Sie sind auf Seite 1von 13

[Ide@s CONCYTEG, 6 (72): Junio, 2011] ISSN: 2007-2716

Cmo citar: Ober. C. A. and R. B. Gupta (2011), Nanoparticle Technology for Drug Delivery, Ide@s CONCYTEG, 6 (72), pp. 714-726.

Nanoparticle Technology for Drug Delivery


Courtney A. Ober1 Ram B. Gupta2

Resumen
La tecnologa de nanopartculas se espera que revolucione la manera en que se lleva a cabo la administracin de frmacos. Las tecnologas de nanopartculas tienen la capacidad de mejorar la eficacia de los medicamentos, reducir al mnimo los efectos secundarios, y proporcionar una entrega especfica, slo para nombrar algunos. Con el fin de aprovechar las aplicaciones y ventajas de las nanopartculas, una comprensin fundamental de sus propiedades, produccin, y caracterizacin es necesaria. En esta revisin se tratar de explorar estos temas en relacin con la administracin de frmacos. Palabras clave: administracin de frmacos, caracterizacin de nanopartculas, produccin de nanopartculas.

Summary
Nanoparticle technology is expected to revolutionize the way in which drug delivery is conducted. Nanoparticle technologies have the capacity to improve drug efficacy, minimize side-effects, and provide targeted delivery, just to name a few. In order to exploit the applications and advantages of nanoparticles, a fundamental understanding of their properties, production, and characterization is necessary. This review will seek to explore these topics as they relate to drug delivery. Keywords: drug delivery, nanoparticle characterization, nanoparticle production.

Ms. Courtney A. Ober obtained a B.S. in chemical engineering from the University of Virginia in Charlottesville, VA. She is currently pursuing a Ph.D. in chemical engineering at Auburn University in Auburn, AL. Her current research interests are nanoparticles for drug delivery, nanomixing, and pharmaceutical cocrystals. Email: CourtneyOber@gmail.com 2 Dr. Ram B. Gupta is WVW chair professor in chemical engineering at Auburn University in Auburn, AL. He obtained his Ph.D. from the University of Texas in Austin, TX. His current research interests are supercritical carbon dioxide technology, nanomedicine, and liquid fuels from biomass. Email: gupta@auburn.edu
1

ISBN 978-607-8164-02-8

714

[Ide@s CONCYTEG 6(72): Junio, 2011]

Nanoparticles: Small Size, Big Advantages

T
the

he

most

defining

property

of

Decreasing particle size to the nanoscale dramatically increases the surface area for a given quantity of material. In addition to increased surface area, the percentage of molecules on the surface also increases. These effects are shown in Table 2 for spherical particles of a 1 nm drug molecule (Gupta and Kompella, 2006).

nanoparticles, their small size, offers a number of unique advantages for

drug delivery. Table 1 compares the size of various biological entities, illustrating that the nanometer scale is found frequently in biological systems (Gupta and Kompella, 2006). By matching the treatment scale with biological entities to be treated, nanoparticles offer a number of treatment strategies unachievable with conventional medicine. For example, 100 nanometer (nm) particles can diffuse into the submuscosal layer of the gastrointestinal tract while larger microparticles et al., 1996). are excluded, persisting predominantly in the epithelial lining (Desai

Table 2. Surface area and percentage of surface molecules for different particle sizes

Particle diameter (nm) 1 10 100 1,000 10,000

Surface Area (nm ) 12.6 1260 1.26 105 1.26 107 1.26 109
2

Surface molecules (%) 100.00 27.10 2.97 0.30 0.03

Table 1. Size comparison of various biological entities

Source: self-elaboration.

Object DNA (diameter) Ribosome Virus Bacterium Red blood cell Human hair (diameter) double

Size (nm) helix 3 10 100 1,000 5,000 50,000 The increased surface area of nanoparticles can significantly increase the dissolution of poorly water-soluble drugs, which are estimated to comprise 40% of drugs under development (Lipinski, 2001, 2002). The relationship between surface area and drug dissolution is governed by the NoyesWhitney equation,

Source: self-elaboration.

