throbber
RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`Application of a Quality by Design Approach to the Cell Culture
`Process of Monoclonal Antibody Production, Resulting in the
`Establishment of a Design Space
`
`HIROAKI NAGASHIMA, AKIKO WATARI, YASUHARU SHINODA, HIROSHI OKAMOTO, SHINYA TAKUMA
`
`API Process Development Department, Chugai Pharmaceutical Company, Ltd., Tokyo, Japan
`
`Received 11 August 2013; revised 9 September 2013; accepted 13 September 2013
`
`Published online 3 October 2013 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23744
`
`ABSTRACT: This case study describes the application of Quality by Design elements to the process of culturing Chinese hamster ovary
`cells in the production of a monoclonal antibody. All steps in the cell culture process and all process parameters in each step were
`identified by using a cause-and-effect diagram. Prospective risk assessment using failure mode and effects analysis identified the following
`four potential critical process parameters in the production culture step: initial viable cell density, culture duration, pH, and temperature.
`These parameters and lot-to-lot variability in raw material were then evaluated by process characterization utilizing a design of experiments
`approach consisting of a face-centered central composite design integrated with a full factorial design. Process characterization was
`conducted using a scaled down model that had been qualified by comparison with large-scale production data. Multivariate regression
`analysis was used to establish statistical prediction models for performance indicators and quality attributes; with these, we constructed
`contour plots and conducted Monte Carlo simulation to clarify the design space. The statistical analyses, especially for raw materials,
`identified set point values, which were most robust with respect to the lot-to-lot variability of raw materials while keeping the product
`quality within the acceptance criteria. C° 2013 Wiley Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 102:4274–
`4283, 2013
`Keywords: antibody; biotechnology; cell culture; design of experiments; design space; failure mode and effects analysis; mathematical
`models; multivariate analysis; proteins; quality by design
`
`INTRODUCTION
`
`Quality by Design (QbD) is a science- and risk-based approach
`to pharmaceutical development and manufacture applicable to
`all stages of a product’s lifecycle.1,2 The QbD approach for bio-
`pharmaceutical manufacturing processes had its beginnings in
`2002 with the Current Good Manufacturing Practices for the
`21st Century initiative launched by the United States Food
`and Drug Administration,3 and the QbD concepts introduced
`there were subsequently followed by the International Con-
`ference on Harmonisation of Technical Requirements for Reg-
`istration of Pharmaceuticals for Human Use (ICH) guidance
`documents.4–7 The traditional approach to pharmaceutical de-
`velopment has been a rigid manufacturing process producing
`a product of variable quality that can be controlled by speci-
`fications; that approach is being replaced by an enhanced ap-
`proach based on QbD elements. Biopharmaceutical manufac-
`turing processes must be developed to produce a product that
`above all meets the needs of the patients while maintaining
`consistent efficacy and safety. Deep understanding of the re-
`lationships among process parameters, quality attributes, and
`
`Abbreviations used: CE-SDS, capillary electrophoresis-sodium dodecyl sulfate;
`CEX, cation exchange chromatography; CHO, Chinese hamster ovary; CPP,
`critical process parameter; CQA, critical quality attribute; DoE, design of ex-
`periments; FMEA, failure mode and effects analysis; HPLC, high-performance
`liquid chromatography; IVCD, initial viable cell density; mAb, monoclonal anti-
`body; QbD, quality by design; RPN, risk priority number; TOST, two one-sided
`t-test; VCD, viable cell density.
`Correspondence
`to: Hiroaki Nagashima (Telephone: +81-3-3968-6115;
`Fax: +81-3-3968-6259; E-mail: nagashimahra@chugai-pharm.co.jp)
`
`Journal of Pharmaceutical Sciences, Vol. 102, 4274–4283 (2013)
`C° 2013 Wiley Periodicals, Inc. and the American Pharmacists Association
`
`efficacy and safety is necessary to achieve such pharmaceutical
`development.
`The QbD approach is a more scientific, systematic, and com-
`prehensive approach that builds quality into the process in-
`stead of the traditional process of testing the quality of the
`product throughout the biopharmaceutical’s lifecycle (e.g., de-
`velopment, manufacture). It is said that pharmaceutical com-
`panies can understand their products more deeply and proceed
`with development more flexibly and robustly through a QbD
`approach, and if the development and manufacture of biophar-
`maceuticals are carried out according to QbD elements using
`appropriate tools, both pharmaceutical companies and regula-
`tory agencies will be able to reap the resulting benefits.8 For
`example, the design space for a manufacturing process is one of
`the most important components in the QbD approach. The ICH
`guidance documents define design space and working within
`the design space is not considered as a change. Some of the
`expected benefits of QbD for the biopharmaceutical industry
`thus include a reduction in the number of postapproval pro-
`cess changes and an increase in flexibility for pharmaceutical
`companies.9,10
`Interpretations of the application of QbD described in the
`guidelines differ within the industry, and concrete case studies
`of QbD as applied to biopharmaceutical drug substances are
`still limited. Some reports show original QbD approaches that
`focus on risk assessment and process characterization using de-
`sign of experiments (DoE) for the processes of cell culture11–20
`and purification,13,15,21,22 and the CMC Biotech Working Group
`published a mock case study covering all of the QbD elements
`for an antibody drug substance and drug product.23 Interpre-
`
`4274
`
`Nagashima et al., JOURNAL OF PHARMACEUTICAL SCIENCES 102:4274–4283, 2013
`
`Pfizer v. Genentech
`IPR2017-020(cid:20)(cid:28)
`Genentech Exhibit 2046
`
`

