Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter September 1, 2016

Organic solvents in electromembrane extraction: recent insights

  • Chuixiu Huang

    Chuixiu Huang received her BS in Chemical Engineering from Huazhong University of Science and Technology (China) in 2002, and she received her MS in Analytical Chemistry from Lund University (Sweden) in 2012. She received her PhD in Pharmaceutical Chemistry from University of Oslo (Norway) in 2016 with focus on the development and applications of electromembrane extraction (EME) for bioanalysis. She has previously published a few articles on EME. Currently, she is a postdoctoral student at the University of Oslo with focus on the development and discovery of new liquid membranes for EME.

    EMAIL logo
    , Astrid Gjelstad

    Astrid Gjelstad is currently an associate professor at the Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo. She received her PhD degree in 2009 and performed her doctoral training also at School of Pharmacy, University of Oslo. The academic field of interest is development of new microextraction technologies for rapid and selective isolation of drugs and peptides from small amounts of biological matrices. The microextraction technology is based on artificial liquid membranes where studies of the transport across the membranes forced by either passive diffusion (liquid-phase microextraction, LPME) or electrokinetic migration (electromembrane extraction, EME) are special focus areas. Another research focus comprises new inventions in the area of dried blood spots (DBS).

    and Stig Pedersen-Bjergaard

    Stig Pedersen-Bjergaard (SPB) was born in Copenhagen in 1966. In 1996, SPB was appointed as Associate Professor at the School of Pharmacy, University of Oslo (Oslo, Norway) and, from year 2000, as full Professor at the same institution. In 2008, SPB was appointed as full Professor (part time) at Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen (Copenhagen, Denmark). SPB has specialized in analytical micro extraction technologies on the development and applications of artificial liquid membranes and on electro-kinetic separation methods. SPB has published more than 155 papers in international journals, given more than 85 presentations at international symposia, contributed to 7 patent applications, and authored two international student textbooks in pharmaceutical analysis. SPB is Contributing Editor for Trends in Analytical Chemistry and is a member of the Editorial board for Journal of Chromatography A, Analytica Chimica Acta, Journal of Pharmaceutical Analysis, and International Journal of Analytical Chemistry.

Abstract

Electromembrane extraction (EME) was invented in 2006 as a miniaturized sample preparation technique for the separation of ionized species from aqueous samples. This concept has been investigated in different areas of analytical chemistry by different research groups worldwide since the introduction. Under the influence of an electrical field, EME is based on electrokinetic migration of the analytes through a supported liquid membrane (SLM), which is an organic solvent immobilized in the pores of the polymeric membrane, and into the acceptor solution. Up to date, close to 150 research articles with focus on EME have been published. The current review summarizes the performance of EME with different organic solvents and discusses several criteria for efficient solvents in EME. In addition, the authors highlight their personal perspective about the most promising organic solvents for EME and have indicated that more fundamental work is required to investigate and discover new organic solvents for EME.

Introduction

Prior to analysis of biological and environmental samples by chromatography and mass spectrometry, sample preparation is usually required. The purpose of the sample preparation is to make the samples compatible with the analytical instruments. During sample preparation, the complexity of the sample is reduced by removal of major sample matrix components. This is important to avoid contamination of the instruments and to avoid interferences during analyte detection. Finally, analytes are often pre-concentrated during sample preparation to enable trace level detection.

Sample preparation is traditionally performed by liquid-liquid extraction (LLE) or solid-phase extraction (SPE). Over the last two decades, however, substantial efforts have been reported to develop different microextraction techniques as alternatives to LLE and SPE, including solid-phase microextraction (SPME) (Arthur and Pawliszyn 1990), stir bar sorptive extraction (SBSE) (Baltussen et al. 1999), single drop microextraction (SDME) (Jeannot and Cantwell 1996), hollow-fiber liquid-phase microextraction (HF-LPME) (Pedersen-Bjergaard and Rasmussen 1999), dispersive liquid-liquid microextraction (DLLME) (Rezaee et al. 2006), solidified floating organic drop microextraction (SFODME) (Dadfarnia et al. 2008), parallel artificial liquid membrane extraction (PALME) (Gjelstad et al. 2013), and electromembrane extraction (EME) (Pedersen-Bjergaard and Rasmussen 2006). Microextraction techniques are interesting for several reasons. First, the consumption of hazardous organic solvents is strongly reduced or even eliminated by using microextraction techniques. This represents a major saving, and laboratory personnel are no longer exposed to vapors of harmful compounds. Second, using microextraction techniques, sample evaporation and reconstitution are avoided. This represents a major improvement in terms of work-flow simplicity, duration, risk for analyte loss, and risk for sample contamination. Third, several microextraction techniques are readily automated, and this is an important issue in modern high-throughput laboratories. Finally, the extraction efficiency can be tuned with several microextraction techniques. Thus, both exhaustive extraction and soft extraction may be accomplished. Soft extraction is highly interesting in cases where equilibria between target analytes and the sample matrix are not to be disturbed.

EME is one of the recently developed microextraction techniques (Pedersen-Bjergaard and Rasmussen 2006). In EME, target analytes are extracted in their ionic form from an aqueous sample solution through a water-immiscible organic solvent immobilized as a thin liquid membrane (supported liquid membrane, SLM) in the pores of a polymeric membrane and into an aqueous acceptor solution. The driving force for the extraction is an electrical field sustained across the SLM. Currently, approximately 150 research papers have been published on EME, covering fundamental aspects about mass transfer, technical configurations, and potential future applications. Also, several reviews on EME have been published in recent years (Lee et al. 2008, Gjelstad 2010, Majors 2010, Gjelstad and Pedersen-Bjergaard 2011, Petersen et al. 2011a,b, Basheer et al. 2012, Gjelstad and Pedersen-Bjergaard 2013, Krishna Marothu et al. 2013, Gjelstad and Pedersen-Bjergaard 2014, Rezazadeh et al. 2014a,b,c, Soltani and Jouyban 2014, Carasek and Merib 2015, Gjelstad et al. 2015, Huang et al. 2015a,b,c,d,e,f, Seip et al. 2015, Ocaña-González et al. 2016). In the current review, which is not an exhaustive review on EME, we focus especially on the different organic solvents that have been used as the SLM in EME before 2016. This is highly important because the chemical composition of the SLM plays a major role for the analyte mass transfer and the selectivity in EME. Overview and understanding of this are highly important for future development and optimization of EME and for the long-term propagation of the EME principle into routine analytical laboratories.

EME principle

A typical set-up of EME is illustrated in Figure 1. The EME equipment comprises a sample solution located in a sample reservoir (typically a glass vial), an SLM located in the pores in the wall of a porous hollow fiber, an acceptor solution located in the lumen of the hollow fiber, two electrodes (anode and cathode) connected to an external power supply, and an agitation system to agitate the entire set-up. During EME, the target analyte is extracted in the fully ionized state through the SLM and into the acceptor solution under the influence of the electrical field. After EME has been completed, the acceptor solution is collected and principally can be analyzed by any analytical techniques, typically by high-performance liquid chromatography (HPLC-UV) or liquid chromatography-mass spectrometry (LC-MS).

Figure 1: Schematic illustration of electromembrane extraction (EME) with hollow fiber membrane.
Figure 1:

Schematic illustration of electromembrane extraction (EME) with hollow fiber membrane.

The pH value in the sample is adjusted (if necessary) to ensure full ionization of the target analytes. This is important to ensure efficient electrokinetic migration in the system. The SLM volume is typically 10–15 μl of 2-nitrophenyl octyl ether (NPOE) or another water-immiscible organic solvent immobilized in the pores in the wall of the porous hollow fiber. The SLM is easily immobilized by dipping the dry and porous hollow fiber in the solvent for a few seconds, and the SLM is immediately immobilized by capillary forces. One electrode is inserted in the sample solution, while the second electrode is situated in the acceptor solution. For extraction of cationic species, the cathode (–) is located in the acceptor solution, whereas the direction of the electrical field is reversed for extraction of anionic species. Thus, the selectivity of the EME system can be determined by the direction of the electrical field. In addition, research has demonstrated that the selectivity in EME also is dependent on the magnitude of the electrical field, and this can easily be tuned by the external power supply (Domínguez et al. 2012). Finally, the selectivity and the efficiency of the EME system are highly dependent on the chemical properties of the SLM. The latter will be reviewed and discussed in detail below.

In addition, EME can be performed in a variety of technical configurations and formats. Thus, in 2009, drop-to-drop EME was designed to extract basic drugs from a drop of stagnant sample (10 μl) into a drop of stagnant acceptor phase (10 μl) with an SLM of NPOE using a porous polymeric polypropylene flat membrane (Petersen et al. 2009). Based on this research, on-chip EME was developed to extract basic drugs from dynamic samples into stagnant (Petersen et al. 2010) or dynamic (Petersen et al. 2011a,b, Petersen et al. 2012) acceptor solutions. Recently, three-hollow fiber EME was designed for exhaustive extraction of hydrophobic basic drugs using larger acceptor solution volume (Eibak et al. 2012a,b). However, using the three-hollow fiber arrangement in EME was more complex than using a single hollow fiber in operation. Therefore, an exhaustive EME device was developed using a sheet of thin flat porous membrane, which enables much larger capacity of acceptor solution volume (up to 600 μl has been tested) (Huang et al. 2014). With this new and improved technical EME set-up, exhaustive EME of basic drugs (Huang et al. 2014), acidic drugs (Huang et al. 2015a,b,c,d,e,f), and peptides (Huang et al. 2015a,b,c,d,e,f) has been achieved. Dual EME (Seidi et al. 2012, Arjomandi-Behzad et al. 2013, Tabani et al. 2013a,b,c, Ara et al. 2015) and all-in-one EME (Koruni et al. 2014) were tailor-made for simultaneous EME of cations and anions or ions with different polarities. Parallel EME (Eibak et al. 2014a,b) was proposed to improve the throughput of sample pretreatment using 96-well technology (Eibak et al. 2014a,b). EME has also been coupled with other sample preparation techniques such as ultrasound-assisted emulsification microextraction (Guo and Lee 2012), dispersive liquid-liquid microextraction (Seidi et al. 2013), solid phase microextraction (Rezazadeh et al. 2013a,b, Rezazadeh et al. 2014a,b,c, Abedi and Ebrahimzadeh 2015, Fakhari et al. 2015a,b, Rezazadeh et al. 2015a,b), and liquid-phase microextraction (Huang et al. 2015a,b,c,d,e,f).

With the advantages mentioned above and sufficient sample clean-up capability (Figure 2), EME has been used for the extraction of basic drugs (Fotouhi et al. 2011, Seidi et al. 2011a,b, Šlampová et al. 2012, Hasheminasab et al. 2013, Fakhari et al. 2014, Kubáň and Boček 2014a,b, Liu et al. 2014, Sun et al. 2014), acidic drugs (Balchen et al. 2007, Payan et al. 2011, Hasheminasab et al. 2013, Fakhari et al. 2015a,b), amino acids (Strieglerová et al. 2011a,b, Rezazadeh et al. 2013a,b, Rezazadeh et al. 2014a,b,c, Rezazadeh et al. 2015a,b), small peptides (Balchen et al. 2008, Balchen et al. 2009, Balchen et al. 2012), organic pollutants (Alhooshani et al. 2011, Suh et al. 2015a,b, Zhang et al. 2015a,b), and inorganic cations (Basheer et al. 2008, Kubáň et al. 2011, Hosseiny Davarani et al. 2015) and anions (Hu et al. 2011, Nojavan et al. 2014) from biological fluids and environmental samples with different matrix components, respectively. However, EME is still facing some challenges such as pH shift and high system-current generated during EME and limited recoveries for polar species as discussed in previous reviews (Majors 2010, Gjelstad and Pedersen-Bjergaard 2011, Petersen et al. 2011a,b, Seip et al. 2012, Gjelstad and Pedersen-Bjergaard 2013, Gjelstad and Pedersen-Bjergaard 2014, Rezazadeh et al. 2014a,b,c, Carasek and Merib 2015, Gjelstad et al. 2015, Huang et al. 2015a,b,c,d,e,f, Seip et al. 2015, Ocaña-González et al. 2016). From the authors’ point of view, discovery or development of new, stable, and efficient SLMs for EME is the most important element for future propagation of the concept.

