Protein formulations and methods of making same

Information

  • Patent Grant
  • 11191834
  • Patent Number
    11,191,834
  • Date Filed
    Tuesday, December 29, 2020
    3 years ago
  • Date Issued
    Tuesday, December 7, 2021
    2 years ago
Abstract
The invention provides an aqueous formulation comprising water and a protein, and methods of making the same. The aqueous formulation of the invention may be a high protein formulation and/or may have low levels of conductity resulting from the low levels of ionic excipients. Also included in the invention are formulations comprising water and proteins having low osmolality.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Dec. 28, 2020, is named 13371-218-999 SEQLIST.txt and 4638 bytes in size, and is identical to the paper copy to the Sequence Listing.


BACKGROUND OF THE INVENTION

A basic principle of pharmaceutical protein formulations is that certain instabilities must be overcome. Degradation pathways of proteins can be separated into two distinct classes, involving chemical instability and physical instability. Chemical instabilities lead to the modification of the protein through bond formation or cleavage. Examples of chemical instability problems include deamidation, racemization, hydrolysis, oxidation, beta elimination and disulfide exchange. Physical instabilities, on the other hand, do not lead to covalent changes in proteins. Rather, they involve changes in the higher order structure (secondary and above) of proteins. These include denaturation, adsorption to surfaces, aggregation and precipitation (Manning et al., Pharm. Res. 6, 903 (1989)).


It is generally accepted that these instabilities, which can have great effect on the commercial viability and efficacy of pharmaceutical protein formulations, can be overcome by including additional molecules in the formulation. Protein stability can be improved by including excipients that interact with the protein in solution to keep the protein stable, soluble and unaggregated. For example, salt compounds and other ionic species are very common additives to protein formulations. They assist in fighting denaturation of proteins by binding to proteins in a non-specific fashion and increasing thermal stability. Salt compounds (e.g., NaCl, KCl) have been used successfully in commercial insulin preparations to fight aggregation and precipitation (ibid. at 911). Amino acids (e.g., histidine, arginine) have been shown to reduce alterations in proteins' secondary structures when used as formulation additives (Tian et al., Int'l J. Pharm. 355, 20 (2007)). Other examples of commonly used additives include polyalcohol materials such as glycerol and sugars, and surfactants such as detergents, both nonionic (e.g., Tween, Pluronic) and anionic (sodium dodecyl sulfate). The near universal prevalence of additives in all liquid commercial protein formulations indicates that protein solutions without such compounds may encounter challenges with degradation due to instabilities.


The primary goal of protein formulation is to maintain the stability of a given protein in its native, pharmaceutically active form over prolonged periods of time to guarantee acceptable shelf-life of the pharmaceutical protein drug. Maintaining the stability and solubility of proteins in solution, however, is especially challenging in pharmaceutical formulations where the additives are included into therapeutics. To date, biologic formulations require additional excipients to maintain protein stability. Typically, liquid pharmaceutical formulations contain multiple additives for stability. For example, a liquid formulation for patient self-administration of human growth hormone, Norditropin SimpleXx®, contains the additives mannitol (a sugar alcohol), histidine and poloxamer 188 (a surfactant) to stabilize the hormone.


Pharmaceutical additives need to be soluble, non-toxic and used at particular concentrations that provide stabilizing effects on the specific therapeutic protein. Since the stabilizing effects of additives are protein- and concentration-dependent, each additive being considered for use in a pharmaceutical formulation must be carefully tested to ensure that it does not cause instability or have other negative effects on the chemical or physical make-up of the formulation. Ingredients used to stabilize the protein may cause problems with protein stability over time or with protein stability in changing environments during storage.


Typically, long shelf-life is achieved by storing the protein in frozen from (e.g., at −80° C.) or by subjecting the protein to a lyophilization process, i.e., by storing the protein in lyophilized form, necessitating a reconstitution step immediately before use and thus posing a significant disadvantage with regard to patient convenience. However, freezing a protein formulation for storage may lead to localized high concentrations of proteins and additives, which can create local extremes in pH, degradation and protein aggregation within the formulation. In addition, it is well known to those skilled in the art that freezing and thawing processes often impact protein stability, meaning that even storage of the pharmaceutical protein in frozen form can be associated with the loss of stability due to the freezing and thawing step. Also, the first process step of lyophilization involves freezing, which can negatively impact protein stability. In industry settings, a pharmaceutical protein may be subjected to repeated freeze-thaw processing during Drug Substance manufacturing (holding steps, storage, re-freeze and re-thaw to increase timing and batch size flexibility in Drug Product fill-finishing) and during subsequent Drug Product fill-finishing (lyophilization). Since it is well known that the risk of encountering protein instability phenomena increases with increasing the number of freeze-thaw cycles a pharmaceutical protein encounters, achieving formulation conditions that maintain protein stability over repeated freeze-thaw processes is a challenging task. There is a need in the biopharmaceutical industry for formulations that can be frozen and thawed without creating undesired properties in the formulations, especially gradients of pH, osmolarity, density or protein or excipient concentration.


Often protein-based pharmaceutical products need to be formulated at high concentrations for therapeutic efficacy. Highly concentrated protein formulations are desirable for therapeutic uses since they allow for dosages with smaller volumes, limiting patient discomfort, and are more economically packaged and stored. The development of high protein concentration formulations, however, presents many challenges, including manufacturing, stability, analytical, and, especially for therapeutic proteins, delivery challenges. For example, difficulties with the aggregation, insolubility and degradation of proteins generally increase as protein concentrations in formulations are raised (for review, see Shire, S. J. et al. J. Pharm. Sci., 93, 1390 (2004)). Previously unseen negative effects may be caused by additives that, at lower concentrations of the additives or the protein, provided beneficial effects. The production of high concentration protein formulations may lead to significant problems with opalescence, aggregation and precipitation. In addition to the potential for nonnative protein aggregation and particulate formation, reversible self-association may occur, which may result in increased viscosity or other properties that complicate delivery by injection. High viscosity also may complicate manufacturing of high protein concentrations by filtration approaches.


Thus, pharmaceutical protein formulations typically carefully balance ingredients and concentrations to enhance protein stability and therapeutic requirements while limiting any negative side-effects. Biologic formulations should include stable protein, even at high concentrations, with specific amounts of excipients reducing potential therapeutic complications, storage issues and overall cost.


As proteins and other biomacromolecules gain increased interest as drug molecules, formulations for delivering such molecules are becoming an important issue. Despite the revolutionary progress in the large-scale manufacturing of proteins for therapeutic use, effective and convenient delivery of these agents in the body remains a major challenge due to their intrinsic physicochemical and biological properties, including poor permeation through biological membranes, large molecular size, short plasma half life, self association, physical and chemical instability, aggregation, adsorption, and immunogenicity.


SUMMARY OF THE INVENTION

The invention is directed towards the surprising findings that proteins formulated in water maintain solubility, as well as stability, even at high concentrations, during long-term liquid storage or other processing steps, such as freeze/thawing and lyophilization.


The present invention relates to methods and compositions for aqueous protein formulations which comprise water and a protein, where the protein is stable without the need for additional agents. Specifically, the methods and compositions of the invention are based on a diafiltration process wherein a first solution containing the protein of interest is diafiltered using water as a diafiltration medium. The process is performed such that there is at least a determined volume exchange, e.g., a five fold volume exchange, with the water. By performing the methods of the invention, the resulting aqueous formulation has a significant decrease in the overall percentage of excipients in comparison to the initial protein solution. For example, 95-99% less excipients are found in the aqueous formulation in comparison to the initial protein solution. Despite the decrease in excipients, the protein remains soluble and retains its biological activity, even at high concentrations. In one aspect, the methods of the invention result in compositions comprising an increase in concentration of the protein while decreasing additional components, such as ionic excipients. As such, the hydrodynamic diameter of the protein in the aqueous formulation is smaller relative to the same protein in a standard buffering solution, such as phosphate buffered saline (PBS).


The formulation of the invention has many advantages over standard buffered formulations. In one aspect, the aqueous formulation comprises high protein concentrations, e.g., 50 to 200 mg/mL or more. Proteins of all sizes may be included in the formulations of the invention, even at increased concentrations. Despite the high concentration of protein, the formulation has minimal aggregation and can be stored using various methods and forms, e.g., freezing, without deleterious effects that might be expected with high protein formulations. Formulations of the invention do not require excipients, such as, for example, surfactants and buffering systems, which are used in traditional formulations to stabilize proteins in solution. As a result of the low level of ionic excipients, the aqueous formulation of the invention has low conductivity, e.g., less than 2 mS/cm. The methods and compositions of the invention also provide aqueous protein formulations having low osmolality, e.g., no greater than 30 mOsmol/kg. In addition, the formulations described herein are preferred over standard formulations because they have decreased immunogenicity due to the lack of additional agents needed for protein stabilization.


The methods and compositions of the invention may be used to provide an aqueous formulation comprising water and any type of protein of interest. In one aspect, the methods and compositions of the invention are used for large proteins, including proteins which are larger than 47 kDa. Antibodies, and fragments thereof, including those used for in vivo and in vitro purposes, are another example of proteins which may be used in the methods and compositions of the invention.


Furthermore, the multiple step purification and concentration processes that are necessary to prepare proteins and peptide formulations often introduce variability in compositions, such that the precise composition of a formulation may vary from lot to lot. Federal regulations require that drug compositions be highly consistent in their formulations regardless of the location of manufacture or lot number. Methods of the invention can be used to create solutions of proteins formulated in water to which buffers and excipients are added back in precise amounts, allowing for the creation of protein formulations with precise concentrations of buffers and/or excipients.


In one embodiment, the invention provides an aqueous formulation comprising a protein and water, wherein the formulation has certain characteristics, such as, but not limited to, low conductivity, e.g., a conductivity of less than about 2.5 mS/cm, a protein concentration of at least about 10 μg/mL, an osmolality of no more than about 30 mOsmol/kg, and/or the protein has a molecular weight (Mw) greater than about 47 kDa. In one embodiment, the formulation of the invention has improved stability, such as, but not limited to, stability in a liquid form for an extended time (e.g., at least about 3 months or at least about 12 months) or stability through at least one freeze/thaw cycle (if not more freeze/thaw cycles). In one embodiment, the formulation is stable for at least about 3 months in a form selected from the group consisting of frozen, lyophilized, or spray-dried.


In one embodiment, proteins included in the formulation of the invention may have a minimal size, including, for example, a Mw greater than about 47 kDa, a Mw greater than about 57 kDa, a Mw greater than about 100 kDa, a Mw greater than about 150 kDa, a Mw greater than about 200 kDa, or a Mw greater than about 250 kDa.


In one embodiment, the formulation of the invention has a low conductivity, including, for example, a conductivity of less than about 2.5 mS/cm, a conductivity of less than about 2 mS/cm, a conductivity of less than about 1.5 mS/cm, a conductivity of less than about 1 mS/cm, or a conductivity of less than about 0.5 mS/cm.


In one embodiment, proteins included in the formulation of the invention have a given concentration, including, for example, a concentration of at least about 1 mg/mL, at least about 10 mg/mL, at least about 50 mg/mL, at least about 100 mg/mL, at least about 150 mg/mL, at least about 200 mg/mL, or greater than about 200 mg/mL.


In one embodiment, the formulation of the invention has an osmolality of no more than about 15 mOsmol/kg.


In one embodiment, the invention provides an aqueous formulation comprising water and a given concentration of a protein, wherein the protein has a hydrodynamic diameter (Dh) which is at least about 50% less than the Dh of the protein in a buffered solution at the given concentration. In one embodiment, the Dh of the protein is at least about 50% less than the IA of the protein in phosphate buffered saline (PBS) at the given concentration; the Dh of the protein is at least about 60% less than the Dh of the protein in PBS at the given concentration; the Dh of the protein is at least about 70% less than the Dh of the protein in PBS at the given concentration.


In one embodiment, the invention provides an aqueous formulation comprising a protein, such as, but not limited to, an antibody, or an antigen-binding fragment, wherein the protein has a hydrodynamic diameter (Dh) of less than about 5 μm. In one embodiment, the protein has a Dh of less than about 3 μm.


Any protein may be used in the methods and compositions of the invention. In one embodiment, the formulation comprises a therapeutic protein. In one embodiment, the formulation comprises an antibody, or an antigen-binding fragment thereof. Types of antibodies, or antigen binding fragments, that may be included in the methods and compositions of the invention include, but are not limited to, a chimeric antibody, a human antibody, a humanized antibody, and a domain antibody (dAb). In one embodiment, the antibody, or antigen-binding fragment thereof, is an anti-TNFα, such as but not limited to adalimumab or golimumab, or an anti-IL-12 antibody, such as but not limited to J695. In addition, the formulation of the invention may also include at least two distinct types of proteins, e.g., adalimumab and J695.


In yet another embodiment of the invention, the formulation may further comprise a non-ionizable excipient. Examples of non-ionizable excipients include, but are not limited to, a sugar alcohol or polyol (e.g., mannitol or sorbitol), a non-ionic surfactant (e.g., polysorbate 80, polysorbate 20, polysorbate 40, polysorbate 60), and/or a sugar (e.g., sucrose). Other non-limiting examples of non-ionizable excipients that may be further included in the formulation of the invention include, but are not limited to, non-trehalose, raffinose, and maltose.


In one embodiment, the formulation does not comprise an agent selected from the group consisting of a tonicity modifier, a stabilizing agent, a surfactant, an anti-oxidant, a cryoprotectant, a bulking agent, a lyroprotectant, a basic component, and an acidic component.


The formulation of the invention may be suitable for any use, including both in vitro and in vivo uses. In one embodiment, the formulation of the invention is suitable for administration to a subject via a mode of administration, including, but not limited to, subcutaneous, intravenous, inhalation, intradermal, transdermal, intraperitoneal, and intramuscular administration. The formulation of the invention may be used in the treatment of a disorder in a subject.


Also included in the invention are devices that may be used to deliver the formulation of the invention. Examples of such devices include, but are not limited to, a syringe, a pen, an implant, a needle-free injection device, an inhalation device, and a patch.


In one embodiment, the formulation of the invention is a pharmaceutical formulation.


The invention also provides a method of preparing an aqueous formulation comprising a protein and water, the method comprising providing the protein in a first solution, and subjecting the first solution to diafiltration using water as a diafiltration medium until at least a five fold volume exchange with the water has been achieved to thereby prepare the aqueous formulation. In one embodiment, the protein in the resulting formulation retains its biological activity.


The invention further provides a method of preparing an aqueous formulation of a protein, the method comprising providing the protein in a first solution; subjecting the first solution to diafiltration using water as a diafiltration medium until at least a five-fold volume exchange with the water has been achieved to thereby prepare a diafiltered protein solution; and concentrating the diafiltered protein solution to thereby prepare the aqueous formulation of the protein. In one embodiment, the protein in the resulting formulation retains its biological activity.


In one embodiment, the concentration of the diafiltered protein solution is achieved via centrifugation.


In one embodiment, the diafiltration medium consists of water.


In one embodiment, the first solution is subjected to diafiltration with water until a volume exchange greater than a five-fold volume exchange is achieved. In one embodiment, the first solution is subjected to diafiltration with water until at least about a six-fold volume exchange is achieved. In one embodiment, the first solution is subjected to diafiltration with water until at least about a seven-fold volume exchange is achieved.


In one embodiment, the aqueous formulation has a final concentration of excipients which is at least about 95% less than the first solution.


In one embodiment, the aqueous formulation has a final concentration of excipients which is at least about 99% less than the first solution.


In one embodiment, the first protein solution is obtained from a mammalian cell expression system and has been purified to remove host cell proteins (HCPs).


In one embodiment, the method of the invention further comprises adding an excipient to the aqueous formulation.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows the SEC chromatogram of Adalimumab reference standard AFP04C (bottom line), Adalimumab DS (drug substance before (middle line) and after DF/UF processing (top line).



FIG. 2 shows the impact of sorbitol (a non-ionizable excipient) and NaCl (ionizable excipient) concentrations on the hydrodynamic diameter (Dh) of Adalimumab monomer upon addition of the excipient compound to DF/UF-processed Adalimumab monomer.



FIG. 3 shows the IEC profile of J695 reference standard (bottom graph) and J695 DS, pH adjusted to pH 4.4 (top graph).



FIG. 4 shows the IEC profile of J695 after DF/UF with Milli-Q water, pH 4.7 (top graph), and J695 DS before DF/UF, pH adjusted to pH 4.4 (bottom curve).



FIG. 5 graphically depicts the correlation of hydrodynamic diameter (z-average) and concentration of Adalimumab (dissolved in WFI). X: determined with an SOP using 1.1 mPas as assumed sample viscosity. y: determined with an SOP using 1.9 mPas as assumed sample viscosity.



FIG. 6 graphically depicts the correlation of hydrodynamic diameter (peak monomer) and concentration of Adalimumab (dissolved in WFI). X: determined with an SOP using 1.1 mPas as assumed sample viscosity. y: determined with an SOP using 1.9 mPas as assumed sample viscosity



FIG. 7 graphically depicts the correlation of hydrodynamic diameter (z-average) and concentration of J695 (dissolved in WFI). X: determined with an SOP using 1.1 mPas as assumed sample viscosity: determined with an SOP using 1.9 mPas as assumed sample viscosity



FIG. 8 graphically depicts the correlation of hydrodynamic diameter (peak monomer) and concentration of J695 (dissolved in WFI). X: determined with an SOP using 1.1 mPas as assumed sample viscosity. y: determined with an SOP using 1.9 mPas as assumed sample viscosity.



FIG. 9 shows the sum of lysine 0, 1 and 2 of Adalimumab [%] in dependence on Adalimumab concentration in water for injection.



FIG. 10 shows the sum of peak 1 to 7 of J695 [%] in dependence on J695 concentration in water for injection.



FIG. 11 shows the sum of acidic peaks of J695 [%] in dependence on J695 concentration in water for injection.



FIG. 12 shows the sum of basic peaks of J695 [%] in dependence on J695 concentration in water for injection (WFI).



FIG. 13 shows the efficiency of the dialysis performed in Example 12, in terms of the reduction of components responsible for osmolality and conductivity of the formulation (BDS, 74 mg/ml, 10 ml sample volume, SpectraPor7 MWCO10k).



FIG. 14 shows the stability of pH levels in dialyzed Adalimumab Bulk Solutions. pH levels before and after dialysis against deionized water (1:1,000,000) are shown. (BDS, 74 mg/ml, 10 ml sample volume, SpectraPor7 MWCO10k)



FIG. 15 shows bottle mapping density data for 250 mg/ml and 200 mg/ml low-ionic Adalimumab solutions after freeze thaw.



FIG. 16 shows bottle mapping pH data for 250 mg/ml and 200 mg/ml low-ionic Adalimumab solutions after freeze thaw.



FIG. 17 shows bottle mapping concentration data for 250 mg/ml and 200 mg/ml low-ionic Adalimumab solutions after freeze thaw.



FIG. 18 shows bottle mapping osmolality data for 250 mg/ml and 200 mg/ml low-ionic Adalimumab solutions after freeze thaw.



FIG. 19 shows bottle mapping conductivity data for 250 mg/ml and 200 mg/ml low-ionic Adalimumab solutions after freeze thaw.



FIG. 20 shows SEC analysis of low-ionic Adalimumab (referred to as D2E7 in FIG. 20) solutions that were either stored at 2-8° C. for 8.5 months after DF/UF (bottom curve) or stored at −80° C. for 4.5 months after DF/UF (top curve).



FIG. 21 shows the stability of the monoclonal antibody 1D4.7 formulated in various solutions and in water before freeze-thaw procedures (T0) and after each of four freeze-thaws (T1, T2, T3 and T4).



FIG. 22 shows the stability of the monoclonal antibody 13C5.5 formulated in water and with various buffers before freeze-thaw procedures (T0) and after each of four freeze-thaws (T1, T2, T3 and T4). Blank=WFI control sample.



FIG. 23 shows the stability of the monoclonal antibody 13C5.5 formulated in water and with various excipients added, before freeze-thaw procedures (T0) and after each of four freeze-thaws (T1, T2, T3 and T4). Blank=WFI control sample.



FIG. 24 shows the impact of the concentration of Adalimumab (WFI formulation) and solution pH on solution viscosity.



FIG. 25 shows turbidity data for Adalimumab solutions (WFI formulations) of various concentrations and pH values.



FIG. 26 shows hydrodynamic diameter (Dh) data for Adalimumab solutions (WFI formulations) at various pH values and concentrations.



FIG. 27 shows a size distribution by intensity graph (Dh measurements) for Adalimumab in water solutions, pH 5, at various concentrations.



FIG. 28 shows size distribution by intensity for 100 mg/mL Adalimumab in water at various pH levels.



FIG. 29 also shows size distribution by intensity for 100 mg/mL Adalimumab in water at various pH levels.



FIG. 30 shows monomer content (SEC) for Adalimumab in water.



FIG. 31 shows aggregate content (SEC) for Adalimumab in water.



FIG. 32 shows the viscosity of two J695 solutions (WFI formulations) as a function of solution temperature.



FIG. 33 graphically depicts 1D4.7 antibody stability as measured by subvisible particle (>1 μm) during repeated freeze/thaw (f/t) cycles for a number of different formulations.



FIG. 34 graphically depicts 13C5.5 antibody stability as measured by subvisible particle (>10 μm) during repeated freeze/thaw (f/t) cycles for a number of different formulations.



FIG. 35 graphically depicts 13C5.5 antibody stability as measured by subvisible particle (>1 μm) during repeated freeze/thaw (f/t) cycles for a number of different formulations.



FIG. 36 graphically depicts 7C6 antibody stability as measured by subvisible particle (>1 μm) during repeated freeze/thaw (f/t) cycles for a number of different formulations.





DETAILED DESCRIPTION OF THE INVENTION
I. Definitions

In order that the present invention may be more readily understood, certain terms are first defined.


As used herein, the term “acidic component” refers to an agent, including a solution, having an acidic pH, i.e., less than 7.0. Examples of acidic components include phosphoric acid, hydrochloric acid, acetic acid, citric acid, oxalic acid, succinic acid, tartaric acid, lactic acid, malic acid, glycolic acid and fumaric acid. In one embodiment, the aqueous formulation of the invention does not include an acidic component.


As used herein, the term “antioxidant” is intended to mean an agent which inhibits oxidation and thus is used to prevent the deterioration of preparations by the oxidative process. Such compounds include by way of example and without limitation, acetone, sodium bisulfate, ascorbic acid, ascorbyl palmitate, citric acid, butylated hydroxyanisole, butylated hydroxytoluene, hydrophosphorous acid, monothioglycerol, propyl gallate, methionine, sodium ascorbate, sodium citrate, sodium sulfide, sodium sulfite, sodium bisulfate, sodium formaldehyde sulfoxylate, thioglycolic acid, sodium metabisulfite, EDTA (edetate), pentetate and others known to those of ordinary skill in the art.


The term “aqueous formulation” refers to a solution in which the solvent is water.


As used herein, the term “basic component” refers to an agent which is alkaline, i.e., pH greater than 7.0. Examples of basic components include potassium hydroxide (KOH) and sodium hydroxide (NaOH)


As used herein, the term “bulking agent” is intended to mean a compound used to add bulk to the reconstitutable solid and/or assist in the control of the properties of the formulation during preparation. Such compounds include, by way of example and without limitation, dextran, trehalose, sucrose, polyvinylpyrrolidone, lactose, inositol, sorbitol, dimethylsulfoxide, glycerol, albumin, calcium lactobionate, and others known to those of ordinary skill in the art.


The term “conductivity,” as used herein, refers to the ability of an aqueous solution to conduct an electric current between two electrodes. Generally, electrical conductivity or specific conductivity is a measure of a material's ability to conduct an electric current. In solution, the current flows by ion transport. Therefore, with an increasing amount of ions present in the aqueous solution, the solution will have a higher conductivity. The unit of measurement for conductivity is mmhos (mS/cm), and can be measured using a conductivity meter sold, e.g., by Orion Research, Inc. (Beverly, Mass.). The conductivity of a solution may be altered by changing the concentration of ions therein. For example, the concentration of ionic excipients in the solution may be altered in order to achieve the desired conductivity.


The term “cryoprotectants” as used herein generally includes agents, which provide stability to the protein from freezing-induced stresses. Examples of cryoprotectants include polyols such as, for example, mannitol, and include saccharides such as, for example, sucrose, as well as including surfactants such as, for example, polysorbate, poloxamer or polyethylene glycol, and the like. Cryoprotectants also contribute to the tonicity of the formulations.


As used herein, the terms “ultrafiltration” or “UF” refers to any technique in which a solution or a suspension is subjected to a semi-permeable membrane that retains macromolecules while allowing solvent and small solute molecules to pass through. Ultrafiltration may be used to increase the concentration of macromolecules in a solution or suspension. In a preferred embodiment, ultrafiltration is used to increase the concentration of a protein in water.


As used herein, the term “diafiltration” or “DF” is used to mean a specialized class of filtration in which the retentate is diluted with solvent and re-filtered, to reduce the concentration of soluble permeate components. Diafiltration may or may not lead to an increase in the concentration of retained components, including, for example, proteins. For example, in continuous diafiltration, a solvent is continuously added to the retentate at the same rate as the filtrate is generated. In this case, the retentate volume and the concentration of retained components does not change during the process. On the other hand, in discontinuous or sequential dilution diafiltration, an ultrafiltration step is followed by the addition of solvent to the retentate side; if the volume of solvent added to the retentate side is not equal or greater to the volume of filtrate generated, then the retained components will have a high concentration. Diafiltration may be used to alter the pH, ionic strength, salt composition, buffer composition, or other properties of a solution or suspension of macromolecules.


As used herein, the terms “diafiltration/ultrafiltration” or “DF/UF” refer to any process, technique or combination of techniques that accomplishes ultrafiltration and/or diafiltration, either sequentially or simultaneously.


As used herein, the term “diafiltration step” refers to a total volume exchange during the process of diafiltration.


The term “excipient” refers to an agent that may be added to a formulation to provide a desired consistency, (e.g., altering the bulk properties), to improve stability, and/or to adjust osmolality. Examples of commonly used excipients include, but are not limited to, sugars, polyols, amino acids, surfactants, and polymers. The term “ionic excipient” or “ionizable excipient,” as used interchangeably herein, refers to an agent that has a net charge. In one embodiment, the ionic excipient has a net charge under certain formulation conditions, such as pH. Examples of an ionic excipient include, but are not limited to, histidine, arginine, and sodium chloride. The term “non-ionic excipient” or “non-ionizable excipient,” as used interchangeably herein, refers to an agent having no net charge. In one embodiment, the non-ionic excipient has no net charge under certain formulation conditions, such as pH. Examples of non-ionic excipients include, but are not limited to, sugars (e.g., sucrose), sugar alcohols (e.g., mannitol), and non-ionic surfactants (e.g., polysorbate 80).


The term “first protein solution” or “first solution” as used herein, refers to the initial protein solution or starting material used in the methods of the invention, i.e., the initial protein solution which is diafiltered into water. In one embodiment, the first protein solution comprises ionic excipients, non-ionic excipients, and/or a buffering system.


The term “hydrodynamic diameter” or “Dh” of a particle refers to the diameter of a sphere that has the density of water and the same velocity as the particle. Thus the term “hydrodynamic diameter of a protein” as used herein refers to a size determination for proteins in solution using dynamic light scattering (DLS). A DLS-measuring instrument measures the time-dependent fluctuation in the intensity of light scattered from the proteins in solution at a fixed scattering angle. Protein Dh is determined from the intensity autocorrelation function of the time-dependent fluctuation in intensity. Scattering intensity data are processed using DLS instrument software to determine the value for the hydrodynamic diameter and the size distribution of the scattering molecules, i.e. the protein specimen.


The term “lyoprotectant” as used herein includes agents that provide stability to a protein during water removal during the drying or lyophilisation process, for example, by maintaining the proper conformation of the protein. Examples of lyoprotectants include saccharides, in particular di- or trisaccharides. Cryoprotectants may also provide lyoprotectant effects.


The term “pharmaceutical” as used herein with reference to a composition, e.g., an aqueous formulation, that it is useful for treating a disease or disorder.


The term “protein” is meant to include a sequence of amino acids for which the chain length is sufficient to produce the higher levels of secondary and/or tertiary and/or quaternary structure. This is to distinguish from “peptides” or other small molecular weight drugs that do not have such structure. In one embodiment, the proteins used herein have a molecular weight of at least about 47 kD. Examples of proteins encompassed within the definition used herein include therapeutic proteins. A “therapeutically active protein” or “therapeutic protein” refers to a protein which may be used for therapeutic purposes, i.e., for the treatment of a disorder in a subject. It should be noted that while therapeutic proteins may be used for treatment purposes, the invention is not limited to such use, as said proteins may also be used for in vitro studies. In a preferred embodiment, the therapeutic protein is a fusion protein or an antibody, or antigen-binding portion thereof. In one embodiment, the methods and compositions of the invention comprise at least two distinct proteins, which are defined as two proteins having distinct amino acid sequences. Additional distinct proteins do not include degradation products of a protein.


The phrase “protein is dissolved in water” as used herein refers to a formulation of a protein wherein the protein is dissolved in an aqueous solution in which the amount of small molecules (e.g., buffers, excipients, salts, surfactants) has been reduced by DF/UF processing. Even though the total elimination of small molecules cannot be achieved in an absolute sense by DF/UF processing, the theoretical reduction of excipients achievable by applying DF/UF is sufficiently large to create a formulation of the protein essentially in water exclusively. For example, with 6 volume exchanges in a continuous mode DF/UF protocol, the theoretical reduction of excipients is ˜99.8% (ci=e−x, with ci being the initial excipient concentration, and x being the number of volume exchanges).


The term “pharmaceutical formulation” refers to preparations which are in such a form as to permit the biological activity of the active ingredients to be effective, and, therefore may be administered to a subject for therapeutic use.


A “stable” formulation is one in which the protein therein essentially retains its physical stability and/or chemical stability and/or biological activity upon storage. Various analytical techniques for measuring protein stability are available in the art and are reviewed in Peptide and Protein Drug Delivery, 247-301, Vincent Lee Ed., Marcel Dekker, Inc., New York, N.Y., Pubs. (1991) and Jones, A. Adv. Drug Delivery Rev. 10: 29-90 (1993), for example. In one embodiment, the stability of the protein is determined according to the percentage of monomer protein in the solution, with a low percentage of degraded (e.g., fragmented) and/or aggregated protein. For example, an aqueous formulation comprising a stable protein may include at least 95% monomer protein. Alternatively, an aqueous formulation of the invention may include no more than 5% aggregate and/or degraded protein.


The term “stabilizing agent” refers to an excipient that improves or otherwise enhances stability. Stabilizing agents include, but are not limited to, α-lipoic acid, α-tocopherol, ascorbyl palmitate, benzyl alcohol, biotin, bisulfites, boron, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), ascorbic acid and its esters, carotenoids, calcium citrate, acetyl-L-camitine, chelating agents, chondroitin, chromium, citric acid, coenzyme Q-10, cysteine, cysteine hydrochloride, 3-dehydroshikimic acid (DHS), EDTA (ethylenediaminetetraacetic acid; edetate disodium), ferrous sulfate, folic acid, fumaric acid, alkyl gallates, garlic, glucosamine, grape seed extract, gugul, magnesium, malic acid, metabisulfite, N-acetyl cysteine, niacin, nicotinomide, nettle root, ornithine, propyl gallate, pycnogenol, saw palmetto, selenium, sodium bisulfite, sodium metabisulfite, sodium sulfite, potassium sulfite, tartaric acid, thiosulfates, thioglycerol, thiosorbitol, tocopherol and their esters, e.g., tocopheral acetate, tocopherol succinate, tocotrienal, d-α-tocopherol acetate, vitamin A and its esters, vitamin B and its esters, vitamin C and its esters, vitamin D and its esters, vitamin E and its esters, e.g., vitamin E acetate, zinc, and combinations thereof.


The term “surfactants” generally includes those agents that protect the protein from air/solution interface-induced stresses and solution/surface induced-stresses. For example surfactants may protect the protein from aggregation. Suitable surfactants may include, e.g., polysorbates, polyoxyethylene alkyl ethers such as Brij 35®, or poloxamer such as Tween 20, Tween 80, or poloxamer 188. Preferred detergents are poloxamers, e.g., Poloxamer 188, Poloxamer 407; polyoxyethylene alkyl ethers, e.g., Brij 35®, Cremophor A25, Sympatens ALM/230; and polysorbates/Tweens, e.g., Polysorbate 20, Polysorbate 80, and Poloxamers, e.g., Poloxamer 188, and Tweens, e.g., Tween 20 and Tween 80.


As used herein, the term “tonicity modifier” is intended to mean a compound or compounds that can be used to adjust the tonicity of a liquid formulation. Suitable tonicity modifiers include glycerin, lactose, mannitol, dextrose, sodium chloride, magnesium sulfate, magnesium chloride, sodium sulfate, sorbitol, trehalose, sucrose, raffinose, maltose and others known to those or ordinary skill in the art. In one embodiment, the tonicity of the liquid formulation approximates that of the tonicity of blood or plasma.


The term “water” is intended to mean water that has been purified to remove contaminants, usually by distillation or reverse osmosis, also referred to herein as “pure water”. In a preferred embodiment, water used in the methods and compositions of the invention is excipient-free. In one embodiment, water includes sterile water suitable for administration to a subject. In another embodiment, water is meant to include water for injection (WFI). In one embodiment, water refers to distilled water or water which is appropriate for use in in vitro assays. In a preferred embodiment, diafiltration is performed in accordance with the methods of the invention using water alone as the diafiltration medium.


The term “antibody” as referred to herein includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”) or single chains thereof. An “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.


The term “antigen-binding portion” of an antibody (or simply “antibody portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., TNFα, IL-12). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH or VL domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. In one embodiment of the invention, the antibody fragment is selected from the group consisting of a Fab, an Fd, an Fd′, a single chain Fv (scFv), an scFva, and a domain antibody (dAb).


Still further, an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. These other proteins or peptides can have functionalities that allow for the purification of antibodies or antigen-binding portions thereof or allow for their association with each other or other molecules. Thus examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric single chain variable fragment (scFv) molecules (Kipriyanov et al. (1995) Human Antibodies and Hybridomas 6:93-101) and the use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov et al. (1994) Mol. Immunol. 31:1047-1058). Antibody portions, such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques.


Two antibody domains are “complementary” where they belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature. For example, a VH domain and a VL domain of an antibody are complementary; two VH domains are not complementary, and two VL domains are not complementary. Complementary domains may be found in other members of the immunoglobulin superfamily, such as the Vα and Vβ (or gamma and delta) domains of the T-cell receptor.


The term “domain” refers to a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases may be added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain. By single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains. It therefore includes complete antibody variable domains and modified variable domains, for example in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain at least in part the binding activity and specificity of the full-length domain.


Variable domains of the invention may be combined to form a group of domains; for example, complementary domains may be combined, such as VL domains being combined with VH domains. Non-complementary domains may also be combined. Domains may be combined in a number of ways, involving linkage of the domains by covalent or non-covalent means.


A “dAb” or “domain antibody” refers to a single antibody variable domain (VH or VL) polypeptide that specifically binds antigen.


As used herein, the term “antigen binding region” or “antigen binding site” refers to the portion(s) of an antibody molecule, or antigen binding portion thereof, which contains the amino acid residues that interact with an antigen and confers on the antibody its specificity and affinity for the antigen.


The term “epitope” is meant to refer to that portion of any molecule capable of being recognized by and bound by an antibody at one or more of the antibody's antigen binding regions. In the context of the present invention, first and second “epitopes” are understood to be epitopes which are not the same and are not bound by a single monospecific antibody, or antigen-binding portion thereof.


The phrase “recombinant antibody” refers to antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor et al. (1992) Nucl. Acids Res. 20:6287-6295) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of particular immunoglobulin gene sequences (such as human immunoglobulin gene sequences) to other DNA sequences. Examples of recombinant antibodies include chimeric, CDR-grafted and humanized antibodies.


The term “human antibody” refers to antibodies having variable and constant regions corresponding to, or derived from, human germline immunoglobulin sequences as described by, for example, Kabat et al. (See Kabat, et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). The human antibodies of the invention, however, may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.


Recombinant human antibodies of the invention have variable regions, and may also include constant regions, derived from human germline immunoglobulin sequences (See Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. In certain embodiments, however, such recombinant antibodies are the result of selective mutagenesis or backmutation or both.


The term “backmutation” refers to a process in which some or all of the somatically mutated amino acids of a human antibody are replaced with the corresponding germline residues from a homologous germline antibody sequence. The heavy and light chain sequences of a human antibody of the invention are aligned separately with the germline sequences in the VBASE database to identify the sequences with the highest homology. Differences in the human antibody of the invention are returned to the germline sequence by mutating defined nucleotide positions encoding such different amino acid. The role of each amino acid thus identified as candidate for backmutation should be investigated for a direct or indirect role in antigen binding and any amino acid found after mutation to affect any desirable characteristic of the human antibody should not be included in the final human antibody. To minimize the number of amino acids subject to backmutation those amino acid positions found to be different from the closest germline sequence but identical to the corresponding amino acid in a second germline sequence can remain, provided that the second germline sequence is identical and colinear to the sequence of the human antibody of the invention for at least 10, preferably 12 amino acids, on both sides of the amino acid in question. Backmuation may occur at any stage of antibody optimization.


The term “chimeric antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species, such as antibodies having murine heavy and light chain variable regions linked to human constant regions.


The term “CDR-grafted antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.


The term “humanized antibody” refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more “human-like”, i.e., more similar to human germline variable sequences. One type of humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences.


Various aspects of the invention are described in further detail in the following subsections.


II. Methods of Invention

Generally, diafiltration is a technique that uses membranes to remove, replace, or lower the concentration of salts or solvents from solutions containing proteins, peptides, nucleic acids, and other biomolecules. Protein production operations often involve final diafiltration of a protein solution into a formulation buffer once the protein has been purified from impurities resulting from its expression, e.g., host cell proteins. The invention described herein provides a means for obtaining an aqueous formulation by subjecting a protein solution to diafiltration using water alone as a diafiltration solution. Thus, the formulation of the invention is based on using water as a formulation medium during the diafiltration process and does not rely on traditional formulation mediums which include excipients, such as surfactants, used to solubilize and/or stabilize the protein in the final formulation. The invention provides a method for transferring a protein into pure water for use in a stable formulation, wherein the protein remains in solution and is able to be concentrated at high levels without the use of other agents to maintain its stability.


Prior to diafiltration or DF/UF in accordance with the teachings herein, the method includes first providing a protein in a first solution. The protein may be formulated in any first solution, including formulations using techniques that are well established in the art, such as synthetic techniques (e.g., recombinant techniques, peptide synthesis, or a combination thereof). Alternatively, the protein used in the methods and compositions of the invention is isolated from an endogenous source of the protein. The initial protein solution may be obtained using a purification process whereby the protein is purified from a heterogeneous mix of proteins. In one embodiment, the initial protein solution used in the invention is obtained from a purification method whereby proteins, including antibodies, expressed in a mammalian expression system are subjected to numerous chromatography steps which remove host cell proteins (HCPs) from the protein solution. In one embodiment, the first protein solution is obtained from a mammalian cell expression system and has been purified to remove host cell proteins (HCPs). Examples of methods of purification are described in U.S. application Ser. No. 11/732,918 (US 20070292442), incorporated by reference herein. It should be noted that there is no special preparation of the first protein solution required in accordance with the methods of the invention.


Proteins which may be used in the compositions and methods of the invention may be any size, i.e., molecular weight (Mw). For example, the protein may have a Mw equal to or greater than about 1 kDa, a Mw equal to or greater than about 10 kDa, a Mw equal to or greater than about 47 kDa, a Mw equal to or greater than about 57 kDa, a Mw equal to or greater than about 100 kDa, a Mw equal to or greater than about 150 kDa, a Mw equal to or greater than about 200 kDa, or a Mw equal to or greater than about 250 kDa. Numbers intermediate to the above recited Mw, e.g., 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 153, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, and so forth, as well as all other numbers recited herein, are also intended to be part of this invention. Ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included in the scope of the invention. For example, proteins used in the invention may range in size from 57 kDa to 250 kDa, from 56 kDa to 242 kDa, from 60 kDa to 270 kDa, and so forth.


The methods of the invention also include diafiltration of a first protein solution that comprises at least two distinct proteins. For example, the protein solution may contain two or more types of antibodies directed to different molecules or different epitopes of the same molecule.


In one embodiment, the protein that is in solution is a therapeutic protein, including, but not limited to, fusion proteins and enzymes. Examples of therapeutic proteins include, but are not limited to, Pulmozyme (Dornase alfa), Regranex (Becaplermin), Activase (Alteplase), Aldurazyme (Laronidase), Amevive (Alefacept), Aranesp (Darbepoetin alfa), Becaplermin Concentrate, Betaseron (Interferon beta-1b), BOTOX (Botulinum Toxin Type A), Elitek (Rasburicase), Elspar (Asparaginase), Epogen (Epoetin alfa), Enbrel (Etanercept), Fabrazyme (Agalsidase beta), Infergen (Interferon alfacon-1), Intron A (Interferon alfa-2a), Kineret (Anakinra), MYOBLOC (Botulinum Toxin Type B), Neulasta (Pegfilgrastim), Neumega (Oprelvekin), Neupogen (Filgrastim), Ontak (Denileukin diftitox), PEGASYS (Peginterferon alfa-2a), Proleukin (Aldesleukin), Pulmozyme (Dornase alfa), Rebif (Interferon beta-1a), Regranex (Becaplermin), Retavase (Reteplase), Roferon-A (Interferon alfa-2), TNKase (Tenecteplase), and Xigris (Drotrecogin alfa), Arcalyst (Rilonacept), NPlate (Romiplostim), Mircera (methoxypolyethylene glycol-epoetin beta), Cinryze (C1 esterase inhibitor), Elaprase (idursulfase), Myozyme (alglucosidase alfa), Orencia (abatacept), Naglazyme (galsulfase), Kepivance (palifermin) and Actimmune (interferon gamma-1b).


The protein used in the invention may also be an antibody, or antigen-binding fragment thereof. Examples of antibodies that may be used in the invention include chimeric antibodies, non-human antibodies, human antibodies, humanized antibodies, and domain antibodies (dAbs). In one embodiment, the antibody, or antigen-binding fragment thereof, is an anti-TNFα and/or an anti-IL-12 antibody (e.g., it may be a dual variable domain (DVD) antibody). Other examples of antibodies, or antigen-binding fragments thereof, which may be used in the methods and compositions of the invention include, but are not limited to, 1D4.7 (anti-IL-12/IL-23 antibody; Abbott Laboratories), 2.5(E)mg1 (anti-IL-18; Abbott Laboratories), 13C5.5 (anti-IL-13 antibody; Abbott Laboratories), J695 (anti-IL-12; Abbott Laboratories), Afelimomab (Fab 2 anti-TNF; Abbott Laboratories), Humira (adalimumab) Abbott Laboratories), Campath (Alemtuzumab), CEA-Scan Arcitumomab (fab fragment), Erbitux (Cetuximab), Herceptin (Trastuzumab), Myoscint (Imciromab Pentetate), ProstaScint (Capromab Pendetide), Remicade (Infliximab), ReoPro (Abciximab), Rituxan (Rituximab), Simulect (Basiliximab), Synagis (Palivizumab), Verluma (Nofetumomab), Xolair (Omalizumab), Zenapax (Daclizumab), Zevalin (Ibritumomab Tiuxetan), Orthoclone OKT3 (Muromonab-CD3), Panorex (Edrecolomab), Mylotarg (Gemtuzumab ozogamicin), golimumab (Centocor), Cimzia (Certolizumab pegol), Soliris (Eculizumab), CNTO 1275 (ustekinumab), Vectibix (panitumumab), Bexxar (tositumomab and I131 tositumomab), an anti-IL-17 antibody Antibody 7 as described in International Application WO 2007/149032 (Cambridge Antibody Technology), the entire contents of which are incorporated by reference herein, the anti-IL-13 antibody CAT-354 (Cambridge Antibody Technology), the anti-human CD4 antibody CE9y4PE (IDEC-151, clenoliximab) (Biogen IDEC/Glaxo Smith Kline), the anti-human CD4 antibody IDEC CE9.1/SB-210396 (keliximab) (Biogen IDEC), the anti-human CD80 antibody IDEC-114 (galiximab) (Biogen IDEC), the anti-Rabies Virus Protein antibody CR4098 (foravirumab), and the anti-human TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-2) antibody HGS-ETR2 (lexatumumab) (Human Genome Sciences, Inc.),


and Avastin (bevacizumab).


Techniques for the production of antibodies are provided below.


Polyclonal Antibodies


Polyclonal antibodies generally refer to a mixture of antibodies that are specific to a certain antigen, but bind to different epitopes on said antigen. Polyclonal antibodies are generally raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl2, or R1NCNR, where R and R1 are different alkyl groups. Methods for making polyclonal antibodies are known in the art, and are described, for example, in Antibodies: A Laboratory Manual, Lane and Harlow (1988), incorporated by reference herein.


Monoclonal Antibodies


A “monoclonal antibody” as used herein is intended to refer to a hybridoma-derived antibody (e.g., an antibody secreted by a hybridoma prepared by hybridoma technology, such as the standard Kohler and Milstein hybridoma methodology). For example, the monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567). Thus, a hybridoma-derived dual-specificity antibody of the invention is still referred to as a monoclonal antibody although it has antigenic specificity for more than a single antigen.


Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Thus, the modifier “monoclonal” indicates the character of the antibody as not being a mixture of discrete antibodies.


In a further embodiment, antibodies can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990). Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks et al., Bio/Technology, 10:779-783 (1992)), as well as combinatorial infection and in vivo recombination as a strategy for constructing very large phage libraries (Waterhouse et al., Nuc. Acids. Res., 21:2265-2266 (1993)). Thus, these techniques are viable alternatives to traditional monoclonal antibody hybridoma techniques for isolation of monoclonal antibodies.


Antibodies and antibody fragments may also be isolated from yeast and other eukaryotic cells with the use of expression libraries, as described in U.S. Pat. Nos. 6,423,538; 6,696,251; 6,699,658; 6,300,065; 6,399,763; and 6,114,147. Eukaryotic cells may be engineered to express library proteins, including from combinatorial antibody libraries, for display on the cell surface, allowing for selection of particular cells containing library clones for antibodies with affinity to select target molecules. After recovery from an isolated cell, the library clone coding for the antibody of interest can be expressed at high levels from a suitable mammalian cell line.


Additional methods for developing antibodies of interest include cell-free screening using nucleic acid display technology, as described in U.S. Pat. Nos. 7,195,880; 6,951,725; 7,078,197; 7,022,479, 6,518,018; 7,125,669; 6,846,655; 6,281,344; 6,207,446; 6,214,553; 6,258,558; 6,261,804; 6,429,300; 6,489,116; 6,436,665; 6,537,749; 6,602,685; 6,623,926; 6,416,950; 6,660,473; 6,312,927; 5,922,545; and 6,348,315. These methods can be used to transcribe a protein in vitro from a nucleic acid in such a way that the protein is physically associated or bound to the nucleic acid from which it originated. By selecting for an expressed protein with a target molecule, the nucleic acid that codes for the protein is also selected. In one variation on cell-free screening techniques, antibody sequences isolated from immune system cells can be isolated and partially randomized polymerase chain reaction mutagenesis techniques to increase antibody diversity. These partially randomized antibody genes are then expressed in a cell-free system, with concurrent physical association created between the nucleic acid and antibody.


The DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc. Natl. Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.


Typically such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.


Chimeric or hybrid antibodies also may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins may be constructed using a disulfide-exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.


Humanized Antibodies


Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting non-human (e.g., rodent) CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some framework (FR) residues are substituted by residues from analogous sites in rodent antibodies. Additional references which describe the humanization process include Sims et al., J. Immunol., 151:2296 (1993); Chothia et al., J. Mol. Biol., 196:901 (1987); Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993), each of which is incorporated by reference herein.


Human Antibodies


Alternatively, it is now possible to produce transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production. For example, it has been described that the homozygous deletion of the antibody heavy-chain joining region (JH) gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge. See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90:2551 (1993); Jakobovits et al., Nature, 362:255-258 (1993); Bruggermann et al., Year in Immuno., 7:33 (1993). Human antibodies can also be derived from phage-display libraries (Hoogenboom et al., J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581-597 (1991)).


In one embodiment, the formulation of the invention comprises an antibody, or antigen-binding portion thereof, which binds human TNFα, including, for example, adalimumab (also referred to as Humira, adalimumab, or D2E7; Abbott Laboratories). In one embodiment, the antibody, or antigen-binding fragment thereof, dissociates from human TNFα with a Kd of 1×10−8 M or less and a Koff rate constant of 1×10−3 s−1 or less, both determined by surface plasmon resonance, and neutralizes human TNFα cytotoxicity in a standard in vitro L929 assay with an IC50 of 1×10−7 M or less. Examples and methods for making human, neutralizing antibodies which have a high affinity for human TNFα, including sequences of the antibodies, are described in U.S. Pat. No. 6,090,382 (referred to as D2E7), incorporated by reference herein.


In one embodiment, the human neutralizing, antibody, or an antigen-binding portion thereof, having a high affinity for human TNFα is an isolated human antibody, or an antigen-binding portion thereof, with a light chain variable region (LCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO:3, or modified from SEQ ID NO:3 by a single alanine substitution at position 1, 4, 5, 7 or 8, and with a heavy chain variable region (HCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO:4, or modified from SEQ ID NO:4 by a single alanine substitution at position 2, 3, 4, 5, 6, 8, 9, 10 or 11. Preferably, the LCVR further has a CDR2 domain comprising the amino acid sequence of SEQ ID NO:5 (i.e., the D2E7 VL CDR2) and the HCVR further has a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 6 (i.e., the D2E7 VH CDR2). Even more preferably, the LCVR further has CDR1 domain comprising the amino acid sequence of SEQ ID NO:7 (i.e., the D2E7 VL CDR1) and the HCVR has a CDR1 domain comprising the amino acid sequence of SEQ ID NO:8 (i.e., the D2E7 VH CDR1).


In another embodiment, the human neutralizing, antibody, or an antigen-binding portion thereof, having a high affinity for human TNFα is an isolated human antibody, or an antigen-binding portion thereof, with a light chain variable region (LCVR) comprising the amino acid sequence of SEQ ID NO:1 (i.e., the D2E7 VL) and a heavy chain variable region (HCVR) comprising the amino acid sequence of SEQ ID NO:2 (i.e., the D2E7 VH). In certain embodiments, the antibody comprises a heavy chain constant region, such as an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region. Preferably, the heavy chain constant region is an IgG1 heavy chain constant region or an IgG4 heavy chain constant region. Furthermore, the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region. Preferably, the antibody comprises a kappa light chain constant region.


In one embodiment, the formulation of the invention comprises an antibody, or antigen-binding portion thereof, which binds human IL-12, including, for example, the antibody J695 (Abbott Laboratories; also referred to as ABT-874) (U.S. Pat. No. 6,914,128). J695 is a fully human monoclonal antibody designed to target and neutralize interleukin-12 and interleukin-23. In one embodiment, the antibody, or antigen-binding fragment thereof, has the following characteristics: it dissociates from human IL-1α with a KD of 3×10−7 M or less; dissociates from human IL-1β with a KD of 5×10−5 M or less; and does not bind mouse IL-1α or mouse IL-1β. Examples and methods for making human, neutralizing antibodies which have a high affinity for human IL-12, including sequences of the antibody, are described in U.S. Pat. No. 6,914,128, incorporated by reference herein.


In one embodiment, the formulation of the invention comprises an antibody, or antigen-binding portion thereof, which binds human IL-18, including, for example, the antibody 2.5(E)mg1 (Abbott Bioresearch; also referred to as ABT-325) (see U.S. Patent Application No. 2005/0147610, incorporated by reference herein).


In one embodiment, the formulation of the invention comprises an anti-IL-12/anti-IL-23 antibody, or antigen-binding portion thereof, which is the antibody 1D4.7 (Abbott Laboratories; also referred to as ABT-147) (see WO 2007/005608 A2, published Jan. 11, 2007, incorporated by reference herein).


In one embodiment, the formulation of the invention comprises an anti-IL-13 antibody, or antigen-binding portion thereof, which is the antibody 13C5.5 (Abbott Laboratories; also referred to as ABT-308) (see. PCT/US2007/19660 (WO 08/127271), incorporated by reference herein).


In one embodiment, the formulation of the invention comprises an antibody, or antigen-binding portion thereof, which is the antibody 7C6, an anti-amyloid β antibody (Abbott Laboratories; see PCT publication WO 07/062852, incorporated by reference herein).


Bispecific Antibodies


Bispecific antibodies (BsAbs) are antibodies that have binding specificities for at least two different epitopes. Such antibodies can be derived from full length antibodies or antibody fragments (e.g., F(ab′)2 bispecific antibodies).


Methods for making bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829 and in Traunecker et al., EMBO J., 10:3655-3659 (1991).


According to a different approach, antibody variable domains with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant domain sequences.


The fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light chain binding, present in at least one of the fusions. DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of no particular significance.


In a preferred embodiment of this approach, the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690 published Mar. 3, 1994. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986).


Bispecific antibodies include cross-linked or “heteroconjugate” antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.


Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature. The following techniques can also be used for the production of bivalent antibody fragments which are not necessarily bispecific. For example, Fab′ fragments recovered from E. coli can be chemically coupled in vitro to form bivalent antibodies. See, Shalaby et al., J. Exp. Med., 175:217-225 (1992).


Various techniques for making and isolating bivalent antibody fragments directly from recombinant cell culture have also been described. For example, bivalent heterodimers have been produced using leucine zippers. Kostelny et al., J. Immunol., 148(5): 1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab′ portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. The “diabody” technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific/bivalent antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific/bivalent antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al., J. Immunol., 152:5368 (1994).


In one embodiment, the formulation of the invention comprises an antibody which is bispecific for IL-1 (including IL-1α and IL-1β). Examples and methods for making bispecific IL-1 antibodies can be found in U.S. Provisional Appln. No. 60/878,165, filed Dec. 29, 2006.


