You are on page 1of 10

Available online at www.sciencedirect.

com

Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273 282 www.nanomedjournal.com

Review Article: Pharmacology

Emerging nanopharmaceuticals
Willie E. Bawarski, Pharm D, Elena Chidlowsky, Pharm D, Dhruba J. Bharali, PhD, Shaker A. Mousa, PhD, MBA
The Pharmaceutical Research Institute at Albany College of Pharmacy, Rensselaer, New York, USA

Abstract

A budding interest in nanopharmaceuticals has generated a number of advancements throughout recent years with a focus on engineering novel applications. Nanotechnology also offers the ability to detect diseases at much earlier stages, such as finding hidden or overt metastatic colonies often seen in patients diagnosed with breast, lung, colon, prostate, and ovarian cancer. Diagnostic applications could build upon conventional procedures using nanoparticles, such as colloidal gold, iron oxide crystals, and quantum dots. Additionally, diseases may be managed by multifunctional agents encompassing both imaging and therapeutic capabilities, thus allowing simultaneous monitoring and treatment. A detailed evaluation of each formulation is essential to expand our current nanopharmaceutical repertoire. However, the safety and long-term effects of nanoformulations must not be overlooked. This review will provide a brief discussion of the major nanopharmaceutical formulations as well as the impact of nanotechnology into the future. 2008 Elsevier Inc. All rights reserved.
Nanoparticles; Micelles; Dendrimers; Liposomes; Quantum dots

Key words:

Novel approaches to drug delivery and formulation using nanotechnology are revolutionizing the future of medicine. A practical definition of nanotechnology, that is unconstrained by an arbitrary size limitation is proposed by Bawa 1 as the design, characterization, production, and application of structures, devices, and systems by controlled manipulation of size and shape at the nanometer scale (atomic, molecular, and macromolecular scale) that produces structures, devices, and systems with at least one novel/superior characteristic or property. Nanomedicine, the medical application of nanotechnology, promises an endless range of applications from biomedical imaging to drug delivery and therapeutics. Nanomedicine results from the manipulation of atoms and molecules that can range in size from 1 to 100 nm. In perspective, a nanometer is one-billionth of a meter, which is analogous to comparing the size of a marble to the size of the Earth. At this size, quantum physics begins

Received 2 January 2008; accepted 3 June 2008. No conflict of interest was reported by the authors of this article. Corresponding author. E-mail address: mousas@acp.edu (S.A. Mousa). 1549-9634/$ see front matter 2008 Elsevier Inc. All rights reserved. doi:10.1016/j.nano.2008.06.002

to take over and particles begin to show entirely different physicochemical properties. Over recent years, advancements in drug delivery have facilitated the targeting of specific tissues. With the advent of nanotechnology, these targeted tissues are now becoming specific organelles within individualized cells. Nanomedicine has blossomed into a billion-dollar industry because of these compounds inherent ability to overcome solubility and stability issues, localize drug delivery, as well as to diagnose via in vivo imaging. Coupled with genomic tailoring, nanomedicine may soon spawn the much-anticipated individualized medicine. Upcoming innovations in nanomedicine may even generate multifunctional entities capable of simultaneously diagnosing, delivering therapeutic agents, and monitoring treatment. Several particle types and structures have been discovered. Noteworthy structures include polymeric micelles, dendrimers, quantum dots (QDs), and solid nanoparticles. Another Food and Drug Administration (FDA)approved nanomedicine, Abraxane (Abraxis, Los Angeles, California), an albumin-bound nanoparticle formulation of paclitaxel, is a prime example of the budding success offered by conjugated polymers. Abraxane is used for metastatic breast

Please cite this article as: W.E. Bawarski, E. Chidlowsky, D.J. Bharali, S.A. Mousa, Emerging nanopharmaceuticals, Nanomedicine: NBM 2008;4:273-82, doi:10.1016/j.nano.2008.06.002

274

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282

cancer. Abraxane for injectable suspension evades the hypersensitivity reaction associated with Cremophor EL (BASF, Florham Park, New Jersey), the solvent in traditional paclitaxel. This nanoparticle has a size of around 100 nm and offers the ability to solubilize insoluble or poorly soluble drugs, avoiding the need for the toxic organic solvent. Abraxane is generally injected into a vein (intravenous infusion) over 30 minutes. The notable side effects of Abraxane include hair loss, infection due to low white blood cell count, fatigue and weakness, low red blood cell count, mouth or lip sores, joint and muscle pain, stomach upset and diarrhea, and cardiovascular effects. Although these structures may promise endless opportunities, their safety should not be ignored. The reactivity of these tiny particles may be due to their large surface area in comparison to their overall mass. Semiconductor metals, such as colloidal gold and iron oxide crystals, are commonly used and have demonstrated toxicity. Additionally, increased pulmonary toxicity was noted with carbon nanotubes when compared to that of the carbon black and carbonyl iron particles seen in mice and rats. 2,3 These tiny particles easily permeate the skin and blood-brain barrier, leading to several potential toxicities. Research should be carried out to fully investigate any toxicity issues associated with these structures. Nanopharmaceutical templates Nanotechnology provides endless opportunities across a wide array of industries. Some of these include titanium dioxide (TiO2) nanoparticles found in sunscreen and cosmetics, silver (Ag) nanoparticles in clothing and disinfectants, and cerium oxide (CeO2) nanoparticles as a fuel catalyst. The National Nanotechnology Initiative (NNI) defines nanotechnology as the understanding and control of matter at dimensions of roughly 1 to 100 nanometers, where unique phenomena enable novel applications, 4 allowing fabrication of devices on the nanoscale. Nanomedicine is the medical application of a broad range of materials and devices on the nanoscale so as to assess, preserve, and restore health and well-being. It takes advantage of nanoscale formulations to optimize drug delivery and to facilitate noninvasive imaging. In addition to mainstream nanomaterials like fullerenes and nanoparticles, nanomedicine uses biomolecules and nanoelectronic biosensors to create therapeutic and diagnostic formulations. Despite sharing one common name, nanomedicine signifies countless approaches to diagnosis and treatment; many different nanoscale drug delivery systems can be created from countless combinations of nanomaterials and molecules, and these carriers can be customized for working in specific tissues or individual patients via attachment of surface ligand molecules. We will review some of the commonly used nanopharmaceutical formulations. However, new advancements are constantly being made that allow multifunctional nanoformulations to deliver drugs while simultaneously collecting diagnostic information.

