Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MAMMALIAN CELL CULTURE
Document Type and Number:
WIPO Patent Application WO/2023/148775
Kind Code:
A1
Abstract:
The present invention relates to method culturing of mammalian cells expressing recombinant proteins. The cell culture method of the present invention implements the N-1 seed stage and the N stage production phase in the same bioreactor. In particular the cell culture method of the present invention provides for consistency in product quailty and productivity upon scale up of the early stage cell culture methods.

Inventors:
DHUNDI RITESH M (IN)
TRIPATHI DIWAKAR MANI (IN)
CHATTERJEE SUDIPTA (IN)
PARUPUDI L C PAVANI (IN)
Application Number:
PCT/IN2023/050112
Publication Date:
August 10, 2023
Filing Date:
February 05, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DR REDDY’S LABORATORIES LTD (IN)
International Classes:
C12N5/00; C12P21/00
Other References:
JYOTHILEKSHMI I., JAYAPRAKASH N. S.: "Trends in Monoclonal Antibody Production Using Various Bioreactor Syst", JOURNAL OF MICROBIOLOGY AND BIOTECHNOLOGY, HAN'GUG MI'SAENGMUL SAENGMYEONG GONG HAGHOE,KOREAN SOCIETY FOR MICROBIOLOGY AND BIOTECHNOLOGY, KOREA, vol. 31, no. 3, 28 March 2021 (2021-03-28), Korea, pages 349 - 357, XP093084332, ISSN: 1017-7825, DOI: 10.4014/jmb.1911.11066
ST. AMAND ET AL.: "Identifying a robust design space for glycosylation during monoclonal antibody production", BIOTECHNOLOGY PROGRESS, vol. 32, no. 5, 2016, pages 1149 - 1162, XP072296891, DOI: 10.1002/btpr.2316
BEN MADSEN, JEFF HURD, CHRIS BRAU AND NEPHI JONES: "Simplify Upstream Process Development and Scale-Up: Single-Use 5:1 Turndown-Ratio Bioreactor Technology", BIOPROCESS INTERNATIONAL, 13 December 2017 (2017-12-13), pages 1 - 9, XP009548085
XU JIANLIN, XU XUANKUO, HUANG CHAO, ANGELO JAMES, OLIVEIRA CHRISTOPHER L., XU MENGMENG, XU XIA, TEMEL DENIZ, DING JULIA, GHOSE SAN: "Biomanufacturing evolution from conventional to intensified processes for productivity improvement: a case study", MABS, LANDES BIOSCIENCE, US, vol. 12, no. 1, 1 January 2020 (2020-01-01), US , XP093084334, ISSN: 1942-0862, DOI: 10.1080/19420862.2020.1770669
Download PDF:
Claims:
CLAIMS We claim: 1. A cell culture method for production of a recombinant biotherapeutic composition having a target glycosylation profile, the said process comprising, (a) providing/culturing mammalian cells expressing the said recombinant biotherapeutic, (b) performing the N-1 seed stage and production stage in the same bioreactor, (c) a 5:2 turndown ratio is used for seed stage to production stage expansion along with calculated O2 volumetric mass transfer coefficient (kLaO2) from Van’t Riet equation (d) maintaining the CO2 volumetric mass transfer coefficient (kLaCO2) of about 0.10 h-1 to about 0.27 h-1, and the O2 volumetric mass transfer coefficient (kLaO2) of about 8.0 h-1 to about 10.5 h-1 (e) recovering the said recombinant biotherapeutic composition from the culture, wherein, the target glycosylation profile is characterised in terms of glycan variants including G0F glycans, total afucosylated glycans, galactosylated glycans. 2. A cell culture method of claim 1, wherein the production stage includes a temperature shift. 3. A cell culture method of claim 1 or 2, wherein the production phase is operated in perfusion mode. 4. A cell culture method of claims 2-3, wherein the temperature difference of the cell culture before and after the temperature shift from about 7 °C to about 2°C.

5. A cell culture method of claims 2-4, wherein the cell culture temperature before the temperature shift is about 37°C and the cell culture temperature after the temperature shift is selected from about 30°C, about 31°C, about 32°C, about 33°C, about 34°C, about 35°C. 6. A cell culture method of claims 2-5, wherein the recombinant biotherapeutic composition so produced comprises of about 51% - about 53% G0-F, about 3.0% – about 3.7 %TAF, about 46% - about 48% galactosylated glycans. 7. A cell culture method of claims 2-6, wherein the recombinant biotherapeutic is an IgG1 monoclonal antibody.

Description:
MAMMALIAN CELL CULTURE FIELD OF INVENTION The present invention relates to method culturing of mammalian cells expressing recombinant proteins. In particular the cell culture method provided ensures consistency in product quality during scale up operations. BACKGROUND OF INVENTION Recombinant biotherapeutics like monoclonal antibodies have evolved into a major therapeutic category in the last quarter century. They are invariably produced using genetically modified cells, amongst which mammalian cells are predominantly favoured. Like any other pharmaceutical drugs, development of bioprocess methods are undertaken at smaller scales in early stages which helps save time and resources. However scaling up of those early stage bioprocess methods to manufacturing scales poses additional challenges due to the inherent complexities of the recombinant biotherapeutics and the cell culture systems used for their production. Recombinant biotherapeutics are heterogeneous molecules and the ones produced in mammalian cell cultures undergo post-translational modifications. These modifications are more often critical to the quality and efficacy of those molecules. Therefore the aim of bioprocess methods development is to ensure favourable productivity as well as product characteristics. It is well recognised that various aspects of mammalian cell culture parameters provide levers to control the product quality and productivity. It is imperative that biotherapeutics production process is well characterised so as to identify key process parameters that would affect the product quality and quantity. The present application provides experimentally characterised cell culture parameters which ensures consistent product quality and productivity upon scale-up of early scale methods to manufacturing scales. SUMMARY OF THE INVENTION Mammalian cell culture methods for production of biotherapeutics invaraibly involves early scale development in smaller bioreactors which provides eficiency in terms of resources and time. Several process parameters can be screened in parallel to arrive at a process that provides favourable productivity and product profile. However scaling up of those early stage process to manufacturing scale poses is a challenge as it is known that various cell culture parameters can affect the product quality as well and the quantity. Herein a scale up strategy based on volumetric mass transfer coefficients of O 2 and CO 2 , which ensures productivity as well as product quality. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1: Contour plots of agitation vs aeration for k L aO 2 values at (a) 600L, (b) 800L and (c) 1000L scales with 20µm sparger. Figure 2: Variation of k L aO 2 with incremental air flow rate and agitation with 20µm sparger at 1000L scale. Figure 3: kLaCO 2 at 95, 110 and 125 RPM with Δ change in pH units over air flow rates through DHS sparger at 1000L scale. Figure 4: Viable cell densities cell cultures (a) N-1 seed bioreactor stage (b) production stage Figure 5: Viability of cell culture (a) N-1 seed bioreactor stage, (b) production stage Figure 6: Air flow rate utilized for day wise stripping of excessive pCO 2 in the production batches DETAILED DESCRIPTION Definitions The term “about” refers to a range of values that are similar to the stated reference value to a range of values that fall within 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1 percent or less of the stated reference value. The term “afucosylated glycans” refers to glycans wherein fucose is not linked to the non-reducing end of N-acetlyglucosamine and includes M3NAG, G0, G1A, G1B and G2. Further, the term “total afucosylated glycans” refers to glycans wherein fucose is not linked to the non-reducing end of N- acetlyglucosamine and includes mannose glycans. Without limitation, examples of total afucosylated glycans include G0, G1A, G1B, G2, M3- M9NAG, M3-M9 (see Table 1). As used herein, the term biotherapeutics and recombinant biotherapeutics has been used interchangeably. It refers to biologic products that can be produced in various expression systems in-vitro using recombinant DNA technology. The exemplary biotherapeutics include but not limited to cytokines, growth factors, hormones, interferons and antibodies. The terms “cell culture medium”, “culture medium”, "media", "medium", as used herein refer to a solution containing nutrients which are required to support the growth of the cells in cell culture. A "basal medium" refers to a cell culture medium that contains all of the essential ingredients useful for cell metabolism. This includes for instance amino acids, lipids, carbon source, vitamins and mineral salts. DMEM (Dulbeccos' Modified Eagles Medium), RPMI (Roswell Park Memorial Institute Medium) or medium F12 (Ham's F12 medium) are examples of commercially available basal media. Alternatively, said basal medium can be a proprietary medium fully developed in-house, in which all of the components can be described in terms of the chemical formulas and are present in known concentrations. The term “cell culture process” as used herein refers to a process of culturing a population of cells that are capable of producing recombinant protein of interest or antibody. It is well known that proteins expressed in eukaryotic cells undergo post- translation modifications and most notable of them is glycosylation which involves covalent addition of sugar residues to the polypeptide chain forming the protein. The term "glycan" refers to the sugar residue which can be a monosaccharide or polysaccharide moiety. The term "glycoprotein" refers to protein or polypeptide having at least one glycan moiety. Thus, any polypeptide attached to a saccharide moiety is termed as glycoprotein. Recombinant biotherapeutics including monoclonal antibodies are examples of glycoproteins. The term "glycoform" or "glycovariant" used interchangeably herein refers to various oligosaccharide entities or moieties linked to the glycoprotein. Examples of such glycans and their structures are listed in Table 1. However, Table 1 may not be considered as limitations of this invention. The term “G0F glycans” refers to glycan moieties with fucose linked to the non- reducing end of N-acetlyglucosamine, and does not contain any terminal galactose residues (see Table 1). The term “galactosylated glycans” refer to glycan moieties containing terminal galactose residues such as G1A, G1B, G1AF, G1BF, G2, G2F and G2SF (see Table 1). The term “high mannose glycans” refers to glycan moieties containing unsubstituted terminal mannose sugars (see Table 1). High mannose glycans contain more than 4 mannose residues attached to the GlcNAc2) core. Table 1: Representative table of various glycans. The terms "perfusion" or “perfusion process” refers to a cell culture process wherein high cell densities and viabilities are achieved and maintained for extended period of time by use of cell retention device/system together with continuous media exchange. Fresh growth medium (“n+1” culture medium) is added or one or more times during cell culture and the spent culture medium (“n” culture medium, which was already present in the bioreactor), which may contain the recombinant product expressed by the cells, is harvested from the bioreactor continually or one or more times during cell culture. The term “concentrated fed batch” (CFB) refers to a perfusion cell culture wherein ultrafiltration membranes are used to retain cells as well as the recombinant product being expressed by the cell inside the bioreactor while removing the waste product in the spent media. The fresh growth medium (“n+1” culture medium) used in the CFB may be different than the spent culture medium (“n” culture medium). Further, the fresh growth medium (“n+1” culture medium) may be a concentrated form than the spent culture medium (“n” culture medium). In some examples, the fresh growth medium (“n+1” culture medium) may be added as a dry powder. The “product quality” as used herein refers to desirable attributes in the biotherapeutic molecule. This may include, but are not limited to the specific glycosylation pattern or glycovariant composition of the biotherapeutic molecule. The term "reference product" refers to a currently or previously marketed recombinant biotherapeutic, also described as the "originator product" or "branded product" serving as a comparator in the biosimilarity studies. The term “biosimilar” interchangeably used with biosimilar drug refers to a biotherapeutic drug which is highly similar to the reference product in terms of purity, structure, and bioactivity and further have been determined to have no meaningful difference, in terms of safety, purity or potency (safety and effectiveness), from the reference product. The term “target/predetermined glycosylation profile” refers to the glycosylation profile of a recombinant biotherapeutic that would form an acceptance criteria for biosimilarity of a biosimilar to the reference product. As used herein, the term “turndown ratio” refers to the ratio of the maximum working volume to the minimum working volume of a bioreactor. For example, a bioreactor with a maximum operating volume of 200 liters and a minimum operating volume of 40 liters has a turndown ratio of 5:1. The term “temperature shift” refers to the change in temperature during the cell culture process in order to produce the therapeutic antibody. The term “normal temperature shift” refers to a temperature shift wherein the cell is cultured at a first temperature for a period of time and change the cell culture temperature to a second temperature, wherein the second temperature is lower than the first temperature. The term “reverse temperature shift” refers to a temperature shift wherein the cell is cultured at a first temperature for a period of time and change the cell culture temperature to a second temperature, wherein the second temperature is higher than the first temperature. The term “viability” refers to a measure of the metabolic state of a cell population which is indicative of the potential for growth. The viability of cells may be measured by methods well known in art such as classical trypan blue exclusion assay, propidium iodide (PI) exclusion assay, fluorescein di-acetate (FDA) inclusion assay. The term “viable cell density (VCD)” is defined as number of live cells per unit volume. Unless defined otherwise, the technical and scientific terms used herein are to be accorded the same meaning as would be commonly understood by one of skill in art to which the subject matter herein belongs. Description of the Embodiments Stainless steel bioreactors (SSBs) and fed-batch methods have been the mainstays of biotherapeutic industry. But of late single-use bioreactors (SUBs) and concetrated fed-batch (CFB) based methods are fast becoming the tools of choice in biopharmaceutical industry. The risks associated in operation and time associated with traditional stainless steel bioreactors has been a major driver for adoption of SUBs while CFB based methods are being favoured for reduction in cost of goods provided. The challenges of scaling up of a bioprocess methods is shared commonly by SSBs and SUBs – it is important to ensure that the product profile and productivity arrived at in early, smaller scales are maintained upon scaling up the process. In a typical cell culture bioprocess the influence of the process parameters like temperature, pH, pCO 2 , pO 2 on the cell physiology, metabolism, product formation kinetics and the product critical quality attributes (CQAs) cannot be ignored. Amongst the above enumerated factors, pCO 2 is a critical process parameter owing to the multiple roles CO 2 plays in the production of biotherapeutics using mammalian cell culture. pCO 2 impacts the cell culture performance by altering the internal organelle pH of the mammalian cells affecting the growth and metabolism. The pCO 2 also affects post translational modifications present on biotherapeutics thus influencing their quality attributes. Hence it is imperative that pCO 2 in the cell culture medium be maintained at optimal levels and ensure that it does not exceeds its threshold. CO 2 accumulation is the most commonly faced challenge during bench scale process scale-up to a manufacturing scale, due to longer residence time of the gas in large scale bioreactors. The CO 2 levels in cell culture is dependent on parameters like bioreactor shape, volume, agitation rate, aeration etc. The volumetric mass transfer coefficient of CO 2 (kLaCO 2 ) is an operational process parameter which can factors in these variables. Hence, an understanding of the mass transfer capacity of CO 2 is imperative to control the gas not to exceed its threshold value. Furthermore, a CO 2 stripping model can also be developed and implemented in large scale manufacturing process with the k L a data generated for CO 2 gas. In an embodiment, the cell culture process of the present invention is applicable to various mammalian cell lines that can be engineered to express a biotherapeutics, including but not limited to Chinese hamster cells (CHO), baby hamster kidney (BHK) cells, human embryo kidney (HEK) cells, mouse myeloma (NS0) cells, and human retinal cells. The cell culture methods of the present invention is not limited to any specific class of recombinant biotherapeutics. In exemplary aspects the cell culture methods of the present invention is applicable to monoclonal antibodies. In an embodiment the cell culture process of the present invention is applicable to stainless steel bioreactors. In another embodiment the cell culture process of the present invention is applicable to single use bioreactors. In an embodiment the cell culture process of the present invention is applicable to fed batch cell cultures. In an embodiment, the cell culture of the present invention is applicable to perfusion cell cultures. In yet another embodiment the, cell culture method of the present invention is applicable to concentrated fed batch cell cultures. In an embodiment, the present application provides for a cell culture method for production of a recombinant biotherapeutic composition having a target glycosylation profile, the said process comprising, (a) providing/culturing mammalian cells expressing the said recombinant biotherapeutic, (b) performing the N-1 seed stage and production stage in the same bioreactor, wherein a 5:2 turndown ratio is used for seed stage to production stage expansion along with calculated O 2 volumetric mass transfer coefficient (k L aO 2 ) from Van’t Riet equation (c) performing the production phase in a perfusion mode (d) maintaining the CO 2 volumetric mass transfer coefficient (k L aCO 2 ) of about 0.10 h -1 to about 0.27 h -1 , and the O 2 volumetric mass transfer coefficient (k L aO 2 ) of about 8.0 h -1 to about 10.5 h -1 (e) recovering the said recombinant biotherapeutic composition from the culture, wherein, the target glycosylation profile is characterised is terms of glycan variants including G0F glycans, total afucosylated glycans, galactosylated glycans. In an embodiment, the cell culture process of present invention would comprise use of a temperature shift in the production phase so as to obtain recombinant biotherapeutic composition with the target glycoprofile. In an embodiment, the cell culture process of the present invention would comprise more than one temperature shift, wherein the individual temperature shift might be result in subsequent lower temperature or higher temperature. For example in a cell culture process having two temperature shift, the following combinations are encompassed: 1st high temperature → 2nd low temperature → 3rd lower temperature, 1st high temperature → 2nd low temperature → 3rd high temperature, 1st low temperature → 2nd high temperature → 3rd low temperature. In a preferred embodiment, the cell culture method of present invention includes a single temperature shift, wherein the second temperature is lower than the first temperature. In yet another embodiment, the cell culture method of present invention includes a single temperature shift which marks the start of production phase in the production stage of cell culture. As an exemplification of the temperature shift strategy, the production stage of the cell culture is initiated at about 37°C and is lowered to about 35°C on about day 6 of the production stage and the culture is continued at this temperature till harvest. In an embodiment, the cell culture of the present invention uses alternating tangential flow for perfusion. The cell culture methods of the present invention is not limited to any specific class of recombinant biotherapeutics. In exemplary aspects the cell culture methods of the present invention is applicable to cytokines, growth factors, hormones, interferons and antibodies. In an embodiment, the cell culture of the present invention is applicable in production of recombinant biotherapeutics like darbepoetin, rituximab, trastuzumab, pertuzumab, bevacizumab, etanercept, aflibercept, abatacept, denosumab etc. Those skilled in the art will recognize that several embodiments are possible within the scope and spirit of this invention. The invention will now be described in greater detail by reference to the following non-limiting examples. The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope. EXAMPLES: Estimation of k L aO 2 and k L aCO 2 : Single use bioreactor setup: A sparger customized Single Use Bioreactor (SUB) of 1000L scale (Xcellerex XDR1000, GE Healthcare) was utilized for the study. The sparger configuration is a dual sparger system with 20µm X 8 sparger discs for air, O 2 and CO 2 ; and a Drilled Hole Sparger (DHS) with 20 holes of 2mm hole diameter for sparging air to strip off CO 2 were used. The bioreactor specifications being 1.5 aspect ratio of 1000L maximum working volume capacity, 30 inch tank internal diameter, single use bottom mounted 0.33 inch M40E impeller located at 15 o off centre to the tank. Experimental Design: A full factorial DOE was used to create design space with the three key factors: volume, agitation and aeration. Experiments were carried out in static gassing out method with volume factor at three levels 600L, 800L and 1000L, agitation rates of 95-125 RMP and aeration rates of 2-8 LPM.1XPBS was used as test solution with a pH of 7.39 and 281 mOSM/Kg osmolality, mimicking the properties of in-house cell culture media in use. The head space air exchange is performed for 3 cycles to reduce the residual nitrogen concentration between each set of study in the gassing phase resulting in a more precise and real time measurement of oxygen transfer. For the k L aO 2 measurements, air was sparged through 20µm x 8 sparger discs at 37°C process temperature and the %DO change was captured in every 20 second interval. The kLaCO 2 experiment was carried out by sparging CO 2 gas through T-sparger with 20 drilled holes of 2mm diameter at 37°C and correspondingly the ΔpH, pCO 2 values were recorded. The trials were carried out with the highest volume first, followed by gradual removal of the required test solution from the bioreactor for k L aO 2 . However kLaCO 2 study was limited to 1000L capacity. kLa measurement: The volumetric mass transfer coefficient of oxygen has been calculated from general mass balance equation . Where, C* - Saturation concentration of oxygen in liquid phase, C L - Concentration of oxygen in liquid phase, t – time required A graph is being plotted and the slope is determined, considering a straight line equation. The O 2 concentration in liquid (mmol/L) is calculated by gas solubility from Henry’s Law: P = Gas phase partial pressure of oxygen; H = Henry’s coefficient at temperature T; yo2 = mole fraction of Oxygen Compensating Henry’s constant for temperature by the below relationship: H -1 = 1.385 - 0.02635(T - 20) + 0.0004288 (T - 20) 2 Equation 3 Where T is Temperature in Centigrade (°C). The unit of Henry’s constant is atm m 3 /mol The mass transfer coefficient of dissolved CO 2 has been calculated from the overall mass balance equation Solving the equation with time limits ‘0’ to ‘t’ the equation becomes, The k L a is determined from the slope of the straight line equation. The liquid solubility of CO 2 (mmol/L) is calculated by Henry’s law. where HCO2 is Henry’s law constant for CO 2 , and [CO 2 ]aq is in mm Hg Compensating Henry’s constant for temperature by the below relationship: Where T is absolute Temperature in Kelvin (°K). The unit of Henry’s constant is in mm Hg L/M The most commonly used correlation for k L aO 2 is expressed in terms of power input per unit volume and superficial gas velocity and is known as Van’t Riet equation. where A, α and β are constants. Cell expansion and ATF setup: A CHO cell line producing IgG1 monoclonal antibodies and an in-house process utilizing proprietary defined basal media and feed media were operated. The seed was expanded from vial thaw in shake flasks till 700mL for 3 passages every 3-4 days in 1Xbasal media with incubator settings 37°C temperature maintenance, 5% CO 2 and 75% humidity. The culture from shake flask was inoculated to rocking wave bioreactors (WAVE Bioreactor, GE Healthcare). The culture was further scaled-up into 50L SUB (Xcellrex XDR50, GE Healthcare) and 5:2 turndown ratio seed expansion in 1000L SUB (Xcellrex XDR1000, GE Healthcare). The 5:2 turndown ratio is atypical from the regular practical applications, however in the current process design compensating the N-1 seed stage and the production bioreactor stage the half maximal working volume was developed. During the production bioreactor stage the seed bioreactor is topped up to the maximum capacity in concentrated fed-batch mode. For perfusion in concentrated fed-batch (CFB) mode, an alternating tangential flow (ATF) system with a 50 kDa filter was used (ATF10, Repligen). The ATF was operated at a pressure and exhaust flowrates (ATF exchange rate) of 54 LPM. The initial seeding density was 1.5±0.2 million cells/mL with ≥90% viability. A temperature shift was applied on the 6 th day of the production bioreactor stage whereby the culture temperature was reduced from 37°C to 35°C. The process was maintained at 40±10% DO, agitation rate of 110 RPM, and 7.05 ± 0.1 pH. Online process monitoring was done every 4 hours and daily offline sampling was performed by collecting cell culture samples from the bioreactor. Viable cell density (VCD) and viability were measured using the manual counting trypan blue exclusion method and on Vi-CELL XR cell counter, Beckman Coulter. Offline pH, pO 2 , and pCO 2 were measured using Rapid lab 348- Siemens blood gas analyzer. Results: The experimentally determined K L aO 2 are extrapolated as contour plots in Figure 1. The KLaO 2 values increases linearly with the flow rate and the agitation speed for all the for all the bioreactor volumes determined. Figure 2 depicts the K L aO 2 values determined for commonly used manufacture scales and conditions, viz.1000L with agitation rates ranges between 95-125 RPM and 2 LPM flow rate variation; the highest KLaO 2 value resulted with the largest flow rate and agitation speed. The Van’t Riet constants (A - 4230, α - 0.28, β – 0.72) were calculated based on the generated data sets. The current study aims at banking the KLaCO 2 data using 2mm X 20 DHS sparger, which can also be a measure of estimating the stripping capacity of the sparger during an on-going cell culture process at 1000L scale. Figure 3 shows the KLaCO 2 measurements with change in pH units in relation to the aeration supplied through DHS at the same agitation rates used for measuring K L aO 2 . In practice, this data has the decisive advantage while choosing the precise air flow rate through DHS sparger for stripping out a determined amount of pCO 2 while a cell culture process batch is in operation. Efficient pCO 2 removal mechanism can be strategized employing Figure 3, with calculated stripping capacity of each flow rate with regards to linear pH unit variability at each of the agitation rates. The cell culture profile results for the N-1 seed bioreactor stage mentioned in Figure 4 and Figure 5 were plausible and consistent with all the five production batches executed for the study, which is indicative of metabolic similarity between the two stages. In continuation to the seed bioreactor, the production bioreactor was operated in CFB mode, wherein adequate aeration and agitation strategy was implemented at full scale. An efficient pCO 2 control mechanism was employed to maintain optimal pCO 2 through-out the cultivation for cellular metabolism. The viable cell count cited in Figure 4 was consistent for all the five batches executed, importantly because the pO 2 was tightly maintained with the desired consumption rate. Figure 6 presents the air flow rate that was used for stripping the excessive pCO 2 in the five 1000L batches. The measurements from Figure 6 quantitatively describes the proportionate air flowrate employed from DHS sparger for step-to-step stripping of the excessive pCO 2 in the running batch, which if not degassed in appropriate can retard the cell growth and pose toxicity for the culture. The glycosylation profile and the titer of the product in the 1000L production batches (Table 1) was consistent, which shows that KLaCO 2 based aeration/agitation strategy enables achievement of a target product profile without any loss in productivity. Table 1. The product quality and titer in the various 1000L production batches