Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209...

10
Sensors and Actuators B 209 (2015) 505–514 Contents lists available at ScienceDirect Sensors and Actuators B: Chemical jo ur nal home page: www.elsevier.com/locate/snb Surface characterization and antifouling properties of nanostructured gold chips for imaging surface plasmon resonance biosensing Sweccha Joshi a,b , Paola Pellacani c , Teris A. van Beek a , Han Zuilhof a , Michel W.F. Nielen a,d,a Laboratory of Organic Chemistry, Wageningen University, Dreijenplein 8, 6703 HB Wageningen, The Netherlands b TI-COAST, Science Park 904, 1098 XH Amsterdam, The Netherlands c Plasmore S.R.L, Via Deledda 4, 21020 Ranco (VA), Italy d RIKILT Wageningen UR, P.O. Box 230, 6700 AE Wageningen, The Netherlands a r t i c l e i n f o Article history: Received 2 September 2014 Received in revised form 6 November 2014 Accepted 15 November 2014 Available online 4 December 2014 Keywords: Nanoplasmonics Imaging SPR Surface characterization Zwitterionic polymers Poly(ethylene glycol) Miniaturization a b s t r a c t Surface Plasmon Resonance (SPR) optical sensing is a label-free technique for real-time monitoring of biomolecular interactions. Recently, a portable imaging SPR (iSPR) prototype instrument, featuring a nanostructured gold chip, has been developed. In the present work, we investigated the crucial first steps, prior to eventual use of the nanostructured iSPR chip, i.e., its surface modification, in-depth surface characterization and the antifouling performance. Results were compared with conventional flat (i)SPR gold chips having the same surface chemistries, viz. different types of polyethylene glycol and zwitteri- onic polymers. Characterization of the (i)SPR chips before and after surface modification was performed using atomic force microscopy (AFM), scanning electron microscopy (SEM), water contact angle (WCA), X-ray photoelectron spectroscopy (XPS) and direct analysis in real time high resolution mass spectrom- etry (DART-HRMS). The antifouling properties were then studied using the nanostructured chip in the portable iSPR instrument and the flat gold chip in conventional SPR setup. The zwitterionic polymer sur- face chemistries showed the best antifouling properties. Comparison of the nanostructured iSPR chips with conventional flat (i)SPR gold chips showed that the latter perform slightly better in terms of surface modification as well as antifouling properties. The portable iSPR instrument is almost as sensitive as conventional iSPR (IBIS) and nine times less sensitive than conventional SPR (Biacore 3000). The nano- structured iSPR chip, along with the portable instrument, offers the advantage of about ten-fold reduction in instrument size, weight and costs compared to conventional (i)SPR instruments using flat gold, thus making it highly interesting for future biosensing applications. © 2014 Elsevier B.V. All rights reserved. 1. Introduction Surface Plasmon Resonance (SPR) based biosensors have emerged as fast, sensitive, and label-free techniques for real- time monitoring of biomolecular interactions [1,2]. The desire to measure multiple biointeractions in parallel has triggered the development of a new platform known as imaging SPR (iSPR) [2–4]. In iSPR, the reflected light is collected by a charge-coupled device (CCD) camera, which allows real-time visualization of the change in reflectivity at multiple spots on the sensor surface [5]. Conventional iSPR instruments, although quite successful in biosensing applications [6–8], are rather heavy and costly, and Corresponding author at: Laboratory of Organic Chemistry, Wageningen Uni- versity, Dreijenplein 8, 6703 HB Wageningen, The Netherlands. E-mail address: [email protected] (M.W.F. Nielen). should be considered as high-end laboratory-based biosensing equipment. Considering the demand for bringing the lab to the sample, a portable iSPR prototype has been developed recently [9,10], which offers the potential for in-field and at-line biosens- ing applications. Unlike conventional SPR (Fig. 1), the miniaturized iSPR instrument uses nanostructured gold instead of flat gold as a sensor chip surface. The nanostructured surface is made up of a periodic alternation of poly(methyl methacrylate) and gold [11], as shown in Figs. 1a and 2. This forms a metal-dielectric pattern that acts as a metallic nanograting [12,13], thus elim- inating the use of expensive and delicate prism-based optics and contributing to miniaturization, portability and low costs of the instrument. Additionally, the periodicity of a nanostructu- red gold surface is known to influence the SPR signal [14]. Due to advantages over flat surfaces, nanostructured gold has also found application in surface-enhanced Raman spectroscopy (SERS) [15–17]. http://dx.doi.org/10.1016/j.snb.2014.11.133 0925-4005/© 2014 Elsevier B.V. All rights reserved.

Transcript of Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209...

Page 1: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

Sg

SMa

b

c

d

a

ARRAA

KNISZPM

1

ettd[dc[b

v

h0

Sensors and Actuators B 209 (2015) 505–514

Contents lists available at ScienceDirect

Sensors and Actuators B: Chemical

jo ur nal home page: www.elsev ier .com/ locate /snb

urface characterization and antifouling properties of nanostructuredold chips for imaging surface plasmon resonance biosensing

weccha Joshia,b, Paola Pellacanic, Teris A. van Beeka, Han Zuilhofa,ichel W.F. Nielena,d,∗

Laboratory of Organic Chemistry, Wageningen University, Dreijenplein 8, 6703 HB Wageningen, The NetherlandsTI-COAST, Science Park 904, 1098 XH Amsterdam, The NetherlandsPlasmore S.R.L, Via Deledda 4, 21020 Ranco (VA), ItalyRIKILT Wageningen UR, P.O. Box 230, 6700 AE Wageningen, The Netherlands

r t i c l e i n f o

rticle history:eceived 2 September 2014eceived in revised form 6 November 2014ccepted 15 November 2014vailable online 4 December 2014

eywords:anoplasmonics

maging SPRurface characterizationwitterionic polymersoly(ethylene glycol)iniaturization

a b s t r a c t

Surface Plasmon Resonance (SPR) optical sensing is a label-free technique for real-time monitoring ofbiomolecular interactions. Recently, a portable imaging SPR (iSPR) prototype instrument, featuring ananostructured gold chip, has been developed. In the present work, we investigated the crucial firststeps, prior to eventual use of the nanostructured iSPR chip, i.e., its surface modification, in-depth surfacecharacterization and the antifouling performance. Results were compared with conventional flat (i)SPRgold chips having the same surface chemistries, viz. different types of polyethylene glycol and zwitteri-onic polymers. Characterization of the (i)SPR chips before and after surface modification was performedusing atomic force microscopy (AFM), scanning electron microscopy (SEM), water contact angle (WCA),X-ray photoelectron spectroscopy (XPS) and direct analysis in real time high resolution mass spectrom-etry (DART-HRMS). The antifouling properties were then studied using the nanostructured chip in theportable iSPR instrument and the flat gold chip in conventional SPR setup. The zwitterionic polymer sur-face chemistries showed the best antifouling properties. Comparison of the nanostructured iSPR chipswith conventional flat (i)SPR gold chips showed that the latter perform slightly better in terms of surface

modification as well as antifouling properties. The portable iSPR instrument is almost as sensitive asconventional iSPR (IBIS) and nine times less sensitive than conventional SPR (Biacore 3000). The nano-structured iSPR chip, along with the portable instrument, offers the advantage of about ten-fold reductionin instrument size, weight and costs compared to conventional (i)SPR instruments using flat gold, thusmaking it highly interesting for future biosensing applications.

