Bioreactor Integration and PAT for Real-Time Control of Upstream Bioprocesses
eBook
Published: October 27, 2025
Credit: Thermo Fisher Scientific
Upstream bioprocessing underpins biopharmaceutical production and spans multiple scales from bench to commercial manufacturing. Optimizing efficiency and ensuring consistent product quality across these stages is critical.
Conventional monitoring methods can be slow and offer limited visibility, creating a need for technologies that deliver continuous, accurate measurement and control which supports seamless scale-up.
This eBook highlights advanced process monitoring and scalable bioreactor integration solutions that enable real-time control and optimized upstream production - acting as the true “eye of the bioreactor.”
Download this eBook to explore:
- The latest strategies in process analytical technology for real-time monitoring and control of critical process parameters
- How Raman spectroscopy enables fed-batch and perfusion cell culture long-term with minimal variability
- Scalable bioreactor solutions that streamline seed train scale-up
PAT-enabled scaling and
optimization of upstream
bioprocesses
Process Raman spectroscopy and bioreactor
application note compendium
Compendium
Introduction
Scaling up a bioprocess is a complex, multi-variable
challenge. Designing processes with scalable parameters is
essential for ensuring confidence when transitioning from the
research laboratory to clinical and production operations.
The application notes in this compendium show how
modern bioreactors and process Raman technology can
enhance bioprocess development and enable advanced
process control strategies from pilot to production scale.
Raman spectroscopy as a PAT tool in a continuous
perfusion run 5
Real-time metabolite monitoring with process Raman 14
Monitor and maintain glucose concentrations in real-time 17
Table of Contents
Additional upstream processing resources
Streamlining seed train scale-up through utilization of the
turndown ratio of a single use bioreactor 23
Part 1: Intuitive bioprocess scale-up from
bench scale to pilot scale 29
Demonstration of scalability for multiple cell lines 36
Chemometric model transferability across process
Raman instruments 43
Measure key variables with process Raman for
enhanced control 47
Product solutions 51
PAT-enabled advanced process control for
cell culture
PAT-enabled advanced process
control for cell culture
Using Raman Spectroscopy as a Process Analytical
Technology Tool in a 50-Day Continuous Perfusion Run
Application note
Authors
Mike Bates¹, Kevin Broadbelt¹,
Jan Ott², Vivian Ott², Regine Eibl²,
Lin Chen¹, David Kuntz¹, Lin Zhang¹,
Mathew Zustiak¹, and Sue Woods¹
(Thermo Fisher Scientific)1
School of Life Sciences and Facility
Management, Institute of Chemistry
and Biotechnology, ZHAW Zurich
University of Applied Sciences², 8820
Wädenswil, Switzerland
Highlights
• Implementation of Process Raman as a PAT tool for in-line and real-time
monitoring of critical process parameters, including titer, cell densities,
and nutrient concentrations in mAb-producing, high-cell-density perfusion
CHO cell culture, is discussed.
• The Thermo Scientific™ MarqMetrix™ All-In-One Process Raman Analyzer with
Thermo Scientific™ Lykos™ PAT Software, with 21 CFR Part 11 compliance,
is used for in-line, real-time, calibration-free monitoring of a bioreactor during a
50-day continuous perfusion run.
Summary
The biopharmaceutical industry is driven by the need to increase production and
reduce costs while maintaining product quality. One effective way to achieve
this goal is to streamline the monitoring process of biologics production, thus
allowing more effective control of production parameters. In this application note,
we introduce a case study using Process Raman for in-line, real-time monitoring
of specific critical process parameters (CPPs) in high-density mammalian
perfusion cell cultures reaching 100–130 million cells mL-1. The Thermo Scientific™
MarqMetrix™ All-In-One Process Raman Analyzer System is shown to enable
in-line measurements of CPPs, including glucose, lactate, ammonium, product titer,
and cell viability, over the course of a 50-day continuous perfusion bioreactor run.
Background
In recent years, Raman spectroscopy has gained popularity
as a process analytical technology (PAT) tool that enables
real-time monitoring and control of critical bioprocessing
parameters that are key to the successful production of
therapeutic drugs. The product portfolio of biologics is
broadening, and implementation of different spectroscopic
PAT tools can address the limitations of traditional off-line
analytical methods for such products. The production of
biotherapeutic products requires high process efficiency while
ensuring product quality and minimized manufacturing costs¹.
Implementing PAT tools in biopharmaceutical manufacturing is
a critical priority identified by the FDA, with the goal of allowing
rapid development and access to novel therapeutics and
existing medications without compromising product quality²,
³.
Cell culture processes are labor intensive due to the frequent
sample analyses required. When operating bioreactors in
fed-batch mode, nutrients are periodically supplemented via
a bolus feeding strategy using predetermined volumes of
concentrated nutrients. While batch production is a
well-vetted manufacturing method, this production strategy can
be slow and inefficient. Recent advances in biomanufacturing
processes involve continuous processing. Continuous
bioprocessing technologies, whether upstream or downstream,
can increase speed while decreasing the costs of producing
these essential biologics⁵. Perfusion cell culture is a process
that uses filters to keep cells in a bioreactor while continuously
exchanging culture medium. Fresh medium replenishes
nutrients and carbon sources, while cellular waste and medium
depleted nutrients are removed. Key advantages of bioreactors
operated in perfusion mode include flexibility, lower cost,
improved quality, and greater speed.
In this study, we focused on the continuous perfusion operation
mode for suspension cell culture. The goal of continuous
perfusion is to develop a process that maintains a steady state
in which productivity and product quality can be sustained
long-term with minimal variability, with bioreactors running for
30 – 90 days.
Raman spectroscopy is a laser‐based method for generating
a chemical fingerprint of a sample²,
³. A key advantage of
Raman spectroscopy is its ability to measure numerous
analytes in a non‐destructive manner, in situ, and with low
interference from water. Fortuitously, numerous analytes with
distinct Raman fingerprints enable monitoring of CPPs such
as nutrient feed levels, cell metabolites, cell growth profiles,
product levels, and product quality attributes¹,
². The MarqMetrix
All-In-One Process Raman Analyzer with Thermo Scientific
Lykos PAT Software is designed to offer accurate, reliable,
real-time identification and quantification of numerous CPP
analytes. Process Raman is extremely advantageous when
adopted into a continuous perfusion culture system. Utilizing
an immersed Raman optical sensor provides real-time process
information about the CPPs, unlike traditional monitoring
systems, which require manual sampling and off-line analysis¹,
².
Noted advantages include an increase in data acquisition
frequency, the opportunity for rapid correction of any detected
process parameter deviations, and reduction of contamination
risk due to a reduced offline sampling.
This application note describes the integration of the
MarqMetrix All-In-One Process Raman Analyzer with the
50L Thermo Scientific™ DynaDrive™ bioreactor to perform
in-line measurements of CPPs in a continuous perfusion run
(See Figures 1-3). Here, we highlight the integration of the
MarqMetrix All-In-One Process Raman Analyzer System to
perform in-line measurements of glucose, lactate, ammonium,
viable cell density (VCD), total cell density (TCD), and titer.
This PAT tool provides accurate prediction models for several
parameters and metabolites and shows high correlations
with offline measurements. The simultaneous measurements
of metabolites, product titer, and protein concentration
allow for real-time process control, demonstrating the
effectiveness of process Raman spectroscopy as a PAT tool in
biopharmaceutical industries.
Materials and methods
Cell line and medium
For the two 50 L perfusion cultivations, a trastuzumabproducing CHO K1 cell line was used. The basal medium was
0.66x concentrated High-Intensity Perfusion CHO medium
(Gibco™), whereas 1x concentrated medium was used as the
feed medium. Both media were supplemented with 4 mmol L-1
l-glutamine (Gibco™) and 1% Anti-Clumping Agent (Gibco™).
Inoculum production
Inoculum production was carried out over a period of 10 days.
The first three passages were carried out at a shake flask
scale. A 10 L wave-mixed bioreactor (Cellbag 10 L, Cytiva) with
a working volume of 5 L was used for the last passage.
Perfusion cultivations at 50 L scale
For the 50 L perfusion cultivations, the ATF version of the
DynaDrive S.U.B. (Thermo Scientific) with the corresponding
G3Pro Bioprocess Controller was used. Cell retention was
realized with Repligen’s XCell ATF6 single-use version and
the corresponding XCell LS controller at an average ATF flow
rate of 17 L min-1. The cultivation was started with a VCD of
3x105
cells mL-1. Perfusion was started after an initial 3-day
batch phase. The perfusion rate was limited to 1 d-1. The bleed
was controlled with the permittivity probe IncyteArc (Hamilton
Bonaduz) to keep a viable cell volume of 100–130 mm3
mL-1
constant. To keep the bioreactor volumes constant at 50 L,
the harvest pump was controlled by the bioreactor weight.
Glucose concentration was either kept constant at 2 g L-1 with
a CIT Sens Bio glucose sensor (C-CIT) for the first run or was
manually controlled for the second run. To prevent foaming, a
1:10 diluted antifoam C emulsion (Sigma Aldrich) was added
quasi-continuously. In all cultivations, the pH was controlled
to a value of ≤7.15 by adding CO₂ via the drilled hole sparger.
DO was controlled at 40% by adding N₂, air and O₂ using the
corresponding sparger. An overlay gassing rate of 0.05 vessel
volumes min-1 with air was chosen. The stirrer speed was set to
136.5 rpm (corresponding to 40 W m-3) in the DynaDrive.
Analytics
Once a day, cell-specific parameters, such as the VCD,
viability, and cell diameter, were determined with a Cedex
HiRes analyzer (Roche Diagnostics, Basel, Switzerland). The
Cedex Bio analyzer (Roche Diagnostics) was used to check
the substrate and metabolite concentrations of glucose,
lactate, glutamine, and ammonium as well as the IgG titer.
Figure 2. 50L Thermo Scientific DynaDrive S.U.B. for cell
culture applications.
Figure 1. Scheme of perfusion process configuration and integration of an in-line Raman sensor (MarqMetrix All-In-One Process
Raman Analyzer with Lykos PAT Software).
High-Intensity
Perfusion CHO
Medium (Gibco)
50L Thermo Scientific
DynaDrive S.U.B.
bioreactor
XCell LS
Lab controller
Thermo Scientific™ MarqMetrix™ All-In-One Process
Raman Analyzer with Thermo Scientific™ Lykos PAT Software
Fresh
Media
Spent
Media
Additionally, an augmentation approach was used to improve
the models. Data from two ZHAW perfusion runs was
collected. One run was used to augment training data to
improve the prediction accuracy for the other. The run used to
augment the training data was weighted equally to the rest of
the training data despite having far fewer samples because this
data was assumed to have greater similarities to the prediction
run. In this manner, the model was able to learn from the bulk
data in the regular training set but was fine-tuned specifically
to give the best predictions on ZHAW data. This approach
optimizes the benefits of using both a broad general dataset
and a specifically targeted but much smaller dataset.
Results
To analyze the chemometric modeling results, let us first
define the key figures of merit: bias and RMSEP. Bias is the
average difference between predicted values and reference
values. The Root Mean Squared Error of Prediction (RMSEP)
is the combined error of bias and precision, where precision
is the randomness (noise) around the mean of the predicted
values, assuming there is no bias. Furthermore, the quality of a
chemometric model is typically evaluated using the Q-residuals
and Hotelling’s T-squared values. The Q-residuals are used
to quantify how well the model fits the raw data. Q-residuals
should typically be less than 1, which indicates that the model
accounts for all the variance in the spectra. A high Q-residual
value indicates an observation not well explained by the model,
suggesting it may be an outlier. Hotelling’s T-squared values
measure the distance of each observation from the model’s
center in the space of the retained components. A high
T-squared value expresses an observation far from the model’s
center, which could also suggest it as a potential outlier. Both
Q-residuals and Hotelling’s T-squared values are important
tools for model diagnostics in chemometrics.
While the results of applying generalized chemometric models,
based on bolus-fed CHO cell lines, produced prediction
errors of approximately 1 g L-1 for glucose and lactate, these
generalized models produced poor results when predicting
ammonium, titer, VCD and TCD. In contrast, the augmentation
of the generalized models using one ZHAW run to
predict the other ZHAW run resulted in prediction errors of
0.36 g L-1 for glucose and 0.37 g L-1 for lactate. Furthermore,
the augmentation of the generalized model enabled accurate
predictions for ammonium (RMSEP = 0.95 mmol L-1)
and titer (RMSEP = 0.36 g L-1) while also providing prediction
accuracy for VCD & TCD of ≈10 million cells mL-1, which
translates to +/- 10% of the stationary phase concentrations
of ≈100 million cells mL-1.
Thermo Scientific MarqMetrix All-In-One Process
Raman Analyzer Measurements
Measurements were performed using the MarqMetrix
All-In-One Process Raman Analyzer System, with the optical
Bioreactor ball probe directly immersed in the bioreactor
(50L). Each Raman spectra resulted from an average of
60 measurements, with an integration/exposure time of 1 sec
and a laser power setting of 450 mW. The total acquisition time
per data spectra was 2 minutes, with a timestamp matched
between the MarqMetrix All-In-One Process Raman Analyzer
and offline instrument analysis to build the model.
Chemometric Model building
Independent data from multiple MarqMetrix All-In-One
Process Raman Analyzers and numerous bioreactors were
used to create models for each analyte. The training datasets
were collected from 12–24 samples per bioreactor to create
each chemometric model. In-line and at-line measurements
were aligned using timestamps between the MarqMetrix
All-In-One Process Raman Analyzer and the at-line instrument,
the Nova Flex II. All data was reviewed before building the
models. In addition, an algorithm was implemented to remove
data spikes in the spectra caused by cosmic rays. The spectral
region of interest was selected, and multiple spectra were
averaged to increase signal-to-noise ratios such that each
measurement corresponded to a ten-minute read-time. The
spectra were pre-processed to remove differences in the
baseline due to fluorescence and other effects. Spectra were
also normalized to remove differences in absolute intensity
between various bioreactor types. Partial Least Squares
(PLS) models were created for each analyte of interest, and
leave-out-one-run cross-validation was performed to test
the optimization of each model. Analytes of interest include
glucose, lactate, ammonium, VCD, TCD, and titer.
Figure 3. MarqMetrix All-In-One Process Raman Analyzer
with optical MarqMetrix Bioreactor BallProbe™ immersed in a
5L glass bioreactor.
Residual
# Occurrences
–0.5 0.0 0.5 12.0
D. Residuals
Reference
Predicted
0.0 2.5 5.0 7.5 10.0 12.5
C. Gluc (g/L) Predicted vs. Reference
Panel C in each figure demonstrates the linearity of the
CHO-cell training data, augmented with a weighted ZHAW run
(green) and a ZHAW run used as a prediction data set (purple).
One reason for applying the weighted-ZHAW augmentation to the
independent training data set was the very small linear range for
many analytes in these perfusion runs. In contrast to the small
linear range of CPPs such as glucose, lactate, ammonium, and
titer, the cell density values, VCD and TCD, exhibited a wide range
from 20–130 million cells mL-1. The results demonstrate excellent
correlations between the model prediction data obtained from
the Thermo Scientific MarqMetrix All-In-One Process Raman
Analyzer and the offline data for numerous CPPs (Table 1).
Figures 4-9 show the correlation between offline data and
predictions made by applying the chemometric models to the
spectra collected using the Thermo Scientific MarqMetrix
All-In-One Process Raman Analyzer. In each Figure, the
predicted vs. offline data is plotted in panel A. These figures
demonstrate the ability to monitor changes in various CPPs
in real-time with a high degree of accuracy. Panel B in each
figure speaks to the quality of the chemometric model, as the
Q-residuals and Hotelling’s T-squared values for each analyte
model exhibit very low values; this indicates a good fit of the
model with negligible unaccounted variance or outliers.
