throbber
International Scholarly Research Network
`ISRN Pharmacology
`Volume 2012 Article ID 623139 15 pages
`doi1054022012623139
`
`Review Article
`Designing Paclitaxel Drug Delivery Systems Aimed at Improved
`Patient Outcomes Current Status and Challenges
`
`Madhu S Surapaneni1 Sudip K Das and Nandita G Das
`
`1
`
`College ofPharmacy Idaho State University Pocatello ID 83201 USA
`Sciences Butler University Indianapolis IN 46208 USA
`2 Department of Pharmaceutical
`
`Correspondence
`
`should be addressed to Nandita G Das ndasbutleredu
`
`Received
`
`3 April 2012 Accepted 29 April 2012
`
`Academic Editors G A Gudelsky T Irie JA Mico and BN Wu
`Copyright 0 2012 Madhu S Surapaneni et al This is an open access article distributed under the Creative Commons Attribution
`License which permits unrestricted use distribution and reproduction in any medium provided the original work is properly
`cited
`
`agents derived from natural sources It has a wide spectrum
`is one of the most widely used and effective antineoplastic
`Paclitaxel
`of antitumor activity particularly against ovarian cancer breast cancer nonsmall cell lung cancer head and neck tumors Kaposis
`is a highly lipophilic compound with a log P value of 396 and very poor aqueous solubility
`sarcoma and urologic malignancies It
`of less than 001 mgmL In addition the compound lacks functional groups that are ionizable which could potentially lead to an
`increase in its solubility with the alteration in pH Therefore the delivery of paclitaxel
`is associated with substantial
`challenges
`the introduction of Abraxane only commercial formulation was solution of paclitaxel
`in cremophor which caused severe
`Until
`in recent years a number of approaches have been reported to solubilize paclitaxel
`side effects However
`using cosolvents and
`approaches have been reported for passive targeting of tumors using nanopartides
`inclusion complexes In addition innovative
`liposomes emulsions micelles implants pastes and gels All approaches for delivery of improved therapeutic
`nanosuspensions
`outcome have been discussed in this paper
`
`1 Introduction
`
`Paclitaxel Taxol
`
`ive antineoplastic
`
`is a pseudoalkaloid
`
`is one of the most widely used and effect
`derived from natural sources It
`agents
`that has a taxane ring as its nucleus
`C47H51N014 MW 85393 1 Figure 1 Its identification
`was the result of screening 35000 plant species for antitumor
`activity by the US National Cancer Institute NCI in 1958
`In 1963 Dr Monroe Wall and Dr M C Wani of Research
`Triangle Institute North Carolina demonstrated
`the
`extract of the inner bark of Pacific Yew Taxus brevifolia
`family Taxaceae had antitumor activity Although the pure
`form of the drug was isolated in 1969 it was not until 1971
`it was identified as paclitaxel and the structure published
`that
`
`that
`
`2 Paclitaxel has a wide spectrum of antitumor activity
`
`particularly against ovarian cancer breast cancer nonsmall
`lung cancer head and neck tumors Kaposis sarcoma
`cell
`
`and urologic malignancies 3 For instance researchers at
`the Johns Hopkins Oncology Center have reported a 30
`ovarian cancer and
`response rate in patients with advanced
`
`who were previously treated with high dose chemotherapy
`4 5 The response rate in patients with metastatic breast
`cancer was found to be 56 4 6
`to the nuclear mechanism of action of a
`In contrast
`majority of other cytotoxic drugs which act by attacking
`such as DNA and RNA paclitaxel
`the nuclear components
`acts by a unique mechanism of action The main site of
`however unlike
`is the microtubules
`action of paclitaxel
`the vinca alkaloids which induce depolymerization of the
`microtubules paclitaxel acts during the mitotic phase of cell
`division and promotes the polymerization of the tubulin pro
`teins and their assembly thereby stabilizing the microtubules
`and making them dysfunctional while arresting cell growth
`inhibits angiogenesis
`related cell migration proliferation and collagenase
`secre
`and
`
`cytotoxic properties contributes
`anticancer
`
`to paclitaxels efficacy as an
`
`7 It has been shown that paclitaxel
`tion 8 The combination of both the antiangiogenic
`agent 9 In spite of the remarkable prospect of
`
`as an antitumor drug it
`to formulate
`is difficult
`paclitaxel
`owing to its extreme lipophilic nature The formulation and
`
`Abraxis EX2054
`Actavis LLC v Abraxis Bioscience LLC
`1PR201701101 1PR201701103 1PR201701104
`
`

`

`2
`
`ISRN Pharmacology
`
`OH
`
`FIGURE 1 Structure of Paclitaxel 2a4a5p7p10p13 a 410bisacetyloxy13
`oyl oxy 17dihydroxy9oxo520epoxytax11en2y1
`benzoate
`
`2R3S3 benzoylamino2hydroxy3phenylpropan
`
`10
`
`6
`
`5
`
`8
`
`3
`
`1 21
`
`FIGURE 2 Taxane ring
`
`clinical problems related to paclitaxels solubility
`toxicity
`and development of drug resistance have led to innovative
`of alternative dosage
`research leading to the development
`forms that can overcome the above mentioned problems
`to discuss the challenges and various
`intends
`This paper
`related aspects of the formulation of paclitaxel
`
`2 The Paclitaxel Molecule and Its
`Delivery Challenges
`
`from its natural source Taxus brevi
`Extraction of paclitaxel
`folia causes the death of several plants to yield a few grams
`of the drug which is not a practical option A completely
`synthetic method of production of paclitaxel has not yet been
`developed due to the challenges presented by its structural
`complexity 1012 Current industrial production of pacli
`is done by plant cell fermentation technology whereby
`line and
`is extracted from a cultured
`Taxus cell
`
`taxel
`
`paclitaxel
`purified by chromatography
`is a diterpenoid centered around a bulky com
`and fused taxane ring Figure 2 composed
`of a
`plex
`number of hydrophobic substituents which makes it a highly
`lipophilic compound with a log P value of approximately 4
`and aqueous solubility of less than 001 mgmL 1315
`The compound lacks potentially ionizable functional groups
`which could lead to an increase in its solubility with alter
`ation in pH Among nonaqueous solvents the solubility is
`found to be 46 mM in ethanol 20 mM in methylene
`chloride or acetonitrile and 14 mM in isopropanol
`tertiarybutanol and dimethyl sul
`also soluble in methanol
`foxide 4 16
`The common approaches to increase aqueous solubility
`such as formation of salts and addition of charged com
`feasible options for paclitaxel 17
`pledng agents are not
`Synthesis of prodrugs to improve the aqueous solubility
`
`Paclitaxel
`
`It
`
`is
`
`has been attempted The 2 position on the
`of paclitaxel
`structure of paclitaxel may be the ideal position for the inser
`tion of functional groups to create prodrugs of paclitaxel
`because several derivatives of 2 acylpaclitaxel
`can quickly
`in the blood 18 A C7
`hydrolyze back into paclitaxel
`prodrug ester of paclitaxel has also been prepared because the
`arrangement of the C7 hydroxyl group does not appear to
`in influencing cytotoxicity 19 A paclitaxel ester
`be a factor
`in the presence of a strong electron withdrawing substituent
`the ester was
`like an alkoxy group in the a position of
`synthesized by Nicolaou et al to facilitate the quickening
`of the hydrolytic cleavage 20 These prodrugs have shown
`cytotoxic activity comparable to paclitaxel against cancer cell
`formulated using PEG
`lines in vitro Prodrugs of paclitaxel
`has been a promising approach because PEG imparts supe
`rior aqueous solubility 21 22 Nevertheless
`the pursuit of
`designing prodrugs of paclitaxel remains primarily academic
`focused on the design of water
`as industrial efforts have
`soluble derivatives or structural analogues of paclitaxel
`
`3 Commercially Available Dosage
`Forms of Paclitaxel
`
`The formulation of paclitaxel most widely used in the clinical
`setting is the solubilized form of
`the drug brand and
`generic which is diluted before intravenous administration
`castor oil Cre
`The formulation contains polyoxyethylated
`mophor EL in a 1 1 vv mixture with dehydrated ethanol a
`powerful solubilizer combo It
`remains stable in unopened
`vials for 5 years at 4°C 4 Polyoxyethylated
`castor oil
`
`is
`
`also used in the formulation of various other hydrophobic
`agents such as teniposide echinomycin and di
`anticancer
`demnin B 23 However
`is associated with side
`this excipient
`effects such as bronchospasms hypotension and symptoms
`of hypersensitivity 24 25 The constituents of polyoxyethy
`and cause
`induce histamine release
`lated castor oil can
`and hypersensitivity reactions following rapid
`hypotension
`administration of the infusion or as a result of an IV bolus
`administration 26 27 Therefore paclitaxel
`formulations
`must be slowly infused over a period of several hours to
`minimize the intensity and frequency of side effects The
`hypersensitivity reactions were evident at almost every phase
`
`

