Sunteți pe pagina 1din 16

Journal of Pharmaceutical Investigation Online ISSN 2093-6214

DOI 10.1007/s40005-016-0299-z Print ISSN 2093-5552

REVIEW

Analysis and optimization of drug solubility to improve


pharmacokinetics
Raj Kumar Thapa1 · Han-Gon Choi2 · Jong Oh Kim1 · Chul Soon Yong1 

Received: 1 November 2016 / Accepted: 21 December 2016


© The Korean Society of Pharmaceutical Sciences and Technology 2017

Abstract Solubility measurement is one of the key ele- Keywords Bioavailability · Carrier technology ·
ments that need to be considered in drug discovery and Optimization · Pharmacokinetics · Solubility
development process. Hence, its appropriate analysis and
optimization is necessary to enhance the drug’s pharma-
cokinetic and therapeutic effects. In this article, differ- Introduction
ent methods used for solubility analysis and optimization
such as shake flask, potentiometric, turbidimetric, compu- Development of new drug candidates (NDCs) is a complex
tational, miniature device and high throughput automated and lengthy process. Although tremendous investments
methods have been summarized. Additionally, the effect have been made in the field of drug discovery, only a few
of solubility optimization methods (physical, chemical NDCs have been produced so far (Ruell and Avdeedf 2003).
and carrier technology modifications) on pharmacokinetic There are a number of key properties that determine the
enhancement has also been elucidated. New fast, feasible fate of the clinical utilization of an NDC which include but
and automated methods of solubility analysis are necessary not limited to solubility, permeability, pKa and metabolism
for its accurate measurement as well as for minimization of (Ruell and Avdeedf 2003). Among them, the most impor-
errors posed by traditional methods. Among the physical, tant one is the solubility. Generally solubility data are used
chemical and carrier modification methods; higher poten- in decision making regarding the developability of NDCs
tial is exhibited by nanoparticulate drug delivery carriers from earliest drug discovery stages to the development pro-
for optimization of drug solubility and enhancement of cess. Possible changes in purity and synthesis in the devel-
pharmacokinetics. Based on the drug characteristics and opment process bring changes in solubility that necessitates
delivery requirement, appropriate method can be chosen multiple events of solubility measurement. Following drug
for efficient solubility and pharmacokinetics enhancement development the solubility of a drug in physiological media
of lipophilic drugs, especially of BCS class II and class IV. directly impacts its pharmacokinetics, which is ultimately
related to the therapeutic efficacy of the drug. Furthermore,
most of the available drugs are hydrophobic in nature that
limits their solubility in the physiological fluids leading to
* Jong Oh Kim their limited therapeutic effects. Hence, in this review, we
jongohkim@yu.ac.kr will mainly deal with solubility aspects of drugs along with
* Chul Soon Yong its measurement methods. Moreover, the methods of solu-
csyong@yu.ac.kr bility enhancement and optimization have been elucidated.
1
College of Pharmacy, Yeungnam University, 214-1
Solubility aspects
Dae-dong, Gyeongsanbuk-do, Gyeongsan-si 712-749,
South Korea
2 Solubility is defined as the property of a solid, liquid or
College of Pharmacy & Institute of Pharmaceutical Science
and Technology, Hanyang University, 55 Hanyangdaehak-ro, gaseous chemical substance, called solute, to dissolve in
Ansan, Sangnok-gu 426-791, South Korea another solid, liquid or gaseous chemical substance, called

13
Vol.:(0123456789)
R. K. Thapa et al.

solvent, to form a homogeneous solution of the solute in the


solvent. Fundamentally, solubility depends on the solvent
used as well as temperature and pressure (Lachman et  al.
1986). International Union of Pure and Applied Chemistry
(IUPAC) define solubility as the analytical composition of
a saturated solution, expressed in terms of the proportion of
a designated solute in a designated solvent (IUPAC 1997).
Pharmaceutical dosage form of drugs and their admin-
istration route is mostly affected by its solubility. Highly
soluble drugs are usually formulated as sustained release
dosage form for long term drug delivery. In contrast, poorly
soluble drugs are prepared with lipids to enhance their
uptake. Soluble drugs can be administered through any
administration routes in the body. However, hydrophobic
drugs in their native form can only be formed as suspension
or tablet formulations that can be administered either orally
or subcutaneously as a depot. Drug absorption is a com-
plex process that can be affected by several factors such as
physicochemical, physiological and formulation aspects.
Among them the rate and extent of solubilization/dissolu-
tion of drug in target site is one of the key factors (Dahan Fig. 1 BCS classification system
and Miller 2012). However, in the current drug discovery
programs, poor aqueous solubility is the main hindrance
because 40–70% of all NDCs and available drugs exhibit can be accomplished in the drug or can be formulated in a
minimum aqueous solubility resulting in partial absorption suitable carrier system to enhance the drug solubility.
from the gastrointestinal tract (Dahan and Miller 2012).
Interrelationship between solubility and pharmacokinetics

Biopharmaceutics classification system (BCS) Pharmacokinetics is defined as the study of the time course
of drugs and their metabolites through the body. It includes
Extensive research on fundamental parameters of oral determination of absorption, distribution, metabolism and
absorption (i.e. drug’s aqueous solubility and gastrointes- excretion parameters of drug in/from the body (Kufe et al.
tinal permeability) by Amidon et  al., led to the develop- 2003). Solubility of a drug plays a vital role in determining
ment of BCS, wherein drugs are classified into four groups the fate of its pharmacokinetics. Based on the BCS, major
(class I–IV) (Amidon et al. 1995). Utilizing solubility and role of solubility is evident in Class II and IV drugs.
permeability as the key factors, BCS allows the prediction Absorption of a drug is directly dependent on its solubil-
of pharmacokinetic performance of a drug in vivo based on ity as only a soluble drug can be absorbed into the cells.
in vitro measurements of solubility and permeability (Cus- Solubility of a drug is dependent on the intrinsic charac-
todio et al. 2008). Figure 1 depicts the BCS as defined by teristics (lipophilic or hydrophilic nature) of the drug mol-
the FDA after Amidon et al., in which Class I compounds ecule. Although hydrophilic drug molecules can be eas-
possess high solubility and high permeability; Class II ily solubilized in body fluids, their penetration into blood
compounds possess low solubility and high permeability; circulation is limited because of lipophilic nature of the
Class III compounds possess high solubility and low per- biological membrane barrier that inhibits penetration and
meability; and Class IV compounds possess low solubility vice-versa for the lipophilic ones (Alavijeh et  al. 2005).
and low permeability characteristics (U.S. Food and Drug Enhancement of drug solubility is therefore an important
Administration 2015). A drug substance is considered issue, especially for lipophilic drugs, in order to achieve
highly soluble when its highest strength is soluble in 250 better absorption and therapeutic effects.
mL or less of aqueous media over the pH range of 1–6.8 Liver and kidneys are the major organs responsible for
(U.S. Food and Drug Administration 2015). BCS is very drug elimination from the body. Both the organs share
useful in the early stages of drug development and provides metabolic and excretory functions, however, liver is mainly
avenues for improving the predictability of oral absorp- responsible for metabolism and kidney for elimination. Sol-
tion and disposition of NDCs (Custodio et al. 2008). Also, ubility of a drug plays a vital role in determining the drug
based on the classification system, suitable modifications disposition. Low solubility (lipophilic) drugs are generally

13
Analysis and optimization of drug solubility to improve pharmacokinetics

bound to plasma proteins, distributed to a greater extent pH value against the consumed volume of acid/base gives
throughout body and metabolized to a greater extent in the potentiometric titration curve which provides ultimate
the liver. Conversely, high solubility (hydrophilic) drugs information about solubility (Glomme et al. 2005).
exhibit limited distribution and rapid elimination mostly by
renal excretion (Alavijeh et al. 2005).
Hepatic metabolism of poorly water soluble drug occurs Turbidimetry
in two phases, viz. biotransformation and conjugation. Bio-
transformation involves processes like oxidation, reduc- In this method, firstly the compound is dissolved in organic
tion, or hydrolysis that convert lipophilic compounds to solvent [usually dimethyl sulfoxide (DMSO)] which is then
hydrophilic one. The major hepatic enzyme responsible added to a pH 7 buffered solution at intervals of 1  min.
for biotransformation is cytochrome P450. In conjugation Some further aliquots of drug solution are added after the
process, generally the biotransformed compounds receive a first detection of turbidity by light scattering. A plot for
molecular attachment (e.g. glucoronate) that aids in trans- volume added against turbidity can be plotted, from which
port within the body (DiPiro et al. 2010). solubility is estimated by back-extrapolation (Glomme
Excretion may occur for a biotransformed drug (usu- et al. 2005). 50–300 samples can be measured per day by
ally low solubility drugs) or a drug that remains unchanged this method. Major drawback of this method is the use
in the body (usually high solubility drugs). Three main of DMSO that can increase solubility to an unknown or
mechanisms affect the excretion of drugs through kid- unpredictable extent leading to an estimate of kinetic rather
ney which include glomerular filtration, tubular secretion than thermodynamic solubility (Avdeef and Testa 2002).
and tubular reabsorption. Poorly soluble drugs can mostly Another problem is lack of information about the form of
undergo tubular reabsorption that occurs passively in the precipitating drug since fastest precipitating form may not
distal tubules whereas highly water soluble ones are easily be the most stable form (e.g. characteristics shown by poly-
excreted. The most important routes of excretion for drugs morphs). Additionally, pH shifts and possible impurities
and their metabolites include the urine and bile (DiPiro are not detected which might lead to poor estimates.
et al. 2010).
Computational models

Methods of solubility measurement A variety of computational models are developed to pre-


dict aqueous solubility of different compounds. Fairly good
Shake flask method predictions of solubility can be performed by simple linear
regression models using physicochemical characteristics
This is the most reliable and commonly used experimen- like size, lipophilicity and/or hydrogen-bonding capacity
tal method that determines the solubility of a drug at equi- along with more complex neural network models based on
librium (Bergstrӧm et al. 2002). Excess sample is added to electrotopologic descriptors (Huuskonen et al. 1998).
the medium and resulting suspension is shaken for appro-
priate time (24–48  h) at a defined temperature to form a
saturated solution. Once the solution is equilibrated, suc- Miniature device
cessive filtration and quantification of drug’s concentration
is performed, which provides its thermodynamic solubility This device provides a unique method for testing aqueous
(Glomme et al. 2005). The solubility of stable form of drug and non-aqueous equilibrium solubility in a microscale
can be determined with minimal risks of obtaining a super- testing. Very small quantity, approximately 1  mg, is suffi-
saturated solution. Drug concentration can be determined cient for determination of entire pH-solubility profile which
with high performance liquid chromatography (HPLC) that makes it suitable during lead optimization and candidate
can easily separate the impurities from the substance of selection in early drug discovery (when compound is in a
interest, thus minimizing the chances of spurious results. limited quantity). In this method, drug slurry is prepared
and circulated into the tubing at a speed of 4.0  mL/min.
Potentiometric titration Filtrate is then collected from one end of tube at predeter-
mined time and the concentration of dissolved compound is
It is a titration method based on precipitation of desired determined by HPLC. Unlike shake-flask method, the drug
substance leading to a characteristic shift in the middle of slurry is continually filtered through the membrane that
the titration curve. To a solution of ionizable substance leads to minimization of adsorption. Also, rapid equilib-
(drug), accurate volumes of standardized acid or base are rium attained in the device enables short turnaround time
added and pH values are continuously monitored. A plot of (Chen et al. 2004).

13
R. K. Thapa et al.

New methods for solubility measurement Physical modifications

Shake-flask is the most commonly used method for esti- Different methods can be employed for physical modifi-
mation of the thermodynamic solubility of a drug, but it cation of drugs in order to improve their solubility which
is a time-consuming and non-automated process. How- comprises particle size reduction, modification of crystal
ever, a fast, feasible and automated method is preferred habit, and eutectic mixtures.
for the high throughput screening studies. Therefore, new
synthetic methods for solubility measurement have been Particle size reduction
developed, which include the dynamic method (Wang et al.
2008), last crystal disappearance method (Hao et al. 2005) Drug solubility is intrinsically related to particle size of the
and laser monitoring technique (Li et al. 2006). The princi- drug, because smaller particle size ensures an increase in
ple of solubility measurement by synthetic method involves the surface area available for solvation. Particle size reduc-
the disappearance of solid drug from the solute and solvent tion is, therefore, one of the oldest methods to improve drug
mixture that is monitored by laser beam (Li et  al. 2006). solubility, and thereby, bioavailability and pharmacokinet-
Disappearance of the drug occurs either by adding the sol- ics (Williams et  al. 2013). According to the Noyes–Whit-
vent or by increasing the temperature, which is then moni- ney equation, dissolution rate of a drug is increased by
tored by laser. Jouyban-Gharamaleki et  al. recently devel- decrease in particle size and increase in surface area. How-
oped an automated system for determining drug solubility ever, decrease in particle size has little effect on solubility
based on laser monitoring technique (Jouyban-Gharamaleki because of no alteration in the solid-state properties of the
et al. 2015). The system was aimed to be accurate, precise, particles. Ostwald–Freundlich equation explains that solu-
cheap and feasible for determination of drug solubility. The bility significantly increases by reducing particle size below
automated system was able to accurately measure the solu- 1 µm as it increases solvation pressure and also causes dis-
bility of acetaminophen at different temperatures and this ruption of solute–solute interaction that eases solubilization
setup has been proposed for use in pharmaceutical indus- of drug (Junghanns et  al. 2008). Additionally, nanometer
try and academia for faster drug solubility measurements sized particles are now being widely studied for formula-
(Jouyban-Gharamaleki et al. 2015). tion of drugs with low aqueous solubility (Leleux and Wil-
Quartz crystal microbalance is another new technique liams 2014).
introduced for the accurate and reliable determination of
saturation solubility of drugs. Peizoelectricity is responsi- Nanosuspension Nanosuspension is the abbreviated form
ble for vibration of quartz crystal at a resonant frequency of nanocrystal suspension in which pure drug crystals are
infinitely upon stimulation of an alternative potential (Revi- stabilized with minimum stabilizers. The submicron sized
akine et  al. 2011). In addition, resonant frequency varies crystalline particles are stabilized in liquid medium, usually
depending on the mass deposited on the crystal and physi- water and can be prepared by either bottom-up (precipita-
cal property changes around the crystal. The key strength tion) or top-down (size reduction) techniques (Ali et  al.
of this system is its ability of simultaneous detection of 2009). General methods utilized for production of nanosus-
subtle property changes in both solid and liquid phases (Liu pensions include precipitation, pearl milling and high pres-
et al. 2015). sure homogenization.
High pressure homogenization is the most commonly
applied top-down technique for the preparation of nanosus-
pensions in food, cosmetics and pharmaceutical industries.
Optimization of drug solubility and enhancement In this method, the suspension is forced through a thin gap
of pharmacokinetics (5–20 µm) at a high velocity leading to particle size reduc-
tion (Van Eerdenbrugh et al. 2009). In order to prevent the
In order to achieve desired therapeutic effect, the drug blockage of gap during homogenization, pre-milling is
needs to be absorbed in the blood circulation and reach recommended in a sequence of two cycles at 100 bar, two
the target site. Optimization of drug solubility, thereby, is cycles at 200  bar, two cycles at 500  bar, and two cycles
very important to enhance the therapeutic efficacy. Drugs at 1000  bar (Patravale et  al. 2004). Nano-precipitation is
especially from BCS class II and IV require optimization another simple and economical method for preparation of
as they possess limited solubility characteristics. One of the nanosuspension, which involves controlled precipitation of
measures for optimization of drug solubility includes utili- drug molecules to form nanoparticles (Verma et al. 2009).
zation of physical or chemical modification techniques, and Detroja et  al. prepared nanosupension of candesar-
the other includes use of various drug carrier systems for tan cilexetil (anti-hypertensive agent) by media milling
improvement of solubility and bioavailability (Fig. 2). method using zirconium oxide beads (Detroja et al. 2011).

13
Analysis and optimization of drug solubility to improve pharmacokinetics

Fig. 2 Different drug solubility optimization methods (physical, chemical and carrier technology modifications) utilized for the poorly water
soluble drugs

Nanosuspension increased saturation solubility of cande- by pure drug. Also, a threefold increase in the Cmax follow-
sartan cilexetil by 22.44 times as compared to bulk drug. ing oral administration of nanosuspension as compared
Further, a significant reduction in blood pressure follow- to conventional suspension was achieved (Thadkala et  al.
ing nanosuspension administration, when compared to 2014). Elsayed et al. developed nanosuspension of diacerin
plain drug suspension, was observed because of increase in by using a combined bottom-up/top-down technique
absorption and bioavailability of drug (Detroja et al. 2011). (Elsayed et al. 2014). It exhibited significantly higher satu-
Another nanoprecipitation based method was used for ration solubility and dissolution as compared to coarse drug
preparation of glimepiride nanosuspension by Yadav et al. powder with increased relative bioavailability (131.4%)
(Yadav et al. 2012). Pharmacokinetic evaluation following (Elsayed et al. 2014).
oral administration suggested 1.54-fold increase in Cmax
(maximum concentration) by nanosuspension as compared Modification of crystal habit
to conventional form suggesting an increase in the satu-
ration solubility of the drug. Also, a significantly higher In general, drugs are manufactured as crystalline solids
area under the curve (AUC) for glimepiride nanosuspen- that are characterized by a definite internal and external
sion (p < 0.01) suggested nearly twofold improvement in structure, where habit refers to external structure and poly-
relative bioavailability with sustained plasma drug levels. morphic state to internal structure of a crystal. The phys-
Further, in  vivo studies suggested better pharmacokinetic icochemical and biological properties of crystalline moiety
profile and reduced blood glucose levels by nanosuspension are influenced by its polymorphism that refers to definite
(Yadav et al. 2012). arrangement of molecules within a solid (Tiwari 2001).
Amorphous ezetimibe nanosuspensions were prepared Much attention, however, has not been paid for the crystal-
by solvent-antisolvent precipitation technique using Tween line habit unless problems with formulation are detected.
80 as a stabilizer (Thadkala et  al. 2014). The saturation Crystallization is generally employed for the purifi-
solubility of ezetimibe in nanosupension increased by four- cation of a drug using different solvents and processing
fold as compared to conventional suspension. In addition, it conditions that may affect the crystal habit, thus alter-
attained complete dissolution in 1 h compared to only 42% ing the reproducibility. Hence, detailed information on

13
R. K. Thapa et al.

crystal habit ensures maintenance of stability and phys- Co-crystallization Crystal form is crucial to the perfor-
icochemical or biological properties of a drug molecule. mance of a dosage form especially for compounds with
One of the important aspects of defining crystal habit is intrinsic barriers to drug delivery such as low aqueous solu-
to determine its influence on solubility. Drug solubility bility and dissolution in gastrointestinal media along with
depends on the size and number of crystal faces that are low permeability (Yadav et  al. 2009). Crystal engineering
exposed to the dissolution medium (Tiwari 2001). Differ- is one of the techniques to improve solubility and engineer-
ent techniques of crystal habit modification that are used ing of BCS class II, III and IV drugs. It is regarded as the
for solubility enhancement of drugs include amorphous method for design and growth of crystalline molecular sol-
forms and co-crystallization that are explained in detail. ids to improve material properties, which utilizes hydrogen
bond for directed intermolecular interaction in molecular
Amorphous forms One of the methods employed to solids. Co-crystals are based on interaction of these hydro-
enhance the solubility and dissolution rate of a crystalline gen bonds in multicomponent crystals without transfer of
drug is conversion to its amorphous form (Hancock and hydrogen ions to form salts (Yadav et al. 2009). Some of the
Zografi 1997). In contrast to crystalline counterpart, the examples of marketed drug products of racemic co-crystals
amorphous form of a drug is devoid of three dimensional include atenolol, atropine, certirazine, disopyramide, fluox-
long range order of molecular packing, leading to ther- etine, ketoprofen, omeprazole and warfarin (Sun 2013).
modynamically higher energy state as compared to crys- Shevchenko et al. studied the effect of new co-crystal of
talline form. This property of amorphous form imparts itraconazole with malic acid which improved the solubility
higher solubility and rate of dissolution as compared to and dissolution rate of itraconazole by about fivefold with-
crystalline counterpart (Hancock and Zografi 1997). The out affecting the hygroscopicity and stability (Shevchenko
higher energy state, however, causes physical instability et  al. 2012). The study revealed that co-crystallization is
of amorphous forms by possible conversion to a lower a suitable method for enhancement of dissolution rate of
energy state (stable crystalline or metastable form) over poorly water-soluble weak bases. In another study, Huang
time. This conversion can potentially negate any improve- et al. developed baicalein-nicotinamide co-crystal that sig-
ment of solubility or dissolution, and hence, stabilization nificantly improved solubility and dissolution of baicalein
of the amorphous state is necessary (Alonzo et al. 2010). as compared to its crystalline form (Huang et  al. 2014).
Stabilization can be achieved by manipulating mobility Additionally, co-crystal contributed to respective 2.49- and
of molecules, and their inter- and intra-molecular inter- 2.80-fold increase in Cmax and AUC as determined by phar-
actions using techniques such as co-amorphous forms macokinetic studies (Huang et al. 2014).
(Lӧbmann et  al. 2011) or by addition of polymers (Bal-
ani et  al. 2010). Different techniques can be utilized for Eutectic mixtures
the production of amorphous drug forms, which include
mechanical activation of crystalline mass, supercooling of Eutectic mixtures are one of the rarely used systems for the
a melt, condensation from the vapour state or rapid pre- enhancement of drug solubility and pharmacokinetics. In
cipitation from the solution. this method, eutectic formation reduces the melting point
In a study by Murdande et  al., amorphous indometha- of a delivery system. A binary eutectic refers to a mixture
cin exhibited significantly higher concentrations after 2  h of two components that do not interact with each other to
as compared to γ-crystalline form at 25 °C. The apparent form a new chemical entity, rather, at definite ratios, inhibit
solubility enhancement ratio for amorphous indometha- the process of crystallization of one another leading to a
cin was found to be 4.9 with peak concentration appearing system with lower melting point than either of the com-
within 10–20 min (Murdande et al. 2010). In another study, ponents. For the formation of a eutectic mixture, the com-
Nielsen et al. evaluated the effects of three different forms ponents must be miscible in liquid state and immiscible in
of furosemide (amorphous salt, amorphous free acid and solid state (Stott et al. 1998). Eutectic mixture reduces the
crystalline free acid) on its solubility and dissolution rate melting point and thus increases the solubility of drugs.
(Nielsen et al. 2013). In contrast to crystalline form, amor- However, exact mechanism behind enhancement of drug
phous salt and free acid forms resulted in higher apparent absorption and pharmacokinetics is still not clear.
solubility. Amorphous salt form exhibited 8- and 20-fold Formation of eutectic mixture of poly(ethylene gly-
increase in intrinsic dissolution rate as compared to amor- col) (PEG) and fenofibrate reduced the particle size and
phous and crystalline free acid, respectively. In vivo phar- improved the solubility of fenofibrate (Law et  al. 2003).
macokinetic study revealed significantly lower Tmax and Similarly, borneol/menthol eutectic mixture enhanced the
higher Cmax for amorphous salt form as compared to other solubility of daidzein as compared to its suspension form
forms with no significant difference in AUC (Nielsen et al. (Shen et al. 2011). Shen et al. investigated the effect of dif-
2013). ferent ratios of borneol/menthol on intestinal absorption of

13
Analysis and optimization of drug solubility to improve pharmacokinetics

daidzein which was significantly increased with the use of drug is complimentarily paired with a counter ion (Kumar
25:75 ratio of eutectic mixture. Pharmacokinetic study of et  al. 2012). Salt formation assists the drug to be solubi-
daidzein with the eutectic mixture revealed an increase in lized more in its ionic state as compared to the nonionic
Cmax and bioavailability by 2.5- and 1.5-fold respectively, state. Among new drugs introduced by FDA, most are
with reduction in Tmax as compared to daidzein only (Shen developed in their salt forms; with hydrochlorides being
et al. 2011). commonly employed (Serajuddin 2007).
In a study by Tao et  al., the solubility of itraconazole
Chemical modifications was amplified by sixfold in its hydrochloride salt form as
compared to itraconazole only. Additionally, dissolution of
Chemical modification of active pharmaceutical ingredi- the salt form was significantly increased with higher stabil-
ent can be implemented for enhancement of solubility and ity of the salt form (Tao et al. 2009). Milewski et al. synthe-
pharmacokinetics of various poorly water soluble drugs. sized different salt forms of naltrexone (acetate, glycolate,
Different methods utilized for such purpose include deri- lactate, fumarate, and citrate) and compared their efficacy
vatization and salt formation. in transdermal delivery with hydrochloride form (Milewski
et  al. 2012). The naltrexone glycolate form enhanced the
Derivatization solubility and provided 50% enhancement in drug flux
suggesting its great potential for drug delivery. In another
Different derivatives of active drug can be synthesized study, ditolenesulfonate salt of itraconazole was synthe-
for the purpose of drug solubility and pharmacokinet- sized and results revealed multifold increase in solubility of
ics enhancement. Functional group modifications can be salt in water and pharmaceutical solvents as compared to
employed for the synthesis of new derivatives with bet- itraconazole (Kumar et  al. 2012). Furthermore, a 5.5-fold
ter solubility and bioavailability. One of the derivatization increase in dissolution for salt form was evident with potent
methods is the synthesis of prodrugs which are inactive in antifungal effects (Kumar et  al. 2012). Similarly, Thakral
their native form but later change to active form by loss of and Suryanarayanan prepared tris salt of indomethacin
certain functional moieties, leading to solubility and bioa- that resulted in short reconstitution time of the elegant lyo-
vailability enhancement. Other methods include use of den- phile with better solubility and dissolution characteristics
drimer–drug complex [e.g. PAMAM-doxorubicin (Ke et al. (Thakral et al. 2015).
2008) and poly(propylene)imine-famotidine/indomethacin/
amphotericin B (Gupta et  al. 2007)] and drug–polymer Carrier technologies
conjugates [e.g. pectin-cisplatin (Verma et  al. 2008) and
N-(2-hydroxypropyl)-methacrylamide (HPMA)-paclitaxel Poor aqueous solubility limits the bioavailability and hin-
(Erez et al. 2009)]. ders the pharmaceutical development process. A plethora
Several studies utilizing derivatization have been per- of approaches involving different carrier systems have been
formed. In a study by Anwar et  al., authors synthesized extensively investigated for improvement of the aqueous
chitosan–atorvastatin conjugate for oral administration solubility and bioavailability of BCS class II and IV drugs.
(Anwar et  al. 2011). The conjugate exhibited solubility This section includes discussion regarding different carrier
enhancement by nearly 100-fold as compared to free atorv- technologies including self-emulsification, solid dispersion,
astatin. Additionally, the bioavailability of atorvastatin was inclusion complex, polymeric micelles, solid lipid nano-
increased by fivefold with conjugate as compared to its sus- particles (SLNs) and nanostructured lipid carriers (NLCs).
pension form (Anwar et al. 2011). In another study, amino Schematic representation of these systems is presented in
acid conjugate of naproxen was synthesized to evaluate the Fig. 3.
effect on solubility and dissolution capability (Mishra et al.
2012). The naproxen–glycine conjugates enhanced solubil- Self-emulsifying drug delivery systems (SEDDS)
ity and intrinsic dissolution of naproxen by 1.24-fold and
30.9%, respectively as compared to naproxen only with SEDDS are lipid based formulations that possess potential
enhanced therapeutic effects (Mishra et al. 2012). for delivery of drugs with poor aqueous solubility and low
oral bioavailability (Kang et al. 2004). These formulations
Salt formation are a blend of oils, surfactants and co-surfactants that in
suitable proportions lead to rapid formation of oil in water
Salt formation is one of the common approaches for solu- emulsion upon exposure to the aqueous media present in
bility and dissolution enhancement of poorly water soluble the gastrointestinal tract (Atef et al. 2008). Emulsion forma-
drugs (Tao et al. 2009). In this method, the physicochemi- tion is generally assisted by the gastric motility that leads
cal or biopharmaceutical property of ionizable moiety of to production of physically and thermodynamically stable

13
R. K. Thapa et al.

Fig. 3 Schematic representation of different nanoparticulate systems for the enhancement of drug solubility. SMEDDS self-microemulsifying
drug delivery system, SNEDDS self-nanoemulsifying drug delivery system

emulsions with <50 nm droplet size (Maulik et al. 2010). the improved therapeutic effects. Different studies utilizing
Rapid emulsion formation retains the drug in dissolved SMEDDS and SNEDDS are presented in Table 1.
form and small droplet size offers a considerably large Kim et  al. prepared flurbiprofen SMEDDS and com-
interfacial surface area for acceleration of absorption rate pared the pharmacokinetics with the commercial product
of lipophilic drugs (Gershanik and Benita 2000; Kommuru (Kim et  al. 2012). The in  vitro dissolution of flurbiprofen
et al. 2001; Jing-ling et al. 2007). Moreover, lipids enhance in SMEDDS was enhanced by 1.5-fold as compared to the
intestinal lymphatic uptake of drugs that aids in avoiding commercial product. More importantly, the total plasma
the presystemic biotransformation of drug molecules (Khan concentration of the drug in SMEDDS was significantly
et  al. 2012). Moreover, the increase in membrane fluidity higher than in the commercial product. A 3.6-fold increase
due to presence of surfactants and co-surfactant expedite in the AUC was evident with SMEDDS formulation (Kim
transcellular absorption thus enhancing the drug bioavail- et  al. 2012). Similarly, fenofibrate SMEDDS were pre-
ability (Cornaire et al. 2004; Kale and Vandana 2008). Two pared (Lee et al. 2014). Compared to the powder form, the
of the most commonly used SEDDS include: self-micro- SMEDDS formulation presented high drug dissolution pro-
emulsifying drug delivery system (SMEDDS) and self- file (~87% in 30 min). Significantly higher values for AUC
nanoemulsifying drug delivery system (SNEDDS). The and Cmax were observed with nearly 3.6-fold increase in
main difference between them is the size of the emulsion relative bioavailability for the SMEDDS formulation (Lee
formed, which is in micro and nano size respectively. These et  al. 2014). Docetaxel SNEDDS were developed by Seo
systems even facilitate the intestinal lymphatic uptake of and colleagues (Seo et al. 2013). In vivo pharmacokinetic
the drugs mediated by the lipoidal part that can avoid the studies revealed enhanced plasma circulation time for doc-
presystemic biotransformation of drug molecules (Humber- etaxel by SNEDDS. A respective 6- and 12-fold increase in
stone and Charman 1997; O’Driscoll 2002). Subsequently, AUC and Cmax, and 6.5-fold higher absolute bioavailabil-
the bioavailability of the drug can be enhanced resulting in ity were observed for SMEDDS when compared to the oral

13
Table 1 SMEDDS/SNEDDS formulations of different drugs for enhancement of solubility and pharmacokinetics
Carrier technology Drug Effect on solubility and dissolution Effect on pharmacokinetics References

SMEDDS Astilbin Enhanced in vitro drug release profiles Significant enhancement of astilbin bioavailability by Mezghrani et al. (2011)
SMEDDS formulation
SMEDDS Probucol 80% probucol dissolved in medium in 10 min by Significant reduction in Tmax and increase in Cmax were Sha et al. (2012)
SMEDDS as compared to negligible release by crude evident; bioavailability of SMEDDS was 2.1- and 10.2-
probucol even after 60 min fold higher as compared to oil solution and aqueous
suspension of probucol
SMEDDS Berberine hydrochloride Attainment of 100% drug dissolution within 5 h Compared to commercial tablet, Cmax and AUC of Zhu et al. (2013)
SMEDDS was 163.4 and 154.2% higher, respectively
with 2.4-fold increase in relative bioavailability
SMEDDS Fenofibrate Significantly higher dissolution rate as compared to drug Compared to powder form, SMEDDS enhanced Cmax Kim et al. (2013a, b)
powder and bioavailability of fenofibrate by 4.0- and 2.0-fold
respectively
SMEDDS Domperidone Improved in vitro dissolution of domperidone Enhancement of oral bioavailability of domperidone by Jakki et al. (2013)
1.9-fold and Cmax by 2.0-fold as compared to suspension
form
Analysis and optimization of drug solubility to improve pharmacokinetics

SMEDDS Naproxen Significantly enhanced solubility and dissolution rate of – Čerpnjak et al. (2014)
naproxen in SMEDDS formulation compared to pure
naproxen
SNEDDS Lutein Enhanced solubility and dissolution of lutein (~90%) as Cmax and relative bioavailability of lutein in SNEDDS was Shanmugam et al. (2011)
compared to powder form significantly increased with 24.9-/9.1-fold and 12.3-/2.8-
fold as compared to lutein powder/commercial product
respectively
SNEDDS Flurbiprofen Significantly higher dissolution of flurbiprofen as com- Several folds increase in Cmax and AUC of flurbiprofen as Kang et al. (2012)
pared to powder form compared to powder drug
SNEDDS Docetaxel Higher solubility in different dissolution media Enhanced Cmax and AUC with respective 12.0- and 6.5- Seo et al. (2013)
fold increase as compared to drug solution
SNEDDS Carvedilol Enhanced solubility and complete dissolution in nearly Significant enhancement in Cmax and AUC by 134.2 and Singh et al. (2013)
20 min 85.2% respectively as compared to pure drug
SNEDDS Atazanavir Complete drug dissolution attained within 30 min as Cmax and AUC values rose significantly with respective Singh et al. (2014)
compared to pure drug (~30%) 2.1- and 2.5-fold increase as compared to pure drug
SNEDDS Atorvastatin Complete drug dissolution attained within 1 h as com- Augmentation in atorvastatin bioavailability by 193.8 Hashem et al. (2015)
pared to suspension (~50%) and 155.3% as compared to suspension and commercial
tablet formulation

13
13
Table 2 Solid dispersion formulations of different drugs for enhancement of solubility and pharmacokinetics
Drug Preparation method Effect on solubility and dissolution Effect on pharmacokinetics References

Valsartan Spray drying Improvement in drug solubility by 43-fold as compared to pure Higher AUC, Cmax and shorter Tmax with improved bioavailability Yan et al. (2012)
drug of drug by 2.2- and 1.7-fold as compared to powder and com-
mercial product respectively
Carvedilol Spray drying Enhancement of drug solubility and dissolution rate by about – Lee et al. (2013)
11,500- and twofold respectively
Raloxifene Spray drying Augmentation of solubility and dissolution by 30- and twofold Enhanced AUC and Cmax of raloxifene by 3.3- and 2.3-fold Tran et al. (2013)
respectively respectively
Telmisartan Spray drying Improvement of aqueous solubility and dissolution rate by Tmax was significantly reduced and Cmax was significantly Marasini et al. (2013)
40,000- and threefold respectively increased with increment of AUC by 13.4- and 2.1-fold as
compared to telmisartan powder and commercial product
respectively
Tacrolimus Spray drying Compared to drug powder, solubility and dissolution were Increase in Cmax and AUC by 10.0- and 2.0-fold respectively with Cho et al. (2014)
enhanced by 700- and 30-fold respectively reduction in Tmax
Silymarin Spray drying Significant increase in solubility by 650-fold as compared to drug 2.5- and 3.0-fold increase in Cmax and AUC respectively as com- Hwang et al. (2014)
powder pared to commercial product
Ezetimibe Spray drying Increase in solubility and dissolution by 25- and fivefold respec- Significant increase in Cmax and AUC; and decrease in Tmax Rashid et al. (2015)
tively
Sorafenib Spray drying Boosted solubility of sorafenib with nearly 10-fold increase com- Rise in Cmax and AUC by 1.5- and 1.8-fold compared to sorafenib Truong et al. (2015)
pared to free drug powder
Clopidogrel Spray drying 4.6-fold increase in solubility with improved dissolution profile Enhancement of AUC and Cmax by 4.0- and 3.2-fold respectively Kim et al. (2015)
napadisi-
late
Rebamipide Spray drying Improvement in solubility by nearly 200-fold as compared to free Increase in Cmax and AUC by nearly 2.0-fold as compared to both Pradhan et al. (2015)
drug rebamipide powder and commercial product
R. K. Thapa et al.
Analysis and optimization of drug solubility to improve pharmacokinetics

Table 3 Inclusion complexes of different drugs for enhancement of solubility and pharmacokinetics
Host molecule Guest molecule Effect on solubility and dis- Effect on pharmacokinetics References
solution

β-cyclodextrin Noscapine 10.35-fold increase in aque- Cmax and AUC were Madan et al. (2010)
ous solubility of noscapine increased by 4.5- and
as compared to free drug 2.0-fold respectively with
reduction in Tmax
Hydroxypropyl-β- Candesartan Cilexetil Escalation of candesartan – Shaikh and Avachat (2011)
cyclodextrin cilexetil’s solubility by
12-fold as compared to
pure drug
Cycloamylose Flurbiprofen Improvement in solubility Enhancement of Cmax and Baek et al. (2011)
and dissolution by 12- and AUC by 10.0- and 6.5-fold
twofold respectively respectively with reduced
Tmax as compared to free
drug
β-cyclodextrin Methotrexate 10-fold rise in aqueous solu- Enhancement of bioavailabil- Bourkaib et al. (2013)
bility of methotrexate ity by 1.5-fold
Hydroxypropyl-β- Cefdinir Enhanced solubility of – Guo et al. (2013)
cyclodextrin cefdinir by 2.36-fold as
compared to pure drug
Hydroxypropyl-β- Acyclovir Superior solubility and Increased rate and extent Nair et al. (2014)
cyclodextrin dissolution efficiency of of drug absorption with
acyclovir ~160.0% relative bioavail-
ability
Skimmed milk Piroxicam 2.5-fold increase in drug – Sanka et al.( 2014)
solubility as compared to
native form
Hydroxypropyl-β- Pseudolaric acid B 600-fold increase in solubil- – Chi et al. (2015)
cyclodextrin ity as compared to free
drug

docetaxel solution (Seo et al. 2013). These studies provide enhancing the surface area for dissolution of poorly water
evidence for the successful utilization of the SMEDDS and soluble drugs (Chiou and Riegelman 1969). Solid dis-
SNEDDS formulations for solubility and pharmacokinetic persion is found to have particles with high porosity that
enhancement of drugs. hastens the drug release profile thus enhancing the solu-
bility (Vasconcelos and Costa 2007). Similarly, availabil-
ity of drugs in amorphous state within the solid disper-
Solid dispersion sion aids in maintenance of higher drug solubility since
no energy is required to break crystal lattice during dis-
Solid dispersions are dispersions of one or more solid solution (Pokharkar et  al. 2006). Lastly, the enhance-
compounds in an inert matrix that are preferably appli- ment of drug’s wettability by the carrier system helps
cable for structurally diverse, hydrophobic drugs with in improvement of drug’s solubilization (Karvas et  al.
a wide range of physicochemical properties. Gener- 2006). Melting, solvent evaporation or solvent wetting
ally, a solid dispersion contains molecular dispersion of are the conventional methods used for preparation of
drug into hydrophilic carrier for the purpose of improv- solid dispersion that pose several problems including low
ing aqueous solubility and dissolution rate of lipophilic yield, drug’s chemical decomposition and stability prob-
drugs, especially from BCS class II. The mechanism of lems (Tran et  al. 2013). Therefore, new techniques uti-
solubility and dissolution enhancement include parti- lizing high energy processes such as freeze drying, spray
cle size reduction, enhanced porosity, drug’s amorphous drying, evaporative precipitation and melt extrusion are
state and improved wettability of drug. Upon expo- used for better efficacy and safety issues. Different for-
sure of solid dispersion to the aqueous media, the car- mulations of solid dispersions for the drug solubility and
rier dissolves and releases drug as fine colloidal crystals bioavailability enhancement are listed in Table 2.

13
R. K. Thapa et al.

Inclusion complex

Ramasamy et al. (2014)

Puntawee et al. (2015)


Kim et al. (2013a, b)

Zhang et al. (2014)


Mu et al. (2010) Inclusion complex, as the name suggests, uses the cavity

Li et al. (2013)
of a molecule in order to incorporate drugs in it. It is gen-
References

erally employed to improve the solubility, dissolution and


bioavailability of poorly water soluble drugs. The nonpolar
molecule or region of molecule (known as guest) is inserted
into the cavity of another molecule (known as host). The

Escalation of Cmax by 5.0- and 2.0-fold; and


and doxorubicin by 2.0- and 4.0-fold, and
Increase in Cmax and AUC of mitoxantrone

to stiripentol suspension and commercial


3.6-fold increase in docetaxel bioavailabil-

Improvement of Cmax and AUC by nearly

most commonly used host molecules are cyclodextrins.


ity as compared to commercial product

Approximately 400-fold increase in solubil- Enhancement of Cmax and AUC by 20.0-

AUC by 5.0- and 1.5-fold as compared


They are a family of cyclic oligosaccharides namely α, β, γ,
3.0-fold as compared to free drug

2.0- and 8.0-fold respectively δ and ε cyclodextrins incorporating six, seven, eight, nine
and ten (or more) (α-1,4)-linked α-D-glucopyranose units
and 13.5-fold respectively
Effect on pharmacokinetics

having hydrophobic cavity and hydrophilic outer surface.


Cyclodextrins are widely applied in pharmaceutical designs
for major role as solubilizer of poorly water soluble drugs.
The main mechanism of solubilization is their potential
Taxotere®

to form inclusion complex besides other mechanisms like


product

non-inclusion complexation and supersaturation. Differ-


ent methods used for the preparation of inclusion complex

include spray drying, freeze drying, kneading, co-precipi-


tation and extrusion, which exhibit differences in particle
Increase in aqueous solubility by 280-fold
Enhanced solubility of paclitaxel in simu-

Mitoxantrone and doxorubicin Superior pH sensitive solubility profile of

size, complexation rate and degree of amorphous nature of


both mitoxantrone and doxorubicin

end product (Singh et al. 2011). Hence, choice of prepara-


Effect on solubility and dissolution

lated gastric and intestinal fluids

Improved solubility of stiripentol


Table 4 Polymeric micelles incorporating different drugs for enhancement of solubility and pharmacokinetics

tion method is crucial to obtain the desired product. Dif-


ferent studies utilizing inclusion complex for drug solubil-
as compared to free drug
ity of sirolimus in water
Improved drug solubility

ity and pharmacokinetic enhancement are summarized in


Table 3.

Polymeric micelles

Micelles are self-assembled structures made of amphiphi-


lic molecules that contain inner hydrophobic core and outer
hydrophilic shell. Hydrophobic core is generally utilized
as a reservoir for drugs with poor aqueous solubility, and
Andrographolide analogue

hydrophilic shell protects unstable drugs from possible


chemical degradation (Xu et al. 2013). Polymeric micelles
are composed of large molecule surfactants which exhibit
excellent physiological and storage stability, and bio-
Stiripentol
Docetaxel

Paclitaxel

Sirolimus

compatibility. Techniques for preparation of drug-loaded


Drug

polymeric micelles include: chemical copolymerization


(wherein drug is chemically linked with a reactive group
of hydrophobic core) and physical entrapment technique (in
Methoxy poly(ethylene glycol)-polylactide

Poly(ethylene glycol)-b-poly(D,L-Lactide)
D-α-tocopheryl polyethylene glycol suc-
Pluronic F127-polyethyleneimine-folate

which a decrease in the level of a solvent leads to entrap-


Monomethoxy poly(ethylene glycol)-b-
Polyethylene oxide-b-polyacrylic acid

ment of drugs in polymer by solvent diffusion technique)


(Zhang et  al. 2014). These micelles are safe, inexpensive
and stable drug carriers that can incorporate several poorly
and pluronic copolymers

soluble drugs. Targetability of polymeric micelles to spe-


poly(ε-caprolactone)

cific organ can be achieved by using thermo- or pH-sensi-


tive block copolymers or by using a vector molecule such
as peptide, antibody, saccharide or some low-molecular
weight compounds attached to the outer surface of micelles.
Polymer

cinate

Due to their nanoscopic size, ability to solubilize large


amounts of hydrophobic drugs and achieve site-specific

13
Analysis and optimization of drug solubility to improve pharmacokinetics

Table 5 SLN/NLC formulations of different drugs for enhancement of solubility and pharmacokinetics

Carrier Drug Effect on pharmacokinetics References


technol-
ogy

SLN Etoposide Increase in AUC, t1/2 and mean residence time of etoposide as compared to free drug Athawale et al. (2014)
SLN Raloxifene Augmentation in Cmax and AUC by 308.0 and 270.0% respectively Tran et al. (2014a)
SLN Vorinostat Enhancement of Cmax and AUC by nearly 2.0-fold Tran et al. (2014b)
NLC Fenofibrate 4.0-fold escalation in plasma drug concentration and AUC as compared to free drug suspension Tran et al. (2014c)
NLC Oridonin Improvement of relative oral bioavailability by 143.4% Zhou et al. (2015)
NLC Imatinib Nearly 2.0-fold increase in Cmax and AUC of imatinib as compared to free drug Gupta et al. (2015)

delivery, polymeric micelles can potentially obtain desir- and pharmacokinetics of poorly water soluble drugs that
able biopharmaceutical and pharmacokinetic properties are elucidated in Table 5.
of drugs (Ale et  al. 2012). Several studies of polymeric
micelles as promising delivery carrier for enhancement of
solubility and pharmacokinetics are mentioned in Table 4.
Conclusion

Solid lipid nanoparticles and nanostructured lipid carriers In this review, we discussed different techniques that have
been adapted for the drug solubility measurement, optimi-
Solid lipid nanoparticles are lipid based colloidal drug zation (in most cases: improvement) and enhancement of
delivery carriers, with particle size of 50–1000  nm, that pharmacokinetics. Although shake flask method is the most
possess advantages of both polymeric nanoparticles and reliable one, new automated methods for high throughput
micronized emulsions. They usually comprise single lipid solubility measurements are still in demand. Among physi-
which remains solid at ambient temperature. SLNs were cal, chemical and carrier modification methods; nanopar-
generated with the purpose of incorporating diverse array ticle technologies offer promising results for drug solubil-
of chemicals, minimizing drug degradation during ADME ity and pharmacokinetics enhancement. Depending on the
process, and controlling drug release (Kathe et  al. 2014). drug characteristics and delivery requirement, appropriate
They are generally used to improve the bioavailability of method can be chosen for efficient solubility and pharma-
lipophilic drugs that are not amenable to other delivery car- cokinetics enhancement of lipophilic drugs, especially of
riers. Different approaches for preparation of SLNs include BCS class II and class IV.
high pressure homogenization, solvent emulsification
Compliance with ethical standards
evaporation and double emulsion techniques (Kathe et  al.
2014). There are several mechanisms by which SLNs can
Conflict of interest All authors (R.K. Thapa, H.G. Choi, J.O. Kim,
enhance drug solubility. Firstly, SLN prevents chemical and C.S. Yong) declare that they have no conflict of interest.
enzymatic degradation of drugs with the aid of lipids thus
maintaining the enhanced drug concentration. Surfactants
used also contribute to increase the permeability of the bio-
logical membrane or improve the affinity of lipid particles References
with biological membrane for drug absorption. Addition-
ally, bioadhesive natures of SLNs assist entry into biologi- Alavijeh MS, Chishty M, Qaiser MZ et  al (2005) Drug metabolism
and pharmacokinetics, the blood–brain barrier, and the central
cal membranes and enhance the drug retention as well as nervous system drug discovery. NeuroRx® 2:554–571
bioavailability (Hu et al. 2004). Ale EC, Maggio B, Fanani ML (2012) Ordered-disordered domain
Nanostructured lipid carriers are the types of SLNs that coexistence in ternary lipid monolayers activates sphingomyeli-
consist of liquid lipid nano-compartment made up of a mix- nase by clearing ceramide from the active phase. Biochim Bio-
phys Acta 1818:2767–2776
ture of structurally different lipids. These were introduced Ali HS, York P, Blagden N (2009) Preparation of hydrocortisone
to overcome the limitations of SLNs. Imperfections and lat- nanosuspension through a bottom-up nanoprecipitation tech-
tice spacings caused by these lipids result in improvement nique using microfluidic reactors. Int J Pharm 375:107–113
of drug entrapment and loading efficiencies. The prepara- Alonzo DE, Zhang GG, Zhou D et  al (2010) Understanding the
behavior of amorphous pharmaceutical systems during dissolu-
tion method involves mixing of solid with liquid lipids tion. Pharm Res 27(4):608–618
(Gaba et  al. 2015). Several studies have been performed Amidon GL, Lennernas H, Shah VP et al (1995) A theoretical basis
utilizing SLNs and NLCs for improvement of solubility for a biopharmaceutics drug classification: the correlation of

13
R. K. Thapa et al.

in  vitro drug product dissolution and in  vivo bioavailability. Erez R, Segal E, Miller K et al (2009) Enhanced cytotoxicity of a pol-
Pharm Res 12:413–420 ymer-drug conjugate with triple payload of paclitaxel. Bioorg
Anwar M, Warsi MH, Mallick N et  al (2011) Enhanced bioavaila- Med Chem 17:4327–4335
bility of nano-sized chitosan-atorvastatin conjugate after oral Gaba B, Fazil M, Ali A et al (2015) Nanostructured lipid (NLCs) car-
administration to rats. Eur J Pharm Sci 44:241–249 riers as a bioavailability enhancement tool for oral administra-
Atef E, Belmonte AA (2008) Formulation and in vitro and in vivo tion. Drug Deliv 22:691–700
characterization of a phenytoin self-emulsifying drug delivery Gershanik T, Benita S (2000) Self-dispersing lipid formulations for
system (SEDDS). Eur J Pharm Sci 35:257–263 improving oral absorption of lipophilic drugs. Eur J Pharm
Athawale RB, Jain DS, Singh KK et  al (2014) Etoposide loaded Biopharm 50:179–188
solid lipid nanoparticles for curtailing B16F10 melanoma col- Glomme A, März J, Dressman JB (2005) Comparison of a miniatur-
onization in lung. Biomed Pharmacother 68:231–240 ized shake-flask solubility method with automated potentiomet-
Avdeef A, Testa B (2002) Physicochemical profiling in drug ric acid/base titrations and calculated solubilities. J Pharm Sci
research: a brief survey of the state-of-the-art of experimental 94:1–16
techniques. Cell Mol Life Sci 59:1681–1689 Guo B, Zhong S, Li N et al (2013) Dissolution enhancement of cef-
Baek HH, Kwon SY, Rho SJ et  al (2011) Enhanced solubility and dinir with hydroxypropyl-β-cyclodextrin. Drug Dev Ind Pharm
bioavailability of flurbiprofen by cycloamylose. Arch Pharm 39:1638–1643
Res 34:391–397 Gupta U, Agashe HB, Jain NK (2007) Polypropylene imine dendrimer
Balani PN, Wong SY, Ng WK et  al (2010) Influence of polymer mediated solubility enhancement: effect of pH and functional
content on stabilizing milled amorphous salbutamol sulphate. groups of hydrophobes. J Pharm Pharm Sci 10:358–367
Int J Pharm 391:125–136 Gupta B, Poudel BK, Tran TH et  al (2015) Modulation of pharma-
BergstrÓ§m CA, Norinder U, Luthman K, Artursson P (2002) cokinetic and cytotoxicity profile of imatinib base by employing
Experimental and computational screening models for predic- nanostructured lipid carriers. Pharm Res 32:2912–2927
tion of aqueous drug solubility. Pharm Res19:182–188 Hancock BC, Zografi G (1997) Characteristics and significance of
Bourkaib N, Zhou J, Yao J et  al (2013) Combination of the amorphous state in pharmaceutical systems. J Pharm Sci
β-cyclodextrin inclusion complex and self-microemulsifying 86:1–12
drug delivery system for photostability and enhanced oral Hao HX, Hou BH, Wang JK et  al (2005) Solubility of erythritol in
bioavailability of methotrexate: novel technique. Drug Dev different solvents. J Chem Eng Data 50:1454–1456
Ind Pharm 39(6):918–927 Hashem FM, Al-Sawahli MM, Nasr M et al (2015) Custom fractional
Čerpnjak K, Zvonar A, Vrečer F et  al (2014) Development of a factorial designs to develop atorvastatin self-nanoemulsifying
solid self-microemulsifying drug delivery system (SMEDDS) and nanosuspension delivery systems—enhancement of oral
for solubility enhancement of naproxen. Drug Dev Ind Pharm bioavailability. Drug Des Dev Ther 9:3141–3152
13:1–10 Hu L, Tang X, Cui F (2004) Solid lipid nanoparticles (SLNs) to
Chen XQ, Venkatesh S (2004) Miniature device for aqueous and improve oral bioavailability of poorly soluble drugs. J Pharm
non-aqueous solubility measurements during drug discovery. Pharmacol 56:1527–1535
Pharm Res 21:1758–1761 Huang Y, Zhang B, Gao Y et  al (2014) Baicalein-nicotinamide
Chi L, Liu R, Guo T et  al (2015) Dramatic improvement of the cocrystal with enhanced solubility, dissolution, and oral bio-
solubility of pseudolaric acid B by cyclodextrin complexa- availability. J Pharm Sci 103:2330–2337
tion: preparation, characterization and validation. Int J Pharm Humberstone AJ, Charman WN (1997) Lipid-based vehicles for the
479:349–356 oral delivery of poorly water soluble drugs. Adv Drug Deliv
Chiou WL, Riegelman S (1969) Preparation and dissolution char- Rev 25:103–128
acteristics of several fast-release solid dispersion of griseoful- Huuskonen J, Salo M, Taskinen J (1998) Aqueous solubility predic-
vin. J Pharm Sci 58:1505–1510 tion of drugs based on molecular topology and neural network
Cho JH, Kim YI, Kim DW et  al (2014) Development of novel modeling. J Chem Inf Comput Sci 38:450–456
fast-dissolving tacrolimus solid dispersion-loaded prolonged Hwang DH, Kim YI, Cho KH et  al (2014) A novel solid dispersion
release tablet. Eur J Pharm Sci 54:1–7 system for natural product-loaded medicine: silymarin-loaded
Cornaire G, Woodley J, Hermann P et  al (2004) Impact of excipi- solid dispersion with enhanced oral bioavailability and hepato-
ents on the absorption of p-glycoprotein substrates in  vitro protective activity. J Microencapsul 31:619–626
and in vivo. Int J Pharm 278:119–131 IUPAC (1997) Compendium of chemical terminology. Blackwell,
Custodio JM, Wu CY, Benet LZ (2008) Predicting drug disposition, Oxford
absorption/elimination/transporter interplay and the role of Jakki R, Afzal Syed M, Kandadi P et  al (2013) Development of a
food on drug absorption. Adv Drug Deliv Rev 60:717–733 self-microemulsifying drug delivery system of domperidone:
Dahan A, Miller JM (2012) The solubility-permeability interplay in vitro and in vivo characterization. Acta Pharm 63:241–251
and its implications in formulation design and development Jing-ling T, Jin S, Zhong-Gui H (2007) Emulsifying drug delivery
for poorly soluble drugs. AAPS J 14:244–251 systems: strategy for improving oral delivery of poorly soluble
Detroja C, Chavhan S, Sawant K (2011) Enhanced antihypertensive drugs. Curr Drug Ther 2:85–93
activity of candesartan cilexitil nanosuspension: formula- Jouyban-Gharamaleki V, Jouyban-Gharamaleki K, Shayanfar A et al
tion, characterization and pharmacodynamic study. Sci Pharm (2015) An automated system for determining drug solubility
79:635–651 based on laser monitoring technique. J Lab Autom 20(1):3–9
DiPiro JT, Spruill WJ, Wade WE et  al (2010) Concept in clinical Junghanns JUA, Müller RH (2008) Nanocrystal technology, drug
pharmacokinetics. American Society of Health-System Phar- delivery and clinical applications. Int J Nanomed 3:295–310
macists, Bethesda Kale AA, Vandana BP (2008) Design and evaluation of self-emulsi-
Elsayed I, Abdelbary AA, Elshafeey AH (2014) Nanosizing of a fying drug delivery systems (SEDDS) of nimodipine. AAPS
poorly soluble drug: technique optimization, factorial analy- Pharm Sci Tech 9:191–196
sis, and pharmacokinetic study in healthy human volunteers. Kang BK, Lee JS, Chon SK et  al (2004) Development of self-
Int J Nanomed 9:2943–2953 microemulsifying drug delivery systems (SMEDDS) for oral

13
Analysis and optimization of drug solubility to improve pharmacokinetics

bioavailability enhancement of simvastatin in beagle dogs. Int LÓ§bmann K, Laitinen R, Grohganz H et  al (2011) Coamorphous
J Pharm 274:65–73 drug systems: enhanced physical stability and dissolution rate
Kang JH, Oh DH, Oh YK et al (2012) Effects of solid carriers on the of indomethacin and naproxen. Mol Pharm 8:1919–1928
crystalline properties, dissolution and bioavailability of flur- Madan J, Dhiman N, Parmar VK et al (2010) Inclusion complexes
biprofen in solid self-nanoemulsifying drug delivery system of noscapine in beta-cyclodextrin offer better solubility and
(solid SNEDDS). Eur J Pharm Biopharm 80:289–297 improved pharmacokinetics. Cancer Chemother Pharmacol
Karvas E, Ktistis G, Xenakis A et al (2006) Effect of hydrogen bond- 65:537–548
ing interaction on release mechanism of felodipine from nano- Marasini N, Tran TH, Poudel BK et al (2013) Fabrication and eval-
dispersion with polyvinylpyrrolidone. Eur J Pharm Biopharm uation of pH-modulated solid dispersion for telmisartan by
63:103–114 spray-drying technique. Int J Pharm 441:424–432
Kathe N, Henriksen B, Chauhan H (2014) Physicochemical character- Maulik JP, Sanjay SP, Natvarlal MP (2010) Self-microemulsifying
ization techniques for solid lipid nanoparticles: principles and drug delivery system. Int J Pharm Sci Rev Res 4(29):35
limitations. Drug Dev Ind Pharm 40:1565–1575 Mezghrani O, Ke X, Bourkaib N et al (2011) Optimized self-micro-
Ke W, Zhao Y, Huang R et al (2008) Enhanced oral bioavailability of emulsifying drug delivery systems (SMEDDS) for enhanced
doxorubicin in a dendrimer drug delivery system. J Pharm Sci oral bioavailability of astilbin. Pharmazie 66:754–760
97:2208–2216 Milewski M, Pinninti RR, Stinchcomb AL (2012) Naltrexone salt
Khan F, Islam MS, Roni MA et  al (2012) Systematic development selection for enhanced transdermal permeation through
of self-microemulsifying drug delivery systems of atorv- microneedle-treated skin. J Pharm Sci 101:2777–2786
astatin with improved bioavailability potential. Sci Pharm Mishra A, Agrawal S, Pathak K (2012) Naproxen glycine conju-
80(4):1027–1043 gate-synthesis, pharmaceutical preformulation and pharmaco-
Kim DW, Kang JH, Oh DH et al (2012) Development of novel flurbi- dynamic evaluation. Drug Deliv 19:102–111
profen-loaded solid self-microemulsifying drug delivery system Mu CF, Balakrishnan P, Cui FD et al (2010) The effects of mixed
using gelatin as solid carrier. J Microencapsul 29:323–330 MPEG-PLA/pluronic copolymer micelles on the bioavail-
Kim GG, Poudel BK, Marasini N et al (2013a) Enhancement of oral ability and multidrug resistance of docetaxel. Biomaterials
bioavailability of fenofibrate by solid self-microemulsifying 31:2371–2379
drug delivery systems. Drug Dev Ind Pharm 39:1431–1438 Murdande SB, Pikal MJ, Shanker RM et al (2010) Solubility advan-
Kim MS, Kim JS, Cho WK et  al (2013b) Enhanced solubility and tage of amorphous pharmaceuticals: I. A thermodynamic
oral absorption of sirolimus using D-α-tocopheryl polyethyl- analysis. J Pharm Sci 99:1254–1264
ene glycol succinate micelles. Artif Cells Nanomed Biotechnol Nair AB, Attimarad M, Al-Dhubiab BE et  al (2014) Enhanced
41:85–91 oral bioavailability of acyclovir by inclusion complex using
Kim YH, Kim DW, Kwon MS et al (2015) Clopidogrel napadisilate hydroxypropyl-β-cyclodextrin. Drug Deliv 21:540–547
monohydrate loaded surface-modifies solid dispersion: phys- Nielsen LH, Gordon S, Holm R et  al (2013) Preparation of an
icochemical characterization and in vivo evaluation. Biol Pharm amorphous sodium furosemide salt improves solubility and
Bull 38:1033–1040 dissolution rate and leads to a faster Tmax after oral dosing to
Kommuru T, Khan M, Reddy I (2001) Self emulsifying drug delivery rats. Eur J Pharm Biopharm 85:942–951
systems (SEDDS) of coenzyme Q10: formulation development O’Driscoll CM (2002) Lipid-based formulations for intestinal lym-
and bioavailability assessment. Int J Pharm 212:233–246 phatic delivery. Eur J Pharm Sci 15:405–415
Kufe DW, Pollock RE, Weichselbaum RR et  al (2003) Holland-Frei Patravale VB, Date AA, Kulkarni RM (2004) Nanosuspensions:
cancer medicine. BC Decker, Hamilton a promising drug delivery strategy. J Pharm Pharmacol
Kumar N, Bansal G, Kumar S et al (2012) Ditosylate salt of itracona- 56:827–840
zole and dissolution enhancement using cyclodextrins. AAPS Pokharkar VB, Mandpe LP, Padamwar MN et  al (2006) Develop-
Pharm Sci Tech 13:863–874 ment, characterization and solubilization of amorphous form
Lachman L, Lieberman H, Kanig JL (1986) The theory and practice of low Tg drug. Powder Technol 167:20–25
of industrial pharmacy. Lea and Febiger, Philadelphia Pradhan R, Tran TH, Choi JY et  al (2015) Development of a
Law D, Wang W, Schmitt EA et al (2003) Properties of rapidly dis- rebamipide solid dispersion system with improved dissolution
solving eutectic mixtures of poly(ethylene glycol) and fenofi- and oral bioavailability. Arch Pharm Res 38:522–533
brate: the eutectic microstructure. J Pharm Sci 92:505–515 Puntawee S, Theerasilp M, Reabroi S et  al (2015) Solubility
Lee SN, Poudel BK, Tran TH et  al (2013) A novel surface-attached enhancement and in vitro evaluation of PEG-b-PLA micelles
carvedilol solid dispersion with enhanced solubility and disso- as nanocarrier of semi-synthetic andrographolide analogue
lution. Arch Pharm Res 36:79–85 for cholangiocarcinoma chemotherapy. Pharm Dev Technol
Lee DW, Marasini N, Poudel BK et  al (2014) Application of Box- 21:437–444
Behnken design in the preparation and optimization of fenof- Ramasamy T, Kim JH, Choi JY et al (2014) pH sensitive polyelectro-
ibrate-loaded self-microemulsifying drug delivery system lyte complex micelles for highly effective combination chemo-
(SMEDDS). J Microencapsul 31:31–40 therapy. J Mater Chem B 2:6324–6333
Leleux J, Williams RO III (2014) Recent advancements in mechanical Rashid R, Kim DW, ud Din F et al (2015) Effect of hydroxypropyl-
reduction methods: particulate systems. Drug Dev Ind Pharm cellulose and Tween 80 on physicochemical properties and bio-
40:289–300 availability of ezetimibe-loaded solid dispersion. Carbohydr
Li X, Yin Q, Chen W et al (2006) Solubility of hydroquinone in dif- Polym 130:26–31
ferent solvents from 276.65 to 345.10 K. J Chem Eng Data Reviakine I, Johannsmann D, Richter RP (2011) Hearing what you
51:127–129 cannot see and visualizing what you hear: Interpreting quartz
Li Y, Bi Y, Xi Y et  al (2013) Enhancement on oral absorption of crystal microbalance data from solvated surfaces. Anal Chem
paclitaxel by multifunctional pluronic micelles. J Drug Target 83:8838–8848
21(2):188–199 Ruell J, Avdeef A (2003) A measured solution. Mod Drug Disc
Liu LS, Kim JM, Kim WS (2015) Simple and reliable quartz crystal 2003:47–49
microbalance technique for determination of solubility by cool- Sanka K, Munjulury VS, Mohd AB et al (2014) Enhancement of solu-
ing and heating solution. Anal Chem 7:3329–3335 bility, dissolution release profile and reduction in ulcerogenecity

13
R. K. Thapa et al.

of piroxicam by inclusion complex with skimmed milk. Drug Tran TH, Ramasamy T, Troung DH et  al (2014b) Development of
Deliv 21:560–570 vorinostat-loaded solid lipid nanoparticles to enhance pharma-
Seo YG, Kim DH, Ramasamy T et  al (2013) Development of doc- cokinetics and efficacy against multidrug-resistant cancer cells.
etaxel-loaded solid self-nanoemulsifying drug delivery system Pharm Res 31:1978–1988
(SNEDDS) for enhanced chemotherapeutic effect. Int J Pharm Tran TH, Ramasamy T, Cho HJ et al (2014c) Formulation and optimi-
452:412–420 zation of raloxifene-loaded solid lipid nanoparticles to enhance
Serajuddin AT (2007) Salt formation to improve drug solubility. Adv oral bioavailability. J Nanosci Nanotechnol 14:4820–4831
Drug Deliv Rev 59:603–616 Truong DH, Tran TH, Ramasamy T et  al (2015) Preparation and
Sha X, Wu J, Chen Y et al (2012) Self-microemulsifying drug-deliv- characterization of solid dispersion using a novel amphiphilic
ery system for improved oral bioavailability of probucol: prepa- copolymer to enhance dissolution and oral bioavailability of
ration and evaluation. Int J Nanomed 7:705–712 sorafenib. Powder Technol 283:260–265
Shaikh SM, Avachat AM (2011) Enhancement of solubility and per- U.S. Food and Drug Administration (2015) Waiver of in  vivo bio-
meability of candesartan cilexetil by using different pharmaceu- availability and bioequivalence studies for immediate release
tical interventions. Curr Drug Deliv 8(4):346–353 solid oral dosage forms based on a biopharmaceutics classifi-
Shanmugam S, Baskaran R, Balakrishnan P et  al (2011) Solid self- cation system. U.S. Food and Drug Administration, Guidance
nanoemulsifying drug delivery system (S-SNEDDS) contain- for Industry, Silver Spring. http://www.fda.gov/AboutFDA/
ing phosphatidylcholine for enhanced bioavailability of highly CentersOffices/OfficeofMedicalProductsandTobacco/CDER/
lipophilic bioactive carotenoid lutein. Eur J Pharm Biopharm ucm128219.html. Accessed 21 July 2015
79:250–257 Van Eerdenbrugh B, Vermant J, Martens JA et  al (2009) A screen-
Shen Q, Li X, Li W et  al (2011) Enhanced intestinal absorption of ing study of surface stabilization during the production of drug
daidzein by borneol/menthol eutectic mixture and microemul- nanocrystals. J Pharm Sci 98:2091–2103
sion. AAPS Pharm Sci Tech 12:1044–1049 Vasconcelos T, Costa P (2007) Development of a rapid dissolving
Shevchenko A, Bimbo LM, Miroshnyk I et al (2012) A new cocrystal ibuprofen solid dispersion. Pharm Sci 16: 676–681
and salts of itraconazole: comparison of solid-state properties, Verma AK, Sachin K (2008) Novel hydrophilic drug polymer nano-
stability and dissolution behavior. Int J Pharm 436:403–409 conjugates of cisplatin showing long blood retention profile: its
Singh G, Pai RS (2014) Optimized self-nanoemulsifying drug deliv- release kinetics, cellular uptake and bio-distribution. Curr Drug
ery system of atazanavir with enhanced oral bioavailabil- Deliv 5:120–126
ity: in  vitro/in vivo characterization. Expert Opin Drug Deliv Verma S, Gokhale R, Burgess DJ (2009) A comparative study of top-
11:1023–1032 down and bottom-up approaches for the preparation of micro/
Singh A, Worku ZA, Van den Mooter G (2011) Oral formulation nanosuspensions. Int J Pharm 380:216–222
strategies to improve solubility of poorly water-soluble drugs. Wang LC, Ding H, Zhao JH et  al (2008) Solubility of isonicotinic
Expert Opin Drug Deliv 8:1361–1378 acid in 4-methylpyridine + water from (287.65 to 361.15) K. J
Singh B, Singh R, Bandyopadhyay S et al (2013) Optimized nanoe- Chem Eng Data 53:2544–2546
mulsifying systems with enhanced bioavailability of carvedilol. Williams HD, Trevaskis NL, Charman SA et  al (2013) Strategies
Colloids Surf B 101:465–474 to address low drug solubility in discovery and development.
Stott PW, Williams AC, Barry BW (1998) Transdermal delivery from Pharmacol Rev 65:315–499
eutectic systems: enhanced permeation of a model drug, ibupro- Xu W, Ling P, Zhang T (2013) Polymeric micelles, a promising drug
fen. J Control Release 50:297–308 delivery system to enhance bioavailability of poorly water-solu-
Sun CC (2013) Cocrystallization for successful drug delivery. Expert ble drugs. J Drug Deliv 2013:315–340
Opin Drug Deliv 10:201–213 Yadav AV, Shete AS, Dabke AP et  al (2009) Co-crystals: a novel
Tao T, Zhao Y, Wu J et al (2009) Preparation and evaluation of itra- approach to modify physicochemical properties of active phar-
conazole dihydrochloride for the solubility and dissolution rate maceutical ingredients. Indian J Pharm Sci 71:359–370
enhancement. Int J Pharm 367:109–114 Yadav SK, Mishra S, Mishra B (2012) Eudragit-based nanosuspen-
Thadkala K, Nanam PK, Rambabu B et  al (2014) Preparation and sion of poorly water-soluble drug: formulation and in  vitro-in
characterization of amorphous ezetimibe nanosuspensions vivo evaluation. AAPS Pharm Sci Tech 13:1031–1044
intended for enhancement of oral bioavailability. Int J Pharm Yan YD, Sung JH, Kim KK et al (2012) Novel valsartan-loaded solid
Investig 4:131–137 dispersion with enhanced bioavailability and no crystalline
Thakral S, Suryanarayanan R (2015) Salt formation during freeze- changes. Int J Pharm 422:202–210
drying—an approach to enhance indomethacin dissolution. Zhang X, Wang H, Zhang T et  al (2014) Exploring the potential of
Pharm Res 32:3722–3731 self-assembled mixed micelles in enhancing the stability and
Tiwari AK (2001) Modification of crystal habit and its role in dosage oral bioavailability of an acid-labile drug. Eur J Pharm Sci
form performance. Drug Dev Ind Pharm 27:699–709 62:301–308
Tran TH, Poudel BK, Marasini N et al (2013) Preparation and evalu- Zhou X, Zhang X, Ye Y et  al (2015) Nanostructured lipid carriers
ation of raloxifene-loaded solid dispersion nanoparticle by used for oral delivery of oridonin: an effect of ligand modifica-
spray-drying technique without an organic solvent. Int J Pharm tion on absorption. Int J Pharm 479:391–398
443:50–57 Zhu JX, Tang D, Feng L et  al (2013) Development of self-micro-
Tran TH, Ramasamy T, Troung DH et  al (2014a) Preparation and emulsifying drug delivery system for oral bioavailability
characterization of fenofibrate-loaded nanostructured lipid car- enhancement of berberine hydrochloride. Drug Dev Ind Pharm
riers for oral bioavailability enhancement. AAPS Pharm Sci 39:499–506
Tech 15:1509–1515

13

S-ar putea să vă placă și