Method for measuring hydrophobicity of chromatographic resin

文档序号:1836104 发布日期:2021-11-12 浏览:17次 中文

阅读说明:本技术 测量色谱树脂的疏水性的方法 (Method for measuring hydrophobicity of chromatographic resin ) 是由 黄弨 J·J·佩里 徐晅阔 S·高斯 李正剑 W·金 于 2020-03-27 设计创作,主要内容包括:在某些实施方案中,本发明提供了一种测量色谱树脂的疏水性水平的方法。在某些实施方案中,本发明提供了一种选择用于从混合物中纯化目的蛋白的色谱树脂条件的方法,其中所述目的蛋白在色谱法过程中具有低的或没有聚集形成。在某些实施方案中,本发明提供了一种从多种色谱树脂中选择用于从混合物中纯化目的蛋白的色谱树脂的方法,其中所述目的蛋白在色谱法过程中具有低的或没有聚集形成。(In certain embodiments, the present invention provides a method of measuring the level of hydrophobicity of a chromatography resin. In certain embodiments, the present invention provides a method of selecting chromatography resin conditions for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography. In certain embodiments, the present invention provides a method of selecting a chromatography resin from a plurality of chromatography resins for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography.)

1. A method of measuring the level of hydrophobicity of a chromatography resin, comprising:

a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle;

b) contacting the fluorophore-conjugated gold nanoparticles with a chromatography resin in solution;

c) removing the supernatant and washing the chromatography resin with a wash buffer; and

d) quantifying the level of fluorescence intensity of the chromatographic resin,

thereby measuring the level of hydrophobicity of the chromatography resin, wherein the level of fluorescence intensity from (d) is indicative of the level of hydrophobicity of the chromatography resin.

2. The method of claim 1, wherein the chromatography resin is selected from the group consisting of an ion exchange chromatography resin, a hydrophobic interaction chromatography resin, an affinity chromatography resin, and a mixed mode chromatography resin.

3. The method of claim 1, wherein the fluorophore is selected from the group consisting of a boron-dipyrromethane (BODIPY) dye, 8-anilino-1-naphthalenesulfonic Acid (ANS), 4 ' -dianilino-1, 1 ' -binaphthyl-5, 5 ' -disulfonic acid (Bis-ANS), 6-propionyl-2- (N, N-dimethylamino) naphthalene (PRODAN), tetraphenylethylene derivatives, and nile red.

4. The method of claim 3, wherein the fluorophore is a boron-dipyrromethane (BODIPY) dye.

5. The method of claim 1, wherein the gold nanoparticles have a diameter of 5nm, 10nm, 15nm, 20nm, 30nm, 40nm, 50nm, 60nm, 70nm, 80nm, 90nm, or 100 nm.

6. The method of claim 4, wherein the BODIPY is conjugated to GNP by cross-linking the NHS (N-hydroxysuccinimide) group of the BODIPY NHS molecule with a primary amine group pre-attached on the GNP surface.

7. A method of selecting chromatography resin conditions for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography, the method comprising:

a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle;

b) contacting the fluorophore-conjugated gold nanoparticles with a chromatography resin in solution under different conditions;

c) removing the supernatant and washing the chromatography resin with a wash buffer;

d) quantifying the level of fluorescence intensity of the chromatography resin resulting from the different conditions in (b);

e) measuring the hydrophobicity level of the chromatography resin under different conditions, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin; and

f) selecting chromatographic resin conditions wherein the level of hydrophobicity of the chromatographic resin results in low or no aggregate formation during chromatographic purification of the protein of interest.

8. A method of selecting a chromatography resin from a plurality of chromatography resins for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography, the method comprising:

a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle;

b) contacting the fluorophore-conjugated gold nanoparticles with each chromatography resin in solution;

c) removing the supernatant and washing the chromatography resin with a wash buffer;

d) quantifying the level of fluorescence intensity of the chromatographic resin;

e) measuring the hydrophobicity level of the chromatography resin, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin; and

f) selecting a chromatography resin having a level of hydrophobicity that results in low or no aggregate formation during chromatographic purification of said protein of interest.

9. The method of claim 7 or 8, wherein the protein of interest is a monoclonal antibody.

10. The method of claim 7 or 8, wherein the chromatography is cation exchange Chromatography (CEX).

11. The method of claim 7 or 8, wherein the mixture has been obtained by affinity chromatography.

12. The method of claim 7 or 8, wherein the mixture is selected from the group consisting of harvested cell culture fluid, cell culture supernatant, and conditioned cell culture supernatant, cell lysate, and clarified bulk.

13. The method of claim 12, wherein the cell culture is a mammalian cell culture.

14. The method of claim 13, wherein the cell culture is a Chinese Hamster Ovary (CHO) cell culture.

Background

The large-scale economic purification of proteins is an increasingly important issue in the biopharmaceutical industry. Therapeutic proteins are typically produced using prokaryotic or eukaryotic cell lines engineered to express the protein from a recombinant plasmid containing a gene encoding the protein of interest. The separation of the desired protein from the mixture of components supplied to the cells, cellular by-products, and aggregated forms of the protein to a sufficient degree of purity (e.g., sufficient for use as a human therapeutic) presents a formidable challenge to the biological agent manufacturer.

Accordingly, there is a need in the art for improved protein purification methods that can be used to accelerate large scale processing of protein-based therapeutics (such as antibodies).

Disclosure of Invention

In certain embodiments, the present invention provides a method of measuring the level of hydrophobicity of a chromatography resin comprising: (a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle; (b) contacting the fluorophore-conjugated gold nanoparticles with a chromatography resin in solution; (c) removing the supernatant and washing the chromatography resin with a wash buffer; and (d) quantifying the fluorescence intensity level of the chromatography resin, thereby measuring the hydrophobicity level of the chromatography resin, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin.

Optionally, the chromatography resin is selected from the group consisting of ion exchange chromatography resins, hydrophobic interaction chromatography resins, affinity chromatography resins, and mixed mode chromatography resins.

Optionally, the fluorophore is selected from the group consisting of boron-dipyrromethane (BODIPY) dye, 8-anilino-1-naphthalenesulfonic Acid (ANS), 4 ' -dianilino-1, 1 ' -binaphthyl-5, 5 ' -disulfonic acid (Bis-ANS), 6-propionyl-2- (N, N-dimethylamino) naphthalene (PRODAN), tetraphenylethylene derivatives, and Nile Red (Nile Red). Preferably, the fluorophore is a boron-dipyrromethane (BODIPY) dye. Optionally, the gold nanoparticles have a diameter of 5nm, 10nm, 15nm, 20nm, 30nm, 40nm, 50nm, 60nm, 70nm, 80nm, 90nm, or 100 nm. Optionally, the BODIPY is conjugated to GNPs by cross-linking the NHS (N-hydroxysuccinimide) group of the bodipyn NHS molecule with a primary amine group pre-attached on the GNP surface. Alternatively, BODIPY can be conjugated to GNPs using other attachment chemistries, including biotin-linked to-streptavidin; and maleimide-linked to-thiol.

In certain embodiments, the present invention provides a method of selecting chromatography resin conditions for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography, the method comprising: (a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle; (b) contacting the fluorophore-conjugated gold nanoparticles with a chromatography resin in solution under different conditions; (c) removing the supernatant and washing the chromatography resin with a wash buffer; (d) quantifying the level of fluorescence intensity of the chromatography resin resulting from the different conditions in (b); (e) measuring the hydrophobicity level of the chromatography resin under different conditions, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin; and (f) selecting chromatographic resin conditions wherein the level of hydrophobicity of the chromatographic resin results in low or no aggregate formation during chromatographic purification of the protein of interest.

Optionally, the chromatography resin is selected from the group consisting of ion exchange chromatography resins, hydrophobic interaction chromatography resins, affinity chromatography resins, and mixed mode chromatography resins.

Optionally, the fluorophore is selected from the group consisting of boron-dipyrromethane (BODIPY) dye, 8-anilino-1-naphthalenesulfonic Acid (ANS), 4 ' -dianilino-1, 1 ' -binaphthyl-5, 5 ' -disulfonic acid (Bis-ANS), 6-propionyl-2- (N, N-dimethylamino) naphthalene (PRODAN), tetraphenylethylene derivatives, and nile red. Preferably, the fluorophore is a boron-dipyrromethane (BODIPY) dye. Optionally, the gold nanoparticles have a diameter of 5nm, 10nm, 15nm, 20nm, 30nm, 40nm, 50nm, 60nm, 70nm, 80nm, 90nm, or 100 nm. Optionally, the BODIPY is conjugated to GNPs by cross-linking the NHS (N-hydroxysuccinimide) group of the bodipyn NHS molecule with a primary amine group pre-attached on the GNP surface. Alternatively, BODIPY can be conjugated to GNPs using other attachment chemistries, including biotin-linked to-streptavidin; and maleimide-linked to-thiol.

Optionally, the protein of interest is a monoclonal antibody.

Optionally, the mixture is selected from the group consisting of harvested cell culture fluid, cell culture supernatant, and conditioned cell culture supernatant, cell lysate, and clarified bulk. For example, the cell culture is a mammalian cell culture (e.g., Chinese Hamster Ovary (CHO) cell culture). Optionally, the mixture has been obtained by affinity chromatography.

In certain embodiments, the present invention provides a method of selecting a chromatography resin from a plurality of chromatography resins for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography, the method comprising: (a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle; (b) contacting the fluorophore-conjugated gold nanoparticles with each chromatography resin in solution; (c) removing the supernatant and washing the chromatography resin with a wash buffer; (d) quantifying the level of fluorescence intensity of the chromatographic resin; (e) measuring the hydrophobicity level of the chromatography resin, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin; and (f) selecting a chromatography resin having a level of hydrophobicity that results in low or no aggregate formation during chromatographic purification of the protein of interest.

Optionally, the chromatography resin is selected from the group consisting of ion exchange chromatography resins, hydrophobic interaction chromatography resins, affinity chromatography resins, and mixed mode chromatography resins.

Optionally, the fluorophore is selected from the group consisting of boron-dipyrromethane (BODIPY) dye, 8-anilino-1-naphthalenesulfonic Acid (ANS), 4 ' -dianilino-1, 1 ' -binaphthyl-5, 5 ' -disulfonic acid (Bis-ANS), 6-propionyl-2- (N, N-dimethylamino) naphthalene (PRODAN), tetraphenylethylene derivatives, and nile red. Preferably, the fluorophore is a boron-dipyrromethane (BODIPY) dye. Optionally, the gold nanoparticles have a diameter of 5nm, 10nm, 15nm, 20nm, 30nm, 40nm, 50nm, 60nm, 70nm, 80nm, 90nm, or 100 nm. Optionally, the BODIPY is conjugated to GNPs by cross-linking the NHS (N-hydroxysuccinimide) group of the bodipyn NHS molecule with a primary amine group pre-attached on the GNP surface. Alternatively, BODIPY can be conjugated to GNPs using other attachment chemistries, including biotin-linked to-streptavidin; and maleimide-linked to-thiol.

Optionally, the protein of interest is a monoclonal antibody.

Optionally, the mixture is selected from the group consisting of harvested cell culture fluid, cell culture supernatant, and conditioned cell culture supernatant, cell lysate, and clarified bulk. For example, the cell culture is a mammalian cell culture (e.g., Chinese Hamster Ovary (CHO) cell culture). Optionally, the mixture has been obtained by affinity chromatography.

Drawings

Fig. 1 shows a schematic of BODIPY FL NHS functionalization onto Gold Nanoparticles (GNPs).

FIG. 2 shows the hydrophobic ordering of HIC resins by the GNP-BODIPY method.

FIG. 3 shows the hydrophobic ordering of CEX resins by the GNP-BODIPY method.

Figure 4 shows the hydrophobicity of different base matrix materials derived by the GNP-BODIPY method.

Figure 5 shows the effect of GNP size on accessible hydrophobicity for various resin types.

Figure 6 shows the effect of the resin hydrophobic surface accessible to the conjugated mAb molecules. mAb1 is IgG1 and mAb2 is IgG 4. The ratio of pool (pool) HMW to load HMW indicates aggregate formation. Ratios >1 indicate on-column aggregation.

Detailed Description

In certain embodiments, the present invention provides a method of measuring the level of hydrophobicity of a chromatography resin comprising: (a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle; (b) contacting the fluorophore-conjugated gold nanoparticles with a chromatography resin in solution; (c) removing the supernatant and washing the chromatography resin with a wash buffer; and (d) quantifying the fluorescence intensity level of the chromatography resin, thereby measuring the hydrophobicity level of the chromatography resin, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin.

Optionally, the chromatography resin is selected from the group consisting of ion exchange chromatography resins, hydrophobic interaction chromatography resins, affinity chromatography resins, and mixed mode chromatography resins.

Optionally, the fluorophore is selected from the group consisting of boron-dipyrromethane (BODIPY) dye, 8-anilino-1-naphthalenesulfonic Acid (ANS), 4 ' -dianilino-1, 1 ' -binaphthyl-5, 5 ' -disulfonic acid (Bis-ANS), 6-propionyl-2- (N, N-dimethylamino) naphthalene (PRODAN), tetraphenylethylene derivatives, and nile red. Preferably, the fluorophore is a boron-dipyrromethane (BODIPY) dye. Optionally, the gold nanoparticles have a diameter of 5nm, 10nm, 15nm, 20nm, 30nm, 40nm, 50nm, 60nm, 70nm, 80nm, 90nm, or 100 nm. Optionally, the BODIPY is conjugated to GNPs by cross-linking the NHS (N-hydroxysuccinimide) group of the bodipyn NHS molecule with a primary amine group pre-attached on the GNP surface. Alternatively, BODIPY can be conjugated to GNPs using other attachment chemistries, including biotin-linked to-streptavidin; and maleimide-linked to-thiol.

In certain embodiments, the present invention provides a method of selecting chromatography resin conditions for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography, the method comprising: (a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle; (b) contacting the fluorophore-conjugated gold nanoparticles with a chromatography resin in solution under different conditions; (c) removing the supernatant and washing the chromatography resin with a wash buffer; (d) quantifying the level of fluorescence intensity of the chromatography resin resulting from the different conditions in (b); (e) measuring the hydrophobicity level of the chromatography resin under different conditions, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin; and (f) selecting chromatographic resin conditions wherein the level of hydrophobicity of the chromatographic resin results in low or no aggregate formation during chromatographic purification of the protein of interest.

Optionally, the chromatography resin is selected from the group consisting of ion exchange chromatography resins, hydrophobic interaction chromatography resins, affinity chromatography resins, and mixed mode chromatography resins.

Optionally, the fluorophore is selected from the group consisting of boron-dipyrromethane (BODIPY) dye, 8-anilino-1-naphthalenesulfonic Acid (ANS), 4 ' -dianilino-1, 1 ' -binaphthyl-5, 5 ' -disulfonic acid (Bis-ANS), 6-propionyl-2- (N, N-dimethylamino) naphthalene (PRODAN), tetraphenylethylene derivatives, and nile red. Preferably, the fluorophore is a boron-dipyrromethane (BODIPY) dye. Optionally, the gold nanoparticles have a diameter of 5nm, 10nm, 15nm, 20nm, 30nm, 40nm, 50nm, 60nm, 70nm, 80nm, 90nm, or 100 nm. Optionally, the BODIPY is conjugated to GNPs by cross-linking the NHS (N-hydroxysuccinimide) group of the bodipyn NHS molecule with a primary amine group pre-attached on the GNP surface. Alternatively, BODIPY can be conjugated to GNPs using other attachment chemistries, including biotin-linked to-streptavidin; and maleimide-linked to-thiol.

Optionally, the protein of interest is a monoclonal antibody.

Optionally, the mixture is selected from the group consisting of harvested cell culture fluid, cell culture supernatant, and conditioned cell culture supernatant, cell lysate, and clarified bulk. For example, the cell culture is a mammalian cell culture (e.g., Chinese Hamster Ovary (CHO) cell culture). Optionally, the mixture has been obtained by affinity chromatography.

In certain embodiments, the present invention provides a method of selecting a chromatography resin from a plurality of chromatography resins for purifying a protein of interest from a mixture, wherein the protein of interest has low or no aggregate formation during chromatography, the method comprising: (a) mixing a fluorophore with a Gold Nanoparticle (GNP) to form a fluorophore-conjugated gold nanoparticle; (b) contacting the fluorophore-conjugated gold nanoparticles with each chromatography resin in solution; (c) removing the supernatant and washing the chromatography resin with a wash buffer; (d) quantifying the level of fluorescence intensity of the chromatographic resin; (e) measuring the hydrophobicity level of the chromatography resin, wherein the fluorescence intensity level from (d) is indicative of the hydrophobicity level of the chromatography resin; and (f) selecting a chromatography resin having a level of hydrophobicity that results in low or no aggregate formation during chromatographic purification of the protein of interest.

Optionally, the chromatography resin is selected from the group consisting of ion exchange chromatography resins, hydrophobic interaction chromatography resins, affinity chromatography resins, and mixed mode chromatography resins.

Optionally, the fluorophore is selected from the group consisting of boron-dipyrromethane (BODIPY) dye, 8-anilino-1-naphthalenesulfonic Acid (ANS), 4 ' -dianilino-1, 1 ' -binaphthyl-5, 5 ' -disulfonic acid (Bis-ANS), 6-propionyl-2- (N, N-dimethylamino) naphthalene (PRODAN), tetraphenylethylene derivatives, and nile red. Preferably, the fluorophore is a boron-dipyrromethane (BODIPY) dye. Optionally, the gold nanoparticles have a diameter of 5nm, 10nm, 15nm, 20nm, 30nm, 40nm, 50nm, 60nm, 70nm, 80nm, 90nm, or 100 nm. Optionally, the BODIPY is conjugated to GNPs by cross-linking the NHS (N-hydroxysuccinimide) group of the bodipyn NHS molecule with a primary amine group pre-attached on the GNP surface. Alternatively, BODIPY can be conjugated to GNPs using other attachment chemistries, including biotin-linked to-streptavidin; and maleimide-linked to-thiol.

Optionally, the protein of interest is a monoclonal antibody.

Optionally, the mixture is selected from the group consisting of harvested cell culture fluid, cell culture supernatant, and conditioned cell culture supernatant, cell lysate, and clarified bulk. For example, the cell culture is a mammalian cell culture (e.g., Chinese Hamster Ovary (CHO) cell culture). Optionally, the mixture has been obtained by affinity chromatography.

I. Definition of

In order that the disclosure may be more readily understood, certain terms are first defined. As used in this application, each of the following terms shall have the meaning set forth below, unless the context clearly provides otherwise. Additional definitions are set forth throughout this application.

As used herein, the term "protein of interest" is used in its broadest sense to include any protein (natural or recombinant) present in a mixture that it is desired to purify. Such proteins of interest include, but are not limited to, hormones, growth factors, cytokines, immunoglobulins (e.g., antibodies), and molecules containing immunoglobulin-like domains (e.g., molecules containing ankyrin or fibronectin domains).

As used herein, "cell culture" refers to cells in a liquid medium. Optionally, the cell culture is contained in a bioreactor. The cells in the cell culture may be from any organism, including, for example, bacteria, fungi, insects, mammals, or plants. In a specific embodiment, the cells in cell culture comprise cells transfected with an expression construct comprising a nucleic acid encoding a protein of interest (e.g., an antibody). Suitable liquid media include, for example, nutrient media and non-nutrient media. In a specific embodiment, the cell culture comprises a Chinese Hamster Ovary (CHO) cell line in nutrient medium, without being subjected to purification by, for example, filtration or centrifugation.

As used herein, the term "clarified bulk" refers to a mixture from which particulate matter has been substantially removed. The clarified bulk comprises a cell culture or cell lysate in which the cells or cell debris have been substantially removed, for example by filtration or centrifugation.

As used herein, a "mixture" comprises a protein of interest (which is desired to be purified) and one or more contaminants, i.e., impurities. In one embodiment, the mixture is produced by a host cell or organism that expresses the protein of interest (naturally or recombinantly). Such mixtures include, for example, cell cultures, cell lysates, and clarified bulk (e.g., clarified cell culture supernatant).

As used herein, the terms "isolate" and "purify" are used interchangeably and refer to the selective removal of contaminants from a mixture containing a protein of interest (e.g., an antibody).

As used herein, the term "contaminant" is used in its broadest sense to cover any undesirable component or compound within a mixture. In a cell culture, cell lysate, or clarified bulk (e.g., clarified cell culture supernatant), contaminants include, for example, host cell nucleic acids (e.g., DNA) and host cell proteins present in the cell culture medium. Host cell contaminant proteins include, but are not limited to, those proteins that are naturally or recombinantly produced by the host cell, as well as proteins (e.g., proteolytic fragments) related to or derived from the protein of interest, and other process-related contaminants. In certain embodiments, the contaminant precipitate is separated from the cell culture using art-recognized means such as centrifugation, sterile filtration, depth filtration, and tangential flow filtration.

As used herein, "centrifugation" is a process used in industrial and laboratory environments that involves the use of centrifugal force to effect settling of a heterogeneous mixture with a centrifuge. This process is used to separate two immiscible liquids. For example, in the methods of the invention, centrifugation can be used to remove contaminant precipitates from mixtures including, but not limited to, cell cultures or clarified cell culture supernatants or elution pools captured by a capture column.

As used herein, "sterile filtration" is a filtration method using a membrane filter, which is typically a filter having a pore size of 0.2 μm to effectively remove microorganisms or small particles. For example, in the methods of the invention, sterile filtration may be used to remove contaminant precipitates from mixtures including, but not limited to, cell cultures or clarified cell culture supernatants or elution pools captured by a capture column.

As used herein, "depth filtration" is a filtration process that uses depth filters, which are typically characterized by their design to retain particles due to a range of pore sizes within the filter matrix. The capacity of a depth filter is typically defined by the depth of the substrate (e.g., 10 inches or 20 inches) and thus the holding capacity for solids. For example, in the methods of the invention, depth filtration may be used to remove contaminant precipitates from mixtures including, but not limited to, cell cultures or clarified cell culture supernatants or elution pools captured by a capture column.

As used herein, the term "chromatography" refers to a process of separating a solute of interest (e.g., a protein of interest) in a mixture from other solutes in the mixture by diafiltration of the mixture through an adsorbent that adsorbs or retains the solute more or less strongly due to the properties of the solute such as pI, hydrophobicity, size and structure under certain buffer conditions of the process. In the methods of the invention, chromatography may be used to remove contaminants after removing a precipitate from a mixture including, but not limited to, a cell culture or clarified cell culture supernatant or an elution pool captured by a capture column.

The terms "ion exchange" and "ion exchange chromatography" refer to chromatographic processes in which an ionizable solute of interest (e.g., a protein of interest in a mixture) interacts with an oppositely charged ligand attached (e.g., by covalent attachment) to a solid phase ion exchange material under appropriate pH and conductivity conditions such that the solute of interest interacts non-specifically with the charged compound more or less than solute impurities or contaminants in the mixture. Contaminating solutes in the mixture can be washed from the column of ion exchange material or bound to or removed from the resin faster or slower than the solute of interest. "ion exchange chromatography" specifically includes cation exchange chromatography and anion exchange chromatography.

The phrase "ion exchange material" refers to a negatively charged solid phase (i.e., a cation exchange resin or membrane) or a positively charged solid phase (i.e., an anion exchange resin or membrane). In one embodiment, the charge may be provided by attaching (e.g., by covalent attachment) one or more charged ligands (or adsorbents) to the solid phase. Alternatively or additionally, the charge may be an inherent property of the solid phase (e.g., as is the case for silica having an overall negative charge).

"cation exchange resin" refers to a solid phase that is negatively charged and has free cations for exchange with cations in an aqueous solution passing over or through the solid phase. Any negatively charged ligand attached to a solid phase suitable for forming a cation exchange resin may be used, such as carboxylates, sulfonates, and other ligands as described below. Commercially available cation exchange resins include, but are not limited to, for example, those having sulfonate-based groups (e.g., MonoS, MiniS, Source 15S and 30S, SP Sepharose Fast Flow)TMHigh-Performance SP Sepharose, Capto S from GE Healthcare, Toyopearl SP-650S and SP-650M from Tosoh, Macro-Prep High S from BioRad, Ceramic HyperD S, Trisacryl M and LS SP, and Spherodex LS SP from Pall Technologies); those having sulfoethyl-based groups (e.g., Fractogel SE from EMD, Poros S-10 and S-20 from Thermo Fisher Scientific); those having sulfopropyl-based groups (e.g., TSK Gel SP 5PW and SP-5PW-HR from Tosoh, Capto SP ImpRes from GE Healthcare, POROS HS-20, HS 50, and POROS XS from Thermo Fisher Scientific); those having sulfoisobutyl-based groups (e.g., Fractogel EMD SO from EMD3 -) (ii) a Those having a sulfoxyethyl-based groupSome (e.g., SE52, SE53, and Express-Ion S from Whatman); those having carboxymethyl-based groups (e.g., CM Sepharose Fast Flow from GE Healthcare; Hydrocell CM from Biochrom Labs Inc.; Macro-Prep CM from BioRad; Ceramic HyperD CM; Trisacryl M CM, Trisacryl LS CM from Pall Technologies; Matrx Cellufine C500 and C200 from Millipore; CM52, CM32, CM23 and Express-Ion C from Whatman; Toyopearl CM-650S, CM-650M and CM-650C from Tosoh); those having sulfonic and carboxylic acid based groups (e.g., BAKERBOND carbon-sulfo from j.t. baker); those having carboxylic acid-based groups (e.g., WP CBX from J.T Baker; DOWEX MAC-3 from Dow Liquid separators; Amberlite Weak cation exchanger, DOWEX Weak cation exchanger, and Diaion Weak cation exchanger from Sigma-Aldrich; and Fractogel EMD COO-from EMD); those having Sulfonic acid-based groups (e.g., Hydrocell SP from Biochrom Labs inc., DOWEX fine mesh strong acid cation resin from Dow Liquid Separations, UNOsphere S, WP Sulfonic from j.t. baker, Sartobind S membrane from Sartorius, Amberlite strong cation exchanger from Sigma-Aldrich, DOWEX strong cation, and Diaion strong cation exchanger); and those having orthophosphate-based groups (e.g., P11 from Whatman).

"anion exchange resin" refers to a positively charged solid phase, and thus has one or more positively charged ligands attached to it. Any positively charged ligand attached to a solid phase suitable for forming an anion exchange resin may be used, such as quaternary amino groups, commercially available anion exchange resins include DEAE cellulose, POROS PI 20, PI 50, HQ 10, HQ 20, HQ 50, D50 and POROS XQ from Thermo Fisher Scientific, Sartobind Q from Sartorius, MonoQ, MiniQ, Source 15Q and 30Q, Q, DEAE and ANX Sepharose Fast Flow, high efficiency Q Sepharose, Capto Q, QAE SEPHADEXTMAnd FAST Q SEPHAROSETM(GE Healthcare), WP PEI, WP DEAM, WP QUAT from J.T.Baker, Hydrocell DEAE and Hydrocell QA from Biochrom Labs Inc., UNOsphere Q, Macro-Prep DEAE and Macro-Prep High Q, Ceramic HyperD DEAE, Trisacryl M and LS DEAE, Spherodex LS DEAE, QMA Spherosil LS, QMA Spherosil M and Mustang Q from Pall Technologies, DOWEX fine mesh Strong base I and II anion resins and DOWEX MONOSPE 77 (weak base anion) from Dow Liquid separators, Intercept Q membrane, Matrex Cellucine A200, A500, Q500 and Q800 from Millipore, Fractogel EMD TMAE, Fractogel EMD DEAE and Fractogel EMD DMAE from EMD, Amberlite I and II strong anion exchangers, EX DOWEI and Diso weak anion exchangers, Super I and Strong anion exchangers, TSAcer II and Subco 650, QAEI and TSARE 650Q 650-5 from Sigma Aldrich, Tosox Topex Q650 and PW 5, Topex PW 650Q 650 and Q650Q 5 from Dow Liquid separators, Doweq 650 and Q500, Doweq 500, and Q500 and Q800 from Doweq 5, DE23, DE32, DE51, DE52, DE53, Express-Ion D and Express-Ion Q, and Sartobind Q (Sartorius corporation, N.Y., USA).

"Mixed mode ion exchange resin" or "mixed mode" refers to a solid phase covalently modified with cationic, anionic, and/or hydrophobic moieties. Examples of mixed mode ion exchange resins include BAKERBOND ABXTM(J.T.Baker; Philippiberg, N.J.); ceramic hydroxyapatite type I and type II and fluorhydroxyapatite (BioRad; Heracleus, Calif.); MEP and MBI HyperCel (Pall Corporation; East Hills, N.Y.); capto adhere, Capto MMC and Capto MMC imprres (GE Healthcare).

"hydrophobic interaction chromatography resin" refers to a solid phase covalently modified with methyl, ether, phenyl, butyl, hexyl, and octyl chemicals. Hydrophobic interaction chromatography is a separation technique that uses the property of hydrophobicity to separate proteins from each other. In this type of chromatography, hydrophobic groups such as methyl, ether, phenyl, butyl, hexyl and octyl are attached to fixed columns. Proteins passing through a column with hydrophobic amino acid side chains on its surface are able to interact with and bind to the hydrophobic groups on the column. Examples of hydrophobic interaction chromatography resins include: (1) butyl FF, butyl HP, octyl FF, phenyl HP, phenyl FF (high sub), phenyl FF (low sub), Capto phenyl imprmes, Capto phenyl (high sub), Capto octyl, Capto butyl ImpRes, Capto butyl (GE Healthcare, uppsala, sweden); (2) toyopearl Super butyl-550C, Toyopearl hexyl-650C, butyl-650C, phenyl-650C, butyl 600M, phenyl-600M, PPG-600M, butyl-650M, phenyl-650M, ether-650M, butyl-650S, phenyl-650S, ether-650S, TSKgel phenyl-5 PW, TSKgel ether-5 PW (Tosoh Bioscience, Tokyo, Japan); (3) Macro-Prep-butyl, Macro-Prep-methyl (Bio-Rad); (4) sartobind phenyl (Sartorius corporation, new york, usa); and (5) POROS ethyl, POROS benzyl ultra (thermo Fisher scientific).

Protein of interest

In certain aspects, the methods of the invention can be used to purify any protein of interest, including but not limited to proteins having any of a variety of other properties useful for pharmaceutical, diagnostic, agricultural, and/or commercial, experimental, or other applications. In addition, the protein of interest may be a protein therapeutic. In certain embodiments, proteins purified using the methods of the invention may be processed or modified. For example, the protein of interest according to the invention may be glycosylated.

Thus, the invention can be used to culture cells to produce any therapeutic protein, such as a pharmaceutically or commercially relevant enzyme, receptor fusion protein, antibody (e.g., monoclonal or polyclonal antibody), antigen binding fragment of an antibody, Fc fusion protein, cytokine, hormone, regulatory factor, growth factor, coagulation/coagulation factor, or antigen binding agent. The above list of proteins is merely exemplary in nature and is not intended to be a limiting statement. One of ordinary skill in the art will know that other proteins can be produced according to the present invention, and will be able to produce such proteins using the methods disclosed herein.

In a particular embodiment of the invention, the protein purified using the method of the invention is an antibody. The term "antibody" is used in the broadest sense to include monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), antibody fragments, immunoadhesins, and antibody-immunoadhesin chimeras.

An "antibody fragment" includes at least a portion of a full-length antibody and typically includes an antigen-binding or variable region thereof. Examples of antibody fragments include Fab, Fab ', F (ab')2And Fv fragments; a single chain antibody molecule; a diabody; a linear antibody; and multispecific antibodies formed from engineered antibody fragments.

The term "monoclonal antibody" is used in a conventional sense to refer to an antibody obtained from a substantially homogeneous population of antibodies such that the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. This is in contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes) of an antigen, whereas monoclonal antibodies are directed against a single determinant on an antigen. In describing antibodies, the term "monoclonal" indicates that the antibody is characterized as being obtained from a substantially homogeneous population of antibodies, and should not be construed as requiring production of the antibody by any particular method. For example, monoclonal antibodies used in the present invention can be produced using conventional hybridoma technology first described by Kohler et al, Nature 256:495(1975), or they can be made using recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). Clackson et al, Nature 352: 624-; marks et al, J.mol.biol.222:581-597 (1991); and U.S. Pat. nos. 5,223,409; 5,403,484; 5,571,698; 5,427,9085,580,717, respectively; 5,969,108, respectively; 6,172,197, respectively; 5,885,793, respectively; 6,521,404; 6,544,731, respectively; 6,555,313, respectively; 6,582,915, respectively; and 6,593,081) from phage antibody libraries.

Monoclonal antibodies described herein include "chimeric" and "humanized" antibodies in which a portion of the heavy and/or light chain is identical to or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain or chains are identical to or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567; and Morrison et al, Proc. Natl.Acad.Sci.USA 81:6851-6855 (1984)). "humanized" forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequences derived from non-human immunoglobulins. In most cases, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) having the desired specificity, affinity, and capacity, such as mouse, rat, rabbit, or non-human primate. In some cases, Fv Framework Region (FR) residues of the human immunoglobulin are replaced with corresponding non-human residues. In addition, humanized antibodies may contain residues that are not found in the recipient antibody or in the donor antibody. These modifications were made to further refine antibody performance. Generally, the humanized antibody will comprise substantially all of at least one and typically two variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically at least a portion of a constant region of a human immunoglobulin. For further details, see Jones et al, Nature 321:522-525 (1986); riechmann et al, Nature 332: 323-E329 (1988); and Presta, curr, Op, Structure, biol.2:593-596 (1992).

Chimeric or humanized antibodies can be prepared based on the sequence of a murine monoclonal antibody prepared as described above. DNA encoding the heavy and light chain immunoglobulins can be obtained from a murine hybridoma of interest and engineered to contain non-murine (e.g., human) immunoglobulin sequences using standard molecular biology techniques. For example, to generate chimeric antibodies, murine variable regions can be joined to human constant regions using methods known in the art (see, e.g., U.S. Pat. No. 4,816,567 to Cabilly et al). To generate humanized antibodies, murine CDR regions can be inserted into a human framework using methods known in the art (see, e.g., U.S. Pat. No. 5,225,539 to Winter, and U.S. Pat. Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen).

The monoclonal antibodies described herein also include "human" antibodies, which can be isolated from a variety of sources, including, for example, from the blood of human patients, or recombinantly produced using transgenic animals. Examples of such transgenic animals include those with a human heavy chain transgene and a human light chain transchromosome(Medarex, Inc., Princeton, N.J.) (see WO 02/43478),(Abgenix, Inc., Fllimont, Calif.; described, for example, in U.S. Pat. Nos. 5,939,598; 6,075,181; 6,114,598; 6,150,584 and 6,162,963 to Kucherlapati et al), and(Metarex, Inc.; described in, for example, Taylor, L. et al (1992) Nucleic Acids Research 20: 6287. sup. 6295; Chen, J. et al (1993) International Immunology 5: 647. sup. 656; Tuaillon et al (1993) Proc. Natl. Acad. Sci. USA 90: 3720. sup. 3724; Choi et al (1993) Nature Genetics 4: 117. sup. 123; Chen, J. et al (1993) EMBO J.12: 821. sup. 830; Tuaillon et al (1994) J. Immunol.152: 2912. sup. 2920; Taylor, L. et al (1994) International Immunology 6: 579. sup. 591; and Fishwi, D. et al (1996) Biotechnology 76, WO 5: 4835, WO 4631; WO 5,545,807; WO 4619, WO 5; WO 5,625,126; WO 469; WO 5; WO 4631; WO 11; WO 5; WO 5,625,126; WO 11; WO) Biology). The human monoclonal antibodies of the invention can also be prepared using SCID mice in which human immune cells have been reconstituted so that a human antibody response can be generated upon immunization. For example, Wilson et al, U.S. Pat. Nos. 5,476,996 and 5,698,767Such mice are described in (a).

Mixtures containing proteins of interest

The method of the invention can be applied to any mixture containing a protein of interest. In one embodiment, the mixture is obtained from or produced by living cells expressing the protein to be purified (e.g., naturally or by genetic engineering). Optionally, the cells in the cell culture comprise cells transfected with an expression construct comprising a nucleic acid encoding a protein of interest. Methods of genetically engineering cells to produce proteins are well known in the art. See, e.g., Ausabel et al, eds (1990), Current Protocols in Molecular Biology (Wiley, N.Y.), and U.S. Pat. Nos. 5,534,615 and 4,816,567, each of which is specifically incorporated herein by reference. Such methods include introducing nucleic acids encoding and allowing expression of the protein into living host cells. These host cells may be bacterial cells, fungal cells, insect cells or preferably animal cells grown in culture. Bacterial host cells include, but are not limited to, e. Examples of suitable E.coli strains include: HB101, DH5 α, GM2929, JM109, KW251, NM538, NM539, and any E.coli strain that is not capable of cleaving exogenous DNA. Fungal host cells that may be used include, but are not limited to, Saccharomyces cerevisiae (Saccharomyces cerevisiae), Pichia pastoris (Pichia pastoris), and Aspergillus cells. Insect cells that may be used include, but are not limited to, Bombyx mori (Bombyx mori), Spodoptera frugiperda (Mamestra sativa), Spodoptera frugiperda (Spodoptera frugiperda), Trichoplusia ni (Trichoplusia ni), Drosophila melanogaster (Drosophila melanogaster).

Many mammalian cell lines are suitable host cells for expression of a protein of interest. Mammalian host cell lines include, for example, COS, PER. C6, TM4, VERO076, DXB11, MDCK, BRL-3A, W138, Hep G2, MMT, MRC 5, FS4, CHO, 293T, A431, 3T3, CV-1, C3H10T1/2, Colo205, 293, HeLa, L cells, BHK, HL-60, FRhL-2, U937, HaK, Jurkat cells, Rat2, BaF3, 32D, FDCP-1, PC12, M1x, murine myeloma (e.g., myeloma)E.g., SP2/0 and NS0) and C2C12 cells, as well as transformed primate cell lines, hybridomas, normal diploid cells, and in vitro cultured cell lines derived from primary tissues and primary explants. New animal cell lines can be established using methods well known to those skilled in the art (e.g., by transformation, viral infection, and/or selection). Any eukaryotic cell capable of expressing a protein of interest can be used in the disclosed cell culture methods. Many cell lines are available from commercial sources such as the American Type Culture Collection (ATCC). In one embodiment of the invention, cell cultures, e.g., large scale cell cultures, are used with hybridoma cells. The construction of antibody-producing hybridoma cells is well known in the art. In one embodiment of the invention, a cell culture, e.g. a large scale cell culture, uses CHO cells to produce a protein of interest, such as an antibody (see, e.g., WO 94/11026). Various types of CHO cells are known in the art, e.g., CHO-K1, CHO-DG44, CHO-DXB11, CHO/dhfr-And CHO-S.

In certain embodiments, the present invention contemplates monitoring specific conditions of growing a cell culture prior to purifying a protein of interest from the cell culture. Monitoring cell culture conditions allows determining whether the cell culture produces the protein of interest at a sufficient level. For example, small aliquots of the culture are periodically removed for analysis in order to monitor certain cell culture conditions. Cell culture conditions to be monitored include, but are not limited to, temperature, pH, cell density, cell viability, integrated viable cell density, lactate levels, ammonium levels, osmolality, and titer of expressed protein. Those skilled in the art are familiar with many techniques for measuring such conditions/criteria. For example, a hemocytometer, an automated cell counting device (e.g., Coulter inc., Beckman, fullerene, ca) or a cell density check (e.g., cedex. rtm., Innovatis, marvin, pa) may be used. Viable cell density can be determined by staining the culture samples with trypan blue. Lactate levels and ammonium levels can be measured, for example, with a BioProfile 400 chemical analyzer (Nova biomedicalal, waltham, massachusetts) that makes real-time online measurements of key nutrients, metabolites, and gases in cell culture media. Osmolality of a cell culture can be measured by, for example, a freezing point osmometer. HPLC can be used to determine, for example, the level of lactate, ammonium, or expressed protein. In one embodiment of the invention, the level of protein expressed may be determined by using, for example, protein a HPLC. Alternatively, the level of expressed protein can be determined by standard techniques (such as Coomassie staining of SDS-PAGE gels, Western blotting, Bradford assay, Lowry assay, biuret assay, and UV absorbance.

In a particular embodiment, the methods of the invention comprise the effective removal of contaminants from a mixture (e.g., cell culture, cell lysate, or clarified bulk) containing a high concentration of a protein of interest (e.g., an antibody). For example, the concentration of the protein of interest can range from about 0.5 to about 50mg/ml (e.g., 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 mg/ml).

The preparation of the mixture depends initially on the expression pattern of the protein. Some cell systems secrete proteins (e.g., antibodies) directly from the cells into the surrounding growth medium, while others retain the antibodies within the cells. For proteins produced intracellularly, any of a variety of methods (such as mechanical shearing, osmotic shock, and enzymatic treatment) can be used to destroy the cells. Disruption releases the entire contents of the cells into the homogenate and additionally produces subcellular debris that can be removed by centrifugation or filtration. During the course of a protein production run, similar problems arise with directly secreted proteins, albeit to a lesser extent, due to natural cell death and release of host cell proteins within the cell.

In one embodiment, cells or cell debris are removed from the mixture, e.g., to prepare a clarified mass. The methods of the invention may use any suitable method to remove cells or cell debris. If the protein is produced intracellularly, as a first step, particulate debris (host cells or lysed fragments) can be removed, e.g., by a centrifugation or filtration step, to prepare a mixture, which is then subjected to purification (i.e., purification of the protein of interest therefrom) according to the methods described herein. If the protein is secreted into the culture medium, the recombinant host cells can be separated from the cell culture medium by, for example, centrifugation, tangential flow filtration, or depth filtration to prepare a mixture from which the protein of interest is purified.

In another embodiment, the cell culture or cell lysate is used directly without first removing the host cells. Indeed, the method of the invention is particularly well suited to the use of mixtures comprising secreted proteins and host cell suspensions.

The disclosure is further illustrated by the following examples, which should not be construed as further limiting. The contents of all figures and all references, patents and published patent applications cited in this application are expressly incorporated herein by reference in their entirety.

Example 1

The methods herein describe a technique for characterizing the overall hydrophobicity of a chromatography resin using gold nanoparticles functionalized with a fluorescent dye. Currently, retention time measurement of small spherical model proteins (e.g., ribonuclease A, lysozyme) is the standard method for comparing the hydrophobicity of adsorbents, particularly for Hydrophobic Interaction Chromatography (HIC) resins (GE Healthcare, Certificate of Analysis of Phenyl Sepharose)TM6Fast Flow (high sub), test and limit: AS 45-6003-91Ed. AG 3). However, for a wide range of chromatographic types, the general methods of quantifying the hydrophobicity of resins are still lacking and highly desirable. Fluorescent dyes for measuring surface hydrophobicity have been used to characterize the lumped hydrophobic properties of cation exchange Chromatography (CEX) resins (Chen Z, Huang C, Chennamsetty N, Xu X, Li ZJ. J Chromatogr A.2016,1460: 110-. Due to the small size of the dye used (usually<1000 daltons) the hydrophobic resin surface being probed is not as close to the larger biologics protein (usually the larger biologics protein)>10,000 daltons). In addition, the charge characteristics of the fluorescent dyeProperties can also affect the resin hydrophobicity test and complicate the results. Thus, in this approach, we prepared a new probe by functionalizing BODIPY FL NHS (an uncharged hydrophobic sensing fluorescent dye) onto Gold Nanoparticles (GNPs). BODIPY FL emits a fluorescent signal in a non-polar (hydrophobic) environment (Dorh N, Zhu S, Dhungana KB, Pati R, Luo FT, Liu H, Tiwari A.Sci Rep.2015; 5:18337.), while GNP provides similar steric hindrance to that of the biomacromolecule of interest (e.g., monoclonal antibody, mAb) (Robertson J, Rizzello L, Avila-Olias M, Gaitzsch J, Contini C, Mago ń M, Renshaw S and Battagliab G, Sci Rep.2016; 6: 27494.). GNP-BODIPY conjugates were prepared by cross-linking the NHS (N-hydroxysuccinimide) group of the BODIPY FL NHS molecule with a primary amine group pre-attached on the GNP surface. The data show that the new probes are capable of quantifying the overall hydrophobicity of HIC and CEX resins, which have similar potential applications to other resin types such as anion exchange chromatography (AEX), Affinity Chromatography (AC), and mixed mode chromatography (MM) resins. This approach can be optimized for more applications. For example, steric effects can be adjusted by using GNPs of different sizes to measure accessible resin surface hydrophobicity for various target molecules.

Fig. 1 shows a schematic of BODIPY FL NHS functionalization onto Gold Nanoparticles (GNPs).

In the first experiment, applicants demonstrated that this method was able to quantify the overall hydrophobicity of HIC resins. The hydrophobic ordering by the GNP-BODIPY method (hexyl-650C > butyl-650C > phenyl-650C) is in good agreement with what the resin supplier claims. See fig. 2.

In a second experiment, applicants demonstrated that this method was able to quantify the overall hydrophobic character of CEX resins. The GNP-BODIPY method sequences the hydrophobicity of CEX resins from different suppliers (POROS XS > Fractogel SE > Capto SP imprmes). See fig. 3.

In a third experiment, the applicant demonstrated that this method was used to compare the overall hydrophobic properties for resins made from different base matrix materials (both CEX and HIC). See fig. 4.

In a fourth experiment, applicants demonstrated that GNP size (i.e., diameter) was used as an adjustable parameter to probe steric effects and their effect on the accessible hydrophobicity of the chromatography resin. See fig. 5.

Next, the applicant conducted the following experiments of practical significance.

A. Chromatographic process development

This tool facilitates chromatographic (especially refining step) process development and parameter optimization. The hydrophobic nature of CEX resins plays a key role by affecting the product quality of therapeutic proteins (e.g., mabs). For example, depending on the resin and solution conditions used, IgG1 and IgG4 mAb can form High Molecular Weight (HMW) aggregates in CEX steps operating in bind/elute or flow/pass mode. Resin surface hydrophobicity accessible to bound IgG molecules was found to have a good correlation with mAb aggregation propensity in CEX step. The tools developed herein may be used to help develop and optimize CEX processes that achieve optimal column performance and product quality.

Figure 6 shows the effect of the resin hydrophobic surface accessible to the conjugated mAb molecules. mAb1 is IgG1 and mAb2 is IgG 4. The ratio of pool (pool) HMW to load HMW indicates aggregate formation. Ratios >1 indicate on-column aggregation.

B. Chromatography-type independent resin hydrophobicity determination

The determination requires a simple procedure and a very small amount of resin sample. It provides sufficient sensitivity that it can be used for the hydrophobic character of a relatively wide range of resins, regardless of the type of chromatography (e.g., HIC, CEX, AEX, MM, AC, etc.). Using the description described herein, the overall hydrophobicity of the resin can be quantified using about 50 μ L of resin (settled volume) over a 60 minute run time.

An example of a resin hydrophobicity test protocol is shown below.

1. 100 μ L of resin slurry was prepared with 50% resin in 25mM sodium acetate pH 5.5 buffer

2. Add 5. mu.L of 100. mu.L GNP-BODIPY stock solution to the target 10pg BODIPY/. mu.L settled resin

3. Add 25mM sodium acetate pH 5.5 buffer to a final 500. mu.L

4. Tumble mix for 30min and centrifuge at 5000x g for 1min

5. The supernatant was removed and 1mL of 25mM sodium acetate pH 5.5 was added for 10min with tumble mixing

6. Centrifuging at 5000x g for 3min, and completely removing supernatant

7. 50 μ L of 25mM sodium acetate pH 5.5 buffer was added to the resin

8. The resin slurry was transferred to a Uni cuvette (Uncariamed lab, Cat. No.: 201-1009) and excess liquid was removed using filter paper

9. Use of virgin resin syrup (without GNP-BODIPY) as a negative control

10. The Uni cuvette was placed on the platform of a ChemiDoc XRS + (BioRad, Cat. No.: 1708265) instrument

11. Image Lab software (BioRad, Cat. No.: 1708265) was turned on and fluorescein was selected as the screening condition (filter)

12. Fluorescence intensity data obtained by exposure for 0.1 to 1.0 seconds

13. Normalized fluorescence intensity (sample data minus negative control data) for each resin was plotted to obtain relative hydrophobicity.

Equivalent scheme

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 covered by the appended claims.

Is incorporated by reference

All patents, pending patent applications, and other publications cited herein are hereby incorporated by reference in their entirety.

18页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:动脉硬化及动脉硬化相关疾病标志物

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!