Deep Eutectic Solvents For Pharmaceutical Formulation and Drug Delivery Applications

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY

2020, VOL. 25, NO. 7, 779–796


https://doi.org/10.1080/10837450.2020.1735414

REVIEW ARTICLE

Deep eutectic solvents for pharmaceutical formulation and drug delivery


applications
Shahram Emamia and Ali Shayanfarb
a
Department of Pharmaceutics, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran; bPharmaceutical Analysis Research
Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran

ABSTRACT ARTICLE HISTORY


Solvents play key roles in designing drug delivery systems (DDSs). They are used as the reaction Received 15 July 2019
media in the preparation of DDSs and as vehicles for delivery of problematic drugs. The number of Revised 22 February 2020
pharmaceutically acceptable solvents is limited and developing new green ones is of a great of inter- Accepted 24 February 2020
est. A deep eutectic solvent (DES) is a room temperature liquid composed of a mixture of hydrogen
KEYWORDS
bond acceptor and hydrogen bond donor. DESs exhibit interesting physical and chemical characteris- Deep eutectic solvents;
tics such as low vapor pressure, non-inflammability, chemically tailorable, solvency power for a wide active pharmaceutical
range of solutes, and water non-reactivity. Furthermore, they can be easily prepared from low tox- ingredient; drug delivery;
icity, readily available, and inexpensive constituents. Due to these properties, DESs have attracted solubility; nanomaterials;
growing attention as green solvents in different areas of science from chemistry to material engineer- self-assembly; toxicity
ing and biology. In this review, after a brief introduction about DESs, we provide an overview about
current advances made over the last decade in utilizing of DESs for solubilization of water insoluble
drugs, transdermal drug delivery, inorganic nanoparticle synthesis, and designing polymeric and self-
assembled drug carriers. This paper also discusses main challenges and limitations of DESs that
should be considered before using of them.

1. Introduction -biocompatibility, and -sustainability (Egorova and Ananikov 2014;


Amde et al. 2015; Kudłak et al. 2015).
Pharmaceutical production processes require solvents and
In 2003, Abbott et al. (2003) dissolved a variety of solutes in
pharmaceutical industry is one the biggest consumers of organic
the room temperature liquids resulted from mixing two solid
solvents (Grodowska and Parczewski 2010). Organic solvents are
organic materials; urea and a quaternary ammonium salt at a 2:1
involved in synthesis, purification, crystallization, and formulation
molar ratio. The authors employed the term of ‘deep eutectic sol-
processes of active pharmaceutical ingredients (APIs).
vents (DESs)’ to name the formed liquids and suggested DESs as
Furthermore, solvents play the role of vehicles in designing drug
sustainable, biodegradable, and chemically tailorable solvents.
delivery systems (DDSs), for example for parenteral and transder- Since this pioneer work, a rapidly growing interest has been
mal delivery of water-insoluble drugs (Mottu et al. 2000). devoted to DESs because of their unique physical and chem-
However, organic solvents are volatile, toxic, flammable, and have ical properties.
limited solvency (Alonso et al. 2016). Thus, researchers attempt to Due to the similarities between physical characteristics of DESs
find more benign alternatives to problematic organic solvents. and ILs, DESs are often erroneously categorized as a subclass of
The attempts to discover green and designer solvents brought ILs. However, in contrast to ILs that are composed of a pair of
ionic liquids (Wilkes and Zaworotko 1992). Ionic liquids (ILs) are large asymmetric organic cation and inorganic or organic anion,
organic salts with melting points less than 100  C and some of DESs are mixtures of natural molecules. These two different types
them are in liquid state at room temperature (Marrucho et al. of solvents share common properties including low vapor pres-
2014). Ionic liquids exhibit unique characteristics such as low sure, high viscosity, non-inflammability, wide liquid-range, chem-
vapor pressure, non-inflammability, wide liquid-range, chemically ically tailorable, solvency power for a wide range of solutes, and
tailorable, solvency power for a wide range of solutes, and chem- chemical and thermal stability. Despite these similar features,
ical and thermal stability (Mizuuchi et al. 2008). Due to these fea- advantages of DESs over ILs such as water nonreactivity, low- or
tures, ILs have been employed extensively as solvents, cosolvents, nontoxicity, biodegradability, ease of preparation, and inexpensive
and/or materials in different disciplines of chemistry (Plechkova and readily available constituents suggest DESs as versatile alter-
and Seddon 2008) and biology (Egorova et al. 2017). In the field natives to ILs (Smith et al. 2014).
of drug delivery, they have been studied for enhancing transder- In this review, first, we give a brief introduction about DESs.
mal permeability (Moniruzzaman et al. 2010), improving solubility Then we discuss the prominent advances that have been made
(Williams et al. 2014), and developing liquid APIs (Keramatnia over the recent years in the applications of DESs in the different
et al. 2016; Shayanfar and Shayanfar 2018). Nevertheless, toxicity areas of drug formulation. The paper covers designing polymeric
studies on ILs have reported high toxicity, poor-biodegradability, drug delivery systems, delivery of water insoluble APIs, dermal

CONTACT Shahram Emami emami.sh@umsu.ac.ir; shahramemami67@gmail.com Department of Pharmaceutics, School of Pharmacy, Urmia University of
Medical Science, Urmia 571478334, Iran
ß 2020 Informa UK Limited, trading as Taylor & Francis Group
780 S. EMAMI AND A. SHAYANFAR

Figure 1. Formation of a deep eutectic solvent (DES) between choline chloride and urea. HBA: hydrogen bond acceptor, HBD: hydrogen bond donor, Tm:
melting point.

Table 1. Different types of DESs. industrial scale production feasible without need to complex
Type Composition Example facilities. Most researchers have prepared DESs by direct mixing
I Organic salt þ metal salt Choline chloride–ZnCl2(1:2) with gentle heating (Tang and Row 2013). This method does not
II Organic salt þ hydrated metal salt Choline chloride–CaCl2.6H2O (2:1) need solvents and produces no waste and by-products. For
III Organic salt þ hydrogen-bond donor Choline chloride–urea (1:2)
IV Metal chloride þ hydrogen-bond donor ZnCl2–urea (2:7)
thermo-sensitive materials, one can dissolve the components in a
V Nonionic hydrogen-bond acceptor Citric acid–glucose (1:1) suitable solvent then remove the solvent by evaporation (Dai
þ hydrogen-bond donor et al. 2013) or freeze drying (Gutierrez et al. 2009).

1.1.2. Natural deep eutectic solvents


and transdermal delivery, self-assembly of amphiphilic molecules In 2011, Choi et al. (2011) hypothesized that primary plant metab-
in DESs, and synthesis of inorganic nanomaterials. Finally, we dis- olites may form a liquid medium other than two immiscible liquid
cuss some of the main challenges and limitations of DESs that phases of plant cells (water and lipids). This third liquid phase
should be considered before using of DESs. may be involved in certain biosynthetic steps or storage of water
insoluble products. The authors prepared a 30 set of novel DESs
containing natural cellular constituents including sugars, carbox-
1.1. Deep eutectic solvents ylic acids, aminoacids, choline, and water and named the resulted
A DES is a room temperature liquid composed of a mixture of hydro- DESs as natural deep eutectic solvents (NADES). Since then, more
gen bond acceptor (HBA) and hydrogen bond donor (HBD) compo- than 150 NADES composed of sugars, sugar alcohols, polyalco-
nents at a specific molar ratio (Smith et al. 2014). The pure hols, organic acids and bases, and amino acids and also ternary
components are typically solid at ambient condition but formation NADES have been reported (Liu et al. 2018). Due to their natural
of strong intermolecular interactions mainly as hydrogen bonds leads origin, NADESs are considered as non-toxic and biodegrable and
to significant melting point depression of the mixture (Figure 1). environmentally benign alternatives for ILs and DESs.
Based on the compositions, Smith et al. (2014) classified DESs into
four types (types I–IV of Table 1). However, some recently developed 1.1.3. Active pharmaceutical ingredient based DESs
DESs could not belong to the types I–IV. Therefore, we added Type V Many APIs have hydrogen-bond donors or acceptor groups in their
to Table 1 to cover all DESs discussed in the literature. It is worth not- structures; therefore, eutectic systems can be developed composed
ing that in addition to binary mixtures, DESs can also originate from of one or more API. If the formed eutectic systems melt at temper-
three components (Liu et al. 2014; Zhang et al. 2016). Types III and V atures lower than room temperature they are named as API based
include most of the studied DESs in the field of pharmaceutics DESs or Simply API-DESs (Abbott et al. 2017). API-DESs can be
because of their advantages such as power to dissolve chemically formed between an API and excipient or between two APIs.
diverse compounds, low-toxicity, large number of possible combina- Examples of reported API-DESs are ibuprofen–menthol (Aroso et al.
tions, nonreactivity with water, and biodegradability. 2015), lidocaine–1,8 octanediol (Serrano et al. 2012), lidocaine–prilo-
Figure 2 presents examples of common HBAs and HBDs to caine (Brodin et al. 1984), ranitidine hydrochloride–urea, and aspir-
prepare DESs. The structure of HBA and HBD and their molar ratio in–choline chloride (Abbott et al. 2017). API-DESs, also called
determine the physicochemical characteristics of a DES (Radosevic therapeutic deep eutectic systems, have been used to design poly-
et al. 2016). Therefore, a tailor made DES can be customized for a meric drug delivery system (Serrano et al. 2012), overcome poly-
certain purpose by choosing appropriate ratios of HBAs and morphism (Abbott et al. 2017), enhance dissolution rate (Aroso
HBDs. Theoretically around 106 DESs can form by combining com- et al. 2016), increase membrane permeability (Santos et al. 2018),
ponents that show eutectic behavior and many have been devel- and improve transdermal delivery (Stott et al. 1998). Until now,
oped in last few years (Francisco et al. 2013). This huge number most of the research conducted on API-DESs has focused on trans-
of combinations makes DESs as designer solvents. Furthermore, a dermal delivery of APIs using API-DESs, therefore we will discuss
third component such as water can be added to optimize the this topic in a separate sub-section (Section 2.5.1).
properties of the DES.
2. Applications of DESs in drug delivery fields
1.1.1. Preparation of DESs
In contrast to ILs, DESs have an easy way of synthesis because To achieve the desired therapeutic effects, APIs must efficiently
their synthesis involves no chemical reaction (salt formation) and specifically reach to their sites of action. However, transport-
between HBA and HBD constituents. This simplicity makes ing APIs from the administration point to the site of action is
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 781

Figure 2. Structures of some hydrogen bond acceptors and hydrogen bond donors utilized in the preparation of DESs.

often hampered by poor biopharmaceutical properties such as functional additive. By using DESs, synthesis can occur in mild
low solubility in body fluids, low permeability through biological conditions (lower temperature, biocompatible media). In addition,
membranes, chemical instability, and rapid metabolism and elim- availability of numerous nontoxic combinations makes it possible
ination from the body (Emami et al. 2018). To address the men- to modulate DES properties and provide optimized condition for
tioned problems, APIs are formulated as various types of drug each reaction. Therefore, biocompatible and environmentally
delivery systems such as solid dispersions (Serajuddin 1999), coc- friendly synthesis methods can be designed (del Monte et al.
rystals (Emami et al. 2019), and polymeric delivery systems 2014). Some excellent reviews have been published on applica-
(Kumari et al. 2010). In the following section, we attempt to tions of DESs as reaction media for synthesis of different types of
briefly overview potentials of DESs to improve drug delivery. materials (Carriazo et al. 2012; del Monte et al. 2014; Alonso et al.
2016; Mota-Morales et al. 2018).
2.1. Designing polymeric drug delivery systems by DESs
2.1.1. DESs as media for synthesis of polymers
Polymeric carriers improve efficacy of drug delivery by controlling Probably the first attempt to design polymeric carrier by DESs
release, providing cell- and tissue-specific targeted delivery, and was polycondensation of citric acid with 1,8-octanediol to synthe-
protecting from degradations (Tibbitt et al. 2016). Natural (Yang size poly(diol-co-citrate) (Serrano et al. 2012). Poly(diol-co-citrate),
et al. 2015), semi-synthetic (Qi et al. 2015), and synthetic (Englert a biodegradable and biocompatible elastomer, can function as a
et al. 2018; Selselehjonban et al. 2019) polymers are used to tissue-engineering scaffold (Prabhakaran et al. 2012) and shape-
design DDSs. However, there are remaining challenges in green memory polymer for temperature-controlled drug delivery
synthesis, processing, and efficient loading of drugs in poly- (Serrano et al. 2011). The routine synthesis of this elastomer needs
meric carriers. a very high temperature (several minutes at 160–165  C followed
In the last decade, DESs have emerged as alternative reaction by 60 min at 140  C) which can lead to thermal instability of APIs
media for synthesis of functional materials such as nanomaterials intended to be loaded in it. Difficulty in homogeneous loading of
(Chen et al. 2013), metal-organic frameworks (Zhang et al. 2009), high amounts of APIs in the prepared elastomer can pose more
and biocompatible polymers (Sanchez-Leija et al. 2014; challenge to conventional methods. To overcome these obstacles,
Pradeepkumar et al. 2018), as well as enzymatic reactions (Durand Serrano et al. (2012) introduced a DES-assisted synthesis method
et al. 2012). DESs can provide green media for polymerization to prepare poly(octanediol-co-citrate) elastomers containing high
reactions to occur where DESs may act as solvent, monomer, or loading of lidocaine (Figure 3). First, they prepared lidocaine–1,8-
782 S. EMAMI AND A. SHAYANFAR