Diafiltration/Ultrafiltration (also generally referred to herein as DF/UF) selectively utilizes permeable (porous) membrane filters to separate the components of solutions and suspensions based on their molecular size. A membrane retains molecules that are larger than the pores of the membrane while smaller molecules such as salts, solvents and water, which are permeable, freely pass through the membrane. The solution retained by the membrane is known as the concentrate or retentate. The solution that passes through the membrane is known as the filtrate or permeate. One parameter for selecting a membrane for concentration is its retention characteristics for the sample to be concentrated. As a general rule, the molecular weight cut-off (MWCO) of the membrane should be ⅓rd to ⅙th the molecular weight of the molecule to be retained. This is to assure complete retention. The closer the MWCO is to that of the sample, the greater the risk for some small product loss during concentration. Examples of membranes that can be used with methods of the invention include Omega™ PES membrane (30 kDa MWCO, i.e. molecules larger than 30 kDa are retained by the membrane and molecules less than 30 kDa are allowed to pass to the filtrate side of the membrane) (Pall Corp., Port Washington, N.Y.); Millex®-GV Syringe Driven Filter Unit, PVDF 0.22 μm (Millipore Corp., Billerica, Mass.); Millex®-GP Syringe Driven Filter Unit, PES 0.22 μm; Sterivex®0.22 μm Filter Unit (Millipore Corp., Billerica, Mass.); and Vivaspin concentrators (MWCO 10 kDa, PES; MWCO 3 kDa, PES) (Sartorius Corp., Edgewood, N.Y.). In order to prepare a low-ionic protein formulation of the invention, the protein solution (which may be solubilized in a buffered formulation) is subjected to a DF/UF process, whereby water is used as a DF/UF medium. In a preferred embodiment, the DF/UF medium consists of water and does not include any other excipients.


Any water can be used in the DF/UF process of the invention, although a preferred water is purified or deionized water. Types of water known in the art that may be used in the practice of the invention include water for injection (WFI) (e.g., HyPure WFI Quality Water (HyClone), AQUA-NOVA® WFI (Aqua Nova)), UltraPure™ Water (Invitrogen), and distilled water (Invitrogen; Sigma-Aldrich).


There are two forms of DF/UF, including DF/UF in discontinuous mode and DF/UF in continuous mode. The methods of the invention may be performed according to either mode.


Continuous DF/UF (also referred to as constant volume DF/UF) involves washing out the original buffer salts (or other low molecular weight species) in the retentate (sample or first protein solution) by adding water or a new buffer to the retentate at the same rate as filtrate is being generated. As a result, the retentate volume and product concentration does not change during the DF/UF process. The amount of salt removed is related to the filtrate volume generated, relative to the retentate volume. The filtrate volume generated is usually referred to in terms of “diafiltration volumes”. A single diafiltration volume (DV) is the volume of retentate when diafiltration is started. For continuous diafiltration, liquid is added at the same rate as filtrate is generated. When the volume of filtrate collected equals the starting retentate volume, 1 DV has been processed.


Discontinuous DF/UF (examples of which are provided below in the Examples section) includes two different methods, discontinuous sequential DF/UF and volume reduction discontinuous DF/UF. Discontinuous DF/UF by sequential dilution involves first diluting the sample (or first protein solution) with water to a predetermined volume. The diluted sample is then concentrated back to its original volume by UF. Discontinuous DF/UF by volume reduction involves first concentrating the sample to a predetermined volume, then diluting the sample back to its original volume with water or replacement buffer. As with continuous DF/UF, the process is repeated until the level of unwanted solutes, e.g., ionic excipients, are removed.


DF/UF may be performed in accordance with conventional techniques known in the art using water, e.g, WFI, as the DF/UF medium (e.g., Industrial Ultrafiltration Design and Application of Diafiltration Processes, Beaton & Klinkowski, J. Separ. Proc. Technol., 4(2) 1-10 (1983)). Examples of commercially available equipment for performing DF/UF include Millipore Labscale™ TFF System (Millipore), LV Centramate™ Lab Tangential Flow System (Pall Corporation), and the UniFlux System (GE Healthcare).


For example, in a preferred embodiment, the Millipore Labscale™ Tangential Flow Filtration (TFF) system with a 500 mL reservoir is used to perform a method of the invention to produce a diafiltered antibody solution. The DF/UF procedure is performed in a discontinuous manner, with 14 process steps used to produce a high concentration antibody formulation in water. For additional exemplary equipment, solution and water volumes, number of process steps, and other parameters of particular embodiments of the invention, see the Examples section below.


Alternative methods to diafiltration for buffer exchange where a protein is re-formulated into water in accordance with the invention include dialysis and gel filtration, both of which are techniques known to those in the art. Dialysis requires filling a dialysis bag (membrane casing of defined porosity), tying off the bag, and placing the bag in a bath of water. Through diffusion, the concentration of salt in the bag will equilibrate with that in the bath, wherein large molecules, e.g., proteins that cannot diffuse through the bag remain in the bag. The greater the volume of the bath relative to the sample volume in the bags, the lower the equilibration concentration that can be reached. Generally, replacements of the bath water are required to completely remove all of the salt. Gel filtration is a non-adsorptive chromatography technique that separates molecules on the basis of molecular size. In gel filtration, large molecules, e.g., proteins, may be separated from smaller molecules, e.g., salts, by size exclusion.


In a preferred embodiment of the invention, the first protein solution is subjected to a repeated volume exchange with the water, such that an aqueous formulation, which is essentially water and protein, is achieved. The diafiltration step may be performed any number of times, depending on the protein in solution, wherein one diafiltration step equals one total volume exchange. In one embodiment, the diafiltration process is performed 1, 2, 3, 4, 5, 6, 7, 8, 9, or up to as many times are deemed necessary to remove excipients, e.g., salts, from the first protein solution, such that the protein is dissolved essentially in water. A single round or step of diafiltration is achieved when a volume of water has been added to the retentate side that is equal to the starting volume of the protein solution.


In one embodiment, the protein solution is subjected to at least 2 diafiltration steps. In one embodiment, the diafiltration step or volume exchange with water may be repeated at least four times, and preferably at least five times. In one embodiment, the first protein solution is subjected to diafiltration with water until at least a six-fold volume exchange is achieved. In another embodiment, the first protein solution is subjected to diafiltration with water until at least a seven-fold volume exchange is achieved. Ranges intermediate to the above recited numbers, e.g., 4 to 6 or 5 to 7, are also intended to be part of this invention. For example, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.


In a preferred embodiment, loss of protein to the filtrate side of an ultrafiltration membrane should be minimized. The risk of protein loss to the filtrate side of a particular membrane varies in relation to the size of the protein relative to the membrane's pore size, and the protein's concentration. With increases in protein concentration, risk of protein loss to the filtrate increases. For a particular membrane pore size, risk of protein loss is greater for a smaller protein that is close in size to the membrane's MWCO than it is for a larger protein. Thus, when performing DF/UF on a smaller protein, it may not be possible to achieve the same reduction in volume, as compared to performing DF/UF on a larger protein using the same membrane, without incurring unacceptable protein losses. In other words, as compared to the ultrafiltration of a solution of a smaller protein using the same equipment and membrane, a solution of a larger protein could be ultrafiltered to a smaller volume, with a concurrent higher concentration of protein in the solution. DF/UF procedures using a particular pore size membrane may require more process steps for a smaller protein than for a larger protein; a greater volume reduction and concentration for a larger protein permits larger volumes of water to be added back, leading to a larger dilution of the remaining buffer or excipient ingredients in the protein solution for that individual process step. Fewer process steps may therefore be needed to achieve a certain reduction in solutes for a larger protein than for a smaller one. A person with skill in the art would be able to calculate the amount of concentration possible with each process step and the number of overall process steps required to achieve a certain reduction in solutes, given the protein size and the pore size of the ultrafiltration device to be used in the procedure.


As a result of the diafiltration methods of the invention, the concentration of solutes in the first protein solution is significantly reduced in the final aqueous formulation comprising essentially water and protein. For example, the aqueous formulation may have a final concentration of excipients which is at least 95% less than the first protein solution, and preferably at least 99% less than the first protein solution. For example, in one embodiment, to dissolve a protein in WFI is a process that creates a theoretical final excipient concentration, reached by constant volume diafiltration with five diafiltration volumes, that is equal or approximate to Ci e−5=0.00674, i.e., an approximate 99.3% maximum excipient reduction. In one embodiment, a person with skill in the art may perform 6 volume exchanges during the last step of a commercial DF/UF with constant volume diafiltration, i.e., Ci would be Ci e−6=0.0025. This would provide an approximate 99.75% maximum theoretical excipient reduction. In another embodiment, a person with skill in the art may use 8 diafiltration volume exchanges to obtain a theoretical ˜99.9% maximum excipient reduction.


The term “excipient-free” or “free of excipients” indicates that the formulation is essentially free of excipients. In one embodiment, excipient-free indicates buffer-free, salt-free, sugar-free, amino acid-free, surfactant-free, and/or polyol free. In one embodiment, the term “essentially free of excipients” indicates that the solution or formulation is at least 99% free of excipients. It should be noted, however, that in certain embodiments, a formulation may comprise a certain specified non-ionic excipient, e.g., sucrose or mannitol, and yet the formulation is otherwise excipient free. For example, a formulation may comprise water, a protein, and mannitol, wherein the formulation is otherwise excipient free. In another example, a formulation may comprise water, a protein, and polysorbate 80, wherein the formulation is otherwise excipient free. In yet another example, the formulation may comprise water, a protein, a sorbitol, and polysorbate 80, wherein the formulation is otherwise excipient free.


When water is used for diafiltering a first protein solution in accordance with the methods described herein, ionic excipients will be washed out, and, as a result, the conductivity of the diafiltered aqueous formulation is lower than the first protein solution. If an aqueous solution conducts electricity, then it must contain ions, as found with ionic excipients. A low conductivity measurement is therefore indicative that the aqueous formulation of the invention has significantly reduced excipients, including ionic excipients.


Conductivity of a solution is measured according to methods known in the art. Conductivity meters and cells may be used to determine the conductivity of the aqueous formulation, and should be calibrated to a standard solution before use. Examples of conductivity meters available in the art include MYRON L Digital (Cole Parmer), Conductometer (Metrohm AG), and Series 3105/3115 Integrated Conductivity Analyzers (Kemotron). In one embodiment, the aqueous formulation has a conductivity of less than 3 mS/cm. In another embodiment, the aqueous formulation has a conductivity of less than 2 mS/cm. In yet another embodiment, the aqueous formulation has a conductivity of less than 1 mS/cm. In one aspect of the invention, the aqueous formulation has a conductivity of less than 0.5 mS/cm. Ranges intermediate to the above recited numbers, e.g., 1 to 3 mS/cm, are also intended to be encompassed by the invention. For example, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included. In addition, values that fall within the recited numbers are also included in the invention, e.g., 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0 and so forth.


An important aspect of the invention is that the diafiltered protein solution (solution obtained following the diafiltration process of the first protein solution) can be concentrated. By following this process, it has been discovered that high concentrations of protein are stable in water. Concentration following diafiltration results in an aqueous formulation containing water and an increased protein concentration relative to the first protein solution. Thus, the invention also includes diafiltering a protein solution using water as a diafiltration medium and subsequently concentrating the resulting aqueous solution. Concentration of the diafiltered protein solution may be achieved through means known in the art, including centrifugation. For example, following diafiltration, the water-based diafiltrated protein solution is subjected to a centrifugation process which serves to concentrate the protein via ultrafiltration into a high concentration formulation while maintaining the water-based solution. Means for concentrating a solution via centrifugation with ultrafiltration membranes and/or devices are known in the art, e.g., with Vivaspin centrifugal concentrators (Sartorius Corp. Edgewood, N.Y.).


The methods of the invention provide a means of concentrating a protein at very high levels in water without the need for additional stabilizing agents. The concentration of the protein in the aqueous formulation obtained using the methods of the invention can be any amount in accordance with the desired concentration. For example, the concentration of protein in an aqueous solution made according to the methods herein is at least about 10 μg/mL; at least about 1 mg/mL; at least about 10 mg/mL; at least about 20 mg/mL; at least about 50 mg/mL; at least about 75 mg/mL; at least about 100 mg/mL; at least about 125 mg/mL; at least about 150 mg/mL; at least about 175 mg/mL; at least about 200 mg/mL; at least about 220 mg/mL; at least about 250 mg/mL; at least about 300 mg/mL; or greater than about 300 mg/mL. Ranges intermediate to the above recited concentrations, e.g., at least about 113 mg/mL, at least about 214 mg/mL, and at least about 300 mg/mL, are also intended to be encompassed by the invention. In addition, ranges of values using a combination of any of the above recited values (or values between the ranges described above) as upper and/or lower limits are intended to be included, e.g., 100 to 125 mg/mL, 113 to 125 mg/mL, and 126 to 200 mg/mL or more.


The methods of the invention provide the advantage that the resulting formulation has a low percentage of protein aggregates, despite the high concentration of the aqueous protein formulation. In one embodiment, the aqueous formulations comprising water and a high concentration of a protein, e.g., antibodies, contains less than about 5% protein aggregates, even in the absence of a surfactant or other type of excipient. In one embodiment, the formulation comprises no more than about 7.3% aggregate protein; the formulation comprises no more than about 5% aggregate protein; the formulation comprises no more than about 4% aggregate protein; the formulation comprises no more than about 3% aggregate protein; the formulation comprises no more than about 2% aggregate protein; or the formulation comprising no more than about 1% aggregate protein. In one embodiment, the formulation comprises at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% monomer protein. Ranges intermediate to the above recited concentrations, e.g., at least about 98.6%, no more than about 4.2%, are also intended to be part of this invention. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.


Many protein-based pharmaceutical products need to be formulated at high concentrations. For example, antibody-based products increasingly tend to exceed 100 mg/mL in their Drug Product (DP) formulation to achieve appropriate efficacy and meet a typical patient usability requirement of a maximal ˜1 mL injection volume. Accordingly, downstream processing steps, such as diafiltration into the final formulation buffer or ultrafiltration to increase the protein concentration, are also conducted at higher concentrations.


Classic thermodynamics predicts that intermolecular interactions can affect the partitioning of small solutes across a dialysis membrane, especially at higher protein concentrations, and models describing non-ideal dialysis equilibrium and the effects of intermolecular interactions are available (Tanford Physical chemistry or macromolecules. New York, John Wiley and Sons, Inc., p. 182, 1961; Tester and Modell Thermodynamics and its applications, 3rd ed. Upper Saddle River, NL, Prentice-Hall, 1997). In the absence of the availability of detailed thermodynamic data in the process development environment, which is necessary to apply these type of models, intermolecular interactions rarely are taken into account during the design of commercial DF/UF operations. Consequently, DP excipient concentrations may differ significantly from the concentration labeled. Several examples of this discrepancy in commercial and development products are published, e.g., chloride being up to 30% lower than labeled in an IL-1 receptor antagonist, histidine being 40% lower than labeled in a PEG-sTNF receptor, and acetate being up to 200% higher than labeled in a fusion conjugate protein (Stoner et al., J. Pharm. Sci., 93, 2332-2342 (2004)). There are several reasons why the actual DP may be different from the composition of the buffer the protein is diafiltered into, including the Donnan effect (Tombs and Peacocke (1974) Oxford; Clarendon Press), non-specific interactions (Arakawa and Timasheff, Arch. Biochem. Biophys., 224, 169-77 (1983); Timasheff, Annu. Rev. Biophys. Biomol. Struct., 22, 67-97 (1993)), and volume exclusion effects. Volume exclusion includes most protein partial specific volumes are between 0.7 and 0.8 mL/g.5 Thus, for a globular protein at 100 mg/mL, protein molecules occupy approx. 7.5% of the total solution volume. No significant intermolecular interactions assumed, this would translate to a solute molar concentration on the retentate side of the membrane that is 92.5% of the molar concentration on the permeate side of the membrane. This explains why basically all protein solution compositions necessarily change during ultrafiltration processing. For instance, at 40 mg/mL the protein molecules occupy approx. 3% of the total solution volume, and an ultrafiltration step increasing the concentration to 150 mg/mL will necessarily induce molar excipient concentrations to change by more than 8% (as protein at 150 mg/mL accounts for more than 11% of total solution volume). Ranges intermediate to the above recited percentages are also intended to be part of this invention. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.


In accordance with the methods and compositions of the invention, buffer composition changes during DF/UF operations can be circumvented by using pure water as diafiltration medium. By concentrating the protein ˜20% more than the concentration desired in the final Bulk DS, excipients could subsequently be added, for instance, via highly concentrated excipient stock solutions. Excipient concentrations and solution pH could then be guaranteed to be identical as labeled.


The aqueous formulation of the invention provides an advantage as a starting material, as it essentially contains no excipient. Any excipient(s) which is added to the formulation following the diafiltration in water can be accurately calculated, i.e., pre-existing concentrations of excipient(s) do not interfere with the calculation. Examples of pharmaceutically acceptable excipients are described in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980), incorporated by reference herein. Thus, another aspect of the invention includes using the aqueous formulation obtained through the methods described herein, for the preparation of a formulation, particularly a pharmaceutical formulation, having known concentrations of excipient(s), including non-ionic excipient(s) or ionic excipient(s). One aspect of the invention includes an additional step where an excipient(s) is added to the aqueous formulation comprising water and protein. Thus, the methods of the invention provide an aqueous formulation which is essentially free of excipients and may be used as a starting material for preparing formulations comprising water, proteins, and specific concentrations of excipients.


In one embodiment, the methods of the invention may be used to add non-ionic excipients, e.g., sugars or non-ionic surfactants, such as polysorbates and poloxamers, to the formulation without changing the characteristics, e.g., protein concentration, hydrodynamic diameter of the protein, conductivity, etc.


Additional characteristics and advantages of aqueous formulations obtained using the above methods are described below in section III. Exemplary protocols for performing the methods of the invention are also described below in the Examples.


III. Formulations of Invention

The invention provide an aqueous formulation comprising a protein and water which has a number of advantages over conventional formulations in the art, including stability of the protein in water without the requirement for additional excipients, increased concentrations of protein without the need for additional excipients to maintain solubility of the protein, and low osmolality. The formulations of the invention also have advantageous storage properties, as the proteins in the formulation remain stable during storage, e.g., stored as a liquid form for more than 3 months at 7° C. or freeze/thaw conditions, even at high protein concentrations and repeated freeze/thaw processing steps. In one embodiment, formulations of the invention include high concentrations of proteins such that the aqueous formulation does not show significant opalescence, aggregation, or precipitation.


The aqueous formulation of the invention does not rely on standard excipients, e.g., a tonicity modifier, a stabilizing agent, a surfactant, an anti-oxidant, a cryoprotectant, a bulking agent, a lyroprotectant, a basic component, and an acidic component. In other embodiments of the invention, the formulation contains water, one or more proteins, and no ionic excipients (e.g., salts, free amino acids).


In certain embodiments, the aqueous formulation of the invention comprises a protein concentration of at least 50 mg/mL and water, wherein the formulation has an osmolality of no more than 30 mOsmol/kg. Lower limits of osmolality of the aqueous formulation are also encompassed by the invention. In one embodiment the osmolality of the aqueous formulation is no more than 15 mOsmol/kg. The aqueous formulation of the invention may have an osmolality of less than 30 mOsmol/kg, and also have a high protein concentration, e.g., the concentration of the protein is at least 100 mg/mL, and may be as much as 200 mg/mL or greater. Ranges intermediate to the above recited concentrations and osmolality units are also intended to be part of this invention. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.


The concentration of the aqueous formulation of the invention is not limited by the protein size and the formulation may include any size range of proteins. Included within the scope of the invention is an aqueous formulation comprising at least 50 mg/mL and as much as 200 mg/mL or more of a protein, which may range in size from 5 kDa to 150 kDa or more. In one embodiment, the protein in the formulation of the invention is at least about 15 kD in size, at least about 20 kD in size; at least about 47 kD in size; at least about 60 kD in size; at least about 80 kD in size; at least about 100 kD in size; at least about 120 kD in size; at least about 140 kD in size; at least about 160 kD in size; or greater than about 160 kD in size. Ranges intermediate to the above recited sizes are also intended to be part of this invention. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included.


The aqueous formulation of the invention may be characterized by the hydrodynamic diameter (Dh) of the proteins in solution. The hydrodynamic diameter of the protein in solution may be measured using dynamic light scattering (DLS), which is an established analytical method for determining the Dh of proteins. Typical values for monoclonal antibodies, e.g., IgG, are about 10 nm. Low-ionic formulations, like those described herein, may be characterized in that the Dh of the proteins are notably lower than protein formulations comprising ionic excipients. It has been discovered that the Dh values of antibodies in aqueous formulations made using the DF/UF process using pure water as an exchange medium, are notably lower than the Dh of antibodies in conventional formulations independent of protein concentration. In one embodiment, antibodies in the aqueous formulation of the invention have a Dh of less than 4 nm, or less than 3 nm.


In one embodiment, the Dh of the protein in the aqueous formulation is smaller relative to the Dh of the same protein in a buffered solution, irrespective of protein concentration. Thus, in certain embodiments, protein in an aqueous formulation made in accordance with the methods described herein, will have a Dh which is at least 25% less than the Dh of the protein in a buffered solution at the same given concentration. Examples of buffered solutions include, but are not limited to phosphate buffered saline (PBS). In certain embodiments, proteins in the aqueous formulation of the invention have a Dh that is at least 50% less than the Dh of the protein in PBS in at the given concentration; at least 60% less than the Dh of the protein in PBS at the given concentration; at least 70% less than the Dh of the protein in PBS at the given concentration; or more than 70% less than the Dh of the protein in PBS at the given concentration. Ranges intermediate to the above recited percentages are also intended to be part of this invention, e.g., 55%, 56%, 57%, 64%, 68%, and so forth. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included, e.g., 50% to 80%.


Protein aggregation is a common problem in protein solutions, and often results from increased concentration of the protein. The instant invention provides a means for achieving a high concentration, low protein aggregation formulation. Formulations of the invention do not rely on a buffering system and excipients, including surfactants, to keep proteins in the formulation soluble and from aggregating. Formulations of the invention can be advantageous for therapeutic purposes, as they are high in protein concentration and water-based, not relying on other agents to achieve high, stable concentrations of proteins in solution.


The majority of biologic products (including antibodies) are subject to numerous degradative processes which frequently arise from non-enzymatic reactions in solution. These reactions may have a long-term impact on product stability, safety and efficacy. These instabilities can be retarded, if not eliminated, by storage of product at subzero temperatures, thus gaining a tremendous advantage for the manufacturer in terms of flexibility and availability of supplies over the product life-cycle. Although freezing is often the safest and most reliable method of biologics product storage, it has inherent risks. Freezing can induce stress in proteins through cold denaturation, by introducing ice-liquid interfaces, and by freeze-concentration (cryoconcentration) of solutes when the water crystallizes.


Cryoconcentration is a process in which a flat, uncontrolled moving ice front is formed during freezing that excludes solute molecules (small molecules such as sucrose, salts, and other excipients typically used in protein formulation, or macromolecules such as proteins), leading to zones in which proteins may be found at relatively high concentration in the presence of other solutes at concentrations which may potentially lead to local pH or ionic concentration extremes. For most proteins, these conditions can lead to denaturation and in some cases, protein and solute precipitation. Since buffer salts and other solutes are also concentrated under such conditions, these components may reach concentrations high enough to lead to pH and/or redox changes in zones within the frozen mass. The pH shifts observed as a consequence of buffer salt crystallization (e.g., phosphates) in the solutions during freezing can span several pH units, which may impact protein stability.


Concentrated solutes may also lead to a depression of the freezing point to an extent where the solutes may not be frozen at all, and proteins will exist within a solution under these adverse conditions. Often, rapid cooling may be applied to reduce the time period the protein is exposed to these undesired conditions. However, rapid freezing can induce a large-area ice-water interface, whereas slow cooling induces smaller interface areas. For instance, rapid cooling of six model proteins during one freeze/thaw step was shown to reveal a denaturation effect greater than 10 cycles of slow cooling, demonstrating the great destabilization potential of hydrophobic ice surface-induced denaturation.


The aqueous formulation of the invention has advantageous stability and storage properties. Stability of the aqueous formulation is not dependent on the form of storage, and includes, but is not limited to, formulations which are frozen, lyophilized, or spray-dried. Stability can be measured at a selected temperature for a selected time period. In one aspect of the invention, the protein in the aqueous formulations is stable in a liquid form for at least 3 months; at least 4 months, at least 5 months; at least 6 months; at least 12 months. Ranges intermediate to the above recited time periods are also intended to be part of this invention, e.g., 9 months, and so forth. In addition, ranges of values using a combination of any of the above recited values as upper and/or lower limits are intended to be included. Preferably, the formulation is stable at room temperature (about 30° C.) or at 40° C. for at least 1 month and/or stable at about 2-8° C. for at least 1 year, or more preferably stable at about 2-8° C. for at least 2 years. Furthermore, the formulation is preferably stable following freezing (to, e.g., −80° C.) and thawing of the formulation, hereinafter referred to as a “freeze/thaw cycle.”


Stability of a protein can be also be defined as the ability to remain biologically active. A protein “retains its biological activity” in a pharmaceutical formulation, if the protein in a pharmaceutical formulation is biologically active upon administration to a subject. For example, biological activity of an antibody is retained if the biological activity of the antibody in the pharmaceutical formulation is within about 30%, about 20%, or about 10% (within the errors of the assay) of the biological activity exhibited at the time the pharmaceutical formulation was prepared (e.g., as determined in an antigen binding assay).


Stability of a protein in an aqueous formulation may also be defined as the percentage of monomer, aggregate, or fragment, or combinations thereof, of the protein in the formulation. A protein “retains its physical stability” in a formulation if it shows substantially no signs of aggregation, precipitation and/or denaturation upon visual examination of color and/or clarity, or as measured by UV light scattering or by size exclusion chromatography. In one aspect of the invention, a stable aqueous formulation is a formulation having less than about 10%, and preferably less than about 5% of the protein being present as aggregate in the formulation.


Another characteristic of the aqueous formulation of the invention is that, in some instances, diafiltering a protein using water results in an aqueous formulation having improved viscosity features in comparison to the first protein solution (i.e., the viscosity of the diafiltered protein solution is reduced in comparison to the first protein solution.) A person with skill in the art will recognize that multiple methods for measuring viscosity can be used in the preparation of formulations in various embodiments of the invention. For example, kinematic viscosity data (cSt) may be generated using capillaries. In other embodiments, dynamic viscosity data is stated, either alone or with other viscosity data. The dynamic viscosity data may be generated by multiplying the kinematic viscosity data by the density.


In one embodiment, the invention also provides a method for adjusting a certain characteristic, such as the osmolality and/or viscosity, as desired in high protein concentration-water solutions, by adding non-ionic excipients, such as mannitol, without changing other desired features, such as non-opalescence. As such, it is within the scope of the invention to include formulations which are water-based and have high concentrations of protein, where, either during or following the transfer of the protein to water or during the course of the diafiltration, excipients are added which improve, for example, the osmolality or viscosity features of the formulation. Thus, it is also within the scope of the invention that such non-ionic excipients could be added during the process of the transfer of the protein into the final low ionic formulation. Examples of non-ionizable excipients which may be added to the aqueous formulation of the invention for altering desired characteristics of the formulation include, but are not limited to, mannitol, sorbitol, a non-ionic surfactant (e.g., polysorbate 20, polysorbate 40, polysorbate 60 or polysorbate 80), sucrose, trehalose, raffinose, and maltose.


The formulation herein may also contain more than one protein. With respect to pharmaceutical formulations, an additional, distinct protein may be added as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect the other protein. For example, it may be desirable to provide two or more antibodies which bind to TNF or IL-12 in a single formulation. Furthermore, anti-TNF or anti-IL12 antibodies may be combined in the one formulation. Such proteins are suitably present in combination in amounts that are effective for the purpose intended.


Examples of proteins that may be included in the aqueous formulation include antibodies, or antigen-binding fragments thereof. Examples of different types of antibodies, or antigen-binding fragments thereof, that may be used in the invention include, but are not limited to, a chimeric antibody, a human antibody, a humanized antibody, and a domain antibody (dAb). In one embodiment, the antibody used in the methods and compositions of the invention is an anti-TNFα antibody, or antigen-binding portion thereof, or an anti-IL-12 antibody, or antigen binding portion thereof. Additional examples of an antibody, or antigen-binding fragment thereof, that may be used in the invention includes, but is not limited to, 1D4.7 (anti-IL-12/anti-IL-23; Abbott Laboratories), 2.5(E)mg1 (anti-IL-18; Abbott Laboratories), 13C5.5 (anti-Il-13; Abbott Laboratories), J695 (anti-IL-12; Abbott Laboratories), Afelimomab (Fab 2 anti-TNF; Abbott Laboratories), Humira (adalimumab (D2E7); Abbott Laboratories), Campath (Alemtuzumab), CEA-Scan Arcitumomab (fab fragment), Erbitux (Cetuximab), Herceptin (Trastuzumab), Myoscint (Imciromab Pentetate), ProstaScint (Capromab Pendetide), Remicade (Infliximab), ReoPro (Abciximab), Rituxan (Rituximab), Simulect (Basiliximab), Synagis (Palivizumab), Verluma (Nofetumomab), Xolair (Omalizumab), Zenapax (Daclizumab), Zevalin (Ibritumomab Tiuxetan), Orthoclone OKT3 (Muromonab-CD3), Panorex (Edrecolomab), and Mylotarg (Gemtuzumab ozogamicin) golimumab (Centocor), Cimzia (Certolizumab pegol), Soliris (Eculizumab), CNTO 1275 (ustekinumab), Vectibix (panitumumab), Bexxar (tositumomab and I131 tositumomab) and Avastin (bevacizumab).


In one alternative, the protein is a therapeutic protein, including, but not limited to, Pulmozyme (Dornase alfa), Regranex (Becaplermin), Activase (Alteplase), Aldurazyme (Laronidase), Amevive (Alefacept), Aranesp (Darbepoetin alfa), Becaplermin Concentrate, Betaseron (Interferon beta-1b), BOTOX (Botulinum Toxin Type A), Elitek (Rasburicase), Elspar (Asparaginase), Epogen (Epoetin alfa), Enbrel (Etanercept), Fabrazyme (Agalsidase beta), Infergen (Interferon alfacon-1), Intron A (Interferon alfa-2a), Kineret (Anakinra), MYOBLOC (Botulinum Toxin Type B), Neulasta (Pegfilgrastim), Neumega (Oprelvekin), Neupogen (Filgrastim), Ontak (Denileukin diftitox), PEGASYS (Peginterferon alfa-2a), Proleukin (Aldesleukin), Pulmozyme (Dornase alfa), Rebif (Interferon beta-1a), Regranex (Becaplermin), Retavase (Reteplase), Roferon-A (Interferon alfa-2), TNKase (Tenecteplase), and Xigris (Drotrecogin alfa), Arcalyst (Rilonacept), NPlate (Romiplostim), Mircera (methoxypolyethylene glycol-epoetin beta), Cinryze (C1 esterase inhibitor), Elaprase (idursulfase), Myozyme (alglucosidase alfa), Orencia (abatacept), Naglazyme (galsulfase), Kepivance (palifermin) and Actimmune (interferon gamma-1b).


Other examples of proteins which may be included in the methods and compositions described herein, include mammalian proteins, including recombinant proteins thereof, such as, e.g., growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; α-1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VIIIC, factor IX, tissue factor, and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or tissue-type plasminogen activator (t-PA); bombazine; thrombin; tumor necrosis factor-α and -β enkephalinase; RANTES (regulated on activation normally T-cell expressed and secreted); human macrophage inflammatory protein (MIP-1-α); serum albumin such as human serum albumin; mullerian-inhibiting substance; relaxin A-chain; relaxin B-chain; prorelaxin; mouse gonadotropin-associated peptide; DNase; inhibin; activin; vascular endothelial growth factor (VEGF); receptors for hormones or growth factors; an integrin; protein A or D; rheumatoid factors; a neurotrophic factor such as bone-derived neurotrophic factor (BDNF), neurotrophin-3, -4, -5, or -6 (NT-3, NT4, NT-5, or NT-6), or a nerve growth factor such as NGF-β; platelet-derived growth factor (PDGF); fibroblast growth factor such as aFGF and bFGF; epidermal growth factor (EGF); transforming growth factor (TGF) such as TGFα and TGF-β, including TGF-β 1, TGF-β 2, TGF-β 3, TGF-β 4, or TGF-β 5; insulin-like growth factor-I and -II (IGF-I and IGF-II); des(1-3)-IGF-I (brain IGF-I); insulin-like growth factor binding proteins; CD proteins such as CD3, CD4, CD8, CD19 and CD20; erythropoietin (EPO); thrombopoietin (TPO); osteoinductive factors; immunotoxins; a bone morphogenetic protein (BMP); an interferon such as interferon-α, -β., and -γ.; colony stimulating factors (CSFs), e.g., M-CSF, GM-CSF, and G-CSF; interleukins (ILs), e.g., IL-1 to IL-10; superoxide dismutase; T-cell receptors; surface membrane proteins; decay accelerating factor (DAF); a viral antigen such as, for example, a portion of the AIDS envelope; transport proteins; homing receptors; addressins; regulatory proteins; immunoadhesins; antibodies; and biologically active fragments or variants of any of the above-listed polypeptides.


IV. Uses of Invention

The formulations of the invention may be used both therapeutically, i.e., in vivo, or as reagents for in vitro or in situ purposes.


Therapeutic Uses


The methods of the invention may also be used to make a water-based formulation having characteristics which are advantageous for therapeutic use. The aqueous formulation may be used as a pharmaceutical formulation to treat a disorder in a subject.


The formulation of the invention may be used to treat any disorder for which the therapeutic protein is appropriate for treating. A “disorder” is any condition that would benefit from treatment with the protein. This includes chronic and acute disorders or diseases including those pathological conditions which predispose the mammal to the disorder in question. In the case of an anti-TNFα antibody, a therapeutically effective amount of the antibody may be administered to treat an autoimmune disease, such as rheumatoid arthritis, an intestinal disorder, such as Crohn's disease, a spondyloarthropathy, such as ankylosing spondylitis, or a skin disorder, such as psoriasis. In the case of an anti-IL-12 antibody, a therapeutically effective amount of the antibody may be administered to treat a neurological disorder, such as multiple sclerosis, or a skin disorder, such as psoriasis. Other examples of disorders in which the formulation of the invention may be used to treat include cancer, including breast cancer, leukemia, lymphoma, and colon cancer.


The term “subject” is intended to include living organisms, e.g., prokaryotes and eukaryotes. Examples of subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. In specific embodiments of the invention, the subject is a human.


The term “treatment” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder, as well as those in which the disorder is to be prevented.


The aqueous formulation may be administered to a mammal, including a human, in need of treatment in accordance with known methods of administration. Examples of methods of administration include intravenous administration, such as a bolus or by continuous infusion over a period of time, intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intra-articular, intrasynovial, intrathecal, intradermal, transdermal, oral, topical, or inhalation administration.


In one embodiment, the aqueous formulation is administered to the mammal by subcutaneous administration. For such purposes, the formulation may be injected using a syringe, as well as other devices including injection devices (e.g., the Inject-ease and Genject devices); injector pens (such as the GenPen); needleless devices (e.g., MediJector and Biojectorr 2000); and subcutaneous patch delivery systems. In one embodiment, the device, e.g., a syringe, autoinjector pen, contains a needle with a gauge ranging in size from 25 G or smaller in diameter. In one embodiment, the needle gauge ranges in size from 25 G to 33 G (including ranges intermediate thereto, e.g., 25 sG, 26, 26 sG, 27 G, 28 G, 29 G, 30 G, 31 G, 32 G, and 33 G). In a preferred embodiment, the smallest needle diameter and appropriate length is chosen in accordance with the viscosity characteristics of the formulation and the device used to deliver the formulation of the invention.


One advantage of the methods/compositions of the invention is that they provide large concentrations of a protein in a solution which may be ideal for administering the protein to a subject using a needleless device. Such a device allows for dispersion of the protein throughout the tissue of a subject without the need for an injection by a needle. Examples of needleless devices include, but are not limited to, Biojectorr 2000 (Bioject Medical Technologies), Cool. Click (Bioject Medical Technologies), Iject (Bioject Medical Technologies), Vitajet 3, (Bioject Medical Technologies), Mhi500 (The Medical House PLC), Injex 30 (INJEX—Equidyne Systems), Injex 50 (INJEX—Equidyne Systems), Injex 100 (INJEX-Equidyne Systems), Jet Syringe (INJEX—Equidyne Systems), Jetinjector (Becton-Dickinson), J-Tip (National Medical Devices, Inc.), Medi-Jector VISION (Antares Pharma), MED-JET (MIT Canada, Inc.), DermoJet (Akra Dermojet), Sonoprep (Sontra Medical Corp.), PenJet (PenJet Corp.), MicroPor (Altea Therapeutics), Zeneo (Crossject Medical Technology), Mini-Ject (Valeritas Inc.), ImplaJect (Caretek Medical LTD), Intraject (Aradigm), and Serojet (Bioject Medical Technologies).


Also included in the invention are delivery devices that house the aqueous formulation. Examples of such devices include, but are not limited to, a syringe, a pen (such as an autoinjector pen), an implant, an inhalation device, a needleless device, and a patch. An example of an autoinjection pen is described in U.S. application Ser. No. 11/824,516, filed Jun. 29, 2007.


The invention also includes methods of delivering the formulations of the invention by inhalation and inhalation devices containing said formulation for such delivery. In one embodiment, the aqueous formulation is administered to a subject via inhalation using a nebulizer or liquid inhaler. Generally, nebulizers use compressed air to deliver medicine as wet aerosol or mist for inhalation, and, therefore, require that the drug be soluble in water. Types of nebulizers include jet nebulizers (air-jet nebulizers and liquid-jet nebulizers) and ultrasonic nebulizers.


Examples of nebulizers include Akita™ (Activaero GmbH) (see US2001037806, EP1258264). Akita™ is a table top nebulizer inhalation system (Wt: 7.5 kg, B×W×H: 260×170×270) based on Pari's LC Star that provides full control over patient's breathing pattern. The device can deliver as much as 500 mg drug in solution in less than 10 min with a very high delivery rates to the lung and the lung periphery. 65% of the nebulized particles are below 5 microns and the mass median aerodynamic diameter (MMAD) is 3.8 microns at 1.8 bar. The minimum fill volume is 2 mL and the maximum volume is 8 mL. The inspiratory flow (200 mL/sec) and nebulizer pressure (0.3-1.8 bar) are set by the smart card. The device can be individually adjusted for each patient on the basis of a lung function test.


Another example of a nebulizer which may be used with compositions of the invention includes the Aeroneb® Go/Pro/Lab nebulizers (AeroGen). The Aeroneb® nebulizer is based on OnQ™ technology, i.e., an electronic micropump (⅜ inch in diameter and wafer-thin) comprised of a unique dome-shaped aperture plate that contains over 1,000 precision-formed tapered holes, surrounded by a vibrational element. Aeroneb® Go is a portable unit for home use, whereas Aeroneb® Pro is a reusable and autoclavable device for use in hospital and ambulatory clinic, and Aeroneb® Lab is a device for use in pre-clinical aerosol research and inhalation studies. The features of the systems include optimization and customization of aerosol droplet size; low-velocity aerosol delivery with a precisely controlled droplet size, aiding targeted drug delivery within the respiratory system; flexibility of dosing; accommodation of a custom single dose ampoule containing a fixed volume of drug in solution or suspension, or commercially available solutions for use in general purpose nebulizers; continuous, breath-activated or programmable; and adaptable to the needs of a broad range of patients, including children and the elderly; single or multi-patient use.


Aerocurrent™ (AerovertRx corp) may also be used with compositions of the invention (see WO2006006963). This nebulizer is a portable, vibrating mesh nebulizer that features a disposable, pre-filled or user filled drug cartridge.


Staccato™ (Alexza Pharma) may also be used with compositions of the invention (see WO03095012). The key to Staccato™ technology is vaporization of a drug without thermal degradation, which is achieved by rapidly heating a thin film of the drug. In less than half a second, the drug is heated to a temperature sufficient to convert the solid drug film into a vapor. The inhaler consists of three core components: a heating substrate, a thin film of drug coated on the substrate, and an airway through which the patient inhales. The inhaler is breath-actuated with maximum dose delivered to be 20-25 mg and MMAD in the 1-2 micron range.


AERx® (Aradigm) may also be used with compositions of the invention (see WO9848873, U.S. Pat. Nos. 5,469,750, 5,509,404, 5,522,385, 5,694,919, 5,735,263, 5,855,564). AERx® is a hand held battery operated device which utilizes a piston mechanism to expel formulation from the AERx® Strip. The device monitors patients inspiratory air flow and fires only when optimal breathing pattern is achieved. The device can deliver about 60% of the dose as emitted dose and 50-70% of the emitted dose into deep lung with <25% inter-subject variability.


Another example of a nebulizer device which may also be used with compositions of the invention includes Respimat® (Boehringer). Respimat® is a multi-dose reservoir system that is primed by twisting the device base, which is compressed a spring and transfers a metered volume of formulation from the drug cartridge to the dosing chamber. When the device is actuated, the spring is released, which forces a micro-piston into the dosing chamber and pushes the solution through a uniblock; the uniblock consists of a filter structure with two fine outlet nozzle channels. The MMAD generated by the Respimat® is 2 um, and the device is suitable for low dose drugs traditionally employed to treat respiratory disorders.


Compositions of the invention may also be delivered using the Collegium Nebulizer™ (Collegium Pharma), which is a nebulizer system comprised of drug deposited on membrane. The dosage form is administered to a patient through oral or nasal inhalation using the Collegium Nebulizer after reconstitution with a reconstituting solvent.


Another example of a nebulizer device which may also be used with compositions of the invention includes the Inspiration® 626 (Respironics). The 626 is a compressor based nebulizer for home care. The 626 delivers a particle size between 0.5 to 5 microns.


Nebulizers which can be used with compositions of the invention may include Adaptive Aerosol Delivery® technology (Respironics), which delivers precise and reproducible inhaled drug doses to patients regardless of the age, size or variability in breathing patterns of such patients. AAD® systems incorporate electronics and sensors within the handpiece to monitor the patient's breathing pattern by detecting pressure changes during inspiration and expiration. The sensors determine when to pulse the aerosol delivery of medication during the first part of inspiration. Throughout the treatment, the sensors monitor the preceding three breaths and adapt to the patient's inspiratory and expiratory pattern. Because AAD® systems only deliver medication when the patient is breathing through the mouthpiece, these devices allow the patient to take breaks in therapy without medication waste. Examples of AAD® system nebulizers include the HaloLite® AAD®, ProDose® AAD®, and 1-Neb® AAD®.


The HaloLite® Adaptive Aerosol Delivery (AAD)® (Respironics) is a pneumatic aerosolisation system powered by a portable compressor. The AAD® technology monitors the patient's breathing pattern (typically every ten milliseconds) and, depending upon the system being used, either releases pulses of aerosolized drug into specific parts of the inhalation, or calculates the dose drawn during inhalation from a “standing aerosol cloud” (see EP 0910421, incorporated by reference herein).


The ProDos AAD® (Respironics) is a nebulizing system controlled by “ProDose Disc™” system. (Respironics). ProDos AAD® is a pneumatic aerosol system powered by a portable compressor, in which the dose to be delivered is controlled by a microchip-containing disc inserted in the system that, among other things, instructs the system as to the dose to deliver. The ProDose Disc™ is a plastic disc containing a microchip, which is inserted into the ProDose AAD® System and instructs it as to what dose to deliver, the number of doses, which may be delivered together with various control data including drug batch code and expiry date (see EP1245244, incorporated by reference herein). Promixin® can be delivered via ProdoseAAD® for management of Pseudomonas aeruginosa lung infections, particularly in cystic fibrosis. Promixin® is supplied as a powder for nebulization that is reconstituted prior to use.


The I-neb AAD® is a handheld AAD® system that delivers precise and reproducible drug doses into patients' breathing patterns without the need for a separate compressor (“I-Neb”). The I-neb AAD® is a miniaturized AAD® inhaler based upon a combination of electronic mesh-based aerosolisation technology (Omron) and AAD® technology to control dosing into patients' breathing patterns. The system is approximately the size of a mobile telephone and weighs less than 8 ounces. I-neb AAD® has been used for delivery of Ventavis® (iloprost) (CoTherix/Schering AG).


Another example of a nebulizer which may be used with compositions of the invention is Aria™ (Chrysalis). Aria is based on a capillary aerosol generation system. The aerosol is formed by pumping the drug formulation through a small, electrically heated capillary. Upon exiting the capillary, the formulation rapidly cooled by ambient air to produce an aerosol with MMAD ranging from 0.5-2.0 um.


In addition the TouchSpray™ nebulizer (Odem) may be used to deliver a composition of the invention. The TouchSpray™ nebulizer is a hand-held device which uses a perforate membrane, which vibrates at ultrasonic frequencies, in contact with the reservoir fluid, to generate the aerosol cloud. The vibration action draws jets of fluid though the holes in the membrane, breaking the jets into drug cloud. The size of the droplets is controlled by the shape/size of the holes as well as the surface chemistry and composition of the drug solution. This device has been reported to deliver 83% of the metered dose to the deep lung. Details of the TouchSpray™ nebulizer are described in U.S. Pat. No. 6,659,364, incorporated by reference herein.


Additional nebulizers which may be used with compositions of the invention include nebulizers which are portable units which maximize aerosol output when the patient inhales and minimize aerosol output when the patient exhales using two one-way valves (see PARI nebulizers (PARI GmbH). Baffles allow particles of optimum size to leave the nebulizer. The result is a high percentage of particles in the respirable range that leads to improved drug delivery to the lungs. Such nebulizers may be designed for specific patient populations, such a patients less than three years of age (PARI BABY™) and nebulizers for older patients (PARI LC PLUS® and PARI LC STAR®).


An additional nebulizer which may be used with compositions of the invention is the e-Flow® nebulizer (PARI GmbH) which uses vibrating membrane technology to aerosolize the drug solution, as well as the suspensions or colloidal dispersions (TouchSpray™; ODEM (United Kingdom)). An e-Flow® nebulizer is capable of handling fluid volumes from 0.5 ml to 5 ml, and can produce aerosols with a very high density of active drug, a precisely defined droplet size, and a high proportion of respirable droplets delivered in the shortest possible amount of time. Drugs which have been delivered using the e-Flow® nebulizer include aztreonam and lidocaine. Additional details regarding the e-Flow® nebulizer are described in U.S. Pat. No. 6,962,151, incorporated by reference herein.


Additional nebulizers which may be used with compositions of the invention include a Microair® electronic nebulizer (Omron) and a Mystic™ nebulizer (Ventaira). The Microair® nebulizer is extremely small and uses Vibrating Mesh Technology to efficiently deliver solution medications. The Microair device has 7 mL capacity and produces drug particle MMAD size around 5 microns. For additional details regarding the Microair® nebulizer see US patent publication no. 2004045547, incorporated by reference herein. The Mystic™ nebulizer uses strong electric field to break liquid into a spray of nearly monodispersed, charged particles. The Mystic™ system includes a containment unit, a dose metering system, aerosol generation nozzles, and voltage converters which together offer multi-dose or unit-dose delivery options. The Mystic™ device is breath activated, and has been used with Corus 1030™ (lidocaine HCl), Resmycin® (doxorubicin hydrochloride), Acuair (fluticasone propionate), NCE with ViroPharm, and NCE with Pfizer. Additional details regarding the Mystic™ nebulizer may be found in U.S. Pat. No. 6,397,838, incorporated by reference herein.


Additional methods for pulmonary delivery of the formulation of the invention are provided in U.S. application Ser. No. 12/217,972, incorporated by reference herein.


The appropriate dosage (“therapeutically effective amount”) of the protein will depend, for example, on the condition to be treated, the severity and course of the condition, whether the protein is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the protein, the type of protein used, and the discretion of the attending physician. The protein is suitably administered to the patient at one time or over a series of treatments and may be administered to the patient at any time from diagnosis onwards. The protein may be administered as the sole treatment or in conjunction with other drugs or therapies useful in treating the condition in question.


The formulations of the invention overcome the common problem of protein aggregation often associated with high concentrations of protein, and, therefore, provide a new means by which high levels of a therapeutic protein may be administered to a patient. The high concentration formulation of the invention provides an advantage in dosing where a higher dose may be administered to a subject using a volume which is equal to or less than the formulation for standard treatment. Standard treatment for a therapeutic protein is described on the label provided by the manufacturer of the protein. For example, in accordance with the label provided by the manufacturer, infliximab is administered for the treatment of rheumatoid arthritis by reconstituting lyophilized protein to a concentration of 10 mg/mL. The formulation of the invention may comprise a high concentration of infliximab, where a high concentration would include a concentration higher than the standard 10 mg/mL. In another example, in accordance with the label provided by the manufacturer, Xolair (omalizumab) is administered for the treatment of asthma by reconstituting lyophilized protein to a concentration of 125 mg/mL. In this instance, the high concentration formulation of the invention would include a concentration of the antibody omalizumab which is greater than the standard 125 mg/mL.


Thus, in one embodiment, the formulation of the invention comprises a high concentration which is at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 100%, at least about 110%, at least about 120%, at least about 130%, at least about 140%, at least about 150%, at least about 175%, at least about 200%, at least about 225%, at least about 250%, at least about 275%, at least about 300%, at least about 325%, at least about 350%, at least about 375%, at least about 400%, and so forth, greater than the concentration of a therapeutic protein in a known, standard formulation.


In another embodiment, the formulation of the invention comprises a high concentration which is at least about 2 times greater than, at least about 3 times greater than, at least about 4 times greater than, at least about 5 times greater than, at least about 6 times greater than, at least about 7 times greater than, at least about 8 times greater than, at least about 9 times greater than, at least about 10 times greater than and so forth, the concentration of a therapeutic protein in a known, standard formulation.


Characteristics of the aqueous formulation may be improved for therapeutic use. For example, the viscosity of an antibody formulation may be improved by subjecting an antibody protein solution to diafiltration using water without excipients as the diafiltration medium. As described above in Section II, excipients, such as those which improve viscosity, may be added back to the aqueous formulation such that the final concentration of excipient is known and the specific characteristic of the formulation is improved for the specified use. For example, one of skill in the art will recognize that the desired viscosity of a pharmaceutical formulation is dependent on the mode by which the formulation is being delivered, e.g., injected, inhaled, dermal absorption, and so forth. Often the desired viscosity balances the comfort of the subject in receiving the formulation and the dose of the protein in the formulation needed to have a therapeutic effect. For example, generally acceptable levels of viscosity for formulations being injected are viscosity levels of less than about 100 mPas, preferentially less than 75 mPas, even more preferentially less than 50 mPas. As such, viscosity of the aqueous formulation may be acceptable for therapeutic use, or may require addition of an excipient(s) to improve the desired characteristic.


In one embodiment, the invention provides an aqueous formulation comprising water and a human TNFα antibody, or antigen-binding portion thereof, wherein the formulation is excipient-free, wherein the formulation has viscosity which makes it advantageous for use as a therapeutic, e.g., low viscosity of less than 40 cP at 8° C., and less than 25 cP at 25° C. when the protein concentration is about 175 mg/mL. In one embodiment, the concentration of the antibody, or antigen-binding portion thereof, in a formulation having improved viscosity is at least about 50 mg/mL. In one embodiment, the formulation of the invention has a viscosity ranging between about 1 and about 2 mPas.


Non-Therapeutic Uses


The aqueous formulation of the invention may also be used for non-therapeutic uses, i.e., in vitro purposes.


Aqueous formulations described herein may be used for diagnostic or experimental methods in medicine and biotechnology, including, but not limited to, use in genomics, proteomics, bioinformatics, cell culture, plant biology, and cell biology. For example, aqueous formulations described herein may be used to provide a protein needed as a molecular probe in a labeling and detecting methods. An additional use for the formulations described herein is to provide supplements for cell culture reagents, including cell growth and protein production for manufacturing purposes.


Aqueous formulations described herein could be used in protocols with reduced concern regarding how an excipient in the formulation may react with the experimental environment, e.g., interfere with another reagent being used in the protocol. In another example, aqueous formulations containing high concentrations of proteins may be used as a reagent for laboratory use. Such highly concentrated forms of a protein would expand the current limits of laboratory experiments.


Another alternative use for the formulation of the invention is to provide additives to food products. Because the aqueous formulation of the invention consists essentially of water and protein, the formulation may be used to deliver high concentrations of a desired protein, such as a nutritional supplement, to a food item. The aqueous formulation of the invention provides a high concentration of the protein in water, without the concern for excipients needed for stability/solubility which may not be suitable for human consumption. For example, whey- and soy-derived proteins are lending versatility to foods as these proteins have an ability to mimic fat's mouthfeel and texture. As such, whey- and soy-derived proteins may be added to foods to decrease the overall fat content, without sacrificing satisfaction. Thus, an aqueous formulation comprising suitable amounts of whey- and soy-derived proteins may be formulated and used to supplement food products.


Articles of Manufacture


In another embodiment of the invention, an article of manufacture is provided which contains the aqueous formulation of the present invention and provides instructions for its use. The article of manufacture comprises a container. Suitable containers include, for example, bottles, vials (e.g., dual chamber vials), syringes (such as dual chamber syringes), autoinjector pen containing a syringe, and test tubes. The container may be formed from a variety of materials such as glass, plastic or polycarbonate. The container holds the aqueous formulation and the label on, or associated with, the container may indicate directions for use. For example, the label may indicate that the formulation is useful or intended for subcutaneous administration. The container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g., from 2-6 administrations) of the aqueous formulation. The article of manufacture may further comprise a second container. The article of manufacture may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.


The contents of all references, patents and published patent applications cited throughout this application are incorporated herein by reference


This invention is further illustrated by the following examples which should not be construed as limiting.


EXAMPLES

The following examples describe experiments relating to an aqueous formulation comprising water as the solution medium. It should be noted that in some instances, decimal places are indicated using European decimal notation. For example, in Table 31 the number “0,296” is synonymous with “0.296”.