`

`ISRN Pharmacology
`
`3
`
`testing there
`in development in both preclinical and clinical
`is recommended
`that patients be premedicated with
`fore it
`and antihistamines prior to drug infusion to
`corticosteroids
`prevent or minimize the anticipated reactions 26 28 29
`The commercially available paclitaxel solution formulation
`is diluted 520 fold in normal saline or 5 dextrose solution
`
`for administration as an intravenous
`
`infusion therefore
`the formulation after dilution is a concern
`stability of
`of 03
`The diluted formulation results in a concentration
`12 mgmL of drug which is considerably higher than the
`001 mgmL aqueous solubility of paclitaxel posing the risk
`of drug precipitation upon dilution 4 25
`
`4 Nanopartides and Nanosuspensions
`
`tional
`
`To decrease
`associated with the conven
`the toxic effects
`formulation described above and minimize the risk
`of precipitation of paclitaxel upon dilution a nanoparticle
`formulation for paclitaxel was introduced in 2005 under
`the
`for particle forma
`trade name Abraxane The technology
`tion involves a proprietary process that binds unmodified
`albumin to the paclitaxel molecule yielding conjugate masses
`of 130 nm size Following infusion these nanoparticles
`rapidly dissociate to yield an albumin bound drug complex
`Albumin paclitaxel molecules bind to an albumin receptor
`gp60 on endothelial cells which transports paclitaxel
`into
`via the formation caveolae 30
`the extravascular
`space
`An alternate transport pathway
`is considered to be via
`binding of
`the nanoparticles with secreted protein acidic
`rich in cysteine SPARC Since SPARC is overexpressed in
`many solid tumors including bladder and prostate cancers
`leads to a 33 increase in intratumoral
`the nabpaclitaxel
`and a 50 higher dose of paclitaxel delivered
`concentrations
`infusion Moreover
`compared with a conventional
`paclitaxel
`the nab paclitaxel being solvent
`free the infusion time is
`much shorter compared to paclitaxel with Cremophor EL
`31 Ibrahim et al reported on the pharmacokinetics
`of
`Abraxane known as ABI007 during development 32
`Among alternative
`experimental delivery systems of
`paclitaxel nanoparticles of various biodegradable polymers
`and bioadhesive materials have been considered as promis
`can help to improve the bioavailability
`ing Nanotechnology
`of poorly soluble drugs such as paclitaxel enhancing drug
`delivery 33 Nanoparticles
`also have the ability to permeate
`through certain tissues adding to their drug targeting poten
`the drug to the targeted tissue efficiently
`tial They deliver
`without clogging the capillaries while protecting the drugs
`bioactivity and stability 14 3436 Upon incorporation
`of paclitaxel
`into nanoparticles there is a demonstrable
`enhancement
`due to changes
`in drug action
`in tissue
`15 37 Additionally it
`distribution and pharmacokinetics
`has been shown that nanoparticles
`can avoid rapid clearance
`system and can preferentially
`by the reticuloendothelial
`accumulate in solid tumors by escaping the angiogenic vas
`culature permeating the neoplasm 3840 Studies indicate
`carriers result in an
`that drugs delivered in nanoparticle
`extended retention of the drug in the tumors diminution
`in tumor growth and prolonged survival of the test subject
`34 41 42 The multidrug resistance phenotype mediated
`
`100
`
`v
`
`75
`
`T
`
`Al
`
`5
`
`25
`
`0
`
`24
`
`72
`
`120
`
`168
`
`216
`
`Time hours
`
`from PLGA
`FIGURE 3 Cumulative in vitro release of paclitaxel
`Nps Resomer RG 502 PLGANps containing paclitaxel 1 ww
`
`at 37°C and shaken horizontally
`were diluted in PBS incubated
`At preselected time intervals the released drug was separated by
`ultracentrifugation and the residual amount of paclitaxel
`in the nanospheres was determined by HPLC reproduced with
`permission from Fonseca et al 33
`
`present
`
`at
`
`by pglycoprotein in the tumor cells can also be overcome
`drug delivery This is important because
`by nanoparticle
`acquired resistance to paclitaxel has been reported 4345
`An additional advantage
`is their enhanced
`of nanoparticles
`stability both during storage and in biological fluids 46
`Fonseca et al have prepared nanoparticles of paclitaxel by
`using the interfacial deposition method in which they added
`an organic solution of PLGA and paclitaxel
`in acetone
`to
`an aqueous poloxamer 188 solution under magnetic stirring
`room temperature followed by washing
`and harvest
`The in vitro
`ing of nanoparticles
`by ultracentrifugation
`studies were conducted by measuring the residual
`release
`amount of paclitaxel at specific time points after the PLGA
`containing paclitaxel were diluted in PBS and
`nanoparticles
`at 37°C in a horizontal shaker The particles
`incubated
`exhibited a biphasic pattern for paclitaxel
`release during the first
`by an early fast
`release Figure 3 Two different
`by a slower and constant
`copolymers were studied and the in vitro antitumoral
`activity of the drug loaded nanoparticles
`was determined
`by the colorimetric MTT assay after
`incubating
`with paclitaxelloaded nanoparticles
`for various time periods
`Figure 4 33
`Mu and Feng investigated
`the use of doctocopheryl
`succinate vitamin E TPGS or
`1000
`polyethylene
`glycol
`TPGS as a novel emulsifier and matrix material for the
`preparation of nanoparticles 34 The nanoparticles
`loaded
`with paclitaxel were prepared by a modified oil in water
`single emulsion solvent evaporationextraction technique
`The size of the particles ranged from 3001000 nm In vitro
`release from the nanoparticles was studied by suspending the
`in PBS and shaking the tubes hor
`drug loaded nanoparticles
`izontally At specific time points the tubes were centrifuged
`and the supernatant was collected for analysis using HPLC
`nanoparticles were resuspended in the buffer
`Precipitated
`and shaken When different
`types of PLGA were used as
`polymers with 003 wt TPGS as emulsifier an initial burst
`
`release illustrated
`24 hrs followed
`
`the cells
`
`

`

`ISRN Pharmacology
`
`being suitable for parenteral administration The suspension
`form permits targeting of paclitaxel
`to organs like
`dosage
`liver spleen lungs and lymphatic circulation to a certain
`extent by the use of particles of varying size and through
`administration through various routes 4 49 MeriskoLiv
`ersidge et al evaluated the process of wet milling in the for
`mulation of a nanocrystalline
`suspension of poorly soluble
`anticancer drugs including paclitaxel 24 The nanoparticle
`formulation containing 2 wv drug suspension and 1
`wv surfactant stabilizer Pluronic F127 for paclitaxel was
`wet milled on a low energy mill until
`size of
`a final
`less than 400 nm was achieved
`as evaluated by photon
`correlation spectroscopy These suspensions were suitable for
`intravenous bolus injection without any incidence of acute
`toxicity in vivo and the tumor
`regression expressed as the
`percentage of tumor weight was significant 24
`
`5 Liposome Formulations of Paditaxel
`
`from
`
`Liposomes
`lipoid vesicles
`are microscopic
`ranging
`250 A to >20 ium in diameter and offer
`a flexible platform
`to encapsulate both lipophilic and hydrophilic drugs The
`lipophilic drugs incorporate in the lipid bilayer while the
`hydrophilic drugs locate in the vesicle cavity Encapsulation
`in their pharma
`of drugs in liposomes causes a change
`cokinetic and pharmacodynamic
`properties resulting in a
`in toxicity and increase in the potency 4 50
`decrease
`Liposomes
`can be used for specific targeting of cytotoxic
`tissues but they can be rapidly cleared
`compounds
`to select
`by the mononuclear phagocytic system in the liver and spleen
`unless special modifications are made to the phospholipid
`surface 51 The major problems associated with liposomes
`are their limited longterm stability and difficulty in large
`scale manufacturing of sterile liposomes
`Crosasso et al have prepared and characterized sterically
`stabilized paclitaxelcontaining liposomes They compared
`conventional
`paclitaxelloaded liposomes with stealth or
`sterically stabilized
`long circulating paclitaxelcontaining
`these
`in the
`liposomes because
`can
`circulate
`liposomes
`periods The PEGylated liposomes
`blood for prolonged
`using paclitaxel with phospholipids and
`were prepared
`in the molar ratio of 1 30 mol drug mol lipid
`cholesterol
`The formulation containing
`egg yolk phosphatidylcholine
`PC and phosphatidylglycerol PG transesterified from egg
`PC in the ratio 9 1 and paclitaxel
`and lipids in the
`molar ratio of 1 30 was
`found to have
`an incorporation
`efficiency of 95 This formulation retained 90 of the drug
`content over a study period of 2 months in the hydrated
`at 4°C during which the liposomes were physically
`state
`stable Figure 6 51 Incorporation of more than 20
`the incorporation efficiency and
`of cholesterol
`decreased
`physical stability of the formulations The liver and spleen
`and PEGylated liposomes
`distributions of the conventional
`after extraction
`of
`paclitaxel were evaluated
`from the tissue using t butyl methyl ether Figures
`7a and 7b 51 Drug uptake
`and spleen
`in liver
`after 05 and 3 hrs
`was
`lower with PEGylated liposomes
`51
`
`containing
`
`paclitaxel
`
`24
`
`72
`
`120
`
`168
`
`0 25 µgmL
`0 25 tigmL
`025 tigmL
`
`Incubation time h
`x 0025 tigmL
`0 Drug free Nps
`
`a RG 502
`
`120
`
`80
`
`40
`
`0 A
`
`72
`
`4
`
`120
`
`80
`
`40
`
`77
`
`24
`
`72
`
`120
`
`168
`
`0 25 µgmL
`0 25 tigmL
`A6 025 ligmL
`
`Incubation time h
`x 0025 tigmL
`0 Drug free Nps
`
`b RG 755
`
`FIGURE 4 Viability of NCI H69 cells incubated with drug free Nps
`or PtxPLGANps prepared with the RG 502 copolymer a or with
`RG 755 b Cells were seeded into 96 well plates and incubated
`
`of both formulations for 24 72 120
`with different concentrations
`or 168 has described reproduced with permission from Fonseca et
`al 33
`
`release pattern was common to all
`formulations following
`which the release rate dropped to generate nearly constant
`release of drug up to one month Figure 5a 34 When
`TPGS was blended with PLGA in the nanoparticle matrix it
`became more hydrophilic and resulted in an increase in drug
`release rate with an increasing ratio of TPGS Figure 5b
`34
`Sharma et al reported the preparation of paclitaxel con
`crosslinked with
`
`taining polyvinylpyrrolidone nanoparticles
`
`effect
`
`N N methylene bisacrylamide MBA in inverse micro emul
`sion and determined their antitumor
`in murine
`melanoma The in vivo efficacy of these nanoparticles when
`measured using reduction in tumor volume and increased
`time as parameters was significantly greater com
`survival
`of free taxol 38 Si
`concentration
`pared to an equivalent
`Shen and Guofeng also prepared nanoparticles of paclitaxel
`using freeze drying and solvent extractionevaporation tech
`niques Their data indicated that short and saturated chains
`of phospholipids such as dipalmitoylphosphatidylcholine
`DPPC have good emulsifying effect 47
`Muller and Petes evaluated the process of highpressure
`homogenization for the formulation of nanosuspensions of
`poorly soluble drugs 48 Nanosuspension
`formulations of
`insoluble drugs such as paclitaxel offer
`
`the advantage
`
`of
`
`

