WO2008128230A1 - Produits de glycoprotéines de référence et procédés associés - Google Patents

Produits de glycoprotéines de référence et procédés associés Download PDF

Info

Publication number
WO2008128230A1
WO2008128230A1 PCT/US2008/060365 US2008060365W WO2008128230A1 WO 2008128230 A1 WO2008128230 A1 WO 2008128230A1 US 2008060365 W US2008060365 W US 2008060365W WO 2008128230 A1 WO2008128230 A1 WO 2008128230A1
Authority
WO
WIPO (PCT)
Prior art keywords
production
glycan
reference glycoprotein
production parameters
cell
Prior art date
Application number
PCT/US2008/060365
Other languages
English (en)
Inventor
Rajeev Chillakuru
Brian Edward Collins
Carlos J. Bosques
Lakshmanan Thiruneelakantapillai
Dorota A. Bulik
Ian Christopher Parsons
Zachary Shriver
Ganesh Venkataraman
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.
Publication of WO2008128230A1 publication Critical patent/WO2008128230A1/fr

Links

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B50/00ICT programming tools or database systems specially adapted for bioinformatics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins

Definitions

  • the invention relates to reference glycoprotein products and related methods, e.g., methods of making glycoprotein products and methods of designing processes to make reference glycoprotein products having defined physical and functional properties.
  • small molecule drugs These drugs exist as simple chemical structures that are synthetically derived. The active ingredient generally exists as a homogenous product. These small molecule drugs and preparations thereof, can be chemically characterized and are generally readily manufactured through comparatively simple chemical synthesis.
  • a typical glycoprotein product differs substantially in terms of complexity from a typical small molecule drug.
  • the sugar structures attached to the amino acid backbone of a glycoprotein can vary structurally in many ways including, sequence, branching, sugar content, and heterogeneity.
  • glycoprotein products can be complex heterogeneous mixtures of many structurally diverse molecules which themselves have complex glycan structures.
  • GJycosylation adds not only to the molecules structural complexity but affects or conditions many of a glycoprotein's biological and clinical attributes.
  • the reference glycoprotiens are glycoproteins which have been approved for commercial marketing, e.g., by a government agency charged with conferring such approval.
  • the methods allow precise replication of approved marketed reference glycoproteins.
  • Some methods rely on the use of databases which include correlations between production parameters and required glycan characteristics.
  • the database can provide production parameters for incorporation into a production protocol.
  • the methods allows for the replication of reference glycoprotiens.
  • the invention features, a method for making a reference glycoprotein product which has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval, including the steps of:
  • the invention features, a method for making a reference glycoprotein product which has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval, including the steps of:
  • c) optionally, providing a database that correlates, defines, identifies, relates, or provides, each of a plurality of required glycan properties as a correlative function of one or more production parameters or combinations of production parameters;
  • methods, databases, and systems disclosed herein can include or use various types of correlations between production parameters and the glycan characteristics they condition. These are referred to as correlative functions.
  • the production of glycoproteins is a complex process and correlations provided in the databases can reflect this.
  • Exemplary correlative functions include non-linear correlative functions.
  • a nonlinear correlation can reflect a relationship between production parameters and glycan characteristics wherein the effect of two (or more) production parameters acting together on a glycan characteristic is not the same as the combination of a first production parameter (acting alone) on the glycan characteristic together with the effect of a second production parameter (acting alone) on the glycan characteristic.
  • correlative functions useful in the methods, databases and systems described herein include constrained, pleiotropic and tunable correlative functions.
  • constrained correlative functions reflect the complexity of glycoprotein synthesis and can represent relationships characterized by incompatible or undesirable combinations or production parameters or glycan characteristic. E.g., a combination of production parameters may be constrained because it results in an undesirable glycan characteristic.
  • Pleiotropic correlative functions can reflect the varied effect of one or more production parameter on different glycan characteristics.
  • a tunable function is one that can allow for a plurality of inputs, e.g., inputs of differing magnitudes, and a plurality of outputs, e.g., of differing magnitude. It can allow the adjustment of a glycan property by the adjustment of a production parameter.
  • the database includes ten or more, e.g., 20, 25, 50, 100, 1 50, 200, 300, 350, 400, 500, 600, /00, 800, 900 or more, tunable, nonlinear, pleiotropic, or constrained correlations.
  • a selected production parameter is associated with a tunable, nonlinear, pJeioiropic, or constrained correlation.
  • a first production parameter Xl is selected by a correlative function between production parameter Xl and a glycan characteristic Yl and a glycan characteristic Y2 and a second production parameter X2 is selected to modify the affect of Xl on Y2.
  • the invention features, a method for making a reference glycoprotein product which has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval, including:
  • a required production parameter is one which is correlated with a required glycan characteristic
  • a production system e.g., a cell culture system, which incorporates a required production parameter
  • selecting the reference glycoprotein providing a glycan pattern representing glycan structures on the reference glycoprotein, e.g., by releasing glycans from the reference glycoprotein, e.g., by enzymatic digestion, and optionally by separating the released glycans, e.g., Io produce fractions or peaks representing one or more required glycan characteristics, selecting a requis ed glycan characteristic, selecting from the database one or more required production parameters or combinations of production parameters that correlates with the required glycan characteristic.
  • providing a required production parameter includes receiving the identity of the parameter from another entity.
  • a first entity performs one or more of a), b) and c) and a second entity performs one or more of steps i), ii), and iii) and transmits the identity of the required parameter to the first entity.
  • a single entity may perform all steps or may receive or by provided with information or selections needed to practice by one or more second entity.
  • the database includes ten or more, e.g., 20, 25, 50, 100, 150, 200, 300, 350, 400, 500, 600, 700, 800, 900 or more, tunable, nonlinear, pleiotropic, or constrained correlation.
  • a required production parameter is associated with a tunable, nonlinear, pleiotropic, or constrained correlation.
  • the invention features, a method for making a reference glycoprotein product which has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval, including the steps of:
  • production parameters sequentially selecting at least 2, 3, 4 or 5 production parameters to provide the required glycan characteristics, e.g., wherein said production parameters are selected from the group consisting of: cell identity, cell culture conditions, fermentation conditions, isolation conditions, and formulation conditions, and combinations thereof.
  • the invention features, a method, which can be a computer- implemented method, including:
  • glycoprotein property e.g., a glycoprotein characteristic, which is associated with said production parameter
  • the invention features, a method, which is can be a computer- implemented method, including:
  • glycoprotein property e.g., a glycoprotein characteristic, for a reference glycoprotein
  • the database can include one or more of a tunable, nonlinear, pleiolropic, or constrained correlation.
  • a required or selected production parameter is associated with a tunable, nonlinear, pleiotropic, or constrained correlation.
  • embodiments can be computer implemented. In other embodiments the method is not computer implemented, e.g., a database relied on is not computei implemented. Embodiments can include displaying, outputting, or memorializing a selected production parameter or glycan characteristic.
  • Methods disclosed herein allow for designing protocols or selecting conditions for making reference glycoproteins.
  • the methods allow for the choice of production parameters, which when incorporated into a protocol for making a glycoprotein, provide for the incorporation into the glycoprotein of preselected glycan structures required on a reference glycoprotein.
  • the invention features, a method for designing a process to produce a reference glycoprotein product, or selecting an element of a process for making, a reference glycoprotein product wherein the reference glycoprotein has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval, including:
  • c) optionally, providing a database that correlates, defines, identifies, relates. 01 piovides, each of a plurality of required glycan properties as a correlative function of one or more production parameters or combinations of production parameters; and
  • the method is a computer implemented method.
  • the method is not a computer implemented method.
  • the method includes displaying, outputting, or memorializing said selected production parameter.
  • the method includes selecting the glycan properties required by the reference glycoprotein product and then selecting the production parameters, e.g., those selected in d), to provide the required glycan properties.
  • the invention features, a method for designing a process to produce a reference glycoprotein product, or selecting an element of a process for making, a reference glycoprotein product wherein the reference glycoprotein has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval, including the steps of the method including the steps of:
  • production parameters sequentially selecting at least 2, 3, 4 or 5 production parameters to provide the required glycan characteristics, e.g., wherein said production parameters are selected from the group consisting of: cell identity, cell culture conditions, fermentation conditions, isolation conditions, and formulation conditions, and combinations thereof;
  • the database can include one or more of a tunable, nonlinear, pleiotropic, or constrained correlation.
  • a required or selected production parameter is associated with a tunable, nonlinear, pleiotropic, or constrained correlation.
  • embodiments can be computer implemented. In other embodiments the method is not computer implemented, e.g., a database relied on is not computer implemented. Embodiments can include displaying, outputting, or memorializing a selected production parameter or glycan characteristic.
  • Methods, databases and systems described herein can be used in a variety of applications, including methods of quality control or production monitoring for a reference glycoprotien.
  • methods disclosed herein can be used to monitor a glycoprotein made by a defined process to determine if the process actually produces the reference glycoprotein complete with required glycan structures.
  • Rg., if the refernce glycoprotein is analyzed and found not to have a required gycan characteristic methods described herein can be used to select alterations in the production process to tune or alter the process so that it produces a properly glycosylated reference glycoprotein.
  • the invention features, a method of monitoring and/or controlling the production of a reference glycoprotein product wherein the reference glycoprotein has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval.
  • the method includes:
  • the method further includes the step of ) providing an observed glycan characteristic from a reference glycoprotein made by the altered production process and evaluating it as described herein. In embodiments steps b), c) and d) are repeated for a glycoprotein made by the altered production process.
  • the method is repeated, e.g., at predetermined intervals.
  • the observed glycan property was or is determined by: providing a glycan pattern representing glycan structures on the reference glycoprotein made by the preselected production process, e.g., by releasing gl yeans from the reference glycoprotein, e.g., by enzymatic digestion, and optionally by separating the released glycans, e.g., to produce fractions or peaks representing one or more glycan characteristic.
  • the standard value was or is determined by: providing a glycan pattern representing glycan structures on a commercially available reference glycoprotein;
  • the standard value was or is determined by: providing a glycan pattern representing glycan structures on the reference glycoprotein made by the preselected production process, e.g., by a different, earlier run of the preselected process, or by a different production process, e.g., an altered production process, e.g., by releasing glycans from the reference glycoprotein, e.g., by enzymatic digestion, and optionally by separating the released glycans, e.g., to produce fractions or peaks representing one or more glycan characteristics.
  • the database includes one or more of at least a tunable, nonlinear, pleiotropic, or constrained correlation.
  • a selected production parameter is associated with a tunable, nonlinear, pleiotropic, or constrained correlation.
  • the invention features, a database described herein configured for designing a process to produce a reference glycoprotein product, or selecting an element of a process for making, a reference glycoprotein product, wherein the reference glycoprotein has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval.
  • the database is disposed on tangible medium.
  • the database is disposed on a single unit of tangible medium, e.g., on a single computer, or in a single paper document.
  • the database is provided on more than one unit of tangible medium, e.g., on more than one computer, in more than a single paper document, partly on a paper document and partly on computer readable medium.
  • the database is disposed on computer readable medium.
  • every dement of the database is not stored in the same place, compute, memory or location.
  • the database is configured to allow computerized access.
  • the database is configured to allow access without the use of a computer, e.g., it is on a printed document.
  • the database includes a plurality of records wherein a record includes, an identifier for a production parameter or a combination of production parameters, an identifier for a glcyan property, e.g., a functional property conditioned by a glycan, or a glycan characteristic (i.e., a structural characteristic), and a correlative function between the production parameter (or combination) and the glycan property, which e.g., correlates, defines, identifies, relates, or provides, one to the other.
  • the correlative function is a tunable function.
  • the correlative function relates X to Y, where X is a value for an element related to a production parameter and Y is a value for an element related to the glycan property and allows adjustment of the value for X to select or identify a value
  • the database includes at least 10, 20, 30, 40, 50 records.
  • the correlation is a positive or negative correlation.
  • the con-elation was or can be established by empirical testing or by prediction.
  • Io an embodiment (he correlation is qualitative, e.g., positive, negative, or no correlation.
  • the correlation is quantitative, e.g., a positive correlation can be expressed as a series of scores increasingly higher correlation.
  • the correlation is expressed in absolute terms or as relative to a standard, e.g., as more or less, how much, more or less likely to confer a particular glycan characteristic on a protein, as another method.
  • the invention features, a system including:
  • a selector to select a production parameter for a reference glycoprotein based on an input glycoprotein property or to select a glycoprotein property of a reference glycoprotein based on an input production parameter.
  • system is configured for designing a process to produce a reference glycoprotein product, or selecting an element of a process for making, a reference glycoprotein product wherein the reference glycoprotein has been approved for commercial marketing, e.g., by a government agency charged with conferring such approval, including:
  • a database described herein e.g., a database that that correlates, defines, identifies, relates, or provides, each of a plurality of glycan properties as a correlative function of one or more production parameters or combinations of production parameters;
  • the system is configuied to allow the design of a process Io produce a reference glycoprotein product having a preselected glycan property, e.g., to select a production parameter for the use m a method of producing a glycoprotein having a selected glycan property.
  • the query is based on a selected glycan property, e.g., of a reference glycoprotein product, and said query result includes one or more production parameters or combinations of production parameters from the database that correlate with the selected glycan property.
  • the query is based on one or more production parameters from the database that correlate with a selected glycan property and said query result is based on a glycan property con-elated with said production parameter(s).
  • the user interface is configured to allow input of a desired glycan property and said processor is configured to allow output of a query result based on a correlated production parameter.
  • the user interface is configured to allow input of a desired production parameter and said processor is configured to allow output of a query result based on a correlated glycan property.
  • the system is configured to allow input of one or more values of X and output, e.g., a query result, of one or more values of Y, wherein a correlative function in said database relates X to Y, where X is a value for an element related to a production parameter and Y is a value for an element related to the glycan property, and said system is configured for adjustment of the value for X to select or identify a value for Y.
  • system is configured to allow input of one or more values of Y and output of one or more values of X, wherein a correlative function in said database relates X to Y, where X is a value foi an element related to a production parametei and Y is a value for an clement related to the glycan property and the system is configured for adjustment of the value for Y to select or identify a value for X,
  • a production parameter 1 is tunable for an input setting (or value) Xl and the output or setting (or value) for Yl will vary with the setting (or value) of Xl
  • a production parameter 2 is tunable for an input setting (or value) X2 and the output or setting (or value) for Y2 will vary with the setting (or value) of X2.
  • some combination of values or settings for Xl and X2, or Yl and Y2, are not compatible and the solution space, or total number of possibilities for the available combinations of Yl and Y2, is less than the product of number of possibilities for Yl and the number of possibilities for Y2 (or the analogous situation for X1X2).
  • a constraint on solution space is imposed by incompatibilities on combinations of Xl and X2, e.g., they may be concentrations of additives or combinations of additives and cells which cannot be combined for one reason or another.
  • a constraint on solution space is imposed because a combination of Yl and Y2 are synthetically or structurally impossible or result in toxicity to the cell culture or to an unwanted property in a glycoprotein.
  • a correlative function produces a null output or a signal corresponding to an unavailable combination.
  • system is configured with a filter which identifies prohibited or unavailable combinations of X 1X2 or Y] Y2 and labels them or removes them from output.
  • a selection for a value for parameter X2 will be made based at least in part on the value chosen for XJ .
  • the method includes displaying, outputling, or memorializing said selected production parameter.
  • the system includes a coi relative function which is a tunable, nonlinear, pleiotropic, or constrained correlation.
  • a tunable function can allow for a plurality of inputs, e.g., inputs of differing magnitudes, and a plurality of outputs, e.g., of differing magnitude. It can allow the adjustment of a glycan characteristic by the adjustment of a production parameter. .
  • a correlative function relates X to Y, where X is a value for an element related to a production parameter and Y is a value for an element related to the glycan characteristic and allows adjustment of the value for X to select or identify a value for Y or the adjustment of the value for Y to select or identify a value for X.
  • X can be any of a value for concentration of an additive, a value of a by product, a value of a physical parameter, a value of time, a value of cell type, a value of gene expression level of copy number, and, in one or more of those cases, Y the amount of a glycan structure on a refernence glycoprotein.
  • a production parameter 1 is tunable for an input setting (or value) Xl and the output or setting (or value) for Yl will vary with the setting (or value) of XI .
  • a production parameter 2 is tunable for an input setting (or value) X? and the output or setting (or value) for Y7 will vary with the setting (or value) of X7.
  • some combination of values or settings for Xl and X2, or YI and Y2 are not compatible and the solution space, or total number of possibilities for the available combinations of Yl and Y2, is less than the product of number of possibilities for Y 1 and the number of possibilities for Y2 (or the analogous situation for X1X2).
  • a constraint on solution space imposed by incompatibilities on combinations of Xl and X2, e.g., they may be concentrations of additives or combinations of additives and cells which cannot be combined for one reason or another.
  • a constraint on solution space is imposed because a combination of Yl and Y2 are synthetically or structurally impossible or result in toxicity to the cell culture or to an unwanted property in a reference glycoprotein.
  • a correlative function produces a null output or a signal corresponding to an unavailable combination.
  • a nonlinear correlation can reflect a relationship between production parameters and glycan characteristics wherein the effect of two (or more) production parameters acting together on a glycan characteristic is not the same as the combination of the first production parameter (acting alone) on the glycan characteristic together with the effect of the second production parameter (acting alone) on the glycan characteristic.
  • This can be expressed as "X1,X2 -Yl ⁇ Xl-Yl + X2-Y1, in the notation used herein.
  • a correlative function relates values for more than one value for production parameters (e.g., Xl, X2, and so on) to one or more glycan characteristic, e.g., Y, and wherein the effect of the combination, e.g., the combination of Xl and X2, on Y is nonlinear.
  • the correlation is nonlinear when the effect of a plurality of production parameters acting together, e.g., production parameters Xl and X2 (acting together), on one or more glycan characteristics, e.g., Y, is not the same as the combination of Xl (acting alone) on Y together with the effect of X2 (acting alone) on Y.
  • glucosamine results in a decrease in galactosylation (Yl), a decrease in fucosylation (Y2), an increase in high mannose structures (Y3), and an increase in hybrid structures (Y4).
  • uridine gives a decreases high mannose structurictures (Yl) but no change of the other glycan characteristics (Yl . Y?, and Y4). If glucosamine (XJ ) and uridine (X2) are combined all four parameteis, Yl , Y?, Y 3 and Y4, are unchanged.
  • the coi relative function between X ) ,X2 and Y 1 is nonlinear.
  • the correlative function between Xl , X2 and Y2 is nonlinear and the correlative function between Xl , X2 and Y4 is nonlinear.
  • single X correlations which are nonlinear when taken together, are also considered, individually, to be nonlinear.
  • the correlation of glucosamine (Xl) with fucosylation (Y2) and the correlation of glucosamine (Xl) with hybrid structures (Y4) are all nonlinear.
  • the correlation between uridine (X2) with galactosylation (Yl), the correlation of uridine (X2) with fucosylation (Y2) and the correlation of uridine (X2) with hybrid structures (Y4) are considered nonlinear correlations in some embodiments.
  • Constrained correlative functions reflect the complexity of glycoprotein synthesis and can represent relationships characterized by incompatible or undesirable combinations or production parameters or glycan characteristics. E.g., a combination of production parameters may be constrained because it results in an undesirable glycan characteristic.
  • a correlative function relates a value for a first production parameter Xl to a first to a value for a first glycan characteristic Yl, but also identifies either or both of: an additional, e.g., second, glycan characteristic Y2 which is altered by Xl ; and an additional, e.g., second, production parameter X2 which can be used along with the first production parameter, e.g., to modulate, e.g., minimize, the overall effect on a second glycan characteristic Y2.
  • This correlation is referred to as a constrained production parameter, because the use of Xl may require the use of X2 as well to avoid an unwanted affect on glycan characteristic Y2.
  • the selection of a first production parameter may constrain the selection of a second production parameter and makes the selection of a specific second production parameter more or Jess favored, because, e.g., of a positive or negative affect on the conferral of a glycan characteristic on the protein if the second parameter is (or is not) combined with the first.
  • the addition of glucosamine, Xl is correlated with a decrease in galactosylation.
  • Xl is also correlated with an increase in high mannose .
  • the addition of uridine, X2 minimizes the increase in high mannose without abolishing the Xl mediated decrease in galactosylation.
  • Xl is constrained.
  • the Xl-Yl correlation or an X1-Y1,Y2 correlation can identify X2 as an additional production parameter to be considered or altered in conjunction with Xl .
  • Pleiotropic correlative functions can reflect the varied effect of one or more production parameters on different glycan characteristics.
  • a correlative function relates X to a plurality of glycan characteristics, and the relationship is pleiotropic.
  • X is a value for an element related to a production parameter
  • Yl and Y2 are each values for elements related to the glycan characteristics
  • production parameter X confers different effects (in an embodiment these effects are in different directions, e.g., one is increased and the other is decreased, as opposed to one is changed, e.g., increased or decreased, and the other is unchanged) on at least two glycan characteristics.
  • production parameter X the addition of glucosamine to the media, is correlated with a reduction in galactosylation (e.g., Yl), reduction in fucosyJation (e.g., Y2). an increase in high mannose (e.g., Y3) and an increase in hybrid structures (e.g., Y4).
  • galactosylation e.g., Yl
  • fucosyJation e.g., Y2
  • high mannose e.g., Y3
  • hybrid structures e.g., Y4
  • Some of the methods, systems and databases described herein include or relate Io the selection, or the use, of a glycan property or a production parameter. Some specific preferred embodiments of these methods, systems and databases are provided below. In an embodiment, e.g., in step in or a meino ⁇ nerem, a plurality oi pro ⁇ uction parameters or combination thereof are selected sequentially.
  • the method includes identifying at least 2, 3, 4, or 5, required glycan properties.
  • the method includes selecting a combination of production parameters, which combination con-elates with a required glycan property.
  • a glycan property is a functional property of a glycoprotein, e.g., serum half life, receptor binding affinity, or immunogenicity (in an embodiment it is other than immunogenicity).
  • a glycan property is a glycan characteristic, i.e., a structural property.
  • exemplary glycan characteristics include: the presence, absence or amount of a chemical unit; the presence, absence or amount of a component of a chemical unit (e.g., a sulfate, a phosphate, acetate); heterogeneity or microhetcrogenily at a potential glycosylation site or across the entire protein, e.g., the degree of occupancy of potential glycosylation sites of a protein (e.g., the degree of occupancy of the same potential glycosylation site between two or more of the particular protein backbones in a glycoprotein product and the degree of occupancy of one potential glycosylation site on a protein backbone relative to a diffei ent potential glycosylation site on (he same prolein backbone); the core structure of a branched (e.g.
  • glycan sttucture e.g., a complex (e.g., biantennary, triantennaiy, telranlennary, etc.), a high mannose or a hybrid glycan stmcture
  • the relative position of a chemical unit within a glycan e.g., the presence, absence or amount of a terminal or penultimate chemical unit
  • the relationship between chemical units e.g., linkages between chemical units, isomers and branch points.
  • a required glycan property is selected from the group consisting of: galactosyl ation, fucosylation, high mannose, sialylation, and combinations thereof.
  • At least 1, 2, 3 ,4 or more production parameters are selected sequentially, e.g., each is selected on the basis of a correlation between a single production parameter and a glycan characteristic.
  • the first production parameter is tested for the ability confer a selected glycan property (e.g., a glycan characteristic correlated with the first production parameter by the database) on an amino acid sequence, e.g., the amino acid sequence of the reference glycoprotein product.
  • a selected glycan property e.g., a glycan characteristic correlated with the first production parameter by the database
  • the second production parameter is tested for the ability confer a selected giycan property (e.g., a glycan characteristic correlated with the second production parameter by the database) on an amino acid sequence, e.g., the amino acid sequence of the reference glycoprotein product.
  • a selected giycan property e.g., a glycan characteristic correlated with the second production parameter by the database
  • fn an embodiment 2.
  • 3 ,4 or more production parameters aie selected simultaneously e.g., a combination of production parameters is selected on the basis of a correlation between the combination of production parameters and a glycan property, e.g., a glycan characteristic.
  • a method includes, e.g., in step iii: selecting, in a sequential manner, i) a first production parameter, e.g., a primary production parameter, e.g., a parameter related to a ceil line, a process or bioreactor variable, e.g., batch, fed- batch, or perfusion, a purification process or a formulation, from said database, said database including a correlation between said first production parameter and the conferral of a selected glycan property, e.g., a glycan characteristic, on a protein made in a process which includes said first production parameter; and ii) a second production parameter, e.g., a secondary production parameter, from said database, said database including a correlation between said secondary production parameter and the conferral of a selected glycan property, e.g., a glycan characteristic, on a protein made in a process which includes said second production parameter.
  • a first production parameter e.g.,
  • a method includes selection of 1, 2, 3 or more primary production parameters is interspersed with or followed by selection of 1, 2, 3 or more secondary production parameters.
  • a step in the production method is determined by selecting a production parameter which is correlated with the production of glycoprotein having said preselected glycan property, e.g., a glycan characteristic, from a database.
  • a step in the production method is determined by selecting a production parameter from a database in which each of a plurality of production parameters, or combinations of production parameters, e.g.. at least 2, 5, 10, 2.0, 30, 40 or more parameters or combinations or parameters, is correlated with the production of glycoprotein having said preselected glycan property, e.g., a glycan characteristic, when said parameter or combination of parameters is incorporated into a method for making the glycoprotein product.
  • glycan property e.g., a glycan characteristic
  • a production parameter is selected to confer a required glycan property, e.g., a functional property, which is the same as or similar to the corresponding glycan property of said reference glycoprotein product.
  • the production method is different from a published method for making said reference glycoprotein product,
  • a production parameter is selected which is correlated with the conferral on an amino acid sequence of a glycan characteristic, found on the glycoprotein product or is correlated with a glycan characteristic, which is an intermediate and which is positively correlated with the (eventual) presence on the expressed glycoprotein product of a selected glycan characteristic.
  • a method includes selecting the glycan properties required by the glycoprotein product and then selecting the production parameters, e.g., those selected in d) of a method described herein, to provide the required glycan properties.
  • a method includes selecting a combination of production parameters, which combination correlates with a required glycan property.
  • a step, e.g., step d, of a method herein includes selecting a combination of production parameters, which combination correlates with a required glycan property or characteristic.
  • a production parameter is selected which is correlated with the conferral on an amino acid sequence of a glycan characteristic, found on the reference glycoprotein product or is correlated with a lequired glycan characteristic, which is an intermediate and which is positively correlated with the (eventual) presence on the expressed reference glycoprotein product of a required glycan characteristic.
  • the production method is different from the method used by a party, e.g., a pioneer or other party operating under a NDA, BLA, or other government license, to make a commercially available version of the reference glycoprotein product..
  • the method includes providing information resulting from subjecting the reference glycoprotein product to one or more of the analytical method described herein to provide a glycan property, e.g., a required glycan characteristic.
  • the analytical method is applied to one or a plurality of samples of the reference glycoprotein product, e.g., commercially available reference glycoprotein product.
  • the analytical method is applied to one or a plurality of production lots of the reference glycoprotein product, e.g., commercially available reference glycoprotein product.
  • the reference glycoprotein product is selected from Table II.
  • a database has at least 5, 10, 20, 30, 40, 50, 100, 150. 200, 250 correlations records
  • a database provides: a correlation between a first production parameter (or combination of production parameters) and the conferral of a selected glycan property, e.g., glycan characteristic, on a protein made by a process which includes said first parameter; a correlation between a second production parameter (or combination of production parameters) and the conferral of said selected glycan property, e.g., glycan characteristic, on a protein made by a process which includes said second production parameter; and the database is configured so as to allow choice between the first and second parameter.
  • a database provides: a correlation between the use of the combination of a first and second production parameter (or respective combinations) in a process for making said glycoprotein product on the conferral of said selected glycan characteristic on a protein made by said combination process; and allows the provision of information on the affect of the addition of the first or second production parameter (or respective combinations) on the other production parameter (or respective combination) in terms of addition of a selected glycan property, e.g., glycan characteristic, to a protein.
  • a selected glycan property e.g., glycan characteristic
  • a database is configured so as to allow making a decision of whether to include the first, second, or both production parameters in the production of a glycoprotein product
  • a database is configured to allow appreciation that selection of a first production parameter constrains the selection of a second production parameter and makes the selection of a specific second production parameter more or less favored, because, e.g., of a positive or negative affect on the conferral of a glycan property on the protein if the second parameter is combined with the first.
  • a database includes: i) a correlation between a first and second production parameter and conferral of a first selected glycan characteristic on a protein made by a method which includes said first and second (but not a third) production parameter ii) a correlation between said first and third production parameters and conferral of said first selected glycan characteristic on a protein made by a method which includes said first and third (but not said second) production parameter; and allows comparison of (1) the presence, on a protein made by a method which includes the first and second (but not said third) production parameter, of a first selected glycan characteristic with (2) the presence, on a protein made by a method which includes the first and the third but not the second production parameter, and and further allows a choice between the combination of i and the combination of ii on the grounds of optimization of the presence of said first selected glycan characteristic.
  • a database database includes: correlations each of a plurality of species of a first generic production parameter, e.g., variants of a cell type, e.g., a plurality of CHO cells having different sites of insertion, copy number of insertion, or gycoslyation-related genes with the conferral of a of a glycan property, e.g., a glycan characteristic, on a protein made by a method which includes use of the species; and correlations each of a plurality of species of a second generic production parameter, e.g., fermentation variants, e.g., a plurality of fermentation conditions such as, celJ density, batch process, perfusion process, continuous process wilh the conferral of a of a glycan property, eg a glycan characteristic, on a protein made by a method which includes use of the species.
  • a first generic production parameter e.g., variants of a cell type, e.g.,
  • a database includes: a correlation between the combination of a first species of a first production parameter and a first species of a second production parameter with the conferral of a glycan property, e.g., a glycan characteristic, on a protein made by a method which includes combination; a correlation between the combination of a second species of a first production parameter and the first or a second species of a second production parameter with the conferral of a of a glycan property, e.g., a glycan characteristic, on a protein made by a method which includes combination and allows comparison (and choice) between (1) a combination of first species of a first generic production parameter, e.g., a CHO cell having insertion at a first site, and a first species of a second generic production parameter, e.g., batch-process fermentation, and (2) a combination of a different species of the first generic production parameter, e.g., a CHO cell having insertion at a second site, and
  • a database includes: a correlation between a combination of production parameters and the conferral of a selected glycan property, e.g., a glycan characteristic, on a protein made in a process which includes said combination of production parameters, e.g., wherein the combination of production parameters includes a cell and a culture medium.
  • a selected glycan property e.g., a glycan characteristic
  • a database includes: correlations between a cell cultured under each of a plurality of culture conditions, e.g., said cell cultured in each of a first, second, and third medium, and the conferral of a selected glycan property, e.g., a glycan characteristic, on a protein made in a process which includes said cell cultured under one of said culture conditions, e.g.. wherein a selected cell type, e.g., a CHO cell, can be cultured in a plurality of media and each cell/condition combination coi related to Ihe incidence of the same or a different gjycan property, e.g., a glycan characteristic.
  • a selected cell type e.g., a CHO cell
  • a database includes: correlations between each of a plurality of cells cultured under a plurality of conditions, e.g., a first cell type cultured in a first, second, and third medium, a second cell type cultured in the first, second, and third medium, and a third cell type cultured in the first, second, and third medium, and the conferral of a selected glycan property, e.g., glycan characteristic, on a protein made in a process which includes a ceil/condition combination.
  • a selected glycan property e.g., glycan characteristic
  • a database includes a correlation between each of several selected cell types, e.g., different strains or genotype CHO cells, cultured in a plurality of media (cell/condition combinations) to the incidence of a glycan property, e.g., a glycan characteristic.
  • a glycan property e.g., a glycan characteristic.
  • all elements of said database are not on one computer or storage medium or in one location.
  • the database includes one, two, three, or all of a tunable, nonlinear, pleiotropic, or constrained correlation.
  • Some of the methods, systems, and databases herein include, or relate to additional steps in the production of a reference glycoprotein. Specific embodiments arc provided below.
  • a method futthci includes analyzing an amino acid sequence, c g , that of the i efercnce glycoprotein product, produced under said selected combination of production parameters and comparing it with a preselected criterion, e.g., the presence, absence or level of a preselected glycan property, e.g., glycan characteristic.
  • a preselected criterion e.g., the presence, absence or level of a preselected glycan property, e.g., glycan characteristic.
  • amino acid sequence has a preselected relationship with the criterion, e.g., it meets or fails to meet said criteria, selecting the combination.
  • a method further includes altering the conditions of the selected combinations, e.g., by altering the growth medium, based on whether the glycoprotein exhibits the preselected relationship.
  • a method further includes analyzing the glycoprotein produced under said selected combination and comparing it with a preselected criterion, e.g., having a preselected glycan property, e.g., a glycan structure.
  • glycoprotein has a preselected relationship with said preselected criteria, e.g., it meets or fails to meet said criteria, selecting the combination or glycoprotein produced by the combination for further analysis, e.g., alteration of another parameter, e.g., altering the growth medium.
  • a method further includes testing the glycoprotein product made by the production method to see if it has a preselected chemical, biological, or pharmacokinetic, property.
  • a method further includes comparing a preselected chemical, biological, pharmacokinetic or pharmacodynamic, property of the glycoprotein made by the production process with a preselected standard and if the value for said reference glycoprotein product has a preselected relationship with the preselected standard selecting said reference glycoprotein product.
  • Jn an embodiment a property of said glycoprotein product is compared with a properly of said reference glycoprotein product.
  • a method further includes selecting said reference glycoprotein product foi, classification, acceptance or discarding, releasing or withholding, processing into a drug product, shipping, being moved to a new location, formulation, labeling, packaging, releasing into commerce, for being sold, or offered for sale, or submission if information about the glycoprotein product to a third party for review or approval, depending on whether the preselected criterion is met.
  • the designer or user has searched, e.g., by consulting a government or commercial listing of patents, for the existence of a U.S. patent which covers the reference glycoprotein product, or a method of making or using the reference glycoprotein product.
  • said selection includes selecting a value for a parameter from Table I.
  • a method further including expressing the amino acid sequence of said reference glycoprotein product under said selected condition or conditions and determining if the selected condition or conditions is positively correlated with the presence of the required glycan-conditioned property in the glycoprotein.
  • Embodiments of methods described herein include analyzing a reference glycoprotein product for glycan properties, e.g., glycan characteristics. This analysis can be used as a guide for selecting production parameters or in producing a reference glycoprotein. The analysis can be based on information produced by releasing glycan structures from the reference glycoprotein. In this context release means release from all oi at least some of the amino acid portion of the glycoprotein. By way of example the method can use complete or partial enzymatic digestion to release glycan structures e.g., as single saccharides or larger fragments, from a reference glycoprotein.
  • the released glycan structures can be analyzed, e.g., by providing a glycan pattern and comparing it to a predetermined standard, e.g., a reference glycan pattern from a commercially available reference glycoprotein.
  • a glycan pattern as used herein, is a representation of the presence (or absence) of one or more glycan properties.
  • the glycan pattern provides a quantitative determination of one or more glycan properties. The quantitative determination can be expressed in absolute terms or as function of a standard, e.g., an exogenous standard or as a function of another glycan property in the pattern.
  • the elements of a glycan pattern can, by way of example, be peaks or other fractions (representing one or more species) from a glycan structures derived from a glycoprotein, e.g., from an enzymatic digest.
  • Elements can be described, e.g., in defined structural terms, e.g., by chemical name, or by a functional or physical property, e.g., by molecular weight or by a parameter related to purification or separation, e.g., retention time on a column or other separation device.
  • Methods described herein can be used to make a reference glycoprotein having required glycan properties. This includes the design of a process to make such a reference glycoprotein or its production.
  • the analysis can be used to determine if or confirm that a reference glycoprotein has required glycoprotein properties.
  • methods described herein can be used to monitor production processes and to select production parameters to refine a process which produces product which fails to meet a standard, e.g., does not posses a required glycan property.
  • evaluating the reference glycoprotein product e.g., evaluating physiochemical parameters of the reference glycoprotein product, e.g., measuring mass (e.g., using SDS-PAGE or size exclusion chromatography), pi, carbohydrate content, peptide mapping, protein concentration, biological activity of the reference glycoprotein product: recording the evaluation of one or more parameters of the reference glycoprotein product, e.g., providing a certificate of analysis for the reference glycoprotein product' assessing process contaminants of the reference glycoprotein or its cell culture, eg including but not limited to endotoxin content, sterility testing, mycoplasma content, leachates, host (e.g.
  • CHO cell DNA or protein contaminants
  • recording the process contaminants of the reference glycoprotein or its cell culture measuring the reference glycoprotein cell culture process parameters, including but not limited to the production pH 5 cell viability, production, titer, yield, doubling time, DO, and temperature; recording the cell culture process parameters; assessing and recording the process media components, including raw materials source and lot numbers of materials; measuring the reference glycoprotein purification process parameters, including but not limited to the flow rate, pH, temperature, yield, process contaminants, column volume, or elution volume; recording the purification process parameters; and recording a lot number of a reference glycoprotein batch made from a process described herein.
  • the invention features, a reference glycoprotein product or preparation, e.g., a pharmaceutical preparation, of a reference glycoprotein product, made by a process described herein 01 by a process designed by a method described herein.
  • the reference glycoprotein product has the amino acid sequence of a protein from Table II.
  • reference glycoproteins including reference glycopeptides
  • These include naturally occurring and nonnaturally occurring glycoproteins.
  • Representative reference glycoproteins include: antibodies, e.g., IgG, IgM, human, humanized, grafted, and chimeric antibodies, and fragments thereof; fusion proteins, e.g., fusions including human (or other) antibody domains, e.g., Fc or constant region domains growth factors; hormones..
  • the invention features, a computer program product tangibly embodied in an information carrier and including instructions that when executed by a processor perform a method related to a reference glycoprotein described herein.
  • the database is presented or memorialized on a medium.
  • the medium can be, e.g., a traditional paper medium or other medium which displays printed or written symbols which can be directly (e.g., without the aid of a computer) used by a human being.
  • Such a database can exist as set of printed tables, or a card catalogue, which, e.g., show the relationship of production parameters to glycan characteristics.
  • the database can also be presented or memorialized in electronic or other computer readable form.
  • the database need not be deposited on a single unit of medium, e.g., in a single table or book, or on a single computer or network
  • a database e.g., can combine a traditional medium as described above with a computer -readable medium.
  • the database will contain a collection of lecords, wherein each record relates a production parameter to a glycan property by way of a correlative function.
  • the database can be organized in a number of ways, e.g., as a relational dai abase.
  • the database is in a format that can be searched for specific information or records by techniques specific to each database.
  • a computer database is typically a structured collection of records stored in a computer or computers so that a program can consult it to answer queries. Relational databases together with interfaces for queries and query results are particularly preferred. Mapping the ontology of a relational uaia ⁇ ase anows oun ⁇ ing oi correlations useiui in the methods described herein.
  • databases used in such embodiments will generally also include at least 1, 2, 5, 10, 20 or 50 correlations which are were not present in, or which were not retrieved from, publicly accessible information, e.g., in such embodiments the database may contain at least 1, 2, 5, 10, 20, 50 or more non-linear, plietropic, or constrained correlations.
  • Publicly accessible information can include information from a publicly accessible database such as PubMed.
  • a database described herein contains at least 1, 2, 5, 10, 20 or 50 correlations which are not in publicly accessible information, e.g., are not in published documents. The determination of whether a correlation is publicly accessible, e.g., in a published document or database, is made as of the earliest U.S. filing date of a nonprovisional application from which this patent claims priority.
  • FJG. 1 is a block diagram of computing devices and systems.
  • FIG. 2 is a depiction of a representative chromatogram of glycan patterns from human IgG produced in CHO cells.
  • Human IgG was produced from CHO cells, isolated, glycans released, isolated, and fluorescently tagged, prior to resolving on NP-HPLC.
  • FIG. 3 is a depiction of glycan patterns from human IgG produced in CHO cells under distinct process conditions.
  • Human IgG was produced from CHO cells cultured in the presence of elevated uridine, glucosamine, or both. The IgG was isolated, glycans released, isolated, and fiuorescently tagged, prior to resolving on NP-HPLC.
  • a summation of the normalized data for the IgG produced in the presence of elevated uridine, glucosamine, or both is shown as indicated. Data are representative of duplicate determinants and are expressed as a % of the total peak area.
  • Methods described herein include selecting one or more production parameters to produce a glycoprotein having one or more preselected glycan characteristics and, e.g., thus a required glycan property.
  • a glycan property refers to (1) a functional property conferred or conditioned by a glycan structure on a protein or (2) a structural property (referred to herein as a "glycan characteristic").
  • a required functional activity can be correlated with a glycan characteristic or characteristics and based upon that correlation a decision can be made regarding which production parameter OJ combination of production parameters result in a glycoprotein having the preselected glycan characteristic and, thus, functional activity.
  • Activities thai can be selected include, but are not limited to, serum half life, clearance, stability in vitro (shelf life) or in vivo, binding affinity, tissue distribution and targeting, toxicity, inm ⁇ unogenecity, absorption rate, elimination rate, and bioavailability.
  • Methods described herein include providing a glycan characteristic of one or more glycans of a target glycoprotein and, e.g., using that information to determine and/or select a production parameter or parameters to produce a glycoprotein preparation having the same or a similar glycan characteristic as the target glycoprotein preparation.
  • a "glycan characteristic" as used herein includes the presence, absence or amount of a chemical unit; the presence, absence or amount of a component of a chemical unit (e.g., a sulfate, a phosphate, an acetate, a glycolyl, a propyl, and any other alkyl group modification); heterogeneity or microheterogenity at a potential glycosylation site or across the entire protein, e.g., the degree of occupancy of potential glycosylation sites of a protein (e.g., the degree of occupancy of the same potential glycosylation site between two or more of the particular protein backbones in a glycoprotein product and the degree of occupancy of one potential glycosylation site on a protein backbone relative to a different potential glycosylation site on the same protein backbone); the structure of a branched (e.g., the presence, absence or amount of bisecting GIcNAc or phosphomannose structures) or unbranched glycan; the presence, absence
  • a glycan characteristic can, by way of example, be a peak or other fraction (representing one or more species) from glycan structures derived from a reference glycoprotein, e.g., from an enzymatic digest.
  • a glycan characteristic can be described, e.g., in defined structural terms, e.g., by chemical name, or by a functional oi physical property, e.g., by molecular weight or by a parameter related to purification or separation, e g., retention time of a peak in a column or other separation device.
  • a chemical unit is a chemical compound of carbon, hydrogen and oxygen in which the atoms of the later two elements arc in a ratio of 7: 1
  • a chemical unit can be, e.g., an aldehyde or ketone derivative of a polyhydric alcohol, particularly pantahydric and hexahydric alcohols.
  • Examples of chemical units include monosaccharides such as galactose, fucose, sialic acid, mannose, glucose, N- acetylglucosamine (GIcNAc), N-acetylgalactosamine (GaINAc) and ribose, as well as derivatives and analogs thereof. Derivatives ot various monosaccharides are known.
  • sialic acid encompasses over thirty derivatives with N-acetylneuraminic acid and N-glycolylneuraminic acid forming the core structures.
  • Synthetic ganglioside derivatives are described in U.S. Patent No. 5,567,684; bivalent sialyl-derivatized saccharides are described in U.S. Patent No. 5,559,103.
  • Derivatives and analogues of 2- deoxy-2,3-didehydro-N acetyl neuraminic acid are described in U.S. Patent No. 5,360,817.
  • Examples of sialic acid analogs include those that functionally mimic sialic acid, but are not recognized by endogenous host cell sialylases.
  • Sialyltransferases and other enzymes that are involved in sialic acid metabolism often recognize "unnatural" or "modified” monosaccharide substrates (Kosa et al, Biochem. Biophys. Res. Commun., 190, 914, 1993; Fitz and Wong, J., Org. Chem., 59, 8279, 1994; Shames et al, Glycobiology, 1 , 187, 1991 ; Sparks et al, Tetrahedron, 49, 1, 1993; Lin et al, J. Am. Chem. Soc, 114, 10138, 1992).
  • monosaccharide analogs include, but are not limited to, N-levulinoyl mannosamine (ManLev), Neu5Ac ⁇ -methyl glycoside, NeuSAc ⁇ -methyl glycoside, NeuSAc ⁇ -benzyl glycoside, NeuSAc ⁇ -benzyl glycoside, NeuSAc ⁇ -methylglycoside methyl ester, Neu5Ac ⁇ -methyl ester, 9-O-Acetyl-N- acetylneuraminic acid, 9-O-Laclyl-N-acetylneurammic acid, N-azidoacetylmannosamine and O-acetylated variations thereof, and NeuSAc ⁇ -ethyl thioglycoside.
  • examples of sialic acid analogs and methods that may be used to produce such analogs are taught in U.S. Patent No. 5,759,823 and U.S. Patent No, 5,712,254.
  • Examples of derivatives, or analogs, of other monosaccharides include: amidine, amidrazone and amidoxime derivatives of monosaccharides (U.S. Patent No. 5,663,355), 1 ,3,4,6 tetra-0 acetyl N -acylmannosamine or derivative thereof, analogs or derivatives of sugais oi amino sugais having 5 or 6 carbons in the glycosyl ring, including aldoses, deoxyaldoses and ke ' toses without regard for orientation or configuration of the bonds of the asymmetric carbons, This includes chemical units such as ribose, arabinose, xylose, lyxose, aJlose, altrosc, glucose, idose, galactose, talosc, ribulose.
  • Exemplary monosaccharide analogs and derivatives derived from glucose, N-acetylglucosamine, galactose, N acetylgalactosamine, mannose, fucose and sialic acid as taught, for example in U.S. Patent No. 5,759,823.
  • a glycan characteristic can include the presence, absence or amount of various derivatives or analogs of a chemical unit.
  • the glycan characteristic can be the absence, presence or amount of N-acetyl neuraminic acid.
  • a “glycan structure” as used herein refers to at least two chemical units linked to one another. Any linkage, including covalent and non-covalent linkages, is included.
  • a glycan characteristic can further be a comparison of the presence, absence or amount of a chemical unit, a component of a chemical unit or a glycan structure relative to the presence, absence or amount of another chemical unit, another component of a chemical unit or another glycan structure, respectively.
  • the presence, absence or amount of sialic acid relative to the presence absence or amount of fucose can be determined.
  • the presence, absence or amount of a sialic acid such as N-acetylneuraminic acid can be compared, e.g., to the absence, presence or amount of a sialic acid derivative such as N-glycolylneuraminic acid.
  • a "correlative function" as used herein provides a function which defines the relationship, e.g., in a database, between one or more production parameters and one or more glycan properties.
  • one production parameter is correlated to one glycan property.
  • the correlative function can embody a constant value, e.g., a positive constant value in the case of a positive correlation between the presence or use of a production parameter and the conferral of a glycan property on a glycoprotein.
  • Embodiments also include those in which a plurality of species of the production parameter (e.g., different concentrations of a specified additive) are each assigned a different correlati ve constant each having a different constant value.
  • the database could include correlations between concentration 1 and glycan level I , concentration 2 and glycan level 2, and so on.
  • the correlative function can also be "tunable/ 1 e.g., it (or its output) can vary, e.g., in a linear or non-linear fashion, over a range of input values, according to a function.
  • the correlative function can embody a function which relates X to Y, where X is a value for some clement related to a production parameter and Y is a value for some element related to the glycan property (m some embodiments X is the input and Y is the output, in others Y is the input and X is the output).
  • X in the case where the production parameter is the presence of an additive, e.g., glucosamine, in the culture medium, X be the value for the concentration of glucosamine added to the culture medium.
  • Y can be a value for the amount of fucose added to a protein made in a method which uses glucosamine at concentration X.
  • the correlative function relates a value (e.g., an input value) for concentration of glucosamine to a value (e.g., an output value) for the amount of fucosyl moieties on the glycoprotein.
  • a value e.g., an input value
  • a value e.g., an output value
  • the correlative function indicates a lower amount of fucosylation.
  • Such correlative functions are tunable in the sense that one can change the value of X and see the effect on Y. This allows tuning the production parameter to achieve a desired glycan property.
  • the method also allows varying Y to see the effect on X.
  • Functions relating X and Y can be determined, e.g., by empirical trial.
  • a function can be derived by conducting a series of trials at different glucosamine concentrations, plotting glucosamine concentration against observed levels of fucosylation, and deriving an equation which describes the curve of the plotted values
  • production parameter 1 is tunable for an input setting (or value) Xl and the output or setting (or value) foi Yl will vaiy with the setting (oi value) of XL
  • Production parametci 2 is tunable for an input setting (or value) X?
  • the output or setting (oi value) fbi Y? will vaiy with the setting (o ⁇ value) of X2
  • the uumbei of combinations of Y J and Y2 J S equal to the ⁇ ioduc t of niinibci ol possibilities for Y I and the numbej ol possibilities ibi Y?
  • the constraint may also be imposed because a combination of Yl and Y2 are synthetically or structurally impossible or result in toxicity to the cell culture or to an unwanted property in a glycoprotein.
  • a constraint, e.g., a physical or biological constraint, on solution space can be determined or elucidated, e.g., by empirical experimentation.
  • the constraint of solution space (for X1X2 or Yl Y2) can be achieved in a database or system in a number of ways.
  • the correlative function can be designed to produce a null output or a signal corresponding to an unavailable combination. This need not be absolute but could be expressed in degrees of undesirability.
  • a system could be configured with a filter which identifies prohibited or unavailable combinations and labels them or removes them from output.
  • the filter could be provided with specific unacceptable combinations or a rule-based algorithm for exclusion of unacceptable combinations.
  • Nonlinear, constrained and pleiotropic correlations can be used in the methods, systems and databases described herein.
  • a correlative function generally, is the degree to which one phenomenon oi random variable (e.g., production parametei, glycoprotein function, etc ) is associated with or can be predicted from another.
  • correlation usually refers to the degree fo which a lyer predictive relationship exists between random variables as measured by a correlation coefficient.
  • Correlation may be positive (but nevcj largcj than 1 ). i.e., both variables increase or decrease together; negative or inverse (but nevct smaller than 1 ), i.e., one variable increases when the othci decreases; oi zero, i.e., a change in one variable docs not affecl the other
  • one or more stochastic processes may be used for identifying and selecting glycoprotein characteristics, production parameters, or other phenomenon 01 ran ⁇ om vanaoies ana tncir relationships.
  • correlation functions e.g., autocorrelations, cross-correlations, etc.
  • stochastic processes random variable theories or techniques, or probability theories may be used for identifying and selecting glycoprotein characteristics, production parameters, or other phenomenon 01 ran ⁇ om vanaoies ana tncir relationships.
  • covariance functions generalization functions, distributions functions, probability density functions and other types of mathematical representations may be implemented.
  • the methods include identifying one or more glycan characteristics of protein in a target glycoprotein preparation and using that information to determine production parameters for a glycoprotein preparation having the same or similar glycan characteristic as the target preparation.
  • the presence, absence or amount of a chemical unit or the presence, absence or amount of a component of a chemical unit may be determined as described by Geyer and Geyer (2006) Biochim Biophys. Acta 1764(12): 1853- 1869, or by LC, MS, LC/MS, NMR, exoglycosidase treatment, GC, or combinations of these methods.
  • the heterogeneity or microheterogenity at a potential glycosylation site or across the entire protein can be determined, e.g., using the methods described by Larsen et al. (2005) MoI. Cell. Proteomics (2005) 4(2): 107- 1 19 or Forno et al. (2004 Eur. J. Biochem. (2004) 271 (5): 907-919, or LC, MS, LC/MS, GC, PAGE, enzymatic treatment, or combinations of these methods.
  • the core structure of a branched or unbranched glycan is determined, e.g., as described by Geyer and Geyer (2,006) supra, LC, MS, LC/MS, lectin staining, GC, PAGE, enzymatic cleavage or addition, ELlSA, NMR, monosaccharide analysis, or combinations of these methods on the intact glycoprotein, gl ycopeptides, oi released glycan.
  • Exemplary methods thai can be used to deteiniine the presence, absence or amount of a glycan structure and the relative position of a chemical unit within a glycan are described by Geyer and Geyei (2006) .supra, or can include LC, MS, LC/MS, lectin staining, chromatographic methods, enzymatic cleavage, ELISA quantitation, monosaccharide analysis NMR, oi combinations of methods theiein on the glycoprotein, glycopeptides, or released glycan).
  • the relationship between chemical units e.g., linkages between chemical units, isomers and branch points
  • information about a glycan structure or structures can be integrated to describe the glycan characteristics of a complex glycoprotein product.
  • information obtained e.g., by various methods described herein, can be used in a step by step manner to reduce the initial possibilities of glycan characteristics in the target glycan product.
  • the data obtained regarding various glycan characteristics can be integrated using the methods described in U.S. Patent Publication No: 20050065738.
  • Methods described herein include determining and/or selecting a production parameter or parameters for a glycoprotein preparation such that a desired glycan characteristic or characteristics can be obtained upon production of a glycoprotein preparation.
  • production parameters can be selected prior to the production of a glycoprotein preparation that ensure that a desired glycan characteristic or characteristics are obtained.
  • Production parameters that can be selected include, e.g., the cell or cell line used to produce the glycoprotein preparation, the culture media, culture process or bioreactoi variables (e.g , batch, fed- batch, or perfusion), purification process and foirnulation of a glycoprotein preparation.
  • Primal y pioduction parameters include. 1) the types of host; 7) genetics of the host; 3) media type; 4) fermentation platform; 5) purification steps; and 6) formulation.
  • a secondary production parameter is a production parametei (hat is adjustable oi variable within each of the primary production parameters. Examples include: selection of host subclones based on desired glycan properties; regulation of host gene levels constitutive or inducible; introduction of novel genes or promoter elements; media additives (e.g. partial list on Table IV); physiochemical growth properties (e.g. partial list on Table V); growth vessel type (e.g.
  • bioreactor type T flask
  • cell density cell cycle
  • enrichment of pioduct with a desired glycan type e g. by lectin or antibody mediated enrichment, ion-exchange chromatography, CE, or similar method
  • secondary production parameters clear to someone skilled in the art.
  • Methods described herein can include determining a cell or cell line to provide a glycan characteristic the same or substantially the same as a glycan characteristic of a target glycoprotein preparation, e.g., an FDA approved target glycoprotein preparation.
  • the selected cell can be eukaryotic or prokaryotic, as long as the cell provides or has added to it the enzymes to activate and attach saccharides present in the cell or saccharides present in the cell culture medium or fed to the cells.
  • Examples of eukaryotic cells include yeast, insect, fungi, plant and animal cells, especially mammalian cells.
  • Suitable mammalian cells include any normal mortal or normal or abnormal immortal animal or human cell, including: monkey kidney CVl line transformed by SV40 (COS-7, ATCC CRL 1651); human embiyonic kidney line (293) (Graham et al., J. Gen. Virol. 36;59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese Hamster Ovary (CHO), e.g., DG44, DUKX-Vl 1, GS-CHO (ATCC CCL 61, CRL 9096, CRL 1793 and CRL 9618); mouse Sertoli cells (TM4, Mather, Biol Reprod.
  • monkey kidney cells (CVl ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL 1587), human cervical carcinoma cells (HeLa, ATCC CCL 2); buffalo rat liver cells (BRL 3A, ATCC CRL 1442): human lung cells (W 138, ATCC CCl , IS), human hvei cells (Hep G2, HB 8065), mouse melanoma cells (NSO), mouse mammal y lumoi (MM ' I 060562 A I CC CCL/S I ) FRl cells (iVlath ⁇ et a) Annals N Y Acad Set 383 44 46 (1982)), canine kidney cells (MDCK) ( ⁇ PCC CCL 34 and CR I 625 -i), HEK 293 (A KX 1 CFL L) / 3), W) 38 cells (ATCC CCL IS) (ATCC ⁇ meucan Type Cult ⁇ ic Collection, Rockville, Md
  • U8 / cells Al 27 cells, HL60 cells A 549 celJs. SPl 0 cells. DOX cells, SHS 75 Y cells, Jurkat cells, BCP- I cells, GH3 cells, 9L cells, MC3T3 cells, C3H 10T1/2 cells, NIH -3T3 cells and C6/36 cells.
  • the use of mammalian tissue cell culture to express polypeptides is discussed generally in Winnacker, FROM GENES TO CLONES (VCH Publishers, N. Y., N.Y., 1987).
  • Exemplary plant cells include, for example, Arabidopsis thaliana, rape seed, corn, wheat, rice, tobacco etc.) (Staub, et al. 2000 Nature Biotechnology 1(3): 333-338 and McGarvey, P. B., et al. 1995 Bio-Technology 13(13): 14844487; Bardor, M., et al. 1999 Trends in Plant Science 4(9): 376-380).
  • Exemplary insect cells for example, Spodoptera fnigiperda Sf9, Sf21 , Trichoplusia ni, etc.
  • Exemplary bacteria cells include Escherichia coli.
  • yeasts and fungi such as Pichiapastoris, Pichia methanolica, Hansenula polymorpha, and Saccharomyces cerevisiae can also be selected.
  • a cell can be selected for production of a glycoprotein based, e.g., upon attributes of the cell itself which produce or show a preference for production of the desired glycan characteristic or characteristics. Attributes of the cell that may affect glycosylation include the type of cell, cell state, the cell cycle, the passage number, and the metabolic stress level of the cell.
  • a glycoprotein can be produced in a genetically engineered cell, e.g., a genetically engineered animal, yeast, fungi, plants, or other eukaryotic cell expression system.
  • a cell can be a genetically engineered cell which expresses or over expresses a component, e.g., a protein and/or sugar or sugar precursor, which produces a desired glycan characteristic or characteristics.
  • a cell can also be genetically engineered such that the activity of a component, e.g., a protein and/or sugar or sugar precursor, which produces a desired glycan characteristic or characteristics, is increased.
  • the cell can also be genetically engineered to decrease or reduce production of v a) 1OUS chemical units, components of chemical units or glycan structures
  • i he cell can be genetically engineered to produce a nucleic acid antagonist such as antisense or RNAi which results in decreased expression of component involved with the synthesis of a particular, glycan characteristic, e.g., an enzyme and/or sugar or sugar precursor involved in the production of a glycan characteristic or characteristics.
  • the cell can also be genetically engineered to knock out one or more components, e.g., an enzyme and/or sugar or sugar precursor, involved with the synthesis of a particular glycan characteristic, or to produce a less active or inactive mutant of a component, e.g., an enzyme and/or sugar or sugar precursor, involved with synthesis of a particular glycan characteristic or characteristics.
  • the copy number, site of integration and transcription variables can affect the glycan characteristics of a glycoprotein produced by the cell.
  • Components of a cell that result in a desired glycan characteristic or characteristic can include enzymes involved with the addition or removal of a chemical unit, a component of a chemical unit, or production of a desired glycan structure.
  • the cell can be genetically engineered to expresses, overexpress or otherwise increase the activity of one or more enzymes involved in glycosylation.
  • Other embodiments include a cell genetically engineered to reduce, eliminate or otherwise alter the activity of one or more enzymes involved in glycosylation.
  • Exemplary enzymes include enzymes that cleave polysaccharides such as degrading enzymes, enzymes that add monosaccharides to a glycan structure, enzymes that remove a component of a monosaccharide, enzymes that add a component to a monosaccharide and enzymes that convert a chemical unit into a different chemical unit, e.g., convert galactose to a glucose, etc.
  • degrading enzymes include a galactosidase (e.g., alpha-galactosidase and beta-galactosidase), a sialidase (e.g., an alpha 2—>3 sialidase and an alpha 2-»6 siahdasc), a fucosidase (e.g., an alpha 1 ⁇ >2 fucosidase, a alpha l->3 fucosidase, an alpha l-->4 fucosidase and an alpha 1— >6 fucosidase.
  • a galactosidase e.g., alpha-galactosidase and beta-galactosidase
  • sialidase e.g., an alpha 2—>3 sialidase and an alpha 2-»6 siahdasc
  • a fucosidase e.g., an alpha 1 ⁇ >2 fucosidase, a
  • beta -N-Acetylhexosaminidase from Jack Bean cleave non-reducing terminal beta 1 -»2,3,4,6 linked N -acetyl glucosamine, and N- acetylgalactosamine from oligosaccharides whereas alpha N- ⁇ cetylgalactosammidase (Chicken liver) cleaves terminal alpha 1- >3 linked N-acetylgalactosamine from glycoproteins Othei enzymes such as aspaityl N -acetylglucosammidase cleave at a beta linkage aftet a GlcN ⁇ m the core sequence of IN linked ohgosaccba ⁇ dcs
  • Examples of enzymes which add a monosacchaiide to a glycan prudentctuie include giycosyltraosfeiascs such as a sialyltransferase (e g., alpha 2 ->3 sialyltransfci asc oi alpha 2- ⁇ >6 sialyltransferase), a fiicosyitransferase (e.g., alpha 1 ->2 fucosyltransferse, alpha l —»3 fiicosyitransferase, alpha l—>4 fucosyl transferase or alpha ] — >6 fucosyltransferase), a galactosyltransferase (e.g., alpha 1 -»3 galactosyltransferase, beta 1— >4 galactosyl transferase or beta l-»3 galactosyltransferase), a N
  • Examples of enzymes which add, transfer or remove a component of a monosaccharide include: glucoscaminc N-acetyl transferase, N-acetylneuraminate 7-0 (or 9 -0) acetyl transferase, galactose- 1 -phosphate uridyl transferase, N-acetylneuraminate 9- phosphate phosphotase, N-acetylglucoasamine dcacetylase, L-fucose kinase, galactokinase 1, galactose-1 -phosphate uridyl yl transferase, glucokinase 1 , GDP-mannose 4,6 dehydratase, GDP mannose pyrophosphorylase, N-acetylglucosamine sulfotransferase, galactosyl sulfotransferase, glucosamine-phosphate N-
  • exemplary enzymes that can be affected in a genetically engineered cell include N-acetylglucosamine-6-phosphate 2-epimerase, CMP-Neu5Ac hydroxylase, CMP-Neu5Ac synthetase, cyclic sialic acid hydrolase, fucose-1 -phosphate guanyltransferase, UDP-galaclose-4-epimerase, galactose mutaratose, mannosyltransferase, UDP-N-acetylglucosamine 2-epimerase, glucose phosphate isomerase, GDP-mannosyl transferase, mannose phosphate isomerase, N- acetylneuraminate pyruvate lyase, sialic acid cyclase, UDP-glucuronate decarboxylase, CMP -sialic acid transporter, GDP-fucosyl transportei and UDP galactosyl transporter.
  • a selected cell fos production of a glycoprotein can be a genetically engineered cell that has decreased the rxpiession and/oi acuvity of one OJ nioi e proteins involved in ihc glycosyjation
  • the cell i an be genetically engmceied to knock out one ot mor e pj olcnis involved vvuh the synthesis of ⁇ ⁇ anic ⁇ lai glyean chat acio ⁇ stic OJ to ptoduce a less ac ⁇ vc oi maci ivr mutant oj a ⁇ toto ⁇ ⁇ ⁇ ell
  • a cell can be selected which has been genetically engineered for permanent or regulated inactivation of a gene encoding a protein involved with the synthesis of a particular glycan.
  • genes encoding an enzyme such as the enzymes described herein can be inactivated.
  • Permanent or regulated inactivation of gene expression can be achieved by targeting to a gene locus with a transfected plasmid DNA construct or a synthetic oligonucleotide.
  • the plasmid construct or oligonucleotide can be designed to several forms.
  • a selectable marker gene In the case of insertion of a selectable marker gene into coding sequence, it is possible to create an in-frame fusion of an endogenous exon of the gene with the exon engineered to contain, for example, a selectable marker gene. In this way following successful targeting, the endogenous gene expresses a fusion mRNA (nucleic acid sequence plus selectable marker sequence). Moreover, the fusion mRNA would be unable to produce a functional translation product.
  • the transcription of a gene can be silenced by disrupting the endogenous promoter region or any other regions in the 5' untranslated region (5' UTR) that is needed for transcription.
  • regions include, for example, translational control regions and splice donors of introns.
  • a new regulatory sequence can be inserted upstream of the gene that would render the gene subject to the control of extracellular factors.
  • nucleic acid antagonists are used to decrease expression of a target protein, e.g., a protein involved with the synthesis of a glycan characteristic, e.g., an enzyme such as those discussed above.
  • the nucleic acid antagonist is an siRNA that targets mRNA encoding the target protein.
  • Other types of antagonistic nucleic acids can also be used, e.g., a nucleic acid aptamer, a dsRNA, a ribozyme, a triple-helix former, or an antisense nucleic acid.
  • siRNAs are small double stranded RNAs (dsRNAs) that optionally include overhangs.
  • the duplex region of an siRNA is about 18 to 25 nucleotides in length, e.g., about 19, 20, 21 , 22, 23, or 24 nucleotides in length.
  • the siRNA sequences are exactly complementary to the target mRNA.
  • dsRNAs and siRNAs in particular can be used to silence gene expression in mammalian cells (e.g., human cells). See, e.g., Clemens, J. C. et al. (2000) Proc. Natl. Sci. USA 97, 6499-6503; Billy, E. et al. (2001) Proc. Natl. Sci. USA 98, 14428-14433; Elbashir et al. (2001) Nature.
  • Anti-sense agents can include, for example, from about 8 to about 80 nucleobases (i.e. from about 8 to about 80 nucleotides), e.g., about 8 to about 50 nucleobases, or about 12 to about 30 nucleobases.
  • Anti-sense compounds include ribozymes, external guide sequence (EGS) oligonucleotides (oligozymes), and other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and modulate its expression.
  • Anti-sense compounds can include a stretch of at least eight consecutive nucleobases that are complementary to a sequence in the target gene.
  • An oligonucleotide need not be 100% complementary to its target nucleic acid sequence to be specifically hybridizable.
  • An oligonucleotide is specifically hybridizable when binding of the oligonucleotide to the target interferes with the noi mal function of the target molecule Io cause a loss of utility, and there is a sufficient degiec of complementarity to avoid non specific binding of the oligonucleotide to non -target sequences under conditions in which specific binding is desired.
  • Hybiidization of antisense oligonucleotides with mRNA can interfere with one or more of the normal functions of mRNA
  • the functions of mRNA to be interfered with include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, splicing of the RNA to yield one or more mRNA species, and catalytic activity which may be engaged in by the RNA. Binding of specific protein(s) to the RNA may also be interfered with by antisense oligonucleotide hybridization to the RNA.
  • Exemplary antisense compounds include DNA or RNA sequences that specifically hybridize to the target nucleic acid.
  • the complementary region can extend for between about 8 to about 80 nucleobases.
  • the compounds can include one or more modified nucleobases.
  • Modified nucleobases may include, e.g., 5-substituted pyrimidines such as 5-iodouracil, 5-iodocytosine, and C5-propynyl pyrimidines such as C5- propynylcytosine and C5-propynyluracil.
  • modified nucleobases include N4 -(Cl -C12)alkylaminocytosines and N4,N4 --(Cl -C12)dialkylaminocytosines.
  • Modified nucleobases may also include 7-substituted ⁇ 8 ⁇ aza-7-deazapurines and 7- substituted-7-deazapurines such as, for example, 7-iodo-7-deazapurines, 7-cyano-7- deazapurines, 7-aminocarbonyl-7-deazapurines.
  • 6-amino-7- iodo-7-deazapurines 6-amino-7-cyano-7-deazapurines
  • 6-amino-7-aminocarbonyl-7- deazapurines 2-amino-6-hydroxy-7-iodo-7-deazapurines
  • 2-amino-6-hydroxy-7-cyano-7- deazapurines 2-amino-6-hydroxy-7-aminocarbonyl-7-deazapurines.
  • N6 -(Cl -C12)alkylaminopurines and N6,N6 -(Cl -C12)dialkylaminopurines are also suitable modified nucleobases.
  • other 6-substituted purines including, for example, 6-thioguanine may constitute appropriate modified nucleobases.
  • Other suitable nucleobases include 2 thiouracil, 8 bromoadenine, 8 -bromoguanine, 2-fluoroadenine, and 2-fluoroguanine. Derivatives of any of the aforementioned modified nucleobases are also appropriate.
  • Substituents of any of the preceding compounds may include Cl C30 allcyl, C2 -C30 alkenyl, C2 C30 alkynyl, aryl, aralkyl, heteroaryl, halo, amino, arnido, nitro, thio, sulfonyl, carboxyl, alkoxy aJkylcarbonyl, alkoxycarbonyl, and the like.
  • the cells When cells are to be genetically modified for the purposes of expressing or overexpressing a component, the cells may be modified by conventional genetic engineering methods or by gene activation.
  • a DNA molecule that contains cDNA or genomic DNA sequence encoding desired protein may be contained within an expression construct and transfected into primary, secondary, or immortalized cells by standard methods including, but not limited to, liposome-, polybrene-, or DEAE dextran-mediated transfection, electroporation, calcium phosphate precipitation, microinjection, or velocity driven microprojectiles (see, e.g., U.S. Patent No. 6,048,729).
  • Viruses known to be useful for gene transfer include adenoviruses, adeno associated virus, herpes virus, mumps virus, pollovirus, retroviruses, Sindbis virus, and vaccinia virus such as canary pox vims.
  • the cells may be modified using a gene activation approach, for example, as described in U.S. Patent No. 5,641 ,670; U.S. Patent No. 5,733,761 ; U.S. Patent No. 5,968,502; U.S. Patent No. 6,200,778; U.S. Patent No. 6,214,622; U.S. Patent No. 6,063,630; U.S. Patent No. 6, 187,305; U.S. Patent No. 6,270,989; and U.S. Patent No. 6,242,218.
  • the term "genetically engineered,” as used herein in reference to cells, is meant to encompass cells that express a particular gene product following introduction of a DNA molecule encoding the gene product and/or including regulatory elements that control expression of a coding sequence for the gene product.
  • the DNA molecule may be introduced by gene targeting oi homologous recombination, i.e., introduction of the DNA molecule at a particulai genomic site.
  • the promoter and/or expression system can be selected as, e.g., a secondary production parameter.
  • the promoter can be selected, e.g., based upon the host cell being used.
  • the methods described herein can include determining and/or selecting media components or culture conditions which result in the production of a desired glycan characteristic or characteristics.
  • Culture parameters that can be determined include media components, pH, feeding conditions, osmolarity, carbon dioxide levels, agitation rate, temperature, cell density, seeding density, timing and sparge rate.
  • Methods described herein can include one or more of: increasing or decreasing the speed at which cells are agitated, increasing or decreasing the temperature at which cells are cultures, adding or removing media components, and altering the times at which cultures are started and/or stopped.
  • Sequentially selecting a production parameters or a combination thereof, as used herein, means a fust parameter (or combination) is selected, and then a second parameter (or combination) is selected, e.g., based on a constraint imposed by the choice of the first production parameter.
  • the methods described herein can include determining and/or selecting a media component and/or the concentration of a media component that has a positive correlation to a desired glycan characteristic.
  • a media component can be added in or administered ovet the course of glycoprotein production or when there is a change in media, depending oo culture conditions.
  • Media components include components added directly to culture as well as components that are a byproduct of cell culture.
  • Media components include, e.g., buffer, amino acid content, vitamin content, salt content, mineral content, serum content, carbon source content, lipid content, nucleic acid content, hormone content, trace element content, ammonia content, co-factor content, indicator content, small molecule content, hydrolysatc content and enzyme modulator content.
  • Table III provides examples of various media components that can be selected.
  • Exemplary buffers include Tris, Tricine, HEPES, MOPS, PIPES, TAPS, bicine, BES, TES, cacodylate, MES, acetate, MICP, ADA, ACES, glycin amide and acetamidoglycine.
  • the media can be serum free or can include animal derived products such as, e.g., fetal bovine serum (FBS), fetal calf serum (FCS), horse serum (HS), human serum, animal derived serum substitutes (e.g., Ultroser G, SF and HY; non- fat dry milk; Bovine EX-CYTE), fetuin, bovine serum albumin (BSA), serum albumin, and transferrin.
  • animal derived products such as, e.g., fetal bovine serum (FBS), fetal calf serum (FCS), horse serum (HS), human serum, animal derived serum substitutes (e.g., Ultroser G, SF and HY; non- fat dry milk; Bovine EX-CYTE), fetuin, bovine serum albumin (BSA), serum albumin, and transferrin.
  • lipids such as. e.g., palmitic acid and/or steiic acid, can be included.
  • Lipids components include oils, saturated fatty acids, unsatmated fatty acids, glyccrides, steroids, phospholipids, sphingolipids and lipoproteins
  • Exemplary amino acid that can be included or eliminated ( ⁇ om the media include alanine, arginine, asparaginc, aspartic acid, cysteine, glutamic acid, glutaminc, glycine, histidine, proline, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine and valine.
  • vitamins examples include vitamin A (retinoid), vitamin B l (thiamine), vitamin B2 (riboflavin), vitamin B3 (niacin), vitamin B5 (pantothenic acid), vitamin Bb (pyroxidone), vitamin B / (biotin), vitamin B9 (folic acid), vitamin B 12 (cyanocobalamin), vitamin C (ascoibic acid), vitamin D, vitamin E, and vitamin K.
  • Minerals that can be present in the media or eliminated from the media include bismuth, boron, calcium, chlorine, chromium, cobalt, copper, fluorine, iodine, iron, magnesium, manganese, molybdenum, nickel, phosphorus, potassium, rubidium, selenium, silicon, sodium, strontium, sulfur, tellurium, titanium, tungsten, vanadium, and zinc.
  • Exemplary salts and minerals include CaCl 2 (anhydrous), CuSC> 4 5H 2 O, Fe(NCy •9H 2 O, KCl, 1(NO 3 , ICH 2 PO 4 , MgSO 4 (anhydrous), NaCl, NaH 2 PO 4 H 2 O, NaHCO 3 , Na 2 SE 3 (anhydrous), ZnSO 4 8 VH 2 O; linoleic acid, lipoic acid, D-glucose, hypoxanthine 2Na, phenol red, putrescine 2HCl, sodium pyruvate, thymidine, pyruvic acid, sodium succinate, succinic acid, succinic acid » Na » hexahydrate, glutathione (reduced), para- aminobenzoic acid (PABA), methyl linoleate, bacto peptone G, adenosine, cytidine, guanosine, 2'-deoxyadenosine HCl, 2'-
  • the production parameters can include culturing a cell, e.g., CHO cell, e.g., dhfr deficient CHO cell, in the presence of manganese, e.g., manganese present at a concentration of about 0.1 TM to 50 TM.
  • Decreased fucosylation can also be obtained, e.g., by culturing a cell (e.g., a CFIO cell, e.g., a dhfr deficient CHO cell) at an osmolality of about 350 to 500 mOsm. Osmolality can be adjusted by adding salt to the media or having salt be produced as a byproduct as evaporation occurs during production.
  • Hormones include, for example, somatostatin, growth hormone-releasing factor (GRF), insulin, prolactin, human growth hormone (hGH), somatotropin, estradiol, and piogestero ⁇ e
  • Growth factors include, for example, bone morphogeny protein (BMP), epidejmal growth factor (EGF), basic fibroblast growth factoi (bFGF).
  • bFGF basic fibroblast growth factoi
  • NGF nei ve growth factor
  • BDGF bone derived growth facloi
  • TGF beta 1 transforming growth factoi beta I
  • sufactants that can be present or eliminated fiom the media include Tween 80 and pluronic F 68.
  • Small molecules can include, e.g., butyrate, ammonia, non natural sugars, non natural amino acids, chloroquinc, and betaine.
  • ammonia content can be selected as a production parameter to produce a desired glycan characteristic or characteristics.
  • ammonia can be present in the media in a range from 0.001 to 50 mM.
  • Ammonia can be directly added to the culture and/or can be produced as a by product of glutamine or glucosamine.
  • the production parameters selected can include culturing a ceil (e.g., a CHO ceil, e.g., a dhfr deficient CHO cell) in the presence of ammonia, e.g., ammonia present at a concentration of about 0.01 to 50 mM.
  • a ceil e.g., a CHO ceil, e.g., a dhfr deficient CHO cell
  • ammonia e.g., ammonia present at a concentration of about 0.01 to 50 mM.
  • production parameters selected can include culturing a cell (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) in serum containing media and in the presence of ammonia, e.g., ammonia present at a concentration of about 0.01 to 50 mM.
  • a cell e.g., a CHO cell, e.g., a dhfr deficient CHO cell
  • ammonia e.g., ammonia present at a concentration of about 0.01 to 50 mM.
  • butyrate content Another production parameter is butyrate content.
  • the presence of butyrate in culture media can result in increased galactose levels in the resulting glycoprotein preparation.
  • Butyrate provides increased sialic acid content in the resulting glycoprotein preparation. Therefore, when increased galactosylation and/or sialylation is desired, the cell used to produce the glycoprotein (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) can be cultured in the presence of butyrate.
  • a CHO cell e.g., a dhfr deficient CHO cell
  • butyrate can be present at a concentration of about 0.001 to 10 mM, e.g., a bout 2 mM to 10 mM.
  • production parameters selected can include culturing a cell (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) in seium containing media and in the presence of butyrate, e.g., butyrate present at a concentration of about 2.0 to 10 mM
  • a cell e.g., a CHO cell, e.g., a dhfr deficient CHO cell
  • butyrate e.g., butyrate present at a concentration of about 2.0 to 10 mM
  • Such methods can furthei include selecting one oi more of adherent culture conditions and culture in a T flask.
  • a component such as an enzyme, sugar and/or sugar precursors can be added to media or batch fed to cells to affect glycosynthesis.
  • enzymes and substrates such as sugar precursors can be added to the media or batch fed to cells to produce a desired glycan characteristic or characteristics.
  • the cell can use, e.g., endogenous biochemical processing pathways or can be genetically engineered to convert, or process, the exogenously added monosaccharide into an activated form that serves as a substrate for conjugation to a target glycoprotein in vivo or in vitro.
  • Monosaccharides added to a polysaccharide chain can be incorporated in activated form.
  • Activated monosaccharides include UDP-galactose, UDP-glucose, UDP-N-acetylglucosamine, UDP-N-acetylgactosamine, UDP-xylose, GDP-mannose, GDP-fucose, CMP-N-acetylneuraminic acid and CMP-N- acetylglycolylneuraminic acid.
  • Other monosaccharide precursors that can be added to media or batch fed to cells include: N-acetylglucosamine, glucosamine, glucose, galactose, N-acetylgalactosamine, fructose, fucose, glucose-6-phosphate, mannose-6- phosphate, mannose- 1 -phosphate, fructose-6-phosphate, glucosamine-6-phosphate, N- acetylglucosamine-6-phosphate, N-acetylmannosamine, N-acetylneuraminic acid-6- phosphate, fucose- 1 -phosphate, ATP, GTP, GDP, GMP, CTP, CDP, CMP, UTP, UDP, UMP, undine, adenosine, guanosine, cytodine, lactose, maltose, sucrose, frustose 1,6 biphosphate, 2 phosphoenol pyruvate, 2-ox
  • Activated forms of monosaccharides can be generated by methods known in the art.
  • galactose can be activated to UDP-galactose by several ways including: direct phosphorylation at the 1 -position to give GaI-I -P, which can react with UTP to give UDP-galactose; Gal- 1 P can be converted to UDP-galactose via uridyl transferase exchange reaction with UDP glucose that displaces GIc- 1 -P UDP glucose can be derived from glucose by convert] ng glucose to GIc 6 P by hexokmase and then either to Fra 6 P by phosphoglucose isoincrase oi to GIc I P by phosphogiucomutase Reaction oi GIc i -P with U I P forms UDP glucose GDP fucose can be derived from GDP Man by i eduction with CH ?
  • An activated monosaccharide can be incorporated into a polysaccharide chain using the appropriate glycosyltransferase.
  • a sialic acid CMP ⁇ sialic acid onto a polysaccharide chain
  • a sialyltransferase e.g., alpha 2->3 sialyltransferase or alpha 2->6 sialyltransferase, can be used.
  • a fucosyltransferase e.g., alpha l->2 fucosyltransferse, alpha l->3 fucosyltransferase, alpha 1— >4 fucosyltransferase or alpha 1— >6 fucosyltransferase, can be used.
  • Glycosyltransferases for incorporating galactose and GIcNAc include a galactosyltransferase (e.g., alpha l-»3 galactosyltransferase, beta l->4 galactosyltransferase or beta l->3 galactosyltransferase) and a N- acetylglucosaminyltransferase (e.g., N-acetylglucosaminyltransferase I, II or III), respectively.
  • Glycosyltransferases for incorporating other monsaccharides are known.
  • the glycosyltransferase can be added to the media or batch fed to the cell or the cell can use endogenous processing pathways or be genetically engineered to convert or process the exogenously added monosaccharide. genetically engineered to convert or process the exogenously added monosaccharide.
  • enzymes that can be added to the media or batch fed to the cell are described herein.
  • glucosamine present in the media.
  • Glucosamine can be added io the media or batch fed to the cell or the appropriate enzymes and/or substrates can be added to the media or batch fed to cells such that glucosamine is produced.
  • one or more of N -acctylglucosamine, N-acetylglucosaminc 6 phosphate, N-acetylmannosamine or fmstose can be added to the media or batch fed to the cell for production of glucosamine.
  • Cells cultured in the presence of glucosamine can provide decreased levels of fucosylation and/or galactosylation.
  • a cell when reduced fucosylation and/or galactosylation is desired, a cell (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) can be cultured, e.g., in serum containing media, in the presence of glucosamine.
  • the presence of glucosamine in cell culture can also increase the amount of high mannose structures and hybrid structures in a glycoprotein preparation.
  • a cell when increased levels of high mannose or hybrid glycan structures are desired, a cell (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) can be cultured in the presence of glucosamine.
  • Glucosamine can be present, e.g., at a concentration of about 0.001 to 40 mM.
  • the methods can further include having uridine added to the media or batch fed to a cell, e.g., to reduce the level of high mannose structures associated with a protein produced by the cell.
  • uridine added to the media or batch fed to a cell, e.g., to reduce the level of high mannose structures associated with a protein produced by the cell.
  • the addition of cytidine, UTP, OMP and/or aspartate to media or batch fed to cells can also result in the production of uridine during culture.
  • uridine is present at a concentration of about 0.001 to 10 mM.
  • a media component or components that do not affect a glycosylation characteristic or characteristics include selecting a media component or components that do not affect a glycosylation characteristic or characteristics. For example, the presence of glucosamine and uridine in culture does not significantly alter galactosylation, fucosylation, high mannose production, hybrid production or sialylation of glycoproteins produced by a cell (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) cultured in the presence of this combination.
  • a cell e.g., a CHO cell, e.g., a dhfr deficient CHO cell
  • the presence of mannose in culture does not significantly alter galactosylation, fucosylation, high mannose production, hybrid production or sialylation of glycoproteins produced by a cell (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) cultured in the presence of mannose.
  • a cell e.g., a CHO cell, e.g., a dhfr deficient CHO cell
  • the methods described herein can include selecting a media component such as mannose and/or the combination of glucosamine and uridine such that the glycan characteristic or characteristic is not altered by this component (or components) of the media.
  • mannose can be added to the media, batch fed to the cells or can be produced by a cell exposed to the appropriate substrates such as fructose or mannan.
  • mannose is present at a concentration of about 0.001 to 30 mM,
  • Methods described herein can include selecting culture conditions that are correlated with a desired glycan characteristic or characteristics. Such conditions can include temperature, pH, osmolality, shear force or agitation rate, oxidation, spurge rate, growth vessel, tangential flow, DO, CO 2 , nitrogen, fed batch, redox, cell density and feed strategy. Examples of physiochemical parameters that can be selected are provided in Table IV.
  • the production parameter can be culturing a cell undo acidic, neutral or basic pH conditions. Temperatures can be selected from 10 to 42°C Foi example, a temperature of about 28 to 36 0 C does not significantly alter galactosylatioii, fucosylation, high mannose production, hybrid production or sialylation of glycoproteins produced by a cell (e.g., a CHO cell, e.g., a dhfr deficient CHO cell) cultured at these temperatures. In addition, any method that slows down the growth rate of a cell may also have this effect. Thus, temperatures in this range or methods that slow down growth rate can be selected when it is desirable not to have this parameter of production altering glycosynthesis.
  • Temperatures can be selected from 10 to 42°C Foi example, a temperature of about 28 to 36 0 C does not significantly alter galactosylatioii, fucosylation, high mannose production, hybrid production or sialylation of glycoproteins produced by a cell (e.g.,
  • carbon dioxide levels can be selected which results in a desired glycan characteristic or characteristics.
  • CO 2 levels can be, e.g., about 5%, 6%, 7%, 8%, 9%, 10%, 11 %, 13%, 15%, 17%, 20%, 23% and 25% (and ranges in between).
  • the cell when decreased fucosylation is desired, the cell can be cultured at CO 2 levels of about 11 to 25%, e.g., about 15%. CO 2 levels can be adjusted manually or can be a cell byproduct.
  • a wide array of flasks, bottles, reactors, and controllers allow the production and scale up of cell culture systems.
  • the system can be chosen based, at least in part, upon its correlation with a desired glycan characteristic or characteristics.
  • Cells can be grown, for example, as batch, fed-batch, perfusion, or continuous cultures depending on the correlation with a particular glycan characteristic or characteristics.
  • Production parameters that can be selected include, e.g., addition or removal of media including when (early, middle or late during culture time) and how often media is harvested; increasing or decreasing speed at which cell cultures are agitated; increasing or decreasing temperature at which cells are cultured; adding or removing media such that culture density is adjusted; selecting a time at which cell cultures are started or stopped; and selecting a time at which cell culture parameters arc changed,
  • Such parameters can be selected for any of the batch, fed batch, perfusion and continuous culture conditions eg , described below.
  • Batch Culture Batch culture is carried out by placing the cells to be cultured into a fixed volume of cullure medium and allowing the cells to giow Cell numbers increase, usually exponentially, until a maximum is reached, after which growth becomes arrested and the cells die. This may be due either to exhaustion of a nutrient or accumulation of an inhibitor of growth.
  • To recover product cells are removed from the medium either when the cells have died or al an earlier, predetermined point.
  • Batch culture is characterized in that it proceeds in a fixed volume (since nothing is added after placing the cells in the medium), for a fixed duration (dependent on the length of time the cells survive) with a single harvest and with the cells dying or being discarded at the end of the process.
  • Fed-Batch Culture This is a variation on batch culture and involves the addition of a feed to the batch. Cells are cultured in a medium in a fixed volume. Before the maximum cell concentration is reached, specific supplementary nutrients are added to the culture. The volume of the feed is minimal compared to the volume of the culture.
  • a fed-batch culture involves a batch cell culture to which substrate, in either solid or concentrated liquid form, is added either periodically or continuously during the period of growth.
  • Fed batch culture is also characterized in that it usually proceeds in a substantially fixed volume, for a fixed duration, and with a single harvest either when the cells have died or at an earlier, predetermined point. Fed-batch cultures are described, e.g., in U.S. Pat. Nos. 5,672,502.
  • Perfusion Culture In a perfusion culture, medium is perfused through the reactor at a high rate while cells are retained or recycled back into the reactor by sedimentation, centrifugation or filtration. Up to ten reactor volumes of medium is perfused through the bioreactor in a day. The major function of perfusing such a large volume of medium is primarily to remove the metabolites, mainly lactate, from the culture fluid. Perfusion cultures are described, e.g., in U.S. Pat. No. 6,544,788.
  • Continuous Culture In continuous culture, the cells are initially grown in a fixed volume of medium. To avoid the onset of the decline phase, a pumped feed of fresh medium is initiated before maximum cell concentration is reached. Culture, containing a proportion of the cells, is continuously removed from the vessel to maintain a constant volume. The process removes a product, which can be continuously harvested, and provides a continuous supply of nutrients, which allows the cells to be maintained in an exponentially growing state. Continuous culture is characterized by a continuous increase in culture volume, of product and maintenance of exponentially growing culture. There is little or no death or decline phase. In a continuous culture, cells are continuously fed fresh nutrient medium, while spent medium, cells, and excreted cell product are continuously drawn off. Continuous cultures and bioreactors are described, e.g., in U.S. Pat. Nos. 4,764,471 ; 5,135,853; 6, 156,570.
  • a bioreactor is a device or system that supports a biologically-active environment, e.g., a device or system meant to grow cells or tissues in the context of cell culture (e.g., mammalian, plant, yeast, bacterial cells). This process can either be aerobic or anaerobic.
  • Bioreactors are commonly cylindrical, ranging in size from some liter to cube meters, and are often made of stainless steel. On the basis of mode of operation, a bioreactor may be classified as batch, fed batch or continuous (e.g. continuous stirred-tank reactor model).
  • a bioreactor can be used for large culture volumes (in the range 100-10,000 liters).
  • Suspension cell lines can be kept in suspension, e.g., by a propeller in the base of the chamber vessel (e.g., stir tank or stir flask bioreactors) or by air bubbling through the culture vessel. Both of these methods of agitation can give rise to mechanical stresses.
  • Membranes, porous matrices (e.g., ceramic matrices), and polysaccharide gels can be used to protect cells from shear and/or to obtain high cell densities in bioreactors that are productive for periods of weeks or months.
  • Rotary bioreactors use rolling action to keep cells well perfused, akin to roller bottles.
  • the culture chamber can be separated from the feeder chamber by a semipermeable membrane. This allows media to be changed without disturbing the cells.
  • the rotating action in Synthecon's Rotary Cell Culture System creates a microgravity environment, virtually eliminating shear forces. This allows the cell to shift resources from damage control to establishing relationships with other cells, mimicking the complex three dimensional (3 D) matrices found in vivo.
  • Reactoi vessels come in sizes ranging fiom ) 0
  • bioreactors are as follows.
  • the Heraeus miniPERM bioreactor combines an autoclavable o ⁇ tei nutrient container and a disposable inner bioreactor chamber.
  • the appropriate molecular weight cut-off membrane for a desired product e.g.,. a product described herein
  • Hs small size allows it to fit inside standard incubators. Densities greater than 10 cells per ml and product yields of 160 mg in four weeks are possible.
  • New Brunswick Scientific's CELLIGEN PLUS® is a highly flexible system for culture of virtually all eukaryotic cell lines.
  • Features include a double screen impeller for increased O 2 saturation, interactive four-gas control, internal ring sparger, five programmable pumps, computer interface for system control and data logging, and four- channel recorder output.
  • the unit may be used either as a stir tank or fibrous-bed system.
  • the Wave BioreactorTM (from Wave Biotech, LLC) employs an adjustable-speed rocking platform and electric air pump to gently aerate the culture while keeping shear forces low. Smaller cultures and rocking platforms will fit in a standard incubator. Culture medium and cells only contact a presterile, disposable chamber called a cellbag that is placed on a special rocking platform. The rocking motion of this platfo ⁇ n induces waves in the culture fluid. These waves provide mixing and oxygen transfer, resulting in a perfect environment for cell growth that can easily support over 2O x 10 6 cells/ml. The bioreactor requires no cleaning or sterilization, providing the ultimate ease in operation and protection against cross-contamination.
  • Quark Enterprises provides a full range of bioreactors including its Spingro® flasks for high-density culture. These borosilicate stir flasks range from 100 ml to 36 1 and feature Teflon® spin paddles, side vents for probes and easy sampling, and jacketed models for use with a recirculating water bath. All models are completely aiitoclavable.
  • the ProCulture DynaLifl system facilitates perfusion and reduces shear effects by using an extended paddle, side baffles, and bottom coniours Jt is available in a range of sizes, from 125 ml to 36 1.
  • Anothcj example is Braun Biofcxh's BlOSTA T® Bioieactor
  • Stir tanks can provide cell cultures with increased density. Examples include the following.
  • a disposable Stirred Tank Bioreactor (Xcellerex): a scaleable, disposable stir tank bioreactor (XDRTM) that can operate as a stand-alone skid mounted system or is integrated into a FlexFactoryTM.
  • the XDR incorporates process sensors that monitor and control the culture conditions up to 1 ,000Lor 2,00OL working volume scale.
  • FlexFactoryTM is a complete, turnkey, modular production train for biotherapeutics and vaccines.
  • the single-use, disposable components that are central to the FlexFactoryTM, provide it with great flexibility to accommodate new process changes, including production of multiple products at a single site, and to establish manufacturing capacity rapidly, at dramatically lower costs than traditional fixed-tank, hard-piped facilities.
  • Applikon offers a full line of stir tanks, from 2.3 1 bench systems to 10,000 1 production units. Pumps, probes, controllers, and software are also available for all units. Borosilicate glass vessels are available up to 20 L and can be fitted with lip-sealed or magnetically coupled stirrers. Stainless steel BioCla ⁇ 'eTM vessels are designed for moderate to large-scale production and feature a flush-mounted longitudinal sight glass as well as a choice of lip -seal or magnetic stirrers.
  • the reactor has no moving blades to create shear forces, which some mammalian and hyb ⁇ doma cells ai e ⁇ at hcularly sensitive to Media perfuse from the top while oxygen enters through tbr bottom creating a ncai ideal mixing environment
  • K mible- Korites manufactures (he CYTOl Tt I ⁇ glass airlft bioicactoj with an effective volume of 580 nil It JS easily cleaned and fully aiiioclavable for consistent performance and long life. A glass jacket is standard on all models. Other features include a check valve to prevent backfJow in case of pressure drop, vent, infusion and effluent ports, plus three ports for pH, foam level, and dO2 probes.
  • CYTOSTIR® (also from Kimble Kontes) is a line of double sidearm bioreactors in nine sizes, from 100 ml to 36 i.
  • the large, height adjustable stirring blades are constructed of TEFLON ⁇ to minimize cell adhesion and facilitate cleaning Components ate steam- autoclavable.
  • the medium and inoculum are loaded in the beginning and the cells are allowed to grow. There is no addition/replacement of medium, and the entire cell mass is harvested at the end of incubation period.
  • the characteristic features of such bioreactor systems are as follows: (i) continuous depletion of medium, (ii) accumulation of cellular wastes, (iii) alterations in growth rate and (iv) continuous change in the composition of cells.
  • a spin filter bioreactor can be used as a batch bioreactor by closing the inlet for medium and the outlets for medium/medium plus cells.
  • Batch bioreactors are available, e.g., from Rockland Immunochemicals, Inc.
  • fed-batch reactors feed is added, but effluent (and cells) are not removed.
  • fed-batch reactors can be used to maintain cells under low substrate or nutrient conditions without washout occurring Because cells are not removed during the c ⁇ lturing, fed batch bioi ecators arc well suited for the production of compounds produced during very slow oi zero growth.
  • the feed does not need to contain all the nutrients needed to sustain growth
  • the feed may contain only a nitrogen source or a metabolic precursoi
  • the central shaft of bioreactoi houses a spinning, filter, which enables the removal of used medium, free of cells, through the shaft, (2) A stirrer plate magnetically coupled to the central shaft provides continuous stirring; the spinning filter also stirs the culture; (3) The culture is aerated by a sparger, which allows a
  • This bioreactor provides a highly versatile system for control on medium change rate and on cell density; this becomes possible due to the two routes for medium removal, while only one of them allows the removal of cells.
  • a continuous flow bioreactor can be used to grow cells at a specified cell density in an active growth phase; such cultures may either provide inocula for further culture or may serve as a continuous source of biomass yields.
  • bioreactors are based on cells entrapped either in gels, such as, agarose, agar, chitosan, gelatine, gellan, polyacrylamide and calcium alginate, to produce beads, or in a membrane or metal (stainless steel) screen compartment or cylinder.
  • gels such as, agarose, agar, chitosan, gelatine, gellan, polyacrylamide and calcium alginate
  • the membrane screen cylinder containing cells is kept in a chamber through which the medium is circulated from a recycle chamber.
  • the medium flows parallel to the screen cylinder and diffuses across the screen into the cell mass.
  • the membrane/screen compartment housing the cells may be cylindrical or flat, and medium movement may be so adjusted as to flow across the screen compartment rather than parallel to it.
  • Fresh medium is regularly added to and equivalent volume of used medium is withdrawn from the recycling chamber to maintain its nutrient status
  • Cell immobilization changes the physiology of cells as compared to that of cells in suspension. This technique is useful where the biochemical of interest is excreted by the cells into Ihe medium.
  • Immobilized cell reactors can have the following advantages: (i) no risk of cell wash out, (ii) low contamination risk, (iii) protection of cells from liquid shear, (iv) better control on cell aggregate size, (v) separation of growth phase (in a batch/continuous bioreactor) from production stage (in an immobilized cell bioreaetor), (vi) cellular wastes regularly removed from the system, and (vii) cultures at high cell densities.
  • DMSO dimethyl sulfoxide
  • Such culture systems use two or more bioreactors in a specified sequence, each of which carries out a specific step of the total production process.
  • the simplest situation would involve two bioreactors.
  • both the bioreactors can be of batch type: the first bioreactor provides conditions for rapid cell proliferation and favors biomass production, while the second bioreactor has conditions conducive for biochemical biosynthesis and accumulation.
  • the cell biomass is collected from the first stage bioreactor and is used as inoculum for the second stage reactor.
  • the first reactor may be in continuous mode, while the second may be of batch type.
  • the cell mass from this bioreactor serves as a continuous source of inoculum for the second stage batch type bioreactor, which has conditions necessary for embryo development and maturation (but not for cell proliferation).
  • the use of continuous first stage bioreactor can offer one or more advantages, e.g.: (i) avoids the time, labor and cost needed for cleaning, etc. of a batch reactor between two runs, (ii) eliminates the lag phase of batch cultures, and (iii) provides a more homogeneous and actively growing cell population
  • Bioreactors are available for perfusion c ⁇ ltuies Examples are as follows The CellCube® System from Corning Life Sciences piovidcs a fast, simple, and compact method for the mass culture of attachment dependent colls in a continuously perfused bioreactor.
  • the system is an easily expandable system for growing adherent cells in all levels of biomass, viral, and soluble biomolecule production.
  • the basic system uses disposable CellCube®MLodules with from 8,500cm 2 to 85,000cm 2 cell growth surface using the same control package.
  • eenL-uDeiMViocmies nave polystyrene growth surfaces that are available with either the stand tissue culture surface or the advanced Corning CellBIND ⁇ Surface for improved cell attachment.
  • the CellCube® System is comprised of four pieces of capital equipment — the system controller, oxygenator, circulation and media pumps.
  • the digital controller features in-line monitoring of perfusion, pH, dC> 2 , and temperature.
  • Another type of bioreactor is a centrifugal bioreactor, e.g., Kinetic Biosystems' CBR 2000 centrifugal bioreactor. Designed for industrial production, it can achieve densities up to 10,000 times greater than stirred tank bioreactors. Media are fed in through the axle, then forced to the outside by the rotating action where they enter the reaction chamber. Cells are held in suspension by opposing centrifugal force with perfusion. Waste products are removed through the axle and sampled 10 times per hour. Real-time analysis of growth and production parameters means that any perturbation can be adjusted quickly. The end result can be increased product yield and quality. Each chamber is capable of producing 1x10 ' cells with each rotor holding three chambers.
  • the cell densities obtained can be increased by the addition of micro-earner beads.
  • These small beads are 30 J 005 ⁇ in in diametei and can be made, e.g., of dextrari, cellulose, gelatin, glass oi silica, and can mciease the surface area available foi cell attachment.
  • the range of micro -carriers available means that it is possible to grow mosi cell types in this system
  • Solid beads are the most manageable for biomass harvest, while porous beads arc better suited for secreted oi lysate products.
  • Other matrices hold beads stationary, creating a solid bed through which media are perfused.
  • Microcarrier cultures using suspended macroporous beads are readily scaled up. These systems are distinct from conventional surface microcarrier culture in that the cells are immobilized at high densities inside the matrix pores and are protected from the fluidshear.
  • Another advantage of macroporous beads is that they can be inoculated directly from the bulk medium in the same fashion as conventional microcarriers. Suspended bead immobilization systems can be used in a number of different reactor configurations including suspended beds or stirred tank bioreactors.
  • these systems can be scaled up by increasing the volume of the bioreactor and the number of beads. Suspended macroporous bead technologies are also available. In an attempt to mimic the cell culture environment in mammals, these macroporous beads can be collagen-based (e.g., collagen, gelatin, or collagen-glycosaminoglycan).
  • collagen-based e.g., collagen, gelatin, or collagen-glycosaminoglycan
  • Porous ImmobaSil microbeads produced by Ashby Scientific are available in different shapes and sizes for easy adaptation to your particular culture vessel. They are gas permeable, allowing culture densities to reach 3xl O 6 /ml for maximum product yield.
  • Cytopore I beads are optimized for CHO-type cells, while Cytopore II is for adherent cells requiring higher surface-charge density. Cytoline I beads are suited for resilient cells requiring high circulation rates. T he low-density Cytoline Il carrier is optimized for shear-sensitive cells such as hybridomas needing slower circulation. AP Biotech has designed the Cytopilot fluid-bed system perfusion reactor for use with its Cytoline beads.
  • C YTOSTfR ® bioreactors arc available in nine sizes ranging from 100 mL to 36 liters.
  • the borosilicate glass flasks have two large sidearms with screw cap closures that allow easy sampling.
  • the dome in the center of the flask base prevents microcarriers from accumulating directly under the stirring blades.
  • the large, height adjustable TEFLON® stirring blades are designed to provide maximum stirring efficiency to keep microcarriers in suspension at the slow stirring speeds required for tissue culture.
  • Spinner flasks are either plastic or glass bottles with a central magnetic stirrer shaft and side arms for the addition and removal of cells and medium, and gassing with CO 2 -enriched air. Inoculated spinner flasks are placed on a stirrer and incubated under the culture conditions appropriate for the cell line. Cultures can be stirred, e.g., at 100-250 revolutions per minute.
  • Spinner flask systems designed to handle culture volumes of 1-12 liters are available from Techne, Sigma, and Bellco, e.g. (Prod. Nos. Z380482-3L capacity and Z380474-1L capacity). Another example of spinner culture systems is the MantaRay single-use spinner flask.
  • the SuperSpinner H orn B Brauii Biotech is an entry level stir flask that accommodates 500 and 1000 ml cultures and features a bubble-free aeration/agitation system.
  • the Biostat® series of stir vessels handles cuJiure sizes from 50 mi to 10 1 and include complete ready-to -use systems and systems thai integi ate preexisting components.
  • Adherent or suspension cultures can be grown in T flasks, e.g., T-25, T-76, T-225 flasks.
  • the caps can be plug sealed or vented.
  • the flasks can be plastic or glass.
  • the surface of the flasks can be coated, e.g., with hydrophilic moieties that contain a variety of negatively charged functional groups and/or nitrogen-containing functional groups that support cell attachment, spreading, and differentiation.
  • T flasks are available, e.g., from Nunc, Nalgene, Corning, Greiner, Schott, Pyrex, or Costar.
  • Cells can be grown in culture dishes.
  • the surface of the dishes can be coated, e.g., with hydrophilic moieties that contain a variety of negatively charged functional groups and/or nitrogen-containing functional groups that support cell attachment spreading, and differentiation Dishes arc available, e.g., from BD Biosciences, Coming Greiner, Nunc, Nunclo ⁇ . Pyrex.
  • Suspended cells can be grown, e.g., m bioreactors, dishes, flasks, oi roller bottles, e.g., described herein.
  • the CELLine 1 M 1000 (Integra Bioscience, Chur, Switzerland) device is a membrane-based disposable cell culture system. It is composed of two compartments, a cultivation chamber (20 mL) and a nutrient supply compartment (1000 niL) separated by a semipermeable dialysis membrane (10 kD molecular weight cut-off), which allows small nutrients and growth factors to diffuse to the production chamber. Oxygen supply of the cells and CO 2 diffusion occur through a gas-permeable silicone membrane. Antibodies concentrate in the production medium. This culture system requires a CO 2 incubator. For example, for optimal production levels, the device can be inoculated with 5OxIO 6 cells, and 80% of the production medium and the entire nutrition medium changed twice a week.
  • Such systems include roller bottles (discussed herein).
  • An example of a rotation suspension system is the miniPERM (Vivascience, Hannover, Germany) which is a modified roller bottle two-compartment bioreactor in which the production module (35 mL) is separated from the nutrient module (450 mL) by a semipermeable dialysis membrane. Nutrients and metabolites diffuse through the membrane, and secreted antibodies concentrate in the production module. Oxygenation and CO 2 supply occur through a gas-permeable silicone membrane at the outer side of the production module and through a second silicone membrane extended into the nutrition module.
  • the miniPERM must be placed on a roller base inside a CO 2 incubator. It is possible to place- two roller bases together in a 1 80- L CO2 incubator, each holding a maximum of four bioreactors (i.e., the same amount of space is occupied for I A incubations)
  • roller bottles are cylindrical vessels that revolve slowly (between 5 and 60 revolutions per hour) which bathes the cells that are attached to the inner surface with medium. Roller bottles are available typically with surface areas of 1050cm2 (Prod. No. Z352969), The size of some of the roller bottles presents problems since they are difficult to handle in the confined space of a microbiological safety cabinet. Recenuy iuuu u ⁇ iura wmi o ⁇ p ⁇ ucu nmci bui idocb iwvc become available which has made handling large surface area bottles more manageable, but repeated manipulations and subculture with roller bottles should be avoided if possible.
  • a further problem with roller bottles is with the attachment of cells since as some cells lines do not attach evenly. This is a particular problem with epithelial cells. This may be partially overcome a little by optimizing the speed of rotation, generally by decreasing the speed, during the period of attachment for cells with low attachment efficiency.
  • Roller bottles are used in every conceivable application. A good starting point for small labs with periodic scale-up needs, they are also being used for large-scale industrial production. Because the cultures are seeded and maintained in a manner similar to flasks, typically no additional training is necessary. Small racks fit inside standard incubators, eliminating the need for additional capital expenditures.
  • Roller bottles come in a number of configurations: plastic, glass, pleated, flat, vented, or solid. Glass can be sterilized and reused, whereas different plastics and coatings optimize growth for an assortment of cell types. Pleats increase the effective growth surface, thereby increasing product yield without additional space requirements. Vented caps are used for culture in a CO 2 environment, while solid caps are best for culturing in a warm room or unregulated incubator. Roller bottles are available, e.g.. from Corning.
  • Adherent cells can be grown, e.g., in bioreactors, dishes, flasks, or roller bottles, e.g., described herein
  • the surfaces to which the cells adhere can be treated or coated to promote or support cell attachment, spreading, and/or differentiation, Coatings i nclude lysine (e.g , poly D lysine), polyethyleneimine, collagen, glycoprotein (e.g., fibronectin), gelatin, and so forth.
  • Shaker Flasks can be used to provide greater agitation oi cell cultures to improve oxygen or gas transfer, e.g., as compared to stationary cultures. Shaker flasks are available, e.g., from Pyrex and Nalgene,
  • Perfusion systems allow for continuous feeding of the cell chamber from external media bottle, as described herein.
  • Cells are retained in the cell chamber (e.g., bioreactor, bed perfusion bioreactor, packed bed perfusion bioreactor), Suppliers of perfusion systems include DayMoon Industries, Inc., and New Brunswick Scientific.
  • Hollow fibers are small tube-like filters with a predefined molecular weight cutoff. Large bundles of these fibers can be packed into cylindrical modules, which provide an absolute barrier to cells and antibodies while ensuring perfusion of the liquid. Hollow fiber modules can provide a large surface area in a small volume. The walls of the hollow fibers serve as semipermeable ultrafiltration membranes. Cells are grown in the extracapillary space that surrounds the fibers, and medium is perfused continuously inside the fibers. Metabolites and small nutrients freely perfuse between extra- and intracapillary space according to concentration gradients. Culture monitoring can be performed by lactate measurement.
  • Example of such systems include: Cellex Biosciences' AcuSyst hollow fiber reactor.
  • CELL- PH ARM ⁇ system 100 (CP l OO, Bio Vest, Minneapolis, IVTN) which is a fully integrated hollow fibei cell culture system.
  • the cell culture unit consists of two cartridges: one that seives as a cell compartment and the other, as an oxygenation unit.
  • the system is a freestanding benchto ⁇ system with a disposable fjowpath with yields of up to 400 rag/month
  • the CELL PHARM ⁇ system 2500 (CP2500, BioVest) is a hollow fiber cell culture production system that can produce high-scale quantities of a cell -produced product, e.g., of monoclonal antibodies. Unlike CPlOO. it consists of two fiber cartridges for the cells and hence offers a large cell growth surface (3,25 ml). A third cartridge serves for oxygenation of the medium.
  • the FIBERCELLTM (Fibercell Systems Inc., Frederick. MD) hollow-fiber cell culture sysiem is composed of a culture medium reservoir (250 mL) and a 60-mL fiber cartridge (1.2 m2), both connected to a single microprocessor-controlled pump. It is possible to prolong the media supply cycles by replacing the original medium reservoir with a 5-L flask.
  • the FIBERCELL 1 M bioreactor is used inside a CO 2 incubator. Oxygenation occurs by a gas-permeable tubing.
  • the ACUSYST-XCELL ⁇ is designed for large-scale production of secreted proteins, producing 60 to 200 grams of protein per month. Its ACUSYST MINIMAXTM is a flexible research scale benchtop bioreactor capable of producing up to 1O g of protein per month. For single-use or pilot studies of a few weeks' duration, the economical RESCU- PRIMER TK ' produces up to 200 mg per month with a choice of hollow fiber and ceramic matrices.
  • the Unisyn CELL-PHARM ⁇ MICROMOUSETM is a disposable system with a footprint of 1.5 ft2, fitting inside a standard lab incubator. It is capable of producing up to 250 mg of monoclonal antibodies per month for three months.
  • the TECNOMOUSE® by Integra Biosciences is a modular rack system with five separate cassette chambers. Up to five different cell lines can be cultivated for up to 30 weeks, each producing 200 mg of antibodies per month.
  • the integrated gas supply and online monitoring capabilities help to control culture conditions
  • Cell factories an, used for large scale (e g , industrial scale) cell culture and products of bioinate ⁇ als such as vaccines, monoclonal antibodies, 01 pharmaceuticals.
  • the factories can be used for adherent cells 01 suspension culture.
  • the growth kinetics are similar to laboratory scale culture.
  • Cell factories provide a large amount of growth surface in a small area with easy handling and low risk of contamination,
  • a cell factory is a sealed stack of chambeis with common vent and fill ports.
  • a 40-chamber factory can be used in place of 30 roller bottles. Openings connecting the chambers cause media to fill evenly for consistent growth conditions. Vents can be capped or fitted with bacterial air vents.
  • Cell factories can be molded from e.g., polystyrene. Suppliers of cell factories include Nunc.
  • the surface of the factories can be treated to improve growth or cell attachment conditions, e.g., treated with NUNCLON® ⁇ .
  • NUNCLON® CELL FACTORIESTM are low-profile, disposable, polystyrene, ventable chambers that come in stacks of one, two, 10, and 40. Inoculation, feeding, and harvest are straightforward due to the innovative design of the connected plates.
  • Cell culture bags e.g., single use cell culture bags, can be used for growing mammalian, insect, and plant cells.
  • the bags convenience and flexibility for suspension, perfusion, and microcarrier culture.
  • Suppliers of cell culture bags include DayMoon Industries, Inc. and Dunn Labortechnik GmbH.
  • the bag system also offers a greater flexibility in gas transfer between the bag headspace and the environment, and it is capable of both gas diffusion and continuous gas flush. Gas diffusion through the built-in microporous membrane on the screw-cap provides sufficient gas exchange for most cell culture need If required, pressurized ail or gas under 1.5 psi can be added through one of the l ⁇ er port? and vented out through the juembiarte cap
  • OPTIMA 1 M is a single use cell culture bag that offers convenience, capacity and flexibility for gi owing insect, plant and mammalian cells.
  • OPTIMATM is designed for use on conventional laboratory shakers or rocking platforms Available in two standard bags with working capacities up to 4 1, the OPTIMATM is useful for high volume suspension culture, providing a cost-effective alternative to stirred bioreactors.
  • OPTIMA -MINITM bags are designed to fit most laboratory shakers and rocking platforms, requiring no specialized equipment.
  • Production parameters including purification and formulation can be used to produce a glycoprotein preparation with a desired glycan characteristic or characteristics.
  • affinity based methods can be used to separate glycans and/or glycoproteins based on polarity.
  • Reverse-phase chromatography can be used, e.g., with derivatized sugars.
  • Anion- exchange columns can be used to purify sialylated, phosphorylated, and sulfated sugars.
  • Other methods include high pH anion exchange chromatography and size exclusion chromatography can be used and is based on size separation (Hardy).
  • Affinity based methods can be selected that preferentially bind certain chemical units and glycan structures.
  • Matrices such as m--aminophenylboronic acid, immobilized lectins and antibodies can bind particular glycan structures.
  • M-aminophenylboronic acid matrices can form a temporary covalent bond with any molecule (such as a carbohydrate) that contains a 1 ,2-cis-diol group. The covalent bond can be subsequently disrupted to elute the protein of interest, Lectins are a family of carbohydrate-recognizing proteins that exhibit affinities for various monosaccharides.
  • Lectins bind carbohydrates specifically and J evcrsibJy, Primary monosaccharides recognized by lectins include mannosc/glucose, galactose/N- acetylgalactosamine, N -acetyl glucosamine, fucose. and sialic acid ( QProteome Glycoarray Handbook, Qiagen, September 2005, available at; or similar references, Lectin matrices (e.g., columns or arrays) can consist of a number of lectins with varying and/or overlapping specificities to bind glycoproteins with specific glycan compositions.
  • Some lectins commonly used to purify glycoproteins include concavalin ⁇ (often coupled to Sepharose or agarose) and Wheat Germ.
  • AnIi- glycan antibodies can also be generated by methods known in the art and used in affinity columns to bind and purify glycoproteins.
  • Reference glycoprotein products include commercially available products that have market approval, e.g., that have market approval from a government agency 01 are in the process of undergoing cluneal trials for approval from a government agency. This includes products having regulatory approval or in the process of obtaining regulatory approval from the FDA, European Medicines Agency, or the Japanese or Canadian equivalents.
  • the reference glycoprotein product is a glycoprotein product described below in Table fL.
  • Bevacizumab Avastin interferon beta Ia, recombinant Avonex® coagulation factor IX BeneF ⁇ x ⁇ "
  • Figure 1 is a block diagram of computing devices and systems 400, 450.
  • Computing device 400 is intended to represent various forms of digital computers, such as laptops, desktops, workstations, personal digital assistants, servers, blade servers, mainframes, and other appropriate computers.
  • Computing device 450 is intended to represent various forms of mobile devices, such as personal digital assistants, cellular telephones, smartphones, and other similar computing devices.
  • the components shown here, their connections and relationships, and their functions, are meant to be exemplary only, and are not meant to limit implementations of the inventions described and/or claimed in this document.
  • Computing device 400 includes a processor 402, memory 404, a storage device 406, a high-speed interface 408 connecting to memory 404 and high -speed expansion ports 410, and a low speed interface 412 connecting to low speed bus 414 and storage device 406.
  • Each of the components 402, 404 , 406, 408, 4 10, and 412 are interconnected using various busses, and can be mounted on a common motherboard or in other manners as appropriate.
  • the processor 402 can process instructions for execution within the computing device 400, including instructions stored in the memory 404 or on the storage device 406 to display graphical information for a GUI on an external input/output device, such as display 416 coupled to high speed interface 408.
  • multiple processors and/or multiple buses can be used, as appropriate, along with multiple memories and types of memory.
  • multiple computing devices 400 can be connected, with each device providing portions of the necessary operations (e.g., as a server bank, a group of blade servers, or a multi-processor system).
  • the memory 404 stores information within the computing device 400.
  • the memory 404 is a computer-readable medium.
  • the memory 404 is a volatile memory unit or units.
  • the memory 404 is a non-volatile memory unit or units.
  • the storage device 406 is capable of providing mass storage for the computing device 400.
  • the storage device 406 is a computer-readable medium.
  • the storage device 406 can be a floppy disk device, a hard disk device, an optical disk device, or a tape device, a flash memory or other similar solid state memory device, or an array of devices, including devices in a storage area network or other configurations.
  • a computer program product is tangibly embodied in an information carrier.
  • the computer program product contains instructions that, when executed, perform one or more methods, such as those desc ⁇ bed above.
  • the information carrier is a computer- or machine readable medium, such as the memory 404, the storage device 406, memory on processor 402, oi a propagated signal
  • the high speed controller 408 manages bandwidth-intensive operations for the computing device 400, while the low speed controller 412 manages lower bandwidth intensive opei ations Such allocation of duties is exemplary only In ouf- implevnentation, the high speed controllei 408 is coupled to inemoi y 404. display 416 (c g thiough a giaphics proccssoi or accelerator), and to high speed expansion polls 4 J Q, which can accept va ⁇ ous expansion cards (not shown).
  • low- speed contiollei 412 is coupled to storage device 406 and low speed expansion poit 414
  • the low speed expansion port which can include various communication ports (e.g , USB, Bluetooth, Ethernet, wireless Ethernet) can be coupled to one or more input/output devices, such as a keyboard, a pointing device, a seamier, or a networking device such as a switch or router, e.g., through a n el work adapter.
  • the computing device 400 can be implemented in a number of different forms, as shown in the figure. For example, it can be implemented as a standard server 420, or multiple times in a group of such servers. It can also be implemented as part of a rack server system 424. In addition, it can be implemented in a personal computer such as a laptop computer 422. Alternatively, components from computing device 400 can be combined with other components in a mobile device (not shown), such as device 450. Each of such devices can contain one or more of computing device 400, 450, and an entire system can be made up of multiple computing devices 400, 450 communicating with each other.
  • Computing device 450 includes a processor 452, memory 464, an input/output device such as a display 454, a communication interface 466, and a transceiver 468, among other components.
  • the device 450 can also be provided with a storage device, such as a microdrive or other device, to provide additional storage.
  • a storage device such as a microdrive or other device, to provide additional storage.
  • Each of the components 450, 452, 464, 454, 466, and 468 are interconnected using various buses, and several of the components can be mounted on a common motherboard or in other manners as appropriate.
  • the processor 452 can process instructions for execution within the computing device 450, including instructions stored in the memory 464.
  • the processor can also include separate analog and digital processors.
  • the processor can provide, for example, for coordination of the other components of the device 450, such as control of user interfaces, applications run by device 450, and wireless communication by device 450
  • Processor 45? can communicate with a user through control interface 458 and display interface 456 coupled to a display 454
  • the display 454 can be. foi example, a TF r LCD display or an OLED display, oi oiher appjop ⁇ atc display technology. 1 he display interface 456 can comprise appropriate circuitry for driving the display 454 Io present graphical and other information to a user.
  • the control interface 458 can receive commands from a user and conveit them for submission to the processor 452.
  • an external interface 462 can be provide in communication with processor 452, so as to enable near area communication of device 450 with other devices. External interface 462 can provide, for example, for wired communication (e.g., via a docking procedure) or for wireless communication (e.g., via Bluetooth or other such technologies).
  • the memory 464 stores information within the computing device 450.
  • the memory 464 is a computer-readable medium.
  • the memory 464 is a volatile memory unit or units.
  • the memory 464 is a non-volatile memory unit or units.
  • Expansion memory 474 can also be provided and connected to device 450 through expansion interface 472, which can include, for example, a SIMM card interface. Such expansion memory 474 can provide extra storage space for device 450, or can also store applications or other information for device 450.
  • expansion memory 474 can include instructions to carry out or supplement the processes described above, and can include secure information also.
  • expansion memory 474 can be provide as a security module for device 450, and can be programmed with instructions that permit secure use of device 450.
  • secure applications can be provided via the SIMM cards, along with additional information, such as placing identifying information on the SIMM card in a non-hackable manner.
  • the memory can include for example, flash memory and/or MRAM memory, as discussed below.
  • a computer program product is tangibly embodied in an information carrier.
  • the computer program product contains instructions that, when executed, perform one or more methods, such as those described above.
  • the information carrier is a computer- or machine -readable medium, such as the memory 464, expansion memory 474, memory on processor A 52, or a propagated signal.
  • Device -150 can communicate wirelessly through communication interface 466, which can include digital signal processing circuitry where nccaciy.
  • Communication interface 466 can provide for communications under various modes or protocols, such as GSM voice calls, SMS, EMS, or MMS messaging, CDMA, TDM ⁇ , PDC, WCDMA, CDMA2000, or GPRS, among others.
  • Such communication can occur, for example, through radio frequency transceiver 468.
  • short-range communication can occur, such as using a Bluetooth, WiFi, or other such transceiver (not shown),
  • GPS receiver module 470 can provide additional wireless data to device 450, which can be used as appropriate by applications running on device 450,
  • Device 450 can also communication audibly using audio codec 460, which can receive spoken information from a user and convert it to usable digital information. Audio codex 460 can likewise generate audible sound for a user, such as through a speaker, e.g., in a handset of device 450. Such sound can include sound from voice telephone calls, can include recorded sound (e.g., voice messages, music files, etc.) and can also include sound generated by applications operating on device 450.
  • Audio codec 460 can receive spoken information from a user and convert it to usable digital information. Audio codex 460 can likewise generate audible sound for a user, such as through a speaker, e.g., in a handset of device 450. Such sound can include sound from voice telephone calls, can include recorded sound (e.g., voice messages, music files, etc.) and can also include sound generated by applications operating on device 450.
  • the computing device 450 can be implemented in a number of different forms, as shown in the figure. For example, it can be implemented as a cellular telephone 480. It can also be implemented as part of a smartphone 482, personal digital assistant, or other similar mobile device.
  • the systems and the functional operations described in this specification can be implemented in digital electronic circuitry, or in computer software, firmware, or hardware, including the structural means disclosed in this specification and structural equivalents thereof, or in combinations of them.
  • the techniques can be implemented as one or more computer program products, i.e., one or more computer programs tangibly embodied in an information carrier, e.g., in a machine readable storage device or m a propagated signal, for execution by, oi to control the operation of, data processing apparatus, e.g., a programmable processor, a computer, or multiple computers
  • a computer program also known as a program, software, software application, or code
  • processors suitable for the execution of a computer program include, by way of example, both general and special purpose microprocessors, and any one or more processors of any kind of digital computer.
  • the processor will receive instructions and data from a read only memory or a random access memory or both.
  • the essential elements of a computer are a processor for executing instructions and one or more memory devices for storing instructions and data.
  • a computer will also include, or be operatively coupled to receive data from or transfer data to, or both, one or more mass storage devices for storing data, e.g., magnetic, magneto optical disks, or optical disks.
  • Information carriers suitable for embodying computer program instructions and data include all forms of non volatile memory, including by way of example semiconductor memory devices, e.g., EPROM, EEPROM, and flash memory devices; magnetic disks, e.g , internal hard disks or removable disks; magneto optical disks; and CD ROM and DVD ROM disks
  • semiconductor memory devices e.g., EPROM, EEPROM, and flash memory devices
  • magnetic disks e.g , internal hard disks or removable disks
  • magneto optical disks e.g., CD ROM and DVD ROM disks
  • the processor and the memory can be supplemented by, or incorporated in, special purpose logic circuitry.
  • Fo provide for intei action with a user, aspects of the described techniques can be implemented on a computer having a display device, e.g , a CRT (cathode ray tube) 01 I ,CD (liquid ciystal display) monitoi foi displaying information to the usei and a kcyboatd and a pointing K P ⁇ g d mouse oi a trackball, by which Hie ⁇ sci can ⁇ iovidc input to the compntci Otltci kinds of devices can be used to ⁇ tovide lot jntei action with a oset as w ell foi example feedback pj ovided to the us ⁇ can be any form of sensoi y feedback c g visual feedback a ⁇ ditoi y feedback, oi tactile feedback and input fioni the usei can be ieccived in any fo ⁇ n including acoustic, speech, OJ tactile input.
  • the techniques can be implemented m a computing system that includes a back- end component, e.g., as a data seivci, OJ that includes a middleware component, e.g., an application server, or that includes a front-end component, e.g., a client computer having a graphical user interface or a Web browser through which a user can interact with an implementation, or any combination of such back-end, middleware, or front-end components.
  • the components of the system can be interconnected by any form or medium of digital data communication, e.g., a communication network. Examples of communication networks include a local area network ("LAN”) and a wide area network (“WAN”), e.g., the Internet.
  • LAN local area network
  • WAN wide area network
  • the computing system can include clients and servers.
  • a client and server are generally remote from each other and typically interact through a communication network.
  • the relationship of client and server arises by virtue of computer programs running on the respective computers and having a client-server relationship to each other.
  • post-translational modifications e.g., , are addresses in the same way as glycosylation.
  • post- translational modification examples include: proteolysis, racemization, N-O acyl shift, multimerization, aggregation, sugar modification, biotinylation, ncddylation, acylation, formylation, myristoylation, pyroglutamate formation, methylation, glycation, carbamylatio ⁇ , amidation, glycosyl phosphatidylinositol addition, O-metbylaUon, glypiatjon, ubiquitination, SUMOylation, methylation, acetylalion, acctylation, hydroxylation, ubiquitination, SUMOylation, desmosine formation, deamination and oxidation to aldehyde, O -glycosylation, im
  • Example I Correlations Between Various Production Parameters and Glycan Properties for Production in CHO cells
  • Glucosamine content was evaluated at 0, 3mM, 10 niM and 2OmM glucosamine content.
  • Example H Correlation of non linear additive relationships between production parameters and glycan properties for production in CHO cells.
  • PNGase-F Peptide:N-glycosidase F
  • PNGase F can hydrolyze nearly all types of N-glyca ⁇ chains from glycopeptides and/oi glycoproteins.
  • the resulting glycan sample was purified using activated graphitized carbon solid phase extraction cartridges, and labeled on theii reducing termini with a fluorescent tag, 2-benzamide, The labeled glycans were subsequently resolved by NP HPLC using an amide column and their patterns determined. Sec Fig. 2, Glycan profiles were normalized foi protein level and finally expressed as a percentage of the total glycan peak area.
  • the pattern of giycans on the antibody produced in the presence of elevated glucosamine, uridine, or both uridine and glucosamine are illustrated in the Fig. 3.
  • the glycan profile pattern observed on IgG produced in the presence of both uridine and glucosamine is not predicted from the profiles observed from an antibody produced in the presence of uridine or glucosamine individually.
  • the relationship between the production parameters and glycan characteristics represented by peaks A, B, D, M, T and V are nonlinear.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Evolutionary Biology (AREA)
  • Theoretical Computer Science (AREA)
  • Bioethics (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Databases & Information Systems (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Medical Informatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

L'invention concerne des procédés de fabrication de produits de glycoprotéines de référence.
PCT/US2008/060365 2007-04-16 2008-04-15 Produits de glycoprotéines de référence et procédés associés WO2008128230A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US91210007P 2007-04-16 2007-04-16
US60/912,100 2007-04-16

Publications (1)

Publication Number Publication Date
WO2008128230A1 true WO2008128230A1 (fr) 2008-10-23

Family

ID=39570174

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/060365 WO2008128230A1 (fr) 2007-04-16 2008-04-15 Produits de glycoprotéines de référence et procédés associés

Country Status (1)

Country Link
WO (1) WO2008128230A1 (fr)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130123126A1 (en) * 2010-04-07 2013-05-16 Momenta Pharmaceuticals, Inc. Selection and use of host cells for production of glycoproteins
WO2014150655A1 (fr) 2013-03-14 2014-09-25 Momenta Pharmaceuticals, Inc. Procédés de culture de cellules
WO2014159499A1 (fr) 2013-03-14 2014-10-02 Momenta Pharmaceuticals, Inc. Procédés de culture cellulaire
WO2014179601A2 (fr) 2013-05-02 2014-11-06 Momenta Pharmaceuticals, Inc. Glycoprotéines sialylées
WO2014186310A1 (fr) 2013-05-13 2014-11-20 Momenta Pharmaceuticals, Inc. Méthodes de traitement de la neurodégénérescence
WO2015073884A2 (fr) 2013-11-15 2015-05-21 Abbvie, Inc. Compositions de protéines de liaison génétiquement glycomodifiées
US9051593B2 (en) 2009-12-21 2015-06-09 Trustees Of Dartmouth College Recombinant prokaryotes and use thereof for production of O-glycosylated proteins
US9170249B2 (en) 2011-03-12 2015-10-27 Momenta Pharmaceuticals, Inc. N-acetylhexosamine-containing N-glycans in glycoprotein products
EP2856159A4 (fr) * 2012-06-01 2016-04-13 Momenta Pharmaceuticals Inc Méthodes associées au denosumab
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9921210B2 (en) 2010-04-07 2018-03-20 Momenta Pharmaceuticals, Inc. High mannose glycans
US11661456B2 (en) 2013-10-16 2023-05-30 Momenta Pharmaceuticals, Inc. Sialylated glycoproteins
EP4310503A2 (fr) 2015-12-30 2024-01-24 Momenta Pharmaceuticals, Inc. Méthodes associées à des produits biologiques

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
COOPER CATHERINE A ET AL: "GlycoSuiteDB: A curated relational database of glycoprotein glycan structures and their biological sources. 2003 update", NUCLEIC ACIDS RESEARCH,, vol. 31, no. 1, 1 January 2003 (2003-01-01), pages 511 - 513, XP002486792 *

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9051593B2 (en) 2009-12-21 2015-06-09 Trustees Of Dartmouth College Recombinant prokaryotes and use thereof for production of O-glycosylated proteins
US20130123126A1 (en) * 2010-04-07 2013-05-16 Momenta Pharmaceuticals, Inc. Selection and use of host cells for production of glycoproteins
US9921210B2 (en) 2010-04-07 2018-03-20 Momenta Pharmaceuticals, Inc. High mannose glycans
US9890410B2 (en) 2011-03-12 2018-02-13 Momenta Pharmaceuticals, Inc. N-acetylhexosamine-containing N-glycans in glycoprotein products
US9170249B2 (en) 2011-03-12 2015-10-27 Momenta Pharmaceuticals, Inc. N-acetylhexosamine-containing N-glycans in glycoprotein products
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
EP2856159A4 (fr) * 2012-06-01 2016-04-13 Momenta Pharmaceuticals Inc Méthodes associées au denosumab
US9695244B2 (en) 2012-06-01 2017-07-04 Momenta Pharmaceuticals, Inc. Methods related to denosumab
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
WO2014159499A1 (fr) 2013-03-14 2014-10-02 Momenta Pharmaceuticals, Inc. Procédés de culture cellulaire
WO2014150655A1 (fr) 2013-03-14 2014-09-25 Momenta Pharmaceuticals, Inc. Procédés de culture de cellules
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
WO2014179601A2 (fr) 2013-05-02 2014-11-06 Momenta Pharmaceuticals, Inc. Glycoprotéines sialylées
EP3719122A1 (fr) 2013-05-02 2020-10-07 Momenta Pharmaceuticals, Inc. Glycoprotéines sialylées
WO2014186310A1 (fr) 2013-05-13 2014-11-20 Momenta Pharmaceuticals, Inc. Méthodes de traitement de la neurodégénérescence
US11352415B2 (en) 2013-05-13 2022-06-07 Momenta Pharmaceuticals, Inc. Methods for the treatment of neurodegeneration
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US11661456B2 (en) 2013-10-16 2023-05-30 Momenta Pharmaceuticals, Inc. Sialylated glycoproteins
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
WO2015073884A2 (fr) 2013-11-15 2015-05-21 Abbvie, Inc. Compositions de protéines de liaison génétiquement glycomodifiées
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
EP4310503A2 (fr) 2015-12-30 2024-01-24 Momenta Pharmaceuticals, Inc. Méthodes associées à des produits biologiques

Similar Documents

Publication Publication Date Title
EP2528002B1 (fr) Produits de glycoprotéine définie et procédés associés
WO2008128230A1 (fr) Produits de glycoprotéines de référence et procédés associés
Tejwani et al. Glycoengineering in CHO cells: advances in systems biology
Karst et al. Modulation and modeling of monoclonal antibody N‐linked glycosylation in mammalian cell perfusion reactors
Burleigh et al. Synergizing metabolic flux analysis and nucleotide sugar metabolism to understand the control of glycosylation of recombinant protein in CHO cells
Ramakrishnan et al. Crystal structure of β1, 4-galactosyltransferase complex with UDP-Gal reveals an oligosaccharide acceptor binding site
Gupta et al. Genomics and proteomics in process development: opportunities and challenges
US20120329709A1 (en) Production of glycoproteins
Jiang et al. pH excursions impact CHO cell culture performance and antibody N-linked glycosylation
Farrell et al. Application of multi-omics techniques for bioprocess design and optimization in Chinese hamster ovary cells
Bos et al. Optimization and automation of an end‐to‐end high throughput microscale transient protein production process
Agirre Strategies for carbohydrate model building, refinement and validation
AU2011237445B2 (en) Selection and use of host cells for production of glycoproteins
AU2010256455A1 (en) Methods of modulating fucosylation of glycoproteins
WO2011069056A2 (fr) Fucosylation antennaire dans des glycoprotéines de cellules cho
Liu et al. Galactose supplementation enhance sialylation of recombinant Fc-fusion protein in CHO cell: an insight into the role of galactosylation in sialylation
Zhang et al. Combined effects of glycosylation precursors and lactate on the glycoprofile of IgG produced by CHO cells
Strnad et al. Optimization of cultivation conditions in spin tubes for Chinese hamster ovary cells producing erythropoietin and the comparison of glycosylation patterns in different cultivation vessels
Prestegard A perspective on the PDB’s impact on the field of glycobiology
JP6096123B2 (ja) ヒト骨髄性白血病細胞およびそれに由来する細胞を培養するための方法
Aghamohseni et al. A semi-empirical glycosylation model of a camelid monoclonal antibody under hypothermia cell culture conditions
Puri et al. Understanding glycomechanics using mathematical modeling: a review of current approaches to simulate cellular glycosylation reaction networks
Mao et al. Progress toward rapid, at-line N-glycosylation detection and control for recombinant protein expression
Monteil et al. A comparison of orbitally‐shaken and stirred‐tank bioreactors: pH modulation and bioreactor type affect CHO cell growth and protein glycosylation
Syddall et al. Directed evolution of biomass intensive CHO cells by adaptation to sub-physiological temperature

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08745880

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08745880

Country of ref document: EP

Kind code of ref document: A1