You are on page 1of 14

12 (2022) 100259

Contents lists available at ScienceDirect

Biosensors and Bioelectronics: X


journal homepage: www.journals.elsevier.com/biosensors-and-bioelectronics-x

Peptide-based optical biosensors: A promising approach for early-stage


cancer detection
Gurpreet K. Soni , Saima , Priya Manhas , Rohit K. Sharma *
Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Sector-14, Chandigarh, 160014, India

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

Keywords: Cancer is the leading cause of 10 million deaths worldwide in 2020, which is exponentially increasing per year.
Optical cancer detection The early detection and effective treatment of cancer can save millions of lives worldwide. Cancer diagnostic
Peptide nucleic acid methods are scarce due to their high cost, laborious instrumentation, high maintenance, time-consuming, and
Targeting peptide
complex nature. Furthermore, current cancer detection methods are also inconvenient and necessitate the use of
Antifouling peptide
Self-assembled peptide
technical experts. However, peptide-based optical biosensors are fast, and compact with a low detection limit and
Cell-penetrating peptide high selectivity, hence having a lot of potential for diagnosing different types of cancer. The present work aims to
provide an overview of various types of peptide-based optical biosensors (OBS) for the early-stage detection of
cancer. Here, the peptides with high specificity, biocompatibility, small size, and numerous conjugation possi­
bilities incorporated with nanomaterials, are reported for the fabrication of optical biosensors to diagnose cancer.
Peptide-based optical biosensors are known to overcome the limitations of the conventional method by providing
high sensitivity, and portability, with rapid and real-time detection of all types of cancer cells. Moreover, in
comparison to conventional methods, cancer can be detected in a quite few million tumor tissues using optical
biosensors. This review mainly focused on the optical biosensors such as colorimetric, fluorescence, surface
plasmon resonance (SPR), chemiluminescence (CL), and photoelectrochemical derived from the different types of
peptides, including their working principle and conjugation with peptides. We have also discussed the advan­
tages and disadvantages of current peptide-based optical biosensors for early-stage cancer detection, as well as
future prospects.

1. Introduction mammography, sonography, polymerase chain reaction, and


enzyme-linked immunosorbent assays (Kaur et al., 2022). These tech­
Cancer is the second most lethal disease causes the death of 10 niques lack behind due to the laborious instruments, inexperienced staff,
million people worldwide in 2020 (Sung et al., 2021). The death rate high cost, complex setup, painful treatment, high maintenance, high
will be expected to reach 12 million by the year 2030 (World Health sample volume (blood, fluids), and long-time procedural duration
Organization, Fact sheet, 2022). However, it has also been reported that (Mittal et al., 2017). The limitations of these techniques open a new
30–50% of human lives can be saved by the early-stage detection, opportunity for researchers to develop new methods for cancer detec­
diagnosis, and treatment of cancer (Office for National Statistics UK, tion at the initial stage with high sensitivity, low cost, fast response, and
2016). The early-stage detection mainly focuses on the detection of user-friendly nature.
cancer/premalignant at the initial phase, which could increase the Biosensors are integrated receptor-transducer devices, which include
survival rate or morbidity reduction. The effectiveness of early diagnosis the receptor in conjugation with a transducer (physical/chemical) and
can be seen in the ratio of late detection of cervical cancer versus early detector. The receptor part embraces the biological recognition elements
detection reported to be 19.8/3.1 per 100,000 (Crosby et al., 2022). The such as peptides, nucleic acids, antibodies, enzymes, protein, whole
non-recognition ability of the human body toward cancer and less cells, and tissue slices to interact with the analyte (Askim and Suslick,
concentration of biomarkers is a great hurdle in its early detection. The 2017). Depending on the concentration of the analyte (biomarker),
available standard methods for cancer detection include magnetic transducer produces a signal which is quantified by the detector. Bio­
resonance imaging (MRI), molecular breast imaging, tissue biopsy, sensors can be classified into various categories based on the signal

* Corresponding author.
E-mail address: rohitksg@pu.ac.in (R.K. Sharma).

https://doi.org/10.1016/j.biosx.2022.100259
Received 15 May 2022; Received in revised form 8 September 2022; Accepted 22 September 2022
Available online 6 October 2022
2590-1370/© 2022 Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

transducer - optical, optoelectronic, electrochemical, thermometric, The current review article embraces the fabrication and application
magnetic, and piezoelectric (Farré and Barceló, 2007). The utilization of of peptide based optical biosensors for early-stage cancer diagnosis,
optical transducers and biological recognition elements in one probe especially in the last decade. The optical biosensing of cancer bio­
originate the new class of biosensors called optical biosensors. Optical markers has been divided into five parts depending upon the nature and
biosensors are a non-invasive replacement for available biosensing application of peptides - (i) peptide nucleic acid (PNA) based detection
techniques for cancer detection. They offer remarkable advantages over (ii) targeting peptide based detection (iii) antifouling peptide based
electrochemical and other biosensors such as high sensitivity and detection (iv) self-assembled peptide based detection (iv) cell pene­
selectivity, multiple component detection, lower signal-to-noise ratio, trating peptide (CPP) based detection (Fig. 1a). The sensing mechanism
lower immunity to electromagnetic interference, ease in fabrication, of various optical biosensors such as fluorescence, colorimetric, surface
smaller size, direct label free and real time detection (Sołoducho and plasmonic resonance, chemiluminescence, and photo electrochemical is
Cabaj, 2015). Moreover, signal transduction is independent of the target methodized with the usage of a particular peptide. Close attention has
in them and can easily adaptable to the environment. These sensors been given to the recently developed user-friendly biosensors for cancer
provide relevant results in the case of single molecular determination detection. At last, we summarized the loopholes and future direction to
and distant sensing. Also, the incorporation of multidisciplinary fields develop optical biosensors in the field of cancer.
including nano/micro technology, chemistry, molecular biology, mi­
croelectronics, and biotechnology results in the fabrication of optical 2. Peptide based optical biosensors for early-stage cancer
biosensor which provides a reliable device for healthcare (Chen et al., detection
2019). Optical biosensors for cancer basically detect the biomarkers
such as micro ribonucleic acid (miRNA), circulating tumor cells (CTC), Nowadays, peptides are becoming nearly an ideal candidate for
exosomes, proteins, and deoxyribonucleic acid (DNA), which can be fabricating selective and sensitive optical biosensors due to their various
collected from plasma, blood, serum, saliva, and urine (Badr et al., 2014; advantages over nucleic acids, antibodies, and enzymes. A peptide with
Underwood et al., 2020). antifouling, self-assembly, cell penetrating, and target recognition
Over the last decade, optical sensors for cancer detection mainly properties conjugate with an optical probe for the fabrication of peptide
comprise the recognition method, based on antibodies and antigens based optical sensors. The conjugation of peptides can be carried out
(Holford et al., 2012). Although antibodies possess high specificity but before and after the synthesis of optical probe (nanoparticles (NPs),
high production costs, non-availability of synthetic production methods, quantum dots (QDs), and carbon dots (CDs)) (Valetti et al., 2014). The
limited methods for modification, and narrow self-life, restrict their single-step peptide conjugation prior to the formation of the optical
applications in cancer detection (Urbanek et al., 2011) Therefore, arti­ probe shows advantages over other processes in terms of high quantum
ficial molecules such as aptamer and peptide based optical sensors yield, maintenance of peptide activity, and uniform particle size. The
dominating the cancer market in recent years (J. J. Zhang et al., 2017, variety of techniques used for peptide immobilization includes electro­
Roberts and Gandhi, 2022). Aptamers are oligonucleotides with a single static binding, physical adsorption, covalent binding, and specific
strand (ssDNA/RNA) that can preferentially bind to a particular target recognition (Rajchakit and Sarojini, 2017). Peptides with positively or
via the formation of defined architectures. Whereas, peptides are short negatively charged amino acid side chains can adsorb electrostatically
polymers in which amino acids are bonded together via an amide bond. on nanoparticles capped with anionic ligands (citrate, mercaptopro­
Depending upon the nature, number, and sequence of amino acids pionic acid, lipoic acid) or cationic ligands (cysteamine) respectively
peptides show distinctive properties such as high specificity and selec­ (Katz and Willner, 2014). Peptides with phenylalanine can also adsorb
tivity, cell penetration, low toxicity, easy synthesis, and surface modi­ on the surface of nanoparticles via self assembly through the formation
fication. These synthetic materials (aptamer and peptide) efficiently of a specific architecture. The most common approach for peptide
replaced antibodies by taking the advantage of additional features such conjugation to the metallic nanoparticles involves the covalent bonding
as high stability, high specificity, low-cost easy synthesis, and modifi­ via the formation of Au–S bond where the cysteine containing peptides
cation (Liu et al., 2018). Targeting peptides and aptamers with biolog­ can directly synthesize the gold nanoparticles (AuNPs)/silver nano­
ical recognition ability can target the tumor cells and tissues through particles (AgNPs) via the formation of a metal thiolate bond (Jazayeri
outstanding ability with respect to target biomarkers. Peptide nucleic et al., 2019). The second covalent approach is based on click chemistry
acids (PNAs) are another class of peptides that mimic the DNA/RNA by where copper(I)-catalyzed alkyne-azide cycloaddition (CuAAC) can be
replacing the deoxyribose/ribose phosphate backbone with pseudo used for peptide conjugation (Lutz and Zarafshani, 2008). Another
peptide-N-(2-aminoethyl)-glycine (Corkill and Rapley, 2008). The alternative way involves the utilization of suitable bifunctional linkers
non-existence of deoxyribose/ribose sugar in PNAs makes them highly with additional features such as control over the peptide orientation of
stable toward enzymatic degradation in a biological environment. The the nanoparticle surface. The bifunctional linkers (polyethylene glycol
replacement of phosphate backbone with N-(2-aminoethyl)-glycine (PEG), ethyl(dimethylaminopropyl)carbodiimide / N-hydrox­
build stable PNA/DNA and PNA/RNA hybridized pairs, as compared to ysuccinimide (EDC/NHS)) have a thiol group at one side, and carboxylic
DNA/RNA due to the reduction of electrostatic interaction (IgIoi, 1998). acid group on another side (Casciaro et al., 2017). Here, the thiol group
PNA’s unique properties make it superior to aptamers/nucleic acids in helps in the formation of Au–S where the –COOH group is used in the
cancer detection. Moreover, peptide based sensors have several advan­ coupling with the –NH2 group of peptides. Among all covalent conju­
tages over their larger analogues such as enzymes and proteins. Enzy­ gation maintain the properties of peptides along with toxicity minimi­
me/protein based sensors suffer from limited lifetime, high cost, zation of nanoparticles which makes it advantageous over other
complex synthesis methods, activity dependence over pH, inhibitors, conjugation methods.
ionic strength, and temperature which are smoothly overcome by pep­ Optical biosensor is an analytical device incorporating a receptor
tide based sensors (Economou et al., 2017; Shahdeo et al., 2022). Besides (peptides, antibodies, nucleic acids, antibodies, etc.) optical transducer,
this, the problem of low response and low sensitivity with whole cell and detector (Damborský et al., 2016). The transducer part comprises
biosensors can also be solved by this new class of biosensors (Chen et al., optical probes such as AuNPs, AgNPs, QDs, CDs, etc. which are further
2017). Additionally, peptides exhibit special features like antifouling integrated with the receptor. The basic sensing mechanism of optical
property, self-assembly, and cell penetration depending upon the biosensors is based on the alteration of the characteristic signal of the
sequence and nature of amino acids (Wang et al., 2021). The ability of optical probe in the presence of an analyte (Seitz, 1989). The in­
peptides to exhibit numerous properties and advantages over already teractions of receptor moieties with analyte initiate the various bio­
reported methods motivates the authors to gather peptide based optical recognition events such as hybridization, protein/peptide binding,
biosensors in this review article, particularly for early cancer detection. metabolic reaction, etc. which produces the physical or chemical change

2
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

Fig. 1. (a) Peptide based optical biosensors for early-stage cancer detection. (b) Schematic representation for the working principle of optical sensors.

in the system. These changes are then converted to optical/electronic process to create an excited state for luminescent signals (Ma et al.,
signals by the transducer and detected by the detector. The optical 2020). On the other hand, photoelectrochemical (PEC) is opposite to
biosensors can be categorized on the basis of various optical principles ECL which utilizes the light for the excitation of electrons and photo­
such as SPR, absorbance, fluorescence, chemiluminescence, photo­ current to observe signals. The photoelectrochemical sensors work on
luminescence, refractive index, surface-enhanced Raman spectroscopy the change in photocurrent/potential caused by the interactions be­
(SERS), scattering, reflectance, diffraction, etc. covering the UV–visible, tween photoelectrochemical material and target moieties (Shu, and
infrared (IR) and near-infrared (NIR) region of electromagnetic spectra Tang, 2020). Both methods are equally sensitive for the detection of
(Fig. 1b) (Jerónimo et al., 2007; Law et al., 2020). various analytes by reducing the background noise. Based on all these
This review article mainly focuses on the SPR, colorimetric, fluori­ methods, peptide based optical biosensors have been discussed
metric, chemiluminescence, and photoelectrochemical (PE) based op­ depending on the nature and function of the peptide.
tical biosensors. Optical sensors derived from the excitation of surface
plasmons are assigned as SPR sensors (Homola, 2008). SPR phenomena 2.1. Peptide nucleic acid based optical sensors
have been widely employed in the fabrication of biosensors and che­
mosensors. The collective/coherent oscillation of free conduction elec­ Peptide nucleic acids are modified DNA/RNA analogues in which
trons in metal nanoparticles referred to as SPR is categorized into negatively charged phosphate backbone is replaced by neutral N-2-
localized surface plasmon resonance (LSPR) and propagating surface aminoethylglycine repeating units, introduced in 1990 by biochemist
plasmon resonance (PSPR) (Chen and Ming 2012). The LSPR phenom­ Peter Nielsen and organic chemist Ole Buchardt (Singh et al., 2010;
ena is observed in metallic nanoparticles whereas, PSPR is mainly seen Saadati et al., 2019). Considering the superior properties of PNA such as
in metallic films and optical fibers. The enhancement of electromagnetic high chemical stability, easy chemical modification, resistance towards
field by both phenomena is leading to the application of SPR in refrac­ enzyme degradation, stable hybrid complex formation (DNA/PNA,
tive index measurement (Zeng et al., 2005), fluorescence enhancement RNA/PNA, PNA/DNA/PNA) with high melting point and binding en­
(Sokolov et al., 1998), absorbance (Su et al., 2007), SERS (Chen and ergy, high specificity for nucleic acids, stand invasion and extraordinary
Burstein, 1980), etc. Among all these, fluorescence has become the ionic strength, it is widely used for the development of sensors, chips,
utmost used optical technique for the detection of cancer due to its and arrays (Shi et al., 2015). The immobilization of PNA on optical
excellent sensitivity as compared to other optical techniques. Fluores­ converters opens a new path for sensitive and selective determination of
cence occurs when molecules excite by incident light followed by the various cancers (Gupta et al., 2017). A colorimetric sensor based on
emission of photons from a single excited state to the ground state (Shin PNA-gold nanoparticles (PNA-AuNPs) has been developed to detect two
et al., 2021). Various parameters of fluorescence such as fluorescence mutations in exon 11 of c-Kit (Lee et al., 2011). The reported method
intensity, fluorescence decay, high quantum yield, anisotropy, excita­ provides the selective and sensitive (lower limit of detection (LOD) 500
tion, and emission spectra provide great help in data analysis (Li et al., fM) detection of specific mutations by avoiding the uncontrolled ag­
2019). Contradictory to this, colorimetric is an absorption-based oldest gregation of PNA-AuNPs caused by electrostatic interactions. Corre­
analytical method used for the naked-eye detection of various analytes. spondingly, naked eye detection of human c-Myc mRNA in colon tumor
This straightforward technique can simplify the data analysis by eval­ was performed by controlled aggregation of AgNPs by PNA/PNA-RNA
uating the UV–Vis peak with high absorbance and spectral analysis at triplex with a sensitivity of 50 nM (Li et al., 2016). In response to
200–800 nm (Liu et al., 2020). Divergent to all above mentioned tech­ c-Myc mRNA, the AgNPs-PNA platform showed color change based on a
niques, electrochemiluminescence (ECL) is a prominent chem­ single base mismatch and provides visual detection. In another report,
iluminescence technique based on the production of ECL luminophore at seven mutations in codon 12 and 13 of the Kisten rat sarcoma viral
the surface of the electrode under applied voltage (Babamiria et al., oncogene homologue (KRAS) gene were validated with the utilization of
2019). The produced luminophore undergoes an electron transfer AuNPs and PNAs for colorectal cancer (CRC) (Zhao and Lin, 2017). The

3
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

rapid, label free colorimetric method was based on the aggregation of detection was reported by the incorporation of the smartphone (Naor­
AuNPs in the presence of PNA which was further restricted by hybrid ungroj et al., 2021). The reported method provided the quantification of
complex formation between PNA and target DNA. A distinctive Human papillomavirus (HPV) DNA by examining the color change in
approach illustrated the modification of gold nanorods (AuNRs) by PNA, AuNPs induced via pyrrolidinyl peptide nucleic acid (acpcPNA).
complementary to the target DNA for colorimetric sensing of point Another excellent class of optical biosensors belongs to fluorescence
mutations in the KRAS gene due to pancreatic ductal adenocarcinoma in which fluorescently modified PNAs are widely employed in the bio­
(Tadimety et al., 2019). The study successfully differentiated between sensing and bioimaging of cancer cells. A disposable fluorescent
the wild type and mutant sequence of KRAS with a sensitivity of 10 biosensor was reported for early detection of lung cancer based on PNA
ng/mL in the serum sample. In another report, homopurine PNA was and poly(3-alkoxy-4-methylthiophene) as a fluorescent probe (Yildiz
efficiently bonded to homopyrimidine DNA and PNA-DNA/DNA triplex et al., 2013). The positively charged fluorescent probe was fabricated on
which can be further conjugated with cyanine dye (DiSC2) to provide poly(vinylidene fluoride) thin paper, which further corroborated with
color variation (Xu et al., 2020). This system helped in designing of PNA and target miRNA. Based on the formation of a hybrid complex
colorimetric sensing probe for micro-RNA for cancer detection. The between reporter-miRNA/PNA and reporter-miRNA, two types of sig­
advancement in PNA-based colorimetric sensors for cervical cancer nals are produced which led to quantitative and qualitative detection. In

Fig. 2. (a) Schematic representation for tumor cell selective fluorescence detection based on ATP aptamer and PNA/peptide/PNA copolymer. (b) The comparison of
protease activatable aptamer probe (PA-SP) with conventional probe (C-SP) shows the high cell selectivity. Reprint with permission from (Xiang et al., 2021). ©2021
John Wiley & Sons, Inc.

4
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

another report, point mutations in the KRAS oncogene was detected linker in the presence of Ru2+-catalyst to expose rhodamine for miRNA
utilizing the two types of molecular beacons (i) thiazole orange PNA detection was reported (Sadhu and Winssinger, 2013). PNA act as a
molecular beacon (TO-PNA-MB), (ii) phosphothiolated DNA molecular template between the Ru2+-catalyst and enclosed rhodamine in the
beacon (PS-DNA-MB) (Kam et al., 2012). From these two systems, presence of target miRNA. Contradictory, fluorescent biosensor reported
TO-PNA-MB effectively differentiated KRAS and SNP in vitro and in vivo for prostate cancer based on the Michael addition reaction between two
to provide the sensitive and selective detection of KRAS mutations. PNA probes (Metcalf et al., 2016). A convenient method was developed
Similarly, the fluorescence probe from 15/16-mer PNA efficiently for EGFR mutated cancer cells based on fluorescence resonance energy
distinguished the mutant target and wild type HER2 based on the single transfer (FRET) phenomena regulated by fluorescein
base mismatch principle (Metaferia et al., 2013). During the synthesis, isothiocyanate-PNA (FITC-PNA) and dabcyl-DNA (Shigeto et al., 2019).
PNAs can be easily modified with dyes, for example, the modification of The fluorescent signal of FITC-PNA was quenched by dabcyl-DNA,
three different PNAs with three different fluorescent dyes carboxy which was further regained in the presence of target mRNA due to the
fluorescein (FAM)-PNA21, 6-carboxy-X-rhodamine (ROX)-PNA125b formation of the FITC-PNA/mRNA hybrid. Correspondingly, PNA based
and cyanine 5(Cy5)-PNA96 was reported (Ryoo et al., 2013). PNAs were FRET was reported between the gold nanoparticles and carbon dots for
complementary to miRNA (miR-21, miR-125b, and miR-96), providing the HeLa cell line (Gao et al., 2020). PNA induced the aggregation of
quantitative monitoring based on the quenching of dyes with nanosized AuNPs and restrict the FRET between AuNPs and CDs. When PNA/DNA
graphene oxide (NGO). A different type of approach was adopted for the complex formed AuNPs remained free in the solution leading to the
fluorescence detection of KRAS2 mRNA in lung cancer cells (Sonar et al., quenching of CDs by FRET. A bioluminescence resonance energy
2014). Fluorescence of single-stranded PNA-IGF1 tetrapeptide-thiol or­ transfer (BRET) phenomenon was employed for smartphone based
ange complex escalated upon interaction with KRAS2 RNA. Corre­ quantification of three miRNA sequences (miR-21, -31, -141) (Chang
spondingly, sensing of colorectal cancer was reported based on the et al., 2020). The work utilized the PNA for selective DNA detection
fluorescence enhancement of thiazole orange-peptide nucleic acid mo­ which further conjugated with luciferase to form H-Luc-PNA conjugate.
lecular beacons (TO-PNA-MB) (Kam et al., 2014). Thiazole Orange and The displacement of the PNA duplex was carried out by target RNA to
other cyanine dyes absorbed light near 500 nm, are highly sensitive to accomplish logic-gate operation (AND operation). An interesting probe
the local environment, and lead to a remarkable increase in signal/noise fabrication was carried out by coating the covalent complex (PNA-­
ratio upon binding with RNA/DNA. These types of spectral properties of photo-cleavage linker and alginate hydrogel) on a microneedle array
cyanine dyes are not appropriate for in vivo applications due to cell (Sulaiman et al., 2019) (Fig. 3a and b). Treatment of the fabricated
auto-fluorescence. To overcome this limitation, the use of Bis Quinoline microneedle with target DNA sequence was responsible for the forma­
(BisQ) and PNA conjugate to detect single point mutations in cancer tion of the PNA-DNA complex followed by the fluorescence quenching
cells was reported (Kolevzon et al., 2016). Similarly, a red-emitting and target detection (Fig. 3c and d). A distinctive approach was
BisQ-PNA probe was utilized for on-site fluorescent detection of employed based on PNA clamping-assisted fluorescence melting curve
mRNA in human cancer tissues (Hashoul et al., 2019). A completely analysis for the detection of non-small cell lung cancer (J.Y. Han et al.,
distinctive approach was demonstrated for tumor overexpressed prote­ 2016; Kim et al., 2017). This method effectively provided the prediction
ase based on PNA/peptide/PNA copolymer (Fig. 2.) (Xiang et al., 2021). of treatment response and mutation diagnosis for lung cancer in the
The structure switching activity of adenosine triphosphate (ATP) plasma sample.
aptamer was locked by PNA/peptide/PNA copolymer and selective A surface plasmon resonance phenomenon was used for the bio­
cleavage of peptide by protease reduced the hybridization of PNA/ap­ sensing of ctDNA (Bellassai et al., 2021). Sensor fabrication was carried
tamer initiating the tumor cell selective molecular detection. out by PNA complementary to target DNA, oligopeptide CEEEEE,
The single stranded PNAs show strong interactions with various AuNPs@KRAS, and poly-L-lysine based antifouling layer. Target DNA
types of nanomaterials and act as a base for several sensing platforms. captured by the PNA probe initiated the changes in the SPR signal. The
The covalently linked PNA with fluorescent nanodiamonds (NDs) using radiolabeled PNAs showed high potential for non-invasive molecular
EDC/NHS chemistry for the detection of lung cancer was reported, imaging of cancer cells. 64Cu-PNA-carboxyfluorescein/DOTA probe was
where the intrinsic luminescent properties of NDs-PNA complex successfully fabricated for non-invasive PET imaging of miR-146b-5p
quenched upon interaction with target DNA (Gaillard et al., 2014). The and miR-146a for lung and thyroid cancer detection (Croci et al., 2019).
quenching of fluorescent label PNA with nanosheets and molecular
organic frame works (MOF) is the basic model for the detection of 2.2. Targeting peptide-based optical sensors
several analytes. In this context, fluorescent dye labeled PNAs illustrated
for miRNA sensing using the nano metal-organic framework (NMOF, Over the past few years, significant interest has emerged in the
UiO-66) and carbon nitride nanosheets (Wu et al., 2015; Liao et al., development of targeting peptide-based sensors for the early identifi­
2015). A method for the conjugation of PNA on silicon oxide surface was cation and measurement of cancer (Karimzadeh et al., 2018). Antitumor
reported based on the click and thio-yne chemistry for early diagnosis of monoclonal antibodies have been widely studied as targeting agents for
cancer (Veerbeek et al., 2018). Thiol-PNA and azido-PNA were incor­ cancer detection (Martinelli et al., 2009). However, the use of antibodies
porated covalently on silicon surface via 1, 8-nonadyne as branching as a targeting biosensor is associated with some limitations. The anti­
moiety. Depending upon the target DNA quenching of fluorescent dyes tumor monoclonal antibodies have a large size and are non-specifically
took place, providing selective and sensitive detection. Based on the taken up by the liver and reticuloendothelial system (Aina et al., 2002).
conjugation of thiolated PNA with magnetic beads (MBs) multistep On contrary, the targeting peptide-based detectors have benefits over
fluorescent sensing strategies had been reported for lung cancer (Wang antibodies as they are relatively small-sized, less immunogenic, easy to
et al., 2020; D. Zhang et al., 2020). The initiator for atom transfer radical synthesize, exhibit high specificity, and excellent flexibility concerning
polymerization (ATRP) was fabricated on PNA/DNA hybrid carbox­ their conjugation potentials and hence are extensively used for
y/sulfonate-Zr4+-phosphate chemistry. Further, the fluorescent signal of biosensor construction (Wang et al., 2022). The targeting peptide-based
deposit fluorescein-o-acrylate (FA) is accelerated by countless polymer optical biosensors generally involve electrochemiluminescence, fluo­
chains. Similarly, a dual-recognition fluorescent probe for circulating rescence resonance energy transfer, and optical analytical methods. The
tumor DNA (ctDNA) was reported, where the PNA-MBs complex selec­ ECL method is highly sensitive, facile, and generally involves the ben­
tively distinguished the mutated ctDNA from wild type DNAs with a efits of both electrochemical and chemiluminescence methods, owing to
detection limit of 0.3161 pM (Chen et al., 2020). which, it has been widely explored for peptide-based biosensing in the
PNA directed templated reactions offer a fascinating approach to the early detection of cancer (Qi and Zhang, 2020). In this context, recently,
imaging and detection of DNA/RNA. The photocatalyzed cleavage of the a peptide-based electrochemiluminescence biosensor with graphitic

5
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

Fig. 3. (a) Schematic representation for the detection


of circulating nucleic acids in skin interstitial fluid
with the help of hydrogel-coated microneedle plat­
form, microneedle array coated with PNA, and algi­
nate hydrogel for target DNA. (b) Scanning electron
micrograph (SEM) of microneedle without and with
coating. (c) Schematic illustration of working princi­
ple of microneedle based array. (d) Swelling kinetics
of hydrogel coated microneedles. Reprinted with
permission from (Sulaiman et al., 2019). Copyright
(2019) American Chemical Society. (e) Pathway
illustration for the peptide-Lum-AuNPs@g-C3N4
nanoprobe preparation. (f) Schematic representation
for the construction of ECL biosensor for phosphati­
dylserine (PS)-positive exosomes. Reproduced with
permission of Elsevier with (Liu et al., 2021). Creative
Commons Attribution License (5293591498830).

phase carbon nitride nanosheet loaded with luminol capped gold detection limits of 5 ng/mL and 10 pg/mL, respectively.
nanoparticles (Lum-AuNPs@g-C3N4) was reported for the early diag­ Fluorescence resonance energy transfer is another important
nosis of ovarian malignancy through highly sensitive detection of analytical technique for cancer detection, utilizing fluorogenic peptide-
phosphatidylserine positive (PS (+)) exosomes (Fig. 3e and f) (Liu et al., based biosensors. FRET is observed when the emission spectrum of a
2021). donor (fluorophore) is overlapped by the absorption spectrum of an
Similarly, a unique electrogenerated chemiluminescence peptide- acceptor (quencher). In FRET the donor should be at a suitable distance
based biosensor (ECL-PB) has been reported for the specific determi­ from the acceptor in order to accommodate the distance-dependent
nation of prostate-specific antigen (PSA) tumor marker for the early fluorescence resonance energy transfer (Zadran et al., 2012). Recently,
detection and examination of recurrence of prostate cancer (Qi et al., a 5-carboxy-fluorescein (FAM)-labeled peptide containing Arg6 assem­
2014). The peptide with sequence (CHSSKLQK) had been employed as a bled with the negatively charged carbon nanoparticles (CNPs) was used
molecular recognition element based on its target-induced cleavage for convenient and highly sensitive detection of trypsin based on FRET
within Nafion films incorporated with gold nanoparticles and (Ru(bpy)3 phenomenon (Hou et al., 2020). The activation of trypsin is known to
2+
) as ECL emitting species. The reported method identified the PSA exceed Pancreatic secretory trypsin inhibitor (PSTI) control and acti­
with a 0.8 pg/mL detection limit. In another report, an ultra-sensitive vated other proteases for self-digestion, which can cause cell damage
and selective electrochemiluminescence assay was developed for the and may lead to pancreatic cancer. The reported method was simple and
highly efficient detection of cyclin A2 via ECL graphene-up conversion highly selective due to the specific cleavage of the six-arginine based
hybrid (Wu et al., 2014). Cyclin A2 is an effective indicator for synthetic peptide by trypsin and efficient to determine the trypsin ac­
early-stage cancer and the developed ECL biosensor can be potentially tivity, with the detection limit of about 0.7 μM. Similarly, a simple FRET
used for the screening of chemotherapeutic drugs and for early-stage sensing platform based on peptide-functionalized upconversion@silica
evaluation of treatments of cancers. Similarly, a selective and sensitive nanoparticle (UCNP@SiO2@Cy5-pep) had been developed for the in
biosensor based on ECL was reported for simultaneous detection of two vitro and in vivo detection of caspase-9 activity which is significant in
matrix metalloproteinases (MMPs) i.e., MMP-2 and MMP-7, associated diseases diagnosis and for the development of anti-cancer drugs (Nitin
with the growth of cancer (Gao et al., 2017). This multiplexed biosensor et al., 2009). In the fluorophore-conjugated peptide detectors,
was designed based on the co-immobilization of the Ir1 and Ru1 labeled peptide-based nanomaterials have been also explored owing to their
peptides on a gold electrode using self-assembly. The authors reported high affinity and specificity. In this context, HEKMs micelles were re­
that the sensor allowed the detection of MMP-2 and MMP-7 with ported, which consist of a peptide as a recognition element, a matrix

6
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

metalloproteinase-2 (MMP-2)-regulated FRET probe for NIR fluores­ colorectal cancer (Jones et al., 2018). Similarly, a water-soluble probe
cence imaging, a gadolinium probe (Gd) for MRI and the last element consisting of a cyclic peptide (GE-137 peptide) that binds to human
includes the mitochondrial damage segment KLA and the chemotherapy tyrosine kinase c-Met, conjugated to the fluorescent cyanine dye was
drug doxorubicin (DOX) for synergetic antitumor treatment (Wang investigated for the early detection of cancer (Burggraaf et al., 2015).
et al., 2019). Apart from FRET, metal-enhanced fluorescence (MEF) is The authors reported that the intravenous administration of GE-137
one of the evolving detection methods as here the emission intensity can cause-specific accumulation in c-Met expressed tumors. Similarly,
be considerably enhanced by positioning the fluorescent molecules near folate and FITC conjugated EC17 peptide was clinically studied for the
the metal nanostructure. Based on the MEF, a proteolytic enzyme specific intraoperative detection of folate receptor-α targeting ovarian
nanosensor was reported which comprised of AuNPs connected to both cancer using fluorescence imaging (Tummers et al., 2015). As a fluo­
single-strand DNA (ssDNA) and peptide (DEVD) (Fig. 4) (Choi et al., rescent contrast agent, the potential of epidermal growth factor (EGF)
2020). The reported nanosensor was recognized and cleaved by labeled with Alexa Fluor® 647 was evaluated to image epidermal
caspase-3, a proteolytic enzyme that is associated with apoptotic cells growth factor receptor (EGFR) expression in oral neoplasia using
and the diagnosis of cancers (Fig. 4a and b). Based on the immunoflu­ high-resolution fluorescence imaging (Nitin et al., 2009). The report
orescence assay the CSP3 (QQLPSS STSTYP) peptide was identified as an illustrated that the peptide-dye conjugate was efficiently and easily
effective targeting probe that can specifically and sensitively bound to entered in less than 30 min through the oral epithelium with a pene­
SiHa cells and serves as a potential candidate for diagnostic applications tration depth of more than 500 μm. Similarly, an epidermal growth
of cervical cancer (Li et al., 2017). Similarly, a peptide-templated gold factor conjugated with Cy5.5 dye was used as a specific NIR contrast
nanoparticle as a nanosensor was reported for rapid detection of mul­ agent to specifically bind the EGFR-positive tumors in mice (Ke et al.,
tiple post-translational modification (PTM) enzymes (J. Zhang et al., 2003). Correspondingly, the peptide sequence SNFYMPL was used as a
2020). For the early detection of pancreatic cancer, a peptide-22 con­ fluorescent contrast agent that enables specific binding to dysplasia in
jugated to the fluorescent dye cyanine 7 (peptide-22-Cy7) was reported Barrett’s esophagus (Li et al., 2010). The authors reported that the
for targeted imaging based on the high targeting toward the low-density fluorescence intensity was increased with the progression of tissue from
lipoprotein receptor (LDLR) which is usually overexpressed and normal to benign intestinal metaplasia to dysplasia. Also, hepsin, a
distributed over pancreatic cancer cells as biological target (Han et al., prostrate biomarker was detected using heparin targeting peptides via
2019). An AuNPs based biosensor was reported for the determination of phage display techniques (Kelly et al., 2008). The targeted peptides were
chymase activity in kidney tissue (Chang et al., 2018). In this study, an conjugated with fluorescent nanoparticles in order to improve the
AuNPs-based fluorescence peptide probe was constructed containing pharmacokinetics which displayed an efficient biomarker-based detec­
FITCAcp-DRVYIHPFHLDDDDDC peptide comprising an enzyme (chy­ tion of prostate cancer. Likewise, the peptide AADNAKTKSFPV (AAD)
mase) substrate (DRVYIHPFHL), a spacer (DDDDD), and cysteine (C) had been identified using phage display selection to guide tissue biopsy
was conjugated to 16 nm-AuNPs. The authors observed that the reported and for the early detection of gastric cancer (GC) (Zhang et al., 2012).
AuNPs-peptide probe required only 15 min for the detection of chymase Moreover, the reported peptide enables the distinction of the neoplastic
activity. In another report, cationized polyacrylamide (CPAA) and a from normal gastric mucosa. Based on the near-infrared fluorescence
synthetic pentadecapeptide, EPPT1 (YCAREPPTRTFAYWG) were stud­ optical imaging Cy5.5-conjugated mono-, di-, and tetrameric RGD pep­
ied for the detection of biomarkers, needed for the prevention and tides were assessed for tumor-targeting efficacy and to investigate the
efficient treatment of colorectal cancer (Bloch et al., 2012). It has been effect of multimerization on integrin avidity (Cheng et al., 2005). The
observed that fluorescence colonoscopy connected with a fluorescent authors reported that all the dye conjugated peptides are appropriate for
probe specific for a polyp biomarker can improve the early detection of integrin expression imaging and the tetramer showed better imaging
cancer. A tumor-targeting, NIR fluorescent peptide (LS301, sequence characteristics than monomer and dimer. Recently, the FITC-labeled
cypate-cyclo (cGRDSPC)K) was investigated for early detection and peptide (NH2-Lys-Gly-Pro-Arg-Ser-Leu-SerGly-Lys-FITC) conjugated to
endoscopic resection of polyps in a genetically engineered rat model of graphene oxide (GO) by non-covalent or covalent linkages were

Fig. 4. Schematic diagram for the detection of proteolytic enzyme based on MEF-caspase-3 biosensor. The restricted metal enhancement was activated by caspase-3
by increasing the distance between AuNPs and FITC. (a) Fluorescent spectrum of bifunctional AuNPs without ssDNA and (b) with ssDNA after proteolytic reaction by
caspase-3. Reprinted with permission from (Choi et al., 2020). Copyright (2020) American Chemical Society. (c) Description of the modification of the FO-SPR sensor
along with the schematic illustration for the detection of PDGF-BB. (d) Schematic representation for home-made FO-SPR biosensing detection system. Reproduced
with permission of Elsevier (Qian et al., 2019). with Creative Commons Attribution License (5293550401354).

7
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

reported as nanoprobes for MMP9 detection (Lin et al., 2018). In another sensor surface using cationic poly-L-Lysine (PLL) with anionic peptide
study, tetraphenylethylene (TPE) based peptide micelles have been CEEEEE to prevent nonspecific adsorption, whereas PNA was immobi­
studied for the monitoring of cellular apoptosis (Qin et al., 2021). In this lized for targeting ctDNA. Owing to the antifouling property of
study, a self-assemble amphiphilic polymer was constructed by linking a dual-layer and PNA KRAS p.G12D-mutated DNA can be detected with
Poly(histidine) chain polypeptide (H9) with LOD 2.5 aM in human plasma. Apart from these, multifunctional pep­
amino-carboxylpolyethylene glycol (NH2-PEG8-COOH) on one side and tides consisting of a number of domains along with antifouling proper­
TPE was attached to H9 using DEVD peptide as the core. The anticancer ties, have been utilized in early cancer detection. A multifunctional
drug doxorubicin was loaded and based on the aggregation-induced peptide SS-IMVTESSDYSSY-KK-FHYQRDTPKSYN has been utilized for
emission (AIE) phenomenon of TPE, the loading and release of DOX the fabrication of a direct-real-time SPR sensor for PD-L1 exosomes (Mao
were processed. et al., 2021). The peptide comprised of four different regions depending
upon the sequence and type of amino acids- SS (antifouling region),
2.3. Antifouling peptide based optical biosensors IMVTESSD (assembly region), YSSY (binding region), and
FHYQRDTPKSYN (recognizing region). The Ser residues with hydro­
The selective and sensitive performance of any optical sensor in bio philic properties prevented the non-specific adsorption of biomolecules
fluids (saliva, blood, serum plasma, urine, and cell lysate), is highly via hydration. The multifunctional peptide enhanced the interactions
affected by nonspecific adsorption (fouling) and background contami­ among graphene and target molecules and ensure the selective detection
nation or biosensor malfunction (Banerjee et al., 2011). To resolve this of PD-L1 exosomes. In another report, RVRRFFF peptide with anti­
issue numerous antifouling materials had been fabricated on the sensor fouling (FFF) and recognition (RVRR) properties were employed for
surface. Among all, synthetic antifouling peptides suppress the available fluorimetric detection of MDA-MB-231 cells (Li et al., 2019). Nitro­
antifouling materials due to their high biocompatibility, easy synthesis, benzoxadiazole (NBD) modified peptide (RVRRFFF-NBD)
flexible and tunable structures. Antifouling peptides comprise electric self-assembled via π–π interactions and resulted in quenching of fluo­
neutrality and strong hydrophilic properties which makes them superior rescence. Here FFF group provided the antifouling property as well as
resistant materials toward protein adsorption (Ye et al., 2015). Modu­ helped in the self-assembly of the peptide. In the presence of
lation of these properties can be easily carried out by varying the MDA-MB-231 cells self-assembled structure was disrupted due to the
sequence, number, and type of amino acid residues in a particular specific binding of the peptide with the target and resulting in the regain
peptide. The immobilization of antifouling peptides onto the surface of of fluorescence.
the optical sensor includes the electrografting, self-assembly, or poly­ These antifouling peptides are a magnificent candidate for the
merization methods from which self-assembly is the easiest way (Jiang fabrication of PEC sensors, for example, an optical PEC based sensor was
et al., 2020). An ECL ratiometric sensor was reported for DNA methyl­ reported for human chorionic gonadotropin (hCG) using EKEKE­
transferase (MTase) detection and to study the antifouling properties of KEPPPPC (antifouling) and PPLRINRHILTR (recognition) peptide in one
the novel peptide DKDKDKDPPPPC (Li et al., 2019). The characteriza­ probe electrode (Gu et al., 2021). The recognition and antifouling pep­
tion of the peptide with Differential Pulse Voltammetry (DPV) and tide were attached to CBO/Au photocathode via Au–NH2 and Au–S
fluorescein diacetate (FDA) confirmed the antifouling nature of the chemistry respectively. The steric hindrance caused by the peptide-hCG
peptide. The detection principle of the ECL sensor initiates a dual ECL complex reduces the photocurrent and leads to the selective detection of
signal based on the HCR amplification method. High concentrations of the target in complex media. In another report, PEC cytosensor for HeLa
Dam MTase decreased the Au@luminol intensity and increased the ECL cells based on zwitterionic peptide (EESKSESKSGGGGC), As1411
intensity of PTC-NH2 which allowed the selective detection of DNA aptamer, and ITO/TiO2/ZnIn2S4 matrix have been reported (Fan et al.,
MTase. Similarly, another ECL sensor was fabricated by immobilization 2019). The decrease in photocurrent response upon binding of HeLa
of DKDKDKDPPPPC peptide on AuNPs via thiol bond formation (Hao cells to AS1411 aptamer exhibited sensitive and selective detection with
et al., 2020). Further, CdS QDs coupled to the peptide by EDC/NHS LOD of 34 cells/mL. Similarly, the PEC sensor for neuron specific
chemistry were reported to study the electrochemiluminescence reso­ enolase (NES) analysis was fabricated using EKEKEKEPPPPC peptide,
nance energy transfer (ERET) between QDs and Cy5 dye. The ECL based anti-NSE, and monocrystal perovskite magnesium dititanate (MgTi2O5)
sensor successfully reported the detection of carcinoembryonic antigen as a basic matrix (Xu et al., 2022). Peptide immobilized on MgTi2O5
(CEA) based on ratiometric ECL signal getting from Au-luminol (positive surface using polydopamine (PDA) provided the antifouling property as
potential) and QDs (negative potential). Apart from the immobilization well as enhanced the sensitivity of the sensor. Similarly, the PEC
of antifouling peptides on the sensor surface, these peptides were also immunosensor was illustrated in which zwitterionic peptide KAEA­
used for the synthesis of antifouling magnetic beads to detect miRNA 21 KAEAPPPPC was used to provide an antifouling interface to novel het­
in biological media (Hao et al., 2021). The dendritic peptide and hairpin erojunction photocathode Au/ZIS/CIS (Hu et al., 2022). The proposed
DNAzyme are anchored on MBs to acquire an antifouling surface and PEC immunosensor provided a new approach for cardiac troponin I
from the two signal amplification methods, the first one comprised of (cTnI) detection at the pM level.
the DNAzyme on MBs which caused cycle amplification of nucleic acids.
The product of the first amplification (DNA) activated the hybrid chain 2.4. Self-assembled peptide based optical biosensors
reaction (HCR) on the GCE/TiO2 NNs/PANI electrode to generate the
second amplification with respect to the ECL signal. The proposed sensor Self-assembly of bio-molecules such as peptides, proteins, lipids, and
can detect miRNA with an LOD of 0.13 fM, and the antifouling MBs can DNA provides the efficient pathway for the engineering of ordered 1D,
serve as a separating tool. 2D, and 3D nano/micro structures (Singh et al., 2020). Depending upon
An antifouling peptide was utilized for the development of a home- the specific recognition, these ordered structures are basically facilitated
made fiber optic surface plasmon resonance (FO-SPR) sensor for a by the non-covalent interactions including the π-π stacking interactions,
platelet-derived growth factor (PDGF-BB) (Qian et al., 2019). The hydrophobic forces, hydrogen bonding, electrostatic interactions, and
FO-SPR sensor assembly is constructed by immobilization of antifouling van der Waals interactions (Brar et al., 2021). Recently self-assembly of
peptide and PDGF-BB specific aptamer on gold-coated optical fibers peptides has been reported to form ordered nanostructures such as
(Fig. 4c and d). The interaction of PDGF-BB with aptamer resulted in nanospheres, nanotapes, nanotubes, and nanofibrils (Gazit, 2007).
refractive index alteration and detection of a target with a LOD of 0.35 Inspired by these, molecular self-assembled peptides have been widely
pM. A newly developed SPR based sensor for ctDNA had been reported used in the field of bio-sensing and biomedical science. By utilizing the
which consists of a dual-functional surface layer, PNA probe, and AuNPs self-assembly property, a novel peptide nanofibrils based colorimetric
(Bellassai et al., 2021). The dual-functional layer was introduced to the method has been developed for HepG2 detection using B

8
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

(OH)2AEAEAELRARARL-OH (C. Chen et al., 2013). The peptide was motivates the researchers to design the pH-based biosensors for cancer
modified with phenylboronic acid (BA-P) to further conjugate with detection. In this regard, pH responsive nanoparticles were reported
catechol alizarin red dye (ARS) for colorimetric probe fabrication. The based on the self-assembly of oligopeptide ([CH3CONH]-K(DEAP)SKSK
interactions of ARS with BA-P resulted in the self-assembly of peptides (DEAP)-[CONH2]) (Zhao et al., 2014). The peptide had been modified
into nanofibrils and the color changed from wine-red to orange yellow. with fluorescent dye and BHQ-1 for FRET based detection of the tumor.
After the interactions with target HepG2 cells, the self-assembly of The non-fluorescent supramolecular nanoparticles of self-assembled
peptides get disordered, and the change in color can be seen by the peptide quickly changed to disassembled fluorescent peptide mole­
naked eyes. Apart from this, peptides can be self-assembled on the metal cules upon binding with the tumor at pH 6.7–7.1. This pH-based
surface via the formation of a covalent bond between the –SH group of detection is quite useful for easy and effective tumor diagnosis in clin­
Cys and metal. In this context, cysteine terminated WLEAAYQRFLC ical. Similarly, pH triggered hydrophilic peptide (SKDEEWHKNNFPLSP)
(WC10) peptide self-assembled on ECL matrix for human breast cancer and hydrophobic signal molecule tetraphenylethylene based fluorescent
cell (MCF-7) detection (L. Zhang et al., 2017). The high affinity between detection of VEGFR2 was reported (Qian et al., 2018). The peptide was
WC10 and MCF-7 along with enzyme catalysis-based signal amplifica­ modified with TPE to assemble non-fluorescent nanoparticles which
tion and recognition property of lectin Con A provided the highly sen­ regain their fluorescence during the binding with VEGFR2 in acidic pH.
sitive detection with a LOD of 150 cell/mL. The peptides with self-assembling properties can be modified
Self-assembly of peptides is susceptible to molecular environment, a conveniently with various fluorophores (TPE, TAMRA, NBD) for the
small change due to various factors such as pH, solvent, temperature, early diagnosis of cancer. A fluorescent light-up probe for caspase-3 was
etc. disrupting the ordered structure. The growth of cancer cells is illustrated based on the aggregation induced emission (AIE) by TPE-
facilitated by a slightly acidic environment (pH 6.7–7.1), which GFFYK(DVEDEE-Ac) (Han et al., 2016). The –COOH terminal of DVED

Fig. 5. (a) The chemical structures of RVRRFFF (C-3)


and nitrobenzoxadiazole NBD. (b) Schematic repre­
sentation for peptide self-assembly based detection of
furin C-3. Reprinted with permission from (Li et al.,
2019). Copyright (2019) American Chemical Society.
(c) Schematic representation of RGDs and TAT con­
jugated NaYF4:Yb3+/Er3+ NPs for Human Epithelioid
Cervix Carcinoma detection. Reprinted with permis­
sion from (Kostiv et al., 2016). Copyright (2016)
American Chemical Society.

9
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

segment had been cleaved in the presence of caspase-3 resulting in (MMP-7) was carried out based on the CPP peptide (JR2EC) function­
fluorescence activation due to the absence of hydrophilic groups. Fol­ alized gold nanoparticles (GNPs). GNPs were modified through the
lowed by this, enzyme hydrolysis in the biological environment cysteine in the middle of the JR2EC sequence (NAADLEKAIEA­
enhanced the AIE of the TPE-GFFYK peptide via self-assembly. Similarly, LEKHLEAKGPCDAAQLEKQLEQAFEAFERAG), via thiol-gold linkage.
amphiphilic peptide RVRRFFF was modified with nitrobenzoxadiazole MMP-7 can recognize and cleave the peptide which was further recog­
(NBD) fluorophore to fabricate non-fluorescent micelles by nized by GNPs aggregation. A LSPR shift and a color change from red to
self-assembly (Li et al., 2019) (Fig. 5a). The RVRR domain in the peptide blue can detect the MMP-7 in the salivary gland of cancer patients with a
was selectively bound to furin and turn-on the quenched fluorescence in LOD of 5 nM.
the hydrophobic environment (Fig. 5b). An interesting approach was An interesting report was presented on chemiluminescence based on
reported by fabricating the fluorescent sensor for breast cancer detection the cell adhesive RGD and TAT peptide decorated lanthanide NPs
using peptide ANSFLEELRHSSLERECIEEICDFEEAKEIFQN-AN33 and 2D (NaYF4:Yb3+/Er3+) for Human Epithelioid Cervix Carcinoma detection
material molybdenum disulfide (MoS2) (Dou et al., 2019). The peptide (Kostiv et al., 2016). The click chemistry had been applied for decorating
modified TAMRA (tetramethyl-rhodamine) fluorophore (TAM­ the NaYF4:Yb3+/Er3+ NPs with RGD and TAT peptides (Fig. 5c). Further,
RA-AN33) loses its activity during the binding with MoS2 via a the targeting affinity of utilized peptides facilitates further it to get
self-assembled mechanism. The binding of protein C receptor (PROCR) localized at the membrane of HeLa cells helping in the imaging of
with TAMRA-AN33-MoS2 broke the ordered structure and enabled the Human Epithelioid Cervix Carcinoma and also in photothermal therapy.
detection via activation of the fluorophore. In another report, a signal amplification strategy based on Ru(bpy)2+- 3
Peptides comprising the different domains with distinctive ap­ encapsulated silica nanoparticles for the electrochemiluminescent
proaches can be synthesized and utilized for cancer detection without cytosensing and sensitive quantification of drug-induced apoptotic
modification, for example, SPR sensor was fabricated for PD-L1 exosome cancer cells was also carried out (Wu et al., 2012). Here, an
based on SS-IMVTESSDYSSY-KK-FHYQRDTPKSYN and gold chip modi­ arginine-glycine-aspartic acid-serine (RGDS) tetrapeptide was func­
fied with graphene (Mao et al., 2021). Peptide consists of different do­ tionalized with multiwalled carbon nanotubes (MWCNTs). The formed
mains with a different function, where IMVTESSD act as a self-assembly RGDS-MWCNTs composites were then immobilized on a graphene
domain that participated in the formation of ordered and crystallo­ nanotubes surface as an anchorage layer in order to effectively capture
graphic nanostructures. The binding of Pd-L1 exosome disrupted the the cells via the specific combination of RGD domains with integrin
ordered assembly resulting in the alteration of the refractive index value. receptors on the cell surface. Similarly, a simple and sensitive electro­
These changes provided the effective and convenient detection of the generated chemiluminescence biosensor was developed to monitor
Pd-L1 exosome for early stage cancer diagnosis. The reported self matrix metalloproteinase 2 (MMP-2) by employing a CPP peptide
assembled peptides based optical biosensors for early cancer detection (CGPLGVRGK) as a molecular recognition substrate and thus help in the
have been listed in Table 1. detection of the malignancy (Dang et al., 2016). Bis(2, 2′ -bipyr­
idine)-4′ -methyl-4-carboxybipyridine-ruthenium N-succinimidyl ester-
2.5. Cell penetrating peptide based optical sensors bis(hexafluorophosphate) (Ru(bpy)2(mcbpy-O-Su-ester)(PF6)2 (Ru1)
was used as ECL-emitting species and covalently labeled onto the pep­
Cell penetrating peptides act as an astonishing agent for detecting tide through lysine via acylation reaction to form Ru1-peptide. ECL
the targeted sites of diseases and delivering drugs. The first CPP was peptide-based biosensor was further fabricated by self-assembling the
discovered in 1988 via the discovery of the Human immunodeficiency ECL probe onto the surface of the gold electrode. The decreased ECL
virus (HIV) trans activator of transcription protein (TAT) (Figueiredo intensity was linear in the concentration range of 1–500 ng/mL of
et al., 2014). In general, a CPP is a short peptide sequence that either has MMP-2. Moreover, the ECL biosensor is successfully applied to the
35 or less than 35 amino acid residues. Cell penetrating peptides have detection of MMP-2 secreted by living cells, such as HeLa cells.
caught much attention due to their ambiguous characteristics such as In another example, a novel intracellular protease fluorescence
low cytotoxicity, an ability to enable receptor-independent transport “turn-on” sensor based on the nanoconjugate of graphene oxides and
across cell membranes when linked to oligonucleotides or proteins, and peptide substrates was developed and used for early stage malignancy
the ability to cross the cell plasma membrane during the process of detection (Wang et al., 2011). The peptide was cleaved from the
endocytosis. Thus, CPPs are classified into three categories-on the basis GO-peptide conjugate after being transported into cells by intracellular
of their physicochemical properties: cationic, hydrophobic, and proteases which further results in a significant increase in fluorescence
amphipathic. Cationic CPPs contain highly positive net charges that emission because of the release of the fluorophore from GO. The fluo­
originate from the basic short strands of arginine and lysine and are most rescence peaks drastically enhanced with increasing caspase-3 concen­
employed in diagnosis due to their preferable interactions with cell tration in the range from 7.25 to 362 ng/mL and LOD was found to be
surface (El-Sayed and Harashima, 2009). Based on these, CPPs played an 7.25 ng/mL. Further FRET phenomena has been used for MMPs detec­
important role in modern biosensor development in various fields, such tion based on the nona-arginine (R9), octa-glutamate (E8), labeled with
as clinic diagnosis and earlier detection of diseases. A report has suc­ Cy5 and Cy7, respectively, and MMP2/9 cleavable linker (PLGCAG)
cessfully demonstrated the application of CPPs for colorimetric (Savariar et al., 2013). When MMPs in the tumor tissue cleave the linker,
screening and imaging of cancer cells (P. Chen et al., 2013). In this, FRET between Cy7 and Cy5 was interrupted producing red fluorescence
colorimetric detection of the matrix metalloproteinase matrilysin by Cy5 in the tumor tissue. Recently, a group has developed a

Table 1
List of reported self-assembled peptide based optical biosensors for early cancer detection.
Sequence of Peptide Type of Cancer Transduced signal LOD Reference

B(OH)2AEAEAELRARARL-OH Liver Cancer Colorimetric 18.6 mM Chen et al. (2013)


WLEAAYQRFLC (WC10) Breast cancer ECL 150 cell/mL Zhang et al. (2017)
[CH3CONH]-K(DEAP)SKSK(DEAP)-[CONH2] Skin Cancer Fluorescence - Zhao et al., 2014)
SKDEEWHKNNFPLSP Breast Cancer Fluorescence - Qian et al. (2018)
TPE-GFFYK(DVEDEE-Ac Colon Cancer Fluorescence 0.54 pM Han et al., 2016
RVRRFFF Breast Cancer Fluorescence 0.11 ng/μL Li et al. (2019)
ANSFLEELRHSSLERECIEEICDFEEAKEIFQN-AN33 Breast cancer Fluorescence 0.76 nM Dou et al., 2019
SS-IMVTESSDYSSY-KK-FHYQRDTPKSYN Lung Cancer SPR 20 particles/mL Mao et al. (2021)

10
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

cell-permeable biosensor to detect deubiquitinating enzyme (DUB) ac­ addition, mesoporous silica nanoparticles, superparamagnetic iron
tivity in intact cancer cells (Safa et al., 2019). The reporter incorporated oxide nanoparticles (SPIONs), and gold nanoparticles have also been
the previously established RWRWR β-hairpin CPP with a short explored for imaging agents and diagnosis, on account of their low
DUB-binding peptide sequence (LRGG), and a fluorescent molecule cytotoxicity and easy functionalization (Grasso et al., 2019)]. Fluo­
(AFC). They observed that the conjugation of the four amino acid rescently labeled, PEG-coated SPIONs were successfully bioconjugated
binding sequence diminished the net uptake of the peptide-based re­ with gH625-derived peptide from the CPP family for breast cancer im­
porter compared to the uptake of the RWRWR CPP itself. RWRWR up­ aging ((Perillo et al., 2017). The uptake of the bioconjugated NPs into
take in HeLa cells was found to occur rapidly for 90 min to be sufficiently cytosolic low polarity locations seems consistent with the
internalized which clearly demonstrated that the efficiency of the CPP endosomal-lysosomal pathways of endocytosis, which appears to be the
should be considered when designing CPP-conjugated therapeutics or major cellular uptake mechanism for most nanosystems. Thus, in
biosensors. In a similar report, it has been observed that the conjugation contrast to free gH625 crossing cell plasma membranes by direct
of green fluorescent protein (GFP) as a model cargo onto seven different translocation, the CPP-capped PEGylated SPIONs undergo enhanced
CPPs, such as penetratin, octa-arginine, TAT, Transportan, and Xentry endocytosis.
(Patel et al., 2019) has been successfully applied for cancer cell detec­ It has been noticed recently that some CPPs have been used for
tion. The study demonstrated that uptake of the cargo was not only cancer diagnosis using molecular imaging techniques like optical im­
dependent on the CPP but also on the cell line it was tested in, including aging, wherein their sensitivity depends on the efficient delivery of
HeLa, HEK, and HepG2. A group of researchers used nanomaterial such contrast agents to the cytoplasm and/or nuclei of the target tissue.
as quantum dots and developed a novel antibody-free assay to study ACPPs have many advantages over other optical and MR-based probes.
protein kinase (Shiosaki et al.2013). A cationic peptide was modified First, their amenability to dual labeling for MR and fluorescence pro­
with a fluorophore that interacts electrostatically with anionic QDs vides an ideal correlation between tomographic imaging that can detect
causing FRET from the QDs to the fluorophore. The fluorescence lifetime tumors anywhere in the body with higher resolution. This added infor­
of the fluorescent probes which is also called FLIM can also be used for mation may be particularly useful in the staging of certain cancers, in
monitoring biological processes. Based on this, researchers have re­ which tumor infiltration of a capsule plays a critical role in prognosis.
ported a cell-penetrating peptide biosensor for dynamic monitoring of Thirdly, the increase in molecular size further reduces the high back­
phosphorylation by Abl kinase (Damayanti et al., 2013). FLIM, which is ground uptake, previously seen for unadorned ACPP’s and allows for
not confounded by photobleaching or cellular autofluorescence, was additional tumor accumulation based on enhanced permeability and
employed to detect phosphorylation-dependent fluorophore lifetime retention although still remaining small enough to penetrate into solid
shifts (1–2 ns) in living cells. In order to accurately probe diseased sites, tumors. For all of these reasons, ACPPs may be useful for clinical pur­
many CPPs-mediated molecular probes such as activatable CPP (ACPP) poses in the early stage of cancer. Thus in a report, the cell-penetrating
have been used for cancer diagnosis. Further, research has shown that peptide-ACPPD (EEEEEEEEPLGCAGRRRRRRRRRC) has been applied
ACPP can be activated by matrix metalloproteinase over-expressed in for the successful delivery of quantum dots to reveal targeted tumor
tumors and imaging can be carried out using fluorescent agents. In a imaging and thus help in increasing the chances of penetration of the
report, a research group designed and developed a novel intravenously nanomaterial through the cell for highly specific cell imaging of the
administered peptide-dye conjugate (AVB-620) for breast cancer diag­ cancer cells (Olson et al., 2010). Another such theranostic application
nosis (Miampamba et al., 2017). Experimental results indicated that based on the use of CPP-coated silica NPs were described (Yu et al.,
AVB-620 can visualize the tumors under a fluorescence imaging camera 2016; De La Torre et al., 2015). Where the mesoporous silica NPs were
based on the FRET between Cy5 and Cy7. Moreover, AVB-620 was in the coated with CPPs and a fluorogenic helps in the detection of the HeLa
clinical phase I study in patients with breast cancer (Unkart et al., 2017). cell line.
The result indicated that AVB-620 was safe, and well-tolerated at doses In another report, a plasmonic gold nanostar platform was designed
for tumor-specific fluorescence detection which was obtained by to increase the Raman signal via the surface-enhanced resonance Raman
intra-operative imaging of surgical specimens after administration of scattering (SERRS) effect (Fales et al., 2013). SERS imaging and
AVB-620. In addition, a similar type of report has been also documented photodynamic therapy using this construct were demonstrated on
where they also designed and synthesized CPPs-mediated molecular BT-549 breast cancer cells. The TAT peptide allowed for effective Raman
probes as imaging agents (Zhu et al., 2018). They firstly synthesized imaging and photosensitization with the nanoparticle after the 1h in­
three thermally activated delayed fluorescence (TADF) chemicals cubation period. In the absence of the TAT peptide, nanoparticle accu­
(4CzIPN, NAI-DPAC, BTZ-DMAC) and TADF was loaded into amphi­ mulation in the cells was not sufficient to be observed by Raman imaging
philic CPPs (F6G6(rR)3R2) which was capable of self-assembling into or to produce any photosensitization effect after this short incubation
nanoparticles in water to constructed TADF nanoparticles as imaging period. This report shows the first application of combined SERS im­
agents. aging and photosensitization from a theranostic nanoparticle construct.
With the emergence of nanotechnology, CPPs-mediated nanoplat­
forms as imaging agents for diagnosis have attracted widespread 3. Future perspective
attention (Onoshima et al., 2015). Because of their advantages of low
cytotoxicity, good biocompatible, high cell membrane permeability, The present review highlights the recent progress made in the optical
small size, large surface areas, abundant functional group on their sur­ sensing of cancer using peptide-based biosensors. The peptides exhibit
face, and easily modify. One excellent example was based on stability against denaturation, specificity, and chemical flexibility.
CPPs-mediated quantum dots as imaging agents for biological diagnosis These features of peptides can contribute to the future development of
(P. Zhang et al., 2018; Yang et al., 2020). However, CPPs-mediated QDs novel biosensors for targeted cancer detection with an improved limit of
not only possess the advantages of QDs but also have high cell mem­ detection, sensitivity, and range of linearity. It is important to note that
brane permeability. A near-infrared semiconducting polymer dots although the reported peptide-based biosensors for optical sensing are
coated with a CPP (R8-Pdots) for cell tracking in deep organs has been focused on specific biomarker-based cancer detection and showed great
designed (Z. Zhang et al., 2018) which presented low cytotoxicity and potential for the early-stage diagnosis of cancer, still these methods are
high cell membrane permeability for MCF-7 cells. Meanwhile, a clear associated with some limitations. Also, most of the reported detection
and strong fluorescence signal was recorded in vitro and in vivo. The methods can detect one biomarker at a time which limited the simul­
results of cell tracking capability of Pdots in live mice in vivo indicated taneous multiple detections of different cancers present in different parts
that MCF-7 cells labeled with R8-Pdots were visualized in real-time, of the body. Multiple biomarkers-based detections by one biosensor are
which are powerful for whole-body fluorescence imaging in vivo. In highly required for successful cancers diagnosis. It is worth mentioning

11
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

that the peptides incorporated with nanoparticles, nanocavities, 2D Data availability


materials, and fibers can be used for the development of novel optical
biosensors with the high specificity of different biomarkers of cancer. No data was used for the research described in the article.
However, the progression of the development of such novel and sensi­
tive biosensors will primarily depend on the advancement of new ma­ Acknowledgments
terials exhibiting unique shapes and structures. Since most of the
reported peptide-based biosensors show good analysis in solution or cell R.K.S. acknowledges the support of the Department of Science and
lysate but due to their complicated components, the precise molecular Technology (DST), India-Project No. DST/ TDT/AGRO-10/2019. G.K.S.
recognition is difficult in real samples of blood and interstitial fluid. In thanks University grant commission (UGC), India and P.M thanks
this context, material-based synergetic recognition can be a more Council of Scientific and Industrial Research (CSIR), India for their
effective approach. The increasing research for the development of respective research fellowships.
nanomaterials on peptide-based optical sensors demonstrated their
significance in early cancer detection and hence can be used to create Appendix A. Supplementary data
the biosensors for easy and successful clinical diagnostics. Also, peptide-
based biosensors can be used to fabricate the commercial portable bio­ Supplementary data to this article can be found online at https://doi.
chips. The new sensing principles and electrochemical signaling can be org/10.1016/j.biosx.2022.100259.
used for the development of peptide-based biochips models. The
development of biochips of peptide-based biosensors will provide a new References
prospect for the fast and convenient biological analysis of early-stage
detection of cancer. It is important to note that there is a certain gap Aina, O.H., Sroka, T.C., Chen, M., Lam, K.S., 2002. Biopolymers 66, 184–199.
Askim, J.R., Suslick, K.S., 2017. Fluorescent Sensors and Imaging Agents 8, 37–88.
between fundamental research and clinical applications however, the Babamiria, B., Baharia, D., Salimi, A., 2019. Biosens. Bioelectron. 142, 111530.
development of these biochips can move the peptide-based biosensing Badr, H.A., Alsadek, D.M.M., Darwish, A.A., ElSayed, A.I., bekmanov, B.O.,
approach from laboratories to the real world for easy and decentralized Khussainova, E.M., Zhanq, X., Cho, W.C.S., Djansuqurova, L.B., Li, C.Z., 2014. Expert
Rev. Proteomics 11, 227–236.
cancer diagnosis. Banerjee, I., Pangule, R.C., Kane, R.S., 2011. Adv. Mater. 23, 690–718.
Bellassai, N., Agata, R.D., Marti, A., Rozzi, A., Volpi, S., Allegretti, M., Corradini, R.,
4. Conclusion Giacomini, P., Huskens, J., Spoto, G., 2021. ACS Sens. 6, 2307–2319.
Bloch, M., Kam, Y., Yavin, E., Moradov, D., Nissan, A., Ariel, I., Rubinstein, A., 2012. Eur.
J. Pharmaceut. Sci. 47, 904–913.
Interdisciplinary research of biomolecular nanomaterials brings Brar, S.K., Kumar, S., Bhasin, A.K.K., Bhasin, K.K., Sharma, R.K., 2021. Mater. Chem.
great challenges and opportunities to push the development of effective Front. 5, 3110–3118.
Burggraaf, J., Kamerling, I.M.C., Gordon, P.B.G., Schrier, S., Kam, M.L.D., Kales, R.B.,
peptide-based optical biosensors with a special emphasis on cancer
Indrevoll, B., Bjerke, R.M., Moestue, S.A., Yazdanfar, S., Langers, A.M.J.,
detection. Currently, strategies for employing the concept of peptide- Nordmo, M.S., Torheim, G., Warren, M.V., Morreau, H., Voorneveld, P.W.,
based nanomaterials are being diversified toward the different roles in Buckle, T., Leeuwen, F.W.B.V., degårdstuen, L., Dalsgaard, G.T., Healey, A.,
the sensing process including the recognition element. Various aspects Hardwick, J.C.H., 2015. Nat. Med. 21, 955–961.
Casciaro, B., Moros, M., Rivera-Fernández, S., Bellelli, A., Fuente, J.M., Mangoni, M.L.,
of the biosensors, such as type of peptide nanomaterial, functionaliza­ 2017. Acta Biomater. 47, 170–181.
tion tool, detection method, signal amplification, and the corresponding Chang, D., Kim, K.T., Lindberg, E., Winssinger, N., 2020. ACS Sens. 5, 807–813.
sample preparation, selectivity, sensitivity, stability, etc., have also been Chang, H., Sun, Y., Yeh, F., Tseng, I.H., Chang, C., Lin, C., 2018. RSC Adv. 8,
29013–29021.
discussed in detail. In addition, the above-summarized examples of Chen, C.Y., Burstein, E., 1980. Phys. Rev. Lett. 45, 1287–1291.
peptide-based biosensors reported so far in the literature, can be eval­ Chen, C., Xu, X., Wang, Y., Yang, J., Jia, H., Cheng, H., Chu, C., Zhuo, R., 2013. Small 9,
uated with their advantages and limitations so as to stress their potential 920–926.
Chen, P.H., Lin, C., Guo, K.H., yeh, Y.C., 2017. RSC Adv. 7, 29302–29305.
for further development in this field. It is still clear that we have wit­ Chen, P., Selegård, R., Aili, D., Liedberg, B., 2013. Nanoscale 5, 8973–8976.
nessed explosive growth in achievements related to the utilization of Chen, Y., Liu, J., Yang, Z., Wilkinson, J.S., Zhou, X., 2019. Biosens. Bioelectron. X. 144,
peptide-based optical biosensors and their type as targeted, cell pene­ 111693.
Chen, Y., Ming, H., 2012. Photonic Sens. 2, 37–49.
trating, antifouling, self assembles, etc., with nanomaterial-based bio­ Chen, C., He, R., Zhang, Z., Chen, Y., 2020. Analyst 145, 7603–7608.
sensors. We further aimed for providing the reader with an introduction Cheng, Z., Wu, Y., Xiong, Z., Gambhir, S.M., Chen, X., 2005. Bioconjugate Chem. 16,
to the peptide-based biosensor field in cancer detection. Although sig­ 1433–1441.
Choi, J.H., Choi, J.W., 2020. Nano Lett. 20, 7100–7107.
nificant advances in the development of peptide-based optical bio­
Corkill, G., Rapley, R., 2008. Humana Press. https://doi.org/10.1007/978-1-60327-375-
sensors have been made, this field is still in its infancy. The remaining 6_1.
challenges for the subsequent development and application of peptide- Croci, S., Manicardi, A., Rubagotti, S., Bonacini, M., Iori, M., Capponi, P.C., Cicoria, G.,
based optical biosensors as a promising solution should be addressed Parmeggiani, M., Salvarani, C., Versari, A., Corradini, R., Asti, M., 2019. Sci. Rep. 9,
3376.
optimally in future. For this purpose efforts are being ongoing for Crosby, D., Bhatia, S., Brindle, K.M., Coussens, L.M., Dive, C., Emberton, M., Esener, S.,
delivering effective peptide based optical biosensors for early stage Fitzgerald, R.C., Gambhir, S.S., Kuhn, P., Rebbeck, T.R., Balasubramanian, S., 2022.
cancer detection. Science 375, 1244.
Damayanti, N.P., Parker, L.L., Irudayaraj, J.M., 2013. Angew. Chem. 125, 4023–4026.
Damborský, P., Švitel, J., Katrlík, J., 2016. Essays Biochem. 60, 91–100.
CRediT authorship contribution statement Dang, Y.Q., Gao, H., Li, Z., Qi, H., Gao, Q., Zhang, H.C., 2016. Anal. Bioanal. Chem. 408,
7067–7075.
De La Torre, C., Mondragõn, L., Coll, C., García-Fernández, A., Sancenõn, F., Martínez-
Gurpreet K. Soni: Conceptualization, Writing – original draft, Máñez, R., Amorõs, P., Pérez-Payá, E., Orzáez, M., 2015. Chem. Eur J. 21,
Writing – review & editing. Saima: Writing – review & editing. Priya 15506–15510.
Manhas: Writing – review & editing. Rohit K. Sharma: Supervision, Dou, W., Liu, L., Gao, J., Zang, Y., Chen, G., Field, R., James, T.D., Li, J., He, X., 2019.
Chem. Commun. 13235–13238, 55.
Conceptualization, Writing – review & editing, Overall, Supervision, Economou, A., Karapetis, S.K., Nikoleli, G.P., Nikolelis, D.P., Bratakou, S., Varzakas, T.
Research support. H., 2017. https://onlinelibrary.wiley.com/doi/10.1002/9781119105916.ch9.
El-Sayed, A., Harashima, H., 2009. AAPS J. 11, 13–22.
Fales, A.M., Yuan, H., Vo-Dinh, T., 2013. Mol. Pharm. 10, 2291–2298.
Declaration of competing interest Fan, G., Li, Z., Lu, Y., Ma, L., Zhao, H., Luo, X., 2019. Sens. Actuators B Chem. 299,
12996.
The authors declare that they have no known competing financial Farré, M., Barceló, D., 2007. Chapter 16 Sensor, biosensors and MIP based sensors.
https://doi.org/10.1016/B978-044452843-8/50017-1.
interests or personal relationships that could have appeared to influence
the work reported in this paper.

12
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

Figueiredo, I.R., Freire, J.M., Flores, L., Veiga, A.S., Castanho, M.A.R.B., 2014. IUBMB Metcalf, G.A.D., Shibakawa, A., Patel, H., Sita-Lumsden, S., Zivi, A., Rama, N., Bevan, C.
Life 66, 182–194. L., Ladame, S., 2016. Anal. Chem. 88, 8091–8098.
Gaillard, C., Girard, H.A., Falck, C., Paget, V., Simic, V., Ugolin, N., Bergonzo, P., Miampamba, M., Liu, J., Harootunian, A., Gale, A.J., Baird, S., Chen, S.L., 2017.
Chevillard, S., Arnault, J.C., 2014. RSC Adv. 4, 3566–3572. Theranostics 7, 3369–3386.
Gao, H., Dang, Q., Xia, S., Zhao, Y., Qi, H., Gao, Q., Zhang, C., 2017. Sensor. Actuator. B Mittal, S., Kaur, H., Gautam, N., Mantha, A.K., 2017. Biosens. Bioelectron. 88, 217–231.
Chem. 253, 69–76. Naorungroj, J., Teengam, P., Vilaivan, T., Chailapakul, O., 2021. New J. Chem. 45,
Gao, T., Xing, S., Xu, M., Fu, P., Yao, J., Zhang, X., Zhao, Y., Zhao, C., 2020. Microchim. 6960–6967.
Acta 187, 375. Nitin, N., Rosbach, K.J., Naggar, A., Williams, M., Gillenwater, A., Richards-Kortum, R.,
Gazit, E., 2007. Chem. Soc. Rev. 36, 1263–1269. 2009. Neoplasia 11, 542–551.
Grasso, G., Mercuri, S., Danani, A., Deriu, M.A., 2019. J. Phys. Chem. B 123, Office for National Statistics UK, 10 June 2016. Cancer Survival by Stage at Diagnosis for
10622–10630. England Experimental Statistics: Adults Diagnosed 2012, 2013 and 2014 and
Gu, S., Shi, X., Zhang, D., Fan, G., Luo, X., 2021. Anal. Chem. 93, 2706–2712. Followed up to 2015. https://www.ons.gov.uk/peoplepopulationandcommuni
Gupta, A., Mishra, A., Puri, N., 2017. J. Biotechnol. 259, 148–149. ty/healthandsocialcare/conditionsanddiseases/bulletins/cancersurvivalbystagea
Han, A., Wang, H., Kwok, R.T.K., Ji, S., Li, J., Kong, D., Tang, B.Z., Liu, B., Yang, Z., tdiagnosisforenglandexperimentalstatistics/adultsdiagnosed20122013and2014an
Ding, D., 2016. Anal. Chem. 88, 3872–3878. dfollowedupto2015.
Han, J.Y., Choi, J.J., Kim, J.Y., Han, Y.L., Lee, G.K., 2016. BMC Cancer 16, 627. Olson, E. S., Jiang, T., Aguilera, T. A., Nguyen, Q. T., Ellies, L. G., Scadeng, Miriam,
Han, Z., Shang, W., Liang, X., Yan, H., Hu, M., Peng, L., Jiang, H., Fang, C., Wang, K., Tsien, R. Y., 2010. 107, 4311-4316.
Tian, J., 2019. Mol. Imag. Biol. 21, 67–77. Onoshima, D., Yukawa, H., Baba, Y., 2015. Adv. Drug Deliv. Rev. 95, 2–14.
Hao, Q., Lei, W., Niu, S., Ding, C., Luo, X., 2020. Anal. Chim. Acta 1136, 134–140. Patel, S.G., Sayers, E.J., He, L., Narayan, R., Williams, T.L., Mills, E.M., Allemann, R.K.,
Hao, Q., Xu, Q., Niu, S., Ding, C., Luo, X., 2021. Anal. Chem. 93, 10679–10687. Luk, L.Y.P., Jones, A.T., Tsai, Y.H., 2019. Sci. Rep. 9, 6298.
Hashoul, D., Shapira, R., Falchenko, M., Tepper, O., Paviov, V., Nissan, A., Yavin, E., Perillo, E., Hervé-Aubert, K., Allard-Vannier, E., Falanga, A., Galdiero, S., Chourpa, I.,
2019. Biosens. Bioelectron. 137, 271–278. 2017. J. Colloid Interface Sci. 499, 209–217.
Holford, T.R.J., Séamus, H.F., Higson, P.J., 2012. Biosens. Bioelectron. 34, 12–24. Qi, H., Li, M., Dong, M., Ruan, S., Gao, Q., Zhang, C., 2014. Anal. Chem. 86, 1372–1379.
Homola, J., 2008. Chem. Rev. 108, 462–493. Qi, H., Zhang, C., 2020. Anal. Chem. 92, 524–534.
Hou, S., Feng, T., Zhao, N., Zhang, J., Wang, H., Liang, N., Zhao, L., 2020. J. Pharm. Qian, H., Huang, Y., Duan, X., Wei, X., Fan, Y., Fan, D., Gan, D., Yue, S., Cheng, W.,
Anal. 10, 482–489. Chen, T., 2019. Biosens. Bioelectron. 140, 111350.
Hu, Z., Xu, Y., Wang, H., Fan, G., Luo, X., 2022. Chin. Chem. Lett. https://doi.org/ Qian, Y., Wang, W., Wang, Z., Jia, X., Han, Q., Rostami, I., Wang, Y., Hu, Z., 2018. ACS
10.1016/j.cclet.2021.12.088. Appl. Mater. Interfaces 10, 7871–7881.
IgIoi, G.L., 1998. Biochemistry 95, 8562–8567. Qin, W., Wu, Y., Hu, Y., Dong, Y., Hao, T., Zhang, C., 2021. ACS Appl. Bio Mater. 4,
Jazayeri, M.H., Amani, H., Pourfatoll, A.A., Pazoki-Toroudi, H., Sedighimoghaddam, B., 1038–1044.
2019. Sens. Bio-Sens. Res. 9, 17–22. Rajchakit, U., Sarojini, V., 2017. Bioconjugate Chem. 28, 2673–2686.
Jerónimo, P.C.A., Araújo, A.N., Conceição, M., Montenegro, B.S.M., 2007. Talanta 72, Roberts, A., Gandhi, S., 2022. Biosens. Bioelectron. X. 11, 100178.
13–27. Ryoo, S., Lee, J., Yeo, J., Na, H., Kim, Y., Jang, H., Lee, J., Han, S., Lee, Y., Kim, V.N.,
Jiang, C., Wang, G., Hein, R., Liu, N., Luo, X., Davis, J.J., 2020. Chem. Rev. 120, Min, D., 2013. ACS Nano 7, 5882–5891.
3852–3889. Saadati, A., Hassanpour, S., Guardia, M., Mosafer, J., Hashemzaei, M., Mokhtarzadeh, A.,
Jones, J.E., Busi, S.B., Mitchem, J.B., Amos-Landgraf, J.M., Lewis, M.R., 2018. Mol. Baradara, B., 2019. TrAC Trends Anal. Chem. (Reference Ed.) 114, 56–68.
imaging 17, 1–9. Sadhu, K.K., Winssinger, N., 2013. Chem. Eur J. 19, 8182–8189.
Kam, Y., Rubinstein, A., Naik, S., Djavsarov, I., Halle, D., Ariel, I., Gure, A.O., Safa, N., Anderson, J.C., Vaithiyanathan, M., Pettigrew, J.H., Pappas, G.A., Liu, D.,
Stojadinovic, A., Pan, H.G., Tsivin, V., Nissan, A., Yavin, E., 2014. Cancer Lett. 352, Gauthier, T.J., Melvin, A.T., 2019. Pept. Sci. 111, e24092.
90–96. Savariar, E.N., Felsen, C.N., Nashi, N., Jiang, T., Ellies, L.G., Steinbach, P., Tsien, R.Y.,
Kam, Y., Rubinstein, A., Nissan, A., Halle, D., Yavin, E., 2012. Mol. Pharm. 9, 685–693. Nguyen, Q.T., 2013. Cancer Res. 73, 855–864.
Karimzadeh, A., Hasanzadeh, M., Shadjou, N., Guardia, M., 2018. Trends Anal. Chem. Seitz, W.R., 1989. Comput. Methods Progr. Biomed. 30, 9–19.
107, 1–20. Shahdeo, D., Roberts, A., Kesarwani, V., Horvat, M., Chouhan, R.S., Gandhi, S., 2022.
Katz, E., Willner, I., 2014. Angew. Chem. 43, 6042–6108. Biosci. Rep. 42. BSR20212622.
Kaur, B., Kumar, S., Kaushik, B.K., 2022. Biosens. Bioelectron. 197, 113805. Shi, H., Yang, F., Li, W., Zhao, W., Nie, K., Dong, B., Liu, Z., 2015. Biosens. Bioelectron.
Ke, S., Wen, X., Gurfinkel, M., Charnsangavej, C., Wallace, S., Sevick-Muraca, E.M., Li, C., 66, 481–489.
2003. Cancer Res. 63, 7870–7875. Shigeto, H., Ohtsuki, T., Iizuka, A., Akiyama, Y., Yamamura, S., 2019. Analyst 144,
Kelly, K.A., Setlur, S.R., Ross, R., Anbazhagan, A., Waterman, P., Rubin, M.A., 4613–4621.
Weissleder, R., 2008. Cancer Res. 68, 2286–2291. Shin, Y., Gutierrez-Wing, T., Choi, J., 2021. J. Electrochem. Soc. 168, 017502.
Kim, C.G., Shim, H.S., Hong, M.H., Cha, Y.J., Heo, S.J., Park, H.S., Kim, J.H., Lee, J.G., Shiosaki, S., Nobori, T., Mori, T., Toita, R., Nakamura, Y., Kim, C.W., 2013. Chem.
Lee, C.Y., Cho, B.C., Kim, H.R., 2017. Oncotarget 8, 65111–65122. Commun. 49, 5592–5594.
Kolevzon, N., Hashoul, D., Naik, S., Rubinstein, A., Yavin, E., 2016. Chem. Commun. 52, Shu, J., Tang, D., 2020. Anal. Chem. 92, 363–377.
2405–2407. Singh, P., Narang, N., Sharma, R.K., Wangoo, N., 2020. ACS Appl. Bio Mater. 3,
Kostiv, U., Kotelnikov, I., Proks, V., Šlouf, M., Kucka, J., Engstová, H., Ježek, P., 6196–6203.
Horák, D., 2016. ACS Appl. Mater. Interfaces 8, 20422–20431. Singh, R.P., Oh, B., Cho, J., 2010. 79, 153-161.
Law, C.S., Marsal, L.F., Santos, A., Hussain, C.M. (Eds.), 2020. Handb. Nanomater. Anal. Sokolov, K., Chumanov, G., Cotton, T.M., 1998. Anal. Chem. 70, 3898–3905.
Chem.. Elsevier, pp. 201–226. https://www.sciencedirect. Sołoducho, J., Cabaj, J., 2015. IntechOpen. https://doi.org/10.5772/60967.
com/science/article/pii/B9780128166994000098#. Sonar, S.V., Wampole, M.E., Jin, Y., Chen, C., Thakur, M.L., Wickstrom, E., 2014.
Lee, H., Kim, A., Ahn, I., Joo, S., Lee, S.Y., Yoon, K., Lee, K., 2011. Chem. Commun. 47, Bioconjugate Chem. 25, 1697–1708.
11477–11479. Su, X., Teh, H.F., Lieu, X., Gao, Z., 2007. Anal. Chem. 79, 7192–7197.
Li, C., Gao, N., Xue, Q., Ma, N., Hu, Y., Zhang, J., Chen, B., Hou, Y., 2017. Biotechnol. Sulaiman, D.A., Chang, J.Y.H., Chang, Jason Y.H., Bennett, N.R., Topouzi, H., Higgins, C.
Lett. 39, 1463–1469. A., Irvine, D.J., Ladam, S., 2019. ACS Nano 13, 9620–9628.
Li, M., Anastassiades, C.P., Joshi, B., Komarck, C.M., Piraka, C., Elmunzer, B.J., Sung, H., Ferlay, J., Siegel, R.L., Layersanne, M., Soerjomataram, I., Jemal, A., Bray, F.,
Turgeon, K.T., Johnson, T.D., Appelman, H., Beer, D.G., Wang, T.D., 2010. 2021. Ca - Cancer J. Clin. 71, 209–249.
Gastroenterology 139, 1472–1480. Tadimety, A., Zhang, Y., Kready, M.K., Palinski, T.J., Tsongalis, G.J., Zhang, J.X.J., 2019.
Li, X., Cao, C., Wei, P., Xu, M., Liu, Z., Liu, L., Zhong, Y., Li, R., Zhou, Y., Yi, T., 2019. ACS Biosens. Bioelectron. 130, 236–244.
Appl. Mater. Interfaces 11, 12327–12334. Tummers, Q.R.J.G., Hoogstins, C.E.S.H., Gaarenstroom, K.N., Kroon, C.D.K.,
Li, X., Song, J., Chen, B., Wang, B., Li, R., Jiang, H., Liu, J., Li, C., 2016. Sci. Bull. 61, Poelgeest, M.L.E.V., Vuyk, J., Bosse, T., Smit, V.T.H.B.M., Velde, C.J.H.V., Cohen, A.
276–281. F., Low, P.S., Burggraaf, J., Vahrmeijer, A.L., 2015. Oncotarget 7, 32144–32155.
Li, Y., Wang, L., Ding, C., Luo, X., 2019. Biosens. Bioelectron. 142, 111553. Underwood, J.J., Quadri, R.S., Kalva, S.P., Shah, H., Sanjeeviah, A.R., Beg, M.S.,
Li, Z., Askin, J.R., Suslick, K.S., 2019. Chem. Rev. 119, 231–292. Sutphin, P.D., 2020. Radiology 294, 5–17.
Liao, X., Wang, Q., Ju, H., 2015. Analyst 140, 4245–4252. Unkart, J.T., Chen, S.L., Wapnir, I.L., González, J.E., Harootunian, A., Wallace, A.M.,
Lin, C., Zhao, M., Xi, G., Wang, X., Chen, T., 2018. Fullerenes, Nanotubes. Carbon 2017. Ann. Surg Oncol. 24, 3167–3173.
Nanostruct. 26, 38–41. Urbanek, C., Goodison, S., Chang, M., Porvasnik, S., Sakamoto, N., Li, C.Z., Boehlein, S.
Liu, B., Zhuang, I., Wei, G., 2020. Environ. Sci. Nano 7, 2195. K., Rosser, C.J., 2011. BMC Cancer 11, 233.
Liu, M., Yang, T., Chen, Z., Wang, Z., He, N., 2018. Biomater. Sci. 6, 3152–3159. Valetti, S., Mura, S., Noirav, M., Arpico, S., Dosio, F., Vergnaud, J., Desmaele, D.,
Liu, X., Wang, Q., Chen, J., Chen, X., Yang, W., 2021. Talanta 221, 121379. Stella, B., Couvreur, P., 2014. Bioconjugate Chem. 25, 1971–1983.
Lutz, J., Zarafshani, Zoya, 2008. Adv. Drug Deliv. Rev. 60, 958–970. Veerbeek, j., Steen, R., Vijselaar, W., Rurup, W.F., Korom, S., Rozzi, A., Corradini, R.,
Ma, C., Cao, Y., Goa, X., Zhu, J., 2020. Anal. Chem. 92, 431–454. Segerink, L., Huskens, J., 2018. Langmuir 34, 11395–11404.
Mao, Z., Zhao, J., Chen, J., Hu, X., Koh, K., Chen, H., 2021. Sens. Actuators B Chem. 346, Wang, Q., Zhang, X., Chu, T., Chen, J., Ge, R., Yu, A., 2011. Angew Chem. Int. Ed. Engl.
130496. 50, 7065–7069.
Martinelli, E., Palma, R.D., Orditura, M., Vita, F.D., Ciardiello, F., 2009. Clin. Exp. Wang, X., Zhang, Y., Zhao, L., Wang, D., Yang, H., Kong, J., 2020. Anal. Bioanal. Chem.
Immunol. 158, 1–9. 412, 2413–2421.
Metaferia, B., Wei, J.S., Song, Y.K., Evangelista, J., Aschenbach, K., Johansson, P., Wang, Y., Zhang, X., Wan, K.M., Zhou, N., Wei, G., Su, Z., 2021. J. Nanobiotechnol. 19,
Wen, X., Chen, Q., Lee, A., Hempel, H., Gheeya, J.S., Getty, S., Gomez, R., Khan, J., 253.
2013. PLoS One 8, e58870.

13
G.K. Soni et al. Biosensors and Bioelectronics: X 12 (2022) 100259

Wang, Z., Wang, Y., Jia, X., Han, Q., Qian, Y., Li, Q., Xiang, J., Wang, Q., Hu, Z., Zadran, S., Standley, S., Wong, K., Otiniano, E., Amighi, A., Baudry, M., 2012. Appl.
Wang, W., 2019. Theranostics 9, 1728–1740. Microbiol. Biotechnol. 96, 895–902.
Wang, M., Li, L., Zhang, L., Zhao, J., Jiang, Z., Wang, W., 2022. Anal. Chem. 94, Zeng, J., Liang, D., Cao, Z.X., 2005. Proc. SPIE 5855, 667–670. https://doi.org/10.1117/
431–441. 12.623375.
World Health Organization, Fact sheet: Cancer; www.who.int/news-room/fact-sheets/ Zhang, D., Meng, Y., Zhang, C., 2020. Chem. Commun. 56, 213–216.
detail/cancer. Zhang, J.J., Smaga, L.P., Satyavolu, N.S.R., Chan, J., Lu, Y., 2017. J. Am. Chem. Soc. 139,
Wu, L., Wang, J., Yin, M., Ren, J., Miyoshi, D., Sugimoto, N., Qu, X., 2014. Small 10, 17225–17228.
330–336. Zhang, J., Liu, Q., Ba, Y., Cheng, J., Yang, H., Cui, Y., Kong, J., Zhang, X., 2020. Anal.
Wu, W., Han, J., Xue, P., Xu, R., Kang, Y., 2015. Nanoscale 7, 1753–1759. Chim. Acta 1094, 99–105.
Wu, Y.F., Zhou, H., Wei, W., Hua, X., Wang, L.X., Zhou, Z.X., 2012. Anal. Chem. 84, Zhang, L., Wang, Y., Tian, Q., Liu, Y., Li, J., 2017. Biosens. Bioelectron. 89, 1013–1019.
1894–1899. Zhang, P., Cui, Y., Anderson, C.F., Zhang, C., Li, Y., Wang, R., 2018. Chem. Soc. Rev. 47,
Xiang, Z., Zhao, J., Yi, D., Di, Z., 2021. Angew. Chem. 133, 22841–22845. 3490–3529.
Xu, M., Fu, P., Xing, S., Zhao, Y., Zhao, C., 2020. Chembiochem 21, 2667–2675. Zhang, W., Sui, Y., Budha, A., Zheng, J., Sun, X., Hou, Y., Wang, T.D., Lu, S., 2012. World
Xu, R., Xu, K., Du, Y., Li, J., Dai, L., Wu, T., Ren, X., Wei, Q., 2022. Sens. Actuators B J. Gastroenterol. 18, 2053–2060.
Chem. 361, 131702. Zhang, Z., Yuan, Y., Liu, Z., Chen, H., Chen, D., Fang, X., 2018. ACS Appl. Mater.
Yang, Y., Xia, M., Zhang, S., Zhang, X., 2020. Chem. Commun. 56, 145–148. Interfaces 10, 26928–26935.
Ye, H., Wang, L., Huang, R., Su, R., Liu, B., Qi, W., He, Z., 2015. ACS Appl. Mater. Zhao, X., Lin, C., 2017. RSC Adv. 7, 48554-4856.
Interfaces 7, 22448–22457. Zhao, Y., Ji, T., Wang, H., Li, S., Zhao, Y., Ni, G., 2014. J. Contr. Release 177, 11–19.
Yildiz, U., Alagappan, P., Liedberg, Bo, 2013. Anal. Chem. 85, 820–824. Zhu, Z., Tian, D., Gao, P., Wang, K., Li, Y., Shu, X., 2018. J. Am. Chem. Soc. 140,
Yu, C., Qian, L., Ge, J., Fu, J., Yuan, P., Yao, S.C.L., Yao, S.Q., 2016. Angew. Chem. 128, 17484–17491.
9418–9422.

14

You might also like