Rising Star

71
Rising Star The past, present, and future of hydrophilic interaction liquid chromatography LC TROUBLESHOOTING A view of the selectivity landscape BIOPHARMACEUTICAL PERSPECTIVES CZE–MS for characterizing mAbs ANALYSIS FOCUS The evolution of foodomics March 2019 Volume 32 Number 3 www.chromatographyonline.com

Transcript of Rising Star

Rising StarThe past, present, and future of hydrophilic interaction

liquid chromatography

LC TROUBLESHOOTING

A view of the selectivity

landscape

BIOPHARMACEUTICAL

PERSPECTIVES

CZE–MS for characterizing mAbs

ANALYSIS FOCUS

The evolution of

foodomics

March 2019

Volume 32 Number 3

www.chromatographyonline.com

Supelco® analytical products.

Exquisite accuracy. Every time.Maintaining your high standards

requires analytical products you

can rely on, along with the right

documentation and expert guidance.

That's why the entire range of

Supelco® analytical products, from

spectroscopy and spectrometry,

to mobile analysis, chromatography

and titration, are exquisitely

developed to comply with the

same guidelines and regulations

that you do.

So for a fl awless performance

time after time, choose Supelco®

analytical products.

The Life Science Business

of Merck operates as

MilliporeSigma in the

US and Canada.

Merck, the vibrant M and Supelco are trademarks of

Merck KGaA, Darmstadt, Germany or its affi liates. All other

trademarks are the property of their respective owners.

Detailed information on trademarks is available via publicly

accessible resources.

© 2019 Merck KGaA, Darmstadt, Germany and/or its affi liates.

All Rights Reserved.

For more information, please visit:

SigmaAldrich.com/Supelco

Chro

mato

gra

phy

Find what you are looking for!

MACHEREY-NAGEL

Manufacturer of premium

chromatography media

www.mn-net.com

� CHROMABOND® columns with classical and

innovative SPE phases

� Original NUCLEOSIL®, professional NUCLEODUR®

and highly efficient NUCLEOSHELL® HPLC columns

� Robust TLC glass plates, economical aluminum and

polyester sheets

Coupling powersPioneering new fields in ultra-trace analysis, the new GCMS-TQ 8050 NX triple quadrupole couplesthe powers of a world-leading GC and a newlydesigned detector. Both provide outstanding sensi-tivity at femtogram and even sub-femtogram levels.

Superior performance of NX technologiese.g. new flow controller and time management formaintenance

A wide variety of optional products supportstrace analysissuch as autosamplers and inlets

Comfortable, easy change of accessories through the advanced, illuminated GC oven

The GCMS-TQ 8050 NX complements the ShimadzuNX family, coupling the Nexis GC-2030 with thequadrupole series TQ-8050, TQ-8040 or QP-2020.Shimadzu’s NX series provides high-end GCMS solu-tions for every analytical challenge.

www.shimadzu.eu /coupling-powers

LC•GC Europe March 2019108

Editorial Policy:

All articles submitted to LC•GC Europe

are subject to a peer-review process in association

with the magazine’s Editorial Advisory Board.

Cover:

Original materials courtesy: Argus/stock.adobe.

com

Features148 The Rising Role of Foodomics

Lewis Botcherby

LCGC Europe spoke to Alejandro Cifuentes from the Institute of Food

Science Research, in Madrid, Spain, about his current foodomics

research projects, the overall state of the fi eld, and the future of

foodomics.

Columns126 LC TROUBLESHOOTING

Looking for New Selectivity in Reversed-Phase Liquid

Chromatography? A View of the Selectivity Landscape Over

the Past Two Years

Dwight R. Stoll

The offerings of commercially available columns for reversed-phase

liquid chromatography continue to expand. Are these columns similar

or different compared to what is already available?

130 BIOPHARMACEUTICAL PERSPECTIVES

Middle-Up Characterization of the Monoclonal Antibody

Infl iximab by Capillary Zone Electrophoresis–Mass Spectrometry

Klára Michalíková, Elena Dominguez-Vega, Govert W. Somsen, and

Rob Haselberg

The potential of CZE–MS for the middle-up characterization of mAbs is

highlighted.

140 COLUMN WATCH

Perspectives on the Adoption and Utility of 1.0-mm Internal

Diameter Liquid Chromatography Columns

David S. Bell

The utility of 1.0-mm internal diameter (i.d.) columns, and the arenas

in which they play a relatively strong role, are investigated.

144 SAMPLE PREPARARTION PERSPECTIVES

Recent Advances in Solid-Phase Microextraction, Part 1: New

Tricks for an Old Dog

Douglas E. Raynie

Advances in SPME are discussed in this month’s instalment.

160 THE ESSENTIALS

Hydrophobic Interaction Chromatography of Proteins

A discussion of HIC theory and its application to the analysis of proteins

Departments154 Products

158 Events

161 The Applications Book

COVER STORY114 Hydrophilic Interaction Liquid Chromatography: An Update

David V. McCalley

This article is an update on the technique

of HILIC and covers recent ideas on the

mechanism of separation, and how it may

be manipulated to suit the separation of

particular sample types. The advantages of

HILIC are discussed, and also the actual and

perceived disadvantages of the technique.

March | 2019

Volume 32 Number 3

d

It’s the DAWN of a new day.

Learn more at wyatt.com/NextGen

LC•GC Europe March 2019110

‘Like’ our page LCGC Join the LCGC LinkedIn groupFollow us @ LC_GC

Published by

MultiMedia Healthcare

LLC

Vice President/

Group Publisher

Mike Tessalone

[email protected]

Editorial Director

Laura Bush

[email protected]

Editor-in-Chief

Alasdair Matheson

[email protected]

Managing Editor

Kate Jones

[email protected]

Associate Editor

Lewis Botcherby

[email protected]

Associate Publisher

Oliver Waters

[email protected]

Sales Executive

Liz Mclean

[email protected]

Senior Director, Digital

Media

Michael Kushner

[email protected]

Webcast Operations

Manager

Kristen Moore

[email protected]

Project Manager

Vania Oliveira

[email protected]

Digital Production Manager

Sabina Advani

[email protected]

Managing Editor Special

Projects

Kaylynn Chiarello-Ebner

kaylynn.chiarello.ebner@

ubm.com

Art Director

Dan Ward

[email protected]

Subscriber Customer Service

Visit (chromatographyonline.com)

to request or change a

subscription or call our customer

service department on

+001 218 740 6877

Hinderton Point,

Lloyd Drive,

Cheshire Oaks,

Cheshire,

CH65 9HQ, UK

Tel. +44 (0)151 353 3500

Fax +44 (0)151 353 3601

Daniel W. Armstrong

University of Texas, Arlington, Texas, USA

Günther K. Bonn

Institute of Analytical Chemistry and

Radiochemistry, University of Innsbruck,

Austria

Deirdre Cabooter

Department of Pharmaceutical and

Pharmacological Sciences, University of

Leuven, Belgium

Peter Carr

Department of Chemistry, University

of Minnesota, Minneapolis, Minnesota,

USA

Jean-Pierre Chervet

Antec Scientific, Zoeterwoude, The

Netherlands

Jan H. Christensen

Department of Plant and Environmental

Sciences, University of Copenhagen,

Copenhagen, Denmark

Danilo Corradini

Istituto di Cromatografia del CNR, Rome,

Italy

Hernan J. Cortes

H.J. Cortes Consulting,

Midland, Michigan, USA

Gert Desmet

Transport Modelling and Analytical

Separation Science, Vrije Universiteit,

Brussels, Belgium

John W. Dolan

LC Resources, McMinnville, Oregon,

USA

Anthony F. Fell

Pharmaceutical Chemistry,

University of Bradford, Bradford, UK

Attila Felinger

Professor of Chemistry, Department of

Analytical and Environmental Chemistry,

University of Pécs, Pécs, Hungary

Francesco Gasparrini

Dipartimento di Studi di Chimica e

Tecnologia delle Sostanze Biologicamente

Attive, Università “La Sapienza”, Rome,

Italy

Joseph L. Glajch

Momenta Pharmaceuticals, Cambridge,

Massachusetts, USA

Davy Guillarme

School of Pharmaceutical Sciences,

University of Geneva, University of

Lausanne, Geneva, Switzerland

Jun Haginaka

School of Pharmacy and Pharmaceutical

Sciences, Mukogawa Women’s

University, Nishinomiya, Japan

Javier Hernández-Borges

Department of Chemistry

(Analytical Chemistry Division),

University of La Laguna

Canary Islands, Spain

John V. Hinshaw

Serveron Corp., Beaverton, Oregon,

USA

Tuulia Hyötyläinen

VVT Technical Research of Finland,

Finland

Hans-Gerd Janssen

Van’t Hoff Institute for the Molecular

Sciences, Amsterdam, The Netherlands

Kiyokatsu Jinno

School of Materials Sciences, Toyohasi

University of Technology, Japan

Huba Kalász

Semmelweis University of Medicine,

Budapest, Hungary

Hian Kee Lee

National University of Singapore,

Singapore

Wolfgang Lindner

Institute of Analytical Chemistry,

University of Vienna, Austria

Henk Lingeman

Faculteit der Scheikunde, Free University,

Amsterdam, The Netherlands

Tom Lynch

Analytical consultant, Newbury, UK

Ronald E. Majors

Analytical consultant, West Chester,

Pennsylvania, USA

Debby Mangelings

Department of Analytical Chemistry and

Pharmaceutical Technology, Vrije

Universiteit, Brussels, Belgium

Phillip Marriot

Monash University, School of Chemistry,

Victoria, Australia

David McCalley

Department of Applied Sciences,

University of West of England, Bristol, UK

Robert D. McDowall

McDowall Consulting, Bromley, Kent, UK

Mary Ellen McNally

DuPont Crop Protection, Newark,

Delaware, USA

Imre Molnár

Molnar Research Institute, Berlin, Germany

Luigi Mondello

Dipartimento Farmaco-chimico, Facoltà

di Farmacia, Università di Messina,

Messina, Italy

Peter Myers

Department of Chemistry,

University of Liverpool, Liverpool, UK

Janusz Pawliszyn

Department of Chemistry, University of

Waterloo, Ontario, Canada

Colin Poole

Wayne State University, Detroit,

Michigan, USA

Fred E. Regnier

Department of Biochemistry, Purdue

University, West Lafayette, Indiana, USA

Harald Ritchie

Advanced Materials Technology, Chester,

UK

Koen Sandra

Research Institute for Chromatography,

Kortrijk, Belgium

Pat Sandra

Research Institute for Chromatography,

Kortrijk, Belgium

Peter Schoenmakers

Department of Chemical Engineering,

Universiteit van Amsterdam, Amsterdam,

The Netherlands

Robert Shellie

Deakin University, Melbourne, Australia

Yvan Vander Heyden

Vrije Universiteit Brussel, Brussels,

Belgium

Editorial Advisory Board

Subscribe on-line at

www.chromatographyonline.com

The Publishers of LC•GC Europe would like to thank the members of the Editorial Advisory Board

for their continuing support and expert advice. The high standards and editorial quality associated

with LC•GC Europe are maintained largely through the tireless efforts of these individuals.

LCGC Europe provides troubleshooting information and application solutions on all aspects of

separation science so that laboratory-based analytical chemists can enhance their practical

knowledge to gain competitive advantage. Our scientific quality and commercial objectivity

provide readers with the tools necessary to deal with real-world analysis issues, thereby

increasing their efficiency, productivity and value to their employer.

SUBSCRIPTIONS: LC•GC Europe is free to qualifi ed readers in Europe. To apply for a free subscription, or to change your name or address, go to

www.chromatographyonline.com, click on Subscribe, and follow the prompts.

To cancel your subscription or to order back issues, please email your request to

[email protected], putting LCE in the subject line.

Please quote your subscription number if you have it.

MANUSCRIPTS: For manuscript preparation guidelines, visit www.chromatographyonline.com or

call the Editor, +44 (0)151 353 3500. All submissions will be handled with reasonable care, but the

publisher assumes no responsibility for safety of artwork, photographs or manuscripts. Every

precaution is taken to ensure accuracy, but the publisher cannot accept responsibility for the

accuracy of information supplied herein or for any opinion expressed.

DIRECT MAIL LIST: Telephone: +44 (0)151 353 3500.

Reprints: Reprints of all articles in this issue and past issues of this publication are available

(250 minimum). Licensing and Reuse of Content: Contact our official partner, Wright’s Media, about

available usages, license fees, and award seal artwork at [email protected] for more

information. Please note that Wright’s Media is the only authorized company that we’ve partnered

with for MultiMedia Healthcare materials.

© 2019 MultiMedia Healthcare LLC (UK) all rights reserved. No part of the publication may be

reproduced in any material form (including photocopying or storing it in any medium by electronic

means and whether or not transiently or incidentally to some other use of this

publication) without the written permission of the copyright owner except in accord-

ance with the provisions of the Copyright Designs & Patents Act (UK) 1988 or under

the terms of the license issued by the Copyright License Agency’s 90 Tottenham

Court Road, London W1P 0LP, UK.

Applications for the copyright owner’s permission to reproduce any part of this

publication outside of the Copyright Designs & Patents Act (UK) 1988 provisions,

should be forwarded in writing to Permission Dept. fax +1 732-647-1104 or email:

[email protected]. Warning: the doing of an unauthorized act in relation to a

copyright work may result in both a civil claim for damages and criminal prosecution.

10% Post

Consumer

Waste

We’ll focus on the water,

so you can focus on

making a diff erence.

Whether you’re rewriting the textbooks, striving

for analytical perfection or protecting the

vulnerable, your work should be your focus not

your lab water.

That’s why the Milli-Q® portfolio supplies

best-in-class water solutions for every kind of

water purity, because we cater for every kind of

laboratory. And with the right expert support

it means we can save you precious time and

physical space, so you can keep your focus on

making the diff erence that really matters.

To fi nd out more, visit:

MerckMillipore.com/labwater

Merck, the vibrant M and Milli-Q are trademarks of Merck KGaA,

Darmstadt, Germany or its affi liates. All other trademarks are

the property of their respective owners. Detailed information on

trademarks is available via publicly accessible resources.

© 2019 Merck KGaA, Darmstadt, Germany and/or its affi liates.

All Rights Reserved.

The Life Science Business of Merck operates

as MilliporeSigma in the US and Canada.

112 LC•GC Europe March 2019

LCGC ONLINE

Selected highlights of digital content from LCGC Europe and LCGC North America:

PHARMACEUTICAL PERSPECTIVESThe Role of Liquid

Chromatography and Gas

Chromatography in the

Analysis of Illegal Medicines

and Health Products

This review article will give a

general overview of the LC and

GC methods used by analytical

laboratories for the detection and characterization of

suspected illegal medicines and health products.

Read Here: https://goo.gl/j6JNeL

CONNECT WITH LCGCStay in Touch with LCGC and Keep

Updated with the Latest News

Follow us on social media to keep up-to-date

with the latest troubleshooting tips and

technical peer-reviewed articles featured

on our website. Follow @LC_GC on Twitter,

join our LCGC Magazine LinkedIn group, or

Like our page on Facebook. You are also

free to post your questions or discussions for

other members to view and comment on!

LCGC BLOGHPLC Diagnostic Skills—

Noisy Baselines

Just as medical practitioners are

able to discern worrying features

from a variety of medical physics

devices, we need to develop

the skill to identify worrying

symptoms from our HPLC instrument output.

Read Here: https://goo.gl/ABCpHW

NEWSElevated Uranium

Concentrations Found

in Groundwater of Rio

de Janeiro Region

Researchers from the PUC-Rio

in Rio de Janeiro, Brazil,

have found unexpectedly

high uranium concentrations in groundwater samples

from the mountainous region near Rio de Janeiro City.

Read Here: https://goo.gl/qoVweL

WEBCASTSKeep Up-to-Date

with Upcoming and

On-Demand Webcasts

Working in partnership

with industry leaders,

LCGC broadcasts

live technical tutorial-style webcasts, as well as

application-based tutorials. A wide range of topics are

covered and the full list of upcoming and on-demand

webcasts can be found on our website at

www.chromatographyonline.com/LCGCwebseminars

CHROMACADEMYCHROMacademy is now free for all university

students and staff. Sign up today for our

animated and interactive e-learning courses.

Find Out More Here: www.chromacademy.

com/Agilent-uni.html

INTERVIEWGoing Green in Pharmaceutical Analysis

LCGC Europe spoke to Yong

Liu and Adam Socia about

the cost-saving benefits

of implementing green

chromatography in the

pharmaceutical sector, the

importance of AMVI, and effective

practices to reduce solvent

consumption and replace harmful solvents.

Read Here: https://goo.gl/oWruoY

PEER-REVIEWED ARTICLEFundamental and Practical Aspects

of LC and CE Techniques for the

Analysis of Phenolic Compounds

in Plants and Plant-Derived

Food, Part 2: Capillary

Electromigration Techniques

In part 2 of this review article

the authors discuss the

fundamental principles and

practical aspects of electromigration techniques.

Read Here: https://goo.gl/MkVTDG

Photo Credit: Abel Tumik/

Shutterstock.com

Photo Credit: Skyler Ewing/

Shutterstock.com

Photo Credit: dabldy/stock.adobe.com

Photo Credit: Oleksii Fedo-

renko/ Shutterstock.com

Agilent 8890 GC system

Agilent 8860 GC system

Spend More Time on What MattersNew Agilent 8890 and 8860 intelligent GC systems

First, Agilent Intuvo redrew the boundaries of GC intelligence. Now, two new GCs have

extended that intelligence to the entire portfolio of Agilent GC systems. So, you can

have the freedom to work the way you want—while delivering quality data, every time.

The Agilent 8890 GC ensures that your system is working properly, and guides you

through problem resolution. What’s more, mobile access features let you check on

your lab anytime, anywhere.

The Agilent 8860 GC makes routine analysis anything but routine. It monitors system

health, tracks injections, and alerts you to leaks. So, you can plan your work—including

maintenance—rather than react to unexpected downtime.

Learn about the latest additions to our family of intelligent GC systems:

www.agilent.com/chem/gc

© Agilent Technologies, Inc. 2019

It is now almost 30 years since the publication of Alpert’s

landmark paper that named the technique of hydrophilic

interaction liquid chromatography (HILIC) and discussed its

mechanism and applications (1). Alpert clearly recognized

that separations were influenced by the partition of solutes

between a water layer held on the surface of a polar column

and the bulk mobile phase rich in an organic solvent such as

acetonitrile. Additional mechanisms, such as ionic retention

and adsorption, can be superimposed on this process.

Retention increases with increasing polarity of the solute,

broadly opposite to that in reversed phase. Nevertheless,

there are considerable differences in the selectivity of the

techniques, indicating their complementary nature and

orthogonality, which is also an advantage for two-dimensional

(2D) separations. Polar solutes are retained in HILIC that have

little or no retention in reversed phase; for example uracil can

show good retention in HILIC, whereas it is used as a void

volume marker in reversed-phase chromatography.

There are many advantages of HILIC over reversed-phase

chromatography; indeed, for some polar neutral solutes,

there is hardly an alternative to HILIC to achieve sufficient

retention for separation. However, there are real and perceived

disadvantages of HILIC that can provide a barrier to more

widespread uptake of the technique. The aim of this paper is to

provide an update on the mechanism of separation of HILIC,

to discuss the manipulation of its selectivity, its advantages

and limitations (and how they may be overcome), together with

some new applications of the technique.

Which Solutes are Suitable for HILIC?Polar and ionized solutes that are hydrophilic are likely to be

retained in HILIC; as a guide, the log of the solute distribution

ratio between octanol and water can be considered where:

Log Dow = log {[neutral + ionized solute]octanol ] /

[neutral + ionized solute]water} [1]

Log Dow values can be measured experimentally, or

obtained from a number of commercial simulation software

packages. Hydrophobic solutes have a high positive log

D value, that is, they prefer to partition into the relatively

nonpolar (octanol) in a shake flask experiment (this simulates

the bulk mobile phase in HILIC), and thus they are less

retained. Conversely, very hydrophilic solutes have a large

negative value (prefer to partition into water) and thus are

more retained. Typically, solutes with a value of log D > ~+1

will show low retention in HILIC. However, ionic effects can

contribute to retention; for example, the protonated base

nortriptyline (log D ~+1) gives high retention on a silica

column using aqueous acetonitrile buffered at ww pH 3

because of interaction with negatively charged silanols

(2). Figure 1 indicates a rough correlation on three HILIC

stationary phases between log Dow and retention for some

acidic, basic, and neutral solutes. The correlation is best

for TSK amide (r = 0.83), which has a thick neutral polymer

layer that shields stationary phase silanol groups (negatively

charged) from ionic retention of protonated bases or

repulsion of charged acids. The bare silica column shows no

such shielding from ionic effects, resulting in a much poorer

correlation (r = 0.42). Clearly, the retention of neutrals by

partition should depend on the extent of the water-rich layer

Hydrophilic Interaction Liquid

Chromatography: An UpdateDavid V. McCalley, Centre for Research in Biosciences, University of the West of England, Frenchay, Bristol, UK

This article is an update on the technique of hydrophilic interaction liquid chromatography (HILIC) and covers recent ideas on the mechanism of separation, and how it may be manipulated to suit the separation of particular sample types. The advantages of HILIC are discussed, and also the actual and perceived disadvantages of the technique and how the latter can be overcome. Some new applications of HILIC for characterization of biopharmaceuticals, where it can even be applied to the separation of intact proteins, and to applications in metabolomics, will be discussed.

KEY POINTS• HILIC is a valuable and complementary alternative to

reversed phase, especially for separation of polar and

ionized solutes.

• HILIC can be applied to the separation of small

molecules, but also to peptides and even large

proteins, such as monoclonal antibodies.

• HILIC has some advantages over reversed-phase,

such as the ease of coupling of detectors that utilize

solvent evaporation, and low column back pressure.

• Some tips are given on how to overcome

shortcomings of HILIC, such as longer column

equilibration times.

Ph

oto

Cre

dit: A

rgu

s/s

toc

k.a

do

be.c

om

LC•GC Europe March 2019114

on the surface, while ionic retention–repulsion and adsorption

effects will also depend on the particular stationary phase

used. These factors contribute to the substantial selectivity

obtainable by changing the stationary phase in HILIC (see

below).

Whereas HILIC has classically been applied to small

molecules, it has recently been successfully applied to the

separation of peptides and even intact proteins (see below).

The Mobile Phase in HILICOrganic Solvent Concentration: The ratio of organic

solvent to water in the mobile phase is a crucial factor in

controlling overall retention in HILIC. Reducing the water (the

strong solvent) concentration increases retention, which is

the opposite effect to that in reversed phase. Retention can

increase exponentially at high levels of acetonitrile (80–95%)

(3). Although other water-miscible polar organic solvents have

also been used (such as methanol), acetonitrile is by far the

most commonly employed, and usually in the concentration

range ~ 60–95%. As the water concentration increases in

the mobile phase, polar solutes increasingly partition into it,

reducing retention.

Buffers and Mobile Phase pH:

Approaches to pH Measurement: Buffers are necessary to

stabilize the charge both on the solute and the stationary

phase. The choice of buffer is relatively restricted as a result

of solubility problems in high concentrations of acetonitrile;

ammonium formate (AF, ww pKa = 3.75) and ammonium

acetate (AA, ww pKa = 4.75) are commonly used. In addition

to favourable solubility, these buffer salts are volatile and

thus useful for mass spectrometry (MS) detection. High

concentrations of acetonitrile do, unfortunately, influence

pH measurement. Typically, pH is measured in the aqueous

component of the mobile phase (ww pH), but is more

informatively measured after addition of the organic solvent,

directly in the final mixture (ws pH). This measurement (by use

of published delta “correction” factors) can be related to the

true thermodynamic pH of the solution (ss pH):

1

0

-1

-2-4 -3 -2 -1 0 1 2

-4 -3 -2 -1 0 1 2

-4 -3 -2 -1 0 1 2

Atlantis silicar = 0.42

Zwitterionicr = 0.65

TSK amider = 0.83

Log D (AVG)

Log D (AVG)

Log D (AVG)

log

klo

g k

log

k

0.5

-0.5

-1.5

1

0

-1

-2

0.5

-0.5

-1.5

1

0

-1

-2

0.5

-0.5

-1.5

Figure 1: Plots of log k versus average log D (from three calculation programs) on three stationary phases. Mobile phase 5-mM ammonium formate pH 3.0 in 85% acetonitrile. Blue circles = basic solutes; black squares = acidic solutes; red triangles = neutrals. Adapted with permission from reference 2.

115www.chromatographyonline.com

McCalley

ss pH = w

s pH- δ [2]

The ss pH ultimately determines the retention properties of

the solute in the system. Correspondingly, the ionization of

buffers is also governed by their ss pKa values. Unfortunately,

these values are rarely available, leading to the widespread

adherence to the (less correct and less informative) ww pH

and ww pKa values. This difficulty can be considered a

disadvantage of the HILIC technique because it complicates

interpretation and prediction of retention.

Effect of Buffer pH: McCalley studied the use of volatile AF,

AA, and ammonium bicarbonate buffers at ww pH 3.0, 4.4,

6.0, and 9.0 for the analysis of acidic, neutral, and basic

solutes on a relatively inert amide column, containing a low

concentration of acidic silanols and a neutral bonded ligand

(see Figure 2). The neutral solutes, thiourea (peak 3) and

uracil (peak 4), showed little variation in k over the entire pH

range. The bases, procainamide and nortriptyline (peaks 5

and 6), gave increased retention as the pH was increased

from 3 to 6. It is possible that increased silanol ionization

over this range produces increased ionic retention of these

basic solutes. At pH 9, however, their retention dropped,

probably as a result of suppression of solute ionization

and thus diminution of ionic attraction. The strong sulfonic

acids (peaks 1 and 2) showed little retention, which further

decreased as the pH was raised from 3 to 9, attributable to

increased solute repulsion of these fully ionized acids from

increasingly ionized silanols as the pH was raised. The weak

acid 4-hydroxybenzoic acid (peak 7) showed increased

retention as the pH was raised from 3 to 6. The increase in

pH causes increased solute hydrophilicity, reflected in a

decreased log D value as the solute was increasingly ionized.

It may also encourage hydrogen bonding between ionized

acidic solute and neutral polar column groups. However, at

pH 6 the acid was mostly ionized and repulsion effects from

increasingly ionized silanols dominated, causing reduced

solute retention at pH 9. The retention of many weak acids on

silica-based columns can be explained by a balance of the

opposing effects of increased ionization and hydrophilicity

against increased repulsion as the pH is raised (4).

Changing the mobile phase pH is a powerful means of

adjusting the selectivity in a HILIC separation even when

restricting the choice to volatile “mass spectrometer-friendly”

buffer systems.

5 mM AF pH 3.0

7 2

4

4

4neutralsstrong bases3

3,4

3

3

3

4

8

8

6

5

5

5

6

6

6

0 5 10 15 20 25 30 Time (min)

5

8

8

5,6

1

1

1

1+

2

2

2

7

7

71,2,7 acids

5 mM AF pH 4.4

5 mM AA pH 6.0

5 mM AB pH 9.0

Figure 2: Chromatogram of a mixture of acidic, basic, and neutral test compounds on a BEH amide column using 95% acetonitrile containing 5-mM ammonium formate buffer w

w pH 3 and 4.4, ammonium acetate buffer pH 6.0, and ammonium bicarbonate buffer pH 9.0. Peak identities as Figure 3. Adapted with permission from reference 4.

LC•GC Europe March 2019116

McCalley

NovaFFF SoftwareNovaFFF Software

ICP-MSICP-MSRIRIUVUVDLSDLSMALSMALS

The FFF - MALS Platform Next Level Nano, Bio and Polymer Analysis

Contact us for more information: www.postnova.com

NEW

Electrical Asymmetrical

Flow FFF Option!

Effect of Buffer Concentration: An increase in volatile buffer

concentration of AA or AF tends to increase the retention of

neutral compounds. This result may be a result of the salt

increasing the volume of water in the stationary phase layer,

a selective process that may depend on the nature of the

salt. For basic compounds, retention decreases with

increasing buffer concentration on silica-based columns,

as a result of the competitive interaction of the buffer cation

with ionized stationary phase silanols. Conversely, the

insulating effect of buffer ions on charged silanols results in

reduced repulsion from the column and a higher retention

of acidic solutes. Clearly, ionic interaction effects (both

attractive and repulsive) could be minimized—or in

some cases even eliminated—by the use of high buffer

concentrations, although their deliberate suppression

is questionable because they can give rise to useful

selectivity effects. In addition, high buffer concentrations

are undesirable for use in liquid chromatography (LC)–MS

because they can suppress solute ionization and thus reduce

detection sensitivity (3).

Alternative Buffers to AA and AF: Simple organic acid

additives, such as 0.1% formic and acetic acid, have been

employed instead of salt buffers because they give reduced

ion suppression effects in MS; unfortunately, they can give

poor peak shapes for some acidic and basic solutes. The

inferior peak shapes may be a result of the very low ionic

strength of simple acids, especially in high concentrations of

acetonitrile, causing overloading effects even at relatively low

solute concentrations (5). These effects may be in addition to

the lack of deactivating effect of the ammonium ion (present

in the usual salt buffers) on ionized silanols. Stronger acids

(for example, 0.1% trifluoroacetic acid [TFA]) give access to

lower ws pH values while retaining sufficient ionic strength in

high concentrations of acetonitrile to yield satisfactory peak

shapes (6). TFA may also improve peak shape through ion

pairing effects. TFA gives unusually increased retention of

strongly acidic solutes on silica-based columns partially as a

result of reduction of silanol repulsion. In addition, the column

surface may become positively charged, attracting anionic

solutes (see Figure 3).

Alpert explored a much more diverse series of buffer

compounds, including nonvolatile salts that are not suitable

for mass spectrometry (7). In particular, he aimed to study

the effects on retention of kosmotropic ions, such as sulfate,

which are well hydrated, and chaotropic ions, such as

perchlorate, which are poorly hydrated. The former was

found to promote partitioning of charged solutes into the

immobilized aqueous layer. The effects on neutral solutes

were more modest; retention times were unchanged or

increased slightly with an increase in concentration of any

salt. Concentrations of salt ranged from 5–120 mM at ww pH

values of both 3 and 6.

5 6 7

1

2

A215

0.1% TFA

AF pH 3

0 4 8 12Time (min)

7

2

1

5

6 8

4

3

43

8

Figure 3: Chromatograms of acidic, basic, and neutral solutes on an glycan (amide shell) column (Agilent) using 95% acetonitrile containing 0.1% TFA or 5-mM ammonium formate pH 3.0. Peak identities: 1 = p-xylenesulfonic acid; 2 = 2-naphthalene-2-sulfonic acid; 3 = thiourea; 4 = uracil; 5 = nortriptyline; 6 = procainamide; 7 = 4-hydroxybenzoic acid; 8 = cytosine. Adapted with permission from reference 6.

LC•GC Europe March 2019118

McCalley

The Stationary PhaseStationary phases can be divided broadly into those that

have an essentially neutral surface, for example, amide

or diol; those that have an acidic surface, for example,

poly(2-sulfoethyl), which possesses sulfonic acid groups;

and those that are basic, for example, those with alkyl amino

groups. There are also other categories, such as zwitterionic

columns, that can behave in a somewhat similar way to

neutral columns because of the proximity of negatively and

positively charged ligands (but with some superimposed

ionic retention behaviour). Bare silica columns are another

distinct group that show acidic properties because of the

presence of negatively charged silanol groups, particularly

at higher pH. Principal component analysis (8) or correlation

analysis considering retention data for different probe solutes

has demonstrated that columns in these various categories

show distinct retention behaviour.

The retention of neutral solutes is influenced by partitioning

into the water layer on the column surface and it is therefore

hardly surprising that their retention is affected by the

thickness of this layer, which varies on different columns.

Commercially available polymeric zwitterionic phases

have an extensive water layer, which may result from

their thick layers of bonded polymeric stationary phase

ligands. In comparison, diol phases and silica hydride have

thinner water layers. Adsorption may be a more important

mechanism here, especially on the latter phase. Neutral

solute retention is affected by water layer thickness and can

be assessed by measuring the hydrophilic selectivity of the

phase αOH (9):

αOH = k uridine/k deoxyuridine [3]

Dexyuridine has one less –OH group than uridine, resulting

in less hydrophilicity and reduced retention compared with

uridine. Measurements of this parameter show a broad

correlation with estimates of water layer thickness (2). For

example, using 5-mM AF pH 3.0 in 95% acetonitrile gave αOH

as 1.6, 2.3, and 3.0 for a bare silica, amide, and zwitterionic

phase, respectively. Clearly, the zwitterionic phase should

give the greatest retention of neutral solutes.

The propensity of a phase to retain cationic solutes over

neutral solutes can be assessed by measuring its cationic

selectivity α CXT:

α CXT = k TMPAC/k uridine [4]

where TMPAC is trimethylphenylammonium chloride, a

quaternary salt that is ionized under all conditions. Using

5-mM AF in 85% acetonitrile, silica, zwitterionic, diol,

UV215

nm

3

3

3

3

3

3

0 7.5Time (min)

2

2

2

2

2

2

ZIC-cHILIC (phosphorylcholine)

BEH silica

Cortecs silica

Agilent glycan

BEH amide

ZIC-HILIC (sulfobetaine)

1

1

1

1

1

1

Figure 4: Selectivity of different columns using mobile phase 5 mM ammonium formate pH 4.4 in 95% acetonitrile. Peak identities: 1 =  uridine; 2 = 4-OH benzoic acid; 3 = nortriptyline. Adapted with permission from reference 10.

119www.chromatographyonline.com

McCalley

and amide columns gave values of 7.1, 1.0, 0.6, and 0.5,

respectively, for this parameter, demonstrating considerable

preferential retention of cationic solutes on bare silica.

Similarly, the anionic selectivity of columns can be measured

from:

α AS = k BSA/k uridine [5]

where BSA is the strong acid benzenesulfonic acid. Once

again when using 5-mM AF in 85% acetonitrile, amino and

amide columns gave values of 6.6 and 0.33, respectively,

demonstrating the considerably increased selectivity for

acids of the amino phase (4).

Changing the stationary phase is probably the most

effective way to change the selectivity of the separation

(2), as shown in Figure 4 for neutral uridine, the acid

4 –hydroxybenzoic acid, and the base nortriptyline

(10). Both bare silicas show preferential retention of

nortriptyline because of attractive interactions with

negatively charged silanols. The low pore occupancy of

water in silica columns (7–9% in 89% acetonitrile [11])

and their poor hydrophilic selectivity explains their low

retention of uridine. Bonded phases have fewer silanols,

which may in addition be partially shielded, resulting

in low retention of nortriptyline on the zwitterionic and

amide phases. Reduced silanol interactions and good

hydrophilic selectivity on these phases may also explain

their improved retention of neutral and acidic probes.

A “toolbox” of phases of different selectivity might

first contain the neutral and quasi-neutral amide and

zwitterionic columns, which are good “general-purpose”

phases. A bare silica column or phase containing

bonded acidic groups (for example, sulfonic acid)

should be included to give good retention or separation

of bases and an amino column could be used for the

separation of acids. The substantial variation in selectivity

between different columns in HILIC is an advantage over

reversed-phase methods, where interchange of stationary

phases has a relatively smaller effect on selectivity.

Advantages of HILICAs well as the high retention of polar and ionized

compounds, the complementary nature to reversed-phase

chromatography, and the beneficial changes in selectivity

of different stationary phases, HILIC demonstrates

a number of other advantages over reversed-phase

chromatography.

Low Viscosity of the Mobile Phase: Many of

the advantages of HILIC stem from its use of high

concentrations of acetonitrile (typically 60–95%) in the

mobile phase, giving low viscosity, and allowing the use

of long columns, or fast flow with conventional columns. A

45 × 0.46 cm column packed with 2.7-μm shell particles

operated at a flow of 2.0 mL/min generated over 100,000

theoretical plates for four basic drugs in a reasonable

analysis time (<15.0 min) (12). For solutes amenable to either

10.00

9.00

8.00

7.00

6.00

5.00

4.00

3.00

2.00

1.00

0.00

Proca

inam

ide

Nort

ripty

line

Dip

henhyd

ram

ine

TMPA

C

cyto

sine

pyrid

ine

BSApXSA

2-NSA

4-OH b

enzo

ic

Basic compounds

Acidic compounds

Are

a S

/N 9

0%

Ace

ton

itri

le-b

uff

er/

10%

Ace

ton

itri

le-b

uff

er

Figure 5: Signal-to-noise ratios using flow injection analysis coupled with optimized MRM triple quadrupole mass spectrometry for 10 solutes with a HILIC mobile phase (5-mM AF pH 3.0 in 90% acetonitrile) and a reversed phase (5-mM AF pH 3.0 in 10% acetonitrile).

LC•GC Europe March 2019120

McCalley

HILIC or reversed phase (such as moderately hydrophobic

base nortriptyline), the organic-rich mobile phase used in

the former technique provides increased solute diffusion

and thus flatter van Deemter curves in the C-term region

(mass transfer). However, if the increase in solute diffusion is

factored out using reduced plots, reversed phase shows a

slight advantage in the C-term region at fast flow rates. This

means that a somewhat improved performance at high flow

rates is obtained for hydrophobic solutes using reversed

phase rather than hydrophilic solutes using HILIC (13).

However, HILIC is clearly favoured in many cases by the

possibility of longer columns (at normal or reduced flow) to

generate high efficiency.

Peak Shape of Ionogenic Compounds: Peak shapes

of basic compounds can be surprisingly good in HILIC.

The basic drugs diphenhydramine, procainamide,

and nortriptyline gave excellent peak shapes using

a bare silica column with a simple AF buffer pH 3 in

acetonitrile. This column has a high cationic selectivity; in

reversed-phase chromatography, strong ionic interactions

can be associated with poor peak shape, which is

not always true in HILIC. Good peak shapes may be

associated with higher sample capacity in HILIC, with

column efficiency being maintained at much higher solute

mass than in reversed phase (14).

Compatibility with Electrospray Ionization (ESI)-MS

and Other Evaporative Detectors: The low viscosity, high

volatility, and low surface tension of high concentrations of

acetonitrile are conducive to higher sensitivity in ESI-MS (15).

Figure 5 compares the relative signal-to-noise (S/N)

ratio for six basic and four acidic solutes by flow injection

analysis (FIA) ESI-MS using a triple quadrupole MS and

multiple reaction monitoring (MRM) conditions optimized

for each solute. On average, a sensitivity increase of ~3×

for the HILIC conditions (90% acetonitrile–10% 5 mM

AF pH 3.0) compared with reversed-phase conditions

(10% acetonitrile–90% 5 mM AF pH 3.0) was obtained.

The use of FIA avoids introduction of the samples into a

chromatographic surface, which potentially might confound

the results with adsorption effects. Gradient elution of

mixtures of solutes using an appropriate column can also

be used to assess difference in detection sensitivity. While

possible column adsorption effects may indeed confound

the results, as well as possible elution in different mobile

phase composition (dependent on the column), this method

can be used to evaluate the response of solutes in mixtures

simultaneously, as they can be separated on the column.

As FIA has no separation stage, solutes are best evaluated

individually, making the procedure more laborious. It also

arguably presents a more realistic simulation of normal

a) Trastuzumab (Herceptin®)

i) 2.0e+5

i) 1.5e+5 ii) 4.5e+5

ii) 4.5e+5

1.6e+5

1.2e+5 3.6e+5

2.7e+5

1.8e+5

9.0e+4

9.0e+4

6.0e+4

3.0e+4

0.0 0.0

3 5 7 3 5 79 11

3

Fc/2

Fc/2LC

LCFd’ Fd’

Fc/2

LC

Fd’

Fc/2

LC

Fd’

5 7 9 11

Time (min) Time (min)

Time (min) Time (min)

3.6e+5

2.7e+5

1.8e+5

9.0e+4

1.2e+5

8.0e+4

4.0e+4

0.0

b) Trastuzumab B

0.0

3 5 7

TIC

TIC

TIC

TIC

Figure 6: Middle-up analysis of (a) Trastuzumab and (b) Trastuzumab B (biosimilar). Total ion chromatograms using i) reversed-phase LC–MS and ii) HILIC–MS. Adapted with permission from reference 21, copyright 2016, American Chemical Society.

121www.chromatographyonline.com

McCalley

experimental conditions. The average gain in sensitivity was

reported as 7–10 times (16), but improved modern interface

designs showed more modest gains (17). Similar beneficial

increases in sensitivity can be obtained with other mobile

phase evaporation detectors, such as the charged aerosol

detector (18).

Disadvantages of HILIC and How They May be OvercomeSample Injection: Using injection solvents of higher

eluotropic strength than the mobile phase (that is, increased

water concentration) gives increasing deterioration in peak

shape (19). This effect can be problematic if the sample

is not soluble in high concentrations of acetonitrile (that

is, in appropriately “weak” mobile phases). The effect can

be moderated by injecting small volumes. Alternatively,

for small-molecular-weight (MW) compounds, isopropyl

alcohol (IPA) or a mixture of 50:50 acetonitrile–IPA has

been recommended. For drug discovery applications,

dimethylsulfoxide in at least 80% acetonitrile can be used,

whereas for peptide analysis, pure ethanol or IPA is possible

(20).

Use of the mobile phase or a weaker solvent as an

injection solvent to avoid peak distortion is not normally

possible with biopharmaceuticals because of limited

solubility in high concentrations of acetonitrile and

possible protein denaturation or precipitation. Therefore,

in one study, an aqueous sample was injected followed

by a fast gradient ramp incorporating a high percentage

of acetonitrile at the beginning of the method, in addition

to a small injection volume (21), which produced good

results.

Long Equilibration Times: Full isocratic equilibrium of the

column (where retention times stabilized to 99–101% of the

value at “infinite” equilibration time), with buffered acetonitrile

mobile phase, can require more than 20 min (> 40 column

volumes) when purging the 10× 0.21-cm columns at 0.5 mL/

min (10). In isocratic analysis, full equilibration is necessary

because the selectivity of the separation can change with

equilibration time. Full equilibration was found to depend on

the nature of the HILIC stationary phase, the purging flow rate,

and the original or “storage” solvent. These long equilibration

times are not, however, a barrier to the use of gradient elution

in HILIC. In gradient elution, a repeatable partial equilibrium

was demonstrated (10) in an equilibration time of as little as

5 min, implying that HILIC can be reliably used under these

conditions. Selectivity changes can occur in the separation

dependent on the particular equilibration time between

gradient runs. Therefore, this parameter must be kept constant

in a series of analyses; however, this does not appear to be a

problem when using modern HPLC instruments.

Retention Time Instability and Drift: HILIC has sometimes

been found to suffer from irreproducible or drifting retention

times. This problem is often associated with insufficient

equilibration of the column (see above), especially in

isocratic applications. However, one study found retention

time irreproducibility was associated with storage of

the (organic-rich) mobile phase while connected to the

instrument, rather than in tightly sealed bottles, which gave

excellent day-to-day repeatability of retention (22).

Automated QuEChERS Method

• High matrix clean-up due to non-dispersive method

• Also applicable for fatty and coloured matrices

• High robustness in the laboratory routine 24/7

• High degree of automation with online LC-MS

www.LCTech.de

[email protected]

McCalley

Extra care may be necessary in mobile phase preparation.

The commonly used buffer salts AF and AA are hygroscopic

and should be stored in a dessicator at room temperature

to improve reproducibility of buffer preparation. For isocratic

analysis using high concentrations of acetonitrile (low

concentrations of water), some consideration should be

given to preparing the mobile phase by premixing aqueous

and organic liquids by weight, taking into consideration the

density of the liquids. Errors might otherwise result if metering

relatively small volumes of aqueous phase using the HPLC

instrument.

In gradient analysis, particularly with high-pressure mixing

systems that use a separate pump for each solvent flow,

the use of one channel delivering small volumes (difficult to

achieve reproducibly and accurately) throughout the gradient

run should be avoided. This is especially true when the total

flow is relatively low, which is necessary with a small-diameter

column, and when a relatively shallow gradient is employed,

as is often the case with HILIC. Therefore, a gradient from

90% to 80% acetonitrile is best not devised with bottle A

containing 90% acetonitrile and bottle B 0% acetonitrile

at a total flow of 0.4 mL/min. This would result in pump B

delivering only ~0.045 mL/min, even at its maximum flow at

the end of the gradient.

Some New Applications of HILICAnalysis of Glycans “Bottom-Up” Methods: HILIC has

established itself as an essential technique for monitoring

glycans in monoclonal antibody (mAb) drugs designed to

target specific antigens. mAbs have MW ~150,000, of which

about 5% by weight can be glycans. Much analytical work

needs to be performed in the characterization of mAbs

or their biosimilars that have similar efficacy and safety

to the original drug, or biobetters, which are improved

products. Many of the original biopharmaceuticals are

coming off patent, giving scope for the development of

these substitute drugs. Glycosylation is one of the important

causes of microheterogeneity caused by post-translational

modification (PTM) that can occur, for example, during

production and storage; thus, its characterization is of great

importance to enable potential differences in the products

to be assessed.

About ten sugars are commonly found as constituents

of glycans, which may be attached to the Fc fragment of

the mAb. A glycan is a mono-, poly-, or oligosaccharide,

but typically contains ~10 monosaccharides. N -glycans

can be cleaved from the mAb by an enzyme, such as

PNGase-F. The resulting free N -glycans can be analyzed

in their native state or reacted with 2-amino benzamide

(2-AB) or procainamide to give sensitive fluorescent

derivatives. However, 2-AB derivatives are difficult to

detect by ESI-MS because of poor ionization efficiency.

An alternative derivative after PNGase-F deglycosylation,

which apparently gives both good fluorescent and

ESI-MS sensitivity, has recently been proposed (23). In

addition to releasing N -glycans from IGg Fc domains,

the proposed approach also produced complete release

of Fab domain N -glycans. Gradient elution analysis

on a wide-pore (300 Å) amide column was used in

their analysis. Complete release of N -glycans from

Fab domains was obtained using this procedure with

HILIC–MS, showing a peak of mass 148.4 kDa before

deglycosylation and 145.3 kDa after, implying the loss of

two N -glycans.

Characterization of Intact or Large Fragment Protein

Biopharmaceuticals and mAbs: Guillarme and co-workers

(24) employed a wide-pore sub-2-μm amide HILIC

column to successfully characterize intact and digested

(25–100 kDa fragment) protein biopharmaceuticals using

gradients of 65–80% acetonitrile and 0.1% TFA. The

300-Å pore size packing allowed the accommodation

of large biomolecules and fragments without resulting in

restricted diffusion. The separations were reported to be

highly orthogonal to reversed-phase LC, while the kinetic

performance remained comparable. The authors stressed

the following advantages of HILIC: i) compatibility with

MS, ii) reduced requirement of high temperatures that are

necessary in reversed-phase LC to limit undesirably strong

adsorption, and iii) the possibility of coupling columns in

series to gain extra resolving power. Applications were

shown to the analysis of six different insulins (one of the

oldest biopharmaceuticals, RMM~6000); reversed-phase

LC and HILIC were shown to be complementary, with

better separation of insulin and insulin glulisine by HILIC,

but superior separation of insulin and insulin lispro by

reversed-phase chromatography. High efficiencies were

obtained in both (isocratic) analyses. The authors compared

characterization of trastuzumab by HILIC, reversed phase,

and ion exchange, even gaining some results with intact

proteins, although reduction of disulphide bonds prior to

chromatography or partial digestion of the mAb typically

yields better results in terms of both chromatographic

123www.chromatographyonline.com

McCalley

and mass spectrometry characterization. Trastuzumab

(Herceptin) is a mAb widely used for the treatment of some

types of breast cancer. Whether these large molecules

are denatured during separation (which should still allow

for their analytical—if not their preparative—separation,

retaining their biological activity) remains to be confirmed.

HILIC–MS was used to compare originator and

biosimilar therapeutic mAbs at the intact and the so-called

“middle-up” level of analysis, again using a wide-pore

300-Å amide column (21). While “bottom-up” analysis

(where the sample is processed to give the simplest

building blocks, for example, liberated peptides or

glycopeptides) is most often used as the subsequent

analysis of these smaller molecules is easier, it loses

structural information on these complex molecules. In the

HILIC analysis, 0.1% TFA was again used as mobile phase

additive, even though it can cause some ion suppression

in MS detection. It was preferred due to its solubilizing

effect on proteins and its low pH suppression of silanol

ionization. The intact mAb was digested, then reduced to

give Fd’, light chain (LC), and Fc/2 subunits (two of each)

of about 25 kDa, containing attached intact glycans, and

then subjected to reversed-phase and HILIC analysis.

The two techniques were complementary, as can be

seen in Figure 6, which shows middle-up separations of

Trastuzumab (Herceptin) and its biosimilar Trastuzumab

B. Reversed-phase analysis gave a separation of the

three main fragments, but offered information about the

glycosylation pattern only after examination of the MS data.

In contrast, HILIC–MS on the same sample allowed for

a direct and immediate comparison of the glycosylation

profiles. Furthermore, Guillarme (25) demonstrated the use

of HILIC coupled with MS to characterize the antibody-drug

conjugate (ADC) Brentuximab vedotin, which is used in

the treatment of Hodgkin’s lymphoma. ADCs enable the

delivery of cytotoxic drugs (present in this case with an

average drug-to-antibody ratio of four) to therapeutic targets

with an antibody-directed mechanism. As with mAbs, these

materials need to be characterized because of the structural

complexity and heterogeneity. A middle-up approach

with fragment (~25 kDa) analysis using a 300-Å amide

column and a gradient of 85–73% acetonitrile with 0.08%

TFA and 0.02% formic acid was used. It was found that

HILIC analysis offered a completely complementary and

orthogonal set of information to reversed-phase analysis;

elution order was essentially the opposite to that observed

in reversed phase. Only one HILIC–MS run was necessary

to obtain highly important information on structural

microheterogeneity.

HILIC in Metabolomics: The human metabolome consists

of small molecules—generally accepted as having a MW

<1500 Da—dictated by the genes of the individual and

also the individual’s environment, and therefore it consists

of a mixture of endogenous and exogenous compounds.

Whereas nuclear magnetic resonance (NMR) spectroscopy

provides standard analytical methodology common to most

samples and does not require analytical development,

it suffers from spectral overlap and low sensitivity. Thus,

MS, which is considerably more sensitive, coupled with

separation techniques such as gas chromatography

(GC) and LC provides a valuable alternative to NMR.

LC clearly has broader application possibilities than

GC because it is amenable to nonvolatile, highly polar,

and thermally labile samples without derivatization.

The complementary use of HILIC and reversed-phase

chromatography coupled with MS expands the number

of detected analytes and provides considerably more

comprehensive analyte coverage (26). Important classes

of compound that have been analyzed by HILIC include

phospholipids, which are the main constituents of

biological membranes. They are important signalling

molecules and potential biomarkers for ovarian cancer,

diabetes mellitus, and other conditions. They consist of a

polar phosphate head group, two fatty acid chains, and

a glycerol group. HILIC eluents are more compatible with

ESI-MS than normal-phase chromatography eluents, which

are traditionally used for these solutes. Typical mobile

phases are acetonitrile containing AA or AF buffers, in

conjunction with bare silica or diol columns. Organic

acids, sugars, amino acids, nucleosides, and nucleotides,

which can be biomarkers of inherited metabolic disease,

are other examples of metabolites amenable to HILIC.

A comparative study of the analysis of 764 metabolites

using either HILIC or reversed-phase analysis showed that

HILIC methods markedly improved the coverage of polar

metabolite groups, such as phosphates or carbohydrates,

and therefore represented a worthy alternative to

reversed-phase separations (27). Zwitterionic sorbents,

such as those containing sulfobetaine groups, had a

particular broad application range. In addition, selectivity

was highly diverse among the HILIC methods investigated

(which used different stationary phases). For example,

an amide column gave good retention of nucleosides,

whereas a phophorylcholine sorbent was most appropriate

for the separation of carbohydrates.

A further advantage of HILIC over reversed

phase in metabolic studies appears to be that

glycerophospholipids, generally observed in cell and

plasma samples, tend to appear in narrow retention time

ranges instead of covering major parts of the retention

window, which is often found for reversed-phase

separations (28). These compounds give extensive ion

suppression in ESI-MS. For analysis of a broad range

of metabolites, the risk of suppression increases with

increased spreading of the retention window of the

interferents. In the same article, considerable differences

in selectivity for metabolites and matrix interferents were

shown between bare silica, zwitterionic, and amide HILIC

stationary phases. Thus, a particular HILIC column may be

the optimum for each individual application.

ConclusionsHILIC has become an indispensable technique for the

analysis of polar and ionized solutes poorly retained

by traditional reversed-phase methods. For samples

amenable to both HILIC and reversed-phase analysis,

the techniques show a complementary nature. In fact, for

such samples, the use of HILIC may be advantageous

because of the favourable coupling with MS and other

evaporative detectors, the low viscosity of the mobile

phase (allowing the use of long columns), and good peak

shapes for some basic pharmaceuticals.The mechanism

of HILIC separation, however, appears complex, which

can pose a barrier to the more widespread adoption of

LC•GC Europe March 2019124

McCalley

the technique. Nevertheless, a greater understanding of

the effect of some simple parameters could lead to more

straightforward method development. Problems such as

longer equilibration times are not a barrier to the use of

gradient methods. Some new applications, such as the

characterization of biopharmaceuticals, and its use in

metabolomics, indicate good potential for the use of the

technique in these areas, where it is complementary to

reversed-phase methodology.

References(1) A.J. Alpert, J. Chromatogr. 499, 177–196 (1990).

(2) A. Kumar, J.C. Heaton, and D.V. McCalley, J. Chromatogr. A 1276,

33–46 (2013).

(3) D.V. McCalley, J. Chromatogr. A 1523, 49–71 (2017).

(4) D.V. McCalley, J. Chromatogr. A 1534, 64–74 (2018).

(5) J.C. Heaton, J.J. Russell, T. Underwood, R. Boughtflower, and D.V.

McCalley, J. Chromatogr. A 1347, 39–48 (2014).

(6) D.V. McCalley, J. Chromatogr. A 1411, 41–49 (2015).

(7) A.J. Alpert, J. Chromatogr. A 1538, 45–53 (2018).

(8) N.P. Dinh, T. Jonsson, and K. Irgum, J. Chromatogr. A 1218,

5880–5891 (2011).

(9) Y. Kawachi, T. Ikegami, H. Takubo, Y. Ikegami, M. Miyamoto, and N.

Tanaka, J. Chromatogr. A 1218, 5903–5919 (2011).

(10) D.V. McCalley, J. Chromatogr. A 1554, 61–70 (2018).

(11) N.P. Dinh, T. Jonsson, and K. Irgum, J. Chromatogr. A 1320, 33–47

(2013).

(12) D.V. McCalley, J. Chromatogr. A 1193, 85–91 (2008).

(13) J.C. Heaton, X. Wang, W.E. Barber, S.M. Buckenmaier, and D.V.

McCalley, J. Chromatogr. A 1328, 7–15 (2014).

(14) D.V. McCalley, J. Chromatogr. A 1171, 46–55 (2007).

(15) S.R. Needham, P.R. Brown, K. Duff, and D. Bell, J. Chromatogr. A

869, 159–170 (2000).

(16) A. Periat, J. Boccard, J.L. Veuthey, S. Rudaz, and D. Guillarme, J.

Chromatogr. A 1312, 49–57 (2013).

(17) A. Periat, I. Kohler, A. Bugey, S. Bieri, F. Versace, C. Staub, and D.

Guillarme, J. Chromatogr. A 1356, 211–220 (2014).

(18) J.J. Russell, J.C. Heaton, T. Underwood, R. Boughtflower, and D.V.

McCalley, J. Chromatogr. A 1405, 72–84 (2015).

(19) J.C. Heaton and D.V. McCalley, J. Chromatogr. A 1427, 37–44

(2016).

(20) J. Ruta, S. Rudaz, D.V. McCalley, J.L. Veuthey, and D. Guillarme, J.

Chromatogr. A 1217, 8230–8240 (2010).

(21) V. D’Atri, S. Fekete, A. Beck, M. Lauber, and D. Guillarme, Anal.

Chem. 89, 2086–2092 (2017).

(22) N. Gray, J. Heaton, A. Musenga, D.A. Cowan, R.S. Plumb, and N.W.

Smith, J. Chromatogr. A 1289, 37–46 (2013).

(23) M.A. Lauber, Y.Q. Yu, D.W. Brousmiche, Z. Hua, S.M. Koza, P.

Magnelli, E. Guthrie, C.H. Taron, and K.J. Fountain, Anal. Chem. 87,

5401–5409 (2015).

(24) A. Periat, S. Fekete, A. Cusumano, J.L. Veuthey, A. Beck, M.

Lauber, and D. Guillarme, J. Chromatogr. A 1448, 81–92 (2016).

(25) V. D’Atri, S. Fekete, D. Stoll, M. Lauber, A. Beck, and D. Guillarme,

J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1080, 37–41

(2018).

(26) D.Q. Tang, L. Zou, X.X. Yin, and C.N. Ong, Mass Spectrom. Rev. 35,

574–600 (2016).

(27) S. Wernisch and A. Pennathur, Anal. Bioanal. Chem. 408,

6079–6091 (2016).

(28) A. Elmsjo, J. Haglof, M.K. Engskog, I. Erngren, M. Nestor, T.

Arvardsson, and C. Pettersson, J. Chromatogr. A 1568, 49–56

(2018).

David McCalley is Professor of Bioanalytical Science at

the University of the West of England, Bristol. He is a past

winner of the Silver Jubilee medal of the UK Chromatographic

Society. In the past three years, he has given invited lectures

in London, San Francisco, Cork, Paris, Prague, Balaton,

Sandefjord, Cannes, and Princeton. His research is directed

towards the understanding of the fundamental separation

mechanisms that occur in liquid chromatography. These

studies have included the effects of pressure on separations,

superficially porous packings, overloading, reversed-phase,

and hydrophilic interaction chromatography.

the micro-Chip Chromatography Company

Changing the ART of analytical

chromatography with

μPAC™Pillar Array Columns:

These micro-Chip columns feature

a perfectly-ordered separation bed

of free-standing pillars ensuring:

• excellent separation power

• unprecedented reproducibility

• unrivalled robustness

Enhance the data productivity

of your nano-LC/MS system

for complex biological samples.

Discover our products on

www.pharmafluidics.com

or meet us at

Pittcon (March 17-21, Philadelphia, USA)

or ABRF (March 23-26, Texas, USA)

FOLLOW US

125www.chromatographyonline.com

McCalley

LC•GC Europe March 2019126

LC TROUBLESHOOTING

One of the most common questions

practitioners of reversed-phase liquid

chromatography (LC) ask is “Which

column should I use?” There are over

1000 reversed-phase columns that are

commercially available today, and at

first glance the prospect of choosing

just one, or a few, can be daunting.

As I frequently tell my students, the

good news is that we have a lot of

good columns to choose from, but the

bad news is that we have to choose,

because it is impractical to evaluate all

of them, or even a large subset, for a

particular application. A running joke in

the community is that the best column

for an application is the one that’s

already in the laboratory drawer. In

other words, column choice is strongly

influenced by the experience of the

practitioner, and prior good or bad

experiences with particular columns.

We all have our favourites, and tend to

stick with them. A related question is

“Does the latest and greatest column

from manufacturer ABC really represent

a ‘new’ selectivity?” It is difficult

to provide a completely satisfying

general answer to this question.

However, we can leverage existing

data to give us some perspective on

new phases as they are introduced

to the marketplace and we consider

adding them to the collections in our

laboratory drawers. In this month’s

instalment of “LC Troubleshooting”,

I provide a perspective on the

characteristics of the columns

introduced to the commercial market

over the last 24 months, using the

hydrophobic subtraction (HS) model of

reversed-phase selectivity.

Essential Elements of the Hydrophobic Subtraction Model (HSM) of Reversed-Phase Selectivity The largest single collection of publicly

available data that allows comparison

of commercially available columns has

been assembled over the past 15 years

using the HS model of reversed-phase

selectivity. The theoretical foundation

of this model, and examples of its

application for choosing columns of

similar or different selectivities, have

been described in detail elsewhere

(1,2). Readers interested in learning

more about the model itself are

encouraged to consult these resources.

The central idea of the HS model

is that most of the selectivity of a

reversed-phase column, defined

here as the ratio of the retention

factor for compound X (kX) to the

retention factor of ethylbenzene (kEB),

can be accounted for by the sum of

five contributions that are related to

different aspects of physicochemical

interactions between solutes and the

stationary phase. This relationship

is shown in equation 1, where the

parameters H, S*, A, B, and C

account for properties of the stationary

phase related to the hydrophobicity

of the phase (H), interactions that

are size dependent (S*), hydrogen

bond donating (A) and accepting

(B) properties of the phase, and the

ability of the phase to participate in

electrostatic interactions with analytes

(C). The parameters η, σ, β, α, and

κ account for analyte properties that

correspond to the column properties.

For example, η accounts for the

hydrophobicity of the analyte, κ is a

measure of the charge of the analyte in

solution, and so on.

log ( ) = Hη – S* σ+ Αβ + Bα + Cκ

kx

kEB

[1]

The column parameters for

any reversed-phase column can

be determined by experiment

using a mobile phase containing

acetonitrile and phosphate buffer (see

reference 1 for details). The current

characterization protocol involves

measuring retention factors for 16

Looking for New Selectivity in Reversed-Phase Liquid Chromatography? A View of the Selectivity Landscape Over the Past Two YearsDwight R. Stoll, LC Troubleshooting Editor

The offerings of commercially available columns for reversed-phase liquid chromatography (LC) continue to expand. Are these columns similar or different compared to what is already available?

127www.chromatographyonline.com

LC TROUBLESHOOTING

probe solutes and regressing those

values against the known solute

parameters for the 16 probes. Since

the beginning of this work in the

early 2000s, over 700 commercially

available columns have been

characterized in this way, and for the

last five years the characterization work

has been done in my laboratory. These

Figure 1: Distribution of the “similarity factor”, FS, values for each of the 40 new entries into the hydrophobic subtraction model (HSM) database compared to its “nearest neighbour” in the existing set of columns.

25

0-3 3-5 5-7 7-13

Fs Value for Nearest Neighbour

Nu

mb

er

of

Ne

w E

ntr

ies

20

15

10

5

0

Table 1: Reversed-phase stationary

phase types of new entries in the

hydrophobic subtraction model (HSM)

database in the last 24 months

PhaseNumber of

Columns

C18 12

Polar Embedded 6

C8 5

C4 4

Cyano 4

Phenyl 4

AQ 2

Pentafluorophenyl

propyl2

Biphenyl 1

Total 40

Pure Chromatography

ADVANTAGESee What It Can Do for You and Your Lab

Sign up today to access Restek’s

years of chromatography knowledge at

www.restek.com/advantage

LC•GC Europe March 2019128

LC TROUBLESHOOTING

change in selectivity that is tolerable

when trying to identify columns with

similar selectivity (or “equivalent”

columns) (2).

New Entries in the HSM Database in the Last 24 Months Over the last 24 months, we have made

40 new entries for reversed-phase

columns in the publicly available

hydrophobic subtraction model

(HSM) database. A summary of the

numbers of columns in different phase

categories is shown in Table 1. Some

of these columns are truly new, both to

the database and to the commercial

market. Others have been on the market

for some time, but have only been

characterized by the HSM recently.

What’s New, and What’s Not?Using the similarity factor (FS) as a

measure of similarity of the new entries

to columns already in the database,

we can explore a variety of interesting

questions.

Question #1: How many of the new

entries constitute an addition of a “new”

selectivity to the database?

In thinking about this question, I am

defining a “new” selectivity as one for

which there is currently no equivalent

data are accessible through two free,

public websites (3,4).

With the column parameters in hand,

we can then think about how similar or

different any particular pair of columns

is. For this purpose, Snyder and Dolan

and coworkers have advocated for the

use of a “similarity factor”, FS, which

is a weighted distance between two

columns in five-dimensional selectivity

space. The relationship between FS

and the column parameters is shown in

equation 2:

where XH, XS*, etc. are the weighting

coefficients with values of 12.5,

100, 30, 143, and 83, suggested by

Snyder and coworkers (1). When FS is

calculated this way, columns for which

FS < 3 are considered “equivalent”,

meaning that they are effectively

interchangeable, and will produce

very similar chromatograms for most

applications. Several users have told

me that for many applications even

FS < 10 is sufficient to obtain very

similar separations. Of course, this

must be verified experimentally for any

particular application, but it is at least

a good guideline. Columns for which

FS > 100 are considered very different,

and looking for such columns may be

useful when intentionally assembling

a set of columns with very different

selectivities. Interpretation of FS values

between 3 and 100 depends on the

properties of solutes in a particular

mixture of interest, and the degree of

in the database. We can answer this

question by taking each new entry,

one at a time, and calculating FS for

the comparison of the new entry to all

previous entries in the database. If the

smallest FS for all of these comparisons

is greater than 3, then the new entry

constitutes a “new” selectivity. Figure 1

shows the distribution of the minimum

FS values for each new entry that result

from these calculations. We see that,

for roughly half of the new entries (21

out of 40), the new entry is “equivalent”

to a column that was already in the

database (that is, 0 < FS < 3). This

may happen as manufacturers aim to

add a column to their portfolio with a

selectivity that is similar to a phase that

has been quite popular historically.

What is perhaps most interesting

about this result is that this set of 40

new entries does not contain any

selectivities that are radically different

relative to what was already in the

database. Of the 19 “new” selectivities,

the largest minimum FS value (that is,

the FS value for the new column and it’s

“nearest neighbour”) is just 13 out of all

the new entries (this particular column

is a wide-pore C4 column, if you’re

curious). We see that the minimum FS

values for all of the other new entries

lie in the range of 3 to 7, which for

most applications will be only slightly

different relative to other selectivities in

the database (2).

Now, I think one could reasonably

argue that these results are surprising,

but in different directions. On the one

hand, we could say that it is surprising

that all of the new entries are so similar

to existing selectivities. On the other

hand, one could say that it is surprising

that nearly half of the new entries are

unique, because there are only nine

phase groups represented in Table 1.

In other words, shouldn’t a new

pentafluorophenyl (PFP) phase be very

similar to an existing PFP phase? Using

FS as a measure of similarity, both of

the PFP phases in the group of 40 new

entries turn out to be “new” selectivities.

How can this be? I actually think that

we can explain both apparent surprises

by recognizing that the selectivities that

populate each phase group are really

quite diverse. In other words, one C18

is not necessarily equivalent to the next

C18, and one PFP is not necessarily

equivalent to the next PFP, even though

we talk about them as being the “same”

chemistry. This point about the diversity

Figure 2: Distributions of the “similarity factor”, FS, values for (a) all members of the C18 phase group compared to an arbitrary member of the group chosen as a “reference” column, and (b) for members of the pentafluorophenyl (PFP) group compared to a reference column from that phase group.

120 14

12

10

8

6

4

2

0

(a) (b)

0-3

0-15

15-20

20-3

030

-40

50-6

060

-100

40-5

03-6

10-20

20-30

30-50

50-100

100-300

6-10N

um

be

r o

f C

18

Co

lum

ns

Nu

mb

er

of

PFP

Co

lum

ns

100

80

60

Fs Values F

s Values

0

20

40

Fs = √ (X

H (H

1 - H

2))2 + (XS*

(S*

– S*))2 + (XA

(A1 – A

2))2 +

1 2

+ (XB(B

1 – B

2))2 + (XC

(C1 – C

2))2

[2]

One of the most common questions practitioners of reversed-phase liquid chromatography (LC) ask is “Which column should I use?”

129www.chromatographyonline.com

LC TROUBLESHOOTING

of selectivities within a phase group has been discussed for

quite a long time, but I don’t think the extent of the diversity is

widely appreciated. Readers interested in the history of this

issue, some of the origins of the selectivity diversity, and the

consequences for method development are referred to John

Dolan’s article on the topic in this column several years ago

(5).

One way to examine this is to look at the distribution of

FS values for all columns within a particular phase group

compared to some arbitrary “reference” column within that

group. Currently, there are 344 columns in the HSM database

that are identified as “C18”, and 31 that are identified as

“PFP”. Figure 2 shows the distributions of FS values for the

members of each group relative to a randomly selected

member of the same group (that is, the “reference column”).

We see that in each case very few columns within a group

are similar enough to be “equivalent” to the reference column.

In the case of the C18s, just five columns meet that criterion,

and in the case of the PFPs the reference column itself is

unique! In other words, it is far more likely that any C18

chosen from the group will be different from another member

of the group than it is that the two will be equivalent.

Question #2: How many columns that previously had no

equivalent in the database now have at least one equivalent

due to the addition of the new entries?

The previous question was focused on the extent to which

new entries to the database reflect the addition of “new”

selectivities. But, this is not the only way to add value to

the complement of commercially available phases. New

entries that are “equivalent” to columns that previously had

no equivalent in the database are also important, because

they provide users with alternate columns in case a problem

develops with the column originally used to develop a

method (for example, if the manufacturer goes out of

business, or discontinues the product). Of the 689 columns

in the database prior to the addition of the 40 most recent

entries, 329 of those were “unique” in the sense that there

was no other column in the database with FS < 3. Of those

329, 322 still have no equivalent column, even after the

addition of the 40 new entries. In other words, seven of the

40 new entries fill a role as an “equivalent” for a column that

previously did not have one.

SummaryThe continuously changing offerings of reversed-phase

columns present both opportunities and challenges to

practitioners looking to choose a column. On one hand,

it is great to be able to choose from over 1000 different

commercially available options. On the other hand, it can be

difficult to tell whether or not new offerings add truly unique

selectivity that we have not seen before. The hydrophobic

subtraction model of reversed-phase selectivity is a tool,

supported by a freely available database of more than 700

columns, that can be used to answer a variety of questions

along these lines. Continued research on this and related

models will be important to the community as we look to

efficiently develop effective LC methods, even as the number

of commercially available columns continues to increase.

References(1) L.R. Snyder, J.W. Dolan, D.H. Marchand, and P.W. Carr, in Advances

in Chromatography (CRC Press, Boca Raton, USA, 2012), pp.

297–376.

(2) J.W. Dolan and L.R. Snyder, LCGC North Am. 34(9), 730–741 (2016).

(3) hplccolumns.org, (n.d.). http://hplccolumns.org/index.php.

(4) http://www.usp.org/usp-nf/compendial-tools/pqri-approach-column-

equiv-tool, (n.d.).

(5) J.W. Dolan, LCGC Europe 24(3), 142–148 (2011).

Dwight R. Stoll is the editor of “LC Troubleshooting”. Stoll is

a professor and co-chair of chemistry at Gustavus Adolphus

College in St. Peter, Minnesota, USA. His primary research

focus is on the development of 2D-LC for both targeted and

untargeted analyses. He has authored or coauthored more

than 50 peer-reviewed publications and three book chapters

in separation science and more than 100 conference

presentations. He is also a member of LCGC ’s editorial

advisory board. Direct correspondence to: LCGCedit@ubm.

com

InertSustainTh e new standard for your (U)HPLC Methods

for more information:www.glsciences.eu

[email protected]

The good news is that we have a lot of good columns to choose from, but the bad news is that we have to choose.

This point about the diversity of selectivities within a phase group has been discussed for quite a long time, but I don’t think the extent of the diversity is widely appreciated.

BIOPHARMACEUTICAL PERSPECTIVES

Monoclonal antibodies (mAbs)

have taken a prominent and

increasingly important position

in today’s drug development

and production, providing high

specificity and limited side effects

in the treatment of life-threatening

diseases. The production of a mAb

using mammalian cells will often

result in a heterogeneous product,

as it will undergo post-translational

modifications (PTMs) (1).

Moreover, potential degradation or

rearrangements at different stages

of the production, purification, and

storage may lead to structural variants

(1). N-glycosylation is an important

source of heterogeneity of mAbs

and most commonly occurs in the

crystallizable (Fc) domain of the heavy

chain (HC) (2). Fc glycosylation is

fundamental to provide the mAb with

the proper effector functions and

affects quality, pharmacokinetics,

immunogenicity, and potency of the

drug. Glycosylation is considered a

critical quality attribute (CQA) of the

mAb (2). Antibody variants, resulting

from asparagine and glutamine

deamidation, methionine oxidation,

C-terminal lysine truncation, or

N-terminal pyroglutamation, are other

sources of product heterogeneity (3),

and should be assessed at different

stages of the production.

Mass spectrometry (MS) has

become a primary tool for the

characterization of mAbs (4), yielding

molecular weights of the main

protein and variants, identification of

PTMs, and glycosylation patterns.

MS can be used for analysis at the

intact mAb level using a top-down

approach, or for the determination of

peptides obtained after enzymatic

digestion of the mAb (that is,

bottom-up). The choice will mainly

depend on the analytical question,

for example, relating to overall protein

heterogeneity or to specific locations

of protein modification. Intact

mAb analysis might be hindered

by poor ionization efficiencies

and lack of MS resolution. The

so-called middle-up approach

provides a practical alternative,

facilitating the characterization of

mAb heterogeneity by analyzing

mAb subunits of 25–50 kDa (5).

Before analysis, mAbs are cleaved

by reduction of the intra-chain

disulfide bridges (yielding light

chain [LC] and heavy chain [HC])

or by enzymatic cleavage using

papain or IdeS (yielding Fab and

Fc fragments). Middle-up analysis

decreases the overall complexity of

the probed molecules, resulting in a

more straightforward interpretation of

Middle-Up Characterization of the

Monoclonal Antibody Infliximab by

Capillary Zone Electrophoresis–

Mass SpectrometryKlára Michalíková1,2, Elena Dominguez-Vega3,4, Govert W. Somsen3, and Rob Haselberg3, 1Charles University, Faculty

of Pharmacy in Hradec Kralové, Department of Analytical Chemistry, Czech Republic, 2Current address: Czech Academy

of Sciences, Institute of Microbiology, Prague, Czech Republic, 3Division of Bioanalytical Chemistry, Amsterdam Institute of

Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, Netherlands, 4Current address: Center for

Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands

Capillary zone electrophoresis-electrospray ionization time-of-fl ight mass spectrometry (CZE–ESI-TOF-MS) for the characterization of the monoclonal antibody (mAb) infl iximab and its variants is presented. Infl iximab was analyzed using a middle-up approach involving either reduction or digestion with the enzyme IdeS. A multilayer capillary coating of Polybrene-dextran sulfate-Polybrene (PB-DS-PB) in combination with a background electrolyte of 40% acetic acid provided efficient separation of the obtained antibody fragments, that is, light chain (LC) and heavy chain (HC), as well as F(ab’)2 and Fc/2 parts. C-terminal lysine variants were also resolved. Recorded mass spectra of HC and Fc/2 fragments permitted assignment of 13 glycoforms and provided a quantitative profi le, with G0F the most abundant glycoform (~50%). CZE–ESI-TOF-MS represents an efficient means for the straightforward analysis of a monoclonal antibody and its proteoforms.

Ph

oto

Cre

dit: ke

nto

h/S

hu

tte

rsto

ck.c

om

LC•GC Europe March 2019130

the obtained MS data and allowing

more accurate assessment of

glycosylation profiles. It has been

applied to the MS-based assessment

of the conjugation degree of antibody

conjugates (6), and for the analysis

of degradation products in mAbs (7),

modifications in HCs or LCs (8), and

HC glycosylation (9).

When analyzing real-life samples

(which are frequently mixtures),

obtaining good-quality mass

spectra of proteins often requires

separation prior to MS detection.

Several separation techniques can be

employed for the separation of mAbs

and their subunits (10,11). Charge

heterogeneity can be assessed by

ion exchange chromatography (IEC)

and capillary isoelectric focusing

(CIEF), whereas size-exclusion

chromatography (SEC) and capillary

gel electrophoresis (CGE) can

be used for determination of size

variants. Unfortunately, the mobile

phases employed to obtain the

highest separation efficiency by

these methods often show serious

incompatibilities with MS detection.

Therefore, reversed-phase liquid

chromatography (LC) is still most

commonly used in combination with

MS for mAb characterization (12),

although reversed-phase LC may

lack the selectivity to distinguish

certain protein modifications.

Recently, hydrophilic interaction

liquid chromatography (HILIC)–

MS has been shown to be very

useful for resolving glycoforms of

biopharmaceuticals, including mAbs

(13). Capillary zone electrophoresis

(CZE), on the other hand, has shown

good potential for the separation of

closely related charge variants and

isoforms of proteins (14). In addition,

CZE has the intrinsic capacity to

produce narrow peaks for proteins

and—nowadays—can be coupled

rather easily to mass spectrometric

detection. Consequently, over the

last few years, the use of CZE–MS in

middle-up and intact mAb analysis

has increased. It has, for example,

been used to separate mAb subunits

and estimate their molecular weights

(15), to identify PTMs on the intact

and subunit level (16,17), and to make

in-depth glycosylation profiles (18).

The aim of this article is to highlight

the potential of CZE–MS for the middle-

up characterization of mAbs. Infliximab

was selected as a model compound

because it comprises extensive

variation in glycosylation as well as

HC C-terminal lysine variants. Prior to

CZE–MS analysis, infliximab was either

reduced to yield its HCs and LCs, or

digested using the proteolytic enzyme

IdeS, resulting in its F(ab)2 and Fc/2

subunits. CZE separation, CZE–MS

interfacing, and MS conditions were

carefully optimized to achieve detailed

glycoform and C-terminal lysine

profiles of the infliximab fragments.

Materials and MethodsChemicals: Polybrene

(hexadimethrine bromide, PB;

average Mw = 15,000), dextran

sulfate sodium salt (DS; average

Mw > 500,000), methanol,

propan-2-ol (IPA), ethylene glycol

(99.5%), and DL-dithiothreitol (99.0%)

were purchased from Sigma Aldrich.

Acetic acid (99.8%) and ammonium

hydroxide (25% v/v) were obtained

from Merck. A formulation of infliximab

(Remicade) comprised sucrose,

INNOVATION YOU CAN TRUST – PERFORMANCE YOU CAN RELY ON

| fused-core.com | Made in the USA

HALO® and Fused-Core® are registered trademarks of Advanced Materials Technology

You Have Unique mAbsDiscover More With Fused-Core®

BIOCLASS 1000 Å

www.chromatographyonline.com 131

sodium phosphate, and polysorbate

80. FragIT MicroSpin columns

containing immunoglobulin-degrading

enzyme of Streptococcus pyogenes

(IdeS; Genovis) were used for

mAb digestion. Ultra-pure water

was produced by a Milli-Q system

(Millipore). Background electrolyte

(BGE) was prepared daily by diluting

acetic acid with water to a final

concentration of 40% v/v (6.9 M,

~pH 2.0).

Sample Preparation: Reduction

of protein disulfide bonds was

accomplished by incubating the

infliximab formulation with 50-mM

dithiothreitol (DTT) for 10 min at

37 °C. The reaction was quenched

with acetic acid (final concentration,

1% v/v). Digestion with IdeS was

performed following the FragIT

MicroSpin (Genovis) kit instructions.

All final reaction mixtures were

centrifuged three times against six

volumes of water using Vivaspin

3 kDa MWCO filter tubes (GE

Healthcare) and applying 15000 × g

at 25 °C. The sample residue was

reconstituted in water and yielded

a final concentration of 5 mg/mL of

infliximab.

CE System: The CZE experiments

were performed on a PA 800plus

capillary electrophoresis system

(Beckman Coulter). Fused-silica

capillaries had a total length of

80 cm and 50 μm internal diameter

(i.d.) × 375 μm outside diameter

(o.d.) (Polymicro Technologies).

New capillaries were subsequently

rinsed with methanol (5 min), 1

M NaOH (30 min), and water (10

min) at 20 psi. After conditioning,

Polybrene-dextran sulfate-Polybrene

(PB-DS-PB) triple layer coating

was applied using the procedure

described elsewhere (19). Samples

were injected hydrodynamically at

1.5 psi for 5 s (~1% of the capillary

Inte

nsi

ty (

cnts

)

Time (min)

18 20

1

2

3

4

0.5

1.0

1.5

0.5

1.0

1.5

2.0

23400 23500 23600 23700 23800

Inte

nsi

ty (

x10

6cn

ts)

23433.8

23532.5

0.0

0.5

1.0

1.5

2.0

2.5

3.0

(a)

50825.4

0.5

1.0

1.5

2.0

2.5

Inte

nsi

ty (

x1

05

cnts

)

0.0

0.2

0.4

0.6

0.8

1.0

*^^

^ *** *

## *

*

* ** *

x 1

06

x 1

05

x 1

05

14 1612 22

Molecular Weight (Da)

(b)

(c)

0.050953.1

m/z 1302.9

m/z 1374.7

m/z 1378.1

50500 50600 50700 50800 50900 51000 51100 51200

Molecular Weight (Da)

*

*

Figure 2: CZE–MS of reduced infliximab. (a) EIEs for the m/z of the most intense ion observed for the LC (green), HC (black), and HCK (red) subunits; dashed lines indicate the position of the apices of the HC and HCK peaks. (b) Deconvoluted mass spectrum obtained for the peak at 18.0 min (LC). (c) Deconvoluted mass spectra obtained for the peaks at 19.0 min (HC; black) and 19.5 min (HCK; red); glycan structures of assigned HC glycoforms are indicated; symbols: •, mannose; Þ, galactose; , fucose; , N-acetylhexosamine; phosphate adducts are indicated with *; in the top spectrum HCK variants are indicated with #; in the bottom spectrum HC variants without C-terminal lysine are indicated with .̂

Time (min)

10 15 2010 110

40%

50%

30%

25%

20%

15%

10%

0

10

20

30A

bso

rban

ce (

mA

U)

Figure 1: CZE–UV of reduced infliximab using BGEs of increasing concentration of acetic acid in water (v/v).

Mass spectrometry (MS) has become a primary tool for the characterization of mAbs (4), yielding molecular weights of the main protein and variants, identifi cation of PTMs, and glycosylation patterns.

LC•GC Europe March 2019132

BIOPHARMACEUTICAL PERSPECTIVES

volume). A voltage of -30 kV was applied, and the

cartridge temperature was maintained at 25 °C. CZE–UV

measurements were conducted in a 60 cm capillary (50

cm to the detector) treated in the same way as described

earlier. The separation voltage was -25 kV and the

detection wavelength was 214 nm.

Mass Spectrometry: A micrOTOF QII

orthogonal-accelerated quadrupole time-of-flight (QTOF)

mass spectrometer (Bruker Daltonics) was used for

detection of mAb fragments. CZE–MS coupling was

achieved via a co-axial sheath-liquid interface (Agilent

Technologies). Sheath liquid consisting of 1:1 IPA and

water (v/v) was delivered by a 2.5 mL gas-tight syringe

(Hamilton) at a flow rate of 180 μL/h using a syringe

pump (KD Scientific Inc.). Electrospray ionization (ESI)

in positive mode was achieved applying a voltage of

-4.5 kV. Other settings were: nebulizer pressure, 0.4 bar;

dry gas flow rate, 4.0 L/min; dry gas temperature, 190 °C;

ion energy, 5 eV; collision energy, 10 eV; in-source

collision-induced dissociation, 0 eV. CZE–MS data were

analyzed using Bruker Daltonics Data Analysis software.

Molecular weight determinations of proteins were

performed using the “Charge Deconvolution” utility of the

software.

Results and DiscussionThe model compound infliximab is an immunoglobulin

G1 (IgG1) therapeutic antibody comprised of two

LCs (214 amino acids each) and two HCs (450 amino

acids each) interconnected by four disulfide bridges.

From the amino acid sequence of the LC and HC

(Figure S1, see supplementary material: http://www.

chromatographyonline.com/supplementary-information-

middle-characterization-monoclonal-antibody-infliximab-

capillary-zone-elec), the theoretical molecular weights and

isoelectric points of the protein subunits were derived.

CZE–MS Conditions: Reduction of protein disulfide bonds

using DTT represents a straightforward way to generate

LC (~25 kDa) and HC (~50 kDa) fragments of a mAb.

CZE-UV analysis of a reduced infliximab sample was used

for optimization of the separation conditions. A BGE of

0.5% (v/v) acetic acid (pH 3.0) was used as the starting

point, in which both the LC (pI 5.8) and HC (pI 8.2) are net

positively charged. In order to prevent protein adsorption

to the capillary inner wall, a positively charged PB-DS-PB

coating was applied, yielding an anodic electroosmotic

flow. This multilayer coating is fully MS-compatible and

can allow highly efficient protein CZE (19). Using 0.5%

acetic acid as BGE, a partial separation of the mAb

fragments was obtained; this was improved by increasing

the concentration of acetic acid. Analysis of the reduced

infliximab employing a BGE of 10% (v/v) acetic acid

resulted in two well-resolved, narrow, and symmetric peaks

(Figure 1). Further increase of the acetic acid concentration

provided an enhanced protein separation with the later

migrating peak resolving into two bands. A BGE of 40%

(v/v) acetic acid (pH 2.0) was selected for further CZE–MS

experiments. The addition of methanol, IPA, or ethylene

glycol up to 10% (v/v) led to longer protein migration times,

but did not improve the separation any further.

CZE–MS coupling was accomplished using a coaxial

sheath liquid sprayer and the reduced infliximab sample

Kromasil anniversary30 yearsof chromatography

Beyond expectations

30 years has gone since Kromasilwas first introduced to the marketand changed the performanceexpectations on preparative HPLCpacking materials.

Today, Kromasil is offering a full-range product line for analysis andproduction by UHPLC/HPLC and SFC,and stands ready to support yourfuture separations.

We are Nouryon

AkzoNobel Specialty Chemicals is nowNouryon, your partner in essentialchemistry for a sustainable future.

www.kromasil.com

www.chromatographyonline.com

was analyzed. Sheath liquids

consisting of either methanol or

IPA in a 1:1 ratio (v/v) with water

containing 0.1% acetic acid (v/v) were

considered. With both sheath liquids,

reduced infliximab yielded three main

peaks, such as those observed in

CE–UV. From the deconvoluted mass

spectra it could be concluded that

the first and second migrating peaks

were the LC and HC, respectively.

The mass obtained for the third peak

corresponded with the HC with a

C-terminal lysine (HCK). The extra

lysine added one positive charge

to the HC, resulting in an increase

of the protein migration time in the

applied CZE system. More detailed

interpretation of the obtained mass

spectra is given in the section

“Middle-Up Characterization”.

Sheath liquids containing IPA

yielded more than twofold larger

protein peak areas when compared

to sheath liquids with methanol.

Further evaluation of the sheath liquid

composition indicated that acetic

acid (0–1% v/v tested) decreased

the protein signals up to 25% and

that the IPA percentage (25–75% v/v

tested) was optimal at 50%. The flow

rate of the optimum sheath liquid

of 1:1 IPA–water (v/v) was tested

in the range 1–5 μL/min. Between

2–3.5 μL/min, stable baselines and

no change in LC and HC peak areas

were observed. Above 3.5 μL/min,

protein signals decreased because of

effluent dilution, whereas below 2 μL/

min unstable sprays were obtained.

The PB-DS-PB coating in

combination with a BGE of 40% (v/v)

acetic acid and a sheath liquid of 1:1

IPA–water (v/v) at the flow rate 3 μL/

min provided stable separation and

favourable detection conditions.

The repeatability of the method was

examined by successive CZE–MS

analyses (n = 5) of reduced infliximab.

Migration times and peak areas

of the LC, HC, and HCK subunits

were derived from the extracted-ion

electropherograms (EIEs) constructed

for the most intense ion observed in

the mass spectra of the respective

species. Relative standard deviations

(RSDs) for migration times were below

2.0%, whereas peak area RSDs did not

exceed 7.5%. Plate numbers obtained

for the LC and HC peaks were 220,000

and 90,000, respectively.

Middle-Up Characterization:

Reduced Infliximab: CZE–MS

analysis of reduced infliximab yielded

three main peaks (Figure 2[a])

of which the mass spectra are

depicted in Figures S2(a–c) (see

supplementary material: http://

www.chromatographyonline.com/

supplementary-information-middle-

characterization-monoclonal-

antibody-infliximab-capillary-

zone-elec). The deconvoluted

mass spectrum of the first peak

(Figure 2[b]) reveals a main

molecular mass of 23433.8 Da,

which is in good agreement with the

theoretical mass of the infliximab

LC when taking inter-chain disulfide

bond reduction into account.

Considering the observed mass,

the intra-chain bonds are still

intact, most probably because no

denaturing agent was used prior

to DTT addition. The minor signal

at 23532.5 Da (Figure 2[b]) can be

ascribed to a phosphate adduct

of the LC (20). For most proteins

analyzed with CZE–MS, minor

phosphate adducts were observed,

which might be related to an

ion-source contamination.

EIEs derived for the second and

third peak show a partial separation

of the two involved species

(Figure 2[a]). The deconvoluted

mass spectrum of the second peak

(Figure 2[c]) shows a main signal

at 50825.4 Da, which corresponds

to the G0F glycoform of the intact

HC (that is, no intra-chain disulfide

reduction) (Table 1). From the other

masses observed in the deconvoluted

spectrum, 12 additional glycoforms

could be assigned based on known

glycan masses (Figure 2[c] top,

Table 1). The glycoforms varied

between biantenary complex glycans

Inte

nsi

ty (

cnts

)

Time (min)

20 21

0.5

1.0

1.5

2.0

0.2

0.4

0.6

0.8

Inte

nsi

ty (

x1

04

cnts

)

98152.5

0.0

1.5

3.0

4.5

6.0

7.5

9.0

(a)

25199.4

1

2

3

4

5

Inte

nsi

ty (

x1

06

cnts

)

0

1

2

3

4

5

*

**

*

*

*

*

*

* *

x 1

06

x 1

06

x 1

06

18 1917 22

Molecular Weight (Da)

(b)

(c)

025327.5

*

m/z 2231.7

m/z 1201.0

m/z 1207.1

0.2

0.4

0.6

0.8

98000 98100 98200 98300 98400 98500

*

25000 25100 25200 25300 25400 25500 256002490024800

Molecular Weight (Da)

Figure 3: CZE–MS of IdeS-digested infliximab. (a) EIEs for the m/z of the most intense ion observed for the F(ab’)2 (green), Fc/2 (black), and Fc/2K (red) sub-units. Dashed lines indicate the position of the apices of the Fc/2 and Fc/2K peaks. (b) Deconvoluted mass spectrum obtained for the peak migrating at 18.5 min, F(ab’)2. (c) Deconvoluted mass spectra obtained for the peaks at 19.7 min (Fc/2; black) and 20.2 min (Fc/2K; red); glycan structures of assigned Fc/2 glycoforms are indicated; symbols: see Figure 2; phosphate adducts are indicated with *.

CZE has the intrinsic capacity to produce narrow peaks for proteins and—nowadays—can be coupled rather easily to mass spectrometric detection.

LC•GC Europe March 2019134

BIOPHARMACEUTICAL PERSPECTIVES

and high-mannose-type glycans

with and without core fucosylation.

Further minor signals originated from

phosphate adducts or HCK variants

that were not fully separated from the

HC species. For the third peak, a main

mass of 50953.1 Da was obtained

(Figure 2[c] bottom), which matches

the HC G0F glycoform including a

C-terminal lysine. Indeed, for the

other 12 HC glycoforms, C-terminal

lysine variants were observed in

the deconvoluted mass spectrum

(Figure 2[c] bottom; Table S1, see

supplementary material: http://

www.chromatographyonline.com/

supplementary-information-middle-

characterization-monoclonal-antibody-

infliximab-capillary-zone-elec).

IdeS-Digested Infliximab: CZE–

MS analysis of IdeS-digested

infliximab using the same

conditions as stated above yielded

three baseline-separated peaks

(Figure 3[a]) with corresponding

mass spectra (Figure S2[d–f], see

supplementary material: http://

www.chromatographyonline.com/

supplementary-information-middle-

characterization-monoclonal-antibody-

infliximab-capillary-zone-elec). The

deconvoluted mass spectrum of the

first peak showed a main molecular

mass of 98152.5 Da (Figure 3[b]),

which agrees with the theoretical

mass of the F(ab’)2 fragment with

intact inter-chain disulfide bonds.

Some minor signals were observed,

from which one (98250 Da) could be

assigned to a phosphate adduct of the

F(ab’)2 subunit.

Deconvolution of the mass spectra

recorded for the second and third

peak yielded main molecular

masses of 25199.4 and 25327.5 Da,

respectively (Figure 3[c]). The

0

10

20

30

40

50

60

Rela

tive A

bu

nd

an

ce (

%)

Figure 4: Relative abundances of the Fc/2 (blue) and HC (red) glycoforms observed during CZE–MS analysis of IdeS-digested and reduced infliximab, respectively. Symbols: see Figure 2.

Table 1: Observed and theoretical masses of HC and Fc/2 glycoforms with assigned glycan structures

HC Fc/2

Assigned Glycan StructureaObserved

Mass (Da)

Theoretical

Mass (Da)Δ Mass (Da)

Observed

Mass (Da)

Theoretical

Mass (Da)Δ Mass (Da)

50476.3 50476.8 0.5 24850.5 24851.0 0.5

50598.0 50597.9 -0.2 24971.6 24972.1 0.5

50621.3 50623.0 1.7 24996.2 24997.2 1.0

50639.7 50639.0 -0.7 25012 25013.2 1.2

50677.9 50680.0 2.1 25053.4 25054.2 0.8

50783.9 50785.1 1.2 25158.8 25159.3 0.5

50803.0 50801.1 -1.9 25174.4 25175.3 0.9

50825.4 50826.2 0.8 25199.4 25200.4 1.0

50845.0 50842.2 -2.8 25215.7 25216.4 0.7

50950.7 50947.3 -3.4 25319.4 25321.5 2.1

50987.5 50988.3 0.8 25361.6 25362.5 0.9

51007.0 51004.3 -2.8 25378.2 25378.5 0.3

51151.3 51150.5 -0.8 25525.5 25524.7 -0.8

a. Symbols: see Figure 2.

135www.chromatographyonline.com

BIOPHARMACEUTICAL PERSPECTIVES

second peak thus comprises the Fc/2

fragment of infliximab bearing the

G0F glycan structure. The main mass

of the third peak corresponds to the

Fc/2 G0F glycoform with a C-terminal

lysine (Fc/2K). Based on the

observed masses, in addition to the

main glycoform, 12 other glycoforms

could be assigned for both the

native Fc/2 fragment (Table 1) and

the Fc/2 lysine variants (Table S1,

see supplementary material: http://

www.chromatographyonline.com/

supplementary-information-middle-

characterization-monoclonal-

antibody-infliximab-capillary-zone-

elec). The assigned glycoforms nicely

matched those observed for reduced

infliximab. The Fc/2 and Fc/2K

subunits were baseline separated, so

no Fc/2K signals were observed in

the Fc/2 spectrum and vice versa.

Considerations and Comparisons:

Good mass resolution is required

to resolve comigrating compounds.

TOF-MS provided a mass resolution

of 12,000, allowing differentiation

of mass differences of about 2.1 Da

and 4.2 Da among proteins of 25

and 50 kDa, respectively. This

resolving power is quite sufficient for

distinguishing different glycoforms

of HC or Fc subunits. However,

minor mass modifications, such

as the reduction of one disulfide

bridge (Δ = 2 Da) or deamidation

(Δ = 1 Da), cannot be resolved.

These result in broadening of the

MS signal and may contribute to the

uncertainty of the measured mass.

In the present study, observed mass

deviations from theoretical values

did not exceed 2.1 and 3.4 Da for the

Fc/2 and HC fragments, respectively.

This means that the observed

molecular masses can be reliably

assigned to the indicated glycoforms.

Indeed, the assigned structures

are in accordance with glycans

typically found for IgG1 antibodies.

The majority of the glycoforms were

previously indicated for infliximab

applying either bottom-up (21),

released glycan (22), or middle-up

(23) approaches.

From the deconvoluted mass

spectra of the HC and Fc/2,

relative intensities for the assigned

glycoforms can be derived assuming

an equimolar response per glycoform

(Figure 4). The main glycoform

(G0F) represents more than 50%

of the total signal, whereas the low

abundant species are around 0.5%.

This illustrates the excellent dynamic

range of the developed method.

When comparing the glycosylation

profiles obtained for the HC and

Fc/2 subunits, only minor differences

were observed in relative intensities

for the glycoforms, confirming that

the responses reflect glycoform

composition.

In comparison with the middle-up

approach employing reduction,

IdeS digestion exhibited increased

MS signal intensity and better CZE

separation of the subunits. The

smaller size of the Fc/2 fragments

relative to the HC fragments yield

more efficient ionization and narrower

charge state distributions, providing

higher sensitivity and better resolved

mass spectra. From the CZE point

of view, differences caused by

modifications are relatively larger

for Fc/2 fragments and therefore

induce more efficient separations.

The Fc/2 and Fc/2K fragments

were baseline separated by CZE,

whereas this was not the case

for the HC and HCK fragments.

Moreover, no glycoform separation

was obtained for the HC fragments,

whereas partial separation of Fc/2

glycoforms was observed (Figure S3,

see supplementary material: http://

www.chromatographyonline.com/

supplementary-information-middle-

characterization-monoclonal-

antibody-infliximab-capillary-zone-

elec). As the glycans are neutral,

the differences in electrophoretic

mobility of the Fc/2 glycoforms can be

ascribed to size differences. Similar

observations were made in previous

CZE studies of intact glycoproteins

(18,24).

ConclusionsCZE–MS employing a positively

charged, noncovalent coated capillary

and a sheath-liquid interface has been

evaluated for the middle-up analysis

of the antibody infliximab. The

developed method allowed for fast

screening of antibody heterogeneity,

exploiting the combined resolution

of CZE and TOF-MS for reliable

assignment of C-terminal lysine

variants and glycoforms. Independent

of the middle-up approach chosen

(that is, reduction or IdeS digestion),

separation of the subunits was

obtained under the same conditions.

Moreover, highly similar glycoprofiles

were obtained in both cases. Overall,

this work clearly highlights the

potential of the developed CZE–MS

method for the assessment of mAb

microheterogeneity.

AcknowledgementFinancial support from the European

Social Fund and the State Budget

of the Czech Republic, Project no.

CZ.1.07/2.3.00/30.0061, is gratefully

acknowledged.

References(1) X. Wang, Z. An, W. Luo, N. Xia, and Q.

Zhao, Protein Cell 9, 74–85 (2018).

(2) F. Cymer, H. Beck, A. Rohde, and D.

Reusch, Biologicals 52, 1–11 (2018).

(3) S. Fekete, A.L. Gassner, S. Rudaz, J.

Schappler, and D. Guillarme, Trends in

Analytical Chemistry 42, 74–83 (2013).

(4) H. Zhang, W. Cui, and M.L. Gross, FEBS

Letters 588, 308–317 (2014).

(5) A. Beck, F. Debaene, H. Diemer, E.

Wagner-Rousset, O. Colas, A. van

Dorsselaer, and S. Cianférani, Journal of

Mass Spectrometry 50, 285–297 (2015).

(6) E. Wagner-Rousset, M.C. Janin-Bussat,

O. Colas, M. Excoffier, D. Ayoub, J.F.

Haeuw, I. Rilatt, M. Perez, N. Corvaia, and

A. Beck, Mabs 6, 173–184 (2014).

(7) H. Lau, D. Pace, B.X. Yan, T. McGrath,

S. Smallwood, K. Patel, J. Park, S.S.

Park, and R.F. Latypov, Journal of

Chromatography B 878, 868–876 (2010).

(8) J. Zhang, H.B. Liu, and V. Katta, Journal

of Mass Spectrometry 45, 112–120 (2010).

(9) E. Wagner-Rousset, A. Bednarczyk,

M.C. Bussat, O. Colas, N. Corvaia, C.

Schaeffer, A. Van Dorsselaer, and A.

Beck, Journal of Chromatography B 872,

23–37 (2008).

(10) A.L. Capriotti, C. Cavaliere, P. Foglia,

R. Samperi, and A. Laganà, Journal of

Chromatography A 1218, 8760–8776

(2011).

(11) M. Tassi, J. De Vos, S. Chatterjee, F.

Sobott, J. Bones, and S. Eeltink, Journal

of Separation Science 41, 125–144 (2018).

(12) B. Bobály, V. D’Atri, M. Lauber, A. Beck,

D. Guillarme, and S. Fekete, Journal of

Chromatography B 1096, 1–10 (2018).

(13) E. Domínguez-Vega, S. Tengattini, C.

Peintner, J. van Angeren, C. Temporini,

R. Haselberg, G. Massolini, and G.W.

Somsen, Talanta 184, 375–381 (2018).

(14) R. Haselberg, G.J. de Jong, and G.W.

Somsen, Journal of Chromatography A

1159, 81–109 (2007).

(15) M. Han, B.M. Rock, J.T. Pearson, and

D.A. Rock, Journal of Chromatography B

1011, 24–32 (2016).

(16) A.M. Belov, L. Zang, R. Sebastiano, M.R.

Santos, D.R. Bush, B.L. Karger, and A.R.

Ivanov, Electrophoresis 39, 2069–2082

(2018).

(17) K. Jooß, J. Hühner, S. Kiessig, B.

Moritz, and C. Neusüß, Analytical and

Bioanalytical Chemistry 409, 6057–6067

(2017).

(18) R. Haselberg, Th. De Vijlder, R. Heukers,

M.J. Smit, E.P. Romijn, G.W. Somsen, and

E. Dominguez-Vega, Analytica Chimica

Acta 1044, 181–190 (2018).

LC•GC Europe March 2019136

BIOPHARMACEUTICAL PERSPECTIVES

(19) R. Haselberg, G.J. de Jong, and G.W.

Somsen, Analytica Chimica Acta 678,

128–134 (2010).

(20) S.K. Chowdhury, V. Katta, R. Beavis,

and B.T. Chait, Journal of the American

Society for Mass Spectrometry 1,

382–388 (1990).

(21) J. Stadlmann, M. Pabst, D. Kolarich, R.

Kunert, and F. Altmann, Proteomics 8,

2858–2871 (2008).

(22) C. Lee, M. Jeong, J.J. Lee, S. Seo, S.C.

Cho, W. Zhang, and O. Jaquez, mAbs 9,

968–977 (2017).

(23) M. Tsuda, Y. Otani, A. Yonezawa, S.

Masui, Y. Ikemi, M. Denda, Y. Sato,

S. Nakagawa, T. Omura, S. Imai,

T. Nakagawa, M. Hayakari, and K.

Matsubara, Biological and Pharmaceutical

Bulletin 41, 1716–1721 (2018).

(24) R. Haselberg, G.J. de Jong, and G.W.

Somsen, Analytical Chemistry 85,

2289–2296 (2013).

Klára Michalíková works at the

Laboratory of Environmental

Biotechnology of the Academy of

Sciences, Czech Republic, focusing

on emerging organic pollutants

decomposition in the environment

using microorganisms. Prior to this,

she gained extensive knowledge in the

field of capillary electrophoresis and its

hyphenation with mass spectrometry

while obtaining her Ph.D. at the Faculty

of Pharmacy of Charles University,

Czech Republic.

Elena Dominguez Vega received

her Ph.D. degree at the University of

Alcala, Spain. Afterwards, she joined

the group of Professor G.W. Somsen

(Vrije Universiteit Amsterdam) where

she worked on the development of new

electrophoretic technologies to study

protein heterogeneity, conformation, and

affinity. Subsequently, she joined the

Center for Proteomic and Metabolomics

(Leiden University Medical Center)

as senior researcher with a focus

on MS-based methodologies to

characterize intact proteins of

pharmaceutical and biomedical interest.

Govert W. Somsen is a full professor

of biomolecular analysis and analytical

chemistry at the Vrije Universiteit

Amsterdam, Netherlands. His

current research interests include the

compositional and conformational

characterization of intact

biomacromolecules, and the bioactivity

screening of compounds in complex

samples. His group have built up an

internationally recognized expertise

in the hyphenation of separation

techniques with mass spectrometry

and optical spectroscopy for the

analysis of intact proteins.

Rob Haselberg is an assistant

professor at the Vrije Universiteit

Amsterdam where he focuses

on the characterization of intact

(bio)macromolecules, such as

biopharmaceuticals and industrial

polymers. For this, he develops and

applies novel analytical approaches

based on liquid chromatography,

capillary electrophoresis, mass

spectrometry, and optical detection.

Koen Sandra is the editor of

“Biopharmaceutical Perspectives”. He

is the Scientific Director at the Research

Institute for Chromatography (RIC,

Kortrijk, Belgium) and R&D Director at

anaRIC biologics (Ghent, Belgium). He

is also a member of LCGC Europe’s

editorial advisory board. Direct

correspondence about this column to

the editor-in-chief, Alasdair Matheson,

at [email protected]

137www.chromatographyonline.com

BIOPHARMACEUTICAL PERSPECTIVES

The life science business of Merck operates as MilliporeSigma in the U.S. and Canada.

Merck, the vibrant M, Sigma-Aldrich, Sigma, Millipore, Ascentis, Astec, Cerilliant, Carbosieve, Certipur, Supelco and TraceCERT are trademarks of Merck KGaA, Darmstadt, Germany or its DI¿OLDWHV��$OO�RWKHU�WUDGHPDUNV�DUH�WKH�SURSHUW\�RI�WKHLU�UHVSHFWLYH�RZQHUV��'HWDLOHG�LQIRUPDWLRQ�RQ�WUDGHPDUNV�LV�DYDLODEOH�YLD�SXEOLFO\�DFFHVVLEOH�UHVRXUFHV�

�������0HUFN�.*D$��'DUPVWDGW��*HUPDQ\�DQG�RU�LWV�DI¿OLDWHV��$OO�5LJKWV�5HVHUYHG�

/LW��1R��0.B$'����(1�����������

IMAGINE THE NEXT 350 YEARS

Supelco®�DQDO\WLFDO�SURGXFWV�IURP�0HUFN��$FFXUDWH��3UHFLVH��&RQVLVWHQW��

:H�NQRZ�RQO\�WKH�PRVW�DFFXUDWH�DQDO\WLFDO�SURGXFWV�ZLOO�GR��7KDW¶V�ZK\�0HUFN�RIIHUV�WKH�6XSHOFR®

SRUWIROLR�RI�DQDO\WLFDO�SURGXFWV��:KDWHYHU�\RXU�QHHGV��RXU�SRUWIROLR�LV�DOZD\V�¿W�IRU�SXUSRVH��

HQVXULQJ�\RXU�UHVXOWV�DUH�DFFXUDWH��SUHFLVH�DQG�UHSURGXFLEOH��DQG�\RXU�V\VWHPV�IXOO\�FHUWL¿HG�

Our comprehensive portfolio, developed by analytical chemists for analytical chemists, covers a

broad range of analytical solutions, and every product is meticulously quality-controlled to maintain

WKH�LQWHJULW\�RI�\RXU�WHVWLQJ�SURWRFROV��$QG�ZLWK�6XSHOFR® scientists dedicated to your analytical

DSSOLFDWLRQV��WKH�H[SHUWLVH�\RX�QHHG�LV�DOZD\V�RQ�KDQG�

SigmaAldrich.com/Supelco

Merck beginning

ZLWK�D�SKDUPDF\

in Darmstadt,

Germany

16681850

1951

1975

1966

19351827

Establishment of

partnership EMD

Aldrich Chemical

&RPSDQ\�)RXQGHG

Merger to form

Sigma-Aldrich

Corporation

Supelco

)RXQGHG

Sigma Chemical

&RPSDQ\�)RXQGHGTransformation

into a research-based

company

1993

Supelco Acquired

by Sigma-Aldrich

1995Establishment of

Merck KGaA and

public listing

Footprint of Supelco

��������(QWHUV�*&�EXVLQHVV�ZLWK�DGVRUEHQWV�DQG�SDFNHG�*&�FROXPQV

������(QWHUV�FDUERQ�DGVRUEHQW�EXVLQHVV�ZLWK�&DUERVLHYH®

��������(QWHUV�+3/&�ZLWK�683(/&26,/��OLQH�RI�VWDEOH����P�VSKHULFDO�SDUWLFOHV�ZLWK�WUXH�PRQROD\HU�

bonding

��������(QWHUV�WKH�DLU�VDPSOLQJ�PDUNHW�E\�LQWURGXFLQJ�D�OLQH�RI�VROYHQW�GHVRUSWLRQ�WXEHV�IRU�

LQGXVWULDO�K\JLHQLVWV�WR�KHOS�SURWHFW�ZRUNHUV�IURP�EHLQJ�H[SRVHG�WR�WR[LF�FKHPLFDOV

�������(QWHUV�VDPSOH�SUHSDUDWLRQ�EXVLQHVV�ZLWK�6XSHOFOHDQ��63(�WXEHV

������/DXQFKHV�630(�¿EHUV

������,62������±�4XDOLW\�0DQDJHPHQW�6\VWHP

�������6LJPD�$OGULFK�DFTXLUHV�$VWHF®��OHDGHU�LQ�FKLUDO�FKURPDWRJUDSK\

��������)LUVW�WR�PDUNHW�JOREDOO\�)XVHG�&RUH®�SDUWLFOHV�MRLQWO\�ZLWK�$GYDQFHG�0DWHULDOV�7HFKQRORJ\��

introduced Ascentis® Express

�������6XSHOFR® expanded to include all analytical products from Merck KGaA for a comprehensive

range of analytical techniques

Founders of SupelcoDr. Walter Supina

%RUQ�LQ�+DUWIRUG��&RQQHFWLFXW��:DOW�

obtained his doctorate in chemical

HQJLQHHULQJ�LQ�����

Mr. Nicholas Pelick

%RUQ�LQ�6FUDQWRQ��3HQQV\OYDQLD��

1LFN�REWDLQHG�KLV�PDVWHU¶V�GHJUHH�

LQ�ELRFKHPLVWU\�LQ�����

2015

2010

Acquisition

of Millipore

Acquisition of

Sigma-Aldrich

Reference materials

for accurate results

Chemical Standards and

&HUWL¿HG�5HIHUHQFH�0DWHULDOV

Cerilliant®, Certipur®, TraceCERT®

$QDO\VLV�ZLWK�mobility

and ease-of-use

Comprehensive

analytical portfolio

6DPSOH�3UHSDUDWLRQ�

Chromatography,

Titration,

Spectroscopy,

3KRWRPHWU\

Analytical test kits

and test strips

LC•GC Europe March 2019140

COLUMN WATCH

It has been observed over recent

years that 1.0-mm internal diameter

(i.d.) columns continue to be

introduced by manufacturers. The

usage of 1.0-mm i.d. columns in

the laboratory, however, seems

to be scarce. A rudimentary scan

of research articles employing

these smaller-bore columns initially

supported this hunch, and prompted

a more intensive investigation into

the utility of 1.0-mm i.d. columns.

What follows is a synopsis of these

findings.

TerminologyThe terminology surrounding column

geometries is often confusing. Terms

such as “nanobore” and “microbore”

are often used to describe a number

of different formats. For example,

Majors referred to columns having

an internal diameter greater than

3 mm (up to and including 4.6 mm) as

“conventional” or “standard” columns.

The term “microbore” is associated

with 2.1-mm internal diameters.

The term “narrow-bore” is used for

columns of greater than 1.0 but less

than 2-mm i.d., and “capillary” is

employed to describe columns with

internal diameters less than 1.0 mm

(1). In a 2003 article, Gerard Rozing

proposed that 4 mm to 5 mm i.d.

columns be termed “normal-bore”,

2.1 mm < i.d. < 4 mm as “narrow- or

small-bore”, 1.0 mm ≤ i.d. ≤ 2 mm as

“microbore”, 100 μm < i.d. < 1.0 mm

as “capillary”, and “nanobore” for

columns less than 100 μm (2). In

addition, terms describing flow rate,

including nanoflow and microflow,

are often mixed into the column

descriptions. To this author’s

knowledge, there has been no

formal standardization of terminology

relating to internal diameters of liquid

chromatography (LC) columns. For

clarity, actual internal diameters or

specified ranges of internal diameters

are used throughout this work when

describing column geometries.

Manufacturing TrendsTable 1 shows a count of the reported

new product lines introduced based

on LCGC manufacturer surveys over

the past five years (3–7). The data

are first broken down into product

lines intended for reversed-phase

chromatography, as this tends to be

the category most widely used. Within

the reversed-phase category, the

number of product lines that contained

the release of 1.0-mm formats was

counted and presented (authors note:

inclusion was based on data provided

by vendors at the time of submission).

As smaller internal diameter columns

are often associated with the needs of

large molecule separations, columns

designated for large molecule

analyses (peptides, proteins, and

antibodies) are separately counted,

along with the number of these lines

that included 1.0-mm diameters.

The data indicate that the number of

product line introductions targeted

towards reversed-phase separations

has largely stayed constant over the

past five years. The number of product

lines reported to include 1.0-mm

columns, however, seems to have

decreased from four entries in 2014 to

no more than two (2017) in the years

following. Although there have been

nearly the same number of overall

product line introductions geared

towards large molecule separations,

the downwards trend of including

1.0-mm i.d. columns parallels that of

reversed-phase introductions. This

was a surprising result, as smaller

bore columns are often associated

with the needs of large molecule

analysis.

Perspectives on the Adoption and Utility of 1.0-mm Internal Diameter Liquid Chromatography ColumnsDavid S. Bell, Column Watch Editor

One millimetre internal diameter liquid chromatography columns are available from many manufacturers. In this article, the utility of 1.0-mm internal diameter (i.d.) columns, and the arenas in which they play a relatively strong role, are investigated. Further, the advantages and disadvantages of 1.0 mm diameter columns are contrasted with both larger- and smaller-bore formats

The terminology surrounding column geometries is often confusing. Terms such as “nanobore” and “microbore” are often used to describe a number of different formats.

141www.chromatographyonline.com

COLUMN WATCH

Reviewing one major column

supplier website, 94 column

products (including guard systems)

out of 547 (17%) were described

as having 1.0 mm diameters. The

data suggest that column vendors

are paying less attention to 1.0-mm

i.d. column formats, but still retain a

considerable portfolio of 1.0-mm i.d.

options.

User TrendsIn the last high performance

liquid chromatography (HPLC)

column survey published by

Ron Majors in 2012, column

dimensions below 2.1-mm i.d.

were not reported in the top five

employed column formats (1).

An increase in the popularity of

2.1-mm and 3.0-mm columns

from previous surveys (8,9)

was speculated as being due

to the increase in use of mass

spectrometric (MS) detection

in conjunction with liquid

chromatography (LC).

The data presented in Table 2

is based on surveys of end-user

purchasing habits conducted in

2007, 2009, and 2012. The values

noted are weighted averages

attempting to estimate the number

of columns used per year, per

instrument. In a relative sense,

columns based on 3 mm and

larger internal diameters have

historically dominated usage

(and continue to do so). As

noted above, there has been a

significant rise in the use of 2.1-

mm i.d. columns, largely due to

adoption of LC–MS platforms.

Smaller diameters of greater than

or equal to 1.0 mm and less than

2 mm (typically 1.0 mm and herein

noted as such for simplicity)

have risen as well. Lastly, smaller

diameters than 1.0 mm appeared

to rise between 2007 and 2009,

but levelled out according to

the 2012 results. In this treatment,

the lower column usage for the

smaller two diameter classes can

be attributed to the smaller number

of available instruments suitable for

their operation (1).

In an attempt to further understand

the general trends on 1.0-mm

i.d. column usage from the user

perspective, several literature

searches were attempted. The lack

of common terminology, however,

made this task extremely difficult.

It suffices to report that finding

either fundamental or applied

papers specifically using 1.0-mm

i.d. columns proved onerous. The

user results seem to indicate that,

although the 1.0-mm column formats

are available from a number of

manufacturers, the format is not well

adopted.

Why Go Narrow?The desire for narrower columns

stems from several areas of interest.

Smaller column volumes result in less

solvent consumption, and therefore

Smaller column volumes result in less solvent consumption, and therefore may reduce cost and provide for an overall greener separation technology.

BioLC Innovations......with Incredible Reproducibility!

Discover more at www.ymc.de

• SEC for high resolved MAbs

• HIC with exceptional effi ciency

• RP-C4-Widepore with superior stability

• IEX for high recovery

Proteins AntibodiesOligonucleotidesPeptides

LC•GC Europe March 2019142

COLUMN WATCH

Loss of efficiency can also be

caused by poorly packed beds,

inadequate surface treatments, and

frit dispersion. Axial heterogeneity

in the packed bed is also a source

of dispersion. Gritti and Wahab

explained that, using conventional

methods, the packed bed near the

wall of the column is more dense

than that of the bulk packing, which

results in variance in analyte and

mobile phase velocity through the

column (11). For a given column

internal diameter, there exist wall

region (more dense) volumes and

bulk region (more loosely packed)

volumes. For 0.8–1.0-mm columns,

the wall region volume to bulk region

volume ratio is close to 1. This is the

worst case because it maximizes

the potential for the sample to be

impacted by the heterogeneity.

For larger diameter columns, the

wall volume becomes overall less

significant, and, for smaller column

internal diameter, the bulk region

becomes less significant.

Anspach and colleagues,

however, demonstrated that

laboratory-produced columns

with 1.0-mm internal diameters

could produce efficiencies close

to theoretical predictions (12).

Great care in the design of column

hardware (retaining frits, for example),

experimental parameters such as

injection volume, and instrument

configuration to limit extra-column

dispersion was required. Similarly,

Ma and associates determined that

80–90% of the efficiency of a 2.1-mm

i.d. column could be achieved with a

1.0-mm i.d. column for well-retained

analytes when the system was

optimized for minimal extra-column

band broadening (13). Band focusing

using weak injection solvent was

also noted as a potential means to

minimize the impact of extra-column

volume. Without modification of the

HPLC system for the smaller internal

diameter column, Wu and Bradley

may reduce cost and provide

for an overall greener separation

technology. As noted by Lestremau

and associates, reducing the use of

environmentally unfriendly solvents

through reduction in volume, rather

than replacement with greener

solvents, is often preferred from a

selectivity standpoint (10). Smaller

column internal diameters and thus

lower optimal flow rates also give

rise to more efficient desolvation

or ionization in LC–MS analyses

resulting in greater sensitivity.

The disadvantages of smaller-bore

columns lies in the limited loading

capacity and the relative impact of

extra-column band broadening in

a given chromatographic system.

The limited loading capacity may be

offset by the increase in detection

sensitivity when coupled with mass

spectrometry (MS). Extra-column

volume, however, is fixed for a given

system, and, as the column volume is

reduced, band broadening as a result

of the extra-column volume increases.

Lestremau and associates compared

the performance of commercially

available 1.0 mm × 100 mm and

2.1 mm × 100 mm columns using

a modern ultrahigh-performance

liquid chromatography (UHPLC)

system, and determined that, in

isocratic mode, approximately 67%

of the efficiency was obtained on

the 1.0-mm i.d. column versus the

2.1-mm i.d. geometry (10). The impact

of extra-column volume could be

reduced by either increasing the

1.0-mm i.d. column length (increase

the relative volume of the column),

or by converting to gradient mode.

The group reported that in the latter

mode 80% of the efficiency obtained

on the 2.1-mm i.d. column could

be achieved using the 1.0-mm i.d.

column.

Packed columns in 1.0-mm

diameter tubing do not often provide

efficiencies that are theoretically

expected, even when extra-column

dispersion is taken into account.

Table 1: Manufacturing trends—inclusion of 1-mm i.d. columns in modern product

lines

Year

Total Reversed

Phase

Product Lines

Introduced

Number

Including

1.0 mm

Total Large

Molecule

Product Lines

Introduced

Number

Including

1.0 mm

2014 13 4 10 3

2015 13 1 13 1

2016 14 0 9 0

2017 12 2 15 1

2018 9 1 9 1

Table 2: Trends in high performance liquid chromatography (HPLC) column internal

diameter usage (in millimetres)

Survey Year i.d. ≥ 3.0 mm2.0 ≤ i.d. < 3.0

mm

1.0 ≤ i.d. < 2.0

mmi.d. < 1 mm

2007 5 2.2 0.4 0.6

2009 5.8 2.9 0.7 1.3

2012 7.4 5.3 1.3 1.3

The disadvantages of smaller-bore columns lies in the limited loading capacity and the relative impact of extra-column band broadening in a given chromatographic system.

Although 1.0-mm i.d. columns are commercially available for liquid chromatography, the adoption of this format by users is relatively low.

www.chromatographyonline.com

COLUMN WATCH

143

reported a 73% loss of efficiency on a

1.0-mm i.d. column for a well retained

analyte when compared to a 4.6-mm

i.d. column of the same length (14).

Who Uses 1.0-mm i.d. Columns?As noted above, finding research

where 1.0 mm i.d. columns were

specifically utilized was difficult.

For example, in a review of

micro-UHPLC as applied to residue

and contaminant analysis, Blokland

and colleagues made no mention

of column internal diameters in the

region of 1.0 mm, yet described

much research using 2.1 mm and

sub-1.0-mm i.d. columns (15). With

the preconception that 1.0-mm i.d.

columns would be most popular in

large molecule analyses, the many

articles pored over were again found

to be either based on 2.1-mm or

sub-1.0-mm i.d. columns.

Olkowiscz and associates compared

method performance using a 1.0-mm

i.d. column (termed “LC microflow”)

versus “direct”, and a “preconcentration”

system utilizing 75-μm columns for the

quantitation of cardiac troponin T in

mouse heart (16). The group determined

that the preconcentration method

utilizing the 75-μm column provided a

significant gain in sensitivity without loss

of accuracy and precision as compared

to the 1.0-mm i.d. column setup.

One successful usage of 1.0-mm

i.d. columns discovered was

for the analysis of monoamine

neurotransmitters in microdialysis

samples (17). As expected, the group

demonstrated a significant increase

sensitivity using the 1.0-mm i.d.

column as compared to a 2.1-mm

i.d. column. Careful attention to

band broadening as a result of

extra-column volume was noted. In

addition, the group reported the

use of a weak sample injection

solvent to aid in focusing the sample

at the head of the column. In this

case, the 1.0-mm i.d. format fit the

needed sample size, sensitivity

requirements, and speed of analysis

desired.

ConclusionsAlthough 1.0-mm i.d. columns are

commercially available for liquid

chromatography, the adoption of this

format by users is relatively

low. The increase in sensitivity

afforded by the reduction in column

volume from the more popular

2.1 mm to 1.0 mm is confounded

by decreases in efficiency and

loadability. The effort needed to

reduce extra-column volumes or

compensate for instrument band

broadening to focus analytes at

the head of columns appears to

be a strong barrier. In addition, the

packing quality of commercially

available 1.0-mm i.d. columns may

be suspect due to the challenges

of wall effects. The apparent

popularity of sub-1.0-mm i.d.

columns suggests that users

are more apt to move straight to

“capillary” formats rather than deal

with 1.0-mm nuances. In some

cases, however, the 1.0-mm

i.d. format can provide the right

combination of sensitivity and

speed.

References(1) R.E. Majors, LCGC Europe 25(1), 31–39

(2012).

(2) G. Rozing, LCGC Europe 6(6A), 1–7

(2003).

(3) R.E. Majors, LCGC Europe 27(4),

195–207 (2014).

(4) M. Swartz and R.E. Majors,

LCGC Europe 28(4), 232–243 (2015).

(5) D.S. Bell, LCGC Europe 29(4), 214–224

(2016).

(6) D.S. Bell, LCGC Europe 31(4), 234–247

(2018).

(7) D.S. Bell, LCGC Europe 30(4), 196–207

(2017).

(8) R.E. Majors, LCGC North Am. 27(11),

956–972 (2009).

(9) R.E. Majors, LCGC North Am. 25(6),

532–544 (2007).

(10) F. Lestremau, D. Wu, and R. Szücs, J.

Chromatogr. A 1217(30), 4925–4933

(2010).

(11) F. Gritti and M. Farooq Wahab,

LCGC Europe 31(2), 90–101

(2018).

(12) J.A. Anspach, T.D. Maloney, and L.A.

Colón, J. Sep. Sci. 30(8), 1207–1213

(2007).

(13) Y. Ma, A.W. Chassy, S. Miyazaki,

M. Motokawa, K. Morisato, H. Uzu,

M. Ohira, M. Furuno, K. Nakanishi, H.

Minakuchi, K. Mriziq, T. Farkas,

O. Fiehn, and N. Tanaka, J.

Chromatogr. A 1383, 47–57

(2015).

(14) N. Wu and A.C. Bradley, J. Chromatogr.

A 1261, 113–120 (2012).

(15) M.H. Blokland, H.G. Mol, and A.

Gerssen, LCGC Europe 27(s9), 13–18

(2014).

(16) M. Olkowicz, I. Rybakowska, S.

Chlopicki, and R.T. Smolenski,

Talanta, 131, 510–520 (2015).

(17) J. Van Schoors, K. Maes, Y. Van

Wanseele, K. Broeckhoven, and A.

Van Eeckhaut, J. Chromatogr. A

1427, 69–78 (2016).

David S. Bell is a director of

Research and Development

at Restek. He also serves on

the Editorial Advisory Board

for LCGC and is the Editor for

“Column Watch”. Over the past

20 years, he has worked directly

in the chromatography industry,

focusing his efforts on the design,

development, and application of

chromatographic stationary phases

to advance gas chromatography,

liquid chromatography, and

related hyphenated techniques.

His undergraduate studies in

chemistry were completed at the

State University of New York at

Plattsburgh (SUNY Plattsburgh),

USA. He received his Ph.D. in

analytical chemistry from The

Pennsylvania State University, USA,

and spent the first decade of his

career in the pharmaceutical industry

performing analytical method

development and validation using

various forms of chromatography and

electrophoresis. His main objectives

have been to create and promote

novel separation technologies and

to conduct research on molecular

interactions that contribute to

retention and selectivity in an array

of chromatographic processes.

His research results have been

presented in symposia worldwide,

and have resulted in numerous

peer-reviewed journal and

trade magazine articles. Direct

correspondence to: LCGCedit@ubm.

com

The increase in sensitivity afforded by the reduction in column volume from the more popular 2.1 mm to 1.0 mm is confounded by decreases in efficiency and loadability.

The apparent popularity of sub-1.0-mm i.d. columns suggests that users are more apt to move straight to “capillary” formats rather than deal with 1.0-mm nuances.

LC•GC Europe March 2019144

SAMPLE PREPARATION

PERSPECTIVES

The evolution of chemical extractions using sorptive phases follows an interesting history.

The evolution of chemical extractions

using sorptive phases follows an

interesting history. It likely started

with the column chromatography

cleanup procedures that we all

performed in our sophomore organic

chemistry classes and that are

widely used in the organic discipline.

This evolution eventually led to

solid-phase extraction (SPE), which

was developed and popularized

beginning in the 1970s. But the big

breakthrough was the advent of

solid-phase microextraction (SPME).

Perhaps what is most notable is

that as SPME gained acceptance,

a plethora of techniques sprouted

where the researchers seemingly

attempted to place a stationary phase

on any surface that could potentially

be exposed to a sample—inside

needles and capillaries, on vial

surfaces and lids, on stir bars, and

so on. Since the patent protection

on SPME was lifted, several new

advances have been noted. In this

instalment, we provide an overview of

some of these major advances related

to the traditional practice of SPME.

ApplicationsA series of three critical reviews

explored the application of SPME

in environmental (1), food (2), and

clinical (3) analysis. Environmental

samples like soils, sediments,

and wastewater were discussed

and thin-film and cold-fibre SPME

approaches were emphasized (1).

In vivo SPME of living animals and

plants was reviewed for environmental

and ecological studies (4). Particular

interest on quantification methods is

noted. A discussion of SPME used

for food analysis includes emphasis

of new phases, calibration studies,

and discusses the determination

of contaminants and food quality

(2). Another review of food

applications of SPME looked at

volatile, semivolatile, and nonvolatile

components from a diverse set of

food samples (5). Fibres overcoated

with polydimethylsiloxane (PDMS)

did not show the matrix interferences

common in food analysis using direct

immersion SPME (6). We will not

discuss the bioanalytical and clinical

analysis review (3) in this instalment

of “Sample Preparation Perspectives”

because it will be the topic of part 2 in

this series.

Commercial AdvancesOnly recently have multiple SPME

fibres been able to be desorbed

automatically in a single gas

chromatography (GC) sequence

without manual intervention. In

evaluating the Gerstel Multi-Fiber

Exchange (MFX) system,

modifications to the cap and potential

cooling minimized sample storage

stability, defined as less than 10%

sample loss (7). Short-term exposure

and time-weighted average data were

obtained and a new diffusive fibre

holder was investigated.

One new configuration for SPME

sampling is the Custodian SPME

syringe marketed by PerkinElmer.

Designed for field sampling, the

syringe features a ballpoint pen-style

mechanism for situations where

analyst dexterity may be limited.

Another design is the PAL (Prep

and Load) SPME Arrow (8,9). Key

features of the device are a stainless

steel fibre designed to provide a

greatly enhanced sample capacity,

as illustrated in Figure 1. Another

Recent Advances in Solid-

Phase Microextraction, Part 1:

New Tricks for an Old Dog Douglas E. Raynie, Sample Preparation Perspectives Editor

Solid-phase microextraction (SPME) was introduced nearly 30 years ago and since that time has matured into a widely used tool in the arsenal of sample preparation techniques. Simultaneously, it has spawned a host of related techniques where sorbent coatings are placed on stir bars, magnetic particles, vial walls, and so on. Over the past few years, several advances in SPME have been developed, including increasing the sorbent surface area available for extraction, accommodating direct analysis by mass spectrometry (MS), and sorbent overcoating to resist fouling by sugars, lipids, and other macromolecules present in some sample types. These advances are discussed in this month’s instalment. The use of SPME for microsampling of biological systems, so-called bio-SPME, will be the focus of Part 2.

Gas tight valves from the company

that invented them.

Valves for Gas Chromatography

TWO POSITION & MULTI-POSITION VALVES

• For injection, stream selection, and trapping

• Bores from 0.25 mm (.010”) to 4 mm (.156”)

• <GTQ�FGCF�XQNWOG�ƂVVKPIU�HQT�1/32”, 1/16”, 1/8” or 1/4” tubing

• Alloys and polymer composites to meet virtually any system requirement

• Manual, pneumatic, or electric actuation

®

www.vici.com | [email protected] | 800-367-8424

LC•GC Europe March 2019146

SAMPLE PREPARATION PERSPECTIVES

Sorptive Phase DevelopmentHou, Wang, and Guo (14) classified

current SPME phases into inorganic

coatings (metal–organic frameworks,

carbon nanotubes, graphene, and

metal and metal oxides), organic

polymer coatings (polymeric

ionic liquids and molecularly

imprinted polymers), and others

(aptamer-functionalized materials,

nanofibres) and discussed their use.

Carbon nanotubes modified with

PDMS and used as a sol-gel are

shown to have several advantages

for SPME including high porosity,

a long lifetime, and high thermal

stability (15). Wide linear ranges and

low detection limits are obtained with

these devices. Microwave-induced

plasma treatment of stainless steel

fibres led to improved adherence

of multiwalled carbon nanotubes

(16). In general, carbon nanotubes

possess high surface area and large

conjugated π-cloud systems, both

of which lead to superior adsorption

capability. This inexpensive method

provided high thermal stability,

solution resistance, and high

surface area to the SPME fibre.

As an example of metal–organic

frameworks, an aluminium-based

metal–organic framework with a

stated advantage of the SPME Arrow

is that the design minimizes coring

of the GC injector septum, allowing

a greater number of injections

before septa must be replaced.

Although improved sample capacity

is observed, the sorption–desorption

times are significantly increased if

this greater capacity is to be realized.

But the sensitivities gained with the

SPME Arrow device fall between

conventional SPME and stir-bar

sorptive extraction (SBSE).

In-Tube SPMEPerhaps the simplest alternate

form of SPME is in-tube SPME. In this

design, a section of an open-tubular

(capillary) GC column is attached

to a syringe and draw–eject cycles

are used for solute sampling and

desorption. This approach is most

widely used with subsequent liquid

chromatography (LC) analysis and

is often performed in an online

SPME-LC mode. One recent review

(10) reported that most accounts

of in-tube SPME used 250-μm i.d.

columns, though diameters ranging

from 10 to 750 μm have been used,

with lengths ranging from a few

centimetres to 80 cm. There was

no correlation between column

diameter and length, though if

in-tube SPME is combined online

with LC, it must be appropriate for

the separation column. Obviously,

the larger the surface area, the

greater the sample capacity. Solvent

desorption, rather than the thermal

desorption used in traditional

SPME, alleviates concerns with slow

desorption steps. They also report

that typical siloxane stationary

phases are used for extraction of

environmental, food, and industrial

products, while carbon-based

phases like polyethylene glycol

and molecular sieves are used

in bioanalysis, as well as with

environmental and food products.

Others, such as Fernandez-Amado

and colleagues (11), assert that the

type of capillary and coating are

the most influential parameters for

in-tube SPME. They report that, of

the published applications using

the technique, 38.6% involved

environmental analysis, 37.3% were

biomedical, and food applications

accounted for 21.7% of articles.

Other ConfigurationsFused-silica-lined bottles provided

sample stability for the collection and

storage of a headspace subsample

with repetitive analysis using

fibre-based SPME (12). Samples

could be held in the bottle for up to

30 days with no sample loss, and

the 400-mL bottle volume allowed

multiple sampling via SPME.

A passive sampler made from an

SPME device isolated hydrophobic

organic compounds in natural waters

for subsequent measurement and

modelling of bioaccumulation by

benthic invertebrates (13).

10 mm X 100 μm sorption phase

30 mm X 250 μm sorption phase

(a)

(b)

Figure 1: Comparison of (a) a conventional SPME fibre containing a 100 μm × 10 mm coating with 0.6-μL of stationary phase with (b) a PAL SPME Arrow device containing a 250 μm × 30 mm coating with 15.3 μL of stationary phase. Adapted with permission from reference 8.

Only recently have multiple SPME fi bres been able to be desorbed automatically in a single gas chromatography (GC) sequence without manual intervention.

Perhaps the simplest alternate form of SPME is in-tube SPME. In this design, a section of an open-tubular (capillary) GC column is attached to a syringe and draw–eject cycles are used for solute sampling and desorption.

What is most notable is that as SPME gained acceptance, a plethora of techniques sprouted where the researchers seemingly attempted to place a stationary phase on any surface that could potentially be exposed to a sample.

www.chromatographyonline.com

SAMPLE PREPARATION PERSPECTIVES

(16) Z. Tang, Y. Liu, and Y. Duan, J.

Chromatogr. A 1425, 34–41

(2015).

(17) S. Lirio, W.-L. Liu, C.-L. Lin, C.-H. Lin,

and H.-S. Huang, J. Chromatogr. A

1428, 236–245 (2016).

(18) S. Ansari and M. Karimi, Talanta 164,

612–625 (2017).

(19) A. Sarafriz-Yazdi and N. Razavi, TrAc

Trend Anal. Chem. 73, 81–90

(2015).

(20) M. Cui, J. Qiu, Z. Li, M. He, M. Jin, J.

Kim, M. Quinto, and D. Li, Talanta 132,

564–571 (2015).

(21) M.V. Nair and G.S. McKelly, Forensic

Sci. Intl. 268, 131–138 (2016).

“Sample Prep Perspectives” editor

Douglas E. Raynie is a Department

Head and Associate Professor at

South Dakota State University, USA.

His research interests include green

chemistry, alternative solvents,

sample preparation, high-resolution

chromatography, and bioprocessing

in supercritical fluids. He earned

his Ph.D. in 1990 at Brigham Young

University under the direction of

Milton L. Lee. Raynie is a member

of LCGC ’s editorial advisory board.

Direct correspondence about this

column via e-mail to LCGCedit@ubm.

com

polymethacrylate monolith provided

efficient isolation of penicillins in

milk samples using in-tube SPME

(17). Ansari and Karimi reviewed the

application of molecular-imprinted

polymers (MIP) for drug analysis

in the coated, in-tube, monolithic,

sol-gel, and membrane SPME modes

(18). The low cost, high porosity,

reproducibility, and increased

sensitivity are touted as advantages

of MIPs. Another review of MIP-SPME

focused on applications (19). In

another study, hydrofluoric acid

was used to etch the surface of a

stainless steel wire and increase its

surface area (20). Then the wire was

coated with an ionic liquid as the

sorptive phase and, after optimizing

conditions, used for the extraction

of aqueous alkylphenols with

enrichment factors 6.5 to 16.8 times

greater than that with a commercial

polyacrylate fibre with an 85-μm

film. Glass-fibre filters coated with

PDMS in the capillary microextraction

mode were found to be 30-fold more

sensitive than dynamic SPME for

isolating methamphetamine vapours

associated with forensic samples

(21).

ConclusionDespite its longevity in the sample

preparation world, SPME is seeing

a tremendous growth spurt. Recent

advances and those expected in the

foreseeable future centre primarily

on the development of novel sorptive

phases and formats that allow for the

analysis of emerging systems, such

as sampling living systems or harsh

environments.

References(1) E.A. Souza-Silva, R. Jiang, A.

Rodriguez-Lafuente, E. Gionfriddo,

and J. Pawliszyn, TrAC Trend Anal.

Chem. 71, 224–235 (2015).

(2) E.A. Souza-Silva, E. Gionfriddo, and J.

Pawliszyn, TrAC Trend Anal. Chem. 71,

236–248 (2015).

(3) EA. Souza-Silva, N. Reyes-Garces,

G.A. Gomez-Rios, E. Boyaci, B. Bojko,

and J. Pawliszyn, TrAC Trend Anal.

Chem. 71, 249–264 (2015).

(4) J. Xu, G. Chen, S. Huang, J. Qiu,

R. Jiang, F. Zhu, and G. Ouyang, T

rAC Trend Anal. Chem. 85, 26–35

(2016).

(5) C.-H. Xu, G.-S. Chen, Z.-H. Xiong,

Y.-X. Fan, X.-C. Wang, and Y. Liu,

TrAC Trend Anal. Chem. 80, 12–29

(2016).

(6) A. Naccarato and J. Pawliszyn, Food

Chem. 206, 67–73 (2016).

(7) G.A. Gomez-Rios, N. Reyes-Garces,

and J. Pawliszyn, J. Sep. Sci. 38,

3560–3567 (2015).

(8) A. Kremser, M.A. Jochmann, and T.C.

Schmidt, Anal. Bioanal. Chem. 408,

943–952 (2016).

(9) A. Helin, T. Ronkko, J. Parshintsev, K.

Hartonen, B. Schilling, T. Laubli, and

M.-L. Riekkola, J. Chromatogr. A 1426,

56–63 (2015).

(10) P. Serra-Mora, Y. Moliner-Martinez,

C. Molins-Legua, R.Herraez-Hernandez,

J. Verdu-Andres, and P. Campins-Falco,

Comp. Anal. Chem. 76, 427–461

(2017).

(11) M. Fernandez-Amado, M.C.

Prieto-Blanco, P. Lopez-Mahia, S.

Muniategui-Lorenzo, and D. Prada-

Rodriguez, Anal. Chim. Acta 906, 41–57

(2016).

(12) C.A. Harvey, J.C. Carter, J.R. Ertel,

C.T. Alviso, S.C. Chinn, and R.S.

Maxwell, J. Chromatogr. A 1401, 1–8

(2015).

(13) K.A. Maruya, W. Lao, D. Tsukada, and

D.W. Diehl, Chemosphere 137, 192–197

(2015).

(14) X. Hou, L. Wang, and Y. Guo, Chem.

Lett. 46, 1444–1455 (2017).

(15) A. Amiri and F. Ghaemi, Microchim.

Acta 184, 297–305 (2017).

SEPARATION SCIENCE

HPLC2019MILANOSlam

slam

your

science.

win

2000 €(1st prize)

16-20 June 2019Milan, Italy www.hplc2019-milan.org

Despite its longevity in the sample preparation world, SPME is seeing a tremendous growth spurt.

LC•GC Europe March 2019148

ANALYSIS FOCUS: FOODOMICSA

Q. Foodomics has evolved in terms

of knowledge and technology

since the term was coined in 2009.

What is foodomics and why is it

important?

A: Foodomics was defined for the

first time by our research group

in 2009 as “a new discipline that

studies the food and nutrition

domains through the application

and integration of advanced ‘omics’

technologies to improve consumer’s

well-being, health, and knowledge”

(1). Foodomics has subsequently

been applied by many laboratories

and numerous papers have been

published in the literature (2–15).

Basically, we believe that foodomics,

even in its current early stages,

can provide new answers to many

crucial questions that need to be

solved in food science and nutrition.

Q. Could you give a general

overview of the fi eld with regards

to major research areas, the

chromatographic techniques used,

and the analytical challenges that

have been overcome during this

decade?

A: The major research areas of

foodomics include investigations

on food safety, food quality, food

traceability, and food bioactivity.

As you can deduce, the number of

research areas where foodomics

can play a role is therefore huge.

Any food-related research in which

an “omics” methodology is applied

may be considered a part of

foodomics. Apart from miniaturization,

there are two other major analytical

developments that have had a

clear influence on the evolution of

foodomics in the past ten years.

The first is related to the new

and fast sequencing methods that

are categorized together under the

common umbrella of next-generation

sequencing (NGS), which is having

a crucial impact on genomics and

transcriptomics applications. The

second is the manufacture of high

resolution mass spectrometers with

improved features and capabilities

that are becoming available at

more affordable prices, and which

are significantly contributing to the

progression of foodomics in general,

and metabolomics and proteomics in

particular.

In foodomics, we develop

transcriptomics, proteomics, and

metabolomics approaches to study

the safety, quality, and traceability

of foods, or to investigate how foods

(including functional ingredients)

interact with our genome and the

subsequent modifications that these

interactions can generate at the

proteome and metabolome level.

The chromatographic techniques

used in foodomics are those

typically used in proteomics

and metabolomics, such as

liquid chromatography (LC),

ultrahigh-performance liquid

chromatography (UHPLC), nano-LC,

gas chromatography (GC), or capillary

electrophoresis (CE) hyphenated to

high-resolution mass spectrometry

(HRMS).

With these techniques we are

able to obtain massive amounts of

information at different expression

levels, such as proteins and

metabolites.

In addition, we have developed

many comprehensive two-dimensional

liquid chromatography tandem mass

spectrometry (LC×LC–MS/MS)

methods for metabolomics profiling

of different natural compounds in

complex matrices (7,8). Logically, an

important additional step in all these

procedures is the use of adequate

sample preparation techniques (6).

One of the main analytical challenges

that we had to overcome was the

development of the three different

“omics” strategies used in foodomics:

transcriptomic-, proteomic-, and

metabolomic methods.

Considering that practically all

“omics” laboratories around the

world may work in just one of these

The Rising Role of FoodomicsTen years since its official defi nition, foodomics continues to expand the scientifi c knowledge of food and nutrition while resolving many analytical challenges along the way. LCGC Europe spoke to Alejandro Cifuentes from the Institute of Food Science Research, in Madrid, Spain, about his current foodomics research projects, the overall state of the fi eld, and the future of foodomics.

Interview by Lewis Botcherby, Associate Editor, LCGC Europe

Ph

oto

Cre

dit :

Da

nie

l V

inc

ek -

sto

ck.a

do

be

.co

m

One of the main analytical challenges that we had to overcome was the development of the three different “omics” strategies used in foodomics: transcriptomic-, proteomic-, and metabolomic methods.

We believe that foodomics, even in its current early stages, can provide new answers to many crucial questions that need to be solved in food science and nutrition.

Our ASPEC® systems simplify your solid phase extraction tasks even with the most complex matrix.

WE’LL MAKE EXTRACTIONS EASIER.

ASPEC offers food scientists the instruments and consumables

to tackle extractions, along with technical support to help you

easily transfer to a new system.

www.gilson.com/contact

LC•GC Europe March 2019150

ANALYSIS FOCUS: FOODOMICS

“omics” disciplines, the amount

of work we had to face was really

challenging! Collaboration with

other groups and laboratories was,

of course, essential to meet these

challenges. For example, we always

need collaborations for in vivo

experiments because we do not

have the necessary infrastructure

in our institute. As a result, in

our publications we usually have

co-authors from other institutions and

countries.

Q. Data management is a huge part

of any “omics” fi eld and is often

cited as a bottleneck in research.

Is this true for foodomics? And if

so, what is being done to solve this

issue?

A: Data handling is a global problem

for any discipline that generates

huge amounts of data and foodomics

is no exception. This bottleneck

prevents the integration of data from

the different “omic” levels making it

impossible to obtain the biological

meaning from the data generated in

a straightforward way. At the moment

there is no bioinformatic tool that can

put together and handle the data that

we generate from transcriptomics,

proteomics, and metabolomics

experiments simultaneously.

Other typical problems are the

difficult data storage, the slow

processing time, the improvement

needed by metabolomic databases,

the suitable mining of massive

datasets, and the need for

harmonized and standardized

methods to generate and analyze

data, among many others. Fortunately,

there are many groups all around the

world working on these problems and

trying to solve them.

Q. How has data that can be

obtained at present translated

to useful dietary or medical

information?

A: After several years working in

foodomics, we have obtained useful

information about the antiproliferative

effect of several extracts from natural

sources and food by-products.

For example, we have found a

very interesting and active extract

obtained from rosemary using

a green extraction process that

has demonstrated an impressive

antiproliferative activity against

different in vitro and in vivo models

of colon cancer together with low

toxicity.

The biological mechanisms of this

activity were found and explained via

foodomics. This was one of the first

works published in which the three

“omics” levels were simultaneously

interrogated (9). This is the first step

to understanding and demonstrating

scientifically the health impact of a

given food or dietary ingredient before

providing any dietary or medical

recommendation. I will discuss other

aspects of this research in more detail

later.

Although foodomics is a new

discipline and still in its early stages,

the discipline is expected to help to

overcome the controversial messages

that consumers are continuously

receiving from the food industries and

the regulatory agencies about the

benefits and risks of different foods.

Q. From an analytical perspective

what challenges do scientists

involved in foodomics face?

A: As foodomics uses advanced

analytical instruments and “omics”

approaches, the challenges to

be faced are the common ones

for any laboratory working with

these technologies. Thus, for

transcriptomics we must evaluate if

the cost linked to a transcriptomic

study using microarrays compensates

(or not) for the option to do the

same study using NGS of all the

transcripts—something unthinkable a

few years ago.

Regarding proteomics studies,

we are now moving from the

classical procedures, such as using

two-dimensional electrophoresis,

(isoelectric focusing followed by size

separation), imaging comparison,

tryptic hydrolysis, and matrix-assisted

laser desorption–ionization (MALDI)

MS/MS of peptides, to very powerful

strategies using isotopic derivatization

of enzymatically hydrolyzed proteins

followed by nano-LC–HRMS analysis

for identification and quantitation of all

the proteins (and samples) in a single

run.

In metabolomics the challenges

remain linked to getting a picture as

wide as possible of the metabolome

of any biological system. This means

putting together several metabolomics

platforms, such as LC–HRMS plus

GC–HRMS and CE–HRMS, to reduce

the bias induced by the sample

preparation step as much as possible,

and to manage the development

of metabolomics databases. Of

course, all of them share the same

analytical constraints related to speed,

sensitivity, and resolution. There will

always be a desire to improve these

factors in any analytical instrument.

Q. You recently published two

papers using a multianalytical

platform based on pressurized

liquid extraction (PLE), in vitro

assays, and liquid chromatography/

gas chromatography coupled to

high-resolution mass spectrometry

to investigate food by-products

valorization. Why were you

investigating food by-products

valorization, what was novel about

your approach, and what were your

main fi ndings?

A: One unique aspect of the group

involved in publishing these two

papers is the capability of integrating

different areas of expertise and

backgrounds (3,4). One co-author

has expertise in the development

of new green processes to get

valuable extracts from different

raw materials that, ultimately, can

increase the viability and economic

interest of the whole study. Another

important aspect of these studies is

sustainable development, which is

an important goal of our group as

we enter a new “green food era”. In

view of the potential of Colombian

food by-products as a promising

source of bioactive phytochemicals,

a multianalytical platform based on

Although foodomics is a new discipline and still in its early stages, the discipline is expected to help to overcome the controversial messages that consumers are continuously receiving from the food industries and the regulatory agencies about the benefi ts and risks of different foods.

151www.chromatographyonline.com

ANALYSIS FOCUS: FOODOMICS

separation techniques coupled to

HRMS was presented as part of an

integrated valorization strategy for

these by-products and includes PLE

using green solvents and different in

vitro evaluations of their bioactivity.

Thus, a comprehensive

phytochemical profiling analysis

of the compounds extracted

from goldenberries calyx (using

an optimized PLE process) was

performed by LC and GC coupled

to quadrupole time-of-flight tandem

mass spectrometry (QTOF-MS/

MS), applying new and integrated

identification approaches for

the confident identification and

structural elucidation of the bioactive

phytochemicals from the HRMS data.

Q. Could this “multianalytical”

technique be adapted for other

applications?

A: The proposed strategy could be

readily implemented in many other

applications, such as the complete

characterization of preparations with

health-promoting effects intended

to be used as functional foods or

in traditional medicines. And, of

course, for revalorization of any food

by-products by obtaining from them

bioactive compounds with added

value via, for example, some specific

bioactivity.

Q. You mentioned earlier

a metabolomics study you

published last year to investigate

metabolomic changes in cells in

response to rosemary diterpene

exposure (2). What was the aim of

this research, and what were your

main fi ndings?

A: Rosemary diterpenes have

demonstrated diverse biological

activities, such as anticancer and

anti-inflammatory properties, as well

as other beneficial effects against

neurological and metabolic disorders.

In particular, carnosic acid (CA),

carnosol (CS), and rosmanol (RS)

diterpenes have shown interesting

results on anticancer activity.

However, little was known about the

toxic effects of rosemary diterpenes

at the concentrations needed to

exert their antiproliferative effect

on cancer cells. In the mentioned

work, CA, CS, and RS exhibited a

concentration-dependent effect on

cell viability of two human colon

cancer cell lines (HT-29 and HCT116)

after 24 h exposure. The HT-29

cell line was more resistant to the

inhibitory effect of the three diterpenes

than the HCT116 cell line. Among

the three diterpenes, RS exerted the

strongest effect in both cell lines. To

investigate the hepatotoxicity of CA,

CS, and RS, undifferentiated and

differentiated HepaRG cells were

exposed to increasing concentrations

of the diterpenes (from 10 mM to

100 mM). Differentiated cells were

found to be more resistant to the toxic

activity of the three diterpenes than

undifferentiated HepaRG, probably

related to a higher detoxifying function

GREAT FLUIDIC SOLUTIONS

…from OEM customizations to spare parts

Europe: Biotech AB 7HO����������������������LQIR#ELRWHFKïXLGLFV�FRP�USA: Biotech USA LLC 7HO���������������VDOHV#ELRWHFKïXLGLFV�FRP�Japan: BioNik Inc.7HO������������������LQIR#ELRQLNLQF�FRP�

<RXU�'HJDVVLQJ�([SHUWZZZ�ELRWHFKïXLGLFV�FRP

%,27(&+�'(*$6L��35(3��'HJDVVHU�Lj�$Q�LQ�OLQH��PHPEUDQH�GHJDVVHU�

Lj�6XLWDEOH�IRU�DJJUHVVLYH�PHGLD�DQG�RUJDQLF�VROYHQWV

Lj�)ORZ�UDWHV�XS�WR��������PO�PLQ�SHU�FKDQQHO

Lj�'HOLYHUHG�DV�6WDQG�DORQH�RU�2(0�RSHQ�IUDPH

Lj�$YDLODEOH�ZLWK�����FKDQQHOV

The global movement of food and related raw materials will increase in the next few years and it will bring about an increasing number of contamination episodes worldwide.

LC•GC Europe March 2019152

ANALYSIS FOCUS: FOODOMICS

of differentiated HepaRG cells

compared with the undifferentiated

cells.

The metabolic profiles of

differentiated HepaRG cells in

response to CA, CS, and RS were

examined to determine biochemical

alterations and deepen the study

of the effects of rosemary phenolic

diterpenes at the molecular level.

A multiplatform metabolomics

study based on liquid and gas

chromatography hyphenated to HRMS

was used and revealed that rosemary

diterpenes exerted different effects

when HepaRG cells were treated

with the same concentration of each

diterpene. RS revealed a greater

metabolome alteration followed by

CS and CA, in agreement with their

observed cytotoxicity. In this work,

we demonstrated the usefulness of

metabolomics combined with cell

lines for toxicity studies because it

provides valuable information about

early events in the metabolic profiles

of cells after the treatment with the

investigated compounds. Interestingly,

this new analytical method reduces

the use of animals classically used in

toxicology studies.

Q. Are there any other recent

research projects that you

think demonstrate the value of

foodomics?

A: We have recently founded the

first Joint Research Center for Green

Foodomics Research in Beijing

together with the Chinese Academy

of Agricultural Sciences (CAAS), and

in a year a major reference work on

comprehensive foodomics (10 books

and more than 150 book chapters)

will be published (10).

The 65,000 results obtained

in Google using the search term

“foodomics” may be regarded

as additional proof of the interest

generated by foodomics among

research institutions, agencies, food

industries, regulatory laboratories,

scientific instrumentation

manufacturers, and consumers.

In this regard, we are in

collaboration with several Latin

American universities to work

together on foodomics for food waste

and food by-product revalorization,

mainly through different stays,

exchanges, courses, or roundtables.

This interaction with students,

teachers, and local people is

probably one of the most rewarding

experiences that one may have in

science.

Q. What are you currently working

on?

A: Amongst the different bioactivities

studied related to the consumption

of health-promoting food

components, their possible

neuroprotective effect is gaining

importance because of the

huge negative impact that

neurodegenerative diseases have

in society. However, to date, the

mechanisms through which the

neuroprotective activities would

be exerted are largely unknown.

In this regard, foodomics might be

a very useful tool to discover the

neuroprotective effects of dietary

terpenes at a molecular level.

The main aim of the new project in

which we are working is to study the

neuroprotective activity of terpenes

and terpenoids from natural extracts,

such as microalgae, seaweeds, and

food-related by-products from olive

oil and orange juice production,

carrying out a systematic study

of different chemical structures

combined with in vitro and in vivo

Alzheimer’s disease models and

a foodomics study of their activity.

The selection of this family of

compounds is based on previously

published reports highlighting the

possible neuroprotective effect of

some terpenoids, although, up to

now, there still exists a lot of missing

information on how they may exert

this activity. This project proposal

also includes the development of a

new methodology for the valorization

of by-products and fractions based

on the concept of biorefinery and

zero-waste philosophy.

Q. What is the future of

foodomics?

A: In the future of food science, the

first demand will still be to guarantee

food safety. The global movement of

food and related raw materials will

increase in the next few years and it

will bring about an increasing number

of contamination episodes worldwide.

In addition, many food products

containing multiple and processed

ingredients shipped from all over the

world will share common storage

spaces and production lines, which

will make it even more complicated

to face food safety episodes. As a

result, ensuring the safety, quality,

and traceability of food will be even

more complicated and important than

now. It will require the development

of new, more advanced, and more

powerful analytical strategies as

proposed by foodomics.

Foodomics can also help

to investigate and solve other

crucial topics in food science and

nutrition, such as establishing the

global role and functions of the

gut microbiome; obtaining sound

scientific evidence that supports

or refutes the beneficial claims of

functional foods and constituents;

establishing analytical methods

to guarantee food safety, origin,

traceability, and quality, including

the discovery of biomarkers to detect

unsafe products, the understanding

of the stress adaptation responses

of foodborne pathogens, or the early

detection of food safety problems;

and understanding the molecular

basis of biological processes with

agronomic and economic relevance,

such as the interaction between

crops and their pathogens, as well as

physicochemical changes that take

place during fruit ripening.

We are still far from solving many

of the aforementioned challenges.

This is why we need to keep

working, probably for a number of

years, before getting the necessary

perspective (and knowledge) on

these complex and fundamental

topics.

Q. How do you see foodomics

becoming more commonly used in

practice?

A: Food industries and regulatory

agencies have traditionally not been

particularly interested in any new

approaches that may require more

investment or effort.

We are not proposing to use

foodomics as a sledgehammer to

crack a nut. The problem is that there

are many questions that need to be

solved and clarified in food science

and nutrition. These issues are much

bigger and more crucial to our health

than a nut, and, you can be sure, to

address them you really need to have

the right approach, investment, and

knowledge. For example, traditional

153www.chromatographyonline.com

ANALYSIS FOCUS: FOODOMICS

References(1) A. Cifuentes, J. Chromatogr. A 1216,

7109 (2009).

(2) T. Acunha et al., Anal. Chim. Acta

1037(1), 40–151 (2018).

(3) D. Ballesteros-Vivas et al., J.

Chromatogr. A 1584, 155–164 (2019).

(4) D. Ballesteros-Vivas et al., J.

Chromatogr. A 1584, 144–154 (2019).

(5) A. Cifuentes, Ed., Special issue on

Foodomics and Modern Food Analysis

in TrAC-Trends in Analytical Chemistry

96, 1–212 (2017).

(6) T. Martinovic, M.G. Šrajer, and D. Josic,

Electrophoresis 39, 1527–1542 (2018).

(7) L. Montero, M. Herrero, E. Ibáñez, and

A. Cifuentes, J. Chromatogr. A 1313,

275–284 (2013).

(8) L. Montero, V. Sáez, D. von Baer,

A. Cifuentes, and M. Herrero, J.

Chromatogr. A 1536, 205–215 (2018).

(9) C. Ibáñez, A. Valdés, V. García-Cañas,

C. Simó, M. Celebier, L. Rocamora,

A. Gómez, M. Herrero, M. Castro,

A. Segura-Carretero, E. Ibáñez,

J.A. Ferragut, and A. Cifuentes, J.

Chromatogr. A 1248, 139–153 (2012).

(10) A. Cifuentes, Ed., Major References

Work on Comprehensive Foodomics

(Elsevier, To be published in 2021).

(11) A. Cifuentes, Ed., Foodomics.

Advanced Mass Spectrometry in

Modern Food Science and Nutrition

(John Wiley & Sons, Hoboken, New

Jersey, USA. 2013). (The first book

published on foodomics)

(12) M. Herrero et al., Mass Spec. Rev. 31,

49–69 (2012).

(13) T Skov, A.H. Honoré, H.M. Jensen, T

targeted analytical approaches are

the ones typically used by food

industries and regulatory agencies,

and they are useful to detect known

allergens in foods, no doubt.

However, the use of foodomics—in

this case based on proteomics

approaches—might be essential

to obtain the complete de novo

sequencing and identification of

new previously unknown food

allergens.

In addition, the use of

foodomics-based systems, biology

combined with data mining,

interpretation, and integration,

might be a key tool to discover the

molecular mechanisms beneath food

allergies at a physiological level. For

example, you can also think about

the new possibilities for foodomics

related to safety and bioactivity

of new compounds from food

by-products or waste revalorization,

or how to elucidate the complexity

of the food digestome or the food

microbiome, to mention a few. I

fully expect foodomics to lead food

science and nutrition research into a

new era.

Næs, and S.B. Engelsen, TrAC Trends

in Analytical Chemistry 60, 71–79

(2014).

(14) P. Ferranti, Curr. Opin. Food Sci. 22,

102 (2018).

(15) A. Bordoni and F. Capozzi, Current

Opinion in Food Science 4, 124–128

(2015).

Alejandro Cifuentes

is a Professor at the

National Research

Council of Spain

(CSIC) in Madrid,

Spain, and Head

of the Laboratory

of Foodomics and

Director of the Metabolomics Platform

(International Excellence Campus

CSIC + University Autonoma of

Madrid). He is the Founding Director

of the Institute of Food Science

Research and Deputy Director of the

Institute of Industrial Fermentations,

both belonging to CSIC. Alejandro’s

activities include advanced analytical

method development for foodomics,

food quality, and safety, as well as

the isolation and characterization of

natural bioactive compounds and

their effect on human health.

www.pss-polymer.com

PERFECT

SEPARATION SOLUTIONS

Dextran

PEG

PEO

PMMA

Polystyrene

Polystyrene (High Temp)

PullulanWhen Standard means Superior

GPC/SEC ReadyCal Calibration Kits

www.pss-shop.com

LC•GC Europe March 2019154

PRODUCTS

Lab equipment

Trackman

Connected is a tablet

with accessories

and applications

that make pipetting

on 96- and 384-well

plates faster and

more reliable,

reportedly improving

efficiency at the bench by tracking pipetting tasks. Designed to

communicate with Pipetman M Connected via Bluetooth, the

tablet interacts in real-time with the pipette and guides users

through their protocol with PipettePilot.

www.gilson.com

Gilson, Middleton, Wisconsin, USA.

Chromatography software

DataApex has launched

a new version of Clarity

Chromatography software.

Clarity version 8 comes with

a graphically enhanced user

interface, improvements

in MS and GLP options,

and new control modules.

According to the company,

Clarity brings easy operation, user support, and optional

extensions for various applications, such as PDA, MS, GPC,

NGA, and many more. A free demo is available from DataApex’s

website.

www.dataapex.com

DataApex, Prague, Czech Republic.

HPLC columns

Based on core–shell technology, the

Nucleoshell Biphenyl phase from

Macherey-Nagel extends the column

range for fast and efficient HPLC,

according to the company. The

separation principle of the specially

biphenylpropyl modifi ed silica is

determined by π-π and hydrophobic

interactions. Thus, it shows an enhanced retention for aromatic

and unsaturated substances and is recommended for the

sophisticated analytical separations of mycotoxins, phthalates,

pesticides, pharmaceuticals, hormones, DNPH aldehydes, and

many more substances.

www.mn-net.com

Macherey-Nagel GmbH & Co. KG, Düren, Germany.

(U)HPLC columns

YMC-Triart Bio C4 is a

new wide-pore phase

for (U)HPLC. As a result

of its 300-Å pore size, it

is designed for peptide

and protein separations.

Excellent reproducibility is

provided, according to the

company. High pH (1–10)

and temperature (up to

90 °C) stability is offered.

https://ymc.de/

rp-bioseparation.html

YMC Europe GmbH, Dinslaken, Germany.

Columns

Analysts that are working

with analytical or

preparative HPLC, or with

substances of interest that

require normal phase,

reversed phase, HILIC,

GPC/SEC, or ion-exclusion separations can be sure that

Knauer can offer a wide range of columns. To extend

the lifetime, analytical Knauer columns can be equipped

with an integrated precolumn. They are certifi ed in this

confi guration to ensure performance and quality.

www.knauer.net/columns

Knauer Wissenschaftliche Geräte GmbH, Berlin,

Germany.

GC

GL Sciences’s CryoFocus-4 is

a GC cryogenic trap, used to

refocus analytes on the column.

The technology was developed

using knowledge learned with

Optic-4, a multi-mode inlet for gas

chromatography. Cooling is done

using either CO2 (-50 °C) or LN2

(-150 °C). Low temperature trapping is combined with a

fast heating rate (60 °C/s). The result, according to the

company, is very sharp peaks, and improved separation of

volatile and semivolatile compounds.

www.glsciences.eu

GL Sciences B.V., Eindhoven, Netherlands.

155www.chromatographyonline.com

PRODUCTS

GC–MS

An automated GC–MS-based

system determines 3-MCPD,

2-MCPD, and glycidyl fatty

acid esters in edible oil

meeting the requirements of

standard ISO, AOCS, and

DGF methods. Samples

are automatically prepared

and analyzed, including

analyte derivatization and

evaporation of excess reagent and solvent for best limits of

determination and system stability.

www.gerstel.com

Gerstel GmbH & Co. KG, Mülheim an der Ruhr, Germany.

Microchip column

PharmaFluidics has launched a

μPAC trapping column with identical

stationary phase support morphology

compared to the analytical μPAC

columns. By effectively desalting

and preconcentrating the analytes

of interest onto the trap column,

analytical column lifetime and

workfl ow throughput can be

improved, according to the company.

Dilute samples can be loaded in

a bidirectional way at high fl ow

rates and with minimal loss of

chromatographic performance.

www.pharmafl uidics.com

PharmaFluidics, Ghent, Belgium.

Multi-angle static light scattering

Introducing the next

generation DAWN

multi-angle static light

scattering (MALS)

detector for absolute

characterization of the

molar mass and size of

macromolecules and

nanoparticles in solution.

DAWN offers high

sensitivity, a wide range

of molecular weight, size,

and concentration, and a large selection of confi gurations and

optional modules for enhanced capabilities, according to the

company.

https://www.wyatt.com/dawn

Wyatt Technology, Santa Barbara, California, USA.

Flow modulator

LECO has introduced a Flux

fl ow modulator option for

routine GC×GC analysis.

While LECO’s traditional

thermal modulation

alternative is still available

and offers high sensitivity,

the Flux fl ow modulator is a

cost-effective option that makes GC×GC more accessible

and easy to use, according to the company. Another

advantage of the Flux is that it does not require cryogens

to carry out GC×GC, saving the user time and resources

in their laboratory.

www.leco.com/fl ux-gcxgc-fl ow-modulator

LECO Corporation, Saint Joseph, Michigan, USA.

GPC/SEC calibration

ReadyCals allows for the rapid

preparation of reproducible

GPC/SEC calibration curves

without the inconvenience

of individually weighing the

standards, according to the

company. New in the PSS ReadyCal family are the

Dextran ReadyCals: reference material: dextran, nine

different standards with Mp 180–298 000 Da; application:

calibration of aqueous GPC/SEC systems; set contains

3 × 5 vials-1.5 mL for fi ve calibrations.

www.pss-shop.com

PSS GmbH, Mainz, Germany.

HPLC affinity column

Tosoh Bioscience has

introduced TSKgel FcR-IIIA-

NPR, an HPLC affinity column

based on recombinant

FcRIIIa receptor, a key player

in antibody-dependent

cell-mediated cytotoxicity

(ADCC). FcRIIIa affinity chromatography reportedly

allows fast evaluation of biologic activity and glycoform

patterns of antibodies. This method can provide

valuable information during cell line selection, upstream

optimization, or quality control of therapeutic antibodies.

www.tosohbioscience.de

Tosoh Bioscience GmbH, Griesheim, Germany.

LC•GC Europe March 2019156

PRODUCTS

Mass spectrometer

The Thermo Scientifi c ISQ EM single

quadrupole mass spectrometer for

high performance and productivity

standards has a mass range of

10–2000 m/z. The system offers the

power to detect and quantify small

and large molecules, and supports

analytical needs across an extensive

range of applications, from drug development to manufacturing

support and quality control. The system’s heated electrospray

ionization (HESI) and dual HESI/atmospheric pressure chemical

ionization (APCI) probes facilitate the measurement of polar and

nonpolar analytes, enabling application fl exibility. It is integrated

with HPLC systems and fully controlled by Thermo Scientifi c

Chromeleon Chromatography Data System.

www.thermofi sher.com/ISQEM

Thermo Fisher Scientifi c, California, USA.

HPLC system

The Agilent 1260 Infi nity II Prime LC system

provides the highest convenience standard

in the 1260 Infi nity II LC portfolio, as well as

an extended pressure range (up to 800 bar),

excellent quaternary mixing, and specifi cally

designed columns, according to the

company. The automated instrument features

reportedly increase analytical laboratory

efficiency and the intelligent system emulation technology (ISET)

ensures seamless method transfers from many Agilent and

third-party legacy instruments. The improved convenience level of

the system is highlighted with a local user interface—the Agilent

Infi nityLab LC Companion.

www.agilent.com

Agilent Technologies, Inc., California, USA.

FID gas station

The VICI FID gas station

combines the reliability of

the VICI DBS hydrogen and

zero-air generators into one

compact and convenient

package. Available in high and

ultrahigh purity for all GC detector and carrier gas applications.

The generator is available in two styles: fl at for placement under

a GC, or the Tower. Available in H2 fl ow ranges up to 1 L/min and

10.5 bar.

www.vicidbs.com

VICI AG International, Schenkon, Switzerland.

HILIC columns

Hilicon offers a broad range

of hydrophilic interaction

liquid chromatography (HILIC)

products to separate polar

compounds. Three column

chemistries in UHPLC and HPLC,

iHILIC-Fusion, iHILIC-Fusion(+),

and iHILIC-Fusion(P), provide customized and

complementary selectivity, excellent durability, and

ultralow column bleeding, according to the company.

The columns are suitable for the LC–MS analysis

of polar compounds in “omics” research, food and

beverage analysis, pharmaceutical discovery, and

clinical diagnostics.

www.hilicon.com

Hilicon AB, Umeå, Sweden.

Sample automation

Markes’ new Centri multitechnique

platform is an advance in sample

automation and concentration for

GC–MS, according to the company,

and offers four sampling modes: HiSorb

high-capacity sorptive extraction,

headspace, SPME, and thermal

desorption. The company reports

analyte focusing allows increased

sensitivity in all modes, state-of the-art robotics increase

sample throughput, and sample re-collection allows

repeat analysis without having to repeat lengthy sample

extraction procedures.

http://chem.markes.com/Centri

Markes International Ltd., Llantrisant, UK.

LC accessories

Restek has expanded the company’s

line of liquid chromatography

accessories for chromatographers.

High-quality couplers, fi ttings,

unions, tees, and crosses; PEEK and

stainless steel tubing; mobile phase

maintenance and safety products,

including bottle tops, valves, fi lters, and spargers are now

available.

www.restek.com/LCacc

Restek Corporation, Bellefonte, Pennsylvania, USA.

157www.chromatographyonline.com

PRODUCTS

Sample prep

LCTech has introduced an automated

system designed to clean up samples

that need to remain melted in PCB

and dioxin analysis. Three specifi cally

designed heating zones keep the

sample liquid from sample vial to the

fi rst column. The DEXTech Heat,

which is based on the established

DEXTech Pure system, processes

difficult samples, such as stearin

or PFADs. Excellent automated,

reliable results, without clogging, are

produced, according to the company.

www.LCTech.de

LCTech GmbH, Obertaufkirchen,

Germany.

Triple detection

Postnova has introduced the Triple

Detection for thermal fi eld-fl ow

fractionation (FFF) and GPC/SEC.

Triple Detection is the combination of

multi-angle light scattering (MALS),

viscosity detection, refractive index

detection, and UV detection. In a

single separation experiment, Triple Detection provides molar

mass distribution, molecular size distribution, and molecular

structure (branching, composition) of polymers, biopolymers,

polysaccharides, proteins, and antibodies.

www.postnova.com

Postnova Analytics GmbH, Landberg, Germany.

Electrochemical detector

The Decade Elite from Antec

Scientifi c is designed as an

easy-to-use electrochemical

detector that can integrate with any

LC system on the market, according

to the company. The system can

reportedly handle fast eluting

peaks in (U)HPLC and deliver fast

stabilization from dedicated fl ow

cells. When used with the SenCell,

the system is a highly sensitive

electrochemical detector.

www.AntecScientifi c.com

Antec Scientifi c, Zoeterwoude, Netherlands.

Method modelling software

Molnár-Institute’s DryLab

software has a 35-year

history in scientifi c method

modelling. Using a DoE

of twelve input runs, the

software integrates the theory of solvophobic interactions

and linear solvent strength (LSS) to predict the movements

of peaks, selectivity changes, and retention times of any

multidimensional design space. The software’s automation

module creates method sets in the most economic and

ecologic order, executes runs, and acquires results from

the CDS. Mass and other integrated data are retrieved and

ambiguity in peak tracking is reduced to a minimum.

www.molnar-institute.com

Molnár-Institute, Berlin, Germany.

Pesticide reference materials

Spex CertiPrep has introduced

a new pesticide mix to address

the European Commission’s

Regulation 2017/170. The

Commission is amending

Annexes II, III, and V to

Regulation (EC) No 396/2005

of the European Parliament

and of the Council as regards to maximum residue levels

for bifenthrin, carbetamide, cinidon-ethyl, fenpropimorph,

and trifl usulfuron in or on certain products.

www.spexeurope.com

Spex Europe, Dalston Gardens, Stanmore, UK.

Seals

The Bal Seal provides excellent

sealing performance in a broad range

of pressures, temperatures, fl ow rates,

and media, helping pump designers

meet PM requirements of more than

1 M cycles/60 L. The seal allows for

precise control of frictional forces,

resulting in reduced contamination. Designs for UHPLC,

with pressures greater than 20,000 psi, are also available.

www.balseal.com

Bal Seal Engineering, Foothill Ranch, California, USA.

T r i p l e D e t e c t i o nOnline Coupling to FFF and SEC

FFFSEC

PN3621 MALS Detector

Particle SizeRg / Molar Mass

PN3150 RI Detector

Concentration

PN3310 ViscometerDetector

Intrinsic ViscosityBranching

cosity

+

+

on

e

End [%B]

T [°C]

Flow

rate

[mL/m

in]

T [°C]

tG [min]tG [min]

Star

t[%

B]

tG [min]

T [°C]

En

d[%

B]

Start [%B]Flow rate

T [°C]

Flow

rate

[mL/m

in]

tG [min]

LC•GC Europe March 2019158

EVENT NEWS

6–7 May 2019Method Development for the

Separation of Therapeutic Proteins

(Biopolymers)

Molnár-Institute, Berlin, Germany

E-mail: [email protected]

Website: http://molnar-institute.com/

fileadmin/user_upload/Training/

SeminarRegistrationForm.pdf

12–17 May 2019 International Symposium on

Capillary Chromatography (ISCC)

and the GC×GC Symposium

Fort Worth, Texas, USA

E-mail: [email protected]

Website: www.isccgcxgc.com

13–17 May 20195th Workshop on Analytical

Metabolomics

Aristotle University, Thessaloniki, Greece

E-mail: [email protected]

Website: http://biomic.web.auth.gr/

workshop2019

22–23 May 20193rd International Conference and

Exhibition on Petroleum, Refi ning,

and Environmental Technologies

(PEFTEC 2019)

Rotterdam, Netherlands

E-mail: [email protected]

Website: www.ilmexhibitions.com/peftec/

17–18 June 2019 Analytical Quality by Design

Naarden-Bussum, Netherlands

E-mail: [email protected]

Website: www.kantisto.nl/index.php/

agenda/30-agenda-items/38-analytical-

quality-by-design

18–20 June 2019LABWorld China 2019

Shanghai New International Exhibition

Center (SNIEC), Shanghai, China

E-mail: [email protected]

Website: www.pmecchina.com/

labworld/en

7–10 July 2019International Symposium

on Preparative and Process

Chromatography (PREP 2019)

Baltimore, USA

E-mail: [email protected]

Website: www.prepsymposium.org/

Please send any upcoming event

information to Lewis Botcherby at

[email protected]

The 48th International Symposium on High

Performance Liquid Phase Separations and Related

Techniques (HPLC 2019)

The 48th International Symposium on High

Performance Liquid Phase Separations

and Related Techniques (HPLC 2019) will be

held 16–20 June 2019 at the Milano-Bicocca

University, Milan, Italy. This is the first time that

this symposium will be held in Italy.

The HPLC symposium series is the largest,

most recognized international forum on high performance liquid chromatography

(HPLC) and related techniques. It is the ideal location for industrial and academic

researchers to exchange information with other colleagues from all over the world.

Both fundamental and practical aspects of separation science will be covered

during the conference, with the main focus being on new, highly relevant trends

emerging in this field, cutting-edge applications, and innovation in the technology.

Among the many topics that will be covered at HPLC 2019, particular emphasis

will be given to hyphenated techniques, in particular liquid chromatography

coupled to mass spectrometry (LC–MS), design and characterization of

stationary phases, micro- and nano-fluidics, supercritical fluid chromatography

(SFC) and capillary electrophoresis (CE) and their applications in proteomics,

metabolomics, food analysis, and characterization of biopharmaceuticals and

biosimilars. HPLC 2019 will also feature some unique highlights, such as a

session entirely dedicated to HPLC for process analytical technology (PAT) and

quality assessment in preparative chromatography, and a session focusing on

high-performance thin-layer chromatography (HPTLC). A complete list of topics

can be found on the HPLC website.

Thanks to its multidisciplinary character, the symposium

represents an important source for analytical chemists,

biochemists, and engineers seeking practical solutions to

their problems. Researchers will be presenting their work

as lectures in topical scientific sessions or in exciting poster

sessions. We strongly encourage the active participation

of younger scientists in the symposium. A great amount

of effort has been made to guarantee low student fees,

including some special packages designed to give students

the opportunity to attend short courses taught by highly

reputed experts. We are offering 14 short courses on different topics, whose

description can be found online.

A series of awards will be presented during the conference, including the Csaba

Horváth Young Scientist Award, the Best Poster Award, the Uwe Neue Award,

and many more. In addition to the traditional awards, HPLC 2019 will feature new

exciting ideas to recognize the scientific activity of young colleagues, which will be

announced soon on the website and on social media.

A rich scientific and social programme awaits participants, starting with the

opening ceremony in the exclusive location of the “Giuseppe Verdi” Conservatorio

di Milano, the largest music academy in Italy and one of the most important

worldwide. A unique opening event merging music and science will celebrate the

genius of Leonardo da Vinci on the 500th anniversary of his death.

Milan is a cosmopolitan and vibrant centre of Italian culture. The various cultures

of northern Europe and Mediterranean countries merge and as such Milan has

a lot to offer, including art museums, history, opera (La Scala), music, fashion,

design, and Italian culinary delights.

Milan also offers a wide range of accommodation for all budgets, and with its

three international airports it is easily accessible by plane, and also by car or train

for those closer by.

The organizers look forward to welcoming participants in Milan in 2019!

E-mail: [email protected] Website: www.hplc2019-milan.org

EEVVEENNTT NNEEWWSSEVENT NEWS

LC•GC Europe March 2019158

powered by

www.chromacademy.com/hplc_troubleshooting.html

Try it now for FREE @

For more information contact:

Glen Murry: +1 732.346.3056 | [email protected]

Peter Romillo: +1 732.346.3074 | [email protected]

Jacqueline Robertson: +44 (0)1357 522961 | [email protected]

Become the lab expert with our interactive

Get answers fast. Reduce downtime. Increase efficiency.

HPLC Troubleshooter

LC•GC Europe March 2019160

THE ESSENTIALS

Hydrophobic interaction chromatography

(HIC) is a very versatile technique

capable of separating protein

molecules according to differences

in hydrophobicity (in a similar way to

reversed-phase separations). But, unlike

reversed-phase separations, HIC utilizes

nondenaturing mobile phases and is

often employed for protein purification,

within a capture, intermediate purification,

polishing regime. Furthermore, because

of its ability to discriminate between

analytes with only minor differences in

hydrophobicity, HIC has advantages in

separating variants with subtle changes

that can expose more, or less, of a

hydrophobic surface. These variants

are difficult to resolve using other

chromatographic approaches, including

oxidized species (such as methionine),

deamidation (asparagine-aspartic acid),

and isoaspartate formation; as well as the

separation of antibody–drug conjugate

(ADC) variants, and for drug-to-antibody

ratio (DAR) analysis.

HIC uses a somewhat hydrophobic

stationary phase with typically shorter

ligands than in reversed-phase

chromatography (C4 ligands are

popular), and typically the ligand binding

density is lower (bonded phases ligands

are spaced further apart on the stationary

phase surface). A salt solution is used as

the strong eluent to promote adsorption

between the protein hydrophobic

regions and the bonded phase. The

salt concentration is gradually reduced

throughout the run (typically to 0%) to

allow the protein to elute, and elution

order in HIC is usually least hydrophobic

to the most hydrophobic. Crucially, unlike

reversed-phase HPLC, the eluent does

not contain a denaturing organic solvent,

and the native protein structure will be

retained.

With a sufficiently high eluent salt

concentration, the hydrophobic regions

of the protein will be encouraged to

interact with the hydrophobic stationary

phase, and will concentrate to ensure

sharp chromatographic peaks. As

the salt concentration decreases

during the gradient elution, adsorption

to the stationary phase becomes

less favourable, and the proteins will

elute in hydrophobicity order (least

hydrophobic first). The second law of

thermodynamics also plays a part in

protein retention in HIC, and the highly

ordered water cages around the protein

and the stationary phase ligands makes

interaction, and therefore retention,

much less favourable. However, at

higher salt concentrations, these water

cages are disrupted (less ordered), and

therefore retention is more favourable.

Ammonium sulfate is typically used

as the salt in HIC, as it is very effective

at “salting out” the protein (Hofmeister

series for protein retention is LiCl <

NaCl < Na2HPO3 < (NH4)2SO4 <

Na2SO4), which promotes the protein

stationary phase interaction without

causing protein denaturation (2 M

is a typical starting concentration),

while being highly soluble in aqueous

solutions. Care should be taken to avoid

elevated salt concentrations, which

may lead to precipitation due to protein

agglomeration.

Proteins possess complex

three-dimensional structures that result

in areas rich in amino acids possessing

hydrophobic side chains, or areas rich in

ionic side chains. Consequently, proteins

may well be poorly soluble in water

alone, with hydrophobic interactions

as well as ionic interactions able to

occur. Small amounts of buffer help to

disrupt the weak interactions that can

cause insolubility. This process is often

referred to as “salting in” and improves

the protein solubility. It is also important

to maintain a constant pH and so the

use of a buffer is also typical in HIC

chromatography, with 50-mM sodium

phosphate at pH 7 added to both eluent

A and eluent B a typical example.

The pH of the eluent will influence the

degree of ionization of ionogenic amino

acid residues, and therefore retention

will alter as the protein hydrophobicity

changes. Eluent pH values below 5

or above 8.5 tend to dramatically alter

retention and therefore the eluent pH

is typically maintained between these

values and should be optimized on a

case-by-case basis.

Due to the thermodynamic

dependency of the retention mechanism

in HIC, variations in temperature

will also alter retention behaviour.

The relationship between retention

and temperature is complex, and

therefore should be optimized on a

case-by-case basis, using a controlled

room temperature (20–25 oC) as a

reasonable starting point. It is important

that temperature is carefully controlled

in order to ensure run-to-run or

batch-to-batch retention time stability.

Water-miscible additives, such as

detergents, can reduce the binding of

proteins even at low concentrations, as

they compete with the protein for binding

sites on the stationary phase surface.

Therefore, addition of such species

can substantially improve the elution

efficiency of the protein, sharpening

peaks and often improving resolution.

Lately, HIC has been extensively

used for DAR determination in ADCs.

Typically, the protein, usually a

monoclonal antibody (mAb), is fully

reduced prior to analysis of light and

heavy chains to determine the number,

and position, of conjugated small

molecules, which can affect the potency

of the biotherapeutic.

Hydrophobic InteractionChromatography of ProteinsA discussion of hydrophobic interaction chromatography (HIC) theory and its application to the analysis of proteins and biomolecules is presented.

Get the full tutorial at www.CHROMacademy.com/Essentials

(free until 20 April).

More Online:

THEAPPLICATIONS

BOOK

March 2019

www.chromatographyonline.com

CONTENTS

THE APPLICATIONS

BOOK

Food and Beverage

163 Determination of Fatty Acid Esters of 2- and 3-Monochloro-1,2- propanediol (MCPD) and Glycidol in Edible Oil Using GC–MS TQ

Waldemar Weber, Shimadzu Europa GmbH

Medical/Biological

165 Separation and Identification of Non-Covalent Metal Complexes from Bacteria by HILIC–MS

Karen Scholz1, Matthias Baune1, Heiko Hayen1, and Wen Jiang2,

1Institute of Inorganic and Analytical Chemistry, University of Münster,

Münster, Germany, 2HILICON AB

167 Extraction of Loperamide and N-Desmethyl Loperamide from Blood Followed by LC–MS/MS Analysis

Tina Fanning, UCT, LLC

Pharmaceutical/Drug Discovery

168 Add More Confidence to Your UHPLC–MS AnalysisRudolf Köhling, Merck

169 Assessing Antibody ADCC Activity by Affinity HPLCTosoh Bioscience GmbH

170 Reversed-Phase Analyses of Monoclonal Antibodies Using High TemperaturesYMC Europe GmbH

General

172 Identifying Long-Chain and Short-Chain Branching in PolymersChris Deng, Wyatt Technology

Nic

ola

s F

ara

maz/s

tock.a

do

be.c

om

mach

ista

/sto

ck.a

do

be.c

om

p

hasin

/sto

ck.a

do

be.c

om

cassis

/sto

ck.a

do

be.c

om

F16

-ISO

10

0/s

tock.a

do

be.c

om

CONTENTS

162 THE APPLICATIONS BOOK – MARCH 2019

THE APPLICATIONS BOOK – MARCH 2019 163

FOOD AND BEVERAGE

Determination of Fatty Acid Esters of 2- and 3-Monochloro-1,2-propanediol (MCPD) and Glycidol in Edible Oil Using GC–MS TQWaldemar Weber, Shimadzu Europa GmbH

Esterifi ed and free forms of 2-MCPD, 3-MCPD, and glycidol

are heat-induced contaminants found in various types of

processed food (1). Numerous studies have investigated the

levels of these contaminants in oils and fats from different

sources, and mitigation strategies are currently under

development. However, to support future research where

higher sensitivity and selectivity are necessary, analytical

solutions based on triple-quadrupole mass spectrometers will

be required. In the present study, a novel GC–MS TQ method

was developed and validated for the simultaneous analysis

of 2-MCPD, 3-MCPD, and glycidyl fatty acid esters in edible

oil. This method was subsequently applied to quantitation of

these contaminants in commercial edible oil samples.

Methods and Materials

Fatty acid esters of 2-MCPD, 3-MCPD, and glycidol in oil were

extracted and derivatized using phenylboronic acid according to the

AOCS Offi cial Method Cd 29a-13 (2). The analyte standards used

were 1,3-dipalmitoyl-2-chloropropanediol (2-MCPD), 1,2-dipalmitoyl-

3-chloropropanediol (3-MCPD), and glycidyl palmitate (GlyP). The

internal standards were 1,2-dipalmitoyl-3-chloropropanediol-d5

(3-MCPD-d5) and glycidyl palmitate-d5 (GlyP-d5). The extracts were

analyzed on a GCMS-TQ8040 NX system (Shimadzu Corporation).

Separation was performed using a 30 m × 0.25 mm, 1.0-μm

SH-Rxi-1MS capillary column (Shimadzu Corporation). Detailed

instrumental conditions are presented in Table 1, and MRM

parameters for the different analytes are shown in Table 2.

Results

MRM chromatograms of the analytes (using target MRM transitions)

showed good peak shapes (Figure 1). The retention times of

3-MCPD-d5, 3-MCPD, 2-MCPD, glycidol-d5, and glycidol were

18.4 min, 18.6 min, 19.6 min, 21.6 min, and 21.7 min, respectively. Method validation was performed to assess parameters such

as sensitivity, accuracy, precision, linearity, and repeatability using

calibration standards or quality control (QC) samples. Subsequently,

this method was applied to quantitation of the different analytes in

commercially available edible oil samples.

Sensitivity

The sensitivity of the assay was examined by measuring the signal to

noise (S/N) ratio of the analyte peaks. The LOD (limit of detection)

was defi ned as S/N ratio of at least 5. The LOD for this method was

0.003 μg of 2-MCPD and 3-MCPD and 0.006 μg of glycidol. The

limit of quantitation (LOQ) was defi ned as S/N ratio of at least 10.

The LOQ of this method was 0.01 μg of 2-MCPD and 3-MCPD and

0.024 μg of glycidol.

(x100,000,000)

3-MCPD-d5 2-MCPD Glycidol-d5Glycidol3-MCPD

1.0

1.0

18.0 18.5 19.0 19.5 20.0 20.5 21.0 21.5 22.0 22.5

Figure 1: Representative GC–MS (MRM) chromatogram for

3-MCPD-d5, 3-MCPD, 2-MCPD, glycidol-d5, and glycidol.

2-MCPD

Area Ratio

0.75

0.50

0.50

0.250.25

0.00 0.000.0 1.0

2.5

2.0

1.5

1.0

0.5

0.00.0 0.01.0 1.0 2.0

Conc. Ratio Conc. Ratio Conc. Ratio

Area Ratio Area Ratio

3-MCPD Glycidol

Figure 2: Calibration curves for the analytes 2-MCPD, 3-MCPD,

and glycidol where the x-axis is the concentration ratio and y-axis

is the peak area ratio between analyte and IS peak areas.

Table 1: Instrumental conditions used for analysis

Parameter Setting

GC Conditions

Injection Mode/Volume Splitless/0.5 μL

Injector Temperature 250 °C

Flow Control Mode Pressure

Pressure 49.7 kPa

Oven Temperature Program

80 °C (1 min) A 10 °C/min to 170 °C

(5 min) A 3 °C/min to 200 °C A 15 °C/

min to 300 °C A300 °C (15 min)

MS Conditions

Interface Temperature 300 °C

Ion Source Temperature 230 °C

164 THE APPLICATIONS BOOK – MARCH 2019

FOOD AND BEVERAGE

Accuracy and Precision

QC samples at three concentrations (QC1, QC2, and QC3) were

used to investigate the accuracy and precision of the method. High

accuracy and precision were demonstrated for this method as the

accuracies of the QC samples were all within 100 ± 7% and the

%RSD were all < 10% (n = 6).

Linearity and Repeatability

Excellent repeatability of the peak areas of consecutive injections

were achieved for all analytes (< 3%) (n = 6). Eight calibration

standards for 2-MCPD and 3-MCPD ranging 0.010–0.930 μg and

glycidol 0.024–2.130 μg were used to construct the calibration

curves. The calibration curves (Figure 2) for all analytes showed

excellent linearity (R2 > 0.998) (n = 6).

Application of Method for Quantitation of Analytes

in Commercially Available Oil Samples

The validated method was applied to quantitation of esters of

2-MCPD, 3-MCPD, and glycidol in edible oil samples from different

sources (Table 3, Figure 3). Generally, samples containing palm oil

were found to contain the highest levels of the contaminants.

Conclusions

In summary, a novel GC–MS method was developed and validated

for the simultaneous analysis of 2- and 3-MCPD and glycidol fatty

acid esters in edible oil. This method showed more than threefold

improvement in sensitivity compared to the offi cial method currently

available and demonstrated excellent linearity, repeatability,

accuracy, and precision. Application of this method to the analyses

of commercially available edible oil samples confi rmed that samples

containing palm oil show higher levels of contaminants.

References

(1) Federation for European Oil and Proteinmeal Industry, “FEDIOL Q&A on

2- and 3-MCPD and Their Esters and Glycidyl Esters”, (2016).

(2) The American Oil Chemists’ Society, “2- and 3-MCPD Fatty Acid Esters and

Glycidol Fatty Acid Esters in Edi-ble Oils and Fats by Acid Transesterification.

AOCS Official Method Cd29a-13, Cd29b-13 and Cd29c-13”, (2013).

Shimadzu Europa GmbHAlbert-Hahn-Str. 6–10,

D-47269 Duisburg, Germany

Tel.: +49 203 76 87 0

Fax: +49 203 76 66 25

E-mail: [email protected]

Website: www.shimadzu.eu

(x100,000,000)

3-MCPD-d5 2-MCPD Glycidol-d5Glycidol3-MCPD

18.0 18.5 19.0 19.5 20.0 20.5 21.0 21.5 22.0 22.5

(x100,000,000)

3-MCPD-d5 2-MCPD Glycidol-d5Glycidol3-MCPD

18.0 18.5 19.0 19.5 20.0 20.5 21.0 21.5 22.0 22.5

2.0

1.5

1.0

0.5

3.0

2.5

2.0

1.51.0

0.5

Figure 3: Representative MRM chromatograms of olive

oil (top) and kenaf seed oil (bottom) shown below.

Table 2: MRM parameters used in analysis

Start

Time

(min)

End

Time

(min)

Event

Time

(s)

Ch1 m/zCE

1 Ch2 m/z

CE

2 Ch3 m/z

CE

3 Ch4 m/z CE 4 Ch5 m/z CE 5

3-MCPD-d5 17.0 19.2 0.167 150.0>93.1 15 201.0>150.2 10 201.0>93.2 30 203.0>150.1 10 203.0>93.2 25

3-MCPD 17.0 19.2 0.133 147.0>91.1 15 196.0>91.2 25 147.0>65.1 25 198.1>147.2 15 ----- --

2-MCPD 19.2 21.2 0.300 196.0>104.1 20 198.0>104.1 20 196.0>91.2 10 196.0>62.0 25 198.0>91.1 10

Glycidol-d5 21.2 23.0 0.167 150.0>93.1 15 245.0>150.1 10 247.0>150.1 10 245.0>93.1 25 247.0>93.1 25

Glycidol 21.2 23.0 0.133 242.0>147.1 10 240.0>147.1 15 240.0>91.2 30 242.0>91.1 25 ----- --

Table 3: Concentration of contaminants in commercially

available edible oil samples

Sample Analyte Concentration (ppm) (n = 3)

2-MCPD 3-MCPD Glycidol

Kenaf seed oil 0.173 ± 0.007 0.354 ± 0.008 0.367 ± 0.003

Mustard oil < LOQ < LOQ < LOQ

Olive oil < LOQ < LOQ < LOQ

Palm oil A 2.820 ± 0.062 5.313 ± 0.032 8.813 ± 0.131

Palm oil B 0.653 ± 0.032 1.145 ± 0.023 5.891 ± 0.032

Peanut oil 0.491 ± 0.008 0.898 ± 0050 0.589 ± 0.016

Vegetable

cooking oil*1.346 ± 0.018 2.804 ± 0.061 4.273 ± 0.046

*Vegetable cooking oil contains palm olein and soybean oil

THE APPLICATIONS BOOK – MARCH 2019 165

MEDICAL/BIOLOGICAL

Separation and Identifi cation of Non-Covalent Metal Complexes from Bacteria by HILIC–MSKaren Scholz1, Matthias Baune1, Heiko Hayen1, and Wen Jiang2, 1Institute of Inorganic

and Analytical Chemistry, University of Münster, Münster, Germany, 2HILICON AB

Pseudomonas bacteria are a genus of rod-shaped, ubiquitous, and

gram-negative bacteria, which are resistant against commonly used

antibiotics and disinfectants. Pseudomonads are able to survive

in extreme conditions (1) and are increasingly used as whole-cell

biocatalysts. The range of their biotechnological applications is not

limited by the toxicity of used chemicals because of their manifold

survival strategies (2).

Pseudomonads require iron(III) for metabolism and growth.

Under aerobic conditions, the iron(III) is almost insoluble and

the pseudomonas bacteria synthesize yellow-green fl uorescent

molecules called pyoverdines. These molecules can react with

iron(III) and form stable hexadentate iron(III)-complexes in the

extracellular space and transport the metal (1). The pyoverdines

consist of three structural parts: (i) chromophoric unit derived from

2,3-diamino-6,7-dihydroxyqunoline; (ii) strain specifi c peptide

moiety consisting of 6–14 proteinogenic and nonproteinogenic

amino acids; (iii) acyl side chain derived from malic acid, succinic

acid, their mono amides, α-ketoglutaric acid, or glutamic acid.

Pyoverdines are highly polar molecules that conventional

reversed-phase high performance liquid chromatography (HPLC) is

problematic due to insuffi cient retention. Furthermore, we intended

to separate and identify intact metal complexes in our study.

Hydrophilic interaction liquid chromatography (HILIC) is well suited

for the separation of polar compounds and enables the separation

of polar non-covalent metal species (3,4). In this study, we aimed

to use a charge modulated HILIC stationary phase (iHILIC®-Fusion)

to separate different siderophores of the Pseudomonas taiwanensis

VLB120 bacteria.

Experimental

LC–MS System: Agilent 1100er LC system and Q Exactive™ Plus

Hybrid Quadrupole-Orbitrap™ mass spectrometer, equipped with a

Figure 1: Structures of siderophores produced by P. taiwanensis VLB120: four pyoverdines with an acyl side chain derived from

malic acid (1), succinic acid (2), malic acid amide (3), succinic acid amide (4); one pyoverdine (5) with proline in the peptide moiety.

166 THE APPLICATIONS BOOK – MARCH 2019

MEDICAL/BIOLOGICAL

HESI II source, operated in positive ionization mode (ESI+).

Column: 150 × 2.1 mm, 3.5 μm 100 Å, iHILIC®-Fusion (P/N

110.152.0310, HILICON AB, Sweden)

Eluent: A) acetonitrile; B) ammonium bicarbonate (5 mM, pH 7)

and acetonitrile (95:5, v/v);

Gradient Elution: 0–0.75 min, 80% A; 0.75–20 min, from 80% A

to 40% A; 20–25 min, 40% A; 25–27 min, from 40% A to 80% A;

27–40 min, 80% A

Flow Rate: 0.3 mL/min

Column Temperature: 40 °C

Injection Volume: 5 μL

P. taiwanensis VLB120 Cell Supernatants: Cell supernatants of the P.

taiwanensis VLB120 bacteria were provided by Dr. Till Tiso at iAMB

(Institute of Applied Microbiology) and ABBt (Aachen Biology and

Biotechnology, RWTH Aachen University, Aachen, Germany) and

purifi ed by solid-phase extraction as described in reference 5. The

samples for injection contained fi ve pyoverdines that differ in their

acyl side chain and in the peptide moiety (6), shown in Figure 1.

Results and Conclusion

The different siderophores of the P. taiwanensis VLB120 bacteria

and their non-covalent iron and aluminium complexes can be

simultaneously separated and determined by an iHILIC®-Fusion

column hyphenated with electrospray ionization mass spectrometry

(ESI-MS) detection (Figures 2 and 3). Pyoverdines with various

acyl side chains exhibit different levels of hydrophilic partitioning

and electrostatic interaction with the stationary phase. The

extracted ion chromatograms in Figure 3 also demonstrate that the

iHILIC®-Fusion column is even capable of separating the iron(III)

and aluminium(III) complexes of each siderophore. Because of

the smaller radius, aluminium has a stronger polarizing effect on

the siderophores. Therefore, aluminium(III) complex has higher

retention on the HILIC stationary phase. This work demonstrates

that intact non-covalent metal complexes can be separated and

identifi ed by a HILIC–MS method using an iHILIC®-Fusion column.

References

(1) C. Cézard, N. Farvacques, and P. Sonnet, Curr. Med. Chem. 22, 165–186

(2015).

(2) J. Volmer, C. Neumann, B. Bühler, and A. Schmid, Appl. Environ. Microbiol.

80, 6539–6548 (2014).

(3) G. Weber, N. von Wiren, and H. Hayen, J. Sep. Sci. 31, 1615–1622 (2008).

(4) J. Köster, R. Shi, N. von Wiren, and G. Weber, J. Chromatogr. A 1218,

4934–4943 (2011).

(5) M. Baune, Y. Qi, K. Scholz, D.A. Volmer, and H. Hayen, BioMetals 30,

589–597 (2017).

(6) K. Scholz, T. Tiso, L.M. Blank, and H. Hayen, BioMetals 31, 785–795

(2018).

HILICON ABTvistevägen 48, SE-90736, Umeå, Sweden

Tel.: +46 (90) 193469

E-mail: [email protected]

Website: www.hilicon.com

3x106

Time (min)

3

5

0

20

20

40

60

80

100

1510

0

11x106

2x106

Inte

nsi

ty (

-)

Elu

en

t B

(%

)

2x104

1x104

8x103

4x103

0

5b

5a

10 12 14 16 18 20

Time (min)

Inte

nsi

ty (

-)

Figure 2: Extracted ion chromatograms of the iron(III) complexes

of the pyoverdines with an acyl side chain derived from malic

acid (1) or malic acid amide (3), and proline pyoverdine (5).

Figure 3: Extracted ion chromatogram showing

the iron complex (5a) and the aluminum complex

(5b) of the proline pyoverdine (5).

THE APPLICATIONS BOOK – MARCH 2019 167

MEDICAL/BIOLOGICAL

Extraction of Loperamide and N-Desmethyl Loperamide from Blood Followed by LC–MS/MS AnalysisTina Fanning, UCT, LLC

Loperamide is an over-the-counter antidiarrhoeal drug

that has been considered to be safe when used as directed.

However, as the opioid epidemic continues to ravage our

nation there has been an increasing number of reported cases

of loperamide overdose. This application note outlines a simple

three-step method for the extraction of loperamide and its

main metabolite, N-desmethyl loperamide, from blood. UCT’s

Clean Screen® XCEL I column provides users with the same

level of sample clean-up as traditional SPE while allowing the

elimination of timely conditioning and wash steps.

Procedure

Sample Pretreatment

1. To 1 mL blood sample add 3 mL acetate buffer pH 5 and

appropriate amount of internal standard.

2. Vortex samples for 30 s to mix.

SPE Procedure

1. Apply samples to SPE tubes without any preconditioning.

2. Allow samples to fl ow through the column at a rate of 1–2 mL/min.

3. Wash samples with 2 mL of D.I. H2O.

4. Wash SPE column with 2 mL of 98:2 methanol–glacial acetic acid.

5. Dry column for 5 min at full vacuum or pressure.

6. Wash samples with 2 mL of hexane.

7. Dry column for 10 min at full vacuum or pressure.

8. Elute compounds with 2 mL of 78:20:2 DCM–IPA–NH4OH.

9. Collect eluate at a rate of 1–2 mL/min.

10. Evaporate to dryness at < 50 °C.

11. Reconstitute sample in mobile phase.

Instrumental

LC–MS/MS: Thermo Scientifi c™ Dionex™ 3000 (LC) TSQ

Vantage™ (MS/MS)

Column: UCT Selectra® DA HPLC Column

100 × 2.1 mm, 1.8-μm

Guard Column: UCT Selectra® DA Guard Column

10 × 2.1 mm, 1.8-μm

Injection Volume: 1 μL

Mobile Phase A: D.I. H2O + 0.1% formic acid

Mobile Phase B: Acetonitrile + 0.1% formic acid

Column Flow Rate: 0.30 mL/min

Results

Conclusion

This application note effectively outlines a simple three-step method

for the extraction of loperamide and its main metabolite, N-desmethyl

loperamide, from blood. UCT’s Clean Screen® XCEL I column provides

users with the same level of exceptional sample clean-up as traditional

SPE while allowing the elimination of timely conditioning and wash steps

in addition to a concentration step. Sample extracts

are analyzed via UHPLC–MS/MS utilizing UCT’s

Selectra® C18 1.8 μm analytical column.

UCT, LLC2731 Bartram Rd, Bristol, Pennsylvania 19007 USA

Tel.: (800) 385 3153

Website: www.unitedchem.com

Extraction/Analytical Materials

CSXCE106Clean Screen® XCEL I

130 mg / 6 mL SPE Cartridge

SLC-18100ID21-18UMSelectra® C18 HPLC

100 × 2.1 mm, 1.8-μm

MRM transitions (ESI+, 50 ms dwell time)

Compound tR (min) Q1 ion Q3 ion 1 Q3 ion 2

1N-Desmethyl-

loperamide2.83 463.1 252.06 196.03

2 loperamide 2.95 477.1 266.05 209.98

Recovery (%) from blood (n = 3)

Compound 5 ng/mL 10 ng/mL 50 ng/mL

N-desmethyl-loperamide 106 97 96

loperamide 102 95 93

Average recovery 104 96 94.5

168 THE APPLICATIONS BOOK – MARCH 2019

PHARMACEUTICAL/DRUG DISCOVERY

Add More Confi dence to Your UHPLC–MS AnalysisRudolf Köhling, PhD, Merck

For your highly sensitive UHPLC–MS analyses, how can you reduce

noise and additional signals to a minimum? Our new high-end

UHPLC–MS solvents raise the standard for low baseline noise and

clean mass spectra.

Our new range of advanced UHPLC–MS LiChrosolv® solvents

have been developed to exceed all expectations, providing rapid

and reliable results in both ESI/APCI positive and negative ionization

modes.

Thanks to their lowest level of background noise and ion

suppression, this quality ensures the optimum ionization effi ciency

to enable the highest sensitivity. With these features, use of these

solvents can also help to extend column lifetime.

To ensure that you have confi dence in your results, we specify

the lowest possible limit of polyethylene glycol (PEG) impurities in

all our UHPLC–MS solvents.

Our advanced UHPLC–MS LiChrosolv solvents have been

designed to meet the highest requirements of UHPLC–MS

in research and quality control, including proteomics and

metabolomics, as well as environmental, clinical, food, or industrial

testing applications.

A new standard for the unlimited application of ultrahigh-pressure

chromatography has been set.

Features and Benefi ts

• Suitability tested and specifi ed for UHPLC–MS and UHPLC–UV:

for analytical fl exibility

• Specifi ed quality in positive and negative ESI and APCI MS

for lowest detection limits and confi dence in analyses in all

important MS modes (Test 1)

• ESI or APCI (+) < 2 ppb

• ESI or APCI (-) < 10 ppb

• Lowest impurity profi le: for interference-free baselines (Test 2)

• Microfi ltration through 0.2 μm fi lter (Test 3)

• Prolonged lifetime of fi lters and mechanical parts

in HPLC systems

• Reduced risk of column clogging

• Packaged in borosilicate glass bottles: minimized contamination

with metal ions

• Lowest levels of trace metal impurities: for minimized metal ion

adduct formation

• <5 ppb

• Lowest level of polyethylene glycol (PEG) impurities in our entire

UHPLC–MS solvent lineup to give you confi dence in your results

(PEG S/N signal-to-noise-ratio < 50)

LiChrosolv methanol for UHPLC–MS shows a fl at baseline and by

far the lowest impurity profi le compared to the competition. Both

competitor’s high purity UHPLC–MS products A and B show a

baseline drift and signifi cant impurity peaks.

Test 1: UHPLC–MS gradient run with LiChrosolv acetonitrile for

UHPLC–MS shows a clear detection and identifi cation of 1 ppb

reserpine, 500 ppt propazine, and 4 ppb prednisolone, with very

low background interferences.

Test 2: Comparison of LiChrosolv methanol for UHPLC–MS (blue

line) with two competitor UHPLC–MS products.

To read the rest of this application please visit

SigmaAldrich.com/UHPLC–MS.

MerckFrankfurter Strasse 250

Darmstadt, 64293, Germany

Website: www.sigmaaldrich.com

Merck

THE APPLICATIONS BOOK – MARCH 2019 169

PHARMACEUTICAL/DRUG DISCOVERY

Assessing Antibody ADCC Activity by Affi nity HPLCTosoh Bioscience GmbH

Antibody-dependent cell-mediated cytotoxicity (ADCC)

is a crucial mechanism of action (MoA) of anti-tumour

therapeutic antibodies and FcγIIIa receptor plays a key role

in this process by interacting with the N-glycans of IgG Fc

regions. Hence, affi nity chromatography on Fc receptor

ligands can deliver valuable information about expected

ADCC activity and mAb glycoform distribution.

A new HPLC column, TSKgel FcR-IIIA-NPR, is based on a

recombinant FcaIIIa receptor ligand and allows fast assessment of

biologic activity of monoclonal antibodies (mAbs). As Fc-glycans

of antibodies are known to play an important role in Fc-mediated

interactions, the separation pattern of mAbs on TSKgel FcR-IIIA-NPR

can be correlated to FC-glycans as well (1). Terminal galactose

residues increase affi nity to FcaRIIIa while core fucose residues

reduce it. This correlates with the known infl uence of galactose and

fucose on antibody-dependent cell-mediated cytotoxicity (ADCC)

activity. Accordingly, early eluting peaks of TSKgel FcR-IIIA-NPR

represent glycoforms with low ADCC activity, while late eluting

peaks represent glycoforms with high ADCC activity (Figure 1).

Separation of mAb Glycoforms

Figure 2 demonstrates the specifi city of the recombinant FcaRIIIA

ligand for N-glycans of the Fc domain of mAbs. Adalimumab

analyzed with TSKgel FcR-IIIA-NPR shows a typical pattern of three

peaks, corresponding with the molecule’s glycan heterogeneity.

Treatment of adalimumab with PNGase F de-glycosylates the

sample. De-glycosylated adalimumab does not bind to TSKgel

FcR-IIIA-NPR; the N-glycan related peaks are absent.

HPLC Conditions

Column: TSKgel FcR-IIIA-NPR (4.6 mm × 7.5 cm L, 5-μm)

Mobile Phase: A: 50 mmol/L sodium citrate, pH 6.5;

B: 50 mmol/L sodium citrate, pH 4.5

Flow Rate: 1 mL/min

Temp.: 25 °C

Detection: UV @ 280 nm

Sample: 50 μL of adalimumab or PNGase F treated

adalimumab (1 μg/μL)

Conclusion

A rapid 30-min separation on TSKgel FcR-IIIA-NPR allows the analysis

of large numbers of mAb samples to gain valuable fi rst information on

the distribution of glycoforms and expected ADCC activity. This fast and

effi cient method can be applied to purifi ed samples and supernatant

alike and can be used in many phases of development and production

such as cell line screening in early R&D, biosimilar or originator

comparison, upstream development and optimization, monitoring of

glycoengineering, or lot-to-lot comparison in QC.

Reference

(1) M. Kiyoshi et al., Sci. Rep. 8, 3955 (2018) doi:10:1038/s41598-018-

22199-8

TSKgel is a registered trademark of Tosoh Corporation

Tosoh Bioscience GmbHIm Leuschnerpark 4 64347 Griesheim, Darmstadt, Germany

Tel: +49 6155 7043700 Fax: +49 6155 8357900

E-mail: [email protected]

Website: www.tosohbioscience.de

0 5 10 15 20

Retention Time (min)

Low activityHigh activity

Mid activity

AB

S 280 n

m

10 15 20 35 300

2×105

4×105

6×105

8×105

1×106

0

2×105

4×105

6×105

8×105

1×106

0

20

40

60

Asn

% B

80

100

Detector Respon

se (A

U)

Detector Respon

se (A

U)

Retention Time (min)

Adalimumab

10 15 20 35 300

20

40

60

% B

80

100

Retention Time (min)

Column: TSKgel FcR-IIIA-NPR

Mobile phase A: 50 mmol/L citrate, pH 6.5

Mobile phase B: 50 mmol/L citrate, pH 4.5

Flow rate: 1 mL/min

Detection: UV @ 280 nm

Sample: 50 μL of adalimumab or PNGase F treated adalimumab (1 μg/μL)

Adalimumab treated

with PNGase F

Glycans (+)

Glycans (-)

Figure 1: Separation of mAb glycoforms according

to their affi nity to Fc receptor/ADCC activity.

Figure 2: Analysis of adalimumab and de-glycosylated adalimumab

(Data kindly provided by Leila Ghaleh, TU Darmstadt).

170 THE APPLICATIONS BOOK – MARCH 2019

PHARMACEUTICAL/DRUG DISCOVERY

Reversed-Phase Analyses of Monoclonal Antibodies Using High TemperaturesYMC Europe GmbH

Monoclonal antibodies (mAbs) are immunological active proteins,

which bind specifi cally to certain cells or proteins. This will stimulate

the immune system to attack those targets.

mAbs are very important for the treatment of different kinds of

cancer and autoimmune diseases. Nowadays, a broad variety of

therapeutical antibodies are available on the market and several more

are in research and development.

Due to their molecular weight of about 150 kDa, intact antibodies

are usually analyzed by IEX, SEC, or HIC. In addition, reversed-phase

methods are an easy tool as well.

However, a lack of sensitivity and resolution has been a hurdle in

the past. With modern reversed phases addressing the requirements

of these analytes, it is easy to fi nd a suitable method.

Successful analysis in reversed-phase mode for mAbs is

enhanced by employing a temperature stable (> 60 °C), widepore

stationary phase.

This application note shows how to achieve robust

chromatographic results for two commercially available mAbs:

Adalimumab (Humira®) and Bevacizumab (Avastin®).

The increase in temperatures leads to higher sensitivity and a

sharper peak of Adalimumab at temperatures >60 °C (Figure 1).

1400

1200

1000

800

600

200

400

0

2 4 6 8 10

Time (min)

mA

U

40°C

Adalimumab (MW: ca. 148 kDa)

50°C

60°C

70°C

80°C

90°C

Figure 1: Reversed-phase analysis of Adalimumab.

Table 1: Method details

Column YMC-Triart Bio C4 (3 μm, 30 nm) 150 × 3.0 mm

Part No. TB30S03-1503PTH

EluentA) water–TFA (100:0.1)

B) acetonitrile–TFA (100:0.1)

Gradient

Time (min) Eluent B (%)

0 30

15 60

30 90

Flow Rate 0.4 mL/min

Temperature 25 °C

Detection UV at 220 nm

Injection 4 μL (0.5 mg/mL)

THE APPLICATIONS BOOK – MARCH 2019 171

PHARMACEUTICAL/DRUG DISCOVERY

Bevacizumab shows robust results starting from 70 °C, whereas

at lower temperatures no peak was detected (Figure 2).

With YMC-Triart Bio C4, elevated temperatures can easily

be applied, due to stability up to 90 °C. In addition, the surface

comprising 30 nm/300 Å is benefi cial for resolution. In combination

with a wide pH range of 1–10 YMC-Triart Bio C4 is a well-suited tool

for any mAb (U)HPLC method.

YMC Europe GmbHTel.: +49 2064 4270

E-mail: [email protected]

Website: www.ymc.de

1400

1200

1000

800

600

200

400

0

2 4 6 8 10

Time (min)

mA

U 40°C

50°C

60°C

70°C

80°C

90°C

Bevacizumab (MW: ca. 148 kDa)

Figure 2: Reversed-phase analysis of Bevacizumab.

172 THE APPLICATIONS BOOK – MARCH 2019

GENERAL

Identifying Long-Chain and Short-Chain Branching in PolymersChris Deng, Wyatt Technology

Branching affects macroscopic polymer properties such as crystallinity, melting temperature, toughness, ductility, and optical

clarity. Two types of branching are long-chain branching (LCB) and short-chain branching (SCB), wherein the molar mass of

the branches is larger or smaller than the entanglement molar mass, respectively.

Wyatt Technology Corp.6330 Hollister Avenue, Santa Barbara, California 93117, USA

Tel.: +1 (805) 681 9009 Fax: +1 (805) 681 0123

Website: www.wyatt.com E-mail: [email protected]

Size-exclusion chromatography with multi-angle light scattering

(SEC-MALS) characterizes LCB in macromolecules through

conformation plots: log-log plots of the rms radius (radius of gyration,

Rg) versus molar mass (M). These plots have a characteristic slope

of 0.58 for a linear polymer in a thermodynamically good solvent.

Polymers with LCB exhibit a lower slope than the linear analogue.

The difference in slope between a linear polymer and a branched

polymer can be seen in Figure 1.

SCB does not produce such obvious differences relative to linear

analogues. The following analysis of branched silicones reveals the

telltale characteristics of polymers with SCB.

Materials and Methods

Three separate lots of branched silicone polymers were dissolved in

THF for analysis. The instrument setup consisted of an Agilent 1100

HPLC with two PLgel Mixed-C 300 × 7.5 mm columns combined with

a DAWN® MALS detector and an Optilab® differential refractometer.

Data acquisition and analysis were performed using ASTRA® software.

Discussion

SCB creates a dense structure with smaller radii than linear

polymers of the same molecular weight. However, their overall

conformation remains similar to the linear analogue. As a result,

measurements of two polymers, one with and one without SCB,

produce parallel conformation plots. This phenomenon has been

seen in previous studies of branched polyethylene polymers and

poly(p-methylstyrene) comb polymers (1–3).

Polymers may possess both SCB and LCB. In these cases, the

branched polymer conformation slope is lower than that of the linear

analogue, and the plot is shifted down in size. This is seen in Figure 2,

where silicone polymers with a fixed amount of LCB and increasing

amounts of SCB have slopes lower than 0.58 in the conformation

plot. As the amount of SCB increases, the intrinsic viscosity [η] at a

given molar mass decreases, while the slopes remain parallel.

Conclusions

Light scattering implemented as SEC-MALS can effectively and

rapidly characterize branching in polymers. Polymers with LCB

exhibit lower slopes than the corresponding linear polymer, which

differ depending on the extent of LCB. Polymers containing only

SCB exhibit conformation plots parallel to those of their linear

analogues, shifted down in Rg.

If polymers possess both SCB and LCB, then their slopes will be

less than the linear analogue slope, and the plot will be shifted down

relative to non-SCB analogues. Hence SEC-MALS is an essential tool for

analyzing such polymers. For smaller polymers with radii below 10 nm,

a differential viscometer such as a ViscoStar is added to the system to

plot the logs of intrinsic viscosity, rather than radius, versus molar mass.

References

(1) Y. Yu, P.J. Deslauriers, and D.C. Rohlfing, Polymer 46(14), 5165–5182

(2005).

(2) T. Sun et al., Macromolecules 34, 6812–6820 (2001).

(3) W. Radke and A.H.E. Müller, Macromolecules 38, 3949–3960 (2005).

100

10

10 10

Polyethylene

Molar Mass (g/mol)

slopelin

=0.57

slopebr

=0.32

RMS R

ad

ius

(nm

)

5 6

Figure 1: Linear and LCB polyethylene in a conformation

plot. The linear polymer exhibits a slope close to 0.58, which

is the value for a linear polymer in a thermodynamically good

solvent. The branched polymer exhibits a much lower slope.

100

90

80

70

60

50rms

rad

ius

(mm

)

S1

S2

S3

1.0x106

Molar mass (g/mol)5.0x106

40

Figure 2: Conformation plot of three silicone polymers

with both SCB and LCB, two replicates each.

iHILIC -Fusion®

advances HILIC separations in UHPLC and HPLC

HILICONHILICON

μ Excellent durability and ultra-low bleeding for LC-MS

μ iHILIC -Fusion(P): pH 1-10; 5 μm ®

μ iHILIC -Fusion and iHILIC -Fusion(+): pH 2-8; 1.8, 3.5, and 5 μm® ®

μ Charge modulated hydroxyethyl amide/amide HILIC columns

μ Versatile columns for ”Omics” studies and other applications

μ Three complementary selectivities for separation of polar compounds

For more information:

Email: [email protected] | Website: www.hilicon.com© 2019 HILICON AB. All rights reserved. | iHILIC is a registered trademark of HILICON AB, Sweden

®

iHILIC -Fusion®

iHILIC -Fusion(+)®

iHILIC -Fusion(P)®

Silica based Silica based Polymer based

N

NHN

O

NH2

O

OH

OH

N

OO

NH

COOH

O

HN

CONH2

HO

NH

O

OH

O

HN

N

OHO

HN

OOH

NHO

N

O

OH5 proline pyoverdin

www.gerstel.com

3-MCPD? 2-MCPD? Glycidol?

Automated determination of contaminants in edible oil and fat based on ISO 18363-1, AOCS Cd 29c-13, and DGF C-VI 18 (10):

O WorkStation Sample Prep Solution or

O Complete automation incl. GC/MS

O Evaporation step for lowest limits of determination and best system stability

O�Analytical support at your service

Samples are automatically prepared and analyzed, including derivatization and evaporation of excess solvent and reagent for best limits of determination and optimal GC/MS system stability.

What can we do for you?