Dissolution Rate

(1)

ISBN 978-607-8164-02-8

715

Nanoparticle Technology for Drug Delivery Courtney A. Ober y Ram B. Gupta

where A is surface area, D is diffusivity, h is boundary layer thickness, Cs is saturation solubility, and Cb is bulk concentration (Dressman et al., 1998). Nanoization is an advantageous method for increasing the dissolution of poorly water-soluble drugs since it is a technique that can be applied to virtually all pharmaceutical compounds. Due to their small size, nanoparticles are less prone to gravitational settling and can be easily suspended in liquid formulations. The settling velocity, v, of a particle is given by Stokes law,

Brownian motion, allowing the particles to remain suspended in solution. Using nanoparticles in liquid drug suspensions gives a more homogeneous product with longer shelf life and negates the need for shaking before use. Nanoparticles can also offer unique magnetic and optical properties with relevance in targeted treatment, diagnostics, and imaging. For example, ferromagnetic materials lose there magnetization at particle sizes less than 20 nm due to loss of magnetic domains, but still respond to a magnetic field. Such particles can be directed to tumors and locally heated by pulsed electromagnetic radiation, resulting in perforation of the tumor cell membranes and enhanced drug delivery (Gupta and Kompella, 2006). Due to surface plasmon resonance, the color of nanoparticles changes with particle size which can be useful in diagnostic and imaging applications.

(2) where d is particle diameter, g is gravitational acceleration, s is solid density, l is liquid density, and l is liquid viscosity. Resistance to settling results from random thermal motion, Brownian motion, for which the Brownian displacement, x, can be calculated by,

Nanoparticle Production: TopDown or Bottom-Up?


(3) The unique properties of nanoparticles just where kB is the Boltzmann constant, T is absolute temperature, t is time, is liquid viscosity, and d is particle diameter. As particle size decreases to the nanoscale, their settling velocity becomes less than their mentioned can only be exploited if the particles can be commercially produced through safe and economically viable technologies. There exist two categories of

nanoparticle production technologies: top-

ISBN 978-607-8164-02-8

716

[Ide@s CONCYTEG 6(72): Junio, 2011]

down and bottom-up (Reverchon and Adami, 2006). These two categories are illustrated in Figure 1. Top-down technologies utilize mechanical forces to break-down macroscopic particles to nanoscale size. Bottom-up technologies build-up nanoscale particles from molecular solutions. Examples of top-down processes include pearl/ball milling and high pressure homogenization. Examples of bottom-up technologies include supercritical fluid precipitation and emulsification-diffusion.

Pearl/Ball Milling
Traditional micronization equipment, such as jet mills and rotor-stator colloid mills, are ineffective at creating sufficient quantities of nanoparticles. Pearl mills, however, have been found effective at creating nanosuspensions when run for sufficient times (Liversidge et al., 1992; MeriskoLiversidge et al., 2003; Merisko-Liversidge et al., 1996). Pearl mills generally consist of a stainless steel vessel filled with steel, glass, or hard polystyrene balls. Operation can include moving the balls with an impeller while keeping the vessel static or moving the entire vessel such that the balls inside also move. A schematic of a rotating pearl/ball mill apparatus is shown in Figure 2.

Figure 1. Comparison of top-down (top) and bottom-up (bottom) nanoparticle production technologies

Milling

Figure 2. Rotating pearl/ball mill vessel with drug nanoparticles


Precipitation

Source: Gupta and Kompella, 2006

Pearls/Balls Drug nanoparticles

Breaking It Down
The use of mechanical forces to break down macroscopic pearl/ball materials milling and into high nanoscale pressure
Source: self-elaboration.

particles are categorized into two groups: homogenization.

A macromolecular suspension is made of drug particles in a stabilizer or surfactant containing solution. The suspension is then introduced to the pearl/ball mill vessel and the mill is operated until drug particle size is sufficiently reduced, providing a

ISBN 978-607-8164-02-8

717

Nanoparticle Technology for Drug Delivery Courtney A. Ober y Ram B. Gupta

nanosuspension. The stabilizer or surfactant prevents particle agglomeration and promotes nanosuspension stability. Care must be taken to ensure that the balls are not eroded over extended operation and thus contaminate the drug suspension. Also, since milling times can range from hours to days, the drug must be stable at the operational conditions (i.e. temperature) for it to be processed using pearl/ball milling.

Figure 3. Piston gas homogenization

Source: self-elaboration.