Figure 2: Sample clean-up capability of EME: chromatograms of the acceptor solutions after 20-min EME from 40 times diluted plasma (1) and after 10-min EME from 6 times diluted plasma (2), and the standard solution (3) (STD, 5 μg/ml). Reprinted from Ref. Huang et al. 2015f, with permission © 2015 Elsevier B.V.
Figure 2:

Sample clean-up capability of EME: chromatograms of the acceptor solutions after 20-min EME from 40 times diluted plasma (1) and after 10-min EME from 6 times diluted plasma (2), and the standard solution (3) (STD, 5 μg/ml). Reprinted from Ref. Huang et al. 2015f, with permission © 2015 Elsevier B.V.

Organic solvents in EME

From a theoretical point of view and based on the steady-state model, the analyte flux (Ji) in EME is proportional to the diffusion coefficient of the analyte (Di) in the SLM (Gjelstad et al. 2007a,b, Huang et al. 2015a,b,c,d,e,f). This can be expressed using the following equations:

(1)Ji=Dihm(1+νlnχ){χ1χexp(ν)(cihci0exp(ν))} (1)
(2)Di=kT/6πηR (2)

Here, cih and ci0 are the analyte concentrations at the SLM/sample interface and the acceptor solution/SLM interface; hm is the thickness of the SLM;ν is the driving force; χ is the ion balance; T is the absolute temperature (K); k is the Boltzmann constant; η is the viscosity of the organic solvent; and R is the radius of the analyte.

Thus, Ji is inversely proportional to the viscosity of the organic solvent (Equation 3), which means that organic solvents of low viscocity are beneficial in terms of analyte flux (Ji).

(3)Ji1/η (3)

Based on the transient model (Seip et al. 2013a,b, Huang et al. 2015a,b,c,d,e,f), the amount of analyte transferred to the acceptor solution depends on the SLM permeability coefficient from the sample to the acceptor solution (PiDA), the distribution coefficient of the analyte between the organic solvent and the aqueous phases (Kd*), and also the volume of the organic solvent (Vm), as shown in Equation (4). The two former parameters indicate that the affinity of the analyte for the organic solvent affect the EME efficiency.

(4)CAi(t)=VDCDi0CDi0exp(AfPiDAVDt)(VD+KdVm)VA, (4)

where VD, VA, and Vm are the volume of the sample, the acceptor solution, and the SLM; CDi0 and CAi(t) are the analyte concentrations in the sample (at t=0) and in the acceptor solution (at time t); and Af is the contact surface area of the SLM.

From the two mass transfer models discussed above, the EME efficiency is strongly dependent on the properties of the organic solvent. A low viscosity organic solvent is preferable, and high affinity and strong molecular interactions between the organic solvent and the analyte are required for efficient EME.

The stability of an EME system must be addressed for a successful operation. As reported, the system-current across the SLM in an EME system depends strongly on the applied voltage, the resistance of the immobilized organic solvent (SLM), and the two aqueous phases (Moazami et al. 2014). As the two aqueous phases normally display high conductivity, the system-current in EME under a fixed voltage mainly depends on the resistance of the SLM. This is strongly related to the properties of the organic solvent and the amount of solvent in use. Solvents with low water solubility and low volatility are preferred. In addition, because the support of the SLM used in EME is hydrophobic in nature (normally polypropylene), the solvents should possess suitable surface tension in order to be easily and well spread on the hydrophobic material.

In summary, ideal organic solvents in EME should fulfil several criteria, such as a certain hydrophobicity to spread well in the membrane support, strong ability for hydrogen binding interactions or dipole-dipole interactions to facilitate the transfer of analytes from the sample into the SLM, low viscosity to maintain a fast mass transfer of the analytes from the SLM into the acceptor solution, low water solubility and high boiling point to minimize the loss of the SLM, and to maintain the stability of the SLM during EME (Kubáň et al. 2010, Nojavan and Fakhari 2010, Kiplagat et al. 2011, Seidi et al. 2011a,b, Balchen et al. 2012, Davarani et al. 2013a,b, Ramos-Payán et al. 2013, Rezazadeh et al. 2013a,b, Seidi et al. 2013, Tabani et al. 2013a,b,c, Liu et al. 2014, Nojavan et al. 2014, Sanagi et al. 2014, Šlampová et al. 2014a,b, Fakhari et al. 2015a,b, Fotouhi et al. 2015, Suh et al. 2015a,b, Sun et al. 2015). After a comprehensive literature survey, we here give a summary of the organic solvents used up to date in EME, including the following groups: (1) nitroaromatic and other ethers; (2) benzenes; (3) alcohols; (4) ketones and aldehydes; (5) other solvents including alkylated phosphates, nitriles, esters, amines, carboxylic acids, and ionic liquids. In addition, the additives used in EME to facilitate the extraction are reviewed as well.

Nitroaromatic ethers and other ethers

Different ethers have been tested as the SLM in EME, including NPOE (Figure 3), 2-nitrophenyl pentyl ether (NPPE), 2-nitrophenyl phenyl ether, 2-nitrodiphenyl ether, dodecyl-2-nitrophenyl ether, benzyl 2-nitrophenyl ether, phenyl methyl ether (anisole), ethyl phenyl ether (phenetole), di-hexyl ether (DHE) (Balchen et al. 2007, Basheer et al. 2008, Seidi et al. 2011a,b, Seip et al. 2014a,b).

Figure 3: Structures for the representatives of organic solvents in EME.
Figure 3:

Structures for the representatives of organic solvents in EME.

Among all the organic solvents tested, NPOE has been the most popular and the most efficient solvent in EME for basic drugs (Gjelstad 2010, Majors 2010, Petersen et al. 2011a,b, Gjelstad and Pedersen-Bjergaard 2013, Gjelstad and Pedersen-Bjergaard 2014, Gjelstad et al. 2015, Huang et al. 2015a,b,c,d,e,f, Seip et al. 2015). NPOE was selected as the optimal SLM for the first demonstration of the EME concept (Pedersen-Bjergaard and Rasmussen 2006). In this case, five non-polar basic drugs (pethidine, nortriptyline, methadone, haloperidol, and loperamide) were used as model analytes, and the EME recoveries were in the range of 70–79% after EME for 5 min using a voltage of 300 V. After this first successful application in EME, NPOE has been used frequently for EME of non-polar basic drugs (Gjelstad et al. 2007a,b, Petersen et al. 2009, Nojavan and Fakhari 2010, Petersen et al. 2010, Eskandari et al. 2011, Petersen et al. 2011a,b, Seidi et al. 2011a,b, Domínguez et al. 2012, Eibak et al. 2012a,b, Fakhari et al. 2012, Gjelstad et al. 2012, Petersen et al. 2012, Eibak et al. 2012a,b, Kubáň and Boček 2012, Nojavan et al. 2012, Ahmar et al. 2013, Arjomandi-Behzad et al. 2013, Nojavan et al. 2013, Payán et al. 2013, Seidi et al. 2013, Seip et al. 2013a,b, Tabani et al. 2013a,b,c, Dugstad et al. 2014, Eibak et al. 2014a,b, Fakhari et al. 2014, Huang et al. 2014, Kubáň and Boček 2014a,b, Rezazadeh et al. 2014a,b,c, Sanagi et al. 2014, Seip et al. 2014a,b, Tabani et al. 2014, Abdossalami Asl et al. 2015, Asl et al. 2015a,b, Bazregar et al. 2015, Fakhari et al. 2015a,b, Fuchs et al. 2015, Hasheminasab and Fakhari 2015). The success of NPOE for EME of non-polar basic drugs is due to several facts. First, the hydrophobic property of the solvent results in rapid and highly stable immobilization within the porous membrane support. Second, NPOE has extremely low water solubility (6×10–3 g/l) and relatively high boiling point (351°C), and therefore, the SLM is not prone to dissolution into the sample or to evaporation. Third, and highly important, NPOE has high Kamlet Taft values for dipolarity-polarizability (π*) and hydrogen bonding basicity (β). Thus, NPOE effectively interact with positively charged (protonated) drug substances through dipole-dipole interactions and hydrogen bonding interactions. In the latter aspect, NPOE serve as a strong hydrogen bonding acceptor.

Besides NPOE, the closely related solvent NPPE has been studied for EME of non-polar basic drugs (Middelthon-Bruer et al. 2008, Petersen et al. 2009, Eibak et al. 2010, Petersen et al. 2010, Petersen et al. 2011a,b, Rezazadeh et al. 2011, Seidi et al. 2011a,b, Jamt et al. 2012, Rezazadeh et al. 2012, Arjomandi-Behzad et al. 2014, Seip et al. 2014a,b, Ara et al. 2015, Yamini et al. 2015). However, in most cases, NPPE was inferior to NPOE. Thus, only in one case (Middelthon-Bruer et al. 2008), NPPE was selected as the optimal SLM to extract singly charged basic drugs with log P>2 using 50 V, and the recoveries were in the range of 30–81% after 5 min of EME. DHE, which has been used successfully for liquid-phase microextraction (LPME) of acidic drugs (Boström et al. 2014, Huang et al. 2015a,b,c,d,e,f), has been tested several times in EME for acidic and basic drugs. In all cases, however, DHE has been inefficient as SLM (Pedersen-Bjergaard and Rasmussen 2006, Balchen et al. 2007, Payan et al. 2011, Seidiet al. 2011a,b, Hasheminasab and Fakhari 2013, Hasheminasab et al. 2013, Ramos-Payán et al. 2013, Rezazadeh et al. 2013a,b, Asl et al. 2015a,b, Fakhari et al. 2015a,b, Hasheminasab and Fakhari 2015, Hidalgo et al. 2015). This is due to the relatively lower β and π* values of DHE as compared to NPOE, reducing both dipole-dipole and hydrogen bonding interactions with the analytes.