`

`tation of these initiatives by industry has been an ongoing
`process, with some notable industry and regulatory collab-
`orations for biopharmaceuticals, and each pharmaceutical
`company should take on the challenge of this approach and
`find its own way.24,25
`This study shows our QbD approach as applied to the process
`of culturing Chinese hamster ovary (CHO) cells in the produc-
`tion of a monoclonal antibody (mAb). The aim of this QbD case
`study is to understand the cell culture process more deeply
`and to increase the sophistication of the QbD approach itself.
`The QbD approach has four important elements—identification
`of critical process parameters (CPPs), identification of critical
`quality attributes (CQAs), understanding the relationships be-
`tween CPPs and CQAs, and establishment of an appropriate
`control strategy. In this case study, we focused on quality risk
`management using failure mode and effects analysis (FMEA),
`and we conducted a multivariate study using DoE to establish
`a design space.
`Risk assessment was composed of risk identification, risk
`analysis, and risk evaluation; it was carried out for the cell
`culture process by using FMEA on the basis of established
`scientific knowledge and our experimental knowledge. FMEA
`is a common and useful risk analysis tool to find process pa-
`rameters that require further process characterization.12,13,16
`Process characterization experiments are typically conducted
`using DoE to evaluate not only the main effects of parameters
`but also their interactions.11–20 The potential CPPs that were
`identified by this risk assessment of the production culture step
`in the cell culture process were experimentally evaluated on a
`scaled down model by a multivariate study using DoE. A design
`space was established on the basis of those results and checked
`by Monte Carlo simulation. Because lot-to-lot variability of raw
`materials is said to contribute greatly to product quality, safety,
`and process performance,26–28 our multivariate study included
`an evaluation of material variability by finding the interac-
`tions between material lot and process parameters. The scaled
`down model used for process characterization was confirmed
`to be appropriate by demonstrating that its performance was
`comparable to that of a 2500 L bioreactor.
`Through this QbD case study, we were able to understand
`the cell culture process over a range of operation wider than
`that traditionally used. In this article, we present the QbD
`approach we have developed for the cell culture process of a
`mAb currently under clinical development.
`
`MATERIALS AND METHODS
`
`Cell Culture
`
`A CHO cell line expressing a glycosylated mAb was used in
`this case study. Cells were grown in suspension using a serum-
`free cell culture medium. Cells were subcultured using spinner
`flasks (Bellco Glass, Vineland, New Jersey) in a CO2 incubator
`(Thermo Scientific, Waltham, Massachusetts). Production cul-
`ture was performed in fed-batch mode with continuous feeding.
`We used 1 L bioreactors (ABLE, Tokyo, Japan) as the scaled
`down model for process characterization, and a 2500 L biore-
`actor (Hitachi, Tokyo, Japan) for the qualification of the scaled
`down model. Cell culture fluids were centrifuged and filtered to
`remove cells for analysis, and antibodies were purified from the
`fluids with a POROS A20 protein A column (Life Technologies,
`Carlsbad, California).
`
`RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`4275
`
`Analytical Methods
`
`Viable cell density (VCD) and viability were measured by using
