NOVEL PURIFICATION OF ANTIBODIES USING HYDROPHOBIC INTERACTION CHROMATOGRAPHY

Disclosed herein are compositions and methods for purifying antibody products from a sample matrix. In particular, the present invention relates to compositions and methods for purifying antibody products employing hydrophobic interaction chromatography media. In certain embodiments, the invention provides a method for reducing process-related impurities (e.g., host cell proteins), as well as product-related substances, including molecular weight variants (e.g., aggregates and fragments of the antibody product).

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

The present application is a continuation of U.S. application Ser. No. 13/831,181, filed on Mar. 14, 2013, which claims priority to U.S. Provisional Application Nos. 61/651,544, filed on May 24, 2012 and 61/658,920, filed on Jun. 13, 2012, the entire contents of each of the foregoing applications are incorporated herein by reference.

1. INTRODUCTION

The instant invention relates to the field of protein production and purification, and in particular to compositions and processes for controlling the amount of process-related impurities (e.g., host cell proteins and media components) and/or product-related substances (e.g., product charge variants, aggregates, and fragments) present in purified preparations by applying particular chromatography conditions during such protein purification.

2. BACKGROUND OF THE INVENTION

Hydrophobic interaction chromatography (HIC) media interact with hydrophobic regions present on an antibody of interest and the characteristics of that interaction can be calibrated by selecting specific HIC media types and binding conditions. In the context of commercial chromatographic purification, HIC medias are used to separate antibody product present in a variety of sample mixtures, including partially purified samples, e.g., samples that have been subjected to filtration and/or one or more step of affinity, ion exchange, and/or mixed mode chromatography. HIC is conventionally used in such strategies as a means for retaining an antibody product on a chromatographic support, while allowing other components in a partially purified sample, including product-related substances (e.g., product aggregates and fragments) and process-related impurities (e.g., host cell proteins), to be washed from the support and discarded and/or allow for the impurities to be resolved from the antibody product by selective elution of the antibody product. The retained antibody product can then be eluted from the chromatographic support by disrupting the antibody/HIC media interaction and the product can subsequently be subjected to further purification steps, e.g., those relying on charge (e.g., ion exchange chromatography), biological interaction characteristics (e.g., affinity chromatography), and/or size (e.g., ultrafiltration).

There remains a need in the art for high-efficiency methods of purifying antibody products away from product-related substances and process-related impurities at relatively low cost. Reduction of such substances and/or impurities is particularly advantageous in the context of commercially produced recombinant bio-therapeutics as such substances and/or impurities have the potential to impact numerous product characteristics, including, but not limited to, product stability, product safety and product efficacy.

3. SUMMARY OF THE INVENTION

The present invention is directed to compositions and methods for purifying proteins of interest from a sample mixture. In particular, the present invention relates to compositions and methods for purifying proteins employing HIC media. In certain embodiments, the invention provides a method for reducing product-related substances/impurities, including molecular weight variants (e.g., aggregates and fragments of the antibody product), as well as reducing process-related impurities (e.g., host cell proteins). The method involves contacting a sample mixture, e.g., a partially purified cell culture harvest sample, comprising the protein of interest, e.g., and antibody, with a hydrophobic chromatographic media in an aqueous salt solution under loading conditions that permit both the product, i.e., the protein of interest, and non-product proteins to bind to the hydrophobic media and then collecting the unbound product that is substantially reduced in process-related impurities and/or product-related substances from the media. The method further comprises a step whereby the bound product, is subsequently recovered by employing a wash with a similar aqueous salt composition that is present in the load sample while the product-related and process-related impurities remain bound to the HIC media. The wash fraction collected is also substantially reduced in process-related impurities and/or product-related substances

In certain embodiments, the purification strategies of the present invention may include one or more chromatography and/or filtration steps to achieve a desired degree of purification prior to exposure of the sample comprising the protein of interest of interest to the HIC media. For example, in certain embodiments, such pre-HIC chromatography step(s) can include one or more step of ion exchange chromatography and/or affinity chromatography. In certain embodiments, the purification strategies of the present invention may include one or more additional chromatography and/or filtration steps after the HIC purification step. In one aspect the filtration step is a nanofiltration step. In addition, in certain embodiments, the present invention is directed toward pharmaceutical compositions comprising one or more proteins of interest purified by methods described herein.

In certain embodiments, the sample mixture (also referred to as the “source material’ or simply “sample”) comprises a cell culture harvest wherein the cell line is employed to produce specific antibodies of the present invention. In certain embodiments, the sample exposed to the HIC media comprises a cell culture harvest that has undergone one or more purification and/or filtration steps, including but not limited to, one or more step of ion exchange chromatography and/or affinity chromatography.

In certain embodiments, an affinity chromatography step will precede the HIC chromatography step, thereby forming an affinity chromatography sample that can be exposed to the HIC media in the HIC chromatography step. In certain embodiments, the affinity chromatography step is a Protein A, G, A/G, or L affinity chromatography step. There are several commercial sources for Protein A media. One suitable media is MabSelect™ from GE Healthcare. In certain embodiments, the affinity chromatography sample is further subject to a filtration step. Filters well known to those skilled in the art can be used in this embodiment. In one aspect, the filtration step is a depth filtration step.

In certain embodiments, an affinity chromatography step will follow, directly or indirectly, the HIC chromatography step, thereby forming an affinity chromatography sample. In certain embodiments, the affinity chromatography step is a Protein A, G, A/G, or L affinity chromatography step. There are several commercial sources for Protein A media. One suitable media is MabSelect™ from GE Healthcare. In certain embodiments, the affinity chromatography sample is further subject to a filtration step. Filters well known to those skilled in the art can be used in this embodiment. In one aspect, the filtration step is a nanofiltration step.

In certain embodiments, an ion exchange step will precede the HIC chromatography step, thereby forming an ion exchange sample that can be exposed to the HIC media in the HIC chromatography step. In certain embodiments, the ion exchange step is either a cation exchange step or an anion exchange step. In certain embodiments, the ion exchange step is a single ion exchange chromatographic step or can include multiple ion exchange steps such as a cation exchange step followed by an anion exchange step or visa versa. In one aspect, the ion exchange step is a one step procedure. In certain embodiments, the ion exchange step involves a two step ion exchange process. A suitable cation exchange media is a media whose stationary phase comprises anionic groups. Examples of such a cation exchange media include, but are not limited to Fractogel, carboxymethyl (CM), sulfoethyl(SE), sulfopropyl(SP), phosphate(P) and sulfonate(S). In certain embodiments, the cation exchange media is Fractogel™ SO3. This ion exchange capture chromatography step facilitates the isolation of proteins of interest, e.g., antibodies, from a sample. A suitable anion exchange media is a media whose stationary phase comprises cationic groups. Examples of such an anion exchange media include, but are not limited to, Q sepharose, diethylaminoethyl (DEAE), quaternary aminoethyl(QAE), and quaternary amine(Q) groups. In certain embodiments, the anion exchange media is Q Sepharose™. One or more ion exchange step further isolates antibodies by reducing process-related impurities such as host cell proteins and DNA and, where applicable, affinity matrix protein. In certain embodiments, an ion exchange sample is further subject to a filtration step. Filters well known to those skilled in the art can be used in this embodiment. In one aspect, the filtration step is a depth filtration step. In certain embodiments, a filtration step follows the depth filtration step.

In certain embodiments, an ion exchange step will follow, directly or indirectly, the HIC chromatography step, thereby forming an ion exchange sample. In certain of such embodiments, the ion exchange step is either a cation exchange step or an anion exchange step. In certain embodiments, the ion exchange step is a single ion exchange chromatographic step or can include multiple ion exchange steps such as a cation exchange step followed by an anion exchange step or visa versa. In one aspect, the ion exchange step is a one step procedure. In certain embodiments, the ion exchange step involves a two step ion exchange process. A suitable cation exchange media is a media whose stationary phase comprises anionic groups. Examples of such a cation exchange media include, but are not limited to Fractogel, carboxymethyl (CM), sulfoethyl(SE), sulfopropyl(SP), phosphate(P) and sulfonate(S). In certain embodiments, the cation exchange media is Fractogel™ SO3. This ion exchange capture chromatography step facilitates the isolation of antibodies from a sample. A suitable anion exchange media is a media whose stationary phase comprises cationic groups. Examples of such an anion exchange media include, but are not limited to, Q sepharose, diethylaminoethyl (DEAE), quaternary aminoethyl(QAE), and quaternary amine(Q) groups. In certain embodiments, the anion exchange media is Q Sepharose™. In certain embodiments, an ion exchange sample is further subject to a filtration step. Filters well known to those skilled in the art can be used in this embodiment.

In certain embodiments, a mixed mode chromatography step will precede the HIC chromatography step, thereby forming a mixed mode chromatography sample that can be exposed to the HIC media in the HIC chromatography step. Examples of mixed mode medias include, but are not limited to: CaptoAdhere (GE Healthcare), PPA-HyperCel (Pall Life Sciences), and HEA-HyperCel (Pall Life Sciences). In certain embodiments, the mixed mode chromatography step is a CaptoAdhere chromatography step. In certain embodiments, the mixed mode chromatography sample is further subject to a filtration step. Filters well known to those skilled in the art can be used in this embodiment. In certain embodiments, a depth filtration step follows a filtration step.

In certain embodiments, a mixed mode chromatography step will follow, directly or indirectly, the HIC chromatography step, thereby forming a mixed mode chromatography sample. Examples of mixed mode medias include, but are not limited to: CaptoAdhere (GE Healthcare), PPA-HyperCel (Pall Life Sciences), and HEA-HyperCel (Pall Life Sciences). In certain embodiments, the mixed mode chromatography step is a CaptoAdhere chromatography step. In certain embodiments, the mixed mode chromatography sample is further subject to a filtration step. Filters well known to those skilled in the art can be used in this embodiment. In one aspect, the filtration step is a nanofiltration step. In certain embodiments, a depth filtration step follows a filtration step.

In certain embodiments, the present invention is directed toward methods of purifying a protein of interest, e.g., an antibody, from a sample mixture such that the resulting composition is substantially free of host cell proteins (“HCPs”). In addition, the purity of the proteins of interest in the resultant sample can be analyzed using methods well known to those skilled in the art, e.g., size-exclusion chromatography, Protein A HPLC Assay, HCP ELISA, Protein A ELISA, and western blot analysis.

In certain embodiments, the protein of interest is an antibody, such as, but not limited to a human antibody, a humanized antibody, a chimeric antibody, or a multivalent antibody, such as a DVD-Ig.

In certain embodiments, the invention is directed to one or more pharmaceutical compositions comprising an isolated protein of interest, e.g., an antibody, in the context of a process-related impurity-reduced and/or product-related substance-reduced preparation and an acceptable carrier. In certain embodiments, the compositions further comprise one or more pharmaceutical agents.

4. BRIEF DESCRIPTIONS OF THE DRAWINGS

FIG. 1 depicts a process chromatogram for the HIC purification of Adalimumab, wherein a GE CaptoPhenyl column was equilibrated at 1.1 M AmSO4 pH 7.0 (Tris/Acetate) for 10 CVs, Adalimumab was prepared at 1.1 M AmSO4 and loaded to the column at 20 g-protein/L of media, the column was then washed with 10 CVs of the equilibration buffer and a linear gradient from 1.1M to 0M AmSO4 pH 7.0 (Tris/Acetate) over 20 CVs was performed.

FIG. 2 depicts a process chromatogram for the HIC purification of Adalimumab, wherein a GE CaptoPhenyl column was equilibrated with 400 mM NaCit pH 5.6 for 10 CVs, Adalimumab was prepared at 400 mM NaCit pH 5.6, then loaded to the column at 500 g-protein/L-media, and the column was washed with 7 CVs of the equilibration buffer.

FIG. 3 depicts results of an experiment wherein a feed stream was serial diluted to cover a range of load concentrations from 4 to 15 mg/mL and loaded at 500 g/L to a CaptoPhenyl column in 400 mM NaCit pH 5.6; the results indicate the impact that the concentration of loaded protein can have on aggregate reduction.

FIG. 4 depicts an example of hybrid chromatography mode (i.e., a chromatographic mode having characteristics of both batch-elute and flow-through chromatography) wherein, based on the data presented in FIG. 2, the dynamic binding capacity (DBC), conventionally measured at 10% breakthrough, is greater than the equilibrium binding capacity (EBC).

FIG. 5A-B depict the results of experiments wherein aliquots of resin are incubated with a load covering a range of protein concentrations at room temperature for 3 hours, after which the protein solution is then removed, and replaced with equilibration buffer (Wash simulation) and incubated at room temperature for 3 hours (repeated, Wash II). After each incubation, the concentration of the protein solution is measured and used to calculated the amount of protein ((A) monomer D2E7, a.ka. Adalimumab, and (B) aggregate D2E7) bound to the resin (g protein/L resin) and plotted against the concentration of the protein solution at the end of the incubation (e.g. equilibrium).

FIG. 6A-B depict the results outlined in FIGS. 5A-B, highlighting the fact that at initial equilibrium a significant amount of monomer/aggregate is bound to the resin. However, after the protein solution is replaced with equilibration buffer (see arrow), the monomer de-sorbs from the resin and back into solution, where as the aggregate remains bound.

FIG. 7A-B depict a determination of the binding monomer and aggregate D2E7 (based on data provided in FIGS. 5A-B) by fitting the experimental equilibrium binding data to the Langmuir Isotherm using the equation: q=(qmax×Cequil)/(Kd+Cequil); where q=amount of protein bound to resin [=] g/L-resin; qmax=maximum amount of protein bound to resin [=] g/L-resin; Cequil=solution concentration of protein [=] g/L-soln; and Kd=equilibrium dissociation constant.

5. DETAILED DESCRIPTION OF THE INVENTION

The present invention is directed to compositions and methods for purifying antibody products from a sample matrix. In particular, the present invention relates to compositions and methods for purifying antibody products employing HIC media. In certain embodiments, the invention provides a method for modulating the content of product-related substances, including molecular weight variants (e.g., aggregates and fragments of the antibody product), in a purified sample of a protein of interest as well as reducing process-related impurities (e.g., host cell proteins). The method involves contacting a sample mixture, e.g., a partially purified cell culture harvest sample, comprising the protein of interest, e.g., and antibody, with a hydrophobic chromatographic media in an aqueous salt solution under loading conditions that permit both the product, i.e., the protein of interest, and non-product proteins to bind to the hydrophobic media and then collecting the unbound product that is substantially reduced in process-related impurities and/or product-related substances from the media. The method further comprises a step whereby the bound product, is subsequently recovered by employing a wash with a similar aqueous salt composition that is present in the load sample while the product-related and process-related impurities remain bound to the HIC media. The wash fraction collected is also substantially reduced in process-related impurities and/or product-related substances

In certain embodiments, the purification strategies of the present invention may include one or more chromatography and/or filtration steps to achieve a desired degree of purification prior to exposure of the sample comprising the antibody of interest to the HIC media. For example, in certain embodiments, such pre-HIC chromatography step(s) can include one or more step of ion exchange chromatography and/or affinity chromatography. In certain embodiments, the purification strategies of the present invention may include one or more additional chromatography and/or filtration steps after the HIC purification step. In addition, in certain embodiments, the present invention is directed toward pharmaceutical compositions comprising one or more antibodies purified by methods described herein.

For clarity and not by way of limitation, this detailed description is divided into the following sub-portions:

    • 5.1. Definitions;
    • 5.2. Antibody Generation;
    • 5.3. Antibody Production;
    • 5.4. Antibody Purification;
    • 5.5. Methods of Assaying Sample Purity;
    • 5.6. Further Modifications; and
    • 5.7. Pharmaceutical Compositions

5.1. Definitions

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

The term “product”, as used herein refers to a protein of interest, which may be present in the context of a sample comprising one or more process-related impurities and/or product-related substances. In certain embodiments, the product, i.e., the protein of interest, is an antibody or antigen binding fragment thereof.

The term “antibody” includes an immunoglobulin molecule comprised of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region (CH). 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 LCVR or 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 (CDRs), 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 term “antibody”, as used herein, also includes alternative antibody and antibody-like structures, such as, but not limited to, dual variable domain antibodies (DVD-Ig).

The term “antigen-binding portion” of an antibody (or “antibody portion”) includes fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., hIL-12, hTNFα, or hIL-18). 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 comprising 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 comprising the VH and CH1 domains; (iv) a Fv fragment comprising the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546, the entire teaching of which is incorporated herein by reference), which comprises a VH 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, the entire teachings of which are incorporated herein by reference). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see, e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123, the entire teachings of which are incorporated herein by reference). Still further, an antibody may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody with one or more other proteins or peptides. Examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101, the entire teaching of which is incorporated herein by reference) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al. (1994) Mol. Immunol. 31:1047-1058, the entire teaching of which is incorporated herein by reference). 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, as described herein. In one aspect, the antigen binding portions are complete domains or pairs of complete domains.

The terms “Kabat numbering” “Kabat definitions” and “Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci. 190:382-391 and, Kabat, E. A., 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 entire teachings of which are incorporated herein by reference). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.

The term “human antibody” includes antibodies having variable and constant regions corresponding to human germline immunoglobulin sequences as described by 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 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), e.g., in the CDRs and in particular CDR3. The mutations can be introduced using the “selective mutagenesis approach.” The human antibody can have at least one position replaced with an amino acid residue, e.g., an activity enhancing amino acid residue which is not encoded by the human germline immunoglobulin sequence. The human antibody can have up to twenty positions replaced with amino acid residues which are not part of the human germline immunoglobulin sequence. In other embodiments, up to ten, up to five, up to three or up to two positions are replaced. In one embodiment, these replacements are within the CDR regions. However, the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.

The phrase “recombinant human antibody” includes human 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 human antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see, e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295, the entire teaching of which is incorporated herein by reference) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences (see, Kabat, E. A., 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 approach or back-mutation or both.

An “isolated antibody” includes an antibody that is substantially free of other antibodies having different antigenic specificities. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.

The term “Koff”, as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.

The term “Kd”, as used herein, is intended to refer to the dissociation constant of a particular antibody-antigen interaction.

The phrase “nucleic acid molecule” includes DNA molecules and RNA molecules. A nucleic acid molecule may be single-stranded or double-stranded, but in one aspect is double-stranded DNA.

The phrase “isolated nucleic acid molecule,” as used herein in reference to nucleic acids encoding antibodies or antibody portions (e.g., VH, VL, CDR3), e.g. an antibody having a weak binding capacity for a Protein A media (e.g., a canine monoclonal antibody 82.4). The phrase “isolated nucleic acid molecule” is also intended to include sequences encoding bivalent, bispecific antibodies, such as diabodies in which VH and VL regions contain no other sequences other than the sequences of the diabody.

The phrase “recombinant host cell” (or simply “host cell”) includes a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.

The term “host cell proteins” (HCPs), as used herein, is intended to refer to non-target protein-related, proteinaceous impurities derived from host cells.

The term “modifying”, as used herein, is intended to refer to changing one or more amino acids in the antibodies or antigen-binding portions thereof. The change can be produced by adding, substituting or deleting an amino acid at one or more positions. The change can be produced using known techniques, such as PCR mutagenesis.

The term “about”, as used herein, is intended to refer to ranges of approximately 10-20% greater than or less than the referenced value. In certain circumstances, one of skill in the art will recognize that, due to the nature of the referenced value, the term “about” can mean more or less than a 10-20% deviation from that value.

The term “preparative scale”, as used herein, refers to a scale of purification operation that can be readily scaled-up and implemented at large scale manufacturing while still providing desired separation. For instance, one skilled in the field may develop a process using, e.g., a 0.5 cm (i.d.)×20 cm (length) column in the lab, and transfer it to large scale production using, e.g., a 30 cm (i.d.)×20 cm (length) column packed with the same media and operated with the same set of buffers, same linear flow rates (or residence times) and buffer volumes. In preparative scale separation, column bed height is typically ≦about 30 cm and column pressure drop ≦about 5 bar.

The term “aggregates” used herein means agglomeration or oligomerization of two or more individual molecules, including but not limiting to, protein dimers, trimers, tetramers, oligomers and other high molecular weight species. Protein aggregates can be soluble or insoluble.

The term “fragments” used herein refers to any truncated protein species from the target molecule due to dissociation of peptide chain, enzymatic and/or chemical modifications. For instance, antibody fragments include, but not limited to, Fab, F(ab′)2, Fv, scFv, Fd, dAb, or other compositions that contain a portion of the antibody molecule.

The term “charge variants”, as used herein, refers to the full complement of product variants including, but not limited to acidic species, and basic species (e.g., Lys variants). In certain embodiments, such variants can include product aggregates and/or product fragments, to the extent that such aggregation and/or fragmentation results in a product charge variation as seen in an analytical technique used for that purpose.

As used herein, the term “lysine variant heterogeneity” refers to a characteristic of a population of proteins wherein the population consists of proteins of substantially identical amino acid sequence, but where the population exhibits variation in the presence or absence of C-terminal lysine residues.

In certain embodiments, the protein is an antibody, and the distribution of lysine variant heterogeneity comprises a distribution of the lysine variants Lys 0, Lys 1 and Lys 2, wherein the Lys 0 lysine variant comprises an antibody with heavy chains that do not comprise a C-terminal lysine, wherein the Lys 1 lysine variant comprises an antibody with one heavy chain that comprises a C-terminal lysine, and wherein the Lys 2 lysine variant comprises an antibody wherein both heavy chains comprise a C-terminal lysine.

In certain embodiments, C-terminal lysine variants are associated with charge heterogeneities present in protein preparations, for example, monoclonal antibody (mAb) preparations, produced through a cell culture process. These heterogeneities can be detected by various methods, such as, for example, WCX-10 HPLC (a weak cation exchange chromatography), or IEF (isoelectric focusing).

In certain embodiments, the heterogeneity arises from subspecies of protein differing by the presence or absence of C-terminal lysines. For example, the population of proteins may comprise more than one subspecies of lysine variant. In one non-limiting example, the lysine variants may comprise at least two of Lys 0, Lys 1 and Lys 2 lysine variants which can be detected by weak cation exchange chromatography of the expression product of a host cell expressing Adalimumab.

In certain embodiments, the heterogeneity arises from the size of subpopulations having different C-terminal lysine profiles. For example, the population of proteins may comprise more than one subspecies of C-terminal lysine variant, and each of the variants may be present in different amounts. In one non-limiting example, the C-terminal lysine variants may be at least two of the Lys 0, Lys 1 and Lys 2 lysine variants detected by weak cation exchange chromatography of the expression product of a host cell expressing Adalimumab. In certain embodiments, Lys 0, Lys 1 or Lys 2 subspecies are present in different amounts.

In certain embodiments, the heterogeneity arises from both a difference in the amount of lysine variants in the population of proteins and the type of lysine variants present in the population of proteins.

As used herein, the terms “acidic species”, “acidic region” and “acidic species heterogeneity” refer to a characteristic of a population of proteins wherein the population includes a distribution of product-related substances identifiable by the presence of charge heterogeneities. For example, in monoclonal antibody (mAb) preparations, such acidic species heterogeneities can be detected by various methods, such as, for example, WCX-10 HPLC (a weak cation exchange chromatography), or IEF (isoelectric focusing). In certain embodiments, the acidic species identified using such techniques comprise a mixture of product-related substances containing antibody product fragments (e.g., Fc and Fab fragments), aggregates, and/or post-translation modifications of the antibody product, such as, deamidated and/or glycoslyated antibodies.

In certain embodiments, the acidic species heterogeneity comprises a difference in the type of acidic species present in the population of proteins. For example, the population of proteins may comprise more than one acidic species variant.

In certain embodiments, the heterogeneity of the distribution of acidic species comprises a difference in the amount of acidic species in the population of proteins. For example, the population of proteins may comprise more than one acidic species variant, and each of the variants may be present in different amounts.

5.2. Antibody Generation

The term “antibody” as used in this section refers to an intact antibody or an antigen binding fragment thereof.

The antibodies of the present disclosure can be generated by a variety of techniques, including immunization of an animal with the antigen of interest followed by conventional monoclonal antibody methodologies e.g., the standard somatic cell hybridization technique of Kohler and Milstein (1975) Nature 256: 495. Although somatic cell hybridization procedures are preferred, in principle, other techniques for producing monoclonal antibody can be employed e.g., viral or oncogenic transformation of B lymphocytes.

In certain embodiments, the animal system for preparing hybridomas is the murine system. Hybridoma production is a very well-established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known.

An antibody can be, in certain embodiments, a human, a chimeric, or a humanized antibody. Humanized antibodies of the present disclosure can be prepared based on the sequence of a non-human monoclonal antibody prepared as described above. DNA encoding the heavy and light chain immunoglobulins can be obtained from the non-human hybridoma of interest and engineered to contain non-murine (e.g., human) immunoglobulin sequences using standard molecular biology techniques. For example, to create a chimeric antibody, murine variable regions can be linked 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 create a humanized antibody, 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 et al.).

Human monoclonal antibodies can be generated using transgenic or transchromosomic mice carrying parts of the human immune system rather than the mouse system. These transgenic and transchromosomic mice include mice referred to herein as the HuMAb Mouse® (Medarex, Inc.), KM Mouse® (Medarex, Inc.), and XenoMouse® (Amgen).

Moreover, alternative transchromosomic animal systems expressing human immunoglobulin genes are available in the art and can be used to raise antibodies of the disclosure. For example, mice carrying both a human heavy chain transchromosome and a human light chain tranchromosome, referred to as “TC mice” can be used; such mice are described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97:722-727. Furthermore, cows carrying human heavy and light chain transchromosomes have been described in the art (e.g., Kuroiwa et al. (2002) Nature Biotechnology 20:889-894 and PCT application No. WO 2002/092812) and can be used to raise the antibodies of this disclosure.

In certain embodiments, the antibodies of this disclosure are recombinant human antibodies, which can be isolated by screening of a recombinant combinatorial antibody library, e.g., a scFv phage display library, prepared using human VL and VH cDNAs prepared from mRNA derived from human lymphocytes. Methodologies for preparing and screening such libraries are known in the art. In addition to commercially available kits for generating phage display libraries (e.g., the Pharmacia Recombinant Phage Antibody System, catalog no. 27-9400-01; and the Stratagene SurfZAP™ phage display kit, catalog no. 240612, the entire teachings of which are incorporated herein), examples of methods and reagents particularly amenable for use in generating and screening antibody display libraries can be found in, e.g., Ladner et al. U.S. Pat. No. 5,223,409; Kang et al. PCT Publication No. WO 92/18619; Dower et al. PCT Publication No. WO 91/17271; Winter et al. PCT Publication No. WO 92/20791; Markland et al. PCT Publication No. WO 92/15679; Breitling et al. PCT Publication No. WO 93/01288; McCafferty et al. PCT Publication No. WO 92/01047; Garrard et al. PCT Publication No. WO 92/09690; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum Antibod Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; McCafferty et al., Nature (1990) 348:552-554; Griffiths et al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) J Mol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) PNAS 89:3576-3580; Garrard et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS 88:7978-7982; the entire teachings of which are incorporated herein.

Human monoclonal antibodies of this disclosure can also be prepared using SCID mice into which human immune cells have been reconstituted such that a human antibody response can be generated upon immunization. Such mice are described in, for example, U.S. Pat. Nos. 5,476,996 and 5,698,767 to Wilson et al.

The antibodies or antigen-binding portions thereof, of this disclosure can be altered wherein the constant region of the antibody is modified to reduce at least one constant region-mediated biological effector function relative to an unmodified antibody. To modify an antibody of the invention such that it exhibits reduced binding to the Fc receptor, the immunoglobulin constant region segment of the antibody can be mutated at particular regions necessary for Fc receptor (FcR) interactions (see, e.g., Canfield and Morrison (1991) J. Exp. Med. 173:1483-1491; and Lund et al. (1991) J. of Immunol. 147:2657-2662, the entire teachings of which are incorporated herein). Reduction in FcR binding ability of the antibody may also reduce other effector functions which rely on FcR interactions, such as opsonization and phagocytosis and antigen-dependent cellular cytotoxicity.

5.3. Antibody Production

To express an antibody of the invention, DNAs encoding partial or full-length light and heavy chains are inserted into one or more expression vector such that the genes are operatively linked to transcriptional and translational control sequences. (See, e.g., U.S. Pat. No. 6,914,128, the entire teaching of which is incorporated herein by reference.) In this context, the term “operatively linked” is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene. The expression vector and expression control sequences are chosen to be compatible with the expression host cell used. The antibody light chain gene and the antibody heavy chain gene can be inserted into a separate vector or, more typically, both genes are inserted into the same expression vector. The antibody genes are inserted into an expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present). Prior to insertion of the antibody or antibody-related light or heavy chain sequences, the expression vector may already carry antibody constant region sequences. Additionally or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell. The antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).

In addition to the antibody chain genes, a recombinant expression vector of the invention can carry one or more regulatory sequence that controls the expression of the antibody chain genes in a host cell. The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes. Such regulatory sequences are described, e.g., in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990), the entire teaching of which is incorporated herein by reference. It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Suitable regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma. For further description of viral regulatory elements, and sequences thereof, see, e.g., U.S. Pat. No. 5,168,062 by Stinski, U.S. Pat. No. 4,510,245 by Bell et al. and U.S. Pat. No. 4,968,615 by Schaffner et al., the entire teachings of which are incorporated herein by reference.

In addition to the antibody chain genes and regulatory sequences, a recombinant expression vector of the invention may carry one or more additional sequences, such as a sequence that regulates replication of the vector in host cells (e.g., origins of replication) and/or a selectable marker gene. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al., the entire teachings of which are incorporated herein by reference). For example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced. Suitable selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).

An antibody of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell. To express an antibody recombinantly, a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody such that the light and heavy chains are expressed in the host cell and secreted into the medium in which the host cells are cultured, from which medium the antibodies can be recovered. Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Fritsch and Maniatis (eds), Molecular Cloning; A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Ausubel et al. (eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, (1989) and in U.S. Pat. Nos. 4,816,397 & 6,914,128, the entire teachings of which are incorporated herein.

For expression of the light and heavy chains, the expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques. The various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like. Although it is theoretically possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells, such as mammalian host cells, is suitable because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody. Prokaryotic expression of antibody genes has been reported to be ineffective for production of high yields of active antibody (Boss and Wood (1985) Immunology Today 6:12-13, the entire teaching of which is incorporated herein by reference).

Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above. Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, e.g., Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B. licheniformis 41P disclosed in DD 266,710 published Apr. 12, 1989), Pseudomonas such as P. aeruginosa, and Streptomyces. One suitable E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.

In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide encoding vectors. Saccharomyces cerevisiae, or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms. However, a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. waltii (ATCC 56,500), K. drosophilarum (ATCC 36,906), K. thermotolerans, and K. marxianus; yarrowia (EP 402,226); Pichia pastoris (EP 183,070); Candida; Trichoderma reesia (EP 244,234); Neurospora crassa; Schwanniomyces such as Schwanniomyces occidentalis; and filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.

Suitable host cells for the expression of glycosylated antibodies are derived from multicellular organisms. Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified. A variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.

Suitable mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Urlaub and Chasin, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference), NSO myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2), the entire teachings of which are incorporated herein by reference.

Host cells are transformed with the above-described expression or cloning vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.

The host cells used to produce an antibody may be cultured in a variety of media. Commercially available media such as Ham's F10™ (Sigma), Minimal Essential Medium™ ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium™ ((DMEM), Sigma) are suitable for culturing the host cells. In addition, any of the media described in Ham et al., Meth. Enz. 58:44 (1979), Barnes et al., Anal. Biochem. 102:255 (1980), U.S. Pat. Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; or 5,122,469; WO 90/03430; WO 87/00195; or U.S. Pat. No. Re. 30,985 may be used as culture media for the host cells, the entire teachings of which are incorporated herein by reference. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.

Host cells can also be used to produce portions of intact antibodies, such as Fab fragments or scFv molecules. It is understood that variations on the above procedure are within the scope of the present invention. For example, in certain embodiments it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain (but not both) of an antibody of this invention. Recombinant DNA technology may also be used to remove some or all of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigen to which the putative antibody of interest binds. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the one to which the putative antibody of interest binds, depending on the specificity of the antibody of the invention, by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.

In a suitable system for recombinant expression of an antibody of the invention, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr-CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.

When using recombinant techniques, the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. In one aspect, if the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed cells (e.g., resulting from homogenization), can be removed, e.g., by centrifugation or ultrafiltration. Where the antibody is secreted into the medium, supernatants from such expression systems can be first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit.

Prior to the process of the invention, procedures for purification of antibodies from cell debris initially depend on the site of expression of the antibody. Some antibodies can be secreted directly from the cell into the surrounding growth media; others are made intracellularly. For the latter antibodies, the first step of a purification process typically involves: lysis of the cell, which can be done by a variety of methods, including mechanical shear, osmotic shock, or enzymatic treatments. Such disruption releases the entire contents of the cell into the homogenate, and in addition produces subcellular fragments that are difficult to remove due to their small size. These are generally removed by differential centrifugation or by filtration. Where the antibody is secreted, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit. Where the antibody is secreted into the medium, the recombinant host cells can also be separated from the cell culture medium, e.g., by tangential flow filtration. Antibodies can be further recovered from the culture medium using the antibody purification methods of the invention.

5.4. Antibody Purification

5.4.1 Antibody Purification Generally

In certain embodiments, the invention provides methods and compositions for producing a purified or partially purified (e.g., process-related impurity-reduced and/or product-related substance-reduced) protein preparation from a mixture comprising a protein of interest, e.g., an antibody, and at least one process-related impurity or product-related substance. In certain embodiments, the compositions of the present invention include, but are not limited to, process-related impurity-reduced and/or product-related substance-reduced compositions comprising a protein of interest. For example, but not by way of limitation, the present invention is directed to process-related impurity-reduced and/or product-related substance-reduced compositions comprising Adalimumab. Such process-related impurity-reduced and/or product-related substance-reduced compositions process-related impurity-reduced and/or product-related substance-reduced compositions address the need for improved product characteristics, including, but not limited to, product stability, product safety and product efficacy.

In certain embodiments, the present invention is directed to a method for preparing a process-related impurity-reduced and/or product-related substance-reduced composition comprising a protein of interest wherein a chromatographic separation is performed to identify the particular conditions, e.g., salt concentration, pH, temperature, load amount and conditions, and washing conditions, sufficient to elicit the desired fractionation profile of a sample comprising the protein of interest and at least one process-related impurity and/or at least one product-related substance. In certain embodiments, the method will further comprise pooling of the resulting fractions comprising the desired process-related impurity-reduced and/or product-related substance-reduced composition comprising a protein of interest.

In certain embodiments, the purification process of the invention begins at the separation step when the antibody has been produced using production methods described above and/or by alternative production methods conventional in the art. Once a clarified solution or mixture comprising the antibody has been obtained, separation of the protein of interest from process-related impurities, such as the other proteins produced by the cell, as well as any product-related substances such as charge variants and/or size variants (aggregates and fragments), can be performed using a HIC separation step, or a combination of a HIC separation step and one or more purification techniques, including filtration and/or affinity, ion exchange, and/or mixed mode chromatographic step(s), as outlined herein. The essence of each of the chromatographic separation methods is that proteins can be caused either to traverse at different rates down a column, achieving a physical separation that increases as they pass further down the column, or to adhere selectively to the separation medium, being then differentially eluted by different solvents. In some cases, the protein of interest is separated from impurities and or product-related substances when the impurities and/or product-related substances specifically adhere to the column and the protein of interest does not, i.e., the protein of interest is washed from the column, while in other cases the protein of interest will adhere to the column, while the impurities and/or product-related substances are washed from the column.

5.4.2 Primary Recovery

In certain embodiments, the initial steps of the purification methods of the present invention involve the clarification and primary recovery of antibody from a sample matrix. In certain embodiments, the primary recovery will include one or more centrifugation steps to separate the antibody product from the cells and cell debris. Centrifugation of the sample can be run at, for example, but not by way of limitation, 7,000×g to approximately 12,750×g. In the context of large scale purification, such centrifugation can occur on-line with a flow rate set to achieve, for example, but not by way of limitation, a turbidity level of 150 NTU in the resulting supernatant. Such supernatant can then be collected for further purification, or in-line filtered through one or more depth filters for further clarification of the sample.

In certain embodiments, the primary recovery will include the use of one or more depth filtration steps to clarify the sample matrix and thereby aid in purifying the antibodies of interest in the present invention. In other embodiments, the primary recovery will include the use of one or more depth filtration steps post centrifugation to further clarify the sample matrix. Non-limiting examples of depth filters that can be used in the context of the instant invention include the Millistak+ X0HC, F0HC, D0HC, A1HC, B1HC depth filters (EMD Millipore), Cuno™ model 30/60ZA, 60/90 ZA, VR05, VR07, delipid depth filters (3M Corp.). A 0.2 μm filter such as Sartorius's 0.45/0.2 μm Sartopore™ bi-layer or Millipore's Express SHR or SHC filter cartridges typically follows the depth filters.

In certain embodiments, the primary recovery process can also be a point at which to reduce or inactivate viruses that can be present in the sample matrix. For example, any one or more of a variety of methods of viral reduction/inactivation can be used during the primary recovery phase of purification including heat inactivation (pasteurization), pH inactivation, solvent/detergent treatment, UV and Tray irradiation and the addition of certain chemical inactivating agents such as (3-propiolactone or e.g., copper phenanthroline as in U.S. Pat. No. 4,534,972. In certain embodiments of the present invention, the sample matrix is exposed to detergent viral inactivation during the primary recovery phase. In other embodiments, the sample matrix may be exposed to low pH inactivation during the primary recovery phase.

In those embodiments where viral reduction/inactivation is employed, the sample mixture can be adjusted, as needed, for further purification steps. For example, following low pH viral inactivation, the pH of the sample mixture is typically adjusted to a more neutral pH, e.g., from about 4.5 to about 8.5, prior to continuing the purification process. Additionally, the mixture may be diluted with water for injection (WFI) to obtain a desired conductivity.

5.4.3 Hydrophobic Interaction Chromatography

In certain embodiments, the instant invention features methods for producing a process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest, e.g., an antibody, and at least one process-related impurity and/or product-related substance comprising a hydrophobic interaction chromatography (HIC) step.

In performing the HIC separation, the sample mixture is contacted with the HIC material, e.g., using a batch purification technique or using a column or membrane chromatography or monolithic material (referred to as HIC media). Prior to HIC purification it may be desirable to adjust the concentration of the salt concentration in the buffer to achieve desired protein binding to the HIC media.

For example, in the context of batch purification, HIC material is prepared in or equilibrated with a desired equilibration buffer. A slurry of the HIC material is obtained. The protein solution is contacted with the slurry to allow protein adsorption to the HIC material. The solution comprising the process-related impurities and/or product-related substances that do not bind to the HIC material is separated from the slurry, e.g., by allowing the slurry to settle and removing the supernatant. The slurry can be subjected to one or more washing steps and/or elution steps.

In the context of chromatographic separation, a chromatographic apparatus, commonly cylindrical in shape, is employed to contain the chromatographic support material (e.g., HIC material) prepared in an appropriate buffer solution. The chromatographic apparatus, if cylindrical, can have a diameter of about 5 mm to about 2 meters, and a height of 5 cm to 50 cm, and in certain embodiments, particularly for large scale processing, a height of ≦30 cm is employed. Once the chromatographic material is added to the chromatographic apparatus, a sample containing the protein of interest, e.g., an antibody, is contacted to the chromatographic material to induce the separation. Any portion of the solution that does not bind to the chromatographic material, e.g., which may comprise, depending on the HIC material being employed, the protein of interest, process-related impurities, and/or product-related substances, is separated from the chromatographic material by washing the material and collecting fractions from column. The chromatographic material can be subjected to one or more wash steps. If desired, the chromatographic material can then be contacted with a solution of designed to desorb any components of the solution that have bound to the chromatographic material.

Hydrophobic interaction chromatography employs the hydrophobic properties of proteins to achieve separation of even closely-related molecules. Hydrophobic groups on the protein interact with hydrophobic groups of the media or the membrane. In certain embodiments, the more hydrophobic a protein is the stronger it will interact with the column or the membrane. Thus, HIC steps, such as those disclosed herein, can be used to remove a variety of process-related impurities (e.g., DNA) as well as product-related species (e.g., high and low molecular weight product-related species, such as protein aggregates and fragments).

An HIC column or membrane device can be operated in a bind-elute mode, a flow-through mode, or a hybrid mode wherein the product exhibits binding to the chromatographic material, yet can be washed from the column using a buffer that is the same or substantially similar to the loading buffer. The bind-elute mode of operation has been explained above in connection with batch purification. For flow-through, process-related impurities, such as HCPs, and product-related substances, such as aggregates, will, in certain embodiments, depending the particular HIC material employed, bind to the HIC media while product flows through the column. A hybrid mode, in contrast, can involve the use of an HIC media that allows for the product to be immobilized on the chromatographic support in the presence of a loading buffer, but then removed by successive washes of buffer identical to or substantially similar, for example, but not by way of limitation where the salt concentration is adjusted within about 20% of the concentration of the loading buffer. In certain embodiments, a step-wise or linear change in wash conductivity can be used. In the context of this hybrid strategy, process-related impurities and product-relates substances will either bind to the chromatographic material or flow through with a profile distinct from the protein of interest. After loading, the column can be regenerated with water and cleaned with caustic solution to remove the bound impurities before next use.

Hydrophobic interactions are strongest at high salt concentration (and hence the ionic strength of the anion and cation components), therefore, this form of separation is conventionally performed following salt elution step, e.g., the type of elution step typically used in connection with ion exchange chromatography. Adsorption of the antibody to a HIC column is favored by high salt concentrations, but the actual concentrations can vary over a wide range depending on the nature of the protein of interest, salt type and the particular HIC ligand chosen. For example, and not by way of limitation, the salt concentrations shown to be effective in aggregate reduction are in the range of 80 mM-1000 mM, depending on the salt type and HIC adsorbent. Various ions can be arranged in a so-called soluphobic series depending on whether they promote hydrophobic interactions (salting-out effects) or disrupt the structure of water (chaotropic effect) and lead to the weakening of the hydrophobic interaction. Cations are ranked in terms of increasing salting out effect as Ba2+; Ca2+; Mg2+; Li+; Cs+; Na+; K+; Rb+; NH4+, while anions may be ranked in terms of increasing chaotropic effect as PO43−; SO42−; CH3CO3; Cl; Br; NO3; ClO4; I; SCN.

In certain embodiments, the anionic part of the salt is chosen from among sulfate, citrate, chloride, or a mixture thereof. In certain embodiments, the cationic part of the salt is chosen from among ammonium, sodium, potassium, or a mixture thereof. In general, Na+, K+ or NH4+ sulfates effectively promote ligand-protein interaction in HIC. Salts may be formulated that influence the strength of the interaction as given by the following relationship: (NH4)2SO4>Na2SO4>NaCl>NH4Cl>NaBr>NaSCN. In general, salt concentrations of between about 0.75 and about 2 M ammonium sulfate or between about 1 and 4 M NaCl are useful.

In certain embodiments, the HIC adsorbent material is composed of a chromatographic backbone with pendant hydrophobic interaction ligands. For example, by not by way of limitation, the HIC adsorbent material can be composed of convective membrane media with pendent hydrophobic interaction ligands, convective monolithic media with pendent hydrophobic interaction ligands, and/or convective filter media with embedded media media containing the pendant hydrophobic interaction ligands. In certain embodiments, the HIC adsorbent material can comprise a base matrix (e.g., cross-linked agarose or synthetic copolymer material) to which hydrophobic ligands (e.g., alkyl or aryl groups) are coupled. One, non-limiting, example of a suitable HIC media comprises an agarose media or a membrane functionalized with phenyl groups (e.g., a Phenyl Sepharose™ from GE Healthcare or a Phenyl Membrane from Sartorius). Many HIC medias are available commercially. Examples include, but are not limited to, Capto Phenyl, Phenyl Sepharose™ 6 Fast Flow with low or high substitution, Phenyl Sepharose™ High Performance, Octyl Sepharose™ High Performance (GE Healthcare); Fractogel™ EMD Propyl or Fractogel™ EMD Phenyl (E. Merck, Germany); Macro-Prep™ Mehyl or Macro-Prep™ t-Butyl columns (Bio-Rad, California); WP HI-Propyl (C3)™ (J. T. Baker, New Jersey); Toyopearl™ ether, phenyl or butyl (TosoHaas, PA); ToyoScreen PPG, ToyoScreen Phenyl, ToyoScreen Butyl, and ToyoScreen Hexyl are a rigid methacrylic polymer bead. GE HiScreen Butyl FF and HiScreen Octyl FF are high flow agarose based beads.

In certain embodiments, the protein load of the mixture comprising protein of interest is adjusted to a total protein load to the column of between about 50 and 1000 g/L, or between about 250 and 700 g/L, or between about 350 and 500 g/L. In certain embodiments, the protein concentration of the load protein mixture is adjusted to a protein concentration of the material loaded to the column of about 0.5 and 30 g/L, or between about 1 and 20 g/L, or between about 3 and 10 g/L.

In certain embodiments, aggregate concentration is measured and used to as a parameter for controlling aggregate clearance in the present invention. For example, but not by way of limitation, the data presented in the Examples below, supports the novel finding that aggregation concentration influences the aggregate reduction by hydrophobic interaction chromatography. Thus, in certain embodiments, the aggregate concentration is adjusted from about 0.5 to 0.1 g/L, to about 0.1 to 0.05 g/L or to below 0.05 g/L.

In certain embodiments, monomer concentration is measured and used to as a parameter for controlling aggregate clearance in the present invention. For example, but not by way of limitation, the data presented in the Examples below supports the novel finding that control of the concentration of the monomer can be used to achieve improved aggregate clearance. Thus, in certain embodiments, the monomer concentration is adjusted from about 15 to 8 g/L, to about 8 to 4 g/L or to below 4 g/L.

In certain embodiments, monomer and aggregate concentration is measured and used to as a parameter for controlling aggregate clearance in the present invention. For example, but not by way of limitation, the data presented in the Examples below supports the novel finding that control of the monomer and aggregate concentrations within certain ranges can be used to achieve improved aggregate clearance. Thus, in certain embodiments, the monomer concentration is adjusted from about 20 to 15 g/L, about 15 to 8 g/L or to below 4 g/L and the aggregate concentration is adjusted to 0.5 to 0.1 g/L, about 0.1 to 0.05 g/L or to below 0.05 g/L to achieve aggregate reduction in the present invention.

Because the pH selected for any particular purification process must be compatible with protein stability and activity, thus particular pH conditions may be specific for each application. However, because at pH 5-8.5, particular pH values have very little significance on the final selectivity and resolution of a HIC separation, such conditions may be favored. An increase in pH weakens hydrophobic interactions and retention of proteins changes more drastically at pH values above 8.5 or below 5.0. In addition, changes in ionic strength, the presence of organic solvents, temperature and pH (especially at the isoelectric point, pI, when there is no net surface charge) can impact protein structure and solubility and, consequently, the interaction with other hydrophobic surfaces, such as those in HIC media and hence, in certain embodiments, the present invention incorporates purification strategies wherein one or more of the foregoing are adjusted to achieve the desired reduction in process-related impurities and/or product-related substances.

In certain embodiments of the instant invention, control of molecular weight heterogeneity can be attained by purifying a protein of interest from a mixture comprising the protein with adsorbent material containing HIC functional group (also referred to as “HIC media”) and an aqueous salt solution under loading conditions that permits both the protein of interest and non-target proteins to partially bind to the HIC adsorbent, and thereafter flowing through and washing off the protein of interest that is now reduced in aggregate species and impurities. In certain embodiments, such washing steps will employ similar conditions as the loading solution. In certain embodiments, product/process-related substances/impurities as well as bound protein with enhanced aggregate content can be eluted from the HIC adsorbent with a solution having a lower conductivity than the loading solution.

In certain embodiments, HIC chromatographic fractions are collected during the load, wash, and/or elution, and are combined after appropriate analysis to provide a protein preparation that contains the desired, e.g., reduced, level of aggregate species. In certain embodiments, the load pool is combined with certain wash fractions to improve the yield of the process while still achieving the desired, e.g., reduced, aggregate levels in the resulting material.

In certain embodiments, the wash step(s) employed in the context of HIC chromatography can be performed using conditions similar to the load conditions or alternatively by decreasing the conductivity of the wash in a step-wise or linear gradient manner.

In certain embodiments, spectroscopy methods such as UV, NIR, FTIR, Fluorescence, Raman may be used to monitor levels of aggregates and low molecular weight variants (e.g., fragments of the protein of interest) in an on-line, at-line or in-line mode, which can then be used to control the level of aggregates in the pooled material collected from the HIC adsorbent effluent. In certain embodiments, on-line, at-line or in-line monitoring methods can be used either on the effluent line of the chromatography step or in the collection vessel, to enable achievement of the desired product quality/recovery. In certain embodiments, the UV signal can be used as a surrogate to achieve an appropriate product quality/recovery, wherein the UV signal can be processed appropriately, including, but not limited to, such processing techniques as integration, differentiation, moving average, such that normal process variability can be addressed and the target product quality can be achieved. In certain embodiments, such measurements can be combined with in-line dilution methods such that ion concentration/conductivity of the load/wash can be controlled by feedback and hence facilitate product quality control.

In certain embodiments, a combination of HIC (hydrophobic interaction chromatography) and AEX (anion exchange chromatography) and CEX (cation exchange chromatography) and MM (mixed mode chromatography) methods can be used to prepare product-related aggregate- and/or fragment-reduced materials, including certain embodiments where one technology is used in a complementary/supplementary manner with another technology. In certain embodiments, such a combination can be performed such that certain sub-species are removed predominantly by one technology, such that the combination provides the desired final composition/product quality. In certain embodiments, such combinations include the use of additional intervening chromatography, filtration, pH adjustment, UF/DF (ultrafiltration/diafiltration) steps so as to achieve the desired product quality, ion concentration, and/or viral reduction.

5.4.4 Affinity Chromatography

In certain embodiments, the sample matrix is subjected to affinity chromatography to purify the antibody of interest away from process-related impurities and/or product-related substances. As noted above, certain embodiments of the present in invention will employ one or more affinity chromatography steps prior to the HIC purification step, while others will employ an affinity chromatography step after or both before and after the HIC purification step. In certain embodiments, the affinity chromatography media is a Protein A, G, A/G, or L media, although alternative affinity chromatography medias are known in the art. There are a variety of commercial sources for Protein A media. Suitable medias include, but not limited to, MabSelect SuRe™, MabSelect SuRe LX, MabSelect, MabSelect Xtra, rProtein A Sepharose from GE Healthcare, ProSep HC, ProSep Ultra, and ProSep Ultra Plus from EMD Millipore, MapCapture from Life Technologies.

In certain embodiments, the Protein A column can be equilibrated with a suitable buffer prior to sample loading. Following the loading of the column, the column can be washed one or multiple times using a suitable sets of buffers. The Protein A column can then be eluted using an appropriate elution buffer. The eluate can be monitored using techniques well known to those skilled in the art. The eluate fractions of interest can be collected and then prepared for further processing.

The Protein A eluate may subject to a viral inactivation step either by detergent or low pH, provided this step is not performed prior to the Protein A capture operation. A proper detergent concentration or pH and time can be selected to obtain desired viral inactivation results. After viral inactivation, the Protein A eluate is usually pH and/or conductivity adjusted for subsequent purification steps.

The Protein A eluate may be subjected to filtration through a depth filter to remove turbidity and/or various impurities from the antibody of interest prior to additional chromatographic polishing steps. Examples of depth filters include, but not limited to, Millistak+ X0HC, F0HC, D0HC, A1HC, and B1HC Pod filters (EMD Millipore), or Zeta Plus 30ZA/60ZA, 60ZA/90ZA, delipid, VR07, and VRO5 filters (3M). The Protein A eluate pool may need to be conditioned to proper pH and conductivity to obtain desired impurity removal and product recovery from the depth filtration step.

5.4.5 Ion Exchange Chromatography

In certain embodiments, the instant invention provides methods for producing process-related impurity and/or product-related substance-reduced protein preparation from a mixture comprising a protein of interest (i.e., a product) and at least one process-related impurity and/or product-related substance by subjecting the mixture to at least one ion exchange separation step in addition to the HIC step described above. In certain embodiments, the ion exchange step will occur after the above-described Protein A affinity and/or HIC chromatography steps, such that an eluate comprising the protein of interest is obtained. In addition, an ion exchange separation step can be employed after the HIC purification step. As used herein, ion exchange separations includes any method by which two substances are separated based on the difference in their respective ionic charges, either on the protein of interest and/or chromatographic material as a whole or locally on specific regions of the protein of interest and/or chromatographic material, and thus can employ either cationic exchange material or anionic exchange material.

The use of a cationic exchange material versus an anionic exchange material is based on the local charges of the protein of interest in a given solution. Therefore, it is within the scope of this invention to employ an anionic exchange step prior to the use of a HIC step, or a cationic exchange step prior to the use of an HIC step. Furthermore, it is within the scope of this invention to employ only a cationic exchange step, only an anionic exchange step, or any serial combination of the two either prior to or subsequent to the HIC step.

In performing the separation, the antibody sample mixture can be contacted with the ion exchange material by using any of a variety of techniques, e.g., using a batch purification technique or a chromatographic technique, as described above in connection with HIC.

Ion exchange chromatography separates molecules based on differences between the local charges of the proteins of interest and the local charges of the chromatographic material. A packed ion-exchange chromatography column or an ion-exchange membrane device can be operated in a bind-elute mode, a flow-through, or a hybrid mode as discussed above in connection with HIC. After washing the column or the membrane device with the equilibration buffer or another buffer with different pH and/or conductivity, the product recovery is achieved by increasing the ionic strength (i.e., conductivity) of the elution buffer to compete with the solute for the charged sites of the ion exchange matrix. Changing the pH and thereby altering the charge of the solute is another way to achieve elution of the solute. The change in conductivity or pH may be gradual (gradient elution) or stepwise (step elution). The column is then regenerated before next use.

Anionic or cationic substituents may be attached to matrices in order to form anionic or cationic supports for chromatography. Non-limiting examples of anionic exchange substituents include diethylaminoethyl (DEAE), quaternary aminoethyl (QAE) and quaternary amine (Q) groups. Cationic substitutents include carboxymethyl (CM), sulfoethyl (SE), sulfopropyl (SP), phosphate (P) and sulfonate (S). Cellulose ion exchange medias such as DE23™, DE32™, DE52™, CM-23™, CM-32™, and CM-52™ are available from Whatman Ltd. Maidstone, Kent, U.K. SEPHADEX®-based and -locross-linked ion exchangers are also known. For example, DEAE-, QAE-, CM-, and SP-SEPHADEX® and DEAE-, Q-, CM- and S-SEPHAROSE® and SEPHAROSE® Fast Flow, and Capto™ S are all available from GE Healthcare. Further, both DEAE and CM derivitized ethylene glycol-methacrylate copolymer such as TOYOPEARL™ DEAE-6505 or M and TOYOPEARL™ CM-650S or M are available from Toso Haas Co., Philadelphia, Pa., or Nuvia S and UNOSphere™ S from BioRad, Hercules, Calif., Eshmuno® S from EMD Millipore, Billerica, Calif.

5.4.6 Mixed Mode Chromatography

The present invention also features methods for purifying a protein preparation from a mixture comprising a protein of interest comprising a mixed mode chromatography separation step. Mixed mode chromatography is chromatography that utilizes a mixed mode media, such as, but not limited to CaptoAdhere available from GE Healthcare. Such a media comprises a mixed mode chromatography ligand. In certain embodiments, such a ligand refers to a ligand that is capable of providing at least two different, but co-operative, sites which interact with the substance to be bound. One of these sites gives an attractive type of charge-charge interaction between the ligand and the substance of interest. The other site typically gives electron acceptor-donor interaction and/or hydrophobic and/or hydrophilic interactions. Electron donor-acceptor interactions include interactions such as hydrogen-bonding, π-π, cation-π, charge transfer, dipole-dipole, induced dipole etc. The mixed mode functionality can give a different selectivity compared to traditional anion exchangers. For example, CaptoAdhere is designed for post-Protein A purification of monoclonal antibodies, where removal of leached Protein A, aggregates, host cell proteins, nucleic acids and viruses from monoclonal antibodies is performed in flow-through mode (the antibodies pass directly through the column while the contaminants are adsorbed). Mixed mode chromatography ligands are also known as “multimodal” chromatography ligands.

In certain embodiments, the mixed mode chromatography media is comprised of mixed mode ligands coupled to an organic or inorganic support, sometimes denoted a base matrix, directly or via a spacer. The support may be in the form of particles, such as essentially spherical particles, a monolith, filter, membrane, surface, capillaries, etc. In certain embodiments, the support is prepared from a native polymer, such as cross-linked carbohydrate material, such as agarose, agar, cellulose, dextran, chitosan, konjac, carrageenan, gellan, alginate etc. To obtain high adsorption capacities, the support can be porous, and ligands are then coupled to the external surfaces as well as to the pore surfaces. Such native polymer supports can be prepared according to standard methods, such as inverse suspension gelation (S Hjerten: Biochim Biophys Acta 79(2), 393-398 (1964). Alternatively, the support can be prepared from a synthetic polymer, such as cross-linked synthetic polymers, e.g. styrene or styrene derivatives, divinylbenzene, acrylamides, acrylate esters, methacrylate esters, vinyl esters, vinyl amides etc. Such synthetic polymers can be produced according to standard methods, see e.g. “Styrene based polymer supports developed by suspension polymerization” (R Arshady: Chimica e L′Industria 70(9), 70-75 (1988)). Porous native or synthetic polymer supports are also available from commercial sources, such as Amersham Biosciences, Uppsala, Sweden.

5.4.7 Viral Filtration

Viral filtration is a dedicated viral reduction step in the entire purification process. This step is usually performed post chromatographic polishing steps. Viral reduction can be achieved via the use of suitable filters including, but not limited to, Planova 20N™, 50 N or BioEx from Asahi Kasei Pharma, Viresolve™ filters from EMD Millipore, ViroSart CPV from Sartorius, or Ultipor DV20 or DV50™ filter from Pall Corporation. It will be apparent to one of ordinary skill in the art to select a suitable filter to obtain desired filtration performance.

5.4.8 Ultrafiltration/Diafiltration

Certain embodiments of the present invention employ ultrafiltration and/or diafiltration steps to further purify and concentrate the antibody sample. Ultrafiltration is described in detail in: Microfiltration and Ultrafiltration: Principles and Applications, L. Zeman and A. Zydney (Marcel Dekker, Inc., New York, N.Y., 1996); and in: Ultrafiltration Handbook, Munir Cheryan (Technomic Publishing, 1986; ISBN No. 87762-456-9). A preferred filtration process is Tangential Flow Filtration as described in the Millipore catalogue entitled “Pharmaceutical Process Filtration Catalogue” pp. 177-202 (Bedford, Mass., 1995/96). Ultrafiltration is generally considered to mean filtration using filters with a pore size of smaller than 0.1 μm. By employing filters having such small pore size, the volume of the sample can be reduced through permeation of the sample buffer through the filter while antibodies are retained behind the filter.

Diafiltration is a method of using ultrafilters to remove and exchange salts, sugars, and non-aqueous solvents, to separate free from bound species, to remove low molecular-weight material, and/or to cause the rapid change of ionic and/or pH environments. Microsolutes are removed most efficiently by adding solvent to the solution being ultrafiltered at a rate approximately equal to the ultratfiltration rate. This washes microspecies from the solution at a constant volume, effectively purifying the retained antibody. In certain embodiments of the present invention, a diafiltration step is employed to exchange the various buffers used in connection with the instant invention, optionally prior to further chromatography or other purification steps, as well as to remove impurities from the antibody preparations.

5.4.9 Exemplary Purification Strategies

In certain embodiments, primary recovery can proceed by sequentially employing pH reduction, centrifugation, and filtration steps to remove cells and cell debris (including HCPs) from the production bioreactor harvest. In certain embodiments, the present invention is directed to HIC purification steps where an HIC media partially binds antibody product as well as product-related substances/impurities and process-related impurities during the loading step with some antibody product collected during the loading step in the unbound fraction and when the column is subjected to a subsequent wash buffer with substantially similar composition, the antibody product reduced in the said impurities/substances are collected in the wash fraction. The antibody product in the flow through and wash fractions represent the purified or impurity reduced materials. The antibody product is specifically eluted while the product-related and process-related impurities are retained on the media. Examples of buffers that can be used in the context of both the loading and elution steps of the present invention include, but are not limited to, the following: about 0.1 M to about 0.6 M sodium citrate (NaCit), pH 5.6; or about 0.5 M to about 1.1 M ammonium sulfate (AmSO4), pH 7.0 as well as buffers substantially similar, in that any differences are result in insubstantial changes to the elution of product-related or process-related impurities, yet retain the ability to elute antibody product. Such buffers can span a range of varying “hydrophobicities” based on the rationales discussed in section 4.4.3, above.

In certain embodiments, the HIC media employed in the HIC step is CaptoPhenyl (GE) resin. In certain embodiments, the CaptoPhenyl (GE) resin is buffer exchanged into 0.4 M sodium citrate (NaCit), pH 5.6, and then distributed in 100 μL aliquots into microcentrifuge tubes. Each tube is then challenged with 2 mL of antibody produce source material, e.g., a partially purified cell culture harvest sample, in 0.4 M NaCit, pH 5.6, at a range of concentrations from 0.5-15.0 mg/mL and incubated for 3 hours at room temperature with mixing. The resin is allowed to settle and the supernatant removed and replaced with 1 mL of fresh 0.4 M NaCit, pH 5.6, buffer and incubated for 2 hours at room temperature with mixing. This step was repeated one more time.

In alternative embodiments, the CaptoPhenyl (GE) HIC resin can be packed in 1.0 cm×10.0 cm (OmniFit) columns. Antibody product HIC-load can be prepared by diluting the source material, e.g., a partially purified cell culture harvest sample, with a 1.2 M stock solution of sodium citrate (NaCit), pH 5.6, to final concentration in the range of 0.3 to 0.5 M NaCit, pH 5.6. CaptoPhenyl columns can then be equilibrated with 7 column volumes (CVs) of a NaCit buffer, pH 5.6, corresponding to the load concentration. The antibody product solution can then be loaded to the column in the range of 200-500 g/L, after which the column is washed with 20 CVs of the equilibration buffer. The column can then be regenerated (3 CVs f 25 mM sodium phosphate/20% (v/v) isopropyl alcohol, pH 6.5), cleaned in place (3 CVs 1M NaOH, 60 min hold), and stored (5 CVs of 25 mM sodium phosphate/20% (v/v) isopropyl alcohol, pH 6.5). The effluent from the column can be fractionated during the entire run and used to monitor the breakthrough of both the antibody product monomer as well as product and process related impurities; namely aggregates and host cell protein (HCP).

Such HIC purification steps can be preceded by affinity chromatography, for example, but not limited to, the use of Protein A-base affinity chromatography. There are several commercial sources for Protein A media. One suitable media is MabSelect™ from GE Healthcare. An example of a suitable column packed with MabSelect™ is a column about 1.0 cm diameter×about 21.6 cm long (˜17 mL bed volume). This size column can be used for bench scale. This can be compared with other columns used for scale ups. For example, a 20 cm×21 cm column whose bed volume is about 6.6 L can be used for commercial production. Regardless of the column, the column can be packed using a suitable media such as MabSelect™.

In certain aspects, the Protein A column can be equilibrated with a suitable buffer prior to sample loading. An example of a suitable buffer is a Tris/NaCl buffer, pH of about 6 to 8, and in certain embodiments about 7.2. A specific example of suitable conditions is 25 mM Tris, 100 mM NaCl, pH 7.2. Following this equilibration, the sample can be loaded onto the column. Following the loading of the column, the column can be washed one or multiple times using, e.g., the equilibrating buffer. Other washes including washes employing different buffers can be used before eluting the column. For example, the column can be washed using one or more column volumes of 20 mM citric acid/sodium citrate, 0.5 M NaCl at pH of about 6.0. This wash can optionally be followed by one or more washes using the equilibrating buffer. The Protein A column can then be eluted using an appropriate elution buffer. An example of a suitable elution buffer is an acetic acid/NaCl buffer, pH around 3.5. Suitable conditions are, e.g., 0.1 M acetic acid, pH 3.5. The eluate can be monitored using techniques well known to those skilled in the art. For example, the absorbance at OD280 can be followed. Column eluate can be collected starting with an initial deflection of about 0.5 AU to a reading of about 0.5 AU at the trailing edge of the elution peak. The elution fraction(s) of interest can then be prepared for further processing. For example, the collected sample can be titrated to a pH of about 5.0 using Tris (e.g., 1.0 M) at a pH of about 10. Optionally, this titrated sample can be filtered and further processed.

In certain embodiments, the HIC purification step can also be preceded by an ion exchange chromatography step. The ion exchange purification step can occur before, after, or in place of an affinity chromatography step. In certain embodiments, where a Protein A step precedes the ion exchange step, a Protein A eluate is purified using a cation exchange column. In certain embodiments, the equilibrating buffer used in the cation exchange column is a buffer having a pH of about 5.0. An example of a suitable buffer is about 210 mM sodium acetate, pH 5.0. Following equilibration, the column is loaded with sample prepared from HIC purification step above. The column is packed with a cation exchange media, such as CM Sepharose™ Fast Flow from GE Healthcare. The column is then washed using the equilibrating buffer. The column is next subjected to an elution step using a buffer having a greater ionic strength as compared to the equilibrating or wash buffer. For example, a suitable elution buffer can be about 790 mM sodium acetate, pH 5.0. The antibodies will be eluted and can be monitored using a UV spectrophotometer set at OD280nm. In a particular example, elution collection can be from upside 3 OD280nm to downside 8 OD280nm. It should be understood that one skilled in the art may vary the conditions and yet still be within the scope of the invention.

In certain embodiments where a Protein A step precedes an ion exchange step, a Protein A eluate is purified using an anion exchange column. A non-limiting example of a suitable column for this step is a 60 cm diameter×30 cm long column whose bed volume is about 85 L. The column is packed with an anion exchange media, such as Q Sepharose™ Fast Flow from GE Healthcare. The column can be equilibrated using about seven column volumes of an appropriate buffer such as Tris/sodium chloride. An example of suitable conditions is 25 mM Tris, 50 mM sodium chloride at pH 8.0. A skilled artisan may vary the conditions but still be within the scope of the present invention. The column is loaded with the collected sample from the HIC purification step outlined above. In another aspect, the column is loaded from the eluate collected during cation exchange. Following the loading of the column, the column is washed with the equilibration buffer (e.g., the Tris/sodium chloride buffer). The flow-through comprising the antibodies can be monitored using a UV spectrophotometer at OD280nm. This anion exchange step reduces process related impurities such as nucleic acids like DNA, and host cell proteins. The separation occurs due to the fact that the antibodies of interest do not substantially interact with nor bind to the solid phase of the column, e.g., to the Q Sepharose™, but many impurities do interact with and bind to the column's solid phase. The anion exchange can be performed at about 12° C.

In certain embodiments, the cation exchange or anion exchange eluate, depending on which ion exchange step is employed, or employed first, is next filtered using, e.g., a 16 inch Cuno™ delipid filter. This filtration, using the delipid filter, can be followed by, e.g., a 30-inch 0.45/0.2 μm Sartopore™ bi-layer filter cartridge. The ion exchange elution buffer can be used to flush the residual volume remaining in the filters and prepared for ultrafiltration/diafiltration.

