Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Journal of Pharmaceutical Sciences 110 (2021) 3811−3818

Contents lists available at ScienceDirect

Journal of Pharmaceutical Sciences


jo urn a l h om ep ag e : w ww.jp harms ci.o rg

Pharmaceutical Biotechnology

A Highly Sensitive LC-MS/MS Method for Targeted Quantitation of


Lipase Host Cell Proteins in Biotherapeutics
Yunqiu Chena,*, Chong-Feng Xua, Bradley Stanleya, Greg Evangelista,
Alex Brinkmanna, Suli Liua, Sean McCarthyb, Lei Xiongb, Elliott Jonesb, Zoran Sosica,
Bernice Yeunga
a
Biogen, Cambridge, MA, USA
b
SCIEX, Redwood City, California, USA

A R T I C L E I N F O A B S T R A C T

Article history: Identification and accurate quantitation of host cell proteins (HCPs) in biotherapeutic drugs has become
Received 4 May 2021 increasingly important due to the negative impact of certain HCPs on the safety, stability, and other product
Revised 23 August 2021 quality of biotherapeutics. Recently, several lipase HCPs have been identified to potentially cause the enzy-
Accepted 23 August 2021
matic degradation of polysorbate, a widely used excipient in the formulation of biotherapeutics, which can
Available online 28 August 2021
severely impact the stability and product quality of drug products. In this study, we identified three lipase
Edited by Editor Name: Dr K Audus. HCPs that were frequently detected in Chinese hamster ovary (CHO) cell cultures using shotgun proteomics,
including phospholipase B-like 2 (PLBL2), lipoprotein lipase (LPL), and lysosomal acid lipase (LIPA). A targeted
Key Words:
quantitation method for these three lipase HCPs was developed utilizing liquid chromatography coupled
Biopharmaceutical characterization
with tandem mass spectrometry (LC-MS/MS) with high-resolution multiple-reaction-monitoring (MRMhr)
Monoclonal antibody(s)
quantitation. The method demonstrated good sensitivity with low limit of quantitation (LLOQ) around
Liquid Chromatography-Mass Spectrometry
(LC-MS) 1 ng/mL, and linear dynamic range of three orders of magnitude for the three lipase HCPs. It has been applied
Mass spectrometry for the characterization of process intermediates from various in-house monoclonal antibody (mAb) produc-
Proteomic tion. In addition, the method has also been used to evaluate the robustness of clearance for one of the lipase
HCPs, PLBL2, under different column purification process conditions.
© 2021 The Authors. Published by Elsevier Inc. on behalf of American Pharmacists Association. This is an open
access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/)

Introduction where the latter in turn can form insoluble particles that affect the
stability and product quality of biotherapeutics.8−10
Host cell proteins (HCPs), the low-level protein impurities derived Several lipase HCPs have been previously identified to cause poly-
from the host cell line that co-purify with protein therapeutics, must sorbate degradation in biotherapeutic formulations, including phos-
be sufficiently and robustly removed from the final drug product to pholipase B-like 2 (PLBL2),4 lipoprotein lipase (LPL),3 Group XV
an acceptable level. Residual HCPs can have detrimental effects on lysosomal phospholipase A2 Isomer X1 (LPLA2),5 phospholipase A2
biopharmaceutical products, including unwanted immunogenic group VII (PLA2G7),11 and liver carboxylesterase,12 among others. In
effects in patients as well as an impact on stability and efficacy of addition, there could be other HCPs that potentially possess the lipase
drugs.1−2 For example, recently, it has been demonstrated that cer- activities that contribute to the degradation of polysorbates, but their
tain lipase HCPs can be the major cause for enzymatic degradation of functions are yet to be identified or confirmed.13 These lipase HCPs
polysorbate 20 and 80, which are common surfactants present in for- are found to often co-purify with many monoclonal antibodies
mulations of biotherapeutics.3−5 Polysorbate 20 and 80 are used in (mAbs) and other biotherapeutics produced in Chinese hamster
formulations for both preventing surface adsorption and as stabil- ovary (CHO) cells.14,15 As a result, they can affect drug substance sta-
izers against protein aggregation.6,7 Enzymatic degradation of poly- bility and need to be removed from the drug substance to acceptable
sorbate 20 and 80 through hydrolysis of ester bond can result in the levels. It has been demonstrated that lipases present as low as less
formation of degraded polysorbates and free long-chain fatty acids, than 1 ppm can cause significant degradation of polysorbates.5
The total amount of HCP is mostly measured by an enzyme-linked
immunosorbent assay (ELISA) method, which is regarded as an
* Corresponding author at: Biogen Inc, 225 Binney Street, Cambridge, MA 02142,
United States. industry “gold standard” method for the total HCP quantitation for
E-mail address: rachel.chen@biogen.com (Y. Chen). in-process testing and product release.16,17 Though a high-

https://doi.org/10.1016/j.xphs.2021.08.024
0022-3549/© 2021 The Authors. Published by Elsevier Inc. on behalf of American Pharmacists Association. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
3812 Y. Chen et al. / Journal of Pharmaceutical Sciences 110 (2021) 3811−3818

