WO2023089503A1 - Inter-alpha inhibitor protein formulations - Google Patents

Inter-alpha inhibitor protein formulations Download PDF

Info

Publication number
WO2023089503A1
WO2023089503A1 PCT/IB2022/061044 IB2022061044W WO2023089503A1 WO 2023089503 A1 WO2023089503 A1 WO 2023089503A1 IB 2022061044 W IB2022061044 W IB 2022061044W WO 2023089503 A1 WO2023089503 A1 WO 2023089503A1
Authority
WO
WIPO (PCT)
Prior art keywords
formulation
iαi
pαi
buffer
exemplary embodiment
Prior art date
Application number
PCT/IB2022/061044
Other languages
French (fr)
Inventor
Joris HÖFINGHOFF
Euripedes DE ALMEIDA RIBEIRO JR.
Sandra PICHLER
Original Assignee
Takeda Pharmaceutical Company Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Takeda Pharmaceutical Company Limited filed Critical Takeda Pharmaceutical Company Limited
Publication of WO2023089503A1 publication Critical patent/WO2023089503A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • C07K14/8114Kunitz type inhibitors

Definitions

  • Inter-alpha inhibitor proteins are a family of structurally related proteins found in plasma that are involved in inflammatory regulation and wound healing.
  • I ⁇ Ips Inter-alpha Inhibitor (I ⁇ I), which consists of two heavy chains (H1 & H2) and a single light chain (e.g., bikunin), and Pre-alpha Inhibitor (P ⁇ I) consisting of one heavy (H3) and one light chain (e.g., bikunin).
  • I ⁇ Ips are reduced during inflammatory processes such as sepsis and stroke.
  • Prior studies indicate that I ⁇ Ip levels are inversely correlated with morbidity and mortality in severe inflammatory conditions and that patients whose I ⁇ Ip levels had recovered over time exhibited improved outcomes. Also, replenishing with exogenous I ⁇ Ip in experimental models of severe inflammation provided recovery in multiple animal studies across species and indication areas.
  • the invention provides a liquid formulation comprising Inter-alpha Inhibitor Proteins (I ⁇ Ips) and a buffer which is a citrate buffer and/or a histidine buffer, and wherein the formulation has a pH of about 5.0 to about 7.5.
  • I ⁇ Ip Inter-alpha Inhibitor Proteins
  • the I ⁇ Ip is a human I ⁇ Ip.
  • the invention provides a method of purifying an I ⁇ I and a P ⁇ I from a biological material by anion-exchange chromatography, wherein a wash buffer and/or an elution buffer used in the anion exchange chromatography step has a low conductivity.
  • the invention also provides a formulation comprising an I ⁇ I and a P ⁇ I, wherein the I ⁇ I and the P ⁇ I are purified by the method above.
  • the formulation has beneficial features, comprising that (i) less than 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa, (ii) more than 80% of the I ⁇ I and the P ⁇ I does not exist as a protein aggregate, (iii) the formulation has at least 20% higher trypsin inhibitory specific activity than known formulations comprising I ⁇ Ips, (iv) the formulation comprises less than 0.03 mg/ml of Factor II (Prothrombin), (v) the formulation comprising less than 0.01 mg/ml of Factor X, and/or (vi) the formulation comprising less than 0.001 mg/ml of C4 binding protein.
  • beneficial features comprising that (i) less than 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa, (ii) more than 80% of the I ⁇ I and the P ⁇ I does not exist as a protein aggregate, (iii) the formulation has at least 20%
  • administering means a method of giving a dosage of a formulation (e.g., an I ⁇ Ip) to a subject.
  • the formulations utilized in the methods described herein can be administered, for example, orally, intramuscularly, intravenously, intradermally, percutaneously, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, topically, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, by catheter, by lavage, in creams, or in lipid formulations.
  • biological material refers to a sample from a subject (e.g., a mammal, such as a human) that contains I ⁇ Ip.
  • the biological material examples include a blood product material, e.g., whole plasma, cryo-poor plasma, liquid plasma, fresh frozen plasma (FFP), FFP24, frozen plasma (FP), FP24, thawed FFP, thawed FFP24, thawed FP, thawed FP24, source plasma, recovered plasma, solvent/detergent-treated plasma (SDP), platelet-rich plasma (PRP), platelet-poor plasma (PPP), serum, blood, and a diluted or concentrated preparation thereof, milk or colostrum, urine, sputum, and cerebrospinal fluid.
  • a blood product material e.g., whole plasma, cryo-poor plasma, liquid plasma, fresh frozen plasma (FFP), FFP24, frozen plasma (FP), FP24, thawed FFP, thawed FFP24, thawed FP24, source plasma, recovered plasma, solvent/detergent-treated plasma (SDP), platelet-rich plasma (PR
  • the biological material can be from a human, primate, bovine, equine, porcine, ovine, feline, canine, or combinations thereof.
  • the biological material may also be an extract prepared using cells that express I ⁇ Ip, or may be or contain cells that secrete I ⁇ Ip, e.g., cells that have been recombinantly modified to express I ⁇ Ip.
  • chromatography refers to a separation of one or more analytes in a mixture by passing the mixture in a solution or in a suspension through a medium in which the analytes of the mixture move at different rates.
  • a chromatography step can be size exclusion chromatography, ion-exchange chromatography, affinity chromatography, or dye-ligand chromatography. More specifically, a chromatography step, as described herein, can be performed to separate an I ⁇ Ip from a biological material containing the I ⁇ Ip.
  • the term “eluate” refers to a fraction containing an analyte material (e.g., I ⁇ Ip) that is eluted from a medium (e.g., a support material) during a purification step (e.g., a chromatography step).
  • An eluate may be released from the medium by applying an eluent to the medium, thereby releasing the analyte.
  • an eluate can refer to a fraction containing I ⁇ Ip that has been released from a medium following application of an eluent (e.g., an elution buffer, such as a buffer containing a salt) to the medium.
  • an eluent e.g., an elution buffer, such as a buffer containing a salt
  • the term “flow through” refers to a fraction of material or a volume of fluid that passes through a medium (e.g., a medium used for chromatography, such as an anion- exchange resin) without binding.
  • a medium e.g., a medium used for chromatography, such as an anion- exchange resin
  • Additional mobile phase e.g., a fluid, such as buffer with a low (e.g., less than 50 mM salt) or no salt (e.g., sodium chloride)
  • a fluid such as buffer with a low (e.g., less than 50 mM salt) or no salt (e.g., sodium chloride)
  • an analyte e.g., I ⁇ Ip
  • inter-alpha inhibitor protein and “I ⁇ Ip,” and plural forms thereof, refer to multi-component glycoproteins in a family of structurally related serine protease inhibitors.
  • I ⁇ Ips have been shown to be important in the inhibition of an array of proteases including neutrophil elastase, plasmin, trypsin, chymotrypsin, Granzyme K, preprotein convertase, furin, cathepsin G, and acrosin.
  • I ⁇ Ips are found at relatively high concentrations (400-800 mg/L).
  • this family of inhibitors typically includes a combination of polypeptide chains (light and heavy chains) covalently linked by a chondroitin sulfate chain.
  • the heavy chains of I ⁇ Ips (H1, H2, and H3) are also called hyaluronic acid (HA) binding proteins.
  • the major forms of I ⁇ Ips found in human plasma are inter-alpha-inhibitor (I ⁇ I), which contains two heavy chains (H1 and H2) and a single light chain (L), and pre-alpha-inhibitor (P ⁇ I), which contains one heavy (H3) and one light chain (L).
  • I ⁇ Ip is the light chain (also termed bikunin (bi-kunitz inhibitor) with two Kunitz domains), which is known to broadly inhibit plasma serine proteases.
  • Another I ⁇ Ip is the heavy chain-related molecule H4, which circulates in the blood without linkage to bikunin.
  • Yet another I ⁇ Ip is the heavy chain-related molecule H5.
  • I ⁇ I and P ⁇ I present in the plasma fraction have an apparent molecular weight of between about 60 kDa to about 280 kDa.
  • pharmaceutically acceptable excipient means one or more compatible solid or liquid fillers, diluents, or encapsulating substances that are suitable for administration into a human.
  • the excipient can contain an additive, such as a substance that enhances isotonicity and/or chemical stability.
  • an additive such as a substance that enhances isotonicity and/or chemical stability.
  • Such materials are non-toxic to recipients in the amounts and concentrations employed, and can include buffers, such as phosphate, citrate, succinate, acetate, lactate, tartrate, and other organic acids or their salts; tris- hydroxymethylaminomethane (Tris), bicarbonate, carbonate, and other organic bases and their salts; antioxidants, such as ascorbic acid; low molecular weight (for example, less than about ten residues) polypeptides, e.g., polyarginine, polylysine, polyglutamate and polyaspartate; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers, such as polyvinylpyrrolidone (PVP), polypropylene glycols (PPGs), and polyethylene glycols (
  • the terms "prevent,” “preventing,” “prevention,” “prophylactic treatment” and the like refer to reducing the probability of developing a disorder or condition in a subject, who does not have, but is at risk of or is susceptible to developing, a disease, disorder, or condition.
  • processing refers to a biological material that has been modified using one or more sample preparation steps (e.g., filtration, centrifugation, sedimentation, chromatography, etc.) prior to contacting the material to an anion-exchange resin.
  • sample preparation steps e.g., filtration, centrifugation, sedimentation, chromatography, etc.
  • Other examples of processing include a decanting step, a clarification step, a freezing step, a drying step, an evaporation step, an extraction step, a filtration step, a precipitation step, or another purification or preparatory method known in the art.
  • the processing step may remove up to, e.g., 10% or more (w/w) (e.g., 10-30% (w/w), such as 15%, 20%, 25%, or 30% (w/w) or more) of one or more substances from the biological material (e.g., a protein other than an I ⁇ Ip).
  • the terms “purify,” “purifying,” “purification” and the like refer to one or more steps or processes of removing proteins (e.g., proteins other than an I ⁇ Ip) and/or non- proteinaceous substances (e.g., phospholipids and nucleic acids) from a heterologous mixture (e.g., a biological material, such as blood or milk) containing I ⁇ Ip and the other proteins and/or substances to produce a formulation containing an I ⁇ Ip without the other proteins and/or substances present in the original mixture (e.g., a biological material) or in which the proteins other than I ⁇ Ip and/or substances have been reduced by 40% or more by weight (e.g., 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97%, or 99% or more) relative to, e.g., a starting mixture (e.g., a biological material).
  • proteins e.g., proteins other than an I ⁇ Ip
  • non- proteinaceous substances e.g.,
  • proteins that can be removed from a mixture containing an I ⁇ Ip include, but are not limited to, alpha-1 antitrypsin, C1-inhibitor, albumin, a globulin (including immunoglobulins, e.g., IgA, IgG (e.g., of intravenous Ig (IVIg), anti-D IgG, hepatitis B IgG, measles IgG, rabies IgG, tetanus IgG, and Varicella Zoster IgG), IgM, IgD, and IgE), fibrinogen (factor I), prothrombin (factor II), thrombin, anti-thrombin III, factor III, factor V, factor VII, factor VIII, factor IX, factor X, factor XI, factor XII, factor XIII, fibronectin, alpha-2 antiplasmin, urokinase, protein C, protein S, protein Z, protein Z-related protease,
  • pure or “purity” refers to the extent to which an analyte has been isolated and is free of other components.
  • purity of an isolated protein can be expressed with regard to the protein that is free of any contaminants (e.g., one or more unrelated proteins or other substances).
  • purity of an I ⁇ Ip formulation indicates how much of the formulation is I ⁇ Ip by total weight of the isolated material, which may be determined using, e.g., pure I ⁇ Ip as a reference.
  • a level of purity found in the disclosure can be 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, greater than 95%, or greater than 99% (w/w).
  • a “pure” I ⁇ Ip formulation of the disclosure can be greater than 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or up to 70% pure by weight.
  • a “substantially pure” I ⁇ Ip formulation can be substantially free of contaminants or impurities, e.g., greater than 70%, 75%, 80%, 85%, 90%, 95%, or >99% purity by weight.
  • the level of contaminants or impurities is no more than about 20%, 15%, 12%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% by weight.
  • Purity can be determined by detecting a level of a specific analyte (e.g., I ⁇ Ip) using an immunoassay or other technique (e.g., MAb 69.26 – heparin-biotin sandwich ELISA, SDS/PAGE, and/or Western blot) and calculating a percentage of the analyte (w/w) relative to the total protein content (e.g., as determined by a total protein assay (e.g., bicinchoninic acid assay (BCA), Bradford assay, Biuret test, or another assay known in the art)).
  • a specific analyte e.g., I ⁇ Ip
  • an immunoassay or other technique e.g., MAb 69.26
  • the term “subject” refers to a mammal, including, but not limited to, a human or non-human mammal, such as a primate, bovine, equine, porcine, ovine, feline, or canine.
  • the subject may be a patient.
  • substantially unprocessed refers to a biological material that has been minimally modified, if at all, relative to the original source material (e.g., blood).
  • a substantially unprocessed biological material can retain the original content (e.g., the same proteins and/or substances and/or the same ratio of two or more proteins or substances) and/or the original characteristics (e.g., one or more biological activities) of the original source material.
  • a biological material can be substantially unprocessed, such that any prior purification step(s) performed with the material removes less than 10% (w/w) (e.g., less than 0.1%-10% (w/w), such as less than 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, or 9% (w/w)) of one or more proteins or substances from the material.
  • a substantially unprocessed biological material may be one that has not been modified using a sample preparation step (e.g., filtration, centrifugation, sedimentation, chromatography, etc.), in particular, for example, prior to contacting the biological material to an anion-exchange resin.
  • a sample preparation step e.g., filtration, centrifugation, sedimentation, chromatography, etc.
  • the phrase “specifically binds” refers to a binding reaction between an analyte (e.g., a protein, such as an I ⁇ Ip) and a binding agent.
  • a specific binding reaction is one that occurs between an analyte and a binding agent even in the presence of a heterogeneous population of proteins and other biological molecules (e.g., proteins other than I ⁇ Ip in a biological material).
  • Specific binding between an analyte (e.g., an I ⁇ Ip) and a binding agent can be characterized by a Kd of less than about 1000 nM (e.g., between 1 pM and 1000 nM).
  • An analyte (e.g., an I ⁇ Ip) that does not specifically bind to a binding agent can be characterized by a Kd of greater than about 1000 nM (e.g., greater than 1 ⁇ M, 100 ⁇ M, 500 ⁇ M, or 1 mM).
  • the term “support” means any apparatus that contains an agent (e.g., an anion-exchange resin) that can be contacted with a material (e.g., a biological material) containing at least one analyte (e.g., an I ⁇ Ip).
  • a support may be a column, a membrane, a disc, a chip, or other apparatus for chromatography or affinity capture, examples of which are known in the art and described herein.
  • the term “treating” refers to reducing or ameliorating a disorder and/or one or more symptoms associated therewith. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder or symptoms associated therewith be completely eliminated.
  • yield refers to the relative amount of an analyte (e.g., I ⁇ Ip) obtained after a purification step or process as compared to the amount of analyte in the starting material (e.g., the biological material) (w/w). The yield may be expressed as a percentage.
  • the amount of analyte (e.g., I ⁇ Ip) in the starting material and analyte obtained after the purification step can be measured using an immunoassay or assay (e.g., an anti-I ⁇ Ip antibody (e.g., MAb 69.26) – heparin-biotin sandwich ELISA, SDS/PAGE, and/or Western blot).
  • an immunoassay or assay e.g., an anti-I ⁇ Ip antibody (e.g., MAb 69.26) – heparin-biotin sandwich ELISA, SDS/PAGE, and/or Western blot.
  • the methods of the disclosure can be used to produce a yield of purified I ⁇ Ip of about 20% (w/w) or greater relative to the amount present in the original biological material.
  • the methods can be used to produce a yield of purified I ⁇ Ip of about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 65%, 70%, 75%, 80%, 85%, or 90% (w/w) or greater.
  • a liquid formulation comprising Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) and a buffer which is a citrate buffer and/or a histidine buffer, and wherein the formulation has a pH of about 5.0 to about 7.5.
  • the invention provides a liquid formulation comprising between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a citrate buffer, wherein the formulation has a pH of about 5.0 to about 7.5, and the formulation contains essentially no magnesium.
  • the invention provides a liquid formulation comprising between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of about 5.3 to about 5.7, and the formulation contains between about 40 mM to about 110 mM magnesium.
  • the invention also features a liquid formulation comprising Inter-alpha Inhibitor (I ⁇ I) and Pre- alpha Inhibitor (P ⁇ I) comprising significantly low amount of high MW species.
  • the liquid formulations described herein are not lyophilized. In an exemplary embodiment, the liquid formulations described herein do not contain a phosphate buffer. I ⁇ I and/or P ⁇ I [0028]
  • the liquid formulation comprises inter-alpha inhibitor (I ⁇ I) and/or pre-alpha inhibitor (P ⁇ I). In some embodiments, the liquid formulation comprises inter-alpha inhibitor (I ⁇ I) and pre-alpha inhibitor (P ⁇ I).
  • the liquid formulation comprises inter-alpha inhibitor (I ⁇ I) or pre-alpha inhibitor (P ⁇ I). In some embodiments, the liquid formulation comprises inter-alpha inhibitor (I ⁇ I) and not pre-alpha inhibitor (P ⁇ I). In some embodiments, the liquid formulation comprises pre-alpha inhibitor (P ⁇ I) and not inter- alpha inhibitor (I ⁇ I). [0029] In some embodiments, the liquid formulation comprises between 60% to 80% (w/w) I ⁇ I and between 20% to 40% (w/w) P ⁇ I. In some embodiments, the liquid formulation comprises between 60% to 70% (w/w) I ⁇ I and between 20% to 30% (w/w) P ⁇ I.
  • the liquid formulation comprises between 60% to 70% (w/w) I ⁇ I and between 30% to 40% (w/w) P ⁇ I. In some embodiments, the liquid formulation comprises between 62% to 72% (w/w) I ⁇ I and between 28% to 30% (w/w) P ⁇ I. In some embodiments, the liquid formulation comprises between 62% to 72% (w/w) I ⁇ I and between 28% to 38% (w/w) P ⁇ I. [0030] In some embodiments, essentially all of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 90% and about 99.5% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 80% and about 90% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 70% and about 80% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 68% and about 78% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 66% and about 76% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 64% and about 74% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 62% and about 72% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 60% and about 65% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 61% and about 66% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 62% and about 67% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 63% and about 68% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 64% and about 69% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 65% and about 70% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 66% and about 71% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 67% and about 72% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 68% and about 73% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 69% and about 74% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 59% and about 64% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 58% and about 63% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 57% and about 62% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 56% and about 61% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 55% and about 60% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 54% and about 59% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 54% and about 59% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 53% and about 58% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 52% and about 57% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 51% and about 56% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 50% and about 55% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 49% and about 54% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 48% and about 53% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 47% and about 52% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 46% and about 51% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 45% and about 50% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 60% and about 70% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 58% and about 68% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 56% and about 66% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 54% and about 64% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 52% and about 62% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 50% and about 60% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 48% and about 58% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 46% and about 56% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 44% and about 54% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 42% and about 52% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 40% and about 50% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 30% and about 40% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 20% and about 30% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I.
