Optimize mAb Purification for Greater Efficiency
eBook
Published: October 15, 2025
Credit: Thermo Fisher Scientific
The growing complexity of antibody therapeutics has driven rapid innovation in downstream processing. As novel formats such as bispecifics, fragments and antibody–drug conjugates (ADCs) expand, so must the purification toolbox to maintain purification and yield performance while managing cost and throughput.
To maintain product quality and regulatory compliance, bioprocess teams need to expand their purification toolkit with high-performance chromatography tools.
This eBook explores recent developments in purification and polishing, highlighting strategies that streamline processing, help to enhance purity and increase manufacturing scalability across diverse monoclonal antibody (mAb) modalities.
Download this eBook to discover:
- How flow-through and mixed-mode chromatography approaches can improve yield and consistency
- Guidance for selecting resins that balance performance and help cost-efficiencies
- Specialist insights on overcoming purification challenges in complex antibody therapeutics
Efficient techniques for
antibody therapeutic
purificationForeword
Monoclonal antibodies (mAbs) are fast becoming one of the
most sought after therapeutics, with hundreds approved
for a wide range of clinical uses, from cancers to infectious
disease, in the last few decades.
In order to address more difficult therapeutic targets, novel
antibody modalities are being developed, such as bispecific
mAbs, antibody fragments and antibody-drug conjugates.
However, these new modalities present purification
challenges that traditional methods of purification such as
polishing with ion exchange chromatography struggle to
overcome. This results in higher costs and lower yields at
manufacturing scale.
This eBook explores the recent developments in purification
and polishing solutions that can help address the
challenges in downstream processing.
1 thermofisher.com/antibody-derived-therapeuticsForeword 1
The challenges of therapeutic monoclonal antibodies 3
Purification strategies for monoclonal antibody production 7
Purification of complex mab modalities: ask the specialists 8
Advances in flow-through technology to enhance mab polishing 12
5 key considerations for optimizing resin selection for
efficient mab purification 17
Resources 21
Contents
23 thermofisher.com/antibody-derived-therapeutics
Introduction
Since the development of the hybridoma technology in 1975,
monoclonal antibodies (mAbs) have become increasingly
important in both medical research and disease treatment.¹ The
first clinically available mAb, muronomab CD3, was approved
in 1986 to block cytotoxic T cell function and prevent rejection
in transplanted patients.² Since then, hundreds of mAbs have
been approved for clinical use for the treatment of a wide
range of diseases, from cancers and autoimmune disorders to
infectious diseases such as HIV.3,4
Despite the success of mAb therapeutics, developing novel
antibodies is challenging, as many of the best-understood
targets have already been exploited. Remaining targets for
potential novel mAb therapeutics pose challenges such as
poorly accessible sites of action, low immunogenicity and
significant differences in targets between human and the
species used for preclinical testing.⁵ In order to overcome these
roadblocks, novel mAb-based therapies are being developed.
These include bispecific mAbs capable of binding two different
antigens, antibody–drug conjugates (ADCs), isotype-switched
mAbs and antibody fragments.⁶
As biotherapeutics, mAbs must adhere to strict regulatory
requirements to help ensure the safety and efficacy of the final
product.7,8 Due to their production in cell-based expression
systems, the manufacture of mAbs can result in process- and
product-related impurities. These impurities significantly affect
the quality and safety of the final product. Therefore, mAbs
must go through extensive purification steps before release.
However, as the structural complexity of novel mAbs increases,
the traditional purification methods have become less effective,
resulting in high costs and low yields. Recent developments
in affinity purification reagents and polish solutions promise
to simplify purification of complex mAbs and improve the
efficiency of commercial production of novel mAb therapeutics.
Purifying mAbs for clinical use
In order to meet regulatory guidelines, purified mAbs must have
low levels of both process- and product-related impurities,
while maintaining a reasonable and cost-effective yield.
Process-related impurities include host cell proteins (HCPs),
host cell DNA, leached reagents from other purification steps,
process buffers and residual detergents used for viruses.
Product-related impurities include high and low molecular
weight product aggregates, misformed products, viruses,
endotoxins from bacterial expression systems and posttranslational modifications.⁹
Following mAbs harvesting from cell culture by centrifugation
and filtration, the typical mAb purification process begins with
protein A chromatography (Figure 1). Protein A is covalently
immobilized onto porous resins. As the samples pass through
the column, protein A binds to the Fc region of antibodies at
the consensus binding site (CBS), capturing the mAbs and
resulting in relatively high purity and recovery in a single step.⁹
One of the advantages of protein A chromatography is its
sensitivity to low pH. This enables an acidic elution of bound
antibodies, and a subsequent low pH hold to inactivate any
contaminating viruses (in line with regulatory guidelines for viral
safety).10,11
The efficiency of protein A chromatography means that
downstream polishing steps only need to remove a small
amount of the remaining impurities. The most common of
these polishing steps are based on ion exchange, namely
anion exchange (AEX) chromatography and cation exchange
(CEX) chromatography, depending on the impurity to be removed.
The net charge of mAbs varies, depending on the relative pH
of the antibody’s environment. Changing the environment
therefore allows the manipulation of mAb charge and enables
the removal of a wide range of differently charged impurities.12
CEX chromatography binds positively charged antibodies and
impurities, which are then separated based on charge difference,
eluting at different ionic strengths. AEX chromatography binds
negatively charged impurities, while positively charged antibodies
are allowed to flow through. It is used for the removal of host cell
DNA, HCPs, bacterial endotoxins and furhter viral clearance.13,14
The final polishing steps in purification to help ensure the product
meets regulatory specifications, using methods such as ion
exchange, or hydrophobic interaction chromatography (HIC).15
The challenges of therapeutic
monoclonal antibodies4 thermofisher.com/antibody-derived-therapeutics
The Protein A chromatography workflow is highly efficient
and selective for traditional mAbs, as it removes the majority
of impurities in a single robust step. Both AEX and CEX
chromatography have high loading capacities and can
accommodate both bind-elute and flow-through modes, for
efficient purification at manufacturing scale.
The challenges of traditional polishing
Despite the successes of current purification and polishing
processes, these methods still show limitations for both traditional
and more complex antibody technologies. Thus, there is a
significant demand for better manufacturing and purification
strategies. Inherent challenges in mAb production begin even
before the purification stages. mAbs are limited to production
in a live-cell expression system, which can be difficult to scale
to a commercial manufacturing level and can produce variable
yields. Historically, mammalian expression systems were used for
full-length antibodies, though recent advances have also allowed
the use of Escherichia coli.16 Emerging technologies using in vitro,
cell-free systems could reduce some of the challenges associated
with live-cell systems (e.g., reducing the amount of cellular debris
and cell-related impurities). However, these methods are not yet
ready for industrial-scale manufacture.17
Extraction of any high-purity product from a complex mixture
such as a cellular expression system is a challenge, but the
development of emerging mAb technologies has complicated
this process further. The diversity of novel mAb structures has
led to increased process- and product-related impurities, for
which traditional methods are not suitable. For example, bispecific
antibodies may express heavy chains (HCs) and light chains (LCs)
from two different parental mAbs. Should these HCs and LCs
mispair to form a homodimer during production, they represent
an additional impurity that must be removed from the final
product, via additional purification steps or specific resins.18 The
complex structure of bispecific mAbs also leads to increased
levels of aggregation and the generation of antibody fragments,
which can prove difficult to remove by conventional means.18
Other novel modalities, such as antibody fusion proteins, bring
their own unique challenges to purification. In this type of
therapeutic, the long half-lives of antibodies are leveraged to
enhance the effectiveness of shorter-lived therapeutic proteins
such as cytokines. However, these too show increased levels
of misformed and mismatched products and aggregates.
In addition, instability in acidic conditions can give rise to
further aggregation and cleavage during the low pH steps of
conventional purification methods.18
For both standard and novel mAbs modalities, traditional polishing
methods can be further optimized for more efficient purification
at almost every stage of the process. In some cases, the type
of chromatography itself can inhibit efficient purification. Despite
its importance for viral clearance, the low pH hold step results
in aggregate formation, exacerbated by the process of protein
A chromatography itself.19 Protein A can leech into the eluted
fraction during the chromatography, both as fragments and fullsize proteins. This results in additional impurities to be removed
from the final product. Furthermore, the protein A fragments can
be even more difficult to remove than the whole proteins by ion
exchange chromatography.20 HIC is often used as an orthogonal
polishing step during mAb purification due to its high aggregate
clearance capability. However, the high concentrations of
kosmotropic salts used during the process require extra washing,
dilution and ultrafiltration steps to remove the salts from the final
product, reducing overall efficiency.21
In addition to limitations in the current processes, the impurities
to be removed can also cause issues. HCPs are a critical impurity
that must be cleared to ensure the safety of the final product.
