throbber
Pharmaceutical Research, Vol. 13, No. 2, 1996
`
`Report
`
`Formulation and Antitumor Activity
`Evaluation of Nanocrystalline
`Suspensions of Poorly Soluble
`Anticancer Drugs
`
`E. Merisko-Liversidge,1.3 P. Sarpotdar,1 J. Bruno,1
`S. Haij,1 L. Wei,1 N. Peltier,1 J. Rake,1 J.M.
`Shaw,1 S. Pugh,2 L. Polin,2 J. Jones,2 T. Corbett,2
`E. Cooper,1 and G. G. Liversidge1
`
`Purpose. Determine if wet milling technology could be used to
`formulate water insoluble antitumor agents as stabilized nanocrystalline
`drug suspensions
`that retain biological effectiveness following
`intravenous injection.
`Methods. The versatility of the approach is demonstrated by evaluation
`of four poorly water soluble chemotherapeutic agents that exhibit
`diverse chemistries and mechanisms of action. The compounds selected
`were: piposulfan (alkylating agent), etoposide (topoisomerase II inhibi(cid:173)
`tor), camptothecin (topoisomerase I inhibitor) and paclitaxel (antimi(cid:173)
`totic agent). The agents were wet milled as a 2% w/v solids suspension
`containing I% w/v surfactant stabilizer using a low energy ball mill.
`The size , physical stability and efficacy of the nanocrystalline suspen(cid:173)
`sions were evaluated.
`Results. The data show the feasibility of formulating poorly water
`soluble anticancer agents as physically stable aqueous nanocrystalline
`suspensions. The suspensions are physically stable and efficacious
`following intravenous injection.
`Conclusions. Wet milling technology is a feasible approach for formu(cid:173)
`lating poorly water soluble chemotherapeutic agents that may offer a
`number of advantages over a more classical approach.
`KEY WORDS: anticancer agents; poorly water soluble agents; nano(cid:173)
`particles; etoposide; camptothecin; piposulfan and paclitaxel.
`
`INTRODUCTION
`
`Frequently in drug discovery poorly water soluble agents
`are identified as actives using in vitro assays but are discarded
`because they are unable to be formulated for further evaluation
`in vivo. Thus many promising agents are discarded due to
`poor water solubility and the lack of a generally applicable
`formulation approach to address this problem. Though this
`predicament is a likely scenario in any therapeutic/diagnostic
`discovery program it has been a recurring issue in cancer
`research. The development of a number of promising anticancer
`agents have been delayed or abandoned due to issues resulting
`from the poor water solubility of the drug (l).
`Currently, the poorly water soluble chemotherapeutic
`agents used in the clinic and that are in various phases of
`development are formulated by conventional methods. Rou(cid:173)
`tinely, these agents are formulated using a co-solvent plus other
`excipients which act to solubilize the drug and provide for
`stability of the formulation during storage and on injection (2).
`
`1 NanoSystems, Collegeville, Pennsylvania 19426.
`2 Division of Hematology and Oncology, Wayne State University,
`Detroit, Michigan 48202.
`3 To whom correspondence should be addressed.
`
`Commonly, the use of co-solvents produce toxic side effects
`e.g., anaphylaxis, pain at the site of injection, emboli formation
`and paradoxically precipitation which results in poor intrave(cid:173)
`nous bioavailability. For instance, etoposide (VP-16), a semi(cid:173)
`synthetic analog of podophy llotoxin, is a sparingly water soluble
`drug that is used to treat small-cell lung cancer and various
`other neoplasms (3,4). Etoposide being a poorly water soluble
`drug is formulated using benzyl alcohol, polyoxyethylated sor(cid:173)
`bitan ester (Tween 80) and polyethyleneglycol (PEG 300). Prior
`to injection the formulation is diluted then slowly infused. Even
`though etoposide has been used in the clinic for a number of
`years, formulation related toxicity issues still occur (5-7).
`Extensive literature is available on other technologies con(cid:173)
`cerned with delivery issues of hard to formulate chemotherapeu(cid:173)
`tic agents. Although significant progress has been made in each
`of these areas including liposomes (8,9), emulsions (10,11)
`and polymeric carriers (12,13), there is a need for additional
`methodologies that provide a safe, effective and economical
`means for intravenous administration of poorly water soluble
`therapeutics and/or diagnostics.