Although mainstream nanotechnology explores particles between 1 and 100 nm in diameter, the size of the individual particles tested for drug delivery of therapeutic and imaging agents may range from 2 to 1000 nm. 4 However, it has been confirmed that particles larger than 200 nm can activate the human complement system and be cleared from the blood by Kupffer cells. Additionally, splenic filtration captures particles that exceed slit size (200250 nm), and liver filtration (via fenestrae in the sinus endothelium) captures particles greater than 150 nm. Furthermore, tumor capillaries rarely exceed 300 nm in diameter. 5 For the aforementioned reasons, current research on nanopharmaceutical formulations focuses on particles less than 200 nm. The outer surfaces of most recently developed nanoformulations are modified through incorporation of various ligands into a membrane (i.e., specific antigens, antibodies, and receptor ligands) to facilitate targeted delivery to specific tissues. 6 Although some applications of nanotechnology in pharmacology may be questioned, there is an area where the application of nanotechnology application is promising. Specifically, nanoformulations may eliminate the need for conditional administration of drugs, thereby promoting patient compliance and increasing therapeutic effects. Liposomes: progressing to nanopharmaceuticals Liposomes are spherical vesicles composed of amphiphilic phospholipids and cholesterol, which self-associate into bilayers to encapsulate an aqueous interior. 7,8 The amphiphilic phospholipid molecules form a closed bilayer sphere in an attempt to shield their hydrophobic groups from the aqueous environment, while still maintaining contact with the aqueous phase via the hydrophilic head group. Drugs with widely varying lipophilicities can be encapsulated in liposomes, in the phospholipid bilayer, in the entrapped aqueous volume, or at the bilayer interface. Although liposomes vary greatly in size, most are 400 nm or less. Depending upon their size and number of bilayers, liposomes can be classified into three categories: multilamellar vesicles, large unilamellar vesicles, and small unilamellar vesicles. Liposomes can be classified in terms of composition and mechanism of intracellular delivery into five types: conventional liposomes, pH-sensitive liposomes, cationic liposomes, immunoliposomes, and long-circulating liposomes. Although liposome technology was discovered over 40 years ago, liposome-based drug formulations have not entered the market in great number. Some of the major problems limiting the manufacture and development of liposomes are their stability, poor batch-to-batch reproducibility, difficulties in sterilization, and low drug loading. One exceptional success story is the discovery of Doxil (ALZA, Mountain View, California; later, Sequus Pharmaceuticals, Menlo Park, California), a long-acting PEGylated liposomal formulation of doxorubicin. Doxil, known for its significant improvements over doxorubicin, has bridged the gap

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282 Table 1 Some selected nanomedicine products currently on the market Product Doxil Amphocil Ambisome DaunoXome Abelcet Rapamune Emend TriCor Megace ES Company Sequus Pharmaceutical Sequus Pharmaceutical NeXstar Pharmaceutical NeXstar Pharmaceutical (Boulder, Colorado) The Liposome Company (Princeton, New Jersey) Wyeth/Elan (Madison, New Jersey) Merck/Elan (Whitehouse Station, New Jersey) Abbott (Abbott Park, Illinois) PAR Pharmaceutical (WoodCliff Lake, New Jersey) American Biosciences (Blauvelt, New York) BioSante (Lincolnshire, Illinois) Drug Doxorubicin Amphotericin B Amphotericin B Daunorubicin citrate Amphotericin B Sirolimus Aprepitant, MK869 Fenofibrate Megaestrol acetate Formulation Liposome Lipocomplex Liposome Liposome Lipid complex Nanocrystal particles Nanocrystal particles Nanocrystal particles Nanocrystal particles Route of administration Intravenous injection IV infusion IV infusion IV IV infusion Oral Oral oOral Oral Application Kaposi sarcoma in AIDS Serious fungal infections Serious fungal infections Kaposi sarcoma in AIDS Serious fungal infections Immunosuppressant in kidney transplant patients For chemotherapy patient to delayed nausea and vomiting Primary hypercholesterolemiamixed lipidemia, hypertriglyceridemia

275

Treatment of anorexia, cachexia, or an unexplained significant weight loss in patients with a diagnosis of AIDS Metastatic breast cancer Treatment of moderate-to-severe vasomotor symptoms (hot flashes) in menopausal women

Abraxane Elestrin

Paclitaxel Estradiol

Albumin-bound nanoparticles Calcium phosphatebased nanoparticles

IV injection Transdermal

between pharmaceuticals and nanopharmaceuticals. Some of the liposome-based formulations are listed in Table 1. Polymeric micelles: enhancing solubility Micelles are nanosized, spherical colloidal particles with a hydrophobic interior (core) and a hydrophilic exterior (shell). Their main utility is in the preparation of pharmaceutical formulations, notably agents that are regularly soluble in water. 9 Drugs or contrast agents may be entrapped within the hydrophobic core or linked covalently to the surface of micelles. Their individual particle size is less than 50 nm in diameter, which provides obvious benefits over liposomes. Polymeric micelles may circulate for prolonged periods in the blood, evading host defenses. With their property of continued stability in the blood, polymeric micelles can be used to gradually release drugs and facilitate in vivo imaging. To support prolonged systemic circulation, shells of polymeric micelles are designed to be thermodynamically stable and biocompatible. 10,11 Many existing solvents for poorly watersoluble pharmaceuticals, like Cremophor EL (BASF) or ethanol, can be toxic, which limits therapeutic doses and restricts treatment options. Polymeric micelles provide a safer alternative for parenteral administration of poorly water-soluble drugs like amphotericin B, propofol, paclitaxel, and photosensitizers. 12 For the formation of micelles, amphiphilic molecules must have both hydrophobic and hydrophilic segments, where the hydrophilic fragments form the micelle shell and the hydrophobic fragment forms the

core. Thus, in aqueous media, the core of the micelles can solubilize water-insoluble drugs; the surface can adsorb polar molecules, whereas drugs with intermediate polarity can be distributed along with the surfactant molecules in intermediate positions. The mechanism of solubilization and utilization of micelles has been extensively studied by various researchers. A schematic diagram of the formation of micelles from an amphiphilic molecule and the loading of hydrophobic drugs are shown in Figure 1. Similar to liposomes, polymeric micelles can be modified using piloting ligand molecules for targeted delivery to specific cells (i.e., cancer cells). pH-sensitive drug-binding linkers can be added for controlled drug release. 13,14 For that same purpose, micelles can also be formed from stimuliresponsive amphiphilic block co-polymers. Multifunctional polymeric micelles can be designed to facilitate simultaneous drug delivery and imaging. 8 A micelle formulation of an approved therapeutic agent was evaluated in a multicenter, randomized, double-blind, placebo-controlled trial of topical nanoparticle estradiol emulsion (MNPEE; Estrasorb; Novavax, Inc., Malvern, Pennsylvania). The formulation was used in postmenopausal women with moderate to severe vasomotor symptoms (hot flashes). The subjects were given 8.6 mg of estradiol daily, and the treatment lasted 12 weeks. The treatment was both efficacious and well tolerated. MNPEE provided rapid symptom relief and significantly reduced hot flush frequency and severity as compared with placebo from week 4 to the end of the trial (P b 0.001 for both parameters). 15 A list of different polymers/copolymers used

276

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282

Figure 1. Micelles formation. A, Formation of micelles in aqueous media. B, Formation of micelles in aqueous media incorporating drugs.

to prepare micelles loaded with various pharmaceuticals is shown in Table 2. Dendrimers: utilizing multivalent moieties Dendrimers are a unique class of polymeric macromolecules synthesized via divergent or convergent synthesis by a series of controlled polymerization reactions. Characteristically, the structure of these polymers is repeated branching around the central core that results in a nearly-perfect threedimensional geometrical pattern. At higher generations (greater than five) dendrimers resemble spheres with countless cavities within their branches to hold therapeutic and diagnostic agents. In theory, it is possible to synthesize amphiphilic dendrimers with a hydrophobic core inside hydrophilic branching. Dendrimers used in drug delivery and imaging are usually 10 to 100 nm in diameter with multiple functional groups on their surface, rendering them ideal carriers for targeted drug delivery. 16 The structure and function of dendrimers has been well studied. Contemporary dendrimers can be highly specialized, encapsulating functional molecules (i.e., therapeutic or diagnostic agents) inside their core. 17 A polyamidoamine dendrimer that can be synthesized by the repetitive addition of branching units to an amine core (ammonia or ethylene diamine) is an example of such an application. Polyamidoamine cores can function as drug reservoirs and have been studied as vehicles for delivery of drugs, 18 genetic material, 19 and imaging probes. 20,21 Other pharmaceutical applications of dendrimers include nonsteroidal anti-inflam-

matory formulations, antimicrobial and antiviral drugs, anticancer agents, 22 pro-drugs, and screening agents for high-throughput drug discovery. 23 Dendrimers may be toxic because of their ability to disrupt cell membranes as a result of a positive charge on their surface. 24 Dendrimers are useful antiviral agents in that they allow the presentation of multiple ligands on a single molecule as a result of a high number of functional groups. Upon modification with carbohydrate residues, these multivalent molecules can inhibit viral binding. When derivatized with peptides or anionic groups, they can inhibit infection. Because dendrimer synthesis is controlled, they can be made to order to fit target binding sites of specific viruses. 25 The first investigational new drug application for a dendrimer-based drug was submitted to the US FDA in June 2003, and the first clinical trial under this investigational new drug application was completed in 2004. The drug, named VivaGel (SPL7013 Gel) is a vaginal microbicide designed to prevent the transmission of sexually transmitted infections, including the human immunodeficiency virus (HIV) and genital herpes. VivaGel is the first example of a dendrimerbased product and was formulated by Starpharma (Melbourne, Australia). SPL7013 is the active ingredient in VivaGel and is a lysine-based dendrimer with naphthalene disulfonic acid surface groups. Dendrimers such as SPL7013 are polymers that contain a central core, interior branches, and terminal surface groups adapted to specific targets. A polyanionic outer surface of SPL7013 is responsible for multiple target interactions. It was demonstrated that the active surface groups bind to gp120 proteins on HIV's

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282 Table 2 Some block co-polymers used to prepare micelles loaded with various pharmaceuticals Pharmaceuticals incorporated Doxorubicin Polymers/block polymer used for micelles formation Pluronics Poly(aspartic acid)-b-PEG Poly(aspartic acid)-b-PEG Poly(hydroxyl-ethylene oxide) Poly(benzyl-L-aspartate)-b-PEG Poly(2-ethyl-2-oxazoline)-b-poly(L-lactide) Poly(N-isopropylacrylamide)-b-poly (alkyl(meth)acrylate) (pH-sensitive) Poly(L-histidine)-b-PEG (folate-targeted) Poly(L-lactic acid)-b-PEG (folate-targeted) Carboplatin Cisplatin Pluronics Pluronics Polycaprolactone-b-methoxy-PEG Poly(aspartic acid)-b-PEG Poly(glutamic acid)-b-PEG Paclitaxel Poly(delta-valerolactone)-b-methoxy-PEG Polycaprolactone-b-methoxy-PEG Poly(D,L-lactide)-b-methoxy-PEG Poly(2-ethyl-2-oxazoline)-b-poly (-caprolactone) PEG-lipid PEG-PE/egg phosphatidylcholine (mixed micelles) Indomethacin Polycaprolactone-b-methoxy-PEG Poly(benzyl-L-aspartate)-b-PEG Poly(benzyl-L-aspartate)-b-PEG Poly(N-vinyl-2-pyrrolidone)-b-poly (d,L-lactide) Cyclosporine A Amphotericin B Polycaprolactone-b-PEG Poly(benzyl-L-aspartate)-b-PEG PEG-lipid Adriamycin Poly(aspartic acid)-b-PEG Poly(hydroxyl-ethylene oxide) Ibuprofen Ketorolac Camptothecin Chitosan grafted with palmitoyl Poly(N-isopropylacrylamide)-poly (vinylpyrrolidone)-poly(acrylic acid) Poly(aspartic acid)-b-PEG Poly(benzyl-L-aspartate)-b-PEG PEG-lipid PEG-PE/egg phosphatidylcholine (mixed micelles) Phtalocyanine Poly(N-isopropylacrylamide)-b-poly (d,L-lactide) (thermosensitive) Poly(N-isopropylacrylamide)-based (continued on next page) Testosterone Iodine-125 Indium-111 (via DTPA-PE, diagnostic) Gd (via DTPA-PE, diagnostic) Table 2 (continued) Pharmaceuticals incorporated Polymers/block polymer used for micelles formation micelles (pH-sensitive) Poly(N-isopropylacrylamide)-b-poly (alkyl(meth)acrylate) (pH-sensitive) PEG-lipid Poly(D,l-lactide)-b-methoxy-PEG Poly(L-lysine)-b-PEG PEG-lipid PEG-lipid

277

DTPA-PE = Diethylenetriaminepentaacetic acid phosphatidylethanolamine.

surface, preventing CD4 receptor binding by healthy cells, thus preventing transmission of HIV to healthy cells. Because of its size and polyvalent nature, a dendrimer can activate many receptors simultaneously as compared with a small molecule, which can interact with only a single receptor; therefore, these polyvalence dendrimers can lead to new or enhanced biological effects (Figure 2). Quantum dots: enhancing in vivo imaging QDs are colloidal semiconductor nanocrystals ranging from 2 to 10 nm in diameter. QDs can be synthesized from various types of semiconductor materials via colloidal synthesis or electrochemistry. The most commonly used QDs are cadmium selenide (CdSe), cadmium telluride (CdTe), indium phosphide (InP), and indium arsenide (InAs). In bioimaging these particles serve as contrast agents, providing much greater resolution than existing fluorescent dyes. These particles can absorb white light and re-emit it within nanoseconds with different bulk band gap energies corresponding to different combinations of particles. Thus, different QDs can emit different fluorescent light (in wavelength from 400 to 1350 nm). For example, 2-nm QDs will emit green light, whereas 5-nm particles will emit red light. 26,27 QD libraries can be assembled to include particles of various size and composition to support derivation of multicolored images for bimolecular studies, 26 gene expression, 28 cell labeling and tracking, 29 Fluorescence resonance energy transfer (FRET), in vivo imaging, and related applications. 30 Similar to other nanoparticles, QDs can be modified via conjugation of various surface molecules for targeted delivery. 31,32 QDs also provide enough surface area to attach therapeutic agents for simultaneous drug delivery and in vivo imaging, 26,33 as well as for tissue engineering. 34 In vivo cancer targeting and imaging in living animals by QDs was first demonstrated by Gao et al., 35 wherein both subcutaneous injection of QDtagged cancer cells (prostate cancer) and systemic injection of multifunctional QD probes were used to achieve sensitive and multicolor fluorescence imaging of cancer cells. In a

278

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282

Figure 2. Dendrimers work more effectively than a small molecule. A, Small molecules have the ability to interact with only one receptor in a biological system. B, Dendrimers can interact with multiple receptors simultaneously and thus have the potential to increase the biological effect.

recent a study, Bagalkot et al. 33 has used QD-apatmerdoxorubicin (Dox) conjugate for targeted cancer therapy, imaging, and sensing. It was shown that this multifunctional nanoparticle system can deliver Dox to the targeted prostate cancer cells and sense the delivery of Dox by activating the fluorescence of QDs, which concurrently images the cancer cells. Under some conditions QDs can become cytotoxic. 36 It was discovered that CdSe particles may leak cytotoxic cadmium ions after long-term exposure to ultraviolet light, whereas CdTe particles produce reactive oxygen species as a result of the loss of their protective coating after longterm circulation. In both cases, cytotoxicity and cell death were recorded. 37-40 Research is continuing to find biocompatible and stable QD coatings. One recently proposed option was to cover CdSe-ZnS QDs with silica spheres. Resulting CdSe-ZnSSiO2 particles demonstrated consistent fluorescence intensity and predictable behavior. 41 Solid nanoparticles: constructing versatile drug carriers Most commonly used solid nanoparticles (SNPs) are spherical objects made of biodegradable materials, such as proteins (i.e., albumin or collagen), fats, or polymers. 42 First SNPs were constructed to deliver drugs. Ranging in size from 10 to 1000 nm, current SNPs can multitask, providing simultaneous imaging and drug delivery. Analogous to other nanoparticles, SNPs can be modified with surface molecules for guided drug delivery. A major advantage of this formulation is that SNPs can be prepared to provide controlled drug release. 43 SNPs are among the most common current nanoformulations.

In August 2000, the US FDA approved the first nanoparticle-mediated medicine known as Rapamune (sirolimus), an immunosuppressant to prevent organ transplant rejection, developed by Elan (King of Prussia, Pennsylvania). 44 Rapamune was previously available only as an oral solution that requires refrigeration, and must be mixed with water or orange juice before administration. A new tablet developed by using NanoCrystal technology (Elan) is more convenient in terms of storage and administrations. NanoCrystal particles are typically in the size range of 80 to 400 nm and are made by wet-milling a drug substance, water, and a stabilizer. For compounds with negligible water solubility, Elan's proprietary NanoCrystal technology can facilitate formulation development, as well as improving compound activity and final product characteristics. The NanoCrystal technology can be incorporated into all dosage forms, both parenteral and solid, liquid, fast-melt, pulsed release, and controlled-release oral dosage forms. Several years ago an albumin-stabilized nanoparticle formulation of paclitaxel, ABI-007, was designed to overcome insolubility of this agent. 45 The formulation was designed to eliminate the use of its toxic solvent, Cremophor EL (BASF), which allowed increasing administered doses, thus increasing overall drug efficacy. ABI-007, unlike conventional paclitaxel, did not require pre-medication, and showed acceptable toxicities and considerable antitumor activity (80.9% of patients had a complete or partial response). The recommended phase II ABI-007 dose was 230 mg/m 2 every 3 weeks. A phase III trial of that same formulation for metastatic breast cancer was completed, and fast-track status for application was granted by the US FDA in January 2003. In another study the pharmacokinetics of Cremophor-free ABI-007 was

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282

279

compared to traditional Cremophor-ethanol paclitaxel formulation (Taxol; Bristol-Meyers Squibb, New York, New York) in animals and humans. 46 Both formulations were administered intravenously. The study found fecal excretion to be the main elimination pathway for both formulations. Scientists also reported higher paclitaxel clearance and volume of distribution for ABI-007 as compared with paclitaxel in humans (21.13 versus 14.76 L/hr/m 2 (P = .048) and 663.8 versus 433.4 L/hr/m 2 (P = .040), respectively). Some of the selective SNP formulations available in the market are shown in Table 1. A SNP formulation of another cancer agent, Dox, was also evaluated. 47 Dox was incorporated into biodegradable acrylate nanoparticles of polyisohexylcyanoacrylate. The nanoformulation of Dox reported an increase in cytotoxicity and a reduction in cardiotoxicity in preclinical studies. This formulation was tested in human subjects with refractory solid tumors at six dosing regimens (15, 30, 45, 60, 75, and 90 mg/m 2). The drug was infused intravenously over 60 minutes in 250 mL of 5% dextrose in water. No cardiotoxicity was reported, and dose-limiting toxicity was neutropenia; hematologic toxicity appeared at 75 and 90 mg/m 2. Cautiously, the recommended phase II dose was 75 mg/m 2. Solid lipid nanoparticles (SLNs) (made from solid lipids) has attracted significant interest by various researchers since the mid 1990s as an innovative drug delivery carrier system, because of their physical stability, protection of incorporated labile drugs from degradation, controlled release, and excellent tolerability. These versatile SLN formulations can be administered by various routes like parenteral, oral, dermal, ocular, pulmonary, and rectal. Depending on the drugs to be incorporated and the route of administration, SLNs can be synthesized by various techniques, which includes high-pressure homogenization, 48-50 microemulsions, 51-55 solvent emulsificationevaporation or diffusion, 56-58 water-inoil-in-water (w/o/w) double-emulsion method, high speed stirring, and/or ultrasonication. 59,60 Literature describes that SLNs can be used for all parental applications, right from intraarticular to intravenous administration. Many researchers suggest that SLNs have the capability to cross the blood-brain barrier; in support of this theory, a higher amount of drug was found in the brain after intravenous injection. 61-64 Thus, SLNs have the potential to be used as delivery vehicles to the brain for drugs like Dox, 61,62 paclitaxel, 61 camptothecin, 65 and others that cannot cross the blood-brain barrier. Significant efforts have been made by various research groups to study the efficacy of various drugs incorporated in SLNs after oral administration. Successful in vivo studies include oral delivery of tobramycin, 63 clozapine, 64 camptothecin, 65 rifampicin, 66 and isoniazid. 66 Furthermore, the utility of SLNs has been demonstrated through ocular, 67 buccal, 68 and rectal delivery. 69 Several research groups reported successful trials of SLNs in the topical application of all-trans retinoic acid, 70 clobetasol propionate, 71 cosmetics, 72 and in transdermal drug delivery of isotretinoin. 73 All nanoformulations

