epitope mapping of an antibody receptor using hydrogen...

36
Epitope Mapping of an Antibody Receptor Using Hydrogen-Deuterium Exchange MS Enhancing the Sensitivity of Atmospheric Pressure Ionization GC–MS Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates Volume 16 Number 4 October 2018 www.chromatographyonline.com

Upload: others

Post on 24-Jun-2020

11 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

Epitope Mapping of an Antibody Receptor Using

Hydrogen-Deuterium Exchange MS

Enhancing the Sensitivity of

Atmospheric Pressure Ionization GC–MS

Peptide Mapping of Monoclonal Antibodies

and Antibody–Drug Conjugates

Volume 16 Number 4 October 2018

www.chromatographyonline.com

Page 2: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

© 2018 Thermo Fisher Scientifi c Inc. All rights reserved.All trademarks are the property of Thermo Fisher Scientifi c and

its subsidiaries unless otherwise specifi ed. AD65342_EN 07/18

Get confi dent at

thermofi sher.com/confi dentquantitation

Confi dent Quantitation

To achieve your business and scientifi c goals, you need results you can count on. Regardless of

your application, Thermo Scientifi c™ TSQ™ series triple quadrupole mass spectrometers deliver

unprecedented precision for your quantitative workfl ows. High-resolution selective reaction

monitoring (SRM), robustness, reliability, and sensitivity come together—now every user in every

lab can obtain high-confi dence data, regardless of the matrix and molecule analyzed.

Any compound, any matrix, any user

Thermo Scientifi c™ TSQ Quantis™

Triple Quadrupole Mass

Spectrometer

Thermo Scientifi c™ TSQ Fortis™

Triple Quadrupole Mass

Spectrometer

Thermo Scientifi c™ TSQ Altis™

Triple Quadrupole Mass

Spectrometer

Page 3: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

Don’t Wait for AnswersGet them with BaySpec’s Portable Mass Spectrometers

Chemical Warfare

Pathogen Detection Security

Oil and Gas

Food Safety Biomedical

Applications for Onsite Monitoring in Real Time

Features:

Q Miniature linear ion trap, MS/MS capabilityQ High sensitivity, parts-per-trillion level of detectionQ Fully field portable and deployableQ Dual ionization, internal EI and external APIQ Compatible with other ionization sources

Page 4: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

4 Current Trends In Mass Spectrometry October 2018 chromatographyonl ine .com

®

50

% Recycled Paper 1

0-2

0%

Post Consumer W

as

te

MANUSCRIPTS: To discuss possible article topics or obtain manuscript preparation

guidelines, contact the editorial director at: (732) 346-3020, e-mail: Laura.Bush@

ubm.com. Publishers assume no responsibility for safety of artwork, photographs, or

manuscripts. Every caution is taken to ensure accuracy, but publishers cannot accept

responsibility for the information supplied herein or for any opinion expressed.

CHANGE OF ADDRESS: Send change of address to LCGC North America,

P.O. Box 6196, Duluth, MN 55806-6196; provide old mailing label as well as

new address; include ZIP or postal code. Allow 4–6 weeks for change.

RETURN ALL UNDELIVERABLE CANADIAN ADDRESSES TO: IMEX Global Solutions, P.O. Box

25542, London, ON N6C 6B2, CANADA. PUBLICATIONS MAIL AGREEMENT No.40612608.

REPRINT SERVICES: Reprints of all articles in this issue and past issues 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 Advanstar@

wrightsmedia.com for more information. Please note that Wright’s Media is the only

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

C.A.S.T. DATA AND LIST INFORMATION: Contact Melissa Stillwell, (218) 740-6831;

e-mail: [email protected]

INTERNATIONAL LICENSING: Jillyn Frommer, (732) 346-3007,

fax: (732) 647-1104; e-mail: [email protected]

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

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

information storage and retrieval without permission in writing from the publisher. Authorization to

photocopy items for internal/educational or personal use, or the internal/educational or personal

use of 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 customers’ names, addresses,

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 Ameri-

cas 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.

LCGC North America welcomes unsolicited articles, manuscripts, photographs, illustrations and

other materials but cannot be held responsible for their safekeeping or return.

UBM Americas (www.ubmamericas.com) is a leading worldwide media company 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 marketing 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.

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

Fax: (732) 647-1235

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

Edward FantuzziPublisher

[email protected]

Stephanie Shaffer Sales Manager

[email protected]

Brianne Molnar Sales Manager

[email protected]

Michael KushnerSenior Director, Digital Media

[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]

Kristen MooreWebcast Operations Manager

[email protected]

Vania OliveiraProject Manager

[email protected]

Sabina AdvaniDigital Production Manager [email protected]

Kaylynn Chiarello-EbnerManaging Editor, Special Projects

[email protected]

Dan Ward Art Director

[email protected]

Brianne Pangaro Marketing Associate

[email protected]

Melissa Stillwell C.A.S.T. Data and List Information

[email protected]

Wright’s Media Reprints

[email protected]

Jillyn FrommerPermissions

[email protected]

Jesse Singer Production Manager

[email protected]

Wendy Bong Audience Development Manager

[email protected]

Morgan Hight Audience Support [email protected]

Page 5: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

© 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.

Page 6: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

6 Current Trends in Mass Spectrometry October 2018 chromatographyonl ine .com

Articles

October 2018

Epitope Mapping of an Interleukin Receptor for Three Therapeutic Antibodies by Hydrogen-Deuterium Exchange Mass Spectrometry 8

Eric Largy, Caroline Cajot, and Arnaud Delobel

Among all the analytical techniques available for epitope mapping studies, hydrogen–deuterium exchange mass spectrometry (HDX-MS) is usually the fastest and easiest to carry out. We present here the epitope mapping of three distinct monoclonal antibody (mAb) candidates targeting the same antigen, an interleukin receptor. The goal is to establish the binding mode of these mAbs, and explain possible differences observed for in vitro binding and in vivo function.

Enhancing the Sensitivity of Atmospheric Pressure Ionization Mass Spectrometry Using Flow Modulated Gas Chromatography 15

Karl J. Jobst, John V. Seeley, Eric J. Reiner, Lauren Mullin, and Adam Ladak

The past decade has witnessed resurgent interest in coupling GC to atmospheric-pressure chemical ionization (APCI), which is suitable for the high column flows required for using flow modulation. This study assesses the use of GP-APCI with flow modulation for sensitive detection of selected trace organics.

Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates Using Micro-Pillar Array Columns Combined with Mass Spectrometry 20

Koen Sandra, Jonathan Vandenbussche, Isabel Vandenheede, Bo Claerebout,

Jeff Op de Beeck, Paul Jacobs, Wim De Malsche, Gert Desmet, and Pat Sandra

The structural complexity of monoclonal antibodies (mAbs) challenges the capabilities of even the most advanced chromatography and mass spectrometry techniques. This study examines the use of micro-pillar array columns in combination with mass spectrometry for peptide mapping of both mAbs and antibody–drug conjugates (ADCs).

Departments

Product Profiles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31

Application Notes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32

Cover image courtesy of ustas/stock.adobe.com

Page 7: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

NM 1350 Plus Hydrogen Generator

[email protected] www.vicidbs.com +1 (713) 263 6970

One generator.

Enough ultra high purity hydrogen for up to 27 GCs.

The NEW VICI DBS NM Plus 1350 Hydrogen

Generator uses the same space saving cabinet

as the existing NM range, but now with a higher

flow rate of 1350 ml/min.

With the higher flow rate and 11 bar outlet

pressure, only one generator is needed to

supply up to 27 GCs with detector gas.

Improves workplace safety and lab efficiency

Ultra high purity carrier grade gas with

convenient software control and safety alarm

capability. It gives the GC user the option for

on-demand hydrogen gas while managing

safety concerns.

TM

Page 8: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com8 Current Trends in Mass Spectrometry October 2018

Monoclonal antibodies (mAbs) constitute a major and fast-growing biotherapeutic class, thanks notably to their outstanding selectivity for specific targets.

Their physicochemical characterization is complex, because of their size (around 150 kDa), numerous variants caused by post-translational modifications, and their tendency to aggre-gate (1). Furthermore, there is a need for better understanding of how these aspects may alter the structure and function of mAbs, as observed in vivo or in vitro by techniques such as enzyme-linked immunosorbent assay (ELISA) or surface plasmon resonance (SPR). This need prompts the study of the precise nature of the interaction between mAbs and their antigens at the molecular level. Epitope mapping consists of the determination of the binding site of the antigen (coined epitope), and provides several very useful applications:• A way to screen mAb candidates and assist in the devel-

opment of new candidates (2)• A better understanding of the mechanisms of action (3–5)• An additional layer of intellectual property protection (6).

A number of methods can be used to map interactions between proteins. X-ray crystallography is probably the gold

standard for such endeavors (7,8), but it suffers from several drawbacks. Most notably, it is a lengthy process that relies on the amenability of the proteins to crystallize, and “only” yields a snapshot of one energetically accessible conformation. In that regard, solution-based techniques are best suited to study dy-namic systems because they take into account all attainable conformations. Nuclear magnetic resonance (NMR) experi-ments provide similar resolution but are often too difficult to carry out in this context (9). Other methods such as circular dichroism or mutagenesis usually lack in resolution or do not provide direct evidence, respectively.

The hydrogen–deuterium exchange mass spectrometry (HDX-MS) approach is much faster and easier to carry out than the aforementioned methods. Although it does not provide atomic resolution, the approach allows the determination of epitopes at the scale of a few amino acids (regardless of the com-plexity of the sample), requires smaller sample amounts, tolerates impurities and formulants (such as salts and buffers), and oper-ates in solution in near-physiological conditions. Furthermore, it provides insight into the structure and interaction dynamics on a large time scale (from a few seconds to several hours).

Eric Largy, Caroline Cajot, and Arnaud Delobel

The study of protein-protein interactions is critical in the development of biotherapeutics, to

develop new candidates, to understand modes of actions, and to protect intellectual property.

Among all the analytical techniques available for epitope mapping studies, hydrogen–deuterium

exchange mass spectrometry (HDX-MS) is usually faster and easier to carry out. We present here

the epitope mapping of three distinct monoclonal antibody (mAb) candidates targeting the same

antigen, an interleukin receptor. The goal is to establish the binding mode of these mAbs, and

explain possible differences observed for in vitro binding and in vivo function.

Epitope Mapping of an

Interleukin Receptor for Three

Therapeutic Antibodies by

Hydrogen–Deuterium Exchange

Mass Spectrometry

Page 9: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 9

We describe below the epitope map-ping of three distinct mAb candidates targeting the same antigen, an interleu-kin receptor. The goal is to establish the binding mode of these mAbs, and ex-plain possible differences observed for in vitro binding and in vivo function.

Experimental

Principles of the Method

The analytical workflow for HDX-MS is summarized in Figure 1. Proteins in solution are diluted in an excess of deuterium oxide (D2O) so that hydro-gen atoms are exchanged for deuterium atoms on the amide nitrogen of the pep-tide bond (10,11). Side-chain H atoms also exchange, but at too fast a rate, and are consequently entirely back-ex-changed during the liquid chromatog-raphy–MS (LC–MS) analysis (that is, hy-drogen atoms replace deuterium atoms). After quenching the deuteration by low-ering both the temperature and the pH, the protein is digested online by a pro-tease. The resulting peptides are quickly desalted and separated at low tempera-ture. Finally, MS measures the uptake of deuterium using the difference in peptide mass resulting from the mass difference between exchanged deute-rium (2.0141 Da) and hydrogen (1.0078 Da). This exchange of D for H occurs at rates varying by a factor of as much as 108 depending on hydrogen bonding and solvent accessibility (Figure 2) (12). Schematically, hydrogen atoms from accessible regions and hydrogen atoms not involved in H-bonds exchange faster. By measuring hydrogen–deuterium ex-change rates, changes in protein struc-ture and dynamics are determined. Note that the exchange rate is also influenced

by experimental parameters such as the temperature and pH (13).

Concretely, binding a mAb on the antigen buries the amino acids from the binding sites, hence screening them from the bulk medium and ultimately slowing down the H-to-D exchange. In the approach described in this article, the proteins are digested online with a pepsin column, which generates a large number of partially overlapped peptides and allows the analysis of ex-change data at the scale of a few amino

D2O labeling

pH 7.0, 20 °C

Quench

pH 2.5, 1 °C

On-line pepsin

digestion

pH 2.5, 20 °C

Filtering and analysis Identification

LC–MS

separation and

detection,

0 °C

Figure 1: Analytical workflow of hydrogen–deuterium exchange mass spectrometry.

+1-203-949-8697www.ProtonOnSite.com

PROTON ONSITE LAB SERVERS

FEED MULTIPLE UNITS

NOURISH YOUR ANALYTICAL EQUIPMENT

WITH HIGH PURITY GAS TODAY

• H2 and N

2 Servers

• Consistent Purity• Consistent Pressure• Proven Safe• Cost Effective• Eliminates Cylinder

Storage and Delivery Issues

Page 10: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com10 Current Trends in Mass Spectrometry October 2018

acids. The epitope mapping can there-fore be conducted by a straightforward comparison of the exchange data of the antigen unbound and bound to the mAb for each peptide. Peptides for which a statistically significant decrease in deuterium uptake is found are likely to be part of the epitope. By combining the results of all peptides, the epitope can be more finely characterized.

Analytical Workflow

Brief ly, the experiments, which were replicated at least three times, can be de-

scribed as follows: The samples were di-luted ten-fold into a deuterated buffer for 30 min, after which they were quenched at a low pH (2.5) and a low temperature (1.0 °C) to minimize the amount of back exchange. Furthermore, to ease the on-line digestion of the proteins in subop-timal conditions, the quench buffer was complemented with high concentra-tions of a reducing agent that can oper-ate at acidic pH and a chaotropic agent, namely TCEP and guanidine.

Proteins were then injected in the LC system, where they were quickly digested

in an immobilized pepsin column, at 20 °C, then desalted on a reversed-phase cartridge, and separated on an analyt-ical reversed-phase column at 0.0 °C (again, to minimize the back exchange). The system was coupled to a high-res-olution electrospray ionization quadru-pole time-of-flight (ESI-Q-TOF) mass spectrometer for peptide identification and deuteration quantitation purposes, which were assisted by various software solutions (PLGS, UNIFI, and DynamX, all from Waters). Undeuterated peptides (no incubation in D2O) were used as ref-erence point and for MSE-based identi-fication, in which the sequence of the peptides was systematically confirmed by collision-induced dissociation (CI-D)-based fragmentation. Their observed retention times and theoretical isotope profiles were subsequently used to detect the corresponding deuterated peptides and quantify their deuteration.

Hardware

The HDX experiments were conducted using an HDX-2 system from Waters, consisting of an automated sample prepa-ration bench (which performs the incu-bation in deuterium, quenching and in-jection steps at controlled temperature), two pumps (supplying the mobile phases to the digestion, desalting, and analyti-cal columns). A module allows the tem-perature to be set independently for the digestion column on one hand, and de-salting and analysis on the other. Deuter-ation and digestion were performed at 20 °C, while the quench and desalting/sepa-ration at 1 and 0 °C, respectively. Samples and Buffers

The interleukin receptors unbound and bound to the antibody were prepared at around 25 μM in each partner, in an equilibration buffer composed of 10 mM sodium phosphate complemented with sodium chloride (100 mM) and adjusted to pH 6.8. Given that the KD of the mAb–antigen complex lies in the 10-10 M range, we can assume that more than 99.7% of 1:1 complex is formed at this concentra-tion, hence maximizing the observable effects without resorting to the use of an excess of mAb. Given that the epitope mapping is performed by studying pep-tides from the antigen, it is preferable to

100

120

2 3 4 5 6 7 8 10 11 129

100

80

60

40

20

0

8.38

Peptide 126-137n = 60.32%RSD

02.00

Cu

mu

lati

ve

de

tect

ed

pe

pti

de

s

4.00

Retention time

6.00 8.00 10.00 12.00 14.00 16.00

Time

%

Figure 3: Six overlaid LC–MS chromatograms of a randomly selected interleukin receptor peptide

(top), and cumulative peptides elution as a function of time (bottom).

D2O

HD

Figure 2: D-labeling of a protein in the presence of an excess of D2O.

Page 11: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 11

5

60 65 70 75 80 85 90 95 100 105 110

120

180

137 peptides, 99.6% sequence coverage, 6.44 average redundancy

N-glycosylation site reported and observed by HDX-MS

N-glycosylation site unreported but observed by HDX-MS

Disulfide bridge (reported)N-glycosylation concensus sequence (no glycosylation reported nor obseved by HDX-MS)

185 190 195 200 205 210 215 220 225 230 235

125 130 135 140 145 150 155 160 165 170 175

115

10 15 20 25 30 35 40 45 50 55

Figure 4: Sequence coverage of the interleukin receptor. Peptides are depicted by blue bars, disulfide

bridges by green lines, and N-glycosylation sites by orange frames.

minimize the quantity of mAb peptides that may complicate the detection, iden-tification, and deuterium uptake quanti-tation of the antigen peptides, because of the large occurrences of peptide coelution resulting from the nonoptimal separation conditions (10 min gradient at 0 °C).

The deuteration buffer composition was matched to the equilibration buf-fer composition, but in D2O instead of water. The pH was also matched by ad-justing its apparent value at 6.4, given that pD = pH – 0.4 (14). The quench buffer was composed of 100 mM so-dium phosphate, 400 mM TCEP, and 4 M guanidine, and was adjusted at pH 2.3, hence ensuring that the post-quench pH of the solution was 2.5 by 1:1 mixing. Finally, mobile phases involved in the online digestion and analytical separation of the peptides were water and acetonitrile, adjusted to pH 2.5 with 0.2% formic acid.

Results

Analytical Separation

For peptide identification and deute-rium quantitation purposes, it is pref-

REVEALTHE INVISIBLEPhotonis engineers and manufactures custom ion and electron

detectors and inlet tubes for use in mass spectrometers and other

analytical instruments. We offer a comprehensive selection of

detectors suitable for portable and benchtop instruments which

provide superior lifetime and enhanced performance so you will be

the first to identify any unknown matter.

Learn how our technology can refine your analysis. Visit us at

analytica China stand E4.4706 or at www.photonis.com.

Page 12: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com12 Current Trends in Mass Spectrometry October 2018

erable to limit coelution events as much as possible. Although a short gradient at low temperature cannot extensively separate the large number of peptides entering the column, it is still possible to ensure that there is no large skew-ing of peptide elution leading to a very large number of them being eluted in a short time range. Figure 3 illustrates the fairly linear elution of undeuterated peptides that was obtained for the inter-leukin receptor using our experimen-tal conditions, which limited the occur-rences of coelution. Furthermore, six injections of independently prepared samples exhibited very little retention time deviation for a randomly selected peptide (0.32% RSD). Calculated across 399 peptides, the average retention time (RT) standard deviation was as low as 0.009 min, which is critical to ensure proper identification of the deuterated peptides later in the experiment.

Sequence Coverage

The sequence coverage, peptide length, and average peptide redundancy per amino acid are important parameters regarding the quality of the results, notably in terms of confidence and resolution. It is, of course, preferable to cover the protein sequence as much as possible to avoid missing a binding site. Shorter peptides yield higher res-olution results, especially when com-bined with high redundancy where partially overlapped peptides allow narrowing the sequence of the binding site. A high redundancy also increases the confidence in the results because it provides a means to evaluate the con-sistency of the results.

An excellent sequence coverage, together with a very good average re-dundancy, was obtained (99.6%, 6.44 peptides per amino acid; see Figure 4) from 137 peptides identified using both the PLGS and UNIFI software. As ex-pected, the redundancy is not homo-geneous; the N-terminal half contains three disulfide bridges and is thus more difficult to digest than the C-terminal half, which does not have any. Hence, the average redundancies of the 8–90 and 178–225 regions are 4.7 and 8.2, respec-tively. All regions are covered sufficiently well to gather high-quality results.

Pooled SD

93—125

71—85

93—125

93—108

125—14167—93

67—85

10

Deute

rium

up

take

(D

a)

Deute

rium

up

take

(D

a)

Deute

rium

up

take

(D

a)

8

6

4

2

0

-2

8

6

4

2

0

-2

8

6

4

2

0

-2

Complex

mAb A

mAb B

mAb C

DifferenceIL

Figure 5: Deuterium uptakes of the interleukin receptor (blue) and complex (orange) peptides (from

N- to C-terminal), and corresponding difference (green). The pooled standard deviation is shown as

a mirrored grey area. Arbitrary thresholds at ±0.5 and ±1.0 Da are illustrated by dashed and plain

gray lines, respectively.

5

60 65 70

75 80 85 90 95 100 105 75 80 85 90 95 100 105 75 80 85 90 95 100 105

110 120

180 185 190 195 200 205 210 180 185 190 195 200 205 210 180 185 190 195 200 205 210

215 220 225 230 235

Relative fractional uptake (%)

No protection

mAb A mAb B mAb C

Maximum protection

215 220 225 230 235 215 220 225 230 235

125 130 135 140

145 150 155 160 165 170 175 145 150 155 160 165 170 175 145 150 155 160 165 170 175

115 110 120 125 130 135 140115 110 120 125 130 135 140115

10 15 20 25 30 35 5 10 15 20 25 30 35 5 10 15 20 25 30 35

40 45 50 55 60 65 7040 45 50 55 60 65 7040 45 50 55

Figure 6: Heatmap of the deuteration differences between the unbound and complexed interleukin

receptor (bottom). The inferred epitope site 1 (blue, red and orange) and site 2 (green) are shown

on the 3D structure (top).

Page 13: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 13

mA

b B

mA

b A

mA

b C

70

SS

MD

SS

MD

SS

MD

50

30

10

-10

-30

-50

-70

8.0

6.0

4.0

2.0

-2.0

0.0

-4.0

-6.0

-8.0

3.5

2.5

1.5

0.5

-0.5

-1.5

-2.5

-3.5-2.5 -2.0 -1.5 -1.0 -0.5 0.0

Average Log2 fold change

1.0 1.5 2.0 2.50.5

-2.5 -2.0 -1.5 -1.0 -0.5 0.0 0.5 1.0 1.5 2.0 2.5

-3.0 -2.5 -2.0 -1.5 -1.0 -0.5 0.0 0.5 1.0 1.5 2.0 2.5 3.0

Figure 7: Dual-flashlight plot for the discovery of peptides with a statistically significant protection.

Epitope Determination

The interleukin receptor deuteration D was calculated as an average of the ex-perimental replicates and compared peptide per peptide between the two states (bound versus unbound). The differences between both states were calculated by equation 1, where IL and cplx refer to the interleukin receptor and complex, respectively:

ΔD=DIL

-Dcplx

[1]

This calculation means that the bind-ing of the mAb on a given peptide would result in a positive ΔD value for said pep-tide. For instance, such observations can be made for regions 71–85 and 93–125 of the antigen in presence of mAb A (Figure 5), where the difference is above 1.0 Da, an arbitrary threshold often used in the litera-ture (15). mAb B has an additional neigh-boring region above this threshold (region 125–141), whereas mAb C only has a few peptides barely above the limit, suggest-ing a lower binding strength, or possibly a different binding mode resulting in a de-creased shielding of the backbone hydro-gen atoms from the bulk medium. These regions, likely constituting the epitope, are clearly highlighted in a heatmap colored by ΔD (Figure 6). Large regions (regions 1–60, 140–190, and 201–236) show virtu-ally no difference between the two states, whereas other regions (regions 126–139 and 191–200) display slightly higher ΔD values. Incidentally, a secondary epitope site may be hypothesized in the 191–200 region of mAb A only, as also observed by orthogonal techniques (data not shown).

The statistical significance of uptake differences can be assessed by calculat-ing the corresponding pooled standard deviation SD

P, following equation 2,

where SD is the standard deviation in-ferred from the replicates.

SDp= +SD

ILSD

cplx√2 2 [2]

The hypothesized epitopes exhibit differences larger than their corre-sponding pooled standard deviation for several peptides, which suggests that there is a statistically significant binding event taking place within these peptides. A few peptides from the secondary site also ex-hibit this property for mAb A. There is no

Page 14: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com14 Current Trends in Mass Spectrometry October 2018

peptide displaying a significantly negative ΔD in any case, suggesting that no other change in protein conformation leading to more exchangeable hydrogen atoms is taking place upon binding.

To quickly screen all peptides for each complex, an alternative visualization of the results was set up, taking into ac-count both the magnitude of the change in deuteration and the pooled standard deviation. This approach allows the quick identification of the peptides with the most significant changes in deutera-tion upon binding, hence yielding a ro-bust epitope. This result was achieved by calculation of the SSMD (strictly stan-dardized mean difference), following equation 3.

SSMD = = —

DIL

−Dcplx

√2 2+

ΔD

SDpSD

ILSD

cplx

[3]

Each peptide’s SSMD was plotted against the deuteration change magnitude D

IL/D

cplx, expressed as a base 2 logarithm

(Figure 7). As expected, the best peptides (located in the top right-hand corner) are located on the hypothesized primary epi-tope site, for all three mAb candidates. Note that the experimental conditions used in our study (1:1 molar mixture of the two proteins), together with the excel-

lent KD

of the primary epitope, disfavor the discovery of secondary sites.

Finally, the changes in deuteration for the hit peptides are qualitatively evident from the corresponding mass spectra, which further strengthens the epitope determination (Figure 8).

Conclusion

Using HDX-MS, the epitope of three mAbs were straightforwardly inferred, in solution, for an interleukin receptor tar-get. Qualitative and quantitative analysis allowed the identification of the peptidic regions of the antigen that are protected from deuteration upon antibody binding. The resulting epitope location is analo-gous for all three candidates, although one mAb seems to also bind a secondary site.

References

(1) A. Beck, E. Wagner-Rousset, D.

Ayoub, A. Van Dorsselaer, and S.

Sanglier-Cianférani, Anal. Chem.

85, 715–736 (2013). doi:10.1021/

ac3032355.

(2) G.F. Pirrone, R.E. Iacob, and J.R.

Engen, Anal. Chem. 87, 99–118

(2015). doi:10.1021/ac5040242.

(3) V.C. Casina, W. Hu, J. Mao, R. Lu, H.A.

Hanby, B. Pickens, Z.-Y. Kan, W.K.

Lim, L. Mayne, E.M. Ostertag, S. Kacir,

D.L. Siegel, S.W. Englander, and X.L.

Zheng, Proc. Natl. Acad. Sci. U. S. A.

112, 9620–9625 (2015). doi:10.1073/

pnas.1512561112.

(4) J.C. Geoghegan, G. Diedrich, X. Lu, K.

Rosenthal, K.F. Sachsenmeier, H. Wu,

W.F. Dall’Acqua, and M.M. Damschroder,

mAbs 8, 454–467 (2016). doi:10.1080/1

9420862.2016.1143182.

(5) C. Jost, J. Schilling, R. Tamaskovic, M.

Schwill, A. Honegger, and A. Plück-

thun, Structure 21, 1979–1991 (2013).

doi:10.1016/j.str.2013.08.020.

(6) C.G. Sandercock and U. Storz, Nat.

Biotechnol. 30, 615–618 (2012).

doi:10.1038/nbt.2291.

(7) E. Malito, A. Carfi, and M. Bottomley,

Int. J. Mol. Sci. 16, 13106–13140 (2015).

doi:10.3390/ijms160613106.

(8) F. Dall’Antonia, T. Pavkov-Keller, K.

Zangger, and W. Keller, Methods

66, 3–21 (2014). doi:10.1016/j.

ymeth.2013.07.024.

(9) M. Bardelli, E. Livoti, L. Simonelli, M.

Pedotti, A. Moraes, A.P. Valente, and L.

Varani, J. Mol. Recognit. 28, 393–400

(2015). doi:10.1002/jmr.2454.

(10) J.J. Skinner, W.K. Lim, S. Bédard, B.E.

Black, and S.W. Englander, Protein

Sci. 21, 987–995 (2012). doi:10.1002/

pro.2082.

(11) J.J. Skinner, W.K. Lim, S. Bédard, B.E.

Black, and S.W. Englander, Protein Sci.

21, 996–1005 (2012). doi:10.1002/

pro.2081.

(12) S.W. Englander, T.R. Sosnick, J.J. En-

glander, and L. Mayne, Curr. Opin.

Struct. Biol. 6, 18–23 (1996).

(13) Y. Bai, J.S. Milne, L. Mayne, and S.W.

Englander, Proteins 17, 75–86 (1993).

doi:10.1002/prot.340170110.

(14) A.K. Covington, M. Paabo, R.A. Rob-

inson, and R.G. Bates, Anal. Chem.

40, 700–706 (1968). doi:10.1021/

ac60260a013.

(15) J. Fang, C. Doneanu, W.R. Alley, Y.Q. Yu,

A. Beck, and W. Chen, mAbs 8, 1021–

1034 (2016). doi:10.1080/19420862.20

16.1193661.

Eric Largy, Caroline Cajot, and Arnaud

Delobel are with Quality Assistance SA, in

Donstiennes, Belgium. Direct correspondence

about this article to arnaud.delobel@quali-

ty-assistance.be.

78-84

402.0 403.0 404.0 405.0 406.0

m/z

407.0 408.0 14.5 415.5 416.0 417.5

m/z m/z

418.5 419.5 667 669 670 671 672 673 674 675 676 677668

85-93 112-123

Undeuterated reference

Antigen

Complex

Figure 8: Examples of mass spectra of three peptides (peptides 78–84, 85–93, and 112–123) from

the undeuterated antigen reference (blue, top), the deuterated antigen (red, middle), and complex

(green, bottom) samples.

Page 15: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 15

The exposome represents all environmental exposures over the course of a lifetime (1). The identities of most environmental contaminants and their roles

in causing chronic diseases are unknown. This is partly a result of the analytical challenges associated with biomon-itoring trace levels of environmental toxicants in small sample quantities of serum or tissue. It is estimated that standard gas chromatography–mass spectrometry (GC–MS) and liquid chromatography–MS (LC–MS) platforms are not sufficiently sensitive to identify approximately 90% of unknown contaminants (2).

Inspired by advances in comprehensive two-dimensional gas chromatography (GC×GC) (3), Patterson and coworkers elegantly showed that (targeted) sub-femtogram detection of environmental pollutants is achievable using a technique termed cryogenic zone compression (CZC) (4). With CZC, the 3–8 s width of a GC peak is compressed to approxi-mately 200 ms, resulting in a significant enhancement in signal intensity (4–6). This has opened the door to the pos-

sibility of monitoring environmental toxicants using less invasive and inexpensive sampling techniques, such as collection of dried blood spots (7). The cost of requisite hardware, however (such as a thermal modulator), and associated cryogens and refrigeration is a drawback.

Signal enhancement is also possible using a valve-based f low modulator, a technology pioneered by J.V. Seeley (Oakland University, Michigan, USA) (8,9). In contrast to thermal modulation, a f low modulator requires a high carrier gas f low (>10 mL/min) to transfer the column ef-f luent from the modulator into the mass spectrometer. The potential of f low modulation has not been fully re-alized, because of the perception that high column f lows are not compatible with most (electron ionization [EI] and chemical ionization [CI]) mass spectrometers. This is usually addressed by eff luent splitting at the cost of sensitivity. Improved pumping capacity of modern turbo-molecular pumps can mitigate the problem, but still this hyphenation is generally considered difficult (10,11).

Karl J. Jobst, John V. Seeley, Eric J. Reiner, Lauren Mullin, and Adam Ladak

Peak intensity enhancement is one highly desirable outcome of comprehensive two-dimensional gas

chromatography (GC×GC). When coupled to mass spectrometry (MS), such enhancement is usually

achieved with a thermal modulator using a technique called cryogenic zone compression (CZC).

Differential flow modulation is a simple and cost-effective alternative to thermal modulation, but the

requisite high flow rates are generally perceived as being incompatible with most (electron ioniza-

tion [EI] and chemical ionization [CI]) mass spectrometers. The past decade has witnessed resurgent

interest in coupling GC to atmospheric pressure chemical ionization (APCI), which requires high gas

flows to assist ionization. This article reports on the modification of a GC–APCI system with a flow

modulator and evaluates its potential to enhance the sensitivity towards selected trace organics.

Enhancing the Sensitivity of

Atmospheric Pressure Ionization

Mass Spectrometry Using Flow

Modulated Gas Chromatography

Page 16: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com16 Current Trends in Mass Spectrometry October 2018

(a)

(b)

Experimental

The experiments were performed using a Xevo G2-XS quadrupole time-of-f light mass spectrometer (Waters) coupled to a modified 7890B gas chro-matograph (18,19) (Agilent). In GC–APCI, the GC effluent is swept into the ion source by a 350 mL/min make-up flow (N2). A schematic of GC–APCI is shown in Figure 1(a).

Ions formed by a corona discharge needle set to 3 μA were transferred into the mass spectrometer, aided by 20 V potential and cone and auxil-iary gas f lows of 175 L/h and 100 L/h, respectively. In the charge transfer mode (Figure 1[b]), ionization was typically initiated by the formation of N2

•+ and Nn•+ cluster ions, which may

undergo charge exchange with ana-lyte molecules (M) provided the ion-ization energy (IE) of M is lower than the recombination energy (RE) of N2

•+. Non-reactive collisions between the incipient ions and the surround-ing N2 results in collisional cooling that significantly reduces fragmenta-tion and therefore also the detection limits for the molecular ions (M•+). Other ionization schemes are also possible by introducing dopants to the ion source. For example, H2O can be used to promote protonation. O2

•- is the primary charge carrier in negative ion mode APCI, which can result in selective ionization and structure diagnostic fragmentation of halogenated compounds (18,20). The technique of ionizing a sample of molecules by gas-phase ion-mol-

Figure 1: (a) The atmospheric pressure gas chromatography (APGC) ion source; (b) general scheme for positive mode ionization in dry ion source.

Figure 2: The flow modulator used in this study.

GC coupled to atmospheric pres-sure chemica l ionizat ion (APCI) (12–16) is, by contrast, ideally suited to high f lows. In GC–APCI, the GC eff luent is swept into the ion source, which is held at atmospheric pres-sure, by a high f low of nitrogen gas (>100  mL/min). There is a lso the additional benefit that the elevated pressures promote collisional cooling of the incipient ions. Consequently, very little fragmentation occurs, re-sulting in further signal enhance-ment of the molecular ion. With widespread adoption of atmospheric

pressure ionization techniques, such as electrospray ionization (ESI) and APCI, there is growing interest in coupling GC–MS using atmospheric pressure ionizat ion. This ar t icle reports on proof-of-concept exper-iments using a simple, cost-effec-tive f low modulator (17) coupled to a quadrupole t ime-of-f l ight mass spectrometer (QTOF-MS). The re-sults indicate that environmental contaminants can be detected at low femtogram levels using a full-scan-ning instrument capable of nontar-geted analysis.

Page 17: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 17

Figure 3: (a) Comparison of pre- and post-modulation of 500 fg of TCDD; (b) CZC of 500 fg of

13C-TCDD and 2.5 fg of TCDD. Experiments were performed using a Zoex ZX2 thermal modulator.

Columns: 15 m × 0.25 mm, 0.1-μm db-5 (Agilent) and 0.18 mm × 0.18 μm, 40-m db-5 (Agilent)

were employed for experiments (a), and (b) and (c), respectively.

used to explore the potential of cou-pling f low modulation to GC–APCI. The construction, operation, and performance of this device has been described recently (17). Brief ly, the multimode modulator (17) of this study was constructed around a 1/8” outside diameter (o.d.) liner (standard PTV injector liner) (Figure 2).

A piece of tubing or capillary can also be used for primary eff luent col-lection, but the 1/8” o.d. liner provided the volume required (~200 μL) to ac-commodate a 4 s modulation period and 3 mL/min carrier gas f low. One end of the joining liner was attached to a cross union, or as shown in Figure 2, a modified tee. The exit of the pri-mary column (15 m × 0.25 mm, 0.1-μm db-5 [Agilent Technologies]) was in-serted through the cross and then into the joining liner, so that the column exit sat approximately 1 cm from the gooseneck of the liner. The opposite end of the liner was attached to a tee union. A 0.5 m × 0.53 mm Rxi Guard

ecule reactions is well studied (20), and, since its inception, an enormous number of studies have appeared on its applications in analytical MS.

A ZX2 thermal modulator (Zoex) was used for CZC experiments. A multimode modulator (17) designed and constructed by J.V. Seeley was

Page 18: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com18 Current Trends in Mass Spectrometry October 2018

column (Restek) that served as the transfer line to the MS was inserted into the tee and butted against the gooseneck. Auxiliary f low (125 mL/min) was delivered to the tee and cross unions via a two-way, three-port solenoid valve (not shown). A f low restrictor was connected to the cross connection. The operation of the multimode modulator for the purpose of signal enhancement is described below.

Results and Discussion

CZC relies on the same principles of operation as GC×GC, whereby a pulse of cooled nitrogen gas is used to condense and focus GC eff luent

(primary peak), followed by a pulse of heated gas for re-injection (secondary peak). The timing of the two pulses (modulation period) is configured so that the width of a GC peak is com-pressed. This is i l lustrated in Fig-ure 3(a), which displays a ~3 s wide peak prior to modulation and a nar-row (250 ms wide) peak post modula-tion. The tenfold enhancement in sig-nal intensity (Figure 3[a]) enables low femtogram detection of 2378-TCDD (tetrachlorodibenzo-p-dioxin) using QTOF, see Figure 3(c). Note that the identity of the native compound (Fig-ure 3[c]) is confirmed by comparison with the corresponding 13C-labelled compound (Figure 3[b]).

A flow modulator does not condense or focus the GC eff luent. Instead, the effluent is trapped in a sample loop of sufficient volume to accommodate the primary flow. The width of the second-ary peak can still be controlled by the secondary f low. Figures 4(a) and 4(b) show the flow modulator of this study in the collection and injection states, respectively. In the collection state, ef-fluent from the primary column (3 mL/min) backfills into the liner as primary carrier gas escapes via the flow restric-tor. In the injection state (Figure 2[c]), the secondary flow (125 mL/min) is di-verted through the sample loop, flush-ing the accumulated contents into the mass spectrometer.

From the ratio of the primary and secondary f lows, one anticipates a signal enhancement of approxi-mately 40×. As shown in Figure 5(a), the signal-to-noise ratios (S/N) of two polychlorinated biphenyls (101 and 118) are approximately 6:1. When the modulator is activated, this S/N in-creases by a factor of 20 to 124:1 (see Figure 5[c]). Note that the acquisition time of the mass spectrometer is nor-mally set to acquire ~10 data points across the peak. As shown in Figure 5(b), the S/N of the primary peaks in-creases to 24:1 when the acquisition rate is adjusted from 30 Hz to 4 Hz. Still, signal enhancement of approxi-mately 10:1 is evident from the over-laid chromatograms in Figure 5(d).

Summary

A f low modulator was constructed and evaluated using QTOF-MS. Like CZC, flow modulation produces sharp peaks (<200 ms wide) with a significant improvement in signal intensity com-pared to unmodulated peaks. Aside from the obvious implications to quan-titative analysis, the enhanced sensi-tivity also helps address the critical need for nontargeted identification of unknown environmental toxicants. The analytical platform described in this study is capable of acquiring high quality, full scan mass spectra of femtogram level toxicants and as such may be a promising tool in en-vironmental monitoring, biomoni-toring, and exposomic research (20).

Figure 4: The home-built flow modulator and its operation in the (a) collection and (b) injection

states.

Page 19: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 19

Time (min)

Time (min)

Time (min)

Time (min)

References

(1) C.P. Wild, Int. J.Epidemiol. 41, 24

(2012).

(2) S.M. Rappaport, D.K. Barupal, D.

Wishart, P. Vineis, and A. Scalbert,

Environ. Health Perspect. 122, 769

(2014).

(3) L. Ramos, Ed., Comprehensive Two

Dimensional Gas Chromatography

(Elsevier, Amsterdam, Netherlands,

2009).

(4) D.G. Patterson Jr., S.M. Welch, W.E.

Turner, A. Sjodin, and J.-F. Focant, J.

Chrom. A 1218, 3274 (2011).

(5) B. L’Homme and J.-F. Focant, Anal.

Methods 8, 249 (2016).

(6) C. Brasseur, B. L’Homme, and J.-F. Fo-

cant, Organohalogen Compounds 74,

55 (2012).

Figure 5: Extracted ion chromatograms of PCB-101 and PCB-118 without modulation (a) 30 Hz

and (b) 4 Hz; and (c) with modulation; (d) overlaid chromatograms (a–c).

(7) R. Guthrie and A. Susi, Pediatrics 32,

338 (1963).

(8) J.V. Seeley, N.J. Micyus, J.D. McCurry,

and S.K. Seeley, Amer. Lab. 38, 24

(2006).

(9) J.V. Seeley, J. Chrom. A 1255, 24

(2012).

(10) R. Shellie, LCGC Europe 21(11), (2008).

(11) M. Poliak, M. Kochman, and A. Amirav,

J. Chrom. A 1186, 189 (2008).

(12) E.C. Horning, M.G. Horning, D.I. Car-

roll, I. Dzidic, and R.N. Stillwell, Anal.

Chem. 45, 936 (1973).

(13) D.-X. Li, L. Gan, A. Bronja, and O.J.

Schmitz, Anal. Chim. Acta 891, 43

(2015).

(14) A. Ballesteros-Gomez, J. de Boer, and

P.E.G. Leonards, Anal. Chem. 85, 9572

(2013).

(15) C.N. McEwen and R.G. McKay, J.

Am. Soc. Mass Spectrom. 16, 1730

(2005).

(16) C.N. McEwen, R.G. McKay, and B.S.

Larsen, Anal. Chem. 77, 7826 (2005).

(17) J.V. Seeley, N.E. Schimmel, and S.K.

Seeley, J. Chrom. A 1536, 6–15 (2018).

(18) S. Fernando, M.K. Green, K. Organ-

tini, F. Dorman, R. Jones, E.J. Reiner,

and K.J. Jobst, Anal. Chem. 88, 5205

(2016).

(19) D. Megson, M. Robson, K.J. Jobst, P.A.

Helm, and E.J. Reiner, Anal. Chem. 88,

11406–11411 (2016).

(20) A.G. Harris, Chemical Ionization Mass

Spectrometry (CRC Press, Boca Raton,

USA, 1983).

(21) J.R. Sobus, J.F. Wambaugh, K.K. Isaacs,

A.J. Williams, A.D. McEachran, A.M.

Richard, C.M. Grulke, E.M. Ulrich, J.E.

Rager, M.J. Strynar, and S.R. Newton,

J. Exposure Sci. Environ. Epidemiol.

(2017) doi: 10.1038/s41370-017-

0012-y.

Karl Jobst is a development scientist with

the Ontario Ministry of the Environment and

Climate Change (MOECC). His research in-

terests focus on the development and appli-

cation of mass spectrometric techniques for

the analysis of complex environmental sam-

ples. John Seeley is a professor of chemis-

try at Oakland University in Rochester, Mich-

igan, USA. For the past 20 years, his research

has largely focused on the development of

flow modulation GC×GC and its application

to separations of complex mixtures of vola-

tile organic compounds. Eric Reiner is an

adjunct professor at the University of To-

ronto and emeritus mass spectrometry re-

search scientist at MOECC. He has spent over

30 years at MOECC working on the develop-

ment of analytical methods for the analysis

of halogenated persistent organic pollutants

(POPs). Lauren Mullin is a principal scien-

tist at Waters Corporation. Her work focuses

on the development of liquid and gas chro-

matography–mass spectrometry methods

to address food, environmental, and various

small molecule research analyses. Adam

Ladak is a senior strategic scientific mar-

keting manager at Waters Corporation. He is

responsible for food and environmental mar-

ket development worldwide with a focus on

environmental research. Direct correspon-

dence about this article to Adam_Ladak@

waters.com.

Page 20: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com20 Current Trends in Mass Spectrometry October 2018

The discovery of monoclonal antibodies (mAbs) in the 1970s and the realization of their therapeutic potential in the following decades has been a key milestone in

medicine (1–3). Today, over 70 mAbs have received regula-tory approval in the US and Europe, of which over 20 display blockbuster status, and over 50 are in late-stage clinical de-velopment (4–6). With the top-selling mAbs evolving out of patent, there has been a growing interest in the development of biosimilars (5,7,8). In 2013, we witnessed the European approval of the first mAb biosimilars and in 2016, the first mAb biosimilar also reached marketing authorization in the US. Since then, a growing number of mAb biosimilars have reached approval in both Europe and the US. The successes of mAbs have furthermore triggered the development of var-ious next-generation formats. In oncology, antibody–drug conjugates (ADCs) are particularly promising, because they

synergistically combine a specific mAb linked to a biologi-cally active cytotoxic drug via a stable linker. The promise of ADCs is that highly toxic drugs can selectively be delivered to tumor cells, thereby substantially lowering side effects as typically experienced with classical chemotherapy. Currently two ADCs are marketed and over 30 are in clinical trials.

Together with a huge therapeutic potential, mAbs come with an enormous structural complexity. Opposed to small-molecule drugs, mAbs are large (ca. 150 kDa) and heterogeneous (as a result of the biosynthetic process and subsequent manufacturing and storage) making their analysis very challenging. Despite the fact that only a single molecule is cloned, hundreds of possible variants differing in post-translational modifications (N-glycosylation, asparagine deamidation, aspartate isomerization, methionine oxidation), amino acid

Koen Sandra, Jonathan Vandenbussche, Isabel Vandenheede, Bo Claerebout, Jeff Op

de Beeck, Paul Jacobs, Wim De Malsche, Gert Desmet, and Pat Sandra

Monoclonal antibodies are becoming a core aspect of the pharmaceutical industry. Together with

a huge therapeutic potential, these molecules come with a structural complexity that drives state-

of-the-art chromatography and mass spectrometry (MS) to its limits. This article discusses the

use of micro-pillar array columns in combination with mass spectrometry for peptide mapping of

monoclonal antibodies (mAbs) and antibody–drug conjugates (ADCs). Micro-pillar array columns are

produced by a lithographic etching process creating a perfectly ordered separation bed on a silicon

chip. As a result of the order existing in these columns, peak dispersion is minimized and highly effi-

cient peptide maps are generated, providing enormous structural detail. Using examples from the

author’s laboratory, the performance of these columns is illustrated.

Peptide Mapping of Monoclonal

Antibodies and Antibody–Drug

Conjugates Using Micro-Pillar

Array Columns Combined with

Mass Spectrometry

Page 21: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 21

Figure 1: Micro-pillar array column. From left to right: (left) Top view of two parallel 315-μm wide separation channels that have been interconnected

with proprietary flow distributor structures, (middle) SEM image showing a transverse section of a separation channel containing 5-μm diameter

cylindrical pillars, (right) HR-SEM image of the 300-nm porous-shell layer incorporated into a 5-μm diameter pillar.

sequence, and higher order structures may coexist, all contributing to the safety and efficacy of the product. Compared to naked mAbs, ADCs f ur t her add to t he complex it y

because the heterogeneity of the initial antibody is superimposed with the variability associated with the conjugation. Conjugation typically takes place on the amino groups of

lysine residues or on the sulphydryl g roups of i ntercha i n c y s te i ne residues. With 80 to 100 lysine and only eight interchain cysteine residues available, lysine conjugation yields a more heterogeneous mixture of species compared to cysteine conjugation.

Peptide mapping is particularly powerful for the detailed structural characterization of these products and has proven to be of enormous value in demonstrating comparability, for example, between mAb originator and biosimi lar. Character ist ics such a s a m i no ac id sequence and modif icat ions l ike N- and O-glycosylation, glycation, N- and C-terminal processing, deamidation (asparagine, glutamine), aspartate isomerization, succinimide, oxidation (methionine, tryptophan), clipping, sequence variants, cysteine variants (S-S bridges, thioether, free cysteine), and drug conjugation sites can readily be extracted out of the generated peptide map data and at low levels.

Look ing back to t he pept ide mapping of the f irst monoclonal antibodies in the late 1980s and early 1990s , one w i l l not ice a substantial leap forward in technical capabilities. Chromatography and mass spectrometry were at that time of modest performance compared to t he current s tate-of-t he-a r t technology. High performance liquid chromatography (HPLC) separations were performed on columns packed with 5–10 μm porous particles and pumps were operated at 400 bar.

Figure 2: LC–MS chromatograms of a Herceptin tryptic digest. (a) Total compound chromatogram.

(b) Compound chromatograms corresponding to the identified peptides presented in Table 1.

Page 22: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com22 Current Trends in Mass Spectrometry October 2018

Fast atom bombardment (FAB) was used to introduce peptides into low resolution mass spectrometers and peptide identity was further confirmed using Edman degradation following peak collection (16). Today, columns packed w it h sub-2-μ m porous and superficially porous particles operated at system pressures up to 1500 bar and electrospray ionization (ESI) can be used to introduce peptides into high resolution mass spectrometers equipped with a variety of fragmentation modes.

A more re c e nt a d d i t ion to t he ch romatog raphers toolbox

are micro-pi l lar array columns. T he or ig i n of t h is technolog y d ate s bac k to t he l a te 19 9 0 s when Regnier et a l . addressed t he problem of m i n iat u r i z i ng capi l lary electrochromatography (CEC) columns and introduced microfabricated supports as an a lternative for the conventional packed beds (17,18). The theoretical benef it (reduc t ion of t he va n Deemter A-term) of such supports was elucidated only a few years later by Knox (19). In the years to follow, Desmet et a l. conducted severa l quant itat ive studies on K nox’s

argumentation, taking a column f i l led with an array of pil lars as a representative example (20). In 2007, the first micromachined LC columns operated by pressure-driven l iquid f low, later termed micro-

pillar array columns, were reported (21). The inherent high permeability and low “on-column” dispersion obtained by the perfect order of the separation bed makes micro-pillar array-based chromatography unique and offers several benefits for chromatographers. The peak d i s p e r s i o n o r i g i n a t i n g f r o m heterogeneous f low paths in the s e p a r at ion b e d i s e l i m i n at e d (no A-term contr ibut ions) a nd therefore components remain more concentrated (sharp peaks) during separation. The freestanding nature of the pillars also leads to much lower backpressure allowing the use of very long columns. These properties result in excellent chromatographic performance with high resolution and high sensitivity.

This article describes the use of micro-pillar array columns in the characterization of mAbs and ADCs for the first time.

Materials and Methods

Materials

Water and acetonitrile were purchased f rom Biosolve . Tr i f luoroacet ic acid, dithiothreitol (DTT), and 2-iodoacetamide (IAA) were from Sigma-Aldrich. Tris-HCl pH 7.5 was purchased as a 1 M solution (Thermo Fisher Scientific). Porcine sequencing-grade modified trypsin was acquired from Promega and Rapigest from Waters. Herceptin and Kadcyla were obtained from Roche, Remicade from Johnson & Johnson, Adcetris from Seattle Genetics, and the candidate biosimilars from a local biotechnology company.

Sample Preparation

To a volume corresponding to 100 μg of protein, 105 μL of 0.1% Rapigest in 100 mM Tris-HCl pH 7.5 was added followed by the addition of 100 mM Tris-HCl pH 7.5 to a final volume of 192.5 μL. The sample was subsequently reduced

Figure 3: LC–UV 214 nm chromatograms corresponding to the replicate analysis (n = 4) of a

Herceptin tryptic digest.

Figure 4: LC–MS total compound chromatograms of a Herceptin originator and candidate

biosimilar tryptic digest.

Page 23: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 23

at 60 °C for 30 min by the addition of 5  mM DTT (2.5 μL of 400 mM DTT in 100 mM Tris-HCl) and alkylated at 37 °C for 1 h by adding 10 mM IAA (5 μL of 400 mM IAA in 100 mM Tris-HCl). Digestion proceeded for 16 h at 37 °C

using trypsin as protease added at an enzyme to substrate ratio of 1/25 (w/w). Lyophilized trypsin (20 μg) dissolved in 100 mM Tris-HCl (50 μL) was added in a volume of 10 μL giving rise to a final sample volume of 210 μL.

LC–MS

A n U lt i m ate 3 0 0 0 R SL Cna no system (Thermo Fisher Scientific) was used for LC–ultraviolet (UV)–MS measurements. Tryptic digests were analyzed on a 200 cm C18 μPAC column (PharmaFluidics) at 50 °C. Elution was performed with a linear gradient of (A) 0.1% TFA in H2O and (B) 0.1% TFA in 80:20 (v/v) acetonitrile–water, from 2% B to 70% B in 120 min. The flow rate was 500  nL/min. An injection program allowed the introduction of 100-nL sample in between two plugs of mobile phase A. Loop size was 1 μL and samples were kept at 10 °C in the autosampler tray while waiting for injection. UV detection was performed at 214 nm using a 3 nL detection cell.

High-resolution accurate mass measurements were obtained on a 6530 QTOF mass spectrometer (Agilent Technologies) equipped with a dual nano-ESI source operated in posit ive electrospray ionizat ion mode . T he m icro-pi l la r a r ray column was connected via a 30-μm internal diameter (i.d.), 280-μm outside diameter (o.d.) fused silica capillary to a PicoTip emitter (8-μm tip i.d. [New Objective]) via a true zero dead volume conductive union (UH-634 sta inless steel adapter from Idex). To connect the PicoTip emitter to the union, an F-330 blind f it t ing equipped with an M-125 perfluoroelastomer conductive ferrule was used (Idex). Capillary voltage was set at +2.4 kV, drying gas f low rate and temperature were set at 4 L/min and 300 °C, and fragmentor voltage at 175 V. The TOF was calibrated on a daily basis and subsequently operated at high accuracy (<5 ppm) without using reference masses. Data were col lected in centroid mode at a rate of 1 spectrum/s in the extended dynamic range mode (2 GHz) offering a resolution of 9000 at mass-to-charge ratio (m/z) 622.0296. MS/MS experiments were performed in the data-dependent acquisition (DDA) mode. One survey MS measurement was complemented with three data-dependent MS/MS

Figure 5: Extracted ion chromatograms of selected modified peptides measured in Herceptin

originator and candidate biosimilar. Shown are asparagine deamidation, lysine truncation, and

methionine oxidation, which are elevated in the biosimilar compared to the originator. The

location of the modifications is highlighted in the antibody structure.

Page 24: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com24 Current Trends in Mass Spectrometry October 2018

measurements. Double, triple, and quadruple charged precursor ions with the Averagine isotope model were selected based on abundance. After being fragmented twice, a particular m/z value was excluded for 30 s. Selecting the same m/z value twice increases the chance of measuring a particular precursor at its maximum intensity while an exclusion time of 30 s allows MS/MS information on chromatographically resolved isomers to be obtained. The quadrupole was operated at medium resolution and the collision energy varied based on the following equation:

(3.6 * m/z)/100 – 4 [1]

A l l-ions MS/MS ex per i ments were per for med at a col l i s ion energ y of 20 eV. LC data were acquired in Chromeleon (Thermo Fisher Scientif ic) and MS data in MassHunter (Agilent Technologies). Data analysis was performed in MassHunter Qualitative Analysis with BioConfirm add-on.

Results and Discussion

Characteristics of

Micro-Pillar Array Columns

The separation beds of micro-pillar array columns are fabricated by carefully etching the interstitial volumes out of a silicon substrate following lithographic definition of an array of pillars. This creates a stationary phase support structure that is organized in a reproducible and ordered pattern. Concatenation of several of these channels allows long column lengths to be fabricated on a small footprint (22). The most important characteristics of the micro-pillar array separation bed design are: pillar diameter, 5 μm; inter pillar distance, 2.5 μm; pillar length or bed depth, 18 μm; external porosity (Vinterstitial/Vtotal), 59%; bed channel width, 315 μm; and bed length, 200 cm. To increase the retentive surface, the pillars are rendered superficially porous with a typical porous shell thickness of 300 nm and pore sizes in the nanometer range. The porous surface has been uniformly modified with octadecyl alkyl

Figure 6: LC–UV 214 nm peptide map of (a) a Remicade originator and (b) a candidate biosimilar

tryptic digest. Unzoomed and zoomed views.

Figure 7: LC–MS total compound chromatograms of (a) a Remicade originator and (b) a candidate

biosimilar tryptic digest. MS/MS spectra associated with peaks 4 and 5 are presented in Figure 8.

Page 25: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 25

chains to create a hydrophobic stationary phase suited for reversed-phase LC separations. Figure  1 shows some relevant characteristics of the micro-pillar array column.

Because of the high permeability, the 200-cm column used in this study can be operated at moderate LC pump pressures (50 to 300 bar) over a wide range of flow rates (100–1000 nL/min). Van Deemter measurements with heptyl-phenyl ketone demonstrated that a total of 400,000 theoretical plates could be generated at the optimal li nea r solvent veloc it y, corresponding to a f low rate of 200–250 nL/min and generat ing a column backpressure of on ly 70  bar. By increasing the f low rate

up to 750  nL/min, eff iciencies up to 200,000 theoretical plates could be ach ie ved w it h i n 30 m i n at operating pressures well below 250 bar (mobile phase 50% acetonitrile with 0.1% [v/v] FA).

Peptide Mapping of

Herceptin Originator and

a Candidate Biosimilar

Herceptin (scientific international non-proprietary names [INN] name tras-tuzumab) is a humanized IgG1 mAb recombinantly produced in Chinese hamster ovary (CHO) cells and used in the treatment of human epidermal growth factor receptor 2 (HER2) posi-tive breast cancer. Herceptin is open to the European market and evolves out of

patent in the US in 2019 (5). Given its market potential (global sales of $6.6 bil-lion in 2015), dozens of companies are actively developing a Herceptin biosim-ilar. In developing these products, simi-larity to the originator has to be demon-strated and for that, an enormous weight is placed on analytics; both the biosimilar and originator need to be characterized and compared in great detail. Peptide mapping is a powerful method to charac-terize and compare mAbs in great detail.

When digesting Herceptin with the enzyme trypsin, which cleaves the protein next to arginine and lysine residues, 62 identity peptides are formed. Taking into account post-translational modifications and incomplete and aspecific cleavages tak ing place, over 100 peptides w it h va r y i ng phy sicochem ic a l properties present in a wide dynamic concent rat ion ra nge a re to be expected. This is quite a complex sample demanding the best in terms of chromatographic resolution.

Figure 2(a) shows the LC–MS tota l compound chromatogra m of a Herceptin tryptic digest on a m ic ro -pi l l a r a r r ay c o lu m n . The compound chromatograms corresponding to the identi f ied peptides presented in Table  1 are shown in Figure 2(b). Approximately 95% of the sequence is covered by this peptide map and post-translational modifications such as glycosylation, asparagine deamidation, aspartate i s o m e r i z a t i o n , m e t h i o n i n e oxidation, N-terminal cyclization (pyroglutamate), and C-terminal lysine truncation, amongst others, are revealed. Peptides that are not detected are typically small and their signal might be suppressed in the column f low through. The UV 214 nm chromatograms corresponding to replicate analyses (n = 4) of the Herceptin tryptic digest are demonstrated in Figure 3. These measurements are highly precise making this technology attractive to compare different mAb production batches a nd to compa re m Ab originator products to biosimilars.

T h e L C – M S p e p t i d e m a p s of a Hercept in or ig inator a nd

Figure 8: LC–MS/MS spectra of peak 4 and 5 (Figure 7) acquired on the fly in data-dependent

MS/MS mode confirming the threonine to serine substitution in the variable part of the heavy

chain.

Page 26: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com26 Current Trends in Mass Spectrometry October 2018

Table I: Identified peptides in the Herceptin tryptic digest analyzed by LC–MS

RT (min) Mass Sequence Seq Loc. Theor. Mass ∆m (ppm) Modifications

61.29 1880.99676 EVQLVESGGGLVQPGGSLR Hc(001-019) 1880.9956 0.62

66.83 1862.98605 EVQLVESGGGLVQPGGSLR Hc(001-019) 1862.98503 0.55 Pyroglutamate

51.04 1166.57794 LSCAASGFNIK Hc(020-030) 1166.5754 2.18 Cys-Alkylation

71.21 2237.10621 LSCAASGFNIKDTYIHWVR Hc(020-038) 2237.10516 0.47 Cys-Alkylation

55.35 1088.54237 DTYIHWVR Hc(031-038) 1088.54033 1.87

52.54 829.44557 GLEWVAR Hc(044-050) 829.44464 1.12

39.97 1084.52052 IYPTNGYTR Hc(051-059) 1084.51893 1.47 Deamidation

40.39 1083.53637 IYPTNGYTR Hc(051-059) 1083.53491 1.35

41.46 1084.52241 IYPTNGYTR Hc(051-059) 1084.51893 3.21 Deamidation

28.81 681.33403 YADSVK Hc(060-065) 681.33336 0.99

44.14 968.48309 FTISADTSK Hc(068-076) 968.48148 1.66

43.48 1366.66793 NTAYLQMNSLR Hc(077-087) 1366.66634 1.17 Met-Alkylation

53.78 1310.63608 NTAYLQMNSLR Hc(077-087) 1310.62889 5.49 Deamidation

54.96 1309.6462 NTAYLQMNSLR Hc(077-087) 1309.64487 1.02

40.75 1333.56246 AEDTAVYYCSR Hc(088-098) 1333.56087 1.19 Cys-Alkylation

70.51 2840.27436WGGDGFYAMDYWGQ

GTLVTVSSASTKHc(099-124) 2840.2752 -0.3 Met-Alkylation

79.37 2783.25248WGGDGFYAMDYWGQ

GTLVTVSSASTKHc(099-124) 2783.25374 -0.45

58.45 1185.64103 GPSVFPLAPSSK Hc(125-136) 1185.63938 1.4

54.84 1320.67286 STSGGTAALGCLVK Hc(137-150) 1320.67075 1.59 Cys-Alkylation

92.81 6712.27146

DYFPEPVTVSWNSGALTSG

VHTFPAVLQSSGLYSLSSVVTVP

SSSLGTQTYICNVNHKPSNTK

Hc(151-213) 6712.3072 -5.32 Cys-Alkylation

24.21 599.36222 KVEPK Hc(217-221) 599.36426 -3.41

23.25 471.26917 VEPK Hc(218-221) 471.2693 -0.27

21.56 508.19936 SCDK Hc(222-225) 508.19515 8.28 Cys-Alkylation

82.24 2618.30392 THTCPPCPAPELLGGPSVFLFPPK Hc(226-249) 2618.30254 0.53 Cys-Alkylation

80.29 2843.4514 THTCPPCPAPELLGGPSVFLFPPKPK Hc(226-251) 2843.45027 0.4 Cys-Alkylation

41.18 891.44858 DTLMISR Hc(252-258) 891.44841 0.19 Met-Alkylation

41.64 850.42241 DTLMISR Hc(252-258) 850.42186 0.65 Oxidation

45.43 834.42764 DTLMISR Hc(252-258) 834.42694 0.84

64.07 2138.02125 TPEVTCVVVDVSHEDPEVK Hc(259-277) 2138.02016 0.51 Cys-Alkylation

61.55 1676.79346 FNWYVDGVEVHNAK Hc(278-291) 1676.79471 -0.75 Isomerization

62.47 1676.7955 FNWYVDGVEVHNAK Hc(278-291) 1676.79471 0.47

32.86 2957.13923 EEQYNSTYR Hc(296-304) 2957.14427 -1.7 G2F

32.92 2795.08868 EEQYNSTYR Hc(296-304) 2795.09144 -0.99 G1F

33.05 2633.03635 EEQYNSTYR Hc(296-304) 2633.03862 -0.86 G0F

81.52 1807.98677 VVSVLTVLHQDWLNGK Hc(305-320) 1807.98324 1.95 Deamidation

82.11 1807.0017 VVSVLTVLHQDWLNGK Hc(305-320) 1806.99922 1.37

Page 27: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 27

Table I: (Continued) Identified peptides in the Herceptin tryptic digest analyzed by LC–MS

82.92 1807.98553 VVSVLTVLHQDWLNGK Hc(305-320) 1807.98324 1.27 Deamidation

84.33 1789.97519 VVSVLTVLHQDWLNGK Hc(305-320) 1789.97267 1.4 Succinimide

46.50 837.49753 ALPAPIEK Hc(330-337) 837.49601 1.82

23.71 447.26932 TISK Hc(338-341) 447.2693 0.04

50.08 1285.66847 EPQVYTLPPSR Hc(348-358) 1285.66665 1.41

24.31 636.2778 EEMTK Hc(359-363) 636.27888 -1.69

58.87 1160.62392 NQVSLTCLVK Hc(364-373) 1160.62235 1.36 Cys-Alkylation

68.04 2544.11037 GFYPSDIAVEWESNGQPENNYK Hc(374-395) 2544.10812 0.88 Deamidation

68.51 2543.12268 GFYPSDIAVEWESNGQPENNYK Hc(374-395) 2543.12411 -0.56

69.70 2526.09392 GFYPSDIAVEWESNGQPENNYK Hc(374-395) 2526.09756 -1.44 Succinimide

70.26 1872.90824 TTPPVLDSDGSFFLYSK Hc(396-412) 1872.91455 -3.37 Isomerization

71.48 1872.91517 TTPPVLDSDGSFFLYSK Hc(396-412) 1872.91455 0.33

32.14 574.33213 LTVDK Hc(413-417) 574.33263 -0.86

66.07 2800.25791 WQQGNVFSCSVMHEALHNHYTQK Hc(420-442) 2800.25984 -0.69 Cys-Alkylation

44.81 659.34888 SLSLSPG Hc(443-449) 659.34901 -0.19 Lys-Truncation

43.20 787.44611 SLSLSPGK Hc(443-450) 787.44397 2.72

46.55 1934.90169 DIQMTQSPSSLSASVGDR Lc(001-018) 1934.90038 0.68 Met-Alkylation

54.09 1877.87963 DIQMTQSPSSLSASVGDR Lc(001-018) 1877.87891 0.38

35.39 748.39074 VTITCR Lc(019-024) 748.39016 0.78 Cys-Alkylation

53.62 1707.82255 ASQDVNTAVAWYQQK Lc(025-039) 1707.82165 0.53

54.73 1708.80311 ASQDVNTAVAWYQQK Lc(025-039) 1708.80567 -1.5 Deamidation

51.50 1990.97492 ASQDVNTAVAWYQQKPGK Lc(025-042) 1990.97486 0.03 Deamidation

53.37 1989.99336 ASQDVNTAVAWYQQKPGK Lc(025-042) 1989.99084 1.27

54.43 1990.97614 ASQDVNTAVAWYQQKPGK Lc(025-042) 1990.97486 0.64 Deamidation

83.35 4040.11375ASQDVNTAVAWYQQKPG

KAPKLLIYSASFLYSGVPSRLc(025-061) 4040.116 -0.55

76.25 1771.95123 LLIYSASFLYSGVPSR Lc(046-061) 1771.95088 0.2

81.29 4186.90432SGTDFTLTISSLQPEDFAT

YYCQQHYTTPPTFGQGTKLc(067-103) 4186.91062 -1.5 Cys-Alkylation

30.68 487.3006 VEIK Lc(104-107) 487.3006 -0.01

72.96 2101.12277 RTVAAPSVFIFPPSDEQLK Lc(108-126) 2101.1208 0.94

74.60 1945.02035 TVAAPSVFIFPPSDEQLK Lc(109-126) 1945.01968 0.34

80.21 1796.88935 SGTASVVCLLNNFYPR Lc(127-142) 1796.88796 0.77 Cys-Alkylation

21.45 346.18747 EAK Lc(143-145) 346.18524 6.46

34.98 559.31157 VQWK Lc(146-149) 559.31183 -0.48

39.99 2134.96295 VDNALQSGNSQESVTEQDSK Lc(150-169) 2134.96146 0.7

59.63 1501.75181 DSTYSLSSTLTLSK Lc(170-183) 1501.75118 0.42

24.33 624.27451 ADYEK Lc(184-188) 624.27551 -1.6

50.86 1874.92219 VYACEVTHQGLSSPVTK Lc(191-207) 1874.91965 1.35 Cys-Alkylation

26.74 522.25451 SFNR Lc(208-211) 522.25505 -1.03

Page 28: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com28 Current Trends in Mass Spectrometry October 2018

candidate biosimilar are shown in Figure  4. While both peptide maps are highly comparable, differences in post-translational modifications are detected. This is illustrated in the extracted ion chromatograms presented in Figure 5.

Peptide Mapping of

Remicade Originator

and Candidate Biosimilar

Remicade (scient i f ic INN name inf liximab) is a chimeric IgG1 mAb on the market since 1998 targeting t u mou r ne c ro s i s f a c tor a lph a (TNF-α) and consequent ly used in the treatment of autoimmune diseases. Remicade reached global sales of $8.4 billion in 2015. Several Remicade biosimilars have already been approved bot h in Europe and the US and many more are in development (8).

Figures 6 and 7 show the UV and MS total compound chromatograms of a Remicade orig inator and a candidate biosimilar tryptic digest, respectively. Chromatograms are ver y simi lar but some st r i k ing differences are noted (peaks 1–5), wh ich c a n be ex pla i ned upon consulting the corresponding MS data. Peaks 1 and 2 in the originator chromatogram, corresponding to C-terminal heav y chain peptides SLSLSPGK and SLSLSPG respectively, are replaced by peak 3, corresponding to SLSLSPGI in the biosimilar. The origin of the two peaks SLSLSPG and SLSLSPGK in the originator mAb can be explained by the knowledge that heavy chains are historically cloned with a C-termina l lysine but during cell culture production, host cel l carbox y pept idases act on the antibody result ing in the par t ia l remova l of t hese lysine residues . A m issense mutat ion (Ly s" I l e) i n t he C HO c lone producing the candidate biosimilar e x pl a i n s p e a k 3 (SL SL SP GI). Peaks 4 and 5 corresponding to, respectively, heavy chain peptides NYYGSSYDY WGQGTTLTVSSASTK in the candidate biosimilar and NYYGSTYDY WGQGTTLTVSSASTK i n t he or ig i nator aga i n resu lt

Figure 9: LC–UV 214 nm peptide map of Herceptin and Kadcyla.

Figure 10: LC–MS peptide map of Herceptin and Kadcyla.

Figure 11: All-ions MS/MS chromatograms of the Herceptin and Kadcyla tryptic digest. The ion at

m/z 547.2206 was extracted at high mass accuracy.

Page 29: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 29

from a point mutation (Thr" Ser) in the biosimilar CHO clone. The corresponding MS/MS spectra are shown in Figure 8.

According to US and European regulatory authorit ies , identica l primary sequence is primordial to similarity thereby ruling out this candidate biosimilar from further development.

Peptide Mapping

of the Antibody–Drug

Conjugate Kadcyla

The antibody–drug conjugate Kadcyla (ado-trastuzumab emtansine) has been used in the treatment of HER2 positive breast cancer since 2013. It combines the anti-HER2 antibody trastuzumab (Hercept in) with the cy totox ic microtubule-inhibiting maytansine

derivative DM1 conjugated to lysine residues via a non-reducible thioether linker. With a drug distribution of 0 to 8, a drug-to-antibody ratio (DAR) of 3.5, and various lysine residues available for conjugation, thousands of species can be generated. To obtain insight in the drug conjugation sites, peptide mapping is the gold standard technology.

Figures 9 and 10 show, respectively, the LC–UV and MS total compound chromatograms of a Herceptin and Kadcyla tryptic digest. Since Herceptin and Kadcyla have the same amino acid sequence, the majority of the peptide map is identical. Differentiating peaks, corresponding to the DM1 conjugated peptides, are nevertheless observed and are located in the late eluting part of the chromatogram. Indeed, the conjugation of DM1 makes the peptides more hydrophobic explaining the elution behavior. Upon collision-induced dissociation (CID), DM1 conjugated peptides give rise to specific fragments originating from the cytotoxic drug, for example, at m/z 547.2206. This ion can be used to selectively recognize DM1 conjugated peptides in the LC–MS chromatogram. For that, one can operate the QTOF-MS system in the a l l-ions MS/MS mode, which means that the quadrupole is operated in the RF only mode, thereby transferring all peptides to the collision cell where CID takes place. As illustrated in Figure 11, when extracting from the data the specific fragment ion at m/z 547.2206 at high mass accuracy, all conjugated peptides are revealed in the chromatogram of Kadcyla compared to Herceptin. The latter chromatogram is virtually empty, il lustrating the selectivity that is offered by this all-ions MS/MS functionality.

Figure 12 shows the extracted ion chromatograms associated with a selection of identified conjugated peptides. A striking observation is the appearance of isomeric DM1 conjugated peptides, which can be explained by the existence of two stereochemical configurations (diastereomers) of the antibody–drug linkage through maleimide.

Figure 12: Extracted ion chromatograms of selected peptides showing the appearance of isomeric

conjugated peptides. The location of the conjugation site in the mAb structure is shown as well.

Figure 13: LC–MS peptide map of Adcetris. (a) Total compound chromatogram and (b) all-ions

MS/MS chromatogram. The ion at m/z 718.5113 was extracted at high mass accuracy. The location

of the conjugation site in the mAb structure is shown as well.

Page 30: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com30 Current Trends in Mass Spectrometry October 2018

Peptide Mapping of the

Antibody–Drug Conjugate Adcetris

A simi lar st rateg y was appl ied to revea l t he conjugat ion sites o n A d c e t r i s ( b r e n t u x i m a b -vedotin). Approved in 2011, this ADC is directed to CD30, a major ma rker of Hodg k in ly mphoma a nd s y s tem ic a napla s t ic l a rge cel l lymphoma (ALCL). Adcetris combines the antibody brentuximab w i t h t h e a n t i m i t o t i c d r u g monomethylauristatin E (MMAE) conjugated to interchain cysteine residues via a cathepsin cleavable valine-citrulline linker. With only eight residues (four on each half antibody) available for conjugation, Adcetris is much simpler compared to Kadcyla.

Figure 13 shows the LC–MS total compound chromatogram and the a l l-ions MS/MS chromatog ra m of a n Adcet r i s t r y pt ic d iges t , respectively. In analogy to DM1 c o n j u g a t e d p e p t i d e s , M M A E conjugated peptides a lso contain specific fragment ions originating f rom t he c y totox ic mole c u le , t hat is , at m/z 718.5113. W hen ex t rac t i ng t he lat ter ion f rom t he a l l - ions MS/MS d at a , t he conjugated peptides are revealed. Three intense peaks explaining the full conjugation of MMAE at the four interchain cysteine residues in light chain peptide GEC (peak 2), heav y chain pept ides SCDK ( p e a k 1) , a n d T H T C P P C PA PE L L G G P S V F L F PPK PK (p e a k 3) a re obser ved . Of pa r t icu la r interest is the detection of isomeric p a r t i a l l y c o n j u g a t e d h e a v y c h a i n p e p t i d e T H T C PP C PA P ELLGGPSVFLFPPK PK (pea ks a and b), in which only one of the two cysteine residues is conjugated.

ConclusionsIn t he 21st centur y, numerous novel developments were made in LC column technolog y and the best known are columns packed with sub-2-μm porous part icles or sub-3-μm superficially particles. Micro-pi l lar array columns are another novel development offering

highly ef f icient separat ions. In this article, the use of the micro-pillar array column in mAb and ADC peptide mapping has been i l lustrated. In combination with high resolution mass spectrometry, high sequence coverage is obtained and post-translational modifications such as glycosylation, deamidation, isomerization, oxidation, N-terminal cyclization, and C-terminal lysine truncation can be elucidated. Its use in assessing comparability between an originator and biosimilar mAb has furthermore been demonstrated. The resolving power offered by a micro-pillar array column allows an in-depth study of ADC conjugation s i t e s a n d a c c o m m o d a t e s t h e separation of isomeric conjugated peptides. In combination with all-ions MS/MS, conjugated peptides can selectively be recognized to assist data interpretation.

References(1) G. Köhler and C. Milstein, Nature

256, 495–497 (1975).

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

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

Haan, Curr. Opin. Pharmacol. 8,

620–626 (2012).

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

der Walle, Int. J. Pharm. 440, 83–98

(2013).

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

Levine, mAbs 7, 9–14 (2015).

(5) G. Walsh, Nat. Biotechnol. 32, 992–

1000 (2014).

(6) H. Kaplon and J.M. Reichert, mAbs

10, 183–203 (2018).

(7) K. Sandra, I. Vandenheede, E. Du-

mont, and P. Sandra, LCGC Europe

28(s10), 16–23 (2015).

(8) A. Beck and J.M. Reichert, mAbs 5,

621–623 (2013).

(9) S. Panowski, S. Bhakta, H. Raab, P.

Polakis, and J.R. Junutula, mAbs 6,

34–45 (2014).

(10) A. Wakankar, Y. Chen, Y. Gokarn,

and F.S. Jacobson, mAbs 3, 161–172

(2011).

(11) A. Beck and J.M. Reichert, mAbs 6,

15–17 (2014).

(12) A. Beck, L. Goetsch, C. Dumontet,

and N. Corvaia, Nat. Rev. Drug Dis-

cov. 16, 315–337 (2017).

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

Ayoub, A. Van Dorsselaer, and S.

Sanglier-Cianférani, Anal. Chem. 85,

715–736 (2013).

(14) K. Sandra, I. Vandenheede, and

P. Sandra, J. Chromatogr. A 1335,

81–103 (2014).

(15) S. Fekete, D. Guillarme, P. Sandra,

and K. Sandra, Anal. Chem. 88, 480–

507 (2016).

(16) D.J. Kroon, A. Baldwin-Ferro, and

P. Lalan, Pharm. Res. 9, 1386–1393

(1992).

(17) B. He, N. Tait, and F. Regnier, Anal.

Chem. 70, 3790–3797 (1998).

(18) F. Regnier, J. High Resol. Chromatogr.

23, 19–26 (2000).

(19) J.H. Knox, J. Chromatogr. A 960, 7–18

(2002).

(20) P. Gzil, N. Vervoort, G. Baron, and G.

Desmet, Anal. Chem. 75, 6244–6250

(2003).

(21) W. De Malsche, H. Gardeniers, and G.

Desmet, Anal. Chem. 80, 5391–5400

(2008).

(22) W. De Malsche, J. Op de Beeck, S.

De Bruyne, H. Gardeniers, and G.

Desmet, Anal. Chem. 84, 1214–1219

(2012).

Koen Sandra is the editor of “Biophar-

maceutical Perspectives”. He is the Scien-

tific Director at the Research Institute for

Chromatography (RIC, Kortrijk, Belgium)

and the R&D Director at anaRIC biologics

(Ghent, Belgium). Jonathan Vanden-

bussche is an LC–MS Technician at RIC.

Isabel Vandenheede is a Project Man-

ager Biologics at RIC. Bo Claerebout is

R&D Engineer at PharmaFluidics, in Ghent,

Belgium. Jeff Op de Beeck is an Appli-

cation Development Manager at Pharma-

Fluidics. Paul Jacobs is the Chief Oper-

ating Officer (COO) at and co-founder of

PharmaFluidics. Wim De Malsche is an

Associate Professor at the Department

of Chemical Engineering at the Vrije Uni-

versiteit Brussel, in Belgium, and the co-

founder of PharmaFluidics. Gert Desmet

is a Full Professor in Chemical Engineer-

ing at the Vrije Universiteit Brussel, Bel-

gium, and a co-founder of PharmaFluidics.

Pat Sandra is the President of RIC and

Emeritus Professor at Ghent University.

Page 31: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

chromatographyonl ine .com October 2018 Current Trends in Mass Spectrometry 31

PRODUCTS & RESOURCES Multipurpose samplerThe Gerstel MultiPurpose

Sampler (MPS) with filtration

and DNPH elution is designed

to enable automated cleanup

of up to 240 samples or extracts

using individual standard filters.

According to the company,

up to 72 DNPH cartridges

per tray can be eluted to

determine formaldehyde

and lower aldehydes sampled from air.

Gerstel GmbH & Co. KG, Linthicum, MD; www.gerstel.com

Mass spectrometerPerkinElmer’s QSight Triple Quad LC–MS/

MS mass spectrometer is designed for appli-

cations such as food safety, environmental

testing, and industrial research. According

to the company, the mass spectrometer

provides high sensitivity and uptime, and

offers remote support capabilities.

PerkinElmer, Waltham, MA.www.perkinelmer.com/qsight

UHPLC guard cartridgesRestek’s EXP UHPLC

guard column cartridges

are made specifically for

the company’s Raptor

1.8-μm columns. Accord-

ing to the company, the

guards provide protection from particulates and matrix contamina-

tion, extend the lifetime of Raptor columns, and can withstand the

same UHPLC pressures as their analytical column counterparts.

Restek Corporation, Bellefonte, PA.www.restek.com/raptor

HILIC columnsHILIC pac VC-50 columns

from Shodex are designed

for hydrophilic interaction

chromatography analysis of

monoamine type neurotrans-

mitters and oral anti-diabetic

drugs. According to the

company, the columns are

packed in PEEK to decrease

interactions between com-

pounds and housing sides, and are suitable for LC–MS analysis.

Shodex/Showa Denko America, Inc., New York, NY.www.shodexhplc.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

Electron multipliersDiscrete dynode electron multi-

pliers from Photonis are designed

as plug-and-play replacements

for ICP-MS systems. The electron

multipliers reportedly provide a

mass range to 238

U and gating of

less than 6 ns. According to the

company, nearly 200 electron

multiplier types are available for

current and legacy instruments.

Photonis USA, Sturbridge, MA.www.photonis.com

PyrolyzerThe Pyprobe 6000 pyrolyzer

from CDS Analytical is designed

with a pyrolysis sample tube

that is gravity-dispensed

into the system’s Drop-In-

Sample- Chamber (DISC).

According to the company,

the RSD results are within 2%

due to the positioning of the

sample in the pyrolysis coil.

CDS Analytical LLC, Oxford, PA.www.cdsanalytical.com

SEC–MS application noteAn application note from Tosoh

Bioscience titled “Size-Exclusion

Chromatography/ Mass Spectrometry

(SEC/MS) Analysis of a Bispecific

Antibody” reportedly describes a

bispecific T cell engager (BiTE) consisting

of two linked single-chain variable

fragments that was analyzed by SEC–MS

using a TSKgel UP-SW3000, 2-μm column.

Tosoh Bioscience LLC, King of Prussia, PA.www.tosohbioscience.com

Page 32: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

32 PHARMACEUTICAL/DRUG DISCOVERY ADVERTISEMENT

Ineffi ciently designed fl ow splitter technology causes signifi cant

problems that are often overlooked, underestimated, or tolerated simply

due to a lack of product options.

Mott Corporation has developed a breakthrough fl ow splitter tech-

nology for use in HPLC and UHPLC analysis. The new PerfectPeak®

Flow Splitters utilize proprietary porous metal fl ow restrictor technology

as a means of creating a differential fl ow while achieving the appropriate

post-column split ratio. These fl ow splitters are designed for +/-10%

split accuracy with only ~5 μL post-column dwell volumes, signifi cantly

minimizing peak broadening. Additionally, PerfectPeak Flow Splitters

are easier to install, and more convenient to use with their interchange-

able cartridges and fi nger-tight fi ttings.

We subjected new PerfectPeak Flow Splitters to a repeat injection test

to determine the variability between injections.

Repeat Injection Test

The PerfectPeak Flow Splitter was tested for injection-to-injection

reproducibility with a small-molecule analysis method. The method

used was modifi ed from an Agilent application note on the United

States Pharmacopeia (USP) method for the evaluation of caffeine and

acetaminophen (https://www.agilent.com/cs/library/applications/5991-

5920EN.pdf) in which 1 mg/mL of caffeine and acetaminophen was

prepared in deionized water. The method was run under the conditions

shown in Table I.

Results

Peak retention was monitored upon successive injections for fl ow

stability. It was found that upon four consecutive injections, the caffeine

and acetaminophen peaks had a 0.11% relative standard deviation

(RSD). The USP requirements of RSD for replicated injections is no

more than 2.0% variation. The results of the testing can be found in

Figure 2 where it can be seen that peak

resolution was 10.3 on average. Figure

3 highlights the reduction in peak

broadening due to the lower internal

volume of the Mott PerfectPeak Flow

Splitter compared to generic “T” style

New Post-Column Flow Splitters Improve Accuracy and RepeatabilityChristopher J. Martino, James Steele, and Vincent Palumbo, Mott Corporation

Mott Corporation

84 Spring Lane, Farmington, CT 06032

tel. (860) 747-6333

Website: www.mottcorp.comFigure 1: Schematic diagram of the low-pressure gradient experimental test system.

Pump G1312B

Auto Injector G1329B

Column Comp. G1316A

Detector VWD G1314B

Figure 2: Data overlay of the caffeine and acetaminophen analysis from the UV detector at 275 nm showing 0.11% relative standard deviation an average peak resolution of 10.3 after four consecutive injections.

Figure 3: Reductions in peak broadening are observed by converting a method setup from a generic “T” with cut tubing to the Mott PerfectPeak Flow Splitter.

splitting that uses cut

tubing.

Mott’s new Perfect-

Peak Flow Splitters offer

convenience and high

accuracy for 1:1, 2:1,

3:1, 5:1, and 10:1 split

ratios with the use of

interchangeable car-

tridges and fi nger-tight

fi ttings. Each fl ow

splitter is packaged in a small, lightweight tee body that will not

kink instrument lines when connected. The footprint of the device

is under 2 in. in any dimension and rated for up to 15,000 PSIG.

More case studies on improving

peak dispersion and time savings

on system setup can be found at

www.mottcorp.com/splitter.

Table I: Repeat injection method

configuration

ColumnZorbax Eclipse Plus C18

4.6 × 100 mm, 3.5 μm

Mobile PhaseWater:Methanol:Acetic

Acid (69:28:3)

Temperature 45 °C

Flow Rate 1.0 mL/min

Injection Volume 5 μL

Detection UV, 275 nm

Page 33: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

Medical/Biological 33ADVERTISEMENT

The analysis of synthetic cannabinoids and their metabolites can be a

diffi cult task, one that is further complicated by the ever-growing list of

synthetic cannabinoids produced by illicit drugmakers. As shown here,

the retention and selectivity of the Raptor Biphenyl column allows new

drugs to be added to an existing method, providing labs with an important

vehicle for improving effi ciency and productivity.

When developing methods for synthetic cannabinoids and their

metabolites, optimization of analysis time, resolution of isobaric

compounds, method robustness, and the ability to add emerging

compounds are of ultimate importance as they infl uence method

effectiveness and longevity. Because the Raptor Biphenyl column

combines the speed of superfi cially porous particles (SPP) with the

resolution of highly selective USLC technology, we used it to develop

a simple, dilute-and-shoot method for synthetic cannabinoids and

their metabolites in urine. Our original work produced a fast method

that separated all target compounds in less than 7 min.

Here, we expand on our original method and show proof of

concept for how new drugs can be added, while still maintaining

complete resolution of JWH-018 and JWH-073 isobars, as well as

good separation of all target analytes from matrix interferences in

urine. In the ever-changing landscape of illegal drugs, the ability

to add emerging drugs to existing methods allows for much more

effective use of laboratory resources as analysts and instruments

can focus on routine sample analysis rather than new method

development.

Experimental Conditions

For the existing method, samples were prepared at 5  ng/mL in

human urine and diluted 3x in a 0.2 μm PVDF Thomson SINGLE

StEP fi lter vial with water:methanol (50:50) prior to analysis. The

additional compounds were prepared at 50 ng/mL in urine and also

were diluted 3x and fi ltered using the same technique. Analyses

were performed on a Waters ACQUITY UPLC I-Class equipped

with a Xevo TQ-S and a Shimadzu Nexera UHPLC equipped with a

SCIEX API 4500 MS/MS. Instrument conditions were as follows, and

analyte transitions for the original analytes and the new drugs are

provided in Tables I and II, respectively.

Adding New Synthetic Cannabinoids to an Existing LC–MS/MS Method

Sharon Lupo and Frances Carroll, Restek Corporation

Time (min)0.00

m/z 328�155

m/z 344�155

m/z 358�155

0.50 1.00 1.50 2.00 2.50 3.00 3.50 4.00 4.50 5.00 5.50

Time (min)

2.00 2.50 3.00 3.50 4.00 4.50 5.00 5.50

2.00 2.50 3.00 3.50 4.00 4.50 5.00 5.50

2.00 2.50 3.00 3.50 4.00 4.50 5.00 5.50

Figure 1: Original method developed for the combined analysis of synthetic cannabinoids and metabolites in diluted human urine

Page 34: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

34 Medical/Biological ADVERTISEMENT

Table I: Figure 1 analyte identification, retention times, and ion transitions

Figure 1 Peak Identifications tR (min) Precursor Ion Quantifier Product Ion Qualifier Product Ion

1. Pravadoline 2.15 379.29 135.04 114.16

2. AM2233 2.44 459.25 112.2 98.15

3. JWH-200-d5 2.47 390.34 155.07 NA

4. JWH-200 2.48 385.28 155.07 114.16

5. WIN 55, 212 3.34 427.29 155.07 127.14

6. JWH-073 N-butanoic acid 3.39 358.27 155.08 127.11

7. JWH-073 4-hydroxybutyl 3.4 344.24 155.09 127.09

8. JWH-018 N-pentanoic acid 3.49 372.18 155.08 127.14

9. JWH-018 5-hydroxypentyl-d5 3.54 363.5 155.08 NA

10. JWH-018 5-hydroxypentyl 3.55 358.27 155.08 127.11

11. JWH-073 6-hydroxyindole 3.77 344.24 155.09 127.09

12. JWH-073 5-hydroxyindole-d7 3.81 351.21 155.07 NA

13. JWH-073 5-hydroxyindole 3.83 344.24 155.09 127.09

14. JWH-073 7-hydroxyindole 3.92 344.24 155.09 127.09

15. JWH-018 6-hydroxyindole 3.94 358.27 155.08 127.11

16. JWH-018 5-hydroxyindole 3.99 358.27 155.08 127.11

17. JWH-018 7-hydroxyindole 4.08 358.27 155.08 127.11

18. RCS-4 4.15 322.27 135.12 77.09

19. XLR-11 4.21 330.25 232.17 125.1

20. JWH-015-d7 4.27 335.28 155.07 NA

21. JWH-250 4.27 336.28 121.12 91.07

22. JWH-015 4.29 328.26 155.07 127.13

23. AM2201 4.30 360.26 155.07 127.14

24. JWH-203 4.39 340.23 188.18 125.09

25. JWH-073 4.42 328.26 155.07 127.13

26. UR-144 4.44 312.32 214.17 125.1

27. JWH-073 4-hydroxyindole 4.53 344.24 155.09 127.09

28. JWH-018-d9 4.55 351.34 155.07 NA

29. JWH-018 4.57 342.27 155.08 127.11

30. JWH-081 4.64 372.28 185.12 157.09

31. JWH-018 4-hydroxyindole 4.66 358.27 155.08 127.11

32. JWH-122 4.69 356.29 169.12 141.11

33. JWH-019 4.70 356.29 155.07 127.1

34. JWH-210 4.84 370.31 183.12 153.26

Table II: Figure 2 analyte identification, retention times, and ion transitions

Figure 2 Peak Identifications tR (min) Precursor Ion Quantifier Product Ion Qualifier Product Ion

1. AB-FUBINACA 3.15 369.0 253.0 109.1

2. AB-PINACA 3.23 331.2 215.0 286.1

3. Salvinorin A 3.39 433.1 373.0 91.1

4. 5F-PB-22 4.04 377.2 232.1 143.9

5. PB-22 4.30 359.2 214.0 144.0

6. APINACA (AKB-48) 4.76 366.3 135.1 93.1

Page 35: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

Medical/Biological 35ADVERTISEMENT

Analytical column: Raptor Biphenyl (2.7 μm, 50 mm x 3.0 mm;

cat.# 9309A5E)

Guard column: Raptor Biphenyl EXP guard column (2.7 μm,

5 mm x 3.0 mm; cat.# 9309A0253)

Mobile phase A: 0.1% Formic acid in water

Mobile phase B: 0.1% Formic acid in acetonitrile

Gradient: Time (min) %B

0.00 25

1.00 25

5.00 95

5.50 95

5.51 25

7.00 25

Flow rate: 0.6 mL/min

Injection volume: 2 μL

Column temp.: 30 °C

Ion mode: Positive ESI

Results

Chromatographic results for the original method are presented in

Figure 1. Complete resolution of all isobaric compounds and good

separation from major matrix interferences were achieved with a

cycle time of 5 min and a total analysis time of 7 min because of

the unique selectivity and retention of the Raptor Biphenyl column.

To determine whether new drugs could be successfully added

to the original method, fi ve additional synthetic cannabinoids

and salvinorin A (a psychotropic terpenoid) were analyzed under

conditions identical to the established method. As shown in Figure

2, which is an overlay of the new compounds (numbered peaks)

and the existing separation from Figure 1 (unnumbered peaks), all

compounds were separated from early-eluting matrix interferences

as well as from each other without the need for adjustments to the

mobile phase, gradient, or analytical column.

Conclusions

Labs analyzing synthetic cannabinoids are under increasing

pressure to add new compounds to their analytical testing services.

While this can be done through new method development, adding

new compounds to an existing method can save time and resources.

The method shown here demonstrates the advantages of the Raptor

Biphenyl SPP column for this analysis: due to the column’s highly

retentive, selective characteristics, 35 drugs (including isomers and

emerging compounds) can be analyzed quickly and effectively.

Time (min)

0.00 0.50 1.00 1.50 2.00 2.50 3.00 3.50 4.00 4.50 5.00 5.50

1

2

3

4

5

6

Figure 2: Overlay of new drugs (numbered peaks) and existing method chromatogram from Figure 1.

Restek Corporation

110 Benner Circle, Bellefonte, PA 16823

tel. (814) 353-1300

Website: www.restek.com

Page 36: Epitope Mapping of an Antibody Receptor Using Hydrogen ...files.alfresco.mjh.group/alfresco_images/pharma/... · Peptide Mapping of Monoclonal Antibodies and Antibody–Drug Conjugates

www.gerstel.com

Dynamic Headspace

(DHS), Headspace

Automated Pyrolysis

(PYRO)

Extraction, Clean-Up,

Evaporation,

Addition of Standards

Olfactory Detection Port

(ODP)

MAESTRO PrepAhead

productivity

SPME, SBSE,

Thermal desorption

How much packaging is inside your product?

Extractables & Leachables, Migration,

Plasticizers, Contaminants, Allergens,

Off-Odors – the list goes on.

Ensure constant high quality of

polymer-based packaging for:

• Pharmaceuticals

• Food and beverages

• Personal care and consumer products

For the highest Product Quality you can

rely on GERSTEL Solutions for GC/MS and LC/MS.

What can we do for you?