You are on page 1of 76

A Practical Guide to

High Performance
Liquid Chromatography

The life science business of


Merck KGaA, Darmstadt,
Germany operates as
MilliporeSigma in the
U.S. and Canada.
Table of Contents
Mobile phase composition and temperature 35
1. THEORY OF LIQUID CHROMATOGRAPHY 1
Mobile phase properties 35
Mobile phase pH 36
Quanti_cationexperimentsinanalyticalHPLC 4
Detector choice 37
2. SYSTEM SETUP AND SETTINGS 6 Ultraviolet/Visible Absorbance (UV/Vis) 37
Refractive index (RI) 37
Dwell volume and dead volume 6 Fluorescence (FL) 37
Evaporative light scattering (ELS) 38
Detectorresponsetime 7 Electrochemical (EC) 38
Sample injection, mass, and volume overload 8 Mass spectrometer (MS) 38

Thermostatting 10 Generalrecommendations 40
Column hardware 40
Systemcareandmaintenance 11
InstallationofcolumnswithPEEKend_ttings 40
Equilibrating the Column 41
3. M
 ETHOD DEVELOPMENT Column coupling 41
& OPTIMIZATION 13 Column lifetime, cleaning and regeneration 42
Use with mass spectrometers—procedure to maintain
SelectionoftheHPLCmethodandsystem 13 low column bleeding 42
Storage 43
Method goals 13
Common mistakes in analytical method development 13 Systemoptimization 43
Getting started 13
Method validation 44
Sample Preparation 14
Accuracy 44
Overview of Techniques 14
Precision 44
Selectionofinitialconditions 23 Speci_city 44
Limits of detection and quantitation 44
Mode of separation 23
Linearity 44
ChoosetherightHPLCcolumn 24 Robustness 44
The column backbone 24 Analytical solution stability 45
Stationary phase selection 28
ScalingofHPLCmethods 45
Choosing the right column format 30
Scaling from HPLC to UHPLC 45
Mobilephaseselection 32 Adjusting the column length 45
Isocratic elution 32 Scalingthe‘owrate 45
Gradient elution 32 Scaling the injection volume 45
Gradient method development 32 Adjusting gradient time 45

Mobilephasepreparation 33 Scaling an HPLC method 46


FiltrationtoolsforpreparingHPLC/UHPLCbu^ersand
Speedingupascaledmethod 47
mobile phases 33
Solvent and additive purity 34 Ramipril and Related Substances—from HPLC to UHPLC 48
Chemical compatibility 34 Ramipril and Related Substances—from HPLC to UHPLC 49
4. C
 HROMATOGRAPHIC SEPARATION CommonColumnRestorationProcedures 65
OF LARGE MOLECULES 50 Part 1: Bare Silica Columns 65
Part 2a: Silica-Based Reversed Phase Columns
Introduction 50 Analyzing Water-Soluble Compounds 65
Part 2b: Silica-Based Reversed Phase Columns
SizeExclusionChromatography 50 Analyzing Water-Insoluble Compounds 65
Part 3: Polar-Bonded Reversed Phase Columns
IonExchangeChromatography 52 (Amino, Cyano, Diol, Chiral) 65
Part 4: Silica-Based Ion Exchange Columns 65
Hydrophilic Interaction Liquid Chromatography 53
6. R
 EFERENCE MATERIALS IN LC-MS/MS
A~nityChromatography 53
APPLICATIONS: CHALLENGES
Reversed-PhaseChromatography 54 & SELECTION CONSIDERATIONS 66

References 55 Choosingthecorrectreferencematerialquality
gradeforyourneeds 66
5. T
 ROUBLESHOOTING COMMON Who uses reference materials? 66
HPLC ISSUES 56 Metrological traceability and SI Units of your
reference materials 66

Introduction 56 ISO 17034 and quality grades of standards, reference


materials,andcerti_edreferencematerials 66
Issue No. 1: No Peaks/Very Small Peaks 56 What is measured in each grade of reference material? 67
Issue No. 2: No Flow 56 UnderstandingyourreferencematerialCerti_cate
Issue No. 3: No Pressure/Pressure Lower Than Usual 57 of Analysis 68
Issue No. 4: Pressure Higher Than Usual 57 Reference material formats: do you need a neat,
Issue No. 5: Variable Retention Times 57 solution, or matrix material? 69
Issue No. 6: Loss of Resolution 58 Choosing the correct reference material for your
testing purpose 69
Issue No. 7: Split Peaks 58
Whichqualitygradeisthebest_tforpurpose? 70
Issue No. 8: Peaks Tail on Initial and Later Injections 59
Issue No. 9: Tailing Peaks 59 BestPracticeforPreparationofCalibration
Issue No. 10: Fronting Peaks 60 CurvesinMatrix 70
Issue No. 11: Rounded Peaks 60
Issue No. 12: Baseline Drift 61 APPENDIX 71
Issue No. 13: Baseline Noise (regular) 61
Issue No. 14: Baseline Noise (irregular) 61 Abbreviations 71
Issue No. 15: Broad Peaks 62
Issue No. 16: Change in Peak Height (one or more peaks) 63 LinksandLiteratureforDownload 71
Issue No. 17: Change in Selectivity 63
Issue No. 18: Negative Peak(s) 64
Issue No. 19: Ghost Peak 64
1. Theory of liquid chromatography
High performance liquid chromatography (HPLC) is a The volume of connecting tubing has to be taken into
technique for separating analytes dissolved in a liquid, account as well, especially if the volume of the analytical
mobilephasebyusingtheirspeci_cinteractionwithacolumn used is comparably small (e.g., capillary columns
stationary phase (chromatography column). with a typical i.d. of 50 to 200 µm).
Depending on the type of interaction of the sample • Retention time tR (s) of an analyte is the time elapsed
molecules with both mobile and stationary phases, between the injection and detection of an analyte.
di^erenttypesofinteractionscanbepossibleinHPLC

intensity
(insomecasesbordersare‘uid): tR3
tR2
tR1
• Adsorption chromatography: retention of analytes
by reversible binding on a stationary phase (e.g.,
normal phase; for example allows for the separation
ofisomersbyspeci_cadsorption).
• Partition chromatography: retention by reversible
t0
dissolution in a stationary (quasi-) liquid 3-dimensional
layer;di^erentsolubilityordi^erentpartitioncoea-
cients of samples in liquid stationary and liquid mobile
phase, e.g., in reversed phase (RP) LC, hydrophilic t’R3 time
interaction liquid chromatography (HILIC). t’R2
t’R1

• Ion exchange chromatography: retention by Figure 1. Schematic drawing of a chromatogram displaying the isocratic
electrostatic interaction with the stationary phase. separation of three analytes.

• Aanity chromatography: retention comparable to


adsorption chromatography; interaction of tailor-made The column length L (mm) is prede_ned by the user;
stationary phases with biomolecules (lock and key together with the three parameters described above,
principle). the following information can be calculated:

• Size exclusion chromatography: retention by • The correlation between column back pressure and the
permeation into the pores of a stationary phase and particle size of a particulate column can be calculated
steric exclusion of analytes (e.g., gel permeation from:
chromatography,gelchromatography,gel_ltration).
FL
hp=
The following information is generated during a chro- K0 π r2 dp2
matographic run (Figure 1):
hp change in pressure (bar)
• Back pressure p (bar or psi) is determined during the
eluent viscosity (units?)
analysis for the HPLC utilized and is a combination of
pressures generated by the column and the system. F ‘owrate(mL/min)
K0 column permeability (units?)
• Dead time or hold-up time t0 (s) of a compound not
r column radius (mm)
being retained under the given chromatographic
conditions (i.e. a compound that is not interacting dp particle diameter (µm)
with the stationary phase). Often also referred to
as tm. • Linearvelocityorlinear‘owrateu(mm/s):

t0 can also be calculated from the volume vempty of u = L/t0


a column, the porosity of the stationary phase
andthevolumetric‘owrateF(mL/min,seealso Incontrasttothelinearvelocityu,theunitof‘owrate
description below): FismL/
/ min0The
. ‘owratesetfortheHPLCinstrument
can be converted to u with the following equation:
t0 = vempty/F
4F
u=
For totally porous particles, is typically in the dC 2 π T
range of 0.7–0.8. In turn, the (unknown) porosity
of a chromatographic column can be calculated by dC column diameter (mm)
multiplying F and t0.
T total porosity

1
• Net retention time or reduced retention time t’R: • PlateheightH(m)correspondstothequotientofL
(in µm) and N (L is the length of the column utilized
t’R = tr – t0 for the determination of N).
• Chromatographic resolution R can be calculated from:
• Capacity factor or retention factor k is calculated for
every analyte utilizing the respective retention time R = 2 [(tB – tA)/(WA + WB)]
and the dead time of the separation:
It is a measure of the quality of a separation of two
k = (tR – t0)/t0 analytes A and B (baseline separation has been achieved
whenRl1.4)
• Selectivitycoeacientorseparationfactordescribes
the quality of a separation of two compounds A and B: ClassicalHPLCcolumns(particlediameter5m)are
normallyusedatratherhigh‘owratesandabovethe
=k minimum of their van Deemter plot (see details below).
B/kA
As a result of this, the resolution of such a setup can
The selectivity of a chromatographic column is dictated be improvedbyadecreased‘owrateincrease ( oftr).
by the physical properties of the eluent and the sta- By contrast, UHPLC (Ultra High Performance Liquid
tionaryphase(modi_edorunmodi_ed). Chromatography) columns (particle diameter less than
m)2 areoperatedatcomparablylow‘owrates,and
• Plate count N is calculated from: at the minimum of of their van Deemter plot. In this
2
case,anincreaseof‘owratewillimproveresolution.
N = 16 (tR/w) (USPharmacopoeia(USP(_gure2)
• Peak symmetry is described by the symmetry factor
and describes the performance of a column in isocratic or tailing factor AS (or Tusp) and is utilized by USP (USP
mode. It is given as an absolute value (with respect to chapter 621, see Figure 3):
aspeci_ccolumndimension)orpermeterofacolumn
(unit: m-1). As = (A + B)/2A or As = 0.5 (1 + B/A)

Alternatively N can be calculated from: A and B are determined at 5% of the peak height.

N = 5.54 (tR/w1/2)2 (Figure 2) Alternatively the peak shape can be described by the
asymmetry factor calculated from the ratio of B and A.
The base line width w, and the peak width at half In this case B and A are determined at 10% of the
height w1/2 are determined by the software utilized. peak height.

The plate count depends on column length, geometric For symmetrical peaks, the symmetry or asymmetry
properties of the stationary phase (particle size dis- factor is equal to 1. Peak tailing is observed when the
tribution or monolithic skeleton diameter distribution), factor is >1, whereas peak fronting will be visible when
columnpackingparameters,eluent‘owrateandproper the factor is <1.
setup of the system used (avoidance of dead volume,
see also chapter 2).

As a rule of thumb, the plate count for a particle packed


column is roughly equal to L/2dp (dp: particle diameter).
This means that the plate count can be increased by
increasing the length of a column, or decreasing the
particle diameter. h

tangent line
0.05 h
A B

tR baseline
h w1/2
Figure 3. Schematic drawing of data acquired for the calculation of peak
symmetry AS. A and B are determined at 5% of the peak height h.
tR

w baseline

Figure 2. Schematic drawing of data acquired for the calculation of


plate count N.

2
The shape of a chromatographic peak is ideally narrow Thecorrelationbetween‘owrate,plateheightandthe
and symmetric, but in a real system, peak broadening three terms A, B, and C is described by the van Deemter
(and peak asymmetry) can be caused by the column, or plot (Figure 5) and the following equation:
from the tubing and connectors. The following phenomena
H = A + B/u + C*u
are responsible for this type of peak deterioration
(Figure 4): H Heightequivalenttotheoreticalplatecolumn
( length/eaciency)
• Eddy di^usion (A term): Mixing of the sample with A Eddydi^usion
surroundingmobilephase;peakbroadeningbydi^erent B Longitudinaldi^usion
analyte‘owpaths/pathlengthsthroughthecolumn
C Resistance to Mass Transfer
bed. Eddy di^usion is nearly independent from the
‘ow rate and is in‘uenced by particle size and size u Mobile phase linear velocity
distribution, as well as by the homogeneity of the
column packing.

plate height H
• Longitudinalaxial)
( di^usionB ( term)alongbothcolumn
axis and sample concentration gradient. This e^ect
is rather weak and only strong at low (and therefore
impractical)‘owratesofthemobilephase. mass transfer

• Mass transfer (C term) between stationary and mobile van Deemter plot
phase caused by repeated interaction of sample mole-
cules with the stationary phase. The strength of this
e^ectisproportionaltothe‘owrateofthemobile eddydi^usion
phase.
axialdi^usion

linear‘owrateu

Figure 5. Schematic drawing of a van Deemter plot resulting from the


contributions of eddy di^usion, axial/longitudinal di^usion and mass
transfer to column-related peak broadening.

Ideally, the van Deemter plot displays a rather ‘at


course at overall low plate heights H—corresponding to
alargeplatecountoverawiderangeof‘owrates.Each
chromatographic column possesses its own characteristic
plot, with the minimum of the plot (highest plate count
Figure 4. In‘uenceofmasstransfer(top)andeddydi^usion(bottom) NorlowestplateheightHatacharacteristic‘owrate)
on peak broadening when utilizing fully porous particles. describing the optimum performance of the column
(Figure 6). Please note, that for a given column, the van
Deemterplotbecomes‘atterwithincreasingcapacity
Eddydi^usionisdeterminedbytheparticlesizedistri-
factor k of an analyte. Due to this, the comparison of
bution (PSD) or monolithic skeleton and the domain size
vanDeemterplotsofdi^erentHPLCcolumnsonlymakes
for monolithic materials (sum of pore size and particle
sense when prepared utilizing samples with comparable
or skeleton diameter). The smaller the particle/skeleton
capacity factors.
and domain size and the more homogenous the particle/
skeleton size distribution in a homogenous column 25
bed,thesmallertheresultinge^ectoftheAtermon
peak broadening.
20
PlateheightH( µm)

Thecontributionofaxialdi^usiontopeakbroadeningis
in‘uencedbytheviscosityofthemobilephaseandthe 15
time of analysis.
Peak broadening caused by mass transfer increases with 10
increasing ‘ow rate, larger particle size (larger depth
of pores) and increased viscosity of the mobile phase 5
(decreasedvelocityofdi^usion).
0
0 1 2 3 4 5 6
Linear‘owrateu(mm/s)
Figure 6. Van Deemter plots for Purospher™ STAR RP-18 endcapped
cm
01 xmm
1.2 I.D.columnstop:
( m3 particlesize,bottom:m)2
using anthracene as an analyte.
Chromatographic conditions: Detection: UV (254 nm); mobile phase:
acetonitrile/water 75/25 (v/v); sample: anthracene. The capacity factor
is4.1(3mparticlesize)and4.5(2mparticlesize),respectively.

3
Columns: Discovery™ C18,
5.0 cm x 4.6 mm,
1.
2.
Barbital
Phenobarbital Quantification experiments in
analytical HPLC
5 µm particles 3. Butabarbital
Mobile Phase: CH3OH:H2O 4. Mephobarbital
1 5. Pentabarbital
(45:55)
Temp.: 25 °C 6. Secobarbital
Det.: UV, 214 nm The task of analytical HPLC is the chromatographic sepa-
2
ration of analytes within a sample and their subsequent
4 mL/min
1 2
3
identi_cation.Nexttothesequali_cationexperiments,the
4
quanti_cationofcompounds,forexampleintraceanalysis,
1.0 2.0 3.0 4.0 5.0 6.0 7.0
3 mL/min
can be an additional challenge.
5
The aim of setting up a proper chromatographic system
2 mL/min
1.0 2.0 3.0 4.0 5.0 6.0 7.0
usedinquanti_cationexperimentshastobeamaximi-
1 mL/min zation of the intensity of a target analyte peak and a
minimization of background noise. The combination of
1.0 2.0 3.0 4.0 5.0 6.0 7.0

Time (min)
both optimizes the sensitivity of a separation and hence
Figure 7: In‘uenceofFlowrateonresolution. increases the signal to noise ratio (S/N). The former
can best be achieved by utilizing short columns with the
smallest possible internal diameter (I.D.) (Table 1) in
Inpractice,ahigh‘owratespeedsupseparationssigni_- combinationwithhighinjectionvolumesandhigh‘ow
cantly, but can result in loss of resolution as demonstrated rates in gradient mode. Of course, packing quality has
with a 5 µm particulate column in Figure 7. to be maintained as consistently high and independent
InFigurethe,8 detailedviewshowsasigni_cantlossof from column I.D.. At the same time, minimization of
resolution at a ‘ow rate of 4 mL/min in comparison to background noise can be ensured by using the purest
a ‘ow rate of 1 mL/min using the same HPLC column solvents and reagents and using proper sample prepa-
(Discovery® C18 5 µm, 50 x 4.6 mm) ration and handling.

Smaller particles show a very ‘at Van Deemter plot, Table 1.ColumnI.D.typical
, ‘owrateandcalculatedrelativesensitivity.

which makes them better suitable for fast separations Column I.D. Typical‘owrate Relative
using UHPLC. (mm) (µL/min) sensitivity
4.6 1000–6000 1
2.0 200–800 5.3
0.2 0.5–20 530
0.1 0.4–3 2100
0.05 0.1–0.8 8500

4 mL/min
1.0
In qualitative analysis, the intensity of the smallest
detectable analyte signal should be three times higher
1 mL/min than the noise signal, while in quantitative analyses, the
1.0 2.0 3.0 4.0 5.0 6.0 7.0
ratio should be approximately 10:1. In this context, the
Time (min)
limitofquanti_cation(LOQ)isreferredtoasthelowest
Figure 8. Loss of Resolution Under Non-Optimal Flow Rate amount of analyte allowing for a quantitative analysis
withprede_nedaccuracylow ( deviationbetweentrue0 /
and detected value), and repeatability (repeated analysis
In addition to column-related peak broadening, wider
on the same instrument, with the same settings and
peakscanalsobecausedbye^ectsattributedtotheLC
with a low standard deviation). See also the chapter
systemused.Thesee^ectscanbeduetoaxialdi^usion
on method development. The limit of detection (LOD)
withintheinstrument,hydrodynamic‘owpro_lesbeing
iscalculatedat30%ofLOQ.Asanexample, Figure 9
inconsistent across the tubing cross section, or turbulences
combines the chromatogram and the calibration curves
or mixing at corners, edges or in dead volumes.
from the separation of seven pesticides on a monolithic
silica capillary column. LODs were obtained from a serial
dilution of the analyte mixture.

4
4.9E+07 5.0E+06

R2 = 0.9990

Peak area (MS)


3.9E+07 4.0E+06
MS intensity

2.9E+07 3.0E+06

1.9E+07
2.0E+06

1.0E+06
9.0E+06

0.0E+00
-1.0E+06
0 1 2 3 4 5 6 7 8 9
0 1 2 3 4 5 6 7
Retention time (minutes) Injected mass (pg)

5.0E+06 Figure 9. LC-MS base peak chromatogram displaying the separation


of a stock solution of seven pesticides on a monolithic silica capillary
R2 = 0.9991 column (top). Bottom: Calibration curves for both metamitron (peak 1,
Peak area (MS)

4.0E+06 open circles, left) and metolachlor (peak 7, closed triangles, right). The
insets show the complete concentration range covered (including the
non-linear region typical for ion trap overload) while the large diagrams
3.0E+06 display the linear regions of the curves. The limits of detection for the
setuputilizedare0.24pg(0.16ng/L)formetamitronand0.59pg
(0.4ng/L)formetolachlor.
2.0E+06
Chromatographic conditions: Chromolith® CapRod® RP-18 endcapped
15 cm x 0.1 mm I.D. monolithic silica capillary column. Base peak
1.0E+06 chromatogram (m/z 200–290). Mobile phase A: water + 0.1% formic
acid, mobile phase B: acetonitrile + 0.1% formic acid; gradient: 20% B
to 80% B in 10 minutes.
0.0E+00
0 1 2 3 4 5 6 7
Injected mass (pg)

5
2. System setup and settings
Every single HPLC system has its own characteristics,
and these can change over time—either due to wear
of instrument parts, or by a variation of the setup
by the operator. The following chapter describes
importantsystemparametersandtheirin‘uenceon

Intensity
chromatographic results and gives recommendations
for system optimization. Change from
solvent A 1/ hmax
Dwell volume and dead volume to B 2

The dwell volume of an HPLC instrument is the combined


volume of all system parts from the solvent mixer to the
head of the column. Aside from the capillary connecting retention time
dwell time
the outlet of the injector to the inlet of the column, this
volume is usually typical for a given instrument. The Figure 10. Determination of the dwell time or dwell volume of an
HPLC system.
pump unit of an HPLC system can be either a low or a
high pressure gradient system. In case of the former,
dosing is controlled by a valve, and the mixing of up to In contrast to the dwell volume, all volumes from injector
four solvents takes place on the low pressure side of one to the detector cell—except for the column itself—are
single pump. Due to this construction, the dwell volume of called dead volume. This includes the volumes of all
such a pump system is relatively large. In contrast, high- capillary connections between the injector, column and
pressure gradient systems are composed out of two (or detectorcell,aswellasofthecellitselfand_ttings.The
three) pumps and each pump is dedicated to one single dead volume of a setup has to be kept as small as possible
solvent. Mixing is performed on the high-pressure side of in order to avoid peak broadening, peak tailing and a
the pump system and dosing of the solvents is controlled decreasedseparationperformance(seealsode_nition
bytherelative‘owratesofeachofthepumps.Thedwell of dead time in chapter 1). Both isocratic, and gradient
volume of high-pressure gradients is small, but due to the separationsarenegativelya^ectedbylargedeadvolumes.
construction, these systems are comparably expensive. Connectors and detector cells with low volume should
Modern UHPLC instruments (low or high-pressure gradient therefore be used; in addition, short tubing with low I.D.
systems) display very small dwell volumes, a prerequisite is mandatory. When transferring methods from one HPLC
whenworkingatlow‘owrates. system to another, both the dwell and dead volumes have
to be taken into account. Figure 11showsthein‘uence
Depending on the prerequisites of a separation, the of column installation on the chromatographic result. A
design and length of the connecting tubing may change, gradient run was applied in a separation of a tryptic digest
e.g. when working at temperatures below or above room of cytochrome C. Due to setup restraints in the laboratory,
temperature, or when analyzing biomolecules. Under these a capillary with a length of approximately 0.8 m and an
conditions, the use of inert polyetheretherketone (PEEK) i.d. of 50 m was utilized to connect the HPLC injector
rather than of stainless steel tubing is recommended. with the MS source. The chromatography column was
connected either to the outlet of the injector, or to the inlet
In gradient separations, large dwell volumes are undesir-
ofthemassspectrometertodemonstratethedi^erence.
able as they smooth and delay a gradient and cause
increased equilibration times, but as a consequence,
isocraticseparationsarenotnegativelya^ectedbythe
sizeofthedwellvolume.Underveryspeci_cconditions
such as a large contribution of all system parts from the
injector to the column head to the dwell volume, in com-
binationwithlow‘owrates,peakbroadeningmightbe
observed. For this reason, the dwell volume of modern
UHPLCsystemsforoperationatlow‘owratesisparticu-
larly low. The dwell volume of low-pressure gradient
systems is larger in comparison to high pressure gradient
systems. For the determination of the dwell volume or
dwell time of an HPLC system, the chromatographic
column is replaced by a capillary with low I.D. providing 0 1 2 3 4 5 6 7 8 9 10 11 12
suacientandrealisticbackpressure.Waterandwater/ Retention time (minutes)
acetone 99.5/0.5 (v/v) are used as mobile phases A and B Figure 11. In‘uence of column installation on chromatographic
and a steep gradient is run using UV detection (254 nm). performance. The monolithic silica capillary column was connected either
to the outlet of the injector (top), or to the inlet of the mass spectrometer
The dwell time can be directly read out as the delay (bottom).Forconnection,a50mi.d.silicacapillarywasutilized.
between the time of gradient start and the time of Chromatographic conditions: Chromolith® CapRod® RP-18 endcapped
half-maximum signal intensity 1/2 hmax (Figure 10). The 15 cm x 0.1 mm I.D. HPLC column; injection 1 µL; detection nano-ESI-
dwell volume can be calculated from the dwell time and spraypos.m/
( z‘ow
;)057 3 rateµL/
5.3 min;mobilephasev/
( v)A::
water + 0.1 % formic acid, B: acetonitrile + 0.1 % formic acid; gradient:
‘ow rate. 5% to 25% B in 7 min, 25% to 70% in 3 min; temperature 25 °C; sample:
lyophilized cytochrome C digest resuspended in acetonitrile/water 5/95.

6
The internal diameter of the connecting tubing that can Detector response time
be used in a chromatographic setup depends on column
properties (e.g. back pressure issues), chromatographic Most HPLC detectors have a variable response time or
conditions applied (eluent viscosity, ‘ow rate, temper- time constant. If the response time is too slow (the time
ature), as well as the desired performance and resolution. constant is then too long, e.g., 2 seconds), peaks may
It also must be bigger when matrix rich samples are appear broad and show tailing. For narrow peak widths,
injected (at least between injector and column). In good integration of the peak area, and good optical
general, the smallest possible tubing i.d. should be used presentation of the chromatogram, the data system
inordertogainthehighestcolumneaciency.Fornano settings must enable approximately 20 data points to be
andcapillary-LCcolumn
( i.d.
m)0 5 0 5atubingi.d.of acquired during the peak width time. Therefore columns
0.064 mm should be chosen, whereas in micro, narrow- require a fast detector time constant, such as 0.01 seconds
bore, and conventional analytical LC (column I.D. (100 Hz, i.e. 100 data points per second, which means
1–4.6 mm) a tubing i.d. of 0.13 mm is recommended. In 1 data measurement in 10 milliseconds). As a rule of
addition,adetectorwithamicro‘owcellissuggestedthumb, the time constant should be set to 1/10 of the peak
(check pressure tolerance of the detector cell). Larger width. Always choose the lowest response time possible.
detector cells can be used, but peak widths will be By reducing the time constant from 2 to 0.1 seconds, the
wider (or the intensity will be lower; see Figure 12 and plate count can be improved signi_cantly Figure
( 13).
Table 2). In the given examples, a change from an Keep in mind that a decreased time constant improves
analytical to a micro detector cell leads to an increase in peak shape and column performance, but that at the same
performance of 20–25%. time the baseline noise is increased (because of less signal
averaging). This plays a role when performing quantitative
50
analyses, and when high sensitivity is important. Under
theseprerequisites,therewillbeatradeo^betweenhigh
Intensity (mAU)

40 peak intensity and low baseline noise,and the settings


have to be adapted accordingly.
30

50
20
Intensity (mV)

40
10
30

0
0 1 2 3 4 5 6 20
Retention time (minutes)
10
50

0
Intensity (mAU)

40
0.0 0.4 0.8 1.2 1.6 2.0
Retention time (min)
30

20 300

250
Intensity (mV)

10
200
0
150
0 1 2 3 4 5 6
Retention time (minutes) 100
Figure 12.In‘uenceofUVdetectorcellvolumeandcolumntypeon
thepeakintensityandpeakwidth.L4.1 UVmicrocellgreen
( line), 50
1LUVanalyticalcell(yellowline).Top:Purospher=STARRP-18
®
endcapped (3 µm) Hibar HR 5 cm x 2.1 mm I.D., bottom: Purospher™ 0
STAR RP-18 endcapped (3 µm) LiChroCART® HR 5.5 cm x 2.0 mm I.D.. 0.0 0.4 0.8 1.2 1.6 2.0
Chromatographic conditions: Mobile phase: acetonitrile/water 70/30 (v/v),
‘owrate:0.21mL/min,detection254nm,injection0.2µL,sample
Retention time (min)
(in order of elution): uracil 12 mg/L, ethylbenzene 839 mg/L,
Figure 13. Setting the proper detector response time (top: 2 s,
propylbenzene 922 mg/L, butylbenzene 1006 mg/L.
bottom: 0.1 s) in fast separations.
Chromatographic conditions: Chromolith Performance RP-18 endcapped
Table 2. Change of chromatographic parameters when switching from
10 cm x 4.6 mm I.D. HPLC column, mobile phase: acetonitrile/water
an1Ltoa1.4LUVcell(example:butylbenzenepeak).
40/6(v/v),‘owrate:5.0mL/min,injectionvolume10L,detection
Cell Plate Plate 254nm,cell:1L,backpressure5bar,sample(inorderofelution):
Column volume/L Tusp count/m count/% uracil, pyridine, aniline, 4-ethylaniline, benzene.
Purospher™ STAR RP-18e 1.4 1.15 43.000 119
(3 µm) Hibar® HR
5 cm x 2.1 mm I.D. 11 1.29 36.000 100
Purospher™ STAR RP-18e 1.4 1.24 44.000 126
(3 µm) LiChroCART® HR
5.5 cm x 2.0 mm I.D. 11 1.31 35.000 100

7
Sample injection, mass and Massoverloade^ectsdependonthesamplecomplexity,
solubility, and retentivity and are commonly observed
volume overload when trace compounds are analyzed in complex mixtures.
Quantitativeanalysesneedacarefulsystemsetup,In asthis situation, a desirable increase in sensitivity is
an improper combination of injection needle and sample achieved by simply increasing the injection volume. Con-
vial, as well as a high sample viscosity can cause false sequently, a mass overload with the main component is
sample drawing. For example, a combination of a high most likely. As long as the peak shape of trace compounds
draw speed and a highly viscous sample can cause sample isnotnegativelya^ectedandnooverlapofthetraceand
underdosing,because_llingoftheinjectorneedlewiththe main component peaks is observed, this phenomenon can
sample solution can be somewhat slower in comparison to be tolerated. A visible phenomenon of mass overload is
the draw speed of the dosing syringe. In this situation, the the detection of triangular shaped peaks displaying tailing
draw speed has to be decreased in order to enable proper orfronting.Infact,thee^ectofmassoverloadonpeak
sampling. A similar error can occur when the septum of shapeisin‘uencedbytheinteractionbetweenanalyte,
the sample vial is completely airtight and large sample mobile phase, and stationary phase and is a direct result
volumes (e.g., pharma samples) need to be taken. The of the adsorption isotherm of a chromatographic system
use of split septa, specially designed needles or puncture (analyte/mobile phase/stationary phase). The dependency
o^settuningviathesystemsoftwarecanbeofhelpinof the sample concentration in the mobile phase on the
this case. sample concentration in the stationary phase is described
by a Langmuir or anti-Langmuir isotherm, where peak tail-
Make sure that the autosampler syringe is free of any ing (Langmuir behavior) or peak fronting (anti-Langmuir)
airbubblesinordertoavoidanynegativee^ectsonthe can be observed. Depending on the characteristics of the
drawn sample volume. In order to avoid air bubbles, detector, mass overload can lead to a detector overload,
‘ushthesyringemanuallyandalwaysdegaswashing resultingin‘at-toppedpeaks.Thiscancompromisethe
solution prior to use. determination of plate count or resolution, for example.
The stability of, e.g., a UV detector signal varies and In such a situation the linear range of the detector can
isin‘uencedbybothtemperature(temperaturedrift) be increased (and an overload can be avoided) when the
and ageing of the UV lamp applied. Make sure the detection is not performed at the absorption maximum
lamp is thermally equilibrated (stable baseline signal) of an analyte.
prior to an analysis, and that standards are run directly Figure 14 displays a simulated, and rather weak, mass
beforequanti_cationexperimentsin ( orderofincreasingand volume overload experiment conducted by injecting
concentration). identicalvolumesofca^eineatdi^erentconcentratio
An increase of the volume or mass of a sample injected onto a particle packed column.
onto a column—or a decrease of column i.d. while main-
taining the injection volume constant—has a positive
in‘uenceonthesensitivityofananalysis.However,this
approach is limited, as at a certain point both mass and/
or volume overload can be observed in a chromatographic
separation. Under these prerequisites, retention capacities
and peak widths as well as resolution are no longer inde-
pendent from the amount of sample injected. As a rule
of thumb, when the injection volume does not exceed 1%
ofthetotalcolumnvolumethemaximumseparationea-
ciency of a column can be preserved. Table 3 combines
typical‘owratesandsamplemassandvolumeamounts
for the loading capacities of various column formats.
Table 3. Guidelines for typical ‘ow rates and sample mass and
tR tR
volume amounts for the loading capacities of analytical and
Figure 14.Mass(left)andvolumeoverload(right)e^ect,injection
semi-preparative columns.
ofca^einesamples.Injectedmassofca^eineandvolumeofca^eine
ColumnDimension Typical Flow Sample Sample increasing from bottom to top chromatogram. For better comparability
length
( xi.d.inmm) RatesmL/
( min) Amount(mg) VolumeL)( of peak shapes, the peak intensity was not normalized.
Chromatographicconditions:Purospher=STARRP-endcapped 81 m)3(
150 x 1 0.06 i0.5 0.05–1 Hibar® HR 10 cm x 4.6 mm I.D.; mobile phase: acetonitrile/water
250 x 2 0.25 i0.2 0.2–10 90/1(v/v);detection30nm;‘owrate1.0mL/min;temperature:
25°Cmassoverload:injectionvolume5L(sampleconcentrations
250 x 3 0.6 i1 1–20 0.2,51and50g/Lca^eine);volumeoverload:
concentration50g/Lca^eine(injectionvolume:0.2,51
250 x 4 1 i5 5–80
20,and50L).
250 x 10 6 i30 30–500
250 x 25 39 i20 200–3000

8
In contrast to mass overload, volume overload is observed The detection of traces of a previous sample injection in a
when the sample concentration is kept constant but the chromatogram is referred to as sample carry-over. Carry-
injected sample volume is increased. Volume overload overcannegativelya^ectquantitativeandqualitati
resultsinbandbroadening,‘at-toppedpeaks,orpeakanalyses, hence chromatographic columns should display
splitting,andnegativelya^ectsresolutionaswellasplate a low carry-over tendency. When calibration curves are
count (see Figures 15 and 16). A second e^ect of prepared running dilution series, lowest concentrated
volumeoverloadisadecreaseinseparationeaciency. samplesneedtobeanalyzed_rst.Systemconsumables
An additional visible phenomenon can be the so-called (seals, needle) as well as system parts such as connecting
detector overload correlated to the detector properties. capillaries, sample loops and especially the injection valve
In Figure 15, such a detector overload can be observed (stator and rotor seal) can be a source of sample carry-
for seven out of eight analytes (cropped peak tips). over. Although less likely, the chromatographic column
itself can also be the cause of carry over. If carry-
One distinct di^erence between mass and volume
over persists after replacement of these components,
overload is evident: In a mass overload experiment, plate
check for the compatibility of both the autosampler
count drops by one order of magnitude when increasing
wash solution composition and the sample properties.
mass load by one order of magnitude, whereas in a
Otheroptionstoovercomeacarry-overe^ectcanbe
volume overload experiment plate count drops by two
a change of column (selectivity), an adaption of the
orders of magnitude when increasing volume load by one
chromatographic method, the application of washing
order of magnitude. For this reason, it is recommended
steps between chromatographic runs or a needle wash
to work with concentrated sample solutions rather than
procedure. Biological samples (e.g., proteins, peptides)
with large volumes of a diluted sample. If injection of
should be analyzed in biocompatible HPLC systems, in
large sample volumes is necessary, it has to be made sure
which all metal parts are replaced by inert polymeric
that the sample is dissolved in a weak solvent in order to
components (e.g., made out of PEEK). Combining
achieve peak focusing on a chromatographic column.
biological samples with standard HPLC systems will
most likely cause sample carry-over. Figure 17 displays
thesamplecarry-overinananalysisofUV_ltersinsun
lotions on a particulate type HPLC column. After one UV
_lteranalysisrun,threeblankrunswereperformed.A
washing step is necessary in order to remove residual
Tinosorb S (chromatogram peak 4) from the particle
packed column after an overloading experiment. The
sample carry over observed in the _rst blank run is
caused by system parts between injector and column
including stainless steel frits of the column itself.
900
UV intensity (mV)

0 2 4 6 8 10 12 14
Retention time (min)
Figure 15.E^ectofvolumeoverloadonpeakshape.Frombottomto 600
top:10,25and10Linjectionvolume.
Chromatographic conditions: Purospher™ Star RP-18 endcapped (5 µm)
Hibar® HR 15 cm x 4.6 mm I.D.; detection: 247 nm; temperature: 40 °C;
‘owrate:mL/
3 . 1 min;mobilephaseA:water,mobilephaseB:acetonitrile; 300
gradient conditions: 0 min 45% B, 2.5 min 95% B; sample (in order of
elution): acetanilide, acetophenone, propiophenone, butyrophenone,
benzophenone, valerophenone, hexanophenone, heptanophenone.
0
100,000 0 5 10 15 20 24
1% of column volume 10% of column volume

80,000
Retention time (minutes)
Figure 17. Sample carry-over experiment as observed in a comparative
analysisofUV_ltersinsunlotionsonparticlepackedHPLCcolumn.After
60,000
aUV_lteroverloadinjection(topchromatogram,ingreen)threeblank
EaciencyN)(

runs were completed (chromatograms two to four, from top to bottom).


40,000 Chromatographicconditions:Purospher=STARRP-endcapped
81 m)3(
Hibar® HR 10 cm x 4.6 mm I.D.; mobile phase A: acetonitrile, B: water
+ 0.1% phosphoric acid; gradient: 0–9.6 min 67% A, 17.6 min 100% A,
20,000 min
02 A;%0 1 detection:UVnm;
213 ‘owrate:mL/
0.1 min;temper-
ature:ambient;injectionvolume:10L;sample:1EusolexOCR4 1
µg/mL, 2 Eusolex 2292 127 µg/mL, 3 Eusolex 9020 160 µg/mL,
0 4 Tinosorb S 92 µg/mL.
0 50 100 150 200
Injection volume (µL)
Figure 16.Decreaseinseparationeaciencywithincreasinginjection
volume shown on a 50–4.6 mm HPLC column and an analyte with k = 1.6.
Note that if 10% of the total column volume is injected, only about 20%
ofthecolumneaciencyremains.
Chromatographicconditions:SeQuant ® ZIC®-HILIC (5 µm, 200 Å) PEEK

5cmx4.6mmI.D.;detection:254nm;‘owrate:0.5mL/min;mobile
phase: Acetonitrile/ammonium acetate 5 mM 80/20 (v/v); sample: cytosine.

9
Thermostatting mobile phase temperature becomes large, a temperature
gradient may appear inside columns with PEEK housing
Thein‘uenceoftemperatureonachromatographicsepa- thatnegativelya^ectspeakshapeandseparationea-
ration cannot be generalized and an increase of tempera- ciency. Figure 18 shows the in‘uence of temperature
ture can, but does not necessarily lead to, an improve- on the plate height achievable with an RP-18 endcapped
ment of column performance, or a change of selectivity. 50–2 monolithic silica column. As a reason of this it is
Higher temperatures decrease the viscosity of the mobile recommended to install a metal capillary with 1/16‘‘ outer
phase, which can improve mass transfer and separation diameter and 0.2 mm inner diameter (length 30 cm) in
performance, but on the other hand, a loss in performance frontofcolumnswithPEEKhousing.Thehigherthe‘ow
or undesired changes in selectivity are possible. Working rate of the mobile phase, the more important a pre-heated
at elevated temperatures also reduces analysis time, mobile phase is. Another option for thermostatting of elu-
because an increased di^usion coeacient facilitates ents is the combined use of metal capillaries and a water
higher ‘ow rates of the mobile phase. When working bath and a column oven. Depending on the HPLC system,
with highly viscous eluents, an increase in temperature mobile phase pre-heating modules can be a general alter-
might allow for a chromatographic run that is not possible native to the use of metal capillaries (and a water bath).
under ambient room temperature conditions, as in the
caseofelevatedbackpressures,forexample.Low‘ow 30
40 °C w/o capillary 25 °C w/o capillary
rates,areproducibleandsuacientheattransferfrom 40 °C with capillary 25 °C with capillary
25

Plate height (µm)


column oven to column, as well as a pre-column eluent
heater are necessary for the successful implementation of
20
temperature in chromatographic analysis.
Thermostatting HPLC column compartments can be either 15
performed by an air bath (still or circulated air), or a block
heater unit, the latter providing better and faster heat 10
transfer. When combining columns with a block heater, the
5
columns walls display a quasi-isothermal behavior, which
means that the column wall temperature remains constant
0
independent from the mobile phase temperature. In 0.5 1.5 2.5 3.5 4.5 5.5
contrast, in still air thermostats, almost no heat exchange Flow rate (mL/min)
between air and column wall is observed; consequently,
Figure 18. Correlation between column thermostatting setup and plate
such a setup is referred to as quasi-adiabatic. In circulated height achievable with a Chromolith® RP-18 endcapped 5 cm x 2.0 mm
air-water baths, heat exchange between column wall and I.D. column.
air is possible, making this system a somewhat isothermal
setup. Chromatography at elevated temperatures aims
atmaximizingseparationeaciency.Duetothisdesire, Figure 19 displays the separation of doping drugs at
the combination of an adiabatic approach with an eluent an oven temperature of 45 °C. Depending on the eluent
preheater is mandatory in order to avoid the formation of tubing material and internal diameter used, obvious
a radial temperature gradient inside the chromatographic changes in the chromatographic results are visible.
column. In UHPLC experiments, friction of the mobile
phase leads to the generation of heat inside a column. The
application of isothermal conditions then results in a radial
temperature gradient that compromises chromatographic
0 1 2 3
results. In contrast, under adiabatic conditions, an
axial temperature gradient is generated that does not
negativelya^ectperformance.
0 1 2 3

However, it is worth mentioning that the temperature of


a separation should be adjusted to the properties of the
columnhousing,aswellastothespeci_ccharacteristics 0 1 2 3

of the column packing material. A polymeric column


backbone can be operated at higher temperatures without
column deterioration, whereas the solubility of silica-based 0 1 2 3
Retention time (min)
stationary phases increases rapidly under such conditions.
Of course, in addition to the temperature, the pH of an Figure 19. Analysis of a doping drug mixture on a monolithic silica
eluent has to also be taken into account. For details, see column at an oven temperature of 45 °C. Capillary utilized in front of the
column, from top to bottom: No capillary; metal capillary with 1 mm i.d.;
section on mobile phase composition. metal capillary with 0.2 mm i.d.; red PEEK capillary with 0.17 mm i.d.
The length of all capillaries used was identical (30 cm).
Whatever system is used, it is important to work under
Chromatographic conditions: Chromolith® HighResolution RP-18 endcapped
equilibrated conditions. The heat conductivity of a stain- 10 cm x 4.6 mm I.D.; mobile phase A: acetonitrile, mobile phase B:
less steel column housing is high, therefore stationary water;gradient:A% 5 3 toA% 0 1 inmin,
1 min
5 . 3 atA;% 0 1 ‘owrate
phase temperature most likely matches column oven 2 mL/ min; temperature 45 °C; detection: UV 240 nm; injection: 5 µL;
temperature. On the other hand, the heat conduction sample:‘uoxymesterone,boldenone,methandrostenolone,testosterone,
coeacientofcolumnsutilizingPEEKhousingisloweras methyltestosterone, boldenone acetate, testosterone acetate, nandrolone
propionate, testosterone propionate, nandrolone phenylpropionate,
comparedtosteel.Ifthedi^erencebetweenovenand testosterone isocaproate.

10
System care and maintenance Pumpdebrisiscollectedinthepumpinlet_lter.These
compounds might not be visible using UV detection, but
A system suitability test (SST) should be performed itislikelythattheycanbedetectedviaMS.The_lter
with any HPLC system and needs to be independent should therefore be replaced every 1–2 months, or after
of its use. The SST delivers information about the changing from acetonitrile to methanol (or vice versa)
suitability of a combination of a chromatographic in order to obtain lower baseline noise and to generally
column and an HPLC system in terms of selectivity or protect the system including column and detector.
otherprede_nedcriteriaforaspeci_ctypeofsample
under the given chromatographic and instrumental HPLC systems are equipped with a solvent mixture bottle
conditions. In a pharmaceutical environment, such an dedicated to autosampler washing. This mixture normally
SST is mandatory and has to be conducted regularly contains water and approximately 5–10% of an organic
(every workday morning), or prior to a new analysis solvent such as isopropanol for better wettability. It is
sequence. The chromatographic properties of the common practice in many laboratories to keep the compo-
sample utilized in such a test have to be similar to the sition of this mixture constant and utilize it independent
real samples analyzed in subsequent runs. of the type of method (isocratic or gradient) as well as
stationary phase (reversed or normal phase as well as
Knowledge about the system back pressure under HILIC). After an autosampler needle-washing step, part of
speci_cconditionsallowsforacalculationofnet the washing solution is transferred to the chromatographic
column back pressures and a good comparability of system. As long as chromatography is done in reversed
di^erentchromatographiccolumnsunderidenticalphase mode (the organic solvent is the strong solvent),
chromatographic conditions. Changes in system back lowamountsoforganicsolventwillhardlya^ecttheresult
pressure can help to identify system wear. The system of an isocratic or gradient run. By contrast, water is the
back pressure can be determined by performing an strong eluent under HILIC conditions. If the autosampler
HPLC run without installed chromatographic column. washingsolutioncompositionisnotadapted,thee^ect
The HPLC instrument has to be ‘ushed with organic will be comparable to the injection of a sample dissolved
solvent regularly to prevent microbial contamination in a highly aqueous solvent mixture. This can cause peak
anditsnegativee^ects,mainlyonhighlysensitivemass shape issues such as peak splitting or broadening, as well
spectrometric detection. Use alcohols such as methanol as decreased retention. Therefore, the amount of strong
or isopropanol for this means. To a certain degree, solvent in the autosampler washing solution (as well as in
acetonitrile can be contaminated with amines, these the sample solvent itself) has to be kept lower than the
adsorb on the sapphire seats and balls of check valves, content in the mobile phase starting composition.
subsequently polymerize and can block inlet valves. When preparing a sample for reversed phase analysis,
Ceramiccheckvalvesdonotseemtobea^ectedina the sample has to be dissolved completely; ideally in
similarmanner.Thefrequencyof‘ushingdependson the mobile phase (gradient runs: initial composition).
theutilizedeluentsandbu^erconcentration,andshould Solubility of the sample in the eluent has to be tested
be between two to four weeks. If possible, at least 5% of prior to injection. If the sample cannot be dissolved in
organic solvent should be added to the aqueous mobile the eluent, a solution in less than 50% acetonitrile is
phase. An inversion of eluent channels can also help to the best choice. The second choice is 100% water (used
avoidmicrobialcontamination.Ifabu^erwasusedprior up to 500 µL) and avoid (close to) 100% acetonitrile.
toinstrument‘ushing,makesurethesaltissolublein
the organic solvent or that water is used for ‘ushing Under HILIC conditions, the sample has to be dissolved
before switching to an organic solvent. If an HPLC is in 60–100% organic solvent or the initial mobile phase
notusedforprolongedperiodsoftime,‘ushtheentire composition. The sample should not be diluted in water!
instrument with alcohol. Again, always test sample solubility in the mobile phase
prior to injection. When performing a gradient run, the
steepness should not exceed 2–5% eluent composition
change per minute in order to keep the system in
equilibrium.

11
Figure 20illustratesthee^ectofimpropersample amounts of sample dissolved in the mobile phase
solvent composition in the separation of the antiviral (acetonitrile/ammonium acetate 10 mM 90/10 v/v) were
drug oseltamivir and oseltamivir carboxylate on a injectedwithnonegativee^ecttothechromatographic
zwitterionic HILIC column. As water is the strong results. By contrast, injection of samples dissolved in
solvent in this chromatography mode, water content of acetonitrile/ammonium acetate 10 mM 50/50 (v/v) as
the sample solution should be kept as low as possible. a solvent under otherwise identical conditions, led to
In the example shown in the left column di^erent the elution of broad and even splitting peaks.

1.5e5 1.5e5
MS intensity

MS intensity
(counts)

(counts)
1.0e5 1.0e5

0.5e5 0.5e5

0.0e5 0.0e5
0 1 2 3 4 0 1 2 3 4
Retention time (minutes) Retention time (minutes)

1.5e5 1.5e5
MS intensity

MS intensity
(counts)

(counts)
1.0e5 1.0e5

0.5e5 0.5e5

0.0e5 0.0e5
0 1 2 3 4 0 1 2 3 4
Retention time (minutes) Retention time (minutes)

1.5e5 1.5e5
MS intensity

MS intensity
(counts)

(counts)

1.0e5 1.0e5

0.5e5 0.5e5

0.0e5 0.0e5
0 1 2 3 4 0 1 2 3 4
Retention time (minutes) Retention time (minutes)
Figure 20. Multiple reaction monitoring mode analysis of oseltamivir and oseltamivir carboxylate on a zwitterionic HILIC column. Sample diluent:
Acetonitrile/Ammonium acetate 90/10 (v/v) (left), acetonitrile/NH4OAc0/5(v/v)(right).Injectionvolumes:2.5L(top),5.0L(middle),
7.5L(bottom).
Chromatographicconditions:SeQuant ® ZIC®-HILIC(5m,20Å)PEEK5cmx2.1mmI.D.;detection:MS(multiplereactionmonitoringmode,

extractedionchromatogramoverlayofm/z31.and25.);mobilephase:acetonitrile/Ammoniumacetate10mM90/1(v/v);‘owrate
500 µL/min; sample: oseltamivir, oseltamivir carboxylate.

12
3. Method development & optimization
Theoverallprocessisin‘uencedbythenatureoftheThese initial questions will direct the chromatographers
analytes and generally follows the following steps: tode_nethemethodgoal,andto_ndoutrequirements
of the new method. Is high resolution (in separation and
1. Selection of the HPLC method and system
detection), short analysis time, maximum sensitivity, long
2. Sample preparation column lifetime, and/or a column with wide pH stability a
3. Selection of the detector need, or will the method be used at neutral pH and under
4. Selection of initial conditions non-aggressive conditions? True optimization of a method
isabalancebetweenselectivity,speed,andeaciency
5. Choose the right HPLC Column
in order to produce results that are the purpose of the
6. Mobile phase selection application. Ideally, the development should result in a
7. Mobile phase preparation robust method that gives the laboratory a low, overall,
8. Selectivity optimization price-per-injectionandultimatelyacost-eacientassay.
9. System optimization
Common mistakes in analytical method
10. Method validation
development
11. Scaling of HPLC methods
• Inadequate formulation of method goals
• Insuacientknowledgeofchemistry
Selection of the HPLC method • Use of whatever reversed phase HPLC column is
and system available in the lab
Methoddevelopmentisnotdiacultwhenaliterature • Use of wrong instrument set-up
reference can be found for the same or similar needs.
Methods are published in pharmacopeia, in column • Trialanderrorwithdi^erentcolumnsandmobilephases
manufacturer application databases, and as published These mistakes often result in laborious, time-consuming
scienti_c studies. These sources can provide good projects that lead to methods that fail to meet the needs
guidance for the planned work, but what happens when of the laboratory.
references to the compounds of interest do not exist?
Di^erentapproachesarepossible,andtrialanderroris Getting started
the least successful way forward. A chromatographer After de_ning the goal of the method development,
normally has access to a wide variety of equipment, speci_c information of the sample and the analytes
columns, mobile phase compositions and operational should be sought. Di^erent sources are available:
parameters which make high performance liquid e.g. scienti_c journals, chemical databases such as
chromatography method development seem complex. www.pubmed.org (small molecules), ExPASy Proteomics
In this chapter, direction will be given to make method Server http://expasy.org (large biomolecules), and
development intuitive and successful, with emphasis
reference books. Listed below are some of the most
on column selection.
common parameters.

Method goals • Nature of the sample


Methoddevelopmentinthiscontextmeanstode_ne• Number of compounds/analytes present
needs, set goals, and make experimental plans, then to
carryoutthepracticalwork,and_nallyvalidateandput• Chemical structure (functionality)
the new method into routine work. For these reasons, • Molecular weight of the compounds
method development should be started at the desk, and
notinthelaboratory.Anumberofquestionsshould_rst • pKa values
be addressed and answered: • log P and/or log D values (hydrophilicity/hydrophobicity)
• Is the primary goal quantitative or qualitative analysis? • Concentration
• If quantitative analysis is required, what levels of
accuracy and precision are needed? • Sample matrix
• Are reference standards available? • Sample solubility
• How many analytes need to be detected?
Depending on the method requirements, some steps
• Is it necessary to resolve all sample components? will not be necessary. For example, if a satisfactory
• Howmanydi^erentsamplematricesisthemethod separation is found initially, steps 7 and 8 may be omitted.
designed for? The extent to which method validation (step 9) is
• How many samples will be analyzed at one time? investigatedandpursuedwilldependonthe_naluseof
• If qualitative analysis is requested, it is important to the analysis; for example, a method required for quality
de_newhetherthemethodwillbeusedforcharacter- control will require more validation than one developed
ization of unknown sample components or isolation/ foraone-o^analysis.
puri_cationofanalytes.

13
Sample Preparation Overview of Techniques
At a glance, sample preparation enables the following
advantages: Filtration
Filtration is a rather simple but essential component of
• Removal of undissolved particulates that could lead
high-qualityseparationandpuri_cationprocessesforthe
to instrument down-time
removal of undissolved matter and particles from samples
• Decrease all/most of impurities that would interfere priortoHPLCorUHPLCanalysis.Therefore,a_ltration
with the analyte during analysis step has to be part of every sample preparation procedure
• Increase the sensitivity or enrichment of the desired and depending on the throughput of a laboratory syringe
analyte(s) _lters, complete _ltration systems or _lter plates ar
available.Thetypeof_ltrationmembranepore ( diameter
Sample preparation is generally the most time-consuming or
02. m)54.0 shouldbechosenaccordingtospeci_c
and tedious portion in the analysis for small molecules. sample properties:
But it is one of the most, if not the most, important
• LCR (hydrophilic PTFE): Aqueous or mild organic
considerations in the process. The ideal sample prep-
solutions;lowbindingandextractables,_ltrationof
aration prior to analysis increases the compatibility of
protein-containing solutions.
the analyte with the detection technique and removes
interfering impurities from the matrix. Addressing these • Durapore® (PVDF): Aqueous or mild organic
factors allow for cleaner spectra which enables greater solutions; low binding and extractables, clarifying
sensitivity. Further, certain analysis techniques require protein-containing solutions.
the analyzed sample be particle-free meanwhile
the prepared sample needs to be miscible with the • Nylon: Aqueous or organic solutions
resultant technique. • Express=(polyethersulfone(PES):Fast‘owandlow
An important note about the matrix of the analyte sample. protein binding
The matrix tolerance of chromatographic columns and • Fluoropore® (hydrophobic PTFE): biologically inert
detectorsdi^ersdependingonthetypeandproperties with broad compatibility. Compatible with acids,
of the carrier material (monolithic or particulate), and bases, and solvents
on the type of detector used (UV, MS etc.). Especially
• MixedCelluloseesters(MCE):clari_cationofwater,
when utilizing highly sensitive mass spectrometric
detectionandquanti_cation,thesamplepreparation bu^ers,oraqueoussolutions
process has to be performed thoroughly, as matrix
components can cause signal suppression and/or Syringe _lters with high-density polyethylene or
adduct formation with target molecules and therefore polypropylenehousingaresuitableforthefast_ltration
decrease sensitivity (signal-to-noise ratio) and/or of a rather limited number of samples (1–10 per day).
increase complexity of the mass spectrum. Due to Their chemical compatibility is broad and low holdup
this, the chromatographic analysis of samples with volumes make them ideal candidates for the preparation
highmatrixloadcanmakeseveraldi^erentselective of low sample volumes. In addition, e.g. greater than
andspeci_csamplepretreatmentstepsnecessary. 90%drugrecoveryinthe_rstmLof_ltrateispossible
During the sample preparation procedure, undesirable whenutilizing_lterswithPTFEmembrane,indicating
compounds such as particles, lipids or dissolved matter low drug binding to PTFE, making this membrane type
shouldberemoved.Extractionandpuri_cationstepsan asideal candidate for sample preparation prior to
well as preconcentration procedures of low abundance quantitativeanalyses.Syringe_ltersarealsoavailablefor
analytes can be part of the clean-up procedure. Depending roboticsystemsorworkstations.Inthatcase,_ltersare
on the physical state of the sample, various sample usedincombinationwithanautomated_lterchanging
preparation techniques are available: system, and for applications such as dissolution testing
(evaluation of the dissolution rate of solid dosage forms
Liquid samples:
in the digestive tract) or HPLC sample preparation.
• Filtration Nexttotheabove-mentionedmembranes,aglass_ber
• Liquid-liquid extraction (LLE) membranefortheclari_cationofaqueousororganic
• Solid phase extraction (SPE) solutions with high particulate levels is available.
• Solid phase microextraction (SPME) Forhard-to-_ltersamplessamples
( containingparticulate
• Restricted access materials (RAMs) materials as well as samples that are viscous), syringe
_ltration of food and beverage samples such as, e.g.,
Solid samples: juices, honey, soups or salad dressing can be diacult
and the particle load can easily clog the syringe _lter.
• Soxhlet extraction, batch extraction Otherhard-to-_ltersamplesincludemanypharmaceutical
• Matrix solid phase extraction suspensions, shampoos, conditioners, creams and other
household products. Under these circumstances, vacuum-
After extraction of the analytes, the sample may be driven_ltrationofsamplesforliquidchromatographywith
concentrated by evaporation of the solvent. This allows speci_callydesigned_ltrationsystemsand_lterscanbe
the sample to be resuspended in a desired solvent, i.e. an option. Same as for the syringe _lters, low hold-up
the initial mobile phase for liquid chromatography. volumes of such devices deliver high analyte recoveries.

14
Incontrasttosyringe_ltration,manysamplescanbe • Easy handling
treatedatthesametime,making_ltrationsystemsa
• No formation of emulsions
good solution for non-automated, medium throughput
(10samplesperday).Recommended_eldsofuse • High recovery rate and cleaner eluates in comparison
are, for example, drug dissolution testing, food safety to “bulk” LLE
(unknown and known toxin), cosmetics (ingredients and
formulations) and pharmacokinetics/pharmacodynamics • Lower solvent consumption
(PK/PD, drug interaction with the human body). • High batch-to-batch purity and reproducibility

Sorption-based sample preparation


Sample_ltrationisnormallyfollowedbyasecondsample Solid Phase Extraction (SPE)
preparation step depending upon the analytes. After SPE is the most widely used technique for sample clean
adsorption, the analytes are desorbed utilizing small up or preconcentration of analytes from aqueous or liquid
volumes of a suitable solvent which are collected and samples. It can be utilized for analytes covering the
analyzed. The sample preparation procedures in the complete range of polarities and chemical structures and
following section are technically mature and belong to the combinesahighrecoveryrateande^ectiveconcentration
most widely used processes in laboratories worldwide. without the formation of emulsions. In addition, solvent
Improvements in these procedures are aimed at consumption of this easy-to-use and automatable method
increasing degree of automation and/or sensitivity. is very low.
The goal of SPE is the selective extraction of the analyte(s)
Liquid-liquid extraction
from a complex sample for cleaner and more reliable
Classical liquid-liquid extraction (LLE) is a common results by a further analytic technique (e.g. HPLC or
choice when it comes to the treatment of liquid samples. LC-MS). Overall, SPE works on the principle of liquid
It is one of the _rst sample preparation procedures chromatography where analyte(s) are retained by
established and is still used in its original form for the reversable interactions between the analyte(s) and the
analysis of biological samples. LLE “in bulk” is based sorbent surface. These interactions include, but are not
on the transfer of an analyte from an aqueous solution limited to, van-der-Waals forces, hydrogen bonding, dipole-
to a water insoluble solvent using a separation funnel. dipoleforces,andelectrostaticinteractions.Thedi^erent
Disadvantages of this time-consuming approach include sorbents used for SPE contain one or more of these
the formation of emulsions, poor phase separation and intermolecular forces and will be highlighted in more
a comparably high solvent consumption. detail below.
AnotherLLEapproach(thatisin_rstsightsimilartosolid When starting SPE, the more information known about
phase extraction (SPE) procedures (see section below)— the analyte(s) makes selection of the sorbent easier.
uses a solid support, in most cases natural diatomaceous Useful properties include information about the structure,
earth, as a matrix for the adsorption of the aqueous solubility, polarity, lipophilicity, log P’s (hydrophilicity/
sample. The sample distributes itself in the form of a hydrophobicity), concentrations, and pKa’s. Other
thin_lmoverthechemicallyinertmatrixandthusacts factors relating to the conditions of the solution that
as a stationary phase. In a subsequent step, all lipophilic the analyte are in are also important. These include the
analytes are eluted with an organic solvent that is not polarity (aqueous versus organic), ionic strength, matrix
miscible with water (e.g., diethyl ether, ethyl acetate, interference (lipids, salts, fats), and pH to name a few.
halogenated hydrocarbons), then further cleaned up and Table 4 shows one approach to narrowing down what
analyzed. Figure 21 illustrates this process. The advan- type of sorbent to use for SPE. Table 5liststhedi^erent
tages of this type of LLE (also known as SLE or types of sorbents that are available along with their
supported liquid extraction) are: description and general applications.

1. Extrelut® 2. Apply aqueous sample 3. Apply organic solvent 4. Elution


(not miscible with water)

Distribution of the aqueous Lipophilic substance extracted


phase on Extrelut® from the sample
Figure 21. Working principle of Supported-liquid extraction. The sorbent applied is a naturally occurring, pure, wide pore kieselgur (diatomaceous earth).

15
Table 4. Flowchart to help decide which sorbents are best for analyte(s) separation.

SampleMatrix Aqueous(polar,bu^er,water) Organic(slightlypolartonon-polar)


RetentionMechanism Reverse Phase Ion Exchange Normal Phase
Strong Ions Weak Ions
Analyte Property Slightly polar to non-polar Polar to moderately polar compound
Cation Anions Cations Anions
C18, C8, C4, Ph, CN,
SorbentPhase SCX SAX, NH2 WCX NH2, PSA Si, CN, Diol, NH2, PSA, Florisil, Alumina
DPA-6S, HLB

Table 5.Outliningthedi^erentsorbentswithpotentialapplications.

Sorbents
Available Description Applications
Extractions of nonpolar to moderately polar compounds including antibotics,
C18 Octadecyl bonded, end capped silica
barbiturates, essential oils, steroids, surfactants
C8 Octyl bonded, end capped silica Similar to the C18 but slightly less hydrophobic.
ReversePhase

C4 Butyldimethyl bonded, end capped silica Less hydrophobic than C8 and C18, extraction of peptides and proteins
Used to adsorb polar compounds (hydroxyl and phenolic compunds) from
DPA-6S Polymeric hexamide bonded
aqueous and methanolic solutions through strong hydrogen bonding.
Ph Phenyl bonded silica LessretentionthanC8orC18,slightlymorespeci_cforaromaticcompounds
For very hydrophobic analytes that may be irreversibly retained on more
CN Cyanopropyl bonded
hydrophobic sorbents such as C18
Hydrophobic surface enclaved by
HLB For wide range of polar to non-polar analytes.
hydrophilic network
Polymer bonded Used to extract polar analytes through hydrogen bonding from organic
Diol
2,3-dihydroxypropoxypropyl solvents, oils, and lipids
Used to extract polar analytes or analytes with hydroxyl, amines, or
Si Silica gel
heteroatoms.
NH2 Aminopropyl bonded silica Used to extract moderately polar to polar compounds
NormalPhase

Polymeric bonded ethylenediamine—


PSA N- propyl (primary and secondary amine) Similar to amine phase, but higher capacity due to presence of secondary amine
bonded silica
Less retentive than Si or Diol and allows for rapid release of very polar analytes
CN Cyanopropyl bonded
that may irreversibly be retained on very polar sorbents.
No bonded phase, available at neutral pH Extraction of polar compounds such as vitamins, antibiotics, essential oils,
Alumina
(6.5) adsorbent glycosides
Magnesium silicate Medium polar extraction of analytes including alcohols, aldehydes, amines,
Florisil
available as adsorbent ketones, organic acids, and phenols
For extractions of anions, organic acids, nucleic acids, nucleotides and
Quaternaryaminebondedsilicawith surfactants. Elution solution must have a pH < pKa +2 of the analyte and/or
SAX
Cl- counterion (pKa >14) ionic strength > 0.1 mol/L. Not intended for recovery of strong anions (use an
NH2 if intention is recovery)
For extractions of cations when acting as a weak anion exchange, loading of
NH2 Aminopropyl bonded silica (pKa ~ 9.8)
analyte must have a pH <7.8. Elution solution must have a pH >11.8
Polymeric bonded ethylenediamine—
IonExchange

For extractions of cations when acting as a weak anion exchange, loading of


PSA N- propyl (primary and secondary amine)
analyte must have a pH <8. Elution solution must have a pH >13
bonded silica (pKa 10.1 and 10.9
For extraction of cations, antibiotics, drugs, organic bases, amino acids,
Aliphatic sulfonic acid bonded silica with catecholamines, nucleic bases, nucleosides, and surfactants.
SCX
Na+ counterion (pKa < 1) Forelution,solutionmusthaveapHlpKa+2oftheanalyte.Notintendedfor
recovery of strong cations (use a WCX if intention is recovery).
For extrations of cations, amines, antiobiotics, drugs, amino acids, catecholamines,
Carboxylic acid bonded silica with Na+
WCX nucleic acid bases, and nucleosides. Elution solution must have a pH <2.8 and/or
counterion (pKa ~ 4.8)
ionicstrengthl0.1mol/L
Carbon-Based Nonporous with variety of surface area For adsorption of polar and non-polar compounds, depends more on structure/
Packing available Adsorbent size of analyte than intermolecular forces.
For extraction of hydrophobic compounds which contain some hydrophilic
Styrene/divinylbenzene
Chrom P functionality (e.g. aromatics). Elution usually performed with mid- to non-polar
Adsorbent
solvent
Extractionofmulti-residuepesticideanalysis,samplesare_rstextractedwith
Dispersive SPE (dSPE) available with
Other

QuEChERS aqueoussolventsinthepresenceofhighamountsofsaltsand/orbu^ering
di^erentsaltsand/orbu^eringagents
agents followed by SPE clean up.
Molecular MIPs
TheseincludedsorbentsforNSAIDS,nitroimidazoles,‘uoroquinolones,
Imprinted Specialized sorbents whose surfaces has
Beta-agonist,ribo‘avin,aminoglyconsides,andbisphenolA.
Polymers beendesignedtomimicspeci_canalytes.
Designed for the resolution of cis/trans isomers. The cis isomer is better
Chiral Ag-Ion retained on the sorbent compared to the trans isomer. One application is the
fractionation of fatty acids methyl esters (FAMEs)

16
As seen in Table 4, there are three major types of achieved by use of a polar solvent that disrupts these
retention methods: reverse phase (RP), normal phase interactions. Solvents that are commonly used include
(NP), and ion exchange (IE). We will discuss these acetic acid, water, and methanol. Similar to RP, secondary
methods in greater detail. Listed in Table 6 are interactions involving silanols and its deprotonated state
solvents commonly used for SPE in order from most may exist and similar strategies may need to be explored
non-polar to polar in nature as well as the type of if recovery is lower than expected.
separation they are preferred for.
Reverse Phase (RP): RP separations involve a polar Ion Exchange (IE)
matrix/solution (usually aqueous but can also be biological Ion exchange sorbents are used for analytes that are
samples such as plasma or urine) and non-polar or mildly charged in solution (usually aqueous, but sometimes
polar analyte(s). The analyte(s) are retained on the polar organic). Depending on the charge of the analyte
appropriate non-polar sorbent primarily due to Van der in solution this will dictate which type of sorbent is
Waals forces or dispersion forces. Recovery of the analyte required. If the analyte in solution is an anion (negatively
is achieved by using a non-polar solvent that the analyte charge) then a strong anion exchange (SAX) or amine
is miscible in. Common solvents for elution range from (NH2 or PSA sometimes referred to a WAX) sorbent
hexane, tetrahydrofuran, ethyl acetate, or acetonitrile is required. If the analyte instead is positively charge
depending on the analyte(s). When working with RP (cation) than the sorbent to use is either a strong
bonded silicas, some polar secondary interactions with cation exchange (SCX) or weak cation exchange (WCX).
the residual silanols may occur if the elution with the Retention for all four of these sorbents is primarily
nonpolarsolventdoesnoteacientlyelutetheanalyte. electrostatic interactions where the analyte is attracted to
The addition of a more polar solvent (methanol) may the opposite charge on the sorbent. The pH of the analyte
aid in disrupting these secondary interactions. Further solution and the sorbent is a critically important criteria
considerationscouldincludeusinganorganicmodi_erto when performing IE. Table 7 provides an approximation
either increase or decrease the pH. The silanol (Si-OH) will of the pH’s to use while performing IE. At the same time,
deprotonate above pH ~4. Under these circumstances, the the ionic strength of the sample and loading solution are
column may have slight cation exchange properties. This important. If the ionic strength is too high, then poor
can be overcome by using a mixture of acidic methanol or retention of the analyte(s) to the sorbent will occur.
basic methanol or by mixing these with a more nonpolar,
In general, strong ion exchange sorbents (SAX or SCX),
methanol-miscible solvent. A good suggestion is to keep
should only be used with strong anions/cations when
theorganicmodi_ertolessthan2%byvolume.Ifthe
recovery and elution is not important. If recovery of
sample recovered is going to be used for analysis by
these species are important, than a weak ion exchange
mass spectrometry, consider using a modi_er that is
sorbent is recommended. Strong ion exchange sorbents
compatible (e.g. ammonium formate for acids, ammonium
are good for working with analytes that can have the
hydroxide for bases).
charge changed depending on pH. This is highlighted
Normal Phase (NP): NP procedures involve polar ana- in Table 7. The strong ion exchange sorbents have a
lyte(s) in a mid-to non-polar matrix/solution (acetone, loading capacity ~0.2 milliequivalence/gram of sorbent.
chlorinated solvents, hexanes). In this case, the sorbents Equivalenceisde_nedastheconcentrationmultipled
areusuallysilicaormodi_edsilica(CN,2NH , or Diols) by the charge of the ion. For example, a solution of
that enable either hydrogen bonding, dipole-dipole, or 0.2 mM sodium ions and 0.2 mM magnesium ions
dipole-dipole-induced interactions with the polar (more would have equivalence of 0.2 meq (0.2 mM x [+1])
hydrophilic) analyte(s). Recovery of the analyte(s) is and 0.4 meq (0.2 x [+2])

Table 6. Characteristics of Solvents Commonly Used in SPE for RP or NP.

Solvent MiscibleinWater?
Non-Polar Strong RP Weak NP Hexane No
Isooctane No
Carbon tetrachloride No
Chloroform No
Methylene chloride
No
DCM (dichloromethane)
THF (tetrahydrofuran) Yes
Diethyl ether No
Ethyl acetate Poorly
Acetone Yes
Acetonitrile Yes
Isopropanol Yes
Methanol Yes
Water Yes
Polar Weak RP Strong NP
Acetic Acid Yes

17
Table 7. Methodology of using Ion Exchange sorbents and relative pH’s to use.

Analyte Weakacid(-)pKa~5 Stronganions WeakbasespKa


) + ( ~10 Strongcations
Sorbent SAX NH2/PSA/WAX SCX WCX
Approx. pkA pKa ~1 pKa ~10 pKa ~14 pKa ~ 5
charge state always + + at pH < 8 always – – at pH >7
Loading Soln pH >7 pH <8 pH <8 pH >7
Absorption
Analyte (charge) Neg (–) Neg (–) Pos (+) Pos (+)
condition
Sorbent (charge) Pos (+) Pos (+) Neg (–) Neg (–)
Loading Soln pH < 3 pH > 12 pH > 12 pH < 3
Desorption
Analyte (charge) Neutral (0) Neg (–) Neutral (0) Pos (+)
condition
Sorbent (charge) Pos (+) Neutral (0) Neg (–) Neutral (0)
Summary Analyte(s) changes charge Sorbent changes charge Analyte(s) changes charge Sorbent changes charge

Conversely, the use of weak ion exchange columns applicable,centrifugeand/orpre_ltersamplesprior


are generally better for strong ions (whose charge is to loading. Additional details about pretreatment of
mildlye^ectedbypH)asretentionandreleaseofthe samples will be discussed later.
analyte(s) is controlled by charge manipulation of the
3. Washing. This step helps eliminate impurities that
sorbent. This is not to say that weak ion exchange
would impact the results of the desired analyte(s). It
sorbents cannot be used for weak ions. Consideration
is important to have an idea of what the impurities
of the sorbent’s and analyte’s charge state with the
mightbeandhowtheirpropertiesdi^erfromthe
elution solution’s pH are important. Alternative, the
desired analyte(s). Example, if using a RP mechanism,
appropriate ionic strength may also be used to displace
the impurities may be less retained than the desired
the analyte. These ion exchange capacities should be
analyte(s). This is can be exploited by using a slightly
determined for each individual application.
morepolarsolventtowasho^theimpurity,leaving
behind the desired analyte. Another example involves
Solid Phase Extraction Methodology exploitingdi^erencesinpKa,s.Ifthesorbentis
In general, there are four steps when performing SPE. washed with a slightly acidic/basic solution, it could
These 1. Conditioning/Priming, 2. Loading 3. Washing, be used to change the charged state on the impurity
and 4. Elution. andalsotheaanityforthesorbent,leavingbehind
the desired analyte(s). Table 6, lists some common
1. Conditioning and priming the sorbent. Conditioning
solvents and their applicational use for SPE.
is vitally important when working with silica-based
sorbents and is encouraged when desired recoveries 4. Elution. Two smaller aliquots compared to one larger
or separations are not being achieved. Conditioning aliquot generally elutes the desired analyte(s) more
“wets” or solvates the sorbent and equilibrates the eaciently. Flow rate, once again, is an important
sorbent with similar solvent strength and/or pH to factor. The longer the elution solution/solvent is in
optimize retention. During this time, it is important contact with the sorbent, the better the expected
to not completely dry the sorbent out, otherwise it recovery of the desired analyte(s) will be. Another
will defeat the purpose. As general rule, conditioning consideration is whether or not the analyte is less
should be performed between 1 and 2 volumes if retained than the impurities. If this is the case, it is
using a cartridge (ex. Using a 10 mg/1 mL cartridge important to choose an eluent that will be selective for
would suggest 1 to 2 mL of conditioning). the analyte(s) and leave the impurities on the column.
The elution process also provides an opportunity to
2. Loading the sorbent. Depending on the sample,
increase the concentration of the analyte. This is
itmaybeloadeddirectlyonthecolumn.The‘ow
achieved if a smaller volume if feasible is used than
rateisimportant.Theideal‘owrateisonedrop
the volume of sample used during the loading step.
per second when time is not a factor and should
not exceed 2 mL/min in most cases. As a general Withanoverviewofthedi^erentstepsthatarecommon,
rule, the loading capacity of the analyte should therearethreedi^erentschemesthataremostoften
not exceed 5% of the bed weight of the sorbent implored. These are selective extraction, selective
(this does not apply for ion exchange sorbents as washing, and selective elution as seen in Figure 22.
it depends on the meq/g of sorbent, see earlier
discussion). As an example, if the bed weight is In selective extraction (Figure 22, left), the desired
100 mg of sorbent, this implies no more than 5 mg sorbent will bind the selected components of the sample,
of analyte is recommended to be loaded on the which can be the analyte(s) of interest or the impurities.
sorbent. If the sample is rather viscous or the pH is The selected component will be retained on the sorbent
not amenable to the column, presample preparation whiletheebuentwillcontaintherestofthesolution
should be considered. This may involve diluting Ifthedesiredanalyteisretained,thentheebuentis
the sample to reduce the viscosity or adjusting the discarded, and the analyte is collected separately in a
pH to the appropriate range (especially important new collection vessel when released from the sorbent.
for ion exchange). To avoid clogging frits or disks if However, if the impurities are retained and the analyte is
intheebuent,thisiscalledthepass-throughmethod.

18
Loading Loading Wash Wash Elution

Impurity Analyte Sorbent

Figure 22. Visualization of Selective Extraction (left), Selective Washing (Middle) and Selective Elution (Right). The legend of the symbols is
located below the image.

In selective washing (Figure 22, middle), the sorbent will available such as glass. Glass is recommended when
bind both the analyte(s) of interest and impurities when there is concern about trace amounts of leachables
loaded. During the wash step, an appropriate solution is from polypropylene.
added where the impurities are selectively removed, and
theanalyte(s)isretained.Theebuentisthendiscarded,
Well Plates. Similarly
and the analyte is collected separately when released
to cartridges, these are
from the sorbent.
available in a variety of
In selective elution (Figure 22, right), the sorbent will sorbents and bed weights.
bond the analyte(s) of interest and the impurities once These are recommended for
again. Washing steps are selected to remove unwanted processing many samples at a
matrix, while leaving the impurities and analyte(s) of time and are amenable to high throughput automation
interestonthesorbent.Theebuentuptothispoint liquid
is handling systems. These are tailored for smaller
discarded. At this time, a solution/solvent is selective for volumes (less than 2 mL).
the analyte(s) while leaving the impurities on the sorbent.
Once this is completed, further post-elute treatment may
Disks. Available in a variety of
perform at this time. This may include concentration or
sorbents embedded in glass
drying the sample or diluting/resuspending the sample
_ber membranes. Enables
in starting mobile phase.
faster ‘ow rates and less
clogging than PTFE disks
Format of sorbent while extracting organic
Thereareseveraldi^erentformatsinwhichthesorbent analytes/contaminants from
is available, including free sorbent, columns/cartridges, larger aqueous samples.
disks, 96-well plates, and online/inline cartridges. In
addition to the format, the sorbents are also available
indi^erentbedweightsthatallowfordi^erentsample Online/Inline. An alternative
volume capacities. to manual SPE. These are
designed for direct injection of
Columns/cartridges. These come in a variety untreated samples for analysis by
ofsizeswithdi^erentamountsofsorbents LC-MS.Theseareavailableindi^erent
loaded in the cartridge. Bed weights typically sorbents, including one designed to remove
range between 30 mg up to 10 g (see Table 8). phospholipids from biological samples. This is an
The smaller the bed weight, the smaller amount automated SPE which leads to high reproducibility and
ofelutionvolumesrequired.Thisisbene_cial consistent results by removing the human factor. Initial
for sensitive analyses where concentration of con_gurationoftheLCisrequired.
analytes may be quite small. SPE is achieved
through manual processing. Variations are
Bulk Sorbent. This is available in
available where either positive or negative
di^erent sorbents for those either
(vacuum) pressure can be applied to aid in
interested in packing their own cartridges
extraction. Cartridges are primarily made of
or performing dispersive solid phase
polypropylene, however, other mediums are
extractiondSPE)
( orQuEChERsmethod.

19
Table 8.SPECon_gurationGuideforCartridgeSelection

BedWeight Tube Volume MinimumElutionVolume Bed Capacity*


30–100 mg 1 mL 100–200 mL 1.5–5 mg
500 mg 3 mL 1–3 mL 25 mg
0.5–1 g 6 mL 2–6 mL 25–50 mg
2g 12 mL 10–20 mL 0.10 g
5g 20 mL 20–40 mL 0.25 g
10 g 60 mL 40–100 mL 0.50 g

*B
 ed capacity depends on the type of sorbent and analytes being used. Five percent of the bed weight is a general rule and represents the values
in the table.

Restricted Access Materials Dispersive Solid Phase Extraction (dSPE)


This method is another version of on-line sample prepa- Dispersive solid phase extraction is also to referred to as
ration. These are biocompatible and allow for the direct theQuEChERS0
/ quick,
( easy,cheap,e^ective,rugged,
injection of untreated biological samples (haemolyzed and safe) method. In this method, samples are _rst
blood, plasma, serum, milk, saliva, fermentation broth, extracted with aqueous miscible solvents (e.g. acetonitrile)
…) into a chromatography system. They were designed usually in the presence of high amounts of salts and/
as a time-saving, integrated solution. The working orbu^eringagents.UnliketraditionalSPE,thesorbent
principle is based on fractionation of a sample based on is present as a loose powder in pre-weighed quantities.
the molecular weight of both protein fraction and target The presence of salts aid in inducing liquid phase separ-
analytes. Macromolecules and protein matrix compounds ation. Upon shaking the sample with the sorbent, the
only interact with the exterior, hydrophilic surface of mixture is centrifuged, and the organic supernatant is
sorbent(diolmodi_cation)andarenotadsorbed.In removed. Depending on the nature of the sample, the
contrast, small and low molecular weight analytes are supernatant can be either analyzed directly or processed
enriched on the interior hydrophobic surface (C18, C8, further. Acceptance of this methodology is recognized in
or C4) of the column packing material. These materials the area of pesticide analysis in the formalized methods
operate on a dual method of size exclusion and reverse- EN15662:2008 and AOAC 2007.01.
phase/ion-pair chromatography. The access to the
interior is restricted dependent upon the pore diameter Sample Pretreatment
(physicalbarrier)orchemical(di^usion)barrier(e.g.
In this section, we will discuss pretreatment of some
semipermeable surface, protein network formation).
common matrices. Please understand, these conditions
The silica gel particles are 25 mm in size with pore
may require additional optimization depending on the
sizes of 6 nm (60 angstroms) and have a working pH
application or the analyte(s) to be monitored.
stability between 2 and 7.5.

Figure 23.DispersiveSolidPhaseExtractiondSPE) ( Formats.ShownontheleftaretraditionalQuecherstubeswhosetypicalmethodisoutlinedabove.


On the right, a newer format shown is dispersive SPE tips that is comprised of loose sorbent within a micropipette tip, allowing the extraction to be
performed entirely within the tip in a method that entails simple pipetting steps.

20
Most often analytes may be protein bound and these interactions will decreased recoveries. There are several methods
to disrupt these including:
1. S
 hift the pH of the sample to an extreme (pH <3 or >9). By shifting the pH, this can cause proteins to precipitate and/or
change the charge of the protein and/or analyte. If a precipitation occurs, continue working with the supernatant.
Serum,Plasma,and
WholeBlood 2. P
 recipitate the proteins. The most common way to precipitate proteins is to add three equivalents of a polar solvent,
usually acetonitrile. After mixing, the sample is centrifuge to remove the protein pellet and the supernatant is used
for further analysis.
3. Sonication.Thisisachievedbysonicatingthebiological‘uidfor15minutes,followedbydilutingwitheitherwateror
bu^erand_nallycentrifugation.
Thismaynotrequirepretreatment,however,itiscommontodiluteatleast1:withwaterorbu^eradjustedtothe
appropriate pH. A strong acid (ex. 12 M HCl) or base (10 M KOH) is added and heated for 15–20 minutes when interested in
Urine
acid or base hydrolysis. After the solution is cooled it is diluted in an appropriate solution. Alternatively, enzyme hydrolysis
to free bound compounds may be used.
Itiscommonpracticetodilutethemediawithwaterorbu^erattheappropriatepH.Ifthesample
ticulate-
ispar laden,
Cell Culture Media
itwilleitherneedtobe_lteredorvortexedandcentrifugedpriortoextraction.
Milk is often diluted with water or a mixture of water and a polar solvent (such as methanol). Proteins can be precipitated
Milk
by treating with an acid. If precipation occurs, the sample will require centrifugation.
Usually extraction can occur without prior treatment. One exception if it is laden with solid particulates and it clogs the
WaterSamples cartridge.Inthiscase_ltrationmayberequired.Note,that_ltrationmayreducerecoveriesiftheanalytesareboun
the particulates.
These may be able to be extracted without pretreatment usually. One consideration is if the beverage contains alcohol. If RP
Wine,Beerand
is being used, dilution of the sample so the alcohol percent is below 10% may be required. If particulates are present,
AqueousBeverages
_lteringorcentrifugationmayberequired.
Usually processed without pretreatment. An exception is if the juice has high amounts of pulp, which will need centrifuged
FruitJuices or_ltered.Asecondexceptionisifthesampleisviscous,inwhichcasethesampleshouldbediluted.Finally,fruitjuices
are acidic and may need to have the pH adjusted depending upon the analyte(s) of interest.
Since these are typically aqueous solutions, they are usually processed by RP or IE. If the sample is viscous, the sample
Liquid Pharmaceutical
will need to be diluted. Organic extracts of the preparation may be processed by NP.
Hydrocarbons or fatty oils are generally processed by NP. They cannot be diluted with water (immiscible) and if dilution
Oils
is required, a mid- to non-polar solvent is used.
These matrices are typically extracted with mid- to non-polar solvents via a Soxhlet extraction or sonication. Resulting
Soil and Sediment extracts are normally processed by NP to remove interferences. At this time, the sample may be dried and resuspended
in an appropriate solvent/solution for choice of SPE methodology.
Planttissue,fruit, For analytes that can be extracted by RP or IE, the material is homogenized in water and/or a polar organic solvent.
vegetables, Aftercentrifugationor_ltration,thepHofthesamplemaybeadjusted.Iftheanalyte(s)arenotasmisciblewithpolar
andgrains solvents, the sample can be extracted by NP by homogenizing in a less polar solvent.
Meat,Fish,and These can be processed similar to the plant, fruit, and vegetables. In addition, hydrolysis or digestion of meat or tissue
AnimalTissue can be achieved with strong acids (HCl), bases (NaOH, KOH), or enzymatic.
TabletsandOther Thesesolidsshouldbecrushedintoa_nepowderandextractedwiththeappropriatesolvent/solutiondependingu
Solid Pharmaceutical the SPE method being used.
Preparations

21
BioSPME Selection of initial conditions
BioSPME, or bioanalytical solid phase microextraction
devices are comprised of particles embedded in an inert Mode of separation
binder and immobilized onto a durable base, such as a
When selecting the most suitable mode of separation, it
polypropylene 96-pin device.
is dependent on sample solubility and how the analytes
The BioSPME extraction does not require pretreatment ofinterestdi^erfromothercompoundsormatrixinthe
of the sample. The coated portion of the BioSPME pin sample.Thetypeofsurfacemodi_cationusedin‘uences
is preconditioned and quickly rinsed with water prior to the selectivity of a stationary phase. There are several
immersion directly into the sample for extraction. While di^erentmodesofseparationtobedistinguished:
immersed, the binder selectively allows small analytes
of interest to bind to the adsorbent particles, while larger Reversed phase
macromolecules are excluded. The adsorption mechanism In reversed phase mode, the mobile phase is polar and
for BioSPME is based on partitioning of analytes between the stationary phase is less polar. The major distinction
the solution (sample) and the pin coating. The rate of this between analytes is their hydrophobicity where samples
partitioningisdependentupontheaanityoftheanalyte should be soluble in water or a polar organic solvent.
for the phase coating compared to the aanity for the
sample matrix. BioSPME is not an exhaustive technique, The retention mechanism is based on partition of the
rather, after a given amount of time, equilibrium is analytes between quasi-liquid stationary phase layer
achieved between the concentration of analytes in the andmobilephase.Asamodi_cationofthepacking
sample matrix and the pin coating. material for example n-octadecyl (RP-18), n-octyl (RP-8),
n-butyl (RP-4), or phenyl are applied—a combination
This allows for a robust and selective non-exhaustive
with or without an endcapping step is possible. Amino-,
extraction of free analyte that can be employed in both
cyano-anddiol-modi_ed,Penta‘uorophenylPFP ( orF5)
qualitative and quantitative applications. The 96-pin
and RP-Amide stationary phases can also be operated
con_gurationallowsfordirectsamplingfromwell 69 plates
under reversed phase conditions. Furthermore, a so-called
and is compatible with robotic liquid handling systems
providing a fully automated high-throughput methodology. PAH material is available, designed for the selective
separation of polycyclic aromatic hydrocarbons.

Macromolecules Reversed phase mode is mainly applied for the separation


Proteins bound to small analytes of non-polar to medium polar analytes (highly polar
Small analytes of interest and ionic compounds should be combined with ion-pair
reagents), hydrocarbons, alcohols, phenols, amines,
BioSPME Pin
carboxylic acids and derivatives with hydrophobic
molecule parts as well as compounds with hetero-atoms.
Analyte Adsorption

Supporting Core
Typical solvents utilized in reversed phase chromatography
are organic solvents miscible with mixtures of water
Functionalized Particles and aqueous bu^er solutions. In order of decreasing
Embedded in an Inert Binder
miscibility, these are: methanol, acetonitrile, ethanol,
Sample (whole blood, plasma, isopropanol, dimethyl formamide, n-propanol, dioxane and
serum, urine, saliva, etc.)
tetrahydrofuran. In general, all stationary phases can be
Sample Well utilized under highly aqueous conditions. Depending on
thewatercontentinthemobilephase,andthespeci_c
characteristics of the stationary phase (surface coverage),
phase dewetting and a subsequent drop of capacity can
occurafterprolonged‘ushingwithpurewater.Inorder
Extraction Time to avoid such a phenomenon, it is recommended to keep
Figure 24. Depiction of BioSPME Extraction showing analyte adsorption 5% organic solvent in the mobile phase. In case of column
vs extraction time. Small analytes are extracted as macromolecules and
protein bound analytes are excluded from extraction by the coated pin.
dewetting, indicated by a strong decrease of capacity
factors,phasepropertiescanberecoveredby‘ushing
with an organic solvent.
Phenyl stationary phases can be used for the
separation of aromatic compounds. Analyte retention
isperformedvia several di^erent mechanisms,
including-inter-actionsandpartitioningbetween
the mobile phase and the hydrophobic aryl-alkyl phase.
Phenyl stationary phases are in most cases combined
with methanol as an or-ganic solvent in order to
promote aforementioned - interactionswithanalyte
molecules.

Figure 25. Supel™ BioSPME 96-Pin Devices. Available to extract from


96-well plates, these are designed to perform multiple extractions at
the same time using a series of well-plates, whether through manual
or automated process.

22
Normal phase (polar) and medium polar phase Chiral separations
In normal phase, the mobile phase is non-polar while In chiral separations, the chromatographic result is
the stationary phase is more polar. This is the same for determined by the three dimensional structure of a solute.
hydrophilic interaction liquid chromatography. In normal Chiral selectors bonded to a base particle display a broad
phase, the major distinction between analytes is NOT their enantioselectivity and can be used for the chiral separation
hydrophobicity, and where the samples should be soluble ofenantiomersofnumerousdi^erentsubstanceclasses
in a hydrophobic solvent like hexane and the mobile phase (hydrocarbons, steroids, phenol esters and derivatives,
is a weak to moderate solvent for the sample. aromatic amines, heterocycles with 5 to 7-membered
rings) using typical RP or NP eluents (depending on the
An interaction of the active sites of the polar stationary
type of selector).
phase surface with polar parts of the sample molecules
leads to retention in normal phase chromatography. Onewidespreadchiralselectoriscyclodextrin
- covalently
linked to a base particle. Cyclodextrins are cyclic oligosac-
Polarsubstancessuchasunmodi_edSiO 2 (ideally suited charidesconsistingofglycosidically
-4,1- linkedD-glucose
for the separation of fat-soluble vitamins, small organic
units.-cyclodextrinconsistsof7glucoseunits,respec-
molecules or pharmaceuticals) or Al2O3 act as a stationary
tively. Geometrically seen, cyclodextrins can be described
phase.Inaddition,amodi_cationyieldingmediumpolar
as truncated cones, where all secondary hydroxyl groups
phases is possible:
are directed towards the larger opening, whereas primary
• NH2: The (silica) surface is derivatized with an hydroxyl groups form the smaller opening at the other
aminopropyl silane. The resulting phase can be end. The result is a hydrophobic inner cavity, in contrast
applied in the separation of carbohydrates or with the two hydrophilic openings. Since cyclodextrins
oligosaccharides in normal or reversed phase mode. are made up of chiral D-glucose units, the structure
may be regarded as a chiral selector. The enantiomers
• CN: Derivatization with polar cyano entities. For of a racemic substance mixture, due to their opposite
the separation of charged, unpolar to semipolar con_gurations, can now be associated(to di^erent
substances: Tetracycline antibiotics, steroids, organic degrees—with the cyclodextrin molecule. Consequently,
acids, peptides, proteins. diastereomeric “inclusion complexes” are formed based
• Diol:Diolmodi_edsurfacewithspeci_cselectivityfor on hydrophobic interaction (between cavity and guest
compounds with double bonds (azo dyes), peptides, molecule) and stereoselective hydrogen bonds (between
proteins and malto-oligosaccharides. the C2 and C3 hydrogen groups of glucose molecules
and the guest molecule).

Medium polar phases (“polar bonded phases”) can Other available selectors are (macrocyclic) glycoproteins,
generally be utilized both in normal phase and reversed (3,5-dimethylphenylcarbamate)-derivatized cellulose,
phase mode (HILIC) when the eluent properties copper ligands, polycyclic amine polymers or cyclofructans.
are adjusted accordingly. They possess both polar
properties from their functional groups and non-polar Glycoprotein Chiral Phases (CHIROBIOTIC®)
(hydrophobic) properties by -(CH2)n-spacers. A cyano CHIROBIOTIC® phases are based on covalently bonding
modi_edpackingmaterialcanactasanormalphase macrocyclic glycoproteins to a high purity 5 micron
system when combined with an non-polar mobile silica gel in such a way as to establish it’s stability while
phase,whiletheadditionofwater(orbu^er)tothe retaining essential components for chiral recognition.
eluent increases its polarity and allows for separations in CHIROBIOTIC® V and V2 are based on bonding
reversed phase mode. The main application for the diol Vancomycin, which contains 18 chiral centers surrounding
phase is normal phase chromatography, and reversed three pockets or cavities. Five aromatic ring structures
phase chromatography for the amino bonded phase. bridge these strategic cavities. Hydrogen donor acceptor
In normal phase chromatography, the polar bonded sites are readily available close to the ring structures.
phases have the advantage of a shorter conditioning CHIROBIOTIC® V has demonstrated selectivity similar
time than silica phases. to glycoprotein phases except it is stable from 0–100%
Typical analytes in normal phase chromatography are organicmodi_erandexhibitshighsamplecapacity.
non-ionic unpolar to medium-polar compounds such For CHIROBIOTIC® V2, changes to the linkage chemistry
as hydrocarbons, ethers, esters, alcohols, amines or and silica o^er improvements for preparative LC and
carboxylic acids and derivatives. for more demanding chiral separations. CHIROBIOTIC®
Normal phase chromatography works best with isopro- T, T2, and TAG are based on bonding the amphoteric
panol, ethyl acetate, tetrahydrofuran, t-butylmethyl glycopeptide, Teicoplanin, which contains 23 chiral centers
ether, dichloromethane or hexane. These solvents need surrounding four pockets or cavities. For CHIROBIOTIC®
to be mixed according to the characteristics of both T2, changes to the linkage chemistry and silica o^er
analytes and stationary phase characteristics in order to improvements for preparative LC and for more demanding
provide a suitable elution strength. chiral separations. CHIROBIOTIC® TAG has the sugars
removed from the macrocyclic glycopeptide to produce
Depending on the mode in which they are used, an aglycone structure as a variant of CHIROBIOTIC® T.
medium polar phases can be combined with any of the CHIROBIOTIC® R is based on bonding Ristocetin A to high
aforementioned reversed and normal phase eluents purity 5 micron silica.
and solvents.

23
Cellulose DMP Chiral Columns Protein-Based Chiral Columns
Cellulose DMP is a chiral stationary phase (CSP) Hermansson described the use of natural proteins
comprising spherical, high-purity porous silica coated immobilized onto a silica support for chiral separations
with DMPC (3,5-dimethylphenyl carbamate)-derivatized in 1983. Proteins contain a large number of chiral
cellulose, and packed in analytical to preparative size centersofonecon_guration,andmanyothersites
HPLC columns. It separates a wide range of chiral that contribute to the general retention process. We
compounds under normal phase, polar organic, and o^erthreeCSPswithproteinsasthechiralselectors,
SFCconditions,withhigheaciency,highloading CHIRALPAK AGP (a1-acid glycoprotein), CHIRALPAK
capacity, and excellent column lifetime. Performance is CBH (cellobiohydrolase) and CHIRALPAK HSA (human
comparable to other DMPC-derivatized cellulose CSPs. serum albumin). They are typically used in reversed-
phase mode, and perform a wide variety of chiral
Key Features and Application Areas:
separations. CHIRALPAK HSA is also used for drug-
• Classic DMPC-cellulose chiral selectivity binding studies. Solutes are retained by three types of
interactions: ionic (for charged solutes), hydrophobic
• Eacient,rugged,reproducible,andscalable and hydrogen bonding. The relative contribution of
• Low backpressure thedi^erentforcestosoluteretentiondependsonth
nature of the analyte.
• Ideal for chiral analysis in the pharmaceutical
industry and for small analytes in chemical and
environmental areas Choose the right HPLC column
• Routine chiral column method development Chromatographicresolutionismainlya^ectedbythe
screening protocols selectivity ( ), as can be seen inFigure 26 in the
stationary phase selection section of this guide.
• Approximately one-half the cost of most DMPC- Changing the mobile phase composition or the stationary
cellulose columns phase is the most powerful way of optimizing selectivity,
whereas the particle size, pore size, length of the column,
temperature, mobile phase strength have much less
Copper Ligand Exchange (CLC) Chiral Columns e^ect.Therefore,ifsatisfactoryresultsarenotmet,or
The CLC phases are based on coupling an enantiomeric no retention is achieved, it is better to change to another
form of an amine to a proprietary derivative to create selectivityusingadi^erentcolumntypeand/oradi^erent
an appropriate distance for copper coupling. Using the mobile phase.
copper ligand concept, this phase resolves hydroxy
acids like lactic, malic, tartaric and mandelic. This The column backbone
phase can also resolve amino acids and other amines The column backbone of a chromatography column
by the same mechanism. It has been reported that, in can consist of either a particle packing, or a monolithic
addition to amino acids, other bifunctional racemates structure. Independent from the chemical characteristics
like amino alcohols can be resolved. In theory, any of the silica or polymeric backbone, the chromatographic
analyte that can complete the coordination with the propertiesofacolumnarestronglyin‘uencedbythe
copper ion can be resolved. For the CLC-D column,the resulting particle packed or monolithic structure.
L enantiomer generally elutes before D with the
exceptionoftartaricacidwheretheDelutes_rst.The Monolithic HPLC columns
CLC-L column has the opposite elution order and the D
enantiomer elutes before L. Monolithic HPLC columns consist of a single piece of
high-purity and metal free monolithic silica gel (see
scanning electron microscopic image in Figure 26).
The mesopores within the silica skeleton form the

Figure 26. Scanning electron microscopic image of the cross section of a monolithic silica column. The total porosity of the monolith is > 90%.
Left image: Mesoporous silica skeleton, right image: Macroporous transport pore.

24
_neporousstructureandcreatethelargeuniform and make the use of dedicated HPLC systems necessary.
surface area on which adsorption takes place, thereby This needs to be taken into account when considering
enabling high performance chromatographic separation. theapplicationofm2 orsub-m2 particles.
Macroporous transport pores enable rapid ‘ow of
Note that UHPLC equipment and columns are not
the mobile phase and low back pressure analyses.
necessarily required in order to perform ultra-high
The diameter of both types of pores, as well as the
performance separations—it is more a matter of time
diameteroftheskeletoncanbeindependently_ne-
requiredtoachieveaspeci_cseparationeaciency.As
tuned. For this reason, monolithic silica columns can
long as a column, such as a monolithic silica column,
be tailor-made for a large variety of applications such
allowsforhigh‘owratesatatolerablebackpressure,
as small molecule analysis, as well as separation of
such separations can also be performed on robust
large biomolecules (see summarized pore size data of
and economically priced standard HPLC equipment.
monolithic silica analytical HPLC columns in Table 9).
In addition, low-pressure gradients can typically be
Currently, silica monolithic and organic polymer run with up to four eluents without extra costs. In
monolithic columns are available, the latter can often UHPLC separations, and under high-pressure gradient
lacksuacientcapacityandmechanicalstability.Inthis conditions, additional pumps would be necessary to
respect, silica monoliths are advantageous, as they do perform the same experiment.
not swell or shrink in organic solvents. They show much
If the plan is to speed up the separation processes and
higher capacities due to their high surface area and do
accelerateanalysesingeneral,thenincreasingthe‘ow
not exhibit micropores that can lead to peak tailing.
rate of an existing chromatographic method utilizing a
In contrast, a strongly basic eluent pH can be an issue
particle packed column can quickly exceed both column
when working with silica based monoliths. Make sure
and system pressure limits. Monolithic (silica) columns can
that mobile phase pH matches the characteristics of
be an option to overcome those limitations of particulate
your column backbone.
columns and to make the method quicker, as their back
pressure is generally lower. Figure 27 shows an example
Table 9. Pore size data of selected monolithic silica analytical HPLC columns. of speeding up a method using a monolithic column.
Diameter Macropore Mesopore
Product (mm) Modi_cation size(µm) size(Å)
Chromolith® 0.05 RP-18e 2 130
CapRod® 0.2
1 mL/min, 39 bar

Chromolith® 0.1 RP-8e 2 130


CapRod® RP-18e
2 mL/min, 76 bar

Chromolith® 0.1 RP-18e 1 130


CapRod® 0.2
HighResolution 3 mL/min, 115 bar

Chromolith® 2 RP-18e 1.5 130


4 mL/min, 152 bar
Chromolith® 3 RP-8e 2 130
4.6 RP-18e
5 mL/min, 193 bar
Chromolith® 4.6 Phenyl 2 130 0 2 4 6 8
CN Retention time (min)
DIOL
Si Figure 27. Higher throughput on a monolithic silica HPLC column.
NH2 Chromatographic conditions: Chromolith® HighResolution RP-18
endcapped 10 cm x 4.6 mm I.D.; mobile phase: acetonitrile/water
Chromolith® 2 RP-18 1.15 150
40/6(v/v);‘owrate15mL/min;detection254nm;temperature:
HighResolution 4.6 RP-8e
ambient; injection volume 5 µL, sample: 1 thiourea, 2 biphenyl-4-4’-ol,
RP-18e
3 biphenyl-2,2’-ol, 4 biphenyl-4-ol, 5 biphenyl-2-ol.
Protein A
Epoxy
Chromolith® 10 Si 2 130 Comparedtoam5 particulatecolumn,youcanexpect
25
an increase in analysis speed by a factor of four, while
Chromolith® 10 RP-18e 2 130 theseparationeaciencywillbeonthesamelevelasthe
25
theoreticaleaciencyofm5 . 4 particulatecolumnsdata (
Chromolith WP 2 RP-18 2 µm 300 Å for Chromolith®Performance;thetheoreticaleaciencyof
4.6 RP-4
Protein A
Chromolith® HighResolution columns is equivalent to the
Epoxy theoreticaleaciencyofm4.2 particulatecolumns)In .
addition,youcanrun‘owratesofuptoapproximately
9 mL/min under suitable conditions (mobile phase
TheuseofHPLCcolumnscontainingor 2 m3 small composition, column dimension) without reaching HPLC
silica particles often results in high back pressure. This system pressure limits. Alternatively, coupling of multiple
high back pressure can damage both the column and columnscanbeameansofachievinghigheacienciesat
HPLC system; therefore, traditional HPLC columns have a normal pressures. In addition, a low column back pressure
limited length and a limited number of theoretical plates. puts you into the position to replace acetonitrile by more
Attempts have been made to increase the plate count viscous solvents such as isopropanol in MS experiments—
by decreasing the particle size (see also chapter below). an option that can help to improve the sensitivity of a
Resulting UHPLC columns display high back pressures separation.

25
As can be seen from van Deemter plots of monolithic O H
H 3C N
O CH3 3 O NH2 total number
H 3C N N N
silica columns in Figure 28,thein‘uenceofan of injections
O N N O N N
increased‘owratedoesnotsigni_cantlydecrease CH3
1 2
CH3
Cl
10059
separationeaciency.Thisfactmeansthatonecan
work‘exiblyandinawiderangeof‘owrateswith 9405

minimal loss of peak resolution. 8644

35 7836
Chromolith® HR—4.6 mm

MS Intensity
Chromolith® 100—4.6 mm
30 7022
Chromolith® 100—3 mm
Chromolith® 100—2 mm 6049
25
5186
H (µm)

20
4371

15 3533

10 2739

1760
5
902
0
0 1 2 3 4 8
Linear velocity (mm/s)
0.00 0.50 1.00 1.50 2.00 2.50
Figure 28. van Deemter plots of selected monolithic silica HPLC columns. Retntion time (min)

Figure 29. R&D small molecule analysis. Results of everyday standard


If gradients from highly aqueous to highly organic runs testing the performance of a monolithic silica column. The total
number of injections is the combined numbers of test and standard runs.
conditions are run, subsequent reequilibration will be
Chromatographic conditions: Chromolith® Performance RP-18
more time consuming as compared to working in a endcapped 10 cm x 3.0 mm I.D.; mobile phase: A: water + 0.05%
narrow eluent composition range. Monolithic columns formic acid, B: acetonitrile + 0.04% formic acid; gradient: 0 min 99%
will perform this step of the analysis faster than particle A,1.8min0%A,2.5min0%A;‘owrate0.4mL/min(MS;HPLC
‘ow2mL/min,post-columnsplit);injectionvolume1µL;detection
packed columns of similar dimensions. pos.API-MS,m/zrange850;sample:1theophylline,2ca^eine,3
2-amino-5-chlorobenzophenone (1 mMol each) dissolved in methanol/
When working in a high-throughput environment, the water 50:50 (v:v).
propertiesofamonolithiccolumnmightalsobebene_cial.
For in-house pharmaceutical R&D, small molecule analysis
(e.g., analysis of yield and/or byproduct formation) is For the analysis of small sample amounts and/or when
performed on a monolithic silica column. The performance increased sensitivity is needed, large bore analytical
of the setup needs to be checked for consistency by the columns are not suitable. Instead, (monolithic) capillary
injectionofastandardmixtureoftheophylline,ca^eine columns for nano-LC are recommended. They are
and 2-amino-5-chlorobenzophenone every workday availablewithinternaldiametersof50to20mand
morning. Figure 29 displays the results of several of di^erent bonded phases (RP-8e: C8, RP-18e: C18).
those randomly picked test runs (number of injections Column length (15 or 30 cm) should be chosen in
given as the combined numbers of standard and test runs). accordance with sample complexity and they are not
available as one but few columns should be connected
Analytical monolithic silica columns are available in a using coupling unit. Such columns can be operated
range of diameters from 0.05 to 25 mm. Depending on atratherhigh‘owrates(13L/mincomparedto
your speci_c needs, you might pick a small I.D. (e.g., 200–400 nL/min for conventional media on a particle
2 mm) for fast and solvent saving gradient runs in low packed10mI.D.LCcapillarycolumn).Capillary
dead volume HPLC systems combined with MS detection. columns can be combined with online (ESI, nano spray) or
Column length (ranging from 25 to 150 mm) then mainly obineMALDI)
( MSdetection.Majorapplicationareasare
depends on the complexity of a sample and the speed of the separation and analysis of biomolecules (peptides,
analysis needed. For an ultra-fast separation of simple proteins, etc.) and the trace analysis of contaminants
mixtures, a Chromolith® Flash column (25 mm length) in food and environmental samples.
should be chosen, whereas for the separation of more
complex mixtures columns of 100 or 150 mm length are For further details about the proper choice of column
moresuitable.Fordetailsonthespeci_cpropertiesofthe format, see subchapter below.
di^erentbondedstationaryphases,pleaseseesubchapter
above.

26
Particulate HPLC columns (see also Figure 31:p/ucurvesfordi^erentparticulate
Particle packed columns are well known and established in columns in comparison with a monolithic column).
laboratories worldwide. Particles used for column packing
can either be spherically or irregularly shaped with the
former being available as fully or super_cially porous
material. An inherent property of particle-packed columns
is the mutual dependence of particle diameter and back
pressure. In other words, a decrease of particle diameter
isdesirableinordertoincreasecolumneaciency,but
thereisalwaysatradeo^withbackpressure,especially
when it comes to high speed separations. Currently, a
lot of e^ort is put into the development of sub-2 m
particle packed columns to be used in combination with
special UHPLC equipment. Particle size reduction leads to
adecreaseddi^usionpathlengthaswellasaminimized
C term (mass transfer, see chapter 1). As a consequence,
the gain in plate count is substantial, while the increase Figure 31.p/ucurvesforPurospher=STAR(2m)RP-18endcapped
in back pressure is increasing by a factor of four when 5cmx2.1mmI.D.(top),Purospher=STAR(3m)RP-18endcapped
5 cm x 2.1 mm I.D (middle) and Chromolith® FastGradient RP-18
reducing the particle size by half (Figure 30). endcapped 5 cm x 2.0 mm I.D. (bottom) utilizing acetonitrile/water
60/40 (v/v) as a mobile phase and thiourea as a dead time marker.

30,000
1 Regardless of particle diameter, two types of spherical
25,000 N
20,000
dP particles are available: fully porous particles and
super_ciallyporousparticlesFPPs ( andSPPs,respectively).
Eaciency(N)

15,000
FPPs (or “fully porous particles”) are well known and
10,000 established in the chromatographic community, whereas
5,000 SPPs (“fused core particles”) are a relatively new devel-
0 opment. SPPs are prepared by depositing a mesoporous
0 5 10 15 20 shell onto a solid and nonporous core. The particle size
distribution of the core shell particles is narrow, and eddy
400
1 di^usion(Aterm)isreduced.Duetotheshortdi^usion
Pressure (bar)

350 P path, axial dispersion of solutes is reduced and peak


300 dP2 broadening is minimized (see Figure 32).
250
200 SPPs combine the advantages of high eaciency and
150 low back pressure. On the other hand, FPPs provide
100 higher capacities.
50
0.6 µm
0
0 5 10 15 20 0.6 µm

dp (µm)
0.5 µm

Doublingtheeaciencybyhalvingtheparticlesize 0.5 µm ~5 µm 3.3 µm Solid Core


results in a pressure increase by a factor of four. 2.7 µm 1.7 µm Solid Core
~5 µm 3.3 µm Solid Core

2.7 µm 1.7 µm Solid Core


Particle (µm) psi bar N
1.8 5889 406 27,500
2.5 3089 213 20,000 5 µm
2.7 µm
3 2118 146 16,500 2.7 µm 5 µm
5 769 53 10,000
10 189 13 5,000
15 87 6 3,750
20 44 3 2,500

Column length: 10 cm, 3 mm/s linear velocity

Figure 30.E^ectofParticleSizeonEaciencyandPressure Fused-Core Particles


Fused-Core Particles Traditional Porous
Traditional
Particles
Porous Particles

In contrast to particles, monolithic columns show a


very low column backpressure. They owe their rapid
separation speed to their unique bimodal pore structure
of macro and mesopores. The macropores reduce Fused-Core Particles Traditional Porous Particles

columnbackpressureandallowtheuseoffaster‘ow Figure 32. StructureofSPPs(top)anditsin‘uenceonpeakshape:


rates, thereby considerably reducing analysis time. Minimizedpeakbroadeningbyshortdi^usionpath(reducedaxial
dispersion of solutes, middle) and narrow particle size distribution
(reducededdydi^usion,bottom).

27
Super_cially porous particles (SPP) provide smaller,Currently, particle packed columns are based on high-
reduced plate heights leading to higher eaciencies purity, metal-free silica produced from tetraalkoxysilane
narrower and taller peaks, for improved resolution and in a sol-gel process (type B silica). Due to the absence
lowerdetectionlimitsLODs ( andLOQs)( Figure 33).A‘at of metals in the silica structure, this column family can
van Deemter plot and higher linear velocity optimum allow be used for the analysis of acidic, basic, and chelating
higher‘owrateswithminimalcolumneaciencyloss. compounds. In contrast, older type A (or acidic) silica
contains metal ions and the resulting chromatograms
show tailing peaks for basic solutes (Figure 35).

O
O SiOH
O
O SiOH
O
O SiOH
O

Type A Type B

M2O – nSiO2 M= Si (OC2H5)4


Na, K, Fe
Figure 33.ComparisonofvanDeemterplotsfordi^erentparticlesize, [–Si (OH)2 – O – [–Si (OC2H5)2 – O –
FPP particles as compared to a 2.7 µm SPP particle Si (OH)2 – O –]n Si (OC2H5)2 – O –]n

–Si – O – Si – O – –Si – O – Si – O –
O O O O
SPP HPLC and UHPLC columns provide about 40% –Si – O – Si – O –
n
–Si – O – Si – O –
n

moreeaciencyincomparisontocolumnswithfullyporous
particles of the same size. This performance enhancement
SiO2(Na, K, Fe) SiO2
is applicable to all HPLC instruments (in addition to UHPLC
• Metal content • NO Metal content
systems). Due to the lower backpressure of 2.7 µm
particles in comparison to sub-2 µm particles, an increased • Peak-Tailing
- for • Peak-Tailing FREE
basic and chelating separation of basic
‘owratedouble
( inthiscase)canbeappliedprovidingthe compounds and chelating
samebackpressure,separationeaciencyandresolution compounds
asonasub-m2 UHPLCcolumn,justwithashorter
%05
Figure 35. Comparison of Type A and Type B silica
runtime, increasing sample throughput (see Figure 34).
1.Estradiol
2. ß-Estradiol Some stationary phases are prepared utilizing a cross-
Ascentis® Express C18 3. Impurity
linkedsurfacemodi_cationprocess,thereforethese
4. Estrone
0.4mL/min‘owrate
1
2
5.Estronedegradant
columns can be applied under comparably extreme
4 pH conditions, e.g. for the separation of strongly basic
3000 psi compounds utilizing alkaline eluents.
Chromatography columns based on (spherical) alumina
particles are stable in the range of pH 2–12. They are an
5 Twice the excellent alternative if separations need to be performed
3
speed at equal
pressures in the strongly basic pH range with eluents such as
1.0 2.0 3.0 solutions of NaOH for suppression of analyte ionization.
Min
Sub-2 µm competitor 2
0.2mL/min‘owrate Stationary phase selection
1 3000 psi

2
After setting the method goals and careful investigation
4 of the analyte structures (hydrophobicity/hydrophilicity;
functional groups and potential detection possibilities),
select a few appropriate bonded phases along with a
viable detector. The initial column selection is important
3
5 andthechromatographerisadvisednottousethe_rst
reversed phase HPLC column available. Reversed phase
0 2
Min
4 6 liquid chromatography is indeed a workhorse in most
laboratories, and a particulate RP-18 column is often
Chromatographicconditions the_rstchoiceformanychromatographers,butmany
Columns Ascentis® Express C18, 10 cm x 2.1 mm I.D., methods are often developed without utilizing the best
2.7 µm particles (53823-U) and sub-2 µm
particle colum (same dimensions)
or most appropriate selectivity. If the sample is mainly
Mobile phase water/acetonitrile 49:51 (for Ascentis® Express);
of hydrophobic character, having positive log P values
water/acetonitrile 55:45 (for sub-2 µm) and predominantly hydrophobic functional groups, a
Column temp ambient reversed phase column is advisable. Select a C18 or C8
Detector UV, 200 nm bonded phase for good retention and resolution. If the
Injection 1 µL

Figure 34. Comparison between SPP HPLC and UHPLC columns

28
sample molecules have aromatic backbones and C18 The column selectivity has the highest in‘uence on
or C8 columns are unable to resolve all components, resolution in chromatography. Therefore, the selection
then a change to a phenyl column where, in addition to of the best suitable column chemistry for the target
hydrophobicity,-interactionsbetweenthestationary analytes is an important selection parameter. C18 column
phaseandsamplemoleculesprovideadi^erentselectivity. chemistries are typically the _rst choice. Neverthele
As previously mentioned it is, however, important to when a C18 doesn't give the desired separation or the
usealcoholsasthemobilephaseorganicmodi_erwhile samplecontainscompoundsthatareknowntobediacult
working with phenyl columns. Acetonitrile, or any solvent to retain or resolve on a C18, consider other stationary
withdoubleortriplebondsbonds) -( inthebackbone, phase chemistries.
will diminish the interaction and make the phenyl column
If the method is intended for bioanalysis, analysis of
interact only with hydrophobicity.
matrix-rich samples in general, or where proper sample
preparation is unwanted or not possible, a monolithic
Resolution is mainly controlled by selectivity reversed phase column (e.g. Chromolith®) is a supe-
Resolution, R, can be expressed in terms of three rior choice over a particulate column. For example,
parameters(k,,andN)whicharedirectlyrelated Chromolith® RP-18 endcapped is a better choice over
to experimental conditions. The parameters k and Purospher™ STAR RP-18e or any other particulate
,aredeterminedbytheexperimentalconditions HPLC Column for such purposes as it has good matrix
(composition of the mobile phase; stationary phase tolerability and long column lifetime.
chemistry and temperature; see also chapter 1), and
whereNisa^ectedbycolumnlengthL,particlesize If samples are clean and/or good sample preparation
and pore size. Figure 36. willbeincludedinthe_nalmethod,andhighpeak
capacity is needed, a particulate column with small
particles and small pores may be more useful. Choose
[R5] columns which are known to have long lifetime at the
3
operating mobile phase pH. Choose bonded phases
α
2.5 based on high-purity, low-acidity silica for best peak
shape. If the sample consists of polar and hydrophilic
2 analytes, an orthogonal selectivity to reversed phase
shouldbeselected.Ifchiralresolutionisde_nedin
1.5 N
the method goal, then a suitable chiral column should
1 bechosen.Useanalytespeci_cstructureinformation
k (chemical structure, log P values etc.) to choose a proper
0.5 stationary phase (Figure 37 and Table 10). If acidic or
0
basic analytes are present in the sample; reversed phase
1 1.05 1.1 1.15 1.2 1.25 ion suppression (for weak acids or bases), reversed
0 5000 10000 15000 20000 25000
phase ion-pairing (for strong acids or bases) or HILIC
0 5 10 15 20 25
should be used. For low/medium polarity analytes,
Figure 36. Key parameters that control resolution and their overall normal phase HPLC or HILIC are viable techniques.
contribution to changes in resolution

Hydrophobic
Hydrophilic

Log P

8 0 -8
Too much Poor Peak
Aromatic
Isomers retention Shape (Basic Retention too short or inadequate Separation on RP-18
Compounds) compounds
on C18
Polar compounds
Closely related Pi-Pi
with high water HILIC
compounds Interactions content

Reversed phase
HILIC

RP- F5 ZIC
C30 C18 C8 Amide (PFP) Phenyl AQ C18 Amide Diol Cyano Amino OH 5 Si
USP
USP L62 USP L1 USP L7 USP L60 USP L43 USP L11 USP L1 USP L68 USP L20 USP L10 USP L8 USP L95 USP L3 L114

Figure 37.

29
Table 10. Polarity scale of analyte functional groups.

Polarity Functional Group Hybridization IntermolecularForces


Low Methylene s London
Phenyl s/p London
Halide s London, dipole-dipole
Ether s London, dipole-dipole, H bonding
Nitro s/p London, dipole-dipole, H bonding
Ester s/p London, dipole-dipole, H bonding
Aldehyde s/p London, dipole-dipole, H bonding
Ketone s/p London, dipole-dipole, H bonding
Amino s/p London, dipole-dipole, H bonding, Acid-base chemistry
Hydroxyl s London, dipole-dipole, H bonding
High Carboxylic acid s/p London, dipole-dipole, H bonding, Acid-base chemistry

Choosing the right column format Selection by column dimension


Use the column selection guide below (Table 11) to Depending on the scale of a separation and/or a needed
_ndthebestcolumncon_gurationforminimumanalysis separationeaciencyofaseparation,acolumndimension
timewithhigheaciencyandresolution,andmatchup speci_edbycolumninnerdiameter(i.d.)andcolumn
method goals to make sure that the chosen format has length has to be chosen.
the ability to produce resolution for the purpose of the
application, e.g. choose the right column length, column
inner diameter, particle size and pore size.

Table 11.

Columndimension
(lengthxi.d.inmm) Application Reason
4x4 Guard-column Protection from mechanical contamination
x5 2/34.6 Sample contaminated to low extent
x10 4.6/1025
25x4 Precolumn High capacity precolumn
30x2/.134 Method development Short retention time
x5 2/.134 Rapid HPLC and UHPLC Rapid equilibration
x75 4 (if pressure stable) Low solvent consumption (small i.d.)
Low pressure drop
10x2.1 High detection sensitivity Semi-micro column for low injection volumes and low peak dispersion
x125 2/3 (mass selectivity) Low solvent consumption
x150 2.1/3
10x4.6 Standard column Adequate performance for most applications (average performance
x125 4/.6 8000–10000 N/columns)
x150 4.6
250x2/.13 High detection sensitivity Semi-micro column for low injection volumes and low peak dispersion
High performance separation Low solvent consumption
For complex samples
250x4/.6 High performance separation For very complex samples
250x10 Semi-preparative For mg quantities of pure substance on lab scale
250x25 Preparative For g quantities of pure substance

Guidelines for typical flow rate and orientation values for the loading capacities of analytical and semi-preparative columns

Columndimension
(lengthxi.d.inmm) Typical‘owratea Sample amount Sample volume
150x1 0.06 mL/min ~0.05 mg 0.05–1 µL
250x2 0.25 mL/min ~0.2 mg 0.2–5 µL
250x3 0.6 mL/min ~1 mg 1–20 µL
250x4 1 mL/min ~5 mg 5–80 µL
250x10 6 mL/min ~30 mg 30–500 µL
250x25 39 mL/min ~200 mg 200–3000 µL

30
If high mass loadability is needed, or if the sample is 50 50 50

complex, a larger column (in both length and diameter)


is recommended, as it can accommodate more mass. 40 40 40

If working with traditional detectors like UV, RI, FL


etc. then 4.6 or 3.0 mm I.D. columns are suitable. 30 30 30

Dependingonthespeci_cneeds,asmalli.d.forfast
and solvent-saving gradient runs in low dead volume 20 20 20
HPLC systems combined with MS detection may be
selected; for MS, a 2 mm or smaller I.D. column is 10 10 10
generallyrecommended.Ifsuacientresolutionis
achieved, it is also possible to speed up the separation 0 0 0
byincreasingthe‘owrateorshorteningthecolumn
2.0 3.0 2.0 3.0 2.0 3.0
length (e.g., a column of 25 mm length for an ultra- Particle size: 10 µm Particle size: 5 µm Particle size: 3 µm
fast separation). For the separation of more complex N = 5,700 N = 15,199 N = 23,411
Peak H = 23.35 Peak H = 36.38 Peak H = 45.26
mixtures, columns of 100 or 150 mm length are more Rs = 1.42 Rs = 2.32 Rs = 2.84
suitable. When it comes to the analysis of small sample P = 58 bar P = 115 bar P = 153 bar
volumes, and/or when increased sensitivity is needed,
Chromatographic conditions
large bore analytical columns are not suitable. Instead, Column: Ascentis® C18, 150 mm x 4.6 mm
capillarycolumnswithinternaldiametersofto 0 5 m0 2 Flow: 1.8 mL/min
Mobile Phase: 70/30 ACN/Water
for nano-LC are recommended. Column length should Detection: 254 nm
be chosen according to the sample complexity. Temp: 30 °C
Injection Vol: 5 mL
Analytes: 1. o-Xylene (0.04mg/mL)
Silica-based materials are physically strong and will not 2. p-Xylene (0.01mg/mL)
shrink or swell, being compatible with a broad range of
Figure 38.E^ectofParticleSizeonChromatographicEaciency
polar and non-polar solvents, and are therefore often the
initial choice. Most silica based columns are stable from
pH 2–7.5, and, historically, polymeric packing materials Pore size
provided better column stability under pH extremes.
Choose a pore large enough to enclose the target
A polymer-based packing material, like ZIC®-pHILIC,
molecule completely. If the molecule is larger than the
is compressible and may shrink or swell with certain
pore,sizeexclusione^ectswillbeseen,anditwillbe
solvents. Care must therefore be taken if a polymeric
diacultorimpossibletoretain.Forthisreason,reversed
column is used, and the upper back pressure limit is
phase wide-pore materials for the separation of peptides
lower than corresponding silica based stationary phases.
and low molecular weight proteins are available.
Newer, type B high-purity silica-based phases, like
In general, packing materials with a smaller pore size
Purospher™ STAR, are stable at pH 1.5–10.5, with
have higher surface areas and higher capacities than
the surface functional groups bound to the base silica
packing materials with larger pore sizes. A larger
particle at multiple attachment points via polymeric
surface area typically indicates a greater number of
modi_cation.
pores, and therefore a higher overall capacity. Smaller
surface areas equilibrate faster, which is important for
Particle size gradient elution analyses. Larger pores are better for
Smaller particle sizes provide higher separation interaction with large compounds, such as proteins.
eaciencyandhigherchromatographicresolutionthan
largerparticlesizes.However,largerparticlesizesCarbon o^er load
faster‘owratesatlowercolumnbackpressure,and
For silica-based, reversed-phase packing materials,
are less prone to clogging, and for these reasons are
carbon load indicates the amount of functional bonded
moretoleranttomatrixe^ects.Typicalparticlesizes
phase attached to the base material. Phases with lower
rangefrom310m,but2mparticlesizesarealso
carbon loads are more weakly hydrophobic, which may
availabletomaximizeresolutionA5mparticlesize
signi_cantlyreduceretentiontimesoverphaseswith
representsthebestcompromisebetweeneaciency
higher carbon loads. However, a higher carbon load
and back pressure for most non-high throughput
will give higher capacity and often greater resolution,
applications (Figure 38).
especially for compounds of similar hydrophobicity.
Fastandultra-fastseparationson2mand3m Carbon load is not a relevant parameter for columns
particulate silica have become particularly important used in normal phase or HILIC mode.
due to the need for high sample throughput and higher
productivity in daily lab work. With the use of UHPLC Endcapping
methods with short columns, narrow inner diameters,
Silica-based, reversed-phase packing materials
and small particles sizes, it is possible to speed up
have free silanol groups that will interact with polar
analyses up to ten-fold, while decreasing solvent
compounds. Endcapping the bonded phase minimizes
consumption by up to 90% and increasing sensitivity.
these secondary interactions. Choose endcapped
Clogging and high back pressure can be an issue,
phases if secondary interactions with polar compounds
therefore3mparticulatematerialisrecommendedfor
are not wanted. Choose non-endcapped phases if
use with complex samples.
enhanced polar selectivity is desired, for stronger
retention of polar organic compounds.

31
Mobile phase selection truebu^er,butratherapHadjustablesalt)areviable
alternatives depending on detection mode. At high
In previous sections, insight has been given to mobile pH, dipotassium hydrogen phosphate and ammonium
phase recommendations, solvent properties, and carbonatecanbeusedasbu^erstomaintainpHabove
bu^ercomponents.Asummaryofstartingconditions 8. Keep in mind that when working at high pH, only
is presented in this section, along with a discussion columns with wider pH tolerability should be used,
aboutthedi^erencebetweenisocraticandgradient and for this purpose Purospher™ STAR is an excellent
elution. The mobile phase solvent strength is a measure choice.Inorganicbu^ersarenotrecommendedwith
ofitsabilitytoeluteanalyteso^thecolumn.Solvent MS,ELSandCAdetectors.Thesebu^ersarenot
strength is generally controlled by the concentration volatile and may precipitate, and this is also the case
of the solvent with the highest strength; for example, when high percentages of organic solvent are used in
in reverse phase HPLC with aqueous mobile phases, the mobile phase (>70%).
thestrongsolventwouldbetheorganicmodi_erbut
in normal phase and HILIC, the strong solvent would Gradient elution
be the most polar. It is worth pointing out that cyano-
bonded phases are easier to work with than plain silica Often it is not possible to elute all analytes with a single
fornormalphaseseparations.Theaimisto_ndthe mobile phase (isocratic) in the desired k’ (2–5) range.
correct concentration of the strong solvent. With many It is therefore advisable to use gradient elution where
samples, there will be a range of solvent strengths the mobile phase strength, and sometimes also pH
that can be used within the aforesaid capacity limits. andionicstrength,willchangeovertime.E^ectively,
Otherfactors(suchaspH)mayalsoa^ecttheoverall this means that early in the gradient the mobile phase
retention of analytes. elution strength is low, and where the elution strength
isincreasingwithtimeaccordingtoade_nedprogram
that maximizes the number of peaks that can be
Isocratic elution resolved with a given resolution. This method results in
In partition chromatography, the mobile phase should the constant peak width observed in gradient elution,
be a moderate to weak solvent for the samples to compared to isocratic elution where the peak width
achieve peak focusing and not to compromise the increases in proportion to retention time. Gradient
actual separation. A good rule of thumb is to achieve elution is used to solve the general elution problem
a capacity factor (retention factor, k) of 2 to 5 for an for samples containing mixtures of analytes with a
isocratic method. In both RP and HILIC mode, the wide range of polarities. Gradient elution will also give
preferred organic solvent is acetonitrile for several greater sensitivity, particularly for analytes with longer
reasons: favorable UV transmittance, low viscosity, retention times, because of the more constant peak
and being easy to volatilize (important for MS, ELS width (for a given peak area, peak height is inversely
and corona discharge, CA, detectors). Methanol is a proportional to peak width). Common practice in
reasonable alternative, hence it may be worth changing methoddevelopmentistorunascoutinggradient_rst
the organic solvent if resolution is not achieved, to decide whether to use isocratic or gradient elution.
and adjust the percentage organic solvent in the
mobile phase to accomplish maximum resolution and Ifht/tGl0.25usegradientelutionIfht/tGk0.25use
retention. Methanol and other alcohols are also the isocratic elution
preferredchoiceasthemobilephaseorganicmodi_erWhere ht is di^erence in the retention time between
while working with phenyl columns. Acetonitrile or any the _rst peak and last peak in the chromatogram, tG
solvent with double or triple bonds in the backbone is the gradient time; the time over which the solvent
willdiminishthe-interactionandmakethephenyl composition is changed. For most samples (unless they
column interact only with hydrophobicity. are extremely complex), short columns (10–15 cm) are
In reversed phase mode, the initial mobile phase pH recommended to reduce method development time. Such
should be selected with two considerations. Low pH columnsa^ordshorterretentionandequilibrationtimes
that protonates column silanol groups and reduces A‘owrateofmL/
5 . 1 minshouldbeusedinitially.
their chromatographic activity is generally preferred, The direct disadvantages with gradient elution are
especially with non-endcapped columns. Mobile phases the need of a more complex HPLC system, and the
havingpH1to3with205mMbu^er(potassium column requires re-equilibration after every analysis,
dihydrogen phosphate, TFA or formic acid in water) which makes injection-to-injection lengthier than for
is advisable depending on detection mode, and to an isocratic method. Gradient mode is not compatible
increase temperature to reduce analysis time. with all detectors (i.e. RI and EC) and more variables
Mobile phase solvents should be water miscible, have need to be controlled to ensure method reproducibility.
lowviscosity,lowUVcut-o^,benon-reactive,andfor System dwell volume (gradient delay volume) becomes
these reasons, acetonitrile, methanol and THF are used important especially in scaling a separation or
with RP columns. Not all RP methods are suitable under whenever transferring a method between instruments
acidic conditions, and other pH intervals may provide and/or laboratories. Be aware that delay volumes will
di^erentselectivity.AroundneutralpH,dipotassium vary from instrument to instrument.
hydrogen phosphate or ammonium acetate (not a

32
Gradient method development When performing a separation under isocratic conditions,
Good laboratory practice does not allow gradients from a premixed mobile phase has to be utilized. Shifts in
100% aqueous to 100% organic. For method robustness retention times caused by irreproducible mixing of
(for better mixing, to prevent precipitation of salt, avoid a mobile phase by the HPLC pump unit are avoided.
column dewetting, and to provide more robust gradient Premixed eluents have to be prepared by separately
pro_les),itisadvisabletokeepminimum5%ofeach measuring the appropriate volumes of each solvent in
component in all mobile phase bottles. In practice for ordertoavoidanyvolumecontractione^ectsduring
a reversed phase method, this means mobile phase A mixing.
contains 5% organic solvent and 95% aqueous, while Under gradient conditions, the mixer of the pump unit
mobile phase B contains 95% organic solvent and of the HPLC system is responsible for a proper mixing
5% aqueous. of the mobile phase. Either static or dynamic mixing
Initially, run a wide scouting gradient (5-95 % B) chambers exist; make sure that the mixer is switched
over 40–60 minutes. From this run, decide whether on when working with a dynamic mixing chamber. The
isocratic or gradient elution is best for the application. If shift in retention time in a separation of alkyl phenones
gradient mode is a more appropriate alternative, eliminate withadynamicmixerswitchedonandswitchedo^can
sections of the gradient and try to compress the analyte be seen in Figure 39.
peaksinspaceasmuchaspossiblepriortothe_rstand 200
lastelutingpeak.Tofurtherimprovethegradientpro_le

UV Intensity (mV)
and shorten overall cycle times (including re-equilibration),
try to reduce the gradient and total run time. Keep in 150
mindthatasegmentedgradientcanbeane^ectivetool
to improve the separation. If there is a need to improve 100
the separation of two closely eluting peaks; change
the solvent strength by varying the fraction of each
solvent (gradient shape and steepness); change column 50
temperature; change the mobile phase pH (in small
units)use
; di^erentmobilephasesolventsand/orbu^er
0
components;and/oruseadi^erentselectivityby 0.0 0.5 1.0 1.5 2.0 2.5 3.0
changing the stationary phase.
Retention time (min)
Figure 39. Shift in retention time in a separation of alkyl phenones
withadynamicmixerswitchedonandswitchedo^(greenandyellow

Mobile phase preparation chromatogram, respectively).


