Drug delivery and drug targeting with parenteral lipid

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Journal of Controlled Release 223 (2016) 85–98

Contents lists available at ScienceDirect

Journal of Controlled Release

journal homepage: www.elsevier.com/locate/jconrel

Review article

Drug delivery and drug targeting with parenteral lipid


nanoemulsions — A review
Karl Hörmann a, Andreas Zimmer b,⁎
a
Fresenius Kabi Austria GmbH, Hafnerstraße 36, A-8055 Graz, Austria
b
Karl-Franzens-University of Graz, Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology, Member of BioTechMed Graz, Universitätsplatz 1, A-8010 Graz, Austria

a r t i c l e i n f o a b s t r a c t

Article history: Lipid nanosized emulsions or nanoemulsions (NE) are oil in water dispersions with an oil droplet size of about
Received 16 November 2015 200 nm. This size of oil droplets dispersed in a continuous water phase is a prerequisite for the parenteral, namely
Accepted 12 December 2015 intravenous administration. Many parenteral nutrition and drug emulsions on the market confirm the safe use of
Available online 14 December 2015
NE over years.
Parenteral emulsions loaded with APIs (active pharmaceutical ingredients) are considered as drug delivery sys-
Keywords:
Emulsion
tems (DDS). DDS focuses on the regulation of the in vivo dynamics, such as absorption, distribution, metabolism,
Nanoemulsion and extended bioavailability, thereby improving the effectiveness and the safety of the drugs. Using an emulsion
Intravenous as a DDS, or through the use of surface diversification of the dispersed oil droplets of emulsions, a targeted
Parenteral increase of the API concentration in some parts of the human body can be achieved. This review focuses on NE
Drug targeting similar to the marketed once with no or only low amount of additional surfactants beside the emulsifier from
Drug delivery system a manufacturing point of view (technique, used raw materials).
Manufacturing © 2015 Elsevier B.V. All rights reserved.
Materials
Ingredients

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
86
2. Classification of parenteral nanoemulsions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
3. Nanoemulsions for nutrition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
3.1. Biological fate of nanoemulsions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
3.2. Modification of the pharmacological profile of drugs owing to treatment with lipid nanoemulsions . . . . . . . . . . . . . . . . . . . . 87
4. Nanoemulsions as matrix for lipophilic APIs (drug delivery systems) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
4.1. Targeting liver and MPS (mononuclear phagocyte systems) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
4.2. Targets other than the liver . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
4.3. Droplet size . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
5. Nanoemulsion with cationic surface charge . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
6. NE with PEGylation on droplet surface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
6.1. Opsonization and phagocytosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
7. NE with PEGylation on droplet surface with targeting ligand . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
7.1. Peptide mediated targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
7.2. Antibody mediated targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
7.3. Sugar ligands and albumin as targeting moieties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
8. The concept of differential protein adsorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
9. Summary/conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96

⁎ Corresponding author.
E-mail address: andreas.zimmer@uni-graz.at (A. Zimmer).

http://dx.doi.org/10.1016/j.jconrel.2015.12.016
0168-3659/© 2015 Elsevier B.V. All rights reserved.
86 K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98

1. Introduction potential of the components, most usually surfactants (low-energy


emulsification). There are a number of processes for high-energy emul-
The aim of new research in this field is to achieve an improved sification, i.e. high-pressure homogenization (HPH), ultrasonication and
drug delivery and drug targeting by modifying existing and develop- microfluidization. Though this review is focusing on high-energy
ing new emulsions. Intravenous fat, also known as oil or lipid, emul- emulsification as used in the pharmaceutical industry for intravenous
sions have been in medical use for over 5 decades (e.g. Intralipid, emulsions, the low-energy emulsification processes are phase inversion
approved in Europe in 1962). Parenteral nutrition emulsions supply temperature (PIT), phase inversion composition (PIC) and solvent
critical ill patients, which cannot be fed orally, with triglycerides. diffusion [6].
These triglycerides are metabolized further to (essential) fatty During the microfluidization process the liquid is pumped
acids in the human body. The accepted regulatory status of the raw through an interaction chamber at high pressure. The liquid in
materials used for NEs and the safe metabolism is one of the advan- this chamber is then divided into two streams, which are brought
tages of NEs. Another important advantage is the capability of together with ultrahigh velocities [7]. High-pressure homo-
dissolving not water soluble APIs. Many novel therapeutic agents have genization, using a homogenizer from companies such as GEA,
poor or no water solubility. Formulating these substances as lipid NE APV, etc., transfer the liquid using high-pressure from plunger
can be a very smart way to overcome this hurdle and make the effect pumps through a small gap. Droplet size is reduced by high-shear
of these substances possible only. and cavitation forces more so then by high-speed collisions with
All NEs in this review are intended for intravenous administration other droplets which is the main force for microfluidization.
just like the marketed products. As an exception, Mendes et al. [1] Based on a classification of NEs from Tamilvanan et al. [8], parenteral
reported an NE composed of approximately 47.8% phospholipids, NEs can also be classified as:
48.0% cholesteryl oleate, 1.9% free cholesterol and 2.3% triacylglycerols
administered intralesionally to patients with breast cancer 12 h prior – NE for nutrition (phospholipid based)
to surgery directly into or near the tumor. Radioactive labeling with – NE as matrix for APIs (drug delivery systems)
14
C-cholesteryl oleate showed that peritumoral injection has the best – NE with cationic surface charge
concentration effect. Further examples of intratumoral and intramuscu- – NE with PEGylation on droplet surface
lar injections are given in the review on lipid carrier systems by Hashida – NE with PEGylation on droplet surface with targeting ligand.
et al. [2].

2. Classification of parenteral nanoemulsions


3. Nanoemulsions for nutrition
This review focuses on o/w (oil in water) emulsions (Fig. 1). There is
also some current research ongoing with non-aqueous and/or oil in oil The first attempt to administer oil parenterally was at the end of the
emulsions; for example a study [3] used castor oil in silicone oil emul- 17th century when English naturalist William Courten administered
sions. This strategy can be used as a DDS for lipophilic and/or hydrolyt- olive oil intravenously to a dog (1 g olive oil/kg dog). The dog went
ically unstable drugs for intramuscular injection. The release of the drug into severe respiratory distress and died, probably from lung emboli
is substantially slowed down when using such a DDS. [9]. Injection of pure oil is lethal.
For intravenous applications of emulsions, the dispersed oil droplets After further efforts in the following hundreds of years, the first safe
must be below the size of the smallest blood vessels to avoid embolisms. intravenous fat emulsion was developed by Arvid Wretlind and ap-
The smallest blood vessels are present in the lung with diameters down proved in Europe in 1962, and it is still marketed as Intralipid [10,11].
to 5 μm. This is in accordance with one of the parameters noted in the It is an emulsion used for parenteral nutrition and contains 10 or 20%
American Pharmacopeia, USPb729N, PFAT5. The percentage of fat drop- soya bean oil, 1.2% egg phospholipids as emulsifier/surfactant, 2.5%
lets greater than 5 μm must not exceed 0.05% of intravenous applied glycerin for tonicity and sodium hydroxide for pH adjustment (no
emulsions [4]. further ingredients).
The mean droplet size is, therefore, in the submicron area, about NEs for nutritional purposes have no active pharmaceutical ingredi-
200–400 nm and these emulsions are categorized as nanoemulsions ent (API). However, to understand the NE system it is worth having a
(NEs), as well as nanosized emulsions, submicron emulsions, look at this first marketed NE. The first successfully developed
miniemulsions, fine dispersed emulsions, etc. [5]. emulsions for parenteral/intravenous use were developed to meet the
The energy required to produce a NE can be generated by a nutritional needs of critically ill patients. Formerly, only glucose and
mechanical device (high-energy emulsification) or from the chemical amino acid solutions were available. These NEs provide the patient
with high caloric triglycerides and are an enormous benefit for patients
who must be fed parenterally.
As regards oil sources, soybean oil is very often used due to its high
content of essential C18 fatty acids like linoleic acid, and therefore called
long chain triglycerides (LCT). There are some concerns that a high
content of ω-6 polyunsaturated fatty acids may lead to negative side
effects in prolonged administration, such as immunosuppressive
actions (MPS impaired function) and inhibition of lymphocytes.
To overcome these possible negative side effects, NEs were
developed with a 1:1 mixture of LCT and medium chain triglycerides
(MCT; from coconut oil). Other commercially marketed NEs use struc-
tured triglycerides as an oil source, in which long and medium chain tri-
glycerides are bound to the same glycerol backbone, olive oil and, fish
oil. Fish oil is especially interesting owing to its high content of ω-3
polyunsaturated fatty acids, i.e. EPA (eicosapentaenoic acid) and DHA
(docosahexaenoic acid).
Fig. 1. Oil in water emulsion, stabilized through phospolipids on the surface with dissolved To stabilize the fine dispersed oil droplets in the continuous water
drug molecules (red dots), adapted from [4]. phase an amphiphilic emulsifier, which is located at the interface
K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98 87

