Você está na página 1de 11

Enhancement of Drug Solubility in Supramolecular

and Colloidal Systems

ANDRÁS SÜLE,1 LAJOS SZENTE,2 FERENC CSEMPESZ1


1
Laboratory of Colloid and Supramolecular Systems, Eötvös University, Institute of Chemistry, P.O. Box 32,
H-1518 Budapest 112, Hungary
2
Cyclolab Cyclodextrin Research & Development Ltd, P.O. Box 435, H-1525 Budapest, Hungary

Received 20 December 2007; revised 31 March 2008; accepted 13 April 2008


Published online 27 May 2008 in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jps.21437

ABSTRACT: Statins, as efficient HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme-A)


reductase inhibitors are widely used in the management of cardiovascular diseases.
Interactions in aqueous solutions between highly lipophilic statins and cyclodextrins
(CDs) in the absence and the presence of a dissolved polymer or its monomeric
compound, respectively, were studied. The solubility of lovastatin and simvastatin at
various temperatures and pHs were investigated by phase-solubility measurements.
Surface activity of solutes in binary (CD–statin) and ternary (CD–statin–polymer)
systems was studied by determining the surface tension of the solutions. For the
characterization of the CD–statin inclusion complexes, stability constants for associates
of different molar ratios have been calculated. It was shown that complexation may lead
to improvement of the aqueous solubilities of both statins by 1–2 orders of magnitude.
Especially, randomly methylated b-cyclodextrin (RAMEB) showed outstanding solubi-
lizing effects. In binary systems dominantly CD–statin associates of 1:1 molar ratios
form, which exhibit considerable surface activity. RAMEB forms more stable complexes
with these drugs than the native b-CD, and also the surface activity of the former solutes
is higher. In polymer-containing ternary systems the solubility of both statins could be
further improved. The enhanced drug solubilities can be ascribed to the formation of
CD–statin–polymer associates with supramolecular structure. A portion of the surface
active CD–statin complexes are very likely anchored at the macromolecular chains. In
these solutions, the total amounts of solutes are composed of the sum of the ‘‘free’’ binary
and the supramolecular ternary associates. ß 2008 Wiley-Liss, Inc. and the American
Pharmacists Association J Pharm Sci 98:484–494, 2009
Keywords: cyclodextrins; dissolution; inclusion compounds; supramolecular associ-
ates; nanocapsules; macromolecular drug delivery

INTRODUCTION growing amount of papers pointed out that statins


provide crucial benefits when used in either lipid
Improvement of the efficacy and safety of existing lowering therapy or coronary heart disease (CHD)
drugs is a key issue in pharmaceutical sciences therapy. It is well documented that statins inhibit
and the related fields. Over the past few years, a the rate-limiting steps in cholesterol synthesis.
The 3-hydroxy-3-methylglutaryl-like domain of
statins may attach to the appropriate binding site
of the reductase enzyme and therefore, sterically
Correspondence to: Ferenc Csempesz (Telephone: þ36-1-
372-2544; Fax: þ36-1-372-2592; E-mail: csf@chem.elte.hu) prevent the natural substrates to bind.1 Sche-
Journal of Pharmaceutical Sciences, Vol. 98, 484–494 (2009) matic molecular structure of two statins is
ß 2008 Wiley-Liss, Inc. and the American Pharmacists Association illustrated in Figure 1.

484 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009


ENHANCEMENT OF DRUG SOLUBILITY 485

Figure 1. Chemical structure of lovastatin and sim-


vastatin.
Figure 2. (a) Chemical structure of alpha-cyclodex-
trin. (b) Computer-simulated surface model of alpha-
Chemically different statins (such as lovastatin, cyclodextrin.12
simvastatin, pravastatin, atorvastatin) differ
mostly in their absorption parameters, plasma and secondary hydroxyl groups located around
protein binding and solubility, therefore they may their narrower and wider rim, respectively.11 The
exhibit varied efficacy in reducing the low-density most widely used natural cyclodextrins consist of
lipoprotein (LDL) cholesterol levels even at the 6, 7, and 8 glucose units and according to the
same dosages. In general, by doubling the statin number of monomers in the macrocylce, they are
dose, LDL-cholesterol level is reduced by more named as a-, b-, and g-cyclodextrin. It is well
than 7%.2 Nevertheless, 22–45% reduction in the known that CD molecules have a hydrophobic
blood triglyceride levels of hypertriglyceridemic inner cavity, while the large number of hydroxyl
patients and a minor elevation in high-density groups on the outer surface makes them water-
lipoprotein (HDL) cholesterol levels could also be soluble (Fig. 2).
observed. Due to this special molecular structure, lipo-
A hard obstacle in statin therapy is the very low philic guest molecules (or molecule parts) of the
aqueous solubility of the pharmacons. Their appropriate size can be encapsulated in the
bioavailability is, therefore, also low and may cavities of CD molecules, hereby forming inclusion
exhibit high variability in individuals.3,4 In complexes.13 The chemical structure of CDs can be
addition, the HMG-CoA reductase inhibitors are expediently modified by esterification of the 2-, 3-,
associated with two uncommon, but important and 6-hydroxyl groups and also, other functional
side effects: asymptomatic elevation of liver groups can be introduced at these sites. For
enzymes and skeletal muscle abnormalities. instance, these structural changes perturb the
These latter can range from benign muscle pain internal hydrogen-bond network of b-CDs, result-
to myopathy (severe muscle pain and weakness) ing in an elevated water-solubility.14
and life-threatening rhabdomyolysis.5,6 Despite of Extensive studies showed that more than 40%
these shortcomings, monotherapy with either of developed drug molecules exhibit poor bio-
lovastatin or simvastatin is proven to be safe, pharmacological properties, partly because of
but concerning their interactions in complex their diminished dissolution and/or inadequate
delivery systems, many questions may arise.7,8 permeability.15 A great challenge for the present
Both statin molecules are known to have a ring- pharmaceutical research is, therefore, to formu-
closed lactone, and ring-opened hydroxyacid form, late orally administrable dosage forms with
from which the latter is the active substance.9,10 decent bioavailability of pharmacons. To achieve
The ring-closed prodrugs are metabolized to their this, the drug must be dissolved in the gastro-
respective acidic form in liver cells. It is possible intestinal tract and it must be absorbed in a way
however that hydrolysis might occur in aqueous that sufficient drug levels arise at the pharmaco-
media that affects their biopharmacological prop- logically active sites.
erties too. During drug formulation it is, there- To date, only little information is available on
fore, crucial to ensure the appropriate chemical the relevance of colloids in controlling physico-
stability of statins. chemical and pharmacological behavior of drugs
Cyclodextrins (CDs) are peculiar cyclic oligo- in complex delivery systems. Much less is known
mers of a-D-glucose and are extensively used as about how colloidal components may affect non-
complexing agents for lipophilic compounds. Their covalent interactions of pharmacons in aqueous
shapes resemble truncated cones with primary media. Possible use of water-soluble polymers to

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009
486 SÜLE, SZENTE, AND CSEMPESZ

improve inclusion properties of CDs is well Table 1. Ranges of Additive Concentrations in


documented by Loftsson et al.16 Nevertheless, Different Measurements
the effect of colloidal additives on the complex
formation between cyclodextrins and pharma- Concentration
Measurement Component Range (mg/mL)
cons, and interactions of colloids with the formed
supramolecules in complex drug delivery systems Phase solubility a-, b-, g-CD, 0–50
has not been revealed in detail. (binary systems) RAMEB
Complexation of lovastatin and simvastatin Phase solubility RAMEB 0–12
with cyclodextrins, and the effect of water-soluble (ternary systems) PVP 0–0.20
macromolecules and their noncolloidal chemical NEP 0–0.10
Surface tension b-CD, RAMEB 0–10
analogues on the formation of supramolecular
PVP 0.20
complexes are in the focus of the present study.
Solubilization of statins in aqueous cyclodextrin
solutions and its control in the absence and the Millipore type GS 0.22 mm membrane filter, the
presence of a macromolecular colloid and its absorption spectra of the dissolved pharmacon
monomeric compound, respectively, have been were then taken in the range of 190–400 nm
investigated. Possible ways of enhancing the wavelengths. The highly lipophilic character of
aqueous solubility of the drugs in both binary the molecules of both drugs might raise the ques-
and ternary systems have also been discussed. tion of possible drug adsorption to the membrane
filters. Measuring the statin content of both
ethanolic and aqueous solutions throughout 10
MATERIALS AND METHODS consecutive filtration cycles proved that no sig-
nificant adsorption occurs during the separation
Materials procedures.
The statin content of the solutions was deter-
Lovastatin and simvastatin of USP23-grade
mined with a UV-spectrophotometric assay based
(Chiesi Farmaceutici SpA, Parma, Italy), a-, b-,
on the PhEur directives.18 The absorbance of
g-cyclodextrin and RAMEB [randomly methylated
lovastatin and simvastatin was measured at 236
b-cyclodextrin, average degree of methylation: 1.8
and 240 nm, respectively. For these measure-
methyl groups/glucose monomers], as well as PVP
ments a computer-controlled spectrophotometer
K30 polyvinyl pyrrolidone (Fluka AG, Buchs SG,
(Perkin-Elmer Lambda Series 2S instrument) was
Switzerland) and NEP [1-Ethyl-2-pyrrolidinone]
used and the raw spectra were evaluated by
(Sigma-Aldrich Chemie GmbH, Munich, Germany)
OriginLab Origin 7.0 software suite. The solubi-
were used in the experiments. The cyclodextrins
lity isotherms were calculated by using calibra-
were of analytical grade and manufactured by
tion curves determined with ethanolic statin
Cyclolab R&D Ltd (Budapest, Hungary).
solutions of known concentrations. It is worth-
while to mention that compared to aqueous and
CD-containing matrices, the ethanol did not
Solubility Measurements
distort either the UV-spectra of the drugs, or
Phase-solubility studies of the statins in aqueous the response factor. Therefore, throughout these
solutions of several cyclodextrins of different measurements no use of correction factors was
chemical structures were carried out according necessary.19 As noted in the Introduction Section,
to the Higuchi–Connors procedure.17 Various preserving the chemical stability of statins is a
amounts of CD were generally dissolved in dis- crucial factor in these experiments. In aqueous
tilled water and lovastatin or simvastatin was solutions, no changes in the molecular structures
added to the solutions in vast excess (see Tab. 1). of the drugs could be detected either by pre-
Twenty-four hours of temperature-programmed liminary conductometric, circular dichroism, or by
incubation period in a refrigerated bath (HAAKE NMR measurements.
PhoenixII C35P instrument) was ensured for the The solubility of statins was also investigated
dissolution of the pharmacon at 14, 25, 36, and at two moderate pH values, relevant to that of
478C, respectively. After longer equilibration the gastro-intestinal tract. To adjust the pH of
times (3–14 days), no definite increase in drug the solutions, HCl (pH ¼ 1.2, cHCl ¼ 0.08 M,
solubilities could be detected. After removing the cNaCl ¼ 0.034 M) solution and phosphate buffer
nondissolved statin from the solutions with (pH ¼ 7.4, c ¼ 0.25 mM), was used, respectively.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009 DOI 10.1002/jps
ENHANCEMENT OF DRUG SOLUBILITY 487

The buffers were prepared on the basis of the constants, using the total concentrations of the
directives of the European Pharmacopoeia’s reactants:
5th Edition and the Hungarian Pharmacopoeia’s cG ¼ Spbpq ½Gp ½Hq (3)
8th Edition.18,20
For the preparation of statin–CD–polymer cH ¼ Sqbpq ½Gp ½Hq (4)
ternary systems, a specially controlled thermal
program was used in a similar way described by where b10 and b01 values are equal to 1.
Loftsson and Masson.21 The appropriate mixtures Knowing the total concentrations (cG, cH) and
(listed in Tab. 1) were put in a computer-driven assuming realistic bpq values, [G] and [H] can be
refrigerated bath, where the samples were heated determined in an iterative procedure. Recalculat-
up to 708C and held at this elevated temperature ing the values of total concentrations (c), the
for 2 h. After that, the solutions were cooled down minimum of the next equation represents the best
to 258C and kept at this temperature for 22 h. UV- fit for the iterative process:
spectra of solutions indicated that such a rapid
U ¼ Sðccalculated cmeasured Þ2 (5)
thermal treatment does not cause alterations in
the molecular structure of the statins. Seventy
Assuming possible molecular structures of com-
degree Celsius was found as an optimum tem-
plexes that may really form in statin–CD binary
perature where the thermal degradation of
systems, two combinations may have relevance:
components could be avoided and also, after the
(a) 1:1 complex is formed, (b) 1:1 and 1:2 (statin/
incubation period at this temperature sufficient
CD) complexes are both formed. The stability
solubility enhancing effect could be detected (for
constants of 1:1 statin–CD complexes are denoted
more details see Tab. 4).
by b11 (usually denoted by K11). The cumulative
stability constants of 1:2 stain–CD complexes are
Surface Tension Determination denoted by b12. K12 —is the stepwise stability
constant for the complexation step where a second
Surface tension of aqueous solutions was mea-
CD molecule is bound to a 1:1 statin–CD complex
sured by computer controlled Wilhelmy-plate
(b12 ¼ K12
b11 or b12 ¼ K12
K11).
method, using a KSV Sigma 70 instrument.
Accordingly, Eqs. (3) and (4) can be simplified as
In each measurement, the shown data are
follows:
the averages of at least three independent
experiments. In addition, the spectrophotometric cG ¼ ½G þ b11 ½G½H (3a)
assays have been validated by measurements
of 15 samples of the same composition, and cH ¼ ½H þ b11 ½G½H (4a)
standard deviation of the experimental data has
been accordingly calculated at several additive cG ¼ ½G þ b11 ½G½H þ b12 ½G½H2 (3b)
concentrations.
cH ¼ ½H þ b11 ½G½H þ 2b12 ½G½H2 (4b)

Theoretical Approach to the


CD–Statin Association
RESULTS AND DISCUSSION
Interactions that may lead to formation of
inclusion complexes between guest (G) and host Solubility of Pharmacons in Aqueous Media
(H) molecules can be represented as
Aqueous solubility of pharmacologically active
pG þ qH , Gp Hq (1) substances is a key parameter regarding their
biological availability. The solubility of both
where p and q indicate the stoichiometric factors.
lovastatin and simvastatin in water is extremely
The stability constant (b) for any associate can be
poor (Slovastatin ¼ 1.5 mg/mL, Ssimvastatin ¼ 2.0 mg/
defined in the following way:22,23
mL). Improving the aqueous solubility of these
½Gp Hq  pharmacons might offer, therefore, considerable
bpq ¼ (2)
ð½Gp  ½Hq Þ technological benefits without altering their
pharmacodynamic and metabolic (first-pass meta-
A computer program based on the rule of mass bolism above all) behavior. In Figures 3a and b
balance provides evaluation of different bpq and 4 typical solubility isotherms determined for

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009
488 SÜLE, SZENTE, AND CSEMPESZ

RAMEB proved to be effective solubilizer for these


drugs, enhancing their solubilities by two orders
of magnitude. The observed improvement of drug
solubilities can presumably be attributed to the
formation of inclusion complexes between the CD
and the statin molecules.14 The linear sections of
the isotherms support this notion. In the range of
CD concentrations up to about 20 mg/mL, a 1:1
complexation is sufficient to explain the observed
enhancement in solubility. It should be noted here
that b-CD itself has a rather low aqueous solu-
bility and therefore, the solubility of the assumed
b-CD–statin complexes can also be limited.24

Effect of Temperature and pH on Drug Solubilities


Complexation of statins with CD in aqueous
media was investigated at 14, 25, 36, and 478C.
At higher temperatures and neutral pH, the
aqueous solubilities of the natural statins only
slightly increased, but in cyclodextrin solutions
these were considerably enhanced. Data in
Figure 5a–d show that raising the dissolution
Figure 3. (a) Phase-solubility curves for lovastatin in temperature from 14 to 478C results in more than
cyclodextrin solutions, T ¼ 258C, pH ffi 6.5. (b) Phase- threefold increase in the solubility of both statins.
solubility curves for simvastatin in cyclodextrin solu- Somewhat less enhancement of the lovastatin
tions, T ¼ 258C, pH ffi 6.5. solubility could be observed. The solubility of
simvastatin in several CD-solutions could also be
improved.25
lovastatin and simvastatin in several CD solu- The apparent increase in the solubilities of
tions at 258C are shown. The marked points complexed drugs can be ascribed to the improved
and the corresponding error bars in the plots intrinsic solubilities of the uncomplexed pharma-
indicate the mean and the standard deviation, cons, as well. While the complexation constants
respectively, of the experimental data. tend to decrease with increasing temperature
These results well demonstrate that in solutions (Tab. 2), the increased intrinsic drug solubilities
of the cyclodextrins the solubility of both statins is can outweigh the modest decrease of the corre-
significantly increased. At higher CD concentra- sponding stability constants (see also Eq. 2).26,27
tions more than 10-fold increase in the solubility At physiologically relevant acidic and basic
of both pharmacons could be detected. Especially, pH-values, both statin-derivatives exhibited some-
what higher solubilities than in distilled water. In
CD-solutions the drug solubilities could be further
enhanced. Figure 6a and b illustrates that the
solubility of both lovastatin and simvastatin in
acidic solutions is increased by 40–100% in a wide
concentration range of b-CD, relative to the
corresponding values obtained in distilled water.
To evaluate these results, some structural
change of the dissolved statin molecules that
may take place in acidic media should be taken
into account. It is reasonable to assume that at
low pH the inactive lactone form of statins can
Figure 4. Phase-solubility curves for lovastatin and be transformed into active acidic forms.9 Such
simvastatin in RAMEB solutions, T ¼ 258C, pH ffi 6.5. a structural alteration may lead to a less

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009 DOI 10.1002/jps
ENHANCEMENT OF DRUG SOLUBILITY 489

hydrophobic form of statins, which may certainly pH ¼ 7.4, which corresponds to the basicity of
cause increases in the drug solubilities. the intestinal tract. As can be seen in
Higher enhancement in the solubilities of both Figure 6a and b, at this pH the solubility of both
statins were detected in buffered media at lovastatin and simvastatin is enhanced by mostly
100–150%, compared to those of their solubilities
measured in aqueous CD-solutions. Hydrolysis
and ionization of the lactone of statins at this pH,
and their enhanced complexation with CD may
alike contribute to the improvement of drug
solubilities.28
The results of phase solubility measurements
provide solid experimental evidences for the
complexation of statin molecules with cyclodex-
trins. The linear parts of the solubility isotherms
suggest that at low CD concentrations in aqueous
solutions presumably host-guest type inclusion
complexes of 1:1 molar ratios form, but consider-
ing the chemical structure of the statins and the
complexation preference of CDs,14 development of
other assemblies cannot be excluded. A possible
site of inclusion of statin with CD is schemati-
cally illustrated in Figure 7. (Formation of such
associates are supported by preliminary NMR
measurements.)
Considering the theoretical approaches22 dis-
cussed in the section of Materials and Methods,
stability constants for the statin–CD complexes
both of 1:1 and 1:2 molar ratios have been calculat-
ed. The b11 and b12 association constants calculat-
ed by iteration procedure are shown in Table 2.
These results indicate that formation of statin–
CD complexes of 1:2 molar ratios is less likely than
that of 1:1 associates. The K12 stepwise association
constants are lower than the corresponding b11
(K11) values. Also, the RAMEB-statin complexes
of 1:1 molar ratio are more stable than those of the
native b-CD. The appropriate stability constants
for the RAMEB-statin associates are about one
order of magnitude higher.
For comparison purposes, stability constants
have also been calculated directly from the solu-
bility curves using an approach offered by Iga
et al.29
m1
K11 ¼ (6)
S0 ð1 m1 Þ

Figure 5. (a) Effect of temperature on the aqueous


solubility of lovastatin in b-CD solutions, pH ffi 6.5. (b)
Effect of temperature on the aqueous solubility of lovas-
tatin in RAMEB solutions, pH ffi 6.5. (c) Effect of tem-
perature on the aqueous solubility of simvastatin in b-
CD solutions, pH ffi 6.5. (d) Effect of temperature on the
aqueous solubility of simvastatin in RAMEB solutions,
pH ffi 6.5.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009
490 SÜLE, SZENTE, AND CSEMPESZ

Table 2. Stability Constants of Statin–CD Complexes Determined by Iterative Computational Method

Temperature
(8C) Binary System b11 (M 1) K12 (M 1) b12 (M 2)
25 b-CD–lovastatin 1.86  102 2.2  101 1.13  102 1.5  101 2.1  104 3.74  103
36 b-CD–lovastatin 1.75  102 2.1  101 8.46  101 1.2  101 1.48  104 2.75  103
25 b-CD–simvastatin 2.88  102 3.2  101 1.61  102 2.1  101 4.63  104 7.94  103
36 b-CD–simvastatin 2.59  102 2.8  101 1.60  102 2.1  101 4.14  104 7.05  103
25 RAMEB–lovastatin 9.38  102 9.3  101 2.63  101 3.7  100 2.47  104 4.25  103
36 RAMEB–lovastatin 9.00  102 8.8  101 3.01  101 5.2  100 2.71  104 5.38  103
25 RAMEB–simvastatin 1.21  103 1.1  102 2.38  102 2.9  101 2.88  105 4.38  104
36 RAMEB–Simvastatin 7.41  102 7.3  101 6.10  101 8.6  100 4.52  104 7.77  103

where K11 is the stability constant of 1:1 the molecular mechanism of drug inclusion in
assemblies, m1 is the slope of the linear section greater details.
of the solubility curve, S0 its intersection with the
ordinate axis.
The K11 constants calculated from the solubility
isotherms according to Eq. 6 are shown in Table 3. Complexation in Ternary Systems
Estimation of the association constants in such In ternary systems, the effects of a water-soluble
a way provides somewhat higher values for K11, polymer and a chemically analogous compound of
but these data are in line with the corresponding its monomer, respectively, on the solubilization of
values listed in Table 2. No information can be statins by CDs were studied. By adding a third
obtained, however, from these results concerning component to a CD–statin solution, in principle a
conventional competition between the pharmacon
and the additive for the CD cavities may take
place. Such competition may result in a definite
decrease in the amount of the complexed statin.
Changes in the solubilities of statins in CD-
solutions due to the addition of 1-ethyl-2-pyrro-
lidinone (NEP) are illustrated in Figure 8. As
can be seen, the added NEP may notably reduce
drug solubility, presumably due to its preferred
complexation in CD cavities. The higher is the
concentration of the small molecular mass addi-
tive at constant CD concentration, the higher is
the reduction in the solubility of the drug.
These results confirm a distinct competitive
mechanism in the binary interactions between the
CD, the statin and the NEP molecules, res-
pectively. Complexation of the drug with CD is

Figure 6. (a) Aqueous solubility of lovastatin in b-CD


solutions at pH 1.2 and pH 7.4, T ¼ 258C. (b) Aqueous
solubility of simvastatin in b-CD solutions at pH 1.2 and Figure 7. A possible inclusion complexation of sta-
pH 7.4, T ¼ 258C. tins with cyclodextrins.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009 DOI 10.1002/jps
ENHANCEMENT OF DRUG SOLUBILITY 491

Table 3. Stability Constants for 1:1 CD–Statin


Complexes Determined from the Solubility Isotherms

Temperature
(8C) Binary System K11 (M 1)
25 b-CD–lovastatin 1.9  102 3.5  101
25 b-CD–simvastatin 2.1  102 3.8  101
25 RAMEB–lovastatin 1.5  103 2.1  102
25 RAMEB–simvastatin 1.7  103 2.4  102

hindered, and/or the pharmacon is displaced from Figure 9. Effect of PVP on lovastatin solubility in
the cavity of cyclodextrins by the preferred NEP RAMEB solutions, T ¼ 708C/258C, pH ffi 6.5.
molecules.
In polymer-containing systems, just an opposite systems presumably cannot be regarded as
effect of the macromolecular colloid on the solu- real equilibrium solubilities, rather as solubilities
bility of statins could be observed. Figure 9 well representing ‘‘frozen’’ metastable states. The
demonstrates that in solutions of CD–drug– polymer-containing samples stored at 258C for
polyvinyl pyrrolidone, the solubility of both 24 h showed no significant increase in the
statins, under the specified experimental condi- solubility of the statins. Heat treatment proved
tions, definitely increased. to be a crucial step, since the drug solubility could
At the studied CD-concentrations, the dissolved be enhanced only when a short ‘‘thermal jump’’ in
polymer resulted in 5–50% increase in the solu- the storage regimen was implied. In the studied
bility of lovastatin. Based on these solubility temperature range, the increase in the solubility
isotherms, stability constants for polymer con- of the drug seems to be proportional to the extent
taining systems could be estimated according to of ‘‘heat treatment’’ (Tab. 4). Without polymer,
the method of Iga et al.29 This method was such enhance of the drug solubilites could not be
originally proposed for host-quest type binary observed.
systems, so estimation of cumulative stability These results clearly indicate that under
constants for three component systems can suitable conditions, the dissolved macromolecules
only be regarded as a rough approximation. may significantly affect complexation of the drug
Nevertheless, for comparison purposes, cumula- with CD, but in a different way from that as the
tive stability constant for a ternary system of monomeric additives act.
lovastatin-RAMEB-PVP at 0.20 mg/mL polymer Loftsson et al.30 pointed out first that water-
concentration has also been estimated. Its soluble polymers can improve the dissolution of
approximated value is as high as K ¼ 6.5  103. certain guest molecules in cyclodextrin solutions
As noted in the section of Materials and Methods, through complex formation. The enhancement of
these complex systems were subjected to a heat solubility was attributed to the formation of ter-
treatment as described in the experimental nary associates. Possible increase of the solubi-
methods. Drug solubilities observed in such lizing effect of aqueous surfactant solutions by
water-soluble polymers was also shown.31 ‘‘Simple
adsorption’’ of individual surfactant molecules on
the polymer chains, or formation of ‘‘polymer-
aggregate complexes,’’ that is, complex formation
between the micelles and the polymer chains were
supposed as likely outcomes of the surfactant-
polymer interactions.32 Detailed mechanism of
the formation and structure of the ternary
complexes has not been revealed.
For a better understanding of the relevant
interactions in ternary systems of CD, statin and
PVP, surface activities of the solutes in aqueous
Figure 8. Effect of NEP on lovastatin solubility in CD–statin solutions with and without the polymer
RAMEB solutions, T ¼ 708C/258C, pH ffi 6.5. have also been studied.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009
492 SÜLE, SZENTE, AND CSEMPESZ

Table 4. Effect of Heat Treatment on the Solubility of Lovastatin in Ternary Systems

Change in Drug Change in Drug Change in Drug


Change in Drug Solubility, Solubility, Solubility,
PVP Solubility, 2.0 mg/mL 2.7 mg/mL 4.0 mg/mL
Peak Temp. Concentration Distilled RAMEB RAMEB RAMEB
(8C) (mg/mL) Water (%) Solution (%) Solution (%) Solution (%)
25 0.05 <1 <1 1.6 0.5 0.9 0.7
0.10 <1 <1 3.2 0.5 4.1 0.7
0.20 <1 <1 2.7 0.5 3.7 0.7
47 0.05 <1 1.6 0.3 2.2 0.7 1.1 0.9
0.10 <1 3.5 0.3 7.6 0.7 2.4 0.9
0.20 1.2 0.1 4.2 0.3 9.6 0.7 5.1 0.9
70 0.05 2.5 0.2 4.2 0.5 13.3 0.8 42.1 1.1
0.10 3.1 0.2 7.0 0.5 15.9 0.8 40.5 1.1
0.20 3.8 0.2 9.1 0.5 16.3 0.8 48.0 1.1

In Figure 10, the surface tensions of aqueous The enhanced drug solubilities in polymer solu-
CD solutions and CD–simvastatin solutions, tions can be likely ascribed to the formation of
respectively are plotted as a function of the CD ternary associates. At the elevated temperatures
concentration. used during the thermal treatments, host-guest
Considerable surface activities of the solutes in association is shifted towards enhanced complexa-
cyclodextrin solutions are demonstrated by these tion of the drug, that is, the concentration of CD–
curves. At higher CD concentrations, about 15– statin complexes is increased. The surface-active
25 mJ/m2 decrease in the surface tension of water complexes may attach to the dissolved macro-
could be detected. Without simvastatin, only molecules in a way that they are anchored by their
the dissolved RAMEB exhibited surface activity. hydrophobic part at the polymeric chains. Hereby,
It can be concluded from these results that the concentration of the free associates in the
formation of CD–statin complexes leads to the solution may remain the same as that correspond-
development of surface-active solutes, which can ing to the equilibrium concentrations of the binary
accumulate at the air/water interface. In other complexes develop at 258C in polymer-free solu-
words, complexation of statin can also be regarded tions. The total concentration of the dissolved
as a CD-induced ‘‘amphiphilization’’ of the hydro- CD–statin complexes is evidently the sum of that
phobic drug. Dissolved CD–statin complexes with of the free and the bound associates. Accordingly,
an amphiphilic character may thus cause the at low CD-concentrations where self-association
significant reduction in the surface tensions de- of CD-molecules can be disregarded,33 a ‘‘simple
tected in their aqueous solutions. In the polymer- sorption’’ mechanism similar to the development
containing ternary systems, no further decrease of of surfactant-polymer associates can be assumed
surface tensions was measured. in these systems as well, which may lead to the
formation of ternary CD–statin–polymer associ-
ates of supramolecular structure.
A potential benefit in using macromolecular
colloids can also involve a significant improve-
ment of the in vivo bioavailability of these drugs.
Moreover, such ternary assemblies appear to be
better adsorbed to biological membranes, thus
ensuring an enhanced drug delivery and bioavail-
ability.30

CONCLUSIONS

The results of our phase-solubility studies suggest


Figure 10. Surface activity of cyclodextrins and CD– that in aqueous media, a host-guest type inclusion
statin complexes, T ¼ 258C, pH ffi 6.5. complexation of lovastatin and simvastatin with

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009 DOI 10.1002/jps
ENHANCEMENT OF DRUG SOLUBILITY 493

several cyclodextrins takes place. Dominantly self-microemulsifying drug delivery systems


CD–statin associates of 1:1 molar ratios form, (SMEDDS) for oral bioavailability enhancement
development of 1:2 complexes seems to be negli- of simvastatin in beagle dogs. Int J Pharm 274:
gible. More stable complexes are formed with the 65–73.
randomly methylated b-cyclodextrin than with 5. Grundy SM. 1988. HMG-CoA reductase inhibitors
for treatment of hypercholesterolemia. N Engl J
the native b-CD. Complexation leads to definite
Med 319:24–33.
increase in the aqueous solubility of these phar-
6. Tobert JA. 1988. Efficacy and long-term adverse
macons. In distilled water and at moderate effect pattern of lovastatin. Am J Cardiol 62:28J–
pH values, improvement of drug solubilities by 34J.
1–2 orders of magnitude could be observed. 7. Bradford RH, Shear CL, Chremos AN, Dujovne CA,
RAMEB showed outstanding solubilizing effect Franklin FA, Grillo RB, Higgins J, Langendorfer A,
for both statins. Also, the dissolved CD–statin Nash DT, Pool JL. 1994. Expanded Clinical Evalua-
complexes exhibited considerable surface activity, tion of Lovastatin (EXCEL) study results: Two-year
especially those of the RAMEB. efficacy and safety follow-up. Am J Cardiol 74:667–
By adding a macromolecular colloid to the 673.
binary CD–statin systems, the drug solubilities, 8. Haria M, McTavish D. 1997. Pravastatin: A re-
appraisal of its pharmacological properties and
after a certain temperature treatment, could
clinical effectiveness in the management of coron-
be further improved. The enhancement of drug
ary heart disease. Drugs 53:299–336.
solubilities can be attributed to the development 9. Hamelin BA, Turgeon J. 1998. Hydrophilicty/lipo-
of CD–statin–polymer associates of supramolecu- philicity: Relevance for the pharma-cology and clin-
lar structure. In these ternary complexes a portion ical effects of HMG-CoA reductase inhibitors.
of the surface-active solutes are attached to the Trends Pharmacol Sci 19:26–36.
macromolecular chains. The enhanced drug com- 10. Kearney AS, Crawford LF, Mehta SC, Radebaugh
plexation due to the polymeric additive could GW. 1993. The interconversion kinetics, equili-
possibly result in improved dissolution of statins brium, and solubilities of the lactone and hydro-
in the gastrointestinal tract. Inclusion complexa- xyacid forms of the HMG-CoA Reductase inhibitor,
tion might level off interindividual differences in CI-981. Pharm Res 10:1461–1465.
11. Brewster ME, Loftsson T. 2007. Cyclodextrins as
bioavailability of these pharmacons, preserve the
pharmaceutical solubilizers. Adv Drug Deliv Rev
liver preference of such lipophilic compounds9 and
59:645–666.
also, may protect against some types of food 12. Loftsson T, Brewster M. 1996. Pharmaceutical
interactions. Cyclodextrin-based ‘‘nanoencapsula- applications of cyclodextrins. I. Drug solubiliza-
tion’’ of statins might, therefore, offer benefits for tion and stabilization. J Pharm Sci 85:1017–
the standard antihiperlipidemic therapy. 1025.
13. Liu L, Guo QX. 2002. The driving forces in the
inclusion complexation of cyclodextrins. J Incl
ACKNOWLEDGMENTS Phenom Macro 42:1–14.
14. Szejtli J. 1982. Cyclodextrins and their inclusion
The authors are highly indebted to Dr. Á. Buvári- complexes. Budapest, Hungary: Akadémiai Kiadó.
Barcza and late Prof. L. Barcza for their valuable 15. Prentis RA, Lis Y, Walker SR. 1988. Pharmaceu-
suggestions and useful discussions. tical innovation by seven UK-owned pharmaceuti-
cal companies (1964–1985). Br J Clin Pharmacol
25:387–396.
16. Loftsson T, Friðriksdóttir H, Sigurðardóttir AM,
REFERENCES Ueda H. 1994. The effect of watersoluble polymers
on drug-cyclodextrin complexation. Int J Pharm
1. Istvan ES, Deisenhofer J. 2001. Structural mech- 110:69–177.
anism for statin inhibition of HMG-CoA reductase. 17. Higuchi T, Connors KA. 1965. Phase-solubility
Science 292:1160–1164. techniques. Adv Anal Chem Instrum 4:117.
2. Roberts WC. 1997. The rule of 5 and the rule of 7 in 18. European Directorate for the Quality of Medicines
lipid-lowering by statin drugs. Am J Cardiol 80: & HealthCare: European Pharmacopeia 5th
106–107. Edition 5.8 Supplement.
3. Maron DJ, Fazio S, Linton MF. 2000. Current 19. Validation of analytical procedures: methodology
perspectives on statins. Circulation 101:207–213. (CPMP/ICH/281/95).
4. Kang BK, Lee JS, Chon SK, Jeong SY, Yuk SH, 20. Országos Gyógyszerészeti Intézet-Gyógyszerkö-
Khang G, Lee HB, Cho SH. 2004. Development of nyvi Osztály—VIII. Magyar Gyógyszerkönyv

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009
494 SÜLE, SZENTE, AND CSEMPESZ

(Pharmacopoeia Hungarica VIII.) I./237–239, 27. Junquera E, Aicart E. 1997. Potentiometric study of
II./839. the encapsulation of ketoprophen by hydroxypro-
21. Loftsson T, Masson M. 2004. The effects of water- pyl-beta-cyclodextrin. Temperature, solvent and
soluble polymers on cyclodextrins and cyclodextrin salt effects. J Phys Chem B 101:7163–7171.
solubilization of drugs. J Drug Deliv Sci Tec 14:35– 28. Marcin C, White R, Hirsch C, Ferris F, Sykes R,
343. Houck D, Greasham R, Chartrain M. 1991. Biocon-
22. Buvári-Barcza Á, Barcza L. 2000. Changes in the version of the sodium salt of Simvastatin (MK-733)
solubility of b-cyclodextrin on complex formation: to 6-desmethyl-6-a-hydroxymethyl Simvastatin.
Guest enforced solubility of b-cyclodextrin inclusion J Ind Microbiol Biotechnol 8:157–164.
complexes. J Incl Phenom Macro 36:355–380. 29. Iga K, Hussain A, Kashihara T. 1981. New direct
23. Puskás I, Barcza L, Szente L, Csempesz F. 2006. calculation of K1: 1 and K1: 2 complexation con-
Features of the interaction between cyclodextrins stants using solubility method. J Pharm Sci 70:108–
and colloidal liposomes. J Incl Phenom Macro 109.
54:89–93. 30. Loftsson T, Stefánsson E, Friðriksdóttir H,
24. Redenti E, Szente L, Szejtli J. 2000. Drug/cyclodex- Kristinsson JK. 1996. Novel CD-based Drug
trin/hydroxy acid multicomponent systems. Proper- Delivery System. Proceedings of the Eighth Inter-
ties and pharmaceutical applications. J Pharm Sci national Symposium on Cyclodextrons. p 407–
89:1–8. 412.
25. Süle A, Csempesz F. 2005. Hatóanyag-oldékonyság 31. Attwood F, Florence AT. 1983. Surfactant systems.
szabályozása ciklodextrinekkel és kolloidokkal. Their chemistry, pharmacy and biology. London,
Abst. Semmelweis Egyetem PhD Tudományos UK: Chapman and Hall. pp 361–365.
Napok, Budapest, Hungary, p 64. 32. Myers D. 1988. Surfactant science and technology.
26. Zia V, Rajewski RA, Stella VJ. 2000. Thermody- New York, USA: VCH Publishers. pp 142–145.
namics of binding of neutral molecules to sulfobutyl 33. Loftsson T, Másson M, Brewster ME. 2004. Self-
ether b-Cyclodextrins (SBE-b-CDs): The effect of association of cyclodextrins and cyclodextrin com-
total degree of substitution. Pharm Res 17:936–941. plexes. J Pharm Sci 93:1091–1099.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 98, NO. 2, FEBRUARY 2009 DOI 10.1002/jps

Você também pode gostar