Chromatographic conditions
Solvents and bu^ers have to be miscible over the Column: Purospher™ Star RP-18 endcapped (2 µm) Hibar® HR
cm
5 xmm
1.2 I.D.detection:
; UVnm)
742( temperature:
; °C;
04 ‘ow
entirerangeofachromatographicrunandnobu^er rate: 0.54 mL/min; mobile phase A: water, mobile Phase B: acetonitrile;
precipitation must occur. Degassing helps to prevent gradient conditions: 0 min 45% B, 2.5 min 95% B; sample: 1 Urea,
bubble formation and spikes in chromatographic runs; 2 Acetanilide, 3 Acetophenone, 4 Propiophenone, 5 Butyrophenone,
today, the pump module of many modern LC systems 6 Benzophenone, 7 Valerophenone, 8 Hexanophenone, 9 Heptanophenone.
o^ersonlinedegassing.
All eluent bottles have to be labeled properly and a shelf
lifehastobeadded.Foraqueousbu^ers,theshelflife Filtration tools for preparing HPLC/UHPLC
typically is not higher than one week. Exceeding this buffers and mobile phases
time span will, for example, cause microbial growth.
Mobilephasesolventsandbu^ershavetobeofhigh-
Microbial growth can be prevented by adding 5% or more
qualityHPLCgrade,andpreparedfreshly,_ltered
of organic solvent to the aqueous mobile phase. The
m)5 4 . 0 ( anddegassedbeforeuse.Membrane_ltration
shelf life of organic mobile phases is comparable to the
removes contaminating particles from solvents and
manufacturer’s data printed on the original packaging.
mobile phases, increasing column life, minimizing
Make sure that all mobile phase solvents and additives back pressure, and preventing system failure. That’s
used in a chromatographic separation are completely why most HPLC/UHPLC instrument manufacturers
miscible (solvent miscibilities) in order to avoid recommend_ltrationofmobilephasesusingeither0.45
precipitation of additives in the HPLC system and in or0.2m_lters.Membranesthatdisplaythehighest
order to perform reproducible analyses. Solvents being particleretentiontendtobethemoste^ectiveat
completely miscible with all other solvents listed in table x minimizing back pressure.
are: Ethanol, isopropanol, dioxane, tetrahydrofuran,
acetone and glacial acetic acid.

33
Polypropylene membranes exhibit poor particle 1.2E+07

retention,andtherefore_lteringUHPLCmobilephases
1.0E+07
throughpolypropyleneistheleaste^ectiveforreducing

UV Intensity (mV)
backpressurebuildup.Incontrast,_lteringtheeluent
8.0E+06
throughpolytetra‘uoroethylene(PTFE)orPVDF
membrane_lters(e.g.,Omnipore ® and Durapore®), 6.0E+06
enablestheUHPLCsystemtorunwithoutsigni_cant
back pressure buildup. 4.0E+06

Inadditiontothe_ltrationofsolventspriortomobile 2.0E+06

phase preparation, various manufacturers provide


_lterfritswhichcanbeattachedtotheeluenttubing0.0E+06 of 0.0 1 2 3 4 5 6 7
mobilephasebottles.These_ltersadditionallyprotect
the LC system from particulate matter. Stainless steel Retention time (min)
orPTFE_lterfritsshouldbeusedratherthanglass Figure 40.E^ectofequilibrationtimeontheaccumulationofplasticizers
frits:Cleaningofthelatteristimeconsuming,asbu^er on a reversed phase column prior to a gradient run. Equilibration
times with mobile phase A: 0 (blue chromatogram), 15 (red), and
residue is hard to remove. In addition, silica and alkali 60 (green) minutes.
aredissolvedfromtheglass_lterandformadductswith Chromatographic conditions: Mobile phase A: water/acetonitrile 95/5
analyte molecules when working with MS detection. (v/v) + 0.1% formic acid, B: acetonitrile + 0.1% formic acid; gradient:
0min10%A,3min5%A,5min5%A;‘owrate0.4mL/min;
detection pos. ESI-MS (TICs); temperature: 25 °C; sample: plasticizers
Solvent and additive purity (*) were added by the immersion of plastic tubing in aqueous eluent A.
The quality of the solvents as well as of the additives
(bu^ers, salts, acids, bases) utilized in an HPLC
PEEK generally shows chemical resistance to a wide
experimenthastobeadaptedtothespeci_csensitivity
range of organic solvents commonly used in RP and NP
of the detector and the type of elution protocol applied.
applications. Therefore, columns with PEEK hardware
In isocratic separations combined with UV detection,
can be used with the following organic solvents without
isocratic grade solvents will be the matter of choice. In
limitations: Alcohols, acetonitrile, alkanes, dioxane, esters,
contrast, gradient elution protocols and UV detection
and ethers. Restrictions are imposed with respect to the
require gradient grade solvent quality in order to avoid
following solvents (as swelling of PEEK may occur after
elution of solvent contaminations (“ghost peaks”) during
prolonged exposure): The mobile phase should not contain
chromatographic runs.
more than 50% tetrahydrofuran (THF), 5% chlorinated
Mass spectrometry as the most sensitive detection solvent (e.g. dichloromethane), or 5% dimethyl sulfoxide
techniquerequiresspeci_cMSgradequalitysolvents(DMSO). of However, all these solvents can be used as a
highest purity. These solvents maximize the sensitivity component of the sample solution.
of an analysis by minimizing signal suppression, adduct
Do not exceed the pH stability range of the column, this
formation, and baseline noise. In general, the purest
avoidsdamagetothecolumnmodi_cationorcolumnbed.
solvent and reagent quality available has to be used, and
For example, when using a silica based stationary phase,
their contamination has to be avoided by careful handling.
a pH in the range of 2–7.5 is recommended. Higher pH
Contamination in solvents and additives such as acids, values will dissolve the silica, creating voids in the column
basesorbu^erscanaccumulateonthestationaryphase( and leads to peak shape deterioration and peak fronting.
depending on the chromatographic conditions applied for Lower pH values can eventually hydrolyze the bonded
aspeci_cseparationandontheequilibrationtimeprior to
phase. These defects will cause a decrease of retention
a run. Figure 40 shows the accumulation of plasticizers times and loss of resolution and might lead to peak tailing
dissolved in aqueous eluent A on a reversed phase column forbasiccompounds.Across-linkedsurfacemodi_cation
after equilibration for 0, 15, and 60 minutes. While these or a polymeric backbone of a column can increase the pH
compounds would be eluted as very broad peaks under operating range (e.g., to 1.5–10). HPLC columns based
isocratic conditions (and cause an increased background on alumina can typically be run at a pH of 2–12. Carbon-
noise), they elute as distinct, intensive peaks under based HPLC columns (like Supel™ Carbon LC, based
gradient conditions and can interfere with analyte signals. on porous graphitic carbon technology) can operate at
To avoid such ghost peaks use pure solvents and additives pH14withnodeclineineaciencyorlifetime.
andavoidexcessivecolumnequilibrationcolumn ( ‘ushing
Do not use strong acids (e.g. hydrochloric, nitric, and
with approximately 10 column volumes is suacient,
sulfuric acids) in the column and limit the use of strong
alternatively one blank gradient run including subsequent
bases (e.g. sodium, potassium, ammonium hydroxide)
equilibration is an option).
to amounts needed to adjust the pH of the mobile phase.
Chemical compatibility When measuring the pH of mobile phases, the
The composition as well as the pH of the mobile phase has measurement should be carried out in the aqueous
to be chosen in accordance with column housing material, medium before mixing with organic solvents. Although
as well as stationary phase characteristics. Damage to this will not give the actual pH in the mixed aqueous-
thecolumnhardware,modi_cationorcolumnbedcanbe organic eluent, it will give more consistent results than
avoided by following this recommendation. a mixed mobile phase.

34
Mobile phase composition and temperature Mobile phase properties
Verify that solvents are miscible when changing mobile Chromatography on normal phase columns is based on
phases,andthatnobu^erprecipitationwilloccur. the interaction of polar functional groups of analytes
with the polar stationary phase surface. In addition, the
In reversed phase chromatography, water-miscible organic
physical properties of the solvents/eluents play a role,
solvents such as acetonitrile, methanol, isopropanol
as these compete with the analyte molecules during the
andwater,oranaqueousbu^erserveaseluents,and
retention process. Depending on the strength of dipole-
additives such as tetrahydrofuran or dioxane can be used.
dipole and hydrogen bonding interactions of solvents with
Bu^erssuchasphosphate,borate,acetate,carbonate,
a normal phase stationary phase, their elution strength
organic modi_ers and ion pair reagents present no
can be considered as weak or strong. An eluent showing
problems as long as the appropriate pH stability range
weak interaction with the stationary phase is only capable
of the stationary phase and packing material is not
of eluting weakly bonded analytes from the column,
exceeded.Ionpairreagentsareoftendiacultto‘ush
whereas a strong interaction causes elution of strongly
completely from the column. Therefore, columns used
bonded sample molecules. The elution or solvent strength
with these reagents should be dedicated to the particular
of various solvents depends on the type of stationary
analysis involved.
phase used. Table 12 summarizes the solvent strength
0 of various solvents commonly used in normal phase
Normal phase and medium polar phase columns are
generally used with unpolar to semipolar solvents or chromatography mode on silica gel. On alumina the
obtainedvaluesfor 0 are somewhat higher, but show the
mixtures of n-heptane, hexane, cyclohexane, dioxane,
ethyl acetate, methanol, chloroform or dichloromethane. sametrend.Inorderto_ne-tunetheelutionstrengthof
n-heptane and dioxane are typical solvents for adsorption amobilephase,di^erentsolventscanbemixed,either
chromatography. static in isocratic separations or dynamic in gradient runs.

Sometimes, it can be helpful to change the temperature


Table 12 Solvent strength of selected solvents in normal phase
of a chromatographic method, an approach that alters the
chromatography on silica gel.
selectivity of a given system of stationary phase/mobile
phase/analyte and decreases method run times. Mutual Solvent Solventstrength 0

interactions are numerous; therefore, an empirical process n-Pentane 0


isnecessaryforselectivityoptimizationvia_ne-tuning n-Hexane 0
oftemperature.Anadditionale^ectofatemperature
Isooctane 0.01
increase above room temperature is a desired increase in
separationperformancecausedbyaccelerateddi^usion Cyclohexane 0.03
processes. p-Xylene 0.20

When adjusting temperature, one has to keep in mind Diisopropyl ether 0.22
that the chemistry of most chromatographic columns is Toluene 0.22
based on silica, and one negative aspect of this chemistry Diethyl ether 0.29
in an aqueous environment is that raising temperature
Methylene chloride 0.30
dramaticallyincreasessilicasolubility.Duetothisspeci_c
temperature,limitsexistfortheoperationofdi^erent Methyl ethyl ketone
stationary phases (for details see respective columns Acetone 0.43
instructions). Keep in mind that exceeding these limits Dioxane 0.43
will cause a loss of capacity, column performance or even
Methyl acetate 0.46
a breakdown of the column bed. As a rule of thumb, the
operating temperature for silica based HPLC columns Tetrahydrofuran 0.48

should not exceed 60 °C. Depending on the surface tert-Butylmethyl ether 0.48
chemistry of each type of column this value can be lower Ethyl acetate 0.48
or higher. For detailed information, please always refer to Dimethyl sulfoxide 0.48
the column manual.
Diethyl amine
An appropriate method for increasing the lifetime of Nitromethane 0.49
analytical columns is the use of guard columns or guard
Acetonitrile 0.50
cartridges. This principle is based on the saturation of the
mobile phase with silica before entering the analytical Isopropanol 0.60
column and is especially recommended when working Ethanol 0.68
with amino bonded phases. In addition, guard columns Methanol 0.73
protect the analytical columns from solid matter and
prevent clogging. An alternative can be columns with a Ref: [table reproduced from V.R. Meyer, Praxis der Hochleistungs-
polymeric column bed; such columns can tolerate higher Flüssigchromatographie]
temperatures.
Next to the elution strength, the viscosity and UV ab-
For proper thermostatting of columns, not only at elevated sorbance of mobile phase solvents and additives play
temperature, please also see the speci_c sectionan in important role in terms of their suitability for use in
chapter 2. HPLC analyses.

35
The viscosity of an eluent is directly correlated to the 3.5 acetonitrile isopropanol
methanol THF
back pressure of a given HPLC column and system ethanol
3.0
combination. The ‘ow rate range under which a

Viscosity (cP)
separation can be performed—and in turn, the speed 2.5
of an analysis—is enlarged when using low-viscosity.
Another bene_t of low solvent viscosity is enhanced2.0
mass transfer also attributing to the speed of analysis. 1.5
Separation at increased temperature can balance the
negative e^ects of high mobile phase viscosity. The 1.0
viscosity of various aqueous mixtures of important .5
organic solvents is displayed in Figure 41. In general,
low viscosity eluents are favorable for the reasons given 0
0 20 40 60 80 100
above. However, in mass spectrometry detection, it can Percent organic (v/v)
be helpful to substitute 20–30% of a low viscous solvent
Figure 41. Viscosity of aqueous mixtures of acetonitrile, methanol,
such as acetonitrile by isopropanol in order to improve ethanol, isopropanol and tetrahydrofuran.
the sensitivity of an analysis. Under these prerequisites,
back pressure issues can be avoided by utilizing monolithic
type columns. 1.0
acetonitrile
methanol
The UV transmittance or absorbance is another key issue ethanol
0.8 isopropanol
when working with organic solvents. The transmittance

Viscosity (cP)
THF
is in‘uenced by the inherent properties of a solvent,
but also by any contamination which can signi_cantly 0.6
decrease sensitivity. While the absorbance is negligible in
thevisiblerangeoflight,thedi^erenceisratherlargein 0.4
the UV wavelength range from 190 to 300 nm (Figure 42).
Acetonitrile displays the lowest UV absorbance and can 0.2
be used down to a wavelength of 200 nm, while the
application of other organic solvents such as methanol, 0.0
ethanol, is restricted to UV wave-lengths of approximately 190 210 230 250 270 290 310
l 2 0 nm isopropanol > 230 nm, THF > 250 nm. In Percent organic (v/v)
addition,bu^ersoradditivescanalsoa^ectmobilephase Figure 42. UV absorbance of acetonitrile, methanol, ethanol,
transmittance in a negative manner. isopropanol and tetrahydrofuran.

Mobile phase pH 300


The result of a chromatographic separation of ionizable
UV intensity (mAU)

analytes is pH dependent, the exact adherence to a


speci_c,prede_nedpHisthereforeoftennecessary. 200
Figure 43 displays the pH dependent chromatographic
analysisoftheantibioticCefaclor.Whilethee^ectofpH
on this analysis is rather small, shifts in retention time
can be up to several minutes when changing pH from, 100
pH 7.0
e.g., acidic to neutral.
pH 3.5
Bu^ers or bu^er systems are generally used to set a
0 pH <2
pH ionizing or deionizing analytes. The ionic strength
depending
( onbothbu^erconcentrationandioncharge) - 1 2 3 4 5 6
should not be set below 20 mM in order to achieve a Retention time (min)
suacient bu^er capacity. In contrast, ionic strengths Figure 43. pH dependent chromatographic analysis of a Cefaclor
abovemM 0 1 cancausebu^erprecipitationinorganic formulation.
solvents.Verifybu^ersolubilityoverthecompleterangeChromatographic conditions: Purospher™ STAR RP-18 endcapped (2 µm)
Hibar® HR 5 cm x 2.1 mm I.D.; mobile phase A: 10 mM sodium
of0to10%organic.Bu^ers(especiallyforMSuse( phosphatebu^erpH3.5orwater+0.1%TFApH<2or10mMsodium
should always be freshly prepared and utilizing the purest phosphatebu^erpH7,mobilephaseB:acetonitrile;gradient:0min
salt and acid/base quality available. Table 13 summarizes 95%A,1min95%A,6min5%A;‘owrate267µL/min;detectionUV
variousbu^ersystemsincludingtheirpHrange.Notethat 254 nm; injection volume: 0.11 µL; temperature: 30 °C; sample: one
Cefaclor capsule (500 mg) was suspended in 100 mL water, sonicated
the counter ion and its degree of hydration can have an for20minutesand_lteredthrougha0.2mMillex ®_lter.

in‘uenceonthepHofthe_nalmobilephase.

36
Table 13.Bu^errangesofselectedbu^ersystems multiple wavelengths simultaneously. The downside is
Bu^er
thataUVdetectorisnotanalytespeci_candrequires
that the analyte absorb more light than sample matrix
TFA
at the set wavelength. Choose a detection wavelength
Sulfonic acid/NaHSO3
thatmaximizessensitivityandspeci_city,butkeep
Glycine/HCI inmindthatthemobilephasesolventsandbu^er
H3PO4/KH2PO4 components may cause slight shifts in UVmax from
Formic acid reference values. Therefore, it is advisable to check the
Citric acid/Na citrate analyte absorbance in the mobile phase. Mobile phase
Acetic acid/Na acetate solventsandbu^ercomponentsalsohaveUVcut-o^;
Pyridin/formic acid
therefore, make sure to work well above these levels.
Otherwise, there are likely to be problems with reduced
Formic acid/Na formate
sensitivity and increased system noise (unstable and
Pyridin/acetic acid
drifting baseline noise). UV wavelengths below 200 nm
KH2PO4/Na2HPO4 should be avoided because detector noise increases in
BIS-TRIS this region. Higher wavelengths give greater selectivity.
NaHSO3/Na2SO3
TRIS/HCI Refractive index (RI)
Trimethylamine/HCI Refractive index is also a common detection technique,
Na borate/HCI andmeasuresthedi^erenceintherefractiveindexofa
Trimethylamine/CO2 sample cell versus a reference cell. This detector is also
NH4HCO3 a non-selective detection technique, being concentration
(NH4)2CO3/NH3 dependent. The sensitivity is typically 100–1000 times
lower than a UV/Vis detector. The bene_t over a UV
NH3/acetic acid
detector is the possibility to quantify analytes with no
Ethanolamine/HCI
chromophores in the molecular backbone. The drawback is
Na2CO3/NaHCO3 the sensitivity and the fact that RI detectors are typilcally
Na borate/NaOH used in isocratic mode only.
Triethylamine/HCI
Pyrollidine Fluorescence (FL)
Na2HPO4/Na3PO4 Fluorescence detection is speci_c and measures only
pH 0 1 2 3 4 5 6 7 8 9 10 11 12 13 compoundsthat‘uoresce;hence,arequirementofthis
technique. The operation is similar to a UV/Vis detector but
Detector choice wherethedetector‘owcellisusedasthesensorthrough
which excitation light passes axially. A photocell is located
The choice of the detection is critical in HPLC as only at the side of the cell to receive radially emitted light. The
compounds can be analyses if they are detected. Using cell wall is made of special glass to prevent the excitation
a not suitable detector for the compounds of interest the light or other stray light from reaching the photocell.
chromatographic information to this compound will get Whenasolutethat‘uorescesintheexcitationlight‘ows
lost. To select the most appropriate detection mode, four throughthecell,themoleculeexcitesand‘uorescentlight
important parameters should be taken into consideration; passes through the walls of the cell onto the photocell. The
chemical nature of the analytes, potential interferences, excitation light may be light of any wavelength selected
LODandLOQrequired,linearityrange,availabilityand/ from the light source using a monochrometer. Another
or cost of detector. Below are some of the most common monochrometer may also be used to selectively analyze
detection techniques for liquid chromatography presented. the‘uorescentlightandthus,a‘uorescentspectrumcan
Fluorescence, electrochemical or mass detectors should beproducedforexcitationlightofanyspeci_cwavelength
be used for trace analysis. For preparative HPLC, andanexcitationspectrumproducedfor‘uorescentlight
refractive index is preferred because it can handle high ofanyspeci_cwavelength.Toimprovespeci_cityofanLC
concentrations without overloading the detector. analysis,a‘uorescentderivatizationreagentcanbeadded
either
( pre-columnorpost-column)toforma‘uorescent
Ultraviolet/Visible Absorbance (UV/Vis) derivative of the substance of interest. This derivative may
then be selectively detected from other solutes, which,
UV detectors are most commonly used in HPLC. This
if( theydonot‘uoresce)neednotberesolvedfromeach
detector is a robust, inexpensive and versatile detection
other by the separation column. Fluorescence detection is
technique since most compounds absorb light,
up to 1000 times more sensitive than UV/Vis, and is also
especially at low UV wavelengths. It is possible to use
concentration sensitive.
a diode array detector (DAD) and allow monitoring at

37
Evaporative light scattering (ELS) and alternating radio frequency (RF) potentials applied to
ELS is also a non-selective detection technique, but them. The HPLC system handles dissolved analytes under
where the ELS detector (ELSD) is mass sensitive and not ambient pressure (760 Torr) and delivers the sample
concentration dependent. It is an ideal technique for high to the MS, where the detection of the gaseous, ionized
molecular weight compounds, sugars, and less volatile samples is performed under high vacuum conditions
acids. The detector measures the light scattering and (10-5-10-6 Torr). The transfer of the analyte solution
where the amount of scattering is related to the molecular from the LC to the MS is accomplished via an interface.
mass of the analyte, i.e. the more mass the more The interface converts the sample stepwise to an aerosol,
scattering will be seen measured. In the detector, there ionizes it, and removes the solvent. Ions are then focused
are three processes; nebulization of the mobile phase (1), and passed along the middle of the quadrupoles. Their
evaporation of the mobile phase (2) and light scattering movementwilldependontheelectric_eldssothatonly
by analyte particles. In contrast to RI, it works well in ions of a particular mass to charge ratio (m/z) will have
gradient mode. Keep in mind that mobile phase solvents a stable path to the detector. The RF is varied to bring
should be volatile for best performance. ionsofdi^erentm/zintofocusonthedetectorandthus
build up a mass spectrum. Depending on the physical
properties and the molecular mass of the molecules,
Electrochemical (EC) di^erenttypesofinterfacesareused,whichvaryamong
An electrochemical detector requires that the analytes each other by how they ionize the molecules and the
can be oxidized or reduced by an electrical current. pressure applied during this process. At present, all the
Thedetectoroutputisanelectron‘owgeneratedbya common ionization techniques operate under ambient
reaction that takes place at the surface of electrodes. If pressure; i.e. electrospray ionization (ESI), atmospheric
this reaction is complete (exhausting all the analyte), the pressure chemical ionization (APCI), matrix assisted
current becomes zero and the generated total charge laser desorption/ionization (MALDI), and atmospheric
is proportional to total mass of material that has been pressure photo ionization (APPI). ESI and APCI are by far
reacted. This process is called coulometric detection. If the the most widely used in LC-MS hyphenation. The more
mobilephaseiscontinuously‘owingpasttheelectrodes,esoteric techniques, electron ionization (EI) and chemical
the reacting analyte is continuously replaced in the ionization (CI) work under high vacuum conditions with
detector. As long as the analyte is present between the the advantage of being suitable for GC-MS hyphenation.
electrodes, a current will be maintained, albeit varying Quadrupolemassspectrometerscommonlyhavetwo
in magnitude, and is called amperometric detection. An con_gurationswhenusedwithliquid-chromatography,
electrochemical detector requires three electrodes, the either as a simple single quadrupole system or placed in
working electrode (where oxidation or reduction takes tandem. The latter principle, the triple quadrupole mass
place), the auxiliary electrode and the reference electrode spectrometer, enables ion fragmentation studies (tandem
(compensates for changes in the background conductivity mass spectrometry or MS/MS) to be performed.
of the mobile phase). Electrochemical detection is more
sensitivethan‘uorescencedetection,butcommonlynot Electrospray Ionization (ESI)
asselectiveas‘uorescenceandgenerallynotcompatible
with gradient elution. In ESI mode, liquid solutions of charged or polar
substances, delivered with an HPLC system, are
sprayed utilizing a metal capillary (“spray needle”)
Mass spectrometer (MS) and a nebulizer gas (nitrogen) in the MS. Resulting
Mass spectrometry is regarded as an established, droplets are dried (desolvatization) and volatilized,
routine, detection technique. MS detectors can be isolated, analyte ions are transferred to the detector.
coupled to various separation techniques such as liquid Thermal stress is low so the analyte molecules do not
chromatography (LC), thin layer chromatography (TLC), decompose. ESI is almost unlimited regarding molecule
or gas chromatography (GC), where the hyphenation size and suitable for medium to strong polar molecules,
with LC is by far the most frequent setup. In contrast e.g., amines, carboxylic acids, heteroaromatics, and
to more simple detectors, i.e. UV, RI, FL etc., MS sulfonic acids. ESI is applied when fragmentations are
generates data about molecular masses and detailed unwanted and molecular masses of biomolecules have
structuralparametersandtherebyo^ersthepossibility to be determined. ESI-MS is well suited for hyphenation
to discriminate between co-eluting peaks in selected ion withLC,andaslongas‘owratesdonotexceed
monitoring mode. The latter reduces the requirement for maximum 1–2 mL/min (depending on instrumentation),
chromatographic retention and resolution before detection, attainablesensitivityisveryhigh;however,‘owrates
yet it is always better to have retained and completely ofbetween150L/minaremorecommon.Inliquid
resolved peaks to prevent ion suppression or ion solution, molecules are either already ionized, or will
enhancemente^ects.Massanalyzerscanbequadrupole, become protonated or deprotonated by additives in the
magneticsector,time-of-‘ight,iontrap,orioncyclotron sample solution and the mobile phase. To achieve best
resonance type. A quadrupole mass analyzer consists sensitivity, the mobile phases used should be set at
offourparallelrodsthathave_xeddirectcurrenta (DC)pH where analytes are ionized, and a rule of thumb

38
is to use neutral to basic pH (7–9) for acids, whereas Bu^ersdonotonlyadjustthepHoftheeluentand
more acidic pH (3–4) is advisable for basic compounds. lead to ionization of a target molecule, they can also
If the analytes of interest have multiple pKa values and formadductswiththeanalyte.Adducts[M+bu^er],
may change their ionization state, other pH values may e.g. with ammonium, alkali, halogens, formate or
bemorebene_cialbothintermsofionizationofthe acetate, will lead to the detection of an additional peak
analyte and behavior in the column. Thus, depending in the MS spectrum; even a complete suppression of
on the choice of solvent and additives, either positive the analyte signal is possible when the vapor pressure
and/or negative ESI mode can be used. Typically, of the resulting adduct (mainly alkali) is decreased
positive mode is applied in combination with more signi_cantly.Duetothis,andinordertokeepthe
basic molecules, while acid compounds are analyzed in ESIsourceclean,volatilebu^ersarerecommended.
negative mode. 0.1% formic acid is commonly added Non-volatile salts like phosphates, borates, sulfates or
to the mobile phase in positive ESI mode to provide citrates will precipitate in the MS source, block it, and
alowpH(i3)andtoprotonatetheanalyte(s).Acidic cause tedious cleaning procedures.
analytes will be neutralized under such conditions,
accordingly negative ESI mode is preferred and higher Atmospheric pressure chemical ionization
mobilephasepHisrecommended.Volatilebu^erslike (APCI)
ammonium acetate or ammonium formate are used
in the pH range 4.5–7 to deprotonate the analyte(s), This technique is complementary to ESI and also useful
and for high pH, it is possible to use either ammonium for LC-MS hyphenation. It does not require a mobile
carbonate or ammonium hydroxide (aqueous ammonia). phase with conducting properties where acetone or
For both negative and positive ESI, it is a prerequisite acetic acid esters can be used as solvents and thus
that all mobile phase solvents and additives are allows for a coupling of APCI with normal phase
volatile in order to avoid contamination of the mass chromatography. In APCI mode, the analyte solution
spectrometer, and that the total mobile phase ionic is vaporized prior to the ionization. Subsequently
strength is adequate (generally 2–25 mM) to prevent solvent molecules (aqueous-organic, e.g. methanol,
unnecessary down-time for cleaning of the detector. propanol, acetonitrile, acetone etc., combined with
Strong acids like hydrochloric acid or nitric acid are 20mMofavolatileorganicbu^ersuchasformicor
unsuitable for two reasons: they form ion pairs with acetic acid, ammonium acetate, ammonium formate
analyte molecules (analyte signal suppression) and or triethylamine) become ionized with a corona needle
display strong oxidizing properties. where their charge is then transferred to the analyte
molecules via proton transfer or abstraction. APCI is
Tri‘uoroaceticacid(TFA)isaspecialcase:Itiswidely suitable for the analysis of less polar, weakly ionizable
used as an ion-pairing reagent to improve the liquid substances with small or medium molecular weight
chromatographic separation of peptides or proteins. On (analytes without acidic or basic functional groups,
the other hand, TFA can cause strong ion suppression in e.g. hydrocarbons, alcohols, aldehydes, ketones,
mass spectrometry (mainly in negative ESI mode) and esters) and is therefore complementary to ESI, as long
contaminates the LC-MS system. ’A good compromise as the sample is thermally stable and vaporizable.
herewouldbethroughtheuseofdi‘uoroaceticacid Fragmentations are generally observed with APCI.
(DFA). DFA provides the same excellent increase in Highest sensitivity is achieved using acetonitrile,
eaciencyasTFAbutwithoutasmuchionsuppression methanol or water as solvents, and where the degree
nor does it contaminate the MS system as readily as of analyte ionization can be optimized via mobile phase
TFA. Unfortunately, both a quantitative estimation of pH.AsforESI,‘owratesuptomaximum12mL/min
thesee^ectsaswellasgeneralrecommendationsiscan be tolerated. There are other less commonly used
not possible as their strength strongly depends on detection techniques possible to combine with liquid
the MS system used. Triethylamine as an alternative chromatography, such as chemiluminescence nitrogen
additive behaves in a similar manner. If the use of TFA (CLND), radio detectors, charged aerosol (CA, inductive
is unavoidable, a weak acid such as propanoic acid, or coupled plasma (ICP), nuclear magnetic resonance
isopropanol can be added to the mobile phase in order (NMR), but these are not dealt with here.
todecreaseasignalsuppressione^ect.

39
General recommendations Bothmetalandpolymeric_ttings,tubing,andferulesare
available for a proper column installation. Depending on
the application and the setup, an operator can use either
Column hardware
material. The depth of the drill hole of Supelco®-branded
HPLCandUHPLCcolumnscomeinavarietyofdi^erent HPLC column end _ttings is 2 mm [Parker standard
column hardware formats and materials for di^erent format, used by most column manufactures] (as is the
applications (Table 14). Depending on the material visible length of tubing after unmounting of a _tting/
(stainless steel, PEEK) the pressure stability can tubing unit, see Figure 44), while for few other column
vary signi_cantly. manufacturers it is 3 mm. In order to avoid the creation
®
All Supelco columnshaveUNF2 3 0 1 femaleend_ttings of large dead volumes or improper column installation,
that connect to 1/16” capillary tubing. Pre-installed end this fact makes careful handling necessary when working
_ttingsofanytypeofchromatographiccolumnshould with columnsfromdi^erentmanufacturers.Inaddition,
the end _ttingsofsomeHPLCcolumnse. ( g.Chromolith
, ®
not be removed from HPLC columns, because the column
bed might be damaged and the performance reduced. columns) consist of PEEK, while others are made out
For further handling instructions see also the section on of stainless steel. Mounting metal capillaries with 1/16‘‘
column installation. outer diameterandametalcuttingring_xedtoamm 3
drill hole length can damage the PEEK hardware (both
The column hardware of Chromolith® monolithic silica columnhousingandend_tting)andthesilicabedofthe
columns consists of a mechanically stable and chemically aforementioned columns. To avoid any damage, use either
robustpolymerPEEK)( with
, theend_ttingsmadefrom ‘exiblemetalcapillariesmm 5 2 . 0 ( outerdiameter)with
the same material. SeQuant ® particulte columns are
apolyvinylidene‘uoridePVDF)( coneorPEEKcapillaries
packed in a PEEK lined stainless-steel column hardware. with PEEK screws and adjustable plastic ferrules.
Particulate silica for reversed phase and normal phase
HPLC is delivered in stainless steel column hardware with
stainless steel frits to keep the stationary phase particles
in place. The di^erent columns hardwares provide dif-
ferent pressure stabilities (Table 14). Hibar® columns
as well as Chromolith®, SeQuant ®, Ascentis® Express,
Discovery® and Supelcosil™ columns require a separate
pre-column holder for the use of precolumns. LiChroCART®
cartridges allow the direct integration of 4-4 guard-
columns in the cartridge holder “manuCART” which can
bemountedwithouttools(_ngertight).
Drillhole
Cone
Installation of columns with PEEK end fittings
Most HPLC columns are connected to all standard HPLC and UNF thread 10/32”
UHPLC systems with standard 1/16_ttings.
, Shortcapillary Figure 44. Schematic drawing of a Chromolith®end_tting.
tubing is recommended to minimize extra-column volumes.

Table 15

Trademark Pressure
Hardware Trademark Sorbent Column Use Precolumn Material stability
Purospher™ STAR
LiChrospher® Requires
HPLC direct integration of precolumn—
LiChoCART® Superspher® manuCART® Stainless Steel 250 bar
Cartridge no separate holder needed
LiChrosorb® to use

Aluspher®
Purospher™ STAR
LiChrospher®
Hibar® RT HPLC column ready to use column separate precolumn holder required Stainless Steel 400 bar
Superspher®
LiChrosorb®
Hibar® HR Purospher™ STAR UHPLC Colum ready to use column No precolumns available Stainless Steel 1000 bar
®
SeQuant U/HPLC Colum ready to use column separate precolumn holder required PEEK lined Stainless Steel 200–550 bar
Chromolith® U/HPLC Colum ready to use column separate precolumn holder required PEEK 200 bar
Discovery®
Ascentis®
Supelcosil™ HPLC Colum ready to use column separate precolumn holder required Stainless Steel 400 bar
[LiChrospher®]
[LiChorsorb®]
Ascentis® Express
HPLC Colum ready to use column separate precolumn holder required Stainless Steel 600 bar
BIOshell™
Ascentis® Express (2 µm)
BIOshell™ (2 µm) UHPLC Colum ready to use column separate precolumn holder required Stainless Steel 1000 bar
Titan™

40
Equilibrating the Column Reversed phase columns are shipped in acetonitrile/water
In order to conduct reproducible results, every (e.g., 60/40 v/v). As the columns can dry out during
chromatographiccolumnhastobe‘ushedwitheluent stocking and shipping (this is of special importance with
under the starting conditions of a chromatographic run. cartridges, because the columns are closed but not tight
Isocraticseparationsrequire‘ushingwithcolumn 02 5 enough and can dry out easily), the column packing has
volumes of mobile phase, depending on whether a purged to bewettingthoroughlyby‘ushingwithcolumn 02 1
system is used and whether the column is new or has volumes of pure organic solvent (e.g., acetonitrile or
been used before, plus instrument dead volume. Under methanol). Then conditioning of the column with mobile
gradientconditions‘ushingwith510columnvolumes phase has to be continued until the baseline has stabilized.
of the mobile phase will be suacient for reproducible Of course, the miscibility of mobile phases is a prerequisite
runs. Without equilibration, low reproducibility, a shift for successful equilibration.
in retention times, and peak overlap can be observed Normal phase columns (Si, NH2, CN, Diol) are typically
(Figure 45). Note that Table 15 summarizes gross shipped with n-heptane/dioxane (99/1 or 95/5 v/v). It
column volumes. Depending on the stationary phase, the is recommended to perform equilibration with dioxane,
net column volume will be in the range of approximately followed by the mobile phase. If columns are going to be
60 8 % of the listed value. The ‘ow rate utilized for usedwithaqueouseluents,‘ushthecolumnwithethanol
column equilibration is limited by the back pressure or 2-propanol before equilibration with the mobile phase.
generated by the packing material. Due to this, columns Monolithic silica NH2 columns are shipped in acetonitrile/
with a monolithic silica backbone creating a low back water (90/10, v/v). The shipment solvent is described in
pressurecanbeequilibratedsigni_cantlyfaster(and theatColumn Care & Use sheet, it is recommended to start
much higher ‘ow rates) compared to particle packed the equilibration of the column in this solvent mixture,
columnswithparticlesizesof5morless. followed by the mobile phase.

Table 15. Gross column volumes of various column formats (length: Column coupling
100 mm).
The separation eaciency of HPLC columns is mostly
Column I.D. (mm) Grosscolumnvolume(µL) reported as relative separation eaciency in plates pe
4.6 1,662 meter (N/m). For complex separations it can often be
3 707
necessary to use long columns in order to provide the
separation eaciency required for the resolution of all
2 314
compounds of interest. With respect to column coupling,
0.3 7.1 the use of particle packed columns is restricted due to
0.2 3.1 back pressure issues.
0.1 0.79 The typical relative separation eaciency of most
0.075 0.44 monolithic silica HPLC columns (Chromolith® Performance)
is similar to 5 µm particles—about 100,000 N/m. Due to
the low back pressure of the monolithic column bed, these
500 columns can be connected in series up to a length of 1 m,
resulting in more than 80,000 plates (absolute, although
400 somewhatlessthanintheoryduetothein‘uenceofthe
Intensity (mAU)

column couplers used) at relatively low back pressure.


300 Figure 46 displays the separation of alkyl benzenes on
ten coupled monolithic silica columns.
200 6 7
5
UV intensity (mAU)

100
3
0 4
2
0.0 0.5 1.0 1.5 2.0 2.5 3.0
Retention time (min)
Figure 45. Separation of alkyl phenones without and after proper
column equilibration (blue and black chromatogram, respectively).
Chromatographic conditions: Purospher™ STAR RP-18 endcapped (2 µm) 1
Hibar® HR 5 cm x 2.1 mm I.D.; mobile phase A: water, mobile phase
B: acetonitrile; gradient conditions: 0 min 45% B, 2.5 min 95% B;
injectionvolume2.1L;detection:UV(247nm);temperature:40°C; 0 5 10 15 20
‘owrate:mL/
0.54 min;sample:urea,
1 acetanilide,
2 acetophenone,
3 Retention time (min)
4 propiophenone, 5 butyrophenone, 6 benzophenone, 7 valerophenone,
8 hexanophenone, 9 heptanophenone. Figure 46. Isocratic separation of alkyl benzenes on ten coupled
monolithicsilicacolumnsyieldinganabsoluteeaciencyofplates 0 ,38
atabackpressureofbars.
07 Columnswereconnectedutilizingspeci_c
column connectors.
Chromatographic conditions: Ten pieces Chromolith® Performance
RP-18e10cmx4.6mmI.D.mmplusspeci_ccolumncouplers;mobile
phase:acetonitrile/water80/2(v/v);‘owrate2mL/min;detection
UV 210 nm; sample: 1 uracil, 2 toluene, 3 ethyl benzene, 4 propyl
benzene, 5 butyl benzene, 6 pentyl benzene, 7 hexyl benzene.

41
Column lifetime, cleaning and regeneration Exposure of a column to samples or solvents containing
Column lifetime is highly dependent on the sample highly adsorptive components (CRUD; Chromatograph-
properties (particle load, impurities) and the HPLC method ically Retained Undesired Debris) or even particulate
used (eluent pH, temperature), and cannot be generalized. matter will result in increased back pressure, a change in
selectivity,andadropofseparationeaciency.Indetail,
In certain situations 50,000 injections on one column are possible reasons are microbiological impurities in the
possible and in other cases only a few (less than 10). This mobile phase, mechanical abrasion from components
fact depends on the sample properties, method criteria, of the HPLC system, matrix components from biological
andthemethodandsystemsuitabilitycriteriade_ned: samples,orparticlesinthemobilephase.Reversethe‘ow
periodically to prevent particles and non-eluting sample
• platenumbers(eaciency/sensitivity) components from accumulating on the column. This is
• selectivity (how well separated peaks are) only allowed for columns which have the same frits on the
top and the bottom. The column can often be restored to
• tailing factor (how symmetric peaks you expect) original performance by using suitable wash protocols (see
above). When performing solvent rinse regeneration, the
column should be transferred from the analytical HPLC
Column back pressure (how stable the method is and
system to a simple, inexpensive pump. Alternatively,
how little matrix accumulates on the column with time)
disconnect the column from the detector and rinse directly
can also be a critical issue here. to waste. Rinse with a minimum of 20, preferably 30,
Many injections on one column can be expected if one column volumes of each solvent. Every column has insert
ormoreofthefollowingcriteriaareful_lled: sheet with washing recommendations, and users must
follow insert sheet guidelines.
• big gap in retention time between peaks (high resolution)
If a reversed phase column (RP-18, RP-8, Diol, CN,
• low concentration and volumes of sample are injected NH2 if used in RP mode) is strongly contaminated,
ontothecolumn,solventsareofhighqualityand_ltered then pump the following solvents one after the other
(ifbu^ersareadded) through the column for 5 minutes at the upper limit
of the corresponding column optimal ‘ow-rate range:
• peak shape changes are not too relevant
water, acetonitrile, 2-propanol, n-heptane, 2-propanol,
• software can still integrate peak area adequately acetonitrile, water, mobile phase.
For cleaning and regeneration of polar phases (Si,
If many peaks elute within a short time and with big Diol, CN, NH2)connectthecolumninthereverse‘ow
di^erencesinpeakamplitude,andsharpsymmetricaldirection, then pump the following solvents one after the
peaks with no co-elution are expected, a column might other through the column under the same conditions
need to be changed more regularly. Therefore, if a as described above: n-heptane, chloroform, ethanol
method is set up, and it is planned to run the method or 2-propanol, chloroform, heptane, mobile phase. For
overalongperiod,itishighlyrecommendedtode_ne normal phase columns use n-heptane, n-heptane/dioxane
limits of the criteria listed. This fact makes it easy to 50/50, dioxane, n-heptane/dioxane 50/50 as solvents/
decide when to change the column. solvent mixtures. Every column has insert sheet with
washing recommendations, and users must follow insert
For samples with large quantities of contaminants, it is sheet guidelines.
recommended to apply one or more sample preparation
Whenre-reversingthe‘ow,‘ushthecolumnbefore
methods prior to separation (e.g. solid phase extraction,
connecting it to the detector.
_ltration, centrifugation; see also chapter 5). An
alternative or additional option is to apply guard columns. A documentation of HPLC column history makes sense in
It is generally good practice to protect the analytical ordertotraceanyissuespossiblya^ectingcolumnlifetime
column with such a pre-column in order to ensure in a negative manner. This makes sense especially when
maximum column lifetime. Use of a pre-column might acolumnisrununderverydi^erentconditionssample (
result in a slight shift of the chromatographic parameters. load, contaminated sample, extreme pH).
Make sure that the samples and the mobile phases are
clean and particulate free by using HPLC grade solvents Use with mass spectrometers—procedure to
andreagents.Ifbu^ersorothersaltsareused,a_nalmaintain low column bleeding
_ltrationofthemobilephaseshouldbecarriedoutUnless with an LC run is performed utilizing a normal phase
amembrane_lter. column, the stationary phase of every HPLC column
contains covalently bound organic entities altering its
To extend the lifetime of the column, “wash” the
physical properties. Depending on the quality of both
column after use and before storage to remove traces
phasemodi_cationandasubsequentwashingstep
ofsamplesandbu^ersfromthecolumn.Forcleaning
these entities (e.g. octadecyl, cyano, phenyl) can be
of non-polar phases (RP-18, RP-8, Diol, CN, NH2 if used
washedo^thecolumnduringachromatographicrun
in RP mode), connect the column in the reverse ‘ow
and cause weak to severe interfering signals. This
direction. The simplest procedure is to pump 100%
unwanted phenomenon is referred to as “column
methanol or acetonitrile for 5 min at middle optimal
bleeding” and leads to decreased sensitivity in MS.
‘owrate.Ifbu^ershavebeenused,_rstpump% 0 1
water and then methanol. Note: Ion pairing reagents are Aminimizationofcolumnbleedispossibleby‘ushingof
not completely removable from a stationary phase. an RP-18 or RP-8 column prior to connection to an LC/MS

42
instrument using a mixture of isopropanol and 0.1% formic Na+
athalfoptimum‘owrateforapproximatelyminutes. 03
This process cleans any trace organic material out of the N– O
F
column and hence increases sensitivity by decreasing S N
background noise. Plain Si columns should be washed with
dioxaneatmiddleoptimal‘owrateforthesameperiodof
F O N
time. As an alternative, 2–3 gradient runs from strongly
aqueous to strongly organic conditions can also be utilized
to wash a column prior to connecting it to the MS detector. Figure 47. Chemical structure of
pantoprazole sodium. O O
Column bleed will generally be low when the maximum
solvent strength of the mobile phase used is equivalent The original method was developed on a Purospher™ STAR
to methanol or acetonitrile. If stronger solvents are used RP-endcapped
81 cm
5 1 xmm
6 . 4 I.D.columnwithm5
in the mobile phase, e.g. tetrahydrofuran or DMSO, then particles with a total cycle time of 50 minutes (Figure 48).
itisrecommended_rsttopumpapproximately10mL By changing to a Purospher™ STAR RP-18 endcapped
of this stronger mobile phase plus 0.1% formic acid cm
5 xmm 1 . 2 I.D.columnwithm2 particles,lowering
through the column before connecting to the detector. the ‘ow-rate, and altering the gradient pro_le, it was
possible to reduce the total analysis time from 50 to
5 minutes, maintaining sample peak pro_le (with
Storage improved resolution), at low back pressure and high
Reversed phase columns should be stored in pure separationeaciency Figure
( 49).
acetonitrile or a mixture of organic solvent (e.g.
50
acetonitrile or methanol) and water (e.g. 50/50 v/v).
Polar phase columns are best stored in heptane/

11.914/Pantopraole Na

18.025/ImpB
40
dioxane (80/20).
Storingthecolumninbu^ersoratunsuitablepHfora30
prolonged time will shorten the lifetime of the column.
Before extended storage (i.e. over the weekend or 20

14.407/Imp D

15.848/IUnknown
16.514/IUnknown

18.420/IUnknown
long term storage), the columns should be thoroughly

14.861/Imp E
13.799/Imp F
10.791/Imp A
8.088/Imp C
rinsed-outfrombu^ersalts,orion-pairreagents 10
which can cause bacterial growth or precipitate in the
stationary phase or the HPLC system. The following 0
procedureisrecommended,_rst‘ushthecolumnwith
0 5 10 15 20 25 30
102columnvolumesofmobilephaseminusbu^er, Retention time (min)
then with 10–20 column volumes of the shipping
Figure 48.Chromatogramshowingthepuritypro_leofpantoprazole
solvent mixture (acetonitrile/water, 75/25). sodium on a Purospher™ STAR RP-18 endcapped 15 cm x 4.6 mm I.D.
columnwith5mparticles.
Alternatively, follow this protocol if the mobile phase
Chromatographicconditions:Purospher=STARRP-endcapped 81 m)5(
contains a bu^er: Flush with 5 10 column volumes 15 cm x 4.6 mm I.D.; mobile phase A: 1.74 g dipotassium hydrogen
water, then with 20–50 column volumes organic solvent/ phosphate in 1000 mL water, adjusted to pH 7.0 with dilute phosphoric
water (e.g. acetonitrile/water, 1/ ). After ‘ushing acid (330 g/L), mobile phase B: acetonitrile; gradient: 80% A to 20% A in
min,
0 4 min
01 reequilibrationatA;% 0 8 ‘owratemL/ 0 . 1 min;temper-
with 20 column volumes of the storage solvent (e.g. ature°C;0 4 injection:L;detection:
02 UVnm;
0 9 2 sample:ppm 0 6 4 of
preferably acetonitrile), the column can be easily stored. pantoprazole sodium in 1:1 mixture of ACN and 0.001 N NaOH.
By rinsing with acetonitrile, aprotic impurities can also
be removed from the column. It's recommended to users 100,000
2.369

to follow insert sheet recommendations.


75,000

System optimization
4.047

50,000

To_ndthedesiredbalancebetweenresolutionand
1.911

analysis time after satisfactory selectivity has been 25,000


1.236

3.096

achieved, parameters such as column dimension,


3.344
2.860

3.477

4.482
3.611

particlesize,and‘owrateshouldbeoptimized.With 0
a truly scalable stationary phase, these parameters
maybechangedwithouta^ectingcapacityfactors 0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0
Retention time (min)
or selectivity. With the introduction of smaller
particle sizes and narrower column inner diameters, Figure 49.Chromatogramshowingthepuritypro_leofpantoprazole
optimization of the complete HPLC instrumentation is sodium on a Purospher™ STAR RP-18 endcapped 5 cm x 2.1 mm I.D.
columnwith2mparticles.
also needed, and sometimes it is necessary to replace Chromatographicconditions:Purospher=STARRP-endcapped
81 m)2 (
the whole system. An example of a successful complete 5 cm x 2.1 mm I.D.; mobile phase A: 1.74 g dipotassium hydrogen
system optimization is shown for Pantoprazole sodium phosphate in 1000 mL water, adjusted to pH 7.0 with dilute phosphoric
acid (330 g/L), mobile phase B: acetonitrile; gradient: 80% A to 72% A
(Pantoloc, Protium, Pantecta and Protonix; a proton in 1.5 min, 72% aA to 60% A in 2.5 min, 1 min reequilibration at 80% A;
pump inhibitor drug that inhibits gastric acid secretion; ‘owrate0.6mL/min;temperature40°C;injection:7L;detection:
for chemical structure see Figure 47. UV 290 nm; sample: 460 ppm of pantoprazole sodium in 1:1 mixture
of ACN and 0.001 N NaOH.

43
Method validation but depending if it is a pharmaceutical assay or a bio-
analyticalmethod,di^erentacceptancecriteriagovern.
Proper validation of an analytical method is important to In pharmaceutical quality control there are much more
ensure that it will provide similar results, today, tomorrow, stringent method requirements and less variation
next week, next year, i.e. over a long period of time, in amongst samples compared to analysis of patient
di^erentlaboratoriesandindependentoftheanalyst.plasma Not or serum samples. For any assay, the relative
only because of requirements from regulatory authorities, standarddeviationRSD) ( orcoeacientofvariationCV) (
but rather to ensure good manufacturing practice (GMP) is used as indication of the imprecision of the method.
and good laboratory practice (GLP). It is especially From a practical perspective, six to ten replicate
important for pharmaceutical analysis when assurance of injections will give you a good idea of the precision of
thecontinuingeacacyandsafetyofeachmanufactured the method. An analytical method can be accurate but
batch relies solely on the determination of quality. not precise, precise but not accurate, neither, or both.
Guidelines for the validation of analytical methods can be
found at the International Council on Harmonization (ICH). Specificity
The US Food and Drug Administration (FDA) and USP both
refer to ICH guidelines. Speci_city is an important parameter to test in a
validation program as it veri_es the ability of the
Keep in mind that analytical method validation should method to accurately measure the analyte response in
be isolated from the initial selection and development, the presence of all potential sample components. The
whichactuallyareonlythe_rststepsinestablishing analyte response from a solution containing only the
a routine analytical method. Validation means testing analyte is compared with test samples containing the
ofthemethodto_ndoutallowedvariabilityforeach analyte and all potential sample components (placebo,
method parameter. Routine quality control methods synthesis intermediates, excipients, degradation
should guarantee that the analytical results of raw products and impurities). For pharmaceuticals, stress
materials, excipients, intermediates, bulk products or conditions such as heat, light, acid, base and oxidant
_nishedproductsareviable. are typical. For formulated products, heat, light and
humidity are commonly used to stress the samples. The
In this section the most widely applied validation char-
analyte peak is evaluated in all test samples for peak
acteristics are explained; accuracy, precision (repeat-
purity and resolution from the nearest eluting peak.
ability and reproducibility/intermediate precision),
speci_city, limit of detection, limit of quantitation,
linearity, robustness and stability of analytical solutions. Limits of detection and quantitation
TheLODisde_nedastheleastamountofananalyte
Accuracy in a sample that can be detected, and commonly
expressed as the concentration level that is able to
An analytical method is accurate if it gives the right
provideasignal-to-noiseratioofthreeS/( N=3LOQ . ) is
numerical value for the analyte (either mass or
de_nedasthelowestanalyteconcentrationleveltha
concentration) and can be described as the degree
canbequanti_edwithgoodprecisionandaccuracy,
of closeness of measurements of a quantity to its
and providing a signal-to-noise ratio of ten (S/N=10).
actual value. A method almost never gives the exact
LODandLOQcanalsobedeterminedbasedonthe
same results for replicate analyses, which means that
standard deviation of the response and the slope of
the result is presented as the mean or average. A
the calibration curve.
pragmatic way to express accuracy is to present it in
termsofthestandarderror,whichisthedi^erence
between the observed and the expected concentrations Linearity
of the analyte. To determine accuracy, a common The linearity of an analytical method is the capability
practice is to analyze a known amount of standard to generate results that are directly proportional to the
materialunderdi^erentconditionsinaformulation, concentration of analyte in the sample. It is commonly
bulk material or intermediate product to ensure that illustrated as the interval between the upper and lower
nothing interferes with the method. analyte concentration levels that may be determined
with precision and accuracy. Linearity data is often calcu-
Precision latedusingthecalibrationcurvecorrelationcoeacie
and the y-intercept. The RSD, intercept and slope of the
Precision is the ability of repeatedly performing an
calibration curve should also be calculated.
analysiswithalowstandarddeviation.Adi^erentiation
is made between repeatability and reproducibility.
Repeatability is the measure of how easy it is for Robustness
an analyst in a given laboratory to attain the same The method robustness is a measure on how well
result for the same batch of samples (normally by an analytical method remains una^ected by small
injecting the same samples repeatedly at di^erent variations in the experimental conditions, but also how
concentration levels) using the same method and using reliable the method is during normal use. Important
the same equipment and reagents. Reproducibility or parameters to monitor are changes in the mobile phase
intermediate precision measures the variations within composition, mobile phase pH, changes in the gradient
or between days, analysts and equipment. Highly pro_le,changesinthebu^erconcentration,column
reproducible quantitative results should be expected, temperature and injection volume.

44
Analytical solution stability Scaling the flow rate
Analyticalsolutionstabilitycanbedividedintodi^erent Decreasing the internal diameter of the column (e.g. from
sections; recovery, dilution, internal standard addition mm
6 . 4 tomm)1 . 2 requiresrecalculationofcolumn‘ow
etc. If an extraction process is used (either liquid-liquid rate in order to maintain linear velocity. Linear velocity is
or solid phase extraction), it must provide proper analyte de_nedasthedistancewhichmobilephasetravelsover
recovery. A method with low analyte recovery and/ timecm/ ( min)whereas
, ‘owrateisthevolumeofmobile
or where the analyte is degraded during the sample phase that travels over time (mL/min). To maintain the
preparation is not tolerable for routine quality control. same linear velocity through a column with a smaller
Internal or external standards (reference materials) internal diameter, the ‘ow rate must be decreased
should be prepared in such a way that they maintain their proportionally to the column internal diameter according
potency, and produce same response over time. Samples to the equation below.
and standards should be tested for stability to verify
stability over a normal analysis cycle. A rule of thumb is
Flow rate f2 = f1 x d22/d12
that the sample solutions, standard solutions and HPLC
mobile phases should be stable for minimum 24 hours f1 =HPLC‘owrate(mL/min)
underde_nedstorageconditions. f =UHPLC‘owrate
2
d1 = HPLC column i.d. (mm)
d2 = UHPLC column i.d.

Scaling of HPLC methods


Scaling from HPLC to UHPLC
Scaling the injection volume
A transfer of HPLC methods to UHPLC requires scaling
Decreasing the column internal diameter and length,
down from columns with larger inner diameter (e.g.
decreases the overall column volume and sample
4.6 mm i.d.), to columns with a smaller inner diameter
capacity. Therefore, the injection volume must be
(e.g. ~2.1 mm i.d.), and from long columns (e.g. 150 mm
altered. Note that since overall column volume has
length), to short columns (e.g. 50 mm length), in
decreased, it is more important to match the sample
additiontothereductionofparticlesizese. ( g.fromm5
solvent to the starting mobile phase composition.
to2m).
Mismatched sample solvents can cause irreproducible
To ensure equivalent chromatographic separation, it is retentiontimes,eaciencies,andevenchanges
alsonecessarytoscalethe‘owrate,injectionvolume in selectivity. If using a larger injection volume
and the gradient parameters. than calculated, check for peak abnormalities and
irreproducibility that could result from phase overload.
Adjusting the column length
The_rststepistodeterminetheappropriatecolumn Injection volume V2 = V1 x (d22/d12) x (L2/L1)
length in order to maintain the same separation.
Keeping the same column length while decreasing the V1 = HPLC injection volume
particle size will increase the number of theoretical V2 = UHPLC injection volume
plates as well as back pressures. Therefore, when L1 = HPLC column length (mm)
decreasing particle size, column length can be L2 = UHPLC column length
shortened without losing resolution.

Column length L2 = L1 x dp2/dp1 Adjusting gradient time


L1 = HPLC column length When an analytical method is scaled down, the time
L2 = UHPLC column length program of the gradient also needs to be scaled down
dp1 = HPLC particle size to keep the gradient volume the same.
dp2 = UHPLC particle size
Time t2 = t1 x (f1/f2) x (d22/d12) x (L2/L1)

t1 = HPLC time (min)


t2 = UHPLC time

Make sure the dwell volume (gradient delay volume)


of the system has been determined, and take it into
account when scaling a separation or transferring
methods from one HPLC system to another.

45
Scaling an HPLC method 0 0
0

C
Purospher=STAR5mcolumn H2C N
from H2N NH2 H
dimension 15 cm x 4.6 mm I.D.
1. Urea 2. Acetanilide 3. Acetophenone
Purospher=STAR2mcolumn
to
dimension 5 cm x 2.1 mm I.D.
0 0
0

Separation of a mixture of nine alkylphenones (Figure 50)


was scaled from HPLC to UHPLC conditions (Figure 51).
All calculations were following the equations on the 4. Propiophenone 5. Buthyrophenone 6. Benzophenone
previous page.
0 0 0
Flow rate
f2 = 1.3 x 2.12/4.62 = 0.27 mL/min CH3

Time 7. Valerophenone 8. Hexanophenone 9. Heptanophenone


t2 = 15 x (0.5/0.105) x (2.12/4.62) x (50/150) = 5 min
Figure 50. Alkylphenone standards utilized for HPLC method scaling
Injection volume and speeding up HPLC method
V2=10x(2.1/46)x(50/1)=0.7L

Purospher™ STAR RP-18 endcapped, 5 µm Purospher™ STAR RP-18 endcapped, 2 µm


2,000 2,000

1,500

1,000 1,000

500

0 0
–200
0 2 4 6 8 10 12 25 0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 6.0 6.5 7.0 7.5
Retention time (min) Retention time (min)

Column Purospher™ STAR RP-18e (5 µm) Column Purospher™ STAR RP-18e (2 µm)
LiChroCART® 150 x 4.6 mm Hibar® HR 50 x 2.1 mm
Mobilephase A:Milli-Q ® Ultrapure Water Mobilephase A:Milli-Q ® Ultrapure Water
B: Acetonitrile B: Acetonitrile
Gradient Time (min) %A %B Gradient Time (min) %A %B
0.0 55 45 0.0 55 45
15.0 55 45 5.0 55 45
Flow rate 1.3 Flow rate 0.27
Detection UV 247 nm Detection UV 247 nm
Temperature 40 °C Temperature 40 °C
Equilitration 7.5 min Equilitration 2.5 min
Injection 10 µL Injection 0.7 µL
volume volume
Sample Alkylphenone standard Sample Alkylphenone standard
1. Urea 1. Urea
2. Acetanilide 2. Acetanilide
3. Acetophenone 3. Acetophenone
4. Propiophenone 4. Propiophenone
5. Butyrophenone 5. Butyrophenone
6. Benzophenone 6. Benzophenone
7. Valerophenone 7. Valerophenone
8. Hexanophenone 8. Hexanophenone
9. Heptanophenone 9. Heptanophenone

Figure 51. Scaling a method from a HPLC column to a UHPLC column


46
Speeding up a scaled method Scaling from HPLC to UHPLC can speed up the separation
up to 10 times and save solvent by up to 90% at the same
After the method is scaled to a smaller column dimension, time. The separation of 9 alkyl phenones shown in the
thenextstepistoincreasespeedby‘owrate.The example was achieved in 22.5 minutes using a 150–4.6 mm
gradient was adjusted according to the equation: Purospher=STAR5mcolumnat1.3mL/min‘owrate
Time t2 = t1 x f1/f2 (29.5 mL per run). The method was scaled to a column
dimension of 50 2.1 mm using a 2 m material of the
Time t2 = 5 x (0.27/1.08) = 1.25 min same sorbent. The new UHPLC method total run time is
2.15 minutes including re-equilibration of the gradient at a
‘owrateofmL/
8 0 . 1 minmL
3 0 . 2 ( perrun)( Figure 52).

Purospher™ STAR RP-18 endcapped, 2 µm Purospher™ STAR RP-18 endcapped, 2 µm


2,000 1,500

1,500
1,000

1,000

500
500

0 0

0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 6.0 6.5 7.0 7.5 0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6
Retention time (min) Retention time (min)

Column Purospher™ STAR RP-18e (2 µm) Column Purospher™ STAR RP-18e (2 µm)
Hibar® HR 50 x 2.1 mm Hibar® HR 50 x 2.1 mm
Mobilephase A:Milli-Q ® Ultrapure Water Mobilephase A:Milli-Q ® Ultrapure Water
B: Acetonitrile B: Acetonitrile
Gradient Time (min) %A %B Gradient Time (min) %A %B
0.0 55 45 0.0 55 45
5.0 55 45 1.25 55 45
Flow rate 0.27 Flow rate 1.08
Detection UV 247 nm Detection UV 247 nm
Temperature 40 °C Temperature 40 °C
Equilitration 2.5 min Equilitration 0.625 min
Injection 0.7 µL Injection 0.7 µL
volume volume
Sample Alkylphenone standard Sample Alkylphenone standard

Figure 52.Scalingamethodtoahigher‘owrate

47
Ramipril and Related Substances— RamiprilandRelatedSubstances(Purospher=STAR
from HPLC to UHPLC RP-18endcapped(HPLC)
Thebene_tofscalingfromHPLCtoUHPLCisillustrated 240
with the USP 36–NF 31 monograph method for Ramipril O
related compounds, where the liquid chromatograph O

should be equipped with a 210 nm detector and a 180


HN

Intensity (mV)
2504.mmcolumnthatcontains3mpackingL1

Impurity B
O H
(RP-18) and is maintained at a temperature of 65 °C.
120
O N=
Within the scope of allowed monograph method changes, HO =
and only to perform partial revalidation, this method can H

Impurity A
be changed by: 60

• Reductionofparticlesizetomaximum1.5m(50%)
0
• Shortening the column to a length of 75 mm (70%) 0 5 10 15 20 25 30 35 1.4 45
Retention time (min)
• Reduction of inner diameter if linear velocity is
kept constant
Column Purospher™ STAR RP-18e (5 µm)
• Reduction of injection volume as long as limit of Hibar® RT 250 x 4.6 mm
detection (LOD) and linearity is OK. Mobilephase A: Dissolve 2.0 g of sodium perchlorate in a
mixtureofmL
0 8 ofMilli-Q ® Ultrapure Water and

0.5 mL of triethylamine. Adjust pH to 3.6 with


Using the same mobile phases and gradient program phosphoric acid. Add 200 mL acetonitrile and mix.
aspermonograph,thismethodwas_rst_nalizedon B: Dissolve 2.0 g of sodium perchlorate in a
a 250–4.6 mm Purospher™ STAR RP-18 endcapped mixtureofmL
0 3 ofMilli-Q ® Ultrapure Water and

0.5 mL of triethylamine. Adjust pH to 2.6 with


columnwith5mpacking,page20,andthereafter
phosphoric acid. Add 700 mL acetonitrile and mix.
scaled to a 100–2.1 Purospher™ STAR RP-18 endcapped
columnwith2mpacking.TheUHPLCapplicationis Gradient Time (min) %A %B
0.0 90 10
anallowedmonographmodi_cationperUSPguidelines 6.0 90 10
7.0 75 25
but the application using the larger HPLC column is 20.0 65 35
not allowed. It is possible to reduce particle size by a 30.0 25 75
maximum of 50%, but no increase is allowed (Figure 53). 40.0 25 75
45.0 90 10
55.0 90 10
Performance criteria Flow rate 1.0 mL/min
Chromatograph the Resolution solution, and record the Detection UV 210 nm
peak responses as directed for Procedure: the resolution,
Cell 10 µL
R, between ramipril related compound A and ramipril
is not less than 3.0. Similarly chromatograph the Test Temperature 65 °C
solution, and record the peak responses as directed for Diluent Solution A
procedure: the retention time for ramipril is between Injection volume 10 µL
16 and 19 minutes; and the tailing factor for the ramipril
Pressuredrop 61 to 74 Bar (884 to 1073 psi)
peak is between 0.8 and 2.0. Chromatograph the
Standard solution, and record the peak responses as Sample Dissolve 25 mg of sample in diluent and dilute
to 25 mL with same solvent.
directed for Procedure: the relative standard deviation
for replicate injections is not more than 5.0 %. [The
Chromatographic Data
relative retention times are about 0.8 for ramipril related
compound A, 1.0 for ramipril, 1.3 for ramipril related Compound RT (min) RRT Asymmetry
compound B, 1.5 for ramipril related compound C, and 1. Ramipril RS A 18.2 0.82 1.0
1.6 for ramipril related compound D.]
2. Ramipril 22.2 1.00 1.0
NOTE: No Ramipril related compound C and D were 3. Ramipril RS B 30.9 1.39 1.0
available at time of developing this application. It
is therefore not marked as an USP method, despite Figure 53. Scaling USP method for Ramipril from HPLC conditions to
following the monograph experimental conditions. UHPLC conditions

48
RamiprilandRelatedSubstances(Purospher=STAR Ramipril and Related Substances—
RP-18endcapped(HPLC) from HPLC to UHPLC
240 As can be seen on the left page, both columns meet the
O
performance criteria in terms of:
O
180 • The resolution, R, between ramipril related compound
HN
A and ramipril (not less than 3.0)
Intensity (mV)

O H
O N= • The relative retention time between ramipril related

Impurity B
120
compound A (ramipril RS A), ramipril, and ramipril
HO =
H related compound B (ramipril RS B)
60 Impurity A
• The tailing factor for the ramipril peak (between 0.8
and 2.0).
0 • The application using HPLC conditions also meet the
0 2 4 6 8 10 12
Retention time (min) retention time requirement for ramipril
The UHPLC column, Purospher™ STAR RP-18
Column Purospher™ STAR RP-18e (2 µm) endcapped(2m)10x2.1mm,thusseemsto
Hibar® RT 100 x 2.1 mm comply with the monograph method and leads to the
Mobilephase A: Dissolve 2.0 g of sodium perchlorate in a followingbene_ts:
mixtureofmL
0 8 ofMilli-Q ® Ultrapure Water and

0.5 mL of triethylamine. Adjust pH to 3.6 with 1. Faster method (Time-saving: 40 minutes per
phosphoric acid. Add 200 mL acetonitrile and mix. sample or 360%)
B: Dissolve 2.0 g of sodium perchlorate in a
mixtureofmL
0 3 ofMilli-Q ® Ultrapure Water and
(yes... the column length is 60% shorter and this
0.5 mL of triethylamine. Adjust pH to 2.6 with
phosphoric acid. Add 700 mL acetonitrile and mix.
provides 60% time saving but the real gain is to
scale the method to a column with smaller particle
Gradient Time (min) %A %B
0.0 90 10
size and not having to keep same linear velocity).
1.66 90 10
1.93 75 25
2. Higherchromatographicresolutionandeaciency
5.54 65 35
8.31 25 75
11.08 25 75 … but this is not true. The retention time requirement
12.46 90 10 for ramipril is NOT between 16 and 19 minutes. In
15.23 90 10 addition,the‘owratehasnotbeenscaledtomaintain
Flow rate 0.3 mL/min same linear velocity.
Detection UV 210 nm
The monograph method is documented at 1.0 mL/min
Cell 2.5 µL (use 0.1 mm tubing) on 4.6 mm column, and thus the ‘ow rate should be
Temperature 65 °C reduced by a factor or 4.8 for the 2.1 mm i.d. UHPLC
Diluent Solution A
column (for calculations, see page 18). A ‘ow rate of
0.2 mL/min should have been used instead of 0.3 mL/
Injection volume 2 µL
min. With the current experimental conditions, this would
Pressuredrop 196 to 164 Bar (2827 to 2378 psi) give comments from an auditor and very likely a request
Sample Dissolve 25 mg of sample in diluent and dilute for method change.
to 25 mL with same solvent.
ThelargerPurospher=STARRP-18endcapped(5m)
Chromatographic Data
250x4.6mmcolumncande_nitelynotbeused.The
particle size is larger than monograph method and
Compound RT (min) RRT Asymmetry would require complete revalidation and discussion with
1. Ramipril RS A 5.6 0.81 1.1 auditor and authorities. Most likely it would not be an
2. Ramipril 6.9 1.00 1.1
accepted method.
3. Ramipril RS B 9.5 1.38 1.1

Figure 53. Scaling USP method for Ramipril from HPLC conditions to
UHPLC conditions

49
4. C
 hromatographic Separation of
Large Molecules
Introduction whichisresponsibleforthee^ectorfunction,andthe
antigen binding domain (Fab), which, as the name
Biomacromolecules (in particular monoclonal implies, is solely dedicated to binding the antigen to
antibodies;mAbs) have seen a renewed interest in the the antibody. mAbs are glycoproteins that have two
pharmaceutical and biotechnology industry. The reason conserved N-glycosylation sites in the Fc domain. A variety
for this high level of interest resides in the number of ofotherchemicalandenzymaticmodi_cationscanfurther
bene_tsthesebiologicalmoleculeshaveforpatients modify a mAb including methylation, phosphorylation,
including,butnotlimitedto,higheacacyintreatingdeamidation, an oxidation, and conjugation to cytotoxic,
illness,highspeci_cityforatargetreceptororantigen, small molecule drugs (thus resulting in an antibody-drug
wide therapeutic range, and limited undesirable side conjugate; ADC), among many others.3 Therefore, there
e^ects.1 However, due to the fact that these molecules are thousands of potential variant combinations in a single
are complex and are often produced in host cell lines, mAb formulation, and some of these may elicit a lethal
bacteria, or fermentation reactors, these potential response in a patient. In addition to the above-mentioned
therapeuticshavesigni_cantheterogeneitywhichmodi_cations needs to the primary structure of the mAb,
to be evaluated and characterized using analytical modi_cationstothehigher-orderstructureofthemAb
techniques.2 may occur, such as aggregation or clipping, which can also
a^ectthesafetyandeacacyofthetherapeutic. 4
The complexity of such biomacromolecules can be
easily illustrated by examining the structure of a typical Due to the above-mentioned, inherent danger associated
mAb as depicted in Figure 54. with the possible heterogeneity of these biologics, the
Food and Drug Administration (FDA) and the European
IgG1
Medicines Agency (EMA) now require all biopharmaceutical
Va

manufacturing companies to submit a comprehensive


r
ia

n le
L)
R Va n
bl VL

io iab
(V

report detailing the complete characterization of their


i
e )

ha
L gio
(

eg r
R

ig n R
e

(C nt t C
ht

submitted drug formulation. Due to this stringent


n ta h
Ch Con ion

io ns ig

requirement, the need for e^ective, sensitive, and


on
L
Va

ai sta (CL

L)

gi
n nt )
r

ai le H)
eg
(V e R
ia

robust analytical techniques to fully characterize these


R
Fa Ch riab (V
bl
H e

eg o
) gi

R C

biomacromolecules is a key factor for the “biopharma”


H

n
Va
on
ea Fa

market to continue to thrive. This chapter will outline


vy b

n
C

H egio
on

t )

several analytical chromatographic strategies that


y
(C nt
C

an H1
st

av
R

b
ha

H Re
a

st (C
1) g

the biopharmaceutical scientist can employ to fully


in

on

characterize novel biopharmaceuticals.


i on

Hinge Region
N-Linked
Glycan N-Linked Glycan
(CH2) (CH2)
Constant Regions

Constant Regions
Heavy Chain

Heavy Chain

Fc
Size Exclusion Chromatography
Size exclusion chromatography (SEC) is a mode
of chromatography that separates molecules by
(CH3) (CH3) their size (i.e. hydrodynamic radius). This mode of
chromatography does not rely on the interaction of
the analytes with a stationary phase ligand; it is an
Figure 54. Graphic depiction of an IgG1 antibody. Note the structural entropic process meaning that it relies on the random
complexityofthedi^erentdomainsofthemAb. ‘owoftheanalytesthroughthestationaryphase
particles. For most practical purposes, this can be
mAbs are large, tetrameric immunoglobulin G (IgG) envisioned as analytes with a higher molecular weight
molecules with a molecular weight of approximately will elute earlier in the run, since these analytes
150 kDa (150,000 g/mol). These molecules form are fully or partially excluded from the pores of the
a Y-shape composed of four peptide chains: two stationary phase particles, while lower molecular weight
identical light (L) chains with a molecular weight of analytes will elute later in the run, since these analytes
approximately 25 kDa each, and two identical heavy (H) will spend more time navigating the torturous path
chains with a molecular weight of approximately 50 kDa through the particles.5
each. To form the Y-shape, these four polypeptide ThedrivingforceinSECistheexclusione^ectbased
chains associate with each other through the creation of onmolecularweightandsize.Thisexclusione^ectis
inter-andintra-chaindisul_debonds.Fromastructuraldictated by the pore diameter and geometry of the
biology perspective, a mAb can be broken down into stationary phase particle. Therefore, when selecting
two domains: the fragment crystallizable (Fc) domain, an SEC column (or any column) for a separation

50
experiment, one needs to be cognizant of the particle There have been some developments in the manu-
pore size. If the pore size is too small, the analyte(s) of facturing and application of small particle size SEC
interest will not enter the particle and will elute in the columns for biomolecule separations. These new
void volume. For SEC of smaller proteins and peptides, columnsexistas2.0mandsub-2msilicaparticles,
particles with a pore diameter of 150 Å will provide a enabling the biochromatographer to perform high
good linear separation range when creating a molecular eaciencyseparationsinhalfthetimecomparedto3.0
weight calibration curve, shown in Figure 55. Table 16 and4.0mparticles.Recentstudies(7)haveshown
details the molecular weights of the analytes used in this the advantages of using small particle SEC column
study. As seen in Figure 55, proteins with molecular technology.
weights larger than approximately 50 kDa (analytes 1–3)
In addition to recent advances in column technology,
begin to be excluded from the pores of the column,
the mobile phase is also important to consider when
resulting in premature elution, whereas analytes with
performing SEC of biomolecules. One area of focus
molecular weights less than 5 kDa (analytes 8–11)
where the composition of the mobile phase will
interact more with the stationary phase and elute later
dictate success or failure in an SEC experiment is in
in the run. To achieve the best chromatographic results
characterizing ADCs by SEC. ADCs tend to exhibit
with this column, one should operate in the linear
secondary interactions with the stationary phase due
region of the curve (analytes 5–7; molecular weights
to the added cytotoxic drug payload. These interactions
17 kDa–6 kDa).6
will result in broad peak tailing and lower sensitivity
1
of higher-order aggregates. One way to minimize
1000000
these interactions is to add an organic alcohol (i.e.
2 isopropanol, 1-butanol, 1-propanol, etc.) to the mobile
Molecular weight

100000 3 Insulin at 6 kD is phase.Theadditionofanalcoholmodi_eristhought


4
partially excluded to either mask surface silanols on the stationary phase
5
Size 6 or stabilize a particular fold of the protein, thereby
7
10000 dominates reducing the heterogeneity of the analyte and allowing
8 for the analyte to elute with a lower peak volume.8,9
9
10
1000 AnotherimportantaspectofSECisthee^ectofsalt
Adsorption
dominates
and salt type in SEC. Salt is typically used in SEC
11 to minimize electrostatic interactions between the
100 analyte and charged sites on the silica stationary
6 8 10 12 14 16 18
Retention time (min) phase. Typically, most SEC methods will use sodium
or potassium phosphate (100–200 mM, pH 7.0–7.4)
Figure 55. Molecular weight calibration curve for a series of proteins
to perform the separation, though recent research
separated by SEC. Column: Zenix® SEC-150, 30 cm x 4.6 mm I.D.,
3m;MobilePhase:0.2Msodiumphosphatedibasic,pH7.0with has revealed that potassium salts may elicit better
phosphoric acid; Flow Rate: 0.25 mL/min; Column temperature: 25 °C; resolution of dimeric species.10 One problem with
Detector:UV,215nm;Injection:0.5L;Sample:Mixtureofproteins this salt, however, is that it is not compatible with
from Table x, 1 mg/mL, 0.2 M sodium phosphate dibasic, pH 7.0 with
phosphoric acid. Adapted from reference 6. mass spectrometric (MS) detection. Volatile salts, like
ammonium formate, have been used to perform online
SEC/MS for characterizing biotherapeutics. Figure 56
Table 16. Analytes Used in Molecular Weight Calibration Curve
shows the deconvoluted MS spectral results of such
Analyte MW(Da) a study, showing that SEC/MS could be employed in
1. thyroglobulin 667000
drug-to-antibody (DAR) characterization of an ADC.11
2. IgG 150000
Average DAR 3.9
3. BSA 66400
4. ovalbumin 45000 146474
100
5. myoglobin 17000 4
6. thioredoxin 11700
2
7. insulin 5733 145135
6
8. neurotensin 1700 147812

9. vitamin B12 1350


0 8
10. angiotensin II 1000 143799 149147

11. uracil 112 0


143000 144000 145000 146000 147000 148000 149000
Mass
Figure 56. Native SEC/MS spectrum of the SigmaMAb ADC mimic.
Di^erenceinmolecularweightbetweeneachmainpeakis136Da,
corresponding to the molecular weight of two payloads. Conditions:
Column: TSKgel® SuperSW 3000, 30 cm x 2 mm I.D., 4 µm; Mobile phase:
100 mM Ammonium acetate, pH 7.0 (isocratic); Flow rate: 0.07 mL/min;
Column temperature: 35 °C; Detector: ESI-MS, 1000–8000 m/z;
Injection: 1.0 µL; Sample: ADC mimic, 100 µg/mL, 100 mM Ammonium
acetate, pH 7.0. Adapted from reference 11.

51
Hydrophobic Interaction Chromatography Figure 4 also shows the use of isopropyl alcohol in
Hydrophobic interaction chromatography (HIC) is a mode both mobile phases, essentially setting up an alcohol
of chromatography that separates analytes based on the gradient. The reason for this inclusion is the same
degree of interaction between hydrophobic moieties on as was discussed with SEC: some analytes tend to
the analyte and hydrophobic ligands on the stationary interact too strongly with the stationary phase and
phase. Under conditions of high concentrations of salt, need the extra, organic component to promote elution
the hydration layer around a protein may be disrupted from the column. It should be noted here, though, that
enough that it becomes entropically favorable for both SEC and HIC are considered native techniques,
hydrophobic regions of the protein’s surface to interface meaning that the structure and activity of the protein
with the non-polar stationary phase. This phenomenon is preserved. However, employing alcohols in the
is similar to the classical biochemical technique of mobile phases increase the risk of these analytes being
protein salting out, but in HIC’s case, the interactions denatured. One should always check the stability
are between protein-stationary phase ligand rather than of their analytes in dilute, organic solutions prior to
betweendi^erentproteinmolecules.Duetothelower incorporation in a chromatographic method (by using
molecular weight and lower propensity for folding, HIC is a technique like sodium dodecyl sulfate polyacrylamide
not often used for separating peptides. Salt selection in gel electrophoresis; SDS-PAGE). Most proteins will be
13
HICisdictatedbytheHofmeisterseries,whichclassi_esstable in solutions up to 35% organic solvent.
cations and anions in terms of their ability to disrupt the
hydration layer around a protein (chaotropic) or promote
the formation of a hydration layer (kosmotropic). Typical
salts in HIC are ammonium sulfate, potassium sulfate, Ion Exchange Chromatography
and sodium sulfate. Ion exchange chromatography (IEX) is a mode of
The biggest application area currently under investigation chromatography that separates analytes by charge.
forHICisindeterminingtheDARpro_leofanADC.The Proteins and peptides are amphoteric, meaning that they
DARpro_leisonecriticalqualityattribute(CQA)that have both acidic and basic functionalities. The acidic
a biopharmaceutical company needs to determine for portions of a protein include aspartic acid, glutamic
approval by the FDA or EMA. An overly conjugated ADC acid,cysteine,tyrosine,andthecarboxylate
- onthe
can kill both healthy and carcinogenic cells, whereas an C-terminus. The basic portions of a protein include
under-conjugatedADCmaynotbeeaectiveinkilling arginine,histidine,lysine,andtheamine - onthe
carcinogenic cells. In cysteine-linked ADCs, where the N-terminus. Charge variants of a biotherapeutic, another
linker and cytotoxic payload is conjugated through the CQAthatregulatorybodiesrequiremanufacturers
sulfhydryl moiety of a cysteine amino acid, separation to monitor, can be detected and resolved by IEX.
byHICleadstoapro_leofpeakscorrespondingto,2 0 These charge variants can arise from mistranslation
4, 6, and 8 drugs attached to the antibody (Figure 57). of messenger RNA (mRNA) transcripts and/or post-
The keen reader will note the presence of a peak between translational modi_cationssuchasdeamidation,
peaks 2 and 3; this corresponds to a DAR 3 species. It oxidation, or glycosylation, among others.14
is possible that not being able to detect and quantify When choosing a column for an IEX experiment, one
these odd-numbered DAR species could result in an needs to be aware of the isoelectric point (pI) of the
underestimation of the average DAR for an ADC by as native state of the protein. For example, most mAbs
much as 3%.12 have a pI of ~7.4. If the pH of the mobile phase is
lower than 7.4, the mAb will be positively charged and
Elution Order: 3 bind to a cation exchange column. If the pH of the
1. DAR 0 (nativ mAb)
2. DAR 2 mobile phase is above 7.4, the mAb will be negatively
3. DAR 4 charged and bind to an anion exchange column. In
4. DAR 6 addition to cation versus anion exchangers, these can
5. DAR 8
be broken down into weak and strong exchangers
2 based on the pKa for the exchanger.
4 Therearetwomethodsforelutinganalyteso^ofan
5 IEX column: using a salt gradient and using a pH
gradient. The salt gradient approach employs a linear
gradient of a salt (i.e. sodium chloride, potassium
0 10 20 30 40 50 chloride, etc.) to essentially compete with the analyte
min binding to the column. Figure 58 shows a separation
ofdi^erentmAbchargevariants,fromaseriesof
Figure 57. HIC/UV analysis of native SigmaMAb ADC mimic. Conditions:
Column: TSKgel® Butyl-NPR, 10 cm x 4.6 mm I.D., 2.5 µm; Mobile phase: proprietary mAbs, by IEX using a salt gradient.15
[A] 50 mM Potassium phosphate, 1.5 M Ammonium sulfate, pH 7.0 plus
5% (v/v) Isopropyl alcohol, [B] 50 mM Potassium phosphate, pH 7.0
plus 20% (v/v) Isopropyl alcohol; Gradient: 0% B to 100% B in 50 min;
Flow rate: 1.0 mL/min; Column temp.: 35 °C; Detector: UV, 215 nm;
Injection: 5.0 µL; Sample: ADC mimic, 100 µg/mL, 50 mM Potassium
phosphate, pH 7.0. Adapted from Reference 12.

52
40 To determine the pattern of glycosylation on a potential
biotherapeutic,generallyone_rstreleasestheglycans
from the protein using an enzyme such as PNGase F.
30
Afterwards, the released glycans are labeled with a
Intensity (mV)

‘uorescenttaglikeaminobenzamide
-2 orprocainamide.
20 The sample is then subjected to chromatographic
analysis with either ‘uorescence detection or MS
detection. Figure 59 shows chromatographic results of
10 separating a mixture of N-linked glycans released from
human IgG. Baseline resolutions for all of the individual
0
glycans were observed, except for the positional
isomersG1FandG1F,The
. identitiesofthesedi^erent
0 10 20 30 40
Retention time (min)
glycanswerecon_rmedbyLC/MS.
Peak Glycan Structure Peak Glycan Structure
Figure 58. Analysis of mAb charge variants by weak cation exchange 1 G0 9 G2FB
chromatography. Conditions: Column: TSKgel® CM-STAT, 10 cm x 4.6 mm 2 2 G0F 10 G1FSI
I.D.,7m;MobilePhase:[A]20mM2-(N-morpholino)ethanesulfonic 3 G0FB 11 A1
acid(MES)bu^er,pH6.0;[B]20mMMESbu^er,pH6.0+5M 4 G1F 12 A1F
sodium chloride; Gradient: 0% B to 30% B in 15 min; 30% B to 100% 5 G1F’ 13 A1FB
4
B in 2 min; Flow Rate: 1.0 mL/min; Detector: UV, 280 nm; Column
6 G1FB 14 A2
Temperature:25°C;Injection:20L;Sample:mAbsAE,1g/L,
7 G2 15 A2F
20mMMESbu^er,pH6.0Adaptedfromreference15.
8 G2F 16 A2FB
8
5
The pH gradient approach employs a complex bu^er
12
system that gradually changes the pH of the mobile phase 2-AB labeling
3
6
15
resulting in concomitant changes to the ionic state of the agent
1 7
10 16
9 11 13 14
analytes and stationary phase ligands, which leads to
elution of the analytes. There are negatives to each of 0 10 20
thesetechniques:thesaltgradientisnotase^ectiveat Min
resolving similarly charged variants than a pH gradient Figure 59. Analysis of N-linked glycans released from human IgG.
methodwhereasapHgradientrequiresacomplexbu^er Conditions:Column:BIOshell=Glycan,15cmx2.1mmI.D.,2.7m;
system that may cause issues with method reproducibility. Mobile Phase: [A] 100 mM ammonium formate, pH 4.5; [B] Acetonitrile;
Gradient: 74% B to 71% B in 15 min; 71% B to 55% B in 10 min; 55% B
However,somevendorshavemadeavailableo^the-shelf to 45% B in 1 min; Flow Rate: 0.4 mL/min; Column Temperature: 50 °C;
bu^ersolutionsforpHgradientmethodsforresolving Detector:FLR,260nmexcitation,430nmemission;Injection:3.0L;
protein charge variants. Sample:Released,AB 2- labeledglycansfromhumanIgG,10g/mL,
30:70 100 mM ammonium formate, pH 4.5: acetonitrile

Hydrophilic Interaction Liquid Affinity Chromatography


Chromatography Aanitychromatographyisamodeofchromatography
Hydrophilic interaction liquid chromatography (HILIC) is thatreliesonaspeci_cinteractionbetweentheanaly
a mode of normal phase chromatography in which the of interest and the stationary phase ligand. Ideally, no
mobile phase is relatively non-polar and the stationary other component of the sample would interact with the
phase is relatively polar. In HILIC, the strong solvent is ligand, thus only the analyte of interest interfaces with
water(oralowconcentrationbu^er)andtheanalytes the stationary phase. Afterwards, a second solution is
partition between an aqueous-enriched layer adsorbed passed through the column that breaks this interaction,
on the stationary phase and the bulk mobile phase. eluting the analyte.
Retention of analytes is mostly due to the amount of Protein A chromatography is the most common
water adsorbed to the particle and the ionic strength of form of aanity chromatography employed in the
thebu^erusedinthemobilephase.Analytesareeluted biopharmaceutical industry. Protein A is a 42 kDa surface
o^thecolumnbygraduallyincreasingtheamountof protein found in the cell wall of Staphylococcus aureus.
water(bu^er)inthemobilephase. Thisproteinbindsspeci_callytotheheavychaininthe
The key application area in the biopharmaceutical industry Fc region of IgGs, making this an ideal mechanism to
for HILIC is in glycan analysis, though recently there separate IgGs from other components of a sample. Most
have been some publications focusing on the use of new, Protein A columns are manufactured by immobilizing the
wide pore HILIC columns for glycoprotein separations. 16,17 protein on a porous, organic particle. However, monolithic
The pattern of glycosylation is another CQA that format for Protein A chromatography has been produced,
biopharmaceutical manufacturers need to determine allowing for high sample throughput, at various ‘ow
accordingtoregulatoryagencies.Glycosylationcana^ect rates,withoutsacri_cingeaciency. Figure 60 shows
a protein’s folding ability as well as its stability. More the puri_cation of IgG, at di^erent ‘owrates,usinga
importantly,thepatternofglycosylationcana^ectthe Protein A monolith.
antibody’s ability to bind antigens and promote antibody-
dependent cellular cytotoxicity (ADCC) and complement
dependent cytotoxicity (CDC).

53
Absorbance at 280 nm

UV, 215 nm (mAU)


Green = SPP, 1000 Å C4
Yellow = SPP, 400 Å C4
Purple = FPP, 300 Å C4
5 mL/min
90
4 mL/min 70
50
3 mL/min
30
2 mL/min 10
1 mL/min -10
0 2 4 6 8 10 12 14
0 0.5 1 1.5 2 2.5 3 3.5 4
Time (min) Retention time (min)
Figure 60.Puri_cationofIgGusingaProteinAmonolithatdi^erent Figure 61.Middleup/downanalysisofIn‘iximab.LandHrepresent
‘owrates.Conditions:Column:Chromolith ® Protein A, 25 cm x 4.6 mm
light and heavy chain, respectively. Conditions: Column: As indicated,
I.D., Mobile Phase: [A] 100 mM sodium phosphate, pH 7.4; [B] 100 mM 10 cm x 2.1 mm I.D., 1.7 µm (FPP), 3.4 µm (SPP, 400 Å), 2.7 µm (SPP,
sodiumphosphate,pH2.5;Gradient(for2mL/min‘owrate):0%Bfor 1000 Å); Mobile Phase: [A] 70:30 water (0.1% (v/v) TFA): acetonitrile
0.25 min; increase to 100% B in 0.1 min; hold at 100% B for 1 min, (0.1% (v/v) TFA); [B] 50:50 water (0.1% (v/v) TFA): acetonitrile
Flow Rate: 2 mL/min; Column Temperature: 25 °C, Detector: UV, 280 nm, (0.1% (v/v) TFA); Gradient: 20% B to 50% B in 10 min; 50% B to
Injection:L,0 Sample:
1 IgG,g/1 mM
L,0 1 sodiumphosphate,pH. 4 7 100% B in 5 min; Flow Rate: 0.2 mL/min; Column Temperature: 75 °C;
Figure courtesy of Dr. Egidijus Machtejevas. Detector:UV,215nm;Injection:5.0µL;Sample:Reducedin‘iximab,
100 µg/mL, water (0.05% TFA)

Reversed-Phase Chromatography 60000000


DFA Method
Reversed-phase chromatography (RPC) is a mode of
50000000 1
chromatography that separates analytes based primarily
UV Intensity (mV)

4
on hydrophobicity. Unlike HIC, RPC employs a water/ 40000000
2
5
organic mixture for the mobile phase. There are several 3
1. Insulin-Glargine
parameters that can be tuned in an RPC experiment to get 30000000 2. Insulin-Bovine
3. Insulin-Asp
satisfactory peak shape and resolution; these have been 4. Insulin-LisPro
5. Insulin-Human
reviewed in several recent publications.18,19 20000000
6. Insulin-Porcine

Several wide pore RPC columns have been launched by 10000000

column vendors over the past decade. These columns


tend to fall into one of two categories: fully porous 0
3 3.5 4 4.5 5 5.5 6 6.5 7
particle(FPP)packedcolumnsandsuper_ciallyporous
Retention time (min)
particle (SPP) packed columns. SPP packed columns
tendtoprovidemoreeacientseparationsduetobetter 160000000

packingeacienciesofSPPcolumnsaswellasreduced Ammonium Formate Method


140000000
longitudinaldi^usionduetothepresenceofasolid
UV Intensity (mV)

silica core within the particle. Figure 61 showcases 120000000

the advantages of using SPP packed columns over 100000000

FPP packed columns in performing“middle up/down” 80000000


analysisofIn‘iximab,atherapeuticmAb.Notethe
60000000
improved resolution and peak shape with the 1000 Å
SPP column. 40000000

20000000
Another key aspect in RPC of biomacromolecules is
the use of an ion pair reagent in the mobile phase. 0
3 3.5 4 4.5 5 5.5 6 6.5 7
Generally,tri‘uoroaceticacid(TFA)hasbeenused
as a good ion pair reagent as it masks interactions Retention time (min)
between the analytes and free, active silanols on the Figure 62. Separation of insulin variants by LC/MS using di^erent
stationary phase. However, this ion pair reagent can mobilephasemodi_ers.Conditions:Column:BIOshell=A160Peptide
cause severe ion suppression when coupled to MS C18, 15 cm x 2.1 mm I.D., 2.7 µm; Mobile Phase: [A] 75:25 10 mM
ammonium formate, pH 2.6 with formic acid or water (0.1% (v/v)
detection.Therefore,alternatives,likedi‘uoroacetic DFA): acetonitrile; [B] 50:50 10 mM ammonium formate, pH 2.6
acid (DFA) and ammonium formate, should be used for with formic acid or water (0.1% (v/v) DFA): acetonitrile (77:23 A:B);
MS analyses as they tend to provide good ion pairing Flow Rate: 0.2 mL/min; Column Temperature: 75 °C; Detector: MSD,
ESI-(+), TIC 100–3000 m/z; Injection: 0.5 µL; Sample: Mixture of six
capacity while minimizing ion suppression. Figure 62
insulin variants, 100 µg/mL, 10 mM ammonium formate, pH 2.6 with
shows a comparative study of these two ion pairing formic acid. Adapted from Reference 20.
reagents in separating a series of insulin variants. Note
the ~2.7-fold increase in sensitivity using ammonium
formate.20

54
References
1. A. J. Chirino, A. Mire-Sluis, Nat. Biotechnol 22, 12. B. Bobaly, G. M. Randazzo, S. Rudaz, D. Guillarme,
1383 – 1391 (2004). S. Fekete, J. Chromatogr. A 1481, 82 – 91, (2017).
2. K. Sandra, I. Vandenheede, P. Sandra, J. 13. C. N. Pace, S. Trevino, E. Prabhakaran, J. M.
Chromatogr. A 1335, 81 – 103 (2014). Scholtz, Phil. Trans. R. Soc. Lond. B 359, 1225 –
1235 (2004).
3. H. Liu, G. Ponniah, H. M. Zhang, C. Nowak, A. Neill,
N. Gonzalez-Lopez, R. Patel, G. Cheng, A. Z. Kita, 14. E. Wagner-Rousset, S. Fekete, L. Morel-Chevillet,
B. Andrien, mAbs 6, 1145 – 1154 (2014). O. Colas, N. Corvaia, S. Cianferani, D. Guillarme, A.
Beck, J. Chromatogr. A 1498, 147 – 154 (2017).
4. A. Beck, E. Wagner-Rousset, D. Ayoub, A. Van
Dorsselaer, S. Sanglier-Cianferani, Anal. Chem. 85, 15. R. Romling, Reporter, 29.3, 20 – 21 (2011).
715 – 736 (2013).
16. V. D’Atri, S. Fekete, A. Beck, M. Lauber, D.
5. J. H. Knox, High Performance Liquid Guillarme, Anal. Chem. 89, 2086 – 2092 (2017).
Chromatography, Edinburgh University Press,
17. A. Periat, S. Fekete, A. Cusumano, J. Veuthey, A.
1978.
Beck, M. Lauber, D. Guillarme, J. Chromatogr. A
6. R. A. Henry, S. Schuster, “Impact of Pore Exclusion 1448, 81 – 92 (2016).
on Reversed-Phase HPLC Column Performance,”
18. A. Beck, S. Cianferani, A. Van Dorsselaer, Anal.
presented at the Eastern Analytical Symposium
Chem. 84, 4637 – 4646 (2012).
(EAS 2016), Somerset, New Jersey, 2016.
19. A. Beck, G. Terral, F. Debaene, E. Wagner-Rousset,
7. H. K. Brandes, S. Shollenberger, C. E. Muraco,
J. Marcoux, M. C. Janin-Bussat, O. Colas, A. Van
Reporter 34.3, 33 – 34 (2016).
Dorsselaer, S. Cianferani, Expert Rev. Proteomics
8. X. M. Lu, K. Benedek, B. L. Karger, J. Chromatogr. 13, 1 – 26 (2016).
A 359, 19 – 29 (1986).
20. C. E. Muraco, S. Shollenberger, H. K. Brandes,
9. S. Fekete, S. Rudaz, J. Veuthey, D. Guillarme, J. “Reversed-Phase HPLC Analysis of Insulin Variants
Sep. Sci. 35, 3113 – 3123 (2012). and Analogs by UV and Mass Spectral Detection,”
presented at the American Association of
10. A. Goyon, A. Beck, J. Veuthey, D. Guillarme, S.
Pharmaceutical Scientists National Biotechnology
Fekete, J. Pharm. Biomed. Anal. 144, 242 – 251
Conference (AAPS NBC 2016), Boston,
(2017).
Massachusetts, 2016.
11. C. E. Muraco, K. Ray, G. Oden, D. S. Bell, “Using
All the Tools in the Toolbox: Characterization of A
Novel Antibody-Drug Conjugate Mimic by Several
Modes of Chromatography,” presented at the
International Symposium on the Separation of
Proteins, Peptides, and Polynucleotides (ISPPP
2017), Philadelphia, Pennsylvania, 2017.

55
5. Troubleshooting Common HPLC Issues
Introduction Problem No. 2: No Flow
Although HPLC method development has been improved Normal
by advances in column technology and instrumentation,
problems still arise. In this chapter, a systematic means
of isolating, identifying, and correcting many typical

Detector Responce
problems encountered in the practice of HPLC will be
given. The important segments of an HPLC system are
the same, whether using a modular system or “cutting-
edge0UHPLCinstruments.Problemsa^ectingoverall
system performance can arise in each component.
Some common problems are discussed here.

Problem No. 1: No Peaks/Very Small Peaks


Time
Normal
Detector Responce

Problem

Responce
Detector
Time

Time
Probable Causes:
1. Pumpo^.

Problem 2. Flow interrupted/obstructed.


Responce
Detector

3. Leakinthe‘owpath.
4. Air trapped in pump head (revealed by pressure
Time ‘uctuations).

Problem Possible Solutions:


Responce
Detector

1. Start pump.
2. Check mobile phase level in reservoir(s). Check
Time ‘owthroughoutsystem.Examinesampleloopfor
obstruction or air lock. Make sure mobile phase
components are miscible and mobile phase is
Probable Causes: properly degassed.
1. Detectoro^ornotsuaciently/warmedup0.
3. Checksystemforloose_ttings.Checkpumpfor
2. Poor/no connection between instrument and computer. leaks, salt buildup, or unusual noises. Change pump
3. Nomobilephase‘ow. seals if necessary.
4. No sample/deteriorated sample/wrong sample.
4. Disconnect tubing at guard column (if present) or
5. Settings incorrect on detector. analyticalcolumninlet.Checkfor‘ow.Purgepump
athigh‘owrate(e.g.mL/
510, min)prime
, system
Possible Solutions: if necessary (prime each pump head separately). If
1. Turndetectoronorallowsuacienttimefordetector system has check valve, loosen valve to allow air to
to “warm up”. escape.Ifproblempersists,‘ushsystemwith10%
2. Check connectivity between instrument and computer. methanol or isopropanol. If problem still persists,
3. See “No Flow” (Problem No. 2). contact system manufacturer.
4. Besureautosamplervialshavesuacientliquid
and no air bubbles in the sample. Evaluate system
performancewithfreshstandardtocon_rmsample
as source of problem.
5. Check detector status/settings. Auto-zero if necessary.

56
Problem No. 3: No Pressure/Pressure eliminating system components, starting with
Lower Than Usual detector,thenin-line_lter,andworkingbackto
Probable Causes: pump.Replace_lterinpumpifpresent.
1. Leak. 2. Remove guard column (if present) and check
pressure. Replace guard column if necessary. If
2. Mobilephase‘owinterrupted/obstructed. analyticalcolumnisobstructed,reverseand‘ush
3. Air trapped in pump head (revealed by pressure the column, while disconnected from the detector.
‘uctuations). If problem persists, column may be clogged with
strongly retained contaminants. Use appropriate
4. Leakatcolumninletend_tting. restoration procedure (Appendix). If problem still
5. Air trapped elsewhere in system. persists, replace column.

6. Worn pump seal causing leaks around pump head.


7. Faulty check valve. Problem No. 5: Variable Retention Times
8. Faulty pump seals

Detector Responce
Normal
Possible Solutions:
1. Checksystemforloose_ttings.Checkpumpfor
leaks, salt buildup, or unusual noises. Change pump
seals if necessary.
2. Check mobile phase level in reservoir(s). Check
‘owthroughoutsystem.Examinesampleloopfor
obstruction or air lock. Make sure mobile phase Time
components are miscible and mobile phase is
properly degassed. Detector Responce
Problem
3. Disconnect tubing at guard column (if present) or
analyticalcolumninlet.Checkfor‘ow.Purgepump
athigh‘owrate(e.g.,10mL/min),primesystem
if necessary (prime each pump head separately). If
system has check valve, loosen valve to allow air to
escape.
4. Reconnect column and pump solvent at double the Time
‘owrate.Ifpressureisstilllow,checkforleaksat Problem
inlet_ttingorcolumnend_tting.
Responce
Detector

5. Disconnect guard and analytical column and purge


system. Reconnect column(s). If problem persists,
‘ushsystemwith10%methanolorisopropanol.
6. Replace seal. If problem persists, replace piston and
Time
seal.
7. Rebuild or replace valve.
Probable Causes:
8. Replace seals.
1. Leak.
2. Change in mobile phase composition (small changes
Problem No. 4: Pressure Higher Than Usual can lead to large changes in retention times).
Probable Causes:
3. Air trapped in pump. (Retention times increase and
1. Probleminpump,injector,in-line_lter,ortubing. decrease at random times.)
2. Obstructed guard column or analytical column. 4. Columntemperature‘uctuations(especiallyeviden
in ion exchange chromatography).
Possible Solutions: 5. Column overloading. (Retention times usually
1. Remove guard column and analytical column from decrease as mass of solute injected on column
system. Replace with unions and 0.010'' I.D. or exceeds column capacity.)
larger tubing to reconnect injector to detector. Run
pump at 2–5 mL/min. If pressure is minimal, see 6. Sample solvent incompatible with mobile phase.
Solution 2. If not, isolate cause by systematically 7. Column problem. (Not a common cause of erratic
retention. As a column ages, retention times
gradually decrease.)

57
Possible Solutions: Problem No. 7: Split Peaks
1. Checksystemforloose_ttings.Checkpumpfor
leaks, salt buildup, or unusual noises. Change pump
Normal

Detector Responce
seals if necessary.
2. Check make-up of mobile phase. If mobile phase
is machine mixed using proportioning values, hand
mix and supply from one reservoir.
3. Purge air from pump head or check valves. Change
pump seals if necessary. Be sure mobile phase is
degassed. Time
4. Use a reliable column oven. Note that higher
columntemperaturesincreasecolumneaciency. Problem
For optimum results, heat eluant before introducing
it onto column.

Detector Responce
5. Injectsmallervolume(e.g.,1.0Lvs.10.L)
or inject the same volume after 1:10 or 1:100
dilutions of sample.
6. Adjust solvent. Whenever possible, inject samples
in starting mobile phase.
7. Substitutenewcolumnofsametypetocon_rm
column as cause. Discard old column if restoration
procedures fail.
Time

Problem No. 6: Loss of Resolution


Probable Causes:
Normal 1. Contamination on guard or analytical column inlet.
2. Partially blocked frit.
Responce
Detector

3. Small (uneven) void at column inlet.


4. Sample solvent incompatible with mobile phase

Time Possible Solutions:


1. Remove guard column (if present) and attempt
Problem analysis. Replace guard column if necessary. If
analyticalcolumnisobstructed,reverseand‘ush.
Responce
Detector

If problem persists, column may be clogged with


strongly retained contaminants. Use appropriate
restoration procedure (Appendix). If problem still
persists, replace column.
Time 2. Adjust solvent. Whenever possible, inject samples
in starting mobile phase.

Probable Causes:
1. Mobile phase contaminated/evaporated (causing
retention times and/or selectivity to change).
2. Obstructed guard or analytical column.

Possible Solutions:
1. Prepare fresh mobile phase.
2. Remove guard column (if present) and attempt
analysis. Replace guard column if necessary. If
analyticalcolumnisobstructed,reverseand‘ush.
If problem persists, column may be clogged with
strongly retained contaminants. Use appropriate
restoration procedure (Appendix). If problem still
persists, replace column.

58
Problem No. 8: Peaks Tail on Initial and Problem No. 9: Tailing Peaks
Later Injections
Normal
Normal

Detector Responce
Detector Responce

Time
Time
Problem

Detector Responce
Problem
Responce
Detector

Time
Time

Probable Causes:
Probable Causes: 1. Guard or analytical column contaminated/worn out.
1. Sample reacting with active sites. 2. Mobile phase contaminated/evaporated.
2. Wrong mobile phase pH. 3. Interfering components in sample.
3. Wrong column type.
4. Small (uneven) void at column inlet. Possible Solutions:
1. Remove guard column (if present) and attempt
5. Wrong injection solvent.
analysis. Replace guard column if necessary.
If analytical column is source of problem, use
Possible Solutions: appropriate restoration procedure (Appendix). If
1. First check column performance with standard problem persists, replace column.
column test mix. If results for test mix are good, 2. Check make-up of mobile phase.
add ion pairing reagent or competing base or acid
modi_er. 3. Check column performance with a column test mix.

2. Adjust pH. For basic compounds, lower pH usually


provides more symmetric peaks.
3. Try another column type.
4. See Split Peaks (Problem 7).
5. Peaks can tail when sample is injected in stronger
solvent than mobile phase. Dissolve sample in
mobile phase.

59
Problem No. 10: Fronting Peaks Problem No. 11: Rounded Peaks

Normal Normal

Detector Responce
Detector Responce

Time
Time
Problem
Problem

Responce
Detector
Detector Responce

Time

Probable Causes:
1. Detector operating outside linear dynamic range.
Time
2. Column overloaded.
3. Sample-column interaction.
Probable Causes:
4. Detector time constants are set too high.
1. Column overloaded.
2. Sample solvent incompatible with mobile phase. Possible Solutions:
3. Shoulder or gradual baseline rise before a main 1. Reduce sample volume and/or concentration.
peak may be another sample component. 2. Injectsmallervolume(e.g.,1.0Lvs.10.L)or
1:10 or 1:100 dilution of sample.
Possible Solutions:
3. Changebu^erstrength,pH,ormobilephase
1. Injectsmallervolume(e.g.,1.0Lvs.10.L). composition. If necessary, raise column
Dilute the sample 1:10 or 1:100 fold in case of temperature or change column type. (Analysis of
mass overload. solute structure may help predict interaction.)
2. Adjust solvent. Whenever possible, inject samples 4. Reduce settings to lowest values or values at which
in mobile phase. Flush polar bonded phase column no further improvements are seen.
with 50 column volumes HPLC grade ethyl acetate
at23timesthestandard‘owrate,thenwith
intermediate polarity solvent prior to analysis.
3. Increaseeaciencyorchangeselectivityofsystem
to improve resolution. Try another column type if
necessary (e.g., switch from nonpolar C18 to polar
cyano phase).

60
Problem No. 12: Baseline Drift Problem No. 13: Baseline Noise (regular)
Normal

Responce
Detector
Normal

Responce
Detector
Time
Time
Problem
Responce
Detector

Problem

Responce
Detector
Time
Time

Probable Causes:
1. Columntemperature‘uctuation.Evensmallchanges Probable Causes:
cancausecyclicbaselineriseandfall.Mostoftena^ects 1. Air in mobile phase, detector cell, or pump.
refractive index and conductivity detectors, UV detectors
at high sensitivity or in indirect photometric mode. 2. Pump pulsations.
2. Nonhomogeneous mobile phase. Drift is usually to 3. Incomplete mobile phase mixing.
higher absorbance, rather than cyclic pattern from
4. Temperaturee^ect(columnathightemperature,
temperature‘uctuation.
detector unheated).
3. Contaminant or air buildup in detector cell.
4. Plugged outlet line after detector. High pressure 5. Leak.
cracks cell window, producing noisy baseline.
5. Mobilephasemixingproblemorchangein‘owrate. Possible Solutions:
6. Slow column equilibration, especially when 1. Degas mobile phase. Flush system to remove air
changing mobile phase. from detector cell or pump.
7. Mobile phase contaminated, deteriorated, or not 2. Incorporate pulse damper into system.
prepared from high quality chemicals.
8. Strongly retained materials in sample (high 3. Mix mobile phase by hand or use less viscous solvent.
retention factor (k)) can elute as very broad 4. Reducedi^erentialoraddpost-columncooler.
peaks and appear to be a rising baseline. Gradient
analyses can aggravate problem. 5. Checksystemforloose_ttings.Checkpumpfor
9. Detector (UV) not set at absorbance maximum but leaks, salt buildup, or unusual noises. Change pump
at slope of curve. seals if necessary.

Possible Solutions: Problem No. 14: Baseline Noise (irregular)


1. Control column and mobile phase temperature; use
post-column cooler before detector. Normal
Responce
Detector

2. Use HPLC grade solvents, high purity salts, and


additives. Degas mobile phase before use if
instrument is not equipped with automatic degasser. Time
3. Flush cell with methanol or other strong solvent. If
necessary, clean cell with 1 N nitric acid (never with Problem
hydrochloric acid, and never use nitric acid with
Responce
Detector

PEEKtubingor_ttings).
4. Unplug or replace line. Refer to detector manual to
replace window.
5. Correctcomposition/‘owrate.Toavoidproblem, Time
routinelymonitorcompositionand‘owrate.
6. Flush column with intermediate strength solvent,
run 10–20 column volumes of new mobile phase
through column before analysis. Probable Causes:
7. Check make-up of mobile phase. 1. Leak.
8. Useguardcolumn.Ifnecessary,‘ushcolumnwith 2. Mobile phase contaminated, deteriorated, or
strong solvent between injections or periodically prepared from low quality materials.
during analysis.
9. Change wavelength to UV absorbance maximum.
61
3. Air trapped in system. 5. Extra-columne^ects:
4. Air bubbles in detector. a. Column overloaded.
5. Detector cell contaminated. (Even small amounts of b. Detector response time or cell volume too large.
contaminants can cause noise.)
c. Tubing between column and detector too long or
6. Weak detector UV lamp. I.D. too large.
7. Column leaking silica or bonding. d. Detector response time too high.
6. Bu^erconcentrationtoolow.
Possible Solutions:
7. Guard column contaminated/worn out.
1. Checksystemforloose_ttings.Checkpumpfor
leaks, salt buildup, or unusual noises. Change pump 8. Column contaminated/worn out.
seals if necessary.
9. Void at column inlet.
2. Check make-up of mobile phase.
10. Peak represents two or more poorly resolved
3. Select the highest solvent grade appropriate for compounds.
your application. Ensure the mobile phase is freshly
11. Column temperature too low.
prepared.
4. Flush system with strong solvent. Possible Solutions:
5. Purge detector. Install backpressure regulator after 1. Prepare new mobile phase.
detector. Check the instrument manual, particularly
for RI detectors (excessive backpressure can cause 2. Adjust‘owrate.
the‘owcelltocrack). 3. Checksystemforloose_ttings.Checkpumpfor
6. Clean cell. leaks, salt buildup, and unusual noises. Change
pump seals if necessary.
7. Replace UV lamp.
4. Adjust settings.
8. Replace column and clean system.
5. ToresolveExtraColumnE^ects:
a. Injectsmallervolume(e.g.,1.0Lvs.10.L)
Problem No. 15: Broad Peaks or 1:10 and 1:100 dilutions of sample.

Normal b. Reduce response time or use smaller cell.


c. Use as short a piece of 0.007–0.010" I.D. tubing
as practical.
Detector Responce

d. Reduce response time.


6. Increase concentration.
7. Replace guard column.
8. Replace column with new one of same type. If new
columndoesnotprovidenarrowpeaks,‘ushold
column (Appendix), then retest.
Time
9. Replace column.
Problem 10. Change column type to improve separation.
11. Increase temperature. Do not exceed 75 °C unless
Responce
Detector

higher temperatures are acceptable to column


manufacturer.

Time

Probable Causes:
1. Mobile phase composition changed.
2. Mobilephase‘owratetoolow.
3. Leak (especially between column and detector).
4. Detector settings incorrect.

62
Problem No. 16: Change in Peak Height Problem No. 17: Change in Selectivity
(one or more peaks)
Normal
Normal

Detector Responce
Detector Responce

Time

Problem Time
Detector Responce

Problem

Detector Responce
Time

Probable Causes:
1. One or more sample components deteriorated or
column activity changed. Time

2. Leak, especially between injection port and column


inlet (retention would also change). Probable Causes:
3. Inconsistent sample volume. 1. Increase or decrease solvent ionic strength, pH,
oradditiveconcentration(especiallya^ectsionic
4. Detector setting changed. solutes).
5. Weak detector UV lamp. 2. Columnchanged,newcolumnhasdi^erent
6. Contamination in detector cell. selectivity than old column.
3. Sample injected in incorrect solvent or excessive
Possible Solutions: amount(10.2L)ofstrongsolvent.
1. Usefreshsampleorstandardtocon_rmsample 4. Column temperature change.
as source of problem. If some or all peaks are
still smaller than expected, replace column. If
new column improves analysis, try to restore Possible Solutions:
the old column, following appropriate procedure 1. Check make-up of mobile phase.
(Appendix). If performance does not improve,
2. Con_rmidentityofcolumnpacking.For
discard old column.
reproducible analyses, use same column type.
2. Checksystemforloose_ttings.Checkpumpfor Establish whether change took place gradually. If
leaks, salt buildup, or unusual noises. Change pump so, bonded phase may have been stripped. Column
seals if necessary. activity may have changed, or column may be
contaminated.
3. Besuresamplesareconsistent.For_xedvolume
sample loop, use 2–3 times loop volume to ensure 3. Adjust solvent. Whenever possible, inject sample in
loopiscompletely_lled.Besureautosamplervials mobile phase.
containsuacientsampleandnoairbubbles.Check
4. Adjust temperature. Use column oven to maintain
syringe-type injectors for air. In systems with wash
constant temperature.
or‘ushingsteps,besurewashsolutiondoesnot
precipitate sample components.
4. Check settings.
5. Replace lamp.
6. Clean cell.

63
Problem No. 18: Negative Peak(s) Problem No. 19: Ghost Peak

Normal Previous Sample


Detector Responce

Detector Responce
Time Time

Problem Normal

Responce
Detector
Detector Responce

Time
(solvent injected after sample)

Problem

Responce
Detector
Time
Time
(solvent injected after sample)
Probable Causes:
1. Refractive index of solute less than that of mobile
phase (RI detector).
Probable Causes:
2. Samplesolventandmobilephasedi^ergreatlyin
1. Contamination in injector or column.
composition (vacancy peaks).
2. Late eluting peak (usually broad) present in sample.
3. Mobile phase more absorptive than sample
components to UV wavelength.
Possible Solutions:
Possible Solutions: 1. Flush injector between analyses (a good routine
practice). If necessary, run strong solvent through
1. Use mobile phase with lower refractive index.
column to remove late eluting compounds. Include
2. Adjust or change sample solvent. Dilute sample in _nalwashstepingradientanalyses,toremove
mobile phase whenever possible. strongly retained compounds.
3. Change UV wavelength or use mobile phase that 2. a. Check sample preparation.
does not adsorb at chosen wavelength.
b. Include (step) gradient to quickly elute component.

64
Common Column Restoration Part 3: Polar-Bonded Reversed Phase
Columns (Amino, Cyano, Diol, Chiral)
Procedures
To restore these types of columns, the following
*Note: The below volumes are based on optimized procedure should be employed:
procedures for columns that are 4.6 mm I.D. For
di^erentI.D.columns,pleaseconvertvolumesbytaking 1. For a column used in the reversed phase mode
the ratio of the square of the new I.D. to (4.6 mm I.D.)2 (e.g.,organicsolvent/aqueousbu^ermobile
and multiplying this conversion factor to the volume. phase), follow the same cleanup procedure as for
Please check the column Care and Use Guide to ensure silica-based reversed phase columns. For a column
compatibility with these solvents prior to employing these used with nonaqueous mobile phases, use the
strategies to restore the column. following scheme:
2. Flush with the following:
Part 1: Bare Silica Columns
   a. 50 mL chloroform
To restore a bare silica column, the following procedure
should be used:    b. 50 mL methanol
1. 50 mL hexane    c. 50 mL acetonitrile
2. 50 mL methylene chloride    d. 25 mL methylene chloride
3. 50 mL 2-propanol    e. 25 mL methanol
4. 50 mL methanol    f. 25 mL mobile phase
5. 25 mL methylene chloride
6. 25 mL mobile phase Part 4: Silica-Based Ion Exchange Columns
To restore these ion exchange columns, the following
procedure should be employed:
Part 2a: Silica-Based Reversed Phase Columns
1. 50 mL hot (40–60 °C) distilled water
Analyzing Water-Soluble Compounds
To restore a reversed-phase column that was used 2. 50 mL methanol
in analyzing water-soluble compounds, the following 3. 50 mL acetonitrile
procedure should be employed:
4. 25 mL methylene chloride
1. Flush with warm (60 °C) ultrapure water
5. 25 mL methanol
2. 50 mL methanol
6. 25 mL mobile phase
3. 50 mL acetonitrile
4. 25 mL methanol
5. 25 mL mobile phase

Part 2b: Silica-Based Reversed Phase Columns


Analyzing Water-Insoluble Compounds
To restore a reversed-phase column that was used in
analyzing water-insoluble compounds, the following
procedure should be employed:
1. 50 mL 2-propanol
2. 50 mL methylene chloride
3. 50 mL hexane
4. 25 mL isopropanol
5. 25 mL mobile phase

65
6. R
 eference Materials in LC-MS/MS
Applications: Quality Grades &
Selection Considerations
Choosing the correct reference Mass—
Kilogram
material quality grade for Luminous
your needs Intensity—
Candela kg Length—
Metre/Meter

m
cd
Who uses reference materials?
Reference materials are a critical component of the
analyticaltestingwork‘ow.Throughcalibrationof

mol
measurement systems, validation of methods, and Amount of

s
Time—
quality control programs, reference materials ensure Substance—
Second
accuracyintesting.Fromcerti_edreferencematerials Mole
(CRMs) and other quality grades of reference materials,
tocerti_catesofanalysis,metrologicaltraceability,and K A
other concepts, the world of reference materials is vast,
and can be confusing. Temperature— Electric Current—
Kelvin Ampere
This chapter presents critical reference material topics,
o^eringinformationonmetrologicaltraceability,the Figure 63. Metrological Traceability—SI Unit of Measurement
hierarchyofreferencematerials,certi_catesofanalysis,
referencematerialformatsanduses,aswellas_t-for-
purpose selection considerations. Proper selection of ISO 17034 and quality grades of standards,
the right reference material for a laboratory’s testing reference materials, and certified reference
application is important because results are only as materials
accurate as your reference.
Thereferencematerialhierarchyincludes_vemajor
quality grades from national metrology and other
Metrological traceability and SI Units of your primary standards, to CRMs, reference materials
reference materials (RMs), analytical standards, and research grade or
Metrological traceability is an important concept in the researchchemicals.Higherlevelsofcerti_cation
world of reference materials. A fundamental term in and traceability are required with increasing levels
metrological traceability is the International System of quality grade. While national governments give
ofUnits(SI)unitofmeasurement.TheSIde_nesthe standardizationtothetoplevel,speci_cISOguidelines
seven units of measure as the basic set from which all provide standardization for CRMs and RMs. These ISO
other SI units can be derived. The two most common SI requirements include ISO 17034, ISO/IEC 17025, and
units of measure for traceability of reference materials ISO Guide 31.
are the kilogram and mole.
Reference material producers must meet these ISO
Metrological traceability means measurements can requirements to manufacture CRMs or RMs. For both of
be meaningfully compared across di^erence places, thesequalitygrades,Certi_catesofAnalysismustbe
atdi^erenttimes,bydi^erentpeople,usingdi^erentprovidedandtheinformationcontainedwithinisde_ned
equipment. The measurement result must be related to a by the aforementioned ISO guidelines. The quality
reference through a documented and unbroken chain of speci_cationsforthelasttwolevelsarede_nedbyeach
calibrations, tracing back to the SI unit of measurement. individual producer rather than by a national government
orISOaccreditationsspeci_ctoCRMsandRMs.

66
National Metrology Standard (e.g. NIST, JRC, NMI Australia)
Compendial Standard (e.g. USP, EP, BP, JP, IP)
• Issued by an authorized body
• Considered to provide the highest level of accuracy &
traceability

Certi_edReferenceMaterial (CRM) (ISO 17034, 17025)


• Considered to provide the highest level of accuracy,
uncertainty, and traceability to an SI unit of measurement
• Manufactured by an accredited Reference Material Producer

Reference Material (RM) (ISO 17034)


4  Ful_llingISOrequirementswhicharelessdemandingthan
y

for CRMs
ilit

• Manufactured by an accredited Reference Material Producer


cab
Tra

Analytical Standard (ISO 9001)


d
an

4 Certi_cateofAnalysisavailable
ion

4 Levelofcerti_cationvaries
cat
rtifi
Ce

ReagentGrade/ResearchChemical
of

4 MaycomewithaCerti_cateofAnalysis
el

• Are not characterised for use as reference materials


Lev

Figure 64.TheHierarchyofReferenceMaterials(WhataretheDi^erentTypes?

What is measured in each grade of Homogeneity is required for the primary standards,
reference material? CRMs, and RMs, but this parameter will not be found
with the lower quality grades. Uncertainty and
Purity and identity of the material are typically included
traceability information are limited to only primary
intheCerti_cateofAnalysisforeachofthe_vequality
standards and CRMs. In the pharmaceutical world,
grades. Content and stability are required for primary
secondary standards can be CRMs or RMs, but here,
standardsorISO-de_nedCRMsandRMs.
therearetwodi^erenttypesoftraceability:tothe
Analytical standards and research chemicals may or SIunitofmeasurementforISO-de_nedCRMs,and
may not include these two parameters as their inclusion traceability to the primary compendial standard, which
is dependent on the producer. Analytical standards isarequirementspeci_ctopharmaceuticalsecondary
can also in some cases be quality control materials standards.
compliant with ISO Guide 80.

Table 17. TheHierarchyofReferenceMaterials(What,stheDi^erence?

Compendial Analytical Research


Parameter NMI Standard Standard CRM RM Standard Chemical
Purity 4 4 4 4 4 4
Identity 4 4 4 4 4 4
Content 4 4 4 4 maybe
Stability 4 4 4 4 4
Homogeneity 4 4 4 4
Uncertainty 4 4
Traceability 4 4 4
Type Primary Primary or Secondary
Measurement Secondary Standard
Standard or Standard (Pharma)
Primary Standard (Pharma)
(Pharma)

67
Understanding your reference material Be sure to examine the CoA for the producer’s quality
Certificate of Analysis systems,thereferencematerial,scerti_cationprocess,
and supporting information on traceability for a CRM.
WiththeCRMorRMgradecomesaCerti_cateof
The CoA is important since it can give the laboratory
Analysis (CoA). Within the CoA, there are several
information which ensures the reference material’s
quality parameters which are critical to understand:
certi_cationis_tforpurposewithinthetestingmetho
accuracy, consistency, homogeneity, purity, and
or application.
stability.Also,whichpropertyisbeingcerti_edis
important to understand, whether it be concentration,
potency, or content.

Accuracy
Comparison to a primary source
orcerti_edsecondsource(curve/
calibration standard. Comparison of
Certificate of Analysis – Certified Reference Material multiple independent preparations.

Gold standard for ICP


Consistency
Product no.: 38168 Lot-to-lotconsistencyveri_edby
Lot no.: Sample
Description of CRM: Gold metal (high-purity quality) in 5% HCl (prepared with HCl suitable for comparing to the previous lot.
trace analysis and high-purity water, 18.2 MΩ.cm, 0.22 µm filtered).
Expiry date: NOV 2020
Storage: 5-30°C Stability
Density (certified) at 20°C: 1024.2 kg m-3 ± 0.5 kg m-3
Expiration date established though
Constituent Certified values at 20°C and expanded uncertainties, U = k .u (k = 2) [1][2] real-time stability studies.

Gold 976 mg kg-1 ± 2 mg kg-1 1’000 mg L-1 ± 2 mg L-1 Homogeneity


Across the batch of ampoules/vials.

Metrological traceability: Certified values are traceable to the International System of units (SI)
through a metrologically valid weighing process. Details see “Details on
Purity
metrological traceability”. [3] Consistent with the neat material.
Measurement method: The certified value is determined by high-precision weighing of thoroughly
characterized starting materials and verified by measurement against NIST No contamination or degradation.
SRMs or similar CRMs
Intended use: Calibration of ICP, AAS, spectrophotometry or any other analytical technique.
Instructions for handling This reference material shall be stored in the original closed bag between 5°C
and correct use: and 30°C. Before every use of the material the bottle must be shaken well
and its temperature has to be 20°C. If storage of a partially used bottle is
necessary, the cap should be tightly sealed and the bottle should be stored at
reduced temperature (e.g. refrigerator) to minimize transpiration rate. We
highly recommend using this reference material no longer than 15 months
after the aluminum bag was opened.
Health and safety Please refer to the Safety Data Sheet for detailed information about the
information: nature of any hazard and appropriate precautions to be taken.
Accreditation: Sigma-Aldrich Production GmbH is accredited by the Swiss accreditation
authority SAS as registered reference material producer SRMS 0001 in
accordance with ISO 17034 and registered testing laboratory STS 0490
according to ISO/IEC 17025.[4][5]
Certificate issue date: 04 APR 2019
Packaging: 100 mL HDPE bottle sealed with an aluminized bag

ISO 17034 ISO/IEC 17025 ISO 9001 S. Matt – CRM Operations Dr. P. Zell – Approving Officer
SRMS 0001 STS 0490 005356 QM08

Sigma-Aldrich Production GmbH, Industriestrasse 25, 9471 Buchs, Switzerland;


Tel +41-81-755-2511; Fax +41-81-756-5449; www.sigmaaldrich.com
Sigma-Aldrich Production GmbH is a subsidiary of Merck KGaA, Darmstadt, Germany.

Certificate Page 1 of 4 Certificate version 01

Figure 65. The components of a CoA.

68
Reference material formats: do you need a Choosing the correct reference material for
neat, solution, or matrix material? your testing purpose
Referencematerialscanbeusedindi^erentformatsin Forinstrumentquali_cationsandcalibrations,establishi
the testing laboratory depending on product availability and maintaining traceability is key, and the selected
and method requirements. The three formats for reference reference material should help the laboratory achieve
materials are a neat or powder form, in solution, or this. In daily routine system suitability applications, it may
matrix. The Supelco® family of reference materials be important to qualify something that is practical and
include CRMs, RMs, or analytical standards in each easy-to-use,yetreliableandcost-e^ectiveforeveryday
format depending on the testing laboratory’s needs applications In method validation, it is critical to use
and application. highly accurate and precise materials to ensure a method
maintains these parameters. For identity and screening
purposes, proven authenticity and identity are important
attributes of reference materials. For quantitation, assays,
or stability assessment, stable and accurate reference
materials are needed.

Table 18. Di^erentFormats(HowReferenceMaterialsareUsedintheTestingLaboratory

Neat Analyte In Solution InMatrix


Form Vial/Lyophilized Ampoules/Vials/Bottles Ampoules/Vials/Bottles
Uses Weigh daily/weekly to make Ready-made,certi_edand Ready-madeandcerti_edat
stock levels & working solutions ready to use or dilute working level in matrix of choice
Pros Widely available Convenient—Saves time Convenient—Saves time
Flexible to use in a variety of Concentrationiscerti_ed& Remove need to further dilute
applications traceable into matrix of choice
Larger unit sizes available Stabl—protected from Concentration and stability is
evaporation, transpiration, O2 certi_edandtraceable
Cons May be hard to handle: Need correct mix of analytes at Long-term analyte stability in
Hygroscopic, viscous, unstable right concentration and matrix vs. diluent
volumes
Time consuming Diluent compatibility with Special handling considerations
method & storage of biological matrices
Potentially greater week-week
variability in results

Table 19. Use of Reference Materials—Type of Test

Typeoftest UseofRef.Mat. Examples RequirementsoftheRef.Mat.


Instrumentquali_cation/ Establish system performance Annualquali_cations Traceable
Calibration
Measurement accuracy Routine balance calibrations
Routine calibration/ Daily/weekly Pre-use balance calibrations Qualifyassuitableforuse
Systemsuitability
System/methodspeci_c System performance checks for
LC-UV/MS; GC-FID…
Establish routine performance
Method validation Accuracy PharmaQC;Environmentaltesting Accurate
Precision Standards of the analyte(s), Traceable
interferences, impurities
Speci_city&interferences
LOD/LOQ&Linearity
Identity Comparison of unknown to Incoming raw materials in Authenticity
known pharma, food etc.
Screening tests
Contentorassay Quantitationofanalytes Pesticide/toxin limits Certi_edcontent
PharmaQC(APIcontent Traceable
Stabilityassessment Monitor product stability PharmaQC Stable, homogenous
Internal Quality Control Method accuracy Routine quantitation of analytes Certi_edcontent
—pharma/pesticides/diagnostics
Traceable

69
Which quality grade is the best fit for purpose? Keep balances and pipettors maintained and calibrated.
Fit for purpose decisions in selection of reference For all equipment, balances, and pipettors in particular, read the
materials can depend on several factors, from regulatory manufacturer documentation and user guides to understand best
requirements, availability, and type of testing application, practices.

tolevelofaccuracyandsamplematrix.A_tforpurpose To obtain best precision and accuracy, do not use volumes that are
guidance in standard selection can be found in Table 20. less than 20% of the total volume of the pipettor.
If in doubt about the performance of a pipettor, or to develop your
technique, try dispensing water into a container and assessing
gravimetrically.

Best Practice for Preparation of Be sure that solutions are neither hot nor cold to minimize volumetric
errors.
Calibration Curves in Matrix
Considerations when preparing PreparingCalibrationStandards:
calibration standards from CRMs Keep organic content in the control matrix to a minimum—2% or
less is recommended when preparing calibrators. One easy way
toprepareasetofcalibrationstandardsisto_rstprepareseveral
Recommendationsforpreparingcalibrationstandards working solutions (WS) from a higher concentration stock solution.
Prepare each working solution such that, when added to the blank
Store compounds under the recommended conditions to ensure stability.
matrix (plasma or serum for example) at a volume of 2% or less of
After removing stock solutions from storage check, that analytes have the_nalconcentration,theresultingsolutionprovidesthedesired
gone back into solution as in some cases compounds may fall out of concentration.
solution under cold storage conditions. In this case, it may be necessary
— For example, combine 20 µL, of a working solution at 10 µg/mL,
to mix or sonicate solutions for periods of time to aid dissolution.
to 980 µL of control plasma to yield 1 mL of a 200 ng/mL
For neat materials, allow time for the container to warm to room calibration standard. Prepare additional working solutions at
temperature before opening in order to eliminate condensation from appropriate concentrations so that the same volumes can be
forming inside the container. used to prepare a complete calibrator series.
In weighing out neat compounds, it is generally best to weigh out a The strategy of preparing one high calibration standard from a stock
larger mass of material if possible (though this may not always be or working solution and then diluting this standard further is often
economical). consideredlessdesirable,asanyinaccuracyinthe_rstsolutionwill
be carried through the entire calibration series.
Tohelpreducetransferlosses,considerusingsmallvolumetric‘asks
(1 to 10 mL), weigh material into a small aluminum weighing pan, Have a second individual weigh out and prepare solutions of the
anddroptheentirepanintothe‘ask. same compounds separately. Then, check solutions against each
other using an appropriate analytical technique. Solutions should
For organic solutions, use positive displacement pipettors to get show agreement within a few percent of each other.
accurate dispensing of high vapor pressure solvents. Be sure that air
bubbles have been removed from the tip before dispensing.

Table 20. Fit for Purpose Guidance in Standard Selection

Compendial Analytical Reagent


TypeofTest NMI Standard Standard CRM RM Standard Chemical Attribute
Instrument 4 4 4 Traceability &
quali_cation/ Accuracy
Calibration
Routine 4 4 4 4 maybe Quali_ed
calibration/ standard
System (Primary or
suitability secondary)
Method 4 4 4 4 Accuracy,
validation Precision,
Bias
Identity 4 4 4 4 4 4 Authenticity
Content or 4 4 4 4 maybe Quali_ed
assay standard
Stability 4 4 4 4 maybe Quali_ed
assessment standard
Internal 4 4 4 4 maybe Quali_ed
Quality standard
Control
Regulatory/ 4 4 4 4 Quali_ed
Accreditation standard

70
Appendix
Abbreviations Links and Literature for Download
• LC-MS Resource guide
APCI Atmospheric pressure chemical ionization
CA Charged aerosol (detector) • HPLC Columns selection guide for small molecules
separation wallchart
DAD Diode array detector
• Chiral selection guide wallchart
DMSO Dimethyl sulfoxide
EC Electrochemical (detector) • HPLC Troubleshooting Guide “Untangle your liquid
chromatography problems”
ELS Evaporative light scattering (detector)
• Ascentis® Express columns brochure
ESI Electrospray Ionization
FL Fluorescence (detector) • BIOshell™ columns brochure

GC Gas chromatography • Chromolith® columns brochure


HILIC Hydrophilic liquid interaction chromatography • Puropher™ STAR columns brochure
HPLC High performance liquid chromatography • Discovery® BIO columns brochure
i.d. Internal diameter
• SigmaAldrich.com
LC Liquid Chromatography
LLE Liquid-liquid extraction
LOD limit of detection
LOQ limitofquanti_cation
MS Mass spectrometry/spectrometer
NP Normal phase
PEEK Polyetheretherketone
PES Polyethersulfone
PTFE Polytetra‘uoroethylene
PVDF Polyvinyliden‘uoride
RAM Restricted access materials
RI Refractive index (detector)
RP Reversed phase
RSD Relative standard deviation
S/N Signal to noise ratio
SPE Solid phase extraction
SPP Super_ciallyporousparticle
SST System suitability test
TFA Tri‘uoroaceticacid
THF Tetrahydrofuran
TPP Totally porous particle
UHPLC Ultra high performance liquid chromatography
USP US pharmacopoeia
UV/Vis Ultraviolet/Visible (detector)
ZIC Zwitterionic

71
72
MilliporeSigma
400 Summit Drive
Burlington, MA 01803

EMDMillipore.com

To place an order or receive technical assistance in the U.S. and Canada, call toll-free 1-800-645-5476
For other countries across Europe and the world, please visit:
For Technical Service, please visit:

MS_BR6818EN Ver. 1.0


34401
05/2021

You might also like