development of a hghab-based surface acoustic wave …

79
UNIVERSITY GHENT FACULTY OF PHARMACEUTICAL SCIENCES Department Pharmaceutical Analysis Laboratory for Drug Quality and Registration Academic year 2013-2014 DEVELOPMENT OF A HGHAB-BASED SURFACE ACOUSTIC WAVE METHOD FOR THE FUNCTIONAL QUALITY CHARACTERIZATION OF NOTA-MODIFIED SOMATROPINS Sophia BARHDADI Master of Industrial Pharmacy Promoter: Prof. Dr. B. De Spiegeleer

Upload: others

Post on 18-Dec-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

UNIVERSITY GHENT

FACULTY OF PHARMACEUTICAL SCIENCES

Department Pharmaceutical Analysis

Laboratory for Drug Quality and Registration

Academic year 2013-2014

DEVELOPMENT OF A HGHAB-BASED SURFACE ACOUSTIC

WAVE METHOD FOR THE FUNCTIONAL QUALITY

CHARACTERIZATION OF NOTA-MODIFIED SOMATROPINS

Sophia BARHDADI

Master of Industrial Pharmacy

Promoter:

Prof. Dr. B. De Spiegeleer

COPYRIGHT

"The author and the promoters give the authorization to consult and to copy parts of this

thesis for personal use only. Any other use is limited by the laws of copyright, especially

concerning the obligation to refer to the source whenever results from this thesis are cited.”

Promoter Author

Prof. Dr. Bart De Spiegeleer Sophia Barhdadi

ACKNOWLEDGEMENTS

I would like to make use of this opportunity to thank all the people who helped me to bring

this thesis to a successful end.

First of all, I would like to thank Prof. Bart De Spiegeleer for giving me the opportunity to do

my research internship in the DruQuar laboratory, but especially for his guidance and for

pushing me in the right direction for my thesis.

I am also very grateful to Nathalie Bracke for supervising me throughout this past year, for

her endless patience and for kindly answering my many questions. Without your time and

energy, I never would have been able to finish this work. It was a pleasure to work with you

and I wish you all the best with the rest of your PhD.

Furthermore, I want to thank all the lovely people from the lab. They were always there for

support or just a simple talk. These little things helped to create a pleasant work environment.

Thank you to Sarah and Antoon, for your friendship and for the unforgettable moments we

shared in DruQuar.

Last but not least, I would like to thank all my friends and family for their continuous support.

A special thank goes to my parents, who gave me the chance to obtain an additional Master

degree and for always believing in me.

ABSTRACT

Introduction

Somatropin (i.e. recombinant human growth hormone, rhGH) is a biologic drug, currently

approved to treat growth hormone deficiencies. Recently, studies indicate a possible role of

hGH in certain cancers; hence, the oncologic potential of somatropin and analogues/modified

proteins are currently under investigation. Modifications with chelating agents like 1,4,7-

triazacyclononane-1,4,7-triacetic acid (NOTA) allows the incorporation of radiometals for

SPECT/PET-diagnostic (67Ga, 68Ga, 111In) or therapeutic (90Y) purposes. Somatropin has nine

potential binding sites (Lys) for NOTA. Therefore, it is important to characterize the obtained

product under different specified synthesis procedures like the excess of NOTA (1:1, 3:1 and

10:1 molar based) added to somatropin.

Objective

The main objective of this paper is to develop a hGHAb-based SAW method in order to

perform a functional quality characterization of NOTA-modified somatropins.

Methods and Results

Five different ligand immobilisation techniques were explored and compared in terms of

ligand immobilisation density, binding capability and specificity. The covalent amine

immobilisation of somatratopin was found the best immobilisation method (59 pg/mm2

surface-bound ligand, 66.7% RR% and 1.5% NSB%). An appropriate regeneration procedure

was also developed: a mixture of 50 nM NaOH and 0.1% SDS completely removed the bound

hGHAb from the immobilised somatropin surface, while preserving the ligand ‘s activity. The

developed SAW method was used to determine the binding affinities and kinetics of NOTA-

modified somatropins for hGHAb. A KD in sub-nanomolar range was observed for

somatropin and NOTA-modified somatropins.

Conclusion

The NOTA-modifications do not alter the binding affinity of somatropin toward hGHAb. The

hGHAb-based SAW method was successfully used as a system to investigate at least part of

the functional quality of NOTA-modified somatropins. Binding to hGHAb does not

automatically correlate with the binding potency to hGHR expressing tumour cells. These

questions are currently further being explored.

SAMENVATTING

Inleiding

Somatropine (recombinant humaan groeihormoon, rhGH) is een biologisch geneesmiddel, dat

goedgekeurd is voor de behandeling van groeihormoon deficiënties. Recente studies tonen

aan dat humaan groeihormoon een belangrijke rol kan spelen in de ontwikkeling van bepaalde

tumoren. Vandaar dat het oncologische potentieel van somatropine en analogen momenteel

wordt onderzocht. Somatropine werd geconjugeerd met een chelator zoals 1,4,7-

triazacyclononaan-1,4,7-triazijnzuur (NOTA) zodat het radioactief gelabeld kan worden voor

SPECT/PET-diagnostische (67Ga, 68Ga, 111In) en therapeutische (90Y) doeleinden. Het is

daarom belangrijk om de verkregen eindproducten, die gesynthetiseerd werden onder

verschillende omstandigheden (1:1, 3:1 en 10:1 NOTA overmaat), te karakteriseren

Objectieven

Het doel van deze thesis is het ontwikkelen van een hGHAb-gebaseerd SAW methode, die

gebruikt kan worden voor de functionele kwaliteit karakterisatie van NOTA-gemodificeerde

somatropines.

Methoden en Resultaten

Er werden vijf verschillende immobilisatietechnieken onderzocht en met elkaar vergeleken

voor de ligand immobilisatie densiteit, de binding capaciteit en de specificiteit. De

immobilisatie met de aminekoppeling van somatropine gaf de beste resultaten (59 pg/mm2

oppervlakte-gebonden ligand, 66.7% RR% en 1.5% NSB%). Er werd een regeneratie

procedure ontwikkeld voor de gevonden immobilisatietechniek. De meeste geschikte conditie

was een 1:1 mengel van 50 nM NaOH en 0.1% SDS die zorgde voor het complete wegwassen

van gebonden hGHAb van het geimmobiliseerde somatropine oppervlak, waarbij de activiteit

van het ligand werd bewaard. Tot slot werd de ontwikkelde methode toegepast om de

bindingaffiniteit en kinetiek te bepalen van NOTA-gemodificeerde somatropines voor

hGHAb. De gevonden KD (sub-nanomolair) waren equivalent voor somatropine en NOTA-

gemodificeerde somatropines.

Conclusie

De hGHAb-gebaseerd SAW methode werd succesvol toegepast voor karakterisatie van

NOTA-gemodificeerde somatropines. Er werd gevonden dat de NOTA-modificaties bij

somatropine geen verandering veroorzaken in de bindingaffiniteit tegenover hGHAb.

Momenteel wordt er verder onderzoek verricht naar de functionele kwaliteit van humaan

groeihormoon receptor (hGHR) op tumorcellen.

TABLE OF CONTENT

1! INTRODUCTION* 1!

1.1! ANALYTICAL*TOOLS*FOR*THE*FUNCTIONAL*CHARACTERIZATION*OF*BIOPHARMACEUTICALS* 1!1.1.1! LABELLED)LIGAND!BINDING!ASSAYS! 2!1.1.2! ISOTHERMAL!TITRATION!CALORIMETRY! 2!1.1.3! OTHER!BINDING!ASSAYS! 2!1.2! BIOSENSORS* 3!1.2.1! SURFACE!PLASMON!RESONANCE!BIOSENSOR! 3!1.2.2! QUARTZ!CRYSTAL!MICROBALANCE!BIOSENSOR! 5!1.2.3! SURFACE!ACOUSTIC!WAVE!BIOSENSOR! 5!1.2.4! APPLICATIONS! 7!1.3! THE*HUMAN*GROWTH*HORMONE*SYSTEM* 9!1.3.1! SOMATROPIN:!HISTORY,!USE!AND!ABUSE! 9!1.3.2! PHYSIOPATHOLOGY!OF!HUMAN!GROWTH!HORMONE!IN!CANCER! 10!1.4! PEPTIDE=*AND*PROTEIN=BASED*RADIOPHARMACEUTICALS* 13!

2! OBJECTIVES* 15!

3! MATERIALS*AND*METHODS* 16!

3.1! LITERATURE*REVIEW*=*APPLICATIONS*AND*METHOD*VARIABILITY* 16!3.2! REAGENTS*AND*MATERIALS* 16!3.3! SENSOR*CHIP*PREPARATION*FOR*LIGAND*IMMOBILISATION* 17!3.3.1! CLEANING!AND!CHEMICAL!ETCHING!OF!THE!SENSOR!CHIP! 17!3.3.2! PREPARATION!OF!A!CARBOXYMETHYL!DEXTRAN!HYDROGEL!SENSOR!CHIP! 17!3.4! DEVELOPMENT*OF*A*HGHAB=BASED*SAW*METHOD* 18!3.4.1! LIGAND!IMMOBILIZATION! 18!3.4.2! SCREENING!FOR!A!REGENERATION!CONDITION! 19!3.4.3! ANALYTE!INTERACTION!WITH!IMMOBILISED!LIGAND! 20!3.4.4! NON)SPECIFIC!BINDING!OF!ANALYTE!TO!IMMOBILISED!LIGAND!SURFACE! 20!3.4.5! NON)SPECIFIC!BINDING!OF!ANALYTE!TO!DEXTRAN!HYDROGEL!SENSOR!CHIP! 20!3.5! FUNCTIONAL*CHARACTERIZATION*OF*NOTA=MODIFIED*SOMATROPINS* 21!3.5.1! IMMOBILISATION!OF!SOMATROPIN!(DERIVATIVES)!VIA!AMINE!COUPLING! 21!3.5.2! BINDING!EXPERIMENTS!BETWEEN!HGHAB!AND!NOTA)MODIFIED!SOMATROPINS! 21!3.6! DATA=ANALYSIS* 22!3.6.1! FEASIBILITY TEST FOR SAW BINDING EXPERIMENTS! 22!3.6.2! EVALUATION!OF!THE!IMMOBILIZATION!STRATEGIES! 23!

3.6.3! EVALUATION OF THE REGENERATION CONDITIONS! 23!3.6.4! DETERMINATION!OF!DISSOCIATION!CONSTANT! 24!

4! RESULTS* 27!

4.1! LITERATURE*REVIEW*=*APPLICATIONS*AND*METHOD*VARIABILITY* 27!4.1.1! RANDOM!VARIABILITY!IN!KD!VALUES! 27!4.1.2! MODEL!VARIABILITY! 28!4.2! DEVELOPMENT*OF*A*HGHAB=BASED*SAW*METHOD* 29!4.2.1! EXPLORING!LIGAND!IMMOBILISATION!STRATEGIES! 29!4.2.2! EVALUATION!OF!THE!NON)SPECIFIC!BINDING!OF!ANALYTE!TO!DEXTRAN!HYDROGEL!SENSOR!CHIP! 34!4.2.3! SCREENING!FOR!REGENERATION!OF!SOMATROPIN!LIGAND! 35!4.3! FUNCTIONAL*CHARACTERIZATION*OF*NOTA=MODIFIED*SOMATROPINS* 35!4.3.1! IMMOBILIZATION!OF!SOMATROPIN!AND!DERIVATIVES!USING!AMINE!COUPLING! 35!4.3.2! BINDING!EXPERIMENTS!BETWEEN!IMMOBILIZED!SOMATROPIN!(DERIVATIVES)!AND!HGHAB! 37!4.3.3! SPECIFICITY!OF!THE!METHOD! 39!

5! DISCUSSION* 41!

5.1! LITERATURE*REVIEW*=*APPLICATIONS*AND*METHOD*VARIABILITY* 41!5.1.1! RANDOM!VARIABILITY!KD!VALUES! 41!5.1.2! MODEL!VARIABILITY! 42!5.2! DEVELOPMENT*OF*A*HGHAB=BASED*SAW*METHOD* 42!5.2.1! EXPLORING!IMMOBILISATION!PROCEDURES!FOR!THE!IMMOBILISATION!OF!ANTIBODIES! 43!5.2.2! SCREENING!FOR!OPTIMAL!REGENERATION!CONDITIONS! 46!5.3! FUNCTIONAL*CHARACTERIZATION*OF*NOTA=MODIFIED*SOMATROPINS* 46!5.3.1! IMMOBILIZATION!OF!SOMATROPIN!AND!DERIVATIVES!USING!AMINE!COUPLING! 46!5.3.2! BINDING!EXPERIMENTS!BETWEEN!IMMOBILIZED!SOMATROPIN!()DERIVATIVES)!AND!HGHAB! 47!

6! CONCLUSION* 50!

7! REFERENCES* 51!

8! LIST*OF*ATTACHMENTS* 62!

LIST OF USED ABBREVIATIONS

BSA Bovine serum albumin

cGH Cadaveric growth hormone

CM carboxymethylated

DOTA 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid

DruQuaR Drug Quality and Registration

EDC 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide

ELISA Enzyme-linked immunosorbent assay

EMA European Medicines Agency

FDA Food and Drug Administration

FIA Fluorescence immunoassay

GAG Glycosaminoglycan

GH Growth hormone

GHD Growth hormone deficiency

HBS HEPES buffered saline

HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

hGH Human growth hormone

hGHAb Human growth hormone antibody

hGHR Human growth hormone receptor

HTS High throughput screening

IDT Interdigital transducer

IGF Insulin growth factor

ITC Isothermal titration calorimetry

mAb Monoclonal antibody

MUD 11-mercapto-1-undecanol

NET Neuroendocrine tumour

NHS N-Hydroxysuccinimide

NOTA 1, 4, 7-triazacyclononane-1,4,7-triacetic acid

QC Quality control

QCM Quartz crystal microbalance

rhGH Recombinant human growth hormone

RIA Radio immunoassay

SAM Self-assembling monolayer

SAW Surface acoustic wave

SDS Sodium dodecyl sulphate

SPR Surface plasmon resonance

TETA 1,4,8,11-tetraazacyl cotetradecane-N,N',N ",N"'-tetraacetic acid

WADA World Anti-Doping Agency

1

1 INTRODUCTION

1.1 ANALYTICAL TOOLS FOR THE FUNCTIONAL CHARACTERIZATION OF

BIOPHARMACEUTICALS

The biopharmaceuticals or biotechnological medicinal products (“biotech drugs”) are

an increasing success within the pharmaceutical industry. It is expected that by 2015, 50% of

the newly introduced drugs will be biotech drugs [1]. Because the patents of many

biopharmaceuticals are expired or are about to, the opportunity opens for the development of

biosimilars, i.e. copy-versions of an already authorized biopharmaceutical, which are not

identical but highly similar. Many challenges come with the development and regulatory

approval of biosimilars, due to their magnitude (i.e. large primary structure with small

variations possible), post-translational modification (e.g. glycosylation), three dimensional

structure (e.g. T-shape vs. Y-shape antibodies) and protein aggregation. The manufacturers of

the biosimilars have to prove that their product is comparable to the authorized reference

medicinal product at the level of quality, efficacy and safety [2] (Figure 1.1). The complexity

of the authorization requirements of biosimilars illustrates the emerging role of analytical

technologies in the regulatory approval of biosimilars [3].

Figure 1.1: Analytical characterization of biosimilar products to establish biosimilarity - a step-by-step approach. Adapted from reference [4].

A critical aspect of the regulatory approval of biosimilars is the side-by-side

comparison of the functional activity, which is basically achieved by in vitro cell-based assays

[5]. One of the parameters to be evaluated is the binding affinity, which is quantitatively

2

measured as the dissociation constant (KD). Moreover, the European Medicine Agency

(EMA) recommends the use of emerging technologies such as real time binding assays [6].

1.1.1 Labelled-ligand binding assays

In labelled-ligand binding assays radio- and fluorescent-labelled ligands are used for

the measurement of the binding affinity. A radioactive labelled (with radioisotopes such as 3H, 125I, 35S) or fluorescent-labelled ligand is applied to detect and quantify the binding to a

specific target (enzymes, receptors in tissues or cell cultures). The drawback with labelled-

ligand binding assays is the labelling itself, because this can lead to potential alterations in the

binding characteristics. Also the high cost and hazards for the removal of radioactive waste,

encourages the development of label-free (non-radioactive) techniques [7].

1.1.2 Isothermal titration calorimetry

Isothermal titration calorimetry (ITC) determines the binding affinity, enthalpy,

entropy and reaction stoichiometry in a label-free thermodynamic assay by sensing heat

changes caused by the binding reaction [8]. ITC involves two identical cells, a reference cell

(contains buffer or water) and a sample cell (contains the ligand solution). The analyte is

added to the sample cell, which results in enthalpic changes induced by the ligand-analyte

interaction. These changes are measured as the input of power per time required to retain an

equal temperature between both cells [9]. ITC does not require any labelling or

immobilisation, which allows us to study the binding interaction in their native state.

However, large sample volume (>ml) and sample concentration (>µM) are required for a

single experiment [10]. ITC is used to investigate protein-protein interactions, protein-

DNA/RNA interactions, protein-small molecule interactions and enzyme kinetics [11]. ITC is

less suitable for the evaluation of the binding affinity with receptors presented on cells in

culture and tissues as the cells will die because of the lack of oxygen during the experiment

[12].

1.1.3 Other binding assays

Other techniques used for the investigation of ligand-receptor interaction are structure-

based ligand binding assays. Spectroscopic techniques such as NMR and X-ray

3

crystallography are able to monitor changes in the molecular structure that occur upon

interaction [13, 14].

Biosensors are an emerging method for the label-free determination of binding

characteristics in real time. The different techniques and applications will be further discussed

in chapter 1.2.

1.2 BIOSENSORS

A biosensor is an analytical device, which employs immobilized biological

compounds or ligands for detection of analytes in samples. Biosensors can be divided in

different classes according to the mode of transduction: optical, mass, electrochemical and

thermal [15]. They are able to investigate molecular interactions in real time without labelling

of the analyte. Binding affinity and kinetics of an interaction are determined based on the

observed binding rates. As previously mentioned, label-free methods have quite some

advantages over fluorescent-labelling and radiolabelling, e.g. cost and time saving, less

harmful and less false conclusions due to interferences of the labelling agent [16].

Three biosensor systems are currently available on the market; the Surface Plasmon

Resonance (SPR), the Quartz Crystal Microbalance (QCM) and Surface Acoustic Wave

technology (SAW), each is based on different detection platforms.

1.2.1 Surface Plasmon Resonance biosensor

SPR biosensor is the most popular technique among the biosensors. It is the most

commonly used biosensor as it is the most frequently published in biosensor literature (Figure

1.2).

Figure 1.2: A summary of the number of publications per year referencing “surface acoustic wave”, “quartz crystal microbalance” or “surface plasmon resonance” based on the ISI citation database.

4

SPR technology is an optical biosensor that detects changes in the refractive index

near the surface layer of the sensor chip. Surface plasmon resonance is an optical

phenomenon that occurs under the conditions of total internal reflection (TIR): all the incident

light is reflected at a surface-solution interface. Although no light is coming out at the

interface of the solution, an electromagnetic field called ‘evanescent waves field’, extends a

short distance into the medium of lower refractive index.

In the setup of an SPR biosensor, monochromatic and plane-polarized light passes

through a prism, reflects on the backside of the sensor chip surface into a detector (Figure 1.3

top). At a certain angle of incident light, known as the resonance angle, the photons will

interact and be absorbed by oscillating electrons in the metal film (i.e. surface plasmons). This

phenomenon, surface plasmon resonance, causes a reduction in the intensity of reflected light

and can be seen as a dip in the SPR reflection intensity curve (Figure 1.3 bottom).

The surface plasmons are sensitive to fluctuations of electron density at the interface of

two materials. Thus, the resonance angle is dependent on the refractive index of the solution

close to the gold layer. This means that when analyte and ligand interact at the interface, the

resonance angle is modified. The shift in resonance angle is proportional to the mass of the

bound molecules [17].

Figure 1.3: Mechanism of SPR biosensor. Interaction between ligand and analyte can be monitored in real time by analysis of the reflected light. The resonance angle shifts when a molecular binding event takes place that changes the refractive index on the sensor chip surface. Figure adapted from reference [18].

5

1.2.2 Quartz Crystal Microbalance biosensor

In contrast to SPR, where binding processes are measured by changes in refractive

index, the QCM is based on the properties of piezoelectric materials. The principles are

similar to the SAW biosensor, which is discussed further below (1.2.3.1). The difference with

SAW biosensor is that the acoustic waves are propagated along the whole substrate, thus are

not limited to the surface of the quartz crystal.

1.2.3 Surface Acoustic Wave biosensor

Surface acoustic wave biosensors are devices that utilize a high frequency acoustic

wave (100 MHz - 300 MHz), as the name implies [15]. They were first used as filters and

resonators in the telecommunication industry [19]. Later, they were picked up by the bio-

chemical scientific community for sensing applications. These types of biosensors are highly

sensitive towards mass changes and viscoelastic changes on the sensor surface.

1.2.3.1 Principle of SAW sensor technology

Basically, a SAW sensor generates and detects acoustic waves on the surface of a

piezoelectric substrate. When an electric field is applied to a piezoelectric material, this

creates a mechanical stress; and the other way around, imposing an appropriate mechanical

stress generates an electric field. This mechanical property of piezoelectric materials is used

in the setup of the SAW biosensor. Two interdigital transducers (IDT) are placed on the top of

the substrate. The input IDT is used to provide an electric field that acts on the piezoelectric

substrate to form an acoustic wave. The generated wave propagates through the substrate,

where the biological detection occurs and is finally reconverted at the receiving IDT into

electrical signals [20]. Most common piezoelectric material used for acoustic wave devices

are quartz, lithiumtantalate, lithiumniobate and zinc oxide [21].

SAW sensors are capable to detect mass and viscoelastic changes on the sensitive

surface by measuring wave characteristics. When the surface layer experience changes in

bound mass e.g. upon interaction between ligand and analyte, it will perturb the wave

propagation and accordingly cause shifting of the phase. Viscosity changes of the ligand on

the other hand are indicated by a change in wave amplitude [19].

6

Figure 1.4: Basics of a SAW biosensor A. Elements of a SAW sensor chip. The basis of a sensor chip is a piezoelectric crystal such as quartz (=substrate) with IDTs at both ends. On the top the substrate a waveguide layer is applied, which propagates the acoustic wave in the surface layers. The chip is covered with plain gold to allow immobilisation of ligands. B. Detail of gold surface. The interaction between analyte (green) and ligand (blue) is studied at the surface of the chip.

1.2.3.2 Love-wave mode SAW biosensors

Currently the most sensitive SAW sensors are the sensors in Love mode configuration,

where the acoustic wave is guided through a single surface. This physical effect was

discovered by Love in 1911, who observed effects of an earthquake far from the epicentre

because the waves were guided through stratified geological plates. The same principle is

applicable to SAW devices where the wave is propagated in a guiding layer at the top of the

substrate rather than in the bulk of the piezoelectric material. Therefore a maximal amount of

acoustic energy is trapped close to the surface, where it will be strongly susceptible for

alternations, yielding a higher sensitivity of the sensor. The restriction of the energy in the

guiding layer is due to a lower wave propagation velocity in the waveguide layer than in the

substrate [15].

1.2.3.3 SAW biosensors for molecular interaction analysis

SAW technology can be used to characterize interactions between ligands

(immobilized on the sensor chip) and analytes (solution flowing over the ligands). Binding of

the analyte to the ligands alters the phase velocity of the acoustic wave. This change can be

monitored in real time as shown in Figure 1.5. As the analyte binds to the surface, a positive

phase shift is observed, which leads to an increase of the response (= association). At a certain

point an equilibrium is reached, where equal amounts of analyte are associating and

7

dissociating with the ligand (= steady state). When the analyte solution is then replaced by

buffer, a decrease in signal is observed, because of the separation of the ligand-analyte

complex and loss of bound analyte (= dissociation). Binding affinities and kinetics can be

determined by fitting the curves (i.e. the sensorgram) to an appropriate binding model [20].

Figure 1.5: Phase shift increases as the analyte binds to the ligands resulting in the association phase. After equilibrium, the analyte injection is stopped and replaced by buffer flow. The phase signal decreases, which indicates the dissociation phase. The remaining signal after the dissociation phase comes from the remaining associated ligand-analyte complexes

1.2.4 Applications

SAW biosensors are currently been applied in divergent areas such as food analysis

[22], genomics [23], microbiology [24], environmental studies [25], chemical warfare

detection [26] and the medical diagnostics field [27]. The recent evolution in the biosensor

technology has expanded the capabilities of the biosensors, which allows the successful use of

biosensors in the drug development process [28]. In the next paragraphs, examples are given

of the biosensor application in the drug discovery, preclinical and toxicological studies,

formulation development studies and quality control environment.

1.2.4.1 Application of biosensors in drug development

Biosensors can be applied in many areas of the drug development process. Optical

biosensors have found their way into the pharmaceutical industry for ligand fishing, high

throughput screening (HTS), hit confirmation and lead optimization (Figure 1.6) [18, 29].

During hit confirmation, it is often desirable that the KD is within a specific concentration

Associa'on)

Dissocia'on)

Equilibrium)

PHAS

E)SH

IFT)(°))

TIME)(SEC)))

Injec'on)

8

range. The binding constants of receptor-drug complexes are generally situated in the

micromolar to nanomolar range [30].

Further applications in the pharmaceutical field are for example interactions studies

between a new lead compounds and biological membranes. Permeation of biologically active

compounds would result in changes of the viscoelastic properties of the lipid bilayers in the

immobilized model membranes [31]. Another example is the application of a SPR-based

method to predict the binding affinity of new drug compound for serum proteins, using

immobilized albumin or alpha-1-glycoprotein [32].

In addition, these devices have the potential to be used in toxicity studies. Cell toxicity

of new drug compounds can easily be analysed with whole cell systems; cytoskeletal changes

resulting in cellular volume retribution are detected with biosensors in a reproducible manner

[33].

Biosensors could also be useful in the formulation development. A biocompatibility

study between protein pharmaceuticals and a silicon-oil coated surface was performed with a

QCM. This information was used to determine potential interactions and/or adsorption of

proteins to silicon-oil coated packaging [34].

With the development of process analytical monitoring (PAT) within the

biopharmaceutical industry, bioprocess monitoring using biosensors may permit automated,

real time monitoring of critical performance quality attributes. An automated at-line SPR

detection system has recently been described to monitor secreted protein in a bioreactor

culture of transiently infected embryonic kidney cells [35] and automated SPR methods have

also been used to quantify levels of bioactive monoclonal antibody produced by a hybridoma

cell line.

Finally, biosensor assays have a number of advantages over alternative bioassay

techniques such as ELISA, FIA or RIA which are traditionally used for biopharmaceutical

process development and quality control (QC): (i) no need for reporter molecules such as

enzymes/substrates, (ii) monitoring in real time, (iii) reactants retain conformational integrity

and (iv) use of low concentrations of products within a crude sample [36, 37].

9

Figure 1.6: Overview of the application areas for biosensors in drug development process. Figure adapted from reference [18].

1.3 THE HUMAN GROWTH HORMONE SYSTEM

1.3.1 Somatropin: history, use and abuse

Somatropin or recombinant human growth hormone (rhGH) is widely used for the treatment

of growth hormone deficiencies (GHD) in both children and adults [38]. The first

recombinant hGH was produced in 1982 by Genentech Inc. Before the area of recombinant

DNA technology, children with GHD were treated with GH extracted from pituitary glands of

human cadavers (cGH) because bovine or porcine GH was not effective in humans because of

the major differences in molecular structure. When in 1985, several cases of Creutzfeldt-

Jacob disease were reported with patients who had received cGH [39], regulatory authorities

removed cGH from the market and accelerated the approval of rhGH.

The hGH is a single chain protein of 191 amino acid residues, with a molecular weight

of 22 kDa. The protein structure includes four alpha-helices and two disulphide bonds. The

hGH is a hormone secreted in the anterior pituitary gland. The main function of hGH is to

stimulate the synthesis and secretion of insulin-like growth factors (IGFs) in the liver, skeletal

muscle and other tissues. IGFs stimulate the proportional body growth and regulate several

aspects of metabolism. The actions of hGH are characterized as anabolic and lipolytic [40].

Today seven different somatropin formulations have been authorized in the EU

including two as biosimilars [38]. Moreover, somatropin was the first biosimilar approved in

the EU in 2006. This requires standardization of both protein content and specific activity of

10

rhGH to assure the similar efficacy of the different preparations [41]. With the introduction of

biosimilars, questions were raised about the criteria to demonstrate the comparability of these

biotherapeutics with their originator reference products and the development of new

analytical tools to support the required comparability studies [3].

Somatropin is currently only indicated for the treatment of GHD. In children, GHD are

manifested by short stature and growth retardation. The most common cause are congenital

disorders e.g. GHD resulting from mutations in GH or the growth hormone releasing hormone

receptor genes.

In adults, GHD rarely occurs and is often acquired by inflammation, trauma or tumour

of the hypothalamus or pituitary [42]. In addition to the use of rhGH as hormone replacement

therapy, somatropin is approved by the European Medicines Agency (EMA) to correct short

stature in children with Turner syndrome, Prader-Willi syndrome and chronic renal failure

[43]. Somatropin has also been approved by the FDA for the treatment of AIDS-related

cachexia, but this indication was however rejected by the EMA. Therefore, the use of

somatropin for AIDS wasting is considered off-label in Europe.

Somatropin is included in the prohibited list of the World Anti-Doping Agency

(WADA) [44]. rhGH is very popular among athletes as a performance-enhancing substance

because of its anabolic effects (increases lean body mass, reduces body fat) [45-47].

Somatropin is also illegitimately prescribed and/or delivered by doctors as an anti-aging

product to increase vitality [48]. The abuse of somatropin in sports and as an anti-aging

compound resulted in an increase of spurious/falsely-labelled/falsified/counterfeit (SFFC)

formulations [49].

1.3.2 Physiopathology of human growth hormone in cancer

Recent research showed that hGH and human growth hormone receptor (hGHR) might

play a role in the development of several tumours [50]. Acromegaly and other conditions

characterized with high levels of hGH have been associated with an increased risk for colonic

adenomas. However, rhGH was not found to be oncogenic when applied in patients to correct

for low hGH levels: patients with GHD who received hGH replacement therapy are not

associated with an increased risk of carcinomas [50, 51]. Moreover, the up-regulated

11

expression of hGHR has been widely reported in several malignancies e.g. prostate [52, 53],

breast [54], colon [55-57] and gastric cancers [58], indicating the involvement of hGH-system

in these tumours.

1.3.2.1 Prostate cancer

Androgens like testosterone play a key role in the progression of prostate cancer. This

explains the treatment of early-diagnosed Prostate cancer with anti-androgens and in some

cases castration. In the late stage, therapies like radiotherapy and surgery are necessary [59].

Recent studies have suggested the involvement of hGH and hGHR in the

pathophysiology of prostate cancer: (i) the expression of hGHR is increased in prostate cancer

cell lines and in human tissues from patient diagnosed with prostate cancer [53], (ii) the

hGHR expression in prostate cancer cell lines is regulated by steroids and hormones

associated with the development and progression of prostate diseases [60], (iii) the hGH-

controlled proliferation of prostate cancer cells, (iv) stimulation of IGF and the expression of

bèta-oestrogen receptors in prostate cancer cell lines by hGH, and (v) hGH interaction with

IGF and oestrogen to stimulate proliferation of androgen dependent prostate cancer cells [61].

These studies indicate the involvement of hGH and hGHR in prostate cancers, although

aspects of the exact mechanism remain unclear. hGHR may be considered as a target for the

development of new therapeutics against prostate cancer [52].

1.3.2.2 Breast cancer

The current paradigm in oncology state that tumour growth is a pathological

reproduction of the developmental processes. The development of the mammary glands

involves complex interactions between oestrogen, hGH and prolactin. hGH is necessary for

the formation of rapidly proliferating terminal end bud structures in the developing mammary

gland [62]. The involvement of ovarian hormones in the development and progression of

breast cancer is well known [63, 64]. In the last decade, more evidence suggests the same for

(autocrine) hGH (Figure 1.7).

First indication of the association between hGH and breast cancer is the increased

level of hGHR expression in breast cancer tissues [54]. Another indication is the presence of

locally produced hGH. Autocrine hGH promotes cell proliferation and cell survival in

12

mammary cancer cells. In addition, studies have showed that hGH increases the telomerase

activity, which could contribute to the unlimited replication capacity of these cells [62]. More

recent findings suggest that autocrine hGH act as a potential regulator, which promotes

angiogenesis and induces resistance to chemotherapeutic drugs [65, 66].

hGHR antagonism is considered as a potential approach for the treatment of breast

cancer. However, it appears that the hGH signalling in breast cancer cell lines is also

mediated by the association of hGH to prolactin receptor. Therefore, both hGHR and

prolactin receptor should be considered as potential targets in the development for new

therapeutics for breast cancer [67].

Figure 1.7: Role of autocrine hGH in mammary cancer. Figure adapted from reference [62]

1.3.2.3 Neuroendocrine tumours

Neuroendocrine tumours (NET’s) are a heterogeneous group of neoplasms from cells

of the neuroendocrine system. NETs can occur anywhere in the body, but are mostly found in

the gastrointestinal tract (∼70%) and the bronchopulmonary system (∼25%). Some do not

cause symptoms, while other NETs are associated with a wide variety of symptoms due to the

secretion of diverse peptide hormones and bioactive amines [68].

Most NETs overexpress receptors for somatotropin release inhibiting factor or

somatostatin. The cornerstone of the symptomatic treatment for NETs is somatostatin (and

analogues), which inhibit hormone secretion and therefore reduce the hormone-related

13

symptoms as well as tumour progression [69]. A radiolabelled somatostatin analogue is

currently under investigation (phase II) as therapeutic approach for certain types of NETs

(stage IVc with visual tumour uptake of 111In-Octreoscan) [70]. This approach is based on

the linkage of high-energy radioisotope (90Y or 177Lu) to a specific peptide-ligand

(edotreotide) of which receptors (somatostatin receptor subtype 2) are overexpressed on the

tumour cell’s surface [71].

Recently overexpression of hGH and hGHR was found in LCNEC (large cell

neuroendocrine carcinomas) patients suggesting a role for hGH and hGHR in the progression

of NETs [72]. This information could be valuable for the development of peptide- or protein-

based radiopharmaceuticals.

1.4 PEPTIDE- AND PROTEIN-BASED RADIOPHARMACEUTICALS

The radiolabeled somatostatin analogue is one of the many examples of protein-based

radiopharmaceuticals used for the diagnostic and/or treatment of certain cancers. In the past

decades, nuclear medicine has played an important role in the targeting and treatment of

cancers. Most research is focussed on radiolabelled antibody conjugates, i.e. radionuclides

(111In, 68Ga, 99mTC) combined with monoclonal antibodies. The antigen-binding specificity

provides the capability to localize the tumour, while the radionuclide allows the visualization

(diagnostic function) and/or the killing of the cancer cells [73].

In recent years, radiolabelled (small) peptides have gained enormous interest within

the scientific community [74]. The overexpression of peptide receptors in tumour cells (e.g.

somatostatin receptor in NETs) is the basis for the use of peptide-based radiopharmaceuticals.

The receptors on the surface of the tumour cells are potential targets. The most widely used

radiolabelled peptide is 111In-DPTA-octreotide, a radiolabelled analogue of somatostatin used

for scintigraphy of NETs. For the development of a radiolabelled peptide, it is important that

the high binding affinity of the peptide with its receptor remains the same after radiolabelling,

i.e. the chemical modifications do not influence the binding affinity [75].

There are two different methods to link radioisotopes to the ligand. In a direct

labelling process, the radioisotope is directly attached to a functional group of the ligand (e.g. 123I via iodination of tyrosine residues) [74]. Indirect methods involve the use of chemical

14

spacers to link the radionuclide to the ligand. These organic chemical spacers, called

bifunctional chelating agents, contain a reactive group for coupling to the ligand as well as a

metal-chelating group for the complexation of the radionuclide. A wide variety of

bifunctional chelating agents are available e.g. DOTA, NOTA (Figure 1.8) and TETA [76].

For the therapeutic application of a radiolabelled peptide, the internalization of the

peptide ligand – receptor complex into the tumour cells is desirable [75] and studies have

reported the internalization of hGH-hGHR complex [77]. This finding and the overexpression

of hGHR on the surface of various types of cancer cells make hGHR a potential candidate for

therapeutic tumour targeting with radiolabelled somatropin.

Figure 1.8: p-SCN-Bn-NOTA. NOTA is a nine-member cyclic compound and has three carboxylated groups and nitrogens, for six dative bonds with metal ions (e.g. Ga(III)). The thiocyanate groups forms the reactive site for the covalent attachment to free amines (e.g. lysine residues

15

2 OBJECTIVES

Somatropin (i.e. recombinant human growth hormone, rhGH) is a biologic drug,

currently only approved to treat growth hormone deficiencies. Recently, studies indicate a

possible role of hGH in certain cancers; hence, the oncologic potential of somatropin and

analogues/modified proteins is currently under investigation. Modifications with chelating

agents like 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) allows the incorporation of

radiometals for SPECT/PET-diagnostic (67Ga, 68Ga, 111In) or therapeutic (90Y) purposes.

Somatropin has nine potential binding sites (Lys) for NOTA. Therefore, it is important to

characterize the obtained product under different specified synthesis procedures.

The main objective of this paper is to develop a hGHAb-based SAW method in order to

perform a functional quality characterization of NOTA-modified somatropins.

(i) The first part of this master dissertation will discuss whether or not SAW biosensors

are applicable tools in the functional quality control. A thorough literature search

will be performed.

• What is the current status of biosensors in drug development and quality control?

• What are analytical patterns among study results, such as error on the KD

determinations in SAW biosensor experiments?

• Can biosensors be used for the determination of binding differences of chemical

modified proteins? When do authors talk of a significant change in the binding

affinity?

(ii) The second part of this master dissertation will focus on the further development of the

hGHAb-based SAW method (Figure 2.1). More specifically, we will investigate

which immobilisation strategy leads to the most optimal conditions (ligand

immobilisation density, binding capability and specificity). Furthermore, an appropriate

regeneration procedure is required.

(iii) In the third part of this master dissertation we will apply the developed SAW method to

investigate if NOTA-modified somatropins, prepared under different conditions, result

in changes of the binding properties towards hGHAb.

16

3 MATERIALS AND METHODS

3.1 LITERATURE REVIEW - APPLICATIONS AND METHOD VARIABILITY

Relevant research papers concerning SAW biosensor technology were collected by a

thorough literature search in Web of Science and Pubmed databases. Publications were

selected from 2008 until present (2013) with search terms "love wave” or “surface acoustic

wave", and "biosensor” or “immunosensor" (in the title or abstract). Publications were found

suitable for consideration in this review if: (i) the device used in the study was a surface

acoustic wave biosensor, and (ii) the application of the study was a binding assay. Finally,

scientific articles wherein SAM® 5 Blue or S-Sens K5 instrument was mentioned, were

included as well.

The next step was a detailed examination of the articles, for further selection. Criteria

were introduced based on the technical specifications for the love wave biosensor similar to

the SAM®5 or S-Sens K5 instrument: 1) a working frequency within 100 MHz to 300 MHz

and 2) exclusion of studies in the gaseous phase. These criteria especially apply to in-house

assembled biosensors.

3.2 REAGENTS AND MATERIALS

Ammonium hydroxide (28% w/v), 4-(2-Hydroxyethyl)piperazine-1-ethanesulfonic

acid (HEPES), ethanol absolute, 2-propanol, 11-mercapto-1-undecanol, epichlorhydrin,

diglyme, NaOH, dextran from Leuconostoc spp., bromoacetic acid, NaCl, NaIO4, acetic acid,

N-Ethyl-N’-(3-dimethylaminopropyl)carbodiimide (EDC), N-Hydroxysuccinimide (NHS),

ethanolamine, protein G’ from streptococcus sp. (recombinant, expressed in Escherichia coli),

carbohydrazide and sodium dodecylsulfate (SDS) were purchased from Sigma-Aldrich

(Diegem, Belgium). Sodium acetate trihydrate, albumin fraction V (from bovine serum)

(BSA), sodium borohydride and H2O2 (30%, v/v) were procured from Merck (Overijse,

Belgium). Zomacton® 4 mg, (Ferring, somatropin Ph. Eur.) was obtained from the Ghent

University Hospital (Ghent, Belgium). Lyophilized NOTA-modified somatropin was

synthetized at DruQuaR (Ghent University). The monoclonal mouse antibodies against hGH

were purchased from Thermo Fisher Scientific (Aalst, Belgium). All aqueous solutions were

prepared with 18.2 MΩ × cm deionized water, purified in an Arium 611 purification system

(Sartorius, Göttingen, Germany).

17

SAW experiments were performed with the SAM®5 Blue from SAW Instruments

GmbH (Bonn, Germany). Sensor chip, covered with a thin layer of gold, were also purchased

from SAW Instruments. Each chip received a unique number, to record its use-history. All

SAW experiments were performed at 20°C using HBS running buffer (10 mM HEPES (pH

7.4) and 150 mM NaCl) unless otherwise specified. The solutions intended for injection in the

SAW biosensor were prepared in LoBind Eppendorf® tubes or in glass vials.

3.3 SENSOR CHIP PREPARATION FOR LIGAND IMMOBILISATION

Before immobilisation of ligands, cleaning and chemical etching (i.e. removal of all

organic residues incl. immobilised ligand, up to the gold surface) of the sensor chip were

performed. These steps enabled the repeated use of the sensor chips [78].

3.3.1 Cleaning and chemical etching of the sensor chip

The chip was first pre-cleaned by 3 minutes ultrasonification in deionized water to

remove salts and polar compounds (e.g. carbohydrates). Then the chip was ultrasonicated 3

minutes in aceton followed by 3 minutes in isopropanol for the removal of lipophilic and non-

polar compounds (e.g. lipids, proteins, sugars). The surface was dried in a stream of N2. To

remove all organic residues from the gold layer, a chemical etching procedure was performed.

The chip was immersed for 3 minutes in a freshly prepared boiling solution of 5:1:1 deionized

water, ammonia and hydrogen peroxide at 70°C. Next the chip was rinsed in deionized water

and dried in a stream of N2.

3.3.2 Preparation of a carboxymethyl dextran hydrogel sensor chip

A self-assembled monolayer with end-standing hydroxyl groups was formed by

incubating the sensor chip in a 2 mM 11-mercapto-1-undecanol (MUD) solution in ethanol

over night. Afterwards the MUD modified chip was sonicated three times in absolute ethanol.

Next step was the formation of carboxymethyl (CM) dextran hydrogel (Figure 3.1). This was

achieved by the initial activation of the –OH groups with 0.6 M epichlorhydrin in a 1:1

mixture of diglyme and 0.4 M NaOH solution. After an incubation period of 4 hours, the chip

was covered with a 0.303 g/ml dextran solution in 0.1 M NaOH. Finally the dextran hydrogel

was carboxylated by covering the chip in a 1 M bromoacetic solution in 2 M NaOH for at

18

least 20 hours. The chip was rinsed with deionized water and air-dried. The chip was stored at

4°C to prevent microbial growth. Before use, the chip was inserted in the SAM®5 instrument

and conditioned with HBS running buffer (pH 7.4) for approximately 20 min.

Figure 3.1: Scheme of CM-dextran hydrogel modification of the golden surface. Adapted from reference [79].

3.4 DEVELOPMENT OF A HGHAB-BASED SAW METHOD

3.4.1 Ligand immobilization

3.4.1.1 Immobilisation of hGHAb via amine coupling

Prior to the immobilisation of hGHAb on a carboxymethylated (CM)-dextran

modified sensor chip, 10 µl of 0.1 % SDS was injected to wash away all non-specific bound

material on the chip. The chip was then equilibrated for 20 min in HBS running buffer (pH

7.4) in order to get a stable baseline. Next the chip was activated by injection of 130 µl of a

1:1 EDC:NHS mixture at a flow rate of 12.5 µl/min. Three successive injections of 25 µg/ml

hGHAb in 10 mM sodium actetate buffer pH 4.5, were carried out for 670 s to obtain an

effective immobilisation of the antibody. Finally, 130 µl of 1 M ethanolamine (pH 8.5) was

injected to block the remaining active sites, followed by an equilibration time of 30 min to

stabilize the baseline.

3.4.1.2 Immobilisation of hGHAb as somatropin:hGHAb-complex via amine coupling

A 1:1 somatropin:hGHAb solution was prepared to be immobilised directly to the

surface. In a LoBind Eppendorf® tube, equimolar quantities of hGHAb and somatropin in

HBS buffer were incubated for 10 min at room temperature. The hGHAb:somatropin solution

was further diluted to a final concentration of 25 µg/ml hGHAb with 10 mM sodium acetate

buffer (pH 4.5). The immobilisation of the complex was performed according to the protocol

in 3.3.1.1. After the deactivation step with 1 M ethanolamine, a 20 µl 0.1 % SDS injection

19

was performed to dissociate the complex, followed by an equilibration time of 20 min. to

stabilize the baseline.

3.4.1.3 Immobilisation of somatropin via amine coupling

Somatropin was also immobilised to the CM-dextran sensor chip using amine-

coupling chemistry. After the equilibration of the sensor chip, 50 µg/ml somatropin in acetate

buffer pH 4.5 was immobilised to the surface at the same conditions as 3.3.1.1.

3.4.1.4 Immobilisation of hGHAb via aldehyde coupling

The cis-diols of the sialic acids of the glycoproteins on hGHAb were oxidized to

aldehyde functions on ice using 4 µl of a 0.5 mg/ml hGHAb solution mixed with 0.8 µl of a

50 mM sodium m-periodate in 100 mM sodium acetate (pH 5.5) in a LoBind Eppendorf® tube

(final volume: 41.6 µl). The reaction was stopped after 20 min by desalting the mixture on a

desalting-column with 10 mM sodium acetate buffer (pH 4.0). The oxidized antibody was

eluated in 3.5 ml. The sensor chip was activated with a 1:1 NHS:EDC solution according to

3.3.1.1, followed by hydrazide modification using a 87.5 µl-injection of 5 mM

carbohydrazide in water. Any remaining active esters were inactivated with 130 µl injection

of 1 M ethanolamine (pH 8.5). Multiple 140 µl injections of oxidized-hGHAb were

performed in order to achieve an efficient immobilisation. Finally, 0.1 M sodium borohydride

in 0.1 M sodium acetate buffer (pH 4.0) was injected for 20 min to stabilize the hGHAb-

modified surface.

3.4.1.5 Immobilisation of hGHAb via protein G

50 µg/ml protein G (in 10 mM sodium actetate buffer pH 4.5) was immobilised to the

surface following the same protocol as used in 3.3.1.1. After the deactivation of the remaining

active esters, hGHAb was captured to the protein G. Immobilisation of 25 µg/ml hGHAb in

HBS buffer (pH 7.4) was performed by three 140 µl injections.

3.4.2 Screening for a regeneration condition

0.1% SDS, as used in the pilot studies, was not able to fully regenerate (i.e. removal of

the remaining bound analyte without affecting the immobilised ligands) the somatropin-

immobilised surface. Therefore, a screening with different regeneration conditions was

20

performed: 0.5% SDS, acetate buffer pH 4.5, 0.5 NaCl, 5% ethyleenglycol, 1 mM NaOH and

a 25 mM NaOH and 0.05% SDS solution. After 200 µl injection of 100 nM hGHAb, each

regeneration solution was injected over the sensor chip surface for 60s, followed by a

equilibration time of 5 min.

3.4.3 Analyte interaction with immobilised ligand

The interaction between analyte and immobilised ligand was studied. The hGHAb-

immobilised surfaces were investigated with 200 nM somatropin injections. The somatropin-

immobilised surface was studied with 100 nM hGHAb injections. The samples were injected

through the five channels for 600 s at a continuous flow rate of 20 µl/min. All injections were

performed with an initial burst i.e. a brief increase of the flow rate to 150 µl/min at the

beginning of the injection. Changes in phase shift were measured. The dissociation was

observed for 30 min, during which only HBS buffer was passed over the surface. After each

binding cycle, regeneration was carried out for 60 s. A blank injection (running buffer) was

performed prior to each interaction analysis. Subtraction of the response of the blank injection

from the analyte injections removes small but systematic imperfections in the response

curves. Data represent the mean ± SEM of at least two measurements (n ≥ 2).

3.4.4 Non-specific binding of analyte to immobilised ligand surface

The specificity of the somatropin- and hGHAb immobilized surface was studied with

a 100 nM and 200 nM BSA injection, respectively. The samples were injected through the

five channels for 600 s at a continuous flow rate of 20 µl/min. Changes in phase shift were

measured. The dissociation was observed for 30 min, during which only HBS buffer was

passed over the surface. After each binding cycle, regeneration was carried out for 60 s.

3.4.5 Non-specific binding of analyte to dextran hydrogel sensor chip

The non-specific binding of somatropin, hGHAb and BSA to the dextran hydrogel

surface (i.e. without immobilised ligand) was evaluated and compared with each other. The

carboxymethylated dextran chip was first equilibrated for 20 min in HBS running buffer (pH

7.4) in order to get a stable baseline. The chip was then activated by injection of 130 µl of a

1:1 EDC:NHS mixture at a flow rate of 12.5 µl/min. Next, 130 µl of 1 M ethanolamine (pH

8.5) was injected to block the active sites, followed by an equilibration time of 30 min to

21

stabilize the baseline. On each chip, 200 µl somatropin, hGHAb and BSA were injected at a

flow rate of 20 µl/min with HBS running buffer, followed by 800 s dissociation.

3.5 FUNCTIONAL CHARACTERIZATION OF NOTA-MODIFIED SOMATROPINS

3.5.1 Immobilisation of somatropin (derivatives) via amine coupling

Somatropin was immobilised using amine-coupling chemistry. Prior to the

immobilisation of somatropin on CM-dextran modified sensor chip, 10 µl of 0.1% SDS was

injected to remove all non-specific bound material on the chip. The chip was equilibrated for

20 min in HEPES running buffer (pH 7.4) in order to get a stable baseline. Next the chip was

activated by injection of 130 µl of a 1:1 mixture of 0.4 M EDC and 0.1 M NHS at a flow rate

of 12.5 µl/min. Three successive injections of 50 µg/ml somatropin (in 10 mM sodium

actetate buffer pH 4.5) were carried out for 670 s to obtain an effective immobilisation. After

immobilisation of somatropin, 130 µl of 1 M ethanolamine (pH 8.5) was injected to block the

remaining active sites, followed by an equilibration time of 30 min. to stabilize the baseline.

The same protocol was used for the immobilisation of the somatropin derivatives.

3.5.2 Binding experiments between hGHAb and NOTA-modified somatropins

For the binding assay, hGHAb was diluted with HBS buffer covering a concentration

range between 25 nM - 100 nM. The dilutions were prepared in glass vials to prevent leaching

from plastic vials. The samples were injected through the five channels for 600s at a

continuous flow rate of 20 µl/min. All injections were performed with an initial burst, i.e. a

brief increase of the flow rate to 150 µl/min at the beginning of the injection. The dissociation

was observed for 30 min, during which only HBS buffer was passed over the surface. After

the dissociation, regeneration was carried out for 60 s with a regeneration solution consisting

of 25 mM NaOH and 0.05% SDS, followed by two 30 µl buffer injections. For the control

experiments 100 nM BSA was injected under the same conditions, except for the regeneration

step, which was carried out with 0.1% SDS. In addition, a blank injection (running buffer

without analyte) was performed prior to each bindings experiment and its response was

substracted from the analyte injections. Data represent the mean ± SEM of at least three

measurements (n ≥ 3).

22

3.6 DATA-ANALYSIS

3.6.1 Feasibility test for SAW binding experiments

The signal obtained from a SAW biosensor is related to the mass changes caused by the

investigated molecules. A prediction of whether a certain molecule will cause a sufficient

mass change to obtain a detectable signal is useful to estimate the feasibility of the method.

This is mainly determined by the sensitivity of the equipment.

A theoretical formula was previously proposed to evaluate the feasibility of the SAW

biosensor method for the application under study [80]. For proteins, peptides or small

molecules as ligand with known molecular weight following formula was proposed:

! = !!×!"!×!!!×!!"!

!> 0.5! !"!!! (1)

Where: X: the theoretical area-concentration of bound analyte (in pg/mm2)

P: the phase shift (°) related to the bound ligand

Mw2 is the molecular weight of the analyte (in pg/pmol)

Mw1 is the molecular weight of the ligand (in pg/pmol)

S: the conversion factor, is an equipment constant (0.0515°mm2/pg)

F: binding stoichiometry

This formula determines the amount of analyte that can be detected, with the

assumption that all binding sites of the immobilised ligand are available and active for a 1-to-f

binding ratio (f = stoichiometry). In case of somatropin ligand, f is 0.5 because one

somatropin molecule binds to one of the two binding sites on the antibody. Therefore one half

of the hGHAb is occupied. If hGHAb is immobilised, f is theoretically 2, since every antibody

molecule has two binding sites. The theoretically calculated ad hoc signal value has to exceed

instrument detection limit; 0.5 pg/mm2 (area-concentration) to obtain a detectable signal with

an equipment. The amount of theoretically bound analyte will be dependent on the area-

concentration of ligand immobilised on the chip (= P/S in pg ligand/mm2), as well as the Mw

of the ligand and the Mw of the analyte.

23

3.6.2 Evaluation of the immobilization strategies

The phase shifts from the different immobilization techniques were compared using

normalized response values, calculated as follows (2):

!!% = !!!"!#$%& !!"# ×!100 (2)

Where, RR% = the relative ligand response (%)

!analyte = phase shift of analyte injection at 550s (°)

!max = maximum phase shift (°) i.e. phase shift if all immobilised

ligands were occupied calculated with formula (1).

The non-specific binding of the interactions was expressed as a ratio of the response of

BSA and analyte to the ligand immobilised surface.

!"#% = !!!"# !!"!#$%& !×!100 (3)

Where, NSB% = the relative amount of non-specific binding (%)

!BSA = phase shift of 100 nM BSA injection at 550s (°)

!analyte = phase shift of 100 nM analyte injection at 550s (°)

3.6.3 Evaluation of the regeneration conditions

In order to compare the regeneration efficiency (RE%) of the different conditions

following calculations were made:

!"! % = Δ!!!!"#"$"!%&'!"Δ!!!!"#$%&'#!(" ×100!

!!

!" % = !!!"#$%"!!!"#"$"!%&'($(!!) − !!!!"#$%!!"#"$"!%&'($!(!!)!!!!"#$%!!"#$%&!'"! !! − !!!!"#$%"!!"#$%&'#!("(!!)

×100!!!

24

Figure 3.2: Schematic representation of a sensorgram. The marked points are used for the calculation of the regeneration efficiency.

3.6.4 Determination of dissociation constant

The kinetic constants (kon and koff) and dissociation constants (KD) were determined by

fitting the obtained curves to a 1:1 binding model. The one-to-one binding model is based on

[81, 82]:

! + ! ⇌ !"

!(!")!" = ! ! !!" − (!")!!""

3.6.4.1 Association phase algorithm: kon and koff

kon is the association rate constant (units M-1s-1) and koff the dissociation rate constant

(units s-1). In biosensor experiments, the ligand L is immobilized on the sensor surface. The

concentration of complex [AL] is therefore identical to the concentration of bound analyte A.

The concentration of bound analyte is proportional to the phase P, which is detected by the

SAW biosensor. Free ligand concentration [L] is the difference between total and bound

ligand concentration. When the analyte is injected in a flow over the sensor surface, the

analyte solution is constantly replenished and hence, the free concentration of the analyte may

be considered and identical to the total analyte concentration C. The reaction between

immobilized ligand and analyte in solution can thus be assumed to follow pseudo first order

kinetics and since the concentration of complex and free ligand now can be expressed in

terms of analyte phase response P:

t1

t2 t3

25

!"!" = !!" !!"# − ! − !!""!

! ! = !!" 1− !!!!"#! !!Where,

!!!"# = !!"! + !!""

!!" =!!"!!!"#!!"#

!

Figure 3.3: One-to-one kinetic binding model according to (4) with X0: time at which the association begins (i.e. start injection [A]) and Peq: difference between plateau (response at equilibrium) and Y0 (i.e. baseline response).!

For each concentration of analyte A, an apparent rate constant (kobs) is determined

based on the association profile (0 – 550 s) (Figure 3.3). These apparent rate constants are

plotted against the concentration (nM) as illustrated in Figure 3.4. The intercept is the

dissociation rate constant. The slope is the association rate constant.

3.6.4.2 Dissociation phase algorithm: koff

The process of dissociation of the formed complexes AL can also be observed once

the analyte solution has traversed the flow cell and the system reverts to buffer flow. The rate

of dissociation of the formed complexes is described by

! ! = !!!×!!!!!""×!!

(4)

(5)

26

Figure 3.4: kobs in function of analyte concentration [A] .

Figure 3.5: An exponential one phase decay model based on formula (5) with P0: response at the beginning of the dissociation (i.e. the end of the injection and return to buffer flow) and plateau: response at the end of the dissociation.

The obtained data from the dissociation profile will be fitted in the one phase decay

model (Figure 3.5) from which the dissociation rate could be derived. Since the dissociation is

independent of analyte concentration, the koff is calculated as the mean dissociation rate

constant of the different concentrations.

The final KD-value will be calculated with the kon from the association phase and a

weighted mean of the koff values from the association and dissociation phase. The binding

constant (KD expressed in nM) is determined by:

!!""!!"

= !!

Where, koff: the off-rate, kon: the on-rate and KD: the binding constant

kobs=kon*[A]+koff+k o

bs+

27

4 RESULTS

4.1 LITERATURE REVIEW - APPLICATIONS AND METHOD VARIABILITY

The SAW applications can be classified in 9 groups (Figure 4.1), with most

publications in the medicine diagnostics field (48%). An overview of the SAW literature is

given in Attachment 1.

Figure 4.1: SAW biosensor applications.

A thorough literature search was performed to investigate the variability of a SAW

biosensor method. The variability in KD values reported in the publications as well as the

model variability i.e. the error on the fitting of the KD, were analysed.

4.1.1 Random variability in KD values

From the 13 publications which reported an affinity measurement by SAW biosensor,

only two publications performed multiple experiments (n>1) with a total of three different

interaction studies (Figure 4.2). An analysis of the reported standard deviations on the mean

KD resulted in a mean RSD of 60.43% for the three experiments. Figure 4.2 shows that the

RSD for interaction studies with membrane vesicles (RSD of 58.13% and 89.11%) is

relatively larger than the RSD of protein-protein interaction (RSD of 34.06%).

Medicine diagnostics

48%

Food analysis 7%

Biomaterials 4%

Polymer science

4%

Cell biology 3%

Rheology 3%

Microbiology 21%

Environmental science

3%

Genomics 7%

28

Figure 4.2: Variability of repeatability of SAW method. Pr = protein, MV = membrane vesicles, GAG = Glycosaminoglycan

4.1.2 Model variability

The model variability (expressed as RSE%) signifies the error on the fitting of the KD,

i.e. the variability of the goodness-of-fit (Figure 3.3). Large deviations on the fitting were

observed (mean RSE of 38.38%), especially for studies with glycosaminoglycans (Figure

4.3).

Figure 4.3 Variability of the KD fitting. Pr = protein, MV = membrane vesicles, MM = model membrane,

Pe = peptide, GAG = glycosaminoglycan.

0

20

40

60

80

100

Pr/Pr MV/Pr MV/GAG

[24] [123a] [123b]

34

89

58

RSD

(%)

Reference

n=5!

n>3!n>3!

0!10!20!30!40!50!60!70!80!90!

100!

MV

/GA

G!

MV

/GA

G!

MV

/GA

G!

MV

/GA

G!

MV

/GA

G!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

Pr/G

AG!

MV

/Pr!

Pe/P

e!

Pe/P

r!

RNA

/Pr!

MM

/Pr!

Pr/P

e!

Pe/P

r!

Pr/P

r!

[111a]![111b]![111c]![111d]![111e]![111f]![111g]![111h]![111i]![111j]![111k]![111l]![111m]![111n]![111o]![111p]![120]![121]![113]![112]![122a]![122b]![122c]!

31! 34!

58!

97!

70!

50!

1!

22!

62!

12!

71!

19!

44!49!

77!

24!

43!

32!

11!17! 13!

36!

11!

RSE

(%)!

Reference!

29

The linear association between model variability and the KD value was investigated

using the Pearson’s correlation coefficient. The dataset was first tested for homo- and

heteroscedasticity, where RSE (%) was taken as the dependent variable and KD as

independent variable. The data were found to be homoscedastic by plotting the residuals

against the predicted values (Figure 4.4B). This observation was statistically confirmed by the

Breusch-Pagan heteroscedasticity test, whereby a significance level of 0.201 (χ2=1.634) was

obtained which is higher than 0.05, thus maintaining the null hypothesis: the data is

homoscedastic.

Figure 4.4: A. Scatterplot of the model variability (RSE%) and the dissociation constant (KD). B. Scatterplot of the residuals against the predicted values.

The Pearson correlation coefficient was found to be r=0.282, indicating no linear

association between the model variability and the KD value. This was statistically confirmed

with the Student’s t-test; r<rcrit,0.05/21=0.413, the null hypothesis (no significant correlation) is

maintained. In the scatterplot, there is also no pattern observed in the relationship between the

variables in the scatterplot (Figure 4.4A).

4.2 DEVELOPMENT OF A HGHAB-BASED SAW METHOD

4.2.1 Exploring ligand immobilisation strategies

4.2.1.1 Principles of the different immobilisation techniques

Amine coupling is the most used immobilisation strategy in the biosensor field (Figure

4.5A). The carboxyl groups on the surface are first activated by reaction of EDC/NHS,

0

20

40

60

80

100

0 5000 10000 15000

RSE

(%)

KD

A!

B

30

creating reactive succinimide intermediates. The ligand is then linked to the activated surface

via nucleophilic addition of primary amine functional groups, which results in a stable amide

bond. After immobilisation, the remaining active esters are blocked with a small amine-

containing molecule i.e. ethanolamine.

Aldehyde coupling is based on the binding of aldehydes to a hydrazide-modified

surface. Antibodies, or immunoglobulins are glycoproteins; they have a branched

oligosaccharide N-linked to asparagine 297 present in the Fc region. The oligosaccharide

moiety is located away from the antigen binding sites. Therefore immobilisation to the surface

is possible without affecting the reactivity of the antigen binding site [83]. First aldehyde

groups are introduced into the glycoproteins via oxidation of oligosaccharide moieties using

periodate, which oxidize vicinal diols, such as sialic acids (e.g. acetylneuramic acid) [84]. The

carboxylated chip surface is activated with EDC/NHS, followed by an injection of

carbohydrazide to create a surface with hydrazide groups. The oxidized antibodies containing

aldehyde groups are coupled to the modified surface via condensation of aldehydes with the

nucleophile hydrazide (Figure 4.5B).

Figure 4.5: Schemes of IgG immobilisation techniques. A. amine coupling, B. aldehyde coupling and C. capturing via protein G. (R1 = CH2CH3, R2 = (CH2)3N(CH3)2). Figure adapted from [85]

Another method for site-directed immobilisation can be achieved by capturing a

monoclonal antibody (mAb) using protein G, which is a bacterial protein known for its high

affinity to immunoglobulin G (IgG) [86, 87]. The protein G interacts specifically with the Fc

B"

C"

A"

31

domain of the antibody, keeping the antigen-binding region (Fab) free for interaction. One

molecule of protein G can bind maximum two hGHAb, because of steric hindrance [88].

Protein G is first immobilised to the dextran-gold surface via amine coupling chemistry. IgG

is captured with protein G by its Fc region allowing immobilisation in a site-direct manner

with the antigen-binding sites pointing towards the analyte (Figure 4.5C).

The immobilisation of a ligand in complex with the analyte is a relatively new concept

of site-directed immobilisation [89]. The complex is immobilised using amine coupling,

whereby the analyte occupies the analyte binding sites on the ligand and hence, preventing

amine groups to participate in covalent immobilisation on the surface. At the end of the

immobilisation, a regeneration step is included to dissociate the complex. The result would be

a surface immobilised with “oriented” ligand and analyte (Figure 4.6). This immobilisation

technique was performed with a 1:1 somatropin:hGHAb complex.

Figure 4.6: Principle of the immobilisation via somatropin:hGHAb complex

4.2.1.2 Quantitative evaluation of ligand immobilisation

The immobilised ligand density was determined for the different immobilisation

techniques. The immobilisation could be followed (real-time) in a sensorgram. An example of

an immobilisation sensorgram is shown Figure 4.7. In this case somatropin is immobilised to

the dextran surface via amine coupling. The first shift in the baseline occurs after the

activation with EDC/NHS (200-1000s), indicating a chemical modification on the sensor

surface. This was followed by three somatropin injections leading to increased signals upon

immobilisation of somatropin ligand (amine coupling). The final injection of ethanolamine

was used to quench the activated carboxyl-sites. The difference in phase signal after the

activation and at the end of the immobilisation gives an indication of the total amount of

bound ligand.

Interac(on*of*somatropin*(analyte)*to*somatropin:hGHAb*complex*

Theory:*coupling*of*complex*trough*amine>coupling**

After regeneration

Dextran surface!

Ab-somatropin complex

Orientated Ab

32

Figure 4.7: Sensorgram of the online immobilisation of somatropin using amine coupling.

The total amount of surface bound ligand and the theoretical amount of analyte that

can bind, is calculated for each immobilisation technique using formula (1) (Figure 4.8). The

binding stoichiometry of the interaction used for the calculations is f = 2 for immobilised

hGHAb and f = 0.5 for immobilised somatropin (as one antibody can thoeretically bind two

somatropin molecules).

The feasibility test could not be applied to the immobilization of the complex, because

the increased phase shift is a result of immobilization of both hGHAb and somatropin. In the

case of the immobilisation with protein G, the capturing of hGHAb via protein G leaded to a

significant descending drift of the baseline over time. Therefore, the amount of hGHAb was

calculated after the baseline had stabilised. According to the feasibility test, the calculated

amount of analyte that could bind exceeded the theoretical limit of detection of 0.5 pg/mm²

for each immobilisation technique.

The aldehyde coupling however, was not further included in the method development

due to the low amount of hGHAb that was immobilized and the different steps needed in the

chemical process for oxidation of the carbohydrate functions. Also it is known that it is

difficult to control the oxidation reaction conditions [90] and excessive oxidation might not

only affect the carbohydrate moiety but also amino acid residues [91].

33

Figure 4.8: An overview of the amount of immobilised ligand (blue) and ad hoc analyte (red) (n=5).

4.2.1.3 Quantitative evaluation of the analyte-ligand interaction and the non-specific binding

The interaction between analyte and immobilised ligand was studied for the different

immobilisation techniques. The detected phase shifts upon analyte interaction and normalised

response values (RR%) for the different immobilisation techniques are given in Table 4.1.

The non-specific binding was studied with injections of analyte BSA. The phase shift from

BSA was compared to the phase shift from ligand and expressed as a percentage non-specific

binding (%NSB). The difference between NSB and specific binding is illustrated in Figure

4.9.

Table 4.1: Quantitative evaluation of analyte and non-specific binding

Analyte(1) BSA

Phase shift (°) RR% Phase shift (°) NSB%

Amine coupling hGHAb (n=5) 0.28 ± 0.04 9.61 ± 1.59 0.24 ± 0.04 84.54 ± 6.85

Protein G capturing (n=4) 0.29 ± 0.05 12.70 ± 0.63 0.01 ± 0.01 2.80 ± 1.37

Complex (n=5) (2) 0.32 ± 0.17 N/A 0.32 ± 0.22 94.43 ± 28.88 Amine coupling

somatropin (n=2) 6.88 ± 0.77 66.73 ± 9.04 0.10 ± 0.01 1.47 ± 0.37

(1) 200 nM somatropin analyte was used for hGHAb bound surface, 100 nM hGHAb ligand was used for

somatropin bound surface. Idem for BSA analyte.

(2) RR% is not calculated as the amount of immobilised hGHAb is unknown.

200

42

8

57

59

144

26

194

0 50 100 150 200 250

Amine coupling - somatropin

Protein G capturing

Aldehyde coupling

Amine coupling - hGHAb

pg/mm2

Amount bound ligand Theoretical bound analyte

hGH

Ab

ligan

d So

mat

ropi

n

ligan

d

34

The immobilisation of hGHAb using protein G and immobilisation of somatropin

using amine coupling resulted in the highest specificity (i.e. lowest NSB%). However, the

normalised response values (RR%) show that amine coupling of somatropin result in the

highest active ligand surface compared to the protein G captured surface. Therefore the

immobilization technique that will be further used in the method development is the amine

coupling of somatropin.

Figure 4.9: Sensorgram of specific and non-specific binding of different immobilisation strategies. A. hGHAb-immobilised surface (amine coupling), B. Somatropin-immobilised surface (amine coupling).

4.2.2 Evaluation of the non-specific binding of analyte to dextran hydrogel sensor chip

The non-specific binding of somatropin, hGHAb and BSA to the dextran hydrogel

surface was evaluated (Figure 4.10).

Figure 4.10: Non-specific binding of BSA, hGHAb and somatropin to dextran hydrogel surface

-0,05

0

0,05

0,1

0,15

0,2

0,25

-200 300 800 1300

Phas

e (°

)

Time (s)

Somatropin BSA

-1

0

1

2

3

4

5

6

7

-200 300 800 1300

Phas

e (°

)

Time (s)

hGHAb BSA

-0,2

0

0,2

0,4

0,6

0,8

1

0 200 400 600 800 1000 1200 1400 1600

Phas

e sh

ift (°

)

Time (s)

BSA hGHAb Somatropin

A B

35

The maximum phase shifts at 550s observed for BSA, hGHAb and somatropin were

0.37 ± 0.15°, 0.29 ± 0.11° and 0.35 ± 0.14° respectively (n=3). The phase shift from the non-

specific interaction of hGHAb with the dextran hydrogel surface is significantly lower than

the phase shift of the specific interaction to the immobilised somatropin (0.29° ± 0.11° and

6.88 ± 0.77°, respectively).

4.2.3 Screening for regeneration of somatropin ligand

An appropriate regeneration procedure is required for the chosen immobilisation

technique. The regeneration of the somatropin-immobilised surface with 0.1% SDS was not

able to remove the bound hGHAb. The dissociation after the analyte injections was not

complete (i.e. signal did not return to baseline). It is necessary to include a regeneration step

after each analyte binding cycle to remove all bound analyte from the ligand, while

maintaining the ligands’ activity. Screening for an appropriate regeneration condition was

performed with 0.5% SDS, acetate buffer pH 4.5, 0.5 M NaCl, 5% ethyleenglycol, 1 mM

NaOH. None of those conditions was able to return the signal back to baseline (Figure

4.11A). A small drop was observed with 0.5 % SDS. A 1:1 mixture of 25 mM NaOH:0.05%

SDS was used as suggested in reference [92]. This regeneration procedure was able to

regenerate the surface without loss of activity (Figure 4.11B).

4.3 FUNCTIONAL CHARACTERIZATION OF NOTA-MODIFIED SOMATROPINS

The developed SAW method was used to determine the binding affinities and kinetics

of NOTA-modified somatropins for hGHAb.

4.3.1 Immobilization of somatropin and derivatives using amine coupling

Somatropin and derivatives were immobilized to the dextran-gold surface using amine

coupling and identical experimental parameters. The total amount of surface bound

(modified)-somatropin is given in Figure 4.12. The amount immobilised ligand is higher for

unmodified somatropin and 1:1 NOTA somatropin compared to 3:1 NOTA somatropin and

10:1 NOTA somatropin.

36

Figure 4.11: Screening for regeneration conditions. A. The regeneration efficiency (%) obtained from the different regeneration conditions, B. Evaluation of ligand activity after regeneration with NaOH+SDS.

Figure 4.12: Overview of the amount of immobilised somatropin derivatives (pg/mm2)

16 1,5 1,5 0,8

7

107

-10

10

30

50

70

90

110

0.1% SDS

NaOAc NaOH NaCl 0.5% SDS

SDS + NaOH

Reg

ener

atio

n ef

ficie

ncy

(%)

Regeneration condition

59

117

10

31

0

20

40

60

80

100

120

140

Somatropin 1:1 NOTA somatropin

3:1 NOTA somatropin

10:1 NOTA somatropin

pg/m

m2

Amount surface bound ligand

A

B

37

A

4.3.2 Binding experiments between immobilized somatropin (derivatives) and hGHAb

The interaction between somatropin (ligand), the NOTA-modified somatropins

(ligand) and hGHAb (analyte) was studied (Figure 4.14). Figure 4.13 shows the sensorgram

of channel 1.

To obtain the kinetic and affinity constants, kon, koff and KD are calculated using a one-

to-one binding model. Each curve in the sensorgram was fitted using a 1:1 binding model

(with R2 of more then 0.95%). Fitting of the association phase leads to an observed rate

constant kobs for each analyte concentration (Figure 4.13B). The KD was calculated by

dividing the weighted average koff by the kon from the association phase. Table 4.2 gives an

overview of the kinetic data and calculated KD.

Table 4.2: Affinity (KD) and kinetics (kon, koff) of the interaction between somatropin and hGHAb

Channel 1 Channel 3 kon (nM-1s-1) ± SE 1.08E-04 ± 6.51E-06 9.33E-05 ± 5.41E-06

koff (s-1) ± SE 1.59E-03 ± 1,81E-05 1.57E-03 ± 1,43E-05

KD (nM) ± SD 15 ± 1 17 ± 1

Figure 4.13: Somatropin and hGHAb interaction study. A. Overlay plot of phase shifts at different hGHAb concentrations. B. Linear regression of apparent rate constant in function of analyte concentration.

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

)200! 300! 800! 1300!

Phase*shift*(°)*

Time*(s)*

25!nM! 37.5!nM!50!nM! 100!nM!

y = 0,0001x + 0,0018 R² = 0,96758

0

0,002

0,004

0,006

0,008

0,01

0,012

0,014

0 50 100 150

k obs

(s-1

)

Concentration (nM)

B

38

Figure 4.14: Sensorgram of hGHAb on NOTA-modified somatropin immobilized surface (channel 5). A. 1:1 NOTA:somatropin ligand, B. 3:1 NOTA:somatropin ligand and C. 10:1 NOTA:somatropin ligand.

4.3.2.1 Binding experiment between somatropin (ligand) and hGHAb (analyte)

A average KD of 15.7 ± 1.6 nM was determined for the interaction between

somatropin and hGHAb (Table 4.2). The error of the fitting on the KD varies with a RSE

ranging from 5.8 to 6.1%. The variability (CV) between the different channels is 10.3%

(n=2).

-2

0

2

4

6

8

-200 0 200 400 600 800 1000 1200 1400

Phas

e (°

)

Time (s)

25 nM 37.5 nM 50 nM 75 nM 100 nM 100 nM

-0,1

0

0,1

0,2

0,3

0,4

-200 0 200 400 600 800 1000 1200 1400

Phas

e (°

)

Time (s) 25 nM 37.5 nM 50 nM 75 nM 100 nM 100 nM

-0,5 0

0,5 1

1,5 2

2,5

-200 0 200 400 600 800 1000 1200 1400

Phas

e (°

)

Time (s)

25 nM 37.5 nM 50 nM 75 nM 100 nM 100 nM

B

C

A

39

4.3.2.2 Binding experiment between 1:1 NOTA somatropin (ligand) and hGHAb (analyte)

The average KD is 19.0 ± 2.6 nM (n=5). The error of the fitting on the KD varies with

a RSE ranging from 7.1 to 8.4%. The variability (CV%) between the different channels is

13.5%.

4.3.2.3 Binding experiment between 3:1 NOTA somatropin (ligand) and hGHAb (analyte)

The average KD is 15.3 ± 1.1 nM (n=5). The error of the fitting on the KD varies with

a RSE ranging from 8.2 to 12.3%. The variability (CV%) between the different channels is

7.5%.

4.3.2.4 Binding experiment between 10:1 NOTA somatropin (ligand) and hGHAb (analyte)

The average KD of 15.4 ± 2.8 nM (n=5) was determined for the interaction between

10:1 NOTA somatropin and hGHAb. The error of the fitting for on the KD varies from a RSE

of 10.7 to 30.7%. The variability (CV%) between the different channels is 18.3%.

Table 4.3: Summary of binding constant and rate constants of somatropin and derivatives

KD ± SD kon ± SD koff ± SD

Unmodified somatropin (n=2) 15.7 ± 1.6 1.01E-04 ± 1.04E-05 1.58E-03 ± 1.39E-05

1:1 NOTA somatropin (n=5) 19.0 ± 2.6 3.96 E-05 ± 5.95E-06 7.50E-04 ± 1.53E-04

3:1 NOTA somatropin (n=5) 15.3 ± 1.1 8.84E-05 ± 6.85E-06 1.35E-03 ± 2.33E-05

10:1 NOTA somatropin (n=5) 15.4 ± 2.8 7.70E-05 ± 7.84E-06 1.17E-03 ± 1.15E-04

4.3.3 Specificity of the method

As a control experiment, the specificity of the binding assays was validated. The

interaction of the immobilised somatropin (derivatives) surfaces with BSA was evaluated.

The phase shift of BSA (100 nM) was compared to the phase shift of hGHAb (100 nM)

(Table 4.4) and expressed as a percentage non-specific binding (NSB%). The non-specific

interaction was less than 15% for all assays.

40

Table 4.4: Evaluation of the specificity of the method

Ligand Phase shift (°) 100 nM

BSA Phase shift (°) 100 nM

hGHAb % NSB

Somatropin (n=2) 0.10 ± 0.01 6.87 ± 0.63 1.44 ± 0.20

1:1 NOTA:somatropin (n=5) 0.23 ± 0.16 7.27 ± 0.73 3.21 ± 2.19

3:1 NOTA:somatropin (n=5)

0.05 ± 0.01 0.49 ± 0.08 10.90 ± 2.59

10:1 NOTA:somatropin (n=5) 0.18 ± 0.07 1.52 ± 0.39 11.56 ± 5.77

41

5 DISCUSSION

5.1 LITERATURE REVIEW - APPLICATIONS AND METHOD VARIABILITY

SAW biosensor literature was reviewed to gain more information about their current

status in research and to identify the analytical errors (KD variability and model variability)

among the study results. There are a few issues which must be kept in mind when considering

the research outcomes. Half of the publications from 2008 until now did not report a

dissociation constant. Especially for some experiments with in-house assembled biosensors,

no kinetic analysis was performed, hence, no KD was calculated. The aim of the affinity

determinations was mostly for qualitative purposes. The KD was determined to compare the

binding affinities of different compounds for a specific target, where conclusions were made

not statistically, but more intuitively as in ‘same order of magnitude’ or ‘nanomolar and

micromolar range’.

Secondly, an important assumption was made; when no number of experiments “n”

was reported in the publication, the reported standard error of the KD implied the statistics on

the fitting.

5.1.1 Random variability KD values

The investigation of the KD values variability generated a dataset that included only 3

experiments. As a result of this small set of data, no conclusions could be taken for the

variability of SAW biosensor experiments. More data are needed in order to take a

statistically conclusion about the variability of KD values as obtained by SAW methods. The

information about the variability of a SAW method would be valuable to set specifications

and to compare different KD values including answers to questions e.g. is the difference

between two or more KD values significant or caused by random errors in the measurements?

To give an idea about the variability of other label free binding assays; global

benchmark studies, investigating the variability of SPR biosensors, showed that the variability

was approximately 20% [93-95].

42

5.1.2 Model variability

The model variability (RSE%) was also investigated. There was no association was

found between the model variability and the size of KD value (strength of the binding

affinity). The model variability was found significantly large for some KD values (especially

for glycosaminoglycan interactions). The systematic large model variability could be the

result of an incorrect used fitting model. It is important to apply a suited binding model for

the interaction under study, as different models are possible [96]. In all assessed publications,

the 1:1 binding model was (in some cases wrongfully) applied using the Fitmaster® software

(SAW Instruments, Bonn, Germany).

5.2 DEVELOPMENT OF A HGHAB-BASED SAW METHOD

The choice of ligand immobilisation plays an important role in the analytical

performance of label-free biosensor assays. The signal magnitude, regenerability and stability

of the method depend on the amount of immobilised ligand, the remaining activity after

immobilisation and the orientation on the sensor chip surface, all characterized by the

immobilisation strategy [97].

As is the case in HPLC method development, also for SAW methods we need to

quantify the endpoint of the development effort. One of the quantitative criteria is the

feasibility test, which calculates the theoretically amount of analyte (pg/mm2) that can bind to

the surface bound ligand based on their molecular weights and binding stoichiometry. This

amount of analyte should exceed the limit of 0.5 pg/mm2 [98] to obtain detectable signals.

Secondly, ligand immobilization does not always lead to active ligand: covalent

immobilization can lead to inactivation or inaccessible analyte binding sites. Therefore, the

remaining activity of the ligand can be quantitatively compared using the relative ligand

response RR%. In other words, RR% is a normalised value to compare the binding capability

of the same ligand between different immobilisation techniques using a certain analyte

concentration. This value, however, does not necessarily provide information on the absolute

amount of active ligand. The absolute amount of active ligand can be calculated using analyte

concentrations that saturate the surface. From a practical and economical point of view, it is

not always feasible to obtain high analyte concentrations. Therefore, it is sufficient to use one

43

analyte concentration to compare the RR% between different immobilisation strategies during

method development. Thirdly, we have defined the specificity of the interaction expressed as

NSB% using a negative control protein (e.g. BSA). The NSB% expresses the proportion of an

analyte signal related to non-specific binding, such as interaction with the dextran layer and

gold. The NSB% should preferably be as low as possible. For the different ligands

(somatropin and derivatives) a 99% confidence interval was calculated for the NSB% which

amounted [0.08%;13.47%]. The NSB% of the proposed method should not be higher than

15%. Moreover, the choice of chip chemistry is decisive for the NSB%. The fourth criteria

quantitatively evaluates the regeneration between different analyte injection cycles using the

regeneration efficiency (RE%) parameter and additional ligand activity testings. The RE%

should fall within 90-110%.

5.2.1 Exploring immobilisation procedures for the immobilisation of antibodies

Different immobilisation procedures have been described for proteins and antibodies,

which can be categorised in two main groups: random immobilisation and site-directed

immobilisation. The most widely used immobilisation technique is the random and covalent

immobilisation using amine coupling. The method is easy in use and leads to a high

immobilisation density. For hGHAb, a ligand density of 194 pg/mm2 is reported, which is

lower than 2.5 ng/mm2 reported for a close-packed side-on monolayer of antibodies [97, 99,

100]. Moreover, direct amine coupling of hGHAb doesn’t ensure that all the antigen-binding

sites are available for analyte binding (Figure 5.1), hence, the random orientation of the

ligands often results in the loss of antigen binding capability [101]. The specificity of the

direct immobilisation of hGHAb was significantly low. At least 84% of the response is the

result of NSB to the CM-dextran hydrogel layer, which also results in the relatively low

activity/binding capacity of the immobilised hGHAb (RR%= 9.61 ± 1.59% using 200 nM

somatropin).

The density of immobilised hGHAb by aldehyde coupling was extremely low (26.2 ±

10.3 pg/mm2). Several factors could be responsible for the low efficiency; (i) low ligand

concentration used during immobilisation (11.7 µg/ml) or, (ii) insufficient oxidation of the

ligand. The latter could be improved by increasing the time of oxidation, the oxidants

concentration or the temperature of the reaction [90]. This method was not further included in

the method development.

44

Figure 5.1: Schematic overview of random immobilisation between hGHAb and the dextran layer. (A) All antigen binding sites are still available for analyte binding, (B) no analyte binding sites are available, (C) limited antigen binding sites are available and (D) non-covalently bound antibodies with (limited) available antigen binding sites.

The site-directed immobilisation procedures that were applied for hGHAb did not

yield in higher surface densities. The density of hGHAb captured via protein G was initially

higher than the random immobilised hGHAb. However, immediately after immobilisation, a

large amount of hGHAb was released from the surface, as indicated by the descending drift in

the baseline. The phase difference between the stabilized baseline and the response directly

after the immobilisation was 5.6 ± 0.3° (i.e. 108.7 ± 11.7 pg/mm2). It is most likely that the

captured hGHAb was dissociating from the protein G as the interaction with hGHAb is a non-

covalent affinity based interaction.

The distance between the CM-dextran hydrogel surface and the sample flow increases

when a linker molecule like protein G is used. This explains the lower adsorption effects to

the sensor surface and the lower NSB (NSB% = 2.80 ± 1.37%). Moreover, the recombinant

protein G that was used is truncated (i.e. the BSA binding sites are deleted); no interaction

with BSA is theoretically possible. According to [85] and [97], this orientated immobilisation

approach results in the highest binding capacity. Yet, the binding capacity of hGHAb

captured via protein G was unexpectedly low (RR% = 12.70 ± 0.63%, using 200 nM

somatropin). Most likely this is caused by (i) the serious dissociation of the hGHAb-protein G

complex after immobilisation, and (ii) the rapid saturation of the ligand surface without use of

regeneration. The drift in the baseline after immobilisation should be controlled. In that case

more immobilised hGHAb is available for interaction. The optimal buffer condition for IgG -

protein G binding is pH 5. Therefore, a running buffer of pH 5 is recommended for binding of

the antibody to protein G. However, the stability of hGHAb at lower pH range is unknown.

45

Protein A on the other hand needs a buffer of pH 8 for optimal binding, which is more close

to the physiological conditions [102].

The amount of bound orientated hGHAb using complexation couldn’t be determined

because of the ligands’ heterogeneity. It was assumed that the amount of hGHAb will exceed

the limit of detection as immobilisation using amine coupling chemistry results in a high

concentration of immobilised ligands. The results regarding ligand binding capability (RR%)

and specificity (NSB%) are similar with amine coupling of hGHAb.

So far, the immobilisation with hGHAb did not respond to our expectations.

Somatropin has several functional groups (-NH2) that can be involved in amine coupling: nine

lysine residues (K38, K41, K70, K115, K140, K145, K158, K168 and K172) (Figure 5.3) and

the amine-terminus. This makes somatropin an ideal candidate for amine coupling.

Figure 5.2: Crystal structure of recombinant human growth hormone (PDB:3HHR). The lysine residues are indicated in red.

The mass density of immobilised somatropin was lower compared to immobilised

hGHAb. However, the molecular density of immobilised somatropin (∼ 2.7 nmol/mm2) was

twice as high as the amount of immobilised hGHAb (∼ 1.3 nmol/mm2). This could be

explained by the fact that somatropin (Mw 22 125 Da) is a smaller protein than hGHAb (Mw

±150 000 Da), and therefore more molecules will be able to occupy the same surface. The

relative response of the somatropin ligand was higher than for the hGHAb methods and the

NSB was very low (NSB% = 1.5% ± 0.4), making it an attractive method for further

development.

46

5.2.2 Screening for optimal regeneration conditions

In normal circumstances, regeneration of the surface is required to remove the bound

analytes to recover the ligand density and to avoid saturation of the surface. Important is the

maintenance of the bioactivity of the ligand. A quantitative measure for the evaluation of the

regeneration condition can be given by calculation of the regeneration efficiency (RE%).

This should approach 100% as good as possible and must be within [90-110%]. If the

efficiency is lower than 90%, the conditions are weak and were not able to remove all bound

analytes from the immobilized surface. Conversely if the efficiency is higher than 110% the

conditions are too strong, resulting in ligand dissociation.

A solution with a mixture of 50 nM NaOH and 0.1% SDS was found to be an

appropriate regeneration condition to remove all the bound hGHAb from the immobilised

somatropin surface. SDS is a detergent, which binds to proteins by hydrophobic interactions

[103]. Alkali solutions like NaOH will break the interaction between analyte and ligand. The

high pH will negatively charge the protein binding sites, which will dissociate the analyte –

ligand interaction because of electrostatic repulsion [104].

5.3 FUNCTIONAL CHARACTERIZATION OF NOTA-MODIFIED SOMATROPINS

The functional quality of NOTA-modified somatropins was quantitatively

characterized using the developed hGHAb-based SAW method.

5.3.1 Immobilization of somatropin and derivatives using amine coupling

Somatropin and the NOTA-modified somatropin were immobilised on the dextran-

gold surface using amine coupling. It is expected that the amount of immobilised NOTA-

modified somatropin will be lower compared to somatropin. NOTA-modification involves

amine groups of the lysine residues, therefore less lysines will be available for the

immobilisation via amine coupling. The immobilisation of 1:1 NOTA somatropin was

similar to the immobilisation of unmodified somatropin. The high immobilisation efficiency

is due to the significant presence of unmodified somatropin (46%) in the 1:1 NOTA

somatropin sample (Table 5.1).

47

Table 5.1: Overview of the substitution degree (%) of NOTA-modified somatropin samples [105].

Unmodified +1 NOTA +2 NOTA +3 NOTA +4 NOTA 1:1 NOTA somatropin 46 42 12 ND ND 3:1 NOTA somatropin 13 45 35 7 ND

10:1 NOTA somatropin ND 2 39 47 12 ND: Not detectable

The immobilisation of 3:1 NOTA somatropin was similar to 10:1 NOTA

somatropin. Both have a lower total phase shift after the immobilisation. This can be due to

(i) the use of different chips with different chemistry batches and (ii) a reduced amount of

lysine residues for coupling (Table 5.1).

As previously mentioned, the quality of a sensor chip is essential to obtain reliable

measurements. The chips are reused; therefore the sensor surface is chemical etched and re-

modified with a CM-dextran hydrogel layer. Heterogeneity of the CM-dextran hydrogel can

occur, not only between different chips but also on the same sensor chip, density of the

dextran hydrogel is different on different channels. This leads to variability of the data

between different channels. The state of the plain gold surface has a major impact on the

modification. One sensor chip is used more than the other and the more a chip is chemically

etched, the more likely the sensitive areas may be damaged by erosion of gold and the

physical properties (e.g. wettability) may have changed [106]. Validation of the chip quality is

important to assure reproducible measurements. Characterization of a golden surface is

achieved by the investigating the electrochemical activity and elemental composition of the

surface [107].

5.3.2 Binding experiments between immobilized somatropin (-derivatives) and hGHAb

After the immobilization, the binding experiments were performed to determine the

dissociation constants of hGHAb to somatropin and NOTA modified somatropins.

The association rate constants for typical antibody-antigen interactions are in the range

of 105 - 106 M-1s-1 [108]. Our data indicate a slightly slower association rate of the hGHAb to

the immobilized somatropin surface (kon = 1.01 × 10-4 nM−1s-1). The association rate slightly

decreases with the amount of modified somatropin (3.96 × 10-5 nM−1s-1, 8.84 × 10-5 M−1s-1

and 7.70 × 10-5 nM−1s-1 respectively for 1:1, 3:1 and 10:1 NOTA somatropin). The

dissociation rate constants of the modified somatropins were found in the same range as for

48

somatropin (from 7.50 × 10-4 s-1 to 1.17 × 10-3 s-1). The low dissociation rate constants

suggest a relatively stable complex of the immobilized somatropins and hGHAb. Both rate

constants fall in the range of most protein-protein interactions [109]. Overall, these data

demonstrate a very high affinity and specific interaction between somatropin (derivatives) and

hGHAb.

For the kinetic evaluation of the data an appropriate binding model should be chosen.

The choice should be made rationally, e.g. a 1:1 binding model would not be a logical choice

for cooperative interactions; and the model should be evaluated statistically using “goodness

of fit” parameters and standard errors. In most antigen-antibody binding experiments the 1:1

(Langmuir) model is applied [95, 110]. The antigen binding sites of the antibodies are

assumed to be independent of each other; the binding on one Fab fragment will not interact

with the binding of the other Fab fragment.

For the kinetic evaluation, a 1:1 binding model was applied for the hGHAb-

somatropin interaction and the hGHAb-NOTA modified somatropin interaction. The RSE of

the KD-values were below 10% for somatropin and 1:1 NOTA somatropin. However for 3:1

NOTA somatropin and 10:1 NOTA somatropin, the RSE was not always below 10%.

Because the large SE gave an uncertain value of the KD, the average KD could not be

determined with high precision for 10:1 NOTA somatropin.

We determined the KD of the somatropin-hGHAb interaction to be ~ 16 nM, which is

in the range of the high affinity antibodies (low nanomolar – high picomolar range). The KD

values of the somatropin derivatives are in the same order of magnitude (Figure 5.3) and are

not statistically significant different (ANOVA; p>0.05). Thus, NOTA-modification does not

alter the binding affinity toward hGHAb. No significant differences are found in the

binding properties between the products of different synthesis procedures.

49

Figure 5.3: 95% CI of the KD of the somatropin and the different NOTA-modifications of somatropin

0!

5!

10!

15!

20!

25!

Unmodified! 1-1 NOTAsomatropin! 3-1 NOTAsomatropin! 10-1 NOTAsomatropin!

50

6 CONCLUSION

SAW biosensors are currently been applied in divergent areas. SAW biosensors are

particularly used for qualitative purpose while the quantitative aspects are underutilized. A

thorough literature search was performed concerning SAW biosensor technology. Today,

there are insufficient publications that report statistical data to draw a clear conclusion about

the general repeatability of a SAW method.

In this study five different immobilisation techniques were compared and evaluated to

determine the most appropriate SAW method. We have defined several quantitative criteria

for method development including the evaluation of different immobilisation strategies (the

ligand immobilisation density, binding capability (RR%) and specificity (NSB%)), as well as

the evaluation of the regeneration conditions. The covalent amine immobilisation of

somatropin was considered as the best immobilisation method (59 pg/mm2, 66.7% RR% and

1.5% NSB%). A suited regeneration condition was determined using the regeneration

efficiency (RE%), as well as an additional ligand activity testing. The condition combining

SDS and NaOH were able to return the signal back to baseline after a hGHAb injection

without affecting the activity of the immobilised ligands.

The developed SAW method was used to characterize the functional quality of

NOTA-modified somatropins. The NOTA-modification does not alter the binding affinity of

somatropin toward hGHAb. The binding properties of the different synthesized NOTA-

somatropin (1:1, 3:1 and 10:1) were not significantly different from each other. The hGHAb

was successfully used as a system to investigate the functional quality of NOTA-modified

somatropins.

The SAW biosensor is a very promising instrument to act as a functional quality

characterization tool in the development and quality control. However, a validation of real time

binding assays is absolutely necessary to introduce the method as part of the product release

portfolio and regulatory approval of new biotechnological drugs.

51

7 REFERENCES

1. Lawrence, S., Billion dollar babies - biotech drugs as blockbusters. Nature

biotechnology, 2007. 25(4): p. 380-386.

2. European Medicines Agency, Guideline on similar biological medicinal products.

CHMP/437/04

3. Berkowitz, S.A., et al., Analytical tools for characterizing biopharmaceuticals and the

implications for biosimilars. Nat Rev Drug Discov, 2012. 11(7): p. 527-40.

4. Greer F. Analytical characterization of biosimilar products to establish biosimilarity. .

05/01/2014]; Available from:

http://www.sgs.com/~/media/Global/Documents/Brochures/SGS%20Lab%20Biosimil

ar%20Characterization%20Poster%20EN%2013.ashx.

5. European Medicines Agency, ICH guideline S6 (R1) – preclinical safety evaluation of

biotechnology-derived pharmaceuticals. EMA/CHMP/ICH/731268/1998.

6. European Medicines Agency, Guideline on Similar Biological Medicinal Products

containing biotechnology-derived proteins as active Substance: non-clinical and

clinical issues. EMEA/CHMP/BMWP/42832/2005.

7. Leysen J., et al., Receptors: Binding Assays. Stolerman I., Price L. (Ed.) Encyclopedia

of Psychopharmacology: SpringerReference (www.springerreference.com), 2010.

8. Pierce, M.M., C.S. Raman, and B.T. Nall, Isothermal titration calorimetry of protein-

protein interactions. Methods, 1999. 19(2): p. 213-21.

9. Cooper, M.A., Label-free screening of bio-molecular interactions. Analytical and

bioanalytical chemistry, 2003. 377(5): p. 834-842.

10. Jecklin, M.C., et al., Label-free determination of protein-ligand binding constants

using mass spectrometry and validation using surface plasmon resonance and

isothermal titration calorimetry. Journal of Molecular Recognition, 2009. 22(4): p.

319-329.

11. Liang, Y., Applications of isothermal titration calorimetry in protein science. Acta

biochimica et biophysica Sinica, 2008. 40(7): p. 565-576.

12. Roselin, L.S., et al., Recent trends and some applications of isothermal titration

calorimetry in biotechnology. Biotechnology journal, 2010. 5(1): p. 85-98.

13. Fielding, L., NMR methods for the determination of protein-ligand dissociation

constants. Current topics in medicinal chemistry, 2003. 3(1): p. 39-53.

52

14. Sauter, N.K., et al., Binding of influenza virus hemagglutinin to analogs of its cell-

surface receptor, sialic acid: analysis by proton nuclear magnetic resonance

spectroscopy and X-ray crystallography. Biochemistry, 1992. 31(40): p. 9609-9621.

15. Rocha-Gaso, M.I., et al., Surface generated acoustic wave biosensors for the detection

of pathogens: a review. Sensors (Basel), 2009. 9(7): p. 5740-69.

16. Loot, A.E., et al., The suitability of iodinated Angiotensin-(1–7) peptides as

pharmacological tools. Journal of Pharmacological and Toxicological Methods, 2005.

51(1): p. 51-55.

17. Homola, J.ô.Ã., S.S. Yee, and G.n. Gauglitz, Surface plasmon resonance sensors:

review. Sensors and Actuators B: Chemical, 1999. 54(1): p. 3-15.

18. Cooper, M.A., Optical biosensors in drug discovery. Nat Rev Drug Discov, 2002.

1(7): p. 515-28.

19. Lange, K., B.E. Rapp, and M. Rapp, Surface acoustic wave biosensors: a review.

Analytical and Bioanalytical Chemistry, 2008. 391(5): p. 1509-1519.

20. Perpeet, M., et al., SAW sensor system for marker-free molecular interaction analysis.

Analytical Letters, 2006. 39(8): p. 1747-1757.

21. Drafts, B., Acoustic wave technology sensors. Ieee Transactions on Microwave Theory

and Techniques, 2001. 49(4): p. 795-802.

22. Klauke, T.N., et al., Measurement of porcine haptoglobin in meat juice using surface

acoustic wave biosensor technology. Meat Science, 2013. 95(3): p. 699-703.

23. Gronewold, T.M.A., et al., Discrimination of single mutations in cancer-related gene

fragments with a surface acoustic wave sensor. Analytical Chemistry, 2006. 78(14): p.

4865-4871.

24. Blasche, S., et al., The E. coli Effector Protein NleF Is a Caspase Inhibitor. Plos One,

2013. 8(3).

25. Fournel, F., et al., Love wave biosensor for real-time detection of okadaic acid as DSP

phycotoxin. Sensors and Actuators B-Chemical, 2012. 170: p. 122-128.

26. Love, T.E., C. Redmond, and C.N. Mayers, Real time detection of anthrax spores

using highly specific anti-EA1 recombinant antibodies produced by competitive

panning. J Immunol Methods, 2008. 334(1-2): p. 1-10.

27. Krishnamoorthy, S., et al., An interleukin-6 ZnO/SiO2/Si surface acoustic wave

biosensor. Biosensors & Bioelectronics, 2008. 24(2): p. 313-318.

28. Fang, Y., Label-free cell-based assays with optical biosensors in drug discovery.

Assay Drug Dev Technol, 2006. 4(5): p. 583-95.

53

29. Myszka, D.G. and R.L. Rich, Implementing surface plasmon resonance biosensors in

drug discovery. Pharm Sci Technolo Today, 2000. 3(9): p. 310-317.

30. Smith, A.J.T., et al., Beyond Picomolar Affinities: Quantitative Aspects of

Noncovalent and Covalent Binding of Drugs to Proteins. J Med Chem, 2009. 52(2): p.

225-33.

31. Kannisto, K., L. Murtomäki, and T. Viitala, An impedance QCM study on the

partitioning of bioactive compounds in supported phospholipid bilayers. Colloids and

Surfaces B: Biointerfaces, 2011. 86(2): p. 298-304.

32. Frostell-Karlsson, A., et al., Biosensor analysis of the interaction between immobilized

human serum albumin and drug compounds for prediction of human serum albumin

binding levels. J Med Chem, 2000. 43(10): p. 1986-92.

33. Fatisson, J., F. Azari, and N. Tufenkji, Real-time QCM-D monitoring of cellular

responses to different cytomorphic agents. Biosensors and Bioelectronics, 2011.

26(7): p. 3207-3212.

34. Dixit, N., K.M. Maloney, and D.S. Kalonia, Application of quartz crystal

microbalance to study the impact of pH and ionic strength on protein–silicone oil

interactions. International Journal of Pharmaceutics, 2011. 412(1–2): p. 20-27.

35. Jacquemart, R., et al., At-line monitoring of bioreactor protein production by Surface

Plasmon Resonance. Biotechnol Bioeng, 2008. 100(1): p. 184-8.

36. Thillaivinayagalingam, P., et al., Biopharmaceutical production: Applications of

surface plasmon resonance biosensors. J Chromatogr B Analyt Technol Biomed Life

Sci, 2010. 878(2): p. 149-53.

37. Wang, H., et al., Development of biosensor-based SPR technology for biological

quantification and quality control of pharmaceutical proteins. J Pharm Biomed Anal,

2009. 50(5): p. 1026-9.

38. European Medicines Agency. European public assesment reports. 2013 01/01/2014];

Available from: http://www.ema.europa.eu/.

39. Koch, T.K., et al., Creutzfeldt-Jakob disease in a young adult with idiopathic

hypopituitarism: possible relation to the administration of cadaveric human growth

hormone. The New England journal of medicine, 1985. 313(12): p. 731-733.

40. Okada, S. and J.J. Kopchick, Biological effects of growth hormone and its antagonist.

Trends in molecular medicine, 2001. 7(3): p. 126-132.

54

41. Bristow, A.F. and A.M. Jespersen, The Second International Standard for somatropin

(recombinant DNA-derived human growth hormone): preparation and calibration in

an international collaborative study. Biologicals, 2001. 29(2): p. 97-106.

42. Kronenberg, H.M. and R.H. Williams, Williams Textbook of Endocrinology

(Online)2008: Saunders/Elsevier.

43. Hardin, D.S., Treatment of short stature and growth hormone deficiency in children

with somatotropin (rDNA origin). Biologics, 2008. 2(4): p. 655-61.

44. WADA and List Committee, The 2013 Prohibited List. International standard. The

World Anti-Doping Code, 2013: p. 1-9.

45. Diamanti-Kandarakis, E., et al., Hormones in sports: growth hormone abuse.

Hormones (Athens), 2004. 3(1): p. 37-45.

46. He, C. and M. Wu, Detection of doping with recombinant human growth hormone.

Bioanalysis, 2009. 1(5): p. 953-65.

47. Brennan, B.P., et al., Human growth hormone abuse in male weightlifters. Am J

Addict, 2011. 20(1): p. 9-13.

48. Olshansky, S. and T.T. Perls, New developments in the illegal provision of growth

hormone for anti-aging and bodybuilding. JAMA, 2008. 299(23): p. 2792-2794.

49. Graham, M.R., et al., Counterfeiting in performance- and image-enhancing drugs.

Drug Testing and Analysis, 2009. 1(3): p. 135-142.

50. Clayton, P.E., et al., Growth hormone, the insulin-like growth factor axis, insulin and

cancer risk. Nat Rev Endocrinol, 2011. 7(1): p. 11-24.

51. Jenkins, P.J., A. Mukherjee, and S.M. Shalet, Does growth hormone cause cancer?

Clin Endocrinol (Oxf), 2006. 64(2): p. 115-21.

52. Chopin, L.K., et al., Co-expression of GH and GHR isoforms in prostate cancer cell

lines. Growth Horm IGF Res, 2002. 12(2): p. 126-36.

53. Weiss-Messer, E., et al., Growth hormone (GH) receptors in prostate cancer: gene

expression in human tissues and cell lines and characterization, GH signaling and

androgen receptor regulation in LNCaP cells. Mol Cell Endocrinol, 2004. 220(1-2):

p. 109-23.

54. Gebre-Medhin, M., et al., Growth Hormone Receptor Is Expressed in Human Breast

Cancer. The American Journal of Pathology, 2001. 158(4): p. 1217-1222.

55. Yang, X., et al., Growth Hormone Receptor Expression in Human Colorectal Cancer.

Digestive Diseases and Sciences, 2004. 49(9): p. 1493-1498.

55

56. Wu, X., et al., Growth Hormone Receptor Expression is Up-Regulated During

Tumorigenesis of Human Colorectal Cancer. Journal of Surgical Research, 2007.

143(2): p. 294-299.

57. Kabeer, M.A., et al., Expression of growth hormone receptors in colorectal

carcinoma. Diagnostic Histopathology, 2011. 17(8): p. 355-361.

58. Yang, X., et al., Growth hormone receptor expression in human primary gastric

adenocarcinoma. Journal of Biomedical Research, 2012. 26(5): p. 307-314.

59. Denmeade, S.R. and J.T. Isaacs, A history of prostate cancer treatment. Nature

Reviews Cancer, 2002. 2(5): p. 389-396.

60. Bidosee, M., et al., Regulation of growth hormone receptors in human prostate cancer

cell lines. Molecular and Cellular Endocrinology, 2009. 309(1): p. 82-92.

61. Bidosee, M., et al., Growth hormone affects gene expression and proliferation in

human prostate cancer cells. Int J Androl, 2011. 34(2): p. 124-37.

62. Perry, J.K., et al., The contribution of growth hormone to mammary neoplasia. J

Mammary Gland Biol Neoplasia, 2008. 13(1): p. 131-45.

63. Williams, C. and C.Y. Lin, Oestrogen receptors in breast cancer: basic mechanisms

and clinical implications. Ecancermedicalscience, 2013. 7: p. 370.

64. Anderson, E., The role of oestrogen and progesterone receptors in human mammary

development and tumorigenesis. Breast Cancer Research, 2002. 4(5): p. 197-201.

65. Zatelli, M.C., et al., Growth hormone excess promotes breast cancer chemoresistance.

J Clin Endocrinol Metab, 2009. 94(10): p. 3931-8.

66. Brunet-Dunand, S.E., et al., Autocrine human growth hormone promotes tumor

angiogenesis in mammary carcinoma. Endocrinology, 2009. 150(3): p. 1341-52.

67. Xu, J., et al., Growth hormone signaling in human T47D breast cancer cells: potential

role for a growth hormone receptor-prolactin receptor complex. Mol Endocrinol,

2011. 25(4): p. 597-610.

68. Turaga, K.K. and L.K. Kvols, Recent progress in the understanding, diagnosis, and

treatment of gastroenteropancreatic neuroendocrine tumors. CA: A Cancer Journal

for Clinicians, 2011. 61(2): p. 113-132.

69. Öberg, K.E., et al., Role of Somatostatins in Gastroenteropancreatic Neuroendocrine

Tumor Development and Therapy. Gastroenterology, 2010. 139(3): p. 742-753.e1.

70. ClinicalTrial.gov. DOTA-TOC in Metastasized Neuroendocrine Cancer. 17/05/2014];

Available from: http://clinicaltrials.gov/ct2/show/study/NCT00978211.

56

71. Wang, L., et al., Somatostatin receptor-based molecular imaging and therapy for

neuroendocrine tumors. Biomed Res Int, 2013. 2013: p. 102819.

72. JUKIĆ, Z., et al., hGH and GHR Expression in Large Cell Neuroendocrine

Carcinoma of the Colon and Rectum. Anticancer Research, 2012. 32(8): p. 3377-3381.

73. Goldenberg, D.M. and R.M. Sharkey, Novel radiolabeled antibody conjugates.

Oncogene, 2007. 26(25): p. 3734-44.

74. Wynendaele, E., et al., Development of Peptide and Protein Based

Radiopharmaceuticals. Curr Pharm Des, 2013.

75. Fani, M., H.R. Maecke, and S.M. Okarvi, Radiolabeled peptides: valuable tools for

the detection and treatment of cancer. Theranostics, 2012. 2(5): p. 481-501.

76. Hermanson, G.T., Bioconjugate techniques1996: Academic press.

77. Harding, P.A., et al., Growth hormone (GH) and a GH antagonist promote GH

receptor dimerization and internalization. Journal of Biological Chemistry, 1996.

271(12): p. 6708-6712.

78. Vashist, S.K., A method for regenerating gold surface for prolonged reuse of gold-

coated surface plasmon resonance chip. Analytical Biochemistry, 2012. 423(1): p. 23-

25.

79. Lofas, S., Dextran Modified Self-Assembled Monolayer Surfaces for Use in

Biointeraction Analysis with Surface-Plasmon Resonance. Pure and Applied

Chemistry, 1995. 67(5): p. 829-834.

80. Buyst E., Characterizing binding affinity of somatropin and derived structures to

hGHAb by surface acoustic waves and size exclusion chromatography, in

DruQuaR2013, UGhent.

81. Morton, T.A., D.G. Myszka, and I.M. Chaiken, Interpreting Complex Binding-

Kinetics from Optical Biosensors - a Comparison of Analysis by Linearization, the

Integrated Rate-Equation, and Numerical-Integration. Analytical Biochemistry, 1995.

227(1): p. 176-185.

82. Karlsson, R., A. Michaelsson, and L. Mattsson, Kinetic-Analysis of Monoclonal

Antibody-Antigen Interactions with a New Biosensor Based Analytical System. Journal

of Immunological Methods, 1991. 145(1-2): p. 229-240.

83. Sutton, B.J. and D.C. Phillips, The 3-Dimensional Structure of the Carbohydrate

within the Fc-Fragment of Immunoglobulin-G. Biochemical Society Transactions,

1983. 11(2): p. 130-132.

57

84. O'Shannessy, D.J. and M. Wilchek, Immobilization of glycoconjugates by their

oligosaccharides: use of hydrazido-derivatized matrices. Anal Biochem, 1990. 191(1):

p. 1-8.

85. Kausaite-Minkstimiene, A., et al., Comparative Study of Random and Oriented

Antibody Immobilization Techniques on the Binding Capacity of Immunosensor. Anal

Chem, 2010. 82(15): p. 6401-6408.

86. Bjorck, L. and G. Kronvall, Purification and Some Properties of Streptococcal

Protein-G, Protein-a Novel Igg-Binding Reagent. Journal of Immunology, 1984.

133(2): p. 969-974.

87. Akerstrom, B., et al., Protein-G - a Powerful Tool for Binding and Detection of

Monoclonal and Polyclonal Antibodies. Journal of Immunology, 1985. 135(4): p.

2589-2592.

88. Danczyk, R., et al., Comparison of antibody functionality using different

immobilization methods. Biotechnol Bioeng, 2003. 84(2): p. 215-23.

89. Yoon, M., H.J. Hwang, and J.H. Kim, Immobilization of antibodies on the self-

assembled monolayer by antigen-binding site protection and immobilization kinetic

control. Journal of Biomedical Science and Engineering, 2011. 4(04): p. 242.

90. Wolfe, C.A.C. and D.S. Hage, Studies on the Rate and Control of Antibody Oxidation

by Periodate. Anal Biochem, 1995. 231(1): p. 123-130.

91. Clamp, J.R. and L. Hough, Periodate Oxidation of Amino Acids with Reference to

Studies on Glycoproteins. Biochemical Journal, 1965. 94(1): p. 17-&.

92. Kausaite-Minkstimiene, A., et al., Comparative Study of Random and Oriented

Antibody Immobilization Techniques on the Binding Capacity of Immunosensor.

Analytical Chemistry, 2010. 82(15): p. 6401-6408.

93. Rich, R.L., et al., A global benchmark study using affinity-based biosensors.

Analytical biochemistry, 2009. 386(2): p. 194-216.

94. Cannon, M.J., et al., Comparative analyses of a small molecule/enzyme interaction by

multiple users of Biacore technology. Analytical biochemistry, 2004. 330(1): p. 98-

113.

95. Katsamba, P.S., et al., Kinetic analysis of a high-affinity antibody/antigen interaction

performed by multiple Biacore users. Anal Biochem, 2006. 352(2): p. 208-21.

96. Danielson, U.H., Integrating surface plasmon resonance biosensor-based interaction

kinetic analyses into the lead discovery and optimization process. Future Medicinal

Chemistry 2009. 1(8): p. 1399-1414.

58

97. Vashist, S.K., et al., Effect of antibody immobilization strategies on the analytical

performance of a surface plasmon resonance-based immunoassay. Analyst, 2011.

136(21): p. 4431-6.

98. Schlensog, M.D., et al., A Love-wave biosensor using nucleic acids as ligands.

Sensors and Actuators B-Chemical, 2004. 101(3): p. 308-315.

99. Baszkin, A. and D.J. Lyman, The Interaction of Plasma-Proteins with Polymers .1.

Relationship between Polymer Surface-Energy and Protein Adsorption-Desorption.

Journal of Biomedical Materials Research, 1980. 14(4): p. 393-403.

100. Lofas, S., et al., Dextran Modified Gold Surfaces for Surface-Plasmon Resonance

Sensors - Immunoreactivity of Immobilized Antibodies and Antibody Surface

Interaction Studies. Colloids and Surfaces B-Biointerfaces, 1993. 1(2): p. 83-89.

101. O'Shannessy, D.J., M. Brigham-Burke, and K. Peck, Immobilization chemistries

suitable for use in the BIAcore surface plasmon resonance detector. Anal Biochem,

1992. 205(1): p. 132-6.

102. Wright, C., et al., The interaction of protein A and Fc fragment of rabbit

immunoglobulin G as probed by complement-fixation and nuclear-magnetic-

resonance studies. Biochem. J., 1977. 167(3): p. 661-668.

103. Bhuyan, A.K., On the mechanism of SDS-induced protein denaturation. Biopolymers,

2010. 93(2): p. 186-99.

104. Andersson, K., M. Hamalainen, and M. Malmqvist, Identification and optimization of

regeneration conditions for affinity-based biosensor assays. A multivariate cocktail

approach. Anal Chem, 1999. 71(13): p. 2475-81.

105. Bracke, N., et al., Analytical characerization of NOTA-modified somatropins. 2014.

106. Bhalla, V., et al., Chip cleaning and regeneration for electrochemical sensor arrays.

Thin Solid Films, 2010. 518(12): p. 3360-3366.

107. Fischer, L.M., et al., Gold cleaning methods for electrochemical detection

applications. Microelectronic Engineering, 2009. 86(4–6): p. 1282-1285.

108. Kumagai, I. and K. Tsumoto, Antigen–Antibody Binding, in eLS2001, John Wiley &

Sons, Ltd.

109. Mason, D.W. and A.F. Williams, The kinetics of antibody binding to membrane

antigens in solution and at the cell surface. Biochem. J, 1980. 187: p. 1-20.

110. Bee, C., et al., Determining the Binding Affinity of Therapeutic Monoclonal

Antibodies towards Their Native Unpurified Antigens in Human Serum. PLoS ONE,

2013. 8(11): p. e80501.

59

111. Schlesinger, M., et al., The inhibition of the integrin VLA-4 in MV3 melanoma cell

binding by non-anticoagulant heparin derivatives. Thrombosis Research, 2012.

129(5): p. 603-610.

112. Reder-Christ, K., et al., A dry membrane protection technique to allow Surface

Acoustic Wave biosensor measurements of biological model membrane approaches.

Sensors, 2013. 13(9): p. 12392-12405.

113. Gronewold, T.M.A., et al., Monitoring complex formation in the blood-coagulation

cascade using aptamer-coated SAW sensors. Biosens Bioelectron, 2005. 20(10): p.

2044-2052.

114. Stumbaum, M. and U. Reusch, Membrane-Binding Studies with Living Cells. Genetic

Engineering & Biotechnology News, 2012. 32(4): p. 20-21.

115. Lee, J., et al., Sensitive and reproducible detection of cardiac troponin I in human

plasma using a surface acoustic wave immunosensor. Sensors and Actuators B-

Chemical, 2013. 178: p. 19-25.

116. Fertier, L., et al., Love wave immunosensor for antibody recognition using an

innovative semicarbazide surface functionalization. Sensors and Actuators B-

Chemical, 2009. 140(2): p. 616-622.

117. Lee, H.J., et al., Surface acoustic wave immunosensor for real-time detection of

hepatitis B surface antibodies in whole blood samples. Biosens Bioelectron, 2009.

24(10): p. 3120-3125.

118. Treitz, G., et al., Combination of a SAW-biosensor with MALDI mass spectrometric

analysis. Biosens Bioelectron, 2008. 23(10): p. 1496-502.

119. Moll, N., et al., Multipurpose Love acoustic wave immunosensor for bacteria, virus or

proteins detection. Irbm, 2008. 29(2-3): p. 155-161.

120. Maftei, M., et al., Interaction structure of the complex between neuroprotective factor

humanin and Alzheimer's beta-amyloid peptide revealed by affinity mass spectrometry

and molecular modeling. Journal of Peptide Science, 2012. 18(6): p. 373-382.

121. Dr Gusanu, M., B.A. Petre, and M. Przybylski, Epitope motif of an anti-nitrotyrosine

antibody specific for tyrosine-nitrated peptides revealed by a combination of affinity

approaches and mass spectrometry. Journal of Peptide Science, 2011. 17(3): p. 184-

91.

122. Dragusanu, M., et al., On-line bioaffinity-electrospray mass spectrometry for

simultaneous detection, identification, and quantification of protein-ligand

interactions. J Am Soc Mass Spectrom, 2010. 21(10): p. 1643-8.

60

123. Schlesinger, M., et al., Binding between heparin and the integrin VLA-4. Thrombosis

and Haemostasis, 2009. 102(5): p. 816-822.

124. Gronewold, T.M.A., et al., Kinetic Binding Analysis of Aptamers Targeting HIV-1

Proteins by a Combination of a Microbalance Array and Mass Spectrometry (MAMS).

Journal of Proteome Research, 2009. 8(7): p. 3568-3577.

125. Gronewold, T.M.A., et al., Selection process generating peptide aptamers and

analysis of their binding to the TiO2 surface of a surface acoustic wave sensor. Acta

Biomater, 2009. 5(2): p. 794-800.

126. Cui, J.X., et al., Analysis of Responsive Polymer Films Using Surface Acoustic Waves.

Langmuir, 2013. 29(22): p. 6582-6587.

127. Saitakis, M., A. Tsortos, and E. Gizeli, Probing the interaction of a membrane

receptor with a surface-attached ligand using whole cells on acoustic biosensors.

Biosensors & Bioelectronics, 2010. 25(7): p. 1688-1693.

128. Raimbault, V., et al., Molecular weight influence study of aqueous poly(ethylene

glycol) solutions with a microfluidic Love wave sensor. Sensors and Actuators B-

Chemical, 2010. 144(1): p. 318-322.

129. Matatagui, D., et al., Detection of bacteriophages in dynamic mode using a Love-wave

immunosensor with microfluidics technology. Sensors and Actuators B-Chemical,

2013. 185: p. 218-224.

130. Hammer, M.U., et al., Lipopolysaccharide interaction is decisive for the activity of the

antimicrobial peptide NK-2 against Escherichia coli and Proteus mirabilis. Biochem

J, 2010. 427(3): p. 477-88.

131. Dinh, D.H., et al., Novel optimized biofunctional surfaces for Love mode surface

acoustic wave based immunosensors. Sensors and Actuators B-Chemical, 2010.

146(1): p. 289-296.

132. Al-Kaddah, S., et al., Analysis of membrane interactions of antibiotic peptides using

ITC and biosensor measurements. Biophys Chem, 2010. 152(1-3): p. 145-52.

133. Andra, J., et al., Surface acoustic wave biosensor as a tool to study the interaction of

antimicrobial peptides with phospholipid and lipopolysaccharide model membranes.

Langmuir, 2008. 24(16): p. 9148-53.

134. Gammoudi, I., et al., Love-wave bacteria-based sensor for the detection of heavy

metal toxicity in liquid medium. Biosensors & Bioelectronics, 2010. 26(4): p. 1723-

1726.

61

135. Papadakis, G., A. Tsortos, and E. Gizeli, Triple-helix DNA structural studies using a

Love wave acoustic biosensor. Biosensors & Bioelectronics, 2009. 25(4): p. 702-707.

62

8 LIST OF ATTACHMENTS

ATTACHMENT 1: Detailed list of SAW applications in literature and the reported

dissociation constant (6 pages)

Keyw

ords Sum

mary

Biosensor

(frequency in MH

z) K

D (nM)

SD on K

D (nM

) Purpose of K

D determ

ination R

ef.

Medicine/

Diagnostics

Interaction analysis between integrin V

LA-4

and non-anticoagulant heparin derivatives

LG: V

LA-4 [m

embrane vesicles], P-and L-

selectin [Fc-chimera]

AN

: Natalizum

ab [mA

b], heparin and (size fractionated) derivatives [glycosam

inoglycan]

S-Sens K5

(n.r.)

35.1 (V

LA-4/natalizum

ab) 2.22 x 10

3

(VLA

-4/enoxaparin) 4.61 x 10

3 (V

LA-4/tinzaparin)

2.71 x 103

(VLA

-4/RO

-H)

5.89 x 103

(VLA

-4/6-O D

S H)

11.6 x 103

(VLA

-4/2-O D

S H)

5.76 x 103

(P-selectin/tinzaparin) 6.16 x 10

3 (P-selectin/6-O

DS H

) 4.65 x 10

3 (P-selectin/ 6 sach. unit)

2.17 x 103

(P-selectin/8 sach. unit) 3.88

(P-selectin/12 sach. unit) 3.61 x 10

3 (L-selectin/tinzaparin)

3.11 x 103

(L-selectin/6-O D

S H)

4.29 x 103

(L-selectin/6 sach. unit) 7.16 x 10

3 (L-selectin/8 sach. unit)

0.556 x 103

(L-selectin/12 sach. unit)

8.4 (n=1)

0.76 x 103

(n=1) 2.68 x 10

3

(n=1) 2.63 x 10

3 (n=1)

4.12 x103

(n=1) 3.6 x 10

3

(n=1) 0.03 x 10

3 (n=1)

1.35 x 103

(n=1) 2.88 x 10

3 (n=1)

0.26 x 103

(n=1) 2.75 x 10

3 (n=1)

0.70 x 103

(n=1) 1.36 x 10

3 (n=1)

2.10 x 103

(n=1) 5.54 x 10

3 (n=1)

0.277 x 103

(n=1)

• K

D was determ

ined to com

pare binding affinity of different analytes

• Ligand screening

• Q

ualitative

[111]

(n.r.): not reported

Keyw

ords Sum

mary

Biosensor

(frequency in MH

z) K

D (nM)

SD on K

D (nM

) Purpose of K

D determ

ination R

ef.

Medicine/

Diagnostics

Technique to stabilize biological model

mem

branes for SAW

application LG

: Biotinylated m

odel mem

brane (POPC

) [lipids], V

LA-4 expressed into m

odel m

embrane (D

PPC-D

GA

/NTA

) [protein-lipids] A

N: V

CA

M-1 [protein], avidin [protein]

SAM

5 Blue

(n.r.)

9.02 x 10-2

(biotin/avidin) 2.48

(VLA

-4/VC

AM

-1)

n.r. (n=1) 0.41 (n=1)

• K

D used to com

pare with

online ligand im

mobilization

• D

etection of biological interaction

• Q

ualitative

[112]

Investigation to the mode of binding

LG: R

NA

aptamer [R

NA

/DN

A]

AN

: thrombin [protein]

S-Sens K5

(n.r.) 181

20 (n=1)

• K

D used to compare

with SPR

and filter-binding studies

• Q

ualitative

[113]

Binding betw

een Affim

ed TandAb antibody and

target tumor cells

LG: A

b [protein] A

N: target tum

or cells [cells]

SamX

(n.r.)

2 n.r.

• K

D compared w

ith flow

cytometric

analyis •

Ligand screening •

Qualitative

[114]

Detection and determ

ination of cardiac troponin I in hum

an plasma

(sandwich im

munoassay)

LG: capture A

b cTnI [protein] A

N: A

b /cardiac troponin I [protein]

In-house assembled

(200) n.r.

n.r. -

[115]

Evaluation of new chip preparations w

ith sem

icarbazide for Ag/A

b interactions LG

: HA

, FLAG

Ag [protein]

AN

: anti-HA

/FLAG

Ab [protein]

In-house assembled

(116) n.r.

n.r. -

[116]

Detection of hepatitis B

surface antibodies in w

hole blood samples

LG: hepatitis B

Ag [protein]

AN

: hepatitis B A

b [crude samples]

In-house assembled

(200) n.r.

n.r. -

[117]

SAW

-MS, sensor chips w

ith bound complex

were digested and further used for M

ALD

I/MS

peptide mass fingerprinting

LG: throm

bin (via RN

A-aptam

ers) [protein] A

N: antithrom

bin III [protein]

S-Sense K5

(n.r.) n.r.

n.r. -

[118]

(n.r.): not reported

Keyw

ords Sum

mary

Biosensor

(frequency in MH

z) K

D (nM)

SD on K

D (nM

) Purpose of K

D determ

ination R

ef.

Medicine/

Diagnostics

Developm

ent of SAW

method to detect

different pathogens LG

: specific Ab [protein]

AN

: IgG, E. coli, M

13 bacteriophage [protein, bacteria, virus]

In-house assembled

(118) n.r.

n.r. -

[119]

Specificity and affinity analysis between

humanin peptide (24aa) and β-am

yloid (Aβ)

peptides LG

: Aβ [peptide]

AN

: biotinylated humanin [peptide]

S-Sens K5

(n.r.)

0.61 x 103

(Aβ /pep4)

0.53 x 103

(Aβ /pep6)

0.26 x 103

(n=1) n.r.

(n=1)

• K

D determined to

compare affinity of

different analytes •

Elucidation (target) m

olecular pathway

• Q

uantitative, but no further (statistical) analysis

[120]

Binding betw

een tyrosine-nitrated peptides of prostacyclin synthase and anti-3-nitrotyrosine

antibody LG

: peptide 5 [peptide]; AN

: 3-NT-A

b [protein]

S-Sens K5

(n.r.) 63

20 (n=1)

• K

D used for characterisation of affinity

• Q

uantitative, but no further (statistical) analysis

[121]

Online SA

W-ESI-M

S for simultaneous

detection, ID and quantification of protein-

ligand interactions LG

: Aβ (1-16) [m

AB

], Antiα-Syn (C

-20)-R

[polyclonal Ab], Substance P [peptides]

AN

: nitrotyrosine peptide-1 [peptides], hα-Synuclein [protein], C

almodulin [protein]

S-Sens K5

(n.r.)

106.9 (m

Ab/ Peptide 1)

43.6 (Substance P/ C

almodulin)

164.7 (pA

b/ hα-Synuclein)

14.1 (n=1) 15.8 (n=1) 17.5 (n=1)

• K

D determination for

binding affinity study •

Quantitative, but no

further (statistical) analysis

[122]

Interaction analysis between integrin V

LA-4

and heparin LG

: VLA

-4 [mem

brane vesicles] A

N: heparin [glycosam

inoglycan]

S-Sens K5

(n.r.)

10.1 (V

LA-4/V

CA

M-1)

4.61 x 103

(VLA

-4/Tinzaparin)

9 2.68 x 10

3

(n≥3)

• K

D used to confirm

affinity and to com

pare with other

KD

• Target screening

• Q

ualitative

[123]

(n.r.): not reported

K

eywords

Summ

ary B

iosensor (frequency in M

Hz)

KD (nM

) SD

on KD

(nM)

Purpose of KD

determination

Ref.

Medicine/

Diagnostics

Aptam

ers and HIV

proteins + protein identification via M

ALD

I-ToF MS.

Protein was digested on chip, peptides w

ere analysed via peptide m

apping. LG

: biotyinylated aptamers [peptides]

AN

: V3 loop of H

IV-1 envelope gp150 and

HIV

-1 reverse transcriptase [proteins]

S-Sens K5

(n.r.)

406 (S66A

-C6/H

IV-1 V

3) 791

(S68B-C

5/HIV

-1 V3)

555 (S69 A

-C15/ H

IV R

T-984-2)

n.r.

• K

D were com

pared w

ith KD from

titration w

ith nitrocellulose filter

• K

D determined for

kinetics studies. •

Target characterization

• Q

uantitative, but no further (statistical) analysis

[124]

Food analysis

Measurem

ent of protein concentration in com

plex matrices (e.g. m

eat juice) LG

: Ab [protein]; A

N: acute phase protein

(APP) [protein]

Sam5

(n.r.) 0.96

n.r.

• K

inectic evaluation •

Quantitative, no

further (statistical) analysis

[22]

Detection of okadaic acid w

ith Love wave

biosensor LG

: Ab [protein] AN

: okadaic acid [fatty acid]

In-house assembled

(117) n.r.

n.r. -

[25]

Biom

aterials B

inding of peptides to inorganic materials

LG: m

ineral TiO2 ; A

N: aptam

er peptides S-Sens K

5 (n.r.)

81 n.r.

• K

D used to study kinetics

• Q

uantitative, but no further (statistical) analysis

[125]

Polymer

science

Study of the properties of a responsive brush later

LG: 4,5-dim

ethoxy-2-nitrobenzyl methacrylate

[polymer] A

N: U

V and pH

change

In-house assembled

(150) n.r.

n.r. -

[126]

Cell biology

Interaction analysis of cell mem

brane receptors to im

mobilized ligands

LG: protein G

/anti-HLA

[protein] A

N: LG

2 cell suspension [cells]

In-house assembled

(110) n.r.

n.r. -

[127]

(n.r.): not reported

K

eywords

Summ

ary B

iosensor (frequency in M

Hz)

KD (nM

) SD

on KD (nM

) Purpose of K

D determ

ination R

ef.

Rheology

The influence of molecular w

eight on a LW

acoustic sensor; LW-SA

W as a viscom

eter LG

: Polydimethylsiloxane (PD

MS)-chip

AN

: PEG [polym

er]

In-house assembled

(117) n.r.

n.r. -

[128]

Microbiology

Effector protein NIeF of E. coli N

IeF binds to caspase-4, -8 and -9. LG

: Capase-9 [protein]

AN

: NIeF [protein]

Sam5 B

lue (n.r.)

38.7

13.18

• K

D used to proved affinity

• Elucidation (target) m

olecular pathw

ay •

Qualitative

[24]

Developm

ent of a SAW

method for the

detection of bacteriophages LG

: mAB anti-M

13 [protein] AN

: M-13

bacteriophage [virus]

In-house assembled

(163) n.r.

n.r. -

[129]

Binding of antim

icrobial peptides to model

mem

branes of E. coli and P. mirabilis

LG: lipid bilayer [lipids]; A

N: [peptides]

S-Sens K5

(n.r.) n.r.

n.r. -

[130]

Detection of E.coli w

ith optimized surface

LG: E. coli Ab [protein] A

N: E. coli

[bacteria]

In-house assembled

(118) n.r.

n.r. -

[131]

Analysis of m

embrane activities of

antimicrobial peptides

LG: lipid bilayer (D

OC

P) [lipids] AN

: galliderm

in and vancoycin [peptides]

S-Sens K5

(n.r.) n.r.

n.r. -

[132]

Binding of peptides to bilayers

LG: lipid bilayer [lipids] A

N: LL32

[peptides]

S-Sens K5

(n.r.) n.r.

n.r. -

[133]

Environm

ental science

Detection of heavy m

etals in liquid medium

w

ith love wave biosensor

LG: E. coli [bacteria]

AN

: Cadm

ium, M

ercury [metals]

In house assembled

(118) n.r.

n.r. -

[134]

(n.r.): not reported

K

eywords

Summ

ary B

iosensor (frequency in M

Hz)

KD (nM

) SD

on KD (nM

) Purpose of K

D determ

ination R

ef.

Genom

ics

DN

A hybridization analysis of point m

utation cancer related gene fragm

ents LG

: biotynilated DN

A fragm

ents [DN

A]

AN

: DN

A [D

NA

]

S-Sens K5

(148)

144 (w

t BR

CA

1/ wt

BR

CA

1) 123

(mut B

RC

A1/ m

ut B

RC

A1)

30 (w

t ptch/ wt ptch)

108 (m

ut ptch/ mut ptch)

52 (w

t p53/ wt p53)

103 (w

t p53/ mut p53)

139 (m

ut p53/ mut p53)

3.856 x 103

(mut p53/ w

t p53)

n.r.

• K

D is used for detection of m

utations (hit/no-hit)

• Q

ualitative

[23]

Evaluation of geometrical characteristics

of DN

A m

olecules resulting from the

formation of triple-helical D

NA

s LG

: biotinylated DN

A triplexes A

N: D

NA

In-house assembled

(155) n.r.

n.r. -

[135]

(n.r.): not reported