Although the majority of HCPs can be cleared across multiple
polishing steps, the remaining HCPs can be highly difficult to
remove using traditional means.22 Residual HCPs can decrease
product stability and cause unwanted immune reactions in the
recipients.22 Finally, purification bottlenecks can occur at several
steps in the purification process.
Optimizing downstream processing for improved
purification
The challenges of traditional purification and polishing methods
can be overcome through careful optimization and selection of
the most appropriate polishing platforms, ensuring complete
Bioreactor
Protein A
capture Capture
Clarification
Polishing
Low pH VI
Cation
exchange
Anion
exchange
Virus
filtration
Waste products Centrifuge
Cells and cell
debris
HCP, DNA,
fragments
Aggregates,
leached
protein A
HCP, DNA
1 Depth
filtration
2 Depth
filtration
2 Depth
filtration
Figure 1. mAb production process flow diagram. Adapted from
Gillespie et al., 2014.thermofisher.com/antibody-derived-therapeutics 5
removal of impurities. Ion exchange chromatography is a
common post-protein A step in most polishing workflows to
remove high molecular weight contaminants.9 Ion exchange
chromatography can be employed in both bind-and-elute and
flow-through modes, with the optimal method dependent on
the isoelectric point of the mAb to be purified. A version of AEX
chromatography known as weak partitioning chromatography
(WPC) can remove weakly binding impurities, increasing
product yield.⁹
CEX chromatography has been shown to effectively remove
HCPs, and maximum removal can be assured by selecting
an optimal stationary phase with the chemical and physical
properties best for the impurities and mAb in question.23 To
reduce purification bottlenecks and increase throughput, highcapacity resins can be selected for CEX which may allow the
omission of a rate-limiting protein A capture step.24 Washing,
dilution and filtration steps that increase the time cost of HIC
may be reduced through the use of a salt-free method, using
an extremely hydrophobic resin in the flow-through step without
kosmotropic salts.21
As the technology of novel mAb modalities advances, it is only
fitting that the technology of mAb purification also advances
in step. If optimal yield and efficiency are to be achieved, and
regulatory guidelines for purity are to be met, then traditional
purification methods and resins are no longer the best, or only,
choice. With the wide range of resins now available for all types
of chromatography, use of historical and outdated resins is
no longer necessary. Resin screening studies can be used to
easily select the resin with the highest binding capacity, best
selectivity and best resolution for the antibody in question. For
example, the ideal resin for bispecific antibody purification and
removal of mispaired dimers would consist of a small particle
size combined with large pore diameters for perfusive flow and
mass transfer.18
Further advances in efficiency and purification outcomes
have been achieved by the combination of ion exchange
chromatography with other methods of purification such as
HIC. Known as mixed-mode chromatography, the stationary
phases involved consist of both hydrophobic and charged
functional groups. High levels of aggregates and highmolecular-weight impurities can be removed in a single
step. This eliminates the need for multiple ion exchange
chromatography polishing steps, improving cost and timeefficiency, and helping to ensure a highly pure product, even for
complex mAb modalities.25
Conclusion
Chromatography plays an essential role in therapeutic mAb
manufacture and purification. Until recently, lags in the
development of updated resins and methods have decreased
the efficiency and effectiveness of traditional purification
pathways in the face of novel mAb technologies. However, new
resins and methods, such as mixed mode chromatography,
promise to address the challenges posed by more conventional
methods, enabling more thorough removal of impurities in fewer
steps. Selection of the best resin and platform for superior
mAb purification is crucial, but it doesn’t need to be complex or
confusing. The following articles will outline the tools available
for high-quality mAb purification and polishing, and the novel
technologies that can significantly enhance the efficiency and
economy of mAb manufacture.
References
1. Köhler G, Milstein C. Continuous cultures of fused cells secreting antibody of
predefined specificity. Nature. 1975;256(5517):495-497. doi:10.1038/256495a0
2. Todd PA, Brogden RN. Muromonab CD3. A review of its
pharmacology and therapeutic potential. Drugs. 1989;37(6):871-899.
doi:10.2165/00003495-198937060-00004
3. Manis JP. Overview of the therapeutic monoclonal antibodies. UpToDate. Wolters
Kluwer. https://www.uptodate.com/contents/overview-of-therapeuticmonoclonal-antibodies/print. Updated April 10, 2024. Accessed June 11,
2024.
4. Lu R-M, Hwang Y-C, Liu I-J, et al. Development of therapeutic antibodies
for the treatment of diseases. J Biomed Sci. 2020;27(1). doi:10.1186/
s12929-019-0592-z
5. Carter PJ, Lazar GA. Next Generation Antibody Drugs: Pursuit of the “highhanging fruit.” Nat Rev Drug Discov. 2017;17(3):197-223. doi:10.1038/
nrd.2017.227
6. Lyu X, Zhao Q, Hui J, et al. The global landscape of approved antibody therapies.
Antibody Therap. 2022;5(4):233-257. doi:10.1093/abt/tbac021
7. Guideline on development, production, characterisation and specification for
monoclonal antibodies and related products. European Medicines Agency.
2016. https://www.ema.europa.eu/en/documents/scientific-guideline/
guideline-development-production-characterisation-and-specificationmonoclonal-antibodies-and-related-products-revision-1_en.pdf Published
July 21, 2016. Accessed June 11, 2024.
8. Guidance for industry for the submission of chemistry, manufacturing, and controls
information for a therapeutic recombinant DNA-derived product or a monoclonal
antibody product for in vivo use. United States Food and Drug Administration.
https://www.fda.gov/media/77528/download. Published August 1996.
Accessed June 11, 2024.
9. Liu HF, Ma J, Winter C, Bayer R. Recovery and purification process development
for monoclonal antibody production. mAbs. 2010;2(5):480-499. doi:10.4161/
mabs.2.5.12645
10. Jin W, Xing Z, Song Y, et al. Protein aggregation and mitigation strategy in low ph
viral inactivation for monoclonal antibody purification. mAbs. 2019;11(8):1479-
1491. doi:10.1080/19420862.2019.1658493
11. ICH Q5A(R2) Guideline on viral safety evaluation of biotechnology
products derived from cell lines of human or animal origin.
European Medicines Agency. https://www.ema.europa.eu/en/
ich-q5ar2-guideline-viral-safety-evaluation-biotechnology-productsderived-cell-lines-human-or-animal-origin-scientific-guideline. Published
December 14, 2023. Accessed June 11, 2024
12. Schneider Z. Importance of isoelectric point (pI) of antibodies. The Antibody
Society. https://www.antibodysociety.org/new-articles/importanceisoelectric-point-pi-antibodies. Published June 28, 2017. Accessed June 11,
2024.
13. Chahar DS, Ravindran S, Pisal SS. Monoclonal antibody purification and its
progression to commercial scale. Biologicals. 2020;63:1-13. doi:10.1016/j.
biologicals.2019.09.007
14. Curtis S, Lee K, Blank GS, Brorson K, Xu Y. Generic/matrix evaluation of SV40
clearance by anion exchange chromatography in flow‐through mode. Biotechnol
Bioeng. 2003;84(2):179-186. doi:10.1002/bit.107466 thermofisher.com/antibody-derived-therapeutics
15. Arora I. Chromatographic Methods for the purification of monoclonal antibodies
and their alternatives: A Review. Int J Emerg Techn Adv Eng. 2013;3(10):475-481.
16. Rashid MH. Full-length recombinant antibodies from Escherichia coli: production,
characterization, effector function (FC) engineering, and clinical evaluation. mAbs.
2022;14(1). doi:10.1080/19420862.2022.2111748
17. Sifniotis V, Cruz E, Eroglu B, Kayser V. Current advancements in addressing
key challenges of therapeutic antibody design, manufacture, and formulation.
Antibodies. 2019;8(2):36. doi:10.3390/antib8020036
18. Chen SW, Zhang W. Current trends and challenges in the downstream purification
of bispecific antibodies. Antib Ther. 2021;4(2):73-88. doi:10.1093/abt/tbab007
19. Mazzer AR, Perraud X, Halley J, O’Hara J, Bracewell DG. Protein a
chromatography increases monoclonal antibody aggregation rate during
subsequent low ph virus inactivation hold. J Chromatogr A. 2015;1415:83-90.
doi:10.1016/j.chroma.2015.08.068
20. Carter-Franklin JN, Victa C, McDonald P, Fahrner R. Fragments of protein a eluted
during protein a affinity chromatography. J of Chromatogr A. 2007;1163(1-2):105-
111. doi:10.1016/j.chroma.2007.06.012
21. Ghose S, Tao Y, Conley L, Cecchini D. Purification of monoclonal antibodies
by hydrophobic interaction chromatography under no-salt conditions. mAbs.
2013;5(5):795-800. doi:10.4161/mabs.25552
22. Ito T, Lutz H, Tan L, et al. Host cell proteins in monoclonal antibody processing:
Control, detection, and removal. Biotechnol Progr. 2024;e3448. doi:10.1002/
btpr.3448
23. Bruch T, Graalfs H, Jacob L, Frech C. Influence of surface modification on protein
retention in ion-exchange chromatography. J Chromatogr A. 2009;1216(6):919-
926. doi:10.1016/j.chroma.2008.12.008
24. Tao Y, Ibraheem A, Conley L, Cecchini D, Ghose S. Evaluation of high‐capacity
cation exchange chromatography for direct capture of monoclonal antibodies from
high‐titer cell culture processes. Biotechnol Bioeng. 2014;111(7):1354-1364.
doi:10.1002/bit.25192
25. Mixed-mode chromatography. ThermoFisher Scientific. https://www.
thermofisher.com/uk/en/home/life-science/bioproduction/poroschromatography-resin/bioprocess-resins/mixed-mode-chromatography.
html. Accessed June 11, 2024.Monoclonal antibodies (mAbs) are becoming an increasingly
prominent therapeutic agent which can be engineered to treat a wide range of diseases and
disorders, from cancers to autoimmune
diseases.1 Consequently, many developers have multiple mAbs in their pipeline,
increasing the demand for rapid,
efficient purification processes.
Purification of mAbs is a complex,
multistep process. After initial
harvesting and filtration steps,
mAbs undergo capture, affinity
purification and polishing steps to
remove impurities.
However, the complexity of engineered
mAbs can hinder the purification
process, creating mis-formed
products, antibody fragments
and aggregates. Failure of the
process requires modifications
to both the capture step and
the polish strategy.
This infographic will explore
the modifications needed for an
optimized platform process, focusing
on impurity removal and the Thermo
Fisher Scientific solutions available.
As the complexity and sophistication of mAb therapies increases, structural features such as symmetric,
asymmetric and fragment-based bispecifics are becoming more common, resulting in new purification
challenges. The traditional polishing process can have limited success for these mAbs, resulting in
incomplete impurity removal, and risks non-adherence to regulatory guidelines.
Alternative processes, such as hydrophobic interaction chromatography (HIC) and mixed-mode
chromatography (MMC) are emerging as more efficient platforms for more modern, engineered mAbs.
Purification
strategies for
monoclonal
antibody
production
Typically, an affinity step and two polish steps are needed to remove a range of impurities:
A typical purification process consists of three steps: affinity chromatography, often using Protein A, followed by
cation exchange (CEX) then anion exchange (AEX).
Productrelated
impurities (e.g.,
aggregates,
mis-formed
product, dimers,
post-translational
modifications)
Viruses
Host cell
proteins
(HCPs)
DNA
Endotoxins
(from bacterial
expression
systems)
Affinity
Antibody isolation
HCP reduction
Thermo ScientificTM
CaptureSelectTM
Affinity Resins
HIC
• HCP reduction
• Leached ligand removal
• Aggregate, fragments
• Product related isoforms
AEX as needed
• Final HCP removal
• DNA removal
• Virus removal
AEX
• Final HCP removal
• DNA removal
• Virus removal
Thermo ScientificTM
POROSTM Caprylate
Mixed-Mode Cation
Exchange Resins
Thermo ScientificTM
POROSTM AEX Resins
• POROS XQ
• POROS 50 HQ
• POROS 50 D
• POROS 50 PI
Thermo ScientificTM
POROSTM AEX Resins:
•POROS XQ
• POROS 50 HQ
• POROS 50 D
• POROS 50 PI
Thermo ScientificTM
PorosTM CEX Resins:
• POROS XS
• POROS HS
CEX as needed
• Final HCP removal
• DNA removal
• Virus removal
Thermo ScientificTM
POROSTMHIC Resins:
• POROS ETHYL
• POROS BENZYL
• POROS BENZYL
ULTRA
Mixed Mode-CEX
• HCP reduction
• Leached ligand removal
• Aggregate, fragments
• Product related isoforms
References:
1. Quinteros DA, Bermúdez JM, Ravetti S, Cid A, Allemandi DA, Palma SD. Therapeutic use of monoclonal antibodies: General
aspects and challenges for drug delivery. Nanostruct Drug Deliv. Published online 2017:807-833. doi:10.1016/b978-0-323-
46143-6.00025-7
Thermo ScientificTM
CaptureSelectTM
Affinity Resins
High purity and
yield in a single step
Affinity
• Antibody isolation
• HCP reduction
Thermo ScientificTM
POROSTM CEX
Resins:
Cation Exchange
Removal of:
Challenging mAb
platform process
Thermo ScientificTM
POROSTM AEX
Resins:
Anion Exchange
For removal of:
• Final HCP
• DNA
• Viruses
The POROS Caprylate Mixed-Mode Cation Exchange Resins have a superior pore structure for rapid mass transport and
unique selectivity. This can help increase productivity, purity and yield in flow-through modes.
Improve your polish process for engineered mAbs with
Thermo Scientific™ POROS™ Caprylate Mixed-Mode
Cation Exchange Resin
MMC
Typically combines ion exchange and
hydrophobic interaction principles for improved
selectivity and resolution of impurities.
Able to purify diverse populations
of proteins.
Removes high levels of aggregates and a
wide range of impurities.
Suitable for bind-elute and flow-through
chromatography.
HIC
Removes proteins based on interactions
between hydrophobic ligands on the resin with
hydrophobic motifs on proteins.
Salt buffer conditions promote binding and
stabilize mAb structure.
Removes high levels of aggregates and a wide
range of impurities.
Suitable for bind-elute and flow-through
chromatography.
Removal of impurities
A traditional mAb downstream platform process
Typical process steps
Optimizing polish strategies for complex mAbs
Complex mAb process steps
The advantages of Mixed Mode-CEX resins
© 2025 Thermo Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific and its subsidiaries unless otherwise specified. This information is not intended to
encourage use of these products in any manner that might infringe the intellectual property rights of others.
Pharmaceutical Grade Reagent. For Manufacturing and Laboratory Use Only.
Affinity chromatography
Captures proteins based on reversible
protein/ligand interactions, where the ligand
is coupled to a chromatography resin.
Cation exchange
Captures positively charged mAbs and
impurities. Captured mAbs are then eluted by
increasing conductivity or increasing buffer pH.
Anion exchange
Removes negatively charged product and
process-related impurities. It can be used in
both flow-through and bind-elute modes.
• HCP
• Leached ligand
• Aggregates and
fragments
• Product-related
isoforms
• POROS XS
• POROS 50 HS
• POROS XQ
• POROS 50 HQ
• POROS 50 D
• POROS 50 PI
Complex mAb
platform process
High aggregate
selectivity, effective up
to 20% aggregation
Cost-effective
due to reduced
chromatography
steps and reagents
Linear pressureflow curve results
in excellent and
predictable scalability
Wide operating range
80–90% mAb monomer
recovery with minimal
optimization8 thermofisher.com/antibody-derived-therapeutics
Purification of complex mAb
modalities: ask the specialists
As the complexity of novel mAb modalities increases, traditional
purification and polishing processes may no longer be effective.
Outdated resins and archaic polishing methods can decrease the
efficiency and effectiveness of mAb purification, while new mAb
structures can also bring new, difficult to remove impurities. We asked
three in-house specialists at Thermo Fisher Scientific for their advice on
how to overcome the challenges of purifiying complex mAbs, and how
to optimize your polishing strategies for maximum efficiency.
Jett Appel | Field Application Scientist,
Thermo Fisher Scientific
Jett Appel has been a Purification Field Applications Scientist at Thermo Fisher Scientific since
2021, supporting purification development and scale-up for monoclonal antibodies, viral vectors,
nucleic acids, and recombinant proteins. Prior to joining Thermo Fisher, Jett worked at Avid
Bioservices, a contract development manufacturing organization (CDMO) based in Orange County,
CA. During his 5 years of working at Avid, Jett first worked in process development and eventually
transitioned to a senior engineer role in the Manufacturing Sciences & Technology (MSAT) team,
where he supported purification development and technology transfers for monoclonal IgGs,
IgMs, bispecific antibodies, scFvs, and recombinant enzymes up to 2000 L bioreactor scale. Jett
received a bachelor’s degree in Chemical and Biomolecular Engineering from UCLA in 2016. While
at UCLA, he was involved in epigenetics research at the Steve Jacobsen Lab, which included
studying the pathways involved in RNA-directed DNA methylation in Arabidopsis thaliana.
Nicholas Bardol | Manager, Manufacturing Services,
Thermo Fisher Scientific
Nicholas has been working in the pharmaceutical industry for the past 13 years as a process
development scientist focusing on biologics purification and small molecule conjugation. His work
at CDMOs has supported over a dozen novel molecules through GMP Manufacturing and FDA
filings. He joined the Saint Louis, MO, Thermo Fisher Scientific site supporting Pharma Services
location in 2018 as a member of the downstream process development group.
Kelly Flook | Senior Manager, Product Management,
Purification Products, Thermo Fisher Scientific
Kelly Flook is a dynamic product manager specializing in downstream purification of
biotherapeutics and new product development. With over 15 years’ experience in product
development Kelly has an excellent track record of bringing the right product to the market.
She started her career as a scientist specializing in the development of polymer based analytical
columns for HPLC before moving into purification R&D and then product management. Kelly
went to school in the UK, receiving a bachelor’s degree in Analytical Chemistry from University
of Northumbria, Newcastle, and a PhD in Polymer Chemistry from University of Durham.thermofisher.com/antibody-derived-therapeutics 9
Q: What are the main challenges faced in the
purification of complex mAbs compared to
traditional monoclonal antibodies?
Jett Appel (JA): It’s hard to pinpoint the main challenges
because it depends a lot on antibody structure – and antibody
structures are diverse, creating diverse challenges. Often, when
we talk about complex mAbs, we are talking about bispecific or
multispecific antibodies, antibodies where the Fc domain – or
even antibody fragments, such as Fabs or scFvs – are modified.
So, for example, you may not be able to utilize traditional affinity
steps for purification, or it could result in product impurities
that are harder to separate. So, good yield or purity tends to be
obtained differently from these different complex mAbs.
Nicholas Bardol (NB): An increasing number of drug
candidates are bispecific, which are very difficult to separate,
because bispecific expression systems produce a lot of parent
mAb fragments and paratopes as impurities. Most fragments
not only share a lot of the primary characteristics we would
usually exploit in traditional purification strategies, but they can
also be more biologically active. So if they outcompete the drug
candidate for the epitope they’re targeting, then the efficacy of
the drug candidate really diminishes.
Kelly Flook (KF): Because structures are growing more
and more complex, heterogeneity and aggregation become
increasingly challenging. However, it’s not just complex mAbs
that are suffering from increased aggregation levels. As the
industry moves towards more cost-efficient mAb production,
higher cell densities and processes to increase titer can impose
additional challenges downstream with respect to process- and
product-related impurities.
Q: How do the structural characteristics of
complex mAbs impact the purification process
and pose unique challenges?
JA: It depends on the exact structure of the complex mAb.
Bispecific or multispecific antibodies often have different
heavy chains or light chains and this can result in mispairing.
Consequently, these antibodies are very similar in physicochemical
properties and can be hard to separate or may get co-purified with
traditional Protein A affinity steps.
Other formats of complex mAbs – for example, antibodies that
have been multimerized – are much larger than traditional mAbs.
This can result in steric hindrance or lower diffusivities. That’s a
unique challenge because it may lead to lower resin capacities and
therefore larger columns, which could ultimately lower productivity
as scale increases.
NB: In the case of antibody–drug conjugates, they’ve been
modified specifically to take on a drug linker, and that can pose
many challenges – we see issues with free thiol or unbound drug
linker. In the case of bispecifics, it’s hard to use charge-based
separation because the sequence of impurities will overlap a lot with
the sequence of the target antibody.
Just as importantly, we may not be able to use our typical polishing
strategies of anion and cation exchange. Instead, we have to
rely more on things like multimodal or hydrophobic interaction
chromatography (HIC) as a different tool to target another
characteristic of those impurities and see if we can remove them
with a different mechanism than one the industry typically relies on.
Q: What are some of the common impurities or
contaminants encountered during the purification
of complex mAbs? And how are they typically
addressed?
JA: Some common impurities overlap with traditional mAbs –
e.g., host cell proteins, residual DNA and aggregates. Often in
these complex mAbs, we tend to see higher levels of aggregates
because as these structures are engineered the impurities are
either produced more in the cell culture itself or can be generated
under traditional mAb purification parameters, such as exposure
to low pH conditions. The complex mAbs can be less stable and
form impurities inherently within the process.
Other common impurities can be mispaired homodimers, for
example. They’re typically addressed through a traditional ion
exchange to separate the target product from these impurities.
But, as Nick was alluding to earlier, this can often be a
challenge. So, you may need to implement HIC or multimodal
chromatography as well.
NB: I already alluded to impurities that we see with unbound drug
linker or bispecific isoforms, but the biggest one that we deal with
is host cell protein (HCP). In some of the newer mAbs, there is a
lot of change in the pI of the target antibody, and it can overlap
with the HCP pI. Traditionally, antibodies have high pI relative to
the HCPs, making charge-based separation very straightforward:
you bind up your antibody, and flow through all the HCP. But,
suppose the target is sitting right in the middle. Then you either
need multiple steps to capture and resolve everything on either
side of the pI or you have to look at another option like multimodal
chromatography or HIC again, just to get rid of those impurities
that have historically been relatively simple to remove.
KF: Many manufacturers have developed platform purification
workflows for their traditional mAbs, but these don’t always
work for their more complex molecules. This is when you
need to turn to HIC or multimodal chromatography. For
affinity purification, although Protein A continues to be the
workhorse, it’s not always the best option, depending on the
binding domains of the specific antibody. This is where our
CaptureSelect affinity resin product line comes in.10 thermofisher.com/antibody-derived-therapeutics
Q: Could you discuss any specific strategies
or technologies employed to overcome the
challenges associated with complex mAb
purification?
JA: We’ve seen customers implementing unique affinity
technologies. Typically, we’ve utilized traditional protein A
affinity chromatography but we can see that, depending
on how these complex mAbs are engineered, other affinity
ligands that target different domains of the antibody could
be advantageous. For bispecifics, we’ve seen engineered
antibodies where one arm doesn’t have a CH1 domain. So,
for example, you could utilize an affinity resin that targets
CH1. That homodimer won’t bind to the resin and will just flow
through instead of getting co-purified with protein A.
Another interesting strategy that customers use is to leverage
the avidity of the target molecule or the impurity to the resin
and utilize pH gradients. This is used, depending on the
antibody format, when there is more than one specific domain,
or if the target molecule or the impurity has fewer domains
than the other. Where traditional operating conditions for
polishing steps work for standard mAbs, they may not work
for complex mAbs. These may then need a unique set of
conditions; for example, operating cation exchange under a
more alkaline condition may help improve resolution. As Nick
mentioned earlier, other technologies – such as multimodal
chromatography or HIC – can be a useful tool if the standard
methods do not work.
KF: Our ion exchange resins have high capacity and robust salt
tolerance to be able to handle higher titers in bind-and-elute
mode. Using a flowthrough process, your capacity becomes
capacity for the impurity, which is typically <5% at this point,
meaning you can significantly improve productivity and reduce
resin consumption.
NB: I want to highlight a couple of things both upstream and
downstream of purification. The industry has been chasing high
titers in their clone selection for as long as biologics have been
around, but choosing the clone with the highest titer may not be
the best strategy. Instead, choosing the cell line with the lowest
levels of a complex impurity might be the best mechanism. So
be very forward when thinking about clone selection.
Another thing is analytical strategies. A lot of these new
impurities come with bispecifics and isoforms that share the
same charge; using all of the traditional analytical techniques,
you might not get total resolution. They may co-elute and you
won’t know the contaminant is present until the end stages,
or you may chase something that isn’t there. So, looking at
orthogonal assays for size or isoform pattern and charge
heterogeneity can be hugely beneficial. Some less commonly
used techniques can be really powerful here; using 2D-LC and
native mass spec to see those hidden isoforms can save a lot
of time when you’re trying to develop your purification strategy.
Q: Are there any particular purification
techniques or methods that have shown
promising results in effectively purifying
complex monoclonal antibodies?
JA: Utilizing innovative affinity technologies that target different
domains can be useful to leverage the unique structures of these
complex mAbs. That’s one technique that has shown promise.
Innovations looking at different optimization conditions for
polishing steps and unique polishing resins can be a useful
strategy as well. A lot of these complex mAbs may not be
as stable and can form higher-level aggregates, therefore
the ability to operate a wider set of conditions and separate
species using resins that might be more selective for different
product species can be useful.
NB: Affinity capture is the big space where we’re seeing major
changes and alternative mechanisms for purifying complex
mAbs. For bispecifics, they have both a lambda and kappa
light chain, which is something that you can target – you
can capture one side, then capture the other side. That’s a
perfect mechanism to remove all of the fragments of the parent
antibodies that would otherwise need convoluted polishing
strategies to remove.
KF: We recently launched the new POROS Caprylate, our
first mixed-mode cation exchange resin. As many may know,
caprylic acid is used to pull down high levels of aggregates
upstream. Attaching this to the bead allows aggregation to be
effectively removed downstream, reducing aggregation from
20% to less than 2% without the need to then remove the
caprylic acid. An added benefit is that this resin was designed
specifically to be used in flow-through mode to allow high
recovery over a wider range of loads compared to other resins
positioned for aggregate removal in flow-through mode.
Q: How do the scalability and throughput of
complex mAb purification processes compare
to traditional mAbs? And what considerations
need to be taken into account?
JA: It depends on the antibody and the specific challenges
present. Some of these complex mAbs can have lower
capacities, so you may need to utilize resins at lower loading
densities or, in order to achieve appropriate resolution, operate
at lower flow rates. Ultimately, this can lead to larger columns
as you scale up, increased buffer consumption and longer
processing times, which can reduce efficiency.
Therefore, it’s important to optimize early on and utilize different
strategies – e.g., using conditions that allow you to operate at
faster processing times and higher capacities to reduce that
burden as you scale up more complex mAbs.
NB: We’ve seen a need to prioritize resolution, and we
were seeing lower loading on columns through the entirethermofisher.com/antibody-derived-therapeutics 11
downstream process as we tried to get higher resolution. This
can mean more unit operations in manufacturing and taller
polishing columns that are more difficult to work with, as well
as big increases of buffer volumes and longer residence times.
So, the variations may become a little more complicated to
transfer and scale up. To alleviate these issues, many programs
are using very specific pH ranges or non-traditional buffers
(e.g., Good’s buffers such as MES or bicine) that we haven’t
seen used on a manufacturing scale historically. Pressuretolerant resins can also be very beneficial – you can get the
tall bed heights and the high resolution that you need. New
inline dilution strategies to make the buffer volumes more
manageable are also hugely beneficial when you’re scaling up.
Q: What role does process optimization play in
improving the efficiency and yield of complex
mAb purification? And what are some key
factors to consider during optimization?
JA: Process optimization plays a critical role because your goal
is to maximize purity and recovery and still maintain the quality
of the product. But you also want to ensure that it’s scalable,
robust and overall low-cost once you have a fully scaledup process. Monitoring the process is important to better
understand how it operates – ensuring that you’re not operating
at the edge of failure and identifying the parameters that could
impact your process
NB: Complex mAbs tend to have lower bioreactor titers overall
because they are more difficult to grow, so even a couple of
percent gain can be meaningful. Relying more heavily on DOEs
and quality by design frameworks can help labs to get the
most information possible with the least material, so that those
optimization strategies can be executed with just one batch.
Q: What are the potential future advancements
or innovations in complex mAb purification that
could address existing challenges and further
improve the process?
JA: Firstly, the continuing advancements of different affinity
technologies. Different affinity ligands target different domains
– so designing new ligands that are specific for different
domains or have different characteristics that can be leveraged
for separating difficult-to-resolve impurities in these complex
mAbs is important.
Another interesting advancement is the use of chromatography
modeling software. Mathematical models can reduce the
number of DOE experiments required and help us have a
better understanding of the process. We’re even starting to see
customers utilize AI or machine learning techniques for their
large datasets of different antibody sequences and structures.
AI can look at these datasets and use pattern identification to
predict the behavior of different chromatographic resins.
NB: The other thing that we’re seeing customers move towards
is process analytical technologies. Not all of these mAbs can
be tightly controlled just by looking at a UV signal. Having
inline or online detection or mass-based detection – instead
of just looking for chromophores that may not exist or may
completely overlap with your antibody – has real potential to
solve challenging separation issues that we may not be able to
resolve at scale.
KF: Achieving high purity is all about having the right resin for
your molecule. Chromatographic efficiency can be achieved
by reducing the particle size, but that comes with the expense
of flow rate or bead height due to the pressure limitations of
hardware. Modulating or designing the right chemistry or ligand
on the surface to be selective between target and impurities
is a more effective way to increase purity and yield. But as
molecules become more complex, this means more tools are
needed across different variations from the traditional mAb.
mAb developers can engineer in things like a C-tag to enable
selective removal or a specific sequence that interacts with
specific ion exchange or HIC groups.12
Advances in flow-through technology
to enhance mab polishing
Antibody therapeutics
Author
Robert Stairs
Field Applications Scientist,
Thermo Fisher Scientific
Summary
The rapidly evolving landscape of monoclonal antibody (mAb) development has
led to increasing demand for innovative purification approaches, particularly during
the polishing stage. Advances in flow-through technology have enhanced mAb
polishing, with a growing emphasis on hydrophobic interaction chromatography
(HIC) and mixed-mode chromatography. These approaches can remove challenging
species not easily removed by other methods, enhancing product quality and
manufacturability. This makes flow-through technology an essential tool to address
the increasing complexity and variety of mAbs in clinical development.
Increasingly complex antibody molecules
In recent years, the variety of mAbs has expanded beyond traditional IgG formats. The
emergence of alternative antibody derivatives such as antibody-drug conjugates (ADCs),
bispecific mAbs, Fab fragments and Fc-fusion proteins presents unique challenges (Figure 1).
For example, some of these formats display absent or altered protein A binding sites,
overexpression of free light chains or increased propensity for aggregation. These challenges
place considerable pressure on downstream development teams to continuously evolve
and maintain a robust purification approach.
Figure 1. Different mAb modalities that demand additional tools for efficient purification.
IgG Antibody–drug
conjugates
Bi-specific
antibodies
Fc-fusion proteins Fab fragmentsEfficient purification
thermofisher.com/antibody-derived-therapeutics 13
2). This makes scale-up, as well as optimization of flow rate and
process efficiency, more straightforward with respect to column
pressure-drop. The POROS base bead also has large throughpores, which reduces resistance to mass transfer. This translates
to more robust binding capacity and resolution with respect to
flow rate. Moreover, the bead itself has an average diameter of 50
µm. This size allows for a proper balance between resolving power
and the ability to maintain scalability and sufficient pressure flow
characteristics.
Chromatography can be operated in either bind-and-elute mode or
flow-through mode. In bind-and-elute mode, the resin binds both
the product of interest and impurities (such as aggregates), and
then the product of interest is selectively eluted from the column.
This mode is advantageous for separating closely related species,
Key considerations in downstream processing
Developing effective downstream antibody processing involves
balancing multiple factors. First and foremost, product quality
is of paramount concern. Chromatography resins must provide
high resolution to effectively separate the product of interest from
impurities, such as aggregates and host cell proteins (HCPs).
Ideally, resins should offer high capacity and throughput, allowing
researchers to minimize costs, reduce processing times and
manage intermediate pool volumes. POROS chromatography
resins are designed with these factors in mind and allow for simple
downstream processing.
The POROS base beads are comprised of polystyrenedivinylbenzene, a rigid polymer that results in a stable packed bed
and a linear relationship between flow rate and pressure (Figure
Figure 2. Key features of the POROS bead.
Linear pressure flow curve
0.0
0.5
1.0
1.5
2.0
2.5
3.0
0 200
Pressure (bar)
Flow ratehr(cm/ )
Conventional soft resin
17 cm
Figure 3. Comparison of key characteristics of POROS beads compared to select competitor products, highlighting higher capacity
and resolution, independent of flow rate.
POROS HIC resin performance
5 0
10
15
20
25
30
35
Competitor Ethyl POROS Ethyl Competitor low sub phenyl POROS Benzyl Competitor high sub phenyl POROS Benzyl Ultra
Lysozyme Capacity at 5%
Breakthrough (mg/mL)
POROS Competitor
50
10
15
20
25
30
35
40
0 500 1000 1500 2000
Capacity at 10%
breakthrough (mg/mL)
Linear Velocity (cm/hr)
Higher capacity and resolution compared to competitors -
independent of flow rate
8 HIC Presentation | July 2024
POROS resin
19 cm
400 600 800 1000
Poly(styrene-divinylbenzene) backbone Large through-pores 50-micron bead size
Linear velocity (cm/hr)
Superior resolution
Higher capacity and resolution compared to select
competitor products - independent of flow rate14 thermofisher.com/antibody-derived-therapeutics
such as charge variants. However, for aggregate and host cell
protein removal, comparable product quality can be achieved using
flow-through chromatography. In flow-through mode, only impurities
bind to the stationary phase, allowing for higher mass loading,
which results in reduced resin usage, fewer processing steps and
lower buffer consumption. All these benefits contribute to shorter
processing times and a smaller equipment footprint.
POROS hydrophobic interaction chromatography resins
The POROS HIC family of resins includes POROS Ethyl, POROS
Benzyl and POROS Benzyl Ultra resins. POROS Ethyl is the least
hydrophobic while POROS Benzyl and POROS Benzyl Ultra offer
higher hydrophobicity. POROS Benzyl Ultra is designed specifically
for flow-through mode under low-salt conditions. These POROS
HIC resins display higher capacity and resolution independent of
flow rate compared to competitors’ products (Figure 3). Moreover,
they display consistent lot-to-lot performance as well as the linear
pressure-flow drop, which is characteristic of the POROS base
bead, making them ideal for large-scale bioprocessing.
HIC can be used in a range of applications across different
therapeutic areas. For example, it can also be used for enzyme,
recombinant protein and virus purification. It is also useful for
reducing aggregates and other product and process-related
impurities during mAb purification, including ADC purification for
the resolution of individual drug–antibody ratio (DAR) species and
Fc fusion type molecules. The following case studies highlight the
effective use of flow-through applications for HIC.
Case Study 1: Optimizing a mAb purification polishing
step in flow-through mode using POROS HIC
chromatography in flow-through mode
The first case study focuses on mAb A, a clinical-stage antibody
with an existing process involving affinity capture, depth filtration,
low pH hold, anion exchange in flow-through mode and a mixedmode bind-and-elute step to reduce high aggregate levels (>12%)
(Figure 4). Despite achieving 99% monomer purity and 90%
recovery with the mixed-mode step, the throughput was limited to
25 g/L of resin at a 6-minute residence time.
Figure 4. Bind and elute experiment (a) performed with POROS Benzyl Ultra to determine the starting point for flow through
conductivity, with peak elution at ~7 mS/cm. (b) Heat map highlighting aggregate mass removal from high throughput screening
performed using POROS Benzyl Ultra and four salts (ammonium sulfate, sodium citrate, sodium acetate, sodium chloride) from
0–150 mM, pH 5.5–7.5. (c) Verification run to show effective reduction of aggregates in no-salt FT process with high recovery,
carried out at flow rate 500 cm/hr, 1.2 min residence time, load density 80 g/L. (d) Comparison of product quality, with FT showing
an 8% increase in monomer recovery and ~threefold increase in load density.
(c) (d)
5.0% 9.5%
85.5%
0.1% 0.5% 0.1%
99.3%
0.1%
mAb-A Process Mixed-Mode BE POROS Benzyl
Ultra-FT
Load density
(g/L resin) 25 80
Monomer purity
Pool (%) 99 >99
Monomer
recovery (%) 90 98
Residence time
(min) 6 1.2
HCP (ppm) <LLOQ <LLOQ
Load Purified Antibody
(a) (b)
pH 7.5
pH 6.5
pH 5.5
pH 6.8, ~2 mS/cm
mAB A: Bind-Elute POROS Benzyl Ultra
Absorbance 280 nm (mAu)
Conductivity (mS/cm)
Volume (mL)
HMW Dimer Monomer LMW
Conditions: 0–150 mM Salt, pH 5.5–7.5thermofisher.com/antibody-derived-therapeutics 15
To optimize this process, the mixed-mode step was replaced with
a flow-through POROS HIC step. The process development work
involved three stages:
1. Determining optimal flow-through conductivity: A lowloading bind-and-elute experiment was conducted with
a decreasing conductivity gradient to establish a starting
point for flow-through conductivity optimization.
2. High throughput screening (HTS): During this step, various
salt types, concentrations and pH levels were evaluated to
optimize conditions, focusing on POROS Benzyl Ultra.
3. Scale down model: Column loading studies were performed
to assess residence time effects.
Results showed that the POROS Benzyl Ultra resin operated in
flow-through mode provided comparable aggregate removal to the
mixed-mode separation operated in bind-and-elute mode. Although
product quality in terms of aggregate removal was equivalent
in both modes, the flow-through HIC step noticeably improved
recovery, with a boost of 8% (Figure 4). Similarly, column loading
capacity was almost three-fold higher with the flow-through HIC
step, with column loading increased to 80 g/L. Furthermore, the
residence time, or flow rate, was five times faster with the flowthrough HIC step. Thus, the flow-through HIC step matches the
product quality of the mixed-mode bind-and-elute step and is more
efficient with respect to binding capacity and flow rate, resulting in
productivity gains.
Case Study 2: POROS Benzyl Ultra viral clearance and
impurity removal in an ADC manufacturing process
The next case study involved the evaluation of the POROS Benzyl
Ultra resin for viral clearance and impurity removal during an
ADC manufacturing process. The company producing this
ADC utilizes synthetic amino acids that allow for site-specific
conjugation of the drug linker, creating a highly homogenous
DAR. However, this process can result in high levels of
aggregates (7–11%).
The POROS Benzyl Ultra resin was used to reduce high
molecular weight (HMW) species as well as host cell proteins for
pre-conjugated mAbs in three different processes. The results
showed good reduction of host cell proteins and HMW impurities
using high loading densities (Figure 5). In a viral clearance study
for mAb A, yield was comparable across qualification, XMuLVspiked and MVM-spiked runs, averaging 85%. A log reduction
value (LRV) of >5.97 was achieved for XMuLV and a LRV of 4.56
was achieved for MVM, demonstrating effective viral clearance of
a model parvovirus and retrovirus for this process.
Flow-through high aggregate mAb polishing using
POROS Caprylate Mixed-Mode Cation Exchange
Chromatography Resin
Thermo Scientific POROS Caprylate Mixed-Mode Cation
Exchange resin is a unique mixed-mode chromatography tool
designed for high aggregate selectivity in flow-through mode
that became commercially available in 2024. The ligand, caprylic
acid, imparts both hydrophobic and weak cation exchange
characteristics.
POROS Caprylate Mixed-Mode Cation Exchange resin is suitable
for moderate to high aggregate levels (up to 20%) and operates
over a broad pH (4.5–7.0) and conductivity range (10–30 mS/cm).
To demonstrate the aggregate removal capability of the resin
an IgG1 mAb was purified via Protein A capture and subjected
Figure 5. (a) Host cell protein reduction for three different mAbs using POROS Benzyl Ultra and (b) a summary of aggregate
removal for mAb A using two feed streams. *Post POROS Benzyl Ultra HMW levels for mAb B and C were <1%.
(a) (b)16 thermofisher.com/antibody-derived-therapeutics
Figure 6. Loading density studies, performed across three buffer conditions, to confirm that POROS caprylate can facilitate
effective polishing with high monomer yield and purity.
to high and low pH adjustments to generate up to 10% aggregate
in the feed stream. A bench-scale design-of-experiment (DOE)
was performed to evaluate the impact of pH (4.5–6.0) and sodium
chloride concentration (0–500 mM) on the responses of yield and
high molecular weight (HMW) reduction using POROS Caprylate
Mixed-Mode Cation Exchange resin in flow-through mode. Column
loading was kept constant at 100 g/L resin in the DOE. The results
showed >75% monomer recovery and robust aggregate removal
(<2% aggregate) across a wide range of conditions, with monomer
recovery expected to increase with higher column loading.
Next, loading density studies were performed at three different
conditions within the DOE space (Figure 6). For all three
conditions, <2.0% aggregate in the product pool was achievable
with ≥90% monomer recovery at 160–180 g/L loading density.
Additionally, HCP and leached Protein A were reduced by
approximately 95% for all three operating conditions. Further
characterization of HCPs by HPLC-MS/MS showed that POROS
Caprylate Mixed-Mode Cation Exchange resin was able to
reduce the number of individual HCP species from 380 to 79,
with complete removal of many HCPs considered to be high risk
or challenging to remove in mAb processes.
Conclusion
Advancements in flow-through chromatography technology,
particularly with POROS chromatography resins, offers significant
enhancements for polishing in mAb manufacturing processes.
The case studies highlighted here demonstrate the applications
and benefits of these advanced resins, paving the way for more
efficient and effective bioprocessing strategies.
Watch the complete webinar with Robert Stairs here.
25 mM Sodium acetate
25 mM Sodium acetate
25 mM Sodium acetate
POROS resins: Pharmaceutical Grade Reagent. For Manufacturing and Laboratory Use Only. © 2025 Thermo Fisher Scientific Inc.
All rights reserved. All trademarks are the property of Thermo Fisher Scientific and its subsidiaries unless otherwise specified. COL026398
Learn more at thermofisher.com/mixed-mode-chromatography5 key considerations for optimizing resin
selection for efficient mab purification
Introduction
Monoclonal antibodies (mAbs) have become a major class
of therapeutics and are now approved for clinical use in
various fields, including cancer, autoimmune disorders and
infectious diseases such as Ebola and COVID-19.1 In order
to adhere to regulatory guidelines, mAbs must meet certain
purity standards, making purification a critical step in the
bioprocessing workflow.
As the complexity of novel antibody modalities has increased
(e.g., antibody–drug conjugates, bispecific antibodies, etc.),
traditional purification techniques have struggled to meet
required purity levels while also maintaining high yields and
cost-effective processes. Complex engineered antibodies
can be prone to aggregation and fragmentation and may
have increased sensitivity to pH. In addition, they may also
have altered or absent protein A binding sites, complicating
purification further.
Recent advances in purification methods have resulted in the
emergence of novel affinity and polish purification tools that
can help to overcome many of these challenges. Something as
simple as selecting the optimal resin for purification can help
achieve high purity and efficient yields. This guide will discuss
some of the key considerations for mAb purification, helping
you to optimize your purification and polishing processes while
still balancing cost efficiency.
1. Addressing challenges in complex purification
Although leaps are being made in polishing and purification
processes, inherent challenges in the purification of complex
mAbs still arise at every stage of the mAb manufacturing
process. Cellular expression systems lead to high levels
of process-related impurities, such as residual host cell
proteins (HCPs) and host cell DNA. In addition, the inherent
heterogeneity of mAbs leads to product-related impurities such
as aggregates, misformed products and post-translational
modifications.
These impurities must be removed before product release to
meet stringent regulatory and safety guidelines and prevent
adverse effects in recipients. This is a complex process,
especially for novel antibody modalities. Instead of relying on
traditional purification and polishing methods, look to recent
developments and emerging tools in mAb processing for
solutions to complex challenges.
Hydrophobic interaction chromatography (HIC) (which
removes proteins based on hydrophobic interactions
between resins and target proteins), and mixed-mode
chromatography (MMC), which typically combines ion
exchange and HIC properties for improved selectivity, are two
such alternative processes, already addressing a range of
challenges.2,3 For example, Chinese hamster ovary (CHO) cells
are one of the most common cell lines for mAb production.
17 thermofisher.com/antibody-derived-therapeuticsHowever, these cells produce several notoriously difficult-toremove HCPs, including clusterin, which can co-elute with the
target mAb.4 MMC has been shown to be effective in removing
or significantly reducing the most challenging and high-risk
HCPs to acceptable levels.5
2. Evaluating affinity chromatography options for
mAb capture
Affinity chromatography is widely accepted as one of the most
efficient and effective techniques for the initial stages of mAb
purification, due to its specificity, ease of operation, and high yields,
purity, and throughput.6 While other chromatography methods
rely on separation by differences in size or ionic charge, affinity
chromatography relies on specific binding interactions between an
immobilized ligand and the mAb to be purified (Figure 1).
Due to the specificity of affinity chromatography, it is important
to select the right resin for the mAb in question. The most
common resin for affinity purification is protein A, which binds
primarily to the Fc region between the CH2 and CH3 domains
of most IgG subtypes.7,8
Although protein A is considered a trusted resin of choice
for the purification of traditionally structured mAbs, it can
underperform with novel modalities such as bispecific
antibodies (bsAb), due to formats that produce Fc-containing
mispaired product variants, formats that have modified Fc
regions that limit or eliminate binding to protein A, or fragmentbased antibody formats. In addition, the acidic elution used
during protein A chromatography can lead to aggregation of
some modalities, making impurity removal more difficult. In
these cases, alternative resins such as protein L are available.
Though still requiring a low pH, protein L binding is specific
for the variable light chains and can bind most antibody
classes and fragment fragment antigen-binding region (Fab)
fragments.6,9 Other resins based on camelid-derived singledomain antibody fragments have been developed specifically
for bsAbs and antibody fragments. These come in a range of
different binding sites and have high affinity, specificity and
capacity, allowing for a streamlined workflow and high-purity
products.10 They can also enable mild elution, preventing pHinduced aggregation.
In addition to careful resin selection, recovery rates can also be
maximized by optimizing the purification process and its steps.
If bind/elute steps are providing suboptimal monomer recovery,
changing the workflow to a flow-through (FT) operation using
a HIC resin can improve recovery. HIC resins are particularly
well suited for manufacturing-scale workflows, due to their high
linear binding capacity over a wider range of flow rates.
3. Strategies for effective polishing steps
Following mAb affinity capture, product purity of non-complex
antibodies may be as high as 95%. For more complex,
engineered mabs, purity levels of 80% or below are not
uncommon. To achieve higher purity, the next steps in the
downstream mAb purification process are intermediate and
final polishing. This typically consists of a cation exchange
chromatography step (CEX), followed by anion exchange
chromatography (AEX) and sometimes HIC. For relatively clean
feeds, a single polishing step may be sufficient, provided the
product purity and safety requirements are met.
Selecting an appropriate resin combination along with
optimized process conditions enables orthogonal separations
that maximize yields, reduce process and product-related
impurities, and improve process robustness. Resins should
align with the purification step and the charge properties of
the target molecule or impurities to be removed. In AEX, the
chromatography matrix is positively charged, so negatively
charged molecules can be captured. For example, impurities
such as viruses, HCPs, DNA, endotoxins and aggregates
can be captured and removed from the final product. In
comparison, CEX captures positively charged mAbs, which can
then be eluted by increasing the conductivity or buffer pH.
For complex mAbs, different polishing strategies may be
needed. IEX using standard conditions may result in inadequate
impurity removal, risking non-adherence to regulatory guidelines.
HIC and MMC offer unique selectivity and can often be more
Figure 1. Protein A chromatography.
Sample
Washing
buffer
Elution
buffer
Purified
antibodies
Unwanted
components
Target
antibodies
Affinity
resin
Purification
column
Sample loading Washing Elution
18 thermofisher.com/antibody-derived-therapeuticseffective platforms for novel mAb modalities. HIC resins can be
utilized in either bind and elute or flow-through mode, depending
on the hydrophobicity of the resin, the salt concentration and the
salt type used (e.g., kosmotropic salts like ammonium sulfate).
This allows for highly customizable options for targeting specific
impurities, such as product variants in bind and elute mode or
removal of aggregates and HCPs in flow-through mode. HIC
resins can support high resolution, even under low conductivity
conditions often encountered post protein A, making HIC an
excellent option for manufacturing scalability. HIC resins are
often utilized for the purification of engineered mAbs, including
antibody fragments and antibody–drug conjugates. MMC
combines the benefits of IEX and HIC for even greater selectivity
and purification due to a unique stationary phase that contains
both hydrophobic and charged functional groups (Figure 2). MMC
is ideal for a diverse range of proteins and particularly challenging
mAbs with high aggregate levels and complex impurity profiles,
as it can reduce 10% aggregation to around 1% in a single polish
step.3
4. Enhancing recovery rates through optimal resin
selection
A generic purification workflow may be able to meet regulatory
guidelines, but if it isn’t achieving maximal recovery rates, it
could be costing you product and efficiency. Additionally, a
process that works well at a pilot or development scale may
not run efficiently at a manufacturing scale. It’s important to
optimize your workflow in a manner that maximizes recovery
rates while still maintaining high purity and efficiency. To achieve
this, resin screening studies can be used to select the resins
with the highest binding capacity, best selectivity and best
resolution for the antibody in question.
Recovery can be affected by resin characteristics such as
binding capacity, elution efficiency and stability, therefore these
are important aspects to consider when selecting the best
resin. Some engineered antibodies can have altered, blocked
or absent protein A binding sites, causing issues for traditional
protein A-based purification. In these cases, specialized
matrices can be used, designed to target different binding
sites while still maintaining high dynamic binding capacity. For
example, for Fab purification, ligands that bind the constant
heavy (CH1) domain of IgG subclasses, or the kappa and
lambda Fab regions, can be used. For Fc-fusion proteins and
chimeric antibodies, ligands that bind only the constant heavy
domain (CH3) are available.
Resin selection is also key in polishing steps. While IEX is often
considered as a first choice for mAb polishing, HIC resins are
an effective alternative to address high levels of aggregation or
in antibody–drug conjugates (ADCs), where further purification
is needed to refine the drug–antibody ratio (DAR). HIC resins
can operate using various salt types, over a range of salt
concentrations, and come in a range of hydrophobicities. They
offer improved resolution and can help achieve enhanced
clearance of aggregates with higher productivity. You can also
consider mixed-mode (MMC) resins. As previously mentioned,
these resins combine properties of IEX and HIC, and they can
remove even high levels of aggregates in a single flow-through
step, thereby reducing the number of polishing steps and
improving efficiency.11
5. Balancing cost and performance in resin
selection
The core goal of purification is to isolate a high-purity product
in a reliable and cost-efficient manner. In addition to ensuring
optimal performance, economic aspects – such as initial
costs, lifespan and regeneration capabilities – must also be
considered when selecting a resin.
Reducing the number of steps in the downstream purification
process can improve cost-efficiency, as it reduces processing
time and is faster to optimize. A streamlined process can
also reduce the amounts of reagents needed and the labor
and utilities costs. For example, high-performing techniques
such as affinity chromatography for purification and MMC
for polishing can achieve higher yield and purity than other
methods in a single step, increasing efficiency without
compromising recovery. To streamline IEX steps, resins that
have large through-pore structures can allow higher capacity,
while resins optimized for low to high flow rates can help
ease the scale-up process from small-scale to process-scale
production.
O
O
OH
Hydrophobic
Weak cation
exchange
Figure 2. The structure of an MMC resin backbone.
19 thermofisher.com/antibody-derived-therapeuticsPharmaceutical Grade Reagent. For Manufacturing and Laboratory Use Only. © 2025 Thermo Fisher Scientific Inc. All
rights reserved. All trademarks are the property of Thermo Fisher Scientific and its subsidiaries unless otherwise specified. WTP-
10912755 0525
Ensuring protocol validation and compliance can be complex,
costly and time-consuming. Therefore, consider resins that
can help address these issues; for example, resins with animalfree origins can help ease adherence to regulatory guidelines.
Good availability and quality of technical support from the resin
vendor can also help with validation and optimization, while
choosing a vendor with a reliable supply chain and a proven
history of delivering high-quality product can help mitigate the
risk of unforeseen circumstances and facilitate optimal product
manufacture.
Resin reuse is another effective method of improving costefficiency, though this should be approached carefully so as
not to affect performance. Consider the chemical stability
of the resin, dynamic binding capacity over several cycles,
monitoring of product quality attributes, and other parameters
when assessing a column lifecycle. For example, single-domain
VHH antibody fragment affinity resins are inherently stable,
making them suitable for large-scale processing.
Conclusion
The ultimate goal of every mAb purification process is to
produce a high-quality therapeutic that meets stringent efficacy
and patient safety guidelines. However, it is also important to
consider workflow productivity, in order to keep therapeutics
affordable and manufacturing efficient. Significant leaps
in efficiency and performance can be achieved by simple
changes, such as informed resin selection. Selecting the right
resin for your process can address aggregation challenges,
improve cost-efficiency and maximize recovery rates, while still
adhering to regulatory guidelines. Optimize your purification
workflow today and see the real-world benefits to your mAbs.
References
1. Lyu X, Zhao Q, Hui J, et al. The global landscape of approved antibody therapies.
Antib Ther. 2022;5(4):233-257. doi:10.1093/abt/tbac021
2. Hydrophobic Interaction Chromatography. Thermo Fisher Scientific - US.
https://www.thermofisher.com/uk/en/home/life-science/bioproduction/
poros-chromatography-resin/bioprocess-resins/hydrophobic-interactionchromatography.html. Accessed May 7, 2025.
3. Mixed-Mode Chromatography. Thermo Fisher Scientific - US. https://www.
thermofisher.com/uk/en/home/life-science/bioproduction/poroschromatography-resin/bioprocess-resins/mixed-mode-chromatography.
html. Accessed May 7, 2025.
4. Singh SK, Mishra A, Yadav D, Budholiya N, Rathore AS. Understanding the
mechanism of copurification of “difficult to remove” host cell proteins in rituximab
biosimilar products. Biotechnol Prog. 2020;36(2):e2936. doi:10.1002/btpr.2936
5. Kiyonami R, Melani R, Chen Y, De Leon A, Du M. Applying UHPLC-HRAM MS/
MS method to assess host cell protein clearance during the purification process
development of therapeutic mAbs. Int J Mol Sci. 2024;25(17):9687. doi:10.3390/
ijms25179687
6. Arora S, Saxena V, Ayyar BV. Affinity chromatography: A versatile technique for
antibody purification. Methods. 2016;116:84-94. doi:10.1016/j.ymeth.2016.12.010
7. Protein A Chromatography Resins. Thermo Fisher Scientific - US. https://
www.thermofisher.com/uk/en/home/life-science/bioproduction/poroschromatography-resin/bioprocess-resins/protein-a-chromatography-resins.
html. Accessed May 7, 2025.
8. Hober S, Nord K, Linhult M. Protein A chromatography for antibody purification. J
Chromatogr B. 2007;848(1):40-47. doi:10.1016/j.jchromb.2006.09.030
9. Antibody Purification Using Protein L. Thermo Fisher Scientific - US. https://www.
thermofisher.com/uk/en/home/life-science/antibodies/antibody-purificationkits-reagents/antibody-purification-using-protein-l.html. Accessed May 7,
2025.
10.Affinity Chromatography Resins—Antibody Therapeutics. Thermo Fisher Scientific
- US. https://www.thermofisher.com/uk/en/home/life-science/bioproduction/
poros-chromatography-resin/bioprocess-resins/antibody-derivedtherapeutics.html. Accessed May 7, 2025.
11. Chen Y, de Leon A, Flook K. An innovative approach to addressing high aggregate
challenges in engineered monoclonal antibodies. Thermo Fisher Scientific
Bioproduction. https://assets.thermofisher.com/TFS-Assets/BPD/posters/
high-aggregate-engineered-monoclonal-poster.pdf. Accessed May 7, 2025.
20 thermofisher.com/antibody-derived-therapeuticsResources
Gain insight into an innovative
solution for mixed-mode
chromatography, specifically
designed to remove high level of
aggregates in the downstream
process of therapeutic antibodies.
Read more
Learn about a variety of CaptureSelect
resins for affinity purification to
address today’s challenges in the mAb
therapeutics workflow.
Read more
Find your ideal resin match today at
thermofisher.com/resin-selection-tool
POROS: Pharmaceutical Grade Reagent. For Manufacturing and Laboratory Use Only. CaptureSelect: For Research
Use or Further Manufacturing. Not for diagnostic use or direct administration in humans or animals © 2025 Thermo
Fisher Scientific Inc. All rights reserved. All trademarks are the property of Thermo Fisher Scientific and its subsidiaries unless
otherwise specified. TCN-11374750 0725
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