`a Cedex automated cell counter and analyzer (F. Hoffmann-La
`Roche, Basel, Switzerland). Offline pH was measured by us-
`ing a GASTAT blood gas analyzer (Techno Medica, Kanagawa,
`Japan). Antibody titer was measured by using protein A affinity
`high-performance liquid chromatography (HPLC). Charge vari-
`ants were measured by using cation-exchange chromatography
`(CEX) HPLC. N-linked glycosylation (e.g., high-mannose) was
`measured by hydrophilic interaction chromatography HPLC
`after cutting with N-Glycosidase F (F. Hoffmann-La Roche) and
`labeling with 2-aminobenzamide (Sigma–Aldrich, St. Louis,
`Missouri). Whole antibodies (H2L2) were measured by capillary
`electrophoresis-sodium dodecyl sulfate (CE-SDS) using the PA
`800 plus Pharmaceutical Analysis System (Beckman Coulter,
`Brea, California).
`
`Risk Assessment and Process Characterization
`
`A cause-and-effect diagram was used for risk identification, and
`all process steps and process parameters in the cell culture pro-
`cess were defined. The method applied to the risk analysis and
`risk evaluation of process parameters in the cell culture process
`was based on FMEA. In this case study, two study designs gen-
`erated by DoE were integrated and used for the process char-
`acterization. The parameter IVCD, pH, and temperature were
`evaluated by face-centered central composite design, and cul-
`ture duration and material lot were evaluated by full factorial
`design. The process characterization study was performed with
`a total of 30 conditions. For the purposes of this case study, titer
`was chosen as a representative key performance indicator and
`the main peak in CEX, H2L2 in CE-SDS, and high-mannose
`were chosen as representative CQAs with which to analyze
`the impact of process parameters statistically. Multivariate re-
`gression analysis was performed for each performance indi-
`cator and quality attribute to establish mathematical models.
`Main effects and interaction effects of continuous factors on
`responses were considered statistically significant when the F
`ratio—which means the F value of the factor compared with
`the F value of error—was more than 2.0. If a model was judged
`as rational from statistical and scientific views, contour plots
`were generated and superimposed to define the design space.
`Monte Carlo simulation was performed to discover the distri-
`bution of model outputs as a function of random variation in
`the parameters and model noise. To evaluate the differences
`arising from lot-to-lot variability in raw material, the main ef-
`fect of material lot and the interaction effects between material
`lot and the other parameters were also incorporated into the
`prediction models.
`
`Qualification of Scaled Down Model
`
`Culture performance indicators and quality attributes of the
`scaled down model were compared with those of the large-scale
`production bioreactor to demonstrate the appropriateness of
`the scaled down model. We compared data from 7 runs of the
`scaled down model bioreactor, which ran during process devel-
`opment as a control with data from 6 runs of the 2500 L biore-
`actor used for the production of the clinical drug substance. The
`cells used for small-scale culture were derived from the same
`working cell bank as used for large-scale production. Five cell
`tubes were used for the small-scale runs and two cell tubes were
`used for the large-scale runs. Performance indicators, such as
`
`DOI 10.1002/jps.23744
`
`Nagashima et al., JOURNAL OF PHARMACEUTICAL SCIENCES 102:4274–4283, 2013
`
`