  • between about 10% and about 20% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. In some embodiments, between about 1% and about 10% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is I ⁇ I. [0031] In some embodiments, essentially all of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 90% and about 99.5% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 80% and about 90% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • between about 70% and about 80% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 60% and about 70% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 50% and about 60% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 40% and about 50% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 38% and about 48% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • between about 36% and about 46% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 34% and about 44% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 32% and about 42% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 30% and about 35% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 31% and about 36% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • between about 32% and about 37% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 33% and about 38% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 34% and about 39% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 35% and about 40% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 30% and about 40% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • between about 28% and about 38% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 26% and about 36% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 24% and about 34% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 22% and about 32% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 30% and about 35% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • between about 29% and about 34% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 28% and about 33% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 27% and about 32% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 26% and about 31% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 25% and about 30% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • between about 24% and about 29% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 23% and about 28% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 22% and about 27% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 21% and about 26% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 20% and about 25% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • between about 20% and about 30% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 10% and about 20% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I. In some embodiments, between about 1% and about 10% (w/w) of the I ⁇ I and P ⁇ I present in the liquid formulation is P ⁇ I.
  • I ⁇ I/P ⁇ I ratio [0032] In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 1.5 to about 3.5. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 1.7 to about 3.3. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.0 to about 3.0.
  • the I ⁇ I/P ⁇ I ratio is from about 2.0 to about 2.25. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.0 to about 2.2. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.05 to about 2.15. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is about 2.1. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.5 to about 3.0. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.6 to about 2.85. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.65 to about 2.85. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.7 to about 2.85.
  • the I ⁇ I/P ⁇ I ratio is from about 2.75 to about 2.85. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.75 to about 2.80. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is about 2.78. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.6 to about 3.0. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.7 to about 3.0. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.8 to about 3.0. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.9 to about 3.0.
  • the I ⁇ I/P ⁇ I ratio is from about 2.85 to about 2.95. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is from about 2.90 to about 2.95. In an exemplary embodiment, the I ⁇ I/P ⁇ I ratio is about 2.92. I ⁇ I and/or P ⁇ I concentration [0033] In an exemplary embodiment, the liquid formulations comprise between about 5 mg/mL and about 100 mg/mL of I ⁇ I and/or P ⁇ I. In an exemplary embodiment, the liquid formulations comprise between about 10 mg/mL and about 100 mg/mL of I ⁇ I and/or P ⁇ I. In an exemplary embodiment, the liquid formulations comprise between about 10 mg/mL and about 50 mg/mL of I ⁇ I and/or P ⁇ I.
  • the liquid formulations comprise between about 15 mg/mL and about 25 mg/mL of I ⁇ I and/or P ⁇ I. In an exemplary embodiment, the liquid formulations comprise between about 17 mg/mL and about 23 mg/mL of I ⁇ I and/or P ⁇ I. In an exemplary embodiment, the liquid formulations comprise between about 19 mg/mL and about 21 mg/mL of I ⁇ I and/or P ⁇ I.
  • High MW species [0034]
  • less than 20% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 20% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • between about 0.1% and about 20% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 20% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 15% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 15% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • between about 0.1% and about 15% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 15% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 10% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • between about 0.1% and about 10% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 5% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • between about 0.1% and about 5% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.5% and about 5% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.5% and about 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • between about 0.5% and about 4% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.5% and about 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 1% and about 4% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 1% and about 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • between about 2% and about 4% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 2% and about 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • Concentrations of impurities Factor II, Factor X [0035]
  • the formulation contains less than 0.03 mg/mL of Factor II (Prothrombin). In an exemplary embodiment, the formulation contains less than 0.02 mg/mL of Factor II (Prothrombin). In an exemplary embodiment, the formulation contains less than 0.01 mg/mL of Factor II (Prothrombin).
  • the formulation comprising less than 0.01 mg/mL of Factor X. In an exemplary embodiment, the formulation comprising less than 0.005 mg/mL of Factor X. In an exemplary embodiment, the formulation comprising less than 0.001 mg/mL of C4 binding protein. In an exemplary embodiment, the formulation comprising less than 1.5 ⁇ g/mL of Factor II and less than 0.8 ⁇ g/mL of Factor X.
  • NaCl [0036] In an exemplary embodiment, the formulation further comprises essentially no NaCl. In an exemplary embodiment, the formulation further comprises between about 5 mM to about 110 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 40 mM to about 110 mM NaCl.
  • the formulation further comprises between about 40 mM to about 60 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 90 mM to about 110 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 20 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 20 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 5 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 5 mM NaCl. Sucrose [0037] In an exemplary embodiment, the formulation further comprises sucrose.
  • the formulation further comprises between about 1.5% (w/w) and about 2.5% (w/w) sucrose. In an exemplary embodiment, the formulation further comprises between about 1.8% (w/w) and about 2.2% (w/w) sucrose. In an exemplary embodiment, the formulation further comprises between about 1.9% (w/w) and about 2.1% (w/w) sucrose. In an exemplary embodiment, the formulation further comprises between about 1.8% (w/w) and about 2.0% (w/w) sucrose.
  • Glycine [0038] In an exemplary embodiment, the formulation further comprises glycine. In an exemplary embodiment, the formulation further comprises between about 0.5% (w/w) and about 1.5% (w/w) glycine.
  • the formulation further comprises between about 0.8% (w/w) and about 1.2% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 0.9% (w/w) and about 1.1% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 0.7% (w/w) and about 0.9% (w/w) glycine.
  • Sucrose and Glycine [0039] In an exemplary embodiment, the formulation further comprises sucrose and glycine. In an exemplary embodiment, the formulation further comprises between about 1.5% (w/w) and about 2.5% (w/w) sucrose and between about 0.5% (w/w) and about 1.5% (w/w) glycine.
  • the formulation further comprises between about 1.8% (w/w) and about 2.2% (w/w) sucrose and between about 0.8% (w/w) and about 1.2% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 1.9% (w/w) and about 2.1% (w/w) sucrose and between about 0.9% (w/w) and about 1.1% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 1.8% (w/w) and about 2.0% (w/w) sucrose and between about 0.7% (w/w) and about 0.9% (w/w) glycine. Buffer in the formulation [0040] In an exemplary embodiment, the buffer is citrate or histidine.
  • the buffer is citrate. In an exemplary embodiment, the buffer is histidine.
  • Buffer concentration in the formulation [0041] In an exemplary embodiment, the buffer concentration is between about 10 mM and about 30 mM. In an exemplary embodiment, the buffer concentration is between about 15 mM and about 25 mM. In an exemplary embodiment, the buffer concentration is between about 17 mM and about 23 mM.
  • pH [0042] In an exemplary embodiment, the formulation has a pH of about 5.4 to about 6.6. In an exemplary embodiment, the formulation has a pH of about 5.5 to about 6.5. In an exemplary embodiment, the formulation has a pH of about 5.3 to about 5.7.
  • the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the formulation has a pH of about 6.3 to about 6.7. In an exemplary embodiment, the formulation has a pH of about 6.4 to about 6.6.
  • the formulation further comprises between about 40 mM to about 60 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 90 mM to about 110 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 20 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 20 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 5 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 1 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 5 mM magnesium. Stability [0044] In an exemplary embodiment, the formulation is stable.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a citrate buffer, wherein the formulation has a pH of between about 5.0 to about 6.0, and the formulation contains between about 0 mM to about 1 mM magnesium.
  • I ⁇ I Inter-alpha Inhibitor
  • P ⁇ I Pre-alpha Inhibitor
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a citrate buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 0 mM to about 1 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a citrate buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 0 mM to about 1 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a citrate buffer, wherein the formulation has a pH of between about 6.0 to about 7.0, and the formulation contains between about 0 mM to about 1 mM magnesium.
  • I ⁇ I Inter-alpha Inhibitor
  • P ⁇ I Pre-alpha Inhibitor
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a citrate buffer, wherein the formulation has a pH of between about 6.3 to about 6.7, and the formulation contains between about 0 mM to about 1 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a citrate buffer, wherein the formulation has a pH of about 6.5, and the formulation contains between about 0 mM to about 1 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 30 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I), wherein the formulation comprises less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa, wherein the formulation comprises a buffer, and wherein the formulation has a pH of about 5.0 to about 7.5.
  • I ⁇ I Inter-alpha Inhibitor
  • P ⁇ I Pre-alpha Inhibitor
  • the liquid formulation comprises between about 10 mg/mL and about 30 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I), wherein the formulation comprises less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa, wherein the formulation comprises a buffer which is a citrate buffer and/or a histidine buffer, and wherein the formulation has a pH of about 5.0 to about 7.5.
  • I ⁇ I Inter-alpha Inhibitor
  • P ⁇ I Pre-alpha Inhibitor
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 5 mM to about 110 mM magnesium.
  • I ⁇ I Inter-alpha Inhibitor
  • P ⁇ I Pre-alpha Inhibitor
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 40 mM to about 110 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 40 mM to about 110 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 40 mM to about 60 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre- alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 40 mM to about 60 mM magnesium.
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 90 mM to about 110 mM magnesium.
  • I ⁇ I Inter-alpha Inhibitor
  • P ⁇ I Pre-alpha Inhibitor
  • the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (I ⁇ I) and Pre-alpha Inhibitor (P ⁇ I) in a histidine buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 90 mM to about 110 mM magnesium [0048]
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM citrate buffer, between about 40 mM to about 60 mM NaCl, essentially no magnesium, wherein the formulation has a pH of about 6.4 to about 6.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM citrate buffer, between about 40 mM to about 60 mM NaCl, sucrose, glycine, essentially no magnesium, wherein the formulation has a pH of about 6.4 to about 6.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM citrate buffer, between about 40 mM to about 60 mM NaCl, between about 1.5% and about 2.5% sucrose and between about 0.5% and about 1.5% glycine, essentially no magnesium, wherein the formulation has a pH of about 6.4 to about 6.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl and between about 90 mM to about 110 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 90 mM to about 110 mM magnesium, sucrose, and glycine, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 90 mM to about 110 mM magnesium, and between about 1.5% and about 2.5% sucrose and between about 0.5% and about 1.5% glycine, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl and between about 40 mM to about 60 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 40 mM to about 60 mM magnesium, sucrose, and glycine, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 40 mM to about 60 mM magnesium, between about 1.5% and about 2.5% sucrose, and between about 0.5% and about 1.5% glycine, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, between about 90 mM to about 110 mM NaCl and between about 40 mM to about 60 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, between about 90 mM to about 110 mM NaCl, between about 40 mM to about 60 mM magnesium, sucrose, and glycine, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of I ⁇ I and P ⁇ I, between about 17 mM and about 23 mM histidine buffer, between about 90 mM to about 110 mM NaCl, between about 40 mM to about 60 mM magnesium, between about 1.5% and about 2.5% sucrose, and between about 0.5% and about 1.5% glycine, wherein the formulation has a pH of about 5.4 to about 5.6.
  • the I ⁇ Ip can be purified according to methods known in the art, such as those described in PCT Pat App PCT/US04/36848, US Pat App 16/866,241, PCT Pat App PCT/US09/03291, US Pat App 15/439,328, PCT Pat App PCT/US14/12033, PCT Pat App PCT/US18/29436, and US Pat App 17/404,588, each of which is herein incorporated by reference in their entirety.
  • the I ⁇ I and the P ⁇ I can be purified according to a method below: [1] A method of purifying an I ⁇ I and a P ⁇ I from a biological material comprising: (a) applying the biological material comprising the I ⁇ I and the P ⁇ I to a strong anion exchanger and separating a flow through; (b) applying a wash buffer to the strong anion exchanger and separating a flow through; and (c) applying an elution buffer to the strong anion exchanger and collecting an eluate comprising the I ⁇ I and the P ⁇ I; wherein conductivity of the wash buffer is less than 15 mS/cm, wherein pH of the wash buffer is about 5.0 to about 5.5, wherein the elution buffer comprise about 350 mM to about 450 mM NaCl.
  • a method of purifying an I ⁇ I and a P ⁇ I from a biological material comprising: (a) applying the biological material comprising the I ⁇ I and the P ⁇ I to a strong anion exchanger and separating a flow through; (b) applying a first wash buffer to the strong anion exchanger and separating a flow through; (c) applying a second wash buffer to the strong anion exchanger and separating a flow through; (d) applying a third wash buffer to the strong anion exchanger and separating a flow through; (e) applying an elution buffer to the strong anion exchanger and collecting an eluate comprising the I ⁇ I and the P ⁇ I; wherein conductivity of the second wash buffer is less than 15 mS/cm, wherein pH of the second wash buffer is about
  • Characteristics of the formulation comprising the I ⁇ I and the P ⁇ I purified by improved methods of purification have beneficial features below: (i) In an exemplary embodiment, less than 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • less than 3% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa.
  • more than 80% of the I ⁇ I and the P ⁇ I does not exist as a protein aggregates.
  • more than 85% of the I ⁇ I and the P ⁇ I does not exist as a protein aggregates.
  • more than 90% of the I ⁇ I and the P ⁇ I does not exist as a protein aggregates.
  • the formulation has an at least 20% higher trypsin inhibitory specific activity than known formulations comprising I ⁇ Ips.
  • the formulation has an at least 25% higher trypsin inhibitory specific activity than known formulations comprising I ⁇ Ips. In an exemplary embodiment, the formulation has an at least 30% higher trypsin inhibitory specific activity than known formulations comprising I ⁇ Ips. In an exemplary embodiment, the formulation has an at least 40% higher trypsin inhibitory specific activity than known formulations comprising I ⁇ Ips. In an exemplary embodiment, the formulation has an at least 50% higher trypsin inhibitory specific activity than known formulations comprising I ⁇ Ips.
  • the invention provides a method of purifying inter-alpha inhibitor (I ⁇ I) and pre-alpha inhibitor (P ⁇ I) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b) contacting the product of the previous step with a second anion-exchange resin; c) polishing the product of the previous step, thereby producing purified I ⁇ I and P ⁇ I.
  • the blood product material is cryo-poor plasma.
  • the a) comprises: a1) contacting the blood product material in a loading butter to a first anion-exchange resin, under conditions in which I ⁇ I and P ⁇ I adhere to the resin and separate a flow-through; a2) contacting the product of a1) with an elution buffer in which I ⁇ I and P ⁇ I elute from the resin.
