Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Available online at www.sciencedirect.

com

Advanced Drug Delivery Reviews 60 (2008) 757 – 767


www.elsevier.com/locate/addr

Advances in lipid nanodispersions for parenteral drug


delivery and targeting ☆
Panayiotis P. Constantinides a,⁎, Mahesh V. Chaubal b , Robert Shorr c
a
Biopharmaceutical and Drug Delivery Consulting, LLC, Gurnee, IL, USA
b
Baxter Healthcare, Round Lake, IL, USA
c
Cornerstone Pharmaceuticals, Cranbury, NJ, USA
Received 1 September 2007; accepted 20 October 2007
Available online 6 November 2007

Abstract

Parenteral formulations, particularly intravascular ones, offer a unique opportunity for direct access to the bloodstream and rapid onset of drug
action as well as targeting to specific organ and tissue sites. Triglyceride emulsions, liposomes and micellar solutions have been traditionally used
to accomplish these tasks and there are several products on the market using these lipid formulations. The broader application of these lipid
systems in parenteral drug delivery, however, particularly with new chemical entities, has been limited due primarily to the following reasons: a)
only a small number of parenteral lipid excipients are approved, b) there is increasing number of drugs that are partially or not soluble in
conventional oils and other lipid solvents, and c) the ongoing requirement for site-specific targeting and controlled drug release. Thus, there is
growing need to expand the array of targetable lipid-based systems to deliver a wide variety of drugs and produce stable formulations which can
be easily manufactured in a sterile form, are cost-effective and at least as safe and efficacious as the earlier developed systems. These advanced
parenteral lipid-based systems are at various stages of preclinical and clinical development which include nanoemulsions, nanosuspensions and
polymeric phospholipid micelles. This review article will showcase these parenteral lipid nanosystems and discuss advances in relation to
formulation development, processing and manufacturing, and stability assessment. Factors controlling drug encapsulation and release and in vivo
biodistribution will be emphasized along with in vitro/in vivo toxicity and efficacy case studies. Emerging lipid excipients and increasing
applications of injectable lipid nanocarriers in cancer chemotherapy and other disease indications will be highlighted and in vitro/in vivo case
studies will be presented. As these new parenteral lipid systems advance through the clinic and product launch, their therapeutic utility and value
will certainly expand.
© 2007 Elsevier B.V. All rights reserved.

Keywords: Parenteral delivery; Lipid-based systems; Nanoemulsions; Nanosuspensions; Polymeric phospholipid micelles; Manufacturing; Product stability; Cell and
tissue targeting; Biodistribution; Pharmacokinetics

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 758
2. Formulation and manufacturing aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 758
2.1. Nanoemulsions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 758
2.2. Nanosuspensions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 759

Abbreviations: PEG, polyethylene glycol; CMC, critical micelle concentration; PIT, phase-inversion temperature; PE, phosphatidylethanolamine; MTD, maximum
tolerated dose; Cmax, maximum plasma concentration; AUC, area under the plasma concentration–time curve; EPR, enhanced permeability retention; LCM, lipid-
coated microbubbles; LDL, low-density lipoprotein; DSPE, 1,2-Distearoyl-sn-Glycero-3-Phosphoethanolamine or distearoylphosphatidylethanolamine.

This review is part of the Advanced Drug Delivery Reviews theme issue on “Lipid-Based Systems for the Enhanced Delivery of Poorly Water Soluble Drugs”.
⁎ Corresponding author. Biopharmaceutical and Drug Delivery Consulting, LLC, P. O. Box 7565, Gurnee, IL 60031, USA. Tel./fax: +1 847 599 9496; +1 224 430
0383 (Mobile).
E-mail address: constantinpp@aol.com (P.P. Constantinides).

0169-409X/$ - see front matter © 2007 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2007.10.013
758 P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767

2.3. Polymeric phospholipid micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 759


2.4. Formulation considerations for nanoparticulate systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760
3. In vitro/in vivo case studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760
3.1. Nanoemulsions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760
3.2. Nanosuspensions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 763
3.3. Polymeric phospholipid micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 764
4. Conclusions and future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 764
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765

1. Introduction without the inherent creaming, flocculation, coalescence and


sedimentation associated with macroemulsions. They can be
Triglyceride emulsions, liposomes and micellar solutions have formulated in a variety of formulations such as liquids, sprays,
been traditionally employed to solubilize and parenterally deliver foams, creams, ointments and gels.
poorly soluble drugs with several approved products on the market Likewise, nanosuspensions of drugs are submicron colloidal
[1–4]. These formulations, however, incorporate a small number dispersions of drug particles which are stabilized by surfactants
of approved parenteral excipients and have limitations in terms of [6,7]. They can be distinguished from nanoparticles which are
drug solubilization/loading and targeting, sustained release polymeric drug carriers [2,8–10,20] and solid lipid nanoparti-
characteristics, improved efficacy and pharmacokinetics, as well cles, which are lipidic drug carriers [21]. One of the advantages
as improved manufacturing and shelf-life [1–4]. Several of the of nanosuspensions is their use to formulate drugs that are
existing drugs and new chemical entities are not soluble in insoluble in both water and oils and are usually employed when
conventional triglyceride oils/lipids and surfactants. The devel- the use of other lipid-based systems is limited. Particularly in
opment of advanced parenteral systems in recent years, such as the case of high melting point compounds, solubilization in any
nanoemulsions [3–5], nanosuspensions [6,7] and polymeric solvent is difficult, and nanosuspensions can be used to
micelles [2,8–10] was the outgrowth of the aforementioned needs. maintain these drugs in a preferred crystalline state of
Both conventional and advanced parenteral lipid-based sufficiently small size for intravenous administration [7]. The
systems represent a useful alternative to surfactant: alcohol higher drug loading of nanosuspensions compared to nanoe-
solutions, such as Cremophor EL:ethanol admixtures which mulsions for example, is afforded by the dense, solid state of
required to be diluted with saline or dextrose solutions to final nanosuspensions. High drug loading is advantageous since the
paclitaxel concentration of 0.3 to 1.2 mg/ml prior to adminis- administration volume can be significantly reduced [7].
tration followed by long infusion times, typically over 3 h Polymeric micelles are formed from amphiphilic copolymers
[3,4,11,12]. Although such systems are good solvents for poorly containing hydrophobic and hydrophilic blocks and are widely
soluble drugs, they require special infusion apparatus at extra used recently to solubilize a variety of poorly soluble drugs [2,8–
expense and time. The ability of Cremophor to extract toxic 10,20]. There are three major types based on linear block
plasticizers from manufacturing and i.v. infusion tubing, is copolymers: a) common block copolymer micelle, b) drug-
largely responsible for the observed venous irritation and conjugated block copolymer micelle and c) block ionomer
anaphylactic reactions, such as respiratory distress [13,14]. complex micelle [20]. While there are many options for the
Thus, a desired parenteral reformulation of a marketed drug hydrophobic block, polyethylene glycol (PEG) is the most often
should have the following characteristics: a) is Cremophor-free, used hydrophilic segment due to its flexibility, hydrophilicity and
b) exhibits high drug solubilization/loading capacity, c) is at least biocompatibility [2,9,20]. The size of polymeric micelles is
as efficacious as the marketed formulation, d) it has a shelf-life of typically less than 100 nm and are often more stable than
at least 2 years, e) should be easily processed and manufactured conventional surfactant micelles due to their lower critical micelle
using standard equipment and f) is amenable to fast track concentration (CMC) values [2,8–10,20]. In addition to their high
development status by addressing “unmet medical needs”. drug solubilization capacity, extended drug release and circulation
Nanoemulsions or mini-emulsions are transparent or trans- times are other advantages over conventional micelles [20].
lucent oil-in-water (o/w) or water-in-oil droplets with a mean Within the scope of the present review article, the focus is on
droplet diameter in the range between 100 and 500 nm [1,3– polymeric micelles based on polymer-conjugated phospholipids
5,15–18]. They are also known as submicron emulsions and [2,9,10,22] and more comprehensive discussion on other types of
unlike the thermodynamically stable microemulsions, nanoe- polymeric micelles can be found elsewhere [8–10,20].
mulsions are kinetically stable with great stability in suspension
due to their small droplet size [15]. Ostwald ripening is the 2. Formulation and manufacturing aspects
primary instability process, which can be reduced by the
addition of a second less soluble oil phase and/or addition of a 2.1. Nanoemulsions
strongly adsorbed and water-insoluble polymeric surfactant
[15,19]. Advantages of nanoemulsions over macroemulsions or The methods used to produce nanoemulsions can be divided
coarse emulsions include, higher surface area and free energy into the high- and low-energy ones [15]. High-energy methods
P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767 759

