Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e...

91
Fundamental and Regulatory Aspects of UHPLC in Pharmaceutical Analysis Dennis Åsberg DOCTORAL THESIS | Karlstad University Studies | 2017:9 Faculty of Health, Science and Technology Chemistry

Transcript of Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e...

Page 1: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Fundamental and Regulatory Aspects of UHPLC in Pharmaceutical Analysis

Dennis Åsberg

Dennis Å

sberg | Fundamental and R

egulatory Aspects of U

HPLC

in Pharmaceutical A

nalysis | 2017:9

Fundamental and Regulatory Aspects of UHPLC in Pharmaceutical Analysis

Ultra-high performance liquid chromatography (UHPLC) provides a considerable increase in throughput compared to conventional HPLC and a reduced solvent consumption. The implementation of UHPLC in pharmaceutical analysis has accelerated in recent years and currently both instruments are used. There are, however, technical and regulatory challenges converting a HPLC method to UHPLC making it difficult to take full advantage of UHPLC in regulatory-focused applications like quality control in pharmaceutical production. In UHPLC, the column is packed with smaller particles than in HPLC resulting in higher pressure and viscous heating. Both the higher pressure and the higher temperature may cause changes in retention and selectivity making method conversion unpredictable.

Using chromatographic modelling and fundamental theory, this thesis investigates method conversion between HPLC and UHPLC. It reports on the influence of temperature gradients due to viscous heating, pressure effects and stationary phase properties on the separation performance. It also presents a regulatory concept for less regulatory interaction for minor changes to approved quality control methods and how predicable method conversion is achieved by improved understanding.

DOCTORAL THESIS | Karlstad University Studies | 2017:9

Faculty of Health, Science and Technology

Chemistry

DOCTORAL THESIS | Karlstad University Studies | 2017:9

ISSN 1403-8099

ISBN 978-91-7063-757-5 (pdf)

ISBN 978-91-7063-756-8 (print)

Page 2: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

DOCTORAL THESIS | Karlstad University Studies | 2017:9

Fundamental and Regulatory Aspects of UHPLC in Pharmaceutical Analysis

Dennis Åsberg

Page 3: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Print: Universitetstryckeriet, Karlstad 2017

Distribution:Karlstad University Faculty of Health, Science and TechnologyDepartment of Engineering and Chemical SciencesSE-651 88 Karlstad, Sweden+46 54 700 10 00

© The author

ISSN 1403-8099

urn:nbn:se:kau:diva-47852

Karlstad University Studies | 2017:9

DOCTORAL THESIS

Dennis Åsberg

Fundamental and Regulatory Aspects of UHPLC in Pharmaceutical Analysis

WWW.KAU.SE

ISBN 978-91-7063-757-5 (pdf)

ISBN 978-91-7063-756-8 (print)

Page 4: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

“Data! Data! Data!” he cried impatiently. “I can’t make bricks without clay.”

— Arthur Conan Doyle in The Adventures of Sherlock Holmes

iii

Page 5: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

iv

Page 6: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Abstract

Ultra-high performance liquid chromatography (UHPLC) provides a consi-

derable increase in throughput compared to HPLC and a reduced solvent

consumption. The implementation of UHPLC in pharmaceutical analysis,

e.g. quality control, has accelerated in recent years and there is currently a

mix of HPLC and UHPLC instrumentation within pharmaceutical compa-

nies. There are, however, technical and regulatory challenges converting a

HPLCmethod toUHPLCmaking it difficult to take full advantage ofUHPLC

in regulatory-focused applications like quality control in pharmaceutical

production.

Using chromatographic modelling and fundamental theory, this thesis

investigated method conversion between HPLC and UHPLC. It reports on

the influence of temperature gradients due to viscous heating, pressure

effects and stationary phase properties on the separation performance. It

also presents a regulatory concept for less regulatory interaction for minor

changes to approved methods to support efficient life cycle management.

The higher pressure in UHPLC gave a retention increase of up to 40%

as compared to conventional HPLC while viscous heating, instead, reduced

retention and the net result was very solute dependent. Selectivity shifts

were observed even between solutes with similar structure when switching

between HPLC and UHPLC and an experimental method to predict such

selectivity shifts was therefore developed. The peak shape was negatively

affected by the increase in pressure for some solutes since secondary

interactions between the solute and the stationary phase increased with

pressure.

With the upcoming ICH Q12 guideline, it will be possible for the

industry to convert existing methods to UHPLC in a more flexible way

using the deeper understanding and the regulatory concept presented here

as a case example.

v

Page 7: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

vi

Page 8: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Swedish Summary

De flesta läkemedel är molekyler som framställs på kemisk väg genom

en rad delprocesser. Under framställningen bildas olika biprodukter med

snarlik struktur vilka ofta har en okänd fysiologisk effekt. Därför görs alltid

en kvalitetskontroll av läkemedlets renhet där mängden biprodukter mäts.

För att analysera vad ett läkemedel innehåller så är vätskekromatografi

den vanligaste tekniken och läkemedelstillverkare har hundratals sådana

instrument för att kunna analysera alla läkemedel de tillverkar. I vätskekro-

matografi löser man först upp provet, t.ex. en tablett, i ett lösningsmedel

och sedan injiceras det i en vätskeflöde som pumpas genom ett poröst

material packat i ett rör som kallas kolonn. Provets komponenter fastnar

olika hårt påmaterialet i kolonnen vilket medför att det tar olika långt tid att

skölja ur dem och de kommer därmed ut ett och ett, dvs. de har separerats.

Allteftersom komponenterna kommer ut ur kolonnen detekteras de och

varje ämne bildar en topp i ett kromatogram.

vii

Page 9: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Jag har arbetat tillsammans med AstraZeneca för att bättre förstå hur

provkomponenterna separeras och med att utveckla effektivare kromato-

grafiska analysmetoder. Nyckeln till att designa en effektiv analysmetod

är att förstå hur de olika komponenterna fastnar, adsorberar, på de porösa

partiklarna som kolonnen är packad med. Jag har utvärderat och utvecklat

fysikaliska modeller som med hjälp av datorsimuleringar kan förutsäga

separationen. Resultaten jämfördes sedan med riktiga experiment för att

utvärdera hur bra modellen stämde överens med verkligheten.

Under de senaste åren har det varit en trend mot att packa kolonnen

med mindre och mindre partiklar eftersom topparna då blir skarpare och

analysen kan göras snabbare. De nyaste instrumenten kan utföra analyser

upp till 10 gånger snabbare och förbrukar mycket mindre lösningsmedel

vilket gör dem väldigt attraktiva för läkemedelsindustrin. Det finns dock

både tekniska och regulatoriska utmaningar med att byta ut de gamla

instrumenten. Att pumpa vätska genom en kolonn packad med så små

partiklar kräver ett högt tryck (ca 1 000 bar) och det höga trycket påverkar

hur ämnena adsorberar. Dessutom bildas det värme i kolonnen på grund

av friktionen mellan vätska och partiklar vilket ger en ojämn temperatur

i kolonnen som, tillsammans med trycket, påverkar separationen. En stor

del av avhandlingen handlar om att förstå och förutsäga hur separationen

ändras när analysmetoden överförs till ett instrumentmed små partiklar. Att

på ett förutsägbart och robust sätt överföra metoder mellan olika instrument

är viktigt då industrin har en blandning av nya och gamla instrument där

metodutvecklingen ofta sker på nya instrument och rutinanalys på äldre.

Läkemedelstillverkning är dessutom hårt reglerad av myndigheter och

en analysmetod som används för ett marknadsfört läkemedel får inte ändras

utanmyndigheternas godkännande.Då ett läkemedel oftast ärmarknadsfört

i enmängd länder är detmångamyndigheter som ska granska och godkänna

ändringar och därför vill man från industrins sida undvika ändringar om

man kan. Det här har medfört att läkemedel i vissa fall analyseras med

10-20 år gammal utrustning. Under 2017 ska nya riktlinjer släppas för EU,

USA och Japan som tillåter viss flexibilitet, t.ex. möjliggör att ändra en

kromatografisk metod så att de nyaste instrumenten kan användas. Vi

har tagit fram ett förslag på hur metoder kan utvecklas och ändras i linje

med de nya riktlinjerna och demonstrerat det med en kvalitetskontroll

för läkemedlet Nexium. Min förhoppning är att den ökade flexibiliteten

tillsammans med den djupare förståelse för separationen som presenteras i

avhandlingen, ska underlätta för industrin att implementera ny teknik som

kan effektivisera kvalitetskontrollen av läkemedel.

viii

Page 10: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

List of Papers

This thesis is based on the following papers, hereby referred to by their

roman numerals, and reprints are appended at the end of the thesis with

permission from Elsevier.

I Combining Chemometric Models with Adsorption IsothermMea-surements to Study Omeprazole in RP-LC. D. Åsberg, M. Leśko, J.

Samuelsson, A. Karlsson, K. Kaczmarski and T. Fornstedt. Chromato-graphia, 79, 1283-1291 (2016)

II Method Transfer from High-Pressure Liquid Chromatography toUltra-High-Pressure Liquid Chromatography. I. A Thermodyna-mic Perspective. D. Åsberg, M. Leśko, J. Samuelsson, K. Kaczmarski

and T. Fornstedt. Journal of Chromatography A, 1362, 206–217 (2014)

III Method Transfer from High-Pressure Liquid Chromatography toUltra-High-Pressure Liquid Chromatography. II. Temperature andPressure Effects. D. Åsberg, J. Samuelsson, M. Leśko, A. Cavazzini,

K. Kaczmarski and T. Fornstedt. Journal of Chromatography A, 1401,52–59 (2015)

IV A Quality Control Method Enhancement Concept—Continual Im-provement of Regulatory Approved QC Methods. D. Åsberg, M.

Nilsson, S. Olsson, J. Samuelsson, O. Svensson, S. Klick, J. Ennis, P.

Butterworth, D. Watt, S. Iliadau, A. Karlsson, J. T. Walker, K. Arnot, N.

Ealer, K. Hernqvist, K. Svensson, A. Grinell, P.-O. Quist, A. Karlsson

and T. Fornstedt. Journal of Pharmaceutical and Biomedical Analysis, 129,273–281 (2016)

ix

Page 11: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

V A Practical Approach for Predicting Retention Time Shifts dueto Pressure and Temperature Gradients in Ultra-High-Pressure Li-quid Chromatography. D. Åsberg, M. Chutkowski, M. Leśko, J.

Samuelsson, K. Kaczmarski and T. Fornstedt. Journal of Chromato-graphy A, 1479, 107–120 (2017)

VI A Fundamental Study of the Impact of Pressure on the AdsorptionMechanism inReversed-PhaseLiquidChromatography. D.Åsberg,

J. Samuelsson and T. Fornstedt. Journal of Chromatography A, 1457,97–106 (2016)

My contributions to the papers included in the thesis were:

I: I did the planning, the experiments, the DoE calculations and wrote the

manuscript. II: I did the planning, the experiments, the PP/FA/AED calcu-

lations and wrote the manuscript. III: I did the planning, the experiments

and wrote the manuscript together with my coauthors. IV: I participated in

the planning, did a majority of the experiments, all calculations and wrote

the manuscript together with my coauthors. V: I did the planning, the

experiments, developed the empirical approach and wrote the manuscript.

VI: I did the planning, the experiments, the calculations and wrote the

manuscript.

Papers not Included in the Thesis

VII Fast Estimation of Adsorption Isotherm Parameters in GradientElution Preparative Liquid Chromatography. I: The Single Com-ponent Case. D. Åsberg, M. Leśko, M. Enmark, J. Samuelsson, K.

Kaczmarski and T. Fornstedt. Journal of Chromatography A, 1299, 64–70(2013)

VIII Fast Estimation of Adsorption Isotherm Parameters in GradientElution Preparative Liquid Chromatography. II: The CompetitiveCase. D. Åsberg, M. Leśko, M. Enmark, J. Samuelsson, K. Kaczmarski

and T. Fornstedt. Journal of Chromatography A, 1314, 70–76 (2013)

IX Choice of Model for Estimation of Adsorption Isotherm Parame-ters in Gradient Elution Preparative Liquid Chromatography. M.

Leśko, D. Åsberg, M. Enmark, J. Samuelsson, K. Kaczmarski and T.

Fornstedt. Chromatographia, 78, 1293–1297 (2015)

x

Page 12: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

X Estimation ofAdsorption IsothermParameters inGradient ElutionPreparative RP-LC of Peptides in the Presence of an AdsorbingAdditive. D. Åsberg, M. Leśko, T. Leek, J. Samuelsson, K. Kaczmarski

and T. Fornstedt. Chromatographia, under review (2017)

XI The Importance of Ion-Pairing in Peptide Purification byReversed-Phase Liquid Chromatography. D. Åsberg, A. Langborg Weinmann,

T. Leek, R. J. Lewis, M. Klarqvist, M. Leśko, K. Kaczmarski, J. Samu-

elsson, and T. Fornstedt. Journal of Chromatography A, under review(2017)

XII Evaluation of Co-Solvent Fraction, Pressure and Temperature Ef-fects in Analytical and Preparative Supercritical Fluid Chromato-graphy. D. Åsberg, M. Enmark, J. Samuelsson and T. Fornstedt.

Journal of Chromatography A, 1374, 254–260 (2014)

XIII The Effect of Temperature, Pressure and Co-Solvent on a ChiralSupercritical Fluid Chromatography Separation. M. Enmark, D.

Åsberg, J. Samuelsson and T. Fornstedt. Chromatography Today, 7,14–17 (2014)

XIV A Closer Study of Peak Distortions in Supercritical Fluid Chroma-tography as Generated by the Injection. M. Enmark, D. Åsberg, A.

Shalliker, J. Samuelsson and T. Fornstedt. Journal of Chromatography A,1400, 131–139 (2015)

XV EvaluationofScale-Up fromAnalytical toPreparativeSupercriticalFluid Chromatography. M. Enmark, D. Åsberg, H. Leek, K. Öhlén,

M. Klarqvist, J. Samuelsson and T. Fornstedt Journal of ChromatographyA, 1425, 280–286 (2015)

XVI Analytical Method Development in the Quality be Design Frame-work. D. Åsberg, A. Karlsson, J. Samuelsson, K. Kaczmarski and T.

Fornstedt. American Laboratory, 46, 12–15 (2014)

xi

Page 13: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

xii

Page 14: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Contents

Abstract v

Swedish Summary vii

List of Papers ix

1 Introduction 11.1 Trends in Liquid Chromatography . . . . . . . . . . . . . . . 2

1.2 Trends in Pharmaceutical Analysis . . . . . . . . . . . . . . . 3

1.3 Post-Approval Changes . . . . . . . . . . . . . . . . . . . . . . 6

1.4 Aim . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7

2 Models for Liquid Chromatography 92.1 The Mass Balance . . . . . . . . . . . . . . . . . . . . . . . . . 10

2.2 The Energy Balance . . . . . . . . . . . . . . . . . . . . . . . . 11

2.3 The Adsorption Process . . . . . . . . . . . . . . . . . . . . . 12

2.4 The Stochastic Model . . . . . . . . . . . . . . . . . . . . . . . 18

3 UHPLC Fundamentals 213.1 Geometric Scaling . . . . . . . . . . . . . . . . . . . . . . . . . 22

3.2 Effects of Elevated Pressure . . . . . . . . . . . . . . . . . . . 23

3.3 Effects of Viscous Heating . . . . . . . . . . . . . . . . . . . . 25

4 Analytical Method Validation 274.1 Method Performance Parameters . . . . . . . . . . . . . . . . 27

4.2 Robustness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30

4.3 Analytical Quality by Design . . . . . . . . . . . . . . . . . . 32

5 Results and Discussion 355.1 Paper I . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35

5.2 Paper II . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39

xiii

Page 15: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

5.3 Paper III . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42

5.4 Paper IV . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45

5.5 Paper V . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50

5.6 Paper VI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54

6 Concluding Remarks 59

Acknowledgment 61

References 63

Appendix 75Paper I . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75

Paper II . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90

Paper III . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 104

Paper VI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 112

Paper V . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121

Paper VI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142

xiv

Page 16: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Chapter 1

Introduction

The pharmaceutical industry is regulated by governmental agencies to

ensure the safety, quality and efficacy of the produced drug products and

one aspect of the regulatory oversight is the drug application process [1]. In

the EU, drug applications are sent to the EuropeanMedicines Agency (EMA)

and, once approved, are valid in all member states [2]. Analytical methods

are a necessary part of the drug application and are used to generate data

for acceptance, release and stability of the drug, making pharmaceutical

analysis one of the most important branches of applied analytical chemistry

[3]. All analytical methods in the drug application have to be validated to

ensure that they produce consistent results of sufficient quality for their

intended purpose [4].

A majority of all analytical methods for pharmaceutical analysis are

based on high-performance liquid chromatography (HPLC) and the most

common mode is by far reversed-phase liquid chromatography (RPLC) [3,

5]. RPLC is a versatile separation technique well suited to separate and

quantify awide range of organicmolecules and ions commonly encountered

in pharmaceuticals [6]. HPLC was developed in the 60s but there has been

major advances throughout the years and it is still an active research

area. Among the more recent advances are ultra-high-performance liquid

chromatography (UHPLC), which takes advantage of the higher throughput

and/or better column efficiency offered by sub-2 µm stationary phase

particles, and core-shell particles having a solid core surrounded by a

porous shell [7]. However, it has been challenging to convert existing HPLC

methods to UHPLC in a predictable and robust way [8, 9]. In this thesis, I

investigate UHPLC in depth using advanced separation theory to map the

fundamental differences between UHPLC and HPLC in order to facilitate

the implementation of UHPLC in pharmaceutical analysis so that validation

meets modern regulatory guidelines.

1

Page 17: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

1.1 Trends in Liquid Chromatography

There is a trend towards increasing the column efficiency and reducing

the analysis time in order to meet the ever increasing demands from the

chemical and pharmaceutical industry [7, 10]. Increasing the column

efficiency allows for higher peak capacities or separation of closely related

compounds while reducing the analysis time increases throughput by

making it possible to analyze more samples in the same time.

UHPLC takes advantage of narrow-bore columns packed with sub-

2 µm particles which offers shorter analysis time with preserved separation

efficiency [7, 10]. The smaller particles increases the pressure resistance of

the column since it is inversely proportional to the square of the particle

diameter. In addition, the optimal mobile phase velocity is also inversely

proportional to the particle diameter making the operating pressure even

higher at optimal kinetic performance [7]. In 2004, Waters Corp. introduced

the first commercial UHPLC instrument which was marketed under the

trade name Ultra-Performance Liquid Chromatography (UPLC) [11]. A UHPLC

instrument can operate at higher pressure (usually up to 1200 bar) than

HPLC instruments and has decreased extra-column volume to minimize

system dispersion [7, 10]. Generally, a five-fold fold increase in throughput

compared to conventional HPLC can be achieved [8, 12]. UHPLC columns

have an inner diameter of 2.1 mm instead of the conventional 4.6 mm which

results in a lower volumetric flow rate and a lower solvent consumption

[8, 13]. Reducing the solvent consumption is cost-effective and makes the

analysis more environmentally friendly [13].

