40
BIOPHARMACEUTICAL ANALYSIS SUPPLEMENT TO ADVANCES IN Volume 34, Number s11 November 2016 www.chromatographyonline.com A supplement to LCGC North America Advances in Biopharmaceutical Analysis November 2016

ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

BIOPHARMACEUTICAL ANALYSIS

SUPPLEMENT TO

ADVANCES IN

Volume 34, Number s11 November 2016

www.chromatographyonline.com

A s

up

ple

me

nt to

LC

GC

No

rth

Am

eric

a

Ad

va

nce

s in

Bio

ph

arm

ace

utic

al A

na

lysis

N

ove

mb

er 2

01

6

Page 2: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

; ; ;

That’s the problem with relying on column calibration to characterize macromolecules.

You don’t really know if you’re right—you can only assume. Which is why every major pharma-

ceutical and biotechnology company is switching from relative methods to Wyatt Technol-

ogy’s absolute measurements. Our DAWN® multi-angle light scattering (MALS) instru-

ments allow you to determine absolute molecular weights and sizes without relying on

so-called standards, or measurements made in someone else’s lab. Wyatt instruments mea-

sure all of the quantities required for determining absolute molar masses directly. So visit

www.wyatt.com and request our free 32-page Ultimate Guide to Light Scattering. You’ll learn how

to end your dependence on reference standards, which will make you and your products a

whole lot more convincing.

“So you’re sure this protein has a molecular weight of 150,000?” “According to our assumptions, that’s correct.”

DAWN HELEOS II. The most advanced multi-angle light scattering instrument for macromolecular characterization.

Optilab T-rEX. The refractometer with the greatest sensitivity and range.

ViscoStar II. The viscometer with unparalleled signal-to-noise, stable baselines and a 21st-century interface.

Eclipse. The ultimate system for the separation of macromolecules and nanoparticles in solution.

DynaPro Plate Reader II. Automated dynamic light scattering for 96 or 384 or 1536 well plate samples—now with an onboard camera!

© 2009 Leo Cullum from cartoonbank.com. All Rights Reserved. DAWN, Optilab, DynaPro and the Wyatt Technology logo are registered trademarks, and ViscoStar and Eclipse are trademarks of Wyatt Technology Corporation.

Page 3: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

To learn more about how polymer columns can perform

for you, visit www.ham-info.com/0805-1

or call toll free 1-888-525-2123.© 2014 Hamilton Company. All rights reserved.

Images Copyright Rangizzz and Carolina K. Smith, M.D., 2014

Used under license from Shutterstock.com

Polymer HPLC columns have a lot of benefi ts. They don’t require

any functionalization for reversed-phase separations, and rigid

polymeric supports intrinsically resist chemical and pH degradation,

a fundamental problem with silica columns. Plus, polymer’s inertness

to most chemical environments makes it a robust and

economical solution.

Hamilton offers a line of pH stable polymer HPLC columns for

reversed phase, anion exchange, cation exchange and ion exclusion

separations perfect for pharmaceuticals, small molecules, proteins,

peptides, DNA, organic and inorganic ions and more.

pH range of 1–13

Widest chemical compatibility

Temperatures higher than 60 °C

Maximum sample recovery

Longest average life span

Page 4: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

www.chromatographyonline.com4 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016

ArticlesIntroduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5Pat Sandra and Koen SandraAn introduction from the guest editors of this special supplement

Modern Column Technologies for the Analytical Characterization of Biopharmaceuticals in Various Liquid Chromatographic Modes. . . . . . . . . . . . . . . . . 6Szabolcs Fekete, Jean-Luc Veuthey, and Davy GuillarmeThe recent trends in column technology for reversed-phase LC, SEC, ion-exchange

chromatography, and HIC for analysis of biopharmaceuticals are critically discussed.

Monoclonal Antibodies and Biosimilars—A Selection of Analytical Tools for Characterization and Comparability Assessment . . . . . . . . . . . . . . . . . . . . . . . . . 14Koen Sandra, Isabel Vandenheede, Emmie Dumont, and Pat SandraThis article presents a selection of state-of-the-art analytical tools for mAb

characterization and comparability assessment.

Higher Order Mass Spectrometry Techniques Applied to Biopharmaceuticals. . . . . . . . . . . . . . 22Christian G. HuberAn outline of the basic principles of MS techniques used to investigate higher order

structural features of biopharmaceuticals, as well as some insights into applications

relevant to the pharmaceutical industry.

Advances in Liquid Chromatography–Tandem Mass Spectrometry (LC–MS/MS)-Based Quantitation of Biopharmaceuticals in Biological Samples . . . . . . . . . . . . . . 28Nico C. van de MerbelThe technical requirements for a successful LC–MS/MS method for the

quantitation of biopharmaceuticals are presented and the advantages and

disadvantages compared to ligand-binding assays are evaluated.

Analyzing Host Cell Proteins Using Off-Line Two-Dimensional Liquid Chromatography–Mass Spectrometry . . . . . . . . . . . . 35Koen Sandra, Alexia Ortiz, and Pat SandraThe use of off-line 2D-LC–MS for the characterization of HCPs and their monitoring

during downstream processing

November 2016

Cover images courtesy of vitstudio/shutterstock.com

PUBLISHING & SALES

485F US Highway One South, Suite 210, Iselin, NJ 08830(732) 596-0276

Michael J. TessaloneVice President/Group Publisher,[email protected]

Edward Fantuzzi Publisher,

[email protected]

Stephanie ShafferEast Coast Sales Manager,

[email protected]

Lizzy Thomas Sales Manager, [email protected]

EDITORIAL

Laura Bush Editorial Director,

[email protected]

Alasdair Matheson Editor-in-Chief, LCGC [email protected]

Kate Mosford Managing Editor, LCGC Europe

[email protected]

Megan L’Heureux Managing Editor,

Meg.L’[email protected]

Stephen A. Brown Group Technical Editor,

[email protected]

Dan Ward Art Director,

[email protected]

Rajesh Thangappan Graphic Designer

Anne Lavigne Marketing Manager,

[email protected]

Wright’s Media Reprints,

[email protected]

Jesse Singer Production Manager,

[email protected]

Wendy Bong Audience Development Manager,

[email protected]

© 2016 UBM. All rights reserved. No part of this publication may be

reproduced or transmitted in any form or by any means, electronic

or mechanical including by photocopy, recording, or information

storage and retrieval without permission in writing from the

publisher. Authorization to photocopy items for internal/educational

or personal use, or the internal/educational or personal use of specific

clients is granted by UBM for libraries and other users registered

with the Copyright Clearance Center, 222 Rosewood Dr. Danvers, MA

01923, 978-750-8400 fax 978- 646-8700 or visit http://www.copyright.

com online. For uses beyond those listed above, please direct your

written request to Permission Dept. fax 440-756-5255 oremail:

[email protected].

A supplement to LCGC North America

BIOPHARMACEUTICAL

ANALYSIS

BIOPHARMACEUTICAL

ANALYSIS

ADVANCES INADVANCES IN ®

Page 5: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 5www.chromatographyonline.com

Advances in Biopharmaceutical Analysis

An introduction from the guest editors, Pat Sandra and Koen Sandra of the Research Institute for Chromatography,

focusing on recent developments in biopharmaceutical analysis.

When we were asked to edit a follow-up to

the LCGC Europe May 2013 supplement

“Advances in Pharmaceutical Analysis,” we

immediately wanted to highlight the chal-

lenges in biopharmaceutical analysis. Indeed,

within the pharmaceutical industry and also

within our own research activities related

to pharmaceutical analysis, there has been

a remarkable shift from small to large mol-

ecules. On the market since the early 1980s,

protein biopharmaceuticals have seen an

enormous growth in the last decade. It is

even expected that within the current decade,

more than 50% of new drug approvals will

be biological in nature. A dominant role is

thereby played by monoclonal antibodies

(mAbs), of which a substantial number have

reached blockbuster status. The top 10 best-

selling pharmaceuticals are currently heavily

populated by mAbs.

Protein biopharmaceuticals are large and

heterogeneous and their in-depth analysis

during development and also during their

lifetime requires the best of both chroma-

tography and mass spectrometry (MS). In

editing this special issue, we have therefore

selected authorities in the field to illustrate

the state of the art in biopharmaceutical

analysis.

The first contribution, authored by

Szabolcs Fekete, Jean-Luc Veuthey, and Davy

Guillarme, provides an overview of the dif-

ferent liquid chromatography (LC) column

formats recently introduced in the market

for reversed-phase, size-exclusion (SEC),

ion-exchange, and hydrophobic interaction

(HIC) chromatographic analyses of thera-

peutic proteins, mAbs, and antibody–drug

conjugates (ADCs).

In the May 2013 supplement we described

the features of liquid chromatography cou-

pled to mass spectrometry (LC–MS) in the

characterization of protein biopharmaceu-

ticals. With the patents of the first genera-

tion protein biopharmaceuticals expired and

blockbuster mAbs appearing on the market,

activities in biosimilars have exploded in

recent years. More than 15 biosimilars have

already been approved in Europe, and a ver-

sion of filgrastim was launched in the United

States as the first biosimilar toward the end

of 2015. Analytical methods to compare

originators with biosimilars are highlighted

in the second contribution from our team at

the Research Institute for Chromatography.

The antibody market has been reshaped

by various next-generation formats (biospe-

cific mAbs, antibody mixtures, nanobodies,

brain penetrant mAbs, glyco-engineered

formats), and in recent years the ADCs bren-

tuximab vedotin and trastuzumab emtan-

sine have been approved by the European

Medicines Agency (EMA) and the Food

and Drug Administration (FDA). In ADCs,

a cytotoxin is coupled to an antibody that

specifically targets a certain tumor marker.

As such, highly toxic drugs can be delivered

in a targeted fashion to tumor cells without

affecting healthy cells. Compared to naked

mAbs, the conjugation of cytotoxic drugs

further adds to the complexity. The power

of MS to unravel this complexity is illus-

trated in the paper authored by Alain Beck

and by Sarah Cianferani (available online

at: www.chromatographyonline.com/har-

nessing-benefits-mass-spectrometry-depth-

antibody-drug-conjugates-analytical-char-

acterization-0).

The previous two contributions clearly

illustrate the importance of MS in the eluci-

dation of the primary structure of therapeu-

tic proteins. Higher order elements, on the

other hand, can be derived from special MS

technologies such as native MS, ion mobil-

ity MS, hydrogen–deuterium exchange MS,

and chemical cross-linking MS. In the third

contribution, Christian Huber describes the

basic principles of these techniques and illus-

trates their features for the characterization of

higher order structures of some protein bio-

pharmaceuticals.

Traditionally, ligand-binding assays

(LBAs) are applied to study the pharma-

cokinetic behavior of protein biopharma-

ceuticals in biological fluids. LBAs are

characterized by a high throughput and

sensitivity, but may suffer from long devel-

opment times and potential interferences

from other proteins present in the matrix.

In addition, the generation of drug-specific

antibody tools is a time-consuming process.

Liquid chromatography coupled to tandem

mass spectrometry (LC–MS/MS) methods

are used more and more as alternatives to

LBAs, often offering improved figures-of-

merit while at the same time being generi-

cally applicable. Some of the technicalities

and advantages and disadvantages of LC–

MS/MS compared to LBAs for monitor-

ing biopharmaceuticals in biological fluids

are addressed in the fourth contribution by

Nico C. van de Merbel.

The presence of residual host cell proteins

(HCPs) is a potential safety risk in any bio-

pharmaceutical product. Despite enormous

purification efforts, these HCPs may be left

behind from the expression hosts. HCPs are

normally dosed during downstream process-

ing and in the final biopharmaceutical prod-

uct by enzyme-linked immunosorbent assays

(ELISA). As mentioned in the previous paper,

LBAs are more and more complemented

or even replaced by LC–MS/MS and this

is illustrated in the last contribution by our

group. The use of off-line two-dimensional

LC–MS/MS in the characterization of HCPs

is described and the added value of using

multidimensional chromatography is clearly

demonstrated.

We hope that the contributions in this

supplement are of interest and even a source

of inspiration to the numerous analysts in the

biopharmaceutical industry. It was a pleasure

for us to edit and review the contributions

of outstanding (preselected) colleagues. We

would like to thank all of them for their

excellent work.

Pat Sandra Koen Sandra

Page 6: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

6 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Szabolcs Fekete, Jean-Luc Veuthey, and Davy Guillarme

Modern Column Technologies for the Analytical Characterization of Biopharmaceuticals in Various Liquid Chromatographic Modes

The recent trends in column technology for reversed-phase liquid

chromatography (LC), size-exclusion chromatography (SEC), ion-exchange

chromatography, and hydrophobic interaction chromatography (HIC) for

analysis of biopharmaceuticals at the protein level is critically discussed.

Therapeutic proteins are large and het-

erogeneous molecules subjected to a

variety of enzymatic and chemical

modifications during expression, purifica-

tion, and long-term storage. These changes

include several possible modifications, such

as oxidation, deamidation, glycosylation,

aggregation, misfolding, or adsorption,

leading to a potential loss of therapeutic

efficacy or unwanted immune reactions.

Regulatory bodies require a detailed char-

acterization (for example, verifying primary

structure and appropriate post-translational

modifications, secondary and tertiary

structure), lot-to-lot and batch-to-batch

comparisons, stability studies, impurity

profiling, glycoprofiling, determination of

related proteins and excipients as well as

determination of protein aggregates. For

this purpose, a single analytical technique

is generally not sufficient, and a variety of

orthogonal methods are required to fully

describe such a complex sample.

Today, one of the most widely used ana-

lytical techniques for therapeutic protein

characterization is liquid chromatography

(LC). This is probably a direct result of

the remarkable developments of the past

few years, which have enabled a new level

of chromatographic performance. These

developments include ultrahigh-pressure

liquid chromatography (UHPLC), col-

umns packed with wide-pore superficially

porous particles (SPPs), and organic mono-

lith columns, which have allowed a dra-

matic increase in separation efficiency, even

with large intact biomolecules.

This article reviews the possibilities

and trends of current state-of-the-art LC

column technology applied for different

modes of chromatography for the charac-

terization of therapeutic proteins.

Hydrophobic Interaction

Chromatography

Hydrophobic interaction chromatography

(HIC) has been historically used for protein

purification; more recently, the two main

application fields have been in the determi-

nation of the drug-to-antibody ratio (DAR)

of antibody–drug conjugates (ADCs) and

in monitoring post-translational modifica-

tions of monoclonal antibodies (mAbs).

In HIC, proteins are retained and sepa-

rated on the basis of their hydrophobicity as

a result of the van der Waals forces between

the hydrophobic ligands of the stationary

phase and the nonpolar regions of proteins

(1). The binding of proteins to a hydropho-

bic surface is affected by a number of fac-

tors including the type of ligand, the ligand

density on the solid support, the backbone

material of the stationary phase, the hydro-

phobic nature of the protein, and the type

of salt added to the mobile phase. Dur-

ing the separation, a negative salt gradient

(typically from 2–3 M to 0 M) is applied

under aqueous mobile phase at around pH

6.8–7.0. The structural damage to the bio-

molecules is therefore minimal and its bio-

logical activity is maintained (2).

Analytical-scale HIC columns are based

either on silica or polymer particles. Both

porous and nonporous particles are avail-

Ph

oto

cred

it: J

org

Gre

uel

/Get

ty I

mag

es

Page 7: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

In the sample chromatograms shown here, 5 μm non-porous SP-F column was used for the separation of human IgG mAb charge variants. Even higher resolution was achieved using 3 μm particles.

Phone: +1.610.266.8650Email: [email protected]: www.ymcamerica.comStore: store.ymcamerica.com

With over 30 distinctly different phase chemistries, particle sizes from 1.9 μm to 75 μm, plus column confi gurations for Capillary LC, UHPLC, HPLC, and Prep (and bulk material too) … it’s hard to fi nd a bioseparations challenge that we can’t address.

YMC Products for BioseparationsYMC-BioPro SP-F for mAb Charge Variant Analysis

YMC-Pack Diol-SEC is a size exclusion medium utilizing a silica gel base. Diol-120, 200, and 300 are available in 2, 3, and 5 μm particle diameters, and are suitable for separation or molecular weight determination of proteins with molecular weights of 10,000 to several hundred thousand.

Diol-60 is the most suitable for separation of peptides or oligosaccharides whose molecular weights are 10,000 or less.

YMC-Triart is a hybrid reversed phase material, and YMC Meteoric Core is a core-shell material.

Both are well-suited to the rigors of bioseparations – they perform well in aqueous phases, respond well to gradient changes, and offer high resolution and long column life.

YMC-Triart is offered in capillary columns, UHPLC columns, and standard HPLC columns. Meteoric Core is offered in standard HPLC columns. Both are ideal choices for LC-MS.

Original separation using 5μm.

Switch to 3μm resolves additional peak.

YMC Products for Bioseparations

YMC-Pack Diol Size Exclusion (SEC) Columns for Separation of mAb

YMC-Triart and Meteoric Core for Peptide Mapping and Reversed Phase

YMC-Pack Diol, 2μm, 300Å, 150x4.6mmSmaller Particles and Smaller Columns Enable Lower Flowrates, Higher Throughput

5μm

2μmSample: 1mg/mL dilution of 25mg/mL AvastinMobile Phase: 100mM NaPO4 with 200mM NaCl at pH=7.0

Sample PreparationMonoclonal antibody samples were denatured, reduced (BME), and desalted prior to trypsin digestion.

