58
Volume 36 Number 11 November 2018 www.chromatographyonline.com The Analytical Toolbox for Biopharmaceutical Characterization Understanding Stationary-Phase Selectivity for GC INSTRUMENT CONSIDERATIONS IN THE TRANSFER OF CHROMATOGRAPHIC METHODS

Volume 36 Number 11 November 2018  · UBM Americas () is a leading worldwide media com-pany providing integrated marketing solutions for the Fashion, Life Sciences and Powersports

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

V

olu

me

36

Nu

mb

er 1

1, 7

85

–8

40

LC

GC

NO

RT

H A

ME

RIC

A

No

ve

mb

er 2

018

Volume 36 Number 11 November 2018

www.chromatographyonline.com

The Analytical Toolbox for Biopharmaceutical Characterization

Understanding Stationary-Phase

Selectivity for GC

INSTRUMENT

CONSIDERATIONS IN

THE TRANSFER OF

CHROMATOGRAPHIC

METHODS

ES39215_LCGC1118_CV1.pgs 11.05.2018 14:54 UBM blackyellowmagentacyan

Feel good in your method development?

Feel Better with UsTrust our analytical products in every stage of your drug

product development. Whether you’re working on bioanalytical

/&�06��IRUPXODWLRQ��RU�¿QDO�TXDOLW\�FRQWURO�DQG�UHOHDVH��ZH�

have the expertise you need.

)HHO�EHWWHU�ZLWK�IXOO�ZRUNÀRZ�VROXWLRQV�DW

SigmaAldrich.com/feel-better

The life science business of Merck KGaA, Darmstadt, Germany

operates as MilliporeSigma in the U.S. and Canada.

j������0HUFN�.*D$��'DUPVWDGW��*HUPDQ\�DQG�RU�LWV�DɡOLDWHV��$OO�5LJKWV�5HVHUYHG��0LOOLSRUH6LJPD�DQG�WKH�YLEUDQW�0�DUH�WUDGHPDUNV�RI�0HUFN�.*D$��'DUPVWDGW��*HUPDQ\�RU�LWV�DɡOLDWHV��$OO�RWKHU�WUDGHPDUNV�DUH�WKH�SURSHUW\�RI�WKHLU�UHVSHFWLYH�RZQHUV��'HWDLOHG�LQIRUPDWLRQ�RQ�WUDGHPDUNV�LV�DYDLODEOH�YLD�SXEOLFO\�DFFHVVLEOH�UHVRXUFHV����

2018-14251

www.shodexHPLC.com

SUGAR SH1011 8C- Separates via a combination of

ligand exchange and size

exclusion chromatography

- Save money and time with simple

aqueous eluents

SUGAR SH1011 8C- New 8.0 x 100 mm size

- Analyze sugars, organic

acids, and alcohols in under

5 min

- Ideal for fermentation

monitoring and food samples

Rapid Sugar Series : SH1011 8C

6DPSOH�����/��������Z�Y��HDFK

1. Maltotriose

2. Maltose

3. Glucose

���/DFWLF�$FLG

5. Glycerol

6. Acetic Acid

7. Ethanol

788 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

485F US Highway One South, Suite 210 Iselin, NJ 08830

(732) 596-0276 • Fax: (732) 647-1235

UBM Americas (www.ubmlifesciences.com) is a leading worldwide media com-pany providing integrated marketing solutions for the Fashion, Life Sciences and Powersports industries. UBM Americas serves business professionals and consumers in these industries with its portfolio of 91 events, 67 publications and directories, 150 electronic publications and Web sites, as well as educational and direct market-ing products and services. Market leading brands and a commitment to delivering innovative, quality products and services enables UBM Americas to “Connect Our Customers With Theirs.” UBM Americas has approximately 1000 employees and currently operates from multiple offices in North America and Europe.

© 2018 UBM All rights reserved. No part of this publication may be reproduced or trans-

mitted in any form or by any means, electronic or mechanical including by photocopy,

recording, or information storage and retrieval without permission in writing from the

publisher. Authorization to photocopy items for internal/educational or personal use,

or the internal/educational or personal use of specifi c clients is granted by UBM for

libraries and other users registered with the Copyright Clearance Center, 222 Rosewood

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

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

Permission Dept. fax 732-647-1104 or email: [email protected]

UBM Americas provides certain customer contact data (such as customer’s name, ad-

dresses, phone numbers, and e-mail addresses) to third parties who wish to promote

relevant products, services, and other opportunities that may be of interest to you. If you

do not want UBM Americas to make your contact information available to third parties

for marketing purposes, simply call toll-free 866-529-2922 between the hours of 7:30

a.m. and 5 p.m. CST and a customer service representative will assist you in removing

your name from UBM Americas lists. Outside the U.S., please phone 218-740-6477.

LCGC North America does not verify any claims or other information appearing in any

of the advertisements contained in the publication, and cannot take responsibility for

any losses or other damages incurred by readers in reliance of such content.

To subscribe, call toll-free 888-527-7008. Outside the U.S. call 218-740-6477.

MANUSCRIPTS: For manuscript preparation guidelines, see chromatographyon-

line.com/lcgc-author-guidelines, or call The Editor, (732) 596-0276. LCGC welcomes

unsolicited articles, manuscripts, photographs, illustrations, and other materials

but cannot be held responsible for their safekeeping or return. Every precaution is

taken to ensure accuracy, but LCGC cannot accept responsibility for the accuracy

of information supplied herein or for any opinion expressed.

SUBSCRIPTIONS: For subscription and circulation information: LCGC, P.O. Box

6168, Duluth, MN 55806-6168, or call (888) 527-7008 (7:00 a.m.–6:00 p.m. central

time). International customers should call +1-218-740-6477. Delivery of LCGC out-

side the United States is 14 days after printing. For single and back issues, call (800)

598-6008 or (218) 740-6480. (LCGC Europe and LCGC Asia Pacific are available free

of charge to users and specifiers of chromatographic equipment in Western Europe

and Asia and Australia, respectively.)

CHANGE OF ADDRESS: Send change of address to LCGC, P.O. Box 6168, Duluth,

MN 55806-6168; alternately, send change via e-mail to [email protected] or go to the

following URLs:

• Print: http://ubmsubs.ubm.com/?pubid=LCGC

• Digital: http://ubmsubs.ubm.com/?pubid=LCGC&V=DIGI

Allow four to six weeks for change. PUBLICATIONS MAIL AGREEMENT

No. 40612608. Return all undeliverable Canadian addresses to:

IMEX Global Solutions, P.O. Box 25542, London, ON, N6C 6B2,

CANADA. Canadian GST number: R-124213133RT001.

C.A.S.T. DATA AND LIST INFORMATION: Contact Melissa Stillwell,

tel. (218) 740-6831, e-mail [email protected].

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

are available (500 minimum). Licensing and Reuse of Content: Contact

our official partner, Wright’s Media, about available usages, license

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

information. Please note that Wright’s Media is the only authorized

company that we’ve partnered with for Advanstar UBM materials.

INTERNATIONAL LICENSING: Contact Jillyn Frommer, tel.

(732) 346-3007, fax 732-647-1104, or e-mail [email protected].

50

% Recycled Paper 1

0-2

0%

Post Consumer W

as

te

Michael J. Tessalone

Vice President/Group Publisher [email protected]

Edward Fantuzzi

Publisher [email protected]

Stephanie Shaffer

Sales Manager [email protected]

Brianne Molnar

Sales Manager [email protected]

Michael Kushner

Senior Director, Digital [email protected]

Kristen Moore

Webcast Operations Manager [email protected]

Vania Oliveira

Project Manager [email protected]

Sabina Advani

Digital Production Manager [email protected]

Kaylynn Chiarello-Ebner

Managing Editor, Special Projects [email protected]

Brianne Pangaro

Marketing [email protected]

Melissa Stillwell

C.A.S.T. Data and List Information [email protected]

Laura Bush

Editorial Director [email protected]

John Chasse

Managing Editor [email protected]

Jerome Workman, Jr.

Senior Technical Editor [email protected]

Cindy Delonas

Associate Editor [email protected]

Dan Ward

Art Director [email protected]

Rajesh Thangappan

Graphic Designer [email protected]

Wright’s Media

Reprints [email protected]

Jillyn Frommer

Permissions [email protected]

Jesse Singer

Production Manager [email protected]

Wendy Bong

Audience Development Manager [email protected]

Morgan Hight

Audience Support Analyst [email protected]

Thomas W. Ehardt

Executive Vice-President,Senior Managing Director UBM Life Sciences [email protected]

Dave Esola

VP & General ManagerUBM Life Sciences [email protected]

Chro

mato

gra

phy

Find what you are looking for!

MACHEREY-NAGEL

Manufacturer of premium

chromatography media

www.mn-net.com

� CHROMABOND® columns with classical and

innovative SPE phases

� Original NUCLEOSIL®, professional NUCLEODUR®

and highly efficient NUCLEOSHELL® HPLC columns

� Robust TLC glass plates, economical aluminum and

polyester sheets

790 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

Volume 36 Number 11 November 2018

www.chromatographyonline.com

The Analytical Toolbox for Biopharmaceutical Characterization

Understanding Stationary-Phase

Selectivity for GC

INSTRUMENT

CONSIDERATIONS IN

THE TRANSFER OF

CHROMATOGRAPHIC

METHODS

C O N T E N T S

LCGC North America (ISSN 1527-5949 print) (ISSN 1939-1889 digital) is published monthly by UBM LLC, 131 West First Street, Duluth, MN 55802-2065, and is distributed free of charge to users and specifi ers of chromatographic equipment in the United States and Canada. Single copies (prepaid only, including postage and handling): $15.50 in the United States, $17.50 in all other countries; back issues: $23 in the United States, $27 in all other countries. LCGC is available on a paid subscription basis to nonqualifi ed readers in the United States and its possessions at the rate of: 1 year (13 issues), $74.95; 2 years (26 issues), $134.50; in Canada and Mexico: 1 year (13 issues), $95; 2 years (26 issues), $150; in all other countries: 1 year (13 issues), $140; 2 years (26 issues), $250. Periodicals postage paid at Duluth, MN 55806 and at additional mailing offi ces. POSTMASTER: Please send address changes to LCGC, P.O. Box 6168, Duluth, MN 55806-6168. 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.

COLUMNS

796 LC TROUBLESHOOTINGMixing and Mixers in Liquid Chromatography—Why, When, and How Much?

Part II, Injections

Dwight R. Stoll

What happens when we inject a sample into the mobile-phase stream? Many LC practitioners are surprised to learn just how serious the effect of the injected sample solvent can be.

802 SAMPLE PREP PERSPECTIVESA Look Back and A Look Forward—An Annual Check-up on the State of

Sample Preparation

Douglas E. Raynie

We assess the state of the field, first looking back at developments presented at conferences this year, reader questions, and the passing of a pioneer in solid-phase extraction. Then, we look to the future of sample preparation.

806 GC CONNECTIONSStationary Phase Selectivity: The Chemistry Behind the Separation

Nicholas H. Snow

Here, we focus on selectivity: its definition, its importance for generating separations and resolution; and its role in column polarity.

814 FOCUS ON BIOPHARMACEUTICAL ANALYSISAnalytical Characterization of Biotherapeutic Products, Part II:

The Analytical Toolbox

Anurag S. Rathore, Ira S. Krull, and Srishti Joshi

The analytical techniques used for characterizing biotherapeutics have evolved. We review the utility of the traditional tools and discuss the new, orthogonal techniques that are increasingly being used.

838 THE ESSENTIALSHPLC Column Maintenance: Tips for Extending HPLC Column Lifetime

Follow these tips to protect your columns and extend their useful lifetime.

PEER-REVIEWED ARTICLES

824 Instrument Considerations in the Transfer of Chromatographic Methods,

Part II: System Considerations

Thomas E. Wheat

Scientists executing a method transfer often do not have access to the originating system. Thus, alternative approaches to matching chromatographic results must be considered.

830 Chromatography Fundamentals, Part V:

Theoretical Plates: Significance, Properties, and Uses

Howard G. Barth

The number of theoretical plates forms the basis of chromatographic theory, and is a key parameter used in all modes of chromatography for measuring column efficiency. Fortunately, it’s easy to measure.

COVER DESIGN BY Dan Ward

Cover image courtesy of Ioana Davies (Drutu) /

stock.adobe.com

DEPARTMENTS

794 Peaks of Interest

836 Products & Resources

837 Ad Index

VICI Metronics Dynacalibrators® facilitate

accuracy of analytical data from air pollution

monitoring, industrial hygiene surveys, odor

surveys, and other instruments measuring

gas concentration.

In the lab or in the field, all models support

calibrations traceable to NIST standards for

almost any gas analyzer.

The convenient design of our Dynacal®

permeation devices helps to generate and

deliver precise concentrations from ppb to

high ppm, for hundreds of different

compound options.

Base configurations can be customized for

dilution gas and carrier gas flow capacities.

• Dynacal® permeation devices as trace

gas source

• Dependable, accurate and replicable

calibrations

• Proprietary temperature control system,

chamber temperature 0.01˚ C accuracy

• Front panel access to permeation

chamber

• Plumbing and flow configurations

• Manual and automated versions

• CE-Certified

Metronics

®

Setup

Tubes

Auto

Manual

Timed

Graph

Status

Set Point Actual Deviation

Temp. 1

Temp. 2

Flow 1

Flow 2

Flow dil.

Valve 1

Valve 2

deg. C

deg. C

cc/min.

cc/min.

L/min.

Equilibration Time

46.0

58.0

100.0

100.0

1.0

INLINE

INLINE

38.0

0.0

0.0

0.0

0.0

INLINE

INLINE

-8.0

-58.0

-100.0

-100.0

-1.0

0

0

min:sec10:00

PPB: Mode: Manual- - - -

For Sales inquiresin U.S. :[email protected]

792 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

Editorial Advisory Board• Kevin D. Altria – GlaxoSmithKline, Ware, United Kingdom• Jared L. Anderson – Iowa State University, Ames, Iowa • Daniel W. Armstrong – University of Texas, Arlington, Texas • David S. Bell – Restek, Bellefonte, Pennsylvania• Dennis D. Blevins – Agilent Technologies, Wilmington, Delaware• Zachary S. Breitbach – AbbVie Inc., North Chicago, Illinois• Deirdre Cabooter – Department of Pharmaceutical and Pharmacological

Sciences, KU Leuven (University of Leuven), Belgium• Peter Carr – Department of Chemistry,

University of Minnesota, Minneapolis, Minnesota• Jean-Pierre Chervet – Antec Scientific, Zoeterwoude, The Netherlands• André de Villiers – Stellenbosch University, Stellenbosch, South Africa• John W. Dolan – LC Resources, McMinnville, Oregon• Michael W. Dong – MWD Consulting, Norwalk, Connecticut• Anthony F. Fell – School of Pharmacy,

University of Bradford, Bradford, United Kingdom• Francesco Gasparrini – Dipartimento di Studi di Chimica e Tecnologia

delle Sostanze Biologicamente Attive, Università “La Sapienza,” Rome, Italy• Joseph L. Glajch – Momenta Pharmaceuticals, Cambridge, Massachusetts• Davy Guillarme – University of Geneva,

University of Lausanne, Geneva, Switzerland• Richard Hartwick – PharmAssist Analytical Laboratory, Inc.,

South New Berlin, New York• Milton T.W. Hearn – Center for Bioprocess Technology,

Monash University, Clayton, Victoria, Australia• Emily Hilder – University of South Australia, Adelaide, Australia• John V. Hinshaw – Serveron Corporation, Beaverton, Oregon• Kiyokatsu Jinno – School of Materials Science,

Toyohashi University of Technology, Toyohashi, Japan• Ira S. Krull – Professor Emeritus, Department of Chemistry and

Chemical Biology, Northeastern University, Boston, Massachusetts

• Ronald E. Majors – Analytical consultant, West Chester, Pennsylvania• Debby Mangelings – Department of Analytical Chemistry and

Pharmaceutical Technology, Vrije Universiteit Brussel, Brussels, Belgium• R.D. McDowall – McDowall Consulting, Bromley, United Kingdom• Michael D. McGinley – Phenomenex, Inc., Torrance, California• Victoria A. McGuffin – Department of Chemistry,

Michigan State University, East Lansing, Michigan• Mary Ellen McNally – FMC Agricultural Solutions, Newark, Delaware• Imre Molnár – Molnar Research Institute, Berlin, Germany• Glenn I. Ouchi – Brego Research, San Jose, California• Colin Poole – Department of Chemistry,

Wayne State University, Detroit, Michigan• Douglas E. Raynie – Department of Chemistry and Biochemistry,

South Dakota State University, Brookings, South Dakota• Fred E. Regnier – Department of Chemistry, Purdue University,

West Lafayette, Indiana• Koen Sandra – Research Institute for Chromatography, Kortrijk, Belgium• Pat Sandra – Research Institute for Chromatography, Kortrijk, Belgium• Peter Schoenmakers – Department of Chemical Engineering,

University of Amsterdam, Amsterdam, The Netherlands• Kevin Schug – University of Texas, Arlington, Texas• Dwight Stoll – Gustavus Adolphus College, St. Peter, Minnesota• Michael E. Swartz – Stealth Biotherapeutics, Newton, Massachusetts • Caroline West – University of Orléans, France • Thomas Wheat – Chromatographic Consulting, LLC, Hopedale, Massachusetts• Taylor Zhang – Genentech, South San Francisco, California

CONSULTING EDITORS: Jason Anspach – Phenomenex, Inc.; David Henderson – Trinity College; Tom Jupille – LC Resources; Sam Margolis – The National Institute of Standards and Technology; Joy R. Miksic – Bioanalytical Solutions LLC

Pure Chromatography

ADVANTAGESee What It Can Do for You and Your Lab

Sign up today to access Restek’s years of chromatography knowledge at

www.restek.com/advantage

What can we do for you?

www.gerstel.com

(800) 413-8160sa les@gers te lus .com

Thin Film SPME

TICs of a wine using SPME (top) and TF-SPME (bottom).

Taking SPME to the next level

Solvent free extraction/concentration

Larger surface and phase volume

Improved recovery and sensitivity

Wide polarity range, lowest LODs

Headspace or immersion mode

Efficient batch-wise extraction

Welcome to the next level of SPME

SPME

TF-SPME

Insert

Extract

Desorb and Analyze

1

2

3

NEW

794 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

PEAKS of Interest

Shimadzu Celebrates

50th Anniversary

Shimadzu has celebrated 50 years of

business in Europe, with 300 guests

attending an event in Duisburg, Germany

to commemorate the anniversary. The

“Magic Moments Night” took place at

the Mercator Hall, and featured music,

show acts, dinner, speeches, greeting

notes, and a “Walk of History.” The

musical part of the evening was

covered by members of the Duisburg

Philharmonic Orchestra. The show

act performed by “Physikanten & Co”

combined entertainment and science.

Giant vortex rings flew 20–30 m across

the hall, and, in a rapid sequence of

experiments, the fascinating aspects of

carbon dioxide were explored. For the

“Walk of History.” Shimadzu collected

historic advertisements, brochures, and

photographs from exhibitions covering

50 years of corporate history in Europe.

Shimadzu’s Japanese-based Super-

visory and Executive Board flew in from

Japan to attend the party with the Euro-

pean Shimadzu team, as well as Shimad-

zu’s distributors and subsidiaries. The

evening’s program was hosted by Ger-

man television journalist Asli Sevindim.

Waters Opens Food and Water

Center in Singapore

Waters Corporation (Milford, Massachu-

setts) has opened a new International

Food and Water Research Centre (IFWRC)

in Singapore to address the growing chal-

lenges of food and water security and

safety. The center will be led by a scien-

tific advisory panel that will identify mean-

ingful, innovative projects by working with

academic and industrial leaders.

Important research areas such as food

fraud discovery, water contamination

research, food quality enhancement, and

new ingredient and formulation studies

will be prioritized as the research center

seeks to find solutions to food and water

supply challenges around the world.

Researchers will gain access to

IFWRC’s state-of-the-art facilities outfit-

ted with analytical instrumentation from

Waters. In addition, the laboratory will

be staffed with scientists and researchers

who will work closely with project owners

throughout implementation. ◾

CHROMATOGRAPHY

MARKET PROFILE

Chinese Laboratories Share Views in SurveyFor high performance liquid chroma-

tography (HPLC), ultrahigh-pressure LC

(UHPLC), and LC–mass spectrometry

(LC–MS) technologies, China has con-

tinued to provide solid growth opportu-

nities, as a result of the country’s invest-

ments in the pharmaceutical industry, as well as agriculture and food testing. Major

instrument manufacturers including Agilent, Shimadzu, Thermo Fisher Scientific,

and Waters have established a stronghold in the market, and continue to per-

form well. As the market continues to evolve, Chinese HPLC users are expressing

their preferences, gravitating to only a few manufacturers. Some Chinese users

maintain a strong allegiance to indigenous HPLC suppliers. However, they also

recognize that the quality, reliability, and performance from the international HPLC

manufacturers are unrivaled, swaying purchase decisions in favor of global brands.

Top-Down Analytics (TDA) recently surveyed over 200 Chinese HPLC, UHPLC,

and LC–MS users who provided opinions about their instrument and consumables

suppliers. Approximately 30% of the respondents were from pharmaceutical and

biotechnology laboratories. Agriculture, food and beverage, and government labo-

ratories each represented about a fifth of the responses, and LC users from chemical

laboratories accounted for about 14% of the survey participants.

When asked to compare domestic or Chinese LC manufacturers with global

brands, some respondents were quite candid, indicating that the Chinese brands

were improving, but still far from catching up to the quality of the international sup-

pliers. For columns and other consumables, many Chinese laboratories prefer to use

local distributors.

The use of UHPLC continues to increase in popularity, keeping pace with North

American and European trends. The overall market for HPLC, UHPLC, and LC–MS in

China represents a significant share of the overall market, accounting for about 11%

of the worldwide analytical instrument industry. In 2017, TDA estimates there were

about 7000 combined HPLC and LC–MS installations in China. Growth is expected

to remain quite robust for the next few years, as a result of continued expansion of

life science research laboratories and applied markets.

Market size and growth estimates were adopted from TDA’s Industry Data, a

database of technology market profiles and benchmarks, and the 2018 Instrument

Industry Outlook report from independent market research firm Top-Down Analytics

(TDA). Survey data was extracted from TDA’s report, “A Liquid Chromatography

Survey in China: Chinese Scientists Share Their Preferences.” For more information,

contact Glenn Cudiamat, general manager, at (888) 953-5655 or glenn.cudiamat@

tdaresearch.com. Glenn is a market research expert who has been covering the

analytical instrumentation industry for more than two decades.

Sector distribution of Chinese HPLC and LC–MS survey respondents (n = 223).

wyatt.com

© 2018 Wyatt Technology. All rights reserved. All trademarks and registered trademarks are properties of their respective holders.

Triple detection is a whole new ballgame. Our detectors

for multi-angle light scattering, refractive index and

differential viscosity represent the leading technology

in their categories. When our three instruments are

combined, they create an all-new level of capability

that dramatically expands your characterization

capabilities. It’s an example of the whole exceeding

the sum of its parts. Now, you can do more, see more,

and know more. Simultaneously.

Learn how triple detection can help your research team

hit it out of the park at wyatt.com/Triple

Triple detection combines the power

of three—or more—instruments

to determine absolute molecular

weights, sizes and viscosities.

7

4

1

0 .2 3

65

98

TAB ESC DEL

ENTER

DAWNHELEOS-II

7

4

1

0 .2 3

65

98

TAB ESC DEL

ENTER

OptilabT-rEX

0 .

1 2 3

4 5 6

7 8 9

TAB ESC DEL

OK

796 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

LC TROUBLESHOOTING

Dwight R. Stoll

In the previous installment of “LC Trou-

bleshooting” (1), I reviewed the basic

working principles behind the two most

commonly used LC pump designs in use

today: so-called low- and high-pressure

mixing systems. I then discussed why

using a mobile-phase mixer between the

fluid convergence point at the pump and

the sample injector is needed in both

cases, albeit for different reasons. Finally,

I showed the effect of using mixers with

different volumes for different separation

conditions, and discussed some of the

advantages and disadvantages associ-

ated with changing mixer volumes.

This month, I am continuing with the

theme of mixing and mixers, but this time

focusing on what happens when we inject

a sample into the mobile-phase stream,

particularly in cases where there is a mis-

match between the compositions of the

two fluids. This mismatch most commonly

exists as a difference in solvent composi-

tion (for example, injecting an analyte dis-

solved in 100% acetonitrile into a mobile

phase of 20/80 acetonitrile/water) between

the sample and mobile phase. However,

there certainly are situations where differ-

ences in the pH or buffer composition of

the two fluids can also be very important.

It is most certainly true that there are

many analytical situations where the effect

of the injected solvent is practically negli-

gible; for example, injecting 1 μL of sam-

ple into a 150 mm x 4.6 mm i.d. column

that has a volume of about 1.5 mL. However,

I also think many practitioners of LC are sur-

prised to learn just how serious the effect

of the injected sample solvent can be, most

commonly when they observe bad results. I

am hopeful that this installment will shed a

little more light on these issues, and particu-

larly address the question of whether or not

mixing is needed after the injector.

Combining Fluids in LC

Systems—Where and How?

Three of the different ways that fluids are

brought together in LC systems are illus-

trated schematically in Figure 1. The first

two represent the ways fluids are brought

together in either high-pressure or low-

pressure mixing pump systems. Although

I discussed the differences between

these designs in detail in the previous

installment of “LC Troubleshooting,” the

differences are actually quite relevant

to the topic of sample injection, and so

worth repeating here. In short, the major

fundamental difference between the two

approaches (Figure 1A and 1B) is that, in

the first case, the two fluid streams con-

verge in a kind of parallel fashion, so that

the two fluids are always in close contact,

whereas in the second case, small packets

of each fluid are introduced into a single

fluid path in a kind of serial fashion.

The third scenario of Figure 1 illustrates

the way that a sample is introduced into

the mobile-phase stream for nearly all

injectors in use in LC today. This is con-

ceptually similar to the way that fluids are

combined in the case of a low-pressure

mixing pump; that is, volumes of the flu-

ids are introduced to a single flow path in

a serial fashion, or “end to end.” In the

case of a typical sample injection, the

consequence of this is quite striking. If

we assume that a 10 μL portion of sam-

ple is injected into a 120 μm (0.005”) i.d.

tube leading to the LC column; by dividing

the sample volume by the cross-sectional

area of the tube, we find that the sample

could occupy as much as a 90 cm length

of this tubing, bracketed on both ends

by mobile phase. Given the degree of

physical separation of the middle of the

sample plug from the closest mobile

phase fluid (in this case 45 cm), and the

relatively short time it takes for the sample

to reach the column under typical condi-

tions (a few seconds), there is absolutely

no way that the two fluids will actually

mix before the sample reaches the col-

umn. And so, this then leads to the ques-

tion, “Under what circumstances should

a physical mixer be deployed to ensure

that the sample mixes with the mobile

phase before reaching the column?”

Does it Really Matter If the Sample is

Mixed with the Mobile Phase?

As with many things, the answer here is

“It depends.” In my thinking about this,

I divide different situations into two cat-

Is a mixer needed between the injector and column in HPLC?

Mixing and Mixers in Liquid Chromatography– Why, When, and How Much? Part II, Injections

Ico

n i

ma

ge

: J

oe

Zu

gc

ic,

Zu

gc

ic P

ho

tog

rap

he

rs,

Inc

.

© 2018 Hamilton Company. All rights reserved.

Your sample depends on it

More than 50 years of precision liquid

handling experience. Proven accuracy.

Continuous research and development.

Choose an autosampler syringe from

Hamilton Company and be confident

in your results.

Hamilton offers a line of syringes

designed to work with a wide range

of the most popular autosamplers

from Agilent, CTC PAL, Spark Holland,

and more. Each syringe is expertly

handcrafted to maximize sample

integrity, process efficiency, and

new long-life syringe technology.

For more information, visit

www.hamiltoncompany.com/autosampler

or call toll free 1-888-525-2123.

798 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

egories: conditions that favor focusing of

the analyte at the column inlet, and those

that do not. This is most effectively under-

stood by way of example. Figure 2 shows

the chromatograms obtained when a sim-

ple mixture of alkylphenones is injected

into a reversed-phase column followed

by solvent gradient elution, but with two

different injection conditions. Clearly, the

first case (A) yields a very nice separation,

whereas in the second case (B), the five

compounds are not well separated and

the peak shapes are terrible. The difference

here is that, in Case A, the analytes are dis-

solved in a 30/70 acetonitrile/water mixture,

and, in Case B, a 70/30 acetonitrile/water

mixture. The solvent gradient starts at 50%

acetonitrile, and runs to 90% acetonitrile

at the end of the gradient. We explain this

result by recognizing that, in Case A, the

analytes are dissolved in solvent that con-

tains less acetonitrile than the mobile phase

itself (50%) at the beginning of the analysis.

Under these conditions, the analytes will be

well retained by the stationary phase, have

a low velocity, and are effectively “stuck”at

the column inlet. We say that they are

“focused” or “compressed”into a narrow

band, and that this narrow width estab-

lishes a kind of initial condition from which

the rest of the separation (and subsequent

peak broadening) develops. This effect has

been known for decades (2), but also con-

tinues to be a subject of active research (3,4).

On the other hand, in case B, the sample

contains more acetonitrile than the mobile

phase at the starting point of the analysis.

Under these conditions, the analytes will

be poorly retained, have a high velocity

approaching the mobile phase velocity, and

are spread out across a large fraction of the

column bed. This, too, establishes a kind of

initial condition for the analyte bands, but,

in contrast to case A, one that involves very

broad peaks, from which the separation

cannot recover because the peaks will only

get broader as the separation develops.

So, to answer the question that heads

this section, I would say that, generally

speaking, if the injection conditions favor

focusing of the analytes at the column inlet,

as is the case in Figure 2A, then no actual

mixing of the sample and mobile phase is

needed to obtain good results. Indeed,

the chromatogram in Figure 2A is convinc-

ing evidence for this. Of course, there are

always exceptions (see the section later

about viscous fingering); however, this

view should be pretty broadly applicable.

On the other hand, if the injection condi-

tions are not favorable for focusing, as in

Figure 2B, then this can be a real problem,

FIGURE 1: Idealized representation of the different ways two fl uids converge under different circumstances in LC systems: (A) convergence in a binary high-pressure mix-ing pump, (B) convergence at the outlet of a solvent proportioning valve in a low-pres-sure mixing pump, and (C) convergence when a sample is injected into a mobile phase stream that will carry the sample to the LC column.

FIGURE 2: Comparison of chromatograms obtained from injection of samples in (A 30/70

acetonitrile/water, or (B) 70/30 acetonitrile/water. Conditions: column, 50 mm x 2.1 mm i.d.

C18; injection volume, 40 μL; gradient elution from 50-90% acetonitrile from 0–15 s, with water as the aqueous phase; fl ow rate, 2.5 mL/min.; analytes are alkylphenone homologs from ace-tophenone to hexanophenone.

A(a)

(b)

(c)

B

Tube i.d. ~170 μm

Tube i.d. ~120 μm

~90 cmFor 10 μL injected

1500

1000

500

Ab

sorb

an

ce (

mA

U,2

54

nm

)

0

1500

1000

500

0

0 0.1 0.2 0.3 0.4 0.5

0 0.1 0.2

Time (min)

0.3 0.4 0.5

(a)

(b)

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 799

and solving the problem may or may not

require a physical mixer, depending on

how the solution is implemented.

Dealing with Situations Involving

Unfavorable Solvent Mismatch

In my laboratory, we have studied the

effect of the composition and volume of

the injected sample on separation per-

formance (for example, as in Figure 2)

extensively. When I discuss this work with

people, the two things I hear most com-

monly are that: 1) extensive mixing of the

sample with the mobile phase is needed

to achieve good results as in Figure 2A;

and 2), it is physical differences between

the sample and mobile phase (for exam-

ple, viscosity) that are the root cause of

poor results (as in Figure 2B). Here, I’d like

to discuss a few results that I think shed

some light on these issues.

Viscous fingering is a physical phe-

nomenon that can develop when a less

viscous fluid (for example, acetonitrile)

is injected into a more viscous one (for

example, water) that flows into a porous

medium. In this situation, local flow insta-

bilities can develop that produce “fin-

gers” of the injected fluid that appear

to reach into the adjacent fluid (in the

chromatographic context, the mobile

phase). This effect has been known in

preparative chromatography for some

time, but more recently was also visu-

ally demonstrated under analytical scale

chromatography conditions by Samuels-

son, Fornstedt, and coworkers (5). This,

and related work, provides compelling

evidence that viscous fingering can

occur in analytical chromatography col-

umns and probably leads to effects on

chromatographic efficiency (that is, plate

number, or plate height) that cannot be

accounted for using simple plate models

of chromatography.

In our own work, we have adapted a

simple plate model of liquid chroma-

tography that enables us to simulate the

effects of sample solvent composition and

volume on chromatographic peak shape

and efficiency (4,6). To summarize a great

deal of work in this direction, I would say

that, by using this simple plate model, we

can account for a large majority of the

effects of sample solvent composition

and volume on peaks that we observe

in real experiments without invoking the

effects of more complex processes, such

as viscous fingering (for example, we have

been able to faithfully predict results like

those shown in Figure 2 [4]). Neverthe-

less, there are some differences between

simulation and experimental results that

we cannot account for, and it possible

that the viscous fingering could explain

some of these differences. Clearly, more

work on this is needed to more fully

understand these effects.

Our work on this topic has yielded

some experimental results that are

instructive here. First, the peaks shown in

Figure 3 were obtained from experiments

800 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

aimed at understanding the shape of the

injected sample plug injected into the

second dimension column in two-dimen-

sional liquid chromatography. In this case,

the mobile phase was 50/50 acetonitrile/

water, and the sample was the same solu-

tion but spiked with 10 μg/mL of uracil,

which is used to trace the concentration

profile by UV absorbance detection.

In other words, the uracil is a proxy for

other sample solvent components, such

as acetonitrile. These experiments are

done without a column installed, such

that the profile we observe is essentially

the sample profile as it would enter the

LC column. The point I want to empha-

size here is that, even with a relatively

small injection volume of 13 μL, there is a

point in the center of the profile where the

fluid that is detected is essentially pure

sample. In other words, there is very

little mixing between the center of

the sample plug and the surrounding

mobile phase. I think this is relatively

easy to understand when we imagine

what happens inside the system using

the illustration in the third case of Fig-

ure 1. The practical consequence of

this, then, is that if we have analytes

dissolved in a sample with a high con-

centration of acetonitrile and we inject

this into a mobile phase with a much

lower concentration of acetonitrile,

there will be a point where the sample

solvent acts as the mobile phase inside

the column because there is insufficient

mixing with the surrounding mobile

phase. This brings us back to the ques-

tion, “Should we install a mixer between

the injector and the column?” The main

problem with installing a simple mixer

(think spinning stir-bar) in this context is

that effective mixing of the sample will

require a relatively large volume mixer,

which will both effectively increase the

volume of the injected sample, and add

gradient delay volume to the system. If

neither of these issues is detrimental for

the analysis at hand, then adding such

a mixer could be a good solution to the

problem. In our own work on the sample

solvent problem in the context of 2D-LC

where analysis time is a precious resource,

we have developed an approach referred

to as active solvent modulation (ASM) that

works quite well (7).

The last bit of data I’d like to discuss

here actually comes from our work on

ASM, where an injected sample is mixed

with the mobile phase stream in more of

a parallel fashion (that is, like converging

streams in Figure 1A) than the more typ-

ical serial fashion (as in Figure 1C). Fig-

ure 4 shows a comparison of the sample

plug profiles observed when the injected

sample is brought together with mobile

phase in a serial fashion (black traces), or

in a parallel fashion (red traces). In these

cases, the mobile phase was 50/50 aceto-

nitrile/water, and the injected sample was

acetonitrile (4A), or 2-propanol (4B), each

containing 0.1% acetone as a tracer that

is observed by UV absorbance detec-

tion. There are two main points I’d

like to make about these results. First,

here, as in Figure 3, we see that in

the case of serial sample introduction

there is little mixing of the sample with

the surrounding mobile phases. On

the other hand, when the sample and

mobile phase are brought together in

a parallel fashion, the mixing is very

effective, as indicated by the lowered

concentration of acetone detected

during introduction of the sample

plug. Note that the profile is wider in

time because the effective volume of

the sample plug is increased as it is

mixed with mobile phase. The differ-

ences in the extent of dilution of the

acetone (as indicated by the different

peak heights) are related to the differ-

ent viscosities of the two samples. Sec-

ond, there are not obvious differences

in the sample plug profiles observed

for the two sample solvents injected,

even though their viscosities vary by a

factor of about seven.

FIGURE 3: Comparison of sample plug profi les obtained from injections of either 13 or 40 μL of

sample from a conventional fi xed loop injector. The mobile phase was 50/50 acetonitrile water, pumped at 2.5 mL/min., and the sample was the same solvent spiked with uracil at 10 μg/mL. The injector was connected directly to the detector with a short length of 75 μm i.d. tubing. Adapted with permission from ref. (4).

700

600

500

400

300

200

100

0

700

600

500

400

300

200

100

Ab

sorb

an

ce (

mA

U, 254 n

m)

00 1 2 3

0 1 2 3

Time (s)

(b) 40 μL

(a) 13 μL

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 801

Summary

When possible, it is desirable to match the sample solvent to the

mobile-phase composition used in a LC method, and use an injec-

tion volume that is small relative to the volume of the LC column

itself to minimize the effects of the injected sample on separation

performance. However, there are some situations where this is not

possible because of limitations on analyte solubility, or the need to

inject large volumes to improve detection sensitivity. In these situa-

tions, it is helpful to have a detailed understanding of what happens

during the injection process. In situations where the relationship

between the properties of the sample solvent and the mobile phase

favor analyte focusing, most likely a mixer is not needed between

the injection point and the LC column. However, if the situation

does not favor analyte focusing, this can lead to very bad results

(see for example, Figure 2B), and in these cases installing a mixer, or

using an alternate means of sample injection may be helpful.

Acknowledgements

I want to thank Gustavus Adolphus College student Hayley

Lhotka for collecting the data shown in Figure 4.

References

(1) D.R. Stoll, LCGC North Amer. 36(10),746–751 (2018).

(2) L.R. Snyder and D.L. Saunders, J.Chromatogr. Sci. 7, 195–208 (1969). doi:10.1093/chromsci/7.4.195.

(3) S.R. Groskreutz and S.G. Weber, J. Chromatogr. A 1409, 116–124 (2015). doi:10.1016/j.chroma.2015.07.038.

(4) D.R. Stoll, R.W. Sajulga, B.N. Voigt, E.J. Larson, L.N. Jeong, and S.C. Rutan, J. Chromatogr. A. 1523, 162–172 (2017). doi:10.1016/j.chroma.2017.07.041.

(5) J. Samuelsson, R.A. Shalliker, and T. Fornstedt, Microchem. J. 130, 102–107 (2017). doi:10.1016/j.microc.2016.08.007.

(6) L.N. Jeong, R. Sajulga, S.G. Forte, D.R. Stoll and S.C. Rutan, J. Chromatogr. A 1457, 41–49 (2016). doi:10.1016/j.chroma.2016.06.016.

(7) D.R. Stoll, K. Shoykhet, P. Petersson, and S. Buckenmaier, Anal. Chem. 89, 9260–9267 (2017). doi:10.1021/acs.analchem.7b02046.

Dwight R. Stollis the editor of “LC Troubleshooting.” Stoll is a profes-sor and co-chair of chemistry at Gustavus Adolphus College in St. Peter, Minnesota. His primary research focus is the development of 2D-LC for both targeted

and untargeted analyses. He has authored or coauthored more than 50 peer-reviewed publications and three book chapters in separation science and more than 100 conference presentations. He is also a member of LCGC’s editorial advisory board. Direct correspondence to: [email protected]

FIGURE 4: Comparison of sample plug profi les for injections of ei-

ther (A) acetonitrile or (B) 2-propanol into a 50/50 acetonitrile/water mobile phase. Both samples contained 0.1% acetone (v/v). The ASM injection loop volume was 40 μL, and the injection valve was connect-ed directly to the detector using a short length of 120 μm i.d. tubing.

ABOUT THE COLUMN EDITOR

90

80

70

60

50

40

30

20

10

120

100

80

60

40

20

00 5

00 5 10

Sample introduced serially

(a) Sample in acetonitrile

(b) Sample in PrOH

Time (s)

Ab

sorb

an

ce (

mA

U,

22

0 n

m)

Sample introduced in parallel

15 20

10 15 20

( Reliably Sensitive )

Glyphosate Analysis in Food

The Experts for 30 Years

www.pickeringlabs.com

CATALYST FOR SUCCESS

PINNACLE PCX

802 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

Ico

n i

ma

ge

: J

oe

Zu

gc

ic,

Zu

gc

ic P

ho

tog

rap

he

rs,

Inc

.

SAMPLE PREP

PERSPECTIVES

Since I took responsibility for this col-

umn, I’ve tried to focus on techniques,

with a balance of theory, applications, and

operational aspects. I’ve aimed to provide

a balance that will be of interest to both

the bench chemist and the laboratory

supervisor, at all levels of education and

background. Occasionally, trends related

to sample preparation are reported. This

month, the final installment of the year will

take a slightly different approach. Three

vignettes are presented, either looking

back at important, but overlooked, devel-

opments, or prognosticating the future.

The Total Youden Blank

In our previous column (1), we discussed

the role of sample blanks in chemical anal-

ysis, and provided an overview of various

types of blanks. One concerned reader

sent an email to comment on an often

overlooked, but highly important, type of

blank, the total Youden blank. From the

mid-1940s to the early 1970s, W. J. Youden

presented research on the application

of multivariate analysis to data sets from

chemical determinations (2–6). The cali-

brations and methods presented deter-

mine, among other uses, constant errors

with proposed calculations to confront

proportional errors. These methods use a

fractional factorial design to minimize the

number of analyses in assaying several

factors. In single laboratory validations,

the susceptibility of analytical methods to

small changes in parameters is examined.

For example, in sample preparation, we

have several optimization parameters of

varying importance, as illustrated in Table

I (7). This list of optimization parameters

is not exhaustive, but limited to the major

influences. Other extraction methods, and

each step in an analytical procedure, will

contribute different, and additional, opti-

mization parameters. Evaluating each of

these in a one-at-a-time approach would

be prohibitive. When analytical signals are

due solely to the presence of analyte, with

no matrix-based interference, the signal

from the analyte [SA] can be modeled as

[SA] = K + [αA]CA, where K is the Youden

blank or “true sample blank,” [αA] is the

slope of the calibration curve (or analytical

sensitivity), and CA is the analyte amount

(either concentration or weight). Several

reviews or tutorials, such as those in refer-

ences 7–10, provide more detailed treat-

ments of the Youden method.

In Memory of Patrick D. McDonald

and Recent Advances in Solid-

Phase Extraction Symposium

One of the pioneers in the development

of solid-phase extraction (SPE), Patrick

D. McDonald, passed in August 2017. Dr.

McDonald (Figure 1) was hired by Waters

and Associates in 1974, initially to develop

preparative liquid chromatography (LC)

instruments. Later, he was charged to “find

new, faster, more convenient ways to per-

form traditional sample preparation oper-

ations (11).” While others had used adsor-

bents in sample clean-up procedures, his

team went from proposing the use of LC

technology in a June 1977 internal memo to

Douglas E. Raynie

As this is the final “Sample Prep Perspectives” column of the year, it is fitting to assess the state of the field by taking

a look back and a look forward. Specifically, we’ll start by addressing a reader’s email, then look at the state of sample

preparation at two recent conferences. In the August issue, we looked at the role of blanks (samples lacking the analyte of

interest used to determine or track the source of contamination or sample degredation and taken through the analytical

process) in understanding the sample analysis process. One reader suggested that it is appropriate to address the total

Youden blank, an often overlooked and perhaps the most true blank. The total Youden blank completely eliminates the

constant error component arising from any source of bias involved in the measurement. In August 2017, we lost Dr. Patrick

McDonald. McDonald was a Research Fellow at Waters Corporation and one of the pioneers in the development of solid-

phase extraction. At the Fall National Meeting of the American Chemical Society, a symposium was held in his memory.

We’ll review the symposium and McDonald’s contributions to get an up-to-date snapshot of the field of SPE. Finally, at this

summer’s ExTech (International Symposium on Extraction Technologies), an expert panel offered their views on the future

of sample preparation. A summary of this panel discussion is presented.

A Look Back and A Look Forward—An Annual Check-up on the State of Sample Preparation

Minimize complexity. Magnify your focus.

Milli-Q® IQ 7003/7005Integrated Ultrapure and Pure Water System

• Simple and intuitive Q-POD® and

E-POD® dispensers

• Touchscreen display for unparalleled

ease of use

• (ɞRUWOHVV�GDWD�PDQDJHPHQW�DQG�

ensured traceability

• Compact and versatile to

optimize lab space

7R�ɟQG�RXW�PRUH��YLVLW��

EMDMillipore.com/labwater

The life science business of Merck

.*D$��'DUPVWDGW���*HUPDQ\�RSHUDWHV�DV�

MilliporeSigma in the U.S. and Canada.

0LOOLSRUH6LJPD��WKH�YLEUDQW�0��0LOOL�4��(�32'�DQG�4�32'�

DUH�WUDGHPDUNV�RI�0HUFN�.*D$��'DUPVWDGW��*HUPDQ\�RU�

LWV�DɡOLDWHV��$OO�RWKHU�WUDGHPDUNV�DUH�WKH�SURSHUW\�RI�WKHLU�

respective owners. Detailed information on trademarks

is available via publicly accessible resources.

j������0HUFN�.*D$��'DUPVWDGW��*HUPDQ\�DQG�RU

LWV�DɡOLDWHV��$OO�5LJKWV�5HVHUYHG�

804 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

shipping the first “Sample Enrichment and

Purification” (SEP–PAK) product in January

1978. SEP–PAK featured a heat-shrinkable

polyethylene body, triaxial compression

technology, and preparative LC C18-silica

particles. Figure 2 shows the cover of the

original February 1978 marketing brochure

for SEP–PAK cartridges. Note that these

questions concerning the cost, time, and

interferences of sample preparation still res-

onate today, though it is hoped that sam-

ple preparation advances have significantly

improved over the decades and the current

situation reflects the simultaneous advances

in analysis techniques! As SEP–PAK car-

tridges became accepted and other manu-

facturers developed other approaches, J.T.

Baker’s term, solid-phase extraction (SPE),

came to represent the technique.

McDonald also edited Waters’s “Sol-

id-Phase Extraction Applications Guide

and Bibliography.” By the sixth edition in

1995, over 3000 applications were com-

piled. In 1996, he and his team devel-

oped and patented the Waters Oasis

HLB copolymer (12). This sorbent is a

hydrophilic–lipophilic balance copoly-

mer. The water-wettable sorbent retains

analytes over a wide polarity range and

is stable from pH 1 to 14.

In memory of McDonald, a symposium

on recent advances in SPE was held at the

256th National Meeting of the American

Chemistry Society at Boston in August,

organized by Tom Walter, a corporate

fellow at Waters and former associate of

McDonald. The nine invited oral presenta-

tions, listed in Table II, can be considered

an assessment of the current state of the

field. In particular, small-scale biological

samples, matrix removal, and magnetic

sorbent particles are all driving present

research in SPE. This column and peer-re-

viewed analytical chemistry journals review

the current state of SPE periodically.

Views on Future Developments

in Sample Preparation

Last spring, our sister publication fea-

tured an interview conducted by editors

of LCGC with panelists from compa-

nies that exhibited at Analytica 2018

(13). The topic was to assess the trends

and developments in the chromatogra-

phy sector. Specifically, representatives

from Biotage (Paul Roberts), CEM (Alicia

Stell), Eprep (Peter Dawes), Gerstel (Oli-

ver Lerch), Phenomenex (Matt Brusius),

and UCT (Danielle Mackowsky) were

asked their opinions on emerging sam-

ple preparation trends, important recent

developments, obstacles to continued

sample preparation developments, and

the biggest accomplishments in the past

year. While each of the six panelists pro-

vided perspectives from their viewpoints,

TABLE I: Potential factors to be exam-ined in the robustness testing of com-mon sample preparation techniques (from reference 7)

Sample Preparation Technique

Factors

Solid-PhaseExtraction

Sorbent type

Sorbent manufacturer

Sorbent mass

Sample massor volume

Wash solvent

Elution solvent

Evaporationtemperature

Sample pH

Buffer pH

Matrix Solid-PhaseDispersion

Sorbent type

Sorbent manufacturer

Sorbent mass

Sample pH

Buffer pH

Sonication time

Evaporationtemperature

Wash solvent

Elution solvent

Sample massor volume

TABLE II:TT Papers presented at “Recent Advances in Solid Phase Extraction: Symposium inhonor of Patrick D. McDonald,” held at the Fall 2018 National ACS Meeting

Harnessing the Power of Solid-Phase Extraction for Peptide Bioanalysis. M. Lame (Waters)

Solid-Phase Extraction (SPE) in Bioanalytical Method Development for Therapeutic Peptides. K. Lee (Waters)

Development, Validation and Application of a Cation-exchange, Solid-Phase Extraction forthe Determination of Nanoparticle-released Drug Concentrations in Plasma. C. Holliman, W. Song, J. Tweed, Z. Gu (Pfi zer)

Recent Advances in Solid-Phase Extraction for Biological Sam-ples–Fulfi lling the Promise of SPE. J. Danaceau (Waters)

New Developments in SPME. J. B. Pawliszyn (University of Waterloo)

Effective Simplifi ed SPE for Modern Multiresidue Analysis: Recent Developments for Pass-through, Dispersive, and Retention/Elution SPE. M.S. Young, K. Tran (Waters)

Lipid Selective SPE Materials Simplify Sample Preparation and Improve Results. D. Lucas, B.E. Richter, L. Zhao (Agilent Technologies)

Variability of Solute-Sorbent Binding Constants in SPE Materials. D.E. Raynie, S. Pandey, S. Subedi, D. Lucas, B.E. Richter (South Dakota State University)

Porphyrin-based Magnetic Nanocomposites for Effi cient Extraction of Polycyclic Aromatic Hydrocarbons from Water Samples. J. Yu, S. Zhu (China University of Geosciences)

FIGURE 1: Dr. Patrick D. McDonald (1944-2017), Research Fellow at Waters and Associ-ates and inventor of SEP-PAK, Oasis, and oth-er leading advances in SPE. (Photo courtesy of Tom Walter, Waters.).

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 805

common threads centered on develop-

ment of faster and higher throughput

techniques, automation, and accommo-

dation of smaller samples. Given that

this interview is freely available on-line

(http://www.chromatographyonline.com/

trends-and-developments), the reader is

invited to peruse this article.

Meanwhile, at about the same time, the

20th International Symposium on Advances

in Extraction Technology (ExTech) in Ames,

Iowa (June 19–22) took place. During

one feature, conference organizer Jared

Anderson from Iowa State University

assembled a panel that included technical

experts from sample preparation vendors

and academia. I was honored to join the

panel, which also consisted of Veronica

Pino Estevez (Universidad de La Laguna,

Spain), Elia Psillakis (Technical University

of Crete), Janusz Pawliszyn (University of

Waterloo), Bruce Richter (Agilent Technol-

ogies), Dan Cardin (Entech), and Jason

Herrington (Restek). An interesting mix of

views, with some significant convergence,

was noted. Based on the panel discussion,

new developments in the near term were

discussed. These include approaches for

matrix removal (which differ from sample

enrichment); field-based extractions and

other approaches to take the extraction

to the sample; and even one-size-fits-all

approaches to the extraction of com-

plex samples. Pino Estevez and Richter

discussed how separation scientists can

learn from materials science. For exam-

ple, Pino Estevez opined that the prepa-

ration of core-shell magnetic materials

will drive advances in magnetic-particle–

based separations. Psillakis, on the other

hand, offered the view that advanced,

or smart, materials are not necessary for

development of sample preparation. For

example, she has demonstrated success

in the manipulation of vacuum conditions

to provide more efficient extractions and

mentioned the salting-out effect in driving

chemical separations. Neither of these

manipulations (reduced pressure and salt-

ing out) can compete with the effect of

temperature in driving separations. This

led other members of the panel, nota-

bly Psillakis, Pawliszyn, and me, to stress

that what is missing, and limiting future

innovations, is understanding of the fun-

damentals of extraction. For example, at

its heart, all extractions involve manipula-

tions of phase contact, solubility, diffusion,

and phase separation in a thermodynam-

ically consistent manner. Knowledge of

the chemistry of analytical systems and

the impact of various manipulations (such

as vacuum or temperature) can influence

many technologies. Pawliszyn mentioned

the integration of the individual steps in an

analysis scheme, modeling, optimization

of mass transfer, and direct coupling to

mass spectrometry as needs for the future

development of sample preparation.

Pawliszyn noted the lack of indus-

trial chemists at the ExTech conference,

though it is noted that the infrequent

appearance of ExTech symposia in

North America (the previous ExTech in

the United States was in the Black Hills

of South Dakota nearly ten years ago,

in 2009) and the plethora of specialized

meetings can contribute to lower atten-

dance. He observed a conflict between

industrial adoption of new separation

technology with early inventions. Sim-

ilarly, claims that regulatory methods

generally are developed by academics

and technology manufacturers, due to

budgetary and workload issues in lab-

oratories associated with government

agencies, may also minimize industrial

adoption of new sample preparation

technologies. Hence, the drivers for

sample preparation development have

shifted from being the pull of industry

needs to the push of new technologies

in search of meaningful applications.

However, education toward a more com-

plete understanding of solubility and

phase equilibria will guide all analysts in

coming up with the sample preparation

approaches of the future.

References

(1) D.E. Raynie, LCGC North Am. 36(8), 494–497 (2018).

(2) W.J. Youden, Anal. Chem. 19, 946–950 (1947).

(3) W.J. Youden, Biometrics 3, 61 (1947).

(4) W.J. Youden, Mater. Res. Stand. 1, 268–271 (1961).

(5) W.J. Youden, Statistical Techniques for Collaborative Tests (Association of Offi-cial Analytical Chemists, Washington, DC, 1967).

(6) W.J. Youden and E.H. Steiner, Statistical Manual of AOAC (Association of Official Analytical Chemists, Washington, DC, 1975).

(7) E. Karageorgou and V. Samanidou, J. Chromatogr. A 1353, 131–139 (2014).

(8) R.C.C. Castells and M.A. Castillo, Anal. Chim. Acta 423, 179–185 (2000).

(9) A.R. Mauri, M. Llobat, and D. Adria, Anal. Chim. Acta 426, 135–146 (2001).

(10) A.G. Gonzalez and M.A. Herrador, TrAc Trend Anal. Chem. 26, 227–238 (2007).

(11) P.D. McDonald, “James Waters and his Liquid Chromatography People: A Personal Perspective,” http://www.waters.com/webassets/cms/library/docs/wa62008.pdf.

(12) E.S. Bouvier, R.E. Meirowitz, and P.D. McDonald, U.S. Patent 5,976,367 (1996).

(13) LC/GC Editors, The Column, 14, 2-19 (2018).

FIGURE 2: Questions asked on the cover of a February 1978 brochure for Waters’s SEP-PAK cartridges (from reference 11).

Douglas E. Raynie“Sample Prep Perspec-tives” editor Douglas E. Raynie is a Department Head and Associate Pro-

fessor at South Dakota State Univer-sity. His research interests include green chemistry, alternative solvents, sample preparation, high-resolution chromatography, and bioprocessing in supercritical fluids. He earned his PhD in 1990 at Brigham Young University under the direction of Milton L. Lee. Raynie is a member of LCGC ’s edito-rial advisory board. Direct correspon-dence about this column via e-mail to [email protected]

ABOUT THE AUTHORS

806 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

GC

CONNECTIONS

Nicholas H. Snow

Why does a puddle evaporate?

The next time it rains, observe

the puddles of water on the sidewalk or

street after the storm is over. As we know,

the water evaporates at the air tempera-

ture, say 25 oC, yet the boiling point of

water is 100 oC, and the water still evapo-

rates. This is due to the vapor pressure of

water. Water will continue to evaporate

until the air above the puddle becomes

saturated (100% relative humidity). This

relationship can be expressed chemically

by the following equations:

H2O(l)�H2O(g) Kp=PH2O [1]

The vapor pressure of the water is repre-

sented as PH2O, and Kp is the pressure-based

equilibrium constant for the evaporation

process. Figure 1 shows a stylized puddle.

Figure 1A shows some water molecules,

represented by dots, evaporating on a calm

(no wind) day. Figure 1B shows the same

puddle, but with the wind blowing. In which

situation does the water evaporate faster?

On the calm day, there is no wind to carry

the evaporated water molecules away, so

the air above the puddle becomes more

saturated and evaporation slows down.

On the windy day, the wind carries water

molecules away, so the air above the pud-

dle does not approach saturation and the

water evaporates more quickly. In both

cases, the system (surface, puddle and

air above it) is driving toward equilibrium,

saturation of the air above the puddle. A

puddle evaporates faster on a windy day.

Why does rubbing alcohol evaporate

faster than water? Try a simple experi-

ment. Rub a small amount of water on your

arm. As we know, when the water evapo-

rates, energy transfers from your arm to the

water, evaporating the water and making

your arm feel cool. Try again with rubbing

alcohol. The alcohol evaporates faster, and

your arm feels cooler. This difference arises

from differences in the heat of vaporization

and vapor pressure of water and rubbing

alcohol. This is an example of selectivity.

The ability of the water (H2O) or alco-

hol (Alc) to evaporate is governed by

simple chemical equations:

H2O(l) � H2O(g) Kc = [H2O(g)]

Alc(l) � Alc(g) Kc = [Alc(g)] [2]

In this case, the rubbing alcohol mix-

ture is considered a single substance, as it

evaporates as an azeotrope. Rubbing alco-

hol evaporates faster than water, due to

its higher liquid-vapor partition coefficient,

thus its higher vapor pressure.

Taking this further into chromatogra-

phy, all separations are also governed by

a similar phase equilibrium. Equation 3

represents the partitioning of an analyte

(A) from the mobile phase, the phase in

which it enters the column after the injec-

tion, into the stationary phase:

A (mp) ⇔ A (sp) Kc =

[A(sp)][A(mp)] [3]

Kc is the partition coefficient for the pro-

cess of sorption from the mobile phase

into the stationary phase. A(mp) refers to

an analyte dissolved in or moving in the

mobile phase, and A(sp) refers to an ana-

lyte dissolved in or sorbed on a station-

ary phase. A higher Kc indicates stron-

ger analyte attraction to the stationary

phase, leading to longer retention times.

Figure 2 shows this relationship applied

to a column with flowing mobile phase.

Note how this figure looks very much like

the puddle with the wind blowing shown in

Figure 1B. A column in gas chromatography

“The column is the heart of the separation.” Perhaps more accurately, the column is where the chemistry that generates

a separation happens. For chemists and non-chemists alike, the chemistry that drives the utility of a column to solve a

separation problem can be complex and confusing. Selectivity describes the ability of a column to effect a separation. This

installment of “GC Connections” focuses on selectivity, its definition, and its importance for generating separations and

resolution. We will also see how selectivity is the concept that underlies the idea of column polarity. We begin by asking two

simple questions about common observations, then extend these observations into a capillary GC column, and conclude

with an introduction to methods for evaluating the quality, selectivity, and polarity of a stationary phase or column.

Stationary Phase Selectivity: The Chemistry Behind the Separation

Ico

n i

ma

ge

: J

oe

Zu

gc

ic,

Zu

gc

ic P

ho

tog

rap

he

rs,

Inc

.

ADDITIONAL

15%DISCOUNTWITH THIS COUPON

Coupon Code: LCGC 1815 Expires: 12-31-2018

Cornerstone 5x7 PC tip-in_08-2018.indd 1 8/2/18 1:34 PM

NEWEasy-to-Use Website for Small and Medium LaboratoriesEasy-to-Use Website

Low Prices, Every Day

Innovative Brands Not Offered by “Big Box” Dealers

Great Values

General Laboratory Supplies & Equipment, PLUS:

Chromatography Supplies

Life Science Equipment

HPLC Solvent Disposal Systems

CHECK IT OUT!www.CornerstoneScientifi c.com

Cornerstone 5x7 PC tip-in_08-2018.indd 2 8/2/18 1:34 PM

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 807

behaves in a manner very similar to a pud-

dle on a windy day, with the analyte being

analogous to the puddle, the stationary

phase being analogous to the surface and

the mobile phase being analogous to the

wind. Like the rubbing alcohol versus water

example, if the puddle were a mixture of

two liquids, they would evaporate at differ-

ent rates, based on their vapor pressures.

In the column, if there are two analytes

they will move at different rates, based on

the difference in their partition coefficients

and the resulting vapor pressures above the

surface. For two analytes to be separated

on a column, the difference in partition

coefficient gives rise to selectivity, which

gives rise to the separation.

How is selectivity determined and

where does it come from? Figure 3 shows

the calculation of selectivity from a chro-

matogram, with the relevant equations. It

is simply the ratio of the adjusted retention

times (t’R, the difference between the reten-

tion time, tR and the gas hold up time tM) of

two peaks in the chromatogram. It is also

the ratio of the retention factors (k) and the

partition coefficients (Kc). Detailed descrip-

tions of the basic theory behind these rela-

tionships can be found in most basic text-

books on GC (1–3). Ultimately, selectivity

comes from thermodynamics. From Gen-

eral Chemistry, the Gibbs Equation relates

the partition coefficient to the standard free

energy (4).

6Go= -RT lnKc [4]

If two analytes are present, as seen in Fig-

ure 3, this equation becomes:

∆(∆G°) = –RT In and α = = e K

2

K1

K2

K1

-∆(∆G°)

RT

[5]

A more detailed description of the ther-

modynamic relationships involved in gas

chromatography can be found elsewhere

(5). Selectivity comes from the difference in

free energy change for the partitioning of

the analyte(s) from the mobile phase into

the stationary phase, as seen in Equation 3.

We have now seen the fundamental and

thermodynamic basis of selectivity. Next, we

discuss the impact of selectivity on the sep-

aration and resolution.

How does selectivity impact the sepa-

ration and resolution? The first goal of any

chromatographic method development is

to optimize the resolution. Looking again

at the chromatogram in Figure 3, the res-

olution may be calculated as the difference

between the two retention times divided by

the average of the two peak widths:

Rs=

2(tR(2)–t

R(1))

W1+W

2

[6]

If Rs is greater than 1.5, the peaks are

baseline separated. To understand the role

of selectivity in resolution, the fundamental

principles behind resolution in chromatog-

raphy can be considered using the follow-

ing equation:

Rs=

k

1+k

α-1α

√N4

[7]

Resolution is obtained from three basic

principles:

• retention factor (k), which is a measure

of how long the analyte is in the col-

umn. Too small k (< 2), and there is

not enough contact with the stationary

phase for the most effective chromatog-

raphy. Too large k (> 10), and there is a

diminishing return with longer time.

• theoretical plates (N), the measure of

column efficiency. The more theoretical

plates, the better the resolution.

• selectivity (α), the separating power of

the stationary phase based on differ-

ences in the strength of intermolecu-

lar interactions between the stationary

phase and analyte molecules.

Figure 4 shows two chromatograms with

equal selectivity. In Figure 4A, an example

from HPLC, the column has 18,285 theoret-

ical plates and in Figure 4B, in an example

from capillary GC, the column has 120,000

theoretical plates. With k being equal, a

dramatic effect on resolution is seen. Note

that, while the peak maxima are equally

spaced in the two chromatograms, the

peaks are broader in Figure 4A, reducing

the resolution. In classical packed column

GC and in HPLC, with lower N, adjusting

the selectivity is critical to obtaining nearly

all separations. In today’s capillary GC,

selectivity, with high N, is less important

but still must be considered.

The lower the separation efficiency (low

N), the higher the selectivity needed to

achieve separation. In most applications,

FIGURE 1: A) Water evaporating from a puddle in still air. Evaporated water molecules are represented as dots. B) Water evaporating from a puddle with blowing wind. In still air, the water stays above the puddle. In wind, the water is carried away.

FIGURE 2: Analytes partitioning into the stationary phase in a capillary column. Below the normal boiling point most of the molecules (represented here as dots) will partition into the stationary phase. The few dots in the mobile phase are moved along the col-umn by the fl owing mobile phase.

(a) (b)

808 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

selectivity has a critical impact on sep-

aration and resolution. As columns and

instruments become more efficient (more

theoretical plates, N), the need for high

selectivity lessens. In Figure 4A, a selectiv-

ity of 1.04, which could be a nearly 1 min

difference in retention time in a 20 min sep-

aration, is not sufficient to fully resolve the

peaks. If the selectivity calculated by the

equations shown in Figure 3 is one (1.00),

then the two analytes are not separated

at all and cannot be separated no matter

how efficient the column and instrument.

Fortunately, this is not often the case. Hav-

ing seen the basics of the thermodynamic

background of selectivity, we now turn to

methods for evaluating the selectivity of

stationary phases, and to a better under-

standing of the term “polar,” which is often

used to describe stationary phases in GC.

Most of the development of these meth-

ods occurred in the early days of GC, when

packed columns, with much lower efficiency

than capillary columns, were predominant.

What does “polar” really mean in GC?

Stationary phases are often described using

terms such as “polar”, “non-polar”, “mod-

erately polar” and the like. These terms

are often used very broadly, and can easily

generate confusion and misunderstanding.

The first problem with descriptions such

as “polar” is that they require context. As

an example of context, think about other

simple terms: “hot” and “cold”. In the

environs of New York City, in the winter, a

day with a high temperature of 65 oF would

be considered unusually hot, while in sum-

mer, unusually cold. When describing the

weather, the meaning of “hot” and “cold”

depends greatly on the environment. The

meaning of “polarity” requires similar con-

text. In describing columns, any discussion

of “polarity” must be based on specific

intermolecular interactions between the

stationary phase and the analytes. The

strength of these interactions determines

the free energy required for the analytes

to partition into the stationary phase, and

therefore determines the selectivity of the

stationary phase and, along with the col-

umn dimensions, ultimately the separat-

ing ability of the column.

Over the years, numerous tests have

been developed to evaluate the intermo-

lecular interactions that occur in gas chro-

matographic columns. These interactions

include the same ones we learned about

in school: dipole-dipole, dipole-induced

dipole, van der Waal forces, acid-base,

electrostatic interactions, and hydrogen

bonding. These classical studies generally

involve injecting test mixtures containing

a mix of compounds, each selected to

probe a specific interaction. These tests,

and many other aspects of column evalu-

ation, are discussed in great detail in the

textbook by Barry and Grob (6).

The Grob test mix, developed in the late

1970s is the most commonly used mixture

for testing the interactions and quality of

FIGURE 3: Determination of selectivity from chromatogram with defi ning equations. Chromatogram adapted from ChromAcademy (www.chromacademy.com, accessed September 2018).

TABLE II: List of test probes and interactions for the McReynolds test mixture

Test Probe Interaction(s)

Benzene Pi-pi, aromatic and olefi nic hydrocarbons

Ethanol or n-butanol Hydrogen bonding for alcohols, nitriles

2-butanone or 2-pentanone Proton acceptor – ketones, ethers, aldehydes, esters

Nitromethane or nitropropane Dipole-dipole interactions

Pyridine Strong proton acceptor – acid character of column

TABLE I: Components of the Grob Test Mixture and their column performance andchemistry measures.

Component Interaction(s)/Performance Tested

Decane, undecane Column effi ciency

Fatty acid methyl esters C10

, C11

, C12

Column effi ciency

1-octanol Hydrogen bonding, presence of silanol groups

Nonanal Aldehyde adsorption; non-hydrogen bonding

2,6-dimethyl phenol Acid-base interactions

2,6-dimethyl aniline Acid-base interactions

2-ethyl hexanoic acid Irreversible adsorption

Dicyclohexyl amine Irreversible adsorption

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 809

capillary columns (7,8). Every time you purchase a column, you are

provided with a column test mixture chromatogram that either uses

the Grob test mix, or is based on the principles described in the

original papers. Figure 5 shows a typical chromatogram of a column

test mix run on a commercially available column, with the test mix

components identified. There are several quality tests resulting from

this analysis, including reactivity and sensitivity to several intermo-

lecular interactions, column efficiency, and overall separating power.

The most important selectivity calculation is the spacing of the fatty

acid methyl ester peaks. This spacing must be even as each meth-

ylene unit (-CH2-) is added, indicating proper selectivity. The resolu-

tion of adjacent peaks, which also results from selectivity is tested, as

is the retention time and peak shape of each component, which is

determined by the specific intermolecular interactions between that

component and the column wall or stationary phase.

Table I lists the Grob test mix components with the specific

intermolecular interactions they probe. Comparing Table I to the

chromatogram shown in Figure 5, basic performance tests such

as the spacing and shape of the hydrocarbon and fatty acid peaks

indicate a properly installed generally well-performing column.

The acceptable shapes of the alcohol, phenol, aldehyde, and

aniline indicate a column that should perform well for weaker

acids and bases. The very poor shapes of the hexanoic acid and

dicyclohexylamine indicate that this column is likely to adsorb

stronger acids and bases. The Grob test mix thus provides an

excellent snapshot for the quality and performance of a column.

Most column manufacturers use their own variation of the Grob

test mix to demonstrate column quality and performance.

In order to discuss most measures of intermolecular inter-

actions with proper context, retention time data must be cor-

rected to account for differences in column dimensions, includ-

ing length, inside diameter, and stationary phase film thickness.

The classical means for this is calculating the Kovats Retention

Index for each analyte using Equation 8 (9):

I =100 +100x

log (t’R)u – log (t’

R)x

log (t’R)x+1

– log (t’R)x

[8]

The subscript u refers to the analyte. The subscript x refers to

the number of carbon atoms in the normal alkane eluting immedi-

ately prior to the analyte. The subscript x+1 refers to the number

of carbon atoms in the normal alkane eluting immediately follow-

ing the analyte. For example, an analyte eluting between hexane

(C6) and heptane (C7) might have a Kovats Retention Index of 650.

An example calculation is shown in Figure 6. In theory, the Kovats

Retention Index is linearly related to the free energy change for

the sorption process into the stationary phase, as described in

Equation 5, and it accounts for differences in the column dimen-

sions. The Kovats Retention Index of an analyte is a constant for

a given stationary phase and temperature and is independent of

column length, internal diameter and film thickness.

In the 1960s, based on Kovats Retention Indexes, two systems

for assessing stationary phase polarity, or really the strength of

a stationary phase for separating various classes of compounds

were developed by Rorhschneider and McReynolds (10,11).

These are used in column evaluation and are most commonly

termed McReynolds Constants. The experiment is simple. A test

FIGURE 4: Comparison of chromatograms with α = 1.04, con-stant k and differing N. Adapted from ChromAcademy (www.chromacademy.com, accessed September 2018).

Rs = 1.3, α = 1.04, N = 18285 (Peak*)

Rs = 3.0, α = 1.04,

N = 120,000 (Peak*)

α = 1.04

α = 1.04

(b)

(a)

� � �������������� ���������

������������������ ������� ���

� � ���������������������������

� ��������!��� ������∀�����

� � � ������#�����������������

��������������

����������������

����������������

810 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

mixture is injected under isothermal con-

ditions, and the Kovats Retention Index of

each analyte is determined and compared

to the retention index of that test analyte

on a standard non-polar stationary phase.

The McReynolds constant for that analyte

on that column at the given temperature

is the difference between the measured

retention index and the standard reten-

tion index. A list of the five most com-

mon test probes with the interactions

they were proposed to probe is given in

Table II and some constants for common

stationary phases are given in Table III

(12,13). Table III also includes Mondello

polarity numbers, described below.

McReynolds constants can be used to

estimate the ability of a stationary phase

to separate analytes of various compound

classes. For example, a high McReynolds

constant for benzene indicates a stationary

phase with strong pi-pi interactions and

ability to separate aromatic analytes. If the

McReynolds constants are high for all of the

test probes, the stationary phase may be

accurately described as “polar”. Note that

a column may have a high constant for one

test probe, but a lower constant for another,

indicating different degrees of “polarity”

depending on the analyte. McReynolds

constants provide the necessary context

for describing whether a stationary phase

is “polar”. In general, as the McReynolds

constants increase, the stationary phase

may be described as more polar.

This discussion leads to the final ques-

tion: How polar is my column? This

topic has seen renewed interest with

the advent of ionic liquids as station-

ary phases for capillary GC in the mid-

2000s (14,15). An ionic liquid is a molten

organic salt that is in the liquid phase at

or near room temperature. As station-

ary phases, ionic liquids are considered

highly polar, plus they have low vapor

pressure and do not decompose at the

temperatures normally employed in GC.

They seem to extend the polarity range

of stationary phases available in capillary

GC. An interesting difference between

ionic liquid stationary phases and most

traditional capillary GC stationary phases

is that the ionic liquids are liquid salts,

whereas the traditional stationary phases

are liquid polymers. At a molecular level,

the actual partitioning process may be

different for ionic liquid columns than for

traditional polymeric columns. This pos-

sibility merits further research.

In 2011, while evaluating the new ionic

liquid stationary phases, Mondello devel-

FIGURE 5: Chromatogram of a column test mixture. 1) decane, 2) 1-octanol, 3) undec-ane, 4) 2,6-dimethyl phenol, 5) nonanal, 6) 2-ethyl hexanoic acid, 7) 2,6-dimethyl aniline, 8) methyl decanoate, 9) methyl undecanoate, 10) dicyclohexylamine, 11) methyl laurate. column: 5% phenyl polydimenthyl siloxane, 30 m x 0.25 mm x 0.25 μm. Temperature Program: 70 oC/1 min, 5 oC/min to 250 oC.

1 23

5

6

12 14

Retention Time (min)

18 20 22 2416108

7

8 9

10

11

4

TABLE III: McReynolds constants and polarity numbers of selected stationary phases. Data from references 12, 13 and 16.

Stationary Phase Name X’ Y’ Z’ U’ S’ TotalPolarity Number

Squalane 0 0 0 0 0 0 0

Polydimethyl siloxane (DB-1, SPB-1, ZB-1, Rtx-1, etc.) 16 55 44 65 42 222 5

5% Phenyl polydimethyl siloxane (DB-5, SPB-5, ZB-5, Rtx-5, etc.) 33 72 66 99 67 337 8

50% phenyl polydimethyl siloxane (DB-17, SPB-50, Rtx-50, ZB-50, etc.) 119 158 162 243 202 884 20

Polyethylene glycol (WAX) 322 536 368 572 510 2,308 52

1,12-di(tripropylphosphonium) dodecane bis(trifluoro-

methanesulfonyl)amide (SLB-IL-59)338 505 549 649 583 2,624 59

1,9-di(3-vinylimidazolium)nonane bis(trifl uoromethanesulfonyl)imide (SLB-IL-100) 602 853 884 1017 1081 4,437 100

X’ = benzene

Y’ = n-butanol

Z’ = 2-pentanone

U’ = nitropropane

S’ = pyridine

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 811

oped a straightforward overall column

polarity scale, based on McReynolds

constants (16). These values for some sta-

tionary phases are included in Table III. To

determine the polarity number, the five

McReynolds constants are totaled, and

then the ratio of this total to the total for a

highly polar ionic liquid column (SLB-IL100)

is expressed with the SLB-IL100 column

having the value 100. Squalane is the

standard non-polar stationary phase for

McReynolds constant determination (all

McReynolds constants and polarity num-

ber are zero), but in practice, it is rarely

used with capillary columns. The classical

non-polar phases, polydimethyl siloxane

and 5% phenyl polydimethyl siloxane

have very low polarity numbers, so they

are accurately described as non-polar.

50% phenyl polydimethyl siloxane sta-

tionary phase, long considered moder-

ately polar shows a polarity number of

20, while polyethylene glycol phase, tradi-

tionally considered among the most polar

capillary column stationary phases, only

reaches a polarity number of 52.

Broad descriptions of polarity and

polarity numbers are useful but with cau-

tion. In Table III, the n-butanol McReyn-

olds constant is higher for polyethylene

glycol then for SLB-IL-59, yet the over-

all polarity number is lower. SLB-IL-59 is

an overall more polar stationary phase,

yet polyethylene glycol is likely to more

strongly retain polar alcohols. Results such

as this and the advent of ionic liquid col-

umns are leading to new thinking about

column polarity and how it is described.

Conclusions

The ability of a stationary phase to per-

form a separation is based on selectivity,

the difference in the strength of intermo-

lecular interaction between each analyte

and the stationary phase. Selectivity derives

from the partitioning process for the ana-

lytes between the mobile and stationary

phases and it plays a key role in the abil-

ity to achieve desired resolution. Column

quality and polarity are also determined by

the intermolecular interactions that occur

between compound classes of interest and

the stationary phase. Methods including the

Grob test mix, McReynolds constants and

polarity numbers provide the tools to ana-

lyze these interactions and to better under-

stand the chemistry behind the separation.

References

(1) H.M. McNair and J.M. Miller, Basic Gas Chromatography, (John Wiley and Sons, New York, 2nd ed., 2009).

(2) C.M. Poole, Ed., Gas Chromatography (Elsevier, Amsterdam, 2012).

(3) R.L. Grob, Ed., Modern Practice of Gas Chromatography (John Wiley and Sons, New York, 4th ed., 2004).

(4) N. Tro, Chemistry: A Molecular Approach, (Pearson, New York, 4th Ed., 2016) Chapter 18.

(5) N.H. Snow, J. Chem. Educ. 73(7), 592–597 (1996).

(6) E.F. Barry and R.L. Grob, Columns for Gas Chromatography Performance and Selection (John Wiley and Sons, New York, 2007).

(7) K. Grob, Jr., G. Grob, and K.J. Grob, Chro-matogr. 156, 1–20 (1978).

(8) K. Grob, Jr., G. Grob, and K.J. Grob, Chro-matogr. 219, 13–20 (1981).

(9) E. Kovats, Helv. Chim. Acta 41, 1915–1932 (1958).

(10) L. Rohrschneider, J. Chromatogr. 22, 6–22 (1966).

(11) W.O. McReynolds, J. Chromatogr. Sci. 8, 685–691 (1970).

(12) “Technical Bulletin 880: The Retention Index System in Gas Chromatography: McReynolds Constants”, Sigma-Aldrich, https://www.sigmaaldrich.com/Graphics/Supelco/objects/7800/7741.pdf (Accessed September, 2018).

(13) E.F. Barry and R.L. Grob, Columns for Gas Chromatography Performance and Selection (John Wiley and Sons, New York, 2007), pp. 32–44.

(14) C. Yao and J.L. Anderson, J. Chromatogr. A 1216 1658–1712 (2009).

(15) “Introduction to Ionic Liquid Columns” https://www.sigmaaldrich.com/content/dam/sigma-aldrich/docs/Supelco/Posters/1/ionic_liquid_gc_columns.pdf (Accessed September, 2018).

(16) C. Ragonese, D. Sciarrone, P.Q. Tranchida, P. Dugo, G. Dugo, and L. Mondello, Anal. Chem. 83 7947–7954 (2011).

FIGURE 6: Chromatogram and calculation of Kovats retention index. Adapted from ChromAcademy (www.chromacademy.com, accessed September 2018).

Nicholas H. Snowis the Founding Endowed Professor in the Depart-ment of Chemistry and Biochemistry at Seton Hall

University. He is also the university’s Director of Research and Adjunct Pro-fessor of Medical Science. During his 30 years as a chromatographer, he has published more than 60 refereed arti-cles and book chapters and has given more than 200 presentations and short courses. He is interested in the funda-mentals and applications of separation science, especially gas chromatography, sampling, and sample preparation for chemical analysis. His research group is very active, with ongoing projects using GC, GC–MS, two-dimensional GC, and extraction methods including head-space, liquid–liquid extraction, and sol-id-phase microextraction.

John V. Hinshaw“GC Connections” editor John V. Hinshaw is a Senior Scientist at Serveron Cor-poration in Beaver ton,

Oregon, and a member of LCGC’s editorial advisory board. Direct corre-spondence about this column to the author via e-mail: [email protected]

ABOUT THE COLUMN EDITOR

ABOUT THE AUTHOR

Experience the New Proficiency Testing PortalEnsuring trust and confidence through accurate results every time!

After serving thousands of customers and their 400,000 PT samples over 20 years, we are redefining what PT can do for you.

Discover the next generation of proficiency testing

User friendly data entry * Graphical Reports * Trend data with "MyStats" Over 20 new and improved features to enhance your data entry experience!

We provide you with the essential performance data and premium grade testing components that help you build a valuable quality control asset.

Powerful PT web service provided by QuoData QuoData contributes its long-term experience in the field of analytical quality assurance, PT and web services to make the new portal stand out.

Get more out of your proficiency testing with our new PT portal.

To connect to the new portal: MilliporeSigma-pt.com

Explore more at SigmaAldrich.com/PT

Contact: [email protected]

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

© 2018 Merck KGaA, Darmstadt, Germany and/or its affiliates. All Rights Reserved. MilliporeSigma and the vibrant M 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.

Lit. No. MS_AD2982EN Ver. 1.0 2018-12840 10/2018

Lit. No. MS_AD2967EN 2018-17066 10/2018

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

© 2018 Merck KGaA, Darmstadt, Germany and/or its affiliates. All Rights Reserved. MilliporeSigma and the vibrant M are trademarks of Merck KGaA, Darmstadt, Germany or its affiliates. All other trademarks are the property of their respective owners. Detailed informationon trademarks is available via publicly accessible resources.

Brighter

StandardsResults are only as accurate as your reference

We manufacture and fully certify our CRMs to the highest industry standards:

ł Four production sites doubly accredited toISO 17034/Guide 34 and ISO/IEC 17025

ł��8VH�RI�YDOLGDWHG�RU�TXDOL¿HG�WHVWLQJ�PHWKRGV

ł��&HUWL¿FDWH�RI�$QDO\VLV��LQFOXGLQJ�XQFHUWDLQWLHV�and traceability to meet regulatory requirements

ł Real-time assessment of stabilityand setting of expiry datesand storage conditionsbasHG�RQ�VFLHQWL¿F�GDWD

Because results are onlyas accurate as yourUHIHUHQFH��WUXVW�XV�ZLWKyour quality and accuracy.

For more informationSigmaAldrich.com/standards

Ico

n I

ma

ge

: (C

olu

mn

s) J

oe

Zu

gc

ic/Z

ug

cic

Ph

oto

gra

ph

ers

, In

c.

814 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

FOCUS

ON

BIOPHARMACEUTICAL

ANALYSIS

Anurag S. Rathore, Ira S. Krull, and Srishti Joshi

With increasing importance being

attached to structural attributes of

biotherapeutics and subsequent biosimilars,

the analytical techniques used for charac-

terizing these attributes have also evolved.

There is an ever-increasing interest in attain-

ing a higher structural resolution of these

products (1,2). In view of the complexity

exhibited by biopharmaceutical products, it

has become the norm to use a multitude of

orthogonal, high-resolution tools for charac-

terization. These tools are carefully chosen

such that the platform covers all the critical

structural, physicochemical, immunochemi-

cal, and biologically relevant characteristics of

the molecule (3–5). Attributes to be covered

in biotherapeutic characterization, as per

World Health Organization (WHO) guide-

lines to evaluate quality, safety and efficacy

(2), are listed in Table I. This review focuses

on the utility of the traditional characteriza-

tion tools and covers the new generation of

analytical hardware that is increasingly being

used orthogonally to the traditional toolbox.

Tools for Analytical

Characterization

Cost-effectiveness and relative ease of

use have made Poly-Acrylamide Gel

Electrophoresis (PAGE) one of the most

commonly used techniques for protein

analysis. In the case of biotherapeutic

characterization, PAGE is typically used to

estimate the size and isoelectric point of

the molecule. It serves both as a detecting

as well as resolving technique, and relies

on the property of charged molecules to

migrate in an electric field. Proteins are

resolved on a polyacrylamide matrix, either

in their native form (non-denaturing PAGE)

or in reduced form (SDS-PAGE). A combi-

nation of the two can also be employed

for resolving complex molecules such as

monoclonal antibodies (mAbs), where sep-

aration from other proteins is attained in a

non-denaturing first dimension followed by

a denaturing second dimension resolution

(2D-PAGE). Iso-Electric Focusing (IEF), which

resolves molecules based on their iso-elec-

tric point is also a commonly used platform

for 2D-PAGE and provides higher resolution,

either in the first or the second dimension

when compared with 2-D separation based

only on size. Using Trastuzumab as an

example, 2D-PAGE has also been shown to

be a quick and easy method for qualitative

evaluation of charge heterogeneity, stability

and post-translational modifications. (6).

An interesting recent introduction in the 2D

platform has been of 2D-Difference Gel Elec-

trophoresis (2D- DIGE). Proteins are directly

labelled with fluorescent dyes (CyDyes),

pooled and separated on a 2D-PAGE. The

dye binds covalently to ε-amino groups

of lysine residues in proteins, allowing for

accurate quantification of spots. The gels

are scanned using an instrument capable

of detecting different CyDye independently.

2D-DIGE can further be coupled with mass

spectrometry for protein identification (7).

A major advantage of PAGE is its versa-

tility. Gel matrix can be easily modified to

achieve a specific resolution. Gradient gels

(pH gradient) are routinely used to ascer-

tain the isoelectric point. Moreover, PAGE

can be coupled with Mass Spectrometry

for identification on specific gel bands (first

dimension) or spots (second dimension) (8, 9).

High resolution, varied choice of phase

and robustness have made High Performace

Liquid Chromatography (HPLC) a corner-

stone of biopharmaceutical analysis. It is

ubiquitously used for analysis of proteins,

nucleic acids, or small molecules in com-

plex mixtures. Typical separation is achieved

by using a liquid mobile phase and a solid

stationary phase. Diversity in solid phase

chemistry and choice of mobile phase

allows the analyst to fine-tune the type of

interactions allowed between the analyte

and the stationary phase. This yields unpar-

alleled selectivity between a biotherapeutic

and related variants and impurities, which

otherwise may have near identical physi-

cochemical properties. Some of the com-

monly used modalities of HPLC include:

• Ion Exchange (IEX) Chromatography

is used to separate molecules based

on their total charge. It enables the

separation of molecules based on

their charge. The strength of binding

is determined by the affinity of the

In the first part of this series, we discussed the various quality attributes that are pertinent to a biotherapeutic. In this

issue, we will present the current and evolving practices that are being used for analysis of these attributes.

Analytical Characterization of Biotherapeutic Products Part II: The Analytical Toolbox

FOCUSED ON

PRODUCTIVITY

Parker offers a comprehensive range of gas generation solutions. Parker gas generators

deliver convenience, stability, reliability, performance and cost benefits enabling you to

focus on science. Unlike cylinders, which can run out mid-process, gas generators are

able to produce gas continuously with very little maintenance required.

labgasgenerators.com

800.343.4048

PERFORMANCE

SAFETY

EFFICIENCY

PROFITABILITY

816 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

proteins to the “Ion-Exchanger” linked

to the resin (stationary phase). Cat-

ionic exchangers possessing negative

charge bind positively charged entities,

whereas anionic exchangers bind neg-

atively charged entities. Furthermore,

ion-exchangers can be weak or strong

depending upon the range of pH within

which they can sustain their charge. This

influences the range and type (strong/

weak) of binding that can be achieved

(10). Elution is typically achieved by either

altering the pH of the mobile phase or

increasing the ionic concentration of the

mobile phase (salt gradient IEX). A com-

monly used application involves sep-

aration of charged variants, which are

similar in size but differ in charge, using

cation exchange HPLC (11).

• Size Exclusion Chromatography (SEC)

enables separation of molecules based

on their size. The stationary phase con-

sists of a gel containing beads of a

specific pore distribution. The choice

of pore distribution allows the user to

achieve separation of species in the

desired range of size. In the case of

biotherapeutics, a popular application

is the resolution of size based hetero-

geneities, especially aggregates (12).

Co-eluting host cell proteins can also

be resolved using SEC (9).

• Hydrophobic Interaction Chroma-

tography (HIC) separates molecules

based on their hydrophobicity and

their charge. It is a relatively gentle

separation technique as the chosen

conditions are minimally denaturing

and as a result do not significantly

affect the biological activity of the

protein. Traditionally used as a pol-

ishing step in monoclonal antibody

purification, it has also been used

to characterize drug distribution in

antibody-drug conjugates (ADCs) by

exploiting the hydrophobicity of the

conjugated small molecule (13).

• Reverse Phase (RP) Chromatography

exploits reversible adsorption of bio-

molecules based on their hydrophobic-

ity under conditions where the stationary

TTABLE I: ,,, Techniques used for analytical characterization as mentioned in WHO and ICH Q6b guidelines. CD - Circular Dichroism,-DSC - Dynamic Light Scattering, FTIR - Fourier Transform Infra-Red Spectrometry, LC - Liquid Chromatography, MS - Mass Spec--trometry, NMR - Nuclear Magnetic Resonance, Cryo-EM - Cryo Electron Microscopy, IEX - Ion Exchange Chromatography, CE - Cap-

illary Electrophoresis, IEF - Isoelectric Focusing, RP-HPLC - Reverse Phase High Performance Liquid Chromatography, SEC - Size Exclusion Chromatography, UV - Ultraviolet, MALS - Multi Angle Light Scattering, FFF - Field Flow Fractionation, AUC - Analytical

----Ultracentrifugation, SDS-PAGE - Sodium Dodecyl Sulfate Poly Acrylamide Gel Electrophoresis, TEM - Transmission Electron Micros-copy, ELISA- Enzyme Linked Immunosorbent Assay, SPR - Surface Plasmon Resonance, ITC - Isothermal Titration Calorimetry, BLI --- Bio-Layer Interferometry.

Type Attributes

WHO (2013) ICH Q6b (1999) OTHERS

Phys-iochemical character-ization

Primary structure

Amino-acid sequence, Molecular weight, extinc-tion-coeffi cient, disulfi de linkage, N-terminal

methionine, signal/leader sequence, N-/C-termi-nal modifi cations, C-terminal processing, N-ter-minal pyroglutamate, deamidation, oxidation, isomerization, fragmentation, disulfi de bond

mismatch, N-/O-linked oligosaccharide, glyco-sylation, aggregation, C-terminal lysine presence

HPLC, MS (ESI, MAL-DI-TOF), MS/MS

HPLC (SEC, RP,IEX, Affi nity), MS,SEC, SDS-PAGE,

IEF, UV-VISspectroscopy,

Western Blot, CE

MALS,DSC

Glycan structure

Glycan content, glycan structure, glycan pattern,glycosylation site, glycan charge pattern

HPLC, Electropho-resis, MS, MS/MS, UV-FLD, CE, IEF

CE-MS

Higher Order Structure

secondary structure, tertiary struc-ture, quarternary structure

X-ray crystallography, NMR, CD, FTIR, Fluo-

rescence, DSC, protonnuclear magnetic

resonance (IH-NMR), Hydrogen-Deuteri-um exchange MS

CD, NMR

Cryo-EM,Raman

spectros-copy

Biologicalcharacter-ization

Effector function, complement bind-ing and activation, potency

ADCC, CDC, Apopto-sis assay, Fc-y receptor binding, Neonatal Fc

receptor binding

Animal-basedbiological assays, cell culture-basedassays, biochem-

ical assays

SPR, BLI,ITC

Immu-nochemicalcharacter-ization

Product related

Affi nity, avidity, immunoreactivity, epitope char-acterization, glycosylation/ PEGylation profi le

Cell-based assaysBinding as-

say, westernblot, ELISA

SPR, BLI,ITC

Impurities, contam-inants

Fragmentation, amino acid modifi cation, Higher molecular weight species, particles

HPLC, Electrophore-sis, MS, CE, SEC, FFF, AUC, Fluorescence,

Light scattering

HPLC, SEC-HPLC, SDS-PAGE, peptide mapping,

CE, MS, CD

MALS, SEC, MFI

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 817

phase is more hydrophobic than the

mobile phase. It is quite similar to HIC

in principle, however, the RPC medium

is much more hydrophobic than in HIC

and elution is achieved by the use of

non-polar, organic solvents. This makes

it a very desirable technique for cou-

pling with MS for peptide mapping and

other comparability studies. Coupled

with mass spectrometry, the reverse

phase has been extensively used in pri-

mary structure characterization of bio-

therapeutics as well as in comparability

studies of biosimilars (14).

High Performance Capillary Electropho-

resis (CE) is another technique increasingly

being applied in conjunction with MS for

charge variant characterization, isoelectric

focusing and biosimilarity assessment. Mol-

ecules are separated across a fine capillary

with the internal diameter as small as 50 μM

via application of high voltage (~30 kV). This

miniaturized format requires minimal sample

with flow rates in the range of nL/min, mak-

ing it a cost-effective technique. CE coupled

with nano-ESI-MS has been shown to be

particularly useful for N-Glycan analysis of

monoclonal antibodies as well as for detec-

tion of charge variants (15). The availability

of various CE modes such as capillary zone

electrophoresis, capillary gel electrophore-

sis, capillary isoelectric focusing and micel-

lar electrokinetic chromatography allows

for characterization of different attributes

such as intact mass, reduced mass, charge

variants, as well as glycosylation pattern. CE

coupled with MS is increasingly being used

as a complementary platform to traditional

LC-MS for biotherapeutic characterization

(16). Innovative integration of CE with MS,

such as in ZipChip has reduced the analysis

time to under three minutes and bypassed

issues of individual component integra-

tion and capillary damage due to handling.

Demonstrated applications include glyco-

sylation profiling of mAb directly from cell

culture with sample volume requirement of

under fifty microliters (17). The technique

shows much promise in monitoring batch to

batch variation in manufacturing as well as in

biocomparability exercise.

X-ray crystallography is considered the

gold standard for protein structural stud-

ies. It works on the principle of X-ray dif-

fraction. The angle and the intensity of the

diffracted beam from a crystal are used to

construct a 3-dimensional image of the

molecular structure. However, applica-

tion of this technique for characterization

and biocomparability is challenging as the

protein of interest has to be purified and

crystallized, which may not be possible for

biotherapeutics due to the presence of

inherent heterogeneity in the form of sev-

eral post-translational modifications (PTMs).

Although the data obtained is a direct mea-

surement of the crystal structure, the tech-

nique itself is too cumbersome to be used

as a routine analytical technique for biosim-

ilar characterization and comparability (18).

Nuclear Magnetic Resonance (NMR)

exploits the magnetic properties of specific

818 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

atomic nuclei. Although a gold standard for

protein structural studies, its routine applica-

tion in the biotherapeutic industry has been

limited due to several factors, including the

large size of protein biopharmaceuticals, the

relatively low sensitivity of the NMR signals,

and the low natural abundance of active

nuclei. However, in instances where biother-

apeutics of small molecular size are being

characterized, NMR might be utilized to gain

in-depth HOS information (18).

It should be noted that although both

X-ray crystallography and NMR provide

structural details at a near-atomic level, the

techniques differ in their principle, informa-

tion and bottlenecks. X-ray crystallography

requires the formation of the uniform crystal

lattice and utilizes very high energy X-rays

for deflection by the surrounding electron

clouds, NMR is based on the absorption of

electromagnetic radiation in the radio-fre-

quency (RF) range. In NMR, proteins are

analysed in solution and the final image is

a compilation of a number of low energy

states of the protein in different orientations

as compared to single instance images of

X-ray crystallography. Both the techniques

require high concentration, homogenous

sample preparations. With respect to pro-

tein size, NMR is more limiting. Proteins with

a size larger than 40 kDa exhibit a slower

molecular tumbling in solution leading to

spectral overlap and peak broadening. In

terms of information obtained, NMR can

unravel information on structural dynamics

and flexibility more readily than x-ray crys-

tallography which requires time-resolved

experiments to capture structural changes

due to biochemical reactions (19, 20)

Transmission Electron Microscopy (TEM)

is a microscopy technique in which a beam

of electrons is transmitted through a spec-

imen to form an image. Due to the much

smaller wavelength of electrons as com-

pared to light, the resolution of the image is

greater by orders of magnitude with details

up to atomic level, and hence TEM finds

use in the characterization of biotherapeu-

tic aggregates (21). Recent developments

in achieving greater resolution, especially

in Cryo-TEM, have enabled researchers to

observe protein complexes such as mAbs

in their native formulation (without crys-

tallization) (22). The technique is currently

underutilized in the field of biotherapeutics

but has significant potential to grow.

Circular Dichroism (CD) Spectroscopy

is based on the difference observed in the

absorption of left and right-handed circularly

polarized light of a molecule in the presence

of light absorbing chiral groups. This prop-

erty of biomolecules is frequently utilized in

the examination of the secondary structure

of the proteins and is employed for assessing

HOS comparability. Using thermal denatur-

ation, information about molecule stability

as well as folding and unfolding mechanisms

can be elucidated. An application of CD

in the aggregate characterization of mAbs

has also been demonstrated (23, 24).

Fourier Transform Infrared Spectros-

copy (FTIR) is a spectroscopic technique

that monitors the characteristic infrared

absorption of molecules and translates it

into structural information. Similar to CD, it

gives structural information about the sec-

ondary structure of the protein, mainly the

alpha helices and beta sheet. It serves as an

orthogonal technique to CD in characteriza-

tion and biocomparability studies (25).

Although both CD and FTIR can provide

information with regards to components

of the secondary structure of a molecule,

it is only through higher resolution visual-

ization techniques such as CryoTEM, X-ray

crystallography and NMR that the spe-

cific arrangement of these components in

space can be unravelled.

Dynamic Light Scattering (DLS) is com-

monly used to determine the size distribu-

TTABLE II: - Publications per technique since 2010 for biotherapeutics (Google Schol-,ar) LC - Liquid Chromatography, CE - Capillary Electrophoresis, TOF - Time of fl ight,-Q-TOF- Quadruple-time of fl ight, MALDI - Matrix Assisted Laser Desorption ioniza-

tion, TRIPLE-QUAD - Triple Quadrupole, CD - Circular Dichroism, DSC - Dynamic Light Scattering, FTIR - Fourier Transform Infra-Red Spectrometry, NMR - Nuclear Magnetic

Resonance, HDX-MS - Hydrogen/Deuterium exchange-Mass spectrometry, CRYO-TEM - F - Cryo-Transmission Electron Microscopy, TEM - Transmission Electron Microscopy, FFF-- Field Flow Fractionation, SPR - Surface Plasmon Resonance, ITC - Isothermal TitrationCalorimetry. The publication data were generated using Google Scholar for papersfrom 2010-2018 (excluding citations and patents). Search word combination used was

“““““““““““““““ ””””””””””””“technique name” “biopharmaceutical” and “technique name” “biopharmaceutical”and “monoclonal antibody”.

Tool Category

2010-2017 Biopharmaceuticals Monoclonal antibodies

HPLC 7440 1600

CE 1880 1040

HPCE 109 44

ESI-TOF 2140 703

Q-TOF 559 441

MALDI-TOF 2630 835

ORBITRAP 882 603

TRIPLE-QUAD 39 251

HDX-MS 406 665

CD 1750 781

DSC 1670 847

FTIR 1720 362

DLS 4330 1080

NMR 3550 796

CRYO-TEM 144 24

TEM 3760 650

FFF 550 307

SPR 2060 997

ITC 505 156

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 819

tion profile of small particles in biothera-

peutic formulations. The particle size profile

is determined by measuring the random

changes in the intensity of light scattered

from the sample solution. It is often used in

conjunction with SEC and analytical ultracen-

trifugation for aggregate studies. An interest-

ing application of High-throughput platform

in DLS (HT-DLS) has been in screening stud-

ies to quantify viscosity of mAb formulations

(26). Using automated HT-DLS, effects of buf-

fer conditions and temperature on aggre-

gate formation have also been reported (27)

Raman Spectroscopy is used to observe

vibrational, rotational, and other low-fre-

quency modes in a system, generally applied

to study the secondary structure of proteins

by studying the Amide I band between 1600

and 1700 cm−1. An advantage that Raman

spectroscopy offers over other secondary

structure characterizing techniques is its

low susceptibility to water interference as it

detects scattered light and water is inefficient

at scattering in this part of the spectrum (28).

In a recent study, Raman Optical Activity

(ROA) was evaluated as a means to detect

early thermal instability in mAb samples kept

at 50 °C for a month. Significant structural

changes could be observed at one week of

stress. This provides an advantage over SEC

in monitoring aggregation as ROA would

provide information about subtle differences

in tertiary structure whereas Raman/ROA

spectra can elucidate on changes at the

secondary structure level (29). Another inter-

esting recent application of Raman spectros-

copy has been in drug identification, spe-

cifically in the identification of monoclonal

antibodies by exploiting subtle differences

in vibrational modes of the antibodies (30).

It would be interesting to see if this applica-

tion can be further modified to distinguish

between biosimilars and innovator product.

Analytical Ultracentrifugation (AUC) is

a versatile tool for quantitative analysis of

macromolecules in solution. It employs the

principle of centrifugal acceleration to sep-

arate particles based on their size and mass.

Two types of hydrodynamic analyses, namely

sedimentation velocity and sedimentation

equilibrium, are able to make distinctions

in formulation components based on shape

and mass or mass alone, respectively. Sed-

imentation velocity is a mode of choice for

aggregate analysis because it causes physi-

cal separation of molecular species of differ-

ent mass or shape. This is a matrix-free tech-

nique as no column or gel matrix is required

for size fractionation to occur (31). AUC-Sed-

imentation Velocity (AUC-SV) is often used

to quantify high molecular weight species

present in biopharmaceuticals (32).

Multiple Angle Light Scattering (MALS) is

typically used in line with a resolving tech-

nique such as SEC to determine the size of

the different molecular species as they pass

through the MALS detector. It adds a quanti-

tative measure of analysis to techniques like

SEC. As the sample passes through the laser

beam, light is scattered at multiple angles

and the detector collects this data to approx-

imate the sample size (33). It has been used

www.tosohbioscience.com

Tosoh Bioscience and TSKgel and are registered trademarks of Tosoh Corporation.

Accurate results every time!Contact us: 800-366-4875, option #4

New multiple pore size columnsfor extended linear separation range

TSKgel® SuperMultiporeGPC/SEC columns

Individual particles with broad pore size distribution

Semi-micro columns

• Each particle has a linear calibration curve – no infl ection point

• No chromatogram distortion for more accurate molecular weight distributions

• Small particle size (3 to 6 μm) and smaller column dimensions (4.6 mm ID × 15 cm L)

• Higher throughput, higher resolution, reduced solvent consumption

SuperMultipore columns

Pure packings

with multi-pore size distribution

(TSKgel SuperMultiporeHZ column)

Multiple Pore Sizes

101

102

103

104

105

106

107

108

1.5 2.5 3.5 4.5 5.5 6.5

Retention time (minutes)

Log

mo

lar

ma

ss

TSKgel SuperMultiporeHZ-N

TSKgel SuperMultiporeHZ-M

TSKgel SuperMultiporeHZ-H

- 20

0

20

40

60

80 TSKgel SuperMultipore HZ-M

Competitive mixed bed column

100

6 8 10 12 14 16 18 20

-10

10

30

50

70

90

10 20

Retention time (minutes)

De

tec

tor

resp

on

se (

mV

)

30 40

Conventional columns ×4

TSKgel SuperMultiporeHZ-N ×4

TSKgel® SuperMultiporeGPC/SEC columnsTSKgel® SuperMultiporeGPC/SEC columnsTSKgel® SuperMultiporeGPC/SEC columnsTSKgel® SuperMultiporeGPC/SEC columns

820 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

to monitor the formation of soluble, HMWS

so as to better quantify and model non-na-

tive aggregation kinetics in α chymotrypsino-

gen (34). Composition-Gradient Multi-Angle

Light Scattering (CG-MALS), a variation on

MALS, employs a series of unfractionated

samples of different composition or concen-

tration in order to characterize a wide range

of macromolecular interactions. CG-MALS, a

complementary technique to AUC and DLS,

has been used to characterize self-associa-

tion in model mAb molecule (35). Although

not yet commonly used, CG-MALS could

be used orthogonally to Surface Plasmon

Resonance (SPR) to study receptor bind-

ing kinetics in biotherapeutics. Unlike SPR,

CG-MALS would not require binding of

the receptor to any chip and hence would

be a truly label-free technique to quantify

interactions, similar to Isothermal Titration

Calorimetry (ITC). Although the latter is

more suited to study the thermodynamic

parameters of an interaction.

Field Flow Fractionation (FFF) is a unique

separation technique used for analysis of

aggregates. Samples are pumped into a

narrow tube perpendicular to the flow and

separation occurs due to the difference

in mobility of the species in the mixture

under the field applied. Depending upon

the properties of the species in the sample

mixture, different flows such as electrical,

magnetic, thermal-gradient, gravitational

or centrifugal can be applied. FFF serves

as a complementary technique to DLS and

MALS for determining the presence of

sub-micron particles in the sample (36).

Fluorescence spectrometry is based on

the principle that certain molecules called

fluorophores emit light upon excitation by

an external source such as an incandescent

lamp or a laser which produces a spectrum.

This technique is helpful in exploring HOS

of a protein (to an extent), mainly the tertiary

structure via the intrinsic fluorescence of the

protein. Changes in the local environment

of tryptophan, the strongest intrinsic fluo-

rophore, are reflected in the emission spec-

tra of the molecule and are a measure of

change in the protein tertiary structure (37).

In cases where a biomolecule does not have

an intrinsic fluorophore, extrinsic fluorophore

dyes such as Thioflavin-T (ThT) or 1-anili-

no-8-naphthale-nesulfonate (ANS) can be

used to induce fluorescence. Fluorescence is

routinely used in thermal stability studies of

biotherapeutics, especially monoclonal anti-

bodies (38). At best, it is an indirect measure

of changes in a protein’s tertiary structure and

does not provide information about position

and the specific nature of these changes.

Micro-Flow Imaging (MFI) is an up and

coming technique that combines digital

microscopy with microfluidics to capture

and quantify sub-visible particles in the

range of 1 to 300 μm in a solution. It can

provide information about particle size,

concentration and morphology (39). How-

ever, rather than protein characterization, it

is more suited for profiling and classifying

the particulate size in a given formulation.

Mass Spectrometry (MS) is a powerful

and data-intensive technique for deter-

mining protein mass (intact, fragmented

and reduced), sequence, and for probing

and quantifying protein modifications.

MS involves ionization of the sample

fragments followed by their separation

based on their mass to charge ratio (m/z).

By accelerating the ionized particles and

subjecting them to an electric or magnetic

field, the ions get deflected depending on

the mass and charge that they carry. Ions

are detected by an electron multiplier and

the results are displayed as a spectrum of

the relative abundance of different ions

based on the m/z ratio.

Analytical capability of an MS platform is

defined by the kind of ionization source and

the type of mass analyser being used. Exam-

ples of ionization sources include fast atom

bombardment (FAB), chemical ionization

(CI), atmospheric-pressure chemical ioniza-

tion (APCI), electrospray ionization (ESI), and

matrix-assisted laser desorption/ionization

(MALDI). Examples of mass analyzers include

Time-of-flight (TOF), quadrupole mass filter,

and ion-traps. Some of the popular combi-

nations of the two are ESI-TOF, MALDI-TOF

and ESI-Q-TOF (Table II).

MS is usually coupled with LC or CE as

the first dimension of separation. Charac-

terization of complex molecules such as

monoclonal antibodies requires mapping

of the different fragmentation patterns in

MS such as Electron Transfer Dissociation

(ETD) and Collision-Induced Dissociation

(CID) along with the use of different pro-

teases make it possible to map disulphide

links present and the different glycans

attached to the protein (18,40).

Another useful application of MS, when

coupled with Hydrogen/Deuterium

exchange (HDX) is in elucidating protein

conformational dynamics and protein inter-

actions. In the biopharmaceutical industry,

HDX-MS has established itself in the analysis

of protein-small molecule interactions, char-

acterization of bio-therapeutics/biosimilars,

and epitope mapping of biotherapeutics.

The technique relies on isotope labelling to

probe the rate at which protein backbone

amide hydrogens undergo exchange. MS

is then used to monitor the mass-shift as a

result of incorporation of deuterium in the

protein. The rate of exchange provides infor-

mation regarding the conformational mobil-

ity, hydrogen bonding strength, and solvent

accessibility in protein structure (41,42)

Differential Scanning Calorimetry (DSC)

is a versatile technique that measures the

quantity of heat radiated or absorbed by

the sample on the basis of a temperature

difference between the sample and the

reference material. It is used to determine

equilibrium thermodynamic stability and

folding mechanism of proteins and finds

routine use in thermal stability characteriza-

tion of biotherapeutics (43).

Tools for Functional Characterization:

Enzyme-linked Immunosorbent Assay

(ELISA) is a popular diagnostic technique

used to assess the immunogenicity of a ther-

apeutic product. In a typical assay, a ligand,

typically an antigen, is non-specifically or spe-

cifically bound to the polystyrene well of a 96

well microtiter plate. Enzyme-linked antibod-

ies are used for colorimetric detection of a

positive interaction upon addition of the

enzyme substrate. ELISA has several appli-

cations in biotherapeutic characterization. A

common application of this assay format is in

Host Cell Protein analysis where polyclonal

antibodies raised to the host cell are used

to detect the presence of HCPs in the sam-

ple (44). However, with high specificity tech-

niques such as MS (detection) and Surface

plasmon resonance (interaction) becom-

ing affordable and routine, ELISA can only

be used for a precursor technique for quick

estimation with limited confidence prior to

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 821

employing tools with much higher specificity.

Surface Plasmon Resonance (SPR)

(interaction) is a label-free technique

that allows for real-time detection of

biomolecular interactions. The SPR phe-

nomenon occurs when polarized light

strikes an electrically conducting surface

at the interface between two media. In

response, plasmons are produced, which

are electron charged density waves.

These waves reduce the intensity of

reflected light at a specific angle known

as the resonance angle, in proportion to

the mass on a sensor surface. SPR allows

for real-time monitoring of both associ-

ation and dissociation of an interaction,

generating reproducible kinetic data.

Due to its sensitivity, ease of use and

automated data analysis, it is widely used

to study the ligand-receptor kinetics of

monoclonal antibodies (14)

Bio-Layer Interferometry (BLI) is a label-

free, optical analytical technique that ana-

lyzes the interference pattern of white light

reflected from two surfaces: a layer of

immobilized protein on the biosensor tip

and an internal reference layer. Changes

occurring in the number of molecules

bound to the biosensor tip is reflected as a

shift in the interference pattern. Similar to

SPR, binding kinetics can be determined

by this technique. Because of its robust-

ness and ease of implementation, BLI is

gaining application as a complementary

technique to SPR for studying and com-

paring biotherapeutic binding kinetics (44).

Isothermal Titration Calorimetry (ITC) is a

physical technique used to determine the

thermodynamic parameters of interactions

in a solution. It measures the heat released

or absorbed by mixing the two interactants

via titration. It is widely considered an abso-

lute and direct measurement of interaction.

Thermodynamic parameters of an interac-

tion can be assessed via this technique and

it is fast gaining importance as an orthog-

onal technique to SPR for ligand binding

studies in biocomparability (45).

Summary

With the increase in complexity of the bio-

therapeutics undergoing manufacturing, the

need for in-depth analytical characterization

has also increased. As protein molecules,

even minute changes in the structure of the

biotherapeutic can confer altered functional-

ity, sometimes leading to immunogenic reac-

tions in the patients. In view of our depen-

dency for production of these molecules

on biological machinery, the formation of

numerous altered conformations of the mol-

ecule is unavoidable. However, significant

advancements have been made in analytical

methodology increasing our ability to char-

acterize a biotherapeutic. This is highlighted

by the increase in the number of technique

rich publications on analysis of biotherapeu-

tics since 2010 and the introduction of the

US-FDA BPCI Act 2009 (Table II).

Our understanding of the relevance of the

different measured attributes with respect to

the safety and efficacy of a biotherapeutic

has been improving with time and experi-

• Over 5,000 impurity, API and excipient pharmaceutical reference standards

• &RPSUHKHQVLYH�&HUWL¿FDWH�RI�$QDO\VLV

• Expert technical support

• 5 years of manufacturing excellence

lgcstandards.com/mikromol [email protected]

A global leader in the production and distribution of pharmaceutical reference standards

822 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

ence. Some recently evolving techniques

such as CG-MALS for macromolecular

interactions and intrinsic Förster resonance

energy transfer (iFRET) for in vivo target pro-

tein detection are yet to find their place in

the biotherapeutic characterization toolbox

while other well-established techniques such

as Raman spectroscopy are being increas-

ingly used for novel applications such as

drug identification. A desirable direction for

technological advancements would be the

increasing throughput of techniques such

as LC by parallelization (9). Short analysis

time and micro- and nanoplatforms with

minimal sample consumption would help

cut down the cost of analysis associated

with limited and expensive samples such as

monoclonal antibodies.

Despite these advancements, we are yet

to reach a point where a product such as a

complex biotherapeutic, can be completely

fingerprinted in a manner similar to a phar-

maceutical (small molecule) product. More-

over, each technique comes with its unique

limitations and pitfalls. This ensures that the

topic of analytical characterization of bio-

therapeutics will continue to be an area of

research in the time to come.

References

(1) ICH Expert Working Group, Specif. Test Proced. Accept. Critreia Biotechnol. Prod. (March), 1–20 (1999).

(2) WHO, Guidelines on the Quality, Safety and Efficacy of Biotherapeutic Protein Products Prepared by Recombinant DNA Technology, WHO Technical Report Series 814, 91 (2013)

(3) L.A. Bui, S. Hurst, G.L. Finch, B. Ingram, I.A. Jacobs, C.F. Kirchhoff, C.K. Ng, and A.M. Ryan, Drug Discov. Today 20(S1), 3–15 (2015).

(4) A.S. Rathore, Trends Biotechnol. 27(12), 698–705 (2009).

(5) A. AL-Sabbagh, E. Olech, J.E. McClellan, and C.F. Kirchhoff, Semin. Arthritis Rheum. 45(5), S11–S18 (2016).

(6) D. Nebija, C. Noe, E. Urban, and B. Lach-mann, Int. J. Mol. Sci. 15(12), 6399–6411 (2014).

(7) R. Diez, M. Herbstreith, C. Osorio, and O. Alzate, 2-D Fluorescence Difference Gel Electrophoresis (DIGE) in Neuroproteomics Neuroproteomics (CRC Press/Taylor & Francis, 2010). at <http://www.ncbi.nlm.nih.gov/pubmed/21882449>.

(8) C. Reichel and M. Thevis, Bioanalysis 5(5), 587–602 (2013).

(9) S. Fekete, D. Guillarme, P. Sandra, and K. Sandra, Anal. Chem. 88(1), 480–507 (2016).

(10) J. R. Auclair, A. S. Rathore Chitra, and I. S. Krull, LCGC North Am. 36(1), 26–36 (2018).

(11) S.K. Singh, G. Narula, and A.S. Rathore, Electrophoresis 37(17–18), 2338–2346 (2016).

(12) A. Singla, R. Bansal, V. Joshi, and A.S. Rathore, AAPS J. 18(3), 689–702 (2016).

(13) A. Wakankar, Y. Chen, Y. Gokarn, and F.S. Jacobson, MAbs 3(2), 164–175 (2011).

(14) N. Nupur, N. Chhabra, R. Dash, and A.S. Rathore, MAbs 10(1), 143–158 (2018).

(15) K. Wooding, W. Peng, and Y. Mechref, Curr. Pharm. Biotechnol. 17(9), 788–801 (2016).

(16) M. Han, B. M. Rock, J. T. Pearson, Y. Wang, and D. A. Rock, Therapeutic Monoclonal Antibody Intact Mass Analysis by Capillary Electrophoresis–Mass Spectrometryin Cap-ill. Electrophor. Spectrom., 13–34 (Springer International Publishing, Cham, 2016). doi:10.1007/978-3-319-46240-0_3.

(17) S.A. Berkowitz, J.R. Engen, J.R. Mazzeo, and G.B. Jones, Nat. Rev. Drug Discov. 11(7), 527–540 (2012).

(18) S.A. Berkowitz, Analytical Characterizationin Biosimilar Drug Prod. Dev., 15–82 (2017). doi:10.1201/9781315119878-3.

(19) B. Carragher, A. Schneemann, J.J. Sung, S.K. Mulligan, J.A. Speir, K. On, and C.S. Potter, Microsc. Microanal. 21(33), 2014–2015 (2015).

(20) J. T. Yang, C.-S. C. Wu, and H. M. Martinez, Methods Enzymol. 130, 208–269 (1986). doi:10.1016/0076-6879(86)30013-2.

(21) V. Joshi, T. Shivach, N. Yadav, and A.S. Rathore, Anal. Chem. 86(23), 11606–11613 (2014).

(22) L.R. Tsuruta, M. Lopes dos Santos, and A.M. Moro, Biotechnol. Prog. 31(5), 1139–1149 (2015).

(23) F. He, G. W. Becker, J. R. Litowski, O. L. Narhi, D. N. Brems, V. I. Razinkov, Anal. Bio-chem. 399(1), 141–143 (2010).

(24) L. Aileen, A. Seneviratne, G. Ratnaswamy, and J. Park, Pharm. Technol. 38(10), 32–39 (2015).

(25) R.J. Falconer, D. Jackson-Matthews, and S.M. Mahler, J. Chem. Technol. Biotechnol. 86(7), 915–922 (2011).

(26) G. Thiagarajan, E. Widjaja, J.H. Heo, J.K. Cheung, B. Wabuyele, X. Mou, and M. Sha-meem, J. Raman Spectrosc. 46(6), 531–536 (2015).

(27) S.K. Paidi, S. Soumik, R. Strouse, J.B. McGivney, C. Larkin, I. Barman, Anal. Chem. 88(8), 4361–4368 (2016).

(28) T. Arakawa, J.S. Philo, D. Ejima, K. Tsumoto, and F. Arisaka, Bioprocess Int. 4, 42–43 (2006).

(29) L. Wafer, M. Kloczewiak, and Y. Luo, AAPS J. 18(4), 849–860 (2016).

(30) A. Oliva, M. Llabrés, and J.B. Fariña, J. Pharm. Biomed. Anal. 25(5–6), 833–841 (2001).

(31) Y. Li, W. F. Weiss, and C. J. Roberts, J. Pharm. Sci. 98(11), 3997–4016 (2009).

(32) R. Esfandiary et al., J. Pharm. Sci. 102(9), 3089–3099 (2013).

(33) J. Liu, T. Eris, C. Li, S. Cao, and S. Kuhns, Bio-Drugs 30(4), 321–38 (2016).

(34) P. Garidel, M. Hegyi, S. Bassarab, and M. Weichel, Biotechnol. J. 3(9–10), 1201–1211 (2008).

(35) M. Weichel, S. Bassarab, and P. Garidel, Bioprocess Int. (5), 42–52 (2008). at <http://www.bioprocessintl.com/manufacturing/monoclonal-antibodies/probing-ther-mal-stability-of-mabs-by-intrinsic-trypto-phan-fluorescence-182648/>.

(36) M.K. Joubert, Q. Luo, Y. Nashed-Samuel, J. Wypych, and L.O. Narhi, J. Biol. Chem. 286(28), 25118–33 (2011).

(37) A. Guttman, LCGC North Am. 30(5), 412–421 (2012).

(38) C.M. Johnson, Arch. Biochem. Biophys. 531(1–2), 100–109 (2013).

(39) D.G. Bracewell, R. Francis, and C.M. Smales, Biotechnol. Bioeng. 112(9), 1727–1737 (2015).

(40) U. Sinha-Datta, S. Khan, and D. Wadga-onkar, Biosimilars 5, 83–91 (2015).

(41) C.A. Challener, BioPharm Int. 28(1) (2015).

(42) T.K. Dam, M. Torres, C.F. Brewer, and A. Casadevall, J. Biol. Chem. 283(46), 31366–70 (2008).

(43) M.G. Petroff, H. Bao, J.P. Welsh, M. van Beuningen-de Vaan, J.M. Pollard, J.D. Roush, S. Kandula, P. Machielsen, N. Tugcu, and T. O. Linden, Biotechnol. Bioeng. 113(6), 1273–1283 (2016).

(44) V. Kamat and A. Rafique, Anal Biochem. 536, 16-31 (2017)

(45) S. Perspicace, A.C. Rufer, R. Thoma, F. Muller, M. Hennig, S. Ceccarelli, T. Schulz-Gasch and J. Seelig, FEBS Open Bio. 3, 204-211 (2013)

Anurag S. Rathoreis a professor in the Depart-ment of Chemical Engineer-ing at the Indian Institute of Technology in Delhi, India.

Ira S. Krullis a Professor Emeritus with the Department of Chemis-try and Chemical Biology at Northeastern University in

Boston, Massachusetts, and a member of LCGC’s editorial advisory board.

Srishti Joshiis a post-doctoral research fellow in the Bioseparations and Bioprocessing Lab, under the tutelage of Pro-

fessor Anurag. S. Rathore, Department of Chemical Engineering, Indian Insti-tute of Technology, Delhi.

ABOUT THE AUTHORS

Join thousands of chemists and scientists from around the world at Pittcon, the

leading annual conference and exposition for laboratory science. This all-in-one event

ŅýåųŸ� ±� ĘĜčĘěϱĬĜÆåų� ƋåÏĘĺĜϱĬ� ŞųŅčų±ĵØ� ŸĩĜĬĬěÆƚĜĬÚĜĺč� ŸĘŅųƋ� ÏŅƚųŸåŸ� ±ĺÚ� ±� ÚƼĺ±ĵĜÏ�

ĵ±ųĩåƋŞĬ±Ïå�Ņü� ƋĘå� Ĭ±ƋåŸƋ�ŸÏĜåĺƋĜĀÏ� ĜĺŸƋųƚĵåĺƋ±ƋĜŅĺ�±ĺÚ�ŸåųƴĜÏåŸţ��Ƌ±ųƋ�ÏŅĬĬ±ÆŅų±ƋĜĺč�

ƵĜƋĘ�ĜĺÚĜƴĜÚƚ±ĬŸ�Ĝĺ�±�ƴ±ųĜåƋƼ�Ņü�ŸÏĜåĺƋĜĀÏ�ÚĜŸÏĜŞĬĜĺåŸ�±ĺÚ�ĀĺÚ�ŸŅĬƚƋĜŅĺŸ�ƋŅ�ƼŅƚų�čųå±ƋåŸƋ�

laboratory challenges at Pittcon 2019.

Pennsylvania Convention Center | Philadelphia, PA | March 17 - 21 | www.pittcon.org

The Visible DifferenceIn Laboratory Science Expositions

824 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

Transferring chromatographic meth-

ods among users and laborato-

ries is a common and important activity

that has often proven more difficult than

expected. This is the second installment

of a three-part discussion focused on the

most rigorous method transfer leading

to the duplication of results of the estab-

lished methods. The first installment (1)

described the aspects of the method

that specifically affect the transfer of the

method. In the future, the final part of this

series will consider the details of aligning

individual instrument modules. In this part,

we address the chromatographic systems

and how they may be characterized and

compared for use in the transfer. It is gen-

erally assumed that modern instrumenta-

tion delivers the volumes, temperatures,

and so on that are programmed in the

control software. That is generally true for

well-maintained systems. There are, how-

ever, subtle differences in the exact deliv-

ery and conditioning of flow that can have

significant effects on the chromatographic

results. Such details occur in all the system

modules. We discuss here the characteriza-

tion of the systems to identify these opera-

tional differences.

General Considerations

The chromatographic instrument itself is

often the largest contributor to inconsis-

tencies in the transfer of methods. The

common principle applied for all other

considerations, “Use exactly what was used

in the originator’s laboratory,” is desirable

here. It is, however, very often impossible

to maintain that consistency. The instru-

ments used in various laboratories are

often different models or brands, and it

is not usually financially sensible to pur-

chase chromatography instruments for

each specific new method to be imple-

mented. Furthermore, the usable life-

time of a method is often much longer

than that of an instrument. Duplicating

instruments, therefore, may not be pos-

sible to begin and execute a method

transfer. We must, then, consider the

differences among instruments that can

affect method transfer. The transfer of a

method from one instrument to another

may require some adjustment of the

method. Many laboratories adhere to

the guidelines found in Chapter 621 of

the current United States Pharmaco-

peia. The currently applicable chapter

specifically states:

Adjustments to the specified chro-

matographic system may be nec-

essary in order to meet system suit-

ability requirements. Adjustments to

chromatographic systems performed

in order to comply with system suit-

ability requirements are not to be

made in order to compensate for col-

umn failure or system malfunctions.

Adjustments are permitted only when

. . . adjustments or column change

yields a chromatogram that meets all

the system suitability requirements

specified in the official procedure (2).

These guidelines, often mentioned as

“<621>”, specify changes to the method

that may be implemented without revali-

dating the method. The chapter has been

summarized in many places, but the original

document should always be consulted. We

will allude to specific items in these guide-

lines in the context of specific challenges in

method transfer. It should be emphasized

that many laboratories follow these limits

and practices, but they are not universal

regulations. They are absolute require-

ments only for the compendial methods of

the USP.

The Fluid Path

There are important factors associated

with the fluid path in general; the pump-

ing or solvent delivery system; the sample

introduction or injector system, and the

detector. Each component will be consid-

ered for its potential impact on the method

in terms of altered retention time as it may

affect peak identification and resolution;

altered chromatographic selectivity as it

may affect resolution and quantification;

and peak shape as it may affect resolution

and sensitivity.

The fluid path of the instrument includes

all the tubing, and other elements where

liquid moves through the system. To con-

sider the impact on transfer, we must dis-

tinguish between segments that transport

the sample and segments that are only

exposed to the mobile phase. We gen-

erally assume that the fluid is unaltered

during this transport, but this may not be

Thomas E. Wheat

The process of transferring chromatographic methods between users and laboratories is often complicated and time

consuming. This is the second installment of a three-part consideration of this common activity. In part 1, the focus was

on the method itself. In this second part, the techniques and concerns about characterizing the systems in use in the

originating laboratory and the new facility will be described. Given that the scientists executing the transfer often do not

have free access to the originating system, alternative approaches to matching chromatographic results will be considered.

Instrument Considerations in the Transfer of Chromatographic Methods,Part II: System Considerations

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 825

absolutely true. While the instrument may

create trace chemical changes in the mobile

phase, such as metal leaching, this seldom

complicates method transfer, since mate-

rials of construction are consistent across

models and manufacturers. The worrisome

effects are physical. In addition, the tubing

dimensions create some timing differences.

The time to transit the fluid path is often

of great concern to laboratory scientists

using chromatographs and transferring

methods; these scientists frequently com-

ment that tubing must be shortened and

modules placed closer together. In fact,

such changes are almost imperceptible. As

a point of reference, a 3 m piece of 0.005

inch (~127 μm) tubing has a volume of

about 12.7 μL. So the transit time at 1.00

mL/min is only 0.01 min or 0.75 s. Even dou-

bling the tubing diameter only increases the

time to 3 sec. Larger differences are typically

allowed for flow rate or retention time vari-

ability in typical system suitability specifica-

tions. So, there is little reason to focus on

tubing length as a factor in time offset.

The tubing in the system also contrib-

utes some mixing or dispersion, primar-

ily because of laminar flow differences

between the tubing wall and the center.

The magnitude of this effect is dependent

on both the diameter and length of the

tubing. It is important here to distinguish

between parts of the flow path used for

mobile-phase transport only and the parts

downstream from the injector where the

sample is in the flow path. As a general

rule, the upstream parts of the system

can contribute some mixing to blend the

mobile phase. However, as discussed

above, the volumes and the residence

time are so small that there is no significant

contribution to mixing. There is, of course,

substantial solvent blending in all systems,

and we will discuss that below in the con-

text of the pumping system.

Dispersion in the fluid path downstream

from the injector has an impact on both

peak shape and resolution. This has been

considered in detail in other investigations

(3). The largest contribution to dispersion

during sample transport originates with the

tubing diameter, with length as a smaller

contributor. It is, therefore, important during

method transfer to ensure that the tubing

used for connecting the modules of the tar-

get system are the smallest possible diam-

eter and the shortest length. In following

this guideline, however, it is not necessary

to take extreme measures. Consider the

expected volume of the peaks as they elute

from the column. Many standard methods

have peak volumes near or above 50 μL.

The effect of a 10 μL tubing volume will be

relatively small. Even for the most modern

ultra-high performance liquid chromatogra-

phy (UHPLC) methods, with peak volumes

near 10 μL, 0.004 inch (~102 μm) contributes

about 2 μL per foot, and 0.0025 inch (63.5

μm) is less than 1 μL per foot.

When implementing the above tubing

considerations, many scientists overlook

the back pressure that can arise simply

from flow through tubing. At the midpoint

of a water–methanol gradient, at 1 mL/

min, the resistance to flow through 1 m of

a 0.0025 inch (~63.5 μm) tubing generates

a back pressure greater than 5000 psi. For

this reason, the tubing used to assemble a

system should be no smaller than required

for minimizing dispersion.

Pumps

The pumping, or solvent delivery system,

has received the most attention of any

instrument module in terms of its effect on

method transfer. Isocratic separations are

reasonably simple because modern sys-

tems reliably deliver the specified flow rate

from a reservoir of preblended solvent,

with the preparation constraints discussed

in part I of the series. Gradients are much

more complicated to duplicate between

systems. We assume that the pump is

delivering liquid flow at the programmed

rate, and that the percentage composi-

tion accurately corresponds to the pro-

grammed value. We also expect that any

gradient follows the intended profile and

that the specified composition reaches the

column at the intended time. All modern

chromatographic systems closely approach

these assumptions, but there are always

deviations from the ideal. The differences

in the deviations between different brands

or models of pumps create complications

for method transfer. The differences can

affect retention time and selectivity. It is

less well recognized that the differences

can also affect column regeneration and

reequilibration. Most scientists consider

the system volume to be the major source

of the deviations so method transfer strate-

gies are often based on equalizing the

system volume differences.

FIGURE 1: The measurement of the volume of a chromatographic system is based on running a gradient after removing the column. Mobile-phase A = water. Mobile-phase B = water with 10 mg/mL caffeine. Detection wavelength = 273 nm. The strong solvent is spiked with a UV absorbing marker (in this case, caffeine). Mobile-phase gradient: hold at 0% B for 5 min, then 0–100% B in 20 min, 100% B for 5 min, 100–0% B in 5 min; fl ow rate: 1 mL/min. (Reprinted with permission from reference 2).

100.00

80.00

Gra

die

nt

(%)

60.00

40.00

20.00

5.00 10.00

50%

100%

tG

tD

FVD

tG

t1/2 (2)

tG

t1/2

15.00 20.00

=

– –12

25.00 30.00 35.00 40.00

Time (min)

0.00

0.00

Silicon Wafer Technology Makes LC Analysis for Proteomics More powerful and Effective, A User’s ExperienceA Q&A

Geert Van Raemdonck Global Field Support Expert

PharmaFluidics

A s the field of proteomics grows more complex, traditional separation techniques like

nano-liquid chromatography (LC) have a harder time extracting critical information

efficiently. PharmaFluidics’ μPAC™, a breakthrough in column design, helps bring

LC analysis up to speed. LCGC recently sat down with Geert Van Raemdonck, global

field support expert at PharmaFluidics, to discuss this topic. After using the μPAC™ at the

University of Antwerp, Van Raemdonck was convinced about the benefits of micro-pillar

array column technology. His enthusiasm resulted in him joining the team of PharmaFluidics.

LCGC discussed with Van Raemdonck the unique properties of the μPAC™ columns

and got a user’s-eye view of what it’s like to work with them in proteomics applications.

Van Raemdonck also shared his ideas about how future products could meet even more

advanced analytical needs.

LCGC: What are the biggest differences between the μPAC™ columns and conven-tional nano-LC columns?Van Raemdonck: The biggest difference of the column is the way the backbone of the

stationary phase is manufactured. μPAC™ incorporates a chip made of a silicon wafer, and

freestanding pillars are etched out of the wafer, which results in a perfectly ordered structure.

This leads to a high separation performance because there is almost no peak dispersion.

Sharp peaks also lead to higher sensitivity, so that small amounts of molecules can be

detected more easily.

In addition, back-pressure is significantly lower as compared to conventional columns,

which allows you to operate your column at a broader flow range, going from about 300 nL

up to 1 μL per minute.

And last, since the μPAC™ column is etched out of a silicon wafer, there is no batch-to-

batch variation, as every column that is produced is exactly the same.

LCGC: For which applications are these μPAC™ columns best suited?Van Raemdonck: Special applications seen in the field of proteomics are analyses of samples

with low amounts of peptides—for instance, the analysis of biopsies or protein extracts from

a tissue. But, there’s also the opportunity for the analysis of complex samples that require

longer gradients, as for biomarker discovery. The technology is also suitable for chemically

labeled samples like iTRAQ or TMT, where the gradients applied for separation often last

four hours and longer.

Additional applications are in pharmaceutical fields, like in the detection of small subtle

differences in biopharmaceuticals, and biosimilars, like for antibody production and quality

AN EVALUATION OF USING μPAC™ COLUMNS FOR PROTEOMIC APPLICATIONS

control. In metabolomics, it might also be helpful to use

μPAC™ columns because of the low concentrations of mol-

ecules, and also in lipidomics experiments, where a high

resolution is required.

LCGC: Are there any drawbacks to these types of columns compared with classical columns?Van Raemdonck: In my opinion, there are no specific draw-

backs related to the μPAC™ columns. However, I think it’s

important to keep a few things in mind.

First of all, the back-pressure of

the μPAC™ should always stay below

350 bar, or 5,000 PSI, to prevent

any damage to the freestanding

pi l lars that form the separat ion

bed of the column. This maximum

back-pressure is lower than for clas-

sical nano-LC columns, but it also

lowers the shear force of the LC

instrumentation.

Secondly, it’s important to ground

the column in case of applying a high

voltage to the spray emitter of the

mass spectrometer. This prevents

any charg ing ef fects s ince the

column is made of a semi-conductive silicon chip.

Finally, the price of a μPAC™ column is higher than

most commercially available columns. However, since the

lifetime of a μPAC™ column outperforms any conventional

nano-LC column, the price per injection will be at least

equal or even lower.

LCGC: What product improvements would you like to see in the future from a user’s point of view?Van Raemdonck: I think an introduction of trapping columns

would be very useful since this would reduce the loading time

and also offer higher flexibility in the sample volumes that can

be injected into the column.

Further, it would be interesting to have the addition of

some products to our current portfolio—like, for instance,

those with bigger pore sizes in combination with other coat-

ings, like reverse-phase C8 or C4, which could be applied

in peptidomics and top-down applications.

And finally, if it would be possible to integrate the grounding

mechanism, that would also be very handy.

LCGC: Did you encounter any issues during the instal-lation and first use of the column?

Van Raemdonck: I used the μPACTM for the first time when I was

working at the University of Antwerp. The installation was done

perfectly by the team of PharmaFluidics. One thing that you

have to keep in mind is that you have to adapt your methods to

the internal volume of the 200-cm column. This is about 9 μL,

so it’s important that at the end of your gradient, you provide

enough equilibration time to equilibrate your column before you

inject your next sample. If you desire, you can also increase your

flow rate (due to the lower back-pressure) in order to reduce

this equilibration time.

LCGC: Why is grounding the column so important?Van Raemdonck: The column is

made out of a single silicon wafer

that’s semi-conductive. So, when the

column is not grounded and when a

high voltage is applied to the spray

emitter that is used to transfer your

molecules to your mass spectrometer,

the current can actually reach the

column and this will influence the

retention of the column.

So, this will result in some charging

effects that will retain your tryptic

peptides, which will result in significantly broader peaks. And

this discrepancy can be really big—you’ll see it immediately

when a column is not grounded; you’ll be having very broad

peaks. Unfortunately, adequate grounding is often a step

that’s forgotten.

LCGC: For which proteomics applications do you see the biggest potential for the μPACTM technology?Van Raemdonck: On the one hand, there are the small

sample sizes that I already mentioned—like the pro-

tein or peptide extractions from tissues and biopsies.

Furthermore, there are also the top-down proteomics

applications with the C4 or C8 coating. But, there are also

opportunities for protein–protein interactions and host-cell

protein identifications since there will be very tiny amounts

of compounds detected.

There is also a benefit to using the μPAC™ technology for

targeted applications like scheduled parallel reaction moni-

toring, since the elution profiles can be set really narrow.

So, you can include much more of the compounds that you

want to validate in a single run. And further, it could also be

very interesting to use the very stable retention times of the

μPAC™ columns for data-independent analysis.

“Since the μPAC™ column

is etched out of a silicon

wafer, there is no batch-

to-batch variation, as every

column that is produced is

exactly the same.”

828 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

System Volume

The system volume, also called dwell vol-

ume, void volume, or delay volume, is the

amount of liquid from the point where two,

or more, solvents meet and the point where

the blended solvents reach the head of the

column. Two systems with different dwell

volumes, delivering the same gradient, will

give different retention times. It has long

been recognized that characterizing the

volume of a system is the key first step for

maintaining constant retention time in a

method transfer. There are many published

and freely circulated procedures for mea-

suring system dwell volume (4,5). The gen-

eral approach is to prepare two batches of

the same solvent, one of which is spiked

with a UV-absorbing marker, such as uracil,

acetone, propyl paraben, or caffeine. The

column is replaced with a short length of

small inner diameter (i.d.) tubing. A gradi-

ent is run from 100% A to 100% B over 20

min, followed by a return to 100% A over 5

min. A hold, typically 5 min, is incorporated

at 100% B, and at the return to 100% A. The

gradient is observed as the UV absorbance

trace at the wavelength appropriate for the

selected marker. The volume of the system

is best measured at the midpoint of the gra-

dient. The difference in time between the

programmed midpoint and the observed

midpoint is multiplied by the flow rate to

obtain the system dwell volume. Although

it is sometimes suggested that the volume

be measured with an instantaneous step

from A to B or, alternatively, at the first lift-

off of the gradient trace, the midpoint is

much preferred as reflecting a steady state

transfer through the mixing volume of the

system, as discussed below. Although sys-

tem volume values typically are published

by instrument manufacturers, these val-

ues are seldom ideal for method transfer

experiments because the measurement

procedures are not consistent from one

manufacturer to another. It is, therefore,

necessary to measure the particular sys-

tems in use. For most purposes, the best

method is the one recommended by the

manufacturer of the system, but the overrid-

ing consideration is that the same method

be used to measure the volume of both

the original and the target system. A typical

useful procedure is shown in Figure 1 (5).

The measurement of dwell volume does

not ensure perfect replication of the gradi-

ent to be transferred. When the gradient

trace generated during the measurement

is examined in detail, it is always observed

that the offset time or volume is not con-

stant throughout the gradient profile.

This variation is a consequence of sev-

eral factors. Systems generally create the

programmed percentages by blending

volumes of each solvent. There is, how-

ever, some inaccuracy associated with the

nonadditive effects associated with pairs

of solvents. In addition, all instruments

make some adjustment for compressibil-

ity changes. The much larger source of

changes in the gradient profile is the mix-

ing that is incorporated in all systems. Gra-

dient systems must include a volume for

mixing of the proportioned solvents. Many

different designs have been used for mix-

ers, but they all must meet the two criteria

of stable, reproducible retention times and

ripple-free baselines. The ripples reflect

solvent concentration inhomogeneities

that may alter retention and will certainly

complicate quantitative integration of the

peaks. The alternative mixer designs all

are intended to smooth these ripples by

making the solvent changes uniform with

time. To achieve this, the mixer must have

a common volume that is large enough to

span the period of the ripple. This com-

mon volume, however, also distorts the

programmed profile of the gradient. When

a change in composition is initiated, that

change is intended to be uniformly dis-

tributed throughout the mixer volume. But

that change, now much smaller because

it is diluted by the mixer volume, begins

to emerge from the mixer immediately. In

other words, it reaches the head of the col-

umn in much less than the physical volume

of the system. Over the course of the gra-

dient, this phenomenon becomes less

significant as the series of small composi-

tion increments settles into a steady state

change. This is the basis for recommend-

ing that the system volume be measured

at the gradient midpoint. Measurement

at the start of the rise in the gradient

monitoring trace gives a much smaller

volume than the physical volume, or the

steady state condition.

Mixers

The differences among mixers can have

three effects on chromatography. First,

with a smaller system volume, the leading

edge of the gradient reaches the column

and causes peaks that elute very early in

the gradient to be shifted to an earlier

retention time. Second, because the entire

mixer volume must be flushed with the

final conditions of the gradient before that

strong solvent is delivered to the column,

the column may not be regenerated on

one system as compared to the other. This

can lead to a drift in retention times over a

series of injections. Third, on return to ini-

tial, the entire mixer volume must again be

flushed before re-equilibration can begin.

Again, the two systems may differ in the

equilibration to initial conditions for the

next injection. In this case, the two systems

may prove reproducible but different from

one another. These three problems have

different symptoms, and solutions, from a

simple mismatch in measurement of dwell

volume. Strategies for correcting system

volume differences will be discussed in

part III.

Gradient Mixing

There is one additional difference among

chromatographic systems that must be

considered. Pumps are usually classi-

fied as multi-pump gradient, high-pres-

sure mixing systems or as single-pump

gradient, low-pressure mixing systems.

The high-pressure mixing systems cre-

ate specific compositions by varying the

flow from two or more pumps to gen-

erate the desired solvent percentages.

The low-pressure mixing approach uses

a solenoid valve where each port opens

for a percentage of the valve cycle time

corresponding to the programmed com-

position. This series of solvent aliquots is

carried to the pump through a single tub-

ing piece. Some blending of the solvent

segments occurs in the transport tubing

and then in the pump heads as the mobile

phase is brought to the pressure and flow

for the method. Both system designs can

work very well in terms of compositional

accuracy and precision. The system vol-

ume is usually somewhat smaller with a

multi-pump gradient system, whereas the

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 829

single pump system can provide more

convenience as described in part I in the

context of solvent preparation.

There is an additional characteristic asso-

ciated with low-pressure mixing systems

that is not generally recognized. The fluid

in the transfer tubing from the proportion-

ing valve enters the pump head as a block

corresponding to the delivery volume of the

pump head. That series of solvent segments

is mixed, more or less completely, into a sin-

gle packet at the composition specified at

that time. That blended volume of solvent

is then delivered into the flow path towards

the column inlet. Rather than the smoothly

blended continuous gradient that we envi-

sion, the pump transmits a series of small

steps that are somewhat smoothed at the

edges of the packets. This flow and com-

position pattern is further complicated by a

mechanical characteristic of pumps. When

the piston, or plunger, expels the liquid

from the head, it does not dispense all the

liquid, because there is space between the

surface of the piston and the walls of the

pump head. This unswept liquid can be a

substantial fraction of the total volume of

the pump head, on the order of 40% of

its physical volume. That residual volume

mixes with the incoming mobile phase of

the next segment of solvent aliquots. The

result of this sequence of events is that the

mobile phase is diluted with the composi-

tion of the previous pump cycle, which was

also similarly diluted. This contributes

to the larger system volume associated

with single-pump systems, and, more

importantly, adds to the required time to

reach the regeneration step in a gradient

method and also to complete the return

to initial conditions for re-equilibrating for

the next injection.

Tools for Instrument

Calibration in Method Transfer

The physical factors that directly affect

the shape of the gradient in a given

system have elicited interest in alterna-

tives to the common and simple ways

to estimate and use the system volume.

One interesting alternative is the use of

marker compounds to calibrate the sys-

tem. A more general tool, called “Mea-

sure Your Gradient,” has been developed

to evaluate the shape of the gradient

on various systems (6). This tool can be

implemented from an open-source web

tool: http://www.measureyourgradient.

org/index.php. For this approach, the

intent is to recreate the identical gradient

on the two systems in question. The gra-

dient table entries will be different, but

the solvent composition delivered to the

column will be identical. The “Measure

Your Gradient” tool uses a very exactly

defined mixture of test compounds along

with a specified column, mobile phase,

and method. This test is run on both sys-

tems and the retention times are entered

into the software. The gradient is calcu-

lated to give a specific array of retention

times. Because this approach provides a

multi-point calibration that addresses the

several physical factors discussed above

that make gradient transfer imperfect,

it provides a very good adjustment for

method transfer. It is, however, limited

if the information is not available about

both systems in the method transfer.

This problem arises when implementing

methods that are even a few years old

where the systems may no longer exist.

Even in the case where current instru-

ment systems were used to develop the

method, those instruments may not be

available to the laboratory responsible

for the transfer. This same obstacle can

interfere with the conventional measure-

ment strategies discussed above. When

the observed system volume is to be

used to match the gradients delivered

by two systems, the same measurement

protocol must be used on both systems.

That may simply not be possible.

An alternative to measuring the exact

system volume can be suggested for

transfer of the separation method. The

originator method (the method being

duplicated), should provide system suit-

ability criteria specifying the retention

time for each sample component that

is to be measured. Use the established

method on the new system. This initial

experiment may not meet the specifi-

cations, but the major analyte peak will

be recognizable. Compare the retention

time of this peak on the new system with

the time in the specifications. The differ-

ence in retention time multiplied by the

flow rate gives the difference in system

volume between the two systems. This

trial can then be repeated after apply-

ing the strategies discussed in part III

to match the volumes between the two

systems. After a few iterations of this

empirical adjustment, the chromatogram

should be close to meeting specification.

If there are still discrepancies, apply a

correction to the target system that mini-

mizes all the differences in time.

Conclusions

We have considered the contribution

of system characteristics to transferring

chromatographic methods. The possible

differences between the originating and

the target systems can alter resolution

by changing the characteristics of sol-

vent delivery and by altering dispersion

of the separated peaks as they migrate

through the system. Consideration must

also be given to the resistance to flow

within the system tubing and the blend-

ing of solvents. In our final part of this

three-part series, we will consider the

modules that comprise each system and

the ways that they can be aligned for

consistent results.

References

(1) T.E. Wheat, LCGC North Am. 36(9), 693–696 (2018).

(2) General Chapter <621> “Chromatogra-phy” in United States Pharmacopeia 40 National Formulary 35 (USP 40-NF 35, United States Pharmacopeial Convention, Rockville, Maryland, 2017), pp. 508-520.

(3) F. Gritti, T. McDonald, and M. Gilar, J. Chrom. A 1420, 54-65 (2015).

(4) J. W. Dolan, LCGC Europe, 19(6), 336-343 (2006).

(5) P. Hong and P.R. McConville; Waters Corporation White Paper; 720005723EN; ©2016 Waters Corporation (2016).

(6) M.H. Magee, J.C. Manulik, B.B. Barnes, D. Abate-Pella, J.T. Hewitt, P.G. Boswell; J. Chrom. A 1369, 73-82 (2014).

Thomas E. Wheatis a principal scientist with Chromato-graphic Consulting, LLC in Hopedale, Massachusetts. Direct correspon-dence to: [email protected].

830 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

Plates are generated during the elu-

tion of solutes through a chromato-

graphic column and contain a wealth

of information about the separation

process, mainly peak dispersion. It is an

easily measured quantity used to probe

column properties.

We had previously shown in part IV of

this series (1) that the plate concept orig-

inated from fractional distillation and was

later used to interpret results obtained

from countercurrent distribution (CCD)

extractions. At about the same time that

CCD was being developed, A. J. P. Mar-

tin (1910-2002) and R. L. M. Synge (1914-

1996) formulated a theory of chromatog-

raphy using the theoretical plate concept

(2,3). These researchers went on to win the

Nobel Prize in Chemistry in 1952 for the

invention of partition chromatography.

In this article, we will focus on the

development of theoretical plates and

peak broadening, and arrive at relation-

ships that are applicable to liquid chro-

matographic (LC) separations. The more

important relationships from distillation

and CCD will once again be reviewed for

the benefit of the reader.

Background Information

Fractional Distillation

The concept of theoretical plates

was first introduced with respect to

fractional distillation, which required

an accounting of the separat ion

ef ficiency of dif ferent lengths and

designed distillation columns (1). It was

assumed that the distillation process

occurs in stages along the length of a

distillation column, the location of which

varies during the distillation process.

At each stage, equilibrium is reached

between the vapor and liquid phases,

and separation occurs between high-

and low-vapor-pressure components.

The number of theoretical plates is

proportional to column length, and

depends upon column design. It

should be emphasized that distillation

is not a chromatographic process, but

a countercurrent process of rising vapor

in contact with descending distillate

droplets.

To compare distillation efficiencies

among columns of different lengths, col-

umn length, L, is divided by plates, N, to

give the height equivalent of a theoretical

plate, HETP, or simply plate height H:

H = L/N [1]

Although distillation is a complex pro-

cess, each plate can be viewed as a vir-

tual platform on which vapor-liquid equi-

librium occurs.

CCD Extractions

A cleverly designed automated CCD

extraction apparatus, comprised of

multiple two-compartment transfer

tubes, was invented by Craig and Post

(2). Craig tubes consist of upper and

lower compartments, filled respectively

with two immiscible solvents (1,2). The

first tube contains sample dissolved in

either the upper or lower immiscible

liquid phase. After each extraction,

the upper phases are simultaneously

transferred to adjacent tubes, contents

mixed, and the process repeated until all

transfers are completed. The amount of

solute that is equilibrated between the

two phases depends on the distribution

coefficient of the solute, K, and relative

volumes of the two phases (1,3).

Distribution profiles of solutes are

constructed manually by plotting solute

concentration, c, against the number of

transfers, r, or tube location. If we assume

a Gaussian distribution (see Figure 1), the

number of theoretical plates of a CCD

run can be approximated using

N ∝ rK [2]

with a standard deviation defined by

σ = √rpq [3]

where p and q are the respective sol-

ute fractions in the two immiscible

phases at equilibrium. (By convention,

p is the solute fraction in the upper

compartment or mobile phase and q

is the solute fraction in the lower com-

partment or stationary phase). We can

now see that the chromatographic

theory, including peak broadening, is

beginning to take shape.

Martin and Synge’s

Contributions to LC Theories

Martin and Synge sought to improve

upon CCD extractions with a paradigm

shift by inventing partition chromatog-

raphy, a technique that uses a liquid

stationary phase. Their experiments con-

sisted of using LC columns packed with

an adsorbent coated with an immiscible

liquid phase. (In addition, paper chroma-

Howard G. Barth

This month’s “Chromatography Fundamentals” is a continuation of the development of the theoretical plate concept,

with emphasis on its significance, properties, and uses as applied to liquid chromatography.

Chromatography Fundamentals, Part V:Theoretical Plates: Significance, Properties, and Uses

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 831

tography was also introduced by these

Nobel laureates.)

They made the following assumptions,

which are universally valid for chromato-

graphic techniques (4,5):

1. Separation is uniform throughout a

chromatographic column.

2. A column can be divided into equal

lengths, stages, or segments.

3. Within each stage, there is sufficient

time for equilibrium to be reached,

as solutes partition between mobile

and stationary phases.

4. Solutes are sufficiently dilute so that

their retention characteristics, i.e.,

thermodynamic properties, are inde-

pendent of one another.

5. When applied to LC (or GC), each

stage approximates one theoretical

plate.

6. The number of theoretical plates

generated by a solute can be calcu-

lated by representing each peak as a

Gaussian distribution.

7. Each theoretical plate is considered to

be a discrete site (a nano-size separa-

tory funnel, if you wish), in which sol-

utes distribute between two phases.

8. After equilibrium, solute is carried

by the mobile phase to the next

theoretical plate and the process

repeated until components emerge

from the column with characteristic

retention times and peak widths, as

described by a Gaussian distribution.

Martin and Synge published two experi-

mental methods for determining the num-

ber of theoretical plates of an LC peak (4).

Method 1

A Gaussian distribution was used to

describe solute concentration as a func-

tion of retention time t, where tr is peak

maximum, σ is the standard deviation,

and x = t − tr:

2π)e c = (1/√ −1/2 (x2/σ2) [4]

Based on this equation, the following

relationship was derived:

N = 2π (h2l2/A2) [5]

Inflection Point

0.5 h

0.607 h

h

Injection

W = 4

W1/2

2

tr , V

r , or d

FIGURE 1: Gaussian chromatographic peak indicating different methods of measuring theoretical plates. Refer to Table I and equations 6, 12 to 15.

Learn more about SiliaFast™ FaPEx®: ■ Visit: www.silicycle.com/fapex

■ Contact us: [email protected]

C E R T I F I É E I S O 9 0 01 : 2 015 C E R T I F I E D

www.silicycle.com

Unique on the market

Up to 120 X faster than existing methods

Simple

Wide spectrum

Cost effective

SPEED UP YOUR PESTICIDE RESIDUE ANALYSIS

WITH SILIAFAST ™ FaPEx® CARTRIDGES

T

ECHNO

L

OGY

PA

TENTED

832 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

Here l is the distance from the point of

injection to the peak maximum, h is peak

height, and A is peak area.

Equation 5 can now be converted to

the iconic plate equation, where l is set

equal to retention time, tr, and area and

height are transformed into peak width in

units of time (4–11):

N = 16 (t

r )2

(wt)2

[6]

Provided that the peak is a Gaussian

distribution, equations 5 and 6 are equiv-

alent.

Method 2

The second approach used was to

compare LC to CCD extractions. Since

r is equal to one transfer in a CCD

analysis, or by definition, r = n, equation

3 can be written as

rpq ∝ nσ = √ √ [7a]

We know that n is a single plate, i.e.,

n = N, thus equation 7a, becomes

rpq ∝ σ = √ n ∝ N √ √ [7b]

As previously shown (1), after r trans-

fers, the CCD tube that contains the max-

imum concentration of solute, μ, is given

by

μ = rp [8]

Since μ ∝ tr and from equation 2 we know

that r ∝ N, equation 8 can be recast as

tr ∝ N [9]

Taking the ratio of equations 9 and 7b,

we obtain

tr N

N=σ

[10]

Squaring numerator and denominator,

N2

NN= =

(tr )2

σ2

[11]

Letting time be the unit of measurement,

wt=4σt, we obtain the expected result,

N =(t

r)2

(wt/4)2

= 16(t

r)2

(wt)2

[12]

which is identical to equation 6. This clas-

sical equation not only allowed separa-

tion efficiency to be compared among

columns, but also was key for optimiz-

ing LC experimental parameters, as

described below.

Theoretical Plate Representations

Retention and peak width in equation

6 can be adjusted to accommodate the

types of measurements being made.

Thus these values can be expressed in

terms of volume, by multiplying each

term by flow rate, F:

N =16(V

r)2

(wv)2

[13]

For researchers who still rely on strip-

chart recorders, or for estimating plates

from TLC or open columns (3), the follow-

ing equation can be used:

N = 16(d

r)2

(wd)2

[14]

where dr is either the peak maximum

distance on the strip chart or the migra-

tion distance of a solute band or TLC

spot, and wd is the baseline width.

Equations 5, 6, 13, and 14 are similar,

if not identical; however, plate calcula-

tions from planar chromatography and

open-column LC are estimates, since

solute zones are usually not Gaussian.

(Before online computers, peak widths

were calculated manually by drawing

tangents to the two slopes of a peak, as

seen in Figure 1.)

Occasionally, baseline widths may be

problematic to measure reliably because

of noise, drift, peak asymmetry, or the

presence of partially resolved peaks. A

common procedure to avoid these diffi-

culties is to measure peak widths higher

up the peak, as illustrated in Figure 1.

Thus, at one-half peak height, equation

15 is used:

TABLE I: Summary of different representations of calculating theoretical plates from LCdata. Refer to Figure 1.

Eqn Relationship Applicability

6 N = 16N(t

r)

rr2

(wt)2

Commonly used plate equation with units of time.

12 N = 16N(V

r)

rr2

(wv)2

Use when retention volume is required, e.g., SEC.

14 N = 16N(d

r)

rr2

(wd

w )2Use with planar or open-column chromatography.

15 N = 5.54N(t

r)

rr2

(w1/2

)2

Use for asymmetric, skewed, or partiallyresolved peaks; w is measured at 0.5 x ht.w

16 N =N 4(t

r)2

(w2

wwσ)2

Alternative to eqn 14: w is measured between wpeak infl ection points at 0.607 x peak height.

tr, retention time.rr

wt

w = 4σt, peak width in units of time.

Vr,

V retention volume.

wv

= 4σv, peak width in units of volume.

dr, distance of peak maximum.rr

wd

w = 4σd, peak width in units of length.

ww = 2.35σt, peak width at one-half peak height in time units.

w2

wwσ

= 2σt, peak width at 2σ in time units.

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 833

N = 5.54(t

r)2

(w1/2

)2

[15]

We can move still higher up the peak

and measure width between inflection

points, i.e., 2σ, as depicted in Figure 1,

and equation 16:

N = 4(t

r)2

(w2σ

)2

[16]

Since equations 6, 15, and 16 may

give different results depending on peak

shape and overlapping peaks, measure-

ments should be consistent for a given

analysis. These equations are summa-

rized in Table I.

Theoretical Plate Properties

Ta b l e I I l i s t s LC ex p e r i m e n t a l

parameters and their qualitative effects

on the number of theoretical plates,

as shown in the last column. In spite

of the complexity of some of these

relationships, correlations have been

established most notably with injection

volume, f low rate, column length,

particle size, and dead volume. Except

for column length, which is given in the

next section, these relationships will be

covered in subsequent tutorials.

Column Length

Column length, L, is an integral part of

plate theory, since tr ∝ L. Furthermore,

the number of transfers, r, with respect

to CCD, is analogous to column length.

Therefore, from equation 3, σ = √rpq, we

obtain σ ∝ √L and equation 11 becomes:

N = L2/σ2 ∝ L2/L∝ L [17]

Equation 17 indicates that sufficiently

long columns can be used to generate

enough plates to pull apart neighboring

peaks. However, there are three critical

limitations: back pressure, analysis time,

and detectability. Column pressure and

analysis time are both directly propor-

tional to length, and an acceptable col-

umn length can be readily predicted from

TABLE II: Factors infl uencing theoretical plates, N=tr2/σ2 (eqn 11).

Item LC VariablesRelative Influenceon Plate Numbers

Terms Most Affected

1. Mobile phase composition medium tr

2.Stationary phasecomposition

medium tr

3. Gradient profi le strong tr,rr σ2

4. Column temperature strong tr,rr σ2

5. Injection volume medium/strong σ2

6. Injection amount weak σ2

7. Flow rate weak/medium tr, σ2

8. Column i.d. weak σ2

9. Column length strong tr,rr σ2

10. Column confi guration weak/medium σ2

11. Particle size very strong σ2

12. Particle shape medium σ2

13. Pore size weak tr

14.Particle size/ shapeuniformity

medium σ2

15. Packing purity weak/medium tr

16.

Dead volume of inter-connecting tubing: injector-columncolumn-columncolumn-detector

very strong σ2

A breakthrough in

sample automation

and concentration

for GC–MS

NEW

Centri ®

Multi-technique sample

pre-concentration and injection

platform, delivering enhanced

analytical sensitivity and higher

throughput.

ɵ SPME & SPME–trap

ɵ Headspace & HS–trap

ɵ HiSorb sorptive extraction

ɵ Thermal desorption

Find out more chem.markes.com/Centri

834 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

a single measurement using any conve-

nient column length.

As column length increases, peaks

broaden with a corresponding decreased

detector response, defined by equation

4. Decreased detector response, however,

will also affect signal-to-noise ratio, limiting

detection of trace or minor components.

Plate Height

An essential relationship derived

from theoretical plates is plate height or

height equivalent of a theoretical plate,

HETP (see equation 1). Martin and Synge

referred to it as plate “thickness”, in

which the “thicker” or wider the plate, the

lower the separation efficiency. They pro-

posed that plate thickness corresponded

to the depth of the stationary phase and

reasoned correctly that column efficiency

could be improved by decreasing this

layer or increasing the rate of transfer of

solute molecules during partitioning.

Equation 1 can also be expressed dif-

ferently by letting N = L2/σ2:

H = L/N = Lσ2/L2 = σ2/L [18]

Plate height now becomes peak vari-

ance per unit column length, a rela-

tionship used for studying column and

packing properties that govern peak

broadening, as discussed in part VI.

Chromatographic Resolution

Another critical relationship is the reso-

lution, Rs, between adjacent peaks,

Rs = (x2-x1)/4σ = 6x/4σ [19}

where ∆x is the distance between two

peak maxima, and σ is the average stan-

dard deviation of two adjacent peaks,

which we assume are Gaussian. A value

of ≥ 1.5 is required for baseline resolution

of two peaks of equal areas.

Resolution can be related to column

length by considering the behavior of

peaks during elution. As two adjacent

peaks travel through a column, peak max-

ima move apart, according to

Δx ∝ L [20]

Peak dispersion or width, however,

also increases, but not as fast. The rela-

tionship between peak width and column

length becomes apparent by rearranging

equation 18:

σ = (LH)½ [21]

Since H is constant for a given column,

σ ∝ L½ [22]

the resolution equation now can be

expressed as

Rs=∆x/4σ ∝ L/L½ ∝ L½ [23]

Based on this relationship, resolution

will increase by only 40%, each time col-

umn length is doubled. Furthermore, if

we wanted to double resolution, column

length would have to be increased by a

factor of four. The resolution equation will

be examined in more detail in a subse-

quent article of this series.

Theoretical Plates: Range

and Limitations

It is instructive to estimate the minimum

plate count of low-resolution chromato-

graphic methods, like paper, thin-layer, or

open-column chromatography. We can

assume a worst-case scenario in which

a solute tails from the point of applica-

tion to its final resting place. If we let x

be the distance from sample application

to the middle of a sample spot (splotch)

or tailed band, then σ ≈ ½x. The approxi-

mate plate count is N = x2/σ2 ≈ 4. Indeed,

the typical plate count of open columns

seldom exceed 102. Even with these

extraordinarily low plate counts, by mod-

ern standards, these methods were highly

praised for their separation abilities.

Modern, high efficiency columns,

by comparison, can typically range

TABLE III: Application of theoretical plates for selecting and maintaining high columnperformance.

Application Procedure Comments

Column selection

Choose column withrequired N.

Procedure for calculating required N is Ndescribed by resolution equation.b

Column installation

Compare N with listed value.NN should be within range of list-Ned value, if not, reinstall column.

Column comparisons

Compare N vs. mL/min plots Namong potential columns.a

Select column with shallowest slope.

Column stability

Plot N vs. wks of usageNChange column when plate loss reaches

10-20% or if Rs becomes unacceptably low.

a. A more meaningful plot is H vs. linear velocity, see Chromatography Fundamentals, Part VIII.H

b. Chromatography Fundamentals, Part VIII of this series.

TABLE IV: Using theoretical plates to optimize experimental LC conditions.

Experimental Variables

Data Analysis Optimum Value

InjectionVolume

Plot N vs. μN L: plates will be-gin to decrease with increas-

ing injection volume.

Select injection volume within region giving maximum plates,

maintaining adequate s/n.

Sample Con-centration

Plot N vs. N mg/mL: plates will begin to decrease with increas-

ing injection concentration.

Select concentration within region giving maximum plates,

maintaining adequate s/n.

Flow RatePlot N vs. N mL/min: plates will slowly decrease with increasing fl ow rate.

Select fl ow rate to achieve maximum N maintaining Nadequate analysis time.

Column Temperature

Plot N vs. °C: plates will in-Ncrease with temperature.

Select temperature at max allowable value without sac-rifi cing safety or resolution.

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 835

between 104 to less than 105 plates per

column, depending, of course, on col-

umn length. In theory, there is no upper

plate limit, since we can always con-

struct longer columns.

Suppliers sometimes advertise col-

umns based of plates per meter, which

can be misleading since actual column

lengths are significantly shorter. In order

to compare columns, it is best to choose

columns based on actual plates per col-

umn length; for column comparisons,

plate height is preferred, which is inde-

pendent of column length.

Recall that LC plate theory was devel-

oped by assuming Gaussian peak distri-

butions: for non-Gaussian peaks, such as

skewed or tailed shapes, the plate con-

cept is no longer applicable, and plate

counts are usually underestimated, unless

peak width is measured at 50% or 60.7%

of peak height (equations 15 and 16).

Furthermore, plate theory is also based

on the assumption that chromatographic

conditions remain constant throughout

the analysis and that equilibrium has

been reached.

When gradient elution or temperature

programming is applied, equilibrium

may not be attained. In addition, reten-

tion times are dictated by the gradient

profile. As a result, strongly retained

peaks can remain stationary during

part of the gradient and elute at con-

siderably long times with compressed

peak widths, producing artificially high

plate counts. Under these conditions,

the physical significance of theoretical

plates is lost.

Theoretical Plates: Applications

Theoretical plate measurements of

unretained solutes are typically used

in the development and maintenance

of LC methods, as described below

and summarized in Tables III and

IV. Unretained solutes ref lec t the

un i fo r mi t y o f the pac ked bed,

packing particle size, mobile phase

hydrodynamic s , so lu te d i f fus ion

coefficient in the mobile phase, and

extracolumn effects. In addition to

these effects, retained solutes also

probe stationary phase characteristics,

such as its thickness and homogeneity,

and the diffusion coefficients of solutes

within the stationary phase. In this

section, only unretained solutes are

considered for column evaluation, since

they will produce higher plate counts

than retained components, thus are

more sensitive probes.

Most LC peaks have profiles that devi-

ate from Gaussian distributions and are

not strictly symmetrical; consequently,

there will be errors associated with base-

line width measurements. In view of this

imprecision, only two significant figures

are justified for reporting plate counts.

A summary of theoretical plate

applications is outlined below and in

Tables III and IV.

Getting Started with Plates

Based on preliminary analytical runs,

the required number of plates to

effect a separation are estimated; this

procedure will be described in part VII.

With this value, an appropriate column

of required plates can be selected.

After a new column is installed, the

plate number is checked against the sup-

plier’s value to ensure that the column

was correctly plumbed into the LC sys-

tem.

Experimental parameters, such as

injection volume, solute concentration,

flow rate, and column temperature, are

then optimized for maximum plate count.

Column performance typically will dete-

riorate with time, as a result of buildup of

particulates or contaminants, disruption

of packing uniformity, dissolution of silica

packing, loss of bonded phase, or surface

oxidation. Column fidelity is ascertained

by monitoring separation attributes, such

as plate count, solute retention time, and

resolution on a routine basis.

Conclusions

The number of theoretical plates, which

forms the basis of chromatographic

theory, is a key parameter used in

all modes of chromatography for

measuring column ef ficiency. It is

simply the ratio of retention squared

to peak variance, a relationship first

realized by Martin and Synge.

When column length is normalized

with respect to plate number, we obtain

a new parameter, the plate height, which

is a measure of the amount of peak

broadening that occurs when solute trav-

els through a defined length of column.

Solute emerges as a Gaussian distribu-

tion carrying information regarding sol-

ute and column characteristics, as well as

experimental conditions.

Plate number and height are used

to develop and optimize chromato-

graphic methods, details of which will

be presented in subsequent articles

of this series on the fundamentals of

chromatography.

Our next tutorial will review the prop-

erties of the Gaussian distribution with

emphasis on peak variance and its effect

on chromatographic resolution.

References (1) H.G. Barth, LCGC North Am. 36(8),

532–535 (2018).(2) E.W. Berg, Physical and Chemical

Methods of Separation (McGraw-Hill, NY, 1963).

(3) H.G. Barth, LCGC North Am. 36(3), 200-203 (2018).

(4) R J. Magee, Selected Readings in Chromatography (Pergamon Press, Oxford, UK, 1970).

(5) A.J.P. Martin and R.L.M. Synge, Biochem. J., 35, 1359 (1941).

(6) R.L. Grob, Chpt. 2, In: R.L. Grob, Ed., Modern Practice of Gas Chromatography, 2nd ed. (Wiley-Interscience, New York, 1985), pp. 49–114.

(7) S.G. Weber and P.W. Carr, Chpt. 1, In: P.R. Brown and R.A. Hartwick, Eds., High Performance Liquid Chromatography (Wiley-Interscience, New York, 1989), pp. 1–115.

(8) J.C. Giddings, Unified Separation Theory (Wiley-Interscience, New York, 1991).

(9) B.L. Karger, L.R. Snyder, and C. Horvath, An Introduction to Separation Science (Wiley-Interscience, New York, 1973).

(10) P.A. Bristow, Liquid Chromatography in Practice (Handforth, Cheshire, UK, 1976).

(11) L.R. Snyder, J.J. Kirkland, and J.W. Dolan, Introduction to Modern Liquid Chromatography (Wiley, New York, 3rd ed., 2010).

Howard G. Barthis with Analytical Chemistry Consul-tants, Ltd. in Wilmington, Delaware. Direct correspondence to: [email protected]

836 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

PRODUCTS & RESOURCES Reference materialsLGC Standards’ reference materials

are designed for food, beverage, and

environmental analysis. According

to the company, the materials

include those for cannabis, allergens,

veterinary drugs, pesticides, dyes,

toxins, and other organic materials.

LGC Standards,

Manchester, NH. www.lgcstandards.com

Multitechnique sample preparation systemMarkes’ Centri multitechnique system

is designed for sample automation and

concentration for gas chromatography–

mass spectrometry. According to the

company, four sampling modes are

available: HiSorb high capacity sorptive

extraction, headspace, solidphase

microextraction, and thermal desorption.

Markes International,

Llantrisant, UK.chem.markes.com/Centri

Custom HPLC columnsCustom HPLC col-

umns from Hamilton

can be packed with

any of the company’s

stationary phases in

various hardware for-

mats. According to the

company, the stationary

phases are available in

various particle sizes.

Hamilton Company,

Reno, NV. www.hamiltoncompany.com

ColumnsTosoh’s TSKgel PWXL-CP

columns are designed for the

analysis of water-soluble cationic

polymers by size-exclusion chro-

matography. According to the

company, the column packing

material incorporates a cationic

functionality on the surface of

the particles to prevent ionic

adsorption of the polymers.

Tosoh Bioscience, LLC,

King of Prussia, PA.www.tosohbioscience.com

GC mass spectrometersShimadzu’s GCMS NX series of

gas chromatograph (GC)–mass

spectrometers is designed

using the company’s latest Nexis

GC-2030 as the GC unit, which is

equipped with a next generation

flow controller that enables con-

trol and accommodates a variety

of applications. According to the

company, the systems include fea-

tures to facilitate maintenance and

to improve operating efficiency by reducing standby time. Shimadzu

Scientific Instruments, Columbia, MD. www.ssi.shimadzu.com

LC accessoriesAccessories from Restek are

designed for use in liquid chro-

matography analysis. According

to the company, products include

bottle top inlet valves, outlet

valves, couplers, fittings, unions,

tees and crosses, PEEK stainless

steel tubing, mobile-phase main-

tenance and safety products, bot-

tle tops, valves, filters, spargers,

replacement parts, and supplies.

Restek Corporation,

Bellefonte, PA. www.restek.com

Storage rackMacherey-Nagel’s

chemically resistant

foam storage rack

is designed for the

proper organization and

protection of crimping

tools. According to

the company, the

storage rack is part of

its range of vials, caps,

and chromatography

accessories.

Macherey-Nagel Inc., Bethlehem, PA. www.mn-net.com

Thin film solid-phase microextractionGerstel’s thin-film solid-phase microex-

traction device is a 20 x 4 mm carbon

mesh sheet impregnated with sorptive

phase, designed to be placed inside

a sample vial for analyte extraction

from the headspace or liquid phase.

According to the company, the large

surface area, wide polarity range, and

phase volume result in faster extraction

and improved analyte recovery.

Gerstel,

Linthicum, MD.

www.gerstelus.com

WWW.CHROMATOGRAPHYONLINE.COM NOVEMBER 2018 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 837

Ad IndexADVERTISER PAGE ADVERTISER PAGE

Separation systemThe Eclipse DualTech separation

system from Wyatt Technology

is designed for both hollow-fi-

ber flow field-flow fractionation

(HF5) and asymmetric-flow

field-flow fractionation (AF4)

techniques. According to the

company, both techniques may

be integrated into one instru-

ment and coupled to the company’s DAWN HELEOS II detector.

Wyatt Technology Corp., Santa Barbara, CA. www.wyatt.com

SyringesVICI Precision Sampling Pressure-

Lok analytical syringes are made

with polytetrafluoroethylene

(PTFE) plunger tips. According

to the company, the tips are

designed to remain smooth,

without the seizing or residue

of conventional metal plunges,

and have leak-proof seals.

Valco Instruments Co., Inc.,

Houston, TX.www.vici.com

GC columnThe Quadrex 007-65HT GC

column is designed for the

analysis of highmolecular-weight

triglycerides in edible oils via high-

temperature gas chromatography.

According to the company, the

polar high-temperature column

can withstand temperatures up to

365 °C while achieving separation

of the individual triglycerides.

Quadrex Corporation,

Woodbridge, CT.www.quadrexcorp.com

Fixed-ratio flow splittersMott’s PerfecPeak fixed flow

splitters are designed to pro-

vide improved peak resolution

and accurate splitting with a

fingertight design. According to

the company, the design allows

for low internal volume, and the

splitters are equipped with

interchangeable splits and a

0.1-μm replaceable prefilter.

Mott Corporation,

Farmington, CT.www.mottcorp.com

Cornerstone Scientific. . . . . . . . . . . . . . . . . . . 807a, b

Gerstel GmbH & Co. KG. . . . . . . . . . . . . . . . . . . 793

Hamilton Company . . . . . . . . . . . . . . . . . . . . . 797

LGC Standards . . . . . . . . . . . . . . . . . . . . . . . . 821

Macherey-Nagel GmbH & Co. KG . . . . . . . . . . . . . 789

Markes International Ltd. . . . . . . . . . . . . . . . . . . 833

Merck Millipore. . . . . . . . . . . . . . . . . . . . . . . . 813

MilliporeSigma . . . . . . . . . . . . . . . . . . CV2, 803, 812

Mott Corporation . . . . . . . . . . . . . . . . . . . . . . 799

Optimize Technologies . . . . . . . . . . . . . . . . . . . 817

Parker Balston, Inc. . . . . . . . . . . . . . . . . . . . . . . 815

PharmaFluidics . . . . . . . . . . . . . . . . . . . . . 826–827

Pickering Laboratories . . . . . . . . . . . . . . . . . . . . 801

PITTCON. . . . . . . . . . . . . . . . . . . . . . . . . . . . 823

Restek Corporation. . . . . . . . . . . . . . . . . . . . . . 792

Shimadzu Scientific Instruments. . . . . . . . . . . . . . . CV4

Showa Denko America, Inc.. . . . . . . . . . . . . . . . . . 787

Silicycle, Inc. . . . . . . . . . . . . . . . . . . . . . . . . . 831

Sonntek, Inc.. . . . . . . . . . . . . . . . . . . . . . . . . . 809

Tosoh Bioscience. . . . . . . . . . . . . . . . . . . . . . . 819

VICI Harbor Group. . . . . . . . . . . . . . . . . . . . . . 791

Wyatt Technology Corporation . . . . . . . . . . . . . . . 795

THE ESSENTIALSE xcerpts f rom LCGC’s p ro fess iona l

deve lopment p la t form, CHROMacademy.com

HPLC Column Maintenance:

Tips for Extending HPLC Column Lifetime

While the relative cost of HPLC

columns has been reduced

over time, extending column lifetime

remains an important consideration

for most laboratories. The following

tips should help to protect your col-

umns and significantly extend the use-

ful lifetime of most phases.

Always read the manufacturer’s litera-

ture with respect to the recommended

pressure, eluent pH and temperature

operating ranges for the column, and

stick to these ranges.

Note that higher operating temperatures

often go hand in hand with reduced pH

operating ranges. At low pH, the main

symptom of column degradation is typ-

ically loss of efficiency (peak broaden-

ing) and at high pH, peak tailing and an

increase in column back pressure.

Avoid mechanical shock of the column

bed, such as dropping the column, and

ramp the pressure or flow slowly (1 mL/

min/min is ideal) each time eluent flow

is initiated.

Most modern HPLC equipment is capa-

ble of achieving this flow or pressure

ramp automatically through secondary

instrument settings. Bed voiding due

to pressure shock often manifests itself

via split or very badly tailing peaks. Col-

umns may be reversed for analysis if a

replacement is not readily available;

however, the efficiency of the column

is likely to reduce much more quickly

as the bed will ultimately slump in the

opposite direction, leading to the same

chromatographic symptoms.

If columns have dried out, initiate

the flow very slowly (0.1 mL/min/min)

using an eluent containing at least

50% acetonitrile.

If a “standard” (non “aq” or non-polar

embedded phase type) reversed-phase col-

umn is suspected of phase collapse (short-

ening retention times, poor efficiency) due

to use with 100% aqueous mobile phases,

the column should be reactivated at high

flow with 100% acetonitrile at 60 oC (take

care to not precipitate any solid buffers from

the eluent remaining within the column). In

both of these cases, between 50 and 100

column volumes may be required to prop-

erly re-equilibrate or re-activate the phase.

Columns should be properly washed

after each use.

A recommended washing routine may be:

• Current content to 90% acetonitrile at

10% organic per 2 column volumes and

hold for 10 column volumes (again take

care to avoid precipitation of solid buffers

by ramping the acetonitrile concentra-

tion slowly, as recommended here).

• 10% acetonitrile per 2 column volumes to

50:50 acetonitrile:water and hold for 10

column volumes.

• Remove from the system, end cap and

store. End capping of the column is very

important to avoid the phase drying out.

One might use an older HPLC system as

a column “wash station,” which can save

significant amounts of operating time on

“live” instruments.

If samples are likely to contain partic-

ulate matter, choose a good quality

inline filter with the appropriate mesh

size; 0.45 μm for traditional columns and

0.2 μm for UHPLC columns is typical.

If the sample matrix or diluent is likely to

harm the sorbent (due to pH, for exam-

ple) or is particularly chemically dirty or

intractable, a guard column may be used,

and the phase should be matched with

that of the analytical column. Take great

care when selecting the dimensions of the

guard column and connecting to the ana-

lytical column to ensure that the efficiency

of the separation is not compromised.

If column or frit contamination is sus-

pected due to peak splitting or loss of

efficiency, it is possible to reverse the

direction of the column for back flush-

ing purposes, and the column washing

procedure mentioned above is a good

“recipe” for this purpose.

One should uncouple the column from

the detector to avoid fouling, and note

that reversal for flushing should only be

used as a matter of last resort, and that

the original efficiency of the column may

not be achieved.

Remember that column volume may

be estimated using π x r2 x L x 0.6

(the approximate interstitial poros-

ity of silica used for HPLC column

packing materials).

So, for a 150 x 4.6 mm column, this would

approximate to:

3.142 x (2.3)2 x 150 x 0.6 = 1.496 μL or ~1.5

mL

These tips will help extend your column life-

times.

838 LCGC NORTH AMERICA VOLUME 36 NUMBER 11 NOVEMBER 2018 WWW.CHROMATOGRAPHYONLINE.COM

FIND THIS, AND OTHER WEBCASTS, AT

www.CHROMacademy.com/Essentials

(free until December 20).

MORE ONLINE:

Agilent Technologies is offering five years complimentary access to

CHROMacademy for all university students and staff.

CHROMacademy is an intuitive, comprehensive e-learning and trouble-

shooting platform with more than 3,000 pages of content for HPLC,

GC, sample preparation, and hyphenated techniques. No other online

resource offers separation scientists more live streaming events, a

knowledge base, practical solutions, and new technologies in one easy

to navigate website.

Get your free five year membership worth US $1,995* by submitting the

form at www.chromacademy.com/agilent.

* Five years free access to CHROMacademy only available to customers affiliated with an academic

or research institution, conditions apply. A valid university e-mail address if required.

© Agilent Technologies, Inc. 2017

FREECHROMACADEMY MEMBERSHIP

ProviFKng the Ultimate Balance of High-Level Performance and Ease-of-Use in an Integrated HPLC

Prominence-i

Most Trusted Name in LC

Nexera-i

Order consumables and accessories on-line at http://store.shimadzu.comShimadzu Scientific Instruments Inc., 7102 Riverwood Dr., Columbia, MD 21046, USA

Ideal for both R&D and QC environments, the i-Series Plus

emerges in response to your requests for design evolution.

Every i-Series has a touch-screen LCD display for easy, in-

tuitive system control and chromatogram viewing, built-in

degassing, quaternary solvent delivery, autosampler, and

UV or PDA detector. As always, sample injections are the

fastest around (< 14 s), with ultra-low carryover.

Learn more about Shimadzu’s i-Series Plus.

Call (800) 477-1227 or visit us online at

www.ssi.shimadzu.com/iseries

You asked for it, you got it. The

i-Series Plus offers everything you

need in an integrated HPLC:

X Automate sample dilution and reagent/

internal standard addition

X Store up to three 350mm LC columns

X Choose the analysis flow path with

convenient software control

X Add a refractive index or fluorescence

detector

X Achieve wider linear range and

repeatability for the smallest injected

volumes of 1 μL or less