The pharmaceutical industry recognized the benefits UHPLC soon

after its commercialization in 2004 [14]. However, it is unlikely that the

pharmaceutical industry will replace the majority of their HPLC equipment

withUHPLC in the near future due to the cost of investing in new equipment

and a still functioning HPLC park [6]. UHPLC is mainly used at sites

focusing on research and development while production facilities still

largely employ HPLC [8, 15]. A large pharmaceutical company may have

hundreds or even thousands of HPLC systems across its organization and it

is not economically feasible to replace all instruments at once [6]. Therefore,

there is a mix of HPLC and UHPLC equipment in most pharmaceutical

organization creating a demand for efficient and robust method conversion

between instruments.

Furthermore, it can be technically challenging to predictably convert

a method from HPLC to UHPLC. A very large pressure gradient over the

2

Page 18: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

column is generated in UHPLC causing significant temperature gradients

due to viscous heating [7, 16]. Although well established scaling rules

exists when changing the column dimensions and the particle diameter [9],

the temperature gradients and the pressure itself can affect the retention,

selectivity and efficiency of the separation [16]. There has also been some

indications of stationary phase properties differing between 5 µm and

sub-2 µm particles which can further complicate matters [17].

In 2006, stationary phase particles with a solid core and a porous shell,

called core-shell particles, were reintroduced successfully to themarket after

having gained little interest since the 60s [18, 19]. Modern core-shell particles

can show a reduction in minimum plate height of up to 25% compared to

fully porous particles of the same size and have a smaller C-term in the

van Deemter equation (mass transfer coefficient) [7, 20]. Thereby, core-shell

particles can either improve efficiency compared to fully porous particles

or lower the flow resistance keeping the efficiency constant by switching

to a larger particle size [19]. The efficiency gain from core-shell particles

at optimum flow rate is not completely understood, but believed to stem

from a reduction in the longitudinal diffusion term and a reduction of the

eddy dispersion term [19, 20]. The reduction in the longitudinal diffusion

term can be understood since a significant fraction of the column volume is

occupied by a non-porous material through which the solute cannot diffuse

while the reduction of the eddy dispersion term has been attributed to a

higher transcolumn homogeneity of the core-shell columns. Using UHPLC

instrumentation with columns packed with sub-2 µm core-shell particles

is the setup that today yields the lowest height equivalent to a theoretical

plate. However, it has been suggested that such high efficiencies are not

always necessary and core-shell particles have not yet seen widespread

use in pharmaceutical analysis [19]. Although there have been reports of

pharmaceutical applications demonstrating the use of core-shell particles

[21–25].

1.2 Trends in Pharmaceutical Analysis

Historically, the pharmaceutical industry has been slow to adopt new

technologies in their manufacturing and quality control of release batches

resulting in suboptimal performance. One reason for this was a perceived

hindrance from strict governmental regulations which made it difficult to

change an already approved process or analytical method [26]. If a method

would be changed after its approval, an assessment of the modified method

3

Page 19: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

is needed and the change should be approved by the regulatory agencies in

the regions where the drug is marketed [27]. Additionally, the number of

batches that had to bediscardedbecause theydidnotmeet specificationswas

high and the reason for the batch failures was often unknown. To address

these issues, the US Food and Drug Administration (FDA) introduced the

initiative Pharmaceutical cGMP for the 21st Century - A Risk-Based Approachin 2004 [28]. The objective of the initiative was to enhance and modernize

regulation of pharmaceutical manufacturing and product quality. CMC1

and pharmaceutical regulatory programs were evaluated to encourage early

adoption of new technology and implementation of risk-based approaches,

i.e. focusing on factors critical to the safety and quality of the drug product.

The International Council for Harmonisation (ICH) releases guidelines

harmonizing the pharmaceutical development process in the EU, the US

and Japan [4], while it is the governmental agencies in each region that

evaluate and approve drug applications. Following the FDA initiative,

ICH developed a set of guidelines for pharmaceutical manufacturing and

outlined a number of concepts, among them Quality by Design (QbD), to

facilitate a science and risk-based approach to pharmaceutical manufactu-

ring [29–32]. The ICH Q8 guideline states that “pharmaceutical QbD is a

systematic approach to development that begins with predefined objectives

and emphasizes product and process understanding and control based

on sound science and quality risk management” [29]. Quality should be

built into the product by understanding the process and product giving

a more robust manufacturing process less prone to failure [33]. QbD is

also intended to give more regulatory flexibility to the manufacturing

process and, together with a modern quality management system, promote

continual improvement of the process during its lifetime.

The industry has begun implementing parts of the ICH Q8–Q11 gui-

delines and this is often denoted as an enhanced approach to product

development compared to a traditional approach. However, regulatory

agencies are not classifying drug applications as enhanced or traditional

applications as they often contains elements from both approaches. In can

be said to exist a continuum of approaches with the traditional approach,

based on compliance with set specifications, at one end and the science and

risk based, enhanced approach on the opposite end.

After the introduction of QbD in manufacturing, researchers began

1Chemistry, manufacturing and control are drug development activities before the clinical

trials which involves assessing safety, pharmacokinetics, dose, etc. as well as developing the

manufacturing process and suitable packaging.

4

Page 20: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

applying QbD principles also to the development of analytical methods [34].

This extension of QbD is known as analytical QbD and the objective is to

increase the robustness of analytical methods and provide more regulatory

flexibility to make the life-cycle management of the methods more efficient

[35–39]. However, there is currently no international consensus from

regulatory agencies on analytical QbD and no official guidelines have been

released. Therefore, applications that includes analytical QbD elements are

evaluated on a case-by-case basis [40].

A timeline of important guidelines concerning drug development is

shown in Fig. 1.1. An annex to ICH Q8 was added in 2008 which discussed

QbD explicitly and an official FDA guideline referring to the ICH Q8–Q10

documents were released in 2011 [41]. Analytical QbD was first suggested

in 2007 [42] and has recently been elaborated in several book chapters [26,

43, 44]. The next ICH guideline, ICH Q12, is due 2017 and will be about

life-cycle management and post-approval changes, since the current ICH

guidelines have not had the intended impact on these subjects [45].

Figure 1.1: A timeline over important guidelines for pharmaceutical development.

QbD relies on the construction of a design space where the relationship

between critical factors and process quality is known. The design space is

part of the drug application and it is possible to change the process inside

the design space without the change counting as a post-approval change

[33]. Mathematical modeling is then needed to obtain the relationship

between quality attributes and input variables (settings, materiel attributes,

etc.) and the complexity of the model depends on the process. The model

forms the basis of the knowledge about the process on which to base the

design decisions in the drug application [29]. Therefore, there is also a

trend towards increased mathematical modeling and computer simulations

both in pharmaceutical manufacturing and analysis [46].

5

Page 21: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

1.3 Post-Approval Changes

To convert an approved HPLC method to UHPLC, a post approval change

need to be filed to the regulatory agencies. FDA denotes a change to an

approved drug application as a post-approval change [47] while the EMA

denotes it as a post-authorization variation [48]. To avoid confusion, I will

use the FDA term, but the terms are interchangeable. Depending on the

potential of a change to have an effect on the safety or effectiveness of a drug

product, FDA divides changes into three types; major, moderate and minor

changes. A major change requires the submission of a supplement and

approval by FDA prior to distribution of the drug product made using the

change. A moderate change is divided into two subtypes, one requires the

submission of a supplement to FDA at least 30 days before the distribution of

the drug product made using the change and the other allows distribution

as soon as FDA receives the supplement. A minor change has only minimal

potential to have an adverse effect and need only be described in the annual

report. EMA recognizes similar types of changes but denotes them as type

IA, IB and II where type II is similar to a major change, type IB is similar to

a moderate change and type IA is similar to a minor change.

A change to an analytical method outside the specifications given in

the drug application is categorized as moderate or major depending on its

potential to have an adverse impact on the performance of the analytical

method [47]. Formally, converting a HPLC method to UHPLC rank as

type 1B in the EU, meaning that regulatory agencies have 30 days to send

questions or require more information and if they do not, the change can

be implemented when the time period expires [48]. However, in reality

there is always a dialog between the company and regulators before a post

approval change of this kind is implemented.

After a modification, the method should perform equally or better

compared to the original one. The modified method need to be revalidated

and an equivalence protocol between the modified and original method

should be established to get approval from the regulatory agencies [8].

However, it takes time to get the necessary approvals, since a method

can easily be marketed in over 100 countries [6]. Therefore, it may not be

feasible, under current regulations, to change a HPLC method to UHPLC

for a marketed product. Therefore, new drug applications can have two

analytical methods, one for HPLC and one for UHPLC, filed for the same

analysis to avoid changing from HPLC to UHPLC post approval [12, 49].

6

Page 22: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

1.4 Aim

Implementation of new technology for continual improvement of pharma-

ceutical manufacturing and analysis is today essential to stay competitive

and regulatory agencies has began promoting adoption of new technology

in current and upcoming regulatory guidelines. UHPLC has great potential

to improve the throughput in pharmaceutical analysis but both technical

and regulatory challenges for a wide spread implementation exists.

This thesis addresses the technical challenge with predictable method

conversion between HPLC and UHPLC from a fundamental perspective

and the regulatory challenge of efficient post approval change management.

Paper [I] studies the separation of the drug substance omeprazole from two

impurities, and investigates how mechanistic modeling can complement

conventional chemometric models to elucidate the separation process. Ome-

prazole, Fig. 1.2, is a proton-pump inhibitor and the API in the blockbuster

drugs Losec and Nexium [50]. In paper [II] the potential differences in

stationary phase properties between sub-2 µm particles and conventional

HPLC particles were investigated. The effect of elevated pressure and

viscous heating on the retention and peak shape was studied in papers

[III,V,VI] while the conversion of a quality control method from HPLC

to UHPLC was studied in paper [IV]. Furthermore, in paper [IV] a new

concept for handling post-approval changes with minimal regulatory in-

teraction was presented and exemplified with a realistic case example, a

quality control method for the drug Nexium. In the end my hope is that the

knowledge gained from a fundamental understanding of UHPLC would

support analysts to reliably switch between HPLC and UHPLC based on

scientific decision making which, in turn, would increase the regulators’

trust. Our regulatory concept may serve as support for implementing the

upcoming ICH Q12 guideline handling life cycle management of analytical

methods.

Figure 1.2: The structure of the proton pump inhibitor S-omeprazole.

7

Page 23: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

8

Page 24: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Chapter 2

Models for Liquid Chromatography

Amechanistic model is based on assumed physical or chemical processes

which are thought to have given rise to the phenomena which are being

studied. The parameters in a mechanistic model have a physical definition

and can be measured, in theory at least, independently of each other. In

contrast, a statistical model is based on an empirical relationship which

only seeks to best describe the data. The model parameters lack physical

meaning and does not attempt to explain underlying principles of the

studied process.

In liquid chromatography, models are mainly used for two purposes;

to increase the understanding of the chromatographic process and to find

optimal operating conditions for a separation. Mechanistic models have

been used extensively to increase the understanding of the adsorption [51,

52] and mass transfer [53, 54] processes in chromatography while statistical

models are common in method development and validation [43, 44].

For some models, it is important to distinguish between linear and

nonlinear chromatography because they may not be applicable to both [55].

Linear chromatography refers to conditions where the concentration of

adsorbed solute is directly proportional to the solute concentration in the

mobile phase, i.e. the adsorption isotherm is a linear function. This is the

situation often encountered in analytical applications of chromatography.

Nonlinear chromatography refers to a nonlinear relationship between the

solute concentration in the mobile phase and in the stationary phase,

i.e. a nonlinear adsorption isotherm, and is encountered in preparative

chromatography, but also in analytical separations where the peaks are

overloaded. Models of chromatography based on the mass balance of each

component can be applied to both linear and nonlinear conditions while

molecular dynamic models, which are based on a microscopic description

of the solute migration, are not suitable for nonlinear chromatography [56].

9

Page 25: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

2.1 The Mass Balance

The mass balance of a solute is a partial differential equation describing

how the solute is transported through the chromatographic column and

has parameters such as flow rate, bed porosity and adsorption equilibrium

parameters. The equilibrium-dispersive (ED)modelwasused todescribe the

mass balance in paper [I,II,VI]. The EDmodel assumes that the contribution

of all non-equilibrium effects can be lumped together into an apparent

axial dispersion coefficient, that the mobile phase is non-compressible, that

the pure solvent is not adsorbed on the stationary phase and isothermal

conditions [55]. For a radially homogeneous column the ED model is

written as

∂C∂t

+ F∂q∂t

+ w∂C∂z

� Da∂2C∂z2

(2.1)

where z is the length coordinate, t the time coordinate, F is the ratio between

stationary and mobile phase volume and w is the interstitial velocity of the

mobile phase. If w is unknown, it can be replaced by the linear velocity. The

equilibrium concentration of the solute in the stationary phase, q, is relatedto its corresponding mobile phase concentration, C, through the adsorption

isotherm. The apparent axial dispersion coefficient, Da, is calculated from

Da �Lu

2Nap

(2.2)

where L is the length of the column, u is the linear velocity and Nap apparent

plate number. The initial condition used in this work was that the column

was equilibrated with pure mobile phase before each experiment. The

Danckwerts boundary conditions were applied [57]:

wC(0, t) − Da∂C∂z

����z�0

� wC0(t) (2.3a)

∂C∂z

����z�L

� 0 (2.3b)

where C0(t) is the injection profile which is determined experimentally.

The ED model was solved numerically; orthogonal collocation on finite

elements were used to discretize the spatial derivatives [58] and the Adams-

Moulton method implemented in the VODE procedure was used to solve

the system of ordinary differential equations [59]. This numerical method is

robust and has been used extensively in the past to solve the ED model [55].

10

Page 26: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

2.2 The Energy Balance

In UHPLC, heat is generated in the column due to viscous friction when

the mobile phase percolates the column. To describe this process, the

energy balance for the bed and the column wall in cylindrical coordinates

are needed [60, 61]. The energy balance for the bed describing the heat

generated by viscous friction and the energy evacuated from the column

under steady state conditions is

Cp uz∂T∂z

+Cp ur∂T∂r

�1

r∂∂r

(rλeff

∂T∂r

)+∂∂z

(λeff

∂T∂z

)− uz(1−αT)∂P

∂z(2.4)

where Cp is the mobile phase heat capacity, uz and ur are local super-

ficial mobile phase velocities in z and r directions respectively, T is the

temperature, P is the pressure, α is the mobile phase thermal expansion

coefficient and λeff is the effective bed thermal conductivity which depends

on temperature and hence position. The left-hand side of Eq. (2.4) is the

flux of energy carried with the mobile phase by convection. The first and

second term on the right-hand side describes the energy change due to

conduction and the third therm is the heat generated by viscous heating

and lost by thermal expansion of the mobile phase. The energy balance for

the column wall in contact with the surrounding is also needed [60, 61]:

0 � λw

(1

r∂T∂r

)+∂2T∂r2

+ λw

∂2T∂z2

(2.5)

where λw is the column wall conductivity. The column is no longer

isothermal and using the ED model for a radially heterogeneous bed in

cylindrical coordinates and accounting for spatial variations in u and Da

yields [62, 63]:

∂C∂t

+ F∂q∂t

+1

εt

∂(uC)∂z

�∂∂z

(Da,z

∂C∂z

)+

1

r∂∂r

(rDa,r

∂C∂r

)(2.6)

εt is the total porosity of the bed. When the column is packedwith core-shell

particles instead of fully porous ones, the phase ratio is calculated as

F �(1 − εe) (1 − εs)

(1 − ρ3

)εt

(2.7)

where εe is the external porosity, εs is the shell porosity and ρ is the ratio of

the solid core diameter to the external diameter of the core-shell particle.

The ED model performs well modeling elution profiles in non-isothermal

conditions [64] when solving the steady state energy balance, Eq. (2.4) and

Eq. (2.5), for the column prior to the mass balance, Eq. (2.6).

11

Page 27: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

2.3 The Adsorption Process

The adsorption process is the core of modeling RPLC, since it governs

the separation of the sample components and it is characterized by the

adsorption isotherm for each component. There exists a number of different

adsorption isotherm models, both mechanistic and empirical, but this

section only discusses the ones used in the thesis. The adsorption isotherm

needs to be determined experimentally and when doing this, calculating

the adsorption energy distribution is often helpful for selecting the most

appropriate isotherm model.

2.3.1 Adsorption IsothermModels

The adsorption isotherm relates the concentration of a solute adsorbed on

the stationary phase to the concentration in the mobile phase. The most

common liquid-solid adsorption isotherm is the Langmuir isotherm [55]:

q(C) �qsbC

1 + bC(2.8)

where qs is the monolayer saturation capacity and b is the association

equilibrium constant. At the limit C → ∞ we have q � qs, because the

Langmuir model assumes no adsorbate-adsorbate interaction and therefore

only a monolayer of solute molecules covers the stationary phase. The

retention factor, k, is related to the initial slope of the adsorption isotherm,

i.e. for Eq. (2.8) we have

k � F lim

C′→0

dqdC

����C′

� Fqsb � Fa (2.9)

where a is called the Henry constant. If the stationary phase is heterogene-

ous, and contains two discrete types of adsorption sites, the bi-Langmuir

model can be used instead [55]:

q(C) �qs,1C

1 + b1C+

qs,2C1 + b2C

(2.10)

where subscripts 1 and 2 denotes the two adsorption sites and qs � qs,1+ qs,2.

The bi-Langmuir model have been used in paper [II,VI]. If the solute

exhibits multi-layer adsorption, the liquid-solid BET model is suitable [65]:

q(C) � qs

bSC(1 − bLC) (1 − bLC + bSC) (2.11)

12

Page 28: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

where bS is the liquid-solid association equilibrium constant and bL is the

association equilibrium constant between layers of adsorbed molecules.

This adsorption isotherm was used to model the adsorption of omeprazole

in paper [II]. Eq. (2.11) can be limited to solute molecules adsorbing in two

layers and this version of the BET isotherm is written as [66]

q(C) � qs

bSC + 2bSbLC2

1 + bSC + bSbLC2

(2.12)

which is mathematically equivalent to the quadratic model and the Moreau

model, but derived under different assumptions [55]. Eq. (2.12) was

employed in paper [I] to model the adsorption isotherm of the neutral form

of omeprazole.

Adsorption isotherms can be classified into different types based on their

shape using the van derWaals classificationwhichwas originally developed

for gas-solid equilibrium isotherms in the 40s [67]. Type I represents

isotherms which are convex upwards, like the Langmuir isotherm, and

are the most common type of liquid-solid equilibrium isotherms. Both

type II and type III isotherms are possible when there are significant

adsorbate-adsorbate interactions, but are uncommon in RPLC. In type II,

the interaction between the solute and the stationary phase dominates and

the solute will form a complete monolayer before starting to form the next

layer. If the adsorbate-adsorbate interactions dominates, incomplete layers

will form and we have a type III isotherm. The BET isotherm, Eq. (2.11) can

describe both type II and type III depending on the relative strength of the

equilibrium constants. Fig. 2.1 shows characteristic adsorption isotherms of

type I-III, their corresponding overloaded elution profiles and a schematic

illustration of the formation of solute layers as the concentration increases.

In gradient elution, the organic modifier fraction changes continuously

during the migration of the solute through the column. The parameters in

the isotherms are dependent on the fraction of organic modifier, φ, and will

therefore also vary during the run [VII-VIII]. Based on the linear solvent

strength theory, the Langmuir model can be extended to incorporate φ

giving

q(C, φ) � a0e−SaφC

1 + b0e−SbφC

(2.13)

where Sa and Sb are parameters describing the sensitivity to the organic

modifier and a0 and b0 are the isotherm parameters in pure water. The

modifier fraction can be included in the bi-Langmuir and BET models in an

analogues manner which was done in paper [II].

13

Page 29: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Figure 2.1: a) Adsorption isotherms corresponding to type I, II and III. The type

I isotherm was obtained with Eq. (2.8) and type II and III was obtained with

Eq. (2.11). The parameters were qs � 100 g/L, b � bS � 0.1 L/g and bL � 0.04

L/g for type II and bL � 0.06 L/g for type III. b) Corresponding elution profiles

for an 500 µL injection of a 10 g/L sample calculated with Eq. (2.1) using linear

velocity 15 cm/min and Da � 0.058 cm2/min. c) Illustration of the formation of

layers corresponding to the three types.

2.3.2 Adsorption Isotherm Acquisition

The adsorption isotherm parameters for a certain system cannot be deter-

mined from theory and so must be determined experimentally. There are

many different experimentalmethods for determining adsorption isotherms.

Frontal analysis (FA) and the perturbation peak (PP) method are accurate

and well established in the literature. However, they require relatively large

amounts of sample and many experiments [68]. In FA, the dependence

of retention time of breakthrough curves on the solute concentration is

used to determine single component adsorption isotherms. In the PP

method, the column is equilibrated with a certain solute concentration

and a perturbation is created by injecting a small excess or deficiency of

the solute. The retention time of the perturbation peak is related to the

slope of the adsorption isotherm and by introducing perturbations with

the column being equilibrated with different solute concentrations, the

adsorption isotherm can be determined.

Two methods which requires fewer experiments and less sample are the

14

Page 30: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

elution by characteristic point (ECP) method [69] and the inverse method

(IM) [70, 71]. In the ECP method, the diffuse rear of an overloaded elution

profile is related to the adsorption isotherm. In the inverse method, an

adsorption isotherm model has to be chosen a priori, and the parameters

are estimated by optimizing the agreement between calculated and experi-

mental elution profiles. Both the ECP and IM method are limited by the

highest eluted concentration of the elution profile. The inverse method has

some advantages compared to other methods, namely that it can be used to

estimate not only adsorption isotherm parameters but also parameters in

the mass balance such as Da and that it can be used when the conditions in

the column are not constant. For example, the inverse method was used

to estimate adsorption isotherm parameters directly from elution profiles

obtained in gradient elution, Fig. 2.2, in paper [VII,VIII,IX].

Figure 2.2: An illustration of the inverse method in gradient elution; first an initial

guess of the adsorption isotherm parameters in Eq. (2.13) was done followed by a

numerical optimization to the experimental elution profile.

A proven strategy for determining how the adsorption isotherm for a

solute varies with experimental conditions such as the fraction of organic

modifier in the mobile phase, is to determine the the adsorption isotherm

with FA or PP at one reference condition and then use ECP or IM to

determine the isotherm parameters at new conditions [51]. This approach

was used to study the effect of pH [I], organic modifier [II] and pressure

[VI] on the adsorption isotherm.

15

Page 31: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

2.3.3 The Adsorption Energy Distribution

The adsorption energydistribution (AED) assumes a continuousdistribution

of adsorption energies and is used to characterize the heterogeneity of a

surface, thereby determining the number of different types of adsorption

sites present on the stationary phase surface. This information can then be

used to select an appropriate adsorption isotherm model [51].

The equilibrium constant is related to the adsorption energy, εa, through

the Arrhenius equation

b � b0eεa/RT

(2.14)

where b0 is a preexponential factor and R is the universal gas constant. A

fundamental equation of the global adsorption isotherm on heterogeneous

surfaces is [72]

q(C) �∫

f (ln b)θ(b , C)d(ln b) (2.15)

where q(C) is the amount of solute adsorbed at solute concentration C,

f (ln b) is the AED and θ(b , C) the local adsorption model. θ(b , C) is oftenthe Langmuir model, Eq. (2.8), but it is possible to use others like the BET

model, Eq. (2.11), which was done in paper [II]. To obtain the AED from

Eq. (2.15) for a set of experimental adsorption data (C and q), the expectationmaximization (EM) algorithm provides a robust method that converges

with high stability toward the maximum-likelihood estimate [72, 73]. It

does not require any prior knowledge of the distribution function and no

smoothing of the isotherm data. A unimodal and a bimodal AED is shown

in Fig. 2.3 with corresponding adsorption isotherms. It would be difficult

to distinguish between a homogeneous or heterogeneous model from only

the adsorption isotherm data.

2.3.4 Insights Into the Adsorption Mechanism

The stationary phase surface of a C18-bonded porous silica adsorbent is

always, to some extent, heterogeneous [74]. The heterogeneous surface

arise from different sources; e.g. heterogenities of the bare silica surface

due to elemental impurities or bond strains of the silicate tetrahedra [75],

residual silanol groups [76] or heterogenities in the structure of the C18

bonded phase itself [77].

Based on adsorption isotherm measurements and AED calculations, it

has been suggested that the heterogeneity of the stationary phase causes the

solute to adsorb through different interactions on the same adsorbent [74].

The solute interacts with different types of adsorption sites characterized by

16

Page 32: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

-7 -6 -5 -4 -3 -2 -1 0

ln(b)

0

100

200

300

qs [

g/L

]

a)

0 50 100 150

C [g/L]

0

20

40

60

80

100

q [

g/L

]

b)

Langmuir

bi-Langmuir

Figure 2.3: a) AED for surfaces with one and two types of adsorption sites. b)

Corresponding adsorption isotherms where Langmuir is a one site model and

bi-Langmuir is a two site model. (Langmuir: qs � 110 g/L, b � 0.027 L/g and

bi-Langmuir: qs,1 � 100 g/L, b1 � 0.01 L/g, qs,2 � 10 g/L, b2 � 0.2 L/g.)

their adsorption energies. From AED calculations, it is evident that there

are several discrete adsorption sites and low molecular weight compounds

usually show two distinct adsorption sites on an end-capped stationary

phase in RPLC [51, 74]. The adsorption site having the lowest energy, i.e.

the weakest equilibrium constant, has the highest saturation capacity. It

has been proposed that the low energy sites are dispersive interactions

at the interface between the mobile phase and the C18-bounded layer.

The adsorption sites having a higher adsorption energy and thereby a

larger equilibrium constant, usually have a lower saturation capacity and

is located deeper into the C18 layer, closer to the silica surface. The exact

nature of these adsorption sites are not completely understood and depends

on the properties of the solute molecule. A schematic description of a

solute adsorbing on a C18-bonded stationary phase having two different

adsorption sites is given in Fig. 2.4.

Even though the high energy sites are fewer in number, they can

contribute significantly to the retention factor [78]. If the bi-Langmuir

adsorption isotherm is assumed, the retention factor is related to the

adsorption isotherm through Eq. (2.9) and since the low energy sites

have high saturation capacity and low equilibrium constant and vice

versa for the high energy site, they contribute to a similar extent to the

retention factor. Furthermore, a well known reason for peak tailing in liquid

chromatography is column overloading and a solute having heterogeneous

adsorption can have a different linear isotherm range compared to a solute

17

Page 33: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Figure 2.4: Schematic description of the adsorption equilibria for a solute molecule

adsorbing at two types of adsorption sites; one located near the C18-bonded layer

interface and one located deeper in the C18 layer.

having homogeneous adsorption.

Molecular simulations supports the interpretation of multiple adsorp-

tion sites [52]. Calculations of the spatial distribution coefficient for the

solute in the direction perpendicular to the silica surface show how the

solute is distributed in different regions of the stationary phase. For a

polar compound, 1-propanol, the distribution coefficient exhibited two

preferential regions [79]. 1-Propanol adsorb on the interface of the C18

layer and the second region was close to the silica surface, deep into the

C18 bonded layer with the interface region being much larger.

2.4 The Stochastic Model

Another way of describing the chromatographic process is through molecu-

lar dynamic models which looks at the molecular level [56]. A molecular

dynamicmodel is based on a stochastic description of themolecule’s random

migration through the column. Amolecule is assumed to perform a random

number of adsorption-desorption events, denoted n, and for each instance

it adsorbs, it spends a random amount of time adsorbed on the stationary

phase. The average time a solute molecule spends in the moving mobile

phase is called fly-time, τm, and the average time it spends adsorbed on

18

Page 34: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

the stationary phase or in a stagnant part of the mobile phase is called

the sojourn time, τs. τs and τm are average quantities made up of the

individual values of the adsorption time for each adsorption event and the

time spent in the mobile phase between two adsorption events, which are

random quantities. The retention factor, k, is the ratio between τs and τm ,

i.e. k � τs/τm, and the retention time is calculated as tR � nτm + nτs, i.e.

the sum of the time spent moving in the mobile phase nτm and the time

spent in the stationary phase nτs after n adsorption-desorption events.

Stochastic models can be used to study the peak shape in linear chro-

matography and to better understand why some peaks are tailing even at

very lower concentrations. In line with the discussion about heterogeneous

adsorption in Section 2.3, molecular dynamic models also assumes multiple

adsorption sites [80]. Adsorption at a site with high adsorption energy are

characterized by a long τs while adsorption sites with a low adsorption

energy are characterized by a short τs. Adsorption on a heterogeneous

surface can result in peak tailing because the sites with short τs produce the

sharp front of the peak while the sites with long τs give rise to an elongated

tail.

In paper [III], themolecular dynamicmodel developed byGiddings and

Eyring [81] was used to study the peak shape as a function of pressure and

temperature for a tailing, cationic solute. In thismodel, the chromatographic

process is assumed to be a Poisson process and the chromatographic peak

is the probability density function of time spent in the column by the

molecules. If the stationary phase surface is assumed to be covered with

two types of adsorption sites and the relative amount of these sites are

denoted pi , the retention factor is

tR � nτm + n(p1τs,1 + p2τs,2

)(2.16)

The column efficiency for an surface with two adsorption sites is given by

1

N�

1

ND

+ 2np1 + p2

(τs,2/τs,1

)2(

p1 + p2

(τs,2/τs,1

) )2

(k

k + 1

)2

(2.17)

where ND is the dispersion effect from the mobile phase estimated from

an unretained marker. The skew is traditionally used to describe the peak

symmetry and is

S �3

2

√n

p1τ3

s,1 + p2τ3

s,2(p1τ2

s,1 + p2τ2

s,2

)3/2 (2.18)

19

Page 35: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

A symmetrical chromatographic peak has S � 1 and a tailing peak has

S > 1.

Direct evidence for the stochastic model assuming a multi-site adsorp-

tion process have been reported by single molecule spectroscopy [76, 82, 83].

Single molecule spectroscopy techniques provides experimental data of the

adsorption-desorption steps for individual molecules which is not possible

to obtain through conventional chromatographic experiments providing

only the ensemble-averaged information. When studying the adsorption of

a cationic molecule on a commercially available, end-capped C18-bonded

stationary phase with single molecule spectroscopy, strong, randomly dis-

tributed adsorption sites were revealed [84]. Additionally, the trajectory

of single molecules moving through C18-bonded, porous particles were

measured and from the adsorption-desorption events, there was evidence of

heterogeneity in the interaction with the surface [85]. Using single-molecule

data, it is also possible to approximate the overall adsorption site residence

time distribution for a process including both weak and strong binding

sites [86, 87]. With this approach, the experimental elution profiles could

be reproduced and the asymmetry of the peak at different mobile phase

compositions was correctly predicted.

20

Page 36: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Chapter 3

UHPLC Fundamentals

Sub-2 µm particles results in a high pressure resistance, a reduced C term

giving a flatter profile of the van Deemter curve and a lower optimal plate

height [88]. Fig. 3.1 compares the pressure drop and van Deemter curves

for columns packed with 1.7 µm, 3.5 µm and 5.0 µm particles. A higher

throughput and/or efficiency can be achieved in UHPLC at the cost of a

higher operating pressure [8]. This chapter discusses how to geometrically

scale a chromatographic method when the column dimensions and particle

size are changed. However, for a predictable conversion from HPLC to

UHPLC, the potential effects from the high pressure need to be taken into

account. The pressure can affect the chromatography by both influencing

the retention mechanism, and the the mobile and stationary phase by

compressing it [89]. Viscous heating is discussed separately from the other

consequences of high pressure since it is essentially a temperature effect.

Linear velocity

Pre

ssu

re

a)

1.7 µm

3.5 µm

5.0 µm

Linear velocity

HE

TP

b)

1.7 µm

3.5 µm

5.0 µm

Figure 3.1: a) Comparing pressure drops over the column for three particle

diameters. b) Comparing van Deemter curves for these columns.

21

Page 37: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

3.1 Geometric Scaling

Geometric scaling from one column dimension or particle size to another

with preserved selectivity is well established in liquid chromatography

[9]. UHPLC instruments have low extra-column volumes compared to

conventional HPLC to decrease extra-column band broadening and gradient

dwell time [7]. Therefore, these volumes need to be accounted for as well

when scaling a method from HPLC to UHPLC. In isocratic elution, the

injection volume, Vinj, and flow rate, Fv, need to be scaled when changing

the particle diameter, dp, and columndiameter, dc [90]. The injection volume

for the changed method, denoted 2, is related to the injection volume of the

original method, denoted 1, through

Vinj,2 �V0,2

V0,1Vinj,1 (3.1)

where V0 is the column hold-up volume. The flow rate is scaled so that the

reduced linear velocity of the mobile phase is preserved, i.e.

Fv,2 �

d2

c,2

d2

c,1

dp,1

dp,2Fv,1 (3.2)

In gradient elution, the gradient dwell time, td, (the time before the gradient

reaches the column inlet) and gradient slope, β, must also be scaled and

the most straightforward approach is based on the linear solvent strength

theory [91, 92]. The scaled dwell time is then

td,2 �Fv,1

Fv,2

V0,2

V0,1td,1 (3.3)

where the Fv,2 is calculated with Eq. (3.2). For a linear solvent gradient, the

gradient volume should be preserved, i.e.

β2 �Fv,2

Fv,1

V0,1

V0,2β1 (3.4)

Experimental examples of geometric scaling can be found in [90, 91] and

was also performed in paper [IV].

However, even when following the geometric scaling rules rigorously,

the separation performance may shift between HPLC and UHPLC due to

the higher pressure in UHPLC [93–98]. Pressure and temperature effects are

discussed in the subsequent sections while the consequences for method

conversion are discussed in paper [II-V].

22

Page 38: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

3.2 Effects of Elevated Pressure

Atpressures around1000 bar, the columnsteel casing expands, the stationary

phase is compressed and the mobile phase can no longer be regarded as

incompressible [89]. Although these effects have a relativelyminor influence

on routine applications, theymay need to be accounted forwhen performing

fundamental research since they affect important parameters such as the

mobile phase velocity and the bed porosity [99–101]. For conventional

columns, the expansion of the casing is increasing the volume by around

0.1% per 1000 bar [89]. The compression of a C18-bonded stationary phase

can decrease its volume by ca 1% when increasing the pressure 1000 bar

[89]. It is mainly the C18-bonded layer that is compressed and since most

of the surface area is located inside the pores, it is the internal porosity of

the bed which is most affected. The specific volume of the mobile phase

as a function pressure largely depends in its composition, with e.g. water

having a lower compressibility than acetonitrile, and the compressibility

of the mobile phase affects the local flow rate. However, the definition of

the flow rate set by the operator varies between instrument manufacturers

[7]. If the set flow rate is defined at the low pressure side of the pump, the

actual flow rate will be lower than the set one at the column inlet. On the

other hand, if it is defined at the high pressure side of the pump, the actual

flow rate will be higher than the set one at the column outlet.

InHPLC conditions, the pressure has been known to affect retention for a

long time [102–104], but has not gained much attention due to the moderate

pressure found in conventionalHPLC.However, with the commercialization

of UHPLC, the effect of pressure on the retention mechanism became more

important since much larger pressure effects were encountered [93–96].

Since the pressure drop over the column is close to linear which means that

a pressure of 1000 bar during normal operation gives an average pressure

of 500 bar in the column [89], the solute will experience varying pressure

when it travels through the column. The retention factor can be related to

the pressure through [105]

ln (k) � −∆VRT

P + ln

(FF0

)+ ln (k0) (3.5)

where ∆V is the difference in solute molar volume in the stationary phase

and in the mobile phase and subscript 0 indicates a reference condition. In

RPLC ∆V < 0, i.e. the solute molar volume is smaller when the solute is

adsorbed on the stationary phase, and an increase in retentionwith pressure

is observed. This can easily be understood through le Chatelier’s principle;

23

Page 39: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

when the pressure is increasing the system strives to counteract the change

by increasing its volume, and hence lowering the pressure, which is achieved

when the solute spends a longer time in the stationary phase. Non-polar

solutes have a small pressure dependence while polar and charged solutes

have a larger pressure dependence [93–95]. This implies that ∆V is more

negative for polar and charged solutes, i.e. they experience a larger decrease

in volume when adsorbing. McCalley et al. [93–95] have suggested that

polar and charged solutes lose a larger part of their solvation layer when

adsorbing compared to nonpolar solutes (Fig. 3.2). Nonpolar solutes have

a solvation layer consisting of hydrophobic molecules like acetonitrile or

methanol which can more easily penetrate the C18-bonded layer when the

solute is adsorbed. Apart from the polarity, the molecular weight of the

Figure 3.2: Schematic description of the loss of solvation layer upon adsorption for

a polar compound on a C18-bonded stationary phase.

solute has a large influence on its pressure dependence. For example, for a

1000 bar pressure increase the retention factor increases 25-100% for small

molecules, 150% for peptides (∼ 1.3 kDa), 800% for insulin (∼ 6 kDa) and

3000% for myoglobin (∼ 17 kDa) [96].

The effect of pressure on column efficiency, assuming no viscous heating,

is not well investigated since it is difficult to study without causing viscous

heating. However, theoretical calculations shows that pressure has a

negative effect on column efficiency when working above the optimal flow

rate [106, 107]. The decrease in column efficiency was attributed to the

pressure dependence of the solute diffusion coefficient.

24

Page 40: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

3.3 Effects of Viscous Heating

When the mobile phase percolates a column packed with very fine particles

at a sufficiently high flow rate, heat is generated due to viscous friction [108].

Viscous heating is negligible at HPLC pressures (< 400 bar), but becomes

significant in UHPLC and has therefore been given much attention in the

last decade [16]. The power generated by viscous friction is

ÛW � Fv∆P (3.6)

making it directly proportional to both the flow rate and the pressure

drop [109]. The temperature profile in the column is dependent on how

the column is thermostated [60, 61]; in a still air environment the axial

temperature gradient prevails since the conditions are close to adiabatic

while in a water bath the heat is effectively removed from the column wall

and radial temperature gradients dominates (Fig. 3.3a). In still air conditions

the axial gradients are in the range of 10◦C to 20

◦C and in a water bath the

radial gradients are in the range of 0.5 ◦C to 5◦C at around 1000 bar.

Figure 3.3: a) Schematic illustration of the temperature gradient obtained with two

different thermal environments for the column at high flow rate. b) The reduced

height equivalent to a theoretical plate (HETP) and retention factor for omeprazole

as a function of mobile phase velocity on a BEH C18, 1.7 µm column in a still air

column oven and a water bath.

25

Page 41: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Column efficiency is mostly affected by radial temperature gradients

because they cause the velocity profile of the solute band to vary in the

radial direction which makes the solute band broader [61, 110]. Therefore,

the efficiency loss is much larger in a column thermostated with a water

bath since the majority of the generated heat is then removed through the

column walls. In still air conditions, the negative effect of viscous heating

on efficiency is generally quite small which is evident from the van Deemter

curves in Fig. 3.3b.

The retention factor [111, 112] and adsorption isotherm [113, 114] are

functions of temperature and are therefore affected by viscous heating. The

retention factor as a function of temperature is described by the van’t Hoff

equation

ln(k) � −∆HRT

+∆SR

+ ln

(FF0

)+ ln(k0) (3.7)

where H and S are the enthalpy and entropy of transfer of the solute

from the mobile phase to the stationary phase, respectively. Normally, the

retention decreases with increasing temperature in RPLC, i.e. the opposite

to pressure where the retention increases. Viscous heating affects retention

when the column is in adiabatic conditions since the temperature increase

is then the most severe (Fig. 3.3b). Since an increase in pressure increases

retention and an increase in temperature decreases retention, the net effect

for a certain solute is hard to predict and both a net increase and a net

decrease in retention has been observed [98, 115]

26

Page 42: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Chapter 4

Analytical Method Validation

In the manufacturing of a drug product, a control strategy is mandatory to

confirm the quality of the product. In the product control strategy, different

product quality attributes, such as purity and stability, are monitored with

an analytical procedure [27]. All analytical procedures used in the control

strategy are part of the drug application and are reviewed and approved

together with the manufacturing process by regulatory agencies. In the

drug application, all analytical procedures need to be shown to be suitable

for their intended purpose and this is done by validating that theymeet their

performance criteria [4, 116]. In the EU, the drug application for a new drug

product is made to the EMA which appoints two member states to review

the submission and after their approval, the new drug product may be

marketed in all member states. For a drug product marketed in the US, the

approval from the FDA is needed. After the patent has expired for a drug

product, official analytical procedures are published in pharmacopoeias

like the European Pharmacopoeia (EP) and the US Pharmacopoeia (USP).

Validation guidelines varies somewhat but they are all similar to the ones

given in ICH Q2 [4, 27, 117].

4.1 Method Performance Parameters

The quality of the data generated by an analytical method is specified in a

number of performance parameters [118]. When validating an analytical

method, the performance parameters are evaluated to confirm that they

meet specifications. Some performance parameters like accuracy and

reproducibility are only evaluated during the validation while others, like

chromatographic resolution, may also be included in a system suitability test

(SST) [5]. The purpose of a SST is to continuously monitor the performance

of the method to assure that it delivers data of sufficient quality.

27

Page 43: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

4.1.1 Specificity

Specificity is the ability of an analytical method to assess unequivocally the

analyte in the presence of components that are expected to be present in the

sample andmust therefore be assured before other performance parameters

can be validated [4, 119]. In liquid chromatography it means that the analyte

peak should be separated from all other peaks and it is usually assessed by

measuring the resolution factor between the analyte peak and its closest

neighbors. If co-elution is expected, it will also be necessary to test the

purity of the analyte peak with, for example, mass spectrometry. When

assessing specificity, relevant impurities and excipients can be spiked in

realistic concentrations to a sample containing the pure drug substance

and this approach was used in paper [IV] to assess specificity of a quality

control method for the drug product Nexium.

4.1.2 Precision

The precision of amethod is the variability or variance between independent

measurements of the same sample under prescribed conditions [4, 119]. The

variability of a method is made up of a number of different contributions

and is divided in four levels: system precision, repeatability, intermediate

precision and reproducibility. Since variances are additive, the higher levels

includes the contributions from the lower levels, e.g. reapeatibility includes

the variance from the system precision (Fig. 4.1).

Figure 4.1: Example of sources to variability in the different precision levels.

In liquid chromatography, system precision is assessed by repeated

injections of the same sample and describes the variability of the instrument

and is an important parameter in the SST. Repeatability is assessed by

28

Page 44: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

comparing independent test results for identical samples obtained under

identical condition (same equipment, operator, etc.) within a relatively

short time period. It is the sum of instrument variability and variability

from the sample preparation. The intermediate precision assesses the

variability in the procedure when done in the same lab, i.e. also accounting

for different analysts and instruments under a longer time period. Reprodu-

cibility is the variability between laboratories using different analysts and

different equipment when analyzing the same type of sample during an

extended period of time. It is important for methods intended for multiple

laboratories and for official methods reported in e.g. the USP or EP. In

liquid chromatography, precision is often measured as the relative standard

deviation in peak area. In paper [IV], the system precision was assessed in

the SST and the reproducibility was assessed in the validation by analyzing

the same sample in two different laboratories.

4.1.3 Accuracy

The accuracy of a method is how close a measured value is to a true or

accepted reference value and validation of the accuracy should focus on any

systematic bias [4, 119]. The accuracy of an assay of a drug substance should

preferably be done with an absolute measuring technique like titration or

quantitative NMR. For a drug product assay on the other hand, accuracy is

often assessed by measuring the recovery of a spiked sample with liquid

chromatography. Impurities are present in a much lower concentrations

compared to the API so the accuracy of impurity assays are often lower

than for the API. The accuracy was validated by assessing the recovery

of the API omeprazole and two impurities by analyzing spiked samples

containing a placebo matrix in paper [IV].

4.1.4 Linearity and Range

Linearity refers to the relationship between the measured signal and the

analyte concentration or amount [4, 119]. It is not mandatory that the

concentration is directly proportional to the signal, but it is the preferred

case since it is most practical. So, strictly speaking, the intended calibration

model should be assessed and not necessarily the linearity. The range

refers to the analyte concentrations intended for the method and depends

on the method’s purpose. For example, in the assay of a drug substance

the range is normally between 80 and 120% of the test concentration.

Linearity is evaluated in the concentration range by a calibration curve and

29

Page 45: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

statistical measures like the residual values from linear regression or the

confidence intervals from the y-intercept. For LC-UV, it is usually enough

to graphically evaluate the deviation of experimental data from the fitted

calibration curve. Although not a measurement of linearity itself [120], the

correlation coefficient, R2, is often given as support to an assessment of

linearity and was done so in paper [IV].

4.1.5 Detection and Quantification Limit

The detection limit of a method gives the lowest amount of analyte that can

be detected and the quantification limit gives the lowest amount of analyte

that can be quantified [4, 119]. Estimation of detection and quantification

limits can be done with different approaches but they are all based on the

variability of the method at very small analyte concentrations. In paper [IV],

the quantification limit was determined for the impurities in a drug product

based on the residual standard deviation (sr) of the calibration curve as

QL � 10sr/S where S is the slope of the calibration curve. The detection

limit can be determined with the same approach, i.e. DL � 3.3sr/S.

4.2 Robustness

The robustness of a method is its capacity to remain unchanged by small

variations in procedure-related factors such as instrument settings, sample

preparation etc. and it indicates how sensitive the method is to changes in

these factors [4, 121]. Robustness is separated from ruggedness in the USP

with ruggedness being the variation sensitivity in non-procedure-related

factors such as operator, lab etc. and is assessed with the reproducibility

[121].

To assess a method’s robustness, factors are varied in a deliberate way,

often using DoE, and the quantitative influence on performance parameters

is studied [44]. In the case of liquid chromatography, factors that are often

investigated in a robustness test is eluent pH and composition, temperature,

flow rate and stationary phase batch. The robustness test is often performed

directly after the method development, i.e. before the method validation

activities given in Section 4.1, in order to avoid repeating the method

validation if the method should fail the robustness test.

A 2-level fractional factorial design us suitable for robustness studies

since the number of factors are usually large and the range of the factors

limited (in small intervals, curvature and interaction terms can often be

neglected). Examples of common responses in liquid chromatography are

30

Page 46: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

resolution factor, tailing factor and retention factor of the first and last

eluting peaks. It can be advantageous to also include the responses in the

SST. Acceptance criteria are set for each response based on the required

performance and used to evaluate if the robustness is acceptable.

An example of a HPLC robustness assessment using DoE is given in

Fig. 4.2 which demonstrates four cases:

i The response is inside the acceptance criterion and the regression

model is insignificant. This is the ideal outcome which indicates that

the factors do not affect the response inside the design region. This

case is shown for the response impurity area in Fig. 4.2a where the

ANOVA p-value for the null hypothesis that there is no correlation

between the factors and the response is high and the variations in

impurity area are due to random noise.

ii The response is inside the acceptance criterion and the regression

model is significant. This outcome shows that the factors are affecting

the response and that a regression model can be established to

determine if the response is meeting the acceptance criterion in the

whole design region. This is the case for resolution factor and the

corresponding response surface, Fig. 4.2b, shows that it is above its

acceptance criterion in the design region.

iii The response is outside the acceptance criterion and the regression

model is significant. This outcome allows the regression model to

be used for determining a robust part of the original design region.

The retention factor in Fig. 4.2a has a significant regression model and

one experiment giving a value above the acceptance criterion. The

robust region can be determined from the confidence interval based

on the regression model’s variance and is shown in Fig. 4.2c as a

white, dashed line. Above this line the retention factor will be below

10 at 99% confidence level.

iv The response is outside the acceptance criterion and the regression

model is insignificant is the most problematic outcome. The response

is not robust in the design region and no regression model can be

obtained to determine a robust region. The tailing factor in Fig. 4.2a is

an example of this case and more experiments would be needed to

complete the evaluation of the robustness for this response.

31

Page 47: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Figure 4.2: A robustness test with three factors in a fractional factorial level III

design. a) Experimental results for the responses, the ANOVA p-value for the

model with insignificant model displayed in orange and acceptance criteria. b)

Response surface for the resolution factor where temperature was an insignificant

factor and c) response surface for the retention factor at 25◦C.

4.3 Analytical Quality by Design

Quality by Design (QbD) is a systematic approach to product development

wherein understanding the process is paramount and quality is built into

the process resulting in better robustness [29]. Researchers have suggested

that QbD principles can be applied also to the analytical procedures that are

part of the control strategy [34]. Development of an analytical procedure

based onQbD principles includes similar activities as for traditional method

development but are given in amore structuredmanner. Themaindifference

being that in analytical QbD the method performance is largely defined and

understood during the method development step instead of the validation

step [43, 44]. At the beginning of the method development, the method

goals and the performance required to achieve those goals are given in

an analytical target profile (ATP). The ATP specifies what to measure and

32

Page 48: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

acceptance criteria for accuracy, precision, etc.

When a suitable method has been established, a risk assessment is

performed to identify critical method parameters that have a large impact

on the method’s performance. Traditionally, a risk assessment is often done

informally to determine which factors to include in the robustness study.

QbD, on the other hand, promotes a more formal risk assessment where the

result is supplied in the drug application and tools like Ishikawa diagrams,

failure mode effect analysis or mechanistic modeling are employed. An

example of an Ishikawa diagram is given in Fig. 4.3 describing a number

of factors for an HPLC method that can affect its performance. In a failure

mode effect analysis, potential factors are ranked according to severity,

likelihood and detectability to determine their importance [122]. Both these

tools requires knowledge about the analytical procedure to be effective

which can be problematic if there is a lack of experience of the analytical

technique. Mechanistic modeling can then be valuable since the analysis can

be simulated under varying conditions to investigate how certain variables

affects the performance [123].

Figure 4.3: Example of an Ishikawa diagram used to list potentially critical factors

for risk assessment of a HPLC method.

The design space is denoted method operable design region (MODR)

when applied to an analytical method and is essentially a range of settings

for critical parameters, for example pH, temperature and gradient slope

in liquid chromatography, where the method is shown to meet all its

performance criteria. A MODR is often determined using DoE and a

wider factor range is used compared to a conventional robustness study

resulting in significant regression models [43, 44]. The understanding of

33

Page 49: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

how factors affects the responses are based on a mathematical model which

could be either mechanistic or statistical [124]. In the MODR, all acceptance

criteria are met and it is therefore allowed to change variables as long as the

method is operated inside the MODR. Finally a control strategy should be

established which ensures method performance each time the analytical

procedure is employed. The control strategy contains a SST, but the purpose

of the SST is not only to monitor the day-to-day performance of the method

but also to assess the method performance after intended changes. The

method development using the QbD concept is summarized in Fig. 4.4.

QbD promotes life cycle management in where the analytical method is

continuously improved. However, very few examples exists where this has

been realized and is one of the topics of paper [IV].

Figure 4.4: The steps in analytical method development withing the QbD frame-

work.

To appreciate the advantage of QbD, consider a quality control method

for batch release. If the method would fail, the drug cannot be released to

the market. So, if the original column brand becomes unavailable and the

new brand gives slightly worse performance, the success of the method

depends on its robustness. A traditional method may not be robust enough

to handle the reduced performance and the settings may be too tightly

specified to improve the performance. The method would need to be

redeveloped and a post approval change would have to be submitted the

regulatory agencies which could take months and in the meantime batches

will not be released. However, a QbD method would be more robust in the

first place and, if necessary, there is the possibility to do changes within the

MODR without regulatory interaction [43].

34

Page 50: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Chapter 5

Results and Discussion

Throughout paper [I-V], the API omeprazole has been investigated toget-

her with some of its impurities using a BEH C18 stationary phase from

Waters (Milford, MA, US) and a mobile phase consisting of acetonitrile

and phosphate buffer. In paper [I], the adsorption of omeprazole was

studied varying the pH and type of organic modifier and we show how

adsorption isotherm measurements can explain trends observed in a DoE

investigation. Paper [II-III] are concerned with the fundamental differences

between HPLC and UHPLC conditions, i.e. stationary phase chemistry,

pressure and viscous heating. Paper [IV] presents a case example of a

quality control method method being converted from HPLC to UHPLC as a

continual improvement and an approach for handling minor post-approval

changes with minimal regulatory interactions was developed. In paper [V]

an experimental method was developed to predict retention and selectivity

shifts due to the temperature and pressure gradients in UHPLC. Finally,

in paper [VI] the effect of pressure on the adsorption isotherm from a

fundamental point of view was investigated to explain some observations

made in the previous papers.

5.1 Paper I

In the development of an analytical procedure, DoE is often performed to

find the optimum operating conditions and in the validation step, DoE is an

essential part of the robustness study. Traditionally, the data generated in

DoE is modeled with a statistical regression model to find the relationship

between factors and responses. However, recent regulatory initiatives, like

QbD, encourage the understanding of the process or analytical procedure

and a statistical regression model may be inadequate to understand the

underlying reasons for the observed trends.

35

Page 51: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Paper [I] demonstrated how adsorption isotherm modeling can compli-

ment an empirical DoE investigation to explain how eluent pH, temperature

and type of organic modifier affects the separation of omeprazole from

two of its impurities. Adsorption isotherm models provide a valuable tool

for studying the adsorption mechanism in liquid chromatography from a

mechanistic perspective. The separationwas investigated in isocratic elution

with either acetonitrile or methanol as organic modifier. For each modifier,

an experimental designwith pH and temperature as factors were performed

with a sample containing omeprazole and the impurities H168/66 and

H193/61 in relevant concentrations for a quality control method.

Response surfaces from the statistical models displayed some interesting

trends (Fig. 5.1). Methanol and acetonitrile gave quite different trends in

both resolution factor (Rs), where methanol revealed a maximum resolution

at intermediate pH while acetonitrile gave highest resolution at high pH,

and tailing factor (Tf) where omeprazole went from tailing to fronting

with acetonitrile as the pH increased and was relatively unaffected with

methanol.

Figure 5.1: Response surface for a) the resolution factor between omeprazole and

H168/66 and b) the tailing factor for omeprazole with acetonitrile as the organic

modifier. c) Resolution factor and d) tailing factor with methanol as organic

modifier.

The resolution factor between omeprazole and H168/66 was affected by

pH because both compounds behave as weak acids in the this pH interval.

36

Page 52: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

By measuring the retention factor in a wide pH-interval it was evident

that when protonated, H168/66 has longer retention time than omeprazole

with acetonitrile and vice versa for methanol (Fig. 5.2). The crossing of

the lines in Fig. 5.2 represents a shift in elution order and is correlated to

the region where Rs � 0 in the response surface for acetonitrile (Fig. 5.1a).

Therefore, the difference in resolution with acetonitrile and methanol is

due to a selectivity difference for the protonated forms of H168/66 and

omeprazole.

6 8 10 12

s

wpH

0

2

4

6

8

10

k

a) MeCN

H168/66

Omeprazole

6 8 10 12

s

wpH

b) MeOH

Figure 5.2: The retention factor as a function ofs

wpH for omeprazole and H168/66

with a) acetontrile and b) methanol as organic modifier at 30◦C.

The fronting of omeprazole was, at least partially, the result of overloa-

ding since the peak became more symmetrical as the sample concentration

decreased. The shape of an overloaded peak is governed by its nonlinear

adsorption isotherm and first the adsorption isotherm of the protonated

form of omeprazole was obtained using the inverse method. The 2-layer

BETmodel, Eq. (2.12), was selected as the isothermmodel since it gave good

agreement with the experimental data both for acetonitrile and methanol.

Acetonitrile gave elution profiles consistent with a type III isotherm model

(diffuse front) while methanol gave elution profiles consistent with type

I (diffuse rear) as shown in Fig. 5.3a-b. This was reflected in the isotherm

parameters where the equilibrium constant for adsorption on the stationary

phase, bS, was about two times larger for methanol while the equilibrium

constant for adsorption on the solute layer, bL, was similar. The reason

for the weaker interaction with the stationary phase using acetonitrile was

believed to be the thick layer acetonitrile forms, compared to methanol,

at the stationary phase interface [125] which makes it more difficult for

omeprazole to get access to the stationary phase.

Since pH had a large influence on peak shape with acetonitrile, the

37

Page 53: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

adsorption isotherm for omeprazole was also modeled as a function of

pH. The design region includes the pKa of omeprazole, so the adsorption

isotherm needed to account for this. Additionally, the buffer capacity was

not high enough to keep the pH constant when overloaded samples for

the inverse method were injected and the local variations in pH when

the sample zone moved through the column was also accounted for. An

semi-empirical model based on the general Langmuir isotherm, denoted

the pH model, was employed and showed that the fronting increases with

the fraction of deprotonated omeprazole for acetonitrile (Fig. 5.3c). As the

pH increases, the deprotonated form increases which has a lower saturation

capacity than the protonated form. For methanol, this gives deformed

peaks at high pH (Fig. 5.3d).

0

0.5

1

1.5

2

2.5

Concentr

ation [g/L

] a) MeCN, pH = 7.6

Experiments

BET model

2 4 6 8

Time [min]

0

0.5

1

1.5

2

2.5

Concentr

ation [g/L

] c) MeCN, pH = 9.4

Experiments

pH model

0

1

2

3

4 b) MeOH, pH = 8.1

6 8 10 12 14

Time [min]

0

1

2

3

4 d) MeOH, pH = 9.5

Figure 5.3: Overloaded elution profiles at the a-b) lowest pH and c-d) highest pH

used in the experimental design obtained with either acetonitrile or methanol as

organic modifier at 30◦C.

To conclude, this paper investigated the adsorption mechanism for

omeprazole and how pH, temperature and type of organic modifier affected

it and how these effects translated to resolution and tailing. Adsorption

isotherm models were found to be valuable to understand the trends from

response surfaces obtained from DoE data.

38

Page 54: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

5.2 Paper II

An assumption made when changing the particle size and column dimensi-

ons in chromatography, is that the stationary phase is the same. However,

by comparing columns packed with supposedly the same stationary phase

but different particle size (1.7 µm and 5 µm) using the Tanaka characteri-

zation protocol, significant selectivity shifts were observed with certain

column brands [17]. The hydrogen bonding capacity and the silanol activity

were two parameters that varied with the particle size and the result was

large shifts in retention, especially for basic compounds. Furthermore, the

temperature is often higher in UHPLC than in HPLC either due to viscous

heating or a combination of viscous heating and a higher set temperature

of the column oven to reduce the pressure drop over the column. Since

the temperature affects the adsorption isotherm parameters and hence the

retention, saturation capacity and peak shape, the higher temperature in

UHPLC may affect the separation performance.

Therefore the aim of paper [II] was to compare the adsorption isotherms

obtained at 20◦C with a column packed with 3.5 µm particles (denoted

condition I) with those obtained at 40◦C with a column packed with 1.7 µm

particles (condition II) from the same brand of stationary phase to evaluate

the effect of these changes on the adsorption mechanism. Adsorption

isotherms were acquired for one neutral (cycloheptanone, C7), one anionic

(sodium 2-naphtalenesulfonate, SNS) and one cationic (benzyltriethylam-

monium chloride, BTEAC) compound for the two conditions. The flow rate

was kept low with the 1.7 µm particles to avoid temperature and pressure

gradients because they complicates adsorption isotherm determination.

Additionally, we wanted to avoid studying too many factors at the same

time and pressure and temperature gradients were the subject of paper

[III].The adsorption isotherms for BTEAC, C7 and SNS were determined

with the PP method and described by the bi-Langmuir model, Eq. (2.10)

(Table 5.1). The retention factor was lower in condition II for BTEAC

and SNS while it was, rather surprisingly, identical for C7. In RPLC, it is

expected that the retention factor decreases with temperature and the higher

temperature in condition II was believed to be the main reason for the lower

retention. The three compounds are affected differently by temperature,

with C7 being least affected according to additional experiments in paper

[III]. The total porosity was 0.60 in condition I and 0.55 for condition II,

i.e. the ratio between stationary phase and mobile phase was slightly

39

Page 55: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

higher with the 1.7 µm particles, which increases the retention factor in

condition II. Therefore, the identical retention factors for C7 was most likely

a coincidence of the decrease in retention due to higher temperature and

the increase in retention due to lower porosity canceling each other out.

The saturation capacity, which can be seen as a measure of the number of

possible adsorption sites, was very similar for the two conditions while the

equilibrium constant, related to the strength of the interactions, decreased in

condition II. The equilibrium constant is related to the temperature through

Eq. (2.14) and is expected to decrease with increasing temperature as was

the case here. Based in these observations, the nature of the adsorption

mechanism seems very similar in the two conditions and no additional

interactions with, for example, residual silanols were evident.

Table 5.1: Adsorption isotherm parameters obtained at two conditions; I 20◦C and

3.5 µm particles and II 40◦C and 1.7 µm particles.

Solute Condition k a1 qs,1 b1 a2 qs,2 b2

BTEAC

I 10.5 3.72 18.8 0.198 11.8 3.07 3.82

II 9.33 3.22 20.6 0.157 8.34 2.67 3.12

SNS

I 5.94 1.38 50.3 0.0274 7.39 3.57 2.07

II 4.12 1.04 48.7 0.0213 4.09 2.64 1.55

C7

I 4.21 3.93 40.2 0.0978 2.29 n.a. ≈ 0

II 4.21 3.56 51.7 0.0688 1.68 n.a. ≈ 0

The adsorption isotherm for omeprazole (∼ 80% in protonated form1)

was described by the BET model, Eq. (2.11). Due to the limited solubility

of omeprazole in the eluent, there is not much curvature of the isotherm

but it is evident from the overloaded peaks that a type III model like

BET is suitable (Fig. 5.4a-b). The AED displayed one adsorption site for

both conditions which indicates homogeneous adsorption in the studied

concentration interval (Fig. 5.4c). The variation of the isotherm parameters

with acetonitrile was investigated (Fig. 5.4d-f) and was found to follow the

same trends in both conditions. The a parameter shows that the retention

time is slightly lower in condition II and that the difference increased at low

acetonitrile fractions. bS decreased with acetonitrile which was expected

since omeprazole becomes more soluble in the eluent as the acetonitrile

fraction increases. bL shows a maximum around 22% acetonitrile and it

was speculated that at lower fractions omeprazole binds very strongly to

1The paper erroneously states that omeprazole was negatively charged.

40

Page 56: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

the stationary phase due to its low solubility in the eluent making it less

attractive to form multi-layers.

0 0.5 1 1.5 2 2.5

C [g/L]

0

10

20

q [

g/L

]

b)

-4 -3 -2 -1 0

ln(b)

0

2

4

qs [

g/L

]

c)

0.18 0.22 0.26 0.3

φ [v/v]

1

2

3

4

ln(a

)

d)

0.18 0.22 0.26 0.3

φ [v/v]

-9

-8

-7

-6

ln(b

S)

e)

0.18 0.22 0.26 0.3

φ [v/v]

-4

-3

-2

ln(b

L)

f)

3 4 5 6 7 8

k

0

1

2

3

C [

g/L

]

a)1.7 µm, 40°C

3.5 µm, 20°C

Figure 5.4: a) Overloaded omeprazole peaks normalized to retention factor. b)

Adsorption isotherms obtained with the ECP method at 25% acetonitrile. c) AED

calculations with the local BET model. d-f) Isotherm parameters in Eq. (2.11) as a

function of acetonitrile. Reprinted from D. Åsberg, et al., J. Chromatogr. A, 1362,206–207, Copyright 2014, with permission from Elsevier.

We concluded that the adsorption isotherm model did not change

between condition I and II, but the values of the isotherm parameters did

change slightly. This was true for a wide range of acetonitrile fractions

since the isotherm parameters followed the same trend in both conditions

implying that geometric scaling in gradient elution should result in preser-

ved method performance. Since only minor differences in retention and

saturation capacity were observed for this experimental system, we should

not expect any complications related to stationary phase properties when

switching from 3.5 µm BEH C18 particles to the 1.7 µm variant of the same

particles.

41

Page 57: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

5.3 Paper III

The elevated pressure and viscous heating in UHPLC may affect the perfor-

mance of a method when converting it from HPLC to UHPLC. The pressure

and temperature effects are convoluted under normal operation and only the

net effect is observed. In this paper we studied the individual contributions

from pressure and temperature to the overall change in retention and peak

shape. The peak symmetry as a function of pressure and temperature

was investigated with the stochastic model for BTEAC which was the only

compound displaying tailing at low concentrations. The pressure was

studied by using a low flow rate and a restrictor capillary installed after

the columns to avoid causing viscous heating. Different pressures were

obtained by varying the length of the capillary while keeping the flow rate

constant and the main part of the pressure drop was then in the capillary.

The retention factor of omeprazole and the samemodel compounds as in

paper [II] were determined at five pressures with the result for omeprazole

presented in Fig. 5.5a. The increase in retention for a 500 bar pressure

difference was about 40% for omeprazole which is quite significant2. A

20◦C increase in temperature resulted in a decrease in retention of around

20% for omeprazole (Fig. 5.5b). The temperature profile in the column was

calculatedwith the energy balance, Eq. (2.4), and validated through external

temperature sensors attached to the column surface. With the column in a

conventional still air oven, a 782 bar pressure drop over the column and

a flow rate of 1.2 mL/min, the temperature gradient in the axial direction

was 16◦C and the radial temperature gradient was 2

◦C at most (Fig. 5.5c).

The relative effect of pressure and temperature on retention for ome-

prazole and the three model compounds was estimated by comparing the

retention factors at flow rates 0.13 mL/min and 1.2 mL/min. In Fig. 5.5d

the retention factor at 0.13 mL/min is taken as the reference value where

both pressure and viscous heating are negligible. The estimated change in

retention due to pressure (+300 bar) and due to temperature (8.5 ◦C) are

shown together with the experimentally obtained net effect. Even though

the individual effects are relatively large, they cancel each other out for

C7 and SNS and there is only a minor shift in retention when comparing

the experimental data. However, for OM and BTEAC the pressure effect

dominates with the retention factor increasing 10-15% at 1.2 mL/min. Since

2Note that there is an error in the abstract of this paper; the percent of increase in

retention is for a 300 bar pressure increase (corresponding to Fig. 5 in paper [III]), not 500bar as is specified in the abstract.

42

Page 58: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

there are large differences between the compounds it stands to reason that

selectivity shifts could occur which could cause problems when switching

between HPLC to UHPLC conditions. A case where this happened was

later demonstrated in paper [V].

Figure 5.5: Change in the retention factor of omeprazole as a function of a) pressure

and b) temperature. c) The temperature profile inside the column at 1.2 mL/min

(pressure drop 782 bar) with the oven at 40◦C. d) The calculated change in k due

to pressure (gray) and viscous heating (orange) when increasing the flow rate from

0.13 mL/min to 1.2 mL/min. The blue bars represent the experimentally obtained

net effect. Reprinted from D. Åsberg, et al., J. Chromatogr. A, 1401, 52–59, Copyright2014, with permission from Elsevier.

BTEAC was tailing significantly even at low concentrations and the

elution profiles could be described by a 2-site stochastic model. The tailing

of a peak is related to the skew of the statistical distribution describing the

peak and the skew was studied as a function of temperature and pressure

for BTEAC (Fig. 5.6). The stochastic model revealed one adsorption site

with fast adsorption-desorption in the millisecond scale and n1 ≈ 10 000

which is common for symmetrical peaks in RPLC. The tailing was caused

by a second adsorption site which had slow adsorption-desorption with

the solute staying adsorbed up to several seconds before desorption. The

sojourn time, τs,2, decreased with temperature for this type of site (Fig. 5.6a)

probably due to a faster diffusion at high temperature and a decrease in the

43

Page 59: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

association equilibrium constant with temperature, Eq. (2.14). Increasing

the pressure resulted in a longer sojourn time (Fig. 5.6b) which can be

explained by results obtained in paper [VI] and will be discussed in relation

to that paper. The practical consequence is that the skew decreases with

temperature and increases with pressure which can be seen from the

experimental elution profiles in Fig. 5.6. That pressure could affect peak

symmetry had not been reported before and demonstrated that the pressure

effect is not completely understood in liquid chromatography.

30 35 40 45 50

Temperature [°C]

3

4

5

6

7

τs,2

[s]

a) Experiment

Linear fit

30 35 40 45 50

Temperature [°C]

1.25

1.3

1.35

1.4

Skew

c)

4 5 6 7 8

Time [min]

e)

30°C

40°C

50°C

0 200 400 600

Pressure [bar]

b)

0 200 400 600

Pressure [bar]

d)

5 6 7 8 9

Time [min]

f)

51 bar

301 bar

524 bar

Figure 5.6: Results from the stochastic model for BTEAC. The sojourn time for the

adsorption site with slow kinetic, τs,2, as a function of a) temperature (pressure ca

50 bar) and b) pressure (temperature 40◦C). The skew is shown in c) as a function

of temperature and in d) as a function of pressure. e) Chromatograms with varying

temperature and f) with varying pressure. Reprinted from D. Åsberg, et al., J.Chromatogr. A, 1401, 52–59, Copyright 2014, with permission from Elsevier.

44

Page 60: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

5.4 Paper IV

Quality control (QC) methods plays an important role in the overall control

strategy of drug manufacturing. However, efficient life-cycle management

with continual improvements of QC methods are hindered by the lack of an

internationally harmonized legislation for handling post-approval changes.

Currently, a change to a QC method need to be approved by all regulatory

agencies where the drug product is marketed before it can be implemented

which is very time consuming and may result in the QC methods falling

behind the technical development. Fortunately, the ICH Q12 guidelines

are under development and set to be released in 2017 with the purpose

to harmonize the post-approval legislation and facilitate efficient life-cycle

management of both the manufacturing process and analytical procedures.

The aim of this paper was to develop a concept exemplifying how a post-

approval change to a QCmethod could be handled in practice with minimal

interaction with regulatory agencies before implementation.

We based the concept on QbD principles, but the possibility to do

changes inside the original MODR was too limited since it is seldom

possible to predict what flexibility is needed for implementing future

technology. The idea was that for a certain analytical technique, the

specified performance criteria, failure modes and controls would be valid

also after a change and a revalidation would ensure the preserved quality

of the method. The change and revalidation data would be recorded within

the company and presented upon inspection in a “do and tell” manner.

We distinguish between an ATP as defined in the QbD framework as a set

of performance criteria that are independent of the analytical technique

(measuring principle) and a method ATP (mATP) which we define as a set

of performance criteria for a specific technique, e.g. liquid chromatography.

The concept is demonstrated by a case study in which a QC method for

the drug product Nexium based on HPLC is converted to UHPLC as a

continual improvement strategy.

5.4.1 Method Development

The objective of the analytical procedure is specified along with the pro-

perties that need to be measured. The objective in the case study was to

determine the impurity and the API content in the drug product Nexium.

To this end, the concentration of omeprazole and four impurities needed to

be measured in a Nexium sample with HPLC. A set of method performance

criteria were determined which should be met for the method to give data

45

Page 61: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

of sufficient quality. The mATP for the HPLC method is summarized

in Table 5.2 and includes criteria for specificity, accuracy, reproducibility,

quantification limit, linearity and relative response. After the mATP was

established and a suitable working point was found, it was validated and

the result is shown for omeprazole and one impurity in Table 5.2.

Table 5.2: mATP with method performance criteria and some results from the

HPLC method. The specificity was determined from the resolution between

the closest eluting peaks while the reproducibility was assessed by the relative

standard deviation of total impurity area for the same sample analyzed in two

laboratories.

Attribute Criteria Omeprazole H431/41

Specificity No interference Rs � 9.5 for closest peaks

Reproducibility ≥ 5.0% 2.56% in impurity area

Accuracy 90 − 110% 99.6% 103%

Quantification limit ≤ 0.05% 0.0085% 0.0064%

Linearity ≥ 0.9990 1.0000 1.0000

Relative response ≥ 0.7 1.00 1.29

5.4.2 Risk Assessment

The objective of the risk assessment is to identify critical factors that have a

serious impact on the method performance. Failure modes were identified

as the resolution between the critical peak pair being below 2, the tailing

factor of the main peak being outside the range 0.75-1.50, the first peak

eluting in the void and the retention factors for the last eluting peak being

above 30, corresponding to the method run time. Based on previous

experience of this type of sample, e.g. from paper [I], and experiments done

during the working point screening, the eluent pH, the eluent composition

and the column temperature was identified as critical factors. The failure

modes were included both in the robustness study of the MODR and in the

SST.

5.4.3 Method Validation

At this point, the method is known to meet the mATP criteria in the working

point and critical factors influencing the performance has been identified

along with appropriate failure modes. The main objective in the method

validation step is therefore to increase the knowledge of how the critical

46

Page 62: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

factors affect the failure modes and determine the MODR. An expanded

robustness study was undertaken using DoE and statistical modeling. The

response surfaces for the method’s failure modes are shown in Fig. 5.7.

When more than two factors were significant, the settings giving the worst

case are presented. Only the resolution factor was outside specifications

in the design region (shown as the white dotted line in Fig. 5.7). The

MODR was therefore pH = 7.65-8.35 and T = 15.0-25.0◦C and the method

is specified in Table 5.3 with the tolerance levels for the the critical factors.

As for the understanding of the relationship between the critical factors

and the investigated responses, the trends are evident from Fig. 5.7 while

the physical explanation is discussed in depth in paper [I]. The resolutionfactor was largely governed by the difference in pKa between omeprazole

and H168/66 while the fronting of omeprazole was due to a larger fraction

of omeprazole becoming deprotonated at high pH.

Table 5.3: The HPLC method given with the tolerance levels of the critical factors.

Column BEH C18, 3.5 µm, 4.6 × 100 mm

Buffer Phosphate, pH 8.0±0.35, 10 mM (A) and 3 mM (B)

Eluent A 10/90±5, v/v, acetonitrile/phophate bufferEluent B 80/20±5, v/v, acetonitrile/phophate bufferDetection UV 302 nm

Gradient Linear from 0 to 100% B in 30 min

Gradient dwell time 1.04 min

Flow rate 1.00 mL/min

Temperature 20±5◦CInjection volume 20 µL

5.4.4 Control Strategy

The control strategy ensures adequate performance of themethod during its

lifetime, typically in the form of a system suitability test (SST). By carefully

constructing the SST so that it is also applicable outside the original MODR,

it can be used to assess the failure modes after a change to the method.

When converting the method to UHPLC we move outside the original

MODR and the SST should ensure method performance and generation of

reliable results.

The SST for the case study was taken to include the responses in the

robustness study as they were related to the failure modes and the system

47

Page 63: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Figure 5.7: Response surfaces for the HPLC robustness study. a) The resolution

factor between omeprazole and H168/66 with the robust region (the lower 95%-

confidence limit of the predicted resolution factor > 2) above the white dotted line.

b) The tailing factor of omeprazole. c) The apparent retention factor for H431/41

which is the first eluting peak. d) The apparent retention factor for H168/22 which

is the last eluting peak. Reprinted from D. Åsberg, et al., J. Pharm. Biomed. Anal.,129, 273–281, Copyright 2016, with permission from Elsevier.

repeatability. The omeprazole peak was slightly fronting, with a tailing

factor of 0.93, and the resolution factor between omeprazole and H168/66

was 9.5 which is well above the acceptance criteria.

5.4.5 Continual Improvement

As a continual improvement of the method, the column is exchanged to an

UHPLC column (1.7 µm, 2.1 × 50 mm) and the temperature is increased

from 20◦C to 40

◦C. The UHPLC column will increase the throughput by

shortening the analysis time while the higher temperature will reduce the

pressure and increase the resolution between omeprazole and H168/66

(Fig. 5.7a). This change is outside the original MODR and would normally

require the submission of a post-approval change to the regulators before

implementing it. However, what is proposed in the new concept is that the

change would be implemented directly and managed according to internal

change control procedures and presented upon inspection. The original

48

Page 64: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

submissionwould specify how re-validation andmethod equivalencewould

be handled after a minor change.

The original method was geometrically scaled to give the same separa-

tion with the new column and the resulting chromatograms are compared

in Fig. 5.8. From paper [II] we know that the adsorption mechanism are

almost unaffected when changing the particle size of the stationary phase.

When performing the geometrical scaling and keeping the temperature the

same, Fig. 5.8a, the retention increased slightly in the modified method

most likely due to a pressure increase [III]. However, when also increasing

the temperature, Fig. 5.8b, the retention factors decreased somewhat and

the selectivity between omeprazole and H168/66 improved. The analysis

time was reduced by a factor 4 and the solvent consumption was reduced by

a factor 10 when switching to UHPLC. The mATP needs to be re-validated

No

rma

lize

d R

esp

on

se

a)HPLC

UHPLC

0 2 4 6 8 10 12 14 16

Apparent retention factor

No

rma

lize

d R

esp

on

se

b)

Figure 5.8: Chromatograms from the original and modified QC method for a)

the same oven temperature, 30◦C, and b) the temperatures specified in the QC

methods, i.e. 20◦C and 40

◦C. Reprinted from D. Åsberg, et al., J. Pharm. Biomed.

Anal., 129, 273–281, Copyright 2016, with permission from Elsevier.

for the modified method and this was achieved with similar results as for

the original method. The failure modes are equally valid for the modified

method and was not changed. Next, the MODR was adjusted based on the

settings of the modified method as center point by repeating the robustness

study. Since the resolution had improved, all of the design region was

49

Page 65: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

now robust. The SST is still relevant for testing the failure modes and

the modified method passed the SST. As a test for method equivalence,

the original and modified methods were demonstrated to give the same

impurity area in the center point and the extreme points of the experimental

design.

5.4.6 Conclusions

The proposed concept enables implementation of minor post approval chan-

ges within the same measuring principle without prior regulatory approval.

ThemATP, failure modes and controls given in the original drug application

are valid also for the modified method and a complete revalidation of

these elements are performed within the company’s pharmaceutical quality

system and the result archived for future regulatory inspections. If the

presented concept will be reality it means that the pharmaceutical industry

can save time and resources and still maintain high quality QC-methods

while it reduces the burden for regulatory agencies.

5.5 Paper V

We investigated the effect of elevated pressure and viscous heating in paper

[III] and showed that it could complicate method conversion between

HPLC to UHPLC. Here, a case where the selectivity, and hence resolution,

between omeprazole and the impurity H168/66 is seriously affected by

pressure and temperature gradients is presented. At the flow rate 0.08

mL/min omeprazole and H168/66 co-elutes while increasing the flow rate

to 0.50 mL/min results in baseline separation (Fig. 5.9a). This selectivity

shift is mainly due to a change in temperature caused by viscous heating

which is evident from an experimental design in pressure and temperature

space (Fig. 5.9b). In this case, the flow rate could be identified as a critical

factor when, in reality, it is temperature that is the underlying cause of the

selectivity shift.

The aim of this paper was to develop an easy-to-use, empirical method

to predict retention time shifts due to pressure and temperature gradients

inside the column. Such a method would be a valuable tool in routine met-

hod development to assess method robustness and aid method conversion

between HPLC to UHPLC. The strategy was based on the assumption that

by putting the column in a water jacket the generated heat was effectively

removed and the temperature could be kept nearly constant regardless of

flow rate. This assumption was investigated by calculating the temperature

50

Page 66: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Figure 5.9: The effect of flow rate on the selectivity between omeprazole (largest

peak) and H168/66. a) Chromatograms obtained at three flow rates giving average

column pressures of 93, 300 and 484 bar. The column oven was set to 308 K. b) The

response surface for selectivity in pressure and temperature space using a restrictor

capillary to adjust the pressure. Reprinted from D. Åsberg, et al., J. Chromatogr. A,1479, 107–120, Copyright 2017, with permission from Elsevier.

profiles, in the same way as in paper [III], with the column kept in a still

air oven and in a water jacket. In still air, the axial temperature gradient

dominated and the temperature increased about 10◦C from the inlet to the

outlet of the column (Fig. 5.10a). When keeping the column in the water

jacket, the radial temperature gradient dominated and was about 0.7 ◦C

resulting in a case where the column could be controlled within ±1◦C

(Fig. 5.10b). In agreement with Fig. 5.9, no change in selectivity with flow

rate was observed when keeping the column in the water jacket since the

temperature could be kept nearly constant. There is also a clear difference

between water and air thermostating when plotting the retention factor as a

function of average column pressure controlled by the flow rate. In a water

bath, the increase in retention factor is solely due to the increase in pressure

while in the still air oven the retention factor increases more moderately

since an axial temperature gradient is also present (Fig. 5.10c-d). This is

another approach than the one in paper [III] to de-couple the pressure and

temperature effects in UHPLC.

The empirical approach describes the retention factor as a function of

average pressure, Pavg, and the average temperature, Tavg, in the column as

k(T, P) � a1(Tavg − Tavg,0) + a2(Pavg − Pavg,0) + k0 (5.1)

where ai denotes empirical fitting parameters and subscript 0 denotes a

reference condition where the pressure drop over the column is around 100

51

Page 67: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Figure 5.10: Calculated temperature profiles in a) still air (522 bar) and b) water

jacket (494 bar) for the BEH column. Normalized retention factor for c) omeprazole

and d) H168/66. Dashed line is a linear fit to the experimental data (symbols).

Reprinted from D. Åsberg, et al., J. Chromatogr. A, 1479, 107–120, Copyright 2017,with permission from Elsevier.

bar. The DoE investigation in pressure-temperature space confirmed that

Eq. (5.1) is a valid approximation of how the retention factor varies. Pavg

and Tavg are functions of flow rate:

Pavg(Fv) � a3(Fv − Fv,0) + Pavg,0 (5.2)

Tavg(Fv) � a4(Fv − Fv,0) + Tavg,0 (5.3)

The ai parameters have to be determined experimentally in four experiments:

i The first experiment is used to describe a reference condition where

the pressure and temperature gradients are negligible. It is an injection

at a low flow rate where the back pressure is around 100 bar.

ii The second experiment is for determining how the retention factor

depends on temperature. It is an injection at low flow rate, as in (i),

but with the column oven set to a temperature 20◦C higher than in

the first experiment.

52

Page 68: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

iii The third experiment is for determining how the retention factor

depends on pressure. It is an injection at high flow rate (back pressure

around 1000 bar) with the column placed in a water jacket. The

water jacket minimizes the axial temperature gradient and keeps the

temperature ±1◦C so the pressure effect can be measured.

iv The final experiment estimates the pressure and the average column

temperature as a function of flow rate with the column in the standard

still air oven. It is done at high flow rate using the column oven with

the same set temperature as for the reference condition in step (i).

The empirical approach was validated by predicting the retention factor for

omeprazole and three impurities at four flow rates and with two columns

(one with fully porous particles and one with core-shell particles). Rather

surprisingly, considering the approximations done deriving it, the relative

error between the predicted and experimental retention factor was always

less than 1% and in many cases below 0.5%. This level of accuracy is

enough to predict the selectivity shifts observed between omeprazole and

H168/66 (Fig. 5.11). To predict the elution profiles, the column efficiency

was estimated with linear interpolation of the elution profiles in step (i) and

(iv) which was found to be good enough.

7.6 7.8 8 8.2 8.4 8.6

Time [min]

a) 0.2 mL/min

Experiment

Prediction

3.4 3.6 3.8

Time [min]

b) 0.5 mL/min

Figure 5.11: Predicted and experimental chromatograms for a) 0.20 mL/min and

b) 0.50 mL/min for omeprazole and H168/66. Reprinted from D. Åsberg, et al., J.Chromatogr. A, 1479, 107–120, Copyright 2017, with permission from Elsevier.

The empirical approach has the potential to be a practical tool in routine

method development, but there are still some parts left to investigate. While

gradient elution is very common in pharmaceutical analysis, the empirical

approach in its present form assumes isocratic elution where the eluent

viscosity is constant during the run. Extending it to gradient elution would

53

Page 69: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

not be straightforward since there will exist different cases depending on

the viscosity of eluent A and B and was therefore outside the scope of this

paper. Solutes, like proteins and large peptides, which are more sensitive to

pressure and temperature would also be interesting to try in the empirical

approach to see if the linear relationship in Eq. (5.1) still holds.

5.6 Paper VI

A retentionmechanism in RPLC taking the pressure into account hasmainly

been studied by measuring the retention factor at different pressure levels.

However, in such measurements it is difficult to obtain information about

secondary interactions between the solute and the stationary phase which

are often responsible for tailing peaks. With nonlinear adsorption isotherm

measurements on the other hand, it is possible to elucidate secondary

interactions.

The aim of this paper was to determine adsorption isotherms at different

pressures ranging from 100 to 1000 bar and measure how the different

adsorption sites contribute to the overall pressure dependence of the solute.

More specifically, we were interested in why ionic solutes are more affected

by pressure than neutral solutes and why BTEAC in paper [III] became

more tailing as the pressure increased. To this end, a similar approach as

the one in paper [II] was employed where the adsorption isotherms were

determined for three compounds; one cation (BTEAC), one anion (SNS) and

one neutral (antipyrine, AP), but here the pressure was varied instead. AP

replaced cycloheptanone since it had similar molecular weight to BTEAC

and SNS. The pressure was controlled by the same approach as in paper

[III], i.e. by varying the length of a restrictor capillary attached after the

column while keeping the flow rate constant.

The total porosity of the stationary phase is an important property when

deriving adsorption isotherms from experimental data and it was therefore

measured at all pressure levels. A change from εt � 0.552 to εt � 0.560 was

noted when the pressure increased from 100 to 900 bar mostly because of a

compression of the C18-bonded layer of the stationary phase. A variation

in total porosity in this range is negligible for most routine applications

but should be accounted for when determining adsorption isotherms. All

three compounds were found to be best described by the bi-Langmuir

adsorption isotherm when evaluating different models following a rigorous

approach with Scatchard plots, AED, model selection and verification by

predicting elution profiles and comparing them to experimental ones. For

54

Page 70: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

AP, the saturation capacity for the high energy site was around 1% of

the total saturation capacity and the equilibrium constant was about ten

times higher compared to the low energy site. For BTEAC and SNS, the

saturation capacity for the high energy site was instead around 10% of the

total saturation capacity but the equilibrium constant was still about ten

times higher for the high energy site.

Two adsorption sites prevailed for all pressure levels in the AED

(Fig. 5.12) which indicates that the adsorption isotherm model per se doesnot change with pressure, only the numerical parameters do. The ad-

sorption isotherms tended towards a slightly increased saturation capacity

and initial slope with increasing pressure. Based on previous studies (see

Section 2.3) we interpreted the low energy site as adsorption located at

the eluent-C18 layer interface while the high energy represents adsorption

deeper into the C18 layer. Some trends were evident from the isotherm pa-

rameters, although their exact values should be viewed critically since they

are obtained through nonlinear regression which can yield multiple sets of

parameters giving the same functional values. The association equilibrium

constant increases with increasing pressure for both sites most likely due

to the increased pressure driving the equilibrium between the mobile and

stationary phase towards the stationary phase to reduce the solute molar

volume in accordance with Eq. (3.5). The saturation capacity of the low

energy site is almost unchanged, which is to be expected since the surface

area of the stationary phase is not affected by pressure. The saturation

capacity of the high energy site increased with pressure and molecular

simulations have demonstrated an increased de-wetting of a C18 stationary

phase with increasing pressure [126]. We speculated that the increase in

saturation capacity was caused by a de-wetting of the stationary phase

which made adsorption sites near the silica surface, previously occupied by

water, available [127].

The change in partial molar volume of the solute associated with the

adsorption of each of the two sites were derived from the adsorption

isotherms using a combination of Eq. (3.5) and Eq. (2.9) for each site. For

the low energy site the solute molar volume was practically the same for the

three compounds and equal to ∆Vlow ≈ −6 cm3/mol. For SNS and BTEAC,

the high energy site had almost twice as large volmue change while AP

displayed a 3.6 times larger volume change. Based on these results, a more

nuanced retention mechanism accounting for pressure was proposed. Polar

and ionic solutes will have a surrounding polar solvation layer in the mobile

phase and lose parts of this layer when adsorbing on the stationary phase.

55

Page 71: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

3 4 5 6 7 8 9 10 11 12

0

2

4

6

8

10

12q

s [

mM

]a)

6 7 80

0.1

0.2

0 1 2 30

20

40

60

q [

mM

]

b)

109 bar

316 bar

462 bar

612 bar

919 bar

4 5 6 7 8 9 10 11 12 13

0

0.2

0.4

0.6

0.8

1

qs [

mM

]

c)

0 0.5 10

2

4

6

8

10

12

q [

mM

]

b)

88 bar

263 bar

378 bar

508 bar

808 bar

4 5 6 7 8 9 10 11 12 13

ln(b)

0

0.5

1

1.5

2

qs [

mM

]

e)

7 8 90

0.1

0.2

0 0.5 1

C [mM]

0

2

4

6

8

10

12

q [

mM

]

f)

107 bar

311 bar

445 bar

590 bar

900 bar

Figure 5.12: AED and adsorption isotherm for a-b) AP, c-d) BTEAC and e-f) SNS.

Reprinted from D. Åsberg, et al., J. Chromatogr. A, 1457, 97–106, Copyright 2016,with permission from Elsevier.

It is the loss of this solvation layer that causes the change in the solute’s

molar volume. When the solute adsorbs on the low energy site located near

the C18 surface, only a small fraction of the solvation layer is lost resulting

in a small reduction of the molar volume. Adsorption on the high energy

sites, on the other hand, forces the solutes to penetrate into the C18 layer

56

Page 72: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

and a larger portion of the solvation layer is lost making the molar volume

of the solute decrease more (Fig. 5.13). So, a solute with more adsorption

events at the high energy sites will have a retention factor that are more

sensitive to pressure.

Returning to paper [III], we are now better equipped to understandwhy

BTEAC exhibited increased tailing at higher pressures. We can rule out a

decreasing saturation capacitywith pressure based Fig. 5.12 and calculations

presented in paper [VI] showed no reduction in column efficiency with

pressure due to overloading. If we assume that the nature of the adsorption

sites observed in the stochastic modeling and in the adsorption isotherm

model are of similar nature, then the increase in sojourn time for the high

energy site (Fig. 5.6b) can be correlated to the increase in the association

equilibrium constant with pressure for this site. To conclude, an increase in

pressure causes the equilibrium constant for the high energy site to increase

which in turn results in a longer sojourn time in this type of site. The longer

sojourn type increased the skew of the peak and hence a larger tailing factor

was observed.

Figure 5.13: Schematic illustration of adsorption from the mobile phase on to

the stationary phase for a low-energy site at the eluent-C18 layer interface (left)

and a high-energy site near the silica surface (right). The decrease of ∆V for the

solute upon adsorption on the high-energy site is explained by the larger loss of

its solvation layer when it penetrates deeper into the C18 layer. Reprinted from

D. Åsberg, et al., J. Chromatogr. A, 1457, 97–106, Copyright 2016, with permission

from Elsevier.

57

Page 73: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

58

Page 74: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Chapter 6

Concluding Remarks

UHPLC, with its higher throughput and lower solvent consumption, has

been a great leap forward in the evolution of liquid chromatography.

However, the implementation of UHPLC in pharmaceutical analysis has not

been straightforward and there are both technical and regulatory challenges

to overcome. The goal of the thesis has been to study UHPLC in depth and

thereby solve these challenges. My approach was to implement adsorption

isotherms and mechanistic modeling to complement conventional DoE and

statistical regression models to satisfy the trend towards a more science-

based approach to pharmaceutical method development. I demonstrated

that the adsorption mechanism and the surface heterogeneity were not

seriously affected when switching from a HPLC particle size to a UHPLC

particle size using adsorption isotherm modeling. This is an important

finding since the assumption that the stationary phase remains unaltered is

a requirement for reliable geometrical scaling between particle sizes.

Elevated pressure and temperature gradients due to viscous heating are

the two most important factors to account for to achieve predictable method

conversion between HPLC and UHPLC. Their effect on retention and peak

shape was quantified and studied from a fundamental point of view to

investigate how they affected the adsorption mechanism and separation

performance. I found that a secondary, high energy, type of adsorption site

was the main reason for the pure pressure effect. Ionic solutes adsorbed to

a larger degree on the secondary sites and were hence more influenced by

pressure than neutral solutes. I also demonstrated that the combination

of pressure and viscous heating could cause selectivity shifts since the

retention of solutes, even with similar structure, were affected differently.

Therefore, an easy-to-use, experimental approach intended for method

development was developed to predict these selectivity shifts. Together,

these studies provides a better understanding of UHPLC and demonstrates

59

Page 75: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

for the regulatory agencies an approach for studying and controlling critical

factors in UHPLC.

To change from HPLC to UHPLC in the control strategy of a marketed

drug products, a post approval change would need to be submitted and

approved by the regulatory agencies, which is time consuming. Together

with AstraZeneca, I developed a concept to tackle this issue and proposed

an approach for handling minor post approval changes such as this one

without prior regulatory approval. The idea was that as long as the same

analytical technique is used, the method controls (SST and mATP) and

failure modes would be relevant also for settings not included in the original

submission and by re-validating the controls, the method performance

would not be compromised.

The current ICH Q12 draft document provides a framework for post

approval changes in a more predictable and efficient manner across the pro-

ducts life cycle by making it more feasible to do changes with a downgraded

reporting category under the company’s pharmaceutical quality system

[128]. By demonstrating a good product and process knowledge, only

critical input and output parameters affecting the process performance and

product quality need regulatory approval prior to a change, i.e. noncritical

parameters may be changed without prior regulatory interactions. The

elements in the submission that may not be changed without approval

should include a method description with the critical aspects for the in-

tended performance (similar to the mATP) rather than detailed operating

conditions, along with parameters verifying the method continue being fit

for purpose (a SST). Changes to analytical methods for currently marketed

products can also be handled under ICH Q12 in a manner strikingly similar

to that presented paper [IV]. The method is shown to be fit for purpose

by revalidating it according to ICH Q2 (i.e. revalidating the mATP), the

SST is verified and the results are shown to be equivalent or better. If this

is done the change can be made with downgraded regulatory reporting.

AstraZeneca has already begun implementing such an approach for ana-

lytical methods in some of their drug applications, for example with the

drug product naloxegol (Movantik) which was recently approved by the

FDA and the EMA.

Drug manufacturing will be relaying more on science and risk-based

approaches in the coming years where the final quality of the product is put

in focus. I hope that this thesis can provide support for this trend and the

implementation of the ICH Q12 guideline while enabling implementation

of new technology, such as UHPLC, in pharmaceutical analysis.

60

Page 76: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Acknowledgment

Many people have supported me throughout my work with this thesis and

I am very grateful to

my main supervisor Torgny Fornstedt and my assistant supervisor Jörgen

Samuelsson for your support, inspiration and all our fruitful discussions,

Anders Karlsson for initializing this project and patiently teaching me about

the pharmaceutical industry,

Martin Enmark for great cooperation and nice company in Uppsala,

Krzysztof Kaczmarski andMarek Leśko for a long and fruitful collaboration,

all of my coauthors for your effort, expertise and insightful comments,

Mikael Andersen for your invaluable help in the lab and

mypast andpresent colleagues at the department for your pleasant company.

61

Page 77: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

62

Page 78: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

References

[1] S. Cox Gad. Pharmaceutical Manufacturing Handbook: Regulations andQuality. Hoboken: Wiley, 2008. doi: 10.1002/9780470259832

[2] The European Union. Commission Directive 91/507/EEC. OfficialJournal of the European Communities, 34: 32–52, 1991.

[3] S. Görög. The changing face of pharmaceutical analysis. TrAC, 26:12–17, 2007. doi: 10.1016/j.trac.2006.07.011

[4] International Council for Harmonisation. Q2(R1): Validation of Ana-lytical Procedures: Text and Methodology. Nov. 2005. url: www.ich.org

[5] C. F. Richardson and F. Erni. “Regulatory Considerations in HPLC

Analysis”. In: Handbook of Pharmaceutical Analysis by HPLC. Ed. byM.W. Dong and S. Ahuja. Vol. 6. Separation Science and Technology.

Academic Press, 2005. doi: 10.1016/S0149-6395(05)80054-6

[6] R. Holm and D. P. Elder. Analytical advances in pharmaceutical

impurity profiling. Eur. J. Pharm. Sci., 87: 118–135, 2016. doi: 10.1016/

j.ejps.2015.12.007

[7] J. De Vos, K. Broeckhoven, and S. Eeltink. Advances in Ultrahigh-

Pressure Liquid Chromatography Technology and System Design.

Anal. Chem., 88: 262–278, 2016. doi: 10.1021/acs.analchem.5b04381

[8] B. Debrus, E. Rozet, P. Hubert, J.-L. Veuthey, S. Rudaz, and D.

Guillarme. “Method Transfer Between Conventional HPLC and

UHPLC”. In: UHPLC in Life Sciences. Ed. by D. Guillarme and J.-

L. Veuthey. The Royal Society of Chemistry, 2012. doi: 10.1039/

9781849735490

[9] G. K. Webster, T. F. Cullen, and L. Kott. “Method Transfer Between

HPLC and UHPLC Platforms”. In: Ultra-High Performance LiquidChromatography and its Applications. Ed. by Q. A. Xu. Wiley, 2013. doi:

10.1002/9781118533956

63

Page 79: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[10] S. Fekete, J.-L. Veuthey, and D. Guillarme. Comparison of the most

recent chromatographic approaches applied for fast and high reso-

lution separations: Theory and practice. J. Chromatogr. A, 1408: 1–14,2015. doi: 10.1016/j.chroma.2015.07.014

[11] J. R. Mazzeo, U. D. Neue, M. Kele, and R. S. Plumb. Advancing LC

performance with smaller particles and higher pressure.Anal. Chem.,77: 460 A–467 A, 2005. doi: 10.1021/ac053516f

[12] M. W. Dong and K. Zhang. Ultra-high-pressure liquid chromato-

graphy (UHPLC) in method development. TrAC, 63: 21–30, 2014.doi: 10.1016/j.trac.2014.06.019

[13] J. Cielecka-Piontek, P. Zalewski, A. Jelińska, and P. Garbacki. UHPLC:

The Greening Face of Liquid Chromatography. Chromatographia, 76:1429–1437, 2013. doi: 10.1007/s10337-013-2434-6

[14] S. A. C. Wren and P. Tchelitcheff. Use of ultra-performance liquid

chromatography in pharmaceutical development. J. Chromatogr. A,1119: 140–146, 2006. doi: 10.1016/j.chroma.2006.02.052

[15] S. Fekete, J. Schappler, J.-L. Veuthey, and D. Guillarme. Current and

future trends in UHPLC. TrAC, 63: 2–13, 2014. doi: 10.1016/j.trac.

2014.08.007

[16] D. V. McCalley. The impact of pressure and frictional heating on

retention, selectivity and efficiency in ultra-high-pressure liquid

chromatography. TrAC, 63: 31–43, 2014. doi: 10.1016/j.trac.2014.06.

024

[17] P. Petersson and M. R. Euerby. Characterisation of RPLC columns

packed with porous sub-2 µm particles. J. Sep. Science, 30: 2012–2024,2007. doi: 10.1002/jssc.200700086

[18] C. G. Horvath, B. A. Preiss, and S. R. Lipsky. Fast liquid chromato-

graphy. Investigation of operating parameters and the separation of

nucleotides on pellicular ion exchangers. Anal. Chem., 39: 1422–1428,1967. doi: 10.1021/ac60256a003

[19] N. Tanaka and D. V. McCalley. Core–Shell, Ultrasmall Particles,

Monoliths, andOther SupportMaterials inHigh-PerformanceLiquid

Chromatography. Anal. Chem., 88: 279–298, 2016. doi: 10.1021/acs.

analchem.5b04093

[20] G. Guiochon and F. Gritti. Shell particles, trials, tribulations and

triumphs. J. Chromatogr. A, 1218: 1915–1938, 2011. doi: 10.1016/j.

chroma.2011.01.080

64

Page 80: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[21] J. Ruta, D. Zurlino, C. Grivel, S. Heinisch, J.-L. Veuthey, and D.

Guillarme. Evaluation of columns packed with shell particles with

compounds of pharmaceutical interest. J. Chromatogr. A, 1228: 221–231, 2012. doi: 10.1016/j.chroma.2011.09.013

[22] M. Biba, C. J. Welch, J. P. Foley, B. Mao, E. Vazquez, and R. A. Arvary.

Evaluation of core–shell particle columns for ion-pair reversed-phase

liquid chromatography analysis of oligonucleotides. J. Pharm. Biomed.Anal., 72: 25–32, 2013. doi: 10.1016/j.jpba.2012.09.007

[23] J. Fibigr, D. Šatínský, L. Havlíková, and P. Solich. A new method

for rapid determination of indole-3-carbinol and its condensation

products in nutraceuticals using core–shell column chromatography

method. J. Pharm. Biomed. Anal., 120: 383–390, 2016. doi: 10.1016/j.

jpba.2015.12.039

[24] J. Fibigr, D. Šatínský, and P. Solich. A study of retention characte-

ristics and quality control of nutraceuticals containing resveratrol

and polydatin using fused-core column chromatography. J. Pharm.Biomed. Anal., 120: 112–119, 2016. doi: 10.1016/j.jpba.2015.12.014

[25] J. W. Ludvigsson, A. Karlsson, and V. Kjellberg. Core–shell co-

lumn Tanaka characterization and additional tests using active

pharmaceutical ingredients. J. Sep. Sci., 39: 4520–4532, 2016. doi:

10.1002/jssc.201600769

[26] R. W. Bondi Jr. and J. K. Drennen III. “Quality by Design and the

Importance of PAT in QbD”. In: Handbook of Modern PharmaceuticalAnalysis. Ed. by S. Scypinski and S. Ahuja. Vol. 10. Separation Science

and Technology. Academic Press, 2011. doi: 10.1016/B978-0-12-

375680-0.00005-X

[27] M.M.Dantus andM.L.Wells. Regulatory Issues inChromatographic

Analysis in the Pharmaceutical Industry. J. Liq. Chromatogr. RelatedTechnol., 27: 1413–1442, 2004. doi: 10.1081/JLC-120030610

[28] U.S Food and Drug Administration. Pharmaceutical cGMPs for the21st Century – A Risk-Based Approach. Sept. 2004. url: www.fda.gov

[29] International Council for Harmonisation. Q8(R2): PharmaceuticalDevelopment. Aug. 2009. url: www.ich.org

[30] International Council for Harmonisation. Q9: Quality Risk Manage-ment. Nov. 2005. url: www.ich.org

[31] International Council forHarmonisation.Q10: Pharmaceutical QualitySystem. June 2008. url: www.ich.org

65

Page 81: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[32] International Council for Harmonisation. Q11: Development andManufacture of Drug Substances. May 2012. url: www.ich.org

[33] L. X. Yu, G. Amidon, M. A. Khan, S. W. Hoag, J. Polli, G. K. Raju,

and J. Woodcock. Understanding Pharmaceutical Quality by Design.

AAPS J., 16: 771–783, 2014. doi: 10.1208/s12248-014-9598-3

[34] F. G. Vogt and A. S. Kord. Development of quality-by-design analyti-

cal methods. J. Pharm. Sci., 100: 797–812, 2011. doi: 10.1002/jps.22325

[35] J. Kochling, W. Wu, Y. Hua, Q. Guan, and J. Castaneda-Merced. A

platform analytical quality by design (AQbD) approach for multiple

UHPLC-UV and UHPLC–MS methods development for protein

analysis. J. Pharm. Biomed. Anal., 125: 130–139, 2016. doi: 10.1016/j.

jpba.2016.03.031

[36] L. Taevernier, E. Wynendaele, M. D’Hondt, and B. De Spiegeleer.

Analytical quality-by-design approach for sample treatment of BSA-

containing solutions. J. Pharm. Anal., 5: 27–32, 2015. doi: 10.1016/j.

jpha.2014.06.001

[37] H. I. Mokhtar, R. A. Abdel-Salam, and G. M. Hadad. Design Space

Calculation by In Silico Robustness Simulation with Modeling Error

Propagation in QbD Framework of RP-HPLC Method Development.

Chromatographia, 78: 457–466, 2015. doi: 10.1007/s10337-015-2858-2

[38] R. Mallik, S. Raman, X. Liang, A. W. Grobin, and D. Choudhury.

Development andvalidation of a rapidultra-highperformance liquid

chromatography method for the assay of benzalkonium chloride

using a quality-by-design approach. J. Chromatogr. A, 1413: 22–32,2015. doi: 10.1016/j.chroma.2015.08.010

[39] C. Boussès, L. Ferey, E. Vedrines, andK. Gaudin. Using an innovative

combination of quality-by-design and green analytical chemistry

approaches for the development of a stability indicating UHPLC

method in pharmaceutical products. J. Pharm. Biomed. Anal., 115:114–122, 2015. doi: 10.1016/j.jpba.2015.07.003

[40] Document EMA/430501/2013. EMA-FDA pilot program for parallelassessment of Quality-by-Design applications: lessons learnt and Q&Aresulting from the first parallel assessment. Aug. 20, 2013. url: www.

ema.europa.eu/ema/

[41] U.S Food and Drug Administration. Guidance for Industry – ProcessValidation: General Principles and Practices. Jan. 2011. url: www.fda.

gov

66

Page 82: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[42] P. Borman, K. Truman,D. Thompson, P.Nethercote, andM.Chatfield.

The Application of Quality by Design to Analytical Methods. Pharm.Technol., 31: 142–152, 2007.

[43] D. K. Lloyd and J. Bergum. “Application of Quality by Design (QbD)

to the Development and Validation of Analytical Methods”. In:

Specification of Drug Substances and Products. Ed. by C. M. Riley, T. W.

Rosanske, and S. R. Rabel Riley. Elsevier, 2014. doi: 10.1016/B978-0-

08-098350-9.00003-5

[44] R. LoBrutto. “Method Design and Understanding”. In: Method Vali-dation in Pharmaceutical Analysis. Ed. by J. Ermer and P.W.Nethercote.

2:nd. Wiley, 2015. doi: 10.1002/3527604685

[45] International Council for Harmonisation. Final Concept Paper – Q12:Technical and Regulatory Considerations for Pharmaceutical ProductLifecycle Management. July 2014. url: www.ich.org

[46] J. Rantanen and J. Khinast. The Future of Pharmaceutical Manufac-

turing Sciences. J. Pharm. Sci., 104: 3612–3638, 2015. doi: 10.1002/jps.

24594

[47] U.S Food and Drug Administration. Guidence for Industry – Changesto an Approved NDA or ANDA. Apr. 2004. url: www.fda.gov

[48] The European Union. Information and Notices 2013/C 223/01.

Official Journal of the European Communities, 56: 1–79, 2003.

[49] N. Wu, C. J. Welch, T. K. Natishan, H. Gao, T. Chnadrasekaran,

and L. Zhang. “Practical Aspects of Ultrahigh Performance Liquid

Chromatography”. In: Ultra-High Performance Liquid Chromatographyand its Applications. Ed. by Q. A. Xu. Wiley, 2013. doi: 10.1002/

9781118533956

[50] L. Olbe, E. Carlsson, and P. Lindberg. A proton-pump inhibitor

expedition: the case histories of omeprazole and esomeprazole. Nat.Rev. Drug Discov., 2: 132–139, 2003. doi: 10.1038/nrd1010

[51] F. Gritti and G. Guiochon. Critical contribution of nonlinear chroma-

tography to the understanding of retention mechanism in reversed-

phase liquid chromatography. J. Chromatogr. A, 1099: 1–42, 2005. doi:

10.1016/j.chroma.2005.09.082

[52] R. K. Lindsey, J. L. Rafferty, B. L. Eggimann, J. I. Siepmann, and

M. R. Schure. Molecular simulation studies of reversed-phase liquid

chromatography. J. Chromatogr. A, 1287: 60–82, 2013. doi: 10.1016/j.

chroma.2013.02.040

67

Page 83: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[53] F. Gritti and G. Guiochon. Mass transfer kinetics, band broadening

and column efficiency. J. Chromatogr. A, 1221: 2–40, 2012. doi: 10.

1016/j.chroma.2011.04.058

[54] D. Hlushkou, F. Gritti, A. Daneyko, G. Guiochon, and U. Tallarek.

How Microscopic Characteristics of the Adsorption Kinetics Impact

Macroscale Transport in Chromatographic Beds. J. Phys. Chem. C,117: 22974–22985, 2013. doi: 10.1021/jp408362u

[55] G. Guiochon, A. Felinger, D. G. Shirazi, andA.M. Katti. Fundamentalsof Preparative and Nonlinear Chromatography. 2:nd. Academic Press,

2006.

[56] A. Felinger. Molecular dynamic theories in chromatography. J.Chromatogr. A, 1184: 20–41, 2008. doi: 10.1016/j.chroma.2007.12.066

[57] P. V. Danckwerts. Continuous flow systems. Chem. Eng. Sci., 2: 1–13,1953. doi: 10.1016/0009-2509(53)80001-1

[58] K. Kaczmarski, M. Mazzotti, G. Storti, and M. Morbidelli. Modeling

fixed-bed adsorption columns through orthogonal collocations on

moving finite elements. Comput. Chem Eng., 21: 641–660, 1997. doi:

10.1016/S0098-1354(96)00300-6

[59] P. Brown,G. Byrne, andA.Hindmarsh. VODE:AVariable-Coefficient

ODE Solver. SIAM J. Sci. and Stat. Comput., 10: 1038–1051, 1989. doi:

10.1137/0910062

[60] K. Kaczmarski, J. Kostka, W. Zapała, and G. Guiochon. Modeling

of thermal processes in high pressure liquid chromatography: I.

Low pressure onset of thermal heterogeneity. J. Chromatogr. A, 1216:6560–6574, 2009. doi: 10.1016/j.chroma.2009.07.020

[61] K. Kaczmarski, F. Gritti, J. Kostka, and G. Guiochon. Modeling

of thermal processes in high pressure liquid chromatography: II.

Thermal heterogeneity at very high pressures. J. Chromatogr. A, 1216:6575–6586, 2009. doi: 10.1016/j.chroma.2009.07.049

[62] J. Kostka, F. Gritti, K. Kaczmarski, and G. Guiochon. Modified

Equilibrium-DispersiveModel for the interpretation of the efficiency

of columns packed with core–shell particle. J. Chromatogr. A, 1218:5449–5455, 2011. doi: 10.1016/j.chroma.2011.06.019

[63] K. Kaczmarski and G. Guiochon. Modeling of the Mass-Transfer Ki-

netics in Chromatographic Columns Packedwith Shell and Pellicular

Particles. Anal. Chem., 79: 4648–4656, 2007. doi: 10.1021/ac070209w

68

Page 84: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[64] J. Kostka, F. Gritti, G. Guiochon, and K. Kaczmarski. Modeling of

thermal processes in very high pressure liquid chromatography

for column immersed in a water bath: Application of the selected

models. J. Chromatogr. A, 1217: 4704–4712, 2010. doi: 10 .1016/j .

chroma.2010.05.018

[65] F. Gritti, W. Piatkowski, and G. Guiochon. Comparison of the ad-

sorption equilibrium of a few low-molecular mass compounds on

a monolithic and a packed column in reversed-phase liquid chro-

matography. J. Chromatogr. A, 978: 81–107, 2002. doi: 10.1016/S0021-

9673(02)01279-7

[66] F. Gritti and G. Guiochon. Influence of a buffered solution on the

adsorption isotherm and overloaded band profiles of an ionizable

compound. J. Chromatogr. A, 1028: 197–210, 2004. doi: 10.1016/j.

chroma.2003.11.106

[67] S. Brunauer, L. S. Deming, W. E. Deming, and E. Teller. On a Theory

of the van der Waals Adsorption of Gases. J. Am. Chem. Soc., 62:1723–1732, 1940. doi: 10.1021/ja01864a025

[68] A. Seidel-Morgenstern. Experimental determination of single solute

and competitive adsorption isotherms. J. Chromatogr. A, 1037: 255–272, 2004. doi: 10.1016/j.chroma.2003.11.108

[69] J. Samuelsson, T. Undin, A. Törncrona, and T. Fornstedt. Impro-

vement in the generation of adsorption isotherm data in the elution

by characteristic points method—The ECP-slope approach. J. Chro-matogr. A, 1217: 7215–7221, 2010. doi: 10.1016/j.chroma.2010.09.004

[70] E. V. Dose, S. Jacobson, andG.Guiochon. Determination of isotherms

from chromatographic peak shapes. Anal. Chem., 63: 833–839, 1991.doi: 10.1021/ac00008a020

[71] K. Kaczmarski. Estimation of adsorption isotherm parameters with

inverse method—Possible problems. J. Chromatogr. A, 1176: 57–68,2007. doi: 10.1016/j.chroma.2007.08.005

[72] B. J. Stanley and G. Guiochon. Numerical estimation of adsorp-

tion energy distributions from adsorption isotherm data with the

expectation-maximization method. J. Phys. Chem., 97: 8098–8104,1993. doi: 10.1021/j100132a046

69

Page 85: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[73] B. J. Stanley, S. E. Bialkowski, and D. B. Marshall. Analysis of

first-order rate constant spectra with regularized least-squares and

expectationmaximization. 1. Theory and numerical characterization.

Anal. Chem., 65: 259–267, 1993. doi: 10.1021/ac00051a013

[74] F. Gritti and G. Guiochon. Heterogeneity of the Adsorption Mecha-

nism of Low Molecular Weight Compounds in Reversed-Phase

Liquid Chromatography. Anal. Chem., 78: 5823–5834, 2006. doi:

10.1021/ac060392d

[75] U. D. Neue. “Silica Gel and its Derivatization for Liquid Chromato-

graphy”. In: Encyclopedia of Analytical Chemistry. Ed. by R. A. Meyer.

Wiley, 2009. doi: 10.1002/9780470027318.a5915.pub2

[76] L. Kisley and C. F. Landes. Molecular Approaches to Chromato-

graphy Using Single Molecule Spectroscopy. Anal. Chem., 87: 83–98,2015. doi: 10.1021/ac5039225

[77] L. C. Sander, K. A. Lippa, and S. A. Wise. Order and disorder in

alkyl stationary phases. Anal. Bioanal. Chem., 382: 646–668, 2005. doi:

10.1007/s00216-005-3127-2

[78] F. Gritti and G. Guiochon. Effect of the Surface Heterogeneity of

the Stationary Phase on the Range of Concentrations for Linear

Chromatography. Anal. Chem., 77: 1020–1030, 2005. doi: 10.1021/

ac040163w

[79] J. L. Rafferty, L.Zhang, J. I. Siepmann, andM.R. Schure. RetentionMe-

chanism in Reversed-Phase Liquid Chromatography: A Molecular

Perspective.Anal. Chem., 79: 6551–6558, 2007. doi: 10.1021/ac0705115

[80] A. Cavazzini, M. Remelli, F. Dondi, and A. Felinger. Stochastic

Theory of Multiple-Site Linear Adsorption Chromatography. Anal.Chem., 71: 3453–3462, 1999. doi: 10.1021/ac990282p

[81] J. C. Giddings and H. Eyring. A Molecular Dynamic Theory of

Chromatography. J. Phys. Chem., 59: 416–421, 1955. doi: 10.1021/

j150527a009

[82] M. J. Wirth and D. J. Swinton. Single-Molecule Probing of Mixed-

Mode Adsorption at a Chromatographic Interface. Anal. Chem., 70:5264–5271, 1998. doi: 10.1021/ac980632s

[83] M. J. Wirth, M. D. Ludes, and D. J. Swinton. Spectroscopic Obser-

vation of Adsorption to Active Silanols. Anal. Chem., 71: 3911–3917,1999. doi: 10.1021/ac990382v

70

Page 86: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[84] Z. Zhong, M. Lowry, G. Wang, and L. Geng. Probing Strong Ad-

sorption of Solute onto C18-Silica Gel by Fluorescence Correlation

Imaging and Single-Molecule Spectroscopy under RPLC Conditions.

Anal. Chem., 77: 2303–2310, 2005. doi: 10.1021/ac048290f

[85] J. T. Cooper, E. M. Peterson, and J. M. Harris. Fluorescence Ima-

ging of Single-Molecule Retention Trajectories in Reversed-Phase

Chromatographic Particles. Anal. Chem., 85: 9363–9370, 2013. doi:

10.1021/ac402251r

[86] L. Pasti, N. Marchetti, R. Guzzinati, M. Catani, V. Bosi, F. Dondi,

A. Sepsey, A. Felinger, and A. Cavazzini. Microscopic models of

liquid chromatography: From ensemble-averaged information to

resolution of fundamental viewpoint at single-molecule level. TrAC,81: 63–68, 2016. doi: 10.1016/j.trac.2015.08.007

[87] J. N. Mabry, M. J. Skaug, and D. K. Schwartz. Single-Molecule Insig-

hts into Retention at a Reversed-Phase Chromatographic Interface.

Anal. Chem., 86: 9451–9458, 2014. doi: 10.1021/ac5026418

[88] D. T. T. Nguyen, D. Guillarme, S. Rudaz, and J.-L. Veuthey. Chroma-

tographic behaviour and comparison of column packed with sub-2

µm stationary phases in liquid chromatography. J. Chromatogr. A,1128: 105–113, 2006. doi: 10.1016/j.chroma.2006.06.069

[89] M. Martin and G. Guiochon. Effects of high pressure in liquid

chromatography. J. Chromatogr. A, 1090: 16–38, 2005. doi: 10.1016/j.

chroma.2005.06.005

[90] D. Guillarme, D. T. T. Nguyen, S. Rudaz, and J.-L. Veuthey. Method

transfer for fast liquid chromatography in pharmaceutical analysis:

Application to short columns packed with small particle. Part I:

Isocratic separation. Eur. J. Pharm. Biopharm., 66: 475–482, 2007. doi:

10.1016/j.ejpb.2006.11.027

[91] D. Guillarme, D. T. T. Nguyen, S. Rudaz, and J.-L. Veuthey. Method

transfer for fast liquid chromatography in pharmaceutical analysis:

Application to short columns packed with small particle. Part II:

Gradient experiments. Eur. J. Pharm. Biopharm., 68: 430–440, 2008.doi: 10.1016/j.ejpb.2007.06.018

[92] L. R. Snyder and J. W. Dolan. High-Performance Gradient Elution.Hoboken: Wiley, 2007. doi: 10.1002/0470055529

71

Page 87: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[93] M. M. Fallas, U. D. Neue, M. R. Hadley, and D. V. McCalley. Inves-

tigation of the effect of pressure on retention of small molecules

using reversed-phase ultra-high-pressure liquid chromatography. J.Chromatogr. A, 1209: 195–205, 2008. doi: 10.1016/j.chroma.2008.09.021

[94] M. M. Fallas, U. D. Neue, M. R. Hadley, and D. V. McCalley. Further

investigations of the effect of pressure on retention in ultra-high-

pressure liquid chromatography. J. Chromatogr. A, 1217: 276–284,2010. doi: 10.1016/j.chroma.2009.11.041

[95] M. M. Fallas, N. Tanaka, S. M. C. Buckenmaier, and D. V. Mc-

Calley. Influence of phase type and solute structure on changes

in retention with pressure in reversed-phase high performance

liquid chromatography. J. Chromatogr. A, 1297: 37–45, 2013. doi:

10.1016/j.chroma.2013.04.006

[96] S. Fekete, J.-L. Veuthey, D. V. McCalley, and D. Guillarme. The effect

of pressure and mobile phase velocity on the retention properties

of small analytes and large biomolecules in ultra-high pressure

liquid chromatography. J. Chromatogr. A, 1270: 127–138, 2012. doi:

10.1016/j.chroma.2012.10.056

[97] M. R. Euerby, M. A. James, and P. Petersson. The influence of

stationary phase on pressure-induced retention, selectivity and

resolution changes in RP-LC. Anal. Bioanal. Chem., 405: 5557–5569,2013. doi: 10.1007/s00216-013-6973-3

[98] L. Nováková, J. L. Veuthey, and D. Guillarme. Practical method

transfer from high performance liquid chromatography to ultra-high

performance liquid chromatography: The importance of frictional

heating. J. Chromatogr. A, 1218: 7971–7981, 2011. doi: 10 .1016/j .

chroma.2011.08.096

[99] F. Gritti and G. Guiochon. Influence of the pressure on the properties

of chromatographic columns: I. Measurement of the compressibility

of methanol–water mixtures on a mesoporous silica adsorbent. J.Chromatogr. A, 1070: 1–12, 2005. doi: 10.1016/j.chroma.2005.02.007

[100] F. Gritti, M. Martin, and G. Guiochon. Influence of pressure on the

properties of chromatographic columns: II. The column hold-up

volume. J. Chromatogr. A, 1070: 13–22, 2005. doi: 10.1016/j.chroma.

2005.02.008

72

Page 88: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[101] F. Gritti and G. Guiochon. Influence of the pressure on the properties

of chromatographic columns: III. Retention volume of thiourea,

hold-up volume, and compressibility of the C18-bonded layer. J.Chromatogr. A, 1075: 117–126, 2005. doi: 10.1016/j.chroma.2005.03.095

[102] N. Tanaka, T. Yoshimura, and M. Araki. Effect of pressure on solute

retention in the ionization control mode in reversed-phase liquid

chromatography. J. Chromatogr. A, 406: 247–256, 1987. doi: 10.1016/

S0021-9673(00)94033-0

[103] V. L. McGuffin and C. E. Evans. Influence of pressure on solute

retention in liquid chromatography. J. Microcolumn Sep., 3: 513–520,1991. doi: 10.1002/mcs.1220030606

[104] V. L. McGuffin, C. E. Evans, and S.-H. Chen. Direct examination of

separation processes in liquid chromatography: Effect of temperature

and pressure on solute retention. J. Microcolumn Sep., 5: 3–10, 1993.doi: 10.1002/mcs.1220050102

[105] V. L. McGuffin and S.-H. Chen. Molar enthalpy and molar volume

of methylene and benzene homologues in reversed-phase liquid

chromatography. J. Chromatogr. A, 762: 35–46, 1997. doi: 10.1016/

S0021-9673(96)00958-2

[106] M. Martin and G. Guiochon. Pressure dependence of diffusion

coefficient and effect on plate height in liquid chromatography. Anal.Chem., 55: 2302–2309, 1983. doi: 10.1021/ac00264a023

[107] U. D. Neue andM. Kele. Performance of idealized column structures

under high pressure. J. Chromatogr. A, 1149: 236–244, 2007. doi:

10.1016/j.chroma.2007.03.042

[108] F. Gritti and G. Guiochon. Complete Temperature Profiles in Ultra-

High-Pressure Liquid Chromatography Columns. Anal. Chem., 80:5009–5020, 2008. doi: 10.1021/ac800280c

[109] A. d. Villiers, H. Lauer, R. Szucs, S. Goodall, and P. Sandra. Influence

of frictional heating on temperature gradients in ultra-high-pressure

liquid chromatography on 2.1 mm I.D. columns. J. Chromatogr. A,1113: 84–91, 2006. doi: 10.1016/j.chroma.2006.01.120

[110] F. Gritti, M. Martin, and G. Guiochon. Influence of Viscous Friction

Heating on the Efficiency of Columns Operated under Very High

Pressures. Anal. Chem., 81: 3365–3384, 2009. doi: 10.1021/ac802632x

73

Page 89: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[111] L. A. Cole and J. G. Dorsey. Temperature dependence of retention in

reversed-phase liquid chromatography. 1. Stationary-phase conside-

rations. Anal. Chem., 64: 1317–1323, 1992. doi: 10.1021/ac00037a004

[112] L. A. Cole, J. G. Dorsey, and K. A. Dill. Temperature dependence

of retention in reversed-phase liquid chromatography. 2. Mobile-

phase considerations. Anal. Chem., 64: 1324–1327, 1992. doi: 10.1021/

ac00037a005

[113] F. Gritti and G. Guiochon. Adsorption Mechanisms and Effect of

Temperature in Reversed-Phase Liquid Chromatography. Meaning

of the Classical Van’t Hoff Plot in Chromatography. Anal. Chem., 78:4642–4653, 2006. doi: 10.1021/ac0602017

[114] I. Quiñones, J. C. Ford, and G. Guiochon. Multisolute adsorption

equilibria in a reversed-phase liquid chromatography system. Chem.Eng. Sci., 55: 909–929, 2000. doi: 10.1016/S0009-2509(99)00393-0

[115] S. Fekete, J. Fekete, and D. Guillarme. Estimation of the effects of

longitudinal temperature gradients caused by frictional heating on

the solute retention using fully porous and superficially porous

sub-2 µm materials. J. Chromatogr. A, 1359: 124–130, 2014. doi: 10.

1016/j.chroma.2014.07.030

[116] U.S Food and Drug Administration.Guidance for Industry – AnalyticalProcedures and Methods Validation for Drugs and Biologics. July 2015.

url: www.fda.gov

[117] G. A. Shabir. Validation of high-performance liquid chromatography

methods for pharmaceutical analysis: Understanding the differences

and similarities between validation requirements of the US Food and

Drug Administration, the US Pharmacopeia and the International

Conference on Harmonization. J. Chromatogr. A, 987: 57–66, 2003.doi: 10.1016/S0021-9673(02)01536-4

[118] I. Taverniers,M.De Loose, and E. Van Bockstaele. Trends in quality in

the analytical laboratory. II. Analyticalmethodvalidation andquality

assurance. TrAC, 23: 535–552, 2004. doi: 10.1016/j.trac.2004.04.001

[119] J. Ermer. “Method Performance Characteristics”. In: Method Valida-tion in Pharmaceutical Analysis. Ed. by J. Ermer and P. W. Nethercote.

2:nd. Wiley, 2015. doi: 10.1002/3527604685

[120] P. Araujo. Key aspects of analytical method validation and linearity

evaluation. J. Chromatogr. B, 877: 2224–2234, 2009. doi: 10.1016/j.

jchromb.2008.09.030

74

Page 90: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

[121] B. Dejaegher and Y. V. Heyden. Ruggedness and robustness testing. J.Chromatogr. A, 1158: 138–157, 2007. doi: 10.1016/j.chroma.2007.02.086

[122] H.-C. Liu, L. Liu, and N. Liu. Risk evaluation approaches in failure

mode and effects analysis: A literature review. Expert Syst. Appl., 40:828–838, 2013. doi: 10.1016/j.eswa.2012.08.010

[123] E. Stocker, G. Toschkoff, S. Sacher, and J. G. Khinast. Use of me-

chanistic simulations as a quantitative risk-ranking tool within the

quality by design framework. Int. J. Pharm., 475: 245–255, 2014. doi:

10.1016/j.ijpharm.2014.08.055

[124] D. M. Hallow, B. M. Mudryk, A. D. Braem, J. E. Tabora, O. K.

Lyngberg, J. S. Bergum, L. T. Rossano, and S. Tummala. An Example

of Utilizing Mechanistic and Empirical Modeling in Quality by

Design. J. Pharm. Innov., 5: 193–203, 2010. doi: 10.1007/s12247-010-

9094-y

[125] F. Gritti and G. Guiochon. Adsorption Mechanism in RPLC. Effect

of the Nature of the Organic Modifier. Anal. Chem., 77: 4257–4272,2005. doi: 10.1021/ac0580058

[126] J. L. Rafferty, J. I. Siepmann, and M. R. Schure. The effects of

chain length, embedded polar groups, pressure, and pore shape on

structure and retention in reversed-phase liquid chromatography:

Molecular-level insights fromMonte Carlo simulations. J. Chromatogr.A, 1216: 2320–2331, 2009. doi: 10.1016/j.chroma.2008.12.088

[127] F. Chan, L. S. Yeung, R. LoBrutto, andY.V.Kazakevich. Interpretation

of the excess adsorption isotherms of organic eluent components

on the surface of reversed-phase phenyl modified adsorbents. J.Chromatogr. A, 1082: 158–165, 2005. doi: 10.1016/j.chroma.2005.05.078

[128] International Council for Harmonisation. Working Draft Version 6.0 –Q12: Technical and Regulatory Considerations for Pharmaceutical ProductLifecycle Management. Oct. 2016

75

Page 91: Fundamental and - Diva1073458/FULLTEXT02.pdfsnarlik struktur vilka ofta har en okänd fysiologisk e ekt. Därför görs alltid en kvalitetskontroll av läkemedlets renhet där mängden

Fundamental and Regulatory Aspects of UHPLC in Pharmaceutical Analysis

Dennis Åsberg

Dennis Å

sberg | Fundamental and R

egulatory Aspects of U

HPLC

in Pharmaceutical A

nalysis | 2017:9

Fundamental and Regulatory Aspects of UHPLC in Pharmaceutical Analysis

Ultra-high performance liquid chromatography (UHPLC) provides a considerable increase in throughput compared to conventional HPLC and a reduced solvent consumption. The implementation of UHPLC in pharmaceutical analysis has accelerated in recent years and currently both instruments are used. There are, however, technical and regulatory challenges converting a HPLC method to UHPLC making it difficult to take full advantage of UHPLC in regulatory-focused applications like quality control in pharmaceutical production. In UHPLC, the column is packed with smaller particles than in HPLC resulting in higher pressure and viscous heating. Both the higher pressure and the higher temperature may cause changes in retention and selectivity making method conversion unpredictable.

Using chromatographic modelling and fundamental theory, this thesis investigates method conversion between HPLC and UHPLC. It reports on the influence of temperature gradients due to viscous heating, pressure effects and stationary phase properties on the separation performance. It also presents a regulatory concept for less regulatory interaction for minor changes to approved quality control methods and how predicable method conversion is achieved by improved understanding.

DOCTORAL THESIS | Karlstad University Studies | 2017:9

Faculty of Health, Science and Technology

Chemistry

DOCTORAL THESIS | Karlstad University Studies | 2017:9

ISSN 1403-8099

ISBN 978-91-7063-757-5 (pdf)

ISBN 978-91-7063-756-8 (print)