High Pressure Homogenization


High pressure homogenization uses forces of impaction to produce nanoparticle suspensions from microparticle suspensions. The two most common homogenization configurations are piston-gap, shown in Figure 3, and jet-stream, shown in Figure 4. The piston-gap configuration forces a macrosuspension through a small gap (~10 m) causing particle diminution by shear, impaction, Kompella, configuration and cavitation The two (Gupta and 2006). collides jet-stream high-velocity

Figure 4. Jet-stream homogenization

Source: self-elaboration.

streams of macrosuspension causing particle diminution by impaction. The pressures typically required to obtain nanosuspensions are 1000-1500 bar and the number of homogenization cycles can vary from 10-20, depending on the drug (Gupta and Kompella, 2006).

High-pressure homogenization processes are well-suited to scale-up, with such processes already being used in the food industry for homogenization of milk. The limitation of such processes is for hard or tough drugs which are resistance to crack and fracture, and therefore cannot be broken down merely by particle collision.

ISBN 978-607-8164-02-8

718

[Ide@s CONCYTEG 6(72): Junio, 2011]

Top-down technologies for the production of nanoparticles are advantageous because of process simplicity and applicability to a wide range of materials. Although the products obtained by pearl/ball milling and high pressure homogenization were liquid nanosuspensions, technologies such as spray drying can be used to obtain a solid formulation more amenable to the patient. Examples produced of by two drugs commercially nanoparticle

Figure 5. Generic pressure versus temperature phase diagram highlighting the supercritical fluid region

Source: self-elaboration.

top-down

production technologies are Rapamune and Emend (Gupta and Kompella, 2006).

Supercritical carbon dioxide (CO2) is the most commonly used supercritical fluid for pharmaceutical particle production because it is nonflammable, nontoxic, inexpensive, and has mild critical parameters (Tc = 31.3 C, Pc = 73.7 bar). Supercritical CO2 has been used as both a solvent, in the rapid expansion of supercritical solution (RESS), and as an antisolvent, in the supercritical antisolvent (SAS), particle production technologies.

Supercritical Fluids for Production of Nanoparticles


The application of supercritical fluids for the production widespread of use nanoparticles due to a has found of number

advantageous properties. Supercritical fluids exist at temperatures beyond their critical temperature (Tc) and pressures above their critical pressure (Pc), as shown in Figure 5. Supercritical fluids have diffusivities higher than those of traditional liquid solvents, viscosities similar to gases, and densities that can be tuned by small changes in pressure, all of which make them unique reaction media.

Rapid Expansion of Supercritical Solution (RESS)


In the RESS method, the pressure-dependent solubilizing power of supercritical CO2 is exploited. A bulk drug is dissolved in supercritical CO2 in a high pressure vessel. The solution is then depressurized through a nozzle into a collection vessel at ambient conditions. When depressurization occurs, the supercritical CO2 becomes gaseous CO2, in

ISBN 978-607-8164-02-8

719

Nanoparticle Technology for Drug Delivery Courtney A. Ober y Ram B. Gupta

which the drug is not soluble, and the drug precipitates. The faster the rate of depressurization, the smaller the particles will precipitate. A schematic of the RESS process is given in Figure 6 (Gupta and Kompella, 2006).
Figure 6. Schematic of RESS process
Drug + CO2 Nozzle

of CO2 for

many

drugs by

utilizing

supercritical CO2 as an antisolvent. In this method, the drug is dissolved in a liquid organic solvent and this solution is sprayed through a fine nozzle into a high pressure vessel filled with supercritical CO2. As the CO2 dissolves into the liquid solvent, the solubilizing power of the organic solvent is reduced, inducing supersaturation and causing particle precipitation. Excess CO2 is flushed through the vessel to remove residual solvent and the vessel is depressurized to collect the particles. A schematic of the SAS process is shown in Figure 7 (Gupta and Kompella, 2006).

Supercritical CO2

Bulk Drug

Particles

CO2 gas

Source: self-elaboration.

As with all particle production technologies, the conditions under which the process is carried out, such as solubilization temperature, expansion temperature, pressure drop across nozzle, and nozzle geometry, as well as the molecular structure of the drug, greatly effect particle morphology. A

Figure 7. Schematic of SAS process


Drug + Solvent Solvent + CO2

Particles

disadvantage of the RESS process is the limited solubility of many pharmaceutical compounds in supercritical CO2, for which the SAS process may be more suitable.

Supercritical CO2

Source: self-elaboration.

A number of variations on the RESS and SAS processes have also been introduced in the literature to control particle size, reduce agglomeration, and further improve particle

Supercritical Antisolvent (SAS)


Contrary to the RESS process, the SAS process relies on the weak solubilizing power

properties. In general, supercritical CO2 processes reduce organic solvent use and produce dry nanoparticle powders suitable for

ISBN 978-607-8164-02-8

720

[Ide@s CONCYTEG 6(72): Junio, 2011]

direct capsule filling or table compression. Furthermore, a comprehensive compilation of solubility data in supercritical carbon dioxide has been published by Gupta and Shim, which can facilitate selection of an appropriate supercritical CO2 process for the compound under consideration (Gupta and Shim, 2007). Nonetheless, companies have been slow to adopt supercritical technology as high pressure equipment requires additional safety measures.

glycolide (PLGA), poly(lactic acid) (PLA), and polymethacrylate (PMA) while stabilizers include polysorbate, polyvinyl alcohol, albumin, and poloxamer (Bala et al., 2004; O'Donnell and McGinity, 1997).

Figure 8. Schematic of emulsification solvent evaporation process


Homogenizer Drug + Solvent + Polymer droplets Drug + Polymer Nanoparticles

Water + Stabilizer Solvent Evaporation

Emulsification for Polymer and Protein Stabilized Nanoparticles


An emulsion is a metastable dispersion of two or more immiscible liquids in the presence of surfactant. Emulsions can be used to produce nanoparticles by dissolving a drug and polymer in a water-immiscible solvent and adding the mixture dropwise to an aqueous solution containing surfactant. Shear is applied through The homogenization harden or into sonication to decrease droplet size to the nanoscale. droplets nanoparticles by evaporation of the solvent, and can be separated from the aqueous phase by lyophilization. The above described process is termed emulsification solvent evaporation and a schematic of the process is given in Figure 8. Polymers commonly used in this process are poly(d,l-lactide-co-

Source: self-elaboration.

Final particle properties are dictated by the homogenization duration and intensity, type and amount of surfactant, drug to polymer loading, and rate of solvent removal. One advantage of this process is the ability to enhance drug delivery by selection of appropriate poloxamer surfactants. stabilizers, in For example, to addition

stabilizing the nanoparticles, also exhibit mucoadhesive properties which can enhance oral drug delivery (Gupta and Kompella, 2006). An example of a drug product processed by emulsification solvent evaporation is Abraxane, a cancer therapy treatment drug which consists of 130 nanometer paclitaxel nanoparticles stabilized with albumin (Gupta and Kompella, 2006).

ISBN 978-607-8164-02-8

721

Nanoparticle Technology for Drug Delivery Courtney A. Ober y Ram B. Gupta

Nanoparticle Characterization for Drug Delivery


While nanoparticle characterization is quite similar across disciplines, there are a number of clinically relevant parameters which must be considered for drug delivery applications. Such parameters include particle size, size dispersity, structure, surface characteristics, crystallinity, composition, and dissolution. The size of a nanoparticle defines through which biological routes the particle can travel and through which it will be excluded, as will be discussed in a subsequent section. A number of methods can be used for nanoparticle sizing. With counting methods, such as single-particle optical sensing (SPOS) or microscopy, that measure the size of individual particles, a significant number of particles must be measured to ensure an accurate reflection of the sample. Separation methods, such as filtration or field flow fractionation (FFF), physically order a sample according to particle size, taking all particles into account. Often, two the or more Some analysis techniques, such as X-ray diffraction (XRD) and differential scanning calorimetry (DSC), are conducted the same as they would be for macroscopic materials but can provide clinically relevant information for drug nanoparticles. Crystalline drug formulations are most stable, and it is generally desirable that the crystallinity of a ISBN 978-607-8164-02-8 722 size complementary methods can be used for verification. Knowing particle dispersity of a sample is also clinically important. Even through the average particle size might be well below the required limit for intravenous injection, the presence of a few larger microparticles may increase risk of an embolism. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) can be used for direct observation of nanoparticles, with the former more suited to observing particle morphology and the latter more suited to observing the internal structure. Upon entering circulation in a biological system, the surface of nanoparticles will become coated with lipoproteins and other species (Moghimi and Szebeni, 2003). The type and surface coverage of such species are important to predicting the persistence and biodistribution of the nanoparticles. Electrophoresis is one technique that can be used to identify adsorbant surface proteins on a nanoparticle. Surface hydrophobicity also plays a role in determining the in vivo behavior of nanoparticles and can be characterized by hydrophobic interaction chromatography (HIC).

[Ide@s CONCYTEG 6(72): Junio, 2011]

drug

compound

be

maintained

when

converting it to a nanoparticle formulation with better delivery properties. It should be noted, however, that broadening of the XRD peaks will occur for nanoparticles less than 100 nm in diameter (Gupta and Kompella, 2006). From the melting point and enthalpy data obtained through DSC, the phases that exist in a nanoparticle formulation and the degree to which they interact can be determined. For nanoparticles produced to enhance the dissolution of poorly watersoluble drugs, dissolution testing is important. Standard protocols have been developed and apparatuses are commercially available which can measure the release of drug into a physiologically simulated fluid over time. The characterization of drug nanoparticles is heavily dependent For upon their intended properties, more application. others some

Targeted Delivery: Getting the Nanoparticles Where They are Needed


The size of nanoparticles makes them suitable for a number of delivery strategies. Possible strategies include injection, oral delivery, ocular delivery, delivery to the brain, and gene delivery. Unlike microparticles, nanoparticles do not pose risk of embolism when administered intravenously due to their small size. Their intravenous administration is also suitable for targeting tumors, inflamed, and infected vascular regions, all of which are characterized by leaky vasculature. In order to diffuse through these vascular pores, which range in size from 300-700 nm, the nanoparticles should be <250 nm for maximum efficacy (Wu et al., 1993). Injectable nanoparticulate systems could be in the form of crystalline drug nanosuspensions for immediate release or polymeric drug nanoparticles for a more sustained release (Gupta and Kompella, 2006). Drugs are often formulated for oral delivery due to ease of administration and patient convenience. Nanoparticles are no exception, with a number of nanoparticle drugs currently being examined for oral formulation. Primary goals of oral nanoparticle delivery are local

macroscopic techniques are suitable while for characterization techniques tailored to the nanoscale are required. The characterization techniques used for drug nanoparticles are not unlike those used for nanoparticles in other fields, but the findings should be considered in view of their clinical relevance and acceptability.

ISBN 978-607-8164-02-8

723

Nanoparticle Technology for Drug Delivery Courtney A. Ober y Ram B. Gupta

colonic treatment for ulcerative colitis, enhanced bioavailability for poorly watersoluble drugs, and systemic targeting through circulatory uptake from the intestine. Two less understood but critically important drug delivery strategies are ocular delivery and delivery to the brain. Topical drug delivery to the eye is challenging due to natural tear flow, blinking, and multiple tissue and vascular barriers. Furthermore, treating ocular tissue through systemic circulation requires high levels of dosing, which can prove systemically toxic, in order to compensate for the small amount of drug that is actually delivered to the ocular tissue. Nanoparticles may prove relevant for ocular drug delivery due to their improved accessibility to deep ocular tissues, and thus their ability to maintain improved residence time in the eye. Likewise to the challenges encountered in ocular delivery, the blood brain barrier (BBB) is a highly selective, neuroprotective membrane which severely impedes brain, drug delivery to the brain. drug Conventional methods of drug delivery to the including intraventricular diffusion and intracerebral implants, have been highly invasive. Preliminary results, however, have shown that surface-modified nanoparticles may be able to carry drugs across the BBB.

Nanoparticles are also being considered as delivery vehicles for DNA and ribonucleic (RNA) in gene therapy applications. Gene delivery of DNA and RNA to target cells in order to manipulate protein expression has the potential to combat a number of diseases including cystic fibrosis, hemophilia, cancer and AIDS. Drug nanoparticles, due to their unique properties, have been studied for a number of delivery strategies. Their ability to permeate biological membranes, accessibility to remote tissues, and increased residence time in the body offer a more diverse portfolio of treatment options and improve drug efficacy through both local and systemic targeting.

The Big Impact of Nanoparticles


As with any new technology, there still exist quite a few unknowns concerning the use of nanoparticles for drug delivery. In order for drug nanoparticles to become a universally used treatment option, their benefits must significantly outweigh any possible side effects. Due to nanoparticles penetration ability, their accumulation and persistence in the environment could be concerning. Environmental exposure to nanoparticles could cause adverse effects in biological and

ISBN 978-607-8164-02-8

724

[Ide@s CONCYTEG 6(72): Junio, 2011]

ecological nanoparticles

systems. and

The the

diversity many

of

is likely to become an integral part of future drug delivery strategies.

possible

administration routes make regulating this technology a challenge. Due to the significant research and development costs associated with drug nanoparticles and ensuring their safety, nanoparticle treatment options are likely to be expensive, especially at their advent. Ensuring equal access to these technologies for those who need them most will be an additional regulatory challenge. Despite a lack of long-term safety data and a number of regulatory challenges, nanoparticle technology has the potential to revolutionize modern drug delivery. of the A fundamental of understanding properties
Bala, I., S. Hariharan and M. N. Kumar (2004), "PLGA nanoparticles in drug delivery: the state of the art.", Critical reviews in therapeutic drug carrier systems 21, p. 387. Desai, M. P.,V. Labhasetwar, G. L. Amidon and R. J. Levy (1996), "Gastrointestinal uptake of biodegradable microparticles: effect of particle size", Pharmaceutical research 13, pp. 1838-1845. Dressman, J. B., G. L. Amidon, C. Reppas and V. P. Shah (1998), "Dissolution testing as a prognostic tool for oral drug absorption: immediate release dosage forms", Pharmaceutical Research 15, pp. 11-22. Gupta, R. B. and U. B. Kompella, eds. (2006), Nanoparticle Technology for Drug Delivery, Vol. 159. New York: Taylor and Francis. Gupta, R. B. and J. -J. Shim (2007), Solubility in Supercritical Carbon Dioxide, Boca Raton: Francis and Taylor. Lipinski, C. A. (2001), "Avoiding investment in doomed drugs, is poor solubility an industry wide problem?", Curr. Drug Dis., pp. 17-19. Lipinski, C. A. (2002), "Poor aqueous solubility-an industry wide problem in drug discovery", Am. Pharm. Rev. 5, pp. 82-85. Liversidge, G. G., K. C. Cundy, J. F. Bishop and D. A. Czekai (1992), "Surface modified drug nanoparticles", Google Patents.

References

nanoparticles show a number of unique properties such as increased surface area, a greater percentage of surface molecules, ease of suspension, and size-dependent magnetic and optical properties. have been A number of for technologies developed

producing nanoparticles of virtually any type of drug. Nanoparticle characterization for drug delivery to uses traditional analysis are techniques relevant determine physiologically

parameters.

Nanoparticles

amenable to a number of administration routes, which allows them to be delivered both locally and systemically to biological systems. As research continues and more nanoparticle drugs advance to clinical testing and commercial use, nanoparticle technology

Merisko-Liversidge, E., G. G.Liversidge and E. R. Cooper (2003), "Nanosizing: a formulation approach for poorly-water-soluble compounds", European journal of pharmaceutical sciences 18, 113-120.

ISBN 978-607-8164-02-8

725

Nanoparticle Technology for Drug Delivery Courtney A. Ober y Ram B. Gupta Merisko-Liversidge, E., P. Sarpotdar, J. Bruno, S. Hajj, L. Wei, N. Peltier, J. Rake, J.. M. Shaw, S. Pugh and L. Polin (1996), "Formulation and antitumor activity evaluation of nanocrystalline suspensions of poorly soluble anticancer drugs", Pharmaceutical research 13, 272-278. Moghimi, S. M. and J. Szebeni (2003), "Stealth liposomes and long circulating nanoparticles: critical issues in pharmacokinetics, opsonization and protein-binding properties", Progress in lipid research 42, pp. 463-478. O'Donnell, P. B. and J. W. McGinity (1997), "Preparation of microspheres by the solvent

evaporation technique", Advanced Delivery Reviews 28, PP. 25-42.

Drug

Reverchon, E. and R. Adami (2006), "Nanomaterials and supercritical fluids", J. of Supercritical Fluids 37, 1-22. Wu, N. Z., D. Da, T. L. Rudoll, D. Needham, A. R. Whorton and M. W. Dewhirst (1993), "Increased microvascular permeability contributes to preferential accumulation of Stealth liposomes in tumor tissue", Cancer research 53, pp. 3765.

ISBN 978-607-8164-02-8

726

Das könnte Ihnen auch gefallen