You are on page 1of 7

Colloids and Surfaces A: Physicochemical and Engineering Aspects 629 (2021) 127439

Contents lists available at ScienceDirect

Colloids and Surfaces A: Physicochemical and


Engineering Aspects
journal homepage: www.elsevier.com/locate/colsurfa

Detection limit enhancement of fiber optic localized surface plasmon


resonance biosensor by increased scattering efficiency and reduced
background signal
Hyeong-Min Kim , Se-Woong Bae , Jae-Hyoung Park *, Seung-Ki Lee *
Department of Electronics and Electrical Engineering, Dankook University, Yongin 16890, Republic of Korea

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• The effective coverage of AuNPs is


selected on the optical fiber.
• For improvement of scattering effi­
ciency, AuNPs are directly grown on
fiber optic surface.
• Surface etching is executed for reduc­
tion of background in bare surface.
• The enhancement of the LODs is evalu­
ated for several biomarkers.

A R T I C L E I N F O A B S T R A C T

Keywords: To reduce the limit of detection (LOD) in fiber optic localized surface plasmon resonance (FO LSPR) sensors,
Antireflective surface design strategies with high scattering efficiency and low background signal are proposed. Despite the cost
Biosensor effectiveness of fabrication, miniaturization capabilities, and high portability of optic fibers, the LOD is a limiting
Localized surface plasmon resonance
factor for fiber optic sensor applications. To overcome this limitation, we propose methods to increase the
Optical fiber
scattering efficiency and reduce background signal using gold capping and reactive-ion etching (RIE). Specif­
Seed-mediated growth
ically, three strategies are implemented. First, the effective coverage for detection is selected by analyzing the
sensor output according to the density of gold nanoparticles (AuNPs), which have been immobilized on the cross-
section of the optical fiber. Next, the sensitivity is studied by examining changes in scattering efficiency of the
AuNPs grown with different capping times. Third, RIE is performed on the fiber optic surface to reduce the
reflectance in areas where the AuNPs are not fixed, thereby decreasing the background signal. Thyroglobulin,
amyloid beta monomer, and oligomer are measured using the thus-fabricated FO LSPR sensor. The results are
compared with those obtained with a sensor fabricated without using these strategies. The former showed the
lower LOD and higher accuracy for biomarkers of the two.

1. Introduction past few decades, nanotechnology has been widely applied in biosensing
for detecting low antigen concentrations [3,4], because nanostructures
The detection of antigens at low concentrations is essential to pre­ can overcome the limitations of sensor characteristics with their high
vent disease progression with early diagnosis [1]. The demand for surface-to-volume ratio [5]. For instance, localized surface plasmon
technologies capable of highly sensitive, accurate, and preferably resonance (LSPR) has been representative applicant for highly sensitive,
real-time determination of diseases is rapidly increasing [2]. Over the label-free, and real-time detection of biological species. LSPR is caused

* Corresponding authors.
E-mail addresses: parkjae@dankook.ac.kr (J.-H. Park), skilee@dankook.ac.kr (S.-K. Lee).

https://doi.org/10.1016/j.colsurfa.2021.127439
Received 15 May 2021; Received in revised form 19 August 2021; Accepted 21 August 2021
Available online 26 August 2021
0927-7757/© 2021 Elsevier B.V. All rights reserved.
H.-M. Kim et al. Colloids and Surfaces A: Physicochemical and Engineering Aspects 629 (2021) 127439

by the absorption and scattering of light by free electrons that collec­ 2. Materials and methods
tively vibrate in noble metal nanoparticles with subwavelength scale of
the light [6]. When the receptor is introduced on the surface of the 2.1. Materials
nanoparticle, the LSPR principle can be used for immunoassay because
the band by the extinction changes according to variations in the sur­ Sulfuric acid (H2SO4, ≥ 95.0%), hydrogen peroxide (H2O2, ≥
rounding refractive index [7]. The adsorption of antigens generates a 34.5%), sodium citrate dihydrate (C3H4OH(COONa)3⋅2H2O, ≥ 99.0%),
difference in refractive index. In particular, using optical fibers as sub­ isopropanol (C3H8O, ≥ 99.5%), and glycerol (C3H8O3, ≥ 99.0%) were
strate for fixation of nanoparticles shows enhancements in terms of purchased from Daejung Chemicals, Republic of Korea. 3-(ethox­
cost-effectiveness, miniaturization, sample consumption, and ease of ydimethylsilyl)propylamine (APDMES, C7H19NOSi, 97%), gold (III)
handling of biosensors, which has been introduced as the so-called fiber chloride trihydrate (HAuCl4⋅3H2O, ≥ 99.9%), and bovine serum albu­
optic LSPR (FO LSPR) probes [8]. Attempts to achieve the low limit of min (BSA, ≥ 98%) were supplied by Sigma Aldrich, USA. Gold etchant
detection (LOD) for early detection have recently proceeded in FO LSPR (TFA) was purchased from Transene, USA. Borate buffer (pH 8.5, 50
applications. Martínez-Hernández et al. made multi-layered gold nano­ Mm) was obtained from Bioworld, USA. The antibody and antigen of Tg
particle (AuNP) structure by layer-by-layer assembly using poly­ were obtained at a commercial kit of Shinjin Medics, Republic of Korea.
electrolyte [9]. AuNPs and zinc oxide nanoparticles were successively Purified anti-β-amyloid, 1-8 antibody (SIG-39110), Human β-amyloid
immobilized on the tapered region of optical fiber [10]. Although these peptide (1-42) were gained in BioLegend, USA. The Aβ oligomer was
methods show improved sensitivity, they suffer from limitations asso­ prepared by incubating the monomer solution in a convection oven of
ciated with complexity and reproducibility in regard to fabrication 37 ◦ C for 6 h. The optical fiber (FG105LCA, 105-μm core diameter) was
processes. Also, these techniques are confined to the modification of acquired from Thorlabs, USA. For the preparation of aqueous solutions,
metal nanoparticles. Therefore, methods that can simply adjust nano­ we used deionized (DI) water with a resistivity of 18.2 MΩ cm (Aqua
particles and further modify other parts except for nanoparticles are MAX™-ultra, Youngin Chromass, Republic of Korea).
demanded for the development of FO LSPR biosensor which is detect­
able to the small quantity of antigens. 2.2. Synthesis of AuNPs
For the accomplishment of a low LOD in biosensor applications, we
easily induce the increase of scattering efficiency in AuNPs by in situ The so-called Turkevich method, which chemically synthesizes the
controlling the particle size on an optical fiber. In addition, surface spherical AuNPs by reducing gold ions with citrate ions, was used in this
texturing is applied on the fiber optic cross-section for the diminishment study [19]. The gold colloidal solution is prepared by adding sodium
of the background signal. Owing to their usefulness in clinical studies citrate dihydrate aqueous solution to a gold chloride trihydrate aqueous
and early interventions, biosensing devices should exhibit LODs of the solution. The carboxyl groups in sodium citrate reduce the gold ions in
order of pg/ml [11]. LSPR-based sensors are essentially refractometers the gold chloride trihydrate solution, leading to the organization of
[12]. The LOD is commonly given by the smallest detectable difference small needle-like crystals. The AuNP size gradually increased as citrate
in refractive index with reliable results [13]. In other words, the sensi­ ions adsorbed on the surface the crystals. When growth stops owing to
tivity, which is defined as the rate of change in the output according to the consumption of most citrate ions, an inter-particle repulsive force
the refractive index, needs to be improved to reduce the LOD [14]. In acts due to the negative surface charge in carboxyl groups, resulting in a
addition, the sensitivity is directly related to the LOD as: LOD = 3σ/S certain inter-particle distance [20].
[15], where σ is the standard deviation of the measured values and S is We set the target diameter of AuNPs at approximately 50 nm. This
the sensitivity in the used sensor. It is known that the sensitivity of the condition was confirmed that ensure nanoparticle’s reproducibility and
LSPR sensors is closely related to the scattering efficiency of noble metal robustness for fabrication environment in fiber optic configuration
nanoparticles [16]. The reduction of background signal is also associ­ based on the previous study [21]. The synthetic process is as follows. A
ated with enhanced sensitivity because it makes the signal easier to total of 50 ml of 0.01% gold chloride trihydrate solution was vigorously
distinguish [17]. stirred in an oil bath at 105 ◦ C. Then, 0.5 ml of 1% sodium citrate so­
This study focuses on three strategies to enhance the LOD in an FO lution was added and then reacted for 20 min. The colloidal solution was
LSPR biosensor. First, the density of AuNPs on the cross-section of the cooled to room temperature with stirring and stored at 4 ◦ C until use.
optical fiber was optimized. The sensitivities of FO LSPR sensors with The ultraviolet–visible spectrum (UV1800, Shimadzu, Japan) of the
different nanoparticle coverages were analyzed. Second, the scattering prepared colloid is displayed in Fig. S1.
efficiency of the nanoparticles were improved through a gold-capping
process that increases the size of the AuNPs. This also ensures the 2.3. Fabrication of FO LSPR sensor with improved scattering efficiency
durability of the AuNPs immobilized on the fiber optic surface. Third, and reduced background signal
reactive-ion etching (RIE) was conducted on the non-blocked surface by
nanoparticles. AuNPs act as a mask. Because of the decreased reflectance The schematic diagram of the fabrication of the FO LSPR sensor with
in the roughened surface owing to the etching process, the background gold capping and RIE for improved scattering efficiency and reduced
signal by the reflection in the bare surface reduced. That is, the scat­ background signal is shown in Fig. 1. The fabrication process largely
tering efficiency of AuNPs on the optical fiber was controlled and involved the immobilization of AuNPs (Fig. 1(a) and (b)), capping of
improved, and the background from the bare surface without nano­ nanoparticles based on seed-mediated growth (Fig. 1(c) and (d)), and
particles decreases through three methods. To test the effectiveness of bare surface etching by RIE (Fig. 1(e)). The attachment technique of
the applied strategies, the sensitivities were evaluated for each fabri­ AuNPs on the fiber optic end facet has been reported in previous
cation process. Finally, the proposed FO LSPR sensor was employed to research [22]. Briefly, the optical fiber was firstly cut (S326, OFS Fitel,
measure thyroglobulin (Tg), amyloid beta (Aβ) monomer, and oligomer USA) for a clear cross-section in the step of AuNP fixation. Then, Piranha
to determine whether the LOD is enhanced. Gold capping and surface treatment (3:1 mixture of sulfuric acid and hydrogen peroxide) was
etching were conducted in preliminary research, but the focus was to performed for surface cleaning. To functionalize amine groups for the
evaluate the fabrication potential [18]. Here, we measured the impact of adsorption of AuNPs [23], the optical fiber was immersed in a 5%
strategies on sensitivity, quantitatively detected biomarkers, and eval­ APDMES solution diluted in isopropanol (Fig. 1(a)). Finally, nano­
uated enhancements. The clinical applicability of the proposed structure particles were attached onto the optical fiber with a colloidal solution
is assessed here based on the results. (Fig. 1(b)). The density of particles on the fiber optic surface was
adjusted by varying the treatment times in the APDMES and gold
colloidal solutions. A field-emission scanning electron microscopy

2
H.-M. Kim et al. Colloids and Surfaces A: Physicochemical and Engineering Aspects 629 (2021) 127439

Fig. 1. Schematic diagram of the fabrication of FO LSPR sensor for improved scattering efficiency and reduced background signal. The capping process for the
growth of AuNPs and surface etching by RIE are applied.

(FE-SEM, S5200, Hitachi, Japan) image of AuNPs immobilized on a fiber 2.5. Measurement protocol the immune response
optic tip-end is shown in Fig. S2. The average size of nanoparticles was
44.6 ± 5.3 nm, and small aggregation was found on the end face of the The antibody–antigen reaction measurement proceeded within the
optical fiber. fluidic channel. The fluidic channel provides convenience, low sample
To improve the scattering efficiency of nanoparticles, seed-mediated consumption, and a stable measurement environment [31]. Before the
growth-based gold capping was performed [24]. The simplest way of antibody–antigen interaction, the antibody and blocking agent binding
enhancing the scattering efficiency is to increase the size of the nano­ treatments were performed on the surface of the particles. The FO LSPR
particles [25]. We grew nanoparticles on the optical fiber using the sensor surface was flushed with DI water for at least 5 min to stabilize
already-immobilized particles as seeds. Firstly, seeds were coated by a the signal before the antibody binding. Next, the antibody solution
reductant (carboxyl groups in sodium citrate dihydrate) to reduce gold diluted to 20 μg/ml in a borate buffer was injected into the channel. The
ions on the surface of AuNPs (Fig. 1(c)). Then, the capping process for supply was continued for 15 min at a flow rate of 10 μl/min. Antibodies
bigger nanoparticles was carried out and completed in a gold chloride with amine groups are ionically adsorbed on the surface of negatively
trihydrate solution containing the gold ions (Fig. 1(d)). The concentra­ charged nanoparticles [32]. Antibodies with amine groups reveal their
tions of sodium citrate dihydrate and gold chloride trihydrate aqueous positive charge in the buffer solution. Sufficient washing was performed
solutions were 0.5 mM and 1 mM, respectively. to remove unbound antibodies. Then, a 1% BSA aqueous solution was
To weaken the background signal, RIE (Plasmalab 80 Plus, Oxford supplied for 15 min to prevent nonspecific binding except for the anti­
Instruments, Germany) was implemented on the surface of the bare body–antigen reaction [33]. The excess BSA was thoroughly removed
optical fiber to which AuNPs are not fixed. The capping method is used for at least 5 min in a DI water atmosphere. Finally, an immune response
to grow particles on an optical fiber causes physical adsorption between was elicited, which ended within 5 min. After the termination of the
the fiber and nanoparticles, which allows the successful act of the AuNPs immunoassay, the outputs of the sensor were recorded in DI water
as a mask [26]. The area for the transmission of incident light was also before and after antigen binding. The net change of the signal observed
protected by particles. In the FO LSPR sensor output collected from the in DI water before and after the immune response is defined as the
reflective structure in which nanoparticles are formed on the fiber end output by antigen binding. The output changes recorded in real-time
face, the background signal reflected from the bare surface as well as the during a series of reactions for immunoassay are posted in the Fig. S4.
scattered signal from the AuNPs is always present. If the background Owing to the miniaturization of the optical fiber, the entire measure­
signal is diminished to isolate only the LSPR output, the quantification ment of the immune response takes less than 1 h [34]. The conditions
ability of the sensor is enhanced [27]. Reflectance is known to decrease regarding reaction time and concentration have been adapted from
by building a subwavelength structure (antireflective structure) on a previous research [35].
substrate using RIE [28,29]. We executed dry etching (O2/CF4 of
5 SCCM/50 SCCM, power of 150 W, pressure of 55 mTorr) on the fiber 3. Results and discussion
optic surface (Fig. 1(e)).
3.1. Optimization of nanoparticle density

2.4. Optical system for FO LSPR sensor measurement The signal intensity should be sufficient to distinguish the target
from the background [36]. The dense coverage of AuNPs on the optical
Our simple optical system consisted of a light source, a detector, and fiber causes an enhanced interaction between the nanoparticles and the
a 1 × 2 optical fiber coupler (Thorlabs). One end of the coupler had no incident light, and this increases the amount of scattered light [37].
connector and was spliced to the FO LSPR sensor (A-80S, Walfront, Increasing the number of mediators (AuNPs) can also improve sensi­
China). In side of two ends, one end was joined to the light source and tivity by the expanded sensing area. We varied the density of immobi­
the other to the detector. The input from the source excited AuNPs on lized AuNPs on the fiber optic cross-section and analyzed the effect of
the fiber optic surface and scattered signal collected at the detector the density in the sensitivity before gold capping and surface etching are
through a coupler [30]. When the spectra were observed, white light introduced. The coverage is adjusted by controlling the amine func­
(LS-1, Ocean Optics, USA) was used as a source, and a spectrometer tionalization time/particle fixation time (Fig. 1(a)/(b)). Both amine
(SM200, Spectral Products, USA) as a detector. When monitoring im­ functionalization and particle fixation times can affect final AuNP den­
mune responses, a laser (Iflex-2000, Qioptiq, UK) and a photodetector sity on an optical fiber but the amine functionalization time was
(PDA36A, Thorlabs) were used for real-time visualization of the signal. adjusted to be the same as the fixation time of particles for the proper
To minimize external influences, the measurements were conducted management of conditions for the FO LSPR fabrication. The amine
with the fluidic chip in which the FO LSPR sensor was inserted in the functionalization time/particle fixation time was increased from
fluidic channel. A schematic diagram of the system is shown in Fig. S3.

3
H.-M. Kim et al. Colloids and Surfaces A: Physicochemical and Engineering Aspects 629 (2021) 127439

60 min/60 min to 120 min/120 min with an interval of 15 min. Then, 0 min, 60 min, and 120 min are compared. It was confirmed that the
the samples with 60 min/60 min, 75 min/75 min, 90 min/90 min, LSPR intensity increased by approximately 1.91 times owning to the
105 min/105 min, and 120 min/120 min were fabricated. Fig. 2(a) il­ improved scattering efficiency after the particles were grown up to
lustrates the measured density based on the FE-SEM pictures according 120 min. Based on this, the sensitivities were 26.0, 33.1, and 41.0 in
to different fixation times. FE-SEM images of particle distributions on samples that have capping times of 0 min, 60 min, and 120 min,
optical fiber according to different immobilization conditions were respectively. It was demonstrated that the sensitivity was improved in
given in Fig. S5. The density progressively increased with increased proportion to the gold-capping time (Fig. 3(b)). The particle growth time
fixation times. The changes in the sensor output were measured in in the capping process was fixed at 120 min because the increase in the
samples with different coverages (Fig. 2(b)), where the refractive index sensor output was saturated after 120 min (Fig. 3(c)). Here, the in­
was increased from 1.33 to 1.38 with an increment of 0.01. The solution tensities are considered in the resonance peak. The output intensity can
having various refractive indices was prepared by mixing DI water and partly represent the morphology of the AuNPs on the fiber optic surface.
glycerol with different ratios. In each sample, sensitivities (slopes) were
displayed in the inset of Fig. 2(b). Sensitivity is defined as the ratio of the
change in LSPR output at resonance peak versus the variation in the 3.3. Selection in the etching time of the fiber optic surface
refractive index. Although an upward trend in sensitivity was observed
owing to the increased density of AuNPs, the sample with a density of Upon using the reflective structure with the fiber optic tip-end for
31.5% (fixation condition of 105 min/105 min) was the most sensitive measurement, the detector collects both the LSPR signal, which is
(Fig. 2(c)). This is interpreted to be due to a partial decrease in sensi­ scattered from AuNPs, and the background signal, which is reflected
tivity owing to aggregation between nanoparticles in the over-dense from the bare fiber optic surface [40]. Because the meaningful infor­
sample (fixation condition of 120 min/120 min) [38]. In the subse­ mation for LSPR measurement is the scattered light generated from the
quent experiments, the amine functionalization time/particle fixation AuNPs, the reflected light from the bare surface (background signal)
time was set to 105 min/105 min. should be reduced as much as possible [41]. An antireflective surface is
formed by RIE on the fiber optic end face, where capped AuNPs function
as a mask. The etching time was 5 s, 10 s, and 15 s, respectively. The
3.2. Modification in capping time of AuNPs etching process is performed only for a maximum of 15 s because
immobilized particles on the surface are lost if it exceeds 15 s. The
To increase the scattering efficiency and improve the stability of the surface image of the FO LSPR sensor which was etched for more than
nanoparticles on the optical fiber, gold-capping process is conducted. 20 s was provided in Fig. S8. The roughness (μSurf, KORTherm Science,
The enhancement in scattering efficiency was confirmed by measuring Republic of Korea) of the fiber optic end facet was determined for each
the sensitivity in the samples with different growth times. A reductant etching time (Fig. S9) [18,42]. The surface roughness was measured
was treated on the optical fiber to which AuNPs were immobilized for after AuNPs are removed from the surface by an etchant because the
5 min, and the nanoparticles were then grown for up to 120 min in a presence of particles can affect the roughness. The background signal
solution containing gold ions. Spectra were periodically observed during was also recorded in the absence of AuNPs, and it was confirmed that the
growth. In the growth process of AuNPs by capping, the LSPR intensity background decreased in proportion to the etching time (Fig. 4(a)). The
was gradually strengthened with larger AuNPs (increase in the increase in the etching time makes the surface rougher, which weakens
coverage) and improved scattering efficiency. The recorded spectra ac­ the reflectivity from the surface where nanoparticles are not present
cording to the capping time are shown in Fig. S6. Because the increase in (background signal) [43]. In Fig. 4(a), the sensor output increased with
LSPR intensity is insignificant after a capping time of 120 min, it is the lengthened etching time. At this time, background is gradually
believed that the growth is completed with large consumption of the decreased. The sensor output is the LSPR intensity at the resonance peak
reducing agent. After the capping process for 120 min, the mean observed in the proposed FO LSPR sensor. The background is to record
diameter of the nanoparticles increased to 64.0 ± 6.5 nm with a ratio of the signal which is reflected from the bare cross-section of an optical
0.16 nm/min. The magnitude of the standard deviation in the particle fiber after surface treatment and AuNP removal. The increased rough­
diameter was maintained even after the growth procedure [39]. FE-SEM ness in the fiber optic cross-section reduces the reflection of the signal
photographs under different growth times and the correspondingly ob­ from any direction, such as the direction that goes out from the bare
tained changes of AuNP density were presented in Fig. S7. Since the surface and the scattered signal that enters the optical fiber again. In
capping process was applied after AuNPs were fixed on the surface of the other words, the reflected background signal from the bare fiber optic
optical fiber, an increase in the size of the nanoparticles was observed, surface without AuNPs is reduced, and the reflection of the scattered
but the rearrangement and the change in distribution were insignificant. light at which the optical fiber enters from the outside also decreases
In Fig. 3(a), the sensitivities in three samples with capping times of (the increase in the LSPR collection from AuNPs), which has the effect of

Fig. 2. Optimization of fixation time: (a) nanoparticle density at the optical fiber surface according to the amine functionalization time/particle fixation time, (b) the
variation of the measured sensor output at various refractive indices with density, and (c) sensitivity (slope) according to the coverage. The slope is obtained by a
linear regression.

4
H.-M. Kim et al. Colloids and Surfaces A: Physicochemical and Engineering Aspects 629 (2021) 127439

Fig. 3. AuNP growth time (capping time) in samples with different capping times, (a) variation in the measured sensor’s output with the refractive index and (b)
obtained sensitivities based on the linear fitting, (c) the variation of the intensity with the capping time in one sample.

Fig. 4. Results by RIE on the FO LSPR sensor surface: (a) LSPR output and background signal, which are measured according to the surface etching time, (b) the
variation of the FO LSPR sensor output with the refractive index, and (c) variation in the measured sensitivity with the etching time. The background is monitored
with the removed state of AuNPs after surface etching.

increasing the LSPR intensity to be observed on the detector. The effect to determine the degree to which the LOD has been enhanced with the
of the decreased background on sensitivity is evaluated in Fig. 4(b) and proposed methods. The differences in the measured signal in DI water
(c). The FO LSPR sensor with three strategies responded with the coef­ before and after the antibody-antigen reaction are shown in Fig. 5. As
ficient of determination (R2) of 0.981, which means that high correla­ targets, Tg, Aβ monomer, and oligomer were chosen. Tg and Aβ are
tion and linearity between sensor output and change in refractive index. widely known to be specific biomarkers for thyroid cancer and Alz­
R2 was also obtained based on linear regression with the slope. As a heimer’s disease, respectively. A low LOD is required when monitoring
result of assessing the sensitivities in each sample based on the measured for recurrence because Tg should not be produced at all in patients who
value in various refractive indices, it can be seen that the sensitivity have undergone thyroidectomy [44], and the presence of even small
improved in proportion to the etching time of the fiber optic surface. amounts of Tg indicates the recurrence of thyroid cancer. The reference
Therefore, it is desirable to select a 15 s etched FO LSPR sensor for the value of Aβ for determining Alzheimer’s disease is several hundred
enhancement of the LOD. pg/ml, and a lower LOD is needed for early diagnosis [45]. Each con­
In summary, the condition for immobilization of AuNPs on the op­ centration is repeatedly detected three times to compensate for the
tical fiber (density of nanoparticles) for high sensitivity was determined. uncertainty in the measurement. A total of five concentrations of Tg
In addition, the scattering efficiency was improved through a gold- (from 0.1 pg/ml to 1000 pg/ml) were measured by two types of FO
capping process that increases the size of the particles, and the re­ LSPR sensors. The means and standard deviations of the measured
flected background from the smooth surface in the optical fiber was values by antigen binding are illustrated in Fig. 5(a). It was confirmed
reduced by performing an etching process that roughens the surface. The that there were differences in the intensity measured by the FO LSPR
improvement in the sensitivity was estimated when each strategy was sensor with strategies such as AuNP density optimization, scattering
applied. Since the efficiency of sensitivity increase by surface treatment efficiency improvement, and background reduction being higher than
was the most dominant with approximately 2.9 times, the background results obtained by the sensor without the strategies. In the curves of
relief by the surface etching will mainly contribute to the LOD Fig. 5(a), LODs were obtained on the basis of the linear regression
improvement. As a result, the immunoassay is proceeded by an FO LSPR equation and three times the standard deviations in the measured
sensor with conditions of the amine functionalization time/particle values. The LOD was enhanced by a factor of approximately 2.4 times
fixation time of 105 min/105 min, a capping time of 120 min, and an from 5.1 pg/ml to 2.1 pg/ml when compared to the case before the
etching time of 15 s. introduction of the methods for lower LOD. This trend was also the same
in the measurement results of Aβ monomer and oligomer (Fig. 5(b) and
3.4. Confirmation of LOD enhancement (c)). The LOD in the monomer became 2.5 times lower, moving from
111.9 pg/ml to 45.1 pg/ml, and the LOD in the oligomer improved by a
The immune responses are detected in the fabricated FO LSPR sensor factor of 2.2, changing from 33.6 pg/ml to 15.0 pg/ml. These results
with and without the capping and etching process, respectively. This is indicate the effectiveness of our proposed strategies in the enhancement

5
H.-M. Kim et al. Colloids and Surfaces A: Physicochemical and Engineering Aspects 629 (2021) 127439

Fig. 5. Immunoassay results obtained by the proposed sensor and sensor fabricated without gold capping and surface etching: intensity differences gained in DI
water before and after the antibody-antigen interaction in (a) Tg, (b) Aβ monomer, and (c) oligomer, respectively. The various target concentrations were measured.

maximum sensitivity appeared when the time in each step was 105 min.
Table 1
Second, the scattering efficiency and durability were improved by
Comparison of analytical performance for antigens with different biosensor
growing the nanoparticles using the capping process. The LSPR intensity
configurations. MIPs is an abbreviation of molecular imprinting polymers.
and scattering efficiency increased with increase in growth time of
Mechanism Structure Analyte LOD Ref. AuNPs. Particle growth was saturated at a capping time of 120 min.
(pg/ml)
Third, RIE was performed at the optical fiber tip-end using AuNPs as a
Fluorescence Tannylated ferritin Tg 4.3 [50] mask. The surface of the optical fiber was roughened by the etching
nanocage
process, and the reflectivity in the unprotected surface by AuNPs was
Colorimetry MIPs and hemin- Tg 100 [51]
graphene nanosheets lowered accordingly. As a result, the background signal that is reflected
composite in the bare surface without AuNPs was reduced. When the time of the
Electrochemistry Gold electrode on Aβ 140 [52] surface etching exceeded 15 s, AuNPs fell off so the process was carried
nanopillar array monomer out up to 15 s. There were improvements as a result of evaluating the
Ratiometricelectrochemistry Glassy carbon electrode Aβ 500 [53]
monomer
sensitivity of the proposed FO LSPR sensor when each strategy was
LSPR AuNP and etched Tg 2.1 This introduced. The proposed FO LSPR sensor was ultimately applied in
substrate Aβ 45.1 work biosensing applications. Tg, Aβ monomer, and oligomer with various
monomer concentrations were measured to quantify LODs. In the measurements of
the biomaterials, the proposed sensor showed enhanced LODs and CVs
compared to the FO LSPR sensor to which strategies were not intro­
duced. Therefore, by using capping and surface etching, the developed
of LOD. In the proposed sensor, the reduction of the coefficient of FO LSPR sensor with improved scattering efficiency and reduced back­
variation (CV) also contributes to the upgrade of the LOD. The CV is the ground signal can contribute to applications that require a low LOD.
standard deviation/mean as a percentage, which is an index that rep­
resents the spread of the measured values, and it is directly related to the CRediT authorship contribution statement
LOD [46]. After introducing the strategies in the FO LSPR sensor, the CV
decreased by an average of about 8.0%. This is presumed to be due to the Hyeong-Min Kim: Validation, Investigation, Data curation, Writing
slight improvement in the measurement accuracy by the reduced – original draft. Se-Woong Bae: Validation, Investigation, Data cura­
background [47]. In the proposed sensor, a satisfactory correlation of tion. Jae-Hyoung Park: Conceptualization, Methodology, Writing –
the intensity difference and concentration of biomarkers was also ac­ review & editing, Supervision, Resources, Project administration.
quired with an average R2 of 0.930 by linear regression. LODs obtained Seung-Ki Lee: Term, Conceptualization, Methodology, Writing – review
by the proposed FO LSPR sensor are suitable for practical and clinical & editing, Supervision, Resources, Project administration.
applications considering that the reference values of Tg and Aβ are
5 ng/ml (in relapse patient) and 680 pg/ml, respectively [48,49]. Declaration of Competing Interest
In the case of Tg detection, LOD enhanced approximately 2–48 times
compared with other detection methods using fluorescence and color­ The authors declare that they have no known competing financial
imetry in Table 1. For Aβ based on monomer, good performance with interests or personal relationships that could have appeared to influence
respect to electrochemical and ratiometric techniques was as well evi­ the work reported in this paper.
denced considering that recently reported literature only reached the
LOD between 0.14 ng/ml and 0.5 ng/ml. Acknowledgements

4. Conclusion This work was supported by the National Research Foundation of


Korea (NRF) grant funded by the Korea government (MSIT) (Nos. NRF-
In summary, three fabrication conditions were tuned to improve the 2018R1A2B6001361 and NRF-2019R1A2C1083969).
LOD of the FO LSPR sensor. First, the sensor’s sensitivity was optimized
by varying the density of immobilized AuNPs on the fiber optic surface. Appendix A. Supplementary material
To vary the density of AuNPs, the amount of the formed amine group on
the optical fiber was adjusted by the dipping time in the APDMES so­ Supplementary data associated with this article can be found in the
lution, and the amount of electrostatically bound AuNP with amine online version at doi:10.1016/j.colsurfa.2021.127439.
groups was modified by the dipping time in the gold colloidal solution.
As a result of changing the two conditions, it was confirmed that the

6
H.-M. Kim et al. Colloids and Surfaces A: Physicochemical and Engineering Aspects 629 (2021) 127439

References [27] P. Singh, SPR biosensors: historical perspectives and current challenges, Sens.
Actuators B Chem. 229 (2016) 110–130.
[28] Z. Yue, H. Shen, Y. Jiang, Antireflective nanostructures fabricated by reactive ion
[1] S. Arif, S. Qudsia, S. Urooj, N. Chaudry, A. Arshad, S. Andleeb, Blueprint of quartz
etching method on pyramid-structured silicon surface, Appl. Surf. Sci. 271 (2013)
crystal microbalance biosensor for early detection of breast cancer through salivary
402–406.
autoantibodies against ATP6AP1, Biosens. Bioelectron. 65 (2015) 62–70.
[29] X. Ye, X. Jiang, J. Huang, F. Geng, L. Sun, X. Zu, W. Wu, W. Zheng, Formation of
[2] M. Al Mahfuz, M.A. Mollah, M.R. Momota, A.K. Paul, A. Masud, S. Akter, M.
broadband antireflective and superhydrophilic subwavelength structures on fused
R. Hasan, Highly sensitive photonic crystal fiber plasmonic biosensor: design and
silica using one-step self-masking reactive ion etching, Sci. Rep. 5 (2015) 1–10.
analysis, Opt. Mater. 90 (2019) 315–321.
[30] M. Sanders, Y. Lin, J. Wei, T. Bono, R.G. Lindquist, An enhanced LSPR fiber-optic
[3] S. Jiang, M. Saito, M. Murahashi, E. Tamiya, Pressure free nanoimprinting
nanoprobe for ultrasensitive detection of protein biomarkers, Biosens. Bioelectron.
lithography using ladder-type HSQ material for LSPR biosensor chip, Sens.
61 (2014) 95–101.
Actuators B Chem. 242 (2017) 47–55.
[31] J. Roether, K.Y. Chu, N. Willenbacher, A.Q. Shen, N. Bhalla, Real-time monitoring
[4] Y. Wang, J. Zhou, J. Li, Construction of plasmonic nano-biosensor-based devices
of DNA immobilization and detection of DNA polymerase activity by a microfluidic
for point-of-care testing, Small Methods 1 (2017), 1700197.
nanoplasmonic platform, Biosens. Bioelectron. 142 (2019), 111528.
[5] S. Kunwar, M. Sui, P. Pandey, Z. Gu, S. Pandit, J. Lee, Improved configuration and
[32] A. Makaraviciute, A. Ramanaviciene, Site-directed antibody immobilization
LSPR response of platinum nanoparticles via enhanced solid state dewetting of In-
techniques for immunosensors, Biosens. Bioelectron. 50 (2013) 460–471.
Pt bilayers, Sci. Rep. 9 (2019) 1–14.
[33] Y.L. Jeyachandran, J.A. Mielczarski, E. Mielczarski, B. Rai, Efficiency of blocking of
[6] T. Xu, Z. Geng, Strategies to improve performances of LSPR biosensing: structure,
non-specific interaction of different proteins by BSA adsorbed on hydrophobic and
materials, and interface modification, Biosens. Bioelectron. 174 (2021), 112850.
hydrophilic surfaces, J. Colloid Interface Sci. 341 (2010) 136–142.
[7] M. Ashaduzzaman, S.R. Deshpande, N.A. Murugan, Y.K. Mishra, A.P. Turner,
[34] S. Jia, C. Bian, J. Sun, J. Tong, S. Xia, A wavelength-modulated localized surface
A. Tiwari, On/off-switchable LSPR nano-immunoassay for troponin-T, Sci. Rep. 7
plasmon resonance (LSPR) optical fiber sensor for sensitive detection of mercury
(2017) 1–10.
(II) ion by gold nanoparticles-DNA conjugates, Biosens. Bioelectron. 114 (2018)
[8] B.D. Gupta, A.M. Shrivastav, S.P. Usha, Surface plasmon resonance-based fiber
15–21.
optic sensors utilizing molecular imprinting, Sensors 16 (2016) 1381.
[35] M. Uh, S.K. Kang, J.H. Park, D.H. Jeong, H.Y. Lee, S.M. Lee, S.K. Lee, Analysis and
[9] M.E. Martínez-Hernández, J. Goicoechea, F.J. Arregui, Hg2+ optical fiber sensor
optimization of antibody immobilization for immunoassay using fiber-optic
based on LSPR generated by gold nanoparticles embedded in LBL nano-assembled
localized surface plasmon resonance biosensors, Nanosci. Nanotechnol. Lett. 8
coatings, Sensors 19 (2019) 4906.
(2016) 8–12.
[10] G. Zhu, L. Singh, Y. Wang, R. Singh, B. Zhang, F. Liu, B.K. Kaushik, S. Kumar,
[36] D.D. Galvan, V. Parekh, E. Liu, E.L. Liu, Q. Yu, Sensitive bacterial detection via
Tapered optical fiber-based LSPR biosensor for ascorbic acid detection, Photon.
dielectrophoretic-enhanced mass transport using surface-plasmon-resonance
Sens. (2020) 1–17.
biosensors, Anal. Chem. 90 (2018) 14635–14642.
[11] S.H. Yeom, M.E. Han, B.H. Kang, K.J. Kim, H. Yuan, N.S. Eum, S.W. Kang,
[37] H. Yockell-Lelièvre, F. Lussier, J.F. Masson, Influence of the particle shape and
Enhancement of the sensitivity of LSPR-based CRP immunosensors by Au
density of self-assembled gold nanoparticle sensors on LSPR and SERS, J. Phys.
nanoparticle antibody conjugation, Sens. Actuators B Chem. 177 (2013) 376–383.
Chem. C 119 (2015) 28577–28585.
[12] S. Chen, Y. Liu, Q. Yu, W. Peng, Microcapillary-based integrated LSPR device for
[38] M.H. Tu, T. Sun, K.T.V. Grattan, Optimization of gold-nanoparticle-based optical
refractive index detection and biosensing, J. Lightw. Technol. 38 (2020)
fibre surface plasmon resonance (SPR)-based sensors, Sens. Actuators B Chem. 164
2485–2492.
(2012) 43–53.
[13] S.H. Shen, I.S. Wang, H. Cheng, C.T. Lin, An enhancement of high-k/oxide stacked
[39] C. Wei, Q. Liu, Shape-, size-, and density-tunable synthesis and optical properties of
dielectric structure for silicon-based multi-nanowire biosensor in cardiac troponin I
copper nanoparticles, CrystEngComm 19 (2017) 3254–3262.
detection, Sens. Actuators B Chem. 218 (2015) 303–309.
[40] S. Barizuddin, S. Bok, S. Gangopadhyay, Plasmonic sensors for disease detection – a
[14] N. Cennamo, L. Zeni, P. Tortora, M.E. Regonesi, A. Giusti, M. Staiano, S. D’Auria,
review, J. Nanomed. Nanotechnol. 7 (2016), 1000373.
A. Varriale, A high sensitivity biosensor to detect the presence of perfluorinated
[41] S.M. Tabakman, L. Lau, J.T. Robinson, J. Price, S.P. Sherlock, H. Wang, B. Zhang,
compounds in environment, Talanta 178 (2018) 955–961.
Z. Chen, S. Tangsombatvisit, J.A. Jarrell, P.J. Utz, H. Dai, Plasmonic substrates for
[15] J. Guo, Y. Lin, H. Huang, S. Zhang, T. Huang, W. Weng, One-pot fabrication of
multiplexed protein microarrays with femtomolar sensitivity and broad dynamic
fluorescent carbon nitride nanoparticles with high crystallinity as a highly selective
range, Nat. Commun. 2 (2011) 1–9.
and sensitive sensor for free chlorine, Sens. Actuators B Chem. 244 (2017)
[42] E.S. Gadelmawla, M.M. Koura, T.M.A. Maksoud, I.M. Elewa, H.H. Soliman,
965–971.
Roughness parameters, J. Mater. Process. Technol. 123 (2002) 133–145.
[16] K.S. Lee, M.A. El-Sayed, Dependence of the enhanced optical scattering efficiency
[43] H. Toyota, K. Takahara, M. Okano, T. Yotsuya, H. Kikuta, Fabrication of microcone
relative to that of absorption for gold metal nanorods on aspect ratio, size, end-cap
array for antireflection structured surface using metal dotted pattern, Jpn. J. Appl.
shape, and medium refractive index, J. Phys. Chem. B 109 (2005) 20331–20338.
Phys. 40 (2001) L747–L749.
[17] K. Xiao, L. Meng, C. Du, Q. Zhang, Q. Yu, X. Zhang, J. Chen, A label-free
[44] A. Miyauchi, T. Kudo, A. Miya, K. Kobayashi, Y. Ito, Y. Takamura, T. Higashiyama,
photoelectrochemical biosensor with near-zero-background noise for protein
M. Fukushima, M. Kihara, H. Inoue, C. Tomoda, T. Yabuta, H. Masuoka, Prognostic
kinase A activity assay based on porous ZrO2/CdS octahedra, Sens. Actuators B:
impact of serum thyroglobulin doubling-time under thyrotropin suppression in
Chem. 328 (2021), 129096.
patients with papillary thyroid carcinoma who underwent total thyroidectomy,
[18] S.W. Bae, H.M. Kim, J.H. Park, S.K. Lee, Improvement of fiber optic based localized
Thyroid 21 (2011) 707–716.
surface plasmon resonance sensor by optical fiber surface etching and Au capping,
[45] L.C. Brazaca, I. Sampaio, V. Zucolotto, B.C. Janegitz, Applications of biosensors in
Micro Nano Syst. Lett. 7 (2019) 1–7.
Alzheimer’s disease diagnosis, Talanta 210 (2020), 120644.
[19] F. Schulz, T. Homolka, N.G. Bastús, V. Puntes, H. Weller, T. Vossmeyer, Little
[46] J. Leuermann, A. Fernández-Gavela, A. Torres-Cubillo, S. Postigo, A. Sánchez-
adjustments significantly improve the Turkevich synthesis of gold nanoparticles,
Postigo, L.M. Lechuga, R. Halir, Í. Molina-Fernández, Optimizing the limit of
Langmuir 30 (2014) 10779–10784.
detection of waveguide-based interferometric biosensor devices, Sensors 19 (2019)
[20] J. Piella, N.G. Bastus, V. Puntes, Size-controlled synthesis of sub-10-nanometer
3671.
citrate-stabilized gold nanoparticles and related optical properties, Chem. Mater.
[47] C.R. Crick, P. Albella, H.J. Kim, A.P. Ivanov, K.B. Kim, S.A. Maier, J.B. Edel, Low-
28 (2016) 1066–1075.
noise plasmonic nanopore biosensors for single molecule detection at elevated
[21] H.H. Jeong, N. Erdene, J.H. Park, D.H. Jeong, S.K. Lee, Analysis of fiber-optic
temperatures, ACS Photonics 4 (2017) 2835–2842.
localized surface plasmon resonance sensor by controlling formation of gold
[48] M. Ozata, S. Suzuki, T. Miyamoto, R.T. Liu, F. Fierro-Renoy, L.J. DeGroot, Serum
nanoparticles and its bio-application, J. Nanosci. Nanotechnol. 12 (2012)
thyroglobulin in the follow-up of patients with treated differentiated thyroid
7815–7821.
cancer, J. Clin. Endocrinol. Metab. 79 (1994) 98–105.
[22] H.M. Kim, J.H. Park, D.H. Jeong, H.Y. Lee, S.K. Lee, Real-time detection of
[49] D. Bertens, B.M. Tijms, P. Scheltens, C.E. Teunissen, P.J. Visser, Unbiased estimates
prostate-specific antigens using a highly reliable fiber-optic localized surface
of cerebrospinal fluid β-amyloid 1–42 cutoffs in a large memory clinic population,
plasmon resonance sensor combined with micro fluidic channel, Sens. Actuators B
Alzheimer’s Res. Ther. 9 (2017) 1–8.
Chem. 273 (2018) 891–898.
[50] E. Turan, F. Şahin, Z. Suludere, H. Tümtürk, A fluoroimmunodiagnostic
[23] J. Satija, N.S. Punjabi, V.V.R. Sai, S. Mukherji, Optimal design for U-bent fiber-
nanoplatform for thyroglobulin detection based on fluorescence quenching signal,
optic LSPR sensor probes, Plasmonics 9 (2014) 251–260.
Sens. Actuators B: Chem. 300 (2019), 127052.
[24] H. Manoharan, K.C. Dharanibalaji, V.V.R. Sai, Controlled in situ seed-mediated
[51] X. Wang, K. Huang, H. Cui, H. Zhang, L. Zeng, Y. Zhou, T. Jing, Label-free
growth of gold and silver nanoparticles on an optical fiber platform for plasmonic
determination of thyroglobulin using template-probe double imprinted composites,
sensing applications, Plasmonics 15 (2020) 51–60.
Sens. Actuators B: Chem. 313 (2020), 128028.
[25] A. Bansal, J.S. Sekhon, S.S. Verma, Scattering efficiency and LSPR tunability of
[52] Y.M. Park, J. Ahn, Y.S. Choi, J.M. Jeong, S.J. Lee, J.J. Lee, B.G. Choi, K.G. Lee,
bimetallic Ag, Au, and Cu nanoparticles, Plasmonics 9 (2014) 143–150.
Flexible nanopillar-based immunoelectrochemical biosensor for noninvasive
[26] H.M. Kim, D.H. Jeong, H.Y. Lee, J.H. Park, S.K. Lee, Improved stability of gold
detection of Amyloid beta, Nano Converg. 7 (2020) 1–9.
nanoparticles on the optical fiber and their application to refractive index sensor
[53] Y. Yu, P. Wang, X. Zhu, Q. Peng, Y. Zhou, T. Yin, Y. Liang, X. Yin, Combined
based on localized surface plasmon resonance, Opt. Laser Technol. 114 (2019)
determination of copper ions and β-amyloid peptide by a single ratiometric
171–178.
electrochemical biosensor, Analyst 143 (2018) 323–331.

You might also like