Figure 3. Schematic representation of the preparation of lidocaine loaded poly(diol-co-citrate) elastomer by using DES assisted synthesis method. Tm represents melt-
ing point.

octanediol (1:1) API-DES with a melting point of 40  C. Then citric carriers (Mondal et al. 2013). The ability of DESs to dissolve vari-
acid was dissolved in the liquid API-DES. By increasing the tem- ous types of biopolymers ranging from DNA to proteins and poly-
perature of the resulted solution to about 90  C, polycondensation saccharides have suggested them as processing media for these
reaction was started and lidocaine loaded elastomer was formed. valuable materials and some biopolymer-based functional materi-
In the mentioned example, DES-assisted synthesis resulted in a als have been developed in DESs (Mondal et al. 2013; Sharma
homogenous and high drug loading and protected the thermoli- et al. 2013).
able lidocaine from degradation. In recent decade, DNA, owing to its unique physicochemical
With the aim of preparing antimicrobial wound dressings, properties, have been introduced as a useful tool for designing
Garcıa-Arg€uelles et al. (2013) attempted to incorporate high con- materials with desired properties (Han et al. 2017). Mukesh and
centrations of antibacterial compounds into networks of poly(diol- Prasad (2015) used a choline chloride–ethylene glycol (1:2) to pre-
co-citrate) polyester by using DES-assisted polymerization. To this pare different structural formats of DNA including pH reversible
means, first, they prepared API-DESs of antimicrobial agents ion gel having aggregated DNA structures, pH responsive DNA
including tetraethylammonium bromide, hexadecyltrimethylam- micro-hydrogels, and spheroid-shaped DNA micro-structured
monium bromide, and methyl triphenyl phosphonium bromide assemblies. The formed structures would find applications in drug
with 1,8-octanediol. Then citric acid was dissolved in the resulted and gen delivery purposes.
liquids. Finally, polymerization was performed by thermal cross DNA-hybrid nanomaterials can be used for gene- and drug-
linking at 80  C. Resulting biodegradable polysters were cytocom- delivery and also as antimicrobial agents (Bhatt et al. 2015; Zhang
patible with murine L929 fibroblasts and inhibited Escherichia coli et al. 2015). Hybrid materials of DNA with metal nanoparticles
bacteria growth on solid agar. based on iron, gold, and silver have been prepared using DESs as
In another study, 2-hydroxyethylmethacrylate (HEMA), a com- reaction media. As an example, a DNA based material exhibiting
monly used monomer to produce biocompatible polymers, self- magnetic and antibacterial properties was prepared in a DES com-
polymerized in choline chloride–fructose (2:1) in the presence of posed of choline chloride–ethylene glycol (1:2) (Mondal et al.
indomethacin, a model anti-inflammatory drug (Mukesh et al. 2014). For this mean, iron oxide nanoparticles and protonated lay-
2016). The API was immobilized in the formed poly(2-hydroxye- ered dititanate sheets were added to the solution of DNA in the
thylmethacrylate) ion gel and the immobilized API was stable up DES. Then the dual functionalized DNA based material was pre-
to 6 months at room temperature. Drug release studies showed cipitated by isopropyl alcohol (Figure 4). In the same context,
that the formed ion gel was stable in gastrointestinal fluid pH Bhatt et al. (2016) used choline chloride–ethylene glycol (1:2) DES
and showed pH dependent and sustained drug release. In add- for functionalizing of DNA with iron oxide nanoparticles and N-
ition, in vitro cell viability and hemolysis assays showed noncyto- doped graphene. These hybrid martials of DNA can be used for
toxic and hemocompatible nature of the ion gel. drug and gen delivery applications.
In addition to DNA, polysaccharides can also be processed in
2.1.2. DESs as media to process biopolymers DESs. Chitin is an amino-polysaccharide and is obtained from the
Naturally occurring polymers, or simply biopolymers, are derived exoskeletons of shellfish and insects (Rinaudo 2006). This abun-
from living organisms and include proteins, polysaccharides, and dant natural polymer is a promising carrier for wound care and
nucleic acids. Biodegradability, biocompatibility, and low immuno- drug delivery due to its advantages such as, being mucoadhesive
genicity of biopolymers make them promising carriers for drug and low toxicity (Mi et al. 2003; Madhumathi et al. 2010).
delivery applications (Talebian et al. 2018). However, low solubility However, the application of chitin is limited mainly because of its
of most biopolymers in conventional solvents significantly limits insolubility in water and conventional organic solvents (Sharma
the development of functional materials based on these versatile et al. 2013). In a study, DES of choline chloride–thiourea (1:2) was
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 783

Figure 4. Schematic representation of the preparation of the DNA based hybrid material in choline chloride–ethylene glycol (1:2).

Figure 5. Scanning electron microscopy images of star-, snowflake-, and thorn-shaped gold nanoparticles synthesized in choline chloride–urea (1:2) by varying the
water content in the DES. Pure choline chloride–urea (1:2): snowflake-shaped, 5000 ppm of water in the DES: star-shaped, and more than 10 000 ppm of water in the
DES: thorn-shaped. The figure was adapted with permission from (Liao et al. 2008). Copyright (2008) Wiley-VCH.

successfully used to prepare chitin nanofibers with diameter of ambient condition (Figure 5). Since then, DESs have been used
20–30 nm (Mukesh et al. 2014). By using these nanofibers, the for decorating various shape and size controlled nanomaterials
authors were able to prepare bionanocomposite gel beads of cal- with pharmaceutical interests including, calcium phosphate nano-
cium alginate containing 5-flurouracil, an antitumor drug. particles (Karimi et al. 2016), iron oxide magnetic nanoparticles
Addition of chitin nanofibers to calcium alginate beads improved (Chen et al. 2013), Prussian blue nanoparticles (Sheng et al. 2012),
the elasticity of beads and provided a higher released drug in a silver nanoparticles (Bhatt et al. 2015), and carbon nanomaterials
sustained manner in comparison to calcium alginate beads. (Abo-Hamad et al. 2017).
In addition to synthesis, DESs can also be utilized for function-
alizing nanomaterials to give them particular properties.
2.2. DESs in the synthesis of inorganic nanomaterials
Functionalization is the process of modifying the surface of nano-
Inorganic nano-sized materials can be used in imaging or targeted materials by adding desired functional groups through physical or
drug delivery and have found undeniable position in detection chemical bonding (Emadi et al. 2017). DESs have been studied as
and treatment of diseases (Liang et al. 2014). An emerging area of functionalizing agents for carbon nanotube (Abo-Hamad et al.
application of DESs is as media for designing/synthesis of desired 2017) and graphene (Zainal-Abidin et al. 2019). Graphene is a
nanomaterials. Some review articles have been published on promising drug nanocarrier but its drug delivery applications have
applications of DESs in nanotechnology (Wagle et al. 2014; Abo- been hampered due to the cellular toxicity (Guo and Mei 2014).
Hamad et al. 2015; Tome et al. 2018). In this section, we briefly Surface modification with DES has been successfully utilized to
discuss the potential applications of DESs for designing tailor improve biocompatibility, reduce cellular toxicity, and enhance
made nanomaterials with pharmaceutical interests. drug loading of graphene (Zainal-Abidin et al. 2020). For instance,
The first report on application of DESs in this area was by Liao Zainal-Abidin et al. (2019) utilized choline chloride–urea (1:2), cho-
et al. (2008) in 2008. The authors synthesized various shapes of line chloride–glucose (2:1), choline chloride–fructose (2:1), and
gold nanoparticles in choline chloride–urea (1:2) by simply varying choline chloride–malonic acid (1:1) to modify the surface of gra-
the water content in the DES without seeds or stabilizer at phene by sonication of the suspension of oxidized graphene in
784 S. EMAMI AND A. SHAYANFAR

DESs for 3 h at 60  C. The functionalization was confirmed by dissolving of phenolic antioxidants in DESs, as alternatives for
Fourier transform infrared spectroscopy and Raman spectroscopy. DMSO or ethanol, could result in higher antioxidant activities in
Results indicated that not all of the studied DESs could improve fibroblast cell model (Durand et al. 2017). The improvement
the biocompatibility of oxidized graphene. Indeed, oxidized gra- mechanisms probably were through an enhancement of cell
phene modified with choline chloride–urea (1:2), choline chlori- membrane permeation capacity, or an optimization of activity
de–glucose (2:1), and choline chloride–fructose (2:1) were more within the intracellular medium.
toxic to some studied cell lines than untreated oxidized graphene. In the same line, Shamseddin et al. (2017) studied suitability of
The best result obtained for choline chloride–malonic acid (1:1). various DESs to dissolve resveratrol and compared in vitro
Graphene oxide modified with this DES showed lower toxicity in pharmacological activities of the compound dissolved in the
human cell lines and higher loading of tamoxifen compared to selected DES with resveratrol dissolved in DMSO. Resveratrol, a
untreated graphene oxide. The authors attributed the observed powerful antioxidant, is a low-water soluble compound with low
results to differences in added functional groups by investi- oral bioavailability (Pujara et al. 2017). Among the studied DESs,
gated DESs. 1,2-propanediol–choline chloride–water (1:1:1) showed the lowest
cytotoxicity and exhibited highest solvency for resveratrol.
Resveratrol dissolved in this DES could show ten times higher
2.3. Applications of DESs in delivery of poorly water
in vitro pharmacological activity when compared with the com-
soluble APIs
pound dissolved in DMSO. This higher activity in the presence of
Aqueous solubility/dissolution rate plays an important role DES was attributed to improvements in resveratrol diffusion,
through all stages of drug discovery and development process; transport and, cellular uptake.
from early pharmacological and toxicological evaluations of drug
candidates to development of effective pharmaceutical product 2.3.2. DESs as carriers for solubilization of poorly soluble drugs
(Williams et al. 2013; Emami et al. 2015). It is important because In 2009, Morrison et al. (2009) introduced, for the first time, the
only solubilized form of a compound can transport through bio- use of DESs as drug solubilization vehicles. Since then, several
logical membranes and exhibit pharmacological effect. The num- studies have explored the potential of DESs as pharmaceutical sol-
ber of drug candidates with poor aqueous solubility is increasing vents (Table 2). Until now, most of the studies have used choline
and it is estimated that about 90% of drug candidates in develop- chloride based DESs probably because they are the most common
ment pipelines exhibit low aqueous solubility (Thayer 2010). DESs. As it can be seen in Table 2, solubility of poorly soluble
During drug discovery programs, dimethyl sulfoxide (DMSO) is APIs in DESs can dramatically be higher by 6- to 2200-fold com-
widely used as the solvent of water insoluble drug candidates for pared to their water solubility. Generally, a solvent must be able
biological in vitro and in vivo studies (Santos et al. 2003). to dissolve greater than 1 mg/mL of an API to be utilized in
However, recent studies have questioned the safety of DMSO pharmaceutical processing (Marrucho et al. 2014). Considering this
even at low concentrations (Galvao et al. 2014; Li et al. 2016). criteria, DESs can play an important role as solvents and cosol-
Therefore, it is desirable to find nontoxic and biocompatible alter- vents in drug delivery applications.
natives for DMSO. Lu et al. (2016) reported the solubility of aspirin, acetamino-
In addition to challenges in pharmacological tests, when phen, naproxen, ketoprofen, and ibuprofen in 17 types of DESs.
administered through the oral route, poorly soluble drugs usually The studied APIs could be dissolved 17- to 5477 times better by
face with problems such as large dose, insufficient bioavailability, DESs than by water. In addition to DES composition, solubility
delayed onset of action, and erratic absorption (Pouton 2006). also was dependent to molar ratios of the components of DES. All
About 40% of marketed drugs are poorly water soluble (Takagi five drugs showed the highest solubility in tetra propyl ammo-
et al. 2006). Therefore, finding efficient ways to improve solubility nium bromide–propylene glycol (1:2). The authors could not find
and dissolution rate is essential for translating promising new meaningful correlations between drug solubility and physico-
drug candidates into effective therapies and even improving effi- chemical properties of DESs including viscosity, polarity, and pH.
cacy of marketed drugs. Also to be mentioned is the work of Palmelund et al. (2019), who
measured solubilities of 11 APIs in water, conventional pharma-
2.3.1. DESs as solvents for pharmacological evaluations of ceutical solvents, and six DESs with different HBAs and HBDs. The
drug candidates results showed that for water insoluble APIs such as indometh-
An ideal vehicle/solvent for in vitro and in vivo bioactivity tests of acin, aprepitant, celecoxib, cinnarizine, and flufenamic acid the
candidate drug molecules must be nontoxic, inert, and possess solubility was higher in a DES than in water. As an example, solu-
high solvation properties. Recently some studies have explored bility of celecoxib was about 104 times higher in choline chlori-
DESs as suitable solvents for in vitro pharmacological studies of de–lactic acid–water (1:0.9:0.6) than water. Furthermore, for some
drug candidates. In this context, Rozema et al. (2015) investigated of the studied APIs, the solubility was higher in a DES than com-
the possibility of dissolving salsalate, a poorly water-soluble monly used pharmaceutical solvents; glycerin, ethanol, and poly-
compound, in a DES composed of choline chloride–propylene gly- ethylene glycol 300. These data suggest that DESs may be useful
col–water (1:1:1) as an alternative to DMSO for in vitro pharmaco- as carriers for solubilization of poorly soluble drugs. Many more
logical tests. When this DES was used as the solvent, salsalate of such head to head comparisons should be performed between
remained soluble in the aqueous medium of cell culture in higher solubility of APIs in DESs and conventional pharmaceutical sol-
concentrations in comparison to DMSO. These higher dissolved vents to evaluate real potential of DESs.
concentrations allowed authors to generate full dose-response The chemical structure of the components of a DES and their
curves for salsalate. In addition, as DMSO, the studied DES was molar ratio strongly affect solvation properties, probably by
nontoxic in concentrations up to 1%, and did not influence the changing polarity and pH, and introducing new intermolecular
cellular viability. However, one drawback of the studied DES was interactions. Some studies have reported the dependence of the
chemical instability of salsalate in it whereas salsalate was stable API solubility in DESs to these structural features (Morrison et al.
in DMSO up to three months. Another study showed that 2009; Lu et al. 2016; Palmelund et al. 2019). Jelin ski et al. (2019)
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 785

Table 2. Examples of solubility of APIs in water and DESs.


