You are on page 1of 7

Current Drug Targets, 2004, 5, 449-455 449

Drug Delivery Systems: Past, Present, and Future

Rubiana M. Mainardes1 and Luciano P. Silva2,*

1
Pós-Graduação em Ciências Farmacêuticas, Faculdade de Ciências Farmacêuticas, Universidade Estadual Paulista
“Júlio de Mesquita Filho”, Araraquara, 14802-902, Brazil and 2Pós-Graduação em Biologia Animal, Instituto de
Biologia, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Brasília, 70910-900, Brazil

Abstract: Drug delivery systems are essential components of drugs controlled release. In the last decades, several drug
delivery technologies have emerged including capsules, liposomes, microparticles, nanoparticles, and polymers. These
components must be biocompatible, biodegradable, and display a desired biodistribution providing a long-term
availability of the therapeutic at specific target over time.

Key Words: Drug design, microtechnology, nanotechnology, nanospheres, nanocapsules, antibody-based drug delivery.

INTRODUCTION the first known people to actually carry out chemical


processes to therapeutic. A number of systems containing
The applicability of drugs is almost always a delicate
polymers existed and commonly waxes were added with the
compromise between their therapeutic roles and side effects.
drugs to extend their action mechanisms or improve some
Current advances are aiding the discovery and rationale
feature like site specificity. From Industrial Revolution to
design of many new classes of drugs. It is clear that to turn
1970 different drug delivery technologies were developed.
these new advances into clinical effectiveness is crucial to The Industrial Revolution had a major effect on how Phar-
improve specific drug delivery methods opening novel macy was practiced. The advent of the machines allowed the
therapeutic horizons.
large scale production of important systems commonly used
Controlled drug delivery technology represents one of the nowadays, including capsules and microparticles. After
most intriguing frontier areas of science, which involves 1970, site directed drug delivery systems developed very
multidisciplinary fields. Biology, pharmacy, chemistry, rapidly and such strategies have been constantly “updated”
physics, and medicine work together to provide rapid and including liposomes (immunoliposomes, magnetoliposomes),
consistent advances in this vanguard area. nanoparticles (magnetic nanoparticles, polymeric nanopar-
ticles), high technology polymers (dendrimers), and virus.
Therefore, the development of techniques that could
selectively deliver drugs to the pathological site is currently For proper perspective, a simple literature search with the
one of the most important areas of drug research. This keywords "drug" and "delivery"; "drug" and "target"; and
review focuses on the fundamental concepts and approaches "drug" and "design" for the last 13 years is shown in Fig. (1)
of drug delivery systems with particular emphasis on recent revealing the crescent academic importance of these area.
and perspective advances on this area.
DRUG DELIVERY VEHICLES ON A PERSPECTIVE
HISTORICAL PERSPECTIVE OF DRUG DELIVERY VIEW
SYSTEMS
Finding solutions to deliver drugs at specific sites is not a
The drug delivery system concept is not new despite contemporary issue but remains as a crucial biomedical
great progress has recently been made in this area. Modern aspect. The constant development and design of optimized
drug delivery systems have been modeled for centuries and strategies involving polymers, nanoparticles, liposomes, and
their development appear parallel that of Humanity. The others active or passive delivery systems has been a
history of drug delivery technology can be didactically revolution on the way that the drugs exert their actions and
divided in three parts. From Pharmacy origin to Industrial several novel strategies are developed and tested all years.
Revolution, macroscopic delivery systems were developed.
It is impossible to put an exact date when drug delivery POLYMERS IN DRUG DELIVERY
approaches originated since people were practicing Phar-
One of the enduring features of drug delivery techno-
macy before the word even existed. The Ancient Populations
of Mesopotamia, Egypt, China, and indigenous peoples of logies is the central role that polymers play in control of drug
the Western Hemisphere (Aztecs, Mayans, and Incas) were release, and fabrication of drug delivery devices. The need
for polymers with desired biological and physical properties
has generated continuing interest in novel polymer synthesis.
*Address correspondence to this author at the Pós-Graduação em Biologia Classification of polymers in drug delivery applications
Animal, Instituto de Biologia, Universidade de Brasília, Campus can be difficult due to the inherent diversity of chemical
Universitário Darcy Ribeiro, Brasília, 70910-900, Brazil; structures. In broad terms, polymers may be classified as
E-mail: paulinoluciano@unb.br
either biodegradable or nonbiodegradable. Biodegradable

1389-4501/04 $45.00+.00 © 2004 Bentham Science Publishers Ltd.


450 Current Drug Targets, 2004, Vol. 5, No. 5 Mainardes and Silva

Fig. (1). Occurrences in the Institute for Scientific Information - ISI database, including all journals of the Science Citation Index - SCI to
"drug and delivery", "drug and target", and "drug and design". Per year search results for the last 13 years of the used words. Results are
shown for abstract, keyword, and title searches.

devices have garnered much of the recent attention and Polyester-based polymers are the best characterized and
development in drug delivery systems because they do not the most widely studied biodegradable systems. Polylactic
require retrieval or further manipulation after introduction acid (PLA), polyglycolic acid (PGA) and their copolymers
into the body [1]. In the realm of degradable polymers, there polylactic-co-glycolic acid (PLGA) are some of the most
exists another level of classification based upon the mechan- well-characterized biomaterials with regard to design and
ism of erosion. It is essential recognize that degradation is a performance for drug-delivery applications based on
chemical process, whereas erosion is a physical phenomena diffusive and/or erosive biodegradative aspects [1, 3, 8-13].
dependent on dissolution and diffusion processes. Two It is possible to modify the mechanical, thermal and
mechanisms of polymer erosion can be identified: surface biological properties of PLA by altering its stereochemistry.
and bulk erosion. For most biodegradable polymers both Further biodegradability can be tuned by changing the
mechanisms occur, but the relative extent of surface or bulk proportion of PLA and PGA in the copolymer [1, 2]. The
erosion varies radically with the chemical structure of the lactide/glycolide polymers chains are cleaved by hydrolysis
polymer backbone [1, 2]. to form naturally occurring metabolites (lactic and glycolic
A wide array of natural and synthetic biodegradable acids), which are eliminated from the body by the citric acid
polymers has been investigated for prolonged drug release or cycle [1, 3, 7, 14-17].
drug targeting. However, only a few of them are actually The need to develop biodegradable polymers that inhibit
considered biocompatible. Biodegradable polymers are drug release by diffusion mechanisms and allow drug release
degraded in vivo, either enzimatically or non-enzimatically only after the hydrolysis of polymer chains at the surface of
to produce biocompatible, toxicology safe by-products which the device was the motivation for designing the polyor-
are further removed by the normal metabolic pathways. toesters for drug delivery [1]. Polyorthoesters have been
Drugs formulated in polymeric devices are released either by under development since the 1970. To date four polymer
diffusion through the polymer barrier, or by erosion of the families have been developed and they have singular features
polymer material, or by a combination of both diffusion and among all biodegradable polymers, as mechanical properties
erosion mechanisms [3-5]. Both of these mechanisms are can be readily varied by choosing appropriate diols or
influenced by the degradation rate of polymer [6, 7]. The mixture of diols used in their synthesis [2, 18].
polymers selected for the parenteral administration must Polyanhydrides have been investigated as an important
meet several requirements like biocompatibility, drug com-
biomaterial used for short-term release of drugs for more
patibility, suitable biodegradation kinetics and mechanical
than two decades [19]. Polyanhydrides are characterized by
properties, and ease of processing [3]. their rapid degradation followed by fast erosion of material,
Drug Delivery Systems Current Drug Targets, 2004, Vol. 5, No. 5 451

but at the same time can be designed to release drugs that their activity site) and microparticles (e.g. slow release) into
last from days to weeks by suitable choice of monomers [2]. a single system circumventing problems associated with
The more hydrophobic the monomer, the more stable the each system [34].
anhydride bond is to hydrolysis. Aliphatic polyanhydrides
degrade within days whereas aromatic polyanhydrides POLYMERIC MICELLES
degrade over several years [1, 20]. Block copolymer micelles are promising carriers in drug
Since poly-amino acids are chemically and structurally delivery systems. Originally, the block copolymer micelles
related to natural proteins, the synthesis of amino acid-based composed of amphiphilic block copolymers were used as a
polymers has been explored as a potential source of new carrier of various hydrophobic drugs including anticancer
biomaterials. Starting from about 1970, the use of both homo agents [35]. A polymeric micelle usually consists of several
and copolymers of amino acids was studied for a variety of hundred block copolymers and has a diameter ranging from
biomedical applications [21-25]. Poly-amino acids that have 20 to 50nm. There are two spherical concentric regions of a
good biocompatibility have been investigated to deliver low- polymeric micelle, a densely packed core consisting of
molecular-weight compounds. However, amino acid-based hydrophobic blocks and a shell consisting of a dense brush
drug delivery approaches are limited by their antigenic of hydrophilic block. Polyethylene oxide (PEO) is frequently
potentials and poor controlled release due to the dependence the hydrophilic block, but others are being studied.
on enzymes for biodegradation. So far, only a small number Hydrophobic blocks include poly-alpha-hydroxy acids and
of polyglutamic acid and copolymers have been identified as poly-L-amino acids [36, 37]. The molecular weight of the
promising candidate materials for drug delivery [26, 27]. hydrophobic block is usually less than 2000g per mol [38].
A relatively novel class of biodegradable polymers There are a number of reasons why polymeric micelles
belonging to polyphosphoesters has a single backbone are interesting as drug carriers. As a solubilizing agent for
consisting of phosphorous atoms attached to either carbon or hydrophobic drugs they have a clear advantage over low
oxygen. The uniqueness of this class of polymers lies in the molecular weight surfactants in view of the higher stability
chemical reactivity of phosphorous, which enables a wide of the produced micelles [35, 36]. This higher stability is
range of potential side-chains to be attached for manipulating reflected in terms of the usually very low critical micelle
the molecular weight, biodistribution, and biodegradation concentration of polymeric surfactants. This means that
rates of the polymer [2]. polymeric micelles are resistant to dilution effects, upon for
example intravenous administration of the drug formulation.
Chitosan is a polysaccharide derived by deacetylation of
Another important characteristic of micelles, when compared
chitin, a by-product of the seafood industry. Chitosan meets
with other typical delivery methods, is their small and
the important requirements for excipients in drug delivery,
uniform particle size distribution. In theory, particle sizes
including biocompatibility and biodegradability. Chitosan
can go down to the order of 10 nm for non-loaded polymeric
has been investigated as mucoadhesive polymer for years
micelles. This size is still large enough to accomplish passive
[28], and more recently as a permeation enhancer for drug
targeting to cancer and inflamed tissues. As said above, the
delivery in vitro [29] and at mucosal epithelia [30]. The
hydrophilic corona of the micelles may prevent interaction
degree of deacetylation and derivatization with various side- with blood components. This characteristic and their small
chains can be a source of manipulation for site-directed drug
size prevent immune system recognition, and thus long
delivery applications [2, 31].
circulation times in the blood stream may be achieved.
Cyclodextrins (CDs) are potential high-performance Active targeting is also possible by modifying the peripheral
carrier materials that have the ability to alter physical, chain ends of the polymers with targeting specific ligands.
chemical, and biological properties of drugs through the For the release of the drugs once the micelles have reached
formation of inclusion complexes both in solution and solid their targets, degradable or stimuli-responsive micelles have
state. The alpha, beta, and gamma CDs are the most common been recently developed [35- 38].
naturally occurring CDs consisting of six, seven, and eight In addition to conventional therapeutic drugs, block
D-glucopyranose residues, respectively, linked by alpha-1,4 copolymer micelles might play a role in gene delivery.
glycosidic bonds into a macrocycle [2]. Recently, a number
Several groups have utilized highlight electrostatic binding
of CD derivatives and polymers have been suggested to
to form micelle-like complexes of block copolymers with
obtain better inclusion abilities. The hydrophilic and hydro- oppositely plasmid DNA.
phobic CDs are useful for the immediate or prolonged release
type formulations, respectively. Commonly, the combination Lastly, block copolymer form unique thermolar sensitive
of CDs and other carrier materials is useful for optimizing hydrogels for drug delivery and novel micelles for diagnostic
the release rate of drugs [32]. In drug discovery, CDs have imaging [39, 40]. The wide breadth of roles in drug delivery
mainly been used to increase the aqueous solubility and strategies is evident, and owing to their unique ability to
stability of poor hydrophilic drugs. CDs complexation of form nanoscopic supramolecular structures, additional
lipophilic drugs has also been shown to increase drug bio- approaches in drug delivery and similar fields such as tissue
availability [33]. For some years CDs and their hydrophilic engineering will likely emerge next years [35].
derivatives have been described in the literature as solubi-
lizers capable of enhancing the loading capacity of lipo- POLYMERIC NANOPARTICLES AND MICROPAR-
somes and microparticles. Its main purpose was to combine TICLES
some advantages of CDs (e.g. increased drug solubility) with Over the past few decades, there has been considerable
some advantages of liposomes (e.g. targeting of drugs to interest in developing biodegradable nanosized particles as
452 Current Drug Targets, 2004, Vol. 5, No. 5 Mainardes and Silva

effective drug delivery devices because they have the ability delivery strategies are based on binding drugs with magnetic
to deliver a wide range of drugs to varying areas of the body fluids (MFs) that concentrate the drug in the site of interest.
for sustained periods of time [10-11, 41]. More recently, they MFs are stable colloidal suspensions of magnetic nanopar-
have even been found to be efficient vectors to deliver ticles (MNPs) dispersed in organic or inorganic liquid
synthetic small fragments of DNA into cells [42]. carriers [51-54]. MNPs are nanometer-sized ferrite- or
Nanoparticles are roughly defined as submicron-sized magnetite (Fe 3O4)-based spherical particles [55]. In order to
colloidal systems (vary in size from 10 to 1000 nm) biode- be used for biomedical purposes, MNPs must be pre-coated
gradable or not. Depending upon the process used for the with substances enabling them with biostable, biodegradable,
preparation of the nanoparticles, nanospheres or nanocap- and non-toxic properties. Thus, as biocompatible materials,
sules can be obtained. Nanospheres have a matrix-like struc- MNPs are suitable for drug delivery.
ture, where active compounds can be firmly adsorbed at their MNPs react to a remote magnetic force enabling them to
surface, entrapped or dissolved in the matrix. Nanocapsules be attracted to high magnetic flux density and this feature is
have a polymeric shell and an inner core. In this case, the used for drug targeting. The treatment of solid tumors using
active substances are usually dissolved in the core, but may drugs associated with MNPs could improve efficacy by
also be adsorbed at their surface. Similarly to nanoparticles, increasing the drug concentration at the tumor. In addition,
microparticles are a general name for microcapsules and hysteresis loss in the alternative magnetic field is an impor-
microspheres. The above-mentioned differences between tant feature of MNPs, because it enables effective thermo-
spheres and capsules are similar, but their size ranges from 1 therapy.
to 1000 µm for these carriers [10, 17, 43-44].
Although, a number of techniques have been developed LIPOSOMES AND OTHER LIPID ASSEMBLIES
to prepare nanoparticles and reported to date, the choice of Liposomes are the major drug carriers that contain lipids
the technique depends on the nature of the polymer, the drug, in their functional forms. They are spherical self-closed
the intended use, and the duration of the therapy. In general, structures, composed of well-defined lipid layers which
the methods can be classified into two main categories enclose part of the surrounding solvent into their interior.
according to whether the formation of nanoparticles requires Liposomes have diameters ranging from few nanometers to
a polymerization of dispersed monomers or whether it is several micrometers and they can be composed of one or
achieved directly from an emulsification of natural macro- several concentrically oriented membranes.
molecules or preformed polymers [42, 45]. Many methods
are currently available for the preparation of polymeric nano- They can be classified according to structural features as
particles, such as emulsification-evaporation method [46], large multilamellar liposomes (MLVs), small unilamellar
salting-out procedure [47], and nanoprecipitation method vesicles (SUVs) or large unilamellar vesicles (LUVs),
[48]. depending on their size and the number of lipid bilayers.
Their unique features due to the amphiphilic nature of the
Although various polymers are possible to make lipids make them suitable for many drug delivery strategies
nanoparticles for drug delivery systems in vitro, polymeric [56-58]. The hydrophobic bilayer and the hydrophilic inner
materials used to prepare microspheres and nanoparticles for core enable solubilization or encapsulation of hydrophobic
administration into the human body are significantly limited and hydrophilic drugs. Liposomal drug delivery systems not
to a few kinds of acceptable polymers. Among them, PLA, only enable the delivery of higher drug doses, but also a
PGA and their copolymers such as PLGA are one of the targeting of specific cells, tissues, or organs. Several studies
most used biodegradable polymers to make micro- or based on different drug classes have pointed that harmful
nanoparticles for controlled delivery systems [3, 8-13, 49]. side effects can be reduced or avoided by using liposomes
This is largely due to their biocompatibility and to their instead free administration [59]. In addition, liposomes have
resorbability through natural pathways. Additionally, the been extensively studied as inert carriers for the delivery of
degradation rate and consequently the drug release rate can proteins, nucleic acids, and other biologically active agents
be manipulated by varying the ratio of PLA, increasing the into the cells [60].
hydrophobicity, to PGA, increasing the hydrophilicity [2, 7,
9, 10-13]. However, like all other carrier systems, the application of
liposomes in drug delivery has disadvantages. Interactions
Nanoparticles have been extensively investigated in with lipoproteins, increased free radicals production, and
biomedical areas and, specially, in drug delivery systems for complete saturation of the immune system are only some
drug targeting, because their sizes are also acceptable for typical examples of potentially toxic and adverse effects.
intravenous injection [10-11, 43, 49-50] and their property to
allow ligands binding. The advantages of targeted drug Efficient drug delivery systems based on liposomal
delivery to the specific site of body are in the therapy of approaches need to posses special features. First, good
several disease states such as anticancer treatment, bacterial chemical, coloidal, and biological stability are required.
infection, gene therapy, and viral disease. Therefore, the Second, specific target requires structural and/or chemical
application of nanoparticles for drug targeting in vivo has changes related to functional groups incorporation, like
attracted considerable interest to achieve these aims [49]. carbohydrates, antibodies, ligands, growth factors, or
magnetic nanoparticles, optimizing drug effects.
MAGNETIC NANOPARTICLES The field of liposome targeting has developed rapidly in
Magnetically controlled drug targeting is one of the the past 25 years to the controlled delivery of molecules. The
various recent possibilities of drug delivery. Magnetic-based primary emphasis of the majority of these efforts focused on
Drug Delivery Systems Current Drug Targets, 2004, Vol. 5, No. 5 453

strategies for specific liposome targeting. Among the many delivery approaches [77]. Albumin special features include:
schemes devised for controlled drug delivery, three quite 1) abundance and bioavailability; 2) large circulation on the
different approaches have proved particularly promising. body; 3) several potential binding sites of different affinities.

1). Ligand-Conjugated Liposomes DENDRIMERS


Despite of the evident importance of liposome techno- Dendrimers are polymeric macromolecular complexes
logy, it is widely held that only a minor fraction of liposomes that comprise a series of well-defined branches around an
fuse with the cell membrane. Liposomes with recognition inner core [78]. Dendrimers are produced in an iterative
molecules adsorb to the cells much more effectively. This sequence of well-conducted reaction steps, in which each
approach involves direct targeting of the drug by conjugation additional iteration leads to a higher generation dendrimer
with a ligand having a distinct affinity for a particular organ, (that correspond to the number of layers). Since their
tissue, cell, or sub-cellular structure. Such ligands typically discovery in the early 1980s, dendrimers are the best
include carbohydrates [61], hormones [62], lectins [63], examples of controlled hierarchical synthesis allowing the
peptides [64], antibodies [65], and receptor antagonists [66]. generation of complex systems. Special features concerning
dendrimers synthesis include the possibility to finely control
Liposomes bounded with antibodies (immunoliposomes)
the size, composition, and chemical reactivity in a manner
adsorb to the cells much more effectively than free lipo-
that make it works like a machine.
somes. This approach proved to be efficient in delivering
chemotherapeutic [65, 67] and antiviral [68] drugs. Virtually any polymer could be used to generate a dendri-
mer. The first structure that was well characterized was the
2). Magnetism-Based Liposomes polyamidoamine dendrimers or PAMAM dendrimers [79]
which are constructed using a reiterative sequence of steps
Magnetically controlled liposomes, so commonly called
and products up to generation 10 have been obtained. In the
magnetoliposomes, are aimed at concentrating drugs at a
last 10 years dozens of interesting dendrimer types have
specific target site, with the aid of a well-defined magnetic
appeared and investigated displaying suitable applicability.
field [69]. Magnetoliposomes consist of a liposome filled
with nanosized magnetic nanoparticles. A combination of
MICROMACHINES AND NANOMACHINES
immunoliposomes and magnetoliposomes has been recently
suggested displaying many advantages over either method About four decades ago, the quantum theorist and Nobel
alone, including increased specificity and reduced side Laureate Richard Feynman became the first individual to
effects [70]. suggest the miniaturization of structures. In his famous
lecture titled “There's Plenty of Room at the Bottom.”
3). Photosensitizer-Based Liposomes (1959), he examined the infant field of materials nanoscience
speculating at how small a machine could be made and still
Photodynamic therapy is a promising approach for the be consistent with the known physics laws including quan-
treatment of superficial tumors [71-73]. The therapy involves
tum mechanics. Despite of intense discussions concerning
the systemic administration of a photosensitizer, which after
this topic, there was no consensus until two decades ago.
a predefined time interval is followed by irradiation of the
Drexler [80] applied the term molecular nanotechnology in a
tumor region with a non-thermal light [74]. The net result is
serious unified form to describe advanced precise capabi-
that malignant cells become photochemically eradicated.
lities based on molecular assemblers, proposing devices able
However, like any other anticancer therapy, in photodynamic
to guide chemical reactions by manipulating reactive
therapy, health tissue adjacent to the tumor mass is damaged.
molecules with molecular or atomic levels. In addition, he
A lower extent of undesired lesions to normal tissues can be advanced the proposal that the molecular machinery found in
obtained by associating photodynamic agents with passive or
biological structures demonstrates the feasibility of doing
active targeting vehicles, like liposomes [56]. advanced molecular engineering to produce complex arti-
ficial molecular machines with atomically precise parts of
VIRUS nanometer dimensions.
The so called gene therapy relies on nucleic acid carriers. In that way, micro or nanomachines could act as
Viral vectors have been hypothesized and more recently “intelligent” approaches of drug delivery. The importance of
developed to deliver genes into eukaryotic cells to regulate such methodologies in a near future is undoubtedly and
cellular functions or to express useful proteins [75]. Syn- current efforts have been devoted in this area. However,
thetic virus-like supramolecular vectors involve genetically there is no warranty that all potential risks will be taken into
engineered recombinant viruses that carry the therapeutic account.
gene in their capsid, thus protecting it from enzymatic
degradation. Despite several recent advances in this area, the PHARMACOKINETIC AND PHARMACODYNAMIC
rational and safe design of such strategies requires more CONSIDERATIONS OF DRUG DELIVERY SYSTEMS
studies.
The development of controlled drug delivery is based
PROTEINS practically in to modulate the magnitude and duration of
drug action. To enable optimal design of controlled release
Proteins-based drug delivery approaches have been systems, an understanding of the pharmacokinetics and
suggested for years [76]. Albumin is a plasma protein and pharmacodynamics of the drug is essential.
due to several factors it is the best candidate for drug
454 Current Drug Targets, 2004, Vol. 5, No. 5 Mainardes and Silva

The pharmacokinetic basis for the use of current drugs [9] Vert, M.; Schwach, G.; Engel, R. and Coudane, J. (1998) J.
should ideally be associated to the drug delivery system Control. Release, 53(1-3), 85-92.
[10] Soppimath, K.S.; Aminabhavi, T.M.; Kulkarni, A.R. and
used. The optimization of the dose-time profile of drug at Rudzinski, W.E. (2001) J. Control. Release, 70(1-2), 1-20.
target and non-target sites constitute the major aspect during [11] Hans, M.L. and Lowman, A.M. (2002) Current Opinion in Solid
experimental design of therapeutic strategies. The drug State & Materials Science, 6(4), 319-327.
profile at some target organ or cell site is determined by the [12] Sahoo, S.K.; Panyam, J.; Prabha, S. and Labhasetwar, V. (2002) J.
Control. Release, 82(1), 105-114.
rate of input of drug into and elimination from the body, the [13] Vandervoort, J. and Ludwig, A. (2002) Int. J. Pharm., 238(1-2),
rate at which it is delivered to the target site and the rate at 77-92.
which it is removed from that site. The pharmacokinetics [14] Brannon-Peppas, L. (1995) Int. J. Pharm., 116(1-2), 1-9.
plays an important role in developing of novel drug delivery [15] Anderson, J.M. and Shive, M.S., (1997) Adv. Drug Del. Rev.,
systems, since the introduction of this tool enables us to 28(1), 5-24.
[16] Görner, T.; Gref, R.; Michenot, D.; Sommer, F.; Tran, M.N. and
quantitatively predict the disposition of drugs after any Dellacherie, E. (1999) J. Control. Release, 57(3), 59-268.
modification [81]. [17] Panyam, J. and Labhasetwar, V. (2003) Adv. Drug Del. Rev., 55(3),
329-347.
While the pharmacokinetic consequences of controlled [18] Heller, J.; Barr, J.; Ng, S.Y.; Abdellauoi, K.S. and Gurny, R.
release dosage forms are generally well understood, the (2002) Adv. Drug Del. Rev., 54(7), 1015-1039.
pharmacodynamic aspects are less well studied. The lack of [19] Kumar, N.; Langer, R.S. and Domb, A.J. (2002) Adv. Drug Del.
a well-defined relationship between systemic concentration Rev., 54(7), 889-91.
[20] Gopferich, A. and Tessmar, J. (2002) Adv. Drug Del. Rev., 54(7),
and therapeutic effect, and hence the required drug input rate 911-931.
for optimal response, is the major factor currently limiting [21] Shotts, W.J. and Sievers, A.J. (1974) Biopolymers, 13(12), 2593-
the wider application of controlled drug delivery. In general, 2614.
almost all drugs cause side effects or have extraneous [22] Brown, M.D.; Gray, A.I.; Tetley, L.; Santovena, A.; Rene, J.;
activity in addition to their primary therapeutic function. An Schatzlein, A.G. and Uchegbu, I.F. (2003) J. Control. Release,
93(2), 193-211.
important advantage of specific drug delivery formulations is [23] Singh, M. and O'Hagan, D. (1998) Adv. Drug Deliver. Rev., 34(2-
that by narrowing the range of drug concentrations enables 3), 285-304.
the minimization of the adverse effects associated with [24] Sgouras, D.N.F. and Duncan, R. (1994) STP Pharma Sci., 4(1), 87-
elevated drug concentrations. This pharmacodynamic princi- 94.
[25] Philip, V.A.; Mehta, R.C.; DeLuca, P.P. and Mazumder, M.K.
ple has been widely applied as a means to improve drug Particul. Sci. Technol., 15(3), 245-261.
therapy [81-82]. [26] Bourke, S.L. and Kohn, J. (2003) Adv. Drug Del. Rev., 55(4), 447-
466.
The pharmacodynamics of the desired effects and/or [27] Kwon, H-Y.; Lee, J-Y.; Choi, S-W. and Jang, Y. (2001) Colloid
adverse effects may also be influenced by administration via, Surface A, 182(1-3), 123-130.
especially in cases where the drug is delivered directly to its [28] Bernkop-Schnurch, A.; Guggi, D. and Pinter, Y. (2004) J. Control.
site of action. These factors demonstrate the important influ- Release, 94(1), 177-186.
ence of the mode of administration on the pharmacological [29] Cox, D.S.; Raje, S.; Gao, H.L.; Salama, N.N. and Eddington, N.D.
(2002) Pharmaceut. Res., 19(11), 1680-1688.
and clinical outcomes. In addition, they show the need to [30] Thanou, M.; Verhoef, J.C. and Junginger, H.E. (2001) Adv. Drug
include these pharmacodynamic considerations in all stages Deliver Rev., 52(2), 117-126.
from drug development to the optimization of their clinical [31] Borchard, G. and Junginger, H.E. (2001) Adv. Drug Del. Rev.,
use [82-83]. 52(2), 103.
[32] Hirayama, F. and Uekama, K. (1999) Adv. Drug Del. Rev., 36(1),
125-141.
CONCLUDING REMARKS AND FUTURE PERSPEC- [33] Loftssona, T. and Jarvinen, T. (1999) Adv. Drug Del. Rev., 36(1),
TIVES 59-79.
[34] Duchene, D.; Ponchel, G. and Wouessidjewe, D. (1999) Adv. Drug
Despite evident recent advances in drug delivery Del. Rev., 36(1), 29-40.
technologies, a range of new possibilities have been opened [35] Kakizawa, Y. and Kataoka, K. (2002) Adv. Drug Del. Rev., 54(2),
almost all months and need to be investigated. Studies have 203-222.
[36] Lavasanifar, A.; Samuel, J. and Kwon, G.S. (2002) Adv. Drug Del.
been devoted to answer several questions regarding physical Rev., 54(2), 169-190.
and chemical aspects of deliver devices and to better [37] Kwon, G.S., Okano, T. (1996) Adv. Drug Del. Rev., 21(2), 107-
understand the whole of them in biological systems. 116.
[38] Kwon, G.S. (2002) Adv. Drug Del. Rev., 54(2), 167.
REFERENCES [39] Torchilin, V.P. (2002) Adv. Drug Del. Rev., 54(2), 235-252.
[40] Trubetskoy, V.S. (1999) Adv. Drug Del. Rev., 37(1-3), 81-88.
[1] Uhrich, K.E.; Cannizzaro, S.M.; Langer, R.S. and Shakeshelf, K.M. [41] Mu, L. and Feng, S.S. (2002) J. Control. Release, 80(1-3), 129-
(1999) Chem. Rev., 99(11), 3181-3198. 144.
[2] Pillai, O. and Panchagnula, R. (2001) Curr. Opin. Chem. Biol., [42] Couvreur, P.; Couarraze, G.; Devissaguet, J.P. and Puisiex, F.
5(4), 447-451. (1996) in Microencapsulation: Methods and Industrial
[3] Jain, R.A. (2000) Biomaterials, 21(23), 2475-2490. Applications, (Marcel Dekker), New York, pp. 183-184.
[4] Polakovic, M.; Görner, T.; Gref, R. and Dellacherie, E. (1999) J. [43] Allemann, E.; Leroux, J.C. and Gurny, R. (1998) Adv. Drug Del.
Control. Release, 60(2-3), 169-177. Rev., 34(2-3), 171-189.
[5] Faisant, N.; Siepmann, J. and Benoit, J.P. (2002) Eur. J. Pharm. [44] Nishioka, Y. and Yoshino, H. (2001) Adv. Drug Del. Rev., 47(1),
Sci., 15(4), 355-366. 55-64.
[6] Dunne, M.; Corrigan, O.I. and Ramtoola, Z. (2000) Biomaterials, [45] Quintanar-Guerrero, D.; Fessi, H.; Alléman, E.; Doelker, E. (1996)
21(16), 1659-1668. Int. J. Pharm., 143, 133-141.
[7] von Burkersroda, F.V.; Schedl, L. and Göpferich, A. (2002) [46] Gurny, R.; Peppas, N.A.; Harrington, D.D. and Banker, G.S. (1981)
Biomaterials, 23(21), 4221-4231. Drug Dev. Ind. Pharm., 7(1), 1-25.
[8] Park, T.G. (1995) Biomaterials, 16(15), 1123-1130. [47] Allémann, E.; Gurny, R.; Doelker, E. (1992) Int. J. Pharm., 87,
247.
Drug Delivery Systems Current Drug Targets, 2004, Vol. 5, No. 5 455

[48] Fessi, H.; Puisieux, H.; Devissaguet, J.P.; Ammoury, N. Benna, S. [64] Torchilin, V.P.; Levchenko, T.S.; Rammohan, R.; Volodina, N.;
(1989) Int. J. Pharm., 55. Papahadjopoulos-Sternberg, B. and D'Souza, G.G.M. (2003) P.
[49] Jeon, H.J.; Jeong, Y.L.; Jang, M.K.; Park, Y.H.; Nah, J.W. (2000) Natl. Acad. Sci. USA, 100(4), 1972-1977.
Int. J. Pharm., 207, 99-108. [65] Koning, G.A.; Kampsm J.A.A.M. and Scherphof, G.L. (2002)
[50] Labhasetwar,V.; Song ,C.; Levy ,R.J. (1997) Adv. Drug Del. Rev., Cancer Detect. Prev,, 26(4), 299-307.
24, 63-85. [66] Lakkaraju, A.; Dubinsky, J.M.; Low, W.C. and Rahman, Y.E.
[51] Freitas, M.L.L.; Silva, L.P.; Freitas, J.L.; Azevedo, R.B.; Lacava, (2001) J. Biol. Chem., 276(34), 32000-32007.
Z.G.M.; de Bittencourt, P.I.H.; Curi, R.; Buske, N. and Morais, [67] Xu, L.; Huang, C.C.; Huang, W.Q.; Tang, W.H.; Rait, A.; Yin,
P.C. (2003) J. Appl. Phys., 93(10), 8453-8455. Y.Z.; Cruz, I.; Xiang, L.M.; Pirollo, K.F. and Chang, E.H. (2002)
[52] Freitas, M.L.L.; Silva, L.P.; Azevedo, R.B.; Garcia, V.A.P.; Mol. Cancer Ther., 1(5), 337-346.
Lacava, L.M.; Grisolia, C.K.; Lucci, C.M.; Morais, P.C.; Da Silva, [68] Ho, R.J.Y.; Rouse, B.T. and Huang, L. (1987) Biophys. J. , 51(2),
M.F.; Buske, N.; Curi, R. and Lacava, Z.G.M. (2002) J. Magn. A115.
Magn. Mater., 252(1-3), 396-398. [69] Morais, P.C.; Neto, K.S.; Gravina, P.P.; Figueiredo, L.C.; Da Silva,
[53] Bakuzis, A.F.; Neto, K.S.; Silva, L.P.; Azevedo, R.B. and Morais, M.F.; Lacava, Z.G.M.; Azevedo, R.B.; Silva, L.P. and De Cuyper,
P.C. (2001) J. Appl. Phys., 90(2), 891-895. M. (2002) J. Magn. Magn. Mater., 252(1-3), 418-420.
[54] Lacava, B.M.; Azevedo, R.B.; Silva, L.P.; Lacava, Z.G.M.; Neto, [70] Higuchi, A.; Yamamiya, S-I.; Yoon, B.O.; Sakurai, M. and Hara,
K.S.; Buske, N.; Bakuzis, A.F. and Morais, P.C. (2000) Appl. Phys. M. (2003) J. Biomed. Mater. Res. A, 60(1), 34-42
Lett., 77(12), 1876-1878. [71] Tedesco, A.C.; Rotta, J.C.G. and Lunardi, C.N. (2003) Curr. Org.
[55] Morais, P.C.; Garg, V.K.; Oliveira, A.C.; Silva, L.P.; Azevedo, Chem., 7(2), 187-196.
R.B.; Silva, A.M.L. and Lima, E.C.D. (2001) J. Magn. Magn. [72] Lunardi, C.N.; Rotta, J.C.G. and Tedesco, A.C. (2003) J. Porphyr.
Mater., 225(1-2), 37-40. Phthalocya, 7(7), 493-499.
[56] Nunes, S.M.T.; Sguilla, F.S. and Tedesco, A.C. (2004) Braz. J. [73] Rossa, M.M.; Rocha-e-Silva, T.A.A.; Terruggi, C.H.B.; Tedesco,
Med. Biol. Res., 37(2), 273-284. A.C.; Selistre-de-Araujo, H.S.; Borissevich, I.E. and Degterev, I.A.
[57] Martini, A. and Ciocca, C. (2003) Expert. Opin. Ther. Pat., 13(12), (2003) Pharmacol. Res., 48(4), 369-375.
1801-1807. [74] Betz, C.S. and Leunig, A. (2004) HNO, 52(2), 175-190.
[58] Redziniak, G. (2003) Pathol. Biol., 51(5), 279-281. [75] Hughes, R.M. (2004) J. Surg. Oncol., 85(1), 28-35.
[59] Hrynyk, R.; Storm, G.; Metselaar, B. and Langner, M. (2003) Pol. [76] Vyas, S.P.; Bhatnagar, S.; Gogoi, P.J. and Jain, N.K. (1991) Int. J.
J. Pharmacol., 55(6), 1063-1070. Pharm., 69(1), 5-12.
[60] Semple, S.C.; Klimuk, S.K.; Harasym, T.O.; Dos Santos, N.; [77] Wunder, A.; Muller-Ladner, U.; Stelzer, E.H.K.; Funk, J.;
Ansell, S.M.; Wong, K.F.; Maurer, N.; Stark, H.; Cullis, P.R.; Neumann, E.; Stehle, G.; Pap, T.; Sinn, H.; Gay, S. and Fiehn, C.
Hope, M.J. and Scherrer, P. (2001) BBA-Biomembranes, 1510(1-2), (2003) J. Immunol., 170(9), 4793-4801.
152-166. [78] Boas, U. and Heegaard, P.M.H. (2004) Chem. Soc. Rev., 33(1), 43-
[61] Yamazaki, N.; Jigami, Y.; Gabius, H.J. and Kojima, S. (2001) 63.
Trends Glycosci. Glyc., 13(71), 319-329. [79] Yoo, H.; Sazani, P. and Juliano, R.L. (1999) Pharmaceut. Res.,
[62] Ogawa, Y.; Kawahara, H.; Yagi, N.; Kodaka, M.; Tomohiro, T.; 16(12), 1799-1804.
Okada, T.; Konakahara, T. and Okuno, H. (1999) Lipids, 34(6), [80] Drexler, K.E. (1981) P. Natl. Acad. Sci. USA, 78(9), 5275-5278.
643. [81] Hoffman, A. (1998) Adv. Drug Del. Rev., 33(2-3), 185-199.
[63] Gabor, F. and Wirth, M. (2003) STP Pharma. Sci., 13(1), 3-16. [82] Hoffman, A.; Danenberg, H.D.; Katzhendler, I.; Shuval, R.; Gilhar,
D. and Friedman, M. (1998) J. Control. Release, 54(1), 29-37.
[83] Levy, G. (1998) Adv. Drug Del. Rev., 33(2-3), 201-206.

Received: March 19, 2004 Accepted: March 23, 2004

You might also like