Sunteți pe pagina 1din 26

Drug Development and Industrial Pharmacy

ISSN: 0363-9045 (Print) 1520-5762 (Online) Journal homepage: https://www.tandfonline.com/loi/iddi20

A review of methods for solubility determination


in biopharmaceutical drug characterisation

Ardita Veseli, Simon Žakelj & Albin Kristl

To cite this article: Ardita Veseli, Simon Žakelj & Albin Kristl (2019): A review of methods for
solubility determination in biopharmaceutical drug characterisation, Drug Development and
Industrial Pharmacy, DOI: 10.1080/03639045.2019.1665062

To link to this article: https://doi.org/10.1080/03639045.2019.1665062

Accepted author version posted online: 06


Sep 2019.

Submit your article to this journal

Article views: 5

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iddi20
A review of methods for solubility determination in biopharmaceutical

drug characterisation

Ardita Veseli, Simon Žakelj* and Albin Kristl

Department of Biopharmaceutics and Pharmacokinetics, University of Ljubljana, Faculty of


Pharmacy, Ljubljana, Slovenia

Correspondence: Simon Žakelj; E-mail: simon.žakelj@ffa.uni-lj.si

t
ip
cr
us
an
M
ed
pt
ce
Ac
A review of methods for solubility determination in biopharmaceutical

drug characterisation

Abstract

The significance of thermodynamic solubility in biopharmaceutical compound or drug

characterization as well as the importance of having methods that accurately establish it

have been extensively addressed. Nonetheless, its precise determination continues to

t
ip
remain a challenging task to accomplish. Even more so when the number of compounds

cr
to evaluate is high and the available amount of each compound is low, both of which

us
are inevitable for the compound characterization during the drug development process.

Except for the shake-flask method which is still considered as the ʻgold standardʼ in
an
obtaining thermodynamic data, it is currently difficult to say that another satisfactory

model which is routinely used to determine thermodynamic solubility is being applied.


M

Therefore, this review summarizes the various experimental approaches which are
ed

based on the classical shake flask method but have yet attempted to speed up the

experimental process of obtaining such data more conveniently. The most important
pt

experimental features of these approaches are provided to the reader. Some advantages
ce

and disadvantages associated with each approach are also highlighted, consequently

offering a resource to those looking for the most appropriate of the approaches that
Ac

have already fared well at determining the biopharmaceutically relevant drug solubility.

Keywords: Biopharmaceutical drug solubility; Thermodynamic (equilibrium)

solubility; Shake flask method; High throughput; Miniaturized assays


Introduction
It is well established that solubility as a physicochemical and biopharmaceutical property

has acquired an essential role in drug discovery and development. This parameter in

concurrence with permeability must be known to categorize a drug according to the

Biopharmaceutical Drug Classification System (BCS), which was first introduced by Amidon

et al. [1] and serves as a predictive approach to correlate the in vitro drug dissolution with the

in vivo bioavailability. Among all biopharmaceutically unfavourable compound properties,

poor solubility is on the top of the list. Unfortunately, poor aqueous solubility has often led to

t
ip
failure in drug development and has been manifested by the increased presence of BCS class

II and class IV compounds [2-4]. Determining the accurate and biopharmaceutically relevant

cr
solubility of drug substances is a challenge which should not be underestimated. In fact, due

us
to the complicated solubilisation process and solid phase chemistry of drug candidates, the
an
determination of solubility can be a complex task [5].

Moreover, the introduction of various terms such as ʻintrinsic solubilityʼ, ʻapparent


M

solubilityʼ, and ʻkinetic solubilityʼ has subsequently stirred up some confusion in the

terminology of the field [6–10]. In this paper, our focus lies on thermodynamic solubility
ed

which even to this day is considered to be the more accurate representation of true solubility
pt

[6]. Thermodynamic solubility seeks to establish the extent to which the compound dissolves
ce

and it manifests the equilibrium between the solution and undissolved stable polymorph.

These values are not absolute numbers either, since the thermodynamic value depends on the
Ac

properties of the compound and various experimental factors which involve buffer

composition, time for attaining equilibrium, temperature, mixing conditions, polymorphism,

compound purity, and particle size and shape [2,7,8].

Even though, kinetic measurements can be useful for screening in the early discovery

stage, thermodynamic solubility is still preferred in predicting drug properties and


establishing structure property relationship (SPR) during lead optimization. It is also

generally recommended to avoid the incorporation of kinetic solubility data in ADME

prediction tools and employ high quality aqueous solubility measurements instead [2]. In this

review, our goal is to characterize and provide a comparison of available methods used to

experimentally determine thermodynamic solubility. First, we start with the classical shake-

flask method and move onto assays that have tackled the issues of throughput, sample

requirement and analytical sensitivity, all of which are key factors in facilitating such

experimental determinations. Further, the strengths and limitations of these methods are

t
ip
discussed since the type of method employed is also an essential aspect in solubility

cr
assessment.

Shake-Flask Method

us
The shake-flask method which was first introduced more than 50 years ago, is still
an
considered as the most reliable technique in establishing solubility and comprises of the

following steps: (1) sample preparation, (2) equilibration, (3) separation of phases, (4)
M

analysis of the saturated solution and residual solid and (5) data analysis and interpretation.
ed

An amount of excess solid which does not have to be accurately measured is added to the

aqueous buffers [9]. This is because in principle, solubility should not be impacted by the
pt

amount of solids added, as long as we are strictly working with only one species of the solid
ce

and with one species of the solute while the pH of the dissolution medium is constant. When

the solubility of the tested free acid or free base is reasonably low, the second condition is
Ac

easily met by the use of buffers with sufficient capacity. The first condition, however, is often

overlooked in pharmaceutical applications simply because of the overwhelming interest in

the direct measurements of the solubility of salts at biologically relevant pH conditions.

Very few drugs are neither acids nor bases. The salts of free acids or bases are

favoured to some extent due to better technological properties but especially because of their
superior dissolution and solubility. Inevitably, the dissolution of a highly soluble salt at a pH

which favours the un-ionised form will result in precipitation of the less soluble un-ionised

form. In this case, the excess of the solid salt dictates the rate of total dissolution, but the

equilibrium solubility of the salt is never achieved since the precipitation of the free base or

acid – a competing process with its own kinetics – serves as a continuous solute sink. The

competition between the dissolution and the precipitation can result in a steady state solute

concentration which is in fact neither the solubility of the salt nor that of the free base or acid,

but somewhere in between depending on the excess of the solid salt, the medium pH and

t
ip
other experimental factors. In these very common cases, the biological relevance of the

cr
experimental setting including the excess solid is helpful in the biopharmaceutical

interpretation of the acquired results. Below, there is a summary of a few studies in which

us
the authors have observed and attempted to explain some atypical effects of the presence of
an
solid excess in the saturated solution.

Wang et al. [10] presented such a case showing that the quantity of the excess solid
M

used affected the solubility of a dihydrochloride salt of a diprotic weak base. Essentially, the
ed

concentration of the counterion, chloride was altered in concomitance with the change in the

amount of excess solid. In a pH region 2-5 where the monohydrochloride salt dictates the
pt

solubility, the drug which was firstly added as a dihydrochloride salt was converted to its
ce

monohydrochloride salt and chloride ions were released. As a result of this release, the

solubility of the drug was supressed via the common ion effect. However, in this case, the
Ac

physicochemical property of the drug needs to be emphasized. The compound possesses two

pKas (3.1 and 7.7) and the disparity between the two pKas is what contributed to the

alterations in solubility in the pH region 2-5 based on the added amount of solid. Therefore,

the change in solubility should be attributed to the physico-chemical property of the drug

rather than the intrinsic effect of excess solid on solubility [11].


Contrarily, Kawakami et al. [12] investigated the effect of excess solid on the

apparent solubility of free acids and a base at various pH conditions. Out of all the studied

drugs, indomethacin was the one that showed higher dependency of solubility on the amount

of excess solid. The apparent solubility seemed to decrease with an increase in the solid

amount at pH 5 and 6 while it increased with an increase in the solid amount at pH 6.5 and 7.

The authors suggested that the impact of the solid amount on the solubility of indomethacin

seemed to be most likely affected by a competition between the crystallization and

dissolution rates. In fact, the measurement of indomethacin solubility is often associated with

t
ip
difficulties, most likely because several polymorphic solid forms with different solubility of

cr
this drug exist and therefore, its solubility may appear to change over time [13].

Nonetheless, there are other issues in this study that need to be addressed, such as the

us
lack of buffer concentration and buffer capacity values whilst the pH values were not
an
measured once the samples reached equilibrium. Murdande et al. [11] has suggested that the

decrease in indomethacin solubility at pH 5 could be derived from the suppressed ionization


M

of the drug. Whereas, the increase in solubility at pH 7 might ensue from self-association or
ed

partial formation of sodium salt of indomethacin or even the possibility of co-crystal

formation [11]. On the other hand, according to the results obtained in the study by Baka et
pt

al. [7], hydrochlorothiazide solubility did not depend on the amount of solid excess in the
ce

solution, a finding which was not in contradiction with the results of Higuchi et al. [14]. Then

again, the authors did recommend using a small amount of excess (5-10 mg/5ml) for the
Ac

purpose of avoiding difficulties in sampling [7].

It is generally agreed that equilibrium in shake-flask solubility determinations is

reached when the same apparent solubility of the analysed samples is given after different

equilibration time periods. Equilibrium can be frequently reached within 24 hours with good

agitation. However, it is possible to find reported stirring times from 48 h to 2 weeks [15-17].
This is the case especially when working with compounds that are poorly soluble. When the

effective surface area for dissolution is increased, the procedure can be accelerated by either

vortexing or sonicating samples prior to equilibrium evaluation. Throughout the equilibration

process, amorphous material could be added to the samples or sample temperature may be

manipulated [18]. This will help create a supersaturated solution which serves as an

alternative in solving the issue of slow dissolution rate [9]. Poor wettability and tendency to

float are additional challenges that tend to rise when establishing solubility of poorly soluble

compounds [19]. These can often be resolved by means of small glass microspheres which

t
ip
will de-aggregate the particles with agitation or sonication [9] - the two approaches that we

cr
also often utilize in our laboratories.

To obtain biopharmaceutically relevant drug solubility data, it is recommended to

us
implement a physiologically acceptable pH range. The U.S FDA (Food and Drug
an
Administration) guidelines [20] recommend a pH range of 1 – 7.5, while the EMA (European

Medicines Agency) [21] proposes to conduct solubility studies along the pH of 1.2 – 6.8 at a
M

temperature of 37 ± 1 °C. More importantly, the pH should be verified and in some cases
ed

readjusted at the beginning of the experiment and before the separation of phases. This is

particularly crucial in the case of ionizable compounds which exhibit a pH-dependent


pt

solubility [22, 23] and are prone to overwhelming the buffer capacity when their solubility is
ce

high enough. The change of the medium pH caused by the dissolved tested compound results

in underestimated solubility for free acids and bases or in overestimated solubility for their
Ac

respective salts. Apart from adjusting the pH with an appropriate acid or base solution it is

also worth considering the incorporation of a buffer with sufficient buffering capacity in a

repeated experiment.

During the separation of phases, filtration and centrifugation are the obvious choices.

As easy as filtration sounds, in the case of hydrophobic and poorly soluble compounds, filter
sorption may be a significant source of error. This is especially an issue when the sample

volumes are small. When filtration is inconvenient, centrifugation or ultracentrifugation are

chosen instead. The separation of the solid from the saturated solution should take place at

the equilibrium temperature, especially when equilibrium is reached rapidly. Filtration at

ambient temperature can be tolerated for poorly soluble compounds since in this instance,

equilibrium is reached slowly [9]. The saturated solutions are analysed by UV

spectrophotometric method or HPLC analysis. The use of HPLC is more frequent due to its

advantage of detecting impurities and instabilities. Lastly, several techniques allow the

t
ip
identification of any solid-state changes as well. Such techniques include powder X-ray

cr
diffraction (PXRD), differential scanning calorimetry (DSC), microscopy, Raman and

infrared spectroscopy, and solid-state NMR [24-27]. Examining and reporting crystal forms

us
of excess drug solids is of great importance in such studies. In the event that the excess solid
an
is not in the most stable crystalline form, then non-equilibrium solubility data could be

attained. Irrespective of its reliability, the shake flask method is time consuming as a single
M

solubility experiment can take several days or even weeks. This restricts the usefulness of the
ed

method, making it unsuitable for medium to high throughput screening. It requires a large

quantity of the drug which is normally not available at early discovery phase hence limiting
pt

its suitability to the late discovery/early development stage. Hence, several attempts were
ce

made to miniaturize the classical shake-flask method, minimize the necessary solvent volume

and by doing so, to decrease the consumption of the tested compound and to somewhat
Ac

increase the method throughput. The miniaturization inevitably requires compromises or

adaptations regarding the solid handling, sampling or separation of phases etc. as is described

in the examples below and summarized in the Table 2 but these methods still provide

solubility results comparable to those from the shake-flask method, can be used with various

solvents and with various tested compounds.


Miniaturized Shake-Flask Method

A miniaturized version of the shake-flask method was developed by Glomme et al.

[28] with the purpose of determining solubility accurately with a minimized amount of the

drug. It incorporates all steps of the equilibration method but on a smaller scale. An excess

amount of the drug was placed into the Whatman Uniprep filter chamber followed by the

addition of 2 mL of solvent volume. The chamber was closed with a plunger which consisted

of a filtration membrane and a pre-attached cap. The experiment was performed at 37 °C

t
ip
while the sample was agitated at 450 rpm for 24 h. The pH was checked after several hours

cr
and at the end of the experiment. Once the shaking process came to an end, the plunger was

put into the chamber which pushed the filtrate into the reservoir of the plunger. Finally, the

us
HPLC with UV detection was used to determine the concentration of the compound in the
an
filtrate.

The simplicity and inexpensiveness of the method are certainly considered advantageous.
M

Solubility for almost every substance, including the neutral ones can be established while
ed

enabling the use of a vast range of media [28]. However, the use of the Whatman Uniprep

device may sometimes be conducive to drug loss especially for the poorly soluble compounds
pt

due to adsorption on the device and especially the filter membrane [29]. In addition, surface
ce

tension and volume-to-filter surface ratio could be problematic when solubility is established

on a small-scale setting [2]. The necessary amount of substance for one determination is
Ac

dependent mainly on compound solubility. For a poorly soluble compound, <1 mg is

required. In this case, in silico calculations are recommended, but not mandatory to assess the

approximate amount needed [28,30-33]. The HPLC is considered the only high-priced

element of this assay but nowadays, it can be found in most labs.


Nevertheless, it is necessary to proceed with caution when implementing the low-volume

approach. If this method is applied in the analysis of salts of strong acids and bases, then the

combination of the low volume with low buffer capacity may contribute to an unstable pH

value. The miniaturized shake-flask method is not suitable for high throughput measurements

since it generates data for only approximately 20 compounds per week, but it has been stated

to be precise and reliable [28]. The lack of high-throughput seems to be compensated by the

quality of the produced data seeing as this method has been repeated several times [34-40].

Small Scale Shake-Flask Method (SSF Approach)

t
ip
Another modified version of the shake-flask method can be found in the study conducted

cr
by Bergström et al. [41] where the solvent volume was reduced to 1000, 500, 200, 100, and

even 50 µL. No statistically considerable discrepancies were observed in solubility values in

us
this volume range. Thus, it was established that volume reduction does not affect the
an
precision of the method. The study was conducted at room temperature (22.5 ± 1 °C), the

samples were added in Milli-Q water and shaken at a speed of 300 rpm. Since structurally
M

diverse compounds were chosen, samples were withdrawn at different time points. The
ed

samples of the drugs that were expected to dissolve rapidly were withdrawn after 24, 48 and

72 h whereas, for the slowly dissolving drugs the withdrawal took place after 24, 72 and 144
pt

h. To diminish loss of substance and sample volume, the filtration step was replaced by
ce

ultracentrifugation (for 15 min. at 23000 g) and the samples were analysed by high-

performance liquid chromatography (HPLC) system. For the purpose of validation, the
Ac

obtained results were compared to the previously published solubility values from the

traditional shake-flask method. Both sets of values were in good agreement, confirming the

reliability of the SSF method.

In order to expand the SSF approach, Bergström et al. [42] studied an additional 25 bases

over a wide pH range employing a 0.15 M phosphate buffer where the pH of the drug
suspensions was adjusted with either KOH or H3PO4 solutions. The same experimental

conditions as in the former study were employed, except that all samples were withdrawn

after 24 h. The decision was made based on the previous experiment which showed that for

most of the drugs, equilibrium is attained within this time frame [41]. However, in the study

by Avdeef et al. [43], it was tentatively indicated that 24 h might have not been enough for

the practically insoluble drugs to reach equilibrium, additionally suggesting it as a topic for

further research. In both cases some of the investigated drugs were used as their

corresponding HCl salts, so the effect of the HCl salt on solubility, which we have already

t
ip
discussed in this review from the perspective of excess solid used, could be examined. It was

cr
claimed that no effect was observed when the drugs were studied in Milli-Q water

nonetheless, when phosphate buffer was utilized as a solvent, a minor impact on the

us
solubility of the compounds was seen [41,42]. This serves as further indication that when
an
phosphate buffers are incorporated in solubility studies, caution is required due to their

interaction with weak base drugs [44,45].


M

Moreover, concerns about the pH control in the study by Bergström et al. [41] were
ed

expressed by Völgyi et al. [44] regarding two of the analysed compounds namely

promethazine and propafenone. Therefore, implementing a rigorous protocol is vital since


pt

even the slightest alterations may cause inconsistencies in solubility [46]. This technique was
ce

deemed to be suitable for the 96-well microtiter plate format which makes it possible to

establish solubility by using only microgram quantities of samples. Through this format,
Ac

solubility determinations can also be automated allowing high throughput measurements.

Miniature Devices

A miniaturized device was set up to measure the solubility of six compounds with

diverse chemical structures in a microscale setting in the study by Chen and Venkatesh [47].

A tube closed in a continuous loop was filled with the drug slurry (≤ 0.5 mg) in an aqueous
solution. Tubes with two different diameters, 0.8 mm and 1.6 mm were used and the slurry

was continuously pumped through the tube loop. The solid material was trapped by a filter

(0.45 µm pore) and once the solution samples were removed, they were injected into the

HPLC for quantitation. The method was validated by comparing the results to the ones

obtained by shake-flask assay. When a limited amount of drug is available, which is normally

the case in early drug discovery, it was stated that a quantity as low as 0.2 mg can be added to

the smaller tubing to determine solubility.

In the traditional shake-flask method, drug adsorption to the filter membrane might

t
ip
pose a significant obstacle. Hence, a benefit of this method is the minimization of adsorption

cr
due to the drug slurry being constantly filtered through the membrane [46,47]. However, due

to the continuous pumping of the drug slurry, filter blockage could be an issue [48].

us
Equilibrium was reached within 6 h of circulation for most compounds which is seen as an
an
advantage as it facilitates a shorter turnaround time. This can be quite beneficial in building

structure-solubility relationships. With this set-up only 5-10 compounds are tested per roller
M

pump however, the possibility of setting up the device for high throughput measurements
ed

exists according to the authors. Bearing in mind the challenging process of pumping highly

viscous excipients or semi-solid and solid excipients, this technique is mostly restricted to
pt

aqueous or low viscosity pharmaceutical vehicles [46,47]. Additionally, the use of syringe
ce

filters may impose difficulties in terms of solid sample recovery. With most of these filters it

is not possible to reclaim the solid, which in turn hinders the process of determining the
Ac

crystalline form of the solid phase that would influence drug solubility.

Equilibrium 96-well Microtiter Plate Methods

Several studies have reported to have measured thermodynamic solubility by

implementing the solvent evaporation method. Firstly, the compound is dissolved in an

organic solvent, which is later removed by evaporation. This step is followed by the addition
of the aqueous medium to the drug for the purpose of incubation. Once equilibrium is

attained, separation is achieved usually by centrifugation and ultimately solubility is

determined by HPLC [48]. The solvent evaporation methods were developed to tackle

practicality issues such as excessive weighing tasks or automation of the solid dispensing

process of small drug quantities. However, the pre-dissolution of the drugs in organic

solvents, particularly DMSO followed by evaporation has caused somewhat of a concern.

This is due to the limited ability of some of these protocols to distinguish the polymorphous

states of the reconstituted solid samples. Moreover, some of these methods seem to have

t
ip
found their use primarily in in-house solubility measurements as it has also been stated by the

cr
authors themselves [49-51].

The Thermodynamic Solubility Assay (THESA Method) through which it is possible

us
to establish solubility in the lead optimization phase in some of the key solvents has been
an
reported by Alsenz and Kansy [2]. An aqueous buffer solution is added to a glass tube

containing the sample which is afterwards ultrasonicated for 1 h, shaken for 2 h and allowed
M

to stand for another 20 h. No information is provided regarding the temperature control which
ed

would be crucial especially during the 1h long sonication which could cause overheating of

the sample. Once the pH of the saturated solution is determined, the solution is either filtered
pt

or centrifuged and finally, the concentration is determined by ultra-fast HPLC or LC-MS.


ce

The Partially Automated Solubility Screening (PASS Method) which was also

described by Alsenz et al. [52] was developed for selecting vehicles for preclinical studies
Ac

while parallelly allowing thermodynamic solubility measurements in aqueous, non-aqueous,

high and low viscosity solvents in early pre-formulation. Typically, a compound (100 mg) is

suspended or dissolved in heptane to achieve a final concentration of 25 mg/mL.

Subsequently, 20-160 μL of the drug solution or suspension is pipetted into 96-well plates

and the vehicle is removed by means of evaporation in a SpeedVac centrifuge [52]. Very
much like with the “solvent evaporation method”, this approach in relation to the issue of

solid handling also results in a risk of the formation of new physical forms. Concerns have

also been raised due to the dispersion of the crystalline compound in heptane. An accurate

amount of compound in solid suspension can be difficult to dispense, especially when dealing

with low volumes [48]. Therefore, to ensure that the original physical form was maintained

even after vehicle removal, differential scanning calorimetry (DSC) and thermogravimetric

analysis (TGA) were performed. After evaporation, 40-80 μL of solvent and stir bars were

added to each well which were capped, and the plates were stirred at 300 rpm for 24 h at

t
ip
room temperature. Centrifugation was executed twice. The first time (for 1 min. at 2000 g)

cr
and the second time (for 15 min. at 3200 g), to remove any remaining solid from the

supernatant. The samples were eventually quantified by UHPLC. Combined with a robotic

us
liquid-handling system, this technique has a throughput of 45 samples per hour and therefore,
an
>600 solubility measurements can be obtained per week [52].

Arguments as to why this method is useful were provided. Based on the generated
M

results, an example of how digoxin doses may be formulated was given. It is possible to
ed

assess the feasibility of a dosage form intended to be given to animals based on the solubility

in a given solvent, solvent toxicity and maximum dose volumes per route. Estimations of the
pt

in vitro and in vivo behavior of compounds and formulations can be made based on the
ce

solubility results. This technique may also serve as a tool for selecting the suitable excipients

to improve the dissolution rates or bioavailability of low solubility drugs. Nonetheless, the
Ac

inability to analyze changes in the residual solid is the shortcoming of this assay. As a result,

in later stages or when more compound is available, PASS assay is usually replaced by the

SORESOS (Solubility and Residual Solid Screening) assay. Also, when screening viscous,

semi-solid and solid excipients, the PASS method might not be convenient due to the
challenging dispensing process of these excipients into a 96-well plate while using a robotic

liquid-handling system [48].

Wyttenbach et al. [53] established SORESOS (Solubility and Residual Solid

Screening), a combined screening method for solubility measurement and residual solid

screening in aqueous and non-aqueous solvents. This combination provides a new outlook for

the formulators in choosing vehicles for preclinical studies. The assay required 100 µL of

solvent, 10-15 mg of compound and single-use stirring bars per well in a 96-well microtiter

plate. The plate was sealed, and the drug-vehicle slurries were mixed by head-over-head

t
ip
rotation at 20 rpm for 24 hours at room temperature. The liquid and residual solid were

cr
separated by centrifugation at 3,300 g for 5 min and the drug concentration was determined

by UHPLC whereas the residual solid drug was assessed by high-throughput (HT)

us
transmission X-ray Powder Diffraction (XRPD). When assessing residual solid, XPRD is
an
considered as gold standard however, when low amounts of sample are available Raman

microscopy might serve as an alternative as well [53].


M

Caffeine, carbamazepine and piroxicam were chosen as model compounds for


ed

analysis. The results correlated well to the traditional shake-flask method and to other

published data. It is an easy assay to perform and according to the amount of data that it
pt

generates, it is classified as medium throughput. Still, the authors concluded that it is suited
ce

for high throughput experimentation and automation. Due to the parallel measurement of

solubility and of crystal form, it is applicable in drug development and pre-formulation.


Ac

Problems arise when screening high viscosity excipients and therefore, it is limited to

screening of the low viscosity ones [48,53].

Potentiometric approaches

Avdeef [54] was the one who initially introduced potentiometric based approaches

into solubility profiling however, two methods which are relatively demanding from the
perspective of necessary equipment are available at present, the pSol Gemini and CheqSol

systems (Table 1) [2]. For these two methods which run on a semiautomatic basis, the pH

electrode and capillary dispenser tips are placed in the glass tube which is necessary for the

process of titration. After the pH electrode is calibrated and the titration solutions are

standardized, an acid-base blank titration is performed which is needed to calculate solubility

and measure the electrode error. For the pSol method an octanol water partition coefficient

(logP) is required to calculate solubility by using the Hansch equation whereas and an exact

pKa/pKb is fundamental to determine solubility [28,55].

t
ip
With the pSol method, the time to determine solubility depends on the compound

cr
which sometimes might be more than 24 hours. The same can be said concerning the

substance requirements as the amount depends principally on compound solubility. In silico

us
calculations to estimate the amount required are obligatory for pSol and an initial weight of
an
<100 μg is considered sufficient. This amount has been reported to be adequate for a pH

profile determination as well. Additionally, the workload in the laboratory when using pSol is
M

lighter. It is viewed as an economical method in terms of compound consumption and the


ed

Food and Drug Administration (FDA) considers it acceptable for the evaluation of solubility

for BCS classification [56]. Nevertheless, it is limited to ionizable compounds only. The
pt

pH/solubility profiles determined by pSol method have also been referred to as the industry
ce

ʻgold standardʼ [57]. The potentiometric nature of the method also means that the medium

options are limited. Lastly, the method is a low throughput one as it can generate solubility
Ac

data for only about 3 poorly soluble compounds per week [28].

As for CheqSol (Chasing Equilibrium), it was first introduced by Stuart et al [58]. It

has been reported that the technique assesses kinetic, thermodynamic (equilibrium) and

intrinsic solubility. Additional advantages were highlighted when excipients were tested in

terms of drug formulation in a study by Fornells at al. [59] since the method permits the
determination of both, the extent and duration of supersaturation. This is predominantly

significant for poorly soluble drugs as supersaturation is often employed as a strategy to

improve their bioavailability. CheqSol contains five stages which include dissolution, seeking

precipitation, additional precipitation, chasing equilibrium and re-dissolution. Prior

knowledge of the pKa of the compound is necessary. Initially, the kinetic solubility is

established from the concentration of the solution when a precipitate first appears as the pH

of a solution of the drug in ionized form is adjusted by adding acid or base titrant to convert

the substance to its unionized form.

t
ip
As the experiment continues, the equilibrium solubility is subsequently determined in

cr
the presence of precipitated neutral solid, by monitoring pH changes induced by precipitation

and actively seeking equilibrium pH where equal amount of the sample is precipitating and

us
dissolving per unit time. The quantity of the tested solid that is needed for establishing
an
solubility must ensure a concentration above its intrinsic solubility when fully neutral to

allow precipitation once the pH is adjusted accordingly. If substance precipitation is not


M

detected, a larger quantity is required. A minimum of 2-5 mg was reported for the
ed

experimental volume of 10 mL, but for substances that exhibit a higher solubility, larger

amounts are of course necessary [58]. Only 20 - 80 minutes are required for kinetic and
pt

equilibrium solubility determination per sample, making it faster than the pSol method [60].
ce

Alas, the method is also restricted only to ionizable compounds. Seeing as the CheqSol

method essentially considers the Henderson-Hasselbalch equation always valid since


Ac

pH/solubility profiles cannot be directly measured, in some cases it could potentially lead to a

source of systematic error [44]. Nevertheless, the error would not affect the experimentally

established kinetic or intrinsic values but only the pH/solubility profiles.


Column Elution Method

As indicated by the name, the apparatus employed in the column elution method

consists of a microcolumn that contains either glass beads, silica gel or sand all of which are

inert carrier material, together with an excess of test substance. A small plug of glass wool is

used to keep this inert material in place. Constant temperature in the column needs to be

sustained. The preferred eluent is double distilled water however, deionized water with a

resistivity above 10 megohms/cm and a total organic carbon content below 0.01% can be

used. When connected to a recirculating pump, the headspace of the column should have the

t
ip
capacity for at least five bed-volumes of water and five samples. In order to remove water

cr
soluble impurities, the first five bed volumes are discarded. The recirculating pump can be

either a peristaltic or membrane pump by which a flow of approximately 25 mL/h

us
(corresponding to 10 bed volumes per hour) should be efficiently maintained. At this point, it
an
is necessary to be cautious as contamination and/or adsorption may occur with the tube

material. Alternatively, the microcolumn can be connected to a levelling vessel in which a


M

flow rate of approximately 25 mL/h is required too [61,62].


ed

Once the eluate fractions are collected, the next step requires to check if colloidal

matter is present by means of the Tyndall effect. If any particles are present, it indicates a
pt

poor filtering action of the column, rendering the test invalid. Gas or liquid chromatography,
ce

titration, photometric or polarographic methods are appropriate for sample analysis.

Equilibrium is attained when the mass concentration of the eluate is constant. In contrast to
Ac

the shake flask method (Table 1), the column elution method applies to substances with low

solubilities. It is not applicable to volatile substances and in order to determine solubility,

substances must be as pure and stable in water. Nevertheless, further research is still vital in

finding the most suitable carrier material, as the ones previously mentioned may not yet be
optimal. The construction of the column and regulation of the flow rate require optimization

as well [61,62].

Conclusion

Due to the status of solubility in the design of drug compounds as well as in the

development and optimization of drug manufacturing processes, an accurate understanding

and determination of this property is valuable in drug discovery. While medium to high

throughput assays certainly have their advantages, low throughput solubility measurements

are still performed to provide more accurate information relevant to the formulation strategies

t
ip
and overall profile of the drug candidate developability [63]. The idea of having only one

cr
consistent solubility method for both, the discovery and development phase may seem

desirable at first however, various factors such as compound quantity, speed and workflow

us
differences prevent such a set-up [2]. Even today, solubility assays are not ʻone size fits allʼ
an
therefore, different methods are applied at different stages of drug discovery and

development [8]. Thus, recognizing the limitations of each assay is vital so the data could be
M

interpreted properly.
ed

Acknowledgement
pt

This research was financially supported by Slovenian Research Agency (ARRS) [P1-0189].
ce

Disclosure statement
Ac

No potential conflict of interest was reported by the authors.


References
[1] Amidon GL, Lennernas H, Shah VP, et al. A Theoretical Basis for a Biopharmaceutic
Drug Classification: The Correlation of In Vitro Drug Product Dissolution and In Vivo
Bioavailability. Pharm Res. 1995;12:413-420.
[2] Alsenz J, Kansy M. High throughput solubility measurement in drug discovery and
development. Adv Drug Deliv Rev. 2007;59:546–567.
[3] Rinaki E, Valsami G, Macheras P. Classification System: The Central Role of
Dose/Solubility Ratio. Pharm Res. 2003;20:1917–1925.
[4] Di L, Kerns E, Carter G. Drug-Like Property Concepts in Pharmaceutical Design. Curr
Pharm Des. 2009;15:2184–2194.
[5] Wang J, Urban L. The impact of early ADME profiling on drug discovery and
development strategy. Drug Discov. 2004;4:72–96.

t
[6] Elder D, Holm R. Aqueous solubility: Simple predictive methods (in silico, in vitro

ip
and bio-relevant approaches). Int J Pharm. 2013;453:3–11.

cr
[7] Baka E, Comer JEA, Takács-Novák K. Study of equilibrium solubility measurement
by saturation shake-flask method using hydrochlorothiazide as model compound. J
Pharm Biomed Anal. 2008;46:335–341.
[8]
chemists. Drug Discov Today. 2006;11:1012–1018. us
Bhattachar SN, Deschenes LA, Wesley JA. Solubility: it’s not just for physical
an
[9] (TONY)TONG W-Q. Practical Aspects of Solubility Determination in Pharmaceutical
Preformulation. In: Augustijns P, Brewster ME, editors. Solvent systems and their
selection in pharmaceutics and biopharmaceutics. New York (NY): Springer; 2007. p.
M

137-149.
[10] Wang Z, Burrell LS, Lambert WJ. Solubility of E2050 at various pH: A case in which
ed

apparent solubility is affected by the amount of excess solid. J Pharm Sci.


2002;91:1445–1455.
[11] Murdande SB, Pikal MJ, Shanker RM, et al. Aqueous solubility of crystalline and
pt

amorphous drugs: Challenges in measurement. Pharm Dev Technol. 2011;16:187–200.


[12] Kawakami K, Miyoshi K, Ida Y. Impact of the amount of excess solids on apparent
ce

solubility. Pharm Res. 2005;22:1537–1543.


[13] Comer J, Judge S, Matthews D, et al. The intrinsic aqueous solubility of indomethacin.
Ac

Admet Dmpk. 2014;2:18–32.


[14] Higuchi T, Shih F-ML, Kimura T, et al. Solubility Determination of Barely Aqueous-
Soluble Organic Solids. J Pharm Sci. 1979;68:1267–1272.
[15] Lipinski CA, Lombardo F, Dominy BW, et al. Experimental and computational
approaches to estimate solubility and permeability in drug discovery and development
settings. Adv Drug Deliv Rev. 1997;23:3–25.
[16] Mora CP, Martínez F. Solubility of naproxen in several organic solvents at different
temperatures. Fluid Phase Equilib. 2007;255:70–77.
[17] Etzweiler F, Senn E, Schmidt HWH. Method for Measuring Aqueous Solubilities of
Organic Compounds. Anal Chem. 1995;67:655–658.
[18] Loftsson T, Hreinsdóttir D, Másson M. Evaluation of cyclodextrin solubilization of
drugs. Int J Pharm. 2005;302:18–28.
[19] Buch P, Meyer C, Langguth P. Improvement of the wettability and dissolution of
fenofibrate compacts by plasma treatment. Int J Pharm. 2011;416:49–54.
[20] U.S. Food and Drug Administration. Guidance for Industry: waiver of in vivo
bioavailability and bioequivalence studies for immediate-release solid oral dosage
forms based on the Biopharmaceutics. Classification system. [Internet]. [Cited 2019
Aug 16]. Available from: https://www.fda.gov/regulatory-information/search-fda-
guidance-documents/waiver-vivo-bioavailability-and-bioequivalence-studies
immediate-release-solid-oral-dosage-forms
[21] European Medicines Agency. Evaluation of medicines for human use. Committee for
proprietary medicinal products. Note for guidance on the investigation of

t
bioavailability and bioequivalence. [Internet]. [Cited 2019 Aug 16]. Available from:

ip
https://www.ema.europa.eu/en/documents/scientific-guideline/draft-note-guidance-
investigation-bioavailability-bioequivalence_en.pdf

cr
[22] Avdeef A, Fuguet E, Llinàs A, et al. Equilibrium solubility measurement of ionizable
drugs – consensus recommendations for improving data quality. ADMET&DMPK.

[23]
2016;4:117-178.

us
Kawabata Y, Wada K, Nakatani M, et al. Formulation design for poorly water-soluble
an
drugs based on biopharmaceutics classification system: Basic approaches and practical
applications. Int J Pharm. 2011;420:1-10.
[24] Brittain HG. Spectral Methods for the Characterization of Polymorphs and Solvates. J
M

Pharm Sci. 1997;86:405–412.


[25] Pan L, Ho Q, Tsutsui K, Takahashi L. Comparison of chromatographic and
spectroscopic methods used to rank compounds for aqueous solubility. J Pharm Sci.
ed

2001;90:521–529.
[26] Dalvit C, Flocco M, Knapp S, et al. High-Throughput NMR-Based Screening with
pt

Competition Binding Experiments. J Am Chem Soc. 2002;124:7702–7709.


[27] Mohan R, Lorenz H, Myerson AS. Solubility Measurement Using Differential
ce

Scanning Calorimetry. Ind Eng Chem Res. 2002;41:4854–4862.


[28] Glomme A, März J, Dressman JB. Comparison of a miniaturized shake-flask solubility
method with automated potentiometric acid/base titrations and calculated solubilities. J
Ac

Pharm Sci. 2005;94:1–16.


[29] Miller JM, Collman BM, Greene LR, et al. Identifying the Stable Polymorph Early in
the Drug Discovery–Development Process. Pharm Dev Technol. 2005;10:291–297.
[30] Meylan WM, Howard PH. Atom/fragment contribution method for estimating octanol-
water partition coefficients. Perspect Drug Discov Des. 2000;19:67–84.
[31] Bernard F, Frank W. Physicochemical parameters as tools in drug discovery and lead
optimization. In: Bernard T, van de Waterbeemd H, Gerd F, Richard G, editors.
Pharmacokinetic optimization in drug research. Wiley-VCH; 2001. p. 257–274.
[32] Abraham MH, Le J. The correlation and prediction of the solubility of compounds in
water using an amended solvation energy relationship. J Pharm Sci. 1999;88:868–880.
[33] ACD/Solubility [Internet]. Toronto, Ontario [cited 2019 Mar 20] Available from:
https://www.acdlabs.com/products/percepta/predictors.php
[34] Gopi SP, Banik M, Desiraju GR. New Cocrystals of Hydrochlorothiazide: Optimizing
Solubility and Membrane Diffusivity. Cryst Growth Des. 2017;17:308–316.
[35] Banerjee R, Bhatt PM, Ravindra N V., et al. Saccharin Salts of Active Pharmaceutical
Ingredients, Their Crystal Structures, and Increased Water Solubilities. Cryst Growth
Des. 2005;5:2299–2309.
[36] Rajput L. Stable Crystalline Salts of Haloperidol: A Highly Water-Soluble Mesylate
Salt. Cryst Growth Des. 2014;14:5196–5205.
[37] da Silva CC, Cirqueira M de L, Martins FT. Lamivudine salts with 1,2-dicarboxylic

t
acids: A new and a rare synthon with double pairing motif fine-tuning their solubility.

ip
CrystEngComm. 2013;15:6311-6317.
[38] Jantratid E, Janssen N, Chokshi H, et al. Designing biorelevant dissolution tests for

cr
lipid formulations: Case example – Lipid suspension of RZ-50. Eur J Pharm
Biopharm. 2008;69:776–785.

us
[39] Chen J, Wang J, Schwab LP, et al. Benzimidazole Analogs as Potent Hypoxia
Inducible Factor Inhibitors: Synthesis, Biological Evaluation, and Profiling Drug-like
Properties. Anticancer Res. 2014;34:3891–3904.
an
[40] Ruff A, Fiolka T, Kostewicz ES. Prediction of Ketoconazole absorption using an
updated in vitro transfer model coupled to physiologically based pharmacokinetic
M

modelling. Eur J Pharm Sci. 2017;100:42–55.


[41] Bergström CAS, Norinder U, Luthman K, Artursson P. Experimental and
computational screening models for prediction of aqueous drug solubility. Pharm Res.
ed

2002;19:182–188.
[42] Bergström CAS, Luthman K, Artursson P. Accuracy of calculated pH-dependent
aqueous drug solubility. Eur J Pharm Sci. 2004;22:387–398.
pt

[43] Avdeef A. Phosphate Precipitates and Water-Soluble Aggregates in Re-analyzed


Solubility-pH Data of Twenty-five Basic Drugs. Admet Dmpk. 2014;2:43–55.
ce

[44] Avdeef A. Suggested Improvements for Measurement of Equilibrium Solubility-pH of


Ionizable Drugs. Admet Dmpk. 2015;3:84–109.
Ac

[45] Völgyi G, Baka E, Box KJ,et al. Study of pH-dependent solubility of organic bases.
Revisit of Henderson-Hasselbalch relationship. Anal Chim Acta. 2010;673:40–46.
[46] Kerns EH, Di L, Carter GT. In vitro solubility assays in drug discovery. Curr Drug
Metab. 2008;9:879–885.
[47] Chen X-Q, Venkatesh S. Miniature Device for Aqueous and Non-aqueous Solubility
Measurements During Drug Discovery. Pharm Res. 2004;21:1758–1761.
[48] Dai WG, Pollock-Dove C, Dong LC, et al. Advanced screening assays to rapidly
identify solubility-enhancing formulations: High-throughput, miniaturization and
automation. Adv Drug Deliv Rev. 2008;60:657–672.
[49] Tan H, Semin D, Wacker M, Cheetham J. An automated screening assay for
determination of aqueous equilibrium solubility enabling SPR study during drug lead
optimization. JALA - J Assoc Lab Autom. 2005;10:364–373.
[50] Bharate SS, Vishwakarma RA. Thermodynamic equilibrium solubility measurements
in simulated fluids by 96-well plate method in early drug discovery. Bioorganic Med
Chem Lett. 2015;25:1561–1567.
[51] Roy D, Ducher F, Laumain A, Legendre JY. Determination of the aqueous solubility
of drugs using a convenient 96-well plate-based assay. Drug Dev Ind Pharm.
2001;27:107–109.
[52] Alsenz J, Meister E, Haenel E. Development of a partially automated solubility
screening (PASS) assay for early drug development. J Pharm Sci. 2007;96:1748–1762.
[53] Wyttenbach N, Alsenz J, Grassmann O. Miniaturized assay for solubility and residual
solid screening (SORESOS) in early drug development. Pharm Res. 2007;24:888–898.

t
ip
[54] Avdeef A. pH‐ metric Solubility. 1. Solubility‐ pH Profiles from Bjerrum Plots. Gibbs
Buffer and pKa in the Solid State. Pharm Pharmacol Commun. 1998;4:165–178.

cr
[55] McFarland JW, Avdeef A, Berger CM, et al. Estimating the Water Solubilities of
Crystalline Compounds from Their Chemical Structures Alone. J Chem Inf Comput

us
Sci. 2001;41:1355–1359.
[56] FDA, U.S. Department of Health and Human Services Food and Drug Administration
Center for Drug Evaluation and Research (CDER) BP (Guidance 3618FNL) [Internet].
an
[cited 2019 Feb 26]. Available from:
https://www.fda.gov/downloads/Drugs/Guidances/ucm070246.pdf.
M

[57] Valko K, Reynolds DP. High-throughput physicochemical and in vitro ADMET


screening. Am J Drug Deliv. 2005;3:83–100.
[58] Stuart M, Box K. Chasing Equilibrium : Measuring the Intrinsic Solubility of Weak
ed

Acids and Bases. 2005;77:983–990.


[59] Fornells E, Fuguet E, Mañé M, et al. Effect of vinylpyrrolidone polymers on the
solubility and supersaturation of drugs; a study using the Cheqsol method. Eur J Pharm
pt

Sci. 2018;117:227-235.
[60] Box KJ, Völgyi G, Baka E, et al. Equilibrium versus kinetic measurements of aqueous
ce

solubility, and the ability of compounds to supersaturate in solution—a validation


study. J Pharm Sci. 2006;95:1298–307.
Ac

[61] Veith GD, Comstock VM. Apparatus for Continuously Saturating Water with
Hydrophobic Organic Chemicals. J Fish Res Board Canada. 1975;32:1849–1851.
[62] Guidelines for The Testing of Chemicals, OECD 105, Water Solubility (Column
Elution Method-Shake Flask Method), OECD, Paris, France. Organ Econ Coop Dev.
1981 [Internet] [cited 2019 Aug 16]. Available from:
http://www.oecd.org/chemicalsafety/risk-assessment/1948185.pdf
[63] Di L, Fish P V., Mano T. Bridging solubility between drug discovery and
development. Drug Discov Today. 2012;17:486–95.
Table 1. Comparison between shake flask, potentiometric and column elution methods
Shake Flask Method Potentiometric Methods Column Elution Method
(SF) (pSol, CheqSol)
 Simple experimental  Lighter workload  Simple experimental
set-up  Lower compound set-up
 Inexpensive consumption in  Inexpensive apparatus
 Short time to reach
ADVANTAGES

apparatus comparison to SF
 Easy to learn  Useful in drug saturation compared to SF
 Most accurate formulation studies (e.g.
method to date in investigating
excipients)
 Established solubility
values correlate well with
SF

t
 Time consuming  More demanding in  Large drug quantity is

ip
 Labour intensive terms of laboratory required
 Not suitable for high equipment  Better suited carrier

cr
throughput  Limited medium options material is desirable
DISADVANTAGES

screening  Low throughput  Loading of carrier material


 Large drug quantity

us
 Restricted to ionizable can be problematic when
is required compounds the substance is deposited
as an oil or different
an
crystal phase (inaccurate
results may be generated)
 Optimization of column
M

construction and flow rate


are still desired
ed
pt
ce
Ac
Table 2. Comparison of miniaturized methods based on the classical shake flask (SF) method
Miniaturize
96-well Plate
d Shake Small Scale Shake Miniature
Methods (PASS,
Flask Flask Method Devices
SORESOS)
Method [41,42] [47]
[48-53]
[28]
“medium” “medium”, but
“low” with “high”: 600
approx. 20 96-well format and
Throughput possibility to measurements / week
compounds/ automation are
increase for the PASS method
week possible

Fast
Solid dispensing Slow Slow Slow
(solvent evaporation)

Experiment 6h for most 23h (THESA)

t
24h 24h or more
duration compounds 24h (SORESOS)

ip
cr
Filtration
after
Continuous

us
equilibration
Ultra- filtration,
Phase separation , Centrifugation
centrifugation filter blockage
filter
isues
sorption
an
possible

Necessary demanding –
simple, More demanding
M

equipment besides simple complex


consumable equipment in most cases
(U)HPLC device
ed

Solid sample
Yes, especially for
recovery and Difficult Possible Difficult
SORESOS method
analysis
pt

Risk of formation High (PASS), but was


of new solid Low Low Low controlled by DSC and
ce

physical forms TGA

Aplicability in
High, especially PASS
Ac

studies of Possible Possible Low


method
excipient effects

S-ar putea să vă placă și