Solubility in Solubility in
API Log P DES (molar ratio) DES (mg/mL) water (mg/mL) Solubility ratioa Reference
Acetylsalicylic acid 1.1 Propylene glycol–choline chloride (2:1) 202 7.03 29 (Lu et al. 2016)
Acetaminophen 0.5 Propylene glycol–choline chloride (2:1) 324 19.95 16 (Lu et al. 2016)
Ketoprofen 3.1 Propylene glycol–choline chloride (2:1) 253.40 0.34 744 (Lu et al. 2016)
Naproxen 3.2 Propylene glycol–choline chloride (2:1) 45.26 0.06 754 (Lu et al. 2016)
Ibuprofen 4 Propylene glycol–choline chloride (2:1) 183.3 0.07 190 (Lu et al. 2016)
Itraconazole 5.7 Malonic acid–choline chloride (1:1) 22 <0.001 >22 000 (Morrison et al. 2009)
AMG517 – Malonic acid–choline chloride (1:1) 0.4727 <0.0001 >4727 (Morrison et al. 2009)
Griseofulvin 2.2 Malonic acid–choline chloride (1:1) 1 0.007 143 (Morrison et al. 2009)
Danazol 4.2 Malonic acid–choline chloride (1:1) 0.16 <0.0005 >320 (Morrison et al. 2009)
Benzoic acid 1.9 Urea–choline chloride (2:1) 229 3 76 (Morrison et al. 2009)
Berberine 1.3 Proline–urea (2:1) 12.3 2.1 6 (Sut et al. 2017)
Rutin 0.9 Proline–choline chloride (1:2) 2.643 0.12 22 (Faggian et al. 2016)
Itraconazole 5.7 Glycolic acid–cholinchloride (2:1) 6.7 <0.001 >6700 (Li and Lee 2016)
Piroxicam 3.1 Glycolic acid–cholinchloride (2:1) 9.9 0.023 430 (Li and Lee 2016)
Posaconazole 5.5 Glycolic acid–cholinchloride (2:1) 76.8 0.012 6400 (Li and Lee 2016)
Lidocaine 2.4 Glycolic acid–cholinchloride (2:1) 100.60 3.63 28 (Li and Lee 2016)
Daidzein 2.5 Borneol–menthol (1:3) 0.400 0.008 50 (Shen et al. 2011)
Imipenem 3.9 Trimethylglycine–urea (2:3) 11.32 9.92 1.1 (Olivares et al. 2018)
Clavulanic acid 1.5 Trimethylglycine–urea (2:3) 173.62 289.44 0.6 (Olivares et al. 2018)
a
The ratio of solubility of API in DES to solubility in water.

Figure 6. Solubility of curcumin in NADESs based on choline chloride and


selected HBDs at different molar ratios. Molar ratio (choline chloride: HBD). Data
ski et al. (2019).
were taken from Jelin
Figure 7. Solubility of itraconazole and benzoic acid in malonic acid–choline
chloride (1:1) containing different percentage of water. Data were taken from
Morrison et al. (2009).
measured solubility of curcumin in NADESs based on choline
chloride as HBA and sugars and sugar alcohols as HBD. As it can
nonspecific van der Waals interactions. The same research group
be seen from Figure 6, changing HBD type and molar ratio have
significant effect on curcumin solubility. The authors did not pro- subsequently showed that lidocaine forms strong hydrogen bonds
vide possible explanations behind the obtained results. Further with arginine and organic acids when dissolves in arginine–gluta-
studies are needed to shed a mechanistic light on the influence mic acid (1:1), arginine–oxalic acid (1:1), and arginine–tartaric acid
of different structural parameters of DESs on their solvency. (1:1) (Gutierrez et al. 2019).
Presence of water can greatly influence the properties of DESs Taking into account the large number of possible DESs and in
such as viscosity, polarity, and solvency power (Shekaari et al. the absence of predictive tools considerable time- and labor-
2018). Generally, solubility of poorly water soluble drugs in DESs is intensive experimental work is needed to identify the best DES
significantly decreased by addition of water (Figure 7). The trend of solvent for a given API. Therefore, computational methods are
decrease in solubility is dependent on drug structure and DES type necessary to speed the screening process. Recently Palmelund
(Lu et al. 2016). The added water can weaken intermolecular inter- et al. (2019) performed a computational approach based on the
actions, especially hydrogen-bonding interactions, between the conductor-like screening model for real solvents (COSMO-RS) to
components of DES. It has been shown that high amounts of water predict solubility of APIs in DESs. The API solubility calculated by
(>50%) can completely rupture the hydrogen bonds and destroy COSMO-RS was found in good agreement with experimentally
the supramolecular structure of DES (Dai et al. 2015). measured one. The authors concluded that COSMO-RS can help
Despite an exponentially increasing number of published data to determine DESs with high solvation capacity for a given API.
on APIs solubility in DESs, the study of solute-solvent interactions Turning now to in vivo studies, to date, only very few studies
and solubilization mechanisms of APIs by DESs has received little have assessed the oral pharmacokinetics of poorly soluble APIs dis-
attention to date. Gutierrez et al. (2018) utilized density functional solved in DESs (Faggian et al. 2016; Sut et al. 2017). Berberine is a
theory and molecular dynamics methods to investigate the solv- natural plant alkaloid with antidiabetic, antihypertensive, cholesterol
ation process of lidocaine in choline chloride–lactic acid (1:1) and lowering, and anti-inflammatory effects (Jin et al. 2016). It exhibits
b-alanine–lactic acid (1:1). The results of analyses indicated that low oral bioavailability (less than 1%) due to low solubility and
the drug molecule interacts with components of DESs in solution poor membrane permeability (Zhaojie et al. 2014). As an attempt
through formation of a combination of hydrogen bonding and to solve the solubility problem, a series of 38 DESs were prepared
786 S. EMAMI AND A. SHAYANFAR

hydrophobic forces to adopt well-defined patterns or structures


(Dehsorkhi et al. 2014). Amphiphilic molecules including surfac-
tants (Svenson 2004), block copolymers (Mai and Eisenberg 2012),
lipids (Antonietti and Fo €rster 2003), and some peptides (Zhang
et al. 2002) can self-assemble into a number of various supra-
molecular nanostructures such as liposomes, micelles, vesicles,
tubes, fibers, and lamellas in the solution. Using self-assembled
structures as drug carriers are of great interest in pharmaceutical
sciences due to their advantages such as solubilization potential,
targeted delivery, intracellular delivery, prolonged delivery,
improved permeability through membrane barriers, and possibility
of delivery of both hydrophilic and lipophilic drugs (Ariga et al.
2011; Cui et al. 2016).
Figure 8. Solubility of berberine in: water, (A) proline–malic acid (1:2), (B) proli-
ne–urea (2:1), and (C) proline–malic acid–lactic acid–water (1:0.2:0.3:0.5). Data
Self-assembly only occurs in very limited number of solvents.
were from (Sut et al. 2017). Prior to introduction of ionic liquids, only 17 solvents including
water and some H-bonding molecular solvents had been shown
to support self-assembly of amphiphilic molecules (Greaves and
Drummond 2013). Although ionic liquids have begun to attract
interests as self-assembly media, their limitations necessitate
exploring for better alternatives (Greaves and Drummond 2008).
Possibility of the presence of solvophobic interactions in DESs
make them appropriate media for generating ordered structures
of amphiphilic molecules (Bryant et al. 2016). Self-assembly of cat-
ionic (Sanchez-Fernandez et al. 2016), anionic (Pal et al. 2014),
and zwitterionic surfactants (Sanchez-Fernandez et al. 2018) have
been reported in DESs. Sodium lauryl sulfate (SLS), an anionic sur-
factant, forms micelles in pure choline chloride–urea (1:2) (Arnold
et al. 2015). The behavior of SLS differs greatly in this DES in com-
parison to water. Micelles of SLS in the DES are cylindrical in com-
parison to its spherical micelles in water. In addition, critical
micelle concentration of SLS in choline chloride–urea (1:2) is four
times lower than its CMC in water. As another example, Li et al
(Li et al. 2018) showed that a cationic surfactant cetylpyridium
bromide could form various aggregates, including micelles, hex-
agonal, bicontinuous cubic and lamellar phases in choline chlori-
Figure 9. Plasma concentration-time profiles of berberine; (Water) suspended in
de–glycerol (1:2) and choline chloride–ethylene glycol (1:2).
water, (A) dissolved in proline–malic acid (1:2), (B) dissolved in proline–urea (2:1),
and (C) dissolved in proline–malic acid–lactic acid–water (1:0.2:0.3:0.5) in mice Self-assembly of a series of single-chain n-alkyltrimethylammo-
(n ¼ 15, mean ± SD) The figure was adapted with permission from (Sut et al. nium based cationic surfactants has been shown in choline chlori-
2017) Copyright (2017) MDPI. de–glycerol (1:2) by using fluorescence probe, electrical
conductivity, surface tension, small-angle X-ray/dynamic light scat-
tering, and transmission electron microscopy methods (Pal et al.
2015). The authors reported that hexadecyl trimethylammonium
and used to dissolve berberine (Sut et al. 2017). From the several bromide self-assembled as aggregates with a mean size of about
DESs studied, three DESs with higher solvency power for the drug 36 nm in the DES (Figure 10).
(Figure 8) were selected for pharmacokinetic studies. To assess the Beside to surfactants, self-assembly of lipids has also been
bioavailability, mice were orally gavaged with berberine suspended reported in DESs. By using solvent penetration experiments and
in water or dissolved in the selected DESs. Dissolving berberine in small-angle X-ray scattering, Bryant et al. (2016) were able to
DESs, especially in proline–malicacid–lactic acid–water (1:0.2:0.3:0.5), show that phosphatidylcholine lipids dissolved in choline chlori-
led to significant increases in oral bioavailability of berberine de–urea (1:2) DES self-assemble to vesicles above phase transition
(Figure 9). The maximum plasma concentration and the area under temperature of lipids. In another study, the same authors reported
the mean plasma concentration-time curve values of berberine dis- that DESs composed of alkylammonium halide salts and glycerol
solved in this DES were approximately 6- and 4-fold higher than or ethylene glycol support self-assembly of phosphatidylcholine
those of berberine suspension. Improvements in the bioavailability lipids (Bryant et al. 2017). These findings may expand applications
were attributed to solubilization of the drug by DESs. Many more of biocompatible DESs to develop self-assembled drug carriers
such in vivo studies in animal models and then in human subjects and open new ways for designing drug delivery systems.
are required to verify the real potential of DESs in improving bio-
availability of poorly soluble APIs.
2.5. Application of DESs in dermal and transdermal
drug delivery
2.4. Self-assembly of amphiphiles in DESs
Drug delivery across the skin offers distinct advantages over other
Molecular self-assembly is the spontaneous and reversible associ- routes of administration. The advantages are being non-invasive,
ation of amphiphilic molecules by means of non-covalent interac- better patient compliance, avoiding pre-systemic gastrointestinal
tions such as hydrogen bonding, van der Waals, electrostatic, or and hepatic metabolism, lower side effects, direct access to target
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 787

Figure 10. Transmittance electron microscopy images for hexadecyl trimethylammonium bromide [20 mM (left) and 50 mM (right)] in choline chloride–glycerol (1:2).
The figure was adapted with permission from (Pal et al. 2015). Copyright (2015) Wiley-VCH.

Figure 11. Schematic presentation of the preparation of an oil in water emulsion based on a deep eutectic mixture of lidocaine and prilocaine.

or diseased site, and capability for sustained and controlled deliv- at room temperature) by using appropriate excipients and others
ery (Brown et al. 2006). have used DESs as the carrier for APIs.
The major challenge in designing delivery systems for treat-
ment of dermatological diseases (topical or dermal delivery) or for 2.5.1. Using API-DESs for dermal and transdermal drug delivery
systemic delivery through skin (transdermal delivery) is low per- From the historical point of view, the first applications of API-
meability of skin (Alvarez-Roman et al. 2003). The human skin DESs in the field of drug delivery seems to has been reported for
inherently possesses significant barrier properties to protect body topical and transdermal delivery. In 1889, Jules-Aristide Bonain
organs from entrance of foreign toxins and minimize water loss. discovered that phenol, cocaine hydrochloride, and menthol in
The stratum corneum (SC), the non-living, outermost layer of the equivalent proportions form a room temperature liquid and used
skin, especially hinders the permeation of most APIs (Prausnitz the formed liquid mixture for topical anesthesia (Fiala et al. 2010).
et al. 2004). Only a limited number of drugs with low molecular Later in 1984, it was reported that lidocaine and prilocaine forms
weight (<500 Da) and optimum lipophilicity (Log P ¼ 1–3), can a 1:1 eutectic mixture with the melting point of 18  C (Brodin
effectively penetrate through the intact skin (Naik et al. 2000). et al. 1984). Then, this API-DES was formulated as an oil in water
emulsion (Figure 11) called EMLAV (Eutectic Mixture of Local
R
Therefore, appropriate permeability enhancement strategies
should be used for hydrophilic drugs and macromolecules. Anesthetics) cream and was consequently approved by FDA for
Various strategies such as chemical penetration enhancers local anesthesia in 1992 (Nyqvist-Mayer et al. 1986; Eichenfield
(Pham et al. 2016), physical methods (Nanda et al. 2006), and ves- et al. 2002). The analgesic effectiveness of emulsion formulation
icular carriers (Dubey et al. 2006) have been investigated to based on the API-DES is higher than either prilocaine or lidocaine
applied alone (Gajraj et al. 1994). EMLAV cream still is the most
R
improve skin permeability of APIs. These strategies are mainly
based on disrupting the SC barrier or altering the lipid structure. widely used topical anesthetic. In the same line, an aerosol-deliv-
Toxicity and skin irritation of chemical enhancers and infections, ery form containing lidocaine–prilocaine eutectic-like mixture
burns, and scars related to physical methods have necessitated (FortacinTM) was approved to be used for treatment of premature
the introduction of effective and safe enhancement approaches ejaculation in the European Union in 2016 (Porst and Burri 2017).
(Karande and Mitragotri 2009; Banerjee et al. 2017). These marketed examples highlight the great potential of the
Due to the low toxicity and being liquid at room temperature, API-DESs in the area of dermal and transdermal drug delivery.
eutectic mixtures have been investigated for dermal and transder- The mentioned successful examples have motivated research-
mal drug delivery applications. For these purposes, some ers to attempt eutectic formation approaches for transdermal
researches have focused on converting solid APIs to API-DESs (oil delivery of other drugs. API-DESs could enhance the transdermal
788 S. EMAMI AND A. SHAYANFAR

Table 3. API-DESs reported for transdermal delivery applications.


Tm of
API Coformer Mole ratio Tm of API coformer Tm/Tg of API-DES Reference
Ibuprofen Menthol 3:7 76 41 19 (Stott et al. 1998)
Ibuprofen Methyl nicotinate 1:1 (w/w) 76 41 a (Woolfson et al. 2000)
Itraconazole Phenol 1:1 168 39 <0 (Park et al. 2012)
Lidocaine Camphor 1:1 (w/w) 68 175 33 (Gala et al. 2015)
Lidocaine Tetracaine 1:1 (w/w) 68 41 30 (Gala et al. 2015)
Lidocaine Decanoic acid 1:1 68 32 61 (Bica et al. 2011)
Lidocaine Hexanoic acid 1:1 68 3 56 (Bica et al. 2011)
Lidocaine Linoleic acid 1:1 68 5 71 (Bica et al. 2011)
Lidocaine Oleic acid 1:1 68 13 47 (Bica et al. 2011)
Lidocaine Stearic acid 1:1 68 69 43 (Bica et al. 2011)
Lidocaine Ibuprofen 1:1 68 76 27 (Wang et al. 2014)
Lidocaine Menthol 3:7 (w/w) 68 41 26 (Kang et al. 2000)
Lidocaine Phenyl salicylate 3:2 68 41 18 (Lazerges et al. 2010)
Lidocaine Prilocaine 1:1 68 38 18 (Brodin et al. 1984)
Paeonol Menthol – 52 41 a (Wang et al. 2017)
Propranolol Capric acid 3.5:6.5 (w/w) 92 32 15 (Stott et al. 2001)
Propranolol Lauric acid 3:7 (w/w) 92 43 16 (Stott et al. 2001)
Testosterone Menthol 1:4 154 41 39 (Kaplun-Frischoff and Touitou 1997)
a
Liquid at room temperature

permeability of propranolol, testosterone, ibuprofen, itraconazole, It will also be interesting to see whether it is possible to tailor
and lidocaine (Table 3). The enhancement mechanisms have been API-DES properties by changing counterpart.
suggested to be related to penetration enhancing capability of The chemical structure of the molecule remains unaffected
coformers (such as menthol), melting point depression, and when a solid API is converted to a liquid API-DES. This can be a
increased membrane solubility (Stott et al. 1998; Fiala et al. 2010). great advantage for API-DESs from drug development and regula-
Indeed, the oily liquid phase (API-DES) containing high drug con- tory decision making because pharmaceutical properties can be
centration acts as a reservoir, and thus may provide higher driving modified without structural change of the API. However, there are
force for diffusion of drug molecules in to the skin (Nyqvist-Mayer no established regulatory authority guidelines regarding API-DESs.
et al. 1986). For example, formation of an API-DES of testosterone, The answer to the question of do regulatory bodies treat API-
a lipophilic molecule, with menthol, a known permeation enhan- DESs as new chemical entities requiring extensive preclinical
cer, at the molar ratio of 4:1 resulted in drastic melting point pharmacological and toxicology studies or see them as new for-
depression of testosterone from 153.7 to 39.9  C. The permeation mulations? is crucial. The answer could significantly affect the
of this API through mice skin was eightfold higher in the form of interest of industry towards research on API-DESs.
API-DES than the aqueous solution of testosterone. The increase
in permeation was attributed to improved solubility of testoster- 2.5.2. DESs as transdermal delivery vehicles
one in hydroalcoholic vehicle and reducing barrier properties of Another interesting area of research is using DESs as carriers/
vehicles with penetration enhancing power in dermal and trans-
SC by menthol (Kaplun-Frischoff and Touitou 1997). Despite these
dermal drug delivery systems. In this context, Mitragotri group
promising results, there are still many unanswered questions
(Zakrewsky et al. 2014) synthesized a range of ILs and DESs and
about the nature of API-DESs, physicochemical stability, compati-
investigated their cytotoxicity, skin irritation, antimicrobial effect
bility with excipients, and formulating these liquids as an accept-
on bacterial biofilm, and delivery of antibiotics through the skin.
able final dosage form.
The best results were obtained for choline–geranate (1:2) DES.
Hydrogen bond formation between an API and coformer is
This DES showed the lowest toxicity for epithelial cells, minimum
considered as the driving force of the melting point depression
of skin irritation, excellent antimicrobial activity against bacterial
and API-DES formation. Theoretically, API-DES formation is pos-
biofilms, and highest efficiency in improving transdermal delivery
sible for a considerable number of pharmaceutical molecules that
of model hydrophilic molecules. For example, the skin permeation
are either hydrogen bond donors or quaternary ammonium salts of cefadroxil, a hydrophilic antibiotic with a log P of –0.4, was
(Abbott et al. 2017). However, until now API-DES discovery has improved by fivefold when dissolved in choline–geranate (1:2) in
been performed mainly by experimental work of the mixing of comparison to aqueous solution of the drug. The mechanism of
APIs with different coformers in different ratios. Very recently, permeability enhancement was proposed to be the extraction of
Wolbert et al. (2019) performed a thermodynamic modeling to lipids from SC. The results suggest that choline–geranate (1:2)
predict eutectic composition and eutectic temperature of an API/ would be a promising carrier for dermal delivery of antibiotics for
coformer eutectic system. Melting points, melting enthalpies, and the eradication of biofilm-associated infections.
thermodynamic activity coefficients of the API and coformer were Biological macromolecules such as proteins and peptides have
used to construct the prediction. The predictions were made for very low transdermal permeability due to their large sizes (Benson
11 combinations of three APIs and six coformers. The predicted and Namjoshi 2008). Encouraged by low toxicity, low irritability,
eutectic compositions and eutectic temperatures were in good and good penetration enhancing property of choline–geranate
agreement with experimental data indicating the applicability of (1:2) observed for hydrophilic antibiotics, Mitragotri’s group fur-
the proposed model for rapid screening of API-DES formation. ther investigated the efficacy of the DES in enhancing permeation
Certainly, there is abundant room for further progress in develop- of proteins across skin. The research group (Banerjee et al. 2017)
ing rational strategies in order to guide the process of synthesis studied ex vivo skin permeability of bovine serum albumin (MW
of API-DESs and selection of optimal one for a given application. 66 kDa), ovalbumin (MW 45 kDa), and insulin (MW 5.8 kDa)
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 789

3. Challenges and limitations of DESs


In the previous sections we briefly reviewed the potential
pharmaceutical applications of DESs. However, to be selected as
solvents or processing materials by pharmaceutical industry, DESs
should compete with currently used drug delivery systems and
organic solvents. In the oncoming section, discussions will be con-
ducted to investigate some important challenges and limitations
that stand in the way of pharmaceutical applications of DESs.

Figure 12. Percent change in blood glucose level with time after administration 3.1. Cost
of various formulations in nondiabetic rats. Topical application of choline–gera-
nate (1:2) (CAGE) alone (0 U kg1 insulin, green triangles); insulin in buffer Without a doubt, cost is a major factor that should to be consid-
(Insulin-PBS, 10 U mL1 as 25 U kg1 body weight, red squares); insulin in choli- ered for commercial use of DESs. To our knowledge, the commer-
ne–geranate (1:2) (10 U mL1 with total dose 25 U kg1 body weight, blue cialization potential of DESs has not been investigated yet. As the
circles); and subcutaneous insulin injection (1 U kg1 insulin, magenta inverted
triangles) was performed. The figure was adapted with permission from field matures, studies need to be performed to techno-economic
(Banerjee et al. 2017). Copyright (2017) Wiley-VCH. analysis of DESs. At the current stage, it seems that the cost of
starting materials and method of synthesis are the largest contrib-
utors in the way of commercial applications of DESs (Chen et al.
dissolved in choline–geranate (1:2). The DES significantly
2014). Many DES components are cost effective, some of which
improved skin penetrations of these macromolecules and was
are routinely used in pharmaceutical applications as excipients.
more efficient than commonly used chemical penetration
For example, choline chloride, a common component of DESs, is
enhancers; diethylene glycol monoethyl ether and ethanol. The
produced in megaton scales at a reasonably low cost (Jablonsky
results of infrared spectroscopy studies of SC suggested that chol- et al. 2019). From preparation point of view, DESs have a simple
ine–geranate (1:2) enhances permeability by the extraction of SC and easy to scale up production method. Therefore, one person
lipids. In the further step, the authors investigated in vivo blood can assume that perhaps not all but at least some DESs can be as
glucose lowering efficacy of insulin dissolved in choline–geranate inexpensive as conventional organic solvents (Cruz et al. 2017). In
(1:2) applied to the skin of nondiabetic rats (Figure 12). When addition, if a DES is being considered as a component of a drug
insulin was dissolved in the DES resulted in 25 and 40% decrease delivery system, the advantages of the prepared formulation may
in blood glucose in 2 and 4 h, respectively, compared to no sig- outweigh the possible higher costs.
nificant reduction in blood glucose level by insulin dissolved in
phosphate buffer.
Glabridin is a water insoluble polyphenolic flavonoid with anti- 3.2. Structure and classification
oxidant, anti-inflammatory, and skin lightening properties. Liu The structure determines the function. Although recently the
et al. (2017) used a menthol–camphor (1:1) DES as the oil phase number of studies focusing on resolving structures of DESs is
(vehicle) for developments of oil in water nanoemulsion of this increasing (Zahn 2017; McDonald et al. 2018) but the basic ques-
drug. Menthol and camphor have been reported as nontoxic, tion of what is the liquid structure of a DES yet continue to be
nonirritating, and efficient transdermal penetration enhancers largely unanswered. The lack of enough structural information
(Sapra et al. 2008). These natural terpenes form a water-immis- also makes it difficult to establish a generally accepted definition
cible DES with hydrophobic properties. The menthol–camphor and classification of DESs. We clearly need to know more about
DES can act as the penetration enhancer as well as the solvent the liquid structure, dynamic, and molecular interactions of DESs
for transdermal delivery of water insoluble compounds. The to establish the structure  property relationships and rationally
authors compared permeability properties of eutectic-based nano- design tailored DESs.
emulsion with nanoemulsion formulated with isopropyl myristate
and aqueous solution of the drug. Isopropyl myristate is a com-
monly used lipophilic vehicle and penetration enhancer in trans- 3.3. High viscosity
dermal delivery (Engelbrecht et al. 2012). The solubility of Generally, due to the strong hydrogen networks, DESs have
glabridin in the DES was about two times higher than its solubil- 100–1000 times higher viscosities than water and common
ity in isopropyl myristate. More importantly, in vitro skin perme- organic solvents (Tang and Row 2013). High viscosity is a limiting
ability of eutectic based nanoemulsion was about 3-and 7-times factor for DESs applications and can cause problems in pharma-
higher than isopropyl myristate-based nanoemulsion and aqueous ceutical operations such as handling, mixing, and filling. Thus,
solution of the drug, respectively. strategies are needed to reduce the viscosity to a suitable level.
Although DESs have the ability to dissolve solutes with wide Among others, viscosity of a DES can be affected by temperature
range of chemical nature but unlike their analogues ILs, the num- and the presence of a third component such as water. Increasing
ber of published works on utilizing DESs as solvent/vehicle for the temperature leads to strong decrease in viscosity. For
transdermal drug delivery are rare yet. We predict that more data example, the viscosity of choline chloride-urea (1:2) was decreased
will be generated in near future by introducing new DESs, specif- from 1100 to 200 cP when temperature was increased from 20 to
ically hydrophobic ones. DESs can be used as dual-function sol- 40  C (Abbott et al. 2003). Studies have also shown that addition
vents in this area. Taking the advantage of large number of of small amounts of water, up to 10%, to DESs can significantly
possible combinations, a researcher would select a DES based reduce their viscosity while maintaining their solvency power. For
vehicle with desired properties such as wound healing, occlusive, example, addition of 10% of water to choline chloride-urea (1:2)
penetration enhancing, or antibacterial, depending on a spe- DES decreased the viscosity to 1/550 of its original value
cific condition. (Guajardo et al. 2017).
790 S. EMAMI AND A. SHAYANFAR

Table 4. Summary of studies conducted on toxicity of DESs.

HBA HBD Biological system Results Reference


Choline chloride Sugars and alcohols Human cell line Toxic to cells, More toxic than their (Ahmadi et al. 2018)
starting materials
Urea, acetamide, glycerol, and Bacteria and aquatic Strong anti-bacterial activity, (Wen et al. 2015)
ethylene glycol organism Considerable toxic effects on cells
Urea, triethylene glycol, Bacteria and aquatic No inhibition on bacterial growth, More (Hayyan, Hashim, Al-Saadi,
glycerol, and organism toxic than their starting materials et al. 2013; Hayyan,
ethylene glycol Hashim, Hayyan,
et al. 2013)
Urea, acetamide, sugars Bacteria No inhibition on bacterial growth by (Zhao et al. 2015)
and alcohols amine-, alcohol- and sugar-based
DESs, Inhibition on bacterial growth
by acid-based DESs
Glucose, glycerol, and Human and fish Low cytotoxicity for choline chloride- (Radosevic et al. 2015)
oxalic acid cell lines glucose and choline chloride-
glycerol, Moderate cytotoxicity for
choline chloride-oxalic acid
Glycerol, ethylene glycol, Human cell line Relatively high cytotoxicity, More toxic (Hayyan et al. 2015)
triethylene glycol, and urea than their starting materials
Citric acid, Fructose, glucose, sucrose, Human and mouse Relatively nontoxicity by DESs of (Hayyan et al. 2016)
trimetyl glycerol, and malonic acid cell lines choline chloride with sugars and
glycine water, cytotoxicity by choline
chloride-malonic acid
Oxalic acid, urea, sugars, Bacteria and human Inhibition on bacterial growth by most (Radosevic et al. 2018)
Glycerol, proline, sugars cell line of DESs, No toxicity by Most DESs,
Glucose, malic acid, High toxicity by choline chloride-
and proline oxalic acid
Methyltriphenyl Glycerol, ethylene glycol, Bacteria and aquatic Inhibition on bacterial growth, More (Hayyan, Hashim, Al-Saadi,
phosphonium triethylene glycol organism toxic than their starting materials, et al. 2013; Hayyan,
bromide Dependence of cytotoxicity of DESs Hashim, Hayyan,
on the structure of components et al. 2013)

3.4. Hygroscopicity non-toxic, eco-friendly, biodegradable, and benign solvents alter-


native for ILs and other organic solvents (Wen et al. 2015).
Hygroscopicity is the tendency of a material to take up water
However, these assumptions have been disputed by recent stud-
vapor from the surrounding air (Newman et al. 2008). Generally,
ies. Table 4 summarizes the results of DESs toxicity in different
possibility of water sorption is increased, when a crystalline sub-
biological systems. As it can be seen different DESs exhibit differ-
stance is converted to a liquid form because environmental water
ent degrees of toxicity depending on the structure of compo-
can easily dissolve in the disordered liquid state (Balk et al. 2015).
nents. Furthermore, some DESs are much more toxic than their
Due to their vast hydrogen bonding networks and hydrophilic starting materials. For detailed discussion on the topic, the reader
nature, most of DESs are hygroscopic and the adsorption of mois- is referred to a recent comprehensive literature review (Juneidi
ture from air has been reported for some of them (Hammond et al. 2018) on the topic.
et al. 2017). For example, choline chloride-urea (1:2) could absorb Although there are limited published data on toxicity of DESs
about 4% of water after 24 h exposure to a relative humidity of but what these data suggest is that one should not consider DESs
64% and 20  C (Zhao et al. 2011). Another study showed that a priori as non-toxic solvents simply by looking at the toxicity pro-
DESs based on choline chloride and carboxylic acids took up files of their staring components. It seems that there is no general
10–20% of water when were stored for 30 days in contact with air answer to the toxicity question and for each DES in-depth investi-
(Florindo et al. 2014). gations are required to determine the toxicity, biocompatibility,
The presence of water can dramatically affect the structure and biodegradability profiles before applying it to pharmaceutical
and physicochemical characteristics of DESs (Dai et al. 2015). applications.
Introducing of water could enormously reduce the viscosity of
DESs (Yadav and Pandey 2014). Polarity and the ability of DESs to
solubilize solutes are other affected properties (Pandey and 3.6. Long-term stability of APIs in DESs
Pandey 2014). As mentioned earlier, presence of water, even in The long term stability of APIs in the forms of API-DESs or as dis-
low amounts, can cause dramatic changes in solvency power of solved in DESs is another inherent concern. Until now, there are
DESs. In addition, it is predictable that chemical stability of DES very limited studies that evaluate physicochemical stability of APIs
and incorporated API will be reduced by contamination of in DESs under stressed storage conditions of elevated tempera-
DESs with water. For these reasons, it is crucial to evaluate hygro- ture and/or high humidity. Generally speaking, APIs in solid state
scopicity of DESs and their water content should be carefully are more physicochemical stable than liquid state and transform-
monitored and controlled. ing solid APIs to liquid forms can increase the risk of instabilities
(Balk et al. 2015). In the case of APIs dissolved in DESs, enhanced
chemical stability has been reported for some hydrolysis-sensitive
3.5. Toxicity
APIs such as aspirin (Lu et al. 2016), imipenem and clavulanic acid
Most of the components of DESs are non-toxic, biodegradable, (Olivares et al. 2018), salvialonic acid B (Chen et al. 2016), and car-
and pharmaceutically acceptable therefore, DESs are assumed as thamin (Dai et al. 2014) in short-term storage times in comparison
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 791

Table 5. Summary of potentials and challenges of pharmaceutical applications systems. In particular, the ability of DESs to dissolve various types
of DESs. of biopolymers ranging from DNA to proteins and polysaccharides
Potentials Challenges makes them an ideal option to process the biopolymers.
Easy method of preparation Hygroscopicity Despite these promising results, deeper studies on the interac-
Readily available components Absence of techno-economic tions and building principles are needed in order to gain insight
analysis data
Numerous number of possible Long term stability of dissolved APIs into the properties of DESs and to. Reasonable data should
combinations become available regarding toxicity, biocompatibility, biodegrad-
Solvency for biopolymers and Toxicity and safety profile ability, and safety. Also, appropriate strategies should be devel-
water insoluble APIs
oped to reduce viscosity, overcome hygroscopicity, and determine
Non-volatile and non-inflammable High viscosity
Dual-function solvents Unresolved structural properties long-term physicochemical stability.
Supporting self-assembly of Absence of structure-property We searched ClinicalTrialsRegister.eu and ClinicalTrials.gov
amphiphile molecules relationships using the terms, ‘deep eutectic solvents’ OR ‘eutectic’ to retrieve
Reaction media to design tailor
made materials
possible eutectics under clinical investigations. There were 38
Biodegradable studies all of them were about eutectic mixtures of local anes-
Enhancing Permeability of APIs thetics. This indicate that until now, most of the conducted stud-
through biological barriers ies have investigated DESs from a proof-of-concept point of view,
and no considerable attempts have been invested to translate the
findings of basic science to real-world pharmaceutical product
to their water stability. As an example, in a time period of seven development. Overall, we expect that DESs and API-DESs will find
days, the stability of imipenem and clavulanic acid in trimethyl real applications in the field of drug delivery in the near future.
glycine–urea (1:1.5) DES, was 7 and 2.5 times higher, respectively,
compared to water (Olivares et al. 2018). In another study it was
reported that photostability of curcumin dissolved in chloride–gly- Disclosure statement
cerol (1:1) was significantly higher than curcumin dissolved in No potential conflict of interest was reported by the author(s).
methanol and curcumin powder (Jelin ski et al. 2019). However,
these improved stabilities cannot be extrapolated to other APIs
and in each case comprehensive long-term stability data should ORCID
be provided. Shahram Emami http://orcid.org/0000-0002-5182-1728
Ali Shayanfar http://orcid.org/0000-0001-7507-9057
4. Concluding remarks
This paper provides an overview about current advances made References
over the last decade in utilizing of DESs and API-DESs for pharma-
Abbott AP, Ahmed EI, Prasad K, Qader IB, Ryder KS. 2017. Liquid
ceutical formulation and drug delivery applications. Table 5 sum-
marizes the potentials and challenges of DESs in pharmaceutical pharmaceuticals formulation by eutectic formation. Fluid Phase
engineering. Equilib. 448:2–8.
In the field of drug solubilization, published data show that Abbott AP, Capper G, Davies DL, Rasheed RK, Tambyrajah V. 2003.
DESs can effectively dissolve water insoluble APIs and may find Novel solvent properties of choline chloride/urea mixtures.
applications in early pharmacological and toxicological evaluations Chem Commun. 2003(1):70–71.
of drug candidates and development of effective pharmaceutical Abo-Hamad A, Hayyan M, AlSaadi MA, Hashim MA. 2015. Potential
products. However, more studies should investigate solute-solvent applications of deep eutectic solvents in nanotechnology.
interactions and solubilization mechanisms and propose models Chem Eng J. 273:551–567.
to predict API solubility in DESs. Furthermore, in vivo behavior of Abo-Hamad A, Hayyan M, AlSaadi MA, Mirghani MES, Hashim MA.
API dissolved in DES remains largely unknown. Therefore, 2017. Functionalization of carbon nanotubes using eutectic
researchers should plan to conduct studies on cell cultures, ani- mixtures: a promising route for enhanced aqueous dispersibility
mal models, and human subjects to assess the bioavailability and electrochemical activity. Chem Eng J. 311:326–339.
enhancing potential. Ahmadi R, Hemmateenejad B, Safavi A, Shojaeifard Z, Mohabbati
Regarding transdermal drug delivery, most studies have M, Firuzi O. 2018. Assessment of cytotoxicity of choline chlor-
focused on API-DESs and there are scare studies about utilizing ide-based natural deep eutectic solvents against human HEK-
DESs as delivery vehicles. DESs, as dual-function solvent, can act
293 cells: A QSAR analysis. Chemosphere. 209:831–838.
as penetration enhancer, wound healing, or antibacterial in add- n DJ.
Alonso DA, Baeza A, Chinchilla R, Guillena G, Pastor IM, Ramo
ition to their solvent role. On the hand, many APIs can be
2016. Deep eutectic solvents: the organic reaction medium of
designed as API-DESs. The successful case of local anesthetics
the century. Eur J Org Chem. 2016(4):612–632.
(EMLA) indicates that API-DESs are not far from clinical applica-
Alvarez-Roman R, Merino G, Kalia YN, Naik A, Guy RH. 2003. Skin
tions and more designer API-DESs can contribute to improve
pharmaceutical properties of problematic APIs such as solubility, permeability enhancement by low frequency sonophoresis:
permeability, and polymorphism. Indeed, the most promising area lipid extraction and transport pathways. J Pharm Sci. 92(6):
of API-DESs applications seems to be in dermal and transdermal 1138–1146.
drug delivery. Amde M, Liu J-F, Pang L. 2015. Environmental application, fate,
Also, DESs can help to design innovative and sophisticated effects, and concerns of ionic liquids: a review. Environ Sci
materials with pharmaceutical interest such as inorganic nanoma- Technol. 49(21):12611–12627.
terials, functionalized nanomaterials, self-assembled delivery Antonietti M, Fo€rster S. 2003. Vesicles and liposomes: a self-assem-
vehicles, and polymer and biopolymer based drug delivery bly principle beyond lipids. Adv Mater. 15(16):1323–1333.
792 S. EMAMI AND A. SHAYANFAR

Ariga K, Lvov YM, Kawakami K, Ji Q, Hill JP. 2011. Layer-by-layer Cruz H, Jord~ao N, Branco LC. 2017. Deep eutectic solvents (DESs)
self-assembled shells for drug delivery. Adv Drug Del Rev. 63(9): as low-cost and green electrolytes for electrochromic devices.
762–771. Green Chem. 19(7):1653–1658.
Arnold T, Jackson AJ, Sanchez-Fernandez A, Magnone D, Terry AE, Cui W, Li J, Decher G. 2016. Self-assembled smart nanocarriers for
Edler KJ. 2015. Surfactant behavior of sodium dodecylsulfate in targeted drug delivery. Adv Mater. 28(6):1302–1311.
deep eutectic solvent choline chloride/urea. Langmuir. 31(47): Dai Y, van Spronsen J, Witkamp G-J, Verpoorte R, Choi YH. 2013.
12894–12902. Natural deep eutectic solvents as new potential media for
Aroso IM, Craveiro R, Rocha A, ^ Dionısio M, Barreiros S, Reis RL, green technology. Anal Chim Acta. 766:61–68.
Paiva A, Duarte A. 2015. Design of controlled release systems Dai Y, Verpoorte R, Choi YH. 2014. Natural deep eutectic solvents
for THEDES—therapeutic deep eutectic solvents, using super- providing enhanced stability of natural colorants from safflower
critical fluid technology. Int J Pharm. 492(1–2):73–79. (Carthamus tinctorius). Food Chem. 159:116–121.
Aroso IM, Silva JC, Mano F, Ferreira ASD, Dionısio M, Sa-Nogueira Dai Y, Witkamp G-J, Verpoorte R, Choi YH. 2015. Tailoring proper-
I, Barreiros S, Reis RL, Paiva A, Duarte A. 2016. Dissolution ties of natural deep eutectic solvents with water to facilitate
enhancement of active pharmaceutical ingredients by thera- their applications. Food Chem. 187:14–19.
peutic deep eutectic systems. Eur J Pharm Biopharm. 98:57–66. Dehsorkhi A, Castelletto V, Hamley IW. 2014. Self-assembling
Balk A, Holzgrabe U, Meinel L. 2015. Pro et contra’ ionic liquid amphiphilic peptides. J Pept Sci. 20(7):453–467.
drugs – challenges and opportunities for pharmaceutical trans- del Monte F, Carriazo D, Serrano MC, Gutierrez MC, Ferrer ML.
lation. Eur J Pharm Biopharm. 94:291–304. 2014. Deep eutectic solvents in polymerizations: a greener
Banerjee A, Ibsen K, Iwao Y, Zakrewsky M, Mitragotri S. 2017. alternative to conventional syntheses. ChemSusChem. 7(4):
Transdermal protein delivery using choline and geranate 999–1009.
(CAGE) deep eutectic solvent. Adv Healthcare Mater. 6(15): Dubey V, Mishra D, Asthana A, Jain NK. 2006. Transdermal delivery
1601411. of a pineal hormone: melatonin via elastic liposomes.
Benson HAE, Namjoshi S. 2008. Proteins and peptides: strategies Biomaterials. 27(18):3491–3496.
for delivery to and across the skin. J Pharm Sci. 97(9): Durand E, Lecomte J, Barea B, Piombo G, Dubreucq E, Villeneuve
3591–3610. P. 2012. Evaluation of deep eutectic solvents as new media for
Bhatt J, Mondal D, Bhojani G, Chatterjee S, Prasad K. 2015. Candida antarctica B lipase catalyzed reactions. Process
Preparation of bio-deep eutectic solvent triggered cephalopod Biochem. 47(12):2081–2089.
shaped silver chloride-DNA hybrid material having antibacterial Durand E, Lecomte J, Upasani R, Chabi B, Bayrasy C, Barea B,
and bactericidal activity. Mater Sci Eng C. 56:125–131. Jublanc E, Clarke MJ, Moore DJ, Crowther J, et al. 2017.
Bhatt J, Mondal D, Devkar RV, Prasad K. 2016. Synthesis of func- Evaluation of the ROS inhibiting activity and mitochondrial tar-
tionalized N-doped graphene DNA hybrid material in a deep geting of phenolic compounds in fibroblast cells model system
eutectic solvent. Green Chem. 18(15):4297–4302. and enhancement of efficiency by natural deep eutectic solvent
Bica K, Shamshina J, Hough WL, MacFarlane DR, Rogers RD. 2011. (NADES) formulation. Pharm Res. 34(5):1134–1146.
Liquid forms of pharmaceutical co-crystals: exploring the boun- Egorova KS, Ananikov VP. 2014. Toxicity of ionic liquids: eco(cy-
daries of salt formation. Chem Commun. 47(8):2267–2269. to)activity as complicated, but unavoidable parameter for task-
Brodin A, Nyqvist-Mayer A, Broberg F, Wadsten T, Forslund B. specific optimization. ChemSusChem. 7(2):336–360.
1984. Phase diagram and aqueous solubility of the lidocaine- Egorova KS, Gordeev EG, Ananikov VP. 2017. Biological activity of
prilocaine binary system. J Pharm Sci. 73(4):481–484. ionic liquids and their application in pharmaceutics and medi-
Brown MB, Martin GP, Jones SA, Akomeah FK. 2006. Dermal and cine. Chem Rev. 117(10):7132–7189.
transdermal drug delivery systems: current and future pros- Eichenfield LF, Funk A, Fallon-Friedlander S, Cunningham BB.
pects. Drug Deliv. 13(3):175–187. 2002. A clinical study to evaluate the efficacy of ELA-max (4%
Bryant SJ, Atkin R, Warr GG. 2016. Spontaneous vesicle formation liposomal lidocaine) as compared with eutectic mixture of local
in a deep eutectic solvent. Soft Matter. 12(6):1645–1648. anesthetics cream for pain reduction of venipuncture in chil-
Bryant SJ, Atkin R, Warr GG. 2017. Effect of deep eutectic solvent dren. Pediatrics. 109(6):1093–1099.
nanostructure on phospholipid bilayer phases. Langmuir. Emadi F, Amini A, Gholami A, Ghasemi Y. 2017. Functionalized
33(27):6878–6884. graphene oxide with chitosan for protein nanocarriers to pro-
Carriazo D, Serrano MC, Gutierrez MC, Ferrer ML, del Monte F. tect against enzymatic cleavage and retain collagenase activity.
2012. Deep-eutectic solvents playing multiple roles in the syn- Sci Rep. 7(1):42258.
thesis of polymers and related materials. Chem Soc Rev. 41(14): Emami S, Adibkia K, Barzegar-Jalali M, Siahi-Shadbad M. 2019.
4996–5014. Piroxicam cocrystals with phenolic coformers: preparation, char-
Chen J, Li S-F, Yao Z-F, Yang D-W, Zhang L-W. 2016. Improved sta- acterization, and dissolution properties. Pharm Dev Technol.
bility of salvianolic acid B from Radix Salviae miltiorrhizae in 24(2):199–210.
deep eutectic solvents Anal Methods. 8(11):2502–2509. Emami S, Jouyban A, Valizadeh H, Shayanfar A. 2015. Are crystal-
Chen L, Sharifzadeh M, Mac Dowell N, Welton T, Shah N, Hallett linity parameters critical for drug solubility prediction? J Solut
JP. 2014. Inexpensive ionic liquids: [HSO4]-based solvent pro- Chem. 44(12):2297–2315.
duction at bulk scale. Green Chem. 16(6):3098–3106. Emami S, Siahi-Shadbad M, Adibkia K, Barzegar-Jalali M. 2018.
Chen F, Xie S, Zhang J, Liu R. 2013. Synthesis of spherical Fe3O4 Recent advances in improving oral drug bioavailability by coc-
magnetic nanoparticles by co-precipitation in choline chloride/ rystals. Bioimpacts. 8(4):305–320.
urea deep eutectic solvent. Mater Lett. 112:177–179. Engelbrecht TN, Deme B, Dobner B, Neubert R. 2012. Study of the
Choi YH, van Spronsen J, Dai Y, Verberne M, Hollmann F, Arends I, influence of the penetration enhancer isopropyl myristate on
Witkamp G-J, Verpoorte R. 2011. Are natural deep eutectic sol- the nanostructure of stratum corneum lipid model membranes
vents the missing link in understanding cellular metabolism using neutron diffraction and deuterium labelling. Skin
and physiology? Plant Physiol. 156(4):1701–1705. Pharmacol Physiol. 25(4):200–207.
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 793

Englert C, Brendel JC, Majdanski TC, Yildirim T, Schubert S, functionalization of upconversion nanocrystals for imaging and
Gottschaldt M, Windhab N, Schubert US. 2018. Pharmapolymers in vivo drug delivery. Adv Mater. 29(18):1700244.
in the 21st century: synthetic polymers in drug delivery applica- Hayyan M, Hashim MA, Al-Saadi MA, Hayyan A, AlNashef IM,
tions. Prog Polym Sci. 87:107–164. Mirghani ME. 2013. Assessment of cytotoxicity and toxicity for
Faggian M, Sut S, Perissutti B, Baldan V, Grabnar I, Dall’Acqua S. phosphonium-based deep eutectic solvents. Chemosphere.
2016. Natural deep eutectic solvents (NADES) as a tool for bio- 93(2):455–459.
availability improvement: pharmacokinetics of rutin dissolved in Hayyan M, Hashim MA, Hayyan A, Al-Saadi MA, AlNashef IM,
proline/glycine after oral administration in rats: possible appli- Mirghani MES, Saheed OK. 2013. Are deep eutectic solvents
cation in nutraceuticals. Molecules. 21(11):1531. benign or toxic? Chemosphere. 90(7):2193–2195.
Fiala S, Jones SA, Brown MB. 2010. A fundamental investigation Hayyan M, Looi CY, Hayyan A, Wong WF, Hashim MA. 2015. In
into the effects of eutectic formation on transmembrane trans- vitro and in vivo toxicity profiling of ammonium-based deep
port. Int J Pharm. 393(1–2):68–73. eutectic solvents. PLoS One. 10(2):e0117934.
Florindo C, Oliveira FS, Rebelo LPN, Fernandes AM, Marrucho IM. Hayyan M, Mbous YP, Looi CY, Wong WF, Hayyan A, Salleh Z,
2014. Insights into the synthesis and properties of deep eutec- Mohd-Ali O. 2016. Natural deep eutectic solvents: cytotoxic pro-
tic solvents based on cholinium chloride and carboxylic acids. file. Springerplus. 5(1):913.
ACS Sustainable Chem Eng. 2(10):2416–2425. Jablonsky M, Skulcova A, Sima J. 2019. Use of deep eutectic sol-
Francisco M, van den Bruinhorst A, Kroon MC. 2013. Low-transi- vents in polymer chemistry–a review. Molecules. 24(21):3978.
tion-temperature mixtures (LTTMs): a new generation of ski T, Przybyłek M, Cysewski P. 2019. Natural deep eutectic
Jelin
designer solvents. Angew Chem Int Ed. 52(11):3074–3085. solvents as agents for improving solubility, stability and deliv-
Gajraj NM, Pennant JH, Watcha MF. 1994. Eutectic mixture of local ery of curcumin. Pharm Res. 36(8):116.
anesthetics (EMLAV R ) cream. Anesth Analg. 78(3):574–583.
Jin Y, Khadka DB, Cho W-J. 2016. Pharmacological effects of ber-
Gala U, Chuong MC, Varanasi R, Chauhan H. 2015. berine and its derivatives: a patent update. Expert Opin Ther
Characterization and comparison of lidocaine-tetracaine and Pat. 26(2):229–243.
lidocaine-camphor eutectic mixtures based on their crystalliza- Juneidi I, Hayyan M, Hashim MA. 2018. Intensification of biotrans-
tion and hydrogen-bonding abilities. AAPS PharmSciTech. 16(3): formations using deep eutectic solvents: overview and outlook.
528–536. Process Biochem. 66:33–60.
Galvao J, Davis B, Tilley M, Normando E, Duchen MR, Cordeiro MF. Kang L, Jun HW, McCall JW. 2000. Physicochemical studies of lido-
2014. Unexpected low-dose toxicity of the universal solvent caine-menthol binary systems for enhanced membrane trans-
DMSO. FASEB J. 28(3):1317–1330.
port. Int J Pharm. 206(1–2):35–42.
Garcıa-Argu€elles S, Serrano MC, Gutierrez MC, Ferrer ML, Yuste L,
Kaplun-Frischoff Y, Touitou E. 1997. Testosterone skin permeation
Rojo F, del Monte F. 2013. Deep eutectic solvent-assisted syn-
enhancement by menthol through formation of eutectic with
thesis of biodegradable polyesters with antibacterial properties.
drug and interaction with skin lipids. J Pharm Sci. 86(12):
Langmuir. 29(30):9525–9534.
1394–1399.
Greaves TL, Drummond CJ. 2008. Ionic liquids as amphiphile self-
Karande P, Mitragotri S. 2009. Enhancement of transdermal drug
assembly media. Chem Soc Rev. 37(8):1709–1726.
delivery via synergistic action of chemicals. Biochim Biophys
Greaves TL, Drummond CJ. 2013. Solvent nanostructure, the sol-
Acta 1788(11):2362–2373.
vophobic effect and amphiphile self-assembly in ionic liquids.
Karimi M, Hesaraki S, Alizadeh M, Kazemzadeh A. 2016. Synthesis
Chem Soc Rev. 42(3):1096–1120.
of calcium phosphate nanoparticles in deep-eutectic choline
Grodowska K, Parczewski A. 2010. Organic solvents in the pharma-
chloride–urea medium: investigating the role of synthesis tem-
ceutical industry. Acta Pol Pharm. 67(1):3–12.
Guajardo N, Domınguez de Marıa P, Ahumada K, Schrebler RA, perature on phase characteristics and physical properties.
Ramırez-Tagle R, Crespo FA, Carlesi C. 2017. Water as cosolvent: Ceram Int. 42(2):2780–2788.
nonviscous deep eutectic solvents for efficient lipase-catalyzed Keramatnia F, Jouyban A, Valizadeh H, Delazar A, Shayanfar A.
esterifications. ChemCatChem. 9(8):1393–1396. 2016. Ketoconazole ionic liquids with citric and tartaric acid:
Guo X, Mei N. 2014. Assessment of the toxic potential of gra- synthesis, characterization and solubility study. Fluid Phase
phene family nanomaterials. J Food Drug Anal. 22(1):105–115. Equilib. 425:108–113.
Gutierrez A, Aparicio S, Atilhan M. 2019. Design of arginine-based Kudłak B, Owczarek K, Namiesnik J. 2015. Selected issues related
therapeutic deep eutectic solvents as drug solubilization to the toxicity of ionic liquids and deep eutectic solvents—a
vehicles for active pharmaceutical ingredients. Phys Chem review. Environ Sci Pollut Res. 22(16):11975–11992.
Chem Phys. 21(20):10621–10634. Kumari A, Yadav SK, Yadav SC. 2010. Biodegradable polymeric
Gutierrez A, Atilhan M, Aparicio S. 2018. A theoretical study on nanoparticles based drug delivery systems. Colloids Surf B
lidocaine solubility in deep eutectic solvents. Phys Chem Chem Biointerfaces 75(1):1–18.
Phys. 20(43):27464–27473. Lazerges M, Rietveld IB, Corvis Y, Ceolin R, Espeau P. 2010.
Gutierrez M C, Ferrer M L, Mateo C. R, del Monte F. 2009. Freeze- Thermodynamic studies of mixtures for topical anesthesia: lido-
drying of aqueous solutions of deep eutectic solvents: a suit- caine–salol binary phase diagram. Thermochim Acta 497(1–2):
able approach to deep eutectic suspensions of self-assembled 124–128.
structures. Langmuir. 25(10):5509–5515. Li Z, Lee PI. 2016. Investigation on drug solubility enhancement
Hammond OS, Bowron DT, Edler KJ. 2017. The effect of water using deep eutectic solvents and their derivatives. Int J Pharm.
upon deep eutectic solvent nanostructure: an unusual transi- 505(1–2):283–288.
tion from ionic mixture to aqueous solution. Angew Chem Int Li Q, Wang J, Lei N, Yan M, Chen X, Yue X. 2018. Phase behaviours
Ed. 56(33):9782–9785. of a cationic surfactant in deep eutectic solvents: from micelles
Han S, Samanta A, Xie X, Huang L, Peng J, Park S J, Teh D B L, to lyotropic liquid crystals. Phys Chem Chem Phys. 20(17):
Choi Y, Chang Y-T, All A H, et al. 2017. Gold and hairpin DNA 12175–12181.
794 S. EMAMI AND A. SHAYANFAR

Li X, Wang Y-K, Song Z-Q, Du Z-Q, Yang C-X. 2016. Dimethyl sulf- review of toxicity data. PDA J Pharm Sci Technol. 54(6):
oxide perturbs cell cycle progression and spindle organization 456–469.
in porcine meiotic oocytes. PLoS One. 11(6):e0158074. Mukesh C, Mondal D, Sharma M, Prasad K. 2014. Choline chlori-
Liang R, Wei M, Evans DG, Duan X. 2014. Inorganic nanomaterials de–thiourea, a deep eutectic solvent for the production of chi-
for bioimaging, targeted drug delivery and therapeutics. Chem tin nanofibers. Carbohydr Polym. 103:466–471.
Commun. 50(91):14071–14081. Mukesh C, Prasad K. 2015. Formation of multiple structural for-
Liao H-G, Jiang Y-X, Zhou Z-Y, Chen S-P, Sun S-G. 2008. Shape- mats of DNA in a bio-deep eutectic solvent. Macromol Chem
controlled synthesis of gold nanoparticles in deep eutectic sol- Phys. 216(10):1061–1066.
vents for studies of structure–functionality relationships in elec- Mukesh C, Upadhyay KK, Devkar RV, Chudasama NA, Raol GG,
trocatalysis. Angew Chem Int Ed. 47(47):9100–9103. Prasad K. 2016. Preparation of a noncytotoxic hemocompatible
Liu Y-T, Chen Y-A, Xing Y-J. 2014. Synthesis and characterization ion gel by self-polymerization of HEMA in a green deep eutec-
of novel ternary deep eutectic solvents. Chin Chem Lett. 25(1): tic solvent. Macromol Chem Phys. 217(17):1899–1906.
104–106. Naik A, Kalia YN, Guy RH. 2000. Transdermal drug delivery: over-
Liu Y, Friesen JB, McAlpine JB, Lankin DC, Chen S-N, Pauli GF. coming the skin’s barrier function. Pharm Sci Technol Today
2018. Natural deep eutectic solvents: properties, applications, 3(9):318–326.
and perspectives. J Nat Prod. 81(3):679–690. Nanda A, Nanda S, Ghilzai NM. 2006. Current developments using
Liu C, Hu J, Sui H, Zhao Q, Zhang X, Wang W. 2017. Enhanced emerging transdermal technologies in physical enhancement
skin permeation of glabridin using eutectic mixture-based methods. CDD. 3(3):233–242.
nanoemulsion. Drug Deliv Transl Res. 7(2):325–332. Newman AW, Reutzel-Edens SM, Zografi G. 2008. Characterization
Lu C, Cao J, Wang N, Su E. 2016. Significantly improving the solu- of the “hygroscopic” properties of active pharmaceutical ingre-
bility of non-steroidal anti-inflammatory drugs in deep eutectic dients. J Pharm Sci. 97(3):1047–1059.
solvents for potential non-aqueous liquid administration. Med Nyqvist-Mayer AA, Brodin AF, Frank SG. 1986. Drug release studies
Chem Commun. 7(5):955–959. on an oil-water emulsion based on a eutectic mixture of lido-
Madhumathi K, Sudheesh Kumar PT, Abhilash S, Sreeja V, Tamura caine and prilocaine as the dispersed phase. J Pharm Sci. 75(4):
H, Manzoor K, Nair SV, Jayakumar R. 2010. Development of 365–373.
novel chitin/nanosilver composite scaffolds for wound dressing Olivares B, Martinez F, Rivas L, Calderon C, M Munita J, R
applications. J Mater Sci: Mater Med. 21(2):807–813. Campodonico P. 2018. A natural deep eutectic solvent formu-
Mai Y, Eisenberg A. 2012. Self-assembly of block copolymers. lated to stabilize beta-lactam antibiotics. Sci Rep. 8(1):14900.
Chem Soc Rev. 41(18):5969–5985. Pal M, Rai R, Yadav A, Khanna R, Baker GA, Pandey S. 2014. Self-
Marrucho IM, Branco LC, Rebelo LP. 2014. Ionic liquids in pharma- aggregation of sodium dodecyl sulfate within (choline chlori-
ceutical applications. Annu Rev Chem Biomol Eng. 5(1): de þ urea) deep eutectic solvent. Langmuir. 30(44):
527–546. 13191–13198.
McDonald S, Murphy T, Imberti S, Warr GG, Atkin R. 2018. Pal M, Singh RK, Pandey S. 2015. Evidence of self-aggregation of
Amphiphilically nanostructured deep eutectic solvents. J Phys cationic surfactants in a choline chloride þ glycerol deep eutec-
Chem Lett. 9(14):3922–3927. tic solvent. ChemPhysChem. 16(12):2538–2542.
Mi F-L, Shyu S-S, Lin Y-M, Wu Y-B, Peng C-K, Tsai Y-H. 2003. Palmelund H, Andersson MP, Asgreen CJ, Boyd BJ, Rantanen J,
Chitin/PLGA blend microspheres as a biodegradable drug deliv- Lo€bmann K. 2019. Tailor-made solvents for pharmaceutical use?
ery system: a new delivery system for protein. Biomaterials Experimental and computational approach for determining
24(27):5023–5036. solubility in deep eutectic solvents (DES). Int J Pharma X. 1:
Mizuuchi H, Jaitely V, Murdan S, Florence AT. 2008. Room tem- 100034.
perature ionic liquids and their mixtures: potential pharmaceut- Pandey A, Pandey S. 2014. Solvatochromic probe behavior within
ical solvents. Eur J Pharm Sci. 33(4–5):326–331. choline chloride-based deep eutectic solvents: effect of tem-
Mondal D, Bhatt J, Sharma M, Chatterjee S, Prasad K. 2014. A perature and water. J Phys Chem B. 118(50):14652–14661.
facile approach to prepare a dual functionalized DNA based Park CW, Mansour HM, Oh TO, Kim JY, Ha JM, Lee BJ, Chi SC,
material in a bio-deep eutectic solvent. Chem Commun. 50(30): Rhee YS, Park ES. 2012. Phase behavior of itraconazole-phenol
3989–3992. mixtures and its pharmaceutical applications. Int J Pharm.
Mondal D, Sharma M, Mukesh C, Gupta V, Prasad K. 2013. 436(1–2):652–658.
Improved solubility of DNA in recyclable and reusable bio- Pham QD, Bjo €rklund S, Engblom J, Topgaard D, Sparr E. 2016.
based deep eutectic solvents with long-term structural and Chemical penetration enhancers in stratum corneum—relation
chemical stability. Chem Commun. 49(83):9606–9608. between molecular effects and barrier function. J Control
Moniruzzaman M, Tahara Y, Tamura M, Kamiya N, Goto M. 2010. Release. 232:175–187.
Ionic liquid-assisted transdermal delivery of sparingly soluble Plechkova NV, Seddon KR. 2008. Applications of ionic liquids in
drugs. Chem Commun. 46(9):1452–1454. the chemical industry. Chem Soc Rev. 37(1):123–150.
Morrison HG, Sun CC, Neervannan S. 2009. Characterization of Porst H, Burri A. 2017. Fortacin spray for the treatment of prema-
thermal behavior of deep eutectic solvents and their potential ture ejaculation. Urologia. 84(2 suppl):1–10.
as drug solubilization vehicles. Int J Pharm. 378(1–2):136–139. Pouton CW. 2006. Formulation of poorly water-soluble drugs for
Mota-Morales JD, Sanchez-Leija RJ, Carranza A, Pojman JA, del oral administration: physicochemical and physiological issues
Monte F, Luna-Barcenas G. 2018. Free-radical polymerizations of and the lipid formulation classification system. Eur J Pharm Sci.
and in deep eutectic solvents: Green synthesis of functional 29(3–4):278–287.
materials. Prog Polym Sci. 78:139–153. Prabhakaran MP, Nair AS, Kai D, Ramakrishna S. 2012. Electrospun
Mottu F, Laurent A, Rufenacht DA, Doelker E. 2000. Organic sol- composite scaffolds containing poly(octanediol-co-citrate) for
vents for pharmaceutical parenterals and embolic liquids: a cardiac tissue engineering. Biopolymers. 97(7):529–538.
PHARMACEUTICAL DEVELOPMENT AND TECHNOLOGY 795

Pradeepkumar P, Elgorban AM, Bahkali AH, Rajan M. 2018. Natural Serrano MC, Carbajal L, Ameer GA. 2011. Novel biodegradable
solvent-assisted synthesis of amphiphilic co-polymeric nanomi- shape-memory elastomers with drug-releasing capabilities. Adv
celles for prolonged release of camptothecin delivery. New J Mater. 23(19):2211–2215.
Chem. 42(12):10366–10375. Serrano MC, Gutierrez MC, Jimenez R, Ferrer ML, Monte F. 2012.
Prausnitz MR, Mitragotri S, Langer R. 2004. Current status and Synthesis of novel lidocaine-releasing poly(diol-co-citrate) elas-
future potential of transdermal drug delivery. Nat Rev Drug tomers by using deep eutectic solvents. Chem Commun. 48(4):
Discov. 3(2):115–124. 579–581.
Pujara N, Jambhrunkar S, Wong KY, McGuckin M, Popat A. 2017. Shamseddin A, Crauste C, Durand E, Villeneuve P, Dubois G,
Enhanced colloidal stability, solubility and rapid dissolution of Durand T, Vercauteren J, Veas F. 2017. Resveratrol formulated
resveratrol by nanocomplexation with soy protein isolate. J with a natural deep eutectic solvent inhibits active matrix met-
Colloid Interface Sci. 488:303–308. alloprotease-9 in hormetic conditions. Eur J Lipid Sci Technol.
Qi X, Wei W, Li J, Liu Y, Hu X, Zhang J, Bi L, Dong W. 2015. 119(11):1700171.
Fabrication and characterization of a novel anticancer drug Sharma M, Mukesh C, Mondal D, Prasad K. 2013. Dissolution of
delivery system: salecan/poly(methacrylic acid) semi-interpene- a-chitin in deep eutectic solvents. RSC Adv. 3(39):18149–18155.
trating polymer network hydrogel. ACS Biomater Sci Eng. 1(12): Shayanfar S, Shayanfar A. 2018. Ionic liquid forms of carvedilol:
1287–1299. preparation, characterization, and solubility studies. J Pharm
Radosevic K, Bubalo MC, Srcek VG, Grgas D, Dragicevic TL, Innov. 14(4):382–390.
Redovnikovic IR. 2015. Evaluation of toxicity and biodegradabil- Shekaari H, Zafarani-Moattar MT, Shayanfar A, Mokhtarpour M.
ity of choline chloride based deep eutectic solvents. Ecotoxicol 2018. Effect of choline chloride/ethylene glycol or glycerol as
Environ Saf. 112:46–53. deep eutectic solvents on the solubility and thermodynamic
Radosevic K, Canak I, Panic M, Markov K, Bubalo MC, Frece J, properties of acetaminophen. J Mol Liq. 249:1222–1235.
Srcek VG, Redovnikovic IR. 2018. Antimicrobial, cytotoxic and Shen Q, Li X, Li W, Zhao X. 2011. Enhanced intestinal absorption
antioxidative evaluation of natural deep eutectic solvents. of daidzein by borneol/menthol eutectic mixture and microe-
Environ Sci Pollut Res Int. 25(14):14188–14196. mulsion. AAPS PharmSciTech. 12(4):1044–1049.

Radosevic K, Curko N, Gaurina Srcek V, Cvjetko Bubalo M, Sheng Q, Liu R, Zheng J. 2012. Prussian blue nanospheres synthe-
Tomasevic M, Kovacevic Ganic K, Radojcic Redovnikovic I. 2016. sized in deep eutectic solvents. Nanoscale. 4(21):6880–6886.
Natural deep eutectic solvents as beneficial extractants for Smith EL, Abbott AP, Ryder KS. 2014. Deep eutectic solvents
enhancement of plant extracts bioactivity. LWT. 73:45–51. (DESs) and their applications. Chem Rev. 114(21):11060–11082.
Rinaudo M. 2006. Chitin and chitosan: properties and applications. Stott PW, Williams AC, Barry BW. 1998. Transdermal delivery from
Prog Polym Sci. 31(7):603–632. eutectic systems: enhanced permeation of a model drug, ibu-
Rozema E, van Dam AD, Sips HCM, Verpoorte R, Meijer OC, profen. J Control Release. 50(1–3):297–308.
Kooijman S, Choi YH. 2015. Extending pharmacological dose- Stott PW, Williams AC, Barry BW. 2001. Mechanistic study into the
response curves for salsalate with natural deep eutectic sol- enhanced transdermal permeation of a model beta-blocker,
vents. RSC Adv. 5(75):61398–61401. propranolol, by fatty acids: a melting point depression effect.
Sanchez-Fernandez A, Arnold T, Jackson AJ, Fussell SL, Heenan RK, Int J Pharm. 219(1–2):161–176.
Campbell RA, Edler KJ. 2016. Micellization of alkyltrimethylam- Sut S, Faggian M, Baldan V, Poloniato G, Castagliuolo I, Grabnar I,
monium bromide surfactants in choline chloride:glycerol deep Perissutti B, Brun P, Maggi F, Voinovich D, et al. 2017. Natural
eutectic solvent. Phys Chem Chem Phys. 18(48):33240–33249. deep eutectic solvents (NADES) to enhance berberine absorp-
Sanchez-Fernandez A, Moody GL, Murfin LC, Arnold T, Jackson AJ, tion: An in vivo pharmacokinetic study. Molecules. 22(11):1921.
King SM, Lewis SE, Edler KJ. 2018. Self-assembly and surface Svenson S. 2004. Controlling surfactant self-assembly. Curr Opin
behaviour of pure and mixed zwitterionic amphiphiles in a Colloid Interface Sci. 9(3):201–212.
deep eutectic solvent. Soft Matter. 14(26):5525–5536. Takagi T, Ramachandran C, Bermejo M, Yamashita S, Yu LX,
Sanchez-Leija RJ, Pojman JA, Luna-Barcenas G, Mota-Morales JD. Amidon GL. 2006. A provisional biopharmaceutical classification
2014. Controlled release of lidocaine hydrochloride from poly- of the top 200 oral drug products in the United States, Great
merized drug-based deep-eutectic solvents. J Mater Chem B. Britain, Spain, and Japan. Mol Pharm. 3(6):631–643.
2(43):7495–7501. Talebian S, Foroughi J, Wade SJ, Vine KL, Dolatshahi-Pirouz A,
Santos NC, Figueira-Coelho J, Martins-Silva J, Saldanha C. 2003. Mehrali M, Conde J, Wallace GG. 2018. Biopolymers for antitu-
Multidisciplinary utilization of dimethyl sulfoxide: pharmaco- mor implantable drug delivery systems: recent advances and
logical, cellular, and molecular aspects. Biochem Pharmacol. future outlook. Adv Mater. 30(31):1706665.
65(7):1035–1041. Tang B, Row KH. 2013. Recent developments in deep eutectic sol-
Santos F, P.S. Leit~ao MI, C. Duarte AR. 2018. Properties of thera- vents in chemical sciences. Monatsh Chem. 144(10):1427–1454.
peutic deep eutectic solvents of l-arginine and ethambutol for Thayer AM. 2010. Finding solutions. Chem Eng News. 88(22):
tuberculosis treatment. Molecules. 24(1):55. 13–18.
Sapra B, Jain S, Tiwary AK. 2008. Percutaneous permeation Tibbitt MW, Dahlman JE, Langer R. 2016. Emerging frontiers in
enhancement by terpenes: mechanistic view. AAPS J. 10(1):120. drug delivery. J Am Chem Soc. 138(3):704–717.
Selselehjonban S, Garjani A, Osouli-Bostanabad K, Tanhaei A, Tome LIN, Bai~ao V, da Silva W, Brett C. 2018. Deep eutectic sol-
Emami S, Adibkia K, Barzegar-Jalali M. 2019. Physicochemical vents for the production and application of new materials.
and pharmacological evaluation of carvedilol-eudragitV R RS100 Appl Mater Today. 10:30–50.
electrosprayed nanostructures. Iran J Basic Med Sci. 22(5): Wagle DV, Zhao H, Baker GA. 2014. Deep eutectic solvents: sus-
547–556. tainable media for nanoscale and functional materials. Acc
Serajuddin A. 1999. Solid dispersion of poorly water-soluble drugs: Chem Res. 47(8):2299–2308.
early promises, subsequent problems, and recent break- Wang W, Cai Y, Liu Y, Zhao Y, Feng J, Liu C. 2017. Microemulsions
throughs. J Pharm Sci. 88(10):1058–1066. based on paeonol-menthol eutectic mixture for enhanced
796 S. EMAMI AND A. SHAYANFAR

transdermal delivery: formulation development and in vitro biocompatibility of graphene as a potential nano-carrier for tar-
evaluation. Artif Cells Nanomed Biotechnol. 45(6):1–6. geted drug delivery using natural deep eutectic solvents. Chem
Wang H, Gurau G, Shamshina J, Cojocaru OA, Janikowski J, Eng Sci. 195:95–106.
MacFarlane DR, Davis JH, Rogers RD. 2014. Simultaneous mem- Zainal-Abidin MH, Hayyan M, Ngoh GC, Wong WF. 2020.
brane transport of two active pharmaceutical ingredients by Doxorubicin loading on functional graphene as a promising
charge assisted hydrogen bond complex formation. Chem Sci. nanocarrier using ternary deep eutectic solvent systems. ACS
5(9):3449–3456. Omega. 5(3):1656–1668.
Wen Q, Chen J-X, Tang Y-L, Wang J, Yang Z. 2015. Assessing the Zakrewsky M, Lovejoy KS, Kern TL, Miller TE, Le V, Nagy A,
toxicity and biodegradability of deep eutectic solvents. Goumas AM, Iyer RS, Del Sesto RE, Koppisch AT, et al. 2014.
Chemosphere. 132:63–69. Ionic liquids as a class of materials for transdermal delivery and
Wilkes JS, Zaworotko MJ. 1992. Air and water stable 1-ethyl-3- pathogen neutralization. Proc Natl Acad Sci USA. 111(37):
methylimidazolium based ionic liquids. J Chem Soc, Chem 13313–13318.
Commun. (13):965–967. Zhang Y, Hou Z, Ge Y, Deng K, Liu B, Li X, Li Q, Cheng Z, Ma P, Li
Williams HD, Sahbaz Y, Ford L, Nguyen T-H, Scammells PJ, Porter C, et al. 2015. DNA-hybrid-gated photothermal mesoporous sil-
C. 2014. Ionic liquids provide unique opportunities for oral ica nanoparticles for NIR-responsive and aptamer-targeted drug
drug delivery: structure optimization and in vivo evidence of delivery. ACS Appl Mater Interfaces. 7(37):20696–20706.
utility. Chem Commun. 50(14):1688–1690. Zhang S, Marini DM, Hwang W, Santoso S. 2002. Design of nano-
Williams HD, Trevaskis NL, Charman SA, Shanker RM, Charman structured biological materials through self-assembly of pepti-
WN, Pouton CW, Porter C. 2013. Strategies to address low drug des and proteins. Curr Opin Chem Biol. 6(6):865–871.
solubility in discovery and development. Pharmacol Rev. 65(1): Zhang H, Wang Y, Xu K, Li N, Wen Q, Yang Q, Zhou Y. 2016.
315–499. Ternary and binary deep eutectic solvents as a novel extraction
Wolbert F, Brandenbusch C, Sadowski G. 2019. Selecting exci- medium for protein partitioning. Anal Methods. 8(46):
pients forming therapeutic deep eutectic systems—a mechanis- 8196–8207.
tic approach. Mol Pharm. 16(7):3091–3099. Zhang J, Wu T, Chen S, Feng P, Bu X. 2009. Versatile structure-
Woolfson AD, Malcolm RK, Campbell K, Jones DS, Russell JA. 2000. directing roles of deep-eutectic solvents and their implication
Rheological, mechanical and membrane penetration properties in the generation of porosity and open metal sites for gas stor-
of novel dual drug systems for percutaneous delivery. J Control age. Angew Chem Int Ed. 48(19):3486–3490.
Release. 67(2–3):395–408. Zhao H, Baker GA, Holmes S. 2011. New eutectic ionic liquids for
Yadav A, Pandey S. 2014. Densities and viscosities of (choline lipase activation and enzymatic preparation of biodiesel. Org
chloride þ urea) deep eutectic solvent and its aqueous mixtures Biomol Chem. 9(6):1908–1916.
in the temperature range 293.15 K to 363.15 K. J Chem Eng Zhao B-Y, Xu P, Yang F-X, Wu H, Zong M-H, Lou W-Y. 2015.
Data. 59(7):2221–2229. Biocompatible deep eutectic solvents based on choline chlor-
Yang C, Wang X, Yao X, Zhang Y, Wu W, Jiang X. 2015. Hyaluronic ide: characterization and application to the extraction of rutin
acid nanogels with enzyme-sensitive cross-linking group for from Sophora japonica. ACS Sustainable Chem Eng. 3(11):
drug delivery. J Control Release. 205:206–217. 2746–2755.
Zahn S. 2017. Deep eutectic solvents: Similia similibus solvuntur? Zhaojie M, Ming Z, Shengnan W, Xiaojia B, Hatch GM, Jingkai G, Li
Phys Chem Chem Phys. 19(5):4041–4047. C. 2014. Amorphous solid dispersion of berberine with absorp-
Zainal-Abidin MH, Hayyan M, Ngoh GC, Wong WF. 2019. From tion enhancer demonstrates a remarkable hypoglycemic effect
nanoengineering to nanomedicine: a facile route to enhance via improving its bioavailability. Int J Pharm. 467(1–2):50–59.
Copyright of Pharmaceutical Development & Technology is the property of Taylor & Francis
Ltd and its content may not be copied or emailed to multiple sites or posted to a listserv
without the copyright holder's express written permission. However, users may print,
download, or email articles for individual use.

You might also like