`In this study, a delivery system well suited for sparingly
`water soluble chemotherapeutic agents is described wherein the
`compounds are formulated as dispersible particles consisting
`essentially of a crystalline drug substance stabilized with a
`surface modifier. The methods of preparation and characteriza(cid:173)
`tion of nanocrystalline dosage forms suitable for intravenous
`administration of poorly water soluble chemotherapeutic agents
`are presented together with preclinical efficacy data performed
`in the mammary 16C murine tumor model.
`
`MATERIALS AND METHODS
`
`Chemicals
`
`With the exception of piposulfan which was custom syn(cid:173)
`thesized at Kodak Research Laboratories, Rochester, NY.,
`agents were obtained from the following vendors: etoposide
`(Sigma Chemical Co, St. Louis, MO), camptothecin (Aldrich,
`Milwaukee, WI) and paclitaxel (Biolyse Corporation, Port-Dan(cid:173)
`iel, Que.). Pluronic and Tetronic surfactant stabilizers were
`obtained from BASF (Parsnippany, NJ.) and the polyoxyethy(cid:173)
`lated sorbitan esters were purchased from ICI, Wilmington, DE.
`
`Nanoparticle Formulations
`
`Nanoparticle formulations were prepared under aseptic
`conditions as a drug suspension (2% wt/v) containing 1 % (wt/v)
`surfactant stabilizer ( 14, 15). An aqueous suspension containing
`drug and stabilizer was wet milled using preconditioned zirco(cid:173)
`nium oxide media (Zircoa Inc., Solon, OH.) For screening,
`milling was performed in a 28 ml bottle using a media bead
`volume of 7.5 ml and 3.75 ml of the drug/surfactant slurry.
`The slurry was milled on a low energy mill (U.S. Stoneware,
`East Palestine, OH) at 57% of the critical speed. The critical
`speed is defined as the rotational speed of the grinding vessel
`when centrifuging of the grinding media occurs. Milling effi(cid:173)
`ciency was dependent on a number of factors including drug
`substance and choice of stabilizer. The particle size of the slurry
`was assayed daily during milling. Routinely, the drug/surfactant
`slurry was milled to a final size of less than 400 nm based on
`
`272
`
`

`

`Nanosuspensions of Anticancer Drugs
`
`273
`
`photon correlation spectroscopy (PCS) and generally this could
`be achieved in a 4 day period. Milled nanosuspensions were
`evaluated for chemical stability, physical stability, and physical
`stability in plasma. Formulations with acceptable physical sta(cid:173)
`bility in plasma were submitted for efficacy studies. Acceptable
`physical stability was defined as absence of agglomeration or
`negligible particle size growth in the presence of plasma when
`the nanosuspension was incubated with plasma for 60 min
`at 37°C.
`The stabilizers used for each of the formulations were:
`1) polyoxyethylene sorbitan monooleate (Tween 80)1sorbitan
`monooleate (Span 80) for piposulfan; 2) Pluronic F108 for
`camptothecin, and 3) Pluronic F127 for etoposide and paclitaxel
`(Table 1).
`
`Particle Size Analysis
`
`Particle size analysis was determined using photon correla(cid:173)
`tion spectroscopy (PCS). Prior to sizing, samples were diluted
`with freshly filtered deionized water. Sizing measurements were
`routinely performed using the Coulter Model N4MD Submicron
`Particle Analyzer (Coulter, Miami, Fl.) or the Malvern Zetamas(cid:173)
`ter (Malvern Instruments Ltd., Worcester, England) using uni(cid:173)
`modal analysis of intensity distributions for mean particle size·
`determination. PCS results were confirmed using scanning elec(cid:173)
`tron microscopy (SEM). For SEM, samples were diluted and
`an aliquot of the diluted preparation was dried, sputter coated
`and visualized using the Topco SM510 (Topcon Technologies,
`Inc., Pleasanton, Ca.).
