Sie sind auf Seite 1von 12

11

Noninvasive Visualization of
In Vivo Drug Delivery of
Paramagnetic Polymer
Conjugates with MRI
Zheng-Rong Lu, Yanli Wang, Furong Ye, Anagha
Vaidya, and Eun-Kee Jeong
CONTENTS
11.1 Introduction ....................................................................................................................... 201
11.2 Magnetic Resonance Imaging ........................................................................................... 202
11.2.1 Principles of MRI ................................................................................................ 202
11.2.2 Contrast-Enhanced MRI...................................................................................... 203
11.3 MR Imaging of Paramagnetic HPMA Copolymer Conjugates........................................ 204
11.3.1 MR Imaging of a Paramagnetic HPMA Conjugate in an Animal
Tumor Model....................................................................................................... 204
11.3.2 MR Imaging of a Paramagnetic HPMA Conjugate for Bone Delivery............. 206
11.4 MR Imaging of Paramagnetic Poly(L-Glutamic Acid) Conjugates ................................. 206
11.4.1 MR Imaging of PGA(Gd-DOTA) Conjugate in an Animal
Tumor Model....................................................................................................... 206
11.4.2 MR Imaging of PGAMce
6
(Gd-DOTA) Conjugate in an Animal
Tumor Model....................................................................................................... 208
11.5 Summary............................................................................................................................ 210
References .................................................................................................................................... 210
11.1 INTRODUCTION
The conjugation of therapeutics to water-soluble biomedical polymers increases the aqueous solu-
bility of hydrophobic drugs, prolongs in vivo drug retention time, reduces systemic toxicity, and
enhances therapeutic efcacy.
1,2
Polymer drug conjugates can preferentially accumulate in solid
tumor tissues due to the hyperpermeability of tumor blood vessels or the so-called enhanced
permeability and retention (EPR) effect.
3,4
Polymer drug conjugates can also down-regulate or
overcome multiple drug resistance.
5,6
Because of these unique properties, polymer drug conjugates
exhibit higher therapeutic efcacy than the corresponding therapeutics alone. Several polymer drug
conjugates are currently used in clinical cancer treatment, and more are in the pipeline of clinical
201
q 2006 by Taylor & Francis Group, LLC
development. For example, styrenemaleic acid copolymer neocarzinostatin conjugate (SMANCS)
is used for liver cancer treatment in Japan;
7
pegylated adenosine deaminase
8
and asparaginase
9,10
are used for enzyme replacement therapy for immunodeciency and for the treatment of acute
lymphoblastic leukemia, respectively. Many other polymer drug conjugates, including pegylated
interferon a,
11
PEGcamptothecin conjugate,
12
poly(L-glutamic acid) paclitaxel conjugate,
13
poly[N-(2-hydroxypropyl)methacrylamide] (PHPMA)-cis-platinate conjugate,
14
and PHPMA
doxorubicin conjugate
15,16
are in various phases of clinical trials.
The in vivo behavior, including pharmacokinetics, biodistribution, drug delivery efciency and
therapeutic efcacy, of polymer drug conjugates has been traditionally evaluated by using blood
and urine sampling, biopsy-based methods, and symptom-based observations. These methods are
sometimes invasive and cannot accurately provide real time information on the interaction of
polymers with various organs and tissues, the targeting and delivery efciency of the drug delivery
system, and therapeutic response. A large number of animals are also required in preclinical
development. Sometimes, the data obtained by conventional biopsies may be misleading, which
might be one of the causes of the efcacy discrepancy between preclinical development in animal
models and human clinical trials and the failure of some clinical trials due to improper selection of
drug candidates.
In vivo drug delivery by polymer drug conjugates involves circulation of the conjugates in the
blood; interaction with the major organs including the liver, heart, lungs, spleen and kidneys, etc.;
transport to target tissue; and uptake by target cells and drug release. Direct and continuous
evaluation of drug delivery efciency and tissue interaction of polymeric drug conjugates is critical
to develop more efcacious drug delivery systems. Recent advancements in biomedical imaging
technology have provided the essential tools for noninvasive and continuous in vivo evaluation for
drug delivery. Nuclear medicine, including positron emission tomography (PET) and single photon
emission computed tomography (SPECT), is a clinical imaging modality with high detection
sensitivity (ca. 10
K9
M). Magnetic resonance imaging (MRI) is a noninvasive clinical imaging
modality with high spatial resolution. Both imaging modalities are able to noninvasively visualize
in vivo drug delivery with polymeric drug conjugates. The polymer conjugates can be labeled with
imaging probes and noninvasively and continuously monitored with the imaging modalities.
The number of experimental animals can be dramatically reduced in the preclinical development
of the conjugates. Biomedical imaging will provide real-time information of the in vivo behavior of
polymeric drug conjugates. In this chapter, we will focus on noninvasive visualization of in vivo
drug delivery with paramagnetic polymeric conjugates and contrast-enhanced MRI.
11.2 MAGNETIC RESONANCE IMAGING
11.2.1 PRINCIPLES OF MRI
MRI is a clinical imaging modality that produces three-dimensional anatomic images with high
spatial resolution and provides in vivo physiological properties including physiochemical infor-
mation, ow diffusion, and motion in the tissues. The physics of MRI is complicated and has been
detailed in a number of monographs.
17,18
The fundamental principles of MRI are briey reviewed
in this section. A normal human adult has approximately 60% of the body weight as water. Water
protons have a magnetic moment and the orientations of the magnetic moments are random in the
absence of an external magnetic eld. When a patient is placed in a MRI scanner, the proton
magnetic moments align either along or against the static magnetic eld (B
0
) of the scanner and
create a net magnetization pointing in the direction of the main magnetic eld of the scanner.
The magnitude of the magnetization is proportional to the external magnetic eld strength as well
as the amount of protons.
Nuclear magnetic resonance (NMR) measures the change of the magnetization by applying
radiofrequency (RF) pulses. When an RF pulse is applied to create an oscillating electromagnetic
Nanotechnology for Cancer Therapy 202
q 2006 by Taylor & Francis Group, LLC
eld (B
1
) perpendicular to the main eld, the longitudinal magnetization is tipped away from the
static magnetic eld and deviates from the equilibrium state (in longitudinal space). A perfect 908
RF pulse ips the total longitudinal magnetization onto the transverse plane. Immediately after
excitation with the RF pulse, the transverse magnetization then undergoes the decay process.
The decaying transverse magnetization induces an NMR signal as an electromotive force (emf)
on an RF coil. Simultaneously, the nuclear spin system approaches toward the equilibrium state
with a characteristic recovery time T
1
, which depends upon the physical and chemical environment
of the molecules.
Longitudinal T
1
relaxation involves the return of protons from the high energy state back to
equilibrium by dissipating their excess energy to their surroundings. The process is also called spin-
lattice relaxation. Transverse (T
2
) relaxation involves the transfer of the spin angular momentum
among the protons via the interactions such as dipoledipole interaction. The process is called
spinspin relaxation. In biological systems, the T
2
relaxation time is typically shorter than T
1
relaxation time. Because the main magnetic eld of MRI scanners is not perfectly homogenous,
the inhomogeneity of the magnetic eld also causes dephasing of individual magnetizations of
protons, resulting in more rapid loss of transverse magnetization. The process is called
T

2
relaxation.
In MR imaging, the position dependent frequency and phase are encoded into the transverse
magnetization, and the measured signals are Fourier transformed to construct the spatial map
(image). MR images are created based on the proton density, or T
1
and T
2
relaxation rates, and
ow and diffusion properties in different tissues. These parameters vary between different tissues
and create image contrast among the tissues. Tissues with a short T
1
relaxation time give a strong
MR signal, resulting in bright images in T
1
-weighted MRI. Tissues with a short T
2
relaxation time
produce a weak MR signal, resulting in dark images in T
2
-weighted MRI.
11.2.2 CONTRAST-ENHANCED MRI
In many cases, the differences between the MR signal intensities of normal and diseased tissues are
not large enough to produce obvious contrast for denitive diagnosis. Contrast agents are used to
enhance the image contrast between normal and diseased tissues to improve diagnostic sensitivity
and specicity.
19,20
MRI contrast agents are either paramagnetic metal chelates or ultrasmall
superparamagnetic iron oxides. These agents interact with surrounding water molecules and
increase the relaxation rates (1/T
1
and 1/T
2
) of water protons. The change of relaxation rate is
linearly proportional to the concentration of contrast agents, [M]:
1=T
i;obs
Z1=T
i;d
Cr
i
M; (11.1)
where i Z1,2. In Equation 11.1, 1/T
i,obs
is the observed relaxation rate with a contrast agent, 1/T
i,d
the relaxation rate without the contrast agent, and r
i
the relaxivity of the contrast agent. Relaxivity is
a measurement of the efciency of a contrast agent to enhance the relaxation rate of water protons
or the efciency to generate image contrast enhancement. Because the uptake of contrast agents
varies between different tissues, MR image contrast is enhanced in the tissue with a high contrast
agent concentration compared to that with a low concentration.
MRI contrast agents approved for clinical applications are mainly stable paramagnetic chelates,
e.g., Gd(III) chelates
2124
and Mn(II) chelates,
25
and ultrasmall superparamagnetic iron oxide
(USPIO).
26
Gd(III) chelates of DTPA, DOTA, or their derivatives are commonly used for
contrast-enhanced MRI. Both Gd(III) and Mn(II) chelates are mainly used as T
1
contrast agents,
whereas USPIO is used as a T
2
contrast agent. These agents signicantly enhance image contrast
between normal tissue and diseased tissue and improve the diagnostic accuracy. Currently, contrast
agents are used in approximately 30% of clinical MRI examinations. Macromolecular Gd(III)
complexes have also been developed as MRI contrast agents.