  • the first anion- exchange resin is a weak anion exchange resin.
  • the first anion- exchange resin comprises a DEAE moiety.
  • the first anion- exchange resin is DEAE-Sephadex A-50, Toyopearl-GigaCap DEAE-650M, or DEAE- Sepharose FF.
  • the b) comprises: b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which I ⁇ I and P ⁇ I adhere to the resin and separate a flow-through; b2) contacting the product of b1) with an elution buffer in which I ⁇ I and P ⁇ I elute from the resin.
  • the second anion-exchange resin is a strong anion exchange resin.
  • the second anion-exchange resin comprises a quaternary ammonium moiety.
  • the second anion-exchange resin is a weak anion exchange resin.
  • the second anion-exchange resin comprises a DEAE moiety.
  • the second anion-exchange resin is TOYOPEARL GigaCap Q-650M, Capto Q, Poros XQ, Eshmuno Q, Chromalite MQ/F, POROS 50HQ, Q-PuraBead HF, Praesto Jetted Q35, DEAE-Sepharose, or Toyopearl-GigaCap DEAE- 650M.
  • the second anion-exchange resin is DEAE- Sepharose or Toyopearl-GigaCap DEAE-650M.
  • the second anion-exchange resin is TOYOPEARL GigaCap Q-650M, Capto Q, Poros XQ, Eshmuno Q, Chromalite MQ/F, POROS 50HQ, Q-PuraBead HF, Praesto Jetted Q35.
  • the second anion-exchange resin is TOYOPEARL GigaCap Q-650M.
  • the b1) further comprises: the ratio of total protein (in mg) in the product of the previous step to mL of resin is between about 15 mg/mL to about 100 mg/mL.
  • the b1) further comprises: the ratio of total protein (in mg) in the product of the previous step to mL of resin is between about 1 mg/mL to about 50 mg/mL, or between about 5 mg/mL to about 40 mg/mL, or between about 10 mg/mL to about 30 mg/mL. In an exemplary embodiment, the b1) further comprises: the ratio of total protein (in mg) in the product of the previous step to mL of resin is between about 15 mg/mL to about 25 mg/mL.
  • the loading buffer in b1) comprises a conductivity of between about 10-22 mS/cm; a pH range of 6.0-8.0, and a NaCl concentration of between about 50 mM and about 220 mM. In an exemplary embodiment, wherein the loading buffer in b1) comprises a conductivity of between about 15 mS/cm and about 25 mS/cm, or between about 19 mS/cm and about 22 mS/cm. In an exemplary embodiment, wherein the loading buffer in b1) comprises a pH of about 7.0 to about 8.0.
  • the loading buffer in b1) comprises a NaCl concentration of between about 0 mM and about 220 mM, or between about 150 mM to about 250 mM, or between about 180 mM to about 220 mM.
  • the b1) further comprises, after contacting with a loading buffer, contacting with a first wash buffer and separating a flow-through, the first wash buffer has a conductivity of between about 15-25 mS/cm; a pH range of 6.0-8.0; a NaCl concentration of between about 150 mM and about 250 mM, and a column volume (CV) of between about 3 to about 10.
  • the b1) further comprises, after the contacting with the loading buffer, contacting with a first wash buffer and separating a flow-through, the first wash buffer has a conductivity of between about 15-25 mS/cm; a pH range of 6.0-8.0; a NaCl concentration of between about 150 mM and about 250 mM, and a column volume (CV) of between about 3 to about 10.
  • the first wash buffer has a conductivity of between about 19-22 mS/cm.
  • the first wash buffer has a pH of between about 7.0 and about 7.5.
  • the first wash buffer has a NaCl concentration of between about 200 mM and about 250 mM.
  • the column volume is from about 3 to about 8, or about 5 to about 10, or between about 3 to about 5, or between about 5 to about 8.
  • the second wash buffer has a conductivity of between about 0-15 mS/cm; a pH range of 4.2-6.2, a NaCl concentration of between about 0 mM and about 100 mM, and a column volume (CV) of between about 3 to about 10.
  • the second wash buffer has a conductivity of between about 0-15 mS/cm; a pH range of 4.2-6.2, a NaCl concentration of between about 0 mM and about 100 mM, and a column volume (CV) of between about 3 to about 10.
  • the second wash buffer has a conductivity of between about 0 mS/cm and about 15 mS/cm, or about 10 mS/cm to about 15 mS/cm, or about 12 mS/cm to about 13 mS/cm.
  • the second wash buffer has a NaCl concentration of between about 50 mM to about 80 mM, or about 60 mM to about 70 mM. In an exemplary embodiment, the second wash buffer has a NaCl concentration of between about 100 mM to about 200 mM, or about 140 mM to about 160 mM. In an exemplary embodiment, the second wash buffer has a pH of about 5.0 to about 5.4. In an exemplary embodiment, the second wash buffer has a pH of about 4.7 to about 5.7. In an exemplary embodiment, the second wash buffer has a pH of about 4.2 to about 6.2.
  • the column volume is from about 3 to about 8, or about 5 to about 10, or between about 3 to about 5, or between about 5 to about 8.
  • the invention provides a method of purifying inter-alpha inhibitor (I ⁇ I) and pre-alpha inhibitor (P ⁇ I) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which I ⁇ I and P ⁇ I adhere to the resin and separate a flow-through; b2) contacting the product of b1) with an elution buffer in which I ⁇ I and P ⁇ I elute from the resin; c) polishing the product of the previous step, thereby producing purified I ⁇ I and P ⁇ I.
  • the invention provides a method of purifying inter-alpha inhibitor (I ⁇ I) and pre-alpha inhibitor (P ⁇ I) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which I ⁇ I and P ⁇ I adhere to the resin and separate a flow-through, and then contacting with a first wash buffer and separating a flow-through, and then contacting with a second wash buffer and separating a flow-through, wherein the second wash buffer has a conductivity of between about 0 mS/cm and about 15 mS/cm, or about 10 mS/cm to about 15 mS/cm, or about 12 mS/cm to about 13 mS/cm; and b2) contacting the product of b1) with an elution buffer in which I ⁇ I and P ⁇ I
  • the invention provides a method of purifying inter-alpha inhibitor (I ⁇ I) and pre-alpha inhibitor (P ⁇ I) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which I ⁇ I and P ⁇ I adhere to the resin and separate a flow-through, and then contacting with a first wash buffer and separating a flow- through, and then contacting with a second wash buffer and separating a flow-through, wherein the second wash buffer has a conductivity of between about 0 mS/cm and about 15 mS/cm, or about 10 mS/cm to about 15 mS/cm, or about 12 mS/cm to about 13 mS/cm, and the second wash buffer has a pH of about 4.2 to about 6.2, or about 4.7 to about 5.7, or about
  • the b1) further comprises, after the contacting with the second wash buffer, contacting with a third wash buffer and separating a flow-through, the third wash buffer has a conductivity of between about 0-20 mS/cm, a pH range of 6.5-7.5, a NaCl concentration of between about 0 mM and about 200 mM, and a column volume (CV) of between about 3 to about 10.
  • the third wash buffer has a NaCl concentration of between about 50 mM to about 80 mM, or about 60 mM to about 70 mM.
  • the third wash buffer has a pH is from about 4.7 to about 5.7, or from about 4.2 to about 6.2 or from about 7.0 to about 7.5.
  • the elution buffer in b2) has a conductivity of between about 35-40 mS/cm; a pH range of 6.5-7.5, and a NaCl concentration of between about 350 mM and about 450 mM.
  • the elution buffer in b2) has a conductivity of between about 65-80 mS/cm; a pH range of 6.5-7.5, and a NaCl concentration of between about 700 mM and about 800 mM.
  • Therapeutic efficacy of the formulations may be monitored, e.g., by methods known in the art, to determine pre- and post-treatment levels of I ⁇ Ips (e.g., I ⁇ I, P ⁇ I, a heavy chain (e.g., H1, H2, H3, H4, and/or H5), a light chain (e.g., bikunin), or a combination thereof).
  • I ⁇ Ips e.g., I ⁇ I, P ⁇ I, a heavy chain (e.g., H1, H2, H3, H4, and/or H5), a light chain (e.g., bikunin), or a combination thereof).
  • the level of I ⁇ Ips and I ⁇ Ip-related biomarkers can be detected and/or measured e.g., by gas phase ion spectrometry methods, optical methods, electrochemical methods, atomic force microscopy, radio frequency methods, surface plasmon resonance, ellipsometry, and immunological methods.
  • Sedimentation rate can be measured using standard clinical tests (e.g., blood tests).
  • An immunoassay can be used to detect and analyze I ⁇ Ips (e.g., I ⁇ I, P ⁇ I, a heavy chain (e.g., H1, H2, H3, H4, and/or H5), a light chain (e.g., bikunin), or a combination thereof) and/or other biomarker protein levels in a sample.
  • the immunoassay can include: (a) providing an antibody that specifically binds to I ⁇ I and/or P ⁇ I; (b) contacting a sample with the antibody; and (c) detecting the presence of a complex of the antibody bound to the proteins in the sample.
  • Suitable antibodies for use in an immunoassay to detect I ⁇ Ips include, MAb 69.31, MAb 69.26, anti-I ⁇ Ip polyclonal antibody, and anti-bikunin monoclonal or polyclonal antibody.
  • Total Protein in a solution can be measured with a assay such as spectroscopic like UV280, or colorimetry based on BCA (Bicinchoninic acid assay), Biuret assay or Bradford.
  • the liquid formulations comprising I ⁇ I and/or P ⁇ I were prepared (Table 1) by dialyzing an I ⁇ I and/or P ⁇ I-containing Bulk Drug Substance (BDS) against the buffers shown in Table 1. Depending on the protein content the dialyzed solutions were either diluted with the same buffer or concentrated with Vivaspin ultrafiltration tubes to a protein concentration of around 20 mg/mL. Afterwards the solutions were sterile-filtered and filled into glass vials, closed with a rubber stopper and an Aluminum-cap. The vials were stored at 2-8°C over 6 months and the stability was investigated at start, 1 month, 2 months, 4 months and 6 months with biochemical and bio-physical methods. Table 1.
  • Fig.1 Turbidity was investigated at start, 1 month, 2 months, 4 months, and 6 months ( Figure 1). The turbidity of the 5 formulations differed from one another but did not show any relevant change over 6 months at 2-8°C. Visual appearance (particle formation) also did not change over 6 months at 2-8°C.
  • Fig.2 SEC aggregation Size-Exclusion Chromatography (SEC) main peaks were investigated over 6 months ( Figure 2). The lowest decrease of SEC-main peaks (IaI + PaI) was for A, followed by B at 2- 8°C after 6 months ( Figure 2).
  • Example 2 Process Description [0072] Frozen plasma units were pooled for thawing at a maximum temperature of 6°C. Liquid solid separation of the thawed plasma pool was performed through centrifugation. The cryo-poor plasma centrifugate was collected and further processed. [0073] 0.5 g of an ion exchange gel (DEAE-Sephadex) was added per liter centrifugate. After sedimentation the protein/complex was separated from residual supernatant by filtration. The protein/DEAE-Sephadex complex was washed and the protein was subsequently eluted from the DEAE-Sephadex with a sodium chloride solution. The intermediate was stored frozen until further use.
  • DEAE-Sephadex sodium chloride solution
  • wash buffer 1 (20 mM Tris, 250 mM sodium chloride, pH 7.2 ⁇ 0.2) and wash buffer 2 (75 mM glycine, 100 mM sodium acetate 65 mM sodium chloride, pH 5.2 ⁇ 0.2).
  • the equilibration buffer (20 mM Tris, 200 mM sodium chloride, pH 7.2 ⁇ 0.2) was taken as the third wash buffer.
  • Proteins were eluted with Q elution buffer (20 mM Tris, 400 mM sodium chloride, pH 7.2 ⁇ 0.2). The eluate was collected in a jacketed stainless-steel vessel for further processing.
  • the eluate of the Q column was diluted at a ratio of 1:2 (1+1) with purified water prior to EtoxiClear loading.
  • the diluted solution was loaded onto the equilibrated EtoxiClear column. After loading was completed, the column was washed with wash buffer 1 at pH 5.2 (75 mM glycine, 0.1 M AcOH, 200 mM NaCl), followed by washing the column using wash buffer 2 (20 mM Tris, 300 mM sodium chloride 7.2 ⁇ 0.2).
  • wash buffer 1 pH 5.2 (75 mM glycine, 0.1 M AcOH, 200 mM NaCl)
  • wash buffer 2 (20 mM Tris, 300 mM sodium chloride 7.2 ⁇ 0.2.
  • IAIP was then eluted with the EtoxiClear elution buffer (20 mM Tris, 500 mM sodium chloride, pH 7.2 ⁇ 0.2).
  • the IAIP eluate solution was diluted 1:2 (1+1) with purified water.
  • the diluted IAIP eluate was subsequently passed through a 20 nm filter. The pressure during filtration was controlled to not more than 0.9 bar differential pressure.
  • the nanofiltrate was concentrated to a protein concentration of 15 mg/mL through ultrafiltration.
  • the UF membrane used had a nominal molecular weight cut off (NMWCO) of 50,000 daltons or lower.
  • the concentrate was diafiltered against liquid formulation (e.g., liquid formulations described in Table 1). The minimum exchange volume was 10 times of the original concentrate volume. Throughout the ultrafiltration/diafiltration operation, the solution was maintained at 4 to 25°C.
  • the protein/DEAE-Sephadex complex was washed, and protein was subsequently eluted from the DEAE-Sephadex with a sodium chloride solution. The intermediate was stored frozen until further use. [0087] After thawing, the product was filtered through a membrane filter. The filtrate and the post wash were collected for further processing. [0088] A stock solution of Polysorbate 80 and Tri-N-butyl phosphate was prepared in a separate vessel and was added while stirring to a calculated final concentration of 1.0% Polysorbate 80 and 0.30% Tri-N-butyl phosphate, respectively. After addition, the solution was stirred until complete homogenization and subsequently filtered through a membrane filter.
  • the resin was washed with wash buffer 1 (20 mM Tris, 250 mM sodium chloride, pH 7.2 ⁇ 0.2) and wash buffer 2 (75 mM glycine, 100 mM acetic acid 150 mM sodium chloride, pH 5.2 ⁇ 0.2).
  • the Q equilibration buffer (20 mM Tris, 200 mM sodium chloride, pH 7.2 ⁇ 0.2) was taken as the third wash buffer.
  • Proteins were eluted with Q elution buffer (20 mM Tris, 750 mM sodium chloride, pH 7.2 ⁇ 0.2). The eluate was collected in a jacketed stainless-steel vessel for further processing.
  • the Q eluate was diluted at a ratio of 1:4 (1+3) with purified water or with buffer matrix without salt.
  • the diluted solution was loaded onto the equilibrated EtoxiClear column. After loading was completed, the column was washed with wash buffer 1 at pH 5.2 (75 mM glycine, 0.1 M AcOH, 200 mM NaCl) followed by washing the column using wash buffer 2 (20 mM Tris, 300 mM sodium chloride 7.2 ⁇ 0.2).
  • wash buffer 1 pH 5.2 (75 mM glycine, 0.1 M AcOH, 200 mM NaCl) followed by washing the column using wash buffer 2 (20 mM Tris, 300 mM sodium chloride 7.2 ⁇ 0.2).
  • wash buffer 2 (20 mM Tris, 300 mM sodium chloride 7.2 ⁇ 0.2.
  • IAIP was then eluted with EtoxiClear elution buffer (20 mM Tris, 500 mM sodium chloride 7.2 ⁇
  • the IAIP eluate solution was diluted 1:2 (1+1) with purified water.
  • the diluted IAIP eluate was subsequently passed through a 20 nm filter. The pressure during filtration was controlled to not more than 0.9 bar differential pressure.
  • the nanofiltrate was concentrated to a protein concentration of 15 mg/mL through ultrafiltration.
  • the UF membrane used had a nominal molecular weight cut off (NMWCO) of 50,000 daltons or lower.
  • the concentrate was diafiltered against liquid formulation (e.g., liquid formulations described in Table 1). The minimum exchange volume was 10 times of the original concentrate volume. Throughout the ultrafiltration/diafiltration operation, the solution was maintained at 4 to 25°C.
  • the protein/DEAE-Sephadex complex was washed and protein was subsequently eluted from the DEAE-Sephadex with a sodium chloride solution. The intermediate was stored frozen until further use. [0102] After thawing, the product was filtered through a membrane filter. The filtrate and the post wash were collected for further processing. [0103] A stock solution of Polysorbate 80, Tri-N-butyl phosphate and Triton X-100 reduced was prepared and added to the thawed product while stirring to a calculated final concentration of 0.30% Polysorbate 80 and 0.30% Tri-N-butyl phosphate, and 1% Triton X-100 reduced respectively.
  • the solution was stirred until complete homogenization (for at least 30 minutes) and subsequently filtered through a membrane filter.
  • the temperature during S/D treatment was maintained at 19.0 to 25.0°C. The time required was a minimum of 120 minutes.
  • the solution was diluted to a target conductivity of 22.5 ⁇ 2.5 mS/cm with Q equilibration buffer (20 mM Tris-HCl, 200 mM NaCl, pH 7.2 ⁇ 0.1) and pH was adjusted to 7.2 ⁇ 0.1 by addition of HCl.
  • the conductivity of Q equilibration buffer was 21 ⁇ 2 mS/cm.
  • the resin was washed with approximately 8 CV of wash buffer 1 (20 mM Tris-HCl, 250 mM sodium chloride, pH 7.2 ⁇ 0.1, conductivity 26 ⁇ 2 mS/cm) and with approximately 8 CV of wash buffer 2 (75 mM glycine, 100 mM acetic acid 150 mM sodium chloride, 92.5 mM sodium hydroxide, pH 5.2 ⁇ 0.1, conductivity 20 ⁇ 2 mS/cm).
  • the Q equilibration buffer with approximately 5 CV (20 mM Tris-HCl, 200 mM sodium chloride, pH 7.2 ⁇ 0.1, conductivity 21 ⁇ 2 mS/cm) was taken as the third wash buffer.
  • Proteins were eluted with Q elution buffer (20 mM Tris-HCl, 750 mM sodium chloride, pH 7.2 ⁇ 0.1, conductivity 66 ⁇ 2 mS/cm). The eluate was collected in a jacketed stainless-steel vessel for further processing. [0107] The eluate from the Q column was diluted at a ratio of 1:4 (1+3) with dilution buffer (10 mM Tris-HCl, pH 7.2 ⁇ 0.1, conductivity 1 ⁇ 0.5 mS/cm). [0108] The diluted solution was loaded at a ratio of approximately 10 mg total protein per mL resin at a flow rate of 120 cm/h onto the equilibrated EtoxiClear column.
  • This column had a bed height of 10.5 cm and a diameter of 14 cm.
  • the volume of the column was 1.6 L and the residence time was 5.25 min.
  • the column was washed with approximately 8 CV with wash buffer 1 at pH 5.2 (75 mM glycine, 100 mM acetic acid, 200 mM sodium chloride, 92.5 mM NaOH, pH 5.2 ⁇ 0.1, conductivity 25 ⁇ 2 mS/cm) followed by washing the column with approximately 5 CV of wash buffer 2 (20 mM Tris-HCl, 300 mM sodium chloride 7.2 ⁇ 0.1, conductivity 30 ⁇ 2 mS/cm).
  • IAIP was then eluted with approximately 3 CV with EtoxiClear elution buffer (20 mM Tris-HCl, 500 mM sodium chloride 7.2 ⁇ 0.1, conductivity 47 ⁇ 2 mS/cm). [0110] The IAIP eluate solution was diluted 1:2 (1+1) with dilution buffer (10 mM Tris-HCl, pH 7.2 ⁇ 0.1, 1 ⁇ 0.5 mS/cm). [0111] The diluted IAIP eluate was subsequently passed through a 20 nm filter. A clarifying filter was connected in series to protect the nanofilter. The pressure during filtration was controlled to not more than 0.9 bar differential pressure.
  • the nanofiltrate was concentrated to a protein concentration of 15 mg/mL through ultrafiltration.
  • the UF membrane used had a nominal molecular weight cut off (NMWCO) of 30,000 daltons.
  • the concentrate was diafiltered against a diafiltration buffer which resembled liquid formulation. The exchange volume was 10 times of the original concentrate volume. Throughout the ultrafiltration/diafiltration operation, the solution was maintained at 4 to 25°C.
  • the solution was concentrated to a protein concentration of minimum 2.5% w/v. The solution temperature was maintained at 4°C to 25°C. Example 5.
  • Example 4 The product of Example 4 was analyzed for dimer and multimer aggregates of IAIP using high performance Size-Exclusion Chromatography (HP-SEC). Dimer and multimer aggregates of I ⁇ I and P ⁇ I have a molecular weight of more than 500 kDa.
  • the % of product with a molecular weight of more than 500 kDa (Peak 1) was 3.61%.
  • the % of product with a molecular weight of approximately 360 kDa (Peak 2, I ⁇ I) was 63.87%.
  • the % of product with a molecular weight of approximately 250 kDa (Peak 3, P ⁇ I) was 30.47%.
  • the % of product with a molecular weight of approximately 100 kDa, 60 kDa, and less than 50 kDa was 1.29%, 0.51%, and 0.25% respectively (see Table 2).
  • the sum % area of Peaks 2 & 3 representing the purity of IaI and PaI was 94.34%.
  • the ratio of the % area of Peak 2 (IaI) to peak 3 (PaI) was 2.10.
  • the final container concentration was 26.30 mg/mL total protein.
  • Table 2 HP-SEC results of IAIP concentrate Example 6. Wash Buffer Conductivity Effect on Turbidity [0117] The effect of wash buffer conductivity in the Anion Exchange Chromatography step on product turbidity was examined.
  • Cryo-poor plasma centrifugates were prepared and main steps were processed as described in Example 4.
  • S/D treatment was performed using a final concentration of 1% Polysorbate 80 and 0.30% Tri-N-butyl phosphate. No additional filtration after S/D reagent addition was performed.
  • S/D treated and filtered protein solutions were loaded onto an equilibrated TOYOPEARL GigaCap Q-650M column and were washed with wash buffer 1 as described in Example 4. Subsequent wash 2 was performed by applying wash buffer 2 with a conductivity of either 13 mS/cm or 20 mS/cm.

Abstract

Liquid IαI and PαI formulations are described.

Description

INTER-ALPHA INHIBITOR PROTEIN FORMULATIONS PRIORITY CLAIM [0001] This application claims benefit to U.S. Provisional Application Serial No.63/280,057, filed November 16, 2021, which is incorporated herein by reference in its entirety. BACKGROUND [0002] Inter-alpha inhibitor proteins (IαIps) are a family of structurally related proteins found in plasma that are involved in inflammatory regulation and wound healing. The major forms of IαIps are Inter-alpha Inhibitor (IαI), which consists of two heavy chains (H1 & H2) and a single light chain (e.g., bikunin), and Pre-alpha Inhibitor (PαI) consisting of one heavy (H3) and one light chain (e.g., bikunin). IαIps are reduced during inflammatory processes such as sepsis and stroke. Prior studies indicate that IαIp levels are inversely correlated with morbidity and mortality in severe inflammatory conditions and that patients whose IαIp levels had recovered over time exhibited improved outcomes. Also, replenishing with exogenous IαIp in experimental models of severe inflammation provided recovery in multiple animal studies across species and indication areas. [0003] There exists a need for improved formulations for treating diseases and conditions characterized by inflammation and/or low IαIps levels in blood. SUMMARY OF THE INVENTION [0004] In one aspect, the invention provides a liquid formulation comprising Inter-alpha Inhibitor Proteins (IαIps) and a buffer which is a citrate buffer and/or a histidine buffer, and wherein the formulation has a pH of about 5.0 to about 7.5. In some embodiments of any of the foregoing aspects, the IαIp is a human IαIp. [0005] In one aspect, the invention provides a method of purifying an IαI and a PαI from a biological material by anion-exchange chromatography, wherein a wash buffer and/or an elution buffer used in the anion exchange chromatography step has a low conductivity. The invention also provides a formulation comprising an IαI and a PαI, wherein the IαI and the PαI are purified by the method above. The formulation has beneficial features, comprising that (i) less than 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa, (ii) more than 80% of the IαI and the PαI does not exist as a protein aggregate, (iii) the formulation has at least 20% higher trypsin inhibitory specific activity than known formulations comprising IαIps, (iv) the formulation comprises less than 0.03 mg/ml of Factor II (Prothrombin), (v) the formulation comprising less than 0.01 mg/ml of Factor X, and/or (vi) the formulation comprising less than 0.001 mg/ml of C4 binding protein. DEFINITIONS [0006] As used herein, the singular form “a,” “an,” and “the” includes plural references unless indicated otherwise. [0007] As used herein, the term “about” means +/- 10% of the recited value. [0008] As used herein, “administering” means a method of giving a dosage of a formulation (e.g., an IαIp) to a subject. The formulations utilized in the methods described herein can be administered, for example, orally, intramuscularly, intravenously, intradermally, percutaneously, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, topically, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, by catheter, by lavage, in creams, or in lipid formulations. The method of administration can vary depending on various factors (e.g., the formulation being administered and the severity of the condition, disease, or disorder being treated). [0009] The term “biological material” as used herein refers to a sample from a subject (e.g., a mammal, such as a human) that contains IαIp. Examples of the biological material include a blood product material, e.g., whole plasma, cryo-poor plasma, liquid plasma, fresh frozen plasma (FFP), FFP24, frozen plasma (FP), FP24, thawed FFP, thawed FFP24, thawed FP, thawed FP24, source plasma, recovered plasma, solvent/detergent-treated plasma (SDP), platelet-rich plasma (PRP), platelet-poor plasma (PPP), serum, blood, and a diluted or concentrated preparation thereof, milk or colostrum, urine, sputum, and cerebrospinal fluid. Examples of the biological material include a plasma fraction intermediate or a blood product which is produced through one or more process steps (e.g. filtration, centrifugation, sedimentation, chromatography, adsorption, isolation, freezing, thawing, dilution, concentration, virus clearance, etc). The biological material can be from a human, primate, bovine, equine, porcine, ovine, feline, canine, or combinations thereof. The biological material may also be an extract prepared using cells that express IαIp, or may be or contain cells that secrete IαIp, e.g., cells that have been recombinantly modified to express IαIp. [0010] As used herein, the term “chromatography” or “chromatography step” refers to a separation of one or more analytes in a mixture by passing the mixture in a solution or in a suspension through a medium in which the analytes of the mixture move at different rates. For example, a chromatography step can be size exclusion chromatography, ion-exchange chromatography, affinity chromatography, or dye-ligand chromatography. More specifically, a chromatography step, as described herein, can be performed to separate an IαIp from a biological material containing the IαIp. [0011] In this disclosure, the terms "comprises," "comprising," "containing" and "having" and the like can have the meaning ascribed to them in U.S. Patent law and can mean " includes," "including," and the like; "consisting essentially of” or "consists essentially" likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments. [0012] As used herein, the term “eluate” refers to a fraction containing an analyte material (e.g., IαIp) that is eluted from a medium (e.g., a support material) during a purification step (e.g., a chromatography step). An eluate may be released from the medium by applying an eluent to the medium, thereby releasing the analyte. More specifically, an eluate can refer to a fraction containing IαIp that has been released from a medium following application of an eluent (e.g., an elution buffer, such as a buffer containing a salt) to the medium. [0013] As used herein, the term “flow through” refers to a fraction of material or a volume of fluid that passes through a medium (e.g., a medium used for chromatography, such as an anion- exchange resin) without binding. Additional mobile phase (e.g., a fluid, such as buffer with a low (e.g., less than 50 mM salt) or no salt (e.g., sodium chloride)) can be added to ensure that one or more components of a mixture applied to a medium is fully loaded onto the medium, and to achieve initial or additional separation of an analyte (e.g., IαIp) from other components in the mixture. [0014] As used herein, the terms “inter-alpha inhibitor protein” and “IαIp,” and plural forms thereof, refer to multi-component glycoproteins in a family of structurally related serine protease inhibitors. IαIps have been shown to be important in the inhibition of an array of proteases including neutrophil elastase, plasmin, trypsin, chymotrypsin, Granzyme K, preprotein convertase, furin, cathepsin G, and acrosin. In human plasma, IαIps are found at relatively high concentrations (400-800 mg/L). Unlike other inhibitor molecules, this family of inhibitors typically includes a combination of polypeptide chains (light and heavy chains) covalently linked by a chondroitin sulfate chain. The heavy chains of IαIps (H1, H2, and H3) are also called hyaluronic acid (HA) binding proteins. The major forms of IαIps found in human plasma are inter-alpha-inhibitor (IαI), which contains two heavy chains (H1 and H2) and a single light chain (L), and pre-alpha-inhibitor (PαI), which contains one heavy (H3) and one light chain (L). Another IαIp is the light chain (also termed bikunin (bi-kunitz inhibitor) with two Kunitz domains), which is known to broadly inhibit plasma serine proteases. Another IαIp is the heavy chain-related molecule H4, which circulates in the blood without linkage to bikunin. Yet another IαIp is the heavy chain-related molecule H5. IαI and PαI present in the plasma fraction have an apparent molecular weight of between about 60 kDa to about 280 kDa. [0015] As used herein, the term “pharmaceutically acceptable excipient” means one or more compatible solid or liquid fillers, diluents, or encapsulating substances that are suitable for administration into a human. The excipient can contain an additive, such as a substance that enhances isotonicity and/or chemical stability. Such materials are non-toxic to recipients in the amounts and concentrations employed, and can include buffers, such as phosphate, citrate, succinate, acetate, lactate, tartrate, and other organic acids or their salts; tris- hydroxymethylaminomethane (Tris), bicarbonate, carbonate, and other organic bases and their salts; antioxidants, such as ascorbic acid; low molecular weight (for example, less than about ten residues) polypeptides, e.g., polyarginine, polylysine, polyglutamate and polyaspartate; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers, such as polyvinylpyrrolidone (PVP), polypropylene glycols (PPGs), and polyethylene glycols (PEGs); amino acids, such as glycine, glutamic acid, aspartic acid, histidine, lysine, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose, sucrose, dextrins or sulfated carbohydrate derivatives, such as heparin, chondroitin sulfate or dextran sulfate; polyvalent metal ions, such as divalent metal ions including calcium ions, magnesium ions and manganese ions; chelating agents, such as ethylenediamine tetraacetic acid (EDTA); sugar alcohols, such as mannitol or sorbitol; counterions, such as sodium or ammonium; and/or nonionic surfactants, such as polysorbates or poloxamers. Other additives may be also included, such as stabilizers, anti-microbials, inert gases, fluid and nutrient replenishers (i.e., Ringer’s dextrose), electrolyte replenishers, and the like, which can be present in conventional amounts. [0016] As used herein, the terms "prevent," "preventing," "prevention," "prophylactic treatment" and the like refer to reducing the probability of developing a disorder or condition in a subject, who does not have, but is at risk of or is susceptible to developing, a disease, disorder, or condition. [0017] As used herein, the term “processed” refers to a biological material that has been modified using one or more sample preparation steps (e.g., filtration, centrifugation, sedimentation, chromatography, etc.) prior to contacting the material to an anion-exchange resin. Other examples of processing include a decanting step, a clarification step, a freezing step, a drying step, an evaporation step, an extraction step, a filtration step, a precipitation step, or another purification or preparatory method known in the art. The processing step may remove up to, e.g., 10% or more (w/w) (e.g., 10-30% (w/w), such as 15%, 20%, 25%, or 30% (w/w) or more) of one or more substances from the biological material (e.g., a protein other than an IαIp). [0018] As used herein, the terms “purify,” “purifying,” “purification” and the like refer to one or more steps or processes of removing proteins (e.g., proteins other than an IαIp) and/or non- proteinaceous substances (e.g., phospholipids and nucleic acids) from a heterologous mixture (e.g., a biological material, such as blood or milk) containing IαIp and the other proteins and/or substances to produce a formulation containing an IαIp without the other proteins and/or substances present in the original mixture (e.g., a biological material) or in which the proteins other than IαIp and/or substances have been reduced by 40% or more by weight (e.g., 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97%, or 99% or more) relative to, e.g., a starting mixture (e.g., a biological material). Examples of proteins that can be removed from a mixture containing an IαIp include, but are not limited to, alpha-1 antitrypsin, C1-inhibitor, albumin, a globulin (including immunoglobulins, e.g., IgA, IgG (e.g., of intravenous Ig (IVIg), anti-D IgG, hepatitis B IgG, measles IgG, rabies IgG, tetanus IgG, and Varicella Zoster IgG), IgM, IgD, and IgE), fibrinogen (factor I), prothrombin (factor II), thrombin, anti-thrombin III, factor III, factor V, factor VII, factor VIII, factor IX, factor X, factor XI, factor XII, factor XIII, fibronectin, alpha-2 antiplasmin, urokinase, protein C, protein S, protein Z, protein Z-related protease inhibitor, plasminogen, tissue plasminogen activator, plasminogen activator inhibitor-1, plasminogen activator inhibitor-2, von Willebrand factor, factor H, prekallikrein, high-molecular-weight kininogen, and heparin cofactor II. [0019] As used herein, the term “pure” or “purity” refers to the extent to which an analyte has been isolated and is free of other components. In the context of proteins, purity of an isolated protein can be expressed with regard to the protein that is free of any contaminants (e.g., one or more unrelated proteins or other substances). For example, purity of an IαIp formulation indicates how much of the formulation is IαIp by total weight of the isolated material, which may be determined using, e.g., pure IαIp as a reference. A level of purity found in the disclosure can be 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, greater than 95%, or greater than 99% (w/w). A “pure” IαIp formulation of the disclosure can be greater than 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, or up to 70% pure by weight. A “substantially pure” IαIp formulation can be substantially free of contaminants or impurities, e.g., greater than 70%, 75%, 80%, 85%, 90%, 95%, or >99% purity by weight. In some embodiments, the level of contaminants or impurities is no more than about 20%, 15%, 12%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% by weight. Purity can be determined by detecting a level of a specific analyte (e.g., IαIp) using an immunoassay or other technique (e.g., MAb 69.26 – heparin-biotin sandwich ELISA, SDS/PAGE, and/or Western blot) and calculating a percentage of the analyte (w/w) relative to the total protein content (e.g., as determined by a total protein assay (e.g., bicinchoninic acid assay (BCA), Bradford assay, Biuret test, or another assay known in the art)). [0020] As used herein, the term “subject” refers to a mammal, including, but not limited to, a human or non-human mammal, such as a primate, bovine, equine, porcine, ovine, feline, or canine. The subject may be a patient. [0021] The term “substantially unprocessed,” as used herein, refers to a biological material that has been minimally modified, if at all, relative to the original source material (e.g., blood). For example, a substantially unprocessed biological material can retain the original content (e.g., the same proteins and/or substances and/or the same ratio of two or more proteins or substances) and/or the original characteristics (e.g., one or more biological activities) of the original source material. A biological material can be substantially unprocessed, such that any prior purification step(s) performed with the material removes less than 10% (w/w) (e.g., less than 0.1%-10% (w/w), such as less than 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, or 9% (w/w)) of one or more proteins or substances from the material. A substantially unprocessed biological material may be one that has not been modified using a sample preparation step (e.g., filtration, centrifugation, sedimentation, chromatography, etc.), in particular, for example, prior to contacting the biological material to an anion-exchange resin. [0022] As used herein, the phrase “specifically binds” refers to a binding reaction between an analyte (e.g., a protein, such as an IαIp) and a binding agent. A specific binding reaction is one that occurs between an analyte and a binding agent even in the presence of a heterogeneous population of proteins and other biological molecules (e.g., proteins other than IαIp in a biological material). Specific binding between an analyte (e.g., an IαIp) and a binding agent can be characterized by a Kd of less than about 1000 nM (e.g., between 1 pM and 1000 nM). An analyte (e.g., an IαIp) that does not specifically bind to a binding agent can be characterized by a Kd of greater than about 1000 nM (e.g., greater than 1 µM, 100 µM, 500 µM, or 1 mM). [0023] As used herein, the term “support” means any apparatus that contains an agent (e.g., an anion-exchange resin) that can be contacted with a material (e.g., a biological material) containing at least one analyte (e.g., an IαIp). A support may be a column, a membrane, a disc, a chip, or other apparatus for chromatography or affinity capture, examples of which are known in the art and described herein. [0024] As used herein, the term “treating” refers to reducing or ameliorating a disorder and/or one or more symptoms associated therewith. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder or symptoms associated therewith be completely eliminated. [0025] As used herein, the term “yield” refers to the relative amount of an analyte (e.g., IαIp) obtained after a purification step or process as compared to the amount of analyte in the starting material (e.g., the biological material) (w/w). The yield may be expressed as a percentage. In the context of the disclosure, the amount of analyte (e.g., IαIp) in the starting material and analyte obtained after the purification step can be measured using an immunoassay or assay (e.g., an anti-IαIp antibody (e.g., MAb 69.26) – heparin-biotin sandwich ELISA, SDS/PAGE, and/or Western blot). The methods of the disclosure can be used to produce a yield of purified IαIp of about 20% (w/w) or greater relative to the amount present in the original biological material. For example, the methods can be used to produce a yield of purified IαIp of about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 65%, 70%, 75%, 80%, 85%, or 90% (w/w) or greater. DETAILED DESCRIPTION OF THE INVENTION [0026] The invention features a liquid formulation comprising Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) and a buffer which is a citrate buffer and/or a histidine buffer, and wherein the formulation has a pH of about 5.0 to about 7.5. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of about 5.0 to about 7.5, and the formulation contains essentially no magnesium. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of about 5.3 to about 5.7, and the formulation contains between about 40 mM to about 110 mM magnesium. The invention also features a liquid formulation comprising Inter-alpha Inhibitor (IαI) and Pre- alpha Inhibitor (PαI) comprising significantly low amount of high MW species. [0027] In an exemplary embodiment, the liquid formulations described herein are not lyophilized. In an exemplary embodiment, the liquid formulations described herein do not contain a phosphate buffer. IαI and/or PαI [0028] In some embodiments, the liquid formulation comprises inter-alpha inhibitor (IαI) and/or pre-alpha inhibitor (PαI). In some embodiments, the liquid formulation comprises inter-alpha inhibitor (IαI) and pre-alpha inhibitor (PαI). In some embodiments, the liquid formulation comprises inter-alpha inhibitor (IαI) or pre-alpha inhibitor (PαI). In some embodiments, the liquid formulation comprises inter-alpha inhibitor (IαI) and not pre-alpha inhibitor (PαI). In some embodiments, the liquid formulation comprises pre-alpha inhibitor (PαI) and not inter- alpha inhibitor (IαI). [0029] In some embodiments, the liquid formulation comprises between 60% to 80% (w/w) IαI and between 20% to 40% (w/w) PαI. In some embodiments, the liquid formulation comprises between 60% to 70% (w/w) IαI and between 20% to 30% (w/w) PαI. In some embodiments, the liquid formulation comprises between 60% to 70% (w/w) IαI and between 30% to 40% (w/w) PαI. In some embodiments, the liquid formulation comprises between 62% to 72% (w/w) IαI and between 28% to 30% (w/w) PαI. In some embodiments, the liquid formulation comprises between 62% to 72% (w/w) IαI and between 28% to 38% (w/w) PαI. [0030] In some embodiments, essentially all of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 90% and about 99.5% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 80% and about 90% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 70% and about 80% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 68% and about 78% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 66% and about 76% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 64% and about 74% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 62% and about 72% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 60% and about 65% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 61% and about 66% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 62% and about 67% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 63% and about 68% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 64% and about 69% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 65% and about 70% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 66% and about 71% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 67% and about 72% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 68% and about 73% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 69% and about 74% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 59% and about 64% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 58% and about 63% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 57% and about 62% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 56% and about 61% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 55% and about 60% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 54% and about 59% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 54% and about 59% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 53% and about 58% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 52% and about 57% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 51% and about 56% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 50% and about 55% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 49% and about 54% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 48% and about 53% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 47% and about 52% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 46% and about 51% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 45% and about 50% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 60% and about 70% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 58% and about 68% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 56% and about 66% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 54% and about 64% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 52% and about 62% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 50% and about 60% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 48% and about 58% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 46% and about 56% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 44% and about 54% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 42% and about 52% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 40% and about 50% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 30% and about 40% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 20% and about 30% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 10% and about 20% (w/w) of the IαI and PαI present in the liquid formulation is IαI. In some embodiments, between about 1% and about 10% (w/w) of the IαI and PαI present in the liquid formulation is IαI. [0031] In some embodiments, essentially all of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 90% and about 99.5% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 80% and about 90% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 70% and about 80% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 60% and about 70% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 50% and about 60% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 40% and about 50% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 38% and about 48% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 36% and about 46% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 34% and about 44% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 32% and about 42% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 30% and about 35% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 31% and about 36% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 32% and about 37% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 33% and about 38% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 34% and about 39% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 35% and about 40% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 30% and about 40% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 28% and about 38% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 26% and about 36% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 24% and about 34% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 22% and about 32% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 30% and about 35% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 29% and about 34% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 28% and about 33% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 27% and about 32% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 26% and about 31% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 25% and about 30% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 24% and about 29% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 23% and about 28% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 22% and about 27% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 21% and about 26% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 20% and about 25% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 20% and about 30% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 10% and about 20% (w/w) of the IαI and PαI present in the liquid formulation is PαI. In some embodiments, between about 1% and about 10% (w/w) of the IαI and PαI present in the liquid formulation is PαI. IαI/PαI ratio [0032] In an exemplary embodiment, the IαI/PαI ratio is from about 1.5 to about 3.5. In an exemplary embodiment, the IαI/PαI ratio is from about 1.7 to about 3.3. In an exemplary embodiment, the IαI/PαI ratio is from about 2.0 to about 3.0. In an exemplary embodiment, the IαI/PαI ratio is from about 2.0 to about 2.25. In an exemplary embodiment, the IαI/PαI ratio is from about 2.0 to about 2.2. In an exemplary embodiment, the IαI/PαI ratio is from about 2.05 to about 2.15. In an exemplary embodiment, the IαI/PαI ratio is about 2.1. In an exemplary embodiment, the IαI/PαI ratio is from about 2.5 to about 3.0. In an exemplary embodiment, the IαI/PαI ratio is from about 2.6 to about 2.85. In an exemplary embodiment, the IαI/PαI ratio is from about 2.65 to about 2.85. In an exemplary embodiment, the IαI/PαI ratio is from about 2.7 to about 2.85. In an exemplary embodiment, the IαI/PαI ratio is from about 2.75 to about 2.85. In an exemplary embodiment, the IαI/PαI ratio is from about 2.75 to about 2.80. In an exemplary embodiment, the IαI/PαI ratio is about 2.78. In an exemplary embodiment, the IαI/PαI ratio is from about 2.6 to about 3.0. In an exemplary embodiment, the IαI/PαI ratio is from about 2.7 to about 3.0. In an exemplary embodiment, the IαI/PαI ratio is from about 2.8 to about 3.0. In an exemplary embodiment, the IαI/PαI ratio is from about 2.9 to about 3.0. In an exemplary embodiment, the IαI/PαI ratio is from about 2.85 to about 2.95. In an exemplary embodiment, the IαI/PαI ratio is from about 2.90 to about 2.95. In an exemplary embodiment, the IαI/PαI ratio is about 2.92. IαI and/or PαI concentration [0033] In an exemplary embodiment, the liquid formulations comprise between about 5 mg/mL and about 100 mg/mL of IαI and/or PαI. In an exemplary embodiment, the liquid formulations comprise between about 10 mg/mL and about 100 mg/mL of IαI and/or PαI. In an exemplary embodiment, the liquid formulations comprise between about 10 mg/mL and about 50 mg/mL of IαI and/or PαI. In an exemplary embodiment, the liquid formulations comprise between about 15 mg/mL and about 25 mg/mL of IαI and/or PαI. In an exemplary embodiment, the liquid formulations comprise between about 17 mg/mL and about 23 mg/mL of IαI and/or PαI. In an exemplary embodiment, the liquid formulations comprise between about 19 mg/mL and about 21 mg/mL of IαI and/or PαI. High MW species [0034] In an exemplary embodiment, less than 20% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 20% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 20% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 20% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 15% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 15% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 15% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 15% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 10% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 10% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 5% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 5% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.1% and about 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.5% and about 5% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.5% and about 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.5% and about 4% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 0.5% and about 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 1% and about 4% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 1% and about 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 2% and about 4% of total protein in the formulation comprises a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, between about 2% and about 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. Concentrations of impurities (Factor II, Factor X) [0035] In an exemplary embodiment, the formulation contains less than 0.03 mg/mL of Factor II (Prothrombin). In an exemplary embodiment, the formulation contains less than 0.02 mg/mL of Factor II (Prothrombin). In an exemplary embodiment, the formulation contains less than 0.01 mg/mL of Factor II (Prothrombin). In an exemplary embodiment, the formulation comprising less than 0.01 mg/mL of Factor X. In an exemplary embodiment, the formulation comprising less than 0.005 mg/mL of Factor X. In an exemplary embodiment, the formulation comprising less than 0.001 mg/mL of C4 binding protein. In an exemplary embodiment, the formulation comprising less than 1.5 µg/mL of Factor II and less than 0.8 µg/mL of Factor X. NaCl [0036] In an exemplary embodiment, the formulation further comprises essentially no NaCl. In an exemplary embodiment, the formulation further comprises between about 5 mM to about 110 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 40 mM to about 110 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 40 mM to about 60 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 90 mM to about 110 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 20 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 20 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 5 mM NaCl. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 5 mM NaCl. Sucrose [0037] In an exemplary embodiment, the formulation further comprises sucrose. In an exemplary embodiment, the formulation further comprises between about 1.5% (w/w) and about 2.5% (w/w) sucrose. In an exemplary embodiment, the formulation further comprises between about 1.8% (w/w) and about 2.2% (w/w) sucrose. In an exemplary embodiment, the formulation further comprises between about 1.9% (w/w) and about 2.1% (w/w) sucrose. In an exemplary embodiment, the formulation further comprises between about 1.8% (w/w) and about 2.0% (w/w) sucrose. Glycine [0038] In an exemplary embodiment, the formulation further comprises glycine. In an exemplary embodiment, the formulation further comprises between about 0.5% (w/w) and about 1.5% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 0.8% (w/w) and about 1.2% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 0.9% (w/w) and about 1.1% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 0.7% (w/w) and about 0.9% (w/w) glycine. Sucrose and Glycine [0039] In an exemplary embodiment, the formulation further comprises sucrose and glycine. In an exemplary embodiment, the formulation further comprises between about 1.5% (w/w) and about 2.5% (w/w) sucrose and between about 0.5% (w/w) and about 1.5% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 1.8% (w/w) and about 2.2% (w/w) sucrose and between about 0.8% (w/w) and about 1.2% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 1.9% (w/w) and about 2.1% (w/w) sucrose and between about 0.9% (w/w) and about 1.1% (w/w) glycine. In an exemplary embodiment, the formulation further comprises between about 1.8% (w/w) and about 2.0% (w/w) sucrose and between about 0.7% (w/w) and about 0.9% (w/w) glycine. Buffer in the formulation [0040] In an exemplary embodiment, the buffer is citrate or histidine. In an exemplary embodiment, the buffer is citrate. In an exemplary embodiment, the buffer is histidine. Buffer concentration in the formulation [0041] In an exemplary embodiment, the buffer concentration is between about 10 mM and about 30 mM. In an exemplary embodiment, the buffer concentration is between about 15 mM and about 25 mM. In an exemplary embodiment, the buffer concentration is between about 17 mM and about 23 mM. pH [0042] In an exemplary embodiment, the formulation has a pH of about 5.4 to about 6.6. In an exemplary embodiment, the formulation has a pH of about 5.5 to about 6.5. In an exemplary embodiment, the formulation has a pH of about 5.3 to about 5.7. In an exemplary embodiment, the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the formulation has a pH of about 6.3 to about 6.7. In an exemplary embodiment, the formulation has a pH of about 6.4 to about 6.6. Magnesium [0043] In an exemplary embodiment, the formulation further comprises essentially no magnesium. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 150 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 150 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 5 mM to about 110 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 40 mM to about 110 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 40 mM to about 60 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 90 mM to about 110 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 20 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 20 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 5 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 0 mM to about 1 mM magnesium. In an exemplary embodiment, the formulation further comprises between about 1 mM to about 5 mM magnesium. Stability [0044] In an exemplary embodiment, the formulation is stable. Further Embodiments [0045] In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of between about 5.0 to about 6.0, and the formulation contains between about 0 mM to about 1 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 0 mM to about 1 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 0 mM to about 1 mM magnesium. [0046] In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of between about 6.0 to about 7.0, and the formulation contains between about 0 mM to about 1 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of between about 6.3 to about 6.7, and the formulation contains between about 0 mM to about 1 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of about 6.5, and the formulation contains between about 0 mM to about 1 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 30 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI), wherein the formulation comprises less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa, wherein the formulation comprises a buffer, and wherein the formulation has a pH of about 5.0 to about 7.5. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 30 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI), wherein the formulation comprises less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa, wherein the formulation comprises a buffer which is a citrate buffer and/or a histidine buffer, and wherein the formulation has a pH of about 5.0 to about 7.5. [0047] In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 5 mM to about 110 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 40 mM to about 110 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 40 mM to about 110 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 40 mM to about 60 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre- alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 40 mM to about 60 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of between about 5.3 to about 5.7, and the formulation contains between about 90 mM to about 110 mM magnesium. In an exemplary embodiment, the liquid formulation comprises between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of about 5.5, and the formulation contains between about 90 mM to about 110 mM magnesium [0048] In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM citrate buffer, between about 40 mM to about 60 mM NaCl, essentially no magnesium, wherein the formulation has a pH of about 6.4 to about 6.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM citrate buffer, between about 40 mM to about 60 mM NaCl, sucrose, glycine, essentially no magnesium, wherein the formulation has a pH of about 6.4 to about 6.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM citrate buffer, between about 40 mM to about 60 mM NaCl, between about 1.5% and about 2.5% sucrose and between about 0.5% and about 1.5% glycine, essentially no magnesium, wherein the formulation has a pH of about 6.4 to about 6.6. [0049] In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl and between about 90 mM to about 110 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 90 mM to about 110 mM magnesium, sucrose, and glycine, wherein the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 90 mM to about 110 mM magnesium, and between about 1.5% and about 2.5% sucrose and between about 0.5% and about 1.5% glycine, wherein the formulation has a pH of about 5.4 to about 5.6. [0050] In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl and between about 40 mM to about 60 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 40 mM to about 60 mM magnesium, sucrose, and glycine, wherein the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, essentially no NaCl, between about 40 mM to about 60 mM magnesium, between about 1.5% and about 2.5% sucrose, and between about 0.5% and about 1.5% glycine, wherein the formulation has a pH of about 5.4 to about 5.6. [0051] In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, between about 90 mM to about 110 mM NaCl and between about 40 mM to about 60 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, between about 90 mM to about 110 mM NaCl, between about 40 mM to about 60 mM magnesium, sucrose, and glycine, wherein the formulation has a pH of about 5.4 to about 5.6. In an exemplary embodiment, the invention provides a liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of IαI and PαI, between about 17 mM and about 23 mM histidine buffer, between about 90 mM to about 110 mM NaCl, between about 40 mM to about 60 mM magnesium, between about 1.5% and about 2.5% sucrose, and between about 0.5% and about 1.5% glycine, wherein the formulation has a pH of about 5.4 to about 5.6. Methods of Purification [0052] In an exemplary embodiment, the IαIp can be purified according to methods known in the art, such as those described in PCT Pat App PCT/US04/36848, US Pat App 16/866,241, PCT Pat App PCT/US09/03291, US Pat App 15/439,328, PCT Pat App PCT/US14/12033, PCT Pat App PCT/US18/29436, and US Pat App 17/404,588, each of which is herein incorporated by reference in their entirety. Improved Methods of Purification [0053] In an exemplary embodiment, the IαI and the PαI can be purified according to a method below: [1] A method of purifying an IαI and a PαI from a biological material comprising: (a) applying the biological material comprising the IαI and the PαI to a strong anion exchanger and separating a flow through; (b) applying a wash buffer to the strong anion exchanger and separating a flow through; and (c) applying an elution buffer to the strong anion exchanger and collecting an eluate comprising the IαI and the PαI; wherein conductivity of the wash buffer is less than 15 mS/cm, wherein pH of the wash buffer is about 5.0 to about 5.5, wherein the elution buffer comprise about 350 mM to about 450 mM NaCl. [2] The method of [1], wherein conductivity of the wash buffer is less than 13 mS/cm, wherein pH of the wash buffer is about 5.2, wherein the elution buffer comprise about 400 mM NaCl. [3] A method of purifying an IαI and a PαI from a biological material comprising: (a) applying the biological material comprising the IαI and the PαI to a strong anion exchanger and separating a flow through; (b) applying a first wash buffer to the strong anion exchanger and separating a flow through; (c) applying a second wash buffer to the strong anion exchanger and separating a flow through; (d) applying a third wash buffer to the strong anion exchanger and separating a flow through; (e) applying an elution buffer to the strong anion exchanger and collecting an eluate comprising the IαI and the PαI; wherein conductivity of the second wash buffer is less than 15 mS/cm, wherein pH of the second wash buffer is about 5.0 to about 5.5, wherein pH of the first wash buffer and the second wash buffer is higher than the pH of the second wash buffer, wherein the elution buffer comprise about 350 mM to about 450 mM NaCl. [4] The method of [3], wherein conductivity of the second wash buffer is less than 13 mS/cm, wherein pH of the second wash buffer is about 5.2, wherein the elution buffer comprise about 400 mM NaCl. [5] The method of [4], wherein the first wash buffer comprise about 250 mM NaCl, wherein the pH of first wash buffer is about 7.2, wherein the second wash buffer comprise about 65 mM NaCl, wherein the third wash buffer comprise 200 mM NaCl, wherein the pH of the third wash buffer is 7.2. [6] The method of [1] to [5] further comprising polishing step. Characteristics of the formulation comprising the IαI and the PαI purified by improved methods of purification [0054] The formulation comprising an IαI and a PαI purified according to the improved methods of purification have beneficial features below: (i) In an exemplary embodiment, less than 10% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 5% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 4% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. In an exemplary embodiment, less than 3% of total protein in the formulation is a protein or a protein aggregate having molecular weight of more than 500 kDa. (ii) In an exemplary embodiment, more than 80% of the IαI and the PαI does not exist as a protein aggregates. In an exemplary embodiment, more than 85% of the IαI and the PαI does not exist as a protein aggregates. In an exemplary embodiment, more than 90% of the IαI and the PαI does not exist as a protein aggregates. (iii) In an exemplary embodiment, the formulation has an at least 20% higher trypsin inhibitory specific activity than known formulations comprising IαIps. In an exemplary embodiment, the formulation has an at least 25% higher trypsin inhibitory specific activity than known formulations comprising IαIps. In an exemplary embodiment, the formulation has an at least 30% higher trypsin inhibitory specific activity than known formulations comprising IαIps. In an exemplary embodiment, the formulation has an at least 40% higher trypsin inhibitory specific activity than known formulations comprising IαIps. In an exemplary embodiment, the formulation has an at least 50% higher trypsin inhibitory specific activity than known formulations comprising IαIps. [0055] In an exemplary embodiment, the invention provides a method of purifying inter-alpha inhibitor (IαI) and pre-alpha inhibitor (PαI) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b) contacting the product of the previous step with a second anion-exchange resin; c) polishing the product of the previous step, thereby producing purified IαI and PαI. In an exemplary embodiment, the blood product material is cryo-poor plasma. [0056] In an exemplary embodiment, the a) comprises: a1) contacting the blood product material in a loading butter to a first anion-exchange resin, under conditions in which IαI and PαI adhere to the resin and separate a flow-through; a2) contacting the product of a1) with an elution buffer in which IαI and PαI elute from the resin. In an exemplary embodiment, the first anion- exchange resin is a weak anion exchange resin. In an exemplary embodiment, the first anion- exchange resin comprises a DEAE moiety. In an exemplary embodiment, the first anion- exchange resin is DEAE-Sephadex A-50, Toyopearl-GigaCap DEAE-650M, or DEAE- Sepharose FF. [0057] In an exemplary embodiment, the b) comprises: b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which IαI and PαI adhere to the resin and separate a flow-through; b2) contacting the product of b1) with an elution buffer in which IαI and PαI elute from the resin. In an exemplary embodiment, wherein the second anion-exchange resin is a strong anion exchange resin. In an exemplary embodiment, the second anion-exchange resin comprises a quaternary ammonium moiety. In an exemplary embodiment, wherein the second anion-exchange resin is a weak anion exchange resin. In an exemplary embodiment, the second anion-exchange resin comprises a DEAE moiety. In an exemplary embodiment, wherein the second anion-exchange resin is TOYOPEARL GigaCap Q-650M, Capto Q, Poros XQ, Eshmuno Q, Chromalite MQ/F, POROS 50HQ, Q-PuraBead HF, Praesto Jetted Q35, DEAE-Sepharose, or Toyopearl-GigaCap DEAE- 650M. In an exemplary embodiment, wherein the second anion-exchange resin is DEAE- Sepharose or Toyopearl-GigaCap DEAE-650M. In an exemplary embodiment, wherein the second anion-exchange resin is TOYOPEARL GigaCap Q-650M, Capto Q, Poros XQ, Eshmuno Q, Chromalite MQ/F, POROS 50HQ, Q-PuraBead HF, Praesto Jetted Q35. In an exemplary embodiment, the second anion-exchange resin is TOYOPEARL GigaCap Q-650M. [0058] In an exemplary embodiment, the b1) further comprises: the ratio of total protein (in mg) in the product of the previous step to mL of resin is between about 15 mg/mL to about 100 mg/mL. In an exemplary embodiment, the b1) further comprises: the ratio of total protein (in mg) in the product of the previous step to mL of resin is between about 1 mg/mL to about 50 mg/mL, or between about 5 mg/mL to about 40 mg/mL, or between about 10 mg/mL to about 30 mg/mL. In an exemplary embodiment, the b1) further comprises: the ratio of total protein (in mg) in the product of the previous step to mL of resin is between about 15 mg/mL to about 25 mg/mL. [0059] In an exemplary embodiment, wherein the loading buffer in b1) comprises a conductivity of between about 10-22 mS/cm; a pH range of 6.0-8.0, and a NaCl concentration of between about 50 mM and about 220 mM. In an exemplary embodiment, wherein the loading buffer in b1) comprises a conductivity of between about 15 mS/cm and about 25 mS/cm, or between about 19 mS/cm and about 22 mS/cm. In an exemplary embodiment, wherein the loading buffer in b1) comprises a pH of about 7.0 to about 8.0. In an exemplary embodiment, wherein the loading buffer in b1) comprises a NaCl concentration of between about 0 mM and about 220 mM, or between about 150 mM to about 250 mM, or between about 180 mM to about 220 mM. [0060] In an exemplary embodiment, wherein the b1) further comprises, after contacting with a loading buffer, contacting with a first wash buffer and separating a flow-through, the first wash buffer has a conductivity of between about 15-25 mS/cm; a pH range of 6.0-8.0; a NaCl concentration of between about 150 mM and about 250 mM, and a column volume (CV) of between about 3 to about 10. In an exemplary embodiment, wherein the b1) further comprises, after the contacting with the loading buffer, contacting with a first wash buffer and separating a flow-through, the first wash buffer has a conductivity of between about 15-25 mS/cm; a pH range of 6.0-8.0; a NaCl concentration of between about 150 mM and about 250 mM, and a column volume (CV) of between about 3 to about 10. In an exemplary embodiment, the first wash buffer has a conductivity of between about 19-22 mS/cm. In an exemplary embodiment, the first wash buffer has a pH of between about 7.0 and about 7.5. In an exemplary embodiment, the first wash buffer has a NaCl concentration of between about 200 mM and about 250 mM. In an exemplary embodiment, the column volume is from about 3 to about 8, or about 5 to about 10, or between about 3 to about 5, or between about 5 to about 8. [0061] In an exemplary embodiment, wherein the b1) further comprises, after contacting with a first wash buffer, contacting with a second wash buffer and separating a flow-through, the second wash buffer has a conductivity of between about 0-15 mS/cm; a pH range of 4.2-6.2, a NaCl concentration of between about 0 mM and about 100 mM, and a column volume (CV) of between about 3 to about 10. In an exemplary embodiment, wherein the b1) further comprises, after the contacting with the first wash buffer, contacting with a second wash buffer and separating a flow-through, the second wash buffer has a conductivity of between about 0-15 mS/cm; a pH range of 4.2-6.2, a NaCl concentration of between about 0 mM and about 100 mM, and a column volume (CV) of between about 3 to about 10. In an exemplary embodiment, the second wash buffer has a conductivity of between about 0 mS/cm and about 15 mS/cm, or about 10 mS/cm to about 15 mS/cm, or about 12 mS/cm to about 13 mS/cm. In an exemplary embodiment, the second wash buffer has a NaCl concentration of between about 50 mM to about 80 mM, or about 60 mM to about 70 mM. In an exemplary embodiment, the second wash buffer has a NaCl concentration of between about 100 mM to about 200 mM, or about 140 mM to about 160 mM. In an exemplary embodiment, the second wash buffer has a pH of about 5.0 to about 5.4. In an exemplary embodiment, the second wash buffer has a pH of about 4.7 to about 5.7. In an exemplary embodiment, the second wash buffer has a pH of about 4.2 to about 6.2. In an exemplary embodiment, the column volume is from about 3 to about 8, or about 5 to about 10, or between about 3 to about 5, or between about 5 to about 8. [0062] In an exemplary embodiment, the invention provides a method of purifying inter-alpha inhibitor (IαI) and pre-alpha inhibitor (PαI) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which IαI and PαI adhere to the resin and separate a flow-through; b2) contacting the product of b1) with an elution buffer in which IαI and PαI elute from the resin; c) polishing the product of the previous step, thereby producing purified IαI and PαI. In an exemplary embodiment, the invention provides a method of purifying inter-alpha inhibitor (IαI) and pre-alpha inhibitor (PαI) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which IαI and PαI adhere to the resin and separate a flow-through, and then contacting with a first wash buffer and separating a flow-through, and then contacting with a second wash buffer and separating a flow-through, wherein the second wash buffer has a conductivity of between about 0 mS/cm and about 15 mS/cm, or about 10 mS/cm to about 15 mS/cm, or about 12 mS/cm to about 13 mS/cm; and b2) contacting the product of b1) with an elution buffer in which IαI and PαI elute from the resin; c) polishing the product of the previous step, thereby producing purified IαI and PαI. In an exemplary embodiment, the invention provides a method of purifying inter-alpha inhibitor (IαI) and pre-alpha inhibitor (PαI) from a blood product material, said method comprising: a) contacting the blood product material with a first anion-exchange resin; b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which IαI and PαI adhere to the resin and separate a flow-through, and then contacting with a first wash buffer and separating a flow- through, and then contacting with a second wash buffer and separating a flow-through, wherein the second wash buffer has a conductivity of between about 0 mS/cm and about 15 mS/cm, or about 10 mS/cm to about 15 mS/cm, or about 12 mS/cm to about 13 mS/cm, and the second wash buffer has a pH of about 4.2 to about 6.2, or about 4.7 to about 5.7, or about 5.0 to about 5.4; and b2) contacting the product of b1) with an elution buffer in which IαI and PαI elute from the resin; c) polishing the product of the previous step, thereby producing purified IαI and PαI. [0063] In an exemplary embodiment, wherein the b1) further comprises, after the contacting with the second wash buffer, contacting with a third wash buffer and separating a flow-through, the third wash buffer has a conductivity of between about 0-20 mS/cm, a pH range of 6.5-7.5, a NaCl concentration of between about 0 mM and about 200 mM, and a column volume (CV) of between about 3 to about 10. In an exemplary embodiment, the third wash buffer has a NaCl concentration of between about 50 mM to about 80 mM, or about 60 mM to about 70 mM. In an exemplary embodiment, the third wash buffer has a pH is from about 4.7 to about 5.7, or from about 4.2 to about 6.2 or from about 7.0 to about 7.5. [0064] In an exemplary embodiment, the elution buffer in b2) has a conductivity of between about 35-40 mS/cm; a pH range of 6.5-7.5, and a NaCl concentration of between about 350 mM and about 450 mM. In an exemplary embodiment, the elution buffer in b2) has a conductivity of between about 65-80 mS/cm; a pH range of 6.5-7.5, and a NaCl concentration of between about 700 mM and about 800 mM. Assays [0065] Therapeutic efficacy of the formulations may be monitored, e.g., by methods known in the art, to determine pre- and post-treatment levels of IαIps (e.g., IαI, PαI, a heavy chain (e.g., H1, H2, H3, H4, and/or H5), a light chain (e.g., bikunin), or a combination thereof). The level of IαIps and IαIp-related biomarkers can be detected and/or measured e.g., by gas phase ion spectrometry methods, optical methods, electrochemical methods, atomic force microscopy, radio frequency methods, surface plasmon resonance, ellipsometry, and immunological methods. Sedimentation rate can be measured using standard clinical tests (e.g., blood tests). [0066] An immunoassay can be used to detect and analyze IαIps (e.g., IαI, PαI, a heavy chain (e.g., H1, H2, H3, H4, and/or H5), a light chain (e.g., bikunin), or a combination thereof) and/or other biomarker protein levels in a sample. The immunoassay can include: (a) providing an antibody that specifically binds to IαI and/or PαI; (b) contacting a sample with the antibody; and (c) detecting the presence of a complex of the antibody bound to the proteins in the sample. Suitable antibodies for use in an immunoassay to detect IαIps include, MAb 69.31, MAb 69.26, anti-IαIp polyclonal antibody, and anti-bikunin monoclonal or polyclonal antibody. [0067] Total Protein in a solution can be measured with a assay such as spectroscopic like UV280, or colorimetry based on BCA (Bicinchoninic acid assay), Biuret assay or Bradford. [0068] The following examples are intended to illustrate, rather than limit, the invention. EXAMPLES Example 1. Stability study of an IαI and/or PαI liquid formulation [0069] The liquid formulations comprising IαI and/or PαI were prepared (Table 1) by dialyzing an IαI and/or PαI-containing Bulk Drug Substance (BDS) against the buffers shown in Table 1. Depending on the protein content the dialyzed solutions were either diluted with the same buffer or concentrated with Vivaspin ultrafiltration tubes to a protein concentration of around 20 mg/mL. Afterwards the solutions were sterile-filtered and filled into glass vials, closed with a rubber stopper and an Aluminum-cap. The vials were stored at 2-8°C over 6 months and the stability was investigated at start, 1 month, 2 months, 4 months and 6 months with biochemical and bio-physical methods. Table 1. List of IαI and/or PαI liquid formulation for stability study
Figure imgf000028_0001
Fig.1 Turbidity [0070] Turbidity was investigated at start, 1 month, 2 months, 4 months, and 6 months (Figure 1). The turbidity of the 5 formulations differed from one another but did not show any relevant change over 6 months at 2-8°C. Visual appearance (particle formation) also did not change over 6 months at 2-8°C. Fig.2 SEC aggregation [0071] Size-Exclusion Chromatography (SEC) main peaks were investigated over 6 months (Figure 2). The lowest decrease of SEC-main peaks (IaI + PaI) was for A, followed by B at 2- 8°C after 6 months (Figure 2).
Example 2. Process Description [0072] Frozen plasma units were pooled for thawing at a maximum temperature of 6°C. Liquid solid separation of the thawed plasma pool was performed through centrifugation. The cryo-poor plasma centrifugate was collected and further processed. [0073] 0.5 g of an ion exchange gel (DEAE-Sephadex) was added per liter centrifugate. After sedimentation the protein/complex was separated from residual supernatant by filtration. The protein/DEAE-Sephadex complex was washed and the protein was subsequently eluted from the DEAE-Sephadex with a sodium chloride solution. The intermediate was stored frozen until further use. [0074] After thawing, the product was filtered through a membrane filter. The filtrate and the post wash were collected for further processing. [0075] A stock solution of Polysorbate 80 and Tri-N-butyl phosphate was prepared and was added while stirring to a calculated final concentration of 1.0% Polysorbate 80 and 0.30% Tri-N- butyl phosphate, respectively. After addition, the solution was stirred until complete homogenization and subsequently filtered through a membrane filter. [0076] The temperature during S/D treatment was maintained at 19.0 to 25.0°C. The time required was 60 minutes. After incubation, the solution was diluted to a target conductivity of 20 ± 5 mS/cm with purified water and pH was adjusted to 7.2 ± 0.2 by addition of HCl. [0077] Removal of the S/D-mixture was performed by strong anion exchange chromatography with TOYOPEARL GigaCap Q-650M resin. [0078] The S/D treated and filtered protein solution was loaded onto an equilibrated TOYO PEARL GigaCap Q-650M column. The resin was washed with wash buffer 1 (20 mM Tris, 250 mM sodium chloride, pH 7.2 ± 0.2) and wash buffer 2 (75 mM glycine, 100 mM sodium acetate 65 mM sodium chloride, pH 5.2 ± 0.2). The equilibration buffer (20 mM Tris, 200 mM sodium chloride, pH 7.2 ± 0.2) was taken as the third wash buffer. Proteins were eluted with Q elution buffer (20 mM Tris, 400 mM sodium chloride, pH 7.2 ± 0.2). The eluate was collected in a jacketed stainless-steel vessel for further processing. [0079] The eluate of the Q column was diluted at a ratio of 1:2 (1+1) with purified water prior to EtoxiClear loading. [0080] The diluted solution was loaded onto the equilibrated EtoxiClear column. After loading was completed, the column was washed with wash buffer 1 at pH 5.2 (75 mM glycine, 0.1 M AcOH, 200 mM NaCl), followed by washing the column using wash buffer 2 (20 mM Tris, 300 mM sodium chloride 7.2 ± 0.2). [0081] IAIP was then eluted with the EtoxiClear elution buffer (20 mM Tris, 500 mM sodium chloride, pH 7.2 ± 0.2). [0082] The IAIP eluate solution was diluted 1:2 (1+1) with purified water. [0083] The diluted IAIP eluate was subsequently passed through a 20 nm filter. The pressure during filtration was controlled to not more than 0.9 bar differential pressure. [0084] The nanofiltrate was concentrated to a protein concentration of 15 mg/mL through ultrafiltration. The UF membrane used had a nominal molecular weight cut off (NMWCO) of 50,000 daltons or lower. The concentrate was diafiltered against liquid formulation (e.g., liquid formulations described in Table 1). The minimum exchange volume was 10 times of the original concentrate volume. Throughout the ultrafiltration/diafiltration operation, the solution was maintained at 4 to 25°C. After diafiltration, the solution was concentrated to a protein concentration of minimum 2.5% w/v. The solution temperature was maintained at 4°C to 25°C. Example 3. Process Description [0085] Frozen plasma units were pooled for thawing at a maximum temperature of 6°C. Liquid solid separation of the thawed plasma pool was performed through centrifugation. The cryo-poor plasma centrifugate was collected and further processed. [0086] 0.5 g of an ion exchange gel (DEAE-Sephadex) was added per liter centrifugate. After sedimentation the protein/complex was separated from residual supernatant by filtration. The protein/DEAE-Sephadex complex was washed, and protein was subsequently eluted from the DEAE-Sephadex with a sodium chloride solution. The intermediate was stored frozen until further use. [0087] After thawing, the product was filtered through a membrane filter. The filtrate and the post wash were collected for further processing. [0088] A stock solution of Polysorbate 80 and Tri-N-butyl phosphate was prepared in a separate vessel and was added while stirring to a calculated final concentration of 1.0% Polysorbate 80 and 0.30% Tri-N-butyl phosphate, respectively. After addition, the solution was stirred until complete homogenization and subsequently filtered through a membrane filter. [0089] The temperature during S/D treatment was maintained at 19.0 to 25.0°C. The time required was 60 minutes. After incubation, the solution was diluted to a target conductivity of 20 ± 5 mS/cm with purified water and the pH was adjusted to 7.2 ± 0.2 by addition of HCl. [0090] Removal of the S/D-mixture was performed by strong anion exchange chromatography with TOYOPEARL GigaCap Q-650M resin. [0091] The S/D treated and filtered protein solution was loaded onto an equilibrated TOYO PEARL GigaCap Q-650M column. The resin was washed with wash buffer 1 (20 mM Tris, 250 mM sodium chloride, pH 7.2 ± 0.2) and wash buffer 2 (75 mM glycine, 100 mM acetic acid 150 mM sodium chloride, pH 5.2 ± 0.2). The Q equilibration buffer (20 mM Tris, 200 mM sodium chloride, pH 7.2 ± 0.2) was taken as the third wash buffer. Proteins were eluted with Q elution buffer (20 mM Tris, 750 mM sodium chloride, pH 7.2 ± 0.2). The eluate was collected in a jacketed stainless-steel vessel for further processing. [0092] The Q eluate was diluted at a ratio of 1:4 (1+3) with purified water or with buffer matrix without salt. [0093] The diluted solution was loaded onto the equilibrated EtoxiClear column. After loading was completed, the column was washed with wash buffer 1 at pH 5.2 (75 mM glycine, 0.1 M AcOH, 200 mM NaCl) followed by washing the column using wash buffer 2 (20 mM Tris, 300 mM sodium chloride 7.2 ± 0.2). [0094] IAIP was then eluted with EtoxiClear elution buffer (20 mM Tris, 500 mM sodium chloride 7.2 ± 0.2). [0095] The IAIP eluate solution was diluted 1:2 (1+1) with purified water. [0096] The diluted IAIP eluate was subsequently passed through a 20 nm filter. The pressure during filtration was controlled to not more than 0.9 bar differential pressure. [0097] The nanofiltrate was concentrated to a protein concentration of 15 mg/mL through ultrafiltration. The UF membrane used had a nominal molecular weight cut off (NMWCO) of 50,000 daltons or lower. [0098] The concentrate was diafiltered against liquid formulation (e.g., liquid formulations described in Table 1). The minimum exchange volume was 10 times of the original concentrate volume. Throughout the ultrafiltration/diafiltration operation, the solution was maintained at 4 to 25°C. [0099] After diafiltration, the solution was concentrated to a protein concentration of minimum 2.5% w/v. The solution temperature was maintained at 4°C to 25°C. Example 4. Process Description [0100] Frozen plasma units were pooled for thawing at a maximum temperature of 6°C. Liquid solid separation of the thawed plasma pool was performed through centrifugation. The cryo-poor plasma centrifugate was collected and further processed. [0101] 0.5 g of an ion exchange gel (DEAE-Sephadex) was added per liter centrifugate. After sedimentation the protein/complex was separated from residual supernatant by filtration. The protein/DEAE-Sephadex complex was washed and protein was subsequently eluted from the DEAE-Sephadex with a sodium chloride solution. The intermediate was stored frozen until further use. [0102] After thawing, the product was filtered through a membrane filter. The filtrate and the post wash were collected for further processing. [0103] A stock solution of Polysorbate 80, Tri-N-butyl phosphate and Triton X-100 reduced was prepared and added to the thawed product while stirring to a calculated final concentration of 0.30% Polysorbate 80 and 0.30% Tri-N-butyl phosphate, and 1% Triton X-100 reduced respectively. After addition, the solution was stirred until complete homogenization (for at least 30 minutes) and subsequently filtered through a membrane filter. [0104] The temperature during S/D treatment was maintained at 19.0 to 25.0°C. The time required was a minimum of 120 minutes. After incubation, the solution was diluted to a target conductivity of 22.5 ± 2.5 mS/cm with Q equilibration buffer (20 mM Tris-HCl, 200 mM NaCl, pH 7.2 ±0.1) and pH was adjusted to 7.2 ± 0.1 by addition of HCl. The conductivity of Q equilibration buffer was 21 ± 2 mS/cm. [0105] Removal of the S/D-mixture was performed by strong anion exchange chromatography with TOYOPEARL GigaCap Q-650M resin. [0106] The S/D treated and filtered protein solution was loaded at a ratio of approximately 20 mg total protein per mL resin at a flow rate of 120 cm/h onto an equilibrated TOYOPEARL GigaCap Q-650M column. This column had a bed height of 22 cm and a diameter of 14 cm. The volume of the column was 3.4 L and the residence time was 11 min. The resin was washed with approximately 8 CV of wash buffer 1 (20 mM Tris-HCl, 250 mM sodium chloride, pH 7.2 ± 0.1, conductivity 26 ±2 mS/cm) and with approximately 8 CV of wash buffer 2 (75 mM glycine, 100 mM acetic acid 150 mM sodium chloride, 92.5 mM sodium hydroxide, pH 5.2 ± 0.1, conductivity 20 ± 2 mS/cm). The Q equilibration buffer with approximately 5 CV (20 mM Tris-HCl, 200 mM sodium chloride, pH 7.2 ± 0.1, conductivity 21 ±2 mS/cm) was taken as the third wash buffer. Proteins were eluted with Q elution buffer (20 mM Tris-HCl, 750 mM sodium chloride, pH 7.2 ± 0.1, conductivity 66 ± 2 mS/cm). The eluate was collected in a jacketed stainless-steel vessel for further processing. [0107] The eluate from the Q column was diluted at a ratio of 1:4 (1+3) with dilution buffer (10 mM Tris-HCl, pH 7.2 ± 0.1, conductivity 1 ± 0.5 mS/cm). [0108] The diluted solution was loaded at a ratio of approximately 10 mg total protein per mL resin at a flow rate of 120 cm/h onto the equilibrated EtoxiClear column. This column had a bed height of 10.5 cm and a diameter of 14 cm. The volume of the column was 1.6 L and the residence time was 5.25 min. After loading was completed, the column was washed with approximately 8 CV with wash buffer 1 at pH 5.2 (75 mM glycine, 100 mM acetic acid, 200 mM sodium chloride, 92.5 mM NaOH, pH 5.2 ± 0.1, conductivity 25 ± 2 mS/cm) followed by washing the column with approximately 5 CV of wash buffer 2 (20 mM Tris-HCl, 300 mM sodium chloride 7.2 ± 0.1, conductivity 30 ± 2 mS/cm). [0109] IAIP was then eluted with approximately 3 CV with EtoxiClear elution buffer (20 mM Tris-HCl, 500 mM sodium chloride 7.2 ± 0.1, conductivity 47 ±2 mS/cm). [0110] The IAIP eluate solution was diluted 1:2 (1+1) with dilution buffer (10 mM Tris-HCl, pH 7.2 ± 0.1, 1 ± 0.5 mS/cm). [0111] The diluted IAIP eluate was subsequently passed through a 20 nm filter. A clarifying filter was connected in series to protect the nanofilter. The pressure during filtration was controlled to not more than 0.9 bar differential pressure. [0112] The nanofiltrate was concentrated to a protein concentration of 15 mg/mL through ultrafiltration. The UF membrane used had a nominal molecular weight cut off (NMWCO) of 30,000 daltons. [0113] The concentrate was diafiltered against a diafiltration buffer which resembled liquid formulation. The exchange volume was 10 times of the original concentrate volume. Throughout the ultrafiltration/diafiltration operation, the solution was maintained at 4 to 25°C. [0114] After diafiltration, the solution was concentrated to a protein concentration of minimum 2.5% w/v. The solution temperature was maintained at 4°C to 25°C. Example 5. IαI/PαI & Dimer & Multimer Aggregate Analysis by HP-SEC [0115] The product of Example 4 was analyzed for dimer and multimer aggregates of IAIP using high performance Size-Exclusion Chromatography (HP-SEC). Dimer and multimer aggregates of IαI and PαI have a molecular weight of more than 500 kDa. The % of product with a molecular weight of more than 500 kDa (Peak 1) was 3.61%. The % of product with a molecular weight of approximately 360 kDa (Peak 2, IαI) was 63.87%. The % of product with a molecular weight of approximately 250 kDa (Peak 3, PαI) was 30.47%. The % of product with a molecular weight of approximately 100 kDa, 60 kDa, and less than 50 kDa (Peaks 4, 5, 6, respectively) was 1.29%, 0.51%, and 0.25% respectively (see Table 2). [0116] The sum % area of Peaks 2 & 3 representing the purity of IaI and PaI was 94.34%. The ratio of the % area of Peak 2 (IaI) to peak 3 (PaI) was 2.10. The final container concentration was 26.30 mg/mL total protein. Table 2 HP-SEC results of IAIP concentrate
Figure imgf000035_0001
Example 6. Wash Buffer Conductivity Effect on Turbidity [0117] The effect of wash buffer conductivity in the Anion Exchange Chromatography step on product turbidity was examined. [0118] Cryo-poor plasma centrifugates were prepared and main steps were processed as described in Example 4. S/D treatment was performed using a final concentration of 1% Polysorbate 80 and 0.30% Tri-N-butyl phosphate. No additional filtration after S/D reagent addition was performed. S/D treated and filtered protein solutions were loaded onto an equilibrated TOYOPEARL GigaCap Q-650M column and were washed with wash buffer 1 as described in Example 4. Subsequent wash 2 was performed by applying wash buffer 2 with a conductivity of either 13 mS/cm or 20 mS/cm. In addition, a 2-step Q elution was performed with 400 mM and 550 mM NaCl respectively, whereas main IaI and PaI elution occurred in the first eluate with 400 mM NaCl. The turbidity of the Q eluates at 400 mM NaCl either washed with wash buffer 2 at 13 mS/cm or 20 mS/cm were analyzed by comparing online measured intensities of the UV350 wavelength (indicating turbidity) of the respective samples and visual inspection (Figure 3). The sample with the least turbidity (Figure 3a) was subjected to a wash buffer with a conductivity of 13 mS/cm. Figure 3. Visual turbidity check. a) Q eluate (400 mM NaCl) washed with wash buffer 2 with 13 mS/cm showed no turbidity; b) in corresponding Q eluate washed with wash buffer 2 with 20 mS/cm turbidity was observed.

Claims

What is claimed: 1. A liquid formulation which is: a) comprising between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a citrate buffer, wherein the formulation has a pH of about 5.0 to about 7.5, and the formulation contains essentially no magnesium; or b) comprising between about 10 mg/mL and about 100 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI) in a histidine buffer, wherein the formulation has a pH of about 5.3 to about 5.7, and the formulation contains between about 40 mM to about 110 mM magnesium.
2. The formulation of claim 1, wherein the formulation has a pH of about 5.4 to about 5.6.
3. The formulation of claim 1, wherein the formulation has a pH of about 6.4 to about 6.6.
4. The formulation of claim 1, wherein the formulation comprises between about 65% and about 70% (w/w) IαI.
5. The formulation of claim 1, wherein the formulation comprises between about 30% and about 35% (w/w) PαI.
6. The formulation of a preceding claim, having between about 15 mg/mL and about 25 mg/mL of IαI and PαI.
7. The formulation of a preceding claim, having between about 17 mg/mL and about 23 mg/mL of IαI and PαI.
8. The formulation of a preceding claim, having between about 19 mg/mL and about 21 mg/mL of IαI and PαI.
9. The formulation of claims 1-8, wherein the formulation further comprises essentially no magnesium.
10. The formulation of claims 1-8, wherein the formulation further comprises between about 5 mM to about 110 mM magnesium.
11. The formulation of claims 1-8, wherein the formulation further comprises between about 40 mM to about 110 mM magnesium.
12. The formulation of claims 1-8, wherein the formulation further comprises between about 40 mM to about 60 mM magnesium.
13. The formulation of claims 1-8, wherein the formulation further comprises between about 90 mM to about 110 mM magnesium.
14. The formulation of a preceding claim, wherein the formulation further comprises essentially no NaCl.
15. The formulation of claims 1-13, wherein the formulation further comprises between about 5 mM to about 110 mM NaCl.
16. The formulation of claims 1-13, wherein the formulation further comprises between about 40 mM to about 60 mM NaCl.
17. The formulation of claims 1-13, wherein the formulation further comprises between about 40 mM to about 110 mM NaCl.
18. The formulation of claims 1-13, wherein the formulation further comprises between about 90 mM to about 110 mM NaCl.
19. The formulation of a preceding claim, wherein the buffer concentration is between about 10 mM and about 30 mM.
20. The formulation of a preceding claim, wherein the buffer concentration is between about 15 mM and about 25 mM.
21. The formulation of a preceding claim, wherein the buffer concentration is between about 17 mM and about 23 mM.
22. The formulation of a preceding claim, wherein the buffer is citrate and does not contain histidine.
23. The formulation of any one of claims 1-21, wherein the buffer is histidine and does not contain citrate.
24. A liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI), between about 17 mM and about 23 mM citrate buffer, between about 40 mM to about 60 mM NaCl, essentially no magnesium, wherein the formulation has a pH of about 6.4 to about 6.6.
25. A liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI), between about 17 mM and about 23 mM histidine buffer, essentially no NaCl and between about 90 mM to about 110 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6.
26. A liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI), between about 17 mM and about 23 mM histidine buffer, essentially no NaCl and between about 40 mM to about 60 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6.
27. A liquid formulation comprising between about 17 mg/mL and about 23 mg/mL of Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI), between about 17 mM and about 23 mM histidine buffer, between about 90 mM to about 110 mM NaCl and between about 40 mM to about 60 mM magnesium, wherein the formulation has a pH of about 5.4 to about 5.6.
28. The formulation of a preceding claim, wherein the formulation further comprises sucrose and glycine.
29. The formulation of a preceding claim, wherein the formulation further comprises between about 1.5% and about 2.5% sucrose and between about 0.5% and about 1.5% glycine.
30. The formulation of a preceding claim, wherein the formulation further comprises between about 1.8% and about 2.2% sucrose and between about 0.8% and about 1.2% glycine.
31. The formulation of a preceding claim, wherein the formulation further comprises between about 1.9% and about 2.1% sucrose and between about 0.9% and about 1.1% glycine.
32. The formulation of a preceding claim, wherein the formulation further comprises between about 1.8% and about 2.0% sucrose and between about 0.7% and about 0.9% glycine.
33. A liquid formulation comprising Inter-alpha Inhibitor (IαI) and Pre-alpha Inhibitor (PαI), wherein less than 5% of total protein in the formulation comprises a protein or a protein aggregate having a molecular weight of more than 500 kDa.
34. The formulation of claim 33, wherein the IαI/PαI ratio in the formulation is from about 2.0 to about 3.0.
35. A method of purifying inter-alpha inhibitor (IαI) and pre-alpha inhibitor (PαI) from cryo-poor plasma, said method comprising: a1) contacting the processed or substantially unprocessed cryo-poor plasma in a loading butter to a first anion-exchange resin, under conditions in which IαI and PαI adhere to the resin and separate a flow-through; a2) contacting the product of a1) with an elution buffer in which IαI and PαI elute from the resin; b1) contacting the product of the previous step in a loading buffer with a second anion-exchange resin, under conditions in which IαI and PαI adhere to the resin and separate a flow-through, wherein the b1) further comprises the ratio of total protein (in mg) in the product of the previous step to ml of resin is between about 15 to about 100, wherein the loading buffer in b1) comprises a conductivity of between about 10-22 mS/cm, a pH range of 6.0-8.0, and a NaCl concentration of between about 50 mM and about 220 mM, wherein the b1) further comprises, after the contacting with the loading buffer, contacting with a first wash buffer and separating a flow-through the first wash buffer has a conductivity of between about 15- 25 mS/cm, a pH range of 6.0-8.0, a NaCl concentration of between about 150 mM and about 250 mM, and a column volume (CV) of between about 3 to about 10, wherein the b1) further comprises, after the contacting with the first wash buffer, contacting with a second wash buffer and separating a flow-through the second wash buffer has a conductivity of between about 0-15 mS/cm; a pH range of 4.2-6.2, a NaCl concentration of between about 0 mM and about 100 mM, and a column volume (CV) of between about 3 to about 10, wherein the b1) further comprises, after the contacting with the second wash buffer, contacting with a third wash buffer and separating a flow-through the third wash buffer has a conductivity of
between about 0-20 mS/cm; a pH range of 6.5-7.5, a NaCl concentration of between about 0 mM and about 200 mM, and a column volume (CV) of between about 3 to about 10; b2) contacting the product of b1) with an elution buffer in which IαI and PαI elute from the resin, wherein the elution buffer in b2) has a conductivity of between about 35-40 mS/cm; a pH range of 6.5-7.5, and a NaCl concentration of between about 350 mM and about 450 mM; thereby producing purified IαI and PαI.
36. The method of claim 35, wherein the first anion-exchange resin is a weak anion exchange resin.
37. The method of claim 35, wherein the first anion-exchange resin comprises a DEAE moiety.
38. The method of claim 35, wherein the first anion-exchange resin is DEAE- Sephadex A-50, Toyopearl-GigaCap DEAE-650M, or DEAE-Sepharose FF.
39. The method of claim 35, wherein the second anion-exchange resin is a strong anion exchange resin or a weak anion-exchange resin.
40. The method of claim 35, wherein the second anion-exchange resin comprises a quaternary ammonium moiety or a DEAE moiety.
41. The method of claim 35, wherein the second anion-exchange resin is TOYOPEARL GigaCap Q-650M, Capto Q, Poros XQ, Eshmuno Q, Chromalite MQ/F, POROS 50HQ, Q-PuraBead HF, Praesto Jetted Q35, DEAE-Sepharose, or Toyopearl-GigaCap DEAE- 650M.
PCT/IB2022/061044 2021-11-16 2022-11-16 Inter-alpha inhibitor protein formulations WO2023089503A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163280057P 2021-11-16 2021-11-16
US63/280,057 2021-11-16

Publications (1)

Publication Number Publication Date
WO2023089503A1 true WO2023089503A1 (en) 2023-05-25

Family

ID=84364001

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2022/061044 WO2023089503A1 (en) 2021-11-16 2022-11-16 Inter-alpha inhibitor protein formulations

Country Status (1)

Country Link
WO (1) WO2023089503A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7932365B2 (en) * 2003-11-08 2011-04-26 Pro Thera Biologics, Llc Preparation and composition of inter-alpha inhibitor proteins from human plasma for therapeutic use
US9758570B2 (en) * 2008-05-28 2017-09-12 Prothera Biologics, Inc. Preparation and composition of inter-alpha inhibitor proteins from blood

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7932365B2 (en) * 2003-11-08 2011-04-26 Pro Thera Biologics, Llc Preparation and composition of inter-alpha inhibitor proteins from human plasma for therapeutic use
US9758570B2 (en) * 2008-05-28 2017-09-12 Prothera Biologics, Inc. Preparation and composition of inter-alpha inhibitor proteins from blood

Similar Documents

Publication Publication Date Title
US20090318674A1 (en) Process for purification of antibodies
EP2748193B1 (en) Method for reducing the thromboembolic potential of a plasma-derived immunoglobulin composition
EP2945962B1 (en) Methods for isolating blood products from an inter-alpha inhibitor protein-depleted blood product material
US20020099174A1 (en) Filtration of plasma mixtures using cellulose-based filter aids
EP2640413B1 (en) A process for reduction and/or removal of fxi and fxia from solutions containing said coagulation factors
WO2023089503A1 (en) Inter-alpha inhibitor protein formulations
US20230416342A1 (en) Methods for purifying inter-alpha inhibitor proteins
JPH0739375A (en) Purification of plasminogen
US20220363715A1 (en) Process for Purifying C1-INH
US20210087224A1 (en) Compositions and methods for generating modified cryo poor plasma
AU2015268579A1 (en) Method for reducing the thromboembolic potential of a plasma-derived immunoglobulin composition
KR20230148961A (en) Method for purifying immunoglobulin
EP2699594B1 (en) Process for the preparation of a virus-inactivated fv concentrate starting from human plasma, scalable to industrial level
US20230142480A1 (en) A method to produce an immunoglobulin preparation from c-1 inhibitor depleted plasma
AU710566B2 (en) Filtration of plasma mixtures using cellulose-based filter aids

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22813736

Country of ref document: EP

Kind code of ref document: A1