Next Article in Journal
Combined Use of Ionic Liquid-Based Aqueous Biphasic Systems and Microfluidic Devices for the Detection of Prostate-Specific Antigen
Next Article in Special Issue
Recent Advances in Field Effect Transistor Biosensors: Designing Strategies and Applications for Sensitive Assay
Previous Article in Journal
Microfluidics as a Ray of Hope for Microplastic Pollution
Previous Article in Special Issue
Electrochemical Immunosensor Using Electroactive Carbon Nanohorns for Signal Amplification for the Rapid Detection of Carcinoembryonic Antigen
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Label-Free Electrochemical Biosensor Platforms for Cancer Diagnosis: Recent Achievements and Challenges

1
Department of Chemistry, Gebze Technical University, 41400 Kocaeli, Turkey
2
Department of Chemistry, Faculty of Science, Hacettepe University, 06800 Ankara, Turkey
*
Author to whom correspondence should be addressed.
Submission received: 1 December 2022 / Revised: 17 February 2023 / Accepted: 23 February 2023 / Published: 1 March 2023
(This article belongs to the Special Issue Label-Free Biosensor)

Abstract

:
With its fatal effects, cancer is still one of the most important diseases of today’s world. The underlying fact behind this scenario is most probably due to its late diagnosis. That is why the necessity for the detection of different cancer types is obvious. Cancer studies including cancer diagnosis and therapy have been one of the most laborious tasks. Since its early detection significantly affects the following therapy steps, cancer diagnosis is very important. Despite researchers’ best efforts, the accurate and rapid diagnosis of cancer is still challenging and difficult to investigate. It is known that electrochemical techniques have been successfully adapted into the cancer diagnosis field. Electrochemical sensor platforms that are brought together with the excellent selectivity of biosensing elements, such as nucleic acids, aptamers or antibodies, have put forth very successful outputs. One of the remarkable achievements of these biomolecule-attached sensors is their lack of need for additional labeling steps, which bring extra burdens such as interference effects or demanding modification protocols. In this review, we aim to outline label-free cancer diagnosis platforms that use electrochemical methods to acquire signals. The classification of the sensing platforms is generally presented according to their recognition element, and the most recent achievements by using these attractive sensing substrates are described in detail. In addition, the current challenges are discussed.

1. Introduction

Cancer, which causes premature death in almost all countries of the world, maintains its position at first place even if it is sometimes replaced by cardiac disease. In particular, due to demographic effects and the trends of these effects in cancer incidence in different locations, it is expected that instances of cancer will approximately double in the next 50 years globally. However, cancer does not affect the population of all countries at the same rate, and it is predicted that there will be a higher increase in countries that can be classified as low–middle income [1,2]. The Global Cancer Statistics 2020 report shows that the most common cancer in men is prostate cancer, followed by lung cancer, colorectal cancer and liver cancer, whereas breast cancer and cervical cancer are the most commonly diagnosed cancers in women. In addition, according to the same report, what is striking is that an estimated 19.3 million new cancer cases were detected worldwide and approximately 10.0 million deaths were calculated due to cancer only in 2020 [3].
Regardless of the type, the diagnosis and treatment of cancer at an early stage is very important to reduce both cancer incidence and mortality rates. As the traditional cancer detection method, enzyme-linked immunosorbent assay (ELISA), which detects cancer-specific protein biomarkers and is called the gold standard, is widely known [4]. Also, genomic- and proteomic-based molecular methods such as polymerase chain reaction (PCR), immunohistochemistry (IHC) and radioimmunoassay (RIA) are used for cancer diagnosis [5]. In addition, various clinical tools such as magnetic resonance imaging (MRI), positron emission tomography (PET), endoscopy, sonography, X-ray, computed tomography (CT) and biopsy are extensively utilized [5,6,7]. However, although the mentioned methods and technologies are efficient, most of them are expensive, time-consuming, invasive and limited to the laboratories of some hospitals. Especially with imaging methods, detecting cancer tumors below the millimeter size may be inconclusive. Similarly, invasive methods such as biopsy have the same problems and difficulties in diagnosing early-stage cancer tumors [6].
Early-stage cancer diagnosis increases the survival rate of the patient [8]. In addition, early diagnosis offers several advantages that lead to more appropriate treatment for the patient and even reduce the severity of the cancer [9]. One of the biggest problems limiting early diagnosis in cancer detection is the nonappearance of obvious symptoms in the early stages of cancer; the other is not detecting sufficiently sensitive biomarkers [10]. From an economic point of view, it is known that the costs used for cancer treatment are increasing rapidly, and this cost is expected to increase up to USD 246 billion by 2030. Therefore, detecting cancer at an early stage can reduce the potential economic burden for the patient and society [11]. There is a crucial need to develop low-cost, sensitive, non-invasive (bio)sensors for early-stage cancer diagnosis. In general, biological biomarkers show the genetic characteristics of cancer cells and diagnosis/monitoring of cancer with a biomarker-based biosensor is seen as one of the most promising approaches [12]. These biomarkers can be deoxyribonucleic acid (DNA), ribonucleic acid (RNA), hormones, protein, enzymes and specific cells that can be found in human bodily fluids such as urine, serum, plasma and blood [13,14]. For this aim, electrochemical sensors have been widely used in the field of cancer diagnosis. There are valuable studies in the literature that include various approaches to detect different cancer types such as breast cancer [15], ovarian cancer [16], prostate cancer [17], pancreatic cancer [18] and lung cancer [19]. Electrochemical sensors are prominent tools because they are sensitive, selective, fast, cost-effective, instrumentable and can be performed as on-site analysis [20,21]. Different electrochemical sensing methods such as potentiometric, amperometric, conductometric, impedimetric and voltammetric are used to convert the obtained signal into useful analytical data. Detection methods in biosensors can be grouped as labeled or unlabeled depending on the use of labels as electroactive molecules or nanomaterials. However, labeled systems are complex and expensive as they require an extra labeling process. Conversely, label-free biosensors have shorter analysis time and simplicity and they offer good advantages [22,23,24].
In this review, current label-free cancer diagnosis platforms in the literature, including the last three years, in which the electrochemical method is used as a signal converter, are detailed. Biorecognition elements and mechanisms used in biosensor design for cancer diagnosis are emphasized. In addition, the immobilization method and immobilization matrices, which are important parameters for the activity and stability of a biorecognition element, are also the subject of this study. Finally, current challenges and future perspectives are discussed.

2. Electrochemical Techniques as a Sensing Mechanism

Electroanalytical studies are included as a sub-discipline of analytical chemistry, which includes charge transfers in addition to oxidation–reduction reactions [25]. Biorecognition elements, which are one of the parameters that make up the biosensor, are important components for the analyte to be detected. This component needs to be used with a converter so that a meaningful signal can be generated according to the analyte concentration [26]. In an electrochemical transducer system, detectable signals such as current, potential, impedance and conductivity are obtained as a result of the interaction of samples with a bioreceptor. In connection with these signals, electrochemical biosensors are included in various classifications such as amperometric, potentiometric, impedimetric and conductometric. In addition, voltammetric techniques are important and sensitive techniques to help analyte determination [27,28]. Electrochemical detection systems, which provide analytical advantages such as low cost, simple design and portable features, are platforms that can make sensitive and selective detections even in body fluids with complex matrices such as serum [29,30]. Therefore, these detection systems have attracted great attention in biosensor technology owing to their unique properties.
Voltammetric techniques have been commonly utilized. For example, differential pulse voltammetry (DPV), where a pulse is applied to the electrode and provides current measurement. Before the pulse is applied and at the end of each pulse, the current is measured and the difference between the currents is calculated. This procedure effectively reduces the background current due to linear increase, thus resulting in a faradaic current with no capacitive current. The biggest advantage of DPV is a low capacitive current, which leads to high sensitivity. Small steps in DPV also lead to narrower voltammetric peaks, and therefore, DPV is often used to distinguish analytes with similar oxidation potentials. Thus, this technique is preferred in electrochemical cancer biosensors as it exhibits very sensitive properties against the reduction and oxidation of bio-electrochemical species [31,32]. Cyclic voltammetry (CV) is one of the most common methods to obtain information about redox potentials and to investigate the mechanisms and kinetic parameters involved in the reactions of electroactive analytes. In this method, the current between the working and counter electrodes is monitored, but changes in the potential of the working electrode due to the reference electrode are also controlled [33]. In the electrochemical impedance spectroscopy (EIS) technique, the impedance change in both faradaic and non-faradaic modes is measured. As an example, in the measurement system in the faradaic mode, the change in the electron transfer rate caused by the aptamer–analyte interaction is examined. In measurement systems taken in non-faradaic mode, the surface capacitance change due to the aptamer–analyte connection is detected [34]. In the amperometric technique, the working electrode is kept at a constant potential that is sufficient to reduce or oxidize the analyte of interest and the resulting current is monitored over time. Potential selection is critical as only one potential is applied in this technique. Due to the monitoring of current over time at a constant potential, all dynamic changes in the current can be observed [31]. On the other hand, in a potentiometric system based on potential measurements, the principle of changing the potential with the concentration of the analyte is used in the measuring system with the help of a reference electrode with a fixed electrode potential. Besides cancer diagnosis, electrochemical techniques are also highly preferred in routine laboratory analysis and clinical and environmental monitoring analysis [35].
When electrochemical techniques are compared with each other, it is observed that each of them can have limitations in different aspects. For example, the sensitivity of the potentiometric method depending on the environment and temperature is an important limitation. For the limitations of other methods, it can be said that redox elements are needed in the amperometric technique, whereas EIS is sensitive to the environment and requires theoretical stimulation for data analysis [36]. Voltammetric techniques show high selectivity and sensitivity due to the voltammetric peak potential applied to the analyte. However, one of the major problems encountered with these techniques is obtaining overlapping voltammetric responses due to very similar oxidation peak potentials. Various recently developed materials and protocols are used to overcome this problem [37]. Besides this, choosing an appropriate sensing technique for analyte detection can minimize the limitations. Additionally, parameters such as pretreatments applied to the working electrode and the biofunctionality of the electrodes can have a great impact on the precise and effective determination [34].
The electrochemical transformations occurring at the interface of the label-free sensing platform are determined by the affinity between the analyte and the biorecognition elements, regardless of the use of labels [29]. Thanks to the detectable signals obtained by electrochemistry, these techniques are widely preferred not only for cancer detection and follow-up but also for the accurate and sensitive detection of analytes in areas such as the detection of different diseases and environmental and food control [38,39,40,41,42,43]. In an electrochemical biosensor, two different reactions can be observed as a result of the interaction of the electrode surface and the analyte: the first is the positive read signal called “signal-on”, and the other is the negative read signal called “signal-off” [44,45].
Label-free electrochemical biosensors are particularly interesting and important for studies in the biomedical field. In this type of electrochemical biosensor, the information in the reaction is converted into an electrical signal by the direct transfer of electrons between the electrode surface and the biorecognition elements as a result of the interaction between the biomolecule and the analyte [46]. Additionally, the surface characteristics of the electrodes significantly support improving the sensitivity of the biosensor. Therefore, surface modification is also important for good analytical performance. At this point, nanomaterials have been in the scope of scientists. The use of nanomaterials of different sizes, shapes and morphologies together with electrochemical transducers makes it possible to improve properties. Nanowires, metal/metal oxide nanoparticles, carbon nanotubes, graphene or graphene-like structures and conductive nanostructures such as polymers have provided more sensitive biosensors with high surface/volume ratios [47,48]. The scope of this study mainly covers the discussion of the technological developments and also problems/limitations in the development of label-free biosensors containing different biorecognition elements to serve cancer diagnosis.
Despite a lot of effort and good progress in the field of biosensors, it is seen as an inconsistency that only a few of them find a place in the commercial market. The first example of commercial biosensor is the enzymatic glucose biosensor, which is expected to have a market of USD 38 billion by 2027 [49]. This biosensor currently holds approximately 75% of the global biosensor market. There are still outstanding challenges, both to overcome the current constraints and to making the products available commercially. Firstly, understanding the mechanisms of biocatalytic work and charge transfers and also improvements in the properties of biorecognition elements that provide selectivity should be considered. In addition, the use of various nanoparticles and hydrogels has been reported to improve existing deficiencies, although not completely [50,51]. For this purpose, researchers are conducting detailed studies about the effects of parameters on biomolecule (such as enzymes) immobilization and the effect of these parameters on the performance of the biosensor platforms [52]. However, since the biomolecule redox reaction processes are still not fully known, in situ inspection techniques are used for evaluation [53]. Some of the obstacles in the transformation of biosensor studies from laboratory to commercial products are performance and nonspecific surface interaction problems in various body fluids, which have complex matrices [49].
Although electrochemical methods provide several advantages, each method may also have limitations. It is particularly important to focus on and discuss these limitations to put the developed technologies into clinical practice. Reducing or overcoming all the disadvantages could help to develop more accurate and sensitive electrochemical cancer biosensors. More effective platforms for early diagnosis can be created with a multidisciplinary study. In addition, the detection of new cancer biomarkers will greatly benefit the facilitation of early-stage diagnosis and thus the management and control of the cancer disease process. It is expected that the label-free electrochemical methods will increase in reliability after the difficulties we have mentioned have been overcome. As a result, they will find a regular use in the clinical field. To strengthen this reliability, novel and advanced electrochemical cancer biosensors with different perspectives need to be developed.

3. Importance of a Label-Free Electrochemical Sensing Platform

A typical electrochemical biosensor is expected to convert signals that are related to the presence of the analyte molecules into measurable quantities with the help of the biorecognition unit. In some cases, various markers/labels or tags are used for the detection of the analyte and the signal is obtained in conjunction with them. These biosensor systems are called label-based biosensors. The use of these labels, which are commonly classified as radioactive-, fluorescent- or chemiluminescence-based, is time consuming and laborious because it requires an extra process. More importantly, it is thought that in this case, the affinity between the biorecognition element and the analyte may be adversely affected. To eliminate these limiting factors, unlabeled detection systems have become highly preferred in recent years. If a direct measurement is made with the biorecognition system, this is called a label-free biosensor system [54].
In a typical label-free biosensor design, sensing can be performed by converting it to optical [55], mechanical [56] or electrical [57] signals and more accurate information can be provided as biorecognition systems are directly used. Within this classification, electrochemical label-free biosensors can be used actively in the field and can be also implanted in the body to detect biological analytes, increasing their future potential [58]. Various electrodes with different biorecognition elements and composite designs have been developed for analytes such as gliotoxin [59], microRNA (miRNA) [60], bacterial pathogens [61] and aflatoxin-B1 [62] in this biosensor group, which combines the advantages of both the electrochemical method and the label-free platform. For the continuation of the remarkable progress of the mentioned electrochemical label-free biosensors, a better understanding of the current working processes is required for the creation of sensitive and selective biosensing systems that find application in wider use. Based on this idea, we have detailed and discussed cancer studies classified on different biorecognition elements.

4. Biorecognition Elements for Label-Free Electrochemical Cancer Diagnosis

Basically, antibodies, aptamers, nucleic acids and cells are immobilized to surfaces/interfaces to achieve affinity and selective biorecognition. In this part, the classification of the label-free electrochemical cancer detection systems is divided into categories according to the type of the biorecognition element. Besides this classification, electrode material and the detection technique are also highlighted. Figure 1 demonstrates the schematic presentation of the label-free electrochemical cancer biosensors with successful electrode modifications, such as nanotechnology-based materials, biorecognition immobilization protocols and some of the powerful electrochemical detection techniques.

4.1. Nucleic-Acid-Based Label-Free Cancer Biosensors

Nucleic acids are natural biopolymers that store genetic information in humans and almost all organisms [63]. Nucleic acids include DNA and RNA, which are composed of nucleotides. The well-known specific hybridization feature between nucleic acid chains also constitutes the main detection principle of DNA biosensors [64]. The development of biosensors for the detection of DNA sequences is important because of its application in gene identification, molecular diagnosis and drug screening [65]. Nucleic acids can be affected by environmental conditions such as temperature and pH [66]. Nevertheless, in many studies electrochemical signal amplification by means of nucleic acids has been successfully developed for cancer applications [67,68].
Studies in recent years show that excessive secretion of microRNAs is associated with malignancies that cause cancer [15,69,70,71]. In one study, Zhao et al. proposed MXene-molybdenum disulfide (MoS2) constructs with thionine and gold nanoparticles for the label-free electrochemical detection of microRNA-21, which plays an important role in the emergence of cancer associated with proliferation/differentiation in cells. The modification of the prepared nanocomposite on glassy carbon electrode (GCE) was performed by drop casting. Then, the hairpin capture probe was dropped onto the modified electrode. The hybridization event was carried out in the presence of the target and a hairpin probe 2. The detection method was square wave voltammetry (SWV). Thanks to this structure, the capture probe immobilization was improved, the amplification of the electrochemical signal was achieved and microRNA-21 detection in the linear measurement range of 100 fM to 100 nM was obtained with a detection limit of 2 fM [72].
Pothipr et al. described a gold nanoparticle-dye/poly(3-aminobenzylamine)/two-dimensional molybdenum selenide (MoSe2)-based electrochemical label-free biosensor for breast cancer diagnosis that could detect cancer antigen 15-3 and microRNA-21 simultaneously. Based on the complexity of the immune system in the human body and therefore the inadequacy of cancer assays using single biomarker systems, they introduced this bidirectional detection platform produced on a two-screen printed carbon electrode. DPV was used for the evaluation of the electrochemical performance of the biosensor and the detection limit was found to be 1.2 fM for microRNA-21 detection [73]. Jafari-Kashi et al. presented a DNA biosensor for the detection of cytokeratin 19 fragment 21-1, which is associated with lung cancer. They preferred DPV as an electrochemical technique to examine the interaction between the capture probe and target using GCE modified with reduced graphene oxide, polypyrrole, silver nanoparticles and single-stranded DNA (ssDNA). With this technique, no peak was detected before DNA hybridization, but a distinctive peak was obtained after hybridization according to the oxidation of guanine. They declared that the label-free DNA biosensor showed a good result for detection of cytokeratin 19 fragment 21-1, with a wide linear measurement range and a 2.14 fM limit of detection [74]. Avelino et al. presented a polypyrrole film containing DNA immobilized chitosan/zinc oxide nanoparticles for the diagnosis of myelocytic leukemia by BCR/ABL fusion gene detection. Oxidation and reduction steps were observed in line with the voltammetric measurements taken in 10 mM [Fe(CN)6]3–/4–. It is also stated that the biosensor was designed as a result of bioactivity tests and could be used as a new biosensing platform that enabled the identification of early-stage cancer [75].

4.2. Aptamer-Based Label-Free Cancer Biosensors

Aptamers are single-stranded DNA or RNA molecules that can usually be synthesized using an in vitro method. In fact, RNA-based aptamers were first found in 1990, followed by DNA-based aptamers, with the development of in vitro selection/amplification for the isolation of RNA sequences that could specifically bind to molecules [76]. In aptamer-based electrochemical sensors, it is necessary to be able to detect the conformational changes caused by the presence of the aptamer on the electrode surface for obtaining a signal [77]. Aptamers are widely used in the development of biosensors due to their high specificity, easy synthesis, simple modification and high chemical stability [78]. They offer the advantages of more cost-effective production, easy modification and thermal stability, especially when compared with monoclonal antibodies. After the aptamers are immobilized on a conductive matrix, their redox-active moieties allow the formation of aptamer–target complexes and thus the design of various electrochemical biosensors with the realized electron transfer properties [76]. The most important problem in this electrochemical process can be the generation of a determinable signal between the target analyte and the aptamer. In order to solve this problem, electrochemically active labeling units such as hemin [79], ferrocene [80] and methylene blue [81] have been introduced. However, labeling of aptamers introduces known disadvantages such as time consumption, poor affinity performance and cost [82].
In recent years, aptamers have attracted great interest in electrochemical label-free biosensor design, which has applications in the diagnosis and follow-up of various cancers. Label-free aptasensors also require an increased surface area to improve weak signal intensity. Nanomaterials contribute greatly to increasing the surface area because they act as electron-transfer tunnels, which increase the electrical communication between the redox regions of the aptamer and the electrode surface [83]. Zhang et al. developed a label-free aptasensor for the detection of cancer antigen 125 by immobilizing aptamer on the surface of nickel hexacyanoferrate nanocubes/polydopamine functionalized graphene. DPV was utilized for electroanalytical studies in this work, which was designed to provide a detectable electrochemical response with the help of increasing surface area and conductivity. Thanks to the insulating structure formed as a result of the combination of aptamer and cancer antigen 125 (CA125), or in other words aptamer–CA125 complex, the peak current value decreased as the analyte concentration increased. The linear measurement range and limit of detection were calculated as 0.10 pg mL−1–1.0 μg mL−1 and 0.076 pg mL−1, respectively. The measurements were carried out in phosphate buffer solution (PBS) [82]. In another study, a paper-based electrochemical label-free aptasensor was fabricated for the detection of epidermal growth factor receptors. Interestingly, the concept of origami as a valve for a paper-based biosensor was used in this study. As a result of the biochemical reaction, the data became an electrochemical response with the presence of the nanocomposites containing amino functionalized graphene/thionine/gold. This system in the form of origami was designed to increase the penetration of the liquid and shorten the time taken for flow, resulting in a shorter test time. The linear concentration range obtained with the sensor was from 0.05 ng mL−1 to 200 ng mL−1 and it had a detection limit of 5 pg mL−1 [84].

4.3. Antibody-Based Label-Free Cancer Biosensors

Antibodies are protective proteins produced by the immune system in response to the presence of antigens, including pathogens and toxic materials [78]. Biosensors that offer the advantages of high binding affinity and specificity and use antibodies for biorecognition take the advantage of the high affinity between antibodies and antigens for detection and are called immunosensors [85,86]. However, there are some parameters that limit their use. Apart from being adversely affected by environmental conditions and having difficulties for storage, it can be said that the production of polyclonal antibodies in animals is difficult and costly. Moreover, polyclonal antibodies may lack selectivity as they can have affinity for different epitopes [87]. With the help of the new and improved sensor interfaces developed in recent years, some disadvantages have been overcome and many antibody-based sensitive and selective label-free electrochemical biosensors have been designed. Also, these limitations pave the way for the development of new forms of biorecognition units that can replace antibodies, thus introducing new biosensor projections to the field.
Various electrochemical techniques have been used for antibody-based biosensors for gastric cancer [88], breast cancer [89,90,91,92], ovarian cancer [93,94,95,96], bladder cancer [97], colorectal cancer [98], lung cancer [99], prostate cancer [100,101,102,103,104,105], liver cancer [106] and more. In a study for a label-free electrochemical immunosensor developed for early-stage detection of prostate cancer, the surface of the indium tin oxide electrode was firstly coated with chitosan and reduced graphene oxide, and then the specific polyclonal anti-prostate-specific antigen (PSA) antibody as a recognition element was immobilized on the surface. It was determined that a linear decrease had been observed in the peak current values of the redox probe by using DPV with increasing concentrations of the antigen. It is reported that the linear measurement range determined for prostate-specific antigen detection was between 1 pg mL−1 and 5 ng mL−1, and the limit of detection was 0.8 pg mL−1 [107].
CA125 was detected by DPV using a layer-by-layer assembly of ordered mesoporous carbon, gold nanoparticles and MgAl-layered double hydroxides containing ferrocene carboxylic acid composite. It is explained that the conductivity increased significantly with the addition of the ferrocene component to the composite. The electrochemical performance of the biosensor was determined based on the change of the peak current observed in the voltammogram at +0.27 V according to the ferrocene in the presence of different CA125 antigen concentrations. It is stated that the peak current value obtained with the increase in the CA125 concentration changed inversely, since the complex formed between the antigen and the antibody. The linear measuring range and limit of detection of the biosensor were described as 0.01 U mL−1–1000 U mL−1 and 0.004 U mL−1, respectively [108]. A label-free sandwich type biosensor was developed for the electrochemical detection of cytokeratin fragment antigen 21-1 (CYFRA 21-1), a lung cancer biomarker. An antibody–antigen–antibody sandwich structure was formed between the 4-(2-trimethylsilylethinyl)benzoic acid gold electrode used as a bridge and the poly(ε-caprolactone)-b-poly(ethylene oxide) copolymer. The linear concentration range and limit of detection for the sensor determined by electrochemical impedance spectroscopy were declared as 1.0 pg mL−1 to 10 ng mL−1 and 0.125 pg mL−1, respectively. According to the impedance results, the electrochemical responses showed a linear response with the concentration of CYFRA 21-1 [109].
Liu et al. developed a gold nanoparticle/polyethyleneimine/reduced graphene oxide nanocomposite for the electrochemical detection of matrix metalloproteinase-1, a cancer biomarker, based on the knowledge that gold nanoparticles were supportive in maintaining the reversibility of redox reactions in electroanalytical reactions. They determined that the biosensor performance obtained by DPV had an operating range of 1 ng mL−1 to 50 ng mL−1. In this work, the peak current value obtained from voltammetry decreased due to the increased antigen concentration blocking on the electrode surface. In the electrochemical measurements taken in 5 mM Fe(CN)63−/4− medium, it is stated that an insulating layer was formed due to the antigen–antibody complex, and therefore, a repulsive electrostatic interaction occurred between the antigen and Fe(CN)63−/4− [110]. Zhu et al. also developed a carbon-based nanocomposite to take advantage of its high surface area and good conductivity properties. The surface was used for the construction of an immunosensor for the detection of alpha-fetoprotein, which is a liver cancer biomarker. They calculated a linear measurement range of 0.10 ng mL−1 to 420 ng mL−1 and a limit of detection of 0.03 ng mL−1 using square wave voltammetry, a method that could suppress background current and provide sensitivity to the biosensor system [106].

4.4. Cell-Based Label-Free Cancer Biosensors

The use of cells as a biorecognition element dates back to the early 1970s and it is still preferred today. Cells offer an interesting alternative to other biorecognition units such as antibodies, enzymes and nucleic acids thanks to their relatively easy production and lower cost than antibodies and purified enzymes. As an example, since whole cells offer a multi-enzyme alternative, they can be preferred in the development of biosensors for the simultaneous determination of various analytes. In addition, cell-based biosensors enable in situ monitoring using suitable substrates [78,111,112]. However, some limitations such as maintenance and immobilization of cells can arise [113].
Human cervical carcinoma (HeLa) cells were used as a biorecognition unit in an electrochemical label-free cytosensor to evaluate the anticancer activity of pinoresinol, which had biological properties such as anticancer, anti-inflammatory and antifungal effects. HeLa cells were immobilized on a GCE surface modified with multi-walled carbon nanotubes and gold nanoparticles, and the performance of the biosensor was evaluated by electrochemical impedance spectroscopy with different pinoresinol concentrations. The limit of detection value for the biosensor, which showed a linear correlation with the pinoresinol concentration range of 102 to 106 cells mL−1, was reported as 102 cells mL−1 [114]. Another cell-based label-free electrochemical biosensor was developed to investigate the interactions of cancer cells (HepG2 cells and A549 cells) with molecules and to screen anticancer drugs. Cancer cells were immobilized on the GCE coated with N-doped graphene–Pt nanoparticles–chitosan and polyaniline. It is stated that this electrode surface might be suitable for examining different cell lines by changing the targeted cells as a result of the electrochemical properties examined by DPV with its large surface area and catalytic properties [115].
Liu et al. carried out the detection of cell surface glycan that played an important role in processes such as cancer cell metastasis by means of a nano channel ion channel of porous anodic alumina hybrid combined with an electrochemical detector. Thus, the enhanced ionic current caused by the array nano channels along with the ionic current rectification gave a precise current response. The alumina was functionalized with aminopropyltriethoxysilane and glutaraldehyde to immobilize the cell surface glycan. The linear working range was obtained from 10 fM to 10 nM, and the limit of detection was calculated to be approximately 10.0 aM. It is stated that this biosensor was a promising alternative that could be used in cancer diagnosis and an important platform for label-free detection of cell surface glycan [116].
Despite the advantages of cell-based electrochemical biosensors, there are also various disadvantages faced by designers such as reproducibility and inability to selectively place cells at detection sites [117]. In addition, some difficulties in terms of electrochemical techniques such as amperometric and impedimetric have been reported in the literature. For example, the difficulties often observed in electrochemical impedance spectroscopy-based studies are that the measured electrochemical response is the total change produced by a set of cells and poor selectivity. Emerging technology, nanomaterial selection, new immobilization matrices, integration of different transducer mechanisms and advances in the control of the sensor interface are some of the promising approaches to overcome these challenges [105,106].

5. Immobilization Strategies of Biorecognition Elements

Biorecognition element immobilization or its integration is one of the important processes to be considered, since this step thoroughly affects the analytical performance of all types of biosensors. The efficient immobilization of the biorecognition element is a process applied to overcome the problems such as loss of activity and stability by integrating biomolecules into a suitable support material. The immobilization methods are classified as adsorption, covalent bonding, cross-linking, etc., according to the type of the biomolecule to be immobilized and the structure of the immobilization surface [118]. These methods are illustrated in Figure 2.
In Table 1, the immobilization methods used by some of the studies within the scope of this review are indicated. Some cancer detection studies in the literature for recent years, different biorecognition units, other biosensor components and the parameters used in these studies are listed. Metals, metal oxides, conductive polymers, biopolymers, carbon-based structures, quantum dots and their composites [93,100,107,109,119,120] have been used as the immobilization matrices for label-free electrochemical cancer biosensors. In general, electrostatic interactions can have negative effects on the stability of the biorecognition element or the repeatability of the biosensor [121,122]. However, these methods, which have very simple processes, are still actively used in the surface immobilization of many electrodes. The entrapment method also offers specific properties and contributes to the improvement of chemical and thermal stability. However, leakage and low biological activity limit this method. To overcome the leakage problem, crosslinkers are preferred in the immobilization step. However, at this stage, excessive chemical requirements are necessary [123].
In the study of Yaiwong et al., an immunosensor for label-free electrochemical cancer detection was developed. Electrostatic interaction was carried out for the immobilization of the anti-metalloproteinase-7 (MMP-7) capture antibody, which was used as a biorecognition element, on the surface of the screen-printed carbon electrode (SPCE) coated with two-dimensional (2D) MoS2/graphene oxide [124]. More commonly, immobilization methods by covalent or cross-linking over carboxyl or amine groups are robust and reproducible ways to obtain an effective biosensor interface. Glutaraldehyde or carbodiimide structures that act as bridges in these binding reactions are preferred [121]. As an example, Yan et al. coated the surface of an indium tin oxide electrode with chitosan-modified reduced graphene oxide nanocomposite for prostate cancer detection. In order to detect prostate-specific antigens with this biosensor, they immobilized the recognition antibodies onto the electrode surface by covalent bonding. Chitosan naturally provided a large number of amino groups to the electrode surface, and glutaraldehyde, a bifunctional bridge, was used for covalent immobilization of the anti-PSA antibody with amino groups. Thus, a label-free electrochemical immunosensing platform based on antibody–antigen affinity was developed [107].
Echeverri et al. immobilized the anti-β-1,4-galactosyltransferase-V (β-1,4-GalT-V) antibody biorecognition element on the self-assembled monolayer (SAM)-coated SPCE by covalent bonding for the detection of colorectal cancer. The SAM provided a carboxylic acid group that allowed for antibody binding [98]. Generally, N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) pairs are used for this type of covalent bonding. In this way, a bridge is formed between the amine and carboxyl groups and a high binding efficiency is achieved [121]. Although covalent bonding seems to offer good efficiency and is an advantageous method, it can also have various disadvantages in some cases. For example, denaturation may occur due to the undesirable site orientation of the biorecognition element, and in addition, the bridging compounds are needed to use in the covalent bonding reaction. Therefore, there can be a decrease or disappearance of the biocatalytic effect expected from the biorecognition unit [125]. Moreover, covalent bonding, which causes a tight binding, can also restrict the movement of the biorecognition elements, which may also cause a loss of activity [126].
Table 1. Electrochemical-based label-free biosensors for cancer detection: biorecognition elements, sensor platforms, immobilization methods and electroanalytical performances.
Table 1. Electrochemical-based label-free biosensors for cancer detection: biorecognition elements, sensor platforms, immobilization methods and electroanalytical performances.
Biorecognition
Elements
Other ComponentsImmobilization MethodCancer TypeAnalyteElectrochemical
Technique
Limit of
Detection
Linear RangeReferences
Biotinylated DNA probeMulti-walled carbon
nanotubes non-covalently functionalized with avidin
Covalent bindingBreast cancerBRCA1Electrochemical impedance spectroscopy330 aM1.0 fM–10 nM[67]
Three-dimensional
(3D) DNA walker
Au nanoparticles/encapsulation of glucose oxidase in zeolitic imidazolate framework-8 (ZIF-8)Electrostatic adsorptionCancerMicroRNADifferential pulse voltammetry29 pM0.1 nM–10 μM[68]
Hairpin probe (H1)MXene/MoS2/thionine/Au nanoparticlesAdsorptionCancerMicroRNA-21Square wave voltammetry26 fM100 fM–100 nM[72]
Anti-CA 15-3 antibodiesGold nanoparticle-dye/poly(3-aminobenzylamine)/two dimensional
MoSe2/graphene oxide
Covalent bindingBreast cancerCA 15-3Differential pulse voltammetry0.14 U mL−10.0–500 U mL[73]
DNA-21Gold nanoparticle-dye/poly(3-aminobenzylamine)/two dimensional
MoSe2/graphene oxide
AdsorptionBreast cancerMicroRNA-21Differential pulse voltammetry1.2 fM0.0–1000 pM[73]
DNAReduced-graphene oxide/polypyrrole/silver nanoparticlesCovalent bindingLung CancerCytokeratin 19
fragment 21–1 (CYFRA21-1)
Differential pulse voltammetry2.4 fM1.0 × 10−14–1.0 × 10−6 M[74]
DNA-aptamer probeα-Fe2O3/Fe3O4@AuAdsorptionOvarian cancerCA125Differential pulse voltammetry2.99 U mL−15–125 U mL−1[77]
Aminoated CA 125 aptamersNickel hexacyanoferrate nanocubes/polydopamine
functionalized graphene
Covalent bindingOvarian cancerCA125Differential pulse voltammetry0.076 pg mL−10.10 pg mL−1–1.0 μg mL−1[82]
AptamerGraphene oxide functionalized with aspartic acidCovalent bindingCancerCytochrome cDifferential pulse voltammetry0.74 nM10 nM–100 μM[83]
Epidermal growth factor receptor (EGFR) aptamersAmino-functionalized graphene/
thionine/gold particle nanocomposites
Covalent bindingCancerEGFRDifferential pulse voltammetry5 pg mL−10.05–200 ng mL−1[84]
CA72-4 antibodiesCarbon nanotube–graphene oxide hybridCovalent bindingGastric cancerAntigen
72-4
Differential pulse voltammetry0.4 U mL−12.0–80.0 U mL−1[88]
Calreticulin antibodyElectrodeposited single-walled carbon nanotubes and polymerized oxiran-2-yl methyl 3-(1H-pyrrol-1-yl) propanoate monomerCovalent bindingBreast cancerCalreticulinElectrochemical impedance spectroscopy0.0046 pg mL−10.015–60 pg mL−1[89]
Cancer antigen (CA 15-3) antibodyTernary silver/titanium dioxide/reduced graphene oxides nanocompositeCovalent bindingBreast cancerCA 15-3 antigenAmperometry0.07 U mL- 10.1–300
U mL−1
[90]
Anti-cancer antigen (CA125) antibodyBoron nitride nanosheetsPhysical adsorptionOvarian
cancer
CA125Differential pulse voltammetry1.18 U mL−15–100 U mL−1[94]
Anti-nuclear matrix protein 22 (NMP22)Reduced graphene oxide/tetraethylene pentamine/Cu-based metal organic
frameworks deposited silver nanoparticles
Covalent bindingBladder cancerNMP22Differential pulse voltammetry33.33 fg mL−10.1 pg mL−1–1000 ng mL−1[97]
Anti-β-1,4-galactosyltransferase-V (β-1,4-GalT-V) antibodySelf-assembled monolayer-coated screen-printed gold electrodeCovalent bindingColorectal cancerβ-1,4-GalT-V glycoproteinElectrochemical impedance spectroscopy7 pM5–150 pM[98]
Prostate-specific membrane antibody (PSMA)Cysteamine-modified gold nanoparticlesCross-linkingProstate cancerPSMA proteinDifferential pulse voltammetry0.47 ng mL−10–5 ng mL−1[100]
Anti-prostate-specific antigen (PSA)Chitosan, graphene, ionic liquid and ferrocene cryogelChemical adsorptionProstate cancerPSADifferential pulse voltammetry4.8 × 10−8 ng mL−11.0 × 10−7–1.0 × 10−1 ng mL−1[103]
Anti-alpha-fetoprotein (AFP)MnO2 functionalized mesoporous carbon hollow sphereCross-linkingLiver cancerAFPSquare wave voltammetry0.03 ng mL−10.10–420 ng mL−1[106]
Polyclonal anti-PSA antibodyChitosan–graphene-modified indium tin oxide electrodeCovalent bindingProstate cancerProstate-specific antigenAmperometry0.8 pg mL−11–5 ng mL−1[107]
Primary antibody (Ab1)Linear poly(ε-caprolactone)-
b-poly(ethylene oxide) copolymer
Cross-linkingLung cancerCYFRA 21-1Electrochemical impedance spectroscopy0.125 pg mL−11 pg mL−1–10 ng mL−1[109]
Human cervical carcinoma (HeLa) cellsCarboxylated multiwalled
carbon nanotubes/gold nanoparticles
AdsorptionCervical cancerPinoresinolElectrochemical impedance spectroscopy102 cells mL−1102–106 cells mL−1[114]
Anti-matrix metalloproteinase (MMP)-7 capture antibodiesTwo-dimensional molybdenum disulfide/graphene
oxide nanocomposite
Electrostatic interactionsPancreatic and colorectal cancersMMP-7Differential pulse voltammetry0.007 ng mL−10.010–75 ng mL−1[124]
Antibodies specific to IL-8 (Anti-IL-8)Silver molybdate nanoparticlesCovalent bindingOral cancerIL-8Differential pulse voltammetry90 pg mL−11 fg mL−1–40 ng mL−1[127]
Human epidermal growth factor receptor 2 (HER2) antibodyFe3O4/TMU-21/multi-walled carbon nanotubesCross-linkingBreast cancerHER2Amperometry0.3 pg mL−11.0 pg mL−1–100 ng mL−1[128]
Carcinoembryonic antigen (CEA) antibodyFe3O4@Au nanoparticlesAdsorptionCancerCEALinear sweep voltammetry0.10 pg mL−10.001–100 ng mL−1[129]
DNAExo-III-assisted target recycling and dual enzymesCovalent bindingOral cancerORAOV1Electrochemical impedance spectroscopy0.019 fM0.05 fM–20 pM[130]
Capture strandAu nanoparticlesAdsorptionBrain cancersCerebrospinal fluid microRNAsDifferential pulse voltammetry56 fM0.5–80 pM[131]
CEA aptamer (AptGAC-
P)
6-Mercapto-1-hexanol (MCH)/cpDNA2/goldAdsorptionCancerCEADifferential pulse voltammetry0.24 ng mL−12–45 ng mL−1[132]
Carcinoembryonic antigen aptamerAu nanoparticlesSelf-assemblyLung cancerCEAElectrochemical impedance spectroscopy0.085 ng ml−10.2–15.0 ng ml−1[133]
AS1411 aptamerReduced graphene oxide–chitosan–gold nanoparticleCovalent bindingBreast cancerMCF-7 cancer cellsElectrochemical impedance spectroscopy4 cells mL−11 × 10–1 × 106 cells mL−1[134]
DNA aptamerGold electrodeCovalent bindingCancerCluster of differentiation-44 (CD44)Electrochemical impedance spectroscopy0.087 ng mL−10.1–1000 ng mL−1[135]
PDGF-BB affinity aptamersCarboxyl-functionalized photoresist-derived carbonCovalent bindingCancerPlatelet-derived growth factor-BB (PDGF-BB)Cyclic voltammetry7 pM0.01–50 nM[136]
DNAGraphene
oxide–chitosan/polyvinylpyrrolidone–gold nano urchin
Covalent bindingLung CancermiR-141Square wave voltammetry0.94 fM2.0–5.0 × 105 fM[137]
DNA-21Graphene/polypyrrole/gold nanoparticlesElectrostatic interactionCancermiRNA-21Differential pulse voltammetry0.020 fM1.0 fM to 1.0 nM[138]
DNAChitosan-capped gold
nanoparticles
Electrostatic interactionCervical cancerHPV-16Cyclic voltammetry/square wave voltammetry1.0 pM1 pM–1 μM[139]
ssDNAL-cysteine
functionalized ZnS quantum dots
Covalent bindingOvarian cancermiR-200aElectrochemical impedance spectroscopy8.4 fM1.0 × 10−14–1.0 × 10−6 M[140]
ssDNAReduced graphene oxide/polyaniline nanofibersElectrostatic interactionBreast cancerBRCA1Differential pulse voltammetry3.01 × 10−16 M1.0 × 10−15–1.0 × 10−7 M[141]
acpcPNA-T9 probeAg@Au core–shell nanoparticles electrodeposited on graphene quantum dotsAdsorptionCancermiRNA-21Chronoamperometry5 pM5 pM–5 mM[142]
Although the immobilization of biorecognition elements on the surface of the biosensing platform is a very important step for the design of sensitive, selective and long operational lifetime biosensors, it is clear that each method has several advantages and disadvantages. Various factors such as the immobilization matrix and the charge or functional groups of the biorecognition units guide the selection of the appropriate method, and thus, effective interfaces are created.

6. Label-Free Electrochemical Cancer Biosensors for Point-of-Care Applications

Label-free electrochemical biosensors have a high capability of being adapted into point-of-care (POC) systems that can be used for outside the laboratory testing to minimize the need for healthcare services such as hospitals [14,143,144,145]. In POC testing particularly, microfluidic devices have attracted great attention lately for effective and accurate cancer diagnosis owing to their ability to separate analytes at a good resolution in a rapid reaction time and to minimize the handling errors and costs [143]. As a result, promising detection systems with high performances are acquired with the elimination of the need for trained personnel. Recently, in the study by Keyvani et al., a POC sensing device for the detection of cervical cancer was developed for whole blood. This system identified cancer circulating DNA with high purity by the help of a graphene oxide-dependent electrochemical sensor platform by using differential pulse voltammetry [146]. In another study, Ming et al. fabricated a cellulose-paper-based POC testing with the modification of amino redox graphene, thionine, streptavidin integrated gold nanoparticles and chitosan for the detection of biomarker 17β-estradiol, which may be associated with breast cancer. The detection strategy, realized with differential pulse voltammetry in phosphate buffer solution, was carried out via the interaction of the target biomarker and its biotin-modified aptamer on the surface of the paper. The linearity of the label-free sensor was between 10 pg mL−1 and 100 ng mL−1, with a limit of detection value of 10 pg mL−1 [147].
Besides microfluidic devices, multiplex systems that can detect multiple analytes associated with cancer have several advantages in terms of label-free point-of-care testing. As an example, Kuntamung and his colleagues achieved simultaneous detection of breast cancer biomarkers: mucin1, cancer antigen 15-3 and human epidermal growth factor receptor 2 depending on the formed antibody and antigen interactions. For this purpose, redox species and antibody-conjugated polyethylenimine-modified gold nanoparticles were utilized as the modification elements of a SPCE. In addition to multiplex detection performance, the label-free biosensor kept 90% of its initial responses obtained via voltammetry [92]. In another approach that contained the fabrication of a flexible screen-printed electrode system, carcinoembryonic antigen was detected on graphene–ZnO nanorods deposited on a polyethylene terephthalate substrate with a screen-printed electrode by Chakraborty et al. ZnO nanorods were functionalized with aptamers and the resulting surface improved the mass transport through the electric field application. This system was integrated into smartphone interface technology and a handheld potentiostat. The linearity of the label-free sensor was between 0.001 pg mL−1 and 10 pg mL−1, with a limit of the detection value of 1 fg mL−1 by using electrochemical impedance spectroscopy. The results were also validated using a commercial ELISA kit [148].
The use of label-free POC testing in cancer diagnosis is in increasing demand in recent years since POC systems yield rapid decisions, more frequent testing to monitor wellness, eliminate the need for trained staff and utilize small specimen volumes. In addition, they are cost-effective. Despite these advantages, they are still more open to false positives or negatives and incorrect interpretations. Also, these sensing platforms have a risk of external interference since the environment is not as well controlled as in laboratories. In some cases, the sampling procedure can be inconvenient, such as in cancer diagnosis protocols. Indeed, POC-based electrochemical cancer biosensors are not yet available on the market. One of the additional reasons for this issue could be the distance between physicians and electrochemical biosensor developers. It is believed that multidisciplinary studies between them will improve the quality of the developed platforms. Additionally, shelf-life and production control are important parameters to improve their commercialization capacity [149,150,151,152]. However, electrochemically based POC systems are promising tools for the accurate and fast detection of cancer with their overall characteristics.

7. Conclusions and Future Perspectives

In the current review, we have summarized the recent achievements and progresses around label-free electrochemical biosensors that are utilized for cancer detection. Since the type of biorecognition element is an important key parameter to enhance the selectivity of the detection, the classification of the biosensors is made according to the types of recognition elements. Besides the achievements, the current challenges are also outlined in detail. Label-free detection systems are in urgent demand owing to their properties, including reducing labored modification steps and interference effects.
The growing demand on clinical research and the medical industry for cancer studies has pushed scientists to perform early detection with practical analytical tools instead of time-consuming and back-breaking methods. In addition to detection, isolation of the cancer cells is also important to increase the survival rates and quality of life. The design and development of early-cancer diagnosis platforms has been one of the hot topics of the last decades. The recent advances in the field of cancer diagnosis show that electrochemical sensing methodologies have an important impact on the accurate, rapid and sensitive detection of cancer types. Particularly, label-free electrochemical biosensors maintain predominant features to obtain reliable, cost-effective and selective cancer diagnosis that can serve for future implementations. With the addition of advanced materials such as nanomaterials, not only sensitivity of the biosensors but also the selectivity of them can be significantly improved. Surface modification makes bare electrode substrates available and suitable for biorecognition element immobilization. Recent studies on label-free and electrochemical biosensing of cancers indicate how promising and operational these biosensors are. It is certain that their advantages will certify more powerful medical applications in the near future with the support of growing materials science technology.

Author Contributions

V.S.: Conceptualization, Writing—Original draft preparation, Review and Editing, Supervision; F.K.: Conceptualization, Writing—Original draft preparation, Review and Editing, Supervision, Project Administration. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

F.K. acknowledges Turkish Academy of Sciences as an associate member.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Soerjomataram, I.; Bray, F. Planning for tomorrow: Global cancer incidence and the role of prevention 2020–2070. Nat. Rev. Clin. Oncol. 2021, 18, 663–672. [Google Scholar] [CrossRef] [PubMed]
  2. Bray, F.; Jemal, A.; Grey, N.; Ferlay, J.; Forman, D. Global cancer transitions according to the Human Development Index (2008–2030): A population-based study. Lancet Oncol. 2012, 13, 790–801. [Google Scholar] [CrossRef] [PubMed]
  3. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  4. Qian, L.; Li, Q.; Baryeh, K.; Qui, W.; Li, K.; Zhang, J.; Yu, Q.; Xu, D.; Liu, W.; Brand, R.E.; et al. Biosensors for early diagnosis of pancreatic cancer: A review. Transl. Res. 2019, 213, 67–89. [Google Scholar] [CrossRef] [PubMed]
  5. Cui, F.; Zhou, Z.; Zhou, H.S. Measurement and analysis of cancer biomarkers based on electrochemical biosensors. J. Electrochem. Soc. 2019, 167, 037525. [Google Scholar] [CrossRef]
  6. Khanmohammadi, A.; Aghaie, A.; Vahedi, E.; Qazvini, A.; Ghanei, M.; Afkhami, A.; Hajian, A.; Bagheri, H. Electrochemical biosensors for the detection of lung cancer biomarkers: A review. Talanta 2020, 206, 120251. [Google Scholar] [CrossRef]
  7. Li, G.; Wu, J.; Qi, X.; Wan, X.; Liu, Y.; Chen, Y.; Xu, L. Molecularly imprinted polypyrrole film-coated poly (3,4-ethylenedioxythiophene):polystyrene sulfonate-functionalized black phosphorene for the selective and robust detection of norfloxacin. Mater. Today Chem. 2022, 26, 101043. [Google Scholar] [CrossRef]
  8. Ahlquist, D.A. Universal cancer screening: Revolutionary, rational, and realizable. NPJ Precis. Oncol. 2018, 2, 23. [Google Scholar] [CrossRef] [Green Version]
  9. Sadighbayan, D.; Sadighbayan, K.; Khosroushahi, A.Y.; Hasanzadeh, M. Recent advances on the DNA-based electrochemical biosensing of cancer biomarkers: Analytical approach. TrAC Trends Anal. Chem. 2019, 119, 115609. [Google Scholar] [CrossRef]
  10. Wang, H.; Peng, R.; Wang, J.; Qin, Z.; Xue, L. Circulating microRNAs as potential cancer biomarkers: The advantage and disadvantage. Clin. Epigenet. 2018, 10, 59. [Google Scholar] [CrossRef] [Green Version]
  11. Shih, Y.-C.T.; Sabik, L.M.; Stout, N.K.; Halpern, M.T.; Lipscomb, J.; Ramsey, S.; Ritzwoller, D.P. Health economics research in cancer screening: Research opportunities, challenges, and future directions. JNCI Monogr. 2022, 59, 42–50. [Google Scholar] [CrossRef]
  12. Hasan, M.R.; Ahommed, M.S.; Daizy, M.; Bacchu, M.S.; Ali, M.R.; Al-Mamun, M.R.; Aly, M.A.S.; Khan, M.Z.H.; Hossain, S.I. Recent development in electrochemical biosensors for cancer biomarkers detection. Biosens. Bioelectron. X 2021, 8, 100075. [Google Scholar] [CrossRef]
  13. Chen, Y.; Sun, L.; Qiao, X.; Zhang, Y.; Li, Y.; Ma, F. Signal-off/on electrogenerated chemiluminescence deoxyribosensors for assay of early lung cancer biomarker (NAP2) based on target-caused DNA charge transfer. Anal. Chim. Acta 2020, 1103, 67–74. [Google Scholar] [CrossRef]
  14. Pacheco, J.G.; Silva, M.S.V.; Freitas, M.; Nouws, H.P.A.; Delerue-Matos, C. Molecularly imprinted electrochemical sensor for the point-of-care detection of a breast cancer biomarker (CA 15-3). Sens. Actuators B 2018, 256, 905–912. [Google Scholar] [CrossRef] [Green Version]
  15. Pothipor, C.; Jakmunee, J.; Bamrungsap, S.; Ounnunka, K. An electrochemical biosensor for simultaneous detection of breast cancer clinically related microRNAs based on a gold nanoparticles/graphene quantum dots/graphene oxide film. Analyst 2021, 146, 4000–4009. [Google Scholar] [CrossRef]
  16. Li, S.; Hu, C.; Chen, C.; Zhang, J.; Bai, Y.; Tan, C.S.; Ni, G.; He, F.; Li, W.; Ming, D. Molybdenum disulfide supported on metal–organic frameworks as an ultrasensitive layer for the electrochemical detection of the ovarian cancer biomarker CA125. ACS Appl. Bio Mater. 2021, 4, 5494–5502. [Google Scholar] [CrossRef]
  17. Dou, Y.; Zhenhua, L.; Su, J.; Song, S. A portable biosensor based on Au nanoflower interface combined with electrochemical immunochromatography for POC detection of prostate-specific antigen. Biosensors 2022, 12, 259. [Google Scholar] [CrossRef]
  18. Chen, S.-C.; Chen, K.-T.; Jou, A.F.-J. Polydopamine-gold composite-based electrochemical biosensor using dual-amplification strategy for detecting pancreatic cancer-associated microRNA. Biosens. Bioelectron. 2021, 173, 112815. [Google Scholar] [CrossRef]
  19. Liu, Q.; Xie, H.; Liu, J.; Kong, J.; Zhang, X. A novel electrochemical biosensor for lung cancer-related gene detection based on copper ferrite-enhanced photoinitiated chain-growth amplification. Anal. Chim. Acta 2021, 1179, 338843. [Google Scholar] [CrossRef]
  20. Jing, L.; Xie, C.; Li, Q.; Yang, M.; Li, S.; Li, H.; Xia, F. Electrochemical biosensors for the analysis of breast cancer biomarkers: From design to application. Anal. Chem. 2021, 94, 269–296. [Google Scholar] [CrossRef]
  21. Kuralay, F.; Bayramlı, Y. Electrochemical determination of mitomycin C and its interaction with double-stranded DNA using a poly(o-phenylenediamine)-multi-walled carbon nanotube modified pencil graphite electrode. Anal. Lett. 2021, 54, 1295–1308. [Google Scholar] [CrossRef]
  22. Li, G.; Qi, X.; Wu, J.; Xu, L.; Wan, X.; Liu, Y.; Chen, Y.; Li, Q. Ultrasensitive, label-free voltammetric determination of norfloxacin based on molecularly imprinted polymers and Au nanoparticle-functionalized black phosphorus nanosheet nanocomposite. J. Hazard. Mater. 2022, 436, 129107. [Google Scholar] [CrossRef]
  23. Hai, X.; Li, Y.; Zhu, C.; Song, W.; Cao, J.; Bi, S. DNA-based label-free electrochemical biosensors: From principles to applications. TrAC Trends Anal. Chem. 2020, 133, 116098. [Google Scholar] [CrossRef]
  24. Reta, N.; Saint, C.P.; Michelmore, A.; Prieto-Simon, B.; Voelcker, N.H. Nanostructured electrochemical biosensors for label-free detection of water-and food-borne pathogens. ACS Appl. Mater. Interfaces 2018, 10, 6055–6072. [Google Scholar] [CrossRef] [PubMed]
  25. Jadon, N.; Jain, R.; Sharma, S.; Singh, K. Recent trends in electrochemical sensors for multianalyte detection—A review. Talanta 2016, 161, 894–916. [Google Scholar] [CrossRef] [PubMed]
  26. Yahaya, M.L.; Noordin, R.; Razak, K.A. Advanced nanoparticle-based biosensors for diagnosing foodborne pathogens. In Advanced Biosensors for Health Care Applications; Inamuddin Mohammad, A., Khan, R., Asiri, A.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2019; Chapter 1; pp. 1–43. [Google Scholar]
  27. Sharma, H.; Mutharasan, R. Review of biosensors for foodborne pathogens and toxins. Sens. Actuators B 2013, 183, 535–549. [Google Scholar] [CrossRef]
  28. Dükar, N.; Tunç, S.; Öztürk, K.; Demirci, S.; Dumangöz, M.; Sönmez Çelebi, M.; Kuralay, F. Highly sensitive and selective dopamine sensing in biological fluids with one-pot prepared graphene/poly(o-phenylenediamine) modified electrodes. Mater. Chem. Phys. 2019, 228, 357–362. [Google Scholar] [CrossRef]
  29. Hassan, R.Y.A. Advances in electrochemical nano-biosensors for biomedical and environmental applications: From current work to future perspectives. Sensors 2022, 22, 7539. [Google Scholar] [CrossRef]
  30. Sanko, V.; Şenocak, A.; Oğuz Tümay, S.; Orooji, Y.; Demirbas, E. An electrochemical sensor for detection of trace-level endocrine disruptor bisphenol A using Mo2Ti2AlC3 MAX phase/MWCNT composite modified electrode. Environ. Res. 2022, 212, 113071. [Google Scholar] [CrossRef]
  31. Patel, B.A. Electrochemistry for Bioanalysis; Elsevier: Amsterdam, The Netherlands, 2021. [Google Scholar]
  32. Laborda, E.; González, J.; Molina, Á. Recent advances on the theory of pulse techniques: A mini review. Electrochem. Commun. 2014, 43, 25–30. [Google Scholar] [CrossRef]
  33. Rezaei, B.; Irannejad, N. Electrochemical detection techniques in biosensor applications. In Electrochemical Biosensors; Ensafi, A.A., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; Chapter 2; pp. 11–43. [Google Scholar]
  34. Forouzanfar, S.; Alam, F.; Pala, N.; Wang, C. A review of electrochemical aptasensors for label-free cancer diagnosis. J. Electrochem. Soc. 2020, 167, 067511. [Google Scholar] [CrossRef]
  35. Özbek, O.; Berkel, C.; Isildak, O.; Isildak, I. Potentiometric urea biosensors. Clin. Chim. Acta 2022, 524, 154–163. [Google Scholar] [CrossRef]
  36. Sharifi, M.; Avadi, M.R.; Attar, F.; Dashtestani, F.; Ghorchian, H.; Rezayat, S.M.; Saboury, A.A.; Falahati, M. Cancer diagnosis using nanomaterials based electrochemical nanobiosensors. Biosens. Bioelectron. 2019, 126, 773–784. [Google Scholar] [CrossRef]
  37. Chillawar, R.R.; Tadi, K.K.; Motghare, R.V. Voltammetric techniques at chemically modified electrodes. J. Anal. Chem. 2015, 70, 399–418. [Google Scholar] [CrossRef]
  38. Kuralay, F.; Tunç, S.; Bozduman, F.; Oksuz, L.; Uygun Oksuz, A. Biosensing applications of titanium dioxide coated graphene modified disposable electrodes. Talanta 2016, 160, 325–331. [Google Scholar] [CrossRef]
  39. Pourali, A.; Rashidi, M.R.; Barar, J.; Pavon-Djavid, G.; Omidi, Y. Voltammetric biosensors for analytical detection of cardiac troponin biomarkers in acute myocardial infarction. TrAC Trends Anal. Chem. 2021, 134, 116123. [Google Scholar] [CrossRef]
  40. Walker, N.L.; Dick, J.E. Oxidase-loaded hydrogels for versatile potentiometric metabolite sensing. Biosens. Bioelectron. 2021, 178, 112997. [Google Scholar] [CrossRef]
  41. Sciuto, E.L.; Petralia, S.; van der Meer, J.; Conoci, S. Miniaturized electrochemical biosensor based on whole-cell for heavy metal ions detection in water. Biotechnol. Bioeng. 2021, 118, 1456–1465. [Google Scholar] [CrossRef]
  42. Hussein, H.A.; Kandeil, A.; Gomaa, M.; El Nashar, R.M.; El-Sherbiny, I.M.; Hassan, R.Y.A. SARS-CoV-2-impedimetric biosensor: Virus-imprinted chips for early and rapid diagnosis. ACS Sens. 2021, 6, 4098–4107. [Google Scholar] [CrossRef]
  43. Sanko, V.; Şenocak, A.; Oğuz Tümay, S.; Çamurcu, T.; Demirbas, E. Core-shell hierarchical enzymatic biosensor based on hyaluronic acid capped copper ferrite nanoparticles for determination of endocrine-disrupting bisphenol A. Electroanalysis 2022, 34, 561–572. [Google Scholar] [CrossRef]
  44. Yan, Y.; Qiao, Z.; Hai, X.; Song, W.; Bi, S. Versatile electrochemical biosensor based on bi-enzyme cascade biocatalysis spatially regulated by DNA architecture. Biosens. Bioelectron. 2021, 174, 112827. [Google Scholar] [CrossRef] [PubMed]
  45. Mahshid, S.S.; Flynn, S.E.; Mahshid, S. The potential application of electrochemical biosensors in the COVID-19 pandemic: A perspective on the rapid diagnostics of SARS-CoV-2. Biosens. Bioelectron. 2021, 176, 112905. [Google Scholar] [CrossRef] [PubMed]
  46. Vestergaard, M.; Kerman, K.; Tamiya, E. An overview of label-free electrochemical protein sensors. Sensors 2007, 7, 3442–3458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Grieshaber, D.; MacKenzie, R.; Vörös, J.; Reimhult, E. Electrochemical biosensors-sensor principles and architectures. Sensors 2008, 8, 1400–1458. [Google Scholar] [CrossRef]
  48. Ozcelikay, G.; Kurbanoglu, S.; Yarman, A.; Scheller, F.W.; Ozkan, S.A. Au-Pt nanoparticles based molecularly imprinted nanosensor for electrochemical detection of the lipopeptide antibiotic drug Daptomycin. Sens. Actuators B 2020, 320, 128285. [Google Scholar] [CrossRef]
  49. Colombo, R.N.P.; Sedenho, G.C.; Crespilho, F.N. Challenges in biomaterials science for electrochemical biosensing and bioenergy. Chem. Mater. 2022, 34, 10211–10222. [Google Scholar] [CrossRef]
  50. George, S.M.; Tandon, S.; Kandasubramanian, B. Advancements in hydrogel-functionalized immunosensing platforms. ACS Omega 2020, 5, 2060–2068. [Google Scholar] [CrossRef]
  51. Bhalla, N.; Pan, Y.; Yang, Z.; Payam, A.F. Opportunities and challenges for biosensors and nanoscale analytical tools for pandemics: COVID-19. ACS Nano 2020, 14, 7783–7807. [Google Scholar] [CrossRef]
  52. Wandelt, K. Encyclopedia of Interfacial Chemistry: Surface Science and Electrochemistry; Elsevier: Amsterdam, The Netherlands, 2018. [Google Scholar]
  53. Sedenho, G.C.; Hassan, A.; de Souza, J.C.P.; Crespilho, F.N. In situ and operando electrochemistry of redox enzymes. Curr. Opin. Electrochem. 2022, 34, 101015. [Google Scholar] [CrossRef]
  54. Sang, S.; Wang, Y.; Feng, Q.; Wei, Y.; Ji, J.; Zhang, W. Progress of new label-free techniques for biosensors: A review. Crit. Rev. Biotechnol. 2016, 36, 465–481. [Google Scholar] [CrossRef]
  55. Lee, S.-L.; Kim, J.; Choi, S.; Han, J.; Seo, G.; Lee, Y.W. Fiber-optic label-free biosensor for SARS-CoV-2 spike protein detection using biofunctionalized long-period fiber grating. Talanta 2021, 235, 122801. [Google Scholar] [CrossRef]
  56. Yen, Y.-K.; Chiu, C.-Y. A CMOS MEMS-based membrane-bridge nanomechanical sensor for small molecule detection. Sci. Rep. 2020, 10, 2931. [Google Scholar] [CrossRef] [Green Version]
  57. Faria, H.A.M.; Zucolotto, V. Label-free electrochemical DNA biosensor for zika virus identification. Biosens. Bioelectron. 2019, 131, 149–155. [Google Scholar] [CrossRef]
  58. Syahir, A.; Usui, K.; Tomizaki, K.; Kajikawa, K.; Mihara, H. Label and label-free detection techniques for protein microarrays. Microarrays 2015, 4, 228–244. [Google Scholar] [CrossRef] [Green Version]
  59. Wang, H.; Huang, Y.; Xiog, M.; Wang, F.; Li, C. A label-free electrochemical biosensor for highly sensitive detection of gliotoxin based on DNA nanostructure/MXene nanocomplexes. Biosens. Bioelectron. 2019, 142, 111531. [Google Scholar] [CrossRef]
  60. Han, S.; Liu, W.; Yang, S.; Wang, R. Facile and label-free electrochemical biosensors for microRNA detection based on DNA origami nanostructures. ACS Omega 2019, 4, 11025–11031. [Google Scholar] [CrossRef] [Green Version]
  61. Vu, Q.K.; Tran, Q.Y.; Vu, N.P.; Anh, T.-L.; Le Dnag, T.T.; Tonezzer, M.; Nguyen, T.H.H. A label-free electrochemical biosensor based on screen-printed electrodes modified with gold nanoparticles for quick detection of bacterial pathogens. Mater. Today Commun. 2021, 26, 101726. [Google Scholar] [CrossRef]
  62. Singh, A.K.; Dhiman, T.K.; Lakshmi, G.B.V.S.; Solanki, P.R. Dimanganese trioxide (Mn2O3) based label-free electrochemical biosensor for detection of Aflatoxin-B1. Bioelectrochemistry 2021, 137, 107684. [Google Scholar] [CrossRef]
  63. Karunakaran, C.; Rajkumar, R.; Bhargava, K. Introduction to biosensors. In Biosensors and Bioelectronics, 1st ed.; Karunakaran, C., Bhargava, K., Benjamin, R., Eds.; Elsevier: Amsterdam, The Netherlands, 2015; Chapter 1; pp. 1–68. [Google Scholar]
  64. Du, Y.; Dong, S. Nucleic acid biosensors: Recent advances and perspectives. Anal. Chem. 2017, 89, 189–215. [Google Scholar] [CrossRef]
  65. Kuralay, F.; Erdem, A. Gold nanoparticle/polymer nanocomposite for highly sensitive drug–DNA interaction. Analyst 2015, 140, 2876–2880. [Google Scholar]
  66. Fu, Z.; Lu, Y.-C.; Lai, J.J. Recent advances in biosensors for nucleic acid and exosome detection. Chonnam Med. J. 2019, 55, 86–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Mujica, M.L.; Rubianes, M.D.; Rivas, G. A multipurpose biocapture nanoplatform based on multiwalled-carbon nanotubes non-covalently functionalized with avidin: Analytical applications for the non-amplified and label-free impedimetric quantification of BRCA1. Sens. Actuators B 2022, 357, 131304. [Google Scholar] [CrossRef]
  68. Kong, L.; Lv, S.; Qiao, Z.; Yan, Y.; Zhang, J.; Bi, S. Metal-organic framework nanoreactor-based electrochemical biosensor coupled with three-dimensional DNA walker for label-free detection of microRNA. Biosens. Bioelectron. 2022, 207, 114188. [Google Scholar] [CrossRef] [PubMed]
  69. Gao, J.; Wang, C.; Chu, Y.; Wang, S.; Sun, M.Y.; Ji, H.; Gao, Y.; Wang, Y.; Han, Y.; Song, F.; et al. Poly-L-lysine-modified graphene field-effect transistor biosensors for ultrasensitive breast cancer miRNAs and SARS-CoV-2 RNA detection. Anal. Chem. 2022, 94, 1626–1636. [Google Scholar] [CrossRef]
  70. Khodadoust, A.; Nasirizadeh, N.; Seyfati, S.M.; Taheri, R.A.; Ghanei, M.; Bagheri, H. High-performance strategy for the construction of electrochemical biosensor for simultaneous detection of miRNA-141 and miRNA-21 as lung cancer biomarkers. Talanta 2023, 252, 123863. [Google Scholar] [CrossRef]
  71. Meng, F.; Yu, W.; Chen, C.; Guo, S.; Tian, X.; Miao, Y.; Ma, L.; Zhang, X.; Yu, Y.; Huang, L.; et al. A versatile electrochemical biosensor for the detection of circulating microRNA toward non-small cell lung cancer diagnosis. Small 2022, 18, 2200784. [Google Scholar] [CrossRef]
  72. Zhao, J.; He, C.; Wu, W.; Yang, H.; Dong, J.; Wen, L.; Hu, Z.; Yang, M.; Hou, C.; Huo, D. MXene-MoS2 heterostructure collaborated with catalyzed hairpin assembly for label-free electrochemical detection of microRNA-21. Talanta 2022, 237, 122927. [Google Scholar] [CrossRef]
  73. Pothipor, C.; Bamrungsap, S.; Jakmunee, J.; Ounnunkad, K. A gold nanoparticle-dye/poly (3-aminobenzylamine)/two dimensional MoSe2/graphene oxide electrode towards label-free electrochemical biosensor for simultaneous dual-mode detection of cancer antigen 15-3 and microRNA-21. Colloids Surf. B 2022, 210, 112260. [Google Scholar] [CrossRef]
  74. Jafari-Kashi, A.; Rafiee-Pour, H.-A.; Shabani-Nooshabadi, M. A new strategy to design label-free electrochemical biosensor for ultrasensitive diagnosis of CYFRA 21–1 as a biomarker for detection of non-small cell lung cancer. Chemosphere 2022, 301, 134636. [Google Scholar] [CrossRef]
  75. Avelino, K.Y.P.S.; Oliveira, L.S.; Santos, M.R.; Lucena-Silva, N.; Andrade, C.A.S.; Oliveira, M.D.L. Electrochemical DNA biosensor for chronic myelocytic leukemia based on hybrid nanostructure. Bioelectrochemistry 2022, 147, 108176. [Google Scholar] [CrossRef]
  76. Song, S.; Wang, L.; Li, J.; Fan, C.; Zhao, J. Aptamer-based biosensors. TrAC Trends Anal. Chem. 2008, 27, 108–117. [Google Scholar] [CrossRef]
  77. Ni, Y.; Ouyang, H.; Yu, L.; Ling, C.; Zhu, Z.; He, A.; Liu, R. Label-free electrochemical aptasensor based on magnetic α-Fe2O3/Fe3O4 heterogeneous hollow nanorods for the detection of cancer antigen 125. Bioelectrochemistry 2022, 148, 108255. [Google Scholar] [CrossRef]
  78. Sawant, S.N. Development of biosensors from biopolymer composites. In Biopolymer Composites in Electronics, 1st ed.; Sadasivuni, K.K., Kim, J., AlMaadeed, M.A., Ponnamma, D., Cabibihan, J.-J., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; Chapter 13; pp. 353–383. [Google Scholar]
  79. Zhou, X.; Pu, Q.; Yu, H.; Peng, Y.; Li, J.; Yang, Y.; Chen, H.; Weng, Y.; Xie, G. An electrochemical biosensor based on hemin/G-quadruplex DNAzyme and PdRu/Pt heterostructures as signal amplifier for circulating tumor cells detection. J. Colloid Interface Sci. 2021, 599, 752–761. [Google Scholar] [CrossRef]
  80. Nabok, A.; Abu-Ali, H.; Takita, S.; Smith, D.P. Electrochemical detection of prostate cancer biomarker PCA3 using specific RNA-based aptamer labelled with ferrocene. Chemosensors 2021, 9, 59. [Google Scholar] [CrossRef]
  81. Wei, X.; Wang, S.; Zhan, Y.; Kai, T.; Ding, P. Sensitive identification of microcystin-LR via a reagent-free and reusable electrochemical biosensor using a methylene blue-labeled aptamer. Biosensors 2022, 12, 556. [Google Scholar] [CrossRef]
  82. Zhang, F.; Fan, L.; Liu, Z.; Han, Y.; Guo, Y. A label-free electrochemical aptasensor for the detection of cancer antigen 125 based on nickel hexacyanoferrate nanocubes/polydopamine functionalized graphene. J. Electroanal. Chem. 2022, 918, 116424. [Google Scholar] [CrossRef]
  83. Sadrabadi, E.A.; Benvidi, A.; Yazdanparast, S.; Amiri-zirtol, L. Fabrication of a label-free electrochemical aptasensor to detect cytochrome c in the early stage of cell apoptosis. Microchim. Acta 2022, 189, 279. [Google Scholar] [CrossRef]
  84. Wang, Y.; Sun, S.; Luo, J.; Xiong, Y.; Ming, T.; Liu, J.; Ma, Y.; Yan, S.; Yang, Y.; Yang, Z.; et al. Low sample volume origami-paper-based graphene-modified aptasensors for label-free electrochemical detection of cancer biomarker-EGFR. Microsyst. Nanoeng. 2020, 6, 32. [Google Scholar] [CrossRef]
  85. Holford, T.R.J.; Davis, F.; Higson, S.P.J. Recent trends in antibody based sensors. Biosens. Bioelectron. 2012, 34, 12–24. [Google Scholar] [CrossRef]
  86. Liu, X.; Liu, J. Biosensors and sensors for dopamine detection. View 2021, 2, 20200102. [Google Scholar] [CrossRef]
  87. Dover, J.E.; Hwang, G.M.; Mullen, E.H.; Prorok, B.C.; Suh, S.-J. Recent advances in peptide probe-based biosensors for detection of infectious agents. J. Microbiol. Methods 2009, 78, 10–19. [Google Scholar] [CrossRef] [PubMed]
  88. Wei, C.; Xiao, J.; Liu, S.; Wang, Z.; Chen, L.; Teng, W. Simple and label-free electrochemical immuno determination of the gastric cancer biomarker carbohydrate antigen 72-4 with a carbon nanotube-graphene oxide hybrid as the sensing platform and ferrocyanide/ferricyanide as the probe. Anal. Lett. 2022, 55, 1306–1317. [Google Scholar] [CrossRef]
  89. Aydın, E.B.; Aydın, M.; Sezgintürk, M.K. Impedimetric detection of calreticulin by a disposable immunosensor modified with a single-walled carbon nanotube-conducting polymer nanocomposite. ACS Biomater. Sci. Eng. 2022, 8, 3773–3784. [Google Scholar] [CrossRef] [PubMed]
  90. Shawky, A.M.; El-Tohamy, M. Signal amplification strategy of label-free ultrasenstive electrochemical immunosensor based ternary Ag/TiO2/rGO nanocomposites for detecting breast cancer biomarker CA 15-3. Mater. Chem. Phys. 2021, 272, 124983. [Google Scholar] [CrossRef]
  91. Ortega, F.G.; Regiart, M.D.; Rodríguez-Martínez, A.; de Miguel-Pérez, D.; Serrano, M.J.; Lorente, J.A.; Tortella, G.; Rubilar, O.; Sapag, K.; Bertotti, M.; et al. Sandwich-type electrochemical paper-based immunosensor for claudin 7 and CD81 dual determination on extracellular vesicles from breast cancer patients. Anal. Chem. 2020, 93, 1143–1153. [Google Scholar] [CrossRef]
  92. Kuntamung, K.; Jakmunee, J.; Ounnunkad, K. A label-free multiplex electrochemical biosensor for the detection of three breast cancer biomarker proteins employing dye/metal ion-loaded and antibody-conjugated polyethyleneimine-gold nanoparticles. J. Mat. Chem. B 2021, 9, 6576–6585. [Google Scholar] [CrossRef]
  93. Chen, Z.; Li, B.; Liu, J.; Li, H.; Li, C.; Xuan, X.; Li, M. A label-free electrochemical immunosensor based on a gold–vertical graphene/TiO2 nanotube electrode for CA125 detection in oxidation/reduction dual channels. Microchim. Acta 2022, 189, 257. [Google Scholar] [CrossRef]
  94. Öndeş, B.; Evli, S.; Uygun, M.; Uygun, D.A. Boron nitride nanosheet modified label-free electrochemical immunosensor for cancer antigen 125 detection. Biosens. Bioelectron. 2021, 191, 113454. [Google Scholar] [CrossRef]
  95. Biswas, S.; Lan, Q.; Xie, Y.; Sun, X.; Wang, Y. Label-free electrochemical immunosensor for ultrasensitive detection of carbohydrate antigen 125 based on antibody-immobilized biocompatible MOF-808/CNT. ACS Appl. Mater. Interfaces 2021, 13, 3295–3302. [Google Scholar] [CrossRef]
  96. Rafique, S.; Tabassum, S.; Akram, R. Sensitive competitive label-free electrochemical immunosensor for primal detection of ovarian cancer. Chem. Pap. 2020, 74, 2591–2603. [Google Scholar] [CrossRef]
  97. Rong, S.; Zou, L.; Zhu, L.; Zhang, Z.; Liu, H.; Zhang, Y.; Zhang, H.; Gao, H.; Guan, H.; Dong, J.; et al. 2D/3D material amplification strategy for disposable label-free electrochemical immunosensor based on rGO-TEPA@ Cu-MOFs@ SiO2@AgNPs composites for NMP22 detection. Microchem. J. 2021, 168, 106410. [Google Scholar] [CrossRef]
  98. Echeverri, D.; Orozco, J. β-1, 4-Galactosyltransferase-V colorectal cancer biomarker immunosensor with label-free electrochemical detection. Talanta 2022, 243, 123337. [Google Scholar] [CrossRef]
  99. Kuntamung, K.; Sangthong, P.; Jakmunee, J.; Ounnunkad, K. A label-free immunosensor for the detection of a new lung cancer biomarker, GM2 activator protein, using a phosphomolybdic acid/polyethyleneimine coated gold nanoparticle composite. Analyst 2021, 146, 2203–2211. [Google Scholar] [CrossRef]
  100. Kabay, G.; Yin, Y.; Singh, C.K.; Ahmad, N.; Gunasekaran, S.; Mutlu, M. Disposable electrochemical immunosensor for prostate cancer detection. Sens. Actuators B 2022, 360, 131667. [Google Scholar] [CrossRef]
  101. Martínez-Rojas, F.; Castañeda, E.; Armijo, F. Conducting polymer applied in a label-free electrochemical immunosensor for the detection prostate-specific antigen using its redox response as an analytical signal. J. Electroanal. Chem. 2021, 880, 114877. [Google Scholar] [CrossRef]
  102. Gui, J.-C.; Han, L.; Du, C.-X.; Yu, X.-N.; Hu, K.; Li, L.-H. An efficient label-free immunosensor based on ce-MoS2/AgNR composites and screen-printed electrodes for PSA detection. J. Solid State Electrochem. 2021, 25, 973–982. [Google Scholar] [CrossRef]
  103. Choosang, J.; Khumngern, S.; Thavarungkul, P.; Kanatharan, P.; Numnuam, A. An ultrasensitive label-free electrochemical immunosensor based on 3D porous chitosan–graphene–ionic liquid–ferrocene nanocomposite cryogel decorated with gold nanoparticles for prostate-specific antigen. Talanta 2021, 224, 121787. [Google Scholar] [CrossRef]
  104. Chen, S.; Xu, L.; Sheng, K.; Zhou, Q.; Dong, B.; Bai, X.; Lu, G.; Song, H. A label-free electrochemical immunosensor based on facet-controlled Au nanorods/reduced graphene oxide composites for prostate specific antigen detection. Sens. Actuators B 2021, 336, 129748. [Google Scholar] [CrossRef]
  105. Mishra, S.; Kim, E.-S.; Sharma, P.K.; Wang, Z.-J.; Yang, S.-Y.; Kaushik, A.K.; Wang, C.; Li, Y.; Kim, N.-Y. Tailored biofunctionalized biosensor for the label-free sensing of prostate-specific antigen. ACS Appl. Bio Mater. 2020, 3, 7821–7830. [Google Scholar] [CrossRef]
  106. Zhu, X.; Dai, Y.; Sun, Y.; Liu, H.; Sun, W.; Lin, Y.; Gao, D.; Han, R. Rapid fabrication of electrode for the detection of alpha fetoprotein based on MnO2 functionalized mesoporous carbon hollow sphere. Mater. Sci. Eng. C 2020, 107, 110206. [Google Scholar] [CrossRef]
  107. Yan, L.; Zhang, C.; Xi, F. Disposable amperometric label-free immunosensor on chitosan–graphene-modified patterned ITO electrodes for prostate specific antigen. Molecules 2022, 27, 5895. [Google Scholar] [CrossRef] [PubMed]
  108. Wu, M.; Liu, S.; Qi, F.; Qiu, R.; Feng, J.; Ren, X.; Rong, S.; Ma, H.; Chang, D.; Pan, H. A label-free electrochemical immunosensor for CA125 detection based on CMK-3 (Au/Fc@MgAl-LDH)n multilayer nanocomposites modification. Talanta 2022, 241, 123254. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, Y.; Wang, X.; Fang, X.; Yuan, X.; Yang, H.; Kong, J. Label-free electrochemical immunoassay for detecting CYFRA 21-1 using poly (ε-caprolactone)-b-poly(ethylene oxide) block copolymer. Microchem. J. 2021, 165, 106119. [Google Scholar] [CrossRef]
  110. Liu, X.; Lin, L.-Y.; Tseng, F.Y.; Tan, Y.-C.; Li, J.; Feng, L.; Song, L.; Lai, C.-F.; Li, X.; He, J.-H.; et al. Label-free electrochemical immunosensor based on gold nanoparticle/polyethyleneimine/reduced graphene oxide nanocomposites for the ultrasensitive detection of cancer biomarker matrix metalloproteinase-1. Analyst 2021, 146, 4066–4079. [Google Scholar] [CrossRef] [PubMed]
  111. Liu, Q.; Wu, C.; Cai, H.; Hu, N.; Zhou, J.; Wang, P. Cell-based biosensors and their application in biomedicine. Chem. Rev. 2014, 114, 6423–6461. [Google Scholar] [CrossRef] [PubMed]
  112. Gupta, N.; Wu, C.; Cai, H.; Hu, N.; Zhou, J.; Wang, P. Cell-based biosensors: Recent trends, challenges and future perspectives. Biosens. Bioelectron. 2019, 141, 111435. [Google Scholar] [CrossRef]
  113. Lu, X.; Ye, Y.; Zhang, Y.; Sun, X. Current research progress of mammalian cell-based biosensors on the detection of foodborne pathogens and toxins. Crit. Rev. Food Sci. Nutr. 2021, 61, 3819–3835. [Google Scholar] [CrossRef]
  114. Zhou, H.; Huang, R.; Su, T.; Li, B.; Zhou, H.; Ren, J.; Li, Z. A c-MWCNTs/AuNPs-based electrochemical cytosensor to evaluate the anticancer activity of pinoresinol from Cinnamomum camphora against HeLa cells. Bioelectrochemistry 2022, 146, 108133. [Google Scholar] [CrossRef]
  115. Li, C.; Cui, Y.; Ren, J.; Zou, J.; Kuang, W.; Sun, X.; Hu, X.; Yan, Y.; Ling, X. Novel cells-based electrochemical sensor for investigating the interactions of cancer cells with molecules and screening multitarget anticancer drugs. Anal. Chem. 2020, 93, 1480–1488. [Google Scholar] [CrossRef]
  116. Liu, F.-F.; Zhao, X.-P.; Liao, X.-W.; Liu, W.-Y.; Chen, Y.-M.; Wang, C. Ultrasensitive and label-free detection of cell surface glycan using nanochannel-ionchannel hybrid coupled with electrochemical detector. Anal. Chem. 2020, 92, 5509–5516. [Google Scholar] [CrossRef]
  117. Ding, L.; Du, D.; Zhang, X.; Ju, H. Trends in cell-based electrochemical biosensors. Curr. Med. Chem. 2008, 15, 3160–3170. [Google Scholar] [CrossRef]
  118. Sandhyarani, N. Surface modification methods for electrochemical biosensors. In Electrochemical Biosensors, 1st ed.; Ensafi, A.A., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; Chapter 3; pp. 45–75. [Google Scholar]
  119. Cruz-Pacheco, A.F.; Quinchia, J.; Orozco, J. Cerium oxide–doped PEDOT nanocomposite for label-free electrochemical immunosensing of anti-p53 autoantibodies. Microchim. Acta 2022, 189, 228. [Google Scholar] [CrossRef]
  120. Ghanavati, M.; Tadayon, F.; Bagheri, H. A novel label-free impedimetric immunosensor for sensitive detection of prostate specific antigen using Au nanoparticles/MWCNTs-graphene quantum dots nanocomposite. Microchem. J. 2020, 159, 105301. [Google Scholar] [CrossRef]
  121. Luo, X.; Davis, J.J. Electrical biosensors and the label free detection of protein disease biomarkers. Chem. Soc. Rev. 2013, 42, 5944–5962. [Google Scholar] [CrossRef]
  122. Burcu Aydın, E.; Aydın, M.; Sezgintürk, M.K. Biosensors and the evaluation of food contaminant biosensors in terms of their performance criteria. Int. J. Environ. Anal. Chem. 2020, 100, 602–622. [Google Scholar] [CrossRef]
  123. Agrahari, S.; Gautam, R.K.; Singh, A.K.; Tiwari, I. Nanoscale materials-based hybrid frameworks modified electrochemical biosensors for early cancer diagnostics: An overview of current trends and challenges. Microchem. J. 2022, 172, 106980. [Google Scholar] [CrossRef]
  124. Yaiwong, P.; Semakul, N.; Bamrungsap, S.; Jakmunee, J.; Ounnunkad, K. Electrochemical detection of matrix metalloproteinase-7 using an immunoassay on a methylene blue/2D MoS2/graphene oxide electrode. Bioelectrochemistry 2021, 142, 107944. [Google Scholar] [CrossRef]
  125. Hartmann, M.; Kostrov, X. Immobilization of enzymes on porous silicas—Benefits and challenges. Chem. Soc. Rev. 2013, 42, 6277–6289. [Google Scholar] [CrossRef]
  126. Eş, I.; Vieira, J.D.G.; Amaral, A.C. Principles, techniques, and applications of biocatalyst immobilization for industrial application. Appl. Microbiol. Biotechnol. 2015, 99, 2065–2082. [Google Scholar] [CrossRef]
  127. Pachauri, N.; Lakshmi, G.B.V.S.; Sri, S.; Gupta, P.K.; Solanki, P.R. Silver molybdate nanoparticles based immunosensor for the non-invasive detection of Interleukin-8 biomarker. Mater. Sci. Eng. C 2020, 113, 110911. [Google Scholar] [CrossRef]
  128. Ehzari, H.; Samimi, M.; Safari, M.; Gholivand, M.B. Label-free electrochemical immunosensor for sensitive HER2 biomarker detection using the core-shell magnetic metal-organic frameworks. J. Electroanal. Chem. 2020, 877, 114722. [Google Scholar] [CrossRef]
  129. Butmee, P.; Tumcharern, G.; Thouand, G.; Kalcherd, K. An ultrasensitive immunosensor based on manganese dioxide-graphene nanoplatelets and core shell Fe3O4@Au nanoparticles for label-free detection of carcinoembryonic antigen. Bioelectrochemistry 2020, 132, 107452. [Google Scholar] [CrossRef] [PubMed]
  130. Zhang, D.; Wang, Y.; Jin, X.; Xiao, Q.; Huang, S. A label-free and ultrasensitive electrochemical biosensor for oral cancer overexpressed 1 gene via exonuclease III-assisted target recycling and dual enzyme-assisted signal amplification strategies. Analyst 2022, 147, 2412–2424. [Google Scholar] [CrossRef] [PubMed]
  131. Wang, C.; Liu, Y.; Chen, R.; Wang, X.; Wang, Y.; Wei, J.; Zhang, K.; Zhang, C. Electrochemical biosensing of circulating microRNA-21 in cerebrospinal fluid of medulloblastoma patients through target-induced redox signal amplification. Microchim. Acta 2022, 189, 105. [Google Scholar] [CrossRef]
  132. Zhai, X.-J.; Wang, Q.-L.; Cui, H.-F.; Song, X.; Lv, Q.-Y.; Guo, Y. A DNAzyme-catalyzed label-free aptasensor based on multifunctional dendrimer-like DNA assembly for sensitive detection of carcinoembryonic antigen. Biosens. Bioelectron. 2021, 194, 113618. [Google Scholar] [CrossRef]
  133. Wang, Y.; Chen, L.; Xuan, T.; Wang, J.; Wang, X. Label-free electrochemical impedance spectroscopy aptasensor for ultrasensitive detection of lung cancer biomarker carcinoembryonic antigen. Front. Chem. 2021, 9, 721008. [Google Scholar] [CrossRef]
  134. Shafiei, F.; Saberi, R.S.; Mehrgardi, M.A. A label-free electrochemical aptasensor for breast cancer cell detection based on a reduced graphene oxide-chitosan-gold nanoparticle composite. Bioelectrochemistry 2021, 140, 107807. [Google Scholar] [CrossRef]
  135. Zhou, J.; Cheng, K.; Chen, X.; Yang, R.; Lu, M.; Ming, L.; Chen, Y.; Lin, Z.; Chen, D. Determination of soluble CD44 in serum by using a label-free aptamer based electrochemical impedance biosensor. Analyst 2020, 145, 460–465. [Google Scholar] [CrossRef]
  136. Forouzanfar, S.; Alam, F.; Pala, N.; Wang, C. Highly sensitive label-free electrochemical aptasensors based on photoresist derived carbon for cancer biomarker detection. Biosens. Bioelectron. 2020, 170, 112598. [Google Scholar] [CrossRef]
  137. Khodadoust, A.; Nasirizadeh, N.; Taheri, R.A.; Dehghani, M.; Ghanei, M.; Bagheri, H. A ratiometric electrochemical DNA-biosensor for detection of miR-141. Microchim. Acta 2022, 189, 213. [Google Scholar] [CrossRef]
  138. Pothipor, C.; Aroonyadet, N.; Bamrungsap, S.; Jakmunee, J.; Ounnunkad, K. A highly sensitive electrochemical microRNA-21 biosensor based on intercalating methylene blue signal amplification and a highly dispersed gold nanoparticles/graphene/polypyrrole composite. Analyst 2021, 146, 2679–2688. [Google Scholar] [CrossRef]
  139. Pareek, S.; Jain, U.; Bharadwaj, M.; Chauhan, N. A label free nanosensing platform for the detection of cervical cancer through analysis of ultratrace DNA hybridization. Sens. Bio-Sens. Res. 2021, 33, 100444. [Google Scholar] [CrossRef]
  140. Moazampour, M.; Zare, H.R.; Shekari, Z. Femtomolar determination of an ovarian cancer biomarker (miR-200a) in blood plasma using a label free electrochemical biosensor based on L-cysteine functionalized ZnS quantum dots. Anal. Methods 2021, 13, 2021–2029. [Google Scholar] [CrossRef]
  141. Xia, Y.M.; Li, M.-Y.; Chen, C.-L.; Xia, M.; Zhang, W.; Gao, W.-W. Employing label-free electrochemical biosensor based on 3D-reduced graphene oxide and polyaniline nanofibers for ultrasensitive detection of breast cancer BRCA1 biomarker. Electroanalysis 2020, 32, 2045–2055. [Google Scholar] [CrossRef]
  142. Farshchi, F.; Saadati, A.; Fathi, N.; Hasanzadeh, M.; Samiei, M. Flexible paper-based label-free electrochemical biosensor for the monitoring of miRNA-21 using core–shell Ag@Au/GQD nano-ink: A new platform for the accurate and rapid analysis by low cost lab-on-paper technology. Anal. Methods 2021, 13, 286–1294. [Google Scholar] [CrossRef]
  143. Kaya, H.K.; Çağlayan, T.; Kuralay, F. Functionalized nanomaterial-based electrochemical sensors for point-of-care devices. In Functionalized Nanomaterial-Based Electrochemical Sensors Principles, Fabrication Methods, and Applications, 1st ed.; Hussain, C.M., Manjunatha, J.G., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; Chapter 14; pp. 309–335. [Google Scholar]
  144. Ebrahimi, G.; Samadi Pakchin, P.; Shamloo, A.; Mota, A.; de la Guardia, M.; Omidian, H.; Omid, Y. Label-free electrochemical microfluidic biosensors: Futuristic point-of-care analytical devices for monitoring diseases. Microchim. Acta 2021, 189, 252. [Google Scholar] [CrossRef]
  145. Surucu, O.; Öztürk, E.; Kuralay, F. Nucleic acid integrated technologies for electrochemical point-of-care diagnostics: A comprehensive review. Electroanalysis 2021, 34, 148–160. [Google Scholar] [CrossRef]
  146. Keyvani, F.; Debnath, N.; Ayman Saleh, M.; Poudineh, M. An integrated microfluidic electrochemical assay for cervical cancer detection at point-of-care testing. Nanoscale 2022, 14, 6761–6770. [Google Scholar] [CrossRef]
  147. Ming, T.; Cheng, Y.; Xing, Y.; Luo, J.; Mao, M.; Liu, J.; Sun, S.; Kong, F.; Jin, H.; Cai, X. Electrochemical microfluidic paper-based aptasensor platform based on a biotin–streptavidin system for label-Free detection of biomarkers. ACS Appl. Mater. Interfaces 2021, 13, 46317–46324. [Google Scholar] [CrossRef]
  148. Chakraborty, B.; Das, A.; Mandal, N.; Samanta, N.; Das, N.; Chaudkur, C.R. Label free, electric field mediated ultrasensitive electrochemical point-of-care device for CEA detection. Sci. Rep. 2021, 11, 2962. [Google Scholar] [CrossRef]
  149. Laocharoensuk, R. Development of electrochemical immunosensors towards point-of-care cancer diagnostics: Clinically relevant studies. Electroanalysis 2016, 28, 1716–1729. [Google Scholar] [CrossRef]
  150. Dai, Y.; Liu, C.C. Recent advances on electrochemical biosensing strategies toward universal point-of-care systems. Angew. Chem. Int. Ed. 2019, 131, 12483–12496. [Google Scholar] [CrossRef]
  151. Syedmoradi, L.; Norton, M.L.; Omidfar, K. Point-of-care cancer diagnostic devices: From academic research to clinical translation. Talanta 2021, 225, 122002. [Google Scholar] [CrossRef] [PubMed]
  152. Lopes, L.C.; Santos, A.; Bueno, P.R. An outlook on electrochemical approaches for molecular diagnostics assays and discussions on the limitations of miniaturized technologies for point-of-care devices. Sens. Actuators Rep. 2022, 4, 100087. [Google Scholar] [CrossRef]
Figure 1. Label-free electrochemical cancer biosensors: electrode modifications such as nanotechnology-based materials, biorecognition immobilization protocols and some of the powerful electrochemical detection techniques.
Figure 1. Label-free electrochemical cancer biosensors: electrode modifications such as nanotechnology-based materials, biorecognition immobilization protocols and some of the powerful electrochemical detection techniques.
Biosensors 13 00333 g001
Figure 2. Various immobilization methods for the biorecognition elements.
Figure 2. Various immobilization methods for the biorecognition elements.
Biosensors 13 00333 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sanko, V.; Kuralay, F. Label-Free Electrochemical Biosensor Platforms for Cancer Diagnosis: Recent Achievements and Challenges. Biosensors 2023, 13, 333. https://0-doi-org.brum.beds.ac.uk/10.3390/bios13030333

AMA Style

Sanko V, Kuralay F. Label-Free Electrochemical Biosensor Platforms for Cancer Diagnosis: Recent Achievements and Challenges. Biosensors. 2023; 13(3):333. https://0-doi-org.brum.beds.ac.uk/10.3390/bios13030333

Chicago/Turabian Style

Sanko, Vildan, and Filiz Kuralay. 2023. "Label-Free Electrochemical Biosensor Platforms for Cancer Diagnosis: Recent Achievements and Challenges" Biosensors 13, no. 3: 333. https://0-doi-org.brum.beds.ac.uk/10.3390/bios13030333

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop