advances in pharmaceutical analysisimages2.advanstar.com/pixelmags/lcgc-eu/pdf/2016-10-sp.pdf ·...

44
SUPPLEMENT TO Advances in Pharmaceutical Analysis October 2016 Volume 29 Number s10 www.chromatographyonline.com

Upload: others

Post on 09-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

SUPPLEMENT TO

Advances in

Pharmaceutical Analysis

October 2016

Volume 29 Number s10

www.chromatographyonline.com

Page 2: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Joanna Simpson

comfortable with on the

spot sampling at home

�����������������������������������

���������������������

����������������������������������� ������

�������������������������

Dried blood spot (DBS) sampling is an emerging technology in the clinical and pharmaceutical laboratory,

offering easy sample collection, transport and storage. Our revolutionary DBS Autosampler™ maintains the

integrity of the sample through automation, offering time and cost savings. Innovative patented Flow-through

desorption technology (FTD™)* eliminates tedious punching and costly robotics. Automation of the entire

workflow for DBS analysis in minutes, providing maximum sensitivity without any manual intervention.

BE

TT

ER

SAMPLE

CA

RE

Spark Holland B.V.P.O. box 3887800 AJ EmmenThe Netherlands

P. +31 591 631 700F. +31 591 630 035E. [email protected]. www.sparkholland.com

Head Offi ce:P. de Keyserstraat 87825 VE EmmenThe Netherlands

*US 8586382 B2

Page 3: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Ion chromatography? Metrohm is the better deal!

• Because we make sure your applications

run on your Metrohm IC – guaranteed!

• Because when you have a question, our

specialists are just a phone call or visit

away – guaranteed!

• Because we train you to become an IC

expert yourself – guaranteed!

www.metrohm.com

Ask for

a free demo

now!

Page 4: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

4 Advances in Pharmaceutical Analysis October 2016

Advances in

Pharmaceutical Analysis

8 Advances in Pharmaceutical Analysis: Introduction

Pat Sandra and Davy Guillarme

An introduction from the guest editors of this special supplement

9 Advances in Sample Preparation for Biological Fluids

Lucie Nováková

This article explores microextraction techniques, selective approaches, on-line sample preparation, and dried

matrix spots that aim to provide solutions to sample preparation problems in bioanalysis.

16 Quality by Design: A Tool for Separation Method Development in Pharmaceutical Laboratories

Karen Gaudin and Ludivine Ferey

The trends observed when applying QbD to the development of separation methods in pharmaceutical

analysis are discussed.

26 High-Throughput Analysis of Drugs and Metabolites in Biological Fluids Using Quan–Qual Approaches

Ronald de Vries, Rob J. Vreeken, and Filip Cuyckens

Recent high-resolution MS instruments can be used for both quantitative and qualitative measurements, making

these a worthwhile alternative to triple quadrupole MS systems for quantitation, as well as a powerful tool for

identification of unknowns within the same analysis.

31 Contemporary Analysis of Chiral Molecules

Eric Francotte

The state-of-the-art in chiral recognition separation methods with an emphasis on LC and SFC is presented.

38 Characterization of Counterfeit and Substandard Medicines Using Capillary Electrophoresis

Julie Schappler and Serge Rudaz

As a simple, reliable, and cost-efficient technique, CE can offer significant benefits in counterfeit drug analysis.

Ph

oto

Cre

dit: G

ett

yp

ho

to/G

ett

y Im

ag

es

Page 5: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

PHARMACEUTICAL Q� HEALTH SCIENCES Q� FOOD Q� ENVIRONMENTAL Q� CHEMICAL MATERIALS

©2016 Waters Corporation. Waters and The Science of What’s Possible are registered trademarks of Waters Corporation.

INNOVATIVE

DMPK SOLUTIONS

Page 6: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

6 Advances in Pharmaceutical Analysis October 2016

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

5IF�1VCMJTIFST�PG�-$t($�&VSPQF�XPVME�MJLF�UP�UIBOL�UIF�NFNCFST�PG�UIF�&EJUPSJBM�"EWJTPSZ�#PBSE�GPS�

UIFJS�DPOUJOVJOH�TVQQPSU�BOE�FYQFSU�BEWJDF��5IF�IJHI�TUBOEBSET�BOE�FEJUPSJBM�RVBMJUZ�BTTPDJBUFE�XJUI�

-$t($�&VSPQF�BSF�NBJOUBJOFE�MBSHFMZ�UISPVHI�UIF�UJSFMFTT�FGGPSUT�PG�UIFTF�JOEJWJEVBMT�

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

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

LOPXMFEHF�UP�HBJO�DPNQFUJUJWF�BEWBOUBHF��0VS�TDJFOUJGJD�RVBMJUZ�BOE�DPNNFSDJBM�PCKFDUJWJUZ�QSPWJEF�

SFBEFST�XJUI�UIF�UPPMT�OFDFTTBSZ�UP�EFBM�XJUI�SFBM�XPSME�BOBMZTJT�JTTVFT �UIFSFCZ�JODSFBTJOH�UIFJS�

efficiency, productivity and value to their employer.

Kevin Altria

(MBYP4NJUI,MJOF �)BSMPX �&TTFY �6,

Daniel W. Armstrong

6OJWFSTJUZ�PG�5FYBT �"SMJOHUPO �5FYBT �64"

Günther K. Bonn

*OTUJUVUF�PG�"OBMZUJDBM�$IFNJTUSZ�BOE�

3BEJPDIFNJTUSZ �6OJWFSTJUZ�PG�*OOTCSVDL �

"VTUSJB

Deirdre Cabooter

Department of Pharmaceutical and

1IBSNBDPMPHJDBM�4DJFODFT �6OJWFSTJUZ�PG�

-FVWFO �#FMHJVN

Peter Carr

%FQBSUNFOU�PG�$IFNJTUSZ �6OJWFSTJUZ

PG�.JOOFTPUB �.JOOFBQPMJT �.JOOFTPUB �64"

Jean-Pierre Chervet

"OUFD�-FZEFO �;PFUFSXPVEF �5IF�

Netherlands

Jan H. Christensen

Department of Plant and Environmental

4DJFODFT �6OJWFSTJUZ�PG�$PQFOIBHFO �

Copenhagen, Denmark

Danilo Corradini

Istituto di Cromatografia del CNR, Rome,

Italy

Hernan J. Cortes

H.J. Cortes Consulting,

.JEMBOE �.JDIJHBO �64"

Gert Desmet

5SBOTQPSU�.PEFMMJOH�BOE�"OBMZUJDBM�

4FQBSBUJPO�4DJFODF �7SJKF�6OJWFSTJUFJU �

#SVTTFMT �#FMHJVN

John W. Dolan

LC Resources, Walnut Creek, California,

64"

Anthony F. Fell

Pharmaceutical Chemistry,

6OJWFSTJUZ�PG�#SBEGPSE �#SBEGPSE �6,

Attila Felinger

Professor of Chemistry, Department of

"OBMZUJDBM�BOE�&OWJSPONFOUBM�$IFNJTUSZ

6OJWFSTJUZ�PG�1ÏDT �1ÏDT �)VOHBSZ

Francesco Gasparrini

Dipartimento di Studi di Chimica e

5FDOPMPHJB�EFMMF�4PTUBO[F�#JPMPHJDB-

NFOUF�"UUJWF �6OJWFSTJU�i-B�4BQJFO[Bw �

Rome, Italy

Joseph L. Glajch

Momenta Pharmaceuticals, Cambridge,

.BTTBDIVTFUUT �64"

Jun Haginaka

School of Pharmacy and Pharmaceutical

4DJFODFT �.VLPHBXB�8PNFO�T�

6OJWFSTJUZ �/JTIJOPNJZB �+BQBO

Javier Hernández-Borges

%FQBSUNFOU�PG�"OBMZUJDBM�$IFNJTUSZ �

/VUSJUJPO�BOE�'PPE�4DJFODF�6OJWFSTJUZ�PG�

Laguna, Canary Islands, Spain

John V. Hinshaw

4FSWFSPO�$PSQ� �)JMMTCPSP �0SFHPO �64"

Tuulia Hyötyläinen

VVT Technical Research of Finland,

Finland

Hans-Gerd Janssen

7BO�U�)PGG�*OTUJUVUF�GPS�UIF�.PMFDVMBS�

4DJFODFT �"NTUFSEBN �5IF�/FUIFSMBOET

Kiyokatsu Jinno

School of Materials Sciences, Toyohasi

6OJWFSTJUZ�PG�5FDIOPMPHZ �+BQBO

Huba Kalász

4FNNFMXFJT�6OJWFSTJUZ�PG�.FEJDJOF

#VEBQFTU �)VOHBSZ

Hian Kee Lee

/BUJPOBM�6OJWFSTJUZ�PG�4JOHBQPSF �

Singapore

Wolfgang Lindner

*OTUJUVUF�PG�"OBMZUJDBM�$IFNJTUSZ �

6OJWFSTJUZ�PG�7JFOOB �"VTUSJB

Henk Lingeman

'BDVMUFJU�EFS�4DIFJLVOEF �'SFF�6OJWFSTJUZ �

"NTUFSEBN �5IF�/FUIFSMBOET

Tom Lynch

#1�5FDIOPMPHZ�$FOUSF �1BOHCPVSOF �6,

Ronald E. Majors

"OBMZUJDBM�DPOTVMUBOU �8FTU�$IFTUFS

1FOOTZMWBOJB �64"

Debby Mangelings

%FQBSUNFOU�PG�"OBMZUJDBM�$IFNJTUSZ�

BOE�1IBSNBDFVUJDBM�5FDIOPMPHZ �7SJKF�

6OJWFSTJUFJU �#SVTTFMT �#FMHJVN

Phillip Marriot

.POBTI�6OJWFSTJUZ �4DIPPM�PG�$IFNJTUSZ �

7JDUPSJB �"VTUSBMJB

David McCalley

%FQBSUNFOU�PG�"QQMJFE�4DJFODFT �

6OJWFSTJUZ�PG�8FTU�PG�&OHMBOE �#SJTUPM �6,

Robert D. McDowall

.D%PXBMM�$POTVMUJOH �#SPNMFZ �,FOU �6,

Mary Ellen McNally

%V1POU�$SPQ�1SPUFDUJPO /FXBSL �

%FMBXBSF �64"

Imre Molnár

.PMOBS�3FTFBSDI�*OTUJUVUF �#FSMJO �(FSNBOZ

Luigi Mondello

%JQBSUJNFOUP�'BSNBDP�DIJNJDP �'BDPMU�

EJ�'BSNBDJB �6OJWFSTJU�EJ�.FTTJOB �

Messina, Italy

Peter Myers

Department of Chemistry,

6OJWFSTJUZ�PG�-JWFSQPPM �-JWFSQPPM �6,

Janusz Pawliszyn

%FQBSUNFOU�PG�$IFNJTUSZ �6OJWFSTJUZ�PG�

Waterloo, Ontario, Canada

Colin Poole

8BZOF�4UBUF�6OJWFSTJUZ �%FUSPJU

.JDIJHBO �64"

Fred E. Regnier

%FQBSUNFOU�PG�#JPDIFNJTUSZ �1VSEVF�

6OJWFSTJUZ �8FTU�-BGBZFUUF �*OEJBOB �64"

Harald Ritchie

5SBKBO�4DJFOUJGJD�BOE�.FEJDBM �.JMUPO�

,FZOFT �6,

Koen Sandra

Research Institute for Chromatography,

,PSUSJKL �#FMHJVN

Pat Sandra

Research Institute for Chromatography,

,PSUSJKL �#FMHJVN

Peter Schoenmakers

Department of Chemical Engineering,

6OJWFSTJUFJU�WBO�"NTUFSEBN �"NTUFSEBN �

The Netherlands

Robert Shellie

5SBKBO�4DJFOUJGJD�BOE�.FEJDBM �

3JOHXPPE �7JDUPSJB �"VTUSBMJB

Yvan Vander Heyden

7SJKF�6OJWFSTJUFJU�#SVTTFM �

#SVTTFMT �#FMHJVN

Editorial Advisory Board

Published by

UBM EMEA

Vice President/Group Publisher

Mike Tessalone

[email protected]

Editorial Director

Laura Bush

[email protected]

Editor-in-Chief

Alasdair Matheson

[email protected]

Managing Editor

Kate Mosford

[email protected]

Assistant Editor

Lewis Botcherby

[email protected]

Sales Manager

Oliver Waters

[email protected]

Sales Executive

Liz Mclean

[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, UKTel. +44 (0)151 353 3500

Fax +44 (0)151 353 3601

Subscribe on-line at

www.chromatographyonline.com

46#4$3*15*0/4��-$r($�&VSPQF�JT�GSFF�UP�RVBMJàFE�SFBEFST�JO�&VSPQF� To apply for a free subscription, or to change your name or address, go to

XXX�DISPNBUPHSBQIZPOMJOF�DPN �DMJDL�PO�4VCTDSJCF �BOE�GPMMPX�UIF�QSPNQUT�

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

NBHB[JOFT!TVQFSGJMM�DPN �QVUUJOH�-$&�JO�UIF�TVCKFDU�MJOF�

Please quote your subscription number if you have it.

MANUSCRIPTS:�'PS�NBOVTDSJQU�QSFQBSBUJPO�HVJEFMJOFT �WJTJU�XXX�DISPNBUPHSBQIZPOMJOF�DPN�PS�

DBMM�UIF�&EJUPS ��������������������"MM�TVCNJTTJPOT�XJMM�CF�IBOEMFE�XJUI�SFBTPOBCMF�DBSF �CVU�

UIF�QVCMJTIFS�BTTVNFT�OP�SFTQPOTJCJMJUZ�GPS�TBGFUZ�PG�BSUXPSL �QIPUPHSBQIT�PS�NBOVTDSJQUT��&WFSZ�

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

����NJOJNVN��$POUBDU�#SJBO�,PMC�BU�8SJHIU�T�.FEJB ������5JNCFSMPDI�1MBDF �5IF�8PPEMBOET �59�

�������5FMFQIPOF���������������FYU�������&NBJM��CLPMC!XSJHIUTNFEJB�DPN�

ª������"EWBOTUBS�$PNNVOJDBUJPOT�6,�-UE��"MM�SJHIUT�SFTFSWFE��/P�QBSU�PG�UIJT�

publication may be reproduced in any material form (including photocopying or storing it in any

NFEJVN�CZ�FMFDUSPOJD�NFBOT�BOE�XIFUIFS�PS�OPU�USBOTJFOUMZ�PS�JODJEFOUBMMZ�UP�TPNF�

PUIFS�VTF�PG�UIJT�QVCMJDBUJPO�XJUIPVU�UIF�XSJUUFO�QFSNJTTJPO�PG�UIF�DPQZSJHIU�PXOFS�

FYDFQU�JO�BDDPSEBODF�XJUI�UIF�QSPWJTJPOT�PG�UIF�$PQZSJHIU �%FTJHOT���1BUFOUT�"DU�

6,������PS�VOEFS�UIF�UFSNT�PG�B�MJDFODF�JTTVFE�CZ�UIF�$PQZSJHIU�-JDFOTJOH�

"HFODZ ����5PUUFOIBN�$PVSU�3PBE �-POEPO�8�1��-1 �6,���"QQMJDBUJPOT�GPS�UIF�

DPQZSJHIU�PXOFS�T�QFSNJTTJPO�UP�SFQSPEVDF�BOZ�QBSU�PG�UIJT�QVCMJDBUJPO�TIPVME�CF�

GPSXBSEFE�JO�XSJUJOH�UP�1FSNJTTJPOT�%FQU �)JOEFSUPO�1PJOU �-MPZE�%SJWF �&MMFTNFSF�

1PSU �$IFTIJSF �$)����)2��8BSOJOH��5IF�EPJOH�PG�BO�VOBVUIPSJ[FE�BDU�JO�SFMBUJPO�UP�B�

DPQZSJHIU�XPSL�NBZ�SFTVMU�JO�CPUI�B�DJWJM�DMBJN�GPS�EBNBHFT�BOE�DSJNJOBM�QSPTFDVUJPO�

10% Post

Consumer

Waste

Page 7: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Absolute Molecular Weights andSizes with your UHPLC…

and our ģDAWN™ MALS Detector

Absolute molar

masses: 200 g/mol

to 1x107 g/mol

RMS radii (rg):

10 to 50 nm

Shapes and

branching ratios

Protein conjugates,

copolymers and more

www.wyatt.com | [email protected] | 805-681-9009

Page 8: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

In the LCGC Europe October

2015 supplement “Advances in

Biopharmaceutical Analysis”, the

analytical challenges in the analysis

of large and heterogeneous protein

biopharmaceuticals were addressed.

Biopharmaceuticals are indeed

the fastest growing area within the

pharmaceutical industry but innovation

in small-molecule drugs is still of great

importance in the sector. It is estimated

that research in both fields, small and

large, will recognize an even distribution.

This supplement therefore focuses on

recent advances in small-molecule drug

analysis, complementing the October

2015 supplement.

Giving a complete and detailed

overview of all innovations in

contemporary analytical methods

for pharmaceutical analysis in a

supplement is unrealistic and this

issue is therefore based on selecting

some important keywords within this

research domain: sample preparation

for biological fluids, quality by

design (QbD), the power of mass

spectrometry (MS), chiral recognition,

and considerations on the analysis of

counterfeits medicines.

Sample preparation is the first

step in an analytical procedure for

biofluids and its importance is often

underestimated. The first contribution,

authored by Lucie Nováková, provides

an overview of conventional sample

preparation techniques and presents

the most recent developments for high

throughput bioanalysis. Microextraction

techniques, on-line sample preparation,

and selective methods, as well as

dried matrix (blood) spot analyses, are

presented in detail.

Analytical procedures are critical

processes in drug development and

quality control. Quality by design (QbD)

is gaining widespread acceptance

in the pharmaceutical industry

and is also more and more widely

implemented in analytical work. Karen

Gaudin and Ludivine Ferey describe

the present trends in applying QbD

to separation methods (particularly

in liquid chromatography [LC]) in

pharmaceutical analysis. A detailed

description of tools involved in QbD

methods are presented, and the

benefits of implementing QbD in the

laboratory are provided.

Developments in MS have gained

momentum in recent years and

are very rapidly implemented into

pharmaceutical laboratories. Ronald

de Vries, Rob J. Vreeken, and

Filip Cuyckens show how recent

high-resolution MS instruments can

be used for both quantitative and

qualitative measurements, making

these a worthwhile alternative to “the

golden standard” triple quadrupole

MS systems for quantitation, as well

as a powerful tool for identification of

unknowns within the same analysis.

A significant number of

small-molecule drugs are single

enantiomers and chiral resolution

methods are required during the

synthetic routes of these drugs and

for the final active pharmaceutical

ingredient (API). Eric Francotte is an

expert in chiral separation methods

and he describes the state-of-the-art in

chiral recognition separation methods

with emphasis on LC and supercritical

fluid chromatography (SFC). This

contribution is a follow-up of the work

described in LCGC Europe April 2016

entitled: “Practical Advances in SFC

for the Purification of Pharmaceutical

Molecules” (1).

Counterfeits or fake medicines

are a growing problem, especially in

developing regions. They may contain

the active ingredients in a lower dose

or even not at all, they may contain

another active ingredient, or they may

be contaminated with toxic ingredients.

Because counterfeits are produced

under uncontrolled conditions and

have not been assessed by health

authorities, they may be very harmful to

the health of users. Control of imported

medicines in developing countries is

therefore of the utmost importance,

but the analytical technique must

be sufficiently simple, reliable, and

cost-efficient. Julie Schappler and

Serge Rudaz describe how capillary

electrophoresis (CE) can fulfil these

requirements. As CE is now recognized

as a valuable technique by numerous

Pharmacopoeia, it is indeed a good

candidate in counterfeit analysis.

We hope that the contributions in this

supplement are of interest to the LCGC

Europe readers. Editing and reviewing

the contributions was a pleasure for

us and we thank our colleagues for an

excellent job.

Reference(1) E. Francotte, LCGC Europe 29(4),

194–204 (2016).

Advances in Pharmaceutical AnalysisPat Sandra1 and Davy Guillarme2, 1Research Institute for Chromatography, Kortrijk, Belgium, 2University of Geneva,

University of Lausanne, Geneva, Switzerland

An introduction from the guest editors of this special supplement from LCGC Europe revealing recent

developments in small-molecule drug analysis.

An additional paper titled

“On-Line Two-Dimensional

Liquid Chromatography

(2D-LC) in the

Analysis of Pharmaceuticals”

by Pat Sandra,

Gerd Vanhoenacker,

Mieke Steenbeke, Frank David,

Koen Sandra, Claudio Brunelli,

and Roman Szucs will be

featured in an upcoming

issue of LCGC Europe.

Advances in Pharmaceutical Analysis October 20168

Pat Sandra Davy Guillarme

Page 9: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

The determination of drug

concentrations in biological matrices

is an important aspect of the drug

development process. Bioanalytical

methods are needed to obtain

information on drug profiles from

animal toxicokinetic studies and from

clinical trials, including bioequivalence

studies. These results are used to

make critical decisions supporting

the safety and efficacy of a drug

substance or drug product. Biological

fluids are not convenient for direct

analysis using instrumental methods

because of high sample complexity

and the high content of many

interfering compounds. Generally,

very complex liquid samples including

whole blood, plasma, serum, urine,

or saliva are handled in bioanalysis.

The target analytes are often

present at very low concentrations,

while the interfering compounds

are abundant. Among the most

important, salts and phospholipids are

responsible for matrix effects in liquid

chromatography–mass spectrometry

(LC–MS) analysis. Proteins are

damaging for the analytical

instrumentation because they can

irreversibly adsorb onto the stationary

phase, resulting in a substantial loss

of column efficiency, an increase in

back pressure, or system clogging.

To prevent these issues, adequate

sample preparation is mandatory in

bioanalytical methods (1,2,3).

With a well-designed sample

preparation technique, isolation,

cleanup, and preconcentration of the

analytes of interest from the complex

biological matrices can be achieved,

while the interfering compounds can

be removed. Sample preparation

is an integral part of a bioanalytical

method influencing all further steps

of the analysis, with a crucial impact

on the accuracy and precision of

the final results. Unfortunately, it is

still the most labour-intensive and

time-consuming step of the analytical

procedure, representing 60–80% of

the total analysis time. This is in strong

contrast to modern, fast LC methods

and detection approaches and makes

sample preparation a limiting step to

fast bioanalysis (1,4).

The selection of an appropriate

sample preparation technique is

made with regards to the analyte type,

sample type and amount available,

requested selectivity and sensitivity of

the procedure, extraction time, solvent

consumption, and the possibility of

automation. A well-designed sample

preparation technique should involve a

small amount of sample and a simple

method, which is “just adequate” prior

to analysis. This feature is important

because complicated multistep

procedures can introduce errors.

Sample preparation techniques used

for the treatment of biological fluids

can be classified into two main groups

based on the history and frequency

of their use: conventional methods

and modern approaches to sample

preparation.

Conventional Sample

Preparation Techniques

Conventional sample preparation

approaches involve well-established

and well-optimized techniques,

which are commercially available

and widespread across analytical

laboratories. Straightforward,

reliable, and high-throughput

sample preparation techniques

are a guarantee for achieving

accurate results and meeting

rigorous requirements for method

validation in bioanalysis (5,6) within

a reasonable time frame. These

conventional approaches involve

protein precipitation (PP), liquid–liquid

extraction (LLE), and solid-phase

extraction (SPE) (1,7,8).

Protein Precipitation (PP): Protein

precipitation (PP) followed by

centrifugation (or another precipitate

separation step) belongs among

the leading sample preparation

techniques in modern bioanalysis,

despite its very low selectivity and

clean-up efficiency. Important

benefits, such as very fast sample

treatment, easy and fast method

optimization, minimum number of

steps, and no requirement for special

equipment has brought this method to

the attention of bioanalytical scientists.

However, method sensitivity might

be compromised and matrix effects

may be serious. Another important

drawback of PP is the difficulty of

automating the centrifugation step. To

facilitate the separation of precipitated

phase and supernatant, new solutions

have recently become available in the

form of well-plates to enable filtration

of the precipitated samples (1,7,8).