Example 1: Diafiltration/Ultrafiltration with Adalimumab and J695

Materials and Methods


Adalimumab and J695 were diafiltered using pure water. After an at least 5-fold volume exchange with pure water, the protein solutions were ultrafiltered to a final target concentration of at least 150 mg/mL. Osmolality, visual inspection and protein concentration measurements (0D280) were performed to monitor the status of the proteins during DF/UF processing.


Size exclusion chromatography and ion exchange chromatography were used to characterize protein stability in each final DF/UF product as compared to the starting formulation, e.g., drug substance (DS) starting material and protein standard. Drug substance or “DS” represents the active pharmaceutical ingredient and generally refers to a therapeutic protein in a common bulk solution.

    • Adalimumab Drug Substance, (Adalimumab extinction coefficient 280 nm: 1.39 mL/mg cm). Drug Substance did not contain polysorbate 80. DS composition: 5.57 mM sodium phosphate monobasic, 8.69 mM sodium phosphate dibasic, 106.69 mM sodium chloride, 1.07 mM sodium citrate, 6.45 mM citric acid, 66.68 mM mannitol.
    • Adalimumab solution used for dynamic light scattering (DLS) measurements: Adalimumab solution that was diafiltered using pure water as exchange medium was adjusted to 1 mg/mL concentration by diluting the Adalimumab solution with Milli-Q water and excipient stock solutions (excipients dissolved in Milli-Q water), respectively.
    • J695 Drug Substance, (J695 extinction coefficient 280 nm: 1.42 mL/mg cm). DS composition: Histidine, Methionine, Mannitol, pH 5.8, and polysorbate 80.
    • Millipore Labscale™ Tangential Flow Filtration (TFF) system, equipped with a 500 mL reservoir. The Labscale TFF system was operated in discontinuous mode at ambient temperature according to Millipore Operating Instructions. Stirrer speed was set to approx. 1.5, and the pump speed was set to approximately 3. The target inlet and outlet pressures were 15 mm psig approximately 50 mm psig, respectively.
    • Minimate™ Tangential Flow Filtration capsule, equipped with an Omega™ PES membrane, 30 kDa cut-off. The capsule was rinsed for 30 min with 0.1 N NaOH and for another 30 min with Milli-Q water.
    • 780 pH meter, Metrohm, equipped with pH probe Pt1000, No. 6.0258.010, calibrated with buffer calibration solutions VWR, pH 4.00 buffer solution red, Cat. No. 34170-127, and pH 7.00 buffer solution yellow, Cat. No. 34170-130.
    • Varian 50 Bio UV visible spectrophotometer, AI 9655, with a fixed Cary 50 cell was used for protein concentration measurements (280 nm wavelength). A 100 μL protein sample was diluted with water (Milli-Q water for HPLC) to a final volume of 50.00 mL for protein concentration measurements of all J695 samples and the Adalimumab solution after DF/UF. Concentration of all other Adalimumab samples was monitored by diluting 40 μL sample solution with 1960 μL Milli-Q water. Disposable UV cuvettes, 1.5 mL, semi-micro, Poly(methyl methacrylate) (PMMA), were used for concentration measurements, Milli-Q water was used as OD 280 blank.
    • Milli-Q water for HPLC grade was used as DF/UF medium.
    • A Malvern Zetasizer Nano ZS, Instrument No. AI 9494 was used for DLS measurements.
    • Hellma precision cells, suprasil, Type No. 105.251-QS, light path 3 mm, center 8.5, were used for DLS measurements (filled with 75 μL sample, Malvern Mastersizer Nano ZS, Item No. AI 9494).
    • Knauer Osmometer Automatic, Instr. No. 83963, Berlin, Germany, was used for osmolality measurement (calibrated with 400 mOsmol/kg NaCl calibration solution, Art. No. Y1241, Herbert Knauer GmbH, Berlin, Germany).
    • 250 mL Corning cell culture flasks, 75 cm2, polystyrene, sterile, Corning, N.Y., USA, were used for storage of the protein solutions after the DF/UF operation.
    • Sodium chloride: J.T. Baker was used for preparing a 2M NaCl stock solution. The stock solution was used to prepare 1 mg/mL Adalimumab solution in pure water with various concentrations of NaCl (10, 20, 30, and 50 mM)
    • D-sorbitol, Sigma Chemical Co., St. Louis, Mo. 63178 was used for preparing a 200 mg/mL sorbitol stock solution. The stock solution was used to prepare 1 mg/mL Adalimumab solution in pure water with various concentrations of sorbitol (10, 20, 30, and 40 mg/mL).


      HPLC Methods
    • Adalimumab, SEC analysis: Sephadex 200 column (Pharmacia Cat. No. 175175-01, S/N 0504057). Mobile phase 20 mM sodium phosphate, 150 mM sodium chloride, pH 7.5, 0.5 mL/min flow rate, ambient temperature, detection UV 214 nm and 280 nm. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 50 μg (duplicate injection).
    • Adalimumab, IEC analysis: Dionex, Propac WCX-10 column (p/n 054993) along with a corresponding guard column (p/n 054994). Separation conditions: mobile phase A: 10 mM sodium phosphate, pH 7.5; mobile phase B 10 mM Sodium phosphate, 500 mM Sodium chloride, pH 5.5. 1.0 mL/min flow rate, ambient temperature. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 100 μg, duplicate injection.
    • J695, SEC analysis: Tosoh Bioscience G3000swxl, 7.8 mm×30 cm, 5 (Cat. No. 08541). Mobile phase 211 mM Na2SO4/92 mM Na2HPO4, pH 7.0. 0.3 mL/min flow rate, ambient temperature, detection UV 214 nm and 280 nm. Each sample was diluted to 2.5 mg/mL with Milli-Q water, sample injection load 50 μg (duplicate injection).
    • J695, IEC analysis: Dionex, Propac WCX-10 column (p/n 054993) along with a corresponding guard column (p/n 054994). Separation conditions: mobile phase A: 10 mM Na2HPO4, pH 6.0; mobile phase B 10 mM Na2HPO4, 500 mM NaCl, pH 6.0. 1.0 mL/min flow rate, 35° C. temperature. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 100 μg. J695 Reference standard 29001BF was run in triplicate as a comparison and was diluted to 1 mg/ml in Milli-Q water based on the concentration from the Certificate of Analysis.


      Calculation of the Protein Concentration


      Calculation Formula:






E
=



-
l



g


(

I

I
0


)



=



ɛ
·
c
·
d


c

=

E

ɛ
×
d









ε—absorption coefficient


c—concentration


d—length of cuvette that the light has to pass


E—absorbance


I0—initial light intensity


I—light intensity after passing through sample







ɛ
Adalimumab

=

1.39


mL

mg
×
cm










ɛ

J





695


=

1.42


mL

mg
×
cm










ɛ
HSA

=

1.042


mL

mg
×
cm








1.1: DF/UF Processing of Adalimumab


DF/UF experiments are carried out following the standard operating procedures of the DF/UF equipment manufacturers. For example, the Millipore Labscale™ TFF system was equipped with a 500 mL reservoir and the system operated in discontinuous mode at ambient temperature, in accord with Millipore operating instructions. Stirrer speed was set to approximately 1.5, and the pump speed was set to approximately 3. The target inlet and outlet pressures were 15 mm psig and approximately 50 mm psig, respectively, and the target pressures were monitored to ensure that they were not exceeded.


A Minimate™ Tangential Flow Filtration capsule equipped with an Omega™ PES membrane (Pall Corp., Port Washington, N.Y.), 30 kDa MWCO, was used. The capsule was rinsed for 30 min with 0.1 N NaOH and for another 30 min with Milli-Q water before use.


Approximately 500 mL of Adalimumab solution were placed into the TFF reservoir and DF/UF processing was started in discontinuous mode. Table 1 provides details on the In-Process-Control (IPC) data characterizing the DF/UF process.









TABLE 1







Overview on Adalimumab DF/UF Processing













Volume of
Approx.






Milli-Q
volume of
Adalimumab

Adalimumab



water
Adalimumab
concentration

concentration


Process
added
solution in
of retentate
Osmolality
of permeate


Step
(mL)
retentate (mL)
(mg/mL)
(mOsmol/kg)
(mg/mL)





 1

500
54.66
305



 2

400
68.33
297
3.15


 3

300





 4
250
550
43.73
169
1.39


 5

300


4.45


 6
250
550
47.27
 93
2.58


 7

250





 8
250
500





 9

250





10
250
500





11

250





12
250
500
52.24
 9
1.24


13

300
90.27
   7.5



14

130
213.87 

4.08





Fields filled with “—” indicate that no IPC samples were pulled at that step.






The DF/UF processing was stopped after an approximate 5-fold volume exchange (1 volume exchange accounting for approx. 250 mL diafiltration medium). Assuming an ideal 100% excipient membrane permeability, the theoretical final excipient concentration reached by the experiment parameters applied is Ci(250/500)5=0.03125*Ci, with Ci being the initial concentration. The maximum excipient reduction was therefore 96.875% (if constant volume diafiltration would have been used, the theoretical excipient reduction with 5 diafiltration volumes would have been Ci e−5=0.00674, i.e. an approximate 99.3% maximum excipient reduction). Adalimumab solution was drained from the TFF system to a 250 mL cell culture flask (low-volume rinse of the TFF system was performed using WFI yielding a 175.05 mg/mL concentration; without the rinse, the retentate concentration was 213.87 mg/mL). Samples were pulled for determination of pH, osmolality and Adalimumab concentration. Additionally, samples were pulled for characterization by SEC and IEC. Characteristic parameters of the Adalimumab solution before and after DF/UF processing, respectively, are listed in Table 2.









TABLE 2







Impact of DF/UF processing on Adalimumab solution












solution
solution




before
after



parameter
DF/UF
DF/UF















pH
5.19
5.22



concentration (mg/mL)
54.66
175.05



osmolality (mOsmol/kg)
305
24



*SEC data (% aggregate,
0.26
00.50



monomer,
99.74
99.50



fragment)
0.00
0.00



*IEC data (acidic regions, lys
13.89
14.07



0,
62.05
61.97



lys 1,
19.14
18.51



lys 2, %)
4.83
4.73







*samples were subjected to one freeze/thaw step (−80° C./25° C.) before analysis via SEC and IEC






In the course of DF/UF processing, Adalimumab concentration exceeded 210 mg/mL. Throughout the experiment, the protein solution remained clear, and no solution haziness or protein precipitation, which would have indicated Adalimumab solubility limitations, was observed. Compared to the original Adalimumab DS solution (˜55 mg/mL), Adalimumab solution diafiltered by using pure water as DF/UF exchange medium revealed lower opalescence, despite a more than 3-fold increase in protein concentration (˜175 mg/mL).


1.2: Adalimumab Characterization Via Chromatography



FIG. 1 shows a SEC chromatogram of an Adalimumab reference standard (Adalimumab standard (bottom line)) compared to the Adalimumab drug standard solution before (middle line) and after (top line) the DF/UF processing procedure. Note that all samples were frozen at −80° C. prior to analysis.


Table 3 also contains the IEC chromatogram data (note all samples were frozen at −80° C. prior to analysis).









TABLE 3







IEC Data of Various Adalimumab Samples













% Acidic
% Acidic
% 0
% 1
% 2


Sample Name
Region 1
Region 2
Lys
Lys
Lys





Reference standard
2.69
11.66
60.77
19.42
5.40


Adalimumab DS
2.51
11.38
62.05
19.14
4.83


Adalimumab, after
2.26
11.81
61.97
18.51
4.73


DF/UF










1.3: Impact of Excipients on Adalimumab Hydrodynamic Diameter (Dh)


It was previously determined that the hydrodynamic diameter of J695, as determined by dynamic light scattering (DLS) measurements, was notably decreased when formulating J695 into pure water. J695 in WFI had a Dh of ˜3 nm, far below the values that are expected for immunoglobulins. Upon addition of low amounts of ionizable NaCl, the Dh values increased to ˜10 nm (independent of the NaCl concentration). Addition of non-ionizable mannitol increased J695 solution osmolality, but had no effect on J695 Dh.


In order to assess the impact of excipients on the hydrodynamic diameter of Adalimumab that had been processed according to the above DF/UF procedure, the Adalimumab solution from the DF/UF experiment was used to formulate Adalimumab solutions in pure water, but with varying levels of NaCl (0-50 mM) and sorbitol (0-40 mg/mL), respectively. The impact of sorbitol (a non-ionizable excipient) and NaCl (ionizable excipient) concentrations on Dh of Adalimumab monomer is displayed in FIG. 2.


The hydrodynamic diameter of Adalimumab monomer in pure water was 2.65 nm. The Adalimumab Dh response to salt and non-ionic excipients was identical to the J695 response seen previously. Adalimumab Dh was virtually not impacted by the presence of sorbitol. Low concentrations of NaCl induced the monomer hydrodynamic diameter to increase to expected levels of ˜11 nm. These findings demonstrate that protein hydrodynamic diameters as measured by dynamic light scattering are crucially impacted by the presence of ionizable excipients. Absence of ionizable excipients also is linked to solution viscosities.


These findings have implications for high-concentrated protein solutions: the lower the hydrodynamic diameter, the lower the spatial volume proteins occupy. In high-concentration scenarios, the viscosities of protein solutions that are prepared by using water as DF/UF exchange medium will be substantially lower than the viscosities of traditional protein formulations containing considerable amounts of ionizable buffer excipients. The Adalimumab data confirmed this, as viscosities of 200 mg/mL Adalimumab solutions in water for injection were found to be well below 50 mPas, independent of pH (e.g. pH 4, pH 5, and pH 6). More data on the effect of pH on Dh can be found in Example 17 below.


Overall, these findings are useful in high-concentration protein formulation activities, where viscosity related manufacturing and dosing/delivery problems are well known. Furthermore, these findings show that the osmolality values of final Drug Product can be adjusted with non-ionizable excipients such as sugars or sugar alcohols as desired without inducing an increase in protein Dh and solution viscosity, respectively.


1.4: DF/UF Processing of J695 (Anti-IL12 Antibody)


Approximately 200 mL of J695 solution were adjusted to pH 4.4 with 1 M phosphoric acid and filled into the TFF reservoir (pH adjustment was made to ensure a positive zeta-potential of J695 monomers and thus avoid a potential impact of uncharged protein monomer on data). Then, 300 mL of Milli-Q water were added to the TFF reservoir, and DF/UF processing was started in discontinuous mode. 250 mL reservoir volume, 250 mL of Milli-Q water were added, and DF/UF processing was started again. The DF/UF processing was stopped after a total of 5 volume exchange steps were performed (1 volume exchange accounting for approx. 250 mL).


Assuming an ideal 100% excipient membrane permeability, the theoretical final excipient concentration reached by the experiment parameters applied is Ci(250/500)5=0.03125*Ci, with Ci being the initial concentration. The maximum excipient reduction is therefore 96.875% (if constant volume diafiltration would have been used, the theoretical excipient reduction with 5 diafiltration volumes would have been Ci e−5=0.00674, i.e. an approx. 99.3% maximum excipient reduction). J695 solution was drained from the TFF system to a 250 mL cell culture flask (no rinse of the TFF system was performed). Samples were pulled for determination of pH, osmolality and J695 concentration. Additionally, samples were pulled for characterization by SEC and IEC. Characteristic parameters of the J695 solution before and after DF/UF processing, respectively, are listed in Table 4.









TABLE 4







Impact of DF/UF Processing on J695 Solution












solution
solution




before
after



parameter
DF/UF
DF/UF















pH
4.40
4.70



concentration (mg/mL)
122.9
192.8



osmolality (mOsmol/kg)
265
40



SEC data (% aggregate,
0.41
0.69



monomer,
98.42
98.11



fragment)
1.18
1.21



IEC data
92.00
92.11



(sum of isoforms,
5.17
5.30



acidic species,
2.83
2.59



basic species, %)










As with Adalimumab, the DF/UF experiments on J695 substantiate the principal possibility of processing and formulating J695 by using pure water as exchange medium in DF/UF operations. Both SEC and IEC data suggest no substantial impact on J695 stability while DF/UF processing for an overall period of 1.5 days (process interruption overnight) at ambient temperature using Milli-Q water as diafiltration medium. Throughout the experiment, the protein solution remained clear, indicating no potential J695 solubility limitations.


1.5: J695 Characterization


Table 5 describes the percentages for aggregate, monomer and fragment content for the three solutions as determined by SEC chromatogram.









TABLE 5







Data from SEC chromatogram













%
%
%




Aggregate
Monomer
Fragment



Sample Name
content
content
content







Reference standard
0.45
98.00
1.56



J695 before DF/UF
0.41
98.42
1.18



J695 after DF/UF
0.69
98.11
1.21











FIG. 3 shows the IEC profile of J695 reference standard (bottom graph) and J695 DS, pH adjusted to pH 4.4 (top graph).


Only a small increase in aggregate content was observed in the J695 samples after DF/UF processing.



FIG. 4 shows the IEC profile of J695 after DF/UF with Milli-Q water, pH 4.7 (top graph), and J695 DS before DF/UF, pH adjusted to pH 4.4 (bottom curve).


As depicted in FIG. 4, the DF/UF step had no notable impact on J695 stability when monitored by IEC. The differences between the two J695 reference standards (refer to FIG. 3) can be attributed to differences in the manufacturing processes between the 3000 L and 6000 L DS campaigns. Table 6 highlights more details on IEC data.









TABLE 6







IEC Data of Various J695 Samples











Sample Name
0 glu (1)
other isoforms
acidic
basic





Reference standard
43.57
50.06
4.47
1.90


J695
35.74
56.26
5.17
2.83


J695 after DF/UF
36.59
55.52
5.30
2.59










1.6: Conclusion


The above example provides a diafiltration/ultrafiltration (DF/UF) experiment where water (Milli-Q water for HPLC) was used as diafiltration medium for monoclonal antibodies Adalimumab and J695.


Adalimumab was subjected to DF/UF processing by using pure water as DF/UF exchange medium and was formulated at pH 5.2 at high concentrations (>200 mg/mL) without inducing solution haziness, severe opalescence or turbidity formation. Upon one subsequent freeze/thaw step, SEC and IEC data suggested no notable difference between Adalimumab solution formulated in water via DF/UF processing and the original Adalimumab DS.


J695 also was also subjected to DF/UF processing by using pure water as DF/UF exchange medium and was formulated at pH 4.7 without impacting J695 stability (visual inspection, SEC, IEC).


When formulated using such a DF/UF processing, the hydrodynamic diameter (Dh) of Adalimumab monomer was approx. 2.65 nm. The presence of non-ionic excipients such as sorbitol in concentrations up to 40 mg/mL was shown to have no impact on Dh data, whereas ionic excipients such as NaCl already in low concentrations were demonstrated to induce the Adalimumab monomer Dh to increase to approx. 11 nm (such Dh data are commonly monitored for IgG). Similar findings were made earlier for J695.


In conclusion, processing and formulating proteins using pure water as DF/UF exchange medium is feasible. Assuming an ideal 100% excipient membrane permeability, the theoretical final excipient concentration reached by the constant volume diafiltration with 5 diafiltration volumes would be Ci e−5=0.00674, i.e. an approx. 99.3% maximum excipient reduction. Using 6 diafiltration volume exchanges, an theoretical 99.98% maximum excipient reduction would result.


Examples 2 to 5 describe experimental execution with respect to three different proteins which were concentrated into an aqueous formulation, and Examples 6 to 11 describe analysis of each of the aqueous formulations.


Materials and Methods for Examples 2-11





    • Adalimumab protein solution (10 mg/mL) in water for injection, Protein Drug Substance (DS) Material (49.68 mg/mL), DS contains Tween 80, Adalimumab, Adalimumab Drug Product (DP) (40 mg, solution for injection, filtered solution from commercial production). Protein absorption coefficient 280 nm: 1.39.

    • J695 protein solution (10 mg/mL) in water for injection, Protein Drug Substance (DS) (54 mg/mL), DS contains Tween 80. Absorption coefficient 280 nm: 1.42

    • HSA protein solution (10 mg/mL) in water for injection, DP without Tween 80, Grifols Human Serum Albumin Grifols®, 20%, 50 mL. Absorption coefficient 280 nm: 1.042

    • 6 R vial and 10R vial

    • vial preparation: the vials were washed and autoclaved

    • stoppers, 19 mm, West, 4110/40/grey

    • sample repositories (e.g., Eppendorf sample repository, Safe-Lock or simple snap-fit, 1-2 mL)

    • single-use syringes, sterile, 20 mL; NormJect, 10 mL

    • single use filter units (filter Millex®-GV, Syringe Driven Filter Unit, PVDF 0.22 μm; Millex®-GP, Syringe Driven Filter Unit, PES 0.22 μm, Sterivex®0.22 μm Filter Unit)

    • Vivaspin concentrators (cut off 10 kDa, PES; cut off 3 kDa, PES)

    • Pipettes (e.g., Eppendorf, max.: 1000 μL)

    • Water for injection

    • Centrifuge (Eppendorf) and Centrifuge No. 1 (temperature-controlled)

    • Diafiltration equipment: Millipore Labscale™ TFF System, Millipore diafiltration membrane: Adalimumab: Polyethersulfone; J695: Polyethersulfone; HSA: regenerated cellulose

    • pH probe (Metrohm pH-Meter, protein-suitable probe, biotrode 46)

    • Laminar-Air-Flow bench, Steag LF-Werkbank, Mp.-No. 12.5

    • NaCl; mannitol

    • Gemini 150 Peltier Plate Rheometer, Malvern

    • Rheometer MCR 301 [temperature-controlled P-PTD 200 (plate with Peltiertempering)] and cone/plate measurement system CP50/0.5° Ti as well as CP50/1° (stainless steel); Anton Paar

    • Capillary viscometer, Schott, capillaries: type 537 20, type 537 10, type 537 13

    • 1M hydrochloric acid (J. T. Baker)


      Analytics

    • UV/VIS spectrophotometry (OD 280 nm); Photon Correlation Spectroscopy (PCS): for approximately 10 mg/mL and approximately 20 mg/mL: 1.1 mPas, 3 runs, 30 s, one measurement, 25° C., from approximately 30 mg/mL and above: 1.9 mPas, 30 s, 3 runs, one measurement, 25° C.

    • Size Exclusion Chromatography (SEC) and Ion Exchange Chromatography (IEC), as described below.

    • viscosity measurement: different viscometers with individual and different set-ups were used


      Calculation of the Protein Concentration


      Calculation Formula:









E
=



-
1







g


(

I

I
0


)



=



ɛ
·
c
·
d


c

=

E

ɛ
×
d









ε—absorption coefficient


c—concentration


d—length of cuvette that the light has to pass


E—absorbance


I0—initial light intensity


I—light intensity after passing through sample







ɛ
Adalimumab

=

1.39


mL

mg
×
cm










ɛ

J





695


=

1.42


mL

mg
×
cm










ɛ
HSA

=

1.042


mL

mg
×
cm








Viscosity Data and Calculation for Adalimumab


Adalimumab commercial formulation (approximately 194 mg/mL) density:






ρ
=

1
.
05475


g

cm
3








Adalimumab commercial formulation (approximately 194 mg/mL) kinematic viscosity:


K—constant of the capillary


t—the time the solution needs for passing the capillary [s]


v—kinematic viscosity






v
=


K
×
t

=


0
.
03159



mm
2


s
2


×
279
.
36





s

=

8.825



mm
2

s









Adalimumab commercial formulation (approximately 194 mg/mL) dynamic viscosity:


η—dynamic viscosity


ρ—density






η
=


v
×
ρ

=


8
.
825








mm
2


s

×
1
.
05475


g

cm
3



=

9
.
308





mPas








Viscosity Data and Calculation for Human Serum Albumin


HSA commercial formulation (approximately 200 mg/mL) density:






ρ
=

1
.
05833


g

cm
3








HSA commercial formulation (app. 200 mg/mL) kinematic viscosity:


K—constant of the capillary


t—the time the solution needs for passing the capillary [s]


v—kinematic viscosity






v
=


K
×
t

=


0
.
01024



mm
2


s
2


×
337
.
69





s

=

3
.
46



mm
2

s









HSA commercial formulation (approximately 200 mg/mL) dynamic viscosity:


η—dynamic viscosity


ρ—density






η
=


v
×
ρ

=


3
.
46



mm
2

s

×
1
.
05833


g

cm
3



=

3
.
662





mPas








HSA in WFI (approximately 180 mg/mL) density:






ρ
=

1
.
07905


g

cm
3








HSA in WFI (approximately 180 mg/mL) kinematic viscosity:


K—constant of the capillary


t—the time the solution needs for passing the capillary [s]


v—kinematic viscosity






v
=


K
×
t

=


0
.
09573



mm
2


s
2


×
185
.
3





s

=

17
.
72



mm
2

s









HSA in WFI (approximately 180 mg/mL) dynamic viscosity:


η—dynamic viscosity


ρ—density






η
=


v
×
ρ

=


17
.
72



mm
2

s

×
1
.
07905


g

cm
3



=

19
.
121





mPas








General Experimental Execution for Arriving at High Concentration Formulation


Generally, the process of the invention for arriving at a high concentration, salt-free, protein formulation includes diafiltration of the initial drug substance material, followed by a procedure to increase the concentration of the drug substance in the solution. This may be done in separate procedures or may be done in separate or coinciding steps within the same procedure.


Diafiltration


A sufficient amount of Drug Substance (DS) material (depending on protein concentration of DS) was subjected to diafiltration. Prior to diafiltration, the DS material was diluted with water for injection ˜10 mg/ml. Note that in total approximately 540 mL of a 10 mg/mL solution was needed for the experiment.


Water for injection was used as diafiltration medium. The number of diafiltration steps performed was 5 to 7 (one diafiltration step equals one total volume exchange). In-Process-Control (IPC) samples were pulled prior to diafiltration and after diafiltration step (200 μL for osmolality, 120 μL for SEC).


Diafiltration with TFF equipment is performed by applying the following parameters:

    • stirrer: position 2
    • pump: position 2
    • pressure up-stream/inlet: max 20-30 psi
    • pressure down-stream/outlet: max 10 psi


(Parameters used in this experiment were derived from manufacturer's recommendations. One with skill in the art would be able to alter the parameters of equipment operation to accommodate a particular protein or variances in equipment, formulation, viscosity, and other variables.)


After diafiltration, protein concentration was assessed by means of OD280. If protein concentration was >10 mg/mL, the protein concentration was adjusted to 10 mg/mL by appropriately diluting the solution with water for injection.


Concentration


20 mL of diafiltrated protein solution (e.g., Adalimumab, J695, HSA) were put into a Vivaspin 20 Concentrator. The concentrator was closed and put into the centrifuge. The protein solution was centrifuged at maximum speed (5000 rpm).


Sample Pulls


Samples of the concentrated solutions were pulled at: 10 mg/mL and then every 10 mg/mL (20, 30, 40 mg/ml etc.) or until the protein aggregates visibly, and samples were analyzed as follows:

    • The protein solution was homogenized in the Vivaspin concentrator and filled in an adequate vial.
    • Optical appearance was inspected directly in the vial.
    • 300 μL was used for UV spectroscopy, 160 μL for PCS, 1204, for SEC and 300 μL, for IEX.
    • the samples for SEC and IEX were stored at −80° C.


      Dynamic Light Scattering (DLS) Protocol


Dynamic light scattering was performed using the Zetasizer Nano ZS (Malvern Instruments, Southborough, Mass.) equipped with Hellma precision cells (Plainview, N.Y.), suprasil quartz, type 105.251-QS, light path 3 mm, center Z8.5 mm, with at least 60 sample fill, using protein samples as is and placed directly in measurement cell. Prior to measurement, the cell window was checked to verify that the solution was free of air bubbles or particles/dust/other contaminants that may impact DLS measurement. Measurements were taken under standard operating procedures (“general purpose” mode, 25° C., refractive index set to 1.45, measurement mode set to “manual”, 1 run per measurement, each comprising 3 measurements of 30 s each, type of measurement set to “size”). Dispersion Technology Software, version 4.10b1, Malvern Instruments, was used to analyze data. About 70 μL of a sample solution were filled in precision cell for analysis of hydrodynamic diameters (Dh). Default sample viscosity was set 1.1 mPas for low concentrated protein solutions (e.g. <5 mg/mL). Underlying measurement principles concluded that minimal differences between real viscosity values of the sample solution to be measured and the use of default viscosities does not impact DLS data readout substantially. This was verified by performing DLS measurements of low protein concentration solutions (<5 mg/mL) where solution viscosities were determined and taken into account in subsequent DLS measurements. For all samples with higher protein concentration, viscosities were determined and taken into account during DLS measurements.


Example 2: Formulation Comprising High TNFα Antibody Concentration

2.1: Diafiltration


Prior to diafiltration, Adalimumab (49.68 mg/mL) was diluted with water for injection to a concentration of approximately 15 mg/mL. Therefore 140.8 mL Adalimumab solution (49.68 mg/mL) were filled in a 500 mL volumetric flask. The flask was filled up to the calibration mark with water for injection. The volumetric flask was closed and gently shaken for homogenization of the solution. The TFF labscale system was flushed with water. Then the membrane (PES) was adapted and was also flushed with 1 L distilled water. Next, the TFF labscale system and the membrane were flushed with approximately 300 mL of water for injection. The diluted Adalimumab solution was then filled in the reservoir of the TFF. A sample for an osmolality measurement (300 UV spectrophotometry (500 μL) and a sample for SEC analysis (120 μL) were pulled. The system was closed and diafiltration was started. The DF/UF (diafiltration/ultrafiltration) was finished after 5 volume exchanges and after an osmolality value of 3 mosmol/kg was reached. The pH-value of the Adalimumab solution after diafiltration was pH 5.25.


Diafiltration with TFF equipment was performed by applying the following parameters:

    • stirrer: position 2
    • pump: position 2
    • pressure up-stream/inlet: max 20-30 psi
    • pressure down-stream/outlet: max 10 psi


After diafiltration, protein concentration was assessed by means of OD280. The concentration was determined to be 13.29 mg/mL.


The Adalimumab solution was sterile filtered.


The TFF and the membrane were flushed with approximately 1 L distilled water and then with 500 mL 0.1M NaOH. The membrane was stored in 0.1M NaOH, the TFF was again flushed with approximately 500 mL distilled water.


2.2: Protein Concentration


Prior to concentrating the antibody, the protein concentration was again assessed by means of OD280. Adalimumab concentration was determined to be 13.3 mg/mL. The Adalimumab solution was then diluted to 10 mg/mL. 375.94 mL of Adalimumab solution (13.3 mg/mL) was filled in a 500 mL volumetric flask and the flask was filled up to the calibration mark with water for injection (WFI). 75.19 mL of Adalimumab solution (13.3 mg/mL) was also filled in a 100 mL volumetric flask, and filled up to the calibration mark with pure water, i.e., water for injection (WFI). Both flasks were gently shaken for homogenization. The solutions from both flasks were placed in a 1 L PETG bottle. The bottle was gently shaken for homogenization.


Four Vivaspin 20 concentrators (10 kDa) were used. In three Vivaspins, 20 mL of Adalimumab solution (10 mg/mL) were filled (in each). In the fourth Vivaspin device, water was filled as counterbalance weight while centrifuging. The concentrators were closed and put into the centrifuge. The Adalimumab solution was centrifuged applying 4500×g centrifugation force (in a swing out rotor).


2.3: Sample Pull


Samples of the concentrated Adalimumab solution were pulled when they reached a concentration of 10 mg/mL and at each subsequent 10 mg/mL concentration increment increase (at 20 mg/mL, 30 mg/mL, 40 mg/mL etc. until approximately 200 mg/mL). At 40 minute intervals, the concentrators were taken out of the centrifuge, the solution was homogenized, and the solution and the centrifuge adapters were cooled for approximately 10 min on ice. After each 10 mg/mL concentrating increment, the solutions in the concentrators were homogenized, the optical appearance was checked and samples were pulled for analysis via UV (300 μL), PCS (160 μL), SEC (120 μL) and IEC (300 μL). After sample pulls, the concentrators were filled up to approximately 20 mL with Adalimumab solution (10 mg/mL).


Visual Inspection and PCS analysis of protein precipitation were used to determine the solubility limit of Adalimumab protein (i.e. isoforms) in the solution.


At a concentration of approximately 80 mg/mL it became obvious that the Adalimumab solution was not opalescent anymore, opalescence being a known characteristic of Adalimumab solutions having a high amount of fragment. Therefore, it was suspected that fragmentation might have occurred during experiment execution. For further analysis, a sample of Adalimumab solution (approximately 80 mg/mL) was analyzed by SEC. The remainder of the solution in each Vivaspin, as well as the rest of Adalimumab solution (10 mg/mL), was removed to 50 mL falcon tubes and stored at −80° C. The SEC analysis showed a purity of 99.6% monomer.


The solution was thawed in water bath at 25° C. and sterile filtered. Afterwards the solutions from 3 falcon tubes were place into each Vivaspin. The concentrators were filled to approximately 20 mL and the concentration was continued. The experiment was finished as a concentration of approximately 200 mg/mL was reached.


All SEC and IEC samples were stored at −80° C. until further analysis. UV and PCS were measured directly after sample pull. The rest of the concentrated Adalimumab solution was placed in Eppendorf repositories and stored at −80° C.


Table 7 shown below describes calculation of volumes of protein solution to be refilled into the concentrators while concentrating Adalimumab solution. The scheme was calculated before experiment execution to define at which volume samples have to be pulled. The duration of centrifugation is shown in Table 8.









TABLE 7







Centrifugation Scheme

















volume







10 mg/ml





volume
concentration
protein




step
(ml)
(mg/ml)
solution


















 0
20
10




conc.
 1
10
20




sampling
 2
9
20




filling
 3
20
14.5
11



conc.
 4
9.66
30




sampling
 5
8.66
30




filling
 6
20
18.66
11.34



conc.
 7
9.33
40




sampling
 8
8.33
40




filling
 9
20
22.49
11.67



conc.
10
8.99
50




sampling
11
7.99
50




filling
12
20
25.98
12.01



conc.
13
8.66
60




sampling
14
7.66
60




filling
15
20
29.15
12.34



conc.
16
8.32
70




sampling
17
7.32
70




filling
18
20
31.96
12.68



conc.
19
7.99
80




sampling
20
6.66
80




filling
21
20
34.46
13.01



conc.
22
7.65
90




sampling
23
6.65
90




filling
24
20
36.6
13.35



conc.
25
7.32
100




sampling
26
6.32
100




filling
27
20
38.44
13.68



conc.
28
6.98
110




sampling
29
5.98
110




filling
30
20
39.9
14.02



conc.
31
6.65
120




sampling
32
5.65
120




filling
33
20
41.07
14.35



conc.
34
6.31
130




sampling
35
5.31
130




filling
36
20
41.86
14.69



conc.
37
5.98
140




sampling
38
4.98
140




conc.
39
4.64
150
154.14







174.14

















TABLE 8







Centrifugation times required for


concentrating the Adalimumab solution








concentration



[from -> to]



[mg/ml]
time [min]











10 to 20
15


20 to 30
20


30 to 40
27


40 to 50
30


50 to 60
40


60 to 70
50


70 to 80
60


80 to 90
67


 90 to 100
80


100 to 110
100


110 to 200
206










Results from the concentration of Adalimumab are also shown below in Table 12.


2.4: Viscosity Measurement


Adalimumab solutions comprising either 50 mg/mL in WFI or 200 mg/mL in WFI were measured for viscosity. 50 mg/mL and 200 mg/mL in WFI were measured using a Gemini 150 Peltier Plate Rheometer, Malvern, and the 200 mg/mL in WFI solution was also measured via rheometer MCR 301 [temperature-controlled P-PTD 200 (plate with Peltier tempering)] and cone/plate measurement system CP50/1 (stainless steel); Anton Paar).


Adalimumab solutions (200 mg/mL) in repository tubes were thawed and homogenized in a 6R vial. 1 mL Adalimumab (200 mg/mL) was diluted with 3 mL WFI to obtain the diluted solution (for a 50 mg/ml Adalimumab solution).


For the rheometer Gemini 150 approximately 2 mL were needed and for the MCR 301 less than 1 mL was needed for measurement.


Adalimumab (approximately 194 mg/mL) in the commercial formulation was obtained by using Vivaspin tubes. The tubes were filled with Adalimumab solution in commercial buffer and centrifugation was applied until a 194 mg/mL concentration was reached. Viscosity was measured with the capillary viscometer Schott.


2.5: Summary


In sum, Adalimumab was concentrated from 50 mg/mL to approximately 194 mg/mL in Vivaspin 20 tubes in four different tubes. At the beginning, 20 mL of Adalimumab solution (50 mg/mL) were in every tube (four tubes). At the end of the concentration, 5 mL of Adalimumab solution (approximately 194 mg/mL) were in every tube. The concentration step was performed at 5000 rpm (approximately 4500 g). After every hour, the oblong beakers and the protein solution in the Vivaspin tubes were cooled in crushed ice for approximately 10 to 15 min. The density was measured with density measurement device DMA 4500, Anton Paar. Further analysis of the high Adalimumab concentration formulation is provided in Examples 5 to 11.


Example 3: Formulation Comprising High Concentration 11-12 Antibody

3.1: Diafiltration


Prior to diafiltration, IL-12 antibody J695 (54 mg/mL) was diluted with water for injection to a concentration of approximately 15 mg/mL. This was done by placing 150 mL J695 solution (54 mg/mL) in a 500 mL volumetric flask and filling the flask to the calibration mark with water for injection. The volumetric flask was closed and gently shaken for homogenization of the solution. The TFF labscale system was flushed with water. Then the polyethersulfone membrane (PES) was adapted and was also flushed with 1 L of distilled water. Afterwards the TFF labscale system and the membrane were flushed with approximately 300 mL of water for injection. Next, the diluted J695 solution was placed in the reservoir of the TFF. A sample for osmolality measurement (300 μL), UV spectrophotometry (500 μL) and a sample for SEC analysis (120 μL) were pulled. The system was closed and diafiltration was started. After 200 mL of processing the DF volume, the diafiltration was stopped and another sample for UV measurement was pulled. The DF/UF was stopped after 1800 mL diafiltration volume (approximately factor 3.5 volume exchange), reaching an osmolality value of 4 mosmol/kg. The pH-value of the J695 solution after diafiltration was pH 6.48.


Diafiltration with TFF equipment was performed by applying the following parameters:

    • stirrer: position 2
    • pump: position 2
    • pressure up-stream/inlet: max 20-30 psi
    • pressure down-stream/outlet: max 10 psi


After diafiltration, the protein concentration was assessed by means of OD280. The concentration was determined to be 16.63 mg/mL.


The J695 solution was sterile filtered.


The TFF instrument and the membrane were flushed with approximately 1 L of distilled water and then with 500 mL 0.1M NaOH. The membrane was stored in 0.1M NaOH and the TFF was again flushed with approximately 500 mL of distilled water.


3.2: Concentrating


Prior to concentrating, the J695 solution was diluted to 10 mg/mL: 316 mL of J695 solution (16.63 mg/mL) was placed in a 500 mL volumetric flask and the flask was filled to the calibration mark with water for injection (WFI). Additionally, 64 mL of J695 solution (16.63 mL) was placed in a 100 mL volumetric flask and filled to the calibration mark with WFI. Both flasks were gently shaken for homogenization. The solutions from both flasks were placed in a 1 L PETG bottle. The bottle was gently shaken for homogenization.


Four Vivaspin 20 concentrators (10 kDa cut-off) were used. 20 mL of J695 solution (10 mg/mL) were place in each of three Vivaspins. In the fourth Vivaspin concentrator device, water was filled as counterbalance weight while centrifuging. The concentrators were closed and put into the centrifuge. The J695 solution was centrifuged applying 4500×g centrifugation force (in a swing out rotor).


3.3: Sample Pull


Samples of the concentrated J695 solution were pulled when they reached a concentration of 10 mg/mL and at each subsequent 10 mg/mL concentration increment increase (at 20 mg/mL, 30 mg/mL, 40 mg/mL etc. until 200 mg/mL). After every 40 minutes, the concentrators were taken out of the centrifuge, the solution was homogenized, and the solution and the centrifuge adapters were cooled for approximately 10 min on ice. After every 10 mg/mL concentration increase, the solutions in the concentrators were homogenized, the optical appearance was checked and samples were pulled for UV (300 μL), PCS (160 μL), SEC (120 μL) and IEC analysis (300 μL). After sample pulls, the concentrators were filled up to approximately 20 mL with J695 solution (10 mg/mL).


Visual Inspection and PCS analysis were used to determine the solubility (i.e., to check for potential precipitation) and stability of J695.


At the conclusion of the experiment, a concentration of approximately 200 mg/mL was reached.


All SEC and IEC samples were stored at −80° C. for further analysis (see below). UV spectrophotometry and PCS measurements were taken directly after each sample pull. The rest of the concentrated J695 solution was placed in Eppendorf repositories and stored at −80° C.


Details regarding the centrifugation scheme are provided above in Table 7. The duration of the J695 centrifugation are provided in Table 9.









TABLE 9







Centrifugation Times used to


Concentrate the J695 Solution










concentration




[from -> to]




[mg/mL]
time [min]














10 to 20
13



20 to 30
22



30 to 40
27



40 to 50
38



50 to 60
45



60 to 70
80



70 to 80
90



80 to 90
105



 90 to 100
165



100 to 200
270











3.4: Impact of Excipients on the Hydrodynamic Diameter of J695


In this experiment the impact of sodium chloride and mannitol, separately, on the hydrodynamic diameter of J695 was analyzed. For this purpose, stock solutions of sodium chloride (12 mg/mL) and of mannitol (120 mg/mL) were prepared. 1.2 g NaCl was weighed in a beaker, which was filled with approximately 70 mL of WFI, and 12.002 g of Mannitol was weighed in a beaker which was filled with approximately 70 mL of WFI. The two solutions were stirred for homogenization. Each solution was placed in a volumetric flask, which was filled to the calibration mark with WFI. The flasks were gently shaken for homogenization.


Approximately 8 mL of J695 solution (approximately 200 mg/mL) was thawed at 37° C. The solution was filled in a 10R vial and homogenized. Seven 2R vials were filled with 500 μL J695 solution (approximately 200 mg/mL) each. The filling scheme is described in Table 10 below.









TABLE 10







Filling Scheme for Preparation of J695 Solutions Containing


Different Concentrations of NaCl or Mannitol















vol.
vol.






NaCl
mannitol





volume
stock
stock





ABT-874
solution
solution
vol.



concentration
(200 mg/ml(
(12 mg/ml)
(12 mg/mL)
WFI


excipient
excipient
[μL]
[μL]
[μL]
[μL]







500


500


NaCl
 2 mg/mL
500
167

333


NaCl
 4 mg/mL
500
333

167


NaCl
 6 mg/mL
500
500




mannitol
20 mg/mL
500

167
333


mannitol
40 mg/mL
500

333
167


mannitol
60 mg/mL
500

500










The 2R vials were gently homogenized via shaking. Thereafter, PCS and osmolality measurements were taken of the different J695 solutions (100 mg/mL).


To prepare samples for PCS analysis, the cuvettes were first flushed with 50 μL of the sample. Then measurements were taken using 100 μL of the sample.


Further analysis of the high J695 concentration formulation is provided in Examples 5 to 11.


Example 4: High Concentration Human Serum Albumin (HSA) Formulation

4.1: Diafiltration


Prior to diafiltration, HSA solution (200 mg/mL, commercial formulation) was diluted with water for injection to a concentration of 15.29 mg/mL. To achieve this, 38 mL HSA (200 mg/mL) were filled in a 500 mL volumetric flask. The flask was filled to the calibration mark with water for injection. The volumetric flask was closed and gently shaken for homogenization of the solution. The TFF labscale system was flushed with water. Then the membrane (regenerated cellulose) was adapted and was also flushed with 1 L of distilled water. Afterwards the TFF labscale system and the membrane were flushed with approximately 300 mL water for injection. Next, the diluted HSA solution was filled in the reservoir of the TFF. Samples for osmolality measurement (300 UV spectrophotometry (500 μL) and a sample for SEC analysis (120 μL) were pulled. The system was closed and diafiltration was started. After diafiltration of approximately 300 mL of volume, a UV measurement of the permeate was taken. The permeate revealed a concentration of 2.74 mg/mL, indicating that protein was passing through the membrane. The diafiltration was stopped after approximately 500 mL of DF, and another sample for UV measurement was pulled (HSA concentration 11.03 mg/mL). The DF/UF was finished after 950 mL of diafiltration volume (approximately 2 volume exchanges) and after reaching an osmolality value of 4 mosmol/kg. The pH-value of the HSA solution after diafiltration was pH 7.13.


UV spectrophotometric measurement of the permeate was performed three times (n=3).


Diafiltration with TFF equipment was performed by applying the following parameters:

    • stirrer: position 2
    • pump: position 2
    • pressure up-stream/inlet: max 20-30 psi
    • pressure down-stream/outlet: max 10 psi


After diafiltration, protein concentration was assessed by means of OD280. The concentration was determined 9.41 mg/mL.


The HSA solution was sterile filtered. The TFF and the membrane were flushed with approximately 1 L of distilled water. Afterwards an integrity test was done (see Operating Instructions Labscale™ TFF System, page 5-3 to 5-5, 1997). The volume flow was 1.2 mL/min, thus, the integrity test was passed (acceptable maximal limit 3 mL/min). The membrane was once more flushed with 500 mL of distilled water and then with 500 mL of 0.05 M NaOH. The membrane was stored in 0.05 M NaOH, the TFF was again flushed with approximately 500 mL of distilled water.


4.2: Concentration Process


Prior to concentrating the HSA protein solution, the concentration was assessed by means of OD280 and was determined to be 9.52 mg/mL. Four Vivaspin 20 concentrators (10 kDa) were used. 20 mL of HSA solution (9.52 mg/mL) were placed in each of 3 Vivaspin concentrators. In the fourth Vivaspin, water was filled as counterweight balance while centrifuging. The concentrators were closed and put into the centrifuge. The HSA solution was centrifuged applying 4500×g centrifugation force (in a swing out rotor).


4.3: Sample Pull


Samples of the concentrated HSA solution were pulled when the concentration reached 10 mg/mL and subsequently after every 10 mg/mL concentration increment increase (at 20 mg/mL, 30 mg/mL, 40 mg/mL etc. until approximately 180 mg/mL). Every 40 minutes the concentrators were taken out of the centrifuge, the solution was homogenized, and the solution and the centrifuge adapters were cooled for approximately 10 min on ice. After every 10 mg/mL concentration increment increase, the solutions in the concentrators were homogenized, the optical appearance was checked and samples were pulled for analysis via UV (300 μL), PCS (160 μL), SEC (120 μL) and IEC (300 μL). After the sample pull, HSA solution (9.52 mg/mL) was added to the concentrators, up to approximately 20 mL.


When the projected concentration for the HSA solution in the concentrator reached approximately 20 mg/mL, permeate was measured via OD280, revealing a concentration of 0.5964 mg/mL. The concentration of the HSA solution was only 15.99 mg/mL, which was less than expected. A sample of concentrated HSA in WFI (10 mg/mL) was analyzed via SEC to scrutinize for potential fragmentation. The HSA solution (15.99 mg/mL) in the Vivaspins was placed in falcon tubes and stored at −80° C. The remainder of the original HSA solution (9.52 mg/mL) used to fill the concentrators was also stored at −80° C.


SEC analysis was performed to determine whether the HSA protein underwent degradation, producing small fragments that could pass through the membrane. The SEC analysis, however, revealed a monomer amount of 92.45% for 10 mg/mL HSA in WFI with virtually no fragments.


The solutions that were stored at −80° C. were thawed at 25° C. and sterile filtered. The solutions in the falcon tubes were transferred in one Vivaspin 20 concentrator each (3 kDa cut-off). The Vivaspins were filled up with HSA solution (9.52 mg/mL) and centrifuged (refer to 3.2 concentration process described above).


Visual Inspection and PCS analysis were used to determine the solubility limit of HSA.


At the completion of the experiment, a concentration of approximately 180 mg/mL HSA was reached.


All SEC and IEC samples were stored at −80° C. until further analysis. UV and PCS measurements were performed directly after sample pull. The rest of the concentrated HSA solution was placed in Eppendorf repositories and stored at −80° C.


An overview of the centrifugation scheme is described above in Table 7. The duration of the centrifugation used to concentrate HSA is described in Table 11.









TABLE 11







Centrifugation Times Necessary


to Concentrate HSA Solution








concentration



[from-> to]



[mg/mL]
time [min]











10 to 20
9


20 to 30
30


30 to 40
40


40 to 50
50


50 to 60
80


60 to 70
90


70 to 80
110


80 to 90
130


 90 to 100
170


100 to 180
360










4.4: Impact of the pH Value on the Hydrodynamic Diameter of HSA


The following part of the experiment was performed to evaluate a potential impact of pH on the hydrodynamic diameter of HSA when the protein is dissolved in WFI. Four 6R vials were filled with 5.09 mL HSA solution (9.83 mg/mL), and pH values from 3 to 6 were set up with 1M HCl (actual pH: 3.04, 3.99, 5.05, 6.01). These solutions were each transferred to a separate 10 mL volumetric flask. The flasks were then filled to the calibration mark and gently shaken for homogenization.


The HCl solutions were placed in 10R vials and analyzed via PCS. The solutions were sterile filtered and measured again via PCS. Also, 5.09 mL HSA solution (9.83 mg/mL) were transferred in a 10 mL volumetric flask and this was filled with WFI to the calibration mark. The flask was gently shaken for homogenization and then the solution was sterile filtered and measured via PCS.


Sample preparation for PCS measurement:


The cuvettes were flushed with 50 μL of sample. Measurement was performed with 100 μL of sample volume.


4.5: Viscosity Measurement


For HSA in commercial formulation (200 mg/mL) and for HSA in WFI (approximately 180 mg/mL) the viscosity was measured with a capillary viscometer (Schott, MP.-No. 33.2).


A 15 mL aliquot was pulled from a 50 mL bottle of commercial formulation HSA. HSA in WFI was thawed at approximately 20° C. and approximately 9 mL were aliquotted in a Falcon tube. The density was measured with density measurement device DMA 4500, Anton Paar.


Further analysis of the high HSA concentration formulation is provided in Examples 5 to 11.


Example 5: Analysis of High Protein Formulations—Optical Appearance

In contrast to Adalimumab in the commercial formulation, Adalimumab in WFI did not reveal opalescence. J695 also did not reveal any opalescence phenomena when dissolved in WFI. Despite the fact that the protein concentration of Adalimumab was 80 mg/mL and 200 mg/mL in WFI, there was virtually no opalescence observed. In contrast, the commercial formulation comprising 50 mg/mL Adalimumab revealed notable opalescence in the commercial formulation. Thus, the use of pure water, i.e., WFI, as a dissolution medium had a positive effect on protein solution opalescence.


It was a surprising observation that (in addition to being soluble at all at such a high protein concentration) Adalimumab in WFI appeared to have a low viscosity, even at higher concentrations such as 200 mg/mL.


Depending on the concentration, the optical characteristics/color of HSA solutions changed from clear and slightly yellow (10 mg/mL in WFI) to clear and yellow (approximately 180 mg/mL in WFI).


During the concentration process, no precipitation was observed for the Adalimumab solution and HSA solution. Precipitation would have been an indication for solubility limitations. The solutions stayed clear until the experiment was finalized. It is to be highlighted that the experiments were not finished because potential solubility limits were approached and precipitation occurred, but were finished because the solution volumes remaining in the concentrators were not sufficient to proceed with concentrating (i.e. lack of material). It appears very likely that the solubility limits of Adalimumab, J695, and HSA are well beyond 220 mg/mL.


In the J695 solution a crystal like precipitate was observed when the high-concentrated solution was stored over night at 2-8° C. in the concentrators (approximately 120 mg/mL). The crystal like precipitate redissolved after approximately 3-5 min when the solution was stored at ambient temperature. Thus the environment created by dissolving J695 at high concentration in pure water provides conditions where protein crystallization might be performed by mere temperature cycling (e.g., from ambient temperature to 2-8° C.).


Example 6: Analysis of High Protein Formulations—Protein Concentration

The calculation of the protein concentrations is provided above in the Materials and Methods section.