Figure 4. Modeling results for glucose monitoring in the 50-day perfusion run of the bioreactor. (A) shows the modeling results
generated from the process Raman spectra compared to the results of offline analysis. (B) shows the Q-residuals vs. Hotelling’s
T² values. (C) shows the linearity of the predicted vs. offline values. (D) shows the histogram of residuals between predicted and
offline values.
0.0
0.2
0.4
0.6
0.8
1.0
T²
A. Glucose – Predicted vs. Offline
Q
0.0 0.5 1.0
0.0
0.5
Glucose (g L-1)
2023-07-15 2023-07-22 2023-08-01 2023-08-08 2023-08-15 2023-08-22
1.0
1.5
2.0
2.5
B. Gluc Q vs. T²
0.0
2.5
5.0
7.5
10.0
12.5
2.5
5.0
7.5
10.0
12.5
0.0
Model
Timeseries
Offline
Model
ZHAW
1=1
Predicted
Offline
A. Ammonium – Predicted vs. Offline
A. Lactate – Predicted vs. Offline
Figure 6. Modeling results for ammonium monitoring in the bioreactor’s 50-day perfusion run. (A) shows the modeling results generated
from the process Raman spectra compared to the results of offline analysis. (B) shows the Q-residuals vs. Hotelling’s T² values. (C)
shows the linearity of the predicted vs. offline values. (D) shows the histogram of residuals between predicted and offline values.
Figure 5. Modeling results for lactate monitoring in the bioreactor’s 50-day perfusion run. (A) shows the modeling results generated
from the process Raman spectra compared to the results of offline analysis. (B) shows the Q-residuals vs. Hotelling’s T² values.
(C) shows the linearity of the predicted vs. offline values. (D) shows the histogram of residuals between predicted and offline values.
Residual
# Occurrences
–0.6 –0.4 –0.2 0.0
D. Residuals
Reference
Predicted
0 1 2 3
C. Lac (g/L) Predicted vs. Reference
0.0
0.2
0.4
0.6
0.8
1.0
T²
Q
0.0 1.0 2.0
0.5
Lactate (g L-1)
2023-07-15 2023-07-22 2023-08-01 2023-08-08 2023-08-15 2023-08-22
1.0
1.5
B. Lac Q vs. T²
0
1
2
3
2
4
6
8
10
0
Model
Timeseries
Offline
Model
ZHAW
1=1
Predicted
Offline
0.0
0.5 1.5 0.2
Residual
# Occurrences
0 1
D. Residuals
Reference
Predicted
0 5 10 15 20 25
C. NH₄+ (mmol/L) Predicted vs. Reference
0
4
6
8
10
12
T²
Q
0.0 1.0 2.0
1
NH₄+ (mmol L-1)
2023-07-15 2023-07-22 2023-08-01 2023-08-08 2023-08-15 2023-08-22
2
3
B. NH₄+ Q vs. T²
0
10
20
25
2.5
5.0
7.5
10.0
12.5
0.0
0
0.5 1.5 2
Model
Timeseries
Offline
Model
ZHAW
1=1
Predicted
Offline
4
2
5
15
Figure 8. Modeling results for Total Cell Density (TCD) monitoring in the bioreactor’s 50-day perfusion run. (A) shows the modeling
results generated from the process Raman spectra compared to the results of offline analysis. (B) shows the Q-residuals vs. Hotelling’s
T² values. (C) shows the linearity of the predicted vs. offline values. (D) shows the histogram of residuals between predicted and
offline values.
Figure 7. Modeling results for Viable Cell Density (VCD) monitoring in the 50-day perfusion run of the bioreactor. (A) shows the
modeling results generated from the process Raman spectra compared to the results of offline analysis. (B) shows the Q-residuals
vs. Hotelling’s T² values. (C) shows the linearity of the predicted vs. offline values. (D) shows the histogram of residuals between
predicted and offline values.
A. TCD – Predicted vs. Offline
A. VCD – Predicted vs. Offline
Residual
# Occurrences
0 20
D. Residuals
Reference
Predicted
0 50 100
C. VCD Predicted vs. Reference
0
1
2
3
T²
Q
0.0 1.0 2.0
40
VCD (10⁶ cells mL-1)
2023-07-15 2023-07-22 2023-08-01 2023-08-08 2023-08-15 2023-08-22
60
80
B. VCD Q vs. T²
–25
25
75
125
2
4
6
10
12
0
20
0.5 1.5
Residual
# Occurrences
0
D. Residuals
Reference
Predicted
0 50 100
C. TCD Predicted vs. Reference
0
1
2
3
T²
Q
0.0 1.0 2.0
40
TCD (10⁶ cells mL-1)
2023-07-15 2023-07-22 2023-08-01 2023-08-08 2023-08-15 2023-08-22
60
80
B. TCD Q vs. T²
0
50
100
125
5.0
7.5
10.0
12.5
15.00
0.0
20
0.5 1.5 20
100
25
75
100
Predicted
Offline
Model
Timeseries
Offline
Model
ZHAW
1=1
0
50
100
8
Model
Timeseries
Offline
Model
ZHAW
1=1
Predicted
Offline
2.5
-20
Analyte Units RMSEP Bias
Glucose g L-1 0.36 -0.15
Lactate g L-1 0.37 0.32
Ammonium mmol L-1 0.95 -0.71
VCD million cells mL-1 10.78 0.47
TCD million cells mL-1 10.59 0.62
Titer g L-1 0.36 -0.27
Table 1. Summary of Chemometric Modeling Results for this 50-day Perfusion Run.
Figure 9. Modeling results for titer monitoring in the 50-day perfusion run of the bioreactor. (A) shows the modeling results generated
from the Process Raman spectra compared to the results of offline analysis. (B) shows the Q-residuals vs. Hotelling’s T² values.
(C) shows the linearity of the predicted vs. offline values. (D) shows the histogram of residuals between predicted and offline values.
A. Titer – Predicted vs. Offline
Residual
# Occurrences
0.00 0.50
D. Residuals
Reference
Predicted
0 2 4
C. Titer Predicted vs. Reference
0.0
0.4
0.6
1.0
T²
Q
0.0 1.0 2.0
0.5
IgG Titer (g L-1)
2023-07-15 2023-07-22 2023-08-01 2023-08-08 2023-08-15 2023-08-22
1.0
1.5
B. Titer Q vs. T²
–1
1
3
5
2
4
6
10
12
0
0.0
0.5 1.5
2.0
0
2
4
8
Conclusion
The study in this application note highlights an approach to
using Raman spectroscopy, specifically Process Raman, for
in-line, real-time monitoring of a high cell density mammalian
cell culture run in a bioreactor operated in perfusion mode
The MarqMetrix All-In-One Process Raman Analyzer provides
exceptional data quality, which, when combined with
multivariate PLS modeling, enables in-line, real-time monitoring
of CPPs glucose, lactate, ammonium, titer, and cell density
measurements. The ability to obtain accurate cell density
measurements in a wide range from 20-130 million cells mL-1
showcases the robustness and utility of these chemometric
models and the efficacy of utilizing the MarqMetrix All-In-One
Process Raman Analyzer to collect high-quality spectral data.
Great data lead to robust and accurate chemometric models,
thereby unlocking the potential of Process Raman as a critical
PAT tool in biopharmaceutical processes.
The simultaneous measurements of metabolites and product
titer are of special interest as they will allow for greater
process control of cultivation and purification parameters
within continuous biomanufacturing processes. Additionally,
Process Raman greatly enhances the operator’s understanding
of the CHO cell perfusion process as these CPPs are
measured repeatedly in short intervals in a non-destructive
manner. This study demonstrates the capability of Process
Raman spectroscopy as a PAT tool that can pair seamlessly
with automation systems to improve yields and enhance
product quality.
The results of the perfusion cultivations discussed
in this application note have been published in detail,
https://www.mdpi.com/2227-9717/12/4/806.⁶
Predicted
Offline
Model
Timeseries
Offline
Model
ZHAW
1=1
0.2
0.8
–0.25 0.25 0.75
References
1. Research, C. for D. E. and. PAT — A Framework for Innovative Pharmaceutical
Development, Manufacturing, and Quality Assurance. U.S. Food
and Drug Administration. https://www.fda.gov/regulatory-information/
search-fda-guidance-documents/pat-framework-innovative-pharmaceuticaldevelopment-manufacturing-and-quality-assurance (accessed 2023-03-21).
2. Real time monitoring of multiple parameters in mammalian cell culture bioreactors
using an in-line Raman spectroscopy probe. Nicholas R Abu-Absi ¹, Brian M Kenty,
Maryann Ehly Cuellar, Michael C Borys, Sivakesava Sakhamuri, David J Strachan,
Michael C Hausladen, Zheng Jian Li. Biotechnol Bioeng. 2011 May;108(5):1215-21.
doi: 10.1002/bit.23023. Epub 2010 Dec 22
3. Quick generation of Raman spectroscopy based in-process glucose control to
influence biopharmaceutical protein product quality during mammalian cell culture.
Brandon N Berry ¹, Terrence M Dobrowsky ¹, Rebecca C Timson ¹,
Rashmi Kshirsagar ¹, Thomas Ryll ¹, Kelly Wiltberger ² Biotechnol Prog 2016
Jan-Feb;32(1):224-34. doi: 10.1002/btpr.2205. Epub 2015 Dec 21.
4. Modernizing the Way Drugs Are Made: A Transition to Continuous Manufacturing |
FDA, Sau (Larry) Lee, Ph.D., Deputy Director of the Office of Testing and Research,
and Chair of the Emerging Technology Team, Office of Pharmaceutical Quality, CDER
5. Integrated Continuous Pharmaceutical Technologies – A Review. András Domokos,
Brigitta Nagy, Botond Szilágyi, György Marosi, and Zsombor Kristóf Nagy.
Organic Process Research & Development 2021 25 (4), 721-739 DOI: 10.1021/acs.
oprd.0c00504
6. Scaling Fed-Batch and Perfusion Antibody Production Processes in Geometrically
Dissimilar Stirred Bioreactors. Vivian Ott, Jan Ott, Dieter Eibl, Regine Eibl.
Processes. 2024, 12(4), 806.
Learn more at thermofisher.com/marqmetrixAIO
For research use only. Not for use in diagnostic procedures. For current certifications, visit thermofisher.com/certifications
© 2024 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific
and its subsidiaries unless otherwise specified. MCS-AN1068-EN 5/24
Real time metabolite monitoring using the MarqMetrix
All-In-One Process Raman Analyzer and the
500L Dynadrive Single-Use Bioreactor (S.U.B.)
Application note
Authors
Juan Villa, Matthew Zustiak, Elizabeth
Amoako, David Kuntz, Lin Zhang, Kevin
Broadbelt and Sue Woods (Thermo
Fisher Scientific)
Background
The capability of Raman spectroscopy to reflect small changes in complex aqueous
systems has expanded the application of this technique to analyze biopharmaceutical
processes such as cell growth in bioreactors. When the Raman spectrometer is
utilized as a continuous process analyzer of these complicated chemical systems, this
technology can monitor biopharmaceutical production processes real-time,
in-situ and non-destructively. The ability of Raman Spectroscopy to detect changes of
numerous metabolites during bioreactor processes has elevated this technology to a
robust Process Analytical tool.
Thermo Scientific™ MarqMetrix™ Single-Use Bioreactor BallProbe™ Sampling Optic
with TouchRaman™ immersion technology
Reusable Raman Spectroscopy analysis optical probes offer benefits such as
improved process repeatability and reliability by reducing run-to-run variability.
With the MarqMetrix All-In-One Process Raman Analyzer, there are a wide range of
probes available. The MarqMetrix Single-Use Bioreactor BallProbe Sampling Optic is
designed to meet the requirements of the bioprocess industry and can be used with
the MarqMetrix All-In-One Process Raman Analyzer. These probes are quick and
easy to swap and connect, are durable and can handle sterility practices including
offline autoclaving.
Thermo Scientific™ DynaDrive™ Single-Use Bioreactor (S.U.B.), for perfusion cell
culture applications
The DynaDrive Single-Use Bioreactor (S.U.B.), the latest advancement in S.U.B.
technology, offers improved performance and scalability for large volume
bioproduction. The cuboid-shaped tank offers several key advantages over legacy
S.U.B. designs including superior mixing and mass transfer capabilities as well as
improved scalability.
This application note describes the integration of the MarqMetrix All-In-One Process
Raman Analyzer with the 500L DynaDrive Single-Use bioreactor to perform in-line
measurements of critical process parameters (CPPs). Utilizing continuously generated
spectral data throughout a cell growth culture run, accurate prediction models for
several parameters and metabolites were developed using this integrated system.
Process Raman analysis
Materials and methods
Cell culture and feeding strategy
Cell culture was performed in a 500L DynaDrive S.U.B, containing
a working volume of approximately 320L of cell culture medium,
and inoculated with 0.5*106 cells/mL at a temperature of 36.5 °C,
pH= 6.9+/- 0.3, DO = 50%). The pH level was controlled by CO2
gassing and sodium carbonate additions, as needed. The cells
were grown in a chemically defined medium and fed daily with a
two-step feeding process, starting at day 3. The first feed media
was added at 4% by weight based on the starting volume and
the second feed media was added at 0.4%. The temperature
was shifted to 33 °C on day 6. The run terminated after 14 days.
The bioreactor was covered to protect from stray light. After
autoclaving, the MarqMetrix Single-Use Bioreactor BallProbe
Sampling Optic was inserted into the DynaDrive S.U.B. during the
run for in-line, real-time spectral Raman data generation.
Many pre-processing techniques were tested, including the
Savitzky Golay filter with derivatives, Automatic Whitaker
Smoothing, Extended Multiplicative Scatter Correction, SNV,
and mean centering. The best pre-processing techniques used
varied, based on which specific parameter of interest was
modelled. Partial Least Squares (PLS) models were created for
each property of interest and cross-validation was performed
to test the optimization of each model. Properties of interest
included glucose, lactate, glutamine, glutamate, TCD, VCD, and
other common metabolites generated during the bioreactor
culture run.
Results
In this work, continuous in-line Raman spectroscopy was applied
to a fed-batch CHO cell culture process. The in-line spectral
data was correlated to the offline analytical data acquired for
parameters of interest. The use of Raman spectroscopy to monitor
process parameters first requires chemometric model building
with an externally calibrated data set (independent offline data). To
assess the accuracy of the MarqMetrix All-In-One Process Raman
Analyzer predicted values, bioreactor samples were collected
daily and analyzed for comparison. The root mean square error
of calibration (RMSEC), root mean square error of cross validation
(RMSECV) and root mean square error of prediction were
calculated for each parameter (RMSEP). The error was averaged
based upon the prediction of the model to identify the RMSECV
which is used to construct the model. The RMSEP is used to test
the model against “new” data that the model has not seen. The
coefficient of variation, R², was recorded for each PLS model. The
value is used to determine the amount of variation of the Y variable
which the model predictors (X variables) can explain.
It is important to note that the combined use of several large,
independent data sets from bioreactor runs of the same CHO
culture process produced predictive chemometric models that
are more accurate and robust. For this study, five independent
datasets from previous bioreactor runs were combined to train
a large chemometric model. The calibration model was then
applied to the spectral data obtained during this DynaDrive S.U.B
run. The data indicates that the model was able to accurately
predict this new dataset, and that model predictions were highly
correlated with data measurements collected offline for numerous
metabolites as shown in Table 1.
Figure 2. Thermo Scientific MarqMetrix All-In-One Process Raman Analyzer.
Figure 1. 500L Thermo Scientific DynaDrive S.U.B. for cell culture applications.
MarqMetrix All-In-One Process Raman
Analyzer measurements
Measurements were performed using the MarqMetrix All-InOne Process Raman Analyzer, with the MarqMetrix Single-Use
Bioreactor BallProbe Sampling Optic of the MarqMetrix All-In-One
Process Raman Analyzer directly immersed in the bioreactors
(500L) D. Each Raman spectra was the result of an average of
20 measurements with an integration/exposure time of 3 sec,
and laser power setting at 450 mW. The total acquisition time per
data spectra was 2 minutes, with a timestamp matched between
the MarqMetrix All-In-One Process Raman Analyzer and off-line
instrument analysis to build the model.
Chemometrics, model building
Independent data from multiple MarqMetrix All-In-One Process
Raman Analyzer instruments, probes, and bioreactor types were
used to create models. The training datasets were collected
from 45 samples per bioreactor to create each chemometric
model. The spectral data was reviewed, and outlier spectral
spikes caused by cosmic rays were removed. The spectral
region of interest was selected, and the spectra were preprocessed to remove the baseline and maximize signal to noise.
For research use only. Not for use in diagnostic procedures. For current certifications, visit thermofisher.com/certifications
© 2024 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific
and its subsidiaries unless otherwise specified. TS_MCS_AN_DynaDrive_MMAIO_0824
Learn more at thermofisher.com/marqmetrixAIO
Table 1. Correlation of model prediction with offline data analysis.
Metabolite Predicted R2
Predicted RMSEC RMSECV RMSEP
Glucose (g/L) 0.98 0.43 0.49 0.40
Lactate (g/L) 0.92 0.15 0.18 0.25
Glutamine (mmol/L) 0.92 0.42 0.48 0.58
Titer 0.92 0.21 0.25 0.37
Cell Viability (%) 0.94 1.72 2.29 1.83
Figure 3. Thermo Scientific DynaDrive S.U.B. Chemometric Model
Plots- Comparison of Raman Model vs Offline Analytical Data for Important
Bioreactor Parameters.
Conclusions
The MarqMetrix All-In-One Process Raman Analyzer provides
accurate in-line, real-time measurements of the critical process
parameters glucose, glutamine, and lactate as well as total
and viable cell densities in the 500L DynaDrive S.U.B. using
the reusable MarqMetrix Single-Use Bioreactor BallProbe
Sampling Optic with TouchRaman immersion technology. The
correlation analysis shows excellent agreement between the
model prediction data and the offline analytical data, indicating
the robustness of the model applied to the parameters shown
in Table 1.
0
2
4
6
8
2022
Glucose
DynaDrive S.U.B. Gluc (g/L) RMSEP:0.4
Gluc (g/L)
8/25 8/27 8/29 8/31 9/01 9/03 9/05 9/07
Predicted
Offline
0.0
0.5
1.0
1.5
2.0
2022
Lactate
DynaDrive S.U.B. Lac (g/L) RMSEP:0.25
Lac (g/L)
8/25 8/27 8/29 8/31 9/01 9/03 9/05 9/07
Predicted
Offline
0.0
1.0
2.0
3.0
3.5
2022
Titer
DynaDrive S.U.B. Titer RMSEP:0.37
Titer
8/25 8/27 8/29 8/31 9/01 9/03 9/05 9/07
Predicted
Offline
0
1
2
3
6
2022
Glutamine
DynaDrive S.U.B. Gln RMSEP:0.58
Gln
8/25 8/27 8/29 8/31 9/01 9/03 9/05 9/07
Predicted
Offline
80
85
90
95
100
2022
Cell Viability
DynaDrive S.U.B. Viability RMSEP:1.83
Viability
8/25 8/27 8/29 8/31 9/01 9/03 9/05 9/07
Predicted
Offline
0.5
1.5
2.5
4
5
Use of Lykos and TruBio Software Programs for
Automated Feedback Control to Monitor and Maintain
Glucose Concentrations in Real Time
Application note
Authors
Juan Villa, Matthew Zustiak,
David Kuntz, Lin Zhang,
Nimesh Khadka, Kevin Broadbelt
and Sue Woods
(Thermo Fisher Scientific)
Summary
In this study, the Thermo Scientific™ MarqMetrix™ All-In-One Process Raman Analyzer
with Thermo Scientific™ Lykos™ PAT Software and Thermo Scientific™ TruBio™ 6.0
Bioprocess Control Software was used to provide in-line feedback control of glucose
in both Fed-Batch and Perfusion CHO cell culture processes without the need
for operator intervention. This ability to monitor and maintain the desired glucose
concentration in real time leads to improved process consistency and product
quality, supporting crucial mAb post-translational modification of the product
(e.g., glycosylation). An OPC-UA (Open Platform Communications United Architecture)
connection between the Lykos PAT software and the TruBio 6.0 bioprocess control
software was used for feedback control of one of the integrated pumps on the G3Lab
controller supplying a concentrated glucose solution. Both Fed-Batch and Perfusion
cultures were maintained automatically without operator process intervention.
Introduction
Process analytical technology (PAT) enables manufacturers to measure and control
a process based on the product’s critical quality attributes (CQAs) in real time.
Enhanced control of critical process parameters (CPPs) optimizes quality while
reducing the cost and time of product development and manufacturing. With PAT,
these CPPs and, in some instances, CQAs can be measured in real time, therefore
leading to gains in essential real-time process information and the ability to create
a quality-by-design workstream. In recent years, process Raman spectroscopy
has gained popularity as a PAT tool that enables real-time monitoring and control
of critical bioprocessing parameters which are key to the successful production of
therapeutic drugs. Successful production implies high process efficiency, high and
consistent product quality, and minimized manufacturing costs¹. Implementing PAT
tools in biopharmaceutical manufacturing continues to receive much interest,
allowing rapid development and access to therapeutics and existing medications
without compromising high quality⁵,
⁶.
Complex, multivariate, and univariate instrument data are
interpreted, and based on this information, critical process
parameters are predicted and, where necessary, adjusted
to optimize the outcome of the process. Analytical results
make it possible to predict the quality of the end material and
understand how altering CPPs will affect the process and
product. In turn, by executing experiments with real-time quality
predictions, the relationships between the CPPs and CQAs
can be established to develop true process understanding.
Armed with this knowledge, it is possible to ‘close the loop’ and
control the process using those quality predictions. Integration
of continuous monitoring into a bioprocess and the application
of analytical data is crucial for understanding the process and
proactively addressing challenges. A key challenge is in-line
monitoring of critical quality attributes such as glycosylation
that affect the bioproduct’s stability, immunogenicity, safety,
and potency. Maintaining the glucose concentration at a
steady level in the bioreactor is extremely important to ensure
consistent glycosylation of the product while optimizing product
yields¹,
². This online continuous monitoring and control also
allow for the reduction of manual sampling and feeding of the
bioreactor, which are costly and inefficient and increase the risk
of contamination each time the sterile system is accessed.
This application note describes the use of the MarqMetrix
All-In-One Process Raman Analyzer with Lykos PAT Raman
Software to monitor glucose levels in a bioreactor and to
trigger automated addition of feed so as to maintain the
desired concentration. Communication between Lykos and
the TruBio 6.0 control software was executed using
OPC-UA⁹. A system that incorporates a sensitive Raman
Process Analyzer and an established complex feedback
loop allows for real-time monitoring of a critical parameter
while maintaining stable concentrations. This system frees
the operator from manual sampling and feeding, thereby
eliminating the risks of contamination to the health of the cells.
Materials & Methods
Bioreactor Control System and Process Parameters
Process monitoring and control were performed using the
MarqMetrix All-In-One Process Raman Analyzer and TruBio 6.0
Bioprocess Control Software powered by the Emerson DeltaV
Distributed Control System. Cell culture was performed in a
HyPerforma Glass reactor and a HyPerforma G3Lab Controller.
Fed-Batch strategy
A 3L reactor was prepared with an initial working volume of 1.5L
of SPM (ExpiCho stable production media + 6mM glutamine
+ 1 g/L pluronic) culture medium and inoculated with ExpiCho
cells at 0.6*10⁶ cells/mL. Bioreactor environmental control
parameters were set at a temperature of 36.5°C, a pH of 7.0, and
a dissolved oxygen content of 40% air saturation. The pH level
was controlled as needed using CO₂ gassing and 1M sodium
carbonate additions.
Feeding Strategy
The cells were grown in a chemically defined medium and
were fed using a continuous feeding process starting on
day 3. The feed media (2X EFFICIENT FEED C+) was added at
3% reactor weight daily. The run was terminated after 14 days.
The bioreactor was covered to protect it from stray light.
Perfusion strategy
Fresh basal medium addition was initiated on day 3 at
0.5 reactor volumes/day while spent media was removed
through a proprietary perfusion device at the same rate, thereby
maintaining the reactor volume at a constant level. The
perfusion rate was increased stepwise up to 2 volumes per day
by day 5 and held at that rate for the duration of the culture.
Glucose control Strategy
Glucose feedback control was initiated on day 3 for the fed-batch
culture and day 0 for the perfusion culture, using the glucose
value provided by Lykos to the TruBio 6.0 Bioreactor Control
Software, to control the glucose concentration in the bioreactors
at 3.0 g/L for the fed-batch and 4.0 g/L for the perfusion cultures.
MarqMetrix All-In-One Process Raman
Analyzer Monitoring
Measurements were performed using the MarqMetrix All-In-One
Process Raman Analyzer, with the optical BioReactor Ball Probe
of the MarqMetrix All-In-One Process Raman Analyzer directly
immersed in the bioreactors⁷,
⁸. Each Raman spectrum was the result
of an average of 20 measurements with an integration/exposure
time of 3000 milliseconds and laser power set at 450 mW. The
total acquisition time per data spectra was 2 minutes (1 min dark
spectrum correction and 1 min sample spectra) with a timestamp
matched between the MarqMetrix All-In-One Process Raman
Analyzer and offline instrument analysis to align the online and offline
results. Measurements were taken every 2 minutes, and based on
these measurements, the pump rate was adjusted automatically.
Table 1. Media and Cell Line Summaries.
Bioreactor Run Summaries
Run Mode: Fed-Batch Run Mode: Perfusion
• 1 Bioreactors
(5L glass vessel)
• Inoculation:
0.6*10⁶ cells ExpiCHOs
• Initial media:
ExpiCHO SPM
• Feed media, EFC+
• Run time: 14 days
• 1 Bioreactor
(3L glass vessel)
• Inoculation:
0.6*10⁶ cells ExpiCHOs
• Initial media:
HipCHO
• Feed media:
HipCHO (perfusion)
• Run time: 10 days
LyKos PAT Software
The Lykos provided user access to the MarqMetrix All-InOne Process Raman Solution and data access for the stored
Raman spectra. As part of this application note, two new
capabilities were developed: A process analysis engine that
applies a chemometric model to generate a process value, and
an OPC-UA server which allows automated data access.
Process Control Integration
Glucose addition rate control by the TruBio 6.0 Process Control
System was based on process measurements from the Lycos
PAT Software via OPC-UA.
Chemometric Model building
Independent data from multiple MarqMetrix All-In-One
Process Raman Analyzers and 3 bioreactors were used to
create models. The training datasets were collected from
12–24 samples per bioreactor to create each chemometric
model. In-line and at-line measurements were aligned using
timestamps between the MarqMetrixAll-In-One Process
Raman Analyzer and the at-line instrument, a Flex2 cell culture
analyzer from Nova Biomedical. All data was reviewed before
building the models. In addition, an algorithm was implemented
to remove data spikes in the spectra caused by cosmic
rays. The spectral region of interest was selected, and each
measurement corresponded to ten minutes. The spectra were
pre-processed to remove the baseline and, maximize the
signal-to-noise ratio, and correct for path length differences.
Partial Least Squares (PLS) models were created for each
property of interest, and leave-out-one-run cross-validation was
performed to test the optimization of each model. Properties
of interest included glucose, lactate, and titer generated during
the bioreactor culture run.
Lykos PAT Software communicated directly with the bioreactorcontrolling software to send the glucose concentration
measured in the bioreactor. A MarqMetrix All-In-One Process
Raman Analyzer and Lykos PAT Software were integrated with
TruBio 6.0 Biorprocess Control Software (powered via the
DeltaV Distributed Control Platform from Emerson). Raman
spectroscopy was used to determine the glucose concentration
in the cell culture.
Figure 1A. Process Control Diagram.
Figure 1B. Process Monitoring & Control.
Glucose
Feed
Solution TruBio 6.0
BioProcess
Control System
Process Value
via OPC-UA
Raman Spectra
via Ethernet
MarqMetrix
All-In-One
Process
Raman Solution
MarqMetrix
BallProbe
Sampling Optic
BioReactor
Feed
Pump
Control
LASER Signal
via Fiber
Optic Cable
Lycos PAT
Software
• LASER Pulse Injection
• Raman Spectra Detection
• Generate the spectra
• Read the spectra
• Calculate glucose concentration
• Send the glucose value
to TruBio 6 software control
• Read the glucose concentration
sent by Lykos PAT Raman Software
• Adjust the pump rate for feed
(containing glucose)
Ethernet
BioReactor
LASER and Raman Signal
Feed
Control
Results
During the cell culture process, glucose was consumed by the cells, and
concentration levels stabilized at around 3 g/L for the fed-batch culture and around
4 g/L for the perfusion reactor. Figures 2A & B show that once the target glucose
concentration was reached, the glucose concentrations were precisely maintained
at 3 g/L and 4 g/L for the fed-batch and perfusion cultures, respectively. Traditional
offline samples were collected to measure the glucose and lactate concentrations as
controls to compare the accuracy of the Raman analyzer measurements.
Figure 2A. Fed-Batch
Data from real-time control of glucose and monitoring of lactate and titer for a fed-batch culture are presented here.
Figure 2B. Perfusion
Data from real-time control of glucose and monitoring of lactate for a perfusion culture are presented here.
Glucose Lactate Titer
Correlation of Model Prediction with Offline Data
Metabolite/Variable Predicted R² Predicted RMSEP
Glucose (g/L) 0.890 0.315
Lactate (g/L) 0.929 0.223
Titer (g/L) 0.980 0.200
Correlation of Model Prediction with Offline Data
Metabolite/Variable Predicted R² Predicted RMSEP
Glucose (g/L) 0.95 0.34
Lactate (g/L) 1.00 0.36
Glucose Lactate
Discussion
In the above-described experiments, process data was
monitored by Lykos PAT software and easily integrated
with TruBio bioprocess control software via an OPC-UA
protocol. The control loop demonstrated a very stable
glucose concentration, which was achieved along with
accurate measurements by the MarqMetrix All-In-One
Process Raman Analyzer. Automating the process, as
demonstrated, significantly reduced the intervention of the
operator, who typically must sample the bioreactor for offline
or at-line analyses. Because of this automation, the risk
of contamination and batch rejection relating to operator
error and other deviations is significantly reduced. In this
application note we demonstrated a culture environment that
can ensure maximum productivity in a reproducible manner
using an automated feedback control loop. With real-time
process monitoring by Thermo Fisher Scientific’s MarqMetrix
All-In-One Process Raman Analyzer with Lykos PAT Raman
Software and feedback control enabled by TruBio 6.0
Bioprocess Control Software, we have demonstrated a
real-world application of PAT and the ability to implement a
process developed with a quality-by-design approach.
References
1. Ghaderi D, Zhang M, Hurtado-Ziola N, et al. Production platforms for biotherapeutic
glycoproteins. Occurrence, impact, and challenges of non-human sialylation.
Biotechnol Genet Eng Rev. 2012; 28:147–75.
2. Durocher Y, Butler M. Expression systems for therapeutic glycoprotein production.
Curr Opin Biotechnol. 2009; 20:700–7.
3. Swiech K, Picanco-Castro V, Covas DT. Human cells: new platform for recombinant
therapeutic protein production. Protein Expr Purif. 2012; 84:147–53.
4. Research, C. for D. E. and. PAT — A Framework for Innovative
Pharmaceutical Development, Manufacturing, and Quality Assurance. U.S.
Food and Drug Administration. https://www.fda.gov/regulatory-information/
search-fda-guidance-documents/pat-framework-innovative-pharmaceuticaldevelopment-manufacturing-and-quality-assurance (accessed 2023-03-21).
5. Real time monitoring of multiple parameters in mammalian cell culture bioreactors
using an in-line Raman spectroscopy probe. Nicholas R Abu-Absi, Brian M Kenty,
Maryann Ehly Cuellar, Michael C Borys, Sivakesava Sakhamuri, David J Strachan,
Michael C Hausladen, Zheng Jian Li. Biotechnol Bioeng. 2011 May;108(5):1215-21.
doi: 10.1002/bit.23023. Epub 2010 Dec 22.
6. Quick generation of Raman spectroscopy based in-process glucose control to
influence biopharmaceutical protein product quality during mammalian cell culture.
Brandon N Berry , Terrence M Dobrowsky , Rebecca C Timson , Rashmi Kshirsagar,
Thomas Ryll, Kelly Wiltberger Biotechnol Prog 2016 Jan-Feb;32(1):224-34. doi:
10.1002/btpr.2205. Epub 2015 Dec 21.
7. Real-time metabolite monitoring using the Thermo Scientific™ Ramina™ Process
Analyzer System. Juan Villa, Mathew Zustiak, Elizabeth Amoako, David Kuntz, Lin
Zhang and Kevin Broadbelt (Thermo Fisher Scientific).
8. Real-time metabolite monitoring using the Thermo Scientific™ Ramina™ Process
Analyzer System and the Thermo Scientific™ 500L HyPerforma™ Dynadrive™
Single-Use Bioreactor (S.U.B.). Juan Villa, Mathew Zustiak, Elizabeth Amoako, David
Kuntz, Lin Zhang and Kevin Broadbelt (Thermo Fisher Scientific).
9. Lange, J et al., Frank Iwanitz, and Thomas Burke, OPC From Data Access to Unified
Architecture, 4th rev. Ed., OPC Foundation – Softing (VDE Verlag GMBH, 2010.
Learn more at thermofisher.com/marqmetrixAIO
For research use only. Not for use in diagnostic procedures. For current certifications, visit thermofisher.com/certifications
© 2024 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific
and its subsidiaries unless otherwise specified. MCS-AN1012-EN 2/24
Additional upstream
processing resources
Streamlining seed train scale-up through utilization of the
turndown ratio of the DynaDrive S.U.B.
Single-use technologies
Application note | DynaDrive Single-Use Bioreactors
Introduction
Upstream bioproduction has seen a substantial movement in the industry toward
single-use systems. This has been driven primarily by the need to reduce contamination
risk and cleaning requirements when compared to stainless steel systems, and to allow
for faster changeover of equipment between batches. At the same time, bioprocessing
manufacturing processes have matured significantly, and intensification of cell culture
processes has pushed the limits of these legacy single-use systems. In recent years,
Thermo Fisher Scientific has brought enhancements to the Thermo Scientific™
HyPerforma™ Single-Use Bioreactor (S.U.B.) platform with 5:1 S.U.B. and enhanced
S.U.B. options, which have allowed for higher turndown ratios and optimized mixing and
gassing strategies for intensified processes. Now Thermo Fisher Scientific has launched
a next-generation bioreactor for the biopharmaceutical industry with the Thermo
Scientific™ DynaDrive™ S.U.B. (Figure 1).
Keywords
DynaDrive S.U.B.,
single-use bioreactors,
scalability, turndown
Figure 1. DynaDrive S.U.B.s in 50 L, 500 L, 3,000 L, and 5,000 L sizes.
S.U.B. seed train options
During seed train expansion, cell cultures are sequentially
increased in volume and cell population to provide enough
starting material to enter batch, fed-batch, or perfusion
production unit operations. Small-scale cell cultures, usually
in less than 1 L of working volume, are typically maintained in
flasks on an orbital shaking platform. Shake flask culture requires
careful handling within a biosafety cabinet in a separate, higherclassification cleanroom to limit the risk of contamination. Due to
the heightened risk associated with these open-unit operations,
it is desirable for bioprocess engineers to move the cell culture
into a closed system as early as possible in the scale-up process.
A commonly chosen closed system for expansion steps after
shake flasks is the rocking-motion bioreactor. This single-use
closed system employs a platform to mix cells and gases to
maintain viable cultures. After expanding in the rocker, cultures
are transferred to stirred-tank S.U.B.s for further expansion prior
to inoculating the N-stage production vessel (Figure 2A).
Enhancements to our legacy S.U.B. systems, with the launch of
the 5:1 and the enhanced S.U.B. systems, allowed for working
volumes as low as 20% of the final working volume (e.g., 10 L in
a 50 L vessel), which has allowed for scale-up of the seed train
within the vessel and elimination of some seed train vessels
(Figure 2B). With the innovative design features of the 50 L
Building on our extensive experience and nearly two decades
of end users’ feedback, the DynaDrive S.U.B. employs a new
agitator drive technology with carefully engineered hardware that
enables exceptional performance. The DynaDrive S.U.B. allows
for a higher turndown ratio of at least 10:1 and up to 20:1 in the
larger sizes, with a reliable power input of up to 80 W/m³, and
scalability through each size.
The innovative impeller design enables a higher turndown ratio
than previously seen in single-use systems, offering working
volumes as low as 5 L in the 50 L S.U.B. These higher turndown
ratios open a new paradigm of what is possible with seed trains,
potentially eliminating multiple vessels and reducing logistical and
operating costs dramatically while increasing the efficiency of the
seed train through reduced connections and transfer losses.
DynaDrive S.U.B., cell cultures in working volumes as low as
5 L can be grown in a stirred-tank vessel, allowing for scale-up
processes to happen at even earlier stages. This extremely high
turndown ratio eliminates the need for rockers and extra steps
in the seed train process further on, with the ability to seed the
5,000 L DynaDrive S.U.B. at 20:1 directly from the 50 L scale.
This essentially means that the seed train process can be limited
to two reactors, with each having scale-up steps that take place
within them. For example, the 5 L minimum working volume
of the 50 L DynaDrive S.U.B. enables cell culture scale-up to
transfer directly from shake flasks to the S.U.B. After the 5 L
culture reaches a viable cell density suitable for expansion,
additional culture medium can be added to the vessel as a
second passage within the 50 L DynaDrive S.U.B. Once that
culture reaches a viable cell density suitable for expansion,
it can then be used to seed the 5,000 L DynaDrive S.U.B. at
a 250–500 L working volume, which subsequently can be
passaged within the reactor for inoculation of the production run
(Figure 2C).
Alternatively, because of the flexibility of the DynaDrive S.U.B.
systems, the 500 L DynaDrive S.U.B. can be used in the seed
train in place of the 5,000 L DynaDrive S.U.B., increasing the
required reactors to three, which allows for increased overall
bioreactor production and throughput. In this scenario, the seed
train would still utilize the 5 L minimum working volume of the
50 L DynaDrive S.U.B., but after reaching a viable cell density
suitable for expansion, these cells would be used to seed the
500 L DynaDrive S.U.B. at a 25 L or 50 L volume. When ready,
this would be expanded within the 500 L vessel and then
used to inoculate the 5,000 L DynaDrive S.U.B. production run
(Figure 2D).
Since every additional aseptic connection is a potential vector
for contamination, the high turndown ratios of the DynaDrive
S.U.B. systems enable risk reduction for bioprocess scale-up
operations. Performing expansions in the same vessel saves time
in setup and reduces the quantity of sterile connections needed
for the overall seed train expansion.
2
Figure 2. Improving facility efficiency with increased S.U.B. turndown ratios and careful logistical planning. Using bioreactors capable of
high turndown ratios, such as the DynaDrive S.U.B.s, can enable a streamlined seed train for cell expansion. Benefits of high turndown ratios include
increased facility space through reduction of bioprocess units, reduction of operator labor (e.g., setups, takedowns, and sterile or aseptic transfers),
and reduction of logistical risks. (A) Example of a 2:1 turndown ratio operation that incorporates a rocking-motion bioreactor and stirred-tank S.U.B.s.
(B) With the removal of at least one or more bioprocess units (e.g., rocker and S.U.B.), the cell expansion requires 20–25% less operator intervention
and up to 50% fewer single-use Thermo Scientific™ BioProcess Containers, compared to a 2:1 turndown ratio operation. The high-intensity portions
of operator labor (dark blue) of both the 5:1 and the DynaDrive S.U.B. seed trains are spaced at intervals of 2–4 days, reducing risk of operator error
through fatigue or haste. (C) Using the 5,000 L DynaDrive S.U.B. at a turndown ratio of 10:1 as the N-1 bioreactor allows for a smaller footprint in the
facility, less setup, and fewer sterile or aseptic transfers. (D) Alternatively, utilizing the 500 L DynaDrive S.U.B. in the seed train allows the 5,000 L
DynaDrive S.U.B. to remain a dedicated production vessel. Maximum split ratio = 10; minimum cell density = 0.3 x 10⁶ cells/mL.
Phase
Day 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15
Incubator
Rocker
Incubator
Incubator
Incubator
Final
production
phase
2,000 L
Final
production
phase
Final
production
phase
Fed-batch production phase
Fed-batch production phase
Fed-batch production phase
Fed-batch production phase
2,000 L
Seed
train
250 L
2:1 50 L
2:1
2:1 turndown maximum
S.U.B. count: 4
Seed-train cell expansion phase
Phase Seed-train cell expansion phase
Phase Seed-train cell expansion phase
Phase Seed-train cell expansion phase
DynaDrive 10:1 turndown
S.U.B. count: 3
Operator
labor 10:1
50 L
10:1
500 L
10:1
5,000 L
Final
production
phase
5,000 L
10:1
Seed train
50 L
10:1
DynaDrive 10:1 turndown
S.U.B. count: 2
Operator
labor 5:1
Operator
labor 10:1
50 L
5:1
5:1 turndown maximum
S.U.B. count: 3
Operator
labor 2:1
500 L
5:1
A
B
C
D
3
Application and benefits
By utilizing DynaDrive S.U.B.s in seed trains, not only are there
ideal turndown ratios of scaling within vessels, but the system
has been optimized to provide ideal mixing and mass transfer,
even at low turndown ratios, as shown in Table 1.
Also shown in Table 1 is a comparison of the monitoring
capabilities of each system. In comparing the single-use closed
system options for seed train expansion, stirred-tank S.U.B.s
allow for better monitoring and control than rocker systems.
Due to design limitations of the rocker bioreactors, there are
limited online process monitoring capabilities, whereas the
50 L DynaDrive S.U.B. can support temperature, pH, and DO
probes as well as an additional probe at the 10:1 turndown
ratio, with space for more probes at higher volumes (Table 1).
This capability provides greater design space for bioprocess
monitoring and enhanced process analytical technology (PAT)
capabilities for quality by design (QbD) approaches.
Table 1. Highlighted performance comparisons and monitoring capabilities of the HyPerforma, enhanced HyPerforma,
and DynaDrive S.U.B. systems.
S.U.B. Rocker HyPerforma Enhanced HyPerforma DynaDrive
Generation Rocker 2:1 5:1 Enhanced
fed-batch
Enhanced
perfusion DynaDrive
Vessel size 10 to 50 L 50 to 2,000 L 50 to 500 L 50 to 5,000 L
Scalable volumes 1 to 25 L 25 to 2,000 L 10 to 2,000 L 12.5 to 500 L 25 to 500 L 5 to 5,000 L
Turndown ratio 5:1 2:1 5:1 4:1 2:1 10:1 or 20:1
Sparge Overlay only Frit and drilled-hole sparger (DHS) Enhanced DHS
kLa at 20 W/m³
with DHS only No data ≤10 hr–1 ≤20 hr–1 ≤25 hr–1 20–30 hr–1
Max kLa No data ≤15 hr–1 30–40 hr–1 >40 hr–1
Max P/V No data 40 W/m³ 100 W/m³ 80 W/m³
Dissolved oxygen
(DO) sensor
Must be singleuse, limited
options available
Standard or single-use available, multiple options
pH sensor
Must be singleuse, limited
options available
Standard or single-use available, multiple options
Temperature
control
Electric,
heat only Jacket, temperature control unit (TCU)
Additional sensor
port options None available
2 probe belts,
no options
below first
probe belt
2 probe belts plus 1 DO and 1 pH option
at low turndown ratio
Additional
port options
at low
turndown
ratio
4
0
10
20
30
40
50
60
70
80
90
100
0
5
10
15
20
25
0 3 6 9 12 15 18
VCD (x 106 cells/mL)
Time (day)
50 L, VCD 500 L, VCD 5,000 L, VCD
50 L, viability 500 L, viability 5,000 L, viability
Viability (%)
Figure 3. VCD and viability of ExpiCHO-S cell culture from N-1
through N-stage production culture in the 50 L, 500 L, and
5,000 L DynaDrive S.U.B.s. Each N-1 culture was seeded at a 10:1
volume. The low viability seen in the N-1 culture in the 50 L S.U.B.
occurred in a prototype BPC with known effects on cell health at low
working volumes. Production BPCs have been shown to have reduced
impact on cell health and growth across multiple cell clones tested.
The following case study was done to evaluate the performance
of the DynaDrive S.U.B.s across all scales from 50 to 5,000 L
in a fed-batch process using CHO cells (Table 2), as well as to
evaluate the performance of the cells during the seed train steps
within these bioreactors.
Methods
Cells were expanded in shake flasks, the 50 L DynaDrive S.U.B.,
or both through the N-2 stage. The N-1 stage for each seed
train was carried out at a 10:1 turndown ratio within the N-stage
vessel. Fresh production medium was added to the S.U.B. after
3 days, resulting in the culture starting at the proper N-stage
production volume and initial seed density. Operating conditions
for both the N-1 and N-stage production vessels are described
in Table 3. During the production run, daily bolus feeds of 2X
concentrated EfficientFeed C+ AGT Supplement were added
from days 3 to 13 through either a subsurface (5,000 L S.U.B.)
or top feed (50 and 500 L S.U.B.) line. Following addition of
EfficientFeed C+ AGT Supplement, glucose was supplemented
in the same way on an as-needed basis after taking glucose
measurements to bring the final glucose concentration to
>4 g/L. Cell counts, viability, gases, nutrients, and metabolites
were measured offline daily. All three DynaDrive S.U.B. volumes
were tested in N-stage production runs. The 500 L and 5,000 L
production runs came from the same seed train, with the 500 L
N-1 being inoculated with cells from the 5,000 L N-1 stage
reactor, prior to the start of that 5,000 L N-stage production run.
Case study
Seed train evaluation using ExpiCHO-S cell line in 14-day fed-batch run
Table 2. Cell line evaluated in 50–5,000 L
DynaDrive S.U.B.s.
Cell type Gibco™ ExpiCHO-S™ Cells
Production medium Gibco™ ExpiCHO™ Stable Production Medium
Feed supplement Gibco™ EfficientFeed™ C+ AGT™ Supplement
Titer range ~3 g/L
Table 3. Operating parameters for evaluation of ExpiCHO-S cells in the 3 scales of DynaDrive S.U.B.s.
S.U.B. 50 L 500 L 5,000 L
Target starting volume 5 L, 35 L* 50 L, 350 L* 500 L, 3,500 L*
Seeding density 0.3 x 10⁶ cells/mL, 0.7 x 10⁶ cells/mL*
Temperature 37°C (N-1 and days 0–5), 34°C (days 5–14)
pH 6.8–7.2
pH control Acid control: sparged CO₂
Base control: 1 N NaOH
Acid control: sparged CO₂
Base control: 1 N NaOH
Acid control: sparged CO₂
through the macro DHS
Base control: 1 N NaOH
Agitation 140 rpm, 120 rpm* 60 rpm
26 rpm for N-1
37 rpm (days 0–3)
33 rpm (days 3–14)
DO 40%
Air crossflow/headspace (slpm) 1 6 10–20
DO cascade Air supplemented with
O₂ through DHS
Air supplemented with
O₂ through DHS
Air supplied to both
macro and micro DHS
O₂ supplemented
through micro DHS
Feeding strategy
Daily bolus of 1.05 L EfficientFeed
C+ AGT Supplement and glucose
(as needed)
Daily bolus of 10.5 L EfficientFeed
C+ AGT Supplement and glucose
(as needed)
Daily bolus of 105 L EfficientFeed
C+ AGT Supplement and glucose
(as needed)
* The N-1 culture conditions are listed first, followed by the N-stage culture conditions.
5
For Research or Further Manufacturing. Not for diagnostic use or direct administration into humans or animals.
© 2021, 2022 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific and its
subsidiaries unless otherwise specified. EXT3848 1022
Learn more at thermofisher.com/dynadrive
Results
Viable cell density (VCD) and viability (Figure 3) for the N-1 and
production cultures show consistent growth profiles among the
cultures, with similar cell density and viability trends at each step
in the seed train. The viability at the end of the run was above
80% in all systems, which was within expectations.
Conclusion
Overall, the ability of the DynaDrive S.U.B. systems to operate at
a 10:1 or 20:1 turndown ratio enables more efficient cleanroom
utilization, reduces risk of contamination, and simplifies seed
train expansion operations. This innovative product offering
from Thermo Fisher Scientific enables consolidation of unit
operations into fewer vessels and helps provide a more flexible
manufacturing system for upstream bioprocessing.
Application note | DynaDrive S.U.B.
Part 1: Intuitive bioprocess scale-up from
bench scale to pilot scale
A comparative study of Thermo Scientific single-use bioreactors
Introduction
The transition from bench scale to pilot scale is often viewed as a critical step in
bioproduction process development, generally because of physical differences
of the separate systems. The ability to generate scalable parameters is critical to
enable confidence when progressing to clinical and production scales. An example
of scaling up a CHO-based bioprocess from bench scale to pilot scale using the
Thermo Scientific™ DynaDrive™ Single-Use Bioreactor (S.U.B.) is outlined here,
highlighting similarities and differences in control parameters and outcomes. In general,
the culture carried out in the DynaDrive S.U.B. displayed key performance indicators that
matched or exceeded those of the bench-scale bioreactor, including IgG titer, specific
productivity, and cell density.
Keywords
Single-use bioreactor, DynaDrive,
CHO, fed-batch, scale-up,
Efficient-Pro, bench scale, pilot scale
Single-use bioprocessing
Methods
IgG-producing CHO-K1 cells were thawed and propagated
following standard procedures to establish a suitable
seed train. The reactors used in this study included the
3 L Thermo Scientific™ HyPerforma™ Glass Bioreactor, the
Thermo Scientific™ 5:1 HyPerforma™ S.U.B. (50 L), and the
DynaDrive™ S.U.B. (50 L). Cell expansion was performed in a
series of shake flasks until a sufficient number of cells were
generated to inoculate the bioreactors. Control parameters used
for the n-stage operation of each bioreactor are displayed in
Tables 1–3.
Table 1. Operation and control parameters.
Parameter
3 L
HyPerforma
Glass Bioreactor
50 L
HyPerforma
S.U.B.
50 L
DynaDrive
S.U.B.
n-1 stage seed
volume — 10 L 5 L
Target initial/
final volume 1.7/2 L 36/50 L 36/50 L
Seed density
(x10⁶ cells/mL) 0.3 0.3 0.3
Temperature
set point (°C) 37 37 37
Agitation (rpm) 350 183 105
Power input per
volume (W/m³) 100* 20 20
Tip speed
(m/sec) 1.01 1.06 0.59
Impeller
configuration
2 down-pumping
pitched-blade
impellers
1 down-pumping
pitched-blade
impeller
3 down-pumping
pitched-blade
impellers
Sparger
configuration
Drilled pipe
sparger
7 x 800 µm holes
Drilled-hole
sparger (DHS)
360 x 178 µm
pores
DHS
1,448 x 80 µm
pores
Target glucose
conc. (g/L) 3 3 3
Foam control
High-threshold
output 45 45 45
Foam alarm
delay (sec) 60 60 60
Splash delay
(sec) 5 5 5
* Approximate. Power number for the impeller configuration used has not been determined at time of
publication.
Table 2. Dissolved oxygen (DO) control gassing strategy.
Parameter
3 L
HyPerforma
Glass Bioreactor
50 L
HyPerforma
S.U.B.
50 L
DynaDrive S.U.B.
DO set point (%) 40 40 40
DO PID
Gain 0.07 0.10 0.10
Reset 200 200 200
O₂
Controller
output 15 100% 15 100% 15 100%
MFC scaling 0 0.5 slpm 0 5 slpm 0 3 slpm
N₂
Controller
output 0 40% 0 30% 0 30%
MFC scaling 0.15 0 slpm 1 0 slpm 1 0 slpm
Air
Overlay 0.2 slpm 5 slpm 5 slpm
Sparge
controller
output
— — 15 30 45%
MFC scaling — — 0 0.5 0 slpm
Table 3. pH control strategy.
Parameter
3 L
HyPerforma
Glass Bioreactor
50 L
HyPerforma
S.U.B.
50 L
DynaDrive
S.U.B.
pH set point 7.15 7.15 7.15
pH PID
Gain 0.04 0.04 0.04
Reset 200 200 200
pH deadband Not enabled Not enabled Not enabled
CO₂
Controller
output –100 0% –100 0% –100 0%
MFC scaling 0.08 0 slpm 2 0 slpm 2 0 slpm
Base Not enabled Not enabled Not enabled
2 Bioprocess scale-up from bench scale to pilot scale thermofisher.com/sut
Although a power input per volume (P/V) of 20 W/m³ was
targeted for the 50 L S.U.B.s, the P/V was approximately
100 W/m³ in the 3 L glass bioreactor culture. While the difference
in P/V between scales is large, this is not a significant concern
as P/V is frequently an impractical or unmeaningful scaling
parameter at the benchtop scale. In this case, an agitation rate
was selected for the 3 L glass bioreactor culture that provided for
tip speed comparable to that of the 50 L HyPerforma S.U.B.
Gibco™ Efficient-Pro™ medium was used throughout the
expansion and in the bioreactors for the n-stage production
process. Gibco™ Efficient-Pro™ Feed 1 was used to supplement
the production run cultures from day 3 onward, feeding 2.25%
of the current vessel volume daily (see Equation 1). A 2 M glucose
solution was also used to supplement the cultures, targeting a
Equation 1: Daily feed rate
Feed rate (mL/min) = (Current volume (L) ) ( ) ( ) ( ) 1,000 mL
1 L
0.0225
day
1 day
1,440 min
3 g/L glucose concentration upon starting the fed-batch phase
of the process. Gibco™ FoamAway™ Irradiated AOF Antifoaming
Agent was used to control excess foam during operation via the
use of the foam probe and pump triggered by Thermo Scientific™
TruBio™ automation software “Foam Hi Lim Out” as a remote
set point for the pump.
Reactors were sampled daily, and measurements were recorded
for cell count, cell size, cell viability, metabolites, and protein titer.
3 Bioprocess scale-up from bench scale to pilot scale thermofisher.com/sut 3
Results
Culture growth was similar among all reactors, with peak viable
cell densities (VCD) between 60.4 x 10⁶ cells/mL and 72.0 x 10⁶
cells/mL (Figure 1). Comparing cell viability, some discrepancies
between scales were apparent (Figure 2). The 50 L HyPerforma
and DynaDrive S.U.B. cultures had comparable cell viability
profiles, with the viability of the 3 L glass bioreactor culture
trending slightly higher throughout the latter half of the process.
Protein production was also similar among the conditions tested,
with peak protein titer reaching between 3.48 and 3.73 g/L
(Figure 3). Productivity behavior is further elucidated by evaluating
specific productivity (QP), which is shown in terms of pg/cell
per day (Figure 4). Excluding a couple of minor deviations, the
specific productivity lies within a similar range for each of the
vessels during the fed-batch portion of the process, with the
overall trend being similar across all conditions.
Figure 1. Viable cell density (VCD) profiles for CHO-K1 cells
in the 3 L glass bioreactor, 50 L HyPerforma S.U.B., and 50 L
DynaDrive S.U.B.
0
10
20
30
40
50
60
70
80
0 2 4 6 8 10 12 14
VCD (x 106 cells/mL)
Day
3 L HyPerforma Glass Bioreactor 50 L HyPerforma S.U.B. 50 L DynaDrive S.U.B.
Figure 2. Viability profiles for CHO-K1 cells in the 3 L glass
bioreactor, 50 L HyPerforma S.U.B., and 50 L DynaDrive S.U.B.
0
10
20
30
40
50
60
70
80
90
100
Viability (%)
0 2 4 6 8 10 12 14
Day
Lorem ipsum 3 L HyPerforma Glass Bioreactor 50 L HyPerforma S.U.B. 50 L DynaDrive S.U.B.
Figure 3. IgG titer profiles for CHO-K1 cells in the 3 L glass
bioreactor, 50 L HyPerforma S.U.B., and 50 L DynaDrive S.U.B.
0 2 4 6 8 10 12 14
Day
Lorem ipsum 3 L HyPerforma Glass Bioreactor 50 L HyPerforma S.U.B. 50 L DynaDrive S.U.B.
4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0.0
lgG titer (g/L)
Figure 4. Specific productivity (QP, a 3-point moving average)
profiles for CHO-K1 cells in the 3 L glass bioreactor, 50 L
HyPerforma S.U.B., and 50 L DynaDrive S.U.B. QP is expressed in
terms of picograms IgG per cell per day.
2
4
0
6
8
10
12
14
0 2 4 6 8 10 12 14
QP (pg/cell per day)
Day
3 L HyPerforma Glass Bioreactor 50 L HyPerforma S.U.B. 50 L DynaDrive S.U.B
4 Bioprocess scale-up from bench scale to pilot scale thermofisher.com/sut
The lactate concentration in each reactor increased over the
first few days, with a primary peak on day 4, after which the
concentration dropped for 4–6 days before increasing again until
process termination (Figure 5). While the lactate concentration
in the 3 L culture was lower than that of both 50 L cultures
during the primary peak, the lactate accumulation rate was
higher in the vessel from day 10 and beyond, resulting in a
higher final concentration. The relatively larger increase in lactate
concentration in the 3 L glass bioreactor culture coincided with
a drop in pCO₂ near the end of the process, falling as low as
~25 mmHg, whereas pCO₂ in both 50 L cultures was maintained
at 40–80 mmHg from day 3 onward.
Scaled total gas flow rates (vessel volume per minute, VVM)
show a lower O₂ sparge rate in the glass reactor and the
DynaDrive S.U.B. compared to the HyPerforma S.U.B.
(Figure 6). Nitrogen flows are also shown in the figure, and
any sparged air flow rates were treated as 21% O₂ and 78% N₂.
A direct comparison of the gassing requirements of the glass
bioreactor and S.U.B.s is somewhat difficult due to the relatively
high P/V in the glass reactor and varying rates of sparged N₂
in each condition. Even so, it can be seen from the similarity in
scaled flow rates that the 50 L DynaDrive S.U.B. serves as a
straightforward option when considering scalability from bench
to pilot scale. Inspection of the CO₂ sparge rates (Figure 7) and
referring to Figure 5 show that acceptable pCO₂ concentrations
were maintained in the 50 L cultures even while the CO₂ flow rate
was insignificant.
Figure 5. Lactate and pCO₂ profiles for CHO-K1 cells in the 3 L glass
bioreactor, 50 L HyPerforma S.U.B., and 50 L DynaDrive S.U.B.
0 2 4 6 8 10 12 14
Day
Lorem ipsum 3 L HyPerforma
Glass Bioreactor lactate
50 L HyPerforma
S.U.B. lactate
50 L DynaDrive
S.U.B. lactate
50 L DynaDrive
S.U.B. pCO2
3 L HyPerforma
Glass Bioreactor pCO2
50 L HyPerforma
S.U.B. pCO2
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
1.6
1.8
Lactate (g/L)
0
20
40
60
80
100
120
140
160
pCO2 (mmHg)
Figure 6. Oxygen and nitrogen gas flow through the drilled-hole
sparger for each vessel in terms of vessel volume per minute.
2 L HyPerforma
Glass Bioreactor O2
50 L HyPerforma
S.U.B. O2
50 L DynaDrive
S.U.B. O2
0 2 4 6 8 10 12 14
Day
Lorem ipsum 3 L HyPerforma
Glass Bioreactor O2
50 L HyPerforma
S.U.B. O2
50 L DynaDrive
S.U.B. O2
3 L HyPerforma
Glass Bioreactor N2
50 L HyPerforma
S.U.B. N2
50 L DynaDrive
S.U.B. N2
0.00
0.01
0.02
0.03
0.04
0.05
0.06
0.07
0.08
Gas flow (VVM)
Figure 7. pH profile and carbon dioxide gas flow through the
drilled-hole sparger in terms of vessel volume per minute.
0 2 4 6 8 10 12 14
Day
Lorem ipsum 3 L HyPerforma
Glass Bioreactor pH
50 L HyPerforma
S.U.B. pH
50 L DynaDrive
S.U.B. pH
3 L HyPerforma
Glass Bioreactor CO2
50 L HyPerforma
S.U.B. CO2
50 L DynaDrive
S.U.B. CO2
0.025
0.020
0.015
0.010
0.005
0.000
CO2 flow rate (VVM)
7.30
7.25
7.20
7.15
7.10
7.05
7.00
6.95
6.90
6.85
6.80
pH
5 Bioprocess scale-up from bench scale to pilot scale thermofisher.com/sut 5
Totals for feeds and supplements at the time of run termination
are provided in Table 4. Antifoam requirements are significantly
lower in the 3 L culture compared to the 50 L cultures due to
much smaller sparged gas flow rates, larger pore sizes in the
drilled-hole sparger, and the foam sensor's proximity to the liquid
level in the smaller culture.
Table 4. Total feed and supplement (from calibrated
pump totalizer) added by the end of process.
Parameter
3 L
HyPerforma
Glass Bioreactor
50 L
HyPerforma
S.U.B.
50 L
DynaDrive
S.U.B.
Total feed 442 mL 10.0 L 9.9 L
Total 2 M
glucose solution 160 mL 3.2 L 2.6 L
Total FoamAway
agent 4.7 mL 350 mL 280 mL
6 Bioprocess scale-up from bench scale to pilot scale thermofisher.com/sut
For Research Use or Further Manufacturing. Not for diagnostic use or direct administration into humans or animals.
© 2024 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific and
its subsidiaries unless otherwise specified. COL028238 0724
Learn more at thermofisher.com/sut
Author
Jace Parkinson, Engineer II, Systems Design, Thermo Fisher Scientific, Logan, UT
Discussion and conclusion
Considerations for scale-up of cell culture processes frequently
center around agitation and gassing strategies. The control
parameters used in this study were found to produce
comparable results when scaling from benchtop glass
bioreactors to Thermo Scientific™ S.U.B.s at the 50 L scale.
Cell growth profiles, metabolism, and productivity were
maintained in similar ranges in each of the bioreactors. While
an aggressive CHO-K1 clone was used, with VCD reaching
65 x 10⁶ cells/mL in the DynaDrive S.U.B., the cell cultures were
easily maintained within the design constraints of the reactors,
with mixing and gassing demands being met in the glass
bioreactor and the S.U.B.s without difficulty. The final protein
titer at the time of harvest was 3.48 g/L in the glass bioreactor
and 3.73 g/L in the DynaDrive S.U.B., with productivity being
maintained during the duration of the culture.
Ordering information
Product Cat. No.
Efficient-Pro AGT Medium A5322303
Efficient-Pro AGT Feed 1 A5209102
FoamAway Irradiated AOF Antifoaming Agent A1036902
HyPerforma G3 Lab Controller F100-2695-001
HyPerforma G3 Lite Controller F100-2701-001
HyPerforma G3 Pro Controller F100-2961-001
HyPerforma Glass Bioreactor (3 L) F100-2680-002
HyPerforma 5:1 Single-Use Bioreactor (50 L) SUB0050.9500
Bioprocess Container for HyPerforma 5:1 S.U.B. (50 L) SH31073.01
DynaDrive Single-Use Bioreactor (50 L) DDB0050.1011
DynaDrive Bioprocess Container (50 L) SH31192.01
Scale-up evaluation of the DynaDrive S.U.B.s
Part 1: ExpiCHO-S Cells and CHO-S Cells (cGMP banked)
Single-use technologies
Application note | DynaDrive S.U.B.
Introduction
As a molecule approaches commercial launch and more is known about the potential
market demand, companies are often faced with the decision to scale up or scale out
their manufacturing processes. Generally, when scale-up production vessels of more
than 2,000 L are required, this decision also involves moving from single-use bioreactors
(S.U.B.s) to traditional stainless-steel systems.
Additionally, more recently developed process intensification methods have allowed
manufacturers increased product output, pushing titers past 10 g/L in some cases.
These output achievements require increased production efficiency and input, pushing
many bioreactor systems past their limits. S.U.B. quality requirements, robustness, and
functional performance can all become constraints, especially at scales up to 2,000 L.
For example, as oxygen transfer rate (OTR) becomes a limiting factor, most traditional
S.U.B.s rely primarily on increased sparging flow to increase oxygen mass transfer.
Keywords
Single-use bioreactor,
DynaDrive S.U.B.,
scalability, fed-batch
Maintaining a dissolved oxygen (DO) target in high-demand
cell cultures can be increasingly difficult due to limitations in
the amount of mixing power that can be distributed effectively
through the drivetrain of traditional S.U.B.s. Sparging through
a micro-sparger has become a widely used strategy to improve
OTR in traditional S.U.B.s and typically requires a secondary
sparger to facilitate removal or stripping of dissolved CO₂
(measured as the partial pressure of CO₂, or pCO₂). Some cell
lines, however, are sensitive to the higher shear produced by
micro-sparging, and process scale-up cannot depend on this
method alone to ensure sufficient O₂ delivery or CO₂ removal.
A next generation of S.U.B., the Thermo Scientific™ DynaDrive™
S.U.B., with vastly improved mixing and mass transfer
performance, is now enabling scale-up to 5,000 L and process
intensification. Previous limits are no longer a burden for the
DynaDrive S.U.B., and it continues to leverage known and
acknowledged benefits of legacy units. DynaDrive S.U.B.s are
multifunction reactors for a range of applications, including
intermediate-scale production of preclinical, clinical, and
commercial material, as well as perfusion for both production
and N-1 seed processes. Additionally, each DynaDrive S.U.B.
includes features that are improved over legacy and alternative
S.U.B. options:
• Each system is equipped with a Thermo Scientific™
BioProcess Container (BPC) load assist device, reducing
handling and setup time, increasing safety, and providing
consistent BPC loading. BPC loading can be accomplished
in less time at the 50 L and 500 L scales compared to legacy
S.U.B.s, and in less than 45 minutes at the 3,000 L and
5,000 L scales.
• Best-in-class enhanced drilled-hole sparger (DHS) provides
repeatable and reliable performance that users of S.U.B.s
have embraced due to its linear scale-up benefits.
• Revolutionary drivetrain design with multiple impellers allows
increased power input and efficiency while offering reduced
shear rates.
• Cuboid design contributes to better BPC fit and increased
baffled-like mixing efficiency, and allows more productive use
of facility footprint.
• 10:1 or better turndown ratio reduces facility requirements
and investment costs while increasing flexibility in seed train
applications and all aspects of scale-up.
• Continuous mixing during harvest and minimal hold-up
volume (<1%) after drain.
• Improved exhaust system for the 3,000 L and 5,000 L
S.U.B.s, allowing for increased gas flow rates and utmost
reliability typically required for production-scale cultures.
These major design changes have enabled a power-to-volume
(P/V) ratio of up to 80 W/m³ in all sizes, t95 mixing times of less
than 60 sec, and kLa performance of at least 40 hr–1 at all scales
(Table 1).
Additionally, the DynaDrive S.U.B. allows for process scale-up
and transfer from legacy S.U.B.s, offering benefits of consistent
BPC film, assurance of supply, robust quality controls, BPC
integrity, and industry-leading BPC customization options. End
users can continue using previously qualified traditional and
single-use sensing options as well as inlet and exhaust filters and
other peripheral components integrated through high-strength
porting and line sets.
Goal
The goal of this study was to evaluate the performance of the
DynaDrive S.U.B. across 50 L–5,000 L scales using two different
cell lines (Table 2) together with previously developed processes
specific to those cell lines for manufacturing up to a 2,000 L
scale. These experiments were designed to demonstrate that the
DynaDrive S.U.B. could be successfully implemented for use with
multiple cell lines across scales with standard scale-up criteria.
Both cell lines were subjected to a 14-day fed-batch run at full
working volume for each scale.
Table 1. Comparison of DynaDrive S.U.B. capabilities.
Parameter 50 L S.U.B. 500 L S.U.B. 5,000 L S.U.B.
Maximum
volume 50 L 500 L 5,000 L
Turndown ratio 10:1 20:1 20:1
kLa >50 hr–1 >50 hr–1 40 hr–1
t
95 mixing times <30 sec <40 sec <60 sec
Maximum
P/V ratio 80 W/m³ 80 W/m³ 80 W/m³
Table 2. Cell lines evaluated in 50 L–5,000 L
DynaDrive S.U.B.s.
Parameter Cell line 1 Cell line 2
Cell type Gibco™ ExpiCHO-S™
Cells
Gibco™ CHO-S™ Cells
(cGMP banked; part of
the Gibco™ Freedom™
CHO-S™ Kit)
Production
medium
Gibco™ ExpiCHO™
Stable Production
Medium (SPM)
Gibco™ Dynamis™
Medium
Feed
supplement Gibco™ EfficientFeed™ C+ AGT™ Supplement
Titer range Medium: ~3 g/L Low: ~1 g/L
Cell line
characteristics Platform cell line Legacy cell line
Case study 1 2
2 DynaDrive S.U.B. thermofisher.com/dynadrive DynaDrive S.U.B. thermofisher.com/dynadrive
Case study 1
Scale evaluation using ExpiCHO-S Cells in a 14-day fed-batch run
Methods
Cells were expanded in shake flasks or pilot-scale S.U.B.s
through the N-2 stage. The N-1 stage for each seed train
was performed at a 10:1 turndown ratio. Fresh production
medium was added to the S.U.B. after 3 days, resulting in
the culture starting at proper N-stage production volume and
initial seed density. Operating conditions are described in
Table 3. Daily bolus feeds of 2X concentrated EfficientFeed
C+ AGT Supplement were added from day 3 to 13 via either
a subsurface (5,000 L S.U.B.) or top feed line (50 L and 500 L
S.U.B.s). Glucose was supplemented in the same manner on an
as-needed basis after taking a glucose measurement following
addition of EfficientFeed C+ AGT Supplement to bring the final
glucose concentration to >4 g/L. Cell counts, viability, dissolved
gases, nutrients, and metabolites were measured offline daily.
Titer samples were filtered and frozen daily starting on day 6 for
batch testing at the culmination of the run.
Table 3. Operating parameters for evaluation of ExpiCHO-S Cells in 3 scales of DynaDrive S.U.B.s.
S.U.B. 50 L 500 L 5,000 L
Target starting volume 35 L 350 L 3,500 L
Seeding density 0.7 x 10⁶ cells/mL
Temperature 37°C (days 0–5)
34°C (days 5–14)
pH 6.8–7.2
pH control Acid control: sparged CO₂
Base control: 1 N NaOH
Acid control: sparged CO₂
Base control: 1 N NaOH
Acid control: sparged CO₂
through macro DHS
Base control: 1 N NaOH
Agitation 120 rpm 60 rpm 26 rpm (days 0–3)
33 rpm (days 3–14)
DO 40%
Air headspace 1 slpm 6 slpm 10–20 slpm
DO cascade Air supplemented with O₂
through DHS
Air supplemented with O₂
through DHS
Air supplied to both macro and
micro DHS; O₂ supplemented
through micro DHS
Feeding strategy
Daily bolus of 1.05 L EfficientFeed
C+ AGT Supplement, and glucose
(as needed)
Daily bolus of 10.5 L EfficientFeed
C+ AGT Supplement, and glucose
(as needed)
Daily bolus of 105 L EfficientFeed
C+ AGT Supplement, and glucose
(as needed)
Figure 1. VCD and viability comparison of ExpiCHO-S Cells in the
3 DynaDrive S.U.B.s.
0
10
20
30
40
50
60
70
80
90
100
0
5
10
15
20
25
0 2 4 6 8 10 12 14
Viability (%)
VCD (x 106 cells/mL)
Time (day)
50 L, VCD 500 L, VCD 5,000 L, VCD
50 L, viability 500 L, viability 5,000 L, viability
Figure 2. Titer results for ExpiCHO-S Cells in the 3 DynaDrive
S.U.B.s.
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
0 2 4 6 8 10 12 14
IgG (g/L)
Time (day)
50 L 500 L 5,000 L
DynaDrive S.U.B. thermofisher.com/dynadrive 3
Figure 3. pCO₂ profile measurements for ExpiCHO-S Cells in the
3 DynaDrive S.U.B.s.
0
20
40
60
80
100
120
0 2 4 6 8 10 12 14
pCO2 (mm Hg)
Time (day)
50 L 500 L 5,000 L
Figure 4. Lactate and NH₄⁺ profiles for ExpiCHO-S Cells in the 3
DynaDrive S.U.B.s.
0
0.5
1
1.5
2
2.5
3
3.5
4
4.5
5
0
1
2
3
4
5
6
7
8
9
10
0 2 4 6 8 10 12 14
NH4+ (mM)
Lactate (g/L)
Time (day)
50 L, lactate 500 L, lactate 5,000 L, lactate
50 L, NH4+ 500 L, NH4+ 5,000 L, NH4+
Figure 5. DHS gas flow rates for ExpiCHO-S Cells in the 3
DynaDrive S.U.B.s.
0.000
0.005
0.010
0.015
0.020
0.025
0.030
0.035
0.040
0.045
0 2 4 6 8 10 12 14
DHS gas flow (VVM)
Time (day)
5,000 L, O2 VVM
5,000 L, micro DHS, air VVM
500 L, O2 VVM
500 L, air VVM
50 L, O2 VVM
50 L, air VVM
5,000 L, macro DHS, air VVM
Results
Viable cell density (VCD) and viability for the cultures show
consistent growth profiles among the cultures, with similar cell
density and viability trends (Figure 1). Peak VCDs were similar at
~20 x 10⁶ cells/mL, and the end-of-run viability was above 80%
in all systems. IgG concentration measured 2.7–3.0 g/L on day
14 and is within ranges observed historically (Figure 2). While CO₂
levels in the 5,000 L culture were within expected conditions,
levels in the 50 L and 500 L cultures were slightly lower than
anticipated, indicating possible over-stripping of CO₂ (Figure 3).
Metabolite data collected offline indicated healthy cultures with
maintained glucose and low levels of metabolic byproducts,
including lactate and ammonium, staying within ranges observed
historically (Figure 4). Minimum glucose concentrations were
maintained according to the culture protocol by feeding with
EfficientFeed C+ AGT Supplement daily, and a glucose solution
when needed.
The gas flow rates were controlled based on culture oxygen
demand (Figure 5). For the 50 L and 500 L cultures, DO was
maintained by first sparging air, then supplementing with O₂
through the single DHS. For the 5,000 L culture, DO was
maintained by sparging air through the macro DHS at a constant
flow rate and sparging an air–O₂ mix through the micro DHS.
Using these strategies, DO was maintained at 40 ± 5% with no
major fluctuations during the duration of the run. Importantly, gas
flow requirements for the cultures remained very low at all scales,
never reaching past 0.035 vessel volumes per minute (VVM) for
either the 50 L or 500 L S.U.B. For the 5,000 L S.U.B., gas flow
requirements were below 0.01 VVM for the micro DHS with a
constant 0.005 VVM for the macro DHS. These low gas flow
rates in conjunction with the relatively low power inputs used at
low rpm represent only about 30% of the available performance
capacity of the system. The sparge strategy of the 5,000 L S.U.B.
was adjusted on days 3–6 to successfully keep pCO₂ levels
below 80 mm Hg for the balance of the cell culture run.
4 DynaDrive S.U.B. thermofisher.com/dynadrive DynaDrive S.U.B. thermofisher.com/dynadrive
Case study 2
Scale evaluation using CHO-S Cells (cGMP banked) in a 14-day fed-batch run
Methods
Cells were expanded in shake flasks or pilot-scale S.U.B.s until
seeding into the production vessels at either a 10:1 or 20:1
turndown ratio. The 50 L and 500 L S.U.B.s were brought to full
volume with fresh production medium after 1–2 days, while the
5,000 L S.U.B. was filled to a 5:1 turndown ratio after 3 days
and then full production volume after another day of growth.
Operating conditions for each bioreactor are described in Table 4.
A 2X concentration of EfficientFeed C+ AGT Supplement was
added continuously from day 3 to 11 through either a subsurface
(5,000 L S.U.B.) or top feed line (50 L and 500 L S.U.B.s). Glucose
was supplemented in a continuous drip as needed depending on
culture demands, to maintain glucose concentrations of 1–3 g/L.
Cell counts, viability, dissolved gases, metabolites, and nutrients
were measured offline daily. Titer samples were filtered and
frozen daily starting on day 6. Titer samples were batch tested at
the end of the run.
Results
VCD and viability for the cultures show similar growth profiles
among the cultures, with similar peak VCD and viability trends
(Figure 6). The 5,000 L culture exhibited slightly slower growth
and lower peak VCD compared with the 500 L culture (VCD of
23 x 10⁶ and 26 x 10⁶ cells/mL, respectively) but was still within
expected ranges. Productivity throughout the run was within
expected ranges (Figure 7). The 50 L culture exhibited the lowest
harvest titer, likely due to low pCO₂ in the latter half of the run.
Metabolites and gas flow demands were similar in concentration
and magnitude compared to the ExpiCHO-S cell cultures.
Glucose was maintained in the S.U.B.s at around 1–5 g/L,
while lactate and ammonium concentrations were within ranges
observed historically (data not shown).
Table 4. Operating parameters for evaluation of CHO-S Cells in 3 scales of DynaDrive S.U.B.s.
S.U.B. 50 L 500 L 5,000 L
Target starting volume 42.5 L 425 L 4,250 L
Seeding density 0.3 x 10⁶ cells/mL
Temperature 37°C
pH 6.8–7.2
pH control Acid control: sparged CO₂
Base control: not applicable
Acid control: sparged CO₂
Base control: not applicable
Acid control: sparged CO₂
through macro DHS
Base control: not applicable
Agitation 120 rpm 68 rpm 31–41 rpm
DO 30%
Air headspace 3 slpm 6 slpm 10–20 slpm
DO cascade N₂ and O₂ through the DHS N₂ and O₂ through the DHS N₂ and O₂ through the micro DHS
Feeding strategy
7.5 L of EfficientFeed C+ AGT
Supplement added on continuous drip
from day 3 to 11, and glucose as needed
75 L of EfficientFeed C+ AGT Supplement
added on continuous drip from day 3 to
11, and glucose as needed
750 L of EfficientFeed C+ AGT
Supplement added on continuous drip
from day 3 to 11, and glucose as needed
Figure 6. VCD and viability comparison of CHO-S Cells in the 3
DynaDrive S.U.B.s.
0
10
20
30
40
50
60
70
80
90
100
0
5
10
15
20
25
30
35
40
45
50
0 2 4 6 8 10 12 14
Viability (%)
VCD (x 106 cells/mL)
Time (day)
50 L, VCD 500 L, VCD 5,000 L, VCD
50 L, viability 500 L, viability 5,000 L, viability
Figure 7. Titer results for CHO-S Cells in the 3 DynaDrive S.U.B.s.
50 L 500 L 5,000 L
1.2
1.0
0.8
0.6
0.4
0.2
0.0 0 2 4 6 8 10 12 14
lgG (g/L)
Time (day)
DynaDrive S.U.B. thermofisher.com/dynadrive 5
Discussion
As cell cultures are scaled to large-volume S.U.B.s, several
factors can become more difficult to manage and control. Often,
decisions related to culture parameters and operation thresholds
must be balanced to provide the culture with the best opportunity
for success. The DynaDrive S.U.B. assists with ensuring culture
success due to specifically designed aspects of the S.U.B.,
including the geometrically scalable agitator drivetrain and the
uniquely linear sparge performance of the DHS, which both
directly benefit mass transfer and mixing abilities.
The agitator drivetrain of the DynaDrive S.U.B. with multiple
impellers at optimal locations allows for optimal and scalable
power input. The cultures performed in these case studies
were operated at a power input of approximately 20 W/m³, far
below the maximum recommended 80 W/m³. Mass transfer in
this design is more proportional to the power input of a system
due to the mixer’s ability to both disperse and retain sparged
gas. Therefore, if a culture were to demand more mass transfer,
increased mixer speeds above those tested here would be an
option for improved performance.
The gas flow rates observed in these studies were far below the
maximum rated gas flow rates for each system. As with power
input for each system, mass transfer is shown to be linearly
proportional and scalable to gas flow rate in the DynaDrive
S.U.B.s. For the 50 L and 500 L ExpiCHO-S cell cultures, total
gas flow rate through the DHS was never higher than 0.035
VVM, less than 25% of the maximum 0.15 VVM limit. For the
5,000 L ExpiCHO-S cell culture, total gas flow rate through the
micro DHS reached only 0.007 VVM (30 slpm) for most of the
culture. Additionally, the 5,000 L culture used a minimal macro
DHS flow rate of only 15 slpm of air to maintain CO₂ stripping
and to provide additional O₂ mass transfer. These chosen gas
flow rates resulted in easily maintained O₂ mass transfer to
support cultures of light to moderate demand with cell densities
at 20–30 x 10⁶ cells/mL. More demanding clones are expected
to require higher gas flow rates or agitation but still remain within
design limits of the DynaDrive S.U.B.s. It is important to note that
the DHSs were designed with specific pore quantities and sizes
to provide these high levels of mass transfer at low gas flow rates,
removing the need for other sparger types such as the legacy
sintered sparger.
One of the biggest concerns when scaling to larger S.U.B. sizes
is the ability to control dissolved CO₂ concentrations within ideal
physiological ranges (60–100 mm Hg). While spargers in smaller
S.U.B.s typically provide sufficient CO₂ stripping capability due to
their often oversized bubbles, shorter liquid column heights, and
favorable overlay surface-to-volume ratios, spargers for larger
S.U.B.s can be limited in providing sufficient CO₂ stripping while
maintaining required O₂ mass transfer at reasonable gas flow
rates. The 5,000 L DynaDrive S.U.B. is equipped with 3 separate
DHSs to provide optimal gassing to drive mass transfer for both
gases of interest. The two larger macro DHSs provide an amount
of O₂ mass transfer while also providing more CO₂ stripping
capability due to the larger bubbles created, while the single
micro DHS provides higher O₂ mass transfer due to the smaller
bubbles created. Balancing these spargers in tandem with
agitation in these cultures allowed pCO₂ to remain within desired
ranges for each culture, thus providing a very generous operating
design window in anticipation of future process intensification
that may be requested by the end user.
Culture conditions in a S.U.B. are highly variable and must be
balanced with multiple inputs and outputs. Through experience
and tracking online and offline readings, setpoints can be
balanced for gas flow rates, agitation, pH, DO, and feed flow
rates. Generally speaking, metabolite buildup such as lactate and
respired CO₂ lead to acidic conditions in the reactor that require
the addition of base to balance the culture pH. Additionally,
especially in larger vessels, buildup of pCO₂ can be detrimental to
cell health. The S.U.B.s in this study were able to maintain pCO₂
and pH conditions through the employed gassing strategies,
leading to pCO₂ levels maintained within physiological conditions.
The 50 L and 500 L cultures actually showed a propensity to
exhibit too much CO₂ stripping. While the pH in each culture
was balanced sufficiently, more optimal gassing and pH control
strategies could be employed in the future to provide more
optimal growth and production conditions.
Finally, each reactor was seeded at a low working volume: 10:1
turndown ratio in the 50 L S.U.B. and 20:1 turndown ratio in the
500 L and 5,000 L S.U.B.s. This has been shown to reduce the
complexity of seed train requirements by eliminating intermediate
vessels, consumables, and operation space. While using a
low turndown ratio in a production vessel such as the 5,000 L
DynaDrive S.U.B. is not ideal for some commercial applications
seeking to maximize daily productivity, this feature allows for
flexibility in manufacturing spaces not previously available.
6 DynaDrive S.U.B. thermofisher.com/dynadrive DynaDrive S.U.B. thermofisher.com/dynadrive
For Research Use or Further Manufacturing. Not for diagnostic use or direct administration into humans or animals.
© 2021, 2022 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific and its
subsidiaries unless otherwise specified. EXT3458 0822
Learn more at thermofisher.com/dynadrive
Authors: Jordan Cobia, Product Manager;
Ben Madsen, R&D Manager
Conclusions
The DynaDrive S.U.B.s were able to support both clones in this
study and provide controlled conditions to achieve target VCDs
and titers while maintaining high viability. This was achieved by
choosing simple scale-up parameters among the S.U.B. sizes,
including consistent power input and gas flow rates. The low gas
flow rates and agitation rates tested provided sufficient mass
transfer to maintain DO setpoints while maintaining pCO₂ levels at
or below maximum limits. Additionally, the ability of each S.U.B.
to be seeded at low volume allowed reduction in seed train
complexity by using the S.U.B. at the N-1 culture stage. Fewer
manipulations, media preparations, and fluid transfers, and less
overall consumption of resources have the potential to noticeably
mitigate risk, reduce waste, and lower operating costs.
Overall, the different scale-up processes demonstrated in these
studies show the versatility of the DynaDrive S.U.B. to maintain
culture setpoints compared to historical processes with simple
scale-up criteria. The modified drivetrain and sparging, when
compared to legacy S.U.B. products, did not have adverse
effects on the cell cultures, thus enabling a much larger
design space for process development and production than
previously available. Additionally, the DynaDrive S.U.B. provides
consistent, scalable performance from 50 L to 5,000 L. With
this demonstrated consistency, the DynaDrive S.U.B. will enable
users to scale up their process with minimal process changes to
meet demand for commercial therapeutics.
Raman spectroscopy
Application note
MarqMetrix All-In-One Process Raman Analyzer –
Chemometric model transferability across instruments
Summary
• The Thermo Scientific™ MarqMetrix™ All-In-One Process Raman Analyzer represents a
breakthrough in process analytical technology (PAT) by providing in-line, real-time and
actionable monitoring of multiple analytes in complex systems.
• This paper demonstrates the successful transferability of one chemometric model
across 10 different MarqMetrix All-In-One Process Raman Analyzers while maintaining
an average prediction error of 0.21 g/L, 0.16 g/L and 0.3 g/L for glucose, glutamine, and
lactate, respectively in a mixture.
• Measurement accuracy and precision is maintained when applying a chemometric
model across multiple instruments, ensuring users do not have to spend time or
resources to re-build a model for a new MarqMetrix All-In-One Process Raman Analyzer
or new autoclavable probe.
Introduction
The MarqMetrix All-In-One Process Raman Analyzer is a Raman spectroscopy instrument
designed to offer rapid, robust, scalable, and reliable identification, quantification, and
characterization of molecules during any phase of R&D process development. It is an
“all-in-one” instrument utilizing easily exchangeable, autoclavable probes to meet the
various analytical needs for, but not limited to, upstream bioprocess monitoring, or
characterization of fill and finish products.
Chemometric analysis allows users to develop a data analysis model to monitor the
concentration of multiple analytes from their analyzer. There is a significant investment
made from a time and resource perspective to build an accurate and robust chemometric
model. As a result, it is imperative for a chemometric model to be used between
instruments to fully leverage the value of this investment. Once a chemometric model
is developed, it can be used with any MarqMetrix All-In-One Process Raman Analyzer
Authors
Mike Bates, Nimesh Khadka, David Kuntz,
Lin Zhang, Sue Woods
to monitor multiple reactors in parallel with high fidelity of model
accuracy. The measurement accuracy and precision maintained
when transferring chemometric models across different system
hardware ensures that customers do not have to re-build a model
when using a new instrument or new autoclavable probe.
Experimental set up
To demonstrate the transferability of a complex chemometric model,
we evaluated three relevant analytes: glucose, glutamine and
lactate in Gibco™ DMEM growth media. All three analytes occur in
bioreactors in the g/L concentration ranges with glucose occurring
in the range of 0-12 g/L, glutamine 0-2.5 g/L and lactate 0-20 g/L.
A chemometric model was developed by collecting spectra from
a set of calibration standards using one MarqMetrix All-In-One
Process Raman Analyzer. Within the relevant concentration ranges
for each analyte, 24 samples with randomized concentrations were
selected using the uniform design method2
. The model was then
applied to spectra from a different set of 8 validation samples,
measured using 10 unique MarqMetrix All-In-One Process Raman
Analyzers. The acquisition parameters were optimized to maximize
the signal-to-noise ratio of the spectral features corresponding to
the analyte concentrations. Optimized parameters were 15 second
integration time, 450 mW laser power and 10 replicate on-board
signal averaging with automatic dark correction. These acquisition
parameters resulted in a sample collection period of about 5
minutes. The model was able to predict the concentration of all
three analytes in the 8-sample validation set with a high degree of
accuracy and precision.
Each MarqMetrix All-In-One Process Raman Analyzer evaluated
in this study included a unique hardware set comprised of
spectrometer box, fiber optic cables and Thermo Scientific™
MarqMetrix™ Bioreactor BallProbe™ Sampling Optic tip. Average
prediction error for each analyte was 0.21 g/L for glucose, 0.16 g/L
for glutamine and 0.3 g/L for lactate. These results demonstrate the
excellent transferability of this complex chemometric model across
numerous MarqMetrix All-In-One Process Raman Analyzers.
Model analysis
A single PLS model was built using Eigenvector Solo software
to predict all three analytes in each mixture. The PLS calibration
model was built using 24 calibration samples with 3 replicates per
sample. All calibration spectra were collected using one MarqMetrix
All-In-One Process Raman Analyzer. The Raman fingerprint region
between 870-3096 cm-1 was used to build the model. A SavitskyGolay smoothing filter was applied to remove the random noise and
improve the signal to noise ratio. Next, baselines were corrected
followed by scattering correction and normalization. In addition, all
data were mean-centered before model building. The calibration
model was built using the cross-validation strategy of leave-onesample-out. The 3 replicates for the same sample were carried
together in this process. After these crucial pre-processing and
cross-validation steps were performed, the resulting optimized
model with 4 latent variables was selected. Results of the
calibration and cross-validation of the model are shown in Table 1.
Spectra for the 8-sample validation set were subsequently
collected on 10 different MarqMetrix All-In-One Process Raman
Analyzers. The chemometric model was applied to the validationset spectra for each MarqMetrix All-In-One Process Raman
Analyzer to predict the analyte concentrations. The results
discussed below show that high degree of accuracy and precision
were maintained for the prediction of all three analytes across all 10
MarqMetrix All-In-One Process Raman Analyzers.
Results
The development of robust chemometric models requires a
significant investment of time and resources. To ensure that
this investment provides long term value for our customers,
we have demonstrated excellent transferability of chemometric
models across numerous MarqMetrix All-In-One Process Raman
Analyzers. The correlation plot in Figure 1 shows the predicted vs.
reference values for glucose (Fig.1a), lactate (Fig.1b) and glutamine
(Fig.1c). Each plot contains an
overlay of the predicted values
for all 10 MarqMetrix All-In-One
Process Raman Analyzers. The
precision of the chemometric
model across 10 MarqMetrix AllIn-One Process Raman Analyzers
provides customers with
consistent results. Furthermore,
the accuracy of the model can be
seen from the results of Table 2.
Analyte
Model Parameter Glucose Glutamine Lactate
RMSEC (g/L) 0.079 0.075 0.146
RMSECV (g/L) 0.095 0.088 0.176
Bias (g/L) 4.79E-05 -1.94E-05 -2.50E-05
CV Bias (g/L) -2.25E-04 -6.45E-04 -4.42E-03
R2
Calibration 0.9995 0.9902 0.9994
R2
Cross-Validation 0.9993 0.9864 0.9991
Table 1. Calibration and cross-validation results for Chemometric
model.
Results
The development of robust chemometric models requires a significant investment of time and
resources. To ensure that this investment provides long term value for our customers, we have
demonstrated excellent transferability of chemometric models across numerous Thermo Scientific
Ramina Process Analyzers. The correlation plot in Figure 1 shows the predicted vs. reference values for
glucose (Fig.1a), lactate (Fig.1b) and glutamine (Fig.1c). Each plot contains an overlay of the predicted
values for all 10 Thermo Scientific Ramina Process Analyzers. The precision of the chemometric model
across 10 Thermo Scientific Ramina Process Analyzers provides customers with consistent results.
Furthermore, the accuracy of the model can be seen from the results of Table 2.
0
2
4
6
8
10
12
0 2 4 6 8 10 12
Predicted Glucose
(g/L)
Reference Glucose (g/L)
Glucose Instrument 1 Instrument 2
Instrument 3 Instrument 4
Instrument 5 Instrument 6
Instrument 7 Instrument 8
Instrument 9 Instrument 10
Linear (1:1 Fit)
0
5
10
15
20
0 2 4 6 8 10 12 14 16 18 20
Predicted Lactate
(g/L)
Reference Lactate (g/L)
Lactate Instrument 1 Instrument 2
Instrument 3 Instrument 4
Instrument 5 Instrument 6
Instrument 7 Instrument 8
Instrument 9 Instrument 10
Linear (1:1 Fit)
Results
The development of robust chemometric models requires a significant investment of time and
resources. To ensure that this investment provides long term value for our customers, we have
demonstrated excellent transferability of chemometric models across numerous Thermo Scientific
Ramina Process Analyzers. The correlation plot in Figure 1 shows the predicted vs. reference values for
glucose (Fig.1a), lactate (Fig.1b) and glutamine (Fig.1c). Each plot contains an overlay of the predicted
values for all 10 Thermo Scientific Ramina Process Analyzers. The precision of the chemometric model
across 10 Thermo Scientific Ramina Process Analyzers provides customers with consistent results.
Furthermore, the accuracy of the model can be seen from the results of Table 2.
0
2
4
6
8
10
12
0 2 4 6 8 10 12
Predicted Glucose
(g/L)
Reference Glucose (g/L)
Glucose Instrument 1 Instrument 2
Instrument 3 Instrument 4
Instrument 5 Instrument 6
Instrument 7 Instrument 8
Instrument 9 Instrument 10
Linear (1:1 Fit)
0
5
10
15
20
0 2 4 6 8 10 12 14 16 18 20
Predicted Lactate
(g/L)
Reference Lactate (g/L)
Lactate Instrument 1 Instrument 2
Instrument 3 Instrument 4
Instrument 5 Instrument 6
Instrument 7 Instrument 8
Instrument 9 Instrument 10
Linear (1:1 Fit)
Figure 1. Prediction correlation plot across 10 Thermo Scientific Ramina Process Analyzers.
Table 2 shows the average prediction error for each analyte on each Thermo Scientific Ramina Process
Analyzer and the average error for each analyte across all 10 Thermo Scientific Ramina Process
Analyzers. All parameters demonstrate a high degree of measurement accuracy with <0.5 g/L prediction
error. Furthermore, in some cases, the prediction error is <0.1 g/L. This level of accuracy is in line with
other published results for chemometric modeling3 and is on par with other relevant measurement
techniques for bioreactor monitoring.
Table 2. Prediction Error (RMSEP) Calculated from Chemometric Modeling
Hardware Average Prediction Error (g/L)
Glucose Glutamine Lactate
Instrument 1 0.18 0.05 0.20
Instrument 2 0.11 0.13 0.40
Instrument 3 0.12 0.15 0.21
Instrument 4 0.18 0.14 0.26
Instrument 5 0.18 0.08 0.40
Instrument 6 0.40 0.11 0.25
Instrument 7 0.13 0.38 0.32
Instrument 8 0.21 0.11 0.26
Instrument 9 0.47 0.15 0.44
Instrument 10 0.09 0.34 0.23
Average Error
(across 10 systems) 0.21 0.16 0.30
With this accuracy and precision, controlling glucose concentrations within a bioreactor in the typical 2-
4 g/L range can be readily achieved. Furthermore, through continuous monitoring and feedback
utilization, even tighter control is possible leading to improved process and product consistency.
Chemometric models developed using Thermo Scientific Ramina Process Analyzer have demonstrated
exceptional performance for multi-analyte monitoring in full-scale bioreactor studies. (Real time
0
0.5
1
1.5
2
2.5
0.00 0.50 1.00 1.50 2.00 2.50
Predicted Glutamine
(g/L)
Reference Glutamine (g/L)
Glutamine
Instrument 1 Instrument 2
Instrument 3 Instrument 4
Instrument 5 Instrument 6
Instrument 7 Instrument 8
Instrument 9 Instrument 10
Linear (1:1 Fit)
Figure 1. Prediction correlation plot across 10 MarqMetrix All-In-One Process Raman Analyzers.
A
B
C
© 2023 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific and its
subsidiaries unless otherwise specified. TS_MCS_AN_MMAIO_TRANS_1123
Learn more at thermofisher.com/MarqMetrixAIO
Table 2 shows the average prediction error for each analyte on
each MarqMetrix All-In-One Process Raman Analyzer and the
average error for each analyte across all 10 MarqMetrix All-InOne Process Raman Analyzers. All parameters demonstrate a
high degree of measurement accuracy with <0.5 g/L prediction
error. Furthermore, in some cases, the prediction error is <0.1
g/L. This level of accuracy is in line with other published results
for chemometric modeling3
and is on par with other relevant
measurement techniques for bioreactor monitoring.
With this accuracy and precision, controlling glucose
concentrations within a bioreactor in the typical 2-4 g/L range can
be readily achieved. Furthermore, through continuous monitoring
and feedback utilization, even tighter control is possible leading
to improved process and product consistency. Chemometric
models developed using MarqMetrix All-In-One Process Raman
Analyzer have demonstrated exceptional performance for multianalyte monitoring in full-scale bioreactor studies. (Real time
metabolite monitoring using the MarqMetrix All-In-One Process
Raman Analyzer and the Thermo Scientific™ 500L HyPerforma™
Dynadrive™ SingleUse Bioreactor (S.U.B.) MarqMetrix All-In-One
Process Raman Analyzer Real time App note
Conclusion
Customers will benefit from generating chemometric models
which are transferrable between multiple MarqMetrix All-In-One
Process Raman Analyzers. The ability of the MarqMetrix All-InOne Process Raman Analyzer to utilize the same chemometric
model across multiple units provides users with the confidence of
measurement accuracy and precision. Advanced signal processing
and model optimization may be employed to further increase the
level of prediction performance. This example simply provides
a benchmarking reference for the development of chemometric
models using MarqMetrix All-In-One Process Raman Analyzers.
References
1. FDA. Guidance for Industry: PAT—a Framework for Innovative
Pharmaceutical Development, Manufacturing, and Quality
Assurance, via FDA Website. 2004.. https://www.fda.gov/
media/71012/download. Accessed October 17th 2022.
2. Zhang, Lin, et al. “Uniform design applied to nonlinear
multivariate calibration by ANN.” Analytica Chimica Acta 370.1
(1998): 65-77.
3. Buckley, Kevin, and Alan G. Ryder. “Applications of Raman
spectroscopy in biopharmaceutical manufacturing: a short
review.” Applied spectroscopy 71.6 (2017): 1085-1116
Average prediction error (g/L)
Hardware Glucose Glutamine Lactate
Instrument 1 0.18 0.05 0.20
Instrument 2 0.11 0.13 0.40
Instrument 3 0.12 0.15 0.21
Instrument 4 0.18 0.14 0.26
Instrument 5 0.18 0.08 0.40
Instrument 6 0.40 0.11 0.25
Instrument 7 0.13 0.38 0.32
Instrument 8 0.21 0.11 0.26
Instrument 9 0.47 0.15 0.44
Instrument 10 0.09 0.34 0.23
Average error
(across 10 systems) 0.21 0.16 0.30
Table 2. Prediction error (RMSEP) calculated from Chemometric
modeling.
MarqMetrix All-In-One Process Raman Analyzer
Raman spectroscopy
Introduction
Manufacturing processes can be extremely complex, especially
biopharmaceutical manufacturing. With processes that rely heavily
on living organisms to generate products of interest, such as
those within the Biopharma industry, complete understanding
of the process is essential to success, because the more you
know about the process, the more control you can exercise. The
primary problem process analytical instruments and technologies
have sought to alleviate has always been a lack of visibility into the
underlying reactions. What you see, and also understand, you can
control and optimize.
Subtle variations in the environment can have a huge impact on
yield and quality. The consequences can range from failed batches
and inefficient use of resources to products that don’t meet quality
specifications, and limited data to help make improvements over
time. Ultimately, patient safety and stakeholder interests are at risk.
We are dedicated to making instruments that help our customers
gain greater control of their processes that involve chemistry. One
of the most powerful and advantageous analytical technologies that
we use is Raman spectroscopy. It’s extremely accurate and versatile,
generates very rapid results, covers a multitude of functions, and is
non-destructive to whatever sample or substance is being analyzed.
Application note
Until recently, Raman Spectroscopy required a well-trained technician
to operate, and demanded complex, bulky, and expensive equipment
that also made it at times unsuitable for in-line or field use.
We have introduced the Thermo Scientific™ MarqMetrix™ All-In-One
Process Raman Analyzer, a compact, easy-to-use, reliable, and
affordable system that leverages the power of Raman spectroscopy
to measure key variables in a process so that they may be controlled.
The MarqMetrix All-In-One Process Raman Analyzer also makes
Raman technology more accessible to non-experts through its
simplified user interface. Since it is also small, it is uniquely qualified to
solve a common problem faced by those who use chemistry to make
things, i.e., portability and ease of use whether in the field or in the
process development space.
What is RAMAN spectroscopy?
The underlying technology, Raman spectroscopy, is not new; it
has been around since the 1950s. What Raman instruments have
lacked until now is the size, reliability, ease of use, and affordability
that enables manufacturers to integrate Raman spectroscopy into
their production processes where it can be instrumental in improving
efficiency and quality. Before MarqMetrix All-In-One Process Raman
Analyzer, integrating Raman technology into a manufacturing process
was problematic. There were challenges with the hardware; it was
difficult to use and required a scientist on staff to operate and maintain
it. Reliability and cost were also issues. Better hardware was needed,
as well as a simplified, more user-friendly interface. These features and
advantages are now realized in MarqMetrix All-In-One Process Raman
Analyzer, but what exactly is Raman spectroscopy all about?
Non-destructive analysis and process monitoring
The technology begins with a laser that is directed at a substance
or sample through a fiberoptic cable with a probe at the end. The
energy from the laser light causes covalently bonded molecules in the
substance to vibrate and the light from the laser may scatter elastically
(the same laser energy is released as what caused the molecule
to vibrate) or inelastically (some of the laser energy is absorbed by
the molecule and a lesser amount of energy is released than what
caused the molecule to vibrate). Some of this inelastically scattered
light makes its way back into a detector within the MarqMetrix All-InOne Process Raman Analyzer. The detector collects and interprets
the light scattered from the sample to generate a “picture” called a
Raman spectrum. What makes this technology powerful is that the
Raman spectrum of a molecule is unique, and for that reason it’s
sometimes referred to as its molecular fingerprint. Just as fingerprints
may be used to identify people, we use the Raman spectrum to
identify a given substance because the fingerprint can tell us not only
qualitatively what something is, but also quantitatively how much
there is of it- one may determine both the identity and concentration
of a given analyte of interest. The non-destructive nature of Raman
spectroscopy provides a tremendous value-add. This is very important
because it can be integrated directly into a production line to measure
and analyze on a continuous basis, serving as a process monitor. This
is what is called in-line or online measurement. Raman technology
is also fast, and most substances can be measured in a matter of
seconds or less.
Versatile, precise, providing a wealth of information
Additionally, the Raman spectrum provides us with a great deal
of information about the substance being tested; every one of the
peaks in the Raman spectrum tells us something unique about what
we’re testing, providing us with many opportunities to identify what
we’re looking for. This is extremely important when measuring in a
bioreactor, for example, containing many molecules of different types.
The next advantage is that there’s a direct and linear relationship
between the concentration of a given substance and the intensity
of the peaks in the spectrum. This means that building quantitative
models is easier. With a relatively small number of samples, we can
build a model that accurately predicts concentration across our range
of detection. We can also measure substances in all forms, whether
they’re solid, liquid, gas, powder, or slurry. A final advantage is that
unlike other forms of spectroscopy, water does not distort Raman
measurements; therefore, we can see clearly what’s happening in the
aqueous solution like that found inside of a bioreactor. Competitively,
MarqMetrix All-In-One Process Raman Analyzer is better suited for
inline and online measurements in aqueous-based solutions than
competing technologies, but perhaps its greatest advantage is that it is
non-destructive.
Process control
There are Raman applications that span the entire biopharma
manufacturing process. Verifying the integrity of raw materials is the
first step where MarqMetrix All-In-One Process Raman Analyzer can
help ensure that the process gets off to a good start. In practice, we’ve
observed notable variations in the chemical composition of growth
media, variations significant enough to have an impact on yields and
cycle times. It’s important to be able to detect changes in the material
in real time. For example, customers want to know what’s happening
in the bioreactor so that they can make adjustments if necessary.
This is true regardless of whether or not manufacturing is done via
batch or continuous flow, and MarqMetrix All-In-One Process Raman
Analyzer works in either scenario. Do the cells have the right amount
of glucose? Are too many secondary metabolites building up? Are
the cells beginning to produce the product of interest? How much
product has been produced, and does it have the right characteristics?
MarqMetrix All-In-One Process Raman Analyzer enables answers to all
of these questions, and because the unit can interface with third-party
control systems, adjustments can be made in near real-time to optimize
conditions. Finally, when capturing finished product downstream,
manufacturers need to know exactly when and how much protein is
coming out of a purification column, for example. These are just a few
examples of how MarqMetrix All-In-One Process Raman Analyzer
helps achieve higher efficiency and quality in chemistry-dependent
manufacturing.
Robust yet simple operation
MarqMetrix All-In-One Process Raman Analyzer is easy to use. A
technician with no prior Raman experience can typically begin taking
measurements within 15 minutes of removing the instrument from
the box. Facilities across the country can use MarqMetrix All-In-One
Process Raman Analyzer to take hundreds of measurements daily
without the need for any scientific staff to maintain and calibrate the
instrument.
MarqMetrix AllIn-One Process
Raman Analyzer is
compact. With a
footprint less than
one square foot
and three inches
tall, MarqMetrix
All-In-One Process Raman Analyzer is sized to be placed at or near
the point of measurement and with no moving parts other than
its cooling fan, MarqMetrix All-In-One Process Raman Analyzer
is designed to be reliable and stable. Up time exceeds 99% and
calibrations performed in the factory remain accurate for years.
By being easy to use and reliable, MarqMetrix All-In-One Process
Raman Analyzer is also less costly to install and operate over time,
with a very low cost of ownership.
Software built to simplify chemometrics
Maximize the power of the MarqMetrix All-In-One Process
Raman Analyzer with Thermo Scientific™ Lykos™ PAT Software,
a remote control and monitoring software designed to simplify
the complexities of chemometrics. Lykos PAT Software facilitates
data acquisition and analysis by displaying, storing, and exporting
bioprocess monitoring data in real-time, enabling remote control and
monitoring of the MarqMetrix All-In-One Process Analyzer
and providing an intuitive workflow to streamline processes for
nonRaman spectroscopists and Raman experts alike.
A wide range of
easily-swappable
probes are
available
The MarqMetrix
All-In-One Process
Raman Analyzer can
be adapted to almost
any R&D production
or process system. It
features a Thermo Scientific™ MarqMetrix™ Fiber Head and Thermo
Scientific™ MarqMetrix™ BallProbe™ Sampling Optic that fits any need
due to a wide array of easily interchangeable probes. Swapping
the probe is easy; simply unscrew the fastener, remove the probe,
swap in another, and tighten it down again. Our tapered fitting allows
for easy indexing, requires no alignment, and there’s no need to
recalibrate the system.
The MarqMetrix All-In-One Process Raman Analyzer lineup of
standard fiber probes allows measurement of compounds in any
form, e.g., solids, liquids, gas, slurries, pastes, and gels. It can
measure by contact or immersion with
its performance half inch ball probe
and/or standard ball probes which
range in size from 1/2” down to
1/8”. For higher temperatures
and harsh chemicals, there are
process ball probes that are gold
sealed and temperature safe
Learn more at thermofisher.com/MarqMetrixAIO
© 2023 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific and its
subsidiaries unless otherwise specified. TS_MCS_BROCHURE_MMAIO_2_1123
to 350 degrees Celsius. MarqMetrix BallProbe Sampling Optics
are also available in bioreactor-specific versions with a seal and
nut to allow for immersion in a reaction vessel, be it a dedicated
stainless-steel bioreactor or a single-use bioreactor. These Thermo
Scientific™ MarqMetrix™ Bioreactor BallProbe™ Sampling Optics are
autoclavable, either while attached to the bioreactor, or on their own
when cleaning and sterilizing the ball probe separately.
Integration into a high-pressure flow system is simplified with
patented flow cell technology operable at 2500 psi. Custom probes
are also available.
At Thermo Fisher, we’re realizing the full potential of Raman
technology by building better instruments, and we’re all about
making Raman spectroscopy more accessible to help our
customers control chemistry. We’re opening up new applications
for Raman and increasing the returns on existing applicationsreturns that come in the form of faster cycle times, higher yields,
and better quality.
For research use only. Not for use in diagnostic procedures. For current certifications, visit thermofisher.com/certifications
© 2024 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific
and its subsidiaries unless otherwise specified. MCS-CM1177-EN 11/24
Learn more at thermofisher.com/marqmetrixaio
Single-use bioreactor solution
The DynaDrive Bioreactor Platform is a high-performance, single-use
bioreactor system designed for optimized mixing, mass transfer, and scalability
in biopharmaceutical production.
Process Raman analyzer
The MarqMetrix All-In-One Process Raman Analyzer is a compact, powerful
spectrometer for nondestructive real-time analysis of critical process parameters
directly in-line with processes. It can be used for direct process monitoring and
control in upstream and downstream bioprocessing.
Learn more at thermofisher.com/dynadrive
Brought to you by
Download the eBook for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version