Liquid–Liquid Extraction (LLE):

Liquid–liquid extraction is based

on a transfer of the analyte from the

aqueous sample to a water-immiscible

solvent. Because of the high

consumption of organic solvents

and thus the production of a large

Advances in Sample Preparation for Biological FluidsLucie Nováková, Department of Analytical Chemistry, Faculty of Pharmacy, Charles University, Hradec Králové,

Czech Republic

Sample preparation techniques in bioanalysis are multistep, time-consuming, and labour-intensive

procedures that can take up 60–80% of the total analysis time. Sample preparation is often the limiting

step of fast bioanalysis and the most error-prone part of the analytical method. There is currently a

focus on improving the sample preparation process by shortening sample preparation time, cutting the

cost of analysis, decreasing sample volume and solvent consumption, reducing the number of sample

preparation steps, and adapting the whole process for automation. This article explores microextraction

techniques, selective approaches, on-line sample preparation, and dried matrix spots that aim to provide

solutions to sample preparation problems in bioanalysis.

9www.chromatographyonline.com

Ph

oto

Cre

dit: sim

on

257

9/G

ett

y Im

ag

es

Page 10: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

volume of environmental pollutants in

a conventional LLE setup, miniaturized

versions of LLE are currently preferred.

Such extraction is performed in glass

vials or small test tubes (9) using

typically 50–100 μL of sample and

about 600–2000 μL of organic solvent

(10,11). The amount of solvents and

sample can be further decreased in

modern microextraction approaches

(see “Microextraction Techniques

Based on LLE”).

LLE is a simple and straightforward

extraction technique. Advantages

include no requirement for special

equipment or skills, lower cost

compared to SPE, and potential to

remove matrix effects in

LC–MS because ionized compounds

such as salts do not partition

into the organic layer (3). Some

disadvantages arise when large-scale

LLE is performed, such as bubble

and emulsion formation. The high

consumption of toxic organic solvents

and production of a large amount of

organic waste make LLE expensive

and environmentally harmful.

Another principal drawback of LLE

is its unsuitability for hydrophilic

compounds, which might be a

significant issue in bioanalysis

because some drugs and many

metabolites possess polar structures.

Last, but not least, most of the LLE

procedures require the evaporation

of nonpolar organic solvent and its

reconstitution in the mobile phase

to reach sample preconcentration

and to obtain a sample compatible

for injection into the LC system.

This step substantially prolongs the

extraction time and may be critical

for sample recovery because of

solubility issues (1,7,8,9). In order

to improve the extraction efficiency,

salting-out (SALLE) (12), sugaring-out

(SULLE) (13), or in-vial derivatization

approaches may be applied in LLE (9).

Although LLE is widely and

commonly used in bioanalysis, its

on-line coupling and full automation is

difficult and requires substantial effort.

An important approach to facilitate

and automate LLE is supported liquid

extraction (SLE). SLE columns contain

specially processed wide-pore

diatomaceous earth, a chemically inert

matrix, which only acts as a holder for

the aqueous sample. A 48- or 96-well

plate format enables automation

and high sample throughput using

a positive-pressure manifold. The

adoption of this approach has

initially been very slow because of

the well-established conventional

LLE procedures in routine practice.

However, there has been an increased

interest in this arrangement as a result

of this miniaturization and automation

(14,15).

Solid-Phase Extraction (SPE):

SPE is probably the most widely

accepted sample preparation

technique, enabling sample cleanup

and preconcentration of the target

analytes. The extraction of these

analytes is based on partitioning

between a solid phase (SPE cartridge)

and a liquid phase (sample). The

selection of SPE sorbent is the key

factor, defining important parameters

of selectivity, affinity, and capacity.

SPE protocols are more selective

compared to those of LLE because

of the selectivity of these sorbents.

Moreover, cleanup is also enabled

during individual washing steps.

Further advantages include lower

consumption of organic solvents

(compared to conventional LLE),

high recovery, removal of nonvolatile

salts, ease of operation, and ease of

automation (1,16,17).

Currently, SPE is most widely

performed in a conventional way using

SPE cartridges. Other formats involve

SPE discs and well plates. An SPE

extraction protocol involves several

steps, including sorbent activation

and conditioning, sample load, a

washing step, and, finally, elution of

the analytes. It is performed using

an SPE vacuum manifold, and so the

requirements on equipment are slightly

higher than in LLE or PP. It is important

to point out that manual SPE is quite a

time-consuming, multistep procedure,

especially when the evaporation of

SPE eluate and its reconstitution in

mobile phase are needed. SPE is

also relatively expensive because the

cartridges are manufactured

for single use only and require

relatively large amounts of organic

solvents. Therefore, modern

development in SPE is focused on

miniaturization (see “Microextraction

Techniques Based on SPE”),

automation (see “On-Line Sample

Preparation Techniques”), enhanced

selectivity (see “Sample Preparation

with High Selectivity”), and new

materials (see “New Materials in

Sample Preparation”) (1,7).

Because phospholipids are an

important source of matrix effects,

the use of hybrid precipitation and

SPE plates for the simultaneous

removal of precipitated proteins and

phospholipids has recently become

popular (18).

Modern Advances

in Sample Preparation

The research and development

of modern sample preparation

techniques is focused on

miniaturization and facilitation of the

sample preparation step. One of

the main goals is to decrease both

sample and solvent volumes, to be in

agreement with the green principles in

chemistry (19). Further requirements

include simpler equipment, a

reduction of handling steps, and a

shortening of sample preparation time,

with the final objective of reducing

sample preparation cost and the

susceptibility to errors (1,7,8). As

ultrahigh-performance LC (UHPLC)

and other fast-LC approaches are

now replacing conventional high

performance LC (HPLC) methods,

Advances in Pharmaceutical Analysis October 201610

Nováková

Figure 1: (a) Schematic illustration of the extraction well using a supported liquid membrane. (b) PALME setup: (A) 96-well donor plate, (B) acceptor plate with supported liquid membranes, and (C) and lid. Photo provided by Astrid Gjelstad, University of Oslo, Norway.

Page 11: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

there is a need for fast and high

throughput sample preparation

techniques.

Microextraction Techniques Based

on LLE: Microextraction approaches

based on liquid-phase extraction are

a very extensive group of techniques.

They may use very simple devices,

quite a complex setup, or membrane

assistance to achieve better phase

separation (20). These techniques

provide important benefits such

as almost solventless extraction

and very high preconcentration

factors (21). Basic classification of

the liquid-phase microextraction

techniques based on the extraction

principle involves single drop

microextraction (SDME), dispersive

liquid–liquid microextraction (DLLME),

and membrane-supported–LLE.

The membrane techniques include

various hollow-fibre liquid-phase

microextraction (HF-LPME)

configurations (22) and its advanced

variant electromembrane extraction

(EME) (21). The last development

of membrane supported-LLE is

represented by parallel artificial

liquid membrane extraction

(PALME). Despite a growing number

of applications of liquid-phase

microextraction approaches in the

scientific literature, the adoption in

routine bioanalytical laboratories

is much slower. Indeed, their use

may be limited in routine bioanalysis

because of the following drawbacks:

manipulation with the devices may

require skilful operation; method

development may be time-consuming

in more complex setups; longer

extraction times compared to

other techniques to accomplish

the same task; and only a small

preconcentration factor obtained

from a low amount of biological

sample compared to other matrices,

where the enrichment factors

can be much higher. Among the

discussed LLE-based microextraction

approaches, PALME (23,24), EME (25)

— both in 96-well plate format — and

DLLME show the greatest potential

in bioanalysis. The latter has been

more widely applied in many fields

and also in bioanalysis, particularly

because of its speed and ease of use,

which is unique among the LLE-based

microextraction approaches (20).

To perform DLLME, an immiscible

solvent and a miscible disperser

solvent are injected into an aqueous

sample, leading to the formation of

a cloudy solution of fine droplets of

an extraction solvent. As a result of

the high surface contact between

extraction solvent droplets and

aqueous samples, high recovery and

enrichment factors are obtained even

when using very small volumes of the

extraction solvent (20).

PALME has been recently

developed to take advantage of

clean extracts obtained when using

back-extraction and to facilitate the

automation of the LLE procedure

(23,24). Similar to HF-LPME, the

analytes are extracted with the help of

a supported liquid membrane (SLM).

The extraction setup is composed of

a 96-well donor and acceptor plates

(Figure 1). The acceptor plate contains

96 artificial membranes that are

impregnated with an organic solvent

to create the SLMs. The pH gradient

across the SLM serves as the driving

force for mass transfer. The aqueous

acceptor solution can be directly

analyzed with LC–MS/MS (23).

EME is another technique using

a similar setup to the one used in

HF-LPME (SLM) with the addition of a

power supply and two electrodes. A

power supply provides d.c. potential

to enhance the extraction rate of

ionizable analytes from the donor

solution (sample) to the acceptor

phase, which is contained in the

lumen of a hollow fibre (25).

Microextraction Techniques Based

on SPE: Microextraction approaches

based on SPE involve three different

groups of extraction techniques

using diverse extraction principles. A

miniaturized version of SPE is applied

in microextraction by packed syringe

(MEPS) and μ-SPE in pipette tips

(μ-SPE-PT). In MEPS, the extraction

is made using a specially adapted

syringe handled manually or with an

automated analytical syringe (26,27).

The solid packing material (1–4 mg)

is inserted into the barrel of a syringe

as a plug or between the needle and

the barrel as a cartridge. In μ-SPE-PT,

the sorbent is firmly placed into

the pipette tip. Both commercially

available or laboratory-made sorbents

can be used for extraction. A great

advantage of the latter is the option

to tailor-make sorbents using a

combination of different chemistries

(Figure 2). This approach originates

from proteomics and is sometimes

designated as STop And Go

11www.chromatographyonline.com

Nováková

(a) (b)

(c)

Figure 2: Microextraction using μ-SPE-PT with laboratory-made pipette tips. (a) Preparation of the pipette tips. (b) Resulting μ-SPE-PT tip containing three combined sorbent layers: C18-polymer-CAX. (c) Extraction procedure using centrifuge as a driving force. Photos and figure provided by Ota Blahoušek and Ondrej Novák, Palacky University Olomouc.

Page 12: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Extraction tips (StageTips) (28,29,30).

In μ-SPE-PT extraction procedures,

pipetting or centrifugation may be

used as a driving force to perform

the individual steps of SPE. The latter

enables simultaneous treatment of a

large number of samples (Figure 2[c]).

In both μ-SPE-PT and MEPS

techniques, the same protocol used

for conventional SPE is applied with

much lower sample and solvent

volumes. Straightforward method

development (a result of the similarity

with conventional SPE) is another

very important benefit. It is easy

and straightforward to automate the

procedure in μ-SPE-PT, but somewhat

more challenging for MEPS, requiring

a dedicated autosampler.

The second approach, which is

called dispersive SPE (d-SPE), is

used in μ-dSPE-PT or in QuEChERS

(quick, easy, cheap, effective, rugged,

and safe) extractions. Compared

to the previous approach, it differs

in the sorbent placement and the

extraction process. In μ-dSPE-PT,

the extraction also takes place in the

standard pipette tips. The sorbent

is loosely placed between the two

frits. This variant is also designated

as disposable pipette tips extraction

(DPX). However, sorbent price is

higher and stationary phase choice is

more limited compared to traditional

SPE (31). Individual extraction steps

are performed using a standard

pipette, which allows the sorbent

to be dynamically mixed with the

extracted material and leads to a rapid

adsorption equilibrium between solid

phase and the analyte. In this way,

every sorbent particle actually faces

analyte several times. This is contrary

to conventional SPE, where a greater

amount of sorbent is needed because

the analyte comes into contact with

the sorbent particles only once. This

extraction procedure leads to fast and

efficient extractions (8).

The third solid-phase–based

extraction principle is different from

conventional SPE. Here, adsorption,

absorption, and desorption processes

take place on the solid sorbent,

allowing the need for solvents to be

completely eliminated. The sorbent

is placed on the fused-silica fibre in

the modified syringe in solid-phase

microextraction (SPME) (32,33), in

a piece of capillary in in-tube SPME

(32), or on the stir bar in stir-bar

sorptive extraction (SBSE) (34).

In SPME, the coated fibre can be

moved in and out using a plunger.

Using such simple equipment, all

steps — extraction, preconcentration,

derivatization, and transfer to

the chromatographic system —

are integrated into one device.

Various materials such as PDMS

(polydimethylsiloxane) or many other

materials may be used for the fibre

coating. New types of SPME fibres

use biocompatible coatings prepared

by immobilizing various sorbents with

polyacrylonitrile (35). Since 1990,

when SPME was first introduced,

a series of modifications and

improvements have been proposed.

Initially, either direct immersion

(DI-SPME) or head-space fibre

(HS-SPME) were applied; however,

in vivo SPME sampling devices

and fully automated 96-well format

multifibre SPME are now preferred

(33). The main benefits of SPME

include no need for solvents, the ease

of automation, minimal equipment

requirements, good linearity, and

relatively high sensitivity. Some of

the drawbacks include a longer time

needed for extraction, limited capacity

and fragility of SPME fibre, generally

lower recoveries than those using

LLE and SPE, and the incidence of

carryover effects. The use of modern

SPME in routine laboratories is still

limited.

On-Line Sample Preparation

Techniques: The use of on-line

sample preparation techniques

reduces sample manipulation and

provides high preconcentration

factors, recoveries, high speed, and

throughput. SPE using 96- or 384-well

plate formats in fully automated robotic

workstations, semi-automated, or in

an off-line configuration is therefore

the most well-established technique in

bioanalysis (36,37).

On-line SPE technology in

combination with LC–MS/MS analysis

has experienced many developments

in recent years and enables faster

and precise determination in both

conventional and miniaturized

arrangements (see “Microextraction

Techniques Based on SPE”). On-line

hyphenation is accomplished using

switching valves and an additional

pump. The experimental parameters

to be optimized in on-line SPE are the

type of sorbent, the solvents used

in the different SPE steps, and their

flow-rates (extraction time). In addition,

further pretreatment, such as filtration,

centrifugation, or protein precipitation,

might sometimes be required prior to

on-line SPE. Other constraints may

include reduced sorbent capacity,

too strong retention, slow kinetic of

the sorption process, and possible

absorption of the analyte on the system

tubes (9,36,38). Specific approaches

to remove macromolecules from the

biological fluids include molecularly

imprinted polymers (MIPs),

restricted-access media (RAM), and

turbulent flow chromatography (TFC).

RAM sorbents are used for the

direct injection of biological fluids

into a chromatographic system to

enable the fractionation of a biological

Advances in Pharmaceutical Analysis October 201612

Nováková

Complex

formation

Cross-linker

Polymerization

Monomer

+

+Template

Template

Template

Template

removal

Figure 3: Schematic procedure of the synthesis of a molecularly imprinted polymer (MIP).

Page 13: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

sample into a protein matrix and analyte fraction. This will

lead to the extraction and enrichment of low molecular

compounds into the interior phase via partition. The

exclusion of macromolecules can be accomplished using

the outer surface of the RAM particle as a physical or a

chemical barrier. The use of RAM sorbents for the direct

and repeated analyses of biological fluids is already well

established. However, the complete elution of the analytes

from RAM on the analytical column might sometimes be

quite challenging (36,39).

TFC enables the direct injection of biological fluids and the

separation of small analyte molecules from the macromolecular

matrix based on the low-diffusion coefficients of proteins

using turbulent flow. The generation of turbulent-flow requires

short, narrow-bore columns, packed with large-size particles

(typically 50 mm × 1.0 mm, 20–60 μm) and flow-rates in the

range of 4–5 mL/min. High-molecular-weight compounds are

quickly eluted using a pure water or buffer mobile phase and

are usually directed to the waste. The retained small molecular

weight analytes are subsequently eluted onto an analytical

column for the chromatographic separation using an organic

mobile phase (36).

To increase the limited capacity and facilitate its

automation, on-line coupling of SPME with separation

techniques has been established and termed in-tube

SPME. In this setup, a capillary column is placed as the

injection loop in a standard autosampler. While the typical

advantages attributed to the on-line setup are obtained

with this technique, there are also several drawbacks,

such as the need for more complex instrumentation with

commercially unavailable adjustments; a requirement

for very clean samples, because the capillary can be

easily clogged; low extraction efficiency; selectivity;

and mechanical stability. To overcome these issues,

development is focused on coupling in-tube SPME with

miniaturized LC techniques and the preparation of new

extraction phases (36,40,41).

Sample Preparation with High Selectivity: The selectivity

of SPE can be further improved using specific sorbents,

such as MIPs, immunoaffinity sorbents, or aptamers.

MIPs are stable synthetic polymers possessing

tailor-made recognition sites, which can specifically retain

target analyte molecules. The preparation of MIPs involves

the copolymerization of a complex formed by the template

molecule and a functional monomer with a cross-linking

agent in the presence of a suitable porogenic solvent

(Figure 3). After removing the template, the resulting cavity

is complementary to the target analytes in terms of size,

shape, and functionality. An SPE cartridge is the most

popular form of MIP used in sample preparation. However,

growing interest in miniaturization has led to the use of MIPs

in SPME, SBSE, MEPS, μ-SPE, membranes, and magnetic

beads. High extraction selectivity and capability to

eliminate the matrix effects are definitely the most important

benefits of MIPs. Other advantages include reusability,

ease of use, and low cost of preparation. Some features

that need to be improved in future research include the

need for higher yields of specific binding sites, the need for

some rules for MIP design, and the application of MIPs to

aqueous samples (42,43).

Immunoaffinity-based SPE (IA-SPE) uses sorbents

with attached antibodies to obtain high selectivity from

13www.chromatographyonline.com

Nováková

Ɣ Highly cost effective - ideal for method development

Ɣ Porous, solid-core and biomolecule kit options

ƔWide range of particle sizes and complementary phases

Ɣ Microbore to analytical ids in a wide range of lengths

Ɣ Excellent peak shape, efficiency, reproducibility & lifetime

Contact your local distributor or visit www.ace-hplc.comACE is a registered trademark of Advanced Chromatography Technologies Ltd

ACEMethod Development Kits

Intelligent Solutions forUHPLC & HPLC Method Development

®

Page 14: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

antigen–antibody interaction.

For small molecules, the target

analyte is typically coupled to the

carrier protein (hapten) to achieve

an immune response. While the

cross-reactivity of antibodies with

structural analogues is considered as

a negative feature in immunoassay,

it is exploited in immunoaffinity

extraction. An important limitation

of IA-SPE is the difficult, expensive,

and time-consuming synthesis

of antibodies with no certainty of

success, which probably prevents

wider use of IA-SPE in routine

bioanalytical laboratories (44).

Aptamer-functionalized

materials (AFMs) are promising,

specific-recognition materials in

sample preparation, providing

many advantages such as high

specificity and binding affinity,

good stability, low cost, nontoxicity,

ease of synthesis, and easy and

controllable modification. Aptamers

are the artificial single-stranded

oligonucleotides generated by an in

vitro selection process called SELEX

(systematic evolution of ligands by

exponential enrichment). By folding

into distinct secondary or tertiary

structures, aptamers can bind to

certain targets with extremely high

specificity. So far, they have been

widely used in biosensors or in

immunoassays, while their use in

sample preparation in bioanalysis is

still at the research stage (45,46).

New Materials in Sample

Preparation: New types of materials

have been introduced in line with the

green analytical chemistry concept.

Among them are sol-gel-based

materials, which allow inorganic and

organic-inorganic hybrid polymers

with typically higher thermal and

chemical stability, controlled

morphology, surface properties, and

pore structures to be obtained due to

the controlled synthesis conditions.

These sorbents are successfully used

in SPE, SBSE, SPME, and MEPS (47).

Ionic liquids (ILs) are used to

enhance extraction efficiency and

selectivity in LLE-based procedures

and as sorbents in SPE-based

techniques. They are liquid salts at

room temperature, with a melting point

lower than 100 ºC. Their structure

consists of organic cations derived

from Lewis bases and polyatomic

anions. The addition of different

structures enables their hydrophobic or

hydrophilic abilities to be tailored (47).

Along with the development of

nanotechnology, various nanomaterials

have been introduced into the sample

preparation domain. Nanomaterials

refer to a special kind of materials

with nanometric scales. Compared

to conventional materials, some

exceptional properties, such as ultrahigh

specific area and increased surface

activity, are facilitating the application

of nanomaterials in sample preparation.

Further advantages include tunable

compositions, various morphologies,

and flexible functionalization.

Nanomaterials used in sample

preparation include nanoparticles and

nanoporous materials (48,49).

Magnetic separation techniques

(Figure 4) (50) are an interesting

approach used in sample preparation

and provide several advantages, such

as efficient, gentle, and nondestructive

separation, especially for large

molecules. Magnetic separation

techniques are therefore able to

facilitate or accelerate many separation

and purification procedures because

of rapid isolation of analytes using an

external magnetic field (51,52).

Dried Matrix Spot Analysis:

Dried matrix spot (DMS) analysis

has recently gained attention in

bioanalytical routine laboratories.

Although dried blood spot (DBS)

analysis is the most widely used

(53,54), other matrices, such as dried

saliva spots (DSS) (55), dried plasma

spots (DPS) (56), dried urine spots

(DUS), and even cerebrospinal fluid

(CSF) sample spots (57), have been

successfully analyzed.

The DBS and other matrix spot

sampling is minimally invasive and

uses substantially lower sample

volumes (<100 μL for four spots)

compared to other procedures. DBS

uses capillary blood from a finger

prick with a lancet. After collection,

the samples are dried on the special

sample collection cards. As the

samples are collected, transported,

and stored in dry form, the handling

and storage of such biological

material becomes easier and may

lead to increased stability of some

unstable compounds. For subsequent

analysis, the disk is punched out from

the spot and this disk is extracted for

the isolation of target analytes, which

is typically followed by LC–MS/MS.

Despite some challenges,

such as proper DBS calibration,

analyte stability, the influence of

hematocrit on blood spot size,

and the need for clinical validation

because of the differences between

venous, whole blood, and plasma, its

wider use in bioanalysis is expected

in the future, as it is a fast, relatively

simple, and straightforward sample

preparation technique (53,54).

The importance of microsampling

in bioanalysis is confirmed by the

development of a new microsampling

Advances in Pharmaceutical Analysis October 201614

Nováková

Vortex Magnetic separation

Elution

Discard liquid

supernatant

HPLC

analysis

MNPs ADR Impurities Magnet

Figure 4: A schematic procedure of magnetic SPE. Extraction of Adriamycin hydrochloride from human plasma. Adapted with permission from reference 50.

Page 15: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

approach termed volumetric

absorptive microsampling (VAMS).

This approach should compensate

for the area bias and homogeneity

issues associated with conventional

DBS (58,59).

Conclusions

Modern method development in

bioanalysis is focused on speed

of analysis, efficiency, selectivity,

sensitivity, low cost, miniaturization,

and automation to obtain high sample

throughput and high quality data.

While significant improvements have

been made in instrumental methods

— both in chromatographic separation

and MS/MS detection — the sample

preparation step still remains the most

time-consuming and labour-intensive

aspect of a bioanalytical method,

particularly when compared to

ultra-fast chromatography. The efforts

to improve this situation have resulted

in the development of many new

sample preparation techniques, which

have helped to overcome some of the

drawbacks of the conventional sample

preparation techniques.

References(1) L. Nováková, J. Chromatogr. A 1292, 25

(2013).

(2) V. Pucci, S. Di Palma, A. Alfieri, F. Bonelli,

and E. Monteagudo, J. Pharm. Biomed.

Anal. 50, 867 (2009).

(3) M. Jemal, Z. Ouyang, and Y.-Q. Xia,

Biomed. Chromatogr. 24, 2 (2010).

(4) A. Kaufmann, Trends Anal. Chem. 63, 113

(2014).

(5) Committee for Medicinal Products for

Human Use, Guideline on Bioanalytical

Method Validation (EMA, London, UK,

2011).

(6) U.S Department of Health and Human

Services, Food and Drug Administration,

Guidance for Industry, Bioanalytical Method

Validation (2001).

(7) L. Nováková and H. Vlcková, Anal. Chim.

Acta 656, 8 (2009).

(8) P. L. Kole, G. Venkatesh, J. Kotecha, and

R. Sheshala, Biomed. Chromatogr. 25, 199

(2011).

(9) L. Ramos, J. Chromatogr. A 1221, 84

(2012).

(10) J.-W. Gao, Y.-M. Yuan, Y.-S. Lu, and M.-C.

Yao, Biomed. Chromatogr. 26, 1482 (2012).

(11) A.E.-H. de Mendoza, I. Imbuluzqueta, M.A.

Campanero, D. Gonzalez, A. Vilas-Zornoza,

X. Agirre, H. Lana, G. Abizanda, F. Prosper,

and M.J. Blanco-Prieto, J. Chromatogr. B

879, 3490 (2011).

(12) D. Jain, R. Athawale, A. Bajaj, and S.

Shrikhande, J. Chromatogr. B 970, 86

(2014).

(13) J. Zhang, F. Myasein, H. Wu, and T.A.

El-Shourbagy, Microchem. J. 108, 198

(2013).

(14) S.X. Peng, T.M. Branch, and S.L. King,

Anal. Chem. 73, 708 (2001).

(15) L. Nováková, V. Desfontaine, F. Ponzetto,

R. Nicoli, M. Saugy, J.-L. Veuthey, and

D. Guillarme, Anal. Chim. Acta 915, 102

(2016).

(16) M.C. Henion, J. Chromatogr. A 856, 3

(1993).

(17) A. Andrade-Eiroa, M. Canle, V.

Leroy-Cancellieri, and V. Cerdà, Trends

Anal. Chem. 80, 641 (2016).

(18) C. Bylda, R. Thiele, U. Kobold, and D.A.

Volmer, Analyst 139, 2265 (2014).

(19) J. Płotka, M. Tobiszewski, A.M. Sulej, M.

Kupska, T. Górecki, and J. Namiesnik, J.

Chromatogr. A 1307, 1 (2013).

(20) J.A. Ocaña-Gonzáles, R.

Fernándes-Torres, M.Á. Bello-Lopéz, and

M. Ramos-Payán, Anal. Chim. Acta 905,

8 (2016).

(21) F. Pena-Pereira, I. Lavilla, and C. Bendicho,

Spectrochim. Acta Part B 64, 1 (2009).

(22) S. Pedersen-Bjergaard and K.E.

Rasmussen, J. Chromatogr. B 817, 3

(2005).

(23) K.S. Ask, T. Bardakci, M.P. Parmer,

T.G. Halvorsen, E.L. Øiestad, S.

Pedersen-Bjergaard, and A. Gjelstad, J.

Pharm. Biomed. Anal. 129, 229 (2016).

(24) L.E.E. Eibak, M.P. Parmer, K.E. Rasmussen,

S. Pedersen-Bjergaard, and A. Gjelstad,

Anal. Bioanal. Chem. 406, 431 (2014).

(25) L.E.E. Eibak, K.E. Rasmussen, E.L.

Øiestad, S. Pedersen-Bjergaard, and A.

Gjelstad, Anal. Chim. Acta 828, 46 (2014).

(26) M. Abdel-Rehim, Anal. Chim. Acta 701,

119 (2011).

(27) M.M. Moein, A. Abdel-Rehim, and M.

Abdel-Rehim, Trends Anal. Chem. 67, 43

(2015).

(28) J. Rappsilber, Y. Ishihama, and M. Mann,

Anal. Chem. 75, 663 (2003).

(29) Y. Ishihama, J. Rappsilber, and M. Mann, J.

Proteome Res. 5, 988 (2006).

(30) J. Svacinová, O. Novák, L. Placková,

R. Lenobel, J. Holík, M. Strnad, and K.

Doležal, Plant Methods 8, 17 (2012).

(31) D.C.M. Bordin, M.N.R. Alves, E.G. de

Campos, and B.S. De Martinis, J. Sep. Sci.

39, 1168 (2016).

(32) H. Kataoka and K. Saito, J. Pharm. Biomed.

Anal. 54, 926 (2011).

(33) B. Bojko, E. Cudjoe, G.A. Gómez-Ríos, K.

Gorynski, Ruifen Jiang, N. Reyes-Garcés,

S. Risticevic, É.A.S. Silva, O. Togunde, D.

Vuckovic, and J. Pawliszyn, Anal. Chim.

Acta 750, 132 (2012).

(34) F.J. Camino-Sánchez, R.

Rodríguez-Gómez, A. Zafra-Gómez, A.

Santos-Fandilla, and J.L. Vílchez, Talanta

130, 388 (2014).

(35) F.S. Mirnaghi, Y. Chen, L.M. Sidisky, and J.

Pawliszyn, Anal. Chem. 83, 6018 (2011).

(36) O. Núñez, H. Gallart-Ayala, C.P.B. Martins,

P. Lucci, and R. Busquets, J. Chromatogr.

B 927, 3 (2013).

(37) L. Krcmova, D. Solichova, and P. Solich,

Talanta 115, 973 (2013).

(38) M. Rogeberg, H. Malerod, H.

Rogerg-Larsen, C. Aass, and S.R. Wilson,

J. Pharm. Biomed. Anal. 87, 120 (2014).

(39) S. Souverain, S. Rudaz, and J.-L. Veuthey,

J. Chromatogr. B 801, 141 (2004).

(40) M. Fernández-Amado, M.C. Prieto-Blanco,

P. López-Mahía, S. Muniategui-Lorenzo,

and D. Prada-Rodríguez, Anal. Chim. Acta

906, 41 (2016).

(41) Y. Moliner-Martinez, R. Herráez-Hernández,

J. Verdú-Andrés, and C. Molins-Legua,

Trends Anal. Chem. 71, 205 (2015).

(42) Y. Hu, J. Pan, K. Zhang, H. Lian, and G. Li,

Trends Anal. Chem. 43, 37 (2013).

(43) E. Turiel and A. Martín-Esteban, Anal.

Chim. Acta 668, 87 (2010).

(44) N. Delaunay, V. Pichon, and M.-C. Hennion,

J. Chromatogr. B 15 (2000).

(45) Q. Yuan, D. Lu, X. Zhang, Z. Chen, and W.

Tan, Trends Anal. Chem. 39, 72 (2012).

(46) F. Du, L. Guo, Q. Qin, X. Zheng, G. Ruan,

J. Li, and G. Li, Trends Anal. Chem. 67, 134

(2015).

(47) B.H. Fumes, M.R. Silva, F.N. Andrade, C.E.

Domingues, and F.M. Lanças, Trends Anal.

Chem. 71, 9 (2015).

(48) L. Xu, X. Qi, X. Li. Y. Bai, and H. Liu,

Talanta 146, 714 (2016).

(49) Y. Wen, L. Chen, J. Li, D. Liu, and L. Chen,

Trends Anal. Chem. 59, 26 (2014).

(50) N. Ma, L. Zhang, R. Li, Y. Zhou, Z. Cai, Ch.

Dong, and S. Shuang, Anal. Methods 6(17),

6736–6744 (2014).

(51) J. He, M. Huang, D. Wang, Z. Zhang, and

G. Li, J. Pharm. Biomed. Anal. 101, 84

(2014).

(52) M. Wierucka and M. Biziuk, Trends Anal.

Chem. 59, 50 (2014).

(53) Y. Enderle, K. Foerster, and J. Burhenne, J.

Pharm. Biomed. Anal. in press (2016).

(54) N. Zheng, L. Yuan, Q.C. Ji, H. Magnus,

Y. Song, Ch. Frost, J. Zeng, A.-F. Aubry,

and M.E. Arnold, J. Chromatogr. B 988, 66

(2015).

(55) N. Zheng, J. Zheng, Q.C. Jin, A. Angeles,

A.-F. Aubry, S. Basdeo, A. Buzescu, I.S.

Landry, N. Jariwala, W. Turley, R. Burrell,

and M.E. Arnold, Anal. Chim. Acta in press

(2016).

(56) W. Li, J. Doherty, S. Favara, Ch. Breen, J.

Flarakos, and F.L.S. Tse, J. Chromatogr. B

991, 46 (2015).

(57) B. Rago, J. Liu, B. Tan, and Ch. Holliman,

J. Pharm. Biomed. Anal. 55, 1201 (2011).

(58) P. Dennif and N. Spooner, Anal. Chem.

86(16), 8489–8495 (2014).

(59) L. Mercolini, M. Protti, M.C. Catapano,

J. Rudge, and A.E. Sberna, J. Pharm.

Biomed. Anal. 123, 186 (2016).

Lucie Nováková is an associate

professor in the Department of

Analytical Chemistry, Charles

University, Faculty of Pharmacy in

Hradec Králové, Czech Republic.

She has a Masters degree in

pharmacy and obtained her Ph.D.

in pharmaceutical analysis from

Charles University, Faculty of

Pharmacy. Her research is focused

on fast LC and SFC techniques,

especially UHPLC, UHPSFC, and

their coupling to MS. She is involved

in a wide variety of research projects

focused on pharmaceutical analysis,

plant analysis, doping control, and

bioanalytical methods. An important

part of her research therefore lies in

the sample preparation step, where

the focus is put on the current trends

enabling facilitation, miniaturization,

and reduction of time and sample

requirements. She has published over

75 scientific articles with about 1650

citations and is widely involved in

teaching and education activities, such

as HPLC and SFC training courses,

seminars, and congresses.

15www.chromatographyonline.com

Nováková

Page 16: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

The concept of quality by design

(QbD) was first introduced in 2004 by

the US Food and Drug Administration

(FDA), as an essential pharmaceutical

quality model to be considered in

the development of new products

and processes (1). Later, QbD

was supported by the International

Conference on Harmonization (ICH).

Since the adoption of the ICH Q8

(R2) guideline encouraging the

implementation of a QbD approach

in pharmaceutical development, QbD

has become a key methodology in the

pharmaceutical industry (2). Indeed,

the number of publications discussing

QbD has exponentially increased over

the last 10 years, as shown in Figure 1.

In the ICH Q8 (R2) guideline, QbD

is defined as “a systematic approach

to development that begins with

predefined objectives and emphasizes

product and process understanding

based on sound science and quality

risk management” (2). The main

purpose is to improve the quality of

pharmaceutical products and ultimately

patient safety by designing quality

directly into pharmaceutical processes.

Processes are better understood

compared to the quality-by-testing

(QbT) approach, which is traditionally

implemented in the pharmaceutical

industry. In fact, QbT does not ensure

product quality since processes and

products cannot be sufficiently known

and controlled.

A QbD approach is mainly

applied to the development of

new pharmaceutical formulations,

as encouraged by the specific

guideline (2). However, in the ICH

Q8 (R2) guideline, the need to

gain a greater understanding of

all processes involved in product

development to achieve a proper

control strategy is outlined. By

supporting drug development

and quality control (QC) activities,

analytical procedures form an integral

part of pharmaceutical processes.

Analytical methods play a key role

throughout a drug product life cycle

because they ensure efficacy and

safety of a pharmaceutical product

by including scientific and regulatory

knowledge, as well as QC needs.

As a result, QbD is being adopted

more and more by pharmaceutical

and biopharmaceutical analysts to

achieve a higher quality of analytical

methods and thus a higher quality of

the pharmaceutical product. The ICH

Q8 (R2) guideline is completed by the

ICH Q9 (3) and Q10 (4) guidelines,

which provide a comprehensive

overview of the basic tools that can be

used to effectively build quality into

products.

This article presents examples

of applications of QbD in the

development of separation methods

for pharmaceutical analysis. The

global methodology of QbD is

summarized, followed by a focus on

some essential tools and concepts

related to QbD strategy. Finally,

this article concludes on how QbD

stands out as a powerful tool for the

pharmaceutical analyst.

Applications

Table 1 classifies the use of QbD

for the development of separation

methods dedicated to pharmaceutical

analysis in four main applications: the

simultaneous analysis of an active

pharmaceutical ingredient (API) and

its related substances; multimolecule

separation; enantioselective

separation; and natural product

analysis. All of these applications are

encountered in the pharmaceutical

industry and are challenging because

of the simultaneous analysis of an

important number of molecules and

the difficulty of separating molecules

with closely related properties.

Related substance analysis

represents one of the most

challenging analytical applications

required in QC and research

and development activities. As

these methods are included in

the Common Technical Document

(CTD) of a pharmaceutical product,

the robustness of the method is

an added value. Samples to be

analyzed can be highly complex

because of the combination of

unreacted starting materials,

impurities originating from starting

materials, unreacted intermediates,

reaction byproducts, and degradation

products. Compounds with closely

related structures are present at

very different concentrations in the

Quality by Design: A Tool for Separation Method Development in Pharmaceutical Laboratories

Karen Gaudin and Ludivine Ferey, School of Pharmaceutical Sciences, Bordeaux University, Bordeaux, France

Quality by design (QbD) has gained in importance in the pharmaceutical industry and is supported

by several regulatory documents (ICH, FDA). The aim is to ensure product quality through a better

understanding of products and processes during pharmaceutical development. As analytical procedures

are critical processes of pharmaceutical product development and quality control (QC), QbD has become

a key tool in the development of analytical methods. This article outlines the general trends observed

when applying QbD to the development of separation methods in pharmaceutical analysis. The main

pharmaceutical applications are reviewed along with a detailed description of tools involved in QbD

methodology. A focus on QbD benefits for the pharmaceutical industry is provided.

Ph

oto

Cre

dit: R

alf H

iem

isc

h/G

ett

y Im

ag

es

Advances in Pharmaceutical Analysis October 201616

Page 17: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

samples, making the development of

separation methods difficult. However,

these impurities usually provide no

benefit to patients and can actually

constitute risks to patient safety or

drug efficacy. There is a great need

therefore to be able to control them

during the manufacturing process

and in the final product. To deal with

such expectation, a QbD strategy

has proved to be appropriate. For

the study of related compounds

in a QC context, the most current

techniques coupled to QbD are high

performance liquid chromatography

(HPLC) and ultrahigh-pressure

liquid chromatography (UHPLC)

in reversed-phase mode (5–13).

However, as QbD provides a better

understanding of the development

process, more complex and recent

techniques that suffer from robustness

problems (such as, hydrophilic

interaction liquid chromatography

[HILIC] and capillary zone

electrophoresis [CZE]) could take

advantage of this approach (14–18).

The second important

pharmaceutical application using QbD

is the separation of several APIs for

the detection of counterfeit medicines

(19–23) or for homologous molecule

analysis in QC (24). When a high

number of compounds (above 10)

needs to be simultaneously separated,

the method must be specific and

robust to distinguish one API from

another. QbD is the most appropriate

tool to speed up the development of

such a method, while also improving

knowledge acquired on the analytical

method.

Enantioselective separations

represent a fundamental field of

research in drug analysis because

the potential biological activity of

chiral compounds is mostly down to

one enantiomer only. Optical purity,

that is, enantiomeric excess (ee),

determination represents a major

challenge for the analyst. Indeed,

the main enantiomer is the active

one and is present in high amounts

in the sample, whereas the impurity

enantiomer is expected to be under

0.1% of the main enantiomer. To

quantify the chiral impurity, the

complete separation of the two

enantiomers is required with a low

limit of quantitation for the impurity

enantiomer. However, enantiomers

have closely related physico-chemical

properties, making the separation

challenging. CZE is a powerful

technique for chiral analysis because

of its high efficiency, fast analysis,

and the low amounts of chiral selector

required. Cyclodextrins are the most

commonly used chiral selectors in

CZE. The association of QbD with

CZE proves that robust methods for

enantioseparations can be achieved

(25,26).

The final application combining

QbD and separation method

development in pharmaceutical

analysis is the characterization of

natural products. Traditional medicine

still holds an important place in curing

disease in developing countries. This

also includes occidental countries

with a greater use of phytotherapy

and an increased consumption

of natural medicines and food

supplements. Efficient analytical

methods are required for structural

identification, classification of

varieties or geographical origin, and

purification purposes. The safety and

quality aspects of finished herbal

medicinal products depend on source

materials of high complexity and

variability because they can include

hundreds of constituents. Fast,

selective, and accurate analytical

methods are therefore needed. QbD

can manage this natural heterogeneity

to provide the best controlled

separations (27–29). In addition, the

optimization offered by QbD can

combine separation parameters

with those of the extraction that may

be intercorrelated (27). Therefore,

the optimum condition and the

design space are designed taking

into account the influence of both

separation and extraction methods.

The main use of QbD in the

optimization of separation method

development lies in the separation

of samples composed of a large

number of molecules. This approach

17www.chromatographyonline.com

Gaudin and Ferey

ICH Q8 (R2)

Pharmaceutical development

2009

1990

250

200

150

100

50

0

1995 2000 2005 2010 2015

Final concept paper

ICH Q8 Pharmaceutical development

2003

Year

Nu

mb

er

of

pu

blica

tio

nFigure 1: Evolution of the number of publications based on the QbD approach following its acceptance by regulatory authorities. Source: Scopus search of the chemical abstracts database from 1990–2015.

QbDstrategy

Qualitycontrol

Method goals

Method selection

Risk assessment

Knowledge space

Design space

Method validation

and control

strategy

-�� �%�#!��#������'�&.#������ ���!%�%����(���&�� ��$� &�-�� M� &��'���&�$�%��

-�� ��!&��"%&*#������%��'�e#!$!%����+'��!$�%�-�� �� !"�*����������!�"�'�!&�!!,�!&%

-�� ��.%���(!��!#!���t-�� M� &��)�#!��!&%

-�� Scr��%!%�����!�%�-�� ����$!%�!&%�&����!!��#���&���������$��������� ��%�� �!r� )��!�!&%��&$�!%�

-�� �'!$!,�!&%����!�%�-�� ���'&%����(������$� &�&#&�-�� ���!���!#!��(%�!&%-�� Wo�"!%��'&!%��%�����!�%��'����)�#!��!&%����obustness

-�� ��!!��#��(�#!���!�(�������)-�� M� &����&(!%����)�#(�!&%�&���!�����%���'����&�� ��$� &��&� .%���%��''�&�� �&��$� &���&%�!!&%��&������#��&��� !�)��$� &�� �&�#��� ��!������

Figure 2: QbD approach for analytical method development.

Page 18: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

takes advantage of the design of

experiments to find the optimum

method for the separation and to

provide method robustness. QbD

is a powerful tool that can offer a

guarantee of separation quality.

QbD Methodology

Traditional univariate methods vary

only one factor at a time (OFAT)

between consecutive experiments.

Such method development strategies

can lead to nonoptimized methods

and offer no warranty of robustness.

On the contrary, by conducting a

systematic development, QbD allows

optimal method performance to be

obtained.

An implementation of the QbD

approach during analytical method

development involves several steps,

as described in Figure 2.

QbD requires the determination of

predefined objectives of the analytical

method, called the analytical target

profile (ATP). The intended purpose

of the method can be stability testing,

quantitative determination, or a limit

test. This means that the goals of the

method — including its performance

—must be precisely described before

the development. For separation

methods, the objectives could be

described as the separation of

compounds with an identification

or quantification purpose, with

no specification of the separation

technique at this step.

The method is selected based on its

ability to fulfill the objectives specified

in the ATP. A significant amount of

knowledge of analytical technologies

and the drug helps with the selection

of an appropriate method. Through

method scouting experiments and

prior knowledge, the most suitable

analytical technique is chosen: HPLC,

UHPLC, or CZE. At the same time,

Critical Quality Attributes (CQAs)

are selected. They are defined in

ICH Q8 (R2) as “physical, chemical,

biological, or microbiological property

or characteristic that should be

within an appropriate limit, range,

or distribution to ensure the desired

product quality” (2). These elements

convert the intended purpose

of the method into performance

criteria. Therefore, the CQAs are a

measure of method performance in

accordance with the ATP — in other

words, the method specifications.

In chromatographic methods,

resolution of the critical pairs

(5,7,8,17,18,21,25), time differences

between chromatographic peaks

(19,20,22), analysis time (24–26),

and even robustness as allowed by a

QbD approach (5,21) are examples of

CQAs to evaluate separation quality.

Once the CQAs are selected, the

method under development follows a

Advances in Pharmaceutical Analysis October 201618

Gaudin and Ferey

1

1

1

1

1

-1

-1 -1

0

00

0.5

0.50.5

-0.5

-0.5-0.5

(12)

(11)

(10)

(3)

(7)

(8)

(9)

(6)

(1)

(2)

(5)

(4)

1

0

0

0

X3

X3

X3

X2

X2

X2

X1

X1

X1

-1

-1-1

(a)

(b)

(c)

Figure 3: Response-surface designs for three factors: (a) central composite design (at least 15 experiments), (b) Box-Behnken design (at least 13 experiments), and (c) Doehlert design (at least 13 experiments).

Page 19: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

(QT�RTQFWEV�KPHQTOCVKQP��YYY�XYT�EQO

+/2418+0)�

241&7%6+8+6;�+0�

*2.%�#0#.;5+5�

0GY�JKIJ�TGUQNWVKQP�EQNWOP|VGEJPQNQI[

8GT[�NQY�U[UVGO�FYGNN�XQNWOG

6QQN�NGUU�EQNWOP�ƂVVKPI

5WRGTKQT�TGUQNWVKQP�CPF�UGPUKVKXKV[

%QPVKPWKPI�VJG�VTCFKVKQP�QH�

OCPWHCEVWTKPI�JKIJN[�TQDWUV�*2.%�

U[UVGOU�DWV�PQY�YKVJ�CFFKVKQPCN�

PQXGN�PGY�VGEJPQNQIKGU��6JG�

%JTQOCUVGT7NVTC4U�GHHQTVNGUUN[�

FGNKXGTU�C�VQR�ENCUU�RGTHQTOCPEG�

CPF�VJG�ƃGZKDKNKV[�PGGFGF�HQT�

VQFC[oU�NCDQTCVQT[�

0GGF�C�FGOQPUVTCVKQP!�'OCKN��EJTQOCVQITCRJ["GW�XYT�EQO

“ChromasterUltra Rs

with ChromSwordAuto 5

method development

platform exclusively

available through

VWR.”

Solvent switching

valves

Easy to maintain

6 column switching valve

Ideal for use with

QbD method

development software

ChromSwordAuto 5.0

Continuing the Hitachi tradition of

manufacturing highly robust HPLC

systems but now with additional

novel new technologies. The

ChromasterUltra Rs effortlessly

delivers a top class performance

and the fexibility needed for

today’s laboratory.

Page 20: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

quality risk assessment to investigate

Critical Process Parameters (CPPs).

CPPs are “parameters whose

variability has an impact on CQAs

and therefore should be monitored

or controlled to ensure the process

produces the desired quality” (2).

Risk assessment tools can be used

to identify and rank parameters with

a potential impact on method quality,

based on prior knowledge and

initial experimental data. A fishbone

diagram, also known as Ishikawa

diagram, identifies potential factors

and classifies risks associated with

these factors into groups related

to instrumentation, materials,

methods, chemicals and reagents,

operators, and laboratory environment

(5,9,30,31). Risk factors can be

further ranked and prioritized through

a Failure Mode and Effect Analysis

(FMEA) approach (32). This approach

guarantees that all parameters of the

analytical method were listed and their

influence evaluated.

High-risk parameters, known as

Critical Process Parameters (CPPs),

are then assessed experimentally

through screening designs, which are

first-degree Design of Experiments

(DOEs). For separation methods,

mobile phase or buffer pH often

appears as a CPP when ionizable

compounds have to be separated,

since pH has an impact on resolution

between compounds (5,8–20,25,26).

Others factors identified as CPPs are

flow rate (6,9,27); gradient slope (5,

7–13,19–23,28,29) in reversed-phase

HPLC; percentage organic solvent

in the initial and final mobile phases

(6,10,14,15,28,29) in reversed-phase

HPLC and HILIC–HPLC; buffer

concentration (14–18) in HILIC–HPLC

and CZE; and column or capillary

temperature in HPLC and CZE. During

this stage, the knowledge space is

fixed, which corresponds to the range

of CPP variation.

DOEs based on higher-order

polynomial models are then

performed to optimize CPPs through

a response-surface methodology, that

is, the modelling of the CQAs as a

function of the CPPs.

The main goal of a QbD approach

is, in fine, to define the Method

Operable Design Region (MODR)

or the Design Space (DS). This is

the multidimensional subspace of

the experimental domain where

assurance of analytical method

quality is provided. This domain

is determined using a desirability

function. Further information about

the concept of DS for analytical

methods is provided later in the

article. A detailed review on DS

for analytical methods is also available

in the literature (33). The final step

of the QbD approach is the

validation of the DS to prove

experimentally that the ATP is

achieved inside the DS. The range of

each CPP is thus validated and the

robustness domain of the achieved

method is described.

To use the optimized method

routinely in QC laboratories, a formal

validation is necessary (34–36).

The final step is the implementation

of a control strategy to ensure

that the method will perform as

expected during its routine use

(4). Some responses measuring

method performance, known as

system-suitability criteria, are selected

and monitored at each analytical run

during routine applications. Therefore,

risk assessment and DS definition

involved in QbD approach can help

to identify an appropriate control

strategy (36).

Advances in Pharmaceutical Analysis October 201620

Gaudin and Ferey

4 5

55

50

45

40

35

30

50

45

40

35

30

3.5

Gra

die

nt

slo

pe

pH

pHp

H

3

2.5

0.2 0.3

Imp. A efficiency

Imp. A efficiency

Imp. A efficiency

Imp. A efficiency

Imp. A efficiency

Imp. A efficiency

EtOH volume

EtOH volume

EtOH volume

EtOH volume

EtOH volume

EtOH volume

Mobile phase volume

Mobile phase volume

Mobile phase volume

Mobile phase volume

Mobile phase volume

Mobile phase volume

Imp. C efficiency

Imp. C efficiency

Imp. C efficiency

Imp. C efficiency

Imp. C efficiency

Rs DXM/Imp.A

Rs DXM/Imp.A

Rs DXM/Imp.A

Rs DXM/Imp.A

Rs DXM/Imp.A

Rs DXM/Imp.C

Rs DXM/Imp.C Rs DXM/Imp.C

Rs Imp. A/Imp. C

0.40.25 0.35

Flow rate Flow rate

0.2 0.3 0.40.25 0.35

0.2 0.3 0.40.25 0.35

Flow rate

2 3 42.5 3.5

Gradient slope

2 3 42.5 3.5

Gradient slope

Temperature

Tem

pera

ture

Tem

pera

ture

30 40 5035 45

2 4

44

Figure 4: Contour plots of the seven CQAs of interest (Rs DXM/Imp.A, Rs DXM/Imp.C, Rs Imp.A/Imp.C, Imp.A efficiency, Imp.C efficiency, mobile phase volume, and EtOH volume) as a function of the four studied CPPs. In the white zone, all CQAs fulfill the requirements (Rs >2.5, Imp.A >3000 theoretical plates, Imp.C >30,000 theoretical plates, mobile phase volume <1.2 mL, and EtOH volume <0.35 mL). Adapted with permission from reference 9.

Page 21: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

21www.chromatographyonline.com

Gaudin and Ferey

Table 1: Applications of QbD to the development of separation methods in pharmaceutical analysis

API + Related Substances Analysis

Technique Analyte Matrix Modelling ApproachDesign Space

Determination/ValidationReference

Reversed-

phase

HPLC–DAD

Abacavir, Lamivudine, and Dolutegravir + 11 related substances + excipients

In-house manufactured tablets

Central composite designFactors: pH, column temperature, gradient slope3 levels

Monte Carlo simulationsDetermination of DS through process capability index calculation

(5)

Reversed-

phase

HPLC–DAD

Pitavastatin calcium + 15 related substances

Forced degradationLivalo tablets

Central composite designFactors: % solvent, column temperature, flow rate5 levels

Desirability plotsValidation of DS through 2 verification trials, one fixing all the factors at the higher ranges and another fixing all the factors at the lower ranges

(6)

Reversed-

phase

UHPLC–DAD

Ebastine + related substances

Oro-dispersible tablets

Full factorial designFactors: gradient time, 2 levels, column temperature, 2 levels, ternary composition of the organic eluent, 3 levels, 12 experiments (2 × 2 × 3)

3D resolution mapsValidation of DS through 6 experiments visually selectedSimulated robustness testing (729 experiments) by varying 6 factors

(7)

Reversed-

phase UHPLC–

DAD

Omeprazole + related substances

Standard

Full factorial designFactors: gradient time, 2 levels, column temperature, 2 levels, pH, 3 levels12 experiments (2 × 2 × 3) in two organic solvents

3D resolution mapsValidation of DS through 6 experiments visually selected Simulated robustness testing (729 experiments) by varying 6 factors

(8)

Reversed-

phase

UHPLC–DAD

Dextromethorphan + impurities + excipient

Syrup

Central composite designFactors: flow rate, gradient slope, pH, column temperature5 levels

Response surface overlayValidation of DS through a PB design

(9)

Reversed-

phase

UHPLC–UV

Mixture of benzalkonium chloride

Preserved drug formulation

Stepwise optimization using commercial softwareFactors: pH, gradient slope, % solvent in initial and final mobile phases

Response surface overlayRobustness validation of optimum during validation

(10)

Reversed-

phase

UHPLC–DAD

Amlodipine besylateBisoprolol fumarate+ related substances

Standards

Full factorial designFactors: gradient time, 2 levels, column temperature, 2 levels, pH, 3 levels12 experiments (2 × 2 × 3)

3D resolution mapsSimulated robustness testing (729 experiments) by varying 6 factorsand 3 experiments fixing worst case conditions

(11)

Reversed-

phase

UHPLC–DAD

2 active pharmaceutical ingredients + 9 impurities

Eye drop solution

Full factorial designFactors: gradient time, 2 levels, column temperature, 2 levels, pH, 3 levels12 experiments (2 × 2 × 3)

3D resolution mapsDS validation through 12 experiments by varying the 3 factors at the edge of the DS Simulated robustness testing by varying 6 factors

(12)

Reversed-

phase UHPLC–

UV–TOF-ESI-

MS/MS

Imatinib mesylate + related substances

Forced degradation

Box–Behnken designFactors: gradient slope, column temperature, pH3 levels

Response surface overlayRobustness validation of optimum during validation

(13)

HILIC–

HPLC–UV

Iohexol + related substances

Omnipaque Solution for parenteral use

Box-Behnken designFactors: % solvent, pH, buffer concentration3 levels

Monte Carlo simulationsValidation of DS through a fractional factorial design 2 3-1 Robustness validation of optimum through a PB design

(14)

HILIC–

HPLC–UV

Bilastine + related substances

Nixar tablets

Box-Behnken designFactors: % solvent, pH, buffer concentration3 levels

Monte Carlo simulationsRobustness validation of optimum through a fractional factorial design 2 5-2

(15)

CZE–DADZolmitriptan + 5 related substances

Zomigtablets

Box-Behnken design Factors: buffer concentration, pH, temperature3 levels

Monte Carlo simulationsValidation of DS through a PB design Robustness validation of optimum through a PB design by expanding the number of factors

(16)

CD–CZE–DAD

Metformin hydrochloride + 5 related substances

Metforalcoated tablets

Doehlert design Factors: injection time, 3 levels, anionic CD concentration, 7 levels, buffer concentration, 5 levels, pH, 7 levels

Monte Carlo simulationsValidation of DS through a PB design Robustness validation of optimum through a PB design by expanding the number of factors

(17)

CZE–DADGlibenclamide + 2 related substances

Eugucontablets

Box-Behnken designFactors: voltage, injection time, buffer concentration, pH, 3 levels

Monte Carlo simulationsValidation of DS and robustness of optimum through a 23 full factorial design

(18)

Page 22: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Advances in Pharmaceutical Analysis October 201622

Gaudin and Ferey

Table 1: Contd....

Multimolecule Separation

Technique Analyte Matrix Modelling Approach Design Space Determination Reference

Reversed-

phase

HPLC–DAD

19 antimalarial drugsdivided in two groups

Arteplus pharmaceutical formulation

Full factorial designFactors: pH, 5 levels, gradient time, 3 levels, column temperature, 3 levels

Monte Carlo simulationsOne method for each groupNo robustness validation

(19)

Reversed-

phase

HPLC–DAD

18 non-steroidal anti-inflammatory drugs, 5 pharmaceutical conservatives, paracetamol, chlorzoxazone, caffeine, salicylic aciddivided into five groups

5 capsules from the Democratic Republic of Congo

Augmented central composite designFactors: pH, 7 levels, gradient slope, 5 levels, column temperature, 5 levels

Monte Carlo simulationsOne method for each groupRepeatability at the optimal conditions

(20)

Reversed-

phase

UHPLC–UV

15 antipsychotic drugs

Standards

Full factorial design Factors: column temperature and gradient time3 levels

Monte Carlo simulationsDetermination of DS through robustness criterion Cp calculation Experimental validation of robustness

(21)

Reversed-

phase

UHPLC–DAD

19 antibioticsdivided into 3 groups

Powder for injection

D-optimal experimentsFactors: pH, 6 levels, gradient time, 3 levels, column temperature, 3 levels

Monte Carlo simulationsRepeatability at the optimal conditions

(22)

Reversed-

phase

HPLC–UV

8 antidiabetic drugsGlimepiride plustablets

Full factorial design on 3 columnsFactors: pH, 3 levels, ternary solvent ratio, 3 levels, gradient time, 2 levels

DS through modelling error propagationIn-silico robustness testing

(23)

MEKC–DAD 7 triptans

AlmogranAuradolZomigtablets

Central composite designFactors: SDS concentration, n-butanol and ethanol percentages5 levels

Monte Carlo simulationsValidation of DS through a PB design Robustness validation of optimum through a PB design by expanding the number of factors

(24)

Enantioselective Separation

Technique Analyte Matrix Modelling Approach Design Space Determination Reference

CD–MEKC–

DAD

Ambrisentan + related substances

VolibrisCoated tablets

Central composite face centred designFactors: voltage, CD concentration, pH, 3 levels

Monte Carlo simulationsValidation of DS through a PB designRobustness validation of optimum through a PB design by expanding the number of factors

(25)

CD–CZE–DAD LevosulpirideLevobrenAmpoules

Doehlert design Factors: anionic CD concentration, 5 levels, neutral CD concentration, 7 levels, pH, 7 levels, voltage, 3 levels

Monte Carlo simulationsValidation of DS through a PB design Robustness validation of optimum through a PB design by expanding the number of factors

(26)

Natural Product Analysis

Technique Analyte Matrix Modelling Approach Design Space Determination Reference

SPE/reversed-

phase UHPLC–

UV/ELSD

Isoflavonoids, triterpenoid saponins

Pharmaceutical liquid herbal preparation of Shenqi Fuzheng

Box–Behnken designFactors: flow rate, column temperature, ELSD evaporation temperature, and ELSD gas flow rate3 levels

Monte Carlo simulationsRobustness validation of optimum during validation

(27)

Reversed-

phase

HPLC–UV

13 aporphine alkaloids

Leave extras of Spirospermum penduliflorum Thouars

Full factorial designFactors: pH, 3 levels, gradient slope, 3 levels, methanol proportion at the beginning of the gradient, 4 levels

Monte Carlo simulationsNo experimental validation of DS or robustness of optimum

(28)

Reversed-

phase

HPLC–DAD

7 tertiary alkaloidsLeave extras of Strychnos usambarensis

Full factorial designFactors: proportions of organic solvents in the initial phase, gradient slope, % acetonitrile, 3 levels

Monte Carlo simulationsNo experimental validation of DS or robustness of optimum

(29)

CD: cyclodextrin, DS: design space, ELSD: evaporative light scattering detector, PB: Plackett-Burman

Page 23: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

DOE and Response Modelling

DOEs are key components of a

QbD methodology. Compared to the

OFAT approach, DOEs minimize the

number of experiments, while gaining

knowledge about the analytical

method. Such multivariate strategies

allow the evaluation of the effects of

the factors and their interactions, the

modelling and the prediction of the

responses as a function of the factors,

and, in fine, the determination of the

DS. The factors are the CPPs and the

responses the CQAs.

Method optimization often begins

with a screening phase followed by

an optimization step using screening

and response-surface designs,

respectively.

Screening Designs: After a risk

prioritization, where a high number

of factors have been revealed

as potentially critical on method

performance, screening designs are

used to identify which parameters

are effectively critical. The aim is

the selection of CPPs. Such designs

allow the evaluation of a relatively

high number of factors (qualitative

or quantitative) — mostly at two

levels in a relatively low number of

experiments. The relation between

the responses and the factors is

described by a first-order polynomial

model. An estimation of the main

effects of factors on the considered

responses, mainly the CQAs, can

be assessed by calculating model

coefficients for the linear terms.

Factors with the most significant

affect on the analytical method are

retained as CPPs. Interaction effects

between factors are rarely evaluated,

while higher order terms, for example,

quadratic effects, cannot be studied.

Indeed, two-level screening designs

don’t allow modelling of curvature.

Such designs are therefore not

sufficient to investigate a process

involving interactions and higher-order

effect terms. The most often applied

screening designs are two-level

fractional factorial or two-level

Plackett-Burman designs (9,27,37).

Response-Surface Designs: Once

identified, CPPs are further studied

through a response-surface method to

predict CQAs. For a given number of

parameters, many more experiments

are required compared to first degree

screening designs. The analysis of

the results is focused on building

a mathematical model linking the

responses (CQAs) to the factors

(CPPs). More than two levels for each

factor are needed to fit quadratic

or higher-order-term polynomial

functions and, thus, to model the

curvature in the function responses.

Interactions between factors and

higher-order effect terms can then be

evaluated.

Full factorial designs

(7,8,11,12,19,21,23,28,29), central

composite designs (5,6,9,20,24,25),

Box-Behnken (13–16,18,27), and

Doehlert designs (17,26) are

commonly used. Figure 3 shows the

experimental domains resulting from

the study of three factors for central

composite designs, Box-Behnken

designs, and Doehlert designs.

D-optimal designs can also be

performed in particular cases, for

example, when experimental space

is constrained or standard factorial

designs require too many runs (22).

Such DOEs are essential tools

to define the DS of an analytical

method (33). Indeed, the model

enables response values (CQAs)

within the investigated experimental

domain to be predicted. Moreover,

model-parameter uncertainty and

the prediction uncertainty of the

model can be estimated, providing

information on the probability of

meeting the specifications imposed on

CQAs. The resulting DS corresponds

to a subspace of the experimental

domain where a level of quality is

ensured, that is, the CQAs meet the

specifications with a given probability.

Response Modelling: Responses

can be modelled in different ways.

The first approach uses optimization

software for an automated method

development. Such software is mostly

dedicated to reversed-phase HPLC

and based on the well-known linear

solvent-strength theoretical model.

Several software systems are available

commercially (7,8,11,12,38). However,

such theoretical models do not usually

take into account all the parameters

that may impact the analytical method.

Moreover, the variability or the method

quality (the robustness) is not included

in the method development.

A second approach is the use of

empirical models based on DOEs

and multiple linear regressions (MLR).

Such models link the responses to

the factors investigated by fitting an

appropriate polynomial model on the

data obtained from DOEs. Software

used for such empirical approaches

is that classically dedicated to

experimental designs. However, there

is software that is appropriate for QbD

developments by combining DOE,

MLR, and some specific CQAs to

evaluate method robustness (21,39).

Design Space: As mentioned

previously, the main aim in

applying QbD to analytical method

development is to establish a DS.

In the ICH Q8 (R2) guideline, DS is

defined as “the multidimensional

combination and interaction of input

variables (such as material attributes)

and process parameters that have

been demonstrated to provide

assurance of quality” (2).

Most of the time, DS is represented

graphically through response surface

overlay. A graph displaying multiple

CQA objectives is plotted using

software (Figure 4). This example

deals with the development of a

UHPLC method for the simultaneous

analysis of dextromethorphan (DXM)

and its main degradation products

(9). Contour plots of the seven studied

CQAs as a function of the four CPPs

are overlaid and DS is represented by

the white zone where all CQAs meet

predefined criteria.

Up to now, the DS of an analytical

method has been mainly defined

through mean-response surfaces.

However, mean-response surfaces

do not provide any guarantee about

method quality, and so it is impossible

to know with which probability the

CQAs will reach the performance

criteria. As “assurance of quality”

is clearly required by ICH Q8 (R2)

guideline (2), the level of assurance

that the analytical method will meet

the specifications must be known.

This is why a group of researchers

highlighted the need to consider

the prediction uncertainty by taking

into account the model-coefficient

uncertainty, that is, the model

uncertainty (40). The Monte Carlo

study is the most implemented

strategy (5,14–26). By including

model coefficient uncertainty, a

model’s predictive errors can be

calculated. This provides access

to the distributions of the modelled

responses for each operating

condition of the DOE. Consequently,

when establishing the DS, a level

23www.chromatographyonline.com

Gaudin and Ferey

Page 24: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

of quality assurance can be set by

imposing a probability threshold

above which the CQAs meet the

requirements. As an example,

Debrus et al. performed Monte Carlo

simulations to compute and model

a “robustness criteria”, called Cp.

As such criterion was a quantitative

measure of the variation of the

predicted responses, it was modelled

as an additional CQA to guarantee

the quality of the method (21). Quality

should be assured inside the DS and

as specified by ICH Q8 guideline

“working within the design space is

not considered as a change” (2), DS

should represent a zone of robustness

of the analytical method.

The final step of the QbD method

is to experimentally validate the DS.

To limit the number of experiments,

DOEs are usually required. Screening

designs, such as two-level fractional

factorial or two-level Plackett-Burman

designs, are sufficient to achieve

this validation. To validate the whole

DS, CPPs are usually varied from the

lowest to the highest limits of the DS

borders (6,9,14,16–18 24–26). DS

represents the robustness zone of the

method, and so a classical robustness

study verifying the influence of

small changes of the CPPs around

the working point on the CQAs

performance is not required. However,

after DS determination, it may be

preferred to perform a robustness

study before or juxtaposed to a

complete method validation (6,14,

16–18,24–26). Additional parameters

may be added, but the range of

variation of each parameter is often

reduced in comparison to the values

defined by the DS.

Another approach has been

proposed by Molnár et al. Using

chromatography modelling software,

a robustness testing is simulated over

more than 700 conditions by enlarging

the number of tested factors compared

to the number of CPPs retained for

the optimization step. Some selected

points are then experimentally

checked (7,8,11,12).

Some authors limit the DS validation

to a traditional robustness validation of

the optimum point, leading to methods

with a smaller robustness domain

because the method operable region

is also reduced (10,13,27). Finally,

in several papers DS validation and

robustness validation have been

replaced by a single repeatability

study at optimal conditions (20,22)

without verifying if the DS is correctly

defined. This latter approach cannot

provide a DS for the analytical method

under development, while robustness

inside is not experimentally checked.

This conflicts with the objectives of

QbD to develop a robust method.

Conclusion

A QbD approach for analytical method

development aims at determining a

multivariate domain, called the design

space (DS), where method critical

parameters have been demonstrated

to provide assurance of quality.

In fact, the analytical method is

no longer restricted to one single

combination of parameters, but is

now identified by a set of operating

conditions defining the DS. Inside

the DS, the method achieves the

predefined performance with a high

level of probability.

Many benefits are intrinsic to QbD

for the pharmaceutical industry.

Robust methods are developed so that

fewer method failures may

appear during routine use in QC

laboratories. Moreover, higher flexibility

is achieved because moving inside the

DS is not considered a change and

does not require any further regulatory

approval. QbD offers a deeper

understanding of the analytical method

through the evaluation of the main

and interaction effects of the method

parameters on its performance.

This additional knowledge gained

during development is crucial for

pharmaceutical analysts. Method

validation becomes less prone to

unpredictable errors, analyst reactivity

is improved to face future operating

problems, and strong scientific

justification may influence future

regulatory frameworks. Finally, method

transfer from one laboratory to another

is simpler because the adaptation of

the transferred method will be eased

by the knowledge of its DS at the

receiving site.

Throughout the product life cycle,

companies have the opportunity to

evaluate innovative approaches to

improve product quality. QbD, through

a more systematic approach, plays a

major role in assessing the potential of

new techniques and thus contributes

to continual improvement within the

pharmaceutical industry.

References(1) US Food and Drug Administration

(FDA), Department of Health and

Human Services, Pharmaceutical

cGMPs for the 21st Century —

A Risk-Based Approach, Final

report, September 2004 (http://

www.fda.gov/downloads/Drugs/

DevelopmentApprovalProcess/

Manufacturing/Questionsand Answers

onCurrentGoodManufacturing

PracticescGMPforDrugs/UCM176374.

pdf)

(2) International Conference on

Harmonization (ICH) of Technical

Requirements for Registration of

Pharmaceuticals for Human Use, Topic

Q8 (R2): Pharmaceutical Development

(ICH, Geneva, Switzerland, 2009).

(3) International Conference on

Harmonization (ICH) of Technical

Requirements for Registration of

Pharmaceuticals for Human Use, Topic

Q9: Quality Risk Management (ICH,

Geneva, Switzerland, 2005).

(4) International Conference on

Harmonization (ICH) of Technical

Requirements for Registration of

Pharmaceuticals for Human Use, Topic

Q10: Pharmaceutical Quality System

(ICH, Geneva, Switzerland, 2008).

(5) T. Tol, N. Kadam, N. Raotole,

A. Desaib, and G. Samanta, J.

Chromatogr. A 1432, 26–38 (2016).

(6) B. Hariram, R. Suresh Kumar, A.

Jaya Shree, D. Venugopala Rao,

L. Kalyanaraman, and K. Srinivas,

Chromatogr. 78, 1017–1029 (2015).

(7) A.H. Schmidt and I. Molnár, J. Pharm.

Biomed. Anal. 78–79, 65–74 (2013).

(8) A.H. Schmidt and M. Stanic, LCGC

North America 32, 126–148 (2014).

(9) C. Boussès, L. Ferey, E. Vedrines, and

K. Gaudin, J. Pharm. Biomed. Anal.

115, 114–122 (2015).

(10) R. Mallik, S. Raman, X. Liang,

A.W. Grobin, and D. Choudhury, J.

Chromatogr. A 1413, 22–32 (2015).

(11) R. Kormany, I. Molnár, J. Fekete, D.

Guillarme, and S. Fekete, Chromatogr.

77, 1119–1127 (2014).

(12) K. Monks, I. Molnár, H.J. Rieger, B.

Bogati, and E. Szabo, J. Chromatogr. A

1232, 218–230 (2012).

(13) Z. Ren, X. Zhang, H. Wang, and X. Jin,

RSC Adv. 6, 13050–13062 (2016).

(14) M. Jovanovic, T. Rakic, A. Tumpa, and

B. Jancic Stojanovic, J. Pharm. Biomed.

Anal. 110, 42–48 (2015).

(15) J. Terzic, I. Popovic, A. Stajic, A.

Tumpa, and B. Jancic Stojanovic, J.

Pharm. Biomed. Anal. 125, 385–393

(2016).

(16) S. Orlandini, B. Pasquini, C. Caprini,

M. Del Bubba, S. Pinzauti, and S.

Furlanetto, Electrophoresis 36,

2642–2649 (2015).

(17) S. Orlandini, B. Pasquini, R. Gotti,

A. Giuffrida, F. Paternostro, and S.

Furlanetto, Electrophoresis 35,

2538–2545 (2014).

(18) S. Furlanetto, S. Orlandini, B. Pasquini,

and C. Caprini, Anal Bioanal Chem 407,

7637–7646 (2015).

(19) B. Debrus, P. Lebrun, J.K. Mbinze, F.

Lecomte, A. Ceccato, G. Caliaro, J.

Mavar Tayey Mbay, B. Boulanger, R.D.

Marini, E. Rozet, and Ph. Hubert, J.

Chromatogr. A 1218, 5205–5215 (2011).

(20) J.K. Mbinze, P. Lebrun, B. Debrus, A.

Advances in Pharmaceutical Analysis October 201624

Gaudin and Ferey

Page 25: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Dispas, N. Kalenda, J. Mavar Tayey

Mbay, T. Schofield, B. Boulanger, E.

Rozet, Ph. Hubert, and R.D. Marini, J.

Chromatogr. A 1263, 113–124 (2012).

(21) B. Debrus, D. Guillarme, and S. Rudaz,

J. Pharm. Biomed. Anal. 84, 215–223

(2013).

(22) J.K. Mbinze, A. Dispas, P. Lebrun, J.

Mavar Tayey Mbay, V. Habyalimana,

N. Kalenda, E. Rozet, Ph. Hubert, and

R.D. Marini, J. Pharm. Biomed. Anal.

85, 83–92 (2013).

(23) H.I. Mokhtar, R.A. Abdel-Salam, and

G.M. Hadad, J. Chromatogr. A 1399,

32–44 (2015).

(24) B. Pasquini, S. Orlandini, M. Del

Bubba, E. Bertol, and S. Furlanetto,

Electrophoresis 36, 2650–2657 (2015).

(25) S. Orlandini, B. Pasquini, C. Caprini,

M. Del Bubba, M. Dousa, S. Pinzauti,

and S. Furlanetto, J. Chromatogr. A

doi:10.1016/j.chroma.2016.06.082

(2016).

(26) S. Orlandini, B. Pasquini, M. Del

Bubba, S. Pinzauti, and S. Furlanetto,

J. Chromatogr. A 1380, 177–185 (2015).

(27) L. Wang and H. Qu, Anal. Bioanal.

Chem. 408, 2133–2145 (2016).

(28) M.H. Rafamantanana, B. Debrus, G.E.

Raoelison, E. Rozet, P. Lebrun, S.

Uverg-Ratsimamanga, P. Hubert, and

J. Quetin-Leclercq, J. Pharm. Biomed.

Anal. 62, 23–32 (2012).

(29) I. Nistor, M. Cao, B. Debrus, P. Lebrun,

F. Lecomte, E. Rozet, L. Angenot,

M. Frederich, R. Oprean, and P.

Hubert, J. Pharm. Biomed. Anal. 56,

30–37 (2011).

(30) S. Orlandini, S. Pinzauti, and S.

Furlanetto, Anal. Bioanal. Chem. 405,

443–450 (2013).

(31) K. Ishikawa, What is Total Quality

Control? The Japanese Way (Prentice-

Hall, Englewood Cliffs, NJ, USA, 1985).

(32) D. Stamatis, Failure Modes and Effects

Analysis, Second Edition (ASQ Quality

Press, Milwaukee, WI, USA, FMEA from

Theory to Execution, 2003).

(33) E. Rozet, P. Lebrun, B. Debrus, B.

Boulanger, and P. Hubert, Trends Anal.

Chem. 42, 157–167 (2013).

(34) International Conference on

Harmonization (ICH) of Technical

Requirements for registration of

Pharmaceuticals for Human Use,

Topic Q2 (R1): Validation of Analytical

Procedures: Text and Methodology

(ICH, Geneva, Switzerland, 2005).

(35) C. Hubert, S. Houari, E. Rozet, P.

Lebrun, and Ph Hubert, J. Chromatogr.

A 1395, 88–98 (2015).

(36) F.G. Vogt and A.S. Kord, J. Pharm. Sci.

100, 797–812 (2001).

(37) D. Awotwe-Otoo, C. Agarabi, P.J.

Faustino, Muhammad J. Habib, S. Lee,

M.A. Khan, and R.B. Shah, J. Pharm.

Biomed. Anal. 62, 61–67 (2012).

(38) C. Horváth, W. Melander, and I. Molnár,

J. Chromatogr. 125, 129–156 (1976).

(39) R. Cela, E.Y. Ordonez, J.B. Quintana,

and R. Rodil, J. Chromatogr. A 1287,

2–22 (2013).

(40) P. Lebrun, B. Govaerts, B. Debrus, A.

Ceccato, G. Caliaro, P. Hubert, and B.

Boulanger, Chemom. Intell. Lab. Syst.

91, 4–16 (2008).

Karen Gaudin completed a Ph.D.

in analytical chemistry in 1999 at

the School of Pharmacy of Paris XI

University in France. She is a professor

and director of a research team focusing

on analytical and pharmaceutical

developments at the University of

Bordeaux. These main developments

concern strategies to analyze APIs or

lipids in new pharmaceutical products

using various separation techniques

(reversed phase, HILIC) and detectors

(CAD) eventually in green chemistry and

QbD. She has published more than 50

papers in reputed journals and serves

as an editorial board member of repute.

Ludivine Ferey has completed a

Ph.D. in analytical chemistry at the

Industrial Physics and Chemistry

Higher Educational Institution (ESPCI)

in Paris, France. She is an associate

professor in the ChemBioPharm

team (ARNA, INSERM U1212) at the

University of Bordeaux in France. Her

research is focused on the use of

chemometric tools, mostly QbD, for

the development and optimization of

separation analytical methods in the

field of pharmaceutical analysis.

25www.chromatographyonline.com

Gaudin and Ferey

>ƵĚŐĞƌ�Ɛ�ĞdžƉĞƌƟƐĞ�ĂŶĚ�Yď��ĂƉƉƌŽĂĐŚ�ƚŽ�ĚƌƵŐ�ŐůLJĐŽƐLJůĂƟŽŶ�ĂŶĂůLJƐŝƐ�ĐĂŶ�ŚĞůƉ�LJŽƵ�ĂĐŚŝĞǀĞ�ƐƵĐĐĞƐƐ���

/ƚ�ŝƐ�ƵƐĞĚ�ďLJ�ŽǀĞƌ�ŚĂůĨ�ŽĨ�ƚŚĞ�ǁŽƌůĚ�Ɛ�ƚŽƉ�Ϯϱ�ƉŚĂƌŵĂ�ĐŽŵƉĂŶŝĞƐΎ

www.ludger.com

Are you seeking regulatory approval for your glycosylated biopharmaceutical?

ΎĂƐ�ũƵĚŐĞĚ�ďLJ�'ůŽďĂů�ĂƚĂ�ĨƌŽŵ�ϮϬϭϰ�ŐůŽďĂů�ƐĂůĞƐ

Kits & Reagents ZĞůŝĂďůĞ�ƚĞĐŚŶŽůŽŐLJ�ĨŽƌ�ŐůLJĐĂŶ�ƐĂŵƉůĞ�

ƉƌĞƉĂƌĂƟŽŶ�ŝŶĐůƵĚŝŶŐ�ƐƚĂŶĚĂƌĚƐ#�ĐůĞĂŶ$ƵƉ�

ĂŶĚ�ůĂďĞůŝŶŐ�ŬŝƚƐ

High-Throughput Glycomics�ŽůůĂďŽƌĂƟǀĞ�ZΘ��ƉƌŽŐƌĂŵŵĞƐ�ƚŽ�ƐƚƵĚLJ�

ŐůLJĐŽƐLJůĂƟŽŶ�ŝŶ�ĚŝƐĞĂƐĞ�ĂŶĚ�ĨŽƌ�

ďŝŽŵĂƌŬĞƌ�ĚŝƐĐŽǀĞƌLJ

*O\FRSURÀOLQJ�6HUYLFHV �ĞƚĂŝůĞĚ�ĂŶĂůLJƐŝƐ�ŽĨ�ďŽƚŚ�ŝŶŶŽǀĂƚŽƌ�ĚƌƵŐƐ�ĂŶĚ�

ďŝŽƐŝŵŝůĂƌƐ�ďLJ�h,W>�#�D^#�ĂŶĚ�>�$D^

Ludger Ltd, Oxfordshire UK

Dr. Louise Royle+HDG�RI�*O\FRSURÀ[email protected]

'U��&ODLUH�0RUJDQ+HDG�RI�%XVLQHVV�'HYHORSPHQW

FODLUH�PRUJDQ#OXGJHU�FRP

Page 26: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Liquid chromatography coupled to

tandem mass spectroscopy (LC–MS/

MS) using triple quadrupole mass

spectrometers (TQ-MS) has evolved

into a mainstream approach for

quantitative bioanalysis of drugs and

metabolites over the past 20 years.

The high selectivity of triple quadrupole

instruments in the selected reaction

monitoring (SRM) mode makes the

technique very suitable for targeted

analysis in a high-throughput mode with

short run times. The disadvantage of this

approach is that only information on the

analytes for which SRM mass transitions

are put into the method are obtained.

Qualitative information on other analytes

will not be obtained using this approach.

The sensitivity and dynamic range

of the latest generation of HRMS

instruments now approaches that

of TQ-MS, but at a higher and still

increasing resolution. When HRMS

is used, no SRM transitions for the

analytes of interest need to be entered

into the method. The system is run

in full-scan MS mode and selectivity

is achieved by the high resolution of

the system, instead of by selecting

a specific precursor–product ion

combination in SRM. As a result, a much

richer dataset is obtained, containing

not only quantitative information on the

analytes of interest, but also qualitative

information on the other analytes present

in the sample.

An example of the different data types

obtained by HRMS and TQ-MS is shown

in Figure 1. For TQ-MS, only information

on the parent drug is obtained, whereas

with HRMS, information on adducts,

in-source fragments, metabolites, and

endogenous compounds, including

biomarkers and background ions, is

obtained.

It can be very useful to obtain both

quantitative information on the drug and

qualitative information on other analytes

that are present in the sample, and not

be limited by only the analytes for which

SRM mass transitions were entered. In

the literature, this approach is referred

to as a quan–qual approach, and the

terminology quan–qual is mainly used

in the context of obtaining quantitative

information on the drug and qualitative

information on drug metabolites (1–3).

For example, in metabolic stability

studies, quantitative data on the drug,

as well as qualitative data of the in vitro

metabolites, can be relevant (4). In this

way, not only the metabolic stability

of the drug is assessed, but also

information is obtained on the metabolic

hotspot(s) of the molecule, which is

relevant for compound optimization to

improve metabolic stability.

Our group, however, prefers to use

a broader definition for quan–qual,

because there is much more potential in

the use of the qualitative data besides

looking for drug metabolites. Looking

at the presence and regulation of

endogenous metabolites can be of

interest, although the number and type

of biomarkers that can be followed will

be limited by the more generic sample

preparation and LC conditions applied

for quan–qual analyses. The availability

of all ion information can be beneficial

for speeding up method development,

or for the resolution of analytical or

other issues. A few examples of this are

given below. Certain compounds, like in

formulations (for example, PEG 400 and

polysorbate [tween]) or endogenous

compounds, such as phospholipids,

can cause ion suppression and

influence the quantitative result if

they are coeluted with the analyte.

Therefore, it is useful to know what is

coeluted with the analyte(s) of interest,

so this information can be used for

potential troubleshooting. If an in vitro

experiment does not seem to provide

the expected result, it might be useful to

check whether the right cofactors were

present and in which form (for example,

to determine the ratio of glutathione

[GSH] and oxidized GSH in a reactive

metabolite screening), or to check if

cells were present during the in vitro

experiments by looking for the presence

of phospholipids via their marker ions at

m/z 104 and 184.

Quan–qual approaches have clear

advantages compared to doing “quan”

alone but there are some caveats to

take into account. First, for quantitative

targeted analysis, very short analytical

run times (typically 0.5–1.5 min) are

preferred to enable the analysis of

many samples in a short timeframe. For

qualitative analysis, significantly longer

analytical runtimes are more beneficial.

For drug metabolite identification, longer

analytical runtimes (typically 10–20 min)

High-Throughput Analysis of Drugs and Metabolites in Biological Fluids Using Quan–Qual Approaches

Ronald de Vries, Rob J. Vreeken, and Filip Cuyckens, Pharmacokinetics, Dynamics & Metabolism, Janssen R&D,

Beerse, Belgium

The new generation of high-resolution mass spectrometry (HRMS) systems offers high sensitivity,

dynamic range, resolution, accuracy, and scan-to-scan reproducibility, enabling high-throughput

quantitative analyses in combination with information-rich qualitative data. The most recently released

HRMS systems offer an alternative to triple quadrupole (TQ)-MS systems. This provides a huge opportunity

to obtain quantitative and qualitative information from one analysis, but also requires a different mindset

and expertise to make the right choices and compromises to get the most information from your sample.

Ph

oto

Cre

dit: d

ow

ell/

Ge

tty Im

ag

es

Advances in Pharmaceutical Analysis October 201626

Page 27: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

are important to reduce the risk of

coelution of isobaric metabolites and, in

this way, to correctly identify metabolites.

Therefore, we see quan–qual more

as a quantitative approach providing

additional qualitative information “for

free”, rather than using the approach as

a replacement for separate metabolite

identification studies.

When metabolite identification is (one

of) the main goal(s) of the study, it could

be better to reanalyze a small selection

of the samples using a longer analytical

runtime and use these data for metabolite

identification, rather than analyzing

all samples with a longer analytical

runtime (5). Also, if obtaining biomarker

information is one of the main goals, it

may be better to rerun samples using a

method where sample preparation and

chromatography have been optimized for

the analysis of the required biomarkers.

Furthermore, the large amount of

data produced by quan–qual analysis

increases the risk of confusion. It is

important to ensure that the qualitative

data produced are used with a focus

on answering specific key questions,

rather than spending time trying to

extract all available information on

a routine basis simply because it

is available. The availability of the

qualitative data should be used to avoid

repetition of experiments and analyses

if specific questions come up later in

the development programme. Data

can be reevaluated at the moment that

the specific questions arise, rather

than directly after the data has been

acquired. For example, if a critical in

vivo experiment is performed, where

only quantitative data on the drug

are requested, it might be useful to

perform the analysis in quan–qual mode

but initially only use the quantitative

data. If, later on, questions arise (for

example, the potential involvement of a

metabolite explaining a pharmacokinetic

or pharmacodynamic [PK or PD]

disconnect, or the presence of a human

metabolite in animal species), it is easy

to go back to the qualitative data months

or years after analyses to find the answer.

In this paper, sample preparation

methods and mass spectrometry

approaches suitable for high throughput

quan–qual analysis are presented. High

performance liquid chromatography

(HPLC) optimization for high throughput

quan–qual analysis is also discussed.

Furthermore, potential uses, challenges,

and future perspectives for quan–qual

analysis will be discussed.

Sample Preparation

Since the qualitative part of a quan–

qual analysis is an untargeted analysis

with a potential interest in molecules

27www.chromatographyonline.com

Cuyckens et al.

20

0

40

60

80

100

Re

lati

ve

Ab

un

da

nce

20

0

40

60

80

100

Re

lati

ve

Ab

un

da

nce

0.0 0.5 1.0 1.5 2.0 2.5

Time (min)

0.0 0.5 1.0 1.5 2.0 2.5

Time (min)

(a)

(b)

metabolite

parent drug

adduct

in source fragment background ion

biomarker

Figure 1: (a) Triple quad MS (TQ-MS) data. (b) High-resolution MS (HRMS) data.

0

0.9 1.1 1.3 1.5

Time (min)

1

1

3

3

3

3

4

2

2

22

2

5

5

4

4

4

24

4

4

22

25

50

75

100

%

0

25

50

75

100

%

(a)

(b)

2.7 2.9 3.1 3.3 3.5 3.7

Time (min)

Figure 2: Separation of buspirone (1) and its major metabolites (2: +O; 3: -2H; 4: +O-2H; 5: +O+gluc) in rat hepatocytes with: (a) a traditional metabolite profiling chromatographic method using a 100-mm length porous 1.7-μm particle packed column at 0.4 mL/min and (b) a shorter 50-mm solid core 1.6-μm particle packed column at a higher 0.9 mL/min flow rate. Adapted with permission from reference 6.

Page 28: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

that are unidentified at the start of

the analysis, the sample preparation

should be kept to a minimum to avoid

any potential degradation or recovery

issues of the compounds of interest.

This is in line with what is done in

metabolite profiling studies and might

be in contrast with quantitative assays

that often use liquid–liquid extraction

(LLE) and solid-phase extraction (SPE)

to improve the specificity and sensitivity

of the assay. For quan–qual analyses,

typically one or multiple volumes of

an organic solvent (usually 3 or more

volumes of acetonitrile) are added to the

sample to stop any remaining enzymatic

activity, break the analyte–protein

binding, and precipitate the majority

of the proteins. The samples are then

centrifuged and the supernatant

injected onto the LC–MS system.

Liquid Chromatography

Optimization

Metabolites are often structurally

related molecules. Therefore, longer

LC gradients are typically used in

metabolite profiling and metabolomics

studies to reduce the risk of missing

coeluting isobaric compounds.

Quantitative LC–MS runs are usually

very short, allowing a higher throughput

of the analysis of large(r) batches of

samples. A compromise needs to be

made providing sufficient throughput

with decent separation power. This

is also a function of the batch size

because there is no use in running

short gradients if the instrument sits

idle for a couple of hours because the

run finishes in the middle of the night.

Nevertheless, longer runs also result in

larger file sizes and processing times,

and a higher potential for MS drift.

Longer LC gradients do not

necessarily give rise to the separation

of more compounds in one run —

expressed as peak capacity for

gradient elution. The effect of a longer

run time on separation power is often

overestimated because an increase

in gradient time gives a less than

proportional increase in peak capacity.

When chromatographic conditions are

optimized using shorter LC columns

packed with sub-2 μm particles in

combination with higher flow rates

and the extracolumn dead volume

is reduced, it is perfectly feasible to

achieve peak capacities similar to

those obtained in traditional metabolite

profiling studies in 2–3-fold shorter

run times. This was demonstrated

on a mix of pharmaceutical

standards and an in vitro incubation

of buspirone containing multiple

isobaric metabolites (6). The latter is

illustrated in Figure 2, which shows a

slightly better separation between the

major metabolites of buspirone in rat

hepatocytes using a 2.5-fold shorter

generic method, which applies a

short (5 cm) ultrahigh-pressure liquid

chromatography (UHPLC) column

packed with solid core 1.6-μm particles

in combination with a higher flow rate.

While the peak capacity of the LC

separation is worthwhile optimizing

because metabolites are often isobaric

compounds, the selectivity provided by

mass spectrometry — high-resolution

MS in particular — has a much bigger

impact on the total peak capacity of the

overall analytical assay.

Mass Spectrometry Methods

To take full benefit of the optimized

chromatographic systems boosted for

peak capacity, the mass spectrometry

methods should provide high-quality

quantification and a maximum of

qualitative information in accordance

with the short timeframe of high

resolution chromatographic peaks.

To obtain high-quality quantitative

information with good reproducibility,

linearity, and sensitivity, an adequate

description of the chromatographic

peak is essential. This requires typically

>12 data points per chromatographic

peak. Current attainable peak widths

when using UHPLC are around 2–3 s

and are distinctively smaller than

traditional LC peak widths. Current

HRMS systems are well equipped

with respect to scan speed to obtain

at least 12 data points across the

peak in full scan mode, even with

these narrow peak widths. From full

scan HRMS data, relevant qualitative

information can be obtained. Analytes

can already be identified to a certain

extent because information on the

likely elemental composition can be

obtained based on the accurate mass

measurement and isotope ratios, or

by using very high-resolution systems

(Fourier-transform MS) one can even

look at the isotopic fine structure. Scan

routines to obtain MS/MS spectra can

be added to the MS method to obtain

additional qualitative information to aid

in structure identification, for example,

when coeluting isobaric metabolites

show differences in fragmentation.

However, when additional scan modes

are added to the mass spectroscopy

method, in addition to the full scan

mode, challenges can arise with

regards to the available acquisition

time for these experiments. Optimizing

both the “quan” and “qual” world will

result in a “sweet spot” of settings

where, on the one hand, an acceptable

number of data points allows reliable

quantitation and, on the other, MS/MS

spectra of sufficient quality further aids

identification.

The MS/MS scan routines can be

divided into targeted (data-dependent

[7,8]) and untargeted (data-independent

[8,9]) strategies (see Figure 3). Targeted

approaches relate to acquisition of

product ion scans of preselected

m/z values. A targeted approach

can be (but does not need to be)

part of a data-dependent strategy.

A data-independent approach is by

definition untargeted.

Data-dependent strategies, such

as Data Directed Analysis (DDA) or

Information Dependent Analysis (IDA),

Advances in Pharmaceutical Analysis October 201628

Cuyckens et al.

Ion source

DDA/IDA

SWATH

MSe/MSall

Targeted

Un-Targeted

Collision cell

TOF

TOF

TOF

Q1

Figure 3: An overiew of different quan–qual MS acquisition strategies. Information or Data Dependent Acquisition (IDA or DDA), Sequential Window Acquisition of all THeoretical fragment ion spectra (SWATH), MS/MS of all ions (MSall or MSe). Adapted with permission from reference 8.

Page 29: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

have been known for more than a

decade and were first used on triple

quadrupole instruments. The system

acquires data in full-scan mode and

upon passing a threshold (either

intensity [untargeted] or intensity at

predefined m/z values [targeted]), the

system automatically switches to one

or more product-ion scans of that m/z

value to obtain product ion spectra

that can be used for identification

purposes. Instead of using a predefined

list of m/z values (targeted approach),

MS/MS spectra can also be acquired

through different parameters, such as

the 10 most intense ions. This can be

combined with dynamic inclusion and

exclusion lists, isotope pattern filtering

(if the analytes of interest contain

chlorine, the isotope pattern filtering

can be used to obtain MS/MS spectra

of only chlorine-containing analytes),

dynamic background subtraction,

and on-the-fly mass defect filtering to

maximize the number of MS/MS spectra

obtained for relevant analytes, while

reducing the acquisition of MS/MS

spectra of irrelevant background ions.

(Mass-defect filtering is often used in

drug metabolism and filters data on the

basis of the mass-defect of the parent

compound. Unrelated compounds are

then dynamically excluded from MS/

MS acquisition.) The use of HRMS

also allows for “thresholding” based

on accurate mass instead of nominal

mass, further reducing the number

of product ion spectra of irrelevant

background ions. Despite all these

available filtering algorithms, the

disadvantages of data-dependent

strategies remain. Taking individual

MS/MS spectra requires substantial

scanning time and hence some

relevant, often lower abundant, analytes

might be missed. Another disadvantage

of a data-dependent strategy is that

it relies heavily on prior knowledge

of the sample and that it cannot be

used as a generic method, especially

for parameters such as mass defect

filtering, isotope filtering, and using a

predefined list of m/z values for each

drug. Although MS/MS spectral quality

is often superior to that obtained in

data-independent strategies (discussed

below), it is not usually the method of

choice for quan–qual analyses, which

ideally comprise the maximum amount

of information to allow questions that

come up long after the analyses have

finished to be addressed.

In data-independent strategies, no

precursor ions are selected for MS/MS

as it is in data-dependent or targeted

approaches. Instead, MS allows the

passage of all precursor ions at the

same time and no selection of ions on

whatever criteria needs to be made.

Both full scan and MS/MS spectra

of all incoming ions are obtained via

this so-called MSall (also called MSE)

approach by alternately acquiring

spectra at low-collision energy (full

MS) and high-collision energy (MS/

MS). If the UHPLC separation of the

different analytes in the sample is

adequate, good quality MS/MS spectra

can be obtained via this approach.

However, when the complexity of the

sample increases, resulting in coelution

of analytes, mixed MS/MS spectra

are obtained that complicate their

interpretation. One way to obtain cleaner

MS/MS spectra in MSall mode, even for

low-abundant analytes coeluting with

high-abundance interfering analytes,

is to transmit smaller m/z ranges (for

example, 25 amu) instead of the whole

m/z range to pass through the first MS

analyzer and subsequently produce

MS/MS spectra for each separate

window (multiplexed MS/MS, MSX,

SWATH). By consecutively stepping up

the m/z range of ions passing through

the MS system, the complete mass

range of interest is “scanned” over.

Data deconvolution by the vendor

software relates the precursor and

associated product ions. Parameters

like scan speed, width of each window,

number of windows selected, scan

speed or dwell time for each window,

and overlap between windows will

affect the information obtained for

each compound. The disadvantage of

multiplexed MS/MS is that the required

scan time is significantly longer than for

MSall, and inversely proportional to the

width of the m/z windows. Therefore,

the multiplexed MS/MS approach is

usually not compatible with quan–qual

analysis where full MS or MSall is to be

preferred, unless future developments

in HRMS technology result in large

improvements in scan speed. Another

technique resulting in cleaner MS/MS

spectra in MSall mode without affecting

scan times is ion mobility spectrometry

(IMS). In IMS, ions are separated based

on size, shape, and charge rather than

on m/z. The combination of retention

time (LC) and drift time (IMS) alignment

results in cleaner MS/MS spectra

because precursor and product ions

have identical drift times, while coeluting

background ions and their product ions

might be separated in the ion mobility

device preceding the collision cell

(10). In full MS, ion mobility separation

can be beneficial, providing advanced

selectivity and, thus, better detection

limits. The IMS separation also has some

disadvantages: it affects the dynamic

range of the detector (earlier saturation),

the resolving power of current IMS

systems is often not adequate enough,

and it has an impact on scan speed;

however, in general, it is still compatible

with average UHPLC peak widths.

Challenges and Future

Perspectives

There are a lot of advantages to using

HRMS systems for the acquisition

of both quantitative and qualitative

information in the same analyses.

The number of applications is also

gradually growing in different (mainly

non-regulated) fields. Nevertheless, the

majority of LC–MS quantification is still

triple quadrupole-based since quan–

qual approaches and high-resolution

quantification, in general, also come

with some challenges, as described in

previous publications (11,12).

Historically, most laboratories

focusing on LC–MS quantification are

equipped with triple quadrupoles.

The replacement of an MS system

requires substantial investment, which

can hamper a rapid shift to HRMS.

In the last few years HRMS systems

have become available offering

high sensitivity, dynamic range,

resolution, accuracy, and scan-to-scan

reproducibility at a price similar to

modern triple quadrupole systems.

Most of these are still tandem mass

spectrometers (mainly quadrupole

time-of-flight [QTOF]). However, the

selectivity provided by the narrow

extraction windows, based on accurate

mass in full MS, is sufficient. Therefore,

a growing choice in single-stage

high-resolution systems will make the

switch easier because the investment

will be lower than a high-end triple

quadrupole or a tandem HRMS

system. In addition, the ease of use

will be improved over a tandem HRMS

and TQ-MS system. HRMS systems

were historically designed to be used

primarily by more experienced MS

users. The newly released TOF systems

are focused on quantification and

29www.chromatographyonline.com

Cuyckens et al.

Page 30: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

ease of use, rather than on flexibility in

scan speed, resolution, and a myriad

in MSn or MSall scanning options. As

is the case in any application, any

good hardware is only useful when

appropriate application software

is available. Many good uni- and

multivariate analysis approaches

and structure identification tools are

available for metabolite profiling,

typically done on HRMS systems.

There is still room for improvement

because identification of molecules

based on mass spectrometry can be

time-consuming and requires expertise.

Another bottleneck in the qualitative

part of the quan–qual analysis is

that appropriate blank samples (for

example, from vehicle dosed animals)

are often lacking. This gives rise to false

positives upon extraction of metabolites

from the background.

Quantification tools based on narrow

extraction windows are also available

in most, if not all, vendor software.

Processing times might be longer

compared to triple quadrupole data

processing because of the exponentially

larger file sizes linked to the richness

of the data, which also require much

larger servers for file storage. However,

a lot more can potentially be done with

the available MS data besides looking

at one accurate mass extracted ion

chromatogram of the analyte(s) of

interest, as is currently the standard for

quantitative processing. Besides the

[M + H]+ ion chromatogram, the ion

chromatograms of the isotopes could be

automatically processed to check their

correspondence with the theoretical

isotope distribution. This could serve as

a peak purity check to highlight potential

coeluting isobaric species or other

coeluting compounds. Isotope peaks

could be processed instead of the

primary isotope in case of interference

or summed to increase signal-to-noise

ratios, especially in cases where the

isotope peaks are equally abundant

(bromine-containing compounds,

multiple charged ions). Isotopes could

also be used in detector saturation

(and no saturation of the ionization)

to increase the dynamic range by

processing a less abundant isotope

peak. The richness of the accurate mass

data provides many more opportunities,

such as automatically highlighting the

presence of adducts and in-source

fragments, automated assignment of

the optimal narrow extraction window,

and alerting the user for coelution with

potentially interfering matrix compounds

(phospholipids, PEG from the

formulation) to name a few. These extra

capabilities and improved ease-of-use

of HRMS systems are key to convincing

the majority of the LC–MS quantification

user community to make the switch

to high-resolution MS quantification

and quan–qual analysis in those

cases where additional information

on metabolites or background ions

might be useful in the future, and

where there is no need for that extra

2–5-fold more sensitivity provided by

the newest high-end triple quadrupole

systems. Furthermore, increasing scan

speed and the resolution of both the

mass spectrometer and ion mobility

separation in newer HRMS instruments

will inevitably lead to more adaptation

to quan–qual workflows. The pace

with which the capabilities of modern

high-resolution systems increase and

the computer power and software

tools improve will dictate the speed

of implementation. However, the split

between “quantitative” and “qualitative”

bioanalytical departments, lack of

experience with high-resolution MS, and

often a more conservative approach

in regulatory environments are some

of the other hurdles to overcome for a

widespread application.

References(1) K.P. Bateman, M. Kellmann, H. Muenster,

R. Papp, and L. Taylor, J. Am Soc Mass

Spectrom. 20, 1441 (2009).

(2) W. Korfmacher, Bioanalysis 3(11), 1169

(2011).

(3) L. King, Bioanalysis 6(24), 3337 (2014).

(4) G. Backfish, B. Reder-Hilz, J.

Hoeckels-Messemer, et al., Bioanalysis

7(6), 671 (2015).

(5) M.F. Grubb, W.G.Humphreys, and

J.L. Josephs, Bioanalysis 4(14), 1747

(2012).

(6) A.-C. Dubbelman, F. Cuyckens, L. Dillen,

G. Gross, T. Hankemeier, and R.J.

Vreeken, J. Chromatogr. A 1374, 122–133

(2014).

(7) M.R. Anari, R.I. Sanchez, R. Bakhtiar, R.B.

Franklin, and T.A. Baillie, Anal. Chem.

76(3), 823 (2004).

(8) X. Zhu, Y. Chen, and R. Subramanian,

Anal. Chem. 86(2), 1202–1209 (2014).

(9) K.P. Bateman, J. Castro-Perez, M. Wrona

et al., Rapid Commun. Mass Spectrom.

21(9), 1485 (2007).

(10) S. Blech and R. Laux, Int. J. Ion Mobil.

Spec. 16(5), 5 (2013).

(11) E.N. Fung, M. Jemal, and A.-F. Aubrym,

Bioanalysis 5(10), 1277 (2013).

(12) L. Dillen and F. Cuyckens, Bioanalysis

5(10), 1145 (2013).

Ronald de Vries graduated in

organic and analytical chemistry at

the Free University of Amsterdam, in

the Netherlands. After working in a

contract laboratory for 7 years, he joined

Janssen R&D in 1998. At Janssen

R&D, he worked in the bioanalytical

department supporting both clinical and

non-clinical bioanalysis. He provided

the bioanalytical support for various

drug development programmes and

led the method establishment group

responsible for development of new

bioanalytical assays using LC–MS/

MS. Since 2014, he has worked in the

drug metabolism group, focusing on

metabolite identification using high

resolution mass spectrometry and

radiodetection. He has (co-)authored

more than 40 peer-reviewed scientific

publications.

Rob J. Vreeken has over 25 years

experience in quantitative and qualitative

analysis by hyphenated MS techniques

in a wide variety of applications. He has

worked in academia, service providers,

and in industry, and is currently

employed at Janssen Pharma R&D. His

team focuses on quantitative assays

in early discovery and exploratory

pharmacokinetics, dynamics, and

metabolism. Next to semi-HT compound

exposure analysis, the team implements

metabolomics strategies to collect

information on efficacy through

semi-quantitative PD-marker analysis.

He is also an associate professor at M4I,

Maastricht University in the Netherlands,

where he focuses on quantitative mass

spectrometry imaging techniques for

pharmaceutical markers. He has (co-)

authored close to 100 peer-reviewed

scientific papers and is a frequent

presenter at international symposia.

Filip Cuyckens is a Scientific Director

& Fellow at Janssen R&D. He is

responsible for analytical sciences

in the pharmacokinetics, dynamics,

and metabolism department,

focusing on metabolite profiling and

identification of drugs in discovery

and development, and quantification

of drug candidates, metabolites, and

biomarkers in biological matrices. He

earned a pharmacist degree in 1998,

a degree in industrial pharmacy in

2002, and a Ph.D. in pharmaceutical

sciences in 2003. He has (co-)authored

more than 50 peer-reviewed scientific

publications, and is a member of the

associate editorial board of Rapid

Communications in Mass Spectrometry

and board member of the Belgian

Society for Mass Spectrometry.

Advances in Pharmaceutical Analysis October 201630

Cuyckens et al.

Page 31: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Analysis of enantiomers is a critical topic

in the life science, food, fragrance, and

environment industries, and is also key

in asymmetric synthesis and catalysis.

Enantioselective chromatography

has evolved as a powerful tool for the

analysis of chiral compounds and is

now established as the method of

choice in almost all academic and

industrial laboratories dealing with

chiral molecules. Over the past 35

years intense efforts have been made

to obtain reliable tools for this purpose

and various methods and techniques

have been developed and optimized for

the accurate determination of the chiral

composition of enantiomeric mixtures

(Table 1).

The techniques in Table 1 do not all

have the same importance but all have

been applied. While the formation of

diastereoisomers prior to analysis was

the common practice in the past, today

the respective enantiomers of almost

any type of small chiral molecules can

be directly “discriminated”. Among all

techniques, liquid chromatography (LC)

and supercritical fluid chromatography

(SFC) are now the most used methods

for enantioselective analysis in the

pharmaceutical environment. Gas

chromatography (GC) is still used,

but more for volatile substances and

amino acids. There are numerous

applications of capillary electrophoresis

(CE), including drugs, but most of these

applications have been performed in

academic environments. It is used less

in the pharmaceutical industry, but might

still be useful and even the best method

for particular applications. Spectroscopic

methods, such as nuclear magnetic

resonance (NMR) spectroscopy,

vibrational circular dichroism (VCD)

spectroscopy, and mass spectrometry

(MS), are also occasionally used, but

not to the extent of the chromatographic

methods.

As many chromatographic techniques

are now available for the analysis of chiral

molecules, the choice is usually driven

by the application, that is, for volatile

compounds, GC might be preferred,

while for preparative purposes, LC or

SFC will be favoured.

The types of application covered

by the enantioselective analytical

techniques are quite broad and the

major ones are listed in Table 1. This

article focuses on the contemporary

enantioselective analytical techniques

with an emphasis on pharmaceutical

chiral compounds and, therefore, only

the most contemporary approaches are

discussed.

Analysis of Chiral Molecules

by HPLC

Over the past 30 years, HPLC on

chiral stationary phases has evolved

as a powerful method to separate

enantiomers. HPLC is applicable to

enantiomers of almost all types of small

chiral molecules, it is compatible with

almost all kinds of functional groups

(basic, acidic, neutral), and it can usually

be applied directly without previous chiral

derivatization to diastereoisomers. In the

classical pharmaceutical environment, it

concerns about 50% of the molecules,

the others being achiral or more complex

molecules, such as small and large

peptides, monoclonal antibodies, and

antibody–drug conjugates.

Hundreds of chiral stationary phases

(CSPs) have been commercialized for

HPLC, but the practical experience of

the last 25 years has led to a ‘“natural”

selection of the phases, and it is

generally recognized that a relatively

limited number of columns cover at

least 90% of all applications (Figure 1).

These “all-rounder” phases consist of

polysaccharide derivatives, particularly

from cellulose and amylose. It has

already been shown more than 40 years

ago that cellulose triacetate (CTA) has

unique chiral recognition properties

in enantioselective chromatography

(1), and the very first enantioselective

pharmaceutical application on this

polysaccharide phase was reported

more than 30 years ago (2). However,

it is the process elaborated by Yoshio

Okamoto and his group in Japan that

has made possible the preparation

of robust and high-performing

polysaccharide-based CSPs, which have

been commercialized and are suitable

for validated analytical processes (3).

Other column manufacturers have

produced generic versions of these

polysaccharide-based phases. Most

of the reported applications were

performed in the normal-phase mode,

but the CSPs can also be used in the

reversed-phase mode.

While normal-phase mode is generally

applied to determine enantiomeric

excesses of synthetic samples or for

preparative purposes, reversed-phase

conditions are usually preferred for

samples obtained from biological

matrices (stereoselective metabolism,

toxicity, and chiral stability). Figure 2

shows a typical example of chiral

separation, illustrating the increasing

complexity of the investigated drugs (with

multiple chiral centres) and the possibility

offered by the available tools to achieve

the separation of the four stereoisomers

under normal-phase conditions or under

Contemporary Analysis of Chiral MoleculesEric Francotte, FrancotteConsulting, Separation Sciences, Basel, Switzerland

The first high performance liquid chromatography (HPLC) column for enantioselective chromatography

was introduced commercially in 1981. This chromatographic mode has now become the method of

choice for the analysis of chiral pharmaceutical compounds, making previous approaches, such

as optical rotation, almost completely obsolete. However, supercritical fluid chromatography (SFC)

has been gaining increasing recognition as a complementary technique to HPLC for pharmaceutical

enantioselective analysis. Gas chromatography (GC) and capillary electrophoresis (CE) remain very

useful for particular applications.

31www.chromatographyonline.com

Ph

oto

Cre

dit: G

ett

yp

ho

to/G

ett

y Im

ag

es

Page 32: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

polar organic conditions, which are

more appropriate to investigate aqueous

samples on polysaccharide-based

CSPs.

As a result of the complexity of

the mechanism of interaction with

polysaccharide-based phases, it

is almost impossible to predict the

chiral separation on these phases.

Small variations of the structure of the

analyte, of the polysaccharide CSP, or

of the mobile-phase composition can

dramatically affect the separation, and

even cause the inversion of the elution

order of the enantiomers. Therefore,

in most cases, method development

is required to identify appropriate

conditions to separate the enantiomers.

Many factors can influence the

separation, and it is common practice

to screen a broad combination of CSPs

and mobile phases. Numerous strategies

have been proposed by different groups

for screening chiral stationary phases

(4–6). However, because of the rapidly

evolving market for chiral columns,

the setup has to be regularly adjusted.

The screening is usually performed on

single columns, which are successively

tested, but this mode is not very efficient,

particularly if a large number of chiral

substances need to be processed. This

is still the most common approach, but

it is now widespread to use switching

valves sequentially connected to the

different columns in an automated

fashion, which has the advantage of

being a relatively low-cost setup.

The screening process can be

accelerated by using an isochronal setup

(7), but the use of HPLC systems capable

of working in parallel clearly shows the

highest effectiveness (8). Our group

developed this strategy several years ago,

and it is currently used as the standard

procedure for chiral method development

(8). Under the standard setup, using

columns of 15 cm × 0.46 cm and a flow

rate of 0.8 mL/min (20 min per run), up

to 20 different conditions (CSP or mobile

phases) can be tested each hour. The

system is totally automated. The parallel

setup might also be useful if the analysis

of many samples of the same molecule is

required, for example, for the screening

of catalysts in asymmetric synthesis, for

samples from biological investigations, or

for samples from formulation optimization

studies. In these cases, the same CSP is

used on all channels of the parallel unit.

The necessity of running efficient

parallel screening approaches for chiral

method development has become even

more essential since the introduction of

the immobilized polysaccharide phases.

Indeed, with this new generation of chiral

columns, it is possible to apply strong

solvating modifiers like ethyl acetate,

tetrahydrofuran, dichloromethane,

chloroform, or dimethoxymethane

that are not tolerated by the classical

non-immobilized polysaccharide phases,

considerably increasing the potential to

improve the separation by modulating the

mobile phase. This is a great advantage

because it could help to optimize the

retention times and selectivity, which are

crucial parameters for enantioselective

analysis.

Various approaches have been

developed for the preparation of

immobilized polysaccharide phases (9),

including a particularly simple process

that our group developed (7,8,10)

and which has led to the introduction

of several CSPs of this type on the

market (11,12). Screening approaches

incorporating these phases have been

described (8,13).

For racemic substances that cannot

be resolved on polysaccharide-based

CSPs, a few other columns are generally

tested in a secondary screening. All

these phases have been designed by

bonding small chiral molecules to silica

gel. These phases include an amino

tetrahydrophenanthrene-based phase

(14), the cyclodextrin-based CSPs (15),

the cinchona alkaloid ionic phases (16),

the cyclofructan (17), the macrocyclic

peptides (18), and glycoprotein phases

(19), which together share about 10% of

the applications of the enantioselective

analyses performed in the

pharmaceutical field. The quinine-based

phase (20), the α-1-acid glycoprotein

(AGP) (or orosomucoid) (21), and the

cyclofructan (22) phases might exhibit

excellent chiral recognition power for

polar or ionic compounds, which often

fail on polysaccharide-based CSPs.

Figure 3 shows the analytical separation

of the enantiomers of free amino acids on

the new quinine-based ionic CSP (20).

Thousands of pharmaceutical

applications have been reported

attesting to the effectiveness of the

enantioselective HPLC technique.

Modern enantioselective HPLC is very

efficient (≤5-μm particle size) and an

enantiomeric ratio lower than 99.9/0.1

can usually be easily determined.

The method is also routinely applied

for validated processes (23,24) and

for enantioselective drug metabolism

assessment (25). Figure 4 shows a

recent application of the simultaneous

quantification of the enantiomers of the

drug lacosamide and its chiral impurity

on an immobilized polysaccharide-

based phase (24). Another typical

application of a high enantiomeric

excess (ee) determination for the drug

Pemetrexed also exemplifies that even

polar analytes can be well resolved on

polysaccharide-based phases (26).

It must also be mentioned that a

considerable number of enantioselective

Table 1: Major techniques for enantioselective analysis and major applications fields.

Techniques Major Application Fields

Chromatography Asymmetric synthesis and catalysis

t��$BQJMMBSZ�FMFDUSPQIPSFTJT�$& Chiral stability

t��&MFDUSPLJOFUJD�DISPNBUPHSBQIZ�&,$ Drugs

t��$BQJMMBSZ�FMFDUSPDISPNBUPHSBQIZ�$&$ t�4ZOUIFTJT

t�(BT�DISPNBUPHSBQIZ�($ t�#JPMPHJDBM�BDUJWJUZ

t��-JRVJE�DISPNBUPHSBQIZ�-$ �)1-$ t��.FUBCPMJTN �QIBSNBDPLJOFUJD

t��4VQFSDSJUJDBM�GMVJE�DISPNBUPHSBQIZ�4'$ t�'PSNVMBUJPO

t�2VBMJUZ�DPOUSPM

Spectroscopy Environment

t�0QUJDBM�SPUBUJPO Fragrances

t�.BTT�TQFDUSPNFUSZ�.4 Food

t��/VDMFBS�NBHOFUJD�SFTPOBODF�/.3 Pesticides (insecticides, fungicides)

t��7JCSBUJPOBM�DJSDVMBS�EJDISPJTN�7$% t�4ZOUIFTJT

t�#JPMPHJDBM�BDUJWJUZ

Sensor t�.FUBCPMJTN

t�&MFDUSPDIFNJDBM t�'PSNVMBUJPO

t�&MFDUSJDBM t�2VBMJUZ

t�(SBWJNFUSJD�NBTT

t��0QUJDBM�DPMPSJNFUSZ �GMVPSFTDFODF �413

Advances in Pharmaceutical Analysis October 201632

Francotte

Page 33: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

methods are developed in the context of

preparative separations or asymmetric

synthesis of drugs and drug intermediates.

For this purpose the polysaccharide-

based phases dominate again, with

more than 95% of the applications in the

pharmaceutical industry.

Recently, it has been shown that

ultrafast chiral HPLC can be achieved

within a few seconds, but no practical

applications have been reported so far

(27–28).

Analysis of Chiral Molecules

by SFC

Intensive research activities on

supercritical fluid chromatography

(SFC) in open tube capillaries and in

packed columns have been performed

since the early 1980s (29), but it took

many years until the technique was

really adopted. The poor acceptance

was mainly a result of the limited

robustness and low reliability of the

available instrumentation. From the

beginning of the new millennium,

the situation radically changed with

the significant improvement of the

new SFC instruments, and it was,

in fact, preparative enantioselective

chromatography that rescued packed

SFC (pSFC). Enantioselective separation

has been the major SFC application

for about 10 years and has now been

adopted by most pharmaceutical

companies for this purpose.

pSFC is very similar to normal-phase

HPLC—it “just” uses liquid CO2 as

the major component of the mobile

phase. However, in its supercritical

state, CO2 exhibits unique properties

that gives it some advantages in terms

of chromatographic performance

because of the low viscosity of the

fluid. Other advantages include

speed, efficiency, safety, costs,

and environmental impact (green

technology) (30).

Even though SFC is usually achieved

under sub-critical conditions, most

of these advantages remain. These

reasons have contributed to the rapid

establishment of packed SFC as a

powerful technique for enantioselective

analysis. Moreover, the switch from LC

to SFC was helped because the same

“chiral” columns can be used for either

LC or SFC.

The suitability of SFC for

enantioselective analysis of drugs has

been reviewed in a series of review

articles and book chapters, containing

an extended list of drug applications

(31–36). However, enantioselective

separations that can be achieved

on a particular CSP by HPLC do

not necessarily work under SFC

conditions (and vice versa). This is not

surprising because the mobile phases

are different and such effects are

observed in enantioselective HPLC on

polysaccharide-based stationary phases

when slight modifications of the modifier

composition are performed. Based on

our own experience, about 10% of the

chiral pharmaceuticals are better or

only resolved by HPLC, while the same

proportion is better resolved under SFC

conditions.

As for LC, any resolution of a new

racemic compound in SFC requires

method development. Several groups

have reported on their own process

flow (13,37–40), but because the chiral

column market is constantly evolving,

the setup has to be regularly adapted,

even though the preferred columns

33www.chromatographyonline.com

Francotte

CCCCChhhheeemmiccaallllyy iinnertt

TTeemmppperaturreee stableuuupp tto 20000ººC

Hamilton Bonaduz AG

CH-7402 Bonaduz

Switzerland

[email protected]

www.hamiltoncompany.com

HDHT. First cement-free syringefor PAL Combi-xt headspace autosamplers

Page 34: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

remain those based on polysaccharides.

Similar to HPLC, a parallel setup with

eight columns operating simultaneously

can be used for screening CSPs in

SFC (40,41). The instrument permits the

rapid identification of optimal conditions

(chiral column, mobile phase) to perform

the enantiomeric separations. Using a

standard configuration with columns of

15 cm × 0.46 cm, a flow rate of 3 mL/

min (7 min for each run), and a gradient

of 5–40% modifier within 5 min, up to

60 different conditions can be tested

within 1 h. In the primary screening,

eight polysaccharide-based CSPs have

been selected (40,41). Analogously

to enantioselective HPLC, almost

any combination of CO2 with organic

modifiers is feasible with the immobilized

polysaccharide-based CSPs because

they tolerate most organic solvents

(40,42).

Hundreds of pharmaceutical

applications have been published and

reported over the last 10 years and

surely many more have been performed,

in particular within the pharmaceutical

companies that apply SFC for hundreds

of new chiral molecules every year.

Although enantioselective SFC

is now well adopted and even the

method of choice in drug discovery

in most pharmaceutical companies,

there is only a very limited number of

validated methods (43,44) because

the SFC technique has not yet been

fully embraced by the pharmaceutical

development community. However,

considering the availability of robust and

high performing analytical instruments,

and the recent publication of numerous

papers demonstrating the possibilities

of SFC for pharmaceutical investigations

in a good laboratory practice/

manufacturing (GLP/GMP) environment,

it can be anticipated that this situation

will rapidly change in the next few years

(45,46).

Analysis of Chiral Molecules

by GC

GC was a major technique for

enantioselective analysis for two

decades (1980–2000). It was researched

intensively and reached a high level of

advancement (47). Numerous stationary

phases have been developed and a

few have been commercialized. The

most used phases are based on amino

acids and cyclodextrins (Figure 1). GC

is particularly appropriate for volatile

molecules, which is not often the case for

chiral drug compounds. However, it

is regularly applied to determine

the purity of volatile pharmaceutical

intermediates and it remains a standard

approach for enantioselective analysis

of amino acids. Nevertheless, the

introduction of the new cinchona

alkaloid ionic phases, which show a

wide application range, might change

this in the future (20). These latter ionic

phases have the advantage that they do

not require a derivatization of the amino

acids before analysis.

GC is very efficient and generally

shows a high separation power. Very

low selectivity values (alpha values

lower than 1.1) are sufficient to achieve

accurate ee determinations. Easy

coupling of GC to MS was also a major

Advances in Pharmaceutical Analysis October 201634

, ,

R

Amylose derivatives1 - acid glycoprotein (LC) Cinchona alkaloids

(LC, SFC, CE)

Quinine derivative (8S, 9R, 1S, 2S)

Quinidine derivative (8R, 9S, 1R, 2R)

Saccharide chains (~45%)

N-acetylglucosamine

Mannose

Galactose

Fucose

Sialic acid

Peptide chain

183 amino acids

(LC, SFC, CE, CEC)

Cellulose derivatives

(GC, LC, SFC, CE, CEC)

Cyclodextrins derivatives

(GC, LC, SFC, CE)

L-Valine-tert.butylamide

(GC)

Tetrahydro-

phenanthrene amine(LC, SFC)

Cyclofructans

CF6

(GC, LC, SFC, CE)

R

R

R

R

R

R

R

R

R

R

R

R

R

RR

O

O

O

O

OO

O

O η

η

η

β

β γ

α

α

O O

O

O

O

O

+

O

O

(S)

(S)

(S)

S

S

1

2 9

8

O

O

nOH

OH

RORO

RO

RO

RO RO

RO

OR

OR

OR OR

OR

OR

OR

OR

OR

OR

OR

OH

HO

H

HH

H

HN

HN

NH

NH

NH

H

H

HO

Cl

Cl NH

NH

HO

HO

HO

HOHO

OH

OH

OH

OH

NH

NH2

HOOH

OH

NH

NH

NO2

NO2

H

H

O

O

OO

O

O

O

O

O O

OO

NH

N

NH

H2N

O

O

O

O

O

O

O

O

O

O

OR

OROR

OR

OROR

NHNHSi

O

O

OR

OR OR

OR

OR

RO RO

RO

RO

RO

RO

RO

O

OO

O

O

O

O O O

O

O

O

O

O

O

O

O

O O

O

O

O

O

O

O

O

O

O

O

O

O

O

O

O

Figure 1: Structures of the most used chiral selectors for enantioselective chromatography. Applied techniques are in brackets.

Page 35: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

advantage compared to normal-phase

chiral separation for many sensitive

and biological applications. The

usefulness of GC for enantioselective

analysis has been recently reviewed by

C. Morrisson (48).

Recent applications include the chiral

analysis of nonprotein amino acids (49)

or pesticides in humans (50). The latest

examples of validated methods are the

enantioselective analysis for (L)-pidolic

(51) and the chiral GC–MS method for

the determination of free D-amino acids

ratio in human urine for gestational

diabetes studies (52).

Similar to LC and SFC, chiral

GC requires method development.

Although it is usually done by screening

GC columns one by one, Schafer

and colleagues proposed a parallel

chromatography screening system

(53). Interestingly, most of the current

applications are still performed on the

GC phases developed decades ago, but

there is still some research ongoing to

develop new and more efficient chiral GC

columns. Recent developments include

the utilization of cellulose derivatives

in coated open tubular capillaries,

which has been reported by Zhang

and colleagues (54). The chiral nematic

mesoporous silica column exhibits very

good chiral recognition abilities for a

broad variety of racemic compounds.

Enantioselective GC remains a popular

technique for the analysis of chiral

compounds and is the second technique

in importance after LC.

Analysis of Chiral Molecules

by Capillary Electrophoresis

(CE) and Micellar Electrokinetic

Chromatography (MEKC)

From the mid-1980s, enantioselective

capillary electrophoresis and micellar

electrokinetic chromatography have

been the focus of intensive research. CE

BOE�.&,$�IBWF�UIF�BCJMJUZ�UP�TFQBSBUF�

according to charges and are true

orthogonal methods of analysis because

they work through different mechanisms

compared to GC, LC, or SFC.

Much academic work has been

QFSGPSNFE�JO�$&�BOE�.&,$ �BOE�UIFSF�JT�

a vast amount of original papers on this

topic. Various reviews compiling extensive

lists of pharmaceutical applications are

available (55–57). Interestingly, as for the

other separation techniques, the chiral

selectors derived from saccharides

have developed as the most widely

applicable. The cyclic oligosaccharides

cyclodextrins clearly cover the majority

of the applications. The method has

undoubtedly some advantages in terms of

cost and practical operation.

Although numerous applications

for a wide variety of chiral drugs have

been reported, most of the published

works have been performed in

academic environments and CE has

not reached the level of acceptance

of enantioselective LC analysis in the

pharmaceutical industry. The technique

35www.chromatographyonline.com

Francotte

O7.0

2

8.2

4

8.3

9

9.9

9

10

.71

11

.68

a b c

c d a b

d

0 5 10 15 20 25

0 5 10 15 20

10

.34

17

.29

O

R2

R1R3

CF3

NH

NH

Time (min)

Time (min)

Figure 2: Method development for stereoselective drug metabolism analysis of a chiral bis-amido drug. Separation of the four stereoisomers under (a) normal-phase mode, column: 0.46 × 25 cm, 5-μm Chiralcel OZ (Chiral Technologies), mobile phase: heptane/ethanol/diethylamine 70/30/0.03, flow rate: 1 mL/min; (b) polar organic mode, column: 0.46 × 25 cm, 5-μm Chiralpak IE (Chiral Technologies), mobile phase: ethanol 5 mm + 0.03% diethylamine, flow rate: 0.5 mL/min. Elution order of the individual stereoisomers a–d is indicated.

O

O O

0 1 2 3 4 5 6 7Time (min)

O

OH

NH

NH2 NH2

OH

OH OH

DL-Pro

DL-Nva DL-Nle

DL-Hpr

(a)

(b)

0 1 2 3 4 5 6 7Time (min)

0 1 2 3 4 5 6 7

Time (min)

0 1 2 3 4 5 6 7

Time (min)

(d)

(c)

NH

Figure 3: Separations of the enantiomers of selected α amino acids with cyclic and acyclic side chains. Column: 150 × 3 mm, 5-μm Chiralpak ZWIX(+) (Chiral Technologies); mobile phase: 50 mM formic acid + 25 mM diethylamine as additives in methanol/acetonitrile/water 49:49:2; detection: ELSD. Adapted with permission from reference 20.

Page 36: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

is still suffering from its lack of robustness

and relatively low sensitivity, even

though some progress has been made

to improve this (58). The question of

the limited integration of CE in the

pharmaceutical industry was recently

discussed (59) and this trend is reflected

by the small number of CE publications

from this sector over the last three

years. Nevertheless, a few practical

applications have been achieved in the

industrial pharmaceutical environment,

including validated processes with UV or

MS detection (60–63).

Analysis of Chiral Molecules

by Spectroscopic Methods

Enantioselective analyses by

spectroscopic methods have drawn

the attention of many research groups

for a long time. In particular, NMR

using a chiral solvating agent (CSA)

(64) has been, besides optical rotation

(αD), a standard approach for the

determination of optical purity before

1980, at a time where no chiral stationary

phases were available. NMR using

CSA is a simple method and does

not need previous derivatization of

the analyte with a chiral molecule to

form diastereoisomers. It was much

used until the end of the century, but

because of its limited accuracy (± 3%)

NMR has been progressively replaced

by chromatographic techniques. The

value of the technique has recently

been reviewed (65) and applications to

pharmaceutical drugs are still regularly

published (66–68). A number of new

chiral solvating agents showing excellent

chiral discrimination properties were also

developed (69), giving accuracy values

approaching those of chromatography

on chiral stationary phases but still lower

(about 1% of the minor enantiomer).

Nevertheless, it is definitely less broadly

applicable than LC or GC.

MS has also been applied to

determine ee (70), and the promise of the

method has recently been reviewed (71).

The feasibility of using an MS instrument

solely for chiral recognition has

clearly been demonstrated and a first

practical application for the quantitative

determination of the chiral purity of an

antibiotic drug by flow-injection MS/

MS has been reported by Wu and

colleagues two years ago (72). However,

this approach is not broadly applicable

and it is likely that it will not reach the

level of popularity of the chromatographic

techniques.

VCD has also been reported to be a

suitable technique for ee determination

(73,74) and might be useful in particular

cases (75,76), but it cannot compete with

more generally applicable techniques for

daily use.

Enantioselective Sensors

The development and application of

enantioselective sensors is a topic of

increasing interest and might possibly

become a useful technique for ee

determination. Progress in this field

has recently been reviewed (77).

Very recent applications include the

discrimination of the enantiomers

of amino acids or ascorbic acid by

applying electrochemical processes

(78–80) or the enantiomers of naproxen

by recording change in the optical

properties (colorimetry) (81). Although

this approach requires the development

of a specific sensor for each particular

application, it can be of great interest

in diagnostic or environmental analysis

and has the advantage that it can be

miniaturized.

Conclusion

A broad variety of chromatographic and

spectroscopic approaches are available

for the enantioselective analysis and

determination of the chiral purity of

chiral drugs. Among all approaches,

chromatography is clearly the preferred

technique. Methods are easy to

elaborate with a relatively limited number

of chiral stationary phases, mainly

made from polysaccharide derivatives

in LC or SFC, and from cyclodextrins

or amino acids in GC, as the chiral

selectors. Interestingly, the number of

chiral selectors that have emerged as

the most widely applicable is relatively

limited, whatever the technique, and

even more fascinating is that the great

majority are composed of or contain a

large amount of saccharides. Compared

to the chromatographic methods,

spectroscopic approaches are usually

less accurate and are not so generally

applicable, even though their usefulness

has been demonstrated in some cases.

References(1) G. Hesse and R. Hagel, Chromatographia

6, 277−280 (1973).

(2) E. Francotte, H Stierlin, and J.W. Faigle, J.

Chromatography A 346, 321–31 (1985).

(3) J. Shen and Y. Okamoto, Chem. Rev. 116,

1094−1138 (2016).

(4) $�,��&TTFS �3�.��#MBDL �BOE�%��7PO�-BOHFO �

Pharmaceutical Discovery 4(9), 26–32

(2004).

(5) R. Sneyers, T. Vennekens, T. Huybrechts,

I. Somer, G. Torok, and S. Vrielynck, LCGC

Europe 20(6), 320–335 (2007).

(6) A. Younes, D. Mangelings, and Y. Vander

Heyden, J. Pharm. Biomed. Anal. 55(3),

414–423 (2011).

(7) H. Wetli and E. Francotte, J. Sep. Sci. 30,

1255–1261 (2007).

(8) E. Francotte, D. Huynh, and H. Wetli, G.I.T.

Laboratory Journal Europe 10, 46–48

(2006).

(9) J. Shen, T. Ikai, and Y. Okamoto, J.

Chromatogr. A 1363, 51–61 (2014).

(10) E. Francotte and D. Huynh, J. Pharm.

Biomed. Anal. 27, 421–429 (2002).

(11) C. Amoss, G. Cox, P. Franco, and T.

Zhang, LCGC’s The Application Notebook

(September 2008).

(12) +��-FF �8�-��8BUUT �+��#BSFOEU �5�2�:BO �:��

Huang, F. Riley, M. Hardink, J. Bradow, and

P. Franco, J. Chromatogr. A 1374, 238–246

(2014).

(13) P. Franco and T Zhang, in Chiral

Separations: Methods and Protocols, (�,�

Scriba, Ed. (Methods in Molecular Biology,

vol. 970), pp. 113–126.

Advances in Pharmaceutical Analysis October 201636

Francotte

0.25

0.030

0.025

0.020

0.015

0.010

0.005

0.000

-0.005

-0.010

0.20

0.15

0.10

0.05

0.00

Ab

sorb

an

ce AU

Ab

sorb

an

ce AU

Column: Chiralpak-IC

MP: n-Hexane: Ethanol

85:15 (v/v) (R)-

Imp

-1

(R)-LAC

(R)-LAC

(S)-LAC

(S)-

Imp

-1

(S)-LAC

O O

O

O

O

OH

NHNH

NH

H3C

H3C

CH3

LAC (lacosamide)

0.15% spiked chromatogram

Imp-1 (impurity)

NH0.00 2.00 4.00 6.00 8.00 10.00 12.00 14.00 16.00

0.00 2.00 4.00 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00 24.00

Time (min)

Time (min)

Figure 4: Separation of the enantiomers of the lacosamide drug and its chiral impurity on 250 mm × 4.6 mm, 5-μm Chiralpak-IC column (Chiral Technologies); mobile phase: n-hexane/ethanol 85:15 (v/v); flow rate: 1.0 mL/min; oven temperature: 27 °C. Adapted with permission from reference 24.

Page 37: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

(14) C.J. Welch, J. Chromatogr. 666, 3–26

(1994).

(15) X. Zhang, Y. Zhang, and D.W. Armstrong, in

Comprehensive Chirality, E.M. Carreira and

H. Yamamoto, Eds. (Elsevier, Amsterdam,

Netherlands, 2012), pp. 177–199.

(16) M. Lämmerhofer and W. Lindner, J.

Chromatogr. A 741, 33–48 (1996).

(17) P. Sun, C. Wang, Z.S. Breitbach, Y. Zhang,

and D. Armstrong, Anal. Chem. 81(24),

10215–26 (2009).

(18) A. Berthod, in Comprehensive Chirality,

E.M. Carreira and H. Yamamoto, Eds.

(Elsevier, Amsterdam, Netherlands, 2012),

pp. 227–262.

(19) J. Haginaka, in Comprehensive Chirality,

E.M. Carreira and H. Yamamoto, Eds.

(Elsevier, Amsterdam, Netherlands, 2012),

pp. 153–176.

(20) T. Zhang, E. Holder, P. Franco, and W.

Lindner, J. Sep. Sci. 37, 1237–1247 (2014).

(21) H.P. Jadhav and D.B. Pathare, Int. J. Pharm.

& Pharm. Sci. 7(7), 77–80 (2015).

(22) ,��)SPCPOPWÈ �+��.PSBWDÓL �+��-FIPUBZB �

and D.W. Armstrong, Anal. Methods 7,

4577–4582 (2015).

(23) Y.-Y. Zhang, X.-W. Liu, L.-J. Zhu, and M.

Yuan, Biomed. Chromatogr. 28(7),

1030–1035 (2014).

(24) ,��$IBSBHPOEMB �J. Chromatogr. Sep.

Techn. 6(5), 280. doi:10.4172/2157-

7064.1000280 (2015).

(25) Z. Shen, C. Lv, and S. Zeng, Journal of

Pharmaceutical Analysis 6, 1–10 (2016).

(26) ,��3BNVMV �#�.��3BP �1��.BEIBWBO �

.�-��%FWJ �.�,��4SJOJWBTV �BOE�,�#��

Chandrasekhar, Chromatographia 65(3/4),

249–252 (2007).

(27) D.C. Patel, Z.S. Breitbach, M.F. Wahab, C.L.

Barhate, and D.W. Armstrong, Anal. Chem.

87(18), 9137–9148 (2015).

(28) O.H. Ismail, A. Ciogli, C. Villani, M. De

Martino, M. Pierini, A. Cavazzini, D.S. Bell,

and F. Gasparrini, J. Chromatog. A 1427,

55–68 (2016).

(29) .�-��-FF�BOE�,�&��.BSLJEFT �Analytical

Supercritical Fluid Chromatography and

Extraction (Chromatography Conferences,

Inc. Provo, Utah, USA, 1990).

(30) C. Brunelli, A. Pereira, F. David, M. Dunkle,

and P. Sandra, LCGC Europe 23(8),

396–405 (2010).

(31) C. West, Cur. Anal. Chem. 10, 99–120

(2014).

(32) ,��,BMJLPWB �5��4MFDIUPWB �+��7J[LB �BOE�&��

Tesarova, Anal. Chim. Acta 82, 1–33 (2014).

(33) J.M. Plotka, M. Biziuk, C. Morrison, and

J. Namiesnik, Trends in Anal. Chem. 56,

74–89 (2014).

(34) ,��%F�,MFSDL �%��.BOHFMJOHT �BOE�:��7BOEFS�

Heyden, J. Pharm. Biomed. Anal. 69, 77–92

(2012).

(35) D. Mangelings and Y. Vander Heyden, J.

Sep. Sci. 31, 1252–1273 (2008).

(36) $�+��8FMDI �.��#JCB �+�3��(PVLFS �(��,BUI �

P. Augustine, and P. Hosek, Chirality 19,

184–189 (2007).

(37) C. Hamman, M. Wong, I. Aliagas, D.F.

Ortwine, J. Pease, D.E. Schmidt, and J.

Victorino, J. Chromatogr A. 1305, 310–319

(2013).

(38) N. Hicks, J. Stafford, and V.S. Sharp, LCGC

North America 31(8), 622–628 (2013).

(39) R.M. Woods, Z.S. Breitbach, and D.W.

Armstrong, LCGC Europe 28(1), 26–33

(2015).

(40) E. Francotte, LCGC Europe 29(4), 194–204

(2016).

(41) E. Francotte, “SFC – The Universal

Separation Technique?”, paper presented

at the 4th International Conference on

Packed-Column SFC, Stockholm, Sweden,

2010.

(42) E. Francotte and G. Diehl, “Extending

the SFC Applicability in the Field of

Enantiomeric Separations”, paper

presented at the 2nd International

Conference on Packed-Column SFC,

Zurich, Switzerland, 2008.

(43) Y. Huang, in Pharmaceutical Analysis �(�,��

Webster, Ed. (Pan Stanford Publishing, CRC

Press, Boca Raton, Florida, USA, 2014), pp.

225–263.

(44) Z. Yang, X. Xu, L. Sun, X. Zhao, H. Wang,

J.P. Fawcett, Y. Yang, and J. Gu, J.

Chromatogr. B 1020, 36–42 (2016).

(45) A. Grand-Guillaume Perrenoud, C.

Hamman, M. Goel, J.-L. Veuthey, D.

Guillarme, and S. Fekete, J. Chromatogr. A

1314, 288–297 (2013).

(46) M.B. Hicks, E.L. Regalado, F. Tan, X. Gong,

and C.J. Welch, J. Pharm. Biomed. Anal.

117(5), 316–324 (2016).

(47) V. Schurig and M. Juza, Adv. Chromatogr.

52, 117–168 (2014).

(48) C. Morrison, in Comprehensive Chirality,

E.M. Carreira and H. Yamamoto, Eds.

(Elsevier, Amsterdam, Netherlands, 2012),

pp. 333–353.

(49) S. Fox, H. Strasdeit, S. Haasmann, and H.

Brueckner, J. Chromatogr. A 1411, 101–109

(2015).

(50) C. Corcellas, E. Eljarrat, and D. Barcelo,

Anal. Bioanal. Chem. 407(3), 779–786

(2015).

(51) J.J. Salisbury, M. Li, and A. Boyd, J.

Pharmac. Biomed. Anal. 120, 79–83

(2016).

(52) M.P. Lorenzo, D. Dudzik, E. Varas, M.

Gibellini, M. Skotnicki, M. Zorawski, W.

Zarzycki, F. Pellati, and A. Garcia, J.

Pharmac. Biomed. Anal. 107, 480–487

(2015).

(53) W. Schafer, S.E. Hamilton, Z. Pirzada, and

C.J. Welch, Chirality 24(1), 1–4 (2012).

(54) J.-H. Zhang, S.-M. Xie, M. Zhang, M. Zi,

P.-G. He, and L.-M. Yuan, Anal. Chem.

86(19), 9595–9602 (2014).

(55) B. Chankvetadze, Capillary Electrophoresis

in Chiral Analysis�8JMFZ �$IJDIFTUFS �6, �

1997).

(56) S. Fanali, J. Chromatogr. A 875, 89–122

(2000).

(57) H. Lu and G. Chen, Anal. Meth. 3(3),

488–508 (2011).

(58) E. Sanchez-Lopez, M.L. Marina, and A.L.

Crego, Electrophoresis 37, 9–34 (2016).

(59) C. Sänger-van de Griend, LCGC North

America 30(11), 954–971 (2012).

(60) '�"��"HVJBS �$�.��EF�(BJUBOJ �BOE�,�#�

Borges, Electrophoresis 32(19), 2673–2682

(2011).

(61) J. Schappler, D. Guillarme, J. Prat, J.-L.

Veuthey, and S. Rudaz, Electrophoresis

29(10), 2193–2202 (2008).

(62) :��2J�BOE�9��;IBOH �Cell Biochem. Biophys.

70(3), 1633–1637 (2014).

(63) Z.-I. Szabo, D.-L. Muntean, L. Szocs

Levente, B. Noszal, and G. Toth, Chirality

28(3), 199–203 (2016).

(64) D. Parker, Chem. Rev. 91, 1441−1457

(1991).

(65) T.J. Wenzel, Topics in Current Chemistry

341, 1–68 (2013).

(66) X. Liang, R. Gopalaswamy, F. Navas, E. J.

Toone, and P. Zhou, J. Org. Chem. 81(10),

4393–4398 (2016).

(67) M.M. Cavalluzzi, A. Lovece, C. Bruno, C.

Franchini, and G. Lentini, Tetrahedron:

Asymmetry 25(24), 1605–1611 (2014).

(68) J. Redondo, A. Capdevila, and S. Ciudad,

Chirality 25(11), 780–786 (2013).

(69) P. Borowiecki, Tetrahedron: Asymmetry

26(1), 16–23 (2015).

(70) M. Shizumaa, H. Imamurab, Y. Takaib,

H. Yamadab, T. Takedaa, S. Takahashib,

and M. Sawadab, Intern. J. Mass Spect.

210/211, 585–590 (2001).

(71) H. Awad and A. El-Aneed, Mass

Spectrometry Rev. 32(6), 466–483 (2013).

(72) -��8V �'�(��7PHU �BOE�%�2��-JV �Anal. Chem.

85(10), 4869–4874 (2013).

(73) L.A. Nafie, F. Long, T.B. Freedman, H.

Buijs, A. Rilling, J.-R. Roy, and R. Dukor,

AIP Conference Proceedings 430, 432–434

(1998).

(74) P.L. Polavarapu and J. He, Analytical

Chemistry 76(3), 61A–67A (2004).

(75) -��,PUU �+��1FUSPWJD �%��1IFMQT �3��3PHJOTLJ �

and J. Schubert, Applied Spectroscopy

68(10), 1108–1115 (2014).

(76) M.E. Said, P. Vanloot, I. Bombarda, J.-V.

Naubron, E.M. Dahmane, A. Aamouche,

M. Jean, N. Vanthuyne, N. Dupuy, and C.

Roussel, Anal. Chim. Acta 903, 121–130

(2016).

(77) ,��.BOPMJ �.��.BHMJVMP�BOE�-��5PSTJ �Topics

in Current Chemistry 341, 133–176 (2013).

(78) H. Gou, J. He, Z. Mo, X. Wei, R. Hu, Y.

Wang, and R. Guo, J. Electrochem. Soc.

163(7), B272–B279 (2016).

(79) 2��#J �4��%POH �:��4VO �9��-V �BOE�-��;IBP �

Anal. Biochem. 508, 50–57 (2016).

(80) *��1BOEFZ�BOE�3��,BOU �Biosensors &

Bioelectronics 77, 715–724 (2016).

(81) '��,FTIWBSJ �.��#BISBN �BOE�"��'BSTIJE �

Anal. Methods 7(11), 4560–4567 (2015).

Eric Francotte received his Ph.D. in

organic chemistry from the University of

Louvain in Belgium, and spent two years

as a postdoctoral fellow at the University

of Geneva in Switzerland. He joined

former Ciba-Geigy (Novartis) in 1980

where he established a centre of expertise

for the development and application of

chiral polymers for the chromatographic

resolution of chiral compounds. After 10

years he moved to Novartis pharma as an

executive director and was responsible

for a global technology platform dealing

with separation sciences. His major

achievements include the invention of

an innovative process to immobilize

polysaccharide derivatives as chiral

stationary phases. Several phases

arising from this technology have been

marketed as the result of an external

collaboration and are considered as the

new gold standards in enantioselective

chromatography. He was also a

pioneer in the implementation of new

chromatographic technologies, such

as SMB and SFC. He is the author or

co-author of numerous publications,

patents, and editor of several books.

He has chaired various international

symposia in the field of preparative

chromatography, chirality, and SFC. He

is now an independent consultant for

separation science.

37www.chromatographyonline.com

Francotte

Page 38: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

The ever-growing problem of drug

purity and counterfeit medicine needs

to be addressed. It is estimated that

an important portion of all world trade

in branded pharmaceutical products

is counterfeit, leading to a great health

risk and public health concern. As

witnessed by numerous official sources,

including the World Health Organization

(WHO), the proportion of counterfeit

medicines has tremendously increased,

representing more than 50% in some

developing regions of the globe (1). The

primary reported cases of counterfeits

are affiliated to emerging countries, where

economically middle-income markets are

most represented (that is, Sub-Saharan

African countries, as well as Asian and

Latin American regions). According to

health-related government agencies,

the most prevalent therapeutic category

affected by counterfeits is anti-infectives

(21.1%), with the much needed antibiotic,

antiparasitic, and antiretroviral medicines

(2). Discovering and determining

counterfeit drugs is mandatory not only

for these countries, but also for others,

particularly when considering the internet

market (3). In order to characterize

counterfeit and substandard medicines

(Figure 1), different levels of action can be

applied. The evaluation of the packaging

based on a comparison with the genuine

sample and the search for manipulation

signs is appropriate for detecting

counterfeits, whereas chemical methods

should be implemented to authenticate

the samples and provide the chemical

composition of substandards (7).

Characterization of Counterfeit

and Substandard Medicines with

Chemical Analysis

There is an urgent need to set up tools

and workflows to fight against counterfeit

and substandard medicines and their

associated health risks. Evaluating

the quality of active pharmaceutical

ingredients (APIs) and excipients usually

requires a broad variety of modern

analytical methodologies, allowing the

qualitative and quantitative determination

of the content and the purity of the drugs

(8).

Methods for qualitative analysis include

basic but critical physical, organoleptic,

and haptic tests, as recommended

by international Pharmacopoeia.

Colorimetric assays using chemical

assays reacting with functional groups

is another traditional way to identify drug

compounds, although false-positive

results may be obtained because of

the low specificity of such assays,

circumventing the determination of

structurally similar molecules, such as

impurities. These tests are therefore

mainly implemented as a first and

rapid screening to establish only the

presence versus absence of an API in

a medicine. Spectroscopic techniques,

such as Raman, near infrared (NIR),

and nuclear magnetic resonance (NMR)

spectroscopy, are used to control the

quality of pharmaceuticals because

they provide a wealth of information on

the molecular structure of the APIs and

excipients, as well as on their spatial

distribution. Separation techniques, such

as thin-layer chromatography (TLC), high

performance liquid chromatography

(HPLC), gas chromatography (GC),

and capillary electrophoresis (CE), are

more selective assays and can be used

for qualitative as well as quantitative

determination. They are fully established

and have been added to almost all

monographs of major Pharmacopoeia for

quality control (QC) purposes. The higher

selectivity of hyphenated techniques

(that is, with mass spectrometry [MS])

offers detailed pictures of an analyte or

an analyte mixture, while the structure

of unknown contaminants can be

elucidated and determined in very small

quantities. In modern QC laboratories

these analytical instruments are mostly

available, allowing for an extensive

product testing during production

and during the distribution within the

respective supply channels. However,

they need trained people to use them

and samples generally need to be

brought to the laboratory to be analyzed.

In emerging countries, comprehensive

QC of circulating medicines is hardly

achieved because of relatively

young healthcare systems, restricted

laboratory capacities, weak analytical

infrastructures, and chaotic distribution

logistics. Therefore, a sequential

Characterization of Counterfeit and Substandard Medicines Using Capillary ElectrophoresisJulie Schappler and Serge Rudaz, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU,

Geneva, Switzerland

The proportion of counterfeit medicines has increased dramatically. Combatting this issue is complex, and

various levels of action are necessary. The quality control (QC) of imported batches using simple, reliable,

and cost-efficient analytical approaches is vital. Capillary electrophoresis (CE) is becoming important

because the analysis is achieved in a capillary with small dimensions, and is usually filled with an aqueous

buffer. No organic solvent is required and injection volumes are in the nanolitre range, which is convenient

because of the low availability of reference substances and reduces the environmental impact. CE is now

recognized by numerous Pharmacopeia and can be used for counterfeit and substandard characterization

as a validated analytical procedure that adheres to international guidelines.

Ph

oto

Cre

dit: H

ero

Im

ag

es/G

ett

y Im

ag

es

Advances in Pharmaceutical Analysis October 201638

Page 39: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

approach is recommended, according to

the goal of the analysis, the localization of

the laboratory, and the overall technical

skills of the analyst (Figure 2). In a first

approach, a large screening can be

performed with a nondestructive method,

for example, vibrational spectroscopy

(Raman, NIR). These methods give rapid

and low false-negative results, can be

brought to the field where the samples

can be tested, are sufficiently easy to

handle by people with little training, and

can be conducted without special, or

reduced, sample preparation (9). When

a sample does not comply in terms

of identity or quantity, a confirmatory

analysis should be done with an

orthogonal approach, such as that

afforded by separation techniques. As

previously mentioned, separation-based

devices including GC, LC, or CE are often

not portable, therefore the suspicious

samples are generally brought to the

laboratory where the analysis takes place

by trained people (10).

Rodionova et al. applied the

sequential approach for the

detection of counterfeited ampoules

of dexamethasone (11). First, NIR

measurements with chemometric

data processing were performed

directly in the closed ampoules of the

injectable formulations. The technique

was able to detect small differences in

the samples and emphasized reliable

discrimination between genuine and

counterfeit samples, using a modelling

set that included samples from various

batches (Figure 3[a]). Samples were then

subjected to GC–MS, HPLC coupled to

a diode array detector (DAD), and CE

coupled to an ultraviolet (UV) detector

for confirmation. GC–MS did not find any

differences between genuine and fake

samples, avoiding the discrimination

between both samples. HPLC–DAD

revealed that samples from different

batches were identical, but for some

impurities they differed in quantity. A

new impurity (marked with an asterisk)

was found in a fake sample (Figure 3[b])

and MS was further applied for its

identification. According to the high

efficiency and orthogonal selectivity of

the separation, CE–UV reliably found

three impurities in the fake sample that

were not present in the genuine sample

(Figure 3[c]). In this study, the CE method

showed the most informative results

for discriminating samples with close

chemical composition.

As can be seen from this study,

chromatographic approaches can be

applied for the confirmatory analysis,

but they suffer from several drawbacks

that often hamper their use in emerging

countries: (i) they are expensive to

acquire, maintain, and run; (ii) high

amounts of reference material and

organic solvents are needed; (iii) the

consumables and the chemicals may be

prohibitive and difficult to obtain.

In this context, CE has a role to

play in this fight because it offers

a low consumption of sample and

reference material, little use of organic

solvent, affordable consumables and

maintenance, and a basic requirement of

chemistry knowledge.

Characterization of Counterfeit

and Substandard Medicines Using

Capillary Electrophoresis

The well-established CE technology can

be applied for the characterization of

counterfeit and substandard medicines.

In this article we present several CE

applications, as well as developments

that were made in this context towards

the instrumentation and the methods.

Applications: Vidal et al. developed

a method based on CE coupled to

capacitively coupled contactless

conductivity detection (C4D) for

the determination of an important

counterfeited medicine, sildenafil, used

for treating erectile dysfunction (12).

Experiments were performed with a

laboratory-made CE device equipped

with two C4D detectors in series. The first

detector afforded a low efficiency but

fast detection, while the second detector

was used to obtain higher efficiency

results. The background electrolyte

(BGE) was made of acetic acid 0.5 M

and the capillary was coated prior to a

series of analyses with a cationic coating

to prevent analyte adsorption on to the

capillary wall. An interesting feature of

C4D detection was the possibility to

detect in the same run, not only sildenafil,

but also its associate salt (citrate).

Thus, commonly counterfeited tadalafil

or vardenafil tablets (which should be

exempt of citrate) containing sildenafil

could be easily detected.

39www.chromatographyonline.com

Schappler and Rudaz

Counterfeits Su

bst

an

dard

s

Fails quality standards

IllegalLegal

Meets quality standards

Illegitimatebad treatment

Legitimatebad treatment

Legitimategood

treatment

Illegitimategood

treatment

Figure 1: Categories of medicines. The medicines failing quality standards are considered “substandards” and they can be legitimate or illegitimate. The unapproved medicines are considered “counterfeits” and they can meet or not meet the quality specifications.

Laboratory

On-site

Pharmacopeia QC

2. Confirmatory assays

1. Large screening

Sequential approach for the characterization ofcounterfeits and substandardsin emerging countries

Governmental

authorities

for example, separation techniques,

hyphenated techniques

for example, colorimetric assays,

spectroscopic techniques

Figure 2: Proposed hierarchic testing of medicines using different analytical approaches.

Page 40: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

Antimalarial drugs are another major

class of counterfeit medicines that are

dramatically present on the African

market. Lamalle et al. developed a

CE–UV method for 15 antimalarials (13).

As these molecules cannot be ionized

at the same pH, micellar electrokinetic

chromatography (MEKC) was preferred

over capillary zone electrophoresis,

allowing the separation of neutral

compounds. Preliminary experiments

were first performed to select the

most crucial factors (including pH,

surfactant concentration, acetonitrile

proportion, and temperature). Next,

an experimental design methodology

was applied to enable the prediction

of optimal conditions for best analyte

separation in less than 8 min. Finally,

the method was successfully applied to

the quality control of African antimalarial

medicines for their qualitative and

quantitative content. At the same time,

another European laboratory developed

a MEKC–UV method for artesunate and

amodiaquine in fixed-dose combination

tablets (14).

Capillary electrophoresis was also

used to determine adulterants in

pharmaceuticals. De Carvalho et al.

applied CE–C4D for the determination

of adulterants in herbal weight loss

products collected in pharmacies

located in different Brazilian states.

A first study emphasized that either

anxiolytics, diuretics, or laxatives were

added to 4% of the weight products

collected (15). In a second study, CE

analyses revealed that more than 30% of

the analyzed formulations were found to

contain diuretics in the final composition

(16). These drugs have a low incidence

of side effects, but are associated

with several metabolic disorders

when administrated in high doses.

The developed CE procedure, which

enables a rapid and selective screening,

is now considered by governmental

organizations for inspecting commercial

dietary supplement formulations.

In another study, Cianchino et al.

developed a CE method with a

basic BGE to obtain a characteristic

fingerprint of Hedera helix L., used as

a cough treatment in Argentina (17).

The developed approach enabled the

determination of adulterants, such as

synthetic drugs used in respiratory

diseases (for example, ephedrine,

codeine, diphenhydramine) in

phytopharmaceutical formulations. The

fingerprint could also help to distinguish

differences between Hedera helix L.

from various sources.

Instrumental Advances:

Introducing high-quality separation

analysis into the daily fight for major

health and public safety issues and doing

it at an affordable cost is challenging.

Taking into account the limited technical

and financial resources in most

developing countries, a few initiatives

have emerged to provide sustainable

and affordable CE instruments that can

be easily implemented at a low cost by

organizations within these countries. For

example, a collaboration between Swiss

academic institutions and hospitals was

implemented to build a low-cost CE

device and help transitional countries

with limited means to fight against

counterfeit and substandard medicines

(18). A series of iteratively refined and

field-proofed CE units were built. The

first prototype addressed most of the

mechanical and electronic issues, and

included an original UV detection device

based on a LED technology. The second

generation improved the software and

the apparatus ergonomics, while the

last prototype included modifications

to improve the system robustness.

Methods were developed for drugs

selected from among a list provided by

several African partners. Marini et al.

evaluated the analytical performance

of this low-cost CE equipment for drug

quantification (19). A complete validation

study with reduced requirements

regarding calibration purposes was

Advances in Pharmaceutical Analysis October 201640

Schappler and Rudaz

(a)

(b)

(c)

4.0

3.0

2.0

5500 7000

5

5 6 7 8

10 15 20 25

8500 10000

1.0

1000

800

600

400

0.8

0.6

0.4

0.2

0.0

-0.2

-0.4

200

0

0.0

cm-1

Time (min)

Time (min)

F2

G2

G1

G1

Absorbance

units (AU)

Absorbance

units (m

AU)

Absorbance

units (m

AU)

F2

Figure 3: Detection of counterfeited ampoules of dexamethasone using several analytical approaches. (a) A raw NIR spectra of 30 genuine samples (blue lines) and 15 fakes (red lines); (b) HPLC–DAD chromatograms of a fake (F2) and genuine (G1 and G2) samples; (c) CE–UV electropherograms of a fake (F2) and genuine (G1) sample. Adapted with permission from reference 11.

Page 41: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

performed on medicines representing the

most targeted pharmacological groups

by counterfeiting, that is, antimalarial,

diuretic, and anti-infective products.

A comparison study was performed

with commercial CE equipment and

equivalent quantitative performance

was obtained between both devices,

demonstrating the great potential of this

simple and low-cost device (19). Finally,

concrete positive results in emerging

countries (mostly in Africa and in Asia)

were obtained, thanks to cooperation at

a geographically well-distributed series

of sites, implementing the prototype

instruments there and successfully

training the local personnel to accomplish

reliable identification and quantification of

pharmaceuticals. Sarr et al. implemented

the device at the Senegalese National

Medicines Control Laboratory where

a CE method was developed and

validated for the quality assessment

of metronidazole-based drugs (20).

Eleven metronidazole samples were

taken from various markets in Benin and

Senegal and analyzed by CE–LED/UV.

Fortunately, all collected samples were

compliant with USP specifications. The

results obtained with the low-cost CE

apparatus were confirmed with an official

chromatographic method described in

the US Pharmacopoeia. Similar results

for both methods with comparable

precision were obtained, demonstrating

that low-cost CE analysis could be a real

alternative in developing countries for

drug QC to protect population health.

It should be noted that although

portability of the device is not afforded

by the main suppliers, CE can also

present a great potential in point-of-care

diagnostics, where the speed of the

analysis, the device compactness, and

the sample volume are critical factors.

Gregus et al. developed a homemade

novel, simple, portable CE instrument

equipped with C4D for the analysis

of small volumes of biological fluids

(21). The instrument is light (<5 kg),

all necessary parts including a tablet

computer are accommodated with

small dimensions (20 × 33 × 17 cm),

hydrodynamic injection is performed

with less than 10 μL sample volume, and

the device can continuously operate for

at least 10 h. Several applications were

demonstrated, including the diagnostics

of respiratory tract diseases (for example,

asthma, chronic obstructive pulmonary

disease, cystic fibrosis).

Recent Method Developments: To

analyze a high number of compounds

and benefit from CE with basic chemistry

knowledge and training, simple and

generic methods should be applied to

determine counterfeit and substandard

medicines. In this context, Schappler

et al. developed a strategy based on

multiple injections to simultaneously

characterize and quantify APIs in

medicines in one single run, using an

external calibration and an internal

standard (22). Two generic CE–UV

methods were developed, using a BGE

at an acidic or a basic pH for basic or

acidic compounds, respectively. More

than 80 drugs from the list of the 200

essential medicines defined by WHO

could be analyzed with this approach.

Both methods were fully validated

according to international guidelines for

two important APIs (metronidazole and

ampicillin). They were further applied

for QC of formulations and detection of

substandards in samples from Tanzania.

This methodology was also implemented

for the simultaneous identification and

quantification of insulin formulations,

obtained from regular and parallel

markets, by CE with time-of-flight mass

spectrometry (TOF-MS) detection (23).

Particular attention was paid to the BGE

composition and acetonitrile added to the

BGE to enable direct MS coupling as well

as reduction of the protein adsorption.

The multiple injection approach offered

an alternative way to compensate for

ionization variability and matrix effect in

the absence of stable isotope-labelled

compounds for insulin. Two injections

were performed in the same analytical

run, the first one with a standard of insulin

at a known concentration and the second

one with the sample to be identified and

quantified. Figure 4(a) shows the total

ion electropherogram (TIE) obtained

with this multiple injection approach. The

41www.chromatographyonline.com

Schappler and Rudaz

(a) 2

x106

a

a

b

b

x103

1

1

0

x105

4

2

0 2 4 6

1000 1500

6 7 8

2000

Counts versus Acquisition Time (min)

Counts versus Acquisition Time (min)

Counts versus Mass-to-Charge (m/z)

8

[M+3H]3+

1452.921

[M+4H]4+

1936.557

7.349

6.518

(b)

(c)

Figure 4: Analysis of insulin by CE–TOF-MS. (a) Total ion electropherogram (TIE); (b) extracted mass spectrum used for identification purpose; (c) extracted ion electropherogram (XIE) used for quantification purposes. Adapted with permission from reference 23.

Page 42: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

(M+3H)3+ and (M+4H)4+ multicharged

ions were detected as the major extracted

ions (1937 and 1453 m/z, respectively,

(Figure 4[b]) and enabled identification

of insulin in unknown samples. The

extracted ion electropherogram (XIE)

was reconstructed using both ions and

integration was achieved for quantification

purposes (Figure 4[c]). It should be

noted that this approach is currently not

adapted for emerging countries because

of the expense and the need for highly

trained MS coupling.

Lamalle et al. alternately used

MEKC–UV to distinguish between human,

bovine, and porcine insulins in less than

12 min and easily determined if human

insulin was replaced by bovine or porcine

insulin in counterfeited pharmaceutical

formulations (24).

Conclusions

The expanding use of counterfeit drugs

in developed and emerging countries

is a growing problem and is associated

with challenges in QC. CE features high

performance (qualitative and quantitative

methods can be implemented and

validated), high sustainability (one

analysis consumes less than 1 μL

of sample, reference material, and

solvent), and low costs (analyses and

maintenance are affordable), and so it

is currently recognized as a contributing

technique that helps health organization

laboratories to spot check medicines

in an effort to monitor counterfeit and

substandard drugs and therefore deliver

high-quality medicines to vulnerable

populations.

A few issues still remain to fully

implement CE technology in emerging

countries. Because CE relies on a high

electric field, the input electric power has

to be reliable, which is, unfortunately,

not the case, with many countries

experiencing unpredictable power

breakdowns. Further improvements

should therefore be implemented in

CE equipment to render the device

more autonomous, sustainable, and

accessible, at even lower costs: (i) a

renewable energy source to supply high

voltage regardless of the local electricity

facilities; (ii) a universal detection system

to broaden the analysis range to any

pharmaceutical drug; (iii) a reduction of

the whole device dimensions to obtain

a disposable and portable system; and

(iv) integrated software to simplify data

treatment and reporting by the user. In

addition to continuous improvements

of the methodological and technical

aspects of the analytical device,

educational programmes should also

be developed and implemented (for

example, with e-learning tools) to provide

adequate initial and continuing guidance,

as well as sustainable and readily

deliverable training.

In this battle, a big stake is the share

of the information. Counterfeit medicines

are distributed through international

routes and it can be difficult to predict

them. Efforts should be made to quickly

and accurately detect points of entry and

locate where to perform the first step of

the analysis. The contributions of forensic

scientists will be decisive in all technical

aspects, including profiling seizures

with chemical and packaging data. The

interpretation of data on the production

and the distribution of counterfeits has

the potential to be a reliable approach to

aid in the understanding of the organized

crime phenomenon behind counterfeiting

and to enable efficient and operational

strategies for decision making (25). In

this context, citizen science projects

(for example, Hackathon at CERN [26])

are emerging that collect information

on medicines with devices that can be

used by anyone (such as smartphones

to take pictures, geolocalize products,

and create and curate databases).

This approach is particularly attractive

and complementary to chemical

analysis because it involves the global

population in the constant battle against

counterfeiting. Another important aspect

of medicines relates to the trust in drugs

by the patients themselves. Blockchain

databases, when combined and fully

integrated with analytical approaches,

offer an additional tool to strengthen

the trust and monitor the quality in drug

distribution chains, and should be

investigated further.

References(1) Bulletin of the World Health Organization,

88(4), 241–320 (April 2010).

(2) T. Mackey, B. Liang, P. York, and T. Kubic,

Am. J. Trop. Med. Hyg. 92, 59–67 (2015).

(3) B. Baert and B. De Spiegeleer, Anal.

Bioanal. Chem. 398, 125–136 (2010).

(4) R. Marini, J. Kindenge, M. Montes, B.

Debrus, P. Lebrun, J. Mantanus, E.

Ziemons, C. Rohrbasser, S. Rudaz, and P.

Hubert, Chemistry Today 28, 10–14 (2010).

(5) http://www.who.int/medicines/regulation/

ssffc/definitions/en/

(6) A. Attaran, S. Basheer, R. Bate, D. Benton,

J. Chauvin, L. Garret, I. Kickbush, J. Kohler,

K. Midha, P. Newton, S. Nishtar, P. Orhii,

and M. McKee. BMJ 1–6 (2012).

(7) K. Degardin and Y. Roggo, Drug Testing

Analysis 8, 388–397 (2015).

(8) R. Martino, M. Malet-Martino, V. Gilard, and

S. Balayssac, Anal. Bioanal. Chem. 398,

77–92 (2010).

(9) O. Rodionova and A. Pomerantsev, Trends

Anal. Chem. 29, 795–803 (2010).

(10) L. Höllein, E. Kaale, Y. Mwalwisi, M.

Schulze, and U. Holzgrabe, Trends Anal.

Chem. 76, 60–70 (2016).

(11) O. Rodionova, A. Pomerantsev, L.

Houmoller, A. Shpak, and O. Shpigun, Anal.

Bioanal. Chem. 397(5), 1927–1935 (2010)

(12) D. Vidal, M. Augelli, and C. do Lago, Anal.

Methods 5, 2041–2045 (2013).

(13) C. Lamalle, R. Marini, B. Debrus, P. Lebrun,

J. Crommen, P. Hubert, A.-C. Servais, and

M. Fillet, Electrophoresis 33, 1669–1678

(2012).

(14) N. Amin, M.-D. Blanchin, M. Aké, J.

Montels, and H. Fabre, Malaria J. 11,

149–155 (2012).

(15) L. De Carvalho, P. Cohen, C. Silva,

A. Moreira, T. Falcao, T. Dal Molin, G.

Zemolin, and M. Martini, Food Additives &

Contaminants: Part A 29, 1661–1667 (2012).

(16) A. Moreira, M. Motta, T. Dal Molin, C. Viana,

and L. De Carvalho, Food Additives &

Contaminants: Part A 30, 1230–1237 (2013).

(17) V. Cianchino, C. Ortega, G. Acosta, L.

Martinez, and M. Gomez, Pharmazie 62,

262–265 (2007).

(18) C. Rohrbasser, D. Rhème, S. Décastel,

S. Roth, M. Montes, J.-L. Veuthey, and S.

Rudaz, Chimia 63, 890–891 (2009).

(19) R. Marini, E. Rozet, M. Montes, C.

Rohrbasser, S. Roht, D. Rhème, P.

Bonnabry, J. Schappler, J.-L. Veuthey, P.

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

Anal. 53, 1278–1287 (2010).

(20) S. Sarr, E. Diop, A. Diop, A. Atouni, S.

Ndiaye, C. Wafo Tchounga, R. Gueye,

K. Thiam, T. Wane, A. Sarr, B. Ndiaye, S.

Rudaz, and Y. Diop, Int. J. Modern Anal.

Sep. Sci. 5, 1–11 (2016).

(21) M. Gregus, F. Foret, and P. Kubán, J.

Chromatogr. A 1427, 177–185 (2016)

(22) J. Schappler, E. Reginato, E. Assane Diop,

and S. Rudaz, Spectra Analyse 298, 63–73

(2014).

(23) A. Staub, S. Rudaz, J.-L. Veuthey, and

J. Schappler, J. Chromatogr. A 1217(51),

8041–8047 (2010).

(24) C. Lamalle, D. Roland, J. Crommen, A.-C.

Servais, and M. Fillet, Electrophoresis 36,

2504–2506 (2015).

(25) K. Degardin, Y. Roggo, and P. Margot,

Forensic Sci. Int. 248, 15–32 (2015).

(26) http://theport.ch/home/the-port-2016#topic-

pier71

Julie Schappler is a pharmacist and

holds a Ph.D. from the University of

Geneva. She is currently a lecturer and

research associate. She heads the

unit of capillary electrophoresis and

sample preparation, which works on

developing methods to improve analysis

performance, while reducing analysis

time and cost.

Serge Rudaz is an associate professor

at the University of Geneva. He is a

research group leader and member of the

management board of the Swiss Centre

for Applied Human Toxicology (SCAHT)

Foundation. He is also President of the

Competence Center in Chemical and

Toxicological Analysis (ccCTA).

Advances in Pharmaceutical Analysis October 201642

Schappler and Rudaz

Page 43: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

TSKgel PROTEIN A-5PW IS SPECIFICALLY DESIGNED FOR THE DETERMINATION OF mAb CONCENTRATION DURING

CELL LINE SELECTION OR UPSTREAM BIOPROCESS OPTIMIZATION AND CONTROL. READ MORE ON BIT.LY/29T9DeP.

RECOMBINANT LIGAND BOUND TO TSKgel MATRIX

MORE THAN 2000 INJECTIONS PER COLUMN

mAb QUANTITATION FROM 0.05 TO 200 MICROGRAM

LESS THAN 2 MIN RUNTIMES ALLOW HIGH THROUGHPUT

FAST AND ACCURATE ANTIBODY TITER ANALYSIS WITH TSKgel PROTEIN A-5PW

Page 44: Advances in Pharmaceutical Analysisimages2.advanstar.com/PixelMags/lcgc-eu/pdf/2016-10-sp.pdf · 2018-08-28 · Absolute Molecular Weights and Sizes with your UHPLC… and our ģDAWN™

www.gerstel.com

Performance- Enhancing Automation

Headspace and Dynamic Headspace (DHS)

SPE, Online SPE and Dispersive SPE

Derivatization and Addition of Standards

Agitate, Heat, Mix

Twister (SBSE), SPME and Thermal Desorption

MAESTRO PrepAhead for enhanced productivity

Efficiently automated Solutions for Pharmaceutical Analysis: Extractables & Leachables (E&L) Residual Solvents (USP<467> / OVI) Fatty Acid Methyl Esters (FAMEs) Genotoxic Impurities (GTI/PGI) Metabolomics Automated Sample Preparation

GERSTEL Solutions for GC/MS and LC/MS –For enhanced performance and highest quality of results.

A

Headspace and Dynamic Headspace (DHS)

SPE, Online SPE andDispersive SPE