Alcohols

Aliphatic alcohols including 1-butanol, 1-pentanol, 1-hexanol, 1-heptanol, 1-octanol, 1-nonanol, 1-decanol, 1-undecanol, 1-dodecanol, 2-undecanol,iso-amyl alcohol, 2-ethylhexanol, 2-octyl-1-dodecanol, and 2-hexyl-1-decanol have been tested in EME as the SLM (Lee et al. 2009, Seidi et al. 2011a,b, Strieglerová et al. 2011a,b, Guo and Lee 2012, Seip et al. 2014a,b, Huang et al. 2015a,b,c,d,e,f, Suh et al. 2015a,b, Tak et al. 2015, Zhang et al. 2015a,b, Hosseiny Davarani et al. 2016, Rahmani et al. 2016, Zahedi et al. 2016). Both experiments with pure solvents and with carriers added to the solvent have been reported. Alcohols were found to be useful SLMs with/without carriers for EME of several acidic drugs, peptides, and metal ions because of their high hydrogen bonding acidity (α) and basicity (β) and high/moderate polarizability (π*) (Balchen et al. 2007, Basheer et al. 2010, Payan et al. 2011, Strieglerová et al. 2011a,b, Tabani et al. 2013a,b,c, Koruni et al. 2014, Huang et al. 2015a,b,c,d,e,f). Among the aliphatic alcohols, 1-octanol (Figure 3) was one of the most frequently used EME SLMs with/without carriers to extract peptides (Balchen et al. 2012), metal ions (Kubáň et al. 2011, Hosseiny Davarani et al. 2015), acidic drugs (Payan et al. 2011, Koruni et al. 2014), chlorophenols (Lee et al. 2009, Šlampová et al. 2013), and inorganic anions (Safari et al. 2013). 1-Nonanol (Figure 3) was used to replace 1-octanol to improve the stability of the EME system for peptides (Huang et al. 2015a,b,c,d,e,f, Huang et al. 2015a,b,c,d,e,f). 1-Nonanol provided a SLM of higher stability than 1-octanol due to lower water solubility. Besides 1-octanol, 1-heptanol (Figure 3) has been successfully used as the SLM to extract acidic drugs (Balchen et al. 2007, Huang et al. 2015a,b,c,d,e,f) and to extract inorganic anions (Kiplagat et al. 2011, Nojavan et al. 2014). 1-Butanol, 1-pentanol, 1-hexanol, and 2-ethylhexanol were not suitable SLMs for EME due to water solubility. 1-Decanol, 1-undecanol, 1-dodecanol, 2-undecanol, isoamylalcohol, 2-ethylhexanol, 2-octyl-1-dodecanol, and 2-hexyl-1-decanol were unsuccessful SLMs. Most of them are highly hydrophobic alcohols with relatively high viscosity and less prominent hydrogen bonding capability (α and β).

Phenols including eugenol, m-cresol, di-sec-butylphenol, 5-isopropyl-2-methylphenol, 2-isopropyl phenol, and 2-methoxy-4-methylphenol have been tested in EME for peptides and basic drugs without success because of partial water solubility and due to high current (Balchen et al. 2011, Seip et al. 2011, Seip et al. 2014a,b).

Other alcohols including benzyl alcohol, 2-phenylethanol,2-nitrophenylethyl alcohol, tetrahydrofurfuryl alcohol, furfuryl alcohol, (+)-santolina alcohol, (–)-perillyl alcohol, and cyclohexanol have been investigated in EME (Lee et al. 2009, Payan et al. 2011, Seip et al. 2011, Davarani et al. 2013a,b, Nojavan et al. 2013, Ramos-Payán et al. 2013, Seip et al. 2014a,b, Song et al. 2015). However, they were not useful SLMs because of high current and inefficient mass transfer (Seip et al. 2011, Seip et al. 2014a,b).

Ketones

Some ketones including 2-nitroacetophenone, 2-octanone, benzyl methyl ketone, acetophenone, 2-decanone,cyclohexanone, isophorone, methyl ethyl ketone, acetyl acetone, 2-nonaone, 2-undecanone, 6-undecanone, methylacetophenone, 2,6-dimethyl-4-heptanone, α-tetralone, 3,3,5-trimethylcyclohexanone, and 1-chloropinacolone have been tested as the SLM (Pedersen-Bjergaard and Rasmussen 2006, Balchen et al. 2007, Balchen et al. 2009, Seip et al. 2011, Strieglerová et al. 2011a,b, Ahmar et al. 2013, Seip et al. 2014a,b). Among these tested ketones, 2-octanone, 2-nonaone, 2-decanone, 2-undecanone, 6-undecanone, and 2,6-dimethyl-4-heptanone were efficient for EME of non-polar drugs. The other ketones mentioned were not suitable SLMs mainly because of the high current and due to significant water solubility (Seip et al. 2014a,b). In addition, with the assistance of an ion-pair reagent (DEHP), 2-octanone and 2-nonaone (Figure 3) were successful organic solvents for EME of several model peptides (Seip et al. 2011).

Aromatics and substituted aromatics

Benzene, benzene derivatives [1-ethyl-2-nitrobenzene (ENB), 1-isopropyl-4-nitrobenzene (IPNB), 2,4-dimethyl-1-nitrobenzene, nitrobenzene, 1,2,4-trifluoro-5-nitrobenzene, 1,2,4-trifluoronitrobenzene, (2,2-dimethyl-1-propyl) benzene, pentylbenzene, bromobenzene, and chlorobenzene], 4-nitro-m-xylene, o-xylene, toluene, 2-nitrotoluene, cumene, and 3-nitrostyrene have all been tested as SLMs in EME (Balchen et al. 2007, Balchen et al. 2009, Kubáň et al. 2011, Seidi et al. 2011a,b, Seip et al. 2014a,b). Most of them are not suitable, but some nitrobenzene derivatives, namely, IPNB, ENB, and 2,4-dimethyl-1-nitrobenzene (Figure 3), were found to be successful SLMs in EME because of high dipolarity-polarizability (π*) and high hydrogen bonding basicity (β) (Gjelstad et al. 2009, Jamt et al. 2012, Šlampová et al. 2012, Eibak et al. 2014a,b, Seip et al. 2014a,b, Šlampová et al. 2014a,b). For example, IPNB was used to extract amitriptyline, fluoxetine, and haloperidol from undiluted human plasma and urine using 20 V (Eibak et al. 2014a,b). Stimulating drugs including cathinone; methamphetamine; 3,4-methylenedioxy-amphetamine; 3,4-methylenedioxy methamphetamine; ketamine; and 2,5-dimethoxy-4-iodoamphetamine were extracted successfully from undiluted whole blood and post mortem blood by EME with an SLM of ENB (Jamt et al. 2012). ENB also has been used to extract some basic drugs from diluted human urine and serum (Šlampová et al. 2012). In addition, several basic drugs were extracted from undiluted biological samples including human urine and serum using μ-EME and ENB as the free liquid membrane (Kubáň and Boček 2014a,b).

Toluene has been studied intensively as the SLM in EME by different research groups (Basheer et al. 2008, Basheer et al. 2010, Alhooshani et al. 2011, Davarani et al. 2013a,b, Safari et al. 2013, Liu et al. 2014, Sanagi et al. 2014, Šlampová et al. 2014a,b, Zhang et al. 2015a,b). For example, toluene was used as the SLM in EME to determine the concentration of lead ions in amniotic fluid, blood serum, lipstick, and urine samples (Basheer et al. 2008). In another example, haloacetic acids and aromatic acetic acids were extracted from wastewater by EME with an SLM of toluene (Alhooshani et al. 2011). In addition, simultaneous extraction of acidic and basic drugs at neutral sample pH was achieved with an SLM of toluene by EME under 300 V (Basheer et al. 2010). However, toluene is not recommended because of volatility, which might cause inconsistent results (Lee et al. 2009).

Alkanes and substituted alkanes

Alkanes and related derivatives including 1-nitropropane, nitropentane, 1-hexane, 1,10-dichlorodecane, dichloromethane (DCM), hexadecane, nitrocyclohexane, n-tetradecane, nitromethane, n-hexane, and kerosene (lamp oil) have been studied (Pedersen-Bjergaard and Rasmussen 2006, Kjelsen et al. 2008, Basheer et al. 2010, Alhooshani et al. 2011, Seidi et al. 2011a,b, Guo and Lee 2012, Nojavan et al. 2013, Seip et al. 2014a,b, Šlampová et al. 2014a,b, Mamat and See 2015, Suh et al. 2015a,b). However, very low or no recoveries were obtained from all cases. This was mainly due to high volatility causing serious losses of the SLM during EME (Basheer et al. 2010, Nojavan et al. 2013) or due to low values of α, β, and π*(Suh et al. 2015a,b).

Ionic liquids

Most recently, ionic liquids (ILs) including imidazoliumhexafluorophosphate-based ILs[CnMIm][PF6] (n=4, 6, 8), 1-butyl-3-methylimidazolium-hexafluorophosphate ([C4MIm][PF6]), 1-hexyl-3-methylimidazolium-hexafluorophosphate ([C6MIm][PF6]) (Figure 3), and 1-octyl-3-methylimidazolium-hexafluorophosphate ([C8MIm][PF6]) were applied for extraction of basic and acidic compounds under very low voltage conditions (Sun et al. 2014, Sun et al. 2015). With ionic liquid-based EME (IL-EME), strychnine and brucine were determined in human urine (Sun et al. 2014). In another work, IL-EME was used to determine chlorophenoxy acid herbicides in pig kidney (Sun et al. 2015). IL-EME has to be accomplished with very low voltage (1.5–7.5 V) to avoid high and unstable system-current due to high conductivity. However, it can also be a great advantage for EME that efficient EME can be obtained with very low consumption of energy by using ionic liquid as SLM.

Solvent mixtures

In addition, some mixed organic solvents have been tested for peptides and acidic drugs (Balchen et al. 2009, Balchen et al. 2010, Balchen et al. 2011, Payan et al. 2011, Seip et al. 2011, Balchen et al. 2012, Ramos-Payán et al. 2013, Tabani et al. 2013a,b,c, Huang et al. 2015a,b,c,d,e,f). For example, the mixture of 1-octanol with 4-nitro-m-xylene and di-isobutylketone, 1-octanol with di-isobutyl ketone, 4-nitro-m-xylene with di-isobutyl ketone, and 1-octanol with NPOE provided efficient EME of peptides from water samples (Balchen et al. 2009). Mixtures of organic solvents (a mixture of 1-heptanol and NPOE) were used to reduce and stabilize the system-current in EME (Huang et al. 2015a,b,c,d,e,f).

Miscellaneous solvents

Alkylated phosphates (including dibutyl phosphate, tributyl phosphate, triisopropyl phosphate, tris(2-butoxyethyl) phosphate, tris(2-ethylhexyl) phosphate (TEHP), di-(2-ethylhexyl) phosphate (DEHP), and trixylyl phosphate), tri-tert-butylborate, benzyl chloride, different amines (including trihexyl amine, tert-butylamine, dibenzylamine, 2-ethylhexyl-4-(dimethylamino) benzoate, and octylamine), carboxylic acids (including, 1-octanoic acid, linoleic acid, caprylic acid, and nonanoic acid), nitriles (including cinnamonitrile and 3-tolunitrile), aldehydes (heptane aldehyde, benzaldehyde, nonanal and undecanal), peppermint oil, silicone oil AS 4, and different esters (including dodecyl acetate, dibutyl phthalate, and dioctyl phthalate) have been investigated as SLM in EME but without major success (Balchen et al. 2007, Balchen et al. 2009, Seidi et al. 2011a,b, Seip et al. 2014a,b). Unsuccessful EME with the above tested organic solvents was mainly due to significant water solubility, causing high current and an unstable SLM system. Additionally, solidified solvents (polymer inclusive membranes) have also been used successfully in EME, where the organic solvent was mixed with cellulose triacetate and polymerized after the evaporation of dichloromethane (See and Hauser 2011, Schmidt-Marzinkowski et al. 2013, See et al. 2013, Mamat and See 2015).

Additives in the SLM

In most of the EME publications, EME was conducted using pure solvents as the SLM. However, addition of ion-pair reagents in the SLM is mandatory for EME of polar substances, and additives in the SLM could enhance the EME efficiency.

Alkylated phosphates are the most intensively studied ion-pair reagents in EME of basic drugs, peptides, amino acids, and metal ions. Investigated phosphates include di-(2-ethylhexyl) phosphate (DEHP), tris-(2-ethylhexyl) phosphate (TEHP), diphenylphosphate, tridecyl phosphate (TDP), dibutyl phosphate (DBP), trixylyl phosphate, triphenyl phosphate, dihexadecyl phosphate, dibenzyl phosphate, 2-cyanoethyl phosphate, 1-naphtyl phosphate, tris(2-butoksyethyl) phosphate, bis(4-nitrophenyl) phosphate, tri-isopropyl phosphate, tributyl phosphate, tri(8-quionlyl) phosphate, tri octyl phosphine oxide, hexamethylenediamine tetra (methylene phosphonic acid), amino tris (methylenephosphonic acid), and triethyl phosphate (Balchen et al. 2008, Seip et al. 2011, Davarani et al. 2013a,b). Among those DEHP, TEHP, TDP, and DBP, whose structure is shown in Figure 4, were the most efficient ones (Middelthon-Bruer et al. 2008, Seip et al. 2011, Strieglerováet al. 2011a,b, Rezazadeh et al. 2013a,b). For example, with pure NPPE as the SLM, only singly charged basic drugs with log P>2 were extracted by EME. However, the addition of TEHP in NPPE as the SLM was required for EME of medium polar basic drugs (1<log P<2), and addition of DEHP into the SLM was crucial for EME of the most polar drugs with log P<1 (Middelthon-Bruer et al. 2008). This main difference was due to the fact that DEHP can form complexes with positively charged species by ionic interactions, while TEHP can form complexes with the analytes by hydrogen bonding. Thus, for EME of hydrophilic substances like amino acids or peptides, DEHP is always present in the SLM (Balchen et al. 2008, Balchen et al. 2009, Balchen et al. 2011, Seip et al. 2011, Strieglerová et al. 2011a,b, Balchen et al. 2012, Seip et al. 2012, Rezazadeh et al. 2013a,b, Rezazadehet al. 2014a,b,c, Huang et al. 2015a,b,c,d,e,f, Rezazadeh et al. 2015a,b). In addition, in order to improve the affinity between the SLM and the analytes, the combination of DEHP (strong ionic interaction) and TEHP (strong hydrogen bonding) was used (Rezazadeh et al. 2012, Rezazadeh et al. 2013a,b, Ahmar et al. 2014, Fashi et al. 2015, Hosseiny Davarani et al. 2015, Rezazadeh et al. 2015a,b).

Figure 4: Structures for the representatives of additives in EME.
Figure 4:

Structures for the representatives of additives in EME.

Amines including tetrabutyl ammonium hydroxide 30-hydrate, tetrabutylammonium phosphate, 1-hexyltriethyl ammonium phosphate, 1-octyltriethyl ammonium phosphate, 1-dodecyltriethyl ammonium phosphate, trimethyloctyl ammonium bromide, decyltrimethyl ammonium bromide, dodecyltrimethyl ammonium bromide, cetyltrimethyl ammonium bromide (CTAB), and tetraoctylammonium bromide have been tested as the carrier for EME of anions (Balchen et al. 2007, Lee et al. 2009, Tabani et al. 2013a,b,c, Suh et al. 2015a,b). However, in most cases, the addition of amines in the organic solvent was found to be unsuccessful because of high system-current, bubble formation in the EME system (Balchen et al. 2007), and too strong ionic interaction (Suh et al. 2015a,b). Most recently, CTAB (Figure 4) was successfully used as the carrier to extract polar acidic drugs in an all-in-one EME system (Koruni et al. 2014).

Nanoparticles have been introduced into the SLM to improve the EME performance for basic and acidic compounds by involving both solid-phase microextrcation(SPME) and EME principles (Hasheminasab and Fakhari 2013, Hasheminasab et al. 2013, Hasheminasab et al. 2014, Ramos-Payán et al. 2014, Song et al. 2015). Carbon nanotubes (CNT) were added into the organic solvent to extract buprenorphine with success (Hasheminasab and Fakhari 2013). Nanotubes dispersed in 1-octanol was also used as the SLM to extract acidic drugs (Hasheminasab et al. 2013). In addition, some acidic drugs were extracted by EME using silver nanoparticles in the SLM (Ramos-Payán et al. 2014). The addition of nanoparticles in the organic solvent resulted in more efficient EME (Hasheminasab and Fakhari 2013, Hasheminasab et al. 2013, Ramos-Payán et al. 2014).

Other carriers such as tri-tert-butyl borate, dithizone, 8-hydroxyquinoline, acids (including sodium-2-ethylhexylsulphuric acid, triisopropylnaphtalenesulfonicacid, hydroxyisobutyric acid, 1-naphtoic acid, caprylic acid, stearic acid, phenylboronic acid, and hydroxyisobutyric acid), and crown ethers (including 15-crown-5 ether, 18-crown-6, benzo-18-crown-6, dibenzo-18-crown-6 (Figure 4), dicyclohexano-18-crown-6, and 4-amino-dibenzo-18-crown-6) have been tested for EME of peptides and metal ions (Balchen et al. 2008, Balchen et al. 2011, Kubáň et al. 2011, Seip et al. 2011, Šlampová et al. 2014a,b). Among all these tested carriers, crown ethers were useful for selective EME of hydrophobic peptides (Balchen et al. 2011) and K+ (Šlampová et al. 2014a,b) due to the complexation.

Other parameters in EME

As discussed above, the system-current in the EME system and the EME efficiency strongly depends on the properties of the organic solvent. Besides that, to construct a stable and efficient EME system, some other operation parameters such as the SLM volume, extraction voltage, pH in sample and acceptor solution, acceptor solution composition, and extraction time also need to be optimized.

It has been found that the SLM volume also affects the system-current and the EME recovery of some small peptides (Huang et al. 2015a,b,c,d,e,f). Larger SLM volume (≥7.5 μl) could reduce and stabilize the system-current during EME. In addition, the recoveries of peptides were improved by using an optimal SLM volume of 10 μl.

Extraction voltage can affect the selectivity, efficiency, and stability in EME. In one example, all five basic drugs were extracted under higher voltage (50 V), whereas only two basic drugs possessing the lowest polar surface area were extracted with lower voltage (5 V) (Domínguez et al. 2012). Generally, high voltage resulted in higher EME recovery but also higher system-current. The latter affects the EME efficiency in turn. Normally, unstable EME system leads to a reduction in EME efficiency and also irreproducible results. Thus, the extraction voltage should be carefully optimized after the optimization of the organic solvent to propose the most efficient EME system but without causing any stability problems.

The pH in sample and in the acceptor solution should ionize the analytes of interests. In addition, buffer solution is preferable to be used to overcome the pH shift because of the electrolysis during the EME process (Šlampová et al. 2007, Huang et al. 2015a,b,c,d,e,f, Kubáň and Boček 2015, Šlampová et al. 2015). The composition of the acceptor solution affects the EME efficiency as well as the system-current (Huang et al. 2015a,b,c,d,e,f).

The optimal extraction time for EME depends strongly on the type of analytes and the parameters that we discussed above. For pharmaceuticals, normally, the optimal extraction time is within 15 min, and longer extraction time will result in lower recovery because of the back-extraction due to the pH shift in the acceptor solution. In a recent publication, the extraction time was reduced to 30 s (Song et al. 2015).

Future directions

As discussed in the current review, a large number of different organic solvents have been tested as the SLM in EME. Only a few of these solvents have been highly efficient, and this supports that several criteria have to be fulfilled for an EME solvent. Clearly, the solvent has to be immiscible with water (low polarity) and of low volatility in order to avoid leakage to the sample and evaporative losses. In addition, the successful EME solvent also has to be carefully balanced in terms of dipolarity-polarizability (π*), hydrogen bonding acidity (α), and hydrogen bonding basicity (β). For future development of EME, the number of efficient EME solvents should definitely be increased, especially for anionic analytes and polar analytes. Thus, more research in this field is required. Further testing of pure organic solvents may be one approach, but testing of solvent additives insoluble in water may be even more interesting. Future additives may be based on ionic interactions, but also carriers with high α and β values should be tested. The latter type of carriers will not contribute to excessive current in the EME system, and this is an important practical point that should not be overlooked.

About the authors

Chuixiu Huang

Chuixiu Huang received her BS in Chemical Engineering from Huazhong University of Science and Technology (China) in 2002, and she received her MS in Analytical Chemistry from Lund University (Sweden) in 2012. She received her PhD in Pharmaceutical Chemistry from University of Oslo (Norway) in 2016 with focus on the development and applications of electromembrane extraction (EME) for bioanalysis. She has previously published a few articles on EME. Currently, she is a postdoctoral student at the University of Oslo with focus on the development and discovery of new liquid membranes for EME.

Astrid Gjelstad

Astrid Gjelstad is currently an associate professor at the Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo. She received her PhD degree in 2009 and performed her doctoral training also at School of Pharmacy, University of Oslo. The academic field of interest is development of new microextraction technologies for rapid and selective isolation of drugs and peptides from small amounts of biological matrices. The microextraction technology is based on artificial liquid membranes where studies of the transport across the membranes forced by either passive diffusion (liquid-phase microextraction, LPME) or electrokinetic migration (electromembrane extraction, EME) are special focus areas. Another research focus comprises new inventions in the area of dried blood spots (DBS).

Stig Pedersen-Bjergaard

Stig Pedersen-Bjergaard (SPB) was born in Copenhagen in 1966. In 1996, SPB was appointed as Associate Professor at the School of Pharmacy, University of Oslo (Oslo, Norway) and, from year 2000, as full Professor at the same institution. In 2008, SPB was appointed as full Professor (part time) at Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen (Copenhagen, Denmark). SPB has specialized in analytical micro extraction technologies on the development and applications of artificial liquid membranes and on electro-kinetic separation methods. SPB has published more than 155 papers in international journals, given more than 85 presentations at international symposia, contributed to 7 patent applications, and authored two international student textbooks in pharmaceutical analysis. SPB is Contributing Editor for Trends in Analytical Chemistry and is a member of the Editorial board for Journal of Chromatography A, Analytica Chimica Acta, Journal of Pharmaceutical Analysis, and International Journal of Analytical Chemistry.

References

Abdossalami Asl, Y.; Yamini, Y.; Seidi, S.; Ebrahimpour, B. A new effective on chip electromembrane extraction coupled with high performance liquid chromatography for enhancement of extraction efficiency. Anal. Chim. Acta2015, 898, 42–49.10.1016/j.aca.2015.09.052Search in Google Scholar PubMed

Abedi, H.; Ebrahimzadeh, H. Electromembrane-surrounded solid-phase microextraction coupled to ion mobility spectrometry for the determination of nonsteroidal anti-inflammatory drugs: a rapid screening method in complicated matrices. J. Sep. Sci.2015, 38, 1358–1364.10.1002/jssc.201401350Search in Google Scholar PubMed

Ahmar, H.; Fakhari, A. R.; Tabani, H.; Shahsavani, A. Optimization of electromembrane extraction combined with differential pulse voltammetry using modified screen-printed electrode for the determination of sufentanil. Electrochim. Acta2013, 96, 117–123.10.1016/j.electacta.2013.02.049Search in Google Scholar

Ahmar, H.; Tabani, H.; Hossein Koruni, M.; Davarani, S. S. H.; Fakhari, A. R. A new platform for sensing urinary morphine based on carrier assisted electromembrane extraction followed by adsorptive stripping voltammetric detection on screen-printed electrode. Biosen. Bioelectron.2014, 54, 189–194.10.1016/j.bios.2013.10.035Search in Google Scholar PubMed

Alhooshani, K.; Basheer, C.; Kaur, J.; Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard S.; Lee, H. K. Electromembrane extraction and HPLC analysis of haloacetic acids and aromatic acetic acids in wastewater. Talanta2011, 86, 109–113.10.1016/j.talanta.2011.08.026Search in Google Scholar PubMed

Ara, K. M.; Raofie, F.; Seidi, S. Simultaneous extraction and determination of trace amounts of olanzapine and fluoxetine from biological fluids: comparison of conventional hollow fiber supported liquid phase microextraction and pulsed electrically assisted liquid-phase microextraction techniques. Anal. Methods2015, 7, 7840–7851.10.1039/C5AY00345HSearch in Google Scholar

Arjomandi-Behzad, L.; Yamini, Y.; Rezazadeh, M. Pulsed electromembrane method for simultaneous extraction of drugs with different properties. Anal. Biochem.2013, 438, 136–143.10.1016/j.ab.2013.03.027Search in Google Scholar PubMed

Arjomandi-Behzad, L.; Yamini, Y.; Rezazadeh, M. Extraction of pyridine derivatives from human urine using electromembrane extraction coupled to dispersive liquid-liquid microextraction followed by gas chromatography determination. Talanta2014, 126, 73–81.10.1016/j.talanta.2014.02.066Search in Google Scholar PubMed

Arthur, C. L.; Pawliszyn, J. Solid phase microextraction with thermal desorption using fused silica optical fibers. Anal. Chem.1990, 62, 2145–2148.10.1021/ac00218a019Search in Google Scholar

Asl, Y. A.; Yamini, Y.; Rezazadeh, M.; Seidi, S. Electromembrane extraction using a cylindrical electrode: A new view for the augmentation of extraction efficiency. Anal. Methods2015a, 7, 197–204.10.1039/C4AY02327GSearch in Google Scholar

Asl, Y. A.; Yamini, Y.; Seidi, S.; Amanzadeh, H. Dynamic electromembrane extraction: Automated movement of donor and acceptor phases to improve extraction efficiency. J. Chromatogr. A.2015b, 1419, 10–18.10.1016/j.chroma.2015.09.077Search in Google Scholar

Balchen, M.; Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Electrokinetic migration of acidic drugs across a supported liquid membrane. J. Chromatogr. A2007, 1152, 220–225.10.1016/j.chroma.2006.10.096Search in Google Scholar

Balchen, M.; Reubsaet, L.; Pedersen-Bjergaard, S. Electromembrane extraction of peptides.“ J. Chromatogr. A2008, 1194, 143–149.10.1016/j.chroma.2008.04.041Search in Google Scholar

Balchen, M.; Halvorsen, T. G.; Reubsaet, L.; Pedersen-Bjergaard, S. Rapid isolation of angiotensin peptides from plasma by electromembrane extraction. J. Chromatogr. A2009, 1216, 6900–6905.10.1016/j.chroma.2009.08.037Search in Google Scholar

Balchen, M.; Jensen, H.; Reubsaet, L.; Pedersen-Bjergaard, S. Potential-driven peptide extractions across supported liquid membranes: investigation of principal operational parameters. J. Sep. Sci.2010, 33,1665–1672.10.1002/jssc.201000025Search in Google Scholar

Balchen, M.; Hatterud, A. G.; Reubsaet, L.; Pedersen-Bjergaard, S. Fundamental studies on the electrokinetic transfer of net cationic peptides across supported liquid membranes. J. Sep. Sci.2011, 34, 186–195.10.1002/jssc.201000703Search in Google Scholar

Balchen, M.; Lund, H.; Reubsaet, L.; Pedersen-Bjergaard, S. Fast, selective, and sensitive analysis of low-abundance peptides in human plasma by electromembrane extraction. Anal. Chim. Acta2012, 716, 16–23.10.1016/j.aca.2011.02.058Search in Google Scholar

Baltussen, E.; Sandra, P.; David, F.; Cramers, C. Stir bar sorptive extraction (SBSE), a novel extraction technique for aqueous samples: theory and principles. J. Microcolumn Sep.1999, 11, 737–747.10.1002/(SICI)1520-667X(1999)11:10<737::AID-MCS7>3.0.CO;2-4Search in Google Scholar

Basheer, C.; Tan, S. H.;Lee, H. K. Extraction of lead ions by electromembrane isolation. J. Chromatogr. A2008, 1213, 14–18.10.1016/j.chroma.2008.10.041Search in Google Scholar

Basheer, C.; Lee, J.; Pedersen-Bjergaard, S.; Rasmussen, K. E.; Lee, H. K. Simultaneous extraction of acidic and basic drugs at neutral sample pH: A novel electro-mediated microextraction approach. J. Chromatogr. A2010, 1217, 6661–6667.10.1016/j.chroma.2010.04.066Search in Google Scholar

Basheer, C.; Alhooshani, K.; Nuhu, A. A.; Kanimozhi, S.; Lee, H. K. Sample preparation of complex biological samples in the analysis of trace-level contaminants. Comprehensive Sampling and Sample Preparation2012, 3, 681–700.10.1016/B978-0-12-381373-2.00109-5Search in Google Scholar

Bazregar, M.; Rajabi, M.; Yamini, Y.; Asghari, A.; Abdossalami Asl, Y. In-tube electro-membrane extraction with a sub-microliter organic solvent consumption as an efficient technique for synthetic food dyes determination in foodstuff samples. J. Chromatogr. A2015, 1410, 35–43.10.1016/j.chroma.2015.07.084Search in Google Scholar PubMed

Boström, M. L.; Huang, C.; Engström, H.; Larsson, E.; Berglund, O.; Jönsson, J. A. A specific, highly enriching and green method for hollow fiber liquid phase microextraction of ionizable pharmaceuticals from fish tissue. Anal. Methods2014, 6, 6031–6037.10.1039/C4AY00470ASearch in Google Scholar

Carasek, E.; Merib, J. Membrane-based microextraction techniques in analytical chemistry: a review. Anal. Chim. Acta2015, 880, 8–25.10.1016/j.aca.2015.02.049Search in Google Scholar PubMed

Dadfarnia, S.; Salmanzadeh, A. M.; Shabani, A. M. H. A novel separation/preconcentration system based on solidification of floating organic drop microextraction for determination of lead by graphite furnace atomic absorption spectrometry. Anal. Chim. Acta2008, 623, 163–167.10.1016/j.aca.2008.06.033Search in Google Scholar PubMed

Davarani, S. S. H.; Morteza-Najarian, A.; Nojavan, S.; Pourahadi, A.;Abbassi, M. B. Two-phase electromembrane extraction followed by gas chromatography-mass spectrometry analysis. J. Sep. Sci.2013a, 36, 736–743.10.1002/jssc.201200838Search in Google Scholar PubMed

Davarani, S. S. H.; Moazami, H. R.; Keshtkar, A. R.; Banitaba, M. H.;Nojavan, S. A selective electromembrane extraction of uranium (VI) prior to its fluorometric determination in water. Anal. Chim. Acta2013b, 783, 74–79.10.1016/j.aca.2013.04.045Search in Google Scholar PubMed

Domínguez, N. C.; Gjelstad, A.; Nadal, A. M.; Jensen, H.; Petersen, N. J.; Hansen, S. H.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Selective electromembrane extraction at low voltages based on analyte polarity and charge. J. Chromatogr. A2012, 1248, 48–54.10.1016/j.chroma.2012.05.092Search in Google Scholar PubMed

Dugstad, H. B.; Petersen, N. J.; Jensen, H.; Gabel-Jensen, C.; Honoré Hansen, S.; Pedersen-Bjergaard,S. Development and characterization of a small electromembrane extraction probe coupled with mass spectrometry for real-time and online monitoring of in vitro drug metabolism. Anal. Bioanal. Chem.2014, 406, 421–429.10.1007/s00216-013-7378-zSearch in Google Scholar PubMed

Eibak, L. E. E.; Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Kinetic electro membrane extraction under stagnant conditions–fast isolation of drugs from untreated human plasma. J. Chromatogr. A2010, 1217, 5050–5056.10.1016/j.chroma.2010.06.018Search in Google Scholar PubMed

Eibak, L. E. E.; Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Exhaustive electromembrane extraction of some basic drugs from human plasma followed by liquid chromatography-mass spectrometry. J. Pharm. Biomed. Anal.2012a, 57, 33–38.10.1016/j.jpba.2011.08.026Search in Google Scholar PubMed

Eibak, L. E. E.; Hegge, A. B.; Rasmussen, K. E.; Pedersen-Bjergaard, S.;Gjelstad, A. Alginate and chitosan foam combined with electromembrane extraction for dried blood spot analysis. Anal. Chem.2012b, 84, 8783–8789.10.1021/ac301996nSearch in Google Scholar PubMed

Eibak, L. E. E.; Parmer, M. P.; Rasmussen, K. E.; Pedersen-Bjergaard, S.; Gjelstad, A. Parallel electromembrane extraction in a multiwell plate. Anal. Bioanal. Chem.2014a, 406, 431–440.10.1007/s00216-013-7345-8Search in Google Scholar PubMed

Eibak, L. E. E.; Rasmussen, K. E.; Øiestad, E. L.; Pedersen-Bjergaard, S.; Gjelstad, A. Parallel electromembrane extraction in the 96-well format. Anal. Chim. Acta2014b, 828, 48–52.10.1016/j.aca.2014.04.038Search in Google Scholar PubMed

Eskandari, M.; Yamini, Y.; Fotouhi, L.; Seidi, S. Microextraction of mebendazole across supported liquid membrane forced by pH gradient and electrical field. J. Pharm. Biomed. Anal.2011, 54, 1173–1179.10.1016/j.jpba.2010.12.006Search in Google Scholar PubMed

Fakhari, A. R.; Tabani, H.; Nojavan, S.; Abedi, H. Electromembrane extraction combined with cyclodextrin-modified capillary electrophoresis for the quantification of trimipramine enantiomers. Electrophoresis2012, 33, 506–515.10.1002/elps.201100426Search in Google Scholar PubMed

Fakhari, A. R.; Koruni, M. H.; Ahmar, H.; Shahsavani, A.; Movahed, S. K. Electrochemical determination of dextromethorphan on reduced graphene oxide modified screen-printed electrode after electromembrane extraction. Electroanalysis2014, 26, 521–529.10.1002/elan.201300323Search in Google Scholar

Fakhari, A. R.; Hasheminasab, K. S.; Aladaghlo, Z.; Koruni, M. H. Surfactant-assisted electromembrane extraction combined with capillary electrophoresis as a novel technique for the determination of acidic drugs in biological fluids. Electrophoresis2015a, 36, 3034–3041.10.1002/elps.201500268Search in Google Scholar PubMed

Fakhari, A. R.; Sahragard, A.; Ahmar, H.; Tabani, H. A novel platform sensing based on combination of electromembrane-assisted solid phase microextraction with linear sweep voltammetry for the determination of tramadol. J. Electroanal. Chem.2015b, 747, 12–19.10.1016/j.jelechem.2015.01.032Search in Google Scholar

Fashi, A.; Yaftian, M. R.; Zamani, A. Determination of melamine in dairy products using electromembrane-LPME followed by HPLC. Food Chem.2015, 188, 92–98.10.1016/j.foodchem.2015.04.132Search in Google Scholar PubMed

Fotouhi, L.; Yamini, Y.; Molaei, S.; Seidi, S. Comparison of conventional hollow fiber based liquid phase microextraction and electromembrane extraction efficiencies for the extraction of ephedrine from biological fluids. J. Chromatogr. A2011, 1218, 8581–8586.10.1016/j.chroma.2011.09.078Search in Google Scholar PubMed

Fotouhi, L.; Yamini, Y.; Hosseini, R.; Rezazadeh, M. Determination of phenazopyridine in biological fluids using electromembrane extraction followed by high-performance liquid chromatography. Can. J. Chem.2015, 93, 702–707.10.1139/cjc-2014-0460Search in Google Scholar

Fuchs, D.; Jensen, H.; Pedersen-Bjergaard, S.; Gabel-Jensen, C.; Hansen, S. H.; Petersen, N. J. Real time extraction kinetics of electro membrane extraction verified by comparing drug metabolism profiles obtained from a flow-flow electro membrane extraction-mass spectrometry system with LC-MS. Anal. Chem.2015, 87, 5774–5781.10.1021/acs.analchem.5b00981Search in Google Scholar PubMed

Gjelstad, A. Electromembrane extraction: the use of electrical potential for isolation of charged substances from biological matrices. LC GC Eur.2010, 23, 152–164.Search in Google Scholar

Gjelstad, A.; Pedersen-Bjergaard, S. Electromembrane extraction: a new technique for accelerating bioanalytical sample preparation. Bioanalysis2011, 3, 787–797.10.4155/bio.11.13Search in Google Scholar PubMed

Gjelstad, A.; Pedersen-Bjergaard, S. Recent developments in electromembrane extraction. Anal. Methods2013, 5, 4549–4557.10.1039/C3AY40547HSearch in Google Scholar

Gjelstad, A.; Pedersen-Bjergaard, S. Electromembrane extraction-Three-phase electrophoresis for future preparative applications. Electrophoresis2014, 35, 2421–2428.10.1002/elps.201400127Search in Google Scholar PubMed

Gjelstad, A.; Andersen, T. M.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Microextraction across supported liquid membranes forced by pH gradients and electrical fields. J. Chromatogr. A2007a, 1157, 38–45.10.1016/j.chroma.2007.05.007Search in Google Scholar PubMed

Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Simulation of flux during electro-membrane extraction based on the Nernst-Planck equation. J. Chromatogr. A2007b, 1174, 104–111.10.1016/j.chroma.2007.08.057Search in Google Scholar PubMed

Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Electromembrane extraction of basic drugs from untreated human plasma and whole blood under physiological pH conditions. Anal. Bioanal. Chem.2009, 393, 921–928.10.1007/s00216-008-2344-xSearch in Google Scholar PubMed

Gjelstad, A.; Jensen, H.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Kinetic aspects of hollow fiber liquid-phase microextraction and electromembrane extraction. Anal. Chim. Acta2012, 742, 10–16.10.1016/j.aca.2011.12.039Search in Google Scholar PubMed

Gjelstad, A.; Rasmussen, K. E.; Parmer, M. P.; Pedersen-Bjergaard, S. Parallel artificial liquid membrane extraction: micro-scale liquid-liquid-liquid extraction in the 96-well format. Bioanalysis2013, 5, 1377–1385.10.4155/bio.13.59Search in Google Scholar PubMed

Gjelstad, A.; Pedersen-Bjergaard, S.; Seip, K. F. Electromembrane extraction as a rapid and selective miniaturized sample preparation technique for biological fluids. Bioanalysis2015, 7, 2203–2209.10.4155/bio.15.150Search in Google Scholar PubMed

Guo, L.; Lee, H. K. Electro membrane extraction followed by low-density solvent based ultrasound-assisted emulsification microextraction combined with derivatization for determining chlorophenols and analysis by gas chromatography-mass spectrometry. J. Chromatogr. A2012, 1243, 14–22.10.1016/j.chroma.2012.04.054Search in Google Scholar PubMed

Hasheminasab, K. S.; Fakhari, A. R. Development and application of carbon nanotubes assisted electromembrane extraction (CNTs/EME) for the determination of buprenorphine as a model of basic drugs from urine samples. Anal. Chim. Acta2013, 767, 75–80.10.1016/j.aca.2012.12.046Search in Google Scholar PubMed

Hasheminasab, K. S.; Fakhari, A. R. Application of nonionic surfactant as a new method for the enhancement of electromembrane extraction performance for determination of basic drugs in biological samples. J. Chromatogr. A2015, 1378, 1–7.10.1016/j.chroma.2014.11.061Search in Google Scholar PubMed

Hasheminasab, K. S.; Fakhari, A. R.; Shahsavani, A.; Ahmar, H. A new method for the enhancement of electromembrane extraction efficiency using carbon nanotube reinforced hollow fiber for the determination of acidic drugs in spiked plasma, urine, breast milk and wastewater samples. J. Chromatogr. A2013, 1285, 1–6.10.1016/j.chroma.2013.01.115Search in Google Scholar PubMed

Hasheminasab, K. S.; Fakhari, A. R.; Koruni, M. H. Development of carbon-nanotube-assisted electromembrane extraction in the two-phase mode combined with GC for the determination of basic drugs. J. Sep. Sci.2014, 37, 85–91.10.1002/jssc.201300480Search in Google Scholar PubMed

Hidalgo, C. R.; Ramos-Payán, M.; Ocaña-González, J. A.; Martín-Valero, M. J.; Bello-López, M. A. Agar films containing silver nanoparticles as new supports for electromembrane extraction. Anal. Bioanal. Chem.2015, 407, 1519–1525.10.1007/s00216-014-8375-6Search in Google Scholar PubMed

Hosseiny Davarani, S. S.; Sheikhi, N.; Nojavan, S.; Ansari, R.; Mansori, S. Electromembrane extraction of heavy metal cations from aqueous media based on flat membrane: method transfer from hollow fiber to flat membrane. Anal. Methods2015, 7, 2680–2686.10.1039/C5AY00243ESearch in Google Scholar

Hosseiny Davarani, S. S.; Moazami, H. R.; Memarian, E.; Nojavan, S. Electromembrane extraction through a virtually rotating supported liquid membrane. Electrophoresis2016, 37, 339–346.10.1002/elps.201500296Search in Google Scholar PubMed

Hu, Z.; Chen, H.; Yao, C.; Zhu, Y. Determination of inorganic anions in ethyl acetate by ion chromatography with an electromembrane extraction method. J. Chromatogr. Sci.2011, 49, 617–621.10.1093/chrsci/49.8.617Search in Google Scholar PubMed

Huang, C.; Eibak, L. E. E.; Gjelstad, A.; Shen, X.; Trones, R.; Jensen, H.; Pedersen-Bjergaard, S. Development of a flat membrane based device for electromembrane extraction: a new approach for exhaustive extraction of basic drugs from human plasma. J. Chromatogr. A2014, 1326, 7–12.10.1016/j.chroma.2013.12.028Search in Google Scholar PubMed

Huang, C.; Jensen, H.; Seip, K. F.; Gjelstad, A.; Pedersen-Bjergaard, S. Mass transfer in electromembrane extraction-The link between theory and experiments. J. Sep. Sci.2015a, 00, 1–10.10.1002/jssc.201500905Search in Google Scholar PubMed

Huang, C.; Seip, K. F.; Gjelstad, A.; Shen, X.; Pedersen-Bjergaard, S. Combination of electromembrane extraction and liquid-phase microextraction in a single step: simultaneous group separation of acidic and basic drugs. Anal. Chem.2015b, 87, 6951–6957.10.1021/acs.analchem.5b01610Search in Google Scholar PubMed

Huang, C.; Seip, K. F.; Gjelstad, A.; Pedersen-Bjergaard, S. Electromembrane extraction for pharmaceutical and biomedical analysis – Quo vadis. J. Pharm. Biomed. Anal.2015c, 113, 97–107.10.1016/j.jpba.2015.01.038Search in Google Scholar PubMed

Huang, C.; Gjelstad, A.; Pedersen-Bjergaard, S. Selective electromembrane extraction based on isoelectric point: Fundamental studies with angiotensin II antipeptide as model analyte. J. Membr. Sci.2015d, 481, 115–123.10.1016/j.memsci.2015.02.006Search in Google Scholar

Huang, C.; Gjelstad, A.; Pedersen-Bjergaard, S. Exhaustive extraction of peptides by electromembrane extraction. Anal. Chim. Acta2015e, 853, 328–334.10.1016/j.aca.2014.10.017Search in Google Scholar PubMed

Huang, C.; Gjelstad, A.; Seip, K. F.; Jensen, H.; Pedersen-Bjergaard, S. Exhaustive and stable electromembrane extraction of acidic drugs from human plasma. J. Chromatogr. A2015f, 1425, 81–87.10.1016/j.chroma.2015.11.052Search in Google Scholar PubMed

Jamt, R. E. G.; Gjelstad, A.; Eibak, L. E. E.; Øiestad, E. L.; Christophersen, A. S.; Rasmussen, K. E.;Pedersen-Bjergaard, S. Electromembrane extraction of stimulating drugs from undiluted whole blood. J. Chromatogr. A2012, 1232, 27–36.10.1016/j.chroma.2011.08.058Search in Google Scholar PubMed

Jeannot, M. A.; Cantwell, F. F. Solvent microextraction into a single drop. Anal. Chem.1996, 68, 2236–2240.10.1021/ac960042zSearch in Google Scholar PubMed

Kiplagat, I. K.; Doan, T. K. O.; Kubáň, P.; Boček, P. Trace determination of perchlorate using electromembrane extraction and capillary electrophoresis with capacitively coupled contactless conductivity detection. Electrophoresis2011, 32, 3008–3015.10.1002/elps.201100279Search in Google Scholar PubMed

Kjelsen, I. J. Ø.; Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Low-voltage electromembrane extraction of basic drugs from biological samples. J. Chromatogr. A2008, 1180, 1–9.10.1016/j.chroma.2007.12.006Search in Google Scholar PubMed

Koruni, M. H.; Tabani, H.; Gharari, H.; Fakhari, A. R. An all-in-one electro-membrane extraction: development of an electro-membrane extraction method for the simultaneous extraction of acidic and basic drugs with a wide range of polarities. J. Chromatogr. A2014, 1361, 95–99.10.1016/j.chroma.2014.07.075Search in Google Scholar PubMed

Krishna Marothu, V.; Gorrepati, M.; Vusa, R. Electromembrane extraction–a novel extraction technique for pharmaceutical, chemical, clinical and environmental analysis. J. Chromatogr. Sci.2013, 51, 619–631.10.1093/chromsci/bmt041Search in Google Scholar PubMed

Kubáň, P.; Boček, P. Capillary electrophoresis with capacitively coupled contactless conductivity detection: a universal tool for the determination of supported liquid membrane selectivity in electromembrane extraction of complex samples. J. Chromatogr. A2012, 1267, 96–101.10.1016/j.chroma.2012.07.010Search in Google Scholar PubMed

Kubáň, P.; Boček, P. Micro-electromembrane extraction across free liquid membranes. Extractions of basic drugs from undiluted biological samples. J. Chromatogr. A2014a, 1337, 32–39.10.1016/j.chroma.2014.02.046Search in Google Scholar PubMed

Kubáň, P.; Boček, P. Micro-electromembrane extraction across free liquid membranes. Instrumentation and basic principles. J. Chromatogr. A2014b, 1346, 25–33.10.1016/j.chroma.2014.04.047Search in Google Scholar PubMed

Kubáň, P.; Boček, P. The effects of electrolysis on operational solutions in electromembrane extraction: the role of acceptor solution. J. Chromatogr. A2015, 1398, 11–19.10.1016/j.chroma.2015.04.024Search in Google Scholar PubMed

Kubáň, P.; Šlampová, A.; Boček, P. Electric field-enhanced transport across phase boundaries and membranes and its potential use in sample pretreatment for bioanalysis. Electrophoresis2010, 31, 768–785.10.1002/elps.200900561Search in Google Scholar PubMed

Kubáň, P.; Strieglerová, L.; Gebauer, P.; Boček, P. Electromembrane extraction of heavy metal cations followed by capillary electrophoresis with capacitively coupled contactless conductivity detection. Electrophoresis2011, 32,1025–1032.10.1002/elps.201000462Search in Google Scholar PubMed

Lee, J.; Lee, H. K.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Environmental and bioanalytical applications of hollow fiber membrane liquid-phase microextraction: A review. Anal. Chim. Acta2008, 624, 253–268.10.1016/j.aca.2008.06.050Search in Google Scholar PubMed

Lee, J.; Khalilian, F.; Bagheri, H.; Lee, H. K. Optimization of some experimental parameters in the electro membrane extraction of chlorophenols from seawater. J. Chromatogr. A2009, 1216, 7687–7693.10.1016/j.chroma.2009.09.037Search in Google Scholar PubMed

Liu, Y.; Zhang, X.; Guo, L.; Zhang, Y.; Li, Z.; Wang, Z.; Huang, M.; Yang, C.; Ye, J.; Chu, Q. Electromembrane extraction of salivary polyamines followed by capillary zone electrophoresis with capacitively coupled contactless conductivity detection. Talanta2014, 128, 386–392.10.1016/j.talanta.2014.04.079Search in Google Scholar PubMed

Majors, R. E. Electromembrane extraction: the use of electrical potential for isolation of charged substances from biological matrices. LCGC N. Am.2010, 28, 92–112.Search in Google Scholar

Mamat, N. A.; See, H. H. Development and evaluation of electromembrane extraction across a hollow polymer inclusion membrane. J. Chromatogr. A2015, 1406, 34–39.10.1016/j.chroma.2015.06.020Search in Google Scholar PubMed

Middelthon-Bruer, T. M.; Gjelstad, A.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Parameters affecting electro membrane extraction of basic drugs. J. Sep. Sci.2008, 31, 753–759.10.1002/jssc.200700502Search in Google Scholar PubMed

Moazami, H. R.; Nojavan, S.; Zahedi, P.; Davarani, S. S. H. Electronic simulation of the supported liquid membrane in electromembrane extraction systems: improvement of the extraction by precise periodical reversing of the field polarity. Anal. Chim. Acta2014, 841, 24–32.10.1016/j.aca.2014.06.048Search in Google Scholar PubMed

Nojavan, S.; Fakhari, A. R. Electro membrane extraction combined with capillary electrophoresis for the determination of amlodipine enantiomers in biological samples. J. Sep. Sci.2010, 33, 3231–3238.10.1002/jssc.201000242Search in Google Scholar PubMed

Nojavan, S.; Pourahadi, A.; Davarani, S. S. H.; Morteza-Najarian, A.; Beigzadeh Abbassi, M. Electromembrane extraction of zwitterionic compounds as acid or base: comparison of extraction behavior at acidic and basic pHs. Anal. Chim. Acta2012, 745, 45–52.10.1016/j.aca.2012.07.044Search in Google Scholar PubMed

Nojavan, S.; Tahmasebi, Z.; Bidarmanesh, T.; Behdad, H.; Nasiri-Aghdam, M.; Mansori, S.; Pourahadi, A. Electrically enhanced liquid-phase microextraction of three textile azo dyes from wastewater and plant samples. J. Sep. Sci.2013, 36, 3256–3263.10.1002/jssc.201300546Search in Google Scholar PubMed

Nojavan, S.; Bidarmanesh, T.; Memarzadeh, F.; Chalavi, F. Electro-driven extraction of inorganic anions from water samples and water miscible organic solvents and analysis by ion chromatography. Electrophoresis2014, 35, 2446–2453.10.1002/elps.201400072Search in Google Scholar PubMed

Ocaña-González, J. A.; Fernández-Torres, R.; Bello-López, M. A.;Ramos-Payán, M. New developments in microextraction techniques in bioanalysis. A review. Anal. Chim. Acta2016, 905, 8–23.10.1016/j.aca.2015.10.041Search in Google Scholar PubMed

Payan, M. R.; Lopez, M. A. B.; Torres, R. F.; Navarro, M. V.; Mochon, M. C. Electromembrane extraction (EME) and HPLC determination of non-steroidal anti-inflammatory drugs (NSAIDs) in wastewater samples. Talanta2011, 85, 394–399.10.1016/j.talanta.2011.03.076Search in Google Scholar PubMed

Payán, M. D. R.; Li, B.; Petersen, N. J.; Jensen, H.; Hansen, S. H.; Pedersen-Bjergaard, S. Nano-electromembrane extraction. Anal. Chim. Acta2013, 785, 60–66.10.1016/j.aca.2013.04.055Search in Google Scholar PubMed

Pedersen-Bjergaard, S.; Rasmussen, K. E. Liquid-liquid-liquid microextraction for sample preparation of biological fluids prior to capillary electrophoresis. Anal. Chem.1999, 71, 2650–2656.10.1021/ac990055nSearch in Google Scholar PubMed

Pedersen-Bjergaard, S.; Rasmussen, K. E. Electrokinetic migration across artificial liquid membranes: New concept for rapid sample preparation of biological fluids. J. Chromatogr. A2006, 1109, 183–190.10.1016/j.chroma.2006.01.025Search in Google Scholar PubMed

Petersen, N. J.; Pedersen, J. S.; Poulsen, N. N.; Jensen, H.; Skonberg, C.; Hansen, S. H.; Pedersen-Bjergaard, S. On-chip electromembrane extraction for monitoring drug metabolism in real time by electrospray ionization mass spectrometry. Analyst2012, 137, 3321–3327.10.1007/978-1-4939-2353-3_15Search in Google Scholar PubMed

Petersen, N. J.; Pedersen, J. S.; Poulsen, N. N.; Jensen, H.; Skonberg, C.; Hansen, S. H.; Petersen, N. J.; Jensen, H.; Hansen, S. H.; Rasmussen, K. E.; Pedersen-Bjergaard, S. Drop-to-drop microextraction across a supported liquid membrane by an electrical field under stagnant conditions. J. Chromatogr. A2009, 1216, 1496–1502.10.1016/j.chroma.2008.12.079Search in Google Scholar PubMed

Petersen, N. J.; Jensen, H.; Hansen, S. H.; Foss, S. T.; Snakenborg, D.; Pedersen-Bjergaard, S. On-chip electro membrane extraction. Microfluid. Nanofluid.2010, 9, 881–888.10.1007/s10404-010-0603-6Search in Google Scholar

Petersen, N. J.; Rasmussen, K. W.; Pedersen-Bjergaard, S.; Gjelstad, A. Electromembrane extraction from biological fluids. Anal. Sci.2011a, 27, 965–972.10.2116/analsci.27.965Search in Google Scholar PubMed

Petersen, N. J.; Foss, S. T.; Jensen, H.; Hansen, S. H.; Skonberg, C.; Snakenborg, D.; Kutter, J. P.; Pedersen-Bjergaard, S. On-chip electro membrane extraction with online ultraviolet and mass spectrometric detection. Anal. Chem.2011b, 83, 44–51.10.1021/ac1027148Search in Google Scholar PubMed

Rahmani, T.; Rahimi, A.;Nojavan, S. Study on electrical current variations in electromembrane extraction process: relation between extraction recovery and magnitude of electrical current. Anal. Chim. Acta2016, 903, 81–90.10.1016/j.aca.2015.11.024Search in Google Scholar PubMed

Ramos-Payán, M.; Villar-Navarro, M.; Fernández-Torres, R.; Callejón-Mochón, M.; Bello-López, M. A. Electromembrane extraction (EME) – an easy, novel and rapid extraction procedure for the HPLC determination of fluoroquinolones in wastewater samples. Anal. Bioanal. Chem.2013, 405, 2575–2584.10.1007/s00216-012-6664-5Search in Google Scholar PubMed

Ramos-Payán, M.; Fernández-Torres, R.; Pérez-Bernal, J. L.; Callejón-Mochón, M.; Bello-López, M. T. A novel approach for electromembrane extraction based on the use of silver nanometallic-decorated hollow fibers. Anal. Chim. Acta2014, 849, 7–11.10.1016/j.aca.2014.08.020Search in Google Scholar PubMed

Rezaee, M.; Assadi, Y.; Milani Hosseini, M. R.; Aghaee, E.; Ahmadi, F.; Berijani, S. Determination of organic compounds in water using dispersive liquid-liquid microextraction. J. Chromatogr. A2006, 1116, 1–9.10.1016/j.chroma.2006.03.007Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S. Electromembrane extraction of trace amounts of naltrexone and nalmefene from untreated biological fluids. J. Chromatogr. B: Analyt. Technol. Biomed. Life Sci.2011, 879, 1143–1148.10.1016/j.jchromb.2011.03.043Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S. Electrically assisted liquid-phase microextraction for determination of β 2-receptor agonist drugs in wastewater. J. Sep. Sci.2012, 35, 571–579.10.1002/jssc.201100869Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S.; Esrafili, A. One-way and two-way pulsed electromembrane extraction for trace analysis of amino acids in foods and biological samples. Anal. Chim. Acta2013a, 773, 52–59.10.1016/j.aca.2013.02.030Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S.; Ebrahimpour, B. Electromembrane surrounded solid phase microextraction: a novel approach for efficient extraction from complicated matrices. J. Chromatogr. A2013, 1280, 16–22.10.1016/j.chroma.2013.01.034Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S. Microextraction in urine samples for gas chromatography: a review. Bioanalysis2014a, 6, 2663–2684.10.4155/bio.14.215Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S.; Tahmasebi, E.; Rezaei, F. Electromembrane surrounded solid phase microextraction followed by injection port derivatization and gas chromatography-flame ionization detector analysis for determination of acidic herbicides in plant tissue. J. Agric. Food Chem.2014b, 62, 3134–3142.10.1021/jf500017rSearch in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S.; Arjomandi-Behzad, L. Voltage-step pulsed electromembrane as a novel view of electrical field-induced liquid-phase microextraction. J. Chromatogr. A2014c, 1324, 21–28.10.1016/j.chroma.2013.11.034Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S. Application of a new nanocarbonaceous sorbent in electromembrane surrounded solid phase microextraction for analysis of amphetamine and methamphetamine in human urine and whole blood. J. Chromatogr. A2015b, 1396, 1–6.10.1016/j.chroma.2015.03.077Search in Google Scholar PubMed

Rezazadeh, M.; Yamini, Y.; Seidi, S.; Aghaei, A. Pulsed electromembrane extraction for analysis of derivatized amino acids: A powerful technique for determination of animal source of gelatin samples. Talanta2015a, 136, 190–197.10.1016/j.talanta.2015.01.007Search in Google Scholar PubMed

Safari, M.; Nojavan, S.; Davarani, S. S. H.; Morteza-Najarian, A. Speciation of chromium in environmental samples by dual electromembrane extraction system followed by high performance liquid chromatography. Anal. Chim. Acta2013, 789, 58–64.10.1016/j.aca.2013.06.023Search in Google Scholar PubMed

Sanagi, M. M.; Mohamad Hanapi, N. S.; Ismail, A. K.; Wan Ibrahim, W. A.; Saim, N.; Yahaya, N. Two-phase electrodriven membrane extraction combined with liquid chromatography for the determination of tricyclic antidepressants in aqueous matrices. Anal. Methods2014, 6, 8802–8809.10.1039/C4AY01700ESearch in Google Scholar

Schmidt-Marzinkowski, J.; See, H. H.; Hauser, P. C. Electric field driven extraction of inorganic anions across a polymer inclusion membrane. Electroanal.2013, 25, 1879–1886.10.1002/elan.201300176Search in Google Scholar

See, H. H.; Hauser, P. C. Electric field-driven extraction of lipophilic anions across a carrier-mediated polymer inclusion membrane. Anal. Chem.2011, 83, 7507–7513.10.1021/ac201772gSearch in Google Scholar PubMed

See, H. H.; Stratz, S.; Hauser, P. C. Electro-driven extraction across a polymer inclusion membrane in a flow-through cell. J. Chromatogr. A2013, 1300, 79–84.10.1016/j.chroma.2013.01.062Search in Google Scholar PubMed

Seidi, S.; Yamini, Y.; Saleh, A.; Moradi, M. Electromembrane extraction of levamisole from human biological fluids. J. Sep. Sci.2011a, 34, 585–593.10.1002/jssc.201000735Search in Google Scholar PubMed

Seidi, S.; Yamini, Y.; Heydari, A.; Moradi, M.; Esrafili, A.; Rezazadeh, M. Determination of thebaine in water samples, biological fluids, poppy capsule, and narcotic drugs, using electromembrane extraction followed by high-performance liquid chromatography analysis. Anal. Chim. Acta2011b, 701, 181–188.10.1016/j.aca.2011.05.042Search in Google Scholar PubMed

Seidi, S.; Yamini, Y.; Rezazadeh, M.; Esrafili, A. Low-voltage electrically-enhanced microextraction as a novel technique for simultaneous extraction of acidic and basic drugs from biological fluids. J. Chromatogr. A2012, 1243, 6–13.10.1016/j.chroma.2012.04.050Search in Google Scholar PubMed

Seidi, S.; Yamini, Y.; Rezazadeh, M. Combination of electromembrane extraction with dispersive liquid-liquid microextraction followed by gas chromatographic analysis as a fast and sensitive technique for determination of tricyclic antidepressants. J. Chromatogr. B: Analyt. Technol. Biomed. Life Sci.2013, 913914, 138–146.10.1016/j.jchromb.2012.12.008Search in Google Scholar PubMed

Seip, K. F.; Stigsson, J.; Gjelstad, A.; Balchen, M.; Pedersen-Bjergaard, S. Electromembrane extraction of peptides-Fundamental studies on the supported liquid membrane. J. Sep. Sci.2011, 34, 3410–3417.10.1002/jssc.201100558Search in Google Scholar PubMed

Seip, K. F.; Gjelstad, A.; Pedersen-Bjergaard, S. The potential application of electromembrane extraction for the analysis of peptides in biological fluids. Bioanalysis2012, 4, 1971–1973.10.4155/bio.12.163Search in Google Scholar PubMed

Seip, K. F.; Jensen, H.; Sønsteby, M. H.; Gjelstad, A.; Pedersen-Bjergaard, S. Electromembrane extraction: distribution or electrophoresis? Electrophoresis2013a, 34, 792–799.10.1002/elps.201200587Search in Google Scholar PubMed

Seip, K. F.; Gjelstad, A.; Pedersen-Bjergaard, S. Electromembrane extraction from aqueous samples containing polar organic solvents. J. Chromatogr. A2013b, 1308, 37–44.10.1016/j.chroma.2013.07.105Search in Google Scholar PubMed

Seip, K. F.; Faizi, M.; Vergel, C.; Gjelstad, A.; Pedersen-Bjergaard, S. Stability and efficiency of supported liquid membranes in electromembrane extraction – a link to solvent properties Microextraction Techniques. Anal. Bioanal. Chem.2014a, 406, 2151–2161.10.1007/s00216-013-7418-8Search in Google Scholar PubMed

Seip, K. F.; Jensen, H.; Kieu, T. E.; Gjelstad, A.; Pedersen-Bjergaard, S. Salt effects in electromembrane extraction. J. Chromatogr. A2014b, 1347, 1–7.10.1016/j.chroma.2014.04.053Search in Google Scholar PubMed

Seip, K. F.; Gjelstad, A.; Pedersen-Bjergaard, S. The potential of electromembrane extraction for bioanalytical applications. Bioanalysis2015, 7, 463–480.10.4155/bio.14.303Search in Google Scholar PubMed

Šlampová, A.; Kubáň, P.; Boček, P. Additional considerations on electrolysis in electromembrane extraction. J. Chromatogr. A2007, 1429, 364–368.10.1016/j.chroma.2015.12.028Search in Google Scholar PubMed

Šlampová, A.; Kubáň, P.; Boček, P. Electromembrane extraction using stabilized constant d.c. electric current–a simple tool for improvement of extraction performance. J. Chromatogr. A2012, 1234, 32–37.10.1016/j.chroma.2011.11.035Search in Google Scholar PubMed

Šlampová, A.; Kubáñ, P.; Boćek, P. Determination of chlorophenols in environmental samples using electromembrane extraction and capillary electrophoresis. Chem. Listy2013, 107, 447–448.Search in Google Scholar

Šlampová, A.; Kubáň, P.; Boček, P. Effects of selected operational parameters on efficacy and selectivity of electromembrane extraction: chlorophenols as model analytes. Electrophoresis2014a,35, 2429–2437.10.1002/elps.201400096Search in Google Scholar PubMed

Šlampová, A.; Kubáň, P.; Boček, P. Fine-tuning of electromembrane extraction selectivity using 18-crown-6 ethers as supported liquid membrane modifiers. Electrophoresis2014b, 35, 3317–3320.10.1002/elps.201400372Search in Google Scholar PubMed

Šlampová, A.; Kubáň, P.; Boček, P. Quantitative aspects of electrolysis in electromembrane extractions of acidic and basic analytes. Anal. Chim. Acta2015, 887, 92–100.10.1016/j.aca.2015.06.040Search in Google Scholar PubMed

Soltani, S.; Jouyban, A. Biological sample preparation: attempts on productivity increasing in bioanalysis. Bioanalysis2014, 6, 1691–1710.10.4155/bio.14.118Search in Google Scholar PubMed

Song, X. Y.; Chen, J.; Shi, Y. P. Electromembrane extraction based on carbon nanotubes reinforced hollow fiber for the determination of plant hormones. New J. Chem.2015, 39, 9191–9199.10.1039/C5NJ01683ESearch in Google Scholar

Strieglerová, L.; Kubáň, P.; Boček, P. Rapid and simple pretreatment of human body fluids using electromembrane extraction across supported liquid membrane for capillary electrophoretic determination of lithium. Electrophoresis2011a, 32, 1182–1189.10.1002/elps.201000620Search in Google Scholar PubMed

Strieglerová, L.; Kubáň, P.; Boček, P. Electromembrane extraction of amino acids from body fluids followed by capillary electrophoresis with capacitively coupled contactless conductivity detection. J. Chromatogr. A2011b, 1218, 6248–6255.10.1016/j.chroma.2011.07.011Search in Google Scholar PubMed

Suh, J. H.; Eom, H. Y.; Kim, U.; Kim, J.; Cho, H. D.; Kang, W.; Kim, D. S.; Han, S. B. Highly sensitive electromembrane extraction for the determination of volatile organic compound metabolites in dried urine spot. J. Chromatogr. A2015a, 1416, 1–9.10.1016/j.chroma.2015.09.004Search in Google Scholar PubMed

Suh, J. H.; Lee, H. Y.; Kim, U.; Eom, H. Y.; Kim, J.; Cho, H. D.; Han, S. B. Simultaneous determination of benzene, toluene, ethylbenzene, and xylene metabolites in human urine using electromembrane extraction combined with liquid chromatography and tandem mass spectrometry. J. Sep. Sci.2015b, 38, 4276–4285.10.1002/jssc.201500969Search in Google Scholar PubMed

Sun, J. N.; Chen, J.; Shi, Y. P. Ionic liquid-based electromembrane extraction and its comparison with traditional organic solvent based electromembrane extraction for the determination of strychnine and brucine in human urine. J. Chromatogr. A2014, 1352, 1–7.10.1016/j.chroma.2014.05.037Search in Google Scholar PubMed

Sun, J. N.; Shi, Y. P.; Chen, J. Development of ionic liquid based electromembrane extraction and its application to the enrichment of acidic compounds in pig kidney tissues. RSC Advances2015, 5, 37682–37690.10.1039/C5RA01029BSearch in Google Scholar

Tabani, H.; Fakhari, A. R.; Zand, E. Low-voltage electromembrane extraction combined with cyclodextrin modified capillary electrophoresis for the determination of phenoxy acid herbicides in environmental samples. Anal. Methods2013a, 5, 1548–1555.10.1039/c3ay26252aSearch in Google Scholar

Tabani, H.; A. R.; Shahsavani, A. Simultaneous determination of acidic and basic drugs using dual hollow fibre electromembrane extraction combined with CE. Electrophoresis2013b, 34, 269–276.10.1002/elps.201200330Search in Google Scholar PubMed

Tabani, H.; Fakhari, A. R.; Shahsavani, A.; Behbahani, M.; Salarian, M.; Bagheri, A.; Nojavan, S. Combination of graphene oxide-based solid phase extraction and electro membrane extraction for the preconcentration of chlorophenoxy acid herbicides in environmental samples. J. Chromatogr. A2013c, 1300, 227–235.10.1016/j.chroma.2013.04.026Search in Google Scholar PubMed

Tabani, H.; Fakhari, A. R.; Shahsavani, A.; Gharari Alibabaou, H. Electrically assisted liquid-phase microextraction combined with capillary electrophoresis for quantification of propranolol enantiomers in human body fluids. Chirality2014, 26, 260–267.10.1002/chir.22308Search in Google Scholar PubMed

Tak, V.; Kabra, A.; Pardasani, D.; Goud, D. R.; Jain, R.; Dubey, D. K. A glass capillary based microfluidic electromembrane extraction of basic degradation products of nitrogen mustard and VX from water. J. Chromatogr. A2015, 1426, 16–23.10.1016/j.chroma.2015.11.023Search in Google Scholar PubMed

Yamini, Y.; Seidi, S.; Pourali, A.; Rezazadeh, M. Electrical field-stimulated liquid-phase microextraction for trace analysis of pyridine and its derivatives in cigarette extract. J. Iran. Chem. Soc.2015, 12, 503–511.10.1007/s13738-014-0507-6Search in Google Scholar

Zahedi, P.; Davarani, S. S. H.; Moazami, H. R.; Nojavan, S. Surfactant assisted pulsed two-phase electromembrane extraction followed by GC analysis for quantification of basic drugs in biological samples. J. Pharm. Biomed. Anal.2016, 117, 485–491.10.1016/j.jpba.2015.10.002Search in Google Scholar PubMed

Zhang, X.; Guo, L.; Zhang, D.; Ge, X.; Ye, J.; Chu, Q. Sensitive determination of bromate in water samples by capillary electrophoresis coupled with electromembrane extraction. Food Anal. Methods2015a, 9, 393–400.10.1007/s12161-015-0208-5Search in Google Scholar

Zhang, X.; Zhang, H.; Liu, Y.; Guo, L.; Ye, J.; Chu, Q. Sensitive determination of five priority haloacetic acids by electromembrane extraction with capillary electrophoresis. Chin. J. Chem.2015b, 33, 235–240.10.1002/cjoc.201400633Search in Google Scholar

Received: 2016-3-5
Accepted: 2016-7-1
Published Online: 2016-9-1
Published in Print: 2016-12-1

©2016 Walter de Gruyter GmbH, Berlin/Boston

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Downloaded on 28.3.2024 from https://www.degruyter.com/document/doi/10.1515/revac-2016-0008/html
Scroll to top button