In order to accomplish the ultratfiltration/diafiltration step, the filtration media is prepared in a suitable buffer, e.g., 20 mM sodium phosphate, pH 7.0. A salt such as sodium chloride can be added to increase the ionic strength, e.g., 100 mM sodium chloride. This ultrafiltration/diafiltration step serves to concentrate the anti-IL-12, anti-TNFα, or anti-IL-18 antibodies, remove the sodium acetate and adjust the pH. Commercial filters are available to effectuate this step. For example, Millipore manufactures a 30 kD molecular weight cut-off (MWCO) cellulose ultrafilter membrane cassette. This filtration procedure can be conducted at or around room temperature.

In certain embodiments, the sample from the capture filtration step above is subjected to a second ion exchange separation step. In certain embodiments, this second ion exchange separation will involve separation based on the opposite charge of the first ion exchange separation. For example, if an anion exchange step is employed after HIC purification, the second ion exchange chromatographic step may be a cation exchange step. Conversely, if the HIC purification step was followed by a cation exchange step, that step would be followed by an anion exchange step. In certain embodiments the first ion exchange eluate can be subjected directly to the second ion exchange chromatographic step where the first ion exchange eluate is adjusted to the appropriate buffer conditions. Suitable anionic and cationic separation materials and conditions are described above.

In certain embodiments, a mixed mode chromatography step will precede the HIC chromatography step, thereby forming a mixed mode chromatography sample that can be exposed to the HIC media in the HIC chromatography step. Examples of mixed mode medias include, but are not limited to: CaptoAdhere (GE Healthcare), PPA-HyperCel (Pall Life Sciences), and HEA-HyperCel (Pall Life Sciences). In certain embodiments, the mixed mode chromatography step is a CaptoAdhere chromatography step. In certain embodiments, the mixed mode chromatography sample is further subject to a filtration step. Filters well known to those skilled in the art can be used in this embodiment. In one aspect, the filtration step is a nanofiltration step. In certain embodiments, a depth filtration step follows a filtration step.

In certain embodiments of the invention, the eluate from the hydrophobic chromatography step is subjected to filtration for the removal of viral particles, including intact viruses, if present. A non-limiting example of a suitable filter is the Ultipor DV50™ filter from Pall Corporation. Other viral filters can be used in this filtration step and are well known to those skilled in the art. The HIC eluate is passed through a pre-wetted filter of about 0.1 μm and a 2×30-inch Ultipor DV50™ filter train at around 34 psig. In certain embodiments, following the filtration process, the filter is washed using, e.g., the HIC elution buffer in order to remove any antibodies retained in the filter housing. The filtrate can be stored in a pre-sterilized container at around 12° C.

In a certain embodiments, the filtrate from the above is again subjected to ultrafiltration/diafiltration. This step is important if a practitioner's end point is to use the antibody in a, e.g., pharmaceutical formulation. This process, if employed, can facilitate the concentration of antibody, removal of buffering salts previously used and replace it with a particular formulation buffer. In certain embodiments, continuous diafiltration with multiple volumes, e.g., two volumes, of a formulation buffer is performed. A non-limiting example of a suitable formulation buffer is 5 mM methionine, 2% mannitol, 0.5% sucrose, pH 5.9 buffer (no Tween). Upon completion of this diavolume exchange the antibodies are concentrated. Once a predetermined concentration of antibody has been achieved, then a practitioner can calculate the amount of 10% Tween that should be added to arrive at a final Tween concentration of about 0.005% (v/v).

Certain embodiments of the present invention will include further purification steps. Examples of additional purification procedures which can be performed prior to, during, or following the ion exchange chromatography method include ethanol precipitation, isoelectric focusing, reverse phase HPLC, chromatography on silica, chromatography on heparin Sepharose™, further anion exchange chromatography and/or further cation exchange chromatography, chromatofocusing, SDS-PAGE, ammonium sulfate precipitation, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography (e.g., using protein G, an antibody, a specific substrate, ligand or antigen as the capture reagent).

In certain embodiments the unbound flow through and wash fractions can be further fractionated and a combination of fractions providing a target product purity can be pooled.

In certain embodiments the protein concentration can be adjusted to achieve a differential partitioning behavior between the antibody product and the product-related substances such that the purity and/or yield can be further improved.

In certain embodiments the loading can be performed at different protein concentrations during the loading operation to improve the product quality/yield of any particular purification step.

In certain embodiments the column temperature, can be independently varied to improve the separation efficiency and/or yield of any particular purification step.

In certain embodiments, the loading and washing buffer matrices can be different or composed of mixtures of chemicals, while achieving similar “hydrophobic interaction” behavior such that the above novel separation can be effected.

In certain embodiments, the loading and washing buffers can be different, in terms of ionic strength or pH, while remaining substantially similar in function in terms of the washout of the product achieved during the wash step.

In certain embodiments, the loading & washing steps can be controlled by in-line, at-line or off-line measurement of the product related impurity/substance levels, either in the column effluent, or the collected pool or both, so as to achieve the target product quality and/or yield.

In certain embodiments, the loading concentration can be dynamically controlled by in-line or batch or continuous dilutions with buffers or other solutions to achieve the partitioning necessary to improve the separation efficiency and/or yield.

In certain embodiments, additives such as amino acids, sugars, PEG, etc can be added to the load or wash steps to modulate the partitioning behavior to achieve the separation efficiency and/or yield.

In certain embodiments, the separation can be performed on any type of hydrophobic interaction media such as membranes, monoliths or depth filters that have hydrophobic interaction characteristics.

Mixed mode media can also be employed to enable this method, provided the same functionality is achieved by appropriately adjusting the column loading and/or washing conditions.

5.5. Methods of Assaying Sample Purity

5.5.1 Assaying Host Cell Protein

The present invention also provides methods for determining the residual levels of host cell protein (HCP) concentration in the isolated/purified antibody composition. As described above, HCPs are desirably excluded from the final target substance product. Exemplary HCPs include proteins originating from the source of the antibody production. Failure to identify and sufficiently remove HCPs from the target antibody may lead to reduced efficacy and/or adverse subject reactions.

As used herein, the term “HCP ELISA” refers to an ELISA where the second antibody used in the assay is specific to the HCPs produced from cells, e.g., CHO cells, used to generate the antibody of interest. The second antibody may be produced according to conventional methods known to those of skill in the art. For example, the second antibody may be produced using HCPs obtained by sham production and purification runs, i.e., the same cell line used to produce the antibody of interest is used, but the cell line is not transfected with antibody DNA. In an exemplary embodiment, the second antibody is produced using HPCs similar to those expressed in the cell expression system of choice, i.e., the cell expression system used to produce the target antibody.

Generally, HCP ELISA comprises sandwiching a liquid sample comprising HCPs between two layers of antibodies, i.e., a first antibody and a second antibody. The sample is incubated during which time the HCPs in the sample are captured by the first antibody, for example, but not limited to goat anti-CHO, affinity purified (Cygnus). A labeled second antibody, or blend of antibodies, specific to the HCPs produced from the cells used to generate the antibody, e.g., anti-CHO HCP Biotinylated, is added, and binds to the HCPs within the sample. In certain embodiments the first and second antibodies are polyclonal antibodies. In certain aspects the first and second antibodies are blends of polyclonal antibodies raised against HCPs. The amount of HCP contained in the sample is determined using the appropriate test based on the label of the second antibody.

HCP ELISA may be used for determining the level of HCPs in an antibody composition, such as an eluate or flow-through obtained using the process described above. The present invention also provides a composition comprising an antibody, wherein the composition has no detectable level of HCPs as determined by an HCP Enzyme Linked Immunosorbent Assay (“ELISA”).

5.5.2 Assaying Charge and Size Variants

In certain embodiments, the levels of product-related substances, such as acidic species and other charge variants, in the chromatographic samples produced using the techniques described herein are analyzed. For example, but not by way of limitation, the acidic species and other charge variants present in the Adalimumab process samples can be quantified according to the following methods. Cation exchange chromatography was performed on a Dionex ProPac WCX-10, Analytical column 4 mm×250 mm (Dionex, CA). An Agilent 1200 HPLC system was used as the HPLC. The mobile phases used were 10 mM Sodium Phosphate dibasic pH 7.5 (Mobile phase A) and 10 mM Sodium Phosphate dibasic, 500 mM Sodium Chloride pH 5.5 (Mobile phase B). A binary gradient (94% A, 6% B: 0-20 min; 84% A, 16% B: 20-22 min; 0% A, 100% B: 22-28 min; 94% A, 6% B: 28-34 min) was used with detection at 280 nm.

In certain embodiments, the levels of aggregates, monomer, and fragments in the chromatographic samples produced using the techniques described herein are analyzed. In certain embodiments, the aggregates, monomer, and fragments are measured using a size exclusion chromatographic (SEC) method for each molecule. For example, but not by way of limitation, a TSK-gel G3000SW×L, 5 μm, 125 Å, 7.8×300 mm column (Tosoh Bioscience) can be used in connection with certain embodiments, while a TSK-gel Super SW3000, 4 μm, 250 Å, 4.6×300 mm column (Tosoh Bioscience) can be used in alternative embodiments. In certain embodiments, the aforementioned columns are used along with an Agilent or a Shimazhu HPLC system. In certain embodiments, sample injections are made under isocratic elution conditions using a mobile phase consisting of, for example, 100 mM sodium sulfate and 100 mM sodium phosphate at pH 6.8, and detected with UV absorbance at 214 nm. In certain embodiments, the mobile phase will consist of 1×PBS at pH 7.4, and elution profile detected with UV absorbance at 280 nm. In certain embodiments, quantification is based on the relative area of detected peaks.

5.6. Further Modifications

The purified proteins, e.g., antibodies, of the present invention can be modified. In some embodiments, the proteins are chemically modified to provide a desired effect. For example, pegylation of antibodies or antibody fragments of the invention may be carried out by any of the pegylation reactions known in the art, as described, e.g., in the following references: Focus on Growth Factors 3:4-10 (1992); EP 0 154 316; and EP 0 401 384, each of which is incorporated by reference herein in its entirety. In one aspect, the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule (or an analogous reactive water-soluble polymer). A suitable water-soluble polymer for pegylation of the antibodies and antibody fragments of the invention is polyethylene glycol (PEG). As used herein, “polyethylene glycol” is meant to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (Cl—ClO) alkoxy- or aryloxy-polyethylene glycol.

Methods for preparing pegylated antibodies and antibody fragments of the invention will generally comprise the steps of (a) reacting the antibody or antibody fragment with polyethylene glycol, such as a reactive ester or aldehyde derivative of PEG, under suitable conditions whereby the antibody or antibody fragment becomes attached to one or more PEG groups, and (b) obtaining the reaction products. It will be apparent to one of ordinary skill in the art to select the optimal reaction conditions or the acylation reactions based on known parameters and the desired result.

Generally the pegylated antibodies and antibody fragments have increased half-life, as compared to the nonpegylated antibodies and antibody fragments. The pegylated antibodies and antibody fragments may be employed alone, together, or in combination with other pharmaceutical compositions.

An antibody of the invention can be derivatized or linked to another functional molecule (e.g., another peptide or protein). For example, an antibody of the invention can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate associate of the antibody with another molecule (such as a streptavidin core region or a polyhistidine tag).

One type of derivatized antibody is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such linkers are available from Pierce Chemical Company, Rockford, Ill.

Useful detectable agents with which an antibody of the invention may be derivatized include fluorescent compounds. Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme, it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.

5.7. Pharmaceutical Compositions

The proteins of interest, e.g., antibodies and antibody-binding portions thereof, of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the pharmaceutical composition comprises an antibody of the invention and a pharmaceutically acceptable carrier. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it is desirable to include isotonic agents, e.g., sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody.

The antibodies and antibody-binding portions thereof, of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration. The antibody or antibody-portions can be prepared as an injectable solution containing, e.g., 0.1-250 mg/mL antibody. The injectable solution can be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule or pre-filled syringe. The buffer can be L-histidine approximately 1-50 mM, (optimally 5-10 mM), at pH 5.0 to 7.0 (optimally pH 6.0). Other suitable buffers include but are not limited to sodium succinate, sodium citrate, sodium phosphate or potassium phosphate. Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form). Cryoprotectants can be included for a lyophilized dosage form, principally 0-10% sucrose (optimally 0.5-1.0%). Other suitable cryoprotectants include trehalose and lactose. Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 24%). Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L-methionine (optimally 5-10 mM). Other suitable bulking agents include glycine, arginine, can be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%). Additional surfactants include but are not limited to polysorbate 20 and BRIJ surfactants.

In one aspect, the pharmaceutical composition includes the antibody at a dosage of about 0.01 mg/kg-10 mg/kg. In another aspect, the dosages of the antibody include approximately 1 mg/kg administered every other week, or approximately 0.3 mg/kg administered weekly. A skilled practitioner can ascertain the proper dosage and regime for administering to a subject.

The compositions of this invention may be in a variety of forms. These include, e.g., liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The form depends on, e.g., the intended mode of administration and therapeutic application. Typical compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. One mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In one aspect, the antibody is administered by intravenous infusion or injection. In another aspect, the antibody is administered by intramuscular or subcutaneous injection.

Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile, lyophilized powders for the preparation of sterile injectable solutions, the methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, e.g., by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, e.g., monostearate salts and gelatin.

The antibodies and antibody-binding portions thereof, of the present invention can be administered by a variety of methods known in the art, one route/mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978, the entire teaching of which is incorporated herein by reference.

In certain aspects, an antibody or antibody-binding portion thereof, of the invention may be orally administered, e.g., with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.

Supplementary active compounds can also be incorporated into the compositions. In certain aspects, an antibody or antibody-binding portion thereof, of the invention is co-formulated with and/or co-administered with one or more additional therapeutic agents that are useful for treating disorders. Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies. It will be appreciated by the skilled practitioner that when the antibodies of the invention are used as part of a combination therapy, a lower dosage of antibody may be desirable than when the antibody alone is administered to a subject (e.g., a synergistic therapeutic effect may be achieved through the use of combination therapy which, in turn, permits use of a lower dose of the antibody to achieve the desired therapeutic effect).

It should be understood that the antibodies of the invention can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the present invention. The additional agent also can be an agent which imparts a beneficial attribute to the therapeutic composition, e.g., an agent which affects the viscosity of the composition.

6. EXAMPLES 6.1. Materials & Methods

6.1.1. Chromatography Method

Except where noted, the materials and methods described in connection with the instant example were also employed in the examples of Sections 6.2., 6.3., and 6.4., below.

Pre-packed media columns were used in the following experiments, except where specified. The column was equilibrated in a buffer system with appropriate pH and conductivity. The column load was prepared from Protein A affinity chromatography eluates or concentrated CEX chromatography elutes by buffer exchange (if the eluates were with different buffer components from the mixed mode target buffer system) or addition of the stock solutions and/or water to obtain the target pH and conductivity as specified (if the eluates were with the same buffer components as the mixed mode target buffer system). The prepared load material was filtered and loaded on the column according to the target load amount (g protein/L media) as specified followed by washing with the equilibration buffer or buffer similar to equilibration buffer with volumes as specified. The column Flow Through/Wash were collected as fractions or as a pool. HIC column was cleaned with 20% Isopropyl Alcohol solution. 1M NaOH solution was used for column cleaning.

6.1.2. Buffer Preparation Method

Buffers were prepared targeting a specific salt concentration in a buffered system, and titrating to a specific pH with the conjugate acid or base. For example, an 800 mM Ammonium Sulfate (AmSO4) pH 7.0 solution was made by dissolving AmSO4 salt in a 20 mM Tris-Acetate buffered solution, titrating with acetate, and subsequently bringing up to volume with water to achieve the desired AmSO4 concentration. Load samples were prepared targeting a specific salt concentration by addition of concentrated salt solution in a buffered system, and titrating to a specific pH with the conjugate acid or base. For example, an 800 mM AmSO4 pH 7.0 load was made by mixing the load in a 1:1 ratio with a 1600 mM AmSO4 pH 7.0 stock buffer in a 40 mM Tris-Acetate, and subsequently titrating with Tris or acetate to achieve a final pH 7.0.

6.1.3. Size Exclusion Chromatography

The molecular weight distribution of collected samples were quantified according to the following methods. Size exclusion chromatography (SEC) was performed using a TSK-gel G3000SW×L, 5 μm, 125 Å, 7.8×300 mm column (Tosoh Bioscience) on an HP Agilent HPLC system. Injections were made under isocratic elution conditions using a mobile phase of 200 mM sodium sulfate, 100 mM sodium phosphate, pH 6.8, and detected with absorbance at 214 nm. Quantification is based on the relative area of detected peaks.

6.1.4. Host Cell Protein (HCP) ELISA

HCP assay is based on process specific antigen based ELISA. Sample dilutions were applied to achieve readings within the calibration range. The limit of quantitation of the assay is 0.625 ng/mL.

6.1.5. UV Spectroscopy A280

UV A280 was used to determine protein concentrations for the samples post protein A elution. The assay was performed on an Agilent UV Spectrophotometer following the method. The protein concentration was determined using Beer-Lambert's Law, A=εlc, where A is Absorbance, ε is the extinction coefficient, l is the path length, and c is the concentration. The absorbance was taken at 280 nm, the path length was 1 cm, and the extinction coefficients were 1.39 for Adalimumab, 1.38 for mAb B, and 1.43 for mAb C.

6.2. Example HIC 1 Determining Operating Conditions Appropriate For an mAb:Media:Buffer Combination

The demonstration of the current invention for a specific antibody & media is provided in this example, and consists of

    • 1. Choosing a salt concentration that allows product and impurities to bind at a given pH.
    • 2. Loading a small amount of protein to the column and then performing a linear gradient elution by decreasing the salt concentration.
    • 3. Determining salt concentration range in which the protein elutes from the hydrophobic interaction media.

In this example, adalimumab and GE CaptoPhenyl were chosen. The column was equilibrated at 1.1 M AmSO4 pH 7.0 (Tris/Acetate) for 10 CVs. Adalimumab was prepared at 1.1 M AmSO4 and loaded to the column at 20 g-protein/L of resin. The column was washed with 10 CVs of the equilibration buffer. A linear gradient from 1.1M to 0M AmSO4 pH 7.0 (Tris/Acetate) over 20CVs was performed. The process chromatogram is shown in FIG. 1.

This process can be repeated for any given mAb-media combination for a given buffer system. Table 1 shows the DOE parameters determined using the method described above for adlimumab in AmSO4 pH 7.0 (Tris/acetate) for 3 different HIC adsorbents.

TABLE 1 Example Experimental Design Scope determined from LGE with different resins Adlimumab - Ammonium Sulfate pH 7.0 (Tris/Acetate) Resin Buffer Concentration Range Tosoh Hexyl 250-750 mM GE CaptoPhenyl 300-650 mM GE Butyl FF 800-950 mM

In practicing the current invention, the aggregate reduction desired can be achieved by appropriate pooling of the load and wash fractions. By collecting and subsequently determining the product quality of each fraction throughout the load and wash, the accumulative aggregate reduction and accumulative yield can be calculated using the weighted averages up to a given fraction. Additionally, the instantaneous yield can be estimated by comparing the protein recovered against the total protein loaded to the column at a given fraction. Sample calculations are shown below:

Sample Calculation A: Accumulative Yield Up to a Given Fraction

Accumulative Yield = Accumulated Protein Mass Recovered up to Fraction Total Mass Protein Load

Sample Calculation B: Accumulative Aggregate Reduction Up to a Given Fraction

Accumulative Aggregate Reduction = Load Agg % - Accumulated Aggregate Mass Recovered up to Fraction Accumulated Total Protein Mass Recovered up to Fraction

Sample Calculation C: Instantaneous Yield Up to a Given Fraction

Instantenous Yield = Accumulated Protein Mass Recovered up to Fraction Total Protein Mass Loaded to Column at Fraction

The demonstration of the current invention for a specific antibody & resin is provided in this example, and consists of

    • 1. For a given salt concentration and optionally pH and hydrophobic interaction media.
    • 2. Loading the hydrophobic interaction media in excess of the dynamic binding capacity for the product for the given condition.
    • 3. Washing the column with a buffer containing a similar salt concentration and optionally pH used for the equilibration and loading steps.
    • 4. Collecting fractions throughout the loading and wash steps and subsequently determining the product quality profile (e.g. Aggregate, HCP etc.)

In this example, adalimumab and GE CaptoPhenyl were chosen. The experiment was performed at 400 mM sodium citrate (NaCit) pH 5.6. The column was equilibrated with 400 mM NaCit pH 5.6 for 10 CVs. Adalimumab was prepared at 400 mM NaCit pH 5.6 and loaded to the column at 500 g-protein/L-resin. The column was washed with 7 CVs of the equilibration buffer. The process chromatogram is shown in FIG. 2. Fractions were collected and analyzed for product quality and the accumulative yield and accumulative aggregate reduction calculated, shown in Table 2. From this example, it is clear to one skilled in the art to determine a run condition which delivers a targeted product quality and/or step yield.

This general approach is used to evaluate the performance for a given operating condition for any resin/mAb/buffer combination.

TABLE 2 Accumulative Yield and AR Reduction from FIG. 2 Fraction Load Accumulative Recovery Accumulative ΔAgg D1  8 g/L 0% 0.82% D2  45 g/L 4% 0.77% D3  82 g/L 12% 0.71% D4 119 g/L 19% 0.67% D5 156 g/L 26% 0.62% D6 193 g/L 33% 0.56% D7 231 g/L 41% 0.51% E1 268 g/L 48% 0.47% E2 305 g/L 55% 0.43% E3 342 g/L 62% 0.40% E4 379 g/L 70% 0.37% E5 416 g/L 77% 0.34% E6 454 g/L 84% 0.32% E7 491 g/L 91% 0.29% F1 500 g/L 93% 0.29% F2 WASH 99% 0.28% F3 WASH 100% 0.28% F4 WASH 101% 0.29% F5 WASH 101% 0.29%

6.3. Example HIC 2 Demonstration of aggregate reduction with HIC resins

This data set is compiled to demonstrate the aggregate reduction achieved with six different HIC adsorbents. Each resin was evaluated with a 500 g/L load of adalimumab at a NaCit concentration near, and slightly higher than, the peak elution concentration determined from the process outlined in Example 6.2. Table 3 outlines the results from these experiments.

TABLE 3 Effect of HIC Resins on aggregate reduction of Adalimumab HIC Resin NaCit, pH 5.6 ΔAgg Yield Butyl 400 mM 1.5% 99.8% 450 mM 1.2% 85.7% Hexyl 240 mM 1.2% 93.9% 300 mM 1.1% 100.9% Phenyl 400 mM 1.5% 96.5% 450 mM 1.2% 90.7% Octyl 350 mM 0.4% 98.5% 400 mM 0.1% 103.3% GE Butyl FF 550 mM 1.2% 88.1% 600 mM 1.7% 83.0% PPG 450 mM 0.2% 97.5% 600 mM 1.0% 38.1%

6.4. Example HIC 3 Demonstration of Aggregate Reduction with Other Antibodies, mAb B and mAb C

Aggregate reduction technology of the current invention has been demonstrated with multiple antibodies using HIC adsorbents. Antibodies have different hydrophobic properties, leading to interaction behavior on a HIC column that differs from one antibody to another. Therefore the impact of salt type and concentration is different for each antibody.

Table 4 and Table 5, presented below, provide the data obtained for mAb B and mAB C. The data clearly demonstrates that the aggregate reduction technology is effective for alternatives to adalimumab.

TABLE 4 AR reduction for mAb B, pI ~9.1 HIC Resin AmSO4, pH 5.0 ΔAgg Yield Hexyl 370 mM 0.8% 100% 710 mM 0.6% 93% Phenyl 340 mM 0.6% 95% 790 mM 0.5% 95% Butyl 840 mM 0.6% 99% 1000 mM  0.6% 96%

TABLE 5 AR reduction for mAb C, pI ~7.0 HIC Resin AmSO4, pH 5.0 ΔAgg Yield Hexyl  80 mM 5.0% 89.0% 330 mM 4.5% 99.8% Phenyl 130 mM 3.5% 92.8% 480 mM 2.9% 92.8% Butyl 690 mM 5.2% 93.5% 880 mM 5.4% 87.9%

6.5. Example HIC 4 Demonstration of Aggregate Reduction with Different Salt Concentrations—Adalimumab

Ion concentration is a key variable in the performance of hydrophobic interaction chromatography. For every combination of antibody/resin/pH there is a range of ion concentrations that provide aggregate reduction; the strategy outlined in Example 6.2. can be followed to determine the aggregate reduction and the corresponding recovery for each salt concentration.

Table 6, below, shows the effect of salt concentration on aggregate reduction and step yield. In this example CaptoPhenyl and adalimumab were chosen, and evaluated at a loading of 200-500 g/L in NaCit pH 5.6 at the concentration specified. The data demonstrates that the aggregate reduction can be effectively achieved over a range of salt concentrations, and that the salt concentration and column loading can be balanced to achieve a desired step yield and final product quality

TABLE 6 Effect of Ion Concentration on Aggregate Reduction NaCit pH 5.6 Load Yield ΔAgg 300 mM 200 g/L 92% 0.59% 350 g/L 96% 0.33% 500 g/L 97% 0.24% 400 mM 200 g/L 90% 0.76% 350 g/L 94% 0.43% 500 g/L 96% 0.35% 500 mM 200 g/L 85% 1.09% 350 g/L 91% 0.97% 500 g/L 94% 0.86%

6.6. Example HIC 5 Demonstration of Aggregate Reduction with Different Buffer Systems with Adalimumab

In addition to the salt concentration, the salt anion and cation types are key variables in hydrophobic interaction chromatography. The invention has been demonstrated with ammonium sulfate, sodium sulfate, and sodium citrate. As one skilled in the art would appreciate the optimal salt concentration and optionally pH are different for each salt type and was derived by using the strategy outlined in Example 6.2. Table 7 shows the data of aggregate reduction and corresponding recovery for the different anion/cation types and different HIC adsorbents.

TABLE 7 Effect of Anion/Cation Type Aggregate Reduction Resin Buffer System Load Yield ΔAgg CaptoPhenyl 630 mM AmSO4 pH 7.0 300 g/L 95% 2.1% 300 mM AmSO4 pH 7.0 300 g/L 99% 1.1% 425 mM NaSO4 pH 7.0 300 g/L 95% 1.9% 240 mM NaSO4 pH 7.0 300 g/L 101% 1.1% 500 mM NaCit pH 5.6 350 g/L 91% 1.0% 300 mM NaCit pH 5.6 350 g/L 96% 0.2% Tosoh Hexyl 725 mM AmSO4 pH 7.0 300 g/L 94% 1.7% 275 mM AmSO4 pH 7.0 300 g/L 103% 0.9% 460 mM NaSO4 pH 7.0 300 g/L 97% 0.7% 180 mM NaSO4 pH 7.0 300 g/L 101% 0.6% 440 mM NaCit pH 5.6 300 g/L 87% 0.5% 150 mM NaCit pH 5.6 300 g/L 97% 0.5% Butyl FF 800 mM AmSO4 pH 7.0 300 g/L 100% 0.7% 1000 mM AmSO4 pH 7.0 300 g/L 94% 1.6% 750 mM NaSO4 pH 7.0 300 g/L 96% 1.8% 700 mM NaSO4 pH 7.0 300 g/L 101% 1.7% 700 mM NaCit pH 5.6 300 g/L 98% 1.6% 600 mM NaCit pH 5.6 300 g/L 95% 1.5%

6.7. Example HIC 6 Demonstration of Aggregate Reduction with Different Loading

Furthermore, the strategy outlined in Example 6.2. to reduce aggregates through careful control of ion concentration, ion type, HIC adsorbent, and pH can be applied to various ranges of protein loading. Aggregate reduction for a range of protein loadings (e.g. 250-700 g/L) for CaptoPhenyl using a 400 mM NaCit pH 5.6 buffer is shown in Table 8, displaying a robust aggregate reduction across an expansive loading range.

TABLE 8 Impact of Column loading Load Yield ΔAgg ΔAgg/LoadAgg 250 g/L 95% 0.29% 87% 500 g/L 100% 0.25% 77% 700 g/L 100% 0.21% 65%

6.8. Example HIC 7 Demonstration of Aggregate Reduction with Different Load Concentration—Adalimumab

In addition to the strategy outlined in Example 6.7. to reduce aggregates through careful control of ion concentration, ion type, and HIC adsorbent, it has been shown that the concentration of the load protein can have an effect on aggregate reduction. In this example, a feed stream was serial diluted to cover a range of load concentrations from 4 to 15 mg/mL and loaded at 500 g/L to a CaptoPhenyl column in 400 mM NaCit pH 5.6. The effect of decreasing the concentration of the load protein is shown in FIG. 3.

6.9. Example HIC 8 Demonstration of HCP Reduction in Addition to Aggregate Reduction

HIC chromatography can also be effective in reducing host cell protein (HCP) levels. In the present invention, it has been demonstrated that HCP levels can be effectively reduced under operating conditions selected for aggregate reduction.

Table 9 shows HCP removal achieved along with aggregate reduction. The data clearly shows that other process related substances/impurities can be achieved using the current invention on the HIC adsorbents, and hence functions as an effective polishing step in the large scale purification of monoclonal antibodies.

TABLE 9 HCP Removal during HIC Chromatography HCP NaCit pH 5.6 Load Yield ΔAgg Load Pool 300 mM 200 g/L 92% 0.59% 1398 ng/mg NA 350 g/L 96% 0.33% 150 ng/mg 500 g/L 97% 0.24% 348 ng/mg 200 g/L 99% 0.34%  38 ng/mg  5 ng/mg 400 mM 200 g/L 90% 0.76% 1599 ng/mg 104 ng/mg 350 g/L 94% 0.43% 148 ng/mg 500 g/L 96% 0.35% 350 ng/mg 350 g/L 97% 0.35%  38 ng/mg  6 ng/mg 500 mM 200 g/L 85% 1.09% 1528 ng/mg 169 ng/mg 350 g/L 91% 0.97% 203 ng/mg 500 g/L 94% 0.86% 301 ng/mg 500 g/L 87% 0.35%  38 ng/mg  11 ng/mg

6.10. Example HIC 9 Demonstration of Impact of Dynamic and Equilibrium Binding

In the HIC-based separation strategies described herein, the measured dynamic binding capacity (DBC), which is conventionally measured at 10% breakthrough, was found to be greater than the amount of protein that remained bound after washing the column (a.k.a equilibrium binding capacity, EBC) with a buffer with similar pH and salt concentration to the equilibration and load conditions. For example, but not by way of limitation, FIG. 4 shows an example of the DBC and EBC for the data presented in FIG. 2. In addition, Table 10 shows effect of salt type, concentration, and HIC resin on DBC and EBC values for Adalimumab.

TABLE 10 Comparison of DBC and EBC values for Adilmumab Resin Buffer System ΔAgg DBC EBC CaptoPhenyl 630 mM AmSO4 pH 7.0 2.1% 27 g/L 16 g/L 300 mM AmSO4 pH 7.0 1.1%  6 g/L  4 g/L 425 mM NaSO4 pH 7.0 1.9% 22 g/L 15 g/L 240 mM NaSO4 pH 7.0 1.1%  6 g/L  4 g/L Butyl FF 1000 mM AmSO4 pH 7.0 1.6% 17 g/L 11 g/L 800 mM AmSO4 pH 7.0 0.7%  4 g/L  4 g/L 750 mM NaSO4 pH 7.0 1.8% 29 g/L 13 g/L 700 mM NaSO4 pH 7.0 1.7% 22 g/L 11 g/L 700 mM NaCit pH 5.6 1.6% 39 g/L 24 g/L 600 mM NaCit pH 5.6 1.5% 17 g/L 11 g/L

6.11. Example HIC 10 Combinations of HIC with Alternative Separation Strategies

The methods described herein for reducing aggregates using HIC can be used as an independent operation or in combination with other process steps that provide additional aggregate reduction or those providing additional complementary and supplementary purification. Data for specific separation strategies is provided in Tables 11 and 12. For example, but not by way of limitation, the following process combinations can be used:

    • 1. Affinity→HIC
    • 2. Affinity→AEX→HIC
    • 3. Affinity→Mixed Mode→HIC

TABLE 11 Aggregate reduction with different source materials HCP Load Source Buffer Condition Load Yield ΔAgg LRF Protein A 400 mM NaCit pH 5.6 500 g/L 96% 1.49% NA Eluate 450 mM NaCit pH 5.6 500 g/L 91% 1.22% NA Protein A/ 300 mM NaCit pH 5.6 200 g/L 92% 0.59% 1.0 AEX FTW 400 mM NaCit pH 5.6 350 g/L 94% 0.43% 1.0 500 mM NaCit pH 5.6 500 g/L 94% 0.86% 0.7 Protein A/ 300 mM NaCit pH 5.6 200 g/L 99% 0.34% 0.8 Mixed Mode 400 mM NaCit pH 5.6 350 g/L 97% 0.35% 0.8 FTW 500 mM NaCit pH 5.6 500 g/L 87% 0.35% 0.5

TABLE 12 Complete Process Train with Protein A Capture - AR, HMW and HCP reduction % HMW Process Yield (%) reduction HCP LRF Clarified Harvest 97.00% n/a n/a Prt-A Eluate Pool 89.60% n/a 1.87 Viral Inactivated 99.70% 0.07 0.39 Filtrate MM FT pool 91.90% 0.83 1.63 HIC FT-pool 98.50% 0.23 0.46 VF(FT) Filtrate 96.10% No reduction 0.1 BDS (FT) 103.80% No reduction 0.13

6.12. Example HIC 11 Hybrid HIC Binding Mechanism

By estimating the partitioning coefficient Kp, it can be demonstrated that certain strategies described in the instant application do not fall under the category of “Weak-Partitioning (WP)” or “Flow-Through Overload (FT)” modes as those are described in the art, e.g., US2007/0060741. For example, FIG. 5A-B depict the results of experiments wherein aliquots of resin are incubated with a load covering a range of protein concentrations at room temperature for 3 hours, after which the protein solution is then removed, and replaced with equilibration buffer (Wash simulation) and incubated at room temperature for 3 hours (repeated, Wash II). After each incubation, the concentration of the protein solution is measured and used to calculated the amount of protein ((A) monomer D2E7, a.ka. Adalimumab, and (B) aggregate D2E7) bound to the resin (g protein/L resin) and plotted against the concentration of the protein solution at the end of the incubation (e.g. equilibrium).

FIG. 6A-B depict the results outlined in FIGS. 5A-B, highlighting the fact that at initial equilibrium a significant amount of monomer/aggregate is bound to the resin. However, after the protein solution is replaced with equilibration buffer (see arrow), the monomer de-sorbs from the resin and back into solution, where as the aggregate remains bound.

FIG. 7A-B depict a determination of the binding monomer and aggregate D2E7 (based on data provided in FIGS. 5A-B) by fitting the experimental equilibrium binding data to the Langmuir Isotherm using the equation: q=(qmax×Cequil)/(Kd+Cequil); where q=amount of protein bound to resin [=] g/L-resin; qmax=maximum amount of protein bound to resin [=] g/L-resin; Cequil=solution concentration of protein [=] g/L-soln; and Kd=equilibrium dissociation constant.

By fitting the experimental data, the qmax and Kd for the monomer and the aggregates can be calculated.

Species Qmax [mg/mL] Kd [mg/mL] Monomer 41.9 0.47 Aggregate 6.0 0.01

Significantly, qmax for both monomer/aggregate and the Kd values (i.e. strength of binding) are similar to those of strong hydrophobic interactions, therefore it is not expected for this interaction to be “reversible.” In addition, by calculating Kp where:

Species Qmax[mg/mL] Kd[mg/mL] K p Q C Q max K d Monomer 41.9 0.47  90 Aggregate  6.0 0.01 600

it is apparent that the instant technique does not fall within the category of flow-through (where Kp≦1) or weak portioning (where Kp=1-10), but rather fall within the category of bind and elute (where Kp≧10).

Patents, patent applications, publications, product descriptions, GenBank Accession Numbers, and protocols that may be cited throughout this application, the disclosures of which are incorporated herein by reference in their entireties for all purposes. For example, but not by way of limitation, U.S. patent application Ser. No. 13/803,808, filed on Mar. 14, 2013; U.S. patent application Ser. No. 13/830,583, filed on Mar. 14, 2013; U.S. patent application Ser. No. 13/829,989, filed on Mar. 14, 2013; U.S. patent application Ser. No. 13/830,976, filed on Mar. 14, 2013; and U.S. patent application Ser. No. 13/804,220, filed on Mar. 14, 2013 are incorporated herein by reference in their entireties for all purposes.

Claims

1. A method for producing a preparation comprising a process-related impurity-reduced and/or product-related substance-reduced composition comprising a protein of interest from a sample mixture comprising a protein of interest and at least one process-related impurity and/or product-related substance, said method comprising: wherein the flow through and/or wash fractions constitute the process-related impurity-reduced and/or product-related substance-reduced composition comprising a protein of interest preparation

(a) subjecting said sample mixture to a hydrophobic interaction media and collecting the unbound fraction;
(b) contacting the said hydrophobic interaction media after loading with a solution that is substantially similar in hydrophobic interaction functionality to the load sample and collecting the wash;

2. The method of claim 1, wherein prior to subjecting said sample mixture to a hydrophobic interaction chromatography media the sample mixture is subjected to an affinity chromatographic media.

3. The method of claim 2, wherein the affinity chromatographic media is a Protein A, G, A/G, or L media.

4. The method of claim 2, wherein the affinity chromatographic media is MabSuRe Protein A media.

5. The method of claim 1, wherein prior to subjecting said sample mixture to a hydrophobic interaction chromatography media the sample mixture is subjected to an ion exchange chromatography media.

6. The method of any one of claim 5, wherein the ion exchange media is selected from a cation exchange media and an anion exchange media.

7. The method of claim 6, wherein the ion exchange media is an anion exchange media selected from media comprising diethylaminoethyl (DEAE), quaternary aminoethyl (QAE) and quaternary amine (Q) group ligands.

8. The method of claim 6, wherein the ion exchange media is a cation exchange media selected from media comprising carboxymethyl (CM), sulfoethyl(SE), sulfopropyl(SP), phosphate(P) and sulfonate(S) ligands.

9. The method of claim 1, wherein prior to subjecting said sample mixture to a hydrophobic interaction chromatography media the sample mixture is subjected to a mixed mode chromatography media.

10. The method of claim 9, wherein the mixed mode media is CaptoAdhere resin.

11. The method of claim 1 wherein said antibody is a human antibody, humanized antibody, a chimeric antibody, or a multivalent antibody, or an antigen-binding portion thereof.

12. The method of claim 1, wherein said HIC media comprises at least one hydrophobic ligand.

13. The method of claim 12, wherein said at least one hydrophobic ligand is selected from the group consisting of alkyl-, aryl-ligands, and combinations thereof.

14. The method of claim 12, wherein at least one hydrophobic group is selected from the group consisting of butyl, hexyl, phenyl, octyl, or polypropylene glycol ligands.

15. The method of claim 12, wherein said HIC comprises a column.

16. The method of claim 16, wherein said column is selected from the group consisting of: Capto Phenyl; Phenyl Sepharose™ 6 Fast Flow with low or high substitution; Phenyl Sepharose™ High Performance; Octyl Sepharose™ High Performance; Fractogel™ EMD Propyl; Fractogel™ EMD Phenyl; Macro-Prep™ Methyl; Macro-Prep™ t-Butyl columns; WP HI-Propyl (C3)™; Toyopearl™ ether, phenyl or butyl; ToyoScreen PPG; ToyoScreen Phenyl; ToyoScreen Butyl; ToyoScreen Hexyl; GE HiScreen Butyl FF; and HiScreen Octyl FF.

17. The method of claim 1, wherein said hydrophobic interaction chromatography sample is subjected to a filtration step.

18. The method of claim 17, further comprising a depth filtration step, nanofiltration step, ultrafiltration step or absolute filtration step or combination thereof.

19. A pharmaceutical composition comprising a process-related impurity-reduced and/or product-related substance-reduced composition comprising a protein of interest and a pharmaceutically acceptable carrier

20. The pharmaceutical composition of claim 19, wherein said composition is substantially free of product-related aggregates, product-related fragments, and HCPs.

Patent History
Publication number: 20160115193
Type: Application
Filed: Dec 22, 2015
Publication Date: Apr 28, 2016
Inventors: Matthew Omon Herigstad (Charlestown, MA), Linda E. Rich (Worcester, MA), Stephen Ming-teh Lu (Worcester, MA), Natarajan Ramasubramanyan (Westborough, MA)
Application Number: 14/978,553
Classifications
International Classification: C07K 1/22 (20060101); C07K 16/24 (20060101); C07K 16/00 (20060101);