throughput, sensitive and robust method, ELISA-based HCP quantita- across three orders of magnitude. The reproducibility and accuracy
tion is highly dependent on the HCP coverage of the capture antibod- of the method were also thoroughly evaluated. The method has
ies and can miss the detection of certain HCP species.16−18 More been applied in process development and characterization for the
recently, an orthogonal liquid chromatography coupled with a tan- monitoring of lipase clearance in downstream processes, as well
dem mass spectrometry (LC-MS/MS)-based proteomics method has as to demonstrate the robustness of lipase HCP clearance in a
been increasingly utilized in the profiling of HCP population and spike-in study.
abundance during process development and characterization.19−21
The proteomics-based method has the advantage of both identifying
Material and Methods
individual HCP species and providing relative quantitation of their
abundance. Since the identification and quantitation can be per-
Materials
formed simultaneously on thousands of HCP species, the proteomics-
based method also has the benefit of providing information on
The peptide standards used for lipase quantitation were synthe-
critical HCPs. However, despite continuous improvement on the rela-
sized from New England Peptide (Gardner, MA, USA). A total of 18
tive quantitation capability of the shotgun proteomics, it remains a
peptides were synthesized, including nine “light” peptides (not
challenge to accurately quantify individual HCPs, especially HCPs at
labeled) and their heavy-isotope-labeled (13C6,15N2-Lysine or
extremely low levels (<10 ppm), in a fast and high-throughput 13
C6,15N4-Arginine) counterparts, which serve as internal standards
manner.22,23
(IS) (Table 1). The recombinant PLBL2 protein was obtained from
Due to the criticality and high risk of the lipase HCPs, several tar-
MyBiosource, Inc. (San Diego, CA, USA). Urea, Tris, dithiothreitol
geted quantitation methods have been developed to specifically mea-
(DTT), and iodoacetamide (IAM) were from Sigma-Aldrich (St. Louis,
sure the level of lipase HCPs in process and drug substance samples.
MO, USA). Trypsin/Lys-C mix (mass spec grade) was from Promega
These include lipase activity assays that measure the hydrolysis rate
(Madison, WI, USA). Trifluoroacetic acid (TFA), formic acid (FA), water
of lipid-mimic substrates with fluorescent or colorimetric detec-
and acetonitrile were LC-MS grade and from Thermo Fischer Scien-
tion,24 immuno-assays including ELISA and Western blot methods
tific (Waltham, MA, USA). The protein drug substance and process
that target specific lipases,25 activity-based protein profiling (ABPP)
intermediate samples, including cell culture harvest sample to be
approach for serine hydrolase enrichment and identification,11 as
loaded onto Protein A affinity column (ProA load), Protein A column
well as LC-MRM (multi-reaction monitoring)-based targeted quanti-
purified product (ProA eluate), the purified product from Column 2
tation methods.3,5,12,25 The LC-MRM-based method has long been
and Column 3, were produced at Biogen.
used as method of choice for selective, sensitive and accurate quanti-
tation to support pharmacokinetic assessment of small molecules
and biotherapeutics. Recently, Gao et al. has demonstrated the sensi- Sample Digestion
tivity, robustness and accuracy of an LC-MRM-based quantitation
method for two lipase HCPs, PLBL2 and LPLA2, and that the results Samples (»300 mg therapeutic proteins) were denatured and
were comparable to the ELISA-based method.25 reduced by adding 75 mL of 8 M urea, 5 mL of 1 M Tris, pH 8.0, and
In this work, a high-resolution LC-MRM (LC-MRMhr) based tar- 1 mL of 500 mM DTT to a final volume of 100 mL. They were incu-
geted quantitation method was developed for the quantitation of bated at room temperature (RT) for about 30 min. After alkylation by
three potentially high-risk lipase HCPs, including PLBL2, LPL and adding 8 mL of 200 mM IAM and incubation at RT for 30 min in the
lysosomal acid lipase (LIPA), which have been detected by a shot- dark, the samples were diluted 6-fold by adding 492 mL of 50 mM
gun proteomics method in multiple CHO-based mAb processes. Tris (pH 8.0), and digested with 20 mg Trypsin/Lys-C mix overnight
With synthesized peptides used as quantitation standards and at 37°C. Six mL of TFA was added to 0.1% (v/v) to stop the digestion. A
heavy-isotope-labeled peptides as internal standards, the method mAb drug substance (mAb-A) was also prepared in the same way
demonstrated sensitivity around 1 ng/mL and dynamic range and used as matrix for standard curve.

Table 1
Three lipase HCPs Identified From Shotgun Proteomics Analysis, and the Optimized MRMhr Transitions Used in the LC-MS/MS Quantitation Method.

Protein Size (kDa) Peptide Retention Peptide Sequence MRM Transition Collision Energy (eV)
Number Time (min)
Precursor(charge) Product Ion (fragment type)

lipoprotein lipase (LPL) 52.8 1 11.1 GLGDVDQLVK 522.29 (2+) 873.46 (y8) 23
GLGDVDQLVK* (IS) 526.30 (2+) 881.48 (y8)
2 17.8 LSPDDADFVDVLHTFTR 649.99 (3+) 661.34 (y5) 35
LSPDDADFVDVLHTFTR* (IS) 653.32 (3+) 671.35 (y5)
3 16.7 SIHLFIDSLLNEENPSK 652.67 (3+) 817.36 (y7) 30
SIHLFIDSLLNEENPSK* (IS) 655.35 (3+) 825.38 (y7)
lysosomal acid lipase/ 45.6 4 18.8 WPEVIIEDLFGHK 528.28 (3+) 698.87 (y122+) 25
cholesteryl ester WPEVIIEDLFGHK* (IS) 530.95 (3+) 702.88 (y122+)
hydrolase-like (LIPA) 5 7.3 VNVYTSHSPAGTSVQNLR 643.99 (3+) 858.94 (y162+) 27
VNVYTSHSPAGTSVQNLR* (IS) 647.33 (3+) 863.94 (y162+)
6 7.2 GNTWSLK 403.22 (2+) 533.31 (y4) 20
GNTWSLK* (IS) 407.22 (2+) 541.32 (y4)
putative phospholipase 65.4 7 12.5 LTLLQLK 414.78 (2+) 614.43 (y5) 20
B-like 2 (PLBL2) LTLLQLK* (IS) 418.79 (2+) 622.44 (y5)
8 11.2 SVLLDAASGQLR 615.34 (2+) 817.41 (y8) 30
SVLLDAASGQLR* (IS) 620.35 (2+) 827.42 (y8)
9 8.4 AFIPNGPSPGSR 600.31 (2+) 868.41 (y9) 25
AFIPNGPSPGSR* (IS) 605.32 (2+) 878.43 (y9)
* heavy-isotope-labeled peptides (13C6,15N2-Lysine or 13C6,15N4-Arginine) used as internal standards
Y. Chen et al. / Journal of Pharmaceutical Sciences 110 (2021) 3811−3818 3813

Shotgun Proteomics for HCP Identification phases consisted of 0.1% (v/v) formic acid in water (A) and in acetoni-
trile (B). The LC gradient starts with 2% B for 2 min, then increases to
Six hundred mg of samples were spiked with 0.1 mg (»167 ppm) 10% B within 1 min, ramping up to 35% B within 19 min, followed by
of yeast enolase, which serves as an internal standard for label-free 1 min wash step at 80% B and re-equilibration at 2% B for 10 min. The
quantitation. The samples were digested in a similar way as described LC eluate before 4 min and after 20 min was diverted to waste. The
in the above section. After digestion, 185 mL of the digest was puri- mass spectrometer is operated in positive mode using a DuoSprayTM
fied using Bravo desalting RPS cartridges (Agilent AssayMap). The Turbo V Ion Source with the following parameters: curtain gas at 30;
desalted samples were speed vacuumed to dryness and re-consti- ion source gas 1 and 2 at 50; temperature at 600°C; ion spray voltage
tuted in 0.1% (v/v) formic acid. Twenty mL of sample (50 mg) was at 5500 eV. The mass spectrometry acquisition starts with a full scan
loaded for LC-MS analysis using an Orbitrap Fusion mass spectrome- in the TripleTOFÒ 6600 System with 0.1 s acquisition time, followed
ter (Thermo Scientific, MA, US) and the LC separations were per- by “Product Ion” scans with 200 ms acquisition time using high sensi-
formed using a Waters Acquity CSH column (1.8 mm, 2.1 mm £ 15 tivity mode, with “enhance mass” around the m/z of the targeted
cm) using 0.1% (v/v) formic acid buffers containing water and aceto- product ion (detailed in the Results and Discussion section). The MS/
nitrile. The LC gradient started with a 5 min hold at 2% B, a 85 min MS acquisitions were scheduled to occur within three LC time peri-
gradient elution from 3% to 25% B, ramping to 35% B within 20 min, ods: 4 to 10 min; 10 to 15 min; 15 to 20 min. MRMhr transitions
followed by washing at 80% B for 5 min and re-equilibration for were multiplexed according to the peptide elution time, and three
10 min. Electrospray ionization (ESI) parameters on the Orbitrap pairs of peptides (3 light and 3 heavy-labeled) are scheduled within
mass spectrometer included spray voltage at 3.5 kV, ion transfer tube each LC time period. The data was processed by extracting the tar-
temperature at 325°C, vaporizer temperature at 275°C, sheath gas geted product ions using 0.1 Da mass window using MultiQuantTM
and auxiliary gas at 35 and 10 arbitrary unit, respectively. The mass Software 3.0 (Sciex, CA). Calibration curves were calculated based on
spectrometer detection included a full scan in the orbitrap (m/z 300 the linear regression of peak area ratio (peak area of analyte divided
to 1500) with resolution at 120k and data-dependent MS2 scans on by IS) against the analyte concentration, applying a weighting factor
the top abundant ions within a cycle time of 1 s. Precursor ions were of 1/x. Assuming a complete digest, the obtained peptide concentra-
fragmented using HCD at 25% collision energy and detected in the tion can be used to calculate the lipase HCP concentration using the
ion trap. The LC-MS raw files were searched against a custom data- following equation:
base that comprises the CHO proteome database (CHO_refseq down- HCP concentration ðg=LÞ ¼ peptide concentrationðmol=LÞ  size of
loaded from https://www.chogenome.org/) and the sequences of HCP ðg=molÞ
yeast enolase and protein therapeutics, using the Sequest search The relative content of HCP compared with mAb protein
engine with the Proteome Discoverer software (ver. 2.1, Thermo Sci- (expressed as ng/mg or ppm) can be further calculated by dividing
entific). The HCPs were quantitated by a “Hi3” label-free method, the HCP concentration (ng/mL) by the mAb concentration (mg/mL) in
where the top-three most abundant peptides from each HCP were the digest.
used for quantitation. The peak areas of HCPs were compared with
the spiked-in yeast enolase and an estimated ppm value was calcu- Results
lated.
Identification of Lipase HCPs Using Shotgun Proteomics
Internal Standard and Standard Curve Preparation
To identify the lipase HCPs present in the in-house mAb processes
Nine heavy-isotope-labeled peptides were dissolved in using the CHO host, a shotgun proteomics characterization was per-
50:50 ACN/water and mixed to a final concentration of 5 mM as the formed on cell culture harvest and Protein A column eluate samples
IS stock solution. The nine light peptides were prepared in a similar from multiple in-house produced mAbs. The data was searched
way to a 5 mM standard stock solution. The IS stock was further against the CHO proteome database. Several HCPs with potential
diluted to 100 nM using matrix digest during sample preparation. enzymatic activities were identified and three lipase-related HCPs
After sample digestion, 6 mL of IS solution (100 nM) was spiked in to were identified from multiple mAb samples, including lipoprotein
600 mL of sample digest or matrix digest to achieve 1 nM final con- lipase (LPL), lysosomal acid lipase/cholesteryl ester hydrolase-like
centration. The standard curve was prepared by spiking light peptide (LIPA) and putative phospholipase B-like 2 (PLBL2), as shown in
standard stock into the digested matrix using serial dilution to the Table 1. These three lipase HCPs are commonly present in multiple
following concentrations: 6.25, 12.5, 25, 50, 100, 250, 500, 1000, mAb cell culture samples. PLBL2 and LPL have previously been
2500 pM. The QC peptides were prepared from a separate stock with reported to be associated with polysorbate degradation,3−4 and LIPA
serial dilution to the following concentrations: 16, 80, 400 and was recently identified from an activity-based protein profiling
2000 pM. (ABPP) study as an active hydrolytic enzyme that may cause polysor-
bate degradation.11 Thus we decided to develop a platform, MRM-
Targeted Lipase LC-MS/MS Analysis based quantitation method for the three lipase HCPs in order to
achieve better sensitivity of detection and higher throughput to sup-
The digested samples were either directly injected into LC-MS or port process development.
purified using an Agilent Bravo desalting RPS cartridge before injec-
tion. For the desalting step, 185 mL of the digest was purified using LC-MRMhr Method Development for Lipase HCP Quantitation
RPS cartridges, speed vacuumed to dryness and re-constituted in
56 mL of 0.1% (v/v) formic acid, which equated to 3.3-fold concentra- From the list of peptides identified in the shotgun proteomics
tion of the sample. Twenty mL of samples or standards, containing experiment, three tryptic peptides from each lipase HCP were
10 mg (direct injection) or 33 mg (Bravo-desalted) protein, were selected for targeted quantitation (Table 1). These peptides were
loaded for analysis on a TripleTOFÒ 6600 mass spectrometer (SCIEX, selected as they had high MS signal intensity, low propensity to post-
CA, USA) coupled with a Shimadzu HPLC system. Both standards and translational modifications and no mis-cleavages. These peptides
samples are analyzed in duplicate. LC separation was performed on a were also confirmed to be unique peptides for each HCP protein in

Waters Acquity CSH C18 column (2.1 £ 100 mm, 1.7 mm, 130 A) the CHO proteome through database searching by Blastp. To achieve
using 0.2 mL/min flow rate and column temperature at 55°C. Mobile the purpose of absolute quantitation, the selected peptides were
3814 Y. Chen et al. / Journal of Pharmaceutical Sciences 110 (2021) 3811−3818

represent the samples to be analyzed and contain minimal interfer-


ence with quantitation. We screened several in-house and commer-
cially available purified mAbs and found that an in-house mAb-A
drug substance provided a relatively clean matrix with lowest back-
ground lipase detected. The mAb-A digestion matrix showed no
interference with peptide 5 and peptide 7 in the extracted ion chro-
matograms (XICs). It exhibited a background signal at around 10% of
LLOQ level for peptide 1, thus the impact on the quantitation is
deemed minimal (Supplementary Figure 2).
The MRMhr-based quantitation method demonstrated good sen-
sitivity for the selected lipase peptides. The low limit-of-quantitation
(LLOQ) of the nine selected peptides range from 6.25 pM to 100 pM,
as shown in Table 2. The peptide with the best sensitivity and optimal
linear range was chosen as the quantifier peptide for each lipase, i.e.,
peptide 1 (GLGDVDQLVK) for LPL, peptide 5 (VNVYTSH-
Fig. 1. A scheme of scheduled MRM. The LC-MS time is divided into three periods and SPAGTSVQNLR) for LIPA, and peptide 7 (LTLLQLK) for PLBL2. The
MRM scans of the 9 peptides are scheduled according to their elution time. (A) Total other peptides were also monitored simultaneously to provide addi-
ion chromatogram (TIC) of the matrix digest spiked with 10 nM synthetic peptides. (B)
tional confirmation of the quantitation results. The LLOQ for the
Extracted ion chromatograms (XICs) of the nine peptides with peaks labeled by pep-
tide number.
quantifying peptides was determined to be 12.5 pM for peptide 1
(LPL), 25 pM for peptide 5 (LIPA), and 6.25 pM for peptide 7 (PLBL2).
The signal-to-noise (S/N) ratio for the quantifying peptides is at least
chemically synthesized along with their heavy-isotope-labeled coun- 10 at the LLOQ level, as shown in Supplemental Figure 2. The assay
terparts (13C6,15N2-Lysine or 13C6,15N4-Arginine labeled), which are showed a linear dynamic range around three orders of magnitude for
used as quantitation standards and internal standards, respectively. all three lipases, with regression coefficient (r) above 0.99 (Fig. 2). A
We chose to use synthetic tryptic peptides as standards to develop detailed examination of the standard curve demonstrated that the
the method, as many HCPs are not commercially available as purified measured concentrations of peptide standards are within 80-115% of
recombinant proteins and attempting to purify individual HCPs in- the theoretical values for peptide concentration above LLOQ, and
house can be resource-intensive, especially when trying to produce within 130% accuracy at the LLOQ level, with variation less than 20%
and characterize the heavy-isotope labeled HCP proteins. Synthetic among triplicate analysis as shown in Supplementary Table 1.
peptides, on the other hand, can be easily designed and synthesized The LC-MRM method was evaluated for its intra-run and inter-run
to achieve a quick development timeline. reproducibility and quantitation accuracy using QC peptides as well
An LC-MS/MS-based MRMhr quantitation workflow was devel- as a mAb process intermediate sample. QC peptides were prepared at
oped for the nine tryptic peptides on a TripleTOFÒ mass spectrometer four different levels, including 16, 80, 400 and 2000 pM to cover a
coupled with reversed phase LC separation. The MRM transitions wide concentration range. They were prepared for two separate runs,
were optimized for the nine pairs of peptides, including 9 target pep- with three replicates for Run 1 and five replicates for Run 2. As shown
tides and their heavy-isotope labeled counterparts, as shown in in Supplementary Table 2, for peptide 1 (LPL), the accuracy of the QC
Table 1. We optimized the choice of precursor and product ions, as peptides was within 75%-110% for the intra-run, and 80%-105% for
well as the collision energy to achieve the optimal signal intensity inter-run. The precision was within 7% coefficient of variation (CV)
and selectivity. A scheduled MRM strategy was applied, where the both intra-run and inter-run for peptide concentration 80-2000 pM,
20-min LC gradient was divided into three 5-min windows and pepti- with higher variation within 25% CV for QC peptide at 16 pM. For
des are monitored only during their expected elution windows peptide 7 (PLBL2), the accuracy of the QC peptides was within 75-
(Fig. 1). The scheduled MRM was used to accommodate multiplexing 115% for the intra-run, and 80%-105% for inter-run. The precision
of peptides and maintain adequate scan time to ensure reproducibil- was within 15% CV intra-run and inter-run for peptide concentration
ity of peak integration and quantitation. 80-2000 pM, while the precision for 16 pM level was within 25% CV.
This LC-MRMhr workflow utilizes a hybrid triple quadrupole For peptide 5 (LIPA), the accuracy of the QC peptides was within 75-
time-of-flight (TOF) mass spectrometer, where the peptide precur- 115% for the intra-run, and 75%-105% for inter-run. The precision
sors were selected in the triple quadrupole and product ions are was within 15% CV intra-run and inter-run for peptide concentration
detected in the TOF analyzer with high mass accuracy. While the MS/ 80-2000 pM. In addition to QC peptides, to assess the reproducibility
MS scans are acquired in full scan mode in the TOF system, one prod- of sample preparation including enzyme digestion, a mAb process
uct ion from each scan was selected for monitoring using the intermediate sample was digested and analyzed in two different
“enhancement” function of the tripleTOF instrument. With enhance- runs, each with six separate preparations. The quantitation results of
ment mode, ions are briefly trapped in the collision cell and released peptide 1 (LPL), peptide 7 (PLBL2), and peptide 5 (LIPA) are 915 pM,
as a packet. The release of trapped ions is timed so that the m/z of 174 pM, and 108 pM, respectively. The intra-run variation is within
interest arrives in the accelerator precisely in time to be pushed into 20% CV (n = 6), and the inter-run variability is with 15% for peptide 1
the TOF. In this way it is possible to increase the duty cycle of a partic- and 7, and 24% for peptide 5 (n = 12) (Supplementary Table 3).
ular m/z to close to 100%, which in turn increases the signal intensi- The lipase HCP concentration in the sample can be calculated
ties of product ion and boosts method sensitivity. We found the based on the peptide concentration and the size of HCP, assuming
enhancement function increased the signal intensities of the selected a complete digest of HCP. Based on the size of three lipase HCPs
peptides by 2- to 4-fold across the concentration range from 50 pM (Table 1), the LLOQ of the lipase HCPs that can be quantified by
to 10 nM (Supplementary figure 1). At the same time, the high the LC-MRM method is estimated to be 0.7 ng/mL for LPL,
resolving power of the TOF analyzer can minimize the background 0.4 ng/mL for PLBL2, and 1.1 ng/mL for LIPA, as shown in Table 2.
interference from matrix and allow quantitation based on a single In HCP analysis, the relative content of HCP compared with the
isotopic ion. therapeutic protein, expressed as ng HCP per mg of mAb (ng/mg,
Matrix selection is very important for the LC-MRM method devel- or ppm), is very critical for monitoring HCP clearance during pro-
opment, as the selected matrix used for standard curve should cess development and for risk assessment of human exposure to
Y. Chen et al. / Journal of Pharmaceutical Sciences 110 (2021) 3811−3818 3815

Table 2
Low Limit of Quantitation (LLOQ) and Linear Range of the Lipase HCP Quantitation Method.

Protein Peptide Number Peptide LLOQ (pM) Linear Range (pM) Calculated LLOQ of HCP (ng/mL)b
a
Lipoprotein lipase (LPL) 1 12.5 12.5-2500
2 25 25-2500 0.7
3 50 50-2500

Lysosomal acid lipase (LIPA) 4 100 100-2500


5a 25 25-2500 1.1
6 25 25-2500

putative phospholipase B-like 2 (PLBL2) 7a 6.25 6.25-2500


8 12.5 12.5-2500 0.4
9 25 25-1000
a
Peptides denoted are quantifiers for each HCP.
b
LLOQ of HCP is calculated based on peptide LLOQ and the size of HCP, assuming a complete digestion.

Fig. 2. Standard curves for (A) peptide 1 (GLGDVDQLVK), (B) peptide 5 (VNVYTSHSPAGTSVQNLR) and (C) peptide 7 (LTLLQLK) quantitation. The linear regression (using 1/x weight-
ing) and the R values are shown.

HCP. We further calculated the relative concentration of lipase Analysis of Process Intermediate Samples to Understand Lipase HCP
HCPs compared with mAb in the digested sample. In the LC-MRM Clearance
method, the digested sample contains »0.5 mg/mL of mAb; it can
be further concentrated using a desalting cartridge by 3.3-fold The targeted HCP quantitation method was applied to monitor the
resulting in »1.67 mg/mL of mAb in the digested sample. For the abundance of lipase HCPs during each step of the downstream purifi-
direct injection runs, the calculated LLOQ of the three lipase HCPs cation process for several in-house produced mAbs. The mAb purifi-
are between 0.8 to 2.2 ppm, and for the runs with sample pre-con- cation process typically consists of three column steps, including the
centration, the LLOQ can reach <1 ppm level (supplementary Protein A affinity column and two additional polishing columns. In-
Table 4). We have examined that the standard curve for the direct process samples, including Protein A column load and eluate, the
injection runs and concentrated sample runs have no significant products from second and third columns, were analyzed by the LC-
difference in terms of LLOQ and dynamic range. The peptide at MRMhr method for lipase HCP monitoring. The quantitation results
LLOQ level can be reliably detected with sufficient S/N ratio with- for process intermediates from four monoclonal antibodies, including
out being affected by the concentration of matrix. two IgG1 mAbs (A and B), one IgG1/IgG4 hybrid mAb (C) and one
The accuracy of the method was also assessed by spiking known IgG4 mAb (D) are shown in Fig. 3.
amounts of recombinant PLBL2 into a mAb drug substance at four The quantitation results showed that the 3-column process in
levels, including 1, 5, 10, and 100 ppm, before digestion and the LC- general demonstrated the capability of effectively removing the three
MS/MS analysis. The quantitation results of PLBL2 based on peptide 7 lipase HCPs to a level lower or close to LLOQ for the four mAbs used in
is shown in Table 3. The accuracy falls between 100% and 121% for the analysis. However, a close examination of the lipase clearance
the spike-in range between 1 ppm and 100 ppm, and the %CV calcu- revealed that, for different mAbs and processes, the starting abun-
lated based on triplicate analysis are generally within 15%. It is worth dance of lipase HCPs and the clearance at each column step can be
mentioning that the quantitation results obtained from the other two highly divergent. For example, PLBL2 was detected with an abun-
peptides from PLBL2, peptide 8 and 9, are highly consistent with dance ranging from 200 to 800 ppm in the Protein A column load,
quantitation by peptide 7 without a significant peptide-to-peptide and its abundance in the Protein A column eluate was detected any-
bias (Supplementary Table 5). This spike-in experiment demon- where between <1 ppm to more than 100 ppm for the four mAbs.
strated that the method could provide accurate quantitation of PLBL2 For mAb A and mAb B (both IgG1 mAbs), the level of PLBL2 was
lipase HCP for mAb drug substance samples across a wide concentra- detected at lower than 5 ppm in the Protein A column eluate, which
tion range. Due to limited commercial availability, the quantitation can be translated to approximately 100-fold reduction by the Protein
accuracy for the other two lipase HCPs were not assessed in this A column purification. On the other hand, PLBL2 was present at »100
study. and »20 ppm for mAb C and mAb D after Protein A column. After the
3816 Y. Chen et al. / Journal of Pharmaceutical Sciences 110 (2021) 3811−3818

Table 3
Accuracy of quantitation demonstrated by recombinant PLBL2 spike-in study.
Shown are the quantitation results based on peptide 7. Digested samples were pre-
concentrated using a desalting cartridge before injection into LC-MS analysis. Each
spiked sample was analyzed in triplicate with %CV shown.

PLBL2 Measured Calculated %CV (N = 3) %Accuracy


spike-in peptide PLBL2
amount (ppm) 7 conc. (pM) amount (ppm)

1 26.3 1.0 6% 103%


5 155.5 6.1 12% 121%
10 289.9 11.3 4% 113%
100 3004.9 117.0 6% 117%

Column-2, PLBL2 was cleared to <LLOQ level for mAb C, while for
mAb D (IgG4), PLBL2 was still present at »7 ppm. It was further
removed by Column-3 down to a level that is below LLOQ. It is recog-
nized that the process conditions for the four mAbs, such as wash
buffer, pH, loading, etc., are different, and these parameters will affect
individual HCP clearance. Nevertheless, this data suggested that
PLBL2 may possess a slightly different affinity in terms of protein-
protein interactions to mAb C and mAb D as compared to mAb A and
B, which could possibly explain its different clearance pattern during
the purification process. Indeed, the similar observations were
reported by Tran et al. that PLBL2 binds more strongly to IgG4 mAbs
due to protein-protein interactions.26
LPL and LIPA also showed a molecule- and process-dependent
clearance during the purification. LPL was detected in the Protein A
column load between 200 to 1200 ppm from the four mAbs. After
Protein A column, its abundance was reduced to anywhere between
3 to 100 ppm in the Protein A column eluate, and was further cleared
down to close to 1 ppm level after Column-2 and Column-3. On the
other hand, LIPA was detected all below 20 ppm in the Protein A col-
umn eluates and was further removed to below LLOQ after the Col-
umn-2 for the four mAbs.

Robustness of PLBL2 Clearance


Fig. 3. Concentrations of (A) PLBL2, (B) LPL and (C) LIPA in process intermediate sam-
To further understand the robustness of lipase HCP clearance by ples from four mAbs: mAb A and mAb B (IgG1), mAb C (IgG1/IgG4 hybrid) and mAb D
the 3-column process, we carried out a lipase clearance “challenging (IgG4). Samples with HCP level below LLOQ are labeled as “*”.

study,” where recombinant PLBL2 was spiked into process intermedi-


ates, including the Column-2 load and Column-3 load. This spike-in (0.8 ppm) for both antibodies. The Column-3 process was further
study was intended to create some worst-case scenarios where the challenged with higher PLBL2 spike-in at around 500 and 300 ppm
lipase HCPs are present at abnormally high concentration, so that the for mAb A and mAb D, respectively. The measured PLBL2 concentra-
downstream process can be evaluated for its robustness to success- tion in the Column-3 product was still below the LLOQ in both cases
fully clear the HCPs. PLBL2 is one of the best-studied lipase HCPs with (Fig. 4b). This translated to more than 2 logs of reduction of PLBL2 by
commercial availability and, therefore, was chosen for the robustness the Column-3 process for the two antibodies studied, which demon-
study. strated that Column-3 is critical and has a strong capacity for PLBL2
As shown in Fig. 3, PLBL2 was present at very low level (»1 ppm) clearance in mAb A and mAb D processes.
after Protein A column purification for mAb A and was further
reduced to <0.2 ppm after the Column-2 purification. To understand Discussion
the robustness of the Column-2 process for PLBL2 clearance, recombi-
nant PLBL2 was spiked into the Column-2 load of mAb A to a final In this study, we demonstrated a workflow starting with shotgun
concentration of 74 ppm. After Column-2 purification, PLBL2 concen- proteomics to identify potentially high-risk lipase HCPs followed by
tration was measured to be around 17 ppm in the product (Fig. 4a). developing an LC-MRMhr-based method for the quantitation. We
The results indicated that in an extreme condition where PLBL2 is evaluated the sensitivity, linearity, reproducibility and accuracy of
present at higher-than-normal level in Protein A column eluate, a the lipase HCP quantitation method. The method can achieve sensi-
second column alone does not have sufficient capacity to clear PLBL2 tivity of 6.25 to 25 pM for the three lipase HCPs, corresponding to
to a level comparable to the 3-column process for mAb A. around 1 ng/mL of HCP in the digested sample. The method is sensi-
Column-3 is crucial for PLBL2 clearance in the downstream pro- tive enough to detect lipase HCP present at 1-2 ppm level in a mAb
cess of a few mAbs such as mAb D. The spike-in study was carried out sample.
on the Column-3 process, where recombinant PLBL2 was spiked into ELISA assay has been the gold standard assay for HCP analysis in
the Column-3 load of mAb A (IgG1) and mAb D (IgG4) at two different the industry owing to its superior specificity, sensitivity, and high
levels. In the lower spike-in level, PLBL2 concentration was measured throughput. However, the ELISA assay can be limited by the availabil-
to be 80 ppm and 66 ppm, respectively, for mAb A and mAb D load; ity of reagents that recognizes specific HCP, hence the development
after the Column-3 purification, PLBL2 was not detected above LLOQ timeline can be much longer than for LC-MS/MS method. In addition,
Y. Chen et al. / Journal of Pharmaceutical Sciences 110 (2021) 3811−3818 3817

Fig. 4. PLBL2 clearance robustness study. PLBL2 concentration in column load (L) and product (P) was determined, where recombinant PLBL2 was spiked into (A) Column-2 load of
mAb A and (B) Column-3 load for mAb A and mAb D at different levels.

even when there are commercial HCP specific ELISA kits available, We also realize that the targeted HCP method may possess some
not all of them are specific for CHO host and often times the cross- limitations. First, the quantitation accuracy can be potentially
species reactivity is unknown. The targeted LC-MS/MS method, on affected by the matrix, especially early in-process samples with
the other hand, offers similar selectivity and sensitivity approaching higher HCP complexity, such as cell culture harvest samples. None-
low-ppm to sub-ppm levels. It was recently demonstrated that quan- theless, it is acknowledged that the accuracy of HCP quantitation is
titation results of process intermediate samples from LC-MRM more important in the drug substance and later process intermedi-
method and a PLBL2-specific ELISA method showed strong correla- ates, as the residual HCPs will more likely affect product quality and
tion across a wide concentration range for different mAb molecules,25 stability. In addition, the method could still provide relative trend
indicating the two assays can provide comparable results for high- information of HCP abundance for earlier intermediates. One mitiga-
risk HCP monitoring. There are several advantages of developing an tion strategy we implemented is to ensure the digestion enzymes are
LC-MS/MS method for targeted HCP quantitation. First, an LC-MS/MS placed in a sufficiently high enzyme-to-sample ratio to ensure a
method has the benefit of versatility that any HCPs that are detect- near-complete digestion, and to select peptides that are less prone to
able by mass spectrometry can be put through a similar development mis-cleavage. An alternative to using peptide standards is to use
process for a targeted quantitation assay. In addition,25 the use of recombinantly purified HCP proteins as standards, which will com-
synthetic peptides as quantitation standards makes absolute quanti- pensate for the digestion efficiency and recovery issues to a large
tation possible, and provides a tool for faster assay development in extent.25 However, this approach still cannot eliminate the matrix
response to newly identified HCPs when recombinant protein stand- effect and also faces issues as many HCPs are not commercially avail-
ards may not be available. Finally, the targeted LC-MS/MS can be mul- able, or the purity can be too low to serve as a standard. A third
tiplexed by incorporating multiple HCP targets into one assay. With approach that can potentially minimize matrix effect is to spike
proper scheduling of the MS/MS events according to the retention heavy-isotope labeled peptides or proteins into samples, and the
time of the target peptides, additional MRM transitions can be quantitation can be achieved through comparing the signals of target
included in a 20-min gradient, so that even more HCPs can be moni- peptides with the heavy-isotope labeled peptides. This method, how-
tored by a single method. ever, will require spiking-in heavy-isotope labeled peptides at a few
Compared with LC-MRM methods using low resolution mass different levels to accommodate for the wide dynamic range of the
spectrometers, this study has demonstrated that a LC-MRMhr HCPs in different samples.
method could achieve similar sensitivity, accuracy, and reproducibil- In this study, we have shown that the LC-MRMhr method can be
ity. We found the enhancement function utilized in the LC-MRMhr used as a convenient tool to quantify the three lipase HCPs in process
method played a critical role to improve the method sensitivity. Com- intermediate samples to support process development, where the
paring the standard curves obtained by using enhancement vs. no- clearance of lipase HCPs can be monitored throughout the upstream
enhancement in the method, the LLOQ was 2- to 10-fold higher if and downstream processes. In addition, we also performed a prelimi-
enhancement was not used, and there was no significant change of nary spike-in study to demonstrate the robustness of PLBL2 clearance
linear range comparing the two setups (Supplementary Table 6). In during various column steps and identified certain column steps and
addition, the high-resolution mass detection provided additional parameters that are critical for the clearance of this lipase. We antici-
level of specificity and selectivity due to the high mass accuracy. As pate that this kind of clearance robustness study will be useful in
shown in Supplementary Figure 2, there are very minimal interfer- understanding HCP removal under different process conditions and
ence with the target peptide in the detection window even at LLOQ could facilitate decision-making in process development to allow the
level. The use of heavy-isotope-labeled peptides as internal standards most robust HCP clearance.
further improves method specificity by facilitating accurate assign-
ment of target peaks, which sometimes can be difficult due to sample Conclusions
low concentration and presence of interfering peaks. The use of IS
also minimizes potential run-to-run variations due to variance in There has been an ever-increasing interest and urgent need to iden-
sample matrices and instrument performance, which helps improv- tify and quantify HCP abundance, especially high-risk HCP species that
ing method robustness. Finally, using a high-resolution mass spec- can have a negative impact on product quality, safety, and stability dur-
trometry allows to perform both the HCP discovery by shot-gun ing the development and manufacturing of biological therapeutics.
proteomics and targeted MRM method development on the same Some problematic HCPs require very sensitive quantitation methods
instrument, which minimizes variability of peptide behavior between and differentiation with other HCPs, such as lipases, as even trace
different instrumentation. amounts of such HCPs can cause significant product quality and
3818 Y. Chen et al. / Journal of Pharmaceutical Sciences 110 (2021) 3811−3818

stability issues. In this study, we demonstrate that an LC-MRMhr-based 9. Kishore RSK, Kiese S, Fischer S, Pappenberger A, Grauschopf U, Mahler H-C. The
lipase HCP quantitation method can be developed with a streamlined degradation of polysorbates 20 and 80 and its potential impact on the stability of
biotherapeutics. Pharm Res. 2011;28(5):1194–1210.
protocol within a short timeline, with relative ease in securing suitable 10. Labrenz SR. Ester hydrolysis of polysorbate 80 in mAb drug product: evidence in
standards. The LC-MRMhr method can be used in both early-stage pro- support of the hypothesized risk after the observation of visible particulate in mAb
cess development to guide HCP removal as well as in late-stage process formulations. J Pharm Sci. 2014;103(8):2268–2277.
11. Li X, Chandra D, Letarte S, et al. Profiling active enzymes for polysorbate degrada-
characterization. We anticipate that the LC-MRMhr quantitation tion in biotherapeutics by activity-based protein profiling. Anal Chem. 2021;93
method could complement the proteomics-based HCP profiling (23):8161–8169.
method by providing more sensitive and accurate quantitation for cer- 12. Zhang S, Xiao H, Li N. Rapid polysorbate 80 degradation by liver carboxylesterase
in a monoclonal antibody formulated drug substance at early stage development. J
tain high-risk, problematic HCP species. The current method can be
Pharm Sci. 2020;109(11):3300–3307.
further augmented to include other HCP species and used either as a 13. MacDonald ML, Hamaker N, Lee KH. Bioinformatic analysis of Chinese hamster
platform or molecule-specific method for HCP monitoring. ovary host cell protein lipases. AIChE J. 2018;64(12):4247–4254.
14. Valente KN, Lenhoff AM, Lee KH. Expression of difficult-to-remove host cell protein
impurities during extended Chinese hamster ovary cell culture and their impact on
Acknowledgements continuous bioprocessing. Biotechnol Bioeng. 2015;112(6):1232–1242.
15. Levy NE, Valente KN, Lee KH, Lenhoff AM. Host cell protein impurities in chro-
The authors are grateful of Christopher Barton for great discussion matographic polishing steps for monoclonal antibody purification. Biotechnol Bio-
eng. 2016;113(6):1260–1272.
and suggestions.
16. Bracewell DG, Francis R, Smales CM. The future of host cell protein (HCP) identifi-
cation during process development and manufacturing linked to a risk-based man-
Supplementary Materials agement for their control. Biotechnol Bioeng. 2015;112(9):1727–1737.
17. Gunawan F, Nishihara J, Liu P, et al. Comparison of platform host cell protein ELISA
to process-specific host cell protein ELISA. Biotechnol Bioeng. 2018;115(2):382–389.
Supplementary material associated with this article can be found 18. Zhu-Shimoni J, Yu C, Nishihara J, et al. Host cell protein testing by ELISAs and the
in the online version at doi:10.1016/j.xphs.2021.08.024. use of orthogonal methods. Biotechnol Bioeng. 2014;111(12):2367–2379.
19. Madsen JA, Farutin V, Carbeau T, et al. Toward the complete characterization of
host cell proteins in biotherapeutics via affinity depletions, LC-MS/MS, and multi-
References variate analysis. mAbs. 2015;7(6):1128–1137.
20. Yang F, Walker DE, Schoenfelder J, et al. A 2D LC-MS/MS strategy for reliable detec-
1. Vanderlaan M, Zhu-Shimoni J, Lin S, Gunawan F, Waerner T, Van Cott KE. Experi- tion of 10-ppm level residual host cell proteins in therapeutic antibodies. Anal
ence with host cell protein impurities in biopharmaceuticals. Biotechnol Prog. Chem. 2018;90(22):13365–13372.
2018;34(4):828–837. 21. Kreimer S, Gao Y, Ray S, et al. Host cell protein profiling by targeted and untargeted
2. Goey CH, Alhuthali S, Kontoravdi C. Host cell protein removal from biopharmaceu- analysis of data independent acquisition mass spectrometry data with parallel
tical preparations: towards the implementation of quality by design. Biotechnol reaction monitoring verification. Anal Chem. 2017;89(10):5294–5302.
Adv. 2018;36(4):1223–1237. 22. Doneanu CE, Anderson M, Williams BJ, Lauber MA, Chakraborty A, Chen W.
3. Chiu J, Valente KN, Levy NE, Min L, Lenhoff AM, Lee KH. Knockout of a difficult-to- Enhanced detection of low-abundance host cell protein impurities in high-purity
remove CHO host cell protein, lipoprotein lipase, for improved polysorbate stability monoclonal antibodies down to 1 ppm using ion mobility mass spectrometry cou-
in monoclonal antibody formulations. Biotechnol Bioeng. 2017;114(5):1006–1015. pled with multidimensional liquid chromatography. Anal Chem. 2015;87
4. Dixit N, Salamat-Miller N, Salinas PA, Taylor KD, Basu SK. Residual host cell protein (20):10283–10291.
promotes polysorbate 20 degradation in a sulfatase drug product leading to free 23. Zhang Q, Goetze AM, Cui H, et al. Comprehensive tracking of host cell proteins dur-
fatty acid particles. J Pharm Sci. 2016;105(5):1657–1666. ing monoclonal antibody purifications using mass spectrometry. mAbs. 2014;6
5. Hall T, Sandefur SL, Frye CC, Tuley TL, Huang L. Polysorbates 20 and 80 degradation (3):659–670.
by group XV Lysosomal phospholipase A2 Isomer X1 in monoclonal antibody for- 24. Jahn M, Zerr A, Fedorowicz FM, Brigger F, Koulov A, Mahler HC. Measuring lipolytic
mulations. J Pharm Sci. 2016;105(5):1633–1642. activity to support process improvements to manage lipase-mediated polysorbate
6. Jones MT, Mahler HC, Yadav S, et al. Considerations for the use of polysorbates in degradation. Pharm Res. 2020;37(6):118.
biopharmaceuticals. Pharma Res. 2018;35(8):148. 25. Gao X, Rawal B, Wang Y, et al. Targeted host cell protein quantification by LC-MRM
7. Khan TA, Mahler HC, Kishore RS. Key interactions of surfactants in therapeutic pro- enables biologics processing and product characterization. Anal Chem. 2020;92
tein formulations: a review. Eur J Pharma Biopharma. 2015;97(Pt A):60–67. (1):1007–1015.
8. Dwivedi M, Blech M, Presser I, Garidel P. Polysorbate degradation in biotherapeutic 26. Tran B, Grosskopf V, Wang X, et al. Investigating interactions between phospholi-
formulations: identification and discussion of current root causes. Int J Pharm. pase B-Like 2 and antibodies during Protein A chromatography. J Chromatogr A.
2018;552(1-2):422–436. 2016;1438:31–38.

You might also like