Operating ParametersMobile Phase A: 0.1% TFA in HPLC WaterMobile Phase B: 0.1% TFA in AcetonitrileColumn Temp: 40°C Flowrate: 0.6mL/minInj. Volume: 10uL Detection: 215nm

GradientStep Time Flow %A %B

1 0 0.6 98 22 0.67 0.6 98 23 14 0.6 55 454 16 0.6 0 1005 16.67 0.6 98 26 20 0.6 98 2

Porous

Non-Porous

Particle Sizes:

3, 5, 6, 10, 30, 75 μm

Particle Sizes:

3, 5, 6, 10, 30, 75 μm

Page 8: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

8 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Table I: Recent state-of-the-art and some widely used “reference” columns applied for the separation of therapeutic proteins in HIC, SEC, ion-exchange, and reversed-phase LC modes

Column Name ChemistryParticle size/Macropore Size (μm)

Max Temperature

(°C)pH Range

Max Pressure

(bar)

HIC Columns

TSKgel (Tosoh)

Butyl-NPR C4 (non porous) 2.5

50 2–12

200

Ether-5PW Ether (porous) 1050

Phenyl-5PW Phenyl (porous) 10

Protein-Pak Hi Res HIC (Waters) C4 2.5 60 2–12 200

Thermo

MAbPac HIC-Butyl C4 5 60 2–12 300

MAbPac HIC-20 Alkylamide 5 60 2–9 400

ProPac HIC-10 Amide/ethyl 5 60 2.5–7.5 300

Ion-Exchange Columns

Proswift (Thermo)(monolith)

SAX-1SStrong anion exchange

(quaternary amine)

Information not

available

70

2–12 70

WAX-1SWeak anion exchange

(tertiary amine)60

WCX-1SWeak cation exchange

(carboxylic acid)60

SCX-1SStrong cation exchange

(sulfonic acid)60

TSKgel (Tosoh)

SCXStrong cation exchange

(sulfonic acid)5

45

2–14

50

SuperQ-5PWStrong cation exchange

(trimethylamino)10 2–12

SP-STATStrong cation exchange

(sulfopropyl)7, 10 3–10

Q-STATStrong anion exchange

(quaternary ammonium)7, 10 3–10

Bio Mab (Agilent)Weak cation exchange

(carboxylate)

1.7 3 5 10

80 2–12

270 410 550 680

Antibodix (Supelco, Sepax)Weak cation exchange

(carboxylate)

1.7 3 5 10

80 2–12

270 410 550 680

Protein-Pak Hi Res IEX (Waters)

SPStrong cation exchange

(sulfopropyl)7

60 3–10

100

CM Weak cation exchange

(carboxymethyl)7 100

QStrong anion exchange

(quaternary ammonium)5 150

MAbPac SCX-10 (Thermo)Strong cation exchange

(sulphonic acid)

3 5 10

60 2–12480 480 200

Bio-Pro (YMC)

QAQA-F

Strong anion exchange (quaternary ammonium)

5 60 2–12

30 120

SPSP-F

Strong cation exchange (sulfopropyl)

30 120

SEC Columns

Thermo Silica-based 3 60 2.5–7.5 200

YMC-Pack Diol-SEC Diol modified silica-based 5 40 5–7.5 200

Acclaim SEC-300 (Thermo)Hydrophilic polymethacrylate

resin5 60 2–12 1200

TSKgel SW aggregate (Tosoh) Diol 3 30 2.5–7.5 120

TSKgel SW mAb (Tosoh) Diol 4 30 2.5–7.5 120

SRT-SEC (Sepax) Surface-coated silica-based 5Information not

available2–8.5

Information not available

Zenix-SEC (Sepax) Surface-coated silica-based 3 ~250 2–8.5 80

Page 9: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 9www.chromatographyonline.com

able. Highly cross-linked nonporous

poly(styrene–divinylbenzene) (PS–DVB)

and polymethacrylate-based particles are

frequently used in protein separations as a

result of their advantageous mass transfer

properties (the main contribution to the

band broadening of large biomolecules,

namely trans-particle mass transfer resis-

tance is negligible). Table I summarizes

the most widely used and the latest HIC

columns applied for mAb and ADC sepa-

rations.

These materials can now withstand pres-

sure drops of up to 100–400 bar. Columns

are typically packed with 10-, 7-, 5-, 3-, and

even 2.5-μm particles. Column diameters

between 2 mm and 8 mm are available, but

4.6-mm i.d. columns are the most widely

used in current HIC applications. It is

worth mentioning that there is a need for

150 mm × 2.1 mm column formats, which

are often applied for the analysis of proteins

in modern chromatographic practice.

HIC allows both the characterization

of the distribution of drug-linked species

and the determination of average DAR of

ADCs (3). Conjugation of the drug-linker

to the antibody increases the hydrophobic-

ity; therefore, HIC appears as a suitable

tool to separate the different DAR species.

A good example of the HIC profile of a

native IgG1 ADC is shown in Figure 1 (3).

Recently off-line mass spectrometric

(MS) detection was applied for the char-

acterization of brentuximab-vedotin. Each

individual HIC peak was collected, buffer

exchanged, and analyzed by native MS

(4). HIC was also successfully applied for

monitoring various post-translational mod-

ifications, including proteolytic fragments,

domain misfolding, tryptophan oxidation,

and aspartic acid isomerization in therapeu-

tic mAbs (5).

Ion-Exchange Chromatography

Ion-exchange chromatography is widely

used for the characterization of therapeutic

proteins and can be considered as a

reference marker and powerful technique

for the qualitative and quantitative

evaluation of charge heterogeneity.

Table I: Continued

Column Name ChemistryParticle Size/Macropore Size (μm)

Max Temperature

(°C)pH Range

Max Pressure

(bar)

SEC Columns (continued)

Bio SEC (Agilent) surface-coated silica-based3 Information not

available2–8.5 240

5

Acquity UPLC BEH SEC (Waters) diol modified hybrid-based1.7

60 2–8 6002.5

Reversed-Phase LC Columns

ProSwift (Thermo)(Monolith)

RP-1S

Phenyl

1 70 1–14 200

RP-2H 2.2 70 1–14 200

RP-3U 5.1 70 1–14 200

RP-10RInformation not available

80 1–10 300

Acquity BEH 300 (Waters) C18, C4 1.7 80 1–12 1000

Zorbax (Agilent)

300SB RRHD C18, C8 1.8 80 1–8 1200

Poroshell SB300 C18, C8, C35 (0.25-μm thickness)

90 1–8 600

Poroshell 300Ex-tend

C185 (0.25-μm thickness)

60 2–11 600

AdvanceBio RP-mAb C8, C4, diphenyl3.5 (0.25-μm

thickness)90 1–8 600

Aeris (Phenomenex)

Widepore C18, C8, C43.6 (0.2-μm thickness)

90 (C18,C8), 60 (C4)

1.5–9 600

Peptide C18

3.6 (0.5-μm thickness)

2.6 (0.35 μm thickness)

1.7 (0.22-μm thickness)

90 1.5–9600

1000

Halo (Advanced Materials Technology)

Peptide C18, CN

2.7 (0.5-μm thickness)

4.6 (0.6 μm thickness)

100 1–9 600

Protein C18, C83.4 (0.2-μm thickness)

90 1–9 600

Flare Widepore (Diamond Analytics) C183.6 (0.1-μm thickness)

100 1–13 400

Page 10: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

10 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Among the different ion-exchange

modes, cation-exchange chromatography

is the most widely used for protein

characterization (6).

Two modes of elution are often applied

for protein characterization, namely the

salt-gradient and the pH-gradient. In

salt-gradient mode, solutes are eluted in

order of increasing binding charge, which

correlates more or less with the isoelectric

point (pI) and equilibrium constant. In

this case, the mobile-phase pH is kept

constant, while the ionic strength is

continuously increased. In pH-gradient

mode, the ionic strength is kept constant

and the pH is varied during the gradient

program. This mode of elution is often

referred to as chromatofocusing.

Regarding the stationary phase, there are

two main aspects that need to be considered

for successful ion-exchange separation: the

strength of interaction and associated reten-

tion (strong or weak ion exchanger), and

the achievable peak widths (efficiency) (7).

Both cation and anion exchangers can be

classified as either weak or strong exchang-

ers. Weak cation exchangers are composed

of a weak acid that gradually loses its

charge as the pH decreases (for example,

carboxymethyl groups), while strong cat-

ion exchangers are composed of a strong

acid that is able to sustain its charge over

a wide pH range (for example, sulfopropyl

groups). On the other hand, strong anion

exchangers contain quaternary amine func-

tional groups, while weak anion exchanger

possesses diethylaminoethane (DEAE)

groups. As a rule of thumb, it is preferable

to begin the method development with a

strong exchanger, to ensure that a broad pH

range can be worked on. Strong exchangers

are also useful if the maximum resolution

occurs at an extreme pH. However, silica-

based ion exchangers can only be operated

in a restricted pH range. In contrast, poly-

meric ion exchangers can be used over a

wide pH range.

Commercially available ion-exchange

columns are based on silica or polymer

particles but organic–polymeric monoliths

are also available. Both porous and

nonporous particles are available but

for large molecules, which possess low

diffusivity, nonporous materials are clearly

preferred. Highly cross-linked nonporous

PS–DVB materials are most frequently

used in protein separations because of their

high pH stability (2 ≤ pH ≤ 12). These

materials can now withstand a pressure

drop of up to 500–600 bar in some cases.

Columns packed with 10-, 5-, or 3-μm

nonporous particles are often used, but

sub-2-μm materials are also available to

perform UHPLC separations (see Table

I). Suitable peak capacity can be attained

with large biomolecules on those columns

within a reasonable analysis time (for

example, 15–20 min). However, some

limitations can be expected in terms of

loading capacity and retention when

applying nonporous materials.

A recent study systematically compared

the latest state-of-the-art cation exchanger

columns applied for the characterization of

therapeutic mAbs in pH- and salt-gradient

modes (8).

Figure 2 shows an example of the

separation of four intact antibody charge

variants using a 100 mm × 4.6 mm, 5-μm

strong cation polymeric exchanger column

packed with nonporous particles and a

20-min long gradient (7)

Size-Exclusion Chromatography

Size-exclusion chromatography (SEC) is a

powerful technique for the qualitative and

quantitative evaluation of protein aggre-

gates. The main advantage of SEC is the

mild mobile phase conditions that permit

the characterization of proteins with mini-

mal impact on the conformational struc-

ture and local environment.

200

150

100

50

1210

DAR 4

DAR 6

Time (min)

Sig

nal

DAR 8

DAR 2

DAR 0

8640

Figure 1: HIC separation of an ADC for the determination of DAR. Adapted and re-produced with permission from reference 3, ©American Chemical Society.

Sig

nal

2015

Time (min)

1050

1 2 3 4

Figure 2: Ion-exchange separation of four intact mAbs (natalizumab [1], cetuximab [2], adalimumab [3], and denosumab [4]). Adapted and reproduced with permission from reference 7, ©Elsevier.

Page 11: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 11www.chromatographyonline.com

SEC separates biomolecules according

to their hydrodynamic radius. The sta-

tionary phase consists of spherical porous

particles with a carefully controlled pore

size, through which the biomolecules

diffuse based on their molecular size

difference using an aqueous buffer as

the mobile phase. Basically, SEC is an

entropically controlled separation pro-

cess in which molecules are separated

on the basis of molecular size differences

(filtering) rather than by their chemical

properties (8). Therefore, retention fac-

tor (thermodynamic) in SEC is different

from other chromatographic modes. Here,

the thermodynamic retention factor is the

fraction of the intraparticle pore volume

that is accessible to the analyte (9).

Since no retention occurs in SEC, large

pore volumes (high porosity) are required

to ensure appropriate resolution. Gener-

ally, this large pore volume is provided by

long-and wide-bore columns. In routine

SEC applications, a 30-cm column length

with internal diameters (i.d.) of 7.8, 8.0, or

10 mm is generally employed. These SEC

columns are referred to as standard-bore col-

umns. Now, several vendors offer narrow-

bore columns with 4.6-mm i.d. and 15-cm

length that are packed with very efficient,

small particles of ~3 μm. Similar separation

power can be attained using these columns

as with 5-μm particles in 30-cm standard-

bore columns, but the analysis time can be

reduced by a factor of 3 to 4 (10).

There are mainly two types of SEC

packing materials: silica, with or without

surface modification, and cross-linked

polymeric packings, which possess non-

polar (hydrophobic), hydrophilic, or ionic

character (8). The most common silica

packing consists of chemically bonded

1,2-propanediol functional groups that

provide a hydrophilic surface. This sta-

tionary phase blocks or reacts with many

of the acidic silanol groups allowing the

surface to be neutralized. Bare silica is

also a suitable packing material for non-

aqueous polar or nonpolar organic mobile

phases; however, it is not recommended

with aqueous mobile phases because of

the presence of active silanol sites. The lat-

est type of silica-related packing is an eth-

ylene-bridged hybrid inorganic-organic

(BEH) material that is currently avail-

able at particle sizes of 1.7 μm—the first

sub-2-μm SEC packing—and 2.5 μm

(11). Compared to regular silica packings,

BEH particles have improved chemical

stability as well as reduced silanol activity.

The 1.7-μm BEH material can be oper-

ated at up to 600 bar.

There have been a number of different

hydrophilic cross-linked packings devel-

oped for the SEC of biopolymers. Most of

these packings are proprietary hydroxyl-

ated derivatives of cross-linked polymeth-

acrylates (10). Unusual polymeric pack-

ings for aqueous SEC include sulfonated

cross-linked polystyrene, polydivinylben-

zene derivatized with glucose or anion

exchange groups, a polyamide polymer,

and high-performance, crossed-linked

agarose (8).

Today, columns for aqueous and non-

aqueous SEC applications with pore sizes

of 125 to 900 Å are commercially avail-

able (12). Very fast separations of peptides,

myoglobin, and insulin aggregates have

been demonstrated with 1.7-μm SEC

columns (13). These columns were also

applied for the characterization of recom-

binant mAbs (11).

Applying 1.7- and 2.5-μm particles

in SEC has opened up a new level of

separation performance, but it should be

kept in mind that on very fine particles,

the separation quality is improved at the

cost of pressure (and frictional heating

temperature gradients). Therefore, there

is a risk of creating on-column aggregates

when analyzing sensitive proteins under

high-pressure (>200 bar) conditions (11).

Reversed-Phase

Liquid Chromatography

In reversed-phase liquid chromatography

(LC), the solute retention is predominantly

mediated through hydrophobic interac-

tions between the nonpolar amino acid

residues of the proteins and the bonded

n-alkyl ligands of the stationary phase.

Compared to the HIC mode, the reversed-

phase LC mobile phase typically consists

of water, acetonitrile or methanol, and

0.1–0.2% trifluoroacetic acid or formic

acid. The separation mechanism is based

on a combination of solvophobic and elec-

trostatic interactions, the latter being gov-

erned by the interaction of trifluoroacetic

acid with basic side chains of a few amino

acids (that is, arginine, lysine, and histi-

dine) and the N-terminus as well as ionic

interactions between the positive charges

at the surface of the protein and the nega-

tively charged residual silanols (14). The

efficiency of reversed-phase LC is always

superior to other chromatographic modes,

and its superior robustness makes it well

suited for use in a routine environment (15).

Current reversed-phase LC stationary

phases used for proteins analysis can be

classified as silica-based particulate mate-

rials and organic monoliths. The pore

size of particulate phases is an important

factor that must be considered. For the

analysis of peptides and small proteins, a

pore size of 100–200 Å may be acceptable.

However, porous materials with pore sizes

of greater than 200 Å are mandatory for

the separation of larger proteins or mAbs

fragments because the solute molecular

64 5

Time (min)

31 20

Sig

nal

L H

HC

2xLC2xHC

Reduction

+

Figure 3: Reversed-phase LC analysis of reduced IgG1 mAb. Unpublished results from the authors’ laboratory.

Page 12: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

12 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

diameter must be approximately one-

tenth the size of the pore diameter to

avoid the restricted diffusion of the solute

and to allow the total surface area of the

sorbent material to be accessible. An aver-

age pore size of 250–300 Å is often men-

tioned as the reference value for protein

separations, but recently it was shown that

400 Å particles completely eliminated

restricted diffusion effects for molecules

up to about 500 kDa.

The two main trends today in reversed-

phase LC analysis of therapeutic proteins

are the use of fully porous small par-

ticles (FPPs) (sub-2-μm) and superficially

porous particles (SPPs), which possess

particle sizes between 3 and 4 μm.

Columns packed with FPPs have

constraints in separation speed and

efficiency because of limitations in the

stationary phase mass transfer, which

results from the relatively long diffusion

times required for proteins to cross the

porous structure. Therefore, Horváth

first applied the concept of SPPs in the

late 1960s (16,17). They were initially

intended for the analysis of macromol-

ecules such as peptides and proteins. SPPs

are made of a solid, nonporous silica core

surrounded by a porous shell layer. They

have similar properties to the fully porous

materials conventionally used in high per-

formance liquid chromatography (HPLC).

The rationale behind this concept was to

improve column efficiency by shortening

the diffusion path that molecules must

travel, in addition to improving their mass

transfer kinetics.

It was recently shown that columns

packed with wide-pore 3.6-μm and 3.4-

μm SPPs showed significant gain in anal-

ysis time and peak capacity compared to

FPPs for intact protein analysis (18,19).

These wide-pore SPPs are now available

with C4, C8, and C18 chemistries and

can be operated up to 600 bar. Figure 3

shows an example of fast separation of

heavy-chain (Hc) and light-chain (Lc)

variants of an IgG1 mAb performed on a

wide-pore C4 SPP column.

In another study, efficiency and analy-

sis times of 1.7-μm SPPs and FPPs were

compared for peptides and moderate size

intact proteins (20). This study suggests a

two-fold increase in terms of achievable

peak capacity and analysis time for large

proteins when using SPPs compared to

FPPs of the same size. For the separation

of peptides and moderate size proteins, a

160 Å SP packing was also introduced

(21,22). Recently, 1.3- and 1.6-μm SPPs

were also applied for peptide mapping of

mAb samples (23,24). By combining long

columns (200–300 mm) with extended

analysis time, peak capacity around 1000

can be reached with 1.3-μm SPPs for

0.5–2 kDa peptides.

An alternative, carbon-nanodiamond-

based C18 superficially porous mate-

rial was recently introduced (25). The

core of this material is a carbonized

poly(divinylbenzene) particle with a

diameter of approximately 3.4-μm.

Poly(allylamine)-nanodiamond het-

ero-layers are deposited onto the surface of

the carbonized core by a modified layer-

by-layer method. The resulting core–shell

is synthesized to a shell thickness of ca. 0.1

μm and a finished particle size of 3.6 μm.

This superficially porous carbon-based

material was successfully applied for real-

life protein separations.

Another interesting alternative to SPPs

was proposed by Hayes and colleagues

(26). The so-called sphere-on-sphere

(SOS) approach provides a simple and

fast one-pot synthesis in which the thick-

ness, porosity, and chemical substituents

of the shell can be controlled by using

the appropriate reagents and conditions

(27). SOS particles have been shown to

be microporous with a pore diameter of

less than 2 nm. However, while the sur-

face of the material might not exhibit

significant porosity, when packed into a

HPLC column the spaces between surface

nanospheres provide superficial macropo-

rosity. It has been proposed that for large

molecules, larger pores as well as reduc-

tion of the shell thickness can be advan-

tageous, because of the shorter diffusion

distance and greater access to the surface

area of the material (28). SOS particles

were demonstrated to have similar chro-

matographic performance compared to

commercial SPP materials (26). Figure

4 shows the separation of reduced ADC

(brentuximab-vedotin) fragments on a

column packed with SOS particles.

As an alternative to particle-based sta-

tionary phase formats for the LC separa-

tions of proteins, organic polymer-based

monoliths offer some advantages, includ-

ing high permeability and rapid mass

transfer (29). Polymeric monolithic sta-

tionary phases have shown great poten-

tial for the reversed-phase LC separations

of large biomolecules, including intact

proteins, oligonucleotides, and peptides.

With this material, the mass transfer is

mainly driven by convection, rather than

diffusion, because of the absence of mes-

opores (30). The fact that the solvent is

forced to pass through the macropores of

the polymer because of pressure leads to

faster convective mass transfer compared

to the slow diffusion process into the stag-

nant pore liquid that is present in porous

beads packed columns. As a result of their

open channel structure, monoliths gener-

ally possess a high permeability, allowing

the application of elevated flow rates at

moderate back pressure. It was previously

demonstrated that polymeric stationary

Sig

nal

128 10Time (min)

62 40

H1

H0

L1L0

H2H3

Figure 4: Reversed phase LC separation of reduced ADC (brentuximab-vedotin) frag-ments. Unpublished results from the authors’ laboratory.

Page 13: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 13www.chromatographyonline.com

phases led to superior performance over

silica-based materials in the reversed

phase analysis of very large proteins (MW

> 50 kDa) (31).

Conclusion

There is always a need to use several chro-

matographic methods to draw reliable con-

clusions regarding the quality of biophar-

maceuticals. Ion-exchange chromatography,

SEC, and HIC are historical techniques

and are still used in any laboratory deal-

ing with the analytical characterization of

mAbs or ADCs. These techniques were

known to offer poor resolving power, which

is why the stationary phases employed in

ion-exchange chromatography, SEC, and

HIC have strongly evolved over the last few

years, in terms of chemistries, dimensions,

and chemical stability.

The most important improvements for

protein analysis were brought to reversed-

phase LC materials. In the past, this tech-

nique was rarely used for biopharmaceuti-

cal characterization. However, because

this is the only chromatographic approach

directly compatible with MS, providers

have improved and developed their exist-

ing materials. The performance that can

be achieved today with columns packed

with wide pore sub-2-μm fully porous or

sub-4-μm superficially porous are highly

competitive, and even if the selectivity of

reversed-phase LC is still limited for sepa-

rating charge or size variants of proteins,

this is (at least partially) compensated by

the high kinetic performance generated by

modern reversed-phase LC columns.

Acknowledgments

The authors acknowledge Alain Beck

(Pierre Fabre, Saint-Julien Genevois, France)

for providing mAb and ADC samples, and

Stephanie Schuster (Advanced Materials

Technology) and Tony Edge and Richard

Hayes (Thermo Fisher Scientific) for pro-

viding stationary phases.

Davy Guillarme wishes to thank the

Swiss National Science Foundation for

support through a fellowship to Szabolcs

Fekete (31003A_159494).

References

(1) C.J.V. Oss, R.J. Good, and M.K. Chaudhury,

J. Chromatogr. 376, 111–119 (1986).

(2) J.A. Querioz, C.T. Tomaz, and J.M.S. Cabral,

J. Biotech. 87, 143–159 (2001).

(3) L.N. Lee, J.M.R. Moore, J. Ouyang, X. Chen,

M.D.H. Nguyen, and W.J. Galush, Anal.

Chem. 84, 7479–7486 (2013).

(4) F. Debaene, A. Boeuf, E. Wagner-Rousset, O.

Colas, D. Ayoub, N. Corvaïa, A. Van Dors-

selaer, A. Beck, and S. Cianférani, Anal. Chem.

86, 10674–10683 (2001).

(5) M. Haverick, S. Mengisen, M. Shameem, and

A. Ambrogelly, mAbs 6, 852–858 (2001).

(6) S. Fekete, A. Beck, and D. Guillarme, Am.

Pharm. Rev. 18, 59–63 (2015).

(7) S. Fekete, A. Beck, and D. Guillarme, J.

Pharm. Biomed. Anal. 111, 169–176 (2015).

(8) H.G. Barth and G.D. Saunders, LCGC North

Am. 30, 544–563 (2012).

(9) P. Hong, S. Koza, and E.S.P. Bouvier, J. Liq.

Chrom. Rel. Techn. 35, 2923–2950 (2012).

(10) S. Fekete, A. Beck, J.L. Veuthey, and D. Guil-

larme, J. Pharm. Biomed. Anal. 101, 161–173

(2014).

(11) S. Fekete, K. Ganzler, and D. Guillarme, J.

Pharm. Biomed. Anal. 78–79, 141–149 (2013).

(12) E. Gazal, “Can Size Exclusion Chromatography

(SEC) Be Done on Sub-3-μm Particles?,”

presented at the 17th annual meeting of the

Israel Analytical Chemistry Society, Tel Aviv,

Israel, 2014.

(13) S.M. Koza, P. Hong, and K.J. Fountain,

“Advantages of Ultra Performance Liquid

Chromatography Using 125 Å Pore Size, Sub-

2-μm Particles for the Analysis of Peptides

and Small Proteins,” poster presented at

Medimmune, Rockville, Maryland, 2012.

(14) S. Fekete, J.L. Veuthey, and D. Guillarme, J.

Pharm. Biomed. Anal. 69, 9–27 (2012).

(15) K. Sandra, I. Vandenheede, and P. Sandra, J.

Chromatogr. A 1335, 81–103 (2014).

(16) C. Horvath, B.A. Preiss, and S.R. Lipsky, Anal.

Chem. 39, 1422–1428 (1967).

(17) C. Horvath and S.R. Lipsky, J. Chromatogr.

Sci. 7, 109–116 (1969).

(18) S. Fekete, R. Berky, J. Fekete, J.L. Veuthey,

and D. Guillarme, J. Chromatogr. A 1236,

177–188 (2012).

(19) S.A. Schuster, B.M. Wagner, B.E. Boyes, and

J.J. Kirkland, J. Chromatogr. A 1315, 118–126

(2013).

(20) S. Fekete, K. Ganzler, and J. Fekete, J. Pharm.

Biomed. Anal. 54, 482–490 (2011).

(21) F. Gritti and G. Guiochon, J. Chromatogr. A

1218, 907–921 (2011).

(22) S.A. Schuster, B.M. Wagner, B.E. Boyes, and

J.J. Kirkland, J. Chromatogr. Sci. 48, 566–571

(2010).

(23) S. Fekete and D. Guillarme, J. Chromatogr. A

1320, 86–95 (2013).

(24) B. Bobály, D. Guillarme, and S. Fekete, J. Sep.

Sci. 37, 189–197 (2014).

(25) B. Bobály, D. Guillarme, and S. Fekete, J.

Pharm. Biomed. Anal. 104, 130–136 (2015).

(26) R. Hayes, P. Myers, T. Edge, H. Zhang, Ana-

lyst 139, 5674–5677 (2014).

(27) (A. Ahmed, W. Abdelmagid, H. Ritchie, P.

Myers, and H. Zhankg, J. Chromatogr. A

1270, 194–203 (2012).

(28) L.E. Blue and J.W. Jorgenson, J. Chromatogr. A

1218, 7989–7995 (2011).

(29) C. Viklund, F. Svec, J.M.J. Fréchet, and K.

Irgum, Chem. Mater. 8, 744–750 (1996).

(30) M. Petro, F. Svec, I. Gitsov, and J.M.J. Fréchet,

Anal. Chem. 68, 315–321 (1996).

(31) S. Fekete, J.-L. Veuthey, S. Eeltink, and D.

Guillarme, Anal. Bioanal. Chem. 405, 3137–

3151 (2013).

Szabolcs Fekete holds a PhD degree

in analytical chemistry from the Techni-

cal University of Budapest, Hungary. He

worked at the Chemical Works of Gedeon

Richter Plc at the analytical R&D depart-

ment for 10 years. Since 2011, he has

worked at the University of Geneva in

Switzerland. He has contributed 70 jour-

nal articles and authored book chapters.

His main interests include liquid chroma-

tography, column technology, pharmaceu-

tical, and protein analysis.

Jean-Luc Veuthey is professor at

the School of Pharmaceutical Sciences,

University of Geneva, Switzerland. He has

also acted as President of the School of

Pharmaceutical Sciences, Vice-Dean of the

Faculty of Sciences, and finally Vice-Rector

of the University of Geneva. His research

domains include development of separa-

tion techniques in pharmaceutical sciences,

and, more precisely, the study of the

impact of sample preparation procedures

in the analytical process; fundamental

studies in liquid and supercritical chroma-

tography; separation techniques coupled

with mass spectrometry; and analysis of

drugs and drugs of abuse in different

matrices. He has published more than 300

articles in peer-reviewed journals.

Davy Guillarme holds a PhD degree

in analytical chemistry from the University

of Lyon, France. He is senior lecturer at

the University of Geneva in Switzerland.

He has authored 140 journal articles relat-

ed to pharmaceutical analysis. His exper-

tise includes HPLC, UHPLC, HILIC, LC–MS,

SFC, and analysis of proteins and mAbs.

He is an editorial advisory board member

of several journals including Journal of

Chromatography A, Journal of Separation

Science, and LCGC North America. ◾

Page 14: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

14 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Koen Sandra, Isabel Vandenheede, Emmie Dumont, and Pat Sandra

Monoclonal Antibodies and Biosimilars—A Selection of Analytical Tools for Characterization and Comparability Assessment

Monoclonal antibodies (mAbs) have emerged as important therapeutics

for the treatment of life-threatening diseases including cancer and

autoimmune diseases. With the top-selling mAbs evolving out of patent

there has been a growing interest in the development of biosimilars.

In demonstrating comparability to the originator product, biosimilar

developers are confronted with an enormous analytical challenge. This

article presents a selection of state-of-the-art analytical tools for mAb

characterization and comparability assessment.

It was Paul Ehrlich who, around 1900,

reported on “magic bullets” to cure a

wide range of diseases, thereby indi-

rectly referring to antibodies (1,2). The

development of the hybridoma tech-

nology by Köhler and Milstein, which

allowed the production of monoclonal

antibodies (mAbs), bridged the gap

between concept and clinical reality (3).

Since the approval of the first therapeu-

tic murine mAb in 1986, advances in

antibody engineering have allowed the

production of chimeric (mouse–human),

humanized, and human monoclonal

antibodies, thereby substantially improv-

ing safety and efficacy and paving the

way for the full exploitation of mAbs for

therapeutics purposes (4,5). More than

40 mAbs are now marketed in the United

States and Europe for the treatment of a

variety of diseases including cancer and

autoimmune diseases (6,7).

In 2013, 18 mAbs displayed block-

buster status and six of these prod-

ucts had sales greater than $6 billion

(Humira, Remicade, Enbrel, Rituxan,

Avastin, and Herceptin). MAbs are cur-

rently considered to be the fastest grow-

ing class of therapeutics with sales grow-

ing from $39 billion in 2008 to almost

$75 billion in 2013, a 90% increase.

Sales of other recombinant protein bio-

pharmaceuticals have only increased

by 26% in the same time period, while

small-molecule drugs are stagnating

(6,7). The successes of their predeces-

sors have triggered the development of

various next-generation mAb formats

such as bispecific mAbs, antibody–drug

conjugates (ADC), antibody mixtures,

antibody fragments (nanobodies, Fab),

Fc fusion proteins, and brain penetrant

mAbs next to glyco-engineered formats

(4,5,8). With several hundreds of prod-

ucts in preclinical development, the

future looks very bright.

The knowledge that the top-selling

mAbs are, or will become, open to the

market in the coming years has resulted

in an explosion of biosimilar activi-

ties. Last year witnessed the European

approval of the first two monoclonal

antibody biosimilars (Remsima and

Inflectra), which both contain the same

active substance, inf liximab (9). Remi-

cade, inf liximab’s blockbuster origina-

tor, reached global sales of $8.9 billion

in 2013. It is clear that the biosimilar

market holds great potential, but it is

simultaneously confronted with major

hurdles. In contrast to generic versions

of small molecules, exact copies of

recombinant mAbs cannot be produced

because of differences in the cell clon-

ing and the manufacturing processes

used. Even originator companies expe-

rience lot-to-lot variability. As a con-

sequence, regulatory agencies evaluate

Ph

oto

cred

it: J

un

os/G

etty

Im

ages

Page 15: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 15www.chromatographyonline.com

biosimilars based on their level of simi-

larity to, rather than the exact replica-

tion of, the originator. In demonstrating

similarity, an enormous weight is placed

on analytics, and both the biosimilar

and originator need to be characterized

and compared in great detail. In con-

trast to small-molecule drugs, mAbs are

large and heterogeneous (as a result of

the biosynthetic process and subsequent

manufacturing and storage), making

their analysis very challenging (10–13).

This article reports on selected state-of-

the-art chromatographic and mass spec-

trometric tools for detailed mAb charac-

terization and comparability assessment.

Protein A Chromatography

for Clone Selection

Protein A from Staphylococcus aureus has

a very strong affinity for the Fc domain

of IgG, allowing its capture from com-

plex matrices such as cell culture super-

natants. Affinity chromatography mak-

ing use of Protein A is the gold standard

in therapeutic mAb purification and

typically represents the first chromato-

graphic step in downstream processing.

Protein A chromatography finds applica-

tions beyond this large-scale purification.

At the analytical scale it is being used

early on in the development of mAbs

for the high-throughput determination

of mAb titer and yield directly from

cell culture supernatants and to purify

microgram amounts of material for fur-

ther measurements by techniques such as

mass spectrometry (MS) and chromatog-

raphy (14–16).

Figure 1 shows an overlay of the

protein A chromatograms of 12 trastu-

zumab-producing Chinese hamster

ovarian (CHO) clones, generated in the

framework of a Herceptin biosimilar

development program. Herceptin (sci-

entific INN name trastuzumab) is being

used in the treatment of HER2 positive

breast cancer. It is open to the European

market and evolves out of patent in the

United States in 2019 (17). Given its

market potential (global sales of $6.5

billion in 2013), dozens of companies

are actively developing a Herceptin

biosimilar. The unbound CHO mate-

rial is eluted in the f low-through while

the mAb is captured and only released

after lowering the pH. From these chro-

matograms, a distinction can already

be made between low and high mAb

producing clones. Absolute mAb con-

centrations can be determined by link-

ing the peak areas to an external cali-

bration curve constructed by diluting

Herceptin originators. Obtained mAb

titers are visualized in the bar plot in

Figure 1. From the findings, clear deci-

sions can be made for further biosimilar

development, that is, high trastuzumab

producing clones can be selected and

taken further in development.

Next to the mAb titer, the second

important criterion in clone selection is

based on the structural aspects. In the case

of biosimilar development, the structure

should be highly similar to the originator

product, within the originator batch-to-

batch variations. Figure 2 shows the ion

mobility (IM) quadrupole time-of-flight

(QTOF) MS measurements of inter-chain

reduced Herceptin and protein A–puri-

fied trastuzumab from a high titer CHO

84876

1106

The Ascentis Express column line is completely scalable from UHPLC to

legacy HPLC systems. Make method development seamless across your

organization with three particle sizes – 2.0, 2.7, and 5.0 μm and nine

unique phase chemistries:

Ascentis® Express UHPLC

and HPLC ColumnsFaster HPLC on Any System

For videos, product information, ordering and real-time

availability, visit sigma-aldrich.com/express

<�C18

<�C8

<�RP-Amide

<�Phenyl Hexyl

<�Biphenyl

<�F5

<�ES-Cyano

<�HILIC (Si)

<�OH5

©2016 Sigma-Aldrich Co. LLC. All rights reserved. Sigma-Aldrich and Supelco are trademarks

of Sigma-Aldrich Co. LLC, registered in the US and other countries. Ascentis is a registered

trademark of Sigma-Aldrich Co. LLC. Solutions within is a trademark of Sigma-Aldrich Co. LLC.

Sigma-Aldrich Corp. is a subsidiary of Merck KGaA, Darmstadt, Germany.

Page 16: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

16 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

clone. Samples were introduced into the

MS system via a reversed-phase on-line

desalting cartridge and light (Lc) and

heavy chain (Hc) were resolved in the IM

drift cell. Two Lc forms with identical

m/z and molecular weight (MW) but with

a different drift time, hence conformation,

are highlighted. Deconvoluted spectra

reveal that clone derived trastuzumab and

originator display the same Lc and Hc

MW values. In addition, the same N-gly-

cans, which are of the complex type, are

observed on the Hc of the originator and

clone derived mAb. These are considered

the most important attributes of biosimi-

larity according to US and European

regulatory authorities (primary sequence

should be identical and glycosylation

should be preserved). While glycosylation

is similar from a qualitative perspective,

quantitative differences are observed. Our

experience in clone selection studies has

found that it is not always the case that

MW values are identical between origi-

nators and mAbs derived from high-titer

clones or subclones. In these situations,

mAbs are typically not taken further in

development.

Reversed-Phase LC–MS Analysis

of Intact, Reduced, Papain, and

IdeZ Cleaved mAb

When a mAb is taken further in devel-

opment, a detailed characterization

and comparability assessment has to be

performed. Structural characteristics

such as amino acid sequence and com-

position, molecular weight and struc-

tural integrity, N- and O-glycosylation,

N- and C-terminal processing, S-S

bridges, deamidation (asparagine, glu-

tamine), aspartate isomerization, and

oxidation (methionine, tryptophan)

need to be assessed. In that respect,

reversed-phase liquid chromatography

(LC) is extremely powerful. Figure 3

shows highly efficient reversed-phase

LC–ultraviolet (UV) chromatograms

obtained on intact, inter-chain reduced,

papain-digested, and nonreduced and

reduced IdeZ-cleaved Herceptin. All

of these chromatograms are generated

using exactly the same chromatographic

conditions making use of widepore sub-

2-μm C8 particles, elevated column

temperatures (80 °C), and trif luoroace-

tic acid as ion-pairing reagent in a water–

acetonitrile mobile phase system. Under

these conditions, many of the challenges

encountered in performing reversed-

phase LC of proteins (peak tailing, peak

broadening, and adsorption) are tackled

(18–19). Moreover, these conditions are

compatible with MS, which allows an

in-depth characterization and compara-

bility assessment of mAbs.

Figure 4 shows the reversed-phase

LC–UV–MS analysis of IdeZ-cleaved

and TCEP-reduced Herceptin originator

and biosimilar. IdeZ or immunoglobu-

lin-degrading enzyme from Streptococ-

cus equi ssp zooepidemicus is a highly

specific protease similar to IdeS that

cleaves mAbs at a single site below the

hinge region, yielding F(ab′)2 and Fc/2

fragments (20,21). Following reduction,

the F(ab′)2 fragment is converted into

the Lc and Fd′. From the simultaneously

0,8

0,7

0,6

0,5

0,4

0,3

0,2

0,1

0,03

Co

nce

ntr

ati

on

(m

g/m

L)

6 8 9 10 14 24

Clone

Time (min)

mA

U

21.510.50

700

600

500

400

300

200

100

25 26 27 28 32

0

mAb

Figure 1: Overlaid UV 280 nm protein A chromatograms of 12 trastuzumab-produc-ing CHO clones with graphical representation of the mAb titer.

mAb structure

mAbs are tetrameric immunoglobulin G (IgG) molecules with a MW of 150 kDa

composed of two light (Lc – 25 kDa) and two heavy (Hc – 50 kDa) polypep-

tide chains connected through inter-chain disulphide bridges. Twelve intra-chain

disulphide bridges, four within each Hc and two within each Lc, furthermore

guarantee its structural integrity. Six different globular domains, that is, one vari-

able (VL) and one constant domain (CL) for the Lc and one variable (VH) and

three constant domains (CH1, CH2, CH3) for the Hc, are recognized. The struc-

ture can also be divided in the antigen-binding fragment (Fab), composed of VL,

CL, VH, and CH1 and the crystallizable fragment (Fc) composed of CH2 and

CH3. Antigen-binding is mediated by the Fab fragment while the Fc fragment is

responsible for the effector function, that is, antibody-dependent cell-mediated

cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). All mAbs

are glycoproteins with two conserved N-glycosylation sites in the Fc region that

can be occupied with complex and high mannose type N-glycans. These glycan

structures are known to play a role, amongst others, in the effector function.

Page 17: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 17www.chromatographyonline.com

acquired MS data it can be deduced

that peaks b, b′, d, d′, g, and g′, corre-

sponding to, respectively, Fc/2, Lc and

Fd′, are identical in both the origina-

tor and biosimilar. The measured MW

values obtained are well below 0.005%

of the theoretical MW values, which is

expected when using high-resolution

and accurate mass instrumentation.

Upon examining the spectra of the Fc/2

fragment, the biosimilar appears to be

enriched in the N-glycan G0F while a

more even distribution between G0F

and G1F is observed in the originator.

This is also ref lected in the chromato-

graphic peak shape. The broader peak

b′ indicates a partial separation of the

G0F and G1F species, with the former

eluting slightly later. Several other differ-

entiating peaks are observed in the sepa-

ration of the biosimilar, that is, peaks a,

c, e, and f. Compared to peak b, peak a

displays a 128 Da mass increase, which

can be explained by the presence of a

C-terminal lysine. To provide some more

background on this particular event, the

Hc is cloned with a lysine residue at the

Reduction

Reduction

Papain

2 * Lc 2 * Hc

2 * Fab 2 * Fc

1 * F(ab)’2

F(ab)’2 Fd’

Fc/2

Lc

Fc

Fab

Hc

Lc

2 * Fc/2 2 * Lc 2 * Fd’ 2 * Fc/2

Fc/2

IdeZ

6 8 10 12 14 16 18

6 8 10 12 14 16 18

20

64 8 10 12 14 16 18 20 228 10 12 14 16 18 20 22

Time (min)

Time (min)

Time (min)

Time (min)

Time (min)

10 12 14 16 18 20 22

mA

U

mA

U

mA

Um

AU

mA

U

200

150

100

50

0

500

400

300

200

100

0

500

400

300

200

100

0

500

400

300

200

100

0

240

200

160

120

80

0

40

35

30

251000 1500

Drift Time (ms) vs. m/z

2000 2500 1290 1295 1300 13101305

Drift Time (ms) vs. m/z

1315

195025

30

35

40

32

31

34

33

36

35

38

37

1952 1954 1956 1958Drift Time (ms) vs. m/z

1960

Heavy chain

44+

12+

12+ 11+10+

9+

13+

13+

14+15+

16+17+

18+

40+38+

36+

42+

Light chain

Light chain

Light chain (12+)

Light chain (12+)

Light chain 1 (18+)

G0G0F G1F

G2F

Heavy chain (39+)

23100 23200 23300

23439.9

23440.1

0

1

2

3

x106

0

1

2

3

x106

0

1

2

3

4

x106

0

1

2

x106

Counts vs. Deconvoluted Mass (amu) Counts vs. Deconvoluted Mass (amu)

23400 23500 23600 23700 23800 23900 49800 50200

50598.4

50760.6

50922.4

50922.850452.3

50598.150760.7G0F

G0 G2F

Originator

Biosimilar

Originator

Biosimilar

G1F

50600 51000 51400 51800

Figure 3: Reversed-phase LC–UV separations of intact, dithiothreitol (DTT)-reduced, papain-digested, nonreduced IdeZ-cleaved and tris(2-carboxyethyl)phosphine (TCEP) reduced IdeZ-cleaved Herceptin. These represent extremely powerful separations for comparability assessment and for detailed characterization. Conditions are compat-ible with MS allowing identification of the observed peaks.

Figure 2: IM-QTOF-MS profile of inter-chain reduced Herceptin (top). Deconvoluted light and heavy chain spectra of a Herceptin originator and a trastuzumab-producing clone (bottom).

Page 18: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

18 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

C-terminus. During protein matura-

tion, this lysine is removed by host cell

carboxypeptidases. This process is more

dominant in the host cell producing the

originator product than in the host cell

producing the biosimilar mAb. From the

MS data it can be deduced that peak c

originates from the Lc plus 1 and 2 hex-

ose units. This potentially originates

from a glycation event, which appears to

be negligible in the originator mAb. Peak

e shows a 1 Da mass increase compared

to peak d, indicating a deamidation in

the Lc. This event is apparent in both

the originator and biosimilar with an

increased occurrence in the biosimilar.

In analogy with peak c, peak f displays

162 Da spacings on Fd′, which is indica-

tive of glycation.

Reversed-Phase LC–MS

Analysis for Peptide Mapping

As previously demonstrated, protein mea-

surement is extremely powerful but does

not provide the complete picture. While

it is indicative for identity and highlights

dominant modifications, it does not pro-

Fc/2Lc Fd’

Biosimilar

Originator

b

a

b’

8 9 10 11 12 13

Response Units vs. Acquisition Time (min)

x10 2

x10 2

2

1.5

1

0.5

2.5

x10 3

x10 4

x10 4

5

x10 3

8

6

4

2

0

x10 4

8

x10 5

1.25

1

0.75

0.5

0.25

0

6

4

2

0

Fc/2 + K(G0F) a

b

b’

25365.2

Fd’ + 1 Hex

Fd’ + 2 Hex

f

g

g’

25707.6

Fd’

25384.0

Fd’25384.3

25546.1

x10 3

x10 4

x10 3

x10 5

1

0.5

0

4

2

S

S

S

S

S

N

DS

S

S

S

0

2

3

2

1

0

0

7.5

5

2.5

Lc + 2 Hex

Lc + 1 Hex

Lcd

c

d’

e

23766.8

23605.4

23443.8

Lc + deam23444.7

Lc23443.7

Fc/2 (G0F)

NHYTQKSLSLSPG

NHYTQKSLSLSPGK

25237.0

Fc/2 (G1F)25399.2

Fc/2 (G1F)

Fc/2 (G2F)

25399.1Fc/2 (G0F)

Fc/2 (G0)

25236.9

25090.7

25560.9

25254.0

4

3

2

1

0

4

3

2

1

0

2.5

2

1.5

1

0.5

024800 24900 25000 25100 25200 25300 25400

Counts vs. DeconvolutedMass (amu)

24800 25000 25200 25400 25600 25800 26000 26200 26400 2660024600

Counts vs. DeconvolutedMass (amu)

22800 23000 23200 23400 23600 23800 24000 24200 24400

Counts vs. DeconvolutedMass (amu)

25500 25600 25700 25800 25900 26000

2

1.5

1

0.5

0

0

14 15 16 17 18 19 20 21

d’g’

c

d

ef

g

Figure 4: Reversed-phase LC–UV–MS analysis of IdeZ-cleaved and TCEP-reduced Herceptin originator and biosimilar and decon-voluted MS spectra associated with the annotated peaks.

T45 + G2F

Originator

Originator

Biosimilar

Biosimilar

Originator

Biosimilar

Originator

Biosimilar

T45 + G0F

T45 + G1Fb

T45 + G1FaT45 + G0

T45 + G0F

T45 + G1F T45 + G0

T62

T62

T3

T3

13.1%T3 deam

10.5%T62+K

0

20 40 60 80

100

120

0

20

40

60

80

5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33

10.6 10.8 11 11.2 15.4 16.2 17 17.8 18.4 18.8 19.2

Response Units vs. Acquisition Time (min)

Figure 5: Reversed-phase LC–UV peptide map of Herceptin originator and biosimi-lar with detail in some specific regions showing post-translational modifications. T45: EEQYNSTYR, T62: SLSLSPG, T3: ASQDVNTAVAWYQQK. Peak identities were as-signed by the simultaneously acquired MS and MS/MS data.

Page 19: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 19www.chromatographyonline.com

CEX

SEC

HIC

Asndeamidation

mA

Um

AU

mA

U

1000

800

600

400

200

35

350

300

250

200

150

100

50

0

30

25

20

15

10

5

0

2

10 12 14 16 18 20

4 6 8 10 12 14 16

0

12.5 15 17.5 20 22.5 25 27.5 30 32.5

Buffer excipients

Dimer0.4%

Time (min)

Time (min)

Time (min)

Figure 6: CEX, SEC, and HIC separations of Herceptin. These techniques are used in characterization and release testing.

vide the actual amino acid sequence nor

does it localize the modifications. For

example, the measurement presented in

Figure 4 reveals a deamidation on the Lc

(peak e) but it cannot be traced back to a

specific asparagine or glutamine residue.

The Lc of the measured mAb contains

six asparagine and 15 glutamine resi-

dues, which are all prone to this chemical

modification. These characteristics can

further be assessed at the peptide level

following proteolytic digestion. When

digesting Herceptin with the enzyme

trypsin, which cleaves the protein next to

arginine and lysine residues, 62 identity

peptides are formed. Taking into account

post-translational modifications and

incomplete and aspecific cleavages taking

place, more than 100 peptides with vary-

ing physicochemical properties in a wide

dynamic concentration can be expected.

This is a particularly complex sample and

demands the best in terms of separation

technique. Again, reversed-phase LC

is the method of choice to resolve these

complex mixtures. Figure 5 shows the

UV peptide maps of both the origina-

tor and biosimilar. By taking advantage

of the simultaneously acquired MS data,

more than 99% of the peptide sequence

can be covered in both the originator

and biosimilar thereby confirming iden-

tity. While peptide maps are highly com-

parable, differences in post-translational

modifications can be detected (Figure

5). Obtaining a good knowledge of all

of these modifications is important since

they could be critical to the potency and

safety of a mAb. A deviating glycosylation

profile between originator and biosimilar

is already revealed at the protein level (Fig-

ure 4). At the peptide level, the different

N-glycosylated variants are nicely resolved

chromatographically and are shown to be

located on peptide EEQYNSTYR. Again,

the undergalactosylation of the biosimilar

is apparent. The peptide map also reveals

the presence of a lysine at the C-terminal

peptide of the heavy chain (SLSLSPGK)

and slightly increased deamidation in a

light chain peptide (ASQDVNTAVAWY-

QQK). This particular peptide contains

four potential deamidation sites (3 Gln

and 1 Asn). Based on MS measurement

one cannot discriminate between the

four sites. Upon performing MS/MS and

carefully interpreting the fragment ions

observed, the deamidation can be traced

back to the N (11). This deamidation in

fact corresponds to the deamidation event

observed in the reduced IdeS digest (Fig-

ure 4). At that time this event could be

linked to the Lc but could not be traced

back to a specific residue.

As discussed, mAb digests can be quite

complex and their analysis demands the

best in terms of separating power. If one-

dimensional (1D) separations are not able

to provide the separation power needed,

one can opt for two-dimensional (2D)

LC. Compared to 1D-LC, 2D-LC and

especially comprehensive LC (LC×LC)

will drastically increase resolution. We

have recently described the analysis of

Herceptin originator and biosimilar

digests on the combination reversed-

phase LC×reversed-phase LC (22). It is

important to point out that orthogonality

in reversed-phase LC×reversed-phase LC

peptide mapping is only obtained when

operating the two reversed-phase LC col-

umns at different pH values. This is a

direct result of the zwitterionic nature of

peptides, which gives rise to major selec-

tivity differences at pH extremes. These

reversed-phase LC×reversed-phase LC

Page 20: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

20 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

peptide maps provide a wealth of infor-

mation and allow both identity and

purity to be assessed. This makes it an

attractive technology for the comparison

of different production batches and to

compare innovator biopharmaceuticals

with biosimilars.

Native Chromatographic Tools:

Size-Exclusion Chromatography,

Cation-Exchange Chromatogra-

phy, and Hydrophobic Interac-

tion Chromatography

In contrast to reversed-phase LC,

size-exclusion chromatography (SEC),

ion-exchange chromatography, and

hydrophobic interaction chromatog-

raphy (HIC) are nondenaturing tech-

niques that provide complementary

information to the afore-mentioned

chromatographic mode (Figure 6).

These techniques are used early on in

mAb characterization and comparability

assessment and are subsequently applied

in routine testing. A major advantage

of these chromatographic modes com-

pared to reversed-phase LC is that they

preserve the structure, and so minor

variants can be collected and subjected

to complementary techniques such

as potency determination. SEC, ion-

exchange chromatography, and HIC are

not directly compatible with MS because

of the presence of nonvolatile salts in

the mobile phases. The identification

of peaks requires their collection and

subsequent desalting or dilution before

MS measurement. Desalting of the col-

lected fractions can be performed in an

automated manner using a setup such as

a small reversed-phase cartridge hyphen-

ated to an MS system.

Ion-exchange chromatography is an

excellent tool to highlight charged vari-

ants that might arise from modifications

such as deamidation, lysine trunca-

tion, or N-terminal cyclization (23,24).

Since most therapeutic mAbs have a

higher proportion of basic residues,

cation-exchange chromatography is the

most commonly used technique. The

cation-exchange separation of Herceptin

(Figure 6) highlights the asparagine

deamidation discussed earlier. A deami-

dation renders a protein more acidic,

which explains this earlier elution.

SEC is the chromatographic mode

with the lowest efficiency or resolution

of the afore-mentioned techniques, but

it is extremely powerful when determin-

ing aggregation and fragmentation. It is

recognized that aggregates may stimulate

immune responses and it is therefore very

important to measure this critical quality

attribute. Aggregation can typically not

be assessed using the other chromato-

graphic modes discussed. Figure 6 pres-

ents the SEC analysis of Herceptin and

illustrates that dimers can be measured

accurately at levels as low as 0.4%.

In recent years, HIC has been revisited

mainly from the perspective of ADC’s

governing a separation based on the

number of conjugated drugs allowing

the drug-to-antibody ratio (DAR) to be

determined. In the separation of naked

mAbs it is useful to highlight heteroge-

neities originating from oxidation, aspar-

tate isomerization, deamidation, succin-

imide formation, C-terminal lysine, and

clipping. The HIC analysis of Herceptin

gives rise to a single chromatographic

peak (Figure 6).

Hydrophilic Interaction

Liquid Chromatography

for Glycan Profiling

As demonstrated, glycosylation can be

(a) OriginatorG0F

G0F

G1Fa

G1FaMan5G0

F-G

lcN

Ac

G1Fb

G1Fb

G2FG0

Biosimilar

LU14

12

10

8

6

4

2

0

LU

14

16

10 12.5 15 17.5 20 22.5 25 27.5

(b) Biosimilar

Biosimilar: 4x

Biosimilar: 8x

Biosimilar: 16x

Biosimilar: 24x

LU

10

10

5

0

LU

8

4

0

LU

8

4

0

LU8

4

0

LU8

4

0

12.5 15 17.5 20 22.5 25 27.5

10 12.5 15 17.5 20 22.5 25 27.5

10 12.5 15 17.5 20 22.5 25 27.5

10 12.5 15 17.5 20 22.5 25 27.5

10 12.5 15 17.5 20 22.5 25 27.5

Time (min)

Time (min)

Time (min)

Time (min)

Time (min)

Time (min)

Time (min)

10 12.5 15 17.5 20 22.5 25 27.5

12

10

8

6

4

2

0

Figure 7: (a) Overlaid HILIC-FLD chromatograms of the 2-AB labeled N-glycans en-zymatically released from Herceptin originator and Protein A purified biosimilar. (b) N-glycan profiles of the biosimilar obtained by growing the CHO clone at dif-ferent galactose, uridine, and manganese chloride concentrations. Separations were performed on superficially porous HILIC particles.

Page 21: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 21www.chromatographyonline.com

revealed at both the protein and peptide

level. A detailed insight into the sugars,

however, can only be obtained follow-

ing their removal from the protein–pep-

tide backbone. This is preferably done

enzymatically using the deglycosidase

PNGase F. The liberated sugars are sub-

sequently labeled via reductive amina-

tion to improve their chromatographic

separation and detectability (f luores-

cence or mass spectrometric detection).

The f luorescence trace is typically used

for quantitative purposes while the MS

trace is used for qualitative purposes.

Figure 7a displays the analysis of 2-ami-

nobenzamide (2-AB) labelled Herceptin

originator and biosimilar N-glycans

using hydrophilic interaction chroma-

tography (HILIC) with f luorescence

detection (FLD). In this particular case,

a column packed with superficially

porous HILIC particles compatible

with 600 bar high performance liquid

chromatography (HPLC) instrumenta-

tion was used. This measurement pro-

vides information on the glycans and

allows structural isomers, that is, G1Fa

and G1Fb which differ in the position-

ing of the galactose residue either on

the α1-3 or α1-6 branch of the complex

type glycan, to be resolved.

The same type of complex N-glycans

are observed on both the originator and

biosimilar but quantitative differences

are revealed with an overexpression of

G0F species on the biosimilar, which is

in accordance with the measurements

performed at protein and peptide level.

Since glycosylation is a critical quality

attribute, this undergalactosylation does

not make the product similar enough to

be considered by regulatory authorities

as a Herceptin biosimilar.

The biosimilar-producing CHO

cell culture medium was subsequently

tuned by feeding uridine (U), galac-

tose (G), and manganese chloride (M)

at different concentrations (25). These

are the substrates and activator of the

galactosyltransferase responsible for

donating galactose residues to G0F and

G1F acceptors. Figure 7b shows the

N-glycan profiles obtained by growing

the biosimilar producing CHO clone at

different U, G, and M concentrations.

It is observed that the ratio G1F–G0F

increases with increasing concentration

of U, G, and M. From these results it

can be concluded that conditions can

be found that allow the glycosylation of

the biosimilar to fit within the origina-

tor specifications.

Conclusion

In the development of biosimilars, a com-

prehensive comparability exercise involv-

ing the originator product is required

to demonstrate similarity in terms of

physicochemical characteristics, efficacy,

and safety. In that respect, an enormous

weight is placed on analytics and the

analytical package for a biosimilar mAb

submission is considerably larger than

that of a stand-alone mAb. Structural

differences define the amount of pre-

clinical and clinical studies required. A

wide range of analytical tools providing

complimentary information is available

to guide biosimilar development.

Acknowledgments

The authors acknowledge Maureen

Joseph (Agilent Technologies, Wilming-

ton, Delaware), David Wong (Agilent

Technologies, Santa Clara, California)

and Lindsay Mesure (Promega, Leiden,

The Netherlands).

References

(1) K. Strebhardt and A. Ullrich, Nat. Rev.

Cancer 8, 473–480 (2008).

(2) L.M. Weiner, R. Surana, and S. Wang, Nat.

Rev. Immunol. 10, 317–327 (2010).

(3) G. Köhler and C. Milstein, Nature 256,

495–497 (1975).

(4) N.A.P.S. Buss, S.J. Henderson, M.

McFarlane, J.M. Shenton, and L. de Haan,

Curr. Opin. Pharmacol. 8, 620–626 (2008).

(5) J.G. Elvin, R.G. Couston, and C.F. van der

Walle, Int. J. Pharm. 440, 83–98 (2013).

(6) D.M. Ecker, S.D. Jones, and H.L. Levine,

mAbs 7, 9–14 (2015).

(7) G. Walsh, Nat. Biotechnol. 32, 992–1000

(2014).

(8) G. Walsh, Nat. Biotechnol. 28, 917–924

(2010).

(9) A. Beck and J.M. Reichert, mAbs 5, 621–

623 (2013).

(10) K. Sandra, I. Vandenheede, and P. Sandra,

J. Chromatogr. A 1335, 81–103 (2014).

(11) K. Sandra, I. Vandenheede, and P. Sandra,

LCGC Europe May Supplement, 10–16

(2013).

(12) A. Beck, S. Sanglier-Cianférani, and A.

Van Dorsselaer, Anal. Chem. 84, 4637–

4646 (2012).

(13) A. Beck, E. Wagner-Rousset, D. Ayoub, A.

Van Dorsselaer, and S. Sanglier-Cianférani,

Anal. Chem. 85, 715–736 (2013).

(14) E. Dumont, I. Vandenheede, P. Sandra, K.

Sandra, J. Martosella, P. Duong, M. Joseph,

Agilent Technologies Application Note

5991-5124EN (2014).

(15) E. Dumont, I. Vandenheede, P. Sandra, K.

Sandra, J. Martosella, P. Duong, M. Joseph,

Agilent Technologies Application Note

5991-5125EN (2014).

(16) E. Dumont, I. Vandenheede, P. Sandra, K.

Sandra, J. Martosella, P. Duong, M. Joseph,

Agilent Technologies Application Note

5991-5135EN (2014).

(17) www.gene.com

(18) A. Staub, D. Guillarme, J. Schappler, J.L.

Veuthey, and S. Rudaz, J. Pharm. Biomed.

Anal. 55, 810–822 (2011).

(19) S. Fekete, J.L. Veuthey, and D. Guillarme,

J. Pharm. Biomed. Anal. 69, 9–27 (2012).

(20) G. Chevreux, N. Tilly, and N. Bihoreau,

Anal. Biochem. 415, 212–214 (2011).

(21) C. Hosfield, P. Compton, L. Fornelli, P.

Thomas, N.L. Kelleher, M. Rosenblatt,

and M. Urh, Promega Poster Part#PS260

(2015).

(22) G. Vanhoenacker, I. Vandenheede, F.

David, P. Sandra, and K. Sandra, Anal.

Bioanal. Chem. 407, 355–366 (2015).

(23) I. Vandenheede, E. Dumont, P. Sandra, K.

Sandra, M. Joseph, Agilent Technologies

Application Note 5991-5273EN (2014).

(24) I. Vandenheede, E. Dumont, P. Sandra, K.

Sandra, M. Joseph, Agilent Technologies

Application Note 5991-5274EN (2014).

(25) M.J. Gramer, J.J Eckblad, R. Donahue, J.

Brown, C. Schultz, K. Vickerman, P. Priem,

E.T. van den Bremern J. Gerritsen, and P.H.

van Berkel, Biotechnol. Bioeng. 108, 1591–

1602 (2011).

Koen Sandra is Director at the

Research Institute for Chromatography

(RIC) in Kortrijk, Belgium.

Isabel Vandenheede is a Protein

Analyst at the Research Institute for Chro-

matography (RIC).

Emmie Dumont is an LC–MS Specialist

at the Research Institute for Chromatog-

raphy (RIC).

Pat Sandra is Chairman at the Re-

search Institute for Chromatography (RIC)

and Emeritus Professor at Ghent Univer-

sity in Ghent, Belgium. ◾

Page 22: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

22 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Christian G. Huber

Higher Order Mass Spectrometry Techniques Applied to Biopharmaceuticals

The recent trends in mass spectrometric techniques—including native

mass spectrometry (MS), ion mobility spectrometry (IMS), hydrogen–

deuterium exchange MS (HDX MS), and chemical cross-linking MS

(CXMS)—used to elucidate higher-order structures of protein complexes

and the practical implementations of these methods are discussed.

Since its invention in the early

20th century (1), mass spec-

trometry (MS) has been used

to discover new chemical elements

and their isotopes (2), explore mar-

tian soil for organic matter (3), and

study biological processes by profiling

proteomes or metabolomes (4,5). Bio-

polymers, and especially proteins, are

the subject of intensive investigation.

They are characterized by different

levels of structural organization, rang-

ing from the primary structure repre-

sented by the amino acid sequence,

over secondary structural elements

such as α-helices and β-sheets, and

the three-dimensional (3D) orienta-

tion of the polypeptide chain, and

f inally to the assembly of subunits

into protein complexes.

Analysis of protein structure using

MS was first possible in the mid-1980s,

when the soft ionization techniques of

electrospray ionization (ESI) (6) and

matrix-assisted laser desorption–ion-

ization (MALDI) (7) were introduced.

Implementation of MS for protein

analysis initia lly focused on large-

scale identification (8) and the deter-

mination or confirmation of primary

structure (9), whereas newer technol-

ogies have laid the ground work for

the study of tertiary and even higher

order structures of protein molecules

and complexes.

The analysis of biopharmaceuticals

(therapeutic proteins developed for

disease treatment) requires analytical

techniques that are able to elucidate

the various structural levels to ensure

their eff icacy and safety in patients.

Several MS techniques are indispens-

able in the toolbox of physicochemi-

cal characterization methods available

for the analysis of therapeutic proteins

(10). Some of the methods used in

the elucidation of higher order struc-

tural elements of proteins—including

native MS, ion mobility MS, hydro-

gen–deuterium exchange MS, and

chemical cross-lining MS—are dis-

cussed in this article.

Native Mass Spectrometry

Experiments using electrospray ion-

ization mass spectrometry (ESI–MS)

for the analysis of intact proteins were

first performed by J.B. Fenn’s group

(6). The study used strongly dena-

turing conditions created by using

50–90% organic solvent contain-

ing acetic acid or trif luoroacetic acid

and was beneficial for the detection

of multiple-charge protein ions with

a quadrupole mass spectrometer with

an upper nominal mass limit of m/z

1500. However, it was soon discovered

that the charge-state distribution of

electrosprayed proteins was signif i-

cantly inf luenced by the protein struc-

Ph

oto

cre

dit

: D

ou

g A

rman

d/G

etty

Im

ages

Page 23: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 23www.chromatographyonline.com

ture prevalent under nondenaturing or

denaturing conditions (11). This dis-

covery then led to the implementation

of native MS.

Native MS aims to maintain the

3D structure of proteins or protein

complexes as much as possible during

an experiment (12) by using condi-

tions that ref lect the protein’s native

environment. The overall charge of a

protein ion is limited by the number

of ionizable functionalities that are

accessible on the surface for charg-

ing, predominantly through proton-

ation or deprotonation, leading to the

observation of low-charged species in

mass spectra. Weakly bound, nonco-

valent complexes—including proteins

interacting with inhibitors, cofactors,

metal ions, carbohydrates, or pep-

tides—can be preserved during the

electrospray process facilitating the

study of the structure, stoichiometry,

and association constant of such bio-

molecular complexes (13). The reduc-

tion of charge requires the use of mass

spectrometers with an extended mass

range such as time-of-f light (TOF)

(14), or more recently orbital ion trap

(15) mass analyzers to detect the low-

charged protein species.

Trastuzumab (INN; trade name

Herceptin) is a monoclonal antibody

that interferes with the human epider-

mal growth factor receptor 2 (HER2)

and is used to treat HER2-positive

breast cancers. Figures 1a and 1c

illustrate the difference between mass

spectra of trastuzumab when analyzed

under denaturing (Figure 1a) versus

nondenaturing conditions (Figure 1c).

Under denaturing conditions (Figure

1a), charge states from 33+ to 60+ are

detected in an m/z range of 2200–4500,

whereas nondenaturing conditions

(Figure 1c) yield charge states of 22+

to 28+ at m/z 5200–6700. Deconvolu-

tion of both mass spectra gives equiva-

lent masses for the uncharged species

with masses in the range of 147–148

kDa and also reveals several different

protein species that represent the dif-

ferent glycoforms of the monoclonal

antibody. Such analysis can therefore

readily reveal the glycosylation pat-

tern of the protein, a highly important

quality parameter of recombinant bio-

pharmaceuticals.

Native MS has been shown to be a

highly efficient tool for determining

binding stoichiometry of a monoclonal

antibody with its antigen. Humanized

murine monoclonal antibody (hzmAb),

directed against the junctional adhe-

sion molecule A (JAM-A) to have anti-

proliferative and antitumoral prop-

erties, was titrated with its antigen

and then analyzed using native MS

to reveal noncovalent complex stoi-

chiometries (17). Three species were

detected when equimolar amounts of

antibody and its target antigen were

incubated including free antibody, 1:1,

and 1:2 antibody–antigen complex.

bc

a

1000

20

00

40

00

60

00

80

00

40

00

2000 3000 4000

3000 4000 5000 6000 7000 8000

m/z

m/z

5 10

Drift Time (millisec)

15 20 25

5 10

Drift Time (millisec)

15 20 25 30

d

e

(a) (b)

(c) (d)

45+

42+

23+27+

25+

ESI -sourceIon

guide Quadrupole TWIMS cell

Trap Transfer

Pusher Detector

Reflectron

Figure 1: MS and IMS analysis of intact trastuzumab. (a) and (c): Intact MS analy-sis of trastuzumab. ESI-TOF mass spectra of trastuzumab in denaturing (a) or na-tive (c) conditions. The inserts shows an extended view of the 44+ (a) and 25+ (c) charge states with resolution of the different glycoforms: (a) 147 917.1 ± 1.1 Da (G0/G0F), (b) 148 061.7 ± 0.8 Da (G0F/G0F), (c) 148 222.4 ± 0.9 Da (G0F/G1F), (d) 148 383.8 ± 0.8 Da (G1F/G1F), and (e) 148 544.3 ± 1.0 Da (G1F/G2F). (b) and (d): IMS analysis of trastuzumab. Ion mobility mobilograms of trastuzumab in de-naturing (b) or native (d) conditions. IMS data obtained in native conditions (d) reveal small amounts of dimeric mAb. Adapted and reproduced with permission from reference 16, ©American Chemical Society.

Figure 2: Schematic diagram of a quadrupole-traveling wave ion mobility–time of flight instrument.

Page 24: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

24 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Two molar equivalents of antigen led

to an almost quantitative formation of

the 1:2 complex, while eightfold molar

excess yielded a 1:4 complex with a

small portion of 1:3 complex.

Ion Mobility Spectrometry

Developed in the 1960s, ion mobil-

ity spectrometry (IMS) enables the

generation of size- and conformation-

dependent information that is not pos-

sible using MS alone. When coupled

to MS this technique has the potential

to separate isomers, isobars, and con-

formers; significantly reduce chemical

background; and detect aggregates of

biopharmaceuticals. IMS separation

of ions is possible using differing sepa-

ration powers, analyte detection, and

hyphenation to mass spectrometry

(18), including drift-time ion mobil-

ity spectrometry (DTIMS); aspiration

ion mobility spectrometry (AIMS);

field-asymmetric waveform ion mobil-

ity spectrometry (FAIMS); and trav-

eling-wave ion mobility spectrometry

(TWIMS). DTIMS and TWIMS are

the two principles most often used in

commercial instruments. In a DTIMS

device ions are moved through a uni-

form, linear-field drift tube filled with

a so-called “buffer gas” through a small,

uniform electric f ield. The moving

ions are attenuated by collisions with

the buffer gas depending on their over-

all charge and collision cross-section

(18). Ions with multiple charges and a

small cross-sectional area move faster

through the drift cell than low-charge

ions with large collisional cross-sec-

tions. TWIMS is a type of IMS that

utilizes traveling waves created by a

series of ring-shaped electrodes that

split the structure of the drift cell into

a series of segments (Figure 2). Instead

of a uniform linear field, a high field

is applied to one segment of the cell

that is subsequently swept through the

cell in the direction of ion migration.

Consequently, movement and sepa-

ration of ions in the mobility cell is

accomplished by means of pulses of an

electric field passing through.

An example for the appl icat ion

of T W I M S t o t he a n a l y s i s of

monoclonal antibodies is illustrated

in Figures 1b and 1d. IMS analysis

of t ra stuzumab under denaturing

condit ions revea ls a la rge number

of highly charged species clustering

at dri f t t imes between 10 and 15

ms, while native conditions clearly

dist inguish between the dif ferent,

low-charged species in a drift time

range of 7–25 ms. This contrast in

drift behavior is advantageous for the

analysis of more complex mixtures

of biopharmaceutica ls, particularly

when looking at sequence variants

o r o t h e r p o s t - t r a n s l a t i o n a l

modif ications such as ox idation or

pyroglutamate formation.

A n o t h e r p r a c t i c a l e x a m p l e

o f I M S c h a r a c t e r i z a t i o n o f

biopharmaceut ica l s ( les s directed

towards higher order elucidation) is

outlined in Figure 3. Here, a reduced

mouse monoclonal antibody (IgG1,

κ) sample comprising of both heavy

and l ight cha ins was int roduced

into an ESI-quadrupole-IMS-TOF

system (19). The two-dimensiona l

(2D) ion mobilogram-mass spectrum

depicted in Figure 3a clearly shows

that light and heavy chains can be

readily separated as different species

without any other upfront separation

technique. Multiple charged species

related to the light and heavy chains

were d i f f e r ent i a t ed u s i ng m a s s

spectra extracted from the encircled

areas in Figure 3a without mutually

interfering signals. Extracted mass

spectra (Figures 3b and 3d) were

deconvoluted using a ma x imum

entropy algorithm, yielding spectra

Light Chain

Heavy Chain

2000

1500

1000

700

10026+

(b)

(a)

(d)

(e)(c)

28+ 23+

20+

17+

52+

45+

41+

36+

800 1200 1600 2000 2400 28000

%

100

024050 24150 24250 24350 24450 49600 49800 50000 50200 50500

massmass

50249

49779

49924

50086

** *

*

** *

* *

2422624177

24199

%

100

0

%

100

800 1200 1600 2000 2400 2800

m/zm/z

0

%

3.2 6.4

Drift Time (millisec)

m/z

9.6 12.8

Figure 3: On-line LC–MS analysis of a completely reduced IgG1 antibody using ion mobility-TOF mode. (a) Two-dimensional plot of ion drift time versus m/z for the reduced IgG1 obtained using the ion mobility separation (7.5 V pulse). (b) Combined raw mass spectrum of the light chain. (c) Deconvoluted mass spec-trum of the light chain. (d) Combined raw mass spectrum of the heavy chain. (e) Deconvoluted mass spectrum of the heavy chain. Adapted and reproduced with permission from reference 19, ©John Wiley and Sons.

Page 25: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 25www.chromatographyonline.com

of uncharged species, as i l lustrated

in Figures 3c and 3e. As expected,

the light chain is detected as a single

species, while the spectrum of the

heavy chain revea ls at lea st three

glycoforms, characterized by different

ga lactose content in the at tached

N-glycan (162 Da mass difference).

This example nicely demonstrates the

benefits of an additional dimension

of separation, although an upfront

sepa rat ion method such a s high

performance liquid chromatography

(HPLC) or capillary electrophoresis

(CE) may be necessary—especia l ly

for the quantitative analysis of trace

amounts of impurities that is often

indispensable for quality control.

Hydrogen–Deuterium

Exchange Mass Spectrometry

The native state of proteins is gener-

ally characterized by a tightly folded,

compact structure that exposes a

well-def ined surface to its environ-

ment. Denaturation under conditions

such as high temperature, extreme pH,

adsorption to surfaces, or dissolution

in organic solvent results in the protein

unfolding and forming a significantly

different surface. Similarly, interac-

tions of a protein with other molecules

such as small drugs, nucleic acids, or

other proteins can lead to a significant

change in surface properties.

Denaturing and complex formation

can also have a profound inf luence on

the exchangeability of protons at the

surface of a protein molecule—the

acidic protons of the carboxyl groups

or acidic side chains of aspartate and

glutamate are normally the most eas-

ily and rapidly exchanged while the

amide protons of the protein back-

bone are much less prone to exchange.

Exchange can be monitored by dissolu-

tion of a protein in heavy water (D2O),

which leads to a hydrogen exchange by

deuterium in a few minutes to hours.

In proteins, the exchange rates for

the different hydrogen atoms strongly

depend on accessibility and therefore

on protein conformation or associa-

tion into higher order structures. The

substitution of exchangeable hydrogen

atoms with deuterium atoms forms the

basis of hydrogen–deuterium exchange

mass spectrometry (HDX-MS) (20).

A schematic workf low of HDX-MS

is depicted in Figure 4. In brief, a

protein with exchangeable hydrogens

is dissolved for different periods of

time at ambient or elevated incuba-

tion temperature (20–40 °C) in deu-

terated water (buffered to pH around

7). Depending on exchangeability,

hydrogen atoms are replaced by deu-

terium atoms during the incubation

time, before the exchange is quenched

upon acidif ication and cooling to

0 °C. Proteins are then digested under

quenching conditions, and the result-

ing peptides are separated by low-tem-

perature HPLC, and finally analyzed

by tandem mass spectrometry (MS/

MS) upon fragmentation either by col-

lision-induced dissociation (CID) or

electron-transfer dissociation (ETD).

Characteristic mass shifts in the frag-

ment ions are indicative for the pres-

ence and positions of deuterium atoms.

Analysis of the kinetics of deuterium

uptake yields information about the

accessibility of hydrogens at different

positions in the protein, which allows

valuable insights into the 3D structure

of proteins or protein complexes.

HDX-MS has been successfully used

to compare 3D structures of biophar-

maceuticals, which is essential to dem-

onstrate manufacturing consistency to

regulatory agencies or provide a proof

of structural equivalence between an

originator biopharmaceutica l and

its biosimilar. The advantage of this

approach is that it probes the whole

molecule instead of just certain sub-

structures. The results of an interroga-

tion of the 3D structure of interferon-

β-1a, a 20-kDa cytokine used to treat

multiple sclerosis (traded under the

names Avonex (Biogen), Rebif (Merck

Serono or Pfizer), or CinnoVex (Cin-

naGen) as a biosimilar, are shown in

Figure 5 (22). Hydrogen exchange

rates determined for f ive different

peptic peptides effectively show the

impact of different manufacturing

conditions as well as post-translational

modif ications—modif ication with

poly(ethylene glycol) (PEG); or oxi-

dation at C17, M1, M36, M62, and

M117—on protein structure.

No s i g n i f i c a n t a l t e r a t i on i n

h y d r o g e n – d e u t e r iu m e x c h a n g e

prof i le was observed, even though

p r o d u c t i o n i n v o l v e d d i f f e r e n t

batches, using different cell media,

and was subjected to N-termina l

modification with PEG. A significant

i mpa c t w a s howe ve r f ou nd f o r

methionine or cysteine ox idat ion,

and because the ox idized peptides

i n c o r p o r a t e d m o r e d e u t e r i u m

compared to the reference analogues,

it could be concluded that oxidized

i nte r f e ron-β-1a i s more so lvent

exposed and less hydrogen bonded.

H HH H

Z7

t1

H/D exchange

Quench

(pH 2.5, 0oC )

(pepsin, pH 2.5, 0oC )

Cooled

LC-MS

Time

co

nte

nt

Time

(protein)

Gas-phase

Solution-phase

cleavage

cleavage

(peptide)

Gas-phase

cleavage

D2O

t2

t3

t4

C7

C6

C5

C4

C3

C2

C1

R1

N

O

NN

NN

NN

NOH

OOOO

O O O

2

R3

R5

R7

R8

R6

R4

R2

Z6

Z5

Z4

Z3

Z2

Z1

H

H

H

H

H

HH

H

HH

H

H

H

H

HH

H

H

H

H

HH

H H

HH

H H

H

H

HH H

H

H

D

DD

D

DD

D

D

DDD

D

D

D D

D

D

H

HH

H

H

H

H H

H

H

HH H

H

H

D

DD

D

DD

D

D

DDD

D

D

co

nte

nt

D

D

D

H

HH

H

H

H

HH

H

H

HH H

H

H

D

DD D

D D

D

D

DDD

D

D

D

H

HH

H

H

H

Figure 4: Principle of hydrogen/deuterium exchange mass spectrometry. Adapted and reproduced with permission from reference 21, ©American Chemical Society.

Page 26: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

26 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Although this example convinc-

ingly demonstrated the applicability of

HDX-MS for revealing structural dif-

ferences in homogenous biopharma-

ceuticals, the authors also pointed out

that it is not capable, at the moment,

to detect conformational differences

in coexisting, low-level (<10%) com-

ponents of the population (22).

Chemical Cross-Linking

Mass Spectrometry

Three-dimensional structures of pro-

teins can be determined with atomic

resolution by using high-resolution

methods such as X-ray crystallography

or nuclear magnetic resonance (NMR)

spectroscopy, but the high amount

of sample required for these meth-

ods (typically in the milligram range)

makes them impractical for biological

studies. In comparison, low-resolution

structural data generated by chemical

cross-linking MS (CXMS) uses much

less sample amounts (in the nanogram

to picogram range) to generate highly

valuable data (23). Low-resolution

structural information is obtained by

chemically cross-linking functional

groups in a protein by means of a

bifunctional cross-linker, which gives

information about the distance of the

cross-linked functional groups in a

protein molecule or a protein complex.

The most common functional groups

available for cross-linking in proteins

are the lysine amino groups. Sulfydryl

groups of cysteines are another possi-

bility, but they can become involved

in the 3D structure of a protein par-

ticularly when created by reduction of

disulfide-bridges in the native protein.

Although formaldehyde is the oldest

cross-linking reagent, the most com-

monly utilized reagents are based on

bifunctional N-hydroxy-succinimide

esters, which readily react with free

amino groups (and in a side reaction

also with hydroxyl groups of tyrosine)

to create a stable amide or imide bond

upon release of N-hydroxysuccinimide.

Depending on the length of the cross-

linking spacer, different distances of

amino acids can be probed, ranging

from (almost) zero for formaldehyde to

6.4 Å for disulfosuccinimidyl tartrate,

11.4 Å for bis(sulfosuccinimidyl)suber-

ate (10 atoms), and 16.1 Å for ethylene

glycol bis(sulfosuccinimidyl succinate

(14 atoms) (24).

Figure 6 shows an outline of a cross-

linking experiment. After the forma-

tion of intramolecular or intermolecu-

lar cross-links, the protein or protein

complex is proteolytically digested

and the resulting peptides are ana-

lyzed via HPLC–MS/MS. Because the

crosslink remains unaffected by the

proteolysis, cross-linked amino acids

are revealed through the correspond-

ing cross-linked peptides. To more

easily identify cross-linked products,

isotope-labeled cross-linking reagents

with 50% heavy isotope exchange can

be used. Thus, cross-linked peptides

are recognized by 1:1 doublets of mass

signals for the light and heavy ver-

sions, which is usually achieved with

the help of computer-based searching

algorithms (25). The distance infor-

mation obtained from the cross-link-

ing experiment is then used to build

and verify structural models for pro-

teins or protein complexes.

Chemical cross-linking can also

be utilized to directly analyze stabi-

lized protein complexes. For example,

disuccinimidyl suberate, as well as

1,1′-(suberoyldioxyl)bisazabenzotri-

azole) were used as cross-linkers to

stabilize the complex between the

bovine prion protein and a specif ic

1. (8 - 15)FLQRSSNF

76543210

0 1 10 100 1000

5

4

3

2

1

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

5678

43210

0 1 10 100 1000

76543210

0 1 10 100 1000

5

4

3

2

1

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

5678

43210

0 1 10 100 1000

76543210

0 1 10 100 1000

5

4

3

2

1

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

5678

43210

0 1 10 100 1000

76543210

0 1 10 100 1000

5

4

3

2

1

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

12

10

8

6

4

2

00 1 10 100 1000

5678

43210

0 1 10 100 1000

Rela

tive a

bu

nd

an

ce (

Da)

Rela

tive a

bu

nd

an

ce (

Da)

Rela

tive a

bu

nd

an

ce (

Da)

Rela

tive a

bu

nd

an

ce (

Da)

2. (62 - 67)MLQNIF

3. (88 - 101)LANVYHQINHLKTV

4. (121 - 134)HLKRYYGRILHYLK

5. (154 - 162)FYFINRLTG

(a)Differentbatches

(c)Different

media

(b)Pegylated

(d)Oxidized

1. (8- 15)

Time (min) Time (min) Time (min) Time (min) Time (min)

3. (88- 101)

5. (154- 162)2. (62- 67)

4. (121- 134)

Figure 5: Deuterium incorporation graphs generated for five interferon-β-1a (IFN) peptic peptides from four different HDX MS comparability experiments. In each graph, the reference IFN data are the black lines with closed triangles, while the experimental IFN data, to which it is being compared, is the red line with open circles. Row a: Comparison of two different large scale IFN batches prepared over eight years apart; row b: Comparison of IFN versus N-terminally PEGylated IFN; row c: Comparison of IFN produced using different cell culture media and growth conditions; row d: Comparison of IFN versus oxidized IFN (oxi-dation of Met and Cys residues C17, M1, M36, M62, and M117 was 100%). Adapt-ed and reproduced with permission from reference 22, ©John Wiley and Sons.

Page 27: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 27www.chromatographyonline.com

antibody against it, the antibody

3E7 (26). Direct analysis of the reac-

tion products by matrix-assisted laser

desorption–ionization MS revealed

both the free prion protein and the

free antibody together with 1:1 and

1:2 antibody–prion protein complexes.

Conclusions

In conclusion, MS, tradit iona l ly

regarded as one of the most important

analytical methods for the determina-

tion of the primary structure of pro-

teins, is increasingly contributing to

the elucidation of diverse fundamental

aspects of the tertiary and even quater-

nary structure of proteins and protein

complexes. In spite of providing less

spatial resolution, the major strength

of MS-based investigations compared

to NMR spectroscopy or X-ray crys-

tallography lies within the compara-

tively low amounts of sample required

for successful analysis, typically a few

picograms to nanograms. Such stud-

ies are, however, only feasible with

substantial support through elabo-

rate computational algorithms and

workf lows, which requires significant

involvement of bioinformatics into

data evaluation.

Acknowledgments

The financial support by the Austrian

Federal Ministry of Economy, Family,

and Youth, the National Foundation

of Research, Technology, and Devel-

opment, and by a Start-up Grant of

the State of Salzburg is gratefully

acknowledged.

References

(1) J.J. Thomson, Proceedings of the Royal

Society of London Series a-Containing

Papers of a Mathematical and Physical

Character 89, 1–20 (1913).

(2) F.W. Aston, Nature 105, 547–547, (1920).

(3) K. Biemann, Proc. Natl. Acad. Sci. U.S.A.

104, 10310–10313 (2007).

(4) R. Aebersold and M. Mann, Nature 422,

198–207 (2003).

(5) O. Fiehn, Plant Mol. Biol. 48, 155–171

(2002).

(6) C.K. Meng, M. Mann, and J.B. Fenn, Z .

Phys. 10, 361–368 (1988).

(7) M. Karas and F. Hi l lenkamp, Anal .

Chem. 60, 2299–2301 (1988).

(8) J.K . Eng, A.L. McCormack, and J.R .I.

Yates, J. Am. Soc. Mass Spectrom. 5, 976–

989 (1994).

(9) H. Nau and K . Biemann, Abstracts of

Papers of the American Chemical Society

62–62 (1974).

(10) R.J. Falconer, D. Jackson-Matthews, and

S.M. Mahler, J. Chem. Technol. Biotech-

nol. 86, 915–922 (2011).

(11) J.A. Loo, H.R. Udseth, and R.D. Smith,

Biomed. Environ. Mass Spectrom. 17, 411–

414 (1988).

(12) M. Przybylski and M.O. Glocker, Angew.

Chem. Int. Ed. 35, 806–826 (1996).

(13) J.A. Loo, Int. J. Mass spectrom. 200, 175–

186 (2000).

(14) M.C . Fitzgerald, I. Chernushevich, K .

G. Standing, C. P. Whitman, and S. B.

Kent, Proc. Natl. Acad. Sci. U.S.A . 93,

6851–6856 (1996).

(15) S. Rosati, R .J. Rose, N.J. Thompson,

E . van Duijn, E . Damoc, E . Denisov,

A. Makarov, and A. J. R. Heck, Angew.

Chem.-Int. Ed. 51, 12992–12996 (2012).

(16) A. Beck, S. Sanglier-Cianferani, and A.

Van Dorsselaer, Anal. Chem. 84, 4637–

4646 (2012).

(17) C . Atmanene, E . Wagner-Rousset, M.

Malissard, B. Chol, A. Robert, N. Cor-

vaia, A. Van Dorsselaer, A. Beck, and

S. Sanglier-Cianferani, Anal. Chem. 81,

6364–6373 (2009).

(18) A.B. Kanu, P. Dwivedi, M. Tam, L. Matz,

and H.H. Hill, J. Mass Spectrom. 43,

1–22 (2008).

(19) P. Olivova, W. Chen, A.B. Chakraborty,

and J.C. Gebler, Rapid Commun. Mass

Spectrom. 22, 29–40 (2008).

(20) V. Katta and B.T. Chait, J. Amer. Chem.

Soc. 115, 6317–6321 (1993).

(21) K .D. Rand, M. Zehl, and T.J.D. Jor-

gensen, Acc. Chem. Res. 47, 3018–3027

(2014).

(22) D. Houde, S.A. Berkowitz, and J.R .

Engen, J. Pharm. Sci. 100, 2071–2086

(2011).

(23) A. Sinz, Mass Spectrom. Rev. 25, 663–682

(2006).

(24) A. Sinz, J. Mass Spectrom. 38, 1225–1237

(2003).

(25) A. Leitner, T. Walzthoeni, and R. Aeber-

sold, Nature Protocols 9, 120–137 (2014).

(26) C. Bich, S. Maedler, K. Chiesa, F. DeGia-

como, N. Bogliotti, and R. Zenobi, Anal.

Chem. 82, 172–179 (2010).

(27) h t t p : //d a l t o n l a b . i q m .u n i c a m p . b r /

research.html.

Christian Huber was educated as

an analytical chemist from 1985 to 1993

at the University of Innsbruck, Austria.

Following a lecturing qualification at

the University of Innsbruck in 1997, he

held the chair of analytical chemistry

position at Saarland University in

Germany from 2002 to 2008. In 2008,

he was made a professor of chemistry

for biosciences at the Department of

Molecular Biology of the University of

Salzburg, Austria. His research interests

include bioanalytical chemistry and

proteome and metabolome analysis, as

well as in-depth characterization of

therapeutic proteins. ◾

Proteolysis

LC-MS/MS

Cross-linking

Map of cross-links

Intra and inter-chainSelection of structural models cross-links

Set of distance restraints

Protein complexProtein 1

Protein 2

Protein 3

Figure 6: Schematic of a workflow for chemical cross-linking of protein complexes followed by digestion and analysis by HPLC–MS/MS. Reproduced with permission from reference 27.

Page 28: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

28 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

Nico C. van de Merbel

Advances in Liquid Chromatography–Tandem Mass Spectrometry (LC–MS/MS)-Based Quantitation of Biopharmaceuticals in Biological Samples

Liquid chromatography coupled to tandem mass spectrometry

(LC–MS/MS) has recently become a more popular alternative to

traditional ligand-binding assays for the quantitative determination

of biopharmaceuticals. LC–MS/MS offers several advantages such

as improved accuracy and precision, better selectivity, and generic

applicability without the need for raising analyte-directed antibodies.

Here we discuss the technical requirements for a successful LC–MS/MS

method for the quantitation of biopharmaceuticals and evaluate the

advantages and disadvantages compared to ligand-binding assays.

The development of protein-based

pharmaceuticals, or biopharma-

ceuticals, is by far the fastest

growing part of the pharmaceutical

industry today. With more than 1500

biopharmaceuticals in clinical devel-

opment and more companies shifting

their research and development (R&D)

efforts towards this sophisticated and

relatively profitable class of drugs, the

pharmaceutical landscape has changed

beyond recognition compared to 20 or

even 10 years ago. As a result, the field

of bioanalysis that supports drug devel-

opment by measuring the concentra-

tions of drugs or relevant endogenous

molecules in biological samples has also

seen many changes. The quantitative

determination of biopharmaceuticals

has traditionally been the domain of

ligand-binding assays, such as enzyme-

linked immunosorbent assay (ELISA).

However, in the past few years there

has been a clear increase in the applica-

tion of alternative analytical platforms,

in particular liquid chromatography

coupled to tandem mass spectrometry

(LC–MS/MS), which has been the

workhorse for small-molecule bioanaly-

sis for more than 20 years (1–5).

Over the past decade, there have

been many advances in the LC–MS/

MS-based quantitation of biopharma-

ceuticals, both from an analytical and

a conceptual point of view. In this

article, an overview is given of the

many aspects of this field of analytical

research by reference to a selection of

recent applications.

Protein Digestion

MS/MS remains the detection tech-

nique of choice for the quantitative

determination of biopharmaceuticals

because of its sensitivity and wide-

spread availability in the pharmaceu-

tical and related industries. However,

the use of LC–MS/MS to quantify

biopharmaceuticals is more complex

than for small molecules because it

is not directly compatible with mol-

ecules with a mass above around

5000 Da. The ions of larger analytes

are distributed over many dif fer-

ent charge states and usually do not

readily fragment, which considerably

reduces sensitivity.

Therefore, a typical step in the

analysis is the (enzymatic) digestion

of a biopharmaceutical into a mixture

of smaller peptides, followed by the

analysis of the digest and quantitation

of one or more so-called signature pep-

tides as a measure for the intact pro-

Ph

oto

cre

dit

: G

I p

ho

toS

tock

/Get

tyIm

ages

Page 29: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

www.chromatographyonline.com

tein. Digestion is usually performed using the enzyme tryp-

sin, which cleaves the amino acids chain in proteins after a

lysine or arginine. Trypsin is popular because it is readily

available at a reasonable price and can cleave proteins into

peptides of a size (500–2000 Da) that is well suited for MS/

MS detection.

Protein digestion enormously increases the complexity of a

biological sample. Matrices such as plasma contain proteins

at a total concentration of around 80 mg/mL and, when no

further cleanup of the sample is performed, each of these

proteins is cleaved into a series of peptides that are all of a

similar size and have more or less comparable physicochemi-

cal and analytical properties. Therefore, it is often challeng-

ing to detect low concentrations of a signature peptide in

a digest, because of the presence of so many endogenous

peptides, which all consist of combinations of the same 20

amino acids and often occur at much higher levels than the

signature peptide itself.

Despite the selective nature of MS/MS detection, chro-

matograms of digested biological samples often contain

many background peaks originating from endogenous pep-

tides that show a response at the mass transition of the sig-

nature peptide. Figure 1 shows this effect for a fixed con-

centration of digested salmon calcitonin in the presence of

increasing amounts of digested plasma (6). The selectivity of

the method is clearly affected by the presence of endogenous

background peptides. As a result, method sensitivity is also

heavily impacted—in this case the achievable lower limit of

quantitation (LLOQ ) increases 100-fold, from 0.2  ng/mL

(60 pM) in the absence of matrix peptides to 20 ng/mL (6

nM) in the presence of 50% of digested plasma.

A review of current literature (1,4) shows that a typical

LLOQ for a biopharmaceutical in plasma or serum, only

treated by digestion, is in the high nanograms-per-milliliter

to low micrograms-per-milliliter range (corresponding to

low nanomolar levels for many proteins). Figure 2 shows an

example chromatogram for a signature peptide of recombi-

nant human α-glucosidase at its LLOQ of 0.5 μg/mL (5 nM)

in human plasma (7).

Signature Peptide Selection

The possibilities for selecting a proper signature peptide

are usually rather limited. First and foremost, it is essential

that the selected signature peptide has a unique amino acid

sequence that does not naturally occur in any of the endog-

enous matrix proteins. Selection of a non-unique signature

peptide results in an overestimation of analyte concentra-

tions, because the same amino acid sequence that is released

from endogenous proteins would contribute to the overall

signal. This often disqualifies a large number of the theo-

retical signature peptides, particularly for biopharmaceuti-

cals with a high degree of similarity to endogenous proteins,

such as humanized antibodies (if these need to be quantified

in human plasma). In addition, other criteria are applied to

ensure robustness of the LC–MS/MS assay. Peptides con-

taining unstable amino acids, such as methionine and tryp-

tophan that can be oxidized, or glutamine and asparagine

that can be deamidated, are usually disregarded to avoid

ANYTHING BUT TRIVIAL

Things that appear clean...

Things that are guaranteed clean.

CL

ASS

10K

CLEANROOM

PA

CK

ED

GUARANTEED

Are all clean vials the same? Level of cleanliness

IS�NOT�TRIVIAL��%VERY�-ICRO,ITER�CERTIlED�VIAL�IS������

CLEANED�AND�PACKED�IN�A�#LASS��������#LEANROOM�

to ensure consistency of your results.

'ET�A�SAMPLE�IN�YOUR�LAB�BY�VISITING� go.wheaton.com/microliter-samples

Page 30: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

30 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

losses during analysis, although forced

oxidation of a signature peptide to a sta-

ble oxidized product has been success-

fully used (8). Similarly, peptides with

(variable) post-translational modifica-

tions—such as O- or N-glycosylated

amino acids—are typically excluded

because these would introduce unde-

sirable heterogeneity. In addition, pep-

tides that are too small, too large, too

polar, or too hydrophobic might cause

analytical problems because of adsorp-

tion, sub-optimal chromatographic

behavior, or limited selectivity and sen-

sitivity. In the end, there may be just a

few out of the many potential signature

peptides that can be successfully used

in practice.

Protein Extraction

An obvious way to improve selectiv-

ity and sensitivity of an LC–MS/MS

method is to remove interfering matrix

proteins before digestion, which can

be achieved by applying immunocap-

ture (IC) techniques. Magnetic beads

or other resins are coated with a pro-

tein that displays a high binding affin-

100

0

1.75 2.00 2.25 2.50

2.94

0%

1%

5%

10%

20%

50%

2.93

1.84 2.644.24

3.723.583.22

2.922.64

2.50

2.332.162.041.76

2.162.33

2.50

2.54

2.91

3.23 3.74 4.23 4.54 4.86 5.055.44

5.40

5.754.944.514.384.20

3.723.54

3.21

2.902.812.47

2.302.14

2.031.76

2.002.22

2.74

3.043.19

3.453.66 4.20 4.41

4.72

5.24

1.76 1.932.04

5.454.874.554.24

2.75 3.00 3.25 3.50 3.75 4.00 4.25 4.50 4.75 5.00 5.25 5.50 5.75 6.0

1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50 3.75 4.00 4.25 4.50 4.75 5.00 5.25 5.50 5.75 6.0

1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50 3.75 4.00 4.25 4.50 4.75 5.00 5.25 5.50 5.75 6.0

1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50 3.75 4.00 4.25 4.50 4.75 5.00 5.25 5.50 5.75 6.0

1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50 3.75 4.00 4.25 4.50 4.75 5.00 5.25 5.50 5.75 6.0

1.75 2.00 2.25 2.50 2.75 3.00 3.25 3.50

Time (min)

Time (min)

Time (min)

Time (min)

Time (min)

Time (min)

3.75 4.00 4.25 4.50 4.75 5.00 5.25 5.50 5.75 6.0

%

100

0

%

100

0

%

100

0

%

100

0

%

100

0

%

Figure 1: LC–MS/MS (m/z 561.9 to m/z 204.0) chromatograms of a signature peptide of 2 ng/mL salmon calcitonin in samples containing increasing amounts of human plasma digest. Analyte peak at 2.9 min. Adapted and reproduced with permission from reference 6, ©American Chemical Society.

Page 31: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 31www.chromatographyonline.com

2.94

1000

800

600

400

200

Inte

nsi

ty, cp

sIn

ten

sity

, cp

s

0

1000(b)

(a)

800

600

400

200

0

0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0

0.5 1.0 1.5 2.0

Time (min)

Time (min)

2.5 3.0 3.5 4.0 4.5 5.0

1.04

1.04

2.96

3.20

3.43 4.054.12

1.441.52 1.62 2.50 2.64

3.173.40

3.49 3.69

4.014.11

Figure 2: LC–MS/MS (m/z 616.1 to m/z 1030.7) chromatogram of the signature pep-tide of recombinant human α-glucosidase in human plasma; (a) blank and (b) 0.5 μg/mL, pretreated with digestion only.

ity towards the analyte, typically an

antibody raised against the analyte or

the pharmacological target to which a

biopharmaceutical is directed. By mix-

ing the sample with a suspension of

the beads or passing it through a car-

tridge filled with the resin, the analyte

is selectively isolated from the complex

sample. This approach is particularly

popular for the quantitation of endog-

enous proteins such as biomarkers, for

which well-characterized immunologi-

cal reagents are widely available.

One example is an LC–MS/MS

method for parathyroid hormone

(PTH) in human serum (9). A sample

of 1 mL was treated by IC with poly-

styrene beads coated with murine

anti-PTH antibodies and the trapped

analyte digested with trypsin. The IC

treatment allowed the quantitation

of PTH down to 40 pg/mL (4 pM)

in serum, which shows the enormous

clean-up potential of this approach.

A completely 15N-labeled form of

PTH was added to the sample as an

internal standard at the very begin-

ning of the sample handling proce-

dure. In general, it is desirable that a

stable-isotope-labeled or other closely

related form of the protein analyte be

included in the method as an internal

standard, to correct for the variability

of the extraction procedure. This is

also one of the drawbacks of extract-

ing a biological sample before digestion,

because such a protein-based internal

standard can usually only be obtained

by biotechnological means, which may

be difficult, if not impossible (4).

The disadvantages associated with the

use of immunological reagents—such

as their potentially limited availability,

varying quality, and the interference

of matrix proteins with the extraction

efficiency—have prompted research-

ers to investigate alternative so-called

antibody-free extraction approaches

(5). An interesting technique is immo-

bilized-metal affinity chromatography

(IMAC), which is based on the interac-

tion of metal ions, such as Ni2+, with

amino acids that feature strong elec-

tron donor groups, such as histidine.

Proteins with such amino acids on their

surface will be selectively captured by

IMAC resins. As an example, the bio-

pharmaceutical recombinant human

tumor necrosis factor-related apop-

tosis-inducing ligand (rhTRAIL) has

been quantified in human and mouse

serum down to 20 ng/mL (340 pM)

by removing 95% of matrix proteins,

while recovering >70% of the analyte

with IMAC (8).

Another technique is solid-phase

extraction (SPE) with ion-exchange

materials, which separates proteins

based on their isoelectric point (pI).

Proteins with a relatively high pI bear a

net positive charge and can be trapped

on a cation-exchange resin at neutral

or slightly alkaline pH, at which many

endogenous proteins with a lower pI

will be negatively charged and thus

not be captured. The extraction of

rhTRAIL with strong-cation exchange

SPE was found to have a similar clean-

up potential to IMAC, with an analyte

recovery of 70% and a protein removal

eff iciency of 99%. As an illustra-

tion, Figure 3 shows chromatograms

obtained for 10 ng/mL (170 pM) of

rhTRAIL in human serum, which was

extracted by strong cation exchange or

IMAC, followed by trypsin digestion

and LC–MS/MS analysis of the signa-

ture peptide.

Peptide Extraction

Removal of interfering matrix com-

ponents is also possible after diges-

tion, that is, at the peptide level. This

approach has some distinct advantages.

From a practical point of view, the opti-

mization of an SPE procedure is more

straightforward because of the wide

availability of a range of materials that

are commonly used for small-molecule

extractions and because of the more

predictable extraction behavior of

smaller peptides compared to that of

intact proteins.

The accuracy and precision of extrac-

tions may be inf luenced by protein-

protein interactions in samples (such as

binding of a biopharmaceutical to its

target or to anti-drug antibodies), or

the occurrence of aggregates. If a sam-

ple is first subjected to digestion, these

interactions will no longer inf luence

the extraction because all proteins will

have been cleaved to peptides that are

much less likely to bind to one another

with a high affinity.

No less importantly, peptide extrac-

tion does not need a protein-based

internal standard; it can instead per-

Page 32: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

32 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

form very well when using a stable-

isotope labeled form of the signature

peptide (4,6), which is considerably less

expensive and easier to obtain. It may,

however, be difficult to achieve suffi-

cient selectivity because the peptides in

a plasma digest are much more similar

to each other than the plasma proteins

were before digestion. Again, the high-

est selectivity and sensitivity is achieved

by applying immunocapture, which in

this case uses immobilized antibodies

raised against the signature peptide.

This approach is most widespread in the

field of biomarker analysis, where the

number of analytes is relatively limited

and assays are relevant to many research

groups around the globe. Large cleanup

efficiencies can be achieved in this way,

as was reported for the endogenous pro-

teins α1-antichymotrypsin (1453-fold

enrichment relative to matrix proteins)

and TNF-α (573-fold enrichment) (10).

IC at the peptide level is less popular

in biopharmaceutical analysis, probably

because of the general drawbacks of

antibody-based reagents with regard to

availability and batch-to-batch repro-

ducibility. A more generic approach

for peptide extraction from a digest is

to use conventional ion-exchange SPE,

but this needs to be carefully optimized

to obtain sufficient selectivity. A digest

of a protein-rich biological sample

(such as plasma) contains a multitude

of peptides, which all have carboxylic

and amine groups, and the signature

peptide can only be separated from

the excess of endogenous background

peptides if its pI value is sufficiently

different. Typically, the pH and ionic

strength of the loading, washing, and

elution steps need to be carefully opti-

mized for a selective extraction.

A biopharmaceutical nanobody was

quantified down to 10 ng/mL (360 pM)

in rabbit and human plasma by trypsin

digestion followed by SPE on a weak-

anion exchange phase (11). The signa-

ture peptide contained three carboxylic

acid groups and was strongly retained

by the positively charged SPE phase at

pH 5; many endogenous peptides with

less negative charges were not trapped

during sample loading or were removed

from the SPE material by a washing

step with 300 mM sodium chloride.

The mixed-mode SPE phase, which

also contained reversed-phase groups,

was subsequently neutralized at a high

pH and the (relatively polar) signature

peptide was eluted, while some less

polar endogenous peptides remained

bound by reversed-phase interactions.

In this way, two dimensions of selectiv-

ity (ion exchange and reversed phase)

were used to isolate the signature pep-

tide from the plasma digest. Figure 4

illustrates that many interfering peaks

were removed from the chromatogram

with this approach and that selectivity

was clearly improved. Of course, cat-

ion-exchange SPE can be applied in the

same way in case the signature peptide

100

%

0

100

%

0

2.32

1.513.90

4.27 5.07 5.25

5.71 5.84

4.59

2.843.52

4.06

4.20 4.55

5.03

5.305.47

6.195.88

(a)

(b)

Time (min)

Time (min)

Figure 4: LC–MS/MS (m/z 752.0 to m/z 773.3) chromatograms of the signature peptide of 10 ng/mL of a nanobody in human plasma (a) without or (b) with solid-phase extraction of the plasma digest. Analyte peak at 4.6 min. Adapted and reproduced with permission from reference 11, ©Future Science Ltd.

100 2.58e3

6.63

6.51

6.65

6.78

6.94

7.007.13

7.237.49

7.55

7.737.91

6.71

6.79 6.92

7.097.507.55

7.63

7.817.90

6.60 6.80 7.00 7.20 7.40 7.60 7.80

[rhTRAIL]=10 ng/mL [rhTRAIL]=10 ng/mL

SCX

0

%

100 2.02e3

6.60 6.80 7.00 7.20 7.40 7.60 7.80

Time (min)Time (min)

IMAC

0

%

Figure 3: LC–MS/MS (m/z 729.0 to m/z 942.4) chromatograms of the signature peptide of 10 ng/mL rhTRAIL in human serum and the corresponding blanks, pretreated with SCX or IMAC before digestion. Adapted and reproduced with permission from reference 5, ©Future Science Ltd.

Page 33: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 33www.chromatographyonline.com

100

012.50 13.00

12.70

(a)

Time (min) Time (min)

(b)

%100

013.00 14.00 15.00

15.4414.72

13.52

12.76

%Figure 5: LC–MS/MS (m/z 456.6 to m/z 852.5) chromatograms of the signature pep-tide (a) of 0.2 ng/mL of rhTRAIL spiked to dog saliva, plus corresponding blank, or (b) of endogenous human TRAIL in unspiked human saliva. Samples pretreated with IMAC before and SCX after digestion.

has multiple positive charges, and even

reversed-phase SPE might be an option

if the signature peptide is particularly

hydrophobic.

Combined Protein

and Peptide Extraction

As illustrated above, generic protein

or peptide extractions typically result

in LLOQs in the low nanograms-per-

milliliter (mid to high picomolar) range,

while IC extraction at the protein or

peptide level enables quantitation down

to mid picograms-per-milliliter (low to

mid picomolar) concentrations. If more

sensitivity is required, one option is to

combine protein and peptide extrac-

tions. Excellent selectivity and sensitiv-

ity can be reached even without anti-

body-based extraction materials, as was

shown for rhTRAIL in saliva (12). After

IMAC extraction of the protein analyte

and trypsin digestion, the digest was

further purified using SPE on a strong

cation exchange cartridge. Because of

the presence of four basic amino acids

in the signature peptide, the digest was

acidified before loading onto the SPE

phase. The peptide was then trapped

and endogenous peptides were removed

by washing with 200 mM sodium chlo-

ride. After elution at alkaline pH, the

signature peptide was quantified using

LC–MS/MS. As shown in Figure 5, a

TRAIL concentration as low as 0.2 ng/

mL (3.4 pM) could be quantified in

both dog and human saliva. In prin-

ciple, protein or peptide extractions

can be combined in many ways and as

long as the separation mechanisms are

orthogonal, improved selectivity and

sensitivity can be expected compared

to a single-extraction approach.

The ultimate combination of protein

and peptide extraction is IC of the pro-

tein analyte followed by digestion and

IC of the signature peptide. Although

this requires two specifically raised

antibodies and is by no means a generic

approach, it can result in impressive

sensitivities. The biomarker interleu-

kin-21 (IL-21) was quantified in human

serum and monkey tissues with an

LLOQ of 0.78 pg/mL (0.05 pM). This

was achieved by combining off-line

magnetic bead-based protein extraction

using an anti-IL-21 antibody with on-

line enrichment of the signature pep-

tides using immobilized anti-peptide

antibodies (13). Figure 6 shows repre-

sentative chromatograms. It is impor-

tant to realize that the obtained LLOQ

corresponds to a molar concentration

of the protein, which is five orders of

magnitude lower than that shown in

Figure 2 (digestion only). This con-

vincingly demonstrates the enormous

cleanup capability of this combination

of techniques.

LC–MS/MS Versus ELISA

Compared to ligand-binding assays,

LC–MS/MS has a number of analyti-

cal advantages such as a larger linear

dynamic range; (usually) higher accuracy

and precision because of the possibility

REMOVE KINKSfrom Stainless Steel HPLC Tubing for

BETTER FLOW���� EffEfforortltlesesslsly y reremomoveve k kininksk bby y plp acaccining g tutubiingng

inintoto tthehe pprecic sis onon h holo e ofo tthehe B Beetaa-ToT ol-22™

HPHPLCC TTubbing g StStraaigighthtneer and d clc ammpingn dowwn

���� ExExpepep rtrtlyly resestooree tthehe ooutu err diaiamemeteter r ofof

ststaiainlnlesess s ststeeeel tutubib ngng w witi hohoutut ccomomprpromomissiningg

ththe e e ininnnenerr didiamameteterer

�� EaEasisis lylyy bbenenend d d tutut bibib ngngng w wwitithohooutut kkinininkikingngng t thehe iinsnsididee

�� EnEnEnhahahancncce e e dadadatatata c ccoloo leelectctctioioon nn wiwiw ththth i i impmpmprororovevevedd d flofloflowww

Beta Tool-2™ HPLC

Tubing Straightner46046046020-20-20 BTBTBT

Learn more at www.mtc-usa.com

Page 34: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

34 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

to apply internal standards (4); the ability

to quantify multiple analytes simultane-

ously; and the fact that it does not neces-

sarily require immunological reagents (5).

The last point can be especially critical,

because such reagents may be problem-

atic to obtain or show a large batch-to-

batch variability, which makes compari-

son of results between laboratories or

over a longer period of time difficult,

if not impossible. The disadvantages of

LC–MS/MS include its generally higher

operational cost; more limited sample

throughput; and less favorable concen-

tration sensitivity. In addition, with the

digestion step that is generally needed

for LC–MS/MS, the three-dimensional

(3D) structure of a protein analyte is lost

and the analytical principle is therefore

not related to the complex molecular

structure of a protein, which determines

its pharmacological activity.

Now that more and more reports are

appearing that compare newly devel-

oped LC–MS/MS methods with exist-

ing ELISAs for the same protein ana-

lyte, it is becoming increasingly clear

that both techniques do not always give

superimposable concentration results

(14,15). Although in the world of small-

molecule quantitation, two different

results for the same sample would be

seen as proof that at least one of them

is incorrect, this is not necessarily true

for biopharmaceuticals. It should be

realized that, in contrast to small mol-

ecules, LC–MS/MS and ELISA only

use a small part of the protein molecule

for the actual quantitation, the signa-

ture peptide and the binding epitope,

respectively, and this may represent as

little as a few percent of the entire mol-

ecule. Furthermore, both techniques

are based on quite different biochemi-

cal principles, to which the structurally

complex and often heterogeneous bio-

pharmaceuticals may respond in dif-

ferent ways. Thus, neither LC–MS/MS

nor ELISA should be regarded as the

ultimate quantitation technique for bio-

pharmaceuticals, but rather as comple-

mentary tools for obtaining quantitative

information about this complicated but

very interesting class of compounds.

Acknowledgments

Stichting Technische Wetenschappen

(STW) and Samenwerkingsverband

Noord-Nederland (SNN) are gratefully

acknowledged for providing financial

support for part of the work described

in this paper.

References

(1) R. Bischoff, K.J. Bronsema, and N.C. van

de Merbel, Trends Anal. Chem. 48, 41–51

(2013).

(2) G. Hopfgartner, A. Lesur, and E. Varesio,

Trends Anal. Chem. 48, 52–61 (2013).

(3) I. van den Broek, W.M. Niessen, and

W.D. van Dongen, J. Chromatogr. B 929,

161–179 (2013).

(4) K.J. Bronsema, R. Bischoff, and N.C. van

de Merbel, J. Chromatogr. B 893-894,

1–14 (2012).

(5) D. Wilffert, R. Bischoff, and N.C. van de

Merbel, Bioanalysis 7, 763–779 (2015).

(6) K.J. Bronsema, R. Bischoff, and N.C. van

de Merbel, Anal. Chem. 85, 9528–9535

(2013).

(7) K.J. Bronsema, R . Bischoff, W.W.M.P.

Pijnappel, A.T. van der Ploeg, and N.C.

van de Merbel, Anal. Chem. 87, 4394–

4401 (2015).

(8) D. Wilffert, C.R. Reis, J. Hermans, N.

Govorukhina, T. Tomar, S. de Jong, W.J.

Quax, N.C. van de Merbel, and R. Bischoff,

Anal. Chem. 85, 10754–10760 (2013).

(9) V. Kumar, D.R. Barnidge, L.S. Chen, J.M.

Twentyman, K.W. Cradic, S.K. Grebe,

and R.J. Singh, Clin. Chem. 56, 306–313

(2010).

(10) J.R . Whiteaker, L. Zhao, H.Y. Zhang,

L.C. Feng, B.D. Piening, L. Anderson,

and A.G. Paulovich, Anal. Biochem. 362,

44–54 (2007).

(11) K.J. Bronsema, R. Bischoff, M.P. Bouche,

K. Mortier, and N.C. van de Merbel, Bio-

analysis 7, 53–64 (2015).

(12) D. Wilffert, unpublished results.

(13) J. Palandra, A. Finelli, M. Zhu, J. Mas-

ferrer, and H. Neubert, Anal. Chem. 85,

5522–5529 (2013).

(14) N.C. van de Merbel, K.J. Bronsema, and

M. Nemansky, Bioanalysis 4, 2113–2116

(2012).

(15) P. Bults, N.C. van de Merbel, and R .

Bischoff, Expert Rev. Proteomics 12, 355–

374 (2015).

Nico van de Merbel is scientific

director at the bioanalytical laboratories

of PRA Health Sciences in Assen, The

Netherlands and Lenexa, Kansas, and

honorary professor at the University of

the Groningen, The Netherlands. ◾

400

350

300

250

200

150

100

50

QLI

DIV

DQ

LK In

ten

sity

(a) (b)

04.5 5.0

Time (min) Time (min)5.5

400

350

300

250

200

150

100

50

04.5 5.0 5.5

Figure 6: LC–MS/MS chromatograms of two mass transitions (m/z 592.8 to m/z 943.5 in red and m/z 592.8 to m/z 830.5 in blue) of the signature peptide of IL-21 in human serum; (a) blank and (b) 0.78 pg/mL. Samples pretreated with immuno-capture both before and after digestion. Adapted and reproduced with permis-sion from reference 13, ©American Chemical Society.

Page 35: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 35www.chromatographyonline.com

Koen Sandra, Alexia Ortiz, and Pat Sandra

Analyzing Host Cell Proteins Using Off-Line Two-Dimensional Liquid Chromatography–Mass Spectrometry

Protein biopharmaceuticals are commonly produced recombinantly in

mammalian, yeast, or bacterial expression systems. In addition to the

therapeutic protein, these cells also produce endogenous host cell

proteins (HCPs) that can contaminate the biopharmaceutical product,

despite major purification efforts. Since HCPs can affect product

safety and efficacy, they need to be closely monitored. Enzyme-linked

immunosorbent assays (ELISA) are recognized as the gold standard for

measuring HCPs because of their high sensitivity and high throughput,

but mass spectrometry (MS) is gaining acceptance as an alternative

and complementary technology for HCP characterization. This article

reports on the use of off-line two-dimensional liquid chromatography–

mass spectrometry (2D-LC–MS) for the characterization of HCPs and

their monitoring during downstream processing.

In contrast to small-molecule drugs

that are commonly synthesized by

chemical means, protein biophar-

maceuticals result from recombinant

expression in nonhuman host cells.

As a result, the biotherapeutic is co-

expressed with hundreds of host cell

proteins (HCPs) with different physi-

cochemical properties present in a wide

dynamic concentration range. During

downstream processing, the levels of

HCPs are substantially reduced to a

point considered acceptable to regula-

tory authorities (typically <100 ppm–

ng HCP/mg product). These process-

related impurities are considered as

critical quality attributes because they

might induce an immune response,

cause adjuvant activity, exert a direct

biological activity (such as cytokines),

or act on the therapeutic itself (for

example, proteases) (1,2). To men-

tion some specif ic examples, during

the clinical development phase of

Omnitrope, Sandoz’s human growth

hormone biosimilar expressed in E .

coli, adverse events associated with

residual HCPs were encountered. The

European Medicines Agency (EMA)

only granted approval after additional

purification steps for HCP clearance

were incorporated (3–5). Scientists at

Biogen Idec demonstrated fragmenta-

tion of a highly purified monoclonal

antibody as a result of residual Chi-

nese hamster ovarian (CHO) cell pro-

tease activity in the drug substance,

despite an enormous purif ication

effort undertaken (protein A aff in-

ity chromatography with subsequent

orthogonal purif ication steps by cat-

ion- and anion-exchange chromatog-

raphy) (6). The authors of the study

state that it is of utmost importance

to identify residual protease activity

early in process development to allow

a revision of the purification scheme

or ultimately to knockdown the spe-

cific protease gene.

Multicomponent enzyme-linked

Ph

oto

Cre

dit

: M

on

ty R

aku

sen

/Get

ty I

mag

es

Page 36: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

36 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

immunosorbent assay (ELISA) is pres-

ently the workhorse method for HCP

testing because of its high throughput,

sensitivity, and selectivity (1,2). Poly-

clonal antibodies used in the test are

typically generated by the immuniza-

tion of animals with an appropriate

preparation derived from the produc-

tion cell, minus the product-coding

gene. However, ELISA does not com-

prehensively recognize all HCP spe-

cies—that is, it cannot detect HCPs

to which no antibody was raised, it

only provides information on the total

amount of HCPs without providing

insight in individual HCPs, and, in a

multicomponent setup, it has a poor

quantitation power. In that respect,

MS nicely complements EL ISA

because it can provide both qualitative

and quantitative information on indi-

vidual HCPs. In recent years, various

papers have appeared dealing with the

mass spectrometric (MS) analysis of

HCPs (2,3,7–12). These studies typi-

cally rely on bottom-up proteomics

approaches in which peptides derived

from the protein following proteolytic

digestion are handled. A clear trend

is observed toward the use of upfront

multidimensional chromatography

to tackle the enormous complexity

and wide dynamic range (2,3,7,8,10).

Compared to one-dimensional liq-

Supernatants

Buffer exchange/desalting

Reduction/alkylation/digestion

Database search

Protein ID/Quant

High pH reversed-phase LC x low pH reversed-phase LC

QTOF MS/MS

Time (min)

0 5 10 15 20 25 30 35

mA

U

0

200

400

600

800

1000

1200

1400

Fraction 11-19

7x10

0

7x10

0

7x10

0

7x10

0

7x10

0

7x10

0

7x10

0

7x10

0

7x10

0

MS counts vs. Acquisition Time (min)

4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40

Fraction 12

Fraction 13

Fraction 14

Fraction 15

Fraction 16

Fraction 17

Fraction 18

Fraction 19

Fraction 11

Figure 1: Workflow for the character-ization of HCPs using off-line 2D-LC–MS/MS.

Figure 2: First-dimension reversed-phase LC–UV 214 nm chromatogram of a selected downstream manufacturing sample. HPLC system: Agilent Technologies 1200; Col-umn: 150 mm × 2.1 mm, 3.5-μm Waters XBridge BEH C18; mobile-phase A: 10 mM NH4HCO3 pH 10; mobile-phase B: acetonitrile; flow rate: 200 μL/min; gradient: 5–50% B in 30 min; column temperature: 25 °C; injection volume: 50 μL; fraction interval: 1.5 min (300-μL fractions).

Figure 3: Second dimension LC–MS/MS chromatograms of selected fractions (Fig-ure 2). HPLC system: Thermo Scientific Ultimate3000 RSLC nano; MS system: Agilent Technologies 6530 Q-TOF; column: 150 mm × 75 μm, 3-μm Thermo Scientific Acclaim PepMap100 C18; precolumn: 20 mm × 75 μm, 3-μm Acclaim PepMap100 C18 (Thermo Scientific); mobile-phase A: 2% acetonitrile, 0.1% formic acid; mobile-phase B: 80% acetonitrile, 0.1% formic acid; loading solvent: 2% acetonitrile, 0.1% formic acid; flow rate: 300 nL/min (nano pump), 5 μL/min (loading pump); gradient: 0–60% B in 60 min; column temperature: 35°C; injection volume: 20 μL.

Page 37: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

NOVEMBER 2016 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS 37www.chromatographyonline.com

uid chromatography (1D-LC), two-

dimensional LC (2D-LC) drastically

increases peak capacity as long as

the two dimensions are orthogonal

(13). In a 1D chromatographic setup

the separation space is dominated by

peptides derived from the therapeutic

protein, in 2D-LC the increased peak

capacity allows one to look substan-

tially beyond the therapeutic peptides

and detect HCPs at low levels. Three

recent papers using 2D-LC–MS/

MS demonstrate that HCPs can be

revealed at levels as low as 10 ppm

(2,3,7). In these cases, label-free quan-

tification was based on the three most

intense tryptic peptides making use of

single-point calibration against spiked

exogenous proteins.

An off-line 2D-LC–MS/MS setup

was used in our laboratory for the

characterization of HCPs throughout

the downstream manufacturing of a

therapeutic enzyme recombinantly

expressed in yeast. The workf low is

schematically presented in Figure 1.

Supernatant was collected at different

purification steps. Following desalting

of the supernatant, the proteins were

reduced using dithiothreitol (DTT)

and alkylated using iodoacetamide

(IAM) before overnight trypsin diges-

tion. The peptide mixture was subse-

quently subjected to 2D-LC–MS/MS.

In successfully applying 2D-LC ,

the selectivity of the two separation

mechanisms toward the peptides

must differ substantially to maximize

orthogonality and, hence, resolution.

Various orthogona l combinations

targeting different physicochemical

properties of the peptides have been

described. Bottom-up proteomics

setups initially relied on the com-

bination of strong-cation exchange

and reversed-phase LC to separate by

charge in the first dimension and by

hydrophobicity in the second dimen-

sion (13–15). In recent years, various

researchers have shifted their efforts

to the combination of reversed-phase

LC and reversed-phase LC (13,16–19).

The orthogonality in this nonobvious

combination is mainly directed by the

mobile-phase pH, in this instance,

high pH in the f irst dimension and

low pH in the second dimension,

and by the zwitterionic nature of the

peptides. In contrast to the combina-

tion of strong-cation exchange and

reversed-phase LC , where the f irst

dimension has an intrinsic low peak

capacity, the combination of reversed-

phase LC in both dimensions benefits

from the high peak capacities of the

two independent dimensions, which

results in an overall high peak capac-

ity of the 2D setup.

In the characterization of yeast

HCPs, we opted to use reversed-phase

LC in both dimensions with the first

dimension operated at pH 10 and

the second dimension at pH 2.6. An

acidic pH is preferred in the second

dimension since it maximizes MS sen-

Recombinant therapeutic enzyme

99.53

45

99.47

46

0.44

0.008

0.0130.43

0.015

7 1

211

20.000.07

5

0.054

0.023

99.77

0.0346

3

0.06

3

0.09

9

Exogenous glycosidase Metallopeptidase (HCP)

Serine carboxypeptidase 1 (HCP) Aspartyl peptidase (HCP)

Pe

rce

nta

ge

Pe

rce

nta

ge

Pe

rce

nta

ge

Pe

rce

nta

ge

Pe

rce

nta

ge

Pe

rce

nta

ge

Purification stage

1 2 3 1 2 3

Purification stage

Purification stage

1 2 3 1 2 3

Purification stage

Purification stage

1 2 3 1 2 3

Purification stage

Serine carboxy peptidase 2 (HCP)

Figure 4: Evolution of the therapeutic enzyme, the exogenous glycosidase, and some selected HCPs throughout the final stages of downstream manufacturing. The numbers on the bars represent the relative abundances and the number of unique peptides identified and quantified. Relative abundances were calculated based on the MS signal of identified pep-tides. Note: the therapeutic enzyme contains various fully occupied glycosylation sites. These glycopeptides are not identi-fied by the MS/MS search engine and therefore not taken into account in the calculation of relative abundances.

Page 38: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

38 ADVANCES IN BIOPHARMACEUTICAL ANALYSIS NOVEMBER 2016 www.chromatographyonline.com

sitivity for peptides. Figure 2 shows

the f irst dimension ultraviolet (UV)

214-nm chromatogram of a selected

downstream manufacturing sample.

A reversed-phase LC column with an

internal diameter of 2.1 mm was used,

which allowed substantial amounts of

sample to be loaded, in this particu-

lar case the amount corresponding to

115 μg of protein. The peptides were

nicely spread throughout the aceto-

nitrile gradient and 22 fractions were

collected and further processed after

drying and reconstitution in 50 μL of

low-pH mobile-phase A (2% acetoni-

trile and 0.1% formic acid).

The second dimension consisted of

a reversed-phase LC capillary column

with an internal diameter of 75 μm,

which was directly coupled through a

nanospray interface to high resolution

quadrupole time-of-f light (QTOF)-

MS operated in the data-dependent

acquisition (DDA) mode. The LC–

MS/MS traces of some selected frac-

tions are shown in Figure 3 illustrat-

ing good orthogonality between first

and second dimension separations.

The MS system was programed so

that an MS survey measurement pre-

ceded three dependent MS/MS acqui-

sitions. Precursors selected twice for

collision-induced dissociation (CID)

were placed in an exclusion list. Gen-

erated MS/MS spectra were subjected

to database searching (yeast proteins

and therapeutic enzyme sequence)

and relative protein quantif ication

was performed from total protein

intensities computed by the Spectrum

Mill search engine. Total intensity

is the sum of intensities for all spec-

tra of peptides belonging to a given

protein. Figure 4 shows the evolu-

tion of the therapeutic enzyme and

some selected HCPs throughout the

f inal stages of purif ication. Of par-

ticular interest, during downstream

manufacturing, a nonyeast-derived

glycosidase was added to shape the

glycosylation profile of the therapeu-

tic enzyme (in between stage 1 and 2).

This glycosidase temporarily reduced

the purity of the therapeutic enzyme

but was rapidly cleared. The HCPs

detected were mainly proteases, which

inf luenced stability of the therapeu-

tic enzyme. While some were clearly

reduced throughout the process (ser-

ine carboxypeptidase 1 and aspartyl

peptidase), others were enriched (ser-

ine carboxypeptidase 2 and metallo-

peptidase). While these proteases were

present at low levels (<0.1%), stability

studies have shown that they act on

the protein. With the identity of these

proteases revealed, they could be the

subject of a gene knockout to increase

product stability.

It is important to note that none of

the HCPs reported could be identi-

f ied using 1D-LC–MS/MS operated

under exactly the same conditions

as reported in the legend of Figure

3. Column load was evidently much

lower compared to the 2D-LC–MS/

MS analysis (4 μg versus 115 μg).

In conclusion, off-line 2D-LC–MS/

MS represents a valuable new tool for

the characterization of HCPs and their

monitoring throughout downstream

processing. The use of multidimen-

sional chromatography substantially

increases peak capacity and improves

the dynamic range providing access to

otherwise unmined HCPs. Based on the

output of the 2D-LC–MS/MS experi-

ment, processes can be adjusted and

identified HCPs can be incorporated

in single product ELISAs or in targeted

multiple reaction monitoring (MRM)

MS assays for routine monitoring.

References

(1) F. Wang, D. R ichardson, and M.

Shameem, BioPharm Int. 28 , 32–38

(2015).

(2) Q. Zhang, A.M. Goetze, H. Cui, J.

Wylie, S. Trimble, A. Hewig, and G.C.

Flynn, mAbs 6, 659–670 (2014).

(3) C .E . Doneanu, A. Xenopoulos, K .

Fadgen, J. Murphy, S.J. Skilton, H.

Prentice, M. Stapels, and W. Chen, mAbs

4, 24–44 (2012).

(4) M. Pavlovic, E. Girardin, L. Kapetanovic,

K. Ho, and J.H. Trouvin, Horm. Res. 69,

14–21 (2008).

(5) J . G e i g e r t , T h e C h a l l e n g e o f

CMC R e gu l a t o r y Comp l i anc e for

Biopharmaceuticals and Other Biologics

(Springer Science & Business Media,

Heidelberg, Germany, 2013).

(6) S.X . Gao, Y. Zhang, K . Stansberry-

Perkins, A. Buko, S. Bai, V. Nguyen, and

M.L. Brader, Biotechnol . Bioeng. 108,

977–982 (2011).

(7) M.R. Schenauer, G.C. Flynn, and A.M.

Goetze, Anal. Biochem. 428, 150–157

(2012).

(8) J.H. Thompson, W.K. Chung, M. Zhu,

L. Tie, Y. Lu, N. Aboulaich, R. Strouse,

and W. Mo, Rapid Comm. Mass Spectrom.

28, 855–860 (2014).

(9) V. Reisinger, H. Toll, R .E . Mayer, J.

Visser, and F. Wolschin, Anal. Biochem.

463, 1–6 (2014).

(10) C .E . Doneanu and W. Chen, Methods

Mol. Biol. 1129, 341–350 (2014).

(11) K. Bomans, A. Lang, V. Roedl, L. Adolf,

K . Kyriosoglou, K . Diepold, G. Eberl,

M. Mølhøj, U. Strauss, C . Schmalz,

R . Vogel, D. Reusch, H. Wegele, M.

Wiedmann, and P. Bulau, PLoS One 8,

e81639 (2013).

(12) M.R. Schenauer, G.C. Flynn, and A.M.

Goetze, Biotechnol. Bioeng. 29, 951–957

(2013).

(13) K . Sandra, M. Moshir, F. D’hondt,

R . Tuytten, K . Verleysen, K . Kas, I.

François, and P. Sandra, J. Chromatogr.

B 877, 1019–1039 (2009).

(14) M.P. Washburn, D.A. Wolters, and J.R.

Yates, Nat. Biotechnol . 19, 242–247

(2001).

(15) D.A. Wolters, M.P. Washburn, and

J.R. Yates, Anal. Chem. 73, 5683–5690

(2001).

(16) N. Delmotte, M. Lasaosa, A. Tholey, E.

Heinzle, and C .G. Huber, J. Proteome

Res. 6, 4363–4373 (2007).

(17) M. Gilar, P. Olivova, A.E. Daly, and J.C.

Gebler, Anal . Chem. 77, 6426–6434

(2005).

(18) G. Vanhoenacker, I. Vandenheede, F.

David, P. Sandra, and K. Sandra, Anal.

Bioanal. Chem. 407, 355–366 (2015).

(19) K . Sandra, K . Mortier, L. Jorge, L.C .

Perez, P. Sandra, S. Priem, S. Poelmans,

and M.P. Bouche, Bioanalysis 6, 1201–

1213 (2014).

Koen Sandra is Director at the

Research Institute for Chromatography

(RIC) in Kortrijk, Belgium.

Alexia Ortiz is a Proteomics Re-

searcher at the Research Institute for

Chromatography (RIC).

Pat Sandra is Chairman at the

Research Institute for Chromatography

(RIC) and Emeritus Professor at Ghent

University in Ghent, Belgium. ◾

Page 39: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

LCGC’s global digital magazine provides unique and

timely applications, news, and interviews especially

relevant to separation scientists across industry sectors.

SIGN UP FOR FREE AT chromatographyonline.com/column-subscribe

Page 40: ADVANCES IN BIOPHARMACEUTICAL ANALYSISimages2.advanstar.com/PixelMags/lcgc-na/pdf/2016-11-sp.pdf · of Biopharmaceuticals in Various Liquid Chromatographic Modes The recent trends

www.tosohbioscience.com

TSKgel and Tosoh Bioscience are registered trademarks of Tosoh Corporation.

• High resolution• Improved peak shape• Increased effi ciency• High throughput

• Robust • Reproducible• Easily transferrable between UHPLC and HPLC systems

DUAL FUNCTIONALITY FOR USE IN BOTH UHPLC AND HPLC SYSTEMS

TSKgel UP-SW3000, 2 μm columns offer:

Yielding methods that are:

THE NEW GOLD STANDARD FOR MONOCLONAL

ANTIBODY CHARACTERIZATION

Dete

ctor

resp

onse

(m

AU)

Retention time (minutes)0

-0.501 2 3 4 5 6 7 8 9 10 11 12 13 14 15

0.50

1.50

2.50

3.50

4.50

0.00

1.00

2.00

3.00

4.00

5.00 UHPLC system

1 - H

MW

S - 6

.475

5 - L

MW

S - 9

.200

3 - M

ain

peak

- 7.

408

2 - T

riple

LC

- 7.0

67

4 - F

ab/c

- 7.

950

Improvedresolution inLMW side

Improvedresolution inHMW side

TSKgel UP-SW3000

TSKgel UP-SW3000

TSKgel® UP-SW3000, 2 μm UHPLC/HPLC SEC ColumnsTSKgel® UP-SW3000, 2 μm UHPLC/HPLC SEC ColumnsTSKgel® UP-SW3000, 2 μm UHPLC/HPLC SEC ColumnsTSKgel® UP-SW3000, 2 μm UHPLC/HPLC SEC Columns

Contact us today to get your TSKgel UP-SW3000 column:

800-366-4875, option #3