`
`Plasma Stability Assays
`
`Stability of nanoparticle suspensions in plasma was moni(cid:173)
`tored by photon correlation spectroscopy (PCS) and light
`microscopy. Rat plasma was delipidated prior to use by centrifu(cid:173)
`gation at 150,000 X g for lhr. The supernatant was carefully
`decanted and filtered through a 0.1 micron filter. The plasma
`was then pretested in the Coulter N4MD submicron particle
`analyzer to ensure that the sample was particle-free. Nanosus~
`pensions of piposulfan, camptothecin, etoposide and paclitaxel
`were diluted 1:2, 1:10, and 1:100 with lipid-free plasma. Sam(cid:173)
`ples were vortexed and incubated at 37°C. For PCS analysis,
`samples were diluted with water and assayed. Presence or
`absence of aggregation was also monitored using the Leica
`DMRB optical microscope with a 100 X phase-contrast
`objective.
`
`Animal Studies
`Studies were performed in accordance with Wayne State
`University Medical School's animal use and care administrative
`
`policies. As previously described (16), tumor fragments (-30
`mg) were seeded subcutaneously by trocar into 7-10 week-old
`female C3H mice (National Institute of Health, Bethesda, Md.).
`Chemotherapy of either a nanoparticle suspension or a control
`formulation was initiated within 6 days of tumor implantation.
`For chemotherapy, animals were injected via the tail vein using
`a multiple injection regimen. Dose and dosing schedule were
`selected based on the toxicity of the compound with the inten(cid:173)
`tion of administering the drug at its maximum tolerated dose
`(MTD).
`To assess antitumor effectiveness, median tumor burden
`of treated animals was compared to median tumor burden of
`untreated controls. Results are expressed as a percentage deter(cid:173)
`mined by comparing the average weight of tumors in treated
`animals (expressed as "T") to the average weight of tumors in
`untreated controls (expressed as "C"). This value is expressed
`as percent TIC. A TIC = 0% is indicative of a highly active
`agent while a TIC >42% is considered inactive (16).
`
`RESULTS
`
`Particle Size Reduction
`
`Figure 1 shows the particle size reduction profile achieved
`during ball milling. Generally, within the first 24 h a significant
`reduction in particle size is observed. Depending on the drug
`core and stabilizer utilized, additional milling rendered finer
`particle dispersions. For this study, nanosuspensions were
`milled for 4 days and particle size analysis was performed every
`12 hrs. After four days of milling nanosuspensions of piposulfan
`and etoposide with a mean diameter -200 nm were harvested.
`For camptothecin and paclitaxel, milling was continued for an
`additional 3 to 4 days to further reduce particle size. As shown
`in Table 1, routinely nanosuspensions were prepared with a
`mean particle size that averaged 200-250 nm for all four
`agents studied.
`
`Characterization of Nanoparticle Drug Suspensions
`
`In Figure 2, the particle size distribution for nano-piposul(cid:173)
`fan, nano-etoposide, nano-camptothecin and nano-paclitaxel is
`shown. Intensity distributions were obtained using photon corre(cid:173)
`lation spectroscopy (PCS). Different drug cores with an optimal
`stabilizer produce a relatively uniform dispersion. The gross
`uniformity or homogeneity of the dispersions was verified using
`scanning electron microscopy (SEM). In Figure 3, the scanning
`electron micrographs show representative images of the nano(cid:173)
`particle drug suspensions. The milled nanosuspensions for each
`agent were homogeneous. However, the morphology of the
`
`Table 1. Average Particle Size: Nano-Suspensions•
`
`Compound (solubility)h
`
`Piposulfan (125µg/ml)
`Camptothecin (50µg/ml)
`Etoposide (200µg/ml)
`Paclitaxel (< 10 µg/ml)
`
`Formulation
`
`Average Size in nm. (number of batches)
`
`2% Piposulfan, 0.33% Tween 80, 0.67% Span 80
`2% Camptothecin, 1% F108
`2% Etoposide, 1% F127
`2% Paclitaxel, 1 % F127
`
`210.2 ± 38.9 (8)
`202.3 ± 30.5 (6)
`256.2 ± 53.0 (12)
`279.2 ± 29.60 (7)
`
`• Chemotherapeutic agents were roller milled as a 2% wt/v suspension using surfactant satbilizers at a core to surfactant ratio of 2: 1. Samples
`were sized using photon correlation spectroscopy (PCS). The average size was determined using the said number of batches.
`b Aqueous solubility @ 25°C.
`
`

`

`274
`
`Merisko-Liversidge et al.
`
`1000
`
`'i
`Po.
`i
`....
`..
`.
`''o...
`\....
`-0-..--o ••
`···-~:....
`...........
`··0.---0---0----0
`.. ......
