Host Cell Protein Analysis Using Agilent AssayMAP Bravo and 6545XT AdvanceBio LC/Q-TOF

Similar documents
Quantification of Host Cell Protein Impurities Using the Agilent 1290 Infinity II LC Coupled with the 6495B Triple Quadrupole LC/MS System

Ensure your Success with Agilent s Biopharma Workflows

Enhanced Analysis of Host Cell Proteins Using the Agilent 6545XT AdvanceBio LC/Q-TOF

Clone Selection Using the Agilent 1290 Infinity Online 2D-LC/MS Solution

A Unique LC-MS Assay for Host Cell Proteins(HCPs) ) in Biologics

Precise Characterization of Intact Monoclonal Antibodies by the Agilent 6545XT AdvanceBio LC/Q-TOF

Profiling Glycosylation of Monoclonal Antibodies at Three Levels Using the Agilent 6545XT AdvanceBio LC/Q TOF

Fast and Efficient Peptide Mapping of a Monoclonal Antibody (mab): UHPLC Performance with Superficially Porous Particles

Reproducible LC/MS Peptide Separation Using Agilent AdvanceBio Peptide Plus Columns

PLRP-S Polymeric Reversed-Phase Column for LC/MS Separation of mabs and ADC

Put the PRO in Protein Characterization

Highly Confident Peptide Mapping of Protein Digests Using Agilent LC/Q TOFs

Application of Agilent AdvanceBio Desalting-RP Cartridges for LC/MS Analysis of mabs A One- and Two-dimensional LC/MS Study

Application Note. Authors. Abstract. Automated Protein Sample Preparation for LC/MS

A Comprehensive Approach for Monoclonal Antibody N-linked Glycan Analysis from Sample Preparation to Data Analysis

A Highly Accurate Mass Profiling Approach to Protein Biomarker Discovery Using HPLC-Chip/ MS-Enabled ESI-TOF MS

Analysis of Monoclonal Antibody (mab) Using Agilent 1290 Infinity LC System Coupled to Agilent 6530 Accurate-Mass Quadrupole Time-of-Flight (Q-TOF)

Separation of Native Monoclonal Antibodies and Identification of Charge Variants:

Critical Quality Attribute Assessment by Peptide Mapping Using LC/MS with Superficially Porous Columns

Agilent AssayMAP Bravo Platform AUTOMATED PROTEIN AND PEPTIDE SAMPLE PREPARATION FOR MASS SPEC ANALYSIS

Application Note. Authors. Abstract. Ravindra Gudihal Agilent Technologies India Pvt. Ltd. Bangalore, India

Application Note # ET-20 BioPharma Compass: A fully Automated Solution for Characterization and QC of Intact and Digested Proteins

CQA Assessment by Peptide Mapping Using LC/MS with an AdvanceBio Peptide Plus Column

Application Note. Author. Abstract. Biotherapeutics and Biologics. Sonja Schneider Agilent Technologies, Inc. Waldbronn, Germany

Analysis of Monoclonal Antibody N-glycans by Fluorescence Detection and Robust Mass Selective Detection Using the Agilent LC/MSD XT

Application Note. Abstract. Author. Biopharmaceutical

Disulfide Linkage Analysis of IgG1 using an Agilent 1260 Infinity Bio inert LC System with an Agilent ZORBAX RRHD Diphenyl sub 2 µm Column

An Integrated Workflow for Intact and Subunits of Monoclonal Antibody Accurate Mass Measurements

ProteinPilot Report for ProteinPilot Software

Enabling routine characterization of proteins. Agilent MassHunter BioConfirm software

N-Glycan Profiling Analysis of a Monoclonal Antibody Using UHPLC/FLD/Q-TOF

Robustness of the Agilent Ultivo Triple Quadrupole LC/MS for Routine Analysis in Food Safety

Peptide Mapping: A Quality by Design (QbD) Approach

Maximizing Chromatographic Resolution of Peptide Maps using UPLC with Tandem Columns

LC/MS Based Quantitation of Intact Proteins for Bioanalytical Applications

Accelerate mab Characterization Using Automated Sample Prep

Reducing Cycle Time for Charge Variant Analysis of Monoclonal Antibodies

Benefits of 2D-LC/MS/MS in Pharmaceutical Bioanalytics

Cell Clone Selection Using the Agilent Bio-Monolith Protein A Column and LC/MS

Jet Stream Proteomics for Sensitive and Robust Standard Flow LC/MS

Characterize Fab and Fc Fragments by Cation-Exchange Chromatography

Analysis of Nucleosides Using an Agilent Infinity II High Speed UHPLC with the 6130 Single Quadrupole Mass Selective Detector

Peptide Mapping of Glycoprotein Erythropoietin by HILIC LC/MS and RP-LC/MS

Charge Heterogeneity Analysis of Rituximab Innovator and Biosimilar mabs

Bravo with AssayMAP. Automated Sample Preparation

Characterize mab Charge Variants by Cation-Exchange Chromatography

A highly sensitive and robust 150 µm column to enable high-throughput proteomics

High-resolution Analysis of Charge Heterogeneity in Monoclonal Antibodies Using ph-gradient Cation Exchange Chromatography

Agilent Technologies. Carl Myerholtz, Ph.D. Senior Director of QTOF Technology. FAPESP/Agilent Workshop August 30,

Chromatographic Workflows for Biopharmaceutical Characterization

Agilent 1290 Infinity II 2D-LC Solution Biopharmaceutical Polymer Analysis. WCBP Jan 2017 Washington, DC

Separate and Quantify Rituximab Aggregates and Fragments with High-Resolution SEC

Seamless Method Transfer from an Agilent 1260 Infinity Bio-inert LC to an Agilent 1260 Infinity II Bio-inert LC

A Comprehensive Workflow to Optimize and Execute Protein Aggregate Studies

Jet Steam Proteomics with Automated Sample Prep for Superior Peptide Quantitation

Isotopic Resolution of Chromatographically Separated IdeS Subunits Using the X500B QTOF System

Application Note. Authors. Abstract. Clinical Research

Application Note. Authors. Abstract. Automated Peptide Sample Preparation for LC/MS

Universal Solution for Monoclonal Antibody Quantification in Biological Fluids Using Trap-Elute MicroLC-MS Method

Vendor Neutral Host Cell Protein Analysis

Application Note. Biopharma. Authors. Abstract. James Martosella, Phu Duong Agilent Technologies, Inc Centreville Rd Wilmington, DE 19808

Application Note. Author. Introduction. Ravindra Gudihal Agilent Technologies India Pvt Ltd

Proteins. Patrick Boyce Biopharmaceutical Marketing Manager Waters Corporation 1

AdvanceBio HIC: a Hydrophobic HPLC Column for Monoclonal Antibody (mab) Variant Analysis

Sample Preparation Automation and QQQ Workflows for Peptide Quantitation in DMPK

Rapidly Characterize Antibody- Drug Conjugates and Derive Drug-to-Antibody Ratios Using LC/MS

Cell Culture Optimization Using an Agilent Bio-Monolith Protein A Column and LC/MS

Fraction Analysis of Cysteine Linked Antibody-Drug Conjugates Using Hydrophobic Interaction. chromatography. Agilent 1260 Infinity II Bio-Inert System

Analysis of Intact and C-terminal Digested IgG1 on an Agilent Bio MAb 5 µm Column

Cell Signaling Technology

Developing Quantitative UPLC Assays with UV

Enabling Systems Biology Driven Proteome Wide Quantitation of Mycobacterium Tuberculosis

Biotherapeutic Non-Reduced Peptide Mapping

Agilent AssayMAP Platform BIOLOGICS SAMPLE PREPARATION INTELLIGENTLY AUTOMATED

Ultra-Sensitive Quantification of Trastuzumab Emtansine in Mouse Plasma using Trap-Elute MicroLC MS Method

Analysis of Monoclonal Antibodies and Their Fragments by Size Exclusion Chromatography Coupled with an Orbitrap Mass Spectrometer

Advanced QA/QC characterization MS in QC : Multi Attribute Method

Quantification of genotoxic "Impurity D" in Atenolol by LC/ESI/MS/MS with Agilent 1200 Series RRLC and 6410B Triple Quadrupole LC/MS

Expand Your Research with Metabolomics and Proteomics. Christine Miller Omics Market Manager ASMS 2017

Automation for LC/MS Sample Preparation: High Throughput In-Solution Digestion and Peptide Cleanup Enabled by the Agilent AssayMAP Bravo Platform

Simultaneous Quantitation of a Monoclonal Antibody and Two Proteins in Human Plasma by High Resolution and Accurate Mass Measurements

Automated Scouting of Stationary and Mobile Phases Using the Agilent 1290 Infinity II Method Development Solution

Combination of Isobaric Tagging Reagents and Cysteinyl Peptide Enrichment for In-Depth Quantification

The Agilent Metabolomics Dynamic MRM Database and Method

Generating Automated and Efficient LC/MS Peptide Mapping Results with the Biopharmaceutical Platform Solution with UNIFI

Antibody Analysis by ESI-TOF LC/MS

New Approaches to Quantitative Proteomics Analysis

Application Note. Author. Abstract. Biopharmaceuticals. Verified for Agilent 1260 Infinity II LC Bio-inert System. Sonja Schneider

Appendix. Table of contents

: presented at ASMS 2015

Biotherapeutic Peptide Mapping Information Dependent Acquisition (IDA) Method

Introduction. Benefits of the SWATH Acquisition Workflow for Metabolomics Applications

Characterization of mab aggregation using a Cary 60 UV-Vis Spectrophotometer and the Agilent 1260 Infinity LC system

Mobile Phase Optimization in SEC Method Development

AdvanceBio Peptide Mapping

A streamlined drug-to-antibody ratio determination workflow for intact and deglycosylated antibody-drug conjugates

Streamlined and Complete LC-MS Workflow for MAM

Mass Spectrometry Analysis of Liquid Chromatography Fractions using Ettan LC MS System

Monitoring of Mammalian Cell Culture Media with HILIC LC/MS

Application Note LCMS-87 Automated Acquisition and Analysis of Data for Monitoring Protein Conjugation by LC-MS Using BioPharma Compass

Transcription:

Application Note Biotherapeutics and Biosimilars Host Cell Protein Analysis Using Agilent AssayMAP Bravo and 6545XT AdvanceBio LC/Q-TOF Authors Linfeng Wu, Shuai Wu and Te-Wei Chu Agilent Technologies, Inc., Santa Clara, CA, USA Introduction Use of biopharmaceuticals such as monoclonal antibodies (mabs) has been growing rapidly. Since biopharmaceuticals are generated from biological sources, some of the low-level host cell proteins (HCPs) could remain in the final products even after multiple purification steps. Due to their potential to affect product safety and efficacy, HCP level must be monitored and controlled in drug products according to regulatory requirements 1. Traditionally, enzyme-linked immunosorbent assay (ELISA) is the standard method of choice for quantifying HCPs in protein therapeutics. However, ELISA lacks the specificity and coverage to identify and quantify individual HCPs. Therefore, LC/MS technologies have become a choice for HCP analysis. The main challenge during LC/MS analysis of HCPs exists in the coelution of low-abundance HCP peptides with the highly abundant peptides from the drug product. This requires better separation of the peptides and broad dynamic range of the LC/MS system.

This study demonstrates an HCP analysis workflow including the Agilent AssayMAP Bravo platform for automated sample preparation, the Agilent 6545XT AdvanceBio LC/Q TOF for LC-MS/MS analysis, and the vendor neutral software from Protein Metrics for data analysis (Figure 1). The AssayMAP Bravo platform was used for automated sample preparation, which included protein digestion, desalting, and on-cartridge high-ph reversed-phase (HPRP) fractionation. Both digested samples with or without HPRP fractionation were subjected to LC-MS/MS analysis to examine the platform performance. A new acquisition method, Iterative MS/MS, has been shown to improve protein identification. This standard-flow LC/Q-TOF system demonstrates broad dynamic range, excellent reproducibility, and semiquantitative capability. Our results also showed that all HCPs above ppm were identified with high confidence by coupling on-cartridge sample fractionation with Iterative MS/MS. Sample preparation Online separation Detection AssayMAP Bravo Enzymatic digestion and HPRP fractionation Experimental Materials Human IgG1 mab (an R&D product from a partner) was produced from Chinese Hamster Ovary (CHO) cells and purified with protein A. Proteomics Dynamic Range Standard Set (UPS) was purchased from Sigma-Aldrich. Instrumentation Agilent AssayMAP Bravo system Agilent 19 Infinity II LC system including: Agilent 19 Infinity II High Speed Pump G71A Agilent 19 Infinity II Multisampler G7167B 19 Infinity II LC System AdvanceBio Peptide Plus column Figure 1. Overview of Agilent HCP Workflow. Agilent 19 Infinity II Thermostatted Column Compartment G7116B 6545XT AdvanceBio Q-TOF Iterative MS/MS Sample preparation Data processing and report Protein Metrics UPS was spiked into the mab at a 1:1, ratio. A second sample, comprising the mab without UPS spiking, was prepared and analyzed in parallel as a negative control to ensure that the identification of UPS proteins was not due to the presence of sequence homologs in the control sample. Both samples were reduced, alkylated, trypsin-digested, and desalted using the AssayMAP Bravo system. The samples were then aliquoted; one aliquot was subjected directly to LC-MS/MS analysis using the Agilent 6545XT AdvanceBio LC/Q-TOF system, and the other aliquot was fractionated using HPRP method on the AssayMAP Bravo system, followed by LC-MS/MS analysis. Agilent 6545XT AdvanceBio LC/Q TOF Agilent Dual Jet Stream ESI source

HPRP fractionation The digested sample was fractionated into six fractions on an Agilent Reversed Phase (RP-S) cartridge using the preconfigured Protein Sample Prep WorkBench Fractionation application on the AssayMAP Bravo (Figure ). RP-S cartridges were first primed with 7 % acetonitrile (ACN),.1 % trifluoroacetic acid (TFA) and equilibrated with.1 % TFA. Then, 15 μg of digested sample was loaded onto each cartridge, and eluted into six fractions using 1 mm ammonium formate buffer (ph 1) with a stepwise increase of ACN (1, 15,, 5, 3, and 9 %). LC/MS analysis LC separation was performed on an Agilent AdvanceBio Peptide Plus column (.1 15 mm,.7 µm, p/n 67595 9) with.4 ml/min flow rate using a 6 minute LC method (Table 1). Each sample was analyzed on the 6545XT AdvanceBio LC/Q-TOF system using either a conventional Auto MS/MS method or an Iterative MS/MS method as indicated (Table ). Table 1. Liquid chromatography parameters. Table. MS parameters. LC Parameters Analytical column AdvanceBio Peptide Plus,.1 15 mm,.7 μm (p/n 67595-9) Mobile phase A Mobile phase B Flow rate Gradient Stop time Column temperature 6 C H O,.1 % formic acid 9 % ACN,.1 % formic acid.4 ml/min 1. minutes & 3 %B 5. minutes & 1 %B 53. minutes & 9 %B 55. minutes & 9 %B 55.1 minutes & 3 %B 6 minutes Drying gas temperature 9 C Drying gas flow Nebulizer MS Parameters 13 L/min 35 psi Sheath gas temperature 75 C Sheath gas flow Isolation width Dynamic exclusion within run MS Mass range MS Acquisition rate MS/MS Mass range MS/MS Acquisition rate Acquisition mode Iterative MS/MS 1 L/min Narrow (~1.3 m/z) 1 spectrum release after. minutes 5 1,7 m/z 1 spectra/second 5 1,7 m/z 3 spectra/second Iterative MS/MS or Auto MS/MS as indicated Mass error tolerance: 15 ppm RT exclusion tolerance:. minutes to +.4 minutes Figure. Fractionation application user interface with default settings. 3

Data processing The raw data files were processed using software from Protein Metrics Inc. One raw data file was searched using Preview to generate Byonic parameters. All raw data files were searched against a Uniprot CHO K1 protein database that was concatenated with mab and UPS protein sequences using Byonic. The search parameters included semispecific trypsin digestion, up to two missed cleavages, ppm precursor mass tolerance, 3 ppm fragment mass tolerance, fixed cysteine (C) alkylation, variable methionine (M) oxidation, asparagine (N) and glutamine (Q) deamidation, and other variable modifications as suggested by the Preview analysis. The Byonic result files were imported into Byologic for further detailed analysis on a smaller set of protein sequences, and for result reporting. A minimum score of 15 for MS search was used to filter peptides. LC/MS Injection 1 LC/MS Injection 1 6.4.. 1.8 1.6 1.4 1. 1..8.6.4. 3 4 5 6 7 8 9 3 Acquisition time (min) m/z Results and discussion Comparison of Iterative MS/MS and Auto MS/MS The Agilent 6545XT AdvanceBio Q-TOF system provides a new data acquisition method, Iterative MS/MS, which improves identification of low-abundance precursors (Figure 3). Using this method, the protein digest sample was subjected to multiple LC-MS/MS analyses. The first analysis was performed as a conventional Auto MS/MS run using data dependent acquisition. In the following Iterative LC-MS/MS analyses, precursors that were previously selected for MS/MS fragmentation were automatically excluded on a rolling basis with customizable mass error tolerance and retention time exclusion tolerance. As a result, more precursors were automatically interrogated by LC-MS/MS. Additional injections Precursors selected for MS/MS Rolling excluded precursors Figure 3. A diagram of an automated Iterative MS/MS acquisition method. We first compared Iterative MS/MS to Auto MS/MS using the LC/Q-TOF system without any offline fractionation that is, where the purified mab with spiked-in UPS was analyzed in both methods. UPS is the proteomics dynamic range standard produced by Sigma-Aldrich, which is a complex protein mixture containing 48 human proteins at six concentrations ranging from 5 amoles to 5 pmoles. Spiking UPS into the m/z mab sample at a 1:1, ratio (w/w) before digestion resulted in protein levels ranging from.4 to 313 ppm, among which 19 proteins were present above one ppm. This spiking sample mimics the wide dynamic range of the HCPs present in the therapeutic proteins. It also allows us to evaluate the sensitivity and dynamic range of different methods being investigated in this study. 4

Table 3 shows the total numbers of unique peptide sequences from mab and UPS proteins that were identified by each acquisition method. Peptides with the same amino acid sequence but different modifications were counted as one unique peptide sequence. The sample loading amount for each injection was 3 μg, and three injections were carried out for each acquisition method. Overall, Iterative MS/MS identified more unique peptide sequences per protein across a wide dynamic range. All the spiked-in proteins above 8 ppm level were identified with high confidence. All additional results presented here were performed by Iterative MS/MS. Chromatographic reproducibility and dynamic range We also assessed the chromatographic reproducibility and dynamic range using the above dataset acquired by three injections of Iterative MS/MS analyses. Figure 4 shows the overlaid chromatograms of the base peak chromatogram (BPC) and Extracted Ion Chromatogram of four coeluting precursor ions from three Iterative LC-MS/MS runs. Due to the high sample Table 3. Number of unique peptide sequences identified by Iterative MS/MS or Auto MS/MS acquisition methods. Protein accession Molecular weight (kda) Protein spiking level (ppm) Three injections per method Iterative MS/MS Auto MS/MS mab_hc 49.7 NA 419 38 mab_lc 4. NA 1 186 ALBU_HUMAN_spike 66.4 313. 46 43 CAH_HUMAN_spike 9.1 137.3 19 15 CAH1_HUMAN_spike 8.7 135.6 9 9 LEP HUMAN_spike 16. 76. 4 1 HBB_HUMAN_spike 15.9 74.8 1 1 HBA_HUMAN_spike 15.1 71.3 7 6 UBIQ_HUMAN_spike 1.6 5. 6 6 CO5_HUMAN_spike 8.6 4.4 4 4 CATA_HUMAN_spike 59.6 8.1 SUMO1_HUMAN_spike 38.8 18.3 3 1 NQO1_HUMAN_spike 3.7 14.5 PRDX1_HUMAN_spike. 1.4 3 PPIA_HUMAN_spike. 9.5 4 4 MYG_HUMAN_spike 17.1 8. 1 loading amount on the column, many of the signals from mab peptides saturated the MS detector, which showed a plateau in the BPC. Details about those four coeluting peptide precursors are summarized in Table 4. Excellent chromatographic reproducibility was observed for the coeluting peptides, and quantitative reproducibility was excellent across an extremely broad dynamic range (peak intensities from 6.76 1 3 to 1.38 1 8 ). For the lowest-abundance myoglobin peptide, which was spiked in at 8 ppm, we observed an RSD value of 1.3 %. These data demonstrated the excellent in-spectrum dynamic range and reproducibility of the 6545XT AdvanceBio LC/Q-TOF system. 1 6.4.. 1.8 BPC EPQVYTLPPSR Zoom SAVTALWGK TIAQDYGVLK 1.6 1.4 1. 1..8 ALELFR.6.4. 4 6 8 1 1 14 16 18 4 6 8 3 3 34 36 38 4 4 44 46 48 5 5 54 56 58 Acquisition time (min) Figure 4. Overlaid chromatograms of three LC-MS/MS runs for BPC and extracted ion chromatograms of four selected peptide precursors. 5

Semiquantification We also explored semiquantitative analytical capabilities of this workflow solution using the above data files acquired by three injections of Iterative MS/MS analyses. The total extracted ion chromatogram (XIC) for each identified UPS protein using all the measured peptides was normalized to the highest protein in the sample, which is mab heavy chain in this study, and the values were exported directly from Byologic software. Figure 5 shows the normalized XIC per protein (Y-axis on the left) in the mab control sample (blue) and the UPS spiking sample (orange) plotted with the actual UPS protein level in the spiking sample (green, Y-axis on the right). As expected, there are no XIC signals from UPS proteins in the mab control sample, demonstrating the high specificity of this workflow solution. In the UPS spiking sample, the normalized XIC was correlated with the actual protein spiked-in level in ppm. These results indicate that the normalized XIC values reported from Byologic software offer a semiquantitative estimate of HCP abundance. Table 4. Data of the four selected peptide precursors. Peptide Precursor ion (m/z) Mass error (ppm) Intensity Intensity %RSD Protein spiking level (ppm) Protein name ALELFR 374.78-1.1 6.76E+3 1.3 % 8 Myoglobin TIAQDYGVLK 554.349-1.8 1.51E+5 6. % 1.4 Peroxiredoxin 1 SAVTALWGK 466.7659 4.8 1.36E+5 6. % 74.8 Hemoglobin subunit beta EPQVYTLPPSR 643.844 1. 1.38E+8 1. % NA mab Normalized XIC per protein.1.1.8.6.4. Normalize XIC in mab control Normalized XIC in UPS spiking sample Actual level in UPS spiking sample (ppm) 4 35 3 5 15 1 5 Actual UPS protein spiked-in level (ppm) Figure 5. Comparison between normalized XIC per protein and actual UPS spike-in level. The XIC per protein was normalized to mab heavy chain, and their values in mab control sample (blue) and UPS spiking sample (orange) were plotted with the actual protein levels in the UPS spiking sample (green). 6

Improving identification by HPRP fractionation using AssayMAP Bravo Several core protein sample preparation tasks have been automated using the Agilent AssayMAP Bravo sample preparation platform (for example, protein digestion, peptide cleanup, immuno-affinity purification, and so forth) with micro-chromatography cartridges and task centric automation protocols 4. With the same automation platform, we demonstrated improvements on identification sensitivity by HPRP fractionation using RP-S cartridges. One hundred fifty micrograms of digested sample were loaded on each RP-S cartridge and eluted into six fractions using an easy-to-use high-ph fractionation protocol. Each fraction was analyzed by two Iterative MS/MS runs. Figure 6 shows the TIC signal from the LC-MS/MS analysis of unfractionated sample compared to HPRP eluates, which demonstrated the chromatographic separation on the RP-S cartridge. 1 7 1 6 5 1 7 1 7 1 7 1 7 1 7 1 7 1 1 1 3 5 7 9 11 13 15 17 19 1 3 5 7 9 31 33 35 37 39 41 43 45 47 49 51 Acquisition time (min) Unfractionated HPRP Flowthrough HPRP Fraction 1 HPRP Fraction HPRP Fraction 3 HPRP Fraction 4 HPRP Fraction 5 HPRP Fraction 6 Figure 6. TIC signal from unfractionated and HPRP fractionated sample. 7

The MS/MS spectra of peptides from low-abundance spiked-in proteins were manually inspected to ensure high spectral quality and no homolog contamination from endogenous CHO HCPs. All the spiked-in proteins above ppm were identified with high confidence, demonstrating an improvement in the identification sensitivity using HPRP fractionation (Table 5, compared to Table 3). Figure 7 shows an example MS/MS spectrum for peptide IEEIFK from the human KCRM protein, which was spiked in at ppm. Table 5. Identification of unique peptide sequences from the UPS spiking proteins using AssayMAP HPRP fractionation coupled with two Iterative MS/MS runs per fraction. Protein accession Molecular weight (kda) Protein spiking level (ppm) No. unique peptide sequences ALBU_HUMAN_spike 66.4 313. 79 CAH_HUMAN_spike 9.1 137.3 3 CAH1_HUMAN_spike 8.7 135.6 15 LEP_HUMAN_spike 16. 76. 6 HBB_HUMAN_spike 15.9 74.8 HBA_HUMAN_spike 15.1 71.3 14 UBIQ_HUMAN_spike 1.6 5. 9 CO5_HUMAN_spike 8.6 4.4 6 CATA_HUMAN_spike 59.6 8.1 14 SUMO1_HUMAN_spike 38.8 18.3 11 NQO1_HUMAN_spike 3.7 14.5 8 PRDX1_HUMAN_spike. 1.4 9 PPIA_HUMAN_spike. 9.5 11 MYG_HUMAN_spike 17.1 8. CYB5_HUMAN_spike 16. 7.6 EGR_HUMAN_spike 6.4 3. 1 SYHC_HUMAN_spike 58..7 5 KCRM_HUMAN_spike 43.1. 3,5, Intensity 1,5 1, 5 1 3 4 5 6 7 Mass-to-charge (m/z) Figure 7. MS/MS spectrum of peptide IEEIFK originated from the KCRM protein spiked at ppm. 8

The impact of HPRP fractionation on identifying endogenous CHO HCPs was also examined. Figure 8 shows the number of identified CHO proteins between unfractionated and HPRP fractionated samples at a 1 % protein false discovery rate. The unfractionated sample was analyzed by three Iterative MS/MS runs, and the HPRP fractionated samples were analyzed by two Iterative MS/MS runs per fraction. The results show a more than three-fold increase using HPRP fractionation coupled with Iterative MS/MS (138 versus 38). All the above results demonstrate the advantage of on-cartridge HPRP fractionation in improving identification of HCPs. Conclusions A host cell protein analysis workflow including the AssayMAP Bravo platform for automated sample preparation, the 6545XT AdvanceBio LC/Q-TOF for LC-MS/MS analysis, and the vendor neutral software from Protein Metrics for data analysis, has been demonstrated. The AssayMAP Bravo platform using microchromatography cartridges and task-centric automation protocols has brought unprecedented reproducibility, scalability, flexibility, and ease-of-use to sample preparation automation. Iterative MS/MS improves protein identification coverage. Using LC-MS/MS coupled with Iterative MS/MS acquisition, all the spiked-in standard proteins above 8 ppm were identified with high confidence. Number of CHO proteins 16 14 1 1 8 6 4 38 Nonfractionation with three Iterative runs Figure 8. Comparison of number of identified endogenous CHO HCPs between unfractionated and HPRP fractionated samples coupled with Iterative MS/MS acquisition. Broad dynamic range and excellent reproducibility were demonstrated on this standard-flow LC/Q-TOF system. By adding HPRP fractionation using AssayMAP Bravo, all the spiked-in proteins above ppm were identified with high confidence. Data files acquired by data dependent acquisition allow simple and fast protein identification and semiquantification. 138 HPRP Fractionation with two Iterative runs 9

References 1. ICH Q6B Specifications: Test Procedures and Acceptance Criteria for Biotechnological/Biological Products.. Automation for LC/MS Sample Preparation: High Throughput In-Solution Digestion and Peptide Cleanup Enabled by the Agilent AssayMAP Bravo Platform. Agilent Technologies, publication number 5991-957EN. 3. Automation of Sample Preparation for Accurate and Scalable Quantification and Characterization of Biotherapeutic Proteins Using the Agilent AssayMAP Bravo Platform. Agilent Technologies, publication number 5991-487EN. 4. Agilent AssayMAP Bravo Technology Enables Reproducible Automated Phosphopeptide Enrichment from Complex Mixtures Using High Capacity Fe(III)-NTA Cartridges. Agilent Technologies, publication number 5991-673EN. www.agilent.com/chem For Research Use Only. Not for use in diagnostic procedures. This information is subject to change without notice. Agilent Technologies, Inc. 18 Printed in the USA, July, 18 5991-93EN