ArticlePDF Available

Local carboplatin delivery and tissue distribution in livers after radiofrequency ablation

Authors:

Abstract and Figures

This study investigated the local drug pharmacokinetics of intralesional drug delivery after radiofrequency ablation of the liver. We hypothesized that the tissue architecture damaged by the ablation process facilitates the drug penetration in the liver and potentially enlarges the therapeutic margin in the local treatment of cancer. The delivery rate and tissue distribution of carboplatin, an anticancer agent, released from poly(D,L-lactide-co-glycolide) implants into rat livers after radiofrequency ablation were quantified by atomic absorption spectroscopy. Results showed that carboplatin clearance through blood perfusion was significantly slower in the ablated livers, leading to a more extensive tissue retention and distribution of the drug. The concentration of Pt at the implant-tissue interface ranged from 234 to 1440 microg Pt/(g liver) in the ablated livers over 144 h versus 56 to 177 microg Pt/(g liver) in the normal tissue. The maximum penetration distance at which Pt level reached above 6 microg/g (calculated based on a reported IC90 value for carboplatin) was 8-10 mm and 4-6 mm in ablated and normal liver, respectively. Histological analysis of the necrotic lesions showed widespread destruction of tissue structure and vasculature, supporting the initial hypothesis. This study demonstrated that intralesional drug delivery could provide a sustained, elevated concentration of anticancer drug at the ablation boundary that has the potential to eliminate residual cancer cells surviving radiofrequency ablation.
Content may be subject to copyright.
Local carboplatin delivery and tissue distribution in livers
after radiofrequency ablation
A. Szymanski-Exner,
1
A. Gallacher,
2
N. T. Stowe,
3
B. Weinberg,
1
J. R. Haaga,
4
J. Gao
1,4
1
Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio
2
Department of Analytical Chemistry, Ricerca, LLC, Concord, Ohio
3
Department of Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio
4
Department of Radiology, University Hospitals of Cleveland, Cleveland, Ohio
Received 9 December 2002; accepted 20 December 2002
Abstract: This study investigated the local drug pharma-
cokinetics of intralesional drug delivery after radiofre-
quency ablation of the liver. We hypothesized that the tissue
architecture damaged by the ablation process facilitates the
drug penetration in the liver and potentially enlarges the
therapeutic margin in the local treatment of cancer. The
delivery rate and tissue distribution of carboplatin, an anti-
cancer agent, released from poly(D,L-lactide-co-glycolide)
implants into rat livers after radiofrequency ablation were
quantified by atomic absorption spectroscopy. Results
showed that carboplatin clearance through blood perfusion
was significantly slower in the ablated livers, leading to a
more extensive tissue retention and distribution of the drug.
The concentration of Pt at the implant–tissue interface
ranged from 234 to 1440 g Pt/(g liver) in the ablated livers
over 144 h versus 56 to 177 g Pt/(g liver) in the normal
tissue. The maximum penetration distance at which Pt level
reached above 6 g/g (calculated based on a reported IC
90
value for carboplatin) was 8–10 mm and 46 mm in ablated
and normal liver, respectively. Histological analysis of the
necrotic lesions showed widespread destruction of tissue
structure and vasculature, supporting the initial hypothesis.
This study demonstrated that intralesional drug delivery
could provide a sustained, elevated concentration of anti-
cancer drug at the ablation boundary that has the potential
to eliminate residual cancer cells surviving radiofrequency
ablation. © 2003 Wiley Periodicals, Inc. J Biomed Mater Res
67A: 510-516, 2003
Key words: drug delivery; tissue distribution; atomic ab-
sorption spectroscopy; pharmacokinetics; radiofrequency
ablation
INTRODUCTION
Image-guided radiofrequency (RF) ablation has
been successfully used in recent years as a minimally
invasive therapy to treat unresectable liver tumors.
1–8
In this procedure, imaging methods such as X-ray
computed tomography, magnetic resonance imaging,
or ultrasound guides the percutaneous insertion of a
needle electrode to the site of a tumor. Electric current
at radio frequency is applied through the needle, lead-
ing to ionic agitation, increased temperature, and fi-
nally coagulative necrosis.
2–6
The procedure is simple,
quick, and can be performed on an outpatient basis.
3,4
Studies have shown that although RF ablation is able
to destroy majority of the tumor tissue, tumor recur-
rence has been reported in many cases because of the
incomplete elimination of all the cancer cells.
3,6,7
To
address this limitation, other minimally invasive in-
terventional procedures, such as intralesional drug
therapy, are being explored.
Currently, research efforts in our group focus on the
development of a combination therapy consisting of
RF ablation and local chemotherapy for the treatment
of solid tumors. The therapy includes the following
steps: first, a small drug-loaded cylindrical implant,
the polymer millirod, will be fabricated using estab-
lished methods.
9
This device is composed of an active
agent entrapped in a biodegradable poly(lactic-co-gly-
colic acid) (PLGA) matrix. Second, under image guid-
ance, a tumor will be treated with RF ablation fol-
lowed by implantation of millirod(s) directly into
tumor tissues to provide site-specific delivery of the
selected agent. The active agent will be released from
the polymer matrix directly into tumor tissue at a
controlled rate over time to eliminate the residual
cancer cells.
Local drug delivery is a powerful technique that has
the potential to become a mainstream alternative to
Correspondence to: J. Gao; e-mail: jmg23@po.cwru.edu
Contract grant sponsor: National Institutes of Health; con-
tract grant number: R21 CA93993
© 2003 Wiley Periodicals, Inc.
conventional systemic chemotherapy. This mode of
drug administration can deliver the drug to a site of
action at a significantly higher concentration than is
possible with intravenous injection or oral delivery. It
can also maintain the concentration within the thera-
peutic window to avoid unnecessary toxicity. Partic-
ularly, local delivery is beneficial for drugs (especially
anticancer drugs) with narrow therapeutic indices or
short in vivo half-lives to maximize their therapeutic
efficacy.
For a local drug therapy to attain maximum efficacy,
it must deliver the therapeutic dose of a drug to the
target area without substantially affecting the normal
tissues. Frequently, the drug released from the im-
plant must travel a significant distance and reach the
site of action in its active state, and its ability to do so
will depend highly on the properties of the drug as
well as on the property of the tissue. Other groups
have examined the effects of cell density on drug
distribution in tissue and have determined that a de-
creased cell density due to apoptosis enhances tissue
penetration.
10–12
Currently, data on local drug release
and tissue pharmacokinetics are limited but need to be
quantified for the successful development of a local
drug delivery system. This is true especially in com-
plex tissue environments, such as thermally ablated
tissue. Because the viable and ablated tissue structure
varies drastically, with the ablated region lacking vi-
able cells and vasculature, we predict that this differ-
ence will significantly affect the local drug pharmaco-
kinetics in tissues.
In this study, we examined the release kinetics and
tissue distribution of carboplatin as it was released
from the polymer millirod in ablated liver tissues. The
drug distribution profiles provide information on the
local drug pharmacokinetics in ablated tissues and
were correlated with tissue structure from histology
analysis. Results from this study provide the funda-
mental understanding of structure-property relation-
ships between in vivo drug transport properties and
liver structures, which is essential for the future de-
sign of sustained release formulations in conjunction
with RF ablation for the treatment of liver tumors.
MATERIALS AND METHODS
Materials
PLGA (lactide:glycolide 1:1, 0.65 dL/g inherent viscos-
ity) was purchased from Birmingham Polymers, Inc. (Bir-
mingham, AL). Carboplatin (cis-diammine(1,1-cyclobutane-
dicarboxylato) platinum) and poly(vinyl alcohol) (13–23
kDa) was purchased from Sigma-Aldrich (Milwaukee, WI).
D()-Glucose was purchased from Fluka (Milwaukee, WI).
Phosphate-buffered saline (PBS), sodium hydroxide (NaOH,
0.2M), nitric acid (70%, trace metal grade), and methylene
chloride were obtained from Fisher Scientific (Pittsburgh,
PA). Single-element platinum standard (NIST traceable) was
purchased from CPI International (Santa Rosa, CA). Teflon
tubes were purchased from McMaster–Carr Supply Com-
pany (Cleveland, OH). Sprague–Dawley rats were obtained
from Charles River Laboratories (Wilmington, MA).
Polymer implant fabrication and in vitro
characterization
Millirod implants were fabricated according to a previ-
ously established compression-heat molding procedure.
9
Briefly, PLGA microspheres (approx. 4 m diameter) were
mixed with carboplatin to form a uniform mixture, and
D()-glucose was added to the mixtures in order to expedite
the rate of release. The homogeneously mixed powder was
placed in a mold and compressed at 4.6 10
6
Pa at 90°C for
2 h. The resulting cylindrical millirods have an average
diameter of 1.62 mm and a theoretical loading density of
10% carboplatin with 35% glucose (w/w).
The in vitro release of carboplatin from the millirods was
measured in PBS (pH 7.4). Typically, segments of millirods
(approx. 8 mm in length) were submerged in 10 mL of PBS
and placed in an orbital shaker (New Brunswick Scientific,
model C24) at 37°C and 100 rpm agitation. At each sampling
point, the millirod was removed from the vial and placed
into 10 mL of fresh PBS. The retained sample was analyzed
using flameless, graphite-furnace atomic absorption spec-
troscopy (GF-AAS, PerkinElmer model 4100ZL). This
method has been a widely accepted method of examining
levels of various metals in tissue and body fluids.
13–15
The
method is a spectrophotometric technique based on the ab-
sorption of radiant energy by atoms.
16
First, five single-
element platinum standards (NIST traceable) were prepared
in deionized water at concentrations of 10, 25, 50, 100, and
500 g Pt/L to establish the calibration curve. The linear
correlation coefficients for all the curves (area counts for Pt
at 265.9 nm vs. concentration) were greater than 0.999 before
analysis. The platinum concentration in the retained sample
(Pt, g/mL) was determined by quantitative serial dilution
(250) in water followed by their interpolative assay against
the standard curve. The platinum content was then con-
verted to reflect the total carboplatin release based on the
weight percentage of Pt to the drug molecule. The average
amount of carboplatin remaining in the implants was calcu-
lated from this data and was standardized to the initial
measured length of each implant. The total concentration of
carboplatin was calculated based on measurements of the
drug in completely degraded implants.
RF ablation and millirod implantation in rat livers
Animal procedures followed an approved protocol by the
Institutional Animal Care and Use Committee. Male Spra-
gue–Dawley rats (250–350 g) were anesthetized using an
intraperitoneal injection of sodium pentobarbital (4 mg/100
g). The abdomen was swabbed with betadine solution, and
CARBOPLATIN DISTRIBUTION AFTER RF ABLATION 511
mercaine (bupivacaine, 0.1 mL) was injected subcutaneously
at the incision site prior to the start of surgery. The liver was
exposed through an incision in the midsection. In the ani-
mals undergoing ablation, the liver capsule of the medial
lobe was first perforated with an 18-gauge hypodermic nee-
dle, and the liver tissue was ablated with a 19-gauge needle
electrode (Radionics
, Burlington, MA 01803) at 90 3°C for
2 min. The ablation procedure followed a previously estab-
lished method.