`

`4276
`
`RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`Figure 1. A part of the cause-and-effect diagram identifying the process steps and associated process parameters in the cell culture process.
`All identified process parameters were evaluated for further risk assessment. CQA: critical quality attribute, KPI: key performance indicator.
`
`cell growth and titer, and quality attributes, such as CEX vari-
`ants and whole antibody, were compared between the scales.
`Comparability between the scaled down model and the large-
`scale bioreactor was judged by using the mean ± 3SD of each.
`
`RESULTS AND DISCUSSION
`
`Risk Assessment for Process Parameters
`
`Quality risk assessment comprises risk identification, risk
`analysis, and risk evaluation. A cause-and-effect diagram—also
`called a fishbone diagram or Ishikawa diagram—is commonly
`used in many fields for risk identification.21,29 All process steps
`and all process parameters in the cell culture process were iden-
`tified by cause-and-effect diagram. Part of this cause-and-effect
`diagram is shown in Figure 1, indicating that the inoculum
`train step has 11 parameters, the production medium reconsti-
`tution step has 10 parameters, the feed medium reconstitution
`
`step has 12 parameters, and the production culture step has 16
`parameters. In total, the cell culture process was found to have
`14 steps and 176 process parameters (data not shown).
`Risk analysis and evaluation should be the qualitative or
`quantitative process of linking the likelihood of occurrence and
`severity of harm and the ability to detect the harm.5 Although
`many risk analysis tools are known, and no one tool or set
`of tools is applicable to every situation, FMEA is one of the
`most common and useful tools for risk analysis in pharmaceu-
`tical process development.12,13,16 The risks of all of the process
`parameters in the 14 cell culture steps were analyzed and eval-
`uated using FMEA. Each process parameter was assessed in
`terms of three factors: severity of impact, probability of occur-
`rence, and likelihood of detection (Table 1). Severity of impact
`was ranked on a five-level scale with a score of 1–11; a score
`of 11 indicates that the parameter has already been found to
`have a deleterious impact on CQAs or key performance indica-
`tors. Probability of occurrence was ranked on a four-level scale
`
`Table 1. Definitions Used for FMEA
`
`Score
`
`Definition
`
`1
`2
`4
`
`7
`11
`
`1
`2
`3
`5
`
`1
`3
`5
`
`(A)
`It is known that the parameter does not affect CQAs and KPIs because we already have data.
`The parameter is not expected to affect CQAs and KPIs based on experience and prior knowledge (Other cell lines’ data show no effect).
`The parameter might affect CQAs and KPIs based on experience, prior knowledge, and literature (Some other cell lines’ data show
`effects).
`The parameter is expected to affect CQAs and KPIs based on experience and prior knowledge (Other cell lines’ data show effects).
`It is known that the parameter affects CQAs and KPIs because we already have data.
`
`(B)
`Never in the foreseeable future.
`Never, but might happen.
`Rare (less than once per 20 batches, below 5%)
`Sometimes (more than once per 20 batches, over 5%)
`
`(C)
`Change can be detected and dealt with immediately.
`Change can be detected and dealt with within a day.
`Change cannot be detected or dealt with.
`
`(A) Severity of impact, (B) probability of occurrence, and (C) likelihood of detection.
`CQA, critical quality attribute; KPI, key performance indicator.
`
`Nagashima et al., JOURNAL OF PHARMACEUTICAL SCIENCES 102:4274–4283, 2013
`
`DOI 10.1002/jps.23744
`
`

