throbber
R E V I E W S
`
`CYCLODEXTRIN-BASED
`PHARMACEUTICS: PAST, PRESENT
`AND FUTURE
`
`Mark E. Davis* and Marcus E. Brewster‡
`
`Abstract | Cyclodextrins are cyclic oligomers of glucose that can form water-soluble inclusion
`complexes with small molecules and portions of large compounds. These biocompatible, cyclic
`oligosaccharides do not elicit immune responses and have low toxicities in animals and humans.
`Cyclodextrins are used in pharmaceutical applications for numerous purposes, including
`improving the bioavailability of drugs. Current cyclodextrin-based therapeutics are described and
`possible future applications discussed. Cyclodextrin-containing polymers are reviewed and their
`use in drug delivery presented. Of specific interest is the use of cyclodextrin-containing polymers
`to provide unique capabilities for the delivery of nucleic acids.
`
`Cyclodextrins (CDs) comprise a family of cyclic
`oligosaccharides, and several members of this family
`are used industrially in pharmaceutical and allied
`applications. CDs are manufactured from starch, one
`of the two glucose-containing polymers produced by
`photosynthesis (the other is cellulose). Starch consists
`of D-glucopyranoside building blocks that have both
`α-1,4- AND α-1,6-GLYCOSIDIC LINKAGES. The degradation of
`starch (which is primarily derived from corn, but also
`from potatoes and other sources) by the enzyme gluco-
`syltransferase generates, by chain splitting and
`intramolecular rearrangement, primary products that
`are cyclic oligomers of α-1,4-D-glucopyranoside, or
`CDs. FIGURE 1 shows several schematic representations
`of β-CD. CDs derive their system of nomenclature
`from the number of glucose residues in their structure,
`such that the glucose hexamer is referred to as α-CD,
`the heptamer as β-CD and the octomer as γ-CD (FIG. 2).
`There are literally thousands of variations of CDs that
`have variable ring size and random or site-specific
`chemical functionalization. A comprehensive overview
`of all aspects of CDs is available1.
`The earliest reference to a substance that was later
`recognized as a CD was published in 1891 (REF. 2). By
`1953, Freudenberg et al.3 had received the first patent on
`the use of CDs in drug formulations. This patent covered
`
`most of the important concepts that are used even today,
`including the improvement of drug properties such as
`increased aqueous solubility and increased stability
`towards drug oxidation. Currently, CDs have found uses
`in many applications, such as in agrochemicals, pharma-
`ceuticals, fragrances, foods and so on. This review will
`concentrate on the pharmaceutical uses of CDs.
`
`The basis of CDs as pharmaceutical excipients
`The three-dimensional structure of CDs endows them
`with properties that are useful for pharmaceutical appli-
`cations. Because of the large number of hydroxyl
`groups on CDs, they are water-soluble. The water solu-
`bilities of α-, β- and γ-CD at ambient conditions are
`approximately 13%, 2% and 26% (weight by weight
`(w/w)), respectively (for β-CD this is approximately
`18.8 g per l or 16.6 mM)1. The lower solubility of β-CD
`compared with α-CD, even though the former contains
`a higher number of hydroxyl groups than the latter, is
`due to the formation of an internal hydrogen-bond
`network between the secondary hydroxyl groups. The
`disruption of hydrogen bonding via molecular
`manipulation gives rise to an increase in water solu-
`bility. For example, hydroxypropyl-β-CD (HPβCD)
`has an aqueous solubility of 60% (w/w) or more4.
`Although the entire CD molecule is water soluble, the
`
`α-1,4- AND α-1,6- GLYCOSIDIC
`LINKAGES
`The D-glucopyranoside unit
`contains six carbons and two
`of these units can be chemically
`linked from the 1-carbon of a
`unit to either the 4-carbon or
`the 6-carbon of the second unit.
`
`*Chemical Engineering,
`California Institute of
`Technology, Pasadena,
`California 91125, USA.
`‡Johnson & Johnson
`Pharmaceutical Research
`and Development,
`Turnhousteweg 30,
`2340 Beerse, Belgium.
`Correspondence to M.E.D.
`e-mail:
`mdavis@cheme.caltech.edu
`doi:10.1038/nrd1576
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 3 | DECEMBER 2004 | 1 0 2 3
`
`Hopewell EX1068
`Hopewell v. Merck
`IPR2023-00480
`
`1
`
`

`

`OH
`
`OH
`
`O
`
`OH
`
`O
`
`O H
`O
`
`O H
`
`O
`
`HO
`
`O
`
`H
`O
`
`H O
`
`H
`O
`
`O
`
`OO
`H
`
`H
`
`OH
`
`O O
`
`R E V I E W S
`
`HO
`
`O H
`
`O
`
`H O
`
`O H
`
`O
`
`HO
`
`O
`
`HO
`O
`
`OH
`
`HO
`
`O
`
`OH
`
`O
`OH
`
`OH
`Figure 1 | Schematic representations of β-cyclodextrin. The open and closed arrows point to primary and secondary
`hydroxyl groups, respectively. The cyclodextrin (CD) architecture is a cup that is 0.79 ± 0.01 nm from top to bottom (primary OH
`face to secondary OH face), and is slightly larger on the face containing secondary hydroxyl groups. The cavity (0.47–0.53,
`0.60–0.65 and 0.75–0.83 nm for α-, β- and γ-CD, respectively) and exterior diameters of the CDs (1.46 ± 0.04, 1.54 ± 0.04 and
`1.75 ± 0.04 nm for α-, β- and γ-CD, respectively, for the faces containing secondary hydroxyl groups) expand as the number of
`glucopyranoside units increase1.
`
`interior of the cup is relatively apolar and creates a
`HYDROPHOBIC micro-environment. CDs therefore have
`HYDROPHILIC cavity exteriors and hydrophobic cavity
`interiors. These properties are responsible for their
`aqueous solubility and ability to encapsulate hydro-
`phobic moieties within their cavities, and the incorpo-
`ration of ‘guest’ molecules in CD inclusion complexes
`in aqueous media has been the basis for most pharma-
`ceutical applications. A dynamic equilibrium between
`free CDs, free drug molecules and their formed inclu-
`sion complexes is established if drug molecules are of
`
`HYDROPHOBIC
`An affinity for, and propensity
`to dissolve in, non-polar solvents
`such as hydrocarbons.
`
`HYDROPHILIC
`An affinity for, and propensity
`to dissolve in, water and other
`polar solvents.
`
`a
`
`OH
`
`O
`
`O
`OH
`
`OH
`
`O
`
`OH
`O
`
`OH
`
`OH
`
`O
`
`HO
`
`OH
`
`O
`
`HO
`
`O
`
`b
`
`O
`OH
`
`OH
`
`HO
`
`O
`
`O
`OH
`
`OH
`O
`
`O
`
`HO
`OH
`
`HO
`
`OH
`O
`
`HO
`
`O
`
`sufficient size and have appropriate properties for the
`formation of inclusion complexes. FIGURE 3 schematically
`illustrates this dynamic equilibrium for 1:1 and 1:2
`drug–CD complexes. The formation of inclusion
`complexes is possible with the entire drug molecule or
`only a portion of it. FIGURE 3C presents models of how
`α-, β- and γ-CD can form inclusion complexes with
`prostaglandin E2. Because of cavity size, α-CD com-
`plexes well with aliphatic chains and molecules such as
`polyethylene glycol (PEG), whereas β-CD is appropriate
`for aromatic rings, such as that in paclitaxel.
`For CDs to be pharmaceutically useful, they must be
`biocompatible. CDs show resistance to degradation by
`human enzymes; CDs injected intravenously into
`humans are therefore essentially excreted intact via the
`kidney. However, bacterial and fungal enzymes (amy-
`lases) can degrade CDs. Ingested CDs can therefore be
`metabolized in the colon prior to excretion. The toxicities
`of CDs are dependent on their route of administration.
`For example, the dose that causes 50% death (LD50)
`values of α-, β- and γ-CD administered intravenously
`into mice are approximately 1.0 g per kg5, 0.79 g per kg5
`and more than 4.0 g per kg6, respectively. β-CD has an
`affinity for cholesterol and can extract it and other lipid
`membrane components from cells. At sufficiently high
`concentrations, β-CD can cause haemolysis of ery-
`throcytes. Additionally, parenteral administration of
`β-CD is not possible because of its poor solubility
`(which leads to microcrystalline precipitation in the
`kidney), as well as the fact that it forms complexes
`with cholesterol that accumulate in the kidney and
`produce renal tubule damage. Functionalized β-CDs
`can mitigate these problems.
`Chemically modified CDs result from etherification
`or the introduction of other functional groups at the 2-,
`3- and 6-hydroxyl groups of the glucose residues. These
`changes improve solubility through two mechanisms: by
`breaking the 2-OH–3-OH hydrogen bonds, and by pre-
`venting crystallization due to creation of a statistically
`substituted material that is made up of many isomeric
`components and gives rise to an amorphous product.
`
`O
`
`HO
`
`OH
`
`O
`
`OH
`
`OH
`
`OH
`
`HO
`
`HO
`O
`
`O
`
`OH
`
`O
`OH
`
`HO
`
`O
`
`O
`
`OH
`
`OH
`O
`
`HO
`OH
`
`O
`
`O
`OH
`
`OH
`
`O
`
`HO
`
`HO
`
`OH
`
`HO
`
`O
`
`O
`
`OH
`
`OH
`
`OH
`
`O
`
`OH
`
`O
`
`OH
`
`c
`
`HO
`
`O
`
`O
`
`HO
`
`O
`OH
`
`OH
`
`O
`
`OH
`
`OH
`
`OH
`O
`
`HO
`
`HO
`
`O
`
`HO
`
`HO
`O
`
`OH
`
`O
`
`OH
`
`O
`HO
`
`O
`
`OH
`
`OH
`
`HO
`O
`
`O
`HO
`Figure 2 | Schematic representations of cyclodextrins. α-CD (a), β-CD (b) and γ-CD (c)
`contain 6, 7 and 8 glucopyranoside units, respectively. The molecular masses of α-, β- and γ-CD
`are 972, 1,135 and 1,297 Da, respectively.
`
`O
`
`OH
`
`1024 | DECEMBER 2004 | VOLUME 3
`
`www.nature.com/reviews/drugdisc
`
`2
`
`

`

`R E V I E W S
`
`response in mammals. Because of these highly desirable
`properties, CDs have found numerous pharmaceutical
`applications; reviews on the use of CDs in drug delivery
`are available7–13.
`
`Pharmaceutical applications of CDs
`Current pharmaceutical research has a number of drivers,
`including the nature of the drugs being developed, the
`need for generating orally bioavailable dosage forms
`and the preparation of solubilized parenteral formula-
`tions. Drug discovery has evolved over the years to the
`point that high-throughput screening techniques have
`become routine. These approaches put significant evo-
`lutionary pressure on emerging drug candidates, and
`this has led to a systematic increase in molecular mass,
`lipophilicity and a decrease in water solubility for lead
`compounds over time14,15. This, in turn, has had a sig-
`nificant impact on what is required from drug delivery
`formulators, in that the number of formulation options
`has had to be increased to address the larger diversity of
`challenges presented.
`For a drug to be orally available, the compound
`must dissolve and be absorbed through the gastro-
`intestinal tract in such a way that it generates adequate
`drug levels at the pharmacologically active site to ensure
`that the desired action is obtained in a reproducible
`manner. Retrospective studies show that >40% of drug
`failures in development can be traced to poor biophar-
`maceutical properties, specifically poor dissolution or
`poor permeability16. In recognition of the importance of
`these factors, the FDA and other drug regulatory
`organizations have defined a Biopharmaceutical Classi-
`fication System in which drugs are divided into four
`types on the basis of their solubility and permeability
`characteristics (FIG. 4)17–19. High-throughput drug dis-
`covery methodologies are increasingly selecting difficult
`Type II compounds, and CDs can be an important
`enabling technology for these compounds in partic-
`ular20,21. By increasing the apparent water solubility of
`a drug candidate, formulations can be generated such
`that a Type II material behaves like a Type I compound,
`with a resulting increase in oral bioavailability20,22 (BOX 1).
`The reasons for the inclusion of CDs in a particular
`formulation can vary widely (BOX 2)23, and are specific
`to the circumstance — that is, the specific physico-
`chemical issues that have to be overcome and the
`administration route24–26.
`
`Initial applications of α- and β-CDs: prostaglandin
`and nonsteroidal anti-inflammatory agents. CDs first
`came to the fore in marketed products as drug delivery
`technologies that enabled the development of various
`prostaglandins27,28. One of first of these compounds,
`PGE2, a substance with potent oxytocin-like effects,
`was of interest as a possible agent for the induction of
`labour in childbirth29,30. As with other members of the
`E-type prostoglandins, these compounds are highly
`unstable, and this feature complicated their formula-
`tion and development. β-CD complexes of PGE2
`resulted in a significant increase in their solid-state
`stability, and a product designed along these lines
`
`Drug
`
`1:1 drug–CD complex
`
`CD
`
`+
`
`+
`
`CD
`
`Drug
`
`1:2 drug–CD complex
`
`CO2H
`
`b
`
`O
`
`HO
`
`OH
`
`CO2H
`
`c
`
`O
`
`HO
`
`OH
`
`CO2H
`
`OH
`
`A
`
`B
`
`Ca
`
`O
`
`HO
`
`Figure 3 | Schematic illustration of the association of free cyclodextrin (CD) and drug to
`form drug–CD complexes. A | 1:1 drug–CD complex. B | 1:2 drug–CD complex. C | Proposed
`models of inclusion complexes between prostaglandin E2 and (a) α-CD, (b) β-CD and (c) γ-CD.
`Adapted from REF. 1.
`
`The complexity of these mixtures can be appreciated
`by considering β-CD. For this compound, there are 21
`hydroxyl functional groups and therefore 221 –1 possible
`combinations for substitutions (that is, more than 2
`million). If an optically active centre is introduced, as in
`the case of 2-hydroxypropylation, the number of geo-
`metrical and optical isomers is truly astronomical, given
`that the β-CD nucleus contains 28 chiral centres. It is
`conceivable that the pharmaceutical performance of
`these isomeric mixtures can change with the extent and
`degree of substitution, and so these factors have to be
`assessed and specified in the excipient. In practice, this
`is done by analogy with other chemically modified
`pharmaceutical starches and celluloses, such as hydroxy-
`propyl cellulose and hydroxylpropylmethyl cellulose.
`Both the European Pharmacopeial monograph and the
`proposed United States Pharmocopeial monograph on
`HPβCD, for example, specify that the material should
`have a molar substitution (expressed as the number of
`hydroxypropyl groups per anhydroglucose unit)
`between 0.4 and 1.5; this means 2.8–10.5 hydroxypropyl
`functional groups per cyclodextrin molecule. They also
`specify that less than 1.5% unmodified β-CD should be
`present. The molar substitution can be determined
`using nuclear magnetic resonance and infra-red
`methods. Two functionalized CDs, hydroxypropyl β-CD
`(HPβCD) and sulphobutyl ether β-CD (SBEβCD), are
`available in FDA-approved products for human use (see
`below). In addition, CDs do not produce an immune
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 3 | DECEMBER 2004 | 1 0 2 5
`
`3
`
`

`

`On the basis of this administration route, the stabilizing
`effect of α-CD on PGE1 and the suitability of this CD
`for parenteral use (unlike β-CD), formulations of
`PGE1/α-CD complexes were developed. In 1979,
`alprostadil alphadex (Prostavasin) was approved for
`the treatment of peripheral vascular complications,
`including Buerger’s disease29,30. The compound also
`showed activity against chronic arterial occlusions and
`arteriosclerosis.
`Another vascular malady that can be treated
`with alprostadil alphadex is male erectile dysfunction,
`for which the complex is given by intracavernous
`injection31–34. The PGE1–α-CD complex was found to be
`effective in subjects who were not responsive to sildenafil
`citrate (Viagra; Pfizer), an oral inhibitor of phospho-
`diesterase-535. In a study of 67 patients that failed
`sildenafil citrate therapy at 50 and 100 mg, 85–90%
`reported improvements in erectile function when
`PGE1–α-CD was self-administered after ‘at-home’ treat-
`ment. Caverject Impluse is approved for use in the
`United States on the basis of these medical needs.
`The complexity of the administration route prompted
`the development of more convenient dosing options,
`including intravenous dosage forms. These drivers
`resulted in a modified formulation (Prostandin), which
`was approved in 1982 in Japan and subsequently in a
`number of other countries, including Germany29,30.
`A third example of a prostaglandin marketed as a CD
`complex is limaprost alphfadex (Opalmon/ Prorenal;
`Ono)29,30,36,37. This prostaglandin analogue was devel-
`oped for the treatment of vascular disease and was
`shown to have improved antiplatelet aggregation and
`vasodilation activity relative to PGE1. Importantly, the
`compound was orally available and showed a good sepa-
`ration between its therapeutic action and unwanted side
`effects (mainly oxytocin-like effects). The limoprost–α-
`CD complex was found to be safe and effective in the
`treatment of Buerger’s disease and was approved in 1988.
`Nonsteroidal anti-inflammatory drugs (NSAIDs) are
`a mainstay for the treatment of pain38. The major
`drawbacks of these otherwise useful compounds
`include upper gastrointestinal irritation and bleed-
`ing39. Piroxicam (Feldene; Pfizer) is an illustrative
`example. The drug is useful in the treatment of
`osteoarthritis and rheumatoid arthritis, as well as gout,
`acute musculoskeletal disorders and dysmenorrhea40–43.
`It has a relatively long pharmacokinetic half-life, meaning
`that it can be taken once a day, in contrast to many other
`NSAIDs. The parent drug is also poorly water soluble
`(~30 μg per ml), poorly WETTABLE (that is, with a contact
`angle of 70°) and highly crystalline (with a melting
`point of 202 °C and a ΔHmelt of 106 J per g)44–47. This
`imparts to the compound poor dissolution properties,
`as well as dissolution-limited oral pharmacokinetics.
`CDs were applied to this compound in an effort to
`improve several properties, including safety and drug
`dissolution rate. These improved characteristics reduced
`gastrointestinal irritation, and allowed more rapid drug
`absorption and a more rapid onset of the analgesic
`effect. Complexation studies indicated that a molar ratio
`of 2.5 per 1 was optimal for the drug–CD combination
`
`R E V I E W S
`
`Type III
`permeability
`Solubility,
`←
`←
`
`Type I
`←permeability
`Solubility,
`←
`
`Type IV
`Solubility, permeability
`←
`
`←
`
`Type II
`←permeability
`Solubility,
`
`←
`
`Solubility
`
`WETTABILITY
`The wettability of a liquid is
`defined as the contact angle
`between a droplet of the liquid
`in thermal equilibrium on a
`horizontal surface.
`
`Permeability
`
`Figure 4 | Biopharmaceutical Classification System
`(BCS) characterization of drugs based on solubility
`and permeability measures. Drug solubility is defined by
`the dose/solubility ratio, with a soluble drug defined as one
`in which the highest dose intended for human use will
`dissolve in 250 ml of water (the so-called FDA glass of
`water). Permeability can be defined by various in vivo or
`in vitro assays, but a permeable drug is one associated with
`≥90% oral bioavailability or ≥90% absorption as assessed
`by urinary excretion data.
`
`(dinoprostone betadex; Prostarmon E; Ono) was
`approved for the Japanese market in 1976 (TABLE 1).
`Prostarmon E is highly effective and represented a signif-
`icant medical advance, especially for the induction of
`labour in oxytocin-insensitive individuals, but also for
`its tendency to produce less bleeding after delivery.
`The second prostaglandin marketed as a CD com-
`plex was PGE1 (alprostadil alphadex; Prostavasin/
`Edex/Caverject/Prostandin; Schwarz Pharma). PGE1
`relaxes smooth muscle and increases blood flow, and
`was initially developed as a therapeutic to treat periph-
`eral circulatory disorders. Given its limited metabolic
`stability, initial therapies with PGE1 required intra-
`arterial administration to obtain useful clinical results.
`
`Box 1 | Solubilization with cyclodextrins
`
`+
`
`Drug
`
`Kc
`
`Cyclodextrin
`
`Kc = K1:1 =
`
`[D–CD]
`[D] [CD]
`
`Concentration of CD [M]
`
`Concentration of drug [M]
`
`Cyclodextrins (CDs) can enhance
`apparent water solubility by forming
`dynamic, non-covalent, water-soluble
`inclusion complexes as depicted in the
`figure. This interaction is an equilibrium
`governed by an equilibrium constant, Kc.
`The nature of the complex, as well as the
`numerical value of the equilibrium
`constant, can be derived from from
`measuring a particular property of the
`complex as a function of drug and CD
`concentrations. In phase-solubility
`analysis, the increased solubility is
`assessed as a function of CD
`concentration.As illustrated, a number of
`solubility profiles are possible, each giving
`insight into the type of complex formed,
`as well as its stoichiometry. An A-type
`profile (red line) represents the formation
`of soluble CD complexes, whereas B-type
`systems (blue line) indicate the formation
`of complexes of limited solubility.
`
`1026 | DECEMBER 2004 | VOLUME 3
`
`www.nature.com/reviews/drugdisc
`
`4
`
`

`

`and, given the low piroxicam dose (20 mg), this did not
`add excessive bulk to the formulation48. The apparent
`solubility of piroxicam in the β-CD formulation was
`increased fivefold relative to the uncomplexed drug sub-
`stance. This enhanced solubility was associated with an
`increased dissolution rate and higher plasma levels at
`early time in humans, which, in turn, directly correlated
`with an increased absorption rate48,49. There was no
`change in terminal half-life or total area under the curve
`when piroxicam or the piroxicam–β-CD complex were
`compared. With regard to efficacy, piroxicam–β-CD has
`been demonstrated to have a faster onset of action in a
`number of clinical trials48,50,51.
`Although differences in the tolerability of piroxicam
`and its β-CD complex require the analysis of epidemio-
`logical data, acute studies might provide some useful
`insight. Several studies that have assessed the endo-
`scopic appearance of the stomach of volunteers taking
`either piroxicam or the piroxicam–β-CD complex
`have revealed significantly better outcomes in the case
`the complexed drug48,52,53.
`of administration of
`Similarly, radio-adhesive substrates demonstrated fewer
`gastric lesions with the piroxicam–β-CD complex relative
`to piroxicam48,54. Several branded piroxicam–β-CD
`products are available, including Brexin (Chiesi) and
`Cicladol (Chiesi).
`
`Applications of randomly methylated β-CD. Randomly
`methylated β-CD (RMβCD) (FIG. 5) provides good bio-
`compatibility and useful complexing efficiencies, and is
`beginning to be used in marketed products throughout
`the world. An eye drop preparation of the antibiotic
`chloramphenicol has been developed by Oftalder and
`marketed as Clorocil in Portugal55. The recently intro-
`duced nasal product Aerodil, which contains RMβCD, is a
`complex between the indicated CD and β-oestradiol56–60.
`A number of potential advantages are apparent for
`such an oestradiol delivery system. Although the safety
`of hormone-replacement therapy has been the subject of
`recent debate61,62, its efficacy in reducing menopausal
`symptomatology is well established. Traditional dosing
`strategies include oral administration of conjugated
`equine oestrogens, oestradiol esters or micronized
`oestradiol, as well as the use of transdermal patches to
`deliver this sex hormone63–65. Although both routes have
`a number of limitations, both provide relatively con-
`stant blood levels of drug, in contrast to endogenous
`oestrogen release, which tends be more pulsatile.
`The RMβCD-based nasal product avoids a number
`of issues related to oral or transdermal administration56.
`Nasal administration results in direct systemic uptake,
`and so the first-pass effect is reduced or eliminated. In
`addition, the nasal route is convenient, non-invasive and
`provides for consistency of drug absorption. Drug
`uptake is rapid, and peak plasma concentrations are
`achieved within 10–30 min of drug dosing. Drug levels
`also dissipate rapidly and return close to baseline within
`2 hours56. This pattern is more akin to physiological
`oestrogen secretion than that associated with oral or
`transdermal approaches. Another feature of this route is
`that, unlike oral dosing of oestrogens that generate a
`
`R E V I E W S
`
`Box 2 | Pharmaceutical applications of CDs
`
`Cyclodextrins (CDs) can be used to achieve the
`following:
`• Enhance solubility
`• Enhance bioavailability
`• Enhance stability
`• Convert liquids and oils to free-flowing powders
`• Reduce evaporation and stabilize flavours
`• Reduce odours and tastes
`• Reduce haemolysis
`• Prevent admixture incompatibilities
`
`high oestrone/oestradiol ratio, nasal delivery gives rise
`to a more physiological ratio of the two hormones56–60.
`A number of clinical trials have confirmed these
`product design principles. Studd et al. studied 420 post-
`menopausal women and found that the nasal product
`based on oestradiol and RMβCD was effective in amelio-
`rating menopausal symptoms as early as four weeks
`after initiation of therapy in a dose-dependent manner,
`and continued to improve the post-menopausal symp-
`toms even after 12 weeks56. The minimum effective
`oestradiol dose was 200–400 μg per day. These doses
`were similar in efficacy to 2 mg of oestrogen adminis-
`tered orally, although the reduced systemic exposure
`results in a potentially improved safety profile. Inter- and
`intrasubject variability was less than that demonstrated
`in the oral oestrogen group.
`
`Applications of hydroxypropylated β-CD. Two hydroxy-
`propylated CDs (HPβCDs) have been approved in var-
`ious world markets (United States and Europe)25,27,66–69.
`HPβCD (FIG. 5) is available in registered oral, intra-
`venous, buccal, rectal and ophthalmic products, whereas
`HPγCD is available in an eye drop formulation that
`contains the anti-inflammatory agent diclofenac
`sodium70. Of the HPβCD products, the oral and intra-
`venous solutions of itraconazole (Sporanox; Janssen)
`have the most widespread use71.
`Itraconazole is a triazole-type drug that exerts its
`effect by inhibiting fungal cytochrome P450 and
`inhibiting the biosynthesis of ergosterol, an essential
`component of the fungal membrane. The compound is
`noteworthy in that it was the first approved orally
`bioavailable agent with significant clinical activity
`against both Candida spp. and Aspergillus spp., the
`two most common human fungal pathogens72–75.
`Formulation development for this drug was complicated
`by its challenging set of physico-chemical properties,
`which include a pKa of 4, a LOG P >5 and an aqueous
`solubility at neutral pH estimated at 1 ng per ml71,76. The
`production of solid oral dosage forms was eventually
`made possible by using solid solution technology in
`which the drug and a polymeric carrier (hydroxypropyl
`methylcellulose (HPMC)) were sprayed on inert sugar
`spheres to form a thin film. As the film dissolves in the
`stomach, the molecularly dissolved drug is released at
`supersaturated levels. The co-dissolving HPMC inhibits
`
`LOG P
`The logarithm of the partition
`coefficient of a substance in
`octanol–water.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 3 | DECEMBER 2004 | 1 0 2 7
`
`5
`
`

`

`R E V I E W S
`
`Intravenous
`Oral
`Oral
`
`Prostavastin, Caverject, Edex
`Pansporin T
`Opalmon, Prorenal
`
`Europe, Japan, United States
`Japan
`
`Table 1 | Approved and marketed drug–cyclodextrin* complexes in various world markets
`Drug
`Administration route
`Trade name
`Market
`α-Cyclodextrin
`Alprostadil (PGE1)
`Cefotiam hexetil HCl
`Limaprost
`β-Cyclodextrin
`Oral
`Benexate
`Dermal
`Dexamethasone
`Topical
`Iodine
`Sublingual
`Nicotine
`Oral
`Nimesulide
`Sublingual
`Nitroglycerin
`Oral
`Omeprazole
`Dinoprostone (PGE2)
`Sublingual
`Oral
`Piroxicam
`Oral
`Tiaprofenic acid
`2-Hydroxypropyl-β-cyclodextrin
`Cisapride
`Rectal
`Hydrocortisone
`Buccal
`Indomethacin
`Eye drops
`Itraconazole
`Oral, intravenous
`Mitomycin
`Intravenous
`Randomly methylated β-cyclodextrin
`17β-Oestradiol
`Nasal spray
`Chloramphenicol
`Eye drops
`Sulphobutylether β-cyclodextrin
`Voriconazole
`Intravenous
`Ziprasidone maleate
`Intramuscular
`2-Hydroxypropyl-γ-cyclodextrin
`Europe
`Voltaren
`Diclofenac sodium
`Eye drops
`*Commercial suppliers of pharmaceutical-grade cyclodextrins are Roquette, Cerestar, Wacker Chemie and CyDex. See figures 2 and 5
`for schematic representations of cyclodextrins and functionalized cyclodextrins, respectively.
`
`Ulgut, Lonmiel
`Glymesason
`Mena-Gargle
`Nicorette
`Nimedex, Mesulid
`Nitropen
`Omebeta
`Prostarmon E
`Brexin
`Surgamyl
`
`Propulsid
`Dexocort
`Indocid
`Sporanox
`Mitozytrex
`
`Aerodiol
`Clorocil
`
`Japan
`Japan
`Japan
`Europe
`Europe
`Japan
`Europe
`Japan
`Europe
`Europe
`
`Europe
`Europe
`Europe
`Europe, United States
`United States
`
`Europe
`Europe
`
`Vfend
`Geodon, Zeldox
`
`Europe, United States
`Europe, United States
`
`nucleation and crystallization such that the supersatu-
`rated drug solutions are stable for long enough to allow
`significant absorption and oral bioavailability.
`Given the weakly basic nature of itraconazole, the pH
`of the stomach must be sufficiently low to ensure good
`dissolution and the formation of a stable solution78,79. In
`certain subpopulations, stomach pH can be a limiting
`factor for bioavailability.AIDS patients, for example, often
`suffer from hypochlorhydria and so treating opportunis-
`tic fungal infections with itraconazole in the patients can
`be problematic80. Similarly, the chemotherapeutic agents
`given to cancer patients can reduce gastrointestinal func-
`tion by inducing mucositis, and an individual undergoing
`autologous bone marrow transplants can experience
`graft-versus-host gut disease81.
`To better prevent or treat fungal infections in these
`conditions, a liquid oral formulation of itraconazole
`was developed, in addition to a parenteral dosage form
`for treating disseminated systemic infection, as well as for
`use in administering loading doses. HPβCD was chosen
`as the functional excipient to enable both of these
`
`formulations71. It was reasoned that the oral solution
`might improve bioavailability in the described sub-
`populations because no phase transition is required.
`The oral and intravenous formulations that were
`developed proved to be safe and effective in numerous
`clinical trials, and this resulted in their market introduc-
`tion in the United States and Europe (the oral solution
`in 1997 and the intravenous product in 1999)81–83. The
`oral product is indicated in empiric therapy of febrile
`patients with suspected fungal infections, as well as
`for the treatment of oropharyngeal and oesophageal
`candidiasis81,84. The aqueous 40% w/v HPβCD solution
`contains 10 mg per ml itraconazole, which represents an
`increase in apparent solubility of five to six orders of
`magnitude. The oral bioavailability of itraconazole is
`more consistent in the subpopulations described
`(fraction absorbed 85% and oral bioavailability
`55%)81,85. On the basis of an oral dose of 200 mg itra-
`conazole, the dose of HPβCD is 8 g per day. The
`intravenous product is also approved for empiric
`therapy as well as for blastomycosis (pulmonary and
`
`1028 | DECEMBER 2004 | VOLUME 3
`
`www.nature.com/reviews/drugdisc
`
`6
`
`

`

`CH2OR
`O
`
`ROCH2
`
`O
`
`O
`OR
`
`RO
`
`RO
`
`O
`
`CH2OR
`
`OR
`
`O
`
`O
`
`OR
`
`ROCH2
`
`OR
`
`O
`
`RO
`
`RO
`
`O
`
`RO
`
`CH2OR
`
`O
`
`OR
`
`OR
`
`O
`
`O
`
`RO
`
`RO
`
`O
`
`RO
`
`O
`
`R E V I E W S
`
`Dimethyl-β-cyclodextrin (DMβCD)
`Trimethyl-β-cyclodextrin (TMβCD)
`Randomly methylated-β-cyclodextrin (RMβCD)
`Hydroxyethyl-β-cyclodextrin (HEβCD)
`2-Hydroxypropyl-β-cyclodextrin (HPβCD)
`3-Hydroxypropyl-β-cyclodextrin (3HPβCD)
`2,3-Dihydroxypropyl-β-cyclodextrin (DHPβCD)
`2-Hydroxyisobutyl-β-cyclodextrin (HIBβCD)
`Sulphobutylether-β-cyclodextrin (SBEβCD)
`Glucosyl-β-cyclodextrin (G1βCD)
`Maltosyl-β-cyclodextrin (G2βCD)
`
`-CH3 or -H
`-CH3
`-CH3 or -H
`-CH2CH2OH or -H
`-CH2CHOHCH3 or -H
`-CH2CH2CH2OH or -H
`-CH2CHOHCH2OH or -H
`-CH2C(CH3)2OH or -H
`-(CH2)4SO3Na or -H
`-glucosyl or -H
`-maltosyl or -H
`
`CH2OR
`Figure 5 | Graphical representations of β-cyclodextrin and its pharmaceutically relevant chemical derivatives.
`
`O
`
`CH2OR
`
`extra-pulmonary), histomycosis (pulmonary and dis-
`seminated, non-meningeal) and aspergillosis (pul-
`monary and non-pulmonary), in the latter case in
`individuals refractory or intolerant to amphotericin
`B81,84. Based on intravenous doses of itraconazole
`between 200 and 400 mg and a formulation containing
`10 mg itraconazole in a 40% HPβCD solution, the intra-
`venous dose of HPβCD is between 8 and 16 g per day.
`
`Applications of sulphobutyl ether βCD. The sulphobutyl
`ether derivatives of β-CD (SBEβCD; FIG. 5) represent the
`newest CD derivative to be approved25,86–88. In addition,
`γ-CD derivatives of these useful excipients are also
`available. Two products were introduced in 2002 in the
`United States and Europe, including an intravenous
`formulation of the antifungal agent voriconazole
`(Vfend; Pfizer) and an intramuscular dosage form for
`the antipsychotic agent ziprasidone (Zeldox; Pfizer).
`In addition, a number of compounds are under devel-
`opment in areas including parenteral, oral sustained
`release, pulmonary and ophthalmalogical drug delivery.
`Voriconazole is a triazole antifungal agent that, unlike
`itraconazole, which evolved from the miconazole-
`ketoconazole series, is structurally related to flucon-
`azole89–91.Voriconazole is effective against Candida spp. as
`well as Aspergillus spp., and is also reported to be effective
`against such emerging pathogens as Scedospotium and
`Fusarium spp.89–91. As with itraconazole, it can partici-
`pate in numerous drug interactions. The compound is
`also poorly water soluble (~0.2 mg per ml at pH 3) and
`is not stable in aqueous solutions because it forms an
`inactive enantiomer. Unlike itraconazole, which has a
`log P >5, voriconazole is more amphiphilic and has
`a log P of 1.8. These factors complicate the preparation
`of a parenteral dosage form92.
`SBEβCD was used in this formulation problem
`and resulted in a dosage form containing 3.2 g of the
`CD and 200 mg of the active principle84. The supplied
`vial is diluted with water for injection to give a 10 mg
`per ml solution of voriconazole in a 16% w/v
`SBEβCD solution; this is then further diluted such
`that the administration solution contains 5 mg per
`ml or less of the active principle. The intravenous dose
`suggested is 3–6 mg per kg, which means

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket