973 Var. 2045 7.5 ± 0.2 Var. 1758 Var. 1830 Var. 1902 Var. 1974 Var. 2046 5.5 ± 0.1 Var. 1759 Var. 1831 Var. 1903 Var. 1975 Var. 2047 5.6 ± 0.1 Var. 1760 Var. 1832 Var. 1904 Var. 1976 Var. 2048 5.7 ± 0.1 Var. 1761 Var. 1833 Var. 1905 Var. 1977 Var. 2049 5.8 ± 0.1 Var. 1762 Var. 1834 Var. 1906 Var. 1978 Var. 2050 5.9 ± 0.1 Var. 1763 Var. 1835 Var. 1907 Var. 1979 Var. 2051 6.0 ± 0.1 Var. 1764 Var. 1836 Var. 1908 Var. 1980 Var. 2052 6.1 ± 0.1 Var. 1765 Var. 1837 Var. 1909 Var. 1981 Var. 2053 6.2 ± 0.1 Var. 1766 Var. 1838 Var. 1910 Var. 1982 Var. 2054 6.3 ± 0.1 Var. 1767 Var. 1839 Var. 1911 Var. 1983 Var. 2055 6.4 ± 0.1 Var. 1768 Var. 1840 Var. 1912 Var. 1984 Var. 2056 6.5 ± 0.1 Var. 1769 Var. 1841 Var. 1913 Var. 1985 Var. 2057 6.6 ± 0.1 Var. 1770 Var. 1842 Var. 1914 Var. 1986 Var. 2058 6.7 ± 0.1 Var. 1771 Var. 1843 Var. 1915 Var. 1987 Var. 2059 6.8 ± 0.1 Var. 1772 Var. 1844 Var. 1916 Var. 1988 Var. 2060 6.9 ± 0.1 Var. 1773 Var. 1845 Var. 1917 Var. 1989 Var. 2061 7.0 ± 0.1 Var. 1774 Var. 1846 Var. 1918 Var. 1990 Var. 2062 7.1 ± 0.1 Var. 1775 Var. 1847 Var. 1919 Var. 1991 Var. 2063 7.2 ± 0.1 Var. 1776 Var. 1848 Var. 1920 Var. 1992 Var. 2064 7.3 ± 0.1 Var. 1777 Var. 1849 Var. 1921 Var. 1993 Var. 2065 7.4 ± 0.1 Var. 1778 Var. 1850 Var. 1922 Var. 1994 Var. 2066 7.5 ± 0.1 Var. 1779 Var. 1851 Var. 1923 Var. 1995 Var. 2067 5.5 Var. 1780 Var. 1852 Var. 1924 Var. 1996 Var. 2068 5.6 Var. 1781 Var. 1853 Var. 1925 Var. 1997 Var. 2069 5.7 Var. 1782 Var. 1854 Var. 1926 Var. 1998 Var. 2070 5.8 Var. 1783 Var. 1855 Var. 1927 Var. 1999 Var. 2071 5.9 Var. 1784 Var. 1856 Var. 1928 Var. 2000 Var. 2072 6   Var. 1785 Var. 1857 Var. 1929 Var. 2001 Var. 2073 6.1 Var. 1786 Var. 1858 Var. 1930 Var. 2002 Var. 2074 6.2 Var. 1787 Var. 1859 Var. 1931 Var. 2003 Var. 2075 6.3 Var. 1788 Var. 1860 Var. 1932 Var. 2004 Var. 2076 6.4 Var. 1789 Var. 1861 Var. 1933 Var. 2005 Var. 2077 6.5 Var. 1790 Var. 1862 Var. 1934 Var. 2006 Var. 2078 6.6 Var. 1791 Var. 1863 Var. 1935 Var. 2007 Var. 2079 6.7 Var. 1792 Var. 1864 Var. 1936 Var. 2008 Var. 2080 6.8 Var. 1793 Var. 1865 Var. 1937 Var. 2009 Var. 2081 6.9 Var. 1794 Var. 1866 Var. 1938 Var. 2010 Var. 2082 7   Var. 1795 Var. 1867 Var. 1939 Var. 2011 Var. 2083 7.1 Var. 1796 Var. 1868 Var. 1940 Var. 2012 Var. 2084 7.2 Var. 1797 Var. 1869 Var. 1941 Var. 2013 Var. 2085 7.3 Var. 1798 Var. 1870 Var. 1942 Var. 2014 Var. 2086 7.4 Var. 1799 Var. 1871 Var. 1943 Var. 2015 Var. 2087 7.5 Var. 1800 Var. 1872 Var. 1944 Var. 2016 Var. 2088

2. Stabilizing Agents

The pharmaceutical compositions provided herein will typically comprise one or more buffering agents or pH stabilizing agents suitable for intravenous, intravitreal, subcutaneous, and/or intramuscular administration. Non-limiting examples of buffering agents suitable for formulating the storage stable compositions provided herein include glycine, histidine, proline, or other amino acids, salts like citrate, phosphate, acetate, glutamate, tartrate, benzoate, lactate, gluconate, malate, succinate, formate, propionate, carbonate, or any combination thereof adjusted to an appropriate pH. Generally, the buffering agent will be sufficient to maintain a suitable pH in the formulation for an extended period of time.

In a preferred embodiment, the stabilizing agent employed in the storage stable, labile therapeutic protein formulations provided herein is an amino acid. Non-limiting examples of amino acids include, isoleucine, alanine, leucine, asparagine, lysine, aspartic acid, methionine, cysteine, phenylalanine, glutamic acid, threonine, glutamine, tryptophan, glycine, valine, proline, selenocysteine, serine, tyrosine, arginine, histidine, ornithine, taurine, combinations thereof, and the like. In one embodiment, the stabilizing amino acids include arginine, histidine, lysine, serine, proline, glycine, alanine, threonine, and a combination thereof. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In yet another preferred embodiment, the amino acid is histidine.

For purposes of further stabilizing the compositions provided herein, the amino acid will typically be added to the formulation at a concentration between 5 mM and 0.75 M. In one embodiment, at least 100 mM of the amino acid is added to the formulation. In another embodiment, at least 200 mM of the amino acid is added to the formulation. In yet another embodiment, at least 250 mM of the amino acid is added to the formulation. In yet other embodiments, the formulations provided herein will contain at least 25 mM, 50 mM, 75 mM, 100 mM, 150 mM, 200 mM, 250 mM, 300 mM, 350 mM, 400 mM, 450 mM, 500 mM, 550 mM, 600 mM, 650 mM, 700 mM, 750 mM, or more of the amino acid.

In one embodiment, the concentration of buffering agent in the formulation will be at or about between 5 mM and 500 mM. In certain embodiments, the concentration of the buffering agent in the formulation will be at or about 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 50 mM, 75 mM, 100 mM, 125 mM, 150 mM, 175 mM, 200 mM, 225 mM, 250 mM, 275 mM, 300 mM, 325 mM, 350 mM, 375 mM, 400 mM, 425 mM, 450 mM, 475 mM, 500 mM or higher.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

3. Excipients

In certain embodiments, the storage stable labile therapeutic protein aqueous compositions provided herein further comprise one or more excipients. Non-limiting examples of excipients that can be included in the formulations provided herein include non-ionic surfactants, bulking agents (e.g., sugars and sugar alcohols), antioxidants, polysaccharides, and pharmaceutically acceptable water-soluble polymers (e.g., poly(acrylic acid), poly(ethylene oxide), poly(ethylene glycol), poly(vinyl pyrrolidone), hydroxyethyl cellulose, hydroxypropyl cellulose, and starch).

In one embodiment, the excipient is an agent for adjusting the osmolarity of the composition. Non-limiting examples of osmolarity agents include mannitol, sorbitol, glycerol, sucrose, glucose, dextrose, levulose, fructose, lactose, trehalose, polyethylene glycols, phosphates, calcium chloride, calcium gluconoglucoheptonate, dimethyl sulfone, and the like.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant, and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment; the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol, and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In another embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: a labile therapeutic protein; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In one embodiment, the pH of the formulation is between 5.5 and 7.0. In a specific embodiment, the pH of the formulation is between 5.5 and 6.5. In yet another embodiment, the pH of the formulation is between 6.0 and 7.0.

4. Administration

Formulations of the storage stable composition provided herein are delivered to the individual by any pharmaceutically suitable means of administration. Various delivery systems are known and can be used to administer the composition by any convenient route. In one embodiment the compositions of the invention are administered systemically. For systemic use, the composition is formulated for parenteral (e.g. intradermal, subcutaneous, transdermal implant, intracavernous, intravitreal, transscleral, intracerebral, intrathecal, epidural, intravenous, intracardiac, intramuscular, intraosseous, intraperitoneal, and nanocell injection) or enteral (e.g., oral, vaginal or rectal) delivery according to conventional methods. The formulations can be administered continuously by infusion or by bolus injection. Some formulations encompass slow release systems. Preferred routes of administration will depend upon the indication being treated, managed, or prevented.

Single or multiple administrations of the compositions 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, 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 therapeutic protein, and the discretion of the attending physician.

In a preferred embodiment, the storage stable compositions provided herein are formulated for parenteral administration including, but not limited to, intradermal, subcutaneous, transdermal implant, intracavernous, intravitreal, transscleral, intracerebral, intrathecal, epidural, intravenous, intracardiac, intramuscular, intraosseous, intraperitoneal, and nanocell injection administration. In one preferred embodiment, the compositions provided herein will be formulated for intravenous administration. In another preferred embodiment, the compositions provided herein will be formulated for subcutaneous administration. In yet another preferred embodiment, the compositions provided herein will be formulated for intramuscular administration.

B. Immunoglobulins

In one aspect, the present invention provides storage stable, aqueous immunoglobulin compositions formulated at mildly acidic to neutral pH with a moderate concentration of a metal chloride salt and a stabilizing agent.

Any immunoglobulin may be stabilized by the formulations provided herein. Non-limiting examples of immunoglobulin preparations that may be stabilized include, plasma-derived immunoglobulin preparations, recombinant polyclonal or monoclonal preparations, minibodies, diabodies, triabodies, antibody fragments such as Fv, Fab and F(ab)2 or fragmented antibodies such as monovalent or multivalent single chain Fvs (scFv, sc(Fv)2, minibodies, diabodies, and triabodies such as scFv dimers) in which the variable regions of an antibody are joined together via a linker such as a peptide linker, and the like. Recombinant antibodies include murine antibodies, rodent antibodies, human antibodies, chimeric human antibodies (e.g., human/murine chimeras), humanized antibodies (e.g., humanized murine antibodies), and the like. In preferred embodiments, the recombinant antibody is a human, chimeric human, or humanized antibody suitable for administration to a human. In a preferred embodiment, the immunoglobulin in a full length, or near full length immunoglobulin, which will generally be more labile then engineered fragments thereof.

Generally, storage stable immunoglobulin formulations provided herein will be stabilized at room temperature (i.e., between 20° C. and 25° C.) for an extended period of time. For example, in one embodiment, a storage stable, aqueous immunoglobulin composition will be stable when stored at room temperature for at least about 2 months. In another embodiment, the composition will be stable for at least about 3 months. In yet other embodiment, the composition will be stable for at least 1 about month, or at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or more months. In a preferred embodiment, the composition will be stable for at least about 6 months. In a more preferred embodiment, the composition will be stable for at least about 1 year. In a more preferred embodiment, the composition will be stable for at least about 2 years.

In one embodiment, the storage stable, aqueous immunoglobulin composition will be stable for at least six months at a temperature between about 28° C. and about 32° C. In a specific embodiment, the storage stable, aqueous immunoglobulin composition will be stable for at least one year at a temperature between about 28° C. and about 32° C. In a more specific embodiment, the storage stable, aqueous immunoglobulin composition will be stable for at least two years at a temperature between about 28° C. and about 32° C. In another embodiment, the storage stable, aqueous immunoglobulin composition will be stable for at least one month at a temperature between about 38° C. and about 42° C. In a specific embodiment, the storage stable, aqueous immunoglobulin composition will be stable for at least three months at a temperature between about 38° C. and about 42° C. In a more specific embodiment, the storage stable, aqueous immunoglobulin composition will be stable for at least one year at a temperature between about 38° C. and about 42° C.

In one embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 10%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 9%. In a more preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 8%. In a more preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 7%. In a more preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 6%. In a more preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 5%. In a more preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 4%. In a more preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 3%. In a most preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 2%.

In one embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 10% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 9% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 8% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 7% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 6% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 5% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 4% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 3% and the percentage of immunoglobulin in the monomeric state is no less than 85%. In a most preferred embodiment, the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state no more than 2% and the percentage of immunoglobulin in the monomeric state is no less than 85%.

In one embodiment, wherein the labile therapeutic protein is an antibody or fragment thereof, the stability of the composition may be determined by monitoring the loss of anti-antigen titer. The level of anti-antigen titer may be determined, for example, by an immunoassay. A variety of immunoassay formats may be used for this purpose. For example, solid-phase ELISA immunoassays are routinely used to determine antigen titer (see, e.g., Harlow & Lane, Using Antibodies, A Laboratory Manual (1998) for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity). In one embodiment, a 20% loss of anti-antigen titer will correspond to an unstable composition. In other embodiments, a 10% loss of anti-antigen titer, or a 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, or higher loss of anti-antigen titer will correspond to an unstable composition.