between oil and water, is required due to their lipophilic and hydrophil- 3.2. Modification of the pharmacological profile of drugs owing to treat-
ic molecule section. ment with lipid nanoemulsions
Most of the NEs currently on the market use egg yolk phospholipids
as an emulsifier with the main chemical substance being phosphatidyl- There are some studies in which a marketed NE is used to alter the
choline. In a recent review on lecithin based NEs [12], the peculiarities of pharmacological profile of a drug or diagnostic. Liu et al. [18] used a pre-
lecithin as an emulsifier were presented. The seldom used soybean treatment with Intralipid to increase the blood half-time of nano- and
phosphatidylcholine is reported to be less prone to hydrolysis than micron-sized superparamagnetic iron-oxide particles. The proposed
egg yolk PC during shelf life [13]. The combination of egg yolk phospho- mechanism behind this is, that due to the pre-treatment with a lipid
lipids and synthetic surfactants are often used in research to further sta- NE, the MPS capacity for uptake is decreased, which means that
bilize critical NEs, e.g. PEGylated phosphatidylethanolamine (PEG-PE), other drugs vehicles can benefit from a pre-treatment with a well-
Pluronic® F68, etc. established and inexpensive NE.
For the adjustment of tonicity almost all marketed NEs use glycerin Assink et al. [19] showed that after a life-threatening intoxication
in a typical amount of 2.2–2.5% to achieve an osmolality in the same with verapamil, a calcium channel blocker, for which there is no
range as that of blood — around 300 mOsm/kg. antidote, a treatment with a lipid NE is beneficial.
Among the other ingredients of approved NEs are free fatty acids A further study, carried out by Fettiplace et al. [20], used bupivacaine
(oleic acid) as co-emulsifier, stabilizers such as EDTA, and antioxidants as an intoxication model drug. Therein, they reported on a proposed
such as α-tocopherol. mechanism of detoxification via transient drug scavenging and an addi-
tional cardiotonic effect of a lipid NE. This effect couples to accelerate
movement of the toxin away from organs influenced by the toxin. In
3.1. Biological fate of nanoemulsions vivo and in silico models of toxicity were combined and the contribution
of the multi-modal activity, i.e. scavenging and cardiotonic actions, of
An interesting question is, where the “life” of oil droplets of intrave- lipid NEs was tested.
nous emulsions ends and if this can be used for drug targeting. Accord- Another study with bupivacaine also underlined the enhanced elim-
ing to Rossi [14], oil droplets of an intravenous emulsion are cleared ination effect of lipid NE administration and examined the localization
either by the mononuclear phagocyte systems (MPS; e.g. Kupffer cells of the drug after enhanced clearance by the NE [21]. In a rat study
of the liver and splenic macrophages) or metabolized as endogenous they revealed that concentration of bupivacaine in the NE group was
chylomicrons. Chylomicrons (see Fig. 2) are about 75–1000 nm in size lower in heart, brain, lung, kidney, and spleen compared to the control
and metabolized in the blood stream, which results in a release of free group, but higher in the liver.
fatty acids from the triglycerides. The remnants of the chylomicrons
(30–50 nm) are cleared in the liver by low- density lipoprotein 4. Nanoemulsions as matrix for lipophilic APIs (drug delivery
receptors. systems)
There are many parameters that influence the biological fate of
emulsions' oil droplets in the human body, Table 1 lists a number The incorporation of lipophilic drugs in o/w emulsion is a smart way
of these. (For interpretation of the references to color in this figure to overcome solubility problems. However, the commercial use of
legend, the reader is referred to the web version of this article.) pharmaceutical drug NEs is limited to very few products; see the non-
The impact of long chain ω-3 polyunsaturated fatty acids (PUFA) exhaustive list below regarding the commercially available NE drug
on the metabolism was studied by Ton et al. [17]. Soybean oil (LCT), delivery systems (Table 2).
MCT/LCT/ω-3 (5:4:1, wt/wt/wt), and MCT/ω-3 (8:2, wt/wt) A great deal of research is ongoing in this field, mainly due to the fact
emulsions were prepared and radio-labeled with 1α,2α (n)-3 H that many of the newly discovered drugs have a low solubility in water.
cholesteryl oleoyl ether. In a mice in vivo study they found the The drug's lipophilicity incorporated in a lipid NE is an important factor
MCT/ω-3 (8:2, wt/wt) NE to be cleared more efficiently from blood here for biodistribution. If the lipophilicity is low, the drug may be
and that ω-3 PUFA seems to be directed more towards extrahepatic released quite quickly from the oil droplet to the blood stream, where
tissues. sink conditions exist due to the dilution of the administered drug in
the blood stream and subsequently further metabolized. If the lipophi-
licity is high, the drug may be retained in the oil droplet, so the oil
droplet can function as a drug delivery system or aid in drug
targeting. Takino et al. reported, that the log P, used as measuring
parameter for lipophilicity, should be N 9 to prevent fast release of
the drug to blood serum [23]. However, there are many other authors
proposing other log P limits, both lower and higher. Nevertheless, a
good strategy for increasing circulation time and consequently
changing the biodistribution of a drug may be the increase of the drug's
lipophilicity by esterification with a fatty acid such as oleate, palmitate,
valerate, etc.
Ganta et al. [24] developed a NE with the cancer treatment drug
chlorambucil and evaluated the pharmacokinetics and anticancer activ-
ity in mice. The NE consists of soybean oil (10%w/v) as the lipid phase,
and egg phosphatidylcholine (1.8% w/v) and cholesterol (0.2% w/v) as
the surfactant/co-surfactant and 0.2% w/v chlorambucil. All these
ingredients were dissolved in chloroform and formed a lipid film after
evaporation of the chloroform. After addition of the aqueous phase
containing glycerol (2.21%) for tonicity the emulsion was formed
using high-energy ultrasonication. The NE produced in the aforemen-
tioned manner, with a particle size of 182.7 ± 0.8 nm, was administered
Fig. 2. Schematic chylomicron structure with ApoA, ApoB, ApoC, ApoE (apolipoproteins); intravenously to C57 BL/6 male mice. The NE loaded with chlorambucil
T (triacylglycerol); C (cholesterol); green (phospholipids), from [15]. showed improved pharmacokinetics compared to chlorambucil
88 K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98

Table 1
Parameters influencing the metabolism as lipoproteins, the capture by the mononuclear phagocyte system (MPS) and the elimination from the blood circulation of NEs after parenteral
administration, adapted from [16].

Factors Metabolism as lipoprotein Capture by MPS Elimination from the blood


circulation

Poor Extensive Poor Extensive Slow Rapid

Particle size Large Small Small Large Small Large


Emulsifier DPPC EYPC DPPC DSPC DPPC EYPC
DSPC – SM – SM DSPC
SM – – – – –
Co-emulsifier Poloxamers – Poloxamers – Poloxamers –
HCO-60 – HCO-60 – HCO-60 –
PEG – PEG – PEG –
Polysorbates – Polysorbates – Polysorbates –
Solutol – Solutol – Solutol –
– – – – Cholesterol –
– – – – CO –
Cationic lipid SA/OA – SA/OA – SA/OA –
Oil phase LCT MCT – – LCT MCT
– – – – SLS SLM
Opsonization – – Low Large Low Large
Instability of oil droplets in blood stream – – – – Low Large
Overloading of MPS Large Small

DPPC, dipalmitoylphosphatidylcholine; DSPC, distearoylphosphatidylcholine; SM, sphingomyelin; EYPC, egg yolk phosphatidylcholine; HCO-60, polyoxyethylene-(60)-hydrogenated cas-
tor oil; PEG-PE, phosphotidylethanolamine derivative with polyethylene glycol; CO, cholesteryl oleate; SA, stearylamine; OA, oleylamine; LCT, long chain triglycerides; MCT, medium chain
triglycerides; SLS, structured lipid with short chain fatty acids; C8–C10; SLM, structured lipid with medium chain fatty acids, C4.

solution, e.g. AUC was roughly doubled (32.4 ± 0.1 μg/ml h vs. 16.9 ± activity compared with a Cheliensisin A injection solution consisting of
0.1 μg/ml h) and growth suppression rate of the colon-38 adenocarcino- cremophor:ethanol:phosphate-buffered saline PBS (1:1:5, v/v/v).
ma in the mouse was significantly greater. Clarithromycin is a semisynthetic antibacterial drug with low solu-
Commercial docetaxel injections contain ethanol and Tween 80, bility in long chain triglycerides like soybean oil and MCT oil. Tocopherol
whereas the latter is known to cause hypersensitivity. An alternative or its derivatives (tocol emulsions) can be used as another oil source. In
formulation of docetaxel in a NE [25] was reported to have the same the formulation optimization study of Li et al. [27] tocopherol succinate
anticancer efficiency while showing less toxicity. MCT oil containing is used to formulate clarithromycin in a less painful injectable NE. This
3% (w/v) oleic acid was used as a lipid phase to dissolve docetaxel. The was confirmed with a rabbit ear vein irritation test. The NE was com-
aqueous phase consisted of glycerin 2.5% (w/v) and the surfactants posed of tocopherol succinate, soybean oil, MCT oil and oleic acid in
lecithin and poloxamer 188 at 1.3% (w/v) each. Both phases were the oil phase, whereas oleic acid formed a lipophilic ionpair with
heated, mixed thoroughly and high-pressure homogenization, resulting clarithromycin and increased the drug loading of the NE. In the water
in a NE with a mean droplet size of 169 nm. phase glycerin was included for tonicity, poloxamer 188 as a non-ionic
An example of overcoming poor water solubility while also enhanc- surfactant for additional stability and NaOH for pH adjustment. Both
ing chemical stability is reported by Zhao et al. [26]. In this study, phases were mixed using an Ultraturrax, followed by high-pressure
Cheliensisin A, a novel anticancer drug derived from a natural source, homogenization.
was formulated in a lipid NE. Interestingly, to overcome any stability is- The non-steroidal anti-inflammatory drug (NSAID) diclofenac as a
sues, the lipid emulsion was lyophilized after filtration through a 0.8 μm water based solution has a short half-time of 1–2 h, which results in a
filter and finally gamma sterilized. As a lipid phase 200.0 g medium number of injections per day in the case of, for example, the treatment
chain triglycerides were chosen, in which 2.0 g Cheliensisin A, 40.0 g of arthritic conditions. The formulation of diclofenac as an acid in lieu
soybean phospholipids and 0.3 g vitamin E were dissolved. The lipid of the sodium salt in a lipid NE can be a solution to overcoming pain
phase was mixed with 600 ml double distilled water by a high-shear at the injection site and also reduced dosing. Ramreddy et al. [28] devel-
mixer, followed by high-pressure homogenization. After final dilution oped a 200–250 nm NE composed of 100 mg soya bean oil, 12 mg egg
to 1000 ml with double distilled water the cryoprotection agent sucrose lecithin, 2.5 mg diclofenac acid, α-tocopherol acetate 2 mg, cholesterol
was added (10% w/w). 0 to 3 mg, oleic acid 2.5 mg or, alternatively, stearyl amine 3 mg (to
Due to the formulation as a NE, the IC50 (50% inhibitory concentra- form negatively or positively charged NEs), 22.5 mg glycerin in double
tion) decreased to at least one third for several cancer cell types, most distilled water up to 1 ml. The oil phase and the water phase, including
cell types showed a remarkably lower decrease. The in vivo activity glycerin, were mixed at 70 °C and then sonicated. Incorporated
was studied in pulmonary metastasis of colon cancer-bearing BALB/c diclofenac acid NEs improved the pharmacokinetics in rats owing to
mice model, showing significantly increased life span and anti-tumor an increase in AUC, elimination half-life and MRT in comparison to the

Table 2
Marketed nanoemulsions as drug delivery systems, adapted from [22].

Drug Marketed name Manufacturer Market Indication

Alprostadil palmitate Liple® Mitsubishi Tanabe Pharma Japan Vasodilator, platelet inhibitor
Clevidipine Cleviprex® The Medicines Company US Calcium channel blocker
Dexamethasone-palmitate Limethasone® Mitsubishi Tanabe Pharma Japan Rheumatoid arthritis
Diazepam Diazemuls® Actavis Nordic Europe etc. Sedative
Etomidate Etomidat-Lipuro® Braun Melsungen Europe Anesthetic
Flurbiprofen axetil Lipfen® Green Cross Japan Nonsteroidal analgesic
Propofol Diprivan® Astra Zeneca Worldwide Anesthetic
Vitamins A, D, E, K Vitalipid® Fresenius Kabi Europe etc. Parenteral nutrition
K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98 89