`····•·:·;·;::a:········ .. ····•
`.........
`
`....
`
`:=-
`·;;;
`i::
`B
`..s
`!:I<
`
`7.5
`
`5
`
`2.5
`
`0 0.5
`
`1.5
`
`2
`
`2.5
`
`3
`
`3.5 4
`
`600
`
`500
`
`400
`
`300
`
`200
`
`.......
`...........
`'a.
`
`. . . . . .......
`.........
`.
`o .......... .
`··o. ... ...
`.... •··.
`··-."(). ............ '()-............ 0
`............ :::·. ........ , ..........
`···-.. :~:~~:·:~:·:·:·:·:·• ............. .
`.....
`
`1.5
`
`2
`
`2.5
`
`3
`
`3.5
`
`4
`
`Milling Time {Days)
`Fig. 1. Effectiveness of particle size reduction using wet milling tech(cid:173)
`nology to formulate water insoluble chemotherapeutic agents. Com(cid:173)
`(piposulfan-D; · camptothecin-+ ; etoposide__.; and
`pounds
`paclitaxel-0) were roller milled using the surfactant stabilizers
`described in Table 1. During milling, particle size was monitored every
`12 hrs. For particle size analysis samples were diluted with deionized
`water, vortexed and sized using photon correlation spectroscopy (PCS).
`Data show weight average diameters of particles in suspension.
`
`various nanosuspensions differed from the cuboidal appearance
`of nanocamptothecin (Fig. 3B) to the rather elongated rod-like
`structures of nano-paclitaxel (Fig. 3D). Also, as monitored by
`both PCS and SEM, heterogeneity with respects to particle
`size and shape appears to be primarily governed by interactive
`properties of both the drug core and the surfactant stabilizer
`used during milling. When optimally stabilized, the suspensions
`did not aggregate and remained physically stable for at least
`four weeks post preparation (Fig. 4).
`Since the intended use of these nanosuspensions is for
`intravenous administration, physical stability in the presence
`of plasma was monitored using particle size analysis and optical
`microscopy. Nanosuspensions were diluted with plasma and
`incubated at 37°C . As shown in Figure 5, for camptothecin
`and paclitaxel no significant change could be detected in the
`mean particle size of the preparations throughout a 60 min
`incubation period. For piposulfan and etoposide, particle growth
`was observed but there was no evidence of particle aggregation
`and/or agglomeration based on both PCS sizing and optical
`
`0
`
`0
`0
`
`0
`0
`N
`
`0
`0
`0
`0
`~ V
`
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`~ ~ ~ ~ ~ 0
`
`Particle Diameter (nm)
`Fig. 2. Size distribution profiles of nanoparticle suspensions were
`obtained using the Malvern Submicron Particle Sizer. Samples were
`diluted with deionized water, vortexed for -20 sec and sized. Size
`distribution profiles and cumulant z averages are shown for: • nanopi(cid:173)
`posulfan (z Ave = 299.9); 0 nanocamptothecin (z Ave = 227.8; o
`nanoetoposide (z Ave = 155.4); and• nanopaclitaxel (z Ave = 291.3).
`
`microscopy. Physical stability of the nanocrystalline suspen(cid:173)
`sions was also not affected by time of incubation and dilution.
`Samples diluted 1:100 with plasma and incubated at 37°C for
`24 hrs remained physically stable.
`
`Efficacy of Nanoparticle Drug Suspensions
`
`To evaluate efficacy, the nanocrystalline drug suspensions
`were tested in mice previously injected with mammary adeno(cid:173)
`carcinoma (16/C). The nanosuspensions were compared with
`the conventional formulation of the same agent or an agent
`of similar mechanism of action. Results of these studies are
`illustrated in Table 2. For all nanoparticle suspensions evaluated,
`tumor regression expressed as the percentage of tumor weight
`in treated animals to tumor weight in untreated controls was
`significant. In addition, the suspensions were suitable for intra(cid:173)
`venous bolus injection as suggested without increased incidence
`of acute toxicity in comparison to controls. For certain drugs,
`such as piposulfan, only the nanoparticle formulation was toler(cid:173)
`ated as an i.v. injectable. Control formulations were extremely
`toxic unless administered subcutaneously.