After formulation, the storage stable, aqueous immunoglobulin compositions provided herein are typically sterile filtered and dispensed into a sterile containment vessel, which is sealed air-tight, for example, using a rubber stopper. Immunoglobulin compositions in the air-tight vessels are preferably protected from ambient light by storage in a dark place, the use of a tinted vessel material (typically glass or plastic), and/or covering the surface of the vessel with an opaque substance.

In certain embodiments, the headspace air in the containment vessel is replaced with an inert gas. The inert gas helps to maintain an inert atmosphere above the liquid composition. In one embodiment, the liquid is overlaid with inert gas. In another embodiment the liquid is degassed before overlaying it with inert gas, meaning that residual oxygen in the atmosphere may vary. In the context of the present invention, when an immunoglobulin composition is stored in a vessel in which the headspace air has been replaced with an inert gas, the composition has been overlaid with inert gas, or the composition is degassed prior to overlaying with inert gas, the composition is said to be “stored under inert gas.” Non-limiting examples of inert gasses than may be used in conjunction with the present invention include, nitrogen, argon, carbon dioxide, helium, krypton, and xenon. In one particular embodiment, the inert gas is nitrogen. In another particular embodiment, the inert gas is argon.

1. Plasma-Derived Immunoglobulins

Preparations of concentrated immunoglobulins (especially IgG) isolated from pooled human plasma are used for treating a variety of medical conditions, including immune deficiencies, inflammatory and autoimmune diseases, and acute infections. One IgG product, intravenous immunoglobulin or IVIG, is formulated for intravenous administration, for example, at a concentration of at or about 10% IgG. Concentrated immunoglobulins may also be formulated for subcutaneous or intramuscular administration, for example, at a concentration at or about 20% IgG.

Generally, plasma-derived immunoglobulin preparations formulated according to the present invention can be prepared from any suitable starting materials, for example, recovered plasma or source plasma. In a typical example, blood or plasma is collected from healthy donors. Immunoglobulins are isolated from the blood or plasma by suitable procedures, such as, for example, precipitation (alcohol fractionation or polyethylene glycol fractionation), chromatographic methods (ion exchange chromatography, affinity chromatography, immunoaffinity chromatography, etc.) ultracentrifugation, and electrophoretic preparation, and the like. (See, e.g., Cohn et al., J. Am. Chem. Soc. 68:459-75 (1946); Oncley et al., J. Am. Chem. Soc. 71:541-50 (1949); Barandun et al., Vox Sang. 7:157-74 (1962); Koblet et al., Vox Sang. 13:93-102 (1967); U.S. Pat. Nos. 5,122,373 and 5,177,194; PCT/US10/36470; and WO 2010/138736 the disclosures of which are hereby incorporated by reference in their entireties for all purposes).

In many cases, immunoglobulins are prepared from gamma globulin-containing compositions produced by alcohol fractionation and/or ion exchange and affinity chromatography methods well known to those skilled in the art. For example, purified Cohn Fraction II is commonly used as a starting point for the further purification of immunoglobulins. The starting Cohn Fraction II paste is typically about 95 percent IgG and is comprised of the four IgG subtypes. The different subtypes are present in Fraction II in approximately the same ratio as they are found in the pooled human plasma from which they are obtained. The Fraction II is further purified before formulation into an administrable product. For example, the Fraction II paste can be dissolved in a cold purified aqueous alcohol solution and impurities removed via precipitation and filtration. Following the final filtration, the immunoglobulin suspension can be dialyzed or diafiltered (e.g., using ultrafiltration membranes having a nominal molecular weight limit of less than or equal to 100,000 daltons) to remove the alcohol. The solution can be concentrated or diluted to obtain the desired protein concentration and can be further purified by techniques well known to those skilled in the art.

Furthermore, additional preparative steps can be used to enrich a particular isotype or subtype of immunoglobulin. For example, protein A, protein G or protein H sepharose chromatography can be used to enrich a mixture of immunoglobulins for IgG, or for specific IgG subtypes. See generally, Harlow and Lane, Using Antibodies, Cold Spring Harbor Laboratory Press (1999); Harlow and Lane, Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory Press (1988); and U.S. Pat. No. 5,180,810, the disclosures of which are hereby incorporated by reference in their entireties for all purposes.

As will be recognized by one of skill in the art, immunoglobulin compositions isolated from pooled plasma contain impurities carried over from the starting plasma. Typically, pharmaceutically acceptable plasma-derived immunoglobulin compositions will contain at least 90% immunoglobulins, preferably at least 95% immunoglobulins, more preferably at least 98% immunoglobulins; most preferably at least 99% immunoglobulins, expressed as a function of total protein content. For example, GAMMAGARD® LIQUID (Baxter International; Deerfield, Ill.) is a plasma-derived immunoglobulin composition formulated at 100 g/L protein. According to the specifications, at least 98% of the protein is immune globulin, the average immunoglobulin A (IgA) concentration is 37 μg/mL, and immunoglobulin M is present in trace amounts (GAMMAGARD® LIQUID Prescribing Information). Accordingly, unless otherwise specified, an immunoglobulin composition provided herein comprising; consisting essentially of; or consisting of “a plasma-derived immunoglobulin” may contain up to 10% plasma protein impurities carried through during the manufacturing process.

In a particular embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 90% IgG immunoglobulins. In a specific embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 95% IgG immunoglobulins. In a more specific embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 98% IgG immunoglobulins. In a yet more specific embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 99% IgG immunoglobulins. In certain embodiments, the IgG immunoglobulin composition isolated from pooled plasma further comprises IgA and/or IgM immunoglobulins.

In another embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 10% IgA. In a specific embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 25% IgA. In a more specific embodiment, immunoglobulin composition isolated from pooled plasma comprises at least 50% IgA. In yet other embodiments, the immunoglobulin composition isolated from pooled plasma comprises at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more IgA. In certain embodiments, the IgA immunoglobulin composition isolated from pooled plasma further comprises IgG and/or IgM immunoglobulins.

In another embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 10% IgM. In a specific embodiment, the immunoglobulin composition isolated from pooled plasma comprises at least 25% IgM. In a more specific embodiment, immunoglobulin composition isolated from pooled plasma comprises at least 50% IgM. In yet other embodiments, the immunoglobulin composition isolated from pooled plasma comprises at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more IgM. In certain embodiments, the IgM immunoglobulin composition isolated from pooled plasma further comprises IgG and/or IgA immunoglobulins.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In one embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a plasma-derived immunoglobulin; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a plasma-derived immunoglobulin; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a plasma-derived immunoglobulin; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the plasma-derived immunoglobulin comprises IgG.

a. Hyper-Immune Immunoglobulins

In a specific embodiment, the storage stable, plasma derived immunoglobulin composition is a hyper-immune immunoglobulin preparation. For example, in certain embodiments, the hyper-immune preparation may be an anti-tetanus, anti-D, anti-varicella, anti-rabies, anti-CMV, anti-hepatitis A, or anti-hepatitis B immunoglobulin preparation.

As demonstrated in Example 3, plasma derived anti-tetanus and anti-D preparations are stabilized by the addition of between about 100 mM and about 200 mM of an alkali metal chloride salt (e.g., sodium chloride) at a pH between about 5.5 and about 6.5. As shown in FIGS. 2 and 3, maximum stability for the hyper-immune immunoglobulin formulations is found between pH 5.5 and 6.0.

Accordingly, in one embodiment, the present invention provides a storage stable, plasma derived hyper-immune immunoglobulin aqueous composition comprising between about 75 mM and about 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between about 5.5 and about 6.5. In a preferred embodiment, the composition comprises between about 100 mM and about 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between about 5.5 and about 6.5. In another preferred embodiment, the composition comprises between about 100 mM and about 200 mM of an alkali metal chloride salt, a stabilizing agent, and a pH between about 5.5 and about 6.0. In a preferred embodiment, the salt is sodium chloride. In another preferred embodiment, the salt is potassium chloride.

In one embodiment, the storage stable, plasma derived hyper-immune immunoglobulin aqueous compositions provided herein have a protein concentration of between about 30 g/L and about 250 g/L. In certain embodiments, the protein concentration of the hyper-immune immunoglobulin composition is between about 50 g/L and about 200 g/L, or between about 70 g/L and about 150 g/L, or between about 90 g/L and about 120 g/L, or any suitable concentration within these ranges, for example about 30 g/L, or about 35 g/L, 40 g/L, 45 g/L, 50 g/L, 55 g/L, 60 g/L, 65 g/L, 70 g/L, 75 g/L, 80 g/L, 85 g/L, 90 g/L, 95 g/L, 100 g/L, 105 g/L, 110 g/L, 115 g/L, 120 g/L, 125 g/L, 130 g/L, 135 g/L, 140 g/L, 145 g/L, 150 g/L, 155 g/L, 160 g/L, 165 g/L, 170 g/L, 175 g/L, 180 g/L, 185 g/L, 190 g/L, 195 g/L, 200 g/L, 205 g/L, 210 g/L, 215 g/L, 220 g/L, 225 g/L, 230 g/L, 235 g/L, 240 g/L, 245 g/L, 250 g/L, or higher. In a preferred embodiment, the aqueous hyper-immune immunoglobulin composition will have a concentration of between about 100 g/L and about 170 g/L.

The storage stable plasma derived hyper-immune immunoglobulin aqueous compositions provided herein will be stabilized at room temperature for an extended period of time. For example, in one embodiment, the storage stable, aqueous hyper-immune immunoglobulin composition will be stable for at least about 2 months. In another embodiment, the composition will be stable for at least about 3 months. In yet other embodiment, the composition will be stable for at least 1 about month, or at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or more months. In a preferred embodiment, the composition will be stable for at least about 6 months. In a more preferred embodiment, the composition will be stable for at least about 1 year. In a more preferred embodiment, the composition will be stable for at least about 2 years.

2. Recombinant Immunoglobulins

In one aspect, the present invention provides storage stable, recombinant immunoglobulin preparations. Methods for obtaining recombinant antibodies, such as recombinant human antibodies are well known in the art. For example, a desired human antibody having a binding activity for a desired antigen can be obtained by in vitro immunizing human lymphocytes with the desired antigen or a cell expressing the desired antigen and fusing the immunized lymphocytes to human myeloma cells. A desired human antibody can also be obtained by immunizing a transgenic animal having all human antibody gene repertoires with an antigen (see, International Publications Nos. WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, WO 96/33735). Methods for obtaining a human antibody by panning using a human antibody library are also known. For example, phages binding to an antigen can be selected by expressing the variable regions of a human antibody as single chain antibody fragments (scFv) on phage surfaces by a phage display method. The DNA sequences encoding the variable regions of the human antibody binding to the antigen can be determined by analyzing the genes of the selected phages. A whole human antibody can be obtained by preparing a suitable expression vector containing the determined DNA sequences of the scFv fragments binding to the antigen. These methods have already been well known from WO 92/01047, WO 92/20791, WO 93/06213, WO 93/11236, WO 93/19172, WO 95/01438, and WO 95/15388.

Methods for the expression of recombinant immunoglobulins are also well known in the art. For example, recombinant antibodies can be expressed in tissue or cell culture after transforming a recombinant gene for the construct into a suitable host. Suitable eukaryotic cells for use as hosts include animal, plant and fungal cells. Known animal cells include (1) mammalian cells such as CHO, COS, myeloma, BHK (baby hamster kidney), HeLa and Vero cells; (2) amphibian cells such as Xenopus oocytes; or (3) insect sells such as sf9, sf21 and Tn5. Known plant cells include cells of Nicotiana such as Nicotiana tabacum, which can be used as callus cultures. Known fungi include yeasts such as Saccharomyces spp., e.g. Saccharomyces serevisiae and filamentous fungi such as Aspergillus spp., e.g. Aspergillus niger. Prokaryotic cells can be used as producing systems using bacterial cells. Known bacterial cells include E. coli and Bacillus subtilis. Antibodies can be obtained by transforming these cells with an antibody gene of interest and culturing the transformed cells in vitro.

In one embodiment of the present invention, the media used to express a recombinant protein can be animal protein-free and chemically defined. Methods of preparing animal protein-free and chemically defined culture mediums are known in the art, for example in US 2008/0009040 and US 2007/0212770, which are both incorporated herein for all purposes. “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.

Typically a recombinant antibody formulated as provided herein is specific for a polypeptide associated with a disease or disorder. Non-limiting examples of such polypeptides include macrophage migration inhibitory factor (MIF), tissue factor pathway inhibitor (TFPI); alpha-1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as Factor II (prothrombin), Factor III (platelet tissue factor), Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor XI, Factor XII, Factor XIII, and von Willebrand factor; anti-clotting factors such as Antithrombin III (ATIII), Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -beta; enkephalinase; RANTES (regulated on activation normally T-cell expressed and secreted); human macrophage inflammatory protein (MIP-1-alpha); a serum albumin such as human serum albumin; Muellerian-inhibiting substance; relaxin A-chain; relaxin B-chain; prorelaxin; mouse gonadotropin-associated peptide; a microbial protein, such as beta-lactamase; DNase; IgE; a cytotoxic T-lymphocyte associated antigen (CTLA), such as CTLA-4; inhibin; activin; vascular endothelial growth factor (VEGF); receptors for hormones or growth factors; protein A or D; rheumatoid factors; a neurotrophic factor such as bone-derived neurotrophic factor (BDNF), neurotrophin-3, -4, -5, or -6 (NT-3, NT-4, NT-5, or NT-6), or a nerve growth factor such as NGF-b; platelet-derived growth factor (PDGF); fibroblast growth factor such as aFGF and bFGF; epidermal growth factor (EGF); transforming growth factor (TGF) such as TGF-alpha and TGF-beta, including TGF-b1, TGF-b2, TGF-b3, TGF-b4, or TGF-b5; a tumor necrosis factor (TNF) such as TNF-alpha or TNF-beta; insulin-like growth factor-I and -II (IGF-I and IGF-II); des(1-3)-IGF-I (brain IGF-I), insulin-like growth factor binding proteins; CD proteins such as CD3, CD4, CD8, CD19, CD20, CD22 and CD40; erythropoietin; osteoinductive factors; immunotoxins; a bone morphogenetic protein (BMP); an interferon such as interferon-alpha, -beta, and -gamma; colony stimulating factors (CSFs), e.g., M-CSF, GM-CSF, and G-CSF; interleukins (ILs), e.g., IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9 and IL-10; superoxide dismutase; T-cell receptors; surface membrane proteins; decay accelerating factor; viral antigen such as, for example, a portion of the AIDS envelope; transport proteins; homing receptors; addressins; regulatory proteins; integrins such as CD11a, CD11b, CD11c, CD18, an ICAM, VLA-4 and VCAM; a tumor associated antigen such as HER2, HER3 or HER4 receptor; and fragments of any of the above-listed polypeptides.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 5.5 to 7.0. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid.

In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In one embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal, chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 5.5 to 7.0. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 5.5 to 7.0. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a recombinant immunoglobulin; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a recombinant immunoglobulin; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a recombinant immunoglobulin; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.0 and 7.0. In a specific embodiment, the recombinant immunoglobulin is a monoclonal antibody. In a more specific embodiment, the recombinant antibody is an anti-MIF antibody.

C. Coagulation Proteins

Surprisingly, it was found that the stabilizing effects of alkali metal chloride salts (e.g., sodium and potassium chloride) are applicable to a wide range of labile therapeutic proteins ranging from plasma derived immunoglobulin preparations to recombinant coagulation factors. Accordingly, in one aspect, the present invention provides storage stable, aqueous compositions of labile coagulation factors formulated at mildly acidic to neutral pH with a moderate concentration of a metal chloride salt and a stabilizing agent.

In certain embodiments, the labile coagulation factor is a plasma derived protein or preparation. In other embodiments, the labile coagulation protein is a recombinantly expressed coagulation protein. Methods for manufacturing recombinant and plasma derived coagulation factors are well known in the art. Non-limiting examples of labile coagulation factors that may be formulated according to the methods provided herein include, Factor II (prothrombin), Factor III (platelet tissue factor), Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor XI, Factor XII, Factor XIII, von Willebrand Factor (vWF), and the like. In a preferred embodiment, the labile coagulation protein is selected from Factor VII, Factor VIII, Factor IX, and von Willebrand Factor (vWF). In another preferred embodiment, the labile coagulation protein is a vitamin-K dependent protein complex, for example, comprising Factor II, Factor IX, and Factor X, or comprising Factor II, Factor VII, Factor IX, and Factor X.

In one embodiment, the labile therapeutic coagulation protein is isolated from pooled plasma, i.e., plasma-derived coagulation factors. Methods for the isolation of many different coagulation factors are well known in the art. For example, Furuya et al. (“Implementation of a 20-nm pore-size filter in the plasma-derived factor VIII manufacturing process” Vox Sang. 2006 August; 91(2):119-25) describe a method for the purification of a virally reduced plasma-derived Factor VIII composition from pooled human plasma. Similarly, Kisiel et al. (“Activation of Bovine Factor VII (Proconvertin) by Factor XIIa (Activated Hageman Factor)” Biochemistry (1977) 16 (9):4189-4193) and Broze and Majerus (“Purification and Properties of Human Coagulation Factor VII” J Biol Chem. 1980 Feb. 25; 255(4):1242-7) describe methods for the purification of plasma-derived Factor VII. Methods for the purification of plasma-derived Protein K dependent coagulation complexes (e.g., Prothromplex, FEIBA) are described, for example, in U.S. Pat. Nos. 5,409,990 and 5,281,661. Finally, among other teachings, U.S. Pat. No. 4,786,726 and PCT Publication No. WO 2007/046631, describe purification of plasma-derived Factor FIX.

In other embodiments, the labile therapeutic coagulation protein is expressed recombinantly, i.e., recombinant coagulation factors. Methods for the expression and purification of many different coagulation factors are well known in the art. For example, U.S. Pat. Nos. 5,470,954; 6,100,061; 6,475,725; 6,555,391; 6,936,441; 7,094,574; 7,253,262; 6,919,311; 7,544,660; and 7,381,796, all of which are hereby incorporated by reference in their entireties for all purposes, describe the expression and purification of recombinant Factor VIII. Methods for the manufacture of recombinant Factor IX are also well known in the art and are described in, for example, U.S. Patent Application Publication 2008/207879, U.S. Pat. Nos. 4,770,999, and 5,521,070. Likewise, methods for the manufacture of recombinant Factor VII are described in, for example, U.S. Patent Application Publication Nos. 2010/120093 and 2009/047723.

In one embodiment of the present invention, the media used to express a recombinant coagulation factor can be animal protein-free and chemically defined. Methods of preparing animal protein-free and chemically defined culture mediums are known in the art, for example in US 2008/0009040 and US 2007/0212770, which are both incorporated herein for all purposes. “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.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In one embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a′ pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: a coagulation factor; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: a coagulation factor; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: a coagulation factor; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the coagulation factor is a core coagulation factor.

1. Factor VIII

As demonstrated in Example 5, recombinant Factor VIII (rFVIII) is stabilized by the addition of between about 100 mM and about 200 mM of an alkali metal chloride salt (e.g., sodium chloride) at a pH between about 6.0 and about 7.5. In one specific embodiment, methods are provided for the stabilization of Factor VIII formulated at a pH between 6.0 and 7.5, preferably at a pH from 6.5 to 7.0.

In one embodiment, the storage stable, aqueous FVIII compositions provided herein have a protein concentration of between 0.05 g/L and 10 g/L. In certain embodiments, the protein concentration of the FVIII composition is between 0.05 g/L and 2 g/L, or between 0.1 g/L and 1 g/L, or between 0.1 g/L and 0.5 g/L, or any suitable concentration within these ranges, for example 0.05 g/L, or 0.06 g/L, 0.07 g/L, 0.08 g/L, 0.09 g/L, 0.1 g/L, 0.15 g/L, 0.2 g/L, 0.25 g/L, 0.3 g/L, 0.35 g/L, 0.4 g/L, 0.45 g/L, 0.5 g/L, 0.55 g/L, 0.6 g/L, 0.65 g/L, 0.7 g/L, 0.75 g/L, 0.8 g/L, 0.85 g/L, 0.9 g/L, 0.95 g/L, 1 g/L, 1.25 g/L, 1.5 g/L, 1.75 g/L, 2.0 g/L, 2.25 g/L, 2.5 g/L, 2.75 g/L, 3.0 g/L, 3.25 g/L, 3.5 g/L, 3.75 g/L, 4.0 g/L, 4.25 g/L, 4.5 g/L, 4.75 g/L, 5.0 g/L, 5.5 g/L, 6.0 g/L, 6.5 g/L, 7.0 g/L, 7.5 g/L, 8.0 g/L, 8.5 g/L, 9.0 g/L, 9.5 g/L, 10.0 g/L, or higher. In a preferred embodiment, the aqueous FVIII composition will have a concentration of between 0.1 g/L and 0.5 g/L. In yet other embodiments, the concentration of Factor VIII in a storage-stable aqueous formulation is 0.05±0.01 g/L, or 0.06±0.01 g/L, 0.07±0.01 g/L, 0.08±0.01 g/L, 0.09±0.01 g/L, 0.1±0.01 g/L, 0.15±0.01 g/L, 0.2±0.02 g/L, 0.25±0.02 g/L, 0.3±0.03 g/L, 0.35±0.03 g/L, 0.4±0.04 g/L, 0.45±0.04 g/L, 0.5±0.05 g/L, 0.55±0.05 g/L, 0.6±0.06 g/L, 0.65±0.06 g/L, 0.7±0.07 g/L, 0.75±0.07 g/L, 0.8±0.08 g/L, 0.85±0.08 g/L, 0.9±0.09 g/L, 0.95±0.09 g/L, 1±0.1 g/L, 1.25±0.12 g/L, 1.5±0.15 g/L, 1.75±0.17 g/L, 2.0±0.2 g/L, 2.25±0.22 g/L, 2.5±0.25 g/L, 2.75±0.27 g/L, 3.0±0.3 g/L, 3.25±0.32 g/L, 3.5±0.35 g/L, 3.75±0.37 g/L, 4.0±0.4 g/L, 4.25±0.42 g/L, 4.5±0.45 g/L, 4.75±0.47 g/L, 5.0±0.5 g/L, 5.5±0.55 g/L, 6.0±0.6 g/L, 6.5±0.65 g/L, 7.0±0.7 g/L, 7.5±0.75 g/L, 8.0±0.8 g/L, 8.5±0.85 g/L, 9.0±0.9 g/L, 9.5±0.95 g/L, 10.0±1 g/L, or higher.

In one embodiment, the storage stable, aqueous FVIII composition will be stable under refrigeration (i.e., between about 2° C. and about 8° C.) for at least about 1 month. In other embodiments, the storage stable, aqueous FVIII composition will be stable under refrigeration for at least about 2 months. In a preferred embodiment, the storage stable, aqueous FVIII composition will be stable under refrigeration for at least about 3 months. In yet another embodiment, the storage stable, aqueous FVIII composition will be stable under refrigeration for at least about 6 months. In yet other embodiments, the storage stable, aqueous FVIII composition will be stable under refrigeration for at least about 2 weeks, or at least about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 weeks or at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or more months under refrigeration.

Accordingly, in one aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 125 mM to 175 mM of an alkali metal chloride salt; a stabilizing agent; and a pH of from 6.0 to 7.5. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting essentially of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein consisting of: a labile therapeutic protein; a stabilizing agent; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a more specific embodiment, the stabilizing agent is an amino acid. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In one embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; an amino acid; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM glycine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 5 mM to 500 mM glycine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM glycine. In a more specific embodiment, the composition contains from 100 mM to 400 mM glycine. In a more specific embodiment, the composition contains from 150 mM to 350 mM glycine. In a more specific embodiment, the composition contains from 200 mM to 300 mM glycine. In a more specific embodiment, the composition contains 250±25 mM glycine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM proline; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 5 mM to 500 mM proline; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM proline. In a more specific embodiment, the composition contains from 100 mM to 400 mM proline. In a more specific embodiment, the composition contains from 150 mM to 350 mM proline. In a more specific embodiment, the composition contains from 200 mM to 300 mM proline. In a more specific embodiment, the composition contains 250±25 mM proline. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; from 5 mM to 500 mM histidine; and a pH of from 6.0 to 7.5. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 5 mM to 500 mM histidine; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the composition contains from 25 mM to 500 mM histidine. In a more specific embodiment, the composition contains from 100 mM to 400 mM histidine. In a more specific embodiment, the composition contains from 150 mM to 350 mM histidine. In a more specific embodiment, the composition contains from 200 mM to 300 mM histidine. In a more specific embodiment, the composition contains 250±25 mM histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; an antioxidant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; an amino acid; an antioxidant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; a sugar and/or sugar alcohol; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In another embodiment, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; an amino acid; a sugar and/or sugar alcohol; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains calcium.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; mannitol; trehalose; calcium; a non-ionic surfactant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains an antioxidant. In a specific embodiment, the concentration of mannitol is 38±7 g/L. In another specific embodiment, the concentration of trehalose is 10±2 g/L. In another specific embodiment, the concentration of histidine is 12±2 mM. In another specific embodiment, the concentration of calcium is 1.9±0.4 mM. In another embodiment, the non-ionic surfactant is polysorbate-80 at a concentration of 0.15±0.03 g/L.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; mannitol; trehalose; calcium; a non-ionic surfactant; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains an antioxidant. In a specific embodiment, the concentration of mannitol is 38±7 g/L. In another specific embodiment, the concentration of trehalose is 10±2 g/L. In another specific embodiment, the concentration of histidine is 12±2 mM. In another specific embodiment, the concentration of calcium is 1.9±0.4 mM. In another embodiment, the non-ionic surfactant is polysorbate-80 at a concentration of 0.15±0.03 g/L.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; from 75 mM to 200 mM of an alkali metal chloride salt; an amino acid; mannitol; trehalose; calcium; a non-ionic surfactant; Tris; glutathione; and a pH of from 6.0 to 7.5. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific, embodiment, the composition also contains an antioxidant. In a specific embodiment, the concentration of mannitol is 38±7 g/L. In another specific embodiment, the concentration of trehalose is 10±2 g/L. In another specific embodiment, the concentration of histidine is 12±2 mM. In another specific embodiment, the concentration of calcium is 1.9±0.4 mM. In another embodiment, the non-ionic surfactant is polysorbate-80 at a concentration of 0.15±0.03 g/L.

In one aspect, the present invention provides a storage stable, aqueous composition comprising: Factor VIII; an amino acid; mannitol; trehalose; calcium; a non-ionic surfactant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains an antioxidant. In a specific embodiment, the concentration of mannitol is 38±7 g/L. In another specific embodiment, the concentration of trehalose is 10±2 g/L. In another specific embodiment, the concentration of histidine is 12±2 mM. In another specific embodiment, the concentration of calcium is 1.9±0.4 mM. In another embodiment, the non-ionic surfactant is polysorbate-80 at a concentration of 0.15±0.03 g/L.

In a specific embodiment, the present invention provides a storage stable, aqueous composition consisting essentially of: Factor VIII; an amino acid; mannitol; trehalose; calcium; a non-ionic surfactant; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains an antioxidant. In a specific embodiment, the concentration of mannitol is 38±7 g/L. In another specific embodiment, the concentration of trehalose is 10±2 g/L. In another specific embodiment, the concentration of histidine is 12±2 mM. In another specific embodiment, the concentration of calcium is 1.9±0.4 mM. In another embodiment, the non-ionic, surfactant is polysorbate-80 at a concentration of 0.15±0.03 g/L.

In a more specific embodiment, the present invention provides a storage stable, aqueous composition consisting of: Factor VIII; an amino acid; mannitol; trehalose; calcium; a non-ionic surfactant; Tris; glutathione; and an alkali metal salt/pH combination selected from any one of variations 1 to 2088, as set forth in Table 1, Table 2, Table 3, and Table 4. In a specific embodiment, the amino acid is glycine, proline, or histidine. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline. In another preferred embodiment, the amino acid is histidine. In one embodiment, the metal chloride salt is sodium chloride. In another embodiment, the metal chloride salt is potassium chloride. In a preferred embodiment, the pH of the formulation is between 6.5 and 7.5. In a specific embodiment, the composition also contains an antioxidant. In a specific embodiment, the concentration of mannitol is 38±7 g/L. In another specific embodiment, the concentration of trehalose is 10±2 g/L. In another specific embodiment, the concentration of histidine is 12±2 mM. In another specific embodiment, the concentration of calcium is 1.9±0.4 mM. In another embodiment, the non-ionic surfactant is polysorbate-80 at a concentration of 0.15±0.03 g/L.

D. Methods for Stabilizing Labile Proteins

In the context of the present invention, a labile therapeutic protein is unstable when formulated at mildly acidic to neutral pH in the absence of an alkaline metal chloride salt. Surprisingly, it has been found that a wide range of labile therapeutic proteins are stabilized by the addition of a moderate concentration (i.e., between about 75 mM and about 200 mM, preferably between about 100 mM and about 200 mM) of an alkaline metal chloride salt. This effect of alkali metal chloride salts provides methods for stabilizing aqueous formulations of labile therapeutic proteins. Accordingly, in one aspect of the present invention, methods are provided for the stabilization of an aqueous labile therapeutic protein composition. These methods allow for aqueous formulations of labile therapeutic proteins at mildly acidic to neutral pH, which previously required lyophilization, freezing in the presence of several stabilizers, or formulation at extreme pH values.

In one embodiment, the method comprises the addition of alkali metal chloride salt to a final concentration of between about 75 mM and about 200 mM to an aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 7.5. In a preferred embodiment, the method further comprises the addition of a stabilizing agent, such as an amino acid, to the formulation.

In another embodiment, the method comprises the addition of alkali metal chloride salt to a final concentration of between about 100 mM and about 200 mM to an aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 7.5. In a preferred embodiment, the method further comprises the addition of a stabilizing agent, such as an amino acid, to the formulation.

In another embodiment, the method comprises the addition of alkali metal chloride salt to a final concentration of between about 100 mM and about 200 mM to an aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 7.0. In a preferred embodiment, the method further comprises the addition of a stabilizing agent, such as an amino acid, to the formulation.

In yet another embodiment, the method comprises the addition of alkali metal chloride salt to a final concentration of between about 75 mM and about 200 mM to an aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 7.0. In a preferred embodiment, the method further comprises the addition of a stabilizing agent, such as an amino acid, to the formulation.

In certain embodiments, the methods provided herein for the stabilization of a labile therapeutic protein will further comprise the addition of a stabilizing agent to the formulation. In a preferred embodiment, the stabilizing agent will be an amino acid. Exemplary amino acids that may be used for this purpose include, without limitation, arginine, histidine, lysine, serine, proline, glycine, alanine, threonine, and a combination thereof. In a preferred embodiment, the amino acid is glycine. In another preferred embodiment, the amino acid is proline.

For purposes of further stabilizing the compositions provided herein, the amino acid will typically be added to the formulation at a concentration between about 25 mM and about 0.75 M. In one embodiment, at least about 100 mM of the amino acid is added to the formulation. In another embodiment, at least about 200 mM of the amino acid is added to the formulation. In yet another embodiment, at least about 250 mM of the amino acid is added to the formulation. In yet other embodiments, the formulations provided herein will contain at least about 25 mM of the amino acid, or at least about 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, or more of the amino acid.

In a preferred embodiment, the labile therapeutic protein is a human protein, a humanized protein, or a chimeric human protein. The human protein may be either purified from a natural source (e.g., pooled human plasma) or expressed recombinantly, for example in a mammalian cell or tissue culture. In a preferred embodiment, the labile therapeutic protein is a plasma-derived protein, preferably a plasma-derived coagulation factor or immunoglobulin preparation. In another preferred embodiment, the human, humanized, or chimeric protein in a recombinant antibody or fragment thereof.

In one embodiment, the methods for stabilizing a formulation of a labile therapeutic protein provided herein comprise the addition of between about 75 mM and about 200 mM of an alkali metal chloride salt to the formulation. In a preferred embodiment, the methods for stabilizing a formulation of a labile therapeutic protein provided herein comprise the addition of between about 100 mM and about 200 mM of an alkali metal chloride salt to the formulation. In certain embodiments, the methods comprise the addition of between about 150 mM and about 200 mM of an alkali metal chloride salt. In other embodiments, the method comprises the addition of between about 150 mM and about 200 mM of an alkali metal chloride salt. In yet other embodiments, the comprise the addition of at or about 70 mM or at or about 75 mM, 80 mM, 85 mM, 90 mM, 95 mM, 100 mM, 105 mM, 110 mM, 115 mM, 120 mM, 125 mM, 130 mM, 135 mM, 140 mM, 145 mM, 150 mM, 155 mM, 160 mM, 165 mM, 170 mM, 175 mM, 180 mM, 185 mM, 190 mM, 195 mM, 200 mM, 205 mM, 210 mM, 215 mM, or at or about 220 mM of an alkali metal chloride salt. In one preferred embodiment, the alkali metal chloride salt is sodium chloride. In another preferred embodiment, the alkali metal chloride salt is potassium chloride.

The methods for stabilizing a labile therapeutic protein provided by the present invention comprise formulating the labile protein, in the presence of an alkali metal chloride salt, at a mildly acidic to neutral pH. Generally, this includes pH values between about 5.5 and about 7.5. However, the range of pH values at which any individual labile therapeutic protein is stabilized by the addition of a moderate level (i.e., between about 75 mM and about 200 mM, preferably between about 100 mM and about 200 mM) of an alkali metal chloride salt may vary slightly, dependent upon the properties of the individual protein. In a preferred embodiment, the method will comprise formulating a storage stable composition of a labile therapeutic protein at a pH between about 5.5 and about 7.0. In another embodiment, the method will comprise formulating a storage stable composition of a labile therapeutic protein at a pH between about 5.5 and about 6.5. In other embodiments, the pH of the stabilizing formulation will be between about 6.0 and about 7.0. In another embodiment, the pH of the stabilizing formulation will be between about 5.5 and about 6.0. In one embodiment, the pH of the stabilizing formulation will be between about 6.0 and about 6.5. In another embodiment, the pH of the stabilizing formulation will be between about 6.5 and about 7.0. In another embodiment, the pH of the stabilizing formulation will be between about 6.0 and about 7.5. In another embodiment, the pH of the stabilizing formulation will be between about 6.5 and about 7.5. In another embodiment, the pH of the stabilizing formulation will be between about 7.0 and about 7.5. In yet other embodiments, the pH of the stabilizing formulation may be at or about 5.5, or at or about 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, or at or about 7.5.

In certain embodiments, the method will further comprise formulating the storage stable, labile therapeutic protein for parenteral administration including, but not limited to, intradermal, subcutaneous, transdermal implant, intracavernous, intravitreal, transscleral, intracerebral, intrathecal, epidural, intravenous, intracardiac, intramuscular, intraosseous, intraperitoneal, and nanocell injection administration. In one preferred embodiment, the compositions provided herein will be formulated for intravenous administration. In another preferred embodiment, the compositions provided herein will be formulated for subcutaneous administration. In yet another preferred embodiment, the compositions provided herein will be formulated for intramuscular administration.

In certain embodiments, the method for stabilizing a labile therapeutic protein will comprise formulating the protein at a final concentration of between about 0.05 mg/mL to about 250 mg/mL. In certain embodiments, the labile protein will be formulated at a final concentration of about 0.05 mg/mL, 0.06 mg/mL, 0.07 mg/mL, 0.08 mg/mL, 0.09 mg/mL, 0.1 mg/mL, 0.2 mg/mL, 0.3 mg/mL, 0.4 mg/mL, 0.5 mg/mL or about 1 mg/mL, 2 mg/mL, 3 mg/mL, 4 mg/mL, 5 mg/mL, 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, 10 mg/mL, 11 mg/mL, 12 mg/mL, 13 mg/mL, 14 mg/mL, 15 mg/mL, 16 mg/mL, 17 mg/mL, 18 mg/mL, 19 mg/mL, 20 mg/mL, 21 mg/mL, 22 mg/mL, 23 mg/mL, 24 mg/mL, 25 mg/mL, 26 mg/mL, 27 mg/mL, 28 mg/mL, 29 mg/mL, 30 mg/mL, 35 mg/mL, 40 mg/mL, 45 mg/mL, 50 mg/mL, 55 mg/mL, 60 mg/mL, 65 mg/mL, 70 mg/mL, 75 mg/mL, 80 mg/mL, 85 mg/mL, 90 mg/mL, 95 mg/mL, 100 mg/mL, 110 mg/mL, 120 mg/mL, 130 mg/mL, 140 mg/mL, 150 mg/mL, 160 mg/mL, 170 mg/mL, 180 mg/mL, 190 mg/mL, 200 mg/mL, 210 mg/mL, 220 mg/mL, 230 mg/mL, 240 mg/mL, 250 mg/mL, or higher concentrations, depending upon the characteristics of the protein being formulated, the intended therapeutic use of the protein, and the preferred method of administration.

In one embodiment, the method will comprise formulating the labile therapeutic protein at a low final protein concentration of between about 0.05 mg/mL and about 20 mg/mL. In another embodiment, the final protein concentration may be between about 0.5 mg/mL and about 15 mg/mL. In another embodiment, the final protein concentration may be between about 0.5 mg/mL and about 10 mg/mL. In another embodiment, the final protein concentration may be between about 0.5 mg/mL and about 5 mg/mL. In one embodiment, a composition with a final protein concentration as described above will be formulated for intravenous administration.

In other embodiments, the method will comprise formulating the labile therapeutic protein at a moderate final protein concentration of between about 5 mg/mL and about 25 mg/mL. In another embodiment, the final protein concentration may be between about 10 mg/mL and about 25 mg/mL. In another embodiment, the final protein concentration may be between about 15 mg/mL and about 25 mg/mL. In another embodiment, the final protein concentration may be between about 20 mg/mL and about 25 mg/mL. In one embodiment, a composition with a final protein concentration as described above will be formulated for subcutaneous or intramuscular administration.

In certain embodiments, the final protein concentration may be between about 0.5% and about 25%. In another embodiment, the final protein concentration may be between about 0.5% and about 20%. In another embodiment, the final protein concentration may be between about 0.5% and about 15%. In another embodiment, the final protein concentration may be between about 0.5% and about 10%. In another embodiment, the final protein concentration may be between about 0.5% and about 5%. In one embodiment, a composition with a final protein concentration as described above will be formulated for intravenous administration.

In certain embodiments, the final protein concentration may be between about 5% and about 25%. In another embodiment, the final protein concentration may be between about 10% and about 25%. In another embodiment, the final protein concentration may be between about 15% and about 25%. In another embodiment, the final protein concentration may be between about 20% and about 25%. In one embodiment, a composition with a final protein concentration as described above will be formulated for subcutaneous or intramuscular administration.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile plasma derived protein at a pH between about 5.5 and about 7.5, the method comprising adding an alkali metal chloride salt to the formulation at a final concentration of between about 75 mM and about 200 mM. In a preferred embodiment, the compositions comprise between about 100 mM and about 200 mM of an alkali metal chloride salt. In another preferred embodiment, the mildly acidic to neutral pH is between about 5.5 and about 7.0. In a preferred embodiment, the alkali metal chloride salt is sodium chloride. In another preferred embodiment, the alkali metal chloride salt is potassium chloride.

The methods provided herein allow for the stabilization of a labile protein at a mildly acidic to neutral pH for an extended period of time. For example, in one embodiment, the storage stable, aqueous labile therapeutic protein composition will be stable for at least about 2 months. In another embodiment, the composition will be stable for at least about 3 months. In yet other embodiment, the composition will be stable for at least 1 about month, or at least about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, or more months. In a preferred embodiment, the composition will be stable for at least about 6 months. In a more preferred embodiment, the composition will be stable for at least about 1 year. In a more preferred embodiment, the composition will be stable for at least about 2 years.

Dependent upon the individual characteristics of the labile protein being stabilized in the methods provided herein, the formulation will be stabile for an extended period of time at a temperature between about 2° C. and about 42° C. In one embodiments, a labile therapeutic protein will be stabilized by the methods provided herein when stored under refrigeration, i.e., stored at a temperature between about 2° C. and about 8° C. In another embodiment, a labile therapeutic protein will be stabilized by the methods provided herein when stored at room temperature, i.e., stored at a temperature between about 20° C. and about 25° C. In other embodiments, the protein may be stabilized when stored at a temperature between about 28° C. and about 32° C. In yet another embodiment, the protein may be stabilized when stored at a temperature between about 38° C. and about 42° C. The temperatures at which a labile therapeutic protein will be stabilized by the methods provided herein will be dependent upon the characteristics of the individual protein, which can readily be determined by one of skill in the art.

In certain embodiments, the methods provided herein for the stabilization of a labile therapeutic protein comprise the addition of a stabilizing agent. In one embodiment, the stabilizing agent comprises one or more buffering agents or pH stabilizing agents suitable for intravenous, intravitreal, subcutaneous, and/or intramuscular administration. Non-limiting examples of buffering agents suitable for formulating the storage stable compositions provided herein include glycine, histidine, or other amino acids, salts like citrate, phosphate, acetate, glutamate, tartrate, benzoate, lactate, gluconate, malate, succinate, formate, propionate, carbonate, or any combination thereof adjusted to an appropriate pH. Generally, the buffering agent will be sufficient to maintain a suitable pH in the formulation for an extended period of time.

In some embodiments, the concentration of buffering agent in the formulation will be at or about between 5 mM and 500 mM. In certain embodiments, the concentration of the buffering agent in the formulation will be at or about 5 mM, 10 mM, 15 mM, 20 mM, 25 mM, 50 mM, 75 mM, 100 mM, 125 mM, 150 mM, 175 mM, 200 mM, 225 mM, 250 mM, 275 mM, 300 mM, 325 mM, 350 mM, 375 mM, 400 mM, 425 mM, 450 mM, 475 mM, 500 mM or higher.

In another embodiment, the stabilizing agent will comprise an agent for adjusting the osmolarity of the composition. Non-limiting examples of osmolarity agents include mannitol, sorbitol, glycerol, sucrose, glucose, dextrose, levulose, fructose, lactose, polyethylene glycols, phosphates, calcium chloride, calcium gluconoglucoheptonate, dimethyl sulfone, and the like.

In a preferred embodiment, the stabilizing agent employed in the storage stable, labile immunoglobulin formulations provided herein will be an amino acid. Stabilizing amino acids include arginine, histidine, lysine, serine, proline, glycine, alanine, threonine, and a combination thereof. In a preferred embodiment, the amino acid is glycine. For purposes of further stabilizing the compositions provided herein, the amino acid will typically be added to the formulation at a concentration between about 25 mM and about 0.75 M. In one embodiment, at least about 100 mM of the amino acid is added to the formulation. In another embodiment, at least about 200 mM of the amino acid is added to the formulation. In yet another embodiment, at least about 250 mM of the amino acid is added to the formulation. In yet other embodiments, the formulations provided herein will contain at least about 25 mM of the amino acid, or at least about 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, or more of the amino acid.

In a specific embodiment, the labile plasma derived protein is an immunoglobulin preparation. As demonstrated in Examples 1 and 2, plasma derived IgG preparations are stabilized by the addition of between about 100 mM and about 200 mM of an alkali metal chloride salt (e.g., sodium chloride) at a pH between about 6.0 and about 7.0. As shown in FIG. 1, maximum stability for the IgG formulations is found between pH 6.0 and 6.5.

Accordingly, in one embodiment, the present invention provides method for stabilizing a plasma derived IgG aqueous composition formulated at a pH between about 6.0 and about 7.0, the method comprising adding between about 75 mM and about 200 mM, preferably between about 100 mM and about 200 mM of an alkali metal chloride salt to the formulation. In a preferred embodiment, the method further comprises adding a stabilizing agent to the formulation. In another preferred embodiment, the method comprises the addition of between about 75 mM and about 200 mM, preferably between about 100 mM and about 200 mM, of an alkali metal chloride salt to a plasma derived immunoglobulin preparation formulated at a pH between about 6.0 and about 6.5. In a preferred embodiment, the salt is sodium chloride. In another preferred embodiment, the salt is potassium chloride.

In one embodiment, the method for stabilizing a labile plasma derived immunoglobulin composition will comprise formulating the protein at a final concentration of between about 30 g/L and about 250 g/L. In certain embodiments, the method comprises formulating the immunoglobulin composition at a final protein concentration of between about 50 g/L and about 200 g/L, or between about 70 g/L and about 150 g/L, or between about 90 g/L and about 120 g/L, or any suitable concentration within these ranges, for example about 30 g/L, or about 35 g/L, 40 g/L, 45 g/L, 50 g/L, 55 g/L, 60 g/L, 65 g/L, 70 g/L, 75 g/L, 80 g/L, 85 g/L, 90 g/L, 95 g/L, 100 g/L, 105 g/L, 110 g/L, 115 g/L, 120 g/L, 125 g/L, 130 g/L, 135 g/L, 140 g/L, 145 g/L, 150 g/L, 155 g/L, 160 g/L, 165 g/L, 170 g/L, 175 g/L, 180 g/L, 185 g/L, 190 g/L, 195 g/L, 200 g/L, 205 g/L, 210 g/L, 215 g/L, 220 g/L, 225 g/L, 230 g/L, 235 g/L, 240 g/L, 245 g/L, 250 g/L, or higher. In a preferred embodiment, the aqueous IgG composition will have a concentration of at or about 100 g/L. In a related embodiment, the aqueous IgG composition will have a concentration of between about 70 g/L and about 130 g/L. In another preferred embodiment, the aqueous IgG composition will have a concentration of at or about 200 g/L. In a related embodiment, the aqueous IgG composition will have a concentration of between about 170 g/L and about 230 g/L.

In one aspect, the present invention provides methods for stabilizing recombinant immunoglobulin preparations at a pH between about 5.5 and about 7.5 comprising the addition of between about 75 mM and about 200 mM of an alkali metal chloride salt to the formulation. In a preferred embodiment, the compositions comprise between about 100 mM and about 200 mM of an alkali metal chloride salt. In another preferred embodiment, the mildly acidic to neutral pH is between about 5.5 and about 7.0. In a preferred embodiment, the methods further comprise the addition of a stabilizing agent to the formulation. In one preferred embodiment, the salt is sodium chloride. In another preferred embodiment, the salt is potassium chloride.

In another specific embodiment, the storage stable, immunoglobulin composition is a recombinant antibody preparation. As demonstrated in Example 4, a recombinant anti-MIF monoclonal antibody preparation is stabilized by the addition of between about 100 mM and about 200 mM of an alkali metal chloride salt (e.g., sodium chloride) at a pH between about 5.5 and about 6.5.

Accordingly, in one embodiment, the present invention provides methods for stabilizing aqueous compositions of recombinant immunoglobulins formulated at a pH between about 5.5 and about 6.5 by the addition of between about 75 mM and about 200 mM, preferably between about 100 mM and about 200 mM, of an alkali metal chloride salt and optionally, a stabilizing agent to the formulation. In a preferred embodiment, the salt is sodium chloride. In another preferred embodiment, the salt is potassium chloride.

In one embodiment, the methods for stabilizing a recombinant immunoglobulin aqueous composition provided herein comprise the formulation of the recombinant immunoglobulin at a final protein concentration of between about 1 g/L and about 250 g/L. In certain embodiments, the protein concentration of the recombinant immunoglobulin composition is between about 50 g/L and about 200 g/L, or between about 70 g/L and about 150 g/L, or between about 90 g/L and about 120 g/L, or any suitable concentration within these ranges, for example about 1 g/L, 2 g/L, 3 g/L, 4 g/L, 5 g/L, 6 g/L, 7 g/L, 8 g/L, 9 g/L, 10 g/L, 11 g/L, 12 g/L, 13 g/L, 14 g/L, 15 g/L, 16 g/L, 17 g/L, 18 g/L, 19 g/L, 20 g/L, 25 g/L, 30 g/L, or about 35 g/L, 40 g/L, 45 g/L, 50 g/L, 55 g/L, 60 g/L, 65 g/L, 70 g/L, 75 g/L, 80 g/L, 85 g/L, 90 g/L, 95 g/L, 100 g/L, 105 g/L, 110 g/L, 115 g/L, 120 g/L, 125 g/L, 130 g/L, 135 g/L, 140 g/L, 145 g/L, 150 g/L, 155 g/L, 160 g/L, 165 g/L, 170 g/L, 175 g/L, 180 g/L, 185 g/L, 190 g/L, 195 g/L, 200 g/L, 205 g/L, 210 g/L, 215 g/L, 220 g/L, 225 g/L, 230 g/L, 235 g/L, 240 g/L, 245 g/L, 250 g/L, or higher.

In another embodiment, the present invention provides methods for stabilizing a labile coagulation factor formulated at a pH between about 5.5 and about 7.5 comprising the addition of between about 75 mM and about 200 mM of an alkali metal chloride salt to the formulation. In a preferred embodiment, the compositions comprise between about 100 mM and about 200 mM of an alkali metal chloride salt. In another preferred embodiment, the mildly acidic to neutral pH is between about 5.5 and about 7.0. In a preferred embodiment, the methods further comprise the addition of a stabilizing agent to the formulation. In one preferred embodiment, the salt is sodium chloride. In another preferred embodiment, the salt is potassium chloride.

In certain embodiments, the labile coagulation protein is a plasma derived protein or preparation. In other embodiments, the labile coagulation protein is a recombinantly expressed coagulation protein. Methods for manufacturing recombinant and plasma derived coagulation factors are well known in the art. Non-limiting examples of coagulation factors that may be formulated according to the methods provided herein include, Factor II (prothrombin), Factor III (platelet tissue factor), Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor XI, Factor XII, Factor XIII, von Willebrand Factor (vWF), and the like. In a preferred embodiment, the labile coagulation protein is selected from Factor VII, Factor VIII, Factor IX, and von Willebrand Factor (vWF). In another preferred embodiment, the labile coagulation protein is a vitamin-K dependent protein complex, for example, comprising Factor II, Factor IX, and Factor X, or comprising Factor II, Factor VII, Factor IX, and Factor X.

In one specific embodiment, methods are provided for the stabilization of Factor VIII formulated at a pH between about 6.5 and about 7.0. As shown in Example 5, recombinant Factor VIII (rFVIII) is stabilized by the addition of between about 100 mM and about 200 mM of an alkali metal chloride salt (e.g., sodium chloride) at a pH between about 6.5 and about 7.0.

Accordingly, in one embodiment the present invention provides a method for stabilizing an aqueous composition of FVIII, the method comprising formulating a FVIII composition at a pH between about 6.5 and about 7.0 with between about 75 mM and about 200 mM, preferably between about 100 mM and about 200 mM, of an alkali metal chloride salt. In a preferred embodiment, the alkali metal chloride salt is sodium chloride. In another preferred embodiment, the alkali metal chloride salt is potassium chloride.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 7.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 75 mM and about 200 mM. In a preferred embodiment, the compositions comprise between about 100 mM and about 200 mM of an alkali metal chloride salt. In another preferred embodiment, the mildly acidic to neutral pH is between about 5.5 and about 7.0. In a preferred embodiment, the salt is sodium chloride. In certain embodiments, the method further comprises the addition of a stabilizing agent. In one embodiment, the stabilizing agent is an amino acid. In preferred embodiments, the amino acid is glycine or proline.

In another embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 6.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 100 mM and about 200 mM. In a preferred embodiment, the salt is sodium chloride. In certain embodiments, the method further comprises the addition of a stabilizing agent. In one embodiment, the stabilizing agent is an amino acid. In preferred embodiments, the amino acid is glycine or proline.

In one embodiment of the methods provided herein, the labile therapeutic protein is a human or humanized protein. In another embodiment of the methods provided herein, the labile therapeutic protein is a recombinant protein. In yet another embodiment of the methods provided herein, the labile therapeutic protein is a plasma-derived protein.

In a specific embodiment of the methods provided herein, the labile therapeutic protein is an immunoglobulin. In one embodiment, the immunoglobulin is an IgG preparation. In another embodiment, the immunoglobulin is a recombinant antibody.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile immunoglobulin preparation at a pH between about 5.5 and about 7.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 75 mM and about 200 mM, wherein the method stabilizes the immunoglobulin composition for at least 6 months when stored at a temperature at or below about 42° C., for example, at a temperature between about 38° C. and about 42° C. In a preferred embodiment, the pH of the immunoglobulin formulation is between about 5.5 and about 6.5.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile immunoglobulin preparation at a pH between about 5.5 and about 7.0, the method comprising addition of an alkali metal chloride salt at a concentration of between about 100 mM and about 200 mM, wherein the method stabilizes the immunoglobulin composition for at least 6 months when stored at a temperature at or below about 42° C., for example, at a temperature between about 38° C. and about 42° C. In a preferred embodiment, the pH of the immunoglobulin formulation is between about 5.5 and about 6.5.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile immunoglobulin preparation at a pH between about 5.5 and about 7.0, the method comprising addition of an alkali metal chloride salt at a concentration of between about 100 mM and about 200 mM, wherein the method stabilizes the immunoglobulin composition for at least about one year when stored at a temperature at or below about 32° C., for example, at a temperature between about 28° C. and about 32° C. In a preferred embodiment, the pH of the immunoglobulin formulation is between about 5.5 and about 6.5.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile immunoglobulin preparation at a pH between about 5.5 and about 7.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 75 mM and about 200 mM, wherein the method stabilizes the immunoglobulin composition for at least about one year when stored at a temperature at or below about 32° C., for example, at a temperature between about 28° C. and about 32° C. In a preferred embodiment, the pH of the immunoglobulin formulation is between about 5.5 and about 6.5.

In a specific embodiment of the methods provided herein, the labile therapeutic protein is a labile coagulation protein. In one embodiment, the coagulation protein is Factor VIII. In another embodiment, the coagulation protein is Factor VII. In yet another embodiment, the coagulation protein is Factor IX. In another embodiment, the coagulation protein is a protein K-dependent coagulation complex, for example, comprising Factor II, Factor IX, and Factor X, or comprising Factor II, Factor VII, Factor IX, and Factor X.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile coagulation protein at a pH between about 5.5 and about 7.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 75 mM and about 200 mM, wherein the method stabilizes the coagulation protein for at least 3 months when stored at a refrigerated temperature, for example, at a temperature between about 2° C. and about 10° C.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile coagulation protein at a pH between about 5.5 and about 7.0, the method comprising addition of an alkali metal chloride salt at a concentration of between about 100 mM and about 200 mM, wherein the method stabilizes the coagulation protein for at least 3 months when stored at a refrigerated temperature, for example, at a temperature between about 2° C. and about 10° C.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile coagulation protein at a pH between about 5.5 and about 7.0, the method comprising addition of an alkali metal chloride salt at a concentration of between about 100 mM and about 200 mM, wherein the method stabilizes the coagulation protein for at least 6 months when stored at a refrigerated temperature, for example, at a temperature between about 2° C. and about 10° C.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile coagulation protein at a pH between about 5.5 and about 7.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 75 mM and about 200 mM, wherein the method stabilizes the coagulation protein for at least 6 months when stored at a refrigerated temperature, for example, at a temperature between about 2° C. and about 10° C.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile coagulation protein at a pH between about 5.5 and about 7.0, the method comprising addition of an alkali metal chloride salt at a concentration of between about 100 mM and about 200 mM, wherein the method stabilizes the coagulation protein for at least about one year when stored at a refrigerated temperature, for example, at a temperature between about 2° C. and about 10° C.

In one embodiment, the present invention provides a method for stabilizing an aqueous formulation of a labile coagulation protein at a pH between about 5.5 and about 7.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 75 mM and about 200 mM, wherein the method stabilizes the coagulation protein for at least about one year when stored at a refrigerated temperature, for example, at a temperature between about 2° C. and about 10° C.

E. Specific Embodiments

In one embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: between about 75 mM and about 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH between about 5:5 and about 7.5.

In one embodiment, the present invention provides a storage stable, aqueous composition of a labile therapeutic protein comprising: between about 100 mM and about 200 mM of an alkali metal chloride salt; a stabilizing agent; and a pH between about 5.5 and about 7.0.

In a specific embodiment of the compositions provided above, the labile therapeutic protein is a human or humanized protein.

In a specific embodiment of the compositions provided above, the protein is a recombinant protein.

In a specific embodiment of the compositions provided above, the protein is a plasma-derived protein.

In a specific embodiment of the compositions provided above, the protein is an immunoglobulin.

In a specific embodiment of the compositions provided above, the immunoglobulin is an IgG preparation.

In a specific embodiment of the compositions provided above, the protein concentration of the IgG preparation is at least about 100 mg/mg.

In a specific embodiment of the compositions provided above, the protein concentration of the IgG preparation is at least about 200 mg/mL.

In a specific embodiment of the compositions provided above, the immunoglobulin is a recombinant antibody.

In a specific embodiment of the compositions provided above, the composition is stable for at least 6 months when stored at between about 38° C. and about 42° C.

In a specific embodiment of the compositions provided above, the composition is stable for at least 1 year when stored at between about 28° C. and about 32° C.

In a specific embodiment of the compositions provided above, the pH of the composition is between about 5.5 and about 6.5.

In a specific embodiment of the compositions provided above, the protein is a coagulation factor.

In a specific embodiment of the compositions provided above, the coagulation factor is Factor VIII.

In a specific embodiment of the compositions provided above, the pH of the composition is between about 6.0 and about 7.0.

In a specific embodiment of the compositions provided above, the pH of the composition is 6.5±0.2.

In a specific embodiment of the compositions provided above, the composition retains at least 80% of its Factor VIII activity when stored at a temperature between about 2° C. and about 8° C. for at least 3 months.

In a specific embodiment of the compositions provided above, the coagulation factor is Factor VII.

In a specific embodiment of the compositions provided above, the coagulation factor is Factor IX.

In a specific embodiment of the compositions provided above, the coagulation factor is von Willebrand Factor (vWF).

In a specific embodiment of the compositions provided above, the coagulation factor is a protein K-dependent coagulation complex.

In a specific embodiment of the compositions provided above, the protein K-dependent coagulation complex comprises the coagulation factors Factor II, Factor IX, and Factor X.

In a specific embodiment of the compositions provided above, the protein K-dependent coagulation complex further comprises Factor VII.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 3 months in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 2 months in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 1 month in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the compositions provided above, the labile protein is stable for less than 2 weeks in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the compositions provided above, the concentration of the labile therapeutic protein is at least about 50 mg/mL.

In a specific embodiment of the compositions provided above, the concentration of the labile therapeutic protein is at least about 100 mg/mL.

In a specific embodiment of the compositions provided above, the concentration of the labile therapeutic protein is at least about 150 mg/mL.

In a specific embodiment of the compositions provided above, the composition is formulated for subcutaneous or intramuscular administration.

In a specific embodiment of the compositions provided above, the alkali metal chloride salt is sodium chloride.

In a specific embodiment of the compositions provided above, the stabilizing agent is an amino acid.

In a specific embodiment of the compositions provided above, the amino acid is glycine.

In a specific embodiment of the compositions provided above, the concentration of the amino acid is at least about 100 mM.

In a specific embodiment of the compositions provided above, the composition is stable for at least about 3 months.

In a specific embodiment of the compositions provided above, the composition is stable for at least about 6 months.

In a specific embodiment of the compositions provided above, the composition is stable for at least about 1 year.

In a specific embodiment of the compositions provided above, the composition is stable for at least about 2 years.

In one embodiment, the present invention provides a method for stabilizing a aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 7.5, the method comprising addition of an alkali metal chloride salt at a concentration of between about 75 mM and about 200 mM.

In one embodiment, the present invention provides a method for stabilizing a aqueous formulation of a labile therapeutic protein at a pH between about 5.5 and about 7.0, the method comprising addition of an alkali metal chloride salt at a concentration of between about 100 mM and about 200 mM.

In a specific embodiment of the methods provided above, the solution further comprises a stabilizing agent.

In a specific embodiment of the methods provided above, the labile therapeutic protein is a human or humanized protein.

In a specific embodiment of the methods provided above, the protein is a recombinant protein.

In a specific embodiment of the methods provided above, the protein is a plasma-derived protein.

In a specific embodiment of the methods provided above, the protein is an immunoglobulin.

In a specific embodiment of the methods provided above, the immunoglobulin is an IgG preparation.

In a specific embodiment of the methods provided above, the protein concentration of the IgG preparation is at least about 150 mg/mg.

In a specific embodiment of the methods provided above, the protein concentration of the IgG preparation is at least about 200 mg/mg.

In a specific embodiment of the methods provided above, the immunoglobulin is a recombinant antibody.

In a specific embodiment of the methods provided above, the method stabilizes the immunoglobulin composition for at least 6 months when stored at between about 38° C. and about 42° C.

In a specific embodiment of the methods provided above, the method stabilizes the immunoglobulin composition for at least 1 year when stored at between about 28° C. and about 32° C.

In a specific embodiment of the methods provided above, the pH of the composition is between about 5.5 and about 6.5.

In a specific embodiment of the methods provided above, the protein is a coagulation protein.

In a specific embodiment of the methods provided above, the coagulation protein is Factor VIII.

In a specific embodiment of the methods provided above, the pH of the composition is between about 6.0 and about 7.0.

In a specific embodiment of the methods provided above, the pH of the composition is 6.5±0.2.

In a specific embodiment of the methods provided above, the method stabilizes the composition such that at least 80% of the Factor VIII activity is retained after storage at a temperature between about 2° C. and about 8° C. for at least 3 months.

In a specific embodiment of the methods provided above, the coagulation protein is Factor VII.

In a specific embodiment of the methods provided above, the coagulation protein is Factor IX.

In a specific embodiment of the methods provided above, the coagulation protein is von Willebrand Factor (vWF).

In a specific embodiment of the methods provided above, the coagulation protein is a protein K-dependent coagulation complex.

In a specific embodiment of the methods provided above, the protein K-dependent coagulation complex comprises the coagulation factors Factor II, Factor IX, and Factor X.

In a specific embodiment of the methods provided above, the protein K-dependent coagulation complex further comprises Factor VII.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 3 months in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 2 months in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 1 month in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the methods provided above, the labile protein is stable for less than 2 weeks in an aqueous formulation containing less than about 50 mM of an alkali metal chloride salt at a pH between about 5.5 and about 7.5.

In a specific embodiment of the methods provided above, the concentration of the labile therapeutic protein is at least about 50 mg/mL.

In a specific embodiment of the methods provided above, the concentration of the labile therapeutic protein is at least about 100 mg/mL.

In a specific embodiment of the methods provided above, the concentration of the labile therapeutic protein is at least about 150 mg/mL.

In a specific embodiment of the methods provided above, the composition is formulated for subcutaneous or intramuscular administration.

In a specific embodiment of the methods provided above, the alkali metal chloride salt is sodium chloride.

In a specific embodiment of the methods provided above, the stabilizing agent is an amino acid.

In a specific embodiment of the methods provided above, the amino acid is glycine.

In a specific embodiment of the methods provided above, the concentration of the amino acid is at least about 100 mM.

In a specific embodiment of the methods provided above; the method increases the time of stability for the composition by at least 25%.

In a specific embodiment of the methods provided above, the method increases the time of stability for the composition by at least 50%.

In a specific embodiment of the methods provided above, the method increases the time of stability for the composition by at least 100%.

In a specific embodiment of the methods provided above, the method increases the time of stability for the composition by at least 200%.

IV. Examples Example 1

To determine the role pH and salt concentration have on a plasma-derived 20% IgG composition, a two year stability study was conducted. This study revealed that the inclusion of sodium chloride and/or the formulation at neutral to mildly acid pH imparted a stabilizing effect on the 20% IgG composition.

Briefly, two IgG compositions prepared from pooled plasma according to the Gammagard SD process outlined in Teschner et al. (Vox Sang. 2007 January; 92(1):42-55) were concentrated to a final protein concentration of 20%. These preparations were then divided into several samples which were differentially formulated at pHs 6.5, 7.0, or 7.5 with and without 50 mM sodium chloride. The aqueous formulations were then stored at between 28° C. and 32° C. for 24 months. After the two year incubation period, the molecular size distributions of the IgG in the various formulations were investigated by high performance size exclusion chromatography (HP-SEC), the results of which are provided in Table 5.

TABLE 5 Molecular size distribution of 20% IgG (IGSC61 and IGSC63) formulations after two years storage at 28° C. to 32° C. T Poly- Oligo/ Sample (° C.) mers Dimers Monomers Fragments IGSC61 pH 6.5 24 months 28-32 3.57 20.68 68.92 6.83 pH 7.0 24 months 28-32 10.32 19.78 60.96 8.94 pH 7.5 24 months 28-32 11.76 20.01 57.70 10.53 pH 6.5 3 g/L 24 months 28-32 3.16 20.52 69.75 6.57 NaCl pH 7.0 3 g/L 24 months 28-32 9.53 19.68 61.69 9.11 NaCl pH 7.5 3 g/L 24 months 28-32 10.26 20.25 58.97 10.53 NaCl IGSC63 pH 6.5 24 months 28-32 2.34 20.99 68.84 7.83 pH 7.0 24 months 28-32 3.48 20.95 64.37 11.20 pH 7.5 24 months 28-32 6.00 23.23 58.39 12.39 pH 6.5 3 g/L 24 months 28-32 1.92 19.33 70.36 8.40 NaCl pH 7.0 3 g/L 24 months 28-32 2.63 21.10 64.98 11.29 NaCl pH 7.5 3 g/L 24 months 28-32 5.51 23.13 59.24 12.12 NaCl

The results shown in Table 5 indicate that, within the pH range of 6.5 to 7.5, increases in pH result in increased aggregation of the IgG preparation, as shown by the increasing percentage of IgG polymers in the formulations at pH 7.0 and 7.5 compared to the formulations at pH 6.5. Inclusion of 50 mM sodium chloride in each formulation stabilizes the lower molecular weight IgG species, resulting in a nearly 14% reduction in the level of IgG polymers in the samples. This is in contrast to IgG compositions formulated as low pH (4.4 to 4.9), which are destabilized by the addition of sodium chloride.

Example 2

To further characterize the stabilizing effect that sodium chloride has on IgG compositions formulated at mildly acid to neutral pH, an accelerated stability study was performed. For the accelerated study, elevated temperatures (38° C. to 42° C.) were used to simulate longer time periods at room temperature (20° C. to 25° C.). Briefly, a 20% IgG composition, prepared as in Example 1, was divided into samples that were formulated with increasing salt concentrations (0 mM, 50 mM, 100 mM, and 150 mM) at mildly acid to neutral pHs (pH 5.5, 6.0, 6.5, 7.0, and 7.5). The aqueous formulations were then stored at between 38° C. and 42° C. for 6 months. After the 6 month incubation period, the molecular size distributions of the IgG in the various formulations were investigated by high performance size exclusion chromatography (HP-SEC). The percentage of IgG aggregates present in the various formulations is shown in FIG. 1.

As seen in FIG. 1, the stability of the immunoglobulin preparation is dependent upon both the pH and the salt concentration of the formulation. Addition of 100 mM or 150 mM sodium chloride in the formulation provided additional stability, as compared to the protective effects seen for 50 mM sodium chloride in Example 1, at pHs between 6.0 and 7.5. Significantly, inclusion of 150 mM sodium chloride in of IgG compositions formulates at pHs between 6.0 and 7.5 reduced IgG aggregation by more than 50% on average. Optimal stabilization was seen in formulations containing 150 mM sodium chloride at pH 6.5, in which aggregate formation was reduced more than 50% as compared to formulations with 50 mM sodium chloride and about 60% as compared to formulations with no sodium chloride. Consistent with previous observations, the addition of sodium chloride in IgG formulations at lower pH (5.5) results in a destabilizing effect.

Example 3

To evaluate the stabilizing effects of sodium chloride on other immunoglobulin preparations formulated at mildly acidic to neutral pH, preparations of Partobulin® NG (Baxter Biosciences) and Tetabulin® NG (Baxter Biosciences) were formulated accordingly and tested for stability, via aggregation formation, and activity, via anti-antigen potency, over a 6 month time frame. Partobulin® is a plasma-derived human anti-D antigen immunoglobulin preparation used for antenatal anti-D prophylaxis in Rh(D) negative pregnant women carrying Rh(D) positive fetuses, as well as for the treatment of Rh(D) negative persons after incompatible transfusions of Rh(D) positive blood or erythrocyte concentrate. Tetabulin® is a plasma-derived human tetanus immunoglobulin used for post-tetanus exposure prophylaxis and therapy of clinically manifest tetanus. Both Partobulin® and Tetabulin® are typically formulated at between 100 g/L and 170 g/L human protein (of which at least 90% are immunoglobulin G) for intramuscular administration.

Briefly, feasibility lots of Partobulin® NG and Tetabulin® NG were stored at low pH for one year at 2° C. to 8° C. prior to the addition of sodium chloride to 150 mM and adjustment of the pH to between 5.5 and 7.5. Prior to the formulation with sodium chloride and pH adjustment, the bulk was formulated at pH 4.7 in the presence of 250 mM glycine like for GGL/KIOVIG. The stability of the formulations at 28° C. to 32° C. was then tracked for 6 months by periodically monitoring the Tetanus anti-toxin or Anti-D titers as well as the molecular size distributions of the formulations.

Aggregate formation over the course of 6 months for Tetabulin® NG and Partobulin® NG are shown in FIGS. 2 and 3, respectively. Strikingly, no additional immunoglobulin aggregation occurred over the entire six months for either preparation formulated at pH 5.5 or 6.0 with 150 mM NaCl. Similarly, only slight aggregation was seen beginning at six months for both compositions at pH 6.5. In comparison, significant aggregation occurred within three months of storage at 28° C. to 32° C. for both Tetabulin® NG and Partobulin® NG formulated at pH 7.0 and 7.5. After six months of storage, both the Tetabulin® NG and Partobulin® NG formulations at pH 7.0 contained twice the starting amount of aggregates and both formulations at pH 7.5 contained about three times more immunoglobulin aggregates than at the start of the storage period.

Despite the increased aggregation seen in the Tetabulin® NG formulations at pH 7.0 and 7.5, no significant loss in Tetanus anti-toxin titer was found over the course of the six month storage period (FIG. 4). Conversely, a 25% to 40% drop in anti-D titer was observed over the six month storage period over the entire pH range (FIG. 5). However, the loss of anti-D titer was reduced at pH 5.5 and 6.0.

Example 4

To determine if the stabilizing effect of sodium chloride at mildly acidic to neutral pH was specific to plasma-derived immunoglobulin preparations or broadly applicable to all immunoglobulin products, the stability of a recombinant anti-MIF antibody was determined in an accelerated stability assay. Briefly, samples of bulk recombinant anti-MIF antibody at 120 g/L were formulated with 150 mM sodium chloride and 0.25 M glycine at pH 4.5, 5.6, 6.5, and 7.3 and stored at between 38° C. and 42° C. for three months.

The molecular size distributions of the IgG in the various formulations were determined by high performance size exclusion chromatography (HPLC-SEC) at the start of the storage period, after two weeks, 1 month, and 3 months of storage. The results of these characterizations are provided in Table 6.

TABLE 6 Molecular size distribution in Anti-MIF formulations during storage at 38° C. to 42° C. Oligo-/ Formulation Sample T ° C. Aggregates Dimers Monomers Fragments No NaCl pH Bulk 2-8 <0.1% 3.7% 96.1% 0.2% 5.99 150 mM NaCl pH 4.5 1F Start 2-8 <0.1% 4.1% 95.6% 0.2% pH 5.6 2F Start 2-8 <0.1% 3.8% 96.1% 0.1% pH 6.5 3F Start 2-8 <0.1% 3.8% 96.0% 0.2% pH 7.3 4F Start 2-8 <0.1% 3.8% 96.0% 0.2% 150 mM NaCl pH 4.5 1F 2 Weeks 38-42 0.3% 5.4% 91.4% 3.0% pH 5.6 2F 2 Weeks 38-42 0.1% 5.5% 93.6% 0.9% pH 6.5 3F 2 Weeks 38-42 0.3% 6.2% 93.0% 0.5% pH 7.3 4F 2 Weeks 38-42 0.4% 7.7% 91.3% 0.6% 150 mM NaCl pH 4.5 1F 1 Month 38-42 0.1% 5.4% 90.0% 4.5% pH 5.6 2F 1 Month 38-42 0.1% 7.0% 91.3% 1.7% pH 6.5 3F 1 Month 38-42 0.4% 8.4% 90.2% 1.0% pH 7.3 4F 1 Month 38-42 0.7% 12.2% 85.9% 1.2% 150 mM NaCl pH 4.5 1F 3 Months 38-42 0.3% 7.9% 83.7% 8.0% pH 5.6 2F 3 Months 38-42 0.3% 13.4% 84.1% 2.2% pH 6.5 3F 3 Months 38-42 1.5% 14.6% 81.7% 2.2% pH 7.3 4F 3 Months 38-42 4.3% 24.0% 69.6% 2.0%

As seen in Table 6, fragmentation of the recombinant anti-MIF antibody is most pronounced at acidic pH (pH 4.5), even after only 2 weeks storage. Likewise, as seen for the concentrated IgG preparation in Example 1 and Tetabulin® and Partobulin® formulations in Example 3, sodium chloride reduced aggregation of the recombinant anti-MIF antibodies at mildly acid conditions (pH 5.6 and 6.5), while aggregation occurred more quickly at neutral pH (pH 7.5).

To further characterize the stabilizing effects of sodium chloride in these formulations, anti-MIF titers were determined by ELISA for each formulation at the start of the storage period, and after 2 weeks, 1 month, and 3 months of storage at elevated temperatures (38° C. to 42° C.). As seen in FIG. 6, despite low aggregation rates for the recombinant anti-MIF antibodies formulated at acid pH (pH 4.5), the Anti-MIF titer of this formulation dropped by 40% to 50% over the three month storage. Similarly, the Anti-MIF titer of the antibody preparation formulated at neutral pH (pH 7.3) was reduced by at least 20% over three months. However, the antibody preparations formulated at mildly acidic pH (pH 5.6 and 6.5) lost only about 10% of their starting Anti-MIF titer over the three month storage at elevated temperatures.

Example 5

Factor VIII, a coagulation factor, is a highly labile therapeutic protein that is commonly formulated as a lyophilized powder that must be reconstituted prior to administration. In order to determine if the addition of sodium chloride can also stabilize non-immunoglobulin labile protein formulations at mildly acidic to neutral pH, stability tests were performed on recombinant Factor VIII formulations. Briefly, frozen recombinant Factor VIII was slowly thawed and formulated by buffer exchange according to the reconstituted formulation for ADVATE (recombinant FVIII; Baxter Healthcare Corporation, Westlake Village, Calif.), given in Table 7. Samples of the FVIII preparation were then aliquoted and the pH adjusted to pH 5.5, 6.0, 6.5, 7.0, 7.5, or 8.0 with and without addition of 150 mM sodium chloride to give formulations with a starting Factor VIII activity in solution of 500 IU/mL. The samples were stored at 2° C. to 8° C. for 12 weeks, during which time the residual FVIII activity was periodically monitored.

In the absence of sodium chloride in the formulation, a continuous drop in FVIII activity was seen for all formulation (FIG. 7), with the most pronounced loss of activity seen at the extremes of the investigated pH range. Strikingly, all of the rFVIII samples except for the formulation at pH 6.5 lost at least 20% of their starting activity within four weeks. Furthermore, most of the samples lost at least 40% of their starting FVIII activity within 9 weeks and at least 60% of their FVIII activity within 12 weeks. The notable exception to this was the sample formulated at pH 6.0, which appears to be a statistical outlier. It was also noticed that all of the formulations lacking sodium chloride became turbid within two weeks of storage at 2° C. to 8° C.

Conversely, addition of sodium chloride in the rFVIII formulations resulted in a marked increase in the stability of FVIII activity at pH above 6.0 (FIG. 8). Surprisingly, at pH 6.5 and 7.0, the addition of 150 mM sodium chloride leads to a aqueous formulation in which FVIII activity is stable at 2° C. to 8° C. for at least 12 weeks. At the lower end of the investigated pH range (pH 5.5) addition of sodium chloride showed no stabilizing effect of the Factor VIII activity. Notably, the FVIII formulations at pH 6.5 and 7.0 remained clear for at least three weeks, while all other formulations became turbid in this same time period. Notably, these samples showed only slight turbidity over the entire course of the storage period.

TABLE 7 Reconstituted formulation for ADVATE ®. ADVATE ® Formulation 38 g/L mannitol 10 g/L trehalose 108 mEq/L sodium 12 mM histidine 12 mM Tris 1.9 mM calcium 0.15 g/L polysorbate-80 0.10 g/L glutathione

Example 6

The effect sodium chloride has on the stability of an IgG composition formulated at acidic pH (4.4 to 4.9) was investigated. Briefly, an IgG composition was formulated with 0.25 M glycine, pH 4.4 to 4.9, at a final protein concentration of 10%. Sodium chloride was then added to aliquots of the formulation to a final concentration of 0 mM, 10 mM, 50 mM, or 100 mM. The molecular size distribution was then monitored by HPLC analysis after 1, 3, 6, 9, 12, 19, and 24 months of storage under refrigerated conditions (between 2° C. and 8° C.). The fixation of complement in the samples was also measured to determine the tolerability of the formulation. The results of the analysis are given in Table 8.

As can be seen in Table 8, Storage of the 10% IgG preparation at between 2° C. and 8° C., with different amounts of sodium chloride added, revealed that the presence of sodium chloride seriously impacts the ACA value and therefore the tolerability of the product, as well as the oligo/dimmer content of the samples. Compared to the final container without sodium chloride, which meets the required specifications for the product over the entire storage time, a content of 10 mM sodium chloride is sufficient to reduce the storage period to 19 months, a content of 50 mM to 12 months and a concentration of 100 mM to 9 months.

TABLE 8 Influence of sodium chloride content on the stability of a 10% IgG preparation stored under refrigeration. HPLC ACA-Titer Aggregates Oligo/Dimers Monomers Fragments CH50 U/cons. No NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.41 6.83 92.76 0.00 20.3% after incubation 0.43 7.52 91.92 0.15 36.2%  1 month +2 to +8° C. 0.22 7.28 92.28 0.22 41.2%  3 months +2 to +8° C. 0.55 11.88 87.37 0.19 31.6%  6 months +2 to +8° C. 0.38 9.19 90.08 0.35 44.0%  9 months +2 to +8° C. 0.30 7.55 91.98 0.18 33.7% 12 months +2 to +8° C. 0.62 7.50 91.54 0.34 42.5% 19 months +2 to +8° C. 0.74 12.43 86.55 0.28 46.2% 24 months +2 to +8° C. 0.59 8.76 90.53 0.11 46.9% 10 mM NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.32 7.13 92.55 0.00 25.7% after incubation 0.35 8.08 91.39 0.17 47.7%  1 month +2 to +8° C. 0.25 9.79 89.76 0.21 46.8%  3 months +2 to +8° C. 0.53 12.52 86.72 0.23 35.0%  6 months +2 to +8° C. 0.44 11.13 88.26 0.18 42.9%  9 months +2 to +8° C. 0.36 8.22 91.12 0.29 40.6% 12 months +2 to +8° C. 0.63 10.57 88.65 0.15 48.0% 19 months +2 to +8° C. 0.86 13.32 85.59 0.23 49.0% 24 months +2 to +8° C. 0.88 9.15 89.96 0.00 50.5% 50 mM NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.25 7.46 92.29 0.00 38.2% after incubation 0.30 9.30 90.19 0.21 57.0%  1 month +2 to +8° C. 0.29 10.47 89.02 0.22 58.3%  3 months +2 to +8° C. 0.58 13.61 85.57 0.23 35.8%  6 months +2 to +8° C. 0.49 11.82 87.39 0.33 47.7%  9 months +2 to +8° C. 0.46 11.21 88.07 0.26 43.6% 12 months +2 to +8° C. 0.58 11.36 87.86 0.20 45.9% 19 months +2 to +8° C. 0.89 14.36 84.58 0.17 58.7% 24 months +2 to +8° C. 0.80 11.16 88.04 0.00 67.9% 100 mM NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.24 7.41 92.34 0.00 43.9% after incubation 0.29 9.72 89.88 0.11 59.7%  1 month +2 to +8° C. 0.31 10.92 88.63 0.14 54.5%  3 months +2 to +8° C. 0.64 13.40 85.81 0.15 45.9%  6 months +2 to +8° C. 0.50 11.96 87.31 0.24 41.1%  9 months +2 to +8° C. 0.46 11.15 88.17 0.22 48.2% 12 months +2 to +8° C. 0.59 11.88 87.36 0.17 51.8% 19 months +2 to +8° C. 0.91 14.26 84.49 0.35 68.4% 24 months +2 to +8° C. 0.80 11.30 87.89 0.00 73.8%

To confirm the negative effect sodium chloride has on the stability of immunoglobulin compositions formulated at acidic pH, the experiment was repeated with storage at room temperature (28° C. to 30° C.). As can be seen in Table 9, the results of the second experiment confirm the data obtained for storage between 2° C. and 8° C. The ACA values are generally lower, while the fragmentation and aggregation is accelerated. The negative effect of the addition of sodium chloride at acidic pH is confirmed with this experiment.

TABLE 9 Influence of sodium chloride content on the stability of a 10% IgG preparation stored at room temperature. HPLC ACA-Titer Aggregates Oligo/Dimers Monomers Fragments C′H50 U/cons. No NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.41 6.83 92.76 0.00 20.3% after incubation 0.43 7.52 91.92 0.15 36.2%  1 month +28 to +30° C. 0.24 6.68 92.8 0.27 37.5%  3 months +28 to +30° C. 0.65 9.92 88.92 0.50 29.8%  6 months +28 to +30° C. 0.48 8.06 90.39 1.08 37.8%  9 months +28 to +30° C. 0.59 8.07 90.15 1.19 28.7% 12 months +28 to +30° C. 1.18 8.59 88.69 1.54 32.4% 19 months +28 to +30° C. 1.83 12.63 83.28 2.26 35.9% 24 months +28 to +30° C. 2.32 11.41 83.98 2.29 36.2% 10 mM NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.32 7.13 92.55 0.00 25.7% after incubation 0.35 8.08 91.39 0.17 47.7%  1 month +28 to +30° C. 0.24 7.59 91.81 0.37 42.5%  3 months +28 to +30° C. 0.56 10.50 88.47 0.46 34.2%  6 months +28 to +30° C. 0.48 9.33 89.16 1.03 37.8%  9 months +28 to +30° C. 0.61 9.01 89.33 1.05 39.3% 12 months +28 to +30° C. 0.98 10.47 87.04 1.51 39.7% 19 months +28 to +30° C. 1.86 13.06 83.11 1.98 44.4% 24 months +28 to +30° C. 2.43 12.78 82.40 2.39 37.8% 50 mM NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.25 7.46 92.29 0.00 38.2% after incubation 0.30 9.30 90.19 0.21 57.0%  1 month +28 to +30° C. 0.27 9.98 89.50 0.26 45.1%  3 months +28 to +30° C. 0.48 11.38 87.63 0.51 36.1%  6 months +28 to +30° C. 0.61 10.54 87.98 0.87 44.0%  9 months +28 to +30° C. 0.78 97.4 88.32 1.17 46.7% 12 months +28 to +30° C. 1.12 11.39 86.15 1.34 51.8% 19 months +28 to +30° C. 1.93 13.20 82.33 2.53 58.3% 24 months +28 to +30° C. 2.79 12.94 81.78 2.49 50.5% 1000 mM NaCl. 10% Protein. pH 4.7/0.25M Glycine prior incubation 0.24 7.41 92.34 0.00 43.9% after incubation 0.29 9.72 89.88 0.11 59.7%  1 month +28 to +30° C. 0.31 10.45 88.86 0.38 50.0%  3 months +28 to +30° C. 0.52 11.47 87.51 0.50 35.0%  6 months +28 to +30° C. 0.64 10.73 87.66 0.96 47.2%  9 months +28 to +30° C. 0.77 9.54 88.54 1.15 56.1% 12 months +28 to +30° C. 1.10 11.22 88.26 1.43 58.9% 19 months +28 to +30° C. 1.71 12.79 83.02 2.47 64.8% 24 months +28 to +30° C. 2.30 12.63 82.48 2.60 60.8%

It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes.

Claims

1. A storage stable, aqueous composition of a labile therapeutic protein comprising:

(a) a labile therapeutic protein;
(b) from 75 mM to 200 mM of an alkali metal chloride salt;
(c) an amino acid; and
(d) a pH of from 5.5 to 7.5.

2. The composition of claim 1, comprising from 100 mM to 200 mM of an alkali metal chloride salt.

3. The composition of claim 2, comprising from 125 mM to 175 mM of an alkali metal chloride salt.

4. The composition of claim 3, comprising 150±15 mM an alkali metal chloride salt.

5. The composition of claim 1, wherein the alkali metal chloride salt is sodium chloride.

6. The composition of claim 1, therein the alkali metal chloride salt is potassium chloride.

7. The composition of claim 1, wherein the amino acid is selected from the group consisting of glycine, proline, and histidine.

8. The composition of claim 7, wherein the amino acid is glycine.

9. The composition of claim 7, wherein the amino acid is proline.

10. The composition of claim 7, wherein the amino acid is histidine.

11. The composition of claim 1, wherein the concentration of the amino acid is from 50 mM to 500 mM.

12. The composition of claim 11, wherein the concentration of the amino acid is from 100 mM to 400 mM.

13. The composition of claim 11, wherein the concentration of the amino acid is from 150 mM to 350 mM.

14. The composition of claim 11, wherein the concentration of the amino acid is from 200 mM to 300 mM.

15. The composition of claim 11, wherein the concentration of the amino acid is from 225 mM to 275 mM.

16. The composition of claim 11, wherein the concentration of the amino acid is 250±10 mM.

17. The composition of claim 1, wherein the pH of the composition is from 5.5 to 7.0.

18. The composition of claim 1, wherein the pH of the composition is from 5.5 to 6.5.

19. The composition of claim 1, wherein the pH of the composition is from 5.5 to 6.0.

20. The composition of claim 1, wherein the pH of the composition is from 6.0 to 7.5.

21. The composition of claim 1, wherein the pH of the composition is from 6.0 to 7.0.

22. The composition of claim 1, wherein the pH of the composition is from 6.0 to 6.5.

23. The composition of claim 1, wherein the pH of the composition is from 6.5 to 7.5.

24. The composition of claim 1, wherein the pH of the composition is from 6.5 to 7.0.

25. The composition of claim 1, wherein the pH of the composition is from 7.0 to 7.5.

26. The composition of claim 1, wherein the labile therapeutic protein is a human or humanized protein.

27. The composition of claim 1, wherein the labile therapeutic protein is an immunoglobulin.

28. The composition of claim 27, wherein the immunoglobulin is an IgG immunoglobulin.

29. The composition of claim 27, wherein the immunoglobulin is a polyclonal immunoglobulin.

30. The composition of claim 27, wherein the immunoglobulin is a monoclonal immunoglobulin.

31. The composition of claim 27, wherein the immunoglobulin is a recombinant immunoglobulin.

32. The composition of claim 27, wherein the immunoglobulin is enriched from pooled human plasma.

33. The composition of claim 27, wherein the concentration of the immunoglobulin is 50±5 g/L.

34. The composition of claim 27, wherein the concentration of the immunoglobulin is less than 50 g/L.

35. The composition of claim 27, wherein the concentration of the immunoglobulin is at least 50 g/L.

36. The composition of claim 35, wherein the concentration of the immunoglobulin is from 50 g/L to 150 g/L.

37. The composition of claim 36, wherein the concentration of the immunoglobulin is 100±10 g/L.

38. The composition of claim 35, wherein the concentration of the immunoglobulin is at least 100 g/L.

39. The composition of claim 38, wherein the concentration of the immunoglobulin is 150±15/L.

40. The composition of claim 35, wherein the concentration of the immunoglobulin is from 150 g/L to 250 g/L.

41. The composition of claim 40, wherein the concentration of the immunoglobulin is 200±20 g/L.

42. The composition of claim 35, wherein the concentration of the immunoglobulin is at least 200 g/L.

43. The composition of claim 27, wherein at least 95% of the protein in the composition is immunoglobulin.

44. The composition of claim 43, wherein at least 95% of the protein in the composition is IgG immunoglobulin.

45. The composition of claim 44, wherein at least 98% of the protein in the composition is IgG immunoglobulin.

46. The composition of claim 27, wherein the composition is stable for at least 6 months when stored at between 28° C. and 32° C.

47. The composition of claim 46, wherein the composition is stable for at least 1 year when stored at between 28° C. and 32° C.

48. The composition of claim 46, wherein the composition is stable for at least 2 year when stored at between 28° C. and 32° C.

49. The composition of claim 27, wherein the composition is stable for at least 1 month when stored at between 38° C. and 42° C.

50. The composition of claim 49, wherein the composition is stable for at least 3 months when stored at between 38° C. and 42° C.

51. The composition of claim 49, wherein the composition is stable for at least 6 months when stored at between 38° C. and 42° C.

52. The composition of claim 27, wherein the composition is stable for at least 1 year when stored at between 28° C. and 32° C.

53. The composition of claim 46, wherein the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is between 0% and 5%.

54. The composition of claim 53, wherein the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is between 0% and 2%.

55. The composition of claim 46, wherein the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is from 0% to 5% and the percentage of immunoglobulin in the monomeric state is from 80% to 100%.

56. The composition of claim 55, wherein the composition is considered stable as long as the percentage of immunoglobulin in the aggregated state is from 0% to 2% and the percentage of immunoglobulin in the monomeric state is from 85% to 100%.

57. The composition of claim 1, wherein the labile therapeutic protein is a coagulation factor.

58. The composition of claim 57, wherein the coagulation factor is selected from the group consisting of Factor VII, Factor VIII, Factor IX, a protein K-dependent coagulation complexand, and von Willebrand Factor (vWF).

59. The composition of claim 57, wherein the coagulation factor is Factor VIII.

60. The composition of claim 59, wherein the pH of the composition is between 6.0 and 7.0.

61. The composition of claim 60, wherein the pH of the composition is 6.5±0.2.

62. The composition of claim 27, wherein the composition retains at least 80% of its Factor VIII activity when stored at a temperature between 2° C. and 8° C. for at least 1 month.

63. The composition of claim 1, wherein the labile protein is stable for less than 3 months in an aqueous formulation containing:

(a) from 0 mM to 50 mM of an alkali metal chloride salt;
(b) an amino acid; and
(c) a pH of from 5.5 to 7.

64. The composition of claim 1, wherein the composition is formulated for subcutaneous and/or intramuscular administration.

65. A method for stabilizing an aqueous composition of a labile therapeutic protein, the method comprising formulating the composition at a pH between 5.5 and 7.0, wherein the formulated composition comprises:

(a) a labile therapeutic protein;
(b) from 75 mM to 200 mM of an alkali metal chloride salt; and
(c) an amino acid.
Patent History
Publication number: 20120076779
Type: Application
Filed: Sep 16, 2011
Publication Date: Mar 29, 2012
Applicants: Baxter Healthcare S.A. (Glattpark (Opfikon)), Baxter International Inc. (Deerfield, IL)
Inventors: Harald Arno Butterweck (Vienna), Theresa Friederike Bauer (Vienna), Lucia Hofbauer (Eggenburg), Wolfgang Teschner (Vienna), Oliver Zoechling (Vienna), Hans-Peter Schwarz (Vienna), Christa Mayer (Wolfsthal), Meinhard Hasslacher (Vienna)
Application Number: 13/235,307