US20190194298A1 - System and method for characterizing drug product impurities - Google Patents

System and method for characterizing drug product impurities Download PDF

Info

Publication number
US20190194298A1
US20190194298A1 US16/223,463 US201816223463A US2019194298A1 US 20190194298 A1 US20190194298 A1 US 20190194298A1 US 201816223463 A US201816223463 A US 201816223463A US 2019194298 A1 US2019194298 A1 US 2019194298A1
Authority
US
United States
Prior art keywords
cells
antibody
drug product
protein drug
impurities
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/223,463
Other languages
English (en)
Inventor
Shunhai Wang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority to US16/223,463 priority Critical patent/US20190194298A1/en
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WANG, Shunhai
Publication of US20190194298A1 publication Critical patent/US20190194298A1/en
Priority to US17/677,499 priority patent/US20220169704A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/30Partition chromatography
    • B01D15/305Hydrophilic interaction chromatography [HILIC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/15Medicinal preparations ; Physical properties thereof, e.g. dissolubility
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • G01N33/6857Antibody fragments
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • C12Q1/6872Methods for sequencing involving mass spectrometry
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y305/00Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5)
    • C12Y305/01Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5) in linear amides (3.5.1)
    • C12Y305/01052Peptide-N4-(N-acetyl-beta-glucosaminyl)asparagine amidase (3.5.1.52), i.e. glycopeptidase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N2030/022Column chromatography characterised by the kind of separation mechanism
    • G01N2030/027Liquid chromatography
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/88Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86
    • G01N2030/8809Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample
    • G01N2030/8813Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample biological materials
    • G01N2030/8831Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample biological materials involving peptides or proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6848Methods of protein analysis involving mass spectrometry

Definitions

  • the invention is generally directed to protein separation methods and cell culture methods.
  • LMW Low molecular weight
  • HMW high molecular weight
  • antibody dimer species are both examples of product-related impurities that contribute to the size heterogeneity of mAb products.
  • the formation of HMW species within a therapeutic mAb drug product as a result of protein aggregation can potentially compromise both drug efficacy and safety (e.g. eliciting unwanted immunogenic response) (Rosenberg A S. The AAPS journal, 8:E501-7 (2006); Moussa E M, et al. Journal of pharmaceutical sciences, 105:417-30(2006)).
  • LMW species of any therapeutic protein may result from host cell protease activity during production.
  • LMW species often have low or substantially reduced activity relative to the monomeric form of the antibody, while exposing novel epitopes that can lead to immunogenicity or potentially impact pharmacokinetic properties in vivo (Vlasak J, Ionescu R. mAbs, 3:253-63 (2011)).
  • HMW and LMW species are considered critical quality attributes that are routinely monitored during drug development and as part of release testing of purified drug substance during manufacturing.
  • Proteolytic fragments may also be observed.
  • the proposed identity of each minor band can be supported by N-terminal sequencing via Edman degradation, in-gel tryptic digestion followed by mass spectrometry analysis, and western blot analysis using anti-Fc and anti-light chain antibodies.
  • any proposed structures resulting from these methods cannot be unambiguously confirmed at the intact protein level.
  • sample preparation conditions employed in SDS-PAGE experiments can generate LMW artifacts through disulfide bond scrambling, which can lead to overestimations of minor LMW species (Zhu Z C, et al. Journal of pharmaceutical and biomedical analysis, 83:89-95(2013)).
  • CE-SDS capillary electrophoresis-sodium dodecyl sulfate
  • RPLC reversed-phase chromatography
  • mass spectrometry can be used to detect free light chain and associated post-translational modifications (e.g. cysteinylation and glutathionylation) present in mAb drug products.
  • RPLC often lacks sufficient resolution to separate LMW species and thus fails to elucidate the complete LMW profile.
  • H2L species in mAb drug products has never been reported by RPLC-based intact mass analysis, owing to its low abundance and poor resolution from the main intact antibody.
  • Another MS-based technique that is promising for characterizing mAb product-related impurities is native electrospray ionization mass spectrometry (Native ESI-MS), which is particularly informative when coupled with size exclusion chromatography (SEC) (Haberger M, et al. mAbs, 8:331-9 (2016)).
  • SEC size exclusion chromatography
  • LMW species identified in native SEC-MS analysis are often not the same as those identified by SDS-PAGE or CE-SDS, due to significantly different experimental conditions used between methods.
  • the sample preparation required for SDS-PAGE and CE-SDS often starts with protein denaturation, where the non-covalent interactions between the N-terminal regions of HC-LC pairs and the C-terminal regions of the HC-HC pairs are disrupted.
  • LMW impurities such as H2L, half antibody, and free light chain species are able to dissociate from the mAb molecule if the interchain disulfide bonds are broken.
  • native SEC-MS analyzes the mAb samples under near native conditions, permitting the strong non-covalent interchain interactions to be preserved and allowing the four-chain structure of the mAb molecule to be maintained even if the interchain disulfide bonds are broken.
  • LMW protein drug product impurities uses hydrophilic interaction chromatography (HILIC) coupled to mass spectrometry analysis. After removal of the N-linked glycans from the protein drug product, for example an antibody drug product, the elution of LMW impurities from the HILIC column is determined by the size of the molecular weight species. In some embodiments, the HILIC separation is performed under denaturing conditions, making the detection of LMW forms using this method highly comparable to both SDS-PAGE and CE-SDS methods.
  • LMW drug product impurities include, but are not limited to light chain, half antibody, H2L, H2, HL, HC, peptide backbone-truncated species, and combinations thereof.
  • the disclosed HILIC-MS systems and methods provide detailed LMW identification information.
  • the reliable identification and detailed structural information revealed by HILIC-MS analysis is highly valuable for in-depth characterization of LMW impurities in protein drug products, which is often required for late-stage molecule development.
  • the disclosed HILIC-MS system and methods use gentler sample preparations than either SDS-PAGE or CE-SDS does, it is less likely to generate LMW artifacts.
  • the HILIC-MS systems and methods can be used as a semi-quantitative analysis to compare the LMW impurity profile between samples or simply applied qualitatively.
  • One embodiment provides a protein drug product containing a protein drug and an excipient, wherein the protein drug product comprises between about 0.05 and about 30.0% w/w of low molecular weight protein drug impurities.
  • the protein drug product can be an antibody, a fusion protein, recombinant protein, or a combination thereof
  • the drug product contains between about 0.05% to about 25%, or about 0.05% to about 15%, or about 0.05% to about 10%, or about 0.05% to about 5%, or about 1 to about 25%, about 1 to about 15%, about 1 to about 10%, or about 1 to about 5% w/w of low molecular weight protein drug impurities.
  • Still another embodiment provides a method of producing an antibody, including the steps of culturing cells producing the antibody in a cell culture, obtaining a sample from the cell culture, characterizing and quantifying low molecular weight impurities in the sample according to the method described above and modifying one or more culture conditions of the cell culture to reduce the amount of characterized low molecular protein drug impurities produced during cell culture of the antibody.
  • the sample is taken during the cell culture at any interval. In other embodiments, the sample is taken following production culture, following protein harvest or following purification.
  • the one or more conditions of the cell culture that are changed to reduce the amount of low molecular weight protein drug impurities can be selected from the group consisting of temperature, pH, cell density, amino acid concentration, osmolality, growth factor concentration, agitation, gas partial pressure, surfactants, or combinations thereof.
  • the cells can be eukaryotic or prokaryotic.
  • the cells can be Chinese Hamster Ovary (CHO) cells (e.g. CHO K1, DXB-11 CHO, Veggie-CHO), COS cells (e.g. COS-7), retinal cells, Vero cells, CV1 cells, kidney cells (e.g.
  • the cells are hybridoma or quadroma cells. Still another embodiment provides an antibody produced by the methods described herein.
  • the system includes a hydrophilic interaction liquid chromatography system including a hydrophilic interaction liquid chromatography (HILIC) column linked to mobile phase A and mobile phase B as exemplified herein, and the HILIC column is in fluid communication with a mass spectroscopy system.
  • HILIC hydrophilic interaction liquid chromatography
  • the invention relates to use of the method according to the invention for determination of stability and forced degradation studies of a protein drug product.
  • the invention relates to use of a system according to the invention for determination of stability and forced degradation studies of a protein drug product.
  • FIGS. 1A and 1B are chromatograms of HILIC separation of mAb-1 drug substance sample.
  • FIG. 1A is the ultraviolet profile and
  • FIG. 1B is the HILIC profile.
  • the UV signal was amplified 10 times in FIG. 1A to better visualize the LMW impurities.
  • FIG. 2A shows the extracted ion chromatograms (XICs) of different light chain variants using the m/z of the most abundant charge state.
  • FIG. 2B shows the deconvoluted mass spectrum of light chain with ⁇ -elimination.
  • FIG. 2C shows the deconvoluted mass spectrum of light chain modified by 2-mercaptoacetic acid, unmodified light chain and light chain with C-terminal residues clipped.
  • FIG. 2D shows the deconvoluted mass spectrum of light chain with cysteinylation.
  • FIG. 2E shows the deconvoluted mass spectrum of light chain with glutathionylation.
  • FIG. 3A shows the deconvoluted mass spectrum of Fab fragments.
  • FIG. 3B shows the deconvoluted mass spectrum of half antibody.
  • FIG. 3C shows the deconvoluted mass spectrum of Fab-truncated fragments.
  • FIG. 3D shows the deconvoluted mass spectrum of H2.
  • FIG. 4 shows The HILIC-UV analysis of deglycosylated mAb-1 sample treated by DTT (arrow #1) and L-cysteine (arrow #2). Arrow #3 points to the trace with untreated mAb-1.
  • the signal of untreated sample and L-cysteine treated sample were amplified 10 times and 2 times, respectively, to better visualize the LMW impurities.
  • FIG. 5A shows light chain species identified in HILIC-MS analysis of mAb-1 after treated with DTT.
  • FIG. 5B shows heavy chain species identified in HILIC-MS analysis of mAb-1 after treated with DTT.
  • FIG. 5C shows half antibody species identified in HILIC-MS analysis of mAb-1 after treated with DTT.
  • FIG. 5D shows heavy chain dimer species identified in HILIC-MS analysis of mAb-1 after treated with DTT.
  • FIG. 5E shows H2L species identified in HILIC-MS analysis of mAb-1 after treated with DTT.
  • FIG. 5F shows full antibody species identified in HILIC-MS analysis of mAb-1 after treated with DTT.
  • FIG. 6A shows an MS2 spectrum of light chain C-terminal peptides from Lys-C digestion under non-reduced conditions wherein the C-terminal Cys is modified by iodoacetamide.
  • FIG. 6B shows an MS2 spectrum of light chain C-terminal peptides from Lys-C digestion under non-reduced conditions wherein the C-terminal Cys is modified by unknown modification of +89.98 Da.
  • FIG. 7A shows the extracted ion chromatogram (XIC) of light chain C-terminal peptide with unknown modification of +89.98 Da non-reduced Lys-C digests.
  • FIG. 7B shows the extracted ion chromatogram (XIC) of light chain C-terminal peptide with unknown modification of +89.98 Da from reduction of the non-reduced Lys-C digests.
  • FIG. 8 is the proposed structure (2-mercaptoacetic acid) of the unknown modification.
  • FIG. 9 is an exemplary HILIC-MS system.
  • low molecular weight (LMW) protein drug impurity includes but is not limited to precursors, degradation products, truncated species, proteolytic fragments including Fab fragments, Fc or heavy chain fragments, ligand or receptor fragments, H2L (2 heavy chains and 1 light chain), H2 (2 heavy chains), HL (1 heavy chain and 1 light chain), HC (1 heavy chain), and LC (1 light chain) species.
  • a LMW protein drug impurity can be any variant which is an incomplete version of the protein product, such as one or more components of a multimeric protein.
  • Protein drug impurity, drug impurity or product impurity are terms that may be used interchangeably throughout the specification and include LMW protein drug impurities.
  • LMW drug or product impurities are generally considered molecular variants with properties such as activity, efficacy, and safety that may be different from those of the desired drug product.
  • Degradation of protein product is problematic during production of the protein drug product in cell culture systems.
  • proteolysis of a protein product may occur due to release of proteases in cell culture medium.
  • Medium additives such as soluble iron sources added to inhibit metalloproteases, or serine and cysteine proteases inhibitors, have been implemented in cell culture to prevent degradation (Clincke, M.-F., et al, BMC Proc. 2011, 5, P 115).
  • C-terminal fragments may be cleaved during production due to carboxyl peptidases in the cell culture (Dick, L W et al, Biotechnol Bioeng 2008; 100:1132-43). Subsequently, there is a need to profile and monitor LMW impurities.
  • Protein refers to a molecule comprising two or more amino acid residues joined to each other by a peptide bond. Protein includes polypeptides and peptides and may also include modifications such as glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, alkylation, hydroxylation and ADP-ribosylation. Proteins can be of scientific or commercial interest, including protein-based drugs, and proteins include, among other things, enzymes, ligands, receptors, antibodies and chimeric or fusion proteins.
  • Proteins are produced by various types of recombinant cells using well-known cell culture methods, and are generally introduced into the cell by genetic engineering techniques (e.g., such as a sequence encoding a chimeric protein, or a codon-optimized sequence, an intronless sequence, etc.) where it may reside as an episome or be intergrated into the genome of the cell.
  • genetic engineering techniques e.g., such as a sequence encoding a chimeric protein, or a codon-optimized sequence, an intronless sequence, etc.
  • Antibody refers to an immunoglobulin molecule consisting of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain has a heavy chain variable region (HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region contains three domains, CH1, CH2 and CH3.
  • Each light chain has a light chain variable region and a light chain constant region.
  • the light chain constant region consists of one domain (CL).
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • 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” includes reference to both glycosylated and non-glycosylated immunoglobulins of any isotype or subclass.
  • the term “antibody” includes antibody molecules prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell transfected to express the antibody.
  • the term antibody also includes bispecific antibody, which includes a heterotetrameric immunoglobulin that can bind to more than one different epitope. Bispecific antibodies are generally described in US Patent Application Publication No. 2010/0331527, which is incorporated by reference into this application.
  • Fc fusion proteins comprise part or all of two or more proteins, one of which is an Fc portion of an immunoglobulin molecule, which are not otherwise found together in nature. Preparation of fusion proteins comprising certain heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al., Proc. Natl. Acad. Sci. USA 88: 10535, 1991; Byrn et al., Nature 344:677, 1990; and Hollenbaugh et al., “Construction of Immunoglobulin Fusion Proteins”, in Current Protocols in Immunology, Suppl.
  • Receptor Fc fusion proteins comprise one or more extracellular domain(s) of a receptor coupled to an Fc moiety, which in some embodiments comprises a hinge region followed by a CH2 and CH3 domain of an immunoglobulin.
  • the Fc-fusion protein comprises two or more distinct receptor chains that bind to a one or more ligand(s).
  • an Fc-fusion protein is a trap, such as for example an IL-1 trap or VEGF trap.
  • Cell culture refers to the propagation or proliferation of cells in a vessel, such as a flask or bioreactor, and includes but is not limited to fed-batch culture, continuous culture, perfusion culture and the like.
  • HILIC or HILIC chromatography refers to hydrophilic interaction liquid chromatography or hydrophilic interaction chromatography, and is considered a well-known term of the art.
  • any protein of interest suitable for expression in prokaryotic or eukaryotic cells can be used in the engineered host cell systems provided.
  • the protein of interest includes, but is not limited to, an antibody or antigen-binding fragment thereof, a chimeric antibody or antigen-binding fragment thereof, an ScFv or fragment thereof, an Fc-fusion protein or fragment thereof, a growth factor or a fragment thereof, a cytokine or a fragment thereof, or an extracellular domain of a cell surface receptor or a fragment thereof.
  • Proteins of interest may be simple polypeptides consisting of a single subunit, or complex multisubunit proteins comprising two or more subunits.
  • the protein of interest may be a biopharmaceutical product, food additive or preservative, or any protein product subject to purification and quality standards.
  • the protein product is an antibody, a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a multispecific antibody, a bispecific antibody, an antigen binding antibody fragment, a single chain antibody, a diabody, triabody or tetrabody, a Fab fragment or a F(ab′)2 fragment, an IgD antibody, an IgE antibody, an IgM antibody, an IgG antibody, an IgG1 antibody, an IgG2 antibody, an IgG3 antibody, or an IgG4 antibody.
  • the antibody is an IgG1 antibody.
  • the antibody is an IgG2 antibody.
  • the antibody is an IgG4 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG4 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG1 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG1/IgG4 antibody.
  • the antibody is selected from the group consisting of an anti-Programmed Cell Death 1 antibody (e.g. an anti-PD1 antibody as described in U.S. Pat. Appln. Pub. No. US2015/0203579A1), an anti-Programmed Cell Death Ligand-1 (e.g. an anti-PD-L1 antibody as described in in U.S. Pat. Appln. Pub. No. US2015/0203580A1), an anti-Dll4 antibody, an anti-Angiopoetin-2 antibody (e.g. an anti-ANG2 antibody as described in U.S. Pat. No. 9,402,898), an anti-Angiopoetin-Like 3 antibody (e.g.
  • an anti-Programmed Cell Death 1 antibody e.g. an anti-PD1 antibody as described in U.S. Pat. Appln. Pub. No. US2015/0203580A1
  • an anti-Dll4 antibody e.g. an anti-Angiopoetin
  • an anti-AngPtl3 antibody as described in U.S. Pat. No. 9,018,356 an anti-platelet derived growth factor receptor antibody (e.g. an anti-PDGFR antibody as described in U.S. Pat. No. 9,265,827), an anti-Erb3 antibody, an anti-Prolactin Receptor antibody (e.g. anti-PRLR antibody as described in U.S. Pat. No. 9,302,015), an anti-Complement 5 antibody (e.g. an anti-C5 antibody as described in U.S. Pat. Appln. Pub. No US2015/0313194A1), an anti-TNF antibody, an anti-epidermal growth factor receptor antibody (e.g. an anti-EGFR antibody as described in U.S.
  • an anti-GCGR antibody as described in U.S. Pat. Appln. Pub. Nos. US2015/0337045A1 or US2016/0075778A1
  • an anti-VEGF antibody an anti-IL1R antibody
  • an interleukin 4 receptor antibody e.g. an anti-IL4R antibody as described in U.S. Pat. Appln. Pub. No. US2014/0271681A1 or U.S. Pat Nos. 8,735,095 or 8,945,559
  • an anti-interleukin 6 receptor antibody e.g. an anti-IL6R antibody as described in U.S. Pat. Nos.
  • an anti-IL1 antibody an anti-IL2 antibody, an anti-IL3 antibody, an anti-IL4 antibody, an anti-IL5 antibody, an anti-IL6 antibody, an anti-IL7 antibody, an anti-interleukin 33 (e.g. anti-IL33 antibody as described in U.S. Pat. Appln. Pub. Nos. US2014/0271658A1 or US2014/0271642A1), an anti-Respiratory syncytial virus antibody (e.g. anti-RSV antibody as described in U.S. Pat. Appln. Pub. No. US2014/0271653A1), an anti-Cluster of differentiation 3 (e.g.
  • an anti-CD3 antibody as described in U.S. Pat. Appln. Pub. Nos. US2014/0088295A1 and US20150266966A1, and in U.S. Application No. 62/222,605
  • an anti-Cluster of differentiation 20 e.g. an anti-CD20 antibody as described in U.S. Pat. Appln. Pub. Nos. US2014/0088295A1 and US20150266966A1, and in U.S. Pat. No. 7,879,984
  • an anti-CD19 antibody an anti-CD28 antibody
  • an anti-Cluster of Differentiation-48 e.g. anti-CD48 antibody as described in U.S. Pat. No.
  • an anti-Fel d1 antibody e.g. as described in U.S. Pat. No. 9,079,948
  • an anti-Middle East Respiratory Syndrome virus e.g. an anti-MERS antibody as described in U.S. Pat. Appln. Pub. No. US2015/0337029A1
  • an anti-Ebola virus antibody e.g. as described in U.S. Pat. Appln. Pub. No. US2016/0215040
  • an anti-Zika virus antibody e.g. an anti-Lymphocyte Activation Gene 3 antibody (e.g. an anti-LAG3 antibody, or an anti-CD223 antibody)
  • an anti-Nerve Growth Factor antibody e.g.
  • the bispecific antibody is selected from the group consisting of an anti-CD3 ⁇ anti-CD20 bispecific antibody (as described in U.S. Pat. Appln. Pub. Nos.
  • an anti-CD3 ⁇ anti-Mucin 16 bispecific antibody e.g., an anti-CD3 ⁇ anti-Muc16 bispecific antibody
  • an anti-CD3 ⁇ anti-Prostate-specific membrane antigen bispecific antibody e.g., an anti-CD3 ⁇ anti-PSMA bispecific antibody
  • the protein of interest is selected from the group consisting of abciximab, adalimumab, adalimumab-atto, ado-trastuzumab, alemtuzumab, alirocumab, atezolizumab, avelumab, basiliximab, belimumab, benralizumab, bevacizumab, bezlotoxumab, blinatumomab, brentuximab vedotin, brodalumab, canakinumab, capromab pendetide, certolizumab pegol, cemiplimab, cetuximab, denosumab, dinutuximab, dupilumab, durvalumab, eculizumab, elotuzumab, emicizumab-kxwh, emtansinealirocumab
  • the protein of interest is a recombinant protein that contains an Fc moiety and another domain, (e.g., an Fc-fusion protein).
  • an Fc-fusion protein is a receptor Fc-fusion protein, which contains one or more extracellular domain(s) of a receptor coupled to an Fc moiety.
  • the Fc moiety comprises a hinge region followed by a CH2 and CH3 domain of an IgG.
  • the receptor Fc-fusion protein contains two or more distinct receptor chains that bind to either a single ligand or multiple ligands.
  • an Fc-fusion protein is a TRAP protein, such as for example an IL-1 trap (e.g., rilonacept, which contains the IL-1RAcP ligand binding region fused to the Il-1R1 extracellular region fused to Fc of hIgG1; see U.S. Pat. No. 6,927,004, which is herein incorporated by reference in its entirety), or a VEGF trap (e.g., aflibercept or ziv-aflibercept, which comprises the Ig domain 2 of the VEGF receptor Flt1 fused to the Ig domain 3 of the VEGF receptor Flk1 fused to Fc of hIgG1; see U.S. Pat. Nos.
  • IL-1 trap e.g., rilonacept, which contains the IL-1RAcP ligand binding region fused to the Il-1R1 extracellular region fused to Fc of hIgG1; see U.S. Pat. No. 6,927,
  • an Fc-fusion protein is a ScFv-Fc-fusion protein, which contains one or more of one or more antigen-binding domain(s), such as a variable heavy chain fragment and a variable light chain fragment, of an antibody coupled to an Fc moiety.
  • a “fed-batch cell culture” or “fed-batch culture” refers to a batch culture wherein the cells and culture medium are supplied to the culturing vessel initially, and additional culture nutrients are slowly fed, in discrete increments, to the culture during culturing, with or without periodic cell and/or product harvest before termination of culture.
  • Fed-batch culture includes “semi-continuous fed-batch culture” wherein periodically whole culture (which may include cells and medium) is removed and replaced by fresh medium.
  • Fed-batch culture is distinguished from simple “batch culture” whereas all components for cell culturing (including the animal cells and all culture nutrients) are supplied to the culturing vessel at the start of the culturing process in batch culture.
  • Fed-batch culture may be different from “perfusion culture” insofar as the supernatant is not removed from the culturing vessel during a standard fed-batch process, whereas in perfusion culturing, the cells are restrained in the culture by, e.g., filtration, and the culture medium is continuously or intermittently introduced and removed from the culturing vessel. However, removal of samples for testing purposes during fed-batch cell culture is contemplated. The fed-batch process continues until it is determined that maximum working volume and/or protein production is reached, and protein is subsequently harvested.
  • continuous cell culture relates to a technique used to grow cells continually, usually in a particular growth phase. For example, if a constant supply of cells is required, or the production of a particular protein of interest is required, the cell culture may require maintenance in a particular phase of growth. Thus, the conditions must be continually monitored and adjusted accordingly in order to maintain the cells in that particular phase.
  • cell culture medium and “culture medium” refer to a nutrient solution used for growing mammalian cells that typically provides the necessary nutrients to enhance growth of the cells, such as a carbohydrate energy source, essential (e.g. phenylalanine, valine, threonine, tryptophan, methionine, leucine, isoleucine, lysine, and histidine) and nonessential (e.g. alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine) amino acids, trace elements, energy sources, lipids, vitamins, etc.
  • Cell culture medium may contain extracts, e.g.
  • Chemically defined medium refers to a cell culture medium in which all of the chemical components are known (i.e. have a known chemical structure). Chemically defined medium is entirely free of animal-derived components, such as serum- or animal-derived peptones. In one embodiment, the medium is a chemically defined medium.
  • the solution may also contain components that enhance growth and/or survival above the minimal rate, including hormones and growth factors.
  • the solution may be formulated to a pH and salt concentration optimal for survival and proliferation of the particular cell being cultured.
  • a “cell line” refers to a cell or cells that are derived from a particular lineage through serial passaging or subculturing of cells.
  • the term “cells” is used interchangeably with “cell population”.
  • cell includes any cell that is suitable for expressing a recombinant nucleic acid sequence.
  • Cells include those of prokaryotes and eukaryotes, such as bacterial cells, mammalian cells, human cells, non-human animal cells, avian cells, insect cells, yeast cells, or cell fusions such as, for example, hybridomas or quadromas.
  • the cell is a human, monkey, ape, hamster, rat or mouse cell.
  • the cell is selected from the following cells: Chinese Hamster Ovary (CHO) (e.g. CHO K1, DXB-11 CHO, Veggie-CHO), COS (e.g.
  • the cell comprises one or more viral genes, e.g. a retinal cell that expresses a viral gene (e.g. a PER.C6® cell).
  • the cell is a CHO cell. In other embodiments, the cell is a CHO K1 cell.
  • Multisubunit therapeutic proteins particularly monoclonal antibody (mAb)-based therapeutics are inherently heterogeneous with respect to size due to their complex multi-chain structure and the propensity to accommodate multiple enzymatic and chemical post-translational modifications.
  • mAb monoclonal antibody
  • the levels of size variants within a mAb drug product can be readily quantitated by a variety of biophysical methods, unambiguous identification of those product-related impurities has been particularly challenging.
  • Hydrophilic interaction chromatography coupled with mass spectrometry analysis was used to identify low levels of lower molecular weight (LMW) impurities present within a purified drug substance lot.
  • LMW lower molecular weight
  • the HILIC method separates mAb-related LMW impurities with a size-based elution order.
  • the subsequent mass measurement from a high-resolution accurate mass spectrometer provides direct and unambiguous identification of a variety of low-abundance LMW impurities in a single LC-MS analysis.
  • Free light chain, half antibody, H2L species (antibody possessing a single light chain) and peptide backbone-truncated species can all be confidently identified and elucidated in great detail, including the truncation sites and associated post-translational modifications.
  • Such detailed information that cannot be acquired by traditional purity assays e.g. SDS-PAGE and CE-SDS
  • SDS-PAGE and CE-SDS is of great importance to elucidate how the LMW impurities are formed, making this new method a valuable addition to analytical characterization portfolio.
  • the system includes a hydrophilic interaction liquid chromatography system including a hydrophilic interaction liquid chromatography (HILIC) column linked to mobile phase A and mobile phase B, and the HILIC column is in fluid communication with a mass spectroscopy system.
  • the HILIC column is suitable for use with deglycosylated proteins.
  • the disclosed system contains a Waters ACQUITYTM UPLC Glycoprotein Amide column (300 ⁇ , 1.7 ⁇ m, 2.1 ⁇ 150 mm) with a Waters ACQUITYTM UPLC system. The column is typically operated at 60° C.
  • Exemplary mobile phases include 0.1% TFA in water as mobile phase A, 0.1% TFA in acetonitrile as mobile phase B, and the flow rate was set to 0.2 mL/min.
  • the UV traces are typically recorded at 215 and 280 nm.
  • the separation is achieved over 55 min with an initial 0.5 min hold at 15% A, followed by an increase to 25% A over the next 0.5 min, and another linear increase to 40% A over the next 40 minutes.
  • the gradient can be ramped to 100% A over 1 min and held for 2 minutes, before dropping to 15% A in 1 min, and then maintained at initial condition for over 10 minutes to equilibrate the column for the next run.
  • the UPLC is directly coupled to a mass spectrometer, for example a Thermo Scientific Q Exactive hybrid quadrupole Orbitrap mass spectrometer.
  • the capillary voltage is typically set at 4.0 kV, with a sheath gas flow rate of 40 arbitrary units and auxiliary gas flow rate of 15 arbitrary units.
  • the capillary temperature is generally set at 350° C. and the probe heater temperature is generally set at 400° C.
  • Mass spectra are acquired in the mass range of m/z 800-4000. In one embodiment, the raw data is deconvoluted using Intact MassTM software developed by Protein Metrics.
  • the disclosed systems and methods can be used to characterize LMW protein drug impurities.
  • One embodiment provides a method for characterizing low molecular weight protein drug product impurities including the steps of deglycosylating a protein drug product sample, separating protein components of the protein drug product sample by hydrophilic interaction chromatography, and analyzing the separated protein components by mass spectroscopy to characterize low molecular weight protein drug product impurities in the protein drug product sample.
  • the protein drug product sample is taken from or purified from a fed-batch cell culture, a continuous cell culture or a perfusion cell culture.
  • Exemplary protein drug product includes but are not limited to an antibody, a fusion protein, recombinant protein, or a combination thereof.
  • Exemplary low molecular weight protein drug product impurities include but are not limited to precursors, degradation products, truncated species, proteolytic fragments including Fab, ligand or receptor fragments or heavy chain fragments, free light chain, half antibody, H2L, H2, HL, HC, or a combination thereof.
  • Another embodiment includes a step of reducing the sample.
  • exemplary reducing agents include but are not limited to dithiothreitol (DTT, CAS 3483-12-3), beta-mercaptoethanol (BME, 2BME, 2-ME, b-mer, CAS 60-24-2), 2-aminoethanethiol (2-MEA-HCl, also called cysteamine-HCl, CAS 156-57-0), Tris (2-carboxyethyl) phosphine hydrochloride, (TCEP, CAS 5961-85-3), cysteine hydrochloride (Cys-HCl, CAS 52-89-1), or 2-mercaptoethanesulfonic acid sodium salt (MESNA).
  • DTT dithiothreitol
  • BME beta-mercaptoethanol
  • 2-aminoethanethiol also called cysteamine-HCl, CAS 156-57-0
  • One embodiment provides a method of producing an antibody including the steps of culturing cells producing the antibody, for example in a fed-batch culture, obtaining a sample from the cell culture, characterizing and quantifying low molecular weight impurities in the sample using the systems and methods disclosed herein and modifying one or more culture conditions of the cell culture to reduce the amount of characterized low molecular protein drug impurities produced during cell culture of the antibody.
  • the conditions are changed to have the protein drug impurities in a range of 0.05% and 30.0%, preferably 0.05% to 15%, 0.05% to 10%, 0.05% to 5%, or 0.05% to 2% (w/w).
  • the one or more conditions of the cell culture that are changed to reduce the amount of low molecular weight protein drug impurities are selected from the group consisting of temperature, pH, cell density, amino acid concentration, osmolality, growth factor concentration, agitation, gas partial pressure, surfactants, or combinations thereof.
  • the cells producing the antibody are Chinese hamster ovary cells. In other embodiments, the cells are hybridoma cells.
  • Another embodiment provides an antibody produced according the methods provided herein have 1 to 5%, 5 to 10%, 10 to 15%, 15 to 20% protein drug impurities.
  • mAb-1 a recombinant IgG1 mAb (mAb-1) made by Regeneron was used.
  • Peptide-N-Glycosidase F PNGase F, #P0704L
  • Tris-hydrochloride pH 7.5 solution #15567-027) was purchased from Invitrogen
  • dithiothreitol DTT, #20291
  • L-cysteine #168149-25G was purchased from Sigma-Aldrich.
  • Acetonitrile (LC-MS grade, #A955-4) and trifluoroacetic acid (TFA, #PI28904) were purchased from Fisher Scientific. Milli-Q water was provided in-house.
  • the mAb-1 sample was diluted to a final concentration of 5 ⁇ g/ ⁇ L using 100 mM Tri-HCl (pH 7.5). PNGase F was added at an enzyme to substrate ratio of 1 unit/10 ⁇ g protein. The deglycosylation reaction was conducted at 37° C. for 3 hours. To initiate the limited reduction by DTT, a 20 ⁇ g aliquot of the deglycosylated mAb-1 sample was reduced with 5 mM DTT before immediate injection onto the HILIC column for online UV and mass spectrometry analysis.
  • a Waters ACQUITY UPLC Glycoprotein Amide column (300 ⁇ , 1.7 ⁇ m, 2.1 ⁇ 150 mm) was used on a Waters ACQUITY UPLC system for all HILIC separations.
  • the column was operated at 60° C.
  • the mobile phases were 0.1% TFA in water as mobile phase A, 0.1% TFA in acetonitrile as mobile phase B, and the flow rate was set to 0.2 mL/min.
  • the UV traces were recorded at 215 and 280 nm. The separation was achieved over 55 min with an initial 0.5 min hold at 15% A, followed by an increase to 25% A over the next 0.5 min, and another linear increase to 40% A over the next 40 minutes.
  • the gradient was then ramped to 100% A over 1 min and held for 2 minutes, before dropping to 15% A in 1 min, and then maintained at initial condition for over 10 minutes to equilibrate the column for the next run.
  • the UPLC was directly coupled to a Thermo Scientific Q Exactive hybrid quadrupole Orbitrap mass spectrometer.
  • the capillary voltage was set at 4.0 kV, with a sheath gas flow rate of 40 arbitrary units and auxiliary gas flow rate of 15 arbitrary units.
  • the capillary temperature was set at 350° C. and the probe heater temperature was set at 400° C.
  • Mass spectra were acquired in the mass range of m/z 800-4000. The raw data were deconvoluted using Intact MassTM software developed by Protein Metrics.
  • a recombinant IgG1 mAb (mAb-1) drug substance sample was used as a model molecule.
  • PNGase F photodiode array
  • FIGS. 1A and 1B mass spectrometry analysis
  • TIC total ion chromatogram
  • PNGase F major glycan form
  • FIGS. 2A-2E Multiple light chain-related impurities were detected in HILIC-MS analysis of the mAb-1 sample ( FIGS. 2A-2E ).
  • the extracted ion chromatograms (XICs) of each species suggested that they also exhibited different retention times during HILIC separation ( FIG. 2A ).
  • the cysteinylated light chain (+ ⁇ 119 Da) was identified as the major form of all light chain species present in mAb-1 sample and it is the only species visible by UV ( FIG. 2D ).
  • the cysteinylation may occur as a result of the thiol-disulfide exchange reaction between the inter-heavy and light chain disulfide bond and a free cysteine molecule, which can be found in cytoplasm.
  • glutathionylated light chain (+ ⁇ 306 Da) was also identified, with a retention time slightly later than the cysteinylated light chain ( FIG. 2E ). Similar to the cysteinylation process, free glutathione (GSH) molecule, which can also be found in cytoplasm, should be responsible for this modification.
  • GSH glutathione
  • Another interesting LC variant with a delta mass of + ⁇ 90 Da from predicted mass was also observed ( FIG. 2C ). Subsequent peptide mapping analysis using recombinant Lys-C protease located this modification to light chain C-terminal Cys residue.
  • Half antibody is formed as a result of inter-heavy and heavy chain disulfide bonds at the hinge region scrambling into intra-heavy chain disulfide bonds.
  • the four-chain structure of a mAb molecule remains undisrupted by this scrambling, owing to the strong non-covalent interaction between the two heavy chain C-terminal regions.
  • a method with denaturing conditions such as SDS-PAGE or CE-SDS
  • the two half antibody molecules will dissociate from each other and appear as LMW impurities.
  • the half antibody molecule was confidently identified ( FIG. 3B ).
  • the good agreement between the measured mass (73,099.5 Da) and the predicted mass (73,100.1 Da) suggested that, unlike extensively modified free light chain, no substantial modification was associated with the formation of the half antibody, consistent with the disulfide bond scrambling pathway.
  • Fab-truncated species were also identified in mAb-1 sample by the HILIC-MS method ( FIG. 3C ). On the HILIC column, those species were eluted right before the main peak as a partially resolved shouldering peak. The subsequent mass measurement confirmed the identity of the Fab-truncated species and truncation sites were located at the hinge region consistent with those observed in the Fab fragments. Despite the inferior resolution in separation compared to electrophoretic methods, the HILIC-MS method prevails in high fidelity and specificity by accurate mass measurement, making unambiguous identification possible.
  • the Fab-truncated species are very difficult to remove from the main species during purification, due to the preserved Fc region that binds to Protein A.
  • Monoclonal antibody molecules missing a Fab arm are expected to exhibit compromised potency as one of the two target binding sites is not present.
  • losing one Fab arm is detrimental for drug activity, since both Fab arms are essential to achieve the designed therapeutic functions. Therefore, the ability to detect and identify those species directly in a drug substance sample is highly important because time and resources are not only saved, but also avoids the introduction of possible artifacts during the enrichment process.
  • the H2L species comprised of two heavy chains and one light chain, are frequently observed as the most abundant LMW impurities by SDS-PAGE and CE-SDS methods, but their identification is usually only supported by indirect and insufficient evidence.
  • N-terminal sequencing analysis of the H2L-containing minor band on SDS-PAGE can reveal the first several amino acid residues of both heavy chain and light chain, and the signal intensity ratio between heavy chain and light chain might suggest a 2 to 1 ratio.
  • In-gel digestion of the H2L-containing minor band followed by LC-MS based peptide mapping analysis can confidently confirm the presence of both heavy chain and light chain. However, it is difficult to establish an accurate ratio between heavy chain and light chain.
  • H2L species Both methods provide partial identification of H2L species, but neither can reveal a complete structure.
  • HILIC-MS method offers direct identification of H2L species at the intact protein level.
  • mAb-1 product a homogeneous H2L species was observed to co-elute with the Fab-truncated species slightly ahead of the intact antibody.
  • the detected mass (122,851.0 Da) of the H2L species was approximately 34 Da smaller than the predicted molecular weight of a H2L molecule (122,884.5 Da).
  • the ⁇ -elimination of a heavy chain cysteine residue presumably the one originally forming the inter-heavy and light chain disulfide bond, was the root-cause of forming H2L species in mAb-1 sample.
  • LMW impurities with different heavy chain and light chain combinations can be generated, including free light chain, free heavy chain, half antibody, heavy chain dimer and H2L species.
  • these predicted LMW species were detected in the HILIC separation with the elution order consistent with their relative size.
  • two more LMW species, specifically, the free heavy chain species and the heavy chain dimer species were observed to elute between the free light chain and half antibody, and slightly after the half antibody (partially resolved peak), respectively.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Plant Substances (AREA)
US16/223,463 2017-12-22 2018-12-18 System and method for characterizing drug product impurities Abandoned US20190194298A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/223,463 US20190194298A1 (en) 2017-12-22 2018-12-18 System and method for characterizing drug product impurities
US17/677,499 US20220169704A1 (en) 2017-12-22 2022-02-22 System and method for characterizing drug product impurities

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762610029P 2017-12-22 2017-12-22
US201862743632P 2018-10-10 2018-10-10
US16/223,463 US20190194298A1 (en) 2017-12-22 2018-12-18 System and method for characterizing drug product impurities

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/677,499 Division US20220169704A1 (en) 2017-12-22 2022-02-22 System and method for characterizing drug product impurities

Publications (1)

Publication Number Publication Date
US20190194298A1 true US20190194298A1 (en) 2019-06-27

Family

ID=65201678

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/223,463 Abandoned US20190194298A1 (en) 2017-12-22 2018-12-18 System and method for characterizing drug product impurities
US17/677,499 Pending US20220169704A1 (en) 2017-12-22 2022-02-22 System and method for characterizing drug product impurities

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/677,499 Pending US20220169704A1 (en) 2017-12-22 2022-02-22 System and method for characterizing drug product impurities

Country Status (14)

Country Link
US (2) US20190194298A1 (es)
EP (1) EP3727629A1 (es)
JP (2) JP7321159B2 (es)
KR (1) KR20200103728A (es)
CN (2) CN111479618B (es)
AR (1) AR114045A1 (es)
AU (1) AU2018392334A1 (es)
BR (1) BR112020010615A2 (es)
CA (1) CA3084059A1 (es)
IL (1) IL275404A (es)
MX (1) MX2020006639A (es)
SG (1) SG11202004380QA (es)
TW (2) TW202402787A (es)
WO (1) WO2019126123A1 (es)

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7087411B2 (en) 1999-06-08 2006-08-08 Regeneron Pharmaceuticals, Inc. Fusion protein capable of binding VEGF
US6927004B2 (en) 2002-03-08 2005-08-09 Asml Netherlands B.V. Mask for use in lithography, method of making a mask, lithographic apparatus, and device manufacturing method
EP2261230B1 (en) * 2002-09-11 2017-05-10 Chugai Seiyaku Kabushiki Kaisha Protein purification method
RU2337968C2 (ru) * 2003-01-09 2008-11-10 Дженентек, Инк. Способ очистки гетерологичного полипептида
ES2398076T3 (es) 2006-06-02 2013-03-13 Regeneron Pharmaceuticals, Inc. Anticuerpos de alta afinidad contra el receptor de IL-6 humano
US7608693B2 (en) 2006-10-02 2009-10-27 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to human IL-4 receptor
PL2178916T3 (pl) 2007-07-31 2015-08-31 Regeneron Pharma Ludzkie przeciwciała przeciwko ludzkiemu CD20 i sposób ich zastosowania
US8309088B2 (en) 2007-08-10 2012-11-13 Regeneron Pharmaceuticals, Inc. Method of treating osteoarthritis with an antibody to NGF
JO3672B1 (ar) 2008-12-15 2020-08-27 Regeneron Pharma أجسام مضادة بشرية عالية التفاعل الكيماوي بالنسبة لإنزيم سبتيليسين كنفرتيز بروبروتين / كيكسين نوع 9 (pcsk9).
EP2975051B1 (en) 2009-06-26 2021-04-14 Regeneron Pharmaceuticals, Inc. Readily isolated bispecific antibodies with native immunoglobulin format
JO3417B1 (ar) 2010-01-08 2019-10-20 Regeneron Pharma الصيغ المستقرة التي تحتوي على الأجسام المضادة لمضاد مستقبل( interleukin-6 (il-6r
JO3340B1 (ar) 2010-05-26 2019-03-13 Regeneron Pharma مضادات حيوية لـعامل تمايز النمو 8 البشري
JOP20190250A1 (ar) 2010-07-14 2017-06-16 Regeneron Pharma صيغ مستقرة تحتوي على الأجسام المضادة لمضاد عامل نمو الأعصاب
AR083044A1 (es) 2010-09-27 2013-01-30 Regeneron Pharma Anticuerpos anti-cd48 y usos de los mismos
CN106267189B (zh) 2010-10-06 2021-02-26 瑞泽恩制药公司 含有抗白介素-4受体(il-4r)的抗体的稳定制剂
JO3756B1 (ar) 2010-11-23 2021-01-31 Regeneron Pharma اجسام مضادة بشرية لمستقبلات الجلوكاجون
AR087329A1 (es) 2011-06-17 2014-03-19 Regeneron Pharma Anticuerpos humanos contra proteina 3 de tipo angiopoietina humana
LT2780368T (lt) 2011-11-14 2018-03-12 Regeneron Pharmaceuticals, Inc. Kompozicijos ir būdai raumenų masės padidinimui ir raumenų sustiprinimui, specifiškai antagonizuojant gdf8 ir (arba) aktiviną a
EP2807190B1 (en) 2012-01-23 2018-12-26 Regeneron Pharmaceuticals, Inc. Stabilized formulations containing anti-ang2 antibodies
JO3820B1 (ar) 2012-05-03 2021-01-31 Regeneron Pharma أجسام مضادة بشرية لـ fel d1وطرق لاستخدامها
TW201843172A (zh) 2012-06-25 2018-12-16 美商再生元醫藥公司 抗-egfr抗體及其用途
EA028244B1 (ru) 2012-08-13 2017-10-31 Ридженерон Фармасьютикалз, Инк. АНТИТЕЛА К PCSK9 C pH-ЗАВИСИМЫМИ ХАРАКТЕРИСТИКАМИ СВЯЗЫВАНИЯ
JOP20200236A1 (ar) 2012-09-21 2017-06-16 Regeneron Pharma الأجسام المضادة لمضاد cd3 وجزيئات ربط الأنتيجين ثنائية التحديد التي تربط cd3 وcd20 واستخداماتها
JO3405B1 (ar) 2013-01-09 2019-10-20 Regeneron Pharma الأجسام المضادة لمضاد مستقبل عامل النمو المشتق من الصفائح الدموية - بيتا واستخداماتها
JO3532B1 (ar) 2013-03-13 2020-07-05 Regeneron Pharma الأجسام المضادة لمضاد انترلوكين-33 واستعمالاتها
TWI659968B (zh) 2013-03-14 2019-05-21 再生元醫藥公司 針對呼吸道融合病毒f蛋白質的人類抗體及其使用方法
US9637535B2 (en) 2013-03-15 2017-05-02 Regeneron Pharmaceuticals, Inc. IL-33 antagonists and uses thereof
TWI641620B (zh) 2013-08-21 2018-11-21 再生元醫藥公司 抗-prlr抗體及其用途
US20160251441A1 (en) 2013-10-25 2016-09-01 Medimmune, Llc Antibody purification
US11092574B2 (en) * 2013-12-24 2021-08-17 Waters Technologies Corporation Materials for hydrophilic interaction chromatography and processes for preparation and use thereof for analysis of glycoproteins and glycopeptides
US10119944B2 (en) * 2013-12-24 2018-11-06 Waters Technologies Corporation Materials for hydrophilic interaction chromatography and processes for preparation and use thereof for analysis of glycoproteins and glycopeptides
TWI680138B (zh) 2014-01-23 2019-12-21 美商再生元醫藥公司 抗pd-l1之人類抗體
TWI681969B (zh) 2014-01-23 2020-01-11 美商再生元醫藥公司 針對pd-1的人類抗體
JP6632984B2 (ja) 2014-03-11 2020-01-22 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. 抗EGFRvIII抗体およびその使用
TWI701042B (zh) 2014-03-19 2020-08-11 美商再生元醫藥公司 用於腫瘤治療之方法及抗體組成物
EP3636073B1 (en) 2014-05-05 2023-11-15 Regeneron Pharmaceuticals, Inc. Humanized c5 and c3 animals
JO3701B1 (ar) 2014-05-23 2021-01-31 Regeneron Pharma مضادات حيوية بشرية لمتلازمة الشرق الأوسط التنفسية - بروتين كورونا فيروس الشوكي
EA201790377A1 (ru) 2014-09-16 2017-08-31 Регенерон Фармасьютикалз, Инк. Антитела к глюкагону и их применения
TWI710573B (zh) 2015-01-26 2020-11-21 美商再生元醫藥公司 抗伊波拉病毒醣蛋白之人類抗體

Also Published As

Publication number Publication date
CN115925882A (zh) 2023-04-07
EP3727629A1 (en) 2020-10-28
KR20200103728A (ko) 2020-09-02
CN111479618A (zh) 2020-07-31
WO2019126123A1 (en) 2019-06-27
MX2020006639A (es) 2020-09-14
BR112020010615A2 (pt) 2020-10-27
CA3084059A1 (en) 2019-06-27
TWI811277B (zh) 2023-08-11
IL275404A (en) 2020-07-30
US20220169704A1 (en) 2022-06-02
JP2021507878A (ja) 2021-02-25
AU2018392334A1 (en) 2020-05-28
AR114045A1 (es) 2020-07-15
CN111479618B (zh) 2022-08-02
TW201938581A (zh) 2019-10-01
TW202402787A (zh) 2024-01-16
SG11202004380QA (en) 2020-06-29
JP2023139186A (ja) 2023-10-03
JP7321159B2 (ja) 2023-08-04

Similar Documents

Publication Publication Date Title
US11525823B2 (en) System and method for characterizing protein dimerization
JP7349998B2 (ja) サイズバリアントおよび電荷バリアントである薬物製品不純物を特徴解析するためのシステムおよび方法
US20220169704A1 (en) System and method for characterizing drug product impurities

Legal Events

Date Code Title Description
AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WANG, SHUNHAI;REEL/FRAME:048884/0263

Effective date: 20190327

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION