Skoči na glavni sadržaj

Pregledni rad

https://doi.org/10.5599/admet.1709

Recent advances in electrochemical sensors and biosensors for monitoring drugs and metabolites in pharmaceutical and biological samples

Charaf Laghlimi
Abdelaziz Moutcine
Abdelilah Chtaini
Jalal Isaad
Adil Soufi
Younes Ziat
Hassan Amhamdi
Hamza Belkhanchi


Puni tekst: engleski pdf 1.101 Kb

str. 151-173

preuzimanja: 111

citiraj

Preuzmi JATS datoteku


Sažetak

Various applications of electrochemical sensors and biosensors have been reported in many fields. These include pharmaceuticals, drug detection, cancer detection, and analysis of toxic elements in tap water. Electrochemical sensors are characterised by their low cost, ease of manufacture, rapid analysis, small size and ability to detect multiple elements simultaneously. They also allow the reaction mechanisms of analytes, such as drugs, to be taken into account, giving a first indication of their fate in the body or their pharmaceutical preparation. Several materials are used in the construction of sensors, such as graphene, fullerene, carbon nanotubes, carbon graphite, glassy carbon, carbon clay, graphene oxide, reduced graphene oxide, and metals. This review covers the most recent progress in electrochemical sensors used to analyze drugs and metabolites in pharmaceutical and biological samples. We have highlighted carbon paste electrodes (CPE), glassy carbon electrodes (GCE), screen-printed carbon electrodes (SPCE) and reduced graphene oxide electrodes (rGOE). The sensitivity and analysis speed of electrochemical sensors can be improved by modifying them with conductive materials. Different materials used for modification have been reported and demonstrated, such as molecularly imprinted polymers, multiwalled carbon nanotubes, fullerene (C60), iron(III) nanoparticles (Fe3O4NP), and CuO micro-fragments (CuO MF). Manufacturing strategies and the detection limit of each sensor have been reported.

Ključne riječi

Pharmaceuticals and drugs; screen printed carbon electrodes; glassy carbon electrodes; carbon nanotubes

Hrčak ID:

304490

URI

https://hrcak.srce.hr/304490

Datum izdavanja:

4.6.2023.

Posjeta: 559 *




Introduction

Pharmacokinetics aims to determine a drug's pathway from administration to excretion [1]. For clinical diagnosis, there is a need for information on how medicines work in the human body and the dose that should be administered [2]. It is, therefore, necessary to quantify drugs in physiological fluids such as serum, urine, sweat and saliva, as well as in pharmaceutical tablets. Several sensitive and accurate methods have been used to quantify and detect different types of drugs, such as spectrophotometry [3,4], fluorimetry [3], RP-HPLC [4], HPLC-diode array detector, TLC-densitometric methods [5] and capillary zone electrophoresis [6]. However, these techniques are very expensive and require the use of expensive solvents, specialised technicians, long sample preparation times and expensive instrumentation, all of which increase the cost and duration of the analysis [2]. For this reason, the pharmacokinetic community strives to provide alternative methods of drug surveillance that are easy, inexpensive, sensitive and precise [7]. In the last decades, electrochemical sensors have become an interesting alternative technique due to their high capacity to analyse drugs in different physiological fluids [8-10]. Graphene has considerable sensitivity and selectivity and often has a large potential window to accommodate the redox process involved. It also has a significant electron transfer process due to its planar sites and the presence of σ and π bonds, making it an attractive material for sensor manufacture [11,12]. Modification of the carbon paste electrode offers high sensitivity, low cost and rapid analysis without any pre-treatment [13-21]. The modification is an effective way to reduce the overpotential and increase the sensitivity of the sensors [22]. Various electrodes based on carbon as a conductor have been used for the electroanalysis of drugs [23,25].

In this review, we report on a new advance in carbon graphite-based electrochemical sensors used to analyse different types of drugs in pharmaceutical tablets and a variety of physiological fluids such as serum, blood, sweat and urine. The different modifiers used to improve sensor performance and increase the electroactive surface area are studied. Sensitivity, detection limit and method of preparation have been highlighted to provide comprehensive information on all these techniques and their use in drug analysis.

Electrodes based on modified carbon paste for detecting various drugs and metabolites

Carbon paste electrodes (CPE) have a very large electroactive surface area, which can be renewed for a variety of applications. In addition, it has low ohmic resistance and high stability, reproducibility and lifetime, allowing it to analyse various drugs, revealing the oxidation and reduction processes involved due to its wide potential range. The sensitivity and speed of analysis of CPE can be significantly improved by modifying them with conductive materials. Different types of modifiers have been used. The modified CPE has been used to examine various drugs in various samples, including human urine, pharmaceutical formulations, plasma and serum.

The carbon paste electrode modified with poly(EBT) [26] exhibits very high electrocatalytic and semiconducting for detecting methdilazine hydrochloride (MDH), an antihistamine drug, in Dilosyn syrup and human urine using the SWV method (Figure 1). The MDH oxidation peak is observed at 0.675V with higher intensity compared to unmodified CPE. The surface area of poly-EBT/CPE is calculated to be 0.097 cm2, which is 2.30 times higher than that of CPE. The developed sensor shows good accuracy in both media (average recovery is 98.14 % in Dilosyn syrup and 97.4 % in human urine). The developed sensor has a low LOD of the order of 10-8 mol l-1 in the range of 0.1-50 μmol l-1 compared to other methods such as visible spectrophotometry (3.23 μmol l-1) [27], ultra-high performance reversed phase chromatography (0.254 μmol l-1 ) [28] and spectrophotometric method (1.62 μmol l-1 ) [29].

The researchers [30] reported that CPE/nanozeolite type X outperformed CPE/nanozeolite type A with a LOD of 0.2 μmol l-1 for paracetamol and 8 mmol l-1 for epinine.

A recent study described the analysis of the antifungal agent ketoconazole (KTC) in pharmaceutical and urine samples using Ce-BTC MOF/IL/CPE (cf.Table 1) [31]. The modifiers are synthesised according to the method of Liu [32]. The electrochemical behaviour is studied by chronoamperometry (CA), differential pulse voltammetry (DPV), cyclic voltammetry (CV) and linear sweep voltammetry (LSV). The LOD of Ce-BTC MOF/IL/CPE is 0.04 μmol l-1 in the range of 0.1-110.0 μmol l-1 . The sensitivity is found to be 0.1342 μA μmol-1 l.

J. Zoubir et al. [33] used silver nanoparticles to modify carbon graphite by electrodeposition. The fabricated sensor was used to detect metronidazole in milk and tap water with a detection limit of 0.206 μmol l-1 in the range 1-1000 μmol l-1 .

O. Vajdle et al. [34] used the drop coating method to modify CPE with gold nanoparticles. AuNPs/CPE were used to analyse four macrolide antibiotics by SWV. AuNPs/CPEs are used for azithromycin (AZI) detection in Hemomycin® by the SWV method. The oxidation peak of the AZI is identified at 0.77 V on the AuNPs/CPE, while the roxithromycin (ROX) peak is found at 0.65 V by CPE in the Runac®. Both electrodes were found to be valid by comparison with HPLC-DAD measurements.

On the other hand, nitric acid and sulphuric acid are used to increase -OH and -COOH groups on the surface of MWCNTs, which increases the surface area of the material [35]. In addition, the numerous carboxyl groups inhibit π-π interactions, which leads to a decrease in the adsorbency of MWCNTs [36]. Ofloxacin (OFX) adsorbs on MWCNT by binding its fluorine group to the -OH group of the CNT [37-40]. Flake graphite (FG) is used to increase the conductivity of MWCNTs weakened by the adsorption of compounds owing to its high degree of crystallisation. For this reason, M. Elfiky et al. [41] prepared an electrode by mixing graphite powder with FG and MWCNTs. The sensor has a larger electroactive area of 2.08 cm2 compared to the unmodified CPEs, with only 1.14 cm2. This finding is due to cracks on the irregular compact layers of the electrode. The electrode presented by the [10%FG/5%MW] CPE shows excellent performance in the analysis of ofloxacin in a commercial formulation (Ofloxacin® tablets) and in human urine samples with a LOD of 0.18 nmol l-1. OFX peak is observed at 0.85V by SW-AdAS

The analysis of AZI, an alkaline chemical, by HPLC presents a difficulty due to its adsorption on Si-OH chromatographic materials [42]. Therefore, electrochemiluminescence detection is considered an alternative method for the detection of this antibiotic due to its sensitivity and high productivity [43,44]. Electrochemiluminescence (ECL) is based on the redox process involved in the formation of excited states that are able to emit light. The concept of molecularly imprinted polymers (MIP) is based on imprinting a molecular cavity on the surface of the polymer using a template molecule, which is then removed while the polymer is still imprinted. This MIP can then be used to capture and detect a target molecule similar to the template molecule and specifically binds to a functional group in the cavity in the same way that an antibody binds to its antigen. There are many ways to make MIPs, such as soft lithography applied to giant molecules. L. Hu et al. [45] successfully analysed azithromycin (AZI) in urine and serum samples using MIP/CP ECL sensor, with MIP used as a recognisor. The oxidation peak of the amino groups of AZI [31] is observed at 0.8 V with LOD of 23 pmol l-1 in the range of 0.10-400 nmol l-1. The analytical results show the MIP/CP ECL sensor and HPLC agreement.

Clay has attractive characteristics, like high specific surface area and cation exchange capacity [46,47]. Bakary Tigana Djonse Justin et al. [48] recently fabricated a titanium dioxide modified carbon clay paste electrode (CPEA/TiO2/UV) that allows the analysis of ascorbic acid in pharmaceutical tablets using CV.The sensitivity of the electrode is enhanced by photoactivation of TiO2 by light irradiation (with a 100 W lamp), which produces electron (e)/hole (h+) pairs upon absorption of appropriate light energy [49]. The detection limit is 0.732 μmol l-1in the range of 0.15-0.850 μmol l-1 .

V. Vinoth et al. [50] modified a graphite carbon electrode with CuO microflowers (MFs) by solubilising them in Nafion to facilitate the modification. This electrode was named CuO MFs/Nafion/GC. The CuO MFs/Nafion/GC exhibited perfect electrocatalytic properties, selectivity in the presence of interfering molecules, stability and reproducibility. The amperometric analysis of glucose showed a sensitivity of 3.1 μA μmol-1 l cm-2 and LOD of 6.48 μmol l-1 between 10 to 120 μmol l-1 at +500 mV. This detection limit is very good compared to other sensors used for the same purpose such as NiO-SWCNT (907 μmol l-1 ) [51].Table 1 shows the different types of modified CPEs as well as the drugs detected and their detection medium, the limit of detection (LOD), concentration range and other characteristics.

The year of publication is indicated in each table in order to highlight the most recent publications and make them directly accessible to the reader.

Electrodes based on modified glassy carbon for detecting various drugs and metabolites

Glassy carbon is a type of carbon with an amorphous structure like glass and ceramics. Glassy carbon is an interesting material for electrochemical sensors because of its chemical resistance, low density and low electrical resistance. Glassy carbon electrodes (GCE) have also played an important function in electrochemical sensors and biosensors for drug analysis. The electron transfer rate at the GCE surface can be increased by various modifiers for the detection of medicines in diverse media.

The GCE/ZnO@NDCS/GOx is an enzymatic biosensor used to analysis glucose in serum (cf.Table 2) [54]. The biosensor had a reproducible sensitivity of 231.7 μA mol-1 l cm-2 with a LOD of 6.3 μmol l-1 at 0.57 V. The sensor selectivity is demonstrated via the addition of some interfering potentials such as ascorbic acid, dopamine, fructose, uric acid and mannose (Man) with glucose, resulting in only a small increase in current for each species. Due to the easy transfer of electrons through this biosensor, the response time is very short (<3 s). The biosensor is well suited for glucose analysis with recovery between 99.73-100.14 % and retains about 95.3 % of the initial response during 50 days of storage in human blood.

Acyclovir (ACV) (9-(2-hydroxyethoxymethyl)guanine) is a drug largely employed for the therapy of viral skin infections and neuritis [55-57]. Overdoses lead to adverse effects in patients [58,59]. The FeMoO4 compound is able to catalytically fix nitrogen [60] due to its interesting electrocatalytic property [61]. It also has interesting redox kinetics, making it a desirable material for electrode formation. Recently, Y. Wei et al. [62] prepared a sensor using graphene oxide composites loaded (ultrasonically) with ferrous molybdate (FeMoO4) for the analysis of ACV in pharmaceuticals by LSV. The oxidation peak of ACV is observed at 1.1 V. FeMoO4-GO/GCE and has two linearity intervals (0.1-10 and 10-100 μmol l-1 ) with a LOD of 20 nmol l-1. The reaction of ACV on the developed electrode is diffusion controlled with 2e- and 2H+ transferred. The active surface area of FeMoO4-GO/GCE is 1.59 times larger than that of FeMoO4/GCE, which clearly shows that the FeMoO4-GO improves the performance of the sensor.

Y. Huang et al. [63] fabricated a biosensor to study the human umami taste receptor (hT1R1) and the umami substances, such as monosodium glutamate (MSG), using a multilayer material to modify the glassy carbon electrode (Figure 2). During the fabrication of this electrode, a human umami taste receptor (hT1R1) was attached to the layers formed by the AuNPs [64]. Horseradish peroxidase (HRP) is used for direct electron transfer to the multilayer material formed [65]. The researchers suggest that hT1R1 is likely a receptor used by the body to sense nitrogen, opening up a new way of studying nutrient and drug adsorption.

In 2023, the same researchers [66] created another biosensor by attaching colon cancer and adjacent tissues to GCE to visualise the kinetics of responding to C and N nutrient receptors such as glucose and sodium lactate. In order to do this, they mixed solutions of starch gum with an aldehyde base and sodium alginate, which were spread over two microporous polycarbonate membranes into which the colon tissues were placed to build a layered assembly aligned to the GCE (Figure 3). Researchers found that the cells had different sensitivities to lactate, suggesting the possibility of using this nutrient to treat colon cancer. Colon cancer tissue is insensitive to lactate, whereas adjacent tissue is sensitive.

CoCo2O4 nanorods embedded in hexagonal boron nitride are used to modify the GCE for ronidazole determination [67]. The electrode synthesis presents a nano-LOD of 3 nmol l-1 between 0.01 and 1345 μmol l-1and a higher sensitivity of 5.845 μA μmol-1 l cm-2 using DPV. This sensitivity is explained by the enhanced adsorption and transport of mass ions via the formation of aggregates of hexagonal boron nitride (h-BN) and spinel cobalt oxide nanorods (CoCo2O4 NRs). CoCo2O4 NRs are used for their low cost, good stability and other properties. The h-BN, formed from boron and nitrogen, has a structure similar to graphene [68], which makes it an electroactive material [69,70].

Methotrexate (MTX) is a drug that inhibits the growth of tumour cells. It is applied for the treatment of certain types of cancer, notably breast cancer [71] and pulmonary cancer [72]. GO-Nafion-GCE sensor is constructed by dispersing graphite oxide in a solution containing Nafion-ethanol and then using it to modify the GCE [73]. Modified electrode stabilisation is achieved by using a CV of 0.1 V/s and in the potential between 0.5 and 1.2 V. GO-Nafion-GCE is able to detect methotrexate in MTX injection and urine with a LOD of 9 nmol l-1 between 0.4 and 20 μmol l-1 , that is close to the LOD of detection of the same drug by 3DPG-CNT/GCE [74]. The electrolyte solution used for the analysis of MXT is perchloric acid (0.03 mol l-1). A mixed adsorption-diffusion phenomenon controls the reaction process of MXT on the electrode surface.

Dopamine (DA) and paracetamol (PA) detection in synthetic urine was performed using a GCE-ERGO/polyCoTAPc electrode [75]. First, GCE was modified by electrochemically reduced graphene oxide (ERGO), which was simultaneously reduced and deposited from graphene oxide (GO). Finally, GCE-ERGO/polyCoTAPc was formed by electropolymerisation of cobalt (II) tetra-amino phthalocyanine. The developed electrode sensitivity was 1.32 μA mol-1 l cm-2 for PA and 8.39 μA mol-1 l cm-2 for DA. LOD is 0.10 μmol l-1 for PA and 0.095 μmol l-1 for DA using DPV.

The modification of GCE by QDs-P6LC-PEDOT:PSS allowed the amoxicillin (AMX) analysis in synthetic urine, whole milk and pharmaceuticals [76]. AMX oxidation peak is 0.88V and the reaction process on QDs-P6LC-PEDOT:PSS/GCE is irreversible controlled by diffusion process with an equal number of proton and electron exchange. The researchers found that the alkaline medium facilitates the deprotonation of AMX during the oxidation reaction. Furthermore, the detection limit is found to be 50 nmol l-1 in the range 0.90-69 μmol l-1 .

To form graphene (GR)-ZnO/GCE, X. Yue et al. [80] first polished the surface of GCE with a suspension of Al2O3 powder on a polishing cloth. Then, 8 μl of GR-ZnO nanocomposite suspension is used to modify the glassy carbon electrode. GR-ZnO/GCE was used for the simultaneous detection of sulfamethoxazole (SMZ) and trimethoprim (TMP) in urine and serum. SMZ and TMP oxidation peaks were successfully located at the Ep (SMZ) = 0.85 V and Ep (TMP) = 1.06 V by the DPV method, with a slight shift in peak potentials compared to those observed on the GCE. The researchers explained this by the ability of the GR to enhance ZnO nanorods conductivity and the reciprocal ability of the ZnO nanorods to avoid aggregation of the GR by reducing the van der Waals force. SMZ and TMP oxidation reactions are irreversible. They are controlled by adsorption. SMZ LOD is 0.4 μmol l-1 between 1-220 and 0.3 μmol l-1of TMP in the range of 1-180 μmol l-1 . The same drugs have been detected by GCE modified with GO and Ag nanoparticles (GC/rGO-AgNP) [81] using DPV with LOD (SMZ) = 0.6 μmol l-1 and LOD (TMP) = 0.4 μmol l-1 between 1.0 and 10.0 mmol l-1. In addition, SMZ and TMP exhibit irreversible oxidation peaks with ΔETMP-SMZ= 1.14-0.92 = 0.22V.

A further study has shown that the nickel ferrite/rGO (NiFe2O4/rGO) film is an excellent modifier of the GCE. Clenbuterol was analyzed in pig urine samples using the electrode produced [82]. The LOD is 0.17 μmol l-1 between 0.99 and 18.03 μmol l-1 using DPV.

Due to the excellent selectivity of chitosan [83] and the high selectivity and ease of preparation of the MIP method [84], Y. Wu et al. [85] fabricated a sensor called MIP-MWCNTs/GCE for the detection of tryptophan (Trp), a possible cause of schizophrenia [86], by depositing a printed chitosan film on the MWCNT pre-modified GCE surface. This pre-modification with MWCNTs aims to enhance the response of molecularly imprinted polymer electrodes [87]. MWCNTs are a type of carbon nanotube with highly active sites due to the presence of more concentric tubes [88], which gives them high adhesive activity and good conductivity. Y. Wu et al. [85] demonstrated hydrogen bonding between chitosan and Trp using Fourier transform infrared (FTIR) spectroscopy. The extraction reagent (ethanol) used during the extraction process causes the chitosan film to deflate and etch, forming a porous structure capable of efficiently binding Trp molecules to these imprinted sites (Figure 4). The oxidation process of Trp involves the exchange of an identical number of e- and H+. Analysis of Trp in human serum by MIP-MWCNTs/GCE showed a very encouraging recovery between 96.5 and 102.5 %. In the presence of several interferents, the Trp peak is very intense compared to the other substances. The MIP-MWCNTs/GCE sensor has a low LOD (1 nmol l-1) compared to the GCE sensor modified with AuNP and MWCNTs [89].

Daunorubicin is an anticancer drug [90] that requires dose control [91] to avoid adverse effects, including cardiac arrest. H. Karimi-Maleh et al. [92] attempted to develop a glassy carbon electrode modified with a nanocomposite of Pt/SWCNTs and ds-DNA (biorecognition) to eventually form ds-DNA/Pt/SWCNTs/GCE. DVP shows a positive displacement of the daunorubicin oxidation peak (from 847 to 882 mV), confirming the mutual intercalation reaction of ds-DNA (guanine base) and daunorubicin. Furthermore, the equilibrium constant of the association is 5.044×103 mol-1 l with a nanometre LOD (1.0 nmol l-1) over a concentration between 4.0 nmol l-1 and 250.0 μmol l-1.

Using the Fe3O4/MWCNT/GCE electrode, T. Bhengo et al. [93] obtained oxidation peaks of sulfamethoxazole and trimethoprim at 910 and 1120 mV, respectively, with detection limits of 11.0 nmol l-1 for SMZ and 21.0 nmol l-1 for TMP using the DPV method. Similarly, Rajasekhar Chokkareddy et al. [94] deposited IL-f-ZnONPs@MWCNTson the clean surface of GCE by dispersing them in dimethylformamide (cf.Table 2).To prepare a solution containing SMZ, the researchers crushed two Sandoz Co-Trimoxazole (Pharmaceuticals) tablets and dissolved part of the powder in methanol. After shaking, the solution was refined and diluted with PBS (pH 6.0). The resulting solution was then added to the pharmaceutical urine samples. The cyclic voltammetry oxidation peak of the SMX was found to be at 620 mV. The reaction process on IL-f-ZnONPs@MWCNTs@GCE is diffusion controlled and the detection limit is 0.1 ng/ml in the range of 0.1-10 ng ml-1. In addition, Ag-MWCNT/MTOAC/GCE [95] was successfully used to detect SMX in Co-trimoxazole (mg per tablet), Cotrim paediatric and human urine by DPV. The samples were diluted with phosphate solution to reduce the matrix effect and then introduced into the electrochemical cell. The irreversible oxidation of the SMX is observed at +0.85 V and involves the exchange of an equal number of e- and H+ and is controlled by adsorption.

On the other hand, the simultaneous analysis of ascorbic acid (AA), dopamine and uric acid in human urine was investigated using a GCE-based sensor modified with 2D titanium carbide nanoplatelets (MXene) [96]. All three analytes showed remarkably distinct oxidation peaks. AA was observed at 0.001 V, while dopamine and uric acid were at 210 mV and 330 mV, respectively. It should be noted that 2D titanium carbide (MXene) nanoplatelets are mentioned in the literature as having a high conductivity of 9880 S cm-1 [102], higher than that of graphene. In particular, MXene groups, such as fluor, oxygen and hydroxyl, increase its hydrophilicity and allow rapid access to the analyte. It is therefore used as an ideal material for sensors [103].

Table 2 shows the different types of modified GCEs as well as the drugs detected and their detection medium, the limit of detection (LOD), concentration range and other characteristics.

Electrodes based on modified graphene oxide for detecting various drugs and metabolites

Graphene oxide reduced (rGO) is obtained by thermal, electrochemical or chemical treatment of graphene oxide using substances such as NaBH4 or aluminium powder. This reduction removes oxidised functional groups (which increases the sensitivity of the GO and makes it insulating) and creates a defect structure (active sites) characteristic of rGO. These properties have enabled rGO to have high electrochemical activity compared to graphene and graphene oxide.

F. Zhou et al. [104] successfully fabricated a Nafion/GOx/Au-ZnO/rGO/ITO electrode. The researchers found that the rate and efficiency of electron transfer were augmented by the deposition of AuNPs and UV irradiation, allowing a significant increase in glucose sensitivity (cf.Table 3). LOD of this sensor is 0.2 μmol l-1 by amperometric method between 0 and 9.5 mmol l-1 with a higher Michaelis-Menten constant (15.54 mmol l-1) than that of non-UV irradiated Nafion/GOx/Au-ZnO/rGO/ITO. The sensors have exceptional detection accuracy for blood glucose measurement.

Regorafenib (REG) or Regonix is an anticancer drug used in several types of cancer [105,106]. Monitoring the concentration of REG in serum or blood seems to be of great interest as doses above certain limits can cause adverse effects on vital organs of the body [107]. REG is detected in real samples by Pd-Ru/rGO using the DPV technique [108] (cf.Table 3). The acidic graphene neutrality is obtained from pomegranate peel extract (PPE). In fact, the redox current peak is linear with sv (v = potential sweep rate), indicating that the REG reaction occurring at Pd-Ru/rGO is diffusion controlled, with a constant heterogeneous charge transfer rate equal to K0 = 2.29 s-1, calculated based on Laviron's equation (cf.Eq. (1),(2) and(3)) [109]. Furthermore, the REG oxidation involves the exchange of 2e- and 2H+.

(1)
ADMET-11-1709-e001.jpg
(2)
ADMET-11-1709-e002.jpg
(3)
ADMET-11-1709-e003.jpg

Table 3 shows the different types of modified rGOs as well as the drugs detected and their detection medium, limit of detection (LOD), concentration range and other characteristics.

Electrodes based on modified screen-printed carbon electrodes for detecting various drugs and metabolites

The screen-printed electrode (SPE) consists of a substrate (PVC, ceramic) onto which miniature electrodes can be printed using ink based on conductive materials. Various substances are employed to increase the sensitivity of the screen-printed electrode, including fullerenes (C60) and iron(III) nanoparticles (Fe3O4NP).

The antibiotic drug furaltadone (FLT) was detected by a screen-printed carbon electrode (SPCE) decorated with Cu/Ni/TiO2/MWCNTs nanocomposites [111]. FLT reduction peak on Cu/Ni/TiO2/MWCNTs nanocomposites was located at -470 mV with a significant current density of -39.17 μA. The reduction of the R-NO2 group to R-NHOH was performed by electron transfer from HOMO to LUMO in the fabricated electrode and the LOD is equal to 0.0949 μmol l-1 with a high sensitivity of 1.9288 μA μmol-1 l cm-2 in water. We also find that erythronmycin (ERY) is detected by sodium dodecyl sulfate-modified SPCEs (SPCEs/SDS) by flow injection analysis with amperometric detection in PBS of pH 8.5 [112]. Peak detection of ERY is 0.7 V with an LOD of 0.19 μmol l-1.

A SPE coated with C60, rGO and Nafion (NF) is used to analysis antibiotic metronidazole (MTZ) in real samples (cf.Table 4) [113]. Reduction of GO with NaBH4 results in the disappearance of oxygen groups. Fullerenes are used because of their ability to operate at low potentials, which is advantageous for a sensor to avoid interference effects [114,115]. To stabilise the response of the C60-rGO-NF/SPE, the researchers applied two cyclic voltammetry cycles between 0 and -1500 mV at v = 20 mV s-1 in a 1 mol l-1 electrolyte solution of KOH and then the electrode was placed in PBS (Neutre) to apply v = 50 mV s-1 (scan rate) between 550 and -50 mV. The MTZ reduction peak observed at -0.9 V, representing the exchange of 4e- and 4H+. The intensity of the MTZ reduction peak on C60-rGO-NF/SPE is five times higher than that observed on SPE. This sensitivity is explained by the porosity of C60, rGO and NF, which increases the surface area of C60-rGO-NF/SPE. The LOD by SWV is 0.21 μmol l-1in the range 0.25-34.0 μmol l-1.

GP-CAc/PVC is a screen-printed electrode (SPE) [116], estimated to cost 0.016 $ per unit compared to 7 $ for a commercial electrode, used to determine levodopa (L-dopa) in two commercial drugs, Parkidopa® and Ekson®. These drugs are prescribed to treat Parkinson's disease by reducing the severity of symptoms. To maintain the conductive material of the electrode, researchers have used polymeric materials such as cellulose [117], polyvinyl [118], epoxy glue, nail polish [119] and cellulose acetate (CAc) [120]. In this case, the conductive ink for the electrode is obtained by mixing cellulose acetate and graphite powder in attendance of acetone/cyclohexanone (apolar solvents). The adhesive stencil is used to draw the relief of three electrodes, which are then attached to the PVC. Conductive ink is then applied to the stencil to print the electrodes on the PVC. The stencil is immediately removed to allow the ink to run off. The GP-CAc/PVC has an electroactive surface area of 0.48 cm2 with a sensitivity of 0.101 μA μmol-1 l owing to the significant porosity of the cellulose acetate. The detection limit by SWV is 0.06 μmol l-1 between 8.00 and 100 μmol l-1. The researchers found that at pH 5 (pH ≥ 5.0), L-dopa exhibits an irreversible reaction with an oxidation peak located at 0.25 V caused by exchanging the 2H+ and 2e-. Furthermore, at pH 2 (pH ≥ 4.0), the behaviour of L-dopa changes towards a reversible system with Ep(oxy) = 0.27V and Ep(red) = -0.01V.

On the other hand, SPE modified with the CuO/Co3O4 nanocomposite integrated MWCNTs [121] has a very low LOD of 0.223 pmol l-1 between 10-12 and 10-2 mol l-1 range for urea determination by electrochemical impedance spectroscopy (EIS) (Fig. 5). This performance is due to the combination of CuO and Co3O4, which have higher electronic conductivity than Co3O4 or CuO alone.

Table 4 shows the different types of modified SPEs as well as the drugs detected and their detection medium, limit of detection (LOD), concentration range and other characteristics.

Conclusions and perspectives

This review discusses the different types of recently published electrochemical sensors for the detection of drugs and metabolites in pharmaceutical and biological samples. CPE has a very large electroactive surface area, which can be renewed for a variety of applications. In addition, it has low ohmic resistance and high stability, reproducibility and lifetime, giving it the ability to analyse various drugs, revealing the oxidation and reduction processes involved due to its wide potential range. The sensitivity and speed of analysis of CPE can be significantly improved by modifying them with conductive materials. Various modifiers were used, including organic modifiers like Poly-Eriochrome Black T (Poly-EBT), benzene tricarboxylic acid (BTC), metal-organic frameworks (MOFs) or inorganic modifiers like silver nanoparticles (AgNPs), CuO microfragments (CuO MFs) and TiO2. On the other hand, the improvement of CPE can be achieved by using a carbon nanomaterial modifier such as flake graphite (FG), multi-wall carbon nanotubes (MWCNTs) and carbon quantum dots (CQDs). The modified carbon past electrode has been used for the determination of various drugs such as methdilazine hydrochloride (MDH), ketoconazole (KTC), metronidazole (MTZ), sulfamethoxazole (SMZ) in diverse samples such as human urine, pharmaceutical formulations, plasma and serum. Modification with silver nanoparticles is also encouraged due to their biocompatibility, sensitivity, stability and ability to increase peak intensity due to their high conductivity. Similarly, metal oxide nanomaterials such as CuO MFs have an essential contribution to the performance of CPE owing to their wide surface area. L. Hu et al. fabricated a selective electrochemiluminescence (ECL) sensor by modifying an CPE with a MIP. The sensor showed a very low LOD of 0.023 nmol l-1 to analyse azithromycin (AZI) in real samples.The very low LOD of 0.18 nmol l-1 is also found by a CPE modified with lamellar graphite and MWCNT for the analysis of ofloxacin in pharmaceutical tablets and human urine samples. Similarly, CPE modified with Fe3O4/ZIF-67/ILCPE nanocomposite was applied for the analysis of SMZ in urine and water with a LOD of 5.0 nmol l-1.

The transfer rate of electrons at the surface of glassy carbon electrodes (GCE) can be increased by various modifiers such as nitrogen-doped carbon sheets, glucose oxidase, cobalt (II) polymer tetraamino phthalocyanine (polyCoTAPc), methyltrioctyl ammonium chloride (MTOAC) and guanine (ds-DNA). In addition, inorganic modifiers such as iron molybdate (FeMoO4), spinel cobalt oxide nanorods (CoCo2O4 NRs), hexagonal boron nitride (h-BN), cobalt (II) tetra-aminophthalocyanine polymer (polyCoTAPc), cadmium telluride quantum dots (QDs) and titanium carbide (MXene) nanosheets (Ti-C-Tx) have been used for the same purpose. Similarly, a variety of carbon types have been reported in the literature to improve GCE sensitivity, including GO, electrochemically reduced graphene oxide (ERGO), Printex 6L carbon (P6LC), CMK-3 mesoporous carbon (CMK-3), carbon black (CB), graphene (GR), reduced graphene (rGO), graphene-like carbon architecture (HPG) and carboxylic MWCNT (MWCNTseCOOH). The modified GCE was used to determine the drugs in the different matrices. The drugs analysed were: ronidazole, methotrexate, dopamine, amoxicillin, nimesulide, lomefloxacin, sulphamethoxazole, trimethoprim, tryptophan and daunorubicin. Platinum nanoparticles have high electrocatalytic properties. Similarly, DNA nanostructures have high selectivity and affinity for the analyte, which can be used to modify the GCE used for electrochemical drug analysis. The conductivity of the glassy carbon electrodes can be increased by using polymers such as polyaniline (PANI). The combination of various modifiers with high conductivity electrochemical materials such as MIPs, MWCNTs, SWCNTs, ds-DNA and Pt has further increased the sensitivity of the sensors. MIP-MWCNTs/GCE and ds-DNA/Pt/SWCNTs/GCE showed a very low LOD of 1 nmol l-1 for drug determination. In addition, the modification with 3D-HPG/PTH shows a similar nanometric detection limit. In 2023, a group of researchers successfully fabricated a glassy carbon electrode modified with CoCo2O4 NRs/h-BN used to analysis the MTR in pharmaceuticals, with a sensitivity of 5.845 μA μmol-1 l cm-2 and a LOD of 3 nmol l-1.

AuNPs have been employed as modifiers for rGO due to their considerable catalytic activity, biocompatibility and high electrochemical potential, which allows the redox process generated at the electrode surface to be detected. Other materials are employed to improve the sensitivity of rGOs, including glucose oxidase, indium tin oxide (ITO), metal-organic framework (MOF TMU-22) and palladium/ruthenium nanoparticles (Pd-Ru). The modification of rGO with Pd-Ru resulted in a very low LOD of 1.6 nmol l-1 for the determination of regorafenib in various samples like human blood, plasma and pharmaceutical formulations.

A variety of conductive compounds are available to increase the sensitivity of the screen-printed electrode, like sodium dodecyl sulphate (SDS), Nafion (NF), fullerene (C60), cellulose acetate, graphite powder, antimony oxide nanoparticles (AONP) and iron (III) nanoparticles (Fe3O4NP). In addition, multi-wall carbon nanotubes (MWNTs) have a high active surface area and conductivity. The specific structure of these nanomaterials gives them the potential to be an interesting candidate for drug detection electrochemical sensors. The combination of metal oxide and carbon nanotubes significantly increases the sensitivity of SPE. Recently, H. S. Magar et al. fabricated an SPE using a combination of CuO/Co3O4 and MWCNTs. The sensor was used for urea analysis with a very low LOD of 0.223 pmol l-1. Not all electrode types mentioned in this paper reported this value. In addition, a LOD of 2.5 nmol l-1 was found for nano-Au/MWNTs-ZnO/SPE used to analysis EPI in whole blood and pharmaceutical samples.

The use of nanomaterials and metal oxides seems promising to improve the sensitivity of the electrodes, but there are many problems, such as difficulties in fabrication, handling and characterisation, which encourage investment in finding simpler fabrication methods or extraction from plants and animal secretions.

Acknowledgements

We are grateful to Mr. Haroun Laghlimi (My dear son), Mrs. Fatna NÂna and Mrs. Ikram Ait Lebbadfor their encouragement and support.

Notes

[1] Conflicts of interest Conflict of interest: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

List of abbreviations

C60

Buckminsterfullerenes

CA

Chronoamperometry

CPE

Carbon paste electrodes

CPEA

Carbon clay paste electrode

CV

Cyclic voltammetry

DPV

Differential pulse voltammetry

EBT

Eriochrome black T

ECL

Electrochemiluminescence

EIS

Electrochemical impedance spectroscopy

ERGO

Electrochemically reduced graphene oxide

FG

Flake graphite

FTIR

Fourier transform infrared

GCE

Glassy carbon electrode

GO

Graphene oxide

GP

Graphite powder

GR

Graphene

HOMO

Highest occupied molecular orbital

HPLC-DAD

High performance liquid chromatography-diode array detector

LOD

Limit of detection

LSV

Linear scan voltammetry

LUMO

Lowest unoccupied molecular orbital

MIP

Molecularly imprinted polymer

MWCNTs

Multi-wall carbon nanotubes

MXene

M: transition metal, X: carbon and/or nitrogen, ene: similarity to graphene

NF

Nafion

PBS

Phosphate-buffered saline

PVC

Polyvinyl chloride

rGO

Reduced graphene oxide

SDLSV

Second-order derivative linear sweep voltammetry

SPC

Screen-printed carbon

SPCE

Screen-printed carbon electrode

SW-AdAS

Square wave adsorptive anodic stripping voltammetry

SWCNT

Single-walled carbon nanotubes

SWV

Square wave voltammetry

TLC-densitometric

Thin layer chromatography densitometric

References

[1] 

Somvanshi S.B.; Kharat P.B.; Jadhav K.M.; Thorat N.D.; Townley H., Nanomaterials and pharmacokinetics, in Nano-Pharmacokinetics and Theranostics, Academic Press, 2021, p. 1-14. https://doi.org/10.1016/B978-0-323-85050-6.00007-4 https://doi.org/10.1016/B978-0-323-85050-6.00007-4

[2] 

Khorshed A.A.; Khairy M.; Elsafty S.A.; Banks C.E.. Disposable screen-printed electrodes modified with uniform iron oxide nanocubes for the simple electrochemical determination of meclizine, an antihistamine drug. Analytical Methods 11 (2019) 282-287. https://doi.org/10.1039/C8AY02405G https://doi.org/10.1039/C8AY02405G

[3] 

Ibrahim F.; El-Yazbi A.; Wagih M.; Barary M.. Sensitive inexpensive spectrophotometric and spectrofluorimetric analysis of ezogabine, levetiracetam and topiramate in tablet formulations using Hantzsch condensation reaction. Spectrochimica Acta A 184 (2017) 47-60. https://doi.org/10.1016/j.saa.2017.04.078 https://doi.org/10.1016/j.saa.2017.04.078

[4] 

Tamilselvi N.; Rajasekaran A.. Stability-indicating RP-HPLC method for the determination of ezogabine and identification of its degradation products. Journal of Pharmaceutical Sciences and Research 8 (2016) 19–23.

[5] 

Habib N.M.; Abdelrahman M.M.; Abdelwhab N.S.; Ali N.W.. Validated chromatographic methods for the analysis of two binary mixtures containing pyridoxine hydrochloride. Journal of Association of Official Agricultural Chemists International 100 (2017) 414-421. https://doi.org/10.5740/jaoacint.16-0213 https://doi.org/10.5740/jaoacint.16-0213

[6] 

Peraman R.; Manikala M.; Kondreddy V.K.; Yiragamreddy P.R.. A stability-indicating RP-HPLC method for the quantitative analysis of meclizine hydrochloride in tablet dosage form. Journal of Chromatographic Science 53 (2015) 793-799. https://doi.org/10.1093/chromsci/bmu127 https://doi.org/10.1093/chromsci/bmu127

[7] 

Mohamed M.A.; Fayed A.S.; Hegazy M.A.; Salama N.N.; Abbas E.E.. Fully optimized new sensitive electrochemical sensing platform for the selective determination of antiepileptic drug ezogabine. Microchemical Journal 144 (2019) 130-138. https://doi.org/10.1016/j.microc.2018.08.062 https://doi.org/10.1016/j.microc.2018.08.062

[8] 

Karimi-Maleh H.; Darabi R.; Karimi F.; Karaman C.; Shahidi S.A.; Zare N.; Baghayeri M.; Fu L.; Rostamnia S.; Rouhi J.; Rajendran S.. State-of-art advances on removal, degradation and electrochemical monitoring of 4-aminophenol pollutants in real samples: A review. Environmental Research 222 (2023) 115338. https://doi.org/10.1016/j.envres.2023.115338 https://doi.org/10.1016/j.envres.2023.115338

[9] 

Hojjati-Najafabadi A.; Mansoorianfar M.; Liang T.; Shahin K.; Karimi-Maleh H.. A review on magnetic sensors for monitoring of hazardous pollutants in water resources. Science of The Total Environment 824 (2022) 153844. https://doi.org/10.1016/j.scitotenv.2022.153844 https://doi.org/10.1016/j.scitotenv.2022.153844

[10] 

Buledi J.A.; Mahar N.; Mallah A.; Solangi A.R.; Palabiyik I.M.; Qambrani N.; Karimi F.; Vasseghian Y.; Karimi-Maleh H.. Electrochemical quantification of mancozeb through tungsten oxide/reduced graphene oxide nanocomposite: a potential method for environmental remediation. Food and Chemical Toxicology 161 (2022) 112843. https://doi.org/10.1016/j.fct.2022.112843 https://doi.org/10.1016/j.fct.2022.112843

[11] 

Cheraghi S.; Taher M.A.; Karimi-Maleh H.; Karimi F.; Shabani-Nooshabadi M.; Alizadeh M.; Al-Othman A.; Erk N.; Yegya Raman P.K.; Karaman C.. Novel enzymatic graphene oxide based biosensor for the detection of glutathione in biological body fluids. Chemosphere 287 (2022) 132187. https://doi.org/10.1016/j.chemosphere.2021.132187 https://doi.org/10.1016/j.chemosphere.2021.132187

[12] 

Mohanraj J.; Durgalakshmi D.; Rakkesh R.A.; Balakumar S.; Rajendran S.; Karimi-Maleh H.. Facile synthesis of paper based graphene electrodes for point of care devices: A double stranded DNA (dsDNA) biosensor. Journal of Colloid and Interface Science 566 (2020) 463-472. https://doi.org/10.1016/j.jcis.2020.01.089 https://doi.org/10.1016/j.jcis.2020.01.089

[13] 

Moutcine A.; Laghlimi C.; Ifguis O.; Smaini M.A.; El Qouatli S.E.; Hammi M.; Chtaini A.. A novel carbon paste electrode modified by NP-Al2O3 for the electrochemical simultaneous detection of Pb (II) and Hg (II). Diamond and Related Materials 104 (2020) 107747. https://doi.org/10.1016/j.diamond.2020.107747 https://doi.org/10.1016/j.diamond.2020.107747

[14] 

Moutcine A.; Laghlimi C.; Ziat Y.; Smaini M.A.; El Qouatli S.E.; Hammi M.; Chtaini A.. Preparation, characterization and simultaneous electrochemical detection toward Cd (II) and Hg (II) of a phosphate/zinc oxide modified carbon paste electrode. Inorganic Chemistry Communications 116 (2020) 107911. https://doi.org/10.1016/j.inoche.2020.107911 https://doi.org/10.1016/j.inoche.2020.107911

[15] 

Laghlimi C.; Ziat Y.; Moutcine A.; Hammi M.; Zarhri Z.; Ifguis O.; Chtaini A.. A new sensor based on graphite carbon paste modified by an organic molecule for efficient. Chemical Data Collections 31 (2021) 100595. https://doi.org/10.1016/j.cdc.2020.100595 https://doi.org/10.1016/j.cdc.2020.100595

[16] 

Laghlimi C.; Ziat Y.; Moutcine A.; Hammi M.; Zarhri Z.; Maallah R.; Ifguis O.; Chtaini A.. Analysis of Pb (II), Cu (II) and Co (II) in drinking water by a new carbon paste electrode modified with an organic molecule. Chemical Data Collections 29 (2020) 100496. https://doi.org/10.1016/j.cdc.2020.100496 https://doi.org/10.1016/j.cdc.2020.100496

[17] 

Smaini M.A.; Smaini I.; Ennachete M.; Laghlimi C.; Saâdane H.; Moutcine A.; Chtaini A.. Electrochemical determination of adenosine by natural phosphate modified carbon paste electrode: analytical application in serum. Sensing and Bio-Sensing Research 23 (2019) 100272. https://doi.org/10.1016/j.sbsr.2019.100272 https://doi.org/10.1016/j.sbsr.2019.100272

[18] 

Ifguis O.; Moutcine A.; Laghlimi C.; Ziat Y.; Bouhdadi R.; Chtaini A.; Moubarik A.; Mbarki M.. Biopolymer-Modified Carbon Paste Electrode for the Electrochemical Detection of Pb (II) in Water. Journal of Analytical Methods in Chemistry 2022 (2022). https://doi.org/10.1155/2022/5348246 https://doi.org/10.1155/2022/5348246

[19] 

Maallah R.; Moutcine A.; Laghlimi C.; Smaini M.A.; Chtaini A.. Electrochemical bio-sensor for degradation of phenol in the environment. Sensing and Bio-Sensing Research 24 (2019) 100279. https://doi.org/10.1016/j.sbsr.2019.100279 https://doi.org/10.1016/j.sbsr.2019.100279

[20] 

Moutcine A.; Ifguis O.; Samaini M.A.; Ennachete M.; Sâadane H.; Laghlimi C.; Chtaini A.. Simultaneous electrochemical determination of heavy metals by an electrode modified CPE-NP-Al2O3. Materials Today: Proceedings 53 (2022) 404-407. https://doi.org/10.1016/j.matpr.2022.01.388 https://doi.org/10.1016/j.matpr.2022.01.388

[21] 

El Hamdouni Y.; El Hajjaji S.; Szabo T.; Trif L.; Felhősi I.; Abbi K.; Labjar N.; Harmouche L.; Shaban A.. Biomass valorization of walnut shell into biochar as a resource for electrochemical simultaneous detection of heavy metal ions in water and soil samples: Preparation, characterization, and applications. Arabian Journal of Chemistry 15 (2022) 104252. https://doi.org/10.1016/j.arabjc.2022.104252 https://doi.org/10.1016/j.arabjc.2022.104252

[22] 

Mohamed M.A.; Yehia A.M.; Banks C.E.; Allam N.K.. Novel MWCNTs/graphene oxide/pyrogallol composite with enhanced sensitivity for biosensing applications. Biosensors and Bioelectronics 89 (2017) 1034–1041. https://doi.org/10.1016/j.bios.2016.10.025 https://doi.org/10.1016/j.bios.2016.10.025

[23] 

Lotfi S.; Veisi H.. Electrochemical determination of clonazepam drug based on glassy carbon electrode modified with Fe3O4/R-SH/Pd nanocomposite. Materials Science and Engineering : C 103 (2019) 109754. https://doi.org/10.1016/j.msec.2019.109754 https://doi.org/10.1016/j.msec.2019.109754

[24] 

Fallah M.; Rahimnejad M.; Asghary M.; Mashkour M.. An electrochemical sensor based on a carbon paste electrode for the determination of buserelin. Analytical Methods 12 (2020) 33-38. https://doi.org/10.1039/C9AY01760G https://doi.org/10.1039/C9AY01760G

[25] 

Zarezadeh A.; Rajabi H.R.; Sheydaei O.; Khajehsharifi H.. Application of a nano-structured molecularly imprinted polymer as an efficient modifier for the design of captopril drug selective sensor: Mechanism study and quantitative determination. Materials Science and Engineering: C 94 (2019) 879-885. https://doi.org/10.1016/j.msec.2018.10.042 https://doi.org/10.1016/j.msec.2018.10.042

[26] 

Shetti N.P.; Ilager D.; Malode S.J.; Monga D.; Basu S.; Reddy K.R.. Poly (eriochrome black T) modified electrode for electrosensing of methdilazine. Materials Science in Semiconductor Processing 120 (2020) 105261. https://doi.org/10.1016/j.mssp.2020.105261 https://doi.org/10.1016/j.mssp.2020.105261

[27] 

Basavaiah K.; Charan V.S.. The Use of Chloranilic Acid for the Spectrophotometric Determination of Three Antihistamines. Turkish Journal of Chemistry 26 (2002) 653-661. https://journals.tubitak.gov.tr/chem/vol26/iss5/3/

[28] 

Raghu M.S.; Cijo K.B.; Xavier M.; Prashanth K.N.. Development and Validation of Stability-Indicating RP-UPLC Method for the Determination of Methdilazine in Bulk Drug and in Pharmaceutical Dosage Form. International Scholarly Research Network Chromatography 2012 (2012) 1-8. https://doi.org/10.5402/2012/916932 https://doi.org/10.5402/2012/916932

[29] 

Raghu M.S.; Basavaiah K.. Simple, sensitive and selective spectrophotometric methods for the determination of methdilazine in pharmaceuticals through charge transfer complex formation reaction. Proceedings of the Indian National Science Academy 79 (2013) 65-78. http://eprints.uni-mysore.ac.in/10806/

[30] 

Kemmegne-Mbouguen J.C.; Tchoumi F.P.. Synthesis of nanozeolites type A and X from quartz-rich Cameroonian kaolin: application to the modification of carbon paste electrode for acetaminophen and epinine electrochemical sensing. Journal of Solid State Electrochemistry 27 (2023) 1-15. https://doi.org/10.1007/s10008-022-05355-z https://doi.org/10.1007/s10008-022-05355-z

[31] 

