Diagnoza raka mózgu i terapia z nanoplatforms.pdf

(430 KB) Pobierz
doi:10.1016/j.addr.2006.09.012
Advanced Drug Delivery Reviews 58 (2006) 1556
1577
www.elsevier.com/locate/addr
Brain cancer diagnosis and therapy with nanoplatforms
Yong-Eun Lee Koo a , G. Ramachandra Reddy b , Mahaveer Bhojani c ,
Randy Schneider d , Martin A. Philbert d , Alnawaz Rehemtulla c ,
Brian D. Ross e , Raoul Kopelman a,
a Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
b Molecular Therapeutics, Inc., Ann Arbor, MI 48109, USA
c Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
d Department of Environmental Sciences, University of Michigan, Ann Arbor, MI 48109, USA
e Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
Received 9 August 2006; accepted 13 September 2006
Available online 28 September 2006
Abstract
Treatment of brain cancer remains a challenge despite recent improvements in surgery and multimodal adjuvant therapy.
Drug therapies of brain cancer have been particularly inefficient, due to the blood brain barrier and the non-specificity of the
potentially toxic drugs. The nanoparticle has emerged as a potential vector for brain delivery, able to overcome the problems of
current strategies. Moreover, multi-functionality can be engineered into a single nanoplatform so that it can provide tumor-
specific detection, treatment, and follow-up monitoring. Such multitasking is not possible with conventional technologies. This
review describes recent advances in nanoparticle-based detection and therapy of brain cancer. The advantages of nanoparticle-
based delivery and the types of nanoparticle systems under investigation are described, as well as their applications.
© 2006 Elsevier B.V. All rights reserved.
Keywords: Nanoparticles; Blood brain barrier; Drug delivery; MRI; Chemotherapy; Photodynamic therapy; Targeting
Contents
1. Introduction .................................................... 1557
2. Drug delivery methods for the brain ....................................... 1559
2.1. Chemical modification of a drug and prodrugs .............................. 1559
2.2. Temporary disruption of the BBB ..................................... 1559
2.3. Local delivery into brain.......................................... 1559
This review is part of the Advanced Drug Delivery Reviews theme issue on Particulate Nanomedicines Vol. 58/14, 2006.
Corresponding author. Tel.: +1 734 764 7541; fax: +1 734 936 2778.
E-mail address: kopelman@umich.edu (R. Kopelman).
0169-409X/$ - see front matter © 2006 Elsevier B.V. All rights reserved.
207684776.001.png 207684776.002.png
Y.-E.L. Koo et al. / Advanced Drug Delivery Reviews 58 (2006) 1556 1577
1557
2.4. Convection-enhanced delivery (CED) ................................... 1560
2.5. Carrier/receptor-mediated delivery..................................... 1560
3. Nanoparticle delivery system ........................................... 1560
3.1. Nanoparticle platform ........................................... 1561
3.2. Nanoparticle synthesis and characterization ................................ 1562
4. Magnetic nanoparticles for MRI ......................................... 1563
4.1. Iron oxide core surface-coated with polymer ............................... 1563
4.2. Nanoparticles with incorporated iron oxide ................................ 1564
5. Dual imaging nanoparticles (MRI and optical imaging) ............................. 1565
6. Nanoparticles for chemotherapy ......................................... 1565
6.1. Solid lipid nanoparticles (SLNs)...................................... 1565
6.2. Poly(butylcyanoacrylate) (PBCA) nanoparticles.............................. 1566
7. Targeted multi-functional PAA nanoparticles for PDT and MRI ......................... 1567
7.1. In vitro targeting.............................................. 1568
7.2. MRI .................................................... 1569
7.3. PDT .................................................... 1569
7.4. Bioelimination study ........................................... 1570
8. Conclusions.................................................... 1571
Acknowledgements .................................................. 1572
References ....................................................... 1572
1. Introduction
brain tumors, however, do not readily allow quantita-
tion of the actual tumor volume since a lot of extra-
cellular water (edema) can build up around the tumor
site, making exact discrimination of tumor margins
difficult. Moreover, the delivery of contrast agents is
inefficient, due to the blood
Brain tumors constitute a profound and unsolved
clinical problem although significant strides have been
made in the treatment of many other cancer types. The
incidence of primary brain tumors in the United States
has been estimated at approximately 43,800 per year
[1
brain barrier (BBB). The
BBB is a very specialized system of endothelial cells
that separates the blood from the underlying brain
cells, providing protection to brain cells and preserving
brain homeostasis. The use of contrast agents often
allows estimates of tumor domains from the largest
cross-sectional area of contrast enhancement, indicat-
ing a compromised BBB. However, the contrast agents
tend to diffuse away from the vessel, making precise
measurements of the location of the disrupted BBB
somewhat displaced. Finally, even in a tumor
surrounded by an extensive zone of edema, there are
most likely regions of infiltrating tumor cells which are
not apparent. Therefore imaging is typically used to
locate and stage neoplasm and visualize a tumor before
biopsy or at the time of surgery [4] .
The current practice of waiting for altered neuro-
logical function, neurological exam and pathological/
microscopic evaluation/confirmation of the malignan-
cy usually requires that the tumor (benign or malig-
nant) develops either a significant mass or potential
for migration in the neuraxis before invasive surgical
3] and 18,500 of these are expected to be ma-
lignant. Currently brain tumors account for at least
12,690 deaths in the United States yearly and are the
most common cause of cancer-related death for chil-
dren 0
3] .
The earliest stages of intracranial cancer remain
difficult to detect and treat. This problem is confound-
ed by the location of several brain tumors that lie
adjacent to or within anatomical structures critical for
basic motor, cognitive, reflexive and other functions.
As with most other tumors, early detection and reme-
diation correlates with a positive prognosis. Currently
an invasive biopsy is the preferred method to confirm
the diagnosis of cancer as it can provide information
about histological type, classification, grade, potential
aggressiveness and other information that may help
determine the best treatment. Modern imaging techni-
ques such as CT, PET, ultrasound and MRI are rapidly
emerging as standards in the detection of tumors and
cancers. These imaging scans of malignant human
14 years of age [1
1558
Y.-E.L. Koo et al. / Advanced Drug Delivery Reviews 58 (2006) 1556 1577
or non-invasive neuroradiological therapies are
invoked.
Treatment of brain tumors, therefore, has histori-
cally consisted of surgery followed by adjuvant
therapy such as radiation therapy, chemotherapy and
photodynamic therapy (PDT). Despite recent improve-
ments in surgical and adjuvant therapy for brain
tumors, the multimodality approach currently used in
the treatment of malignant brain tumors does not
produce a meaningful improvement in patient outcome
[5] . Each treatment modality has limiting factors, as
stated below.
Surgery is invasive but currently the primary mode
of treatment for the vast majority of brain tumors due
to difficulties in finding a tumor at early stages [6] .
One of the greatest challenges in brain tumor surgery is
achieving a complete resection without damaging cru-
cial structures near the tumor bed. Unfortunately, neo-
plastic tissue that is easily detected radiographically, is
virtually indistinguishable from normal brain. While
surgery is the recommended initial treatment for brain
tumors, it is rarely capable of eradicating all tumor
cells [7] . Furthermore, surgery is not an option when
eloquent structures are likely to be damaged during a
resection. To address the inability of current surgical
techniques to reliably eradicate residual or unresect-
able tumor, adjuvant radiation and chemotherapy regi-
mens have been developed.
Radiation therapy, chemotherapy and PDT are non-
invasive and often used as adjuvant therapy after sur-
gery but may also be effective for curing early-stage
tumors. Radiation therapy usually results in a delayed,
but well-documented, decline in cognitive function
in adults, in addition to posing the risk of secondary
malignancy in the irradiated area [8] . In children, ra-
diation therapy is known to interfere with brain de-
velopment [9] . The efficiency of radiation therapy is
often hindered by diffusely invasive characteristics of
brain tumors as well as the emergence of radiation-
resistant populations.
Most chemotherapeutic agents have a low thera-
peutic index. They are toxic and can affect not only
cancer cells but also healthy cells, which leads to severe
systemic side effects, generally resulting in morbidity
or mortality in the patient. The chemotherapeutic treat-
ment of brain cancer is further restricted due to the
ability of the BBB to exclude a wide range of anti-
cancer agents. Another limiting factor is the develop-
ment of multi-drug resistance (MDR) by the cancer
cells. A combinational chemotherapy, i.e. the use of
more than one drug, is a common practice in clinical
oncology. However, cancer cells often develop resis-
tance against a wide variety of chemotherapeutic drugs,
due to the very effective drug efflux system P-glyco-
protein or multi-drug resistance-associated protein
(MDRP) [10,11] . The P-glycoprotein is an ATP-de-
pendent transporter responsible for the cellular extrusion
of a number of drugs. It is expressed in many tis-
sues, including the luminal membrane of the cerebral
endothelium.
The combination of chemotherapy and radiation
therapy has been implemented with variable success in
adult brain tumors [12] but also carries significant
treatment-related morbidity. Moreover, the improve-
ments in outcome demonstrated with the use of
combination therapy are minimal: a prospective ran-
domized controlled study on temozolomide, the most
effective and best tolerated agent for treating gliomas,
demonstrated an increase in the median two-year
survival of only 2.5 months in patients with newly
diagnosed glioblastoma receiving radiation and temo-
zolomide, compared to those receiving radiation thera-
py alone [13] .
PDT involves the delivery of photosensitizers (PS)
such as Photofrin ® to tumors, combined with local
excitation by the appropriate wavelength of light,
resulting in the production of singlet oxygen and other
reactive oxygen species which initiate apoptosis and
cytotoxicity in many types of tumors, with minimal
systemic toxicity. PDT has emerged as a promising
method for overcoming some of the problems inherent
in classical cancer therapies [14
25] . Recently it was reported that
PDT of primary and recurrent gliomas resulted in an
increase in patient median survival [26] . The efficacy
of PDT for brain cancer is also limited by the BBB
and MDR, just like chemotherapy, as it requires the
delivery of the PS to the brain.
17] . It is more selec-
tive and less toxic than chemotherapy because the drug
is not activated until the light is delivered. PDT was
initially applied clinically to cutaneous and bladder
malignancies that can easily be exposed to light. How-
ever, PDT is also an interesting approach for the treat-
ment of malignant gliomas, as it offers a localized
treatment approach. Several investigations have been
made on the application of PDT for the treatment of
brain tumors [18
Y.-E.L. Koo et al. / Advanced Drug Delivery Reviews 58 (2006) 1556 1577
1559
The therapeutic efficacy of chemotherapy and PDT
can be greatly improved by efficient delivery of the
drugs to the specific tumor location. The recent mo-
lecularly-targeting approach allows the medical inter-
vention to affect only cancer cells but not the normal
cells, based on molecular recognition processes
(ligand
category. Therefore, delivery of drugs to the brain
needs a special strategy to bypass the BBB and thus to
achieve high intratumoricidal drug concentrations
within the central nervous system (CNS). Various
strategies have been explored for manipulating the
BBB, as summarized below.
receptor or antibody
antigene interaction)
31] . This innovative approach is inherently
different from classical modalities. It has the potential
to improve the therapeutic efficacy or imaging contrast
enhancement, by increasing the amount of therapeutic
or contrast agents delivered to the specific site, and to
minimize toxicity, or imaging background signal, by
reducing systemic exposure. The promise of the
molecularly targeted approach in imaging is that one
may be able to obtain dramatic contrast enhancement
so as to detect the tumor at an earlier stage than
possible by current methods, with sensitivity good
enough to avoid an invasive biopsy. Since the specific
molecular signature of one brain tumor may be dif-
ferent from that of another, and can not be differ-
entiated based upon traditional anatomical imaging,
the ability to diagnose brain tumors based on their
genetic presentation, in a targeted manner, would be of
great value. By the same notion, the approach of
delivering a therapeutic agent in a targeted manner
should give clinicians the ability to treat cancer or to
manage it as a chronic disease, thus preventing it from
progressing to its later, more virulent stages. Towards
more efficient chemotherapeutic treatment of brain
cancer, there have been continuous efforts to develop
special delivery methods designed to overcome the
BBB. Proper combination of these methods and the
molecular-targeting approach should be a key factor
for achieving an improved therapeutic efficacy.
2.1. Chemical modification of a drug and prodrugs
Lipid solubility is a key factor in enhancing passive
diffusion into the BBB. Chemical modification of the
drug itself into a more lipophilic and neutral form as
well as a prodrug approach have been investigated.
The prodrug approach involves the administration of the
drug in a form that is inactive or weakly active, but
readily able to penetrate the BBB and then to be con-
verted into the active form within the brain. Both
approaches have pharmacokinetic difficulties, as lipidi-
zation may bring in undesirable pharmacokinetic ef-
fects, such as increased uptake by the reticuloendothelial
system and increasing non-specific plasma protein
binding when administered intravenously [32] .For
example, several lipophilic variants of BCNU were
clinically tried but have not shown improved clinical
efficacy over BCNU [33] .
2.2. Temporary disruption of the BBB
The BBB can be permeabilized using either osmotic
disruption by certain hyperosmolar agents, such as
mannitol, or biochemical opening by bradykinin ana-
logs such as RMP-7. This leads to a reversible opening
of the tight junction, but is not specific enough to
disallow CNS entry of toxins and unwanted molecules,
thus potentially resulting in significant damage. The
experimental studies have clearly shown an increased
penetration of the drug into the brain parenchyma, but
the clinical studies did not show improvement in the
efficacy of the drug with concurrent use of these agents.
Therefore, this has not translated into clinical efficacy
[34] .
2. Drug delivery methods for the brain
In contrast to the open endothelium of the peripheral
circulation, the tightly fused junctions of the cerebral
capillary endothelium, the anatomic basis for the BBB,
essentially form a continuous lipid layer that effectively
restricts free diffusional movement of molecules into
and out of the brain. Only small, electrically neutral,
lipid-soluble molecules (molecular weight up to
500 Da) can penetrate the BBB by passive diffusion
and most chemotherapeutic agents do not fall into this
2.3. Local delivery into brain
This method has been achieved by direct infusion
of a drug via a catheter or implantation of a gel wafer, a
polymer matrix containing a drug. It is, however, a
highly invasive procedure that requires neurosurgery
[27
1560
Y.-E.L. Koo et al. / Advanced Drug Delivery Reviews 58 (2006) 1556 1577
3 weeks. Clinical trials have
shown that Gliadel wafers can lengthen survival time
and help control symptoms of high grade gliomas for
longer times than surgery and radiotherapy alone [35] .
To date this is the most efficient method of delivery of
drugs into the brain.
key factor for the efficacy of cancer detection and
therapy. The utilization of the nanoparticle as a po-
tential vector for brain or other site-specific delivery
has the following advantages, due to its excellent
engineerability and non-toxicity:
1. The loading/releasing of active agents (drugs/con-
trast agents) can be controlled. The drugs are loaded
into nanoparticles by encapsulation, adsorption or
covalent linkage. The loaded amount is controllable
by changing the size of the nanoparticles or the
number of linkers inside and on the surface of the
nanoparticles. Each nanoparticle can carry a large
amount of molecular therapeutic and/or contrast
agents. Release of the agents may occur by desorp-
tion, diffusion through the NP matrix, or polymer
wall, and/or NP erosion, which can all be controlled
by the type of the nanoparticle's polymer matrix,
i.e., having it become swollen or degradable in the
tumor environment.
2. Specific molecular-targeting factors can be attached
for localized binding to and/or uptake by the tumor
cells, as well as for passage through the blood
2.4. Convection-enhanced delivery (CED)
While invasive it is currently an area of active in-
vestigation for drug delivery to the CNS. This method
utilizes convection so as to supplement diffusion for the
distribution of certain compounds and thus treat much
larger volumes of brain than can be achieved by dif-
fusion alone. The convection results from a simple
pressure gradient and is independent of molecular
weight, resulting in greater pharmacokinetic advan-
tages over systemic administration [34] . The CED
delivery system is currently used in two clinical treat-
ment trials for high grade gliomas [36,37] .
brain barrier when appropriate. It should be noted
that the selective delivery of nanoparticles to tumor
is sometimes achieved due to the
2.5. Carrier/receptor-mediated delivery
tumor
vasculature, which is known as the enhanced per-
meability and retention (EPR) effect [39
leaky
The CNS (or brain) has transport routes that
overcome the BBB by other than passive diffusion,
such as carrier/receptor-mediated influx or transcytosis
[38] , in order to receive essential polar metabolites
such as glucose, amino acids and lipoprotein. These
carriers/receptors can be used to deliver drugs to the
CNS. It requires the discovery and development of
receptor specific ligands, which can be attached directly
to the drug of interest or the drug delivery system, such
as nanoparticle and liposome. This methodology has
been receiving significant attention with the remarkable
development of nanotechnology and non-invasiveness,
compared to the other delivery methods listed above.
The combination of nanoparticles and delivery methods
is especially promising, as shown below.
42] . This
and tumor-specific targeting moieties on the surface
turn the nanoparticles into very efficient delivery
vectors for tumors. Moreover, the use of targeted
nanoparticles can achieve the delivery of large
amounts of therapeutic or imaging agents per tar-
geting biorecognition event, which is a major clini-
cal advantage over simple immunotargeted drugs.
3. A hydrophilic coating can be given to the nano-
particle to provide reduced uptake by the RES,
resulting in both increased delivery of the nano-
particles to tumor sites and reduced toxicity to other
body tissues.
4. The nanoparticle matrix provides protection, for the
active agents, from enzymatic or environmental
degradation.
5. The nanoparticles can alleviate the problem posed by
theMDR of cancer cells against many drugs; done by
masking the drugs entrapped within the nanoparti-
cles. This feature may enhance the delivery of drugs
that are normally excluded from tumors.
3. Nanoparticle delivery system
The ability to deliver effective concentrations of
contrast or therapeutic agents selectively to tumors is a
and special equipment. To date, Gliadel ® Wafer
(BCNU-loaded biodegradable polymer) is the only
wafer approved for clinical use in the US; it releases
the chemotherapy drug directly into the brain as the
polymer degrades over 2
Zgłoś jeśli naruszono regulamin