commercial diclofenac solution Voveran®. The use of cholesterol should homogenization and had a surface potential of −45 mV. The anticancer
form a more rigid surface of the oil droplet than when egg lecithin is activity of these NEs was tested on human mammary tumor cells. The
used alone. However, the diclofenac levels in blood serum were lower IC50 of NE against the human mammary tumor cells MCF-7 and
for cholesterol containing NEs, which may be attributed to enhanced MDA-MB-231 with only TRF was 14 and 7 μM. The IC50 decreased to
uptake of oil droplets through LDL receptors in tissues like liver, kidney, 10 μM and 4.8 μM for those NEs containing additional simvastatin.
brain, spleen, etc. Positively charged NEs showed lower blood serum As an alternative to inhalation, an NE as drug delivery system can be
levels, also when compared with the plain NE without cholesterol and used for isoflurane, a halogenated ether administered for general
stearyl amine. This may be explained by an increased uptake of positive- anesthesia [35]. The NE was produced with the high-pressure homoge-
ly charged NE in negatively charged cell membranes. nization technique and had a droplet size of 150 ± 0.78 nm. The lipid
A poorly water-soluble anti-cancer substance isolated from the tra- phase was formed by soybean lecithin S75 (1% w/v) in 15 ml MCT oil,
ditional Chinese herb magnolia, Honokiol, was studied as both a solu- isoflurane was added just before homogenization with the water
tion and an NE [29]. The NE was prepared with high-pressure phase. The water phase consisted of polysorbate 80 (2% w/v), sorbitol
homogenization and consisted of soybean lecithin, Synperonic F68 (2% v/v) and 64.5 ml water.
and glycerol as the water phase and soybean oil with dissolved Within preclinical evaluation, the isoflurane emulsion showed
Honokiol as the lipid phase. Here again, the half-life of the 187 nm NE significantly decreased necessary doses compared to inhaled isoflurane
was substantially longer than that of the solution (8.0 min versus at the same levels of clinical biomarkers. However, minor adverse
4.3 min), AUC was greater and plasma clearance reduced. The tissue dis- effects like tachypnea, edema, and erythema were recognized when
tribution results in tumor-burdened mice indicated that the NE has bet- isoflurane-loaded nanoemulsions were administered.
ter targeting properties for lung, brain and tumor tissues than a Chen et al. [36] prepared a disulfiram-loaded lipid emulsion in his
Honokiol solution. study. Incorporating the cancer drug disulfiram in a lipid emulsion
seems to minimize the instability of the drug in blood. The optimization
4.1. Targeting liver and MPS (mononuclear phagocyte systems) of the NE formulation finally ended in the following composition
resulting in better stability during autoclaving and storage under heat
Through incorporation of lipophilic drugs in oil droplets of an o/w stress: 10% (w/v) MCT oil, 0.6% (w/v) soybean phosphatidylcholine
emulsion the distribution of the drugs in the human body can be S100, 0.6% (w/v) Pluronic F68, 0.2% (w/v) oleic acid, 2.25% (w/v)
influenced. Using emulsions as drug delivery systems can increase the glycerin, and 0.5% (w/v) disulfiram. Interestingly, in this study CO2 and
concentration of the drug in liver and/or MPS enormously. a buffer were used to maintain a stable pH value at the optimal range
A study with primaquine [30], a drug used to treat various stages of of 4.5 to 5, whereas in most NEs on the market sodium hydroxide is
parasitic malaria, showed that incorporation of the drug in an emulsion, used to adjust the pH. The emulsion was prepared using a high-shear
versus the solution, enhanced the liver uptake by a factor of 2.5. The mixer and high-pressure homogenization.
chylomicron-like emulsion was prepared from olive oil, phosphatidyl- Tumor cells grow rapidly and, consequently, have a great need for
choline (chicken eggs), lyso-phosphatidylcholine, cholesterol, and cholesterol to build new cell membranes. Natural carriers of
cholesteryl oleate by thin lipid layer hydration, followed by vortexing cholesterol-esters in the blood are low-density and high-density
and sonication. The API primaquine bisphosphate was complexed lipoproteins (LDL and HDL). Shawer et al. [37] built on the known
with Sodium lauryl sulfate and egg phosphatidic acid respectively and theory of an increased number of LDL-receptors for the development
co-dissolved with the lipid fraction of the emulsion. of a NE drug delivery system. Cholesteryl ester of carborane, cholesteryl
A new 2-(allylthio)pyrazine (2-AP)-loaded NE, a drug which has 1,12-dicarba-closo-dodecaborane-1-carboxylate (BCH) was incorporat-
hepatoprotective effects against toxicants, was developed by Jang et al. ed as drug in the NE, which can be used for boron neutron capture ther-
[31], varying oil to drug ratio, oil to lecithin ratio and tested some apy of cancers. The NE was produced as follows: all lipid fractions were
additional co-surfactants. The resulting NE consisted of 2-AP (1%), soy- dissolved in chloroform and mixed together in a ratio (w/w) of:
bean oil (4%), soybean lecithin (4%) and 91% water and was found to be triolein:egg phosphatidylcholine:lysophosphatidylcholine:cholesterol
isotonic. In comparison with a 2-AP solution, the 2-AP emulsion showed oleate:cholesterol:BCH, 70:22.7:2.3:3.0:2.0:2.0, respectively. After re-
significantly faster clearance of the blood stream and higher accumula- moval of the solvent, the water phase was added and the mixture was
tion in the organs, especially the liver (about double the amount com- sonicated to obtain the final NE. In in-vitro studies, the interaction of
pared to solution). this developed NE with human lipoprotein was shown by electrophore-
An NE loaded with SQ641, which is the most potent analogue of sis. Additionally, a sufficient uptake of BCH for cancer therapy was
capuramycin antibiotics and has a bactericidal effect against Mycobacte- achieved, or at minimum an absorption on rat 9 l glioma cells.
rium tuberculosis, was reported to target lung and spleen in a tuberculo- Another example of the reduction of toxic effect by incorporating a
sis mouse model [32]. Interestingly, SQ641 itself was not active in vivo drug into a NE is described by [38]. In this study, the cancer treatment
which may be caused by its low water solubility. The NE was made of substance N-oleyl-daunorubicin was formulated in a cholesterol-rich
phospholipid Phosal 53 MCT (Lipoid GmbH, Ludwigshafen, Germany) NE that binds to low-density lipoprotein receptors. The maximum
using α-tocopheryl polyethylene glycol 1000 succinate (TPGS) as tolerated dose was found to be 65-fold higher than commercial Dauno-
surfactant. rubicin; moreover, the anti-cancer effects, namely tumor growth inhibi-
tion and survival rates, were better. This is also a further example of
4.2. Targets other than the liver increasing the lipophilicity of a molecule by addition of a fatty acid
ester. Oleyl is derived from oleic acid C18H34O2, which is classified as a
NEs could also be used as drug delivery systems to target tissues in monounsaturated omega-9 fatty acid, abbreviated with a lipid number
the body other than lung and MPS. A cancer treatment was proposed of 18:1 cis-9.
by Alayoubi et.al., using tocotrienol rich fraction (TRF) of vitamin E A cholesterol rich NE comprising paclitaxel oleate was used for the
[33] and TRF and simvastatin [34]. In a later study, a viscous 70/30 treatment of atherosclerosis in rabbits [39]. In this study some of the
blend of TRF and medium chain triglycerides (MCT, Miglyol 812) was rabbits were fed a 1% cholesterol diet, which caused atherosclerotic
used as a lipid phase of the NE, in which simvastatin was dissolved at lesions. In the aortic arch of cholesterol-fed rabbits, the uptake of radio-
9% w/w loading in some formulations. Phospolipids (Lipoid E80S, active labeled NE with 14C-cholesteryl oleate was double that observed
composed of 64–79% phosphatidylcholine and 12–18% phosphatidyl- in animals fed only regular feed. The treatment with NE with paclitaxel
ethanolamine), Tween 80 and poloxamer 188 were used as surfactants. oleate reduced the lesion areas of cholesterol-fed animals by 60% and
The approximately 200 nm sized NE was prepared by high-pressure inhibited smooth muscle cell proliferation, without showing toxic
90 K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98

effects. Consequently, this drug delivery system appears to be a promis- lipid film was rehydrated and vortexed rigorously. After high-pressure
ing cardiovascular therapy approach minus toxicity effects. homogenization the DOX-OA NE was obtained. A pharmacokinetic
The same NE was also used in a study in eight patients with gyneco- study was conducted in mice showing significantly higher AUC and cir-
logic cancers [40]. The NE was prepared from 40 mg cholesteryl oleate, culation time in blood than DOX solution and a positively altered tissue
20 mg egg phosphaditylcholine, 1 mg triolein and 0.5 mg cholesterol as distribution. The DOX concentration in the heart was found to be lower
the lipid phase, which was sonificated in water and homogenized in a than for the DOX solution. The dosage of DOX is limited due to its known
two-step process with KBr to obtain a cholesterol rich NE. Interestingly, cardiotoxic side effects. Interestingly, the release pattern of the DOX-OA
paclitaxel oleate was added to the finished NE as an ethanolic solution. NE was biphasic with a “burst” release in the beginning and a sustained
After sonification the liquid was centrifuged to remove unbound pacli- release until end. This may be explained by DOX-OA, which is located in
taxel oleate. A double labeling was performed using 3H-paclitaxel oleate the oil droplet core but also at the surface within the egg phospholipids
and 14C-cholesteryl ester. Comparing blood decay curves of both labels, layer. Compared to literature data of PEGylated liposome the DOX-OA
which are in principle identical, showed that the drug paclitaxel oleate NE showed shorter circulation time. This may be related to the above-
stays within the NE drug delivery system until the end. Further, an al- mentioned “burst” release portion and the absence of stealth agents
most identical increase of concentration in the cancer tissue of 3.6 like PEG and Poloxamers.
times (compared with commercial paclitaxel) and 3.5 times for the cho-
lesterol rich NE confirm that the whole drug delivery system is incorpo-
rated by cancer cells, supported by overexpression of low-density 4.3. Droplet size
lipoprotein receptors which internalize cholesterol containing NE oil
droplets. Size is an important factor for the biodistribution of the oil droplets
A new NE, incorporating the anti-epileptic drug carbamazepine for a in the blood stream. As discussed before under biological fate of NEs,
drug delivery to the brain, was developed by [41]. 1-O-alkylglycerol/lec- the oil droplets of intravenous emulsion (which are around 200 nm)
ithin blends were used as emulsifiers, e.g. 0.25% w/w soy lecithin and are captured by the MPS quiet fast. Smaller oil droplets, or general nano-
1.25%w/w 1-O-decylglycerol. Soybean oil (10% w/w) as the lipid phase particles, stay longer in the blood circulation because they are not recog-
dissolved the emulsifier and 0.1% w/w carbamazepine. After adding nized by the MPS. Besides that, the enhanced permeation retention
the water phase containing glycerol (2.5%w/w), the emulsion was (EPR) effect is found and used in cancer research (Fig. 3). Tumor tissue
formed by high-pressure homogenization. In a tissue distribution is fast growing and has therefore defective vascular architecture. This
study in mice showed a significant uptake in all tissues for all NEs tested. and impaired lymphatic drainage let nanoparticles diffuse into tumor
1-O-alkylglycerols are reported to improve the brain delivery, which tissue and let them concentrate there.
was best shown using the NE containing 1-O-decylglycerol, which in- Seki et al. [47] reported on artificial lipoprotein-like particles, lipid
creased the brain availability 2.37 times. However, the tissue ranking nano-sphere (LNS®). These LNS are 25–50 nm in diameter. They are
was still lung N liver N spleen N brain N kidney N heart. composed of soybean oil and egg lecithin like commercially available
A study which also targeted the brain was carried out by Wen et al. emulsions for parenteral nutrition (e.g. Intralipid). The soybean oil and
[42], which compared solid lipid nanoparticles (SLN), nanostructured egg lecithin ratio differs as follows, for LNS the ratio is 1:1, whereas in
lipid carriers (NLC), and NEs. These three categories where produced commercial emulsions the ratio is 1:0.12. Additionally, the high-
by using 100% cetyl palmitate, different ratios of cetyl palmitate and pressure homogenization for this LNS is done at 1000 bar, whereas the
squalene and 100% squalene as the lipid phase, respectively. To obtain NE used for comparison is carried out at 500 bar. It seems that this is
a positive charge for all three categories of nanoparticles, Forestall, a cat- the reason for the enormous size reduction of 200–300 nm of commer-
ionic surfactant, was used. As a dye, sulforhodamine B was incorporated cial emulsions to 25–50 nm of LNS.
for imaging. An in vivo study in rats investigating the accumulation of The biological destination of oil droplets from parenteral nutrition
the dye in the brain revealed that the retention time was most emulsion with diameters of 200–300 nm is, as mentioned above, the
prolonged, from 20 to 50 min, using NEs. It seems that due to the MPS and liver. Further, these droplets are cleared from blood circulation
flexibility of NE's oil droplets, compared to more rigid nanostructured quite rapidly. LNS, on the other hand, showed much higher levels in the
lipid carriers and solid lipid nanoparticles, the uptake into the brain blood (AUC three times higher than NEs with 200–300 nm) and much
overcoming the brain blood barrier is enhanced. lower levels in the liver and spleen. Some other studies describe larger
A breviscapine, also known as scutellarin, a substance from a particles or oil droplets being eliminated from circulation by macro-
traditional Chinese herb, lipid emulsion was evaluated in an in vivo phages in a more pronounced way than smaller particles.
pharmacokinetics study in rats [43]. The lipid phase consisted of For example, Qi et al. [48] studied lipid emulsions made of different
soybean oil (10.0%), oleic acid (0.9%), lecithin (1.5%), and poloxamer oils (LCT, LCT/MCT, LCT/MCT/fish oil, fish oil) and saw a significant dif-
188 (0.4%). The latter two were used as surfactants. The water phase in- ference (p b 0.01) between large- and small-sized emulsions containing
cluded glycerol (2.5%). In comparison to the commercial product, the same oil mixture. Unfortunately, the exact sizes were not measured;
breviscapine injection, the NE has about 1.5 fold higher AUC. In a further they were compared with gel chromatography. The difference in drop-
study dealing with breviscapine NE, the AUC could be increased more let size was achieved by oil to egg yolk phosphatidylcholine weight ratio
than 14 times [44]. The main exposure of breviscapine was found in of 4:1 for the large-sized emulsions and 1:1 for the small-sized
blood plasma and lungs. emulsions. As the surface of the oil droplets in both formulations is cov-
In the study carried out by Zhang et al. [45], a lipid NE with the anti- ered by egg lecithin molecules, the change in biodistribution here is only
cancer drug Doxorubicin was developed with a diameter of approx. influenced by the size of the oil droplets.
200 nm, where focus was put on the utilization of raw materials An interesting in vitro and in vivo study about the influence of the
which are already approved for intravenous use and a production pro- droplet size on the anti-allergic and anti-inflammatory activities of a
cess which is easy to scale-up (high-pressure homogenization). To im- curcumin NE was conducted by Onodera et al. [49]. The tested curcumin
prove the poor lipophilicity of Doxorubicin (logP − 1.32 for the free NE was prepared by a thin-film hydration method. Soybean oil and
base) an ionic complex was formed with oleic acid (DOX-OA, logP of HEPC were dissolved in the organic solvent chloroform, as curcumin
1.81), which led to an increase of entrapment efficacy from about 30% and Tween-80 separately. All three solutions were mixed and dried
(DOX alone) to over 90% for DOX-OA. This complex (300 mg DOX-OA) afterwards by rotary evaporation and vacuum desiccation. The so ob-
was dissolved in 200 ml ethanol containing 600 mg lipoid E80, 50 mg tained dry thin film was hydrated with distilled water and sonicated
vitamin E, and 400 mg soybean oil. The solvent was eliminated with a at 25–55 °C till the desired oil droplet size of 50 nm, 100 nm and
rotary evaporator resulting in a thin lipid layer. With 50 ml water this 200 nm was obtained. Remarkable, both in vivo tests in orally fed
K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98 91