`
`DISCUSSION
`
`A technology is described for formulating water insoluble
`anticancer agents as nanoparticle drug suspensions that are
`stable. The method is versatile and suitable for many agents
`. whose solubility is less than 200 ug/ml. The potential value of
`this approach was demonstrated using two well-known drugs,
`etoposide and paclitaxel, which are in the clinic but have report(cid:173)
`edly been associated with formulation-related safety issues (6-
`8,17-19). The other agents
`investigated, piposulfan and
`camptothecin, are older drug candidates that could not be suc(cid:173)
`cessfully formulated as an injectable. In the case of piposulfan
`
`

`

`Fig.· 3: Nanocrystalline drug suspensfons were analyzed using scanning electroll microscopy (SEM). Unmllled drug
`substance was visualized at 500 X magnification and is shown on the left side of the figure as: A) piposulfan; B)
`camptothecin; C) etoposide; and D) paclitaxel. The corresponding nanocrystalline suspensions (5,000 X) are shown on
`the right side of the figure labeled as: E) nanopiposulfan; F) nanocamptothecin; G) nanoetoposide; and H) nanopaclitaxel.
`
`

`

`l
`
`276
`
`e .s
`
`CD
`.!::!
`(JJ
`
`400
`
`350
`
`300
`
`250
`
`200
`
`150
`
`JOO
`
`50
`
`0
`
`Initial Size
`
`4 Weeks
`
`•
`
`Nanopiposulfan
`
`El Nanocamptothecin
`ISi Nanopaclitaxel
`Nanoetoposide
`•
`Fig. 4. Physical stability of nanocrystal suspensions were studied at
`ambient temperature for a four week period. The antitumor agents ·
`were roller milled as described in Table 1 and Figure 1. The suspensions
`were then stored at room temperature for four weeks and re-sized
`using PCS. The graph compares mean particle size of the preparations
`immediately after milling with the mean particle size of the suspension
`following storage at room temperature for one month.
`
`350
`
`300
`
`250
`
`200
`
`'E
`..s
`fJ u;
`GI u
`t:
`:.
`
`·····--o------()--~--------------------0
`
`'"·+·--···-........ _ .... _, __ ,..._. _____ .,,_, ....... _,., ....... .
`
`..
`
`150
`
`•··•••·••••••••••••
`...
`
`0
`
`20
`
`40
`
`60
`
`Time (minutes)
`Fig. 5. The physical stability of nanocrystal drug suspensions was
`studied in the presence of plasma. Nanopiposulfan-D, nanocampto(cid:173)
`thecin-+, nanoetoposide-11, and nanopaclitaxel-0 were diluted
`l :2 with plasma and incubated for 60 min at 37°C. For particle size
`analysis samples were diluted with deionized water and sized using
`PCS. PCS size analysis was confirmed using optical microscopy.
`
`Merisko-Liversidge et al.
`
`drug development was abandoned, whereas, solubility issues
`associated with the camptothecins are being aggressively pur(cid:173)
`sued via the identification of water soluble analogs (20-23).
`The solubility of the anticancer agents chosen for this study
`range from -200 ug/ml to less than 4.0 ug/ml (24,25) and as
`demonstrated in this study can be readily formulated as an
`aqueous suspension of fine particles using a low energy wet
`milling process .
`The nanoparticle suspensions of piposulfan, camptothecin,
`etoposide and paclitaxel that are described were generated using
`a conventional ball mill. As the data show, in the presence of
`the selected surfactant stabilizer(s) the procedure was effective
`in producing nanoparticles of pure drug substance. Generally,
`the higher molecular weight polymeric stabilizers were optimal
`for effective particle size reduction, shelf stability, and preven(cid:173)
`tion of agglomeration in the presence of blood proteins. For
`instance, stable suspensions -250 nm in diameter were obtained
`for camptothecin, etoposide and paclitaxel using the pluronic
`block co-polymers Fl 08 and Fl 27. The higher molecular weight
`Pluronics have been shown to be excellent stabilizers for various
`colloidal delivery systems (26). In addition, these surfactant
`coatings have been known to reduce opsonization of particulate
`drug carriers and enhance delivery of the desired agent to
`various anatomical targets which would be advantageous for
`passive delivery to solid tumors (27 ,28). The pharmacokinetic
`properties of these nanoparticle drug suspensions are being
`studied. However, since the technology described in this study
`is relatively new and the biodistributional properties of colloidal
`nanocarriers are dictated by a complexity of interactions, it
`would be surprising if the blood clearance of these nanosuspen(cid:173)
`sions is not rapid. Currently, methodology is being developed
`so that properties of the nanosuspensions, e.g. size, surface
`characteristics and shape can be modulated to optimize delivery.
`Though the effects of the comminution process on the
`physical state of the drug was not performed, previous studies
`using X-ray diffraction have shown that the dispersion process
`did not change the crystal structure of the compound (14). In
`this study the electron micrographs of the milled dispersions
`suggest that the process generates nanocrystalline drug particles.
`However, comparisons between pre and post processed materi(cid:173)
`als remains to be studied.
`For certain drugs such as piposulfan, generating a stable
`nanocrystalline suspension proved challenging. After screening
`a series of surfactants and surfactant combinations, the use of
`a Tween 80 and Span 80 mixture provided optimal physical
`stabilization. As shown in Figure 1 and 2, using this surfactant
`combination, particle size of the suspension was reduced to
`250 nm in -4 day period. For piposulfan the su1factant mixture
`adequately wets the drug substance and provides steric stabiliza(cid:173)
`tion. However, as judged from the plasma stability data shown
`in Figure 5, though the preparation does not agglomerate in
`plasma, the particles apparently interact with plasma proteins
`resulting in an overall increase in mean particle size of the
`preparation. This interaction does not appear to compromise
`the safety and efficacy of the suspension (Table 2).
`As shown in Table 2, the efficacy of oncologic agents when
`formulated as nanoparticles was satisfactory. For etoposide and
`paclitaxel, novel nanoparticle suspensions were compared to
`currently used clinical formulations. Etoposide is formulated
`using polyethylene glycol, Tween 80 and benzyl alcohol while
`paclitaxel is dissolved in a mixture of Cremophor EL and
`ethanol. To avoid acute toxicity, both formulations must be
`
`

`

`Nanosuspensions of Anticancer Drugs
`
`Table II. Efficacy of Nanoparticle Suspensions Against Early Stage Mammary Adenocarcinoma 16C in CH3 Mice•
`
`Formulation
`
`NanoPiposulfan
`Comparator I
`No Treatment
`NanoCamptothecin
`Comparator II
`
`No Treatment
`NanoEtoposide
`Comparator III
`
`No Treatment
`NanoPaclitaxel
`Comparator IV
`No Treatment
`
`Schedule
`
`BID 1-4
`BID 1-4
`NA
`d 1-3
`d 1-3
`d 1-3
`NA
`d 1,5
`d 1,3,5
`d 1,3,5
`d 1,3,5
`NA
`d 3.6
`BID 3,4,5
`NA
`
`Total Dose
`(mg/kg)
`
`Drug Deaths
`(#deaths/total)
`
`Median Tumor Burden
`(mg)
`
`356
`360
`NA
`40
`20
`12.5
`NA
`125
`102
`69
`45
`NA
`88
`100
`NA
`
`015
`2/5
`015
`015
`515
`1/5
`015
`015
`515
`4/5
`0115
`015
`015
`315
`015
`
`0 (0-234)
`32 (0-63)
`3582 (1352-4678)
`907 (284-2514)
`NA
`969 (352-1224)
`2576 (627-3054)
`0 (0-88)
`NA
`75 (75)
`171 (0-260)
`1387 (0-2016)
`53 90-196)
`158 (138-839)
`2674 (0-3043)
`
`277
`
`TIC
`%
`
`0
`1.0
`NA
`35
`NA
`38
`NA
`0
`NA
`5
`12
`NA
`3
`7
`NA
`
`• Formualtions were injected intravenously using the stated schedule and dosage. Comparator formulations were: a) Comparator I: cyclophosamide
`in saline; 2) Comparator II: camptothecin in 5% ethanol, 2% POE40; c) Comparator III: etoposide in 7% ethanol, 3% POE40 and d) Comparator
`IV: paclitaxel in 50% ethanol, 50% Cremophor. Once a day and twice daily injections are denoted by "d" and "BID" respectively.
`
`diluted prior to use and slowly infused. In the present study,
`these formulations were slowly injected, whereas, nanoparticu(cid:173)
`late suspensions were administered via a rapid bolus injection.
`The data show that in the mammary 16C murine tumor model,
`the performance of nano-etoposide and nano-paclitaxel was
`comparable or slightly improved over the response elicited by
`the conventional formulations. In addition, the data clearly show
`that when administered as a fine particle preparation, dosages
`could be increased without incidence of acute toxicity, abnormal
`weight loss, or organ pathology observed during necropsy. Since
`the surfactants used in the nanocrystal formulations were
`selected so as to prevent agglomeration in the presence of
`plasma, the improved acute safety profile of the suspension
`was anticipated. However, other factors such as an altered phar(cid:173)
`macokinetic profile should be considered.
`Our data suggest that the ability to formulate water insolu(cid:173)
`ble chemotherapeutic agents as nanocrystal drug suspensions
`may offer a number of advantages over a more classical
`approach. First, since these fine particle drug suspensions are
`well tolerated following intravenous injection, nanoparticles
`provide a convenient remedy for administering high doses of
`drug without the risks routinely associated with conventional
`formulations containing cosolvents. Second, the potential for
`being able to safely increase the dosage of a particular drug
`may lead to the identification of new medical indications and
`usages for the given drug. Lastly, the technology described in
`this study provides a convenient platform for targeted delivery
`of water insoluble therapeutic and/or diagnostic agents.
`
`REFERENCES
`
`1. J. R. Robinson. Recent advances in formulation of poorly absorbed
`drugs. In: Current Status on Targeted Drug Delivery to the gastro(cid:173)
`intestinal tract, pp. 59--63. Capsugel Library, ( 1993 ).
`2. J. G. Nairn. Solutions, Emulsions, Suspensions and Extracts. In:
`A. R. Gennaro (ed.), Remington's Pharmaceutical Sciences,
`pp.1519-1544. Easton: Mack Publishing Co. (1990).
`3. L. M. Slevin. The clinical pharmacology of etoposide. Cancer,
`67:319-329 (1993).
`
`4. P. Rubin (ed.) Clinical Oncology. Philadelphia: W. B. Saunders
`(1993).
`5. D. J. O'Dwyer and R. B. Weiss. Hypersensitivity reactions
`induced by etoposide. Cancer Res, 68:959-961 (1984).
`6. J.M. Bennett, G. H. Lymann, P.A. Cassileth, J. H. Glick, M. M.
`Okem. A phase II trial of VP-16213 in adults with acute myeloid
`Leukemia. Am. J. Clinic. Oncol. Cancer Clinical Trials, 7:471-
`473 (1984).
`7. M. M. Hudson, H.J. Weinstein, S.S. Donaldson, C. Greenwald,
`L. Kun, N. J. Tarbell, W. A. Humphrey, C. Rupp, N. M. Marina,
`J. Wilimas, M. P. Link. Acute hypersensitivity reactions to etopo(cid:173)
`side in a VEPA regimen for Hodgkin's disease. J. Clin. Oncol.,
`11:1080-1084 (1993).
`8. T. M. Allen. Long circulating (sterically stabilized) liposomes for
`targeted drug delivery. TIPS, 15:215-220 (1994).
`9. D. Lasic. Applications of liposomes in pharmacology and medi(cid:173)
`cine. In: Liposomes from physics to applications, pp. 262-471.
`Amsterdam: Elsevier Press (1993).
`10. S. S. Davis, C. Washington, P. West, L. Ilium, G. G. Liversidge,
`L. Stemson, R. Kirsh. Lipid emulsions as drug delivery systems.
`Ann N.Y. Acad. Sci. 507:76-78 (1987b).
`11. B. Sjostrom, B. Bergenstahl, B. Kronberg. A method for the
`preparation of submicron particles of sparingly water-soluble
`drugs by precipitation in oil-in-water emulsions. II: Influence of
`the emulsifier, the solvent, and the drug substance. J. Pharm. Sci.
`82:584-589 (1993).
`12. P. Couvreur, L. Roblot-Treupel, M. F. Poupon, F. Brasseur, F.
`Puisieux. Nanoparticles as microcarriers for anticancer drugs.
`Adv. Drug Delivery Rev., 5:209-230 ( l 990).
`13. R. H. Muller. In: Colloidal carriers for controlled drug delivery
`and targeting. Stuttgart: Wissenschaftliche ( 1991 ).
`14. G. G. Liversidge and K. C. Cundy. Particle size reduction for
`improvement of oral bioavailability of hyrophobic drugs: I. Abso(cid:173)
`lute oral bioavailability of nanocrystalline danazol in beagle dogs.
`Int J. Pharm (1995, in press).
`15. N. Kondo,T. Iwao, H. Masuda, K. Yamamouchi, Y. Ishihara,
`N. Yamada, T. Haga, Y. Ogawa, K. Yokoyama. Improved Oral
`Absorption of a Poorly Water-Soluble Drug, H0-221, by Wet(cid:173)
`Bead Milling Producing Particles in Submicron Region. Chem.
`Pharm. Bull. 41:737-740 (1993).
`16. T. H. Corbett, D. P. Griswold, B. J. Roberts, J.C. Peckham, F. M.
`Schable. Biology and therapeutic response of a mouse mammary
`adenocarcinoma (16/C) and its potential as a model for surgical
`adjuvant chemotherapy. Cancer Treat. Reports, 62:1471-1488
`(1978).
`17. J. L. Grem, K. D. Tutsch, K. J. Simon, D. B. Alberti, J. K. V.
`
`

`

`278
`
`Merisko-Liversidge et al.
`
`Wilson, D. C. Tormey. Phase I study of taxolol administered
`as a short i.v. infusion daily for 5 days. Cancer Treat. Rep.
`71:1179-1184 (1987).
`18. J. Koeller, T. Brown, K. Havlin, J. Craig, D. D. Von Hoff. A
`phase I study of taxol given as a 6 hour infusion every 21 days.
`Invest. New Drug 7:451(1989).
`19. D. M. Peereboom, R. C. Donehower, E. A. Eisenhauer, W. P.
`McGuire, W. P., N. Onetto, J. L. Hubbard. Successful re-treatment
`with taxol after hypersensitivity reactions. J. Clin. Oncol., 11:885-
`890 (1993).
`20. N. A. Nelson, R. W. Talley, M. L. Reed, A. M. Evans, B. L. Isaacs,
`P. Huffman, J. Lousi. Midwest cooperative group evaluation of
`piposulfan (A-20968) in cancer. Cancer Clin. Pharm. Ther. 8:385-
`391 (1967).
`21. J. J. Van Dyk, G. Falkson, H. C. Falkson. Clinical experience
`(NSC-
`with 1,4-dihyracryloylpiperazine dimethanesulfonate
`47774 ). Cancer Chem. Rep. 52:275-286 (1968).
`22. F. M. Muggia, P. J. Creavan, H. H. Hansen, M. H. Cohen, H.,
`0. S. Selawry. Phase I clinical trial of weekly and daily treatment
`with camptothecin (NSC-100880): Correlation with Preclinical
`Studies. Cancer Chem. Rep. 56:515-521 (1972).
`
`23. W. J. Slichenmyer, E. K. Rowinsky, R. C. Donehower, S. H.
`Kaufmann. The current status of camptothecin analogues as anti(cid:173)
`tumor agents. J. Natl. Cancer Inst. 85:271-291(1993).
`24. K. C. Nicolau, C. Reimer, M. A. Kerr, D. Rideout, W. Wrasidio.
`Design, synthesis, and biological activity of protaxols. Nature
`364:4~66 (1993).
`25. I. A. Darwish, A. T. Florence, A. M. Saleh. Effects of hydrotropic
`agents on the solubility, precipitation and protein binding of etopo(cid:173)
`side. J. Pharm. Sci. 78:577-588 (1989).
`26. G. Borchard, J. Kreuter. Interaction of serum components with
`poly(methylmethacrylate) nanoparticles and the resulting body
`distribution after intravenous injection in rats. J. Drug Targeting
`1:15-19 (1993).
`27. S. E. Dunn, A. Brindley, S. S. Davis, M. C. Davies, L. Ilium.
`Polystyrene-poly(ethylene glycol) PS-PEG 2000 particles as
`model systems for site specific drug delivery: The effect of PEG
`surface density on the in vitro cell interactions and in vivo biodis(cid:173)
`tribution. Pharm. Res., 11: 1016-1022 0994).
`28. R. Gref, Y. Minamitake, M. T. Peracchia, V. Trubetskoy, V. Tor(cid:173)
`chilin, R. Langer. Biodegradable long-circulating polymeric
`nanospheres. Science 263:1600-1603 (1994).
`
`

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