include high-pressure homogenization and microfluidization


which can be used at both laboratory and industrial scale as well
as ultrasonification which is primarily used at laboratory scale
[15,16,19]. These high-energy methods although very effective in
reducing droplet size, are undesirable for labile drugs and
macromolecules, such as proteins and nucleic acids. For these
labile molecules low-energy emulsification methods are preferred
which include, spontaneous emulsification, the solvent-diffusion
method and the phase-inversion temperature (PIT) method [15–
17,19,23,24]. A lipophilic drug can be added to the oil phase
whereas a hydrophilic one can be solubilized in the aqueous phase.
Co-solvents can be used, if needed, to aid drug solubilization.
During high-pressure homogenization, the coarse dispersion
of the oil and aqueous phase is passed through a small inlet Fig. 1. A typical plot showing the particle size mean and 99% for a model drug
orifice at an operating pressure in the range of 500–5000 psi, nanosuspension as a function of homogenization cycle numbers.
where the emulsion mixture is subjected to intense turbulence
and hydraulic shear which then produces a fine emulsion with an drug to have a reasonably high melting point, so that the crystal
extremely small droplet size [25]. Microfluidization uses a high- surface does not experience phase transition that can inhibit
pressure positive displacement pump operating at very high binding of surfactants [6,7].
pressures, up to 20,000 psi, which forces the emulsion product Nanosuspensions utilize similar high-energy processing
through the interaction chamber which consists of a series of equipment as described for nanoemulsions. The amount of
microchannels. The emulsion flows through the microchannels energy required to break a drug crystal is significantly higher
on to an impringement area resulting in very fine emulsion than that required to reduce the particle size of a globule. Hence
droplets [26]. The operating pressure and the number of passes high pressures and multiple passes are required in producing
of the coarse emulsion through the interaction chamber of the nanosuspensions. Besides high-pressure homogenization and
microfluidizer determine the particle size of the fine emulsion. microfluidization, media milling has also been utilized to
The higher the operating pressure and the number of passes, the produce nanosuspensions. In each case, the particle size is
smaller the droplet size of the final emulsion. The resulting reduced by successive passes through the high-energy impact
nanoemulsion can then be filtered through a 0.2 μm filter under zones. A typical particle size reduction profile is shown in Fig. 1.
nitrogen to remove any large particles present resulting in a It is seen that the particle size decreases rapidly in the early
uniform nanoemulsion. High-energy emulsification methods phase, followed by a more gradual reduction. Besides particle
can produce both o/w and w/o nanoemulsions. size reduction, homogenization processes also allow rapid
Among the low-energy emulsification methods, solvent stabilization of the particles by facilitating contact and binding
diffusion and PIT generate o/w nanoemulsions, whereas of the surfactants to the newly formed crystal surfaces [6,7].
spontaneous emulsification produces w/o nanoemulsions [15– Aside from the high-energy processes, nanosuspensions can
17,19,23,24]. By rapid cooling of the microemulsion form in also be prepared directly by crystallization or precipitation. If
the phase-inversion zone, or dilution with water whatever the the crystallization conditions are carefully chosen to maximize
temperature, very stable o/w nanoemulsions can be produced the number of nuclei in a rapid supersaturation environment, the
[19]. Dilution with water is more practical and adaptable and particles produced would be very small — even in the
preferred for industrial and pharmaceutical applications. How- submicron range. Crystal growth inhibitors can be utilized to
ever, it has recently been shown that under certain conditions, restrict growth of the newly formed particles and surfactants can
the PIT method can also produce w/o nanoemulsions [19]. This be used to prevent their aggregation. Efficient removal of the
involves the inclusion of a lipophilic PEG-surfactant to generate solvent is important in such a process, since presence of the
a microemulsion within the phase-inversion zone which upon solvent can lead to Ostwald ripening [7].
dilution with suitable oil produces highly monodispersed w/o Finally, the microprecipitation and high-energy approaches
nanodroplets [19]. This type of nanoemulsions with an aqueous can also be combined to exploit the advantages of both
core has increasing demand for hydrophilic drug delivery and processes. Such a combination process can be more efficient as
targeting, particularly for peptides/proteins [27] but is beyond compared to homogenization alone. The particle size achieved
the scope of this review article. with the combination process was lower than that seen for direct
homogenization. Furthermore, the combination process allows
2.2. Nanosuspensions for the possibility of aseptic processing for thermally sensitive
molecules [7].
Nanosuspensions are useful as injectable dosage forms for
poorly soluble drugs. Stabilization of the nanoparticles in a 2.3. Polymeric phospholipid micelles
suspension form requires strong binding of surfactants onto the
particle surface. Furthermore high energy is often required in There are several methods to incorporate drugs into micelles.
production of such particles — both these aspects require the These include, dialysis of organic solvents or micellar solutions of
760 P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767