Fig. 3. Schematic illustration of the enhanced permeation and retention effect (EPR), from [46].

mice and in vitro cell culture tests revealed the advantageous properties mechanisms to increase the half-time blood circulation time of NEs
of 100 nm curcumin NE despite curcumin NE with 50 nm or 200 nm. the enhancement of hydrophilicity is due to:
For drug delivery systems, an increased circulation time is often re-
quired to develop their desired capabilities, e.g. to store an API within - polyoxyethylene (POE)
the lipid core for a certain time period and maintenance of a (constant) - or polyethylene glycol (PEG) chains on the surface.
release from the oil droplet to the blood and surrounding tissue. Or, for
the altered biodistribution, especially when the MPS like liver or spleen
is not the primary target, there is some time needed to achieve an The surface can be modified by adding an emulsifier with such POE
accumulation effect in the desired parts of the body. Particle and/or oil or PEG chains in the manufacturing process. Alternatively, the surface
droplet size is an important factor; additional influencing parameters of an existing NE can be modified by the simple addition of other surfac-
are listed in Table 1. tants or stabilizers. After an incubation time (24 h under gentle mixing
is often used) the additional surfactant is admixed to the existing NE.
5. Nanoemulsion with cationic surface charge The addition or substitution of surfactant molecules follows the Nernst
equation. According to Nernst, partitioning coefficients between the
Cationic surface charges on nanoparticles, in general, are supposed water phase and the stabilizer layer of the NE of all surfactants in the
to support the transfer of such nanoparticles through negatively system forms the surface of the NE [51]. This elegant method is used
charged bio-membranes. However, in the last few years, research with e.g. for incorporating PEG2000-DSPE into an existing NE.
these cationic NEs has diminished. Bruxel et al. [50] investigated cation- POEs are often used in the form of triblock-copolymers, marketed as
ic NEs as oligonucleotide delivery systems for the treatment of Plasmo- Poloxamers or Pluronics. In Table 3 commonly used surfactants from
dium falciparum topoisomerase II (Malaria). Oligonucleotide alone this group are listed with their most important characteristics and
show increased degradation in the body and their ability to enter infect- main application areas. According to the schematic structure below, it
ed red blood cells is very low due to their high molecular weight and is obvious that the inner, hydrophobic part will be located at the surface
hydrophilicity. of the oil core, whereas both hydrophilic POE chains will orientate to-
The NE composed of 8% MCT oil, 2% egg lecithin, 2.25% glycerol, and wards the continuous water phase. By the thus increased hydrophilicity,
water to 100% was produced by spontaneous emulsification. A clear in comparison to the most used EYP of the oil droplet, the elimination
ethanolic solution of the lipid phase was slowly added to the rate of the NE is decreased. A recent review on triblock-copolymers
water phase. After removal of the solvent ethanol under 50 °C with and their interface-interactions was provided by Torcello-Gómez et al.
reduced pressure the emulsion was formed. The positive surface [52].
charge of the NE was realized by adding 0.132% 1,2-dioleoyl-3- Of the abovementioned mechanisms the PEGylation is the most
trimethylammonium-propane (DOTAP). Different +/− charge ratios commonly used. PEGylation simply refers to the coverage of a particle
were adjusted by adding different amounts of negatively charged oligo- or oil droplet surface by the covalently grafting, entrapping, or
nucleotide, which formed together with the positively charged NE ion adsorbing of PEG chains (Fig. 5). The first PEGylated drug was developed
pairs. Complexes with a high charge ration (+6/−1) and/or the cation- in 1970 to make use of the advantages of PEG: very good solubility in
ic NE alone caused increased hemolysis. This is attributed to the pres- hydrophilic and hydrophobic phases, high hydration resulting in a
ence of more cationic charges, which can interact with the red blood high hydrodynamic volume, non-toxic and non-immunogenic, FDA ap-
cells. Parasite growth and reinfection capacity was evaluated in vitro, proved material for internal use, and available in different molecular
whereas the best results were obtained with the highest charge ration weights with low polydispersity [53].
(+4/−1), which showed no increased hemolysis. To be successful in prolonging the circulation time there are a num-
ber of points to consider. The molecular weight of the PEG chain must be
6. NE with PEGylation on droplet surface 2000 or greater, as indicated by most studies for polymeric nanoparti-
cles. Further, the PEG-chain density on the particle surface and the con-
To achieve targets in the human body other than MPS or liver with formation can contribute to the stealth effect. An optimal surface
NE, an extension of blood circulation time is required. This also means coverage is achieved when there are no gaps in the PEG-chain coverage
that clearance rate is decreased, which may prevent toxic effects due of the particle and the density is relatively low to allow the single PEG-
to less accumulation in the clearance tissues, too. As regards important chains free movement. This leads to a “mushroom” configuration,
92 K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98

Table 3
Physicochemical characteristics of polyoxyethylene (Pluronics and Poloxamers), adapted from [52].

Pluronic Poloxamer MW CMC at 37 °C POE units POP units HLB Administration (in vivo studies)

L61 P181 2000 1.1 ∗ 10−4 2 30 3 Subcutaneous, oral


P85 P235 4600 6.5 ∗ 10−5 26 40 16 Intravenous, intraperitoneal, subcutaneous, ocular
F68 P188 8400 4.8 ∗ 10−4 75 29 29 Parenteral, ocular, percutaneous, topical, oral, rectal
F127 P407 12,600 2.8 ∗ 10−6 100 65 22 Intravenous, intramuscular, subcutaneous, percutaneous, oral, ocular, buccal, rectal, topical

Pluronic, tradename; Poloxamer, generic term; MW, molecular weight; CMC, critical micelle concentration in mol/l; POE, polyoxyethylene; POP, polyoxypropylene; HLB, hydrophilic-li-
pophilic balance.

where parts of the PEG-chain are freely moveable, which is a low energy added as surfactant. DOTAP, placed on the surface of the NE contributes
conformation. Attachment of opsonins would compress these PEG- to the cationic charge of the whole vehicle, which should ease the trans-
chains, reducing their freedom to move and increasing their energy con- fer through the membrane of the endothelial cells. Also placed on the
formation. This energy change produces an opposing repulsive force, surface is PEG2000-DSPE, which may help to prolong the circulation
which can be higher than the attractive forces between opsonin and time in the vasculature. The NE was produced via microfluidization
particle. At higher PEG-chain density only a “brush” configuration is technique.
possible due to limited space available between the PEG-chains [54]. Improving the chlorambucil NE in the previous study [24], Ganta
The biodistribution of NEs coated with phosphatidylethanolamine et al. also decided to add PEG2000-DSPE to the chlorambucil NE to
derivatives with three different molecular weight PEGs was studied by enhance the circulation time in the body and, at the same time, the
Liu and Liu [55]. Castor oil was used as the lipid phase, different mix- pharmacokinetics and tissue distribution [58]. The chlorambucil NE
tures of surfactant were added to the oil in weight ratio 1.5:0.6, whereas with PEG2000-DSPE showed improved pharmacokinetics when given
the 0.6 consists of egg yolk phosphatidylcholine to 100% or it was divid- intravenously to C57 B/6 mice compared to plain chlorambucil NE or
ed into 0.2 phosphatidylcholine and 0.4 PEG-PE with different chain chlorambucil solution. For example, the AUC was 1.7-fold higher com-
lengths. The NEs, with about 200 nm average diameter, were prepared pared to NE or 2.7-fold higher compared to the solution. The half-time
by dissolving the lipids in chloroform. After removal of the solvent, the was increased 1.3-fold compared to NE and 7.6-fold compared to
lipid film was resuspended in PBS, pH 7.4, vortexed and subsequently solution.
homogenized with a Tissue-Tearor. Paclitaxel, one of the most commonly used anti-cancer agents, was
Among the tested molecular weights of PEG (1000, 2000 and 5000), used in another study for incorporation in an NE with a particle size of
the PEG-2000 was able to extend the circulation time of NEs best, which 50 nm [59]. The commercially available formulation consists of
was accompanied by a decrease in liver accumulation. PEG-5000 was at Cremorphor EL and ethanol (50:50% v/v) due to the low water solubility
the same level, whereas PEG-1000 was at the level of the NE with PC of paclitaxel. To increase the solubility of paclitaxel in lipid emulsions, a
only. The most likely reason for the prolonged circulation time is the in- prodrug, paclitaxel oleate was used. Besides eliminating the toxic effects
creased hydrophilicity of the oil droplet surface. Further, the formation of the Cremorphor EL/ethanol mixture, the NE with paclitaxel oleate
of a steric barrier may also contribute to this effect. demonstrated significantly greater AUC, higher Cmax and lower Vss in
Alayoubi et al. [56] investigated the effect of poloxamer or a pharmacokinetic study in rabbits. The NE was composed of 5.6 μmol
1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- triolein, 6.4 μmol egg phosphatidylcholine, 1.52 μmol polysorbate 80
[amino(polyethylene glycol)2000] (PEG2000-DSPE) surfaces of (poly-oxyethylenesorbitan monooleate), 0.36 μmol PEG-PE, and
tocotrienol-rich fraction's NE. Though the PEG molecules amount was 120 nmol paclitaxel oleate (or unesterified paclitaxel). All these ingredi-
equimolar in both NEs, the different observed effects were found to be ents were dispersed in a solvent and mixed together. After evaporation
determined by different steric confirmation of the PEG chains. of the solvent, the lipid fraction was lyophilized. PBS was added as the
PEG2000-DSPE has more dense brush confirmation than poloxamer water phase, the mixture was vortexed and then sonicated.
and is believed to have more repellent action against other hydrophobic In a newer study [60], Paclitaxel was used without derivatization
surfaces, e.g. red blood cells and blood proteins like immunoglobulin G into a prodrug. In a pharmacokinetic study in mice, same sized
(IgG). Accordingly, this PEG2000-DSPE NE has a 7-fold increased liposomes and NE (around 100 nm) were prepared. The emulsion
half-life time after i.v. injection in rats. Poloxamer NEs, however, consisted of egg yolk phosphatidylcholine, Tween 80, Tricaprylin (C8)
showed higher uptake and lower IC50 against MCF-7 tumor cells and Tricaproin (C6), and Paclitaxel in a weight ratio of
in vitro. 160:120:300:100:1.2. For the manufacturing of the PEGylated NE an ad-
Desphande et al. [57] engineered an omega-3 polyunsaturated fatty ditional PEG-DSPE was added to the mixture (5 mol%). The lipid phase
acid (PUFA) emulsion incorporating 17β-estradiol and C6-ceramide for was dissolved in chloroform and mixed. The dried oil phase was
possible treatment of arterial restenosis. All three substances were mixed with the water phase including 2.25% (w/v) glycerol. Afterwards,
reported to have, in principle, a positive effect on restenosis. However, the mixture was sonificated. The outcome of this study was that the NE
extensive protein binding and their high lipophilicity hampered the released the Paclitaxel in the blood circulation very rapidly, even using a
substances in reaching their cellular targets. A NE tailored as a drug PEGylated NE, while the liposome formulation performed quite well.
delivery system for omega-3 PUFA, 17β-estradiol and C6-ceramide, This poor performance of the NE may be contributed to the only slightly
which delivers the three substances specific to endothelial cells and lipophilic drug (log p 4.7), which can be related to a biofate more similar
arterial vascular smooth muscle cells, was achieved in this report. to water based solution and/or less probably the usage of triglyceride
Flaxseed oil rich in omega-3 PUFAs was used as an oil phase, and egg with relatively short chains (C6, C8) which may also contribute to a
phosphatidylcholine (Lipoid 80, DOTAP and PEG2000-DSPE were faster clearance of the blood stream due to faster lipid metabolism.
K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98 93