`

`ISRN Pharmacology
`
`5
`
`50
`
`40
`
`30
`
`20
`
`10
`
`A E5X E7
`
`ID Es
`
`10
`
`20
`
`30
`
`40
`
`0
`
`10
`
`20
`
`30
`
`40
`
`Released time days
`
`Released time days
`
`a
`
`M1
`
`M2A M3
`
`b
`
`70
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`Qc`
`
`V
`
`r24
`
`80
`
`70
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`
`Releasedpaclitaxel
`
`80
`
`70
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`
`Releasedpaclitaxel
`
`10
`
`20
`
`30
`
`40
`
`10
`
`20
`
`30
`
`40
`
`Released time days
`
`E7
`
`E13
`
`e E14
`
`c
`
`9 E8
`
`EIS
`
`E16
`
`Released time days
`
`d
`
`prepared under various experiment parameters a E5 PLA E7 PLGA
`FIGURE 5 In vitro release curves of paclitaxel
`loaded nanoparticles
`75 25 E8 PLGA 50 50 b ratio for PLGATPGS M1 2 1 M2 1 1 M3 1 2 c PLGA 75 25 concentrationE70125
`E13 0188 E14 025 d PLGA 50 50 concentrationE8 0125 E15 0188 E16 025 reproduced with permission from Mu and Feng
`34
`
`liposome for
`Ceruti et al also compared conventional
`mulations of paclitaxel with stealth long circulating lipo
`somes The PEGylated liposomes were long circulating with
`an elimination halflife of nearly 50 hrs compared to less
`liposomes 52 The group
`than 10 hrs with conventional
`attempted to improve the solubilization and targeting of
`paclitaxel using watersoluble prodrugs and incorporated
`2 succinylpaclitaxel
`2 methylpyridinium acetate pacli
`taxel and 2mPEGpaclitaxel in liposomes The 2 succinyl
`liposomes had superior stability at 4°C at both pH
`58 or 74 for up to 6 days but at 37°C the percentage of
`released increased from 4 to 12 Figures 8a
`and 8b 52 The halflife of this liposomal prodrug was
`than the free drug liposome formulations and the
`cytotoxic activity was similar to that of the free drug 52
`Sharma and Straubinger prepared liposome formulations of
`
`longer
`
`paclitaxel
`
`paclitaxel
`
`paclitaxel using phospholipids in the molar ratio of 1 33
`with phosphatidylcholine PC and phosphatidylglycerol
`PG transestrified from egg PC in the ratio 9 1 The
`liposomes were stable for more than 2 months at 4°C
`and 1 month at 20°C The stability of these liposomes as
`a function of PG and storage temperature
`is shown
`Figure 9 53 The results are expressed as percentage of
`taxol concentration
`remaining in the liposomes at
`initial
`different time points The formulations were well tolerated
`in mice when given via intravenous IV and intraperitoneal
`IP bolus doses The maximum tolerated dose was found
`to be >200 mgkg for liposomal paclitaxel whereas it
`is only
`30 mgkg by IV and 50 mgkg by IP route for free paclitaxel
`53
`Sampedro et al prepared liposomes using mixtures of
`phospholipids such as Ldimyristoyl phosphatidylcholine
`
`

`

`ISRN Pharmacology
`
`05
`
`3
`
`24
`
`Time after injection hours
`a
`
`25
`
`20
`
`0
`
`t4 15
`
`ac
`
`10
`
`40
`
`35
`
`30
`
`o 25
`00
`
`w
`
`20
`
`15
`
`10
`
`5
`
`05
`
`3
`
`24
`
`Time after injection hours
`b
`FIGURE 7 a Biodistribution in Balbc mice in plasma white
`liver black and spleen grey 05 3 and 24 hrs after
`injection of
`liposomes Standard deviations were below
`conventional
`5 of the mean values reproduced with permission from Crosasso
`et al 51 b Biodistribution in Balbc mice in plasma white
`liver black and spleen grey 05 3 and 24 hrs after injection of
`liposomes Standard deviations were below 5
`PEGylated paclitaxel
`of the mean values reproduced with permission from Crosasso et
`al 51
`
`paclitaxel
`
`paclitaxel
`
`phase 58 Paclitaxel
`has
`limited
`solubility in the oil
`solubility in soybean and similar vegetable oils therefore it
`prevents the use of simple 0W emulsion for formulation
`57
`Tarr and Yalkowsky prepared an 0W emulsion of pacli
`taxel using triacetin as the internal phase It was found that
`is highly soluble in triacetin 75 mgmL Regard
`less the drug precipitates when the emulsion is diluted
`approximately nine times with 5 dextrose 59 Tarr et al
`prepared the first parenteral emulsion of paclitaxel contain
`in a vehicle composed of 50
`ing 1015 mgmL paclitaxel
`triacetin 15 soylecithin 15 pluronic F68 and 20
`ethyl oleate About 10 of glycerol was added to prevent
`creaming 60 Although this system was stable for a period
`of 6 months when stored at 4°C the amount of vehicle
`triacetin needed for delivering the therapeutic
`containing
`dose of paclitaxel was found to be toxic when administered
`activity was
`intravenously to mice and no antitumor
`reported 60
`
`120
`
`100
`
`80
`
`60
`
`40
`
`initial
`
`of
`
`Encapsulateddrug
`
`20 H
`
`6
`
`10
`
`15
`
`
`
`2020
`
`25
`
`Time days
`
`FIGURE 6 Release of paclitaxel
`from conventional
`and
`squares
`triangles liposomes in storage conditions at 4°C
`PEGylated
`solid symbols or in human plasma at 37°C outline symbols
`reproduced with permission from Crosasso et al 51
`
`glycerol DMPG
`DMPC and Ldimyristoylphosphatidyl
`with or without
`cholesterol by the standard evapora
`tionhydration method Combination of the two phospho
`lipids in the ratio of 73 and 9 1 with the addition of 5
`cholesterol ww conferred optimal results The cytotoxicity
`of these liposome formulations in L1210 cells was found
`than that of free paclitaxel 54
`to be significantly greater
`in both
`Bartoli et al compared
`the efficacy of paclitaxel
`and liposome formulations They tested solu
`nanoparticle
`in DMSO and Cremophor EL against
`tions of paclitaxel
`liposomal formulations both in vitro and in vivo in P388 and
`L1210 leukemia models and concluded that
`the liposome
`formulation of paclitaxel was superior both in vitro and in
`formulation was found to be
`vivo whereas the nanoparticle
`toxic mainly because of its composition 55 Straubinger
`and
`a series of
`et al developed
`liposome formulations
`the in vitro tests revealed that
`retained
`the liposomes
`growth inhibitory activity while showing a delay of tumor
`progression against colon 26 a taxolresistant murine tumor
`56
`
`6 Emulsion Systems
`
`aqueous phase
`
`An emulsion is a heterogenous
`system comprising of an
`dispersed in an oil phase or vice
`versa
`stabilized by an emulsifier 0W emulsion formulations for
`the delivery of anticancer
`drugs have generated interest
`that many approved emulsifiers
`but are limited by the fact
`hemolytic reactions 57 The drug should
`be
`produce
`incorporated completely into the dispersed phase to form
`a stable emulsion therefore the formulation of a drug as
`0W emulsion is possible only when the drug has adequate
`
`

`

`ISRN Pharmacology
`
`7
`
`100
`
`80 1
`
`60
`
`40
`
`20
`
`0
`
`14
`
`12
`
`Parlitaxelreleased
`
`0
`
`20
`
`40
`
`60
`
`80
`
`100
`
`120
`
`140
`
`160
`
`0
`
`5
`
`I
`
`10
`
`15
`
`20
`
`25
`
`30
`
`Time hours
`a
`
`Time hours
`b
`
`FIGURE 8 a Paclitaxel
`triangles pointing up and 2MPApaclitaxel
`release at 4°C from 2PEGpaclitaxel circles 2succinylpaclitaxel
`triangles pointing up and 2MPA
`triangles pointing down in PBS buffer pH 58 2PEGpaclitaxel squares 2succinylpaclitaxel
`triangles pointing down in PBS buffer pH 74 reproduced with permission from Ceruti et al 52 b Paclitaxel
`paclitaxel
`triangles pointing down and 2MPApaclitaxel hexagons
`in PBS buffer
`37°C from 2PEGpaclitaxel circles 2succinylpaclitaxel
`pH 74 2PEGpaclitaxel squares 2succinylpaclitaxel
`triangles pointing up and 2MPApaclitaxel diamonds in PBS buffer pH 58
`reproduced with permission from Ceruti et al 52
`
`release at
`
`t
`
`110
`
`100
`
`90
`
`80
`
`70
`
`60
`
`50
`
`a n
`
`30
`
`20
`
`10
`
`110
`
`100
`
`90
`
`80
`
`70
`
`initial
`
`of
`
`60
`
`°o
`
`50
`
`Taw
`
`40
`
`30
`
`20
`
`10
`
`0
`
`0
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`0
`
`5
`
`10
`
`15
`
`20
`
`25
`
`30
`
`35
`
`40
`
`Time days
`Time days
`b
`a
`FIGURE 9 In these formulations the PG PC ratio was varied open squares PC only filled squares PG PC 19 open circles PG PC
`3 7 filled circles PG PC 5 5 open triangles PG PC 73 filled triangles PG onlyreproduced with permission from Sharma and
`Straubinger 53
`
`Kan et al prepared 0W emulsions in which triacylglyc
`erols were used to solubilize paclitaxel
`incorporated with
`various emulsifiers 61 The antitumor activity was tested
`both in vitro and in vivo The solubility of paclitaxel
`oils such as tributylin tricaproin tricaprylin soybean oil
`corn oil cottonseed oil and mineral oil was determined
`
`in
`
`Table 1 61 Paclitaxel had poor solubility in Tricaprylin
`C80 with 8 carbons per hydrocarbon
`chain compared
`to the short chain triacylglycerols such as tributyrin C40
`and tricaproin C60 Triacetin C20 resulted in a high
`solubility of paclitaxel 75 mgmL while mineral oil
`led to
`structure The
`poor solubility because of its hydrocarbon
`
`

`

`8
`
`ISRN Pharmacology
`
`TABLE 1 Solubility of paclitaxel
`permission from Kan et al 61
`
`in various oils reproduced with
`
`Oil
`
`Tributyrin
`
`Tricaproin
`
`Tricaprylin
`
`Corn oil
`
`Soybean oil
`
`Cotton seed oil
`
`Mineral oil
`
`Solubility of paclitaxel mgg
`962
`
`903
`
`119
`
`023
`
`018
`
`014
`
`Not detected
`
`1H
`
`08 H
`
`06
`
`04
`
`E
`
`cn
`
`02 H
`
`OH
`
`0
`
`30
`
`40
`
`50
`
`60
`
`Time day
`
`FIGURE
`
`11 Survival
`
`ratio of ascitic
`
`tumor bearing mice
`represents the group treated with
`control
`represents untreated
`60 mgkg paclitaxel emulsion and
`represents the group treated
`with same quantity of paclitaxel
`free emulsion vehicle reproduced
`with permission from Kan et al 61
`
`El
`
`1
`
`4111111111
`
`1
`
`08
`
`06
`
`E
`
`04 H
`
`02 H
`
`OH
`
`01
`
`10
`
`100
`
`1000
`
`FIGURE
`
`Paciltaxel nM
`rate of HeLa S3 tumor cells exposing
`10 Survival
`emulsions and Diluent 12 with or without paclitaxel
`the formulated emulsion with paclitaxel
`represents Diluent 12
`in 1 mL 50 ethanol
`and 50 Cre
`containing 6 mg paclitaxel
`mophor EL respectively 0 represents emulsion without paclitaxel
`and El
`reproduced with
`represents Diluent 12 without paclitaxel
`permission from Kan et al 61
`
`to
`
`represents
`
`emulsions were prepared by dissolving egg phosphatidyl
`choline EPC Tween 80 and paclitaxel
`in the oil phase
`and mixing it with the aqueous phase containing 225 wv
`and extrusion through a
`of glycerol followed by sonication
`022 µm filter The concentration of paclitaxel
`in emulsion for
`50 inhibition IC50 of HeLa cells is approximately 30 nM
`in Diluent 12 6 mg of paclitaxel
`which is similar to paclitaxel
`in 1 mL of 50 50 ethanol Cremophor EL The survival of
`HeLa S3 cells exposed to the test emulsions and Diluent
`is shown in Figure 10 61
`12 with or without paclitaxel
`Ascitictumorbearing mice were injected with the paclitaxel
`emulsion 60 mgkg and the vehicle
`the vehicle did not
`increase the life span of the tumor bearing mice but
`the
`span significantly Figure 11
`emulsion increased the life
`61 Even though the IC50 values are similar the paclitaxel
`lifespan than the Diluent 12
`emulsion resulted in a longer
`formulation when given in equivalent
`doses of
`30 mgkg Figure 12 61
`
`paclitaxel
`
`0
`
`
`
`1010
`
`20
`
`30
`
`40
`
`50
`
`60
`
`Time day
`
`FIGURE 12 Comparison oftreatment of Diluent 12 and emulsion on
`represents untreated control A
`the ascitic tumor bearing mice El
`represents group treated with 30 mgkg emulsion and
`represents
`group treated with Diluent 12 reproduced with permission from
`Kan et al 611
`
`Constantinides
`
`et al developed an injectable emulsion
`formulation containing paclitaxel 810 mgmL using Vita
`min E as the oil phase 25 The mean droplet diameter
`and 99 cumulative particle size distribution was less than
`02 µm therefore the emulsion could be filter sterilized A
`in PEG
`preemulsion was prepared by adding octocopherol
`the surfactants TGPS and Pluronic F127
`400 containing
`with or without paclitaxel
`to degassed water for injection It
`
`

`

`ISRN Pharmacology
`
`9
`
`20
`
`18
`
`16
`
`60 14
`
`2 12
`
`10
`
`8
`
`8
`
`A 6
`
`4
`
`5
`
`10
`
`15
`
`t h
`
`120
`
`100
`
`e
`
`80
`
`60
`
`40
`
`v
`
`20
`
`CL7
`
`0
`
`0
`
`f5
`
`10
`
`100
`
`1000
`
`10000
`
`Concentration nM
`
`48 v 72
`FIGURE 14 Inhibition of HeLa cell proliferation by paclitaxel oleate
`in lipid emulsions Incubation times were 24
`and 96 h The effect of paclitaxel
`in Cremophor ELethanol at
`48 h A is shown as comparison Control growth = 100 Values are
`mean ± SD n = 4 reproduced with permission from Lundberg et
`al 64
`
`350
`
`14 300
`
`250
`
`t
`
`200
`
`2
`
`150
`
`100
`
`2
`
`50
`
`10
`
`20
`
`30
`
`40
`
`50
`
`Time hours
`
`FIGURE 15 Drug concentration
`versus time curves of 3Hpaclitaxel
`in Cremophor EL ethanol 1 1
`vv v and 3Hpaclitaxel
`in lipid emulsion
`3Hpaclitaxel
`oleate in lipid emulsion
`single iv bolus Values are mean ± SD n = 3 P < 005 versus
`from Lundberg et al 64
`
`paclitaxel
`
`in Cremophor ELethanol
`
`reproduced with permission
`
`following
`
`reconstituted with sterile water 63 In
`be successfully
`research the group incorporated a paclitaxel
`subsequent
`derivative which consisted of a paclitaxel oleate in a nanosize
`lipid drug carrier emulsion and evaluated its pharmacoki
`netic profile 64 The dose
`response curve in Figure 14
`shows that paclitaxel oleate is cytotoxic and the cytotoxic
`activity increases with an increase in incubation time 64
`The comparative pharmacokinetic
`profiles of paclitaxel
`lipid
`formulation and paclitaxel
`emulsion Cremophorethanol
`in an oleate emulsion are shown in Figure 15 64 It was
`of paclitaxel
`the plasma concentration
`suggested
`oleate emulsion was increased as a result of interaction with
`
`in the
`
`that
`
`lipoproteins
`
`FIGURE 13 Mean plasma concentration
`istration of paclitaxel
`injection
`
`0 in rats n = 5 reproduced with permission from He et al
`
`of paclitaxel after IV admin
`and paclitaxel microemulsion
`
`9 w
`
`as mixed vigorously at 45°C followed by homogenization
`and sterile filtration through a 02 pm Posidyne filter Drug
`from this emulsion was slower compared to Taxol
`both in the presence or absence of human serum albumin
`tolerated and more
`The tocopherol
`emulsion was better
`in the melanoma tumor model mice
`efficacious than Taxol
`
`release
`
`25
`He et al prepared and evaluated a microemulsion formu
`lation of paclitaxel with lower levels of solubilizers anticipat
`ing less hypersensitivity reactions 9 The hypersensitivity
`
`tests were done on guinea pigs While the Taxol vehicle led
`to frequent nose scratch tremble sneeze erect hair twitch
`or dyspnea with a grade of hypersensitivity equal to 3 the
`microemulsion vehicle resulte

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