You are on page 1of 12

Journal of Drug Targeting, 2010; 18(9): 645–656

REVIEW ARTICLE

Cyclodextrins for drug delivery


A.L. Laza-Knoerr, R. Gref, and P. Couvreur
UMR CNRS 8612, Faculté de Pharmacie, Univ. Paris-Sud, 5 rue JB Clément, 92296 Châtenay-Malabry, France
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

Abstract
Cyclodextrins (CDs) are macrocyclic oligosaccharides composed of α(1,4)-linked glucopyranose subunits.
These molecules possess a cage-like supramolecular structure, comparable with the structures of crown
ethers, cryptands, spherands, cyclophanes, or calixarenes. However, it took 50 years to establish the molecu-
lar structure of CDs. Owing to their capability to form inclusion complexes with a variety of guest molecules,
CDs are considered as the most important supramolecular host family among all supramolecular structures
mentioned above. They can form complexes with various types of molecules including inorganic, organic,
or organometallic that can be radical, cationic, anionic, or neutral molecules. This phenomenon bears the
name “molecular recognition,” while the selectivity in the formation of complexes with enantiomeric species
as guests is called “chiral recognition.” In addition, the properties of the molecules forming the complexes
with CDs can be modified significantly. As such, a large number of scientists have attempted to elaborate
and evaluate various CD derivatives that are able to complex a variety of drugs, enhancing by this way their
in vivo solubility and activity. Moreover, a large number of publications describe CD uses in other fields such
For personal use only.

as foods, textile, cosmetics, or agriculture. This review reports on the recent developments of CDs in drug
delivery using various routes of administration.
Keywords:  Cyclodextrin; drug delivery; inclusion complexes; targeting; toxicology

Introduction and history complexes of CDs were determined and in 1961, the nat-
ural existence of δ-, ζ-, ξ-, and even η-CD was reported
The cyclodextrins (CDs) have been known for over 100 (Valle, 2004). The first patent in CDs was reported in 1953
years. They were first discovered in 1891 by Villiers, a (Freundenberg, Cramer, & Plieninger, 1953) and the first
French scientist (Villiers, 1891). He isolated about 3 g fundamental review on CDs and derivatives was pub-
of crystalline substance from 1000 g starch and deter- lished by French (1957).
mined its composition to be (C6H10O5)2·3H2O. Villiers has Until the end of the 1960s, the CD structure, physical
named this product “cellulosine” because the substance and chemical properties, methods of preparation, and
appeared to be resistant at acid hydrolysis and did not corresponding inclusion complexes have been investi-
have reducing properties. gated in detail by many authors (Higuchi and Connors,
In this so-called discovery period (Szejtli, 1998), a few 1965; Cramer, Saenger, & Satz, 1967).
years later (1903), Franz Schardinger published an article Summarizing the literature until the end of 1960s, CDs
(Schardinger, 1903) suggesting that “crystalline dextrin” were considered to be promising molecules, particularly
would be a better name than “cellulosine” to identify because of their industrial potential, but they remained
these molecules. Later, he changed the name “crystalline as expensive substances (fabrication in small quantities)
dextrin” into “α- and β-CD.” However, in the followings and their utilization in products for human use question-
years, CDs were named “Schardinger CDs” in his honor. able because of unknown potential toxic effect.
In the second stage (from 1930s to the 1970s), the Following the last period from the 1970s until present,
structures of α- and β-CDs were first determined by after adequate toxicological studies, CDs are today used
X-ray diffraction in 1942, followed by the γ-CD structure as successful “new” pharmacological excipients. From
in 1948. At the same time, structures of some inclusion 1981, when the first symposium on CDs was organized,

Address for Correspondence:  UMR CNRS 8612, Faculté de Pharmacie, Univ. Paris-Sud, 5 rue JB Clément, 92296 Châtenay-Malabry, France. E-mail: patrick.
couvreur@u-psud.fr

(Received 13 October 2009; revised 08 January 2010; accepted 13 January 2010)

ISSN 1061-186X print/ISSN 1029-2330 online © 2010 Informa UK, Ltd.


DOI: 10.3109/10611861003622552 http://www.informahealthcare.com/drt
646   A.L. Laza-Knoerr, R. Gref, and P. Couvreur

the total number of CD-related publications increased hydrophobic cavity, CDs can form inclusion complexes
over 15,000 by the end of 1997 (Szejtli, 1998). Every year, with a wide range of hydrophobic molecules as guests
CDs are the subject of approximately 1000 additional (Figure 1A and B) (Ritter &Tabatabai, 2002). In aqueous
research articles and abstracts, with a large number in solution, the hydrophobic CD cavity is occupied by water
the pharmaceutical branch, and many CDs and deriva- molecules bounded by “weak forces” (energetically
tives are included in the Handbook of Pharmaceutical unfavored). Thus, water molecules can be substituted
Excipients. by hydrophobic molecules such as drugs (“guests”)
Considering the price of β-CD, while in 1970 1 kg with the formation of “host” (CD dissolved): “guest”
costed about 2000 euros, today it is only several euros (drug molecules) complexes (Szejtli, 1998). Owing to
(∼450 euros) per kilogram. Thus, a large number of CD the size of the internal cavity, one or two hydrophobic
derivatives are produced industrially and used in the guest molecule(s) can be entrapped by one, two, or even
manufacture of drug carriers, cosmetics, or in catalysis, three CDs (Szejtli, 1998; Songa et al., 2009).
agricultural, and food industries. The stability of the complex formed depends on
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

how the “guest” molecule fits into the hydrophobic


cavity (a whole molecule or part of it). The ratio of
Chemistry and properties host–guest complex is frequently 1:1, 2:1, 2:2, or 1:2.
The formed inclusion complexes can be isolated as
CDs are cyclic α-1,4-glucans composed of six to more stable crystalline substances. The equilibrium estab-
than 100 glucose units (Qi, Mokhtar, & Zimmermann, lished between dissociated and associated species can
2007). They are natural products resulting from intramo- be characterized by the complex stability constant, Ka
lecular transglycosylation reaction of starch degraded by (Szejtli, 1998).
CD glucanotransferase (CGTase) enzyme (Szejtli, 1998). In summary, many reviews and articles describe the
The enzymatic product is generally a mixture of CDs properties and structure of CDs and their complex forma-
including α-, β-, and γ-CDs consisting of six, seven, or tion with various drug molecules. A large number of CD
For personal use only.

eight glucose units, respectively (Figure 1 and Table 1) derivatives have been reported in drug delivery, yielding
(Szejtli, 1998; Zhaofeng et al., 2007). They are the most the improvement in bioavailability and the increase in
extensively studied and utilized products from the CD aqueous solubility and stability. Besides, CDs and CD
family. derivatives can also convert liquid drugs into microcrys-
X-ray investigations on the structure of CD have talline powders, prevent drug–drug and drug–excipient
shown that due to their primary (C6) and secondary interactions, reduce ocular and gastrointestinal irritation,
hydroxyls group (C2 and C3) as well as due to their or diminish bad taste and smell.

OH
OH
O
A OH O O
OH
O O OH
OH HO
O O O O
O HO OH O
O OH
HO HO OH HO
OHHO HO O
O O OH HO O O OH HO O
HO HO
OH HO OH HO HO
O OH OH OH OH
α-CD O O γ-CD
O β-CD
HO O HO O
OH HO OH HO OH HO
OH HO O
O O OH HO O O OH HO O
OHHO
HO HO HO
O OH HO OH
O O O HO OH
O O O
OH OH OH
HO O O O O
HO
O
HO
HO

B OH
H 6
4 5 O
H
HO H 1 H
3 2
OH
H O
7

Figure 1.  Chemical structures of α-, β-, and γ-CDs (A) (Zhaofeng et al., 2007) and (B).
Cyclodextrins for drug delivery   647

Table 1.  Characteristics/properties of CDs.


Type of cyclodextrin
Characteristics α-CD β-CD γ-CD
Number of glucopyranose units 6 7 8
Molecular weight (g/mol) 972 1135 1297
Solubility in water at 25°C (%, w/w) 145 18.5 233
[α]D 25°C 150 ± 5 162.5 ± 5 177.4 ± 5
Internal diameter (Å) 4.7–5.3 6.0–6.5 7.5–8.3
External diameter (Å) 14.6 15.4 17.5
Volume of the cavity (Å3) 174 262 427
Approx. cavity volume in 1 mol CD (mL) 104 157 256
Cristal forms (from water) Hexagonal plates Monoclinic parallelograms Quadratic prisms
Cristal water (wt %) 10.2 13.2–14.5 8.13–17.7
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

Diffusion constant at 40°C 3.443 3.224 3.000


Hydrolysis by A. oryzae α-amylase Negligible Slow Rapid
pK (by potentiometry) at 25°C 12,332 12,202 12,081

lowest water solubility. In α-CD molecule the hydrogen


bond belt is incomplete, because one glucopyranose
Hydrophobic cavity
unit is in a distorted position. As a result, of the six
6 (Å) possible bonds only four can be established. The γ-CD
molecule is a non-coplanar, flexible structure, the most
Hydrophilic soluble of three natural CDs (Szejtli, 1998).
7,9 (Å) outer shell A large number of CD derivatives have been synthe-
For personal use only.

sized: 2-hydroxypropyl-β-CD (2-HP-β-CD) (Gould &


Scott, 2005), methyl-β-CD (M-β-CD) (Uekama & Irie,
6,5 (Å) 1987), or sulfated amphiphilic α-, β-, and γ-CDs (Dubesa
et  al., 2003) principally with the aim to improve the
15,4 (Å) apparent solubility or the biological activity of various
active molecules.
Figure 2.  Structure of β-CD. For example, the introduction of sulfate groups onto
the hydroxyl groups of CDs confers them a higher aque-
Compared with α-CDs and β-CDs, γ-CD with a large ous solubility and a wide range of biological activities
internal cavity and high solubility exhibits more favorable such as antiviral, anti-lipemic, anti-angiogenic, or anti-
properties. Therefore, many groups of scientists are inter- inflammatory properties (Dubesa et al., 2003).
ested in γ-CD (Takada et  al., 2003; Hirano et  al., 2005; Amphiphilic CDs were also synthesized to increase
Jansook & Loftsson, 2008) and many attempts have been the contact of CDs (having high external hydrophilicity)
made to modify the properties of CGTase to increase with lipidic biological membranes in vivo (Memisoglu-
the production of γ-CD (Biwer, Antranikian, & Heinzle, Bilensoy et al., 2006). Depending on their structure, the
2002). amphiphilic CDs have been named: “skirt”-CDs (Zhang
The solubility of β-CD in water was determined to et  al., 1991), “lollipop”-CDs (Bellanger & Perly, 1992),
be 18.6 mg/mL (Loftsson & Frioriksdottir, 1998). Such “bouquet”-CDs (Canceill et  al., 1992), and “cup-and-
relatively low water solubility limits β-CD applications ball”-CDs (Lin, 1995).
since the complex formation with lipophilic compounds Recently, attention has been focused on the synthesis
frequently results in the precipitation of solid cyclodex- and usage of CD polymers. Regarding the synthesis of
trin complexes. The aqueous solubility of the natural this type of water-soluble polymer, the general method
CDs is often much lower than the comparable linear or involves effective cross-linking agents like epichlorohy-
branched dextrins. drin (Renard et  al., 1997), diisocyanates (Cadars et  al.,
The C-2-OH group of one glucopyranoside unit can 2005), polycarboxylic acids (Martel et  al., 2005; Zhao
form a hydrogen bond with the C-3-OH group of the et al., 2009), or anhydrides (Gireka et al., 2005).
adjacent glucopyranose unit. In the CD molecule, a com- Epichlorohydrin is the most popular cross-linking
plete secondary belt is formed by these H bonds; there- agent used in chemistry and industry for the synthesis
fore, the β-CD has a rather rigid structure (Figure 2). of β-CD polymers. A large quantity of epichlorohydrin
This intramolecular hydrogen bond formation is the might be wasted in the reaction, because the reaction
explanation for the observation that β-CD has the between β-CD and epichlorohydrin requires a high
648   A.L. Laza-Knoerr, R. Gref, and P. Couvreur

concentration of sodium hydroxide in order to activate lower toxicity. For example, 2-HP-β-CD is well tolerated
the hydroxyls of CD and to accelerate the hydrolyzation in animal tests (rats, mice, and dogs) particularly by oral
of epichlorohydrin (Renard et al., 1997). route (Gould & Scott, 2005). Furthermore, after intrave-
Unfortunately, most of the cross-linking agents nous administration (mice, monkey, rats, dogs, or rab-
(epichlorohydrin, diisocyanates, anhydrides) utilized bits) 2-HP-β-CD revealed neither lethal nor carcinogenic
in the synthesis of poly-CDs have a potential toxic effect effects and did not show any consequence on embryo-
for human health and environment. Comparatively, citric fetal development (rats or rabbits) (Gould & Scott, 2005).
acid features a lower toxicity and is environment friendly. Human experience with CD derivatives, especially SBE
The condensation between β-CD and citric acid has β-CD (Captisol), a polyanionic variably substituted
been carried out at a temperature not higher than 170°C sulfobutyl ether of β-CD and 2-HP-β-CD, indicates that
and without any organic solvent (Martel et  al., 2005). these two CDs are well tolerated in humans and have no
However, the drawback of this method lies in the weak adverse effects on the kidneys or other organs following
reactivity of citric acid compared with epichlorohydrin either oral or intravenous administration (Stella & He,
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

or diisocyanates. 2008).
CDs polymers in association with other polymers
constitute another area of current interest. For example,
promising results have been obtained by mixing hydro- Drug delivery
phobically modified dextrans bearing dodecyl moieties
with β-CD polymer. Owing to the inclusion of hydropho- Owing to the remarkable ability of CDs to form inclusion
bic side chains of dextran into CD cavities, spontaneous complexes with a variety of drug molecules, numerous
association occurs in solution (Daoud-Mahammed applications have been developed in the drug delivery
et al., 2007a, 2007b; Gref et al., 2006). Encapsulation of field using almost all routes of administration.
lipophilic drugs such as benzophenone and tamoxifen
in this type of system was achieved by their inclusion
Nasal administration
For personal use only.

(complexation) in the remaining free cavities of CDs.


Application of CD solution directly onto the mucosa
(nasal, buccal) is regarded as safe for hydrophilic and
Toxicological consideration natural CDs in a wide range of concentrations while for
methylated CD derivatives, the application time and the
The toxicity profiles of CDs first depend on the route of concentration should be controlled (Asai et  al., 2002;
administration used. After oral administration of CD, Boulmedarat et al., 2005).
a partial or complete hydrolysis of CDs was observed. In rats, the nasal absorption of insulin has been
When used by the oral route, CDs can be hydrolyzed by notably improved by the use of CDs (Iriea et al., 1992).
amylases from the digestive tract, similarly like starch It was reported that methylated CDs were more potent
(Gerlóczy et al., 1985). γ-CD with a large internal cavity is enhancers of insulin adsorption than the parent CDs or
more easily hydrolyzed than α-CD which exhibits a small 2-HP-β-CD. On the other hand, Asai et al. (2002) demon-
internal cavity whereas β-CD is in between. Whether the strated that 20% RM-β-CD (RM-randomly methylated)
hydrolysis is partial or total, CDs are non-toxic when causes serious damage to the rat nasal mucosa and 10%
administered through oral route. CDs can form inclusion solution causes irritation. In the same topic, Boulmedarat
complexes with other compounds present in the diges- et al. (2005) indicated that the application of 10% solu-
tive tract (such as biliary salts) in a reversible manner tion of RM-β-CD on epithelial cells presented toxic and
(Duchene, Bochot, & Loftsson, 2009). inflammatory effects, depending on the exposure time.
It is reported that when administered in high doses in Nasal preparations must, however, be critically evalu-
animals by subcutaneous route, β-CD might cause neph- ated for their possible effects on the mucociliary func-
rotoxicity, decrease in body weight, and decrease in liver tions (defence of the respiratory tract against bacteria,
weight (Perrin et al., 1978). Intravenous administration allergens, etc.). In this view, on the contrary to steroids
was shown to induce similar effects, due to the compl- free, the CD complexes did not present any effects on the
exation of some blood constituents, such as cholesterol. epithelial membranes probably because their cavities are
By parental route, and especially the intravenous one, shielding the intrinsic toxicity of the included molecules
β-CDs can destabilize the red blood cell membrane by (Koizumi et al., 1987). Also, a nasal spray containing estra-
forming inclusion complexes with some of their constitu- diol solubilized in 2,6-di-O-methyl-β-CD (DM-β-CD) was
ents, leading to hemolysis (Duchene, Bochot, & Loftsson, found very well tolerated by patients over a period of 6
2009). months (Hermes et al., 1992). In another study, Kondo,
However, some derivatives of CDs can be used as Irie, and Uekama (1996a) have demonstrated that a
alternatives to the parent α-, β-, or γ-CDs because of their proper use of CDs in nasal morphine preparations may
Cyclodextrins for drug delivery   649

provide with adequate analgesia for chronic and acute Table 2.  Some examples of the use of CDs in ocular delivery.
pain, offering a progress in patient comfort. CD derivatives
In the same topic, a nasal spray containing benzodi- (abbreviation) Drugs References
azepines solubilized in CD was reported by Loftsson et al. HP-β-CD Acetazolamide (Fridriksdottir, Loftsson, &
(2001). It was possible to minimize the amount of CD Stefánsson, 1997; Granero
et al., 2008; Palma et al., 2009
needed to solubilize the benzodiazepine drug by keep-
Hydrocortisone Davies, Wang, & Tucker, 1997
ing the drug/CD molar ratio close to one or by adding a
Dipiverfrin Jarho et al., 1997
water-soluble polymer to the drug/CD complex.
SBE-β-CD Dipiverfrin Jarho et al., 1997
Recently, Al Omaria et  al. (2009) described a novel Pilocaprine Järvinen et al., 1994
inclusion complex of ibuprofen (Ibu) with native CDs
and 2HP-β-CD. Without CD, Ibu exhibits an irritant effect
on the oral cavity, throat, and pharynx and this makes its For example, the increase in water solubility and effi-
administration unpleasant. However, its complexation cacy of acetazolamide, a carbonic anhydrase inhibitor,
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

with natural or modified CDs helped circumventing these was observed in aqueous solutions of 2-HP-β-CD by the
inconveniences. formation of acetazolamide:2-HP-β-CD complex. This
In conclusion, CDs may improve the drug bioavail- derivative of CD was found to be non-toxic toward the
ability and help bypassing the nasal barrier. ocular tissue (because it was unable to penetrate biologi-
cal membranes such as the eye cornea) and its aqueous
formulation was well tolerated (Fridriksdottir, Loftsson,
Ocular drug delivery & Stefánsson, 1997; Granero et  al., 2008; Palma et  al.,
In the ophthalmology field, drug formulations are gener- 2009).
ally applied to the surface of the eye for two purposes: (1) Promising results were also obtained by mixing 2-HP-
to provide intraocular treatment throughout the cornea β-CD with hydrocortisone (HC) (Davies, Wang, & Tucker,
for diseases like glaucoma and (2) to treat the outside of 1997) to formulate a stable topical ophthalmic solution. A
For personal use only.

the eye for infections (conjunctivitis, blepharitis, keratitis soluble 1:1 inclusion complex was formed with enhanced
sicca). Eye-drops are the conventional drug dosage forms chemical stability and water solubility.
for 90% of currently used ophthalmic formulations. A CD derivatives were successfully used to increase
major problem in ophthalmic drug delivery is, however, the aqueous stability of dipivefrin too (dipivalyl epine-
the rapid drug loss caused by drainage and high tear phrine, DPE) (Jarho et  al., 1997). The effect of 2-HP-
fluid. Drug has to penetrate into the eye through con- β-CD or sulfobutyl ether β-cyclodextrin (SBE-β-CD) in
junctiva, cornea, and/or sclera membranes (lipophilic the presence of DPE was studied with isolated rabbit
membranes). These membranes are surrounded by an cornea. The authors reported the formation of strong
aqueous tear fluid and by a mucin layer. Thus, ophthal- inclusion complexes (SBE-β-CD/DPE and HP-β-CD/
mic drugs have to be water soluble (hydrophilic) to pen- DPE) together with a dramatic increase in the aqueous
etrate the exterior surface of the eye (tear fluid and mucin stability of DPE (at room temperature and at pH values
layer), but at the same time they have to display sufficient of 5 and 7.4).
lipophilicity to penetrate the ocular barrier into the eye. Järvinen et  al. (1994) reported the complexation of
As such, after ophthalmic application of drug formula- a large number of β-CD with pilocarpine ophthalmic
tions, only less than 5% can penetrate the cornea and the drug. The authors highlighted that the aqueous stabil-
intraocular tissues (Lang, 1995). ity of pilocarpine increased at pH 7 in the presence of
In this context, CDs may represent an alternative SBE-β-CD. Further, the same author (Järvinen, Järvinen,
approach to increase the solubility and corneal perme- & Urtti, 1995) showed that SBE-β-CD solution improved
ability of drugs. ocular absorption by adding poly(vinyl-alcohol) (PVA)
Indeed, the inclusion complexes of ophthalmic drugs which increased the viscosity and also reduced eye irrita-
with CDs were found to increase the drug aqueous solu- tion. Consequently, CDs may be useful in the formulation
bility without affecting their chemical structure and/or of ophthalmic suspension by complexation with a large
activity (Loftsson & Järvinen, 1999; Loftsson & Stefánsson, number of ophthalmic drugs.
2002; Brewster & Loftsson, 2007; Alexanian et al., 2008;
Gaspar de Araújo et al., 2008; Granero et al., 2008; Araújo
Oral drug delivery
et  al., 2009; Nagarwala et  al., 2009; Palma et  al., 2009).
Thus, CDs were found to be capable of delivering the At present, oral administration of inclusion complexes
entrapped hydrophobic drugs directly to the ocular sur- of CDs with many poorly water-soluble drugs is possible
face, after the passage of the mucin layer. Some examples because of the natural α- and β-CDs, unlike γ-CD (very
are summarized in Table 2. large cavity) are not hydrolyzed by human saliva.
650   A.L. Laza-Knoerr, R. Gref, and P. Couvreur

CDs (α-CD, β-CD, γ-CD) and their derivatives • The formation of a “host–guest” complex CD mol-
[sulfated-α-CD, sulfated-β-CD, carboxymethyl-β-CD ecule (with bad tasting), impeding its reaction with
(CM-β-CD), and 2-HP-β-CD] have been utilized in oral the taste buds.
hygiene products to bind large sized malodorous com- • The interaction of the CDs with the so-called gate-
pounds residing in the mouth (organic acids, amines, keeper proteins localized in the taste bud paralyzing
amino acids) (Lantz et al., 2006). The authors highlighted them.
the high affinity of lactic and succinic acids for CM-β-CD
and a slightly higher binding to sulfated-β-CD than to The bitter taste of coffee results from overcooking,
native β-CD (Table 3). The explanation lies in the pres- extended standing in either cold or hot state, which are
ence of carboxylic groups in the structure of both lactic effectively reduced by mixing 0.1 until 10.2% (w/v) β-CD.
and succinic acids which interact with hydrogen bond CDs form complexes with polyphenols and chlorogenic
donor/acceptor sites in carboxylate and sulfate groups acid, preventing by this way the bad sensation in the
attached to both CD derivatives (CM-β-CD, sulfated- mouth (Hamilton & Healy, 1970).
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

β-CD). An important tool in this regard is the use of CDs and


Very important characteristics of CDs and deriva- their derivatives via dynamic complex formation so that
tives for oral delivery purposes stems from their ability the complexes overcome undesirable physicochemical
to modify the bioavailability of drugs by increasing the properties including low aqueous solubility, poor dis-
time period of drug release and the drug dissolution rate solution rate, and limited drug stability. CDs have been
(Loftsson, Matthíasson, & Másson, 2003) whereas at the utilized extensively in pharmaceutical formulations to
same time CDs may decrease local GI tract irritation or enhance drugs’ bioavailability (Loftsson et  al., 2005;
bitter taste of drugs (Sjejtli & Szente, 2005). Following Carrier, Miller, & Ahmed, 2007; Mallick et  al., 2007).
interactions are possible in order to eliminate the bitter In this topic, many recent reviews deal with the use of
taste: CDs as excipients for the oral delivery of various drugs
(Soares, Carvalho, & Veiga, 2007; Stella & He, 2008;
For personal use only.

Table 3.  Some examples of the use of CDs in oral delivery. Shimpi, Chauhan, & Shimpi, 2004; Stickley, 1994; Fahr
CD derivatives & Liu, 2007; Challa et al., 2004). Thus, the usefulness of
(abbreviation) Active molecules References CDs has been demonstrated in the case of various drugs
α-CD Peptides Haeberlin et al., 1996 such as cilostazol (Patel & Rajput, 2009), piroxicam
CM-β-CD Lactic acid and Lantz et al., 2006 (Lister, Acerbi, & Cadel, 1993), flurbiprofen (Tokumura,
succinic acid Muraoka, & Machida, 2009), paclitaxel (Agüeros et al.,
β-CD Nicotine Carlsson & Andersson, 2009), andragrapholide (Ren et  al., 2009), gefitinib
1991
(Phillip Lee et  al., 2009), various peptides (Haeberlin
Polyphenols and Hamilton & Healy, 1970
chlorogenic acid
et al., 1996), efavirenz (Sathigari et al., 2009), diclofenac
Peptides Haeberlin et al., 1996 (Miro et  al., 2009), flavonoid dioclein (Rezende et  al.,
Flavonoid dioclein Rezende et al., 2009 2009), triclosan (Dinge & Nagarsenker, 2008), and many
Cilostazol Patel & Rajput, 2009 others.
Flurbiprofen Tokumura, Muraoka, &
Machida, 2009
Gefitinib Phillip Lee et al., 2009 Dermal drug delivery
Piroxicam Lister, Acerbi, & Cadel,
Dermal drug delivery has gained popularity in the past
1993
Efavirenz Sathigari et al., 2009
decade. Many drugs have been successfully formulated
γ- CD Peptides Haeberlin et al., 1996
by this route. The transdermal drug transport and release
Cilostazol Patel & Rajput, 2009 is restricted by the stratum corneum which represents
HP-β-CD Peptides Haeberlin et al., 1996 the main barrier toward the administration of active
Triclosan Dinge & Nagarsenker, compounds by this route. Therefore, many methods to
2008 increase drug absorption have been tested.
Cilostazol Patel & Rajput, 2009 Four different approaches have been proposed to
Diclofenac Miro et al., 2009 enhance drug delivery through the skin (Matsuda &
Efavirenz Sathigari et al., 2009 Arima, 1999):
Paclitaxel Agüeros et al., 2009
Andragrapholide Ren et al., 2009 (1) Enhancement in the release of drugs from the
Gefitinib Phillip Lee et al., 2009 transdermal pharmaceutical preparation bases.
DM-β-CD Peptides Haeberlin et al., 1996 (2) Improvement of drug retention in the skin or of the
Cilostazol Patel & Rajput, 2009 flux of drug through the skin.
Cyclodextrins for drug delivery   651

(3) Increase in the tissue targeting (localized drug HP-β-CD enhanced the dermal delivery into the skin
delivery). (proved by the increase of their stability constants). PVP
(4) A combination of (1), (2), and (3). acted as a co-enhancer. More recently, Lopez et al. (2000)
reported a protective effect of 2-HP-β-CD ascribed to an
In order to meet these criteria, many scientists have uti- increase in the solubility of dexamethasone without the
lized surfactants, liposomes, permeation enhancers, ion need of any tertiary agent.
pairs, etc. Furthermore, the combined effects of CD complexa-
In the past decade, various kinds of chemically modi- tion and entrapment into liposomes was investigated
fied CDs too have been prepared in order to optimize and highlighted for the first time by McCormack and
the dermal delivery of drugs (both for systemic and local Gregoriadis (1994). This method revealed an increased
use). drug solubility and stability as well as a better controlled
Some representative examples are summarized in in vivo release.
Table 4. In general, CD derivatives improve the solubil- For example, Maestrellia et  al. (2006a) developed
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

ity and stability of hydrophilic drugs in dermal aqueous by this way an efficient formulation of ketoprofen.
solution preparations as well as in other type of dermal Ketoprofen is an analgesic used for the treatment of
preparations (ointments). osteoarthritis but it presents a low water solubility. When
For example, Kawahara et al. (1992) reported that the administered by the oral route, it presents a gastrointesti-
release rates of indomethacin from gel ointment and nal irritation. For this reason, transdermal administration
hydrophilic petrolatum were accelerated by complexa- has been used as an interesting alternative.
tion with diethyl-β-CD. In rats, the in vivo absorption of Permeability studies across artificial membranes sim-
indomethacin from gel ointment was also accelerated by ulating the skin behavior with the complex ketoprofen–
complexation with diethyl-β-CD. 2-HP-β-CD incorporated in liposome have showed a
Siguroardottir and Loftsson (1995) reported the effect prolonged release effect. The entrapment efficiency of
of the addition of a polymer (tertiary agent) to a solution ketoprofen–2-HP-β-CD in liposomes has been found
For personal use only.

containing a corticoid and CDs on the delivery of the to depend on both liposome preparation and complex
drug through hairless female mice skin. The results sug- concentration in the aqueous phase used for liposome
gested that the addition of a small quantity of hydroxy- preparation.
propylmethylcellulose (HPMC) or polyvinylpyrrolidone Regarding the CD polymers, Szeman et al. (1987) high-
(PVP) to the suspension containing dexamethasone–2- lighted that the plasma concentrations of the antifungal
agent tolnaftate was increased by complexation with the
β-CD polymer via dermal delivery. To evaluate the percu-
Table 4.  Some examples of the use of CDs in dermal delivery.
CD derivatives
taneous absorption of this formulation, the experiences
(abbreviation) Drugs References were conduced by topical application of the drug powder
α-CD Miconasole Tenjarla et al., to the skin of mice.
1998 Some types of CDs (parent and derivatives) may
β-CD Betamethasone Otagiri et al., affect the permeability of drugs through the skin via
1984 the interactions with some components of skin (choles-
4-Biphenylacetic Arima et al., 1996 terol or proteins). Thus, the effects of β-CD, randomly
acid
methylated β-CD (RAMEB) and 2-HP-β-CD on the skin
Chloramphenicol Rahman et al.,
1991
have already been investigated (Legendre et  al., 1995).
DM-β-CD Butylparaben, Okamoto et al.,
RAMEB was reported to decrease the transdermal flux
indomethacin, sulfanilic 1986 of piribedil due to the formation of an inclusion complex
acid RAMEB:piribedil as shown by RMN. On the other hand,
4-Biphenylacetic acid Arima et al., 1996 the penetration of S-9977 hydrochloride was increased
M-β-CD, CM-β-CD, Hydrocortisone Siguroardottir & twofold with the addition of RAMEB, in spite of the fact
maltosyl-β-CD Loftsson, 1995 that this drug did not interact with this CD derivative.
HP-β-CD Dexamethasone Lopez F.P. Renata Therefore, inclusion of skin components into CD have
et al., 2000
to be taken in consideration since it may influence skin
Ketoprofen Maestrellia et al.,
2006
penetration of the drugs in various ways.
Miconasole Tenjarla et al., Among the three derivatives of CDs (β-CD, RAMEB,
1998 and 2-HP-β-CD), only RAMEB extracted significant
β-CD polymer Tolnaftate Szeman, Ueda, & amounts of the major lipid classes from isolated stratum
Szejtli, 1987 corneum (SC). In conclusion, some β-CDs (parent and
Indomethacin Kawahara et al., derivatives) are capable of modifying the skin barrier
1992 function. As long as the drug molecules do not form an
652   A.L. Laza-Knoerr, R. Gref, and P. Couvreur

inclusion complex with CDs, this results in an enhance- Table 5.  Some examples of the use of cyclodextrin in rectal delivery.
ment of the drug diffusion through the skin. CD derivatives
Finally, with or without tertiary agent, CDs (parents (abbreviation) Drugs References
or derivatives) are good agents for the delivery of dermal α-CD Morphine Kondo, Irie, & Uekama,
drugs and due to their high molecular weights, they are hydrochloride 1996b
not absorbed through the skin. Cefmetazole Yanagi et al., 1991
β-CD Carmofur Kikuchi et al., 1987
γ-CD Diazepam Frijlink et al., 1992
Rectal drug delivery DM-β-CD Carmofur Masahikoa, Fumitoshia,
& Kaneto, 1987
Rectal drug delivery stands for a very important system
Insulin Watanabe et al., 1992
for a large number of patients, for example, children, Ethyl 4-biphenylyl Arima et al., 1992
infants, patients with nausea, swallowing, or vomiting. acetate
However, the main problems encountered in the admin- HP-β-CD Diazepam Frijlink et al., 1992
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

istration of drugs by rectal route are generally considered Ethyl 4-biphenylyl Arima et al., 1992
to be the following ones: acetate
Tanshinone IIA Ling et al., 2007
(1) the majority of drugs are poorly absorbed through the β-CD Carmofur Kaji et al., 1985
rectal mucosa,
(2) restrictive absorbing surface area, physicochemical properties of the formed inclusion
(3) inefficient dissolution due to the small fluid present complexes.
in the rectum,
(4) drug metabolism into the rectal mucosa and
Novel nanoparticulate drug delivery systems
microorganisms.
Many efforts have been directed to use CD-based mate-
For personal use only.

In order to overcome these disadvantages, many studies rials for the design of solid nanoparticles. In general,
have been conducted consisting in the use of absorption nanoparticles present a higher stability than liposomes
enhancers, mixed micelle and polymers, surfactants, in biological fluids, and a larger surface area than micro-
etc. particles due to their smaller size. Moreover, their high
However, the most important factor in rectal absorp- surface area can lead to intimate contact with biological
tion is the release and the stabilizing effects of drugs from membranes, giving way to the increase in the bioavail-
suppository bases, which inhibits the bioconversion of ability of the entrapped drugs.
drugs in the rectum. As such, many authors have shown Nanoparticles are generally prepared using preformed
that CDs (parents and derivatives) inhibit bioconversion polymers by emulsification methods or nanoprecipita-
of drugs in the rectum (Table 5). tion. Emulsion polymerization is another very frequently
However, the complexation of drugs with CDs (parents employed method, for example, using alkyl cyanoacr-
and derivatives) makes the drugs insoluble in oleagi- ylates as monomers. Indeed, cyanoacrylates offer the
nous suppository base. For example, Kondo, Irie, and advantage of being biodegradable as well as easy to pre-
Uekama (1996b) reported that the combination of α-CD pare, since the polymerization process occurs in aque-
and xanthan gum (polysaccharide polymer) reduced the ous medium without any polymerization initiator except
bioconversion of morphine (first pass metabolism) in water. Thus, CD-loaded nanoparticles were obtained
the rectal mucosa. Hence, it appeared that xanthan gum in acid conditions by anionic polymerization of alkyl
combined with α-CD exhibited a good stability of the cyanoacrylates in the presence of parent or modified
drug and facilitated the transport of morphine through CDs (Monza Da Silveira et al., 1999). Active compounds
the rectal mucosa. such as taxol and doxorubicin were entrapped in these
In comparison with parent CD (β-CD), DM-β-CD CD-based nanoparticles with loadings ranging between
significantly enhanced the rectal absorption of hydro- 0.01 and 300 mg/g of nanoparticles (Monza Da Silveira
phobic drugs like carmofur (1-hexylcarbamoyl-5-fluor- et al., 1999).
ouracil, HCFU) in oleaginous suppository (Masahikoa, Studies carried out with amphiphilic “skirt-shaped”
Fumitoshia, & Kaneto, 1987). A higher stability of CDs (obtained by the esterification of primary hydroxyl
DM-β-CD complexes compared with parent β-CD ones groups with C6 to C14 alkyl chains) showed that these
was demonstrated. derivatives were capable of forming spontaneous nano-
In conclusion, a large number of reports have dem- particles which could be loaded with a series of steroid
onstrated that the effects of CDs in rectal drug delivery drugs (progesterone, testosterone, hydrocortisone)
depends on the type of vehicle (hydrophilic or oleagi- (Duchene, Wouessidjewe, & Ponchel, 1999). In this type of
nous) on the presence of a tertiary element and on the nanoparticles, the drugs were most probably molecularly
Cyclodextrins for drug delivery   653

pCD MD & Alonso, 2007). The encapsulation yields were up to


dextran
β-CD
58.8% for insulin and 44.6% for heparin. The release pro-
file of insulin was shown to be very fast (84–97% within
molecule alkyl side chain
15 min), whereas heparin remained associated with the
of interest alkyle (C12)
nanoparticles for several hours.
Among these diverse assemblies, micelle-like core-
shell nanospheres mediated by host–guest interactions
are recognized as promising nanocarriers for drug and
gene delivery in a recent year (Zhang & Ma, 2009). In
this work, the author’s selected β-CD as a host unit for
the construction of a hydrophilic host block that is cova-
lently linked with another hydrophilic segment, while
loaded hydrophobic substances, either small molecules or
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

nanoparticles macromolecules, are employed as guest components.


Figure 3.  Schematic illustration of the interaction between poly-
Core-shell nanoassemblies can form by host–guest inter-
β-CD and modified dextran (MD) in the nanoassemblies (Gref et al., action mediated spontaneous assembly in an aqueous
2006). solution. For example, the core-shell aggregates could
be assembled by polyethylene glycol (PEG) block and
dispersed in the alkyl chain hydrophobic domains and a polyaspartamide block carrying β-CD units on the
were found to be released very rapidly. side chain b-PCD (PEG-b-PCD) (aqueous solution) in
Recently, a breakthrough CD-based nanotechnology the presence of hydrophobic compound, mediated by
has been developed by the single mixing (at room tem- host–guest interactions between CD and hydrophobic
perature) of two aqueous solutions of polymers consist- compound.
ing of β-CD polymer (poly-β-CD) and a hydrophobically Thus, CD-based nanoparticles offer an interesting
For personal use only.

modified alkyl chain dextran (MD) (Gref et al., 2006). In alternative to soluble CD (or poly-CD) for the encap-
this system, nanoparticle production yields could reach sulation and the delivery of hydrophobic drugs which
up to 95%. The authors further established that, in these are capable of forming molecular inclusion complexes
nanoassemblies, all alkyl chains were included within the with CDs in aqueous media. The protection of unstable
CD cavities. The ways to load drugs into those nanopar- compounds from degradation in biological media is an
ticles were either to fashion an inclusion complex with additional advantage of this type of technology.
poly-β-CD before mixing with MD or by direct loading
into the preformed nanoassemblies. Noteworthy, the
incorporation of benzophenone or tamoxifen in such Conclusions
nanocarriers did not hinder the sequestration of the
dextran alkyl moieties in β-CD cavities and thus did not This review outlines the current applications of natural
impede the formation of the corresponding associative and chemically modified CDs in the design of advanced
nanoassemblies (100–200 nm) (Figure 3). dosage forms. It clearly appears that the CDs have a wide
Nanoparticles were prepared by ionic gelation using range of applications in different areas of drug delivery
CD and chitosan (CS) mixtures (Maestrellia et al., 2006b). and pharmaceutical industry, due to their complexa-
Briefly, aqueous solutions containing different concen- tion ability with a large number of drugs. In general, the
trations of 2-HP-β-CD were incubated with CS for 24 h most common pharmaceutical applications of CDs are
under magnetic stirring and then filtered (cellulose fil- to enhance the solubility, stability, safety, and bioavail-
ters). The cross-linking agent tripolyphosphate (TPP) was ability of drug molecules.
added to this solution leading to the controlled gelation CDs interact with poorly water-soluble drugs resulting
of CS in the form of NPs. Finally, the NPs were isolated by in an increase in their apparent water solubility, phenom-
centrifugation and resuspended in water. enon based on the ability of CDs to form non-covalent
The higher solubilization as well as the protection of dynamic inclusion complexes in solution. Other solubi-
the sensitive drugs triclosan and furosemide facilitated lizing attributes include the ability of CDs to form non-
the oral absorption of these molecules. The drug content inclusion–based complexes and to form and stabilize
in these chitosan/CD nanoparticles was increased, in the supersaturated drug solutions. The increase in solubility
presence of 2-HP-β-CD due to their high solubilizing often also leads to an increase in the dissolution rates,
properties. thus improving the oral bioavailability of many drugs.
Another type of nanoparticle based on CS, tripolyphos- Moreover, a number of CD-based products have reached
phate and CM-β-CD were elaborated for the delivery of the market based on their ability to camouflage undesir-
macromolecular drugs (i.e., insulin or heparin) (Krauland able physicochemical properties of the drugs. Indeed,
654   A.L. Laza-Knoerr, R. Gref, and P. Couvreur

the bioadaptability and multifunctional characteristics of Canceill J, Jullien L, Lacombe L, Lehn J-M. (1992). Channel-type
molecular-structures. 2. Synthesis of bouquet-shaped mol-
CDs make them capable of alleviating several undesirable ecules based on a beta-cyclodextrin core. Helv Chim Acta, 75,
properties of the drug molecules through the formation 791–812.
of inclusion complexes. The drug:CD complexes can be Carlsson T, Andersson SB. (1991). Smoking substituted containing an
inclusion complex of nicotine and a cyclodextrin compound. In
further used in constructing new classes of drug delivery EP0506774; PCT INT., W.O. 9109599 (CA 115; 273283).
systems such as nanoparticles, liposomes, or site-specific Carrier RL, Miller LA, Ahmed I. (2007). The utility of cyclodextrins for
prodrugs. enhancing oral bioavailability. J Control Release, 123, 78–99.
Challa R, Ahuja A, Ali J, Khar RK. (2004). Cyclodextrins in drug deliv-
The CDs have extensively been used to formulate ery: an updated review. Curr Drug Deliv, 1, 351–360.
insoluble, volatile, or irritating drug molecules. Because Cramer F, Saenger W, Satz HC. (1967). Inclusion compounds. ΧΙХ.
of their continuing ability to find novel applications in The formation of inclusion compounds of alpha cyclodextrin in
aqueous solutions, thermodynamics and kinetics. J Am Chem
drug delivery, the CDs are expected to solve challenges Soc, 89, 14–20.
related to the delivery of novel drugs through different Daoud-Mahammed S, Couvreur P, Gref R. (2007a). Novel self-assem-
routes of administration. bling nanogels: stability and lyophilisation studies. Int J Pharm,
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

332, 185–191.
Daoud-Mahammed S, Ringard-Lefebvre C, Razzouq N, et al. (2007b).
Spontaneous association of hydrophobized dextran and poly-be-
Declaration of interest ta-cyclodextrin into nanoassemblies. Formation and interaction
with a hydrophobic drug. J Colloid Interface Sci, 307, 83–93.
Davies MN, Wang G, Tucker IG. (1997). Evaluation of hydrocortisone/
The authors report no conflicts of interest. The authors hydroxypropyl-beta-cyclodextrin solution for ocular delivery. Int
alone are responsible for the content and writing of the J Pharm, 156, 201–209.
Dinge A, Nagarsenker M. (2008). Formulation and evaluation of fast
paper. dissolving films for delivery of triclosan to the oral cavity. AAPS
PharmSciTech, 9, 349–356.
Dubesa A, Degobertb G, Fessib H, Parrot-Lopez H. (2003). Synthesis
and characterisation of sulfated amphiphilic α-, β- and
References γ-cyclodextrins: application to the complexation of acyclovir.
Carbohydr Res, 338, 2185–2193.
For personal use only.

Al Omaria MM, Daraghmeha NH, El-Barghouthib MI, et al. (2009). Duchene D, Bochot A, Loftsson T. (2009). Les cyclodextrines et leurs
Novel inclusion complex of ibuprofen tromethamine with cyclo- utilisations en pharmacie et cosmétologie. STP Pharma Pratiques,
dextrins: physico-chemical characterization. J Pharm Biomed 19, 15–27.
Anal, 50, 449–458. Duchene D, Wouessidjewe D, Ponchel G. (1999). Cyclodextrins and
Alexanian C, Papademou H, Vertzoni M, Archontaki H, Valsami G. carrier systems. J Control Release, 62, 263–268.
(2008). Effect of pH and water-soluble polymers on the aqueous Fahr A, Liu X. (2007). Drug delivery strategies for poorly water-soluble
solubility of nimesulide in the absence and presence of beta- drugs. Expert Opin Drug Deliv, 4, 403–416.
cyclodextrin derivatives. J Pharm Pharmacol, 60, 1433–1439. French D. (1957). The Schardinger dextrins. Adv Carbohydr Chem,
Agüeros M, Ruiz-Gatón L, Vauthier C, et al. (2009). Combined hydrox- 12, 189–260.
ypropyl-beta-cyclodextrin and poly(anhydride) nanoparticles Freundenberg K, Cramer F, Plieninger H. (1953). Inclusion com-
improve the oral permeability of paclitaxel. Eur J Pharm Sci, 38, pounds of physiologically active organic compounds. German
405–413. Patent 895769.
Araújo J, Gonzalez E, Egea MA, Garcia ML, Souto BE. (2009). Fridriksdottir H, Loftsson T, Stefánsson E. (1997). Formulation and
Nanomedicines for ocular NSAIDs: safety on drug delivery. testing of methazolamide cyclodextrin eye drop solutions.
Nanomed Nanotech Biol Med, In Press. J Controlled Release, 44, 95–99.
Arima H, Adachi H, Irie T, K U. (1996). Possible enhancing of the cuta- Frijlink HW, Paiotti S, Eissens AC, Lerk CF. (1992). The effects of
neous permeation of 4-biphenylacetic acid by beta-­cyclodextrin cyclodextrins on drug release from fatty suppository bases III.
derivatives in hydrophilic ointment. Chem Pharm Bull, 44, Applications of cyclodextrin derivatives. Eur J Pharm Biopharm,
582–586. 38, 174–179.
Arima H, Kondo T, Irie T, Uekama K. (1992). Enhanced rectal absorp- Gaspar de Araújo MV, Barbosa Vieira EK, Silva Lázaro G, et al. (2008).
tion and reduced local irritation of the anti-inflammatory drug Sulfadiazine/hydroxypropyl-β-cyclodextrin host-guest system:
ethyl 4-biphenylylacetate in rats by complexation with water- characterization, phase-solubility and molecular modelling.
­soluble beta-cyclodextrin derivatives and formulation as oleagi- Bioorg Med Chem, 16, 5788–5794.
nous suppository. J Pharm Sci, 81, 1119–1125. Gerlóczy A, Fónagy A, Keresztes P, Perlaky L, Szejtli J. (1985). Absorption,
Asai K, Morishita M, Katsuta H, et al. (2002). The effects of water-sol- distribution, excretion and metabolism of orally adminis-
uble cyclodextrins on the histological integrity of the rat nasal tered 14C-beta-cyclodextrin in rat. Arzneimittelforschung, 35,
mucosa. Int J Pharm, 246, 25–35. 1042–1047.
Bellanger N, Perly B. (1992). NMR investigations of the conformations Gireka T, Kozlowskia CA, Koziola JJ, Walkowiakb W, Korusc I. (2005).
of new cyclodextrin-based amphiphilic transporters for hydro- Polymerisation of b-cyclodextrin with succinic anhydride.
phobic drugs—molecular lollipops. J Mol Struct, 15, 215–226. Synthesis, characterisation, and ion flotation of transition met-
Biwer A, Antranikian G, Heinzle E. (2002). Enzymatic production of als. Carbohydr Polym, 59, 211–215.
cyclodextrins. Appl Microbiol Biotechnol, 59, 609–617. Gould S, Scott RC. (2005). 2-Hydroxypropyl-beta-cyclodextrin
Boulmedarat L, Bochot A, Lesieur S, Fattal E. (2005). Evaluation of (HP-beta-CD): a toxicology review. Food Chem Toxicol, 43,
buccal methyl-beta-cyclodextrin toxicity on human oral epithe- 1451–1459.
lial cell culture model. J Pharm Sci, 94, 1300–1309. Granero GE, Maitre MM, Garnero C, Longhi MR. (2008). Synthesis,
Brewster ME, Loftsson T. (2007). Cyclodextrins as pharmaceutical characterization and in vitro release studies of a new acetazola-
solubilizers. Adv Drug Deliv Rev, 59, 645–666. mide-HP-beta-CD-TEA inclusion complex. Eur J Med Chem, 43,
Cadars S, Foray MF, Gadelle A, Gerbaud G, Bardet M. (2005). High- 464–470.
resolution solid-state 13C NMR study of per(3,6-anhydro)-a- Gref R, Amiel C, Molinard K, et al. (2006). New self-assembled nano-
cyclodextrin based polymers and of their chromium complexes. gels based on host-guest interactions: characterization and drug
Carbohyr Polym, 61, 88–94. loading. J Control Release, 111, 316–324.
Cyclodextrins for drug delivery   655

Haeberlin B, Gengenbacher T, Meinzer A, G., F. (1996). Cyclodextrins— Loftsson T & Frioriksdottir H. (1998). The effect of water-soluble poly-
useful excipients for oral peptide administration. Int J Pharm, mers on the aqueous solubility and complexing abilities of beta-
137, 103–110. cyclodextrin. Int J Pharm, 163, 115–121.
Hamilton RM, Healy RE. (1970). Eliminating undesirable taste from Loftsson T, Järvinen T. (1999). Cyclodextrins in ophthalmic drug deliv-
coffee and tea extracts and products, by treatment with a cyclo- ery. Adv Drug Deliv Rev, 36, 59–79.
dextrin. US33528819 (C.A. 74:12014). Loftsson T, Stefánsson E. (2002). Cyclodextrins in eye drop formula-
Hermes WAJJ, Belder CWJ, Merkus JMWM, Hooymans PM, Verhoef J, tions: enhanced topical delivery of corticosteroids to the eye.
Merkus FWHM. (1992). Intranasal administration of estradiol in Acta Ophthalmol Scand, 80, 144–150.
combination with progesterone to oophorectomized women: a Loftsson T, Gudmundsdóttir H, Sigurjónsdóttir JF, et  al. (2001).
pilot study. Eur J Obstet Gynecol Reprod Biol, 43, 65. Cyclodextrin solubilization of benzodiazepines: formulation of
Higuchi T, Connors KA. (1965). Phase-solubility techniques. Adva Anal midazolam nasal spray. Int J Pharm, 212, 29–40.
Chem Instr, 4, 212–217. Loftsson T, Matthíasson K, Másson M. (2003). The effects of organic
Hirano K, Ishihara T, Ogasawara S, et  al. (2005). Molecular cloning salts on the cyclodextrin solubilization of drugs. Int J Pharm, 262,
and characterisation of a novel gama-CGTase from alkalophilic 101–107.
Bacillus sp. Appl Microbiol Biotechnol, 67, 1–9. Loftsson T, Jarho P, Másson M, Järvinen T. (2005). Cyclodextrins in
Iriea T, Wakamatsua K, Arimaa H, Aritomia H, Uekama K. (1992). drug delivery. Expert Opin Drug Deliv, 2, 335–351.
Enhancing effects of cyclodextrins on nasal adsorption of insulin Lopez RFV, Collet JH, Bentley MVLB. (2000). Influence of cyclodextrin
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

in rats. Int J Pharm, 84, 129–139. complexation on the in vitro permeation and skin metabolism of
Jansook P, Loftsson T. (2008). gammaCD/HPgammaCD: synergistic dexamethasone. Int J Pharm, 200, 127–132.
solubilization. Int J Pharm, 363, 217–219. Maestrellia F, González-Rodríguezb ML, Rabascob AM, Muraa P.
Jarho P, Jarvinen K, Urtti A, Stella VJ, Jarvinen T. (1997). The use of (2006a). Effect of preparation technique on the properties of lipo-
cyclodextrins of ophtalmic formulations of dipivefrin. Int J somes encapsulating ketoprofen–cyclodextrin complexes aimed
Pharm, 153, 225–233. for transdermal delivery. Int J Pharm, 312, 53–60.
Järvinen K, Järvinen T, Thompson DO, Stella VJ. (1994). The effect of Maestrellia F, Garcia-Fuentes M, Mura P, Alonso MJ. (2006b). A new
a modified beta-cyclodextrin, SBE4-beta-CD, on the aqueous drug nanocarrier consisting of chitosan and hydoxypropylcyclo-
stability and ocular absorption of pilocarpine. Curr Eye Res, 13, dextrin. Eur J Pharm Biopharm, 63, 79–86.
897–905. Mallick S, Pattnaik S, Swain K, De PK. (2007). Current perspectives of
Järvinen K, Järvinen T, Urtti A. (1995). Ocular absorption following solubilization: potential for improved bioavailability. Drug Dev
topical delivery. Adv Drug Deliv Rev, 16, 3–19. Ind Pharm, 33, 865–873.
Kaji Y, Uekama K, Yoshikawa H, Takada K, Muranishi S. (1985). Martel B, Ruffin D, Weltrowski M, Lekchiri Y, Morcellet M. (2005).
Selective transfer of 1- hexylcarbamoyl-5-fluorouracil into lym- Water-soluble polymers and gels from the polycondensation
phatics by combination of beta-cyclodextrin polymer complexa- between cyclodextrins and poly(carboxylic acid)s: a study of the
For personal use only.

tion and absorption promotor in the rat. Int J Pharm, 24, 79–89. preparation parameters. J Appl Polym Sci, 97, 433–442.
Kawahara K, Ueda H, Tomono K, Nagai T. (1992). Effect of diethyl Masahikoa K, Fumitoshia H, Kaneto U. (1987). Improvement of
beta-cyclodextrin and the release and absorption behaviour of oral and rectal bioavailabilities of carmofur by methylated
indometacin from ointment bases. STP Pharm Sci, 2, 506–513. β-cyclodextrin complexations. Int J Pharm, 38, 191–198.
Kikuchi M, Hirayama F, Uekama K. (1987). Improvement of oral Matsuda H, Arima H. (1999). Cyclodextrins in transdermal and rectal
and rectal bioavailabilities of carmofur by methylated beta- delivery. Adv Drug Deliv Rev, 36, 81–99.
­cyclodextrin complexations. Int J Pharm, 38, 191–198. McCormack B, Gregoriadis G. (1994). Drugs-in-cyclodextrins-in lipo-
Koizumi K, Okada Y, Kubota Y, Utamuta T. (1987). Inclusion complexes somes: a novel concept in drug delivery. Int J Pharm, 112, 249–258.
of poorly water-soluble drugs with glucosyl-cyclodextrins. Chem Memisoglu-Bilensoy E, Hincal AA, Bochot A, Trichard L, Duchene D,
Pharm Bull, 35, 3416. eds. (2006). Amphiphilic cyclodextrins and microencapsulation.
Kondo T, Irie T, Uekama K. (1996a). Effects of cyclodextrin derivatives In: Microencapsulation, 2nd and revised edition. Benita S. (ed.).
on systemic absorption of morphine through nasal cavity and New York: Taylor & Francis Ltd.
entry into central nervous system in rats. Drug Deliv Syst, 11, 99. Miro A, Rondinone A, Nappi A, Ungaro F, Quaglia F, La Rotonda M.
Kondo T, Irie T, Uekama K. (1996b). Combination effects of (2009). Modulation of release rate and barrier transport of
α-cyclodextrin and xanthan gum on rectal absorption and Diclofenac incorporated in hydrophilic matrices: role of cyclo-
metabolism of morphine from hollow-type suppositories in rab- dextrins and implications in oral drug delivery. Eur J Pharm
bits. Biol Pharm Bull, 19, 280–286. Biopharm, 72, 76–82.
Krauland AH, Alonso MJ. (2007). Chitosan/cyclodextrin nanoparti- Monza Da Silveira A, Ponchel G, Duchene D, Couvreur P, Puisieux F.
cles as macromolecular drug delivery system. Int J Pharm, 340, (1999). Nanoparticles comprising polyisobutylcyanoacrylate and
134–142. cuclodextrins. France, WO 99/43359.
Lang JC. (1995). Ocular drug delivery conventional ocular formula- Nagarwala RC, Kantb S, Singha PN, Maitic P, Pandita JK. (2009).
tions. Adv Drug Deliv Rev, 16, 39–43. Polymeric nanoparticulate system: a potential approach for
Lantz AW, Rodriguez MA, Wetterer SM, Armstrong DW. (2006). ocular drug delivery. J Control Release, 136, 2–13.
Estimation of association constants between oral malodor com- Okamoto H, Komatsu H, Hashida M, Sezaki H. (1986). Effects of
ponents and various native and derivatized cyclodextrins. Anal β-cyclodextrin and di-O-methyl-β-cyclodextrin on the percuta-
Chim Acta, 557, 184–190. neous absorption of butylparaben, indomethacin and sulfanilic
Legendre JY, Rault I, Petit A, et al. (1995). Effects of beta-cyclodextrins acid. Int J Pharm, 30(1), 35–45.
on skin: implications for the transdermal delivery of piribedil and Otagiri M, Fujinaga T, Sakai A, Uekama K. (1984). Effects of beta-
a novel cognoition enhancing-drug, S-9977. Eur J Pharm Sci, 3, ­cyclodextrin and gama-cyclodextrins on release of betametha-
311–322. sone from ointment bases. Chem Pharm Bull, 32, 2401–2405.
Lin J. (1995). Synthèse des cyclodextrins amphiphiles et étude de Palma SD, Tartara LI, Quinteros D, Allemandi DA, Longhi MR,
leur incorporation dans des phases phospholipidiques. Thesis, Granero GE. (2009). An efficient ternary complex of acetazola-
Université de Paris VI. mide with HP-ss-CD and TEA for topical ocular administration.
Ling W, Xuehuab J, Weijuan X, Chenrui L. (2007). Complexation of J Control Release, 138, 24–31.
tanshinone IIA with 2-hydroxypropyl-beta-cyclodextrin: effect Patel SG, Rajput SJ. (2009). Enhancement of oral bioavailability of
on aqueous solubility, dissolution rate, and intestinal absorp- cilostazol by forming its inclusion complexes. Eur J Pharm
tion behavior in rats. Int J Pharm, 341, 58–67. Biopharm, 73, 202–204.
Lister RE, Acerbi D, Cadel S. (1993). Supermolecular inclusion of Perrin JH, Field FP, Hansen DA, Mufson RA, Torosian G. (1978).
piroxicam with beta-cyclodextrin: a review of its pharmacologi- beta-Cyclodextrin as an aid to peritoneal dialysis. Renal toxic-
cal properties in laboratory animals. Eur J Rheumatol Inflamm, ity of beta-cyclodextrin in the rat. Res Commun Chem Pathol
12, 6–11. Pharmacol, 19, 373–376.
656   A.L. Laza-Knoerr, R. Gref, and P. Couvreur

Phillip Lee YH, Sathigari S, Jean Lin YJ, et  al. (2009). Gefitinib- Szejtli J. (1998). Introduction and general overview of cyclodextrin
cyclodextrin inclusion complexes: physico-chemical charac- chemistry. Chem Rev, 98, 1743–1754.
terization and dissolution studies. Drug Dev Ind Pharm, 35, Szeman J, Ueda H, Szejtli J, et  al. (1987). Enhanced percutaneous
1113–1120. absorption of homogenized tolnaftate/beta-cyclodextrin poly-
Qi Q, Mokhtar MN, Zimmermann W. (2007). Effect of ethanol on the mer ground mixture. Drug Des Deliv, 1, 325–332.
synthesis of large-ring cyclodextrins by cyclodextrin glucan- Takada M, Ide T, Yamamoto T, et al. (2003). Novel cyclodextrin glu-
otransferases. J Incl Phenom Macrocycl Chem, 57, 95–99. canotransferase, process for producing the same and process
Rahman AAA, Khidr SH, Ahmed SM, Aboutaleb AE. (1991). Evaluation for producing cyclodextrin by using this enzyme. USA Patent
of chloramphenicol-beta-cyclodextrin inclusion complex. Eur J 20030194796 A1.
Pharm Biopharm, 37, 34–37. Tenjarla S, Puranajoti P, Kasina R, Mandal T. (1998). Preparation,
Ren K, Zhang Z, Li Y, et al. (2009). Physicochemical characteristics and characterisation and evaluation of miconazole-cyclodextrin
oral bioavailability of andrographolide complexed with hydroxy- complexes for improved oral and topical delivery. J Pharm Sci,
propyl-beta-cyclodextrin. Pharmazie, 64, 515–520. 87, 425–429.
Renard E, Deratani A, Volet G, Sebille B. (1997). Preparation and Tokumura T, Muraoka A, Machida Y. (2009). Improvement of oral bio-
characterisation of water soluble high molecular weight beta- availability of flurbiprofen from flurbiprofen/beta-cyclodextrin
­cyclodextrin–epichlorohydrin polymers. Eur Polym J, 33, 49–57. inclusion complex by action of cinnarizine. Arch Pharm Res, 32,
Rezende BA, Cortes SF, De Sousa FB, et al. (2009). Complexation with 155–156.
Journal of Drug Targeting Downloaded from informahealthcare.com by Dalhousie University on 04/13/13

beta-cyclodextrin confers oral activity on the flavonoid dioclein. Uekama K, Irie T, eds. (1987). Industrial uses of methylated cyclo-
Int J Pharm, 367, 133–139. dextrins derivatives. In: Cyclodextrins and Their Industrial Uses.
Ritter H, Tabatabai M. (2002). Cyclodextrin in polymer synthesis: a Duchene D. (eds). Paris: Editions de Santé.
green way to polymers. Prog Polym Sci, 27, 1713–1720. Valle EMM. (2004). Cyclodextrins and their uses: a review. Process
Sathigari S, Chadha G, Lee YH, et al. (2009). Physicochemical charac- Biochemistry, 39, 1033–1046.
terization of efavirenz-cyclodextrin inclusion complexes. AAPS Villiers A. (1891). Sur la fermetation de la fécule de pommes par
PharmSciTech, 10, 81–87. l’action du ferment butyrique. Compt Rend Acad Sci, 112, 536.
Schardinger F. (1903). Uber thermophile Backterien aus verschiedenen Watanabe Y, Matsumoto Y, Seki M, Takase M, Matsumoto M. (1992).
Speisen und Milch, sowie uber einige Umsetzungsprodukte Absorption enhancement of polypeptide drugs by cyclodextrins.I.
derselben in kohenhydrathaltigen Nahrlosungen, darunter krys- Enhanced rectal absorption of insulin from hollow-type sup-
tallisierte Polysacaccharide (Dextrine) aus Starke. Z Untersuch positoires containing insulin and cyclodextrins in rabbits. Chem
Nahr u Genussm, 6, 865. Pharm Bull, 40, 3042–3047.
Shimpi S, Chauhan B, Shimpi P. (2004). Cyclodextrins: applica- Yanagi H, Ishikura N, Kenmotsu H. (1991). Effect of inclusion compl-
tion in different routes of drug administration. Pharm Res, 21, exation of decanoic acid with alfa -cyclodextrin on rectal absorp-
201–230. tion of cefmetazole sodium suppository in rabbits. Yakugaku
For personal use only.

Siguroardottir MA, Loftsson T. (1995). The effect of polyvinylpyrro- Zasshi, 111, 65–69.
lidone on cyclodextrin complexation of hydrocortisone and its Zhang J, Ma PX. (2009). Polymeric core-shell assemblies mediated by
diffusion throught hairless mouse skin. Int J Pharm, 126, 73–78. host-guest interactions: versatile nanocarriers for drug delivery.
Sjejtli J, Szente L. (2005). Elimination of bitter, disgusting tastes of Angew Chem Int Ed Engl, 48, 964–968.
drugs and foods by cyclodextrins. Eur J Pharm Biopharm, 61, Zhang P, Ling CC, Coleman AW, Parrot- Lopez H, Galons H. (1991).
115–125. Formation of amphiphilic cyclodextrins via hydrophobic esteri-
Soares AF, Carvalho RdeA, Veiga F. (2007). Oral administration of pep- fication at the secondary hydroxyl face. Tetrahedron Lett, 32,
tides and proteins: nanoparticles and cyclodextrins as biocom- 2769–2770.
patible delivery systems. Nanomed, 2, 183–202. Zhao D, Zhao L, Zhu C-S, Huang W-Q, Hu J-L. (2009). Water-insoluble
Songa LX, Baia L, Xub MX, Hea J, Pana SZ. (2009). Inclusion complexa- b-cyclodextrin polymer crosslinked by citric acid: synthesis and
tion, encapsulation interaction and inclusion number in cyclo- adsorption properties toward phenol and methylene blue. J Incl
dextrin chemistry. Coord Chem Rev, 253, 1276–1284. Phenom Macrocycl Chem, 63, 195–201.
Stella VJ, He Q. (2008). Cyclodextrins. Toxicol Pathol, 36, 30–42. Zhaofeng L, Miao W, Feng W, et al. (2007). γ-Cyclodextrin: a review
Stickley RG. (1994). Solubilizing excipients in oral and injectable for- on enzymatic production and applications. Appl Microbiol
mulations. Med Res Rev, 14, 353–386. Biotechnol, 77, 245–255.

You might also like