`

`RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`4277
`
`Table 2. Results of Failure Mode and Effects Analysis for the Production Culture Step
`
`Process Parameter
`
`Control Space
`
`Characterization Range
`
`IVCD (×105 cells/mL)
`Duration (day)
`pH (day 4–14)
`Feed start timing (h)
`pH control start timing (h)
`Cell age (day)
`pH (upper, day 0–3)
`Minimum glucose concentration (g/L)
`Feed rate (%)
`Dilution rate (%)
`Inner pressure (MPa)
`Temperature (◦C)
`Agitation power (rpm)
`Viability (%)
`Working volume (%)
`Dissolved oxygen (%)
`
`2.03–2.79
`13.4–13.8
`6.70–6.88
`72.00–72.02
`72.00–72.32
`–
`7.16–7.28
`–
`24.6–28.6
`7.2–8.1
`0.013–0.017
`36.7–37.2
`18.0–18.1
`98.8–99.8
`−0.4–0.1
`28.8–70.9
`
`1.20–2.80
`13.0–14.0
`6.65–6.95
`60.00–96.00
`60.00–96.00
`130
`<7.40
`>0.2
`20.0–40.0
`6.0–9.0
`0.010–0.020
`36.5–37.5
`17.0–19.0
`>80.0
`−10.0–10.0
`>10.0
`
`S
`
`7
`7
`7
`4
`4
`7
`4
`4
`4
`2
`2
`7
`2
`4
`2
`2
`
`O
`
`D
`
`RPN
`
`2
`2
`2
`2
`2
`1
`2
`2
`2
`2
`2
`2
`2
`2
`2
`2
`
`5
`5
`3
`5
`5
`5
`3
`3
`3
`5
`5
`1
`3
`1
`1
`1
`
`70
`70
`42
`40
`40
`35
`24
`24
`24
`20
`20
`14
`12
`8
`4
`4
`
`IVCD, initial viable cell density; S, severity of impact score; O, probability of occurrence score; D, likelihood of detection score; RPN, risk priority number.
`
`with a score of 1–5; a score of 5 indicates that the parame-
`ter has the highest probability of deviating from the control
`space. Likelihood of detection was ranked on a three-level scale
`with a score of 1–5; a score of 5 indicates that the parameter
`cannot be detected or cannot be dealt with even if detected.
`Considering that the manufacturing process is operated within
`a certain control space, characterization ranges should be wider
`than the control space. Characterization ranges were defined
`as “ coefficient × control space” and were used throughout the
`risk assessment and process characterization as ranges within
`which the set point values of process parameters were changed.
`A coefficient of 3 was basically used to calculate the character-
`ization ranges; however, when the resulting ranges were so
`wide that product quality or process performance had to devi-
`ate beyond preferable ranges, a coefficient of less than 3 was
`used. In the risk assessment, the severity of impact for each
`process parameter was evaluated within the characterization
`range, whereas probability of occurrence and likelihood of de-
`tection for each process parameter were evaluated at the edges
`of the control spaces. The risk analysis was conducted on the
`basis of information taken from the literature, experimental
`and manufacturing data on this and other similar processes
`during developments, and general scientific principles.
`Table 2 summarizes the risk analysis results for the produc-
`tion culture step. Although in the risk analysis, the severity
`of impact scores of process parameters were defined with re-
`spect to each quality attribute and performance indicator, the
`highest score is taken as the severity of impact score. Each
`risk priority number (RPN) was calculated by multiplying the
`scores for severity of impact, probability of occurrence, and like-
`lihood of detection, and the resulting scores were used to judge
`which process parameter should be subsequently evaluated in
`the process characterization. The RPN threshold was deter-
`mined by experts in each function and, based on an acceptable
`level of risk at this stage, was set at 40. IVCD, culture duration,
`and pH, each of which had an RPN of over 40, were defined as
`potential CPPs and were evaluated in the process characteri-
`zation. From among all the process steps, FMEA identified no
`potential CPPs other than those for the production culture step
`(data not shown).
`
`Parameters, which had an RPN below the threshold, were
`carefully reviewed by the risk assessment team. Although tem-
`perature in the production culture step was not defined as a
`potential CPP by FMEA, this parameter was evaluated in the
`process characterization for two reasons. One reason was that
`the control space and the characterization range for tempera-
`ture were very narrow because temperature in the large-scale
`bioreactor had been strictly controlled and the risk posed by
`variations in temperature might have been undervalued in the
`FMEA. The other reason was the concern that there are poten-
`tial interaction effects between temperature and other process
`parameters. For these two reasons, temperature was also de-
`fined as a potential CPP. Material lot—medium lot and other
`materials lot in medium reconstitution step—was also defined
`independently of the FMEA as a factor that should be evalu-
`ated in the production culture step because lot-to-lot variation
`in raw material has the potential risk of leading to variability in
`quality attributes and performance indicators. Material lot for
`the production culture was evaluated to statistically analyze
`its variability. Material lot for the subculture was not included
`because the purpose of that step is only to passage and expand
`cells for production culture and the impact on quality attributes
`and performance indicators was considered to be limited. In to-
`tal, four process parameters and material lot were evaluated in
`process characterization.
`
`Process Characterization
`
`The risk assessment stage of our QbD case study identified
`four potential CPPs (IVCD, culture duration, pH, and tem-
`perature) and material lot in the production culture step for
`experimental evaluation in the process characterization stage.
`The actual impact of these potential CPPs and material lot
`on quality attributes and performance indicators was evalu-
`ated by using a DoE-based multivariate study. Because the
`four continuous factors (temperature, pH, IVCD, and duration)
`and the one nominal factor (material lot) cannot be comprehen-
`sively evaluated by a single experimental design, a three-level
`face-centered central composite design to evaluate IVCD, pH,
`and temperature was integrated with a two-level full factorial
`design to evaluate culture duration and material lot to create
`
`DOI 10.1002/jps.23744
`
`Nagashima et al., JOURNAL OF PHARMACEUTICAL SCIENCES 102:4274–4283, 2013
`
`

`

`4278
`
`RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`Figure 2. Multivariate regression analysis for main peak in CEX. (a) Distribution of main peaks in CEX. (b) Correlation between actual values
`and predicted values from the multivariate regression model. (c) Residual analysis for the multivariate regression model. (d) Significant factors
`affecting main peak in CEX. IVCD, initial viable cell density.
`
`a single multivariate experimental design. All of the main ef-
`fects of the five experimental factors and all of the interaction
`effects between the factors could be estimated independently in
`this experimental design. Because the ranges evaluated during
`process characterization are typically wider than the operating
`ranges encountered during manufacturing, we used the char-
`acterization ranges as the experimental ranges during process
`characterization (The characterization range of temperature
`was reset as 36.0–38.0). A total of 30 fed-batch production cul-
`ture runs were conducted using 1 L bioreactors, and the titer,
`main peak in CEX, H2L2 in CE-SDS, and high-mannose were
`used as potential key performance indicators or potential CQAs
`to statistically analyze the impact of process parameters. The
`acceptance criteria for the main peak in CEX and for H2L2 in
`CE-SDS were set using the specifications for manufacturing,
`and the acceptance criterion for high-mannose was set using
`the mean + 3SD of data obtained during process development.
`The acceptance criterion for titer was not set because titer does
`not affect the efficacy or safety of the mAb in patients.
`As a representative example of the procedure used, the
`statistical analysis using JMP software (SAS Institute, Cary,
`North Carolina) for the main peak in CEX is described in detail
`below, although the same analysis was performed for each qual-
`ity attribute and performance indicator. First, the distribution
`of CEX main peaks was monitored by histogram to confirm that
`the data were normally distributed (Fig. 2a). In cases where the
`data were not normally distributed (e.g., viability), a variable
`
`transformation such as logit transformation or Box–Cox trans-
`formation was applied before performing statistical analysis
`that assumes the analyzed data are normally distributed. The
`main effects, interaction effects, and quadratic effects with an
`F ratio of over 2.0 were considered to be significant and were
`incorporated into a multivariate regression model. The actual
`values and the predicted values by the model were shown in a
`scatter plot (Fig. 2b).
`As an indication of how well the mathematical model fitted
`the experimental data, rather than using R2, which increases
`mechanically as more factors are incorporated into the model,
`we used adjusted R2 that takes into consideration the number
`of factors incorporated. A lack of fit is also a useful indication to
`confirm whether sufficient factors including interaction effects
`and quadratic effects are incorporated into the model or not.
`The error measured for these exact replicates is called pure
`error and is the portion of the sample error that cannot be ex-
`plained or predicted by the mathematical model. A lack of fit
`can be significantly greater than pure error if sufficient factors
`including interaction effects and quadratic effects are not in-
`corporated into the model. The adjusted R2 and the P value of
`lack of fit of the mathematical model were 0.914 and 0.302, re-
`spectively, indicating the appropriateness of the mathematical
`model. Residuals of mathematical models should be randomly
`distributed around zero; a scatter plot of residuals by predicted
`values indicated that no notable tendency can be observed
`(Fig. 2c). The statistical analysis showed that the main peak
`
`Nagashima et al., JOURNAL OF PHARMACEUTICAL SCIENCES 102:4274–4283, 2013
`
`DOI 10.1002/jps.23744
`
`

`

`Table 3. Design Space for the Production Culture Step
`
`Potential CPP
`
`IVCD (×105 cells/mL)
`pH
`Culture duration (day)
`Temperature (◦C)
`
`Range
`
`1.2–2.8
`6.65–6.90
`13–14
`36–38
`
`The range of pH in the design space is narrower than the characterization
`range because of the deviation of high-mannose from the desired criterion.
`CPP, critical process parameter; IVCD, initial viable cell density.
`
`in CEX was most significantly influenced by the main effects of
`temperature, pH, IVCD, and culture duration; by the interac-
`tion effects between temperature and pH and between temper-
`ature and duration; and by the quadratic effects of temperature
`and pH (Fig. 2d). Similar results were obtained for the other
`responses (data not shown), and the prediction models were
`used for the further analysis.
`The four statistical prediction models (for main peak in CEX,
`titer, H2L2 in CE-SDS, and high-mannose) were then depicted
`as contour plots to clarify the multivariate acceptable range in
`the production culture step (Fig. 3). In this case study, spec-
`ifications or mean + 3SD from the process development data
`were used as acceptance criteria. Deviation beyond the accep-
`tance criteria is shown in the contour plots as shaded areas.
`Although no deviation was observed for the main peak in CEX
`and H2L2 in CE-SDS, high-mannose levels were higher than
`the acceptable criteria in some ranges. These results indicate
`that the ranges of some parameters should be narrower than
`the characterization range to establish an appropriate design
`space for the production culture step. One possible constraint
`is to narrow the range of pH from 6.65–6.95 to 6.65–6.90 be-
`
`RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`4279
`
`cause pH had the biggest impact on high-mannose and this
`constraint could completely remove all shaded areas from the
`contour plots. Table 3 shows an example design space for the
`production culture step in the cell culture process.
`Another description of a design space can be achieved by ap-
`plying constraints to the prediction models, as has been shown
`in other case studies.12,13,16,18,20,21 In such a case, the area within
`the criteria with values that predict high-mannose can be de-
`fined as the design space. Although this design space will be
`wider than the design space shown in Table 3, the description
`is complicated and difficult to understand intuitively. If con-
`straints are not critical for the process, such as in the current
`case study, a simpler description of the design space might be
`better.
`The distributions of model outputs with random variation in
`the parameters and model noise were evaluated by Monte Carlo
`simulation. The set point value and the perturbative range
`observed in the large-scale production were used as the mean
`and SD of each parameter, respectively. The relative sums of
`mean error of the models were used as the SD of response, and
`a total of 5000 runs were evaluated in the simulation. Figure 4a
`shows the statistical parameters used for the simulation, and
`the results of the simulations are shown in Figures 4b–4e as
`histograms. The minimum values of main peak in CEX, titer,
`and H2L2 in CE-SDS, and the maximum value of high-mannose
`were respectively 79.69%, 94.02%, 87.03%, and 7.30%, which
`were all within the acceptable criteria. These results indicate
`that the production culture step in the cell culture process was
`robust even considering possible noise such as mathematical
`model noise and perturbative noise in large-scale production.
`To promote better understanding of the effect of raw ma-
`terials in the cell culture process, further statistical analysis
`was performed. The main effect of material lot and the interac-
`tion effects between material lot and the other four parameters
`
`Figure 3. Understanding the design space in terms of contour plots. Internal x- and y-axes are temperature and pH, respectively. External
`x- and y-axes are IVCD and culture duration, respectively. Deviation beyond the acceptance criteria is shown as shaded areas. The dashed line
`shows an example of a constraint to establish a design space. IVCD, initial viable cell density.
`
`DOI 10.1002/jps.23744
`
`Nagashima et al., JOURNAL OF PHARMACEUTICAL SCIENCES 102:4274–4283, 2013
`
`

`

`4280
`
`RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`Identifying the distribution of model outputs using Monte Carlo simulation. (a) Statistical parameters used for the simulation.
`Figure 4.
`The set point value and perturbative range observed in large-scale production were used as the mean and SD of each parameter, respectively.
`The relative sums of mean error of models were used as SD of response. Results of 5000 runs in the simulation for (b) main peak in CEX, (c) titer,
`(d) H2L2 in CE-SDS, and (e) high-mannose. IVCD, initial viable cell density.
`
`Figure 5. Robustness analysis for the variability of material lot using the profiles of interaction between material lot and parameters for main
`peak in CEX, titer, H2L2 in CE-SDS, and high-mannose. Each line in the interaction profile represents each material lot. IVCD, initial viable
`cell density.
`
`Nagashima et al., JOURNAL OF PHARMACEUTICAL SCIENCES 102:4274–4283, 2013
`
`DOI 10.1002/jps.23744
`
`

`

`RESEARCH ARTICLE – Pharmaceutical Biotechnology
`
`4281
`
`Figure 6. Qualification of scaled down model. (a) Mean ± 3SD of viable cell density from 2500 L scale production runs (n = 6) are shown
`as dashed lines. Viable cell densities from the scaled down model runs (n = 7) are shown as solid lines. (b) Main peak in CEX, (c) titer, and
`(d) high-mannose were compared between the scales. Results are expressed as mean ± 3SD. VCD, viable cell density.
`
`were incorporated into the four statistical prediction models
`to evaluate differences in variability of material lot. Figure 5
`shows the profiles of interaction between material lot and
`parameters for main peak in CEX, titer, H2L2 in CE-SDS, and
`high-mannose. For each parameter, the set point value that
`results in the smallest difference in response between the dif-
`ferent material lots means that the set point is more robust
`with respect to the variability of the material lot as compared
`with other set points. The effect of temperature on H2L2, pH on
`H2L2, IVCD on H2L2, and IVCD on high-mannose were each af-
`fected by material lot. Although a lower temperature is pr

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket