You are on page 1of 60

Volume 38 Number 6, 313-372

Volume 38 Number 6 June 2020


www.chromatographyonline.com
LCGC NORTH AMERICA

LC PRACTICAL APPLICATIONS DATA HANDLING


Stability Studies MULTIDIMENSIONAL Controlling Peak
and Testing of Integration to Ensure
Pharmaceuticals SEPARATIONS FOR Data Integrity
BIOTHERAPEUTIC
GC CHARACTERIZATION THEORY &
Troubleshooting FUNDAMENTALS
Reduced Peak Starting Up Your Laboratory
June 2020

Size in GC After a Pandemic Shutdown


MACHEREY-NAGEL
Columns and supplies
Chromatography

Your solutions at www.mn-net.com

www.mn-net.com
www.mn-net.com
www.mn-net.com
What does it take to solve
one of the biggest challenges
in chromatography?

Be one of the first to see


what PREMIER can do for you.

waters.com/PREMIER
316  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

MANUSCRIPTS: For manuscript preparation guidelines, see chromatographyonline. 485F US Highway One South, Digital Production Manager
com/lcgc-author-guidelines, or call The Editor, (732) 596-0276. LCGC welcomes unso- Suite 210 Sabina Advani
licited articles, manuscripts, photographs, illustrations, and other materials but can- Iselin, NJ 08830 SAdvani@mjhlifesciences.com
not be held responsible for their safekeeping or return. Every precaution is taken to (732) 596-0276
Fax: (732) 647-1235 Managing Editor, Special Projects
ensure accuracy, but LCGC cannot accept responsibility for the accuracy of informa- Kaylynn Chiarello-Ebner
tion supplied herein or for any opinion expressed. KEbner@mjhlifesciences.com
PUBLISHING/SALES
SUBSCRIPTIONS: For subscription and circulation information: LCGC, P.O. Box
Senior Vice President,
457, Cranbury, NJ 08512-0457, or email mmhinfo@mmhgroup.com. Delivery of MARKETING/OPERATIONS
Industry Sciences
LCGC outside the United States is 14 days after printing. (LCGC Europe and Michael J. Tessalone Marketing Manager
LCGC Asia Pacific are available free of charge to users and specifiers of chro- MTessalone@mjhlifesciences.com Brianne Pangaro
matographic equipment in Western Europe and Asia and Australia, respectively.) BPangaro@mjhlifesciences.com
Associate Publisher
Edward Fantuzzi C.A.S.T. Data and List Information
CHANGE OF ADDRESS: Send change of address to LCGC, P.O. Box 457, EFantuzzi@mjhlifesciences.com Melissa Stillwell
Cranbury, NJ 08512-0457; alternately, send change via e-mail to MStillwell@mmhgroup.com
fulfill@mjhassoc.com or go to the following URL: Sales Manager
http://mmhpubs.mmhgroup.com/Welcome.aspx?pubid=LCGC Brianne Molnar Reprints
BMolnar@mjhlifesciences.com Alexandra Rockenstein
Allow four to six weeks for change. PUBLICATIONS MAIL AGREEMENT
ARockenstein@mjhlifesciences.com
No. 40612608. Return all undeliverable Canadian addresses Senior Director, Digital Media
to: IMEX Global Solutions, P.O. Box 25542, London, ON, N6C Michael Kushner Audience Development Manager
6B2, CANADA. Canadian GST number: R-124213133RT001. MKushner@mjhlifesciences.com Jessica Stariha
JStariha@mmhgroup.com
C.A.S.T. DATA AND LIST INFORMATION: Contact Melissa Stillwell,
EDITORIAL
tel. (218) 740-6831, e-mail MStillwell@mmhgroup.com.
CORPORATE
REPRINTS: Contact Michael J. Tessalone, Editorial Director
e-mail: MTessalone@mjhlifesciences.com Laura Bush Chairman & Founder
INTERNATIONAL LICENSING: Contact Kim Scaffidi, LBush@mjhlifesciences.com Mike Hennessy Sr
e-mail: kscaffidi@mjhassoc.com
Managing Editor Vice Chairman
John Chasse Jack Lepping
CUSTOMER INQUIRIES: Customer inquiries can be forwarded directly to MJH JChasse@mjhlifesciences.com
Life Sciences, Attn: Subscriptions, 2 Clarke Drive, Suite 100, Cranbury, NJ President & CEO
08512; e-mail: mmhinfo@mmhgroup.com Senior Technical Editor Mike Hennessy Jr
Jerome Workman
JWorkman@mjhlifesciences.com Chief Financial Officer
Neil Glasser, CPA/CFE
ycled P
Rec a Associate Editor
%
Cindy Delonas Executive Vice President,
pe
50

r
10-20%

Global Medical Affairs &


aste

CDelonas@mjhlifesciences.com
Corporate Development
rW

Po
s t C s u me
on Creative Director, Publishing Joe Petroziello
Melissa Feinen
mfeinen@mdmag.com Executive Vice President,
© 2020 MultiMedia Pharma Sciences, LLC. All rights reserved. No part of this publi- Operations
Senior Art Director Tom Tolvé
cation may be reproduced or transmitted in any form or by any means, electronic or
Gwendolyn Salas
mechanical including by photocopy, recording, or information storage and retrieval
gsalas@mjhlifesciences.com Senior Vice President, Content
without permission in writing from the publisher. Authorization to photocopy items
Silas Inman
for internal/educational or personal use, or the internal/educational or personal Graphic Designer
use of specific clients is granted by MultiMedia Pharma Sciences, LLC. for libraries Courtney Soden Senior Vice President, I.T.
and other users registered with the Copyright Clearance Center, 222 Rosewood Dr., csoden@mjhlifesciences.com & Enterprise Systems
Danvers, MA 01923, (978) 750-8400, fax (978) 646-8700, or visit http://www.copyright. John Moricone
com online. For uses beyond those listed above, please direct your written request
to Permission Dept. email: ARockenstein@mjhlifesciences.com Senior Vice President, Audience
CONTENT MARKETING
Generation & Product Fulfillment
MultiMedia Pharma Sciences, LLC. provides certain customer contact data (such as Joy Puzzo
Custom Content Writer
customer’s name, addresses, phone numbers, and e-mail addresses) to third parties
Allissa Marrapodi
who wish to promote relevant products, services, and other opportunities that may Vice President, Human
AMarrapodi@mjhlifesciences.com
be of interest to you. If you do not want MultiMedia Pharma Sciences, LLC. to make Resources & Administration
your contact information available to third parties for marketing purposes, simply Webcast Operations Manager Shari Lundenberg
email mmhinfo@mmhgroup.com and a customer service representative will assist Kristen Moore
you in removing your name from MultiMedia Pharma Sciences, LLC. lists.
Vice President, Business
KMoore@mjhlifesciences.com
Intelligence
LCGC North America does not verify any claims or other information a ­ ppearing in Project Manager Chris Hennessy
any of the advertisements contained in the publication, and cannot take responsi- Vania Oliveira
bility for any losses or other damages incurred by readers in reliance of such content. Executive Creative Director,
VOliveira@mmhgroup.com
Creative Services
To subscribe, email mmhinfo@mmhgroup.com. Jeff Brown
Quantification of Adeno-
Associated Virus Aggregation
The Field-Flow Fractionation Platform

Adeno-associated viruses (AAVs) are increasingly used for gene therapy due
to their versatility and safety. One of the biggest concerns for manufacturing
a uniform AAV suspension is the presence of viral aggregates, which
can create problems with transduction efficiency, biodistribution, and
immunogenicity. These large AAV aggregates are challenging to separate
and characterize by traditional column-based chromatography techniques
such as size exclusion chromatography (SEC).

Asymmetrical Flow Field-Flow Fractionation


with Multi Angle Light Scattering (AF4-
MALS) can separate and size large AAV
aggregates, and discern a difference in
aggregate concentration due to the
stressing protocol. Some or all of the large
aggregates would be filtered out by SEC,
resulting in incorrect determination of the
aggregate content or the false conclusion
that no aggregates are present.

For more details visit www.postnova.com and search for ‘virus’.

NovaFFF Software
MALS DLS UV RI ICP-MS

Contact us for more information: www.postnova.com


318  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

CONTENTS
COLUMNS

320 LC TROUBLESHOOTING
Recovering from a COVID-19 Shutdown:
Tips and Tricks for Starting Up, Part I

Volume 38 Number 6, 313-372


Dwight R. Stoll and Tony Taylor
When you restart liquid chromatography (LC) instrumentation that Volume 38 Number 6 June 2020
www.chromatographyonline.com

was idle during the COVID-19 shutdown, you need to follow a


systematic approach. Otherwise, problems may appear in days or
weeks following startup.

LCGC NORTH AMERICA


325 PERSPECTIVES IN MODERN HPLC
Stability Studies and Testing of Pharmaceuticals:
An Overview
LC PRACTICAL APPLICATIONS DATA HANDLING

Kim Huynh-Ba and Michael W. Dong Stability Studies


and Testing of
Pharmaceuticals
MULTIDIMENSIONAL
SEPARATIONS FOR
Controlling Peak
Integration to Ensure
Data Integrity
BIOTHERAPEUTIC

Determining product shelf life is a regulatory requirement for


GC CHARACTERIZATION THEORY &
Troubleshooting FUNDAMENTALS
Reduced Peak Starting Up Your Laboratory

June 2020
Size in GC After a Pandemic Shutdown

pharmaceuticals and many other regulated consumer products. In this


comprehensive overview of stability studies and testing, we summarize COVER IMAGERY BY
current regulatory requirements, share industry practices for forced KENTOH -
degradation, and explain approaches for reduced testing and data stock.adobe.com
evaluation to expedite stability study timelines.

338 FOCUS ON BIOPHARMACEUTICAL ANALYSIS


Multidimensional Separation Techniques for
Characterization of Biotherapeutics
Anurag S. Rathore, Ramesh Kumar, and Ira S. Krull DEPARTMENTS
Multidimensional separations, in which two or more separation 355 The Application
methods are coupled, are a valuable analytical tool for higher Notebook
peak capacity and improved selectivity for the analysis of complex
samples like biotherapeutics.

371 FUNDAMENTALS
Troubleshooting Gas Chromatography:
Reduced Peak Size (Loss of Sensitivity)
Tony Taylor
There are many potential causes of reduced peak size in gas
chromatography (GC), and an inexperienced GC user may not
know where to begin the troubleshooting process. Here, we
review potential causes for reduced peak size in GC systems.

FEATURE ARTICLE

346 Are You Controlling Peak Integration to Ensure Data Integrity?


R.D. McDowall
Chromatography data systems (CDS) have been at the center of
multiple FDA 483 citations and warning letters, with an emphasis
on peak integration and interpretation of chromatograms. Here,
we review the issues associated with ensuring compliance when
performing peak integration.

LCGC North America (ISSN 1527-5949 print) (ISSN 1939-1889 digital) is published monthly by MultiMedia Healthcare, LLC., 2 Clarke Drive, Suite 100, Cranbury, NJ 08512, and is
distributed free of charge to users and specifiers of chromatographic equipment in the United States and Canada. LCGC is available on a paid subscription basis to nonquali-
fied readers in the United States and its possessions at the rate of: 1 year (13 issues), $83.95; 2 years (26 issues), $151.20; in Canada and Mexico: 1 year (13 issues), $107.10; 2 years
(26 issues), $168.53; in all other countries: 1 year (13 issues), $157.50; 2 years (26 issues), $281.40. Periodicals postage paid at Trenton, NJ 08650 and additional mailing offices.
POSTMASTER: Please send address changes to LCGC, P.O. Box 457, Cranbury NJ 08512-0457. PUBLICATIONS MAIL AGREEMENT NO. 40612608, Return Undeliverable Canadian
Addresses to: IMEX Global Solutions, P. O. Box 25542, London, ON N6C 6B2, CANADA Canadian GST number: R-124213133RT001. Printed in the USA.
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  319

Editorial Advisory Board


• Kevin D. Altria – GlaxoSmithKline, Ware, United Kingdom • Ronald E. Majors – Analytical consultant, West Chester, Pennsylvania
• Jared L. Anderson – Iowa State University, Ames, Iowa • Debby Mangelings – Department of Analytical Chemistry and
• Daniel W. Armstrong – University of Texas, Arlington, Texas Pharmaceutical Technology, Vrije Universiteit Brussel, Brussels, Belgium
• David S. Bell – Restek, Bellefonte, Pennsylvania • R.D. McDowall – McDowall Consulting, Bromley, United Kingdom
• Dennis D. Blevins – Agilent Technologies, Wilmington, Delaware • Michael D. McGinley – Phenomenex, Inc., Torrance, California
• Zachary S. Breitbach – AbbVie Inc., North Chicago, Illinois • Victoria A. McGuffin – Department of Chemistry,
• Ken Broeckhoven – Department of Chemical Engineering, Michigan State University, East Lansing, Michigan
Vrije Universiteit Brussel, Brussels, Belgium • Mary Ellen McNally – FMC Agricultural Solutions, Newark, Delaware
• Deirdre Cabooter – Department of Pharmaceutical and Pharmacological • Imre Molnár – Molnar Research Institute, Berlin, Germany
Sciences, KU Leuven (University of Leuven), Belgium • Glenn I. Ouchi – Brego Research, San Jose, California
• Peter Carr – Department of Chemistry, • Colin Poole – Department of Chemistry,
University of Minnesota, Minneapolis, Minnesota Wayne State University, Detroit, Michigan
• Jean-Pierre Chervet – Antec Scientific, Zoeterwoude, The Netherlands • Douglas E. Raynie – Department of Chemistry and Biochemistry,
• André de Villiers – Stellenbosch University, Stellenbosch, South Africa South Dakota State University, Brookings, South Dakota
• John W. Dolan – LC Resources, McMinnville, Oregon • Fred E. Regnier – Department of Chemistry, Purdue University,
• Michael W. Dong – MWD Consulting, Norwalk, Connecticut West Lafayette, Indiana
• Anthony F. Fell – School of Pharmacy, • Koen Sandra – Research Institute for Chromatography, Kortrijk, Belgium
University of Bradford, Bradford, United Kingdom • Pat Sandra – Research Institute for Chromatography, Kortrijk, Belgium
• Francesco Gasparrini – Dipartimento di Studi di Chimica e Tecnologia delle • Peter Schoenmakers – Department of Chemical Engineering,
Sostanze Biologicamente Attive, Università “La Sapienza,” Rome, Italy University of Amsterdam, Amsterdam, The Netherlands
• Joseph L. Glajch – JLG AP Consulting, Cambridge, Massachusetts • Kevin Schug – University of Texas, Arlington, Texas
• Davy Guillarme – University of Geneva, University of Lausanne, Geneva, Switzerland • Dwight Stoll – Gustavus Adolphus College, St. Peter, Minnesota
• Richard Hartwick – PharmAssist Analytical Laboratory, Inc., • Michael E. Swartz – Stealth Biotherapeutics, Newton, Massachusetts
South New Berlin, New York • Caroline West – University of Orléans, France
• Milton T.W. Hearn – Center for Bioprocess Technology, • Thomas Wheat – Chromatographic Consulting, LLC, Hopedale,
Monash University, Clayton, Victoria, Australia Massachusetts
• Emily Hilder – University of South Australia, Adelaide, Australia
• John V. Hinshaw – Serveron Corporation, Beaverton, Oregon CONSULTING EDITORS:
• Kiyokatsu Jinno – School of Materials Science, Jason Anspach – Phenomenex, Inc.; David Henderson – Trinity College;
Toyohashi University of Technology, Toyohashi, Japan Tom Jupille – LC Resources; Sam Margolis – The National Institute of
• Ira S. Krull – Professor Emeritus, Department of Chemistry and Standards and Technology; Joy R. Miksic – Bioanalytical Solutions LLC
Chemical Biology, Northeastern University, Boston, Massachusetts

WinGPC UniChrom
GPC/SEC, IPC and 2D of
(Bio)Polymers and Proteins
Macromolecular Chromatography
Data System
Ensure Compliance and
Data Integrity Data Evaluation PERFECT
Instrument control for all major SEPARATION SOLUTIONS
LCs and specialty detectors MultiWorkstation
Request a Live Demo at your Desk:
Data Evaluation, Client/Server info@pss-polymer.com
or MultiWorkstation Client/Server www.pss-polymer.com
320  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

LC TROUBLE SHOOTING

Recovering from a COVID-19 Shutdown:


Tips and Tricks for Starting Up, Part I
COVID-19-related laboratory shutdowns are sure to cause a myriad of problems with liquid chromatography (LC) instrumentation
across the globe. Taking a systematic approach to restarting these systems will save time and money in the long run, by
preventing problems that may otherwise appear in days or weeks following startup.

Dwight R. Stoll and Tony Taylor

I n March of this year, many organizations took


unprecedented steps to halt the spread of
COVID-19, including severely restricting work
As we discussed the topics to address
in this installment, we decided to focus
on the challenges that are likely to
is, solvent bottles, lines, pump, and flow
path upstream from the column) is free
of contamination and blockages.
in laboratories, or even shutting down entire appear most often. When the pandemic A final general cautionary note here
laboratories, buildings, and work sites for has passed, and we all have the chance is that great care should be taken to
weeks at a time. While some of these shut- to discuss problems we encountered avoid precipitation of salts inside the
downs were planned days in advance and and strange observations we made, LC system. Aside from evaporation of
executed well, I have heard many stories from we’re sure that the list will be very long, water from buffer solutions, precipita-
scientists indicating that the shutdowns were and highly diverse. In the following sec- tion of buffer salts can also easily occur
sudden, and did not allow time to properly tions we’ve tackled specific aspects of when a buffer solution comes into con-
prepare their analytical instrumentation to LC systems and their operation, and tact with a pure organic solvent. This
be idle for weeks or months at a time. Unfor- tried to anticipate the most common is because most commonly used buf-
tunately, this means that these scientists are shutdown-related problems and their fer salts (although inorganic salts, such
going to encounter many challenges when solutions. In next month’s installment, as potassium phosphate, are generally
they return to the laboratory, which will neces- we’ll address other important topics, worse than organic salts such as ammo-
sarily include a lot of troubleshooting to figure including column cleaning and health nium acetate) are highly insoluble in
out why their systems are not working prop- assessment, overall LC system health, organic solvents, such as methanol and
erly before they can return to their normal and making the decision to start collect- acetonitrile (1). So, one should avoid—
experience of producing high quality data. ing data again. at all costs—a situation where a LC sys-
For this month’s installment of “LC Trouble- We strongly encourage you to take tem component containing an aqueous
shooting,” I’ve decided to commit the entire care and use a systematic approach to buffer is flushed immediately with a
column to providing some tips and tricks that bringing your LC system back on-line. pure organic solvent. Such a change-
should be useful as scientists go about the A little effort at this point with preven- over should involve flushing the buf-
Icon Image: Joe Zugcic / Zugcic Photographers, Inc.

process of restarting their laboratories. I’ve tative measures can save a lot of time fer first with water, and then the
invited Tony Taylor to join me in sharing some and money later on when you are ready organic solvent.
of the advice and procedures he has devel- to start collecting useful data again.
oped over the years. Although we’ve never Remember that the column is the heart Solvents and Buffers
worked through pandemic-related laboratory of any LC system, and arguably the most Figure 1 shows a picture of a buffer bot-
shutdowns and startups, some of our expe- sensitive to potential shutdown-related tle I observed in an LC laboratory about
riences with troubleshooting liquid chroma- problems. Be sure to disconnect the a year ago. The solution was dilute (1
tography (LC) problems during normal times column from the flow path before doing mM) phosphoric acid in water. Even
should prove very useful under these strange anything else with the system, and only though this is fairly acidic (pH ~2.7),
circumstances as well. reconnect it to the flow path after you there was a very healthy microbial com-
Dwight Stoll are sure that the rest of the system (that munity growing inside (affectionately
Bio Fermented Alcohol Detection
Utilizing Isocratic Cation Exchange
HPLC with Hamilton’s HC-75 Column

Concern over detrimental greenhouse gases produced from the and lead to less carbon monoxide side products.2 Production of ethanol
combustion of fossil fuels has been growing for the past few decades. from biological sources produce side products, specifically propanol
Identification of new and more diverse methods of energy use has been and methanol amongst other ketones, fatty acids, and esters. Due to
investigated around the world. One such avenue is combustible alcohols azeotropic formation of ethanol with water it is difficult to distill pure
produced from biomasses like sugar beet or sugar cane. To reduce 200 proof ethanol products. However, HPLC analysis provides a good
dependence on fossil fuels, a concerted effort has been made to develop platform for ethanol determination after biomass fermentation.3 To aid
processes where biomass can be fermented biologically by bacteria or the detection of ethanol purity, Hamilton has developed a method utilizing
yeast to produce combustible alcohols. Combustible alcohols are desired the HC-75 (H+ Form) HPLC column, which provides good separation
over other technologies due to the convenience of switching from petrol of the desired analytes. With ~8% crosslinking of the PS-DVB backbone
based fuels. Ethanol is already added to most gasolines to increase the resulting porous gel stationary phase matrix allows for enhanced
octane efficiency. With anywhere from 5–25% ethanol being added to interactions between stationary phase and analyte. Detection was
petrol products in most of the world and up to 100% ethanol fuel in Brazil.1 accomplished with refractive index, but other methods of detection
Additionally, the combustion of these alcohols tend to be more efficient can be applied (i.e. UV, or mass spectrometry).

Column Information
Packing Material HC-75 (H + Form), 9 µm 3

P/N 79544

Chromatographic Conditions
2
Gradient Isocratic

Temperature 80°C
1
Injection Volume 10 µL

Detection Refractive Index

Dimensions 305 x 7.8 mm 0 5 10 15 20 25


Time (minutes)
Eluent A 0.55 mM Succinic Acid
Compounds: 1: 2-Propanol 2: Ethanol 3: Methanol
Flow Rate 0.6 mL/min

1) E. Gnansounou, A. Dauriat, Ethanol fuel from biomass: A review. J. Sci. & Indust. Res.
(64) 2005, 809–21.
2) S. Onuki, J. A. Koziel, W. S. Jenks, L. Cai, D. Grewell, J.H. van Leeuwen, Taking ethanol
quality beyond fuel grade: A review. J. inst. Brew. (122) 2016, 588–98.
3) S.Y. Lee, R. Sankaran, K.W. Chew, et al. Waste to bioenergy: a review on the recent
conversion technologies. BMC Energy. 1:4, 2019.
©2020 Hamilton Company. All rights reserved.
All other trademarks are owned and/or registered by Hamilton Company in the U.S. and/or other countries.
Author: Adam L. Moore, PhD, Hamilton Company Lit. No. L80105 — 05/2020

Web: www.hamiltoncompany.com Hamilton Americas & Pacific Rim Hamilton Europe, Asia & Africa
Hamilton Company Inc. Hamilton Central Europe S.R.L.
USA: 800-648-5950 4970 Energy Way
Reno, Nevada 89502 USA
str. Hamilton no. 2-4
307210 Giarmata, Romania
Europe: +40-356-635-055 Tel: +1-775-858-3000 Tel: +40-356-635-055
Fax: +1-775-856-7259 Fax: +40-356-635-060
To find a representative in your area, please visit hamiltoncompany.com/contacts. sales@hamiltoncompany.com contact.lab.ro@hamilton-ce.com
322  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

to simply not use that solvent bottle any case, if the solvent inlet filter is
again for that instrument, because if porous—whether sintered glass or stain-
thoroughly cleaning the bottle requires less steel—just throw it away, and replace
detergent or other cleaning agents, the it with a new one. These porous filters are
potential for problems associated with perfect environments to support micro-
those cleaning agents is greater than bial growth, and getting them truly clean
the benefit of having a clean bottle to after they have been contaminated is
work with. For example, some deter- extremely difficult. Inlet filters that are
gents can be difficult to remove from based on screens or mesh are more eas-
glassware, to the point where they are ily cleaned. Remove the filter from the
not detectable by MS, and thus what solvent line, remove the visible debris
seems like a simple bottle cleaning step with a brush, and then soak in detergent,
can turn into a background contaminant followed by several rinses with water,
peak in the MS that is detectable for and finally organic solvent. Sonicating
months. When using MS detection, we sintered glass filters should be avoided,
feel the best solution to a badly con- but sonicating plastic or metal ones is
taminated solvent bottle is a brand new okay. As with the bottle, rinse the filter a
FIGURE 1: Image of an LC solvent bottle solvent bottle. In other words, just get few times with the solvent you intend to
with visible (cloudy) microbial growth. rid of the contaminated bottle without use it with before putting it in the clean
bothering to try to clean it. bottle. For the solvent line itself, it is best
known as “floaties”). The good news On the other hand, if the contami- to remove the line from the pump and
in this particular case is that the scien- nated bottle is on an instrument using attach a syringe (10 mL or so will do) so
tists were able to recover their pumping a less sensitive detector (for example, that you can pull large quantities of rins-
system without any long-term effects; ultraviolet-visible [UV-vis] spectros- ing solvent through the line. For older
however, the column they were using copy or an evaporative light scatter- systems that have degasser modules
at the time suffered a performance loss, ing detector [ELSD]), then the bottle separate from the pump itself, discon-
and was never quite the same after- usually can be cleaned without major nect the solvent line at the pump so that
ward. We expect that this experience impact on the rest of the system. After you can pull cleaning solvents through
is going to be repeated in hundreds of discarding the contaminated solvent, the degasser also. Pulling detergent
laboratories as a result of the COVID- wash thoroughly with a detergent solution through the degasser should
related laboratory shutdowns. Many of designed for use with glassware, rinse be avoided, because this may persist in
the aqueous buffer solutions used in four times with HPLC-grade water, and the tubing for a long time. We find that
LC (and even high-performance liquid finally with HPLC- grade organic sol- using warm water (up to 60 °C) is quite
chromatography [HPLC] grade water, vent. We find that working through a helpful for flushing the internal tubing
if given enough time and exposure to series of organic solvents of increasing of the degasser. For newer pumps with
laboratory dust) are environments quite dipolarity—such as cyclohexane, aceto- integrated degassers, it should be suf-
favorable to microbes, particularly those nitrile, and methanol (in that order)—is ficient to disconnect the solvent line at
in the middle of the pH range. Simply effective for removing contaminants of the inlet to the degasser on the pump
stated, many scientists are going to varying degrees of dipolarity. Before unit. Using the same steps as with the
return to laboratories where they will be use again on the instrument, be sure to bottle and filter, pull each liquid through
able to see visible microbial growth in rinse with whatever solvent you plan to the solvent line as a means of rinsing the
the solvent bottles of their LCs. put in the bottle. inside of the line. For older systems with
So…what to do? Now, if you come back to your labo- degassers that have large internal vol-
Occasionally we have problems in our ratory and find microbes in your solvent umes, something on the order of 50 mL
own laboratories with solvent bottles bottles, then the solvent line feeding the of each liquid will be enough to give the
that become contaminated somehow, pump is also contaminated, in addition solvent line and degasser a good flush.
whether resulting from a bad lot of sol- to the solvent bottle itself. Before put- With newer systems where you only need
vent from a vendor, or an instrument ting this solvent line back into the solvent to flush the line external to the pump; 10
operator pouring something into the bottle you have just cleaned, consider mL or so should be good enough.
bottle that does not belong in there. what exactly you will do with the solvent
Whenever this happens, our approach line. Again, if this is an LC–MS instrument, Dirty and Dried-Out Pumps
depends on whether or not the instru- you should seriously consider replacing Before attempting to move any liquid
ment is using mass spectrometric (MS) the entire solvent line and solvent inlet through the pump itself, it is best to dis-
detection. If it does, our preference is filter that goes in the solvent bottle. In connect the high pressure outlet capil-
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  323

lary from the pump and connect a waste valve. These can also suffer from partial care to be sure that, if the rest of the sys-
line running to a waste container to col- or total obstruction, leading to inconsis- tem contains a buffer solution, the first
lect whatever goes through the pump tent flow (if on the inlet side), or higher solvent pumped from the clean pump
until you are confident that the pump is than expected pressure (if on the outlet to the rest of the system is water, so that
delivering clean, debris-free solvent. If it side). If you know you are dealing with precipitation of salts can be avoided.
is obvious that one or more solvent lines a situation where it is likely these filters
has been contaminated with microbes, or screens have been fouled due to pro- Component Blockages Due
it will be best to flush the pump thor- longed inactivity of the pump, microbial to Precipitation and Debris
oughly with clean solvent before con- growth, or other debris, it will be a good Some users will return to their labora-
necting the pump to the rest of the idea to simply replace these filters or tories to find that there is a significant
system. Our approach to this step is to screens at the beginning of your startup obstruction somewhere in their LC sys-
put all of the solvent lines associated process. If you want to try cleaning them tem that is preventing flow at reason-
with the pump (typically two for a binary before replacing, remove and sonicate able pressures. This obstruction could
pump or four for a quaternary pump) in warm water for 10 min, followed by result from debris associated with con-
into a single bottle of HPLC-grade IPA for 10 minutes. If problems persist taminated solvents, pump or valve seal
water, and run 30 mL of water through that are attributed to these parts, just material if salt precipitation occurs in
each channel of the pump. Next, put all replace them entirely. those components, or precipitation of
of the solvent lines into a single bottle If the pump has dried out entirely salts inside of some other components
of isopropyl alcohol (IPA), and run 30 mL because it was left on with an empty such as a connecting capillary. The first
of it through each channel. Finally, move solvent bottle, it can be difficult to step in resolving such obstructions is
each solvent line back to its respective remove air bubbles from pump heads to clean the pump as discussed above,
solvent bottle (for example, one in water, during startup. Air bubbles can also and make sure it will pump water with
one in buffer, and one in acetonitrile), accumulate in pump heads if the sys- a waste tube connected directly to
and flush each channel with 30 mL of the tem is not used for an extended period the high pressure outlet of the pump.
solvent intended for use in the method. of time. In our experience, the best way Then, systematically add one connec-
It is important to recognize that there to resolvate a completely dry pump or tion or component at a time, working
are some filters in modern LC systems remove air bubbles from the pump your way from the pump outlet toward
that might be easy to overlook. It is heads is to purge thoroughly with IPA, the detector. In most systems, the next
always helpful to consult the technical because it wets most materials used in component after the pump will be the
manual for your LC system to see where LC pumps well. autosampler, with a fairly long connect-
these might be. For example, some Once the pump has been thoroughly ing capillary between them. Disconnect
pumps have porous frits or screens on purged with clean solvent and it is deliv- this capillary from the sampler, con-
the upstream side of the inlet check ering the expected flow from each chan- nect it to the pump, and see if you can
valve. Other pumps have porous frits on nel at low pressure, it can be connected pump water through this capillary at
the downstream side of the outlet check to the rest of the system. Again, take reasonable pressure; what pressure is

Unbeatable accuracy
again and again
The precise and time-pressured nature of your work
mean you need products you can rely on, every time.
That’s why every product across the comprehensive
Supelco® portfolio is meticulously tested against the
tightest specifications to ensure the highest industry
standards and compliance with applicable regulations.
So for unbeatable accuracy every time, choose
Supelco® analytical products.

For more information, please visit:


SigmaAldrich.com/SuccessReplicated

MilliporeSigma, the vibrant M and Supelco are trademarks of Merck KGaA, Darmstadt,
Germany or its affiliates. All other trademarks are the property of their respective owners.
Detailed information on trademarks is available via publicly accessible resources.
© 2020 Merck KGaA, Darmstadt, Germany and/or its affiliates. All Rights Reserved.
The Life Science Business of Merck KGaA, Darmstadt, Germany operates as
MilliporeSigma in the US and Canada.
324  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

reasonable depends on the dimen- mission of light through the flow cell.
THEIRS IS sions of the capillary. A benchmark to Most manufacturers recommend flush-
FAMILIAR. remember is that the pressure drop ing the flow cell with warm water (up

OURS, across a 300 mm length of 120-µm


i.d. (0.005 in.) capillary when pumping
to 60 °C), followed by IPA. If excessive
noise or poor sensitivity are observed

SUPERIOR. water at room temperature and 1 mL/


min is about 10 bar. If you double the
after cleaning, the flow cell windows
may have to be replaced.
length of this capillary, the expected
pressure will double. If you double Summary
the diameter, the expected pressure Given the variety of ways different labo-
drops by a factor of four. In our experi- ratories were shut down early on in the
ence, it is best to simply replace the global COVID-19 outbreak, it is likely
capillary if the pressure is more than that LC users will encounter a wide vari-
two times higher than the expected ety of problems with their instruments
pressure after 5 min of flushing with when they return to their laboratories
water. Once you have verified that this and resume work with their instruments.
capillary is okay, either by flushing or In this installment of “LC Troubleshoot-
replacing it, then the next likely com- ing,” we have highlighted some of the
ponent is the autosampler itself. Here problems we expect users to encounter
there are multiple sources of poten- most frequently, and made suggestions
tial obstruction, including the sampler for resolving these problems in a system-
needle and needle seat capillary in the atic and efficient way. Taking the time to
case of a flow-through needle design, thoroughly clean and examine your LC
or a sample loop capillary if using a system when you return to the labora-
fixed volume injection design. If the tory will pay dividends later by avoiding
pressure increases dramatically when costly interruptions to normal work.
the sampler is reconnected, then it is
likely that one of these components is References
obstructed. Occasionally, this can be (1) A.P. Schellinger and P.W. Carr, LCGC
resolved by removing the capillary and North Amer. 22(6), 544–548 (2004).

sonicating in warm water, but the easy


fix is to simply replace a capillary that ABOUT THE COLUMN EDITOR
you know is obstructed based on the
Dwight R. Stoll is the edi-
pressure it adds to the system when tor of “LC Troubleshooting.”
it is in the flow path. You should keep Stoll is a professor and the
adding one component at a time to co-chair of chemistry at Gus-
tavus Adolphus College in St.
the flow path until you discover the
Peter, Minnesota. His primary research
one that is obstructing the flow. focus is on the development of 2D-LC for
Finding the component in a sys- both targeted and untargeted analyses.
tem that is leading to higher-than- He has authored or coauthored more
than 60 peer-reviewed publications and
expected pressures can be tricky
four book chapters in separation science
sometimes, but most of the time the and more than 100 conference presenta-
ION CHROMATOGRAPHY BY
systematic approach described here tions. He is also a member of LCGC’s edi-
will identify the culprit. torial advisory board. Direct correspond-
ence to: LCGCedit@mmhgroup.com
One other component that deserves
special mention here is the flow cell of
optical detectors. Most UV and fluo- ABOUT THE CO-AUTHOR
Get the support your lab deserves. rescence detectors have quartz com- Tony Taylor is the Chief Sci-
Learn more at LeaveTheFamiliar.com
ponents in contact with the mobile ence Officer of Arch Sciences
phase that can be damaged if left in Group and the Technical
contact with highly alkaline solutions Director of CHROMacademy.
Direct correspondence to:
for extended periods, or contami- LCGCedit@mmhgroup.com.
© 2019 Metrohm USA, Inc. Metrohm and design® is a registered trademark of Metrohm Ltd.
nated by microbial growth or other
particles that will reduce the trans-
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  325

PER SPEC TI V E S
IN MO DERN HPLC

Stability Studies and Testing of


Pharmaceuticals: An Overview
This installment is the first of a series of three white papers on stability studies and testing of pharmaceuticals, as well as the
development and validation of stability-indicating high performance liquid chromatography (HPLC) methods. The series is
co-authored by Kim Huynh-Ba, a subject-matter expert on stability testing and regulatory compliance, and Michael Dong,
the columnist on “Perspectives in Modern HPLC.” This first installment provides a comprehensive and updated overview
of stability studies and testing of small molecule drugs, current regulatory requirements, and industry practices for forced
degradation, as well as possible approaches for reduced testing and data evaluation to expedite stability study timelines.
Kim Huynh-Ba and Michael W. Dong

D etermining product shelf life is a reg-


ulatory requirement for pharmaceu-
ticals and many other regulated consumer
best practices, and regulatory expecta-
tions of stability programs.
The stability profile, a critical quality attri-
Stability Studies in
New Drug Development
During drug discovery, the medicinal
products. The shelf life of medicines is set bute (CQA) of a pharmaceutical entity, is chemist focuses on synthesizing new
following stringent regulations; therefore, based primarily on the physicochemical chemical entities (NCEs) that interact selec-
efficient application of stability science is properties of the drug substance (DS) and tively with the molecular targets (such as
critical. The shelf life (expiration dating or drug product (DP). The DS is the active receptors or enzymes), leading to potential
expiry) is displayed on labels of pharma- pharmaceutical ingredient (API), with small disease mitigation (1). The structural motifs
ceutical products to ensure the integrity, amounts of impurities and degradation of a leading NCE candidate are then opti-
quality, and potency of the product when products. A DP typically contains a single DS; mized for biological activities and safety by
used within that time period. Shelf life is when the DP formulation has more than one the synthesis of a series of analogs. The
established using data that are generated DS, it is referred to as a combination product. optimization tools include in vitro and in
to verify the label claim, and approved For example, many over-the-counter (OTC) vivo animal studies, (such as biochemical
by the regulatory agencies. An expiration products contain multiple active ingredients. target binding, biomarkers, or animal mod-
date is required by regional laws to ensure For small-molecule orally delivered DPs, els), bioavailability (pharmacokinetics), and
the safety, efficacy, and quality of the drug such as tablets or capsules, a typical expira- toxicological evaluations (1,2). The desired
products, and that these criteria are main- tion period is two to five years under room NCEs need to be reasonably stable to be
tained throughout the labeled shelf life of temperature storage. Table I summarizes the viable drug candidates, which are scaled
the pharmaceutical product. characteristics of stability studies and testing. up for clinical trials using simple formula-
Most companies have established stan- Each is discussed further in this paper. tions. Eventually, the final DS is formulated
Icon Image: Joe Zugcic / Zugcic Photographers, Inc.

dard operating procedures (SOPs) to sup- In this paper, we use solid oral dos- into a commercial drug product, and sub-
plement regulatory guidelines to provide age forms, such as tablets or capsules, as mitted for regulatory approval.
more specific details, to ensure that stabil- examples. Four types of testing for orally In most pharmaceutical companies, the
ity studies are appropriate for their specific available products, listed in Table II, are nomination of an NCE to the status of a
product types. Nevertheless, stability pro- discussed. Biological products are not drug development candidate triggers the
grams and their practices can vary widely, covered here. formation of a multidisciplinary technical
particularly between large and small Other dosage forms such as parenterals development team. This team is responsi-
pharmaceutical companies, often due to also have similar types of tests, including ble for taking the drug candidate into clini-
the depth of knowledge and available identity, chemical testing (assay and impu- cal trials in humans, and eventually to drug
resources. The primary aim of this paper is rities), physical testing (pH, clarity, particu- approval, production, and commercializa-
to increase understanding of the science, lates), and sterility tests as appropriate. tion. The team is responsible for activities
326  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

referred to in regulatory documents under


TABLE I: Summary of characteristics of stability studies and testing
the heading of “Chemistry, Manufacturing,
Stability Study and Control” (CMC), and thus the team is
Stability Testing Characteristics
Characteristics
often called the CMC team.
Stability studies must be • Drug regulations require expiration dates to be posted on Stability studies for clinical trial materi-
conducted to meet the labels for all pharmaceutical products.
regulatory expectations • Stability data of DS and DP are submitted to regulatory agencies. als (CTMs) are conducted to monitor their
CQAs and help identify which formulation
• Formal stability studies are used to justify storage conditions
and the expiration dating of a packaged product. will result in a successful candidate for reg-
Stability studies fall • Other studies are secondary or those used to support the ulatory submission. In the United States,
into different types primary stability data set.
an expiration date is not required on the
• Stress studies include photostability studies and studies that sup-
port temperature excursions throughout the supply chain. label of a CTM, but this is required in many
• Stability studies are conducted at several environmental condi- countries, such as those in Europe. Forced
tions based on the desired labeled storage conditions. degradation studies (also referred to as
• For products to be stored at room temperature, studies are stress studies) are used to challenge the
Studies are conducted conducted at room temperature and accelerated
under multiple storage conditions. Intermediate condition can be studied if samples stability-indicating power of the analytical
conditions at accelerated method. The accelerated studies, which
conditions may not meet room temperature specifications. require storage under higher tempera-
• Refrigerated or freezing conditions are used for
products that are not chemically stable at room tempera- ture and humidity compared to normal
ture (such as biologics). room-temperature conditions, allow the
• Attributes that change over time are required to be samples to degrade at a faster rate. Data
assessed in a stability study. from accelerated studies may be extrapo-
• The chemical, physical, microbiological, and lated to project the results of a longer-term,
Stability-indicating
performance characteristics of DP must meet
analytical procedures
acceptance criteria throughout the shelf life.
controlled-room-temperature study. Most
must be followed pharmaceutical products exhibit a linear
• Forced degradation studies are performed to challenge the
stability-indicating power of the method and to degradation trend. Based on the stability
evaluate the major degradative pathways of the API. data, a retest period is assigned to the DS,
• Stability testing consumes a significant part of internal and an expiration date to the DP.
Stability studies are
analytical testing and outsourcing resources of
resource intensive The CMC team typically consists of scien-
pharmaceutical companies.
tists from various functions, such as process
chemistry (synthetic organic chemistry), for-
mulation (pharmaceutics), analytical chem-
TABLE II: Four categories of stability tests for tablet or capsule drug products istry (analytical development and quality
Category Typical Tests control [QC]), pharmacokinetics and drug
metabolism (PKDM), outsourcing, regu-
The primary stability-indicating assay is typically an HPLC-UV
Chemical testing method for potency and chemical impurities. Others are tests latory affairs, supply chain, and project
for chiral impurities (HPLC-UV) and moisture (Karl Fisher) management. The degree of involvement
Tests for appearance, color, solid-state characterization (crystallinity, depends on the development phase. For
Physical testing
polymorph by X-ray power diffraction [XRPD]), thermal methods) example, the analytical development chem-
Performance testing
Dissolution (or disintegration) tests to measure the ist is more involved in the discovery and
rate of release of the solid dosage form. early-phases of development to support DS
Microbial limit testing Tests indicated in USP General Chapters <61> and <62> and DP processes. In contrast, quality con-
trol chemists and regulatory staff are more
active in the later clinical phases for CTM
TABLE III: Storage conditions established by the ICH Q1A (R2) guideline for zone I/II. manufacturing and regulatory filings (3,4).
Some of the team activities include:
Label
Storage Conditions Storage Conditions to be Studied • conducting the necessary synthetic
Conditions
process scale-up of the DS (process
Long-term storage 25 ± 2 °C and 60 ± 5% RH
Controlled chemistry staff)
room Intermediate condition 30 ± 2 °C and 65 ± 5% RH
• performing detailed characterization of
temperature
Accelerated condition 40 ± 2 °C and 75 ± 5% RH the API, developing and validating ana-
Refrigerated Refrigerated condition 5 ± 3 °C lytical procedures for DS and DP, and
Freezer Freezing condition –20 ± 5 °C identification of critical impurities and
degradation products (analytical devel-
opment staff)
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  327

• finding the optimum solid-state form the quality of CTMs. This method is typi- Good Manufacturing Practices [GMP]) sup-
(salt, crystallinity) or polymorph of the cally a “composite” analytical procedure porting the establishment of expiration
API (analytical staff) that determines both the potency of the dates for regulatory filings (5,6). Details on
• conducting stability studies to support API and chemical impurities of the DS. Ide- the development and validation of the sta-
clinical development and registration ally, it can also be used for the assessment bility-indicating methods will be covered
(analytical and QC staff) of DP samples. Once a feasible analytical in the second and third installments of the
• designing and developing processes for procedure is established, forced degra- series. Forced degradation and stability
formulations of CTMs and the final com- dation studies are performed to evaluate studies are discussed in later sections here.
mercial DP (pharmaceutics staff) the specificity of the analytical procedure. A better understanding of stability
• setting acceptance criteria for CQAs This method is used in the release testing studies and their regulatory requirements
(specifications) to monitor clinical quality of the early clinical batches and testing of is essential for the analytical chemist for
and establishing commercial specifica- samples from initial stability studies to sup- several reasons (6,7). First, these stud-
tions (QA and regulatory staff) port clinical trials (3–5). ies require many different tests (shown
• manufacturing CTMs (CMC team and out- The stability-indicating assay and in Table II) on multiple batches stored at
sourcing and project management staff) impurities method is typically a gradient different storage conditions and pulled at
• assembling the CMC submission pack- reversed-phase HPLC method with UV numerous time intervals. Second, the pro-
age for regulatory filings (CMC team detection that can separate the API and cess development or formulation scientists
and regulatory staff). all the known impurities and degradation may request many experimental batches
In this series of white papers on stability products (5). This method is validated to of DS or formulations to be placed on sta-
testing, we focus on the tasks performed ensure adequate analytical performance bility, leading to redundant testing, bring-
by the analytical development and QC (specificity, linearity at 100% target concen- ing little added value if not managed with
scientists within the CMC team. The first tration and expected impurities concentra- a science- and risk-based approach. There-
task for the analytical chemist assigned to tions, accuracy, precision, sensitivity, and fore, a better understanding of the design,
the CMC team is to develop a reasonable solution stability) for “formal stability stud- intent, and best practices of stability stud-
stability-indicating method for monitoring ies” (defined as studies conducted under ies and a proper interpretation of associ-

Changing the ART of analytical chromatography with μPACTM capLC columns:

Unrivalled separation power Micromachined reproducibility Flow rate flexibility Plug-and-play connectivity
1 - 15 µl/min

Designed for increased robustness and throughput


without losing sensitivity.

Discover our products on


www.pharmafluidics.com
328  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

dissolution (for solid dosage forms), mois-


TABLE IV: An example of stress conditions for drug substance
ture, and additional physical character-
Factor Being izations such as X-ray powder diffraction
Conditions Time
Studied
(XRPD) tests for crystallinity and polymorph
a) Open dish
Q1B exposure levels form. This set of tests (customized for the
Light b) In the immediate container
(2x to 10x) specific DS or DP, or compendial tests) is
c) In the secondary container
Heat (solution) a) 60 °C Up to 24 h documented in the product specifications.
They must be used to assess the identity,
Heat (solid) a) 50 °C, 60 °C, 70 °C, 80 °C Up to 24 h
physical form (color, size, and crystallin-
Accelerated a) Open dish 40 °C/75% RH or greater
stability b) In the immediate container
Up to 7 d ity), purity (chemical and chiral purity) (3,4),
water content, performance (dissolution),
a) 3% peroxide at RT* a) 30 min to 2 h
Peroxide and microbial activity (4,14,15), to ensure
b) 3% peroxide at <40 °C b) 30 min to 2 d
that drug quality is maintained throughout
a) 0.1–1 N HCl (at RT) a) 30 min to 2 h
Acid (solution) the stability studies.
b) 0.1–1 N HCl (at 60 oC) b) 30 min to 2 d
a) 0.1–1 N NaOH (at RT) a) 30 min to 2 h
In a stability study, representative
Base (solution) batches of pharmaceuticals are stored in
b) 0.1–1 N NaOH (at 60 °C) b) 30 min to 2 d
* RT = room temperature controlled stability chambers at different
temperatures and relative humidity (RH)
ated regulations can lead to more efficient cedures, establishment of acceptance cri- levels in proposed packaging (containers,
implementation of stability programs. teria, forced degradation studies, DS and closure systems) (6,7). These storage con-
DP method validation, and stress tests (3,4, ditions represent long-term, accelerated,
Objectives of this Article 12). In early phases, stability studies may or stress storage conditions. Samples are
In this installment, we discuss: be conducted under limited accelerated periodically removed from these chambers
• regulations and regulatory expectations conditions, and often without a fully vali- according to a pre-approved schedule,
of stability programs dated suite of analytical methods. CTMs and tested to verify that the pharmaceu-
• the goals of forced degradation stud- are released against a set of acceptance tical products meet specifications within
ies and examples of forced-degradation criteria (such as specifications) for clinical the established expiration date under the
protocols use by the Quality Assurance (QA) group. recommended storage and packaging
• a strategy for conducting stability stud- Once a product proceeds to Phase III conditions. Formal stability testing to sup-
ies to expedite regulatory filings development and preparation for product port regulatory filings must be conducted
• modern predictive stability software registration, GMP regulations must be fol- with validated methods according to GMP
tools and statistical approaches to expe- lowed for finished pharmaceutical products regulations (13,16). These stability data
dite stability studies in a science- and starting from the registration batches (13). are also used to support the manufactur-
risk-based approach. The registration batches and commer- ing, registration, shipping, and transporta-
cial products destined to be distributed tion of commercial products. Furthermore,
Regulations and Regulatory for patients’ use must undergo stringent stability data are collected through clinical
Expectations of Stability Programs release testing by QC to meet the estab- formulation phases, process development,
Stability is a Critical Quality lished commercial specifications for identity, proposed packaging selection to establish
Attribute for Pharmaceuticals purity, potency, and quality of the finished the drug product’s shelf life, storage condi-
In the Quality by Design (QbD) approach, product. While QC performs release test- tions, and commercial specifications.
CQAs describe the properties or attri- ing of the registration batches and issues Post-marketing studies are conducted
butes pertinent to product quality (8–11). a Certificate of Analysis (CoA), it is officially to verify that any changes to raw material,
For pharmaceutical products, CQAs are released and signed-off by the QA group manufacturing steps, or container or closure
characteristics that impact the safety and (or by a “Qualified Person” [QP] in Euro- systems have not affected the DP through
efficacy of drug products. Acceptable pean Union pharmaceutical regulation) for its expiration date based on product com-
limits or product specifications must be market distribution after receiving approval. mitment. Any planned change of raw mate-
established and verified by release test- A portion of the registration batches is gen- rial, manufacturing, or packaging must be
ing (11). Stability studies of DS and DP are erally used for “formal (registration) stabil- evaluated to determine if additional stability
conducted throughout the drug develop- ity studies,” and upon commercialization, studies or regulatory filings are needed.
ment process, from the preclinical stage to stability samples are evaluated annually in The United States Food and Drug Admin-
final product approval, with the study size marketed product stability programs. istration (FDA) has determined that the sta-
dependent on the phase of development. Stability testing of CTMs typically bility profile is a critical attribute of a pharma-
The initial analytical development activities includes tests for appearance, identifica- ceutical product. Therefore, stability studies
include the development of analytical pro- tion and quantification (assay), impurities, are necessary to support any submission
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  329

such as an Investigational New Drug (IND), ment process, according to ICH Q1A (R2) Forced degradation studies are per-
New Drug Application (NDA), or Abbrevi- and ICH Q2 (R1) (8,16). Although regula- formed to investigate the major degrada-
ated New Drug Application (ANDA) filing (1). tions mandate that these studies must be tive pathways of the DS and DP and support
The stability data packages within the CMC performed, the guidance does not provide the initial development of the DS stability-
section are typically the most extensive non- directions on procedures or conditions to indicating methods (3,5,12). High-stress
clinical sections in these submissions. They use. The actual protocols employed appear conditions are employed to subject DS and
include tabulated data, graphs, narratives to to vary widely in different organizations. An DP to conditions more severe than acceler-
summarize the stability profile of the pack- overview of the chemistry fundamentals of ated conditions to serve several purposes:
aged product to justify specifications, and drug forced degradation studies and best • to provide insight into the degradation
proposed expiry for the product. practices can be found in books, articles, pathways of DS and DP and their mech-
and other resources (17–20). anisms, facilitating the development
Harmonization of Regulations: ICH
Quality Guidelines for Stability Studies

YOUR LAB WILL FETCH MORE CASH


Given that stability testing is expensive and
labor-intensive, it is imperative to understand
the regulatory requirements and their intent
to avoid unnecessary studies. In addition,
regulations vary significantly from country to
country, which impacts the cost of new drug
development and the registration timeline
for a global product launch. Therefore, in
the early 1990s, the International Council for
Harmonisation of Technical Requirements
for Pharmaceuticals for Human Use (ICH),
consisting of representatives from regulatory
agencies and scientists from pharmaceutical
industries, was formed. The council’s role is
to harmonize the quality requirements for
pharmaceuticals to minimize redundant test-
ing, and reduce the time and cost of new
product development. The ICH stability
guideline, ICH Q1, was the first quality guide-
line established by the council, indicating the
significant need for harmonization of global
stability requirements.
The ICH initially harmonized three geo-
graphic regions: the United States, the
European Union, and Japan, which encom-
passed Zone 1 and Zone 2 of the World
WITH A PROTON ONSITE GENERATOR
Health Organization (WHO) climatic zones.
• H2, N2 and Zero Air • Proven Safe
However, many countries outside Zone 1
and 2 joined the ICH, or voluntarily adopted • Consistent Purity • Cost Effective
these guidelines later with various modifica- • Consistent Pressure • Eliminates Cylinder
tions. The current stability guideline (Q1A Storage and Delivery
[R2]) harmonized the storage conditions, Issues
frequency of required testing, and the mini-
mum amount of data needed for registra-
tion (6–8). Table III lists the stability storage
conditions established by ICH Q1A (R2) (8).

Forced Degradation Studies


Forced degradation studies, or stress test-
ing, are required by regulations and scientific
necessity during the initial method develop-
+1.203.949.8697
www.ProtonOnSite.com
330  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

TABLE V: An elaborate forced degradation protocol used in an automated laboratory Thermal Degradation
The effect of temperature is studied by expos-
Stressing Condition Temperature (°C) Stressing Time (days)
ing the drug substance to high temperatures
Water 70 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7 in increments of 10 oC from 50 to 80 oC. Ther-
0.1 N HCl 70 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7 mal degradation is the primary degradation
0.1 N NaOH 70 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7 pathway of the DS in the solid state.
0.3% H2O2 Ambient 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7, 10, 14
Solution State Acid/Base Degradation
Cool White Fluores-
Ambient 0, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 5, 7, 10, 14 Solution-state degradation mechanisms may
cent Light (solution)
Cool White Fluores- be different, and should also be explored.
Ambient 0, 1, 2, 3, 5, 7, 9, 10, 11, 12, 13, 14
cent Light (solid) If the drug substance is not soluble under
Suntest—UV and aqueous conditions, then a small amount
Ambient 8 h (1xICH), 16 h (2xICH)
Visible Light (solid) (up to 10%) of organic solvent (acetonitrile
PH 2 Buffer 70 0, 1, 3, 5, 7 or methanol) can be used to solubilize the
PH 4 Buffer 70 0, 1, 3, 5, 7 DS, and the acid or base can then be added
PH 6 Buffer 70 0, 1, 3, 5, 7 to stress the samples. Attention should
be given to the functional groups pres-
PH 8 Buffer 70 0, 1, 3, 5, 7
ent in the drug molecule when selecting
PH 10 Buffer 70 0, 1, 3, 5, 7
a co-solvent (20).
Week 1: 60 It is not advisable to use a high concen-
In solid form at
Week 2: 70 0, 1, 3, 5, 7, 8, 10, 12, 14, 15, 17, 19, 21
ambient humidity
Week 3: 80
tration of acid or base, because doing so is
neither practical nor necessary. A few papers
Week 1: 60
In solid form at 75%RH Week 2: 70 0, 1, 3, 5, 7, 8, 10, 12, 14, 15, 17, 19, 21 have suggested a series of extensive steps
Week 3: 80 of pH changing from pH 2 to pH 10 buffer. It
may not be necessary as a routine practice,
unless for a specific DS or as part of a stress
Design of Experiments (DoE) to establish a
TABLE VI: Typical forced degradation studies for drug product of a solid-dosage form
stability model (21).
Forced Degradation Study Suggested Conditions
Heat Expose drug products at 50 ºC for up to 1 month Photostability
Expose drug products to 40 ºC and Photostability studies are essential, and
Humidity
75% RH and 25 ºC and 90% RH must be performed to generate primary
Photostability
Expose drug product in a petri dish without light-induced degradation products by
cover at twice or three times the ICH Q1B exposure level subjecting one representative batch of the
DS to the light exposure listed in ICH Q1B.
The standard light exposure is 1.2 million
of stable formulations and selection of in both solid and solution states. Table IV lux hours of visible light and 200 lux hours
suitable packaging lists an example set of stress conditions of near UV, with storage conditions con-
• to obtain samples to verify method for DS. An important goal of forced deg- trolled at room temperature.
specificity and structure elucidation of radation studies is to generate a potential These studies can be repeated when
significant degradation products level of degradation products that may there is a change of DS samples, such as a
• to allow the differentiation of impurities form during manufacturing and on stabil- new synthetic route, with different crystal-
and degradation products from the DS, ity storage. Typically, forced degradation linity form, different supplier, or a change
excipients, and other interference studies are conducted until a 5–20% loss of DP formulation.
• to facilitate the rapid establishment of of API is observed (18,19). This range is set During the method development phase,
CQAs for selection or elimination of to produce a reasonable amount of deg- forced degradation samples are typically
selected testing. radation products to facilitate method evaluated using a preliminary mass spec-
• to rapidly identify any excipient incom- development and to avoid secondary trometry (MS) compatible HPLC method
patibility issues with the DS degradation products from an unstable with a photodiode array detector (PDA)
• to generate potential data to support degradant (which are not observed under to collect information on peak area under
the justification of specifications. actual stability conditions). It should be the curve. The MS and PDA data are used
Forced degradation studies are con- noted that the conditions should be for peak tracking, peak purity assessment,
ducted under high temperature, humidity, selected based on the physicochemical identification of degradation products,
acid/base, oxidative, and light conditions properties of the individual DS and DP. impurities, and interferences (3,5,19).
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  331

Table V shows an example of a more extensive, elaborate forced these chambers be controlled within ±2 oC and ±5% RH for
degradation protocol used by a pharmaceutical laboratory to col- chambers operated at 25 °C/60% RH, 30 °C/65% RH, and 40
lect data for most of their NCEs using a four-temperature thermal °C/75% RH. The refrigerated condition is controlled at 5 oC ± 3
stressing condition matrix (ambient, 60 oC, 70 oC, 80 oC) for acid, oC (2 to 8 oC) with ambient humidity (monitored but not con-
base, oxidative, light, humidity, buffered solution, and prolonged trolled). These chambers are equipped with sensors for the
stressed duration (up to 21 days). This sample-intensive protocol continuous monitoring of temperature and relative humidity.
was made feasible by this centralized and automated laboratory These data are captured electronically or by chart recorders
specializing in supporting forced degradation studies for the with backup power. Excursions will be noted with an alarm
development site. The laboratory utilized automated sample stor- system. When the chambers are out of tolerance (see above),
age and retrieval system called “Powderium” (a powder dispens- investigations must be performed to identify the cause of the
ing workstation), robotics, programmable liquid handling, and issue and determine the impact of the stored samples (7).
refrigerated storage of quenched samples (18,19). The stability chambers are fully qualified and maintained, and
Table VI provides a summary of forced degradation studies per- their access is limited to authorized personnel to ensure that sam-
formed on solid dosage forms. Note that acid, base, and oxidative ple removal is tracked and controlled. Maintenance and service
exposures are not used, because these conditions are not repre- records must be available for inspection during audits. Downtime
sentative of realistic storage or exposure situations. The length of must be limited, and a backup plan must be developed to avoid
exposure should be evaluated carefully, depending on the dosage issues that can affect sample storage and data integrity. An annual
forms or the formulation. For example, capsule formulations or chamber inventory program is also necessary to track the sample
film-coated tablets may not be able to withstand a temperature storage and disposition at any time.
higher than 60 oC for an extended length of time.
Similar to the DS, a goal of 5% to 20% loss of active ingredient Stability Study Strategies
is recommended. If DS or DP is stable and the desired degrada- to Expedite “First-in-Human” Clinical Trials
tion level cannot be reached under high-stress conditions, stud- Minimizing “Time to Market” is an essential strategy for most
ies should be terminated. Overstressing a sample may lead to pharmaceutical companies (1). Given that by regulatory agencies
secondary degradation products that are not present in formal expect to see three months of stability data in IND filings to enable
stability studies. In contrast, understressing may lead to insufficient
degradation products for method development or identification
(12). For each condition of the forced degradation study, the chro-
matogram is compared with that of a control sample to document
the mass balance of the API (% loss of API) and % degradation
(normalized peak area %). For all conditions, the peak shapes
and peak purity (by PDA and MS) of all components should be
assessed for coelution or presence of interferences (3–5).
Accelerated and stress studies are performed to induce quicker
degradation than what would be observed in recommended stor-
age conditions. Results from these studies are used to estimate the
stability profile of the DS and DP at long term storage conditions,
establish the actual degradation pathways, and demonstrate the
intrinsic stability of the DS. It is recommended that these studies
be implemented under GMP conditions because the results of
these studies may be used as supporting information in regulatory
document packages. Full shelf-life studies are still needed to verify
the retest date of the DS and expiration date of the DP.
While discussions on forced degradation studies for NCEs in
NDA applications are generally plentiful, those for ANDA are often
insufficient and may result in regulatory deficiencies. For instance,
the impurity profiles may not be completely derived or sufficiently
discussed (20). In addition, the labeling for generic drug products
should be concordant with that of the reference listed drug (RLD)
and with a compendial monograph, as applicable (20).

Stability Storage
Stability samples are kept in a controlled temperature in humid-
ity stability chambers. The ICH Q1A (R2) guideline requires that
332  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

Most tools are science- and risk-based,


TABLE VII: Typical stability studies to support early-phase clinical trials
and have a robust statistical underpin-
Intended Storage Conditions to Be Studied Time Points (in Months) ning that conforms to the principles of
25 ºC and 60% RH Time point zero, 1, 2, 3, 6, 9, QbD and DoE, which are endorsed by
Room temperature or 30 ºC and 65% RH and 12, or end of the study regulatory agencies for pharmaceutical
40 ºC and 75% RH 1, 2, 3, 6 development (9).
Time point zero, 1, 2, 3, 6, 9,
2–8 ºC
Refrigerated and 12, or end of study Modern Predictive
25 ºC and 60% RH 1, 2, 3, 6 Methods and Approaches
One widely used approach is the Acceler-
ated Stability Assessment Program (ASAP),
TABLE VIII: Example of a simple bracketing design which uses a modified Arrhenius approxi-
mation. This approach uses degradation
Strength 50 mg 100 mg 250 mg 500 mg
kinetics at a few stressed conditions in 1–2
Batch A B C A B C A B C A B C
weeks, and extrapolates to lower tempera-
50 mL T T T - - - - - - T T T tures and humidity for longer timeframes.
Container
100 mL - - - - - - - - - - - - The advantages of this strategy are that
size
250 mL T T T - - - - - - T T T the length of these experiments is short
and the experiments use fewer resources.
The accuracy of this model assumes a first-
order reaction, which is often the case for
TABLE IX: Examples of factors to consider in bracketing
hydrolysis-based degradation (6,22). The
disadvantage is that the presumed kinetic
1. Container size model limits the degradation to less than
2. Container wall thickness
3. The surface area of the volume
a percent and requires extrapolation of
4. The volume of the product three factors simultaneously: time, temper-
5. The weight of the product ature, and humidity. These disadvantages
6. The permeability rate of water per dosage unit
7. The strength of a similar drug/excipient drug ratio
are minimized when the DS and DP have a
simple kinetic pathway, and the primary sta-
bility drivers are temperature and humidity
(23). Another drawback could occur when
Phase I clinical trials, it is customary to per- stability protocols that are listed for different the kinetic route is complex or nonlinear
form preliminary accelerated stability stud- dosage forms at different drug develop- or involves physical changes, which may
ies on the Good Laboratory Practices (GLP) ment phases are discussed elsewhere (6,7). result in the linear model not being accu-
toxicological DS and prototype DP batches Results of the formal stability studies used to rate. Recently, this approach was also used
to provide supporting data for quicker IND determine the drug products’ expiration are to select appropriate packaging based on
submissions. Early-phase stability studies are included in the stability section of the NDA its thermal and moisture-barrier character-
typically short, as shown in Table VII, and the or ANDA. istics, and to optimize the drug:excipient
study conditions depend on where the clini- ratio of the formulation based on the
cal trials will take place. During Phase I, the Predictive Tools to moisture content (22). This approach has
analytical procedures are usually not fully vali- Establish Product Expiry gained popularity, mainly to set expiry for
dated, though specificity, accuracy, linearity, Stability data are used to establish the clinical materials, and has been accepted
and precision studies should be completed. expiry of finished products according to by multiple regulatory agencies (24).
These time-saving strategies allow the phar- US FDA Good Manufacturing Practices Another successful tool is the Predic-
maceutical companies to start Phase I clinical regulation 21 CFR 211.137 (21). The data tive Characterization Study (PCS), a rapid
trials as soon as possible after the nomina- from the accelerated condition are used to development of robust stability models
tion of the drug candidates. determine a tentative expiration date, but using a semi-empirical design space. It is
Characterization studies of the API and DP testing at full shelf life is necessary to con- a systematic study that evaluates the prod-
continue during formulation development. It firm the approved expiry. uct data of samples exposed to a series
is recommended that the forced degrada- In this section, we discuss examples of storage conditions to build predictive
tion studies be repeated before the release of useful software tools and statistical stability models to establish the design
testing and the initiation of the formal stabil- approaches to evaluate stability data and space and control strategy based on the
ity studies under GMP regulations and ICH to expedite stability studies for establish- QbD concept, such as setting specifica-
Q1A (R2) guideline (8). Examples of standard ing product expiry. tions and selecting proposed container–
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  333

closure systems. This approach is also found acceptable by many size) encompass and represent the stability behaviors of products of
regulatory agencies (23–25). the intermediate levels, eliminating the need for testing at the inter-
Improved modeling tools have enabled stability predictions mediate levels. A bracketing schedule can be applied to multiple
with accelerated timeframes compared to those of the traditional strengths of identical or closely related formulations.
extrapolation approach. Besides providing a more accurate way Table VIII shows an example in which the 100 mL container
to justify an expiry assignment, these models are used to increase size is bracketed by the 50 mL and 250 mL container sizes; thus,
the understanding of the critical factors that influence the quality stability samples stored in the 100 mL containers are not tested.
of the DS and DP, as demonstrated in empirical science and risk- Similarly, the 100 mg and 250 mg tablet strengths are bracketed
based approaches detailed in ICH Q8–Q11. These approaches by the 50 mg and 500 mg strengths. Assuming that the stability
are often utilized by large pharmaceutical companies with bet- profiles of these different strength formulations are the same
ter modeling resources and expertise. A regulatory template and the compositions of these tablets are similar, regardless of
has been shared to standardize on critical elements to use risk- strength, then the testing of 100 mg and 250 mg tablets may
based predictive stability data for setting up shelf life to support not be needed. ICH Q1A (R2) requires that three batches (A, B,
clinical development. (26) C) be made available for submission. According to the guide-
lines, a pharmaceutical company could reduce its testing from
Trending Analysis potentially 36 configurations to only 12 configurations, resulting
The purpose of the stability program is to establish a retest period in significant savings of resources and time (6,7).
for DS or shelf life for DP that will apply to all future commercial Table IX lists some factors to which a bracketing approach can
batches. Stability data of CQAs of individual batches should potentially apply, assuming that the container–closure system
remain within specification throughout the assigned retest period and the headspace of the containers do not have any impact.
of the DS or shelf life of DP. Therefore, the trending of all stability The disadvantage of the bracketing concept is that when one of
data is very important. Most degradation trends of API are linear; the results is out-of-specification; all bracketed configurations
however, the degradation pathway could be a linear, quadratic, or data could then be at risk. Full testing would need to be acti-
cubic function. The typical ICH Q1E approach for the determina- vated mid-study, complicating the data set, and negating the
tion of shelf life through the analysis of data based on a quantita- benefit of any testing reduction. In addition, all bracketed fac-
tive attribute that is expected to change over time is to determine
the period of time at which the 95% one-sided (or two-sided) con-
fidence limit for the mean curve intersects the acceptance crite-
rion (27,28). The annual commitment lots should also be evaluated
against the product trend to assure the consistency of the stability
profile of the DP and verify if there is any unintended change that
may impact the DP.

Reduced Testing with


Bracketing and Matrixing
Conducting stability programs is expensive and time consuming.
To reduce costs and increase the efficiency of the stability pro-
gram while maintaining regulatory compliance, many companies
employ bracketing or matrixing options that reduce the amount
of testing and resources required (6,7). ICH Q1A (R2) and ICH
Q1D are guidelines to refer to when reduced testing is applied.
Although the bracketing and matrixing concepts are included
in multiple global stability guidelines, limited numbers of pub-
lished applications can be found. It is even more challenging to
provide examples for the matrixing for an NCE. Here, we explain
the similarities and differences between these approaches to
illustrate this application.

Bracketing
Bracketing refers to a study design in which only the extreme vari-
ables, such as extreme strengths, container sizes, or container fills,
are tested. The plan assumes that the stability behaviors of prod-
ucts manufactured or packaged at these extreme levels (such as, for
example, highest vs. lowest strength, or largest vs. smallest package
334  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

TABLE X: An example of a two-thirds factorial matrixing design


annual product monitoring stability stud-
ies rather than for DPs from NCEs.
Timepoint Months on stability
Batch 0 3 6 9 12 18 24 36 Matrixing
A T T - T T - T T Matrixing is a statistical design of the sta-
Strength 1 B T T T - T T - T bility schedule in which a selected group
C T - T T T T T T of samples of the total number of pos-
sible samples is tested at a specific time
A T - T T T T T T
point. At the next time point, a different
Strength 2 B T T - T T - T T
group should be tested. At each time
C T T T - T T - T
point, the stability data of that group of
T = for stability testing scheduled for that timepoint samples will represent the stability of the
whole set of studies.
TABLE XI: Examples of factors for which matrixing can be considered Similar to the bracketing approach, this
design also assumes that all the batches
have similar stability profiles; thus, there
1. Different batches is no need to generate all the results. The
2. Different concentrations
3. Different filling long-term trends of all configurations are
4. Different container size approximately linear across the whole set
5. Different closure composition of all studies, and the comparative sta-
6. Different closure system
bility of each presentation can be evalu-
ated with the reduced schedule. Many
options are available with the matrixing
tors (for example strengths/formulations if only a subset will be commercialized. concept, which could reduce the testing
and package configurations) will need to Due to the risk, the bracketing concept schedule by a third, a half, or two thirds,
be registered in all end markets, even is used more often for post-approval or depending on the aggressiveness of the

TABLE XII: Example of a stability data record (6)

STABILITY ANALYTICAL RECORD


Sample Name: Manufacturing Date:
Storage condition:
Lot#: Manufacturing Site:
Sample Orienta-
Study #: Expiration Date:
tion (if applicable):
Protocol #: Testing Site:
Packaging Information:
Study Start Date: Packaging Site:
Packaging Date:
Study Purpose:
Acceptance Time
Test Name Method 1 Mo 2 Mo 3 Mo 6 Mo 9 Mo 12 Mo
Criteria Zero
Pull Date
Test Date
Appearance
Assay
Impurities
Individual
Total
Dissolution
Average
% RSD
Range
Moisture
Completed By: Date:
Reviewed By: Date:
Approved By: Date:
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  335

statistical extrapolations and risk assess- TABLE XIII: Questions about the conclusions of a stability report
ment for the design (6,7).
Table X shows an example of a two-
thirds factorial matrixing design. In this ► What is the stability profile of the product?
► Are all lots within the stability trend of the product?
schedule, there are two strengths and ► Can differences be attributed to a particular factor such as packaging or strength?
three batches made of each strength. ► Do all tests meet specifications?
All presentations are tested at time zero, ► Is there any test closely monitored? Are there any tests to be performed
more often?
12 months, and the end of the study (36 ► Is there any package closely monitored? Is there any package to be tested
months) for the highest possible precision more often?
as these time points are critical to the DP ► Do all tests meet specifications?
► Do the data support the expiration date?
stability profile. For all other time points (3, ► Are there any stability trends?
6, 9, 18, and 24 months), only two-thirds ► Discuss any out-of-specification investigation.
of the presentations get tested at a rotat- ► Discuss any statistical evaluation of the data set.
►  ould the stability database be satisfactory to the proposed or approved expiration
W
ing schedule. Therefore, all configurations dating?
are tested at different time points for the
same amount of times in total.
The matrixing approach requires that
all samples be placed in stability cham- lifecycle state (such as NDA, ANDA, or according to ICH Q1E or statements
bers, including at the timepoints that supplement NDA). The stability data- that no overall trends and little variabil-
are reduced. Therefore, when there is an base comprises laboratory data gen- ity are observed in the dataset. Table
instability issue, the testing schedule can erated and stored electronically or as XIII lists typical questions that should
be changed to test all configurations. If printed reports. These stability data are be addressed in the stability conclusion.
some data points are not available, there entered into stability reports, which are “Poolability” of different batches should
will be a minimum impact on the entire essential to communicate information be evaluated to determine the consis-
study of multiple presentations. internally for audit purposes, product tency of the representative stability data
Table XI lists some examples of factors investigations, justification of specifica- and trends with respect to the manufac-
where matrixing can be used to reduce tions, to support product development, turing process and analytical method,
testing. Matrixing also provides more or simply as a communication tool. All by comparing the intercepts and the
flexibility because different reduced detailed information in the report must slopes of all batches (6,7,24).
testing options can be performed for be verified for accuracy. For an NDA or
various tests depending on the test- ANDA submission, the stability report is Summary and Conclusions
ing confidence. Realistically, matrixing typically the most extensive non-clinical A robust and science-based stability
should be used only for label storage section to summarize the stability data program is required for the registration
conditions. The accelerated stability data of the product during its shelf life (6,28). and commercialization of any pharma-
may provide some insight into the sta- Stability data can be presented in a tab- ceutical product. Regulations in this
bility profile of the product at the label ular format, in a graphical representation, area are well established; however, the
storage conditions. Because matrixing is a narrative, or a combination of formats. application and interpretation of the
a statistical concept, evaluation of data Table XII is an example of a stability data regulations vary between companies
can be more involved, and assessment record. The data tables can be generated and regions. ICH harmonizes the main
of all the presentations as a complete manually in a document or printed out regions to help companies use a stan-
set is more complicated, which explains from a laboratory information manage- dardized approach.
why this approach is not used widely. ment system (LIMS) or stability manage- This article provides a high-level
These reduced testing approaches are ment software. All information and data summary of regulations and regula-
discussed in detail in many global guide- must be reviewed and verified. The con- tory expectations of stability programs.
lines; however, a lack of knowledge of clusion of the stability report discusses It reviews the goals and practices of
matrixing among regulatory reviewers whether the data indicate that the prod- forced degradation studies to generate
and internal quality and regulatory pro- uct continues to meet the quality specifi- sample data to confirm method speci-
fessionals may hinder the use of this cations or acceptance criteria established ficity. A strategy for conducting accel-
application to reduced stability testing. for the study and whether the data sup- erated stability studies on preliminary
port the proposed or approved expiration batches to expedite initial regulatory
Stability Reports and dating period (6,7). filing is described. Finally, predictive
Regulatory Submissions Stability reports should include a sta- stability software-based and statistical
Stability reports are required in all phases tistical evaluation of the currently avail- approaches such as trending, bracket-
of regulatory submissions regardless of able data and a discussion of the results ing, and matrixing are explained. 
336  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

Acknowledgments (12) H. Bruemmer and N. Belikova, Pharm. (26) H. Williams, Pharm Technol. 42(8),
The authors express their gratitude to Technol. 41(9), 46–52 (2017). 42– 47 (2018).

the following colleagues for their time (13) 21 Code of Federal Regulations (CFR), (27) WHO Expert Committee on Specifica-
Part 211.194(a), Current Good Manufac- tions for Pharmaceutical Preparations
and efforts to provide timely reviews of turing Practice for Finished Pharmaceu- Fifty-second report, annex 10, Stabil-
the manuscript to improve content accu- tical Products (US Government Printing ity testing of ac tive pharmaceutical
racy and clarity: Jane Weitzel of Zinata, Office, Washington, DC, 2019). ingredients and finished pharmaceuti-
(14) United States Pharmacopeia Conven- cal products (World Health Organiza-
Jing Capucao of J&J, Mike Shifflet of tion, 2018).
tion, USP 37/NF 32, General Chapter
Johnson and Johnson Consumer Health USP <1225>, Validation of Compendial (28) International Council for Harmonisa-
Care, Adrijana Torbovska of Farmahem, Methods, (United States Pharmacopeial tion of Technical Requirements for
Alice Krumenaker of TW Metals, LLC; Convention, Rockville, Maryland, 2013). Pharmaceuticals for Human Use (ICH)
(15) United States Pharmacopeia Conven- Q1E, Evaluation of Stabilit y Data
Mark Shapiro of MCS Pharma Consult- (Geneva, Switzerland, 2003).
tion, USP 37/NF 32, General Chapter USP
ing, Madhavi Mahavadi of BioMarin <1226>, Verification of Compendial Pro-
Pharmaceuticals, and Joshua Ayers of cedures (United States Pharmacopeial
ASQ Solutions. Convention, Rockville, Maryland, 2013,
revised 2018). ABOUT THE CO-AUTHOR
(16) International Council for Harmonisation
References of Technical Requirements for Pharma-
Kim Huynh-Ba
(1) R.G. Hill and H.P. Rang, Eds., Drug Dis- ceuticals for Human Use (ICH) Q2 (R1), is the managing director
covery and Development: Technology in Validation of Analytical Procedures: of Pharmalytik LLC. (www.
Transition (Churchill Livingston, Elsevier, Methodology (Geneva, Switzerland, pharmaly tik.com), which
Edinburgh, Scotland, 2nd Ed., 2012) November 1996, updated 2015). provides consulting ser-
(17) S.W. Baertschi, K.M. Alsante and R.A.
vices in Stability Sciences, Quality
(2) M.W. Dong, Drug Development Process,
Short Course presented at Pittcon, Phila- Reed, Eds., Pharmaceutical Stress Management Systems, and Analytical
delphia, Pennsylvania, March 2019. Testing: Predicting Drug Degradation Development. She is an Adjunct Pro-
(CRC Press, Boca Raton, Florida, 2nd fessor at Temple University’s School
(3) M.W. Dong, LCGC North Am. 33(11),
Ed., 2011). of Pharmacy and Illinois Institute of
764–775 (2015).
(18) H.T. Rasmussen, W. Li, D. Redlich, M.I.
Technology (IIT). Kim is a member of
(4) D. Kou, L. Wigman, P. Yehl, and M.W. the US Pharmacopeia’s Council of
Dong, LCGC North Am. 33(12), 900–909, Jimidar, HPLC Method Development, In
Handbook of HPLC in Pharmaceutical Experts, and the chair of the Chemical
(2015). Medicines Monograph IV Expert Com-
Analysis, S. Ahuja and M. W. Dong, Eds.,
(5) M.W. Dong, HPLC and UHPLC for Practic- (Elsevier, Amsterdam, 2005), Chapter 6. mittee, USP Good Documentation
ing Scientists (John Wiley & Sons, Hobo- Practices Expert Panel, USP Organic
ken, New Jersey, 2nd Ed., 2019), Chap- (19) M.W. Dong and H.T. Rasmussen, HPLC
Method Development Short Course, Impurities of Drug Substance and
ters 9–11. Drug Products Expert Panel. She is on
Eastern Analytical Symposium, Somer-
(6) K. Huynh-Ba, Ed., Handbook of Stabil- set, New Jersey, 2004. the editorial board of AAPS Open, the
ity Testing of Pharmaceutical Products: Journal of GXP Compliance, and the
Regulations, Methodologies and Best (20) R. Maheswaran, Pharm. Technol. 36(5),
1–6 (2012).
Journal of Validation Technology. Kim
Practices (Springer, New York, 2009). has authored ~30 technical publica-
(7) K. Huynh-Ba, Ed., Pharmaceutical Stabil- (21) 21 Code of Federal Regulations (CFR), tions and is the editor of two books on
ity Testing to Support Global Markets Part 211.137, Current Good Manufactur-
stability testing to support registration
(Springer, New York, 2010). ing Practice for Finished Pharmaceuti-
in global markets.
cal Products (US Government Printing
(8) International Council for Harmonisation Office, Washington, DC, 2019).
of Technical Requirements for Pharma-
ceuticals for Human Use (ICH) Q1A (R2), (22) K. Waterman, Understanding and Pre- ABOUT THE AUTHOR
Stability Testing of New Pharmaceutical dicting Pharmaceutical product Shelf life,
Products (Geneva, Switzerland, 2003). In Handbook of Stability Testing of Phar- Michael W. Dong
maceutical Products: Regulations, Meth- is a principal of MWD
(9) International Council for Harmonisation odologies and Best Practices (Springer, Consulting, which provides
of Technical Requirements for Pharma- New York, 2009), Chapter 6. training and consulting
ceuticals for Human Use (ICH) Q8(R2), services in HPLC and UHPLC,
(23) K. Huynh-Ba et al., Meeting Report,
Pharmaceutical Development (Geneva, Analytical Approaches to Ensure Prod- method improvements, pharmaceutical
Switzerland, 2009). uct Quality – AAPS Joint Face-to-Face analysis, and drug quality. He was formerly
(10) International Council for Harmonisation Meeting of the Stability, the Pharmaceu- a Senior Scientist at Genentech, Research
of Technical Requirements for Pharma- tical Impurities, and the CMC Statistics Fellow at Purdue Pharma, and Senior
ceuticals for Human Use (ICH), Q3A(R2), Focus Group, AAPS Open 3 (1), 2017. Staff Scientist at Applied Biosystems/
Impurities in New Drug Substance, https://aapsopen.springeropen.com/ PerkinElmer. He holds a PhD in Analytical
Q3B(R2), Impurities in New Drug Prod- articles/10.1186/s41120-017-0011-z Chemistry from City University of New York.
ucts (Geneva, Switzerland, 2006). (24) H. Williams et al., Pharm. Technol. 41(3), He has more than 100 publications and
(11) International Council for Harmonisa- 52–57 (2017). a best-selling book in chromatography.
tion of Technical Requirements for (25) D. Lavrich, Rapid Development of He is an editorial advisory board
Pharmaceuticals for Human Use (ICH) Robust Stability Models Using Semi- member of LCGC North America and
Q6A, Specifications: Test Procedures Empirical Design Space, AAPS Webi- the Chinese American Chromatography
and Acceptance Criteria for New Drug nar, March 24, 2016. https://aapsopen. Association. Direct correspondence to:
Substances and New Drug Products, s pring eropen.com /ar ticles/10.118 6/ LCGCedit@MMHGroup.com
(Geneva, Switzerland, 1999). s41120-017-0018-5#article-info
338  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

FOCUS
ON
BIOPHAR M ACEU TIC AL
ANALYSIS

Multidimensional Separation Techniques


for Characterization of Biotherapeutics
Multidimensional separations aim to combine two or more separation methods, and these are increasingly being employed to
overcome the limitations of one-dimensional separations. The outcome typically includes higher peak capacity and improved
selectivity. Such an approach can be invaluable when analyzing complex mixtures. In this article, we discuss some commonly used
two-dimensional systems, their pros and cons, and their applications in the biopharmaceutical industry.

Anurag S. Rathore, Ramesh Kumar, and Ira S. Krull

T he quality of a biotherapeutic is
defined by its various biological,
immunological, structural, and physi-
of large proteins may generate more
than 100 peptides. Given the random
distribution of peaks, even a system
Based on how the eluate is trans-
ferred from one dimension to another,
MD separations can be classified as
cochemical attributes. Some of these with a peak capacity of 10,000 would on-line and off-line. The MD system is
are post-translational modifications be able to resolve only 98% of the mix- said to be on-line when the eluate of
(PTMs), other product-related het- ture of 100 tryptic peptides (6). Thus, one dimension is directly transferred
erogeneities, process related impu- in the case of biopharmaceuticals, to the next dimension, and separation
rities, and host cell related impuri- which are mostly large molecules, use occurs in real time in both the dimen-
ties. An array of different techniques of one-dimensional ( 1D) chromatogra- sions. On the other hand, in the case
contributes to the analytical toolbox. phy usually does not yield complete, of off-line systems, eluates from the
These are utilized for characterization resolved product coverage (6,7). first dimension are collected as frac-
of the biotherapeutic product, and Multidimensional (MD) separation tions, and then later subjected to sep-
especially the myriad of other species systems, in which two or more sepa- aration in the next dimension (9,12).
that are also present in the final drug ration methods are coupled, can be Both approaches have some advan-
product (1–3). One of the most com- used in such cases to increase the tages and disadvantages (Table I).
monly used analytical techniques for peak capacity of the system. This will Additionally, MD approaches can
routine process evaluations and qual- then allow the complete resolution, it also be classified as heart-cutting or
ity control in the industry is reversed- is hoped, of all constituent analytes comprehensive ( 1 D× 2 D), depending
phase liquid chromatography-mass (8). These systems improve sample on the part of the 1 D elution con-
Icon Image: (Columns) Joe Zugcic / Zugcic Photographers, Inc.

spectrometry (LC–MS/MS). Due to its resolution manifold by first resolving tent that is transferred in the second
robustness, reproducibility, and easy the components in the first dimension. dimension. In the heart-cutting MD
coupling with the MS, LC–MS has The eluate is then transferred and fur- approach, only a part of the 1 D elu-
become the state-of-the-art technol- ther resolved in the second or subse- ate is subjected to separation in the
ogy for routine characterization of quent dimensions (9). Such a separa- next dimension. However, in a com-
biotherapeutics in the industry (3,4). tion was first reported with the use of prehensive approach, the whole elu-
However, LC-based separations suf- two-dimensional (2D) paper chroma- ate of the 1 D step is subjected to a
fer from a significant limitation aris- tography in 1944 (8). Since then, differ- 2D separation (9).

ing from incomplete separation and ent separation techniques have been Whether on-line or off-line, an MD
co-elution of target analytes (5). One coupled in a two-dimensional or mul- separation system is useful if it fulfills
reason for lack of baseline resolution tidimensional manner, and have been two fundamental requirements, as
is that LC can generate peak capaci- proven useful for the analysis of com- suggested by Giddings (8). First, the
ties up to 1000, and proteolytic digest plex biological samples (9–11). separation techniques in the MD set
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  339

up must be orthogonal. This means TABLE I: Advantages and disadvantages of on-line and off-line coupling
that the resolving principles of the
Parameter On-line Off-line
separation techniques must be based
on different chemical or physical prop- Speed Fast (12) Slow (12)
erties of the analytes. Thus ideally, for Automation Feasible (10) Difficult (10,13)
a given analyte, the peak distribution Mobile phase compositions Limited options (12) More options (12)
in the two dimensions must be uncor- Optimization Difficult (12) Easy (12)
related. The second principle states Sample manipulation
that the resolution of the first dimen- Not allowed (10) Possible (10)
between dimensions
sion must not be lost during separa- Resolution Less (9) More (9)
tion in subsequent dimensions (8,12).

Multidimensional LC–LC Coupling been combined. Such couplings have applied for the characterization of bio-
One of the most successful MD sys- been successfully implemented for the therapeutics, are discussed below. In
tems, widely accepted by both aca- analysis of biopharmaceuticals (11,15). all these methods, reversed-phase LC
demia and industry alike, is that of However, not all LC modes can be has been used for the second-dimen-
2D–LC-based separations (14). These combined with each other, due to sol- sion separation, likely because of the
separations involve separation of com- vent immiscibility and other compat- high-quality reversed-phase LC col-
pounds at two different conditions, ibility issues (16). These 2D-LC systems umns available and their compatibility
using either different modes of LC or have been widely accepted by the bio- with MS (34).
using a single-mode operated at two pharmaceutical industry. Some of the
different conditions. Many LC modes, applications of these techniques in the Reversed-phase LC x
such as ion exchange chromatography industry are summarized in Table II. Reversed-phase LC Coupling
(IEC), size exclusion chromatography Some of the most commonly used, This separation system involves resolv-
(SEC) and reversed-phase LC, have 2D-LC methods, which have been ing the analytes via reversed-phase

ADVANTAGE
See What It Can Do for You and Your Lab

Sign up today to access Restek’s


years of chromatography knowledge at
www.restek.com/advantage

Pure Chromatography
340  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

TABLE II: Biopharmaceutical applications for multidimensional separation techniques

1st Dimension 2nd Dimension Detectors Applications References


HIC RPLC MS Product quality of antibody–drug conjugates (17)
SEC WCX DAD Aggregate analysis and charge variant profiling (18)
(AEX and
RPLC CAD and MS Polysorbate stability in mAb formulations (19)
RPLC)/CEX
IEC/SEC RPLC MS Charge and size variants of mAb (20)
HIC Native SEC MS Drug to antibody ratio of ADCs (21)
HIC RPLC MS Structural isoforms and PTM analysis of ADCs (21)
SEC RPLC MS mAb degradation products (22)
SEC RPLC DAD/MS Impurity and stability analysis of ADCs (23)
CEX RPLC MS mAb isoform and subunit analysis (24)
SCX RPLC DAD and MS Peptide mapping of mAb (6)
HILIC RPLC DAD and MS Peptide mapping of mAb (6)
RPLC RPLC DAD and MS Peptide mapping of mAb (6)
RPLC RPLC MS Host cell protein analysis (25–27)
SEC iCIEF CCD Charge heterogeneity of mAb heavy and light chains (28)
RPLC/IEX CGE PDA Disulfide isomer heterogeneity in mAb (29)
RPLC CZE UV and LIF Peptide mapping of an immunoconjugate (30)
RPLC CZE MS Peptide mapping of mAb (31)
CIEF CZE MS Charge variant analysis of mAb (32)
CZE CZE MS Charge variant analysis of mAb (33)
Abbreviations: ADC: antibody drug conjugate; AEX: anion exchange chromatography; CAD: charged aerosol detector; CCD: charge coupled device; CEX:
cation exchange chromatography; CGE: capillary gel electrophoresis; CIEF: capillary isoelectric focusing; CZE: capillary zone electrophoresis; DAD: diode ar-
ray detector; HIC: hydrophobic interaction chromatography; HILIC: hydrophilic interaction liquid chromatography; iCIEF: imaged capillary isoelectric focusing;
IEC: ion exchange chromatography; LIF: laser induced fluorescence; mAb: monoclonal antibody; MS: mass spectrometer; PDA: photodiode array detector;
PTM: post translational modification; RPLC: reversed-phase liquid chromatography; SEC: size exclusion chromatography; SCX: strong cation exchange chro-
matography; UV: ultraviolet; WCX: weak cation exchange chromatography.

The impact of the use of different


TABLE III: Advantages and limitations of commonly used 2D separation techniques
cell lines and purification strategies
Techniques Advantages Limitations on the composition of HCPs in bio-
SCX-RPLC
Most commonly Low peak capacity, poor recovery pharmaceuticals was analyzed via an
used method (36) of hydrophobic peptides (36)
on-line, comprehensive 2D-LC–MS/
RPLC-RPLC High peak capacity (37) Only partially orthogonal (45) MS analysis. Six PTG1 mAb samples
HILIC-RPLC High orthogonality (36) Peptide precipitation (35) were trypsinized and characterized
SEC-RPLC High orthogonality (37) Low peak capacity (37) using MS with high pH reversed-phase
IEC-RPLC High orthogonality (37) Low peak capacity (37) LC (pH 10) in the first dimension, and
then with low pH reversed-phase LC
High orthogonality, peak Eluate and injection volume
LC-CE (pH 2.5) in the second dimension.
capacity, and resolution; fast (8) disparity, difficult coupling (9)
Abbreviations: SCX: strong cation exchange RPLC: reversed phase liquid chromatography HILIC: hydrophilic in- The 2D-LC system was quite stable,
teraction liquid chromatography SEC: size-exclusion chromatography IEC: ion exchange chromatography LC: liquid with excellent reproducibility of frac-
chromatography CE: capillary electrophoresis.
tionation in the first dimension, and
overall retention times in the second
LC in both dimensions, but at two dif- This difference in peptide retention dimension. A reproducibility of 0.1%
ferent pH conditions. The principle is mechanisms is utilized to constitute in retention time was obtained over
that at high pH (such as pH 10), pep- the two dimensions of this separation 48 h of separation, and the reproduc-
tide retention on the reversed-phase system (35). For instance, researchers ibility of the chromatographic setup
LC column is based on solvophobic have reported an on-line 2D-LC–MS/ was independent of the number of
interactions, while at low pH, both MS approach for a comprehensive fractionations in the first dimension. A
solvophobic and electrostatic inter- analysis of host cell proteins (HCPs) total of 33 HCPs were identified over 5
actions determine peptide retention. in monoclonal antibodies (mAbs) (27). orders of magnitude, with the lowest
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  341

concentration at 16 ppm. To decrease tic peptides of trastuzumab. Although phase LC in the second dimension,
analysis time and improve peak shape, the system offers high orthogonal- as they are eluted with the change in
the second dimension was operated ity, concerns persist due to the pres- the pH or ionic concentration of the
at a high flow rate and high tempera- ence of a high organic content in the mobile phase. The advantage of using
ture, which then led to a 15% loss in mobile phases (35). this system is its ease of use, as well
peak capacity as compared to that as the minimal sample manipulation
obtained with standard LC conditions SEC x Reversed-Phase LC Coupling requirements. Both cation-exchange
(27). Limited orthogonality is, how- Size-exclusion chromatography (SEC) x and anion-exchange chromatography
ever, a major issue of this approach reversed-phase LC separates analytes have been coupled to reversed-phase
(36). Column stability and possible on the basis of size and hydrophobic- LC to separate protein mixtures and
erosion of fused silica tubes at high ity in the first and second dimensions, tryptic digests (3,37). For example,
pH are some of the other factors that respectively. The system is thus highly rapid analysis of charge variants of
must be considered before setting up orthogonal, and it is useful for peptide mAbs without much sample manipu-
this separation system (35). separations. Synthetic polymers and lation was performed, with a generic
biopolymers are some of the major 2D-LC–MS method. Two independent
HILIC x Reversed-Phase LC Coupling application areas of this system (37). gradients were used on a single chro-
One of the best orthogonalities dem- A 2D-LC–MS based, semi-automated matographic system, consisting of
onstrated is exhibited by the hydro- platform was developed for charac- two independent column compart-
philic interaction chromatography terization of impurities in monoclonal ments, with built-in switching valves.
(HILIC) x reversed-phase LC combina- antibodies, with a week-long data Reversed-phase trap cartridges were
tion (36). In the first dimension, HILIC collection time. Breakdown products used in the second dimension for
separates analytes mainly based on were identified in stress induced mAb desalting first dimension fractions
their separation between the mobile samples, using SEC–reversed-phase before MS analysis. These cartridges
phase and water-enriched layer LC–MS/MS analysis. The low molecu- enabled fast conditioning and re-
adsorbed onto the stationary phase. lar weight breakdown products were equilibration of columns, with minimal
In the second dimension, reversed- enriched by fractionation via SEC in backpressure, during the switching of
phase LC separates them on the basis the first dimension, and the fractions columns. The poorly resolved and low
of their hydrophobicity. The use of this were resolved via C8 chromatography abundance peaks of IEC in the first
setup has been recently reported in a in the second dimension. Both on-line dimension, were preconcentrated on
study in which a commercially avail- and off-line mass spectrometry analy- the reversed-phase traps using mul-
able 2D-LC system was used for the ses were performed, using the C8 elu- tiple injections. Using an in-process
bottom-up analysis of trastuzumab. ate. A part of the C8 eluate was thus sample of a mAb, fractions of interest
A novel dual loop interface was used directed to the MS while a major part were collected from IEC, and then sub-
for transferring the samples between was collected as fractions. While on- sequently analyzed via reversed-phase
the two dimensions. The use of high line analysis was used for intact mass LC, combined with an on-line elec-
elution strength and a high volume of analysis, the off-line set up included trospray ionization (ESI)-time-of-flight
injection solvent, however, led to com- analysis under both reducing, as well (TOF-MS) based detection. Chain spe-
plete or partial elution of polar pep- as non-reducing conditions, so as to cific information about mAbs was also
tides in the void volume. Several pre- elucidate impurity structure. Various deduced by a disulfide reduction step.
cautions, such as high flow rate and low anticipated products, as well as unpre- This was incorporated as an optional
temperature of the second dimension, dicted modifications, such as frag- step into the workflow in an on-line
were taken to minimize peptide break- ment antigen-binding (Fab) fragments manner. Contamination, sample loss,
through in reversed-phase LC, but it and hydrolyzed, free light chain, were and degradation were minimized with
could not be completely eliminated. identified. The approach was also pro- the use of an on-line setup. The whole
Unfortunately, the HILIC x reversed- posed to be useful for charge variant system was fully automated to allow
phase LC combination was challeng- and aggregation analysis of mAbs. for high throughput analysis, minimum
ing, and offered limited coverage of sample handling, and rapid analysis.
the separation space as well. The study IEC x Reversed-Phase LC Coupling The system was shown to be useful
compared HILIC x reversed-phase LC In this mode of duality HPLC, ion for charge, sequence, and chemically
with strong cation exchange (SCX) exchange chromatography (IEC) is unstable mAb variant analysis. Minor
x reversed-phase LC and reversed- used in the first dimension, which glycoforms were also identified at the
phase LC x reversed-phase LC, and it separates molecules based on their intact protein level, due to the sepa-
found that the other two techniques charge. Analytes are transferred from ration of charged isoforms in the first
are superior for the separation of tryp- IEC in the first dimension to reversed- dimension by IEC (20).
342  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

in order to hasten the separation


process, often deteriorate the over-
(a) all, analytical performance of the
500
2D setup (27).
mAU (214 nm)

400

300
Multidimensional LC-CE Coupling
Due to the limited orthogonality and
200
other disadvantages associated with
100
LC modes in the second dimension,
0 alternative separation techniques,
20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57
Time (min) having different separation principles,
(b) x10 7 have been attempted to improve the
1
3
2.8 separation further. Capillary electro-
2.6
2.4
phoresis (CE) is an attractive alterna-
Counts (x107)

2.2
2
1.8 tive in this regard, because it separates
1.6
1.4 analytes based on their charge to size
1.2
1
0.8
ratios, complementary to all modes
0.6
0.4 of LC (10,39). Electrophoresis with
0.2

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
chromatography has been described
Acquisition Time (min) as a powerful 2D-separation method
by Giddings (9). The use of CE as the
second dimension in 2D systems can
FIGURE 1: Off-line LC-CE-MS of a protein digest. A separation of a protein digest was performed thus provide high efficiency and rela-
using reversed-phase LC in the first dimension and capillary electrophoresis (CE) in the second tively fast separations as compared to
dimension. (a) Separation profile of the digest with reversed-phase LC in the first dimension. Sol- LC-based methods. However, special
vent A and solvent B were 0.1% formic acid in water and 0.1% formic acid in acetonitrile, respec-
tively. Overlapping peaks at 38–39 min are circled in green. (b) CE–MS profile of the fraction,
coupling techniques are required for
containing the peaks circled in green in panel a, in the second dimension. CE separation was CE based setups, due to the use of
performed in a 56 cm long polyvinyl alcohol coated capillary at 30 kV. Background electrolyte was small volumes and high electric fields
2% acetic acid and detection was performed at 214 nm. (13). The history of coupling chroma-
tography and electrophoresis dates
The coupling of SCX in the first or less correlated selectivity, and a back to 1948, and since then the cou-
dimension and reversed-phase LC in lower than maximum peak capacity pling has been shown to improve the
the second is the most widely used is usually achieved (10,36). Another resolution of various biological and
2D-LC system, especially in bottom- major limitation of the 2D–LC system environmental samples (9).
up proteomics-based studies. Ana- is that the separation processes have Various methods based on LC–
lytes are separated on the basis of longer analysis times. Various meth- CE, CE–LC, and SEC–CE have been
their charge in the first dimension, ods to speed up the LC-based separa- reported in the literature, primarily for
and hydrophobicity in the second tions via altering column length, tem- proteome and peptide level analysis.
dimension (35). This method, however, perature, and particle size have been An in-depth review of the applica-
suffers from a significant limitation reported. However, each of these tions of different modes of CE in MD
in terms of the peak capacity in the methods has its own limitations (37). In separations has been recently pub-
first dimension. The SCX chromato- 2D-LC, for example, to achieve faster lished (13). The ability of CE to resolve
gram is sparsely populated with posi- separation, the second dimension LC LC peaks has been discussed in the
tively charged tryptic peptides, most column is run at high flow rates, which literature (40), and is demonstrated
of which (+2 and +3 charged pep- compromises resolution. The second in Figure 1. In this work, the tryptic
tides) tend to be eluted close to each dimension can also be made faster digest of a protein was resolved using
other. Another major limitation of this by operating the column at high tem- reversed-phase LC in the first dimen-
method is the limited orthogonality of perature, which reduces the viscosity sion, then fractionated, with each
the system (about 50%). Furthermore, of the mobile phase, and allows the fraction subjected to CE–MS analy-
hydrophobic peptides may be eluted user to increase the eluent flow rate, sis in the second dimension. Several
with a poor peak shape, or may be but this may affect the stability of overlapping peaks were observed in
incompletely recovered with SCX (36). the stationary phase and the analyte LC that were efficiently resolved by
The major limitation with these sys- (38). Thus, the use of high tempera- CE in the second dimension (Figure
tems, however, is that they show more ture and other operating parameters, 1). For instance, the peaks from 38
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  343

to 39 min (circled in green in Figure mixture, immunoconjugates of a mAb, was also useful for obtaining a deeper
1a), remained unresolved in reversed- and disulfide isomer heterogeneity of understanding about the conjugation
phase LC, as observed in the corre- IgG2-mAbs (9,10,40–43). For example, process. Although this method was
sponding UV chromatogram. CE–MS a size exclusion chromatography- well suited for analyzing reduced and
of the corresponding fraction showed imaged capillary isoelectric focusing denatured mAbs, and offered several
baseline resolution of the same peaks (SEC-iCIEF) method was proposed for advantages over 2D PAGE, it required
(Figure 1b). the quantitative charge heterogeneity large amounts of protein sample
Although these reports clearly analysis of heavy and light chains of a for analysis (28).
demonstrate the utility of LC–CE in mAb. Over 10 different mAbs, includ-
biotherapeutics, LC–CE suffers from ing both IgG1 and IgG4 subtypes, Four-Dimensional (4D) HPLC-MS
significant limitations in terms of injec- were characterized using this platform. MD setups are not limited to just two-
tion and elution volume disparities, The platform was found to be less dimensional analysis. Use of more
between the LC and CE dimensions. time-consuming, less labor-intensive, than two dimensions is also emerg-
There is also the need to isolate CE more reproducible, and allowed fully ing for the analysis of biotherapeu-
from the other parts of the system, due quantitative analysis of charge variants tics. For instance, recently, an on-line
to its high electric field. Due to these compared to the traditional 2D poly- 4D HPLC-MS approach was reported
reasons, LC–CE has still not gained as acrylamide gel electrophoresis (PAGE) for characterization of mAb variants
much attention as 2D-LC (9). A com- analysis. Results were obtained in a sin- (44). The mAb sample was resolved
parative analysis of the advantages gle day, but only with automated SEC and fractionated using IEC in the first
and disadvantages of the commonly and iCIEF platforms. The method was dimension. These fractions were then
used MD methods is given in Table III. also used for characterization of mAbs transferred to the 2D reversed-phase
Nevertheless, the coupling of LC with under accelerated stability conditions, column, where they were reduced
CE has been described for analysis and it was proposed to be useful for and washed. The reduced mAb chains
of natural compounds, peptide frag- generating simplified electrophero- were then eluted onto the third col-
ments, tryptic peptides of a protein grams for conjugated antibodies. It umn, where they were digested.
344  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

The digested peptides were then detected using LC–MS in development, and data analysis strategies in the future are
the fourth dimension. The whole system was fully automated expected to make it a routine characterization technique for
from fractionation to peptide mapping. Prefractionation by quality control processes in the industry. Various modes are
IEC allowed reduction and digestion of separated charge available for 2D-LC systems, but these have limited orthogo-
variants, resulting in higher signal intensity and sensitivity. nality and peak capacity. Recent years have witnessed the
The coupling of MS/MS allowed for localizing the modifica- growth of CE as a method of choice for MD separation sys-
tions at the amino acid level. The system was highly sensi- tems. Although these are not yet as widely used as 2D LC-
tive, and it was able to detect deamidation present, even at based methods, interest in their deployment is growing rap-
0.5% of the main peak. Five out of six oxidation sites were idly. They represent one of the major developments in the
identified in the study. The system, however, showed lim- industry which should become more obvious in the future.
ited retention of small and hydrophilic peptides. Although
the authors presented IEC in the first dimension, the 4D References
set-up was designed so that SEC could be coupled in place (1) A.J. Reason, A. Weiskopf, and A.S. Rathore, BioPharm Intl. 27(7),
of the IEC for the characterization of size variants. 1–8 (2014).
(2) A.S. Rathore, I.S. Krull, and S. Joshi, LCGC North Am. 36(6),
Conclusions 376–384 (2018).
Biopharmaceuticals are complex molecules, and they (3) A.S. Rathore, I.S. Krull, and S. Joshi, LCGC North Am. 36(11),
814–822 (2018).
demand complex, analytical procedures with maximum
(4) M. Pioch, S.C. Bunz, and C. Neusüß, Electrophoresis 33(11),
resolving power. To meet this need, one-dimensional analyti- 1517–1530 (2012).
cal methods are not sufficient for achieving complete analy- (5) S.K. Grebe and R.J. Singh, Clin. Biochem. Rev. 32, 5–31 (2011).
sis. Multidimensional systems offer an alternative, promis- (6) G. Vanhoenacker, I. Vandenheede, F. David, P. Sandra, and K.
ing approach in this regard. 2D-LC is commonly used as a Sandra, Anal. Bioanal. Chem. 407(1), 355–366 (2015).
multidimensional separation system for biopharmaceutical (7) J.R. Lill, Analytical Characterization of Biotherapeutics (John
analysis, and it has now been accepted by many biophar- Wiley & Sons, Hoboken, New Jersey, 1st Ed., 2017) pp. 1-14.
maceutical companies. Simplified instrumentation, method (8) W. Grochocki, M.J. Markuszewski, and J.P. Quirino, Electropho-
resis 36(1), 135–143 (2015).
(9) L. Ranjbar, J.P. Foley, and M.C. Breadmore, Anal. Chim. Acta
950, 7–31 (2016).
(10) P. Cesla, P. Dinisová, and J. Fischer, Sens. Electroanal. 6, 357–
368 (2011).
(11) B.W.J. Pirok, D.R. Stoll, and P.J. Schoenmakers, Anal. Chem.
91(1), 240–263 (2019).
(12) C.R. Evans and J.W. Jorgenson, Anal. Bioanal. Chem. 378, 1952–
1961 (2004).
(13) F.J. Kohl, L. Sánchez-Hernández, and C. Neusüß, Electrophore-
Over 25 Years of sis 36(1), 144–158 (2015).
(14) X. Wang, S. Buckenmaier, and D. Stoll, J. Appl. Bioanal. 3(5),
Quality HPLC and 120–126 (2017).
(15) H. Wang and S. Hanash, J. Chromatogr. B 787, 11–18 (2003).
SFC Columns (16) P.W. Carr and D.R. Stoll, Two-dimensional Liquid Chromatogra-
phy (Agilent Technologies, 2015).
(17) R.E. Birdsall, H. Shion, F.W. Kotch, A. Xu, T.J. Porter, and W.
Chen, MAbs 7(6), 1036–1044 (2015).
(18) S. Schneider, Online 2D-LC Characterization of Monoclonal
Antibodies with Size Exclusion and Weak Cation Exchange
Chromatography (Agilent Technologies, 2016).
(19) Y. Li, D. Hewitt, Y. K. Lentz, J. A. Ji, T. Y. Zhang, and K. Zhang,
Anal. Chem. 86(10), 5150–5157 (2014).
(20) M. Alvarez, G. Tremintin, J. Wang, M. Eng, Y. Kao, J. Jeong, V.T.
Offering a wide range Ling, and O. V Borisov, Anal. Biochem. 419(1), 17–25 (2011).
of phases and dimensions for (21) J.J. Gilroy and C.M. Eakin, J. Chromatogr. B 1060, 182–189 (2017).
both analytical and prep (22) M.T. Mazur, R.S. Seipert, D. Mahon, Q. Zhou, and T. Liu, AAPS J.
14(3), 530–541 (2012).
(23) Y. Li, C. Gu, J. Gruenhagen, K. Zhang, P. Yehl, N.P. Chetwyn, and
C.D. Medley, J. Chromatogr. A 1393, 81–88 (2015).
www.pci-hplc.com | 609.860.1803
(24) D.R. Stoll, D.C. Harmes, J. Danforth, D. Guillarme, S. Fekete, and
A. Beck, Anal. Chem. 87(16), 8307–8315 (2015).
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  345

(25) K. Sandra, A. Ortiz, and P. Sandra, LCGC (35) F. Xie, R.D. Smith, and Y. Shen, J. Chro- ABOUT THE CO-AUTHOR
Europe 28(10s), 45–48 (2015). matogr. A 1261, 78–90 (2012).
(26) J.H. Thompson, W.K. Chung, M. Zhu, L. (36) M. Gilar, P. Olivova, A.E. Daly, and J.C.
Ramesh Kumar is a grad-
Tie, Y. Lu, N. Aboulaich, R. Strouse, and Gebler, Anal. Chem. 77(19), 6426–6434 uate student at the Depart-
W.D. Mo, Rapid Commun. Mass Spec- (2005). ment of Chemical Engineer-
trom. 28(8), 855–860 (2014). ing at the Indian Institute of
(37) D.R. Stoll, X. Li, X. Wang, P.W. Carr,
Technology in Delhi, India.
(27) C.E. Doneanu, A. Xenopoulos, K. Fad- S.E.G. Porter, and S.C. Rutanb, J. Chro-
gen, J. Murphy, S.J. Skilton, H. Prentice, matogr. A 1168(1–2), 3–43 (2007).
M. Stapels, and W. Chen, MAbs 4(1), (38) M. Kivilompolo, J. Pó, and T. Hyö-
24–44 (2012). tyläinen, LCGC Europe 24(5), 232–243 ABOUT THE COLUMN EDITOR
(28) R.P. Vanam, M.A. Schneider, and M.S. (2011).
Marlow, MAbs 7(6), 1118–1127 (2015). (39) D. Chen, X. Shen, and L. Sun, Analyst Ira S. Krull is a Professor
(29) R.K. Rober t s, H.J. Holovic s, M.R. 142(12), 2118–2127 (2017). Emeritus with the Depart-
Thompson, and C.W. Demarest, Elec- (40) R. Aebersold and H. D. Morrison, J. ment of Chemistry and
trophoresis 31(3), 448–458 (2010). Chromatogr. 516, 79–88 (1990). Chemical Biology at North-
(30) J. Liu, H. Zhao, K.J. Volk, S.E. Klohr, E.H. eastern University in Bos-
(41) S.E. Rudnick, V.J. Hilser, and G.D.
Kerns, and M.S. Lee, J. Chromatogr. A Worosila, J. Chromatogr. A 672(1–2),
ton, Massachusetts, and a member of
735, 357–366 (1996). 219–229 (1994). LCGC’s editorial advisory board.
(31) R. Kumar, R.L. Shah, and A.S. Rathore, (42) H.J. Issaq, K.C. Chan, C. Liu, and Q. Li,
J. Chromatogr. A (2020). doi:10.1016/j. Electrophoresis 22(6), 1133–1135 (2001).
chroma.2020.460954
(43) E. Tamizi and A. Jouyban, Electrophore-
(32) J. Hühner, K. Jooß, and C. Neusüß, Elec- ABOUT THE COLUMN EDITOR
sis 36, 831–858 (2015).
trophoresis 38(6), 914–921 (2017).
(44) C. Gstöttner, D. Klemm, M. Haberger, A. Anurag S. Rathore is a
(33) K. Jooß, J. Hühner, S. Kiessig, B. Moritz, Bathke, H. Wegele, C. Bell, and R. Kopf, professor in the Depart-
and C. Neusüß, Anal. Bioanal. Chem. Anal. Chem. 90(3), 2119–2125 (2018). ment of Chemical Engineer-
409(26), 6057–6067 (2017). (45) C. Song, M. Ye, G. Han, X. Jiang, F. ing at the Indian Institute of
(34) K. Sandra and P. Sandra, Bioanalysis Wang, Z. Yu, R. Chen, and H. Zou, Anal. Technology in Delhi, India.
7(22), 2843–2847 (2015). Chem. 82(1), 53–6 (2010).

HPLC Columns for


Organic Acid Analysis
Over 40 years of experience
providing high quality polymeric HPLC
columns for the analysis of samples
containing carbohydrates and
organic acids.

Benson polymeric TM

bensonpolymeric.com
775.356.5755

LCGC0719 Benson HPH Ad pr2f.indd 1 2019-07-12 3:46 PM

40419080306_BENSON
magenta
cyan
yellow
black POLYMERIC_pgxx_HPH_1-1.pgs 07.23.2019 20:56 ADVANSTAR_PDF/X-1a
346  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

Are You Controlling Peak Integration to Ensure Data Integrity?


Peak integration and interpretation of chromatograms is one of the current areas when inspectors are looking to ensure
the integrity of chromatographic data and results. Are you in control of integration, or are you integrating into compliance?
R D McDowall

C hromatography Data Systems (CDS)


have been at the center of multiple
FDA 483 citations and warning letters since
form to appropriate standards of identity,
strength, quality, and purity (5).”
• Manipulation of a poorly defined analyti-
cal procedure and associated data analy-
sis to obtain passing results
the Able Laboratories fraud case in 2005 This is a relatively simple regulation to • Missing data or unreliable data
(1). Between 2005 and 2015, the inspection understand; everything done in the labo- • Data or information submitted to the
emphasis was mainly on unofficial testing, ratory including peak integration must be application that were potentially unreli-
deletion of data files, time traveling, and no scientifically sound. A similar requirement able or misleading and the relevance of
segregation of duties for users. As compa- for scientific soundness in the laboratory is these data or information
nies have changed their working practices to found in Section 11.12 of EU GMP Part 2 • Unexplained or inappropriate gaps in a
stop these practices, the regulatory focus has (also known as ICH Q7) for active pharma- chromatographic or analytical sequence
moved to peak integration and invalidated ceutical ingredients (6,7). • A pattern of inappropriately disregarding
out of specification (OOS) results. This “Data As chromatography is a comparative test results
Integrity Focus” column will review compliant analytical technique, all injections should be • Inadequate or lack of justification for not
approaches to peak integration, and the next integrated in the same way as much as pos- reporting data/information.
article will discuss OOS investigations. sible. However, this generalization is tested More detail on this revised version of CPG
Compliant peak integration has been of near the limits of quantification and where 7346.832 can be found in a recent “Focus on
interest since 2015, enough so that I have complex mixtures are separated. Quality” column (14).
published three previous articles in LCGC Next, we have: Although there have been many regula-
publications in that time. The first discussed tory guidance documents from the MHRA,
manual intervention and manual integration 21 CFR 211.194(a): “Laboratory records shall WHO, FDA, and PIC/S (15–18), none of these
in 2015 (2), and in the first series of “Data include complete data derived from all tests mention peak integration. The best guid-
Integrity Focus,” Mark Newton and I dis- necessary to assure compliance with estab- ance for peak integration is PDA Technical
cussed peak integration and the sequence lished specifications and standards, includ- Report 80, which has a large section on chro-
of integrating data files in a run (3). Most ing examinations and assays,….. (5).” matographic integration, and the guidance
recently, Heather Longden and I revisited document is good in that it illustrates both
that first 2015 article in 2019, and revised the The requirement for laboratory records acceptable and unacceptable peak integra-
overall approach (4). is also simple to understand, but appar- tion practices (19).
We’ll start our peak integration journey by ently difficult to follow. Complete data ICH M10 section 3.3.6 outlines the cur-
looking at the applicable GMP regulations means “everything,” including poor injec- rent thinking for integration of chromato-
and guidance documents. tions and poor integration and was dis- grams in bioanalysis (20).
cussed in “Data Integrity Focus” last year Chromatogram integration and reintegra-
A Regulatory Refresher (8) and other articles (9–11). tion should be described in a study plan, pro-
It is important to understand the regulatory The EU GMP regulations are not as sim- tocol or standard operating procedure (SOP).
requirements for laboratory controls and ple to interpret, as Chapter 4 refers to raw Any deviation from the procedures
records as these provide a major input to any data which is not defined (12); a discussion described a priori should be discussed in the
discussion about integration. The US GMP of the meaning of raw data has been pre- Bioanalytical Report.
regulations for laboratory controls include sented earlier, and is equivalent to com- The list of chromatograms that required
this requirement: plete data in the US GMP regulations (8). reintegration, including any manual inte-
If you are subject to FDA’s Pre-Approval grations, and the reasons for reintegration,
21 CFR 211.160(b): “Laboratory controls shall Inspections (PAI), Compliance Program should be included in the Bioanalytical
include the establishment of scientifically Guide (CPG) 7346.832 for inspectors was Report. Original and reintegrated chromato-
sound and appropriate specifications, stan- updated in September 2019 (13). Under grams, and initial and repeat integration
dards, sampling plans, and test procedures Objective 3, the data integrity audit, the results, should be kept for future reference,
designed to assure that components, drug following areas are specifically mentioned and submitted in the Bioanalytical Report for
product containers, closures, in-process in the new version for the inspector to comparative BA/BE (bioavailability or bio-
materials, labeling, and drug products con- focus on: equivalence) studies.
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  347

Gone is the burdensome FDA require- even if it is to get “proper” integration. Sum- not to describe the principles of peak integra-
ment for a manager to preapprove any marizing these citations, we have the follow- tion, as this can be found in the excellent book
manual integration (21), but this is replaced ing areas of non-compliance: by Normal Dyson (23), or in tutorials and man-
by control via a plan or procedure, together • Integrating into compliance uals from CDS suppliers. A suggested peak
with a listing of chromatograms requiring • Failure to retain integration parameters integration workflow is shown in Figure 1 (2,
manual integration (moving baselines) and (for example, a lack of complete data) 24), and we will discuss various aspects of this
the before and after chromatograms. For a • Lack of a procedure for manual integration workflow throughout this article.
study involving the analysis of >1000 samples, • Inappropriate use of integrate inhibit
this could be quite a large burden especially • Unscientific integration Why Manually Integrate Peaks?
if the elimination phase, is relatively long and • No quality oversight We need to consider why we need to inte-
the drug is slowly metabolized or excreted. In my view, the FDA are wrong in their grate peaks manually. The reason is that
These requirements emphasize the need to requirement for a procedure on manual inte- there are situations where a CDS cannot inte-
have robust and reliable analytical proce- gration. To avoid these issues, we must have grate peaks correctly, and some examples of
dures, as discussed in an earlier “Data Integ- a procedure that is structured and compliant, this are:
rity Focus” article (22). and a scientific approach to peak integra- • Split peaks
tion as a whole. What is required is a holistic • Shoulder peaks
A Parcel of Rogues approach with an SOP for all chromatographic • Tailing peaks
A small selection of FDA 483 observations integration, of which manual integration is an • Baseline noise
and warning letter citations of chromato- important subset. Further information about • Negative peaks
graphic peak integration failures are pre- an integration SOP, including banned prac- • Co-eluting peaks
sented in Table I, with the key problems tices such as peak skimming or enhancing, • Rising or falling baselines
highlighted in bold. One piece of free advice can be found in the articles by McDowall, • Slowly eluting peaks (where the CDS
I would offer is not to say to an inspector that Newton and McDowall and Longden and has difficulty identifying the peak start
chromatographers “play with parameters,” McDowall (2–4). The purpose of the SOP is and end).

Robust Methods, Fewer Injections


Software to streamline method development using in silico optimization

ACD/Method Selection Suite

Generate Robust Minimize Experiments


Methods in silico Build optimized methods
Optimize pH, gradient, with minimal injections
temperature, & more
through simulation Predict the Impact of Track Peaks
4 5 35º 40º Method Changes Automatically
6 7 15º 25º
Visualize the effect of Consistently identify
Co pH Te
varying methods without peaks of interest based
lum m
n p added experiments on spectral similarity

Learn more www.acdlabs.com/methdev


A trademark of Advanced Chemistry Development, Inc. (ACD/Labs)
348  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

TABLE I: A Selection of FDA 483 observations and warning letter citations on peak integration

Company Peak Integration Citation


During the inspection, the investigator found no procedures for manual integration or review of electronic
and printed analytical data for (b)(4) stability samples. Electronic integration parameters were not saved
Unimark Remedies Ltd (25)
or recorded manually. When the next samples were analyzed, the previous parameters were overwritten
during the subsequent analyses.
(T)he software you use to conduct HPLC analyses of API for unknown impurities is configured to permit
extensive use of the “inhibit integration” function without scientific justification. For example,.....your
Divi’s Laboratories (26) software was set to inhibit peak integration at four different time periods throughout the analysis. Your
firm reintegrated multiple chromatograms to determine <redacted> levels; however, the parameters for
the reintegration were not retained.
In addition, our investigators documented many iinstances with extensive manipulation of data with no
explanation regarding why the manipulation was conducted. This manipulation would include changing
Leiner Health Products (27) integration parameters or relabeling peaks such that previously resolved peaks would not be integrated and
included in the calculation for impurities…There was no Standard Operation Procedures (SOP) to describe the
policy, standard practice, and circumstances under which manual integration would be allowed…..
Integration of chromatograms for method STM-0076 <redacted> has been performed inconsistently..... The
recommendation is often, but not always followed and results in this area being integrated. There is a lack of
Kashiv BioSciences (28)
scientific justification to support if the tailing portion should be integrated. The change.....was implemented
without fully evaluating the impact on previously processed data.
Your test methods were not capable of demonstrating the purity of your drugs .....(A)nalysts reprocessed
data up to 12 times, and only included the final result in the report for review by Quality Assurance. Your
Megafine Pharma Ltd (29)
Deputy Manager, Quality Control stated that it is common practice to “play with parameters” to get the
proper integration (29).

You failed to establish adequate test procedures. For example, your analyst manually integrated a HPLC
Hubei Danjiangkou Danao
test for <redacted> API despite the fact that the chromatogram lacked peak resolution.....You lacked an
Pharmaceutical Co Ltd (30)
approved protocol for manual integration or quality oversight of the practice.

Your firm failed to properly integrate co-eluting peaks during impurity testing of phentermine HCL
capsules, which resulted in your analysis failing to detect out-of-specification (OOS) results for at least
KVK-Tech, Inc (31)
one lot of drug product.....Multiple examples of your firm’s failure to properly integrate closely
co-eluting peaks were observed during our inspection.

The reasons for the inability of the integra- tions to this statement. None, repeat none. published in 2018, formally defined manual
tion method may be due to: If the peak integration for the sequence integration as:
• Poor method development and valida- is acceptable, then the reportable results
tion where the analytical procedure or can be calculated and reviewed; high fives “(The) (p)rocess used by a person to modify
the integration method is not optimized all around! However, if the peak integra- the integration of a peak area by modifying
or robust. tion is not acceptable we come to the the baseline, splitting peaks or dropping a
• Complex sample matrices resulting in first decision point: Is manual integration baseline as assigned by the chromatogra-
interfering peaks that may still be present allowed for this analysis? If not, we trigger phy software to overrule the pre-established
after sample preparation, such as biologi- a laboratory investigation. integration parameters within the chromato-
cal samples, contrast media, and fermen- The big problem with the citations about graphic software (19).”
tation media. the lack of a manual integration procedure
• Complex mixtures of similar analytes in Table I is that there is no definition of the Is this definition acceptable?
These situations may result in a heavy term “manual integration.” • It is wordy, repetitious, and could be
manual integration workload, as the better phrased.
CDS method is not able to integrate all Defining Manual Integration • The use of the word “overrule” is conten-
peaks automatically. In a “Questions of Quality” column on tious. Chromatography is a dynamic pro-
integration, it was noted that there was cess, and a CDS application can struggle
A Suggested Peak no definition of manual integration, and it to separate overlapping peaks which are
Integration Workflow: 1 was implicitly defined as manual placement obvious to a trained eye.
The first part of the integration workflow in of the baseline by a chromatographer (2). • The biggest issue with this definition
Figure 1 is that all peak integration in the first Mark Newton and I reiterated this defini- is that there is no mention of scien-
instance must always be conducted using tion in our “Data Integrity Focus” article in tific soundless, as defined in the FDA
automatic integration. There are no excep- 2018 (3). The PDA’s Technical Report 80, also GMP regulations (5).
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  349

Chromatographic Run

Automatic
integration

Manual Complete
Laboratory
No Integration No Acceptable? Yes Analysis
Investigation
OK?

Yes

Manual
Integration
Options

1. Rename
peaks or adjust
windows 2. Adjustment of
integration
parameters 3. Manual
reposition of
baselines

Manual Manual

Intervention Reintegration

FIGURE 1: A suggested peak integration workflow (2).

Therefore, a simpler, more concise, and side auditors recognize the legitimate versus
better definition of manual integration egregious use of integration?
should be “manual repositioning of peak Banning the use of manual integration is a
baselines with scientific justification for common response to avoid questions about
their positioning.” data integrity. However, there are three out-
Implicit within this definition is the use of comes to this crude (and stupid) action:
CDS software; otherwise, you’d be draw- • Laboratories will have to accept poor and
ing baselines on paper. However, this also inconsistent integration
requires that the chromatographer is trained, • Analysts will find a workaround that per-
and, ideally, software technical controls mits them to integrate each run with a
should prevent manual repositioning of different set of integration parameters
baselines where this is not justified by the (typically involves performing quanti-
type of the analysis. fication in a LIMS, or worse, a spread-
sheet, without traceability back to the
Should Manual integration methods)
Integration Be Banned? • Analysts will be forced to spend hours
From the citations in Table I, would it be rea- of their day developing complex and
sonable to ban manual integration in regu- manipulative methods to address varia-
lated laboratories? Let’s think this through. tions between chromatograms with a
Experienced analysts know that chromato- single processing method. Typically, this
graphic analysis can be affected by tempera- will require many “integration events”
ture, humidity, and column history, as well that could even include placing peak
as mobile phase preparation, such that one starts and ends at specific time points;
day’s analysis often varies slightly from the in effect, performing manual integration
previous day’s run. To achieve consistent out- under the guise of an automated method
put and measurement, it is critical to adapt to deceive a reviewer (4).
or optimize factors such as peak detec- Consider, also, the nature of an analysis.
tion threshold or retention time windows My experience is based on analysis of small
to ensure consistent, correct, and accurate heterocyclic molecules; however, I acknowl-
peak identification and measurement (4). But edge that there are biologicals, macro-
how can reviewers, approvers, quality, or out- molecules, and chiral separations that will
350  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

baseline placement. My intention is to iden-


TABLE II: A suggested approach when manual integration is and is not allowed
tify an area for data interpretation that rep-
Manual Integration is Allowed When 1,2 Manual Integration is Not Allowed For resents the middle ground between auto-
matic integration and manual integration.
• Improper peak identification
• Peak shaving or clipping As you can see from Figure 1, there are two
• Peak juicing or enhancement types of manual intervention. Let me eluci-
• Peak split by CDS software
• Raising the baseline above the
• Co-eluting peak date my reasoning.
detector signal
• Well-defined peaks on the • Option 1: Peaks have slipped out of a
• Lowering the baseline below the
shoulders of other peaks
detector signal retention window, and they are not cor-
• For impurity peaks close to the limits
• Selectively adjusting integration
of quantification or detection rectly identified. Otherwise, automatic
events
• Matrix interference integration is acceptable, and all that is
• Insufficient sensitivity
• Negative spikes in the baseline
• Using integration parameters required is to change the peak windows
• Failure to separate peaks
inappropriately (peak inhibition)
in the integration method and repro-
• Resolving poor chromatography
due to instrument failure cess. Peak areas are not changed by this
approach. Providing that all changes to
1If each type of manual integration is allowed by the applicable SOP or analytical procedure.
2
integration parameters are recorded
Manual integration is dependent on the nature of the analyte (simple heterocyclic, biological, or
macromolecule; and sample matrix such as liquid, emulsion, or biological, cell culture medium). by the CDS application (either in a new
version of the integration method or
have broader peaks and there will be the • One position could be if these peaks can- the audit trail), this is acceptable, and
impact of the sample matrix that could not be integrated correctly are you out of can be justified on a rational basis, as
impact resolution of compounds. Where is control? We will consider if this is tenable there are no other changes to the run
the laboratory in the research, development, when we have finished discussing the data. Of course, if manual integration
and manufacturing spectrum? The closer to flow chart. was banned, you would be initiating a
product registration and manufacturing, the In my view, manual integration must laboratory investigation already. This is
analytical procedure should be validated be specifically prohibited in the following not the smartest move.
and its performance understood, and the circumstances: • Option 2: Parameters in the integration or
need for manual integration should be • Symmetrical peaks that have acceptable processing method need to be adjusted
restricted to analysis of impurities near the baseline to baseline fitting following and then applied to all injections in the
limits of quantification or detection, albeit automatic integration. run. An example could be change of
with the caveats about the analyte and • Enhancing or shaving peak areas to meet the peak threshold or minimum area to
matrix above. The key question is, “What SST acceptance criteria or allowing a run reduce the impact of baseline noise. Peak
can be justified scientifically?” to meet the test specification. areas may or may not be changed under
In the wrong hands, with the wrong Now we come to what constitutes “man- his option, but, importantly, there is no
intent, and without a robust training and ual integration. Figure 1 presents three manual placement of the baselines by an
review process, altering chromatographic options that I have selected for discussion; analyst. Again, all changes should be cap-
peak processing parameters has been mis- your laboratory SOP may have more areas tured by the software to ensure complete
used by analysts to falsify results. How can depending on the work performed. The data. Don’t even think of an investigation.
this be managed? ideal outcome you require is consistent and • Option 3: To complete the discussion, if
appropriate manual integration that is scien- permitted, manual placement of baselines
A Suggested Peak tifically defensible. Thus, you need to avoid by the chromatographer is required due to,
Integration Workflow: 2 situations where you have inconsistent or say, a late running peak or noise if under-
Returning to the workflow depicted in Figure inappropriate integration. taking an impurity analysis. Peaks areas will
1, we move to a decision point that deter- be changed by the reintegration. Obviously,
mines if manual integration (whatever that Manual Intervention vs. all changes will be monitored by the CDS
term may cover) is permitted for an indi- Manual Integration and recorded, especially the number of
vidual analytical procedure. If not, the next Let me introduce two terms: manual inte- attempts at integration.
stage is a laboratory investigation. gration and manual intervention (2). The Note that the three options in Figure 1
What methods could we consider for former term you know and love; it has been are shown on different levels. This is my way
inclusion for no manual integration? defined above and is manual placement of illustrating the descent of a chromatog-
• Measurement of active pharmaceutical of baselines by a chromatographer. But rapher into the manual integration nether-
ingredients (APIs) manual intervention? Is this just playing with world. In the first option you may think of the
• Registered methods for finished product words to confuse you? first option as purgatory, and the second as
for the active ingredient Manual intervention is defined as chang- limbo, while the third option is... well, you get
• Stability indicating methods (main peak) ing integration parameters without manual the idea.
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  351

Options 1 and 2 are manual intervention, No Manual


Integration
but the baseline placement is performed Allowed

by the CDS and is not altered by a chroma-


tographer. These are the preferred options, Automatic
Integration
Manual
Integration
System
Suitability Test
Standards
Other
Checks
Samples

and easier to justify scientifically. Manual


intervention is also acceptable, according to
the PDA’s Technical Report 80 (19). Option 3 • Check integration
• Manual
• Only where
permitted
• Must meet
predefined
• Must meet
predefined
• Drift checks
and QC samples • Peak shape
intervention: • Standards & acceptance acceptance • Must meet acceptable
is where a chromatographer goes through adjust parameters samples
the same
treated criteria criteria predefined • Peak areas
generated
acceptance
individual chromatograms and repositions criteria

the baselines where appropriate. This latter


Order of
point is important; it is an exercise of scien- Evaluation

tific judgment that needs to be backed up FIGURE 2: Scope and order of chromatographic integration (3).
by the procedures within the integration
SOP and the changes retained in the audit This places responsibility on any labora- • Standards injections
trail of the CDS. Table II shows suggestions tory to develop robust analytical chromato- • Other checks, such as quality control sam-
for when manual integration is and is not graphic procedures with reliable separations ples and blanks
allowed, depending on the nature of the that are fit for use as discussed in a previous • Samples.
analysis undertaken by a laboratory. “Data Integrity Focus” article (22). A key com- If any of these items fail acceptance crite-
ponent of this approach is that the resultant ria, STOP! Do not review anything involving
Why Use Automatic Integration? peak integration must be consistent. This is a samples until assay criteria are met. Looking
This is a very simple question to answer from subtle, but vital, difference that is not always at sample results prior to accepting the run
a business perspective: sequences where appreciated. could be considered “integrating into com-
integration is automatic are very much pliance,” as you wanted to invalidate testing
quicker to review. In contrast, manual inte- Sequence of Data File Integration you didn’t like (or accept one you did like).
gration is slow, especially for sequences with In 2018, Mark Newton and I discussed the Fresenius Kabi Oncology received a warning
a large number of injections with complex order of integration and processing of any letter from the FDA for such behavior (32).
separations where each injection must be injection sequence (3), and, due to the
integrated manually. Often the review can importance of correct integration, it is worth Is the FDA Inhibiting Integration?
take longer than the actual analysis (3). Con- repeating here. Figure 2 shows the order of The use of inhibit integration function is cur-
sequently, a faster review process means that integration evaluation: rently a hot regulatory topic, as can be seen
analysis is completed quicker, a batch can • Automatic integration and manual from in the FDA regulatory citation of Divi’s
be released sooner, and the organization intervention Laboratories (shown in Table I), where this
cash flow benefits. There is also the bonus of • Manual integration (if permitted) function was used four times in the middle of
lower regulatory scrutiny. • System suitability test injections a chromatogram.

CAMAG® HPTLC PRO


Fully automated sample analysis and
evaluation system for routine quality control

Explore the next dimension of High-Performance


Thin-Layer Chromatography at camag.com
352  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

This citation is based on 21 CFR 211.160(b) (5) that was discussed cions that the excluded peaks might be real impurities, exclud-
earlier in this article, and the key questions to ask are if, where ing these system peaks needs to be carefully documented and
and when can integrate inhibit be used. It has been said in some justified in the method development and validation reports;
audits and inspections that this function cannot be used. This is otherwise, they should be integrated and marked clearly as
an untenable situation, as there is no explicit or implicit statement system peaks.
in the GxP regulations for this attitude. However, it comes back to
scientific soundness, and a laboratory must be able to justify the System Evaluation Injections
use of the function. Trial injections using actual samples feature in many warning let-
Let us consider the following scenarios: ters (33) and Question 13 of the FDA Data Integrity Guidance (16),
• There is baseline perturbation with a large negative peak after which states:
an injection.
• A peak of interest elutes shortly after the negative peak. The use of “FDA prohibits sampling and testing with the goal of achieving a spe-
integrate inhibit is fully justified from the start of the injection, void cific result or to overcome an unacceptable result (e.g., testing different
volume, solvent front, and until the baseline has returned to nor- samples until the desired passing result is obtained).”
mal and before elution of the peak of interest. Otherwise, there is
a large probability that baseline placement of the analyte could be This is correct and should never be acceptable in a GxP
adversely influenced by the negative peak. This would also result laboratory SOP.
in more manual integration. However, consider the following situation: You are analyzing low
• Similar scenarios occur when extraneous peaks are washed from volume samples from a non-clinical study. There is a total volume of
the column, or baseline perturbations from mobile phase change 20 µL plasma sample that is extracted, and there is only enough for
during a gradient elution, or wash at the end of a chromatogram. a single injection from each sample. Ask yourself the question: Are
What is more problematic is the use of integrate inhibit in you going to commit an analytical run of samples without checking
the middle of a run, as cited above. If system, blank, or other that the system is ready? The cost of repeating the study is a high
non-sample peaks occur in the middle of a chromatogram, tra- six figure sum if a run does not work. Therefore, from a practical
ditionally those peaks were not integrated. Because of suspi- perspective, we need a way of checking that a system is ready for
analysis, but does not involve testing into compliance with samples.
Ah, somebody says to use SST injections. The problem is that you
may need several replicates to determine if the system is ready, and
SST should never be started until you are confident that the system
is equilibrated.
Consider the following approach for system evaluation or readi-
SERVING ROYALTY. EXCEEDING EXPECTATIONS. EVERY MOMENT. ness injections or equilibration checks (4):
• The ability to use system evaluation injections must be docu-
mented in an applicable SOP or analytical procedure.
• The minimum column equilibration time needs to be documented in
the method to avoid excessive system readiness injections.
• Only system evaluation injections prepared from a suitable refer-
ence standard can be used to evaluate if the chromatographic sys-
tem ready. Records of the solution preparation must be available.
Ideally, a test mixture which mimics the separation characteristics,
but is easily distinguishable from real samples could be used.
• Should the maximum number of system evaluation injections
that can be made be documented in the procedure before a
problem with the chromatographic system needs to be investi-
• Provider of top brand HPLC instrumentation products
gated? If the cause is thought to be an equilibration issue, waiting
• Equivalent to corresponding OEM products
and injecting again should be sufficient. If the system continues
• Serving customers for over 35 years
• Reduce product repair expenses by up to 30%
to not behave, then an investigation is needed, the cause should
• Lifetime Warranty on manufacturing defects be found, remediated, and documented in the instrument log
book before checking the system evaluation again. If the prob-
www.sciencix.com 800.682.6480 sales@sciencix.com
lem requires maintenance to resolve it (for example, a pump
seal replacement), then requalification of the pump should be
conducted and documented before beginning the analysis.
• Using sample preparations must be prohibited in this procedure,
accompanied with staff training.
WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  353

• System evaluation injections are part of that the factors involved in the separation are of manual integration is a regulatory concern,
complete data for the analytical run, and known and controlled adequately (22). Chro- and use needs to be scientifically sound. But
must be included in the instrument log matographic methods should be developed also be aware that, as discussed earlier, man-
book entries, along with any investiga- with automatic integration and the norm ual integration slows down a process, so see
tion and remediation work on the instru- and not the exception. Management need Rule 1 to get the right method depending
ment. A common practice is to store to understand that adequate time must be on the sample matrix and peaks of interest.
the data from these tests in a separate given to method development and validation.
folder or location to the real analyses. This is especially true for pharmacopoeial Rule 4: Understand how the CDS works
This practice needs careful management methods that never work as written. and how the numbers are generated. This
and documentation, as it becomes dif- requires basic training in the principles of peak
ficult to connect those injections to the Rule 2: Never use default integration integration and how a CDS works. The prob-
official laboratory work. Ideally, all work, parameters, always configure specific lem is that with mergers, acquisitions, and
including system evaluation injections, integration for each method. Without encouraging experiences analysts to retire
should be stored in the same location. exception, peak integration and result pro- and employ younger workers, skills are being
cessing must be defined and validated for eroded, and a CDS can be looked at as a
Five Rules of Integration each method so that all peak windows and black box that always gives the right answers.
An integration SOP was discussed earlier. To names are defined, and, if necessary, any
help understand what should be in it and system peaks are identified. Using a default Rule 5: Use your brain – Think! This rule
the associated training, there are five rules of or generic method results in excessive need is sometimes difficult to follow, but it follows
integration to consider (4): for manual integration to name and calculate on from Rule 4. You can have what appears
peaks, and so forth. to be a perfect separation and peak integra-
Rule 1: The main function of a CDS is not tion, but look at peak start and end place-
to correct your poor chromatography. This Rule 3: Always use automatic integration ment—do they look right? Use the zoom
places greater emphasis on the development as a first option and control manual inte- and overlay functions of the CDS to see of
of robust chromatographic procedures so gration practices. Remember that the use standards and samples have the right peak
354  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  JUNE 2020WWW.CHROMATOGRAPHYONLINE.COM

shape, etc. We discussed in the article on Summary agement and Integrity in Regulated GMP / GDP
second person review the need to have Control of peak integration is imperative in a Environments Draft. (Pharmaceutical Inspec-
tion Convention/Pharmaceutical Inspection
an adequate sized monitor; this is a case regulated laboratory. Good peak integration
Cooperation Scheme, Geneva, 2018).
where one is essential (34). The analyst has requires good chromatography. Good chro-
(19) Technical Report 80: Data Integrity Manage-
the responsibility to execute applicable matography requires a well developed and ment System for Pharmaceutical Laboratories
procedures correctly, which includes cor- robust analytical procedure. The bottom line (Parenteral Drug Association [PDA], Bethesda,
rect peak integration. The reviewer, how- is: Are you in control of the analytical proce- Maryland, 2018).
ever, also has a role to ensure that all inte- dure and peak integration? (20) ICH M10 Bioanalytical Method Validation
Stage 2 (International Council on Harmaoni-
gration (whether automated, optimized, sation: Geneva Switzerland, 2019 [draft]).
or manually placed) follows the method References (21) FDA Guidance for Industry: Bioanalytical
guidance for placing baselines as the SOP (1) Able Laboratories Form 483 Observations. Methods Validation (Food and Drug Admin-
describes, especially when the representa- 2005 23 Dec 2019]; Available from: https:// istration: Silver Spring, Maryland, 2018).
www.fda.gov/media/70711/download. (22) R.D. McDowall, LCGC North. Am. 38(4), 233–
tive area for unresolved peaks are being
(2) R.D. McDowall, LCGC Europe 28(6), 336–342 240 (2020).
estimated. Significant peak area manipula- (2015). (23) N. Dyson, Chromatographic Integration
tion should be easily noticed by an experi- (3) M.E. Newton and R.D. McDowall, LCGC Methods (Cambridge: Royal Society of
enced reviewer. North Am. 36(5), 330–335 (2018). Chemistry, Cambridge, United Kindom, 2nd
(4) H. Longden and R.D. McDowall, LCGC Ed., 1998).
Quo Vadis Peak Integration? Europe 21(12), 641–651 (2019). (24) R.D McDowall, Validation of Chromatography
(5) 21 CFR 211 Current Good Manufacturing Data Systems: Ensuring Data Integrity, Meet-
If you think that peak integration is a regula- ing Business and Regulatory Requirements
Practice for Finished Pharmaceutical Prod-
tory issue now, what will it be like in the future? ucts. (Food and Drug Administration: Sliver (Cambridge: Royal Society of Chemistry,
The May 2019 supplement to LCGC Europe Spring, MD, 2008). Cambridge, United Kindom, 2nd Ed., 2017).
gives an interesting glimpse of the future (6) ICH Q7(R1) - Good Manufacturing Prac- (25) FDA Warning Letter Unimark Remedies,
tice for Active Pharmaceutical Ingredients. India, (Food and Drug Administration: Silver
in which Wahab and associates (35) discuss Spring, Maryland, 2014).
(International Conference on Harmonisation:
advanced signal processing techniques that Geneva, 2016). (26) FDA Warning Letter: Divi’s Laboratories Ltd.
could be used in chromatographic integra- (7) EudraLex - Volume 4 Good Manufactur- (Unit II) ( Warning Letter 320-17-34). (Food
tion. The techniques listed are: ing Practice (GMP) guidelines, Part 2 - Basic and Drug Administration: Silver Spring,
Requirements for Active Substances used as Maryland, 2017).
• Deconvolution of extra column effects
Starting Materials (European Commission, (27) FDA Warning Letter Leiner Health laborato-
by Fourier transformation for removing Brussels, Belgium, 2014). ries (Food and Drug Administration: Rockville,
band broadening. (8) R.D. McDowall, LCGC North. Am. 37(4), 265– Maryland, 2006).
• Peak area extraction by iterative curve 268 (2019). (28) FDA 483 Observations: Kashiv BioSciences.
fitting for partial overlapping peaks in (9) R.D. McDowall, Spectroscopy 28(4), 18–25 (Food and Drug Administration: Silver Spring,
(2013). Maryland, 2018).
a chromatogram.
(10) R.D. McDowall, Spectroscopy 31(11), 18-21 (29) FDA Warning Letter: Magafine Pharma Ltd.,
• Model free approaches for peak infor- US FDA:, Silver Spring, Maryland, 2017)
(2016).
mation extraction, another approach for (30) FDA Warning Letter: Hubei Danjiangkou
(11) R.D. McDowall, Spectroscopy 33(12), 8–11
extracting peak areas from overlapping (2018). Danao Pharmaceutical Co., Ltd. (Food and
peaks in complex matrices. Drug Administration, Silver Spring, Mary-
(12) EudraLex - Volume 4 Good Manufacturing land, 2017).
• Direct resolution by Power Law increases Practice (GMP) Guidelines, Chapter 4 Docu-
mentation (European Commission, Brussels, (31) FDA Warning Letter: KVK-Tech, Inc. (Food
resolution by reducing peak width and Drug Administration, Silver Spring, Mary-
Belgium, 2011).
and trailing. land, 2020).
(13) FDA Compliance Program Guide CPG
• Direct resolution enhancement by 7346.832 Pre-Approval Inspections (Food (32) FDA Warning Letter Fresenius Kabi Oncol-
even derivative peak sharpening and Drug Administration: Silver Spring, ogy (Food and Drug Administration: Silver
Maryland, 2019). Spring, Maryland, 2017) Available from:
also increases resolution by reducing https://www.fda.gov/ICECI/EnforcementAc-
peak width. (14) R.D. McDowall, Spectroscopy 34(12), 14–19 tions/WarningLetters/2017/ucm589941.htm.
(2019).
It is beyond the scope of this article (33) R.D.McDowall, LCGC Europe 27(9), 486–492
(15) MHRA GxP Data Integrity Guidance and Defini- (2014).
to present and discuss what is already in tions (Medicines and Healthcare Products Regu-
Wahab’s paper, but if any of these tech- latory Agency, London, United Kingdom, 2018). (34) R.D. McDowall, LCGC North. Am. 38(3), 163–
172, 193 (2020).
niques are integrated into a chromatogra- (16) FDA Guidance for Industry- Data Integrity and
Compliance With Drug CGMP Questions and (35) M.F. Wahab, G. Hellinghausen, and D.W.
phy data system, then their use needs to Armstrong, LCGC Europe 32(s5), 22–28 (2019).
Answers (Food and Drug Administration: Silver
be justified scientifically. This means from Spring, Maryland, 2018).
development through validation to use of (17) WHO Technical Report Series No.996 Annex
a method. R.D. McDowall
5 Guidance on Good Data and Records Man-
is the director of R.D. McDowall Lim-
If regulators are worried by peak inte- agement Practices (World Health Organisation: ited in the UK. Direct correspondence
gration now, they could be paranoid in Geneva, Switzerland, 2016). to: rdmcdowall@btconnect.com
the future! (18) PIC/S PI-041-3 Good Practices for Data Man-
SUPPLEMENT TO

THE
APPLICATION
NOTEBOOK

June 2020
www.chromatographyonline.com
TABLE OF CONTENTS

THE APPLICATION
NOTEBOOK
Environmental
357 The Extraction of PFAS Molecules from Spiked Soil
Alicia D. Stell, CEM Corporation

Medical/Biological
359 Tailored Analysis of Phospholipid Classes Using
iHILIC-Fusion(+) as First Dimension for Online
Two-Dimensional HILIC–Reversed-Phase LC–MS
Patrick O. Helmer*, Carina M. Wienken*, Wen Jiang†, and Heiko Hayen*,
*Institute of Inorganic and Analytical Chemistry, University of Münster, Münster,

Germany, †Hilicon AB

361 Online SPE and LC–MS Analysis of Thyroid Hormones


in Human Serum
MilliporeSigma

362 Analysis of Fentanyl and Its Analogues in Human


Urine by LC–MS/MS
Shun-Hsin Liang and Frances Carroll,Restek Corporation

364 Chondroitin Sulfate Analyzed by SEC–MALS


Wyatt Technology

Food and Beverage


365 HILIC Columns for Phosphorylated Sugar Analysis
Showa Denko America, Inc.

Pharmaceutical/Drug Discovery
366 Amino Acid Analysis by European Pharmacopeia
to Support Coronavirus Research
Pickering Laboratories, Inc.
367 Size-Exclusion Chromatography for the Impurity
Analysis of Adeno-Associated Virus Serotypes
Stephan M. Koza and Weibin Chen,Waters Corporation
369 An Anion-Exchange Chromatography Method
for Monitoring Empty Capsid Content in
Adeno-Associated Virus Serotype AAV8
Hua Yang, Stephan M. Koza, and Weibin Chen,Waters Corporation

356 THE APPLICATION NOTEBOOK – JUNE 2020


ENVIRONMENTAL

The Extraction of PFAS Molecules from Spiked Soil


Alicia D. Stell, CEM Corporation

Per- and polyfluoroalkyl substances (PFAS) are a group of manufactured Sample Preparation and EDGE Method
chemicals that are used in a wide variety of industries because of their Five grams of clean sandy loam was weighed directly into a Q-Cup®
resistance to stains, grease, and high temperatures. They possess a chain containing the S1 Q-Disc® stack (C9-G1-C9 sandwich). Each sample
of linked carbon atoms with fluorine atoms branching off the main chain. was spiked with 2 ng or 200 ng of standard in HPLC-grade methanol,
The presence of the strong carbon-fluorine bond contributes to the stability resulting in low spike and high spike samples, respectively. Each set of
of these compounds, earning them the nickname “forever chemicals.” spikes was done in triplicate. The sample was extracted with the EDGE
PFAS are used in products such as nonstick cookware, firefighting foam, using 80:20 methanol:water with 0.3% ammonium hydroxide using the
and stain-resistant carpets. method provided. Each set of spikes was extracted on the EDGE, and then
Because of their persistent nature and their widespread use, this group a blank extraction of the system was done with a Q-Cup containing the S1
of substances has leached into the environment with limited methods Q-Disc stack (C9-G1-C9 sandwich) to assess the level of carryover. Each
of remediation. Furthermore, these compounds have been found to extraction was collected in a polypropylene conical tube using an EDGE
bioaccumulate in animals and humans, and exposure in humans has rack. The sample was brought up to a final volume of 20 mL using 80:20
been shown to cause adverse health outcomes, including cancer, infertility, methanol:water with 0.3% ammonium hydroxide, and 20 µL of formic
and endocrine disruption. Thus, the assessment of the levels of PFAS in acid was added to each sample to neutralize the sample. The samples
the environment is important to the health and safety of humans. were then analyzed by Pace Analytical.
The EDGE, an automated solvent extraction system, was used to extract
a subset of PFAS molecules from spiked soil samples. The EDGE was EDGE Method
able to extract the soil samples in less than 10 min. The extraction yielded Q-Disc: S1 stack (C9-G1-C9 sandwich)
excellent recoveries and standard deviations. Furthermore, there was no Extraction Solvent: 80:20 methanol:water with 0.3% ammonium
carryover found between samples. The EDGE is an excellent choice for hydroxide
laboratories seeking to automate their PFAS extraction. Cycle 1
Top Add: 10 mL
Method Bottom Add: 0 mL
Rinse: 0 mL
Reagents Temperature: 65 °C
Clean sandy loam was purchased from MilliporeSigma. A PFAS standard Hold Time: 3 min
containing 24 different compounds (Part number 99207) was purchased Cycle 2
from Absolute Standards, Inc. HPLC-grade methanol, HPLC-grade water, Top Add: 10 mL
HPLC-grade formic acid, and ammonium hydroxide were purchased from Bottom Add: 0 mL
Fisher Scientific. Rinse: 0 mL

THE APPLICATION NOTEBOOK – JUNE 2020 357


ENVIRONMENTAL

Table I: Average recovery results from the low spike and high spike soil samples

Compound Low Spike RSD (n = 3) High Spike RSD (n = 3)

1H, 1H, 2H, 2H-perfluorodecane sulfonic acid (8:2 FTS) 83.00% 7.81% 87.00% 7.00%
1H, 1H, 2H, 2H-perfluorooctane sulfonic acid (6:2 FTS) 80.00% 3.00% 87.67% 7.09%
1H,1H,2H,2H-perfluorohexane sulfonic acid (4:2 FTS) 86.67% 10.02% 87.67% 5.13%
N-ethylperfluoro-1-octanesulfonamidoacetic acid (EtFOSAA) 85.67% 6.66% 86.00% 5.57%
N-methylperfluoro-1-octanesulfonamidoacetic acid (MeFOSAA) 78.67% 1.15% 81.33% 4.16%
Perfluoro-1-butanesulfonic acid (PFBS) 78.00% 3.00% 86.00% 1.73%
Perfluoro-1-decanesulfonic acid (PFDS) 77.00% 6.56% 84.33% 0.58%
Perfluoro-1-heptanesulfonic acid (PFHpS) 81.00% 2.65% 87.00% 2.65%
Perfluoro-1-nonanesulfonic acid (PFNS) 76.00% 6.56% 84.67% 2.89%
Perfluoro-1-octanesulfonamide (PFOSA) 81.00% 8.00% 86.67% 5.03%
Perfluoro-1-pentanesulfonic acid (PFPeS) 78.00% 4.36% 86.33% 0.58%
Perfluorohexanesulfonic acid (PFHxS) 98.33% 2.89% 89.67% 3.79%
Perfluoro-n-butanoic acid (PFBA) 85.65% 4.01% 88.17% 2.20%
Perfluoro-n-decanoic acid (PFDA) 101.33% 7.51% 93.33% 2.89%
Perfluoro-n-dodecanoic acid (PFDoA) 79.67% 7.37% 79.33% 2.08%
Perfluoro-n-heptanoic acid (PFHpA) 90.67% 9.02% 81.67% 3.21%
Perfluoro-n-hexanoic acid (PFHxA) 100.67% 9.02% 91.08% 6.64%
Perfluoro-n-nonanoic acid (PFNA) 96.33% 3.21% 92.67% 2.52%
Perfluoro-n-octanoic acid (PFOA) 82.77% 3.87% 85.53% 2.25%
Perfluoro-n-pentanoic acid (PFPeA) 79.50% 0.71% 85.67% 4.04%
Perfluoro-n-tetradecanoic acid (PFTeDA) 86.67% 4.73% 88.67% 1.15%
Perfluoro-n-tridecanoic acid (PFTrDA) 63.00% 3.46% 68.33% 3.51%
Perfluoro-n-u0ecanoic acid (PFUdA) 76.33% 1.53% 79.67% 3.51%
Perfluorooctanesulfonic acid (PFOS) 84.00% 4.00% 79.60% 0.53%

Temperature: 65 °C Conclusion
Hold Time: 4 min The analytical assessment of PFAS compounds is critical because of their
Wash 1 widespread nature, high stability, and adverse health effects. The EDGE
Wash Solvent: Methanol was able to rapidly and efficiently extract spiked soil samples with excel-
Wash Volume: 10 mL lent recoveries and RSD values. The EDGE also saw no carryover after
Temperature: 50 °C extractions into the subsequent extraction. The EDGE is an excellent ex-
Hold: 3 s traction tool for laboratories seeking to automate their PFAS extractions
Wash 2 with great efficiency.
Wash Solvent: 80:20 methanol:water with 0.3% ammonium hydroxide
Wash Volume: 10 mL
Temperature: None
Hold: None

Results
The recovery data in Table I, from the low and high spikes, indicated that
the samples were extracted with high efficiency, with recoveries ranging
from 63% to 101%. The resulting RSD values were also low, indicating
the recovery data were reproducible. The extraction data from the empty CEM Corporation
Q-Cup indicated that there was no carryover of the spiked compounds 3100 Smith Farm Road, Matthews, NC 28104
within the system, indicating the wash was aggressive enough to remove tel. (800) 726-3331
any residual PFAS compounds. Website: www.cem.com

358 THE APPLICATION NOTEBOOK – JUNE 2020


MEDICAL/BIOLOGICAL

Tailored Analysis of Phospholipid Classes


Using iHILIC-Fusion(+) as First Dimension for Online
Two-Dimensional HILIC–Reversed-Phase LC–MS
Patrick O. Helmer*, Carina M. Wienken*, Wen Jiang†, and Heiko Hayen*,
*Institute
of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany, †Hilicon AB

Phospholipids (PLs) are a large lipid subgroup that is involved 1D HILIC Separation:
in important cell functions in all organisms. They are the Column: 20 × 2.1 mm, 5-µm, 100 Å, iHILIC®-Fusion(+) (P/N
main components of biological membranes and are essential 100.022.0510, HILICON)
for many biologic processes within and between cells, for Eluents: A) ammonium formate solution (35 mM, pH 3.5) and
example, signal transduction, cell growth, and various 95:5 (v/v) acetonitrile; B) acetonitrile
transport processes. The basic building blocks for PLs are fatty Gradient Elution: 0–0.2 min, 97% B; 0.2–0.5 min, from 97%
acids linked to a glycerol backbone and polar head groups to 93% B; 0.5–2.75 min, 93% B; 2.75–7.5 min, from 93% to
(Figure 1). In addition to the main membrane PLs, such as 60% B; 7.5–11 min, 60% B; 11–11.5 min, from 60% to 97%
phosphatidylcholine (PC) and phosphatidylethanolamine B; re-equilibration is parallel to reversed-phase LC separation
(PE), other PL classes are of great importance as well. Some at 97% B.
PLs have significantly lower concentrations. For example, Flow Rate: 0.3 mL/min; 0.05 mL/min (during parallel
in eukaryotic organisms, cardiolipin (CL) is exclusively reversed‑phase LC separation)
located in the inner mitochondrial membrane (1). Thus, Column Temperature: 40 °C
the concentration of CL in total lipid extracts is very low in Injection Volume: 2–20 µL
comparison to the major membrane lipids, such as PLs and Heart-Cut Setup for PL Transfer: An online heart-cut 2D
cholesterol, but also triacylglycerols (TGs) (2,3). Furthermore, HILIC–reversed-phase LC–MS setup was developed for PL
ion suppression effects complicate the mass spectrometric
(MS) analysis, and a tailored analysis of low abundant PL
classes is often required.
In our previous study, we demonstrated the advantage
of performing sample preparation by solid-phase extraction
(SPE) in hydrophilic interaction liquid chromatography
PC
(HILIC) mode (4). The nonpolar fraction of lipids was
first separated from polar PLs using an offline HILIC SPE
PE
method, and thereafter separation by reversed-phase liquid
chromatography (LC) was performed. Offline methods often
require solvent evaporation and reconstituting analytes in
a suitable solvent for the second dimension, which is a CL
time‑consuming approach and risks losing labile PLs. In this
application, a novel online heart-cut two-dimensional (2D)
HILIC–reversed-phase LC–MS method is presented for the
separation of PL classes and nonpolar lipids, where HILIC was
polar
utilized in the first dimension (1D) and reversed-phase LC in
second dimension (2D). TG cholesterol

Experimental
Lipid Standards: Triacylglycerol (TG 48:0), cholesterol (Chol),
phosphatidylcholine (PC 32:0), phosphatidylethanolamine
(PE 32:0), cardiolipin (CL 64:4), and yeast total lipid extract
(Saccharomyces cerevisiae).
LC–MS/MS Setup: Thermo Scientific Ultimate 3000 system nonpolar
with dual gradient pump hyphenated to a Q Exactive™ Plus
Hybrid Quadrupole-Orbitrap™ mass spectrometer. Ionization
Figure 1: Selected structures of nonpolar lipids and polar PLs. HILIC
was performed utilizing electrospray ionization in negative separation of PLs takes place according to their polar head groups
ionization mode as described by Helmer et al. (3). (shown in green).

THE APPLICATION NOTEBOOK – JUNE 2020 359


MEDICAL/BIOLOGICAL

transfer from 1D (HILIC) to 2D (reversed-phase LC). As described


earlier, a six-port valve equipped with a sample loop (300 µL,
PEEK) was utilized as the transfer (Figure 2) (3). At first, the HILIC
eluate was directed to the waste. By switching the valve from A (a) TG PE CL PC
(2_1) to B (1_6) position, the selected PL fraction was transferred lipid standards
into the loop for the separation in the second dimension. Since we
focused on CL in this study, the transfer time window was set at its 1D HILIC (b)
retention time of 6.35–6.95 min.
CL 66:4 S. cerevisiae
CL 68:4
Results and Conclusion
CL 70:4
A tailored analysis of CL species in yeast (S. cerevisiae) was
achieved utilizing two-dimensional HILIC–reversed-phase (c)
LC–MS. In the first dimension, the separation of PL classes was
carried out using a HILIC method. In addition to that, nonpolar 1D-RPLC
2D RPLC
lipids did not show retention and were eluted at the void volume. 0 15 30
As shown in Figure 3(a), the lipid classes (TG, PE, CL, and PC) yeast CL
were well separated according to their different head groups
12 20
with this new method. The application of this method to a yeast Time (min)
total lipid extract (Figure 3[b]) shows the intended coelution
of the CL species in yeast, where one typical CL species is CL
Figure 3: (a) 1D HILIC separation of representative lipid standards;
66:4 with 66 carbon atoms and four double bonds in the four (b) coelution of dominant CL species in yeast (S. cerevisiae) within the
linked fatty acids. transfer window; (c) comparison of reversed-phase LC TICs in 1D (red)
In the developed 2D HILIC–reversed-phase LC separation and 2D (black).
method, a transfer window with all CL species from 1D
HILIC was first determined for the transfer via a heart-cut second dimension separation of low abundant PL classes.
setup to the 2D reversed-phase LC system. Compared to the Interestingly, low abundant CL species can be sensitively
conventional one-dimensional (1D) reversed-phase LC (red), detected and observed, even in the TIC. In summary, the
2D reversed‑phase LC (black) shows a significant decrease automated online 2D method via heart-cut transfer offers a fast
of background signals (mainly TGs), and low abundant CL and gentle sample preparation and clean-up procedure that is
species can even be observed in the total ion current (TIC) very useful for analyzing low abundant PL classes as well as
chromatogram (Figure 3[c]). Furthermore, by the reduction of labile PLs, such as PL oxidation products (3).
the background as observed in 1D reversed-phase LC, higher
CL signal intensities have been observed (3). References
The selective HILIC pre-separation of lipid classes in the (1) J.M. Berg et al., Stryer Biochemistry (Springer), (2018).
first dimension significantly reduced the matrix effects in the (2) M. Lange et al., Chromatographia 82, 77–100 (2018).
(3) P.O. Helmer et al., J. Chromatogr. A. 460918, in press https://doi.
org/10.1016/j.chroma.2020.460918
(4) P.O. Helmer et al., Rapid Commun. Mass Spectrom. 34, e8566 (2020).
https://doi.org/10.1002/rcm.8566

Pump 1 HILIC Waste

2 1
Position A: 2_1
3 Valve 6
Position B: 1_6
4 5

Pump 2 RP-MS

HILICON AB
Tvistevägen 48, SE-90736 Umeå, Sweden
Tel.: +46 (90) 193469
Figure 2: Valve setup for the transfer of PL classes from 1D HILIC E-mail: info@hilicon.com
to 2D reversed-phase (RP) LC via heart-cut approach. Website: www.hilicon.com

360 THE APPLICATION NOTEBOOK – JUNE 2020


MEDICAL/BIOLOGICAL

Online SPE and LC–MS Analysis of Thyroid


Hormones in Human Serum
MilliporeSigma

Thyroid hormones play critical roles in the regulation of biological


processes, including growth, metabolism, protein synthesis, and 4750
1:651.80>605.50(+) CE: -24.0 1:777.70>731.80(+) CE: -28.0

brain development. Specifically, both 3,3’,5,5’-tetraiodo-L-thyronine 4500


4250
4000

(thyroxine or T4) and 3,3’,5-triiodo-L-thyronine (T3) are essential for 3750


3500
3250

development and maintenance of normal physiological functions. For 3000


2750
2500
a clinical laboratory, measurements of total T4 and total T3, along with 2250
2000
1750
estimates of free T4 (FT4) and free T3 (FT3), are important for the 1500
1250

diagnosis and monitoring of thyroid diseases. Most clinical laboratories


1000
750
500

measure thyroid hormones using immunoassays, which offer a relatively 250


0
-250

rapid, high patient sample throughput that lends itself to automation, but 2.0 2.5 3.0 3.5 4.0 4.5 5.0
Time (min)
5.5 6.0 6.5 7.0 7.5

are compromised by problems with assay interference and by changes Figure 1: Representative LC–MS chromatogram of thyroid hormones
in protein levels that alter free hormone availability (1). in human serum with RPA online cartridge.
Liquid chromatography–mass spectrometry (LC–MS) can offer superior
specificity and speed over immunoassays for determination of thyroid time. We used RPA online cartridges with LC–MS analysis for the
hormones in biological matrices such as serum and tissues. The present detection of thyroid hormones from human serum. The samples were
work demonstrates successful online solid-phase extraction (SPE) with LC– protein precipitated with methanol containing ammonium formate
MS for rapid determination of T4, T3, and 3,3’,5’-triiodo-L- thyronine (rT3) and after centrifugation then directly injected for online SPE and
from biological matrices. LC–MS analysis. Sample loading–washing was performed entirely by
the instrument, eliminating the time-consuming solvent evaporation
Experimental and reconstitution steps.
Materials: Supel™ Genie RP-Amide (RPA) and C8 (results not shown) The RPA cartridges captured a trace amount (100 ng/mL × 2 µL in
online SPE cartridges (2 cm × 4.0 mm i.d.); human serum; and protein this case) of thyroid hormones from human serum (Figure 1). All three
precipitation solvent: methanol with 1% (w/v) ammonium formate. analytes were well resolved with a peak width at half height <6 s and
Sample Processing: Human serum spiked with analytes was protein tailing factor from 1.5–1.8. The total run time was within 6 min.
precipitated by vortex mixing with the precipitation solvent at a 1:3 An online SPE–LC–MS method can be used for rapid detection of
ratio. The mixture was then centrifuged at 10,000 ×g for 3 min and thyroid hormones in human serum with minimal hands-on effort and
the supernatant was collected and directly injected for LC–MS analysis. time-consuming steps.
Online SPE–LC–MS Setup: Six-port switching valve and two pumps; one
for sample loading and washing, the other for sample elution. To minimize Reference
potential peak broadening from the cartridges, the flow of sample loading– (1) N. Kahric-Janicic, S.J. Soldin, O.P. Soldin, T. West, J. Gu, and J. Jonklaas,
washing and the subsequent elution are in reversed directions. Thyroid 17(4), 303–11 (2007).

Results and Discussion


Conventional SPE typically involves multiple labor-intensive and time-
consuming steps: conditioning, sample loading, washing, elution, MilliporeSigma
evaporation, and reconstitution. Supel Genie online cartridges were 400 Summit Drive, Burlington, MA 01803
developed to automate the sample preparation process, minimize tel. (800) 645-5476
hands-on time and human error, and reduce overall sample processing Website: www.sigmaaldrich.com

Table I: Ruggedness of online SPE–LC–MS with RPA cartridges from 120 consecutive injections of human serum samples.
Peak Area
Retention Time (Min) Retention Time Reproducibility Peak Area
Analyte MRM Quantifier Reproducibility
(Avg. n = 120) (%RSD, n = 120) (Avg. n = 120)
(%RSD, n = 120)
3,3’,5-triiodo-L-thyronine (T3) 651.8 / 605.5 4.03 0.2 27046 5.1
3,3’,5-triiodo-L-thyronine (rT3) 651.8 / 605.5 4.43 0.2 33723 6.2
3,3’,5,5’-tetraiodo-L-thyronine (T4) 777.7 / 731.8 4.79 0.2 23766 7.7

THE APPLICATION NOTEBOOK – JUNE 2020 361


MEDICAL/BIOLOGICAL

Analysis of Fentanyl and Its


Analogues in Human Urine
by LC–MS/MS
Shun-Hsin Liang and Frances Carroll,Restek Corporation

Abuse of synthetic opioid prescription painkillers such as


fentanyl, along with a rapidly growing list of illicit analogues,
is a significant public health problem. In this study, we
developed a simple dilute-and-shoot method that provides
a fast 3.5-min analysis of fentanyl and related compounds
(norfentanyl, acetyl fentanyl, alfentanil, butyryl fentanyl,
carfentanil, remifentanil, and sufentanil) in human urine by
liquid chromatography–tandem mass spectrometry (LC–MS/
MS) using a Raptor Biphenyl column.
0.00 0.20 0.40 0.60 0.80 1.00 1.20 1.40 1.60 1.80 2.00 2.20 2.40 2.60 2.80 3.00
Time (min)

In recent years, the illicit use of synthetic opioids has skyrocketed,


and communities worldwide are now dealing with an ongoing Figure 1: The Raptor Biphenyl column effectively separated all target
epidemic. Of the thousands of synthetic opioid overdose deaths compounds in urine with no observed matrix interferences. Peak
per year, most are related to fentanyl and its analogues. With elution order: norfentanyl-D5, norfentanyl, remifentanil, acetyl fentanyl-
13C , acetyl fentanyl, alfentanil, fentanyl-D , fentanyl, carfentanil-D ,
their very high analgesic properties, synthetic opioid drugs 6 5 5
carfentanil, butyryl fentanyl, sufentanil-D5, sufentanil.
such as fentanyl, alfentanil, remifentanil, and sufentanil are
potent painkillers that have valid medical applications; however, sample preparation procedure was coupled with a fast (3.5 min)
they are also extremely addictive and are targets for abuse. In chromatographic analysis using a Raptor Biphenyl column. This
addition to abuse of these prescription drugs, the current opioid method provides accurate, precise identification, and quantitation
crisis is fueled by a growing number of illicit analogues, such as of fentanyl and related compounds, making it suitable for a variety
acetyl fentanyl and butyryl fentanyl, which have been designed of testing applications, including clinical toxicology, forensic
specifically to evade prosecution by drug enforcement agencies. analysis, workplace drug testing, and pharmaceutical research.
As the number of opioid drugs and deaths increases, so does
the need for a fast, accurate method for the simultaneous analysis Experimental Conditions
of fentanyl and its analogues. Therefore, we developed this LC–
MS/MS method for measuring fentanyl, six analogues, and one Sample Preparation
metabolite (norfentanyl) in human urine. A simple dilute-and-shoot The analytes were fortified into pooled human urine. An 80 µL
urine aliquot was mixed with 320 µL of 70:30 water–
Table I: Analyte transitions methanol solution (fivefold dilution) and 10 µL of in-
ternal standard (40 ng/mL in methanol) in a Thomson
Precursor Product Ion Product Ion
Analyte Internal Standard SINGLE StEP filter vial (Restek cat. #25895). After fil-
Ion Quantifier Qualifier
Norfentanyl 233.27 84.15 56.06 Norfentanyl-D5 tering through the 0.2 µm PVDF membrane, 5 µL was
13 injected into the LC–MS/MS.
Acetyl fentanyl 323.37 188.25 105.15 Acetyl fentanyl- C6
Fentanyl 337.37 188.26 105.08 Fentanyl-D5
Calibration Standards
Butyryl fentanyl 351.43 188.20 105.15 Carfentanil-D5
and Quality Control Samples
Remifentanil 377.37 113.15 317.30 Norfentanyl-D5 The calibration standards were prepared in pooled human
Sufentanil 387.40 238.19 111.06 Sufentanil-D5 urine at 0.05, 0.10, 0.25, 0.50, 1.00, 2.50, 5.00, 10.0,
Carfentanil 395.40 113.14 335.35 Carfentanil-D5 25.0, and 50.0 ng/mL. Three levels of QC samples (0.75,
Alfentanil 417.47 268.31 197.23 Acetyl fentanyl-13C6 4.0, and 20 ng/mL) were prepared in urine for testing ac-
Norfentanyl-D5 238.30 84.15 — — curacy and precision with established calibration standard
Acetyl fentanyl-13C6 329.37 188.25 — — curves. Recovery analyses were performed on three differ-
ent days. All standards and QC samples were subjected to
Fentanyl-D5 342.47 188.27 — —
the sample preparation procedure described.
Sufentanil-D5 392.40 238.25 — —
LC–MS/MS analysis of fentanyl and its analogues was
Carfentanil-D5 400.40 340.41 — —
performed on an ACQUITY UPLC instrument coupled

362 THE APPLICATION NOTEBOOK – JUNE 2020


MEDICAL/BIOLOGICAL

with a Waters Xevo TQ-S mass spectrometer. Instrument conditions to 50 ng/mL for fentanyl, alfentanil, acetyl fentanyl, butyryl fen-
were as follows, and analyte transitions are provided in Table I. tanyl, and sufentanil; from 0.10 to 50 ng/mL for remifentanil; and
from 0.25 to 50 ng/mL for norfentanyl and carfentanil. All analytes
Analytical column: Raptor Biphenyl (5 µm, showed acceptable linearity with r2 values of 0.996 or greater and
50 mm × 2.1 mm; cat. #9309552) deviations of <12% (<20% for the lowest concentrated standard).
Guard column: Raptor Biphenyl EXP guard column
cartridge, (5 µm, 5 mm × 2.1 mm; Accuracy and Precision
cat. #930950252) Based on three independent experiments conducted on multiple
Mobile phase A: 0.1% Formic acid in water days, method accuracy for the analysis of fentanyl and its ana-
Mobile phase B: 0.1% Formic acid in methanol logues was demonstrated by the %recovery values, which were
Gradient Time (min) %B within 10% of the nominal concentration for all compounds at all
0.00 30 QC levels. The %RSD range was 0.5–8.3% and 3.4–8.4% for in-
2.50 70 traday and interday comparisons, respectively, indicating accept-
2.51 30 able method precision (Table II).
3.50 30
Flow rate: 0.4 mL/min Conclusions
Injection volume: 5 µL A simple dilute-and-shoot method was developed for the quantita-
Column temp.: 40 °C tive analysis of fentanyl and its analogues in human urine. The
Ion mode: Positive ESI analytical method was demonstrated to be fast, rugged, and sen-
sitive with acceptable accuracy and precision for urine sample
Results analysis. The Raptor Biphenyl column is well suited for the analy-
Chromatographic Performance sis of these synthetic opioid compounds, and this method can
All eight analytes were well separated within a 2.5-min gradient be applied to clinical toxicology, forensic analysis, workplace drug
elution (3.5-min total analysis time) on a Raptor Biphenyl column testing, and pharmaceutical research.
(Figure 1). No significant matrix interference was observed to neg-
atively affect quantification of the fivefold diluted urine samples.
The 5-µm particle Raptor Biphenyl column used here is a superfi-
cially porous particle (SPP) column. It was selected for this meth-
od in part because it provides similar performance to a smaller
particle size fully porous particle (FPP) column, but it generates
less system back pressure.

Linearity
Linear responses were obtained for all compounds and the calibra- Restek Corporation
tion ranges encompassed typical concentration levels monitored 110 Benner Circle, Bellefonte, PA 16823
for both research and abuse. Using 1/x weighted linear regression tel. 1 (814) 353-1300
(1/x2 for butyryl fentanyl), calibration linearity ranged from 0.05 Website: www.restek.com

Table II: Accuracy and precision results for fentanyl and related compounds in urine QC samples

QC Level 1 (0.750 ng/mL) QC Level 2 (4.00 ng/mL) QC Level 3 (20.0 ng/mL)

Average Conc. Average % Average Conc. Average % Average Conc. Average %


Analyte %RSD %RSD %RSD
(ng/mL) Accuracy (ng/mL) Accuracy (ng/mL) Accuracy
Acetyl fentanyl 0.761 102 1.54 3.99 99.7 2.08 19.9 99.3 0.856
Alfentanil 0.733 97.6 3.34 3.96 98.9 8.38 20.9 104 6.73
Butyryl fentanyl 0.741 98.9 6.29 3.77 94.3 6.01 20.8 104 4.95
Carfentanil 0.757 101 7.34 3.76 94.0 4.64 20.6 103 4.24
Fentanyl 0.761 102 1.98 3.96 99.1 2.31 19.9 99.6 1.04
Norfentanyl 0.768 103 6.50 4.04 101 1.84 20.1 101 2.55
Remifentanil 0.765 102 3.42 3.97 99.2 3.68 20.8 104 4.14
Sufentanil 0.752 100 1.67 3.93 98.3 1.28 20.1 100 0.943

THE APPLICATION NOTEBOOK – JUNE 2020 363


MEDICAL/BIOLOGICAL

Chondroitin Sulfate Analyzed by SEC–MALS


Wyatt Technology

Chondroitin sulfate is used to treat arthritis and other conditions


involving cartilage. Different preparations of the product have
different molecular properties, which are related in turn to therapeutic DŽůĂƌDĂƐƐǀƐ͘dŝŵĞ භ CSLM
^>D
ϭ͘ϬdžϭϬ
1.0x106ϲ භ CS
^
effectiveness. Such properties can be studied—and quantified—by MALS Signal
D>^^ŝŐŶĂů dRI Signal
ĚZ/^ŝŐŶĂů
light scattering.
Two chondroitin sulfate samples were compared by means of 1.0x105ϱ
ϭ͘ϬdžϭϬ
>ŽǁͲDwǁ Chondroitin
Low-M ŚŽŶĚƌŽŝƚŝŶ
a DAWN multi-angle light scattering (MALS) instrument (Wyatt Sulfate
^ƵůĨĂƚĞ

Mass (g/mol)
DŽůĂƌDĂƐƐ;ŐͬŵŽůͿ
Technology) coupled to size-exclusion chromatography (SEC). A 1.0x104ϰ
ϭ͘ϬdžϭϬ
differential refractive index (dRI) detector served to measure
dRI Signal
ĚZ/^ŝŐŶĂů
concentration. MALS and dRI measurements are combined in

Molecular
ϭ͘ϬdžϭϬ
1.0x103ϯ
ASTRA software to calculate molar mass distributions, moments
such as the weight-average molar mass Mw, and the polydispersity
index Mw/Mn. 1.0x102Ϯ
ϭ͘ϬdžϭϬ
Analysis by MALS relies on first principles and therefore
overcomes the uncertainties associated with column calibration, 1.0x101ϭ
ϭ͘ϬdžϭϬ
which arise from differences in elution properties between the 6.0
ϲ͘Ϭ 8.0
ϴ͘Ϭ 10.0
ϭϬ͘Ϭ 12.0
ϭϮ͘Ϭ 14.0
ϭϰ͘Ϭ
dŝŵĞ;ŵŝŶͿ
Time (min)
sample and reference molecules related to conformation, density,
and column interactions. SEC–MALS also eliminates the need to
perform frequent recalibration. Figure 1: Molar mass versus time for two chondroitin sulfate samples
measured by SEC–MALS. The dRI signals for both samples and the LS signal
for one sample are superimposed.
Results
Figure 1 shows chromatograms and molar masses obtained from
MALS measurements for two samples, CS and CSLM. The dRI trace Conclusions
alone is shown for CSLM in red, while both dRI and MALS signals The SEC–MALS method provides complete information about the mo-
are shown for CS, in blue. Not all of the differences between the two lar mass of chondroitin sulfate, determining distributions and a full
samples appear in standard SEC analysis, but are readily revealed set of moments. The analysis indicated similarities and discrepancies
by SEC–MALS. between the two samples. The presence of a high-molecular-weight
MALS indicates that CS has a Mw of 14.8 ± 0.2 kDa and the peak in CS, not observed by dRI, was detected unequivocally by MALS,
polydispersity index (Mw /Mn) is 1.21 ± 0.03. The low molar mass thanks to its enhanced sensitivity to high-molecular-weight species.
chondroitin sulfate LMCS sample has a Mw of 6.9 ± 0.2 kDa and a The fact that similar molar masses and sizes of each sample were
polydispersity of 1.4 ± 0.1. MALS analysis in fact determines a full eluted at the same time is a good indication of similar conformation
set of moments including Mn, Mw, and Mz, as well as cumulative and and chemical properties. Absolute macromolecular characterization
differential molar mass distributions (not shown). by SEC–MALS provides rich, detailed information without the biases
inherent in standard analytical SEC, for confidence in essential
• Two additional peaks were observed in both samples at late physiochemical properties.
elution times, though at different ratios to the main peak. The
molar masses of these species are calculated by MALS as just
a few hundred g/mol.
• In the CS sample, an early peak at 7 min is observed in the
MALS trace but is barely present in the dRI trace. This high ratio
of MALS to dRI is typical of high-molar-mass species; further
analysis shows that this peak contains a fraction with molar
mass ranging from 40–500 kDa. Wyatt Technology Corporation
6330 Hollister Ave., Santa Barbara, CA 93117
tel. (805) 681-9009
Website: www.wyatt.com

364 THE APPLICATION NOTEBOOK – JUNE 2020


FOOD/BEVERAGE

HILIC Columns for Phosphorylated Sugar Analysis


Showa Denko America, Inc.

Various types of phosphorylated saccharides are involved with


glycolysis, glyconeogenesis, glycometabolism, and other naturally
occurring biological phenomena. One example of a phosphorylated
1. Glucose-6-phosphate
saccharide includes fructose-2,6-bisphosphate, the most powerful 3 (G6P)
activator of phosphofructokinase. Recent research has shown 2. Fructose-6-phosphate
(F6P)
the phosphorylated saccharide intermediates created during 1 3. Glucose-1-phosphate
2
glyconeogenesis and glycometabolism can be used in other ways (G1P)
4. Fructose-1-phosphate
than naturally intended for a number of applications, drastically (F1P)
broadening the interest in phosphorylated saccharide. 4
Phosphorylated saccharides occupy a central position in
carbohydrate metabolism. Numerous sugar interconversions occur
when the sugars are linked to phosphates and di-phosphates.
When this occurs, the phosphorylated saccharides can function
as glycosyl donors in many transglycosylation reactions, providing 0 2 4 6 8 10 12 14 min
a variety of oligosaccharides, glycosides, and polysaccharides.
Phosphorylated sucrose and fructose have been synthesized by
the transglycosylation process in plants, glycogen in animal tis- Figure 1: The analysis of phosphorylated saccharides using the Shodex
sues, and chitin. The isolated phosphorylated sucrose has further VT-50 2D column.
shown to potentially be effective in experimental drugs. How the Column: HILICpak VT-50 2D, Column temperature: 60 ºC, Injection
new drugs interact within the body is directly affected by which volume: 5 µL, Eluent: 25 mM HCOONH4 aq./CH3CN : 80/20, Flow
carbon contains the phosphate group. Phosphorylated fructose rate: 0.3 mL/min. Detector: ESI–MS (SIM Negative: m/z 259).
has been recently suggested to be a part of the creation of ATP. Sample: 1-and 6- phosphorylated saccharides.
Phosphorylated mannose and glucose were found to be potent
inhibitors of lysosomal hydrolase endocytose by fibroblasts, plants, HPLC system was coupled with an ESI–MS (SIM negative: m/z
and bacteria. It was recently proposed that phosphorylated 259) detector.
mannose is an essential component of the recognition marker on
acid hydrolases. This was direct evidence from the phosphorylated Results
mannose’s ability to uptake forms of lysosomal enzymes. The aqueous sample containing the four phosphorylated sugars
With the knowledge of these few applications, the field has was analyzed successfully using HILIC and MS detection with the
been exponentially growing. Shodex HILICpak VT-50 2D column (Figure 1). From this single
Four variations of phosphorylate saccharides were analyzed by a run, each phosphorylated saccharide was prominently separat-
Shodex HILICpak VT-50 2D column under LC–MS conditions. The ed. The closest peaks were glucose-6-phosphate and fructose-
sample contained two phosphorylated glucose and two phosphorylated 6-phosphate, both eluted between 6 and 7 min.
fructose, each phosphorylated at a different carbon, providing different
biological functions. This analytical condition can also be used with other
detectors including reflective index (RI), evaporative light scattering
detector (ELSD), and corona charged aerosol detector (CAD).

Experimental Conditions
The analysis of glucose-6-phosphate, fructose-6-phosphate, glucose- TM

1-phosphate, and fructose-1-phosphate was accomplished using


the Shodex HILICpak VT-50 2D (2.0 mm ID × 150 mm ID, 5 µ), a
HILIC column suitable for LC–MS. The column temperature was Shodex™/Showa Denko America, Inc.
60 ºC and the flow rate was 0.3 mL/min. The eluent conditions 420 Lexington Avenue Suite 2335A, New York, NY, 10170
were 25 mM HCOONH4 aq./CH3CN = 80/20. An injection volume tel. (212) 370-0033 x109
of 5 µL of 1 µM of each sugar was used for the experiment. The Website: www.shodexhplc.com

THE APPLICATION NOTEBOOK – JUNE 2020 365


PHARMA/DRUG

Amino Acid Analysis by European Pharmacopeia


to Support Coronavirus Research
Pickering Laboratories, Inc.

Pharmaceutical companies worldwide are currently searching


for vaccines, therapeutic agents, and prophylactics to combat

Ammonia
COVID-19. The focus on biologic drugs during the current
pandemic highlights the crucial role amino acids analysis
plays in research and production of pharmaceuticals.

Lysine
Threonine
The European Pharmacopoeia (Ph. Eur.) defines requirements for

Methionine

Phenylalanine
Isoleucine
Valine
the qualitative and quantitative composition of medicines, as well

Leucine
as the tests to be carried out on medicines and on substances and
materials used in their production.

Proline
Pickering Laboratories, Inc. offers a complete solution for amino
acids analysis according to Ph. Eur.. This includes the Onyx PCX 0 40 min
10 20 30
post-column derivatization instrument, analytical columns and
GARDs, buffers, and Trione® ninhydrin reagent. The methods pre- Figure 2: Sodium chromatogram of alternative amino acids ana-
lyzed using Ph. Eur. 9.0 methods (3 ug/mL each, 50 uL injection).
sented in this application note were optimized to comply with system
suitability requirements of Ph. Eur. 9.0 methods.

Ammonia
Equipment
• Quaternary HPLC pump, autosampler, UV-vis detector
• Onyx PCX post-column derivatization system

Analytical columns and eluants


Isoleucine
Leucine
Cystine

For Sodium-based methods: High-efficiency sodium cation-exchange


column, 4.6 x 110 mm, catalog number 1154110T. Eluants: Na315,
Cysteine
Proline

Na425, Na640, RG011


For Lithium-based methods: High-efficiency lithium cation-exchange 0 10 20 30 40 50 min

column, 4.6 x 75 mm, catalog number 0354675T. Eluants: 1700–


Figure 3: Lithium chromatogram of amino acids used as reference
1125, Li365, Li375, RG003
solutions for cysteine analysis (3 ug/mL each, 50 uL injection).

Post-column reagent To make it easier to start using Pickering Laboratories methods, we offer
Trione® ninhydrin reagent chemistry kits that include an analytical column, GARD buffers, and
reagents for amino acids analysis. All parts of the kit could be ordered
individually if needed. Please contact Pickering Laboratories if you have
Ammonia

any questions regarding this application.


Serine

Alanine
Threonine

Lysine
Isoleucine
Leucine
Valine

Arginine
Cystine
Proline

Pickering Laboratories, Inc.


min
0 10 20 30 40
1280 Speca Park Way, Mountain View, CA 04043
Figure 1: Sodium chromatogram of amino acids analyzed using Ph. Eur. tel. (800) 654-3330, (650) 694-6700
9.0 methods (3 ug/mL each, 50 uL injection). Website: www.pickeringlabs.com

366 THE APPLICATION NOTEBOOK – JUNE 2020


PHARMA/DRUG

Size-Exclusion Chromatography for the Impurity


Analysis of Adeno-Associated Virus Serotypes
Stephan M. Koza and Weibin Chen,Waters Corporation

Monitoring the size heterogeneity of AAV-based gene LC system: ACQUITY™ UPLC™ H-Class Bio
therapy therapeutics is potentially important to ensure Sample temp.: 6 ˚C
consistent product quality and efficacy. We demonstrate Column temp.: 25 ˚C
that the levels of both high-molecular weight (HMW) and Injection volume: 3 µL
low-molecular weight (LMW) impurities in AAV capsid Column: XBridge™ Protein BEH SEC, 450 Å,
preparations can be separated on a Waters™ 450 Å Protein 3.5 µm, 7.8 × 300 mm
BEH SEC column for a series of AAV serotypes. Fluorescence
detector: Excitation: 280 nm; Emission: 350 nm
As the development of gene therapy products accelerates, the Mobile phase: 10 mM NaH2PO4, 10 mM Na2HPO4,
need to develop sound and efficient analytical strategies to 200–400 m KCl, pH 6.6 (HCl)
help guide the development of manufacturing processes and Flow rate: 0.6 mL/min
evaluate the quality of clinical adeno-associated virus-(AAV)–
based gene therapy materials has become more important. Results and Discussion
Among other critical quality attributes, the levels of potential The upper analyte size limitation for SEC separations of
AAV high molecular weight (HMW) species and AAV fragments proteins is approximately 100–200 nm (subvisible) depending
or low molecular weight (LMW) species may also require on SEC particle size. Above these limits, the analyte may be
monitoring (1). Here we present optimized non-denaturing altered due to shear forces or trapped by the frits or packed
size-exclusion chromatography (SEC) methods that can bed of the column. Therefore, these subvisible entities are
separate soluble AAV for several AAV-CMV-GFP serotypes analyzed using dynamic light scattering and nanoparticle
including AAV1, AAV2, AAV5, AAV6, AAV8, and AAV9. tracking analysis (NTA) methods, among others (2). With these
considerations, the SEC separation of soluble AAV monomer,
Experimental Conditions and HMW, and LMW was evaluated on a 450 Å pore-size
AAV sample: AAV serotypes ranging from 1.6 × 1012 to BEH diol-bonded SEC column. This column was previously
6.7 × 1013 GC/mL demonstrated to be effective in the separation of IgM pentamer

ACQUITY UPLC Protein BEH SEC, 450 Å, 2.5 µm, 4.6 x 300 mm Column
0.05
Flow rate: 0.35 mL/min 5
0.045 Injection volume: 5 µL
0.04 4
0.035
UV (280 nm)

0.03

0.025 3
0.02
2
0.015 1a
0.01 1b
0.005

0
0 0.5 1 1.5 2 2.5 3 3.5 4 4.5 5 5.5 6 6.5 7 7.5 8 8.5 9.5 10 10.5 11 11.5 12 12.5
Minutes

Figure 1: Shown is the SEC separation of several protein structures. Compounds are: 1a. IgM dipentamer (1.8 kDa), 1b. IgM pentamer (900 kDa), 2.
thyroglobulin (667 kDa), 3. apoferritin (443 kDa), 4. β-amylase (200 kDa), 5. IgG (150 kDa). UV absorbance (280 nm) and the identities of the peaks observed
for the IgM sample were confirmed by SEC–MALS analysis.

THE APPLICATION NOTEBOOK – JUNE 2020 367


PHARMA/DRUG

and dipentamer in Waters laboratories (Figure 1) and has a


reported diameter of 35 nm (3). AAV has a total protein and 1.0x107
7

ssDNA molecular weight of 5500 kDa; however, the compact RI


RI
MALS
structure has a diameter of only 25 nm (4). Therefore, it 8
8.0x108
Molar Mass
Molar Mass
was predicted that these high-efficiency SEC particles could

(g/mol)
provide the accessible pore volume needed. A 3.5 µm particle

Mass (g/mol)
6.0x1066
6.9 E6
6.9 E6 (± 0.3 E6)
E6)
size was selected over 2.5 µm particles to reduce potential

MolarMass
sample sieving and shearing effects. Intrinsic protein

(V)
voltage (V)
6
0.15
0.15

detectorvoltage
4.0x106

Molar
fluorescence detection was also employed to provide maximum 0.10
0.10
3.8
3.8 E6
E6 (±
(± 0.03 E6)
optical sensitivity.

detector
0.05
0.05

6
2.0x106
The SEC separation of a null control sample (AAV without
0.00
0.00
3.0 4.0 5.0 6.0 7.0
7.0 8.0 9.0
Minutes
Minutes

ssDNA) was evaluated using a Wyatt microDAWN Multi-Angle


Light Scattering (MALS) detector and a Waters ACQUITY 6.0 6.5 7.0
Minutes
Minutes
Refractive Index (RI) detector (Figure 2). The SEC–MALS data
confirmed that adequate separation between the dimeric and
Figure 2: SEC-MALS of AAV8-null sample using refractive index (RI) for
monomeric AAV forms was observed. Putative multimeric forms concentration measurement are shown. The MALS (red) and RI (blue)
preceding dimer were also observed by fluorescence but could signals are normalized and the average and distribution of determined
not be assigned molecular weights due to their low abundance. molar masses (green) were determined using Wyatt Astra (v. 7.3.1.9) based
on a dn/dc of 0.185 and using a “sphere” model for the icosahedral AAV.
A mobile-phase consisting of 20 mM sodium phosphate,
pH 6.6, with varying amounts of KCl (200–400 mM) was
found to maximize the recovery of dimer and multimer for 0.60
AAV1-GFP AAV6-GFP
30.00

serotypes AAV1, AAV2, AAV5, AAV6, AAV8, and AAV9 (Figure (200 mM KCl) 20.00
0.60
(350 mM KCl)
30.00

20.00

3). We observe that the peak shapes for both the dimer 0.40 10.00

D=1.6%
10.00
0.40
0.00 0.00

HMW species and monomeric AAV forms are symmetrical


0.00 5.00 10.00 15.00 20.00 25.00
0.00 5.00 10.00 15.00 20.00 25.00

0.20 0.20
M=0.5%
F=0.2%
and return to baseline appropriately. In addition, detectable D=0.2%
0.00
0.00

levels of multimeric AAV forms were observed for several of 1.00


AAV2-GFP AAV8-GFP
the serotypes. Significant amounts of LMW forms were only
40.00

(350 mM KCl)
100.00
0.80 2.00
(400 mM KCl)D=1.5%
D=1.1%
20.00
50.00
0.60

observed in AAV9 and AAV6.


FLR (relative)

0.00 M=1.5% 0.00

0.40 0.00 5.00 10.00 15.00 20.00 25.00


1.00
0.00 5.00 10.00 15.00 20.00 25.00

0.20

Conclusions 0.00
0.00

-0.20

A BEH SEC column with an average pore size of 450 Å


AAV5-GFP 20.00

AAV9-GFP 100.00

and a particle diameter of 3.5 µm was demonstrated to be 0.40


(250 mM KCl) 2.00
(250 mM KCl) D= 1.5%
15.00 80.00

10.00 60.00

D=1.2%
40.00
5.00

effective in the separation of AAV monomers from their HMW 0.20


0.00

0.00 5.00 10.00 15.00 20.00 25.00

1.00
20.00

0.00

M=0.9%
0.00 5.00 10.00 15.00 20.00 25.00

F=0.8%
dimers, lower valency multimers, and LMW fragments. The M=0.3%
F=0.3%

minimal amount of ionic strength (KCl) required for optimal 0.00 0.00

peak shape and recovery varied by serotype, and separations 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00

Minutes Minutes
are presented for serotypes AAV1, AAV2, AAV5, AAV6, AAV8,
and AAV9. Figure 3: Shown are the SEC separations of a series of AAV serotype
control samples containing ssDNA coding for green fluorescent protein
(GFP). The peak percentages for dimer (D), multimer (M), and fragments (F)
References
are provided. The chromatogram baselines are zoomed approximately 50x
(1) J. F. Wright, Gene Therapy 15(11), 840–8 (2008). versus the full-scale chromatogram shown in the inset.
(2) (a) J. F. Carpenter, T. W. Randolph, W. Jiskoot, D. J. Crommelin, C. R.
Middaugh, G. Winter, Y. X. Fan, S. Kirshner, D. Verthelyi, S. Kozlowski, K.
A. Clouse, P. G. Swann, A. Rosenberg, and B. Cherney, J Pharm Sci 98(4),
1201–5 (2009); (b) B. Slutter, and W. Jiskoot, Expert Opin. Drug Delivery
13(2), 167–70 (2016).
(3) R. Saber, S. Sarkar, P. Gill, B. Nazari, and F. Faridani, Sci. Iran. 18(6),
1643–1646 (2011).
(4) S. Vernon, J. Stasny, A. Neurath, and B. Rubin, J. Gen. Virol. 10(3), 267–
272 (1971). Waters Corporation
34 Maple Street, Milford, MA 01757
Waters, The Science of What’s Possible, ACQUITY, UPLC, and XBridge are trademarks tel. 508 478 2000
of Waters Corporation. All other trademarks are the property of their respective owners. Website: www.waters.com

368 THE APPLICATION NOTEBOOK – JUNE 2020


PHARMA/DRUG

An Anion-Exchange Chromatography Method


for Monitoring Empty Capsid Content in
Adeno-Associated Virus Serotype AAV8
Hua Yang, Stephan M. Koza, and Weibin Chen,Waters Corporation

The single-stranded DNA (ssDNA) content of AAV capsids in Fluorescence


AAV-based gene therapy preparations impacts the efficacy detector: Excitation: 280 nm; Emission: 350 nm
of this treatment modality. We demonstrate that AAV8 Gradient: 70 mM bis-tris propane, pH 9.0, 100–300 mM
capsids without and with full length ssDNA can be separated tetramethylammonium chloride in 20 min
and their relative abundances determined on a Waters™ at 0.4 mL/min
Protein-Pak™ Hi Res Q strong anion-exchange column.
Results and Discussion
During the recombinant adeno-associated virus (AAV) The AEX separation of empty and full AAV8 capsid was optimized
biomanufacturing process, capsids that do not contain for several parameters including pH and salt types (3). In addition,
ssDNA (empty capsids) are also produced. Empty capsids are intrinsic protein fluorescence was monitored to provide greater
unable to deliver ssDNA to the targeted cells and as a result sensitivity and selectivity, and less baseline drift (2). The separation
are important to monitor during process development and achieved is shown in Figure 1. We observe that the ssDNA “empty”
product manufacturing. An anion-exchange chromatography capsids are partially coelute with the ssDNA “full” capsids while
(AEX) separation driven by differences in surface charge can showing no significant change in the UV absorbance ratio (280
monitor changes in empty capsid levels (1,2). AEX using nm/260 nm, data not shown), indicating the unlikelihood of
fluorescent detection consumes small sample amounts and monitoring AAV8 with partial ssDNA by AEX.
can provide reliable and reproducible results. A series of chromatograms for AAV8 samples with differing
percentages of empty capsids is presented in Figure 2. The
Experimental Conditions mixtures were generated from >99% pure “empty” and “full”
Samples: AAV8-CMV-GFP (full capsid) and AAV8-null (empty capsid) samples. The sample concentrations were normalized based
2 × 1012 capsids/mL; other AAV-CMV-GFP serotypes: ranging from on FLR peak areas from a size-exclusion chromatography
1.3 × 1013 to 6.7 × 1013 GC/mL = genome copies (physical titer)/mL (SEC) separation with fluorescence detection and assumed a
“full” to “empty” fluorescence response factor (RF F/E) of 1.3 (2).
LC system: ACQUITY™ UPLC™ H-Class Bio For AEX, however, 1.9 was calculated for RF F/E (Area 100% Full/
Sample temp.: 10 ˚C Area 100% Empty). The discrepancy in RF F/E values between SEC
Column temp.: 30 ˚C and AEX may be the result of changes in quantum efficiency
Injection volume: 0.2–6 µL under different mobile-phase conditions. RF F/E is then divided
Column: Waters Protein-Pak Hi Res Q, 4.6 × 100 mm into the full capsid peak area to calculate the percentage of

20.00
EU

Empty/full capsid mix


10.00

0.00

20.00
EU

Empty capsid
10.00

0.00

20.00
EU

Full capsid
10.00

0.00
8.00 10.00 12.00 14.00 16.00 18.00 20.00
Minutes
Figure 1: AAV8 ssDNA (CMV-GFP) empty and full capsids are separated on a Protein-Pak Hi Res Q Column using an optimized AEX method (see
experimental conditions for method details).

THE APPLICATION NOTEBOOK – JUNE 2020 369


PHARMA/DRUG

24.00 100
0% Empty
22.00 90 100 x A Empty
12.5% Empty % Empty
80 RF F/E

Measured % Empty
20.00 25% Empty (A Empty +
18.00 50% Empty 70 A Full
16.00
75% Empty 60
100% Empty 50
14.00
40
EU

12.00
30
10.00
20
8.00 y = 0.9264x + 3.2146
10 R = 0.9997
6.00
0
4.00 0 20 40 60 80 100
2.00 Predicted % Empty
0.00

8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00


Minutes

Figure 2: Shown on the left is an overlay of AEX chromatograms for AAV8 samples with differing levels of empty capsid. On the right is the
correlation between predicted and measured empty capsid content. Calculation of the percentage of empty capsid was based on the equation
shown, which corrects for the difference in fluorescence response between empty and full capsids. AEmpty and AFull are the AEX peak areas of the
empty and full capsids, and RFF/E is the fluorescence response factor.

empty capsid in a sample (Figure 2). The accuracy of this


approach depends on the veracity of RF F/E, which requires
4.00
reasonably pure “full” and “empty” samples, and accurate AAV1
EU

2.00

capsid concentrations. While RF F/E was not rigorously defined 0.00

4.00

in this work, the estimate of RF F/E used here effectively


EU

AAV2
2.00

demonstrates the approach to interpretation of the AEX data. 0.00

Determining RF F/E is not essential, if only relative comparisons 2.00


EU

AAV5
of empty capsid levels between samples are required. 0.00
4.00

Samples of several AAV-CMV-GFP serotypes were also


AAV6
EU

2.00

evaluated (Figure 3). All tested serotypes were retained on 0.00

the column, but their retention varied significantly. 10.00


EU

AAV8
0.00

Conclusions
AAV9*
EU

5.00
AEX will not likely provide a measurement of AAV8 capsid
0.00
carrying partial ssDNA, therefore complementary methods F
E
10.00
EU

such as analytical ultracentrifugation (AUC) would be needed AAV E/F mix


0.00
for that analysis. However, due to the technical challenges 2.00 4.00 6.00 8.00 10.00 12.00 14.00
Minutes
16.00 18.00 20.00 22.00 24.00 26.00

and costs of these methods, AEX may still provide utility as


an initial screening tool in support of manufacturing process Figure 3: Chromatograms of AAV1, AAV2, AAV5, AAV6, AAV8,
and AAV9 serotypes (full capsids, CMV-GFP ssDNA). For comparison,
development and as a more precise assessment of changes in
the bottom chromatogram shows the optimized separation of AAV8
empty capsid content in product quality testing. empty and full capsid. Separation conditions are as described in the
Due to differences in capsid protein surfaces, method text for all serotypes except for AAV9 (*) where the salt gradient was
optimization may be needed when separating empty and 0–200 mM KCl.
full capsids of other AAV serotypes, and the AEX method
described here can serve as a useful starting point.

References
(1) X. Fu, et al, Hum. Gene. Ther. Methods 30(4), 144 (2019).
(2) C. Wang, et al, Mol. Ther. Methods Clin. 19, 257 (2019).
(3) H. Yang, et al, Waters Application Note 720006825EN (2020).
Waters Corporation
Waters, The Science of What’s Possible, Protein-Pak, ACQUITY, and UPLC are 34 Maple Street, Milford, MA 01757
trademarks of Waters Corporation. All other trademarks are the property of their tel. 508 478 2000
respective owners. Website: www.waters.com

370 THE APPLICATION NOTEBOOK – JUNE 2020


WWW.CHROMATOGRAPHYONLINE.COM JUNE 2020  LCGC NORTH AMERICA  VOLUME 38 NUMBER 6  371

FUN DA MENTAL S

Troubleshooting Gas Chromatography:


Reduced Peak Size (Loss of Sensitivity)
Tony Taylor

W e are frequently asked about issues


with reduced peak size in gas chro-
matography (GC). There are so many poten-
check that the fuel gas ratios are appro-
priate and that all flow rates are as they
should be (using a flow meter, checking
pressure will be ramped to achieve con-
stant linear velocity of carrier through the
column as the oven temperature increases
tial causes for reduced peak size that an inex- one gas flow at a time), and check all and the carrier gas viscosity reduces.
perienced GC user may not know where to applied voltages according to the manu- • Change the inlet septum to overcome any
begin in the troubleshooting process. Sen- facturers specification. leaks that may be occurring during the
sitivity problems can be grouped by way of • With MS detectors in selected ion mode, injection phase.
their presentation within the chromatogram: check the masses and dwell times of each
ion within each SIM group to ensure that All peaks sizes (heights and or areas)
All peaks sizes (heights and or areas) the cumulative ion count is matched to decrease–peaks are broadened, retention
decrease–retention times do not change. the acquisition method. time shift may occur.
As always, it’s best to start with the most • With MS detectors in scan mode, check • Check that the correct column has been
obvious possible causes.. the MS tune and verify that the repeller installed and column dimensions and car-
• If operating in split mode, check the inlet or accelerator voltage has not increased rier gas flow rate are appropriate.
split ratio in the acquisition method and dramatically (indicating a dirty ion source), • Check the column logs; if the column is
correct, if required. that the electron multiplier or MCP volt- old or has been used with dirty sample
• If splitless mode with pressure pulse, ages have not increased dramatically (indi- matrices, suspect that the column effi-
check the inlet pulse pressure and dura- cating a worn-out detector), and that the ciency might have been reduced.
tion in the acquisition method. ionization energy is correctly set (typically • Run a column test mix and compare with
• Check that the inlet and detector temper- 70 eV for electron ionization). the result obtained originally.
atures have been set correctly within the • Check the detector attenuation range • If loss of column efficiency is suspected,
acquisition method. and set appropriately, as a secondary trim 0.5–1 meters of column at the
• Check that the sample vial contains check. When the detector attenuation is inlet end.
sufficient liquid and use separate vials incorrectly set, the signal-to-noise ratio • Verify that the column has been installed
containing the same sample liquid for for a given peak will not reduce, but the to the correct distance within the GC inlet
repeated injections to rule out the pos- chromatogram will appear ”smaller.” and detector. For flame-based and ioniz-
sibility of sample loss through a compro- ing detectors, check that the gas flows are
mised septum (especially when analytes All peaks sizes (heights and areas) decrease. correct paying particular attention to any
are highly volatile), and confirm the prob- Retention times shift, no evidence of loss make-up gas flow rates.
lem is reproducible. of efficiency (peak broadening) • When all peak heights or areas reduce
• Check that the autosampler syringe • Check that the correct column dimensions and the peaks broaden, the most obvious
plunger is free within the barrel and does have been entered into the data system cause is a loss of efficiency within the chro-
not leak. Observe an injection cycle and and that the correct column is installed, matographic system. This phenomenon
Icon image: Monster Ztudio/Adobe Stock

confirm that sample is being aspirated paying attention to the stationary-phase should also lead a reduction in signal-to-
from the vial and that it the correct volume. film thickness. noise ratio for analyte peaks.
• Check and replace the inlet septum as • Check that the carrier gas is flowing at the
required, and check that the correct liner correct volumetric flow rate using a cali- Tony Taylor is the Chief Sci-
has been installed. brated flow meter. ence Officer of Arch Sciences
Group and the Technical
• Check that the sample preparation or dilu- • Check that the carrier gas flow program-
Director of CHROMacademy.
tions are correct as per the sample prepa- ming method is correct by selecting either Direct correspondence to:
ration method. constant pressure or constant flow oper- LCGCedit@mmhgroup.com.
• With flame based ionizing detectors, ating modes. In the latter, the carrier gas

You might also like