US20220081479A1 - Methods of producing ustekinumab - Google Patents

Methods of producing ustekinumab Download PDF

Info

Publication number
US20220081479A1
US20220081479A1 US17/419,541 US201917419541A US2022081479A1 US 20220081479 A1 US20220081479 A1 US 20220081479A1 US 201917419541 A US201917419541 A US 201917419541A US 2022081479 A1 US2022081479 A1 US 2022081479A1
Authority
US
United States
Prior art keywords
ustekinumab
glycan
cells
level
range
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.)
Pending
Application number
US17/419,541
Other languages
English (en)
Inventor
Ru Zang
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.)
Momenta Pharmaceuticals Inc
Original Assignee
Momenta 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 Momenta Pharmaceuticals Inc filed Critical Momenta Pharmaceuticals Inc
Priority to US17/419,541 priority Critical patent/US20220081479A1/en
Publication of US20220081479A1 publication Critical patent/US20220081479A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • 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
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • Therapeutic antibodies are an important class of therapeutic biological products. Antibody glycosylation and glycan composition can impact antibody activity and effector functions. There remains an ongoing need for improved methods for controlling the glycan profile of antibody products.
  • the present disclosure provides process levers for producing (e.g., manufacturing) ustekinumab with a target level of one or more glycans.
  • the present disclosure provides the insight that a relationship between galactose concentration in a culture media and time of culturing can be used as a process lever for controlling a target level of one or more glycans (e.g., galactosylation) of ustekinumab in a culture.
  • the present disclosure identifies an inverse relationship between culture time and galactose concentration in culture media in controlling a target level of one or more glycans (e.g., galactosylation).
  • the disclosure provides methods of manufacturing ustekinumab with a target level of one or more glycans.
  • Such methods can include providing (e.g., producing, expressing (e.g., in small scale or large scale cell culture) and/or manufacturing) or obtaining (e.g., receiving and/or purchasing from a third party (including a contractually related third party or a non-contractually-related (e.g., an independent) third party)) an ustekinumab test protein (e.g., an ustekinumab drug substance, e.g., a preparation of ustekinumab drug substance, e.g., a batch of test ustekinumab dug substance).
  • a third party including a contractually related third party or a non-contractually-related (e.g., an independent) third party
  • an ustekinumab test protein e.g., an ustekinumab drug substance, e.g., a preparation of ustek
  • the disclosure provides methods of manufacturing a pharmaceutical composition comprising ustekinumab having a target level of one or more glycans, the method comprising: selecting a level of galactose and a time for cell culture, where the level of galactose and the time are inversely related; culturing a population of cells genetically engineered to express ustekinumab under conditions comprising the selected level of galactose and time; harvesting ustekinumab expressed by the population of cells, thereby producing the preparation of ustekinumab; and purifying, concentrating, and/or formulating the ustekinumab preparation to produce pharmaceutical composition comprising ustekinumab if the preparation meets the target level of one or more glycans.
  • the target level of one or more glycans is a target level of a glycan selected from the group consisting of G0F, sialylated glycan, and G2F. In some embodiments, the target level of one or more glycans is a target level of G0F.
  • the disclosure provides methods of manufacturing a pharmaceutical composition comprising ustekinumab having a target level of G0F glycan, the method comprising: selecting a target level of G0F glycan; selecting a level of galactose and a time for cell culture to provide the selected target level of G0F, where the level of galactose and the time are inversely related; culturing a population of cells genetically engineered to express ustekinumab under conditions comprising the selected level of galactose and time; harvesting ustekinumab expressed by the population of cells, thereby producing the preparation of ustekinumab; and purifying, concentrating, and/or formulating the ustekinumab preparation to produce pharmaceutical composition comprising ustekinumab if the preparation meets the target level of G0F glycan.
  • the disclosure provides methods of producing an ustekinumab drug product having a target level of one or more glycans, the method comprising: culturing a population of cells genetically engineered to express ustekinumab under conditions, where the conditions are characterized by parameters including a selected level of galactose and a time for cell culture, and where the level of galactose and the time are inversely related; harvesting ustekinumab expressed by the cell, thereby producing an ustekinumab preparation; and purifying, concentrating, and/or formulating the ustekinumab preparation to produce an ustekinumab drug product if the ustekinumab preparation meets the target level of one or more glycans.
  • the target level of one or more glycans is a target level of a glycan selected from the group consisting of G0F, sialylated glycan, and G2F. In some embodiments, the target level of one or more glycans is a target level of G0F.
  • provided methods include culturing a population of mammalian cells that are genetically engineered to express ustekinumab.
  • mammalian cells are selected from: CHO cells, HEK 293 cells, fibrosarcoma HT 1080 cells, PER.C6 cells, CAP cells, HKB-11 cells, HuH-7 cells, NS0 cells and SP 2/0 cells.
  • the culturing step is performed using continuous culture process.
  • provided methods include culturing that is performed using a perfusion culture process (e.g., an alternating tangential flow filter (ATF)-based perfusion culture process).
  • ATF alternating tangential flow filter
  • provided methods include culturing mammalian cells that are genetically engineered to express ustekinumab (e.g., SP 2/0 cells that express ustekinumab) by a perfusion culture process.
  • a target level of G0F glycan is within a range of 20% to 80% G0F relative to total glycan. In some embodiments, a target level of G0F glycan is within a range of 25% to 65% G0F relative to total glycan.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of G0F glycan within a range of 20% up to 40% G0F relative to total glycan.
  • a method for achieving ustekinumab having a target level of G0F glycan within a range of 20% up to 40% G0F relative to total glycan includes a selected level of galactose at 0 mM and a time for cell culturing is within a range of 7 days to 15 days.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of G0F glycan within a range of 40% to 80% G0F relative to total glycan.
  • a method for achieving ustekinumab having a target level of G0F glycan within a range of 40% to 80% G0F relative to total glycan include a selected level of galactose within a range of 15 mM to 30 mM and a time for cell culture that is within a range of 16 days to 60 days. In some certain embodiments, a time for cell culturing is within a range of 25 days to 42 days.
  • provided methods include selecting a target level of G0F glycan.
  • a target level of G0F glycan is within a range of 20% to 80% G0F relative to total glycan.
  • a target level of G0F glycan within a range of 20% up to 40% G0F relative to total glycan, where the method includes a selected level of galactose at 0 mM and a time for cell culture that is within a range of 7 days to 15 days.
  • a target level of G0F glycan within a range of 40% to 80% G0F relative to total glycan where the method includes a selected level of galactose within a range of 15 mM to 30 mM and a time for cell culture that is within a range of 16 days to 60 days.
  • provided methods further include measuring a level of G0F glycan. In some embodiments, if the measured level of G0F glycan is within a range of 20% to 80% G0F relative to total glycan (e.g., within a range of 20% up to 40% G0F relative to total glycan, e.g., within a range of 40% to 80% G0F relative to total glycan), then a step of purifying, concentrating, and/or formulating the ustekinumab preparation is performed.
  • FIG. 1 depicts comparison of glycan profiles of various preparations and sources of ustekinumab.
  • Abundance of G0F glycan is provided in the upper left panel; abundance of G1F-A glycan is provided in the upper right panel; abundance of G1F-B glycan is provided in the lower left panel; and total sialylation provided in the lower right panel.
  • Solid circles represent reference protein products (“RPPs”) from the U.S. (black solid circles) and Europe (shaded solid circles). Symbols in the right-most portion of each panel represent ustekinumab test preparations cultured at different volumes.
  • RPPs reference protein products
  • Solid shaded squares depicting samples cultured at 3 L Solid shaded squares depicting samples cultured at 3 L, open squares depicting samples from satellite culture, solid triangles depicting samples cultured at 100 L, and solid diamonds depicting samples cultured at 250 L.
  • FIG. 2 depicts G0F glycan profile variation with harvest duration.
  • Left-most column (white) represents Group 1 RPP G0F abundance
  • second column from the left (black) represents Group 2 RPP G0F abundance
  • third column from the left represents the average abundance of G0F from samples cultured between 7 days and 42 days.
  • Shaded columns (starting at fourth column from the left and continuing to the right most column) each represent the G0F abundance from samples cultured an indicated number of days within a range of 7 days (D7) up to 42 days (D42, right most column).
  • FIG. 3 depicts comparisons of major glycan profiles and charge variants between various culture conditions. Walking along the x-axis of each panel from left to right are depicts: Group 1 RPPs (left-most), Group 2 RPPs, ustekinumab cultured at 3 L DS, ustekinumab cultured at 100 L DS, ustekinumab cultured at 250 L DS, NCM-2 ustekinumab preparation, and NCM-1 ustekinumab preparation (right-most).
  • the terms “about” or “approximately,” as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the terms “about” or “approximately” refer to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less of the stated reference value.
  • control, controlled, controlling in reference to controlling a target level of one or more glycans (e.g., galactose, e.g., G0F) of ustekinumab means selecting, maintaining and/or adjusting one or more culture conditions for production of ustekinumab. Adjusting can including increasing or decreasing one or more culture conditions for production of ustekinumab.
  • a controlled target level of one or more glycans relates to production of ustekinumab with a desired level of one or more glycans that has minimal product drift.
  • a controlled target level of G0F will vary no more than 20%, 15%, 10%, or 5% between samples in the same production run and/or batch. In some embodiments, controlling a target level of one or more glycan ensures consistency in the production of ustekinumab (e.g., batch to batch consistency, consistency throughout samples from any particular production process).
  • a “glycan” refers to a compound comprising at least one sugar residue (e.g., monosaccharide). Glycans can be monomers or polymers of sugar residues, and can be linear or branched. A glycan may include natural sugar residues (e.g., glucose, N-acetylglucosamine, N-acetyl neuraminic acid, galactose, mannose, fucose, hexose, arabinose, ribose, xylose, etc.) and/or modified sugars (e.g., 2′-fluororibose, 2′-deoxyribose, phosphomannose, 6′-sulfo N-acetylglucosamine, etc).
  • natural sugar residues e.g., glucose, N-acetylglucosamine, N-acetyl neuraminic acid, galactose, mannose, fucose, hexose,
  • glycan includes homopolymers and heteropolymers of sugar residues.
  • glycan also encompasses a glycan component of a glycoconjugate (e.g., a glycoprotein, glycolipid, proteoglycan, etc.).
  • glycoconjugate e.g., a glycoprotein, glycolipid, proteoglycan, etc.
  • free glycans including glycans that have been cleaved or otherwise released from a glycoconjugate.
  • a “galactosylated glycan” refers to a glycan that includes at least one galactose sugar residue.
  • a galactosylated glycan is a G1, G2, G1F, G2F, A1, and/or A2 glycan.
  • a non-galactosylated glycan includes G0F or G0.
  • a target level of galactosylated glycan may refer to the presence of galactosylated glycan (e.g., G2F) and/or a target level of non-galactosylated glycan (e.g., G0F).
  • isolated refers to a substance and/or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an experimental setting), and/or (2) designed, produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% of the other components with which they were initially associated.
  • isolated agents are about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure.
  • a substance is “pure” if it is substantially free of other components.
  • a substance may still be considered “isolated” or even “pure”, after having been combined with certain other components such as, for example, one or more carriers or excipients (e.g., buffer, solvent, water, etc.); in such embodiments, percent isolation or purity of the substance is calculated without including such carriers or excipients.
  • a biological polymer such as a polypeptide or polynucleotide that occurs in nature is considered to be “isolated” when, a) by virtue of its origin or source of derivation is not associated with some or all of the components that accompany it in its native state in nature; b) it is substantially free of other polypeptides or nucleic acids of the same species from the species that produces it in nature; c) is expressed by or is otherwise in association with components from a cell or other expression system that is not of the species that produces it in nature.
  • a polypeptide that is chemically synthesized or is synthesized in a cellular system different from that which produces it in nature is considered to be an “isolated” polypeptide.
  • a polypeptide that has been subjected to one or more purification techniques may be considered to be an “isolated” polypeptide to the extent that it has been separated from other components a) with which it is associated in nature; and/or b) with which it was associated when initially produced.
  • process lever refers to an element of a culture process (e.g., one or more culture conditions) that can be controlled in order to increase or decrease the abundance of one or more glycans on an antibody product.
  • the present disclosure provides a novel process lever that is a specified relationship between galactose concentration in the culture media and culture time.
  • an inverse relationship between galactose concentration in a culture media and the time of culture in the culture media can be used as a process lever to produce ustekinumab with a target level of one or more glycans (e.g., galactosylation).
  • a process lever includes a selected a level of galactose and time for cell culture to control level of one or more glycans (e.g., galactosylation, e.g., G0F) of ustekinumab (e.g., level of one or more glycans in a ustekinumab culture or preparation).
  • level of galactose and time for cell culture to control level of one or more glycans e.g., galactosylation, e.g., G0F
  • ustekinumab e.g., level of one or more glycans in a ustekinumab culture or preparation.
  • an “N-glycosylation site of an Fc region” refers to an amino acid residue within an Fc region to which a glycan is N-linked.
  • a N-glycosylation site of ustekinumab is located in a heavy chain at position Asn299.
  • a “protein,” as used herein, is a polypeptide (i.e., a string of at least two amino acids linked to one another by peptide bonds). Proteins may include moieties other than amino acids (e.g., may be glycoproteins) and/or may be otherwise processed or modified. Those of ordinary skill in the art will appreciate that a “protein” can be a complete polypeptide chain as produced by a cell (with or without a signal sequence), or can be a functional portion thereof. Those of ordinary skill will further appreciate that a protein can sometimes include more than one polypeptide chain, for example linked by one or more disulfide bonds or associated by other means.
  • recovery refers to the process of rendering an agent or entity substantially free of other previously-associated components, for example by isolation, e.g., using purification techniques known in the art.
  • an agent or entity is recovered from a natural source and/or a source comprising cells.
  • sample(s) refer to separately procured samples.
  • evaluation of separate samples includes evaluation of samples from the same culture run (e.g., at different time points during preparation) or from different culture runs (e.g., different rounds of culture).
  • Target value or target level refers to a predetermined level of one or more particular glycans, such as galactosylated glycans and/or sialylated glycans.
  • a target value is a level of one or more particular glycans, such as galactosylated glycans (e.g., G0, G1, G2, G0F, G1F, G2F or combinations) and/or sialylated glycans (e.g., monosialylated, disialylated, or combinations), in a reference ustekinumab product or described in a specification or master batch record for a pharmaceutical product.
  • a target value is a level of G0F glycans in an ustekinumab product.
  • a target value refers to an absolute level of (e.g., number of moles of) one or more glycans (e.g., galactosylated glycans (e.g., one or more species of galactosylated glycans) and/or sialylated glycans (e.g., one or more species of sialylated glycans)) in a ustekinumab preparation.
  • glycans e.g., galactosylated glycans (e.g., one or more species of galactosylated glycans) and/or sialylated glycans (e.g., one or more species of sialylated glycans)
  • a target value refers to a level of one or more glycans (e.g., galactosylated glycans (e.g., one or more species of galactosylated glycans) and/or sialylated glycans (e.g., one or more species of sialylated glycans)) in a ustekinumab preparation relative to total level of glycans in the ustekinumab preparation.
  • glycans e.g., galactosylated glycans (e.g., one or more species of galactosylated glycans) and/or sialylated glycans (e.g., one or more species of sialylated glycans)
  • a target value is expressed as a “percent”, which refers to the number of moles of one or more glycans (e.g., Fc glycans) relative to total moles of glycans (e.g., Fc glycans) in a ustekinumab preparation. In some embodiments, “percent” refers to the number of moles of one or more PNGase F-released Fc glycans relative to total moles of PNGase P-released Fc glycans detected.
  • Ustekinumab preparation refers to a mixture of ustekinumab proteins obtained according to a particular production method.
  • Ustekinumab proteins in an ustekinumab preparation includes multiple copies of ustekinumab (i.e., having the same or substantially the same amino acid sequence) but have a mixture of glycans associated with the protein.
  • an ustekinumab preparation is prepared using a method and/or system as provided herein. Production methods can include a recombinant preparation step using cultured cells that have been engineered to express ustekinumab (or to express ustekinumab at a relevant level or under relevant conditions).
  • a production method may include an isolation step in which ustekinumab is isolated from certain components of the engineered cells (e.g., by lysing the cells and pelleting the protein component by centrifugation).
  • production method may also include a purification step in which ustekinumab is separated (e.g., by chromatography) from other cellular components, e.g., other proteins or organic components that were used in earlier steps. It will be appreciated that these steps are non-limiting and that any number of additional productions steps may be included.
  • Different ustekinumab preparations may be prepared by the same production method but on different occasions (e.g., different runs or preparations). Alternatively, different ustekinumab preparations may be prepared by different production methods. Two production methods may differ in any way (e.g., expression vector, engineered cell type, culture conditions, isolation procedure, purification conditions, etc.).