. Introduction

Surface Plasmon Resonance (SPR) based biosensors havemerged as fast, sensitive, and label-free techniques for real-ime monitoring of biomolecular interactions [1,2]. The desireo measure multiple biointeractions in parallel has triggered theevelopment of a new platform known as imaging SPR (iSPR)2–4]. In iSPR, the reflected light is collected by a charge-coupledevice (CCD) camera, which allows real-time visualization of the

hange in reflectivity at multiple spots on the sensor surface5]. Conventional iSPR instruments, although quite successful iniosensing applications [6–8], are rather heavy and costly, and

∗ Corresponding author at: Laboratory of Organic Chemistry, Wageningen Uni-ersity, Dreijenplein 8, 6703 HB Wageningen, The Netherlands.

E-mail address: [email protected] (M.W.F. Nielen).

ttp://dx.doi.org/10.1016/j.snb.2014.11.133925-4005/© 2014 Elsevier B.V. All rights reserved.

© 2014 Elsevier B.V. All rights reserved.

should be considered as high-end laboratory-based biosensingequipment. Considering the demand for bringing the lab to thesample, a portable iSPR prototype has been developed recently[9,10], which offers the potential for in-field and at-line biosens-ing applications. Unlike conventional SPR (Fig. 1), the miniaturizediSPR instrument uses nanostructured gold instead of flat gold asa sensor chip surface. The nanostructured surface is made upof a periodic alternation of poly(methyl methacrylate) and gold[11], as shown in Figs. 1a and 2. This forms a metal-dielectricpattern that acts as a metallic nanograting [12,13], thus elim-inating the use of expensive and delicate prism-based opticsand contributing to miniaturization, portability and low costs ofthe instrument. Additionally, the periodicity of a nanostructu-

red gold surface is known to influence the SPR signal [14]. Dueto advantages over flat surfaces, nanostructured gold has alsofound application in surface-enhanced Raman spectroscopy (SERS)[15–17].
Page 2: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

506 S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514

Fig. 1. Schematic representation of (a) prototype imaging SPR setup with nanostructured gold as sensor surface and (b) conventional SPR setup with flat gold as sensors

sbrossaAmbpAtgcaePep“mkttc[st[

enpsatigppc

urface, and the antifouling chemistries used.

A crucial step towards the use of any sensor chip for biosen-or applications, is prevention of non-specific interactions ofiomolecules (biofouling) leading to false positive signals. Thisequires the use of well-defined antifouling chemistries, that notnly help to overcome fouling issues, but also to obtain sufficienturface stability for regeneration and repeated use of the sensorurface. SPR optical sensing has been used extensively to studyntifouling properties of chemically modified flat gold [18–22].

widespread approach to reduce non-specific adsorption is toodify the surface using a coating of hydrophilic polymers. Car-

oxymethylated (CM) dextran coatings are the oldest and veryopular antifouling layers used in commercial SPR sensors [23].s an alternative to CM dextran, poly(ethylene glycol) (PEG)-

ethered alkanethiol chains, terminated with various functionalroups, have been used [20,24]. PEG is a hydrophilic, electri-ally neutral polyether known to be resistant to non-specificdsorption mainly due to steric hindrance and water barrierffects [25]. Careful tuning of the length and density of theEG-tethered chains is, however, required to achieve the desiredffect [26]. These parameters are dependent on the method ofolymer growth, thus it is interesting and relevant to comparegrafting to” [27] and “grafting from” [28] approaches of poly-er attachment. Although PEG has been used extensively, it is

nown to oxidize easily in the presence of oxygen and variousransition metal ions [24]. Recently, interest in finding alterna-ives for PEG is increasing, leading to the development of a newlass of antifouling surfaces composed of zwitterionic polymers22]. The antifouling properties of zwitterionic polymer surfacestem mainly from the strong electrostatic interaction betweenhe opposite charges present and its high hydration capacity27,29,30].

In-depth characterization of the sensor chip prior to antifoulingxperiments, is important to understand the surface properties. Theanostructured gold surface used in the present study, is an integralart of the design of the portable iSPR setup (Fig. 1a). The nano-tructured chip in combination with the iSPR instrument offers thedvantage of reduced instrument size, weight and costs comparedo conventional (i)SPR using flat gold and a prism. The surface mod-fication and subsequent characterization of these nanostructured

old chips has never been reported in the literature. Therefore,roperties of nanostructured iSPR chips were studied and com-ared with those obtained on flat gold chips modified with the samehemistries for future application transferability.

2. Materials and methods

2.1. Chemicals and substrates

16-Mercaptohexadecanoic acid (MHDA), pentafluorophenol(PFP) and N,N′-dicyclohexylcarbodiimide (DCC), poly(ethyleneglycol) 2-aminoethyl ether acetic acid (average MW 1000 and3500) (NH2-PEG-COOH), 2,2′-bipyridine (BiPy), anhydrous cop-per (I) chloride (CuCl), anhydrous copper (II) chloride (CuCl2),(2-(methacryloyloxy)ethyl)dimethyl-3-sulfopropyl)ammoniumhydroxide (SBMA monomer), poly(ethylene glycol) methacrylate(PEGMA monomer, MW ∼ 500), isopropanol, bovine serum albu-min (BSA) and PBS buffer tablets (1 tablet was dissolved in 200 mlof deionized water to make 10 mM PBS pH 7.4) were purchasedfrom Sigma Aldrich (Zwijndrecht, The Netherlands). Absoluteethanol was purchased from Fisher Scientific. All the solvents wereused as obtained, except for dichloromethane (Sigma Aldrich,>99.8%), which was further purified using a Pure Solv 400 solventpurification system (Innovative Technology, Amesbury, USA).11-Mercaptoundec-1-yl 2-bromo-2-methylpropionate (Izo-Br)was purchased from Prochimia (Sopot, Poland). HBSEP buffer(0.01 M HEPES pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% (v/v)Surfactant P20) and 1 M ethanolamine·HCl (pH 8.5) were pur-chased from GE Healthcare (Diegem, Belgium). Goat milk wasbought from a local organic farm. Pale lager (The Netherlands)and Belgian ale were used as beer 1 and beer 2, respectively.Biotin-PEG3-amine and streptavidin were purchased from ThermoScientific (The Netherlands) and Life Technologies Europe BV (TheNetherlands) respectively. Deionized water (18.3 M� cm resisti-vity) was obtained using a Merck (Amsterdam, The Netherlands)water purification system.

Flat gold substrates of Au sputtered on glass were purchasedfrom Xantec (Düsseldorf, Germany). The nanostructured gold chipswere produced using colloidal lithography and plasma-enhancedchemical vapor deposition (PE-CVD) [31]. Briefly, poly(methylmethacrylate) (PMMA) was deposited on the glass substrate byPE-CVD using methyl methacrylate as liquid precursor. Spin coat-ing was then used to cover the film with polystyrene (PS) beads(500 nm with a nominal size dispersion of 10%). Next, the sam-

ple was exposed to oxygen plasma etching to remove the PMMAfrom the areas not covered by the beads and to reduce the size ofthe PS beads to provide the required periodicity. A gold layer of100-200 nm was then deposited on the surface by physical vapor
Page 3: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

Actuat

di2a

2

2

rn1Mat1TiTawbe

2z

psTsfa(bCawrtttcwwasfswTbrtr

iaidovd((fl

S. Joshi et al. / Sensors and

eposition. Finally, the polystyrene beads were removed resultingn a surface with a gold film perforated by PMMA (Figs. 1a and). The combined control over deposition and etching parametersllowed fine tuning of film thickness and PMMA well diameters.