Tajik S.; Sharifi F.; Aflatoonian B.; Di Bartolomeo A.. A New Electrochemical Sensor for the Detection of Ketoconazole Using Carbon Paste Electrode Modified with Sheaf-like Ce-BTC MOF Nanostructure and Ionic Liquid. Nanomaterials 13 (2023) 523. https://doi.org/10.3390/nano13030523 https://doi.org/10.3390/nano13030523

[32] 

Liu K.; You H.; Jia G.; Zheng Y.; Huang Y.; Song Y.; Yang M.; Zhang L.; Zhang H.. Hierarchically Nanostructured Coordination Polymer: Facile and Rapid Fabrication and Tunable Morphologies. Crystal Growth & Design 10 (2010) 790-797. https://doi.org/10.1021/cg901170j https://doi.org/10.1021/cg901170j

[33] 

Zoubir J.; Bougdour N.; Hayaoui W.E.; Radaa C.; Idlahcen A.; Assabbane A.; Bakas I.. Electrochemical Detection of Metronidazole Using Silver Nanoparticle-Modified Carbon Paste Electrode. Electrocatalysis 13 (2022) 386-401. https://doi.org/10.1007/s12678-022-00722-w https://doi.org/10.1007/s12678-022-00722-w

[34] 

Vajdle O.; Šekuljica S.; Guzsvány V.; Nagy L.; Kónya Z.; Ivić M.A.; Mijin D.; Petrović S.; Anojčić J.. Use of carbon paste electrode and modified by gold nanoparticles for selected macrolide antibiotics determination as standard and in pharmaceutical preparations. Journal of Electroanalytical Chemistry 873 (2020) 114324. https://doi.org/10.1016/j.jelechem.2020.114324 https://doi.org/10.1016/j.jelechem.2020.114324

[35] 

Carabineiro S.A.C.; Thavorn-Amornsri T.; Pereira M.F.; Figueiredo J.L.. Adsorption of ciprofloxacin on surface-modified carbon materials. Water Research 45 (2011) 4583-4591. https://doi.org/10.1016/j.watres.2011.06.008 https://doi.org/10.1016/j.watres.2011.06.008

[36] 

Sheng G.D.; Shao D.D.; Ren X.M.; Wang X.Q.; Li J.X.; Chen Y.X.; Wang X.K.. Kinetics and thermosdynamics of adsorption of ionizable aromatic compounds from aqueous solutions by as-prepared and oxidized multiwalled carbon nanotubes. Journal of Hazardous Materials 178 (2010) 505-516. https://doi.org/10.1016/j.jhazmat.2010.01.110 https://doi.org/10.1016/j.jhazmat.2010.01.110

[37] 

Peng H.; Pan B.; Wu M.; Liu R.; Zhang D.; Wu D.; Xing B.. Adsorption of ofloxacin on carbon nanotubes: solubility, pH and cosolvent effects. Journal of Hazardous Materials 211 (2012) 342-348. https://doi.org/10.1016/j.jhazmat.2011.12.063 https://doi.org/10.1016/j.jhazmat.2011.12.063

[38] 

Keiluweit M.; Kleber M.. Molecular-level interactions in soils and sediments: the role of aromatic π-systems. Environmental Science and Technology 43 (2009) 3421-3429. https://doi.org/10.1021/es8033044 https://doi.org/10.1021/es8033044

[39] 

Lin D.; Xing B.. Adsorption of phenolic compounds by carbon nanotubes: role of aromaticity and substitution of hydroxyl groups. Environmental Science and Technology 42 (2008) 7254-7259. https://doi.org/10.1021/es801297u https://doi.org/10.1021/es801297u

[40] 

Lee L.S.; Pignatello J.J.. Evidence π–π for electron donor–acceptor interactions between π-donor aromatic compounds and π-acceptor sites in soil organic matter through pH effects on sorption. Environmental Science and Technology 38 (2004) 4361-4368. https://doi.org/10.1021/es035379e https://doi.org/10.1021/es035379e

[41] 

Elfiky M.; Salahuddin N.; Hassanein A.; Matsuda A.; Hattori T.. Detection of antibiotic Ofloxacin drug in urine using electrochemical sensor based on synergistic effect of different morphological carbon materials. Microchemical Journal 146 (2019) 170-177. https://doi.org/10.1016/j.microc.2018.12.034 https://doi.org/10.1016/j.microc.2018.12.034

[42] 

Chang Y.; Wang L.X.; Li Y.P.; Hu C.Q.. Factors influencing the HPLC determination for related substances of Azithromycin. Journal of Chromatographic Science 54 (2016) 187-194. https://doi.org/10.1093/chromsci/bmv127 https://doi.org/10.1093/chromsci/bmv127

[43] 

Wu B.; Guo Y.; Cao H.; Zhang Y.; Yu L.; Jia N.. A novel mesoporous molecularsieves-based electrochemilumenescence sensor for sensitive detection of azithromycin. Sensors and Actuators B: Chemical 186 (2013) 219-225. https://doi.org/10.1016/j.snb.2013.05.080 https://doi.org/10.1016/j.snb.2013.05.080

[44] 

Liu Y.M.; Shi Y.M.; Liu Z.L.; Tian W.. A sensitive method for simultaneous determination of four macrolides by CE with electrochemiluminescence detection and its applications in human urine and tablets. Electrophoresis 31 (2010) 364-370. https://doi.org/10.1002/elps.200900302 https://doi.org/10.1002/elps.200900302

[45] 

Hu L.; Zhou T.; Feng J.; Jin H.; Tao Y.; Luo D.; Mei S.; Lee Y.I.. A rapid and sensitive molecularly imprinted electrochemiluminescence sensor for Azithromycin determination in biological samples. Journal of Electroanalytical Chemistry 813 (2018) 1-8. https://doi.org/10.1016/j.jelechem.2018.02.010 https://doi.org/10.1016/j.jelechem.2018.02.010

[46] 

Zhu T.T.; Zhou C.H.; Kabwe F.B.; Wu Q.Q.; Li C.S.; Zhang J.R.. Exfoliation of montmorillonite and related properties of clay/polymernanocomposites. Applied Clay Science 169 (2019) 48-66. https://doi.org/10.1016/j.clay.2018.12.006 https://doi.org/10.1016/j.clay.2018.12.006

[47] 

Tournassat C.; Steefel C.I.; Bourg I.C.; Bergaya F.. Surface properties of clay minerals. Developments in Clay Science 6 (2015) 5-31. https://doi.org/10.1016/B978-0-08-100027-4.00001-2 https://doi.org/10.1016/B978-0-08-100027-4.00001-2

[48] 

Justin B.T.D.; Blaise N.; Valery H.G.. Investigation of the photoactivation effect of TiO2 onto carbon-clay paste electrode by cyclic voltammetry analysis. Heliyon 9 (2023) 13474. https://doi.org/10.1016/j.heliyon.2023.e13474 https://doi.org/10.1016/j.heliyon.2023.e13474

[49] 

Pawar M.; Topcu Sendoğdular S.; Gouma P.. A brief overview of TiO2 photocatalyst for organic dye remediation: case study of reaction mechanisms involved in Ce-TiO2 photocatalysts system. Journal of Nanomaterials 2018 (2018) 13. https://doi.org/10.1155/2018/5953609 https://doi.org/10.1155/2018/5953609

[50] 

Vinoth V.; Shergilin T.D.; Asiri A.M.; Wu J.J.; Anandan S.. Facile synthesis of copper oxide microflowers for nonenzymatic glucose sensor applications. Materials Science in Semiconductor Processing 82 (2018) 31-38. https://doi.org/10.1016/j.mssp.2018.03.032 https://doi.org/10.1016/j.mssp.2018.03.032

[51] 

Dung N.Q.; Patil D.; Jung H.; Kim J.; Kim D.. NiO-decorated single-walled carbon nanotubes for high-performance nonenzymatic glucose sensing. Sensors and Actuators B: Chemical 183 (2013) 381-387. https://doi.org/10.1016/j.snb.2013.04.018 https://doi.org/10.1016/j.snb.2013.04.018

[52] 

Xiao N.; Deng J.; Cheng J.; Ju S.; Zhao H.; Xie J.; Qian D.; He J.. Carbon paste electrode modified with duplex molecularly imprinted polymer hybrid film for metronidazole detection. Biosensors and Bioelectronics 81 (2016) 54-60. http://dx.doi.org/10.1016/j.bios.2016.02.041 https://doi.org/10.1016/j.bios.2016.02.041

[53] 

Shahsavari M.; Tajik S.; Sheikhshoaie I.; Beitollahi H.. Fabrication of nanostructure electrochemical sensor based on the carbon baste electrode (CPE) modified with ionic liquid and Fe3o4/zif-67 for electrocatalytic sulfamethoxazole detection. Topics in Catalysis 65 (2021) 1-10. https://doi.org/10.1007/s11244-021-01471-8 https://doi.org/10.1007/s11244-021-01471-8

[54] 

Muthuchamy N.; Atchudan R.; Edison T.N.J.I.; Perumal S.; Lee Y.R.. High-performance glucose biosensor based on green synthesized zinc oxide nanoparticle embedded nitrogen-doped carbon sheet. Journal of Electroanalytical chemistry 816 (2018) 195-204. https://doi.org/10.1016/j.jelechem.2018.03.059 https://doi.org/10.1016/j.jelechem.2018.03.059

[55] 

Heidari A.; Kalantar E.; Dehghan M.; Fallah P.. Herpes simplex encephalitis: successful treatment with acyclovir. Archives of Advances in Biosciences 10 (2019) 56-58. https://doi.org/10.22037/jps.v10i1.19886 https://doi.org/10.22037/jps.v10i1.19886

[56] 

Newman D.J.; Cragg G.M.. Natural products as sources of new drugs from 1981 to 2014. Journal of Natural Products 79 (2016) 629-661. https://doi.org/10.1021/acs.jnatprod.5b01055 https://doi.org/10.1021/acs.jnatprod.5b01055

[57] 

Hong X.; Wang X.; Wang Z.. A rare case report of acyclovir-induced immune thrombocytopenia with tongue hematomas as the first sign, and a literature review. BioMed Central Pharmacology and Toxicology 18 (2017) 1-5. https://doi.org/10.1186/s40360-017-0120-2 https://doi.org/10.1186/s40360-017-0120-2

[58] 

Shen Z.; Yu Q.; Li Y.; Bao Y.; Lu H.. Determination of acyclovir in renal microdialysis fluid and confirmation of renal function index. Drug and Chemical Toxicology 43 (2020) 574-580. https://doi.org/10.1080/01480545.2018.1524474 https://doi.org/10.1080/01480545.2018.1524474

[59] 

Sen S.S.; Si A.; Chakraborty U.; Chandra A.. Stevens-Johnson syndrome-toxic epidermal necrolysis: a fatal cutaneous adverse reaction to oral acyclovir. British Medical Journal Case Reports 13 (2020) 8. https://doi.org/10.1136/bcr-2020-238555 https://doi.org/10.1136/bcr-2020-238555

[60] 

Chu K.; Li Q.; Cheng Y.; Liu Y.. Efficient electrocatalytic nitrogen fixation on FeMoO4 nanorods. ACS Applied Materials & Interfaces 12 (2020) 11789-11796. https://doi.org/10.1021/acsami.0c00860 https://doi.org/10.1021/acsami.0c00860

[61] 

Tang L.; Liu L.; Yang F., FeMoO4-graphene oxide photo-electro-catalyst for berberine removal and hydrogen evolution. International Journal of Hydrogen Energy 44 (2019) 19755-19761. https://doi.org/10.1016/j.ijhydene.2019.03.078 https://doi.org/10.1016/j.ijhydene.2019.03.078

[62] 

Wei Y.; Yao L.; Wu Y.; Liu X.; Feng J.; Ding J.; Li K.; He Q.. Ultrasensitive electrochemical detection for nanomolarity Acyclovir at ferrous molybdate nanorods and graphene oxide composited glassy carbon electrode. Colloids and Surfaces A: Physicochemical and Engineering Aspects 641 (2022) 128601. https://doi.org/10.1016/j.colsurfa.2022.128601 https://doi.org/10.1016/j.colsurfa.2022.128601

[63] 

Huang Y.; Lu D.; Liu H.; Liu S.; Jiang S.; Pang G.C.; Liu Y.. Preliminary research on the receptor–ligand recognition mechanism of umami by an hT1R1 biosensor. Food & function 10 (2019) 1280-1287. https://doi.org/10.1039/C8FO02522C https://doi.org/10.1039/C8FO02522C

[64] 

Venkatakrishnan A.J.; Deupi X.; Lebon G.; Tate C.G.; Schertler G.F.; Babu M.M.. Molecular signatures of Gprotein- coupled receptors. Nature 494 (2013) 185-194. https://doi.org/10.1038/nature11896 https://doi.org/10.1038/nature11896

[65] 

Wei L.; Wang X.; Lu D.; Li Y.; Pang G.; Xie J.. A novel staphylococcal enterotoxin q immunosensor prepared with self-assembly method based on horseradish peroxidase and double-layer gold nanoparticles. Food Analytical Methods 10 (2016) 892-899. https://doi.org/10.1007/s12161-016-0632-1 https://doi.org/10.1007/s12161-016-0632-1

[66] 

Lu D.; Liu D.; Liu Y.; Wang X.; Liu Y.; Yuan S.; Ren R.; Pang G.. Comparative study on the sensing kinetics of carbon and nitrogen nutrients in cancer tissues and normal tissues based electrochemical biosensors. Molecules 28 (2023) 1453. https://doi.org/10.3390/molecules28031453 https://doi.org/10.3390/molecules28031453

[67] 

Karupppaiah B.; Jeyaraman A.; Chen S.M.; Huang Y.C.. Development of highly sensitive electrochemical sensor for antibiotic drug ronidazole based on spinel cobalt oxide nanorods embedded with hexagonal boron nitride. Electrochimica Acta 446 (2023) 142008. https://doi.org/10.1016/j.electacta.2023.142008 https://doi.org/10.1016/j.electacta.2023.142008

[68] 

Pu J.; Zhang K.; Wang Z.; Li C.; Zhu K.; Yao Y.; Hong G.. Synthesis and modification of boron nitride nanomaterials for electrochemical energy storage: from theory to application. Advanced Functional Materials 31 (2021) 1-32. https://doi.org/10.1002/adfm.202106315 https://doi.org/10.1002/adfm.202106315

[69] 

García-Miranda Ferrari A.; Rowley-Neale S.J.; Banks C.E.. Recent advances in 2D hexagonal boron nitride (2D-hBN) applied as the basis of electrochemical sensing platforms. Analytical and Bioanalytical Chemistry 413 (2021) 663-672. https://doi.org/10.1007/s00216-020-03068-8 https://doi.org/10.1007/s00216-020-03068-8

[70] 

Rafiq M.; Hu X.; Ye Z.; Qayum A.; Xia H.; Hu L.; Lu F.; Chu P.K.. Recent advances in structural engineering of 2D hexagonal boron nitride electrocatalysts. Nano Energy 91 (2022) 106661. https://doi.org/10.1016/j.nanoen.2021.106661 https://doi.org/10.1016/j.nanoen.2021.106661

[71] 

Ahn H.K.; Han B.; Lee S.J.; Lim T.; Sun J.M.; Ahn J.S.; Ahn M.J.; Park K.. ALK inhibitor crizotinib combined with intrathecal methotrexate treatment for non-small cell lung cancer with leptomeningeal carcinomatosis. Lung Cancer 76 (2012) 253-254. https://doi.org/10.1016/j.lungcan.2012.02.003 https://doi.org/10.1016/j.lungcan.2012.02.003

[72] 

Cohen E.E.; Licitra L.F.; Burtness B.; Fayette J.; Gauler T.; Clement P.M.; Grau J.J.; del Campo J.M.; Mailliez A.; Haddad R.I.; Vermorken J.B.; Tahara M.; Guigay J.; Geoffrois L.; Merlano M.C.; Dupuis N.; Krämer N.; Cong X.J.; Gibson N.; Solca F.; Machiels J.P.. Biomarkers predict enhanced clinical outcomes with afatinib versus methotrexate in patients with second-line recurrent and/or metastatic head and neck cancer. Annals of Oncology 28 (2017) 2526-2532. https://doi.org/10.1093/annonc/mdx344 https://doi.org/10.1093/annonc/mdx344

[73] 

Huang D.; Wu H.; Zhu Y.; Su H.; Zhang H.; Sheng L.; Liu Z.; Xu H.; Song C.. Sensitive determination of anticancer drug methotrexate using graphite oxide-nafion modified glassy carbon electrode. International Journal of Electrochemical Science 14 (2019) 3792-3804. https://doi.org/10.20964/2019.04.03 https://doi.org/10.20964/2019.04.03

[74] 

Asadian E.; Shahrokhian S.; Zad A. I.; Ghorbani-Bidkorbeh F.. Glassy carbon electrode modified with 3D graphene–carbon nanotube network for sensitive electrochemical determination of methotrexate. Sensors and Actuators B 239 (2017) 617-627. https://doi.org/10.1016/j.snb.2016.08.064 https://doi.org/10.1016/j.snb.2016.08.064

[75] 

Luhana C.; Mashazi P.. Simultaneous detection of dopamine and paracetamol on electroreduced graphene oxide–cobalt phthalocyanine polymer nanocomposite electrode. Electrocatalysis 14 (2022) 1-12. https://doi.org/10.1007/s12678-022-00806-7 https://doi.org/10.1007/s12678-022-00806-7

[76] 

Wong A.; Santos A.M.; Cincotto F.H.; Moraes F.C.; Fatibello-Filho O.; Sotomayor M.D.. A new electrochemical platform based on low cost nanomaterials for sensitive detection of the amoxicillin antibiotic in different matrices. Talanta 206 (2020) 120252. https://doi.org/10.1016/j.talanta.2019.120252 https://doi.org/10.1016/j.talanta.2019.120252

[77] 

Pollap A.; Knihnicki P.; Kuśtrowski P.; Kozak J.; Gołda-Cępa M.; Kotarba A.; Kochana J.. Sensitive voltammetric amoxicillin sensor based on TiO2 sol modified by CMK-3-type mesoporous carbon and gold ganoparticles. Electroanalysis 30 (2018) 2386-2396. https://doi.org/10.1002/elan.201800203 https://doi.org/10.1002/elan.201800203

[78] 

Deroco P.B.; Rocha-Filho R.C.; Fatibello-Filho O.. A new and simple method for the simultaneous determination of amoxicillin and nimesulide using carbon black within a dihexadecylphosphate film as electrochemical sensor. Talanta 179 (2018) 115-123. https://doi.org/10.1016/j.talanta.2017.10.048 https://doi.org/10.1016/j.talanta.2017.10.048

[79] 

Kumar N.; Rosy, Goyal R.N.. Gold-palladium nanoparticles aided electrochemically reduced graphene oxide sensor for the simultaneous estimation of lomefloxacin and amoxicillin. Sensors and Actuators B: Chemical 243 (2017) 658-668. https://doi.org/10.1016/j.snb.2016.12.025 https://doi.org/10.1016/j.snb.2016.12.025

[80] 

Yue X.; Li Z.; Zhao S.. A new electrochemical sensor for simultaneous detection of sulfamethoxazole and trimethoprim antibiotics based on graphene and ZnO nanorods modified glassy carbon electrode. Microchemical Journal 159 (2020) 105440. https://doi.org/10.1016/j.microc.2020.105440 https://doi.org/10.1016/j.microc.2020.105440

[81] 

Golinelli D.L.C.; Machado S.A.S.; Cesarino I.. Synthesis of silver nanoparticle-graphene composites for electroanalysis applications using chemical and electrochemical methods. Electroanalysis 29 (2017) 1014-1021. https://doi.org/10.1002/elan.201600669 https://doi.org/10.1002/elan.201600669

[82] 

Man N.Q.; Tu N.T.T.; Hoan N.T.V.; Vu H.X.A.; Son L.L.; Quyen N.D.V.; Nhiem D.N.; Phong N.H.; Nguyen V.T.; Tuyen T.N.; Khieu D.Q.. Electrochemical determination of clenbuterol with nickel-ferrite/reduced-graphene-oxide-modified electrode. Journal of Nanoparticle Research 25 (2023). https://doi.org/10.1007/s11051-023-05679-5 https://doi.org/10.1007/s11051-023-05679-5

[83] 

Wu S.; Dai X.; Cheng T.; Li S.. Highly sensitive and selective ion-imprinted polymers based on one-step electrodeposition of chitosan-graphene nanocomposites for the determination of Cr(VI). Carbohydrate Polymers 195 (2018) 199-206. https://doi.org/10.1016/j.carbpol.2018.04.077 https://doi.org/10.1016/j.carbpol.2018.04.077

[84] 

Chen L.; Xu S.; Li J.. Recent advances inmolecular imprinting technology: current status, challenges and highlighted applications. Chemical Society Reviews 40 (2011) 2922-2942. https://doi.org/10.1039/C0CS00084A https://doi.org/10.1039/C0CS00084A

[85] 

Wu Y.; Deng P.; Tian Y.. Rapid recognition and determination of tryptophan by carbon nanotubes and molecularly imprinted polymer modified glassy carbonelectrode. Bioelectrochemistry 19 (2020) 107493. https://doi.org/10.1016/j.bioelechem.2019.107393 https://doi.org/10.1016/j.bioelechem.2019.107393

[86] 

He Q.; Liu J.; Liang J.; Liu X.; Li W.; Liu Z.; Ding Z.; Tuo D.. Towards improvements for penetrating the blood–brain barrier—recent progress from a material and pharmaceuticalperspective. Cells 7 (2018) 24. https://doi.org/10.3390/cells7040024 https://doi.org/10.3390/cells7040024

[87] 

Duan D.; Yang H.; Ding Y.; Li L.; Ma G.. A three-dimensional conductive molecularly imprinted electrochemical sensor based on MOF derived porous carbon/carbon nanotubes composites and prussian blue nanocubesmediated amplification for chiral analysis of cysteine enantiomers. Electrochimica Acta 302 (2019) 137-144. https://doi.org/10.1016/j.electacta.2019.02.028 https://doi.org/10.1016/j.electacta.2019.02.028

[88] 

Wei H.; Wang Y.; Guo J.; Qiu B.; Ding D.; Wei S.; Wujcik E.K.; Guo Z.. Synthesis of multifunctional carbon nanostructures. Handbook of Carbon Nano Materials 7 (2015) 89. https://doi.org/10.1142/9789814678919_0003 https://doi.org/10.1142/9789814678919_0003

[89] 

Kooshki M.; Abdollahi H.; Bozorgzadeh S.; Haghighi B.. Second-order data obtained from differential pulse voltammetry: determination of tryptophan at a gold nanoparticles decorated multiwalled carbon nanotube modified glassy carbon electrode. Electrochimica Acta 56 (2011) 8618-8624. https://doi.org/10.1016/j.electacta.2011.07.049 https://doi.org/10.1016/j.electacta.2011.07.049

[90] 

Qiu L.; Zhou G.; Cao S.. Targeted inhibition of ULK1 enhances daunorubicin sensitivity in acute myeloid leukemia. Life Sciences 243 (2020) 117234. https://doi.org/10.1016/j.lfs.2019.117234 https://doi.org/10.1016/j.lfs.2019.117234

[91] 

Nong Q.; Zhang C.; Liu Q.; Xie R.; Dong M.. Effect of daunorubicin on acute promyelocytic leukemia cells using nuclear magnetic resonance spectroscopy-based metabolomics. Environmental Toxicology and Pharmacology 78 (2020) 103382. https://doi.org/10.1016/j.etap.2020.103382 https://doi.org/10.1016/j.etap.2020.103382

[92] 

Alizadeh M.; Orooji Y.; Karimi F.; Baghayeri M.; Rouhi J.; Tajik S.; Beitollahi H.; Agarwal S.; Gupta V.K.; Rajendran S.; Rostamnia S.; Fu L.; Movahed F.S.; Malekmohammadi S.. Guanine-based DNA biosensor amplified with Pt/SWCNTs nanocomposite as analytical tool for nanomolar determination of daunorubicin as an anticancer drug: a docking/experimental investigation. Industrial & Engineering Chemistry Research 60 (2021) 816-823. https://doi.org/10.1021/acs.iecr.0c04698 https://doi.org/10.1021/acs.iecr.0c04698

[93] 

Bhengo T.; Moyo M.; Shumba M.; Okankwo O.J.. Simultaneous oxidative determination of antibacterial drugs in aqueous solutions using an electrode modified with MWCNTs decorated with Fe3O4 nanoparticles. New Journal of Chemistry 42 (2018) 5014-5023. https://doi.org/10.1039/C8NJ00129D https://doi.org/10.1039/C8NJ00129D

[94] 

Chokkareddy R.; Kanchi S.; Redhi G.G.. A novel IL-f-ZnONPs@MWCNTs nanocomposite fabricated glassy carbon electrode for the determination of sulfamethoxazole. Journal of Molecular Liquids 359 (2022) 119232. https://doi.org/10.1016/j.molliq.2022.119232 https://doi.org/10.1016/j.molliq.2022.119232

[95] 

Yari A.; Shams A.. Silver-filled MWCNT nanocomposite as a sensing element for voltammetric determination of sulfamethoxazole. Analytica chimica acta 1039 (2018) 51-58. https://doi.org/10.1016/j.aca.2018.07.061 https://doi.org/10.1016/j.aca.2018.07.061

[96] 

Murugan N.; Jerome R.; Preethika M.; Sundaramurthy A.; Sundramoorthy A.K.. 2D-titanium carbide (MXene) based selective electrochemical sensor for simultaneous detection of ascorbic acid, dopamine and uric acid. Journal of Materials Science & Technology 72 (2021) 122-131. https://doi.org/10.1016/j.jmst.2020.07.037 https://doi.org/10.1016/j.jmst.2020.07.037

[97] 

Wu Y.; Deng P.; Tian Y.; Feng J.; Xiao J.; Li J.; Liu J.; Li G.; He Q.. Simultaneous and sensitive determination of ascorbic acid, dopamine and uric acid via an electrochemical sensor based on PVP-graphene composite. Journal of Nanobiotechnology 18 (2020) 112. https://doi.org/10.1186/s12951-020-00672-9 https://doi.org/10.1186/s12951-020-00672-9

[98] 

Zhang X.; Zhang Y.C.; Ma L.X.. One-pot facile fabrication of graphene-zinc oxide composite and its enhanced sensitivity for simultaneous electrochemical detection of ascorbic acid, dopamine and uric acid. Sensors and Actuators B: Chemical 227 (2016) 488-496. https://doi.org/10.1016/J.SNB.2015.12.073 https://doi.org/10.1016/J.SNB.2015.12.073

[99] 

Yang M.; Guo M.; Feng Y.; Lei Y.; Cao Y.; Zhu D.; Yu Y.; Ding L.. Sensitive voltammetric detection of metronidazole based on three-dimensional graphene-like carbon architecture/polythionine modified glassy carbon electrode. Journal of The Electrochemical Society 165 (2018) B530. https://doi.org/10.1149/2.1311811jes https://doi.org/10.1149/2.1311811jes

[100] 

Tursynbolat S.; Bakytkarim Y.; Huang J.; Wang L.. Ultrasensitive electrochemical determination of metronidazole based on polydopamine/carboxylic multiwalled carbon nanotubes nanocomposites modified GCE. Journal of Pharmaceutical Analysis 8 (2018) 124-130. https://doi.org/10.1016/j.jpha.2017.11.001 https://doi.org/10.1016/j.jpha.2017.11.001

[101] 

Yari A.; Shams A.. Silver-filled MWCNT nanocomposite as a sensing element for voltammetric determination of sulfamethoxazole. Analytica Chimica Acta 1039 (2018) 51-58. https://doi.org/10.1016/j.aca.2018.07.061 https://doi.org/10.1016/j.aca.2018.07.061

[102] 

Zhang C.; Ma Y.; Zhang X.; Abdolhosseinzadeh S.; Sheng H.; Lan W.; Pakdel A.; Heier J.; Nüesch F.. Two-dimensional transition metal carbides and nitrides (MXenes): synthesis, properties, and electrochemical energy storage applications. Energy & Environmental Materials 3 (2020) 29-55. https://doi.org/10.1002/eem2.12058 https://doi.org/10.1002/eem2.12058

[103] 

Halim J.; Kota S.; Lukatskaya M.R.; Naguib M.; Zhao M.Q.; Moon E.J.; Pitock J.; Nanda J.; May S.J.; Gogotsi Y.; Barsoum M.W.. Synthesis and characterization of 2D molybdenum carbide (MXene). Advanced Functional Materials 26 (2016) 3118-3127. https://doi.org/10.1002/adfm.201505328 https://doi.org/10.1002/adfm.201505328

[104] 

Zhou F.; Jing W.; Liu S.; Mao Q.; Xu Y.; Han F.; Wei Z.; Jiang Z.. Electrodeposition of gold nanoparticles on ZnO nanorods for improved performance of enzymatic glucose sensors. Materials Science in Semiconductor Processing 105 (2020) 104708. https://doi.org/10.1016/j.mssp.2019.104708 https://doi.org/10.1016/j.mssp.2019.104708

[105] 

Granito A.; Forgione A.; Marinelli S.; Renzulli M.; Lelasi L.; Sansone V.; Benevento F.; Piscaglia F.; Tovoli F.. Regorafenib combined with other systemic therapies: exploring promising therapeutic combinations in HCC. Journal of Hepatocellular Carcinoma 8 (2021) 477-492. https://doi.org/10.2147/JHC.S251729 https://doi.org/10.2147/JHC.S251729

[106] 

Merzak D.; Kaya S.I.; Ahmet C.; Nurgul K.B.; Mohamed T.; Sibel A.O.. Detailed electrochemical behavior and thermodynamic parameters of anticancer drug regorafenib and its sensitive electroanalytical assay in biological and pharmaceutical samples. Microchemical Journal 170 (2021) 106717. https://doi.org/10.1016/j.microc.2021.106717 https://doi.org/10.1016/j.microc.2021.106717

[107] 

Kim H.; Bang Y.; Lee M.A.; Kim J.W.; Jee H.K.; Chon H.K.; Kang B.; Kang M.J.; Kim I.; Cheon J.; Hwang J.E.; Kang J.H.; Byeon S.; Hong J.Y.; Ryoo B.Y.; Lim B.Y.; Yoo C.. Regorafenib in patients with advanced Child-Pugh B hepatocellular carcinoma: a multicentre retrospective study. Liver International 40 (2020) 2544-2552. https://doi.org/10.1111/liv.14573 https://doi.org/10.1111/liv.14573

[108] 

Sravani B.; Kiranmai S.; Reddy G.R.; Park J.P.; Reddy Y. V.; Madhavi G.. Highly sensitive detection of anticancer drug based on bimetallic reduced graphene oxide nanocomposite. Chemosphere 287 (2022) 132281. https://doi.org/10.1016/j.chemosphere.2021.132281 https://doi.org/10.1016/j.chemosphere.2021.132281

[109] 

Laviron E.. General expression of the linear potential sweep voltammogram in the case of diffusionless electrochemical systems. Journal of Electroanalytical Chemistry and Interfacial Electrochemistry 101 (1979) 19-28. https://doi.org/10.1016/S0022-0728(79)80075-3 https://doi.org/10.1016/S0022-0728(79)80075-3

[110] 

Naghian E.; Shahdostfard F.; Sohouli E.; Safarifard V.; Najafi M.; Rahimi-Nasrabadi M.; sobhani-nasab A.. Electrochemical determination of levodopa on a reduced graphene oxide paste electrode modified with a metal-organic framework. Microchemical Journal 156 (2020) 104888. https://doi.org/10.1016/j.microc.2020.104888 https://doi.org/10.1016/j.microc.2020.104888

[111] 

Vasu D.; Karthi Keyan A.; Sakthinathan S.; Chiu T.W.. Investigation of electrocatalytic and photocatalytic ability of Cu/Ni/TiO2/MWCNTs Nanocomposites for detection and degradation of antibiotic drug Furaltadone. Scientific Reports 12 (2022) 1-16. https://doi.org/10.1038/s41598-022-04890-z https://doi.org/10.1038/s41598-022-04890-z

[112] 

Veseli A.; Mullallari F.; Balidemaj F.; Berisha L.; Švorc Ľ.; Arbneshi T.. Electrochemical determination of erythromycin in drinking water resources by surface modified screen-printed carbon electrodes. Microchemical Journal 148 (2019) 412-418. https://doi.org/10.1016/j.microc.2019.04.086 https://doi.org/10.1016/j.microc.2019.04.086

[113] 

Materón E.M.; Wong A.; Freitas T.A.; Faria R.C.; Oliveira O.N.. A sensitive electrochemical detection of metronidazole in synthetic serum and urine samples using low-cost screen-printed electrodes modified with reduced graphene oxide and C60. Journal of Pharmaceutical Analysis 11 (2021) 646-652. https://doi.org/10.1016/j.jpha.2021.03.004 https://doi.org/10.1016/j.jpha.2021.03.004

[114] 

Kurbanoglu S.; Ozkan S.A.. Electrochemical carbon based nanosensors : a promising tool in pharmaceutical and biomedical analysis. Journal of Pharmaceutical and Biomedical Analysis 147 (2018) 439-457. https://doi.org/10.1016/j.jpba.2017.06.062 https://doi.org/10.1016/j.jpba.2017.06.062

[115] 

Yáñez-Sedeño P.; Campuzano S.; Pingarrón J.M.. Fullerenes in electrochemical catalytic and affinity biosensing. C - Journal of Carbon Research 3 (2017) 21. https://doi.org/10.3390/c3030021 https://doi.org/10.3390/c3030021

[116] 

Carvalho J.H.; Gogola J.L.; Bergamini M.F.; Marcolino-Junior L.H.; Janegitz B.C.. Disposable and low-cost lab-made screen-printed electrodes for voltammetric determination of L-dopa. Sensors and Actuators Reports 3 (2021) 100056. https://doi.org/10.1016/j.snr.2021.100056 https://doi.org/10.1016/j.snr.2021.100056

[117] 

dos Santos S.M.V.; De Oliveira P.R.; De Oliveira M.C.; Bergamini M.F.; Marcolino L.H.. Eletrodos impressos construídos por serigrafia utilizando negro de fumo como material conductor. Revista Virtual de Quimica 9 (2017) 626-640. https://doi.org/10.21577/1984-6835.20170037 https://doi.org/10.21577/1984-6835.20170037

[118] 

Menon H.; Aiswarya R.; Surendran K.P.. Screen printable MWCNT inks for printed electronics. Royal Society of Chemistry Advances 7 (2017) 44076-44081. https://doi.org/10.1039/c7ra06260e https://doi.org/10.1039/c7ra06260e

[119] 

Gevaerd A.; Watanabe E.Y.; Belli C.; Marcolino-Junior L.H.; Bergamini M.F.. A complete lab-made point of care device for non-immunological electrochemical determination of cortisol levels in salivary samples. Sensors and Actuators B: Chemical 332 (2021) 129532. https://doi.org/10.1016/j.snb.2021.129532 https://doi.org/10.1016/j.snb.2021.129532

[120] 

Rungsawang T.; Punrat E.; Adkins J.; Henry C.; Chailapakul O.. Development of electrochemical paper-based glucose sensor using cellulose-4-aminophenylboronic acid-modified screen-printed carbon electrode. Electroanalysis 28 (2016) 462-468. https://doi.org/10.1002/elan.201500406 https://doi.org/10.1002/elan.201500406

[121] 

Magar H.S.; Hassan R.Y.A.; Abbas M.N.. Non-enzymatic disposable electrochemical sensors based on CuO/Co3O4@MWCNTs nanocomposite modified screen-printed electrode for the direct determination of urea. Scientific Reports 13 (2023) 2034. https://doi.org/10.1038/s41598-023-28930-4 https://doi.org/10.1038/s41598-023-28930-4

[122] 

Wang Y.; Xie J.; Tao L.; Tian H.; Wang S.; Ding H.. Simultaneous electrochemical determination of epirubicin and methotrexate in human blood using a disposable electrode modified with nano-Au/MWNTs-ZnO composites. Sensors and Actuators B 204 (2014) 360-367. https://doi.org/10.1016/j.snb.2014.07.099 https://doi.org/10.1016/j.snb.2014.07.099

[123] 

Motsaathebe P.C.; Fayemi O.E.. Electrochemical detection of ascorbic acid in oranges at mwcnt aonp nanocompositefabricated electrode. Nanomaterials 12 (2022) 645. https://doi.org/10.3390/nano12040645 https://doi.org/10.3390/nano12040645

[124] 

Uwaya G.; Fayemi O.E.. Electrochemical detection of ascorbic acid in orange on iron(iii) oxide nanoparticles modified screen printed carbon electrode. Journal of Cluster Science 33 (2022) 1035-1043. https://doi.org/10.1007/s10876-021-02030-7 https://doi.org/10.1007/s10876-021-02030-7

Floating objects

Figure 1. Schematic of A) EBT/CPE preparation, B) polymerisation steps of modified CPE, C) proposed mechanism of eriochrome black T (EBT) polymerisation and D) electrochemical detection of MDH by EBT/CPE. Carried out at the base of the reference [26] with an order License ID of 1346497-1
ADMET-11-1709-g001.jpg
Figure 2. Schematic of the multilayer modification of the GCE to study the electrochemical response of the human umami taste receptor hT1R1 towards umami substances such as sodium glutamate, disodium inosinate and disodium guanylate. Carried out at the base of the reference [63] with an order License ID of 1346500-1
ADMET-11-1709-g002.jpg
Figure 3. Schematic of the multilayer modification of GCE to study the detection kinetics of C and N nutrient receptors. Based on the reference [66]
ADMET-11-1709-g003.jpg
Figure 4. Schematic of MIP-MWCNT/GCE preparation and electrochemical detection of Tryptophan. Based on the reference [85]
ADMET-11-1709-g004.jpg
Figure 5. Schematic of CuO/Co3O4@MWCNTs preparation and electrochemical detection of urea. Based on the reference [121]
ADMET-11-1709-g005.jpg
Table 1. Evaluation of electrochemical properties of modified CPE
ElectrodeAnalyte and matrixMethodLinear dynamic rangeLODYear / Ref
poly-EBT/CPEMDH in Dilosyn syrup and human urineSWV0.1-50 μM0.0257 μM2020 / [26]
CPE/nanozeolite type XParacetamol and epinineLSV0.5-70.0 μMPara = 0.2 μM
Epi = 0.008 μM
2023 / [30]
Ce-BTC MOF/IL/CPEKTC in pharmaceutical and urine samplesChrono-amperometry; DPV; CV; LSV0.1-110.0 μM0.04 μM
S = 0.1342 μA μM-1
2023 / [31]
AgNPs@CPEMTZ in milk and tap water-1-1000 μM0.206 μM2022 / [33]
[10%FG/5%MW] CPEOfloxacin in pharmaceutical tablets human urine samplesSW-AdAS0.60 to 15.0 nM0.18 nM2019 / [41]
MIP/CP ECL sensorAzithromycin urine and serum samplesECL0.10-400 nM0.023 nM2018 / [45]
CPEA/TiO2/UVAA in pharmaceutical tabletsCV150-850 nM0.732 μM2023 / [48]
CuO MFs/Nafion/GCGlucose in real serum sampleAmperometric10-120 μM6.48 μM
S=3.1 μA μM-1 cm-2
2018 / [50]
DMIP/CPEMTZ in Human Serum, Urine and drugDPV0.4-200 μM91 nM2016 / [52]
Fe3O4/ZIF-67 /ILCPESMZ in urine and waterDPV0.01-520.0 M5.0 nM2021 / [53]

[i] poly-EBT: poly-eriochrome black T; Ce: cerium; BTC: benzene tricarboxylic acid; MOF : metal organic framework; Il : ionic liquids; AgNPs: silver nanoparticles; FG: Flake graphite; MW: multi-wall carbon nanotubes; MIP: molecularly imprinted polymer; ECL: electrochemiluminescence; CuO MFs: CuO micro flowers; DMIP: duplex molecularly imprinted polymer; ZIF-67: zeolitic imidazolate framework ; MDH: methdilazine hydrochloride; KTC: ketoconazole ; AA: ascorbic acid; MTZ: metronidazole; SMZ: sulfamethoxazole; S: sensitivity

Table 2. Evaluation of electrochemical properties of modified GCE
ElectrodeAnalyte and matrixMethodLinear dynamic rangeLODYear / Ref
GCE/ZnO@NDCS/GOxGlucose in human blood serumCV; DPV;CAM0.2 and 12 mM6.3 μM
S = 231.7 μA mM-1 cm-2
2018 / [54]
FeMoO4-GO/GCEAcyclovir in drug samplesLSV0.1-10 μM
10-100 μM
20 nM2022 / [62]
CoCo2O4 NRs/h-BN/GCERonidazole water samplesDPV0.01-1345 μM3 nM
S = 5.845 μA μM-1 cm-2
2023 / [67]
GO-Nafion-GCEMTX in methotrexae injection an uine samplesCV0.4-20 μM9 nM2019 / [73]
GCE-ERGO/polyCoTAPcDA and Paracetamol in synthetic urineDPV-0.10 μM for PA
0.095 μM for DA
SPA=1.32 μA μM-1 cm-2
SDA= 8.39 μA μM-1 cm-2
2022 / [75]
QDs-P6LC-PEDOT:PSS/GCEAX in synthetic urine, whole milk and pharmaceuticalsSWV0.90 to 69 μM50 nM2020 / [76]
TiO2/CMK-3/AuNPs/Nafion/GCEAX in pharmaceutical product, mineral and environmental waterCVLDR1= 0.5-2.5 μM
LDR2 = 2.5-133 μM
0.3 μM
S1 = 5071 μA mM−1 cm−2
S2 = 2971 μA mM−1 cm−2
2018 / [77]
CB/DPH/GCEAX inbiological urine, lake and tap waterSWV2.0-18.8μM0.12 μM2018 / [78]
NIM inbiological urine, lake and tap waterSWV0.30-5.0 μM0.016 μM
AuNPs/PdNPs/ErGO/GCEAX inhuman urine samplesSWV30.0-350.0μM9.0 μM
S = 0.0376 μA μM-1
2017 / [79]
LMF human urine samplesSWV4-500 μM81 nM
S = 0.0759 μA μM-1
GR-ZnO/GCESMZ and TMP in urine and human serumDPVLRDSMZ= 1-220 μM
LRDTMP = 1-180 μM
LODSMZ = 0.4 μM
LRDTMP = 0.3 μM
2020 / [80]
GC/rGO-AgNPSMZ and TMP in wastewaters samplesDPV1.0-10.0 mMLODSMZ = 0.6 μM
LRDTMP = 0.4 μM
2017 / [81]
MIP-MWCNTs/GCETrp in the human serum samples.CV, SDLSVs2.0 nM-0.2 μM
0.2 -10 μM
10-100 μM
1.0 nM2020 / [85]
ds-DNA/Pt/SWCNTs/GCEDRN in daunorubicin injectionDVP4.0 nM to 250.0 μM1.0 nM2021 / [92]
IL-f-ZnONPs@MWCNTs@GCESMZ
pharmaceutical urine samples
CV0.1-10 ng ml-10.1 ng ml-12022 / [94]
Ag-MWCNT/MTOAC/GCESMZ in pharmaceutical formulationsand human urineDPV0.05-70 μM0.01 μM2018 / [95]
Ti-C-Tx/GCEAA in human urine samplesCV, DPV100-1000μM4.6 μM2021 / [96]
DA in human urine samplesCV, DPV0.5-50 μM0.06 μM
UA in human urine samplesCV, DPV0.5-4 μM
100-1500 μM
0.075 μM
PVP-GR/GCEAA in human urine samplesLSV4-1000 μM0.80μM2020 / [97]
DA in human urine samplesLSV0.02-100 μM0.002 μM
UA in human urine samplesLSV0.04-100 μM0.02 μM
rGO-ZnO/GCEAAin real plasma and urine samplesDPV50-2350μM3.71μM2016 / [98]
UA in real plasma and urine samplesDPV1-70 μM0.33 μM
DA in real plasma and urine samplesDPV3-330 μM1.08 μM
3D-HPG/PTH/GCEMTZ in pharmaceutical and real wate samplesCV; DPV0.05-70 μM
70−500 μM
1 nM2018 / [99]
Polydopamine/MWCNTseCOOH nanocomposites/GCEMTZ in pharmaceutical and biological samplesDPV5-5000 μM0.25 μM2018/ [100]
Ag-MWCNT/MTOAC/GCESMZ in pharmaceutical formulations and humanurineDPV0.05-70 μM0.01 μM2018 / [101]

[i] NDCS: nitrogen-doped carbon sheets; GOx: glucose oxidase; FeMoO4-GO: ferrous molybdate-graphene oxide; CoCo2O4 NRs: spinel cobalt oxide nanorods; h-BN: hexagonal boron nitride; ERGO: electrochemically reduced graphene oxide; polyCoTAPc: polymer of cobalt (II) tetra-amino phthalocyanine; QDs: cadmium telluride quantum dots; P6LC: Printex 6L Carbon; PEDOT: poly(3,4-ethylenedioxythiophene; PSS: polystyrene sulphonate film; CMK-3: mesoporous carbon CMK-3, AuNPs: gold nanoparticles; DPH: dihexadecyl hydrogen phosphate; CB: carbon black; PdNPs: palladium nanoparticles; GR: graphene; MIP: molecularly imprinted polymer; ds-DNA: guanine-based; IL-f-ZnONPs: ionic liquid functionalised zinc oxide nanoparticles; MTOAC: methyltrioctyl ammonium chloride; Ti-C-Tx: titanium carbide (MXene) nanosheets; PVP: polyvinylpyrrolidone; rGO: Reduced graphene; 3D: three-dimensional; HPG: graphene-like carbon architecture; PTH: polythionine; MWCNTseCOOH: carboxylic MWCNT; RNZ: ronidazole; MTX: methotrexate; DA: dopamine; AX: Amoxicillin, NIM: nimesulide; LMF: lomefloxacin; SMZ: sulfamethoxazole; TMP: trimethoprim; Trp: tryptophan; DNR: daunorubicin, AA: acide ascoorbic; DA: dopamine; UA: uric acid; MTZ: metronidazole; S: sensitivity; CAM: chronoamperometry.

Table 3. Evaluation of electrochemical properties of modified rGO
ElectrodeAnalyte and matrixMethodLinear dynamic rangeLODOther characteristicsYear/Ref.
Nafion/GOx/Au-ZnO/rGO/ITOglucose in bloodAmperometric0-9.5 mM0.2 μMS = 10.93 μA mM-1 cm-22020 / [104]
Pd-Ru/rGORegorafenib in human blood and plasma pharmaceutical formulationDPV0.5-300 nM1.6 nMkS = 2.29 s-12022 / [108]
rGO/TMU-22 MOFLevodopa in human urine and tablet samplesCV; SWV0.1-85 μM
0.1-85 μM
25 nMS = 0.58 μA μM−1
kS = 7.7 s−1
2020 / [110]

[i] AuNPs : gold nanoparticles ; GOx: glucose oxidase; rGO: reduced graphene oxide;ITO: indium tin oxide ; TMU-22 MOF: metal-organic framework; Pd-Ru: palladium/ruthenium nanoparticles; S: sensitivity

Table 4. Evaluation of electrochemical properties of modified SPE
ElectrodeAnalyte and matrixMethodLinear dynamic rangeLOD / Other characteristicsYear / Ref.
Cu/Ni/TiO2/MWCNTs/SPCEFLT in waterCV; DPV10-150 μM0.0949 μM
S = 1.9288 μA μM-1 cm-2
2022 / [111]
SPCEs/SDSERY in waterAmperometric1-15 mg l-10.19 μM2019 / [112]
C60-rGO-NF/SPEMTZ in serum and urine samplesSWV0.25-34.0 μM0.21 μM2021 / [113]
GP-CAc/PVCLevodopa in two commercial drugsSWV8-100 μM0.06 μM2021 / [116]
CuO/Co3O4@MWCNTs/SPEUreaEIS10-12-10-2 M0.223 pM2023 / [121]
Nano-Au/MWNTs-ZnO/SPEMTX in whole blood samples and pharmaceuticalSWV0.02-1.00 μM10 nM2014 / [122]
EPI in whole blood samples and pharmaceuticalSWV0.005-0.2 μM2.5 nM
MWCNT-AONP/SPCEAA in fresh orangesSWV160-640 nM140 nM2022 / [123]
SPCE/CV-Fe3O4NPAASWV10-100 μM15.7μM2021 / [124]

[i] SDS: sodium dodecyl sulfate ; NF: Nafion; C60: fullerene ; CAc: cellulose acetate; GP: graphite powder; PVC: polyvinyl chloride; AONP: antimony oxide nanoparticle; CV: Callistemon viminalis; Fe3O4NP: Iron(III) nanoparticle; FLT: furaltadone; ERY:erythromycin; MTZ:metronidazole ; MTX : methotrexate; EPI : epirubicin; AA: ascorbic acid.


This display is generated from NISO JATS XML with jats-html.xsl. The XSLT engine is libxslt.