You are on page 1of 37

Materials Today Chemistry 26 (2022) 101129

Contents lists available at ScienceDirect

Materials Today Chemistry


journal homepage: www.journals.elsevier.com/materials-today-chemistry/

Recent developments and fabrication of the different electrochemical


biosensors based on modified screen printed and glassy carbon
electrodes for the early diagnosis of diverse breast cancer biomarkers
P. Lakhera a, b, V. Chaudhary a, b, e, A. Jha c, R. Singh d, P. Kush d, **, P. Kumar e, *
a
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
b
CSIR-Central Scientific Instruments Organization, Sector 30-C, Chandigarh e 160030, India
c
Dolphin PG Institute of Biomedical and Natural Sciences, Dehradun, Uttarakhand-249161, India
d
School of Pharmacy, Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University Gangoh, Saharanpur, India
e
Exigo Recycling Pvt. Ltd., Noida 201309, Uttar Pradesh, India

a r t i c l e i n f o a b s t r a c t

Article history: Breast cancer (BC) is the second most common cancer among women, comprising 34% of all cancers,
Received 6 May 2022 affecting ~2.23 million women every year globally. Therefore, early diagnosis of BC is required for suc-
Received in revised form cessful treatment and hence an increased survival rate. Timely detection of various biomarkers subsisting
15 July 2022
in the blood, cells, tissues, or other biological fluids using techniques like radioimmunoassay (RIA),
Accepted 3 August 2022
Available online 6 September 2022
enzyme-linked immunosorbent assay (ELISA), and immunohistochemistry (IHC) can be advantageous in
this direction. Although all these methods are effective but they have their pros and cons, hence an
alternative non-invasive, cost-effective screening technique with high sensitivity and specificity is
Keywords:
Biosensing
required for the point of care (POC) diagnosis. Electrochemical (EC) biosensors are promising tools for
Detection POC diagnosis due to their simplicity, reliability, portability, low cost, fast response, ease of detection
Point of care even with a little sample requirement, magnified sensitivity, low detection limit, and POC testing
Challenges compatibility. Screen-printed electrodes (SPEs), and glassy carbon electrodes (GCEs) are widely used as
Nanomaterials electrodes in EC biosensors due to their easy miniaturization, and magnificent mechanical and electrical
Surface modifications properties, respectively. This comprehensive review focuses on the recent developments (covering the
period of 2015e2022) and fabrication of the different EC bioscaffolds based on modified SPEs and GCEs
for the early diagnosis of diverse BC biomarkers at different molecular levels like genomic, tran-
scriptomic, and proteomic formats. Further, circulating tumor cells (CTCs) and multiplex determination
of BC biomarkers are also discussed by using both electrodes. Considering the magnified sensitivity, most
of the reported biosensors have been made by surface modification of SPEs/GCEs with various nano-
materials (NMs), enzymes, and metals due to their unique properties. Further, the challenges associated
with these biosensors, and future perspectives have been also discussed. Therefore, this review will
impart relevant background for the design and fabrication of innovative SPE- and GCE-based POC devices
for diverse BC biomarker monitoring which would open the way for augmenting cancer diagnosis and
therapeutics. Generally, the principle and techniques remain similar while changing the analyte-bio-
recognition element (BRE), hence this review will be a proven guide for the development of similar kinds
of sensors for other analytes as well.
© 2022 Elsevier Ltd. All rights reserved.

1. Introduction

Cancer is one of the most prominent life-threatening diseases;


possesses more than 200 types; and affects approximately 60 hu-
man organs [1]. Annually, the incidence of new cancer cases in men
and women is 439/10,000. As per International Agency for Research
* Corresponding author.
** Corresponding author.
on Cancer (IARC) report, globally ~17 million cancer cases were
E-mail addresses: preetikush85@gmail.com (P. Kush), parveenkaushik7@gmail. reported in 2018 [2]. It is estimated that cancer cases and the
com (P. Kumar). number of cancer-related deaths are predicted to be 1,762,450 and

https://doi.org/10.1016/j.mtchem.2022.101129
2468-5194/© 2022 Elsevier Ltd. All rights reserved.
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

606,880 in 2022, respectively [3]. According to World Health Or- Screen-printed electrodes (SPEs), and glassy carbon electrodes
ganization (WHO), cancer will be the leading cause of approxi- (GCEs) are widely used as electrodes in EC biosensors due to their
mately 12 million deaths by 2030 [4]. Breast cancer (BC) is the low cost, easy miniaturization [12], easy surface functionalization,
second most common cancer among women, comprising 34% of all and magnificent mechanical and electrical properties [24,25].
cancers, affecting ~2.1e2.3 million women every year globally [5]. Moreover, in situ analysis can be appropriately carried out by using
In 2020, 2.261 million new BC cases were recognized and 0.685 SPEs due to their magnified sensitivity, a linear output, minimum
million people died globally. In India only, the incidence of BC cases power consumption, and ease to operate at room temperature [26].
is predicted to cross 2,05,000 and reach over 2,32,000 by 2025 [6]. Further, the sensitivity and reproducibility of these two types of
The highest prevalence rates were observed in developed coun- electrodes can be augmented by surface modification with various
tries, whereas the lowest rates were reported in Africa (23/100,000 nanomaterials (NMs) [27]. Furthermore, the specificity of the
women) and East Asia (21/100,000 women), but this statistical data electrodes can be increased by the functionalization of the working
can be underestimated due to the lack of comprehensive methods electrodes (WEs) with various specific recognition molecules like
of exact data collection in the related wide region [5]. enzymes, antibodies [28], nucleic acids [1,29,30], and aptamers
Diagnosis and treatment of BC are difficult due to the similarity [12,31,32], etc.
between normal cells and cancer cells [7]. If BC is diagnosed at the There are various reviews about the different EC biosensors for
primary/early stage, a 5-year survival rate may extend to 90% in the screening of BC biomarkers [1,5,28,33e35], but few of them are
developed countries [8], whereas A 5-year survival rate decreases focused on SPEs-based screening of biomarkers [36e38]. Addi-
to 27.4% in metastatic BC patients [5]. Therefore, early diagnosis is tionally, the bibliography about GCEs-based biosensors are exten-
required for the successful treatment of BC with an increased sive, but as per the best of our knowledge, there is no review
survival rate. Various techniques like mammography, sonography, emphasizing GCEs-based EC biosensor for BC biomarkers detection.
thermography, magnetic resonance imaging (MRI), molecular This comprehensive review highlights the fabrication principle,
breast imaging, positron emission tomography (PET), biopsy, configuration, and development of recent (covering the period of
computerized tomography (CT), etc. have been used for the 2015e2022) SPEs and GCEs-based EC biosensors for various BC
screening of BC [7,9]. These all methods are effective but possess biomarkers detection. Further, the challenges and future perspec-
certain limitations like invasiveness, costly, time-consuming, tives have been also discussed. Therefore, this review will impart
requiring trained personnel to perform and interpret the imag- relevant background for the design and fabrication of innovative
ing studies, etc. (Table 1) [7,9e11]. Cancers can be early diagnosed POC devices for diverse BC biomarker monitoring which would
by the detection of various biomarkers present in the blood, cells, open the way for augmenting cancer diagnosis and therapeutics.
tissues, or other biological fluids [1,12]. Biomarkers are certain
biologically relevant molecules (mutated gene, surface receptor 2. Breast cancer pathophysiology and biomarkers
proteins, micro-RNA (miRNA), cancer antigens (CAs), cytokines,
exosomes, circulating tumor cells (CTCs), etc.) that are overex- BC is a diversified disease with various subtypes followed by a
pressed in/or cancer cells, indicate the tumor formation/pro- series of genetic mutations, and DNA damage [39]. BC is classified
gression or metastasis in contrast to normal cells [10]. Various into four subtypes based on antigen Ki-67, estrogen receptor (ER),
biomarkers-based techniques like radioimmunoassay (RIA), human receptor tyrosine-protein kinase erbB-2 (HER2), and pro-
enzyme-linked immunosorbent assay (ELISA), fluorescent in-situ gesterone receptor (PR) [5]. There are various risk factors associated
hybridization (FISH), and immunohistochemistry (IHC) are used with the prevalence of BC like age, family history, geographic
for diagnostic purposes [9,13]. These methods are selective location, low rates of breastfeeding, socioeconomic status, lifestyle
and sensitive but possess certain limitations such as being risk factors (smoking, alcohol, diet, obesity, and physical activity),
expensive, complex, and multistep processes, and require ionizing radiation, and increased breast density, etc. (Fig. 1) [40]. BC
high-quality samples, etc. (Table 1). Therefore, an alternative most often originates from the terminal lobes (lobular) and duct
non-invasive, portable, cost-effective screening technique with (ductal) of the mammary glands. Normal breast cells convert into
high sensitivity and specificity is required for the point of care cancer cells, which are phenotypically similar to the normal ones.
(POC) diagnosis. Breast biopsy demonstrates the molecular and histological sub-
Biosensors are widely used for the detection of various BC bio- types, which play an important role in the therapeutical manage-
markers, biomedical analysis [14], environmental monitoring [15], ment of breast cancer (Fig. 2) [5]. Globally, lobular and ductal
and food manufacturing industries [16] owing to their automation, carcinoma covers 40e75% of all diagnosed BC cases [41], but ductal
low cost, sensitivity, specificity, reliability, and reproducibility. carcinoma covers ~80% of all diagnosed BC [42]. However, some-
Moreover, biosensors are fast, inexpensive, and directly detect times BC can originate from the stromal tissues that integrate the
various biomarkers in biological fluids with minimal invasion fatty and fibrous connective tissues of the breast [43].
[10,17]. Additionally, they can also be used as POC devices at doc- BC biomarkers are classified into stage-dependent biomarkers
tors' clinics/hospitals and homes. For biosensing applications, a and overexpressed biomolecule-based biomarkers (Table 2)
biosensor mainly comprises target molecules (biomarkers), bio- [1,5,9,10]. Further, stage-dependent can be subclassified as diag-
receptor/biorecognition element (BRE), and bio-transducer. The nostic (healthy vs. BC) [44], prognostic (early BC vs. advanced BC)
classification of biosensors mainly depends upon the bioreceptor [45], predictive (provide information regarding the particular
(enzyme, antibody, nucleic acid, cell), and transducer (electro- treatment success probability) [46], and pharmacodynamic/thera-
chemical (EC), optical, thermal, mass, acoustic wave, field-effect peutic biomarkers (a target biomolecule for the therapeutic mo-
transistor, and quartz crystal microbalance) [7,18]. Among various dalities). Biomolecule-based biomarkers are more significant in
types of biosensors, EC biosensors are promising for POC diagnosis contrast to the prognostic or therapeutic but there is a correlation
due to their simplicity, reliability, portability, low cost, fast between the prognostic and diagnostic value of these biomarkers
response, ease of detection even with a little sample, magnified [10,47]. Moreover, BC biomarkers can also be classified as genomic
sensitivity, low detection limit, and POC testing compatibility. (DNA), proteomic (a protein molecule), transcriptomic (messenger-
Moreover, these can be easily converted into electronic wearable RNA molecules (mi-RNAs), and metabolomic (low molecular
biosensors and could be used to obtain results via a portable device weight metabolites) based on their origin, function, and structure
such as a smartphone [15,19e23]. [48]. Fig. 3 summarizes the origination of biomarkers from
2
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Table 1
Various techniques used for the screening of breast cancer and their limitations.

Techniques Limitations

Mammography False-positive and negative results, uncomfortable involves breast compression, low sensitivity, and specificity in young
females (<40 years), can only detect 70% of breast cancers
Sonography Difficult to detect a deep-lying solid tumor, higher cost and highly experienced operators are needed to perform the
experiment
Thermography High cost, false negative, use of radiation, and sophisticated instrument
Magnetic resonance imaging Very expensive, some types of breast cancer escape detection, time-consuming process
Microwave imaging Complex procedure
Positron emission tomography Harmful ionizing radiation
Biopsy Probability to miss tumor cells; unnecessary surgery; can cause tumor metastasis; performed for late-stage; expensive
Computerized tomography Expansive scanner, low sensitivity, and risk of harmful radiation
Fluorescent in-situ hybridization Separate patients into positive and negative groups, semiquantitative results
Radioimmunoassay Complex, and time-consuming process along with radioactivity risk
Enzyme-linked immunosorbent assay Time-consuming, expensive, risk of false-positive, requires professional operators
Immunohistochemistry Complex, and time-consuming process required trained personnel for the analysis

tumorigenesis, migration, and accumulation to the different organs the WEs. This technique is further grouped into cyclic voltammetry
together with the relationship between these biomarkers. (CV), differential pulse voltammetry (DPV), linear sweep voltam-
metry (LSV), and square wave voltammetry (SWV). The ampero-
3. Electrochemical biosensors metric technique is also based on the current-potential relationship
but in this technique, the current is measured resulting from the
An EC biosensor is a device used to measure the EC behavior of oxidation or reduction of electroactive material at a constant po-
the electroactive surface to provide quantitative or semi- tential [48,50]. Chronoamperometry is another amperometric
quantitative analytical information using an EC transducer. Based technique where a square-wave potential is applied to the WEs and
on the transducer detection principle EC biosensors are grouped the current is measured as a function of time [50]. Electrochemical
into voltammetric, amperometric, impedimetric, conductometric, impedance spectroscopy (EIS) is extensively used for the surface
and potentiometric (Table 3). The voltammetric transducers are characterization of electrodes especially modified with bio-
highly sensitive and widely used for biosensing, and measure the materials. Moreover, this technique is used to assess the electrode/
current-potential relationship. In this technique, the current is electrolyte interface and the binding kinetics between the elec-
measured at various forms of potential such as potential sweep, trode and analytes. The immobilized biomolecules on the electrode
potential waveform, step potential, potential ramp, etc. is applied to surface change the interfacial electron transfer resistance and

Fig. 1. Various risk factors associated with breast cancer.

3
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Fig. 2. Origination of breast cancer from the terminal lobular unit (breast functional unit).

capacitance of the application system [50]. Conductometry is based [56]. Additionally, SPE can reduce the size of EC instruments to
on the relationship between conductance and a bio-recognition small pocket-sized ones, resulting in personal and professional use
event. Potentiometry measures the potential difference between [38].
the working and reference electrode. However, its sensitivity is A typical SPE comprises a three-electrode configuration: (1)
lower in contrast to other techniques due to the small potential working electrodes (WEs); (2) counter electrodes (CEs); and (3)
difference during the analytical reaction [51]. reference electrodes (REs). SPEs fabrication is a multistep process
that involves: (1) selection of the mesh or screen (significantly af-
4. The emergence of screen-printed electrodes (SPEs) and fects the SPEs size and geometry; (2) selection and preparation of
glassy carbon electrodes (GCEs) the appropriate viscosity inks (3e10 Pa s); (3) selection of the
suitable substrate material; and finally (iv) printing, drying and
SPEs and GCEs have been widely used as electrode materials for curing [38,57]. Precisely, SPEs are fabricated by a layer-by-layer
the fabrication of various EC biosensors due to reproducibility, and printing of ink on a substrate via using a mesh/sieve followed by
low cost. EC biosensors based on SPE and GCE provide great sta- solidification and drying of ink under thermal/ultraviolet (UV) light
bility and sensitive results which helps in the early detection of exposure. Further, the conductive track is insulated via coating with
many diseases and biomarkers [53]. Various commercial SPEs and protective non-conductive ink/paint (Fig. 4, a). Transparent flexible
GCEs in different configurations that exist with excellent perfor- plastic, paper, glass, alumina, or ceramic surfaces are commonly
mance can also purchase from different suppliers (Table 4) [38,54]. used solid substrates [54,56]. During the SPEs printing, WEs and
This section summarizes the basic fabrication principle and CEs are mostly printed with carbon paste/ink (carbon nanotubes
configuration of SPEs and GCEs for EC sensing. [CNTs], graphene [Gr], fullerene, or graphite), whereas RE is painted
SPEs are inexpensive, reproducible, and disposable EC devices with silver (Ag) ink [58]. Moreover, SPEs can be personalized as per
that can be easily fabricated in bulk without any treatment steps the requirement of ongoing study by using carbon, gold (Au), and
beforehand leading to real-time and on-site sensing [55]. These platinum (Pt) [54,56]. Additionally, the composition of printing ink
diversified materials have diagnostic applications in various areas may be modified by the addition of various substances such as
like the detection of environmental pollutants, organic, and inor- complexing agents, electrochemical (EC) signal modifiers, metal,
ganic contaminants in food, medicines, and agriculture due to easy metal oxides, etc.
miniaturization, portability, less sample requirement, and easy A glassy carbon (GC), also known as polymeric carbon or vit-
surface functionalization [26,54,55]. Moreover, SPEs circumvent reous carbon is a non-graphitic sp2 bonded carbon allotrope pro-
some common problems such as tedious cleaning and memory duced by the controlled pyrolysis of organic polymers at high
effects associated with the use of classical solid electrode materials temperatures (>2000 C) [59]. The GC microstructure comprises
4
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Table 2
Common breast cancer biomarkers for the diagnosis of breast cancer within their normal and abnormal range.

Type of biomarkers Examples Normal range Abnormal range Ref

Biomolecule based Genomic/DNA BRCA1, BRCA2 e e [1]


Transcriptomic/micro-RNA miR-21, 16, 27a, 150, 191 200 aM to 20 pM [1]
Proteomic/Glycoprotein HER2 2e15 ng/mL serum 15e75 ng/mL [3]
EGFR 2e15 mg/L >75.3 mg/L [1]
CEA 2.5 mg/L in non-smokers, 5 mg/L in a smoker [1,49]
MUC1 <31 U/mL 3100 U/mL [3]
CA15-3 <30 U/mL 30e50 U/mL [49]
CA27.29 38 U/mL [1]
ER 0.06 mg/g cytosol [1]
PR 0.06 mg/g cytosol [1]
VEGF 1e177 pg/mL 18e328 pg/mL [3,49]
Stage dependent Prognostic ER e e [1,5,9,10]
PR e e
HER2 e e
BRCA1 e e
Ki67 e e
Osteopontin e e
Mammoglobin e e
Sirtuins e e
Autoantibodies e e
Diagnostic HER2 e e
CEA e e
BRCA1 e e
miR-21, 155, 222 e e
Therapeutic ER e e
PR e e
HER2 e e
CA15-3 e e
CA27.29 e e
Ki67 e e
miR-21 e e
CTC e e

Abbreviations: BRCA1: breast cancer types 1; CA: cancer antigen; CEA: carcinoembryonic antigen; CTC: circulating tumor cells; EGFR: epidermal growth factor receptor;
EpCAM: epithelial cell adhesion molecule; ER: estrogen receptor; HER2: human epidermal growth factor receptor-2; miR: micro RNA; MUC1: mucin 1; PR: progesterone
receptor; VEGF: vascular endothelial growth factor.

discrete fragments of curved carbon planes and exhibits both glassy reactivity, durability, impermeability, chemical inertness, and high
and ceramic properties (Fig. 4, b). It is widely used as electrode electrical conductivity [60]. Moreover, GCEs can be used in various
material in electrochemistry owing to its low cost, broad anodic high-aspect-ratio instruments due to their compatibility with the
potential window, magnified temperature resistance, low 3-dimensional (3D) microfabrication and easy surface

Fig. 3. Origination of biomarkers from tumorigenesis, migration, and accumulation to the different organs together with the relationship between these biomarkers.

5
Table 3

P. Lakhera, V. Chaudhary, A. Jha et al.


Comparative analysis of commonly electrochemical techniques.

Technique Principle Most usage Advantage Limitation Sensitivity Ref

Voltammetry The current is measured at various Diagnosis of various cancer biomarkers Highly sensitive, inexpensive, and Substances having fM- nM [48,50]
forms of potential such as potential such as Osteopontin, IL-10, HER2, PSA easily miniaturized electrochemical activity can be
sweep, potential waveform, step CA153, and tumor cells like HEp-2, measured, and interference in
potential, potential ramp, etc. is applied HT29, HCT sensitivity due to non-Faradaic
to the working electrodes. current
CV: Investigation of the electrochemical
behavior of electroactive material, and
measures the current with ramp-off
potential.
DPV: Measurement of current between
two points of each pulse concerning
potential which is applied in the pulse
form. The height of the DPV curve is
directly proportional to the analyte
concentration.
LSV: The current is measured by the
varied potential at a constant rate
through scanning.
SWV: The current is measured by the
difference between forward and
reversed current
Amperometry Current is measured resulting from the Detection of serum glucose, cancer Inexpensive, fast reproducible, Require redox elements to ng/mL- pg/mL [50,52]
oxidation or reduction of electroactive biomarkers, cancer cells, nucleic acid, reusable, suitable for enzymes' improve the current
material at a constant potential pesticides, and food placement, and appropriate for mass production. Poor resolution due
production even without calibration to matrix effect.
Impedimetry (EIS) The measurement of the current Detection of cancer biomarkers (nucleic Cost-effective, higher signal-to-noise Requirement of the bulky mg/mL- pg/mL [50e52]
6

response to the application of a acid, and protein biomarkers), cancer ratio, label-free detection, fast response, readout device
constant AC potential. cells, DNA hybridization, direct low excitation voltage, insensitive to
observation of antigen-antibody environmental conditions, real-time,
reaction, and enzyme reaction, and on-site detection
Conductometry The change in conductivity/current Detection of biological receptors, Inexpensive, insensitive to light, no Poor selectivity mg/mL- pg/mL [43,51]
flow of a solution is measured due to enzymatic reactions, environmental requirement of the reference electrode,
the change in ionic species pollutants, drugs, and food-borne reusable, easy miniaturization, and less
concentration. This can be attributed to pathogens power consumption due to voltage
the concentration of analytes, reduction
Biorecognition elements, ion
concentration, substrate to product
conversion, etc.
Potentiometry Measures the potential difference Detection of cancer cells, and enzymatic Inexpensive, simple, fast reproducible, Sensitive to environment and Up to pg/mL [50e52]
between working and reference activity reusable, and suitable for enzymes' temperature
electrodes. placement

Abbreviations: CV: cyclic voltammetry; CA153: cancer antigen153; DPV: differential pulse voltammetry; EIS: electrochemical impedance spectroscopy; HER2: Human epidermal growth factor receptor-2; IL: interleukin; LSV:

Materials Today Chemistry 26 (2022) 101129


linear sweep voltammetry; PSA: prostate-specific antigen; SWV: square wave voltammetry.
Table 4

P. Lakhera, V. Chaudhary, A. Jha et al.


Commercial screen printed and glassy carbon electrodes in different configurations [38,74].

Sr. No Company name Electrode type Functionalization Image Dimension/specification Company link

1 Metrohm Available in different Carbon/gold/platinum, silver/ 3.4 x 1.0  0.05 cm. Reference www.dropsens.com/en
DropSens types (2, 3, carbon nanotubes electrode and electric contacts
multielectrode) made of silver

2 Pine Research Instrumentation 3 electrode system Carbon, ceramic/gold platinum Carbon working electrode and www.pineinst.com/echem/
counter electrode, and a silver/
silver chloride reference
electrode. 2/4/5 mm working
electrode

3 BVT technologies 3/8 electrode system Pure gold 38.10 mmx 7.26 mm x http://www.bvt.cz/
pure platinum, pure silver, 0.63 mm.
graphite, carbonreference 8 electrode system has 1
electrode common reference electrode
material
Ag/AgCl/Ag covered by AgCl
4 Rusens Ltd 3 electrode system Carbon paste/silver 10 mm  28 mm x 0.35 mm. http://www.rusens.com/
Polyethylene terephthalate.
Resistance: 10e30 U.

5 Gamry Instruments 3 electrode system Carbon/platinum 1 mm thick, 5 cm long https://www.gamry.com/cells-and-


7

1 cm wide, Alumina Substrate, accessories/electrodes/screen-


working electrode ¼ 12 mm2 printed-electrodes/
Area

6 Gwent Group 3 electrode system Ceramic carbon/platinum/gold Substrate 96% Alumina, 10.12 x www.gwent.org/gem_standard_
51.00  0.50 mm Pitch of electrodes.html
individual electrodes -
2.54 mm, working electrode
-2mm
7 Palm Sense 3 electrode system Platinum/gold/carbon/silver/ Alumina substrate,working https://www.palmsens.com/
silver chloride electrode product-select/
- 1 mm
Silver as reference electrode

Materials Today Chemistry 26 (2022) 101129


material
8 Zimmer and peacock 3 electrode system Gold/carbon/graphene 30mm  10mm  0.635 mm, www.zimmerpeacocktech.com/
working electrode: 2.0884 cm, 2019/02/10/manufacturer-of-
reference electrode: 2.1019 cm, screen-printed-electrodes
counter electrode ¼ 2.1049 cm

9 Zensor 3 electrode system Graphite/gold/graphene/ Diameter of working www.nanochemazone.com/


carbon nanotube/platinum/ electrode:2 mm, 3 mm, 4 mm, product/zensor-screen-printed-
silver (customization possible) 5 mm,reference electrode electrodes
¼ 0.5  1 mm,

(continued on next page)


Table 4 (continued )

P. Lakhera, V. Chaudhary, A. Jha et al.


Sr. No Company name Electrode type Functionalization Image Dimension/specification Company link

10 Basi Research Products 1 electrode system Electrode/gold/carbon paste 7.5 cm length x 6 mm https://www.basinc.com/products/
ec/sve

11 ALS Co. ltd 1 electrode system Customized Glassy carbon electrode/Pt/Au/ https://www.als-japan.com/1408.
10mm/6mm/3mm/4 mm html

12 Pine Research 3 electrode system Customized Glassy carbon electrode 3.0 mm https://pineresearch.com/shop/
disk. Available with Ag/AgCl products/cells-and-glassware/
reference electrode, a platinum standard-volume/web92-gc/
counter electrode, and a set of
polytetrafluoroethylene
stoppers

13 Ossila enabling Material 1 electrode system Customized 2/3/4 mm diameter https://www.ossila.com/products/


Science platinum/gold/graphite working-electrode

14 Wuhan Corrtest Instruments 1 electrode system Non-customized Glassy carbon and www.corrtest.en.made-in-china.
Corp., Ltd polytetrafluoroethylene com
8

3mm/2mm/1mm/4mm/5mm/
6 mm
Teflon diameter- 6 mm
Brass rod- 2 mm

15 Metrohm 1 electrode system Non-customized Glassy carbon, length ¼ 76 mm, https://www.metrohm.com/en-in/


diameter ¼ 2 products/61248040

16 Corrtest Instruments Corp. 1 electrode system Customized Glassy carbon 2mm/3mm/ http://www.csechem.com/
4mm/5mm/6 mm, Teflon electrochemical-accessories/
sleeve ¼ 6mm/8mm/10mm/

Materials Today Chemistry 26 (2022) 101129


working-electrode/glassy-carbon-
12 mm working-electrode.html
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Fig. 4. Conventional electrodes used for electrochemical biosensing. a) screen-printed electrode; b) glassy carbon electrode.

functionalization with various NMs without compromising their The sensitivity, specificity, and EC efficiency of both electrodes
inherent properties [61]. GCE is an intertwining ribbon-like mate- can be enhanced by using the modified electrodes with various
rial that exhibits ordered morphology microscopically with a NMs like graphene (Gr), carbon nanodots (CNDs), quantum dots
mixture of the edge plane and the basal plane of carbon [62]. (QDs), carbon nanotubes (CNTs), nanofibers, metallic/polymeric
However, the electrochemistry of GCE is affected by its cleanliness nanoparticles (NPs), etc [56,64e68]. Further, the analyte-specific
and surface chemistry [63]. Therefore, the surface of GCE should be part of the electrodes can also be modified with various enzymes,
maintained and activated for quick electron transfer kinetics which nucleic acid, aptamer, and antibodies resulting in enzyme-, geno-,
can be carried out by washing, mechanical polishing, plasma apta-, and immunosensors respectively (Fig. 5) [69]. These
treatment, ultrasonication, vacuum heat treatment, and thermal biomolecules can be immobilized on the WEs surface by various
and laser activation [61]. techniques such as microencapsulation, adsorption, crosslinking,

Fig. 5. Fabrication and application of modified screen-printed electrode (SPE), and glassy carbon electrode (GCE) with different doping agents. a) surface modification of both
electrodes with various nanomaterials (1: carbon nanotubes, 2: quantum dots, 3: porous silica, 4: polymers, 5: carbon nanodots, 6: nanofibers, 7: metallic/polymeric nanoparticles,
8: graphene), and analyte-specific part coupled with various biorecognition molecules; b) sample analysis; c) electrochemical measurement of analytes of interest.

9
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

entrapment, or covalent attachment [70]. The modified SPEs with etc.), or redox indicators (ferricyanide/ferrocyanide, quinones/hy-
different agents expedite the electron transfer rate on the electrode droquinones (HQ), anthracyclines, viologens, phenothiazines, qui-
surface with increased output signals that capture the analytes of noxaline derivatives, metal complexes, metal chelates, etc.)
interest [71]. The modified GCEs possess increased surface area resulting in augmented signal intensity, specificity, and improved
[72], corrosion resistance, and high thermal and EC stability [73]. limit of detection (LOD) [185]. The labeling molecule quantifies the
Thus, the modified SPEs and GCEs as WEs in EC biosensors, enable t-DNA/miRNAs in the sample either by direct or competitive
the simple, inexpensive, ultrasensitive, and rapid determination of detection [75]. Direct labeling detection is based on the labeled
various substances. capture hybridized with the targets giving rise to a change in EC
signal response due to the variation in distance between the
5. SPEs and GCEs-based electrochemical biosensors for the labeled molecule and electrode surface in the electroactive species,
detection of breast cancer biomarkers which is proportional to target concentration. The competitive
approach used for the detection of targets is based on the elimi-
BC biomarkers are present at very low concentrations in normal nation of labeled probes from the electrode surface through
cells, but the concentration level increases upon the progression of displacement reactions, or by using various splitting agents like
BC. These biomarkers can be detected at an early stage in saliva, endonuclease, duplex-specific nuclease (DSN), or calcium ions
urine or serum, or stool by using the EC techniques owing to their [186].
simplicity, sensitivity, specificity, convenience, fast detection, and EC genosensor involves direct and indirect/sandwich hybridi-
minimum power consumption [75]. This section summarizes the zation techniques using label-free or labeled reporter probes based
principle and application of various SPEs and GCEs-based (micro- on conformational changes in target DNA (Fig. 6) [35]. To further
electrode/modified electrode) EC biosensors for the detection of explain direct assay encompasses the hybridization reaction be-
diverse BC biomarkers (Table 5). tween the capture probes immobilized on the electrode surface and
target probes and the signal can be collected by amperometry/DPV/
5.1. Detection of genomic biomarkers EIS (Fig. 6a and b). Mousavisani et al. developed a simple, stable,
reproducible, and label-free SPE-based impedimetric DNA
Genomic biomarkers used for the diagnosis of BC employ the biosensor. Initially, a screen-printed carbon electrode (SPCE) was
detection of specific changes in certain genes like amplification, modified with diazonium salt and followed by activation of the
deletion, or mutation of proto-oncogenes, tumor suppressor genes, existing carboxyl group by placing droplets of N-ethyl-N’-(3
cell-cycle regulator genes, etc. Various genetic biomarkers such as dimethylamino propyl) carbodiimide hydrochloride (EDC) and N-
ATM, CYP1A1, CHEK2, FGFR2, RAD1, PTEN, PALB BRIP1 2, TP53, hydroxysuccinimide (NHS). Further, the activated electrode was
TGFB1, MAP3K1, BRCA1, and BRCA2 are used for the diagnosis of BC covered with aminated-DNA and followed by drop-casting of
[48,182]. The antioncogenes BRCA1 and BRCA2 have widely ethanolamine to deactivate the unreacted carboxyl group. The
accepted DNA biomarkers expressed in normal cells and are developed biosensor was evaluated to study the DNA damage by
responsible for DNA repair. These tumor suppressor genes control using EIS. The fabricated biosensor can accurately detect DNA
and regulate the cell cycle and cell division [1,5,10]. Mutations or damage with magnified stability, and reproducibility. Moreover, the
other malfunctions in the BRCA1 genes are responsible for biosensor can also be used to investigate the antioxidant effect of
increased risk of BC (60e80%), ovarian, and prostate cancer and are glutathione in reducing DNA damage [76].
responsible for ~21e40% of inherited BC cases [10,183,184]. More- Sandwich hybridization is the most extensively used technique
over, the specific mutation in BRCA1 5382insC increases the 10- that involves initially binding surface-bound capture DNA (DNA-c)
time risk of BC [22]. The BC can be diagnosed by the genome to the specific part of target DNA (DNA-t; Fig. 6a and b) and fol-
screening of BRCA1 and BRCA2 genes on chromosome numbers 13 lowed by hybridization of a reporter probe (DNA-r; usually labeled
and 17 respectively [48]. with electroactive molecule) to the unhybridized part of DNA (Fig. 6
The principle of EC genomic biosensor (genosensor) involves c) resulting in a sandwich of c-DNA|t-DNA|r-DNA. Further, the hy-
immobilization of sequence-specific probe or capture probe (BRE) bridization reactions into detectable signals can be monitored by
on the electrode surface and followed by the affinity reaction be- using various EC transducers (Fig. 6 d). This method has been
tween the probes and target DNA. Further, the EC transducer explored with diverse modifications at either capturing step or the
transforms the different EC (current, potential, impedance, etc.) or recognition step. In a study, selective biosensing of BRCA1 mutation
non-EC properties (van der Waals interactions, conformal changes, was diagnosed by a carbon dots (CDs) modified screen-printed gold
mass transportation, etc.) that happen during the biorecognition electrode (SPGE) disposable genosensor by using Thionine (Th) as a
process into detectable signals [35]. A typical genosensor comprises redox indicator. The biosensor was developed by firstly drop-
an electrode, capture probe (a molecule immobilized on the elec- casting of CDs on the surface of SPGEs and followed by immobili-
trode surface, and used to recognize and bind to target DNA), re- zation of capture DNA (PROBEBRCA1) on the modified SPGE (PRO-
porter/detector probe (an element that causes the detectable signal BEBRCA1/CDs/SPGE). Further, PROBEBRCA1/CDs/SPGE was hybridized
in response to the EC reaction), and the target DNA/RNA. Single- with the sample of gen BRCA1 comprising wild (WTBRCA1), or
stranded DNA (ss DNA; linear or hairpin), aptamer, RNA se- mutated (MUTBRCA1) types leading to the form WTBRCA1/PRO-
quences, peptides, and DNA-related proteins are commonly used as BEBRCA1/CDs/SPGE, and MUTBRCA1/PROBEBRCA1/CDs/SPGE, respec-
capture or reporter probes (Fig. 6) [35]. tively. After the hybridization, Th was accumulated on the electrode
It is a prerequisite to designing high specificity capture/target surface leading to the recognition of the target single-stranded DNA
DNA for these types of sensors [75]. Other components like inter- (DNAss) using DPV. The results revealed that the developed
mediate linkers and electrode coatings are also used to magnify the biosensor was efficient to discriminate WTBRCA1 and MUTBRCA1 with
genosensor efficiency. Moreover, some sensor encompasses the a detection limit of 55.0 pg/mL [77]. Recently, Feng et al. developed
integrated form of capture and reporter probes as a single unit an SPE-based genosensor by using a tetrahedral-structured probe
resulting in magnified sensitivity. Additionally, the probes can also (TSP) and poly-adenine mediated Au NPs for the magnified detec-
be integrated with various NMs and labeled with different elec- tion of BRCA1 by using CV and amperometry. The sensor was
troactive species like specific enzymes (horseradish peroxidase fabricated by firstly electrodepositing the AuNPs on SPEs and
(HRP), glucose oxidase (GOx), beta-lactamase, urease, and urea, further functionalizing with TSP. Secondly, a stable sandwich
10
Table 5

P. Lakhera, V. Chaudhary, A. Jha et al.


Key characteristics of SPE- and GCE-based electrochemical biosensors for the detection of various breast cancer biomarkers.

Biomarkers Electrode Surface functionalization of Method for electrode Advantage of functionalization Detection method Limit of detection Linear range Ref
type electrodes functionalization

Genomic biomarkers
BRCA1 SPCE Diazonium salt Covalent High, and long-term stability EIS e e [76]
SPGE CDs Drop casting Magnified electron transfer DPV 55.0 pg/mL - [77]
SPGE Au NPs Electrodeposition Fast electron conduction, easy to CV 0.1 fM 1 fM-1 nM [78]
label, and biocompatible
GCE Gr Covalent immobilization Good biocompatibility, large surface CV/ 50 aM 100 aM-1 nM [79]
area to volume ratio, high thermal, Chronoamperometry
conductivity, and electron mobility at
room temperature
GCE PEG/Au-NPs Drop coating Easy fabrication of biosensor along EIS 1.72 fM 50.0 fM-1.0 nM [80]
with, magnified sensitivity,
reproducibility, and antifouling
property
GCE PANI/PEG Galvanostatic technique Large surface area along with DPV 0.0038 pM 0.01 pM-1 nM [81]
magnified antifouling, and
conductivity
GCE PEDOT Electrodeposition Excellent conductivity, DPV 0.03 fM 1.0  1016M-1.0  1010M [82]
biocompatibility, stability, and
antifouling property
GCE GO/P3CA Amide bond Excellent conductivity, high surface CV, DPV, and EIS 3 fM 10 fM-0.1 mM [83]
area enabling more DNA
immobilization
GCE P[DA-b-CD/CTAB])-AgNPs Electrodeposition Excellent conductivity, and more DPV, and SWV 0.003 pg/mL 0.01565e10 pg/mL and [84]
immobilization of primary antibodies 0.625e20 pg/mL
due to large surface area respectively
3.01  1016 M 1.0  1015-1.0  107 M
11

GCE 3D-rGO/PANI Drop coating Reduced charge transfer resistance, CV, DPV, and EIS [85]
and magnified EC activity
15 15 10
GCE MWCNT/MOF@Fe3O4 NPs Drop coating Excellent conductivity, DPV 0.57  10 M 1  10 - 1  10 M [86]
electrocatalytic properties, and
magnified surface area
Transcriptomic biomarkers
miRNA-21 and SPdCE e e e Amperometry 0.6 nM 2.0e10.0 nM [87]
miRNA-205
miRNA-34a SPE CNF e high electrocatalytic effect and high DPV 10.98 mg/mL 25e100 mg/mL [88]
conductivity
miRNA-155 SPGE e e e EIS 5.7 aM 10 aM-1.0 nM [89]
miRNA-155 SPE MBs Electrodeposition Label-free detection of miRNA DPV 29 pmol/L 81 pmol/L-1.2 nmol/L [90]
miRNA-21 SPCE MBs functionalized with His-Tag- e Label-free detection of miRNA Amperometry 0.91 nM 3.0e100 nM [91]
ZFP
miRNA-21 SPCE Au-NPs/rGO Electrodeposition Increased surface area for loading, DPV 5 fM 10 fM-2 pM [92]
efficient electrical conductivity,

Materials Today Chemistry 26 (2022) 101129


biocompatibility, and easy
functionalization
miRNA-21 SPCE Gr/PPy/Au-NPs Drop casting Magnified distribution of DNA probe DPV 0.020 fM 1.0 fM-1.0 nM [93]
on the sensing surface leads to
improve the sensitivity
miRNA-155, miRNA- SPCE Au-NPs/Gr/QDs/GO Drop casting Improved electron transfer ability, CV, SWV, and EIS 0.33, 0.04, and 0.28 fM 0.001e1000 pM [94]
21, and miRNA- high surface area, and easy
210 immobilization of biomolecule
enabling improved analytical
performance
miRNA-21 GCE MWCNT Drop casting Excellent electrocatalytic and DPV 84.3 fM 0.1e500.0 pM [95]
conducting properties
(continued on next page)
Table 5 (continued )

P. Lakhera, V. Chaudhary, A. Jha et al.


Biomarkers Electrode Surface functionalization of Method for electrode Advantage of functionalization Detection method Limit of detection Linear range Ref
type electrodes functionalization

miR-199a-5p GCE GNRs/GO Drop casting The composite provides enhanced EIS 4.5 fM 15 fM- 148 pM [96]
resistance to nucleases and improved
thermostability
MiRNA-24 GCE Polyamidoamine dendrimer Covalent grafting Short time, simple operation, and EIS 3.4 fM 1014M-108M [97]
functionalized PPy immobilized large quantity of DNA
miRNA-155 GCE GNRs/GO sheets Drop casting Increased electrical and thermal DPV 0.6 fM 2.0 fM-8.0 pM [98]
conductivity along with improved
mechanical flexibility and
biocompatibility
miRNA-21 GCE ZrO2 - rGO Drop casting Increased electrical and thermal EIS 4.3 fM 10 fM-100 pM [99]
conductivity along with improved
mechanical flexibility and
biocompatibility
miRNA-21 GCE MBs Drop casting Easily discriminate unwanted EIS 60 aM 0.5e40 fM [100]
constituents
miRNA-21 GCE Au-NPs/CNNS Drop casting Improved electron transfer ability, SWV 2.9 fM 10 fM-1 nM [101]
high surface area, and easy
immobilization of biomolecule
enabling improved analytical
performance
Proteomic biomarkers
Immunosensors
HER2 SPCE Au-NPs Physical adsorption Facilitation of rapid electron transfer EIS 0.01 ng/mL 0.01e100 ng/mL [102]
and providing a biocompatible
surface for the immobilization of
antibody fragments enabling
12

enhanced antigen-binding efficiency


SPCE Zwitterionic hydrogel Drop casting Antifouling properties EIS 77 fM e [103]
SPCE Au Drop casting Biocompatible, non-toxic, chemically CV 34.9 pg/mL 0.001e0.5 ng/mL [104]
inert, improved electrical
conductivity, large surface area,
oxygen transferability, and minor
swelling
SPE e e e CV 4 ng/mL 5 ng/mL-20 ng/mL [105]
SPCE e e e SWV 0.28 ng/mL 1e100 ng/mL [106]
SPCE e e e LSV 2.8 ng/mL 5e50 ng/mL, and 50 [107]
e100 ng/mL
SPCE e e e DPASV 0.29 ng/mL 0.50e50 ng/mL [108]
SPCE e e e DPASV 2.1 ng/mL 10e150 ng/mL [109]
GCE e e e DPV 2.0  105 ng/mL 5.0  104-50.0 ng/mL [110]
GCE Fe3O4@ TMU-21 encapsulated in Drop casting Enhanced electrocatalytic activity, EIS and CV 0.3 pg/mL 1.0 pg/mL-100 ng/mL [111]
MOF and MWCNT large surface area, and porosity

Materials Today Chemistry 26 (2022) 101129


GCE SNGQDs@AuNPs, CoPeBNF, and CoP Drop casting Enhanced electrocatalytic activity, EIS 0.0327 ng/mL, e [112]
eBNF/SNGQDs@AuNPs large surface area, and porosity 0.0454 ng/mL, and
0.1072 ng/mL
GCE WO3/poly-glutamic acid Electrochemical grafting Biocompatible DPV 1 fg/mL 1 ng/mL-1 fg/mL [113]
CEA SPE AuNPs/rGO Electrochemical deposition Enhanced electron transfer, large CV 0.28 ng/mL and 0.5e50 ng/mL, and 250 [114]
surface area 181.5 ng/mL e2000 ng/mL
SPCE Au NPs Elecrodeposition Efficient electron transfer, enhanced CV, EIS 0.05 ng/mL and 0.03 ng/ 1e10 ng/mL, and 0.5e7 ng/ [115]
conductivity, easy functionalization, mL, and 0.01 ng/mL mL
and good biocompatibility
SPE Ag NPs/rGO Elecrodeposition Efficient electron transfer, enhanced CV 0.035 mg/mL 0.05e0.50 mg/mL [116]
conductivity, easy functionalization,
and good biocompatibility
P. Lakhera, V. Chaudhary, A. Jha et al.
SPE rGO-TEPA/Au-MNPs Electrodeposition Magnified electrical conductivity, SWV 1.42 pg/mL 5.0 pg/mL-200.0 ng/mL [117]
improved stability, and augmented
biorecognition efficiency
GCE APTES crosslinked Gr sheets Drop casting Biocompatible, efficient electron EIS 0.2 pg/mL 0.001e20 ng/mL [118]
transfer, improved thermal
conductivity and mechanical
stiffness, and better stability
GCE (SnO2)/rGO and AuNPs Drop casting Large specific surface area, improved EIS 0.17 pg/mL 0.5 pg/mL - 25 ng/mL [119]
electrical conductibility resulting in
magnified loading of antibodies
GCE PdeIr NPs Drop casting Efficient electron transfer, enhanced EIS 0.017 ng 0.05e50 ng/mL [120]
conductivity, easy functionalization,
and good biocompatibility
GCE PDA-Pb2þ/chitosan-Au NPs Drop casting Excellent electrical conductivity, SWV 0.26 fg/mL 1 fg/mL-100 ng/mL [121]
remarkable adsorption capacity
GCE rGO-Au NPs Drop casting Excellent conductivity and large SWV 5.3 pg/mL 50 pg/mL- 650 pg/mL [122]
surface area
MGCE Ag NPs Drop casting Excellent elecroconductivity DPV 0.03 pg/mL 0.0001e20 ng/mL [123]
GCE AueAg/rGO@PDA Drop casting Dual signal amplification due to CV 0.286 pg/mL 0.001 ng/mL- 80 ng/mL [124]
excellent elecroconductivity
GCE TGO/AuNPs @strp Drop casting Large specific surface area, improved DPV 75 fg/mL 100 fg/mL to 5 pg/mL [125]
electrical conductibility resulting in
magnified loading of antibodies
GCE rGO/chitosan/AuNPs Layer by layer deposition Large specific surface area, improved DPV 0.1471 pg/mL 0.3 ng/mL-30 ng/mL [126]
electrical conductibility resulting in
magnified loading of antibodies
GCE 2D-ReS2 nanosheets Drop casting excellent optical electronic, EIS 0.468 pg/mL 0.0005e10.0 ng/mL [127]
vibrational characteristics, and large
surface area
GCE 3DPt/HGO Drop casting Large specific surface area, improved DPV 0.0006 ng/mL 0.001e150 ng/mL [128]
13

electrical conductibility resulting in


magnified loading of antibodies
GCE rGO Drop casting Excellent electrical conductivity, non- CV, EIS 0.05 ng/mL 0.1e5 ng/mL [129]
toxicity, good biocompatibility, and
high surface area
MUC1 SPCE GO-COOH Drop casting Inherent peroxidase-like activity, DPV 0.04 U/mL 0.1U/mL- 2 U/mL [130]
excellent conductive surface
CA 27-29 GCE Au/MoS2/rGO NCs Drop casting Magnified electrochemical Amperometry 0.08 U/mL 0.1e100 U/mL [131]
conductivity
CA 15-3 SPE PPy nanowire and poly (1,5 Electropolymerization Large surface to volume ratio and EIS, CV 0.02 U/mL 0.05e20 U/mL [132]
diaminonaphthalene). magnified conductivity
SPGE e e e EIS, CV, SWV 0.05 U/mL 0.25e10.00 U/mL [133]
SPCE CuS NPs-rGO Drop casting Augmented electrocatalytic activity DPV 0.3 U/mL 1e150 U/mL [134]
towards oxidation of catechol
SPGE CoS2-Gr-AuNPs Drop casting Augmented electrocatalytic activity DPV 0.03 U/mL 0.1e150 U/mL [135]
towards the oxidation of catechol

Materials Today Chemistry 26 (2022) 101129


along with a large surface area
resulting in enhanced immobilization
of antibodies
SPCE Au-rGO Drop casting Excellent electrical conductivity, non- EIS 0.08 fg/mL 0.1 fg/mL-1 mg/mL [136]
toxicity, good biocompatibility, and
high surface area
SPGE MSA SAM Provide narrow carboxylic-acid- DPV 0.95 U/mL 1.0e1000 U/mL [137]
terminated monolayer
GCE Nitrogen-doped Gr sheets Drop casting Excellent electrical conductivity and DPV 0.017 U/mL 0.1e20 U/mL [138]
large surface area
GCE AuPd NCN Drop casting Excellent electrical conductivity, large Chronoamperometry 0.35 fg/mL 0.001 pg/mL-100 ng/mL [139]
surface area, and improved structural
stability
(continued on next page)
Table 5 (continued )

P. Lakhera, V. Chaudhary, A. Jha et al.


Biomarkers Electrode Surface functionalization of Method for electrode Advantage of functionalization Detection method Limit of detection Linear range Ref
type electrodes functionalization

GCE rGO/PDA and NH2 functionalized Electropolymerization Excellent electrical conductivity and SWV 0.002 U/mL 0.002e125 U/mL [140]
mesoporous Si conjugated by Au NPs large surface area
GCE GAs and Pb-CD Drop casting and Excellent electrical conductivity and DPV, EIS 0.03 mU/mL 0.1 mU/mL-100 U/mL [141]
electropolymerization large surface area
GCE Cysteamine A/Au NSs/GQDs Electrodeposition Large surface area for the enhanced SWV 0.11 U/mL 0.16e125 U/mL [142]
immobilization of CA 15-3
GCE Gr ink conjugated with anti-CA15-3 Drop casting Excellent electrical conductivity DPV 15 U/mL 15e250 U/mL [143]
GCE Au NPs Electrodeposition Efficient electron transfer, enhanced EIS 5.06 mU/mL 10e100 U/mL [144]
conductivity, easy functionalization,
and good biocompatibility
ER SPCE e e e Amperometry 19 pg/mL 63e200 pg/mL [145]
PR SPCE e e e Amperometry 22 pg/mL 73e1500 pg/mL [146]
Aptasensors
HER2 SPE PLL Electrostatic adsorption Provide support for aptamer DPV 3.0 ng/mL 10e60 ng/mL [147]
immobilization
SPGE Thiolated DNA aptamer and MCH SAM Provide a bridge between the surface EIS 172 pg/mL 1 pg/mL-1 mg/mL [12]
irregularities and reduce the
adsorption of non-specific proteins
SPCE Au -NPs Drop casting Provide support for aptamer CV, DPV, and EIS 0.001 ng/mL 0.001e100 ng/mL [148]
immobilization
GCE rGO-chitosan film Drop casting Provide amine groups for aptamer DPV 0.21 ng/mL 0.5e2 ng/mL [149]
binding, stability, and large surface
area
GCE ErGO- SWCNT and AuNPs Drop casting (ErGO) and Better electrocatalytic property and CV, DPV, and EIS 50 fg/mL 0.1 pg/mL-1 ng/mL [32]
electrodeposition (AuNPs) magnified immobilization of
biorecognition molecule
14

CEA GCE (MoSe2)/Gr/Au Drop casting (MoSe2/Gr) and Magnified electron transfer DPV 0.03 pg/mL 0.1 pg/mL-100 ng/mL [150]
electrodeposition (AuNPs)
GCE GO Drop casting Better electrocatalytic property SWV 0.05 pg/mL 0.0001 pg/mL-10 ng/mL [151]
GCE AuNPs @ NH2-functionalized Drop casting Excellent electrical conductivity and EIS 9.8  104 ng/mL 1.0 £ 103-100.0 ng/mL [152]
mesoporous Si film large surface area
GCE GO Drop casting Excellent electrical conductivity and SWV 0.1 pg/mL 0.5 pg/mL- 1 ng/mL [153]
large surface area
GCE HAuCl4 Electrodeposition Large surface area EIS 0.023 pg/mL 0.05 pg/mL- 20 ng/mL [154]
MUC1 SPCE CNT Dip coating Excellent conductivity, EIS 0.02 U/mL 0.1e2 U/mL [155]
electrocatalytic properties, and
magnified surface area
GCE rGO/Au NPs Drop casting Excellent electrochemical CV and EIS 0.25 pM 1 pM-1 mM [156]
performance and support for aptamer
immobilization
GCE Au NPs Electrodeposition Excellent conductivity, SWV 0.33 pM 1.0 pM 10 mM [157]
electrocatalytic properties, and

Materials Today Chemistry 26 (2022) 101129


magnified surface area
ER SPGE e e e DPV 0.001 ng/mL 0.001e1000 pg/mL [158]
PR SPCE GQDseNiO-AuNFs/f-MWCNTs Drop casting Efficient immobilization matrix due DPV 1.86 pM 0.01e1000 nM [159]
to COOH group
VEGF SPCE PANI/CNT Drop casting Large surface area and excellent DPV 0.4 pg/mL 0.5e10.0  106 pg/mL [160]
electrocatalytic properties
GCE BSA/Au NCs Drop casting Easy functionalization, large specific DPV, and EIS 0.32 and 0.48 pM 1e125 pM, and 2.5e250 [161]
surface area, efficient conductivity, pM
and low toxicity
GCE PLL Electropolymerization Provide support for aptamer CV 4.6 pmol/L 6.0e20 pmol/L [162]
immobilization
Circulating tumor cells
EpCAM SPE Si NPs and IL Drop casting Large surface area and enhanced DPV 1 cells/mL 5-10000000 cells/mL [163]
conductivity
P. Lakhera, V. Chaudhary, A. Jha et al.
GCE Au-NPs Drop casting Increased electron conductivity EIS, DPV 50 cells/mL 80- 10000000 cells/mL [164]
SKBR3 SPE rGO Drop casting Biocompatibility, large surface area, DPV 21 cells/mL 500e30000 cells/mL [165]
easy functionalization, and low-cost
processing technology
GCE e e e DPV 1 cell/mL 1-80 cells/mL [166]
MCF7 MGCE rGO/MoS2 Drop casting Large surface area, excellent electron DPV 6 cells/mL 15-45 cells/mL [167]
conductivity, and good
biocompatibility
GCE rGO/AuNPs e Large surface area, excellent electron CV, DPV 27 cells/mL 50-7000 cells/mL [168]
conductivity, and good
biocompatibility
GCE Au nanocages/MWCNT-NH2 Drop casting Large surface area, excellent electron DPV 80 cells/mL 100-1000000 cells/mL [169]
conductivity, and good
biocompatibility
GCE PANI Electrodeposition Excellent electron conductivity and DPV 20 cells/mL 50-1000000 cells/mL [170]
good biocompatibility
GCE TiO2 nanotube-rGO Drop casting Large surface area, excellent electron EIS 40 cells/mL 1000-10000000 cells/mL [171]
conductivity, and good
biocompatibility
GCE rGO-chitosan-Au NPs composite Drop casting Excellent electron conductivity and EIS 4 cells/mL 10-1000000 cells/mL [172]
large surface area
GCE VS2/Au NPs Drop casting Large surface area and good DPV 5 cells/mL 10-100000 cells/mL [173]
mechanical strength
MDA-MB-231 GCE Mannose-C2NH2 Electrografting e CV 10 cells/mL 10- 100000 cells/mL [174]
GCE HA-BSA-AuNPs Drop casting Large surface area, excellent electron EIS 128 cells/mL 200- 300000 cells/mL [175]
conductivity, and good
biocompatibility
Multiple targets
CA 15-3, and HER2- SPdCE AuNPs In-situ electrodeposition Large surface area, excellent electron LSV CA 15-3: 5.0 U/mL; CA 15-3: 0e70 U/mL; [176]
ECD conductivity, and good HER2-ECD: 2.9 ng/mL HER2-ECD: 0e50 ng/mL
15

biocompatibility
TNF, and RANKL SPdCE e e e Amperometry TNF: 3.0 pg/mL TNF: 9.9e1000 pg/mL; [177]
RANKL: 2.6 pg/mL; RANKL: 8.6e1000 pg/mL
MUC1, and MCF-7 GCE e e e SWV MUC1: 0.5 nM; MUC1: 1e500 nM; [178]
MCF-7: 50 cells/mL MCF-7: 50e5000 cells/mL
GCE Poly (glutamic acid)/MWCNT NCs Drop casting Large surface area and durability DPV MUC1: 3 ng/mL; MUC1: 5e200 ng/mL; [179]
MCF-7: 25 cells/mL MCF-7: 100
e10000000 cells/mL
CEA, and CA 15-3 GCE Au NPs/3-D Gr hydrogel e Fast electron transfer and large DPV CEA: 11.2 pg/mL; CEA: 5.0  102-60.0 ng/ [180]
surface area for the effective CA 15-3: 1.3  102 U/ mL;
immobilization of more aptamer mL CA 15-3: 5.0  102 -100.0
U/mL
miRNA21, and CA SPGE MoSe2/GO-NCs Large surface area, excellent electron DPV miRNA-21:1.2 fM; [181]
15-3 conductivity, and good CA 15-3: 0.14 U/mL
biocompatibility

Materials Today Chemistry 26 (2022) 101129


Abbreviations: 2D-ReS2: two-dimension rhenium disulfate; 3D:3-dimensional; 3DPt/HGO:3-dimensional graphene oxide-supported platinum metal nanoparticles; APTES: 3-aminopropyltriethoxysilane; Au NPs@ strp: gold
nanoparticles coated with streptavidin; Au: gold; BRCA1: breast cancer types A1; BSA: bovine serum albumin; CA: cancer antigen; cDNA: complementary DNA; CDs: carbon nanodots; CEA: carcinoembryonic antigen; CNNS:
carbon nitride nanosheets; CNT: carbon nanotube; CoPeBNF: cobalt porphyrin binuclear framework; CTC: circulating tumor cells; CuS: copper sulfide; CoS2: cobalt sulfide; CV: cyclic voltammetry; DNA: deoxyribonucleic acid;
DPASV: differential pulse anodic stripping voltammetry; DPV: differential pulse voltammetry; EIS: electrochemical impedance spectroscopy; ErGO: electrochemical reduced graphene oxide; EpCAM: epithelial cell adhesion
molecule; ER: estrogen receptor; GAs: graphene aerogel; GCE: glassy carbon electrode; GNRs: gold nanorods; GO: graphene oxide; Gr: graphene; HA: hyaluronic acid; HAuCl4: chloroauric acid; HCT: herceptin; HER2: human
epidermal growth factor receptors-2; His-Tag-ZFP: His-Tag-Zinc finger protein; IL: ionic liquid; Ir: iridium; LSV: linear sweep voltammetry; MBs: magnetic beads; MCH: 1-mercapto-6-hexanol; MGCE: magnetic glassy carbon
electrode; MIP: molecularly printed polymer; miRNA: micro-RNA; MOF: metal organic framework; MoS2: molybdenum di sulfide; MoSe2: molybdenum selenide; MSA: mercaptosuccinic acid; MWCNT: multiwalled carbon
nanotube; NCN: nanochain network; NCs: nanocomposites; NH2-GSs: amine functionalized graphene sheets; NPs: nanoparticles; P[DA-b-CD/CTAB]): poly (dopamine-beta cyclodextrine-Cetyl trimethylammonium bromide);
P3CA: pyrrole -3-carboxylic acid; PANI: polyaniline; Pd: palladium; PDA-Pb2þ: polydopamine-Pb2þ; PEDOT: poly(3,4-ethylenedioxythiophene); PEG: polyethylene glycol; PLL: Poly-L-Lysine; Poly A: poly-adenine; PPy: pol-
ypyrrole; PR: progesterone receptor; Pt: platinum; Pb-CD: polymeric b-cyclodextrin; QDs: quantum dots; RANKL: Receptor Activator of Nuclear Factor-kB Ligand; rGO: reduced graphene oxide; SAM: self-assembled monolayer;
Si: silica; SKBR3: SloaneKettering breast cancer; SNGQDs@AuNPs: gold nanoparticles functionalized graphene quantum dots; SnO2: stannic oxide; SPCE: screen printed carbon electrode; SPdCE: dual screen-printed carbon
electrode; SPE: screen printed electrode; SPGE: screen-printed gold electrode; SWCNT: single walled carbon nanotube; SWV: square wave voltammetry; TEPA: tetraethylene pentaamine; TGO: thiolated graphene oxide; TiO2:
titanium di oxide; TNF: Tumor Necrosis Factor-alpha; tPA: Human tissue plasminogen activator; TSP: tetrahedral-structured probe; VEGF: vascular endothelial growth factor; VS2: vanadium disulfide; WO3: tungsten trioxide;
ZrO2: Zirconium dioxide.
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

system was formed by the capture DNA, target DNA, and reporter and magnified accuracy due to the integration of conduction
DNA (labeled with Au NPs) (Fig. 7). The genosensing principle was polymer (PEDOT) and antifouling zwitterionic peptide [82].
based on the EC signals of the redox reaction in the presence of Shahrokhian et al. developed a DNA biosensor by using pyrrole-
3,3ˊ,5,5ˊ-tetramethylbenzidine (TMB). The developed biosensor 3 carboxylic acid-coated reduced graphene oxide (rGO) modified
exhibited high sensitivity and specificity with a linear range (LR) GCE (PP3CA/ERGO/GCE). The EC behavior of the redox probe was
from 1 fM to 1 nM, and a LOD of 0.1 fM. The results revealed that the determined by CV, DPV, and EIS resulting in the quantitative
DNAs were stable on the surface of the biosensor due to the determination of BRCA1 in the LR of 10 fMe0.1 mM with a LOD as
sandwich system resulting in increased specificity against the non- low as 3 fM. The results revealed that the PP3CA/ERGO/GCE
complementary sequence and blank [78]. possessed more active sites in contrast to polymer-coated GCE.
In another study, an ultrasensitive DNA biosensor was devel- Moreover, the fabricated ultrasensitive genosensor exhibited a
oped for the attomolar (aM) detection of BRCA1 by using Gr- perfect differentiation between the complementary, non-
modified GCE (GCE/Gr). In this sensor, DNA-c was initially immo- complementary, and mismatched DNA sequences due to the
bilized on the GCE/Gr surface via p-p interaction and followed by polymeric and Gr coating on the GCEs surface. Additionally, the
hybridization to the specific part of DNA-t resulting in the forma- modified GCE was efficient to determine the trace amount of DNA-t
tion of the GCE/Gr/DNA-c|DNA-t complex. Further, the unhybri- in plasma samples [83]. Hasanzadeh et al. developed an ultrasen-
dized part of this complex was conjugated to the Au-NPs labeled sitive immunosensor for the detection of BRCA1 based on modified
DNA-r (DNA-r-Au-NPs) resulting in a sandwich of GCE/Gr/DNA-c| GCE with poly (dopamine-beta cyclodextrin-Cetyl trimethy-
DNA-t|DNA-r-Au-NPs. The analytical signals originated from the EC lammonium bromide) doped with AgNPs (P[DA-b-CD/CTAB])-
oxidation of Au-NPs in presence of perchloric acid was measured by AgNPs and functionalized mesoporous silica. The modified elec-
CV with a magnified LOD (1 aM). Moreover, the genosensor could trodes exhibited augmented surface area and magnified conduc-
detect up to 50 aM DNA-t even with a low amount of sample (6 mL) tivity leading to an increase in the immobilization of HRP-labeled
[79]. antibody (HRP-Ab2). The developed immunosensor exhibited
Wang et al. fabricated low fouling GCEs-based genosensor for magnified sensitivity and specificity for the detection of BRCA1
the detection of BRCA1 mutations. It was a modified GCEs-based with a LOD of 0.003 pg/mL, and an LR from 0.01 to 10 pg/mL and
impedimetric hybrid genosensor where the electrode surface was 0.62e20 pg/mL by DPV, and SWV respectively [84]. Recently, a
modified by cross-linked hydrated polyethylene glycol (PEG) and label-free EC biosensor based on three-dimensional rGO (3D-rGO)
self-assembly of Au-NPs resulting in PEG/AuNPs/GCEs. The capture and PANI-modified GCEs (3D-rGO-PANI/GCE) was developed for
probe (thiol functionalized oligonucleotides, S1) was covalently the detection of BRCA1. The results revealed that PANI and 3D-rGO
attached to the modified electrode surface resulting in the forma- exhibited a synergistic effect leading to reduce charge transfer
tion of S1/PEG/AuNPs/GCEs. Further, the hybridization experiment resistance and magnified EC activity of modified electrode. EC
was performed by immersing the S1/PEG/AuNPs/GCEs into analysis of BRCA1 was carried out by immobilization of capture
different concentrations of target analytes for 2 h followed by probe (ss DNA) on the surface of the modified electrode, and further
rinsing with phosphate buffer saline to remove the non-specific hybridized with the DNA-t. The EC evaluation of the functionalized
bound target analytes. The interfacial change and quantification electrode was carried out by CV, DPV, and EIS. The fabricated
of BRCA1 biomarkers were carried out by EIS. The results revealed biosensor was highly sensitive (1.0  1015-1.0  107 M), and
that the developed hybrid GCE-based biosensor was highly sensi- selective with a LOD of 3.01  1016 M. Moreover, no significant
tive (LOD of 1.72 fM) and highly effective (LR from 50.0 fM to interference was observed in the biosensor selectivity neither in
1.0 nM) for the detection of BRCA1 due to the surface modification the complex real blood samples nor the simple matrix [85]. Very
of PEG and Au- NPs. Moreover, the developed biosensor was recently, a novel EC sandwich-type genosensor was developed to
biocompatible, sufficient to screen the DNA mismatch without any detect the BRCA1 gene using modified GCE with the metal-organic
complex chemistry, and quantify a broad range of serum bio- framework (MOF) with @ iron oxide (Fe3O4) NPs core, and
markers [80]. Hui et al. developed another, antifouling GCEs-based multiwalled carbon nanotube (MWCNT). The genosensor was
genosensor by grafting pegylated polyaniline fiber (PANI) on the fabricated by the immobilization of the capture probe (cDNA) on
surface of GCEs leading to an increase in the surface area, con- the modified electrode followed by casting the target analyte and
ductivity, and antifouling property. The detection of BRCA1 was further hybridization with the redox-cycling ferrocene labeled
based on the hybridization reaction between the target BRCA1 and reporter label probe (r-Fc-DNA). The target was analyzed by
immobilized capture DNA, using methylene blue (MB) as an EC r-Fc-DNA via the electro-catalytic activity of Ferri/Ferrocyanide
indicator. The DPV results revealed that the reduction current of MB resulting in an enhancement of the r-Fc-DNA oxidation peak cur-
was decreased after the hybridization reaction. The developed rent (Fig. 8). The developed genosensor was stable, reproducible,
biosensor was highly sensitive to the BRCA1 with an LR from 0.01 and ultrasensitive with an LR from 1  1015- 1  1010 M and a
pM 1 nM and LOD of 0.0038 pM. Moreover, the developed LOD of 0.57  1015 M [86].
biosensor could easily quantify BRCA1 in complex human serum
[81]. In another study, Wang et al. fabricated PEDOT (poly (3,4- 5.2. Detection of transcriptomic biomarkers
ethylene dioxythiophene) modified GCEs-based antifouling and
highly sensitive EC biosensor for the detection of BRCA1 gene Messenger RNA (mRNA) transcribed from the DNA is widely used
mutation by using DPV. The biosensing results revealed that the as a biomarker owing to its ability to reveal the functional descrip-
DPV peak current was inversely proportional to the target DNA tion of proteins. mRNAs are single-stranded RNA (ssRNA) structures
concentration. Further, the target DNA analysis was similar to resembling ssDNA where thiamine is replaced by uracil, hence
another affinity biosensor and can detect the sample with a higher having a similar detection principle (Fig. 6). The complete set of RNA
target concentration. However, for the detection at a lower target comprises mRNA, microRNA (miRNA), extracellular RNA (exRNA),
concentration, the biosensor needs to be revived to dissociate the and circulatory RNA (cirRNA) is used for the early detection of
bound targets. The developed biosensor was highly selective and different cancers [5,48]. Among these biomarkers, miRNAs are the
ultrasensitive (LR:1.0  1016M-1.0  1010 M; LOD: 0.03 fM), and most extensively non-invasive biomarkers for the detection of
able to detect BRCA1 in human plasma (5% v/v) with low fouling various diseases such as cardiovascular, diabetes, viral infections,

16
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Fig. 6. Working principle of the electrochemical genomic biosensor by using modified screen-printed electrode (SPE)/glassy carbon electrode (GCE). a) immobilization of capture
probe on the working electrode via functional groups; b) specific hybridization reaction between the capture probe and target probe; c) hybrid target DNA with capture probe and
labeled reporter probe; d) electrochemical transduction of affinity reactions into detectable signals.

rheumatoid, neurological disorders, and human cancers miR-21, miR-27a, miR29b, miR- 29c, miR- 93, miR-150, miR-155,
[1,35,187,188]. miRNAs are non-coding endogenous RNA molecules miR-191, miR-200c, miR-210, miR- 213, miR- 222, and miR- 451.
with a length of approximately 18e25 nucleotides [5,186] that play Similarly, the downregulated miRNAs are Let7a-2, miR- 10b, miR-
an important role in various biological processes like cell prolifera- 100, miR-125b, miR-145, miR-193, miR-205, and miR- 497 [10].
tion, apoptosis, differentiation, metabolism, and tumorigenesis [5]. Among these miRNA, miR-21 is the widely used stable target
Typically, these are present in the range of 200 aM to 20 pM [1,189] biomarker for the early detection of BC owing to its higher sensitivity
but abnormal expression (upregulation or downregulation) is (87.6%), and specificity (87.3%) but possesses certain limitations like
associated with cancer, therefore can be used as a diagnostic presence in other cancers, sequence homology with related RNAs,
biomarker. The miRNAs up-regulated in tumor cells are miR-16, and low serum concentration [10,190]. EC nucleic acid-based

Fig. 7. Development and principle of electrochemical genosensor for the detection of BRCA1 based on carbon dots modified screen-printed gold electrode using thionine as a redox
indicator adapted from Ref. [78], MDPI, 2020.

17
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

biosensors (labeled or label-free) provide effective alternatives to probe with a target analyte and further monitored by DPV using Fc
quantify miRNA and can overcome the limitation of various con- as redox reporter (Fig. 9). This enzyme-free biosensor was able to
ventional strategies like In Situ Hybridization (ISH) microarrays, detect the target analyte with a LOD of 5 fM and an LR from 10 fM-2
Reverse Transcriptase PCR (RT-PCR), and Northern Blotting [22,186]. pM. Moreover, the biosensor can directly detect the t-miRNA-21
Recently, various EC miRNAs biosensing approaches have been re- from the diluted samples from BC patients with 2-month storage
ported by using modified SPEs and GCEs (Table 5). Regarding the [92].
application of SPEs for the detection of miRNAs, Rodríguez et al. Recently, a highly sensitive EC biosensor based on modified SPEs
developed A p19-based disposable amperometric magnetic was fabricated for the early screening of miRNA by using DPV. The
biosensor by using SPCE for the simultaneous detection of miR-21 biosensor was composed of modified SPCE with Gr, polypyrrole
and miR-205. The fabricated biosensor comprises antimiR-21 and (PPy), and Au-NPs leading to ameliorating the electron transfer
antimiR-205 probes, chitin-modified magnetic beads (Chitin-MBs), properties and augmenting the intercalation of MB for signal
the p19 viral protein as a capture probe and immobilized on the dual amplification. The fabricated biosensor was operated by observing
SPCE (SPdCE). In this biosensor, SPdCEs were responsible for the the MB signal response because of the number of hybridization
simultaneous detection of two BC by using amperometric tech- products between miRNA-21 and DNA-21 probes immobilized on
niques with the hydrogen peroxide (H2O2)/HQ system. The devel- the electrode surface. The DPV results revealed that the increase in
oped biosensor was able to detect both the t-miRNAs in less than 2h peak current was directly proportional to the concentration of the
together with magnified sensitivity and selectivity with an LR from target. The fabricated biosensor was reproducible, selective, and
2.0 to 10.0 nM and LOD of 0.6 nM. Further, the application of this highly stable possessing an LR of 1.0 fM to 1.0 nM with a LOD of
approach was analyzed by screening the endogenous levels of both 0.020 fM and can detect the miRNA [93]. In another study, the same
t-miRNAs in total RNA [87]. Erdem et al. developed an indicator-free group fabricated a label-free EC biosensor-based SPCE modified
voltammetric miRNA biosensor by using carbon nanofiber modified with Au-NPs/Gr QDs/GO for the detection of miRNA-155, miRNA-
SPEs (CNF-SPEs) for the detection of miRNA-34a. This biosensor was 21, and miRNA-210. Different redox indicators like MB, anthraqui-
composed of amino linked DNA capture probe that was immobilized none (AQ), and polydopamine (PDA) were used as signal indicators
on the CNF-SPEs. The detection of hybridization between the cap- for anchoring capture probes, which hybridized with the comple-
ture probe and the miRNA-34a target was investigated by DPV. The mentary miRNA-155, miRNA-21, and miRNA-210, respectively.
results revealed that the CNF-modified SPEs were reproducible and Quantification of miRNA-155, miRNA-21, and miRNA-210 was car-
stable with an enhanced guanine signal (18-fold) in contrast to the ried out by measuring decreases in the MB, AQ, and PDA EC signals
conventional electrode owing to the high conductivity and respectively. Further, the three well-separated peaks were moni-
increased electrocatalytic effect of CNF. Moreover, the LOD of t- tored by SWV. The fabricated biosensor was ultrasensitive and can
miRNA was found to be 10.98 mg/mL in the LR of 25e100 mg/mL [88]. easily detect the miRNA-155, miRNA-21, and miRNA-210 simulta-
In another study, Cardoso et al. developed a simple ultrasensi- neously with an LR from 0.001 to 1000 pM and LOD of 0.33, 0.04,
tive EC biosensor for the detection of the attomolar concentration and 0.28 fM respectively [94].
of miRNA-155 in BC. The biosensor was developed by immobili- In the recent past, EC biosensors based on modified GCEs, and
zation of anti-miRNA-155 (capture probe) on the SPGEs followed by label-free/labeled probes played an important role in the detection
blocking the non-specific binding area with mercaptosuccinic acid of miRNA. Rafiee-Pour et al. developed a label-free EC biosensor
(MSA). The hybridization between the capture probe and the t- based on MWCNT-modified GCE using MB as a redox indicator. The
miRNA-155 was analyzed by EIS. The developed biosensor was modified electrode was immobilized with ss-DNA and incubated
simple and can detect the miRNA-155 from 10 aM to 1.0 nM with a with the t-miRNA-21. EIS and CV were used to analyze the hy-
LOD of 5.7 aM in human serum samples. Moreover, this strategy can bridization reaction whereas, DPV was used to measure the MB
simultaneously quantify multiple miRNA-155 in biological fluids oxidation peak current. The results revealed that after hybridization
[89]. In another study, Mohammadi et al. developed a SPEs-based peak current was increased. This strategy can successfully detect
label-free miRNA biosensor for the detection of miRNA-155. The the miRNA in an LR from 0.1 to 500.0 pM with a relatively LOD of
biosensing was based on sandwich hybridization of streptavidin 84.3 fM [95]. In another study, the EC biosensor based on GCE
MBs integrated with a biotinylated capture probe with a portion of modified with GO and gold nanorods (GNRs) was developed for the
the microRNA and monitored by DPV. The developed biosensor was detection of miR-199a-5p in serum. The sensor was developed by
highly sensitive with a LOD of 29 pmol/L and LR from 81 pmol/L to immobilization of the thiolated oligonucleotide probe on the
1.2 nmol/L [90]. modified electrode surface and followed by blocking of unspecific
Povedano et al., reported a simple amperometric biosensor for binding by 6-mercapto-1-hexanol solution. The developed
the screening of miRNA using SPCE. The biosensor comprised an biosensor was highly sensitive and can evaluate a low concentra-
MBs functionalized His-Tag-Zinc finger protein (His-Tag-ZFP) cap- tion of serum miR-199a-5p with an LR from 15 fM to 148 pM and
ture probe that binds preferably to RNA hybrids and a biotinylated LOD of 4.5 fM [96]. Recently, a label-free miRNA biosensor was
synthetic complementary RNA detector probe (b-RNA-Dp). Further, developed by using modified GCE with polyamidoamine dendrimer
StrepeHRP conjugation allowed amperometric detection using the functionalized PPy nanowires using EIS. The fabricated biosensor
H2O2/HQ system upon capturing the magnetic bioconjugates at was ultrasensitive with an LR from 1014M-108M and LOD of
SPCEs. The developed biosensor was sensitive (LOD:0.91 nM; 0.34  1014 M. Moreover, the biosensor exhibited ~3.12-time
LR:3.0e100 nM) and can detect the t-miRNAs within 2 h. Moreover, higher sensitivity in contrast to the PPy-modified biosensor [97].
the biosensor can also analyze miRNA-21 from total RNA extracted Direct labeling detection is based on the labeled capture hy-
from adenocarcinoma and epithelial non-tumorigenic breast cells bridized with the t-miRNAs giving rise to a change in EC signal
without target amplification, concentration beforehand, or reverse response, which is proportional to t-miRNAs concentration [186].
transcription steps [91]. In another study, Zouari et al. fabricated an Azimzadeh et al. developed another miRNA biosensor based on
enzyme-free voltammetric biosensor based on Au-NPs and rGO- modified GCE with GNRs attached to the GO sheet surface. Early
modified SPCE for the detection of miRNA-21. The biosensing diagnosis of BC was carried out by the quantification of miRNA-155
assay was based on the sandwich-type hybridization of a capture by using ss-probe and Oracet blue (OB, as an EC indicator), and
probe (thiolated DNA) and a ferrocene-capped AuNPs conjugated signals were measured by DPV. The developed biosensor was able
with streptavidin (FcAuNPsStrep) labeled biotinylated detector to discriminate between the complementary t-miRNA-155, single-
18
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Fig. 8. Fabrication principle of electrochemical sandwich-type genosensor for BRCA1 based on glassy carbon electrode modified with the metal-organic framework (MOF) with @
iron oxide nanoparticles (Fe3O4 NPs) core, and multiwalled carbon nanotube (MWCNT). Reprinted with permission from Ref. [86].

and three-base mismatch, and non-complementary miRNA along integrated with a catalytic hairpin assembly (CHA) was developed
with high specificity and sensitivity (LR: 2.0 fM-8.0 pM; LOD: for the detection of miRNA-21. The CHA assembly comprised two
0.6 fM). Moreover, the biosensor was stable, reproducible, and hairpin DNA molecules; H1 and H2). H1 modified with the amine
exhibited real plasma sample analysis [98]. group was covalently attached to the modified GCE surface via
Another approach used for the detection of t-miRNAs is based on amine coupling chemistry. It was observed that there was no hy-
the elimination of labeled probes from the electrode surface after the bridization reaction between H1 and H2 in the absence of the
hybridization with the target miRNA via displacement reaction [99] miRNA-21 but in the presence of the target, a miRNA hybridization
or using different splitting agents [100]. Regarding this, an impedi- reaction took place between the target analyte and H2 structure
metric miRNA biosensor based on ZrO2erGO modified GCE resulting opening of the H2 closed structure. Later on, H1 was

Fig. 9. Sandwich-type detection of miRNA based on gold nanoparticles (Au-NPs) and reduced graphene oxide (rGO) modified screen-printed carbon electrode (SPCE) using DPV.
Sandwich hybridization involves thiolated DNA as a capture probe (SH-DNAcp) and ferrocene-capped AuNPs conjugated with streptavidin (FcAuNPsStrep) labeled biotinylated
detector probe. Reprinted with permission from Ref. [92].

19
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

hybridized with the unfolded H2 and t-miRNAs were released due to and progression owing to their relative abundance in comparison to
the displacement reaction. Further, H2 was immobilized onto the DNA and RNAs. There are various proteomic biomarkers associated
electrode surface, and the released t-miRNA initiated another next with BC development like the human epidermal growth factor (HER)/
cycle. The developed biosensor was able to determine the target epidermal growth factor receptor (EGFR) family, carcinoembryonic
analyte in the LR from 10 fM to 100 pM with a LOD of 4.3 fM [99]. In antigens (CEA), mucin 1 (MUC1), cancer or carbohydrate antigens (CA;
another study, Cheng et al. developed an immobilization-free CA15-3, CA27-29, CA125), estrogen receptor (ER), and progesterone
impedimetric biosensor by using MBs modified GCEs based on receptor (PR) [10,48]. Other proteomic biomarkers are vascular
duplex-specific nuclease-assisted target recycling (DSNATR) for the endothelial growth factor (VEGF), epithelial cell adhesion molecule
detection of miRNA-21. The capture probe was not hydrolyzed in the (EpCAM), Cyclin E, human epididymal protein 4 (HEP4), cathepsin D
absence of target RNA owing to the less activity of DSN while forming inflammatory proteins (fibrinogen and soluble intracellular adhesion
DNA-RNA heteroduplex in presence of target RNA. Further, DSN molecule 1), cell-free DNA (cfDNA), urokinase-dependent plasmin-
hydrolyzed the target binding portion of the capture probe resulting ogen activator system (uPA), Thomsen-Friedenreich (TF), plasmin-
in free target RNA to hybridized with another new capture probe and ogen activator inhibitor (PAI), amino acid transporter (ASCT2), folic
instigating the second hydrolysis cycle. The fabricated biosensor was acid (FA), beta protein 1(BP 1), tenascin-C (TN-C), matrix metal-
a promising approach for the early diagnosis of BC along with a LR lopeptidase 9 (MMP-9), murine double minute 2 (MDM-2), serine
from 0.5 to 40 fM and LOD of 60 aM. Moreover, the approach hydroxymethyltransferase 2 (SHMT2), apurinic/apyrimidinic endo-
exhibited very little cross-hybridization between similar miRNAs nuclease 1 (APE-1), Human tissue plasminogen activator (tPA), and a
[100]. Recently, Ma et al. developed an effective and simple EC cluster of differentiation 105 and 146 antigens (CD 105, CD146)
biosensor for the detection of miRNA-21 by using Au-NPs/carbon [1,5,37]. Among the various proteomic biomarkers, HER2, CA 15e3, CA
nitride nanosheets (CNNS) modified GCEs, thiol modified aptamer 27e29, and CA125 are FDA-approved for clinical application [48,191].
capture t-DNA probe (Fig. 10). Moreover, MB integrated on the 3’ of EC sensing of proteomic biomarkers is usually carried out by using
capture t-DNA was utilized as an EC signal. Additionally, the selec- antibodies (immunosensors) or aptamer (aptasensors) or a combi-
tivity and sensitivity of the biosensor were magnified by DSN and nation of both as a BRE. Therefore, this section is further divided ac-
CHA assembly. The results revealed that the SWV signal was strong cording to the BRE.
in the absence of t-RNA, whereas the MB signals became poor in the
presence of t-RNA. The DSN can selectively split capture t-DNA of the
DNA-t-RNA hybrid and liberate the t-RNA repeatedly. The variation 5.3.1. Electrochemical immunosensors for the detection of
in the current peak was related to the concentration of miRNA-21. proteomic biomarkers
The developed biosensor was reproducible and stable with an LR This section summarizes the principle and recent examples of EC
of 10 fM to 1 nM and LOD of 2.9 fM [101]. immunosensors (ECIs) based on SPEs and GCEs coupled with various
NMs for the detection of extensively used proteomic biomarkers.
ECIs are robust analytical tools that detect the concentration of the
5.3. Detection of proteomic biomarkers target analyte in a sample by measuring the signals in response to
specific binding between antibodies and antigens [36,37]. These
The proteomic biomarkers (soluble, glycoproteins, or membrane- sensors are usually constructed via the immobilization of BRE (an-
associated glycoproteins) are widely associated with BC development tibodies or antigens) on the electrode surface and coupled to a

Fig. 10. Development of an electrochemical biosensor for the detection of miRNA-21 based on glassy carbon electrode (GCE) modified with gold nanoparticles (Au-NPs)/carbon
nitride nanosheets (CNNS). Reprinted with permission from Ref. [101].

20
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

physical transducer that measures the change in voltage or currents in a detectable signal which is directly proportional to the con-
resulting from complementary binding species (Fig. 11) [51]. These centration of analyte (Fig. 11 b) [37,51,193]. To remove the unbound
sensors possess the inherent specificity of immunoreactions and components a washing step is mandatory after each incubation
intelligibility of commercially available various physical transducers process. Competitive immunoassays also known as limited reagent
leading to magnifying their sensitivity [192]. Mainly two types of assays (limited amount of antibodies) are used for small antigens
ECIs potentiometry and amperometry are widely used for clinical with one epitope in which sample/unknown analyte and labeled
applications [37]. Recently, impedimetric and capacitive sensors analyte are mixed where both compete with each other for the
have also gained interest owing to their direct application for the same binding site of antibodies. Briefly, the limited number of
determination of antigen-antibody reaction without using separa- capture antibodies are immobilized on the electrode surface and
tion step and other reagents [38]. then incubated with the sample mixture having a known concen-
The ECIs work either with direct (label-free) or indirect (labeled) tration of labeled antigens and sample analyte (Fig. 11 c). Quanti-
sensors. The basic principle of both is the same in which the target fication of sample analyte is carried out by the determination of
analyte (usual antigens) is captured by antibodies and followed by labeled antigens concentration those bind to the antibodies. The
blocking of unreacted surface and biorecognition of the analyte signal intensity generated by the labeled analyte is inversely pro-
(Fig. 11). A label-free ECI can detect the physical/chemical changes portional to the sample analyte concentration [51]. Herein, EC
directly during the complex formation without labeling the anti- detection of major proteomic BC biomarkers will be discussed by
gens and antibodies (Fig. 11 a), whereas a labeled immunosensor using immune or aptasensors based on SPEs and GCEs.
encompasses the labeled antibodies/antigens and measures the
signals generated by the labels (enzymes/electroactive species/ 5.3.1.1. Detection of human epidermal growth factor and epidermal
fluorophores/NPs) (Fig. 11 b, c) [36,38]. Direct immunosensors are growth factor receptor. HER and EGFR are cell transmembrane
simple, cost-effective, and can be used for real-time analysis but glycoprotein receptors belonging to the ErbB family. This family
suffers from non-specificity and poor sensitivity. Therefore, a mainly comprises four receptors: (i) EGFR/HER1; (ii) HER2/neu; (iii)
proper blocking agent (surfactant, bovine serum albumin, casein, HER3; (iv) HER4. These receptors are mainly responsible for auto-
etc.) is required to overcome the limitations [51]. However, indirect phosphorylation of tyrosine, activating tyrosine kinase, and insti-
immunosensors are highly sensitive, reliable, and specific as a gating cell proliferation, and differentiation after binding with its
comparison to direct immunosensors, but also face certain limita- ligands like transforming growth factor (TGFa) and epidermal
tions like labeling molecules may influence the antigen-antibody growth factor [1,48]. Among these receptors, HER2/neu is the most
binding efficiency, and the yield of the antigen-antibody complex widely accepted prognostic BC biomarker that is overexpressed in
labeled with various biomolecule may be variable [36]. Moreover, 20e30% of non-invasive BC and leads to growing BC more aggres-
indirect immunosensors can be divided into non-competitive and sively [10,12]. It comprises different domains such as the extracel-
competitive immunosensors [51]. Non-competitive immuno- lular domain (ECD), a transmembrane domain, and an intracellular
sensors are also known as two-site ῾sandwich assays´ which are tyrosine kinase domain [176]. HER2-ECD is a minimal-invasive and
usually applied for huge antigens that possess more than one clinically relevant biomarker for the diagnosis of BC [194,195]. The
epitope. In this method, the target analyte is sandwiched between normal serum concentration of HER2 is 2e15 ng/mL and 15e75 ng/
the excess amount of primary antibodies (capture antibody mL in HER2-positive BC patients. It has been reported that over-
immobilized on electrode surface), and secondary antibodies (a expression of HER2 is responsible for brain metastasis, chemother-
labeled antibody that binds to antigen-antibody complex) resulting apeutic resistance, and recurrence of the disease in BC. Moreover, its

Fig. 11. Working principle of electrochemical immunosensor for the detection of various proteomic biomarkers by using different immunoassays. a) label-free immunoassay; b)
sandwiched immunoassay; c) competitive immunoassay.

21
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

overexpression is also associated with other different cancers such antibodies (immobilized on SPCE) and biotinylated antibodies
as ovarian, stomach, and lung cancer [3]. labeled with streptavidin-QDs (CdSe@ZnS) as a secondary antibody.
Recently, various EC immune/aptasensors with different trans- In the presence of the target analyte, a sandwich complex was
ducers have been developed for the detection of HER2 and HER2- formed between the primary and secondary antibodies, and the
ECD by using label-free [102e104] or sandwich immunoassays immunoreactions were measured by differential pulse anodic
[105e109]. Sharma et al. developed a low-cost and ultra-sensitive stripping voltammetry (DPASV). The developed immunosensor was
label-free impedimetric immunosensor based on AuNPs/SPCE for effective for the detection of HER2-ECD with an LR from 0.50 to
the quantitative detection of HER-2. The AuNPs modified electrode 50 ng/mL and LOD of 0.29 ng/mL. Additionally, the biosensor was
exhibited fast electron transfer and was biocompatible resulting in able to detect the CTC [108]. In another study, HER2 was also
increased surface for the immobilization of single-chain fragment detected by sandwich-type immunoassay based on bare SPCE. The
variable antibody fragments on the electrode surface. The devel- immunosensor was fabricated by the immobilization of the capture
oped immunosensor was ultrasensitive with an LR from 0.01 to antibody on the electrode surface and followed by blocking the free
100 ng/mL and a LOD of 0.01 ng/mL [102]. Another label-free surface with BSA. The affinity reaction between the capture anti-
impedimetric immunosensor based on zwitterionic hydrogel bodies and target antigens was analyzed using a label (CdSe@ZnS
modified SPCE for the detection of HER2 was developed by Cho- QDs) and further monitored by DPASV (Fig. 12). The developed
cholova et al. The immunosensor was fabricated by the immobili- immunosensor responded linearly to HER2-ECD with an LR from 10
zation of an anti-HER2 antibody on the electrode surface and to 150 ng/mL and a LOD of 2.1 ng/mL [109].
subsequently in situ glycoprofiling of HER2 was carried out by using Bare or modified GCE is also used to detect HER2 by using
lectins. The developed biosensor was able to detect the 5 pg/mL of label-free or sandwich immunoassays. Regarding this, a
target analyte with a LOD of 77 fM [103]. Recently, a label-free sandwich-type immunosensor was developed using the nano-
voltammetric immunosensor was designed by the immobilization magnetic platform using GCE. The nanomagnetic platform was
of pegylated cerium oxide-antiHER2 conjugate on the surface of designed by immobilization of antiHER2 functionalized with
Au-modified SPCE. The results revealed that the complex of HER2 3-aminopropyltrimethoxysilane coated Fe3O4 NPs (antiHER2/
with cerium oxide-anti-HER2 conjugate exhibited electron transfer APTMS-Fe3O4) on a bare GCE surface followed by blocking of the
inhibition along with voltammetric peak current reduction, which non-specific binding site of the electrode with bovine serum al-
was proportional to HER2 concentration. The developed label-free bumin (BSA). Further, the various concentration of target antigen
immunosensor showed consistent results with an LR from 0.001 to (HER2) was placed on the modified electrode and followed by
0.5 ng/mL and a LOD of 34.9 pg/mL [104]. placing the secondary antibody (antiHER2/Hyd@AuNPs-APTMS-
Moreover, sandwich-based immunoassays are also used for the Fe3O4) containing Ag ions as a label conjugate. A linear relationship
efficient detection of HER2 by using labels. Tallapragada et al. between the DPV stripping signal of Ag and the log of HER2 con-
designed an SPE (bare) based immunosensor for the detection of centrations was observed with an LR of 5.0  104 e50.0 ng/mL and
HER2 by using sandwich ELISA. The immunosensor was fabricated a LOD of 2.0  105 ng/mL [110]. In another study, a label-free
by the immobilization of mouse anti-human ErbB2 (primary anti- amperometric immunosensor was developed by using modified
body), followed by the incorporation of recombinant human HER2 GCE with MOF and MWCNT. The biosensor was designed by the
antigen. Biotinylated goat anti-human ErbB2 was used as the sec- immobilization of capture antibody (HER2) on the modified elec-
ondary antibody which was linked to streptavidin-conjugated HRP. trode surface and followed by blocking the non-specific binding
Quantification of the target analyte was carried out by measuring site of the electrode with BSA. The electrocatalytic activity of the
the redox reaction by CV. It was observed that the signal increased immunosensor was based on the reduction of H2O2. The ampero-
linearly (LR: 5 ng/mL-20 ng/mL) with an increase in the HER2 metric results revealed that in the presence of the target biomarker,
concentration and a LOD of 4 ng/mL. Moreover, the developed the decline of H2O2 was decreased due to the formation of AgeAb
immunosensor was highly sensitive and biocompatible with the complexes on the electrode. The immunosensor was able to detect
ELISA [105]. In another study, Haiza Lah et al. developed sandwich the target analyte within a range of 1.0 pg/mL-100 ng/mL and a LOD
ECIs using SPCE immobilized with primary antibody (Ab1), and of 0.3 pg/mL [111]. Recently, three label-free biosensors were
lead sulfide QDs-conjugated secondary HER2-antibody (Ab2-PbS developed by the surface modification of GCE with Au functional-
QDs) for the detection of HER2. The developed immunosensor ized Gr-QD (GQD) (SNGQDs@AuNPs), cobalt porphyrin binuclear
exhibited magnified sensing by SWV with an LR from 1 to 100 ng/ framework (CoPeBNF), and CoPeBNF/SNGQDs@AuNPs for the
mL and a LOD of 0.28 ng/mL due to the stripping signal of lead via detection of HER2. The biosensor was developed by the immobi-
acid dissolution [106]. lization of antibodies on the modified electrode surface and signals
Recently, Freitas et al. developed a high-performance EC mag- were measured by EIS. The developed sensors were stable, repro-
netic immunosensor for the detection of HER2-ECD in human ducible, and exhibited lower LOD of 0.0327 ng/mL, 0.0454 ng/mL
cancer cells by using SPCE. The biosensor was based on the sand- and 0.1072 ng/mL respectively [112]. In another recent study,
wich immunoassay where MBs anchored with COOH (COOH-MBs) Nasrollahpour et al. synthesized an EC nano biosensor for the
were used as a capture antibody and biotinylated antibody labeled detection of HER2 using WO3/poly-glutamic acid-modified GCE.
with streptavidin alkaline phosphatase (SAP) as a detector antibody The developed biosensor was stable, specific, and stable with an LR
(biotinylated Ab/SAP). In the presence of HER2-ECD, a sandwich from 1 fg/mL to 1 ng/mL and a LOD of 1 fg/mL [113].
complex (COOH-MBs|HER2-ECD|biotinylated Ab/SAP) was formed
on the SPCE surface. After the addition of the enzymatic substrate, 5.3.1.2. Detection of carcinoembryonic antigen. CEA is a glycopro-
the immunoreactions were measured by LSV. The developed tein secreted by gastrointestinal cells during embryo development
immunosensor was selective for the detection of the target analyte and before birth. It is usually present in lower concentrations unless
with two LR (5e50 ng/mL and 50e100 ng/mL) and a LOD of 2.8 ng/ certain diseases such as pancreatitis, Crohn's disease, ulcerative
mL. Moreover, the biosensor was able to detect the CTC (SK-BR-3) colitis, inflammatory bowel disease, and various type of cancers
with a LOD of 3 cells/mL [107]. Further, the same group developed (adenocarcinoma, gastric, ovarian, lung, colorectal, breast cancer,
another EC magnetic immunosensor for the detection of HER2- etc.) are present. It is an FDA-approved biomarker for the moni-
ECD. The immunomagnetic biosensor was also based on a sand- toring of colon cancer. Moreover, elevated levels of CEA can also
wich immunoassay where COOH-MBs were used as primary differentiate between the different types of cancers like benign/
22
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

metastatic, and aneuploid/diploid. CEA is more prevalent in ductal Interestingly, Li et al. developed an ultrasensitive sandwich-type
carcinoma in contrast to lobular carcinoma suggesting that CEA is immunosensor for the detection of CEA based on a secondary anti-
an early diagnostic marker [196]. However, the increased level of body labeled with palladium (Pd)/Pt nanocages anchored on the
CEA is not specific to BC since it can be observed in different can- surface of amino-functionalized MWCNT (PdPt nanocages/MWCNT-
cers. Therefore, various biosensors like DPV, EIS, SWV, chro- NH2) as a signal amplifier. The immunosensor was fabricated by the
noamperometry, conductometry, fluorescence resonance energy modification of GCE with 3-aminopropyltriethoxysilane (APTES)
transfer (FRET), magnetic DNA nanoprobes, surface plasmon reso- crosslinked Gr sheets (NH2-GSs) leading to increase sensitivity,
nance (SPR), Au interdigitated capacitor transducer, etc., have been electron transfer, and can easily immobilize a large quantity of
developed for the detection of CEA concentration in BC patients capture antibody (anti-CEA). The resulting PdPt nanocages/MWCNT-
suggesting that CEA may act as a promising diagnostic biomarker NH2 and modified GCE exhibited enhanced electrocatalytic activity
for detection of BC [1,48]. Herein, a recently developed EC immu- toward the reduction of H2O2 (Fig. 13). The fabricated sensor
nosensor based on SPE and GCE for the detection of CEA will be exhibited enhanced sensitivity, stability, and augmented immobili-
discussed (Table 5). zation of secondary antibodies owing to the high surface area of the
Chan et al. developed an immunosensor based on modified SPE catalytic label [118].
with AuNPs/rGO for the detection of CEA. The modified SPE Han et al. developed a sandwich-type non-enzymatic ECI for
exhibited higher surface area, increased stability, and augmented CEA based on a nanocomposite (NC) of stannic oxide (SnO2)/rGO
electron transfer kinetics. Moreover, modified SPE was compatible and AuNPs modified GCE resulting in augmented surface area and
with the sandwich ELISA system leading to enhancing the sensi- magnified electron transfer. Further, the modified GCE was immo-
tivity of the sensor toward the detection of CEA. The sandwich-type bilized with the capture antibody. The capture CEA was labeled
immunosensor was fabricated by the immobilization of the pri- with Pd NPs-vanadium pentoxide (V2O5)/MWCNTs and function-
mary antibody, target antigen, and secondary antibody onto the alized with secondary antibodies. The developed composite
modified electrode surface. The developed immunosensor was easy exhibited nexcellent catalytic activity towards the reduction of
to fabricate, cost-effective, and exhibited double EC responses for H2O2. The developed immunosensor was sensitive for CEA with an
the detection of CEA with an LR from 0.5 to 50 ng/mL and LR from 0.5 pg/mL-25 ng/mL and a LOD of 0.17 pg/mL [119]. A label-
250e2000 ng/mL and LOD of 0.28 ng/mL and 181.5 ng/mL, free ECI for CEA was developed by the immobilization of an anti-
respectively [114]. In another study, CEA was detected by a CEA antibody on the GCE modified with Pd-iridium (PdeIr) bime-
sandwich-type immunosensor based on modified SPCE with tallic NPs. The immunocomplex between the CEA antigen and CEA
cysteamine/Au NPs via electrodeposition. The sensor was fabri- antibody results in the blockage of catalytic activity of bimetallic
cated by covalent coupling of different lectins (concanavalin A, NPs to the reduction of H2O2, and a decrease in the response signal.
wheat germ agglutinin, and Lens culinaris agglutinin) on the The developed immunosensor was simple with an LR from 0.05 to
modified electrode surface and followed by coupling with the 50 ng/mL and a LOD of 0.017 ng/mL [120]. Another novel label-free
target antigen and HRP labeled anti-CEA detection probe. It was ultrasensitive amperometric immunosensor for CEA based on GCE
observed that the immunosensor produced a catalytic current modified with PDA-Pb2þ (redox species) and chitosan-Au NPs
signal in the presence of H2O2 and HQ. The immunosensor was able leading to enhancing the current signal, and immobilization of
to differentiate CEA between healthy and cancer serum samples capture antibody (anti-CEA). The immunosensor was fabricated by
with an LR from 1 ng/mL to 10 ng/mL with LOD of 0.05 ng/mL and the immobilization of capture antibody (anti-CEA) on the modified
0.03 ng/mL using wheat-germ agglutinin and concanavalin A as electrode surface and followed by blocking the unbound electrode
BRE, respectively, and in the range of 0.5 ng/mL to 7 ng/mL with surface with BSA and various concentrations of the target antigen
LOD of 0.01 ng/mL using Lens culinaris agglutinin as BRE [115]. (CEA). The developed immunosensor was simple and ultrasensitive
Lee et al. also developed a sandwich-type disposable immu- with an LR from 1 fg/mL to 100 ng/mL and a LOD of 0.26 fg/mL [121].
nosensor based on modified SPEs with AgNPs/rGO leading to Luo et al. fabricated a sandwich-type immunosensor for the
augmenting the surface area for increased electron transfer rate. detection of CEA using SWCNTs@GQDs composites as a signal
The immunosensor was developed by the immobilization of the amplifier. The composite not only amplifies the signal response but
primary antibody on the modified electrode surface and followed also exhibited magnified electrocatalytic activity, and sensitivity.
by coupling with the target antigen and HPR-labeled secondary Additionally, rGO-Au-NPs are used to modify the GCE surface
antibody. The developed immunosensor was simple, rapid, cost- resulting in increased surface area, remarkable conductivity, and
effective, and able to detect the CEA in the LR from 0.05 to dual amplification. The developed immunosensor exhibited excel-
0.50 mg/mL with a LOD of 0.035 mg/mL [116]. Recently, Cao et al. lent specificity and sensitivity toward the detection of CEA with an
developed a novel immunosensor for the detection of CEA based LR from 50 pg/mL to 650 pg/mL, and a LOD of 5.3 pg/mL [122].
on Au and magnetic NPs (MNPs) anchored with rGO- Wang et al. designed an ultrasensitive sandwich-type immuno-
tetraethylenepentamine (rGO-TEPA/Au-MNPs) as a biosensing sensor based on silver/molybdenum disulfide/iron oxide composite
platform and secondary antibody modified with Au@PtNPs as (Ag/MoS2@Fe3O4) and an analogous ELISA method for CEA detec-
detector probe. In the presence of EC substrate, o-phenylenedi- tion. The composite was used as a label for secondary antibodies
amine (OPD) was oxidized to the 2, 20 -diaminoazobenzene leading to an increase the sensitivity and specificity owing to its
(electroactive material) owing to the catalytic effect of Au@PtNPs. magnetic property. The immunosensor was fabricated by 96 well
The SWV results revealed that the EC signals were proportional to microplates and the detection process was accomplished on mag-
the CEA concentration analyte in the sample. The magnetic bio- netic GCE modified with Ag NPs. The DPV signals revealed that the
sensing platform exhibited magnified electrical conductivity current responses were inversely proportional to the CEA concen-
improved stability due to rGO-TEPA and improved biorecognition tration. The developed immunosensor was stable, reproducible,
efficiency owing to Au NPs in contrast to the MNPs. The sandwich and selective with an LR from 0.0001 to 20 ng/mL and a LOD of
immunocomplex possessed excellent magnetism and could be 0.03 pg/mL [123].
separated and immobilized on the SPE surface. The developed In a study, CEA was also quantified by ECI based on modified GCE
biosensor was reliable, selective, and stable with an LR of 5.0 pg/ with AueAg/rGO@PDA-NC as a dual signal amplification. The
mL-200.0 ng/mL, and a LOD of 1.42 pg/mL [117]. developed sensor was stable sensitive, and specific with an LR from

23
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Fig. 12. A sandwich-type electrochemical immunosensor for the detection of HER2 based on a bare screen-printed electrode and quantum dot as an electroactive label. Recreated
with permission from Ref. [109].

0.001 ng/mL to 80 ng/mL, and a LOD of 0.286 pg/mL [124]. In followed by the introduction of Au NPs coated with streptavidin
another study, Nakhjavani et al. proposed an Au/Ag nanohybrid- (AuNPs@strp) resulting in enhanced immobilization of CEA bio-
based sandwich-type ultrasensitive immunosensor for CEA detec- tinylated monoclonal antibody (BmAB). Further, a sandwich system
tion using various strategies. Initially, GCE was modified with was formed after the coupling of secondary antibodies conjugated
thiolated-graphene oxide (TGO) to magnify the surface area, with streptavidin-coated Ag NP. Finally, HRP, HQ, and H2O2 were

Fig. 13. Fabrication and principle of an ultrasensitive sandwich-type electrochemical immunosensor for the detection of CEA based on 3-aminopropyltriethoxysilane crosslinked
graphene sheets (NH2-GSs) modified glassy carbon electrode. Recreated with permission from Ref. [118].

24
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

applied to amplify the EC signals, electron mediator, and sensing These are some of the most important BC biomarkers but are not
analyte respectively. The EC signals were significantly increased in specific or sensitive tools for cancer diagnosis. These are mainly
the presence of HQ and H2O2. the tailored immunosensor exhibited used to observe a person's response to BC treatment and to monitor
excellent performance in an LR from 100 fg/mL to 5 pg/mL with a BC recurrence subsequent to surgery[44,198]. It can only be used as
LOD of 75 fg/mL [125]. Also, CEA was detected by sandwich-type a biomarker if the cancer cell is producing increased amounts of it.
immunoassay based on rGO/chitosan/AuNPs modified GCE as a The normal serum concentration of CA15-3 level is < 30 U/mL in a
biosensing platform and polythionine-Au (PTheAu) composites as healthy person which increased significantly (>100 U/mL) in 30% of
a signal label. The modified electrode with rGO/chitosan/AuNPs patients with BC or ovarian cancer and could be correlated with
effectively augments electron transfer and magnifies the surface tumor stage [49,199]. Various methods are used to detect the CA15-
area leading to enhance immobilization of the primary antibody. 3 but suffer from certain limitations (Table 1) and can not meet the
The developed biosensor was biocompatible, sensitive, and specific clinical demand for early detection of CA15-3 with ultralow con-
with an LR of 0.3 ng/mL to 30 ng/mL and LOD of 0.1471 pg/mL due to centrations. EC sensors are a promising solution to the conundrums
the high conductivity of PTheAu [126]. associated with the early diagnosis of BC.
Recently, a sandwich-type high-performance photo- For CA 27e29 detection, a label-free ultrasensitive ECI based on
electrochemical immunosensor based on two-dimension rhenium GCE modified with Au/MoS2/rGO nanocomposites was developed.
disulfite nanosheets (2D-ReS2) modified GCE as a biosensing plat- The nanocomposites provided magnified EC sensitivity due to the
form. The sensor was fabricated by immobilization of primary additive effect of AuNPs, MoS2, and rGO. Signal enhancement of the
antibody on the modified electrode and followed by the capture of amperometric detection was assessed by measuring the electro-
target CEA, and secondary antibody labeled with alkaline phos- catalytic current response of H2O2 reduction via the immuno-
phatase. The label was able to catalyze vitamin C magnesium reaction between the CA 27e29 antigen and anti-CA 27e29 on the
phosphate to ascorbic acid that can donate electrons to capture modified electrode surface. The fabricated immunosensor was
holes photoexcited from ReS2 nanosheets leading to generating reproducible, stable, sensitive, and specific for CA 27e29 with an LR
photocurrent signals for CEA detection. The developed immuno- from 0.1 to 100 U/mL and a LOD of 0.08 U/mL [131].
sensor was stable, selective, and specific with an LR from 0.0005 to For CA15-3 detection, Nguyen et al. developed an ECI based on
10.0 ng/mL and a LOD of 0.468 pg/mL [127]. In another recent study, SPCE modified with a bilayer film of PPy nanowire and poly(1,5-
CEA was detected by a sandwich-type ECI based on 3-D GO- diaminonaphthalene). The combination of the bilayer was a
supported Pt-NPs (3DPt/HGO) modified GCE as a sensing plat- promising material for biosensing due to its large surface area. The
form, and a secondary antibody labeled with HRP-functionalized developed immunosensor was sensitive and specific for CA 15-3
Au NPs. The surface modification leads to enhancing the surface with an LR from 0.05 to 20 U/mL and a LOD of 0.02 U/mL [132]. In
area, electrical conductivity, and increased immobilization of pri- another study, Gomes et al. proposed an alternative EC device for
mary antibodies. The developed immunosensor exhibited a linear CA 15-3 diagnosis in liquid biopsies based on the combination of a
relationship in the range of 0.001e150 ng/mL and a LOD of biomimetic film with EC detection. The biomimetic film was ach-
0.0006 ng/mL [128]. Very recently, Jozghorban et al. developed a ieved by electro-polymerization of OPD around a CA 15-3 target
label-free immunosensor based on rGO-modified GCE for the which was previously immobilized on the SPGE surface. Further,
detection of CEA. The EC behavior of immunosensor and immu- the protein target was eliminated by trypsin enabling the vacant
noreactions were analyzed by CV and EIS. The developed sensor sites for rebinding (Fig. 14). Furthermore, EC techniques were used
was able to detect the CEA in a human serum sample with an LR to monitor the rebinding of material with CA15-3. The fabricated
from 0.1 to 5 ng/mL and a LOD of 0.05 ng/mL [129]. device exhibited successful characteristics for CA 15-3 detection
and a promising tool for BC screening in POC applications with an
5.3.1.3. Detection of mucin proteins. Mucin (MUC) proteins are LR from 0.25 to 10.00 U/mL and a LOD of 0.05 U/mL [133].
tandem repeat structures with a high proportion of prolines, Amani et al. reported a label-free EC immunosensor for the
threonines, and serines (known as the PTS domain). Its main role is detection of CA15-3 based on SPCE modified with copper sulfide
in the protection of cells from external infection but also plays a (CuS)-rGO composite. The modified electrode exhibited magnifi-
part in signal transduction, apoptosis, transcriptional, and acety- cent activity towards the catechol oxidation as an EC probe. The
lation of histone proteins [48]. The human MUC family includes 21 developed enzyme-free immunosensor was rapid, reproducible,
members (MUC1-MUC21) which are further subdivided into stable, selective, and sensitive with an LR from 1.0 to 150 U/mL and
secreted (MUC2, MUC5AC, MUC5B, and MUC6) and trans- a LOD of 0.3 U/mL [134]. Another label-free and enzyme-free
membrane proteins (MUC1, MUC4, MUC13, and MUC16) [197]. The immunosensor for the detection of CA 15-3 based on SPCE modi-
elevated level of MUC glycoproteins has been used to diagnose the fied with NC of cobalt sulfide (CoS2)-Gr-AuNPs was developed. In
different types of cancers. Among various MUC, MUC1, and CA the developed immunosensor, AuNPs act as the binding site of
(CA27-29, CA15-3, and CA125) are the major biomarkers used for antibodies, and CoS2-Gr was responsible for magnified electro-
BC diagnosis [48]. There are various techniques used for the catalytic activity toward the catechol oxidation and enhanced sur-
screening of these biomarkers but suffer from certain drawbacks face area leading to improve the immobilization of CA 15-3
(Table 1). ECIs are a promising solution to overcome the limitations antibody. Quantification of the target analyte was based on the
owing to their rapid detection and direct readout of the signals hindrance of the electrocatalytic oxidation of the probe after
generated from antigen-antibody reactions. Rauf et al. fabricated a binding of a specific antibody (anti-CA 15-3) with the CA 15-3 an-
disposable ECI based on SPCE modified with COOH-rich GO (GO- tigen. The developed immunosensor was accurate, stable, and
COOH) for the detection of MUC1 using MB as a redox probe. The specific sensitive for CA 15-3 detection in human serum with an LR
authors explored that surface modification of SPCE enhanced the from 0.1 to 150 U/mL and a LOD of 0.03 U/mL [135]. Recently, a
conductivity of the electrodes leading to magnifying the sensitivity sandwich-type EC immunosensor was developed for CA 15-3 with
of the immunosensor [130]. an LR of 0.1 fg/mL-1 mg/mL and LOD of 0.08 fg/mL. The immuno-
CA 27e29, and CA15e3 are the epitope of polymorphic sensor was based on Au-rGO modified SPCE. The augmented
epithelial transmembrane MUC belonging to the MUC1 family. sensitivity was achieved by using secondary antibodies labeled
These are usually overexpressed in colon cancer, lung cancer, with HRP in the presence of H2O2 (signal magnifier), and HQ
ovarian cancer, bladder cancer, endometrium cancer, and BC [48]. (electron mediator) [136]. In another recent study, a novel, cost-
25
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

effective, and disposable immunosensor was fabricated for CA 15-3 binding sites. Further, the modified electrode was incubated with
detection. The immunosensor was fabricated through the deposi- various concentrations of the standard solution of CA 15-3 antigen,
tion of a self-assembled monolayer (SAM) of MSA on SPGE resulting and subsequently, glucose, acetylacetone (ACAC), N-isopropyl
in a narrow COOH-terminated monolayer. Further, the anti-CA 15-3 acrylamide (NIPAM), and N, N0 -methylene-bisacrylamide (MBA)
was conjugated with the COOH- groups present on the electrode was placed on the electrode surface to instigate radical polymeri-
surface. The fabricated immunosensor was more sensitive and zation. Parallelly Cu-MOF-based immunoprobes were prepared
specific for CA 15-3 detection with an LR from 1.0 to 1000 U/mL and through the covalent conjugation of the secondary antibody and
a LOD of 0.95 U/mL in contrast to CA 19-9, and CA 125 [137]. GOx due to the magnified resistance and augmented catalytic ac-
A label-free EC immunosensor for sensitive detection of CA15-3 tivity of Cu-MOF. In the presence of a target antigen, the immu-
based on GCE modified with nitrogen-doped Gr sheets (N-GS). noprobes were specifically bound to the modified electrode via
Surface modification of GCE was responsible for the augmented immunoreaction between antibody and antigen and triggered the
sensitivity of the immunosensor due to the high conductivity of N- radical polymerization. Firstly, glucose was catalyzed in the pres-
GS. The proposed immunosensor was facile and applicable for the ence GOx leading to generating H2O2, which further reacted with
detection of CA 15-3 with an LR from 0.1 to 20 U/mL, and a LOD of ACAC through Cu-MOF (peroxidase mimics) catalysis to form ACAC
0.017 U/mL [138]. An enzyme-free sandwich-type immunosensor radicals for the polymerization of N-isopropyl acrylamide (NIPAM)
was developed based on GCE modified with AuPd nanochain leading to synthesize a new polymer poly (N-isopropyl acrylamide)
network (AuPd NCN). The modified electrode was further incu- (PNIPAM). The EIS results revealed an increase in resistance due to
bated with a primary antibody (anti-CA 15-3) and subsequently the poor conductivity of PNIPAM and Cu-MOF. Due to the novel
with BSA (blocking agent of a non-specific binding site). Further, signal amplification strategy, the developed immunosensor was
different concentrations of CA15-3 were placed onto the electrode potentially sensitive for the CA 15-3 detection with an LR from 10 to
surface and followed by placing AuPd NCNs-secondary antibody 100 U/mL and a LOD of 5.06 mU/mL [144].
suspension. The detection of CA 15-3 was based on the electro- CA125 also known as MUC 16 is mainly associated with ovarian
catalytic activity of AuPd NCN via the H2O2 reduction. The devel- cancer but also associated with non-Hodgkin's lymphoma, meso-
oped immunosensor was reproducible, stable, and sensitive with thelioma, uterus, cervix, pancreas, liver, colon, breast, and lung
an LR from 0.001 pg/mL-100 ng/mL, and a LOD of 0.35 fg/mL [139]. cancers [56,200,201]. Moreover, various non-pathological condi-
Hasanzadeh et al. proposed a sandwich-type immunosensor based tions like menstruation and pregnancy are also responsible for an
on GCE modified with rGO/PDA and NH2 functionalized meso- increased level of CA125 in a healthy population. Various studies
porous silica conjugated by Au NPs enabling magnified surface area revealed that 90% of females have an increased level of CA125 in the
for primary antibodies immobilization and enhanced conductivity. advanced stage of BC or ovarian cancer. However, 50% of cancerous
Under optimal conditions, the proposed immunosensor exhibited patients have a normal range of CA125. Therefore, it is considered a
magnificent analysis of CA 15-3 with an LR from 0.002 to 125 U/mL valuable biomarker for cancer diagnosis as well as for cancer pro-
and a LOD of 0.002 U/mL [140]. gression monitoring, and treatment [56,198,202,203]. Various SPE-
A novel ultrasensitive immunosensor for the CA 15-3 based on and GCE-based EC biosensors have been developed for CA 125 in
GCE modified with polymeric b-cyclodextrin (Pb-CD) loaded 3-D ovarian cancer but as per our knowledge from 2015 to date, no EC
porous Gr aerogel (GAs), and hexacyanoferrate as an EC probe biosensor based on these two electrodes has been developed for
was developed. The surface modification of the electrode was the monitoring of CA 125 in BC.
responsible for enhanced surface area and outstanding conductiv-
ity resulting in large immobilization of antibodies. The developed 5.3.1.4. Detection of other proteomic biomarkers. ER and PR re-
immunosensor was reproducible, stable, and selective for CA 15-3 ceptors are proteomic biomarkers that belong to the superfamily of
with an LR from 0.1 mU/mL-100 U/mL, and a LOD of 0.03 mU/mL steroid hormone nuclear receptors [204,205]. These receptors have
along with a low working potential (0.2 V) due to the collaborative two types of isoforms ERa and Erb, and PR-A and PR-B, respectively.
effect of Pb-CD, and GAs [141]. Hasanzadeh et al. developed an Clinical and experimental studies demonstrate that these receptors
innovative immunosensor for ultrasensitive detection of CA 15-3 in are very important in tumorigenesis, mammary gland develop-
MCF-7 and human plasma using Au nanospears (NSs) electro- ment, and BC development [6]. Moreover, it has been documented
chemically immobilized onto thiolated GQDs modified GCE was that ER is responsible for ductal morphogenesis whereas PR is for
developed. The hybrid interface (CysA/Au NSs/GQDs/GCE) fur- lobuloalveolar differentiation. However, the PR expression is highly
nished a large surface area for the immobilization of CA 15-3 an- dependent on the presence of ER. Therefore, these two biomarkers
tigens together with their stability. Under optimized conditions, the are used in prognosis and predicting response to hormone treat-
developed immunosensor was stable, specific, and sensitive for CA ment. Eletxigerra et al. developed an amperometric EC magneto-
15-3 with an LR from 0.16 to 125 U/mL and a LOD of 0.11 U/mL due immunosensor for the detection of ERa in human serum and cell
to the increased immobilization of anti-CA 15-3 onto the modified lysate samples to differentiate between ERa positive and negative
electrode [142]. BC cells. The capture antibodies were immobilized onto MBs-COOH
A novel immunosensor based on GCE modified with Gr ink and the captured target was sandwiched with biotinylated detector
conjugated with anti-CA15-3 (bioink) was developed for the antibodies labeled with a streptavidin-HRP conjugate. The EC
detection of CA 15-3. The conductivity of bioink was analyzed by monitoring of the reactions was performed at SPCEs using H2O2 as
plotting various conductive patterns on paper using a rollerball HRP substrate and HQ as electron transfer mediator. The developed
pen, and DPV was used to evaluate the EC behavior of the tailored immunosensor was inexpensive, easy to operate, quick, simple, and
immunosensor. The fabricated immunosensor was sensitive and sensitive with an LR from 63 to 200 pg/mL and a LOD of 19 pg/mL
selective for CA 15-3 assay in unprocessed human plasma samples [145]. In another study, the same group developed an EC magne-
with an LR of 15e250 U/mL and a LOD of 15 U/mL [143]. Zhang et al. toimmunosensor for the detection of PR receptors based on SPCE
proposed an impedimetric immunosensor based on GCE modified using functionalized MBs and specific antibodies. The developed
with Au NPs using a cascade catalysis-initiated radical polymeri- immunosensor was accurate for the detection of PR with an LR from
zation signal amplification strategy. The immunosensor was fabri- 73 to 1500 pg/mL and a LOD of 22 pg/mL [146].
cated by the immobilization of CA 15-3 antibodies on the modified VEGF is an important angiogenesis regulator and a signaling
GCE surface followed by dripping of BSA to block the non-specific protein that stimulates vascular endothelial cell proliferation,
26
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

growth, and survival. In mammals, it possesses five members 4.12% sensitivity as compared with the SAM platform (LOD: 179 pg/
(VEGFA, VEGF-B, VEGF-C, VEGF-D, and placenta growth factor), and mL) [12]. In another recent study, Harahsheh et al. fabricated an EC
different isoforms of VEGFA (VEGF121, VEGF121b, VEGF145, aptasensor based on Au-NPs modified SPCE for the selective
VEGF165, VEGF165b, VEGF189, and VEGF206) [1]. Among all iso- detection of HER2. The developed aptasensor was sensitive, and
forms, VEGF165 is associated with the growth and metastasis of fast with a 5 min binding time and exhibited an LR of 0.001e100 ng/
various cancers. Although VEGF is an important biomarker for BC mL and a detection limit of 52.85 mA/ng/mL, and 0.001 ng/mL,
diagnosis but also associated with various diseases like brain in- respectively [148]. In a study, an EC aptasensor was developed by
juries, Parkinson's, rheumatoid arthritis, psoriasis, and other can- using rGO-chitosan film modified GCE, specific HER2 aptamer, and
cers. The normal serum concentration of VEGF is 1e177 pg/mL, and HER2 proteins to recognize and detect HER2 in human serum. The
it can increase up to 18e328 pg/mL in BC patients [3,49]. Palabiyik developed aptasensor exhibited magnified selectivity and sensi-
et al. reported a sensitive label-free impedimetric immunosensor tivity (LR: 0.5e2 ng/mL; LOD: 0.21 ng/mL) for HER2 due to signal
based on SPCE modified with molecularly imprinted polymer (MIP) amplification of MB and increased charged transfer kinetics of rGO-
for the detection of VEGF. The developed immunosensor was chitosan film [149]. Rostambadi et al. developed an impedimetric
reproducible and sensitive with an LR of 20e200 pg/mL and a LOD aptasensor based on GCE modified with AuNPs placed on single-
of 0.08 pg/mL [206]. walled CNT (SWCNT) for the detection of HER2. The biosensor
was designed by the immobilization of an antiHER2 aptamer and
5.3.2. Electrochemical aptasensors for the detection of proteomic characterized by CV, DPV, and EIS. The developed aptasensor was
biomarkers able to recognize HER2 with a LOD of 50 fg/mL and LR from 0.1 pg/
EC aptasensors (aptamers used as a bioreceptor) based on SPE m to 1 ng/mL [32].
and GCE are extensively used for the detection of BC biomarkers Interestingly, He et al. designed a sensitive EC aptasensor for the
due to their low cost, flexibility, stability, versatility, and excep- detection of CEA based on molybdenum selenide (MoSe2)/Gr/Au-
tional binding affinity. Moreover, EC methods require lesser sample modified GCE. The modified electrode was further conjugated with a
quantity and display the capability of conceivable biosensors with thiol-labeled aptamer against CEA. The developed aptasensor was
adequate stability and low detection limit leading to easy minia- sensitive with an LR from 0.1 pg/mL to 100 ng/mL and a LOD of
turization [207]. Aptamers are small nucleic acid ligands of ssDNA, 0.03 pg/mL [150]. In another study, GCE (modified with graphene
RNA, and peptide molecules screened by systematized exponen- oxide [GO]) based EC aptasensor was developed for the detection of
tially enriched ligands (SELEX) [208]. Aptamers or chemical anti- CEA. The aptasensor was based on the sandwich method with
bodies have been widely used as bioreceptor in various sensing sequential immobilization of anti-CEA antibody, CEA protein, and
applications due to their sensitivity, specificity, cost-effectiveness, GNR-aptamer onto GCE. The developed aptasensor was specific,
easy synthesis, and stabilization [209]. Moreover, aptamers can sensitive with an LR from 0.0001 pg/mL-10 ng/mL and LOD of
also selectively bind to the unique biomarkers that are overex- 0.05 pg/mL [151]. Shekari et al. developed another GCE-based label-
pressed to the tumor cell surfaces. Based on the methods the EC free impedimetric aptasensor for the selective detection of CEA. The
aptasensors are classified into the direct, sandwich-type, aptasensor was developed by the immobilization of an NH2-modi-
displacement-type, and folding-type aptasensors. The direct assay fied CEA aptamer on the surface of GCE modified with AuNPs @
is based on the molecular recognition of the target biomarker using amine-functionalized mesoporous silica film. The developed apta-
the aptamer immobilized sensing interface. The sandwich-type sensor was sensitive and specific for the quantification of CEA with
bioassays are similar to antibodies in immunosensors by using an LR from 1.0  103 to 100.0 ng/mL and a LOD of 9.8  104 ng/mL
the binary aptamer (Fig. 15, a). Further, an aptamer can be com- [152]. An aptasensor based on GO-modified GCE immobilized with
bined with antibodies when secondary aptamer is not available anti-CEA antibodies was developed for the detection of CEA. The
leading to form aptamer-antibody (Fig. 15, b) or an antibody- modified electrode was further incubated in CEA solution and fol-
aptamer (Fig. 15, c) sandwich assay. The displacement assay in- lowed by immobilization of AuNPs-aptamer primer conjugate
volves the competition between the authentic target analyte and resulting in the formation of anti-CEA/CEA/AuNPs-aptamer-primer
labeled target analytes for the specific binding site on the biosensor onto the GCE surface. The developed aptasensor exhibited
surface. The folding-type EC aptasensor is based on the binding of enhanced signal amplification due to the integration of the DNA-
electrode-bound DNA probes to cause folding [210]. Recently, EC generated technique with rolling circle amplification (RCA). The
aptasensors are widely used in various applications like clinical developed aptasensor conjugated with RCA was highly sensitive
diagnosis, environment, and food monitoring. Here in this section, with an LR from 0.5 pg/mL to 1 ng/mL and a LOD of 0.1 pg/mL in
we will summarize the application of EC aptasensors based on contrast to the aptasensor without RCA [153]. Interestingly, Zhou
modified SPEs/GCEs for the detection of various proteomic BC et al. proposed an impedimetric aptasensor for CEA detection based
biomarkers. on a cascade catalysis signal amplification strategy. Initially, Cu-
Bezerra et al. developed an aptasensor based on modified SPE based MOF encapsulated with Pt-NPs was prepared to achieve
with Poly-L-Lysine (PLL) for the detection of HER2 in human serum. Pt@CuMOFs with peroxidase-like activity. Then, G-rich CEA aptamer,
The developed biosensor was reproducible, and stable with an LR hemin, and GOx were immobilized onto the surface of Pt@CuMOFs.
from 10 to 60 ng/mL and a LOD of 3.0 ng/mL. Moreover, the In the presence of hemin, the G-rich CEA aptamer converted to
developed aptasensor was sensitive and promising for the detec- hemin@G-quadruplex (hGq) resulting in the Pt@CuMOFs-hGq-GOx
tion of serum HER2 with albumin consumption leading to the complex (signal transduction probe). The developed complex was
molecular diagnosis of BC [147]. Recently, another SPGE-based DNA further immobilized to the modified GCE interface. After the intro-
aptasensors were developed for the detection of HER2 biomarkers duction of glucose and 3,3-diaminobenzidine (DAB), a cascade re-
by using a mixed SAM of 1-mercapto-6-hexanol (MCH) with thio- action was instigated by GOx to catalyze the oxidation of glucose, in
lated aptamer specific to HER2 and a ternary SAM with the same situ generating H2O2. At the same time, the decomposition of
aptamer and 1,6-hexanediol (HDT). Both biosensors were able to generated H2O2 was enhanced by Pt@CuMOFs and hGq along with
detect the target analyte with an LR from 1 pg/mL-1 mg/mL. The the oxidation of DAB and the production of insoluble precipitates
ternary SAM aptasensor exhibited non-specific attachment reduc- enabling the signal amplification. The developed aptasensor was
tion to the electrode surface owing to the antifouling properties of sensitive with an LR from 0.05 pg/mL to 20 ng/mL and a LOD of
HDT. Additionally, this biosensor exhibited 172 pg/mL as LOD and 0.023 pg/mL [154].
27
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Nawaj et al. prepared an aptasensor for MUC1 using SPCE response to personalized anticancer therapy mechanisms. How-
functionalized with CNTs.The aptasensor was easy to fabricate ever, their detection is challenging due to their lower concentration
because there is no requirement for prior modification of SPCE and has not been detected by the affinity-based approach [5,10].
together with a small number of CNTs [155]. A 3-D Gr-based EC There are various techniques such as optical, EC, acoustic wave,
aptasensor was developed for the selective detection of MUC1. The field-effect transistor, photothermal, and quartz crystal microbal-
aptasensor was fabricated by surface modification of GCE with rGO ance to detect CTCs. Among all techniques, EC cytosensing features
modified Au-NPs and followed by immobilization of reduced magnified sensitivity, rapid response, and easy fabrication. More-
thiolated Fc-aptamer. Further, the developed aptasensor was over, these cytosensors can also be used clinically for POC diagnosis
incubated with BSA to block the unspecific binding site. The due to their miniaturization. The working principle of cytosensors
developed aptasensor was specific and sensitive for MUC1 with an is similar to immunosensors (Fig. 11) and aptasensors (Fig. 15). EC
LR from 1 pM to 1 mM, and a LOD of 0.25 pM [156]. In another study, cytosensors quantify the cells via transforming the electrical output
a competitive EC aptasensor was developed for the detection of through the reaction between the biorecognition probes and CTCs.
MUC1. The aptasensor was developed by using cDNA-ferrocene/ Based on the BRE EC cytosensors are further categorized into
Ti3C2 (MXene) modified GCE. The modified electrode was further antibody-based cytosensors and aptamer-based cytosensors.
immobilized with MUC1 aptamer via the AueS bond. The devel- Antibody-based cytosensors are more selective but possess certain
oped aptasensor was very promising for the clinical diagnosis of BC limitations such as the production of purified antibodies is a time-
with an LR of 1.0 pMe10 mM and a LOD of 0.33 pM [157]. consuming and expensive process, and antibodies are sensitive to
Ahirwar et al. developed an EC aptasensor for the rapid and high temperatures and harsh chemicals. Aptamer-based cyto-
sensitive detection of ERa in human BC. The aptasensor was sensors are advantageous as compared to antibody-based due to
developed by the immobilization of the thiolated aptamer on the their small size, cost-effectiveness, high purity, magnified binding
screen-printed graphite electrodes (SPGE) and the change in signals affinity, low toxicity, augmented stability, easy functionalization,
was measured by DPV. It was observed that the modified electrode simple synthesis without immunogenicity, and reusability [7]. This
exhibited an augmented flow of electrons, which starts to bring section summarizes the SPE-/GCE-based EC cytosensors for the
down when more ERa binds to the aptamer-modified electrode. The diagnosis of CTCs.
developed aptasensor was coated with aptamer [158]. A label-free Epithelial cell adhesion molecule (EpCAM) is a transmembrane
EC aptasensor was developed for the detection of PR (P4) via the glycoprotein and a potent biomarker for BC metastasis. This
covalent immobilization of an amine-functionalized P4-specific biomarker is responsible for cell proliferation, adhesion, and
aptamer on the modified SPCE. SPCE was modified with migration. Clinically EpCAM is detected by the CellSearch method
GQDseNiO-AuNFs/f-MWCNTs to develop an effective immobiliza- which is based on the conjugation of EpCAM-specific antibodies
tion matrix with enough COOH groups. Quantification of PR was with ferrofluid leading to magnetic separation. However, this
carried out by measuring the DPV response with increasing PR method can only detect the high EpCAM-expressing cells. Recently,
concentration. The developed aptasensor exhibited great potential Hashkavayi et al. developed an ultrasensitive, EC aptasensor based
for the detection of PR in a human serum sample with an LR from on GCE modified with thiol functionalized silica NPs and ionic
0.01 to 1000 nM and a LOD of 1.86 pM [159]. liquid for the detection of EpCAM. Further, the modified electrode
Recently, Park et al. developed a highly sensitive EC aptasensor was functionalized with gold nanostar structures (GNSTs) using a
for the detection of VEGF165 based on polyaniline/CNT nano- dual signal amplification strategy. The developed aptasensor was
composite (PANI/CNT) modified SPCE with an anti-VEGF165 RNA stable and sensitive with an LR from 5-107 and LOD of 1 cells/mL
aptamer. The developed aptasensor was sensitive to detect the [163]. In another recent study, Zhang et al. developed an ultra-
cancer biomarker with an LR from 0.5 to 10.0  106 pg/mL and a LOD sensitive multifunctional EC cytosensor for rapid capture, specific
of 0.4 pg/mL [160]. A label-free EC aptasensor for VGEF165 based on detection, and release of the captured EpCAM based on GCE
the ῾signal off´ and ῾signal on´ mechanism using an anti-VEGF165 modified with Au NPs. The cytosensor was developed by the
aptamer immobilized BSA-Au nanoclusters/ionic liquid/GCE. The immobilization of anti EpCAM onto the modified electrode fol-
LR of the developed aptasensor using the ῾signal off´ and ῾signal on´ lowed by BSA incubation to block the non-specific binding sites.
strategies were 1e125 pM and 2.5e250 pM with corresponding The capture efficiency of the developed device was monitored by
LODs of 0.32 and 0.48 pM, respectively [161]. In another study, EIS and DPV. The analytical results revealed that under optimal
detection of VEGF was carried out by using GCE electropolymerized conditions the capture response changed linearly with an LR from
with PLL. Further, the modified electrode was functionalized with L 8.0  10 e1  107 cells/mL, and a LOD of 50 cells/mL. After the
amino-aptamer 13 solutions followed by L aptamer12- capture of EpCAM glycine HCl was used to destroy the antigen-
functionalized Ag/Pt bimetallic nanoclusters. The developed label- antibody complex to liberate the captured cells without disturb-
free aptasensor was simple, cost-effective, highly specific for VEGF, ing their viability. The developed cytosensor was reproducible,
and able to detect VEGF concentration in human serum of BC pa- accurate, and stable with a fine recovery [164].
tients with an LR from 6.0 to 20 pmol/L and LOD of 4.6 pmol/L [162]. SloaneKettering breast cancer (SKBR3) is one of the important
BC cells that possess the HER2 antigen on its plasma membrane.
5.4. Detection of circulating tumor cells These cells have been used to overcome Herceptin (HCT) treatment
resistance to HER2-overexpressing BC. Tabrizi et al. developed a
Circulating tumor cells (CTCs) are primary cancer biomarkers sandwich-type cytosensor for SKBR3 using rGO modified SPE as a
that play a pivotal role in the early diagnosis and prognosis of platform for the immobilization of primary HCT antibody (Anti-
cancer and are an important indicator of tumor metastasis. These HCT). In this study, various rGO-tetrasodium 1, 3, 6, 8-pyrene-tetra
cells (single or cluster) are liberated from the tumors and circulated sulfonic acid/metal hexacyanoferrates (rGO-TPA/MHCFnano)
in the blood at low concentrations of up to 10 cells/mL, as compared nanocomposites (rGO-TPA/FeHCF, rGO-TPA/NiHCF, rGO-TPA/CoHCF,
with erythrocytes (109/mL), and leucocytes (106/mL) in total blood. and rGO-TPA/CuHCF) were used as a signal tag for secondary anti-
Based on the size, morphological, and biochemical aspects CTCs are HCT. The results revealed that the cytosensor using rGO-TPA/
significantly different from normal cells leading to the easy detec- FeHCF-labeled secondary anti-HCT exhibited higher sensitivity
tion of cancer and its. Moreover, these cells also predict the nature (30000 cells/mL) in contrast to the other rGO-TPA/MHCF nano--
of cancer and could be used to recognize the chemotherapeutics labeled secondary anti-HCT antibodies due to enhanced
28
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Fig. 14. Detection of CA 15-3 by electropolymerization of o-phenylenediamine on a screen-printed gold electrode (SPGE). Reproduced from Ref. [133] PLOS one, 2018.

immobilization of primary and secondary anti-HCT. The proposed (Fe3O4-NPs conjugated with aptamer/maleimide-polyethylenei-
cytosensor was stable, selective, and sensitive for SKBR3 with a LOD mine) onto the modified electrode surface and followed by the
of 21 cells/mL [165]. Recently, a rapid immunosensor was introduction of the sample. The rGO/MoS2 composite and immu-
developed for the detection of SKBR3 based on GCE modified with nomagnetic beads enhanced the signal via a synergistic catalysis
HCT-conjugated Gr with specific anti-HER2. The developed effect. Moreover, both were utilized as EC signal indicators and
biosensor was simple, inexpensive, and sensitive for SKBR3 even at enhancers without supplementary redox mediators. Further, the
lower concentrations (1 cell), which can be used for real sample developed cytosensor could be revitalized by simply withdrawing
analysis [166]. the magnet. The developed cytosensor was inexpensive, versatile,
MCF-7 is a typical BC cell line with progesterone, estrogen, and efficient, reproducible, and sensitive with an LR of 15e45 cells/mL
glucocorticoid receptors that are used for in vitro BC studies. and a LOD of 6 cells/mL [167]. Another EC cytosensor was devel-
Regarding this, Tian et al. developed an ultrasensitive reusable oped for the rapid detection of MCF-7 using GCE modified with
cytosensor based on magnetic GCE (MGCE) modified with rGO/ rGO/AuNPs composites as a support material and CuO nanozyme as
MoS2 composite enabling increased surface area, rapid electron a signal amplifier. rGO/AuNPs provide enhanced surface area for
transfer, and satisfactory biocompatibility. MGCE is advantageous rapid electron transport, biocompatibility, and stability whereas
for the separation of CTCs labeled with magnetic materials due to CuO nanozyme was responsible for the current signal magnifica-
the presence of a powerful magnetic field. The cytosensor was tion enabling ultrasensitive detection of MCF-7. The developed
fabricated by the immobilization of immunomagnetic beads cytosensor detected MCF-7 through the surface recognition

Fig. 15. Sandwich assays generated by combining aptamer and antibodies. a) binary aptamer sandwich-type; b) antibody-aptamer sandwich-type; c) aptamer-antibody sandwich-
type.

29
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

between specific MUC-1 (over-expressed on the MCF-7 cell mem- cytosensor for specific detection of MDA-MB-231 through the re-
branes) and MUC1 aptamer. Under the optimized conditions, the action between the mannosyl GCE (mannose-C2NH2, mannose-
developed cytosensor was reproducible and selective with an LR C6NH2, and galactose-C2NH2) and the overexpressed mannose re-
from 50e7  103 cells/mL and a LOD of 27 cells/mL. Moreover, the ceptors on the MDA-MB-231 cell surface. The fluorescent micro-
cytosensor can easily detect the MCF-7 from the serum sample due scopy revealed that the modified electrode was biocompatible and
to the peculiar combination of MUC1 and MUC1 aptamer [168]. specific to target cells. It was observed that the GCE modified with
Another, GCE-based label-free and sandwich-type EC biosensor mannose-C2NH2 exhibited magnified sensitivity in contrast to
were developed for the sensitive detection of MCF-7 cells through mannose-C6NH2 whereas, galactose-C2NH2 modified electrode was
DPV. The biosensor was developed by the surface modification of not able to detect the target cells. The developed cytosensor was
GCE with a composite of 3D Gr and Au nanocages/MWCNT-NH2 sensitive and specific for MDA-MB-231 with an LR from 10 to
functionalized with a primary antibody. Further, the developed 100000 cells/mL and a LOD of 10 cells/mL [174]. Overexpression of
immunosensor was dipped in different concentrations of MCF-7 CD44 is associated with various cancer cell surfaces and plays a
solutions and subsequently, ssDNA-secondary antibody solutions pivotal role in cancer development through metastasis, invasion,
were dropped. Due to the NM-based signal amplification, and DNA- adhesion, and angiogenesis. Hyaluronic acid (HA) is a key ligand for
labeled antibodies, the proposed biosensor was stable, specific, and CD44 with high affinity. Recently, Zhou et al. developed a label-free
sensitive for MCF-7 with an LR from 100-1,000,000 cells/mL and a EIS cytosensor for the detection of MDA-MB-231 cells and CD44
LOD of 80 cells/mL [169]. overexpression by using GCE modified with HA-BSA-modified
Biofouling and control of selectivity is a challenge for the con- AuNPs. The modified electrode exhibited enhanced capture of
struction of reusable electrode surfaces, particularly in complex CD44-overexpressing cancer cells (MDA-MB-231) due to specific
media. Liu et al. proposed an EC aptasensor with an antifouling molecular recognition of HA. Under the optimal experimental
interface for the quantification of cancer cells in complex biological conditions, the developed cytosensor was sensitive and specific for
media. The antifouling interface was constructed by covalent MDA-MB-231 with an LR from 2.0  102e 3.0  105 cells/mL and a
attachment of branched zwitterionic peptides on the PANI- LOD of 128 cells/mL [175].
modified GCE. The developed interface exhibited a reduction in
non-specific cell and protein adsorption. The developed aptasensor 5.5. Detection of multiple BC biomarkers
with MUC1 aptamer was able to detect MUC1-positive MCF-7 BC
cells with magnified selectivity and sensitivity with an LR from 50- Detection of a single biomarker is not reliable for early diag-
1,000,000 cells/mL and a LOD of 20 cells/mL. Moreover, the assay nosis, and prognosis of BC due to different molecular events
performance of the cytosensor was the same without any fouling involved in BC development, intersubject variations in the
due to the antifouling peptide [170]. Another aptasensor based on expression of biomarkers, and scarcity of specific biomarkers.
GCE modified with a nanohybrid of TiO2 nanotube-rGO linked with Therefore, multiplex determination of specific biomarkers can lead
MUC1 aptamer was developed for the sensitive detection of MCF- to early, reliable, and personalized diagnosis of BC without false
7 cells. The modified electrode with hybrid provides enhanced positives [22]. EC biosensors are a promising platform for the
surface area enabling more sites for MUC1 immobilization. The multiplex analysis of BC biomarkers at the different or same mo-
developed aptasensor was sensitive for MCF-7 with an LR from 103- lecular level in contrast to conventional techniques. Various SPE/
107 cells/mL and a LOD of 40 cells/mL [171]. Recently, a label-free EC GCE-based EC biosensors have been developed for the simulta-
aptasensor for MCF-7 detection based on GCE modified with an neous determination of multiple BC biomarkers.
rGO-chitosan-Au NPs composite was developed. The aptasensor CA 15-3 and HER2-ECD are HER2-ECD are independent bio-
was developed by immobilization of AS1411 aptamer onto the markers but the combination of these two plays a key role in the
modified electrode surface and followed by incubation in an MCF- diagnosis of high-risk BC patients. Therefore, Marques et al. devel-
7 cell line sample. After incubation, the cell lines interact with the oped a voltammetric sandwich-type immunosensor for the simul-
aptamer resulting in cell capture on the biosensor. The developed taneous determination of CA 15-3 and HER2-ECD based on SPdCE
aptasensor exhibited enhanced signal and magnified detection modified with Au NPs. Further, the modified electrode was coated
limit due to the additive effects of composite enabling enhanced with captured antibodies (anti-CA 15-3, and anti-HER2-ECD), and
surface area and electrical conductivity. The developed aptasensor the free surface was blocked by casein. After incubation with target
was reproducible, stable, sensitive, and selective for MCF-7 with an biomarkers solution and labeled detection antibodies, antigen-
LR from 10-1,000,000 cells/mL and a LOD of 4 cells/mL [172]. In antibody reactions were monitored by LSV. The developed immu-
another recent study, MCF-7 was detected by a highly sensitive nosensor was non-invasive and specific for CA 15-3, and HER2-ECD
biosensor based on the synergistic effect of self-assembled aptamer [176]. In another study, a dual immunosensor was developed for the
inserted Arch-cruciform DNA on vanadium disulfide (VS2)/Au NPs simultaneous determination of Tumor Necrosis Factor-alpha (TNF),
modified GCE. In the absence of cancer cells, a cruciform DNA and Receptor Activator of Nuclear Factor-kB Ligand (RANKL) by us-
tagged with three terminal biotins was bound to arch DNA's top, ing SPdCEs, and neutravidin-functionalized magnetic microbeads
and further attached with streptavidin labeled HRP to catalyze the (Neu-MBs). The proposed biosensor was sandwich-type using bio-
HQ-H2O2 reaction on the modified GCE surface. Furthermore, the tinylated specific capture antibodies, detector antibodies, and HRP-
cruciform DNA was released in the presence of MCF-7 cells and labeled secondary antibodies, functionalized onto Neu-MBs. EC
decreased the concentration of immobilized HRP enabling enzyme- detection was carried out by amperometric transduction at SPdCEs
mediated electrocatalysis inhibition (Fig. 16). The EC response of using the H2O2/HQ system. The proposed immunosensor was
the developed biosensor was negatively correlated with the con- ultrasensitively and offered a detection limit of 3.0 pg/mL and
centration of cancer cells, with an LR from 10 to 100000 cells/mL, 2.6 pg/mL for TNF and RANKL respectively [177].
and a LOD of 5 cells/mL. The developed biosensor may be used for Guo et al. reported a sandwich-type EC immunosensor for the
the diagnosis of CTC in real sample analysis due to the 2-D materials detection of MUC1 and MCF-7 cells based on Ag nanoclusters
and enzyme-based dual signal amplification [173]. mediated signal amplification. The immunosensor was developed
The MDA-MB-231 cell lines are commonly used to diagnose by immobilization of anti-MUC1 on a GCE followed by placing the
late-stage BC and are an acceptable model for triple-negative BC MUC1 sample. Further Ag nanoclusters were added for signal
due to the lack of HER2 [211]. Tang et al. developed a label-free EIS amplification via deposition of metallic silver on Ag nanoclusters,
30
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

Fig. 16. Development and principle of cytosensor for MCF-7 using self-assembled aptamer inserted Arch-cruciform DNA on vanadium disulfide (VS2)/gold nanoparticles (Au NPs)
modified glassy carbon electrode. Adaptee from Ref. [173], MDPI, 2021.

and signals were monitored by SWV. The developed immunosensor was reliable, sensitive, and specific for CA 15-3, and CEA with a
was sensitive and selective for MUC1 and MCF-7 with a detection detection limit of 1.3  102 U/mL, and 11.2 pg/mL, respectively
limit of 0.5 nM, and 50 cells/mL, respectively [178]. Another [180]. Very recently, a label-free EC biosensor was developed for the
sandwich-type immunosensor was developed for the detection of simultaneous dual detection of CA 15-3 and miRNA 21. The
MUC1 and MCF-7 cells using Ag NP labels and a poly(glutamic biosensor was based on poly(3-aminobenzylamine)/two-dimen-
acid)/MWCNT nanocomposite. The immunosensor was developed sional (2D) MoSe2/GO-NC modified dual SPGE, functionalized
by the immobilization of MUC1 binding aptamer on the surface of independently with 2,3-diaminophenazine-Au NPs and toluidine
GCE modified with poly (glutamic acid)/MWCNT nanocomposite, blue-Au NPs. Both of the redox probe-Au NPs were employed as
and followed by the immobilization of another aptamer labeled signaling molecules and aided the immobilization of anti-CA 15-3
with Ag NPs for recognition of MCF-7 cells enabling magnified antibodies and capture DNA-21 probes, respectively. The modified
selectivity and signal amplification. Under the optimal experi- electrodes exhibited a magnified immobilization of capture probe
mental conditions, the developed immunosensor was reproducible, and antibodies due to a high surface-to-volume ratio and superior
stable, selective, and sensitive for MUC1 and MCF-7 with a LOD of conductivity leading to efficient dual detection. The developed
3 ng/mL, and 25 cells/mL, respectively [179]. biosensor was selective, and sensitive with a LOD of 1.2 fM and 0.14
Recently, a sandwich-type EC aptasensor was fabricated for the U/mL for miRNA-21, and CA 15-3 respectively [181].
simultaneous detection of CA 15-3 and CEA using AuNPs/3-D Gr
hydrogel nanocomposite modified GCE as a biosensing substrate, 6. Challenges and future perspectives
and CA 15e3 and CEA aptamers attached to AuNPs/redox probe/Gr
nanocomposite as a biosensing probe. In this study, ferrocene and EC biosensors based on SPEs and GCEs are a promising inex-
hemin were used as redox probes for CA 15-3 and CEA respectively pensive alternative for the non-invasive diagnosis of circulating BC
for dual detection. CA 15e3 and CEA aptamers were immobilized biomarkers at different molecular levels due to their simplicity,
on the surface of the modified electrode. It was observed that the accuracy, portability, intrinsic miniaturization, compatibility with
surface modification was responsible for the augmented surface semiconductor technologies, and standard microfabrication.
area resulting in more immobilization of aptamer on its surface. Despite various research, these biosensors face certain limitations
Additionally, the nanocomposites increased the sensitivity of the like complex protocols, multiple steps, and the preparation of
aptasensor due to its high conductivity. Moreover, the aptamers various reagents required for EC detection. Moreover, the discussed
used as capture and reporter probes made this aptasensor more biosensors are only prototyped and assessed under laboratory
specific in contrast to other BRE. The developed duplex aptasensor conditions and far from the real world. None of them have been
31
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

used for a large number of real patient samples to evaluate their biomarkers are also carried out by using both electrodes. Consid-
reliability and validity [22]. Furthermore, these devices are not ering the magnified sensitivity, most of the reported biosensors
commercially available due to insufficient clinical validation studies have been made by surface modification of SPEs/GCEs with various
regarding the intended motive of describing the procedure, preci- NMs, enzymes, and metals due to their unique properties. The
sion, and selectivity. Additionally, a lack of practical solutions to broad variability of the different approaches shows the prospect of
address the basic analytical issues related to non-specific binding, using various bioreceptors, bioassays, and signal amplification
toxicity, reproducibility, and stability [212]. The lack of communi- strategies in connection with the electrode substrates and EC
cation between the researchers and physicians is another challenge techniques. Moreover, these methodologies deal in a wide range
to the successful development of these devices. Storage and pro- from a simple label-free test involving analyte-induced changes in
cessing of samples is also another challenge due to their perish- various properties of the complex formed to very complicated
ability. Therefore, a simple rapid diagnostic process is required multistep but attractive methodologies enabling better recognition
which includes in situ detection or automatic sample processing. of analytes. However, further research is still required to address
Moreover, a comprehensive validation via the large numbers of real the different challenges like reliability, validity, sensitivity, and
patient samples analysis and the comparison against other avail- selectivity in the determination of multianalyte. More research
able competitive approaches remains to be addressed. For needs to be carried out for the commercialization, and multiplex
commercialization, the developed devices should be reproducible, analysis of various biomarkers via the fabrication of multiarray
stable, and robust through the optimization of different variables biosensors with minimum error. Additionally, a multidisciplinary
by using the design of experiments. collaboration should be taken between the various research/aca-
In the future, advancements in multiplex analysis can magnify demic institutions and hospitals to improve the efficacy of devel-
the sensitivity and specificity of these biosensors by using a single oped devices with ample real patient samples. Recent
sample. Microfluidics-based lab-on-chip devices and recent ap- advancements concerning SPEs and GCEs-based EC biosensor
proaches in nanotechnology open novel directions for designing fabrication for early diagnosis of BC have the potential for simple
multiplexed, miniaturized, and entirely automated biosensors for and cost-effective treatment for society. Therefore, this review will
cancer diagnosis. These portable, high throughput and inexpensive impart relevant background for the design and fabrication of
approaches impart low LOD and magnified sensitivity along with innovative POC devices for diverse BC biomarker monitoring which
simultaneous analysis of different biomarkers on a single chip. would open the way for augmenting cancer diagnosis and
Further, automation in sensor technology to involve various bio- therapeutics.
markers patterns software can enhance the potential of these de-
vices for newly discovered biomarkers for POC testing. Additionally, Author contributions
advances in bioinformatics, medicine, materials science, nano-
technology, and synthetic biology may improve the reliability and Conceptualization, writingdoriginal draft preparation Preeti
practical application of these devices through personalization. Kush, Praveen Lakhera, Vikas Chaudhary, and Amit Jha; wri-
Biofouling and control of selectivity is another challenge for the tingdreview and editing, Parveen Kumar and Ranjit Singh.
construction of reusable electrode surfaces, particularly in complex
media. Biofouling is the colonization of undesirable biological Declaration of competing interest
materials, and bacteria onto the electrode surfaces leading to
hampering the selectivity of EC sensors resulting in false-positive or The authors declare that they have no known competing
false-negative results. Recently, various strategies are used to financial interests or personal relationships that could have
develop an antifouling surface using biocompatible materials (PEG, appeared to influence the work reported in this article.
zwitterions, biomimetic materials, synthetic peptides, etc.) but face
reduced EC sensitivity due to the poor electrical conductivity of the Data availability
polymer backbone [213e215]. Moreover, the properties of different
BRE also reduce the antifouling efficiency under different circum- Review article
stances [213]. These limitations can be overcome by using con-
ducting polymers with other antifouling materials. A better
Acknowledgment
understanding of the fouling of electrodes at any phase can lead to
research for targeted antifouling agents. Thus investigating the
Praveen Lakhera thanks to Indian Council of Medical Research,
phenomena in their interface chemistry on the electrode and with
India for the senior research fellowship award from grant number
BRE can result in long storage life and better performance in the
5/3/8/38/ITR-F/2020-ITR for project titled “Design and develop-
assay. Additionally, a multifunctional probe comprising both
ment of colorimetric technique-based point of care device for the
recognizing and antifouling effects may be another approach to
detection of Glycated Haemoglobin”. Preeti Kush and Ranjit Singh
resolve such limitations.
thank to Shobhit University, Gangoh, Saharanpur, Uttar Pradesh
247341 Saharanpur, U.P.
7. Conclusion

SPEs and GCEs are ideal for analytical purposes due to their References
individually unique properties and can be easily integrated with [1] M. Hasanzadeh, N. Shadjou, M. de la Guardia, Early stage screening of breast
other approaches to fit a specific application. EC biosensors when cancer using electrochemical biomarker detection, TrAC, Trends Anal. Chem.
integrated with SPEs and GCEs, provide a promising inexpensive 91 (2017) 67e76.
[2] J. Ferlay, M. Ervik, F. Lam, M. Colombet, L. Mery, M. Pin ~ eros, A. Znaor,
alternative for the non-invasive diagnosis of circulating BC bio-
I. Soerjomataram, F. Bray, Global Cancer Observatory: Cancer Today, inter-
markers. This comprehensive review focuses on the recent devel- national agency for research on cancer, Cancer Today, Lyon, France, 2018.
opment and fabrication of the EC bioscaffolds based on modified [3] S. Şahin, M.O. Caglayan, Z. Üstündag , Recent advances in aptamer-based
SPEs and GCEs for the early diagnosis of diverse BC biomarkers at sensors for breast cancer diagnosis: special cases for nanomaterial-based
VEGF, HER2, and MUC1 aptasensors, Microchim. Acta 187 (10) (2020) 549.
different molecular levels like genomic, transcriptomic, and pro- [4] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2019, CA A Cancer J. Clin. 69
teomics. Further, CTC and multiplex determination of BC (1) (2019) 7e34.