.2. Surface modification

.2.1. “Grafting to” growth of polyethylene glycolPrior to modification, the Au substrates (flat and nanostructu-

ed) were rinsed with ethanol and water followed by drying withitrogen. The chips were then immersed in a 1 mM solution of6-mercaptohexadecanoic acid (MHDA) in ethanol for 24 h. TheHDA-modified surfaces were removed and rinsed with ethanol

nd water, sonicated in ethanol, and dried with nitrogen. The acid-erminated substrates were activated by immersing overnight in a:1 mixture of 0.4 M PFP and 0.4 M DCC in dichloromethane [32].he PFP-terminated substrates were washed with DCM and thenmmersed in a 1 mg/ml solution of NH2–PEG–COOH in DCM for 24 h.he PEG-modified (PEG1000 and PEG3500) surfaces were removednd rinsed with DCM, sonicated in the same solvent, and driedith nitrogen. Any remaining PFP activated esters were deactivated

y immersion in ethanolamine for 30 min prior to the antifoulingxperiments.

.2.2. “Grafting from” growth of polyethylene glycol andwitterionic polymers

The Au substrates were coated with an atom transfer radicalolymerization (ATRP) initiator by immersing them in 2.5 �l/mlolution of Izo-Br in absolute ethanol at room temperature for 24 h.he cleaning procedures before and after modification were theame as in the case of MHDA. For the polymerization, a procedurerom literature was used [33]. SBMA monomer (4.90 g, 17.5 mmol)nd BiPy (0.14 g, 0.9 mmol) were dissolved in deionized water16 ml) and isopropanol (4 ml). The solution was then degassedy purging with argon for 30 min. CuCl (71.3 mg, 0.72 mmol) anduCl2 (9.7 mg, 0.072 mmol) were weighed in a separate flask inside

glove box (Argon 6.0, O2 < 0.1 ppm, and H2O < 0.1 ppm). The flaskas sealed with a rubber septum to ensure an oxygen-free envi-

onment. The SBMA monomer-BiPy solution was then transferredo the flask containing CuCl/CuCl2 using an argon-flushed double-ipped needle. The solution was stirred under argon for 30 mino completely dissolve the CuCl/CuCl2. Meanwhile, the initiator-oated gold substrate was placed in a separate flask and flushedith argon. The mixture containing monomer, BiPy and CuCl/CuCl2as transferred to the flask with the initiator-coated substrate,

gain using an argon-flushed double-tipped needle. Next, the sub-trate was left to polymerize under argon at room temperatureor 2 h. Finally, the SBMA-modified surface was rinsed with water,onicated in water and dried with nitrogen. The same procedureas used for PEGMA using PEGMA monomer (8.75 g, 17.5 mmol).

he PEGMA monomer was passed through a column of activated,asic aluminum oxide directly prior to the polymerization toemove inhibitors. In case of PEGMA, the substrates were addi-ionally rinsed with and sonicated in methanol to remove anyemaining monomer.

In the case of nanostructured gold, the chip surface was mod-fied only partially, in order to have an unmodified gold region as

reference on the same chip. The possibility of having a referencenside the same chip helps to correct for any fluctuations caused byifferences in illumination of the surface. Simple dipping of (a partf) the chip did not work, due to capillary forces acting on the sol-ent (mainly ethanol). Therefore, an oval PEEK flow-cell (internal

imension: 18 mm × 4 mm, volume: 4 �l) sealed by Viton O-ringssee Fig. S1) was used, providing both resistance to the solventsethanol and dichloromethane) as well as a good sealing. Using thisow-cell the nanostructured gold was modified with MHDA, PFP,

ors B 209 (2015) 505–514 507

PEG1000, PEG3500 and Izo-Br by flowing the respective solutionas mentioned above at a continuous flow rate of 0.1 �l/min over apart of the chip surface. After the partial modification of the surfacewith Izo-Br, subsequent reactions with SBMA and PEGMA had to beperformed under argon and the use of the flow-cell setup was notfeasible. Fortunately, in these cases, the solvent was mainly water,thus simple dipping was feasible at this stage to obtain a partiallymodified surface. The chips were placed in narrow glass tubes andthe solution was carefully transferred to fill the tube from the bot-tom until part of the chip was covered. It was important that thenanostructured chip was only partially submerged, as physisorp-tion of the polymer, although to a lesser extent (as measured byXPS) than the part with initiator, was observed also in the areaswithout the initiator.

2.3. Surface characterization

2.3.1. Atomic Force Microscopy (AFM)The AFM images were taken using a JSPM-5400 (Jeol, Japan)

Scanning Probe Microscope in AC-AFM (“tapping”) mode, andOMCL-AC240TS-R3 (Olympus, Japan) cantilevers.

2.3.2. Scanning Electron Microscopy (SEM)The SEM images were taken using a JAMP-9500F (Jeol, Japan)

Field Emission Auger Microscope. The nanostructured surface wasplaced in a JFC-1300 (Jeol, Japan) auto fine coater for 40 s to obtaina thin layer of gold that reduces charging effects during SEM mea-surements, and helps to get a good focusing.

2.3.3. Static water contact angle measurementsThe wettability of the surfaces was determined by measur-

ing the static water contact angle (WCA) using a DSA-100 (Krüss,Germany) goniometer. Drops (2–5 depending on the sample size)of 3 �l of MilliQ water were dispensed on Au surfaces with a micro-liter syringe with stainless steel needle (diameter = 0.51 mm), andthe CA was determined using a Tangent 2 fitting model.

2.3.4. X-ray Photoelectron Spectroscopy (XPS)XPS analyses of the surfaces were performed using a JPS-

9200 (Jeol, Japan) photoelectron spectrometer. The spectra wereobtained under UHV conditions using monochromatic Al K� X-rayradiation at 12 kV and 20 mA, using analyzer pass energy of 10 eV.The spectra were reprocessed using the CASA XPS peak fit program(version 2.3.16). The curve fitting was performed with a linear back-ground fitting. The thickness of the organic layers on the surfacewas calculated using the following formula:

t = lnIAuo

IAu× �Au × cos � [34]

where t = thickness (in nm) of the monolayer, IAuo = intensity of XPSsignal of Au4f7/2 at 83.9 eV (relative to C1s signal) in unmodifiedgold, IAu = intensity of XPS signal of Au4f7/2 (relative to C1s signal)in modified gold, �Au = effective attenuation length of Au4f elec-trons in the organic films (using a value of 3.858 nm as reported byPetrovykh et al. [35] for DNA films), � = the photoelectron emissiontakeoff angle relative to the surface normal (10◦).