An overview of the concentration of Adalimumab, J695, and HSA into pure water, high protein formulation is provided below in Tables 12-14:









TABLE 12







Concentrations of Adalimumab as Assessed Via OD280


during the Concentration Process












absorb-
average

concen-


sample name
ance
value
dilution
tration














Adalimumab in WFI 10 mg/ml
0.680
0.650
20
9.35



0.695






0.575





Adalimumab in WFI 20 mg/ml 1
1.064
0.813
40
23.40



0.688






0.687





Adalimumab in WFI 20 mg/ml 2
0.781
0.788
40
22.68



0.793






0.791





Adalimumab in WFI 20 mg/ml 3
0.870
0.824
40
23.71



0.883






0.719





Adalimumab in WFI 30 mg/ml 1
0.817
0.807
60
34.84



0.812






0.793





Adalimumab in WFI 30 mg/ml 2
0.839
0.827
60
35.69



0.813






0.829





Adalimumab in WFI 30 mg/ml 3
0.770
0.744
60
32.10



0.729






0.732





Adalimumab in WFI 40 mg/ml 1
0.494
0.491
100
35.35



0.493






0.488





Adalimumab in WFI 40 mg/ml 2
0.499
0.501
100
36.06



0.516






0.489





Adalimumab in WFI 40 mg/ml 3
0.495
0.512
100
36.81



0.523






0.517





Adalimumab in WFI 50 mg/ml 1
0.574
0.585
100
42.11



0.579






0.603





Adalimumab in WFI 50 mg/ml 2
0.671
0.634
100
45.63



0.630






0.601





Adalimumab in WFI 50 mg/ml 3
0.579
0.574
100
41.27



0.574






0.568





Adalimumab in WFI 60 mg/ml 1
0.838
0.837
100
60.21



0.833






0.840





Adalimumab in WFI 60 mg/ml 2
0.793
0.777
100
55.89



0.767






0.770





Adalimumab in WFI 60 mg/ml 3
0.802
0.780
100
56.10



0.759






0.779





Adalimumab in WFI 70 mg/ml 1
0.911
0.878
100
63.15



0.866






0.857





Adalimumab in WFI 70 mg/ml 2
1.012
0.996
100
71.68



0.976






1.001





Adalimumab in WFI 70 mg/ml 3
0.879
0.871
100
62.66



0.874






0.861





Adalimumab in WFI 80 mg/ml 1
0.512
0.510
200
73.45



0.489






0.531





Adalimumab in WFI 80 mg/ml 2
0.542
0.526
200
75.64



0.519






0.517





Adalimumab in WFI 80 mg/ml 3
0.551
0.531
200
76.42



0.511






0.531





Adalimumab in WFI 90 mg/ml 1
0.550
0.547
200
78.64



0.550






0.539





Adalimumab in WFI 90 mg/ml 2
0.549
0.548
200
78.80



0.551






0.543





Adalimumab in WFI 90 mg/ml 3
0.532
0.534
200
76.81



0.533






0.537





Adalimumab in WFI 100 mg/ml 1
0.640
0.628
200
90.36



0.621






0.623





Adalimumab in WFI 100 mg/ml 2
0.748
0.747
200
107.41



0.735






0.757





Adalimumab in WFI 100 mg/ml 3
0.625
0.621
200
89.39



0.616






0.623





Adalimumab in WFI 110 mg/ml 1
0.674
0.669
200
96.19



0.671






0.660





Adalimumab in WFI 110 mg/ml 2
0.693
0.668
200
96.05



0.690






0.620





Adalimumab in WFI 110 mg/ml 3
0.604
0.640
200
92.05



0.664






0.652





Adalimumab in WFI 200 mg/ml 1
0.863
0.698
400
201.00



0.612






0.621





Adalimumab in WFI 200 mg/ml 2
1.055
0.791
400
227.53



0.658






0.659





Adalimumab in WFI 200 mg/ml 3
0.732
0.665
400
191.44



0.648






0.615
















TABLE 13







Concentrations of J695 as Assessed Via OD280 during


the Concentration Process












absor-
average

concen-


sample name
bance
value
dilution
tration














ABT-874 in WFI 10 mg/ml
0.715
0.703
20
9.90



0.708






0.705





ABT-874 in WFI 20 mg/ml 1
0.686

40
19.31


ABT-874 in WFI 20 mg/ml 2
0.684

40
19.26


ABT-874 in WFI 20 mg/ml 3
0.685

40
19.29


ABT-874 in WFI 30 mg/ml 1
0.700

60
29.59


ABT-874 in WFI 30 mg/ml 2
0.703

60
29.70


ABT-874 in WFI 30 mg/ml 3
0.684

60
28.91


ABT-874 in WFI 40 mg/ml 1
0.539

100
37.97


ABT-874 in WFI 40 mg/ml 2
0.540

100
38.02


ABT-874 in WFI 40 mg/ml 3
0.520

100
36.65


ABT-874 in WFI 50 mg/ml 1
0.698

100
49.15


ABT-874 in WFI 50 mg/ml 2
0.653

100
45.95


ABT-874 in WFI 50 mg/ml 3
0.623

100
43.87


ABT-874 in WFI 60 mg/ml 1
0.834

100
58.75


ABT-874 in WFI 60 mg/ml 2
0.781

100
55.02


ABT-874 in WFI 60 mg/ml 3
0.778

100
54.76


ABT-874 in WFI 70 mg/ml 1
1.103

100
77.69


ABT-874 in WFI 70 mg/ml 2
1.102

100
77.62


ABT-874 in WFI 70 mg/ml 3
1.110

100
78.13


ABT-874 in WFI 80 mg/ml 1
0.671

200
94.45


ABT-874 in WFI 80 mg/ml 2
0.746

200
105.06


ABT-874 in WFI 80 mg/ml 3
0.664

200
93.45


ABT-874 in WFI 90 mg/ml 1
0.826

200
116.37


ABT-874 in WFI 90 mg/ml 2
0.809

200
113.92


ABT-874 in WFI 90 mg/ml 3
0.804

200
113.27


ABT-874 in WFI 100 mg/ml 1
0.861

200
121.21


ABT-874 in WFI 100 mg/ml 2
0.993

200
139.80


ABT-874 in WFI 100 mg/ml 3
0.985

200
138.73


ABT-874 in WFI 200 mg/ml 1
0.681
0.805
400
226.67



0.864






0.869





ABT-874 in WFI 200 mg/ml 2
0.690
0.767
400
216.10



0.828






0.784





ABT-874 in WFI 200 mg/ml 3
0.708
0.745
400
209.83



0.789






0.738










Tables 14a and 14b: Concentrations of HSA as Assessed Via OD280 during the Concentration Process












TABLE 14a





sample name
absorbance
dilution
concentration


















HSA in WFI 10 mq/ml
0.515
20
9.88


HSA in WFI 30 mg/ml 1
0.398
60
22.94


HSA in WFI 30 mq/ml 2
0.395
60
22.73


HSA in WFI 30 mg/ml 3
0.400
60
23.00


HSA in WFI 40 mg/ml 1
0.383
100
36.78


HSA in WFI 40 mg/ml 2
0.389
100
37.33


HSA in WFI 40 mg/ml 3
0.368
100
35.29


HSA in WFI 50 mq/ml 1
0.479
100
45.97


HSA in WFI 50 mg/ml 2
0.496
100
47.61


HSA in WFI 50 mq/ml 3
0.465
100
44.61


HSA in WFI 60 mg/ml 1
0.609
100
58.47


HSA in WFI 60 mg/ml 2
0.653
100
62.69


HSA in WFI 60 mg/ml 3
0.568
100
54.52


HSA in WFI 70 mg/ml 1
0.645
100
61.89


HSA in WFI 70 mg/ml 2
0.623
100
59.76


HSA in WFI 70 mg/ml 3
0.618
100
59.28


HSA in WFI 80 mq/ml 1
0.393
200
75.37


HSA in WFI 80 mg/ml 2
0.436
200
83.69


HSA in WFI 80 mg/ml 3
0.363
200
69.67


HSA in WFI 90 mg/ml 1
0.484
200
92.90


HSA in WFI 90 mg/ml 2
0.439
200
84.22


HSA in WFI 90 mq/ml 3
0.419
200
80.50


HSA in WFI 100 mg/mL 1
0.604
200
115.93


HSA in WFI 100 mg/mL 2
0.573
200
110.00


HSA in WFI 100 mg/mL 3
0.585
200
112.30




















TABLE 14b






absorbance





sample name
value
average
dilution
concentration







HSA in WFI 180 mg/ml 1
0.946
0.952
200
182.79



0.950






0.961





HSA in WFI 180 mg/ml 2
0.994
0.929
200
178.24



0.906






0.886





HSA in WFI 180 mg/ml 3
0.843
0.896
200
172.05



0.963






0.884









All three proteins evaluated remained soluble in the concentration ranges evaluated (i.e. >200 mg/mL for Adalimumab and J695, >175 mg/mL for HSA). No indications of insolubility, e.g., the clouding phenomena or precipitation occurring in the solution, were observed. For Adalimumab, the results indicate that, over the concentration range evaluated, all Adalimumab isoforms (i.e. lysine variants) remained soluble, as no precipitation occurred at all. This observation is also consistent with ion exchange chromatography data described in Example 11, which describes that the sum of lysine variants stayed virtually consistent regardless of Adalimumab concentration.


Example 7: Analysis of High Protein Formulations—Viscosity

7.1: Adalimumab Viscosity


The viscosity of Adalimumab (approximately 50 mg/mL) in water for injection was determined to be around 1.5-2 mPas. For Adalimumab (approximately 200 mg/mL) in WFI, two values were determined. The one value determined with cone/plate rheometer from Malvern (Gemini 150) was approximately 6-6.5 mPas, while the other value (measured with cone/plate rheometer from Anton Paar, MCR 301) was approximately 12 mPas. Adalimumab commercial formulation (approximately 194 mg/mL) viscosity:


K—constant of the capillary


t—the time the solution needs for passing the capillary [s]


v—kinematic viscosity


η—dynamic viscosity


ρ—density
















time
K
v
ρ
η


[s]
[mm2/s2]
[mm2/s]
[g/cm3]
[mPas]







279.36
0.03159
8.825
1.05475
9.31









The viscosity of Adalimumab in WFI (approximately 200 mg/mL) was determined to be approximately 12 mPas with the viscosimeter from Anton Paar and approximately 6 mPas determined with the viscometer from Malvern. In contrast, the viscosity of Adalimumab in the commercial formulation (approximately 194 mg/mL) is higher, at 9.308 mPas (measured with the capillary viscometer from Schott).


7.2: Human Serum Albumin Viscosity


HSA commercial formulation (approximately 200 mg/mL) viscosity:


K—constant of the capillary


t—the time the solution needs for running through the capillary [s]


v—kinematic viscosity


η—dynamic viscosity


ρ—density
















time
K
v
ρ
η


[s]
[mm2/s2]
[mm2/s]
[g/cm3]
[mPas]







337.69
0.01024
3.46
1.05475
3.66










HSA in WFI (approximately 180 mg/mL) viscosity:


K—constant of the capillary


t—the time the solution needs for running through the capillary [s]


v—kinematic viscosity


η—dynamic viscosity


ρ—density
















time
K
v
ρ
η


[s]
[mm2/s2]
[mm2/s]
[g/cm3]
[mPas]







185.3
0.09573
17.72
1.07905
19.12









The viscosity of HSA in WFI (approximately 180 mg/mL) was determined to be approximately 19.121 mPas. The viscosity of HSA in the commercial formulation (approximately 194 mg/mL) was determined to be 9.308 mPas (measured with the capillary viscometer from Schott).


7.3: Analysis of the Viscosities of Adalimumab and HSA


The dynamic viscosity of Adalimumab 50 mg/mL in WFI was lower than the viscosity of Adalimumab 200 mg/mL in WFI and in commercial buffer, respectively. For HSA the dynamic viscosity for a concentration of 180 mg/mL in WFI was about six-fold higher than for a concentration of 200 mg/mL in commercial buffer. Thus, it seems that the intensity of viscosity change (i.e. increase and decrease, respectively) due to effects conveyed by pure water as dissolution medium may depend on the individual protein.


Example 8: Analysis of the Hydrodynamic Diameters of High Protein Formulations—Photon Correlation Spectroscopy (PCS)

The following example provides an analysis of the hydrodynamic diameter (Dh) (the z-average of the mean hydrodynamic molecule diameter) of various proteins in aqueous formulations obtained using the DF/UF methods of the invention.


8.1: Adalimumab Hydrodynamic Diameter


As shown in FIGS. 5 and 6, a trend can be observed where the hydrodynamic diameter (Dh) increases with increasing Adalimumab concentration. FIG. 5 shows the correlation between hydrodynamic diameter (z-average) and the concentration of Adalimumab in WFI. FIG. 6 shows the correlation between hydrodynamic diameter (peak monomer) and the concentration of Adalimumab in WFI.


The difference between the Dh determined from the 23.27 mg/mL sample compared to the 34.20 mg/mL sample exists because of assumptions made in the Standard Operating Procedure (SOP) for hydrodynamic diameter measurement. For Adalimumab samples having ≤23.27 mg/mL concentration, PCS measurements were performed with a SOP that assumes a 1.1 mPas value for the viscosity of the samples. For Adalimumab samples having ≥34.20 mPas, a SOP assuming a 1.9 mPas sample viscosity was used. It is known that PCS data are strongly influenced by the given viscosity of the sample solution, as PCS data is based on random Brownian motion of the sample specimen, which is impacted by sample viscosity. Thus, the increase in the hydrodynamic diameter with increasing protein concentration can be explained, as increasing protein concentration raises the viscosity of the solution (higher viscosity leads to lower Brownian motion and higher calculated Dh data). The protein molecules experience a lower random Brownian motion and thus, for a given viscosity, the hydrodynamic diameters of the sample specimen are calculated higher. Overall, the z-average based Dh values and the Dh values of the monomer match well. Additionally, no increase in Dh indicative for protein insolubility is observed with increasing concentration (i.e. high molecular weight aggregates and precipitate (if present) would induce a substantial increase in Dh).


8.2: J695 Hydrodynamic Diameter



FIGS. 7 and 8 show that the hydrodynamic diameter of J695 was relatively independent from the protein concentration until a 114.52 mg/mL concentration was reached. Increasing the J695 concentration from 114.52 mg/mL to 133.25 mg/mL induced a rapid increase in Dn. The hydrodynamic diameter at the 217.53 mg/mL concentration was higher than at 114.52 mg/mL. This finding was not surprising as both protein solutions were measured using the same SOP (assuming same viscosity of 1.9 mPas), when in reality the viscosity increases as the protein concentration increases. The strong increase from 114.52 mg/mL to 133.25 mg/mL thus can be explained as an artifact.


8.3: Human Serum Albumin Hydrodynamic Diameter


The hydrodynamic diameter of HSA in WFI was found to decrease as concentrations rose from 9.88 mg/mL to 112.74 mg/mL. From 112.74 mg/mL to 177.69 mg/mL, however, the hydrodynamic diameter was found to increase.


HSA showed a general tendency of increasing hydrodynamic diameters (peak monomer) with increasing protein concentration which is in-line with underlying theoretical principles. The Dh decrease from 9.88 mg/mL to 22.89 mg/mL is caused by a change in the measurement SOP (switching from assumed viscosity of 1.1 mPas to an assumed viscosity of 1.9 mPas).


Numerical data describing the above is provided in Appendix A.


Example 9: J695: Impact of Excipients of the Hydrodynamic Diameter

Having found the surprising result that proteins can be dissolved in high concentrations in pure water, the impact of ionizable and non-ionizable excipients typically used in parenteral formulations on the hydrodynamic diameter was evaluated. J695 was used as a model protein.


Table 15 shows that the solution osmolality is directly proportional to the concentration of sodium chloride. The osmolality in the protein solution rises along with the NaCl concentration (an almost linear correlation). Interestingly, the hydrodynamic diameter of J695 protein increased with increasing salt concentration. NaCl is an ionic excipient and dissociated into positively charged sodium ions and negatively charged chloride ions which might adsorb at the surface of the protein. Without salt being present, the hydrodynamic diameter of J695 was dramatically lower than what normally is expected for J695 (usually values around 10 nm are determined).


As illustrated in Table 15, the osmolality increased linearly with an increase in concentration of mannitol in the protein solution. In contrast, the hydrodynamic diameter did not show a dependence on mannitol concentration. Mannitol is a non-ionic sugar alcohol/polyol. Mannitol or polyols are used as stabilizers during parenteral formulation development and in final formulations. Mannitol stabilizes the protein by preferential exclusion. As other osmolytes, mannitol is preferentially excluded from the surface of the protein and it is outside of the hydrate shell of the protein. Thus the folded state of the protein is stabilized because the unfolded state, which has a larger surface, becomes thermodynamically less favorable (Foster, T. M., Thermal instability of low molecular weight urokinase during heat treatment. III. Effect of salts, sugars and Tween 80, 134 International Journal of Pharmaceutics 193 (1996); Singh, S. and Singh, J., Effect of polyols on the conformational stability and biological activity of a model protein lysozyme, 4 AAPS PharmSciTech, Article 42 (2003)). However, it is interesting that the osmolality can be adjusted basically as desired—which would be an important feature of the protein findings described herein—without impacting the Dh of the protein. These findings may be useful in high-concentration protein formulation, where viscosity related manufacturing and dosing issues may be present, as the osmolality adjustment with mannitol is not mirrored by an increase in protein Dh (meaning viscosity is expected to remain constant).









TABLE 15





Impact of Excipients on J695 Osmolality and Z-Average

















NaCl




concentration
osmolality
z-average


[mg/ml]
[mosmol/kg]
[nm]





0
16
4.19


2
92
12.2


4
158
16.2


6
230
17





mannitol




concentration
osmolality
z-average


[mg/ml]
[mosmol/kg]
[nm]





0
16
4.19


2
148
5.49


4
276
3.22


6
432
3.54









Example 10: Analysis of High Protein Formulations with Size Exclusion Chromatography (SEC)

For the SEC analysis, samples of Adalimumab, J695 and HSA were diluted to 2 mg/mL before injection. The injection volume for Adalimumab was 20 μL. For J695 and HSA, a 104, injection volume was used.


10.1: SEC Analysis of Adalimumab


The amount of monomer of Adalimumab tended to slightly decrease from 99.4% to 98.8% while concentrating from 9.35 mg/mL to 206.63 mg/mL. That decrease of monomer is associated with an increase in the amount of aggregate in Adalimumab solution from 0.4% to 1.1% while concentrating from 23.27 mg/mL to 206.62 mg/mL, respectively. The amount of fragment remained constant at 0.1%, independent of protein concentration (see table in Appendix B). Thus, Adalimumab was stable in WFI.


Overall, the increase in protein aggregation with increasing protein concentration is deemed only minor. A similar trend in monomer decrease would be expected when either the protein was formulated in a buffer system and when additionally surfactants are added. Adalimumab protein appears to be surprisingly stable when formulated in pure water.


10.2: SEC Analysis of J695


The amount of J695 monomer slightly decreased from 99.4% to 98.6% with increasing protein concentration from 9.99 mg/mL to 217.53 mg/mL. The decrease of monomer was associated with an increase in aggregate from about 0.4% to about 1.2% with increasing protein concentration from 9.99 mg/mL to 217.53 mg/mL. Independent from protein concentration, the amount of fragment was almost constant with 0.17% to 0.23% with increasing protein concentration from 9.99 mg/mL to 217.53 mg/mL.


Overall, the increase in protein aggregation with increasing protein concentration was deemed only minor. A similar trend in monomer decrease would be expected when the protein is formulated in buffer systems and when additional surfactants are added. Thus, J695 protein appears to be surprisingly stable when formulated in pure water.


10.3: SEC Analysis of HSA


The amount of monomer HSA decreased from 95.9% to 92.75% while concentrating from 9.88 mg/ml to 112.74 mg/mL. For the sample with 177.69 mg/mL, an increase in monomer up to 94.5% was determined. The decrease of the amount of monomer goes along with an increase in protein aggregate from 4.1% to 7.25% while concentrating from 9.88 mg/mL to 112.74 mg/mL. Thus, HSA protein also appears to be stable when formulated in pure water.


Numerical data describing the above-mentioned SEC experiments is provided in Appendix B.


Example 11: Analysis of High Protein Formulations—Ion-Exchange-Chromatography (IEC)

For the IEC analysis the samples of Adalimumab, J695 and HSA were diluted to 1 mg/mL before injection. The injection volume for all proteins was 100 μL.


11.1: IEC Analysis of Adalimumab


As shown in FIG. 9, Adalimumab was stable in WFI. FIG. 9 shows a slight trending which may be interpreted as indicating that the sum of lysine variants (lysine 0, 1 and 2) decreases with an increase concentration of Adalimumab in WFI. Overall, however, the percentage of lysine variants varied less than 0.25%.


11.2: IEC Analysis of J695



FIG. 10 shows that the sum of the J695 peaks 1 to 7 is slightly decreasing with increasing J695 concentration. With the decrease in peak 1-7 the sum of acidic and basic peaks is slightly increasing, with the increase in the acidic peaks being a little more pronounced (see FIGS. 11 and 12). The sum of acidic peaks slightly increases from approximately 10.2% to 10.6% and the sum of basic peaks from 0.52% to 0.59%, respectively.


Overall, it can be stated that no major instability effects or insolubility effects of J695 formulations in pure water were observed via IEC.


Numerical data describing the above IEC experiments is provided in Appendix C.


Summary of Findings in Examples 2-11

It was initially thought that transferring proteins, such as antibodies, into WFI would likely induce protein precipitation by concentrating the protein beyond its solubility limit in pure water. The above studies demonstrate that proteins, including antibodies, not only can be transferred into pure WFI at lower concentrations without encountering any precipitation phenomena and solubility limitations, but that, surprisingly, Adalimumab (as well as the other two test proteins) can be concentrated in pure water to ultra-high concentrations beyond 200 mg/mL using UF/DF and centrifugation techniques (e.g., TFF equipment, Vivaspin devices). In addition, Adalimumab opalescence was unexpectedly found to be substantially reduced when the protein was formulated in WFI. Osmolality was monitored to ensure that the Adalimumab buffer medium was completely exchanged by pure, salt free water (i.e. WFI). Moreover, freeze-thaw processing was performed during sample preparation for analysis, and virtually no instability phenomena were observed with SEC and IEC analysis.


The approach of formulating proteins, e.g., Adalimumab, at high concentrations in WFI revealed the potential to reduce viscosity phenomena, which often impedes straightforward Drug Product development at high protein concentrations.


Finally, the hydrodynamic diameter (determined via photon correlation spectroscopy, PCS) of Adalimumab was found to be notably lower in WFI than in commercial buffer (indicative of lower viscosity proneness).


Overall, it was concluded that the findings of antibodies and the globular model protein HSA being soluble in pure water in ultra-high concentrations have potential to provide new insight into fundamental protein regimes and to potentially offer new approaches in protein drug formulation and manufacturing, for instance by:

    • reducing opalescence of high-concentrated protein formulations
    • reducing viscosity of high-concentrated protein formulations
    • enabling to adjust osmolality as desired in protein-WFI solutions by adding non-ionic excipients such as mannitol without changing features such as viscosity and non-opalescence (it was demonstrated for J695 that hydrodynamic diameter and opalescence did not change when mannitol was added, but increased dramatically when NaCl was added)
    • providing a new paradigm in Drug Substance formulation and processing, as it was demonstrated that proteins can be subjected to operations such as DF/UF for concentrating the protein in WFI to ultra-high concentrations and to freeze and thaw without substantial stability implications. Given the background that it is well-known that during DF/UF the composition of protein formulations, especially during processing to high concentrations, necessarily changes (Stoner, M. et al., Protein-solute interactions affect the outcome of ultrafiltration/diafiltration operations, 93 J. Pharm. Sci. 2332 (2004)), these new findings could beneficially be applied by either adjusting the Drug Substance concentration by DF/UF of the protein in pure water, and add excipients at high DS concentrations subsequently (by this avoiding the risk of DS formulation changes during process unit operations). Alternatively, the excipients could be added to Drug Substance during final Drug Product fill-finishing.


Example 12: Preparation of Adalimumab in Water Formulation

The following example illustrates the scaling up the DF/UF procedures resulting in large scale production of adalimumab in water.


12.1: Evaluation of Process Parameters


Dialysis process evaluation studies were performed on a laboratory scale to define suitable parameters for the dialysis of Bulk Adalimumab Drug Solution formulated in a phosphate/citrate buffer system containing other excipients, e.g., mannitol and sodium chloride (FIGS. 13 and 14).


Conductivity measurements may be taken with any commercially available conductivity meter suitable for conductivity analysis in protein solutions, e.g. conductivity meter Model SevenMulti, with expansion capacity for broad pH range (Mettler Toledo, Schwerzenbach, Switzerland). The instrument is operated according to the manufacturers instructions (e.g., if the conductivity sensor is changed in the Mettler Toledo instrument, calibration must be performed again, as each sensor has a different cell constant; refer to Operating Instructions of Model SevenMulti conductivity meter). If the instructions are followed, conductivity measurements can be taken by directly immersing the measuring probe into the sample solution.



FIG. 13 shows the efficiency of the dialysis procedure, in terms of the reduction of components responsible for the osmolality and the conductivity of the formulation containing adalimumab at 74 mg/ml. After a reduction of the solutes in the antibody solution by a factor of 100, osmolality and conductivity measurements largely stabilized at levels far below the original measurements of these parameters from the commercial formulation.



FIG. 14 shows the stability of pH in dialyzed Adalimumab bulk solutions. pH levels before and after dialysis against deionized water (1:1,000,000) are shown for Adalimumab solutions with a range of different initial pH readings. pH levels remained nearly the same in the retentate before and after dialysis.


12.2: Production of High-Concentrated Adalimumab in Water Bulk Drug Solution


In a first step, the formulated bulk drug solution (Phosphate/Citrate Buffer system containing other excipients, e.g., Mannitol and Sodium Chloride) was up-concentrated by Ultrafiltration/Diafiltration to a concentration of approximately 100 mg/ml (12 L scale, Millipore Pellicon 2 Mini Bio-A MWCO 10k columns). In a second step, the up-concentrated solution was dialyzed against deionized water (SpectraPor7 MWCO10k, dilution factor 1:100,000). As a third step the dialyzed solution was up-concentrated by Ultrafiltration/Diafiltration to a concentration of approximately 250 mg/ml using Millipore Pellicon 2 Mini Bio-A MWCO 10k columns.


Table 16a shows the results of analysis of highly concentrated Adalimumab in water (DF/UF processed) bulk drug solutions after Step 3 of the procedure.









TABLE 16a







Osmolality and Conductivity Data for DF/UF


Bulk-Processed Adalimumab













osmolality
conductivity

density
Adalimumab



mosm/kg
mS/cm
pH
g/cm3
cone mg/ml







62
0.95
5.28
1.0764
277.8










12.3: Freeze/Thaw (F/T) Procedure Simulating Manufacturing Conditions

Freezing was conducted using an ultra-low temperature freezer (Revco Ultima II, 20 cu.ft.) with a manufacturing scale load of 47 kg of liquid to be frozen at a temperature below −50° C., typically −70° C. to −80° C. The liquid was packaged in individual bottles of 1.6 kg fill weight (e.g., Nalgene 2 L PETG square media). Freezing was completed after 48 hours. Thawing was conducted in a circulating water bath (e.g., Lindergh/Blue) with a manufacturing scale load of 24 kg at a temperature between 20° C. and 40° C., typically 30° C., until the material was completely thawed.


12.4: Bottle Mapping During Freeze and Thaw


Individual horizontal solution layers in the bottle volume were isolated and analyzed. At protein concentrations of 250 mg/ml and 200 mg/ml, only minimal gradient formation was detected in the Adalimumab water solution, as seen in FIGS. 15 through 19. Freezing and thawing of formulated Adalimumab solutions (solutions with a Phosphate/Citrate Buffer system containing other excipients, e.g., Mannitol and Sodium Chloride) at 250 mg/ml and 200 mg/ml, however, led to the formation of precipitate on the bottom of the bottle.


12.5: Gradient Formation in Commercial and Low-Ionic Formulations of Adalimumab


The formation of gradients by freeze thaw procedures in commercial and low-ionic (water) formulations of Adalimumab was compared. Table 16b shows the results of visual inspection of commercial Adalimumab solutions of various concentrations after a f/t step. The formation of precipitates indicates that instability was created in the solution by the f/t procedure. Above 100 mg/ml, significant precipitate formation was observed. Table 17 shows the analytical data of two 50 mg/ml solutions and one 100 mg/ml low-ionic formulation before the freeze-thaw experiment.









TABLE 16b







Observed Precipitation in Commercial Adalimumab Solutions after F/T















250 mg/ml
220 mg/ml
200 mg/ml
150 mg/ml
120 mg/ml
100 mg/ml
60 mg/ml





Commericial
Precipitate
Precipitate
Precipitate
Precipitate
Partial
Clear
Clear


freezing




Precipitate




process in









ultra low









temp freezer:









−70° C./23° C.
















TABLE 17







Solution Analytical Data before Freeze-Thaw

















Protein





Density
Osmolality
conc



formulation
pH
(g/cm3)
(mOsmol/kg)
(mg/mL)















E167 130 01 CL
low-ionic
5.18
1.0121
5
49.3


50 mg/mL in







water







E167 140 01 CL
Commercial
5.20
1.0224
280
48.7


50 mg/mL in
formulation






buffer







100 mg/mL in
low-ionic
5.32
1.0262
12
99.8


water















About 1600 mL (50 mg/ml solutions) or 800 ml (100 mg/ml solution) of each formulation were placed into PETG bottles and subjected to a conventional freeze (−80° C.) thaw (23° C., water bath) procedures. Samples were then pulled from top, center and bottom of the PETG bottles and analyzed for pH, density, osmolality, and protein concentration. Analysis results are shown in Table 18.









TABLE 18







Analysis of Bottle-Mapped Layers from Frozen/Thawed


Solutions protein content













density
osmolality
(volumetric)


sample
pH
g/cm3
mOsmol/kg
mg/mL










50 mg/ml in water











top
5.20
1.0119
6
48.72


middle
5.19
1.0120
8
49.35


bottom
5.17
1.0120
6
49.76







commercial formulation











top
5.16
1.0165
236
37.9


middle
5.13
1.0221
306
45.58


bottom
5.12
1.0257
368
55.48







100 mg/ml in water











top
5.29
1.0259
13
98.7


middle
5.3
1.0262
16
99.9


bottom
5.28
1.0262
14
101.2









The commercial formulation of Adalimumab revealed significant gradients upon freeze/thaw with regard to density (indicating heterogeneities/gradients of protein and excipients), osmolality (indicating excipient gradients), and protein content. In contrast, no gradients were found in the 50 mg/ml low-ionic Adalimumab formulation upon freeze/thaw.


At higher protein concentrations, gradient formation may sometimes be expected to become worse. However, no gradients were found in the 100 mg/ml low-ionic Adalimumab formulation upon freeze/thaw with regard to pH, density, osmolality and protein concentration.


Example 13: Stability of J695 after DF/UF

The following example provides data on the stability of J695 after DF/UF processing in accordance with the methods of the invention.


Protein samples from J695 in normal DS buffer were analyzed, either after pH adjustment or after diafiltration. pH was adjusted to pH 4.4 with 0.1M phosphoric acid, protein concentration 112 mg/mL. For concentrated samples in WFI, protein samples were diafiltered (DF/UF) against water for approximately 1.5 days at ambient temperature, using a TFF equipped with a 30 kDa RC membrane. The protein concentration after DF/UF was determined approx. 192 mg/mL. pH 4.7.


13.1: Size Exclusion Analysis (SEC) Experimental Procedures

A size exclusion method was developed for the purity assessment of J695. Size exclusion chromatography (SEC) separates macromolecules according to molecular weight. The resin acts as a sieving agent, retaining smaller molecules in the pores of the resin and allowing larger molecules to pass through the column. Retention time and resolution are functions of the pore size of the resin selected.


Each sample was diluted to 2.5 mg/mL with purified water (Milli-Q) based on the stated concentration. 50 μg of each sample was injected onto the column in duplicate. A Tosoh Bioscience G3000swxl, 7.8 mm×30 cm, 5 μm (Cat #08541) SEC column is used for separation. For Buffer A, 211 mM Na2SO4/92 mM Na2HPO4, pH 7.0 was used. Detection was performed at 280 nm and 214 nm. The column was kept at room temperature with a flow rate of 0.3 mL/min.


This chromatography utilized an isocratic gradient with a 100% mobile phase A solvent for 50 minutes duration.


13.2: SEC Data


Table 19 describes data from size exclusion chromatography experiments.









TABLE 19







SEC Analysis Data for J695 Reference Standard,


DS and Post-DF/UF (in water)












ABT-874
load
HM
Monom
Frag
















BF Ref Std
50 u
0.49
97.9
1.28
0.26


BF Ref Std dup
50 u
0.41
98.0
1.29
0.27


BF Ref std avg

0.45
98.0
1.29
0.27


std

0.06
0.05
0.01
0.01


% RS

12.5
0.05
0.55
2.67


DS in Buffer pH 4.4
50 u
0.42
98.3
1.04
0.16


DS in Buffer pH 4.4 dup
50 u
0.40
98.4
1.01
0.13


DS in Buffer pH 4.4 avg

0.41
98.4
1.03
0.15


std

0.01
0.06
0.02
0.02


% RS

3.45
0.06
2.07
14.6


DS UF/DF in Water pH 4.7
50 u
0.69
98.1
1.04
0.14


UF/DF in Water pH 4.7 dup
50 u
0.69
98.0
1.07
0.16


DS UF/DF in Water pH 4.7 avg

0.69
98.1
1.06
0.15


std

0.00
0.04
0.02
0.01


% RS

0.00
0.04
2.01
9.43









The data in Table 19 shows that the commercial formulation of J695 (DS PFS, pH=4.4) has comparable levels of fragments and aggregate as the J695 reference standard. There was a difference noted in the aggregate amount between the commercial formulation J695 control and the J695 that had undergone DF/UF, in water (DS in H2O, pH=4.7, 192 mg/ml): an increase from 0.4% to 0.7% in aggregation was seen. This was not a significant increase and may be due to time spent at room temperature during the UF/DF. There is no change to the fragments.


13.4: IEC (WCX-10) Experimental Procedure


A cation exchange method was developed for the assessment of the heterogeneity of J695 using the Dionex WCX-10 column. Generally, cation exchange chromatography separates protein isoforms according to the apparent pI and the surface charge interaction with the resin. The protein of interest is bound to the column under specific low salt starting conditions and is eluted from the column by increasing the salt concentration through a gradient. Proteins with lower apparent pI bind less tightly to a cation exchange column and are the first to elute and proteins with a higher apparent pI bind tighter and are the last to elute.


Cation exchange chromatography using WCX-10 was used in quality control as a lot release assay. The assay conditions were modified to improve separation of known J695 isoforms.


The sample was diluted to 1.0 mg/mL with purified water (Milli-Q). Reference standard was run in triplicate as a comparison and was diluted to 1 mg/ml in purified water (Milli-Q).


Dionex Propac WCX-10 columns (p/n 054993), along with corresponding guard columns (p/n 054994), were used for separation. Buffers used in the procedure included Buffer A (10 mM Na2HPO4, pH=6.0) and Buffer B (10 mM Na2HPO4, 500 mM NaCl, pH=6.0). Column temperature was maintained at 35° C. and column flow rate was 1 mL/min. Injection volumes were 100 μl for a 100 μg load and detection was performed at 280 nm. Buffer gradients over the course of the chromatographic separation are provided in Table 20.









TABLE 20







Buffer Gradients used in IEC Analysis of J695









Time (min)
% MPA
% MPB












0
75
25


3
60
40


33
40
60


36
0
100


41
0
100


43
75
25


48
75
25










13.5: IEC Data


Table 21 provides results from experiments comparing the J695 Reference Standard to J695 in commercial buffer (DS pH=4.4), as well as a comparison of the commercial buffer formulation to J695 after DF/UF (DF/UF H2O, pH=4.7).









TABLE 21







IEC Data for J695 Reference Standard, Commercial Formulation (DS) and after DF/UF (in water)

















0 glu
0 glu
1 glu
1 glu
1 glu
2 glu
2 glu





(1)
(2 + 2a)
(3)
(4)
(5) + (5a)
(6)
(7)
acidic
basic



















ref std
43.77
7.55
8.00
21.87
4.28
4.82
3.75
4.05
1.92


ref std dup
43.49
7.49
7.98
21.70
4.26
4.81
3.75
4.61
1.90


ref std dup
43.44
7.49
8.00
21.65
4.24
4.81
3.74
4.75
1.89


ref std avg
43.57
7.51
7.99
21.74
4.26
4.81
3.75
4.47
1.90


SD
0.20
0.04
0.01
0.12
0.01
0.01
0.00
0.40
0.01


% RSD
0.45
0.56
0.18
0.55
0.33
0.15
0.00
8.86
0.74


ABT-874 DS pH = 4.4
35.65
14.74
7.26
18.06
6.76
5.32
3.98
5.55
2.70


ABT-874 DS pH = 4.4 Dup
35.82
14.73
7.29
18.14
6.82
5.39
4.06
4.79
2.95


ABT-874 DS pH = 4.4 avg
35.74
14.74
7.28
18.10
6.79
5.36
4.02
5.17
2.83


SD
0.12
0.01
0.02
0.06
0.04
0.05
0.06
0.54
0.18


% RSD
0.34
0.05
0.29
0.31
0.62
0.92
1.41
10.39
6.26


ABT-874 DF/UF H2O
36.57
14.51
7.26
18.09
6.57
5.22
3.91
5.28
2.61


pH = 4.7











ABT-874 DF/UF H2O
36.60
14.43
7.25
18.02
6.66
5.18
4.00
5.31
2.56


pH = 4.7 Dup











ABT-874 DF/UF H2O
36.59
14.47
7.26
18.06
6.62
5.20
3.96
5.30
2.59


pH = 4.7 avg











SD
0.02
0.06
0.01
0.05
0.06
0.03
0.06
0.02
0.04


% RSD
0.06
0.39
0.10
0.27
0.96
0.54
1.61
0.40
1.37










13.6: IEC Analysis Conclusions


There were some differences noted between the J695 Reference Standard and the commercial formulation (DS, pH 4.4). These differences were noted in the initial run of the DS engineering run sample and are attributed to differences in the manufacturing processes between the 3000 L and 6000 L campaigns. There were no notable differences between the DS, pH 4.4 control and the J695 in H2O pH=4.7, 192 mg/ml sample.


Example 14: Stability of Adalimumab after DF/UF and Long-Term Storage at 2-8° C.

The following example provides data showing the stability of Adalimumab in an aqueous formulation in accordance with the methods of the invention, after 22.5 months storage at 2-8° C.


Adalimumab samples for SEC and WCX-10 analysis were diafiltered against water and concentrated to about 177 mg/mL. Samples were stored and analyzed at various time points for stability.


Standard Adalimumab solution (DS, pH approx. 5.2) in commercial Humira buffer was used as a starting material for generating a concentrated solution in water. Protein solution samples were diafiltered (DF/UF) against water for approximately 1.5 days at ambient temperature, using a TFF equipped with a 30 kDa RC membrane. Protein concentration after DF/UF was determined approximately 177 mg/mL, pH 5.2. The sample was stored at 2-8° C. for 22.5 months before analysis.


14.1: SEC Experimental Procedure


A size exclusion method was previously developed to check for the presence of antibody fragments and aggregates. Size exclusion chromatography (SEC) separates macromolecules according to molecular weight. The resin acts as a sieving agent, retaining smaller molecules in the pores of the resin and allowing larger molecules to pass through the column. Retention time and resolution are functions of the pore size of the resin selected.


Each sample was diluted to 1.0 mg/mL with milli Q water and 50 μg of each sample was injected onto the column. For SE-HPLC, a Sephadex 200 column (Pharmacia cat #175175-01, S/N 0504057) or a TSK gel G3000SW (cat #08541; for analysis of 22.5 month samples) were used. The mobile phase of the column comprised 20 mM Sodium phosphate and 150 mM Sodium chloride, pH 7.5. Detection was performed at 280 nm and 214 nm. Columns were kept at ambient temperature and the flow rate was 0.5 mL/min (Sephadex column), or 0.3 mL/min (TSK column).


14.2: SEC Data



FIG. 20 and Table 22 contain results of the analysis of a low-ionic Adalimumab solution stored as a liquid at 2-8° C. for 8.5 months compared to the same solution stored at −80° C. Table 23 contains analysis data for a low-ionic Adalimumab solution stored at 2-8° C. for 22.5 months compared to a reference standard sample of Adalimumab.









TABLE 22







SEC Analysis Data Comparing Adalimumab from Frozen


Storage versus Adalimumab from Long-Term Refrigerated Storage











Sample
Load
% HMW
% Monomer
% LMW





DF/UF against water,
50 μg
0.1
99.6
0.3


177 mg/mL,






4.5 months at −80° C.






DF/UF against water,
50 μg
0.2
99.5
0.3


177 mg/mL,






9 months at 2-8° C.
















TABLE 23







SEC Analysis Data Comparing Adalimumab Reference Standard


against Adalimumab from Long-Term Refrigerated Storage













%
%
%


Sample
Load
HMW
Monomer
LMW














Reference Std. Adalimumab
50 μg
0.31
98.85
0.84


DF/UF against water, 177
50 μg
1.42
97.59
0.98


mg/mL, 22.5 months at






2-8° C.










As can be seen in Table 22, SEC analysis revealed that adalimumab in water was stable even after 9 months at 2-8° C. or for 4.5 months at −80° C., as the percent aggregate (% HMW) and percent fragment (% LMW) were minimal over time.


14.3: SEC Analysis Conclusions


After 8.5 months storage at 2-8° C., the Adalimumab solution (DF/UF against water) revealed a small fraction of high molecular weight (HMW) species (0.2%) and a small fraction of fragment (0.3%). Storage for 4.5 months at −80° C. and subsequent thaw (water bath, 23° C.) did not impact Adalimumab stability (0.1% aggregate, 0.3% fragment).


Analysis of a sample stored for 22.5 months at 2-8° C. also shows comparable fragment content to Adalimumab reference standard (Table 23). However, the aggregate levels detected in the 22.5 month stability sample (1.66%) are somewhat higher than aggregate levels detected in the reference standard.


It is known that self-association of antibodies is highly dependent on the antibody concentration, i.e. the formation of non-covalent aggregate and associate complexes is most pronounced at high protein concentration. This self-association is reversible, and dilution with buffer solution results in reduced self-association tendencies (Liu, J. et al., 94 Journal of Pharmaceutical Sciences 1928 (2004)).


Thus, it is likely that differences in sample preparations and different lag-times between Adalimumab solution dilution (from 177 mg/mL to 1 mg/mL) and subsequent sample analysis by SEC are the reason for the differences in aggregate content of the 8.5 month and the 9 month stability samples.


14.4: IEC Experimental Procedure


A cation exchange method was developed for the assessment of antibody charge heterogeneity using the Dionex WCX-10 column. Cation exchange chromatography separates protein isoforms according to the apparent pI and the surface charge interaction with the resin. The protein of interest is bound to the column under specific low salt starting conditions and is eluted from the column by increasing the salt concentration through a gradient. Proteins with lower apparent pI bind less tightly to a cation exchange column and are the first to elute and proteins with a higher apparent pI bind tighter and are the last to elute.


Before the procedure, samples were diluted to 1.0 mg/mL with milli Q water. Dionex Propac WCX-10 columns (p/n 054993), along with a corresponding guard columns (p/n 05499), were used for separation. Two mobile phase buffers were prepared, 10 mM Sodium phosphate, pH 7.5 (Buffer A) and 10 mM Sodium phosphate, 500 mM Sodium chloride, pH 5.5 (Buffer B). Columns were kept at ambient temperature and the flow rate was 1.0 mL/min. Injection volumes were 100 μl for a 100 μg load and detection was performed at 280 nm. Buffer gradients over the course of the chromatographic separation are provided in Table 24.









TABLE 24







Buffer Gradients used in IEC Analysis of Adalimumab









Time (min)
% MPA
% MPB












0.05
94
6


20
84
16


22
0
100


26
0
100


28
94
6


34
94
6


35
94
6










14.5: Ion Exchange Data


Table 25 shows the ion exchange chromatographic data for the Adalimumab reference standard, commercial formulation (150 mg/ml) and post-DF/UF low-ionic solution before storage. Table 26 shows data for the reference standard compared to the low-ionic solution after 22.5 months of storage at 2-8° C.









TABLE 25







IEC Analysis Data of Adalimumab Reference Standard,


DS/Commercial Formulation and After DF/UF (in water)













% Acidic
% Acidic
%
%
%


Sample Name
Region 1
Region 2
0 Lys
1 Lys
2 Lys





Adalimumab Ref. Std.
2.69
11.66
60.77
19.42
5.40


Adalimumab DS 150
2.51
11.38
62.05
19.14
4.83


mg/ml







Adalimumab
2.26
11.81
61.97
18.51
4.73


diafiltered against







water, 177 mg/ml
















TABLE 26







IEC Analysis Data Comparing Reference Standard to


DF/UF Sample from Long-Term Refrigerated Storage













% Acidic
% Acidic
%
%
%


Sample Name
Region 1
Region 2
0 Lys
1 Lys
2 Lys





Adalimumab Ref. Std.
2.1
10.9
63.8
18.4
4.6


Adalimumab DF/UF
2.7
13.4
62
16.7
4.1


against water, 177







mg/mL, 22.5 months







at 2-8° C.










14.6: Ion Exchange Analysis Conclusions


For the T0 samples, data show no significant difference in the percentage of acidic region 1, 2, 0 Lys, 1 Lys, or 2 Lys (i.e., charge heterogeneity) between reference standard Adalimumab, commercial formulation Adalimumab (used as DS to formulate Adalimumab into water by DF/UF), and Adalimumab diafiltered against water and concentrated to 177 mg/ml (Table 25).


Also, after 22.5 months storage of the 177 mg/mL Adalimumab sample in water, only slight differences in 0 Lys, 1 Lys and 2 Lys fractions can be seen when compared to the Adalimumab reference standard. In summary, no significant chemical instability tendencies are observed when Adalimumab is formulated into water by DF/UF processing and stored for 22.5 months at 2-8° C. at a concentration of 177 mg/mL.


Example 15: Freeze/Thaw Stability of Low-Ionic 1D4.7 Solution

1D4.7 protein (an immunoglobulin G1) anti-IL 12/anti-IL 23 was formulated in water by dialysis (using slide-a-lyzer cassettes, used according to operating instructions of the manufacturer, Pierce, Rockford, Ill.) was demonstrated to be stable during repeated freeze/thaw (f/t) processing (−80° C./25° C. water bath) at 2 mg/mL concentration, pH 6. Data were compared with routine formulations (2 mg/mL, pH 6), and it was found that the stability of 1D4.7 formulated in water exceeded the stability of 1D4.7 formulated in routinely screened buffer systems (e.g. 20 mM histidine, 20 mM glycine, 10 mM phosphate, 10 mM citrate) and even exceeded the stability of 1D4.7 formulations based on universal buffer (10 mM phosphate, 10 mM citrate) with a variety of excipients that are commonly used in protein formulation, e.g. 10 mg/mL mannitol, 10 mg/mL sorbitol, 10 mg/mL sucrose, 0.01% polysorbate 80, 20 mM NaCl.


SEC, DLS and particle counting was performed to monitor protein stability, and particle counting was performed by using a particle counting system with a 1-200 μm measurement range (e.g. particle counter Model Syringe, Markus Klotz GmbH, Bad Liebenzell, Germany). Experiment details are as follows:

    • 1D4.7 formulated in water compared with formulations listed above
    • 4 freeze/thaw cycles applied
    • 30 mL PETG repository, about 25 mL fill, 2 mg/mL, pH 6
    • sampling at T0, T1 (i.e. after one f/t step), T2, T3, and T4
    • analytics: visual inspection, SEC, DLS, subvisible particle measurement



FIG. 21 shows 1D4.7 stability during repeated f/t cycling (−80° C./25° C.), mirrored by formation of subvisible particles >1 μm. 1D4.7 was formulated in universal buffer (10 mM citrate, 10 mM phosphate) and then the following excipient variations were tested: sorbitol (10 mg/mL), mannitol (10 mg/mL), sucrose (10 mg/mL), NaCl (100 mM), and polysorbate 80 (0.01%). 1D4.7 was also formulated in water (by dialysis) with no excipients added at all. Water for injection was also subjected to f/t cycling and subvisible particle testing to evaluate a potential impact of material handling, f/t, and sample pull on particle load.


The stability of 1D4.7 formulated in water upon f/t exceeded the stability of 1D4.7 solutions formulated with excipients typically used in protein formulations. Mannitol, sucrose, and sorbitol are known to act as lyoprotectant and/or cryoprotectant, and polysorbate 80 is a non-ionic excipient prevalently known to increase physical stability of proteins upon exposure to hydrophobic-hydrophilic interfaces such as air-water and ice-water, respectively. Thus, 1D4.7 solutions formulated in water appeared to be stable when analyzed with other methodologies applied (e.g. SEC, visual inspection, etc.).


Example 16: Freeze/Thaw Stability of Low-Ionic 13C5.5 Antibody Solution

13C5.5 anti IL-13 protein formulated in water was demonstrated to be stable during repeated freeze/thaw processing (−80° C./25° C. water bath) at 2 mg/mL concentration, pH 6. Data were compared with routine formulations (2 mg/mL, pH 6), and it was found that the stability of 13C5.5 formulated in water exceeded the stability of 13C5.5 formulated in routinely screened buffer systems (e.g. 20 mM histidine, 20 mM glycine, 10 mM phosphate, 10 mM citrate) and even exceeded the stability of 13C5.5 formulations based on universal buffer (10 mM phosphate, 10 mM citrate) with a variety of excipients that are commonly used in protein formulation (e.g. 10 mg/mL mannitol, 10 mg/mL sorbitol, 10 mg/mL sucrose, 0.01% polysorbate 80, 20 mM NaCl, 200 mM NaCl).


Sample preparation, experiment processing, sample pull and sample analysis was performed in the same way as outlined in Example 15 for 1D4.7.

    • 13C5.5 formulated in water compared with formulations listed above
    • 4 freeze/thaw cycles applied
    • 30 mL PETG repository
    • 2 mg/mL, pH 6
    • sampling at T0, T1, T2, T3, and T4
    • analytics: visual inspection, SEC, DLS, subvisible particle measurement



FIG. 22 shows 13C5.5 stability during repeated f/t cycling (−80° C./25° C.), mirrored by formation of subvisible particles >10 μm. 13C5.5 was formulated in either 10 mM phosphate buffer, 10 mM citrate buffer, 20 mM glycine buffer, and 20 mM histidine buffer. 13C5.5 was also formulated in water (by dialysis) with no excipients added at all. Water for injection was also subjected to f/t cycling and subvisible particle testing to evaluate a potential impact of material handling, f/t, and sample pull on particle load (blank).


The stability of 13C5.5 formulated in water upon f/t exceeded the stability of 13C5.5 solutions formulated in buffers typically used in protein formulations. No instabilities of 13C5.5 solutions formulated in water have been observed with other analytical methodologies applied (e.g. SEC, visual inspection, etc.)



FIG. 23 shows 13C5.5 stability during repeated f/t cycling (−80° C./25° C.), mirrored by formation of subvisible particles >1 μm. 13C5.5 was formulated in universal buffer (10 mM citrate, 10 mM phosphate) and then the following excipient variantions were tested: sorbitol (10 mg/mL), mannitol (10 mg/mL), sucrose (10 mg/mL), NaCl (200 mM), NaCl (20 mM) and polysorbate 80 (0.01%). 13C5.5 was also formulated in water (by dialysis) with no excipients added at all for comparison (pure water). Water for injection was also subjected to f/t cycling and subvisible particle testing to evaluate a potential impact of material handling, f/t, and sample pull on particle load.


The stability of 13C5.5 formulated in water upon f/t exceeded the stability of 13C5.5 solutions formulated with excipients typically used in protein formulations. Mannitol, sucrose, and sorbitol are known to act as lyoprotectant and/or cryoprotectant, and polysorbate 80 is a non-ionic excipient prevalently known to increase physical stability of proteins upon exposure to hydrophobic-hydrophilic interfaces such as air-water and ice-water, respectively.


No instabilities of 13C5.5 solutions formulated in water have been observed with other analytical methodologies applied, (e.g. SEC, visual inspection, etc.).


DLS analysis of 13C5.5 solutions after f/t procedures was performed as described above. An 13C5.5 solution with 0.01% Tween-80 contained significant high molecular weight (HMW) aggregate forms after only 1 f/t step, whereas 13C5.5 in water contained no HMW aggregate forms, even after 3 f/t steps.


Example 17: Impact of Solution pH on Adalimumab in WFI

The following experiments were performed to determine the impact of solution pH on physico-chemical characteristics of highly concentrated Adalimumab formulated in WFI. The following concentrations were tested: 2 mg/mL, 50 mg/mL, 100 mg/mL, 150 mg/mL, 200 mg/mL, and 250 mg/mL.


Materials






    • Adalimumab Drug Substance (DS), commercial material

    • 25° C. water bath (circulating) used for thawing

    • Diafiltration equipment: Sartorius Sartocon Slice, membrane: PES 50 kD, 1000 cm2

    • Diafiltration equipment: Millipore Labscale™ TFF System, membrane: PLCTK 30 kD, regenerated Cellulose, size: 50 cm2

    • Eppendorf Centrifuge 5810 R

    • Amicon Ultra-15 repositories for centrifugation, Ultracel-30k, Regenerated Cellulose 30,000 MWCO

    • Millex GV 0.22 μm, Millipore for sterile filtration of samples

    • Sample repositories (Eppendorf sample repository 1.5 mL, Roth cryovials 5 mL, PETG bottle 125 mL)





Analytics:

    • pH measurement using Biothrode
    • Density measurement
    • Osmolality measurement
    • UV/VIS spectrophotometer for protein concentration measurement
    • Photon Correlation Spectroscopy (PCS)
    • Viscosity measurement
    • Turbidity measurement
    • Size Exclusion Chromatography (SEC)
    • Fourier transform mid infrared spectroscopy (FT-M-IR)


      17.1 Overview of Preparation for DF/UF of Adalimumab Commercial Formulation


The Adalimumab DS solution (120 mg/mL) was divided into 7 volume portions which were adjusted to pH3, pH4, pH5, pH6, pH7, pH8, pH9 with 0.25N NaOH and 0.25N HCl, respectively. Then the samples were diluted with Adalimumab buffer of the respective pH to 100 mg/mL. The solutions revealed a slight cloudyness that disappeared after sterile filtration (0.22 μm, PVDF sterile filter). After dilution, the pH value were monitored again (see Table 27 below).


The following samples of the 100 mg/mL solutions were pulled from each solution:

    • 4 mL for turbidity and subsequent zetapotential measurement
    • 1 mL for viscosity measurement (using dropping-ball viscometer)
    • 0.15 mL for osmolality measurement
    • 2 mL for density measurement
    • 0.15 mL for PCS (sample viscosity taken into account for measurements)
    • 1 mL for FT-M-IR
    • 2 mL for viscosity and static light scattering measurements


The samples for zetapotential, viscosity and static light scattering measurements were frozen (−80° C.). The remaining volumes of pH 4, pH 5, pH 6, pH 7, and pH 8 solutions were subjected to continuous mode diafiltration using water for injection as exchange medium. The samples were first frozen at −80° C. Before DF/UF, the samples were thawed at 25° C. in a Julabo water bath.


17.2 DF/UF and Concentration Procedures


Adalimumab solutions in commercial formulation, with concentrations of 100 mg/ml, with pH levels of 4, 5, 6, 7 and 8, were subject to DF/UF processing and further subject to concentration process with UF in a centrifuge. This section describes the processing of the pH 6 Adalimumab solution as an example. Processing for the other solutions was done in a similar manner.


The Adalimumab solution (100 mg/mL, pH 6) was thawed in a water bath at 25° C. and then homogenized. Then, the solution was subjected to diafiltration using water for injection as exchange medium with TFF equipment M.P. 33.4 by applying the following parameters:

    • stirrer: speed 2
    • pump: speed 1
    • pressure up-stream/inlet: 2-2.4 bar
    • pressure down-stream/outlet: 0.6-0.8 bar
    • membrane: regenerated Cellulose, cut off 30 kD
    • continuous mode DF/UF
    • about 6-fold volume exchange applied during DF/UF operation


After applying 6-volume exchange steps, the concentration of Adalimumab was determined by means of OD280, photometer M.P. 9.7. The osmolality of permeate and retentate was checked.


concentration: 125.1 mg/mL


osmolality permeaet: 57 mOsmol/kg


osmolality retentate: 12 mOsmol/kg


The Adalimumab solution in water after DF was diluted with water for injection to 100 mg/mL and sterile filtered. The following samples were pulled from 100 mg/mL solution after the DF/UF process:

    • 4 mL for turbidity and subsequent zetapotential measurement
    • 1 mL for viscosity measurement
    • 0.15 mL for osmolality measurement
    • 2 mL for density measurement
    • 0.15 mL for PCS (viscosity taken into account during measurement)
    • 0.15 mL for SEC
    • pH—measurement
    • 1 mL for FT-M-IR
    • 2 mL for viscosity and static light scattering measurements


A portion of the 100 mg/mL Adalimumab solution was diluted with water for injection to create 50 mg/mL and 2 mg/mL solutions. The following samples were pulled from both solutions:

    • 4 mL for turbidity and subsequent zetapotential measurement
    • 2 mL for viscosity measurement
    • 0.15 mL for osmolality measurement
    • 2 mL for density measurement
    • 0.15 mL for PCS (viscosity taken into account)
    • pH-measurement


Adalimumab solutions (pH 6, 100 mg/mL) in water were subjected to concentration experiments using centrifugation. Centrifugation was performed with Eppendorf Centrifuge (5810R M.P. 33.57). Each centrifugation step was applied for 15 min. at 4000 rpm. After that, homogenization of sample solution in the centrifuge concentration device was performed by gentle upside-down rotation in order to homogenized the solution and thereby to avoid gel formation in areas immediately adjacent to the membrane. Temperature during concentration was 15° C. The centrifugation was performed to about 250 mg/mL. The concentration was determined by means of measuring OD280, photometer M.P. 9.7. The Adalimumab solutions were then diluted to concentrations of 250 mg/mL, 200 mg/mL and 150 mg/mL.


The following samples were pulled after the concentration procedure and after each individual step of dilution. Sample volumes pulled from 250 mg/mL and 150 mg/mL solutions were:

    • 2 mL for viscosity-measurement
    • 0.15 mL for PCS (viscosity taken into account)
    • 0.15 mL for osmolality measurement
    • 0.15 mL for SEC
    • pH-measurement


      Sample volumes pulled from the 200 mg/mL solution were:
    • 4 mL for turbidity and subsequent zetapotential measurement
    • 1 mL for viscosity measurement
    • 0.15 mL for osmolality measurement
    • 2 mL for density measurement
    • 0.15 mL for PCS (viscosity taken into account)
    • 0.15 mL for SEC
    • pH-measurement
    • 2 mL for analytical work to be performed at ABC (viscosity and static light scattering measurements)


The concentration processing of Adalimumab solution in water was halted at approximately 250 mg/mL at each pH value because the viscosity of Adalimumab solution in water at higher concentrations, and especially at pH values close to the pI (about pH 8.5 for Adalimumab), increased dramatically (viscosities approaching gel formation).


17.3 Visual Inspection of Adalimumab Solutions


After DF/UF and concentration to 250 mg/mL, the Adalimumab solutions in water at various pH appeared less opalescent than the Adalimumab solution in buffer (commercial formulation). All of the Adalimumab solutions in water appeared as clear solutions at each pH value. None of the Adalimumab solutions revealed opalescence after dilution. Overall, during concentration and dilution procedures, no precipitation was observed in Adalimumab solutions in water.


17.4 Viscosity


The viscosity measurements were performed taking into account the density of pH 5 Adalimumab solutions at each of the respective concentrations. A dropping-ball viscometer was used. Viscosities higher than 200 mPa*s were measured using capillary viscometer.



FIG. 24 provides an overview of viscosity data of Adalimumab solutions in water with pH ranging from 4 to 8, at various concentrations (2 mg/mL to 250 mg/mL, in 50 mg/mL concentration steps). There is a clear correlation between solution pH, concentration and viscosity. The viscosity increases with increases of protein concentration, independent of the solution pH. At solution pH values close to the pI of Adalimumab (i.e. pH 7 and pH 8), increases in solution viscosity were most pronounced, especially at higher protein concentrations (i.e. 200 mg/mL, 250 mg/mL).


17.5 Turbidity


As seen in FIG. 25, the same trend was found for turbidity data, (i.e., the turbidity increased with increasing concentration and with increasing pH). All samples were sterile filtered (0.22 μm) before turbidity measurement.


17.6 Hydrodynamic Diameter (PCS)


The PCS measurements were performed taking into account the viscosity for each sample, at each concentration and at each pH value. Solutions at 200 mg/mL and 250 mg/mL were measured but were outside the testing parameters of the Zetasizer nano series (Malvern Instruments) equipment, and consequently the data from these measurements was not analyzed.


The hydrodynamic diameter (Dh) was found to be notably decreased when Adalimumab was formulated in water (Dh about 2 nm at 50 mg/mL, pH 5) in comparison to Adalimumab formulated into commercial formulation (Dh about 7 nm). FIG. 26 illustrates the PCS data (also found in Table 39). Corresponding data tables are shown below in part 17.11.


As shown in FIG. 26, for solutions at pH values of 4, 5 and 6, the Dh of Adalimumab monomer decreased constantly with increased protein concentration. In contrast, solutions with pH values closer to the pI of Adalimumab (i.e., at pH 7 and pH8) showed considerable increases in Dh as concentration increased from 2 mg/mL to 50 mg/mL. As concentrations rose beyond 50 mg/mL in pH 7 and 8 solutions, however, Dh decreased. At a concentration of 150 mg/mL, all of the solutions had lower Dh values than the corresponding pH solution at 2 mg/mL. FIG. 27 shows Dh size distributions for pH 5 solutions of various concentrations. FIG. 28 shows Dh size distributions for five Adalimumab solutions formulated in water, each having a 100 mg/mL protein concentration and a different pH value. FIG. 29 shows data similar to data in FIG. 28, except that the five Adalimumab solutions were formulated in buffer.


17.7 pH-Measurement


Measurements of solution pH were performed at 100 mg/mL before and after DF/UF using water (i.e., performed on Adalimumab formulated in buffer and in water, respectively). Table 27 shows the results. The pH values stay constant at pH 5, pH 6 and pH 7 before and after DF/UF. The solution pH does not change because of a medium change. The pH value at pH 4 slightly increases and at pH 8 slightly decreases after DF/UF using water.









TABLE 27







pH values before and after DF/UF with water















pH 4
pH 5
pH 6
pH 7
pH 8


















Adalimumab 100
4.00
4.99
6.00
7.03
8.00



mg/mL in buffer








Adalimumab 100
4.29
4.98
5.98
7.02
7.67



mg/mL in water











17.8 Osmolality Measurements


During DF/UF of the pH 5 solution samples, solution osmolality was measured after each volume exchange step (i.e., after 100 mL permeate, 200 mL permeate, etc.) to check whether a 5-fold volume exchange is sufficient to reduce osmolality to values below 15 mOsmol/kg. Table 28 shows the results.









TABLE 28







Osmolality change during DF/UF using water, pH 5 solution









Volume




exchange step
Retentate
Permeate


in mL
mOsmol/kg
mOsmol/kg












100
96
166


200
28
115


300
29
89


400
12
67


500
15
49









At pH 4, pH 6, pH 7 and pH 8, the osmolality was measured at the end of the DF/UF process only. Table 29 shows the osmolality results (in mOsmol/kg units) for each pH.









TABLE 29







Osmolality at various pH values, before and after DF/UF with water















pH 4
pH 5
pH 6
pH 7
pH 8


















Adalimumab 100
287
298
297
286
279



mg/mL in buffer








Adalimumab 100
40
13
11
5
5



mg/mL in water















The osmolality measurements were performed with a freezing point viscometer.


17.9 Fragmentation (SEC)


The SEC data show a relative pronounced fragmentation of the protein in ph 4 solutions over the whole concentration range (100-250 mg/mL), while there almost no fragmentation detected at pH ranging from 5 to 8 over the same concentration range. Consequently, the monomer content of pH 4 solutions decreased accordingly (FIG. 30). Aggregate values were found to increase with increasing pH values (from pH 4 to pH 8), independent of the concentration (FIG. 31).


17.10 Conclusions


This experiment was designed to examine the impact of solution pH and protein concentration on viscosity and Dh (hydrodynamic diameter) of Adalimumab solutions formulated in water by DF/UF processing. Such solutions are referred to as low-ionic solutions. A pH range of 4-8 was evaluated, and protein concentrations tested were in a range between 2 and 250 mg/mL.


With regard to viscosity (Section 17.4), it was found that low-ionic Adalimumab solutions have the same characteristics as Adalimumab solutions formulated in the presence of ions (i.e. ionic excipients such as organic buffer components or salts):

    • The higher the protein concentration, the higher solution viscosity. This concentration-viscosity correlation was more pronounced for solutions with pH values close to the Adalimumab pI (i.e., pH 7 and pH 8). Conversely, for solutions at a constant concentration, viscosity correlated with the closeness of the solution's pH value to the pI of Adalimumab.


With regard to DLS data (Section 17.6), the following conclusions can be drawn:

    • Adalimumab Dh values determined by DLS of low ionic Adalimumab solutions were found to be lower than Dh values measured in Adalimumab commercial formulations, especially at very low solution pH.
    • The lower the solution pH, the lower Dh values determined by DLS.
    • The higher the protein concentration, the lower the Dh values in low-ionic Adalimumab solutions of a given pH.


The explanation for this behavior is that the ionic strength (i.e. the presence of ions and ionizable excipients) in protein solutions is crucial for the extent of protein-protein interactions. Especially at lower solution pH, charge-charge repulsions are more pronounced in low ionic Adalimumab solutions. When a protein is formulated in water by using water as exchange medium in DF/UF processing, the amount of ionizable counter ions present that can compose both the Helmholtz layer and the Gouy-Chapman layer is notably reduced. Consequently, intermolecular charge-charge interactions (due to the charges of amino acid residues present at the protein's surface) may be more pronounced than in an environment where ionizable counter ions (e.g. ionizable excipients) are abundant, and charge-charge repulsion between protein monomers (leading to molecule motion in case of charge-charge repulsion) and random Brownian motion contribute to the mobility/motion of the protein molecule measured by DLS. In DLS experiments, greater molecule mobilities are translated into greater molecular diffusion coefficients, which usually are assigned to molecules with smaller hydrodynamic sizes via using the Stokes-Einstein equation. This can explain why the hydrodynamic diameter of proteins is reduced in low-ionic formulations.


Charge-charge interactions between antibody molecules can be repulsive (at lower solution pH) and attractive (at higher solution pH close to the protein's pI).


17.11 Data Tables









TABLE 30







Adalimumab 100 mg/mL in buffer before DF versus water















pH3
pH4
pH5
pH6
pH7
pH8
pH9

















turbity (NTU)
9.9
15.4
28.5
36.3
45.0
48.4
46.5


viscosity (mPa * s)
2.5197
2.7935
3.2062
3.1512
3.5116
3.5494
3.5844


viscosity (mm2/s)
2.4366
2.6991
3.0969
3.0444
3.3893
3.4261
3.4589


density (g/cm3)
1.0341
1.0350
1.0353
1.0351
1.0361
1.0360
1.0363


osmolality (mOsmol/kg)
293
287
298
297
286
279
285


Z-Ave d (nm) PCS
4.3
4.3
6.0
7.3
7.7
8.0
7.8


pH

4.00
4.99
6.00
7.03
8.00
9.03
















TABLE 31







Adalimumab after DF versus water, before


concentration, diluted with water to











pH 4
pH 4
pH 4



2 mg/mL
50 mg/mL
100.5 mg/mL





turbity (NTU)
0.296
1.46
3.30


viscosity (mPa * s)
0.9653
1.4471
2.2411


viscosity (mm2/s)
0.9665
1.4298
2.1834


density (g/cm3)





osmolality (mOsmol/kg)


40


Z-Ave d (nm) PCS
3.37
2.24
1.81


pH


4.29










Adalimumab after concentration and dilution with water to











pH 4
pH 4
pH 4



150.5 mg/mL
219.0 mg/mL
251.8 mg/mL





turbity (NTU)

3.56



viscosity (mPa * s)
4.0283
13.304
48.642


viscosity (mm2/s)
3.8712
12.614
45.567


density (g/cm3)





osmolality (mOsmol/kg)
64
96
141


Z-Ave d (nm) PCS
1.32
0.458
0.162


pH
4.32

4.54










Adalimumab after DF versus water, before concentration,


diluted with water to











pH 5
pH 5
pH 5



2 mg/mL
50 mg/mL
97.5 mg/mL





turbity (NTU)
0.02
1.66
3.54


viscosity (mPa * s)
1.0563
1.6664
2.8661


viscosity (mm2/s)
1.0576
1.6465
2.7924


density (g/cm3)
0.9988
1.0121
1.0264


osmolality (mOsmol/kg)


13


Z-Ave d (nm) PCS
157
32.4
1.3


pH
4.55
4.83
4.98










Adalimumab after concentration and dilution with water to











pH 5
pH 5
pH 5



150.7 mg/mL
200.2 mg/mL
253.0 mg/mL





turbity (NTU)

7.24



viscosity (mPa * s)
7.0866
19.539
79.272


viscosity (mm2/s)
6.8102
18.525
74.26


density (g/cm3)
1.0406
1.0547
1.0675


osmolality (mOsmol/kg)
78
80
96


Z-Ave d (nm) PCS
0.727
0.335
0.255


pH
5.03
5.05
5.08
















TABLE 32







Adalimumab after DF versus water, before concentration,


diluted with water to











pH 6
pH 6
pH 6



2 mg/mL
50 mg/mL
100 mg/mL













turbity (NTU)
0.458
2.24
2.95


viscosity (mPa * s)
1.0696
1.8003
3.1147


viscosity (mm2/s)
1.0708
1.7789
3.0385


density (g/cm3)
0.9989
1.012
1.0251


osmolality (mOsmol/kg)
3 + 11 = 14:2 = 7
27
11


Z-Ave d (nm) PCS
30.8
2.78
2.48


pH
5.72
5.95
5.98
















TABLE 33







Adalimumab after concentration and diluted with water to











pH 6
pH 6
pH 6



146.6 mg/mL
201.8 mg/mL
248.5 mg/mL













turbity (NTU)

9.29



viscosity (mPa * s)
9.0193
32.352
126.06


viscosity (mm2/s)
8.6775
30.709
118.07


density (g/cm3)
1.0394
1.0535
1.0677


osmolality (mOsmol/kg)
37
58
95


Z-Ave d (nm) PCS
0.989
0.355
0.108


pH
5.92
6.05
6.03
















TABLE 34







Adalimumab after DF versus water, before concentration,


diluted with water to











pH 7
pH 7
pH 7



2 mg/mL
50 mg/mL
103.2 mg/mL













turbity (NTU)
0.1
7.13
14.9


viscosity (mPa * s)
1.1252
1.6898
4.2257


viscosity (mm2/s)
1.1268
1.6688
4.1146


density (g/cm3)
0.9986
1.0126
1.027


osmolality (mOsmol/kg)
0
2
5


Z-Ave d (nm) PCS
3.31
4.16
2.89


pH
6.63
6.93
7.02
















TABLE 35







Adalimumab after concentration and diluted with water to











pH 7
pH 7
pH 7



143.0 mg/mL
203.4 mg/mL
251.7 mg/mL













turbity (NTU)

19.3



viscosity (mPa * s)
14.024
74.987
343.881


viscosity (mm2/s)
13.492
70.928
321.144


density (g/cm3)

1.0571
1.0708


osmolality (mOsmol/kg)
65
106
160


Z-Ave d (nm) PCS
1.27
0.346
0.0876


pH
6.9
7.01
7.2
















TABLE 36







Adalimumab after DF versus water, before


concentration, diluted with water to











pH 8
pH 8
pH 8



2 mg/mL
50 mg/mL
96.1 mg/mL













turbity (NTU)
0.41
12.10
28.300


viscosity (mPa * s)
1.261
1.8444
4.3486


viscosity (mm2/s)
1.2625
1.8224
4.2368


density (g/cm3)





osmolality (mOsmol/kg)


5


Z-Ave d (nm) PCS
5.59
5.62
4.28


pH


7.67
















TABLE 37







Adalimumab after concentration and dilution with water to











pH 8
pH 8
pH 8



148.5 mg/mL
200.6 mg/mL
230.7 mg/mL













turbity (NTU)

32.5



viscosity (mPa * s)
20.102
85.5
233.14


viscosity (mm2/s)
19.318
81.066
218.04


density (g/cm3)





osmolality (mOsmol/kg)





Z-Ave d (nm) PCS
1.42
0.398
0.168


pH

7.6
















TABLE 38







PCS data: Adalmumab in buffer
















Z-Ave

Pk1
Pk1
Pk2
Pk2
Pk3
Pk3



d · nm
PDI
d · nm
Area %
d · nm
Area %
d · nm
Area %


















pH 3 100 mg/mL
4.23
0.283
4.43
86.4
54.1
13.6
0
0


pH 4 100 mg/mL
4.3
0.101
4.81
100
0
0
0
0


pH 5 100 mg/mL
6.01
0.065
6.5
100
0
0
0
0


pH 6 100 mg/mL
7.25
0.063
7.82
100
0
0
0
0


pH 7 100 mg/mL
7.64
0.094
8.53
100
0
0
0
0


pH 8 100 mg/mL
7.95
0.099
8.88
100
0
0
0
0


pH 9 100 mg/mL
7.7
0.133
8.98
100
0
0
0
0
















TABLE 39







PCS data: Adalmumab in water
















Z-Ave

Pk1
Pk1
Pk2
Pk2
Pk3
Pk3



d · nm
PDI
d · nm
Area %
d · nm
Area %
d · nm
Area %


















pH 4 2 mg/ml
3.37
0.219
3.39
88.8
73.3
11.2
0
0


pH 4 50 mg/ml
2.24
0.194
2.65
97.7
3300
2.3
0
0


pH 4 100.5 mg/ml
1.81
0.172
2.02
97.4
3390
2.6
0
0


pH 4 150.5 mg/ml
1.32
0.181
1.64
100
0
0
0
0


pH 4 219.0 mg/ml
0.458
0.217
4070
62
0.621
38
0
0


pH 4 251.8 mg/ml
0.162
0.263
0
0
0
0
0
0


pH 5 2 mg/ml
157
0.468
1.88
84.3
181
10.7
17
5


pH 5 50 mg/ml
32.4
0.17
1.6
87.7
15.5
4.8
186
4.7


pH 5 97.4 mg/ml
1.32
0.183
1.52
97.4
3290
2.6
0
0


pH 5 150.7 mg/ml
0.931
0.209
1.36
98.7
3710
1.3
0
0


pH 5 200.2 mg/ml
0.335
0.203
0
0
0
0
0
0


pH 5 253.0 mg/ml
0.107
0.255
0
0
0
0
0
0


pH 6 2 mg/ml
30.8
0.382
2.78
60.9
273
30.2
5070
5


pH 6 50 mg/ml
2.78
0.247
2.68
86.4
1600
7.8
114
5.8


pH 6 100 mg/ml
2.01
0.171
2.48
100
0
0
0
0


pH 6 146.6 mg/ml
0.989
0.219
1.32
96.9
3770
301
0
0


pH 6 201.8 mg/ml
0.355
0.231
0
0
0
0
0
0


pH 6 248.5 mg/ml
0.108
0.301
0
0
0
0
0
0


pH 7 2 mg/ml
3.31
0.211
3.58
93.9
1250
6.1
0
0


pH 7 50 mg/ml
4.16
0.132
4.84
100
0
0
0
0


pH 7 103.2 mg/ml
2.89
0.141
3.39
100
0
0
0
0


pH 7 143.3 mg/ml
1.27
0.212
1.68
100
0
0
0
0


pH 7 203.4 mg/ml
0.346
0.306
0
0
0
0
0
0


pH 7 251.7 mg/ml
0.0876
0.497
0
0
0
0
0
0


pH 8 2 mg/ml
5.59
0.365
3.15
67.4
244
30.2
26.5
2.4


pH 8 50 mg/ml
5.62
0.174
7
100
0
0
0
0


pH 8 96.1 mg/ml
4.28
0.192
4.81
96.9
3640
3.1
0
0


pH 8 148.5 mg/ml
1.43
0.253
1.68
93.9
2910
6.1
0
0


pH 8 200.6 mg/ml
0.398
0.246
4920
100
0
0
0
0


pH 8 230.7 mg/ml
0.168
0.3
0
0
0
0
0
0
















TABLE 40







SEC data












pH
conc. mg/mL
% aggregate
% monomer
% fragmente
Area (mVs)















4
100
0.28
67.95
31.76
45195.348


4
150
0.26
66.07
33.68
44492.803


4
200
0.30
64.59
35.11
52558.050


4
250
0.29
64.40
35.31
48491.299


5
100
1.46
98.44
0.11
48127.249


5
150
1.33
98.56
0.11
43226.397


5
200
1.39
98.50
0.11
43634.282


5
250
1.38
98.52
0.11
41643.062


6
100
2.00
97.90
0.10
44338.373


6
150
2.52
97.37
0.11
41899.182


6
200
2.52
97.37
0.11
43869.183


6
250
2.39
97.50
0.11
34969.456


7
100
2.78
97.12
0.10
46194.824


7
150
4.24
95.65
0.11
47443.014


7
200
3.61
96.29
0.10
41916.220


7
250
3.39
96.50
0.11
38185.208


8
100
3.24
96.65
0.12
42334.491


8
150
3.64
96.18
0.18
40305.890


8
200
3.63
96.25
0.13
40280.342


8
250
3.76
96.05
0.19
32067.297









Example 18: Impact of pH on J695 Viscosity

Viscosity data were generated for J695 after DF/UF processing using water as exchange medium. J695 DS (see Example 1) was diafiltered against water, applying at least 5 DF/UF steps. Viscosity was then determined at various temperatures using a plate-plate viscometer, 100 rpm shear rate, 150 μm gap, 60 mm plate diameter (equipment: Bohlin Geminim viscometer (Malvern Instruments, Southborough, Mass.), temperature range evaluated 8-25° C.).


As seen in FIG. 32, at concentrations of 179 mg/mL and 192 mg/mL, respectively, J695 solution viscosities were below 70 cP at 12° C., below 40 cP at 20° C., and below 30 cP at 25° C.


Example 19: Pharmacokinetics (PK) of an Antibody in Pure Water

The goal of this study was to evaluate potential impact of formulation parameters (i.e. low ionic protein formulation containing water vs conventional protein formulations using ionic excipients such as buffers and salts) on local tolerability and PK after sub-cutaneous (s.c.) dosing with Afelimomab. In addition, systemic toxicity and toxicokinetic data of the formulations was investigated. Protein concentrations used ranged from 50 mg/mL to 200 mg/mL and ionic strengths ranged from 3 mOsm/kg to 300 mOsm/kg.


A single (s.c.) dose feasibility study was carried out with Afelimomab (MAK195F—mouse anti human TNF F(ab′)2 (Abbott Laboratories)) in male Sprague-Dawley rats to assess the local tolerance and toxicity of Afelimomab in rats following s.c. administration of liquid formulations at 50 and 200 mg/kg. Single s.c. doses were followed by an observation/recovery period. Limited blood sampling was carried out to measure circulating Afelimomab levels and assess absorption and half-life. The administered dose volume was 1 mL/kg body weight. The experimental groups included the following:


Experimental Groups


















01
Control (vehicle)



02
50 mg/ml Afelimomab, liquid, standard formulation



07
200 mg/ml Afelimomab, liquid, water formulation



Group A
Observation period 2 days



Group B
Observation period 7 days



Group C
Observation period 14 days











Grouping and Rat Identification (N=1 Per Group)

















Animal number












Group
Group A
Group B
Group C
















01
1
2
3



02
4
5
6



07
19
20
21










The animals were repeatedly observed for clinical signs and mortality on day 1 at 15 min, 1, 3, 5, and 24 hours past administration and at least once daily afterwards. Body weights were measured on the days of dosing (day 1) and necropsy (day 3, 15 or 21, respectively) and twice weekly, if applicable. Blood samples for drug analysis were collected on Day 1 (4 hours past administration), and on Days 2, 3, 5, 8, and 15 as applicable. Prior to necropsy blood was collected and hematological and clinical chemistry parameters were evaluated. Blood smears were prepared of each animal prior to necropsy. At necropsy, macroscopy of body cavities was performed. Organ weight measurement was performed on liver, kidneys, thymus, spleen, and lymph nodes. Preliminary histopathology was performed on the injection site and on liver, kidneys, thymus, spleen, and lymph nodes.


All animals survived the study until scheduled necropsy. The rat administered the water formulation showed crusts in the cervical region from Day 14 to 15. No test item-related effect on body weight was observed. Hematology and clinical chemistry values were variable. No clearly test item-related changes were identified in haematology or clinical chemistry. No test item-related changes were noted in urinalysis. Measurement of organ weights resulted in high variability and no clearly test item-related changes in organ weights.


At gross observation reddening of the subcutis at the area of injection was noted in the rat receiving the water formulation at Day 3. All other changes belonged to the spectrum of spontaneous findings commonly seen in Sprague-Dawley rats of this strain and age.


Microscopic Findings were as follows:

    • No findings in Groups 01, 02
    • Minimal diffuse subcutaneous inflammation in Group 07
    • Focal subcutaneous hemorrhage correlating with reddening on gross pathology in Group 07 (Day 3), thought to be administration related
    • Preliminary immunohistochemistry results of pan-T, suppressor/cytotoxic T cells/natural killer cells, pan-B cells and pan-macrophage markers on the local reactions indicate mainly macrophages and natural killer cells involved in the subcutaneous inflammations/infiltrations. Thus, so far there are no hints for a local immunogenic response to the formulations used.


All other changes belonged to the spectrum of spontaneous findings commonly seen in Sprague-Dawley rats of this strain and age.


Following subcutaneous administration Afelimomab absorption appeared to be fast with maximum serum levels reached 0.2-3 days after injection. The absolute levels of Afelimomab in all samples tested were low. Large variability was observed between the samples, likely because of the limited sampling frequency and the low number of animals used. In most samples, no Afelimomab could be detected in serum after 5-8 days. This drop in serum levels is probably due to the high clearance of the F(ab′)2. The observed T½ for most samples were in the range of 1-2 days in agreement with previous observations. The longer half-life of the low-ionic formulation (7.8 d) may represent a protracted absorption of the sample. Data are presented in Tables 41 and 42.









TABLE 41





Plasma exposure levels of MAK195F




















Time
Concentration (μg/ml)
Average















(day)
Rat 4
Rat 5
Rat 6
(μg/ml)
STD





50 mg/kg liquid
0.167
1.40
1.17
1.38
1.32
0.13


standard
2
0.76
0.97
0.66
0.80
0.16


formulation
3
0.45
0.67
0.47
0.53
0.12



5

LLOQ
LLOQ
LLOQ




8

LLOQ
LLOQ
LLOQ




15


LLOQ
LLOQ















Time
Concentration (μg/ml)
Average















(day)
Rat 19
Rat 20
Rat 21
(μg/ml)
STD





200 mg/kg
0.167
1.27
3.01
3.17
2.48
1.05


water
2
0.17
1.57
1.53
1.09
0.80


formulation
3
LLOQ
1.54
1.56
1.55
0.02



5

0.64
0.66
0.65
0.02



8

0.40
0.37
0.38
0.02



15


0.25
0.25
0.00





LLOQ = below quantitation limit






There were no aggregation state findings for liquids, neither Afelimomab or control substance.


For the low-ionic strength formulation, minimal diffuse s.c. injection site inflammation was seen. Inflammation, either minimal to slight, or slight to moderate, was seen with increased protein concentration (50 mg/mL and 200 mg/mL, respectively). Some local s.c. hemorrhage was seen, correlating with reddening on gross pathology; this was considered to be the consequence of blood vessel puncture during injection. Some subcutaneous reddening at the injection site was observed at Day 3 for the water formulation, but was not considered detrimental. Overall, the formulation was tolerated locally.


In Table 42 below, PK data of conventional liquid formulation vs. the water formulation is presented.









TABLE 42







Pharmacokinetic parameters of MAK 195F after subcutaneous dosing in different formulations.






















Time of









Mean
Last
Last








Residence
Detectable
Detectable


Dose

Half-life
Tmax
Cmax
AUC
Time
Conc.
Conc.


(mg/kg)
Formulation
(day)
(day)
(μg/ml)
(day * μg/ml)
(day)
(day)
(μg/ml)


















 50
liquid
0.5
0.2
1.32
3.3
1.5
3
0.53


200
water
5.9
0.2
2.48
11.8
7.5
15
0.25



formulation

















An increase in duration of detectable serum levels was seen with low ionic formulation (i.e. Afelimomab formulated in water), as seen in Table 42.


In this study, the observed absolute levels of MAK195F in low ionic solution (water) provided a better exposure, longer detectable serum levels and ‘half-life’ than in conventional MAK195F liquid formulation.


Afelimomab half-lives were in the range of 1-2 days in standard formulation in agreement with previous observations for F(ab′)2 molecules. However, a seemingly longer half-life was observed for the low-ionic formulation (7.8 d). Accordingly, the mean residence time of MAK 195F in this formulation appeared to be longer compared to the standard formulation tested.


Example 20: DF/UF of 2.5(E)mg1 (Anti IL-18 Antibody)

Diafiltration/ultrafiltration (continuous mode) of 2.5(E)mg1 bulk solution (59.6 mg/mL) was performed, applying an about 4-fold volume exchange using water for injection (in the following referred to as “water”). The DF/UF operation was controlled by monitoring turbidity, protein concentration (0D280), pH and osmolarity of retentate, and DLS measurements. During DF/UF, permeate osmolarities were also monitored to control the excipient reduction of the 2.5(E)mg1 bulk solutions.


Materials and Methods






    • 2.5(E)mg1 Bulk Drug Substance (methionine, histidine, free of polysorbate 80) (Abbott Bioresearch Center, Worcester, Mass.): 2 PETG bottles with a total of 589.12 g solution, solution concentration 59.6 mg/mL.

    • Ampuwa (water for injection) (Fresenius Medical Care, Waltham, Mass.).

    • Millipore Labscale TFF DF/UF unit including 2×Pellicon XL filter cassettes, Millipore, PLCTK 30 kDa membrane, regenerated cellulose

    • UV/VIS spectrophotometer, Specord 50 using 280 nm wavelength

    • Metrohm pH-meter, type 744 with Biotrode probe No. 57

    • Osmometer: Knauer, K-7400

    • density measurements using equipment of Paar, DMA 4100

    • Laminar air flow box Hereaus

    • turbidity measurments: Hach, 2100AN

    • viscometer: Paar, AMVn

    • scales: Mettler Toledo, AT261 and 33.45

    • filters: Millex AP 20 (fiberglass) and Minisart High Flow Filter (celluloseacetate), 0.20 μm pore size.


      20.1 Experimental Procedures





Thawing of 2.5(E)mg1 DS samples: 2 L PETG bottles containing frozen DS were thawed within 2 hrs using a circulating water bath at 23° C. The thawed DS was clear, slightly opalescent, and free from visible particles.


Concentration of DS by DF/UF: due to the DF/UF unit reservoir volume limit of 530 mL, the 2.5(E)mg1 DS was concentrated to a final volume of 525 mL.


DF/UF using water (buffer exchange): the DS (methionine, histidine, 2.5(E)mg1) was subjected to DF/UF, applying a 4-fold volume exchange. Table 43 gives the amounts of water that were used throughout the experiment and Table 44 provides the experimental parameters.









TABLE 43







DF/UF water volume exchanges











Volume of water used



Volume exchange
(cumulative)







1-fold
 576 mL



2-fold
1152 mL



3-fold
1728 mL



4-fold (end of
2304 mL



experiment)


















TABLE 44







DF/UF procedure parameters










Labscale TFF DF
settings







Pump speed
1.5-2



Pressure of pump
20-30 psi



Stirring speed
~3



Experiment duration
8 hrs










Osmolarity measurement of permeate was performed at about every 200 mL of permeate processed.


After DF/UF against water, the volume of the 2.5(E)mg1 solution was 450 mL and the protein concentration 76.6 mg/mL. This solution, containing 2.5(E)mg1 dissolved essentially in water, was then concentrated.









TABLE 45







DF/UF process parameters for solution concentration










Labscale TFF UF
settings







Pump speed
1.5-2



Pressure of pump
max. 30 psi



Stirring speed
~3



Experiment duration
51 min.



Final weight of
257.83 g



solution:











The concentrated solution (˜130 mg/mL) was subjected to 0.2 μm filtration. The solution was cooled to 2-8° C. and then stored at −80° C.


20.2 Data Collected During DF/UF of 2.5(E)mg1









TABLE 46







In Process control data

















Temperature









solution/









Room


Osmolality
conc


DF
Volume

temperature
turbidity

[mOsmol/
[mg/


steps
[mL]
time
[° C.]
[NTU]
pH
kg]
ml]














2.5(E)mg1
14.5
5.91
150
59.6














 01
0
08:00
19.0/24.1
N/A
5.91
125
65.2


1
575
10:02
24.4/24.4
10.1
5.92
50
70.1


2
1150
11:50
24.3/24.7
6.67
5.94
16
72.8


3
1730
13:50
25.0/24.8
6.55
5.97
6
74.6


ca. 4
2200
15:35
25.8/25.5
10.1
5.97
5
76.7
















TABLE 47







Osmolalty of permeate (fractionated and measured during process)















Temperature








solution/



osmol-




Room
Per-
Permeat
No. Of
ality


Sample

temperature
meate
cumulative
DF/UF
[mOsmol/


no.
time
[° C.]
[ml]
[ml]
steps
kg]
















 03
07:35
N/A
 90
N/A
N/A
125


 1
08:00
19.0/24.1
200
 200
0.3
124


 2
08:50
23.0/24.1
200
 400
0.7
82


 3
09:27
24.0/24.2
200
 600
1.0
53


 4
10:12
24.4/24.4
200
 800
1.4
37


 5
10:50
24.5/24.3
200
1000
1.7
25


 6
11:25
24.6/24.3
200
1200
2.1
16


 7
12:10
24.7/24.3
230
1430
2.5
7


 8
12:55
24.7/24.4
170
1600
2.8
4


 9
13:25
24.8/24.4
200
1800
3.1
2


10
14:15
25.1/24.8
200
2000
3.5
0


11
14:55
25.8/25.5
200
2200
3.8
1
















TABLE 48







Concentration of 2.5(E)mg1 solution after buffer exchange











Solution
Volume in reservoir (i.e.




temperature
retentate)



time
[° C.]
[ml]
pH





15:54
25.9
450
5.94


16:02
26.1
400
5.96


16:07
26.1
375
5.96


16:20
26.2
350
5.96


16:28
26.3
300
5.98


16:35
26.5
275
5.98


16:45
26.5
250
5.99
















TABLE 49







Analytical characterization of concentrated 2.5(E)mg1


solution (before and after 0.2 μm filtration):











lot











parameter
before filtration
after filtration















turbidity
15.4
9.58



[NTU]





osmolality
6
N/A



[mOsmol/kg]





density
1.0346
N/A



[g/ml]





pH
5.99
N/A



Dyn. Viscosity
N/A
7.9998



(25° C.)





[mPas]

















TABLE 50







Dynamic light scattering data (determination of Dh of


monomer and z-average value of Dh = Dh of all


specimen present in solution) during DF/UF









Sample pull














DV



After




1-
DV
DV
DV
concen-
After


DLS data
fold
2-fold
3-fold
4-fold
tration
filtration
















Peak 1








diameter
4.32
3.68
3.54
3.48
2.03
2.13


monomer [nm]
100.0
100.0
100.0
89.6
87.0
100.0


intensity [%]
3.95
3.28
3.20
3.53
2.12
1.89


Z-Average [nm]
0.077
0.106
0.094
0.245
0.287
0.113


Pdl








Peak 2








diameter [nm]
N/A
N/A
N/A
984
411
N/A


intensity [%]



10.4
11.8



Peak 3








diameter [nm]
N/A
N/A
N/A
N/A
42601.2
N/A


intensity [%]
















20.3 Discussion


The experiment demonstrated that 2.5(E)mg1 (buffered in methionine, histidine) can be formulated in essentially water at higher concentration (no solubility limitations observed at 130 mg/mL). After 3 volume exchanges using water osmolality of permeate and retentate were below 10 mOsmol/kg, demonstrating that buffer excipients have been effectively reduced. The opalescence of the 2.5(E)mg1 solution was reduced during DF/UF using water (optimal appearance), mirrored also by reduces turbidity values (nephelometric turbidity units (NTU) of DS starting solution 14.5, after 3-fold volume exchange 6.55, after 4-fold volume exchange 10.5.


As seen with other antibodies, the hydrodynamic diameter as determined by DLS decreased due to excipient reduction (intermolecular charge-charge repulsion adding to random Brownian motion, resulting in higher molecule mobility, translates to lower Dh values calculated). The pH of the 2.5(E)mg1 solution was basically the same before (pH 5.94) and after (pH 5.99) the DF/UF operation.


As shown by DLS monitoring, the 2.5(E)mg1 remained stable during the DF/UF operation. No substantial increase in high molecular weight specimen was detected.


Example 21: Preparation of Adalimumab Formulated in Water and Stability Studies Thereof

The following example describes the stability of a formulation comprising adalimumab originating from processes described in the above examples, i.e., adalimumab was successfully dialyzed into water.


Materials and Methods


3323.6 g Adalimumab solution (71.3 mg/mL) were diafiltered using pure water. After a 7-fold volume exchange with pure water (theoretical excipients reduction, 99.9%), the protein solution was diluted/ultrafiltered to final target concentrations of 220 and 63 mg/mL, respectively. PH, osmolality, viscosity, conductivity, PCS, visual inspection and protein concentration measurements (0D280) were performed to monitor the status of the protein after DF/UF processing.


After DF/UF processing, the protein solutions were sterile filtered (0.22 μm Millipak-60 and Millipak-200 membrane filters) and subsequently filled into BD HyPak SCF™ 1 mL long syringes, equipped with 27.5 G RNS needles and sterile BD HyPak BSCF 4432/50 stoppers. The filling volume was around 0.825 mL per syringe.


After filling the syringes were stored at 2-8° C., 25° C. and 40° C., respectively, and analyzed as indicated in the sample pull scheme depicted below.

    • Adalimumab Drug Substance (Adalimumab extinction coefficient 280 nm: 1.39 mL/mg cm): Drug Substance did not contain polysorbate 80. DS buffer, pH 5.38.
    • Sortorius Sartocon Slice diafiltration system, equipped with Ultrasert PES membrane cassettes (50 kDa and 30 kDa cutoff). The Sartocon Slice system was operated in continuous mode at ambient temperature according to Sartorius Operating Instructions.
    • pH electrodes
    • PerkinElmer UV visible spectrophotometer, Lambda 35, was used for protein concentration measurements (280 nm wavelength). Disposable UV cuvettes, 1.5 mL, semi-micro, Poly(methyl methacrylate) (PMMA), were used for the concentration measurements.
    • Sterilized water for injection Ph. Eur./USP was used as DF/UF medium.
    • A Vogel Osmometer OM815, was used for osmolality measurements (calibrated with 400 mOsmol/kg NaCl calibration solution, Art. No. Y1241, Herbert Knauer GmbH, Berlin, Germany).
    • Anton Paar Microviscosimeter, type AWVn, was used for viscosity assessment of the protein solutions according to Anton Paar Operating Instructions. Viscosity was assessed at 20° C.
    • An InoLab Cond Level 2 WTW device was used for conductivity measurements normalized to 25° C.
    • A Malvern Instruments Zetasizer nano ZS, was used for determination of Z-average values, applying a standard method. Measurements were performed at 25° C., using viscosity data obtained by falling ball viscosimetry (Anton Paar Microviscosimeter, type AWVn, at 25° C.).


      HPLC Methods
    • Adalimumab, SEC analysis: Sephadex 200 column (Pharmacia Cat. No. 175175-01). Mobile phase 20 mM sodium phosphate, 150 mM sodium chloride, pH 7.5, 0.5 mL/min flow rate, ambient temperature, detection UV 214 nm and 280 nm. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 50 (duplicate injection).
    • Adalimumab, IEC analysis: Dionex, Propac WCX-10 column (p/n 054993) along with a corresponding guard column (p/n 054994). Separation conditions: mobile phase A: 10 mM sodium phosphate, pH 7.5; mobile phase B 10 mM Sodium phosphate, 500 mM Sodium chloride, pH 5.5. 1.0 mL/min flow rate, ambient temperature. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 100 μg, duplicate injection.


      Calculation of the Protein Concentration


Calculation formula:






E
=



-
1







g


(

I

I
0


)



=



ɛ
·
c
·
d


c

=

E

ɛ
×
d








ε—absorption coefficient


c—concentration


d—length of cuvette that the light has to pass


E—absorbance


I0—initial light intensity


I—light intensity after passing through sample







ɛ
Adalimumab

=

1.39


mL

mg
×
cm








Sample Pull Scheme


Samples of the prepared solutions are stored at the temperatures listed below and pulled (x) at the indicated time points after study start.


















Temp.
T0
1 m
3 m









 5° C.

x
x



25° C.
x
x
x



40° C.

x
x
























Test parameter
Test method









Visible particles
analogous DAC (EA 4.43)



Subvisible particles
analogous




Ph. Eur./USP EA 4.44



Turbidity
analogous Ph. Eur. (EA 4.42)



Color (visual)
Ph. Eur. (EA 4.50)



pH
Ph. Eur. (EA 4.24)



Size exclusion HPLC
Desribed in the text above



Cation exchange HPLC
Desribed in the text above











DF/UF Processing of Adalimumab


Table 51 describes the adalimumab characteristics after diafiltration.
















TABLE 51






Protein




Con-
PCS



Concen-

Osmo-


duc-
[Z-



tration

lality
Vis-

tivity
aver-



[mg/

[mosmol/
cosity
Visual
[μS/
age/


Sample
mL]
pH
kg]
[cP]
Inspection
cm]
d · nm]






















High
220
5.57
26
27.9
Slightly
1167
0.34


concen-




opalescent,




tration




essentially









free from









visible









particles




Low
63
5.44
5
1.8
Slightly
522
1.85


concen-




opalescent,




tration




essentially









free from









visible









particles










Adalimumab characterization upon storage, including clarity and opalescence, degree of coloration of liquids, SEC, at different temperature degrees is described in Appendix D.


Conclusion


The above example provides a diafiltration/ultrafiltration (DF/UF) experiment where water (sterilized water for injection Ph. Eur./USP) was used as diafiltration medium for the monoclonal antibody Adalimumab.


Adalimumab was subjected to DF/UF processing by using pure water as DF/UF exchange medium and was formulated at pH 5.57 at high concentration (220 mg/mL) and at pH 5.44 at lower concentration (63 mg/mL) without inducing solution haziness, severe opalescence or turbidity formation.


Adalimumab from the DF/UF experiments was stored in SCF syringes at 2-8° C., 25° C. and 40° C. for 3 months. Data obtained points at favorable overall stability of the protein.


In conclusion, processing and formulating proteins using pure water as DF/UF exchange medium is feasible. Assuming an ideal 100% excipient membrane permeability, an approx. 99.9% maximum excipient reduction can be estimated.


Example 22: Stability Studies of Adalimumab Formulated in Water Using Non-Ionic Excipients

The following example describes stability studies of a formulation containing an antibody, i.e., adalimumab, in water with additional non-ionic excipients.


Materials and Methods


Adalimumab material was the same as in example 21 (DF/UF processing). After DF/UF processing, the protein solutions were formulated as denoted in Table 52. Mannitol was chosen as example from the group of sugar alcohols, like mannitol, sorbitol, etc. Sucrose was chosen as example from the group of sugars, like sucrose, trehalose, raffinose, maltose, etc. Polysorbate 80 was chosen as example from the group of non-ionic surfactants, like polysorbate 80, polysorbate 20, pluronic F68, etc. ˜10.7 mL were prepared for any formulation. Osmolality, viscosity and PCS measurements were performed for any formulation after preparation.













TABLE 52





Final protein
Mannitol
Sucrose
Polysorbate 80



concentration
(mg/mL)
(mg/mL)
(% w/w)
Sample Name







 50 mg/mL
50


LI50/01


 50 mg/mL

80

LI50/02


 50 mg/mL
50

0.01
LI50/03


 50 mg/mL

80
0.01
LI50/04


 50 mg/mL
50

0.1
LI50/05


 50 mg/mL

80
0.1
LI50/06


 50 mg/mL


0.01
LI50/07


 50 mg/mL


0.1
LI50/08


200 mg/mL
50


LI200/01


200 mg/mL

80

LI200/02


200 mg/mL
50

0.01
LI200/03


200 mg/mL

80
0.01
LI200/04


200 mg/mL
50

0.1
LI200/05


200 mg/mL

80
0.1
LI200/06


200 mg/mL


0.01
LI200/07


200 mg/mL


0.1
LI200/08











    • Polysorbate 80 stock solution 0.5% and 5% in sterile water for injection:

    • Addition in 1:50 ratio (210 μL to 10.5 mL Adalimumab solution, addition of 210 μL water for injection to samples formulated without surfactant to assure equal protein concentration in all samples)

    • Addition of mannitol/sucrose in solid form (525 mg/840 mg, respectively).





The preparations were sterile filtered (Millex GV, Millipore, 0.22 μm, Ø33 mm, Art. SLGV033RS) and subsequently filled into BD HyPak SCF™ 1 mL long syringes, equipped with 27.5 G RNS needles and sterile BD HyPak BSCF 4432/50 stoppers. The filling volume was around 0.6 mL per syringe.


After filling the syringes were stored at 2-8° C., 25° C. and 40° C., respectively, and analyzed as indicated in the sample pull scheme depicted below.

    • Adalimumab Drug Substance (Adalimumab extinction coefficient 280 nm: 1.39 mL/mg cm): Drug Substance did not contain polysorbate 80. DS buffer, pH 5.38.
    • PH electrodes
    • Sterilized water for injection Ph. Eur./USP was used as DF/UF medium.
    • Mannitol, polysorbate 80, and sucrose, all matching Ph. Eur. quality
    • A Vogel Osmometer OM815, was used for osmolality measurements (calibrated with 400 mOsmol/kg NaCl calibration solution, Art. No. Y1241, Herbert Knauer GmbH, Berlin, Germany).
    • Anton Paar Microviscosimeter, type AWVn, was used for viscosity assessment of the protein solutions according to Anton Paar Operating Instructions. Viscosity was assessed at 20° C.
    • Fluostar Optima, BMG Labtech (absorption measurement at 344 nm in well plates, assessment of turbidity)
    • A Malvern Instruments Zetasizer nano ZS, was used for determination of Z-average values, applying a standard method. Measurements were performed at 25° C., using viscosity data obtained by falling ball viscosimetry (Anton Paar Microviscosimeter, type AWVn, at 25° C.).


      HPLC Methods
    • Adalimumab, SEC analysis: Sephadex 200 column (Pharmacia Cat. No. 175175-01). Mobile phase 20 mM sodium phosphate, 150 mM sodium chloride, pH 7.5, 0.5 mL/min flow rate, ambient temperature, detection UV 214 nm and 280 nm. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 50 μg (duplicate injection).
    • Adalimumab, IEC analysis: Dionex, Propac WCX-10 column along with a corresponding guard column. Separation conditions: mobile phase A: 10 mM sodium phosphate, pH 7.5; mobile phase B 10 mM Sodium phosphate, 500 mM Sodium chloride, pH 5.5. 1.0 mL/min flow rate, ambient temperature. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 100 μg, duplicate injection.


      Sample Pull Scheme


Samples of the prepared solutions were stored at 5° C., 25° C., and 40° C. and pulled at either 1 minute (5° C. and 40° C.) or at T0 and 1 minute (25° C.) after study start. Test parameters were measured according to appropriate methods, e.g., color was determined visually, turbidity was determined at an absorption at 344 nm.


Initial Formulation Characterization


Table 53 described the initial formulation osmolalities and viscosities.












TABLE 53







osmolarity
viscosity


Lot.
comp.
rmosmoll
rmPasl


















LI 50/01
50 mg/mL mannitol
309
1.9796


LI 50/02
80 mg/mL sucrose
272
2.1284


LI 50/03
50 mg/mL mannitol; 0.01% Tween 80
307
1.9843


LI 50/04
80 mg/mL sucrose; 0.01% Tween 80
269
2.1194


LI 50/05
50 mg/mL mannitol; 0.1% Tween 80
307
1.9980


LI 50/06
80 mg/mL sucrose; 0.1% Tween 80
272
2.1235


LI 50/07
0.01% Tween 80
8
1.7335


LI 50/08
0.1% Tween 80
8
1.8162


LI 200/01
50 mg/mL mannitol
396
21.395


LI 200/02
80 mg/mL sucrose
351
21.744


LI 200/03
50 mg/mL mannitol; 0.01% Tween 80
387
21.233


LI 200/04
80 mg/mL sucrose; 0.01% Tween 80
350
21.701


LI 200/05
50 mg/mL mannitol; 0.1% Tween 80
387
21.592


LI 200/06
80 mg/mL sucrose; 0.1% Tween 80
355
21.943


LI 200/07
0.01% Tween 80
27
21.296


LI 200/08
0.1% Tween 80
28
21.889









All formulations of one concentration demonstrated equal viscosities. Those of sucrose containing formulations were slightly higher. The reduced viscosities of the highly concentrated formulations in comparison to the highly concentrated formulation in water (example A, viscosity 27.9 cP) is explained by sample dilution with polysorbate 80 stock solutions or plain water, leading to a final concentration of −215 mg/mL vs. 220 mg/mL in example 21.


Table 54 describes the PCS data determined for each sample.












TABLE 54








PCS



Sample
[Z-average/d · nm]



















LI50/01
2.58



LI50/02
2.22



LI50/03
2.13



LI50/04
2.22



LI50/05
2.25



LI50/06
2.55



LI50/07
2.87



LI50/08
1.94



LI200/01
0.50



LI200/02
0.43



LI200/03
0.36



LI200/04
0.38



LI200/05
0.37



LI200/06
0.41



LI200/07
0.35



LI200/08
0.36











The data provided in Table 54 shows that z-average values do not significantly differ from the values obtained from Adalimumab solutions in non-ionic excipient free systems (63 mg/mL, 1.85 d.nm, 220 mg/mL, 0.34 d.nm, example 21).


Adalimumab Characterization Upon Storage


Appendix E provides data on Adalimumab stability upon storage.


Conductivity of Placebo Solutions


Table 55 describes the influence of non-ionic excipients on the conductivity of the various adalimumab formulations. All placebo solutions were prepared using sterilized water for injection Ph. Eur./USP.












TABLE 55







Polysorbate 80
Conductivity


Mannitol (mg/mL)
Sucrose (mg/mL)
(% w/w)
(μS/cm)










1.1


50


1.2



80

2.2


50

0.01
2.3



80
0.01
1.4


50

0.1
2.6



80
0.1
3.6




0.01
1.2




0.1
2.6










Conclusion


The preparations were stored in SCF syringes at 2-8° C., 25° C. and 40° C. for 1 month. Data obtained from the storage study showed that there was overall stability of the protein in all formulations tested. The stability data was comparable to the stability of samples from example 21. Measurement of the conductivity of non-ionic excipient containing placebo solutions demonstrates a marginal increase of conductivity for some excipients in the range of some μS/cm. PCS measurements demonstrate no significant increase in hydrodynamic diameters in comparison to non-ionic excipient free systems.


In conclusion, processing proteins using pure water as DF/UF exchange medium and formulation with non-ionic excipients is feasible. Adalimumab was also assessed by PCS in a buffer of following composition: 10 mM phosphate buffer, 100 mM sodium chloride, 10 mM citrate buffer, 12 mg/mL mannitol, 0.1% polysorbate 80, pH 5.2. The Adalimumab concentration was 50 mg/mL and 200 mg/mL, respectively. The z-average values were 11.9 d.nm for the 50 mg/mL formulation and 1.01 d.nm for the 200 mg/mL formulation, respectively. Thus, it was clearly demonstrated that hydrodynamic diameters at a given protein concentration are dependent on the ionic strength (clearly higher diameters in salt containing buffers).


Example 23: Preparation of J695 Formulated in Water with Non-Ionic Excipients

The following example describes the preparation of a formulation containing an antibody, i.e., adalimumab, in water with additional non-ionic excipients. The example also describes the stability (as measured for example by SE-HPLC and IEC) of J695 formulated in water with additional non-ionic excipients.


Materials and Methods


2×30 mL J695 solution (˜125 mg/mL) at different pH were dialyzed using pure water applying Slide-A-Lyzer dialysis cassettes. Dialysis of the samples was performed for 3 times against 3 L pure water, respectively (theoretical excipients reduction, 1:1,000,000). The protein solutions were ultrafiltered to final target concentrations of 200 mg/mL, by using Vivaspin 20 concentrators. PH, osmolality, viscosity, conductivity, PCS, visual inspection, HPLC and protein concentration measurements (0D280) were performed to monitor the status of the protein during and after processing.


After processing, the protein solutions were formulated as denoted in the following. Mannitol was chosen as an example to use from the group of sugar alcohols, like mannitol, sorbitol, etc. Sucrose was chosen as an example to use from the group of sugars, like sucrose, trehalose, raffinose, maltose, etc. Polysorbate 80 was chosen as an example to use from the group of non-ionic surfactants, like polysorbate 80, polysorbate 20, pluronic F68, etc. A volume of 0.5 mL was prepared for each of these formulations. PH, osmolality, visual inspection, and HPLC analysis were performed to monitor the status of the protein after sample preparation.









TABLE 56







Description of various J695 formulations











Final protein
Mannitol
Sucrose
Polysorbate 80



concentration
(mg/mL)
(mg/mL)
(% w/w)
Sample Name*





200 mg/mL
50


LI200/01/5


200 mg/mL

80

LI200/02/5


200 mg/mL
50

0.01
LI200/03/5


200 mg/mL

80
0.01
LI200/04/5


200 mg/mL
50

0.1
LI200/05/5


200 mg/mL

80
0.1
LI200/06/5


200 mg/mL


0.01
LI200/07/5


200 mg/mL


0.1
LI200/08/5


200 mg/mL
50


LI200/01/6


200 mg/mL

80

LI200/02/6


200 mg/mL
50

0.01
LI200/03/6


200 mg/mL

80
0.01
LI200/04/6


200 mg/mL
50

0.1
LI200/05/6


200 mg/mL

80
0.1
LI200/06/6


200 mg/mL


0.01
LI200/07/6


200 mg/mL


0.1
LI200/08/6





*The term “/5” or “/6” is added to any sample name to differentiate between samples at pH 5 and 6.








    • Polysorbate 80 stock solution 0.5% and 5% in sterile water for injection:

    • Addition in 1:50 ratio (10 μL to 0.5 mL J695 solution, addition of 10 μL water for injection to samples formulated without surfactant to assure equal protein concentration in all samples)

    • Addition of mannitol/sucrose in solid form (25 mg/40 mg, respectively).

    • J695 Drug Substance (J695 extinction coefficient 280 nm: 1.42 mL/mg cm): Drug Substance did not contain polysorbate 80. DS buffer, pH 6.29.

    • pH electrodes

    • Demineralized and sterile filtered water was used as dialysis medium.

    • Mannitol, polysorbate 80, and sucrose, all matching Ph. Eur. quality

    • A Vogel Osmometer OM815, was used for osmolality measurements (calibrated with 400 mOsmol/kg NaCl calibration solution, Art. No. Y1241, Herbert Knauer GmbH, Berlin, Germany).

    • Anton Paar Microviscosimeter, type AWVn, was used for viscosity assessment of the protein solutions according to Anton Paar Operating Instructions. Viscosity was assessed at 20° C.

    • Fluostar Optima, BMG Labtech (absorption measurement at 344 nm in well plates, assessment of turbidity)

    • Eppendorf Centrifuge 5810 R

    • Slide-A-Lyzer dialysis cassettes, Pierce Biotechnology (Cat No 66830)

    • Vivaspin 20 concentrators, 10 KDa PES membranes (Vivascience, Product number VS2001), used according to standard Operating Instructions

    • PerkinElmer UV visible spectrophotometer, Lambda 35, was used for protein concentration measurements (280 nm wavelength). Disposable UV cuvettes, 1.5 mL, semi-micro, Poly(methyl methacrylate) (PMMA), were used for the concentration measurements.

    • An InoLab Cond Level2 WTW device was used for conductivity measurements normalized to 25° C.

    • A Malvern Instruments Zetasizer nano ZS, was used for determination of Z-average values, applying a standard method. Measurements were performed at 25° C., using viscosity data obtained by falling ball viscosimetry (Anton Paar Microviscosimeter, type AWVn, at 25° C.).


      HPLC Methods

    • J695, SEC analysis: Tosoh Bioscience G3000swxl, 7.8 mm×30 cm, 5 (Cat. No. 08541). Mobile phase 211 mM Na2SO4/92 mM Na2HPO4, pH 7.0. 0.25 mL/min flow rate, ambient temperature, detection UV 214 nm and 280 nm. Each sample was diluted to 2.0 mg/mL with Milli-Q water, sample injection load 20 μg (duplicate injection).

    • J695, IEC analysis: Dionex, Propac WCX-10 column (p/n 054993) along with a corresponding guard column (p/n 054994). Separation conditions: mobile phase A: 10 mM Na2HPO4, pH 6.0; mobile phase B 10 mM Na2HPO4, 500 mM NaCl, pH 6.0. 1.0 mL/min flow rate, 35° C. temperature. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 100 μg.


      Calculation of the Protein Concentration





Calculation formula:






E
=



-
1







g


(

I

I
0


)



=



ɛ
·
c
·
d


c

=

E

ɛ
×
d










    • ε—absorption coefficient

    • c—concentration

    • d—length of cuvette that the light has to pass

    • E—absorbance

    • I0—initial light intensity

    • I—light intensity after passing through sample










ɛ
Adalimumab

=

1.42






mL

mg
×
cm








Processing of J695


J695 in water was characterized prior to the addition of any non-ionic excipients. Table 57 provides details of the J695 characterization during dialysis/ultrafiltration.
















TABLE 57






Protein




Con-




Concen-

Osmolality
Vis-

duc-
PCS



tration

[mosmol/
cosity
Visual
tivity
[Z-average/


Sample
[mg/mL]
pH
kg]
[cP]
Inspection
[μS/cm]
d · nm]







Starting
~125
6.29 (for
N/A
N/A
Slightly
N/A
N/A


material
mg/mL
low pH


opalescent,






samples,


essentially






adjusted to


free from






4.77 with


visible






0.01M


particles






hydro-









chloric









acid)







After
42.5
5.21
7
1.60
Slightly
602
1.5


dialysis,
mg/mL



opalescent,




low pH




essentially









free from









visible









particles




After
56.9
6.30
6
2.11
Slightly
500
2.7


dialysis,
mg/mL



opalescent,




high pH




essentially









free from









visible









particles




After
206
5.40
50
39.35
Slightly
1676
0.21


concen-
mg/mL



opalescent,




tration,




essentially




low pH




free from









visible









particles




After
182
6.46
39
47.76
Slightly




concen-
mg/mL



opalescent,




tration,




essentially
1088
0.21


high




free from




pH




visible









particles












Characterization of Formulations with Non-Ionic Excipients


Following the addition of the various non-ionic excipients to the J695 formulation (see description in Table 56), each formulation was analysed. Results from osmolality and visual inspection, and pH are described below in Table 58.












TABLE 58







Osmolality



Sample
pH
[mosmol/kg]
Visual Inspection


















LI200/01/5
5.39
473
Slightly opalescent, essentially free from visible particles


LI200/02/5
5.38
402
Slightly opalescent, essentially free from visible particles


LI200/03/5
5.37
466
Slightly opalescent, essentially free from visible particles


LI200/04/5
5.37
397
Slightly opalescent, essentially free from visible particles


LI200/05/5
5.37
458
Slightly opalescent, essentially free from visible particles


LI200/06/5
5.37
396
Slightly opalescent, essentially free from visible particles


LI200/07/5
5.36
50
Slightly opalescent, essentially free from visible particles


LI200/08/5
5.36
48
Slightly opalescent, essentially free from visible particles


LI200/01/6
6.43
428
Slightly opalescent, essentially free from visible particles


LI200/02/6
6.42
405
Slightly opalescent, essentially free from visible particles


LI200/03/6
6.43
348
Slightly opalescent, essentially free from visible particles


LI200/04/6
6.43
383
Slightly opalescent, essentially free from visible particles


LI200/05/6
6.42
432
Slightly opalescent, essentially free from visible particles


LI200/06/6
6.42
402
Slightly opalescent, essentially free from visible particles


LI200/07/6
6.43
38
Slightly opalescent, essentially free from visible particles


LI200/08/6
6.43
39
Slightly opalescent, essentially free from visible particles










HPLC Data


Each of the non-ionic excipient containing J695 formulations were also examined using SE-HPLC and IEX. The data from these analyses are provided in Tables 59 and 60 and provide an overview of J695 stability during processing and formulation.









TABLE 59







SE-HPLC results of various J695 formulations











Sum aggregates
Monomer
Sum Fragments


sample name
[%]
[%]
[%]





pH 5
0.608
98.619
0.773


125 mg/mL
0.619
98.598
0.783


starting mat.
0.614
98.608
0.778


pH 6
0.427
98.809
0.764


125 mg/mL
0.392
99.005
0.603


starting mat.
0.409
98.907
0.683


pH 5
0.654
98.604
0.742


42.5 mg/mL
0.677
98.560
0.763


after dialysis
0.666
98.582
0.753


pH 6
0.748
98.541
0.711


56.9 mg/mL
0.739
98.597
0.665


after dialysis
0.743
98.569
0.688


pH 5
0.913
98.416
0.671


206 mg/mL
0.923
98.356
0.721


In Water
0.918
98.386
0.696


pH 5
0.928
98.312
0.760


50 mg/mL mannitol
0.926
98.339
0.736


LI 200/01/5
0.927
98.325
0.748


pH 5
0.925
98.319
0.755


80 mg/mL sucrose
0.929
98.332
0.738


LI 200/02/5
0.927
98.326
0.747


pH 5, 50 mg mannitol
0.942
98.326
0.732


0.01% Tween 80
0.942
98.300
0.758


LI 200/03/5
0.942
98.313
0.745


pH 5, 80 mg sucrose
0.944
98.315
0.741


0.01% Tween 80
0.944
98.339
0.717


LI 200/04/5
0.944
98.327
0.729


pH 5, 50 mg mannitol
0.941
98.348
0.711


0.1% Tween 80
9.967
98.299
0.734


LI 200/05/5
0.954
98.323
0.722


pH 5, 50 mg mannitol
0.944
98.346
0.710


0.1% Tween 80
0.948
98.340
0.712


LI 200/06/5
0.946
98.343
0.711


pH 5, 50 mg mannitol
0.946
98.348
0.706


0.1% Tween 80
0.953
98.328
0.719


LI 200/07/5
0.949
98.338
0.713


pH 5, 50 mg mannitol
0.987
98.313
0.701


0.1% Tween 80
0.994
98.283
0.723


LI 200/08/5
0.991
98.298
0.712


pH 6
1.091
98.169
0.739


182 mg/mL
1.075
98.221
0.703


In Water
1.083
98.195
0.721


pH 6
0.998
98.350
0.652


50 mg/mL mannitol
1.002
98.364
0.634


LI 200/01/6
1.000
98.357
0.643


pH 6
1.028
98.243
0.729


80 mg/mL sucrose
0.983
98.355
0.662


LI 200/02/6
1.006
98.299
0.695


pH 6, 50 mg mannitol
1.005
98.322
0.673


0.01% Tween 80
1.008
98.317
0.676


LI 200/03/6
1.006
98.319
0.674


pH 6, 80 mg sucrose
0.987
98.363
0.649


0.01% Tween 80
0.987
98.321
0.692


LI 200/04/6
0.987
98.342
0.671


pH 6, 50 mg mannitol
0.996
98.326
0.678


0.1% Tween 80
0.996
98.338
0.666


LI 200/05/6
0.996
98.332
0.672


pH 6, 80 mg sucrose
0.998
98.305
0.697


0.1% Tween 80
0.984
98.345
0.671


LI 200/06/6
0.991
98.325
0.684


pH 6
1.000
98.325
0.675


0.01% Tween 80
0.994
98.347
0.659


LI 200/07/6
0.997
98.336
0.667


pH 6
1.003
98.314
0.682


0.01% Tween 80
0.998
98.338
0.664


LI 200/08/6
1.001
98.326
0.673
















TABLE 60







LEX results of various J695 formulations











Sum Acid Peaks
Sum Glutamine
Sum Basic


sample name
[%]
[%]
Peaks [%]





pH 5
4.598

3.303


125 mg/mL
4.599

3.177


starting mat.
4.599
92.162
3.240


pH 6
4.597

3.159


125 mg/mL
4.629

3.156


starting mat.
4.613
92.229
3.158


pH 5
4.706

3.177


42.5 mg/mL
4.725

3.205


after dialysis
4.715
92.094
3.191


pH 6
4.739

3.182


56.9 mg/mL
4.752

3.167


after dialysis
4.746
92.080
3.174


pH 5
4.655

3.167


206 mg/mL
4.676

3.201


In Water
4.666
92.146
3.189


pH 5
4.721

3.321


50 mg/mL mannitol
4.733

3.356


LI 200/01/5
4.727
92.935
3.338


pH 5
4.715

3.299


80 mg/mL sucrose
4.687

3.338


LI 200/02/5
4.701
92.981
3.318


pH 5, 50 mg mannitol
4.767

3.246


0.01% Tween 80
4.736

3.253


LI 200/03/5
4.752
92.999
3.250


pH 5, 80 mg sucrose
4.751

3.257


0.01% Tween 80
4.742

3.229


LI 200/04/5
4.746
92.011
3.243


pH 5, 50 mg mannitol
4.780

3.420


0.1% Tween 80
4.720

3.394


LI 200/05/5
4.750
92.843
3.407


pH 5, 50 mg mannitol
4.756

3.421


0.1% Tween 80
4.894

3.375


LI 200/06/5
4.825
92.777
3.398


pH 5, 50 mg mannitol
4.813

3.425


0.1% Tween 80
4.757

3.413


LI 200/07/5
4.785
92.796
3.419


pH 5, 50 mg mannitol
4.769

3.361


0.1% Tween 80
4.842

3.335


LI 200/08/5
4.806
92.846
3.348


pH 6
4.882

3.452


182 mg/mL
4.886

3.451


In Water
4.884
92.664
3.451


pH 6
4.843

3.456


50 mg/mL mannitol
4.833

3.393


LI 200/01/6
4.838
92.737
3.425


pH 6
4.923

3.407


80 mg/mL sucrose
4.896

3.491


LI 200/02/6
4.909
92.642
3.449


pH 6, 50 mg mannitol
4.864

3.423


0.01% Tween 80
4.899

3.392


LI 200/03/6
4.882
92.711
3.408


pH 6, 80 mg sucrose
4.870

3.320


0.01% Tween 80
4.928

3.369


LI 200/04/6
4.899
92.756
3.345


pH 6, 50 mg mannitol
4.905

3.385


0.1% Tween 80
4.922

3.489


LI 200/05/6
4.914
92.649
3.437


pH 6, 80 mg sucrose
4.973

3.443


0.1% Tween 80
4.962

3.335


LI 200/06/6
4.968
92.644
3.389


pH 6
4.934

3.413


0.01% Tween 80
4.899

3.392


LI 200/07/6
4.916
92.681
3.402


pH 6
4.884

3.410


0.01% Tween 80
4.934

3.366


LI 200/08/6
4.909
92.703
3.388










Conclusion


The above example provides an experiment where water (demineralised and sterile filtered water) was used as dialysis medium for the monoclonal antibody J695.


J695 was subjected to dialysis and concentration processing by using pure water as exchange medium and was formulated at pH 5.40 as well as 6.46 at high concentration (206 and 182 mg/mL, respectively) without inducing solution haziness, severe opalescence or turbidity formation.


J695 from the processing experiment was characterized, and formulated with various non-ionic excipients. Data obtained points at favorable overall stability of the protein in the formulations tested.


In conclusion, processing proteins using pure water as exchange medium and formulation with non-ionic excipients is feasible. Assuming an ideal 100% excipient membrane permeability, an approx. 99.9% maximum excipient reduction can be estimated.


Example 24: Syringeability of Adalimumab Formulated in Water

The formulations from example 21 (63 and 220 mg/mL Adalimumab) were subjected to force measurements upon syringe depletion. 220 mg/mL samples of Adalimumab were diluted to 200 mg/mL, 150 mg/mL and 100 mg/mL, respectively, and were also assessed. A Zwick Z2.5/TN1S was used at a constant feed of 80 mm/min. Finally, viscosity data of the formulations was assessed using an Anton Paar Microviscosimeter, type AWVn, at 20° C. The following data collection suggests that both needle and syringe diameters have a significant effect on the gliding forces upon syringe depletion. Surprisingly, the highly concentrated solution at 220 mg/mL (viscosity 27.9 cP at 20° C.) can be delivered by applying equivalent depletion forces as with the lower concentrated formulation at 63 mg/mL (viscosity 1.8 cP at 20° C.).









TABLE 61







Gliding Force Values obtained for Adalimumab solutions in different packaging systems.












D HyPak SCF ™ 1 mL long




BD HyPak
syringes, equipped with














SCF ™ 1 mL long
25G × ⅝″
26G × ½″
27G × ½″
1 mL Soft-Ject ®



syringes,
needles
needles
needles
Tuberkulin syringes



equipped with
(Sterican)
(Sterican)
(Sterican)
(smaller diameter



27.5G RNS
BD HyPak
BD HyPak
BD HyPak
than BD HyPak


Adaliumab
needles
BSCF
BSCF
BSCF
syringes), equipped


Concentration
BD HyPak BSCF
4432/50
4432/50
4432/50
with 27G × ½ ″


(Viscosity)
4432/50 stoppers
stoppers
stoppers
stoppers
needles (Sterican)





63 mg/mL
 3.9 N






(1.8 cP)







100 mg/mL
3.30 N
1.02 N
1.33 N
1.69 N
1.00 N


(2.9 cP)







150 mg/mL
4.63 N
1.16 N
1.58 N
2.93 N
1.33 N


(7.4 cP)







200 mg/mL
7.25 N
2.16 N
3.24 N
6.25 N
2.55 N


(15.7 cP)







220 mg/mL
14.5 N
2.99 N
3.97 N
9.96 N
3.16 N


(27.9 cP)









The above suggests that even with high concentrations of protein, such formulations are conducive to administration using a syringe, e.g., subcutaneous.


Examples 25-28

Examples 25-28 describe freeze/thaw stability experiments of various antibody formulations containing the antibody formulated in water (referred to in examples 26-28 as low-ionic strength protein formulations). The freeze thaw behavior of a number of antibodies was evaluated by cycling various protein formulations up to 4 times between the frozen state and the liquid state. Freezing was performed by means of a temperature controlled −80° C. freezer, and thawing was performed by means of a 25° C. temperature controlled water bath. About 25 mL of antibody solution each were filled in 30 mL PETG repositories for these experiment series.


Formation of subvisible particles presents a major safety concern in pharmaceutical protein formulations. Subvisible protein particles are thought to have the potential to negatively impact clinical performance to a similar or greater degree than other degradation products, such as soluble aggregates and chemically modified species that are evaluated and quantified as part of product characterization and quality assurance programs (Carpenter, J F et al. Commentary: Overlooking subvisible particles in therapeutic protein products: baps that may compromise product quality. J. Pharm. Sci., 2008). As demonstrated in the examples listed below, a number of antibodies were surprisingly stable—especially with regard to subvisible particle formation—when formulated in the formulation of invention.


Example 25: Freeze/Thaw Stability of Adalimumab Formulated in Water and with Non-Ionic Excipients

The following example describes the stability of an antibody, e.g., adalimumab, in a water formulation and in water formulations in which non-ionic excipients have been added. Aliquots of samples from examples 21 and 22 were subjected to freeze/thaw experiments and analyzed by SE-HPLC. Data was compared to SE-HPLC results derived from freeze/thaw experiments using Adalimumab in a buffer of the following composition: 10 mM phosphate buffer, 100 mM sodium chloride, 10 mM citrate buffer, 12 mg/mL mannitol, 0.1% polysorbate 80, pH 5.2. Adalimumab in this latter buffer was used at 50 mg/mL and 200 mg/mL, respectively. Freeze/thaw cycles were performed in Eppendorf caps, by freezing to -80° C. and storage in the freezer for 8 hours, followed by thawing at room temperature for 1 hour and subsequent sample pull. Each formulation was subjected to 5 cycles, i.e., cycles 0, 1, 2, 3, 4, and 5 described in the tables below.


HPLC Method


Adalimumab, SEC analysis: Sephadex 200 column (Pharmacia Cat. No. 175175-01). Mobile phase 20 mM sodium phosphate, 150 mM sodium chloride, pH 7.5, 0.5 mL/min flow rate, ambient temperature, detection UV 214 nm and 280 nm. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 50 μg (duplicate injection).


Adalimumab Characterization Upon Freeze/Thaw Cycling


Table 62 describes Adalimumab purity during the freeze/thaw experiments. For sample composition, refer to examples 21 and 22.









TABLE 62







Freeze/thaw-Low ionic Adalimumab-50 mg/mL









Fraction monomer [%]















cycle
LI 50/01
LI 50/02
LI 50/03
LI 50/04
LI 50/05
LI 50/06
LI 50/07
LI 50/08





0
99.605
99.629
99.632
99.626
99.619
99.626
99.653
99.655


1
99.743
99.768
99.739
99.759
99.731
99.725
99.686
99.669


2
99.715
99.726
99.571
99.661
99.721
99.721
99.601
99.619


4
99.668
99.689
99.632
99.678
99.523
99.724
99.491
99.542


5
99.627
99.771
99.539
99.772
99.525
99.773
99.357
99.445












Fraction aggregates [%]















cycle
LI 50/01
LI 50/02
LI 50/03
LI 50/04
LI 50/05
LI 50/06
LI 50/07
LI 50/08





0
0.106
0.104
0.119
0.136
0.139
0.145
0.158
0.159


1
0.149
0.134
0.153
0.150
0.149
0.166
0.206
0.201


2
0.192
0.178
0.320
0.242
0.178
0.184
0.319
0.261


4
0.213
0.185
0.239
0.187
0.357
0.170
0.393
0.353


5
0.301
0.151
0.384
0.150
0.398
0.150
0.568
0.484












Fraction fragments [%]















cycle
LI 50/01
LI 50/02
LI 50/03
LI 50/04
LI 50/05
LI 50/06
LI 50/07
LI 50/08





0
0.289
0.267
0.249
0.238
0.242
0.229
0.189
0.186


1
0.108
0.098
0.108
0.091
0.120
0.108
0.107
0.130


2
0.093
0.097
0.108
0.097
0.100
0.094
0.080
0.119


4
0.119
0.126
0.130
0.135
0.120
0.106
0.116
0.105


5
0.072
0.078
0.078
0.078
0.077
0.077
0.075
0.071










Freeze/thaw-Low ionic Adalimumab-200 mg/mL









Fraction monomer [%]















cycle
LI 200/01
LI 200/02
LI 200/03
LI 200/04
LI 200/05
LI 200/06
LI 200/07
LI 200/08





0
99.294
99.296
99.348
99.333
99.313
99.349
99.320
99.290


1
99.286
99.267
99.259
99.256
99.120
99.254
98.999
99.126


2
99.305
99.311
99.249
99.214
99.296
99.288
99.149
99.128


4
99.303
99.272
99.261
99.301
99.296
99.283
99.004
99.061


5
99.320
99.322
99.330
99.331
99.327
99.333
98.939
98.949












Fraction aggregates [%]















cycle
LI 200/01
LI 200/02
LI 200/03
LI 200/04
LI 200/05
LI 200/06
LI 200/07
LI 200/08





0
0.489
0.509
0.491
0.484
0.492
0.488
0.515
0.575


1
0.590
0.574
0.584
0.586
0.680
0.582
0.785
0.718


2
0.604
0.604
0.616
0.630
0.607
0.607
0.731
0.736


4
0.591
0.592
0.612
0.581
0.604
0.596
0.868
0.836


5
0.593
0.586
0.583
0.596
0.597
0.589
0.985
0.981












Fraction fragments [%]















cycle
LI 200/01
LI 200/02
LI 200/03
LI 200/04
LI 200/05
LI 200/06
LI 200/07
LI 200/08





0
0.218
0.196
0.161
0.183
0.195
0.163
0.165
0.135


1
0.124
0.159
0.157
0.159
0.200
0.164
0.216
0.157


2
0.091
0.085
0.135
0.156
0.097
0.105
0.120
0.136


4
0.106
0.136
0.127
0.118
0.100
0.121
0.128
0.103


5
0.087
0.092
0.087
0.073
0.075
0.078
0.076
0.070










Freeze/thaw-Adalimumab Commercial and in water









Fraction monomer [%]












from example A,
from example A,
Standard,
Standard,


cycle
low conc.
high conc.
50 mg/mL
200 mg/mL





0
99.733
99.286
99.374
99.227


1
99.689
99.212
99.375
99.215


2
99.614
99.130
99.370
99.218


4
99.489
99.029
99.361
99.196


5
99.430
98.945
99.362
99.177












Fraction aggregates [%]












from example A,
from example A,
Standard,
Standard,


cycle
low conc.
high conc.
50 mg/mL
200 mg/mL





0
0.186
0.635
0.358
0.502


1
0.226
0.706
0.359
0.516


2
0.304
0.780
0.364
0.513


4
0.428
0.888
0.372
0.535


5
0.485
0.971
0.373
0.553












Fraction fragments [%]












from example A,
from example A,
Standard,
Standard,


cycle
low conc.
high conc.
50 mg/mL
200 mg/mL





0
0.080
0.079
0.268
0.272


1
0.085
0.083
0.266
0.269


2
0.082
0.090
0.266
0.269


4
0.083
0.083
0.267
0.270


5
0.085
0.085
0.265
0.269










Conclusion


The above example provides an experiment where Adalimumab DF/UF processed into water (Sterilized water for injection Ph. Eur./USP) and formulated with various non-ionic excipients was subjected to freeze/thaw cycling. Data obtained (described in Table 62) indicates favorable overall stability of the protein in all formulations tested. All formulations contained above 98.5% monomeric species after 5 freeze/thaw cycles, with minimal amounts of aggregate or fragments as cycles continued.


Example 26: Freeze/Thaw Stability Of Low-Ionic 1D4.7 Solutions

1D4.7 protein (an anti-IL 12/anti-IL 23 IgG1) was formulated in water by dialysis (using slide-a-lyzer cassettes, used according to operating instructions of the manufacturer, Pierce, Rockford, Ill.) and was demonstrated to be stable during repeated freeze/thaw (f/t) processing (−80° C./25° C. water bath) at 2 mg/mL concentration, pH 6. Data were compared with data of various formulations (2 mg/mL protein, pH 6) using buffers and excipients commonly used in parenteral protein formulation development. It was found that the stability of 1D4.7 formulated in water exceeded the stability of 1D4.7 formulated in established buffer systems (e.g. 20 mM histidine, 20 mM glycine, 10 mM phosphate, or 10 mM citrate) and even exceeded the stability of 1D4.7 formulations based on universal buffer (10 mM phosphate, 10 mM citrate) combined with a variety of excipients that are commonly used to stabilize protein formulations, e.g. 10 mg/mL mannitol, 10 mg/mL sorbitol, 10 mg/mL sucrose, 0.01% polysorbate 80, or 20 mM NaCl.


SEC, DLS and particle counting analysis were applied to monitor protein stability, and particle counting was performed using a particle counting system with a 1-200 μm measurement range (particle counter Model Syringe, Markus Klotz GmbH, Bad Liebenzell, Germany). Experiment details are as follows:

    • 1D4.7 formulated in water compared with formulations listed above
    • 4 freeze/thaw cycles applied
    • 30 mL PETG repository, about 20 mL fill, 2 mg/mL protein, pH 6
    • sampling at T0, T1 (i.e. after one f/t step), T2, T3, and T4
    • analytics: visual inspection, SEC, DLS, subvisible particle measurement



FIG. 33 shows 1D4.7 stability during repeated f/t cycling (−80° C./25° C.), mirrored by formation of subvisible particles >1 μm. 1D4.7 was formulated in universal buffer (10 mM citrate, 10 mM phosphate) and then the following excipient variations were tested: sorbitol (10 mg/mL), mannitol (10 mg/mL), sucrose (10 mg/mL), NaCl (100 mM), and polysorbate 80 (0.01%). 1D4.7 was also formulated in water (by dialysis) with no excipients added at all (“water” in FIG. 33). Water for injection was also subjected to f/t cycling and subvisible particle testing to evaluate a potential impact of material handling, f/t, and sample pull on particle load.


The stability of 1D4.7 formulated in water upon f/t exceeded the stability of 1D4.7 solutions formulated with excipients typically used in protein formulations. Mannitol, sucrose, and sorbitol are known to act as lyoprotectant and/or cryoprotectant, and polysorbate 80 is a non-ionic excipient prevalently known to increase physical stability of proteins upon exposure to hydrophobic-hydrophilic interfaces such as air-water and ice-water, respectively.


In summary, 1D4.7 solutions formulated in water appeared to be surprisingly stable when analyzed with various analytical methodologies typically applied to monitor stability of pharmaceutical proteins upon freeze-thaw processing (e.g. SEC, visual inspection, dynamic light scattering, and especially light obscuration).


Example 27: Freeze/Thaw Stability of Low-Ionic 13C5.5 Solutions

13C5.5 (an anti IL-13 IgG1) formulated in water was demonstrated to be stable during repeated freeze/thaw processing (−80° C./25° C. water bath) at 2 mg/mL concentration, pH 6. Data were compared with other formulations (2 mg/mL protein, pH 6), and it was found that the stability of 13C5.5 formulated in water exceeded the stability of 13C5.5 formulated in buffer systems often used in parenteral protein formulations (e.g. 20 mM histidine, 20 mM glycine, 10 mM phosphate, or 10 mM citrate) and even exceeded the stability of 13C5.5 formulations based on universal buffer (10 mM phosphate, 10 mM citrate) that has been combined with a variety of excipients that are commonly used in protein formulation (e.g. 10 mg/mL mannitol, 10 mg/mL sorbitol, 10 mg/mL sucrose, 0.01% polysorbate 80, 20 mM NaCl, 200 mM NaCl).


Sample preparation, experiment processing, sample pull and sample analysis was performed in the same way as outlined in the above examples.

    • 13C5.5 formulated in water compared with formulations listed above
    • 4 freeze/thaw cycles applied
    • 30 mL PETG repository
    • 2 mg/mL, pH 6
    • sampling at T0, T1, T2, T3, and T4
    • analytics: visual inspection, SEC, DLS, subvisible particle measurement



FIG. 34 shows 13C5.5 stability during repeated f/t cycling (−80° C./25° C.), mirrored by formation of subvisible particles >10 μm. 13C5.5 was formulated in either 10 mM phosphate buffer, 10 mM citrate buffer, 20 mM glycine buffer, and 20 mM histidine buffer. 13C5.5 was also formulated in the formulation of invention (by dialysis) with no excipients added at all. Water for injection was also subjected to f/t cycling and subvisible particle testing to evaluate a potential impact of material handling, f/t, and sample pull on particle load (referred to as blank).


The stability of 13C5.5 formulated in water upon f/t exceeded the stability of 13C5.5 solutions formulated in buffers typically used in protein formulations. No instabilities of 13C5.5 solutions formulated in water have been observed with other analytical methodologies applied (e.g. SEC, visual inspection, etc.)



FIG. 35 shows 13C5.5 stability during repeated f/t cycling (−80° C./25° C.), mirrored by formation of subvisible particles >1 μm. 13C5.5 was formulated in universal buffer (10 mM citrate, 10 mM phosphate) and in universal buffer combined with the following excipient variations were tested: sorbitol (10 mg/mL), mannitol (10 mg/mL), sucrose (10 mg/mL), NaCl (200 mM), NaCl (20 mM) and polysorbate 80 (0.01%). 13C5.5 was also formulated in water (by dialysis) with no excipients added at all for comparison (pure water). Water for injection was also subjected to f/t cycling and subvisible particle testing to evaluate a potential impact of material handling, f/t, and sample pull on particle load.


The stability of 13C5.5 formulated in water upon f/t exceeded the stability of 13C5.5 solutions formulated with excipients typically used in protein formulations. Mannitol, sucrose, and sorbitol are known to act as lyoprotectant and/or cryoprotectant, and polysorbate 80 is a non-ionic excipient prevalently known to increase physical stability of proteins upon exposure to hydrophobic-hydrophilic interfaces such as air-water and ice-water, respectively. The low number of subvisible particles in 13C5.5 samples formulated into the formulation of invention was found to be at surprisingly low levels, demonstrating the high safety and stability potential of such formulations.


No instabilities of 13C5.5 solutions formulated in water have been observed with other analytical methodologies applied, (e.g. SEC, visual inspection, etc.).


DLS analysis of 13C5.5 solutions after f/t procedures was performed as described above. Results from the DLS analysis showed that an 13C5.5 solution with 0.01% Tween-80 contained significant high molecular weight (HMW) aggregate forms after only 1 f/t step, whereas 13C5.5 in water contained no HMW aggregate forms, even after 3 f/t steps applied.


In summary, 13C5.5 solutions formulated in water appeared to be surprisingly stable when analyzed with various analytical methodologies typically applied to monitor stability of pharmaceutical proteins upon freeze-thaw processing (e.g. SEC, visual inspection, dynamic light scattering, and especially light obscuration).


Example 28: Freeze/Thaw Stability of Low-Ionic 7C6 Solutions

7C6 (an anti amyloid beta IgG1) formulated in water was demonstrated to be stable during repeated freeze/thaw processing (−80° C./30° C. water bath) at 2 mg/mL concentration, pH 6. Data were compared with other formulations (2 mg/mL protein, pH 6), and it was found that the stability of 7C6 formulated in water exceeded the stability of 7C6 formulated in buffer systems often used in parenteral protein formulations and even exceeded the stability of 7C6 formulations based on universal buffer (10 mM phosphate, 10 mM citrate) that has been combined with a variety of excipients that are commonly used in protein formulation.


The following solution compositions were evaluated for their potential to maintain 7C6 physical stability during freeze/thaw experiments:

    • Phosphate buffer, 15 mM
    • Citrate buffer, 15 mM
    • Succinate buffer, 15 mM
    • Histidine buffer, 15 mM
    • Arginine buffer, 15 mM
    • Low ionic protein formulation, no excipients added
    • Universal buffer, sorbitol (10 mg/mL)
    • Universal buffer, mannitol (10 mg/mL)
    • Universal buffer, sucrose (10 mg/mL)
    • Universal buffer, trehalose (10 mg/mL)
    • Universal buffer, 0.01% (w/w) polysorbate 80


Sample preparation, experiment processing, sample pull and sample analysis was performed in very similar way as outlined in Examples 26 and 27.

    • 7C6 formulated in water compared with formulations listed above
    • 4 freeze/thaw cycles applied
    • 30 mL PETG repository, approx. 20 mL fill
    • 2 mg/mL, pH 6
    • sampling at T0, T1, T2, T3, and T4
    • Analytics: Aβ-antibody stability was assessed by the following methods:
    • Visual inspection of the protein solution was performed in polypropylene round-bottom tubes wherein the samples were filled for light obscuration measurements.


It was carefully inspected against both a black and a white background for signs indicating protein physical instability such as haziness, turbidity and particle formation.

    • Dynamic light scattering (eZetasizer Nano ZS, Malvern Instruments, AI9494; equipped with Hellma precision cells, suprasil quartz, type 105.251-QS, light path 3 mm, center Z8.5 mm, at least 60 μL sample fill, protein sample remaining from light obscuration measurements in PP round-bottom tubes were used for DLS measurements). Automated measurements (1 measurement per sample) were performed.
    • Light obscuration analysis. 3.5 mL of sample were filled in 5 mL round-bottom
    • tube under laminar air flow conditions, measurement was performed in n=3 mode (0.8 mL per single measurement) after an initial 0.8 mL rinse.
    • Size-exclusion chromatography, combined with UV214/UV280 and multi-angle light scattering. Mobile phase: 100 mM Na2HPO4/200 mM Na2SO4, pH 7.0 (49.68 g anhydrous disodium hydrogen phosphate and 99.44 g anhydrous sodium sulfate were dissolved in approx. 3300 mL Milli-Q water, the pH was adjusted to 7.0 using 1 M phosphoric acid, filled to a volume of 3500 mL with Milli-Q water, and the solution was filtered through a membrane filter). SEC column, TSK gel G3000SW (cat. no. 08541) 7.8 mm×30 cm, 5 μm combined with a TSK gel guard (cat. no. 08543) 6.0 mm×4.0 cm, 7 μm. Flow 0.3 mL/min, injection volume 20 μL (equivalent to 20 μg sample), column temperature room temperature, autosampler temp. 2 to 8° C., run time 50 minutes, gradient isocratic, piston rinsing with 10% isopropyl alcohol, detection via UV absorbance, diode array detector: wavelength 214 nm, peak width >0.1 min, band width: 8 nm, reference wavelength 360 nm, band width 100 nm



FIG. 36 shows 7C6 stability during repeated f/t cycling (−80° C./25° C.), mirrored by formation of subvisible particles >1 μm. The stability of 7C6 formulated in water upon f/t for many formulations exceeded the stability of 7C6 solutions formulated in buffers typically used in protein formulations. No instabilities of 7C6 solutions formulated in water have been observed with other analytical methodologies applied (e.g. SEC, visual inspection, dynamic light scattering)


Surprisingly, the stability of 7C6 formulated in water upon f/t exceeded the stability of 7C6 solutions formulated with excipients typically used in protein formulations. Mannitol, sucrose, and sorbitol are known to act as lyoprotectant and/or cryoprotectant, and polysorbate 80 is a non-ionic excipient prevalently known to increase physical stability of proteins upon exposure to hydrophobic-hydrophilic interfaces such as air-water and ice-water, respectively. The low number of subvisible particles in 7C6 samples formulated into the formulation of invention was found to be at surprisingly low levels, demonstrating the high safety and stability potential of such formulations.


In summary, 7C6 solutions formulated in water appeared to be surprisingly stable when analyzed with various analytical methodologies typically applied to monitor stability of pharmaceutical proteins upon freeze-thaw processing (e.g. SEC, visual inspection, dynamic light scattering, and especially light obscuration).


Example 29: Preparation of J695 Formulated in Water and Stability Studies Thereof

Materials and Methods


427.1 g (80 mg/mL) of J695 were diluted to 40 mg/mL and diafiltered using purified water. After a 5-fold volume exchange with purified water (theoretical excipients reduction, 99.3%), the protein solution was ultrafiltered to target concentration of 100 mg/mL. pH, osmolality, density, visual inspection and protein concentration measurements (0D280) were performed to monitor the status of the protein after DF/UF processing.


After DF/UF processing, the protein solution was sterile filtered (0.22 μm Sterivex GV membrane filter) into a 60 mL PETG bottle (Nalgene) and subsequently stored at −80° C. for 3 months.


After thawing at 37° C., the solution was sterile filtered (0.22 μm Sterivex GV membrane filter) and filled into sterile BD Hypak Physiolis SCF™ 1 mL long syringes 29 G, ½ inch, 5-bevel, RNS TPE and closed with sterile BD Hypak SCF™ 1 ml W4023/50 Flur Daikyo stoppers. The filling volume was 1.000 mL per syringe.


After filling, the syringes were stored at 2-8° C. and 40° C., respectively, and analyzed as indicated in the sample pull scheme depicted below.

    • J695 Drug Substance (extinction coefficient at 280 nm: 1.42 mL/mg cm): Drug Substance, pH 6.0, did not contain polysorbate 80.
    • Sartorius Sartocon Slice diafiltration system, equipped with Ultrasert PES membrane cassettes (50 kDa cutoff). The Sartocon Slice system was operated in continuous mode at ambient temperature according to Sartorius Operating Instructions.
    • pH electrodes
    • Perkin Elmer UV/Vis spectrophotometer, Lambda 25, was used for protein concentration measurements (280 nm wavelength). Disposable UV cuvettes, 1.5 mL, semi-micro, were used for the concentration measurements.
    • 0.22 μm filtered purified water was used as DF/UF medium.
    • Anton Paar Density Meter DMA 4100 was used for density measurements
    • A Knauer Osmometer Type ML, was used for osmolality measurements (calibrated with 400 mOsmol/kg NaCl calibration solution, Art. No. Y1241, Herbert Knauer GmbH, Berlin, Germany).


      Analytical Methods
    • J695, SEC analysis: Superdex 200 column (Pharmacia). Mobile phase 92 mM di-sodium hydrogen phosphate, 211 mM sodium sulfate, pH 7.0, 0.75 mL/min flow rate, ambient temperature, detection UV 214 nm. Each sample was diluted to 2.0 mg/mL with mobile phase, sample injection load 20 μg (duplicate injection).
    • J695, IEC analysis: Dionex, Propac WCX-10 column along with a corresponding guard column. Separation conditions: mobile phase A: 20 mM di-sodium hydrogen phosphate and 20 mM sodium acetate, pH 7.0; mobile phase B 20 mM di-sodium hydrogen phosphate, 400 mM Sodium chloride, pH 5.0. 1.0 mL/min flow rate, ambient temperature. Each sample was diluted to 1.0 mg/mL with Milli-Q water, sample injection load 100 μg (duplicate injection).
    • J695, SDS-PAGE analysis: Novex acryl amide slab gels (8-16% for non-reducing conditions, 12% for reducing conditions, Invitrogen), Coomassie staining (Invitrogen). Separation under reducing (β-mercaptoethanol) and non-reducing conditions using Tris-Glycine buffer made of 10× stock solution (Invitrogen).
    • J695, quantitation of buffer components:
      • Mannitol: separation per ReproGel Ca column (Dr. Maisch, Germany) and RI detection, mobile phase: deionized water, 0.6 mL/min flow rate, 20 μL sample injection. Quantitation was performed using external calibration standard curve.
      • Histidine and Methionine: fluorescence labelling of the amino acids with OPA (ortho-phthalic aldehyde) and HPLC separation per ReproSil ODS-3 column (Dr. Maisch, Germany) and fluorescence detection at 420 nm (extinction at 330 nm), mobile phase A: 70% citric acid (10.51 g/L) buffer, pH 6.5, 30% methanol, mobile phase B: methanol, 1.0 mL/min flow rate, 20 μL sample injection. Quantitation was performed using external calibration standard curve.
    • J695, PCS analysis: was performed undiluted at 100 mg/mL in single-use plastic cuvettes at 25° C. using a A Malvern Instruments Zetasizer nano ZS at 173° angle assuming solution viscosity of 4.3875 mPas, refractive index of the protein of 1.450 and refractive index of the buffer solution of 1.335. The averaged results of 20 scans, 20 seconds each, are reported.


      Calculation of the Protein Concentration


Calculation formula:






E
=



-
1



g


(

I

I
0


)



=



ɛ
·
c
·
d


c

=

E

ɛ
×
d










    • ε—absorption coefficient

    • c—concentration

    • d—length of cuvette that the light has to pass

    • E—absorbance

    • I0—initial light intensity

    • I—light intensity after passing through sample










ɛ

J





695


=

1.42


mL

mg
×
cm








Sample Pull Scheme


Samples of the prepared solutions were stored at the temperatures listed below and pulled (x) at the indicated time points after study start (Table 63). Test parameters and methods are described in Table 64.















TABLE 63







Temp.
T0
1 m
3 m
6 m









 5° C.
x
x
x
x



40° C.

x
x
x




















TABLE 64







Test parameter
Test method









Appearance
Visual inspection



Visible particles
analogous DAC (EA 4.43)



Sub-visible particles
analogous




Ph. Eur./USP EA 4.44



Clarity
Ph. Eur. (EA 4.42)



Color (visual)
Ph. Eur. (EA 4.50)



pH
Ph. Eur. (EA 4.24)



Size exclusion HPLC
See above



Cation exchange HPLC
See above



SDS-PAGE
See above



PCS
See above











DF/UF Processing of J695


Table 65 provides the J695 status after diafiltration.













TABLE 65






Protein






Concentration

Osmolality



Sample
[mg/mL]
pH
[mosmol/kg]
Visual Inspection







after
107
6.4
10
Slightly opalescent, slightly


DF/UF



yellow essentially free from






visible particles









After DF/UF the concentrations of the originating buffer components were quantitatively monitored to assess the DF effectiveness. All results were found to be below the practical detection limits (see Table 66) of the corresponding analytical methods (HPLC with RI for Mannitol and fluorescence detection for the methionine and histidine after OPA labeling, respectively).














TABLE 66








Methionine
Histidine
Mannitol



Sample
[mg/mL]
[mg/mL]
[mg/mL]





















before DF/UF
0.669
0.586
18.36



after DF/UF
<0.13
<0.14
<3.20











J695 Characterization upon storage Table 67 below supports the stability of J695 DF/UF at 100 mg/mL upon storage.

















Duration





of
Storage conditions




testing
[° C./% RH]











Test criteria
Specification
[months]
+5
+40/75













Appearance
solution
Initial
complies













1
complies
complies




3
complies
complies




6
complies
complies










Clarity
Report Results,
Initial
≤RSII












Compare to reference suspensions acc.
1





to Ph.Eur.
3
≤RSII
≤RSII




6
≤RSII
≤RSIII










Particulate
Report Result
Initial
2.0 (1)











contamination
Visual Score
1
2.0 (1)
3.0 (1)


Visible particles
(number of samples tested)
3
1.6 (5)
1.0 (5)




6
1.1 (9)
1.6 (9)


Particulate
≥10 μm:≤6000 particles per
initial












contamination
container
≥10 μm
290


Subvisible particles
≥25 μm:≤600 particles per container
≥25 μm
16













1






≥10 μm






≥25 μm






3






>10 μm






≥25 μm






6






≥10 μm
124
54




≥25 μm
1
3


Size Exclusion HPLC
Report Results (%) for
initial













Aggregates (A)
A
0.9



Monomer (M)
M
98.9



Fragments (F)
F
0.2













1






A
0.8
2.3




M
99.0
97.1




F
0.1
0.6




3






A
1.0
3.4




M
98.8
95.1




F
0.1
1.4




6






A
1.4
5.5




M
98.4
85.6




F
0.1
8.9










SDS-PAGE
The predominant banding pattern is comparable
Initial
complies











(Non-reducing
to that of the reference standard.
1
complies
complies


conditions)

3
complies
complies




6
complies
complies










SDS-PAGE
The predominant banding pattern is comparable
Initial
complies











(reducing
to that of the reference standard.
1
complies
complies


conditions)

3
complies
complies




6
complies
complies










PCS
Report Results for
initial
0.9



Z-Average [nm] and

0.23












PDI
1
0.9
1.0





0.23
0.23




3
0.9
1.1





0.23
0.24





0.9
1.3




6
0.23
0.29


Cation Exchange
Report Results (%) for
initial












HPLC
Acidic Species (A)
A
5.9



Main Isoforms (M)
M
91.5



Basic Species (B)
B
2.5













1






A
5.6
10.6




M
92.0
98.9




B
2.4
0.5




3






A
5.7
14.4




M
92.1
85.0




B
2.1
0.6




6






A
6.0
29.6




M
91.6
69.4




B
2.3
1.0










Conclusion


The above example provides a diafiltration/ultrafiltration (DF/UF) experiment where water (0.22 μm filtered purified water) was used as diafiltration medium for the monoclonal antibody J695.


J695 was subjected to DF/UF processing using pure water as DF/UF exchange medium and was formulated at about pH 6.4 at high concentration (100 mg/mL) without inducing solution haziness, severe opalescence or turbidity formation.


J695 from the DF/UF experiments was stored in SCF syringes at 2-8° C. and 40° C. for up to 6 months. Data obtained points to a favourable overall stability of the protein.


In conclusion, processing and formulating proteins using pure water as DF/UF exchange medium is feasible. Assuming an ideal 100% excipient membrane permeability, an approx. 99.3% maximum excipient reduction can be estimated. Evidence is given by specific methods that after DF/UF the excipient concentration is below the practical detection limits.


Example 30: Freeze/Thaw Characteristics and Stability Testing of High Concentration Adalimumab Water Solution—Homogeneity and Physical Stability

Preparation of Low-Ionic Adalimumab Solutions


1.6 L of Drug Substance (DS) material in 2 L PETG bottle was thawed at 25° C. in a water bath, homogenized and subjected to DF/UF using water for injection as a diafiltration exchange medium. Diafiltration was performed in continuous mode with Sartorius Sartocon Slice equipment by applying the following parameter:

    • Pump output: 8%
    • Pressure inlet: max 1 bar (0.8 bar)
    • Membrane: 2×PES, cut off 50 kD


During the diafiltration 5-fold volume exchange was sufficient to reduce osmolality to 8 mOsmol/kg.


In-Process-Control (IPC) samples were pulled prior to diafiltration (SEC, protein concentration by means of OD280, pH, osmolality and density) and after diafiltration (protein concentration by means of OD280, pH, osmolality and density). The IPC-samples were not sterile.


After diafiltration the ˜70 mg/mL Adalimumab formulated in water was diluted to 50 mg/mL with water for injection and the pH value was adjusted to 5.2.


1.6 L of the Adalimumab 50 mg/mL formulated in water pH 5.2 was refilled in 2 L PETG bottle. The remaining volume of Adalimumab solution was subjected to DF/UF to increase the concentration to 100 mg/mL.


The Adalimumab 100 mg/mL formulated in water pH 5.3 was sterile filtered and 0.8 L of them was filled in 1 L PETG bottle.


Analytics






    • Size exclusion chromatography (SEC)

    • pH-measurement

    • Osmolality measurement

    • Density measurement

    • Protein concentration by means of OD280

    • Optical appearance

    • Ion exchange chromatography (IEC)


      Freeze/Thaw Experiment of Adalimumab 1 L Containers





Adalimumab 100° mg/mL formulated in water in 1 L PETG containers was precooled to 2-8° C. and than froze at −80° C., freezing cycle >12 hrs. The frozen samples in 1 L PETG bottles were successively thawed at 25° C. in a water bath. During thawing the bottles of the frozen solutions dipped in the water bath up to liquid level. The following samples were pulled just after thawing without homogenization and after homogenization by 15 and 30 turn top over end.









TABLE 68







Sample pull scheme:











Turns of each bottle
Sample
Analytical tests





1
 0
5 mL top
protein content, osmolality,


2
 0
5 mL middle
pH, density, SEC


3
 0
5 mL bottom



4
15
5 mL top
protein content, osmolality,


5
15
5 mL middle
pH, density, SEC


6
15
5 mL bottom



7
30
5 mL top
protein content, osmolality,


8
30
5 mL middle
pH, density, SEC and


9
30
5 mL bottom
subvisible particles










Characterization of Adalimumab Solutions


Adalimumab 50 mg/mL and 100° mg/mL formulated in water appeared every time clearly, light yellow, not opalescent and without wave pattern after gentle movement.


Also after freezing and thawing the Adalimumab formulated in water did not change the appearance (just after thawing and also after 15 and 30 times turn top over end).


A slight wave patterns were seen after gentle movement of the bottle just after thawing and dipping the needle into the solution during sample pull just after thawing.


In contrast to similar experiments with Adalimumab in commercial buffer the Adalimumab solution 50 mg/mL in water did not show any gradient of protein concentration, density and osmolality.


The Adalimumab solution 100 mg/mL did also not show any gradient of protein concentration, density, osmolality.


Stability was assessed after 6 months storage at −30° C. and −80° C., respectively.


In the following the respective analytical data are outlined:









TABLE 69







Adalimumab 50 and 100 mg/mL, before freeze/thaw processing
















protein content
subvisible particles
















density
osmolality
(gravimertic)
1 mL >=
1 mL >=
1 mL >=



pH
g/cm3
mOsmol/kg
mg/mL
1 μm
10 μm
25 μm

















50 mg/mL in
5.18
1.0121
5
49.3
7953
5
0


water









100 mg/mL in
5.32
1.0262
12
99.8
154
4
2


water
















TABLE 70







Adalimumab 50 mg/mL, pH 5.2 formulated in water, after freeze/thaw processing





















subvisible particles























1 mL
1 mL
1 mL





density
osmolality
protein content
purity (SEC)
>=
>=
>=


turn
sample
pH
g/cm3
mOsmol/kg
mg/mL
%
1 μm
10 μm
25 μm



















0
top
5.20
1.0119
6
48.7
99.597





0
middle
5.19
1.0120
8
49.4
99.576





0
bottom
5.17
1.0120
6
49.8
99.649





15
top
5.20
1.0120
4
49.7
99.649





15
middle
5.18
1.0120
5
49.2
99.678





15
bottom
5.17
1.0120
4
49.1
99.637





30
top
5.19
1.0120
5
49.7
99.647
1280
4
0


30
middle
5.17
1.0120
3
50.4
99.637
2055
13
0


30
bottom
5.18
1.0120
6
48.9
99.611
3889
37
11
















TABLE 71







Adalimumab 100 mg/mL, pH 5.2 formulated in water, after freeze/thaw processing



















protein
purity
subvisible particles



















density
osmolality
content
(SEC)
1 mL >=
1 mL >=
1 mL >=


turn
sample
pH
g/cm3
mOsmol/kg
mg/mL
%
1 μm
10 μm
25 μm



















0
top
5.29
1.0259
13
98.7
99.424





0
middle
5.3
1.0262
16
99.9
99.468





0
bottom
5.28
1.0262
14
101.2
99.48





15
top
5.27
1.0261
13
98.9
99.511





15
middle
5.27
1.0261
16
97.7
99.466






bottom
5.28
1.0261
15
97.0
99.483





30
top
5.29
1.0261
16
96.6
99.439
231
58
49


30
middle
5.28
1.0261
16
97.0
99.467
169
21
9


30
bottom
5.28
1.0261
16
99.3
99.476
131
3
1
















TABLE 72







Adalimumab 100 mg/mL, pH 5.2 formulated


in water, stability after storage















subvisible



SEC


particles


Testing
aggregates
IEC

(1 mL)













time
monomer
sum of lysin
visual
>=1
>=10
>=25


point
fragments
isoforms
appearance
μm
μm
μm
















T0
0.55
85.523
clear,
155
3
1



99.40

no particular






0.05

matter





T6
0.47
82.124
clear,
210
8
5


months,
99.39

no particular





−80° C.
0.14

matter





T6
1.28
81.61
clear,
171
71
51


months,
98.58

no particular





−30° C.
0.14

matter










Conclusion


No significant instabilities of Adalimumab formulated in water at 50 and 100 mg/mL after freeze/thaw processing and after storage at −30° C. or −80° C. for up to 6 months have been observed with the analytical methodologies applied.


Example 31: Freezing and Thawing Process of Adalimumab in Low-Ionic Formulation—Process Design Space Including Protein Content

Preparation of Solution


Adalimumab BDS (Bulk Drug Substance) was thawed in a 23° C. circulating water bath. The solution was up-concentrated to a target concentration of 100 mg/ml for the purpose of volume reduction using a Ultrafiltration/Diafiltration (UF/DF) method (Pellicon “Mini” 2). Two cassettes of Millipore Pellicon 2 tangential flow mini-cassettes with Biomax 10K polyethersulfone were installed in the Pellicon 2 unit. At process start the flow rate was measured at 60 ml/min and feed pressure was 21 psi. The process was stopped at 111.3 mg/ml protein concentration.