detergents [2,9,10,22]. In the former method, the drug in a volatile Table 1


organic solvent is added to a preformed aqueous dispersion of Choice of excipients for parenteral nanosuspensions
polymeric micelles and subsequently the organic solvent is Excipient Function IV SC/ Ocular Pulmonary
removed by evaporation. Alternatively, the drug is dissolved in a IM
detergent solution and upon mixing with a polymeric micelles Surfactant(s) Particle stabilization + + + +
dispersion, excess detergent is removed by dialysis. For the Buffer pH adjustment and control + + + +
Polysaccharides Viscosity enhancement − + + −
preparation of drug-loaded PEG-PE micelles which are discussed
Sugars Tonicity adjuster and/or + + + +
in this review article, the procedure involves [22]: a) a dry film of lyoprotectant
the drug and PEG-PE is formed by evaporation of an organic Additional Bioadhesives, matrices for − + + −
solvent solution of the mixture, and b) hydration of the dry firm polymers sustained release
with an aqueous solution/buffer by intensive shaking to form Preservatives Preservatives for multidose − + + +
products
drug-loaded micelles. Any insoluble drug is precipitated in a
Chelating Scavenging of metal ions + + + +
crystalline form and removed by filtration. In order to improve agents (depends on drug stability)
drug solubilization, if needed, additional micelle-forming com-
pounds, such as egg phosphatidylcholine, are added to PEG-PE
micelles [22]. Several poorly soluble drugs were successfully surfactants used to stabilize the nanoparticles are critical in
solubilized in PEG-PE micelles with loading efficiencies in the defining the stability of the product. Some of the commonly used
range of 1.5 to 50% w/w and tight association of the investigated surfactants for stabilization include poloxamers [28,29], phos-
drugs with the PEG-PE micelles was demonstrated by prolonged pholipids [30], and Tweens [31]. Buffers and tonicity agents play
dialysis against aqueous buffer under sink conditions [22]. an important role in maintaining stability of a suspension —
ionizable species may shield the electrostatic repulsion between
2.4. Formulation considerations for nanoparticulate systems particles, thereby reducing the overall stability.
Given the complex physical forces involved in potential
For each of the type of system discussed above, particle size, destabilization of nanoparticles, formulation of such systems
nature and density of coatings, and rate of dissolution of the requires specific accelerated stress tests. Higher temperature and
submicron particles are the primary factors that impact their fate humidity driven accelerated stability studies typically used for
and efficacy. Hence formulation considerations for all these other dosage forms, can be used to demonstrate long-term
systems can be discussed collectively, and learnings from one stability. However formulation screening requires additional
system can be directly applied to another. stress tests to explore potential long-term destabilizing factors
Physical stability is an essential requirement for a nanoparti- such as sedimentation, Ostwald ripening, and aggregation.
culate system. Poorly formulated particles can aggregate over
time leading to higher number of large particles, thereby creating 3. In vitro/in vivo case studies
a potential safety concern. Nanoparticles may also sediment over
time in storage. If not appropriately stabilized, the sediment may In this section, case studies are presented with various poorly
be difficult to resuspend. Finally, if the drug has finite solubility soluble drugs beginning with nanoemulsions, followed by
in the suspending buffer, the drug particles can dissolve and nanosuspensions and polymeric phospholipid-based micelles.
recrystallize on large particles, a phenomenon commonly known
as Ostwald ripening. Any of these issues can lead to a change in 3.1. Nanoemulsions
particle size and consequently, the behavior of the finished
product. Particle size of the final formulation is particularly Paclitaxel is a member of the class of drugs known as taxanes
important for parenteral dosage forms, since that may also and has been isolated from the bark or leaves (needles) of the
impact safety of the product. Hence development of a stable Pacific Yew tree, Taxus brevifolia and related species. Taxanes
nanosuspension has to take into account all these modes of have been found to be useful in the treatment of various cancers
physical instability. Processes such as spray drying, freezing or such as ovarian, breast, non-small cell lung and head and neck
lyophilization can be implemented to facilitate stabilization of carcinomas although allergic reactions were observed to
the particles. However these processes can cause additional formulation excipients [11,12,32,33]. A difficulty in adminis-
stress to the particles, thereby destabilizing them. For example trating taxanes is that the drug is typically insoluble in water.
drying can cause dehydration of the adsorbed stabilizers, causing Paclitaxel has been formulated in a 1:1 mixture of Cremophor
particle aggregation. Hence excipients and stabilizers have to be EL® (a polyethoxylated castor oil) and ethanol to create Taxol®
carefully chosen to stabilize the particles and prevent particle (Bristol-Myers Squibb, Inc) which per ml contains 6 mg
size change during processing and storage. In addition, the route paclitaxel, 527 mg Cremophor EL and 49.7% (v/v) ethanol.
of administration, desired pharmacokinetic profile, and addi- Reconstitution in a suitable carrier is required (stable for ∼ 12 h)
tional features (such as targeting) have to be considered. Some of along with in-line filtration to remove any drug, which may
the excipients used in nanosuspensions for different routes of crystallize out. Taxol® has been shown to be neurotoxic,
administration are listed in Table 1. Most of the excipients on the causing sensory and sometimes accompanying motor neuro-
list (such as tonicity adjusters, chelating agents, preservatives) pathy in patients. Specific neurological manifestations of this
are commonly used in parenteral solutions. However the condition, are numbness/paraesthesia, tingling/burning in the
P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767 761

charged tumor vessel endothelia. These characteristics were


determined in early preclinical studies as shown in Figs. 2 and 3.
Fig. 2 presents antitumor activity data in mice implanted with
B16 melanoma on a q3dx5 schedule using saline, drug-free
nanoemulsion and Taxol® as controls. Tumor growth over
time was monitored using TOCOSOL paclitaxel at 20, 40 and
60 mg/kg and Taxol® at its MTD of 20 mg/kg. As can be seen
from the data in Fig. 2, Taxol® at 20 mg/kg showed minimal
antitumor activity with a log cell-kill value of 0.5 and 50%
mortality [3,38]. In contrast, TOCOSOL paclitaxel was much
better tolerated with no mortality at 20 and 40 mg/kg and log cell-
kill values of 1.8 and 3.0, respectively [3,38] as evident from the
complete tumor remission at 40 and 60 mg/kg (Fig. 2). The
improved antitumor activity of TOCOSOL paclitaxel compared
Fig. 2. Comparative tumor growth of mice implanted with B16 murine to Taxol® in B16-bearing mice was found to be correlated to its
melanoma. Mice were administered saline, drug-free vehicle control, tocol enhanced tumor uptake as illustrated in Fig. 3. While the total
emulsion (listed as TOCOSOL paclitaxel) and the reference formulation Taxol® paclitaxel concentration in the blood was similar following
on a schedule of q3dx5 (values shown are mean ± SEM). The data points for the administration of these two formulations, paclitaxel concentration
40 and 60 mg/kg groups are overlaid. Reprinted from Ref. [3].

limbs, general diffuse weakness, and a loss of reflexes,


including those maintaining posture and gait [12,13,33].
Cremophor EL® is in part responsible for the neuropathy, as
it is itself not free of significant side effects [13,14].
Attempts to formulate paclitaxel in a long-term stable lipid
emulsion have been challenging [34–37]. The drug is reported to
be insoluble in lipid emulsions such as Intralipid®, which
contains primarily soybean oil, or Liposyn®, which contains a
mixture of soybean and safflower oils [34]. Heating paclitaxel in
either soybean oil or safflower oil, or even upon sonication, does
not result in the dissolution of appreciable amounts of drug, and
addition of paclitaxel to a lipid emulsion during a homogeniza-
tion step meets with equally disappointing results. Emulsions
incorporating up to 15 mg/mL of paclitaxel have been
formulated with triacetin, L-α-lecithin, Polysorbate 80, Pluronic
F-68, ethyloleate and glycerol. However, these emulsions are
toxic and unstable upon storage [35].
There is a clear need for a stable, easily prepared, bio-
compatible, efficacious formulation of taxanes, such as paclitaxel,
exhibiting minimal side effects that would promote uptake into
tumor cells with greater potency against drug-resistant cells. Oil-
in-water nanoemulsions incorporating paclitaxel meet these
criteria as it will be illustrated below.
A vitamin E-based nanoemulsion of paclitaxel (TOCOSOL
paclitaxel) was developed by SONUS Pharmaceuticals and it is
currently in Phase III clinical development [3,4,38–42]. It is a
Cremophor-free formulation of paclitaxel, a ready-to-use
injectable product that incorporates high drug loading (8–
10 mg/ml) in smaller dose administration volumes and shorter
infusion times. Based on the data to date, TOCOSOL paclitaxel
shows greater tumor accumulation than Taxol® and antitumor
efficacy allowing for the delivery of a greater “dose density” to
the tumor [43,44]. TOCOSOL paclitaxel nanodroplets manu-
factured by proprietary high shearing homogenization have a
Fig. 3. Comparative paclitaxel concentration in blood (top panel) and tumor
mean droplet diameter in the range of 40–80 nm, are stable and tissue (bottom panel) upon a single administration of 10 mg/kg dose of Taxol®
neutrally charged, which allows for diffusion into the tumor and TOCOSOL paclitaxel to mice implanted with B16 murine melanoma tumor
interstitium without impediment by the highly negatively cells. Reprinted from Ref. [3].
762 P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767

in the tumor was higher following administration of TOCOSOL LCM could influence increased LDL receptor-mediated
paclitaxel, resulting in 1.5-times higher Cmax and 2.2-times higher endocytosis occurring within the tumor tissue. Enhanced LDL
AUC compared to the parameters produced by Taxol®. Higher receptor-mediated uptake of LCM by tumor tissue may explain
accumulation of TOCOSOL paclitaxel is due to the enhance the rapidity and selectivity of LCM accumulation in tumors, as
permeability retention (EPR) effect [44] as a result of the compared to findings with liposomes [71,72].
extremely small size of the tocol nanoemulsion. Other LDL or alternative lipid receptor gene-family
D'Arrigo et al. [45–64] published a number of studies in the members [73,74] may well participate in this receptor-mediated
early 1990s describing a composition and method to produce uptake of LCM by tumor tissue [65]. For example, apo E and
lipid-coated microbubbles (LCM) as an effective sonographic LPL binding to various lipid particles (as similarly proposed
contrast agent for localization, of experimental tumors in rats above for LCM) is widely reported to facilitate the uptake of the
[45–50]. LCM consists of glycerides, cholesterol and choles- lipid particles [e.g., apo E-enriched β -VLDL or LPL-enriched
terol esters. Most of the tumors examined, were located in the VLDL, β -VLDL, and chylomicrons] by a large “multi-ligand”
brain, liver and subcutaneous tissues. The observed sonographic endocytic receptor known as “LDL receptor-related protein” or
enhancement of tumor images [46–48] suggested LCM LRP [73].
accumulated rapidly and selectively within the cells of a In addition, LCM could likely bind (as do LDL with high
tumor mass. This is an important distinction from other tumor affinity) to macrophage scavenger receptors or be linked to lipid
mass accumulation promoting technologies and has been found raft phenomena. LCMs range in diameter from about 0.5 to
to be composition specific. The result was confirmed by 4 μm, with the vast majority having a diameter of approximately
counter-staining histological samples, from each of the above 0.5 μm or less [59,64].
tumor locations, with either of two lipid-specific stains. Results Further development of the D'Arrigo LCM technology and
provided micrographic documentation of lipid-stained disc-like modification of chemical composition specifically for drug
structures accumulated in the tumor cells but not in the tissues in delivery to tumors as a non-gas containing nanoemulsion with
which the tumors are embedded [46,50]. increased drug payload and solubility of lipids over LCM have
D'Arrigo et al. [45] evaluated LCM drug delivery using been achieved by Cornerstone Pharmaceuticals (www.corner-
glioblastoma C6 cells and gliosarcoma 9L cells, in tissue culture stonepharma.com, Cranbury, NJ) without the use of Cremo-
and orthotopic rat models. After labeling LCM with the phor–ethanol. This improved technology called Emulsiphan,
fluorescent lipophilic dye “diO”, Barbarese et al. [51] reported like LCM has been shown to be rapidly taken up into cancer
that analysis of LCM interactions by confocal laser scanning cells selectively directly in culture and with increased efficacy
microscopy with C6 and 9L cells in culture, showed that LCM in human tumor xenograft mouse models. Antibodies directed
was first adsorb at the surface of the cells, and with time became against the lipid transport domain of the LDL receptor however
localized inside the cells. Binding and internalization proceeded did not block or decrease uptake and in some studies increased
faster at 37 °C than at room temperature. Similar analysis of Emulsiphan uptake. That material that was taken up into cells
such tumors in vivo, from rats injected with diO-labeled LCM, was demonstrated by confocal microscopy using fluorescent
again revealed that labeled LCM was actually inside the tumor labeled preparations. Membrane fusion-lipid raft or other lipid
cells. Barbarese et al. [51] further found that staining of live transport receptors may be involved in the selective uptake.
cells in tissue culture with DAMP, a dye that recognizes acidic Further study to elucidate this mechanism is required. An
compartments, showed that the majority of internalized LCM Emulsiphan lipid–oil–water nanoemulsion has been formulated
was associated with compartments containing DAMP. If the with paclitaxel resulting in the product candidate EmPAC. This
same uptake mechanism were operative in vivo, it would is a ready to infuse drug with increased tumor cell uptake and
indicate that a portion of LCM bypasses the reticuloendothelial greater safety and efficacy than Taxol® or liposomal formula-
system and become endocytosed directly by tumor cells. tions of drug in comparative human tumor cell or xenograft
LCM/Cremophor–ethanol drug delivery was also explored models. Increased uptake and binding of drug to microtubules
using Taxol® (Bristol-Myers Squibb Co., NY, NY) in rat over the same concentration of paclitaxel formulated as Taxol®
orthotopic brain tumor models. Ho et al. [52] reported that the or other approved formulations has been demonstrated in cells
LCM/Cremophor–ethanol Taxol® reduced tumor progression in culture using anti-paclitaxel antibodies.
in Fischer 344 rats inoculated with 9L glioma more effectively In surveying a variety of tumor types it was found that while
than Taxol® alone. most solid tumors would take up the nanoemulsion they did not
Uptake into diverse tumor human types in cell culture or do so to the same degree (Table 2). Level of uptake in parenteral
xenographic models was not reported. Based upon the structure 3 T3 L1 cells was not detectable. As shown in Fig. 4 comparison
of LCM and their known physicochemical properties [53–64], of EmPAC and paclitaxel in Cremophor–ethanol indicated
D'Arrigo has suggested that LCM may bind apolipoprotein B- EmPAC to be substantially more effective in tumor growth
100 (apo B-100), apolipoprotein E (apo E), and/or lipoprotein delay with a 4–10-fold-higher dose of Taxol® required to
lipase (LPL) in the bloodstream [65]. Apo B-100 is known to achieve the same result, depending on the model being studied.
mediate low-density lipoprotein (LDL) binding to cellular LDL The aggressively growing H460 NSLC line was used. That
receptors, and it is widely reported that many tumor cells show increased cellular uptake that translates into increased efficacy
increased LDL receptor expression and activity [66–70]. at lower doses is supported by the observation that microtubule
Consequently, the proposed binding of plasma apo B-100 by sites for taxane binding are saturable. Doses of drug above the
P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767 763

Table 2
Uptake of Emulsiphan nanoparticles by human tumor cell lines
Tumor cell lineage Cell line Fluorescence intensity via N
FACS a (mean ± SEM)
Breast MDA-MB-231 120.02 ± 26.37 4
HS578T 100.65 ± 12.48 4
MX-1 86.51 ± 13.28 3
Lung H460 93.60 ± 20.34 5
A549 48.82 ± 8.07 4
H23 189.03 ± 37.86 4
H522 184.67 ± 40.58 4
Central nervous system SF-539 219.63 ± 23.33 10
SF-268 100.41 ± 7.17 3
SF-295 232.74 ± 45.79 2
U-87 234.07 ± 25.54 2
Colon HT-29 46.44 ± 5.68 12
COLO-205 35.17 ± 6.89 4
SW-620 32.11 ± 7.79 4
HCT-15 31.59 ± 16.66 3 Fig. 5. EMPAC versus Taxol® uptake by brain and lung cells.
Caco-2 52.87 ± 5.52 4
Liver Hep-3B 537.69 ± 27.71 7
Uterine and MES-SA 204.92 ± 39.38 4
drug-resistant uterine MES-SA-DX5 180.57 ± 27.88 4 lower more frequent dosing can be use more safely without
MES-SA-MX2 220.81 ± 32.85 6 risking decreased efficacy. Typically 2–10-fold increases in
Ovarian SKOV-3 65.81 ± 11.42 4
EmPAC uptake can be observed on short-term (1 h) incubations
Pancreatic PAN 02 211.99 ± 35.76 4
PAN 03 73.07 ± 22.30 4 depending on cell line and conditions as for brain SF539 and
a lung A549 cells (Fig. 5).
Average fluorescence intensity per cell was obtained for each fluorescent
Emulsiphan-treated sample, by comparing and subtracting fluorescence from the EmPAC is being prepared for investigation in human clinical
same respective untreated cell line. trials. The Emulsiphan technology has been shown to be
applicable to additional taxanes, camptothecins and other
known chemotherapeutic drugs.
ability to saturate these sites are major contributors to side
effects. 3.2. Nanosuspensions
Tumor growth inhibition studies where cells were exposed
briefly to alternative taxane preparations showed an increased In vivo behavior of nanosuspensions strongly depends on three
“apparent” potency for EmPAC. The data supports the notion factors: (a) particle size distribution; (b) dissolution rates; and
that higher dosing can be made even more effective or that (c) nature and density of coatings [7]. Particles that dissolve
rapidly (in order of seconds to minutes) will show pharmacoki-
netic behavior that is similar to solutions. For example, Clement et
al. demonstrated that a phospholipid-based nanosuspension of
flurbiprofen had the same pharmacokinetic behavior and in vivo
distribution as a high-pH solution formulation of the drug after
intravenous injection in rats [74]. This similarity between the
crystalline suspension and the solution formulations was due to
the rapid dissolution of the particles in the blood. Slow dissolving
particles on the other hand are either taken up by the
reticuloendothelial system, or can be targeted to specific tissues
by making them small and adding specific coatings. For example,
small (b 200 nm), PEGylated nanoparticles are known to have
stealth properties, allowing for passive targeting to tumor cells,
via the process of enhanced permeation retention (EPR) effect
[72,75]. Shenoy et al. [75] tested polyethylene oxide-modified
poly(beta-amino ester) nanoparticles in mice containing human
ovarian carcinoma xenograft (SKOV-3). The PEO-based particles
showed higher tumor selectivity via the EPR effect [44].
Macrophage targeting of nanoparticles has been demonstrated
by Rabinow et al. for a poorly soluble drug, Itraconazole [76]. In
Fig. 4. Comparative tumor growth of H460-tumor bearing mice treated with
EmPAC versus Taxol®. Test agents were administered i.p. 3× weekly at the this study, particles greater than 200 nm were seen to be taken up
indicated doses and percent changes in tumor size values represent mean ± SEM by circulating macrophages. The drug then released from the cells
(n = 10). gradually, allowing for sustained release over 3 days. These
764 P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767

Fig. 6. Selective accumulation of PE-PEG micelles in EL4 T lymphoma tumor in mice: (A) pharmacokinetics, (B) AUC. Reprinted from Ref. [22].

examples demonstrate the importance of in vitro characterization similar to a cyclodextrins-based solution formulation. This
of particle size, surface charge, and dissolution rates in suggests that the particles dissolved upon dilution in blood
qualitatively predicting the fate of the nanosuspension in vivo. allowing for a solution like distribution. Another intravenous
Nanosuspensions have been investigated extensively as a nanoparticle formulation tested in clinical trials was Spartaject
delivery system for poorly soluble drugs. Liversidge et al. Busulfan — a formulation stabilized by dimyritoylphosphatidyl-
demonstrated the efficacy of nanoparticulate formulations of choline, and dilauroylphosphatidyl choline in a buffer containing
various anticancer drugs (piposulfan, camptothecin, etoposide, mannitol [80]. Twelve patients with chronic myeloid leukemia
and paclitaxel) in a CH3 mouse model [77]. Formulation of the were treated in a Phase I/II clinical trial. The nanoparticle
drugs as a nanosuspension led to an improved acute toxicity formulation demonstrated low intra-patient variability compared
profile. Peters et al. developed a nanosuspension of an anti- to the oral formulation. Furthermore, the nanoparticle formulation
infective drug, clofazamine and compared its in vivo biodis- indicated reduced toxicity compared to other formulations.
tribution with a liposomal formulation [78]. It was seen that the Finally, a paclitaxel nanosuspension in albumin matrix has been
nanosuspension with a particle size mean of around 380 nm, approved for the treatment of metastatic breast cancer [81]. Data
primarily targeted the reticuloendothelial system, with a from various clinical studies for this product have been published
distribution quite similar to liposomes. The advantage that [81–83]. Most significantly, a pivotal Phase III trial involving 454
nanosuspensions provided in this situation was better stability, breast cancer patients, showed that a high dose of paclitaxel could
compared to liposomes. Scholer et al. utilized nanosuspensions be administered without any premedication with steroid and
for the intravenous delivery of an antimalarial drug, atova- antihistamines (typically required to alleviate Cremophor related
quone, in a murine model [79]. Intravenously injected hypersensitivity). The infusion time could also be reduced, given
atovaquone nanosuspension was distributed in various sites of the absence of Cremophor EL. The nanosuspension formulation
infection, such as the liver, brain, and lung. Administration of also showed a higher response rate, higher time to tumor
atovaquone nanosuspension at a dose of 10 mg/kg led to serum progression, and lower incidence of grade 4 neutropenia.
drug concentrations similar to those attained by oral adminis-
tration of a 10-fold-higher dose of atovaquone suspension. 3.3. Polymeric phospholipid micelles
Rabinow et al. developed a nanosuspension of Itraconazole
for intravenous administration [76]. This nanosuspension Long circulation times of PEG-PE micelles were demonstrated
showed a unique pharmacokinetic profile, with significant pro- in mice by measuring blood clearance as a function of the mole-
longation of the circulation half-life of the drug, as compared to cular size of the PEG block [22]. As the size of the PEG block
a cyclodextrins-based solution formulation. This prolonged increases, the circulation half-lives in the blood increased
half-life may be a result of sequestration of the particles into the from about 1 to 2 h as a result of steric hindrance against opsonin
macrophages, and subsequent gradual release of the dissolved penetration to the hydrophobic core of the micelle [22].
drug. Preferential accumulation of PEG-PE micelles to tumors is
Dou et al. have developed a unique approach to use shown in Fig. 6 using EL4 T-cell lymphoma murine tumor model
macrophages as a targeting vehicle for delivery of antiretroviral [22]. Micelles from distearoylphosphatidylethanolamine (DSPE)
drugs [30]. A nanosuspension of indinavir was loaded into bone incorporating either PEG750 or PEG2000, showed selective
marrow derived macrophages, and injected into HIV-1-challenged tumor accumulation (Fig. 6). The slightly better accumulation
humanized mice. The targeted delivery system significantly of PEG750-DSPE compared to PEG2000-DSPE may be related to
reduced numbers of virus-infected cells in plasma, lymph nodes, the small cutoff size of the EL4 vasculature [22].
spleen, liver, and lung, and led to CD4 (+) T-cell protection.
A nanosuspension of Itraconazole stabilized by Poloxamer 4. Conclusions and future perspectives
388 was tested in human clinical trials [29]. It was seen that the
nanosuspension allowed for higher tolerable dose. Also, the Nanoemulsions, nanosuspensions and polymeric phospho-
pharmacokinetic profile of the nanosuspension was seen to be lipid micelles constitute novel parenteral formulations of
P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767 765

approved drugs or new chemical entities. They are at different [13] D. Dye, J. Watkins, Suspected anaphylactic reaction to Cremophor EL, Br.
stages of preclinical and clinical development. It is evident from Med. J. 280 (1980) 1353.
[14] H. Gelderblom, J. Verweij, K. Nooter, A. Sparreboom, Cremophor EL: the
the preceding discussion that there have been major develop- drawbacks and advantages of vehicle selection for drug formulation, Eur. J.
ments and advances with these novel parenteral nanocarriers Cancer 37 (2001) 1590–1598.
both at the preclinical and clinical phases. These nanosized [15] T. Tadros, P. Izquierdo, J. Esquena, C. Solans, Formation and stability of
particles with a very large interfacial area can influence the nanoemulsions, Adv. Colloid Interface Sci. 109 (2004) 303–318.
[16] S. Shafiq-un-Nabi, F. Shakeel, S. Talegaonkar, J. Ali, S. Baboota, A. Ahuja,
transport and delivery properties of the incorporated drugs and
R.K. Khar, M. Ali, Formulation development and optimization using
provide for site-specific targeting. Their enhanced delivery nanoemulsion technique: a technical note, AAPS Pharm. Sci. Tech. 8 (2)
characteristics (improved efficacy and better tolerance), along (2007) 28.
with improved biodistribution and pharmacokinetics have been [17] P.P. Constantinides, Seang H. Yiv, Particle size of phase inverted water-in-
well documented with cancer chemotherapeutic agents and oil microemulsions under different dilution and storage conditions, Int. J.
other drugs. Furthermore, improvements in manufacturing Pharm. 115 (1995) 225–234.
[18] E.C. Unger, T. Porter, W. Culp, R. Labell, T. Matsunaga, R. Zutshi,
methods and product stability have been achieved. As these Therapeutic applications of lipid-coated microbubbles, Adv. Drug Del.
new parenteral lipid formulations advance through the clinic Rev. 56 (2004) 1291–1314.
and product launch, their therapeutic utility and value will [19] N. Anton, P. Saulnier, A. Beduneau, J.P. Benoit, Salting-out effect induced
certainly expand. by temperature cycling of a water/nonionic surfactant/oil system, J. Phys.
Chem. B 111 (2007) 3651–3657.
[20] L.Y. Qiu, Y.H. Bae, Polymer architecture and drug delivery, Pharm. Res.
Acknowledgements 23 (2006) 1–30.
[21] S.A. Wissing, O. Kayser, R.H. Muller, Solid lipid nanoparticles for
We would like to thank Dr. Vladimir P. Torchilin of parenteral delivery, Adv. Drug Del. Rev. 56 (2004) 1257–1272.
Northeastern University for the discussion on polymeric [22] A.N. Lukyanov, V.P. Torchilin, Micelles from lipid derivatives of water-
phospholipid micelles and permission to include some of his soluble polymers as delivery systems for poorly soluble drugs, Adv. Drug
Del. Rev. 56 (2004) 1273–1289.
published data in this review article. We would also like to thank [23] T. Foster, W. Von Rybinski, A. Walde, Influence of microemulsion phases
Rajunder Bhasin, Claudia Moore, Sameer Kulkarni and Lan on the preparation of fine-disperse emulsions, Adv. Colloid Interface Sci.
Hnuygen of Cornerstone Pharmaceuticals for their skillful 58 (1995) 119–149.
contributions to the Emulsiphan work. [24] R. Rons, I. Carrera, J. Caelles, J. Rouch, P. Panizza, Formation and
properties of miniemulsions formed by microemulsions dilution, Adv.
Colloid Interface Sci. 106 (2003) 129–143.
References [25] C. Washington, S.S. Davis, The production of parenteral feeding
emulsions by microfluidizer, Int. J. Pharm. 44 (1988) 169–176.
[1] J. Rossi, J.-C. Leroux, Principles in the development of intravenous lipid [26] S. Pinnamaneni, N.G. Das, S.K. Das, Comparison of oil-in-water
emulsions, in: K. Wasan (Ed.), Role of Lipid Excipients in Modifying Oral emulsions manufactured by microfluidization and homogenization,
and Parenteral Drug Delivery, Wiley-Interscience, Hoboken, New Jersey, Pharmazie 58 (2003) 554–558.
2007, pp. 88–123. [27] I. Jorgensen, E.H. Moeller, M. van de Weert, H.M. Nielsen, S. Frokjaer,
[2] V.P. Torchilin, Lipid-based parenteral drug delivery systems: biological Preparing and evaluating delivery systems for proteins, Eur. J. Pharm. Sci.
implications, in: K. Wasan (Ed.), Role of Lipid Excipients in Modifying 29 (2006) 174–182.
Oral and Parenteral Drug Delivery, Wiley-Interscience, Hoboken, New [28] J. Moschwitzer, R.H. Muller, New method for the effective production of
Jersey, 2007, pp. 48–87. ultrafine drug nanocrystals, J. Nanosci. Nanotechnol. 6 (2006) 3145–3153.
[3] P.P. Constantinides, A. Tustian, D.R. Kessler, Tocol emulsions for drug [29] J.W. Mouton, A. van Peer, K. de Beule, A. Van Vliet, J.P. Donnelly, P.A.
solubilization and parenteral delivery, Adv. Drug Del. Rev. 56 (2004) Soons, Pharmacokinetics of itraconazole and hydroxyitraconazole in
1243–1255. healthy subjects after single and multiple doses of a novel formulation,
[4] P.P. Constantinides, J. Han, S.S. Davis, Advances in the use of tocols as Antimicrob. Agents Chemother. 50 (2006) 4096–4102.
drug delivery vehicles, Pharm. Res. 23 (2006) 243–255. [30] H. Dou, C.J. Destache, J.R. Morehead, R.L. Mosley, M.D. Boska, J. Kingsley,
[5] D.K. Sarker, Engineering of nanoemulsions for drug delivery, Cur. Drug S. Gorantla, L. Poluektova, J.A. Nelson, M. Chaubal, J. Werling, J. Kipp, B.E.
Deliv. 2 (2005) 297–310. Rabinow, H.E. Gendelman, Development of a macrophage-based nanoparticle
[6] R.H. Muller, K. Peters, Nanosuspensions for the formulation of poorly platform for antiretroviral drug delivery, Blood 108 (8) (2006 Oct 15)
soluble drugs I. Preparation by a size reduction technique, Int. J. Pharm. 2827–2835.
160 (1998) 229–237. [31] B. Van Eerdenbrugh, L. Froyen, J.A. Martens, N. Blaton, P. Augustijns,
[7] B.E. Rabinow, Nanosuspensions in drug delivery, Nature Rev. 3 (2004) M. Brewster, G. Van den Mooter, Characterization of physico-chemical
785–796. properties and pharmaceutical performance of sucrose co-freeze-dried
[8] D. Sutton, N. Nasongkla, E. Blanco, J. Gao, Functionalized micellar solid nanoparticulate powders of the anti-HIV agent loviride prepared by
systems for cancer targeted drug delivery, Pharm. Res. 24 (2007) media milling, Int. J. Pharm. 338 (2007) 198–206.
1029–1046. [32] E.K. Rowinsky, R.C. Donehower, Paclitaxel (Taxol®), New Engl. J. Med.
[9] V.P. Torchilin, Micellar nanocarriers: pharmaceutical perspectives, Pharm. 332 (1995) 1004–1014.
Res. 24 (2007) 1–16. [33] A.K. Singla, A. Garg, D. Aggarwal, Paclitaxel and its formulations, Int. J.
[10] G.S. Kwon, Polymeric micelles for delivery of poorly water-soluble Pharm. 235 (2002) 179–192.
compounds, Crit. Rev. Ther. Drug Carr. Syst. 20 (2003) 357–403. [34] L.C. Collins-Gold, R.T. Lyons, L.C. Bartholow, Parenteral emulsions for
[11] A. Sparreboom, L. van Zuylen, E. Brouwer, W.J. Loos, P. de Bruijn, drug delivery, Adv. Drug Del. Rev. 5 (1990) 189–208.
H. Gelderblom, M. Pillay, K. Nooter, G. Stoter, J. Verweij, Cremophor [35] B.D. Tarr, T.G. Sambandan, S.H. Yalkowsky, A new parenteral emulsion
EL-mediated alteration of paclitaxel distribution in human blood: clinical for the administration of Taxol®, Pharm. Res. 4 (1987) 162–165.
pharmacokinetic implications, Cancer Res. 59 (1999) 1454–1457. [36] B. Lundberg, A submicron lipid emulsion coated with amphipathic
[12] L. van Zuylen, J. Verweij, A. Sparreboom, Role of formulation vehicles in polyethylene glycol for parenteral administration of paclitaxel (Taxol®l),
taxane pharmacology, Invest. New Drugs 19 (2001) 125–141. J. Pharm. Pharmacol. 49 (1997) 16–21.
766 P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767

[37] P. Simamora, R-M. Dannenfelser, S.E. Tabibi, S.H. Yalkowsky, Emulsion [59] J.S. D'Arrigo, Biological surfactants stabilizing natural microbubbles in
formulations for intravenous administration of paclitaxel, J. Pharm. Sci. aqueous media, Adv. Colloid. Interface Sci. 19 (1983) 253–307.
Technol. 52 (1998) 170–172. [60] J.S. D'Arrigo, C. Saiz-Jimenez, N.S. Reimer, Geochemical properties and
[38] P.P. Constantinides, K.J. Lambert, A.K. Tustian, B. Schneider, S. Lalji, biochemical composition of the surfactant mixture surrounding natural
W. Ma, B. Wentzel, D. Kessler, D. Worah, S.C. Quay, Formulation microbubbles, J. Colloid & Interface Sci. 100 (1984) 96–105.
development and antitumor activity of a filter-sterilizable emulsion of [61] J.S. D'Arrigo, Surface properties of microbubble-surfactant monolayers at
paclitaxel, Pharm. Res. 17 (2000) 175–182. the air/water interface, J. Colloid & Interface Sci. 100 (1984) 106–111.
[39] S.C. Spigel, S.F. Jones, F.A. Greco, J.D. Hainsworth, N.R. Dickson, [62] J.S. D'Arrigo, Stable Gas-in-Liquid Emulsions: Production in Natural Waters
N.T. Willcutt, S.W. Calvert, I.K. Rothman, D.A. Welker, J.L. Pratt, G.C. and Artificial Media, Elsevier Science Publishers, Amsterdam, 1986.
Brandt, H.A. Burris III, TOCOSOL Paclitaxel (S-8184 Vitamin E [63] J.S. D'Arrigo. Surfactant mixtures, stable gas-in-liquid emulsions, and
Paclitaxel Emulsion): Preclinical and Phase I Data, ASCO, 2002. methods for the production of such emulsions from said mixtures. U.S.
[40] A. Gorelov, S. Gorelov, P. Kavlov, O. Golubeva, M. Stewart, Paclitaxel Patent No. 4,684,479 (1987).
Injectable Emulsion: Phase 2a Study of Weekly Administration in Patients [64] J.S. D'Arrigo, T. Imae, Physical characteristics of ultrastable lipid-coated
with Metastatic or Locally Advanced Unresectable or Recurrent Urothelial microbubbles, J. Colloid & Interface Sci. 149 (1992) 592–595.
Transitional Cell Cancer (TCC), June 2004 ASCO, Abstract no. 4586. [65] J.S. D'Arrigo, Uptake of lipid-coated microbubbles by tumors: drug
[41] A. Lissianskaya, M. Gershanovich, N. Ognerubov, O. Golubeva, J. Pratt, delivery by receptor-mediated endocytosis, Bk. Abstr.: 216th Am. Chem.
Paclitaxel Injectable Emulsion: Phase 2a Study of Weekly Administration Soc. Meeting, American Chemical Society, Boston, August 1998.
in Patients with Platinum-Resistant Ovarian Cancer, June 2004 ASCO, [66] S. Vitols, G. Gahrton, A. Ost, C. Peterson, Elevated low density lipoprotein
Abstract no. 5047. receptor activity in leukemic cells with monocytic differentiation, Blood
[42] N. Bogdanova, S. Tjulandin, N. Ognerubov, V. Semlgazov, B. Afanasjev, 63 (1984) 1186–1193.
A. Mahson, D. Krasnozhon, G. Manlkhas, O. Vtoraya, I. Mitashok, [67] D. Gal, M. Ohashi, P.C. MacDonald, H.J. Buchsbaum, E.R. Simpson,
V. Astakhov, M. Byakhov, J. Smith, J. Pratt, M. Bolton, R. Daifuku, A Low-density lipoprotein as a potential vehicle for chemotherapeutic agents
Phase 2b Multicenter Evaluation of the Safety and Efficacy of TOCOSOL and radionucleotides in the management of gynecologic neoplasms, Am. J.
Paclitaxel as Initial Treatment of Patients with Metastatic Breast Cancer Obstet. Gynecol. 139 (1981) 877–885.
(MBC), 2005 SABCS, Abstract No. 1070. [68] S.G. Vitols, M. Masquelier, C.O. Peterson, Selective uptake of a toxic
[43] M.R. Dreher, W. Liu, C.R. Michelich, M.W. Dewhirst, F. Yuan, A. Chilkoti, lipophilic anthracycline derivative by the low-density lipoprotein receptor
Tumor vascular permeability, accumulation, and penetration of macro- pathway in cultured fibroblasts, J. Med. Chem. 28 (1985) 451–454.
molecular drug carriers, J. Natl. Cancer Inst. 98 (2006) 335–344. [69] S. Vitols, C. Peterson, O. Larsson, P. Holm, B. Aberg, Elevated uptake of
[44] A.K. Iyer, G. Khaled, J. Fang, H. Maeda, Exploiting the enhanced low density lipoproteins by human lung cancer tissue in vivo, Cancer Res.
permeability and retention effect for tumor targeting, Drug Discov. Today 52 (1992) 6244–6247.
11 (2006) 812–818. [70] M.J. Rudling, V.P. Collins, C.O. Peterson, Delivery of aclacinomycin A to
[45] J.S. D'Arrigo, R.H. Simon, S.Y. Ho, Lipid-coated uniform microbubbles human glioma cells in vitro by the low density lipoprotein pathway, Cancer
for earlier sonographic detection of brain tumors, J. Neuroimaging 1 Res. 43 (1983) 4600–4605.
(1991) 134–139. [71] S.K. Huang, K.D. Lee, K. Hong, D.S. Friend, D. Papahadjopoulos,
[46] R.H. Simon, S.Y. Ho, C.R. Perkins, J.S. D'Arrigo, A quantitative Microscopic localization of sterically stabilized liposomes in colon
assessment of tumor enhancement by ultrastable lipid-coated microbubbles carcinoma-bearing mice, Cancer Res. 52 (1992) 5135–5143.
as a contrast agent, Invest. Radiol. 27 (1992) 29–34. [72] S.M. Moghimi, J. Szebeni, Stealth liposomes and long circulating
[47] J.S. D'Arrigo, S.Y. Ho, R.H. Simon, Detection of experimental rat liver tumors nanoparticles: critical issues in pharmacokinetics, opsonization and
by contrast-assisted ultrasonography, Invest. Radiol. 28 (1993) 218–222. protein-binding properties, Progress. Lipid Res. 42 (2003) 463–478.
[48] R.H. Simon, S.Y. Ho, D.F. Uphoff, S.C. Lange, J.S. D'Arrigo, [73] M. Krieger, J. Herz, Structures and functions of multiligand lipoprotein
Applications of lipid-coated microbubble ultrasonic contrast to tumor receptors: Macrophage scavenger receptors, and LDL receptor-related
therapy, Ultrasound Med. & Biol. 19 (1993) 123–125. protein (LRP), Ann. Rev. Biochem. 63 (1994) 601–637.
[49] R.H. Simon, S.Y. Ho, J.S. D'Arrigo, A. Wakefield, S.G. Hamilton, Lipid- [74] M. Clement, W. Pugh, I. Parikh, Tissue distribution and plasma clearance
coated ultrastable microbubbles as a contrast agent in neurosonography, of a novel microcrystalline-coated flurbiprofen formulation, Pharmacolo-
Invest. Radiol. 25 (1990) 1300–1304. gist 34 (1992) 204.
[50] J.S. D'Arrigo, Contrast-assisted tumor detection, Drug News & Perspect. [75] D. Shenoy, S. Little, R. Langer, M. Amiji, Poly(ethylene oxide)-modified
4 (1991) 164–167. poly(beta-amino ester) nanoparticles as a pH-sensitive system for tumor-
[51] E. Barbarese, S.Y. Ho, J.S. D'Arrigo, R.H. Simon, Internalization of targeted delivery of hydrophobic drugs: part 2. In vivo distribution and
microbubbles by tumor cells in vivo and in vitro, J. Neuro-Oncology 26 tumor localization studies, Pharm. Res. 22 (2005) 2107–2114.
(1995) 25–34. [76] B. Rabinow, J. Kipp, P. Papadopoulos, J. Wong, J. Glosson, J. Gass,
[52] S.Y. Ho, E. Barbarese, J.S. D'Arrigo, C. Smith-Slatas, R.H. Simon, C. Sun, T. Wielgos, R. White, C. Cook, K. Barker, K. Wood, Itraconazole
Evaluation of lipid-coated microbubbles as a delivery vehicle for Taxol in IV nanosuspension enhances efficacy through altered pharmacokinetics
brain tumor therapy, Neurosurgery 40 (1997) 1260–1268. in the rat, Int. J. Pharm. 339 (2007) 251–260.
[53] J.S. D'Arrigo, Improved method for studying the surface chemistry of [77] E. Merisko-Liversidge, P. Sarpotdar, J. Bruno, S. Hajj, L. Wei, N. Peltier,
bubble formation, Aviat. Space Environ. Med. 49 (1978) 358–361. J. Rake, J.M. Shaw, S. Pugh, L. Polin, J. Jones, T. Corbett, E. Cooper,
[54] J.S. D'Arrigo, Y. Mano, Bubble production in agarose gels subjected to G.G. Liversidge, Formulation and antitumor activity evaluation of
different decompression schedules, Undersea Biomed. Res. 6 (1979) nanocrystalline suspensions of poorly soluble anticancer drugs, Pharm.
93–98. Res. 13 (1996) 272–278.
[55] J.S. D'Arrigo, Physical properties of the nonionic surfactants surrounding [78] K. Peters, S. Leitzke, J. Diederichs, K. Borner, H. Hahn, R. Muller,
gas cavitation nuclei, J. Chem. Phys. 71 (1979) 1809–1813. S. Ehlers, Preparation of a clofazimine nanosuspension for intravenous
[56] J.S. D'Arrigo, Structural features of the nonionic surfactants stabilizing use and evaluation of its therapeutic efficacy in murine Mycobacterium
long-lived bubble nuclei, J. Chem.Phys. 72 (1980) 5133–5138. avium infection, J. Antimicrob. Chemother. 45 (2000) 77–83.
[57] J.S. D'Arrigo, Aromatic proteinaceous surfactants stabilize long-lived [79] N. Scholer, K. Krause, O. Kayser, R.H. Muller, K. Borner, H. Hahn,
gas microbubbles from natural sources, J. Chem.Phys. 75 (1981) O. Liesenfeld, Atovaquone nanosuspensions show excellent therapeutic
962–968. effect in a new murine model of reactivated toxoplasmosis, Antimicrob.
[58] J.S. D'Arrigo, Glycoprotein surfactants stabilize long-lived gas micro- Agents Chemother. 45 (2001) 1771–1779.
bubbles in the environment, in: J.E. Zajic (Ed.), Microbial-Enhanced Oil [80] E. Olavarria, M. Hassan, A. Eades, C. Nilsson, A. Timms, J. Matthews,
Recovery, PennWell Press, Tulsa, 1983, pp. 124–140. C. Craddock, E. Kanfer, J. Apperley, J. Goldman. A phase I/II study of
P.P. Constantinides et al. / Advanced Drug Delivery Reviews 60 (2008) 757–767 767

multiple-dose intravenous busulfan as myeloablation prior to stem cell Ellerhorst, Phase I and pharmacokinetic study of ABI-007, a Cremophor-
transplantation. Leukemia. 14 (200) 1954–1959. free, protein-stabilized, nanoparticle formulation of paclitaxel, Clin.
[81] W. Gradishar, S. Tjulandin, N. Davidson, H. Shaw, N. Desai, P. Bhar, Cancer Res. 8 (2002) 1038–1044.
M. Hawkins, J. O'Shaughnessy, Phase III trial of nanoparticle albumin- [83] N.K. Ibrahim, B. Samuels, R. Page, D. Doval, K.M. Patel, S.C. Rao, M.K.
bound paclitaxel compared with polyethylated castor oil-based Nair, P. Bhar, N. Desai, G.N. Hortobagyi, Multicenter phase II trial of ABI-
paclitaxel in women with breast cancer, J. Clin. Oncol. 23 (2005) 007, an albumin-bound paclitaxel, in women with metastatic breast cancer,
7794–7803. J. Clin. Oncol. 23 (2005) 6019–6026.
[82] N.K. Ibrahim, N. Desai, S. Legha, P. Soon-Shiong, R.L. Theriault, E.
Rivera, B. Esmaeli, S.E. Ring, A. Bedikian, G.N. Hortobagyi, J.A.

You might also like