  • the present disclosure describes, at least in part, the discovery of process levers for producing ustekinumab having targeted levels of one or more glycans (e.g., galactosylation, e.g., G0F glycans). Controlling glycan composition and levels during antibody production is a continuing challenge. Glycosylation of therapeutic antibodies can impact their safety and/or efficacy. (Zhang et al. (2016) Drug Discovery Today 21(5): 740-765). Accordingly, it is important to be able to ensure consistency of glycan composition in the production of ustekinumab (e.g., batch to batch consistency, consistency throughout samples from any particular production process). During characterization of ustekinumab reference protein products (“RPPs”), two distinct glycan populations of ustekinumab RPPs were observed.
  • RPPs reference protein products
  • the present disclosure describes the development of levers for controlling glycan composition during manufacture of ustekinumab.
  • Process levers are described herein to produce ustekinumab with target levels of particular glycans, including each of the RPPs glycan profiles identified.
  • the present disclosure identified a relationship between galactose concentration in a culture media and time of culturing.
  • the present disclosure provides the insight that a relationship between galactose concentration in a culture media and time of culturing (e.g., continuously culturing) can be used as a process lever for controlling a target level of one or more glycans (e.g., galactosylation) of ustekinumab in a culture.
  • the present disclosure further provides the insight that the for controlling the glycan level (e.g., galactosylation), a relationship between a level of galactose and time (i.e., duration of culturing) are inversely related.
  • Culturing methods of the present disclosure for controlling glycan compositions include continuous culture methods (e.g., perfusion culture).
  • the present disclosure provides, in part, methods and processes for manufacturing, preparing, controlling or otherwise generating ustekinumab with particular glycan profiles.
  • Ustekinumab is an antibody that specifically binds to the p-40 subunit of both IL-12 and IL-23. Ustekinumab has been studied in a number of human diseases including psoriasis, psoriatic arthritis, Crohn's disease and multiple sclerosis.
  • ustekinumab includes HCDR1, HCDR2, and HCDR3 sequences that differ by no more than 3 amino acid residues from the HCDR sequences as set forth in SEQ ID NO.: 1 and LCDR1, LCDR2, and LCDR3 sequences that differ by no more than 3 amino acid residues from the LCDR sequences as set forth in SEQ ID NO.: 2.
  • ustekinumab includes HCDR1, HCDR2, and HCDR3 sequences that differ by no more than 2 amino acid residues or by no more than 1 amino acid residue from the HCDR sequences as set forth in SEQ ID NO.: 1 and LCDR1, LCDR2, and LCDR3 sequences that differ no more than 2 amino acid residues or by no more than 1 amino acid residue from the LCDR sequences as set forth in SEQ ID NO.: 2.
  • ustekinumab includes a heavy chain variable domain that differs by no more than 3 amino acid resides from the sequence as set forth in SEQ ID NO.: 1 and a light chain variable domain that differs by no more than 3 amino acid resides from the sequence as set forth in SEQ ID NO.: 2.
  • ustekinumab includes a heavy chain that includes a sequence that differs by no more than 5 amino acids from the sequence as set forth in SEQ ID NO.: 1 and a light chain that includes a sequence that differs by no more than 5 amino acid resides from the sequence of SEQ ID NO.: 2.
  • ustekinumab includes HCDR1, HCDR2, and HCDR3 sequences as set forth in SEQ ID NO.: 1 and LCDR1, LCDR2, and LCDR3 sequences as set forth in SEQ ID NO.: 2.
  • ustekinumab includes a heavy chain variable domain as set forth in SEQ ID NO.: 1 and a light chain variable domain as set forth in SEQ ID NO.: 2.
  • ustekinumab includes a heavy chain comprising a sequence of SEQ ID NO.: 1 and/or a light chain comprising a sequence of SEQ ID NO.: 2.
  • the disclosure provides methods of manufacturing a pharmaceutical composition comprising ustekinumab having a target level of one or more glycans, the method comprising: selecting a level of galactose and a time for cell culture, where the level of galactose and the time are inversely related; culturing a population of cells genetically engineered to express ustekinumab under conditions comprising the selected level of galactose and time; harvesting ustekinumab expressed by the population of cells, thereby producing the preparation of ustekinumab; and purifying, concentrating, and/or formulating the ustekinumab preparation to produce pharmaceutical composition comprising ustekinumab if the preparation meets the target level of one or more glycans.
  • the target level of one or more glycans is a target level of a glycan selected from the group consisting of G0F, sialylated glycan, and G2F. In some embodiments, the target level of one or more glycans is a target level of G0F.
  • the disclosure provides methods of producing an ustekinumab drug product having a target level of one or more glycans, the method comprising: culturing a population of cells genetically engineered to express ustekinumab under conditions, where the conditions are characterized by parameters including a selected level of galactose and a time for cell culture, and where the level of galactose and the time are inversely related; harvesting ustekinumab expressed by the cell, thereby producing an ustekinumab preparation; and purifying, concentrating, and/or formulating the ustekinumab preparation to produce an ustekinumab drug product if the ustekinumab preparation meets the target level of one or more glycans.
  • the target level of one or more glycans is a target level of a glycan selected from the group consisting of G0F, sialylated glycan, and G2F. In some embodiments, the target level of one or more glycans is a target level of G0F.
  • a target level of G0F glycan is within a range of 20% to 80% G0F relative to total glycan. In some embodiments, a target level of G0F glycan is within a range of 25% to 65% G0F relative to total glycan.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of G0F glycan within a range of 20% up to 40% G0F relative to total glycan.
  • a method for achieving ustekinumab having a target level of G0F glycan within a range of 20% up to 40% G0F relative to total glycan includes a selected level of galactose at 0 mM and a time for cell culture that is within a range of 7 days to 15 days.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of G0F glycan within a range of 40% to 80% G0F relative to total glycan.
  • a method for achieving ustekinumab having a target level of G0F glycan within a range of 40% to 80% G0F relative to total glycan include a selected level of galactose within a range of 15 mM to 30 mM and a time for cell culture that is within a range of 16 days to 60 days. In some certain embodiments, a time for cell cultures is within a range of 25 days to 42 days.
  • provided methods further include selecting a target level of G0F glycan.
  • a target level of G0F glycan is within a range of 20% to 80% G0F relative to total glycan.
  • a target level of G0F glycan within a range of 20% up to 40% G0F relative to total glycan, where the method includes a selected level of galactose at 0 mM and a time for cell culture that is within a range of 7 days to 15 days.
  • a target level of G0F glycan within a range of 40% to 80% G0F relative to total glycan where the method includes a selected level of galactose within a range of 15 mM to 30 mM and a time for cell culture that is within a range of 16 days to 60 days.
  • provided methods further include measuring a level of G0F glycan. In some embodiments, if the measured level of G0F glycan is within a range of 20% to 80% G0F relative to total glycan (e.g., within a range of 20% up to 40% G0F relative to total glycan, e.g., within a range of 40% to 80% G0F relative to total glycan), then a step of purifying, concentrating, and/or formulating the ustekinumab preparation is performed.
  • provided methods for manufacturing and/or producing ustekinumab having a target glycan level include a target level of sialylated glycan and/or G2F glycan.
  • a target level of sialylated glycan and/or G2F glycan Provided in Table 1 below are conditions for ustekinumab with target levels of sialylated glycan and/or G2F glycan.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of sialylated glycan within a range of 15% to 30% sialylated glycan relative to total glycan.
  • a method for achieving ustekinumab having a target level of sialylated glycan within a range of 15% to 30% sialylated glycan relative to total glycan includes a selected level of galactose at 0 mM and a time for cell culturing is within a range of 7 days to 15 days.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of sialylated glycan within a range of 5% up to 15% sialylated glycan relative to total glycan.
  • a method for achieving ustekinumab having a target level of G0F glycan within a range of 5% up to 15% sialylated glycan relative to total glycan include a selected level of galactose within a range of 15 mM to 30 mM and a time for cell culture that is within a range of 16 days to 60 days. In some certain embodiments, a time for cell culturing is within a range of 25 days to 42 days.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of G2F glycan within a range of 5% to 10% G2F relative to total glycan.
  • a method for achieving ustekinumab having a target level of G2F glycan within a range of 5% to 10% G2F relative to total glycan includes a selected level of galactose at 0 mM and a time for cell culture that is within a range of 7 days to 15 days.
  • provided methods are directed to manufacturing and/or producing ustekinumab having a target level of G2F glycan within a range of 1% up to 5% G0F relative to total glycan.
  • a method for achieving ustekinumab having a target level of G2F glycan within a range of 1% up to 5% G2F relative to total glycan include a selected level of galactose within a range of 15 mM to 30 mM and a time for cell culture that is within a range of 16 days to 60 days. In some certain embodiments, a time for cell culturing is within a range of 25 days to 42 days.
  • provided methods include culturing a population of mammalian cells that are genetically engineered to express ustekinumab.
  • mammalian cells are selected from: CHO cells, HEK 293 cells, fibrosarcoma HT 1080 cells, PER.C6 cells, CAP cells, HKB-11 cells, HuH-7 cells, NS0 cells and SP 2/0 cells.
  • the culturing step is performed using continuous culture process.
  • provided methods include culturing that is performed using a perfusion culture process (e.g., an alternating tangential flow filter (ATF)-based perfusion culture process).
  • ATF alternating tangential flow filter
  • provided methods include culturing mammalian cells that are genetically engineered to express ustekinumab (e.g., SP 2/0 cells that express ustekinumab) by a perfusion culture process.
  • the target value is a predetermined pharmaceutical product specification or a quality control criterion for a pharmaceutical preparation, e.g., a Certificate of Analysis (CofA), a Certificate of Testing (CofT), or a Master Batch Record.
  • the product specification is a product description in an FDA label, a Physician's Insert, a USP monograph, or an EP monograph.
  • cell culture methods of the present disclosure including culturing at a temperature within a range of 25° C. to 40° C. and with gravity as it is encountered on earth.
  • provided methods of culturing a population of cells is sufficient for expression of an ustekinumab product.
  • Cell culture media generally comprise an appropriate source of energy and compounds which regulate the cell cycle.
  • culture media includes, for example amino acids, vitamins, inorganic salts, and glucose, which is known to those of skill in the art.
  • a cell culture media has a pH of 6 to 8.
  • Media for animal cell culture are well established in the art, and are routinely optimized by the skilled artisan for the particular purpose and/or cell type.
  • methods of the present disclosure include culturing a population of cells via a continuous cell culture system (e.g., perfusion cell culture system).
  • a continuous cell culture system includes bioreactor tank and a cell retention device.
  • a continuous cell culture system includes a bioreactor tank, a cell retention device, a media supply, and a bleed waste collection.
  • a continuous cell culture system includes a population of cells (e.g., a population of cells engineered to express ustekinumab, e.g., a population of cells consisting of cells engineered to express ustekinumab) and cell culture media.
  • a cell retention device is or includes a continuous centrifuge, an alternating tangential flow filter (ATF), a tangential flow membrane filter (TFF), a dynamic filter, a spin-filter, an ultrasonic and dielectrophoretic separator, or a gravity settler.
  • ATF alternating tangential flow filter
  • TDF tangential flow membrane filter
  • dynamic filter a dynamic filter
  • spin-filter a spin-filter
  • ultrasonic and dielectrophoretic separator or a gravity settler.
  • a cell retention device is or includes an ATF.
  • a bioreactor system includes a stirred-tank bioreactor, a cell retention device, a media supply, and a bleed waste collection.
  • a bioreactor system (e.g., a perfusion bioreactor system) includes a sparger.
  • a bioreactor system includes a drilled hole sparger.
  • a bioreactor system includes an open pipe sparger.
  • a bioreactor system includes a sintered sparger.
  • methods of the present disclosure include culturing a population of cells (e.g., a population of cells engineered to express ustekinumab, e.g., a population of cells consisting of cells engineered to express ustekinumab) at a volume within a range of 1 L to 3000 L.
  • provided methods include culturing mammalian cells that are genetically engineered to express ustekinumab at a volume of at least 25 L, 50 L, 100 L, 200 L, 250 L, 400 L, 500 L, 600 L, 800 L, 1000 L, or 2000 L.
  • provided methods include culturing mammalian cells that are genetically engineered to express ustekinumab at a volume of about 25 L to about 250 L.
  • glycans of ustekinumab are analyzed (e.g., measured) by any available suitable method.
  • glycan structure and composition as described herein are analyzed, for example, by one or more, enzymatic, chromatographic, mass spectrometry (MS), chromatographic followed by MS, electrophoretic methods, electrophoretic methods followed by MS, nuclear magnetic resonance (NMR) methods, and combinations thereof.
  • exemplary enzymatic methods include contacting a ustekinumab preparation with one or more enzymes under conditions and for a time sufficient to release one or more glycan(s) (e.g., one or more exposed glycan(s)).
  • the one or more enzymes include(s) PNGase F.
  • Exemplary chromatographic methods include, but are not limited to, Strong Anion Exchange chromatography using Pulsed Amperometric Detection (SAX-PAD), liquid chromatography (LC), high performance liquid chromatography (HPLC), ultra-performance liquid chromatography (UPLC), thin layer chromatography (TLC), amide column chromatography, and combinations thereof.
  • MS mass spectrometry
  • MALDI-MS matrix assisted laser desorption ionization mass spectrometry
  • FTMS Fourier transform mass spectrometry
  • IMS-MS ion mobility separation with mass spectrometry
  • ETD-MS electron transfer dissociation
  • Exemplary electrophoretic methods include, but are not limited to, capillary electrophoresis (CE), CE-MS, gel electrophoresis, agarose gel electrophoresis, acrylamide gel electrophoresis, SDS-polyacrylamide gel electrophoresis (SDS-PAGE) followed by Western blotting using antibodies that recognize specific glycan structures, and combinations thereof.
  • CE capillary electrophoresis
  • CE-MS gel electrophoresis
  • agarose gel electrophoresis agarose gel electrophoresis
  • acrylamide gel electrophoresis acrylamide gel electrophoresis
  • SDS-PAGE SDS-polyacrylamide gel electrophoresis
  • Exemplary nuclear magnetic resonance include, but are not limited to, one-dimensional NMR (1D-NMR), two-dimensional NMR (2D-NMR), correlation spectroscopy magnetic-angle spinning NMR (COSY-NMR), total correlated spectroscopy NMR (TOCSY-NMR), heteronuclear single-quantum coherence NMR (HSQC-NMR), heteronuclear multiple quantum coherence (HMQC-NMR), rotational nuclear overhauser effect spectroscopy NMR (ROESY-NMR), nuclear overhauser effect spectroscopy (NOESY-NMR), and combinations thereof.
  • a cell engineered to express ustekinumab includes one or more nucleic acids that encode an ustekinumab product that includes a heavy chain variable domain as set forth in SEQ ID NO.: 1 and/or a light chain variable domain as set forth in SEQ ID NO.: 2.
  • a cell engineered to express ustekinumab includes one or more nucleic acids that encode an ustekinumab product that includes a heavy chain comprising a sequence of SEQ ID NO.: 1 and/or a light chain comprising a sequence of SEQ ID NO.: 2.
  • a cell engineered to express ustekinumab includes one or more nucleic acids that encode an ustekinumab product that includes HCDR1, HCDR2, and HCDR3 sequences as set forth in SEQ ID NO.: 1 and LCDR1, LCDR2, and LCDR3 sequences as set forth in SEQ ID NO.: 2.
  • a cell engineered to express ustekinumab includes one or more nucleic acids that encode an ustekinumab product that has the same primary amino acid sequence as a protein that has been approved, e.g., under a secondary approval process, for therapeutic or diagnostic use in humans or animals.
  • a cell engineered to express ustekinumab includes one or more nucleic acids that encode an ustekinumab product that differs by no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 residues with an approved therapeutic or diagnostic protein.
  • a cell engineered to express ustekinumab includes one or more nucleic acids that encode a protein that has at least 90, 95, 98, 99% or 100% sequence identity with that of an approved therapeutic or diagnostic protein.
  • a protein preparation or product includes amino acid variants, e.g., species that differ at terminal residues, e.g., at one or two terminal residues.
  • sequence identity compared is the identity between the primary amino acid sequence of the most abundant (e.g., most abundant active) species in each of the products being compared.
  • sequence identity refers to the amino acid sequence encoded by a nucleic acid that can be used to make the ustekinumab product.
  • cells engineered to express ustekinumab are mammalian cells.
  • cells engineered to express ustekinumab are murine cells.
  • a cell engineered to express ustekinumab is from a mouse cell line.
  • Murine (e.g., mouse) cell lines include, for example, mouse myeloma cell lines, e.g., NS0 cells and SP 2/0 cells.
  • cells engineered to express ustekinumab are human cells.
  • a cell engineered to express ustekinumab is from a human cell line.
  • Human cell lines include, for example, HEK 293: human embryonic kidney 293; HT-1080: from a fibrosarcoma with an epithelial-like phenotype; PER.C6: from human embryonic retinal cells immortalized via transfection with the adenovirus E1 gene; CAP: from human amniocytes immortalized through an adenovirus type 5 E1 gene; HKB-11: created through polyethylene glycol fusion of HEK293-S and a human B-cell line; and HuH-7: from a human hepatocellular carcinoma.
  • shear-sensitive cells are selected from: HEK 293 cells, fibrosarcoma HT 1080 cells, PER.C6 cells, CAP cells, HKB-11 cells and HuH-7 cells.
  • a population of cells that express an ustekinumab product as described herein is produced using recombinant methods.
  • Recombinant expression of a gene such as a gene encoding a polypeptide, such as an antibody agent described herein, can include construction of an expression vector containing a polynucleotide that encodes the polypeptide.
  • a vector for the production of the polypeptide can be produced by recombinant DNA technology using techniques known in the art.
  • Known methods can be used to construct expression vectors containing polypeptide coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • an ustekinumab product described herein can be purified by any method known in the art for purification, for example, by chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • ustekinumab can be isolated and purified by appropriately selecting and combining affinity columns such as Protein A column with chromatography columns, filtration, ultra-filtration, salting-out and dialysis procedures (see Antibodies: A Laboratory Manual, Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988).
  • an ustekinumab product can be fused to heterologous polypeptide sequences to facilitate purification.
  • a ustekinumab product produced or manufactured using any of the methods, systems and/or processes described herein can be incorporated into a pharmaceutical composition.
  • Such a pharmaceutical composition may be useful in the prevention and/or treatment of diseases.
  • Pharmaceutical compositions comprising ustekinumab can be formulated by methods known to those skilled in the art (see, e.g., Remington's Pharmaceutical Sciences, 20th Ed., Lippincott Williams & Wilkins, 2000).
  • the pharmaceutical composition can be administered parenterally in the form of an injectable formulation comprising a sterile solution or suspension in water or another pharmaceutically acceptable liquid.
  • the pharmaceutical composition can be formulated by suitably combining the cell product (e.g., a recombinant protein, e.g., a glycoprotein, e.g., an antibody agent) with pharmaceutically acceptable vehicles or media, such as sterile water and physiological saline, vegetable oil, emulsifier, suspension agent, surfactant, stabilizer, flavoring excipient, diluent, vehicle, preservative, binder, followed by mixing in a unit dose form required for generally accepted pharmaceutical practices.
  • a recombinant protein e.g., a glycoprotein, e.g., an antibody agent
  • pharmaceutically acceptable vehicles or media such as sterile water and physiological saline, vegetable oil, emulsifier, suspension agent, surfactant, stabilizer, flavoring excipient, diluent, vehicle, preservative, binder, followed by mixing in a unit dose form required for generally accepted pharmaceutical practices.
  • the amount of active ingredient included in a pharmaceutical preparation is such that a suitable
  • a preparation of ustekinumab includes sucrose as a stabilizer/tonicifier. In some embodiments, a preparation of ustekinumab includes histidine (e.g., L-histidine) as a buffer. In some embodiments, a preparation of ustekinumab includes polisorbate 80 as a surfactant. In some certain embodiments, a preparation of ustekinumab includes histidine (e.g., L-histidine), sucrose, and polisorbate 80.
  • a preparation of ustekinumab is formulated for parenteral administration, e.g., intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection. In some embodiments, a preparation of ustekinumab is formulated for subcutaneous administration.
  • This example identifies a significant shift in glycan composition among lots of commercially available ustekinumab reference protein products (“RPPs”). Specifically, the present example demonstrates that the variability in the glycan composition among lots of commercially available ustekinumab that segregates into two distinct glycan profiles. These two groups of glycan variants are referred to herein as “Group 1,” which are characterized by a relatively high level of G0F glycan, and “Group 2,” which characterized by a relatively low level of G0F glycan.
  • Ustekinumab test products generated by culturing (e.g., by an ATF-based perfusion culture process) with 5 mM galactose for a duration of 42 days at different volumes within a range of 3 L to 250 L were also analyzed.
  • the glycan abundance of these ustekinumab test products is depicted in the right most grouping of each panel of FIG. 1 .
  • the glycan profiles of the ustekinumab test products were within the range of those of Group 1 and Group 2 RPPs.
  • the present example identified two distinct groups of glycan profiles for commercially available ustekinumab RPPs and demonstrated that contemporaneous culture conditions yield ustekinumab test products with intermediate glycan abundances within the ranges of these two groups.
  • the present example demonstrates that harvest collection timing is an element for controlling ustekinumab glycan composition.
  • the abundance of G0F glycan relative to total glycan generally decreased with culture duration.
  • ustekinumab preparations that were harvested at 13 days of culturing had an almost 50% abundance of G0F glycans, while samples with harvested after 34 or more days of culturing had less than 30% G0F glycans relative to total glycan composition.
  • the present disclosure provides the recognition that a decrease in the culture duration (i.e. time till harvest) is coupled with an increase in G0F level; while an increase in the culture duration (i.e. time till harvest) is coupled with a reduction in G0F level.
  • this example demonstrates that increasing culture time can decrease abundance of G0F glycans relative to total glycan composition and further describes that culture time can be varied to control ustekinumab glycan composition.
  • the present example identifies an inverse relationship between culture time and galactose concentration in culture media that can serve as a process lever for controlling ustekinumab glycan levels (e.g., galactosylation).
  • the present example demonstrates production of non-conforming materials (i.e., non-conforming ustekinumab preparations) with engineered glycan profiles.
  • non-conforming materials i.e., non-conforming ustekinumab preparations
  • NCM-1 and NCM-2 two non-conforming ustekinumab preparations
  • NCM-1 preparations were generated by culturing (e.g., by an ATF-based perfusion culture process) with 15 mM galactose and harvesting ustekinumab before day 16 of culturing (e.g., harvesting at time points within a range of day 7 to day 15).
  • NCM-2 preparations were generated by culturing (e.g., by an ATF-based perfusion culture process) in a medium lacking galactose (0 mM galactose) and harvesting ustekinumab after day 25 of culturing (e.g., harvesting at time points within a range of day 34 to day 45). As depicted in FIG.
  • Table 3 provides exemplary target ranges for each glycan as engineered by controlling the process levers of galactose concentration and culture time.
  • inversely related elements of galactose concentration and culture time can be used as process levers to control glycan composition and can be used to produce non-conforming ustekinumab preparations with target glycan levels.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Steroid Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US17/419,541 2018-12-31 2019-12-20 Methods of producing ustekinumab Pending US20220081479A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/419,541 US20220081479A1 (en) 2018-12-31 2019-12-20 Methods of producing ustekinumab

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862786821P 2018-12-31 2018-12-31
PCT/US2019/067916 WO2020142275A1 (en) 2018-12-31 2019-12-20 Methods of producing ustekinumab
US17/419,541 US20220081479A1 (en) 2018-12-31 2019-12-20 Methods of producing ustekinumab

Publications (1)

Publication Number Publication Date
US20220081479A1 true US20220081479A1 (en) 2022-03-17

Family

ID=71407091

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/419,480 Pending US20220033487A1 (en) 2018-12-31 2019-12-20 Methods of Producing Ustekinumab
US17/419,541 Pending US20220081479A1 (en) 2018-12-31 2019-12-20 Methods of producing ustekinumab

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US17/419,480 Pending US20220033487A1 (en) 2018-12-31 2019-12-20 Methods of Producing Ustekinumab

Country Status (12)

Country Link
US (2) US20220033487A1 (es)
EP (1) EP3906257A4 (es)
JP (1) JP2022516510A (es)
KR (1) KR20210131323A (es)
CN (1) CN113474366A (es)
AU (1) AU2019419372A1 (es)
BR (1) BR112021012973A2 (es)
CA (1) CA3125443A1 (es)
EA (1) EA202191840A1 (es)
IL (1) IL284374A (es)
MX (1) MX2021008030A (es)
WO (1) WO2020142275A1 (es)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11719704B2 (en) 2015-12-30 2023-08-08 Momenta Pharmaceuticals, Inc. Methods related to biologics

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112852743B (zh) * 2021-01-25 2021-11-02 江苏荃信生物医药有限公司 用于生产乌司奴单抗的生物类似药的细胞株及生产方法
WO2024092203A1 (en) * 2022-10-28 2024-05-02 Amgen Inc. Compositions of anti-interleukin 12/interleukin 23 antibody

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2484142C2 (ru) * 2009-01-22 2013-06-10 Момента Фармасьютикалз, Инк. Содержащие галактоза-альфа-1,3-галактозу n-гликаны в гликопротеиновых продуктах, полученных из клеток сно
US20140271622A1 (en) * 2013-03-14 2014-09-18 Momenta Pharmaceuticals, Inc. Methods of cell culture
AU2014237635B2 (en) * 2013-03-15 2020-03-12 Janssen Biologics B.V. Manufacturing methods to control C-terminal lysine, galactose and sialic acid content in recombinant proteins
US20170166941A1 (en) * 2014-01-29 2017-06-15 Lg Life Sciences Ltd. Method for modulating galactosylation of recombinant protein through optimization of culture medium
CA2969225C (en) * 2014-12-01 2023-08-22 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
CN105779394B (zh) * 2015-03-20 2020-03-24 广东东阳光药业有限公司 一种降低抗体酸性峰含量和改良抗体糖型的细胞培养方法
AU2017264541A1 (en) * 2016-05-10 2018-12-13 Ares Trading S.A. Methods for modulating protein galactosylation profiles of recombinant proteins using peracetyl galactose
WO2018024770A1 (en) * 2016-08-03 2018-02-08 Formycon Ag Production of biosimilar ustekinumab in cho cells
WO2018224673A1 (en) * 2017-06-08 2018-12-13 Zaklady Farmaceutyczne Polpharma S.A. Improved methods of cell culture

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11719704B2 (en) 2015-12-30 2023-08-08 Momenta Pharmaceuticals, Inc. Methods related to biologics

Also Published As

Publication number Publication date
CA3125443A1 (en) 2020-07-09
AU2019419372A1 (en) 2021-07-08
EP3906257A4 (en) 2022-09-14
WO2020142275A1 (en) 2020-07-09
US20220033487A1 (en) 2022-02-03
JP2022516510A (ja) 2022-02-28
CN113474366A (zh) 2021-10-01
IL284374A (en) 2021-08-31
EA202191840A1 (ru) 2021-09-03
BR112021012973A2 (pt) 2021-09-14
KR20210131323A (ko) 2021-11-02
MX2021008030A (es) 2021-08-05
EP3906257A1 (en) 2021-11-10

Similar Documents

Publication Publication Date Title
US20220081479A1 (en) Methods of producing ustekinumab
US10533045B2 (en) Antibody purification and purity monitoring
ES2732679T3 (es) Procedimientos de cultivo celular
WO2018003983A1 (ja) hANP-Fc含有分子コンジュゲート
HU230065B1 (hu) Baktérium gazdatörzsek
KR20150138273A (ko) 단백질의 피로­글루타민산 형성을 증가시키기 위한 방법
US20220267413A1 (en) Methods For The Treatment Of Neurodegeneration
CA3055438A1 (en) Multispecific antibody constructs binding to muc1 and cd3
CN110865129B (zh) 一种度拉鲁肽中多种修饰水平的检测方法
US20160024179A1 (en) Modified fc fusion proteins
US20210222109A1 (en) Methods of continuous cell culture
EA046459B1 (ru) Способы получения устекинумаба
Srebreva et al. Specificity studies on anti‐histone H1 antibodies obtained by different immunization methods
De la Luz-Hernandez et al. Cancer vaccine characterization: from bench to clinic
EP3239168B1 (en) Derivative of conotoxin peptide kappa-cptx-btl02, preparation method therefor, and uses thereof
EP3287468B1 (en) Two conotoxin peptides, preparation methods therefor, and applications thereof
EP3239167B1 (en) Derivative of conotoxin peptide kappa-cptx-btl01, preparation method therefor, and uses thereof
EP4285934A1 (en) Modified trimannose-oligosacharides, bisected n-glycans comprising said trimannose core and method for obtaining them
WO2018025270A1 (en) Enriched modified vitamin d binding protein compositions and use thereof
SCHAPIRA Isolation and characterization of free cytoplasmic messenger ribonucleoproteins from rabbit reticulocyte
EP2725104A1 (en) Method for preparing polymeric protein composed of monomeric protein produced by fusing protein having immunoglobulin fold structure to protein capable of serving as subunit structure
DE69937872T2 (de) Anti-humaner thymidylat-synthase-monoklonaler Antikörper und Hybridoma, das zu seiner Herstellung fähig ist
CN114149501A (zh) 抗c5抗体及其应用
CN116381067A (zh) 羊驼和骆驼血液中纳米抗体的提取鉴定和开发应用

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

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

Free format text: MISSASSIGNED APPLICATION NUMBER