32
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

[5] V. Gajdosova, L. Lorencova, P. Kasak, J. Tkac, Electrochemical nanobiosensors cancer, in: P. Chandra, Y.N. Tan, S.P. Singh (Eds.), Next Generation Point-of-
for detection of breast cancer biomarkers, Sensors (Basel) 20 (14) (2020) care Biomedical Sensors Technologies for Cancer Diagnosis, Springer
4022. Singapore, Singapore, 2017, pp. 253e278.
[6] R. Ahirwar, N. Khan, S. Kumar, Aptamer-based sensing of breast cancer [34] I. Diaconu, C. Cristea, V. Ha ^rceaga , G. Marrazza, I. Berindan-Neagoe,
biomarkers: a comprehensive review of analytical figures of merit, Expert R. S
andulescu, Electrochemical immunosensors in breast and ovarian cancer,
Rev. Mol. Diagn. 21 (7) (2021) 703e721. Clinica chimica acta, Int. J. Clinical. Chem. 425 (2013) 128e138.
[7] F. Vajhadin, S. Ahadian, J. Travas-Sejdic, J. Lee, M. Mazloum-Ardakani, [35] S. Campuzano, P. Ya n
~ ez-Seden~ o, J.M. Pingarro
 n, Electrochemical genosensing
J. Salvador, G.E. Aninwene 2nd, P. Bandaru, W. Sun, A. Khademhossieni, of circulating biomarkers, Sensors (Basel) 17 (4) (2017).
Electrochemical cytosensors for detection of breast cancer cells, Biosens. [36] F. Arduini, L. Micheli, D. Moscone, G. Palleschi, S. Piermarini, F. Ricci, G. Volpe,
Bioelectron. 151 (2020), 111984. Electrochemical biosensors based on nanomodified screen-printed elec-
[8] C. Nún ~ ez, Blood-based protein biomarkers in breast cancer, Clinica chimica trodes: recent applications in clinical analysis, TrAC, Trends Anal. Chem. 79
acta, Int. J. Clinical. Chem. 490 (2019) 113e127. (2016) 114e126.
[9] L. Wang, Early diagnosis of breast cancer, Sensors (Basel) 17 (7) (2017). [37] A. Sinha, Dhanjai, S.M. Mugo, H. Zhao, J. Chen, R. Jain, Chapter 5 - electro-
[10] S. Mittal, H. Kaur, N. Gautam, A.K. Mantha, Biosensors for breast cancer chemical immunosensors for rapid detection of breast cancer biomarkers, in:
diagnosis: a review of bioreceptors, biotransducers and signal amplification Inamuddin, R. Khan, A. Mohammad, A.M. Asiri (Eds.), Advanced Biosensors
strategies, Biosens. Bioelectron. 88 (2017) 217e231. for Health Care Applications, Elsevier, 2019, pp. 147e169.
[11] I.M. Mostafa, Y. Tian, S. Anjum, S. Hanif, M. Hosseini, B. Lou, G. Xu, [38] E.C. Rama, A. Costa-García, Screen-printed Electrochemical Immunosensors
Comprehensive review on the electrochemical biosensors of different breast for the Detection of Cancer and Cardiovascular Biomarkers, Electroanalys. 28
cancer biomarkers, Sensor. Actuator. B Chem. 365 (2022), 131944. (8) (2016) 1700e1715.
[12] D.C. Ferreira, M.R. Batistuti, B. Bachour, M. Mulato, Aptasensor based on [39] F.M. Alkabban, T. Ferguson, Breast Cancer, StatPearls, StatPearls Publishing
screen-printed electrode for breast cancer detection in undiluted human Copyright © 2021, StatPearls Publishing LLC., Treasure Island (FL), 2021.
serum, Bioelectrochemistry 137 (2021), 107586. [40] M. Begum, S. Karim, A. Malik, R. Khurshid, M. Asif, A. Salim, S.A. Nagra,
[13] L. Li, D. Feng, J. Zhao, Z. Guo, Y. Zhang, Simultaneous fluoroimmunoassay of A. Zaheer, Z. Iqbal, A.M. Abuzenadah, M.H. Alqahtani, M. Rasool, CA 15-3
two tumor markers based on CdTe quantum dots and gold nanocluster (Mucin-1) and physiological characteristics of breast cancer from Lahore,
coated-silica nanospheres as labels, RSC Adv. 5 (128) (2015) 105992e105998. Pakistan, Asian Pacific Journal of Cancer Prevention : Asian Pac. J. Cancer
[14] V. Beral, D. Bull, R. Doll, T. Key, Breast cancer and hormone replacement Prev. APJCP 13 (10) (2012) 5257e5261.
therapy: collaborative reanalysis of data from 51 epidemiological studies of [41] E.A. Rakha, T.C. Putti, D.M. Abd El-Rehim, C. Paish, A.R. Green, D.G. Powe,
52 705, Lancet 350 (1997) 9084. A.H. Lee, J.F. Robertson, I.O. Ellis, Morphological and immunophenotypic
[15] E. Asav, M.K. Sezgintürk, A novel impedimetric disposable immunosensor for analysis of breast carcinomas with basal and myoepithelial differentiation,
rapid detection of a potential cancer biomarker, Int. J. Biol. Macromol. 66 J. Pathol. 208 (4) (2006) 495e506.
(2014) 273e280. [42] A. Bombonati, D.C. Sgroi, The molecular pathology of breast cancer pro-
[16] D.R. Hansberry, A. John, E. John, N. Agarwal, S.F. Gonzales, S.R. Baker, gression, J. Pathol. 223 (2) (2011) 307e317.
A critical review of the readability of online patient education resources from [43] C.E. DeSantis, J. Ma, A. Goding Sauer, L.A. Newman, A. Jemal, Breast cancer
RadiologyInfo, Org. Am. J. Roentgenol. 202 (3) (2014) 566e575. statistics, 2017, racial disparity in mortality by state, CA A Cancer J. Clin. 67
[17] M. Mazloum- Ardakani, Z. Tavakolian- Ardakani, N. Sahraei, (6) (2017) 439e448.
S.M. Moshtaghioun, Fabrication of an ultrasensitive and selective electro- [44] N.J. Ronkainen, S.L. Okon, Nanomaterial-based electrochemical immuno-
chemical aptasensor to detect carcinoembryonic antigen by using a new sensors for clinically significant biomarkers, Materials 7 (6) (2014)
nanocomposite, Biosens. Bioelectron. 129 (2019) 1e6. 4669e4709.
[18] H.A. Alhadrami, Biosensors: Classifications, medical applications, and future [45] A. Rasooly, J. Jacobson, Development of biosensors for cancer clinical testing,
prospective, Biotechnol. Appl. Biochem. 65 (3) (2018) 497e508. Biosens. Bioelectron. 21 (10) (2006) 1851e1858.
[19] S. Carvajal, S.N. Fera, A.L. Jones, T.A. Baldo, I.M. Mosa, J.F. Rusling, C.E. Krause, [46] M. Perfe zou, A. Turner, A. Merkoçi, Cancer detection using nanoparticle-
Disposable inkjet-printed electrochemical platform for detection of clinically based sensors, Chem. Soc. Rev. 41 (7) (2012) 2606e2622.
relevant HER-2 breast cancer biomarker, Biosens. Bioelectron. 104 (2018) [47] C.L. Sawyers, The cancer biomarker problem, Nature 452 (7187) (2008)
158e162. 548e552.
[20] M. Li, D.-W. Li, G. Xiu, Y.-T. Long, Applications of screen-printed electrodes in [48] P. Ranjan, A. Parihar, S. Jain, N. Kumar, C. Dhand, S. Murali, D. Mishra,
current environmental analysis, Curr. Opin. Electrochem. 3 (1) (2017) S.K. Sanghi, J.P. Chaurasia, A.K. Srivastava, R. Khan, Biosensor-based diag-
137e143. nostic approaches for various cellular biomarkers of breast cancer: a
[21] M.H. Naveen, N.G. Gurudatt, Y.-B. Shim, Applications of conducting polymer comprehensive review, Anal. Biochem. 610 (2020), 113996.
composites to electrochemical sensors: a review, Appl. Mater. Today 9 [49] D. Sadighbayan, K. Sadighbayan, M.R. Tohid-kia, A.Y. Khosroushahi,
(2017) 419e433. M. Hasanzadeh, Development of electrochemical biosensors for tumor
[22] S. Campuzano, M. Pedrero, J.M. Pingarro n, Non-invasive breast cancer marker determination towards cancer diagnosis: recent progress, TrAC,
diagnosis through electrochemical biosensing at different molecular levels, Trends Anal. Chem. 118 (2019) 73e88.
Sensors (Basel) 17 (9) (2017). [50] F. Cui, Z. Zhou, H.S. Zhou, Reviewdmeasurement and analysis of cancer
[23] P. Kush, P. Kumar, R. Singh, A. Kaushik, Aspects of high-performance and bio- biomarkers based on electrochemical biosensors, J. Electrochem. Soc. 167 (3)
acceptable magnetic nanoparticles for biomedical application, Asian J. (2020), 037525.
Pharm. Sci. 16 (6) (2021) 704e737. [51] F. Mollarasouli, S. Kurbanoglu, S.A. Ozkan, The role of electrochemical
[24] T. Florence, Anodic stripping voltammetry with a glassy carbon electrode immunosensors in clinical analysis, Biosensors (Basel) 9 (3) (2019).
mercury-plated in situ, J. Electroanal. Chem. Interfacial Electrochem. 27 (2) [52] M. Sharifi, M.R. Avadi, F. Attar, F. Dashtestani, H. Ghorchian, S.M. Rezayat,
(1970) 273e281. A.A. Saboury, M. Falahati, Cancer diagnosis using nanomaterials based
[25] S. Azhari, P. Sathishkumar, R. Ahamad, F. Ahmad, A.R. Mohd Yusoff, Fabri- electrochemical nanobiosensors, Biosens. Bioelectron. 126 (2019) 773e784.
cation of a composite modified glassy carbon electrode: a highly selective, [53] M. Hassler, 3 - other commonly used biomedical coatings: pyrolytic carbon
sensitive and rapid electrochemical sensor for silver ion detection in river coatings, in: M. Driver (Ed.), Coatings for Biomedical Applications, Woodhead
water samples, Anal. Methods 8 (28) (2016) 5712e5721. Publishing, 2012, pp. 75e105.
[26] M. Li, Y.-T. Li, D.-W. Li, Y.-T. Long, Recent developments and applications of [54] N.B. Mincu, V. Lazar, D. Stan, C.M. Mihailescu, R. Iosub, A.L. Mateescu, Screen-
screen-printed electrodes in environmental assaysda review, Anal. Chim. printed electrodes (SPE) for in vitro diagnostic purpose, Diagnostics (Basel,
Acta 734 (2012) 31e44. Switzerland) 10 (8) (2020).
[27] I.H. Cho, D.H. Kim, S. Park, Electrochemical biosensors: perspective on [55] E. Martínez-Perin ~
an, C. Gutierrez-Sa nchez, T. García-Mendiola, E. Lorenzo,
functional nanomaterials for on-site analysis, Biomater. Res. 24 (2020) 6. Electrochemiluminescence biosensors using screen-printed electrodes, Bio-
[28] B. Wang, U. Akiba, J.-i. Anzai, Recent progress in nanomaterial-based elec- sens. (Basel) 10 (9) (2020) 118.
trochemical biosensors for cancer biomarkers: a review, Molecules 22 (7) [56] Z. Taleat, A. Khoshroo, M. Mazloum-Ardakani, Screen-printed electrodes for
(2017) 1048. biosensing: a review (2008e2013), Microchim. Acta 181 (9e10) (2014)
[29] I.E. Tothill, Biosensors for Cancer Markers Diagnosis, Seminars in Cell & 865e891.
Developmental Biology, Elsevier, 2009, pp. 55e62. [57] R.A. Couto, J.L. Lima, M.B. Quinaz, Recent developments, characteristics and
[30] R. Stern, Hyaluronidases in cancer biology, Hyaluronan Cancer Bio. (2008) potential applications of screen-printed electrodes in pharmaceutical and
207e220. Elsevier. biological analysis, Talanta 146 (2016) 801e814.
[31] M. Liu, Z. Wang, T. Tan, Z. Chen, X. Mou, X. Yu, Y. Deng, G. Lu, N. He, An [58] E.C. Rama, A. Costa-García, Screen-printed electrochemical immunosensors
aptamer-based probe for molecular subtyping of breast cancer, Theranostics for the detection of cancer and cardiovascular biomarkers, Electroanalys. 28
8 (20) (2018) 5772. (8) (2016) 1700e1715.
[32] P.F. Rostamabadi, E. Heydari-Bafrooei, Impedimetric aptasensing of the [59] L. Valdevit, J. Bauer, Chapter 13.1 - fabrication of 3D micro-/nanoarchitected
breast cancer biomarker HER2 using a glassy carbon electrode modified with materials, in: T. Baldacchini (Ed.), Three-Dimensional Microfabrication Using
gold nanoparticles in a composite consisting of electrochemically reduced Two-Photon Polymerization, second ed., William Andrew Publishing, 2020,
graphene oxide and single-walled carbon nanotubes, Microchim. Acta 186 pp. 541e576.
(8) (2019) 495. [60] L. Redivo, M. Stredanský, E. De Angelis, L. Navarini, M. Resmini,  
L. Svorc, Bare
[33] S. Singh, A. Deep, G. Mohanta, V.K. Meena, Developments in the electro- carbon electrodes as simple and efficient sensors for the quantification of
chemical bionanosensors for the predictive diagnosis of prostate and breast caffeine in commercial beverages, R. Soc. Open Sci. 5 (5) (2018), 172146.

33
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

[61] S. Sharma, Glassy carbon: a promising material for micro- and nano- [87] R.M. Torrente-Rodríguez, S. Campuzano, E. Lo pez-Herna ndez, V.R. Montiel,
manufacturing, Materials (Basel, Switzerland) 11 (10) (2018). R. Barderas, R. Granados, J.M. Sa nchez-Puelles, J.M. Pingarron, Simultaneous
[62] Y. Yi, G. Weinberg, M. Prenzel, M. Greiner, S. Heumann, S. Becker, R. Schlo €gl, detection of two breast cancer-related miRNAs in tumor tissues using p19-
Electrochemical corrosion of a glassy carbon electrode, Catal. Today 295 based disposable amperometric magnetobiosensing platforms, Biosens.
(2017) 32e40. Bioelectron. 66 (2015) 385e391.
[63] Activation of Glassy Carbon Electrodes, 2020. [88] A. Erdem, E. Eksin, G. Congur, Indicator-free electrochemical biosensor for
[64] C.X. Du, L. Han, S.L. Dong, L.H. Li, Y. Wei, A novel procedure for fabricating microRNA detection based on carbon nanofibers modified screen printed
flexible screen-printed electrodes with improved electrochemical perfor- electrodes, J. Electroanal. Chem. 755 (2015) 167e173.
mance, IOP Conf. Ser. Mater. Sci. Eng. 137 (2016), 012060. [89] A.R. Cardoso, F.T.C. Moreira, R. Fernandes, M.G.F. Sales, Novel and simple
[65] P. Kumar, A. Deep, S.C. Sharma, L.M. Bharadwaj, Bioconjugation of InGaP electrochemical biosensor monitoring attomolar levels of miRNA-155 in
quantum dots for molecular sensing, Anal. Biochem. 421 (1) (2012) breast cancer, Biosens. Bioelectron. 80 (2016) 621e630.
285e290. [90] H. Mohammadi, A. Amine, Spectrophotometric and electrochemical deter-
[66] A.L. Sharma, P. Kumar, A. Deep, Highly sensitive glucose sensing with multi- mination of MicroRNA-155 using sandwich hybridization magnetic beads,
walled carbon nanotubes e polyaniline composite, Polym.-Plast. Technol. Anal. Lett. 51 (3) (2018) 411e423.
Eng. 51 (13) (2012) 1382e1387. [91] E. Povedano, V. Ruiz-Valdepen ~ as Montiel, M. Gamella, V. Serafín, M. Pedrero,
[67] M. Kukkar, A. Sharma, P. Kumar, K.H. Kim, A. Deep, Application of MoS2 L. Moranova, M. Bartosik, J.J. Montoya, P. Y ~ ez-Seden
an ~ o, S. Campuzano,
modified screen-printed electrodes for highly sensitive detection of bovine J.M. Pingarro n, A novel zinc finger protein-based amperometric
serum albumin, Anal. Chim. Acta 939 (2016) 101e107. biosensor for miRNA determination, Anal. Bioanal. Chem. 412 (21) (2020)
[68] A. Leniart, M. Brycht, B. Burnat, S. Skrzypek, An application of a glassy carbon 5031e5041.
electrode and a glassy carbon electrode modified with multi-walled carbon [92] M. Zouari, S. Campuzano, J.M. Pingarro  n, N. Raouafi, Femtomolar direct
nanotubes in electroanalytical determination of oxycarboxin, Ionics 24 (7) voltammetric determination of circulating miRNAs in sera of cancer patients
(2018) 2111e2121. using an enzymeless biosensor, Anal. Chim. Acta 1104 (2020) 188e198.
[69] M.R. Hasan, M.S. Ahommed, M. Daizy, M.S. Bacchu, M.R. Ali, M.R. Al-Mamun, [93] C. Pothipor, N. Aroonyadet, S. Bamrungsap, J. Jakmunee, K. Ounnunkad,
M.A. Saad Aly, M.Z.H. Khan, S.I. Hossain, Recent development in electro- A highly sensitive electrochemical microRNA-21 biosensor based on inter-
chemical biosensors for cancer biomarkers detection, Biosens. Bioelectron. X calating methylene blue signal amplification and a highly dispersed gold
8 (2021), 100075. nanoparticles/graphene/polypyrrole composite, Analyst 146 (8) (2021)
[70] W. Putzbach, N.J. Ronkainen, Immobilization techniques in the fabrication of 2679e2688.
nanomaterial-based electrochemical biosensors: a review, Sensors 13 (4) [94] C. Pothipor, J. Jakmunee, S. Bamrungsap, K. Ounnunkad, An electrochemical
(2013) 4811e4840. biosensor for simultaneous detection of breast cancer clinically related
[71] F. Ricci, G. Adornetto, G. Palleschi, A review of experimental aspects of microRNAs based on a gold nanoparticles/graphene quantum dots/graphene
electrochemical immunosensors, Electrochim. Acta 84 (2012) 74e83. oxide film, Analyst 146 (12) (2021) 4000e4009.
[72] G. Chang, H. Shu, K. Ji, M. Oyama, X. Liu, Y. He, Gold nanoparticles directly [95] H.A. Rafiee-Pour, M. Behpour, M. Keshavarz, A novel label-free electro-
modified glassy carbon electrode for non-enzymatic detection of glucose, chemical miRNA biosensor using methylene blue as redox indicator: appli-
Appl. Surf. Sci. 288 (2014) 524e529. cation to breast cancer biomarker miRNA-21, Biosens. Bioelectron. 77 (2016)
[73] Z. Li, J. Zhang, Y. Zhou, S. Shuang, C. Dong, M.M.F. Choi, Electrodeposition of 202e207.
palladium nanoparticles on fullerene modified glassy carbon electrode for [96] A. Ebrahimi, I. Nikokar, M. Zokaei, E. Bozorgzadeh, Design, development and
methane sensing, Electrochim. Acta 76 (2012) 288e291. evaluation of microRNA-199a-5p detecting electrochemical nanobiosensor
[74] L. Dhanapala, C.E. Krause, A.L. Jones, J.F. Rusling, Printed electrodes in with diagnostic application in Triple Negative Breast Cancer, Talanta 189
microfluidic arrays for cancer biomarker protein detection, Biosensors (2018) 592e598.
(Basel) 10 (9) (2020). [97] D. Wang, J. Wang, A sensitive and label-free electrochemical microRNA
[75] F. Wei, P.B. Lillehoj, C.M. Ho, DNA diagnostics: nanotechnology-enhanced biosensor based on Polyamidoamine Dendrimer functionalized Polypyrrole
electrochemical detection of nucleic acids, Pediatr. Res. 67 (5) (2010) nanowires hybrid, Microchim. Acta 188 (5) (2021) 173.
458e468. [98] M. Azimzadeh, M. Rahaie, N. Nasirizadeh, K. Ashtari, H. Naderi-Manesh, An
[76] S.Z. Mousavisani, J.-B. Raoof, A.P.F. Turner, R. Ojani, W.C. Mak, Label-free DNA electrochemical nanobiosensor for plasma miRNA-155, based on graphene
sensor based on diazonium immobilisation for detection of DNA damage in oxide and gold nanorod, for early detection of breast cancer, Biosens. Bio-
breast cancer 1 gene, Sensor. Actuator. B Chem. 264 (2018) 59e66. electron. 77 (2016) 99e106.
[77] T. García-Mendiola, S. Requena-Sanz, E. Martínez-Perin ~an, I. Bravo, [99] K. Zhang, N. Zhang, L. Zhang, H. Wang, H. Shi, Q. Liu, Label-free impedimetric
F. Pariente, E. Lorenzo, Influence of carbon nanodots on DNA-Thionine sensing platform for microRNA-21 based on ZrO2-reduced graphene oxide
interaction. Application to breast cancer diagnosis, Electrochim. Acta 353 nanohybrids coupled with catalytic hairpin assembly amplification, RSC Adv.
(2020), 136522. 8 (29) (2018) 16146e16151.
[78] D. Feng, J. Su, G. He, Y. Xu, C. Wang, M. Zheng, Q. Qian, X. Mi, Electrochemical [100] J. Zhang, D.Z. Wu, S.X. Cai, M. Chen, Y.K. Xia, F. Wu, J.H. Chen, An
DNA sensor for sensitive BRCA1 detection based on DNA tetrahedral- immobilization-free electrochemical impedance biosensor based on duplex-
structured probe and poly-adenine mediated gold nanoparticles, Bio- specific nuclease assisted target recycling for amplified detection of micro-
sensors (Basel) 10 (7) (2020). RNA, Biosens. Bioelectron. 75 (2016) 452e457.
[79] P.A. Rasheed, N. Sandhyarani, A highly sensitive DNA sensor for attomolar [101] X. Ma, H. Xu, K. Qian, M. Kandawa-Schulz, W. Miao, Y. Wang, Electrochemical
detection of the BRCA1 gene: signal amplification with gold nanoparticle detection of microRNAs based on AuNPs/CNNS nanocomposite with Duplex-
clusters, Analyst 140 (8) (2015) 2713e2718. specific nuclease assisted target recycling to improve the sensitivity, Talanta
[80] W. Wang, X. Fan, S. Xu, J.J. Davis, X. Luo, Low fouling label-free DNA sensor 208 (2020), 120441.
based on polyethylene glycols decorated with gold nanoparticles for the [102] S. Sharma, J. Zapatero-Rodríguez, R. Saxena, R. O'Kennedy, S. Srivastava,
detection of breast cancer biomarkers, Biosens. Bioelectron. 71 (2015) Ultrasensitive direct impedimetric immunosensor for detection of serum
51e56. HER2, Biosens. Bioelectron. 106 (2018) 78e85.
[81] N. Hui, X. Sun, S. Niu, X. Luo, PEGylated polyaniline nanofibers: antifouling [103] E. Chocholova, T. Bertok, L. Lorencova, A. Holazova, P. Farkas, A. Vikartovska,
and conducting biomaterial for electrochemical DNA sensing, ACS Appl. V. Bella, D. Velicova, P. Kasak, A.A. Eckstein, J. Mosnacek, D. Hasko, J. Tkac,
Mater. Interfaces 9 (3) (2017) 2914e2923. Advanced antifouling zwitterionic layer based impedimetric HER2 bio-
[82] G. Wang, R. Han, X. Su, Y. Li, G. Xu, X. Luo, Zwitterionic peptide anchored to sensing in human serum: glycoprofiling as a novel approach for breast
conducting polymer PEDOT for the development of antifouling and ultra- cancer diagnostics, Sensor. Actuator. B Chem. 272 (2018) 626e633.
sensitive electrochemical DNA sensor, Biosens. Bioelectron. 92 (2017) [104] Y.W. Hartati, L.K. Letelay, S. Gaffar, S. Wyantuti, H.H. Bahti, Cerium
396e401. oxide-monoclonal antibody bioconjugate for electrochemical immuno-
[83] S. Shahrokhian, R. Salimian, Ultrasensitive detection of cancer biomarkers sensing of HER2 as a breast cancer biomarker, Sens. Bio-Sens. Res. 27
using conducting polymer/electrochemically reduced graphene oxide-based (2020), 100316.
biosensor: application toward BRCA1 sensing, Sensor. Actuator. B Chem. 266 [105] S.D. Tallapragada, K. Layek, R. Mukherjee, K.K. Mistry, M. Ghosh, Develop-
(2018) 160e169. ment of screen-printed electrode based immunosensor for the detection of
[84] M. Hasanzadeh, M. Feyziazar, E. Solhi, A. Mokhtarzadeh, J. Soleymani, HER2 antigen in human serum samples, Bioelectrochemistry 118 (2017)
N. Shadjou, A. Jouyban, S. Mahboob, Ultrasensitive immunoassay of breast 25e30.
cancer type 1 susceptibility protein (BRCA1) using poly (dopamine-beta [106] Z. Lah, S.A.A. Ahmad, M.S. Zaini, M.A. Kamarudin, An electrochemical sand-
cyclodextrine-Cetyl trimethylammonium bromide) doped with silver wich immunosensor for the detection of HER2 using antibody-conjugated
nanoparticles: a new platform in early stage diagnosis of breast cancer and PbS quantum dot as a label, J. Pharmaceut. Biomed. Anal. 174 (2019)
efficient management, Microchem. J. 145 (2019) 778e783. 608e617.
[85] Y.-M. Xia, M.-Y. Li, C.-L. Chen, M. Xia, W. Zhang, W.-W. Gao, Employing label- [107] M. Freitas, H.P.A. Nouws, E. Keating, C. Delerue-Matos, High-performance
free electrochemical biosensor based on 3D-reduced graphene oxide and electrochemical immunomagnetic assay for breast cancer analysis, Sensor.
polyaniline nanofibers for ultrasensitive detection of breast cancer BRCA1 Actuator. B Chem. 308 (2020), 127667.
biomarker, Electroanalysis 32 (9) (2020) 2045e2055. [108] M. Freitas, H.P.A. Nouws, E. Keating, V.C. Fernandes, C. Delerue-Matos,
[86] H. Ehzari, M. Safari, M. Samimi, Signal amplification of novel sandwich-type Immunomagnetic bead-based bioassay for the voltammetric analysis of the
genosensor via catalytic redox-recycling on platform MWCNTs/Fe(3)O(4) breast cancer biomarker HER2-ECD and tumour cells using quantum dots as
@TMU-21 for BRCA1 gene detection, Talanta 234 (2021), 122698. detection labels, Mikrochim. Acta 187 (3) (2020) 184.

34
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

[109] M. Freitas, M.M. Neves, H.P. Nouws, C. Delerue-Matos, Quantum dots as 29 breast cancer antigen detection, New J. Chem. 42 (13) (2018)
nanolabels for breast cancer biomarker HER2-ECD analysis in human serum, 11046e11053.
Talanta 208 (2020), 120430. [132] V.-A. Nguyen, H.L. Nguyen, D.T. Nguyen, Q.P. Do, L.D. Tran, Electro-
[110] M. Shamsipur, M. Emami, L. Farzin, R. Saber, A sandwich-type electro- synthesized poly(1,5-diaminonaphthalene)/polypyrrole nanowires bilayer
chemical immunosensor based on in situ silver deposition for determination as an immunosensor platform for breast cancer biomarker CA 15-3, Curr.
of serum level of HER2 in breast cancer patients, Biosens. Bioelectron. 103 Appl. Phys. 17 (11) (2017) 1422e1429.
(2018) 54e61. [133] R.S. Gomes, F.T.C. Moreira, R. Fernandes, M.G.F. Sales, Sensing CA 15-3 in
[111] H. Ehzari, M. Samimi, M. Safari, M.B. Gholivand, Label-free electrochemical point-of-care by electropolymerizing O-phenylenediamine (oPDA) on Au-
immunosensor for sensitive HER2 biomarker detection using the core-shell screen printed electrodes, PLoS One 13 (5) (2018), e0196656.
magnetic metal-organic frameworks, J. Electroanal. Chem. 877 (2020), [134] J. Amani, A. Khoshroo, M. Rahimi-Nasrabadi, Electrochemical immunosensor
114722. for the breast cancer marker CA 15-3 based on the catalytic activity of a CuS/
[112] S. Centane, T. Nyokong, The antibody assisted detection of HER2 on a cobalt reduced graphene oxide nanocomposite towards the electrooxidation of
porphyrin binuclear framework and gold functionalized graphene quantum catechol, Mikrochim. Acta 185 (1) (2017) 79.
dots modified electrode, J. Electroanal. Chem. 880 (2021), 114908. [135] A. Khoshroo, M. Mazloum-Ardakani, M. Forat-Yazdi, Enhanced performance
[113] H. Nasrollahpour, A. Naseri, M.-R. Rashidi, B. Khalilzadeh, Application of of label-free electrochemical immunosensor for carbohydrate antigen 15-3
green synthesized WO3-poly glutamic acid nanobiocomposite for early stage based on catalytic activity of cobalt sulfide/graphene nanocomposite, Sens.
biosensing of breast cancer using electrochemical approach, Sci. Rep. 11 (1) Actuator. B Chem. 255 (2018) 580e587.
(2021), 23994. [136] T.S. Martins, J.L. Bott-Neto, O.N. Oliveira, S.A.S. Machado, A sandwich-type
[114] K. Chan, H. Lim, N. Shams, S. Jayabal, A. Pandikumar, N. Huang, Fabrication of electrochemical immunosensor based on Au-rGO composite for CA15-3 tu-
graphene/gold-modified screen-printed electrode for detection of carci- mor marker detection, Microchim. Acta 189 (1) (2021) 38.
noembryonic antigen, Mater. Sci. Eng. C 58 (2016) 666e674. [137] T.S.C.R. Rebelo, J.A. Ribeiro, M.G.F. Sales, C.M. Pereira, Electrochemical
[115] L. Zhao, C. Li, H. Qi, Q. Gao, C. Zhang, Electrochemical lectin-based biosensor immunosensor for detection of CA 15-3 biomarker in point-of-care, Sens.
array for detection and discrimination of carcinoembryonic antigen using Bio-Sens. Res. 33 (2021), 100445.
dual amplification of gold nanoparticles and horseradish peroxidase, Sensor. [138] L. Liu, J. Gao, L. Guo, J. Xu, Nitrogen-doped graphene modified glassy carbon
Actuator. B Chem. 235 (2016) 575e582. electrode for electrochemical determination of breast cancer marker car-
[116] S. Lee, H. Lim, I. Ibrahim, A. Jamil, A. Pandikumar, N. Huang, Horseradish bohydrate antigen 15-3, Int. J. Electrochem. Sci. 12 (2017) 8280e8287.
peroxidase-labeled silver/reduced graphene oxide thin film-modified [139] Y. Liu, X. Weng, K.-K. Wang, Y. Xue, A.-J. Wang, L. Wu, J.-J. Feng, A novel
screen-printed electrode for detection of carcinoembryonic antigen, Bio- enzyme-free sandwich-like electrochemical immunosensor for the detection
sens. Bioelectron. 89 (2017) 673e680. of carbohydrate antigen 15-3 based on hierarchical AuPd nanochain net-
[117] L. Cao, H. Xiao, C. Fang, F. Zhao, Z. Chen, Electrochemical immunosensor works, Sensor. Actuator. B Chem. 247 (2017) 349e356.
based on binary nanoparticles decorated rGO-TEPA as magnetic capture and [140] M. Hasanzadeh, E. Solhi, M. Jafari, A. Mokhtarzadeh, J. Soleymani, A. Jouyban,
Au@PtNPs as probe for CEA detection, Mikrochim. Acta 187 (10) (2020) 584. S. Mahboob, Ultrasensitive immunoassay of tumor protein CA 15.3 in MCF-7
[118] N. Li, Y. Wang, W. Cao, Y. Zhang, T. Yan, B. Du, Q. Wei, An ultrasensitive breast cancer cell lysates and unprocessed human plasma using gold
electrochemical immunosensor for CEA using MWCNT-NH 2 supported PdPt nanoparticles doped on the structure of mesoporous silica, Int. J. Biol. Mac-
nanocages as labels for signal amplification, J. Mater. Chem. B 3 (9) (2015) romol. 120 (Pt B) (2018) 2493e2508.
2006e2011. [141] H. Jia, Q. Tian, J. Xu, L. Lu, X. Ma, Y. Yu, Aerogels prepared from polymeric b-
[119] J. Han, L. Jiang, F. Li, P. Wang, Q. Liu, Y. Dong, Y. Li, Q. Wei, Ultrasensitive non- cyclodextrin and graphene aerogels as a novel host-guest system for
enzymatic immunosensor for carcino-embryonic antigen based on palla- immobilization of antibodies: a voltammetric immunosensor for the tumor
dium hybrid vanadium pentoxide/multiwalled carbon nanotubes, Biosens. marker CA 15-3, Mikrochim. Acta 185 (11) (2018) 517.
Bioelectron. 77 (2016) 1104e1111. [142] M. Hasanzadeh, S. Tagi, E. Solhi, A. Mokhtarzadeh, N. Shadjou, A. Eftekhari,
[120] L. Zheng, X. Niu, J. Zhao, T. Liu, Y. Liu, Y. Yang, A label-free immunosensor for S. Mahboob, An innovative immunosensor for ultrasensitive detection of
CEA based on Pd-Ir bimetallic nanoparticles, J. Nanosci. Nanotechnol. 16 (6) breast cancer specific carbohydrate (CA 15-3) in unprocessed human plasma
(2016) 5984e5990. and MCF-7 breast cancer cell lysates using gold nanospear electrochemically
[121] Z. Tang, Z. Ma, Ultrasensitive amperometric immunoassay for carcinoem- assembled onto thiolated graphene quantum dots, Int. J. Biol. Macromol. 114
bryonic antigens by using a glassy carbon electrode coated with a (2018) 1008e1017.
polydopamine-Pb (II) redox system and a chitosan-gold nanocomposite, [143] A. Saadati, S. Hassanpour, M. Hasanzadeh, N. Shadjou, A. Hassanzadeh,
Microchim. Acta 184 (4) (2017) 1135e1142. Immunosensing of breast cancer tumor protein CA 15-3 (carbohydrate an-
[122] Y. Luo, Y. Wang, H. Yan, Y. Wu, C. Zhu, D. Du, Y. Lin, SWCNTs@GQDs com- tigen 15.3) using a novel nano-bioink: a new platform for screening of
posites as nanocarriers for enzyme-free dual-signal amplification electro- proteins in human biofluids by pen-on-paper technology, Int. J. Biol. Mac-
chemical immunoassay of cancer biomarker, Anal. Chim. Acta 1042 (2018) romol. 132 (2019) 748e758.
44e51. [144] C. Zhang, D. Zhang, Z. Ma, H. Han, Cascade catalysis-initiated radical poly-
[123] Y. Wang, G. Zhao, Y. Zhang, X. Pang, W. Cao, B. Du, Q. Wei, Sandwich-type merization amplified impedimetric immunosensor for ultrasensitive detec-
electrochemical immunosensor for CEA detection based on Ag/MoS2@Fe3O4 tion of carbohydrate antigen 15-3, Biosens. Bioelectron. 137 (2019) 1e7.
and an analogous ELISA method with total internal reflection microscopy, [145] U. Eletxigerra, J. Martinez-Perdiguero, S. Merino, R. Barderas, V. Ruiz-
Sensor. Actuator. B Chem. 266 (2018) 561e569. Valdepen ~ as Montiel, R. Villalonga, J.M. Pingarron, S. Campuzano, Estrogen
[124] Y. Yang, M. Jiang, K. Cao, M. Wu, C. Zhao, H. Li, C. Hong, An electrochemical receptor a determination in serum, cell lysates and breast cancer cells using
immunosensor for CEA detection based on Au-Ag/rGO@PDA nanocomposites an amperometric magnetoimmunosensing platform, Sens. Bio-Sens. Res. 7
as integrated double signal amplification strategy, Microchem. J. 151 (2019), (2016) 71e76.
104223. [146] U. Eletxigerra, J. Martinez-Perdiguero, S. Merino, R. Barderas, V. Ruiz-
[125] S.A. Nakhjavani, H. Afsharan, B. Khalilzadeh, M.H. Ghahremani, S. Carrara, Valdepen ~ as Montiel, R. Villalonga, J.M. Pingarro  n, S. Campuzano, Electro-
Y. Omidi, Gold and silver bio/nano-hybrids-based electrochemical immu- chemical magnetoimmunosensor for progesterone receptor determination.
nosensor for ultrasensitive detection of carcinoembryonic antigen, Biosens. Application to the simultaneous detection of estrogen and progesterone
Bioelectron. 141 (2019), 111439. breast-cancer related receptors in raw cell lysates, Electroanalysis 28 (8)
[126] Y. Lai, H. Huang, Z. Xia, S. Li, Y. Deng, X. Liu, A sandwich-type electrochemical (2016) 1787e1794.
immunosensor using polythionine/AuNPs nanocomposites as label for ul- [147] G. Bezerra, C. Co rdula, D. Campos, G. Nascimento, N. Oliveira, M.A. Seabra,
trasensitive detection of carcinoembryonic antigen, Mater. Exp. 9 (5) (2019) V. Visani, S. Lucas, I. Lopes, J. Santos, F. Xavier Jr., M.A. Borba, D. Martins,
444e450. J. Lima-Filho, Electrochemical aptasensor for the detection of HER2 in human
[127] K. Liu, H. Deng, Y. Wang, S. Cheng, X. Xiong, C. Li, A sandwich-type photo- serum to assist in the diagnosis of early stage breast cancer, Anal. Bioanal.
electrochemical immunosensor based on ReS2 nanosheets for high- Chem. 411 (25) (2019) 6667e6676.
performance determination of carcinoembryonic antigen, Sensor. Actuator. [148] T. Harahsheh, Y.F. Makableh, I. Rawashdeh, M. Al-Fandi, Enhanced apta-
B Chem. 320 (2020), 128341. sensor performance for targeted HER2 breast cancer detection by using
[128] A. Jing, Q. Xu, W. Feng, G. Liang, An electrochemical immunosensor for screen-printed electrodes modified with Au nanoparticles, Biomed. Micro-
sensitive detection of the tumor marker carcinoembryonic antigen (CEA) devices 23 (4) (2021) 46.
based on three-dimensional porous nanoplatinum/graphene, Micro- [149] A. Tabasi, A. Noorbakhsh, E. Sharifi, Reduced graphene oxide-chitosan-aptamer
machines 11 (7) (2020). interface as new platform for ultrasensitive detection of human epidermal
[129] M. Jozghorbani, M. Fathi, S.H. Kazemi, N. Alinejadian, Determination of growth factor receptor 2, Biosens. Bioelectron. 95 (2017) 117e123.
carcinoembryonic antigen as a tumor marker using a novel graphene-based [150] B. He, Differential pulse voltammetric assay for the carcinoembryonic anti-
label-free electrochemical immunosensor, Anal. Biochem. 613 (2021), gen using a glassy carbon electrode modified with layered molybdenum
114017. selenide, graphene, and gold nanoparticles, Microchim. Acta 184 (1) (2017)
[130] S. Rauf, G.K. Mishra, J. Azhar, R.K. Mishra, K.Y. Goud, M.A.H. Nawaz, J.L. Marty, 229e235.
A. Hayat, Carboxylic group riched graphene oxide based disposable elec- [151] Z. Si, B. Xie, Z. Chen, C. Tang, T. Li, M. Yang, Electrochemical aptasensor for
trochemical immunosensor for cancer biomarker detection, Anal. Biochem. the cancer biomarker CEA based on aptamer induced current due to for-
545 (2018) 13e19. mation of molybdophosphate, Microchim. Acta 184 (9) (2017) 3215e3221.
[131] N.A. Alarfaj, M.F. El-Tohamy, H. Oraby, New label-free ultrasensitive elec- [152] Z. Shekari, H.R. Zare, A. Falahati, Developing an impedimetric aptasensor for
trochemical immunosensor-based Au/MoS2/rGO nanocomposites for CA 27- selective labelefree detection of CEA as a cancer biomarker based on gold

35
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

nanoparticles loaded in functionalized mesoporous silica films, [175] Y. Zhou, Y. Wan, M. Yu, X. Yuan, C. Zhang, Hyaluronic acid-based label-free
J. Electrochem. Soc. 164 (13) (2017) B739eB745. electrochemical impedance analysis for cancer cell quantification and CD44
[153] W. Jiang, L. Liu, L. Zhang, Q. Guo, Y. Cui, M. Yang, Sensitive immunosensing of expression, Microchem. J. 160 (2021), 105622.
the carcinoembryonic antigen utilizing aptamer-based in-situ formation of a [176] R.C.B. Marques, E. Costa-Rama, S. Viswanathan, H.P.A. Nouws, A. Costa-
redox-active heteropolyacid and rolling circle amplification, Microchim. Acta García, C. Delerue-Matos, M.B. Gonz alez-García, Voltammetric immuno-
184 (12) (2017) 4757e4763. sensor for the simultaneous analysis of the breast cancer biomarkers CA 15-3
[154] X. Zhou, S. Guo, J. Gao, J. Zhao, S. Xue, W. Xu, Glucose oxidase-initiated and HER2-ECD, Sensor. Actuator. B Chem. 255 (2018) 918e925.
cascade catalysis for sensitive impedimetric aptasensor based on metal- [177] A. Valverde, V. Serafín, J. Garoz, A. Montero-Calle, A. Gonza lez-Corte
s,
organic frameworks functionalized with Pt nanoparticles and hemin/G- M. Arenas, J. Camps, R. Barderas, P. Y ~ ez-Seden
an ~ o, S. Campuzano,
quadruplex as mimicking peroxidases, Biosens. Bioelectron. 98 (2017) 83e90. J.M. Pingarro n, Electrochemical immunoplatform to improve the reliability
[155] M.A. Nawaz, S. Rauf, G. Catanante, M.H. Nawaz, G. Nunes, J.L. Marty, A. Hayat, of breast cancer diagnosis through the simultaneous determination of
One step assembly of thin films of carbon nanotubes on screen printed RANKL and TNF in serum, Sensor. Actuator. B Chem. 314 (2020), 128096.
interface for electrochemical aptasensing of breast cancer biomarker, Sen- [178] Q. Guo, X. Li, C. Shen, S. Zhang, H. Qi, T. Li, M. Yang, Electrochemical
sors 16 (10) (2016). immunoassay for the protein biomarker mucin 1 and for MCF-7 cancer cells
[156] S. Yang, F. Zhang, Q. Liang, Z. Wang, A three-dimensional graphene-based based on signal enhancement by silver nanoclusters, Microchim. Acta 182
ratiometric signal amplification aptasensor for MUC1 detection, Biosens. (7) (2015) 1483e1489.
Bioelectron. 120 (2018) 85e92. [179] S. Yazdanparast, A. Benvidi, M. Banaei, H. Nikukar, M.D. Tezerjani,
[157] H. Wang, J. Sun, L. Lu, X. Yang, J. Xia, F. Zhang, Z. Wang, Competitive elec- M. Azimzadeh, Dual-aptamer based electrochemical sandwich biosensor for
trochemical aptasensor based on a cDNA-ferrocene/MXene probe for MCF-7 human breast cancer cells using silver nanoparticle labels and a pol-
detection of breast cancer marker Mucin1, Anal. Chim. Acta 1094 (2020) y(glutamic acid)/MWNT nanocomposite, Microchim. Acta 185 (9) (2018) 405.
18e25. [180] Z. Shekari, H.R. Zare, A. Falahati, Dual assaying of breast cancer biomarkers
[158] R. Ahirwar, A. Dalal, J.G. Sharma, B.K. Yadav, P. Nahar, A. Kumar, S. Kumar, An by using a sandwichetype electrochemical aptasensor based on a gold
aptasensor for rapid and sensitive detection of estrogen receptor alpha in nanoparticlese3D graphene hydrogel nanocomposite and redox probes
human breast cancer, Biotechnol. Bioeng. 116 (1) (2019) 227e233. labeled aptamers, Sensor. Actuator. B Chem. 332 (2021), 129515.
[159] H.A. Samie, M. Arvand, Label-free electrochemical aptasensor for proges- [181] C. Pothipor, S. Bamrungsap, J. Jakmunee, K. Ounnunkad, A gold nanoparticle-
terone detection in biological fluids, Bioelectrochemistry 133 (2020), dye/poly(3-aminobenzylamine)/two dimensional MoSe2/graphene oxide
107489. electrode towards label-free electrochemical biosensor for simultaneous
[160] Y. Park, M.S. Hong, W.H. Lee, J.G. Kim, K. Kim, Highly sensitive electro- dual-mode detection of cancer antigen 15-3 and microRNA-21, Collo. Surf. B
chemical aptasensor for detecting the VEGF(165) tumor marker with PANI/ Biointer. 210 (2022), 112260.
CNT nanocomposites, Biosens. (Basel) 11 (4) (2021). [182] M. Sharifi, A. Hasan, F. Attar, A. Taghizadeh, M. Falahati, Development of
[161] M. Shamsipur, L. Farzin, M. Amouzadeh Tabrizi, F. Molaabasi, Highly sensi- point-of-care nanobiosensors for breast cancers diagnosis, Talanta 217
tive label free electrochemical detection of VGEF165 tumor marker based on (2020), 121091.
“signal off” and “signal on” strategies using an anti-VEGF165 aptamer [183] P. Balraj, A. Khoo, L. Volpi, J. Tan, S. Nair, H. Abdullah, Mutation Analysis of
immobilized BSA-gold nanoclusters/ionic liquid/glassy carbon electrode, the BRCA1 Gene in Malaysian Breast Cancer Patients, 2002.
Biosens. Bioelectron. 74 (2015) 369e375. [184] L.A. Salas, S.N. Lundgren, E.P. Browne, E.C. Punska, D.L. Anderton,
[162] X.-M. Fu, Z.-J. Liu, S.-X. Cai, Y.-P. Zhao, D.-Z. Wu, C.-Y. Li, J.-H. Chen, Elec- M.R. Karagas, K.F. Arcaro, B.C. Christensen, Prediagnostic breast milk DNA
trochemical aptasensor for the detection of vascular endothelial growth methylation alterations in women who develop breast cancer, Hum. Mol.
factor (VEGF) based on DNA-templated Ag/Pt bimetallic nanoclusters, Chin. Genet. 29 (4) (2020) 662e673.
Chem. Lett. 27 (6) (2016) 920e926. [185] N.J. Ronkainen, S.L. Okon, Nanomaterial-based electrochemical immuno-
[163] A. Bagheri Hashkavayi, B.S. Cha, S.H. Hwang, J. Kim, K.S. Park, Highly sensitive sensors for clinically significant biomarkers, Mater. (Basel, Switzerland) 7 (6)
electrochemical detection of circulating EpCAM-positive tumor cells using a (2014) 4669e4709.
dual signal amplification strategy, Sensor. Actuator. B Chem. 343 (2021), [186] M. El Aamri, G. Yammouri, H. Mohammadi, A. Amine, H. Korri-Youssoufi,
130087. Electrochemical biosensors for detection of MicroRNA as a cancer biomarker:
[164] R. Zhang, Q. You, M. Cheng, M. Ge, Q. Mei, L. Yang, W.-F. Dong, Z. Chang, pros and cons, Biosens. (Basel) 10 (11) (2020).
Multifunctional gold nano-cytosensor with quick capture, electrochemical [187] M. Negrini, M. Ferracin, S. Sabbioni, C.M. Croce, MicroRNAs in human cancer:
detection, and non-invasive release of circulating tumor cells for early cancer from research to therapy, J. Cell Sci. 120 (Pt 11) (2007) 1833e1840.
treatment, Front. Bioeng. Biotechnol. 9 (2021), 783661-783661. [188] X. Tang, G. Tang, S. Ozcan, Role of microRNAs in diabetes, Biochim. Biophys.
[165] M. Amouzadeh Tabrizi, M. Shamsipur, R. Saber, S. Sarkar, N. Zolfaghari, An Acta 1779 (11) (2008) 697e701.
ultrasensitive sandwich-type electrochemical immunosensor for the deter- [189] P. Miao, B. Wang, Z. Yu, J. Zhao, Y. Tang, Ultrasensitive electrochemical
mination of SKBR-3 breast cancer cell using rGO-TPA/FeHCFnano labeled detection of microRNA with star trigon structure and endonuclease medi-
Anti-HCT as a signal tag, Sensor. Actuator. B Chem. 243 (2017) 823e830. ated signal amplification, Biosens. Bioelectron. 63 (2015) 365e370.
[166] Z. Rahimzadeh, S.M. Naghib, E. Askari, F. Molaabasi, A. Sadr, Y. Zare, [190] S. Li, X. Yang, J. Yang, J. Zhen, D. Zhang, Serum microRNA-21 as a potential
M. Afsharpad, K.Y. Rhee, A rapid nanobiosensing platform based on diagnostic biomarker for breast cancer: a systematic review and meta-
herceptin-conjugated graphene for ultrasensitive detection of circulating analysis, Clin. Exp. Med. 16 (1) (2016) 29e35.
tumor cells in early breast cancer, Nanotechnol. Rev. 10 (1) (2021) 744e753. [191] M. Zubair, S. Wang, N. Ali, Advanced Approaches to Breast Cancer Classifi-
[167] L. Tian, J. Qi, K. Qian, O. Oderinde, Y. Cai, C. Yao, W. Song, Y. Wang, An ul- cation and Diagnosis, 2021, p. 11.
trasensitive electrochemical cytosensor based on the magnetic field assisted [192] F.S. Felix, L. Angnes, Electrochemical immunosensors - a powerful tool for
binanozymes synergistic catalysis of Fe3O4 nanozyme and reduced gra- analytical applications, Biosens. Bioelectron. 102 (2018) 470e478.
phene oxide/molybdenum disulfide nanozyme, Sensor. Actuator. B Chem. [193] S.A. Lim, M.U. Ahmed, CHAPTER 1 introduction to immunosensors, immu-
260 (2018) 676e684. nosensors, Royal Soc. Chem. (2019) 1e20.
[168] L. Tian, J. Qi, K. Qian, O. Oderinde, Q. Liu, C. Yao, W. Song, Y. Wang, Copper (II) [194] M.J. Duffy, D. Evoy, E.W. McDermott, CA 15-3: uses and limitation as a
oxide nanozyme based electrochemical cytosensor for high sensitive biomarker for breast cancer, Clin. Chim. Acta 411 (23e24) (2010)
detection of circulating tumor cells in breast cancer, J. Electroanal. Chem. 812 1869e1874.
(2018) 1e9. [195] C. Tse, A.-S. Gauchez, W. Jacot, P.-J. Lamy, HER2 shedding and serum HER2
[169] Y. Yang, Y. Fu, H. Su, L. Mao, M. Chen, Sensitive detection of MCF-7 human extracellular domain: biology and clinical utility in breast cancer, Cancer
breast cancer cells by using a novel DNA-labeled sandwich electrochemical Treat Rev. 38 (2) (2012) 133e142.
biosensor, Biosens. Bioelectron. 122 (2018) 175e182. [196] V.V. Levenson, Biomarkers for early detection of breast cancer: what, when,
[170] N. Liu, J. Song, Y. Lu, J.J. Davis, F. Gao, X. Luo, Electrochemical aptasensor for and where? Biochim. Biophys. Acta 1770 (6) (2007) 847e856.
ultralow fouling cancer cell quantification in complex biological media based [197] D.W. Kufe, Mucins in cancer: function, prognosis and therapy, Nat. Rev.
on designed branched peptides, Anal. Chem. 91 (13) (2019) 8334e8340. Cancer 9 (12) (2009) 874e885.
[171] M. Safavipour, M. Kharaziha, E. Amjadi, F. Karimzadeh, A. Allafchian, TiO2 [198] B. Bohunicky, S.A. Mousa, Biosensors: the new wave in cancer diagnosis,
nanotubes/reduced GO nanoparticles for sensitive detection of breast cancer Nanotechnol. Sci. Appl. 4 (2011) 1.
cells and photothermal performance, Talanta 208 (2020), 120369. [199] J. Wu, Z. Fu, F. Yan, H. Ju, Biomedical and clinical applications of immuno-
[172] F. Shafiei, R.S. Saberi, M.A. Mehrgardi, A label-free electrochemical apta- assays and immunosensors for tumor markers, TrAC, Trends Anal. Chem. 26
sensor for breast cancer cell detection based on a reduced graphene oxide- (7) (2007) 679e688.
chitosan-gold nanoparticle composite, Bioelectrochemistry 140 (2021), [200] M.-H. Schlageter, J. Larghero, B. Cassinat, M.-E. Toubert, C. Borschneck, J.-
107807. D. Rain, Serum carcinoembryonic antigen, cancer antigen 125, cancer antigen
[173] J. Quan, Y. Wang, J. Zhang, K. Huang, X. Wang, H. Jiang, Aptamer embedded 15-3, squamous cell carcinoma, and tumor-associated trypsin inhibitor con-
arch-cruciform DNA assemblies on 2-D VS(2) scaffolds for sensitive detection centrations during healthy pregnancy, Clin. Chem. 44 (9) (1998) 1995e1998.
of breast cancer cells, Biosensors (Basel) 11 (10) (2021). [201] A. Ravelli, J.M. Reuben, F. Lanza, S. Anfossi, M.R. Cappelletti, L. Zanotti,
[174] Y.H. Tang, H.C. Lin, C.L. Lai, P.Y. Chen, C.H. Lai, Mannosyl electrochemical A. Gobbi, C. Senti, P. Brambilla, M. Milani, Breast cancer circulating bio-
impedance cytosensor for label-free MDA-MB-231 cancer cell detection, markers: advantages, drawbacks, and new insights, Tumor Biol. 36 (9)
Biosens. Bioelectron. 116 (2018) 100e107. (2015) 6653e6665.

36
P. Lakhera, V. Chaudhary, A. Jha et al. Materials Today Chemistry 26 (2022) 101129

[202] J. Li, S. Li, C.F. Yang, Electrochemical biosensors for cancer biomarker [209] Z. Li, M.A. Mohamed, A.M. Vinu Mohan, Z. Zhu, V. Sharma, G.K. Mishra,
detection, Electroanalysis 24 (12) (2012) 2213e2229. R.K. Mishra, Application of electrochemical aptasensors toward clinical di-
[203] T. Meyer, G.J. Rustin, Role of tumour markers in monitoring epithelial agnostics, Food Env. Monitor.: Rev. Sens. (Basel) 19 (24) (2019).
ovarian cancer, Br. J. Cancer 82 (9) (2000) 1535e1538. [210] R. Abd-Ellatief, M.R. Abd-Ellatief, Electrochemical aptasensors: current
[204] P. De Cremoux, C. Tran-Perennou, B. Brockdorff, E. Boudou, H. Magdel, status and future perspectives, Diagnostics (Basel, Switzerland) 11 (1)
A. Lykkesfeldt, Validation of real-time RT-PCR for analysis of human breast (2021).
cancer cell lines resistant or sensitive to treatment with antiestrogens, [211] J. Welsh, Chapter 40 - animal models for studying prevention and treatment
Endocr. Relat. Cancer 10 (3) (2003) 409e418. of breast cancer, in: P.M. Conn (Ed.), Animal Models for the Study of Human
[205] E.M. Cormier, M.F. Wolf, V.C. Jordan, Decrease in estradiol-stimulated pro- Disease, Academic Press, Boston, 2013, pp. 997e1018.
gesterone receptor production in MCF-7 cells by epidermal growth factor [212] S. Menon, M.R. Mathew, S. Sam, K. Keerthi, K.G. Kumar, Recent advances and
and possible clinical implication for paracrine-regulated breast cancer challenges in electrochemical biosensors for emerging and re-emerging in-
growth, Cancer Res. 49 (3) (1989) 576e580. fectious diseases, J. Electroanal. Chem. 878 (2020), 114596.
[206] B. Bozal-Palabiyik, M. Lettieri, B. Uslu, G. Marrazza, Electrochemical detection [213] P.H. Lin, B.R. Li, Antifouling strategies in advanced electrochemical sensors
of vascular endothelial growth factor by molecularly imprinted polymer, and biosensors, Analyst 145 (4) (2020) 1110e1120.
Electroanalysis 31 (8) (2019) 1458e1464. [214] M.J. Russo, M. Han, P.E. Desroches, C.S. Manasa, J. Dennaoui, A.F. Quigley,
[207] M. Shahdordizadeh, R. Yazdian-Robati, M. Ramezani, K. Abnous, R.M.I. Kapsa, S.E. Moulton, R.M. Guijt, G.W. Greene, S.M. Silva, Antifouling
S.M. Taghdisi, Aptamer application in targeted delivery systems for diagnosis strategies for electrochemical biosensing: mechanisms and performance
and treatment of breast cancer, J. Mater. Chem. B 4 (48) (2016) 7766e7778. toward point of care based diagnostic applications, ACS Sens. 6 (4) (2021)
[208] D. Ou, D. Sun, X. Lin, Z. Liang, Y. Zhong, Z. Chen, A dual-aptamer-based biosensor 1482e1507.
for specific detection of breast cancer biomarker HER2 via flower-like nano- [215] J. Xu, H. Lee, Anti-biofouling strategies for long-term continuous use of
zymes and DNA nanostructures, J. Mater. Chem. B 7 (23) (2019) 3661e3669. implantable biosensors, Chemosens. 8 (3) (2020).

37

You might also like