improved the stability and efficacy over traditional formulations. One study evaluated the transdermal formulation of naproxen, which utilized increased particle permeability at the nanoscale. 74 In this study, naproxen-loaded nanoparticles were formed into unilaminar films using Eudragit E-100 Rhm GmbH & Co. KG, Darmstadt, Germant. The nanoformulation was similar to films prepared by conventional methods from naproxen-methanol solutions. No organic solvents were used in the novel formulation. The study reported that there was no statistical difference between the amounts of naproxen that penetrated across the stratum corneum for either formulation. Surface modifications: targeting drug delivery Currently, the major struggle surrounding administration of anticancer agents is differentiating between cancerous and normal cells, thereby avoiding systemic toxicity and adverse events associated with conventional therapies. Targeted nanomedicines may aid in evading the adverse effects (such as immunosuppression, cardiomyopathy, and neurotoxicity) of traditional therapies, while also providing improved therapeutic efficacy. Throughout recent years, targeting nanomedicines have rapidly evolved. Passive targeting of tumors by nanoparticles takes advantage of their hyperpermeable cells. The rapid vascularization of tumors results in leaky, defective cells and impaired lymphatic drainage. Nanoparticles ranging from 10 to 100 nm in size then begin to accumulate within tumors because of their ineffective lymphatic drainage. This results in a phenomenon known as the enhanced permeability and retention effect. 75 Consideration of the size and surface properties of nanomedicines is vital. As previously noted, particles must be smaller than 100 nm to avoid uptake by the reticuloendothelial system and their surfaces should be hydrophilic to avoid clearance by macrophages. 76 Recent advances have led to the transformation from passive to active targeting. Active targeting has revolutionized nanomedicine, and this can now be achieved by a number of scientific interactions. Namely, lectin-carbohydrate, ligandreceptor, and antibody-antigen binding have provided the framework for such advancements. 77 These various interactions all generally result in preferential accumulation within the tumor-bearing organ or tumor cancer cells. Toxicity Although there is a tremendous increase in applications of nanoparticles in industrial materials, medical imaging, disease diagnoses, drug delivery, cancer treatment, gene therapy, and other areas, the possible toxic health effects of these nanoparticles associated with human exposure have not been studied properly. There is a good chance that these tiny particles may acquire unique surface properties in their nanosized form and may be toxic, causing adverse health effects. Although the use of nanotechnology in drug delivery,

280

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282

imaging, diagnosis, and cancer therapy may be beneficial, it may cause unintentional human exposure with unknown health effects that can only be imagined at present. Shvedova et al. 78 concluded the possibility of cancer and dermatological disorders associated with an excess in iron, alteration of pigmentation, inflammation, porphyria, and other consequences. In another study the same group observed that exposure to the unrefined single-walled carbon nanotubes can lead to increased pulmonary toxicity due to oxidative stress. 79 Further, it was shown by two other groups that these carbon nanotubes caused granulomas in rats and mice after acute exposure. 2,80 In two separate studies done by Lam et al. 81 and Poon and Burd, 82 the possibility of cytotoxicity was found in lesioned skin, growing human fibroblasts, and keratinocytes after the utilization of crystalline silver nanoparticles. There are also a few reports of toxicity of different inorganic nanoparticles like CdCl2, TiO2, gold nanoparticles, and iron oxide, at different levels. Although there are insufficient data for nanoparticles, a few nanomaterials and their relative cytotoxicity index on murine macrophage cells were described by Soto et al. in 2005. 83 Prospective applications Researchers at Rice University have constructed the world's smallest carthe nanocar. Composed of four C60 molecules (the wheels), connected by organic molecules (the chassis), the nanocar measures just 3-by-4 nm. 84 This provides support that materials can be moved around in a controlled manner at the nanoscale level using fullerenebased technology. Nanocars can be applied to targets and further enhance drug delivery systems. Nanomedicine may be the key to unlocking the innovation of oral delivery of peptides, such as oral insulin. Other examples include hormones, growth factors, clotting factors, and anticoagulants. The major obstacle to delivering peptides and protein medications is their limited bioavailability, inadequate stability, immunogenicity, and limited permeability across biological membranes. Nanotechnology can assist in overcoming these setbacks, allowing increased gastrointestinal absorption of these peptides. More than 200 proteins and peptides have been approved within the United States, and the bulk of these medications are delivered through injection. Nanotechnology will soon permit the fusion of peptides with implantable, oral, topical, and transdermal drug delivery systems. Implantable devices using pulsatile delivery systems capable of controlling drug administration may soon become prevalent. Implantable devices, or nanochips, promise improved disease management and may potentially be applied as antitumor therapy, gene therapy, or vaccines. Nanochips may even be used to assist in repairing damaged tissue, detecting mutated genes, or detecting high hormone levels indicative of certain malignancies. 85 Nanochips may be capable of triggering immediate

responses to inflamed, ischemic, or neoplastic tissues and simultaneously provide therapy to these tissues. Surprisingly, a silicon-based nanochannel has already been used to deliver antitumor compounds locally to unresectable tumors with zero-order kinetics. This implantable device circumvented the inconvenience of frequent local injections using novel nanotechnology applications. 86 Prospective delivery systems include biosensing functionalities with in vivo feedback, which will soon permit qsmartq drug delivery. 87,88 Nanotechnology is beginning to shape the way diseases are diagnosed, treated, and prevented; visions from the recent past will soon become a reality. Throughout recent years the nanotechnology field has grown exponentially and is predicted to be in full swing within the next 25 years. Multifunctional entities capable of detecting diseases, delivering medications, and monitoring will soon be within reach. New advancements in nanotechnology promise desired therapeutic effects while ameliorating side effects associated with many traditional medications. Nanomedicines propose solutions to the age-old problems associated with the solubility, bioavailability, immunocompatibility, cellular uptake, and cytotoxicity of many of these traditional medications. 34 Nanotechnology promises anything but minuscule effects, but most of these visions are hypothetical at this point. Accordingly, most pitfalls of molecular manufacturing have not yet been explored, because the benefits remain the dominant focus of researchers. Many questions surround nanotechnologynamely safety, cost, and ethical considerations. Regarding safety, little attention has been paid to environmental effects and the potential effects on the health of those manufacturing these particles. Of the US $1.2 billion spent on nanotechnology research, only 1% was spent on occupational health and safety research. 89 Granted, the applications of nanotechnology are limitless, but the development of safety guidelines by the government should strongly be considered. References
1. Bawa R. Patents and nanomedicine. Nanomedicine 2007;2:351-74. 2. Warheit DB, Laurence BR, Reed KL, Roach DH, Reynolds GA, Webb TR. Comparative pulmonary toxicity assessment of single-wall carbon nanotubes in rats. Toxicol Sci 2004;77:117-25. 3. Lam CW, James JT, Latch JN, Hamilton Jr RF, Holian A. Pulmonary toxicity of simulated lunar and Martian dusts in mice: II. Biomarkers of acute responses after intratracheal instillation. Inhal Toxicol 2002;14: 917-28. 4. National Nanotechnology Initiative. What is nanotechnology? Available at: http://www.nano.gov/html/facts/whatIsNano.html Accessed 25 November 2007. 5. Moghimi SM, Hunter AC, Murray JC. Long-circulating and targetspecific nanoparticles: theory to practice. Pharmacol Rev 2001;53: 283-318. 6. Torchilin VP. Targeted pharmaceutical nanocarriers for cancer therapy and imaging. AAPS J 2007;9:E128-47. 7. Torchilin VP, Weissing V, editors. Liposomes: a practical approach. 2nd ed. New York: Oxford University Press; 2003.

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282 8. Fahmy TM, Fong PM, Park J, Constable T, Saltzman WM. Nanosystems for simultaneous imaging and drug delivery to T cells. AAPS J 2007;9:E171-80. 9. Torchilin VP. Nanocarriers. Pharm Res 2007;24:2333-4. 10. Gaucher G, Dufresne MH, Sant VP, Kang N, Maysinger D, Leroux JC. Block copolymer micelles: preparation, characterization and application in drug delivery. J Control Release 2005;109:169-88. 11. Adams ML, Lavasanifar A, Kwon GS. Amphiphilic block copolymers for drug delivery. J Pharm Sci 2003;92:1343-55. 12. Kwon GS. Polymeric micelles for delivery of poorly water-soluble compounds. Crit Rev Ther Drug Carrier Syst 2003;20:357-403. 13. Bae Y, Jang WD, Nishiyama N, Fukushima S, Kataoka K. Multifunctional polymeric micelles with folate-mediated cancer cell targeting and pH-triggered drug releasing properties for active intracellular drug delivery. Mol Biosyst 2005;1:242-50. 14. Tang N, Du G, Wang N, Liu C, Hang H, Liang W. Improving penetration in tumors with nanoassemblies of phospholipids and doxorubicin. J Natl Cancer Inst 2007;99:1004-15. 15. Simon JA. Estradiol in micellar nanoparticles: the efficacy and safety of a novel transdermal drug-delivery technology in the management of moderate to severe vasomotor symptoms. Menopause 2006;13:222-31. 16. Wiener EC, Brechbiel MW, Brothers H, Magin RL, Gansow OA, Tomalia DA, et al. Dendrimer-based metal chelates: a new class of magnetic resonance imaging contrast agents. Magn Reson Med 1994;31:1-8. 17. Li Y, Cheng Y, Xu T. Design, synthesis and potent pharmaceutical applications of glycodendrimers: a mini review. Curr Drug Discov Technol 2007;4:246-54. 18. Kojima C, Kono K, Maruyama K, Takagishi T. Synthesis of polyamidoamine dendrimers having poly(ethylene glycol) grafts and their ability to encapsulate anticancer drugs. Bioconjug Chem 2000;11:910-7. 19. Fu HL, Cheng SX, Zhang XZ, Zhuo RX. Dendrimer/DNA complexes encapsulated in a water soluble polymer and supported on fast degrading star poly(DL-lactide) for localized gene delivery. J Control Release 2007;124:181-8. 20. Venditto VJ, Regino CA, Brechbiel MW. PAMAM dendrimer based macromolecules as improved contrast agents. Mol Pharm 2005;2: 302-11. 21. Kobayashi H, Kawamoto S, Jo SK, Bryant Jr HL, Brechbiel MW, Star RA. Macromolecular MRI contrast agents with small dendrimers: pharmacokinetic differences between sizes and cores. Bioconjug Chem 2003;14:388-94. 22. Cheng Y, Wang J, Rao T, He X, Xu T. Pharmaceutical applications of dendrimers: promising nanocarriers for drug delivery. Front Biosci 2008;13:1447-71. 23. Cheng Y, Gao Y, Rao T, Li Y, Xu T. Dendrimer-based prodrugs: design, synthesis, screening and biological evaluation. Comb Chem High Throughput Screen 2007;10:336-49. 24. Mecke A, Uppuluri S, Sassanella TM, Lee DK, Ramamoorthy A, Baker Jr JR, et al. Direct observation of lipid bilayer disruption by poly (amidoamine) dendrimers. Chem Phys Lipids 2004;132:3-14. 25. Rosa Borges A, Schengrund CL. Dendrimers and antivirals: a review. Curr Drug Targets Infect Disord 2005;5:247-54. 26. Larson DR, Zipfel WR, Williams RM, Clark SW, Bruchez MP, Wise FW, et al. Water-soluble quantum dots for multiphoton fluorescence imaging in vivo. Science 2003;300:1434-6. 27. Kaji N, Tokeshi M, Baba Y. Quantum dots for single bio-molecule imaging. Anal Sci 2007;23:21-4. 28. Derfus AM, Chen AA, Min DH, Ruoslahti E, Bhatia SN. Targeted quantum dot conjugates for siRNA delivery. Bioconjug Chem 2007;18:1391-6. 29. Muller-Borer BJ, Collins MC, Gunst PR, Cascio WE, Kypson AP. Quantum dot labeling of mesenchymal stem cells. J Nanobiotechnol 2007;5:9. 30. Alivisatos AP, Gu W, Larabell C. Quantum dots as cellular probes. Annu Rev Biomed Eng 2005;7:55-76.

281

31. Hoshino A, Fujioka K, Oku T, Nakamura S, Suga M, Yamaguchi Y, et al. Quantum dots targeted to the assigned organelle in living cells. Microbiol Immunol 2004;48:985-94. 32. Howarth M, Takao K, Hayashi Y, Ting AY. Targeting quantum dots to surface proteins in living cells with biotin ligase. Proc Natl Acad Sci U S A 2005;102:7583-8. 33. Bagalkot V, Zhang L, Levy-Nissenbaum E, Jon S, Kantoff PW, Langer R, et al. Quantum dot-aptamer conjugates for synchronous cancer imaging, therapy, and sensing of drug delivery based on bi-fluorescence resonance energy transfer. Nano Lett 2007;7:3065-70. 34. Goldberg M, Langer R, Jia X. Nanostructured materials for applications in drug delivery and tissue engineering. J Biomater Sci Polym Ed 2007;18:241-68. 35. Gao X, Cui Y, Levenson RM, Chung LW, Nie S. In vivo cancer targeting and imaging with semiconductor quantum dots. Nat Biotechnol 2004;22:969-76. 36. Derfus AM, Chan WCW, Bhatia SN. Probing the cytotoxicity of semiconductor quantum dots. Nano Lett 2004;4:11-8. 37. Cho SJ, Maysinger D, Jain M, Roder B, Hackbarth S, Winnik FM. Long-term exposure to CdTe quantum dots causes functional impairments in live cells. Langmuir 2007;23:1974-80. 38. Green M, Howman E. Semiconductor quantum dots and free radical induced DNA nicking. Chem Commun (Camb) 2005:121-3. 39. Choi AO, Cho SJ, Desbarats J, Lovric J, Maysinger D. Quantum dot induced cell death involves Fas upregulation and lipid peroxidation in human neuroblastoma cells. J Nanobiotechnol 2007;5:1. 40. Lovric J, Cho SJ, Winnik FM, Maysinger D. Unmodified cadmium telluride quantum dots induce reactive oxygen species formation leading to multiple organelle damage and cell death. Chem Biol 2005;12:1227-34. 41. Zhu MQ, Han JJ, Li AD. CdSe/CdS/SiO2 core/shell/shell nanoparticles. J Nanosci Nanotechnol 2007;7:2343-8. 42. Mo Y, Barnett ME, Takemoto D, Davidson H, Kompella UB. Human serum albumin nanoparticles for efficient delivery of Cu, Zn superoxide dismutase gene. Mol Vis 2007;13:746-57. 43. Langer R, Folkman J. Polymers for the sustained release of proteins and other macromolecules. Nature 1976;263:797-800. 44. Till MC, Simkin MM, Maebius S. Nanotech meets the FDA: a success story about the first nanoparticulate drugs approved by the FDA. Nanotechnol Law Business 2005;2:163-7. 45. Damascelli B, Cantu G, Mattavelli F, Tamplenizza P, Bidoli P, Leo E, et al. Intraarterial chemotherapy with polyoxyethylated castor oil free paclitaxel, incorporated in albumin nanoparticles (ABI-007): Phase II study of patients with squamous cell carcinoma of the head and neck and anal canal: preliminary evidence of clinical activity. Cancer 2001;92:2592-602. 46. Sparreboom A, Scripture CD, Trieu V, Williams PJ, De T, Yang A, et al. Comparative preclinical and clinical pharmacokinetics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cremophor (Taxol). Clin Cancer Res 2005;11:4136-43. 47. Kattan J, Droz JP, Couvreur P, Marino JP, Boutan-Laroze A, Rougier P, et al. Phase I clinical trial and pharmacokinetic evaluation of doxorubicin carried by polyisohexylcyanoacrylate nanoparticles. Invest New Drugs 1992;10:191-9. 48. Schwarz C, Mehnert W, Lucks JS, Muller RH. Solid lipid nanoparticles (SLN) for controlled drug delivery: I. Production, characterization, and sterilization. J Control Release 1994;30:83-96. 49. Muller RH, Schwarz C, Mehnert W, Lucks JS. Production of solid lipid nanoparticles (SLN) for controlled drug delivery. Proc Int Symp Control Rel Bioact Mater 1993;20:480-1. 50. Liedtke S, Wissing S, Muller RH, Mader K. Influence of high pressure homogenisation equipment on nanodispersions characteristics. Int J Pharm 2000;196:183-5. 51. Gasco MR, inventor. Method for producing solid lipid microspheres having a narrow size distribution. United States patent US 5250236. 1993 Oct 5.

282

W.E. Bawarski et al / Nanomedicine: Nanotechnology, Biology, and Medicine 4 (2008) 273282 71. Kalariya M, Padhi BK, Chougule M, Misra A. Clobetasol propionate solid lipid nanoparticles cream for effective treatment of eczema: formulation and clinical implications. Indian J Exp Biol 2005;43: 233-40. 72. Wissing SA, Muller RH. Cosmetic applications for solid lipid nanoparticles (SLN). Int J Pharm 2003;254:65-8. 73. Liu J, Hu W, Chen H, Ni Q, Xu H, Yang X. Isotretinoin-loaded solid lipid nanoparticles with skin targeting for topical delivery. Int J Pharm 2007;328:191-5. 74. Ganem-Quintanar A, Silva-Alvarez M, Alvarez-Roman R, CasasAlancaster N, Cazares-Delgadillo J, Quintanar-Guerrero D. Design and evaluation of a self-adhesive naproxen-loaded film prepared from a nanoparticle dispersion. J Nanosci Nanotechnol 2006;6:3235-41. 75. Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res 1986;46:6387-92. 76. Jain RK. Transport of molecules, particles, and cells in solid tumors. Annu Rev Biomed Eng 1999;1:241-63. 77. Guo X, Szoka Jr FC. Chemical approaches to triggerable lipid vesicles for drug and gene delivery. Acc Chem Res 2003;36:335-41. 78. Shvedova AA, Castranova V, Kisin ER, Schwegler-Berry D, Murray AR, Gandelsman VZ, et al. Exposure to carbon nanotube material: assessment of nanotube cytotoxicity using human keratinocyte cells. J Toxicol Environ Health A 2003;66:1909-26. 79. Shvedova AA, Kisin E, Murray AR, Schwegler-Berry D, Gandelsman VZ, Baron P, et al. Exposure of human bronchial cells to carbon nanotubes caused oxidative stress and cytotoxicity. Proceedings of the Meeting of the Society for Free Radical Research (SFRR) Europe 2003, June 26-29; Ioannina, Greece. Philadelphia: Taylor & Francis Group; 2004. p. 91-103. 80. Lam CW, James JT, McCluskey R, Hunter RL. Pulmonary toxicity of single-wall carbon nanotubes in mice 7 and 90 days after intratracheal instillation. Toxicol Sci 2004;77:126-34. 81. Lam PK, Chan ES, Ho WS, Liew CT. In vitro cytotoxicity testing of a nanocrystalline silver dressing (Acticoat) on cultured keratinocytes. Br J Biomed Sci 2004;61:125-7. 82. Poon VK, Burd A. In vitro cytotoxity of silver: implication for clinical wound care. Burns 2004;30:140-7. 83. Soto K, Carrasco A, Powell TG, Garza KM, Murr LE. Comparative in vitro cytotoxicity assessment of some manufactured nanoparticulate materials characterized by transmission electron microscopy. J Nanoparticle Res 2005;7:145-69. 84. Shirai Y, Osgood AJ, Zhao Y, Yao Y, Saudan L, Yang H, et al. Surfacerolling molecules. J Am Chem Soc 2006;128:4854-64. 85. Mousa SA, Bharali DJ, Armstrong D. From nutraceuticals to pharmaceuticals to nanopharmaceuticals: a case study in angiogenesis modulation during oxidative stress. Mol Biotechnol 2007;37:72-80. 86. Lesinski GB, Sharma S, Varker KA, Sinha P, Ferrari M, Carson WE. Release of biologically functional interferon-alpha from a nanochannel delivery system. Biomed Microdevices 2005;7:71-9. 87. Anderson DG, Burdick JA, Langer R. Materials science. Smart biomaterials. Science 2004;305:1923-4. 88. LaVan DA, McGuire T, Langer R. Small-scale systems for in vivo drug delivery. Nat Biotechnol 2003;21:1184-91. 89. Maynard AD. Nanotechnology: a research strategy for addressing risks. Woodrow Wilson International Center for Scholars: Project on emerging nanotechnologies. 2006. Available from: http://www.nanotechproject.org/file_download/files/PEN3_Risk.pdf.

52. Cavalli R, Marengo E, Rodriguez L, Gasco MR. Effects of some experimental factors on the production process of solid lipid nanoparticles. Eur J Pharm Biopharm 1996;43:110-5. 53. Gasco MR. Solid lipid nanospheres from warm microemulsions. Pharm Technol Eur. 1997:9, 52-4, 6-8. 54. Cavalli R, Caputo O, Marengo E, Pattarino F, Gasco MR. The effect of the components of microemulsions on both size and crystalline structure of solid lipid nanoparticles (SLN) containing a series of model molecules. Pharmazie 1998;53:392-6. 55. Igartua M, Saulnier P, Heurtault B, Pech B, Proust JE, Pedraz JL, et al. Development and characterization of solid lipid nanoparticles loaded with magnetite. Int J Pharm 2002;233:149-57. 56. Sjostrom B, Kaplun A, Talmon Y, Cabane B. Structures of nanoparticles prepared from oil-in-water emulsions. Pharm Res 1995;12:39-48. 57. Shahgaldian P, Da Silva E, Coleman AW, Rather B, Zaworotko MJ. Paraacyl-calix-arene based solid lipid nanoparticles (SLNs): a detailed study of preparation and stability parameters. Int J Pharm 2003;253:23-38. 58. Dubes A, Parrot-Lopez H, Abdelwahed W, Degobert G, Fessi H, Shahgaldian P, et al. Scanning electron microscopy and atomic force microscopy imaging of solid lipid nanoparticles derived from amphiphilic cyclodextrins. Eur J Pharm Biopharm 2003;55:279-82. 59. Eldem T, Speiser P, Hincal A. Optimization of spray-dried and -congealed lipid micropellets and characterization of their surface morphology by scanning electron microscopy. Pharm Res 1991;8:47-54. 60. Speiser P, inventor; Dr. Rentschler Arzneimittel GmbH & Co, assignee. Lipid nano-pellets as excipient system for perorally administered drugs. United States patent US 4880634. 1989 Nov 14. 61. Fundaro A, Cavalli R, Bargoni A, Vighetto D, Zara GP, Gasco MR. Non-stealth and stealth solid lipid nanoparticles (SLN) carrying doxorubicin: pharmacokinetics and tissue distribution after i.v. administration to rats. Pharmacol Res 2000;42:337-43. 62. Zara GP, Cavalli R, Bargoni A, Fundaro A, Vighetto D, Gasco MR. Intravenous administration to rabbits of non-stealth and stealth doxorubicin-loaded solid lipid nanoparticles at increasing concentrations of stealth agent: pharmacokinetics and distribution of doxorubicin in brain and other tissues. J Drug Target 2002;10:327-35. 63. Bargoni A, Cavalli R, Zara GP, Fundaro A, Caputo O, Gasco MR. Transmucosal transport of tobramycin incorporated in solid lipid nanoparticles (SLN) after duodenal administration to rats. Part II tissue distribution. Pharmacol Res 2001;43:497-502. 64. Manjunath K, Venkateswarlu V. Pharmacokinetics, tissue distribution and bioavailability of clozapine solid lipid nanoparticles after intravenous and intraduodenal administration. J Control Release 2005;107:215-28. 65. Yang SC, Lu LF, Cai Y, Zhu JB, Liang BW, Yang CZ. Body distribution in mice of intravenously injected camptothecin solid lipid nanoparticles and targeting effect on brain. J Control Release 1999;59:299-307. 66. Pandey R, Sharma S, Khuller GK. Oral solid lipid nanoparticle-based antitubercular chemotherapy. Tuberculosis (Edinb) 2005;85:415-20. 67. Myles ME, Neumann DM, Hill JM. Recent progress in ocular drug delivery for posterior segment disease: emphasis on transscleral iontophoresis. Adv Drug Deliv Rev 2005;57:2063-79. 68. Smart JD. Buccal drug delivery. Expert Opin Drug Deliv 2005;2:507-17. 69. Mackay M, Phillips J, Hastewell J. Peptide drug delivery: colonic and rectal absorption. Adv Drug Deliv Rev 1997;28:253-73. 70. Yamaguchi Y, Nakamura N, Nagasawa T, Kitagawa A, Matsumoto K, Soma Y, et al. Enhanced skin regeneration by nanoegg formulation of all-trans retinoic acid. Pharmazie 2006;61:117-21.

You might also like