p_lecture_1236694477

Upload: nileshruke

Post on 04-Feb-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/21/2019 p_lecture_1236694477

    1/20

    Journal of Controlled Release 70 (2001) 120

    www.elsevier.com/locate/jconrel

    Review

    Biodegradable polymeric nanoparticles as drug delivery devicesa a , a*Kumaresh S. Soppimath , Tejraj M. Aminabhavi , Anandrao R. Kulkarni ,

    bWalter E. Rudzinski

    aDepartment of Chemistry, Polymer Research Group, Karnatak University, Dharwad 580 003, India

    bDepartment of Chemistry, Southwest Texas State University, San Marcos, TX 78666, USA

    Received 12 June 2000; accepted 28 September 2000

    Abstract

    This review presents the most outstanding contributions in the field of biodegradable polymeric nanoparticles used as drug

    delivery systems. Methods of preparation, drug loading and drug release are covered. The most important findings on surface

    modification methods as well as surface characterization are covered from 1990 through mid-2000. 2001 Elsevier

    Science B.V. All rights reserved.

    Keywords: Nanoparticle; Method of preparation; Surface modification; Drug delivery; Drug targeting

    1. Introduction and reduce the toxicity or side effects [2]. However,

    developmental work on liposomes has been limited

    Over the past few decades, there has been consid- due to inherent problems such as low encapsulation

    erable interest in developing biodegradable efficiency, rapid leakage of water-soluble drug in the

    nanoparticles (NPs) as effective drug delivery de- presence of blood components and poor storage

    vices. Various polymers have been used in drug stability. On the other hand, polymeric NPs offer

    delivery research as they can effectively deliver the some specific advantages over liposomes. For in-

    drug to a target site and thus increase the therapeutic stance, NPs help to increase the stability of drugs /

    benefit, while minimizing side effects [1]. The proteins and possess useful CR properties.

    controlled release (CR) of pharmacologically active Nanoparticles generally vary in size from 10 to

    agents to the specific site of action at the therapeu- 1000 nm. The drug is dissolved, entrapped, encapsu-tically optimal rate and dose regimen has been a lated or attached to a NP matrix and depending upon

    major goal in designing such devices. Liposomes the method of preparation, nanoparticles, nanos-

    have been used as potential drug carriers instead of pheres or nanocapsules can be obtained. Nanocap-

    conventional dosage forms because of their unique sules are vesicular systems in which the drug is

    advantages which include ability to protect drugs confined to a cavity surrounded by a unique polymer

    from degradation, target the drug to the site of action membrane, while nanospheres are matrix systems in

    which the drug is physically and uniformly dis-

    persed. In recent years, biodegradable polymeric NPs*Corresponding author. Fax: 191-836-747-884.E-mail address: [email protected] (T.M. Aminabhavi). have attracted considerable attention as potential

    0168-3659/ 01/ $ see front matter 2001 Elsevier Science B.V. All rights reserved.

    P I I : S 0 1 6 8 - 3 65 9 ( 0 0 ) 0 0 3 3 9 - 4

  • 7/21/2019 p_lecture_1236694477

    2/20

    2 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    drug delivery devices in view of their applications in 2.1.1. Solvent evaporation method

    the CR of drugs, their ability to target particular In this method, the polymer is dissolved in an

    organs/ tissues, as carriers of DNA in gene therapy, organic solvent like dichloromethane, chloroform or

    and in their ability to deliver proteins, peptides and ethyl acetate. The drug is dissolved or dispersed into

    genes through a peroral route of administration [3,4]. the preformed polymer solution, and this mixture isSome general aspects on micro- and nanoparticles then emulsified into an aqueous solution to make an

    have been reviewed earlier [1,511]. A majority of oil (O) in water (W) i.e., O/ W emulsion by using a

    these reviews have dealt with the NPs of poly(D,L- surfactant/ emulsifying agent like gelatin, poly(vinyl

    lactide), poly(lactic acid) PLA, poly(D,L-glycolide) alcohol), polysorbate-80, poloxamer-188, etc. After

    PLG, poly(lactide-co-glycolide), PLGA, and poly- the formation of a stable emulsion, the organic

    (cyanoacrylate) PCA. The present review details the solvent is evaporated either by increasing the tem-

    latest developments on the above mentioned poly- perature/ under pressure or by continuous stirring.

    mers as well as NPs based on chitosan, gelatin, The effect of process variables on the properties of

    sodium alginate and other hydrophilic / biodegradable NPs was discussed earlier [26]. The W/O / W method

    polymers. Surface modification aspects are also has also been used to prepare the water-soluble

    covered in more detail. The PLA, PLG and PLGA drug-loaded NPs [27]. Both the above methods use apolymers being tissue-compatible have been used high-speed homogenization or sonication. However,

    earlier as CR formulations in parentral and implanta- these procedures are good for a laboratory-scale

    tion drug delivery applications [1214]. In addition, operation, but for a large-scale pilot production,

    poly(e-caprolactone), PCL, which was first reported alternative methods using low-energy emulsification

    by Pitt et al. [15,16] for the CR of steroids and are required. In this pursuit, following approaches

    narcotic antagonists as well as to deliver opthalmic have been attempted.

    drugs [17], and poly(alkylcyanoacrylate), PACA, are

    now being developed as NPs. In addition, less 2.1.2. Spontaneous emulsification/solvent diffusion

    frequently used polymers like poly(methylidene method

    malonate) [18], gelatin [19], chitosan [20] and so- In a modified version of the solvent evaporation

    dium alginate [21] will also be included in this method [2830] the water-soluble solvent like ace-

    review. The important published literature within the tone or methanol along with the water insolubleperiod 19902000 is critically reviewed. The review organic solvent like dichloromethane or chloroform

    does not cover the entire literature within this period, were used as an oil phase. Due to the spontaneous

    but the reader is advised to go to the original diffusion of water-soluble solvent (acetone or metha-

    literature in order to get more details. nol), an interfacial turbulence is created between two

    phases leading to the formation of smaller particles.

    As the concentration of water-soluble solvent (ace-

    tone) increases, a considerable decrease in particle

    2. Preparation of nanoparticles size can be achieved.

    Conventionally, NPs have been prepared mainly 2.1.3. Salting out/emulsificationdiffusion method

    by two methods: (i) dispersion of the preformed The methods discussed above require the use ofpolymers; and (ii) polymerization of monomers. organic solvents, which are hazardous to the environ-

    ment as well as to the physiological system [31]. The

    US FDA has specified the residual amount of organic2.1. Dispersion of preformed polymers solvents in injectable colloidal systems [32,33]. In

    order to meet these requirements, Allemann and

    Several methods have been suggested to prepare co-workers have developed two methods of prepar-

    biodegradable NPs from PLA, PLG, PLGA and ing NPs. The first one is a salting-out method [34,35]

    poly(e-caprolactone) by dispersing the preformed while the second one is the emulsificationsolvent

    polymers [2225]. diffusion technique [36,37].

  • 7/21/2019 p_lecture_1236694477

    3/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 3

    2.1.4. Production of NPs using supercritical fluid [44], the solution is charged with the supercritical

    technology fluid in the precipitation vessel containing solute of

    Production of NPs with the desired physicochemi- interest in an organic solvent. At high pressures,

    cal properties to facilitate the targeted drug delivery enough anti-solvent will enter into the liquid phase

    has been a topic of renewed interest in pharma- so that the solvent power will be lowered and theceutical industries. Conventional methods like sol- solute precipitates. After precipitation, when the final

    vent evaporation, coacervation and in situ polymeri- operating pressure is reached, the anti-solvent flows

    zation often require the use of toxic solvents and / or through the vessel so as to strip the residual solvent.

    surfactants. Therefore, research efforts have been When the solvent content has been reduced to the

    directed to develop the environmentally safer en- desired level, the vessel is depressurized and the

    capsulation methods to produce the drug-loaded solid product is collected. A schematic of the SAS

    micron and submicron size particles. If solvent method is shown in Fig. 1. In a modified version of

    impurities remain in the drug-loaded NPs, then these the SAS technique [39], the solid of interest is first

    become toxic and may degrade the pharmaceuticals dissolved in a suitable solvent and then this solution

    within the polymer matrix. Supercritical fluids have is rapidly introduced into the supercritical fluid

    now became the attractive alternatives because these through a narrow nozzle. The supercritical fluidare environmentally friendly solvents and the method completely extracts the solvent, causing the super-

    can be profitably used to process particles in high critical fluid insoluble solid to precipitate as fine

    purity and without any trace amount of the organic particles. This method, also called as gas anti-solvent

    solvent. Literature on the production of drug-loaded (GAS) technique, has been successfully used to

    microparticles using supercritical fluids is enormous produce microparticles as well as NPs.

    [3844]. However, comparatively much less have

    been investigated to produce NPs [39,40]. It is 2.1.5. Polymerization methods

    beyond the scope of the present review to give an Nanoparticles can also be prepared by polymeri-

    entire coverage on supercritical fluid technology; we zation of monomers. Poly(alkylcyanoacrylate)s,

    will discuss only two of the most commonly used PACA, being biodegradable, have been used as tissue

    methods of producing micro- or nanoparticles. adhesives in surgery since these are well tolerated in

    In the rapid expansion of supercritical solution vivo [45,46]. This has prompted intense research(RESS) method the solute of interest is solubilized in activity to study polymerization reactions. Couvreur

    a supercritical fluid and the solution is expanded et al. [47,48] reported the production of NPs (|200

    through a nozzle. Thus, the solvent power of super- nm diameter) by polymerizing mechanically the

    critical fluid dramatically decreases and the solute

    eventually precipitates. This technique is clean be-

    cause the precipitated solute is completely solvent-

    free. Unfortunately, most polymers exhibit little or

    no solubility in supercritical fluids, thus making the

    technique less of practical interest. RESS was very

    popular in the late 80s and early 90s for particle

    production of bioerodible drug-loaded polymers likePLA. A uniform distribution of drug inside the

    polymer matrix can be achieved by this method for

    low molecular mass (,10 000) polymers. However,

    the RESS method cannot be used for high molecular

    mass polymers due to their limited solubility in

    supercritical fluids. For these reasons, much less

    information is found in the literature over the pastFig. 1. Schematic diagram of the SAS method: PV1 and PV2 are

    67 years on this technique [41,43]. two volumetric pumps, N is nozzle, P is precipitation vessel, MVIn the supercritical anti-solvent (SAS) method is micrometric valve and EV is expansion vessel.

  • 7/21/2019 p_lecture_1236694477

    4/20

    4 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    dispersed methyl or ethyl cyanoacrylate in aqueous

    acidic medium in the presence of polysorbate-20 as a

    surfactant without irradiation or an initiator. Here,

    the cyanoacrylic monomer is added to an aqueous

    solution of a surface-active agent (polymerizationmedium) under vigorous mechanical stirring to poly-

    merize alkylcyanoacrylate at ambient temperature.

    Drug is dissolved in the polymerization medium

    either before the addition of the monomer or at the

    end of the polymerization reaction. The NP suspen-

    sion is then purified by ultracentrifugation or by

    resuspending the particles in an isotonic surfactant-

    free medium. The mechanism of polymerization of

    PACA monomer is given below.

    Polymerization follows the anionic mechanism, Fig. 2. Schematic representation for the production of poly-(alkylcyanoacrylate) nanoparticles by anion polymerization.since it is initiated in the presence of nucleophilic

    2 2 2initiators like OH , CH O and CH COO leading3 3to the formation of NPs of low molecular mass due [49], but NP production is not possible above a pH

    to rapid polymerization. Such NPs are degraded very of 3.0, probably due to the aggregation and stepwise

    fast. In order to circumvent this problem and to molecular mass increase at lower pH. Other factors

    produce higher molecular mass as well as stable that influence the formation of NPs include the

    NPs, polymerization must be carried out in an acidic concentration of monomer and the speed of stirring.

    medium (pH 1.03.5). After dispersing the monomer The NPs of PACA have gained wide popularity in

    in an aqueous acidic medium containing surfactant recent years despite some major drawbacks such as

    and stabilizer, polymerization is continued for 34 h use of low pH (around 2) and cytotoxicity [50]. This

    by increasing the pH of the medium to obtain the has lead to the synthesis of new dialkyl-methylidene

    desired products. malonic acid ester monomers [51] and the NPs of

    During polymerization, various stabilizers like poly(methylidenemalonate), PDEMM were prepared,dextran-70, dextran-40, dextran-10, poloxamer-188, and these were found to be non-biodegradable both

    -184, -237, etc are added. In addition, some surfac- in vitro and in vivo [52,53]. To overcome this

    tants like polysorbate-20, -40 or -80 are also used. problem, new derivatives of PDEMM were prepared

    Particle size and molecular mass of NPs depend i.e., ethyl-2-ethoxycarbonylmethylenoxycarbonyl

    upon the type and concentration of the stabilizer acrylate. NPs from these monomers were prepared

    and/ or surfactant used. A schematic representation by the same methods as those adopted for the

    for the production of poly(alkylcyanoacrylate) NPs is preparation of PACA NPs by anionic polymerization

    shown in Fig. 2. The size and molecular mass of NPs [54]. The pH of the polymerization medium critically

    depend upon the pH of the polymerization medium influenced the physicochemical properties of NPs,

  • 7/21/2019 p_lecture_1236694477

    5/20

  • 7/21/2019 p_lecture_1236694477

    6/20

    6 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    3. Drug loading when compared to chromatographic methods, which

    require ultracentrifugation.

    A successful NP system may be the one, which In addition to adsorption and incorporation, a new

    has a high loading capacity to reduce the quantity of method of drug loading for the water-soluble drugs

    the carrier required for administration. Drug loading was proposed by Yoo et al. [85]. In this method, druginto the NPs is achieved by two methods: one, by was chemically conjugated into NPs. The conjugated

    incorporating the drug at the time of NP production doxorubicinPLGA and doxorubicin-loaded PLGA

    or secondly, by adsorbing the drug after the forma- NPs were prepared by the spontaneous emulsion

    tion of NPs by incubating them in the drug solution. solvent diffusion method. The encapsulation ef-

    It is thus evident that a large amount of drug can be ficiency of 96.6% and 3.5% loading of doxorubicin

    entrapped by the incorporation method when com- PLGA conjugate have been achieved. For the un-

    pared to the adsorption [76,77]. Adsorption iso- conjugated doxorubicin, these values were, respec-

    therms for the NP/ drug delivery system give vital tively 6.7% and 0.3% (w/ w).

    information on the best possible formulation, the

    drug binding capacity onto the surface of NPs and

    the amount of drug adsorbed. For instance, Couvreur 4. Drug release

    et al. [78] reported the adsorption of two antineoplas-

    tic drugs viz, dactinimycin and methotrexate onto Drug release from NPs and subsequent biodegra-

    poly(methylcyanoacrylate) and poly- dation are important for developing the successful

    (ethylcyanoacrylate). It was observed that methotrex- formulations. The release rates of NPs depend upon:

    ate was bound to the NPs to a lesser extent than (i) desorption of the surface-bound/ adsorbed drug;

    dactinimycin. Generally, in the case of PACA, it is (ii) diffusion through the NP matrix; (iii) diffusion

    observed that longer the alkyl chain length higher the (in case of nanocapsules) through the polymer wall;

    affinity for the drugs. The capacity of adsorption is (iv) NP matrix erosion; and (v) a combined erosion/

    thus related to the hydrophobicity of the polymer and diffusion process. Thus, diffusion and biodegradation

    the specific area of the NPs. In case of entrapment govern the process of drug release.

    method, an increase in concentration of the mono- Methods to study the in vitro release are: (i)

    mer, increases the association of drug, but a reverse side-by-side diffusion cells with artificial or bio-trend is observed with the drug concentration in the logical membranes; (ii) dialysis bag diffusion tech-

    dispersed solution. This observation was further nique; (iii) reverse dialysis sac technique; (iv) ultra-

    substantiated by Radwan [79] who studied the effect centrifugation; (v) ultrafiltration; or (vi) centrifugal

    of monomer concentration on % drug loading. These ultrafiltration technique. Despite the continuous ef-

    results indicate that there is a need to optimize the forts in this direction, there are still some technical

    amount of monomer available for the drug entrap- difficulties to study in vitro drug release from NPs

    ment. [86,87]. These are attributed to the separation of NPs

    The type of surface-active materials and stabilizers from the release media. In order to separate NPs and

    has an effect on drug loading [80]. Chukwu et al. to avoid the tedious and time-consuming separation

    [81] studied the adsorption of different psycho- techniques, dialysis has been used; here, the suspen-

    pharmacological agents onto NPs of poly- sion of NPs is added to the dialysis bags/tubes of(isobutylcyanoacrylate), PIBCA, in the pH range different molecular mass cut-off. These bags are then

    between 2.0 and 7.4. Adsorption of drugs onto NPs incubated in the dissolution medium [8890].

    followed the Langmuir mechanism [82,83]. In Another technique involves the use of a diffusion

    another study [84], a dielectric method was used to cell consisting of donor and acceptor compartments;

    investigate the adsorption ofb-blockers onto PIBCA this technique was used to separate through the

    NPs. In this method, the NP suspensions were taken artificial/ biological membranes [91]. In this method,

    into a capacitance cell, exposed to a high-frequency kinetic study was not performed under the perfect

    field (10 MHz) and the complex impedance was sink conditions, because the NPs were not directly

    measured. This technique is rapid and inexpensive diluted in the release media, but were separated from

  • 7/21/2019 p_lecture_1236694477

    7/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 7

    the release media through the membrane. Thus, the drug from the core across the polymeric barrier

    amount of drug in the release media did not reflect layer. Hence, theoretically, the drug release should

    the real amount released. In order to avoid the follow the zero-order kinetics. Calvo et al. [17]

    enclosure of NPs in the dialysis bag, Leavy and obtained almost the similar release profiles for

    Benita [92] used a reverse dialysis technique for the indomethacin from both NPs and nanocapsules. ThisO / W emulsion. In this method, NPs were added indicated that the polymer coating does not show any

    directly into the dissolution medium. The same barrier properties for the drug release. The drug

    technique was adopted by Calvo et al. [17] for the release from the nanocapsule takes place mainly by

    evaluation of NPs, nanocapsules and nanoemulsions. the partitioning of the drug; however, the main factor

    However, the method is not very sensitive for controlling the release is the volume of the aqueous

    studying the rapid release formulations; but can only medium. For instance, with higher dilution of the

    be used for the release of formulations having the dissolution media, a faster and complete release of

    release time longer than 1 h [93]. the drug takes place. However, Lu et al. [97]

    Release profiles of the drugs from NPs depend reported that the release of bovine serum albumin

    upon the nature of the delivery system. In the case of from PLA nanocapsule depends upon the molecular

    a matrix device, drug is uniformly distributed/ dis- mass of the polymer, which indicates that the releasesolved in the matrix and the release occurs by may not occur by partitioning of the drug, but may

    diffusion or erosion of the matrix. If the diffusion of be due to diffusion across the polymer coating.

    the drug is faster than matrix degradation, then the The method of drug incorporation into NPs has

    mechanism of drug release occurs mainly by diffu- also shown an effect on drug release. Fresta et al.

    sion, otherwise it depends upon degradation [28]. [90] reported a higher burst up to 6070% for the

    Rapid initial release is attributed to the fraction of NPs loaded with drug by adsorption; here, the burst

    the drug which is adsorbed or weakly bound to the effect is less and the remaining drug release is quite

    large surface area of the NPs, than to the drug slow. This study demonstrated that the incorporation

    incorporated in NPs. Following the dilution of the method has shown better sustained release charac-

    dissolution media under perfect sink conditions the teristics. When the drug is chemically conjugated

    drug partition showed an increase due to the immedi- with PLGA NPs, the release took place over 25 days,

    ate release phase. Later, an exponential delayed whereas with those NPs containing unconjugatedrelease rate is observed probably due to the drug free drug, a rapid release in about 5 days occurred

    diffusion from the matrix [94,95,35,17,28]. Release [85]. Here, the CR properties have been attributed to

    in the matrix type of NPs follows the first-order chemical degradation of the conjugated PLGA,

    kinetics [90,79]. which permitted water solubilization and subsequent

    Recently, Polakovic et al. [96] theoretically release of the drug-conjugated PLGA oligomers into

    studied the release of PLA NPs loaded with varying the medium. In case of drug release from hydrogel

    amounts (732% w / w) of lidocane. Two models NPs, release occurs mainly due to swelling, which

    were used to study the drug release: (i) by crystal can be controlled by either adding the hydrophilic

    dissolution and (ii) by diffusion through the polymer functional groups or by monitoring cross-linking of

    matrix. When the drug loading is ,10% (w/ w) (the the matrix.

    drug is molecularly dispersed), the release kineticsshows a better fit to the diffusion model. The

    existence of lidocane crystals at higher concentration 5. Surface properties of NPs

    (.10%) is observed. Since the drug should dissolve

    first from the crystals and then diffuse from the 5.1. Protein adsorption and phagocytosis of NPs

    matrix, the overall release mechanism could be

    described by the dissolution model. Plasma protein adsorption and phagocytosis of

    In the case of nanocapsules (reservoir-type drug- NPs is a subject that has been widely studied in

    delivery systems) the drug core is coated with the recent years. When the NPs are administered in-

    polymer and the release occurs by diffusion of the travenously they are easily recognized by the body

  • 7/21/2019 p_lecture_1236694477

    8/20

    8 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    immune systems, which are then cleared from the of poly(oxyethylene) in the polymer has drastically

    circulation. Apart from the size of NPs, their surface decreased the protein adsorption when compared to

    hydrophobicity determines the amount of adsorbed the pure polyesters.

    blood components, mainly proteins (opsonins). These In another study by the same group of researchers

    will determine the in-vivo fate of NPs [98,99]. [105], an attempt was made to correlate the ad-Binding of these opsonins onto the surface of NPs, sorption results with the in-vivo circulation of NPs.

    called opsonization, acts as a bridge between NPs The di-block and multi-block copolymers of PEG

    and phagocytes. Hence, for a qualitative and quan- were used as model polymers to show the decrease

    titative understanding of the interaction of blood in adsorption of proteins; these NPs have shown

    proteins with NPs, it is necessary to design long- long-circulating properties. The reduced liver uptake

    circulating NPs by surface modification. of NPs was dependent on the molecular mass and

    In a study by Allemann et al. [100], it was surface density of PEG. The in-vitro protein rejection

    reported that when the PLA NPs are incubated in properties of the PEG-coated poly-

    human plasma and serum, the IgG was found to be (alkylcyanoacrylate) NPs were investigated after

    the major protein along with albumin, apolipopro- when the freeze fracture of NPs were pre-incubated

    tein-E, which were adsorbed on the surface. Compli- with fibrinogen as model blood protein [106]. Thement C components (part of immune system used decrease in protein adsorption onto PEG-coated NPs3for the recognition of foreign surfaces) were also was evident by 2-DPAGE after incubating them in

    adsorbed onto the surface of NPs after incubation in human serum. The NPs were also long-circulating as

    the serum reaching the level of antibody IgG. Blunk proved from in-vivo tests.

    et al. [101] studied the kinetics of protein adsorption

    onto polystyrene NPs and confirmed that albumin 5.2. Surface characterization methods

    and fibrinogen were adsorbed in a highly diluted

    plasma (0.08 and 0.8%). However, in the plasma of Many techniques have been developed and used to

    high concentration (80%), proteins were displaced study the surface modification of NPs. The efficiency

    within seconds or even fractions of a second. The of surface modification can be measured either by

    study indicated that apolipoproteins A-I, C-III, E and estimating the surface charge, density of the func-

    J were the major proteins adsorbed onto NPs. tional groups or an increase in surface hydrophilicity.A two-dimensional polyacrylamide gel electropho- One method used to measure the surface modi-

    resis (2-DPAGE) was used to estimate quantitatively fication is to determine zeta potential (j) of the

    the interaction of plasma proteins with iron oxide aqueous suspension containing NPs. In this method,

    NPs in the presence of plasma proteins stabilized by the mobility of charged particles is monitored by

    polysaccharide. Particles incubated in the plasma applying an electrical potential. The zeta potential

    were separated and were then washed with different values may be positive or negative depending upon

    washing media. The protein adsorbed on NPs was the nature of the polymer or the material used for

    then estimated by 2-DPAGE. By this, it was found surface modification. The extent of surface hydro-

    that fibrinogen, IgG and albumin were the major philicity can then be predicted from the values ofj.

    plasma proteins adsorbed onto NP surface [102,103]. This is a widely used technique to understand the

    In another study by Luck et al. [104], the interaction surface charges of NPs.of proteins with NPs was shown to depend upon the Another commonly used technique is electron

    method of NP preparation. For example, the amount spectroscopy for chemical analysis, ESCA, also

    of several apolipoproteins in plasma protein adsorp- called X-ray photoelectron spectroscopy (XPS). This

    tion patterns of the spray-dried PLGA and PLA NPs technique is based on the emission of electrons from

    were distinctly higher than when compared to the materials, in response to irradiation by photons of

    adsorption patterns of the particles produced by W/ sufficient energy, to cause ionization of the core-

    O / W emulsion technique. Some adsorbed proteins level electrons. These electrons are emitted at ener-

    were found to be specific for particles produced by gies characteristic of the atoms from which they are

    the same method. The presence of hydrophilic chain emitted. Since photons have low penetration energy,

  • 7/21/2019 p_lecture_1236694477

    9/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 9

    only those electrons pertaining to atoms at or near action with the surface even at lower concentrationsthe surface (up to 100 A) escape and these can be of the protecting polymeric layer. The biological

    counted. For each atom type, the number of electrons consequences of steric protection of drug carriers

    emitted is related to the number of atoms of a with the surface-grafted polymers have been dis-

    particular type of atom. Using this technique, surface cussed and clinical applications of the long-circulat-elemental analysis was performed [107]. ing NPs have been studied [10]. A theoretical model

    In another technique, the surface hydrophobicity of repulsion of proteins from the solid substrate was

    of NPs can be directly measured by hydrophobic proposed by Joen et al. [110]. The steric repulsion,

    interaction chromatography. This technique involves van der Waals attractions and hydrophobic inter-

    the column chromatography, which is able to sepa- action free energy have been correlated. The model

    rate materials based on the interaction with a hydro- provides a basis for the prevention of opsonin

    phobic gel matrix [108]. The nanoparticle and the gel deposition. High surface density and long chain-

    interaction is a function of surface hydrophobicity of lengths of PEG are necessary for low protein ad-

    NPs. Propyl agarose gel is used as a stationary phase sorption. However, surface density has a greater

    and elution of NPs can be achieved by using the effect than the chain-length on steric repulsion and

    phosphate buffer. Eluent samples can be collected van der Waals attraction.and the optical density measured spectrophotomet- Bazile et al. [111] developed the NPs based on

    rically at 400 nm. The gel matrix can then be washed methoxy PEGPLA i.e., Me-PEGPLA and blends

    to remove the NPs. of PLA with Me-PEGPLA. These NPs, labeled by14

    introducing C-labeled PLA in the formulation were5.3. Methods of surface modification more slowly captured by the cultured THP-1 mono-

    cytes when compared to pluronic F68-coated PLA

    Surface modification of biodegradable and long- NPs. The half-life of Me-PEGPLA NPs was im-

    circulating polymeric NPs has been achieved mainly proved by a factor of 180 (360 min) when compared

    by two methods: (i) surface coating with hydrophilic to the uncoated and F68-coated NPs. Even though, a

    polymers/ surfactants; and (ii) development of bio- high amount of radioactivity was located in the heart

    degradable copolymers with hydrophilic segments. and blood vessels due to particle circulation, in other

    Some of the widely used surface-coating materials phagocytic organs, radioactivity was found evenare: polyethylene glycol (PEG), polyethylene oxide after 6 h of i.v. administration indicating a delay in

    (PEO), poloxamer, poloxamine, polysorbate (Tween- phagocytosis. Tobio et al. [112] observed much

    80) and lauryl ethers (Brij-35). greater penetration of tetanus toxoid (TT) encapsu-

    lated PEGPLA NPs than PLA NPs after nasal5.3.1. PEG and PEO-coated NPs administration. A high persisting radioactivity was

    PEG-coated NPs have received a lot of attention. found in body compartments up to 8 h after the125

    Gref et al. [109] described the one step method to introduction of I TT-loaded NPs.

    prepare the PEG-coated NPs using amphiphilic Gref et al. [113] reported the preparation of blend

    PEGpolyester diblock copolymer as the starting NPs of PLA with monomethoxy polyoxyethylene

    material and showed that the protective coating (MPOE) by solvent evaporation method using so-

    affecting against the phagocytes depends upon den- dium cholate surfactant. The zeta potentials mea-sity and molecular mass of PEG. They also studied sured at various concentrations of NaCl varied from

    the biodistribution of covalently-attached PEG 255 mV for PLA to 0 mV for blends depending

    PLGA NPs. The protective effect of PEG on carriers upon the composition of MPOE in the NPs. The zeta

    like liposomes, NPs and micelles was studied by potential increased with an increasing amount of

    Torchilin [11] in terms of the statistical behavior of MPOE suggesting that the MPOE chains that are

    polymers. A mechanism was proposed which as- present on the surface of NPs mask the ionized2

    sumes that the surface-grafted chains of flexible and COO end-group of PLA. These results are sup-

    hydrophilic polymers form dense conformational ported by a phagocytosis study on the monocytes.

    clouds thus preventing other polymers from inter- When MPOE content in the blend is greater than

  • 7/21/2019 p_lecture_1236694477

    10/20

    10 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    23%, the MPOE chain adopts a brush-like configu- the PEG coated NPs. Peracchia et al. [121] also

    ration forming a sterically-uncharged barrier, thereby prepared the methoxy PEG cyanoacrylatehexadecyl

    reducing the zeta potential and phagocytosis. cyanoacrylate amphiphilic NPs by polymer precipi-

    Govender et al. [114] examined the drug-encapsu- tation or by solvent evaporation method; the PEG

    lation characteristics of PLAPLA NPs using coating was confirmed by XPS. The particles ex-procaine hydrochloride, a water-soluble drug. The hibited a reduced cytotoxicity and enhanced degra-

    PLAPEG NPs were produced by the nano-precipi- dation. NPs prepared in the presence of PEGs have

    tation technique. The drug-entrapment efficiencies of shown some advantages in preventing opsonization

    these NPs were compared with those of PLGA NPs. and thereby avoiding the MPS uptake [122]. This

    Kim et al. [115] used the ESCA method to evaluate mechanism is explained in Fig. 3.

    the presence of PEG on the surface of indomethacin- PEO-surface modified systems have received an

    loaded Me-PEGPLA NPs. The in-vitro cytotoxicity increased attention in recent years. Jaeghere et al.

    of these NPs did not show any remarkable cytotox- [123] studied the freeze-dried PEO-surface modified

    icity against the normal human fibroblast cells. NPs as a function of PEO chain length and surface

    The optimum surface density of PEG on NPs density to avoid the MPS uptake. NPs were produced

    plays an important role in steric repulsion. These by salting-out method using the blends of PLA andNPs have shown a lower accumulation in the liver, PLAPEO copolymers. In an effort to study the

    but the observed high spleen uptake is due to the effect of surface density of PEO on the compliment

    removal of PEG coating from the surface of NPs, an consumption, Vittaz et al. [124] used the diblock

    important property in spleen targeting [116]. In polymer of PLA and polyethylene oxide (PLA

    addition, the distance between PEG chains on the PEO). It was found that as the PEO density on the

    surface of NPs is critical to avoid the adsorption of surface of the NPs increases, a decrease in compli-

    plasma proteins. For instance, a decrease in the ment consumption is observed due to steric repulsion

    distance between PEG chains on the surface from 6.2 of the surface to proteins. A preliminary study was

    to 5.1 nm drastically decreases the adsorption of made on the synthesis of amphiphilic PEOPPO

    apolipoproteins up to 90%. This further confirms that

    the density of hydrophilic segment on the surface of

    NPs is important in opsonization. However, anyfurther decrease in this distance did not show

    significant effects on the adsorption of plasma pro-

    teins [117].

    Peracchia et al. [118] used the emulsification and

    solvent evaporation method to prepare the diblock

    Me-PEGPLA copolymeric NPs containing 20 and

    33% of lidocaine. They confirmed high-density of

    the surface PEG by ESCA. However, the size of NPs

    produced by the block copolymer was twice as high

    as those of PLGA NPs. This was attributed to an

    increase in the chain length of PEG. Peracchia et al.[119,120] reported the chemical coupling of PEG

    with PBCA NPs prepared by emulsion polymeri-

    zation in the presence of PEG, Me-PEG and (Me) -2PEG. Polymerization was possible only in the pres-

    ence of PEG and Me-PEG as hydroxyl group wasFig. 3. Effect of surface PEG density and its conformation on thenecessary for polymerization and association of PEGopsonization process: (A) opsonization takes place when the

    on the surface of NPs. Higher PEG density wasdensity is low, (B) opsonization is not possible at higher surface

    observed on the surface of NPs when Me-PEG was density and (C) when both the end groups of PEG participate inused. A decrease in hydrophobicity was observed for surface modification.

  • 7/21/2019 p_lecture_1236694477

    11/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 11

    PEO block copolymers (Plunoric) and poly(e-cap- uptake pathways are observed for NPs depending

    rolactone) by bulk polymerization [125]. The size of upon their surface characteristics and the rodent

    the NPs prepared varied from 116 to 196 nm, species. Coating was effective in stimulating the

    depending upon the type of copolymer used. spleen uptake in rats and mice. Spleen uptake of

    Fluoresbrite NPs was higher than Estapor NPs,5.3.2. Poloxamine and poloxamer coated NPs probably due to differences in the surface charac-

    Poloxamer and poloxamine have been widely used teristics of NPs.

    in surface coating studies. Storm et al. [126] pre- Polystyrene-latex nanospheres (PSL-NS, mean

    sented an overview of the advances made up to 1995 diameter, 85 nm) were coated with lactosyl-poly-

    on the surface modification of NPs to oppose the styrene (LPS, high affinity to hepatocytes) to evalu-

    MPS uptake. In a study by Illum and Davis ate their targeting characteristics to hepatocytes and

    [127,128], poloxamer and poloxamine were used as PSL-NS surfaces [133]. Hepatocytes were adhered

    the coating materials to prepare the long-circulating specifically with the LPS-coated dishes made of the

    NPs of polystyrene and poly(methyl methacrylate). A same materials as PSL-NS. Flow cytometry inves-

    prolonged circulation time and reduction in liver tigations showed that the LPS-coated fluorescein-

    uptake in rabbits was found for the poloxamine- isothiocyanate (FITC)PSL-NS were taken up bycoated polystyrene NPs (60 nm size) when compared hepatocytes when compared to the noncoated FITC

    to the uncoated NPs of the same size. A decrease in PSL-NS as a control. These findings indicated that

    hepatic uptake of about 20% for the NPs prepared LPSPSL-NS could target to hepatocytes. The sur-

    with poloxamer-188 and about 40% for the NPs face of LPS on PSL-NS showed higher hydrophil-

    coated with poloxamer-338 was observed. Rabbit icity than PEG-6000, Tween-80, poloxamer-407 and131

    experiments with I-labelled polystyrene poloxamer-908, which indicated that LPSPSL-NS

    poloxamer-407 coated NPs showed the superior may avoid the reticuloendothelial system capture and

    performance over that of poloxamer-338 to avoid the have a long plasma duration after the in-vivo i.v.

    hepatospleenic uptake. adminstration. Plasma coagulation can be prevented

    Rudt and Muller [129] studied the uptake of by the addition of 0.1% of PVA in LPSPSL-NS

    surface modified poloxamine-coated polystyrene NPs solution when LPSPSL-NS were injected. The

    and found extremely low levels of uptake for 100 nm LPSPSL-NS were the potential hepatocyte targetingsize NPs. Compared to poloxamer, poloxamine was carriers for the injectable formulations.

    more effective as a coating material to avoid the liver

    capture of rabbits [130]. Moghimi and Gray [131]

    developed the long-circulating poloxamine-908 5.3.3. Cyclodextrin/carbohydrate coated NPs

    coated polystyrene NPs (60 nm) that are resistant to Carbohydrates were also found to avoid the MPS

    MPS uptake. The time interval of administration was uptake when coated on the surface of NPs. To avoid

    important in maintaining the long circulation time. the MPS uptake the coated NPs with carbohydrate

    Spleen-capture study of the fluorescent-labeled was reported by Cho et al. [134]. The NPs of PLA

    polystyrene NPs coated with poloxamer 407 or and poly(L-lysine)-grafted-polysaccharide were also

    poloxamine 908 was made [132] on two rodent developed for the delivery of DNA [135] and these

    species viz., mouse and rat in order to assess the were found to be resistant against self-aggregationeffect of coating on their intraspleenic distribution. and nonspecific adsorption of the serum proteins.

    Two fluorescent polystyrene NPs used were: Recently, Duchene et al. [136] used amphiphilic

    Estapor (FX-010, 185 nm, Prolabo, France) and cyclodextrin NPs to increase the loading of water-

    Fluoresbrite (Plain YG, 260 nm, Polysciences, soluble drugs and bioavailability of the poorly water-

    UK). A fluorimetric investigation indicated that the soluble drugs intended for targeted delivery by the

    Fluoresbrite NPs were more efficiently trapped by oral or parenteral route. In order to further increase

    the spleen than the Estapor -based NPs in mice and the loading capacity, PIBCA NPs were loaded with

    rats. Results indicate an increase in the size of NPs the natural or hydroxypropyl cyclodextrins. The

    after coating the Estapor NPs. Different spleen loading capacity increased with an increase in stabili-

  • 7/21/2019 p_lecture_1236694477

    12/20

    12 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    ty constant of the inclusion drug/ parent g-cyclo- biological membrane was studied and an increase in

    dextrin and with a decrease in water solubility. the permeation of polysorbate-80 coated NPs

    The PIBCA NPs were prepared by the anionic occurred [91]. A schematic representation of the

    polymerization of isobutylcyanoacrylate in 0.01 M increased drug permeation from the polysorbate-80

    HCl containing 1% poloxamer-188 and in the pres- coated NPs through the biological membrane isence of cyclodextrins. The size, zeta potential and shown in Fig. 4.

    cyclodextrin content were influenced by the nature of Borchardt et al. [142] studied the uptake of

    cyclodextrin. The smallest size particles were ob- polysorbate-80 coated poly(methyl methacrylate)

    tained from hydroxypropyl b-cyclodextrin, but the (PMMA) NPs by bovine brain microvessel endo-

    highest cyclodextrin content was obtained for b- thelial cell monolayers. These NPs showed an in-

    cyclodextrin. The cyclodextrin NPs or the polymeric creased uptake by the endothelial cells of the BBB.

    NPs containing cyclodextrin were useful in targeting Troster et al. [143] demonstrated a nine-fold increase

    the water-insoluble drugs through oral or parentral in the accumulation of radioactivity in the brain area

    route. The presence of cyclodexrins in these NPs has after i.v. administration of polysorbate-80 coated14

    drastically reduced the surface negativity probably C-PMMA NPs. A recent study by Steiniger et al.

    due to their hydrophilicity; hence, the cyclodextrin [144] suggested that polysorbate-80 coated poly-coated NPs may help in avoiding the MPS.

    5.3.4. Polysorbate-coated NPs to penetrate the

    bloodbrain barrier

    Targeting drugs to the brain by crossing the

    bloodbrain barrier (BBB) has been a challenge. In

    this pursuit, many attempts have been made to

    develop novel drug delivery systems. BBB is formed

    by the tight endothelial cell junctions of the capil-

    laries within the brain, which limits the ability of

    many drugs to penetrate through the brain tissue in

    order to enter the central nervous system (CNS). It isknown that many regulators of the brain functions

    such as cytokines, transferrin, enkephalins, endor-

    phins or delta sleep inducing peptides pass through

    BBB from the vessels into brain [137,138] as well as

    some excitatory and depressant amino acids, pene-

    trate poorly through BBB. The poor BBB penetration

    of such substances makes the problem of drug

    targeting to the brain highly pertinent. However, the

    surface modified NPs have been used to deliver the

    anti-inflammatory drugs acting on the CNS because

    these can pass through BBB [139,140].The mechanism of enhancement of drug transport

    from the coated NPs through BBB is due to the

    number of mechanisms: (i) by binding the NPs to the

    inner endothelial lining of the brain capillaries and

    subsequently, particles deliver drugs to the brain by

    providing a large concentration gradient, thus en-

    hancing the passive diffusion; (ii) brain endothelialFig. 4. The schematic representation of the drug uptake through

    uptake by phagocytosis [141]. The effect of surfac- biological membrane from (A) free drug and (B) polysorbate-80tant coated NPs on drug permeation across the coated nanoparticle bound drug [91,141].

  • 7/21/2019 p_lecture_1236694477

    13/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 13

    (alkylcyanoacrylate) NPs are superior over the un- also reported an increased analgesic activity of

    coated NPs to transport drugs across BBB. dalargin-loaded PBCA NPs coated with polysorbate-

    Recently, investigations have been carried out 80 when administered in mice. In a further study,

    [145] with PBCA NPs as well as with the non- Alyautdin et al. [148] found that BBB crossing was

    biodegradable polystyrene (PS) NPs (200 nm in observed for low molecular mass and polar hydro-diameter) to study the transport of analgesic peptide, philic drug like tuborcurarin after loading it into

    dalargin to the brain. Its entry into the CNS of the polysorbate-80 coated PBCA NPs. An in-vivo per-

    mice was evaluated using the tail-flick procedure. fused rat brain was used along with the simultaneous

    Locomotor activity measurements were performed to recording of an electroencephalogram (EEG) since

    compare the toxicity of NPs. BBB permeability of the drug induces epileptic form seizures. An i.v.

    PBCA NPs was studied in-vitro using a co-culture of injection of the NPs demonstrated the appearance of

    bovine brain capillary endothelial cells and rat EEG seizures 5 min after the administration.

    astrocytes. Dalargin associated with PBCA NPs and Schroeder et al. [149] studied the transport of

    polysorbate-80 induced a potent and prolonged anal- dalargin, kytorphin (centrally-acting analgesics) and

    gesia, which was not observed by using polystyrene amitriptyline (antidipressant)-loaded PBCA NPs

    NPs, but not using the PBCA NPs. Locomotor coated with polysorbate-80 across the BBB. In-vivoactivity dramatically decreased in the mice dosed analgesic activity carried out in mice showed a

    with PBCA NPs, but not with the polystyrene NPs. drastic enhancement of analgesia for the drug-loaded

    The in-vitro and in-vivo results suggested that the NPs coated with polysorbate-80. The amitriptylin

    PBCA NPs induce a nonspecific opening of the BBB concentration in the brain increased, but the con-

    in the presence of polysorbate-80 allowing the centration in serum decreased for dextran-stabilized

    transport of dalargin into the CNS. Although polysorbate-80 coated NPs. These results indicate

    polysorbate-80 coated PBCA NPs are useful in that the surface modification of NPs by coating with

    increasing the penetration of drugs into the CNS, polysorbate-80 is effective in drug delivery through

    potential therapeutic applications are limited because BBB.

    of the high systemic NP concentration needed to

    deliver drugs to the CNS.

    In an effort to deliver anticancer drugs to the brain 6. Delivery of proteins and peptides using NPsusing NPs, Gulyaev et al. [146] demonstrated that

    the brain concentration of systemically administered Peptide drugs are attracting, as their role in

    doxorubicin was enhanced by more than 60-fold by physiopathology is better understood and because of

    binding it to polysorbate-80 coated PBCA NPs. the progress made in biotechnology and bioengineer-

    Doxorubicin was selected as a model drug due to its ing. Particularly, the development of DNA-recombi-

    potent antitumor activity and because the drug is not nant technology has made these compounds available

    able to cross the BBB by i.v. injection. Polysorbate- on large scale than in the past. However, the use of

    80 coated NPs reached the brain intact and released peptide in medicine is partly limited by their rapid

    the drug after endocytosis by the brain blood vessel degradation by proteolytic enzymes in the gastroin-

    endothelial cells. High brain concentrations achieved testinal tract; thus, they need to be administered

    in this study suggested a significant improvement in through the parentral route. The biological half-lifethe treatment of brain tumors. of peptides is short and needs frequent administra-

    Alyautdin et al. [147] studied the efficiency of tions. On the other hand, their transport across

    polysorbate-80 coated PBCA NPs in crossing BBB biological barriers is poor due to poor diffusivity and

    to deliver the water-insoluble analgesic drug, loper- lower partition coefficients. In this pursuit, the

    mide, in mice. Intravenous injection of the particulate biodegradable delivery systems have been

    polysorbate-80 coated NPs resulted in a long and proposed for the safe and controlled parentral ad-

    significant analgesic effect, which was measured by ministration of peptides [150].

    the tail flick method, while the uncoated NPs were Proteins and peptides are unstable in PLGA

    unable to produce analgesia. Alyautdin et al. [141] because of the hydrophobicity and acidity of PLGA

  • 7/21/2019 p_lecture_1236694477

    14/20

    14 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    [151]. Another problem is the fast burst release of release insulin from the nanospheres over a span of 6

    protein drugs from the PLGA matrices. In order to h. The 1.6% zinc insulin in PLGA with fumaric

    circumvent these problems, different approaches anhydride oligomer and iron oxide additives was

    have been explored to modify the properties of shown to be active orally and was able to control the

    PLGA matrices by using the hydrogel NPs plasma glucose levels. A number of properties of this[152,153]. This has prompted the development of formulation, including size, release kinetics, bioadhe-

    novel protein delivery systems. In these studies, siveness and ability to traverse the gastrointestinal

    bovine serum albumin (BSA) was encapsulated first epithelium have contributed to its oral efficacy.

    in PVA NPs, which were then loaded into PLGA In recent years, greater advances have been made

    microspheres using the phase separation method. The particularly by the research group of Professor

    protein loaded PLGAPVA composite NPs were Robert Langer at MIT (USA) on the development of

    then characterized and were having the nonporous gene delivery systems. However, a discussion on

    surface to release BSA for over 2 months. In a recent these systems is beyond the scope of this review.

    study by Gasper et al. [154], it was shown that the

    presence of end carboxyl group in PLGA resulted in

    a high protein loading of up to 4.86 mass% and the 7. Conclusions

    release continued for about 20 days. On the other

    hand, the presence of esterified carboxyl end groups The use of biodegradable polymers for the CR of

    in PLGA led to a lower loading (2.65 mass%) of therapeutic agents is now well established. Although

    proteins and a release of up to 14 days. currently there are only a small number of commer-

    The release kinetics and in-vivo effects of NPs cially available products that utilize this technology

    containing PGDF-Receptor b (PDGFRb) tyrphostin (e.g., Lupron Depot ), these polymers have great

    inhibitor, AG-1295, AG-1295-loaded PLA NPs were utility for the CR of several drugs like vaccines,

    prepared by the spontaneous emulsification/ solvent human growth hormone, insulin, anti-tumor agents,

    displacement technique [155]. The in-vitro release contraceptives and also vaccines. Long circulation of

    rate and the impact of drug/ polymer ratio on the size drugs in the body is the key in successful drug

    of NPs were determined. It was shown that by delivery and drug targeting to the site of action.

    modulating the formulation variables, release kinet- Many polymeric NPs have been developed for thisics and particle size were tailor-made to address the purpose. Certainly, surface modification is useful in

    clinical needs. A novel pulmonary delivery system of achieving these goals. From the polymer chemistry

    PLGA nanosphere (400 nm size) encapsulating the viewpoint, it is important to synthesize newer poly-

    physiologically active peptide was developed by mers and copolymers to match the hydrophilic and

    Kawashima et al. [156]. These were prepared by hydrophobic properties. Production of NPs using the

    using the modified emulsion solvent diffusion meth- environmentally friendly processes like supercritical

    od in water. The aqueous dispersions of PLGA fluids is quite a promising area of research to

    nanospheres administered pulmonarily to guinea pig develop the products that are free from the unwanted

    via nebulization reduced significantly the blood toxic residual solvents. Although many important

    glucose level for over 48 h when compared to the goals have been reached in achieving stabilization of

    nebulized aqueous solution of insulin as a reference. drugs in circulation, yet more investigations areAbout 85% of the drug was released from the needed to develop the newer materials in this area.

    nanospheres during the initial burst, followed by a

    prolonged release of the remaining drug for few

    hours in saline solution at 378C. Acknowledgements

    Zinc insulin was successfully encapsulated in

    various polyester and polyanhydride nanosphere We immensely thank the Council of Scientific and

    formulations using the phase inversion nanoencapsu- Industrial Research, New Delhi, India [Grant [

    lation technique [157]. The encapsulated insulin 80(0025)97/ EMR-II] for a major financial support

    maintained its biological activity and was able to of this study. Dr. Walter E. Rudzinski thanks the

  • 7/21/2019 p_lecture_1236694477

    15/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 15

    [17] P. Calvo, J.L. Vila-Jato, M.J. Alonso, Comparative in vitroSouthwest Texas State University, San Marcos for aevaluation of several colloidal systems, nanoparticles,research enhancement grant. We also thank Dr. M.I.nanocapsules and nanoemulsions as ocular drug carriers, J.

    Aralaguppi for his assistance. Pharm. Sci. 85 (1996) 530536.[18] F. Lescure, C. Seguin, P. Breon, P. Bourrinet, D. Roy, P.

    Couvreur, Preparation and characterization of novel poly-(methylidene malonate 2.1.2.)-made nanoparticles, Pharm.ReferencesRes. 9 (1994) 12701277.

    [19] C.A. Farrugia, M.J. Grover, Gelatin behavior in dilute[1] J. Kreuter, Nanoparticles, in: J. Kreuter (Ed.), Colloidal Drug aqueous solutions: Designing a nanoparticulate formulations,

    Delivery Systems, Marcel Dekker, New York, 1994, pp. J. Pharm. Pharmacol. 51 (1999) 643649.219342. [20] R. Fernandez-Urrusuno, P. Calvo, C. Remunan-Lopez, J.L.

    [2] C.G. Knight (Ed.), Liposomes From Physical Structure To Villa-Jato, M.J. Alonso, Enhancement of nasal absorption ofTherapeutic Applications, Elsevier, Amsterdam, 1981. insulin using chitosan nanopartilces, Pharm. Res. 16 (1999)

    [3] R. Langer, Biomaterials in drug delivery and tissue engineer- 15761581.ing: One laboratorys experience, Acc. Chem. Res. 33 (2000) [21] I.C. Aynie, C.Vauthier, E. Fattal, M. Foulquier, P. Couvreur,

    94101. Alginate nanoparticles as a novel carrier for antisense

    [4] R.P. Lanza, R. Langer, W.L. Chick, Principles of Tissue oligonucleotide, in: J.E. Diederichs, R. Muler (Eds.), Future

    Engineering, in: Academic Press, Austin, TX, 1997, pp. Strategies of Drug Delivery With Particulate Systems, Med-

    405427. pharm Scientific Publisher, Stuttgart, 1998, pp. 510.[5] L.B, Peppas, Recent advances on the use of biodegradable [22] C. Vauthier, S. Beanabbou, G. Spenlehauer, M. Veillard, P.

    microparticles and nanoparticles in the controlled drug Couvreur, Methodology of ultradispersed polymer system,

    delivery, Int. J. Pharm. 116 (1995 ) 1 9. S.T.P. Pharm. Sci. 1 ( 1991) 109 116.

    [6] A. Zimmer, J. Kreuter, Microspheres and nanoparticles used [23] E. Allemann, R. Gurnay, E. Doelker, Drug-loaded nanopar-

    in ocular drug delivery systems, Adv. Drug. Deliv. Rev. 16 ticles-Preparation methods and drug targeting issues, Eur. J.

    (1995) 6173. Pharm. 39 (1993) 173191.

    [7] P. Couvreur, L. Grislain, V. Lenaerts, F. Brasseur, P. Guiot, [24] P. Couvreur, C. Dubernet, F. Puisieux, Controlled drug

    in: P. Guiot, P. Couvreur (Eds.), Polymeric Nanoparticles and delivery with nanoparticles: Current possibilities and future

    Microspheres, CRC Press, Boca Raton, Florida, 1986. trends, Eur. J. Pharm. 41 (1995) 213.

    [8] D.F. Raney, Biomimetic transport, rational drug delivery, [25] M.J. Alonso, Nanoparticulate drug carrier technology, in: C.

    Biochem. Pharmacol. 59 (2000) 105114. Cohen, H. Bernstein (Eds.), Microparticulte Systems For the

    [9] K.E. Uhrich, S.M. Cannizzaro, R.S. Langer, K.M. Shakes- Delivery of Proteins and Vaccines, Marcel Dekker, New

    sheff, Polymeric systems for controlled drug release, Chem. York, 1996, pp. 203242.

    Rev. 99 ( 1999 ) 3181 3198. [26] P.D. Scholes, A.G.A. Coombes, L. Illum, S.S. Davis, M. Vert,

    [10] C. Monfardini, F.M. Veronese, Stabilization of substances in M.C. Davies, The preparation of sub-500 nm poly(lactide-

    circulation, Bioconjug. Chem. 9 (1998) 418450. co-glycolide) microspheres for site-specific drug delivery, J.

    [11] V.P. Torchilin, Polymer-coated long-circulating microparticu- Control. Rel. 25 (1993) 145153.

    late pharmaceuticals, J. Microencapsul. 15 (1998) 119. [27] M.F. Zambaux, F. Bonneaux, R. Gref, P. Maincent, E.

    [12] D.L. Wise, T.D. Fellman, J.E. Sanderson, R.L. Wentworth, Dellacherie, M.J. Alonso, P. Labrude, C. Vigneron, Influence

    Lactide/ glycolide acid polymers, in: G. Geregoriadis (Ed.), of experimental parameters on the characteristics of poly(lac-

    Drug Carriers in Biology and Medicine, Academic, London, tic acid) nanoparticles prepared by double emulsion method,

    1979, pp. 237270. J. Control. Rel. 50 (1998) 3140.

    [13] T.M. Jackanicz, H.A. Nash, D.L. Wise, J.B. Gregory, Poly [28] T. Niwa, H. Takeuchi, T. Hino, N. Kunou, Y. Kawashima,

    lactic acid as a biodegradable carrier for contraceptive Preparations of biodegradable nanospheres of water-soluble

    steroids, Contraception 8 (1973 ) 227 234. and insoluble drugs with D,L-lactide/ glycolide copolymer by

    [14] L.C. Andersson, D.L. Wise, J.F. Howes, An injectable a novel spontaneous emulsification solvent diffusion method

    sustained release fertility control system, Contraception 13 and the drug release behavior, J. Control. Rel. 25 (1993)(1976) 375384. 8998.

    [15] C.G. Pitt, M.M. Gratzi, A.R. Jeffcot, R. Zweidinger, A. [29] P. Wehrle, B. Magenheim, S. Benita, The Influence of

    Schindler, Sustained release drug delivery systems II: factors process parameters on the PLA nanoparticle size distribution

    affecting release rate for poly(e-caprolactone) and related evaluated by means of factorial design, J. Pharm. Biopharm.

    biodegradable polyesters, J. Pharm. Sci. 68 (1979) 1534 41 (1995) 1926.

    1538. [30] H. Murakami, H. Yoshino, M. Mizobe, M. Kobayashi, H.

    [16] C.G. Pitt, T.A. Marks, A. Schindler, Biodegradable drug Takeuchi, Y. Kawashima, Preparation of poly(D,L-lactide-co-

    delivery systems based on aliphatic polyesters: application to glycolide) latex for surface modifying material by a double

    contraceptives and narcotic antagonists, in: R. Baker (Ed.), coacervation method, Proced. Intern. Symp. Control. Rel.

    Controlled Release of Bioactive Materials, Academic, New Bioact. Mater. 23 (1996) 361362.

    York, 1980, pp. 19 43. [31] D.T. Birnbaum, J.D. Kosmala, D.B. Henthorn, L.B. Peppas,

  • 7/21/2019 p_lecture_1236694477

    16/20

    16 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    Controlled release of b-estradiol from PLAGA microparti- in: R.S. Manly (Ed.), Adhesion in Biological Systems,

    cles: The effect of organic phase solvent on encapsulation Academic, New York, 1970, pp. 185199.and release, J. Control. Rel. 65 (2000) 375387. [47] P. Couvreur, B. Kante, M. Roland, Les perspectives dutilisa-

    [32] R. Bodmeier, J.W. McGinity, Solvent selection in the prepa- tion des formes microdisperses comme vecteurs intracel-ration of poly(D,L-lactide) microspheres prepared by the lulaires, Pharm. Acta Helv. 53 (1978) 341347.

    solvent evaporation method, Int. J. Pharm. 43 (1988) 179 [48] P. Couvreur, B. Kante, M. Roland, P. Goit, P. Bauduin, P.186. Speiser, Polycyanoacrylate nanocapsules as potential

    [33] R. Arshaday, Preparation of porous and nonporous bio- lysosomotropic carriers: preparation, morphology and sorp-degradable polymeric hollow microspheres, J. Control. Rel.

    tive properties, J. Pharm. Pharmacol. 31 (1979) 331332.17 (1991) 122.

    [49] N. Behan, C. Birkinshaw, N. Clarke, A study of the factors[34] E. Allemann, R. Gurnay, E. Doelker, Preparation of aqueous

    affecting the formation of poly(n-butylcyanoacrylate)polymeric nanodispersions by a reversible salting-out pro-

    nanoparticles, Proced. Intern. Symp. Control. Rel. Bioact.cess: influence of process parameters on particle size, Int. J.

    Mater. 26 (1999) 11341135.Pharm. 87 (1992) 247253.[50] C. Lherm, R.H. Muller, F. Puiseux, P. Couvreur,[35] E. Allemann, J.C. Leroux, R. Gurnay, E. Doelker, Invitro

    Alkylcynoacrylate drug carriers: II. Cytotoxicity ofextended-release properties of drug-loaded poly(D,L-lactic)cyanoacrylate nanoparticles with different alkyl chain length,acid nanoparticles produced by a salting-out procedure,Int. J. Pharm. 84 (1992) 1322.Pharm. Res. 10 (1993) 17321737.

    [51] J.-L. De Keyser, C.J.C. De Cock, J.H. Poupaert, P. Dumont,[36] J.C. Leroux, E. Allemann, E. Doelker, R. Gurnay, New14 14

    Synthesis of C labeled acrylic derivatives: diethyl [3- C]approach for the preparation of nanoparticles by an emulsifi- 14methylidenemalonate and isobutyl [3- C] cyanoacrylate, J.cationdiffusion method, Eur. J. Pharm. Biopharm. 41Lable. Comp. Radiopharm. 27 (1989) 909916.(1995) 1418.

    [52] J.-L. De Keyser, J.H. Poupaert, P. Dumont, Poly(diethyl[37] G.D. Quintanar, Q.A. Ganem, E. Allemann, H. Fessi, E.

    methylidenemalonate) nanoparticles as a potential drug car-Doelker, Influence of the stabilizer coating layer on the

    rier: preparation, distribution and elimination after intraven-purification and freeze drying of poly (DL-lactic acid)

    nanoparticles prepared by emulsification-diffusion technique, ous and peroral administration to mice, J. Pharm. Sci. 80

    J. Microencapsulation 15 (1998) 107119. (1991) 6770.

    [38] J.W. Tom, P.G. Debenedetti, Particle formation with super- [53] T.K.M. Mabela, J.H. Poupaert, P. Dumont, A. Haemers,

    critical fluids a review, J. Aerosol Sci. 22 (1991) 555 Development of poly(dialkyl methylidenemalonate)

    584. nanoparticles as drug carriers, Int. J. Pharm. 92 (1993)

    [39] T.W. Randolph, A.D. Randolph, M. Mebes, S. Yeung, Sub- 7179.

    micron-sized biodegradable particles of poly(L-lactic acid) [54] P. Breton, D. Roy, L. Marchal-Heussler, C. Seguin, P.

    via the gas antisolvent spray precipitation process, Biotech- Couvreur, F. Lescure, New poly(methylidene malonate

    nol. Prog. 9 (1993) 429435. 2.1.2) nanoparticles: Recent developments, in: G. Gre-

    [40] L. Benedetti, A. Bertucco, M. Lora, P. Pallado, in: Atti del 38 goriadis, B. McCormack, G. Poste (Eds.), Targeting of

    Congresso I fluidi Supercriticai e le Loro Applicazioni, I. Drugs, Advances in System Constructs,Vol. 4, Plenum Press,

    Kikic and P. Alessi (Eds.), Trieste, 1995, p. 221. New York, 1994, pp. 161172.

    [41] K. Mishima, K. Matsuyama, D. Tanabe, S. Yamauchi, [55] F. Lescure, C. Seguin, P. Breton, P. Bourrinet, D. Roy, P.

    Microencapsulation of proteins by rapid expansion of super- Couvreur, Preparation and charecterization of novel poly-

    critical solution with a nonsolvent, AIChE J. 46 (2000) (methylidene malonoate 2.1.2.)-made nanoparticles, Pharm.

    857865. Res. 11 (1994) 12701277.

    [42] J.W. Tom, P.G. Debenedetti, Formation of bioerodiable [56] P. Breton, X. Guillon, D. Roy, F. Lescure, G. Riess, N. Bru,

    polymeric microspheres and microparticles by rapid expan- C. Roques-Carmes, Physico-chemical characterization, prep-

    sion of supercritical solution, Biotechnol. Prog. 7 (1991) aration and performance of poly(methylidene malonate 2.1.2)

    403411. nanoparticles, Biomaterials 19 (1998) 271281.

    [43] J.W. Tom, P.G. Debenedetti, R. Jerome, Preparation of [57] N. Bru-Magniez, X. Guillon, P. Breton, P. Couvreur, F.

    poly(L-lactic acid) and composite poly(L-lactic acid)-pyrene Lescure, C. Roques-Carmes, G. Riess, Method for preparingby rapid expansion of supercritical solution, J. Supercrit. malonate methylidene nanoparticles, nanoparticles optionally

    Fluids 7 (1994) 929. containing one or several biologically active molecules,

    [44] S. Mawson, K.P. Johnston, J.R. Combes, J.M. DeSimone, International Patent PCT WO 98/18455, 1998.

    Formation of poly(1,1,2,2-tetrahydroperfluorodecyl acrylate) [58] N. Bru-Magniez,V. Larras, G. Riess, P. Breton, P. Couvreur,

    submicron fibers and particles from supercritical carbon C. Roques-Carmes, Novel surfactant copolymers based on

    dioxide solutions, Macromolecules 28 (1994) 31823191. methylidene malonate, International Patent PCT WO 99/

    [45] K.C. Pani, G. Gladieux, G. Brandes, R.K. Kulkarni, F. 38898, 1999.

    Leonarda, The degradation of n-butyl alpha-cyanoacrylate [59] A.N. Mitra, P.K. Ghosh, S. Sahoo, pH and thermoresponsive

    tissue adhesive, II, Surgery 63 (1968) 481489. hydrogel nanoparticles, Proc. Intern. Symp. Control. Rel.

    [46] F. Leonarda, Hemostatic application of alpha cyanoacrylates: Bioact. Mater. 25 (1998) 234235.

    bonding mechanism and physiological degradation of bonds, [60] A.N. Mitra, P.K. Ghosh, S. Sahoo, Long circulating RES

  • 7/21/2019 p_lecture_1236694477

    17/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 17

    evading hydrophilic nanoparticles, Proc. Intern. Symp. Con- dable comb polyesters, Proc. Intern. Symp. Control. Release.

    Bioact. Mater. 26 (1999) 159160.trol. Rel. Bioact Mater. 25 (1998) 168169.[75] M.A. Breitenbach, W. Kamm, K.-D. Hungere, E. Hund, T.[61] R. Gref, Y. Minamitake, M.T. Peracchia, V. Trubetskoy, V.P.

    Kissel, Oral and nasal administration of tetanus toxoidTorchilin, R. Langer, Biodegradable long circulating poly-loaded nanoparticles consisting of novel charged biodegra-meric nanospheres, Science 18 (1994) 16001603.

    dable polyesters for mucosal vaccination, Proc. Intern. Symp.[62] M. Amiji, K. Park, in: S.W. Shalaby, Y. Ikada, R. Langer, J.Control. Release. Bioact. Mater. 26 (1999) 348349.Williams, (Eds.), Polymers of Biological Significance, ACS

    [76] M.J. Alenso, C. Losa, P. Calvo, J.L. Vila-Jato, Approaches toSymp. Ser. 540, Washington, DC, 1994.improve the association of amikacin sulphate to poly-

    [63] P. Calvo, C. Remunan-Lopez, J.L. Vila-Jato, M.J. Alonso,(cyanoacrylate) nanoparticles, Int. J. Pharm. 68 (1991) 69

    Novel hydrophilic chitosan and chitosan/ polyethylene oxide76.

    nanoparticles as protein carriers, J. Appl. Polym. Sci. 63[77] M. Ueda, A. Iwara, J. Kreuter, Influence of the preparation

    (1997) 125132.methods on the drying release behavior of loperamide-loaded

    [64] P. Calvo, C. Remunan-Lopez, J.L. Vila-Jato, M.J. Alonso, nanoparticles, J. Microencapsulation 15 (1998) 361372.Chitosan and chitosan/ ethylene oxidepropylene oxide [78] P. Couvreur, B. Kante, M. Roland, P. Speiser, Adsorption ofblock copolymer nanoparticles as novel carriers for proteins antineoplastic drugs to polyalkylcyanoacrylate nanoparticlesand vaccines, Pharm. Res. 14 (1997) 14311436. and their release in calf serum, J. Pharm. Sci. 68 (1979)

    [65] R. Fernandez-urrusuno, P. Calvo, C. Remunan-Lopez, J.L. 15211523.Vila-Jato, M.J. Alonso, Enhancement of nasal absorption of [79] M.A. Radwan, In vitro evaluation of poly-

    insulin using chitosan nanoparticles, Pharm. Res. 16 (1999) isobutylcyanoacrylate nanoparticles as a controlled drug15761591. carrier for theophylline, Drug Dev. Ind. Pharm. 21 (1995)

    [66] P. Calvo A.S. Boughaba, M. Appel, E. Fattal, M.J. Alonso, P. 23712375.Couvreur, Oligonucleotidechitosan nanoparticles as new [80] M.A. Egea, F. Gamisani, J. Valero, M.E. Garcia, M.L.

    gene therapy vector, Proc. 2nd World Meeting APGI/APV Garcia, Entrapment of cisplatin into biodegradable poly-

    Paris, 1998, pp. 11111112. alkylcyanoacrylate nanoparticles, Farmaco 49 (1994) 211

    [67] H.-Q. Mao, K. Ray, S.M. Walsh, J.T. August, K.W. Leong, 217.

    DNAchitosan nanoparticles for the gene delivery, Proc. [81] K. Chukwu, U. Ishroder, P. Sommerfeld, B.A. Sabel, Load-

    Intern. Symp. Control. Release. Bioact. Mater. 23 (1996) ing some psychopharmacologic agents onto poly(butyl-

    401402. cyanoacrylate) nanoparticles-a means of targeting to the

    [68] K. Ray, H.-Q. Mao, K.Y. Lin, S.-K. Huang, K.W. Leong, brain and improving therapeutic efficiency, Proced. Intern.

    Oral immunization with DNAchitosan nanoparticles, Proc. Symp. Control. Rel. Bioact. Mater. 26 (1999) 11481149.

    Intern. Symp. Control. Release. Bioact. Mater. 26 (1999) [82] J.Vora, N. Bapat, M. Boroujerdi, Investigation of the relative

    348349. affinity of doxorubicin for neutral and negatively charged

    [69] V.L. Truong-Le, H. Mao, S. Walsh, K.W. Leong, J.T. August, particulate carriers, Drug Dev. Ind. Pharm. 19 (1993) 759

    Delivery of DNA vaccine using gelatinDNA nanoparticles, 771.

    Proc. Intern. Symp. Control. Rel. Bioact. Mater. 24 (1997) [83] J. Martin, P. Macchi, M. Hoffman, P. Maincent, Study of the

    3940. binding mechanisms of drugs onto polyalkylcyanoacrylate

    [70] X.-X. Tian, M.J. Groves, Formulation and biological activity nanoparticles, J. Pharm. Belg. 49 (1994) 498508.

    `of antineoplastic proteoglycans derived from Mycobacterium [84] E. Benoit, O. Prot, P. Maincent, J. Bessiere, Adsorption of

    vaccae in chitosan nanoparticles, J. Pharm. Pharmacol. 51 beta-blocker onto polyisobutylcyanoacrylate nanoparticles

    (1999) 151157. measured by depletion and dielectric method, Pharm. Res. 11

    [71] H. Tokumitsu, H. Ichikawa, Y. Fuukumori, Chitosan (1994) 585588.

    gadopenteic acid complex nanoparticles for gadolinium [85] H.S. Yoo, J.E. Oh, K.H. Lee, T.G. Park, Biodegradable

    neutron-capture therapy of cancer: preparation by novel nanoparticles containing doxorubicinPLGA conjugate for

    emulsiondroplet coalescence technique and characteriza- sustained release, Pharm. Res. 16 (1999) 11141118.

    tion, Pharm. Res. 16 (1999) 18301835. [86] C. Washington, Drug release from microdisperse systems. A

    [72] C. Vautheir, I. Aynie, P. Couvreur, E. Fattal, Pharmacokinetic critical review, Int. J. Pharm. 58 (1990) 112.and tissue disposition of oligonucleotide associated with [87] B. Magenheim, S. Benita, Nanoparticle characterization: a

    alginate nanoparticles, Proc. Intern. Symp. Control. Rel. comprehensive physicochemical approach, S.T.P. Pharm. Sci.

    Bioact. Mater. 25 (1998) 228229. 1 (1991) 221224.

    [73] T. Jung, A. Breitenbach, T. Kissel, Sulfobutylated poly(vinyl [88] M.S. El-Samaligly, P. Rohdewald, H.A. Mohmoud, Poly-

    alcohol)-graft-poly(lactide-co-glycolide) facilitate the prepa- alkylcyanoacrylate nanocapsules, J. Pharm. Pharmacol. 38

    ration of small negatively charged biodegradable nanos- (1986) 216218.

    pheres for protein delivery, J. Control. Rel. 67 (2000) 157 [89] A.M. LeRay, M. Vert, J.C. Gautier, J.P. Benoit, End-chain

    169. radiolabeling and in vitro stability studies of radiolabeled

    [74] A. Breitenbach, G. Nykamp, T. Kissel, Self-assembling poly(hydroxy) nanoparticles, J. Pharm. Sci. 83 (1994) 845

    colloidal carriers for protein delivery: nanoparticulate poly- 851.

    mer protein conjugates with novel water soluble biodegra- [90] M. Fresta, G. Puglisi, G. Giammona, G. Cavallaro, N.

  • 7/21/2019 p_lecture_1236694477

    18/20

    18 K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20

    Micali, P.M. Furneri, Pefloxacin mesilate- and ofloxacin- [105] R. Gref, A. Domb, P. Quellec, T. Blunk, R.H. Muller, J.M.

    loaded polyethylcyanoacrylate nanoparticles; characterization Verbavatz, R. Langer, The controlled intravenous delivery

    of the colloidal drug carrier formulation, J. Pharm. Sci. 84 of drugs using PEG-coated sterically stabilized nanos-

    (1995) 895901. pheres, Adv. Drug Deliv. Rev. 16 (1995) 215233.

    [91] G. Cavallaro, M. Fresta, G. Giammona, G. Puglisi, A.Villari, [106] M.T. Peracchia, S. Harnisch, H. Pinto-Alphandary, A.

    Entrapment of b-lactams antibiotics in poly- Gulik, J.C. Dedieu, D. Desmaele, J. dAngelo, R.H. Muller,ethylcyanoacrylate nanoparticles: studies on the possible in P. Couvreur, Visualization of in vitro protein-rejecting

    vivo application of this colloidal delivery system, Int. J. properties of PEGylated stealth polycyanoacrylate

    Pharm. 111 (1994) 3141. nanoparticles, Biomaterials 20 (1999) 12691275.

    [92] M.Y. Leavy, S. Benita, Drug release from submicron O /W [107] B.D. Ratner, A.B. Johnston, T.J. Lenk, Biomaterial surface,

    emulsion: A new in vitro kinetic evaluation model, Int. J. J. Biomed. Mat. Res. Appl. Biomat. 21 (1987) 5990.

    Pharm. 66 (1990) 29 37. [108] H. Carstensen, B.W. Muller, R.H. Muller, Adsorption of

    [93] R. Jalil, J.R. Nixon, Biodegradable poly(lactic acid) and ethoxylated surfactants on nanoparticles. I. Characterization

    poly(lactide-co-glycolide) microcapsules: problems associ- by hydrophilic interaction chromatography, Int. J. Pharm.

    ated with preparative techniques and release properties, J. 67 (1991) 2937.

    Microencapsul. 7 (1990) 297325. [109] R. Gref, Y. Minamitake, M.T. Peracchia, V. Trubetskoy, R.

    [94] B. Magenheim, M.Y. Levy, S. Benita, A new in vitro Langer, Biodegradable long-circulating polymeric

    technique for the evaluation of drug release profiles from nanoparticles, Science 263 (1994) 16001603.

    colloidal carriers-ultrafiltration technique at low pressure, Int. [110] S.I. Joen, J.H. Lee, J.D. Andrade, P.G. de Gennes, Protein-

    J. Pharm. 94 (1993 ) 115 123. surface interactions in the presence of polyethylene oxide. I.[95] J.M. Rodrigues Jr, H. Fessa, C. Bories, F. Pusieux, J.-Ph. Simplified theory, J. Colloid. Interf. Sci. 142 (1991) 149

    Devissaguet, Premaquine-loaded poly(lactide) nanoparticles: 158.

    physicochemical study and acute tolerance in mice, Int. J. [111] D. Bazile, C. Prud Homme, M. Bassoullet, M. Marlard, G.

    Pharm. 126 (1995) 253 260. Spenlehauer, M. Veillard, Stealth Me-PEG PLA nanoparti-

    [96] M. Polakovic, T. Gorner, R. Gref, E. Dellacherie, Lidocaine cles avoid uptake by the mononuclear phagocyte system, J.

    loaded biodegradable nanospheres. II. modeling of drug Pharm. Sci. 84 (1995) 493498.

    release, J. Control. Rel. 60 (1999) 169177. [112] M. Tobio, R. Gref, A. Sanchez, R. Langer, M.T. Alonso,

    [97] Z. Lu, J. Bei, S. Wang, A method of the preparation of Stealth PLAPEG nanoparticles as protein carriers for nasal

    polymeric nanocapsules without stabilizer, J. Control. Rel. administration, Pharm. Res. 15 (1998) 270275.

    61 (1999) 107112. [113] R. Gref, G. Miralles, E. Dellacherie, Polyoxyethylene-

    [98] H. Muller, K.M. Willis, Surface modification of i.v. inject- coated nanospheres: effect of coating on zeta potential and

    able biodegradable nanoparticles with poloxamer polymers phagocytosis, Polymer Int. 48 (1999) 251256.

    and poloxamine 908, Int. J. Pharm. 89 (1993) 2531. [114] T. Govender, T. Riley, S. Stolnik, M.C. Garnett, L. Illum,

    [99] C.J. Van Oss, Phagocytosis as a surface phenomenon, Annu. S.S. Davis, PLAPEG nanoparticles for site specific deliv-

    Rev. Microbiol. 32 ( 1978) 19 39. ery: drug incorporation studies, J. Control. Rel. 64 ( 2000)

    [100] E. Allemann, G. Patricia, J.C. Leroux, B. Luc, R. Gurnay, 269347.

    Kinetics of blood component-adsorption on poly(D,L-lac- [115] S.Y. Kim, I.G. Shin, Y.M. Lee, Preparation and characteriza-

    tide) nanoparticles: Evidence of compliment C component tion of biodegradable nanospheres composed of methoxy3

    involvement, J. Biomed. Mater. Res. 37 (1997) 229234. poly(ethylene glycol) and DL-lactide block copolymer as

    [101] T. Blunk, M. Luck, A. Calvoer, D.F. Hochstrasser, J.C. novel drug carriers, J. Control. Rel. 56 (1998) 197208.

    Sanchez, B.W. Muller, R.H. Muller, Kinetics of plasma [116] M.T. Peracchia, E. Fattal, D. Desmaele, M. Bensard, J.P.

    protein adsorption on model particles for controlled drug Noel, J.M. Gomis, M. Appel, J. dAngelo, P. Couvreur,

    delivery and drug targeting, Eur. J. Pharm. Biopharm. 42 Stealth PEGylated polycyanoacrylate nanoparticles for

    (1996) 262268. intravenous administration and spleenic targeting, J. Con-

    [102] K. Thode, M. Luck, W. Semmler, R.H. Muller, M. Kresse, trol. Rel. 60 (1999) 121128.

    Determination of plasma protein adsorption on magnetic [117] A. Gessnerl, B.R. Paulke, R.H. Muller, Plasma protein

    iron oxide: sample preparation, Pharm. Res. 14 (1997) adsorption on poly(ethylene-glycol) (PEG) modified poly-905910. styrene nanoparticles: influence of PEG surface density,

    [103] K. Thode, M. Luck, W. Semmler, R.H. Muller, M. Kresse, Proc. Intern. Symp. Control. Rel. Bioact. Mater. 26 (1999)

    Influence of sample preparation on plasma protein ad- 597598.

    sorption patterns on polysaccharide-stabilized iron oxide [118] M.T. Peracchia, R. Gref, Y. Minamitake, A. Domb, R.

    particles and end-terminal microsequencing of unknown Langer, PEG-coated injectable nanospheres prepared from

    proteins, J. Drug Target. 5 (1997) 3543. amphiphilic diblock and multiblock copolymers: investiga-

    [104] M. Luck, K.F. Pistel, Y. Li, T. Blunk, R.H. Muller, T. tion of drug encapsulation and release characteristics, J.

    Kissel, Plasma protein adsorption on biodegradable micro- Control. Rel. 46 (1997) 223231.

    spheres consisting of poly(D,L-lactide-co-glycolide), poly(L- [119] M.T. Peracchia, C. Vauthier, F. Puisieux, P. Couvreur,

    lactide) or ABA triblock copolymers containing poly(oxy- PEGPIBCA nanoparticles with different PEG-coating

    ethylene), J. Control. Rel. 55 (1998) 107120. configurations formed by chemical coupling of PEG,

  • 7/21/2019 p_lecture_1236694477

    19/20

    K.S. Soppimath et al. / Journal of Controlled Release 70 (2001) 1 20 19

    Proced. Intern. Symp. Control. Rel. Bioact. Mater. 23 [132] M. Demoy, J.P. Andreux, C. Weingarten, B. Gouritin, V.

    (1996) 395396. Guilloux, P. Couvreur, Spleen capture of nanoparticles:

    [120] M.T. Peracchia, C. Vauthier, F. Puisieux, P. Couvreur, Influence of animal species and surface characteristics,

    Development of sterically stabilized poly(isobutyl-2- Pharm. Res. 16 (1999) 3741.

    cyanoacrylate) nanoparticles by chemical coupling of poly- [133] T. Shinoda, A. Maeda, S. Kojima, S. Kagatani, Y. Konno,

    (ethylene glycol), J. Biomed. Mater. Res. 34 (1997) 317 T. Sonobe, T. Akaike, Nanosphere coated with lactosyl-326. polystyrene polymer as a targeting carrier to hepatocytes,

    [121] M.T. Peracchia, C. Vauthier, D. Desmaele, A. Gulik, J.C. Drug Deliv. 6 (1999) 147151.

    Dedieu, M. Demoy, J. dAngelo, P. Couvreur, PEGylated [134] C.S. Cho, Y.I. Jeong, T. Ishihara, R. Takei, J.U. Park, K.H.

    nanoparticles from a novel methoxypolyethylene glycol Park, A. Maruyama, T. Akaike, Simple preparation of

    cyanoacrylatehexadecyl cyanoacrylate amphiphilic co- nanoparticles coated with carbohydrate-carrying polymers,

    polymer, Pharm. Res. 15 ( 1998) 550 556. Biomaterials 18 (1997) 323 326.

    [122] M.T. Peracchia, C. Vauthier, C. Passirani, P. Couvreur, D. [135] A. Maruyama, T. Ishihara, S.W. Kim, T. Akaike, Nanoparti-

    Labarre, Compliment consumption by poly(ethylene glycol) cle DNA carrier with poly(L-lysine) grafted polysaccharide

    in different conformations chemically coupled to copolymer and poly(D,L-lactic acid), Bioconjug. Chem. 8

    poly(isobutyl-2-cyanoacrylate) nanoparticles, Life Sci. 61 (1997) 735742.

    (1997) 741761. [136] D. Duchene, D. Wouessidjewe, G. Ponchel, Cyclodextrins

    [123] F.D. Jaeghere, E. Allemann, J.C. Leroux, W. Stevels, J. and carrier systems, J. Control. Rel. 62 (1999) 263268.

    Feijen, E. Doelker, R. Gurny, Formulation and lyoprotec- [137] S. Raeissi, K. Audus, In vitro characterization of blood

    tion of poly(lactic acid-co-ethylene oxide) nanoparticles: brain barrier permeability to delta sleep-inducing peptide, J.Influence on the physical stability and in vitro cell uptake, Pharm. Pharmacol. 41 (1989) 848852.

    Pharm. Res. 16 ( 1999 ) 859 866. [138] B. Zlokovich, The in vivo approaches for studying peptide

    [124] M. Vittaz, D. Razile, G. Spenlehauer, T. Verrecchia, M. interaction at the bloodbrain barrier, Peptides 10 (1989)

    Veillard, F. Puisieux, D. Labarre, Effect of PEO surface 249254.

    density on long-circulating PLAPEO nanoparticles which [139] U. Schroeder, B.A. Sabel, Nanoparticles, a drug carrier

    are very low complement activators, Biomaterials 17 system to pass the bloodbrain barrier, permit central

    (1996) 15751581. analgesic effects of i.v. dalargin injections, Brain Res. 710

    [125] J.C. Ha, S.Y. Kim, Y.M. Lee, Poly(ethylene oxide)poly- (1996) 121124.

    (propylene oxide) poly(ethylene oxide) (Plunoric)/ poly(e- [140] U. Schroeder, P. Sommerfeld, B.A. Sabel, A efficacy of oral

    caprolactone) (PCL) amphiphilic block copolymeric nanos- dalargin-loaded nanoparticle delivery across the blood

    pheres I. Preparation and characterization, J. Control. Rel. brain barrier, Peptide 19 (1998) 777780.

    62 (1999) 381392. [141] R. Alyautdin, D. Gothier, V. Petrov, D. Kharkevich, J.

    [126] S. Storm, S.O. Belliot, T. Daemen, D.D. Lasic, Surface Kreuter, Analgesic activity of the hexapeptide dalargin

    modification of nanoparticles to oppose uptake by the adsorbed on the surface of polysorbate 80-coated poly(butyl

    mononuclear system, Adv. Drug Deliv. Rev. 17 (1995) cyanoacrylate) nanoparticles, Eur. J. Pharm. Biopharm. 41

    3148. (1995) 4448.

    [127] L. Illum, S.S. Davis, Effect of the nonionic surfactant [142] G. Borchardt, L.A. Kenneth, S. Fenlin, J. Kreuter, Uptake

    poloxamer 338 on the fate and deposition of polystyrene of surfactant-coated poly(methyl methacrylate)-nanoparti-

    microspheres following intravenous administration, J. cles by bovine brain microvessel endothelial cell mono-

    Pharm. Sci. 72 (1983) 1086 1089. layers, Int. J. Pharmacol. 110 (1994) 29 35.

    [128] L. Illum, S.S. Davis, The organ uptake of intravenously [143] S.D. Troster, U. Muller, J. Kreuter, Modification of the

    administered colloidal particles can be altered using a body distribution of poly(methyl methacrylate) nanoparti-

    non-ionic surfactant (poloxamer 338), FEBS Letts. 167 cles in rats by coating with surfactants, Int. J. Pharm. 61

    (1984) 7982. (1990) 85100.

    [129] S. Rudt, R.H. Muller, In vitro phagocytosis assay of nano- [144] S. Steiniger, D. Zenker, H.V. Briesen, D. Begley, J. Kreuter,

    and microparticles by chemiluminescence. III. Uptake of The influence of polysorbate 80-coated nanoparticles on

    differently sized surface-modified particles, and its correla- bovine brain capillary endothelial cells in vitro, Proced.tion to particle properties and in vivo distribution, Eur. J. Intern. Symp. Control. Rel. Bioact. Mater. 26 (1999) 790

    Pharm. Sci. 1 (1993) 3139. 791.

    [130] L. Illum, S.S. Davis, R.H. Muller, E. Mak, P. West, The [145] J.C. Olivier, L. Fenart, R. Chauvet, C. Pariat, R. Cecchelli,

    organ distribution and circulation time of intravenously W. Couet, Indirect evidence that drug brain targeting using

    injected colloidal carriers sterically stabilized with a block polysorbate 80-coated polybutylcyanoacrylate nanoparticles

    copolymer poloxamine 908, Int. J. Pharm. 89 (1993) 25 is related to toxicity, Pharm. Res. 16 (1999) 18361841.

    31. [146] A.E. Gulyaev, S.E. Gelperina, I.N. Skidan, A.S. Antropov,

    [131] S.M. Moghimi, T.A. Gray, Single dose of iv injected G.Y. Kivman, J. Kreuter, Significant transport of doxorubi-

    poloxamine coated long-circulating particle triggers macro- cin into the brain with polysorbate 80-coated nanoparticles,

    phage clearance of subsequent doses in rats, Clin. Sci. 93 Pharm. Res. 16 (1999) 15641569.

    (1997) 371379. [147] R.N. Alyautdin, V.E. Petrov, K. Lan