2.3.5. Direct Analysis in Real Time High-Resolution MassSpectrometry (DART-HRMS)

The DART-orbitrap high-resolution mass spectrometer systemconsisted of a DART-SVP ion source (Ion-Sense, Saugus, USA) cou-

pled to an Exactive high resolution MS system (Thermo FisherScientific, San Jose, CA, USA). The MS was calibrated daily usingProteoMassTM LTQ/FT-Hybrid ESI (positive and negative mode)Cal Mix (Sigma Aldrich), which is applicable for the m/z range
Page 4: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

508 S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514

F , (b)

1mhbigDasToAmm

2

iaaaagIc(TiaHibbai(m

3.1. Surface modification and characterization

ig. 2. Surface characterization of unmodified nanostructured Au iSPR chip. (a) AFM

00–2000. XCalibur software (version 2.1) was used for instru-ent control, data acquisition and data processing. For the DART,

elium (flow of ∼3.5 l/min) was used as ionizing gas, with the gaseam temperature set at 400 ◦C. The (i)SPR chips (blank and mod-

fied) were attached to a glass slide using double-sided tape. Thelass slide was then fixed onto a motorized rail placed between theART source and MS inlet. The DART was pointed at an angle of 45◦

bove the chip surface. A detailed description of the technique ofurface measurements can be found in a previous publication [36].he neat solutions were measured by placing a drop of the solutionnto a glass slide using the same settings as for modified samples.ll the surfaces were measured in positive ion mode. The surfacesodified with PFP were additionally characterized in negative ionode.

.4. Antifouling experiments

BSA, beer and milk samples were used to test the antifoul-ng properties of different surface chemistries. BSA was chosen as

general model protein for non-specific adsorption, while beernd milk were chosen as model sample matrices for real-life SPRpplications. SPR measurements on flat gold were performed using

Biacore 3000 (GE Healthcare, Sweden), and on nanostructuredold using an imaging nanoplasmonics instrument (Plasmore Srl.,taly). Prior to the antifouling experiments (see Section 3.2), thehips were washed for 1 min with 5 mM NaOH followed by 3 mint = 0–180 s) with running buffer (HBSEP) to get a stable baseline.he non-specific binding was monitored by injecting BSA (1 mg/mln HBSEP), beer samples (four times diluted in HBSEP for Biacorend undiluted for nanostructured gold), and milk (10% diluted inBSEP) at a constant flow of 20 �l/min. After a 5 min (t = 180–480 s)

njection of the analyte, the channels were washed with runninguffer (HBSEP). The response obtained after 4 min (t = 720 s) ofuffer flow (20 �l/min), relative to the starting baseline, was used

s a measure of the amount of fouling. For comparing the foul-ng from different samples on the different modified gold surfacesflat and nanostructured), the relative response obtained was nor-

alized to the response obtained for bare gold upon addition of

SEM images, and (c) AFM profile of the surface along the horizontal arrow in (a).

1 mg/ml BSA to the respective gold surface. The sensorgrams andbar graphs were plotted using Origin 8.5 and Excel 2010. Sensor-grams from the portable iSPR instrument were smoothened usingthe Savitzky-Golay function in Origin 8.5 (10 points of window,polynomial order 2).

Sensitivity measurement: The sensitivity of the portable iSPRinstrument was compared to that of conventional SPR (Biacore3000) and imaging SPR (IBIS Technologies B.V., Hengelo, TheNetherlands). Calibration curves were constructed with serial dilu-tions of glycerol ranging from 10 to 0.01% for each instrument usingthe respective bare chips. The LODs were calculated as three timesthe standard deviation of the baseline (noise). To allow comparisonof the different instruments, each with their own units of response,the LODs were converted to percentage of glycerol using the fittingequation of the calibration curve.

Biotin-streptavidin binding: PEG3500 modified chips were acti-vated using a 1:1 mixture of 0.4 M EDC and 0.1 M NHS for 10 min.The chips were washed with water and then covered with a 1 mg/mlsolution of biotin–PEG–NH2 in PBS (pH 7.4) for 2 h at room tem-perature. The chips were then washed with PBS and subsequentlyblocked with ethanolamine for 10 min. The modifications withbiotin were confirmed by XPS. The chips were then mounted onthe corresponding instruments to monitor the binding of strepta-vidin (Figs. 6 and S16). A 10 �g/ml solution of streptavidin (dilutedin HBSEP) was introduced (t = 50 s) at a flow rate of 20 �l/min. Aftera 5 min (t = 50–350 s) injection, the chips were washed with run-ning buffer (HBSEP). The response obtained after 30 s (t = 380 s) ofbuffer flow (20 �l/min), relative to the starting baseline, was usedas a measure of the amount of binding.

3. Results and discussion

AFM and SEM were used to investigate the surface morphologyof the nanostructured iSPR chip. Both techniques clearly displaythe nanograting with periodic alternation of gold and poly(methyl

Page 5: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 509

Fig. 3. Surface characterization of modified nanostructured gold. XPS survey scan (left) and C1s narrow scan (right) of (a) PEGMA-modified, and (b) SBMA-modified nano-s m/z

w

mtwmi5h

tructured gold, and (c) DART-HRMS of PEGMA-modified nanostructured gold fromith 1 and 2 respectively.

ethacrylate) (see Fig. 2). Three regions can be seen as indicated byhe arrows in Fig. 2: flat gold, poly(methyl methacrylate) (PMMA)ells, and gold rings surrounding the PMMA. The poly(methyl

ethacrylate) regions with diameters of 200–250 nm and period-

city (distance between the centers of the two PMMA wells) of00–600 nm make up around 20% of the total surface area. Theeight of the gold rings is in the range of 10–20 nm. The surface

170 to 670. The two abundant ion series of oligoethylene oxide chains are marked

contains some defects and residual polystyrene beads as seen inFig. 2b. However, these defects and residual PS beads are minor,and most likely do not affect the overall modification, antifouling

and sensing properties of the chips in a significant fashion.

Since, the chemical modification of these nanostructured iSPRsubstrates has not been studied before, each step of the sur-face modification was carefully characterized using water contact

Page 6: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

510 S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514

Table 1Comparison of contact angle, dry layer thickness determined by XPS, and representative ions measured in DART-HRMS (measured in positive ion mode, unless otherwisementioned) with the corresponding m/z values for different modification on flat and nanostructured gold.a

Coating Contact Angle in◦ Dry thickness in nm Ions in DART m/z

Flat iSPR Flat iSPR

Bare 65 ± 1 71 ± 2 NA NA NA NAMHDA 40 ± 1 63 ± 4 2.0 ± 0.4 2.1 ± 0.9 [HOOC–(CH2)15–S]+ 287.2089,PFP 93 ± 2 88 ± 2 2.1 ± 0.2 2.3 ± 0.8 [C6F5OC( O)(CH2)15–S]+, [C6F5O]− 453.1873 (positive mode),

182.9860 (negative mode)

PEG1000 33 ± 2 58 ± 2 3.7 ± 0.2 2.0 ± 0.5 180.0866 + 44.0257p forp = 0–10

PEG3500 45 ± 3 63 ± 2 4.9 ± 0.8 2.2 ± 0.3

Izo-Br 82 ± 1 81 ± 3 1.8 ± 0.1 1.9 ± 0.5 352.1477, 354.1456

PEGMA 47 ± 2 49 ± 1 21.2 ± 1.0 13.7 ± 1.5 Series 1: C10H22O5N + q × C2H4OSeries 2: C12H22O5N + q × C2H4O

236.1487 + 44.0257q(Series 1) and260.1492 + 44.0257q(Series 2) for q = 0-9

SBMA 21 ± 3 24 ± 2 14.9 ± 1.8 11.1 ± 2.1

e erroa

aHmbrm4aiiendtsefcatBtItflamiasg

oftelflvC

a The values reported in the table are an average of three measurements and thpplicable.

ngle measurements, X-ray photoelectron spectroscopy and DART-RMS and the data were compared to those of flat SPR surfacesodified according to the same procedures. The contact angles of

are gold, MHDA, PEG1000 and PEG3500 modified nanostructu-ed gold (Table 1) were found to be significantly higher than thoseeasured on flat gold (Table 1) and reported for flat gold (65◦, 44◦,

2◦ and 40◦, respectively) [27]. The contact angle values for PEGre difficult to compare with literature values as small differencesn these multistep surface modifications may lead to differencesn contact angle values. Such variations, also reported in the lit-rature [37], may arise due to differences in cleaning procedure,ature of initial self-assembled monolayer and differences in poly-ispersity, purity of end groups and average molecular weight ofhe PEG depending on the supplier. The higher values for nano-tructured gold are probably due to the PMMA regions, which arexpected to have a high contact angle (80–90◦) [38]. With a sur-ace area of 20%, PMMA can significantly affect the contact angle inase of thin layers. Note that the PMMA surface may be converted,t least partially, into a carboxylic acid surface during the plasmareatment step in the manufacturing of the nanostructured chips.ut any remaining hydrophobic PMMA regions will contribute tohe overall contact angle. The contact angles values of PFP andzo-Br modified nanostructured gold are in close agreement withhe observed (Table 1) and reported values for similarly modifiedat gold surfaces (90◦ and 80◦, respectively) [39]. These contactngle values are already close to that of PMMA and thus will not beuch affected. The effect of PMMA regions becomes insignificant

n the case of successful modification with thicker polymers, suchs SBMA and PEGMA, where the contact angles of modified nano-tructured gold are in good agreement with those of modified flatold (Table 1) and previous studies (20◦ and 47◦, respectively) [37].

XPS was used to determine the elemental composition of therganic layer, and also the dry thickness thereof, as determinedrom the attenuation of the Au4f7/2 signal from the bare gold surfaceo the coated ones [35]. The relative atomic percentages of differentlements (Figs. S2–S4) and the layer thickness (Table 1) as calcu-

ated from an XPS survey scan for MHDA, PFP and Izo-Br for bothat and nanostructured gold, are in agreement with the expectedalues of 2.1 nm, 2.3 nm, and 1.8 nm, respectively. Typically, in the1s narrow scan of these monolayers the peak at 286.5 eV is slightly

rs are represented as the standard deviation of the three measurements. NA – not

broader and shifted by 1 eV for the nanostructured iSPR chip rela-tive to flat gold. This is probably due to the underlying PMMA layerthat can still be measured by XPS in case of thin layers (< 10 nm).For PEG1000 and PEG3500 modified nanostructured gold, the drythickness calculated from the XPS survey scan is lower than thaton flat gold (roughly 2 nm versus 4 nm, respectively). According tothe literature, the thickness of “grafted to” PEG is expected to beless than 5 nm [37,40]. Under the conditions of XPS, these layersare in a collapsed state and the effect is more pronounced in thecase of nanostructured gold. This is possibly due to the underlyingPMMA that obstructs the formation of a well packed monolayerwhich, in turn, translates to a lower “grafting to” efficiency in caseof the nanostructured gold. Nevertheless, the increased intensityof the carbon peak at 286.7 eV in the C1s spectrum, correspond-ing to carbon attached to oxygen (Figs. S5 and S6), does confirmthe surface attachment of polyethylene glycol units, however to aslightly lower extent compared to flat gold. The presence of PEGunits on the surface was also confirmed by the representative ionseries observed in DART (see further below). The peak at 285 eV inthe C1s spectrum would be expected to be lower than observed forboth flat and nanostructured gold. This, along with a slightly higherC content from the survey scan (C/O = 2.5:1) and the contact anglevalues, suggest an effect of PMMA on the thin layers formed. In caseof PEGMA (Figs. 3a and S7), a thick layer is formed on both nano-structured and flat gold as seen from the high C and O content in thesurvey scan and a significant peak at 286.7 eV (C1s narrow scan).The dry layer thickness of PEGMA and SBMA modified nanostruc-tured gold, 11 and 14 nm, respectively, are slightly lower than thatof correspondingly modified flat gold (15 and 21 nm, respectively,Table 1). In case of SBMA-modified surfaces (Figs. 3b and S8), therelative percentage of the different elements are in good agreementwith expected values for the monomers, and the ratios of the car-bon peaks (C1s narrow scan) at 285 eV and 286.8 eV are as expectedalmost equal. In line with reports on SBMA-modified silicon nitridesurfaces [41], this indicates the formation of significant (>10 nm)polymer films.

DART-HRMS is a powerful tool for surface analysis complemen-tary to XPS [42], and was applied here to both flat and nanostruc-tured chip surfaces. In all cases, the same ions were observed,although sometimes with different relative intensities. Ions of the

Page 7: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 511

F usingd btain

i(falmwdow(tttgPmo(gdofmofomf[Bs[tbssssbiuPb

ig. 4. SPR sensorgrams generated upon addition of beer 1 by (a) iSPR instrumentashed line indicates the time point where the relative response was measured to o

ntact molecule were observed for small molecules, such as MHDA[HOOC–(CH2)15–S]+), and PFP ([C6F5OC( O)(CH2)15–S]+) resultingrom the scission of the relatively weak Au-S bond (Table 1), ingreement with literature for self-assembled thiol-on-gold mono-ayers [43]. Furthermore, ions after loss of H2 from the intact

olecule were also observed in both cases. In the case of MHDA,e also analyzed the molecule on a glass surface by placing a 5 �lrop of 1 mg/ml methanolic solution and the most intense ionbserved was the ammonia adduct [HOOC–(CH2)15–SH + NH4]+,hich was completely absent on the MHDA-modified gold surface

Fig. S9). This clearly indicates the absence of unbound MHDA inhe modified Au substrates. The PFP-modified surface, when addi-ionally analyzed by DART-HRMS in negative ion mode, showedhe ion [C6F5O]− as a result of in-situ ester hydrolysis (Fig. S10),iving rise to the free alcohol [42]. In case of PEG1000 andEG3500-modified gold, we observed ammonia adducts of frag-ents formed by scission of the C–O bonds from the carboxyl end

f the polymer chain: [HOOC–CH2O(CH2CH2O)pCH2CHO + NH4]+

Table 1, Figs. S11 and S12). Fragments with up to 12 ethylenelycol units were observed. Higher mass fragments are probablyifficult to be thermally desorbed by DART and were thus notbserved. The ammoniated fragments could be easily distinguishedrom the unbound PEG (Fig. S11c), where the most abundant frag-

ents were those representing the protonated primary amine endf the polymers: [HO–CH2CH2(OCH2CH2)pOCH2CH2–NH3]+. Theseragments with a protonated primary amine end were not presentn the surfaces modified with PEG1000 and PEG3500. For Izo-Brodified gold (Fig. S13), the ammonia adduct of the aldehyde,

ormed as a result of oxidation of the molecule, was observed:(CH3)2Br–C–C( O)O(CH2)10–(C O)H + NH4]+ (Table 1). The Izo-r molecule analyzed on glass (5 �l drop of 1 mg/ml methanolicolution) showed only the ammonia adduct of the intact molecule(CH3)2Br–C–C( O)O(CH2)11–SH + NH4]+. In both surface and solu-ion measurements, the isotope pattern confirmed the presence ofromine. In the case of PEGMA-modified gold (Fig. 3c), several ioneries containing ethylene glycol units were seen. Series 1 with atarting composition of C10H22O5N can be explained by two pos-ible structures, one after loss of water from the ions formed bycission of the C–O bond and the other due to scission of the C–Oond from the monomer itself. In fact the same ion series is present

n solution (Fig. S14), thus making it difficult to distinguish betweennbound PEGMA monomer and polymerized PEGMA. However,EGMA monomer placed on the gold surface with initiator coulde completely removed after washing with water and methanol

bare nanostructured gold, and (b) Biacore 3000 using bare flat gold. The vertical the amount of fouling.

followed by sonication in methanol (Fig. S14c). The same proce-dure was unable to remove the bound polymerized PEGMA. Thisproves that the ion series observed for polymerized PEGMA is notdue to unbound monomer. Series 2 with a starting composition ofC12H22O5N can be due to dimers of PEGMA. Series 2 was presentin both PEGMA modified flat (Fig. S14a) and nanostructured gold(Fig. 3c), however the intensity was much lower in case of flatgold. Several other ion series, besides Series 1 and 2 depicted inFig. 3c, were present in case of PEG1000, PEG3500 and PEGMA,but could not be associated yet with a particular structure. Sur-faces modified with SBMA were also studied using DART, but theresults are not included in Table 1, as the ions observed could not berelated to a particular (sub)structure of SBMA. Probably, only ther-mal decomposition product ions are generated from SBMA uponDART ionization.

3.2. Antifouling experiments

The antifouling behavior of the surfaces was evaluated usingthe (i)SPR instruments and BSA, beer and milk as sample matrices(Fig. 4). In case of BSA and beer, as expected, the largest amountof biofouling was seen for bare gold (Fig. 5). Mercaptohexade-canoic acid is only slightly better, due to its amphiphilic nature.In the case of MHDA-modified nanostructured gold, the under-lying PMMA makes the surface more hydrophobic compared toflat Au, thus resulting in still significant fouling upon treatmentwith BSA. The PEG-modified surfaces, with internal hydrophilicityin addition to the terminal hydrophilicity, showed better antifoul-ing properties than MHDA. The surfaces with PEG1000, both forflat and nanostructured gold, have showed better antifouling prop-erties than MHDA but were not able to completely resist foulingby beer. The surfaces modified with PEG3500, SBMA and PEGMAshowed very good antifouling behavior, both towards BSA andbeer. The responses obtained for chips having these three surfacechemistries, for both flat and nanostructured gold, were simi-lar within the measurement error range. This demonstrates thetransferability of the chemistries to the nanostructured gold interms of antifouling behavior. The difference between PEG1000and PEG3500 highlights the importance of chain length for effec-tive antifouling surfaces. The best antifouling performance was

observed for SBMA. This zwitterionic layer is well-known to beresistant to fouling due to the strong electrostatic interactionbetween the charged groups and the water molecules, thus form-ing a strongly bound hydration layer compared to the one formed
Page 8: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

512 S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514

F sampn trumea errors

vodss(t(uctinf

4

tglcbtioa3psm(

poses. As expected due to the strong binding of biotin-streptavidin,the response still remained high during the dissociation phase. Thesignal to noise of the portable iSPR instrument (S/N = 0.85) was

ig. 5. Relative responses measured upon addition of BSA and two different beeranoplasmonics instrument), and (b) flat gold (measured in a conventional SPR insddition of 1 mg/ml BSA. The values are an average of three measurements and the

ia hydrogen bonding in case of PEG [18]. However, in the casef 10% milk, fouling of the nanostructured surface was high andid not improve very much; even SBMA was unable to resist non-pecific adsorption (Fig. S15). Milk can be considered a worst-caseample matrix consisting of fats (3.7%), proteins (3.4%), lactose4.8%) and minerals (0.7%) [44]. Hydrophobic regions in milk pro-eins (casein and whey proteins) and long alkyl chains of fatsfatty acids and triacylglycerols) could have a high affinity for thenderlying hydrophobic PMMA in case of nanostructured gold. Theasein proteins also form micelles consisting of thousands of pro-ein molecules that could contribute to further fouling. Furthernvestigations are needed to elucidate why even SBMA-modifiedanostructured gold in the portable setup is performing less well

or milk versus flat gold in a conventional SPR setup.

. Discussion

Comparison of contact angle, XPS and DART-HRMS data showshat surface modification of nanostructured gold works analo-ously to that of flat gold, although the surface coatings are slightlyess effective. This was expected, given that 20% of the surface isovered with PMMA. The conditions for surface modification maye further optimized depending on specific future applications. Inerms of instrument performance, the refractive index sensitiv-ty of the portable instrument (LOD = 0.09%) is comparable to thatf a commercial imaging SPR instrument (IBIS, LOD = 0.07%), butlmost nine times less sensitive than conventional SPR (Biacore000, LOD = 0.01%). Preliminary regeneration experiments were

erformed using sodium hydroxide to test the stability of the nano-tructured chip. Upon injecting the same beer sample to a PEG3500odified chip for another cycle, similar responses were measured

within the measurement error) as the previous cycle (Fig. S17).

les to unmodified and modified (a) nanostructured gold (measured with imagingnt). The values have been normalized to the response obtained for bare gold upon

are represented as error bars (standard deviation of the three measurements).

Additionally, a proof-of-principle experiment was performed usingbiotin-streptavidin as a model system. With both instruments,upon addition of streptavidin to a surface locally modified withbiotin, a response was observed only in regions modified withbiotin (Figs. 6 and S16). The detection of a relatively small proteinsuch as streptavidin shows that the nanostructured surfaces withappropriate surface chemistries are promising for biosensing pur-

Fig. 6. SPR sensorgram generated upon addition of streptavidin (10 �g/ml diluted inHBSEP) to a nanostructured gold chip modified with PEG3500 and measured by theportable iSPR instrument. Inset: the chip was locally modified with biotin in region2. The sensorgram shown is the response in region 2 after background subtractionof the response recorded in region 1.

Page 9: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

Actuat

aSo((b

5

cdTPpal(cwoacsrtb

S

flPuaf

A

ntAWpf

A

t

R

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

[

S. Joshi et al. / Sensors and

lmost nine times less than that of conventional SPR (Biacore 3000,/N = 7.9). However, the portable iSPR instrument offers severalther key advantages such as reduction in size (14 times), weight8–11 times), and cost (7–12 times) compared to the conventionali)SPR instruments mentioned and has potential for future on-siteiosensing applications.

. Conclusions

Nanostructured chips were modified with different antifoulinghemistries (PEG and zwitterionic polymers) and characterized inetail using AFM, SEM, water contact angle, XPS and DART-HRMS.he antifouling properties of six different surfaces (bare, MHDA,EG1000, PEG3500, SBMA and PEGMA) were studied using theortable iSPR instrument. Chips modified with PEG3500, SBMAnd PEGMA showed the best antifouling properties for the estab-ished fouling standard (BSA) as well as a real-life application matrixbeer). Properties (characterization and antifouling) of the iSPRhips and performance of the iSPR instrument were comparedith flat gold and conventional SPR (Biacore). Minor differences

bserved in surface properties without influence on the over-ll antifouling performance demonstrated transferability of thehemistries. Although the iSPR instrument was nine times lessensitive than the conventional SPR, an approximately ten-foldeduction in size, weight and cost opens up significant possibili-ies for future at-field applications, such as detection of toxins ineer and barley, allergens in food [7].

upporting information

The XPS spectra, DART mass spectra of nanostructured andat gold modified with MHDA, PFP, PEG1000, PEG3500, SBMA,EGMA and SPR sensorgrams generated by the iSPR instrumentpon addition of beer 1 and 10% milk and streptavidin to barend/or differently modified nanostructured and/or flat gold can beound in the supporting information.

cknowledgements

This research received funding from the Netherlands Orga-isation for Scientific Research (NWO) in the framework ofhe Technology Area COAST (project nr 053.21.107) with VUmsterdam, RIKILT, Heineken, Synthon, Technex, EuroProxima,aterproef as partners and Plasmore and Bionavis as associated

artners. We would like to thank Dick J. van Iperen (VU Amsterdam)or constructing the flow-cell (Fig. S1).

ppendix A. Supplementary data

Supplementary data associated with this article can be found, inhe online version, at http://dx.doi.org/10.1016/j.snb.2014.11.133.

eferences

[1] J. Homola, Surface plasmon resonance sensors for detection of chemical andbiological species, Chem. Rev. 108 (2008) 462–493.

[2] R.B.M. Schasfoort, A.J. Tudos, Handbook of Surface Plasmon Resonance, TheRoyal Society of Chemistry, Cambridge, 2008.

[3] B. Rothenhaüsler, W. Knoll, Surface–plasmon microscopy, Nature 332 (1988)615–617.

[4] E. Yeatman, E.A. Ash, Surface plasmon microscopy, Electron. Lett. 23 (1987)1091–1092.

[5] S. Scarano, M. Mascini, A.P.F. Turner, M. Minunni, Surface plasmon resonanceimaging for affinity-based biosensors, Biosens. Bioelectron. 25 (2010) 957–966.

[6] D. Dorokhin, W. Haasnoot, M.C.R. Franssen, H. Zuilhof, M.W.F. Nielen, Imagingsurface plasmon resonance for multiplex microassay sensing of mycotoxins,Anal. Bioanal. Chem. 400 (2011) 3005–3011.

[

ors B 209 (2015) 505–514 513

[7] S.R. Raz, H. Liu, W. Norde, M.G.E.G. Bremer, Food allergens profiling with animaging surface plasmon resonance-based biosensor, Anal. Chem. 82 (2010)8485–8491.

[8] J.B. Beusink, A.M.C. Lokate, G.A.J. Besselink, G.J.M. Pruijn, R.B.M. Schasfoort,Angle-scanning SPR imaging for detection of biomolecular interactions onmicroarrays, Biosens. Bioelectron. 23 (2008) 839–844.

[9] A. Valsesia, P. Colpo, F. Rossi, F. Marabelli, A surface plasmon resonance sensingmethod and sensing system, European Patent, EP 2264438A1 (2010).

10] B. Bottazzi, L. Fornasari, A. Frangolho, S. Giudicatti, A. Mantovani, F. Marabelli,G. Marchesini, P. Pellacani, R. Therisod, A. Valsesia, Multiplexed label-free optical biosensor for medical diagnostics, J. Biomed. Opt. 19 (2014)0170061–01700610.

11] A. Valsesia, F. Marabelli, S. Giudicatti, G.R. Marchesini, F. Rossi, P. Colpo, SPRSensor Device with Nanostructure, World Patent, WO 2013/007448A1 (2013).

12] J. Breault-Turcot, J.-F. Masson, Nanostructured substrates for portableand miniature SPR biosensors, Anal. Bioanal. Chem. 403 (2012)1477–1484.

13] M. Najiminaini, E. Ertorer, B. Kaminska, S. Mittler, J.J.L. Carson, Surface plas-mon resonance sensing properties of a 3D nanostructure consisting of alignednanohole and nanocone arrays, Analyst 139 (2014) 1876–1882.

14] C.J. Alleyne, A.G. Kirk, R.C. McPhedran, N.-A.P. Nicorovici, D. Maystre, EnhancedSPR sensitivity using periodic metallic structures, Opt. Express 15 (2007)8163–8169.

15] M. Fan, G.F.S. Andrade, A.G. Brolo, A review on the fabrication of substratesfor surface enhanced Raman spectroscopy and their applications in analyticalchemistry, Anal. Chim. Acta 693 (2011) 7–25.

16] O.R. Bolduc, J.-F. Masson, Advances in surface plasmon resonance sensing withnanoparticles and thin films: nanomaterials, surface chemistry, and hybridplasmonic techniques, Anal. Chem. 83 (2011) 8057–8062.

17] J.N. Anker, W.P. Hall, O. Lyandres, N.C. Shah, J. Zhao, R.P. Van Duyne, Biosensingwith plasmonic nanosensors, Nat. Mater. 7 (2008) 442–453.

18] J. Ladd, Z. Zhang, S. Chen, J.C. Hower, S. Jiang, Zwitterionic polymers exhibit-ing high resistance to nonspecific protein adsorption from human serum andplasma, Biomacromolecules 9 (2008) 1357–1361.

19] Z. Zhang, S. Chen, Y. Chang, S. Jiang, Surface grafted sulfobetaine polymersvia atom transfer radical polymerization as superlow fouling coatings, J. Phys.Chem. B 110 (2006) 10799–10804.

20] K. Uchida, H. Otsuka, M. Kaneko, K. Kataoka, Y. Nagasaki, A reactivepoly(ethylene glycol) layer to achieve specific surface plasmon resonancesensing with a high S/N ratio: the substantial role of a short underbrushedPEG layer in minimizing nonspecific adsorption, Anal. Chem. 77 (2005)1075–1080.

21] Y. Chang, S.-C. Liao, A. Higuchi, R.-C. Ruaan, C.-W. Chu, W.-Y. Chen, A highlystable nonbiofouling surface with well-packed grafted zwitterionic polysulfo-betaine for plasma protein repulsion, Langmuir 24 (2008) 5453–5458.

22] L. Mi, S. Jiang, Integrated antimicrobial and nonfouling zwitterionic polymers,Angew. Chem. Int. Ed. 53 (2014) 1746–1754.

23] S. Löfås, B. Johnsson, A novel hydrogel matrix on gold surfaces in surface plas-mon resonance sensors for fast and efficient covalent immobilization of ligands,J. Chem. Soc. Chem. Commun. (1990) 1526–1528.

24] D. Leckband, S. Sheth, A. Halperin, Grafted poly(ethylene oxide) brushes asnonfouling surface coatings, J. Biomater. Sci. Polym. Ed. 10 (1999) 1125–1147.

25] S. Chen, L. Li, C. Zhao, J. Zheng, Surface hydration: principles and appli-cations toward low-fouling/nonfouling biomaterials, Polymer 51 (2010)5283–5293.

26] L.D. Unsworth, H. Sheardown, J.L. Brash, Protein-resistant poly(ethylene oxide)-grafted surfaces: chain density-dependent multiple mechanisms of action,Langmuir 24 (2008) 1924–1929.

27] C. Rodriguez-Emmenegger, E. Brynda, T. Riedel, Z. Sedlakova, M. Houska, A.B.Alles, Interaction of blood plasma with antifouling surfaces, Langmuir 25 (2009)6328–6333.

28] B.S. Lee, Y.S. Chi, K.-B. Lee, Y.-G. Kim, I.S. Choi, Functionalization ofpoly(oligo(ethylene glycol) methacrylate) films on gold and Si/SiO2 for immo-bilization of proteins and cells: SPR and QCM studies, Biomacromolecules 8(2007) 3922–3929.

29] J.B. Schlenoff, Zwitteration: coating surfaces with zwitterionic functionality toreduce nonspecific adsorption, Langmuir 30 (2014) 9625–9636.

30] Z.G. Estephan, P.S. Schlenoff, J.B. Schlenoff, Zwitteration as an alternative toPEGylation, Langmuir 27 (2011) 6794–6800.

31] S. Giudicatti, A. Valsesia, F. Marabelli, P. Colpo, F. Rossi, Plasmonic resonancesin nanostructured gold/polymer surfaces by colloidal lithography, Phys. StatusSolidi A 207 (2010) 935–942.

32] J. Lahiri, E. Ostuni, G.M. Whitesides, Patterning ligands on reactive SAMs bymicrocontact printing, Langmuir 15 (1999) 2055–2060.

33] A.T. Nguyen, J. Baggerman, J.M.J. Paulusse, H. Zuilhof, C.J.M. van Rijn, Bioconju-gation of protein-repellent zwitterionic polymer brushes grafted from siliconnitride, Langmuir 28 (2012) 604–610.

34] S. Hofmann, Quantitative analysis (data evaluation), in: Auger- and X-Ray Photoelectron Spectroscopy in Materials Science, Springer-Verlag,Berlin/Heidelberg, 2013, pp. 137.

35] D.Y. Petrovykh, H. Kimura-Suda, M.J. Tarlov, L.J. Whitman, Quantitative char-

acterization of DNA films by X-ray photoelectron spectroscopy, Langmuir 20(2004) 429–440.

36] R.K. Manova, F.W. Claassen, M.W.F. Nielen, H. Zuilhof, T.A. van Beek, Ambientmass spectrometry of covalently bound organic monolayers, Chem. Commun.49 (2013) 922–924.

Page 10: Sensors and Actuators B: Chemical - TI COAST Joshi...S. Joshi et al. / Sensors and Actuators B 209 (2015) 505–514 507 deposition. Finally, the polystyrene beads were removed resulting

5 Actuat

[

[

[

[

[

[

[

[

B

Sf

and holds a special Chair on analytical chemistry at Wageningen University. He

14 S. Joshi et al. / Sensors and

37] T. Riedel, Z. Riedelová-Reicheltová, P. Májek, C. Rodriguez-Emmenegger, M.Houska, J.E. Dyr, E. Brynda, Complete identification of proteins responsible forhuman blood plasma fouling on poly(ethylene glycol)-based surfaces, Lang-muir 29 (2013) 3388–3397.

38] S. Hosseini, F. Ibrahim, I. Djordjevic, L.H. Koole, Recent advances in surface func-tionalization techniques on polymethacrylate materials for optical biosensorapplications, Analyst 139 (2014) 2933–2943.

39] D.M. Jones, A.A. Brown, W.T.S. Huck, Surface-initiated polymerizations in aque-ous media: effect of initiator density, Langmuir 18 (2002) 1265–1269.

40] H.B. Lu, C.T. Campbell, D.G. Castner, Attachment of functionalized poly(ethyleneglycol) films to gold surfaces, Langmuir 16 (2000) 1711–1718.

41] A.T. Nguyen, J. Baggerman, J.M.J. Paulusse, C.J.M. van Rijn, H. Zuilhof, Stableprotein-repellent zwitterionic polymer brushes grafted from silicon nitride,Langmuir 27 (2011) 2587–2594.

42] R.K. Manova, S. Joshi, A. Debrassi, N.S. Bhairamadgi, E. Roeven, J. Gagnon, M.N.Tahir, F.W. Claassen, L.M.W. Scheres, T. Wennekes, K. Schroën, T.A. van Beek, H.Zuilhof, M.W.F. Nielen, Ambient surface analysis of organic monolayers usingdirect analysis in real time orbitrap mass spectrometry, Anal. Chem. 86 (2014)2403–2411.

43] K. Kpegba, T. Spadaro, R.B. Cody, N. Nesnas, J.A. Olson, Analysis of self-assembledmonolayers on gold surfaces using direct analysis in real time mass spectrom-etry, Anal. Chem. 79 (2007) 5479–5483.

44] N.P. Wong, R. Jenness, M. Keeney, E.H. Marth, Fundamentals of Dairy Chemistry,Springer US, New York, 1988.

iographies

weccha Joshi is a PhD candidate at Wageningen University, The Netherlands,ocused on the development of a miniaturized imaging SPR system towards

ors B 209 (2015) 505–514

biosensing applications. She received her MSc degree in chemistry from JacobsUniversity, Germany, in 2012. Her research interests include biosensors, surfacemodification and characterization.

Paola Pellacini is currently enrolled in a PhD school at the Universidad Autonomade Madrid, Spain and she is involved in production and surface characterizationof large area micro and nanostructured substrates. She received her MSc degree inbiomedical engineering in 2008 at Politecnico di Milano, Italy. Her research interestsinclude nanotechnology and material science. In 2010 she joined Plasmore Srl, Italy,to work on the fabrication and development of nanostructured plasmonic sensors.

Teris van Beek is Associate Professor in natural products chemistry at the Labora-tory of Organic Chemistry of Wageningen University, Wageningen, The Netherlands.As such he has worked on the isolation, purification, identification and analysisof bioactive or economically relevant metabolites from plants, microbes, ani-mals or man, such as ginkgolides, antioxidants, pheromones, natural dyes andproteins.

Han Zuilhof is the Chair of organic chemistry at Wageningen University. His inter-ests focus on surface-bound (bio-)organic chemistry and bionanotechnology. He isalso a Distinguished Adjunct Professor of chemical engineering (King AbdulazizUniversity, Jeddah, Saudi Arabia), is the founder of the company Surfix, serves onthe Editorial Advisory Board of Langmuir, Applied Surface Science and AdvancedMaterials Interfaces.

Michel Nielen is Principal Scientist at RIKILT in Wageningen, The Netherlands,

is also Scientific Director of TI-COAST, the Dutch public–private partnership onanalytical science and technology. His research covers a range of technologies foradvanced food analysis, including mass spectrometry, biosensing and combinationsthereof.