Spectra/Por molecularporous membrane tubing was used for dialysis (diameter 48 mm, 18 ml/cm volume, 75 cm length). A volume of 8 L of Adalimumab 100 mg/m at pH 5.2 were transferred to 8 dialysis tubes. Each tube was filled with 1 L of Adalimumab 100 mg/ml. Four tubes equal to 4 L of solution were placed in a container with 36 L of water for injection, i.e. a solution exchange factor of 1:10 was accomplished. The solution was allowed to reach equilibrium before the volume was exchanged against fresh water for injection. The solution exchange was repeated 5 times until a total solution exchange factor of 1:100,000 was reached.


After the solution was completely exchanged by dialysis it was up-concentrated by the second UF/DF step. The second UF/DF step was performed like the first step. A final concentration of 247.5 mg/ml Adalimumab in low-ionic formulation was achieved. The UF/DF was performed with starting material that already contained polysorbate 80. It could be expected that polysorbate 80 accumulated in the final protein solution resulting in a higher polysorbate content than 0.1%.


The up-concentrated bulk solution of 247.5 mg/ml Adalimumab was diluted with WFI to lower protein concentration levels as needed—200 mg/ml, 175 mg/ml, 150 mg/ml, 140 mg/ml, 130 mg/ml, 120 mg/ml, 100 mg/ml, 80 mg/ml, 50 mg/ml, 40 mg/ml, and 25 mg/ml. The bottle fill volume was 1600 ml for all experiments.


Freezing Procedures


A series of increasing freeze rates was used in this evaluation: Ultra-low temperature freezer bottom shelf <Ultra-low temperature freezer middle shelf <Ultra-low temperature freezer top shelf <<Dry ice.


A <−70° C. freezer was used for the experiments (Capacity: 20.2 Cu. Ft. (572 liters). Three shelves were used. Each was loaded with nine 2 L PETG bottles. The bottles were stored at room temperature before being placed in the freezer. Freezing continues for at least 48 hours. For the design space evaluations, three positions with increasing freeze rates were chosen. A front position on the bottom shelf was used for the slowest freeze rate. Faster freeze rates were accomplished at the center position on the middle shelf. The fastest freeze rate in the freezer setup was performed in the back/right position on the top shelf.


For freezing by dry ice, one bottle was completely surrounded by dry ice for at least 8 hours. In a Styrofoam box, the bottom was covered with a layer of dry ice (approx. 3 to 5 cm thick). One bottle was placed standing on top of the dry ice layer. Consequently, the space between the bottle and the inner walls of the styrofoam box was filled with dry ice until every surface but the cap was covered. After freezing time, the bottle was removed and thawed immediately or placed in a −70° C. freezer for storage.


Thawing Procedures


A series of thawing rates was used in this evaluation: Cooled air at 4° C. <<Water bath 23° C. <Water bath 37° C.


Analytics


The following analytics were performed to characterize the samples:

    • Osmolality
    • Conductivity
    • pH
    • Density
    • Protein concentration by direct UV (280 nm)


For the concentration test, samples were diluted with water until an absorbance <1.2 was reached. The absorbance coefficient for the Adalimumab molecule at 280 nm of 1.39 was used.


Characterization of Adalimumab Solutions


Bottle mapping studies revealed a slight tendency towards gradient formation in the bottle volume. Especially for the slower freeze and thaw rates, higher protein concentrations were detected near the bottle bottom. This phenomenon was also reflected in conductivity, density, and osmolality data. The pH appears practically constant in all tested conditions.


In previous investigations regarding the Freeze and Thaw design space for the bottle based system in ultra-low temperature freezers, the appearance of sedimentation was found to be the main failure mode determining the boundaries of the allowable operating range. In this study, this boundary was not observed although the investigated design space covered very wide ranges. The unique behavior of this product is also reflected in the very low tendency to form concentration gradients during this freezing and thawing process. In prior studies it was concluded that the product and process inherent gradient formation is the cause for the appearance of precipitate under certain process conditions. As a result, it was determined that from a process standpoint this system is feasible for Adalimumab in low-ionic formulation pH 5 up to a bulk drug substance concentration of 247.5 mg/ml. The investigated Adalimumab water formulation surprisingly demonstrated superior performance in comparison to other tested Adalimumab formulations.









TABLE 73







Distribution of Protein Concentration, Conductivity, Osmolarity, Density, and pH in the Freshly


Thawed (23° C. water bath) 100 mg/ml Adalimumab in Low Ionic


Formulation Containing Bottles


Freeze & Thaw Conditions: −70 C. Top/23 C. Thaw













sample

osmolarity
conductivity


Adalimumab


name
volume ml
mosm/kg
mS/cm
pH
density g/cm3
conc mg/ml





1
40
11
0.61
5.43
1.021
78.0


2
210
14
0.67
5.43
1.024
92.2


3
225
15
0.70
5.43
1.0253
98.1


4
200
17
0.72
5.46
1.0259
103.9


5
175
18
0.73
5.43
1.0268
100.2


6
180
18
0.74
5.43
1.0275
100.9


7
230
20
0.80
5.46
1.0284
109.1


8
180
22
0.81
5.45
1.0294
111.2


9
150
21
0.82
5.44
1.0307
118.0










Freeze & Thaw Conditions: −70 C. Middle/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
30
8
0.54
5.43
1. 0174
65.3


2
175
18
0.68
5.44
1.0235
91.3


3
200
17
0.70
5.44
1.0245
92.1


4
185
17
0.72
5.44
1.0249
102.9


5
200
13
0.71
5.43
1.0248
95.6


6
200
20
0.73
5.44
1.0262
96.6


7
175
20
0.74
5.44
1.0283
107.5


8
180
20
0.77
5.45
1.0306
116.1


9
200
26
0.82
5.44
1.0346
131.1










Freeze & Thaw Conditions: −70 C. Bottom/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
35
9
0.60
5.41
1.0195
73.2


2
200
13
0.68
5.41
1.0231
89.6


3
225
16
0.70
5.41
1.0241
93.2


4
180
15
0.71
5.41
1.0246
96.8


5
200
15
0.72
5.40
1.0249
95.7


6
200
19
0.73
5.41
1.0259
96.4


7
185
21
0.75
5.42
1.0272
102.6


8
200
26
0.79
5.41
1.0309
116.8


9
175
31
0.85
5.42
1.0372
141.5
















TABLE 74







Distribution of Protein Concentration, Conductivity, Osmolarity, Density, and


pH in the Freshly Thawed (23° C. water bath) 140 mg/ml Adalimumab in Low Ionic


Formulation Containing Bottles


Freeze & Thaw Conditions: −70 C Top/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
PH
g/cm3
conc mg/ml





1
50
36
0.87
5.43
1.0338
130.0


2
215
42
0.90
5.43
1.0354
139.9


3
170
54
0.91
5.43
1.0362
144.9


4
210
41
0.84
5.44
1.0365
141.5


5
200
40
0.93
5.43
1.0364
157.7


6
200
41
0.92
5.43
1.0364
140.0


7
190
41
0..92
5.43
1.0363
143.4


8
180
44
0.82
5.43
1.037
150.1


9
140
45
0.95
5.41
1.038
148.3










Freeze & Thaw Conditions: −70 C. Middle/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
25
32
0.81
5.45
1.0284
112.0


2
175
34
0.84
5.44
1.0307
122.4


3
175
36
0.88
5.44
1.033
133.9


4
200
40
0.90
5.43
1.0342
134.6


5
220
40
0.92
5.43
1.0351
140.9


6
185
45
0.94
5.43
1.0369
143.6


7
210
47
0.97
5.43
1.0384
149.8


8
175
47
0.99
5.43
1.0399
160.3


9
190
48
1.01
5.43
1.0435
168.3










Freeze & Thaw Condition: −70 C. bottom/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
75
28
0.75
5.45
1.0257
88.7


2
180
34
0.82
5.46
1.029
111.6


3
175
34
0.84
5.44
1.0313
123.5


4
220
37
0.86
5.44
1.0322
118.8


5
165
38
0.89
5.45
1.0337
126.3


6
215
44
0.95
5.45
1.0374
137.6


7
210
49
1.00
5.45
1.0407
149.6


8
150
53
1.03
5.43
1.0429
154.9


9
180
60
1.06
5.44
1.0501
183.2
















TABLE 75







Distribution of Protein Concentration, Conductivity, Osmolarity, Density, and pH in the Freshly


Thawed (37° C. water bath) 200 mg/ml Adalimumab in Low Ionic


Formulation Containing Bottles


Freeze & Thaw Conditions: −70 C. Top/37 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
40
37
0.89
5.26
1.0573
197.5


2
210
35
0.96
5.24
1.0573
195.3


3
175
34
0.96
5.22
1.0578
200.3


4
210
36
0.88
5.27
1.0579
193.9


5
210
39
0.89
5.24
1.058
210.6


6
190
39
0.84
5.27
1.058
213.8


7
200
41
0.88
5.27
1.058
206.7


8
170
41
0.88
5.24
1.0581
196.6


9
160
39
0.89
5.29
1.0595
201.9










Freeze & Thaw Conditions: −70 C. Center/37 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
10
31
0.85
5.29
1.0485
170.2


2
185
35
0.89
5.31
1.0505
189.3


3
215
37
0.90
5.33
1.0518
191.7


4
185
38
0.89
5.27
1.0519
191.8


5
200
36
0.90
5.32
1.0528
196.3


6
200
48
0.90
5.28
1.0533
189.3


7
170
37
0.90
5.23
1.0536
193.1


8
215
39
0.91
5.33
1.0552
202.1


9
180
48
0.92
5.31
1.0613
225.5










Freeze & Thaw Conditions: −70 C. Bottom/37 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
50
22
0.96
5.27
1.0361
107.1


2
185
29
0.83
5.26
1.0422
163.0


3
180
38
0.88
5.27
1.0522
201.2


4
185
41
0.90
5.24
1.0535
198.9


5
180
44
0.92
5.28
1.0552
201.4


6
195
40
0.91
5.32
1.0558
201.7


7
180
40
0.91
5.32
1.0560
199.6


8
175
41
0.85
5.26
1.0568
206.2


9
190
48
0.91
5.3
1.0619
229.3
















TABLE 76







Distribution of Protein Concentration, Conductivity, Osmolarity, Density, and pH in the


Freshly Thawed (23° C. water bath) 247.5 mg/ml Adalimumab in Low Ionic


Formulation Containing Bottles


Freeze & Thaw Conditions: −70 C. Top/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
65
46
0.98
5.28
1.0755
260.9


2
190
72
0.97
5.28
1.0751
270.8


3
190
56
0.97
5.28
1.0751
314.7


4
200
49
0.96
5.27
1.0751
274.8


5
200
58
0.96
5.27
1.0752
278.4


6
210
57
0.97
5.28
1.0752
275.0


7
210
76
0.96
5.28
1.0748
276.5


8
175
75
0.96
5.27
1.0754
274.5


9
150
62
0.97
5.28
1.0763
276.3










Freeze & Thaw Conditions: −70 C. Middle/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
80
37
0.95
5.29
1.0671
250.0


2
200
59
0.95
5.32
1.0704
251.3


3
175
51
0.97
5.31
1.0722
262.7


4
215
56
0.98
5.31
1.073
327.1


5
200
48
0.99
5.31
1.0739
267.7


6
200
67
0.98
5.31
1.0744
270.6


7
230
59
0.95
5.32
1.0753
273.2


8
175
70
0.96
5.32
1.0771
273.3


9
175
83
0.96
5.32
1.0825
289.6










Freeze & Thaw Conditions: −70 C. Bottom/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
50
32
0.92
5.24
1.0632
215.3


2
220
59
0.95
5.27
1.069
221.7


3
175
72
0.96
5.27
1.0708
268.1


4
180
58
0.96
5.27
1.0725
260.7


5
210
63
0.96
5.27
1.0729
266.8


6
150
69
0.96
5.28
1.0744
280.3


7
225
50
0.96
5.29
1.0762
280.3


8
200
68
0.95
5.28
1.0789
288.6


9
180
70
0.95
5.29
1.0846
293.0
















TABLE 77





Distribution of Protein Concentration, Conductivity, Osmolarity, Density, and


pH in the Freshly Thawed (23° C. water bath) 247.5 mg/ml Adalimumab in Low Ionic


Formulation Containing Bottles After Dry Ice Freezing







Freeze & Thaw Conditions: Dry Ice Freeze/23 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
50
51
0.94
5.28
1.0643
258.9


2
210
68
0.94
5.29
1.0683
261.9


3
180
50
0.95
5.29
1.0702
251.7


4
190
69
0.95
5.29
1.0732
262.2


5
210
72
0.96
5.31
1.0738
274.4


6
225
63
0.95
5.3
1.0746
265.7


7
160
57
0.95
5.3
1.0747
261.9


8
190
63
0.95
5.31
1.0749
270.9


9
200
50
0.95
5.31
1.075
271.4










Freeze & Thaw Conditions: Dry Ice Freeze/2-8 C. Thaw













sample
volume
osmolarity
conductivity

density
Adalimumab


name
ml
mosm/kg
mS/cm
pH
g/cm3
conc mg/ml





1
50
44
0.96
5.29
1.0665
263.1


2
190
53
0.96
5.31
1.0684
258.1


3
200
56
0.96
5.30
1.0691
247.6


4
200
58
0.96
5.30
1.0693
262.2


5
190
64
0.95
5.31
1.0695
243.2


6
200
61
0.95
5.3
1.0695
266.8


7
175
49
0.96
5.32
1.0697
256.2


8
200
50
0.95
5.31
1.0697
261.2


9
175
48
0.96
5.32
1.0704
247.1









APPENDIX A: PCS DATA

















concen-







tration



peak


concen-
[mg/ml]
z-
z-average
peak
monomer


tration
average
average
average
monomer
average


[mg/ml]
value
[nm]
value [nm]
[nm]
value [nm]















Adalimumab












9.35
9.35
2.08
2.08
2.55
2.55


23.40
23.27
2.30
2.47
2.81
2.87


22.70

2.77

3.01



23.70

2.36

2.78



34.80
34.20
1.56
1.55
1.85
1.87


35.70

1.54

1.82



32.10

1.56

1.93



35.40
36.10
1.61
1.63
1.92
1.92


36.10

1.64

1.93



36.80

1.63

1.92



42.10
43.00
1.75
1.75
2.12
2.12


45.60

1.78

2.15



41.30

1.74

2.10



60.20
57.40
2.06
2.02
2.27
2.37


55.90

2.04

2.45



56.10

1.98

2.39



63.20
65.87
2.11
2.24
2.52
2.67


71.70

2.49

2.89



62.70

2.13

2.61



73.40
75.13
2.38
2.41
2.83
2.89


75.60

2.51

3.01



76.40

2.35

2.82



78.60
78.07
2.53
2.55
2.99
2.99


78.80

2.62

3.01



76.80

2.50

2.96



90.40
95.73
2.80
2.85
3.35
3.41


107.40

2.99

3.55



89.40

2.76

3.33



96.20
94.77
2.88
2.86
3.50
3.50


96.00

2.91

3.61



92.10

2.80

3.38



201.00
206.63
4.52
4.82
5.22
5.74


227.50

5.04

6.12



191.40

4.89

5.89








J695












9.99
9.99
2.28
2.28
1.66
1.66


19.31
19.29
2.05
2.12
1.81
1.81


19.26

2.30

1.79



19.29

2.02

1.84



29.59
29.40
1.78
1.62
1.10
1.16


29.7

1.51

1.15



28.91

1.56

1.22



37.97
37.55
1.51
1.56
1.22
1.23


38.02

1.67

1.22



36.65

1.49

1.24



49.15
46.32
1.64
1.58
1.31
1.29


45.95

1.57

1.30



43.87

1.53

1.26



58.75
56.18
1.60
1.61
1.49
1.47


55.02

1.71

1.38



54.76

1.53

1.53



77.69
77.81
2.64
2.43
2.73
2.61


77.62

2.31

2.57



78.13

2.35

2.52



94.45
97.65
2.11
2.07
2.05
2.05


105.06

2.14

2.05



93.45

1.97

2.04



116.37
114.52
3.69
2.69
1.95
2.00


113.92

2.25

2.06



113.27

2.13

1.99



121.21
133.25
9.78
9.49
11.50
11.00


139.8

9.63

11.10



138.73

9.06

10.40



226.67
217.53
4.94
5.34
4.72
4.84


216.1

6.01

5.25



209.83

5.06

4.55








Human Serum Albumin












9.88
9.88
14.90
14.90
2.32
2.32


22.94
22.89
8.26
8.29
1.2
1.18


22.73

8.28

1.18



23.00

8.33

1.17



36.78
36.47
7.40
7.44
1.22
1.23


37.33

7.80

1.24



35.29

7.12

1.22



45.97
46.06
7.09
6.92
1.27
1.25


47.61

6.54

1.24



44.61

7.13

1.25



58.47
58.56
5.94
6.13
1.3
1.31


62.69

6.04

1.31



54.52

6.41

1.32



61.89
60.31
5.83
6.14
1.33
1.32


59.76

6.57

1.34



59.28

6.01

1.29



75.37
76.24
5.58
5.46
1.4
1.40


83.69

5.14

1.45



69.67

5.67

1.36



92.90
85.87
5.30
5.14
1.49
1.47


84.22

5.05

1.49



80.50

5.08

1.43



115.93
112.74
4.78
4.94
1.68
1.61


110.00

5.04

1.58



112.30

4.99

1.57



182.79
177.69
9.85
9.13
2.27
2.19


178.24

9.29

2.21



172.05

8.26

2.08









APPENDIX B: SEC DATA













Adalimumab















mean

mean

mean

mean


conc.
conc.
monomer
monomer
aggregate
aggregate
fragment
fragment


[mg/ml]
[mg/ml]
[%]
[%]
[%]
[%]
[%]
[%]





9.35
9.35
99.40
99.40
0.50
0.50
0.10
0.10


23.40
23.27
99.60
99.57
0.40
0.40
0.10
0.10


22.70

99.50

0.40

0.10



23.70

99.60

0.40

0.10



34.80
34.20
99.50
99.47
0.50
0.47
0.10
0.10


35.70

99.40

0.50

0.10



32.10

99.50

0.40

0.10



35.40
36.10
99.40
99.40
0.60
0.53
0.10
0.10


36.10

99.40

0.50

0.10



36.80

99.40

0.50

0.10



42.10
43.00
99.40
99.33
0.50
0.57
0.10
0.10


45.60

99.30

0.60

0.10



41.30

99.30

0.60

0.10



60.20
57.40
99.30
99.30
0.60
0.60
0.10
0.10


55.90

99.30

0.60

0.10



56.10

99.30

0.60

0.10



63.20
65.87
99.30
99.27
0.60
0.67
0.10
0.10


71.70

99.20

0.70

0.10



62.70

99.30

0.70

0.10



73.40
75.13
99.20
99.23
0.70
0.70
0.10
0.10


75.60

99.20

0.70

0.10



76.40

99.30

0.70

0.10



78.60
78.07
99.30
99.30
0.60
0.60
0.10
0.10


78.80

99.30

0.60

0.10



76.80

99.30

0.60

0.10



90.40
95.73
99.20
99.13
0.80
0.80
0.10
0.10


107.40

99.10

0.80

0.10



89.40

99.10

0.80

0.10



96.20
94.77
99.10
99.03
0.80
0.87
0.10
0.10


96.00

99.00

0.90

0.10



92.10

99.00

0.90

0.10



201.00
206.63
98.80
98.80
1.10
1.10
0.10
0.10


227.50

98.80

1.10

0.10



191.40

98.80

1.10

0.10



9.99
9.99
99.39
99.39
0.44
0.44
0.17
0.17


19.31
19.29
99.38
99.38
0.44
0.44
0.18
0.19


19.26

99.37

0.44

0.20



19.29

99.38

0.44

0.18



29.59
29.40
99.31
99.33
0.51
0.50
0.18
0.18


29.70

99.32

0.50

0.18



28.91

99.35

0.48

0.17



37.97
37.55
99.31
99.29
0.52
0.52
0.17
0.19


38.02

99.27

0.52

0.21



36.65

99.30

0.51

0.19



49.15
46.32
99.19
99.20
0.60
0.60
0.21
0.20


45.95

99.20

0.60

0.20



43.87

99.20

0.61

0.19



58.75
56.18
99.16
99.16
0.64
0.64
0.21
0.21


55.02

99.17

0.64

0.20



54.76

99.15

0.63

0.22



77.69
77.81
99.11
99.10
0.70
0.70
0.19
0.20


77.62

99.09

0.69

0.22



78.13

99.10

0.70

0.20



94.45
97.65
99.05
99.06
0.72
0.71
0.23
0.22


105.06

99.06

0.72

0.21



93.45

99.07

0.70

0.23



116.37
114.52
98.94
98.91
0.85
0.88
0.21
0.22


113.92

98.91

0.88

0.22



113.27

98.89

0.90

0.22



121.21
133.25
98.87
98.89
0.91
0.90
0.22
0.22


139.80

98.89

0.89

0.22



138.73

98.90

0.89

0.21



226.67
217.53
98.58
98.57
1.19
1.21
0.24
0.23


216.10

98.58

1.18

0.24



209.83

98.54

1.25

0.21










Human Serum Albumin












Peak 1
Peak2
Peak3
Peak4 (HSA)
















Area
Area
Area
Area
Area
Area
Area
Area


sample
[mVs]
[%]
[mVs]
[%]
[mVs]
[%]
[mVs]
[%]





sample 1
59.710
2.312
2.975
0.115
43.159
1.671
2477.282
95.902


C = 9.88 mg/ml










sample 2
102.785
2.685
7.859
0.205
73.588
1.923
3643.350
95187


C = 22.94 mg/ml










sample 3
124.226
3.071
11.038
0.273
83.310
2.059
3826.908
94.597


C = 22.73 mg/ml










sample 4
138.353
3.266
14.525
0.343
88.429
2.087
3994.990
94.304


C = 23.00 mg/ml










sample 5
147.465
3.459
14.537
0.341
91.304
2.141
4010.385
94.059


C = 36.78 mg/ml










sample 6
153.956
3.552
14.707
0.339
94.093
2.171
4071.680
93.938


C = 37.33 mg/ml










sample 7
171.478
3.608
16.064
0.338
105.244
2.214
4459.830
93.839


C = 35.29 mg/ml










sample 8
180.027
3.675
17.392
0.355
109.717
2.239
4592.102
93.731


C = 45.97 mg/ml










sample 9
193.325
3.719
19.206
0.370
116.474
2.241
4868.705
93.670


C = 47.61 mg/ml










sample 10
191.512
3.799
19.167
0.380
112.261
2.227
4718.554
93.594


C = 44.61 mg/ml










sample 11
215.044
4.026
17.870
0.335
118.481
2.218
4989.978
93.421


C = 58.47 mg/ml










sample 12
218.072
4.037
20.088
0.372
122.251
2.263
5041.542
93.328


C = 62.69 mg/ml










sample 13
228.014
4.053
19.957
0.355
126.583
2.250
5251.513
93.343


C = 54.52 mg/ml










sample 14
231.235
4.085
22.518
0.398
127.330
2.250
5279.038
93.267


c = 61.89 mg/ml










sample 15
237.894
41 00
22.939
0.395
130.352
2.246
5411.384
93.258


C = 59.76 mg/ml










sample 16
202.103
4.139
17.178
0.352
108.780
2.228
4554.912
93.282


C = 59.28 mg/ml










sample 17
230.552
4.196
18.565
0.338
123.207
2.242
5122.467
93.224


C = 75.37 mg/ml










sample 18
215.365
4.162
18.136
0.351
110.152
2.129
4830.372
93.358


C = 83.69 mg/ml















Peak 1
Peak2
Peak3
Peak4 (HSA)
















Area
Area
Area
Area
Area
Area
Area
Area


sample
[mVs]
[%]
[mVs]
[%]
[mVs]
[%]
[mVs]
[%]





sample 21
233.866
4.316
21.951
0.405
116.325
2.147
5046.183
93.132


C = 84.22 mg/ml










sample 22
221.816
4.461
18.940
0.381
111.006
2.232
4620.655
92.926


C = 80.50 mg/ml










sample 23
223.187
4.783
16.684
0.358
104116
2.231
4322.732
92.629


C = 115.93 mg/ml










sample 24
209.281
4.718
18.745
0.423
96.430
2174
4111.363
92.686


C = 110.00 mg/ml










sample 25
172.657
4.537
15.457
0.406
80.850
2.125
3536.192
92.932


C = 112.30 mg/ml










sample 26
178.208
4.950
15.254
0.424
80.906
2.247
3325.648
92.379


C = 182.79 mg/ml










sample 27
194.516
4.814
17.323
0.429
90.433
2.238
3738.717
95.520


C = 178.24 mg/ml










sample 28
79.605
2.103
12.876
0.340
74.965
1.981
3617.238
95.576


C = 172.05 mg/ml









APPENDIX C: IEC DATA













Adalimumab











mean conc.
sum Lysin
mean sum


conc. [mg/ml]
[mg/ml]
[%]
[%]





9.35
9.35
86.09
86.09


23.40
23.27
86.15
86.13


22.70

86.12



23.70

86.13



34.80
34.20
86.15
86.11


35.70

86.11



32.10

86.06



35.40
36.10
86.03
86.04


36.10

86.06



36.80

86.03



42.10
43.00
85.98
85.96


45.60

85.95



41.30

85.95



60.20
57.40
85.97
85.96


55.90

85.94



56.10

85.97



63.20
65.87
85.96
85.94


71.70

85.97



62.70

85.90



73.40
75.13
85.99
85.97


75.60

85.98



76.40

85.95



78.60
78.07
86.00
85.97


78.80

85.97



76.80

85.94



90.40
95.73
85.96
85.92


107.40

85.97



89.40

85.83



96.20
94.77
85.93
85.88


96.00

85.87



92.10

85.84



201.00
206.63
85.88
85.90


227.50

85.97



191.40

85.84










J695

















mean
sum
mean sum
sum
mean sum



mean
sum peak
sum
acidic
acidic
basic
basic


conc.
conc.
1-7
peak 1-7
peaks
peaks
peaks
peaks


[mg/ml]
[mg/ml]
[%]
[%]
[%]
[%]
[%]
[%]





9.99
9.99
89.24
89.24
10.24
10.24
0.52
0.52


19.31
19.29
89.32
89.28
10.19
10.21
0.50
0.51


19.26

89.23

10.26

0.52



19.29

89.30

10.19

0.51



29.59
29.40
89.33
89.30
10.14
10.17
0.54
0.53


29.70

89.26

10.20

0.54



28.91

89.32

10.16

0.52



37.97
37.55
89.32
89.30
10.13
10.15
0.56
0.55


38.02

89.27

10.18

0.55



36.65

89.31

10.15

0.55



49.15
46.32
89.07
89.10
10.40
10.37
0.53
0.53


45.95

89.12

10.34

0.54



43.87

89.12

10.36

0.53



58.75
56.18
89.13
89.17
10.36
10.31
0.52
0.53


55.02

89.21

10.27

0.52



54.76

89.18

10.29

0.54



77.69
77.81
89.22
89.17
10.25
10.29
0.53
0.54


77.62

89.09

10.36

0.55



78.13

89.20

10.26

0.55



94.45
97.65
89.20
89.16
10.28
10.30
0.52
0.54


105.06

89.12

10.33

0.55



93.45

89.16

10.29

0.55



116.37
114.52
89.03
89.08
10.41
10.36
0.56
0.55


113.92

89.15

10.31

0.54



113.27

89.06

10.37

0.56



121.21
133.25
89.26
89.13
10.20
10.33
0.54
0.55


139.80

89.07

10.38

0.56



138.73

89.05

10.40

0.55



226.67
217.53
88.72
88.78
10.69
10.63
0.59
0.59


216.10

88.82

10.60

0.58



209.83

88.81

10.60

0.59




















APPENDIX D









Duration






of
63 mg/ml
220 mg/ml


Test Item
Component
Testing
5° C.
5° C.





Clarity and
Turbidity
Initial
3.6
8.0


opalescence

1 month
3.5
8.0




3 month
3.5
7.4


Degree of
B scale
Initial
<B9
=B9


coloration of

1 month
<B9
<B8


liquids

3 month
<B9
<B7


pH
Single value
Initial
5.3
5.4




1 month
5.3
5.4




3 month
5.3
5.4


Particulate
visual score
Initial
2.2
0.2


contamination:

1 month
2.2
0.4


visible particles

3 month
2.1
0.2


Particulate
Particles >= 10
Initial
181
357


contamination:
μm [/Container]
1 month
423
290


subvisible

3 month
216
1762


particles
Particles >= 25
Initial
15
3



μm [/Container]
1 month
11
18




3 month
2
50


Size exclusion
Principal peak
Initial
0.2
0.5


chromatography
(aggregate) [%]
1 month
0.2
0.6


(SE-HPIC)

3 month
0.2
0.7



Principal peak
Initial
99.8
99.4



(monomer) [%]
1 month
99.7
99.3




3 month
99.7
99.2



Principal peak
Initial
0.1
0.1



(fragment)[%]
1 month
0.1
0.1




3 month
0.0
0.0


Cation exchange
1st acidic region
Initial
2.2
2.2


HPIC(CEX-
[%]
1 month
2.2
2.2


HPIC)

3 month
2.1
2.0



2nd acidic
Initial
10.4
10.3



region[%]
1 month
10.2
10.0




3 month
10.4
10.2



Sum of lysine
Initial
86.0
86.1



variants [%]
1 month
85.9
85.9




3 month
86.2
86.1



Peak between
Initial
0.8
0.8



lysine 1 und
1 month
1.0
1.0



lysine 2 [%]
3 month
0.8
0.8



Peaks after
Initial
0.5
0.6



Lysin 2 [%]
1 month
0.7
0.9




3 month
0.5
0.8







Duration
63 mg/ml
220 mg/ml




of
25° C./60%
25° C./60%


Test Item
Component
Testing
R.H.
R.H.





Clarity and
Turbidity
Initial




opalescence

1 month
3.51
8.55




3 month
3.70
7.56


Degree of
B scale
Initial




coloration of

1 month
<B9
<B8


liquids

3 month
<B9
<B7


pH
Single value
Initial






1 month
5.4
5.4




3 month
5.3
5.4


Particulate
visual score
Initial




contamination:

1 month
2.5
0.7


visible particles

3 month
3.4
0.0


Particulate
Particles >=10
Initial




contamination:
μm [/Container]
1 month
412
490


subvisible

3 month
277
4516


particles
Particles >=25
Initial





μm [/Container]
1 month
10
14




3 month
7
128


Size exclusion
Principal peak
Initial




chromatography
(aggregate) [%]
1 month
0.3
0.8


(SE-HPIC)

3 month
0.4
1.1



Principal peak
Initial





(monomer) [%]
1 month
99.6
99.0




3 month
99.4
98.6



Principal peak
Initial





(fragment)[%]
1 month
0.2
0.2




3 month
0.2
0.2


Cation exchange
1st acidic region
Initial




HPIC (CEX-
[%]
1 month
2.5
2.4


HPIC)

3 month
3.4
3.2



2nd acidic
Initial





region[%]
1 month
11.7
11.4




3 month
15.3
14.9



Sum of lysine
Initial





variants [%]
1 month
83.6
83.8




3 month
79.2
79.2



Peak between
Initial





lysine 1 und
1 month
1.2
1.3



lysine 2 [%]
3 month
1.3
1.3



Peaks after
Initial





Lysin 2 [%]
1 month
0.9
1.1




3 month
0.8
1.4







Duration
63 mg/ml
220 mg/ml




of
40° C./75%
40° C./75%


Test Item
Component
Testing
R.N.
R.N.





Clarity and
Turbidity
Initial




opalescence

1 month
3.93
7.80




3 month
3.70
8.10


Degree of
B scale
Initial




coloration of

1 month
=B9
=B8


liquids

3 month
<B8
<B7


pH
Single value
Initial






1 month
5.3
5.4




3 month
5.3
5.4


Particulate
visual score
Initial




contamination:

1 month
6.7
0.5


visible particles

3 month
17.5
0.4


Particulate
Particles >=10
Initial




contamination:
μm [/Container]
1 month
1088
518


subvisible

3 month
166
612


particles
Particles >=25
Initial





μm [/Container]
1 month
16
14




3 month
11
30


Size exclusion
Principal peak
Initial




chromatography
(aggregate) [%]
1 month
0.4
1.4


(SE-HPIC)

3 month
0.8
2.5



Principal peak
Initial





(monomer) [%]
1 month
99.0
98.0




3 month
97.8
96.0



Principal peak
Initial





(fragment)[%]
1 month
0.6
0.6




3 month
1.4
1.5


Cation exchange
1st acidic region
Initial




HPIC (CEX-
[%]
1 month
6.7
6.8


HPIC)

3 month
17.5
17.4



2nd acidic
Initial





region[%]
1 month
25.1
23.6




3 month
40.9
38.6



Sum of lysine
Initial





variants [%]
1 month
64.5
62.0




3 month
36.0
36.0



Peak between
Initial





lysine 1 und
1 month
2.2
2.5



lysine 2 [%]
3 month
2.9
3.1



Peaks after
Initial





Lysin 2 [%]
1 month
1.5
5.2




3 month
1.7
4.8
















APPENDIX E







2-8° C.



















Duration
LI
LI
LI
LI
LI
LI
LI
LI




of
50
50
50
50
50
50
50
50


Test Item
Component
testing
01*2
02*2
03*2
04*2
05*2
06*2
07*2
08*2





Clarity and
Absorption
initial
0.096
0.096
0.095
0.100
0.104
0.105
0.099
0.107


opalescence
(340 nm)
1 month
0.102
0.102
0.093
0.094
0.099
0.101
0.097
0.100










Degree of
visual
initial
clear and colorless


coloration

1 month
clear and colorless

















pH
Single value
initial
5.4
5.4
5.4
5.4
5.4
5.4
5.4
5.4




1 month
5.4
5.5.
5.5.
5.4
5.5.
5.4
5.5.
5.5.


Size exclusion
Principal peak
initial
0.1
0.1
0.1
0.1
0.1
0.1
0.2
0.2


chroma-
(aggregate) [%]
1 month
0.1
0.1
0.1
0.1
0.1
0.1
1.7
0.2


tography
Principal peak
initial
99.6
99.6
99.6
99.6
99.6
99.6
99.7
99.7


(SE-HPLC
(monomer) [%]
1 month
99.7
99.7
99.7
99.7
99.7
99.7
99.7
99.7



Principal peak
initial
0.3
0.3
0.2
0.2
0.2
0.2
0.2
0.2



(fragment) [%]
1 month
0.2
0.2
0.2
0.2
0.2
0.2
0.2
0.2


Cation
1st acidic region
initial
3.8
3.7
3.7
3.7
3.7
3.9
3.8
3.8


exchange
[%]
1 month
3.4
3.4
3.4
3.4
3.4
3.5
3.4
3.4


HPLC
2nd cidic region
initial
10.9
10.7
10.4
10.5
10.4
101
10.3
10.2


(CEX-HPLC)
[%]
1 month
9.2
9.2
9.2
9.2
9.2
9.2
9.2
9.3



Sum of lysine
initial
83.8
84.2
84.4
84.3
84.4
84.6
84.4
84.6



variants [%]
1 month
86.0
86.0
86.0
86.0
86.0
85.9
86.1
86.0



Peak between
initial
0.9
8.3
0.8
0.9
0.8
0.8
0.8
0.8



lysine 1 and
1 month
0.9
0.8
0.8
0.8
0.8
0.8
0.8
0.8



lysine 2 [%]












Peaks after
initial
0.6
0.6
0.6
0.6
0.6
0.6
0.6
0.6



Lysin 2 [%]
1 month
0.6
0.5
0.5
0.5
0.5
0.5
0.5
0.5










25° C.



















Duration
LI
LI
LI
LI
LI
LI
LI
LI




of
50
50
50
50
50
50
50
50


Test Item
Component
testing
01*2
02*2
03*2
04*2
05*2
06*2
07*2
08*2





Clarity and
Absorption
initial
0.096
0.096
0.095
0.100
0.104
0.105
0.099
0.107


opalescence
(340 nm)
1 month
0.106
0.109
0.096
0.099
0.104
0.104
0.096
0.105










Degree of
visual
initial
clear and colorless


coloration

1 month
clear and colorless

















pH
Single value
initial
5.4
5.4
5.4
5.4
5.4
5.4
5.4
5.4




1 month
5.4
5.4
5.4
5.4
5.4
5.4
5.4
5.4


Size exclusion
Principal peak
initial
0.1
0.1
0.1
0.1
0.1
0.1
0.2
0.2


chroma-
(aggregate) [%]
1 month
0.2
0.2
0.2
0.2
0.2
?
0.2
0.2


tography
Principal peak
initial
99.6
99.6
99.6
99.6
99.6
99.6
99.7
99.7


(SE-HPLC
(monomer) [%]
1 month
99.6
99.6
99.6
99.6
99.6
?
99.5
99.5



Principal peak
initial
0.3
0.3
0.2
0.2
0.2
0.2
0.2
0.2



(fragment) [%]
1 month
0.2
0.2
0.2
0.2
0.2
?
0.2
0.2


Cation
1st acidic region
initial
3.8
3.7
3.7
3.7
3.7
3.9
3.8
3.8


exchange
[%]
1 month
4.1
4.2
4.1
4.1
4.2
4.2
4.1
4.2


HPLC
2nd cidic region
initial
10.9
10.7
10.4
10.5
10.4
10.1
10.3
10.2


(CEX-HPLC)
[%]
1 month
10.9
10.9
110
10.9
11.0
11.0
11.0
11.1



Sum of lysine
initial
83.8
84.2
84.4
84.3
84.4
84.6
84.4
84.6



variants [%]
1 month
83.3
83.4
83.3
83.3
83.2
83.2
83.3
83.1



Peak between
1 initial
0.9
83
0.8
0.9
0.8
0.8
0.8
0.8



lysine and
1 month
1.0
0.9
1.0
1.0
1.0
0.9
0.9
1.0



lysine 2 [%]












Peaks after
initial
0.6
0.6
0.6
0.6
0.6
0.6
0.6
0.6



Lysin 2 [%]
1 month
0.7
0.6
0.7
0.7
0.7
0.6
0.6
0.6










40° C.



















Duration
LI
LI
LI
LI
LI
LI
LI
LI




of
50
50
50
50
50
50
50
50


Test Item
Component
testing
01*2
02*2
03*2
04*2
05*2
06*2
07*2
08*2





Clarity and
Absorption
initial
0.096
0.096
0.095
0.100
0.104
0.105
0.099
0.107


opalescence
(340 nm)
1 month
0.100
0.110
0.099
0.101
0.111
0.116
0.105
0.113










Degree of
visual
initial
clear and colorless


coloration

1 month
clear and colorless

















pH
Single value
initial
5.40
5.41
5.41
5.40
5.40
5.40
5.41
5.41




1 month
5.42
5.42
5.43
5.43
5.44
5.43
5.44
5.44


Size exclusion
Principal peak
initial
0.106
0.104
0.119
0.136
0.139
0.145
0.158
0.159


chroma-
(aggregate) [%]
1 month
0.324
0.336
0.322
0.334
0.347
0.349
0.411
0.444


tography
Principal peak
initial
99.605
99.629
99.632
99.626
99.619
99.626
99.653
99.655


(SE-HPLC
(monomer) [%]
1 month
98.924
98.914
98.931
98.914
98.895
98.894
98.845
98.782



Principal peak
initial
0.289
0.267
0.249
0.238
0.242
0.229
0.189
0.186



(fragment) [%]
1 month
0.752
0.751
0.747
0.752
0.758
0.757
0.744
0.773


Cation
1st acidic region
initial
3.8
3.7
3.7
3.7
3.7
3.9
3.8
3.8


exchange
[%]
1 month
5.4
5.8
5.3
5.8
5.4
5.8
5.4
5.5


HPLC
2nd cidic region
initial
10.9
10.7
10.4
10.5
10.4
10.1
10.3
10.2


(CEX-HPLC)
[%]
1 month
29.8
29.8
29.7
29.7
29.8
29.8
30.2
30.7



Sum of lysine
initial
83.8
84.2
84.4
84.3
84.4
84.6
84.4
84.6



variants [%]
1 month
61.2
61.0
61.3
61.0
61.2
60.9
60.9
60.5



Peak between
initial
0.9
8.3
0.8
0.9
0.8
0.8
0.8
0.8



lysine 1
1 month
2.3
2.1
2.3
2.2
2.2
2.2
2.1
2.0



and lysine 2 [%]












Peaks after
initial
0.6
0.6
0.6
0.6
0.6
0.6
0.6
0.6



Lysin 2 [%]
1 month
1.3
1.3
1.3
1.3
1.4
1.3
1.4
1.4










2-8° C.



















Duration
LI
LI
LI
LI
LI
LI
LI
LI




of
200
200
200
200
200
200
200
200


Test Item
Component
testing
01*2
02*2
03*2
04*2
05*2
06*2
07*2
08*2





Clarity and
Absorption
initial
0.181
0.187
0.182
0.192
0.184
0.197
0.191
0.199


opalescence
(340 nm)
1 month
0.181
0.185
0.189
0.181
0.190
0.180
0.192
0.192










Degree of
visual
initial
clear and colorless


coloration

1 month
clear and colorless

















pH
Single value
initial
5.6
5.6
5.6
5.6
5.6
5.5.
5.6
5.5




1 month
5.6
5.6
5.5.
5.5.
5.6
5.5.
5.6
5.6


Size exclusion
Principal peak
initial
0.5
0.5
0.5
0.5
0.5
0.5
0.5
0.6


chroma-
(aggregate) [%]
1 month
0.6
0.6
0.6
0.6
0.6
0.6
0.7
0.7


tography
Principal peak
initial
99.3
99.3
99.3
99.3
99.3
99.3
99.3
99.3


(SE-HPLC
(monomer) [%]
1 month
99.3
99.3
99.2
99.3
99.3
99.2
99.2
99.2



Principal peak
initial
0.2
0.2
0.2
0.2
0.2
0.2
0.2
0.1



(fragment) [%]
1 month
0.2
0.2
0.2
0.2
0.2
0.2
0.2
0.2


Cation
1st acidic region
initial
3.7
3.8
3.6
4.5
3.9
2.7
2.7
2.8


exchange
[%]
1 month
3.4
3.4
3.5
3.5
3.5
3.5
3.5
3.5


HPLC
2nd cidic region
initial
10.4
10.2
98
10.1
95
11.6
11.5
11.3


(CEX-HPLC)
[%]
1 month
9.1
9.0
9.1
9.1
9.0
9.1
9.1
9.1



Sum of lysine
initial
84.4
84.6
85.2
83.9
85.2
84.4
84.3
84.5



variants [%]
1 month
86.3
86.1
86.1
86.2
86.0
86.0
86.1
86.1



Peak between
initial
0.8
0.8
0.8
0.8
0.8
0.8
0.8
0.8



lysine 1












and lysine 2 [%]
1 month
0.6
0.8
0.6
0.6
0.8
0.7
0.6
0.6



Peaks after
initial
0.6
0.6
0.6
0.7
0.6
0.6
0.6
0.6



Lysin 2 [%]
1 month
0.7
0.7
0.7
0.7
0.7
0.7
0.8
0.7










25° C.



















Duration
LI
LI
LI
LI
LI
LI
LI
LI




of
200
200
200
200
200
200
200
200


Test Item
Component
testing
01*2
02*2
03*2
04*2
05*2
06*2
07*2
08*2





Clarity and
Absorption
initial
0.181
0.187
0.182
0.192
0.184
0.197
0.191
0.199


opalescence
(340 nm)
1 month
0.178
0.177
0.198
0.189
0.200
0.194
0.194
0.172










Degree of
visual
initial
clear and colorless


coloration

1 month
clear and colorless

















pH
Single value
initial
5.6
5.6
5.6
5.6
5.6
5.5
5.6
5.5




1 month
5.5
5.6
5.6
5.5
5.5
5.5
5.5
5.5


Size exclusion
Principal peak
initial
0.5
0.5
0.5
0.5
0.5
0.5
0.5
0.6


chroma-
(aggregate) [%]
1 month
0.8
0.8
0.8
0.8
0.8
0.8
0.9
0.9


tography
Principal peak
initial
99.3
99.3
99.3
99.3
99.3
99.3
99.3
99.3


(SE-HPLC
(monomer) [%]
1 month
99.0
99.0
99.0
99.0
99.0
99.0
98.9
98.9



Principal peak
initial
0.2
0.2
0.2
0.2
0.2
0.2
0.2
0.1



(fragment) [%]
1 month
0.2
0.2
0.2
0.2
0.2
0.2
0.2
0.2


Cation
1st acidic region
initial
3.7
3.8
3.6
4.5
3.9
2.7
2.7
2.8


exchange
[%]
1 month
4.0
4.0
4.1
4.1
4.0
4.1
4.1
4.0


HPLC
2nd cidic region
initial
10.4
10.2
9.8
10.1
9.5
11.6
11.5
11.3


(CEX-HPLC)
[%]
1 month
10.6
10.7
10.7
10.7
10.7
10.7
10.7
10.9



Sum of lysine
initial
84.4
84.6
85.2
83.9
85.2
84.4
84.3
84.5



variants [%]
1 month
83.4
83.6
83.4
83.6
83.6
83.5
83.3
83.4



Peak between
initial
0.8
0.8
0.8
0.8
0.8
0.8
0.8
0.8



1 lysine and
1 month
1.0
0.8
0.8
0.7
0.7
0.8
0.9
0.8



lysine 2 [%]












Peaks after
initial
0.6
0.6
0.6
0.7
0.6
0.6
0.6
0.6



Lysin 2 [%]
1 month
0.9
0.9
0.9
0.9
0.9
0.9
0.9
0.9










40° C.



















Duration
LI
LI
LI
LI
LI
LI
LI
LI




of
200
200
200
200
200
200
200
200


Test Item
Component
testing
01*2
02*2
03*2
04*2
05*2
06*2
07*2
08*2





Clarity and
Absorption
initial
0.181
0.187
0.182
0.192
0.184
0.197
0.191
0.199


opalescence
(340 nm)
1 month
0.204
0.202
0.198
0.205
0.199
0.216
0.209
0.224










Degree of
visual
initial
clear and colorless


coloration

1 month
clear and colorless

















pH
Single value
initial
5.57
5.56
5.55
5.56
5.55
5.54
5.55
5.52




1 month
5.54
5.55
5.55
5.55
5.55
5.55
5.53
5.54


Size exclusion
Principal peak
initial
0.489
0.509
0.491
0.484
0.492
0.488
0.515
0.575


chroma-
(aggregate) [%]
1 month
1.308
1.343
1.331
1.340
1.341
1.374
1.453
1.521


tography
Principal peak
initial
99.294
99.296
99.348
99.333
99.313
99.349
99.320
99.290


(SE-HPLC
(monomer) [%]
1 month
97.920
97.876
97.892
97.901
97.898
97.861
97.752
97.701



Principal peak
initial
0.218
0.196
0.161
0.183
0.195
0.163
0.165
0.135



(fragment) [%]
1 month
0.773
0.781
0.777
0.759
0.762
0.765
0.794
0.779


Cation
1st acidic region
initial
3.7
3.8
3.6
4.5
3.9
2.7
2.7
2.8


exchange
[%]
1 month
5.3
5.6
5.3
5.6
5.3
5.6
5.3
5.4


HPLC
2nd cidic region
initial
10.4
10.2
9.8
10.1
9.5
11.6
11.5
11.3


(CEX-HPLC)
[%]
1 month
28.6
28.9
28.5
28.7
28.6
28.9
29.1
29.2



Sum of lysine
initial
84.4
84.6
85.2
83.9
85.2
84.4
84.3
84.5



variants [%]
1 month
62.0
61.7
62.0
61.8
62.0
61.6
61.5
61.4



Peak between
initial
0.8
0.8
0.8
0.8
0.8
0.8
0.8
0.8



lysine 1
1 month
2.2
2.0
2.2
2.0
2.2
2.0
2.0
2.0



and lysine 2 [%]












Peaks after
initial
0.6
0.6
0.6
0.7
0.6
0.6
0.6
0.6



Lysin 2 [%]
1 month
1.9
1.9
2.0
1.9
1.9
1.9
2.0
2.0









EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims. The contents of all references, patents and published patent applications cited throughout this application are incorporated herein by reference.

Claims
  • 1. An aqueous pharmaceutical formulation comprising: (a) an anti-tumor necrosis factor alpha antibody comprising a light chain variable region (LCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO:3, a CDR2 domain comprising the amino acid sequence of SEQ ID NO:5, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 7, and a heavy chain variable region (HCVR) having a CDR3 domain comprising the amino acid sequence of SEQ ID NO:4, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 6, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO:8, wherein the concentration of the antibody is 50 to 200 mg/ml;(b) a surfactant;(c) a polyol; and(d) water;wherein the formulation does not comprise a buffering system, andwherein the pH of the formulation is from 4 to 8.
  • 2. The formulation of claim 1, wherein the surfactant is a non-ionic surfactant.
  • 3. The formulation of claim 2, wherein the surfactant is a polysorbate.
  • 4. The formulation of claim 3, wherein the surfactant is polysorbate-20 or polysorbate-80.
  • 5. The formulation of claim 1, wherein the polyol is a sugar or sugar alcohol.
  • 6. The formulation of claim 5, wherein the polyol is trehalose, sucrose or mannitol.
  • 7. The formulation of claim 1, wherein the surfactant is a non-ionic surfactant, and wherein the polyol is a sugar or sugar alcohol.
  • 8. The formulation of claim 7, wherein the surfactant is a polysorbate.
  • 9. The formulation of claim 8, wherein the surfactant is polysorbate-20 or polysorbate-80.
  • 10. The formulation of claim 7, wherein the polyol is trehalose, sucrose or mannitol.
  • 11. The formulation of claim 8, wherein the polyol is trehalose, sucrose or mannitol.
  • 12. The formulation of claim 9, wherein the polyol is trehalose, sucrose or mannitol.
  • 13. An aqueous pharmaceutical formulation comprising: (a) 100 mg/ml adalimumab;(b) a polysorbate;(c) a polyol, wherein the polyol is trehalose, sucrose or mannitol, and(d) water;wherein the formulation does not comprise a buffering system, andwherein the pH of the formulation is from 4 to 8.
  • 14. The formulation of claim 13, wherein the polysorbate is polysorbate-20 or polysorbate-80.
  • 15. An aqueous pharmaceutical formulation comprising: (a) 100 mg/ml adalimumab;(b) polysorbate-20;(c) a polyol, wherein the polyol is trehalose or sucrose, and(d) water;wherein the formulation does not comprise a buffering system, andwherein the pH of the formulation is from 4 to 6.
  • 16. The formulation of claim 15, wherein the pH is from 5 to 6.
  • 17. The formulation of claim 15, wherein the polyol is trehalose.
  • 18. The formulation of claim 15, wherein the polyol is sucrose.
  • 19. The formulation of claim 16, wherein the polyol is trehalose.
  • 20. The formulation of claim 16, wherein the polyol is sucrose.
RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 16/228,478 filed on Dec. 20, 2018, pending, which is a continuation of U.S. patent application Ser. No. 15/423,503 filed on Feb. 2, 2017, abandoned, which is a continuation of U.S. patent application Ser. No. 15/096,043, filed on Apr. 11, 2016, abandoned, which is a continuation of U.S. patent application Ser. No. 14/796,389, filed on Jul. 10, 2015, abandoned, which is a divisional of U.S. patent application Ser. No. 14/506,576, filed on Oct. 3, 2014, now U.S. Pat. No. 9,085,619, issued on Jul. 21, 2015, which is a continuation of U.S. patent application Ser. No. 13/774,735, now U.S. Pat. No. 8,883,146, issued on Nov. 11, 2014, which is a continuation of U.S. patent application Ser. No. 12/325,049, now U.S. Pat. No. 8,420,081, issued on Apr. 16, 2013, which claims the benefit of priority to U.S. Provisional Application No. 61/004,992, filed on Nov. 30, 2007. The entire contents of each of the foregoing applications are hereby incorporated by reference herein.

US Referenced Citations (271)
Number Name Date Kind
4396608 Tenold Aug 1983 A
4597966 Zolton et al. Jul 1986 A
4681713 Miyagi et al. Jul 1987 A
4876088 Hirao et al. Oct 1989 A
4877608 Lee et al. Oct 1989 A
5237054 Brinks et al. Aug 1993 A
5608038 Eibl et al. Mar 1997 A
5654403 Smith et al. Aug 1997 A
5656730 Lee et al. Aug 1997 A
5702699 Hanisch et al. Dec 1997 A
5945098 Sarno et al. Aug 1999 A
6024938 Corbo et al. Feb 2000 A
6090382 Salfeld et al. Jul 2000 A
6165467 Hagiwara et al. Dec 2000 A
6171586 Lam et al. Jan 2001 B1
6252055 Relton Jun 2001 B1
6258562 Salfeld et al. Jul 2001 B1
6281336 Laursen et al. Aug 2001 B1
6287336 Globerman et al. Sep 2001 B1
6436397 Baker et al. Aug 2002 B1
6485392 Miyazaki et al. Nov 2002 B2
6485725 Hirao et al. Nov 2002 B1
6485932 McIntosh et al. Nov 2002 B1
6509015 Salfeld et al. Jan 2003 B1
6693173 Mamidi et al. Feb 2004 B2
6696056 Cheung et al. Feb 2004 B1
6737405 Roemisch et al. May 2004 B2
6818613 Sharma et al. Nov 2004 B2
6875432 Liu et al. Apr 2005 B2
6991790 Lam et al. Jan 2006 B1
7070775 Le et al. Jul 2006 B2
7220409 Norman et al. May 2007 B2
7223394 Salfeld et al. May 2007 B2
7250165 Heavner et al. Jul 2007 B2
7276239 Le et al. Oct 2007 B2
7318931 Okumu et al. Jan 2008 B2
7541031 Salfeld et al. Jun 2009 B2
7588761 Salfeld et al. Sep 2009 B2
7758860 Warne et al. Jul 2010 B2
7863426 Wan et al. Jan 2011 B2
7919264 Maksymowych et al. Apr 2011 B2
8034906 Borhani et al. Oct 2011 B2
8092998 Stuhlmuller et al. Jan 2012 B2
8093045 Pla et al. Jan 2012 B2
8187836 Hsieh May 2012 B2
8197813 Salfeld et al. Jun 2012 B2
8206714 Salfeld et al. Jun 2012 B2
8216583 Krause et al. Jul 2012 B2
8231876 Wan et al. Jul 2012 B2
8372400 Salfeld et al. Feb 2013 B2
8372401 Salfeld et al. Feb 2013 B2
8414894 Salfeld et al. Apr 2013 B2
8420081 Fraunhofer et al. Apr 2013 B2
8436149 Borhani et al. May 2013 B2
8795670 Krause et al. Aug 2014 B2
8802100 Krause et al. Aug 2014 B2
8802101 Krause et al. Aug 2014 B2
8802102 Krause et al. Aug 2014 B2
8821865 Neu et al. Sep 2014 B2
8883146 Fraunhofer et al. Nov 2014 B2
8911741 Krause et al. Dec 2014 B2
8916157 Krause et al. Dec 2014 B2
8916158 Krause et al. Dec 2014 B2
8932591 Krause et al. Jan 2015 B2
8940305 Krause et al. Jan 2015 B2
9085619 Fraunhofer et al. Jul 2015 B2
9114166 Krause et al. Aug 2015 B2
9340611 Manning et al. May 2016 B2
9340612 Manning et al. May 2016 B2
9346880 Manning et al. May 2016 B2
9382317 Manning et al. Jul 2016 B2
9682145 Manning et al. Jun 2017 B2
9707293 Manning et al. Jul 2017 B2
9724414 Manning et al. Aug 2017 B2
9724415 Manning et al. Aug 2017 B2
9731008 Manning et al. Aug 2017 B2
9731009 Manning et al. Aug 2017 B2
9732152 Krause et al. Aug 2017 B2
9737600 Manning et al. Aug 2017 B2
9738714 Krause et al. Aug 2017 B2
9750808 Krause et al. Sep 2017 B2
9757454 Manning et al. Sep 2017 B2
9770507 Manning et al. Sep 2017 B2
9782479 Manning et al. Oct 2017 B2
9782480 Manning et al. Oct 2017 B2
9789185 Manning et al. Oct 2017 B2
9808525 Manning et al. Nov 2017 B2
9861695 Manning et al. Jan 2018 B2
9950066 Krause et al. Apr 2018 B2
10047141 Puchacz et al. Aug 2018 B2
10155039 Manning et al. Dec 2018 B2
10159732 Manning et al. Dec 2018 B2
10159733 Manning et al. Dec 2018 B2
10195275 Manning et al. Feb 2019 B2
10207000 Manning et al. Feb 2019 B2
10286071 Manning et al. May 2019 B2
10286072 Manning et al. May 2019 B2
10688183 Manning et al. Jun 2020 B2
10716852 Manning et al. Jul 2020 B2
10716853 Manning et al. Jul 2020 B2
10716854 Manning et al. Jul 2020 B2
10722579 Manning et al. Jul 2020 B2
10772959 Manning et al. Sep 2020 B2
10772960 Manning et al. Sep 2020 B2
10780163 Manning et al. Sep 2020 B2
10786566 Manning et al. Sep 2020 B2
10799585 Manning et al. Oct 2020 B2
20030012786 Teoh et al. Jan 2003 A1
20030045454 Okumu et al. Mar 2003 A1
20030049725 Heavner et al. Mar 2003 A1
20030138417 Kaisheva et al. Jul 2003 A1
20030143603 Giles-Komar et al. Jul 2003 A1
20030161828 Abdelghany et al. Aug 2003 A1
20030180287 Gombotz et al. Sep 2003 A1
20030190316 Kakuta et al. Oct 2003 A1
20030206898 Fischkoff et al. Nov 2003 A1
20030219438 Salfeld et al. Nov 2003 A1
20030235585 Fischkoff et al. Dec 2003 A1
20040009172 Fischkoff et al. Jan 2004 A1
20040028667 Norman et al. Feb 2004 A1
20040033228 Krause et al. Feb 2004 A1
20040033535 Boyle et al. Feb 2004 A1
20040038878 Tanikawa et al. Feb 2004 A1
20040105889 Ryde et al. Jun 2004 A1
20040126372 Banerjee et al. Jul 2004 A1
20040126373 Banerjee et al. Jul 2004 A1
20040131614 Banerjee et al. Jul 2004 A1
20040136989 Banerjee et al. Jul 2004 A1
20040136990 Banerjee et al. Jul 2004 A1
20040136991 Banerjee et al. Jul 2004 A1
20040151722 Banerjee et al. Aug 2004 A1
20040156835 Imoto et al. Aug 2004 A1
20040166111 Kaymakcalan et al. Aug 2004 A1
20040170623 Arvinte et al. Sep 2004 A1
20040191243 Chen et al. Sep 2004 A1
20040197324 Liu et al. Oct 2004 A1
20040213785 Yamazaki et al. Oct 2004 A1
20040219142 Banerjee et al. Nov 2004 A1
20040228913 Kumar et al. Nov 2004 A1
20050053598 Burke et al. Mar 2005 A1
20050118163 Mizushima et al. Jun 2005 A1
20050118167 Okada et al. Jun 2005 A1
20050119172 Merkle Jun 2005 A1
20050214278 Kakuta et al. Sep 2005 A1
20060009385 Hoffman et al. Jan 2006 A1
20060083741 Hoffman et al. Apr 2006 A1
20060088523 Andya et al. Apr 2006 A1
20060115472 Li et al. Jun 2006 A1
20060142549 Takeda et al. Jun 2006 A1
20060149042 Konstantinov et al. Jul 2006 A1
20060153846 Krause et al. Jul 2006 A1
20060159653 Saito et al. Jul 2006 A1
20060182740 Yang et al. Aug 2006 A1
20060210557 Luisi et al. Sep 2006 A1
20060246073 Knight et al. Nov 2006 A1
20060269479 Colton et al. Nov 2006 A1
20060269543 Chu Nov 2006 A1
20070020255 Ueno et al. Jan 2007 A1
20070031402 Zhang et al. Feb 2007 A1
20070036779 Bardat et al. Feb 2007 A1
20070041905 Hoffman et al. Feb 2007 A1
20070053871 Li et al. Mar 2007 A1
20070053900 Liu et al. Mar 2007 A1
20070053906 Samaritani et al. Mar 2007 A1
20070065440 Tomlinson et al. Mar 2007 A1
20070065567 Segall et al. Mar 2007 A1
20070071747 Hoffman et al. Mar 2007 A1
20070081996 Hoffman et al. Apr 2007 A1
20070122402 Bolli et al. May 2007 A1
20070172475 Matheus et al. Jul 2007 A1
20070172897 Maksymowych et al. Jul 2007 A1
20070184045 Doctor et al. Aug 2007 A1
20070184050 Ishikawa et al. Aug 2007 A1
20070190047 Brych et al. Aug 2007 A1
20070197439 Zhu et al. Aug 2007 A1
20070202051 Schuschnig Aug 2007 A1
20070202104 Banerjee et al. Aug 2007 A1
20070237762 Winter Oct 2007 A1
20070244299 Jaber Oct 2007 A1
20070253984 Khandke et al. Nov 2007 A1
20070269463 Donovan Nov 2007 A1
20070292442 Wan et al. Dec 2007 A1
20080003220 Gokarn Jan 2008 A1
20080016123 Devraj et al. Jan 2008 A1
20080071063 Allan et al. Mar 2008 A1
20080112953 McAuley et al. May 2008 A1
20080118496 Medich et al. May 2008 A1
20080124326 Rehder et al. May 2008 A1
20080131374 Medich et al. Jun 2008 A1
20080139792 Sek et al. Jun 2008 A1
20080145383 Zauner et al. Jun 2008 A1
20080166348 Kupper et al. Jul 2008 A1
20080193466 Banerjee et al. Aug 2008 A1
20080200655 Sek et al. Aug 2008 A1
20080227136 Pla et al. Sep 2008 A1
20080275220 Friess et al. Nov 2008 A1
20080280345 Turner et al. Nov 2008 A1
20080286280 Kallmeyer et al. Nov 2008 A1
20080311043 Hoffman et al. Dec 2008 A1
20080311078 Gokarn et al. Dec 2008 A1
20090017472 Stuhlmuller et al. Jan 2009 A1
20090028794 Medich et al. Jan 2009 A1
20090054331 Chen et al. Feb 2009 A1
20090068172 Kaymakcalan et al. Mar 2009 A1
20090110679 Li et al. Apr 2009 A1
20090123378 Wong et al. May 2009 A1
20090148406 Jezek Jun 2009 A1
20090148513 Fraunhofer et al. Jun 2009 A1
20090155205 Salfeld et al. Jun 2009 A1
20090226530 Lassner et al. Sep 2009 A1
20090239259 Hsieh Sep 2009 A1
20090258018 Medich et al. Oct 2009 A1
20090271164 Peng et al. Oct 2009 A1
20090280065 Willian et al. Nov 2009 A1
20090291062 Fraunhofer et al. Nov 2009 A1
20090304682 Hoffman et al. Dec 2009 A1
20090317399 Pollack et al. Dec 2009 A1
20100003243 Okun et al. Jan 2010 A1
20100016557 Salfeld et al. Jan 2010 A1
20100021451 Wong Jan 2010 A1
20100028372 Jezek Feb 2010 A1
20100028383 van Gelder et al. Feb 2010 A1
20100034823 Borhani et al. Feb 2010 A1
20100040604 Salfeld et al. Feb 2010 A1
20100040630 Elden et al. Feb 2010 A1
20100113744 Tsvetkova et al. May 2010 A1
20100129379 Carpenter et al. May 2010 A1
20100160894 Julian et al. Jun 2010 A1
20100172862 Correia et al. Jul 2010 A1
20100278822 Fraunhofer et al. Nov 2010 A1
20110002935 Wan et al. Jan 2011 A1
20110014676 Cowan et al. Jan 2011 A1
20110054414 Shang et al. Mar 2011 A1
20110171227 Okun et al. Jul 2011 A1
20110236391 Mahler et al. Sep 2011 A1
20110278822 Lungershausen et al. Nov 2011 A1
20110300151 Okun et al. Dec 2011 A1
20120014956 Kupper et al. Jan 2012 A1
20120028877 Gokarn Feb 2012 A1
20120039900 Stuhlmuller et al. Feb 2012 A1
20120077213 Pla et al. Mar 2012 A1
20120107783 Julian et al. May 2012 A1
20120129185 Maksymowych et al. May 2012 A1
20120171123 Medich et al. Jul 2012 A1
20120177596 Fischkoff et al. Jul 2012 A1
20120258114 Salfeld et al. Oct 2012 A1
20120263731 Fraunhofer et al. Oct 2012 A1
20120282262 Okun et al. Nov 2012 A1
20120282270 Krause et al. Nov 2012 A1
20130004507 Fischkoff et al. Jan 2013 A1
20130028903 Wan et al. Jan 2013 A1
20130122011 Hoffman et al. May 2013 A1
20130156760 Fraunhofer et al. Jun 2013 A1
20130243786 Banerjee et al. Sep 2013 A1
20130273066 Gokarn et al. Oct 2013 A1
20130273067 Gokarn Oct 2013 A1
20140141007 Fraunhofer et al. May 2014 A1
20140141008 Fraunhofer et al. May 2014 A1
20140206526 Gao et al. Jul 2014 A1
20140377275 Neu et al. Dec 2014 A1
20150023977 Fraunhofer et al. Jan 2015 A1
20150071944 Correia et al. Mar 2015 A1
20150361170 Fraunhofer et al. Dec 2015 A1
20160017030 Neu et al. Jan 2016 A1
20160222101 Fraunhofer et al. Aug 2016 A1
20160319011 Gokarn Nov 2016 A1
20160367665 Gokarn Dec 2016 A1
20170143828 Fraunhofer et al. May 2017 A1
20190211093 Neu et al. Jul 2019 A1
20190358322 Fraunhofer et al. Nov 2019 A1
20210070850 Bachmann et al. Mar 2021 A1
Foreign Referenced Citations (78)
Number Date Country
2904458 Jun 2009 CA
41925 Dec 1981 EP
0486526 May 1992 EP
0419251 Sep 1993 EP
0893450 Jan 1999 EP
1174148 Jan 2002 EP
1336410 Aug 2003 EP
1712240 Oct 2006 EP
2637690 Sep 2013 EP
3189831 Jul 2017 EP
3479819 May 2019 EP
3684407 Jul 2020 EP
07236483 Sep 1995 JP
02942412 Aug 1999 JP
WO 1989011298 Nov 1989 WO
WO 1990001191 Feb 1990 WO
WO 1990011091 Oct 1990 WO
WO 1992002616 Feb 1992 WO
WO 1993008837 May 1993 WO
WO 1995003826 Feb 1995 WO
WO 1997004801 Feb 1997 WO
WO 1997029131 Aug 1997 WO
WO 1997045140 Dec 1997 WO
WO 1998001555 Jan 1998 WO
WO 1998022136 May 1998 WO
WO 1998042376 Oct 1998 WO
WO 1998044948 Oct 1998 WO
WO 1998056418 Dec 1998 WO
WO 1999037329 Jul 1999 WO
WO 1999064462 Dec 1999 WO
WO 2000067789 Nov 2000 WO
WO 2001047554 Jul 2001 WO
WO 2002012500 Feb 2002 WO
WO 2002012502 Feb 2002 WO
WO 2002013860 Feb 2002 WO
WO 2002030463 Apr 2002 WO
WO 2002043695 Jun 2002 WO
WO 2002043750 Jun 2002 WO
WO 2002051979 Jul 2002 WO
WO 2002057979 Jul 2002 WO
WO 2002064166 Aug 2002 WO
WO 2002072636 Sep 2002 WO
WO 2002096457 Dec 2002 WO
WO 2003002713 Jan 2003 WO
WO 2003009817 Feb 2003 WO
WO 2003053471 Jul 2003 WO
WO 2003102132 Dec 2003 WO
WO 2004001007 Dec 2003 WO
WO 2004007520 Jan 2004 WO
WO 2004016286 Feb 2004 WO
WO 2004024752 Mar 2004 WO
WO 2004039337 May 2004 WO
WO 2004043750 May 2004 WO
WO 2004050059 Jun 2004 WO
WO 2004055164 Jul 2004 WO
WO 2004066957 Aug 2004 WO
WO 2004102184 Nov 2004 WO
WO 2005072772 Aug 2005 WO
WO 2006012500 Feb 2006 WO
WO 2006014965 Feb 2006 WO
WO 2006031560 Mar 2006 WO
WO 2006064373 Jun 2006 WO
WO 2006081320 Aug 2006 WO
WO 2006138181 Dec 2006 WO
WO-2006138181 Dec 2006 WO
WO 2007003936 Jan 2007 WO
WO 2007074880 Jul 2007 WO
WO 2007095337 Aug 2007 WO
WO 2008015419 Feb 2008 WO
WO 2008108927 Sep 2008 WO
WO 2009015367 Jan 2009 WO
WO 2009073569 Jun 2009 WO
WO 2012065072 May 2012 WO
WO 2014039903 Mar 2014 WO
WO 2018004260 Jan 2018 WO
WO 2018162500 Sep 2018 WO
WO 2019057631 Mar 2019 WO
WO 2020187760 Sep 2020 WO
Non-Patent Literature Citations (260)
Entry
Andya et al., “Mechanism of Aggregate Formation and Carbohydrate Excipient Stabilization of Lyophilized Humanized Monoclonal Antibody Formulations”, AAPS Pharm Sci. 5:1-11 (2003).
Betaseron label, 2003.
Chang et al., “Effect of Sorbitol and Residual Moisture on the Stability of Lyophilized Antibodies: Implications for the Mechanism of Protein Stabilization in the Solid State”, Pharm Sci. 94:1445-1455 (2005).
Copaxone label, 2006.
DeFelippis and Akers, “Excipient Selection”, Chapter 7.4 and 8.2.1 in Pharmaceutical Formulation Development of Peptides, edited by Frokjaer, S and Hovgaard, L. (2000).
Glenski et al., “29 Gauge Needles Improve Patient Satisfaction over 27 Gauge Needles for Daily Glatiramer Acetate Injections”, Drug Healthcare and Patient Safety 81-86 (2009).
Paborji et al., “Chemical and Physical Stability of Chimeric 16, a Mouse-Human Monoclonal Antibody”, Pharm Res. 11:764-771 (1994).
Product Monograph for Humira® dated Jun. 25, 2019.
Abbott “Humira Prescribing Info.” Jan. 31, 2003.
Adalimumab entry from National Library of Medicine website: www.nlm.nih.gov/cgi/mesh; printed on Sep. 28, 2009.
Akers, et al., “Development and Manufacture of Protein Pharmacewuticals (Pharmaceutical Biotechnology)”, Chapter 2: “Formulation Development of Protein Dosage Forms”, 2002, Kluver Academic/Plenum, pub., New York, p. 47-127.
Antoni et al., “Side effects of anti-TNF therapy: Current knowledge,” Clin Exp Rheumatol 2002; 20(suppl. 28):S-152-S-157.
Barrera, et al., “Effects of treatment with a fully human antitumour necrosis factor alpha monoclonal antibody on the local and systemic homeostasis of interleukin 1 and TNFalpha in patients with rheumatoid arthritis,” Ann Rheum. Dis. 2001, 60(7):660-669.
Cada et al. “Adalimumab”, Hospital Pharmacy (2003) 38,6:568-580.
Carpenter & Manning, eds., Rational Design of Stable Protein Formulations, Theory and Practice, Pharmaceutical Biotechnology vol. 13, Kluwer Academic/Plenum Publishers: New York (2002).
Carpenter, et al., “Rational Design of Stable Lyophilized Protein Formulations: Some Practical Advice,” Pharmaceurtical Research, 1997, vol. 14(8):969-975.
Chi, et al., “Physical Stability of Proteins in Aqueous Solution: Mechanism and Driving Forces in Nonnative Protein Aggregation,” Pharm. Res. 20(9), 1325-1336 (Sep. 2003).
Christy et al., “High-performance tangential flow filtration: a highly selective membrane separation process”, Desalination, 144:133-136 (2002).
Cleland et al., The Development of Stable protein Formulations: A Close Look at Protein Aggregation, Deamidation, and Oxidation, 10 Crit. Rev. Ther. Drug Carrier Syst. 10, 307-377 (1993).
Cleland, Jeffrey L. et al., “A Specific Molar Ratio of Stabilizer to Protein is Required for Storage Stability of a Lyophilized Monoclonal Antibody,” Journal of Pharmaceutical Sciences, 2001, vol. 90(3): 310-321.
Daugherty et al. “Formulation and Delivery Issues For Monoclonal Antibody Therapeutics,” Advanced Drug Delivery Reviews (2006) 58:686-706.
Den Broeder et al. Long term anti-tumour necrosis factor a monotherapy in rheumatoid arthritis: effect on radiological course and prognostic value of markers of cartilage turnover and endothelial activation, Ann Rheum Dis, 2002;61:311-318.
EMEA Report “Assessment Report for Simponi EMEA/H/C/000992”, Mar. 3, 2008, pp. 1-70.
Fesinmeyer et al. “Effect of ions on agitation- and temperature-induced aggregation; reactions of antibodies.” Pharm Res. Apr. 2009:26(4):903-13.
Garidal et al. “A rapid, sensitive and economical assessment of monoclonal antibody conformational stability by intrinsic tryptophan fluorescence spectroscopy.” Biotechnol J.2008, 3, pp. 9-10.
Gatlin & Gatlin, “Formulation and Administration Techniques to Minimize Injection Pain and Tissue 1999 Damage Associated with Parenteral Products,” Chapter 17 of Injectable Drug Development: Techniques to Reduce Pain and Irritation (Eds. Gupta & Brazeau).
Gokarn, et al., “Self-Buffering Antibody Formulations,” J. Pharm. Sci. 97(8), 3051-3066 (Aug. 2008).
Harris et al., “Commercial manufacturing scale formulation and analytical characterization of therapeutic recombinant antibodies”, Drug Development Research (2004) vol. 61(3): 137-154.
Helms, et al., “Destabilizing Loop Swaps in the CDRs of an Immunoglobulin VL Domain,” Protein Sci. 4, 2073-2081 (1995).
Hillgren, et al., “Protection mechanism of Tween 80 during freeze-thawing of a model protein,” International Journal of Pharmaceutics, 2002, vol. 237: 57-69.
Holt, et al., “Domain antibodies: proteins for therapy,” Trends in Biotechnology, 2003, vol. 21(11):484-490.
Hovgaard & Frokjaer (eds) “Pharmaceutical Formulation Development of Peptides and Proteins”, CRC Press 1999.
Humira Product label Jul. 30, 2004.
Humira® Label (Jan. 2003) (“Humira® Label”).
International Preliminary Examination Report for Application No. PCT/IB03/04502, dated Feb. 14, 2005.
International Preliminary Examination Report for Application No. PCT/US2008/085066, dated 2009.
International Search Report for Application No. PCT/IB03/04502 dated May 26, 2004.
International Search Report for Application No. PCT/US11/060388 dated May 30, 2012.
International Search Report for Application No. PCT/US2008/085066, dated 2009.
Kamerzell, et al., “Increasing IgG Concentration Modulates the Conformational Heterogeneity and Bonding Network that Influence Solution Properties,” J. Phys. Chem. B. 113(17), 6109-6118 (2009).
Koticha et al., “Rapid Antibody Purification Using Ultrafiltration and Centrifugal Affinity Columns,” BioscienceTechnologies.com, http://www.biosciencetechnology.com/article/2006/07/rapid-antibody-purification-usingultrafiltration-and-centrifugal-affinity-columns.
Krishnamurthy et al., The Stability Factor: Importance in Formulation Development, 3 Curr. Pharm. Biotech. 3, 361-371 (2002).
Laursen et al, “Pain perception after subcutaneous injections of media containing different buffers,” Basic Clin. Pharmacol Toxicol. Feb. 2006; 98:218-21.
Lee, et al., “Toward Aggregation-resistant Antibodies by Design,” Trends in Biotech. 31(11), 612-620 (2013).
Li et al. “Resurrecting Abandoned Proteins with Pure Water: CD and NMR Studies of Protein Fragments Solubilized in Salt-Free Water,” Biophys J 91:4201-4209 (2006).
Liu, J. et al., “Reversible self-association increases the viscosity of a concentrated monoclonal antibody in aqueous solution,” J. Pharm.Sci., vol. 94(9) 1928-1940 (Sep. 2005).
Mahler, et al., “Induction and analysis of aggregates in a liquid IgG1-antibody formulation,” Eur. J. Pharm. Biopharm., 2005, 59(3): 407-17.
Manning et al., “Stability of Protein Pharmaceuticals,” Pharm Res 6:903-918 (1989).
Millipore, “Protein Concentration and Diafiltration by Tangential Flow Filtration” http://wolfson.huji.ac.il/purification/PDF/dialysis/ MILLIPORE TFF.pdf, copyright date 2003 (pp. 1-24).
Paborij et al., “Chemical and Physical Stability of Chimeric L6, a Mouse-Human Monoclonal Antibody,” Pharm. Res. 1994; 11, 5:764-771.
Pall Life Sciences, “Desalting and Buffer Exchange by Dialysis, Gel Filtration or Diafiltration”, Pall Life Sciences Scientific and Technical Report, 2003, p. 1-4.
Pennington et al., “Polyclonal and Monoclonal Antibody Therapy for Experimental Pseudomonas aeruginosa Pneumonia”, Infect. Immun. (1986) vol. 54, p. 239-244.
Post-Filing Date Evidence submitted in EP 03 748 438.
Post-Filing Date Evidence submitted to the European Patent Office in EP 03 748 439 on May 15, 2009.
Presentation at AAPS National Biotechnology Conference, Jun. 24-27, 2007, San Diego Convention Center, by R. Matthew Fesinmeyer, Ph.D.
Rouet, et al., “Stability Engineering of the Human Antibody Repertoire,” FEBS Letters 588, 269-277 (2014).
Salinas, et al., “Understanding and Modulating Opalescence and Viscosity in a Monoclonal Antibody Formulation,” J. Pharm. Sci. 99(1), 82-93 (2010).
Saluja et al., “Ultrasonic Storage Modulus as a Novel Parameter for Analyzing Protein-Protein Interactions in High Protein Concentration Solutions: Correlation with Static and Dynamic Light Scattering Measurements,” Biophysical Journal, 92 234-244. Jan. 2007.
Saluja, “Characterization of Protein-Protein Interactions for Optimizing Formulation and Physical Stability of High Protein Concentration Solutions” Jan. 2007.
Sarciaux et al., “J Effects of buffer composition and processing conditions on aggregation of bovine IgG during freeze-drying”, Journal of Pharmaceutical Sciences (1999) 88(12):1354-61.
Schule et al. “Conformational analysis of protein secondary structure during spray-drying of; antibody/mannitol formulations.” Eur J Pharm Biopharm. Jan. 2007; 65(1):1-9.
Schwartz, “Diafiltration for Desalting or Buffer Exchange,” BioProcess International, May 2003 pp. 43-49.
Shimazato et al., “Suppression of Tumor Necrosis Factor Alpha Production by a Human Immunoglobulin Preparation for Intravenous Use”, Infect. Immun. (1990), vol. 58, p. 1384-1390.
Shire et al., “Challenges in the Development of High Protein Concentration Formulations,” J Pharm Sci 93:1390-1402 (2004).
Shire, “Formulation of Proteins and Monoclonal Antibodies (mAbs),” Monoclonal Antibodies, Meeting the Challenges In Manufacturing, Formulation, Delivery and Stability of Final Drug Product, Woodhead Publishing Series in Biomedicine 77, Chap. 4, 93-120 (Woodland Publishing, Cambridge, UK) (2015).
Sivasai et al., “Cytomegalovirus immune globulin intravenous (human) administration modulates immune response to alloantigens in sensitized renal transplant candidates”, Clin. Exp. Immunol., (2000), vol. 119, p. 559-565.
Szenczi et al. “The effect of solvent environment on the conformation and stability of; human polyclonal IgG in solution” Biologicals Mar. 2006; 34(1):5-14.
Tian et al. “Spectroscopic evaluation of the stabilization of humanized monoclonal antibodies in amino acid formulations,” Intl J Pharm 355:20-31(2007).
Trastuzumab (Herceptin®) Product Insert (2006).
U.S. Appl. No. 60/690,582, filed Jun. 14, 2005 (Gokarn Provisional).
Van de Putte et al. “Efficacy and safety of the fully human anti-tumour necrosis factor alpha monoclonal antibody adalimumab (D2E7) in DMARD refractory patients with rheumatoid arthritis: a 12 week, phase II study,” Ann Rheum Dis 2003;62:1168-1177.
Varasteh et al., Optimization of anti-Rh D immunoglobulin stability in the lyphilization process. Iranian Journal of Basic Medical Sciences, 2008, vol. 11, No. 1, pp. 55-61.
Veys et al., “Pain at the injection site of subcutaneously administered erythropoietin: phosphate-buffered epoetin alpha compared to citrate-buffered epoetin alpha and epoetin beta,” Clinical Nephrology, 49:41-44 1998.
Vidanovic et al: “Effects of nonionic surfactants on the physical stability of immunoglobulin; G in aqueous solution during mechanical agitation.” Pharmazie. Jun. 2003; 58(6):399-404.
Voigt, “Textbook of pharmaceutical technology” VCH, 384, 394, 395 (1987).
Wang et al. “Instability, Stabilization, and Formulation of Liquid Protein Pharmaceuticals,” Int'l J. Pharmaceutics 1999; 185:129-188.
Wang et al.“Antibody Structure, Instability, and Formulation,” J Pharmaceutical Sci (2007) 96:1, 1-26.
Wang et al: “Opalescence of an IgG1 Monoclonal Antibody Formulation is Mediated by Ionic Strength and Excipients”, BioPharm Internatl, vol. 22(4) (2009).
Wang, “Lyophilization and development of solid protein pharmaceuticals,” Int J Pharm 2000; (203), 1-2:1-60.
Weinblatt et al. “Adalimumab, a Fully Human Anti-Tumor Necrosis Factor Monoclonal Antibody, for the Treatment of Rheumatoid Arthritis in Patients Taking Concomitant Methotrexate”, Arthritis & Rheumatism. 2003; 48(1):35-45.
Wolfgang Fraunhofer Dissertation Thesis. Ludwig-Maximilians University, München, “Asymmetrical Flow Field-Flow-Fractionation in Pharmaceutical Analytics—Investigations in Aggregation Tendencies of Pharmaceutical Antibodies,” Posted May 7, 2008.
Zhao et al. “Recent U.S. Patents on Protein Drug Formulation: 2000-2007” Protein Drug Formulation 2008.
AbbVie Biotechnology Ltd., “Annex A—The Humira® Story,” in Opposition Proceeding for EP1406656 (filed on Dec. 22, 2014).
Alberts, Molecular Biology of the Cell, Fourth Edition, New York, NY: Garland Publishing, Inc., pp. 1363-1384 (2002).
Amersham Biosciences, “Ion exchange chromatography: Principles and methods” 1-157 (1999) [Substitute].
Andrew and Titus, Curr. Protoc. Cell Biol., Chapter 16:Unit 16.4, pp. 16.4.1-16.4.10 (2000).
Arakawa et al., Protein Expr. Purif. 36:244-248 (2004).
Avonex® (interferon beta-1a) Label, 1-23 (2001).
Avonex® (interferon beta-1a), Physicians' Desk Reference, PDR 57, pp. 1006-1010 (2003).
Bexxar® (tositumomab and iodine I 131 tositumomab), Physicians' Desk Reference, PDR 60, pp. 1360-1366 (2006).
Bjorck et al., J. Immunol. 133:969-74 (1984).
Boes, Mol. Immunol., 37:1141-1149 (2000).
Campath® (alemtuzumab) Label, 1-14 (2004).
Campath® (alemtuzumab), Physicians' Desk Reference, PDR 57, pp. 975-977 (2003).
Carpenter & Manning, eds., Rational Design of Stable Protein Formulations, Theory and Practice, Pharmaceutical Biotechnology vol. 13, Kluwer Academic/Plenum Publishers: New York (2002). (Full text).
Carter, Nat. Rev. Cancer 1:118-129 (2001).
“Clinical Pharmacology and Biopharmaceutics Review(s),” by Center for Drug Evaluation and Research and Center for Biologics Evaluation and Research, Application No. 125057/0, in Approval Package for Humira® (Approved Dec. 31, 2002).
Davies and Metzger, Annu. Rev. Immunol. 1:87-117 (1983).
“Development Pharmaceutics for Biotechnological and Biological Products (Annex to Note for Guidance on Development Pharmaceutics),” by Committee for Proprietary Medicinal Products, The European Agency for the Evaluation of Medicinal Products (Oct. 21, 1999).
DiJoseph et al., Blood 103:1807-1814 (2004).
Egan, Arzneimittelforschung 49:779-790 (1999).
English translation of Octagam® entry (75 008) in Rote Liste 2005 (Cantor Publishers 2005).
Gronski et al., On the Nature of IgG Dimers: I. Dimers in Human Polyclonal IgG Preparations: Kinetic Studies, 82 Behring. Inst. MITT.127 (1988).
Hardcastle, J. Chem. Ed. 58:726 (1981) [Substitute].
Harinayaran et al., Biotech. Bioeng. 102:1718-1722 (2009).
Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, pp. 1-22 (1988).
Herceptin® (trastuzumab) Label, 1-28 (2002).
Herceptin® (trastuzumab), Physicians' Desk Reference, PDR 57, pp. 1399-1402 (2003).
Humblet, Expert Opin. Pharmacother., 5:1621-1633 (2004).
Hypermol, “Dialysis: an introduction,” Dialysis: Technical Datasheet, 1-2 (2008).
“Injection Tips,” Humira.com, http://www.humira.com/hu/hustore/cgi-bin/ProdSubEV_Cat_205043_SubCat_210170_NavRoot_205042_NavID_301.htm [http://web.archive.org/web/20050317083331/http:// www.humira.com /hu/hustore/cgi-bin/ProdSubEV_Cat_205043_SubCat_210170_NavRoot_205042_NavID_301.htm (Archived Mar. 17, 2005).
IPR2017-00822, Institution Decision Denying Institution of Inter Partes Review, Sep. 7, 2017.
IPR2017-00823, Institution Decision Denying Institution of Inter Partes Review, Sep. 7, 2017.
IPR2017-01008, Institution Decision Denying Institution of Inter Partes Review, Sep. 7, 2017.
IPR2017-01009, Institution Decision Denying Institution of Inter Partes Review, Sep. 7, 2017.
Kotz and Treichel, Chem. & Chem. Reactivity, Chapter 18:Unit 18.3, pp. 851-858 (1999).
Kurnik et al., Biotechnol. Bioeng. 45:149-157 (1995).
Lide, Handbook of Chemistry and Physics, 81st ed., Boca Raton, FL: CRC Press., pp. 7-1, 8-44 to 8-56 (2000).
Meyssami et al., Biotechnol. Prog. 8:149-154 (1992).
Mohan, “Buffers: A guide for the preparation and use of buffers in biological systems,” Calbiochem, 1-32 (2003).
Parham et al., J. Immunol. Methods 53:133-173 (1982).
Patent Interference No. 106,057, Decision on Motions, Sep. 27, 2017.
Patent Interference No. 106,057, Judgement, Dec. 20, 2017.
Pierce Biotechnology, Inc., “Dialysis; an overview,” Technical Resource, 1-2 (2004).
Piper and Fenton, Gut 6:506-508 (1965).
Raptiva® (efalizumab), Physicians' Desk Reference, PDR 59, pp. 1350-1354 (2005).
Raptiva® (efalizumab) Label, 1-34 (2003).
Rousseaux et al., J. Immunol Methods 64:141-146 (1983).
Segel, Biochemical calculations, 2nd ed., New York: John Wiley and Sons, Inc., pp. 1-85 (1976).
Sellers et al., J. Pharm. Sci. 90:785-797 (2001).
Shire, Monoclonal Antibodies, Meeting the Challenges In Manufacturing, Formulation, Delivery and Stability of Final Drug Product, Woodhead Publishing Series in Biomedicine 77 (Woodland Publishing, Cambridge, UK) (2015) (Full text).
Tayyab et al., Biochemical Education, 19:149-152 (1991).
Vermeer and Norde, Biophys. J. 78:394-404 (2000).
Wang and Goodman, Basic & Clinical Immunology, ed. Fudenberg, Stites, Caldwell and Wells, Los Altos, CA: Lange Medical Publication, pp. 15-40 (1976).
Xolair® (omalizumab) Label, 1-17 (2003).
Xolair® (omalizumab), Physicians' Desk Reference, PDR 58, pp. 1374-1376 (2004).
Yang et al., Crit. Rev. Oncol. Hematol. 38:17-23 (2001).
Zeman and Zydney, Microfiltration and Ultrafiltration: Principles and Applications, pp. 380-396; 544-564, Marcel Dekker, Inc., New York (1996).
AbbVie, Inc. “Highlights of Prescribing Information” (Humira Label) Mar. 1, 2017: Retrieved from the Internet: URL: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125057s399lbl.pdf.
Adalimumab Product Approval Information, http://www.fda.gov/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/ApprovalApplications/TherapeuticBiologicApplications/ucm080610.htm (accessed Jan. 23, 2017).
Amgen “Highlights of Prescribing Information” Amjevita, FDA (2016): URL: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/761024lbl.pdf.
Avastin™ Label (Feb. 2004).
Bam et al., Tween Protects Recombinant Human Growth Hormone against Agitation-Induced Damage via Hydrophobic Interactions, 87 J. Pharm. Sci. 1554 (1998).
Banks et al., “Native-State Solubility and Transfer Free Energy as Predictive Tools for Selecting Excipients to Include in Protein Formulation Development Studies”, 101 J. Pharm. Sci. 2720 (2012).
Barnett, et al., “Reduction of Pain and Local Complications When Buffered Lidocaine Solution Is Used as a Local Anesthetic in Conjunction with Hyperthermia Treatments: Results of a Randomized Trial,” Int'l J. Radiation Oncology Biol. Phys. 23(3), 585-591 (1992).
Bekker et al., J. Bone Miner. Res. 19:1059-1066 (2004).
Berg et al., “Exploring Proteins” in Biochemistry, 5th edition, pp. 77-116, New York, W H Freeman (2002).
Bexxar® (tositumomab and iodine I 131 tositumomab) Label, 1-49 (2003).
Binabaji et al., Biotech. Prog. 32:692-701 (2016).
Brazeau, et al., “Current Perspectives on Pain upon Injection of Drugs,” J. Pharm. Sci. 87(6), 667-677 (Jun. 1998).
Burton, D.R. et al., Aspects of the Molecular Structure of IgG Subclasses, 19 Monogr. Allergy 7-35 (1986).
Butler & Hamilton, “Quantitation of Specific Antibodies: Methods of Express, Standards, Solid-Phase Considerations, and Specific Applications,” Ch. 9 in Immunochemistry of Solid-Phase Immunoassay, CRC Press (John E. Butler ed., 1991).
Campath® Label (Aug. 2006).
Carnahan et al., “Epratuzumab, a Humanized Monoclonal Antibody Targeting CD22: Characterization of In Vitro Properties,” Clin. Cancer Res., 9:3982s-90s (2003).
Carpenter et al., “Chapter 7: Freezing- and Drying-Induced Perturbations of Protein Structure and Mechanisms of Protein Protection by Stabilizing Additives”, in Freeze-Drying/Lyophilization of Pharmaceutical and Biological Products 167 (2d ed. 2004).
Carpenter et al., Inhibition of Stress-Induced Aggregation of Protein Therapeutics, 309 Methods in Enzymology 236 (1999).
Castellano et al, The Oncologist 16:136-145 (2011).
Chen et al., Pharm. Res. 20:1952-60 (2003).
Chen, et al., “Aggregation Pathway of Recombinant Human Keratinocyte Growth Factor and Its Stabilization,” Pharm. Res. 11(11), 1581-1587 (1994).
Chong and Wong, Clin. Immunol. 123:129-138 (2007).
Christensen, “Proteins as buffers,” Annals of the New York Academy of Sciences, 133:34-40 (Apr. 1966).
Cleland & Langer, “Formulation and Delivery of Proteins and Peptides: Design and Development Strategies,” Ch. 1 in Formulation and Delivery of Proteins and Peptides, ACS Symposium Series 567, 1-19 (1994).
CNJ-016 (Vaccinia Immune Globulin Intravenous) Label (Jan. 2010).
Dani et al., Pharmaceutics, Preformulation and Drug Delivery: High Concentration Formulation Feasibility of Human Immunoglobulin G for Subcutaneous Administration, 86 J. Pharm. Sci. 1504 (2007).
Dean, “Lange's Handbook of Chemistry,” McGraw-Hill, p. 8.49, 8.65 (9th ed. 1999).
Enbrel® Label (Nov. 1998).
Enbrel® Label (Sep. 2002).
Fayos, et al., “On the Origin of the Thermostabilization of Proteins Induced by Sodium Phosphate,” J. Am. Chem. Soc. 127(27), 9690-9691 (2005).
FDA CDER Application No. 125057/0 “Clinical Pharmacology and Biopharmaceutics Review(s),” by Center for Drug Evaluation and Research and Center for Biologics Evaluation and Research, in Approval Package for Humira® (Approved Dec. 31, 2002).
Flebogamma® Label (Jan. 2004).
Fransson & Espander-Jansson, “Local Tolerance of Subcutaneous Injections,” J. Pharm. Pharmacol., 48:1012-1015 (1996).
Frenken et al., “Identification of the Component Part in an Epoetin Alfa Preparation that Causes Pain after Subcutaneous Injection,” American J. of Kidney Diseases, 22(4): 553-556 (1993).
Gammagard Liquid® Label (Apr. 2005).
Gamunex® Label (Nov. 2005).
Gebhart, “Biotech Company Preparing Several Drugs for Takeoff,” Drug Topics, vol. 145, No. 5, p. 50 (Mar. 5, 2001).
Gelfand, “Differences Between IGIV Products: Impact on Clinical Outcome,” Int'l Immunopharmacology, 6:592-99 (2006).
Gokarn et al., “Excipients for Protein Drugs,” Ch. 17 in Excipient Development for Pharmaceutical, Biotechnology, and Drug Delivery Systems (Ashok Katdare & Mahesh V. Chaubal eds., 2006).
Gottlieb, “Efficacy and Safety of Anti-TNF-α Agents in Psoriasis,” in Anti-TNF-α Therapies in the Treatment of Dermatologic Diseases at 6 (2005) (Supplement to Skin & Allergy News; Produced in Affiliation with the Skin Disease Education Foundation's 29th Annual Hawaii Dermatology Seminar).
Granolleras et al., “Experience of Pain After Subcutaneous Administration of Different Preparations of Recombinant Human Erythropoietin: A Randomized, Double-Blind Crossover Study,” Clinical Nephrology, 36:294-298 (1991).
Handbook of Pharmaceutical Excipients, Pharmaceutical Press (Raymond C. Rowe, Paul J. Sheskey, & Siân C. Owen eds., 5th ed. 2006).
Hanna, The IGIV-C Study Group, “Tolerability of a New Intravenous Immunoglobulin Preparation (IGIV) in Pediatric and Adult Patients,” presented at the 60th Anniversary Meeting of the American Academy of Allergy, Asthma & Immunology (Mar. 10, 2003), in J. Allergy Clinical Immunology, vol. 111, No. 2, part 2, a631.
HepaGam B™ Summary Basis for Approval (Jan. 2006).
Humira® (adalimumab) Label, 1-16 (2002).
Humira® (adalimumab), Physicians' Desk Reference, PDR 58, pp. 470-474 (2004).
Humira® Label (Oct. 2005).
Humira® Label (Nov. 2006).
Humira® Label (Feb. 2007).
Humira® Label (Jan. 2008).
Humira® Label (Feb. 2008).
Humira® Label (Nov. 2015).
Humira® Label (Oct. 2016).
Humphreys, “Top 200 Medicines—Special Report,” Pharmalive (Aug. 12, 2015), http://www.pharmalive.com/special-report-top-200-medicines/.
Ipp, et al., “Adverse Reactions to Diphtheria, Tetanus, Pertussis-Polio Vaccination at 18 Months of Age: Effect of Injection Site and Needle Length,” Pediatrics 83(5): 679-682 (May 1989).
Ito, “Effects of subclass changes on the conformation, stability, and aggregation of antibodies: The case of chimeric, humanized, and human antibodies.” Thesis, Graduate School of Frontier Sciences, University of Tokyo, 2013.
Jefferis et al., “Recognition Sites on Human IgG for Fcγ Receptors: The Role of Glycosylation,” Immunology Letters, 44: 111-117 (1995).
Jorgensen, “Improvement of Patient Convenience in Treatment with Growth Hormone,” J. Pediatric Endocrinology 7(2), 175-180 (1994).
Jorgensen, et al., “Pain Assessment of Subcutaneous Injections,” Ann. Pharmacotherapy 30, 729-732 (Jul./Aug. 1996).
Kivitz et al., “Clinical Assessment of Pain, Tolerability, and Preference of an Autoinjection Pen Versus a Prefilled Syringe for Patient Self-Administration of the Fully Human, Monoclonal Antibody Adalimumab: The Touch Trial”, 28(10) Clin. Ther. (2006) 1619-29.
Krishnan et al., Chapter 16: Development of Formulations for Therapeutic Monoclonal Antibodies and Fc Fusion Proteins, in Formulation And Process Development Strategies For Manufacturing Biopharmaceuticals 383 (2010).
Kuzu, et al., “The Effect of Cold on the Occurrence of Bruising, Haematoma and Pain at the Injection Site in Subcutaneous Low Molecular Weight Heparin” Int'l J. Nursing Studies 38, 51-59 (2001).
Lipman, et al., “Monoclonal Versus Polyclonal Antibodies: Distinguishing Characteristics, Applications, and Information Resources,” ILAR J. 46(3), 258-268 (2005).
McCue et al., “Three Generations of Immunoglobulin G Preparations for Clinical Use,” Reviews of Infectious Diseases, 8:S374-81 (1986).
McDonnell, “Production of Antibodies in Hybridoma and Nonhybridoma Cell Lines,” Ch. 3 in Animal Cell Culture, Cell Engineering vol. 9, 65-88 (M. Al-Rubeai ed., 2015).
Meadows & Hollowell, “‘Off-label’ drug use: an FDA Regulatory Term, Not a Negative Implication of Its Medical Use,” Int'l J. Impotence Research 20:135-144 (2008).
Mezzasalma, et al., “Enhancing Recombinant Protein Quality and Yield by Protein Stability Profiling,” J. Biomolecular Screening 12(3), 418-428 (2007).
Nash, Randomized Crossover Comparison of Injection Site Pain with 40 mg/0.4 or 0.8 mL Formulations of Adalimumab in Patients with Rheumatoid Arthritis, Rhematol Ther 3:257-270 (2016).
Nishida et al., Characterization of novel murine anti-CD20 monoclonal antibodies and their omparison to 2B8 and c2B8 (rituximab), 31 Int'l. J. Oncology 29 (2007).
Nozaki & Tanford, “Examination of Titration Behavior,” Methods Enzymol., 11:715-734 (1962).
“Note for Guidance on Development Pharmaceutics,” by the Committee for Proprietary Medicinal Products (CPMP), The European Agency for the Evaluation of Medicinal Products (Jan. 28, 1998).
Octagam® Label (Mar. 2004).
Olthuis et al., “Characterization of Proteins by Means of their Buffer Capacity, Measured with an ISFET-based Coulometric Sensor-Actuator System,” Biosensors & Bioelectronics, 9:743-751 (1994).
Parslow, “Immunoglobulins & Immunoglobulin Genes,” Ch. 7 in Medical Immunology, Appleton & Lange (Daniel P. Stites, Abba I. Terr, & Tristram G. Parslow eds., 9th ed.1997).
Phillips and Signs, Curr. Protoc. Protein Sci., Unit 4.4, pp. 4.4.1-4.4.15, John Wiley & Sons, Inc. (2004).
Physicians' Desk Reference, pp. 470-474 (58th ed. 2004) (“2003 Humira® Label”).
Physicians' Desk Reference entry for Gamunex®, pp. 872-876 (59th ed. 2005).
Physicians' Desk Reference, pp. 558-559, 914-931, 805-807, 2026-2028, 2295-2297, 2524-2525 (56th ed. 2002).
Physicians' Desk Reference, pp. 925-928 (56th ed. 2002) (“Gamimune® Label”).
Press Release, “Amgen and Immunomedics Announce Emphasis on Development of AMG 412 (Epratuzumab) as Combination Therapy While Closing Single Agent Trial,” PRNewswire-FirstCall (Jan. 23, 2003).
Privigen® Label (Oct. 2016).
Raibekas, et al., “Anion Binding and Controlled Aggregation of Human Interleukin-1 Receptor Antagonist,” Biochemistry 4(29), 9871-9879 (2005).
Randolph & Carpenter, Engineering Challenges of Protein Formulations, 53 Am. Inst. Chem. Eng. J. 1902 (2007).
Remicade® (inflizimab) Label, 1-23 (2002).
Remicade® (inflizimab), Physicians' Desk Reference, PDR 57, pp. 1178-1182 (2003).
Remicade® Label (Aug. 1998).
Ruiz, et al., “Aggregation of Recombinant Human Interferon Alpha 2b in Solution: Technical Note,” AAPS Pharm. Sci. Tech. 7(4), Article 99, E1-E5 (2006).
Schwartz, “Diafiltration for Desalting of Buffer Exchange,” BioProcess Int'l (May 2003).
Schwartzman & Morgan, “Does Route of Administration Affect the Outcome of TNF Antagonist Therapy?,” Arthritis Research & Therapy, 6(Suppl. 2):S19-S23 (2004).
Shao and Zydney, Biotech. Bioeng. 87:286-292 (2004).
Stoner et al., “Protein-Solute Interactions Affect the Outcome of Ultrafiltration/Diafiltration Operations,” J. Pharm. Sci., 93:2332-2342 (2004).
Synagis® Label (Jul. 2004).
Thomson Reuters, “A Bioworld Special Report: Biosimilars: U.S. Market Opportunities and Critical Strategies 2016” (2016).
Tsourounis, Biologic Therapies for the Treatment of Chronic Plaque Psoriasis, Formulary 40:184-199 (Jun. 2005).
Tysabri® Label (Nov. 2004).
Uchiyama, “Liquid Formulation for Antibody Drugs,” Biochem. Phiophys. Acta 2041-2052 (2014).
Uchiyama, “Solution Properties of Antibody Drugs” Pharmacology, 74 (1), 12-18 (2014).
Valente et al., “Colloidal Behavior of Proteins: Effects of the Second Virial Coefficient on Solubility, Crystallization and Aggregation of Proteins in Aqueous Solution”, 6(6) Curr. Pharm. Biotechnol. (2005) 427-36.
Van de Putte, et al., A Single Dose Placebo Controlled Phase I Study of the Fully Human Anti-TNF Antibody D2E7 in Patients with Rheumatoid Arthritis, Arthritis Rheum., 41(9), S57 (Sep. 1998).
Van de Weert & Randolph, Chapter 6: Physical Instability of Peptides and Proteins, in Pharmaceutical Formulation Development Of Peptides And Proteins 107 (2012).
Van Reis and Zydney, “Protein Ultrafiltration,” in Encyclopedia of Bioprocess Technology: Fermentation, Biocatalysis, and Bioseparation, ed. by M.C. Flickinger and S.W. Drew, pp. 2197-2214, John Wiley & Sons, Inc. (1999) [Substitute].
Van Reis and Zydney, “Protein Ultrafiltration,” in Encyclopedia of Industrial Biotechnology: Bioprocess, Bioseparation, and Cell Technology, ed. by Flickinger and Drew, pp. 4142-4162, John Wiley & Sons, Inc. (2010).
Van Reis and Zydney, J. Membrane Sci. 297:16-50 (2007).
Van Slyke, “On the Measurement of Buffer Values and on the Relationship of Buffer Value to the Dissociation Constant of the Buffer and the Concentration and Reaction of the Buffer Solution,” J. Biol. Chem., 52:525-570 (1922).
Vectibix™ Label (Sep. 2006).
Vivaglobin® Label (Jan. 2006).
Yim “Summary Review for Regulatory Action,” Division of Pulmonary, Allergy, and Rheumatology Products (U.S. Food & Drug Administration), of Humira® (2015).
Yu, et al., “Pain Perception Following Subcutaneous Injections of Citrate-Buffered and Phosphate-Buffered Epoetin Alpha,” Int'l J. Artificial Organs 21(6), 341-343 (1998).
Zevalin® Label, Physicians' Desk Reference (Thomson PDR, Montvale, N.J., 60th ed.) (2006).
Zydney and Kuriyel, “Protein Concentration and Buffer Exchange,” in Methods in Biotechnology, vol. 9, Downstream Protein Processing, ed. by M. Desai, pp. 35-46, Humana Press, Totowa, NJ (2000) [Substitute].
Zydney, “5.2 Ultrafiltration / Diafiltration”, Slideshow Presentation at Novo Nordisk and Aventis Pasteur (2004).
Zydney, “Membrane Separations: Membrane Bioseparations,” in Encyclopedia of Separation Science, pp. 1748-1755, Academic Press, Ltd., London (2000).
U.S. Appl. No. 17/137,201, filed Dec. 29, 2020, Not Yet Published, Protein Formulations and Methods of Making Same, Pending.
U.S. Appl. No. 16/228,478, filed Dec. 20, 2018, US 2019/0358322 (Nov. 28, 2019), Protein Formulations and Methods of Making Same, Pending.
U.S. Appl. No. 15/423,503, filed Feb. 2, 2017, US 2017/0143828 (May 25, 2017), Protein Formulations and Methods of Making Same, Abandoned.
U.S. Appl. No. 15/096,043, filed Apr. 11, 2016, US 2016/0222101 (Aug. 4, 2016), Protein Formulations and Methods of Making Same, Abandoned.
U.S. Appl. No. 14/796,389, filed Jul. 10, 2015, US 2015/0361170 (Dec. 17, 2015), Protein Formulations and Methods of Making Same, Abandoned.
U.S. Appl. No. 16/531,889, filed Aug. 5, 2019, Protein Formulations and Methods of Making Same, Abandoned.
U.S. Appl. No. 14/506,576, filed Oct. 3, 2014, US 2015/0023977 (Jan. 22, 2015), Anti-TNF Antibodies Formulations, Patented U.S. 9,085,619 (Jul. 21, 2015).
U.S. Appl. No. 13/774,735, filed Feb. 22, 2013, US 2013/0156760 (Jun. 20, 2013), Protein Formulations and Methods of Making Same, Patented U.S. 8,883,146 (Nov. 11, 2014).
U.S. Appl. No. 12/325,049, filed Nov. 28, 2008, US 2009/0291062 (Nov. 26, 2009), Antibody Formulations and Methods of Making Same, Patented U.S. 8,420,081 (Apr. 16, 2013).
U.S. Appl. No. 13/794,881, filed Mar. 12, 2013, 2013/0195888 (Aug. 1, 2013), Ultrafiltration and Diafiltration Formulation Methods for Protein Processing, Abandoned.
Related Publications (1)
Number Date Country
20210113695 A1 Apr 2021 US
Provisional Applications (1)
Number Date Country
61004992 Nov 2007 US
Divisions (1)
Number Date Country
Parent 14506576 Oct 2014 US
Child 14796389 US
Continuations (6)
Number Date Country
Parent 16228478 Dec 2018 US
Child 17137246 US
Parent 15423503 Feb 2017 US
Child 16228478 US
Parent 15096043 Apr 2016 US
Child 15423503 US
Parent 14796389 Jul 2015 US
Child 15096043 US
Parent 13774735 Feb 2013 US
Child 14506576 US
Parent 12325049 Nov 2008 US
Child 13774735 US