An interesting approach using a two vial formulation of Paclitaxel • Hydrophilicity: The blood serum proteins adhere easier on hydropho-
(25 mg/ml) in PEG400 solution and a commercially available 20% bic surfaces. A hydrophilic surface, which is realized e.g. with PEG
emulsion (LCT:MCT, 1:1, w/w) was investigated by Jing et al. [61]. chains and poloxamer 188, suppresses the attachment of the opsonins
Prior to application, the vial containing Paclitaxel was diluted with the on the surface [54].
emulsion to obtain a 1 mg/ml Paclitaxel emulsion. In comparison to a
conventional Paclitaxel-loaded NE [59], this two-vial formulation A successful drug targeting to the heart in an in vivo study in rats was
showed a lower clearance by MPS and higher accumulation in tumors. shown by using TPGS (D-alpha-tocopheryl polyethylene glycol 1000
Moreover, the anti-tumor efficiency was increased in A2780 and Bcap- succinate), a derivate of the natural vitamin E. In a study with a coen-
37 bearing mice. These advantageous effects of the two-vial formulation zyme Q10 loaded NE TPGS-NE showed better drug targeting properties
seem to be attributed to a greater amount of Paclitaxel in the phospho- to heart tissue compared to a lecithin stabilized NE [65]. The NE was pre-
lipid layer and a smaller proportion in the oil core than a conventional pared with hot high-pressure homogenization, whereas 3 g coenzyme
NE, where Paclitaxel is dissolved in the oil core. Q10 and 3 g TPGS or lecithin were dissolved in 3 g of caprylic/capric
To overcome the extremely short half-life of lycobetaine in blood, an triglyceride which together form the lipid phase. The lipid phase was
anti-cancer drug which shows significant cytotoxic activity against at added to 100 ml of a 45% w/w solution of glycerol in water and mixed
least Lewis lung carcinoma, a lipid NE was developed [62]. A lycobetaine to form the pre-emulsion at 60 °C. The NE was homogenized at
ion pair complex with oleic acid (LBT-OA) was formed by 1200 bar with 6 cycles, ending up in about 50 nm mean oil droplet
co-precipitation method, a strategy that has already been reported to size. The TPGS-NE showed a 2.8 times higher concentration of the en-
increase the lipophilicity of a drug [45]. The NE was prepared by a dogenous antioxidant coenzyme Q10 in the heart compared to lecithin
lipid film hydration, high-pressure homogenization method. LBT–OA, stabilized NE five minutes after intravenous administration to rats.
formed from 100 mg lycobetaine and 250 g oleic acid, was dissolved Higher concentration levels were maintained after 90 min, in summary
with 800 mg phospholipids (lipoid 80) and 500 μl structured triglycer- NE-TPGS had a 2.3-fold greater AUC. The tissues distribution rank order
ides in 200 ml dichloromethane. The solvent was removed by rotary was heart N liver N kidney.
evaporation. The obtained lipid film was re-suspended in 50 ml water,
followed by strong vortexing and high-pressure homogenization. In 7. NE with PEGylation on droplet surface with targeting ligand
contrast to the described LBT-OA-NE, the LBT-OA-PEG-NE was
produced in the same way, but using 760 mg lipoid E80 and 150 mg To improve the specificity of drug targeting systems, many studies
PEG2000-DSPE instead of 600 mg lipoid E80. were carried out with a targeting ligand on the surface of nanoparticles
The biodistribution and in vivo pharmacokinetics was studied in (“active targeting”). To the best knowledge of this author, only these
rats. A remarkable increase of AUC could be achieved by LBT-OA-PEG- few studies of NEs with targeting ligands have been reported so far.
NE compared with LBT-OA-NE and lycobetaine solution (3452.09 mg/
l ∗ minute, 1208.16 mg/l ∗ minute and 112.99 mg/l ∗ minute, respective- 7.1. Peptide mediated targeting
ly). Further, LBT-OA-PEG-NE achieved high concentration in the lung,
whereas lower concentration was observed in heart, liver and kidney. Many different moieties can be used as targeting ligands. The advan-
tage of peptides, in addition to e.g. antibodies, is their size. Antibodies
6.1. Opsonization and phagocytosis themselves are in the size range of nanoparticles (10 ∗ 15 nm). In his
review, Raha et al. [66] focused on peptides used for cancer targeting
A critical characteristic of both drug delivery systems and drug on nanoparticles in general and gave a comprehensive overview.
targeting systems is the ability to avoid the fast uptake through the Jarzyna et al. [67] reported on NEs with three different mean diam-
MPS out of the blood stream. Opsonization and the consequent eters (30, 60, and 95 nm), which had incorporated oleic acid coated
clearance of the particles by the liver macrophages occurs very quickly, iron oxide (Fe2O3) nanocrystals for magnetic resonance imaging
typically up to 90% of the injected drug is taken up by the liver within (MRI) and a fluorescent dye (Cy5.5 coupled on PEG2000-DSPE) for
5 min [63]. Prior to the uptake of the MPS blood proteins, known as op- near infrared (NIRF) imaging. Soybean oil was used as a lipid core,
sonins (e.g., immunoglobulin γ, complement factors and fibrinogen), Distearoyl-sn-glycero-3-phosphocholine (DSPC) was used as the emul-
attach to the surface of foreign particles (Fig. 4). sifier and, additionally, PEG2000-DSPE and Cy5.5 coupled on PEG2000-
If dysopsonins (e.g. serum albumin) attach to the particle surface, DSPE were added for surface modification. The NE was produced as
the MPS uptake is minimized and blood circulation time is consequently follows: the lipophilic components were dissolved in chloroform. The
prolonged. Opsonization depends on a number of factors: mixture was added, dropwise, to the heated water phase. The resulting
crude emulsion was homogenized by sonication. Within this in vivo
• Size: Large particles are more quickly cleared from the blood stream study, the accumulation of this multimodal imaging NE in subcutaneous
by macrophages than small particles. human EW7 tumors in nude mice could be shown with MRI and NIRF
• Charge: Clearance is highest for positively charged particles, lower for imaging.
negative ones and lowest for non-charged particles, which means Based on this work, Gianella et al. [68] reported a study using this
nanoparticles or nano oil droplets with a low zeta potential [51]. platform to incorporate the cancer treatment drug prednisolone
acetate, which is provided as valerate ester to increase lipophilicity.
Further, the PEG2000-DSPE was coupled to αvβ3-specific RGD-
peptides to enhance targeting angiogenesis of cancer tissue
(Fig. 6). RGD (arginylglycylaspartic acid) is a tripeptide and is composed
of L-arginine, glycine, and L-aspartic acid. This platform technology is
considered as “theranostic” by some. Theranostics are a combination
of therapy and diagnostics using nanotechnology. To achieve this,
imaging and therapeutic ingredients are combined in one nanoparticle
such as oil droplets of NEs. Interestingly, the addition of further material
or functionalities does not result in a change in diameter (all around
50 nm) or polydispersity compared to the blank NE. This small
size facilitates long circulation time in the blood as well as the
Fig. 4. Opsonization and phagocytosis of oil droplets, from [11]. uptake in cancer cells. MRI and NIRF imaging revealed significant
94 K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98

Fig. 5. PEGylation avoids opsonization and can prolong circulation time in the blood, from [64].

accumulation in different colon cancer mouse models. The RGD ligand study, the in vitro studies revealed that positive charges of the oil drop-
provided specific interaction with ανβ3 integrin expressing cell lets led to more rapid opsonization and, consequently, swifter uptake in
types. Mice treated with prednisolone acetate valerate emulsions, the MPS. Even the addition of PEG2000-DSPE for increase of the reten-
RGD-prednisolone acetate valerate emulsion and prednisolone acetate tion period in the blood stream could not negate this effect. The NE
valerate + FeO emulsion, showed a significantly inhibited tumor system with the CREKA-PEG2000-DSPE conjugate on the surface
growth compared to blank NEs, FeO emulsions, saline and free prednis- reached AUClast in blood of 263.8 ± 21.81 min ∗ % injected dose/ml,
olone acetate valerate (all at a dose of 10 mg prednisolone acetate val- whereas the DOTAP NE 20.2 ± 1.86 min ∗ % injected dose/ml and the
erate per kg). 17β-estradiol solution 44.9 ± 1.24 min ∗ % injected dose/ml.
The effect of the density of PEG-chains on the surface of the
abovementioned “theranostic” NE was investigated by Hak et al. [69].
This could be a critical parameter defining the circulation time within 7.2. Antibody mediated targeting
the body. The PEG density was varied from 5 to 50 mol%. At low PEG
density the PEG chains are more flexible, they organize in what is Despite the disadvantage of an antibody's large size, there was some
known as a mushroom configuration. At higher PEG density (more interesting research with NEs. A NE stabilized with poly(ethylene gly-
than 10%, which is commonly used for such applications), the distance col)-modified phosphatidylethanolamine (PEG-PE) as a drug carrier
between the PEG chains is low, which transforms the PEG units to a was described by [72]. The anti-B-cell lymphoma monoclonal antibody
brush configuration and may hinder the targeting ligands RGD peptides LL2 was successfully coupled to the surface, which was tested with a rat
in finding their targets. This steric configuration seems to determine the monoclonal anti-idiotype antibody, WN. Direct cellular ELISA revealed
targeting efficiency and specificity, because, interestingly, the best similar binding of emulsion-LL2 complexes compared to free monoclo-
results are achieved at low PEG surface density (Fig. 7). nal antibody to three types of Burkitt's lymphoma cell lines, Raji, Ramos
For drug targeting, the most widely used peptides are still RGD and Daudi. This indicates that the NE coupled with LL2 has the potential
peptides (integrin-targeted), which were the first tumor-targeting as a useful drug delivery system to B-cell malignancies. The basic NE was
peptide discovered [70]. prepared of triolein, Dipalmitoylphosphatidylcholine (DPPC) and poly-
For targeting atherosclerosis, Deshpande et al. [71] developed a NE sorbate 80 in a weight ratio of 80.2:1:0.4. For a sub-study 2 to 8 mol%
which used CREKA-peptide (Cysteine–Arginine–Glutamic acid– PEG2000-Dipalmitoylphosphatidylethanolamine (DPPE, related to
Lysine–Alanine) as the targeting moiety. CREKA seems to target the fi- DPPC) was also added to the oil phase, which showed no effect on
brin clots present in atherosclerotic lesions selectively. In his previous coupling efficiency. After removal of the solvent the lipid film was
study [57], a NE made of flaxseed-oil containing a high level of omega- re-suspended with phosphate buffered saline (PBS), vortexed and
3-fatty acids, in which 17β-estradiol was successfully incorporated. sonicated. As a linking partner for the antibody conjugation 2 mol%
DOTAP was used on the surface of the emulsion for a positive charge DSPE-PEG-VS (related to DPPC) was included, whereas VS indicate the
to enhance the transfer and uptake in endothelial cells. In this new reactive terminus of the PEG chain, a vinylsulfone group.

Fig. 6. Schematic depiction of a multifunctional nanoemulsion, from [68].


K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98 95

Fig. 7. Nanoemulsions with different PEG2000-DSPE content and mushroom/brush configuration indicated, from [69].

A cationic NE with conjugated monoclonal antibody AMB8LK, which palmitate NE inhibits the tumor growth in mice significantly more than
should target over-expressing H-ferritin in tumors, was reported by a paclitaxel palmitate solution or the base cationic NE.
Goldstein et al. [73]. The NE consists of MCT (5%w/w), poloxamer 188 With Monoclonal antibody 34A the lung can be targeted, because
(1%w/w), glycerol (2.25%w/w), lipoid E80 (1%w/w), stearylamine 34A specifically binds to pulmonary endothelial cell surface in mice.
(0.25%w/w), α-tocopherol (0.01%w/w), and water (up to 100%w/w). It As a drug delivery system a long-circulating NE was studied by Song
is worth noting that in this particular study the coupling of the ligand et al. [75] composed of castor oil, phosphatidylcholine and DSPE-PEG
is made with cross linkers graft over stearylamine molecules on the sur- coupled with the antibody (antibody to lipid ratio 2:1 w/w).
face of the oil droplets, and not over DSPE-PEG moieties as commonly After injection into the mouse tail vein 30% of the injected dose
used. In this in vitro study the NE with the monoclonal antibody was found in the lung 30 min after administration. A kinetic study
AMB8LK achieved a 50% increase in cell uptake compared to cationic showed that after 5 min the highest amount was found in the lung
NE without conjugated monoclonal antibody. which then decreased. However, after 4 h there were still 50% of
In a newer study [74], an NE loaded with paclitaxel palmitate and the maximum bound emulsions present in the lung. The lipid phase
anti-HER2 monoclonal antibody (Herceptin) as a targeting ligand was of the NE was composed of 1.5 mg castor oil, 0.2 mg of phosphatidyl-
assessed. NE composition except the new covalent linked monoclonal choline, 0.4 mg of DSPE-PEG-COOH, which was all dissolved in
antibody was the same as above. The water and oil phases were chloroform. After re-suspension in 0.5 ml MES buffer (pH 5.5) the
mixed separately and heated to 70 °C. After unifying the two phases, suspension was vortexed and homogenized resulting in oil droplets
the emulsion was produced via microfluidization process. The paclitaxel of about 200 nm.

Fig. 8. Schematic diagram of the differential protein adsorption concept, from [51].
96 K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98

7.3. Sugar ligands and albumin as targeting moieties It may be useful to evaluate the differential protein adsorption right
in the beginning of the development of a new NE (see Fig. 8) to get a
Drug targeting can also be realized using sugar recognition mecha- better prediction of the distribution in the body.
nisms. Ishida et al. [76] investigated galactosylated emulsions which
are intended to target asialoglycoprotein receptor on hepatocytes in 9. Summary/conclusion
the liver. The ligand chosen was cholesten-5-yloxy-N- (4-((1-imino-2-
D-thiogalactosylethyl)amino)butyl)formamide (Gal-C4-Chol). The NEs for intravenous use are an interesting system for drug delivery
lipid mixture containing soybean oil, egg phosphatidylcholine and systems and for drug targeting. The following advantages can be
Gal-C4-Chol (weight ratio 70:25:5) was dissolved in chloroform. After summarized so far:
vacuum desiccation, the lipid phase was re-suspended in the water
phase followed by sonification. Radiolabeling of the emulsions was • Ease of manufacturing compared to other nanoparticle systems.
realized by addition of 3H markers to the finished NE. The in vivo tests • Oil droplet inside as drug reservoir, higher drug loading as e.g.
in mice revealed a faster clearance in the blood and 3.2-fold increased liposomes [80].
accumulation in the liver compared to a bare NE without Gal- C4-Chol. Even liposome (much smaller loading capacity) can deliver ten
Mannosylated NE incorporated with cholesten-5-yloxy-N-(4- thousands of anticancer drug molecules directly into target cells
((1-imino-2-d-thiomannosylethyl)amino)alkyl) formamide (Man- [81].
C4-Chol) were tested in in vivo studies with mice [77,78]. • NEs are a perfect delivery system for high lipophilic drugs. If the
The lipid phase consisted of soybean oil (70%w/w), egg phosphati- drugs lipophilicity itself is not high enough, there are often easy
dylcholine (25%w/w), and cholesterol or Man-C4-Chol at various ways to increase the lipophilicity through esterification with long
weight ratios and was dissolved in chloroform. After the removal chain fatty acids.
of chloroform, PBS was added as the water phase. The mixture was • Provides lipophilic drugs in a water based formulation, avoiding
then sonificated. Compared with the bare NE and the mannosylated ingredients with unwanted side effects.
NE with 2% Man, the mannosylated NE with 5% and 7% achieved • Due to the water-based formulation NE products have a low
rapid elimination from the blood circulation and were mainly viscosity.
accumulated in the liver, whereas non-parenchymal cells were • In contrast to other nanocarriers, parenteral o/w nanoemulsions
targeted. This strategy could be used to treat liver related diseases have an accepted regulatory status. Many ingredients are already
like liver fibrosis. used for parenteral nutrition emulsions and drug emulsions.
An optimized Diclofenac containing NE was developed, where albu- • Accumulation in the human body of the drug delivery system NE
min was used as a targeting ligand for inflamed areas [79]. Albumin, due itself or its ingredients is no issue due to the metabolism of these
to its non-toxicity and biodegradability, is the perfect ligand targeting ingredients.
tissues found at areas of inflammation (and tumors). There are hypoal- • Long shelf life compared to other nanoparticle systems.
buminemia albondin receptors overexpressed for receptor-mediated • Lipid emulsions prevent drug adsorption onto plastic infusion sets
endocytosis of albumin. After several formulation trials the stability op- and allow the stabilization of labile drugs against enzymatic
timized NE was composed of (related to 1 ml final NE): 2.5 mg degradation or oxidation. The therapeutic advantages include the
diclofenac acid, 100 mg soybean oil, 12.0 mg egg phosphatidylcholine, elimination of irritation and toxicity associated in contrast to
2.5 mg α-tocopherol acetate, 3.0 mg cholesterol, 22 mg glycerin, other dosage forms [11].
1.0 mg bovine serum albumin and 3.0 mg stearylamine. The NE was pre- • Can be a protection against hydrolysis and oxidation, when the
pared at 70 °C, without solvent, using the sonification method. After- drug is incorporated into the oil core of emulsions, e.g. protection
wards, the albumin was coupled to the NH2-groups of stearyl amine of the lacton ring of Hydroxycamptothecin in NE [82].
on the surface of the NE with the aid of 1-Ethyl-3-(3-dіmethylamino)
propyl carbodiimide during an incubation time of 2 h, which led to an
increase in average diameter from approx. 200 nm to 250 nm–
As a possible disadvantage of NEs, the relatively short circulation
300 nm. In a pharmacokinetic study in rats the therapeutic availability time in the body has to be mentioned here, which is related to the
of the drug at the site of inflammation (granuloma air pouch fluid)
safe (and fast) metabolism of NEs in the human body. With size reduc-
was 2.89 times that of drug solution and better than other NEs contain- tion of the oil droplets and/or PEGylation of the surface the half-life time
ing diclofenac without albumin as the targeting ligand. Furthermore,
can be increased efficiently, but still remains short in comparison with
the drug concentration ratio of inflamed areas to blood serum was ana- other, i.e. solid, nanoparticles. Nevertheless, NEs as a drug delivery
lyzed above one at all times, which indicates a good targeting potential
system or system for drug targeting are seen as a very promising
of the ligand albumin. strategy to overcome limitations of conventional drug systems in re-
spect of biodistribution, avoidance of adverse effects and in developing
8. The concept of differential protein adsorption new possibilities for an efficient treatment of many diseases.

It must be recognized that in many in vivo studies there is an


References
unpredictable change in pharmacokinetics when compared to the
in vitro studies. In the blood the drug, incorporated in a nanoparti- [1] S. Mendes, S.R. Graziani, T.S. Vitório, A.F. Padoveze, R. Hegg, S.P. Bydlowski, et al., Up-
cle or NE, comes into contact with more than thousands of proteins, take by breast carcinoma of a lipidic nanoemulsion after intralesional injection into
a few hundred in higher concentrations. Some of the proteins ab- the patients: a new strategy for neoadjuvant chemotherapy, Gynecol. Oncol. 112
(2009) 400–404, http://dx.doi.org/10.1016/j.ygyno.2008.10.018.
sorb on the surface of the NE (protein absorption pattern). These [2] M. Hashida, S. Kawakami, F. Yamashita, Lipid carrier systems for targeted drug and
proteins can determine the organ distribution, e.g. apolipoprotein gene delivery, Chem. Pharm. Bull. (Tokyo) 53 (2005) 871–880, http://dx.doi.org/10.
C III is known to be responsible for the inhibition of MPS uptake, 1248/cpb.53.871.
[3] O. Suitthimeathegorn, J.A. Turton, H. Mizuuchi, A.T. Florence, Intramuscular absorption
especially in the liver. Introducing PEG chains to the surface of NEs
and biodistribution of dexamethasone from non-aqueous emulsions in the rat, Int. J.
leads to an increased albumin absorption, a known dysopsonin, Pharm. 331 (2007) 204–210, http://dx.doi.org/10.1016/j.ijpharm.2006.11.062.
which leads to less cognition by the MPS [51]. Generally, a change [4] K. Hippalgaonkar, S. Majumdar, V. Kansara, Injectable lipid emulsions—advancements,
in emulsifier or co-surfactant may alter the protein absorption opportunities and challenges, AAPS PharmSciTech 11 (2010) 1526–1540, http://dx.
doi.org/10.1208/s12249-010-9526-5.
pattern and hence, can influence the distribution of the drug in [5] K.B. Sutradhar, M.L. Amin, Nanoemulsions: increasing possibilities in drug delivery,
the body. Eur. J. Nanomedicine. 5 (2013) 97–110, http://dx.doi.org/10.1515/ejnm-2013-0001.
K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98 97

[6] A. Maali, M.T.H. Mosavian, Preparation and application of nanoemulsions in the last fortified parenteral lipid emulsions, Colloids Surf. B: Biointerfaces 103 (2013)
decade (2000–2010), J. Dispers. Sci. Technol. 34 (2013) 92–105, http://dx.doi.org/ 23–30, http://dx.doi.org/10.1016/j.colsurfb.2012.10.003.
10.1080/01932691.2011.648498. [34] A.Y. Alayoubi, J.F. Anderson, S.D. Satyanarayanajois, P.W. Sylvester, S. Nazzal, Con-
[7] S. Tamilvanan, S.R. Senthilkumar, R. Baskar, T.R. Sekharan, Manufacturing tech- current delivery of tocotrienols and simvastatin by lipid nanoemulsions potentiates
niques and excipients used during the formulation of oil-in-water type nanosized their antitumor activity against human mammary adenocarcenoma cells, Eur. J.
emulsions for medical applications, J. Excip. Food Chem. 1 (2010) 11–29. Pharm. Sci. 48 (2012) 385–392, http://dx.doi.org/10.1016/j.ejps.2012.12.011.
[8] S. Tamilvanan, Formulation of multifunctional oil-in-water nanosized emulsions for [35] C.L. Krahn, R.P. Raffin, G.S. Santos, L.B. Queiroga, R.L. Cavalcanti, P. Serpa, et al.,
active and passive targeting of drugs to otherwise inaccessible internal organs of the Isoflurane-loaded nanoemulsion prepared by high-pressure homogenization: in-
human body, Int. J. Pharm. 381 (2009) 62–76, http://dx.doi.org/10.1016/j.ijpharm. vestigation of stability and dose reduction in general anesthesia, J. Biomed.
2009.08.001. Nanotechnol. 8 (2012) 849–858, http://dx.doi.org/10.1166/jbn.2012.1449.
[9] A. Wretlind, Development of fat emulsions, JPEN J. Parenter. Enteral Nutr. 5 (1981) [36] X. Chen, L. Zhang, X. Hu, X. Lin, Z. Yu, X. Tang, Formulation and preparation of a sta-
230–235. ble intravenous Disulfiram-loaded lipid emulsion, Eur. J. Lipid Sci. Technol.
[10] A.G. Floyd, Top ten considerations in the development of parenteral emulsions, (2014)http://dx.doi.org/10.1002/ejlt.201400278 (n/a–n/a).
Pharm. Sci. Technol. Today 4 (1999) 134–143. [37] M. Shawer, P. Greenspan, S. ØIe, D.R. Lu, VLDL-resembling phospholipid-submicron
[11] F. Leal-Calderon, M. Cansell, The design of emulsions and their fate in the body fol- emulsion for cholesterol-based drug targeting, J. Pharm. Sci. 91 (2002) 1405–1413,
lowing enteral and parenteral routes, Soft Matter 8 (2012) 10213, http://dx.doi.org/ http://dx.doi.org/10.1002/jps.10117.
10.1039/c2sm26215k. [38] T.C. Contente, I.F. Kretzer, F.B. Filippin-Monteiro, D.A. Maria, R.C. Maranhão, Associ-
[12] V. Klang, C. Valenta, Lecithin-based nanoemulsions, J. Drug Deliv. Sci. Technol. 21 ation of daunorubicin to a lipid nanoemulsion that binds to low-density lipoprotein
(2011) 55–76. receptors enhances the antitumour action and decreases the toxicity of the drug in
[13] S.M. Đorđević, T.S. Radulović, N.D. Cekić, D.V. Ranđelović, M.M. Savić, D.R. Krajišnik, melanoma-bearing mice, J. Pharm. Pharmacol. 66 (2014) 1698–1709, http://dx.doi.
et al., Experimental design in formulation of diazepam nanoemulsions: org/10.1111/jphp.12296.
physicochemical and pharmacokinetic performances, J. Pharm. Sci. 102 (2013) [39] R.C. Maranhão, E.R. Tavares, A.F. Padoveze, C.J. Valduga, D.G. Rodrigues, M.D. Pereira,
4159–4172, http://dx.doi.org/10.1002/jps.23734. Paclitaxel associated with cholesterol-rich nanoemulsions promotes atherosclerosis
[14] J. Rossi, J.-C. Leroux, Principles in the development of intravenous lipid emulsions, regression in the rabbit, Atherosclerosis 197 (2008) 959–966, http://dx.doi.org/10.
in: K.M. Wasan (Ed.), Role Lipid Excipients Modifying Oral Parenter, Drug Deliv, 1016/j.atherosclerosis.2007.12.051.
John Wiley & Sons, New York 2007, pp. 88–123. [40] M.L.N. Dias, J.P. Carvalho, D.G. Rodrigues, S.R. Graziani, R.C. Maranhão, Pharma-
[15] Chylomicrons, By Xvazquez (Own Work. [GFDL (http://www.gnu.org/copyleft/ cokinetics and tumor uptake of a derivatized form of paclitaxel associated to a
fdl.html), CC-BY-SA-3.0 or FAL], via Wikimedia Commons, https://commons. cholesterol-rich nanoemulsion (LDE) in patients with gynecologic cancers, Can-
wikimedia.org/wiki/File%3AChylomicron.svg2008 (accessed July 23, 2015). cer Chemother. Pharmacol. 59 (2007) 105–111, http://dx.doi.org/10.1007/
[16] S. Tamilvanan, Oil-in-water lipid emulsions: implications for parenteral and ocular s00280-006-0252-3.
delivering systems, Prog. Lipid Res. 43 (2004) 489–533, http://dx.doi.org/10.1016/ [41] B. Madhusudhan, D. Rambhau, S.S. Apte, D. Gopinath, 1-O-alkylglycerol stabilized
j.plipres.2004.09.001. carbamazepine intravenous o/w nanoemulsions for drug targeting in mice, J. Drug
[17] M.N. Ton, C. Chang, Y.A. Carpentier, R.J. Deckelbaum, In vivo and in vitro properties Target. 15 (2007) 154–161, http://dx.doi.org/10.1080/10611860601141150.
of an intravenous lipid emulsion containing only medium chain and fish oil triglyc- [42] C.-J. Wen, T.-C. Yen, S.A. Al-Suwayeh, H.-W. Chang, J.-Y. Fang, In vivo real-time fluo-
erides, Clin. Nutr. 24 (2005) 492–501, http://dx.doi.org/10.1016/j.clnu.2005.03.001. rescence visualization and brain-targeting mechanisms of lipid nanocarriers with
[18] L. Liu, T.K. Hitchens, Q. Ye, Y. Wu, B. Barbe, D.E. Prior, et al., Decreased reticuloendo- different fatty ester:oil ratios, Nanomedicine (London) 6 (2011) 1545–1559,
thelial system clearance and increased blood half-life and immune cell labeling for http://dx.doi.org/10.2217/nnm.11.46.
nano- and micron-sized superparamagnetic iron-oxide particles upon pre- [43] L. Wei, G. Li, Y.-D. Yan, R. Pradhan, J.O. Kim, Q. Quan, Lipid emulsion as a drug deliv-
treatment with intralipid, Biochim. Biophys. Acta 1830 (2013) 3447–3453, http:// ery system for breviscapine: formulation development and optimization, Arch.
dx.doi.org/10.1016/j.bbagen.2013.01.021. Pharm. Res. 35 (2012) 1037–1043, http://dx.doi.org/10.1007/s12272-012-0611-z.
[19] M.A.J. Assink, P.E. Spronk, H.J.M. van Kan, A. Braber, Intravenous lipid emulsion in [44] F. Xiong, H. Wang, Y.-J. Chen, K.-K. Geng, N. Gu, J.-B. Zhu, Characterization,
the treatment of verapamil intoxication, Neth. J. Crit. Care 17 (2013) 18–21. biodistribution and targeting evaluation of breviscapine lipid emulsions following
[20] M.R. Fettiplace, K. Lis, R. Ripper, K. Kowal, A. Pichurko, D. Vitello, et al., Multi-modal intravenous injection in mice, Drug Deliv. 18 (2011) 159–165, http://dx.doi.org/
contributions to detoxification of acute pharmacotoxicity by a triglyceride micro- 10.3109/10717544.2010.528068.
emulsion, J. Control. Release 198 (2014) 62–70, http://dx.doi.org/10.1016/j.jconrel. [45] X. Zhang, X. Sun, J. Li, X. Zhang, T. Gong, Z. Zhang, Lipid nanoemulsions loaded with
2014.11.018. doxorubicin-oleic acid ionic complex: characterization, in vitro and in vivo studies,
[21] K. Shi, Y. Xia, Q. Wang, Y. Wu, X. Dong, C. Chen, et al., The effect of lipid emulsion on Pharmazie 66 (2011) 496–505, http://dx.doi.org/10.1691/ph.2011.0379.
pharmacokinetics and tissue distribution of bupivacaine in rats, Anesth. Analg. 116 [46] K. Ogawara, Y. Yoshizawa, K. Un, T. Araki, T. Kimura, Challenges of Drug Delivery
(2013) 804–809, http://dx.doi.org/10.1213/ANE.0b013e318284123e. Systems That Contribute to Cancer Chemotherapy Nanoparticle-based Passive
[22] S. Ganta, M. Talekar, A. Singh, T.P. Coleman, M.M. Amiji, Nanoemulsions in transla- Drug Targeting to Tumors : Considerations and Implications for Optimization, 36
tional research—opportunities and challenges in targeted cancer therapy, AAPS (2013) 698–702.
PharmSciTech (2014)http://dx.doi.org/10.1208/s12249-014-0088-9. [47] J. Seki, S. Sonoke, A. Saheki, H. Fukui, H. Sasaki, T. Mayumi, A nanometer lipid
[23] T. Takino, K. Konishi, Y. Takakura, M. Hashida, Long circulating emulsion carrier emulsion, lipid nano-sphere (LNS), as a parenteral drug carrier for passive
systems for highly lipophilic drugs, Biol. Pharm. Bull. 17 (1994) 121–125. drug targeting, Int. J. Pharm. 273 (2004) 75–83, http://dx.doi.org/10.1016/j.
[24] S. Ganta, J.W. Paxton, B.C. Baguley, S. Garg, Pharmacokinetics and pharmacodynam- ijpharm.2003.12.022.
ics of chlorambucil delivered in parenteral emulsion, Int. J. Pharm. 360 (2008) [48] K. Qi, M. Al-Haideri, T. Seo, Y.A. Carpentier, R.J. Deckelbaum, Effects of particle size
115–121, http://dx.doi.org/10.1016/j.ijpharm.2008.04.027. on blood clearance and tissue uptake of lipid emulsions with different triglyceride
[25] X. Li, L. Du, C. Wang, Y. Liu, X. Mei, Y. Jin, Highly efficient and lowly toxic docetaxel compositions, J. Parenter. Enter. Nutr. 27 (2003) 58–64.
nanoemulsions for intravenous injection to animals, Pharmazie 66 (2011) 479–483, [49] T. Onodera, I. Kuriyama, T. Andoh, H. Ichikawa, Y. Sakamoto, E. Lee-Hiraiwa, et al.,
http://dx.doi.org/10.1691/ph.2011.1015. Influence of particle size on the in vitro and in vivo anti-inflammatory and anti-
[26] D. Zhao, T. Gong, Y. Fu, Y. Nie, L.-L. He, J. Liu, et al., Lyophilized cheliensisin A submi- allergic activities of a curcumin lipid nanoemulsion, Int. J. Mol. Med. 35 (2015)
cron emulsion for intravenous injection: characterization, in vitro and in vivo anti- 1720–1728, http://dx.doi.org/10.3892/ijmm.2015.2186.
tumor effect, Int. J. Pharm. 357 (2008) 139–147, http://dx.doi.org/10.1016/j. [50] F. Bruxel, S. Cojean, A. Bochot, H. Teixeira, C. Bories, P.-M. Loiseau, et al., Cationic
ijpharm.2008.01.055. nanoemulsion as a delivery system for oligonucleotides targeting malarial topo-
[27] J. Li, S. Nie, X. Yang, C. Wang, S. Cui, W. Pan, Optimization of tocol emulsions for the isomerase II, Int. J. Pharm. 416 (2011) 402–409, http://dx.doi.org/10.1016/j.
intravenous delivery of clarithromycin, Int. J. Pharm. 356 (2008) 282–290, http://dx. ijpharm.2011.01.048.
doi.org/10.1016/j.ijpharm.2007.12.046. [51] C.M. Keck, M. Jansch, R.H. Müller, Protein adsorption patterns and analysis on IV
[28] S. Ramreddy, P. Kandadi, K. Veerabrahma, Formulation and pharmacokinetics of nanoemulsions—the key factor determining the organ distribution, Pharmaceutics
diclofenac lipid nanoemulsions for parenteral application, PDA J. Pharm. Sci. 5 (2012) 36–68, http://dx.doi.org/10.3390/pharmaceutics5010036.
Technol. 66 (2012) 28–37, http://dx.doi.org/10.5731/pdajpst.2012.00735. [52] A. Torcello-Gómez, M. Wulff-Pérez, M.J. Gálvez-Ruiz, A. Martín-Rodríguez, M.
[29] J. Zheng, Y. Tang, M. Sun, Y. Zhao, Q. Li, J. Zhou, et al., Characterization, pharmacoki- Cabrerizo-Vílchez, J. Maldonado-Valderrama, Block copolymers at interfaces: inter-
netics, tissue distribution and antitumor activity of honokiol submicron lipid emul- actions with physiological media, Adv. Colloid Interf. Sci. 206 (2014) 414–427,
sions in tumor-burdened mice, Pharmazie 68 (2013) 41–46, http://dx.doi.org/10. http://dx.doi.org/10.1016/j.cis.2013.10.027.
1691/ph.2013.2062. [53] F.M. Veronese, A. Mero, The impact of PEGylation on biological therapies, BioDrugs
[30] A.M. Dierling, Z. Cui, Targeting primaquine into liver using chylomicron emulsions 22 (2008) 315–329, http://dx.doi.org/10.2165/00063030-200822050-00004.
for potential vivax malaria therapy, Int. J. Pharm. 303 (2005) 143–152, http://dx. [54] D.E. Owens, N.A. Peppas, Opsonization, biodistribution, and pharmacokinetics of
doi.org/10.1016/j.ijpharm.2005.07.015. polymeric nanoparticles, Int. J. Pharm. 307 (2006) 93–102, http://dx.doi.org/10.
[31] J.-H. Jang, C.-K. Kim, H.-G. Choi, J.H. Sung, Preparation and evaluation of 2- 1016/j.ijpharm.2005.10.010.
(allylthio)pyrazine-loaded lipid emulsion with enhanced stability and liver [55] F. Liu, D. Liu, Long-circulating emulsions (oil-in-water) as carriers for lipophilic drugs,
targeting, Drug Dev. Ind. Pharm. 35 (2009) 363–368, http://dx.doi.org/10.1080/ Pharm. Res. 12 (1995) 1060–1064, http://dx.doi.org/10.1023/A:1016274801930.
03639040802363696. [56] A. Alayoubi, S. Alqahtani, A. Kaddoumi, S. Nazzal, Effect of PEG surface conformation
[32] B. Nikonenko, V.M. Reddy, E. Bogatcheva, M. Protopopova, L. Einck, C.A. Nacy, Ther- on anticancer activity and blood circulation of nanoemulsions loaded with
apeutic efficacy of SQ641-NE against Mycobacterium tuberculosis, Antimicrob. tocotrienol-rich fraction of palm oil, AAPS J. 15 (2013) 1168–1179, http://dx.doi.
Agents Chemother. 58 (2014) 587–589, http://dx.doi.org/10.1128/AAC.01254-13. org/10.1208/s12248-013-9525-z.
[33] A. Alayoubi, S. Kanthala, S.D. Satyanarayanajois, J.F. Anderson, P.W. Sylvester, S. [57] D. Deshpande, D.R. Janero, M. Amiji, Engineering of an ω-3 polyunsaturated fatty
Nazzal, Stability and in vitro antiproliferative activity of bioactive “Vitamin E” acid-containing nanoemulsion system for combination C6-ceramide and
98 K. Hörmann, A. Zimmer / Journal of Controlled Release 223 (2016) 85–98

17β-estradiol delivery and bioactivity in human vascular endothelial and smooth [70] X. Zhang, H. Eden, X. Chen, Peptides in cancer nanomedicine: drug carriers, targeting
muscle cells, Nanomedicine 9 (2013) 885–894, http://dx.doi.org/10.1016/j.nano. ligands and protease substrates, J. Control. Release 159 (2012) 2–13, http://dx.doi.
2013.02.007. org/10.1016/j.jconrel.2011.10.023.Peptides.
[58] S. Ganta, P. Sharma, J.W. Paxton, B.C. Baguley, S. Garg, Pharmacokinetics and phar- [71] D. Deshpande, S. Kethireddy, F. Gattacceca, M. Amiji, Comparative pharmacokinetics
macodynamics of chlorambucil delivered in long-circulating nanoemulsion, J. Drug and tissue distribution analysis of systemically administered 17-β-estradiol and its
Target. 18 (2010) 125–133, http://dx.doi.org/10.3109/10611860903244199. metabolites in vivo delivered using a cationic nanoemulsion or a peptide-modified
[59] B.B. Lundberg, V. Risovic, M. Ramaswamy, K.M. Wasan, A lipophilic paclitaxel nanoemulsion system for targeting atherosclerosis, J. Control. Release 180 (2014)
derivative incorporated in a lipid emulsion for parenteral administration, J. Control. 117–124, http://dx.doi.org/10.1016/j.jconrel.2014.02.009.
Release 86 (2003) 93–100, http://dx.doi.org/10.1016/S0168-3659(02)00323-1. [72] B.B. Lundberg, G. Griffiths, H.J. Hansen, Conjugation of an anti-B-cell lymphoma
[60] Y. Yoshizawa, Y. Kono, K. Ogawara, T. Kimura, K. Higaki, PEG liposomalization of monoclonal antibody, LL2, to long-circulating drug-carrier lipid emulsions, J.
paclitaxel improved its in vivo disposition and anti-tumor efficacy, Int. J. Pharm. Pharm. Pharmacol. 51 (1999) 1099–1105, http://dx.doi.org/10.1211/
412 (2011) 132–141, http://dx.doi.org/10.1016/j.ijpharm.2011.04.008. 0022357991776787.
[61] X. Jing, L. Deng, B. Gao, L. Xiao, Y. Zhang, X. Ke, et al., A novel polyethylene glycol [73] D. Goldstein, T. Nassar, G. Lambert, J. Kadouche, S. Benita, The design and evaluation
mediated lipid nanoemulsion as drug delivery carrier for paclitaxel, Nanomedicine of a novel targeted drug delivery system using cationic emulsion-antibody conju-
10 (2014) 371–380, http://dx.doi.org/10.1016/j.nano.2013.07.018. gates, J. Control. Release 108 (2005) 418–432, http://dx.doi.org/10.1016/j.jconrel.
[62] H. Zhao, H. Lu, T. Gong, Z. Zhang, Nanoemulsion loaded with lycobetaine–oleic acid 2005.08.021.
ionic complex: physicochemical characteristics, in vitro, in vivo evaluation, and [74] D. Goldstein, O. Gofrit, A. Nyska, S. Benita, Anti-HER2 cationic immunoemulsion as a
antitumor activity, Int. J. Nanomedicine 8 (2013) 1959–1973, http://dx.doi.org/10. potential targeted drug delivery system for the treatment of prostate cancer, Cancer
2147/IJN.S43892. Res. 67 (2007) 269–275, http://dx.doi.org/10.1158/0008-5472.CAN-06-2731.
[63] R.H. Müller, Colloidal Carriers for Controlled Drug Delivery and Targeting: Modifica- [75] Y.K. Song, D. Liu, K. Maruyama, T. Takizawa, Antibody mediated lung targeting of
tion, Characterization and In Vivo Distribution, Taylor & Francis, 1991. long-circulating emulsions, PDA J. Pharm. Sci. Technol. 50 (1996) 372–377.
[64] J.M. Morachis, E.A. Mahmoud, A. Almutairi, Physical and chemical strategies for [76] E. Ishida, C. Managit, S. Kawakami, M. Nishikawa, F. Yamashita, M. Hashida,
therapeutic delivery by using polymeric nanoparticles, Pharmacol. Rev. 64 (2012) Biodistribution characteristics of galactosylated emulsions and incorporated
505–519, http://dx.doi.org/10.1124/pr.111.005363. probucol for hepatocyte-selective targeting of lipophilic drugs in mice, Pharm.
[65] H. Zhou, J. Zhang, Q. Jin, G. Liu, Y. Long, M. Duan, et al., Targeting of coenzyme Q10 Res. 21 (2004) 932–939, http://dx.doi.org/10.1023/B:PHAM.0000029280.39882.ff.
via d-alpha-tocopheryl polyethylene glycol 1000 succinate-based nanoemulsion to [77] W. Yeeprae, S. Kawakami, Y. Higuchi, F. Yamashita, M. Hashida, Biodistribution char-
the heart, Mater. Lett. 109 (2013) 20–22, http://dx.doi.org/10.1016/j.matlet.2013. acteristics of mannosylated and fucosylated O/W emulsions in mice, J. Drug Target.
07.057. 13 (2005) 479–487, http://dx.doi.org/10.1080/10611860500293367.
[66] S. Raha, T. Paunesku, G. Woloschak, Peptide-mediated cancer targeting of [78] W. Yeeprae, S. Kawakami, F. Yamashita, M. Hashida, Effect of mannose density on
nanoconjugates, Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 3 (2011) mannose receptor-mediated cellular uptake of mannosylated O/W emulsions by
269–281, http://dx.doi.org/10.1002/wnan.121. macrophages, J. Control. Release 114 (2006) 193–201, http://dx.doi.org/10.1016/j.
[67] P.A. Jarzyna, T. Skajaa, A. Gianella, D.P. Cormode, D.D. Samber, S.D. Dickson, et al., jconrel.2006.04.010.
Iron oxide core oil-in-water emulsions as a multifunctional nanoparticle platform [79] P. Kandadi, M.A. Syed, S. Goparaboina, K. Veerabrahma, Albumin coupled lipid
for tumor targeting and imaging, Biomaterials 30 (2009) 6947–6954, http://dx. nanoemulsions of diclofenac for targeted delivery to inflammation, Nanomedicine
doi.org/10.1016/j.biomaterials.2009.09.004. 8 (2012) 1162–1171, http://dx.doi.org/10.1016/j.nano.2011.12.006.
[68] A. Gianella, P.A. Jarzyna, V. Mani, S. Ramachandran, C. Calcagno, J. Tang, et al., [80] J. D'Arrigo, Stable Nanoemulsions: Self-assembly in Nature and Nanomedicine,
Multifunctional nanoemulsion platform for imaging guided therapy evaluated in Elsevier, 2011http://dx.doi.org/10.1016/B978-0-444-53798-0.00022-5.
experimental cancer, ACS Nano 5 (2011) 4422–4433, http://dx.doi.org/10.1021/ [81] H.-C. Wu, D.-K. Chang, Peptide-mediated liposomal drug delivery system targeting
nn103336a. tumor blood vessels in anticancer therapy, J. Oncol. 2010 (2010) 723798, http://
[69] S. Hak, E. Helgesen, H.H. Hektoen, E.M. Huuse, P.A. Jarzyna, W.J.M. Mulder, et al., The dx.doi.org/10.1155/2010/723798.
effect of nanoparticle polyethylene glycol surface density on ligand-directed tumor [82] Y. Zhao, J. Gao, X. Sun, H. Chen, L. Wu, W. Liang, Enhanced nuclear delivery and
targeting studied in vivo by dual modality imaging, ACS Nano 6 (2012) 5648–5658, cytotoxic activity of hydro-xycamptothecin using o/w emulsions, J. Pharm. Pharm.
http://dx.doi.org/10.1021/nn301630n. Sci. 10 (2007) 61–70.

You might also like