27,28
These agents have a prolonged
Noninvasive Visualization of In Vivo Drug Delivery of Paramagnetic Polymer Conjugates 203
q 2006 by Taylor & Francis Group, LLC
blood circulation time and preferentially accumulate in solid tumors due to the EPR effect. Macro-
molecular Gd(III) complexes have demonstrated superior contrast enhancement in MR
angiography and cancer imaging in animal models, but are not yet approved for clinical
applications.
Contrast-enhanced MRI is a useful method for noninvasive in vivo evaluation of polymeric
drug delivery systems after the systems are labeled with MRI contrast agents. The real-time
pharmacokinetics and biodistribution of the labeled drug delivery systems can be continuously
visualized by contrast-enhanced MRI. It has a potential to provide accurate four-dimensional
information of in vivo properties of the drug delivery systems.
11.3 MR IMAGING OF PARAMAGNETIC HPMA COPOLYMER CONJUGATES
N-(2-Hydroxypropyl)methacrylamide (HPMA) copolymer is a biocompatible water-soluble poly-
meric carrier for therapeutics.
1
The conjugation of anti-cancer drugs to HPMA copolymers results
in many advantageous features over small molecular therapeutics, including improved water solu-
bility and bioavailability, preferential accumulation of the conjugates in solid tumors, reduced
nonspecic toxicity, and down-regulation of multi-drug resistance.
1,29
A number of HPMA copo-
lymer drug conjugates have been systematically investigated both in vitro and in vivo. Several
HPMA copolymer-anti-cancer drug conjugates are now in various phases of clinical development
for cancer treatment.
30
The incorporation of MRI contrast agents into HPMA copolymer conjugates
allows direct visualization of real-time drug delivery of the conjugates in animal models with
contrast-enhanced MRI. The noninvasive imaging reveals some interesting features of drug
delivery with the conjugates that cannot be obtained with conventional surgery based evaluations.
11.3.1 MR IMAGING OF A PARAMAGNETIC HPMA CONJUGATE
IN AN ANIMAL TUMOR MODEL
HPMA copolymers have been labeled with radioactive probes including
131
I and
99m
Tc for
noninvasive visualization of in vivo drug delivery of the conjugates with g-scintigraphy.
3133
Nuclear medicine is highly sensitive for detecting labeled conjugates, but its poor spatial resolution
cannot provide detailed biodistribution of conjugates in tissues and organs. In comparison, contrast-
enhanced MRI provides clear visualization of the biodistribution of paramagnetically labeled
conjugates with high spatial resolution.
HPMA copolymers were labeled with an MRI contrast agent, Gd-DOTA, and investigated in
an animal tumor model with contrast-enhanced MRI.
34
The structure of a labeled conjugate,
poly[HPMA-co-(MA-Gly-Gly-1,6-hexanediamine-(Gd-DOTA))], is shown in Figure 11.1.
The molecular weight of the polymer conjugate was 25.6 kDa (PDZ1.50) and the Gd content
was 0.33 mmol Gd/g polymer. The T
1
relaxivity of the conjugate was 6.0 mM
K1
s
K1
per attached
Gd(III) chelate at 3 T. Figure 11.2 shows the dynamic, contrast-enhanced, three-dimensional
maximum intensity projection (MIP) (Figure 11.2a) and 2D coronal MR images (Figure 11.2b)
of a mouse bearing a human prostate carcinoma DU-145 xenograft and a Kaposis sarcoma xeno-
graft. The conjugate was administrated intravenously at a Gd equivalent dose of 0.1 mmo1/kg body
weight. The pharmacokinetics and biodistribution of the conjugate were clearly revealed in the
contrast-enhanced MR images.
Strong MRI signal was observed in the heart, liver, kidneys and vasculature in the 3D images at
the initial stage after the injection of the conjugates. The signal intensity decreased over a 60-min
period; meanwhile, the signal intensity in the urinary bladder increased gradually, indicating that
the conjugate was excreted via renal ltration. Signicant contrast enhancement was still visible in
the heart at 60 min. After 22 h, most of the conjugate was cleared from the body except the liver, as
shown in Figure 11.2. The signal intensity in the liver was stronger at 22 h than in the precontrast
image, suggesting signicant interaction of HPMA copolymers with the liver.
Nanotechnology for Cancer Therapy 204
q 2006 by Taylor & Francis Group, LLC
The coronal images cross-sectioning the tumor tissues showed heterogeneous uptake of HPMA
copolymers in two different tumors (Figure 11.2b). The prostate carcinoma had a smaller size, but
stronger MR signal than the Kaposis sarcoma. For Kaposis sarcoma, the contrast enhancement
was mainly observed in the periphery of the tumor tissue and to a lower extent in the inner tumor
tissue. A plausible explanation is that the interstitium of the large tumor was possibly necrotic,
which would limit the access of the conjugate. The results suggested that tumor uptake of HPMA
copolymers may vary from tumor to tumor and with different tumor sizes. It might affect the
efcacy of the polymer conjugates and more detailed studies are needed to understand the
possible correlations.
C
CH
3
O
NH
CH
2
CH OH
CH
3
m
CH
2
C
CH
3
O
NH
CH
2
C
NH
CH
2
n
CH
2
O
O
N N
N N
COO

COO

OOC
Gd
3+
NH(CH
2
)
6
NH C
O
FIGURE 11.1 The structure of poly[HPMA-co-(MA-Gly-Gly-1,6-hexanediamine-(Gd-DOTA))].
FIGURE 11.2 Three-dimensional maximum intensity projection (MIP) (a) and 2D coronal MR images (b) of
the male nude mouse bearing prostate carcinoma DU-145 (left) and human Kaposis sarcoma (SLK) tumor
(right) at different time points. The arrow points to the tumor. Imaging parameters were TRZ7.8 ms,
TEZ2.7 ms, 258 ip angle, and 0.4-mm coronal slice thickness.
Noninvasive Visualization of In Vivo Drug Delivery of Paramagnetic Polymer Conjugates 205
q 2006 by Taylor & Francis Group, LLC
11.3.2 MR IMAGING OF A PARAMAGNETIC HPMA CONJUGATE FOR BONE DELIVERY
Kopecek and coworkers labeled HPMA copolymers (55 kDa) with Gd-DOTA to noninvasively
evaluate macromolecular accumulation in arthritic joints in a rat model. The copolymers were
labeled with uorescein in addition to the paramagnetic metal chelate. The structure of the conju-
gate is shown in Figure 11.3. After intravenous administration of the conjugate in a rat model with
adjuvant-induced arthritis, the biodistribution of copolymers was followed by contrast-enhanced
MRI at various time points post-injection.
35
Contrast-enhanced dynamic MRI clearly revealed the pharmacokinetic properties and biodis-
tribution of the conjugate with high anatomic resolution in the adjuvant-induced arthritic rats
(Figure 11.4). The copolymers had a relatively high molecular weight and a considerable
amount of the copolymers was still circulating in the blood 3 h post-injection. Selective accumu-
lation of HPMA copolymers was gradually shown in the arthritic joints of the adjuvant-induced
arthritis rats. The copolymers were also cleared from the accumulation sites over time as shown by
contrast-enhanced MRI. The uptake and retention of the MR contrast agent labeled polymer
correlated well with the histopathological features of inammation and local tissue damages.
No signicant contrast enhancement was observed in the hind-limb joints of healthy rats in
a control study.
11.4 MR IMAGING OF PARAMAGNETIC POLY(L-GLUTAMIC ACID)
CONJUGATES
Poly(L-glutamic acid) (PGA) is a negatively charged, biocompatible polymer that has been used as
a carrier for drug delivery.
36
It is a homopolymer of L-glutamic acid linked through peptide bonds
and the pendant g-carboxyl groups are suitable to load therapeutic agents via chemical conjugation.
Several poly(L-glutamic acid)-anti-cancer drug conjugates have been developed for cancer treat-
ment.
3739
The conjugation of a MRI contrast agent to PGA enables noninvasive investigation of its
pharmacokinetics and in vivo drug delivery with contrast-enhanced MRI.
11.4.1 MR IMAGING OF PGA(GD-DOTA) CONJUGATE IN AN ANIMAL TUMOR MODEL
Poly(L-glutamic acid)1,6-hexanediamine(Gd-DOTA) conjugate was prepared using a synthetic
procedure similar to that of PGA-cystamine-Gd-DOTA conjugate synthesis
40
to noninvasively
C
CH
3
O
NH
CH
2
CH OH
CH
3
N N
N N
COO
-
COO
- -
OOC
Gd
3+
n
C
CH
3
O
NH
CH
2
CH
2
CH
2
CH
2
m
NH
S
NH
O HO
COOH
O
CH
2
C
CH
3
O
NH
CH
2
CH
CH
2
CH
2
NH
O
FIGURE 11.3 The structure of HPMA-MAFITC copolymers labeled with Gd(III)-DOTA.
Nanotechnology for Cancer Therapy 206
q 2006 by Taylor & Francis Group, LLC
study delivery efciency of the conjugate. The structure of the conjugate is shown in Figure 11.5.
PGA1,6-hexanediamine(Gd-DOTA) conjugates, H a high molecular weight (50 kDa, PDZ
1.12) conjugate and a low molecular weight conjugate (20 kDa, PDZ1.08) were investigated by
contrast-enhanced MRI to study the size effect of conjugates on the efciency of in vivo drug
delivery. The conjugates with different molecular weights had similar Gd content (41.7% for
50 kDa conjugate and 40.2 mol% for 20 kDa conjugate) and T
1
relaxivity (9.44 and 9.20 mM
K
1
s
K1
for 50 and 20 kDa conjugates, respectively). The conjugates were intravenously administered
into female nu/nu athymic mice bearing human breast carcinoma MB-231 xenografts at a dose of
0.07 mmol-Gd/kg. Dynamic contrast-enhanced MRI clearly revealed the size effect of the conju-
gates on their pharmacokinetics and in vivo tumor delivery efciency in the animal model.
Figure 11.6 shows the T
1
-weighted coronal MR images of mice before and at various time
points after injection with the conjugates. Strong contrast enhancement was observed in the heart at
the initial stage post-injection and then gradually faded away for both conjugates. The signal
intensity decreased more rapidly for the low molecular weight conjugate than the high molecular
weight conjugate, validating the concept that the blood circulation of the conjugates is prolonged
N N
N N
COO

COO

OOC
Gd
3+
C
O
CH
CH
2
CH
2
C
NH
NH(CH
2
)
6
NH
O
C
O
n
CH
CH
2
CH
2
C
OH
NH
O
C
m
O
FIGURE 11.5 The structure of poly(L-glutamic acid)1,6-hexanediamine(Gd-DOTA) conjugate.
FIGURE 11.4 The MR images of rats taken at different time points. The acquired images were post-processed
using the maximum intensity projection (MIP) algorithm. P-Gd(III)-DOTA is abbreviated as P-Gd. (A-H) AIA
rat images at baseline (a) and 5 min (b), 1 h (c), 2 h (d), 3 h (e), 8 h (f), 32 h (g), 43 h (h) post-injection of
P-Gd(III)-DOTA. (in) Healthy rat images at baseline (i) and 5 min (j), 1 h (k), 2 h (l), 8 h (m), 48 h (n)
post-injection of P-Gd(III)-DOTA. Arrow points to the diseased joint (Adapted from Wang, D. etal., Pharm.
Res., 21, 1741, 2004. With permission.)
Noninvasive Visualization of In Vivo Drug Delivery of Paramagnetic Polymer Conjugates 207
q 2006 by Taylor & Francis Group, LLC
with increase in molecular weights. The low-molecular-weight conjugate (20 kDa) was cleared
from the blood circulation within 30 min after-injection. The dynamic changes in signal intensity in
the liver correlated well to that in the heart. The enhancement in the liver returned to the back-
ground level 24 h after the injection, indicating low liver uptake of the PGA conjugates.
The contrast enhancement pattern in the tumor tissue was similar to that of HPMA conjugates
in Kaposis sarcoma. Strong signal was observed at tumor periphery and little in the inner tumor
tissue. The dynamic MR images showed that the intensity and duration of tumor enhancement
strongly depended on the size of the conjugates, indicating size-dependent tumor accumulation.
Rapid clearance of the low molecular weight conjugate (20 kDa) from the blood circulation
resulted in low and short accumulation in the tumor tissue. The high molecular weight conjugate
(50 kDa) had a long blood circulation time, resulting in an increased contrast enhancement for at
least 4 h. The enhancement also gradually extended into inner tumor tissue, indicating diffusion of
the conjugate further into the tumor. The results clearly showed that drug delivery with polymeric
conjugate into tumor tissue was a slow process and H conjugates with high molecular weight and
long blood circulation time was more effective for tumor delivery than the conjugate with low
molecular weight. Contrast-enhanced MRI clearly revealed the size effect of the conjugates on their
pharmacokinetics and in vivo drug delivery efciency.
11.4.2 MR IMAGING OF PGAMCE
6
(Gd-DOTA) CONJUGATE
IN AN ANIMAL TUMOR MODEL
Most anti-cancer drugs are lipophilic and the conjugation of lipophilic drugs to a polymeric carrier may
also modify the in vivo behavior of the polymers. Mesochlorin e
6
(Mce
6
) is a lipophilic photosentizer
for photodynamic therapy.
41,42
Aparamagnetic poly(L-glutamic acid) Mce
6
conjugate was prepared to
investigate the pharmacokinetics, biodistribution and drugdeliveryefciency of the PGAMce
6
conju-
gate withcontrast-enhancedMRI.
43
The structure of the conjugate is showninFigure 11.7. APGA1,6-
hexanediamine(Gd-DOTA) conjugate was also prepared from the same poly(L-glutamic acid) as a
control. Mce
6
content was 2.5 mol% in PGAMce
6
1,6-hexanediamine(Gd-DOTA) and Gd content
was 20and29 mol%for PGAMce
6
1,6-hexanediamine(Gd-DOTA) andPGA1,6-hexanediamine
(Gd-DOTA), respectively. The T
1
relaxivity was 8.46 and 8.33 mM
K1
s
K1
for PGAMce
6
1,6-hexa-
nediamine(Gd-DOTA) and PGA1,6-hexanediamine(Gd-DOTA), respectively. The small amount
FIGURE 11.6 Coronal MR slice images of mice bearing MB-231 breast cancer tumors using PGA1,6-hexa-
nediamine(Gd-DOTA) conjugates with different molecular weights. The images were taken at different time
points using a 3D FLASH pulse sequence (1.74-ms TE, 4.3-ms TR, 258 RF tip angle, 120-mm FOV, and
1.6-mm coronal slice thickness).
Nanotechnology for Cancer Therapy 208
q 2006 by Taylor & Francis Group, LLC
of Mce
6
in PGAMce
6
1,6-hexanediamine(Gd-DOTA) signicantly reduced the hydrodynamic
volume and molecular weight distribution of the conjugate. The weight (M
w
) and number (M
n
)
average molecular weights decreased from 101 and 49 kDa of PGA1,6-hexadiamine(Gd-DOTA)
to 49 and 34 KDa for PGAMce
6
1,6-hexadiamine(Gd-DOTA). The lipophilic Mce
6
may alter the
conformation of the polymers, resulting in a smaller hydrodynamic volume for PGAMce
6
1,6-hexa-
diamine(Gd-DOTA).
A contrast-enhanced MRI study demonstrated that PGAMce
6
1,6-hexanediamine(Gd-DOTA)
had signicantly different pharmacokinetics and biodistribution from PGA1,6-hexanediamine(Gd-
DOTA) in female nu/nu athymic mice bearing human breast carcinoma MB-231 xenografts.
Figure 11.8 shows coronal dynamic MR images of mice injected with PGAMce
6
1,6-hexanedia-
mine(Gd-DOTA) and PGA1,6-hexanediamine(Gd-DOTA) at a dose of 0.07 mmol-Gd/kg.
The conjugates demonstrated different dynamic contrast enhancement patterns in the heart and liver.
PGAMce
6
1,6-hexadiamine(Gd-DOTA) had a strong contrast enhancement in the liver and
relatively weak enhancement in the heart, whereas PGA1,6-hexadiamine(Gd-DOTA) exhibited
N N
N N
COO

COO

OOC
Gd
3+
C
O
CH
CH
2
CH
2
C
NH
NH(CH
2
)
6
NH
O
C
O
l
CH
CH
2
CH
2
C
OH
NH
O
C
n
CH
CH
2
C
NH
O
C
m
O
O
NH
=
mesoc (
HN N
NH N
HOOC
CH
2
CH
2
NH
HOOC
O
CH
2
FIGURE 11.7 The structure of PGAMce
6
1,6-hexanediamine(Gd-DOTA).
FIGURE 11.8 Coronal contrast-enhanced MR slice images for PGAMce
6
(Gd-DOTA) conjugate and
PGA(Gd-DOTA) conjugate through the heart (b) before and at 5, 15, 30, 60, and 120 min post-injection.
Noninvasive Visualization of In Vivo Drug Delivery of Paramagnetic Polymer Conjugates 209
q 2006 by Taylor & Francis Group, LLC
strong enhancement in the heart and relatively weak enhancement in the liver. The conjugation of
Mce
6
signicantly altered the pharmacokinetics and biodistribution of the poly(L-glutamic acid). The
results indicated that PGAMce
6
1,6-hexadiamine(Gd-DOTA) had higher liver accumulation than
PGA1,6-hexadiamine(Gd-DOTA). Consequently, it cleared fromthe blood circulation more rapidly
than PGA1,6-hexadiamine(Gd-DOTA), which could also be attributed to the larger
hydrodynamic volume of PGA1,6-hexadiamine(Gd-DOTA).
11.5 SUMMARY
Contrast-enhanced MRI is effective for noninvasive visualization of the real-time pharmacokinetics
and biodistribution of paramagnetically labeled polymer drug conjugates after intravenous admin-
istration. The technique provides three-dimensional anatomic images of soft tissues with high
spatial resolution. The circulation and accumulation of the paramagnetic polymeric conjugates
in organs or tissues result in bright signal for T
1
-weighted images. Dynamic contrast-enhanced
MRI clearly reveals circulation of the conjugates in the blood, interaction with the major organs,
including the liver, heart, kidneys, etc., and accumulation and clearance in the target tissues. It also
provides information on the dynamic changes of the distribution patterns of polymer conjugates in
solid tumor tissues, which cannot be obtained with conventional pharmacokinetic methods. Such a
detailed map of the delivery system deposition within the tissue and its correlation with the local
pathologic features may help to optimize the structure of the polymeric drug conjugate to achieve
high drug delivery efciency. One limitation of contrast-enhanced MRI is the accurate quanti-
cation of the concentrations of the conjugates in the tissues and organs. Several technical factors
control the accurate measurement of concentrations of contrast agents with conventional contrast-
enhanced MRI. For quantitative study of in vivo drug delivery with paramagnetic conjugates, MR
T
1
mapping can be used because T
1
relaxation rate (1/T
1
) is linearly proportional to the concen-
tration of MRI contrast agents. Data acquisition and algorithms to process the data for T
1
mapping
are more complicated than for conventional contrast-enhanced MRI.
REFERENCES
1. Kopecek, J. et al., HPMA copolymer-anticancer drug conjugates: design, activity and mechanism of
action, Eur. J. Pharm. Biopharm., 50, 61, 2000.
2. Kopecek, J. et al., Water soluble polymers in tumor targeted delivery, J. Control. Rel., 74, 147, 2001.
3. Hobbs, S. K. et al., Regulation of transport pathways in tumor vessels: role of tumor type and
microenvironment, Proc. Natl Acad. Sci. USA, 95, 4607, 1998.
4. Maeda, H., Sawa, T., and Konno, T., Mechanism of tumor-targeted delivery of macromolecular drugs,
including the EPR effect in solid tumor and clinical overview of the prototype polymeric drug
SMANCS, J. Control. Rel., 74, 47, 2001.
5. Minko, T., Kopeckova, P., and Kopecek, J., Chronic exposure to HPMA copolymer-bound adria-
mycin does not induce multidrug resistance in a human ovarian carcinoma cell line, J. Controlled Rel.,
59, 133, 1999.
6. Minko, T. et al., HPMA copolymer bound adriamycin overcomes MDR1 gene encoded resistance in a
human ovarian carcinoma cell line, J. Control. Rel., 54, 223, 1998.
7. Abe, S. and Otsuki, M., Styrene maleic acid neocarzinostatin treatment for hepatocellular carcinoma,
Curr. Med. Chem. Anti-Cancer Agents, 2, 715, 2002.
8. Brewerton, L. J., Fung, E., and Snyder, F. F., Polyethylene glycol-conjugated adenosine phosphoryl-
ase: development of alternative enzyme therapy for adenosine deaminase deciency, Biochim.
Biophys. Acta, 1637, 171, 2003.
9. Pinheiro, J. P. et al., Drug monitoring of PEG-asparaginase treatment in childhood acute lympho-
blastic leukemia and non-Hodgkins lymphoma, Leuk. Lymphoma, 43, 1911, 2002.
10. Ettinger, L. J. et al., An open-label, multicenter study of polyethylene glycol-L-asparaginase for the
treatment of acute lymphoblastic leukemia, Cancer, 75, 1176, 1995.
Nanotechnology for Cancer Therapy 210
q 2006 by Taylor & Francis Group, LLC
11. Motzer, R. J. et al., Phase II trial of branched peginterferon-alpha 2a (40 kDa) for patients with
advanced renal cell carcinoma, Ann. Oncol., 13, 1799, 2002.
12. Rowinsky, E. K. et al., A phase I and pharmacokinetic study of pegylated camptothecin as a 1-hour
infusion every 3 weeks in patients with advanced solid malignancies, J. Clin. Oncol., 21, 148, 2003.
13. Auzenne, E. et al., Superior therapeutic prole of poly-L-glutamic acid-paclitaxel copolymer
compared with Taxol in xenogeneic compartmental models of human ovarian carcinoma, Clin.
Cancer Res., 8, 573, 2002.
14. Gianasi, E. et al., HPMA copolymer platinates as novel antitumor agents: in vitro properties, phar-
macokinetics and antitumor activity in vivo, Eur. J. Cancer, 35, 994, 1999.
15. Thomson, A. H. et al., Population pharmacokinetics in phase I drug development: a phase I study of
PK1 in patients with solid tumors, Br. J. Cancer, 81, 99, 1999.
16. Seymour, L. W. et al., Hepatic drug targeting: phase I evaluation of polymer-bound doxorubicin,
J. Clin. Oncol., 20, 1668, 2002.
17. Liang, Z. P. and Lauterbur, P. C., Principles of Magnetic Resonance Imaging, IEEE Press, New York,
1999.
18. Vlaardingerbroek, M. T. and den Boer, J. A., Magnetic Resonance Imaging, 3rd ed., Springer, New
York, 2003.
19. Lauffer, R. B., Paramagnetic metal complexes as water proton relaxation agents for NMR imaging:
theory and design, Chem. Rev., 87, 901, 1987.
20. Merbach, A. E. and Toth, E., The Chemistry of Contrast Agents in Medical Magnetic Resonance
Imaging, John Wiley & Sons, Inc., England, 2001 chap. 12
21. Weinmann, H. J. et al., Characteristics of gadolinium-DTPA complex: a potential NMR contrast
agent, Am. J. Roentgenol., 142, 619, 1984.
22. Kaplan, G. D., Aisen, A. M., Aravapalli, S. R. et al., Preliminary clinical trial of gadodiamide
injection: a new nonionic gadolinium contrast agent for MR imaging, J. Magn. Reson. Imaging, 1,
57, 1991.
23. Magerstadt, M. et al., Gd(DOTA): An alternative to Gd(DTPA) as a T
1,2
relaxation agent for NMR
imaging or spectroscopy, Magn. Reson. Med., 3, 808, 1986.
24. Tweedle, M. F., The proHance story: the making of a novel MRI contrast agent, Eur. Radiol.,
7(Suppl 5), 225, 1997.
25. Youk, J. H., Lee, J. M., and Kim, C. S., MRI for detection of hepatocellular carcinoma: comparison of
mangafodipir trisodium and gadopentetate dimeglumine contrast agents, Am. J. Roentgenol., 183,
1049, 2004.
26. Weissleder, R. et al., Ultrasmall superparamagnetic iron oxide: an intravenous contrast agent for
assessing lymph nodes with MR imaging, Radiology, 175, 494, 1990.
27. Wang, S. C. et al., Evaluation of Gd-DTPA-labeled Dextran as an intravascular MR contrast agent:
imaging characteristics in normal rat tissues, Radiology, 175, 483, 1990.
28. Bryant, L. H. et al., Synthesis and relaxometry of high-generation (GZ5, 7, 9 and 10) PAMAM
dendrimer-DOTA-gadolinium chelates, J. Magn. Reson. Imaging, 9, 348, 1999.
29. Lu, Z. R. et al., Design of novel bioconjugates for targeted drug delivery, J. Control. Rel., 78, 165,
2002.
30. Duncan, R., The dawning era of polymer therapeutics, Nat. Rev. Drug Discov., 2, 347, 2003.
31. Pimm, M. V. et al., Gamma scintigraphy of the biodistribution of
123
I labeled N-(2-hydroxypropyl)-
methacrylamide copolymer-doxorubicin conjugates in mice with transplanted melanoma and
mammary carcinoma, J. Drug Target, 3, 375, 1996.
32. Kissel, M. et al., Synthetic macromolecular drug carriers: biodistribution of poly [(N-2-hydroxypro-
pyl)methacrylamide] copolymers and their accumulation in solid rat tumors, J. Pharm. Sci. Tech., 55,
191, 2001.
33. Mitra, A. et al., Technetium-99m-Labeled N-(2-hydroxypropyl) methacrylamide copolymers:
synthesis, characterization, and in vivo biodistribution, Pharm. Res., 21, 1153, 2004.
34. Wang, Y.L., and Lu, Z.R. Unpublished data 2005.
35. Wang, D. et al., The arhrotropism of macromolecules in adjuvant-induced arthritis rat model: a
preliminary study, Pharm. Res., 21, 1741, 2004.
36. Li, C., Poly (L-glutamic acid)-anticancer drug conjugates, Adv. Drug Deliv. Rev., 54, 695, 2002.
Noninvasive Visualization of In Vivo Drug Delivery of Paramagnetic Polymer Conjugates 211
q 2006 by Taylor & Francis Group, LLC
37. Hurwitz, E., Wilchek, M., and Pitha, J., Soluble macromolecules as carriers for daunorubicin, J. Appl.
Biochem., 2, 25, 1980.
38. Li, C. et al., Complete regression of well-established tumors using novel water-soluble poly (L-
glutamic acid)-paclitaxel conjugates, Cancer Res., 58, 2404, 1998.
39. Zou, Y. et al., Effectiveness of water soluble poly(L-glutamic acid)camptothecin conjugate against
resistant human lung cancer xenografted in nude mice, Int. J. Oncol., 18, 331, 2001.
40. Lu, Z. R. et al., Poly(L-glutamic acid) Gd(III)-DOTA conjugate with a degradable spacer for magnetic
resonance imaging, Bioconjugate Chem., 14, 715, 2003.
41. Lu, Z. R., Kopeckova, P., and Kopecek, J., Polymerizable Fab fragment for targeting of anticancer
drugs, Nature Biotechnol., 17, 1101, 1999.
42. Lu, Z. R. et al., Polymerizable Fab antibody fragments targeted photodynamic cancer therapy in nude
mice, STP Pharm. Sci., 13, 69, 2003.
43. Vaidya, A. et al. Non-invasive in vivo imaging of a polymeric drug conjugate using contrast enhanced
MRI. In Transactions of 32nd Annual Meeting of the Controlled Release Society, 2005.
Nanotechnology for Cancer Therapy 212
q 2006 by Taylor & Francis Group, LLC

Das könnte Ihnen auch gefallen