Chromatographic Method Development
Chromatographic Method Development
Development
Jenny Stanford Series on Pharmaceutical Analytics
Series Editors
Gregory K. Webster, J. Derek Jackson, and Robert G. Bell
Vol. 1 Vol. 3
Supercritical Fluid Chromatography: Chromatographic Method
Advances and Applications in Development
Pharmaceutical Analysis Gregory K. Webster and Laila Kott, eds.
Gregory K. Webster, ed. 2020
2014 978-981-4800-53-2 (Hardcover)
978-981-4463-00-3 (Hardcover) 978-0-429-20172-1 (eBook)
978-981-4463-01-0 (eBook)
Vol. 2
Poorly Soluble Drugs: Dissolution
and Drug Release
Gregory K. Webster, J. Derek Jackson,
and Robert G. Bell, eds.
2017
978-981-4745-45-1 (Hardcover)
978-981-4745-46-8 (eBook)
Jenny Stanford Series on Pharmaceutical Analytics — Volume 3
Chromatographic Method
Development
edited by
Gregory K. Webster
Laila Kott
Published by
Jenny Stanford Publishing Pte. Ltd.
Level 34, Centennial Tower
3 Temasek Avenue
Singapore 039190
Email: [email protected]
Web: www.jennystanford.com
All rights reserved. This book, or parts thereof, may not be reproduced in any
form or by any means, electronic or mechanical, including photocopying,
recording or any information storage and retrieval system now known or
to be invented, without written permission from the publisher.
Preface xxi
1.1 Introduction 1
1.2 Technology and Chromatography 2
1.2.1 New Goal of Method Development: Speed 3
1.3 The Chromatography Market 4
1.4 Chromatographic Method Development 4
1.4.1 Software for Chromatographic Method
Development 5
1.4.2 Flash Chromatography Method
Development 5
1.4.3 Orthogonality in Column Chromatography 5
1.4.4 Unified Approach to Reversed-Phase and
Normal-Phase Chromatography 6
1.4.5 Polysaccharide-Derived Chiral Stationary
Phases for the Separation of Enantiomers 6
1.4.6 Chiral Chromatography: Method
Development 6
1.4.7 Aqueous Normal-Phase Chromatography
Using Type-C Silica Columns 7
1.4.8 Ion Chromatography Columns 7
1.4.9 Ion Chromatography: Method Development 7
1.4.10 Fundamentals, Properties, and Applications
of Stationary Phases of Gas Chromatography
Method Development 8
1.4.11 Gas Chromatography Method Development 8
vi Contents
4.1 Introduction 65
4.2 Traditional View of Orthogonal LC Methods 66
4.3 Column Orthogonality in Reversed-Phase
Chromatography 66
4.3.1 Introduction 66
4.3.2 Hydrophobic Subtraction Theory 68
4.3.2.1 HST for column comparison 69
4.3.2.2 HST values for commercial
reversed-phase columns 69
4.3.2.3 HST for orthogonal reversed-phase
separations 70
viii Contents
Index 531
Preface
[email protected], [email protected]
1.1 Introduction
In the late 1990s, it was not uncommon for laboratories to argue
over whether it was robust to use the new 3 µm packed particle
columns. Colleagues would argue over the merits of using this
“new” particle over the trusted industry standard 5 µm particle.
These memories bring a smile to those of us trying to bring Ultra
High-Performance Liquid Chromatography (UHPLC) and its 1.2 to
2 µm columns into routine use.
The differences between systems designed for the 3 µm and
UHPLC columns are significant. A change to 3 µm particle did not
require new instrumentation. Not only does a change to UHPLC
Disclosure
Laila Kott is an employee of Xenon. Gregory Webster is an
employee of AbbVie. The study contains no proprietary Xenon
or AbbVie data. Xenon and AbbVie jointly participated in the
writing, review, approval, and financial support of this publication.
References
2.1 Introduction
Analytical testing and its control strategy play critical roles during the
entire life cycle of the drug product in the pharmaceutical industry.
Chromatography, especially high performance liquid chromato-
graphy (HPLC), ultra-high performance liquid chromatography
(UHPLC), and gas chromatography (GC), represent the key techniques
employed throughout many aspects of pharmaceutical analyses,
including the determination of assays, dissolution, and impurity
profiles, process control testing, and drug metabolism studies
[1–3]. In general, the use of the trial-and-error approach has been
common practice in developing analytical methods for many
years. However, this method usually requires large numbers of
(DOE-related)
Structure-
based Chromato Method
retention gram scouting/ Method Robustness
Function prediction simulation screening optimization evaluation
Chromato- Offline Online Online Online Online
graphic /Offline /Offline
simulation
software
DOE (n/a) (n/a) Offline Offline Offline
modeling
software
Online: Separation is conducted automatically for predesigned plans (combination
of parameters), thus no data input prior to simulation/calculation is required.
Offline: Separation is conducted independently of software, thus data input for
simulation/calculation is required.
Figure 2.4 A heat map generated using DryLab® software which shows
the relationship between time and resolution (top). A simulated
chromatogram at the spot marked by the black dot in the upper image
(bottom). Reprinted from [23], with permission from Elsevier.
Figure 2.10 Experimental design using the Plackett–Burman design and the resulting Pareto charts of factorial analysis showing
33
the effects of various factors on the modeled responses. Reprinted from [31], with permission from Springer (color altered).
34 Software for Chromatographic Methods Development
Factor
Experimental General Response interaction
Types of DOE design application surface plot investigation
a b
c
Figure 2.15 Surface response modeling of the resolution for the SFC
method as a function of (a) the flow rate, (b) the percentage of ethanol
in the mobile phase, and (c) the outlet pressure. Reprinted from [34],
with permission from Elsevier.
Application of Software to Analytical Method Development 41
Pros
Possibility of significantly reducing experimental/analytical
time, resources, and number of experiments required
Systematic approach enabling the identification and
statistical analysis of the optimal point or space of the
analytical method parameters
Feasibility of chromatogram simulation without carrying
out any physical experiments (only for chromatographic
software)
Easy to understand through a visualized outcome
Easier transfer for knowledge and technical skills via
better understandings of analytical methods
Cons
Possibility of inconsistency between the predicted (simulated)
and obtained chromatogram (Prediction accuracy)
Disadvantages of Using Available Method Development Software Programs 45
Figure 2.18 Comparison of three different 2-D planes of the 3-D resolution
spaces generated using DryLab® for each column. Reprinted from [4],
with permission from Elsevier.
References
J. Robert Bickler
Biotage LLC,
10430 Harris Oaks Blvd., Suite C, Charlotte, North Carolina 28269, USA
[email protected]
3.1 Introduction
Flash chromatography (a.k.a. flash purification) is a rapid
separation and purification technique (hence the term flash)
used by organic, medicinal, and natural product chemists to
isolate one or more compounds in crude chemical mixtures. The
term Flash Chromatography was first coined by W. Clark Still et
al. in his seminal paper “Rapid Chromatographic Technique for
Preparative Separations with Moderate Resolution,” published
in 1978 [1].
Flash chromatography requires only a few items—a solid, inert
sorbent (e.g., silica), a sturdy, inert column (glass, polypropylene),
only one time, TLC results are used as a substitute for scouting
runs.
CV = 1/Rf (3.1)
Table 3.1 The relationship between TLC Rf values and the number of
required column volumes for elution from a flash column
Rf CV
0.1 10
0.2 5
0.3 3.33
0.4 2.5
0.5 2
0.6 1.67
0.7 1.43
0.8 1.25
0.9 1.11
1 1
Figure 3.1 The relationship between Rf from a TLC plate and the number
of CV required to elute the compound is illustrated here. The relationship
is reciprocal allowing the chemist to determine the required solvent
volume to elute each compound in the mixture being separated.
Table 3.2 Common flash solvents and their selectivity group [1]
Table 3.3 Common flash solvents and their strength relative to silica
Solvent Strength
MeOH 0.70
EtOH 0.68
PrOH 0.60
THF 0.53
MeCN 0.51
Acetone 0.50
EtOAc 0.43
Ether 0.40
DCM 0.32
Chloroform 0.26
Toluene 0.22
Hexane —
Heptane —
Isooctane —
For example, let us say you have TLC data showing three
spots with Rf values of 0.6, 0.3, and 0.1. Using Eq. 3.1, we can
convert each Rf into its CV, determine the DCV for each adjacent
pair.
Based on this data, the lower DCV is 1.66 and the suggested
load amount for any cartridge size is found in the column headed
by <2 in Table 3.4. For instance, if the sample size is 0.1 g, the
appropriate cartridge size is 10 g.
Table 3.4 Flash cartridge load estimate (in grams) based on TLC DCV data
DCV
Cartridge
size (g) <2 2–3.9 >4
5 <0.05 0.05–0.25 0.25–0.5
10 <0.1 0.1–0.5 0.5–1.0
25 <0.25 0.25–0.75 0.75–2.5
50 <0.5 0.5–2.5 2.5–5.0
100 <1.0 1.0–5.0 5.0–10
250 <2.5 2.0–7.5 7.5–25
500 <5 5–25 25–50
750 <7.5 6.0–22.5 22.5–75
1000 <10 10–50 50–100
1500 <15 12–45 45–150
While this approach can work, it ignores the fact that the DCV’s
usefulness is in determining the optimal amount of silica for a
particular separation.
The most powerful elution method is the step gradient. It can also
be the most difficult to create and optimize. Step gradients are
more powerful because each compound in the sample mix can
have its separation parameters optimized. This has been found
to be true in many cases, and Fig. 3.4 is a nice example using
the same five-component mix used in Figs. 3.2 and 3.3.
By using an optimized step gradient, the elution of each
compound is determined by step-wise solvent ratio changes.
Some commercially available automated flash chromatography
systems have capability to create an optimized step gradient
from a pair of TLC plate data. In Fig. 3.4, Rf data from 20% and
30% ethyl acetate in hexane TLC runs were used.
Choosing between Normal- and Reversed-Phase 63
3.7 Conclusion
Flash chromatography is a useful technique for the separation
of a broad variety of compounds. It is one of the workhorse
techniques used by chemists who need to isolate compounds
that are recently synthesized, or extracted from a wide variety
of matrices.
References
Orthogonality in Chromatographic
Methods
4.1 Introduction
Throughout chromatographic method development, challenges
often arise when peaks elute with the void volume, co-elute with
each other, or do not elute at all. In these cases, it is beneficial
to evaluate and/or develop orthogonal separation methods.
Chromatographic orthogonality simply refers to a separation
technique, mode, or phase that offers unique selectivity. Early on,
this was most frequently seen as using alternate methods with
dissimilar mechanisms of separation to evaluate the selectivity
profile of the original method. This was often performed by using
a normal-phase method to evaluate a reversed-phase profile but
can also be done using TLC, GC, or capillary electrophoresis
techniques. Coupling mass spectrometric detection provides a
4.3.1 Introduction
For the last two decades, researchers have developed alternative
approaches to method development. The focus changed from
Column Orthogonality in Reversed-Phase Chromatography 67
Prelimimary
Method
ChangeBͲSolvent,
ColumnChange
AcidsorBases
Present?
Yes
No
AdjustTandeith%B
orgradienttimefor 2
Rsш 1.5
ChangepH3Ͳ5units ChangeColumn
Measure|ɷlogɲ|avg
2 2
|ɷlogɲ|avgш0.10 |ɷlogɲ|avg<0.10
1
“Orthogonal”Method
S = dipolarity/polarizability
A = solute hydrogen-bond acidity
B = solute hydrogen-bond basicity
V = molecular volume
Column Properties
e = interaction with n- and π-electrons
s = dipolar interactions
a = hydrogen-bond basicity
b = hydrogen-bond acidity
v = dispersive and cavity effects
One attempt to study silica using LSERs concluded that the LSER
methods typically used to classify differing column interactions
are not appropriate in predicting solute retention [9]. One reason
for this is the difficulty in defining the bulk stationary phase
in a bare silica column [9]. Other reports proved successful in
evaluating bonded normal-phase columns using the same LSER
approach [10] but did report negative values for dispersive and
cavity formation effects. Table 4.5 lists LSER coefficients which
have been determined for a series of traditional normal-phase
columns. This information can be useful in determining the
controlling factors that lead to differences between columns,
as well as, give an idea of which columns of similar phase type
are most unique. As seen in Table 4.5, interactions through
dipolarity and polarizability seem to be fairly consistent among
the tested columns (note, e values are not reported in the table
as they were noted to be insignificant on each test stationary
phase). Interactions through hydrogen bonding give the greatest
differences in the tested columns. For example, this could be
particularly useful in determining which cyanopropyl stationary
phases are most unique. In this case the Spherisorb CN appeared
to be orthogonal to the other cyanopropyl columns. Of course it
is important to note that mobile phase components can greatly
alter interaction types of normal-phase columns and the apparent
differences in selectivity from these evaluations may not always
remain.
74 Orthogonality in Chromatographic Methods
Particle Surface
Stationary Size area
phase Ligand (µm) (m2/g) c s a b v
Hypersil Bare Silica 5 170 –1.21 1.06 2.23 1.56 –0.83
silica
Hypersil APS Aminopropyl 5 170 –1.14 0.94 2.94 1.20 –0.72
NH2
Lichrospher Dihydroxypropyl 5 250 0.94 1.07 2.37 1.47 –0.85
Diol
Lichrospher Cyanopropyl 5 350 –0.89 0.99 1.94 1.04 –0.64
CN
Ultrasphere Cyanopropyl 5 200 –1.10 1.03 1.97 1.08 –0.60
CN
Hypersil CN Cyanopropyl 5 170 –1.16 0.95 1.86 1.15 –0.61
Spherisorb Cyanopropyl 5 220 –0.99 1.00 2.13 1.69 –0.84
CN
Source: Adapted from reference [10].
Figure 4.3 Spider diagram based on LSER models calculated with seven
descriptors and Eq. 4.4, with the retention data measured for the 85
neutral species and 24 ionizable species, for 31 columns. Chromatographic
conditions: CO2–MeOH 90:10 (v/v), 25°C, 150 bar, 1 or 3 mL/min depending
on column dimensions. Reproduced with permission from [16].
s = dipolar interactions
a = hydrogen-bond basicity
b = hydrogen-bond acidity
v = dispersive and cavity effects
d– = electrostatic interaction with anions
d+ = electrostatic interaction with cations
Further, using over 100 probe analytes, a hierarchical cluster
map of 31 stationary phases was produced using normalized
retention data as shown in Fig. 4.4. This figure is a more
practical representation of column uniqueness and is useful in
understanding the orthogonality of such columns. In Fig. 4.4,
columns which are further away from each other should result in
the largest difference in selectivity.
Figure 4.7 Normalized retention time plots for selected 2D-LC systems.
Reproduced with permission from [21].
82 Orthogonality in Chromatographic Methods
4.9 Conclusion
Separation techniques, modes of chromatography, and
stationary phase material all can be orthogonal. Many models for
characterizing orthogonality can be studied and used to make
decisions on screening column sets for method development (as
discussed in Chapter 5). It should be noted that column vendors
specifically choose to commercialize sets of columns that are
orthogonal for their internal test analytes libraries. Thus, if a
column company has two HILIC columns available, they believe
them to be orthogonal. This, however, may not prove true in
all cases when applied to real world sample or using unique
mobile phases. At the end of the day, using the tools and maps
described herein can only guide educated guesses on column
orthogonality; it is still up to the practitioner to find the right
column for the job as is outlined in the proceeding chapter.
Disclosure
Zachary S. Breitbach and Gregory K. Webster are employees of
AbbVie. AbbVie provided no additional financial support for
this work. AbbVie participated in the writing, reviewing, and
approving the publication. The presentation contains no
proprietary AbbVie data.
References
1. Pellett, J., Lukulay, P., Mao, Y., Bowen, W., Reed, R., Mac, M., Mungerc, R.
C., Dolan, J. W., Wrisley, L., Medwide, K., Toltl, N. P., Chan, C., Skibic, M.,
Biswas, K., Wells, K. A., and Snyder, L. R., ’Orthogonal’ separations for
reversed-phase liquid chromatography, J. Chromatogr. A, 2006, 1101,
122–135.
2. Wilson, N. S., Nelson, M. D., Dolan, J. W., Snyder, L. R., Wolcott, R. G., and
Carr, P. W., Column selectivity in reversed-phase liquid chromatography
I. A general quantitative relationship, J. Chromatogr. A, 2002, 961,
171–193.
3. Wilson, N. S., Nelson, M. D., Dolan, J. W., Snyder, L. R., Wolcott,
R. G., and Carr, P. W., Column selectivity in reversed-phase liquid
chromatography II. Effect of a change in conditions, J. Chromatogr. A,
2002, 961, 195–215.
References 83
4. Wilson, N. S., Nelson, M. D., Dolan, J. W., Snyder, L. R., Wolcott, R. G.,
and Carr, P. W., Column selectivity in reversed-phase liquid
chromatography III. The physico-chemical basis of selectivity, J.
Chromatogr. A, 2002, 961, 217–236.
5. Dolan, J. W., Maule, A., Bingley, D., Wrisley, L., Chand, C. C., Angodd,
M., Luntee, C., Kriskoe, R., Winstone, J. M., Homeiere, B. A., McCalley,
D. V., and Snyder, L. R., Choosing an equivalent replacement
column for a reversed-phase liquid chromatographic assay procedure,
J. Chromatogr. A, 2004, 1057, 59–74.
6. Marchand, D. H., Snyder, L. R., and Dolan, J. W., Characterization and
applications of reversed-phase column selectivity based on the
hydrophobic-subtraction model, J. Chromatogr. A, 2008, 1191, 2–20.
7. Snyder, L. R., Dolan, J. W., and Carr, P. W., A new look at the selectivity
of RPC columns, Anal. Chem., 2007, 79, 3254–3262.
8. http://www.hplccolumns.org/database/compare.php.
9. Cheong, W. J., and Choi, J. D., Linear solvation energy relationships
in normal-phase liquid chromatography based on retention data on
silica in 2-propanol/kexane eluents, Anal. Chim. Acta, 1997, 342,
51–57.
10. Park, J. H., Yoon, M. H., Ryu, Y. K., Kim, B. E., Ryu, J. W., and Jang, M. D.,
Characterization of some normal-phase liquid chromatographic
stationary phases based on linear solvation energy relationships,
J. Chromatogr. A, 1998, 796, 249–258.
11. West, C., and Lesellier, E., Characterization of stationary phases
in subcritical fluid chromatography by the solvation parameter
model I. Alkylsiloxane-bonded stationary phases, J. Chromatogr. A,
2006, 1110, 181–190.
12. West, C., and Lesellier, E., Characterisation of stationary phases
in subcritical fluid chromatography by the solvation parameter
model II. Comparison tools, J. Chromatogr. A, 2006, 1110, 191–199.
13. West, C., and Lesellier, E., Characterisation of stationary phases
in subcritical fluid chromatography with the solvation parameter
model III. Polar stationary phases, J. Chromatogr. A, 2006, 1110,
200–213.
14. West, C., and Lesellier, E., Characterisation of stationary phases
in subcritical fluid chromatography with the solvation parameter
model IV. Aromatic stationary phases, J. Chromatogr. A, 2006, 1115,
233–245.
15. West, C., and Lesellier, E., Characterisation of stationary phases
in subcritical fluid chromatography with the solvation parameter
84 Orthogonality in Chromatographic Methods
Gregory K. Webster
AbbVie Inc., Research and Development, 1 N. Waukegan Rd.,
North Chicago, Illinois 60064, USA
[email protected]
5.1 Introduction
Method development for liquid chromatography has not changed
substantially from the groundbreaking work of Snyder and
Kirkland [1] and updated texts [2, 3]. We still need selectivity
and sensitivity in validatable and transferable methods. The
requirements from the USP [4] and ICH [5] have not changed.
What has changed in this new millennium is the need for efficiency
(speed) over “academic” methods. The pharmaceutical industry
develops thousands of methods a year to support rapid drug
development. Methods need to meet their intended goal in their
role of moving the drug development pipeline forward. The
RtC18 ≈ RtC8 > RtPhenyl > RtC4 > RtC2 > RtCN >> Rtsilica
5.3.4 Screening
If early methods are suitable to meet the separation goal, use
them. Otherwise, we need to do some injections! Since method
development is about critical pair resolution [6], not plate
number [7], we need to have the critical pair(s) in mind going
into development. All too often, advocates of computer-assisted
method development are too busy encouraging the use of their
software and they forget to promote that all development starts
with a few injections. We need initial information about the
elution characteristics of the sample and the critical pairs.
Depending on the chromatographer’s experience, there are several
starting points for this. However, they all must include looking
at extremes in pH and mobile phase. Most compounds tend to
have a region of pH where they have optimal resolution.
InitialScreen
ColumnDiameter
Flow=0.4 Flow=0.2
Flow=1.0
Dimensions:100x3.0mm Dimensions:50x2.1mm
Dimensions:150x4.6mm GradientTime:14min GradientTime:7min
GradientTime:20min K*2Ͳk*5:10.8Ͳ27.1 K*2Ͳk*5:4.7–11.7
K*2Ͳk*5:15.3Ͳ38.3
Prelimimary
Method
ChangeBͲSolvent,
ColumnChange
AcidsorBases
Present?
Yes
No
AdjustTandeith%B
orgradienttimefor 2
Rsш 1.5
ChangepH3Ͳ5units ChangeColumn
Measure|ɷlogɲ|avg
2 2
|ɷlogɲ|avgш0.10 |ɷlogɲ|avg<0.10
1
“Orthogonal”Method
Figure 5.1 Model 1 approach with Chromsword®. Figure used with permission.
Reversed-Phase Chromatography 97
5.3.4.3.3 pH control
Automated systems for method development minimize the
number of mobile phases needed by taking advantage of the
Henderson-Hasselbalch equation to control buffer pH. Table 5.8
illustrates a six-level mass spectrometry detection-friendly pH
screen from pH 2.75 to 6.25 using formic acid and ammonium
formate. For sample sets requiring a low UV detection wavelength,
a similar series is easily generated with phosphate buffers.
Other pH series can be found in the literature and product websites.
98 Unified Approach to Chromatographic Method Development
Figure 5.2 Model 2 approach with Fusion®. Fusion QbD-LC Method Development White Paper, V3.2, S-Matrix Corporation. Used with
Normal-Phase Chromatography
permission.
99
100 Unified Approach to Chromatographic Method Development
2.75 100 0
3.16 80 20
3.70 50 50
4.34 20 80
5.42 5 95
6.24 0 100
5.4.5 Screening
5.4.5.1 Mobile phases
Elise et al. recently studied different mobile phase additives for
the achiral SFC separation of 160 proprietary drugs [16]. Using
methanol without additives as a reference, they studied the basic,
acidic and salt additives to methanol. The basic additives studied
were ammonium hydroxide, diethylamine, diethanolamine and
iso-propylamine. The acidic additives selected were trifluoroacetic
acid and water. The salt additives for this study were ammonium
acetate and ammonium formate. Each additive was evaluated for
their effect upon elution capability, peak shapes, UV baseline drift,
and UV and mass responses (signal-to-noise ratios) for the sample
set.
The study used Derringer desirability functions to rank the
performance of the selected additives. The authors concluded that
ammonium acetate at 20 mM was the most effective additive in
terms of chromatographic efficiency and detection. Diethylamine,
trifluoroacetic acid and water yielded surprisingly poor results.
The study also found that ammonium hydroxide provided useful
MS responses and low baseline drift with UV detection.
In a different screening study, Hicks et al. proposed that
diethylamine or formic acid were effective additives for their sample
sets [17].
5.4.5.2 Columns
In an adjacent study, Elise et al. also studied column orthogonality
with SFC for the achiral SFC separation of 160 proprietary
drugs [18]. They evaluated 1.7–2.5 µm fully porous or 2.6–2.7 µm
superficially porous particles, with a variety of stationary phase
chemistries. Because of the varying column dimensions used,
the linear speed and gradient conditions were adjusted to be nearly
equivalent for each of the columns.
In this study, a Derringer desirability functions was used to
rank the selected columns with regard to retention, retention
window and peak shapes. The results enabled the column set to
be segregated in four groupings: (i) 1-aminoanthracene and 2-
ethylpyridine phases, (ii) diol-type phases and the sole sulfobetaine
Normal-Phase Chromatography 105
Figure 5.3 LSER classification of SFC columns. Figure used with permission
from ref. [15].
5.5.5 Screening
5.5.1.1 Mobile phases
Traditionally, acetonitrile is used as the organic phase in
hydrophilic interaction chromatography. Use of small levels of
methanol and isopropanol has illustrated mixed results [25, 26].
Thus, the primary alternative to acetonitrile is the use of acetone.
Periat et al. found that for the 82 drugs they studied using
HILIC, pH 3 and pH 6 buffers at 10 and 50 mM were effective [26].
McCalley found additional selectivity with using trifluoroacetic or
heptafluorobutyric acid additives [27]. However, as these additives
can produce memory effects in columns and system tubing, they
may be best suited for a second tier screen.
110 Unified Approach to Chromatographic Method Development
5.5.1.2 Columns
While literature method development studies [25–28] have looked
to optimize columns, Periat et al. went a step further looking for
column orthogonality for HILIC separations [26]. Their approach
proposed four columns with sub-2 µm particles which seems
like an excellent starting point for early HILIC screening: Agilent
Zorbax RRHD (bare silica), Acquity BEH (hybrid silica), Thermo
Syncronis HILIC (zwitterionic phase) and Acquity BEH amide
(hybrid silica bonded with amide group).
5.6 Conclusion
Reversed-phase chromatography is once again rapidly evolving
with the advent of small particle columns with high pressure
systems optimized for minimal void volumes and fast detection.
Yet, the basics of method development for this mode is still
very much in accordance with the standards set by Snyder and
Kirkland years ago [1]. The principles set forth by this classic work
have simply been updated to reflect technology capabilities. In
addition, automated system opportunities have enabled statistical
and DOE algorithms to facilitate finding optimal separations
conditions.
Supercritical fluid chromatography has, to date, been
very successful in normal-phase applications, particularly in
pharmaceutical analysis. SFC provides several advantages over
traditional normal-phase liquid chromatography in terms of
resolution, throughput, and consumption of hazardous/expensive
solvents. However, these advantages have not been fully utilized
in achiral analysis. There is little doubt SFC will become more
and more popular in achiral analysis in the near future.
HILIC has found a very successful niche in chromatographing
polar impurities that are difficult to resolve by reversed-phase
separations. In particular, HILIC conditions are especially amenable
to mass spectrometric detection for low level sensitivity and high
selectivity.
Disclosure
AbbVie provided no financial support outside of Dr. Webster
being an employee of AbbVie. AbbVie participated in writing,
reviewing, and approving the publication. The chapter contains no
proprietary AbbVie data.
References
Polysaccharide-Derived Chiral
Stationary Phases for the Separation of
Enantiomers
6.1 Introduction
Based on the research work from Prof. Okamoto’s group [1–3],
polysaccharide-derived and silica-supported chiral stationary
phases were introduced in the 1980s. They are considered as the
first choice of chiral stationary phases (CSPs) for enantiomer
separations by liquid chromatography (LC) and supercritical fluid
chromatography (SFC). A considerable number of publications
over three decades have reported and reviewed the applications of
these CSPs by scientists working in the field of enantioseparations
[4–24].
CSP R CSP R
CH3 CH3
H tris(3,5-dimethyl- H tris(3,5-dimethyl-
CHIRALPAK IA N N
phenylcarbamate) CHIRALPAK IB phenylcarbamate)
O CH3 O CH3
Cl
H
CHIRALPAK ID N tris(3-chloro- H tris(3,5-dichloro-
phenylcarbamate) CHIRALPAK IC N
phenylcarbamate)
O Cl
O Cl
Cl
H tris(3,5-dichloro-
CHIRALPAK IE N
phenylcarbamate)
O Cl
H tris(3-chloro-4-methyl-
CHIRALPAK IF N CH3
phenylcarbamate)
O Cl
CH3
H tris(3-chloro-5-methyl-
CHIRALPAK IG N phenylcarbamate)
O Cl
H3C tris((S)-α-methyl-
CHIRALPAK IH O (S) benzylcarbamate)
N
H
CSP R CSP R
CH3 O
H tris(3,5-dimethyl- CHIRALCEL OA triacetate
CHIRALPAK AD N
phenylcarbamate) C
H3
O CH3 O
CHIRALCEL OK tricinnamate
H
N Cl tris(4-chloro-3-methyl-
CHIRALCEL OX
O CH3 phenylcarbamate)
H
CHIRALCEL OZ N CH3 tris(3-chloro-4-methyl-
phenylcarbamate)
O Cl
polar organic (PO) mode and reversed phase (RP) mode to achieve
efficient, fast, and appropriate method which is matched to the
requirements of the project and the molecule.
(a-1) (b-1)
Hexane/2-PrOH/FA
D
Hexane/2-PrOH/FA
80/20/0.1 v/v/v 80/20/0.1 v/v/v
k1 = 1.33 k1 = 1.82
ɲ = 1.63 ɲ = 1.1
Rs = 6.9
Minutes Minutes
Table 6.1 Mobile phases for sample screening with immobilized CSPs
Starting
condition Typical optimization
Solvent mixture (%) range (%)
Primary Alkane/EtOH 80/20 99/1 to 50/50
mobile phases Alkane/2-PrOH 80/20 99/1 to 50/50
Alkane/DCM/EtOH 50/50/2 85/15/0 to 0/80/20
Alkane/MtBE/EtOH 0/98/2 80/20/0 to 0/40/60
Secondary Alkane/EtOAc 50/50 80/20
mobile phases Alkane/THF 70/30 95/5 to 0/100
MeOH 100
ACN 100
Table 6.2 Mobile phases for sample screening with coated CSPs
Sample nature?
Samplenature?
Choosemobilephaseadditive
CO2 /ACN CO2 /THF CO2 /(DCM/MeOH90/10) CO2 /(MtBE/MeOH80/20) CO2 /(EtOAc/MeOH90/10)
70/30v/v 75/25v/v 80/20v/v 75/25v/v 80/20v/v
OH
H
O N Propranolol
0.3%
0.1%
N
N O N
N O N O
Praziquantel
Praziquantel Praziquantel
0
0 5 0
5 10 5
10
15 10 20 15
15 25 20
20 30 25
25
PLQ 30
30
PLQ
PLQ
ln D
lnk
lnk
0.70 0.70
1.5 0.72
1.0 1.0 0.68 0.68
ln D
lnk
7000 7000
25 25
6000 6000 10.0 10.0
Plate count
Plate count
Rs
9.5 9.5
8000 4000 4000 15 Analysis
15 time
3000 N
3000
2 9.0 9.010.5 Rs
7000 10 10
2000 2000
8.5 8.5
6000 ((e)) ((e)) 10.0 ((f)) 5 ((f)) 5
1000 1000
Plate count
20 25 9.530
20 25 35 30 40 35 45 40 50 45 55 50 55
4000 Temperature Temperature
(°C) (°C) Temperature Temperature
(°C) (°C)
3000 9.0
15.0 s
8.1 s (a-2)
Indapamide ((a-1)
T = 25°C T = 40°C
O R = 3.2
Rs Rs = 2.7
O O
S
N
N NH2
H 20 4 s
20.4
Cl
26.4 s
CHIRALCEL OD-3
(3 m, 50 x 4.6 mm i.d.)
0 30 60 0 30 60
Retention time (secon
nd) Retention time (second)
10.8 s 9.6 s
( -1))
(b ( )
(b-2)
O CH3 T = 25°C T = 40°C
Rss = 4.7 Rs = 7.7
NH
O N O
CH3
Mephobarbital
27.6 s
CHIRALPAK AD-3
(3 m, 50 x 4.6 mm i.d.) 38.4 s
(a) 19 (b)
20 17
15
HETP (m)
HETP (m)
15 13
11
10 9
5 5
0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 0.0 0.5 1.0 1.5 2.0
(mm/s) (mm/s)
H3C
H
N
CHIRALPAK AD-H (5 m) O CHIRALPAK IB (5 m)
O O
CHIRALPAK AD-3 (3 m) CHIRALPAK IB-3 (3 m)
CHIRALPAKIAͲU(subͲ2ђђm)
1.5mL/min(ђ=3.54mm
m/s)
Rs =10.0
30s
CHIRALPAKIAͲ3(3ђm)
0.6mL/min(ђ=1.42mm/s)
Rs =10.6
CHIRALPAKIA(5ђm)
0.4mL/min(ђ=0.94mm/s)
Rs =8.8
ond)
Retention time (seco
are shown in Fig. 6.12. It has to be noted that the instrument void
should be optimized when working with columns packed with
small CSP particles in order to maximize the gain in resolution
afforded by the high column efficiency. For this purpose, ultra-
high-performance of liquid chromatography (UHPLC) system is
strongly recommended to run separations with sub‑2 µm packed
columns.
For optimization of analytic method, the adjustment of
linear velocity (or the flow rate on a column with a given internal
diameter) may be helpful in search of the optimal column
efficiency, therefore the improved resolution degree. This is
particularly critical in the attempt to transfer a partial resolution
to a full one. Such an approach is normally more effective with
CSPs of larger particles than smaller particles due to the steeper
slope of the linear region in Van Deemter plot for the larger particle
size. As demonstrated in Fig. 6.13 for enantiomer resolution of
Effects of Flow Rate and Particle Size 139
N OH
H O HO
O N
O 2N N CH3 O O
O
Cl O H O
H
OH O
Oxp
prenolol OH
Ornidazole
2,3-Dibenzoyl-
y
DL-tartaric acid
(a) (c)
(b)
0s 12s 24s 36s 48s 60s 0s 12s 24s 36s 48s 60s 0s 12s 24s 36s 48s 60s
1.3 N 1 / N 1’
Rs / Rs’
S
meter
1.2
Relative param
N
1.1
N
1.0
Mequitazine
0.9
0.8
0.6 0.7 0.8 0.9 1.0 1.1 1.2 1.3
Flow rate
e (mL/min)
NC O
N O CH3
H
O N
OH
CH3O
O CH3
O
CH3O Verapamil
(aͲ1) (bͲ1)
NͲCBZͲDLͲVal
D =1.10 ɲ =1.62
Rs =1.9 Rs=6.8
NC O
N O CH3 H
O N
OH
CH3O
O CH3 O
CH3O O CH3
(bͲ2) NͲCBZͲDLͲNva
Methoxyverapamil
(aͲ2) ɲ =1.21
D =1.28 Rs=2.5
Rs =4.8
5 10 15 20 25 30
0 1 2 3 4
Minutes
Minutes
Propranolol
Pindolol
Propranolol 5.09
5.09
5.75 6.56 0.66
5.75 1.32 3.500.66
6.94
1.32
1.32
Propranolol
Propranolol 5.09
5.09 5.75 0.66
5.75 0.66 1.32
Propranolol
Carazolol HN 7.51
5.09 5.75
7.93 0.66
1.44 1.32
1.09
Metoprolol
Acebutolol O 15.70 7.14 17.14 11.87 4.11 1.32 1.11 2
O O
Acebutolol
Metoprolol 15.70
7.14 17.14
11.87 4.11
1.32 1.11
2.16
Metoprolol O O 7.14 11.87 1.32 2.16 11.57
Metoprolol O 7.14 11.87 1.32 2
O
O
Metoprolol O 7.14 11.87 1.32 2.16
Bisoprolol
Bisoprolol 6.986.98 9.22 1.27 9.22 1.57 6.20 1.27 1
Metoprolol O 7.14 11.87 1.32 2.16
O O
Bisoprolol 6.98 9.22 1.27 1.57
Note: Column size: 250 × 4.6 mm i.d., Particle size: 5 µm. Mobile phase: 1.27
Bisoprolol O O 6.98 9.22 1
O
Hexane/2-PrOH/DEA 80/20/0.1
O v/v/v. Flow rate: 1.0 mL/min, T: 25°C.
O
Bisoprolol 6.98 9.22 1.27 1.57
Bisoprolol Although
O bearing
O the same general
6.98 or basic9.22
structure, these
1.27 1.57
ß‑blockers had veryO large variation in retention factor (k1 ranging
O
from 0.57 to 5.24) and in enantioselectivity (a spreading from
1.00 to 3.67) in function of the aromatic scaffold. The most
striking difference in enantiomer separation was observed
between Alprenolol and Oxprenolol. Both molecules have a single
substituent at position‑2 of the phenoxy group. The substituent is
an allyl group in Alprenolol but an allyloxy group in Oxprenolol.
The structural difference is very subtle but the chromatographic
results were extremely distinctive. Chromatographically, racemic
Alprenolol was eluted very fast in a single peak (Fig. 6.15a), while
racemic Oxprenolol was resolved into enantiomers to a very
large degree with slightly longer retention time for the first peak
(Fig. 6.15b). There is no doubt that the allyloxy group in Oxprenolol
played an essential role in the enantioselective mechanism under
the given chromatographic conditions. This phenomenon with
the same compounds may not be as significant as described here
if the chromatographic conditions are not the same (e.g., changes
in mobile phase and/or in column).
144 Polysaccharide-Derived Chiral Stationary Phases for the Separation of Enantiomers
4.8 OH 5.9 OH
H H
O N O N
Oxprenolol
Alprenolol
13.3
(a) (b)
D = 1.00 D = 3.67
Rs = 0.0 Rs = 13.6
0 2 4 6 8 10 12 14 16 18 PLQ 0 2 4 6 8 10 12 14 16 18 PLQ
12.6
15.8
13.5
O O
Non-standard
F
mobile phases
(4-Fluorophenyl)-
Ȗ-butyrolactone
(a) (b)
6 8 10 12 14 16 18 min
6 8 10 12 14 16 18 min
13.1
13.5
16.0
4.4
14
Regeneration
(d) (c)
6 8 10 12 14 16 18 min 6 8 10 12 14 16 18 min
Figure 6.16 Effect of the non-standard mobile phases and the regeneration:
CHIRALPAK IA (5 µm, 250 × 4.6 mm i.d.); flow rate: 1.0 mL/min;
mobile phase: hexane/EtOH 90/10 v/v. T: 25°C. (a) Initial separation.
(b) Separation obtained after application of DCM‑containing mobile
phase. (c) Separation obtained after application of EtOAc-containing
mobile phase. (d) Separation obtained after column regeneration.
148 Polysaccharide-Derived Chiral Stationary Phases for the Separation of Enantiomers
6.8 Conclusions
The technology advancement in preparing polysaccharide-based
chiral stationary phases has provided an impetus to the field of
chiral separation by chromatography. It gives rise to CSPs with
higher performance, broader application scope, higher versatility
and enhanced robustness. It also makes the approaches of method
development evolve to reach higher success rate in analytical and
preparative separations of chiral compounds.
The establishment of the screening protocols by choosing
the appropriate chiral columns and the suitable mobile phase is
essential for efficient method development. The option to use 3 to
4 immobilized polysaccharide‑derived CSPs in combination with
the 4 most versatile mobile phase systems fits well the current
challenge of fast screening for an increased number of compounds
in a limited time frame. The choice in chromatographic mode
(SFC or HPLC, NP or RP, etc.) can be considered in connection with
the instrument availability, the application purpose and specific
aspects of the sample source and sample properties.
Based on the screening results, the optimization to obtain
the best suited method can be accomplished by fine‑tuning the
mobile phase composition, by using the most appropriate additives
in amount and in nature, by rational use of sample solvent, by
adjustment in temperature and in flow rate, by changing the
column dimension, by considering the particle size in the column
and, of course, without forgetting to try the alternative columns
which may exhibit complementary properties for chiral separation.
It is important to note that many methods issuing the first sample
screening can directly meet the separation objective with no need
of further optimization efforts. Fast or ultra‑fast chiral analysis
can be achieved by running short columns packed with 3 µm or
sub‑2 µm particles on optimized chromatographic systems such
as UHPLC or ultra-high-performance SFC.
References
2. Okamoto, Y., Kawashima, M., and Hatada, K. (1984) J. Am. Chem. Soc.
106 p. 5357.
3. Okamoto, Y., Aburatani, R., and Hatada, K. (1987) J. Chromatogr. A 389
p. 95.
4. Dingenen, J. (1994) Polysaccharide phases in enantioseparations in
A Practical Approach to Chiral Separations by Liquid Chromatography,
ed., Subramanian, G., Chapter 6 (VCH, Weinheim).
5. Francotte, E. (1997) Chromatography as a separation tool for the
preparative resolution of racemic compounds in Chiral Separations,
Applications and Technology, ed., Ahuja, S., Chapter 10 (American
Chemical Society, Washington).
6. Okamoto, Y., and Kaida, Y (1994) J. Chromatogr. A 666 p. 403.
7. Francotte, E. (1994) J. Chromatogr. A 666 p. 565.
8. Okamoto, Y., and Yashima, E. (1998) Angew. Chem. Intl. Ed. Engl. 37
p. 1020.
9. Maier, N. M., Franco, P., and Lindner, W. (2001) J. Chromatogr. A 906
p. 3.
10. Yashima, E. (2001) J. Chromatogr. A 906 p. 105.
11. Tachibana, K., and Ohnishi, A. (2001) J. Chromatogr. A 906 p. 127.
12. Franco, P., Senso, A., Oliveros, L., and Minguillón, C. (2001) J. Chromatogr.
A 906 p. 155.
13. Francotte, E. (2001) J. Chromatogr. A 906 p. 379.
14. Aboul-Enein, H. Y. (2001) J. Chromatogr. A 906 p. 185.
15. Cox, G. (ed.) (2005) Preparative Enantioselective Chromatography
(Blackwell Publishing, Oxford).
16. Zhang, T., and Franco, P. (2007), Chiral Separation Techniques: A Practical
Approach (3rd revised and updated Edition) ed., Subramanian, G.,
Chapter 3 (VCH, Weinheim).
17. Franco, P., and Zhang, T. (2008) J. Chromatogr. B 875 p. 48.
18. Zhang, T., Nguyen, D., and Franco, P. (2010) J. Chromatogr. A 1217
p. 1048.
19. Lee, J., Lee, J. T., Watts, W. L., Barendt, J., Yan, T. Q., Huang, Y., Riley, F.,
Hardink, M., Bradow, J., and Franco, P. (2014) J. Chromatogr. A 1374
p. 238.
20. Khater, S., Lozac’h, M.-A., Adam, I., Francotte, E., and West, C. (2016)
J. Chromatogr. A 1467 p. 463.
21. Francotte, E. (2016) LCGC Eur. 29 p. 194.
150 Polysaccharide-Derived Chiral Stationary Phases for the Separation of Enantiomers
22. Baudelet, D., Schifano-Faux, N., Ghinet, A., Dezitter, X., Barbotin, F.,
Gautret, P., Rigo, B., Chavatte, P., Millet, R., Furman, C., Vaccher, C., and
Lipka, E. (2014) J. Chromatogr. A 1363 p. 257.
23. Rizzo, S., Menta, S., Faggi, C., Pierini, M., and Cirilli, R. (2014) J.
Chromatogr. A 1363 p. 128.
24. Lämmerhofer, M. (2010) J. Chromatogr. A 1217 p. 814.
25. Zhang, T., and Franco, P. (2008) LC‑GC Europe 21(9) p. 430.
26. Cirilli, R., Ferreti, R., Gallinilla, B., La Torre, F., Mai, A., and Rotili, D.
(2006) J. Sep. Sci. 29 p. 1399.
27. De la Puente, M. L., White, C. T., Rivera-Sagredo, A., Reilly, J., Burton, K.,
and Harvey, G. (2003) J. Chromatogr. A 983 p. 101.
28. De la Puente, M. L. (2004) J. Chromatogr. A 1055 p. 55.
29. Andersson, M. E., Aslan, D., Clark, A., Roeraade, J., and Hagman, G.
(2003) J. Chromatogr. A 1005 p. 83.
30. Wetli, H. A., and Francotte, E. (2007) J. Sep. Sci. 30 p. 1255.
31. Matthijs, N., Maftouh, M., and Heyden, Y. V. (2006) J. Chromatogr. A
1111 p. 48.
32. Zhang, T., and Franco, P. (2010) LC‑GC Europe 23(6) p. 302.
33. Franco, P., and Zhang, T. (2013) Chiral Separations, Methods and
Protocols (Second edition), ed., Scriba, G., Chapter 6 (Humana Press),
pp. 113–126.
34. Zhang, T., Nguyen, D., and Franco, P. (2008) J. Chromatogr. A 1191
p. 214.
35. Younes, A. A., Ates, H., Mangelings, D., and Vander Heyden, Y. (2013)
J. Pharm. Biomed. Anal. 75 p. 74.
36. Zhang, T., Kientzy, C., Franco, P., Ohnishi, A., Kagamihara, Y., and
Kurosawa, H. (2005) J. Chromatogr. A 1075 p. 65.
37. Zhang, T., Nguyen, D., Franco, P., Murakami, T., Ohnishi, A., and
Kurosawa, H. (2006) Anal. Chim. Acta 557 p. 221.
38. Zhang, T., Nguyen, D., Franco, P., Isobe, Y., Michishita, T., Murakami,
T. (2008) J. Pharm. Biomed. Anal. 46 p. 882.
39. Zhang, T., and Franco, P. (2004) Int. Lab. 34‑5 p. 24.
40. Cox, G. B. (2008). A screening process for development of
enantioselective supercritical fluid chromatography separation
methods, LC.GC Application notes.
41. Amoss, C., Cox, G. B., Franco, P., and Zhang, T. (2008) CHIRALPAK®
IC™—An Immobilized Polysaccharide Chiral Stationary Phase with a
Unique Chiral Selector, LC.GC Application Notes.
References 151
42. Zhang, T., Schaeffer, M., and Franco, P. (2005) J. Chromatogr. A 1083
p. 96.
43. Cox, G. B., Maier, N. M., Zhang, T., and Franco, P. (2009) LC GC Application
Notes.
44. Zhang, T., Nguyen, D., and Franco, P. (2006) J. Sep. Sci. 29 p. 1517.
45. Ghanem, A., and Aboul Enein, H. Y. (2005) Anal. Chim. Acta 548 p. 26.
46. Ghanem, A., and Naim, L. (2006) J. Chromatogr. A 1101 p. 171.
47. Ali, I., Naim, L., Ghanem, A., and Aboul Enein, H. Y. (2006) Talanta 69
p. 1013.
48. Cirilli, R., Simonelli, A., Ferretti, R., Bolasco, A., and Climenti, P. (2006)
J. Chromatogr. A 1101 p. 198.
49. Cirilli, R., Orlando, V., Ferretti, R., Luciana Turchetto, L., Silvestri, R.,
De Martino, G., and La Torre, F. (2006) Chirality 18 p. 621.
50. Andersson S. (2007) Chiral Separation Techniques: A Practical
Approach (3rd revised and updated Edition), ed., Subramanian, G.,
Chapter 17 (VCH, Weinheim).
51. Cirilli, R., Ferretti, R., Gallinella, B., De Santis, E., Zanitti, L., and La Torre,
F. (2008) J. Chromatogr. A 1177 p. 105.
52. Cirilli, R., Ferretti, R., Gallinella, B., Billa, A. R., Vincieri, F. F., and
La Torre, F. (2008) J. Sep. Sci. 31 p. 2206.
53. Cirilli, R., Ferretti, R., De Santis, E., Gallinella, B., Zanitti, L., and
La Torre, F. (2008) J. Chromatogr. A 1190 p. 95.
54. Ferretti, R., Gallinella, B., La Torre, F., Zanitti, L., Turchetto, L.,
Mosca, A., and Cirilli, R. (2009) J. Chromatogr. A 1216 p. 5385.
55. Pawar, R. S., Grundel, E., Mazzola, E., White, K. D., Krynitsky, A. J., and
J. I. Rader, J. I. (2009) J. Sep. Sci. 33 p. 200.
56. Wang, C., Armstrong, D. W., and Chang, C. D. (2008) J. Chromatogr. A
1194 p. 172.
57. García, P., Franco, P., Alvarez, R., and De Lera., U. R. (2011) J. Sep. Sci.
34 p. 999.
58. Zhang, T., Franco, P., Nguyen, D., Hamasaki, R., Miyamoto, S., Ohnishi, A.,
and Murakami, T. (2012) J. Chromatogr. A 1269 p. 478.
59. Ashok, S., Varma, M. S., and Swaminathan, S. (2012) J. Chromatogr.
Sci. 50 p. 799.
60. Zhou, J., Du, Q. Z., Zhao, S. Z., Sun, F., Li, X. Y., and Zhang, Z. Z. (2014)
J. Chromatogr. Sci. 53 p. 959.
61. Geryka, R., Kalíkováa, K., Vozkaa, J., Plecitáa, D., Schmidb, M. G., and
Tesărová, E. (2014) J. Chromatogr. A 1363 p. 155.
152 Polysaccharide-Derived Chiral Stationary Phases for the Separation of Enantiomers
62. Peluso, P., Mamane, V., Aubert, E., and Cossu, S. (2014) J. Sep. Sci. 37
p. 2481.
63. Harrison, A., Melchionna, M., Franco, P., and Hlavac, J. (2014) J. Chem.
38 p. 5491.
64. Rizzo, S., Menta, S., Benincori, T., Ferretti, R., Pierini, M., Cirilli, R., and
Sannicolo, F. (2015) Chirality 27 p. 888.
65. Geryk, R., Kalíková, K., Vozka, J., and Tesarŏvá, E. (2015) Chromato-
graphia 78 p. 909.
66. Liu, N., Dong, F., Xu, J., Liu, X. G., Chen, Z. L., Tao, Y., Pan, X. L., Chen, X. X.,
and Zheng, Y. Q. (2015) J. Agric. Food Chem. 63 p. 6297.
67. Islam, M. F., and Lee, W. (2015) J. Chosun Natural Sci. 8 p. 111.
68. Rao, R. N., and Kumar, K. N. (2015) J. Chromatogr. Sci. 53 p. 295.
69. Menta, S., Carradori, S., Secci, D., Faggi, C., Mannina, L., Cirilli, R., and
Pierini, M. (2015) J. Org. Chem. 80 p. 11932.
70. López-Ram-de-Víu, P., Gálvez, I., and Díaz-de-Villegas, M. D. (2015)
J. Chromatogr. A 1390 p. 78.
71. Qian, Y., Zhao, X. Q., Zhao, L., Cui, L. L., Liu, L., Jiang, X. L., Liu, Y. J., Gao,
L. P., and Xia, T. (2015) J. Chromatogr. B 1006 p. 1.
72. Cirilli, R., Ferretti, R., Gallinella, B., Turchetto, L., Zanitti, L., and La
Torre, F. (2015) J. Pharm. Biomed. Anal. 50 p. 9.
73. Zanitti, L., Ferretti, R., Gallinella, B., La Torre, F., Sanna, M. L., Mosca, A.,
and Cirilli, R. (2015) J. Pharm. Biomed. Anal. 52 p. 665.
74. Sola, I., Viayna, E., Gómez, T., Galdeano, C., Cassina, M., Camps, P., Romeo,
M., Diomede, L., Salmona, M., Franco, P., Schaeffer, M., Colantuono,
D., Robin, D., Brunner, D., Taub, N., Hutter-Paier, B., and Muñoz-Torrero,
D. (2015) Molecules 20 p. 4492.
75. Moldovan, R. C., Dascal, G. S., Mirel, V., Bodoki, E., and R. Oprean, R.
(2015) Pharmacia 63 p. 909.
76. Adhikari, S., Kang, J. S., and Lee, W. (2016) Chirality 28 p. 789.
77. Srinivasu, G., Nagesh Kumar, K., Thirupathi, C., Lakshmi Narayana, C.,
and Parameswara Murthy, C. (2016) Chromatographia 79 p. 1457.
78. Saduttoa, D., Ferretti, R., Zanitti, L., Casulli, A., and Cirilli, R. (2016)
J. Chromatogr. A 1445 p. 166.
79. Gallinella, B., Ferretti, R., Zanitti, L., Sestili, I., Mosca, A., and Cirilli, R.
(2016) J. Pharm. Anal. 6 p. 132.
80. Kim, S. J., Nam, K. W., Park, B., Md. F. Islam, Md. F., and Lee, W. (2016)
Korean Soc. Biotechnol. Bioeng. J. 31 p. 186.
References 153
81. Lotter, C., Poehler, E., Heiland, J., Mauritz, L., and Belder, D. (2016)
Lab Chip. 16 p. 4648.
82. Török, G., Goetelen, L., Luyckx, R., and Van Broeck, P. (2005) J. Pharm.
Biomed. Anal. 39 p. 425.
83. Huybrechts, T., Török, G., Vennekens, T., Sneyers, R., and Vrielynck, S.
(2007) LCGC Europe 20 p. 319.
84. Ishikawa, A., and Shibata, T. (1993) J. Liq. Chromatogr. 16 p. 859.
85. Zhou, L., Welch, C., Lee, C., Gong, X., Antonucci, V., and Ge, Z. (2009)
J. Pharm. Biomed. Anal. 49 p. 964.
86. Amoss, C., Coryell, B. R., Cox, G. B., and Maier, N. M. (2010) Poster at
the HPLC2010, Boston, USA.
87. Cirilli, R., Ferretti, R., Gallinella, B., and Zanitti, L. (2013) J. Chromatogr.
A 1304 p. 147.
88. Materazzo, S., Carradori, S., Ferretti, R., Gallinella, B., Secci, D., and
Cirilli, R. (2014) J. Chromatogr. A 1327 p. 73.
89. Gallinella, B., Bucciarelli, L., Zanitti, L., Ferretti, R., and Cirilli, R. (2014)
J. Chromatogr. A 1339 p. 210.
90. Ferretti, R., Zanitti, L., Casulli, A., and Cirilli, R. (2016) J. Sep. Sci. 39
p. 1418.
91. Petersson, P., Lundell, N., and Markldes, K. E. (1992) J. Chromatogr. 623
p. 129.
92. Wang, R. Q., Ong, T. T., Tang, W. H., and Ng, S. C. (1012) Trends Anal.
Chem. 37 p. 83.
93. Kalíková, K., Slechtová, T., Vozka, J., and Tesărová, E. (2014) Anal.
Chim. Acta 821 p. 1.
94. Regalado, E. L., and Welch, C. J. (2015) J. Sep. Sci. 38 p. 2826.
95. Lemasson, E., Bertin, S., and West, C. (2016) J. Sep. Sci. 39 p. 212.
96. Cox, G., Stringham, R., and Matabe A. (2002) LC GC North America,
The Application Notebook, August, p. 24.
97. Maftouh, M., Granier-Loyaux, C., Chavana, E., Marini, J., Pradines, A.,
Vander Heyden, Y., and Picard, C. (2005) J. Chromatogr. A 1088 p. 67.
98. Diehl, G., Meishammer, A., Huynh, D., and Francotte, E. (2006)
Presentation at SPICA 2006, Innsbruck, Austria.
99. Zhang, T., Nguyen, D., and Franco, P. (2007) Presentation at the
HPLC2007, Ghent, Belgium.
100. Cox, G. B. (2007) Presentation at the 1st International Conference
on SFC, Pittsburgh, Pennsylvania, USA (September 2007).
154 Polysaccharide-Derived Chiral Stationary Phases for the Separation of Enantiomers
121. Gasparrini, F., Marini, F. F., Pierini, M., and Villani, C. (1999) Enantiomer
4 p. 325.
122. Fulde, K., and Frahm, A. W. (1999) J. Chromatogr. A 858 p. 33.
123. Persson, B. A., and S., Andersson, S. (2001) J. Chromatogr. A 906
p. 195.
124. Balmer, K., Lagerström, P. O., and Persson, B. A. (1992) J. Chromatogr.
592 p. 331.
125. Majors, R. E. (2006) LC‑GC Europe 19 p. 352.
126. Butchart, K., Foster, G., and Temesi, D. (2007) Chromatogr. Today 1
p. 3.
127. Aboul‑Enein, H. Y., and Serignese, V. (1994) Chirality 6 p. 378.
128. Aboul‑Enein, H. Y., and Bakr, S. A. (1997) Chirality 9 p. 10.
129. Toribio, L., del Nozal, M. J., Bernal, J. L., Jimenez, J. J., and Alonso, C.
(2004) J. Chromatogr. A 1046 p. 249.
130. Toribio, L., del Nozal, M. J., Bernal, J. L., Alonso, C., and Jimenez, J. J.
(2005) J. Chromatogr. A 1091 p. 118.
131. Myrdal, P. B., Angersbach, B. S., Karlage, K., and Kuehl, P. J. (2006) J.
Chromatogr. A 1132 p. 315.
132. Gaffney, M. H., Stiffin, R. M., and Wainer, I. W. (1989) Chromatographia
27 p. 15.
Chapter 7
7.1 Introduction
In achiral chromatography, there are many understood interactions
and equilibria that take place between the analytes, the solid phase
of the column and the liquid (mobile) phase. These interactions
can be modeled (see Chapter 2) and often fall within well-
established theories and equations of chromatographic separations.
The same is not true for chiral separations. Chiral separations
depend initially on how well (points of contact) the chiral molecules
interact with the chiral selector on the solid phase of the column.
The packing of the column, the number of chiral selectors and
how they are attached (i.e., coated vs. immobilized) have effects on
the chiral separations and dictate how much method development
space there is within given chromatographic parameters. Many
chiral methods have very small design spaces as the chiral selectors
work under quite specific conditions. For example, in an achiral
assay, if one had two closely eluting peaks, one could adjust the
column (longer, smaller particle size, smaller internal diameter) to
increase the resolution. These parameters will all work for chiral
method development as well. In reverse-phase chromatography,
one can almost always slow down elution by slowing the change
from polar to non-polar liquids. This theoretically can be done ad
infinitium, as the peaks will continue to spread but nonetheless,
what happens to the peaks is highly predictable. Conversely,
in chiral chromatography one may only have a window of 5%
to 10% organic where the interactions with the chiral phase
favor chiral separation. Going below or above that range may
cause the two enantiomers to, once again, merge into one
peak. Overall, the goal of chiral chromatography is to achieve a
resolution (Rs) of at least 1.5 between enantiomer peaks. In some
cases when the 1.5 resolution is not achievable, the method can
still be used, if the small enantiomer peak elutes prior to the parent
peak. This order of elution is a second (and sometimes achievable)
goal when developing a chiral method. Once an enantioselective
stationary phase has been found, then much of the chiral method
development follows the same processes as developing an
achiral method.
These goals lead to the first and most useful part of chiral
chromatographic method development—column screening. This
chapter presents a roadmap for the development of a chiral purity
method.
7.2.1 Polarimeters
Normal light waves vibrate in many planes; however, plane
polarized light is generated when normal light is passed through
an optical polarizing filter. This effect results in a light beam
emerging that vibrates in a single plane (linearly polarized). Linearly
polarized light is rotated when passing through a compound which
is optically active. The degree of rotation depends on both the
concentration of a chiral compound and its molecular structure.
Every optically active substance has its own specific rotation
(degree of rotation in polarized light). The specific rotation of
the molecule, not the absorption characteristics, is what determines
the signal strength using the polarimeter.
Drude’s equation highlights the effect of wavelength on the
specific rotation [a]l, such that
al =
A i
, (7.1)
( l2 – li )
Circularly
polarized light: The two beams of linearly polarized light that are of equal
amplitude and are a quarter wave out of phase.
Start at the End, with the Detectors 161
A=e×c×l (7.2)
162 Chiral Chromatography
and it has been discussed that the circular dichroism (CD) signal
is the difference between the absorbed right (A(r)) and left (A(l))
circularly polarized light:
CD = Ar – A1 (7.3)
( er – el ) × c × l e (7.5)
=
e×c ×l e
which results in an expression independent of concentration and
path-length, provided they are constant for both measurements.
A g-factor of zero would then be indicative of a racemic mix.
Column dimensions
Conditions 4.6 × 250 mm 4.6 × 250 mm 4.6 ×150 mm
Particle size (µm) 10 5 3
Flow rate (mL/min) 1.5 2 3
Starting % MeOH 4 4 1
Hold (min) 4 4 0
Ramp time to 40% 18 9 5
(min)
Hold (min) 3 2 1
Post equilibration time 5 1 0
(min)
Run cycle time (min) 30 16 6
Source: Reproduced with permission from [12].
Typical
Particle dimensions
Manufacturer Type of phase Column name sizes (µm) (mm)
Waters polysaccharide Cell-1 Cell-2 2.5 3.0 × 100
(Trefoil series) Amy-1 (150)
Phemomenex polysaccharide Amylose-1, -2 3, 5 3.0 × 100
(Lux series) Cellulose-1, -2, (150)
-3, -4
iCellulose-5
iAmylose-1
Chiralcel polysaccharide See Figures Sub 2, 3, 5 2.1 (3.0) ×
6.1 and 6.2 in 150
Chapter 6
Regis Pirkle type Whelko (S,S) 1.8 4.6 (3.0) ×
and (R,R) (Whelko 250 (150)
Ulmo (S,S) and O1), 3.5,
(R,R) 5, 10
Next Step—the Type of Chromatography 167
Parameter Settings
pH’s screened 1 to 2 Neutral (6.8) 9.0
Organic phase, Methanol Acetonitrile
first pass
Gradient 90 (aqueous):10 90 (aqueous):10 (ACN) to
(methanol) to 20:80 30:70
Column 4.6 × 100 or 3.0 × 100 or 2.1 × 100 or 150
150 mm [3 µm 150 mm [3 µm mm [3 µm particle
particle size] particle size] size]
Flow rate 1.0 0.75 0.5
(ml/min)
Gradient time 20 to 40 min
Detection Collect all wavelengths, if possible, otherwise 220,
254, 280 nm
Detection Circular dichroism at estimated appropriate
(optional) wavelengths
Detection Mass spectrometry if aqueous phases used are volatile
(optional)
Choose additive based on pKa, choose a range Choose additive based on pKa, choose a range
2 pH units lower: 2 pH units higher:
• TFA: pH range <2.5 • Triethanolamine: pH range 6.8 to 8.8
• Formic acid: pH range 2.8 to 4.8 • Diethanolamine: pH range 7.9 to 9.9
• Acetic acid: pH range <2.5 • Ammonia: pH range 8.2 to 10.2
• Triethanolamine: pH range <2.5 • Ethanolamine: pH range 8.5 to 10.5
Next Step—the Type of Chromatography
Figure 7.2 Flow chart to aid in choosing an additive for chiral screening.
169
170 Chiral Chromatography
Chiral Column
selector Modification(s) name Selective for
Vancomycin None Chirobotic V Neutral molecules, amides,
acids, esters and cyclic
amines
Teicoplanin None Chirobiotic T Underivatized amino
acids, N-derivatized amino
acids, carboxy cyclic acids,
phenols, small peptides,
aromatic molecules, and
aliphatic and cyclic amines
Teicoplanin Sugars Chirobiotic Amino acids and other
aglycone removed TAG amphoteric molecules,
cyclic amino acids, and
neutral molecules such as
diazopenes, hydantoin and
oxazolidinones
Ristocetin A Chirobiotic R Anionic analytes (i.e., α-
hydroxy acids, substituted
aliphatic acids), chiral
alcohols, secondary and
tertiary amines
Phase/chiral Commercially
Column separator available Reference
7.5 Conclusions
As discussed in this chapter and in Chapter 6, the key to quick
chiral method development is first and foremost to find the
conditions and column to achieve the enantiomeric separation.
This can be simplified by screening multiple columns under
multiple conditions, and even further simplified with the use of
chiral detectors. Once conditions that work have been found, then
the method development can be taken off-line from the screening
system for optimization, which can be similar to achiral optimization
at this point. Different approaches and techniques have been
discussed, and all work well for certain compounds. It may take
several attempts to find the right conditions for separation of
enantiomers, but options are many and usually some separation of
enantiomers is found.
Acknowledgments
The authors would like to thank Zachary Breitbach for review of
the chapter and providing useful insights.
Disclosure
Laila Kott is an employee of Xenon. Gregory Webster is an
employee of AbbVie. The study contains no proprietary Xenon or
AbbVie data. Xenon and AbbVie jointly participated in writing,
reviewing, and approving and providing financial support for
this publication.
References
13. Zhang, T., Franco, P. (2010) Finding the best separation for
enantiomeric mixtures, LCGC North America. 28(9), pp. 818–822.
14. Woiwode, U., Neubauer, S., Kaupert, M., Lindner, W., Lammerhofer,
M. (2017) Trends in enantioselective high performance liquid
chromatography, LCGC Europe. (Special issue) 30(6), pp. 34–42.
15. Kennedy, J. H. (1996) Comparison of chiral separations on
polysaccharide chiral stationary phases to an improved Pirkle-type
phase, Journal of Chromatography. 725, pp. 219–24.
16. Siluveru, M., Stewart, J. T. (1996) Stereoselective determination of
R(-)- and S(+)-prilocaine in human serum using a brush-type chiral
stationary phase, Journal of Pharmaceutical and Biomedical Analysis.
15, pp. 389–92.
17. Zhang, X., Ouyang, J., Yang, Y. (2001) A simple method for chiral
separation of ephedrines using (R)-1-naphthylglycine and 3,5,-
dinitrobenzoic acid as stationary phase, Analytical Letters. 34,
pp. 1851–1865.
18. Shao, B.-H., Xu, X.-Z., Wu, Q.-Z., Lu, J.-D., Fu, X.-Y. (2005) Comparative
enantioseparation of 2-arylpropionic acid esters on cellulose
derivative and (S,S)-Whelk-O 1 columns, Journal of Liquid
Chromatography & Related Technologies. 28, pp. 63–80.
19. Shao, B.-H., Xu, X.-Z., Wu, Q.-Z., Lu, J.-D., Fu, X.-Y. (2003) Comparison
of enantioseparation and chiral recognition mechanism of racemic
naproxen esters on (S,S)-Whelk-O 1 and CDMPC chiral columns,
Acta Chimica Sinica. 61, pp. 1635–1640.
20. Madhavan, P., Rao, B. M., Pravin, A., Kumar, P. R., Screenivasulu,
M., Chandrasekhar, K. B. (2007) A validated chiral HPLC method
for the determination of enantiomeric purity of R-β-amino-β-(4-
methoxyphenyl) propionic acid, Chromatographia. 65, pp. 81–84.
21. Szczerba, T. (2011) Summarization of screening hits on the
Whelk-O® 1, RegisPack and RegisCell chiral stationary phases (CSPs),
LCGC North America. (Suppl.), 24 (February) p. 24.
22. Holzheuer, W. B., Wong, M. M., Webster, G. K. (2009) Evaluation
of Pirkle-type stationary phases in chiral method development
screening for compounds of pharmaceutical interest, Critical
Pharmaceutical Analysis. 5, pp. 10–20.
23. McDevitt, T. F., Vicente, G., Webster, G. K., Szczerba, T. (2009) Column
coupling to solve a challenging API separation using the Whelk-O®
1 chiral stationary phase, LC/GC Application Notebook. September
2009 p. 23.
178 Chiral Chromatography
8.1 Introduction
A variety of approaches and stationary phases are available for the
separation of mixtures or analyses of specific compounds based
on the chemical and structural properties of the analyte. A large
fraction of current chromatographic methods utilize a format that
revolutionized separation science more than five decades ago;
chemically bonded stationary phases that are fabricated on silica
as a support material with hydrophobic moieties attached to the
surface. Columns made from these materials are referred to as
reversed-phase (RP) and the most common bonded group is the
straight chain C18 hydrocarbon. Other chain lengths between C4
and C30 have demonstrated the same type of retention based on
hydrophobic interactions with the degree of retention based on
the number of carbon atoms, the amount of the bonded phase and
the properties of the analytes. Other types of bonded moieties with
significant hydrophobic character can also provide RP retention and
many have been synthesized over the years but only a relatively
small number have merited commercial development. While other
contributions to retention can occur on these phases, very often
adsorption effects from residual silanols on the silica surface, the
hydrophobic effects are usually the dominant factor in these types
of chromatographic columns.
However, the analysis of polar (hydrophilic) compounds has
always presented a serious challenge due to the low retentivity
on typical RP stationary phases. In this case, the typical approach
for many analyses was to derivatize the analyte to make it less
polar so that it could be retained on a typical RP column. Only a
few stationary phases were polar enough to provide significant
retention for these types of compounds. The most common being
ion-exchange materials which could be used for analytes possessing
either a permanent positive or negative charge. This could often be
achieved by adjusting the pH of the mobile phase or dealing with
the salts of the parent compound. The relatively few good options
for polar compounds provided opportunities for new approaches
and materials to deal with these challenging types of analytes.
In recent years, there have been two types of chromatographic
materials developed more specifically designed for the analysis of
Silica Hydride 181
Figure 8.5 Calibration curve for homocysteine in blood plasma using the
diamond hydride stationary phase in the aqueous normal phase mode.
Reprinted with permission from ref. 36.
Figure 8.7 Analysis of fructose (1) and glucose (2) in various beverages
using the Diamond Hydride column in the aqueous normal phase mode.
(A) Cola, (B) red wine, and (C) grape juice. Reprinted with permission
from ref. 51.
Figure 8.8 Retention of the drug zanamivir under optimal conditions
on several commercially available HPLC columns. (a) Scherzo SM C18,
(b) Agilent Zorbax SB-Aq, (c) Luna HILIC, (d) Cogent Bidentate C18 and
(e) Cogent Diamond Hydride. Reprinted with permission from ref. 57.
Applications of Silica Hydride-Based Separation Materials 197
8.5 Conclusions
Silica hydride-based separation materials are being recognized
as an alternative to traditional silica stationary phases.
Its unique properties provide chromatographic advantages that
can be exploited for improved and more efficient separations. The
low water content is one feature that has the broadest impact on
differentiating silica hydride from ordinary silica. The resulting
mildly hydrophobic surface surprisingly retains both polar and
nonpolar compounds. The ability to retain polar compounds has
been attributed to the strongly negative surface as determined by
zeta potential measurements. This negative charge is thought to
be similar to oil droplets in organo/aqueous solvents. In this case,
hydroxides from the auto dissociation of water are responsible for
the high negative zeta potential. The excellent analytical
reproducibility of the silica hydride materials is also another
distinguishing feature. The ANP capabilities of silica hydride phases
have produced the majority of applications to date since they
often have provided superior performance to HILIC. However,
rugged and reproducible reversed-phase and organic normal
phase analyses can also be done on silica hydride columns. It is
expected that use of these materials will continue to expand as
more scientists become aware of their unique properties.
References
27. Pesek, J. J., Matyska, M. T., and Lee, P. (2011). Synthesis of a preparative
C30 stationary phase on a silica hydride surface and its application
to carotenoid separation, J. Liq. Chromatogr. Rel. Technol., 34,
pp. 231–240.
28. Rocchi, S., Rocco, A., Pesek, J. J., Matyska, M. T., Capitani, M., and
Fanali, S. (2015). Use of a novel sub-2 µm vancomycin silica hydride
stationary phase, J. Chromatogr. A, 1381, pp. 149–159.
29. Pesek, J. J., Boysen, R. I., Hearn, M. T. W., and Matyksa, M. T. (2014).
Hydride- based stationary phases: A rapidly evolving technology
for the development of new bioanalytical methods, Anal. Methods, 6,
pp. 4496–4503.
30. Pesek, J. J., Matyska, M. T., Fischer, S. M., and Sana, T. R. (2008). Analysis of
hydrophilic metabolites by high-performance liquid chromatography-
mass spectrometry using a silica hydride-based stationary phase,
J. Chromatogr. A, 1204, pp. 48–55.
31. Chalkraft, K. R., and McCarry, B. E. (2013). Tandem LC columns for
the simultaneous retention of polar and nonpolar molecules in
comprehensive metabolomic analysis, J. Sep. Sci., 36, pp. 3478–3485.
32. Gouzy, A., and Larrouy-Maumus, G. (2014). Mycobacterium tuberculosis
exploits asparagine to assimilate nitrogen and resist acid stress
during infection, PLOS Pathogens, 10, e1003928, pp. 1–14.
33. Jenkins, S., Fischer, S. M., Chen, L., and Sana, T. R. (2013). Global LC/MS
metabolomics profiling of calcium stressed and immunosuppressant
treated Saccharomyces cerevisiae, Metabolites, 3, pp. 1102–1117.
34. Pesek, J. J., Matyska, M. T., Loo, J. A., Fischer, S. M., and Sana, T. R.
(2009). Analysis of hydrophilic metabolites in physiological fluids by
HPLC-MS using a silica hydride-based stationary phase, J. Sep. Sci., 32,
pp. 2200–2208.
35. Putluria, N., Shojaiegn, A., and Vasu, V. T. (2011). Metabolic profiling
reveals potential markers and bioprocesses altered in bladder
cancer progression, Cancer Res., 15, pp. 7376–7386.
36. Hellmuth, C., Koletzko, B., and Peissner, W. (2011). Aqueous
normal phase cysteine and methionine by liquid chromatography-
tandem mass spectrometry, J. Chromatogr. B, 879, pp. 83–89.
37. Gouzy, A., and Larrouy-Maumus, G. (2013). Mycobacterium tuberculosis
nitrogen assimilation and host colonization require aspartate,
Nat. Chem. Biol., 9, pp. 674–676.
38. Sana, T. R., Gordon, D. B., Fischer, S. M., Tichy, S. E., Kitagawa, N., Cai, C.,
Gosnell, W. L., and Chang, S. P. (2013). Global mass spectrometry based
206 Aqueous Normal-Phase Chromatography Using Type C Silica Columns
Christopher Pohl
Chemistry R&D, Thermo Fisher Scientific,
1228 Titan Way, Sunnyvale, California 94085, USA
[email protected]
9.1 Introduction
Ion chromatography is a highly specialized branch of liquid
chromatography, generally making use of columns specifically
designed for ion chromatography and intended to be used with ion
chromatography instrumentation. In general, ion chromatography
is performed with a specialized pumping system designed for the
delivery of highly acidic and highly basic mobile phases without risk
of corrosion as a consequence of the extreme pH associated with
such mobile phases. Also, although there are notable exceptions,
ion chromatography is commonly practiced while making use
of a conductivity detector. Because a conductivity detector is a
bulk property detector [1], the conductivity signal arising from
conductive mobile phases is highly susceptible to environmental
effects. For that reason, ion chromatography is usually practiced
in a temperature-controlled environment to minimize thermal
effects on the conductivity signal. In addition, the signal-to-noise
ratio associated with detection of ionic species against a background
conductivity of the mobile phase is relatively poor given the fact
that the concentration of the mobile phase is typically much
greater than the concentration of analytes being detected in the
presence of the mobile phase conductivity. For that reason, most
ion chromatography is practiced making use of a suppressor
device [2, 3]. The suppressor device is unique among analytical
techniques in that it reduces or eliminates the mobile phase
conductivity while simultaneously enhancing the conductivity of
the analyte ion. While the operation of the suppressor device is
beyond the scope of this chapter, it is relevant to the design and
architecture of ion chromatography columns. Because the best
conductivity detector performance is achieved when a strong
acid mobile phase is used for the analysis of cations and an alkali
metal hydroxide mobile phase is used for the analysis of anions,
this necessitates the use of stationary phase materials compatible
with long-term exposure to mobile phases at extreme pH. Thus,
while silica-based materials are the mainstay of HPLC, the poor
hydrolytic stability of silica at high pH and the poor ligand stability
at low pH render silica-based stationary phases a poor choice
for ion chromatography. In contrast, polymeric stationary phases,
especially polymers based on styrenic monomers are exceptionally
stable at high pH and low pH, making columns derived from such
monomers an excellent choice for use in ion chromatography.
For that reason, the bulk of the discussion in this chapter will
focus on polymeric stationary phases suitable for use in ion
chromatography.
Cation-Exchange Phases 211
References
Laila Kott
Xenon, Burnaby, B.C., Canada
[email protected]
10.1 Introduction
The use of ion exchangers has been known throughout history.
Aristotle reported the desalination of seawater using sands and
soils, but the widespread use of synthetic ion exchangers did not
come about until scientists discovered the ion-exchange capabilities
Column
Injector LCpump Detector(s)
andoven
Eluant Columnand
j
Injector p p
LCpump pp
Suppressor ()
Detector(s)
generator
t oven
10.2.1 Columns
Unlike liquid chromatography columns, ion-exchange columns
are predominantly polymer based and due to the nature of the
analyte (ions) are designed to be stable under more extreme pH
conditions than typical (U)HPLC columns. This is in part due
to the elution of analytes in IC being accomplished by acids or
bases, typically methanesulfonic acid, potassium hydroxide or
potassium carbonate. Since the predominant mode of detection for
IC is conductivity, it is important to keep the overall ionic strength
of the eluant as low as possible, which in turn requires less
suppression of the signal. As such, columns with low ion-exchange
capacities are preferred for standard pharmaceutical analyses,
as they require less eluant and therefore, also less suppression
[6]. In IC the type and design of the stationary phase in particular
plays a significant role in controlling selectivity due to the range
of stationary phase chemistries available; this being in contrast
to reversed-phase liquid chromatography, which is dominated
by similar octadecylsilica-based stationary phases [7].
Column selection begins first and foremost with type of
analysis, anionic or cationic. Figure 10.2 shows a representation
of the functional groups found on both the cationic and anionic
column types.
240 Ion Chromatography
10.2.2 Eluants
In ion chromatography, once the ion is retained by the stationary
phase, it falls to the eluant to disrupt the interaction of the
sample ion and the stationary phase to elute the ion. Some ion
chromatographs come with the ability to generate the eluants
in situ/inline, whereas other systems require that eluants are
prepared as they would be for liquid chromatographic separations,
at fixed concentrations. Since pH and ion concentration play such
an important role in the IC elution process, special care must be
taken when preparing IC eluants.
For eluants prepared offline (not using the eluant generators),
they must be prepared with fresh de-ionized water, with
sufficiently low conductivity (18.2 MW/cm at 25°C is typical) and
should be free of organic compounds as not to compromise IC
separation sensitivity. If unsure of the water quality or conductivity,
boil the water to remove any dissolved CO2, which can convert
to carbonate and change the background conductivity of the
eluant. Other considerations when preparing eluants are as
follows:
Components of the System 243
Figures 10.3 and 10.4 show a schematic of how the cation and
anion suppressors exchange out the highly charged eluant and
use the detector effluent to self-regenerate. The detector effluent,
which has only a few anions in it, is directed back through the
suppressor as a regenerant. The water in the regenerant aides in
the suppression and regeneration of the suppressor.
Figure 10.3 Schematic showing the ionic flows through the suppressor to
the detector and then from the detector back through the suppressor
for regeneration: Cation.
10.2.4 Detectors
10.2.4.1 Conductivity detector (CD)
The conductivity detector (CD) is the most common detector for
IC analysis, as all ions are electrically conducting, making this a
Components of the System 247
Figure 10.4 Schematic showing the ionic flows through the suppressor to
the detector and then from the detector back through the suppressor
for regeneration: Anion.
V=R (10.2)
d d
k= = G = KG , (10.4)
AR A
Components of the System 249
where K is the cell constant for the detector, with units of cm–1.
Since conductance of electrolytes will vary with concentration
(C), the equivalent or molar conductance (L) is defined per
1000 cm3 as
1000k (10.5)
L=
C
Combining Eqs. 10.4 and 10.5, we get conductance defined as:
LC
G= (10.6)
1000K
( l + l )C (10.7)
G=
1000K
(272.1) 0.005
G=
10000
G = 0.00013605 S = 136.05 mS
h K
G25 = GT T , (10.8)
h25
where G25 and GT are the conductances at 25°C and T°C and
h25 and hT are the viscosities of water at 25°C and T°C. K is a
constant.
To ensure reproducible results, it becomes important to
control the temperature of the detector.
͊
Figure 10.5 Chromatogram showing the change in the baseline as the eluant gradient changes the effluent conductivity
above the suppressor setting for 1 mM hydroxide, which was set 7 mA.
Method Development 257
10.5 Applications
Most ion chromatographic applications in the pharmaceutical
industry are currently counterion analyses, analyses of ionic
impurities or detection of certain genotoxic impurities, which
often require the detection and separation of one or two ions
only, making method development fairly simple. However, as drug
development expands to new modalities, including compounds
such as nucleotides, amino acids, peptides, polypeptides, and
proteins, all of which are now becoming part of or in fact, the
active pharmaceutical ingredient, ion chromatography will be
further challenged with their analyses. Chapter 9 and Paull and
Nesterenko [7] have compiled lists of commercially available
columns for amino acid and analysis of other biologics, showing
that column developers are expanding on their stationary phase
chemistries, to keep up with the trend towards analyzing more
biologics.
Along with the newer horizons in drug development, there
is continued scrutiny on the environment. There are many EPA
and USP methods currently using ion chromatography and that
number is likely to rise. The monitoring of wastewaters from
sewage plants, mining operations, water runoff from farming,
air quality monitoring are all areas where ion chromatography
is being used. As acceptable limits for human exposure of metals
and other ionic toxins in foodstuffs and drinking water are
decreasing so must the limits of detection in the analytical
methodology. This may lead to more interest in IC-MS methodology,
260 Ion Chromatography
Acknowledgments
The author would like to thank Anthony Landry, Jelena Petrovic,
and Amy McCusker for their help.
References
11.1 Introduction
Gas chromatography (GC) is a separation technique that has been
widely used for the analysis of volatile and semi-volatile chemical
constituents. GC has become a workhorse separation method in
a variety of applications, including petroleum analysis [1], food
sample testing [2], environmental sample screening [3], and
drug analysis [4]. The remarkable work on GC in the 1940s by
A. J. P. Martin and R. L. M. Synge is considered a milestone in the
evolution of modern GC techniques.
Advances in column technology have played a significant role
in transforming the face of GC. Before optimizing any GC analysis,
the selection of an appropriate column type as well as stationary
phase is an important prerequisite. GC columns are broadly
divided into two classes, namely, packed columns and open tubular
columns. In packed columns, a solid material (possessing high
surface area) or a liquid coated on the solid support typically
Introduction 265
phase materials that can impart the desired selectivity and high
thermal stability to satisfy the increasing demands of complex
samples. Today, a wide range of GC columns are available with
different chemical selectivities and thermal properties. As a result,
selection of an appropriate stationary phase that can provide high
selectivity as well as good separation efficiency is an important
task and holds significant impact in GC analysis.
This book chapter provides an overview on the various column
fabrication procedures and different types of stationary phases
utilized in GC analysis. The first section of the chapter focuses on
fundamentals involved in column preparation, evaluation, and their
influence on gas chromatographic separation. The second section
of this chapter encapsulates the properties, applications, and
limitations of current stationary phases such as poly(siloxanes) and
polyethylene glycols. Finally, emerging stationary phases including
ionic liquids (ILs) and PILs will be discussed along with their
advancements with respect to improved chemical selectivities and
thermal stabilities for providing solutions to separating complex
sample.
System constants
Stationary
phase e s a b l Ref.
cPoly(dimethyltrifluoropropylsiloxane).
Stationary phase Commercial identity column Sample types Sample matrix Ref.
100% PDMS AT-1, BP-1, CB, CP-sil-5, DB-1, DB-1 Steroids, organochlorine Air; ginger extract; [81–87]
(non-polar) Equility-1, HP-1 ms, Optima-1 pesticides, mixture of essential crude oil; soil; shale
ms, oils, aromatic and saturated gas; tea.
DB-1 ms, ZB-, AT-1 ms, Sol-gel- hydrocarbons, pyrethroid
ms, HP-1, MDN-1, Optima-1, pesticides, chemical warfare
OV-1, agents
OV-101, SE-30, Rtx-1, SPB-1,
VF-1 ms
5% phenyl, 95% AT-5, AT-5 ms, BP-5, BPX-5 ms, DB-5 Alkylphenol, parabens, aroma Urine, blood; breast [76–80]
dimethysiloxane CP-Sil-8, CP-Sil-8, CB-ms, compounds, polychlorinated milk; particulate
(non-polar) DB-5, Equity-5, HP-5, HP-5 dibenzofurans, polychlorinated matter, pomegranate;
ms, MDN-5, MDN-5 s, MTX-5, dibenzo-p-dioxins, agricultural soils;
Optima-5, Optima-5 ms, OV-23, polybrominated dibenzo-p- fresh water.
Permabond SE-52, PB-5, RTX-5, dioxins, essential oils,
RTX-5 ms, SPB-5, Ultra-2, ZB-5, Polychlorinated biphenyls,
007-5 unsaturated isoprenoids
6% Cyanopropyl AT-624, BP-624, CP-624, CP-30, DB-624 Low-molecular-weight silanol Biogas; gas; [92–97]
phenyl, 94% DB-624, DB-30, DB-VRX, HP- and siloxanes; volatile organic municipal
dimethylsiloxane 624, compounds (VOCs); residual sludge sample;
(mid-polar) HP-30, Optima-624, Optima-30, solvents; odor-active sulfur and pharmaceutical drugs
PE-624, PE-30, RTX-624, RTX- nitrogen compounds; flavoring and intermediates;
Stationary Phase Materials
(Continued)
ZB-624,007-624, 007-502 pork liver powder.
277
278
Representative
Stationary phase Commercial identity column Sample types Sample matrix Ref.
35% phenyl 65% AT-35, BPX-35, DB-35, HP-35, DB-35 Pesticide residues; polycyclic Tobacco; fruit [88–91]
dimethylsiloxane MDN-35, RTX-35, SPB-35 aromatic compounds and samples; cigarette
(mid-polar) tobacco specific N-nitrosamines; smoke; urine; grape
methylmalonic acid (biomarker leaves.
for organic aciduria); plant
primary metabolite profiling
14% cyanopropyl BPX-10, CB-1701, CP-Sil-19CB, DB-1701 Organophosphorus pesticides; Plant; tobacco [101–
phenyl 86% DB-1701, DB-1701P, HP-1701, organochlorine pesticides; products; skimmed 103]
dimethylsiloxane Optima-1701, Rtx-1701, trihalomethanes; trimethyl silyl milk; drinking water;
(mid-polar) SPB-1701, ZB-1701, 007-1701 derived monosaccharides plant.
50% Cyanopropyl BP-225, BPx-50, HP-225, DB-225 Pectic polysaccharides; mono Fruit samples; [98–100]
Stationary Phases for Gas Chromatography Method Development
phenyl 50% DB-225, Optima-225, OV-225, unsaturated fatty acid profiling plasma; serum.
dimethylsiloxane RTX-50, RTX-225, SP-2250,
(polar) ZB-1701, 007-225
Stationary Phase Materials 279
Cations Anions
11.5.1.2 Viscosity
The viscosity of the IL is an important property that influences
the film stability of the stationary phase, particularly at high
temperatures. Owing to long-range columbic forces and
intermolecular interactions (e.g., hydrogen bonding), ILs exhibit
at least two to three orders of magnitude higher viscosities than
organic liquids at room temperature. Within the class of 1-alkyl-
3-methylimidazolium-based ILs, increasing the length of the alkyl
side chain causes a significant increase in viscosity [21, 106].
Effective charge delocalization tends to reduce the viscosity of
ILs, whereas substituents that reduce ion-symmetry often result
in increased viscosity. Halide-based anions enhance the viscosity
of the IL by participating in hydrogen bonding interactions
while [NTf–2] anions tend to reduce the viscosity due to charge
delocalization and poor hydrogen bonding capabilities [32, 107].
Other approaches to increase the viscosity involve incorporation
286 Stationary Phases for Gas Chromatography Method Development
Table 11.3 Chemical composition, MAOT, and system constants of selected ionic liquid-based stationary phases
1-butyl-3-(2-hydroxycyclohexyl) imidazolium –0.29 0.67 3.11 0.31 0.49 Analysis of FAMES, alkanes, and [130]
[BF4]; 250 esters
(Continued)
291
292
References
7. Herraiz, T., Reglero, G., Herraiz, M., Alonso, R., Cabezudo, M. D. (1987).
Micropacked columns: A suitable alternative to very thick capillary
columns, J. Chromatogr. A, 388, pp. 325–333.
8. Ettre, L. (1987). MJE Golay and the invention of open‐tubular (capillary)
columns, J. Sep. Sci., 10, pp. 221–230.
9. Poole, C. F. (2003). The Essence of Chromatography, 1st ed. (Elsevier).
10. Desty, D. (1987). The history and potentiality of capillary columns
in gas chromatography, J. Chromatogr. Sci., 25, pp. 552–563.
11. Dandeneau, R. D., and Zerenner, E. H. (1979). An investigation of
glasses for capillary chromatography, J. High Resolut. Chromatogr.,
2, pp. 351–356.
12. Peters, T. L., Nestrick, T. J., Lamparski, L. L. (1982). Etching borosilicate
glass capillary columns, Anal. Chem., 54, pp. 2397–2398.
13. Grob, K., and Grob, G. (1977). The barium carbonate procedure for
the preparation of glass capillary columns; further informations
and developments, Chromatographia, 10, pp. 181–187.
14. Grob, K. (1980). Persilylation of glass capillary columns. Part 4:
Discussion of parameters, J. High Resolut. Chromatogr., 3, pp. 493–
496.
15. Grob, K., Grob, G., Blum, W., Walther, W. (1982). Preparation on
inert glass capillary columns for gas chromatography: A revised,
comprehensive description, J. Chromatogr. A, 244, pp. 197–208.
16. Hetem, M., Rutten, G., Vermeer, B., Rijks, J., van de Ven, L., de Haan,
J., Cramers, C. (1989). Deactivation with polymethylhydrosiloxane:
A comparative study with capillary gas chromatography and solid-
state 29Si nuclear magnetic resonance spectroscopy, J. Chromatogr.
A, 477, pp. 3–24.
17. Novotný, M., and Bartle, K. (1974). Preparation of thick-film glass
capillary columns by the dynamic coating procedure, J. Chromatogr. A,
93, pp. 405–411.
18. Bouche, J., and Verzele, M. (1968). A static coating procedure for
glass capillary columns, J. Chromatogr. Sci., 6, pp. 501–505.
19. Wang, D., Chong, S. L., Malik, A. (1997). Sol−gel column technology for
single-step deactivation, coating, and stationary-phase immobilization
in high-resolution capillary gas chromatography, Anal. Chem., 69,
pp. 4566–4576.
20. Poole, C. F., and Poole, S. K. (2012). Chromatography Today, 5th ed.
(Elsevier).
References 297
46. Mayer, B. X., Zöllner, P., Lorbeer, E., Rauter, W. (2002). A new
75% diphenyl, 25% dimethyl polysiloxane coated on fused silica
capillary columns for high temperature gas chromatography, J. Sep.
Sci., 25, pp. 60–66.
47. Bradshaw, J. S., Schirmer, M. M., Juvancz, Z., Markides, K. E., Lee, M. L.
(1991). New polar polysiloxane stationary phases containing
cyano, nitrophenyl and 8-quinolinyl units attached to diethylene
oxide side groups, J. Chromatogr. A, 540, pp. 279–291.
48. Martin, S. D., Poole, C. F., Abraham, M. H. (1998). Synthesis and
gas chromatographic evaluation of a high-temperature hydrogen-
bond acid stationary phase, J. Chromatogr. A, 805, pp. 217–235.
49. Blomberg, L. G. (1990). Current aspects of stationary phase
immobilization in open tubular column chromatography, J. Microcolumn
Sep., 2, pp. 62–68.
50. Bottoli, C. B., Chaudhry, Z. F., Fonseca, D. A., Collins, K. E., Collins, C. H.
(2002). Poly (alkylmethylsiloxanes) thermally immobilized on silica
as stationary phases for high-performance liquid chromatography,
J. Chromatogr. A, 948, pp. 121–128.
51. Markides, K., Blomberg, L., Buijten, J., Wännman, T. (1983).
Cyanosilicones as stationary phases in gas chromatography: II. Gum
and rubber phases on fused silica, J. Chromatogr. A, 267, pp. 29–38.
52. Wright, B. W., Peaden, A., Lee, M. L., Stark, T. J. (1982). Free radical
cross-linking in the preparation of non-extractable stationay
phases for capillary gas chromatography, J. Chromatogr. A, 248,
pp. 17–34.
53. Schomburg, G., Husmann, H., Ruthe, S., Herraiz, M. (1982). Crosslinking
of alkylpolysiloxane filsm on various types of glass surfaces including
fused silica using g-radiation of a60cobalt-source. Comparison to
crosslinking by thermal peroxid treatment, Chromatographia, 15,
pp. 599–610.
54. Hubball, J., DiMauro, P., Barry, E., Lyons, E., George, W. (1984).
Developments in crosslinking of stationary phases for capillary gas
chromatography by cobalt-60 gamma radiation, J. Chromatogr. Sci., 22,
pp. 185–191.
55. Zhou, X.-C., Yan, H., Chen, Y.-Y., Wu, C.-Y., Lu, X.-R. (1996). Chiral crown
ether-anchored polysiloxanes as capillary gas chromatography
stationary phases, J. Chromatogr. A, 753, pp. 269–277.
56. Schurig, V. (1984). Gas chromatographic separation of enantiomers
on optically active metal-complex-free stationary phases. New
analytical methods (24), Angew. Chem. Int. Ed., 23, pp. 747–765.
300 Stationary Phases for Gas Chromatography Method Development
57. Ruderisch, A., Pfeiffer, J., Schurig, V. (2003). Mixed chiral stationary
phase containing modified resorcinarene and β-cyclodextrin
selectors bonded to a polysiloxane for enantioselective gas
chromatography, J. Chromatogr. A, 994, pp. 127–135.
58. Zhang, H., Dai, R., Ling, Y., Wen, Y., Zhang, S., Fu, R., Gu, J. (1997).
Resorcarene derivative used as a new stationary phase for capillary
gas chromatography, J. Chromatogr. A, 787, pp. 161–169.
59. Berthod, A., Li, W., Armstrong, D. W. (1992). Multiple enantioselective
retention mechanisms on derivatized cyclodextrin gas chromato-
graphic chiral stationary phases, Anal. Chem., 64, pp. 873–879.
60. Schurig, V., Schmalzing, D., Schleimer, M. (1991). Enantiomer
separation on immobilized chirasil-metal and chirasil-dex by gas
chromatography and supercritical fluid chromatography, Angew.
Chem. Int. Ed., 30, pp. 987–989.
61. Schurig, V. (2002). Chiral separations using gas chromatography,
Trends Anal. Chem.; TrAC, 21, pp. 647–661.
62. Levkin, P. A., Levkina, A., Schurig, V. (2006). Combining the
enantioselectivities of l-valine diamide and permethylated β-
cyclodextrin in one gas chromatographic chiral stationary phase,
Anal. Chem., 78, pp. 5143–5148.
63. Ding, J., Welton, T., Armstrong, D. W. (2004). Chiral ionic liquids
as stationary phases in gas chromatography, Anal. Chem., 76,
pp. 6819–6822.
64. König, W. A., Lutz, S., Mischnick-Lübbecke, P., Brassat, B., Wenz, G.
(1988). Cyclodextrins as chiral stationary phases in capillary gas
chromatography I. Pentylated α-cyclodextrin, J. Chromatogr. A, 47,
pp. 193–197.
65. Armstrong, D. W., Li, W., Chang, C. D., Pitha, J. (1990). Polar-liquid,
derivatized cyclodextrin stationary phases for the capillary gas
chromatography separation of enantiomers, Anal. Chem., 62,
pp. 914–923.
66. Nowotny, H. P., Schmalzing, D., Wistuba, D., Schurig, V. (1989).
Extending the scope of enantiomer separation on diluted methylated
β-cyclodextrin derivatives by high-resolution gas chromatography,
J. High Resolut. Chromatogr., 12, pp. 383–393.
67. Blum, W., and Aichholz, R. (1990). Gas chromatographic enantiomer
separation on tert-butyldimethylsilylated β-cyclodextrin diluted in
PS-086. A simple method to prepare enantioselective glass capillary
columns, J. High Resolut. Chromatogr., 13, pp. 515–518.
References 301
68. Fischer, P., Aichholz, R., Bölz, U., Juza, M., Krimmer, S. (1990).
Permethyl-β-cyclodextrin, chemically bonded to polysiloxane: A
chiral stationary phase with wider application range for enantiomer
separation by capillary gas chromatography, Angew. Chem. Int. Ed.,
29, pp. 427–429.
69. Sicoli, G., Pertici, F., Jiang, Z., Jicsinszky, L., Schurig, V. (2007). Gas-
chromatographic approach to probe the absence of molecular
inclusion in enantioseparations by carbohydrates. Investigation of
linear dextrins (“acyclodextrins”) as novel chiral stationary phases,
Chirality, 19, pp. 391–400.
70. Huang, K., Zhang, X., Armstrong, D. W. (2010). Ionic cyclodextrins
in ionic liquid matrices as chiral stationary phases for gas
chromatography, J. Chromatogr. A, 1217, pp. 5261–5273.
71. Conder, J. R., Fruitwala, N. A., Shingari, M. K. (1983). Thermal
decomposition of polyethylene glycol 20 m and essential oils in
gas—liquid chromatography and the effect of traces of oxygen, J.
Chromatogr. A, 269, pp. 171–178.
72. Sandra, P., and Van Roelenbosch, M. (1981). Mixed phases of
Superox 20M and OV-1 in fused silica and glass capillary gas
chromatography, Chromatographia, 14, pp. 345–350.
73. Lindsay Smith, J. R., and Waddington, D. J. (1969). Gas chromatographic
analysis of aliphatic amines: The use of ethylene glycols as stationary
phases, J. Chromatogr. A, 42, pp. 183–194.
74. Azzouz, A., Rascón, A. J., Ballesteros, E. (2016). Simultaneous
determination of parabens, alkylphenols, phenylphenols, bisphenol
A and triclosan in human urine, blood and breast milk by continuous
solid-phase extraction and gas chromatography–mass spectrometry,
J. Pharm. Biomed. Anal., 119, pp. 16–26.
75. He, D., Simoneit, B. R. T., Xu, Y., Jaffé, R. (2016). Occurrence of
unsaturated C25 highly branched isoprenoids (HBIs) in a freshwater
wetland, Org. Geochem., 93, pp. 59–67.
76. Ren, Z., Xiao, X., Chen, D., Bi, X., Huang, B., Liu, M., Hu, J., Peng,
P. A., Sheng, G., Fu, J. (2014). Halogenated organic pollutants in
particulate matters emitted during recycling of waste printed
circuit boards in a typical e-waste workshop of Southern China,
Chemosphere, 94, pp. 143–150.
77. Tripathi, J., Chatterjee, S., Gamre, S., Chattopadhyay, S., Variyar, P. S.,
Sharma, A. (2014). Analysis of free and bound aroma compounds
of pomegranate (Punica granatum L.), LWT-Food. Sci. Technol., 59,
pp. 461–466.
302 Stationary Phases for Gas Chromatography Method Development
78. Zhu, Z.-C., Chen, S.-J., Zheng, J., Tian, M., Feng, A.-H., Luo, X.-J., Mai,
B.-X. (2014). Occurrence of brominated flame retardants (BFRs),
organochlorine pesticides (OCPs), and polychlorinated biphenyls
(PCBs) in agricultural soils in a BFR-manufacturing region of North
China, Sci. Total Environ., 481, pp. 47–54.
79. An, J., Zhu, B., Wang, H., Li, Y., Lin, X., Yang, H. (2014). Characteristics
and source apportionment of VOCs measured in an industrial
area of Nanjing, Yangtze River Delta, China, Atmos. Environ., 97,
pp. 206–214.
80. Hou, X., Zheng, X., Zhang, C., Ma, X., Ling, Q., Zhao, L. (2014).
Ultrasound-assisted dispersive liquid–liquid microextraction based
on the solidification of a floating organic droplet followed by gas
chromatography for the determination of eight pyrethroid pesticides
in tea samples, J. Chromatogr. B, 969, pp. 123–127.
81. López, L. (2014). Study of the biodegradation levels of oils from the
Orinoco Oil Belt (Junin area) using different biodegradation scales,
Org. Geochem., 66, pp. 60–69.
82. López, L., Lo Mónaco, S., Volkman, J. K. (2015). Evidence for mixed
and biodegraded crude oils in the Socororo field, Eastern Venezuela
Basin, Org. Geochem., 82, pp. 12–21.
83. Maguire-Boyle, S. J., and Barron, A. R. (2014). Organic compounds in
produced waters from shale gas wells, Environ. Sci. Process. Impact.,
16, pp. 2237–2248.
84. Nagashima, H., Kondo, T., Nagoya, T., Ikeda, T., Kurimata, N., Unoke,
S., Seto, Y. (2015). Identification of chemical warfare agents from
vapor samples using a field-portable capillary gas chromatography/
membrane-interfaced electron ionization quadrupole mass
spectrometry instrument with Tri-Bed concentrator, J. Chromatogr. A,
1406, pp. 279–290.
85. Yeh, H.-Y., Chuang, C.-H., Chen, H.-C., Wan, C.-J., Chen, T.-L., Lin, L.-Y.
(2014). Bioactive components analysis of two various gingers
(Zingiber officinale Roscoe) and antioxidant effect of ginger extracts,
LWT-Food. Sci. Technol., 55, pp. 329–334.
86. Aghamohammadi, M., Shahdousti, P., Harooni, B. (2016).
Ultrasound-assisted emulsification microextraction followed
by gas chromatography–flame ionization detection for urinary
methylmalonic acid determination, Microchem. J., 124, pp. 188–194.
87. Harb, J., Alseekh, S., Tohge, T., Fernie, A. R. (2015). Profiling of primary
metabolites and flavonols in leaves of two table grape varieties
References 303
97. Müller, D. C., Degen, C., Scherer, G., Jahreis, G., Niessner, R., Scherer,
M. (2014). Metabolomics using GC–TOF–MS followed by subsequent
GC–FID and HILIC–MS/MS analysis revealed significantly altered
fatty acid and phospholipid species profiles in plasma of smokers,
J. Chromatogr. A, 966, pp. 117–126.
98. Watkins, B. A., Kim, J., Tamez, H., Wenger, J., Thadhani, R., Friedman, A. N.
Serum phospholipid fraction of polyunsaturated fatty acids is the
preferred indicator for nutrition and health status in hemodialysis
patients, J. Nutr. Biochem., 38, pp. 18–24.
99. Qi, D., Fei, T., Sha, Y., Wang, L., Li, G., Wu, D., Liu, B. (2014). A novel fully
automated on-line coupled liquid chromatography–gas chromato-
graphy technique used for the determination of organochlorine
pesticide residues in tobacco and tobacco products, J. Chromatogr. A,
1374, pp. 273–277.
100. Siddique, A., Saied, S., Mumtaz, M., Hussain, M. M., Khwaja, H. A. (2015).
Multipathways human health risk assessment of trihalomethane
exposure through drinking water, Ecotoxicol. Environ. Saf., 116,
pp. 129–136.
101. Yang, Y., Kong, W., Zhao, L., Xiao, Q., Liu, H., Zhao, X., Yang, M. (2015).
A multiresidue method for simultaneous determination of 44
organophosphorous pesticides in Pogostemon cablin and related
products using modified QuEChERS sample preparation procedure
and GC–FPD, J. Chromatogr. B, 974, pp. 118–125.
102. Hallett, J. P., and Welton, T. (2011). Room-temperature ionic
liquids: Solvents for synthesis and catalysis. 2, Chem. Rev., 111,
pp. 3508–3576.
103. Ho, T. D., Zhang, C., Hantao, L. W., Anderson, J. L. (2014). Ionic liquids
in analytical chemistry: Fundamentals, advances, and perspectives,
Anal. Chem., 86, pp. 262–285.
104. Coker, T. G., Ambrose, J., Janz, G. J. (1970). Fusion properties of
some ionic quaternary ammonium compounds, J. Am. Chem. Soc., 92,
pp. 5293–5297.
105. Gordon, J. E. (1965). Fused organic salts. IV.1a characterization of
low-melting quaternary ammonium salts. Phase equilibria for
salt-salt and salt-nonelectrolyte systems. Properties of the liquid
salt medium, J. Am. Chem. Soc., 87, pp. 4347–4358.
106. Poole, C. F. (2004). Chromatographic and spectroscopic methods
for the determination of solvent properties of room temperature
ionic liquids, J. Chromatogr. A, 1037, pp. 49–82.
References 305
107. Bonhôte, P., Dias, A.-P., Papageorgiou, N., Kalyanasundaram, K., Grätzel,
M. (1996). Hydrophobic, highly conductive ambient-temperature
molten salts, Inorg. Chem., 35, pp. 1168–1178.
108. Shashkov, M. V., and Sidelnikov, V. N. (2012). Mass spectral
evaluation of column bleeding for imidazolium-based ionic liquids
as GC liquid phases, Anal. Bioanal. Chem. 403, pp. 2673–2682.
109. Huddleston, J. G., Visser, A. E., Reichert, W. M., Willauer, H. D., Broker,
G. A., Rogers, R. D. (2001). Characterization and comparison of
hydrophilic and hydrophobic room temperature ionic liquids
incorporating the imidazolium cation, Green Chem., 3, pp. 156–164.
110. Kroon, M. C., Buijs, W., Peters, C. J., Witkamp, G.-J. (2007).
Quantum chemical aided prediction of the thermal decomposition
mechanisms and temperatures of ionic liquids, Thermochim.
Acta, 465, pp. 40–47.
111. Lebga-Nebane, J. L., Rock, S. E., Franclemont, J., Roy, D., Krishnan, S.
(2012). Thermophysical properties and proton transport mechanisms
of trialkylammonium and 1-alkyl-1H-imidazol-3-ium protic ionic
liquids, Ind. Eng. Chem. Res., 51, pp. 14084–14098.
112. Poole, C. F., and Poole, S. K. (2011). Ionic liquid stationary phases for
gas chromatography, J. Sep. Sci., 34, pp. 888–900.
113. Payagala, T., Zhang, Y., Wanigasekara, E., Huang, K., Breitbach, Z.
S., Sharma, P. S., Sidisky, L. M., Armstrong, D. W. (2009). Trigonal
tricationic ionic liquids: A generation of gas chromatographic
stationary phases, Anal. Chem., 81, pp. 160–173.
114. Law, G., and Watson, P. R. (2001). Surface tension measurements
of N-alkylimidazolium ionic liquids, Langmuir, 17, pp. 6138–6141.
115. Kilaru, P., Baker, G. A., Scovazzo, P. (2007). Density and surface
tension measurements of imidazolium-, quaternary phosphonium-,
and ammonium-based room-temperature ionic liquids: Data and
correlations, J. Chem. Eng. Data, 52, pp. 2306–2314.
116. Lantz, A. W., Pino, V., Anderson, J. L., Armstrong, D. W. (2006).
Determination of solute partition behavior with room-temperature
ionic liquid based micellar gas–liquid chromatography stationary
phases using the pseudophase model, J. Chromatogr. A, 1115,
pp. 217–224.
117. Armstrong, D. W., He, L., Liu, Y.-S. (1999). Examination of ionic liquids
and their interaction with molecules, when used as stationary
phases in gas chromatography, Anal. Chem., 71, pp. 3873–3876.
306 Stationary Phases for Gas Chromatography Method Development
118. Zhao, Q., Eichhorn, J., Pitner, W. R., Anderson, J. L. (2009). Using the
solvation parameter model to characterize functionalized ionic
liquids containing the tris (pentafluoroethyl) trifluorophosphate
(FAP) anion, Anal. Bioanal. Chem., 395, pp. 225–234.
119. Breitbach, Z. S., and Armstrong, D. W. (2008). Characterization
of phosphonium ionic liquids through a linear solvation energy
relationship and their use as GLC stationary phases, Anal. Bioanal.
Chem., 390, pp. 1605–1617.
120. Delmonte, P., Fardin-Kia, A. R., Kramer, J. K. G., Mossoba, M. M., Sidisky,
L., Tyburczy, C., Rader, J. I. (2012). Evaluation of highly polar ionic
liquid gas chromatographic column for the determination of the
fatty acids in milk fat, J. Chromatogr. A, 1233, pp. 137–146.
121. Zeng, A. X., Chin, S.-T., Nolvachai, Y., Kulsing, C., Sidisky, L. M., Marriott,
P. J. (2013). Characterisation of capillary ionic liquid columns for gas
chromatography–mass spectrometry analysis of fatty acid methyl
esters, Anal. Chim. Acta, 803, pp. 166–173.
122. Do, L., Liljelind, P., Zhang, J., Haglund, P. (2013). Comprehensive
profiling of 136 tetra- to octa-polychlorinated dibenzo-p-dioxins and
dibenzofurans using ionic liquid columns and column combinations,
J. Chromatogr. A, 1311, pp. 157–169.
123. Anderson, J. L., and Armstrong, D. W. (2005). Immobilized ionic
liquids as high-selectivity/high-temperature/high-stability gas
chromatography stationary phases, Anal. Chem., 77, pp. 6453–6462.
124. Hsieh, Y.-N., Ho, W.-Y., Horng, R. S., Huang, P.-C., Hsu, C.-Y., Huang, H.-H.,
Kuei, C.-H. (2007). Study of anion effects on separation phenomenon
for the vinyloctylimidazolium based ionic liquid polymer stationary
phases in GC, Chromatographia, 66, pp. 607–611.
125. Zhang, C., Park, R. A., Anderson, J. L. (2016). Crosslinked structurally-
tuned polymeric ionic liquids as stationary phases for the analysis
of hydrocarbons in kerosene and diesel fuels by comprehensive
two-dimensional gas chromatography, J. Chromatogr. A, 1440,
pp. 160–171.
126. Gonzalez-Alvarez, J., Arias-Abrodo, P., Puerto, M., Viguri, M. E.,
Perez, J., Gutierrez-Alvarez, M. D. (2013). Polymerized phosphonium-
based ionic liquids as gas chromatography stationary phases, RSC
Adv., 3, pp. 21377–21380.
127. Shashkov, M. V., and Sidel’nikov, V. N. (2012). Single cation ionic liquids
as high polarity thermostable stationary liquid phases for capillary
chromatography, Russ. J. Phys. Chem. A, 86, pp. 138–141.
Preparation and Beginning of Performance 307
1 + 6k +11k 2
hmin = dc , (12.1)
12(1 + k )2
Table 12.1 Benefits of modifying column I.D., length, and film thickness
Figure 12.2 Van Deemter curves for N2, He, and H2. Column I.D. = 0.25 mm,
analyte retention factor (k) = 10. Reproduced from [5].
Instrument and Column Optimization 319
Figure 12.3 Relationship between the carrier gas viscosity and temperature.
Reproduced from [5].
Helium Hydrogen
Column I.D. Linear velocity Flow rate Linear velocity Flow rate
(mm) (cm/s) (mL/min) (cm/s) (mL/min)
0.18 20–45 0.3–0.7 40–60 0.6–0.9
0.25 20–45 0.7–1.3 40–60 1.2–2.0
0.32 20–45 1.2–2.2 40–60 2.2–3.0
0.53 20–45 4.0–8.0 40–60 6.0–9.0
Figure 12.9 Mass spectra (electron ionization; EI) for: (A) penehyclidine
and (b) the internal standard, penehyclidine-d5. (A) EI-MS of m/z
315 175 for penehyclidine. (B) EI-MS of m/z 320 180 for penehyclidine-
d5. Reproduced from [35].
GC/Mass Spectrometry 335
Scheme 12.4 Ionization pathways in EI. (A) Electron expulsion, and (B)
electron abstraction.
Abundance Abundance
Element (A) (%) A+1 (%) A+2 Abundance (%)
1H 99.99 2H 0.01 — —
12C 98.91 13C 1.1 — —
14N 99.6 15N 0.4 — —
16O 99.76 17O 0.04 18O 0.02
19F 100 — — — —
28Si 92.2 29Si 4.7 30Si 3.1
31P 100 — — — —
32S 95.02 33S 0.76 34S 4.22
35Cl 75.77 — — 37Cl 24.23
79Br 50.5 — — 81Br 49.5
127I 100 — —
Source: Adapted from [47].
R + dB = C# – 12 H# + 12 N# + 1
Table 12.4 Common fragment losses from the electron ionization of the
molecular ion
Figure 12.13 (A) Original GC-HS method for in-process testing of residual
solvents, and (B) Fast HS-GC method for in-process testing of residual
solvents. Elution orders are the same of solvents peaks in (B) as (A). Credit:
Harry Patel, Umesh Patel, Tien Ho (Process Research & Development,
AbbVie, Inc.).
Considerations for Using GC in the Pharmaceutical Setting 349
Table 12.5 GC-HS conditions for the optimized (fast) GC-HS method
Carrier gas
(mL/min) 1.3 (constant) Temperature program
Split Ratio 30-1 35°C hold for 1.5 min, ramp at 25°C/
min to 90°C, hold for 0 min, ramp
Injector (°C) 210 35°C/min to 235°C, hold for 3.16 min
Detector (°C) 250
FID hydrogen flow 40
(mL/min) Vial incubation time: 10 min
FID air flow 40
(mL/min)
FID nitrogen 25 GC run time: 11 min/GC cycle time:
makeup (mL/min) 17 min
Column
GC-HS oven (°C) 100 624-type phase; 20 m × 0.18 mm ×
1.0 μm
GC-HS loop (°C) 110
GC-HS transfer 130
line (°C)
Figure 12.14 Fast GC direct inject method for residual solvent analysis.
Solvents: (1) Methanol, (2) Pentane, (3) Ethanol, (4) Diethylether, (5) Ethyl
Formate, (6) Acetonitrile, (7) Dichloromethane, (8) t-Butanol, (9) MTBE,
(10) n-Hexane, (11) 1-Propanol, (12) Nitromethane, (13) Methyl Ethyl
Ketone, (14) Ethyl Acetate, (15) Tetrahydrofuran, (16) Chloroform,
(17) Cyclohexane, (18) Carbon Tetrachloride, (19) 1,2-Dimethoxy ethane,
(20) Benzene, (21) Isopropyl Acetate, (22) 2-Methyl Tetrahydrofuran,
(23) n-Heptane, (24) Methyl Cyclohexane, (25) 1,4,-Dioxane, (26)
n-Propyl Acetate, (27) MIBK, (28) Pyridine, (29) Toluene, (30) 2-Hexanone,
(31) Dimethylformamide, (32) Chlorobenzene, (33) Ethyl benzene, (34)
Propylene Glycol/m-Xylene/p-Xylene, (35) o-Xylene, (36) DMSO, (37) NMP.
Credit: Harry Patel, Umesh Patel, Tien Ho (Process Research & Development,
AbbVie, Inc.).
Considerations for Using GC in the Pharmaceutical Setting 351
Table 12.6 GC conditions for fast GC direct injection method for residual
solvent analysis
References
24. Jumtee, K., Bamba, T., and Fukusaki, E. (2009) Fast GC-FID based
metabolic fingerprinting of Japanese green tea leaf for its quality
ranking prediction, J. Sep. Sci., 32, pp. 2296–2304.
25. Yan, X. (2006) Unique selective detectors for gas chromatography:
Nitrogen and sulfur chemiluminescence detectors, J. Sep. Sci., 29,
pp. 1931–1945.
26. Ragunathan, N., Krock, K. A., Klawun, C., Sasaki, T. A., and Wilkins,
C. L. (1999) Gas chromatography with spectroscopic detectors, J.
Chromatogr. A, 856, pp. 349–397.
27. Poole, C. F. (2015) Ionization-based detectors for gas chromatography,
J. Chromatogr. A, 1421, pp. 137–153.
28. Gunnar, T., Mykkänen, S., Ariniemi, K., and Lillsunde, P. (2004)
Validated semiquantitative/quantitative screening of 51 drugs
in whole blood as silylated derivatives by gas chromatography-
selected ion monitoring mass spectrometry and gas chromatography
electron capture detection, J. Chromatogr. B, 806, pp. 205–219.
29. Ho, T. D., Yehl, P. M., Chetwyn, N. P., Wang, J., Anderson, J. L., and
Zhong, Q. (2014) Determination of trace level genotoxic impurities
in small molecule drug substances using conventional headspace
gas chromatography with contemporary ionic liquid diluents and
electron capture detection, J. Chromatogr. A, 1361, pp. 217–228.
30. Kosjek, T., Heath, E., Petrović, M., and Barceló, D. (2007) Mass
spectrometry for identifying pharmaceutical biotransformation
products in the environment, TrAC, Trends Anal. Chem., 26,
pp. 1076–1085.
31. David, F., Alegre, M. R., Vanhoenacker, G., and Sandra, P. (2013)
Determination of genotoxic impurities in pharmaceuticals, LCGC Eur.,
26, pp. 31–34.
32. Girijan, R. (2012) Gas chromatography-mass spectrometry
technique and its applications in pharmaceutical industry, Chem.
Ind. Dig., 25, pp. 59–63.
33. Gillespie, T. A., and Winger, B. E. (2011) Mass spectrometry for
small molecule pharmaceutical product development: A review,
Mass Spectrom. Rev., 30, pp. 479–490.
34. Ji, C., Dean, J. L., Flores, R. A., and Boisvert, S. M. (2009) Quantitative
determination of co-eluting compounds by using extract ion
chromatogram in GC-MS, Chem. Educator., 14, pp. 212–214.
35. Xue, M., Yuan, S., Ruan, J., Qiao, J., Xu, Y., Zhang, Z., and Liu, K. (2006)
Determination of penehyclidine by gas chromatographic-mass
364 Gas Chromatography Method Development
Chromatography (Grob, R. L., and Barry, E. F., eds.), John Wiley & Sons,
Inc., Hoboken, New Jersey, pp. 339–401.
50. Smith, R. M. (2004) Understanding Mass Spectra: A Basic Approach,
2nd ed., John Wiley & Sons, Inc., Hoboken, New Jersey.
51. Zhang, L., Tang, C., Cao, D., Zeng, Y., Tan, B., Zeng, M., Fan, W.,
Xiao, H., and Liang, Y. (2013) Strategies for structure elucidation
of small molecules using gas chromatography-mass spectrometric
data, TrAC, Trends Anal. Chem., 47, pp. 37–45.
52. Hamilton, S. E., Rossington, M. D., and Bertrand, A. (2016) Development
of an automated headspace gas chromatography instrument for the
determination of residual solvents in pharmaceutical compounds
and reaction mixtures, Org. Proc. Res. Dev., 20, pp. 189–194.
53. Nacham, O., Ho, T. D., Anderson, J. L., and Webster, G. K. (2017)
Use of ionic liquids as headspace gas chromatography diluents for
the analysis of residual solvents in pharmaceuticals, J. Pharm. Biomed.
Anal., 145, pp. 879–886.
54. Belal, T. S., Awad, T., and Clark, C. R. (2014) Stability-indicating
determination of trimetazidine dihydrochloride in the presence of
two of its related substances using a direct GC/MS method, J. AOAC
Int., 97, pp. 1514–1518.
55. Belal, T. S., Abdel-Hay, K. M., and Clark, C. R. (2016) Selective
determination of dimenhydrinate in presence of six of its related
substances and potential impurities using a direct GC/MS method,
J. Adv. Res., 7, pp. 53–58.
56. van den Berg, J. J. M., Winterbourn, C. C., and Kuypers, F. A. (1993)
Hypochlorous acid-mediated modification of cholesterol and
phospholipid: Analysis of reaction products by gas chromatography-
mass spectrometry, J. Lipid Res., 34, pp. 2005–2012.
57. Skórka, M., Asztemborska, M., and Żukowski, J. (2005) Thermodynamic
studies of complexation and enantiorecognition processes of
monoterpenoids by α- and β-cyclodextrin in gas chromatography,
J. Chromatogr. A, 1078, pp. 136–143.
58. Fiamegos, Y. C., Nanos, C. G., and Stalikas, C. D. (2004) Ultrasonic-
assisted derivatization reaction of amino acids prior to their
determination in urine by using single-drop microextraction in
conjunction with gas chromatography, J. Chromatogr. B, 813,
pp. 89–94.
59. McGovern, T., and Jacobson-Kram, D. (2006) Regulation of genotoxic
and carcinogenic impurities in drug substances and products,
TrAC, Trends Anal. Chem., 25, pp. 790–795.
366 Gas Chromatography Method Development
60. Reddy, A. V. B., Jaafar, J., Umar, K., Majid, Z. A., Aris, A. B., Talib, J.,
and Madhavi, G. (2015) Identification, control strategies, and
analytical approaches for the determination of potential genotoxic
impurities in pharmaceuticals: A comprehensive review, J. Sep. Sci.,
38, pp. 764–779.
61. David, F., Alegre, M. R., Vanhoenacker, G., and Sandra, P. (2013)
Determination of genotoxic impurities in pharmaceuticals, LCGC Eur.,
pp. 31–34.
62. Elder, D. P., Delaney, E., Teasdale, A., Eyley, S., Reif, V. D., Jacq, K.,
Facchine, K. L., Oestrich, R. S., Sandra, P., and David, F. (2010) The
utility of sulfonate salts in drug development, J. Pharm. Sci., 99,
pp. 2948–2961.
63. Liu, D. Q., Sun, M., and Kord, A. S. (2010) Recent advances in trace
analysis of pharmaceutical genotoxic impurities, J. Pharm. Biomed.
Anal., 51, pp. 999–1014.
64. Alzaga, R., Ryan, R. W., Taylor-Worth, K., Lipczynski, A. M., Szucs, R.,
and Sandra, P. (2007) A generic approach for the determination
of residues of alkylating agents in active pharmaceutical ingredients
by in situ derivatization–headspace–gas chromatography–mass
spectrometry, J. Pharm. Biomed. Anal., 45, pp. 472–479.
65. Ho, T. D., Joshi, M. D., Silver, M. A., and Anderson, J. L. (2012)
Selective extraction of genotoxic impurities and structurally alerting
compounds using polymeric ionic liquid sorbent coatings in solid-
phase microextraction: Alkyl halides and aromatics, J. Chromatogr. A,
1240, pp. 29–44.
66. Byun, J., Henderson, J. P., and Heinecke, J. W. (2003) Identification
and quantification of mutagenic halogenated cytosines by gas
chromatography, fast atom bombardment, and electrospray ionization
tandem mass spectrometry, Anal. Biochem., 317, pp. 201–209.
67. Kataoka, H., Nishioka, S., Kobayashi, M., Hanaoka, T., and Tsugane,
S. (2002) Analysis of mutagenic heterocyclic amines in cooked food
samples by gas chromatography with nitrogen-phosphorus detector,
Bull. Environ. Contam. Toxicol., 69, pp. 682–689.
68. David, F., Jacq, K., Sandra, P., Baker, A., and Klee, M. S. (2010) Analysis
of potential genotoxic impurities in pharmaceuticals by two-
dimensional gas chromatography with Deans switching and
independent column temperature control using a low-thermal-mass
oven module, Anal. Bioanal. Chem., 396, pp. 1291–1300.
Chapter 13
Susanne Fabel
Thermo Fisher Scientific, Dornierstr. 4, D-82110 Germering, Germany
[email protected]
13.1 Introduction
The method transfer between conventional HPLC and UHPLC is
done either to accelerate the method or to gain more resolution
compared to the HPLC method. The opposite transfer from
UHPLC to HPLC could also be a requirement, for example, if in
the R&D department only UHPLC equipment is available and the
methods have to be transferred to HPLC equipment in the QC
laboratory. Conveniently, method transfer for UHPLC to HPLC can
be achieved by applying the same rules and principles for HPLC
to UHPLC methods. In this chapter, the focus is on the method
transfer from conventional HPLC to UHPLC. The theory behind
Figure 13.1 The focus of method transfer from HPLC to an UHPLC method
can be on high resolution for high peak capacity (left, up to 1000 peaks
separated in a single run) or on high speed for high sample throughput
(right, 10 peaks separated in 10 s). Reproduced with permission of
Thermo Fisher Scientific.
1 k a –1
R = N 2 (13.1)
4 1 + k2 a
k2
a= (13.2)
k1
L
N= (13.3)
H
370 Method Transfer between HPLC and UHPLC
B
H = A+ + C u (13.4)
u
10
uopt (13.6)
dp
t1, the calculated analysis time of the UHPLC method t2 with new
column parameters is described by Eq. 13.8.
dp,2 L2
t 2 = t1 (13.8)
dp,1 L1
uLÖ (13.10)
P =
dp2
The pressure drop generated by different particle sizes is
shown in Fig. 13.4. Operating a column filled with 5 µm particles
at 160 bar pressure drop would mean that the same column
filled with 2 µm particles, would in theory generate 1000 bar
pressure drop assuming that the other parameters like linear
velocity, column length, viscosity, and porosity are kept constant.
The optimum flow rate for HPLC to UHPLC method transfer
is influenced by the particle size (see Eq. 13.6). This dependency,
combined with Darcy’s law, allows the conclusion that the added
back pressure is inversely proportional by the third power of
particle size and directly related to the column length. The
added pressure drop for the method transfer from HPLC column
formats to UHPLC column formats can be calculated by Eq. 13.11.
3
dp,1 L
P2 = P1 3
2 (13.11)
dp,2 L1
Figure 13.4 The simulated pressure drop on the column rises with inverse
square of the particle size according to Eq. 13.10 demonstrates the
trade-off for UHPLC separation targeting higher peak capacity or higher
analysis speed. Reproduced with permission of Thermo Fisher Scientific.
Table 13.1 Estimated efficiency, time saving, solvent reduction, and back
pressure increase for method transfer from HPLC method to UHPLC
method for different column geometries generating comparable efficiencies
and resolution
dc,2 2 dp,1
F2 = F1
d (13.12)
c,1 dp,2
2
F1 L2 dc,2
t 2 = t1 (13.13)
F2 L1
dc,1
dc,2 2 L2 dp,2
Vinj, 2 = Vinj,1
d (13.14)
c,1 L1 dp,1
Table 13.2 The pressure drop generated by flow through the capillary is
proportional to the fourth power of the inner diameter of the capillary
according to the Hagen–Poiseuille equation
Figure 13.6 Ideally 30 data points should be recorded for each peak
between the limits of integration. The time constant should be fit to
the set data collection rate. The best settings should be determined by
the narrowest peak. Poor peak symmetry can be improved by reducing
the time constant. High baseline noise can be improved by increasing
the time constant. Reproduced with permission of Thermo Fisher
Scientific.
D1
D2 = (13.15)
3
L2 dp,2
3
L1 dp,1
the scaling of the gradient profiles for new column formats and
flow rates are unavoidable to maintain separation performance.
The theoretical background is known as the gradient volume
principle, introduced by L. Snyder [14]. The gradient volume VG
is defined as the mobile phase volume flowing through a column
at a defined gradient time tG.
VG = F . tG (13.16)
2
F1 L2 dc,2
t g , 2 = t g ,1 (13.17)
F2 L1
dc,1
Figure 13.9 The dwell volume creates a delay before the programmed
solvent compositions reaches the column inlet. This adds an isocratic
hold at the beginning of the gradient. Reproduced with permission of
Thermo Fisher Scientific.
VGDV , 1 Vc ,2
VGDV , 2 = . (13.18)
Vc,1
386 Method Transfer between HPLC and UHPLC
Figure 13.10 The gradient delay volume (dotted line) is defined as the
volume from the mixing of solvents to the column head. By its technical
operating principle the gradient delay volume is lower for high pressure
gradient pumps (top) than for low pressure gradient pumps (bottom).
Gradient delay volume must be considered for optimal gradient
method transfer from HPLC to UHPLC. Reproduced with permission
of Thermo Fisher Scientific.
Figure 13.11 The pseudo isocratic hold while transferring from an HPLC
system with high gradient delay volume (bottom, grey) to a system with
lower gradient volume (top, black) has to be considered for the smooth
transition from HPLC to UHPLC to avoid problems. Reproduced with
permission of Thermo Fisher Scientific.
H 0 1 S 0
ln k = + + ln (13.20)
R T R
13.5 Conclusion
The motivation for transferring a conventional HPLC method to
UHPLC can be better resolution or faster analysis time. Following
simple geometric rules and some practical guidelines allows for
easy transfer the conventional method. A systematic method
speed-up can be made by reducing the particle size, shortening
the column length and increasing the linear velocity of the mobile
phase. For high-resolution chromatography, all of the geometric
rules can greatly impact the separation and should be considered
accordingly. Method transfer tools embedded in chromatography
data software and available online or offline help to easily apply
the geometric rules.
Not only does the pressure capability of the UHPLC
instrument performance for method transfer matter, but the extra-
column volume, gradient delay volume, detection options, and
thermostatting characteristics have to be considered as well.
While there are many facets to consider for successful method
transfer, UHPLC separations give a greater freedom to exploit the
principles of chromatography and get the optimized method to
solve the analytical problem.
Acknowledgements
My thanks go to my colleagues for contribution to the chapter,
especially Holger Franz, Frank Steiner, Sabrina Patzelt, Michael
Heidorn, and Jenny Wong.
References
Small-Molecule Pharmaceutical
Impurities Test Method Validation:
Precision Acceptance Criterion
14.1 Introduction
The International Conference on Harmonization (ICH) guideline
[1] for pharmaceutical small-molecule impurities test method
validation does not provide any guidance in setting numerical
validation performance criteria for linearity, accuracy, and
precision. It specifies a numerical minimum acceptance criterion
See Fig. 14.2, noting that it plots the log of absolute standard
deviation versus log c.
RSDR, Observed
HorRat = 2.0
RSDR, Horwitz
408 Small-Molecule Pharmaceutical Impurities Test Method Validation
Figure 14.5 HorRat = 2.0 line and the quantile regression of the AbbVie
data of seventy two impurity, degradation product or minor component
test method precision validations. The analyte concentration, c (%w/w),
is relative to the whole matrix mass.
Test Method Variability as a Function of Analyte Concentration 411
Acknowledgments
The authors thank the many Abbott and AbbVie analytical chemists
who endeavored to develop and validate the robust pharmaceutical
test methods reflected by the test method validation data set
characterized herein. The authors also thank AbbVie New Chemical
Entity Analytical Chemistry R&D and Non-Clinical Statistics
department senior management for their support of resources
for this standing functional interface within the AbbVie Global
Pharmaceutical R&D Development Sciences CMC function.
References
15.1 Introduction
Recombinant proteins and monoclonal antibodies (mAbs),
although relatively complex, are becoming even more complex
with the advent of mAb conjugates (mAb + conjugated peptide or
protein) and antibody drug conjugates (ADCs) consisting of a
mAb + conjugated small molecules. These more complex molecules
are typically characterized by an array of orthogonal analytical
Additionally, for this approach using both acidic and basic buffers,
no pH adjustment is required. A disadvantage of this approach
is that care must be taken to ensure that ionic strength of the
individual buffers within the mobile phases are minimized to
avoid compromising the resolution of the separation due higher
ionic buffer strength from the combined buffers. Another
disadvantage of this approach is that it requires multiple
acidic buffers and multiple basic buffers, making mobile phase
preparation a more complex process requiring multiple acidic and
basic buffers. Additionally, more experiments are required during
development to insure that the buffering strength of the acidic
and basic buffers is appropriately balanced so that a reasonably
stable pH gradient is achieved. In general, the use of a wider pH
range may compromise the resolution of the separation, and
will generally require the use of shallower gradients to achieve
adequate separation. This is due to the increased rate of pH
change across the stationery phase associated with the wider pH
gradient. However, this drawback is easily overcome by reducing
the pH range of the gradient. The advantages of narrower pH
gradients will be discussed later in this chapter.
that the ionic strength of the individual buffers within the mobile
phases is minimized to avoid compromising the resolution
of the separation due higher ionic strength from the combined
buffers and subsequent pH adjustment. As with option 15.2.1
the drawbacks associated with a wider pH gradient range are
easily overcome by reducing the pH range of the gradient in the
mobile phases. This approach is outlined in the following section.
Mobile Phases Consist of a Single Buěer or Group of Buěers Consisting of Weak A cids
or Bases Which Běr Cover a Specięed pH Range
11
Buěer pH adjusted above molecule pI
10
6
Buěer pH adjusted below molecule pI
5
5.5 6.5 7.5 8.5 9.5 10.5
Mobile Phase Buěering Range (e.g. pH 6 -10)
0.10
µL NaOH RT µL NaOH RT µL NaOH RT µL NaOH RT
1400 22.9 1050 34.3 700 41.2 350 47.9
0.08 M.P. pH
pH = 8.2
8.2 M.P. pH
pH = 7.8
7.8 M.P. pH
pH = 7.4
7.4 M.P. pH
pH = 7.0
7.0
0.06
AU
Allowable
AllowablepH pH
AU
0.04 Range
RangeforforMethod
Method
pH
pH8.0
8.0±±0.1
0.1
0.02
0.00
10.0 15.0 20.0 25.0 30.0 35.0 40.0 45.0 50.0 55.0 60.0
Minutes
Minutes
7.5
pH
7 adjustment
6.5
6
5.5
0 20 40 60 80 100
% Mobile Phase B
0.20
mAb 1 A
CombinĂƟŽn 1
AU
0.10
(pH adjusted)
0.00
0.15
mAb 1
0.10
CombinĂƟŽn 2
AU
0.05
(non-pH adjusted)
0.00
0.00 2.00 4.00 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00 24.00 26.00 28.00 30.00 32.00 34.00 36.00 38.00 40.00 42.00 44.00
Minutes
0.20
mAb 2 B
CombinĂƟŽn 1
AU
0.10
(pH adjusted)
0.00
0.15
mAb 2
0.10
CombinĂƟŽn 2
AU
0.20
mAb 3 C
CombinĂƟŽn 1
AU
0.10
(pH adjusted)
0.00
0.20
mAb 3
CombinĂƟŽn 2
AU
0.10
(non-pH adjusted)
0.00
0.00 2.00 4.00 6.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00 24.00 26.00 28.00 30.00 32.00 34.00 36.00 38.00 40.00 42.00 44.00
Minutes
0.20
mAb 4 D
CombinĂƟŽn 1
AU
0.10
(pH adjusted)
0.00
0.15
mAb 4
0.10
CombinaƟon 2
AU
Figure 15.4 (A–D) Three mAbs (labeled 1–3) and one mAb conjugate
(labeled 4) were compared using combination 1 and combination 2.
The separation is improved for all molecules using combination 2, the
non-pH adjusted buffers.
Impact of Ions on IEC Separations 427
$8
0LQXWHV
Table 15.4 Area% data for acidic, basic, and main peak at various
separation conditions
Buffer
Run time conc. Grad. slope
Name (Min) (mM) (%/min) %Acidic % Main % Basic
mAb 2 45 16 2.4 35.64 37.63 26.73
mAb 2 75 16 1.2 34.40 37.46 28.14
mAb 2 75 10 1.2 36.80 36.68 26.52
mAb 2 75 7 1.2 36.12 37.33 26.56
mAb 2 75 5 1.2 37.06 36.89 26.05
mAb 2 45 7 1.2 36.72 37.36 25.92
Mean 36.12 37.23 26.65
Stdev 0.99 0.36 0.79
%RSD 2.74 0.97 2.98
10
5mM
8
50mM
pH
5
0 20 40 60 80 100
% Mobile Phase B
*Elution pH is calculated from main peak RT data in Fig. 15.5 with the gradient slope
of 2.4%/min to determine the mobile phase composition and subsequent elution
pH which is calculated from the average slope – intercept data from Fig. 15.8.
Buffer pH range
MOPSO 6.2–7.6
HEPES 6.5–7.9
AMPSO 8.3–9.7
BIS-TRIS propane 6.3–9.5
Ammonium acetate 3.8–5.8
Ammonium formate 2.8–4.8
Table 15.9 Retention time and elution pH information from two different
mAbs with the same theoretical pI of 7.3, which were run with Bis-Tris
propane mobile phases at 10 mM
Figure 15.13 Chromatographic overlays of two different mAbs with theoretical pIs of 8.5 and 8.7. Both were run using the same mobile
phases and pH gradient (cation exchange) method used in Table 15.8 and 15.9.
Molecule Information and Method Development 441
Figure 15.14 Chromatographic overlay of a single mAb (IgG 2) with a theoretical pI of 9.1 run using a 5 mM pH gradient, or a 5–10
or 5–20 mM pH/ionic strength gradient (combination) in cation exchange separation mode.
Using pH Gradient Separation for mAbs, ADCs and Other Complex Bio-Pharmaceuticals
Figure 15.15 Chromatographic overlay of a single mAb (IgG 1) with a theoretical pI of 8.9 run using a 5 mM pH gradient, a 5–10 or
5–20 mM pH/ionic strength gradient (combination) with cation exchange separation mode.
Molecule Information and Method Development
443
444
Using pH Gradient Separation for mAbs, ADCs and Other Complex Bio-Pharmaceuticals
Figure 15.16 Chromatographic overlay of two mAbs from Figs. 15.14 and 15.15 (IgG 1, Top and IgG 2, Bottom) with theoretical pIs
of 8.9 and 9.1 respectively, run using a 3–35 or 3–50 mM ionic strength gradients in cation exchange separation mode.
Molecule Information and Method Development 445
Figure 15.17 Separation of a fusion protein (100 kDa, pI 8.35) using CEX
ionic strength pH gradient separation. Top profile shows the impact of
5 mM NaCl in mobile phase B. Bottom profile shows separation (left to
right) of homo and heterodimer species.
Figure 15.18 AEX profile and expanded plot of vaccine with a theoretical pI of ~4.0, and molecular weight of ~55 kDa.
448 Using pH Gradient Separation for mAbs, ADCs and Other Complex Bio-Pharmaceuticals
Figure 15.19 AEX IgG 4 parent mAb and ADC with pH gradient CEX
separation.
Molecule Information and Method Development 449
Figure 15.20 IgG 1 parent mAb and ADC with pH gradient CEX separation.
Both of the profiles in Fig. 15.19 were run using CEX separation
mode. The mobile phases were the same for both separations.
Mobile phase A was 5 mM HEPES, 5 mM AMPSO no pH adjustment.
Mobile phase B was 5 mM Bis-Tris propane no pH adjustment.
The gradient slope for both was 1.5%/min. The column used for
the mAb was a Dionex ProPac WCX-10, 4 × 250 mm. The column
used for the ADC was a MabPac SCX-10, 4 × 250 mm.
The mAb/ADC comparison clearly shows the influence of the
conjugate on the separation. However, the separation conditions
in terms of mobile phases and gradient slopes are the same
for both showing in this case that the conjugation may significantly
impact the chromatographic profile, but separation conditions
are comparable.
Both of the profiles in Fig. 15.20 were also generated in the CEX
separation mode. Based on the higher pI of the molecule (pI 8.9),
an ionic strength gradient was used for these separations.
The mobile phases were the same for both the mAb and the ADC.
Mobile phase A was 3 mM Bis-Tris propane, no pH adjustment.
Mobile phase B was 50 mM Bis-Tris propane no pH adjustment.
The gradient slope for the mAb was 1.5%/min, while the gradient
slope used for the ADC separation was slightly greater at 1.7%/min.
The column used for the mAb and ADC was a MabPac SCX-10,
4 × 250 mm
In this comparison as with the comparison in Fig. 15.19 we see
the influence of conjugation on the separation. The separation
conditions in terms of mobile phases are the same for both
molecules and the gradient slopes are comparable. This further
450 Using pH Gradient Separation for mAbs, ADCs and Other Complex Bio-Pharmaceuticals
Figure 15.21 Separate overlays of two IgG 2 mAbs comparing a wider pH gradient (~7 pH units) to a narrower pH gradient
(~3 pH units).
Platform Application of pH Gradients 453
Figure 15.23 Comparison of two profiles from the same method using columns from different manufacturers with loads of 5 mg (top
profile) and 25 µg (bottom profile).
Mobile Phase Preparation 457
Table 15.11 List of columns which have been used for IEC separations
in this chapter
Figure 15.26 CEX (Top) and AEX (Bottom) profiles for a highly sialylated IgG2 with a theoretical pI of 7.3.
464
Using pH Gradient Separation for mAbs, ADCs and Other Complex Bio-Pharmaceuticals
Figure 15.27 CEX (Bottom) and AEX (Top) profiles for an IgG2 mAb with a theoretical pI of 7.3.
Summary 465
Figure 15.27 shows the IgG2 from Fig. 15.23, with a theoretical
pI of 7.3 which has been separated using the same mobile
phases used in Fig. 15.22 in both CEX and AEX separation modes.
In this case, the CEX separation is shown to be preferable over
the AEX separation mode. The gradient slope for the CEX mode
was 0.7%/min while the gradient slope for the AEX mode was
0.5%/min.
Table 15.12 Summary Table of separation conditions for mAbs, ADCs, and other bio-therapeutics
mAb, IgG1 7 to < 9 150 HEPES/AMPSO Bis-Tris propane ~10–20 mM CEX pH 4D, 11, 12, 13, 23,
mAb IgG1 ≥9 150 N/A Bis-Tris propane 3–35 mM CEX Ionic Strength 16
ADC, IgG1 ~9 150 N/A Bis-Tris propane 3–35 mM CEX Ionic Strength 21
mAb IgG2 7 to < 9 150 HEPES/AMPSO Bis-Tris propane ~5–10 mM CEX pH 4A, 4B, 4C, 10, 11, 13,
23, 24,
mAb IgG2 ≥9 150 N/A Bis-Tris propane 3–35 mM CEX Ionic strength 16, 21
Fusion ~8.4 100 Bis-tris propane + HCL Bis-tris propane 3–60 mM CEX pH-Ionic strength 17
Protein combination
15.13 Summary
Table 15.12 provides a summary of the information from the
separations in this chapter and should serve as a resource for
method development of other mAbs, ADCs, as well as other bio-
therapeutics. Matching the general information in the molecule
and pI columns to another mAb, ADC, or other molecule should
provide useful starting information for developing a method.
For example, if one wanted to develop a separation for a mAb,
IgG1 subclass, with a pI of 8.1, the conditions listed in the first row
of Table 15.12 will likely be suitable for generating an acceptable
separation to start with, and are summarized in Table 15.13.
Further information regarding the separations associated with
each row can be obtained through the figure references in the
far right hand column of Table 15.12.
Parameter Specification
Separation mode CEX
Columns Dionex, Mab Pac, SCX-10, 4 × 250,
Waters, Protein-Pak, Hi Res CM, 4.6 × 100 Agilent,
PL-SCX 4000 Å, 4.6 × 150 mm, 8 µm
Column temp. 35–40°C
Autosampler temp. 2–8°C
Flow rate ~1 mL/min
Detector wavelength 280 nm
Injection volume 50 µL
Sample dilution 1 mg/mL (water or mobile phase A)
Mobile phase A 15 mM HEPES/15 mM AMPSO
Mobile phase B 15 mM Bis-Tris propane
15.14 Conclusions
This chapter describes a simple approach to developing highly
resolving and robust pH gradient IEC methods for mAbs, ADC’s, and
468 Using pH Gradient Separation for mAbs, ADCs and Other Complex Bio-Pharmaceuticals
References
1. Flatman, S., Alan, I., Gerard, J., Mussa, N. J. Chromatogr. B, 2007, 848,
79–87.
2. Schenerman, M., Sunday, B. R., Kozlowski, S., Webber, K., Gazzano-
Sanotora, H., Mire-Sluis, A. BioProcess Int., 2004, 42–52.
3. Harris, R. J. J. Chromatogr., A, 1995, 705, 129–134.
4. Weitzhandler, M., Farnan, D., Horvath, J., Rohrer, J. S., Slingsby, R. W.,
Avdalovic, N., Pohl, C. J. Chromatogr. A, 1998, 828, 365–372.
5. Santora, L. C., Krull, I. S., Grant, K. Anal. Biochem., 1999, 275, 98–108.
6. Moorhouse, K. G., Nashabeh, W., Deveney, J., Bjork, N. S., Mulkerrin, M.
G., Ryskamp, T. J. J. Pharm. Biomed. Anal., 1997, 16, 593–603.
7. Chelius, D., Jung, K., Lueras, A., Rehder, D. S., Dillon, T. M., Vizel, A.,
Rajan, R. S., Li, T., Trauheit, M. J., Bondarenko, P. B. Anal. Chem., 2006
78, 2370–2376.
8. Donato, A., Ciardiello, M., Nigris, M., Piccoli, R., Mazzarella, L., D’Alessio,
G. J. Biol. Chem., 1996, 268, 4745–4751.
9. Hsu, Y.–R., Chang, W.-C., Mendiaz, E. A., Hara, S., Chow, D. T., Mann, M. B.,
Langley, K. E., Lu, H. S. Biochemistry, 1998, 37, 2251–2262.
References 469
10. del la Guntińas, M., Wissiack, R., Bordin, G., Rodrigez, A. R. J. Chromatogr.
B, 2003, 791, 73–83.
11. Santora, L., Kaymakcalan, Z., Sakorafas, P., Krull, I., Grant, K. Anal.
Biochem., 2001, 299, 119–129.
12. Vlasak, J., Ionescu, R. Curr. Pharm. Biotechnol., 2008, 9, 468–481.
Chapter 16
(partially) [1]. Very large solutes (larger than the pores) can be
totally excluded from the pores and will elute within the interstitial
column volume. Based on its mechanism, SEC is applied to
separate solutes of different sizes (molecular weights). Hence, SEC
is commonly used to separate protein aggregates (size variants)
such as monomers, dimers, trimers, and other oligomers or
high-molecular-weight species (HMWS).
Among the different techniques applied for the characterization
of aggregates—such as sodium dodecylsulfate polyacrylamide gel
electrophoresis (SDS-PAGE), asymmetrical field flow fractionation
(AF4), fluorescence spectroscopy, circular dichroism (CD) or light-
scattering-based methods, like multi-angle laser light scattering
(MALLS)—SEC is considered today as the reference technique
for the quantification of protein aggregates. The main advantage
of this approach is the mild elution conditions (non-denaturing)
allowing the characterization of proteins with minimal impact
on their conformational structure and the local environment.
In SEC, we strive for separating the proteins under inert conditions.
Indeed, the separation should ideally depend only on the size of
the solutes. However, in practice, some secondary interactions
(hydrophobic and/or electrostatic) may occur between the proteins
and the stationary phase [2]. To limit such interactions and work
under inert conditions, the pH and ionic strength of the mobile
phase should be carefully controlled. Ideally, the mobile-phase pH
should be set close to the isoelectric point (pI) of the proteins but
it is not applied in practice for basic proteins and a physiological
pH is usually applied. Salts (sodium or potassium chloride) or
other additives (arginine) are also often added to the mobile
phase to avoid or at least limit electrostatic interactions, while a
small proportion of organic modifier (e.g., 10% 2-propanol) can
also be used to limit hydrophobic interactions as much as possible.
The aim of this chapter is to provide some guidelines to
develop the SEC method for proteins and to show the possibilities
of modern SEC (UHP-SEC) separations.
Vp ep t0 – te
kmax
= = = , (16.2)
Ve ee te
where Velution and telution are the elution volume and elution time,
respectively, while Ve and te are the total exclusion volume and
time (determined by the interstitial volume) and t0 is the column
dead time (determined Vp byep thet 0 –sum of interstitial and pore
te
volumes). The kmax =value =
= then ,
defines the possible
Ve ee t Vp ep elution
t –t
window (tW) for the solutes. The elarger the kmax , =the = 0the e ,
=better
Ve ee t
probability to separate the compounds. Figure 16.1 shows ea
typical SEC chromatogram and corresponding te, telution, t0 andV e
t –t
tW. Using currently available state-of-the-art columns, the kmax =is p = p = 0 e ,
Ve ee te
around 1.4–1.8. This parameter—and the associated selectivity—
can be altered by packing a column more densely (or decreasing
particle size) or by using particles with larger pore volumes.
Another important characteristic of SEC columns is the
calibration curve, since it illustrates the size (weight) of analytes
that can be separated on a given column. The calibration curve
474 Size Exclusion Chromatography Method Development for Therapeutic Proteins
where A and B are the slope and intercept of the line, respectively.
As the pore size distribution becomes narrower, the slope
becomes shallower which results in a greater selectivity to
discriminate analytes of similar size. The intercept of the linear
part depends on the average pore diameter. Figure 16.2 shows
Theoretical Aspects 475
B
H = Au01/2 + + C u0 , (16.4)
u0
476 Size Exclusion Chromatography Method Development for Therapeutic Proteins
Figure 16.3 Calculated H–u curve in SEC for a 150 kDa protein using a
column packed with 3.0 µm particles operating at 25°C. Reproduced
with permission from Elsevier.
Figure 16.4 Measured H–u curves for β-lactoglobulin (A) and a therapeutic
monoclonal antibody (B) by using columns packed with 5.0, 2.7, 2.0, and
1.8 µm particles, at ambient temperature. Reproduced with permission
from Elsevier.
Particle Max. temperature Max. pressure
Table 16.1 Some popular state-of-the-art SEC columns and their properties
Name (provider) Material size (µm) Pore size (Å) (˚C) pH range (bar)
Acclaim SEC-300 hydrophilic 5, 7 300, 1000 60 2–12 80
(Thermo) polymethacrylate resin
Acquity UPLC BEH SEC diol modified hybrid-based 1.7, 2.5 125, 200, 450 60 2–8 400
(Waters)
AdvanceBio SEC (Agilent) surface-coated silica-based 2.7 130, 300 80 2–8.5 400
Bio SEC-3 (Agilent) surface-coated silica-based 3 100, 150, 300 80 2–8.5 240
Bio SEC-5 (Agilent) surface-coated silica-based 5 100, 150, 300, 80 2–8.5 240
500, 1000,
2000
SRT-SEC (Sepax) surface-coated silica-based 5 100, 150, 300, 80 2–8.5 240
500, 1000,
2000
TSKgel Ultra SW diol modified silica-based 3 300 30 2.5–7.5 120
aggregate (Tosoh)
TSKgel SW mAb (Tosoh) diol modified silica-based 4 250 30 2.5–7.5 120
TSKgel UP-SW3000 diol modified silica-based 2 250 30 2.5–7.5 250
(Tosoh)
Yarra (Phenomenex) silica-based 1.8, 3, 5 145, 150, 290, 60 2.5–7.5 (except 200 (except for
300, 500 for SEC-X150, 1.8 µm: 480 bar)
X300: 1.5–8.5)
Method Development of Proteins
YMC-Pack Diol-SEC (YMC) diol modified silica-based 3, 5 60, 120, 200, 40 5–7.5 200
300
479
Figure 16.5 Effect of salt (NaCl) concentration on the elution time of an IgG1
monoclonal antibody. Overlaid chromatograms correspond to different
salt concentrations on column C (A) while plots of relative change in k
vs. salt concentrations have been prepared for four different columns (B).
Reproduced with permission from Elsevier.
16.5 Perspectives
Since the introduction of UHPLC systems and columns, which
enabled high resolution, sensitivity, and peak capacity, there have
been a variety of new analytical advancements in LC and LC–MS
based on the use of low dispersion systems [40]. In SEC, the
trend is the same as in the other modes of liquid chromatography,
namely the decrease of particle sizes and column dimensions.
In conventional SEC, 300 mm × 6–8 mm columns are commonly
used, while the recent trend in UHP-SEC is to work with
150 mm × 4.6 mm columns. Many column providers offer such
UHP-SEC columns and efforts are also made to improve the
inertness of the stationary phases. However, further decrease in
SEC column dimension is not expected, due to the loss of intrinsic
column efficiency caused by commercially available LC systems.
This behavior is related to the nature of SEC separations. Indeed,
the band variance depends on the solute retention factor k¢, which
in SEC ranges between –1 and 0. It inherently results in very
low peak variances for columns of low volumes, which would be
too strongly affected by the system variance [12].
To improve the sensitivity of SEC separations or work with
very small amounts of available samples, the use of capillary
columns can be a promising approach. However, important
modifications to a commercially available LC systems are required
to drastically reduce the system volume. Until now, the number of
References 491
References
28. Liu, B., Guo, H., Xu, J., Qin, T., Xu, L., Zhang, J., and Dai, J. (2016).
Acid-induced aggregation propensity of nivolumab is dependent on
the Fc. mAbs, 8(6), 1107–1117.
29. Nicoud, L., Cohrs, N., Arosio, P., Norrant, E., and Morbidelli, M.
(2015). Effect of polyol sugars on the stabilization of monoclonal
antibodies. Biophysical Chemistry, 197, 40–46.
30. Hernández-Jiménez, J., Salmerón-García, A., Cabeza, J., Vélez, C., Capitán-
Vallvey, L. F., and Navas, N. (2016). The effects of light-accelerated
degradation on the aggregation of marketed therapeutic monoclonal
antibodies evaluated by size-exclusion chromatography with
diode array detection. Journal of Pharmaceutical Sciences, 105(4),
1405–1418.
31. Zheng, K., Bantog, C., and Bayer, R. (2011). The impact of glycosylation
on monoclonal antibody conformation and stability. mAbs, 3(6),
568–576.
32. Koza, S., Hong, P., and Fountain, K. J. (2012). Size-exclusion ultra
performance liquid chromatography analysis of insulin (Application
note No. 720004271EN). Retrieved from Waters Corporation
website: http://www.waters.com/webassets/cms/library/docs/
720004271en.pdf.
33. Gjoka, X., Schofield, M., Cvetkovic, A., and Gantier, R. (2014).
Combined protein A and size exclusion high performance liquid
chromatography for the single-step measurement of mAb, aggregates
and host cell proteins. Journal of Chromatography B, 972, 48–52.
34. Haberger, M., Leiss, M., Heidenreich, A. K., Pester, O., Hafenmair,
G., Hook, M., and Bulau, P. (2016). Rapid characterization of
biotherapeutic proteins by size-exclusion chromatography coupled
to native mass spectrometry. mAbs, 8(2), 331–339.
35. Schmidt, A. C., Fahlbusch, B., and Otto, M. (2009). Size exclusion
chromatography coupled to electrospray ionization mass
spectrometry for analysis and quantitative characterization of
arsenic interactions with peptides and proteins. Journal of Mass
Spectrometry, 44(6), 898–910.
36. Liu, H., Gaza-Bulseco, G., and Chumsae, C. (2009). Analysis of
reduced monoclonal antibodies using size exclusion chromatography
coupled with mass spectrometry. Journal of the American Society
for Mass Spectrometry, 20(12), 2258–2264.
37. Li, Y., Gu, C., Gruenhagen, J., Zhang, K., Yehl, P., Chetwyn, N. P.,
and Medley, C. D. (2015). A size exclusion-reversed phase two
dimensional-liquid chromatography methodology for stability and
References 495
17.1 Introduction
Countercurrent chromatography is a separation technique for
the isolation of a mixture using a liquid stationary phase held in
place by centrifugal force. Many texts are available that describe
countercurrent chromatography and related instrumentation
[1–6]. In addition, several papers by Berthod et al. discuss the use
of ionic liquids in centrifugal partition chromatography (CPC)
and CCC [7–12]. It has been suggested, that the high viscosity of
ionic liquids would be applicable in hydrodynamic (planetary
centrifuges) or hydrostatic (sun centrifuges), countercurrent
chromatography [8]. Berthod et al. reported that the use of ionic
liquids in CCC was found to have a number of drawbacks such
as “UV absorbance limiting the use of the convenient UV detector,”
“non-volatility precluding the use of the evaporative light-scattering
detector for continuous detection” and “high viscosity producing
pressure build-up” [8]. The authors conclude that CCC was
unsuitable at that time, as the instrumentation available “was not
able to cope with the high back-pressures generated by the large
viscosities of many ionic liquids” [8]. This view of the use of ionic
liquids in CCC was correct with the instrumentation available at
the time. However, with careful experimental design and the use
of wider coils and pipework, it has been possible to use ionic
liquids in CCC. Several papers have been published that show
the successful use of ionic liquids and related deep eutectic
solvents (DES) in CCC [13–19].
17.2.1 Introduction
Ionic liquid research came about as a result of three discoveries:
(1) the adoption of water stable neutral ionic liquids such as
[C4mim][PF6] [39–41] and [C4mim][BF4] [42, 43], (2) the discovery
that a wide range of chemical reactions can be carried out in ionic
liquids [44–46], and (3) the realization that the number and range,
and diversity of potential ionic liquids that could be synthesized
is enormous [47].
Ionic liquids have unique properties that can help overcome
some of the problems associated with the use of conventional
organic solvents [44, 47]. Ionic liquids are materials composed
entirely of ions, with low melting points (usually <100°C) and
can dissolve a huge range of organic and inorganic compounds,
polymers, bio-molecules and other salts [48]. They have no
measurable vapor pressure at ambient temperatures and pressures
[49], resulting in their non-flammability and fire resistance [50].
These properties and their general stability make many ionic
liquids inherently safe and environmentally friendly solvents [47].
Their use in synthesis [44], catalysis [51] and separations [52]
has expanded significantly in recent years and they are now being
applied in a number of industrial processes [53]. Moreover, the
properties of ionic liquids can be tuned or adjusted by the choice
of the appropriate anion and cation allowing them to be optimized
for a particular application [47].
502 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
2 2
1 1 ) & 6 2 1 1
)& 6 2
2 2
>&PLP@>27I@ >&PLP@>27I@
2
>& PLP@>27I@
1 1 ) & 6 2
2 2
1 1 )& 6 1 6 &)
>&PLP@>17I @ 2 2
1 1 &O
>&PLP@&O
3 &O
Figure 17.1 The structure of the ionic liquids used in Table 17.1.
Step Procedure
1 A range of solvents of varying structure and polarity include n-
hexane, n-heptane, limonene, liquid carbon dioxide,* sub-critical
fluids such as R134a,* as relatively non-polar or low polarity choices;
ethyl ethanoate, MTBE, n-butanol, chloroform, dichloromethane
as mid polarity options; and ethanenitrile, ethanol, methanol and
distilled water as high polarity options. For unknown compounds
or analytes, screen the above solvents as a single liquid to
determine the solvency. Choose those solvents which form a rapid
settling biphasic mixture, and then test the solvency and partition
coefficient in this biphasic mixture.
2 In a 10 ml stoppered measuring cylinder, add a few milligrams of
the analyte to 2 ml of solvent to be used separately and note their
solvency.
3 Mix pairs of solvents known to form biphasic solvent systems when
combined (2 ml of each). For non-polar analytes, n-heptane or
n‑hexane, etc., mixed with ethanol/methanol/ethanenitrile/distilled
water gives some preferred solvent system options. For mid to high
polarity analytes, combinations such as n-butanol, ethyl acetate,
chloroform, dichloromethane, PEG, or dextran with distilled water
are options. Next, if appropriate, measure the partition coefficient
(K) by determining concentration of the target or analyte in both
the upper and lower phase, where K = [analyte]SP/[analyte]MP.
Also note the settling time of the biphasic solvent systems created.
This is accomplished by stoppering the measuring cylinder and
vigorously shaking for 60 s then recording the settling times and
volumes of upper and lower phase.
4 In over 80% of the cases, this simple procedure will accurately
predict the elution position of targets in CCC or CPC. Where
K = 1, the analyte will elute in one coil volume, where K = 0.5 is
half a coil volume, and for K = 2, 2 coil volumes, etc.
5 For cases where the above prediction fails, this is most often
caused by emulsion formation in the CCC or CPC instrument, and
experiments utilizing the instruments, in different modes and
using different injected masses may be needed to optimize the
method.
(Continued)
508 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
Step Procedure
6 For biphasic solvent systems, where inappropriate or unacceptable
settling times and/or partition coefficients are found, the ARP is
used by the addition of a modifier solvent, which is miscible with the
least favored solvent phase, or a solvent which is miscible with the
most favored solvent can be added to adjust the partition coefficient
to preferred range.
7 For laboratory-only methods, simple optimization as in (1) to (6)
can be used with 1, 2, or even 3 additional solvents to rapidly achieve
the required partition coefficients, without the need to test a pre-set
number of set solvent ratios.
8 For process applications and recycling of solvents, the least number
of solvents necessary to obtain the desired distribution ratios is
preferred and often necessary. This can be achieved using ionic
liquid solvent systems that usually have only two or at most three
components. Here the ionic liquid is designed to suit the separation,
rather than using complex mixtures to give optimal separations.
This is where the use of ionic liquids can pay major dividends.
9 For CCC with molecular solvents, it is very important that the
biphasic solvent system chosen has a settling time of less than
60 s and ideally less than 30 s, to achieve good stationary phase
retention. For CPC, slower settling times for the solvent system
can be used successfully, but in our experience, this often achieves
a lower stationary phase retention than is normally found in CCC
for rapid settling biphasic eluents.
*Requires autoclave or high-pressure apparatus.
Abbreviations: MP, mobile phase; SP, stationary phase; MTBE, methyl t-butyl ether or
1-methoxy-1,1-dimethylethane; R134a, 1,1,1,2-Tetrafluoroethane.
target) has a high enough relative loading, it will act as a buffer for
the eluting phase. Having buffered the eluent, initially all compounds
with that pKa will elute. If one solute (ideally your target) is in
relative excess, it will move only, at its buffered pH, giving a square
wave shape signal in the detector. The retention will be determined
by its solubility in the eluent, and will be the only component
to elute. Once this compound has been completely eluted, the
pH gradient will be reestablished and will not depend on other
compounds that can elute at later pHs. The use of this methodology
can often allow an increase in sample loading by 2 to 4 times,
over and above the already high loading of standard CCC/CPC
separations [86]. A list of simple solvent systems for use in CCC is
shown in Table 17.3. The solvent systems that are suitable depend
largely on the polarity of the compounds being separated.
One important factor, particularly with Flash and HPLC
chromatography, is how much mass of analyte can be retained
when complex mixtures are put onto silica or end-capped modern
reverse phase silica chromatography columns. This problem of
sample loss does not occur with CCC/CPC, when the elution/
extrusion separations are carried out. The full mass balance is
nearly always obtained with CCC/CPC type separations. Silica and
reversed-phase silica stationary phases, including fully end-capped
reverse phases, can change the nature of eluting compounds, with
respect to those injected. This can be due to on-column degradation,
and in particular, degradation of acid labile compounds [87].
Generally, CCC/CPC techniques do not decompose compounds,
assuming that the compounds are stable in the solvent system
used. For bio-active samples, if samples are run on a CCC/CPC
instrument, they generally remain bioactive. However, if samples
are run in solid-phase columns, such as Flash or HPLC columns,
the bioactive complex can be damaged, and the bioactivity lost.
CCC/CPC techniques and solvent systems can allow the retention
of the nature or structure of these molecules in the form that
they naturally occur, thereby maintaining the bioactivity. As many
natural bioactive compounds occur as complexes chromatographers
using solid–liquid chromatography denature the complexes found
in their natural state.
512 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
Table 17.3 The choice of standard biphasic solvent systems for molecular
solvents and ionic liquids (in gray background)
17.3 Conclusion
The use of molecular solvents as biphasic eluents has been shown
to be excellent for a huge variety of applications in laboratory
scale preparations of up to kilo or multi-kilo of target. But the
complexity of molecular solvent biphasic eluents with three, four
or on occasions five solvents, becomes less favored in larger-
scale processes. For these larger-scale process applications, of tens
of kilos to many tons per annum, the unique properties of ionic
liquids can provide many advantages, which we have discussed
in the chapter. This can make the additional effort of developing
ionic liquid eluents well worthwhile. The use of ionic liquids in
countercurrent chromatography instruments was initially found
to be problematic due to high backpressures and consequent
low mobile phase flow rates. With careful design of the fluid flow
paths and by eliminating pressure bottlenecks, this problem has
now been solved. Currently there are very few papers in the
literature describing the use of solvent systems containing a high
percentage of ionic liquids in CCC or LLC. Control over the structure
and design of ionic liquids and ionic liquid containing solvent
systems (solvent engineering) has enabled us to select or alter
solute distribution ratios of solutes that are to be separated.
Peptides, proteins, monoclonal antibodies (mAbs), enzymes,
bioactive compounds, precious metals, actinides and lanthanides,
are all ideal candidates for ionic liquid CCC/CPC separation
processes, as all are high value, but can be potentially difficult
References 519
Acknowledgements
We would like to express our gratitude to Invest NI for the
Collaborative Grants for R&D: RD0210506 and RD1111867, the
EPSRC, and the QUILL Research Centre for funding this work. We
would also like to thank Professor Paul Davey and Givaudan for
useful discussions.
References
1. Snyder, L. R., Kirkland, J. J. (1979). Introduction to Modern Liquid
Chromatography, 2nd ed., John Wiley & Sons Inc., New York.
2. Conway, W. D., Petroski, R. J. (1995). Modern Countercurrent
Chromatography, American Chemical Society, Washington, DC.
3. Degenhardt, A., Winterhalter, P. (2001). HSCCC–A powerful tool for
the preparative isolation of bioactive compounds, Pfannhauser W,
Fenwick GR, Khokhar S in Biologically-Active Phytochemicals in Food,
eds. Pfannhauser W, Fenwick GR, Khokhar S, pp. 143–146.
4. Berthod, A. (2009). Countercurrent Chromatography: From the
Milligram to the Kilogram, Grushka E, Grinberg N in Advances in
Chromatography, Vol 47, eds. Grushka E, Grinberg N, CRC Press-Taylor
& Francis Group, Boca Raton, pp. 323–352.
5. Cazes, J. (2009). Encyclopedia of Chromatography, 3rd ed., Taylor &
Francis, Boca Raton.
6. Sutherland, I. A. (2010). Industrial applications of CCC, Cazes J in
Encyclopedia of Chromatography, eds. Cazes J, 3 ed., Taylor & Francis,
Boca Raton, pp. 2116–2119.
7. Berthod, A., Carda-Broch, S. (2003). A new class of solvents for CCC:
Room temperature ionic liquids, J. Liq. Chromatogr. Relat. Technol., 26,
pp. 1493–1508.
8. Berthod, A., Carda-Broch, S. (2004). Use of the ionic liquid 1-butyl-
3-methylimidazolium hexafluorophosphate in countercurrent
chromatography, Anal. Bioanal. Chem., 380, pp. 168–177.
520 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
20. Brown, L., Earle, M. J., Gîlea, M. A., Plechkova, N. V., Seddon, K. R.
(2017). Ionic liquid-liquid chromatography: A new general purpose
separation methodology, Top. Curr. Chem., 74, pp. 1–41.
21. Earle, M. J., Seddon, K. R., Self, R., Brown, L. (2013). Ionic Liquid
Separations. Patent WO2013121218A1.
22. Earle, M. J., Gilea, M. A. (2013). Lentinan extraction process from
mushrooms using ionic liquid. Patent WO2013140185A1.
23. Earle, M., Seddon, K. (2013). Ionic liquid separations. Patent
WO2013121219A1.
24. Earle, M. J., Seddon, K. R. (2013). Ionic Liquid Separations. Patent
WO2013121220A1.
25. Foreiter, M. B., Gunaratne, H. Q. N., Nockemann, P., Seddon, K. R.,
Srinivasan, G. (2014). Novel chiral ionic liquids: physicochemical
properties and investigation of the internal rotameric behaviour in
the neat system, Phys. Chem. Chem. Phys., 16, pp. 1208–1226.
26. Payagala, T., Armstrong, D. W. (2012). Chiral ionic liquids:
A compendium of syntheses and applications (2005–2012), Chirality,
24, pp. 17–53.
27. Franco, P., Blanc, J., Oberleitner, W. R., Maier, N. M., Lindner, W.,
Minguillon, C. (2002). Enantiomer separation by countercurrent
chromatography using cinchona alkaloid derivatives as chiral
selectors, Anal. Chem., 74, pp. 4175–4183.
28. Wang, S. S., Han, C., Wang, S. S., Bai, L. J., Li, S. S., Luo, J. G., Kong,
L. Y. (2016). Development of a high speed counter-current
chromatography system with Cu(II)-chiral ionic liquid complexes
and hydroxypropyl-beta-cyclodextrin as dual chiral selectors for
enantioseparation of naringenin, J. Chromatogr. A, 1471, pp. 155–163.
29. Willauer, H. D., Huddleston, J. G., Rogers, R. D. (2002). Solvent
properties of aqueous biphasic systems composed of polyethylene
glycol and salt characterized by the free energy of transfer of a
methylene group between the phases and by a linear solvation
energy relationship, Ind. Eng. Chem. Res., 41, pp. 2591–2601.
30. Ishii, K., Tanaka, Y., Hata, K., Goto, M., Saitoh, K., Minamisawa, H.,
Shibukawa, M. (2004). Separation of inorganic ions by high-speed
countercurrent chromatography with an aqueous biphasic system,
Bunseki Kagaku, 53, pp. 911–917.
31. Freire, M. G., Claudio, A. F. M., Araujo, J. M. M., Coutinho, J. A. P.,
Marrucho, I. M., Lopes, J. N. C., Rebelo, L. P. N. (2012). Aqueous biphasic
systems: a boost brought about by using ionic liquids, Chem. Soc.
Rev., 41, pp. 4966–4995.
522 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
32. Ina, K., Furuta, R., Kataoka, T., Kayukawa, S., Yoshida, T., Miwa, T.,
Yamamura, Y., Takeuchi, Y. (2011). Lentinan prolonged survival in
patients with gastric cancer receiving S-1-based chemotherapy,
World Journal of Clinical Oncology, 2, pp. 339–343.
33. Xu, X. J., Wang, X. H., Cai, F., Zhang, L. N. (2010). Renaturation of triple
helical polysaccharide lentinan in water-diluted dimethylsulfoxide
solution, Carbohydr. Res., 345, pp. 419–424.
34. Ina, K., Kataoka, T., Ando, T. (2013). The Use of Lentinan for
Treating Gastric Cancer, Anti-Cancer Agents in Medicinal Chemistry,
13, pp. 681–688.
35. Chihara, G., Hamuro, J., Maeda, Y. Y., Arai, Y., Fukuoka, F. (1970).
Fractionation and purification of the polysaccharides with marked
antitumor activity, especially lentinan, from Lentinus edodes (Berk.)
Sing.(an edible mushroom), Cancer research, 30, pp. 2776–2781.
36. Xu, X. J., Zhang, X. F., Zhang, L. N., Wu, C. (2004). Collapse and
association of denatured lentinan in water/dimethlysulfoxide
solutions, Biomacromolecules, 5, pp. 1893–1898.
37. Jiang, Z. G., Du, Q. Z., Sheng, L. Y. (2009). Separation and purification of
lentinan by preparative high speed counter current chromatography,
Chin. J. Anal. Chem., 37, pp. 412–416.
38. Hughes, K., Paisley, N. (2015). Hughes Mushrooms Ltd.
39. Earle, M. J., McCormac, P. B., Seddon, K. R. (1998). Regioselective
alkylation in ionic liquids, Chem. Commun., pp. 2245–2246.
40. Carmichael, A. J., Earle, M. J., Holbrey, J. D., McCormac, P. B., Seddon,
K. R. (1999). The Heck reaction in ionic liquids: A multiphasic
catalyst system, Org. Lett., 1, pp. 997–1000.
41. Earle, M. J., McCormac, P. B., Seddon, K. R. (2000). The first high
yield green route to a pharmaceutical in a room temperature
ionic liquid, Green Chem., 2, pp. 261–262.
42. Holbrey, J. D., Seddon, K. R. (1999). The phase behaviour of 1-alkyl-
3-methylimidazolium tetrafluoroborates, ionic liquids and ionic
liquid crystals, J. Chem. Soc.-Dalton Trans., pp. 2133–2139.
43. Seddon, K. R., Stark, A., Torres, M. J. (2002). Viscosity and density of
1-alkyl-3-methylimidazolium ionic liquids, Abraham MA, Moens
L in Clean Solvents: Alternative Media for Chemical Reactions and
Processing, eds. Abraham MA, Moens L, Amer. Chem. Soc., Washington,
pp. 34–49.
44. Earle, M. J., Seddon, K. R. (2000). Ionic liquids. Green solvents for the
future, Pure Appl. Chem., 72, pp. 1391–1398.
References 523
45. Earle, M. J. (2002). Ionic liquids: Solvents for the twenty-first century,
Rogers RD, Seddon KR in Ionic Liquids: Industrial Applications
for Green Chemistry, eds. Rogers RD, Seddon KR, Amer. Chem. Soc.,
Washington, pp. 90–105.
46. Earle, M. J., Seddon, K. R. (2002). Clean synthesis in ionic liquids,
Trulove PC, DeLong HC, Mantz RA, Stafford GR, Matsunaga M in
Molten Salts Xiii, eds. Trulove PC, DeLong HC, Mantz RA, Stafford GR,
Matsunaga M, Electrochemical Soc Inc, Pennington, pp. 177–189.
47. Freemantle, M. (2010). An Introduction to Ionic Liquids Royal Society
of Chemistry, Cambridge, UK.
48. Wasserscheid, P., Welton, T. (2008). Ionic Liquids in Synthesis, 2nd ed.,
Wiley-VCH, Weinheim, Germany.
49. Earle, M. J., Esperança, J. M. S. S., Gîlea, M. A., Lopes, J. N. C., Rebelo, L. P.
N., Magee, J. W., Seddon, K. R., Widegren, J. A. (2006). The distillation
and volatility of ionic liquids, Nature, 439, pp. 831–834.
50. Kim, H.-T., Kang, J., Mun, J., Oh, S. M., Yim, T., Kim, Y. G. (2016). Pyrrolinium-
based ionic liquid as a flame retardant for binary electrolytes of
lithium ion batteries, ACS Sustain. Chem. Eng., 4, pp. 497–505.
51. Earle, M. J., Wasserscheid, P., Schulz, P. C., Olivier-Bourbigou, H., Favre,
F., Vaultier, M., Kirschning, A., Singh, V., Riisager, A., Fehrmann, R.,
Kuhlmann, S. (2008). Organic Synthesis, Welton T, Wasserscheid P
in Ionic Liquids in Synthesis, eds. Welton T, Wasserscheid P, 2nd ed.,
Wiley-VCH, Weinheim, pp. 265–568.
52. Arce, A., Earle, M. J., Rodriguez, H., Seddon, K. R. (2007). Separation
of aromatic hydrocarbons from alkanes using the ionic liquid 1-ethyl-
3-methylimidazolium bis{(trifluoromethyl) sulfonyl} amide, Green
Chem., 9, pp. 70-74.
53. Plechkova, N. V., Seddon, K. R. (2008). Applications of ionic liquids in
the chemical industry, Chem. Soc. Rev., 37, pp. 123–150.
54. Earle, M. J., Katdare, S. P., Seddon, K. R. (2004). Paradigm confirmed:
The first use of ionic liquids to dramatically influence the outcome of
chemical reactions, Org. Lett., 6, pp. 707–710.
55. Arce, A., Earle, M. J., Katdare, S. P., Rodriguez, H., Seddon, K. R. (2006).
Mutually immiscible ionic liquids, Chem. Commun., pp. 2548–2550.
56. Martins, V. L., Nicolau, B. G., Urahata, S. M., Ribeiro, M. C. C., Torresi,
R. M. (2013). Influence of the Water Content on the Structure and
Physicochemical Properties of an Ionic Liquid and Its Li+ Mixture, J.
Phys. Chem. B, 117, pp. 8782–8792.
57. Wagner, M., Stanga, I., Schroer, W. (2004). Critical viscosity near the
liquid-liquid phase transition in the solution of the ionic liquid 1-
524 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
69. Kohlmann, C., Leuchs, S., Greiner, L., Leitner, W. (2011). Continuous
biocatalytic synthesis of (R)-2-octanol with integrated product
separation, Green Chem., 13, pp. 1430–1436.
70. Dolan, J. W. (2010). Selectivity in reversed-phase LC separations, Part
I: Solvent-type selectivity, LCGC North America, 28, pp. 1022–1027.
71. Lesellier, E. (2015). Sigma pider diagram: A universal and versatile
approach for system comparison and classification Application to
solvent properties, J. Chromatogr. A, 1389, pp. 49–64.
72. Tan, X., Tan, D. M., Brown, L., Yang, D. P., Zhu, L. P., Chen, Y. L., Chang, D.
H. T., Wang, D. M. (2013). Isolation and purification of four bioactive
constituents from alpinia officinarum hance utilizing high-speed
counter-current chromatography, J. Liq. Chromatogr. Relat. Technol.,
36, pp. 1355–1365.
73. Pellett, J., Lukulay, P., Mao, Y., Bowen, W., Reed, R., Ma, M., Munger, R.
C., Dolan, J. W., Wrisley, L., Medwid, K., Toltl, N. P., Chan, C. C., Skibic, M.,
Biswas, K., Wells, K. A., Snyder, L. R. (2006). “Orthogonal” separations
for reversed-phase liquid chromatography, J. Chromatogr. A, 1101,
pp. 122–135.
74. Kamath, G., Bhatnagar, N., Baker, G. A., Baker, S. N., Potoff, J. J. (2012).
Computational prediction of ionic liquid 1-octanol/water partition
coefficients, Phys. Chem. Chem. Phys., 14, pp. 4339–4342.
75. Soskic, M., Plavsic, D. (2005). Modeling the octanol-water partition
coefficients by an optimized molecular connectivity index, J. Chem Inf.
Model., 45, pp. 930–938.
76. Sedykh, A. Y., Klopman, G. (2006). A structural analogue approach to
the prediction of the octanol-water partition coefficient, J. Chem Inf.
Model., 46, pp. 1598–1603.
77. Xie, Y. J., Liu, H., Liu, H. X., Zhai, Z. C., Wang, Z. Y. (2008). Determination
of solubilities and n-Octanol/Water partition coefficients and QSPR
study for substituted phenols, Bull. Environ. Contam. Toxicol., 80,
pp. 319–323.
78. Hsieh, C. M., Lin, S. T. (2009). Prediction of 1-octanol-water partition
coefficient and infinite dilution activity coefficient in water from
the PR plus COSMOSAC model, Fluid Phase Equilib., 285, pp. 8–14.
79. Souza, E. S., Zaramello, L., Kuhnen, C. A., Junkes, B. D., Yunes, R. A.,
Heinzen, V. E. F. (2011). Estimating the octanol/water partition
coefficient for aliphatic organic compounds using semi-empirical
electrotopological index, Int. J. Mol. Sci., 12, pp. 7250–7264.
526 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
80. Bhatnagar, N., Kamath, G., Chelst, I., Potoff, J. J. (2012). Direct
calculation of 1-octanol-water partition coefficients from adaptive
biasing force molecular dynamics simulations, J. Chem. Phys., 137,
pp. 014502.
81. Garrard, I. J. (2005). Simple approach to the development of a CCC
solvent selection protocol suitable for automation, J. Liq. Chromatogr.
Relat. Technol., 28, pp. 1923–1935.
82. Kukula-Koch, W., Koch, W., Angelis, A., Halabalaki, M., Aligiannis, N.
(2016). Application of pH-zone refining hydrostatic countercurrent
chromatography (hCCC) for the recovery of antioxidant phenolics
and the isolation of alkaloids from Siberian barberry herb, Food
Chem., 203, pp. 394–401.
83. Kotland, A., Chollet, S., Autret, J. M., Diard, C., Marchal, L., Renault, J. H.
(2015). Modeling pH-zone refining countercurrent chromatography:
A dynamic approach, J. Chromatogr. A, 1391, pp. 80–87.
84. Vieira, M. N., Leitao, S. G., Porto, P. C. C., Oliveira, D. R., Pinto, S. C.,
Braz Filho, R., Leitao, G. G. (2013). Application of pH-zone-refining
countercurrent chromatography for the separation of indole
alkaloids from Aspidosperma rigidum Rusby, J. Chromatogr. A, 1319,
pp. 166–171.
85. Wang, T., Jiang, X., Yang, L., Wu, S. (2008). pH-gradient counter-current
chromatography isolation of natural antioxidant chlorogenic acid
from Lonicera japonica Thumb. using an upright coil planet centrifuge
with three multi-layer coils connected in series, J. Chromatogr. A, 1180,
pp. 53–58.
86. Ito, Y. (2013). pH-zone-refining counter-current chromatography:
Origin, mechanism, procedure and applications, J. Chromatogr. A,
1271, pp. 71–85.
87. Connolly, B. A. (1987). The synthesis of oligonucleotides containing
a primary amino group at the 5’-terminus, Nucleic acids research, 15,
pp. 3131–3139.
88. Wang, R., Chang, Y., Tan, Z., Li, F. (2016). Applications of choline
amino acid ionic liquid in extraction and separation of flavonoids
and pectin from ponkan peels, Sep. Sci. Technol., 51, pp. 1093–1102.
89. Yeh, C. S., Yu, T., Berthod, A. (1999). Separation of steroids by
countercurrent chromatography using supercritical fluid carbon
dioxide, J. Liq. Chromatogr. Relat. Technol., 22, pp. 345–356.
90. Yu, T., Chang, J. Y. (2001). Measurements of partition coefficients in
water-1,1,1,2-tetrafluoroethane by countercurrent chromatography,
J. Liq. Chromatogr. Relat. Technol., 24, pp. 1607–1615.
References 527
91. Mai, N. L., Ahn, K., Koo, Y. M. (2014). Methods for recovery of ionic
liquids-A review, Process Biochem., 49, pp. 872–881.
92. Faure, K., Bouju, E., Doby, J., Berthod, A. (2014). Limonene in
Arizona liquid systems used in countercurrent chromatography. II
Polarity and stationary-phase retention, Anal. Bioanal. Chem., 406,
pp. 5919–5926.
93. Friesen, J. B., Pauli, G. F. (2005). GUESS - A generally useful estimate
of solvent systems in CCC, J. Liq. Chromatogr. Relat. Technol., 28,
pp. 2777–2806.
94. Earle, M. J., Gordon, C. M., Plechkova, N. V., Seddon, K. R., Welton, T.
(2007). Decolorization of ionic liquids for spectroscopy, Anal. Chem.,
79, pp. 758–764.
95. Costa, F. D., Leitao, G. G. (2010). Strategies of solvent system selection
for the isolation of flavonoids by countercurrent chromatography,
J. Sep. Sci., 33, pp. 336–347.
96. Hopmann, E., Arlt, W., Minceva, M. (2011). Solvent system selection
in counter-current chromatography using conductor-like screening
model for real solvents, J. Chromatogr. A, 1218, pp. 242–250.
97. Yin, L. H., Xu, L. N., Wang, X. N., Lu, B. N., Liu, Y. T., Peng, J. Y. (2010).
An economical method for isolation of dioscin from dioscorea
nipponica makino by HSCCC coupled with ELSD, and a computer-
aided UNIFAC mathematical model, Chromatographia, 71, pp. 15–23.
98. Berthod, A., Friesen, J. B., Inui, T., Pauli, G. F. (2007). Elution-
extrusion countercurrent chromatography: Theory and concepts
in metabolic analysis, Anal. Chem., 79, pp. 3371–3382.
99. Cocalia, V. A., Visser, A. E., Rogers, R. D., Holbrey, J. D. (2008).
Physicochemical Properties, Wasserscheid P, Welton T in Ionic Liquids
in Synthesis, eds. Wasserscheid P, Welton T, 2nd ed., Wiley-VCH,
Weinheim, pp. 89–102.
100. Domanska, U., Wlazlo, M. (2016). Thermodynamics and limiting
activity coefficients measurements for organic solutes and water in
the ionic liquid 1-dodecyl-3-methylimidzolium bis(trifluoromethylsu
lfonyl) imide, J. Chem. Thermodyn., 103, pp. 76–85.
101. Clough, M. T., Crick, C. R., Grasvik, J., Hunt, P. A., Niedermeyer, H.,
Welton, T., Whitaker, O. P. (2015). A physicochemical investigation
of ionic liquid mixtures, Chem. Sci., 6, pp. 1101–1114.
102. Tariq, M., Shimizu, K., Esperança, J. M. S. S., Lopes, J. N. C., Rebelo, L. P.
N. (2015). Viscosity minima in binary mixtures of ionic liquids plus
molecular solvents, Phys. Chem. Chem. Phys., 17, pp. 13480–13494.
528 Countercurrent Chromatography Using Biphasic Molecular Liquids and Ionic Liquids
103. Zhou, T., Chen, L., Ye, Y., Chen, L., Qi, Z., Freund, H., Sundmacher, K.
(2012). An overview of mutual solubility of ionic liquids and water
predicted by COSMO-RS, Ind. Eng. Chem. Res., 51, pp. 6256–6264.
104. Liu, W., Cheng, L., Zhang, Y., Wang, H., Yu, M. (2008). The physical
properties of aqueous solution of room-temperature ionic liquids
based on imidazolium: Database and evaluation, J. Mol. Liq., 140,
pp. 68–72.
105. Zhang, S. J., Sun, N., He, X. Z., Lu, X. M., Zhang, X. P. (2006). Physical
properties of ionic liquids: Database and evaluation, J. Phys. Chem.
Ref. Data, 35, pp. 1475–1517.
106. Pereiro, A. B., Araujo, J. M. M., Teixeira, F. S., Marrucho, I. M., Pineiro,
M. M., Rebelo, L. P. N. (2015). Aggregation behavior and total
miscibility of fluorinated ionic liquids in water, Langmuir, 31,
pp. 1283–1295.
107. NIST Standard Reference Database #147, (2016) http://ilthermo.
boulder.nist.gov/index.html (verified 19-12-2016).
108. Atashrouz, S., Zarghampour, M., Abdolrahimi, S., Pazuki, G.,
Nasernejad, B. (2014). Estimation of the viscosity of ionic liquids
containing binary mixtures based on the Eyring’s theory and a
modified Gibbs energy model, J. Chem. Eng. Data, 59, pp. 3691–3704.
109. Liu, W. W., Cheng, L. Y., Zhang, Y. M., Wang, H. P., Yu, M. F. (2008).
The physical properties of aqueous solution of room-temperature
ionic liquids based on imidazolium: Database and evaluation, J. Mol.
Liq., 140, pp. 68–72.
110. Arce, A., Earle, M. J., Rodriguez, H., Seddon, K. R., Soto, A. (2010).
Isomer effect in the separation of octane and xylenes using the ionic
liquid 1-ethyl-3-methylimidazolium bis{(trifluoromethyl)sulfonyl}
amide, Fluid Phase Equilib., 294, pp. 180–186.
111. Arce, A.; Earle, M. J., Rodriguez, H., Seddon, K. R., Soto, A. (2009).
Bis{(trifluoromethyl)sulfonyl}amide ionic liquids as solvents for
the extraction of aromatic hydrocarbons from their mixtures
with alkanes: effect of the nature of the cation, Green Chem., 11,
pp. 365–372.
112. Arce, A., Earle, M. J., Rodriguez, H., Seddon, K. R., Soto, A. (2008).
1-Ethyl-3-methylimidazolium bis{(trifluoromethyl)sulfonyl}amide as
solvent for the separation of aromatic and aliphatic hydrocarbons by
liquid extraction–extension to C-7- and C-8-fractions, Green Chem., 10,
pp. 1294–1300.
113. Arce, A., Earle, M. J., Rodriguez, H., Seddon, K. R. (2007). Separation
of benzene and hexane by solvent extraction with 1-alkyl-3-
References 529
456, 458, 460, 462, 464, 466, polymeric ionic liquids (PILs) 264,
468 266, 283, 285, 287, 289, 291,
pH gradient separations 417, 293
419–422, 425, 427, 432, polysaccharide 116, 118, 124–128,
435–436, 438, 448, 453, 455, 130, 140, 144, 171, 500
457 potassium 211, 214–216, 218,
pH gradients 220, 243, 245, 250, 484
narrower 419–420, 451–452 programmed-temperature
wider 419, 451–452 vaporization (PTV) 311
pharmaceutical analyses 13, 111, PTV, see programmed-temperature
vaporization
191, 329, 331, 333, 340, 346,
pyridine 344, 350, 418
351, 354
pharmaceuticals 10, 328, 346, 352,
397, 400
phases
QbD, see quality by design
624-type 349, 351 quality by design (QbD) 14
chiral 167, 171–172, 174
HILIC 77–78, 185
hydrocarbon 273–274
ion chromatography 231 refractive index (RI) 89, 123, 195
organic 109, 167–168, 509 relative standard deviation (RSD)
PEG 283 354–355, 400–407, 409, 429
silica hydride 181, 189, 191, 195, response surface methodology
199, 201–202 (RSM) 29, 352
solid 157 retention factor 72, 77, 95, 122,
phenyl 28, 36, 90, 188, 275–279 133, 143, 272, 315, 369, 388
phosphate 126, 226–228, 231, retention of hydrophilic compounds
235, 241, 250, 254, 485 184–185, 191
PILs, see polymeric ionic liquids retention time shifts 422, 430,
Plackett–Burman design 29, 33, 457, 459–460, 465
38, 43 retention volume 474–475
plasma 200, 278, 331–333 reversed phase (RP) 63, 118, 125,
polar analytes 77, 107–108 127–128, 131, 137, 145, 148,
179–180, 186, 188–190
polar compounds 86–87, 180, 183,
reversed-phase liquid
189, 191, 199, 202, 282, 318
chromatography (RPLC) 21,
analysis of 189, 264
239, 477, 490
retention of 185
reversed-phase liquid
separation of 282, 505
chromatography method
polar organic (PO) 66, 79, 108, development 89, 93
118, 122, 127, 144, 163, 172, RI, see refractive index
186, 288, 516 room-temperature ionic liquids
polar solvents 121, 125, 144, 146 (RTILs) 284–285, 350
540 Index