8,17
After ablation, millirods (5–7 mm in
length) were implanted into the needle electrode track. Mil-
lirods of the same composition used in the ablated lobes
were implanted into nonablated livers for comparison. After
implantation, a small piece of cotton was sutured on top of
the implantation site to seal the wound and prevent the
implant from slipping out. The animals received Buprenex
(buprenorphine, 0.05–0.1 mg/kg) after the surgery and were
allowed to recover. The animals were then euthanized with
Fatal Plus (concentrated sodium pentobarbital), and livers
were removed and stored at 80°C. Animals were divided
into six experimental groups with endpoints of 1, 6, 24, 48,
96, and 144 h. Each group consisted of six rats with implants
in normal livers (n3) and ablated livers (n3).
Sample preparation and analysis with atomic
absorption spectroscopy
The platinum content in livers and explanted millirods
was also analyzed with atomic absorption spectroscopy
(AAS). In preparation for AAS analysis, the livers were
divided into three segments around the center (implantation
site) and three 2-mm thick slices were sectioned from each
segment. These were cut into 2-mm wide strips and the six
strips closest to the implant (radial distance of 12 mm) were
retained for analysis. These samples were deposited into
previously weighed, 4-mL glass scintillation vials. The vials
were weighed again to determine the wet mass of each liver
segment. Sample digestion was conducted using a novel
dry-block procedure (compared to the standard microwave
digestion). The 4-mL vials were placed into dry-block incu-
bators and 100 L of 70% nitric acid was added to each vial.
The samples were heated to 70°C for 1 h and digestion was
aided by addition of 50 L hydrogen peroxide after 20, 30,
and 50 min. The clear sample solutions were transferred to
15-mL Falcon centrifuge tubes that were diluted to 5 mL
with distilled water and stored at 4°C until analysis. To
determine carboplatin release in vivo, the explanted milli-
rods were degraded in 2 mL of 0.2M NaOH at 37°C for 1
week. The solution was neutralized with addition of 2 mL of
0.2M nitric acid and filtered with 0.45-m syringe filters
before AAS analysis. The platinum concentrations in the
above solutions were determined by interpolative assay
against the standard curve (10–500 g Pt/L) as described in
the previous section. The g/L measurements were con-
verted to g/(g liver) by multiplication of the sample vol-
ume and division by the original wet tissue weight. The
explant measurements were converted to mg/cm as the
remaining quantity of carboplatin per unit length of the
millirod.
The t
1/2
was calculated based on exponential decay curve
fit to the average release data. The tissue penetration dis-
tance was calculated as the maximum distance at which the
Pt concentration reaches above 6 g/g, a value calculated
based on the IC
90
(10 g/mL)
18
carboplatin concentration to
the VX-2 cancer cells. The area under the concentration-time
curve (AUC) for carboplatin in ablated liver tissue over
144 h was calculated using the trapezoid rule for the im-
plant/tissue interface and ablation boundary. An unpaired,
two-tailed Student t test with a 95% confidence interval was
used to determined significant differences among data sets.
Significant outliers were discarded on basis of the Grubbs
test (␣⫽0.05).
Histological analysis
All liver samples underwent gross histological examina-
tion and microscopic analysis. The diameter of the visible
ablated area perpendicular to the ablation needle tract was
measured three times and an average measurement of the
ablation area was calculated. Representative sections of the
specimens were removed and fixed in 10% formalin solu-
tion. The preserved sections were embedded in paraffin,
sectioned into 5-m slices and stained with hematoxylin and
eosin. The hematoxylin and eosin sections were examined to
determine the normal tissue structure surrounding the im-
planted millirod and the damage inflicted by the implanta-
tion trauma and thermal ablation.
RESULTS
In vitro release of carboplatin in PBS buffer
Figure 1 shows the average release profile of carbo-
platin in PBS buffer (pH 7.4) at 37°C. The data are
represented as carboplatin remaining in the millirod
over time and is standardized by the length of each
implant (mg/cm). The cumulative release follows the
Higuchi model (C
drug
kt
n
,n0.5 0.03) with a
value of t
1/2
at 42 8 h. In addition, 2.0 0.2 and
3.8 0.3 mg carboplatin/cm were released after 24
and 96 h, respectively (n4). The reproducibility of
the fabrication procedure is demonstrated by a rea-
sonably low standard deviation (0.3 mg/cm, n4)
at all time points.
In vivo release of carboplatin in rat livers
The in vivo release of carboplatin in ablated and
normal rat livers is shown in Figure 1. Compared with
the in vitro release, the standard deviation in in vivo
release data is significantly greater, particularly at ear-
lier time points (t24 h). Although carboplatin re-
lease appears to be faster in ablated livers than normal
livers in the first day after implantation, it should be
512 SZYMANSKI-EXNER ET AL.
noted the difference is not statistically significant (e.g.
the pvalue from the Student ttest is 0.13 and 0.28 at 6
and 24 h, respectively). Release half-life was deter-
mined as 51 10 and 44 15 h for normal and ablated
tissue, respectively. When examining the raw data, we
observed a comparable release amount of carboplatin
at longer time points in both systems, with 3.7 0.2
and 3.8 0.5 mg carboplatin/cm released after 96 h in
normal and ablated livers, respectively. This data
demonstrates similar overall release kinetics in the
two liver environments as well as in PBS buffer (3.8
0.3 mg/cm at 96 h).
Distribution of carboplatin in normal and ablated
liver tissues
Figure 2(A,B) shows the tissue distribution of car-
boplatin in platinum concentrations as a function of
distance in ablated (n9) and normal (n9) liver
tissues. Data show significantly more carboplatin were
retained in the ablated tissue over the normal tissue. In
the livers treated with RF ablation [Fig. 2(A)], the
range of platinum in the tissue exceeds 1400 g Pt/(g
liver) at the millirod/tissue interface, and the concen-
tration rises steadily through 48 h and decreases there-
after. In contrast, the range of Pt in the normal liver
[Fig. 2(B)] consistently remains below 200 g Pt/(g
liver), and the concentration varies randomly through-
out the 6 days after implantation. The maximum tissue
penetration distance, at which the Pt concentration
reaches above 6 g/g liver, was found to be between
8–10 mm in the ablated liver at 1 and 96 h. During the
other time points, the maximum distance was between
4 6 mm. In the normal liver, the maximum penetra-
tion distance was found to be between 0–2 mm at all
but one time point and between 4 6 mm in the first
hour.
The change in carboplatin concentration over time
at the implant/tissue interface and the distal ablation
boundary is shown in Figure 2(C,D). At implant/
tissue interface, carboplatin in the ablated livers
reaches 1.4 0.6 10
3
g/g liver (C
max
,n9) at 48 h
and is 24 times higher than that in the normal liver
[61 45 g/g, Fig. 2(C)]. At the approximate ablation
boundary (4 6 mm), the carboplatin concentration
was at least two times higher in the ablated liver at all
time points, and 14 times higher at 144 h [Fig. 2(D)].
Although the differences in measured Pt are consis-
tent, it should be noted that some measurements (es-
pecially the Pt content in normal tissue) at this dis-
tance are below the detection limit of the atomic
absorption technique, and may not be accurate.
The AUC at the implant/tissue interface was calcu-
lated to be 234.6 and 28.5 (mg/g)*h for carboplatin in
ablated and normal liver, respectively. The AUC at the
ablation boundary (or 4 6 mm from the implant) was
calculated to be 4.8 (mg/g)*h in ablated and 0.8 (mg/
g)*h in normal liver. All AUC values are reported in
mg carboplatin (not Pt) for clinical relevance.
Histological analysis
The gross morphology of the tissue surrounding the
millirod was significantly altered in the ablated liver
site. The ablation radius ranged from 3.8 to 5.8 mm
(average radius 4.8 0.9 mm). Millirod implanta-
tion in the ablated lesion resulted in little or no addi-
tional injury to the tissue as observed by the easy
removal of the millirod from the implantation site.
Upon gross examination, the tissue at the millirod
boundary appeared to be identical to that further
away from the implantation site in the ablated area. In
comparison, the implantation site in normal liver
showed a slight morphological change in the sur-
rounding tissue. The tissue adjacent to the implant
appeared white and granular, and the implant was
difficult to remove, possibly indicating an early re-
sponse of the tissue to injury.
Microscopic examination of the implantation site
showed a vastly different tissue structure in the ab-
lated liver tissue with notable changes over 6 days
[Fig. 3(A,D)]. The tissue structure near the implanta-
tion site showed the same architecture as in the rest of
the ablated area [Fig. 3(A,B)]. The cellular structure
was different from normal liver cells and corresponds
Figure 1. Release of carboplatin from millirod implants in
vitro (n4) and in vivo (n3) in ablated () and normal (F)
liver. Dara are shown as weight of carboplatin remaining in
millirod and standardized based on the length of individual
implants. Portions of error bars (reflecting standard devia-
tion measurements) were omitted for clarity. In the in vitro
release (Œ), the standard deviation was below 0.3 mg/cm at
all time points.
CARBOPLATIN DISTRIBUTION AFTER RF ABLATION 513
to features of coagulative necrosis with picnotic nuclei
and disrupted and irregular cytoplasm. The sinusoi-
dal structure was destroyed and the interstitial space
was increased. At the distal edge of the ablated area,
and at the interface of the ablated and normal liver
tissue, a ring of inflammatory infiltrate, composed of
lymphocytes and monocytes, was observed. This ring is
more apparent at later time points starting at 24 h and
increasing through 144 h. The cell nuclei in the necrotic
area become increasingly sparse as the necrosis spreads,
and fibroblasts and granulation tissue are visible at the
distal ablation boundary at 96 and 144 h [Fig. 3(C,D)].
DISCUSSION
The therapeutic efficacy of the intralesional drug
delivery system addressed in this study is highly de-
pendent on the local drug pharmacokinetics in the
tissue environment surrounding the implantation site.
Characterization of the in vivo drug release kinetics
and drug penetration in ablated tissue is critical in
providing the necessary experimental data for the de-
velopment of these delivery systems.
The in vitro release kinetics of carboplatin in PBS
buffer (t
1/2
42 8 h) agrees reasonably well with
Figure 2. Local drug pharmacokinetics of carboplatin in normal and ablated liver over 144 h. (A) Tissue distribution in
ablated liver. Carboplatin distribution increases initially and reaches a maximum at 48 h. The distribution data at 6 h
superimpose with the 96-h data and were omitted. (B) Tissue distribution in normal liver, where no obvious time-dependent
trend was observed. Error bars were omitted for clarity in (A) and (B). (C) Pt concentration at the implant/tissue interface in
normal () and ablated (E) livers. (D) Pt concentration at the distal ablation boundary in ablated livers. Pt concentration at
the same distance in normal liver is shown for comparison. Dashed line represents the IC
90
value of carboplatin to VX-2 cancer
cells from the literature. Error bars reflect standard error (n9).
514 SZYMANSKI-EXNER ET AL.
those in normal liver (t
1/2
51 10 h) and ablated
liver (t
1/2
44 15 h) in vivo. The in vivo release
kinetics showed larger experimental variability, which
may reflect the heterogeneous liver environments in
different animals. Results from current study indicate
slightly faster release of carboplatin from polymer
millirods in ablated liver than normal liver; however,
this difference is not statistically significant. In com-
parison, the retention, distribution and penetration of
carboplatin are significantly different in the two tissue
environments. Carboplatin retention is approximately
one order of magnitude higher in the ablated liver
over normal liver [Fig. 2(A,B)]. In addition, drug pen-
etration is significantly greater in ablated liver with a
maximum penetration distance of 8–10 mm compared
to mostly 2 mm in the normal liver.
The different pharmacokinetics of carboplatin in ab-
lated and normal liver can be correlated to structural
changes in the liver following RF ablation. The histol-
ogy analysis documented these changes in the ablated
lesion over 6 days. During this time it is possible to
observe morphological changes in hepatocytes under-
going coagulative necrosis, where the cells shrink and
nuclei become increasingly sparse as they are cleared
out from the injured area. These results are in agree-
ment with those reported previously by our group
17
as well as others.
5,8
More importantly, the histological
analysis of ablated lesions confirms the severe destruc-
tion of organized sinusoidal capillary structure and
other liver vasculature visible in normal tissue (not
shown). The lack of carboplatin clearance through
blood perfusion in ablated tissue is a determinant
factor for the greater retention and penetration of car-
boplatin. Without drug loss from perfusion, drug dif-
Figure 3. Histological changes in liver tissue after RF ablation . Arrows indicated the location of ablated tissue and implant
position. (A) After 24 h, the boundary between normal and ablated tissue is clearly defined (*). (B) At 48 h, the ring of
inflammatory cells is clearly visible (#). (C) At 96 h, the inflammatory response gives way to fibroblasts and collagen formation
(). (D) At 144 h, the disorganized structure of the fibrous capsule is clearly visible (). The original magnification was 100
for (A) and (B) and 200for (C) and (D). [Color figure can be viewed in the online issue, which is available at www.inter-
science.wiley.com.]
CARBOPLATIN DISTRIBUTION AFTER RF ABLATION 515
fusion becomes the dominant process of transport in
ablated liver, and leads to significantly higher pene-
tration distance over normal liver. Wound healing
response to the heat-induced injury may also affect the
local drug pharmacokinetics. At the ablation boundary,
a ring of inflammatory cells is observed from 24 to 96 h
[Fig. 3(B,C)], and fibroblasts are observed forming a fi-
brous capsule around the ablated area after 96 h [Fig.
3(D)]. The formation of fibrous capsule may cause the
elevated platinum concentration at the ablation bound-
ary at 144 h [Fig. 2(D)] over the other time points.
In our proposed combination therapy, intralesional
drug delivery aims to eliminate the residual cancer
cells surviving RF ablation. Therefore, the ablation
boundary is the targeted site of action and the concen-
tration-time relationships at this location are impor-
tant for an effective carboplatin therapy. Our study
showed that carboplatin reached its therapeutic con-
centration (IC
90
of carboplatin to VX-2 cancer cells) at
the ablation boundary for all the time points over
144 h [Fig. 2(D)]. The AUC value at the ablation
boundary was determined to be 4.8 (mg/g)*h in ab-
lated liver, six times more than normal liver [0.8 (mg/
g)*h] at the same distance. Since clinical AUC values
are normally calculated from the plasma drug concen-
trations, the tissue measurements from this study can-
not be compared with the clinical data. Nonetheless, as
regional and local drug delivery becomes a more ac-
cepted therapy, these measurements may become essen-
tial in describing the local tissue pharmacokinetics and
evaluating the effectiveness of drug delivery systems.
CONCLUSIONS
The data obtained in this study successfully support
our hypothesis that radiofrequency ablation of tissue
increases the maximum penetration distance of a drug
and allows the drug to be consistently elevated above
therapeutic concentration at the site of action. The
tissue penetration was three times higher in the ab-
lated tissue and drug concentration was found to be
an order of magnitude higher at most distances from
the tissue/implant interface as compared with similar
implants in normal livers. The mode of action for this
striking difference is the destruction of vasculature and
loss of perfusion resulting from the ablation process,
which is supported by gross and microscopic examina-
tion of the histology sections. Although this study dem-
onstrates that the drug can reach the ablation boundary,
further studies on a tumor model are necessary to eval-
uate the increased efficacy of the combination therapy
(RF ablation local drug delivery) over RF ablation
alone. These studies are currently being conducted in
our group with the rabbit VX-2 tumor model.
The authors thank Dr. James Anderson for his help with
the histology analysis.
References
1. Becker GJ. 2000 RSNA annual oration in diagnostic radiology:
The future of interventional radiology. Radiology 2001;220:
281–292.
2. Lewin JS, Connell CF, Duerk JL, Chung YC, Clampitt ME,
Spisak J, Gazelle GS, Haaga JR. Interactive MRI-guided radio-
frequency interstitial thermal ablation of abdominal tumors:
clinical trial for evaluation of safety and feasibility. J Magn
Reson Imaging 1998;8:4047.
3. Dodd GD III, Soulen MC, Kane RA, Livraghi T, Lees WR,
Yamashita Y, Gillams AR, Karahan OI, Rhim H. Minimally
invasive treatment of malignant hepatic tumors: at the thresh-
old of a major breakthrough. Radiographics 2000;20:9–27.
4. Rhim H, Dodd GD III. Radiofrequency thermal ablation of
liver tumors. J Clin Ultrasound 1999; 27:221–229.
5. Goldberg SN, Gazelle GS, Compton CC, Mueller PR, Tanabe
KK. Treatment of intrahepatic malignancy with radiofre-
quency ablation: radiologic-pathologic correlation. Cancer
2000;88:2452–2463.
6. Goldberg SN, Gazelle GS. Radiofrequency tissue ablation:
physical principles and techniques for increasing coagulation
necrosis. Hepatogastroenterology 2001;48:359–367.
7. Bartolozzi C, Crocetti L, Cioni D, Donati FM, Lencioni R.
Assessment of therapeutic effect of liver tumor ablation pro-
cedures. Hepatogastroenterology 2001;48:352–358.
8. Boaz TL, Lewin JS, Chung YC, Duerk JL, Clampitt ME, Haaga
JR. MR monitoring of MR-guided radiofrequency thermal ab-
lation of normal liver in an animal model. J Magn Reson
Imaging 1998;8:6469.
9. Qian F, Szymanski A, Gao J. Fabrication and Characterization
of Controlled Release Poly(D,L-Lactide-co-Glycolide) Milli-
rods. J Biomed Mater Res 2001;55:512–522.
10. Au JLS, Jang SH, Wientjes MG. Clinical aspects of drug deliv-
ery to tumors. J Control Release 2002;78:81–95.
11. Kuh HJ, Jang SH, Wientjes MG, Weaver JR, Au JLS. Determi-
nants of Paclitaxel Penetration and Accumulation in Human
Solid Tumor. J Positron Emission Tomogr 1999;290:871–880.
12. Jang SH, Wientjes MG, Au JLS. Enhancement of paclitaxel
delivery to solid tumors by apoptosis-inducing pretreatment:
effect of treatment schedule. J Positron Emission Tomogr 2001;
296:1035–1042.
13. Duffull SB, Robinson BA. Clinical pharmacokinetics and dose
optimisation of carboplatin. Clin Pharmacokinet 1997;33:161–83.
14. Milacic R, Cemazar M, Sersa G. Determination of platinum in
tumour tissues after cisplatin therapy by electrothermal atomic
absorption spectrometry. J Pharm Biomed Anal 1997;16:343–348.
15. Meerum Terwogt JM, Tibben MM, Welbank H, Schellens JH,
Beijnen JH. Validated method for the determination of plati-
num from a liposomal source (SPI-77) in human plasma using
graphite furnace Zeeman atomic absorption spectrometry. Fre-
senius J Anal Chem 2000;366:298–302.
16. Shugar GJ. Atomic Absorption Spectroscopy. In: Shugar R, Bau-
man L, Shugar-Bauman R, editors. Chemical technicians’ ready
reference handbook, 3rd ed. New York: McGraw-Hill, Inc.; 1990.
p 751–763.
17. Szymanski-Exner A, Stowe NT, Lazebnik RS, Salem KA, Wilson
DL, Haaga JR, Gao J. Noninvasive monitoring of local drug
release in a rabbit radiofrequency (RF) ablation model using x-ray
computed tomography. J Control Release 2002;83:415–425.
18. Pauser S, Wagner S, Lippmann M, Pohlen U, Reszka R, Wolf
KJ. Evaluation of efficient chemoembolization mixtures by
magnetic resonance imaging therapy monitoring: An experi-
mental study on the VX2 tumor in the rabbit liver. Cancer Res
1996;56:1863–1867.
516 SZYMANSKI-EXNER ET AL.
... Following the scan, the implants were removed and the remaining drug was extracted into solution. These samples were analyzed with atomic absorption spectroscopy to determine the total amount of drug remaining in each implant (12). Average HU values were calculated within a circular region of interest for each implant and tissue phantom. ...
... Data obtained from the CT images was correlated with atomic absorption spectrometry (AAS) analysis of the retrieved tissue and implants as previously reported (12). To further validate the accuracy of the CT method, we calculated the average drug concentration in a tissue pillbox 0.8 -4 mm from the implant/tissue interface for both the CT and AAS data. ...
... The data from this study were compared with AAS analysis of carboplatin content in both the implants removed immediately after the CT scan and the liver tissue as reported previously (12). It is apparent that the CT data accurately predicts the kinetics in this system despite the small size of the implant, and this is true in both ablated (Fig. 4A) and normal (Fig. 4B) livers. ...
Article
Rationale and Objectives. Computed tomography (CT) was used to noninvasively monitor local drug pharmacokinetics from polymer implants in rat livers before and following radiofrequency ablation. Materials and Methods. Polymer matrixes containing carboplatin (a platinum-containing chemotherapeutic agent) were implanted into rat livers either immediately after radiofrequency ablation (n = 15) or without prior treatment (n = 15). The animals were divided into five subgroups (n = 3 per group) and subjected to a terminal CT scan at 6, 24, 48, 96, or 144 hours. Carboplatin concentration in tissue and within the implant matrix was correlated with CT intensity, and standard curves were produced for each environment. This correlation was used to evaluate the differences in drug transport properties between normal and ablated rat livers. A quantitative image analysis method was developed and used to evaluate the release rate and tissue distribution of carboplatin in normal and ablated liver tissue. The CT data were validated by previously reported atomic absorption spectroscopy measurement of implant and tissue drug levels. Results. Correlation of carboplatin concentration and Hounsfield units results in a linear relationship with correlation coefficients (slopes) of 15 and 4 Hounsfield units/(mg/mL), for carboplatin in tissue and polymer, respectively. Noninvasive monitoring of local pharmacokinetics in normal and ablated tissues indicates that ablation before local carboplatin delivery increases the retention of carboplatin within the polymer matrix and drastically increases the drug retention in the ablated tissue volume (over 3-fold difference) resulting in a higher average dose to the surrounding tissue. At 1.6 turn from the implant boundary, carboplatin concentration is significantly higher in ablated tissue at 48, 96, and 144 hours (P <.05), and reaches 4.7 mg/mL in ablated tissue at 48 hours. In comparison, the concentration in normal liver at 1.6 mm reaches only 0.7 mg/mL at the same time point. The drug penetrates 3.1 mm in ablated liver compared with 2.3 mm in normal liver also at 48 hours. After 144 hours, the drug is still detected at 3.1 mm in ablated liver but not in normal liver. The differences are significant (P <.05) at both 48 and 144 hours. Correlation with chemical analysis suggests that CT data accurately predicts the drug pharmacokinetics in both ablated and normal livers. Conclusion. This work shows that X-ray CT imaging is a useful and promising technique for in vivo monitoring of the release kinetics of locally delivered radiopaque agents. tomography.
... Following the scan, the implants were removed and the remaining drug was extracted into solution. These samples were analyzed with atomic absorption spectroscopy to determine the total amount of drug remaining in each implant (12). Average HU values were calculated within a circular region of interest for each implant and tissue phantom. ...
... Data obtained from the CT images was correlated with atomic absorption spectrometry (AAS) analysis of the retrieved tissue and implants as previously reported (12). To further validate the accuracy of the CT method, we calculated the average drug concentration in a tissue pillbox 0.8 -4 mm from the implant/tissue interface for both the CT and AAS data. ...
... The data from this study were compared with AAS analysis of carboplatin content in both the implants removed immediately after the CT scan and the liver tissue as reported previously (12). It is apparent that the CT data accurately predicts the kinetics in this system despite the small size of the implant, and this is true in both ablated (Fig. 4A) and normal (Fig. 4B) livers. ...
Article
Full-text available
Computed tomography (CT) was used to noninvasively monitor local drug pharmacokinetics from polymer implants in rat livers before and following radiofrequency ablation. Polymer matrixes containing carboplatin (a platinum-containing chemotherapeutic agent) were implanted into rat livers either immediately after radiofrequency ablation (n = 15) or without prior treatment (n = 15). The animals were divided into five subgroups (n = 3 per group) and subjected to a terminal CT scan at 6, 24, 48, 96, or 144 hours. Carboplatin concentration in tissue and within the implant matrix was correlated with CT intensity, and standard curves were produced for each environment. This correlation was used to evaluate the differences in drug transport properties between normal and ablated rat livers. A quantitative image analysis method was developed and used to evaluate the release rate and tissue distribution of carboplatin in normal and ablated liver tissue. The CT data were validated by previously reported atomic absorption spectroscopy measurement of implant and tissue drug levels. Correlation of carboplatin concentration and Hounsfield units results in a linear relationship with correlation coefficients (slopes) of 15 and 4 Hounsfield units/(mg/mL), for carboplatin in tissue and polymer, respectively. Noninvasive monitoring of local pharmacokinetics in normal and ablated tissues indicates that ablation before local carboplatin delivery increases the retention of carboplatin within the polymer matrix and drastically increases the drug retention in the ablated tissue volume (over 3-fold difference) resulting in a higher average dose to the surrounding tissue. At 1.6 mm from the implant boundary, carboplatin concentration is significantly higher in ablated tissue at 48, 96, and 144 hours (P <.05), and reaches 4.7 mg/mL in ablated tissue at 48 hours. In comparison, the concentration in normal liver at 1.6 mm reaches only 0.7 mg/mL at the same time point. The drug penetrates 3.1 mm in ablated liver compared with 2.3 mm in normal liver also at 48 hours. After 144 hours, the drug is still detected at 3.1 mm in ablated liver but not in normal liver. The differences are significant (P <.05) at both 48 and 144 hours. Correlation with chemical analysis suggests that CT data accurately predicts the drug pharmacokinetics in both ablated and normal livers. This work shows that X-ray CT imaging is a useful and promising technique for in vivo monitoring of the release kinetics of locally delivered radiopaque agents.
... Thus, Weinberg et al. demonstrated that these DOX-containing implants were able to provide high DOX concentration at the tumor site, resulting in significant decrease in tumor size compared to the untreated control ( Figure 8) [99]. These PLGA millirods have also been used to deliver other chemotherapy drugs such as carboplatin [102] and 5-fluorouracil [103]. ...
... Thus, Weinberg et al. demonstrated that these DOX-containing implants were able to provide high DOX concentration at the tumor site, resulting in significant decrease in tumor size compared to the untreated control ( Figure 8) [99]. These PLGA millirods have also been used to deliver other chemotherapy drugs such as carboplatin [102] and 5-fluorouracil [103]. [104]. ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is the fifth most common type of cancer diagnosed and the second leading cause of death worldwide. Despite advancement in current treatments for HCC, the prognosis for this cancer is still unfavorable. This comprehensive review article focuses on all the current technology that applies biomaterials to treat and study liver cancer, thus showing the versatility of biomaterials to be used as smart tools in this complex pathologic scenario. Specifically, after introducing the liver anatomy and pathology by focusing on the available treatments for HCC, this review summarizes the current biomaterial-based approaches for systemic delivery and implantable tools for locally administrating bioactive factors and provides a comprehensive discussion of the specific therapies and targeting agents to efficiently deliver those factors. This review also highlights the novel application of biomaterials to study HCC, which includes hydrogels and scaffolds to tissue engineer 3D in vitro models representative of the tumor environment. Such models will serve to better understand the tumor biology and investigate new therapies for HCC. Special focus is given to innovative approaches, e.g., combined delivery therapies, and to alternative approaches—e.g., cell capture—as promising future trends in the application of biomaterials to treat HCC.
... To address this limitation, biodegradable polymer implants were designed to release chemotherapeutic drugs into tumors after RF ablation. These polymer implants in the form of millirods have been tested in normal liver tissue [20,23] as well as in experimental liver tumors in rabbits [28,29]. Despite promising in vivo results, the clinical use of millirods may be limited by the inability to deliver a drug to distal regions of the remnant tumor. ...
Article
Radiofrequency ablation has emerged as a minimally invasive option for liver cancer treatment, but local tumor recurrence is common. To eliminate residual tumor cells in the ablated tumor, biodegradable polymer millirods have been designed for local drug (e.g., doxorubicin) delivery. A limitation of this method has been the extent of drug penetration into the tumor (<5 mm), especially in the peripheral tumor rim where thermal ablation is less effective. To provide drug concentration above the therapeutic level as needed throughout a large tumor, implant strategies with multiple millirods were devised using a computational model. This dynamic, 3-D mass balance model of drug distribution in tissue was used to simulate the consequences of various numbers of implants in different locations. Experimental testing of model predictions was performed in a rabbit VX2 carcinoma model. This study demonstrates the value of multiple implants to provide therapeutic drug levels in large ablated tumors.
... 19 -21 Goldberg et al 22 have shown that both intravenous and intralesional administration of drug either immediately before or after RF ablation can lead to an increase in the coagulation diameter relative to ablation alone. In addition, Szymanski-Exner et al 23 have shown that ablation increases the retention time of carboplatin released from intralesional PLGA implants by slowing clearance caused by perfusion. Furthermore, in a limited patient trial, 10 patients were given intravenous Doxil before percutaneous ablation, and increased necrosis was documented in the tumor site 2-4 weeks after treatment compared with RF only. ...
Article
Full-text available
The purpose of this study was to investigate whether an intralesional chemotherapy depot with or without a chemosensitizer could improve the efficacy of radiofrequency (RF) ablation in treatment of experimental carcinoma in rats. Eighteen BD-IX rats were inoculated with bilateral subcutaneous tumors via injection of DHD/K12TRb rat colorectal carcinoma cells in suspension. Four weeks after inoculation, one tumor in each rat was treated with RF ablation at 80 degrees C for 2 minutes and the other with RF ablation followed by intralesional chemotherapy with carboplatin. The drug was administered via 2 different in situ-forming poly(D,L-lactide-coglycolide) (PLGA) depot formulations either with or without a chemosensitizer. Treatment efficacy was assessed by comparing the change in tumor diameter compared with control, percent of coagulation necrosis and a rating of treatment completeness. Tumors treated with ablation and carboplatin + sensitizer (n = 9) showed a diameter decrease of 49.4 +/- 24.5% at the end point relative to ablation control, while those treated with ablation and carboplatin only (n = 8) showed a 7.1 +/- 12.6% decrease. Use of sensitizer also showed increased tissue necrosis (81.9 +/- 9.7% compared with 68.7 +/- 26.7% for ablation only) and double the number of complete treatments (6/9 or 66.7%) compared with ablation control (3/9 or 33.3%). From these results, we conclude that intralesional administration of a carboplatin and sensitizer-loaded polymer depot after RF ablation has the potential to improve the outcome of ablation by increasing effectiveness of local adjuvant chemotherapy in preventing progression of tumor unaffected by the ablation treatment.
Article
Full-text available
This is the fifteenth ASU to review literature relevant to clinical and biological specimens, foods and beverages. Two of the original three writers are still involved in preparing this Update and the other authors have also been part of the team for several years. However, in accepting a new role within the MAFF Central Science Laboratory Linda Owen will no longer be able to write for the review and we shall miss her valuable contribution. Although there have been many changes and important innovations during the fifteen years most have been gradual developments which were discerned after a few years' consolidation. As far as this year is concerned, those items which we identified in the last review—in vivo analyses by XRF, high resolution ICP-MS and electrospray MS—are featured again. Perhaps the most exciting of the emerging techniques is capillary electrophoresis for separation/speciation coupled to a detection system such as ICP-MS. The potential of these approaches has been hinted at in the last year or so but, with the development of effective coupling technology, results are now beginning to appear.
Article
Full-text available
beta-Lapachone (ARQ 501, a formulation of beta-lapachone complexed with hydroxypropyl-beta-cyclodextrin) is a novel anticancer agent with selectivity against prostate cancer cells overexpressing the NAD(P)H:quinone oxidoreductase-1 enzyme. Lack of solubility and an efficient drug delivery strategy limits this compound in clinical applications. In this study, we aimed to develop beta-lapachone-containing polymer implants (millirods) for direct implantation into prostate tumors to test the hypothesis that the combination of a tumor-specific anticancer agent with site-specific release of the agent will lead to significant antitumor efficacy. Survival assays in vitro were used to test the killing effect of beta-lapachone in different prostate cancer cells. beta-Lapachone release kinetics from millirods was determined in vitro and in vivo. PC-3 prostate tumor xenografts in athymic nude mice were used for antitumor efficacy studies in vivo. beta-Lapachone killed three different prostate cancer cell lines in an NAD(P)H:quinone oxidoreductase-1-dependent manner. Upon incorporation of solid-state inclusion complexes of beta-lapachone with hydroxypropyl-beta-cyclodextrin into poly(D,L-lactide-co-glycolide) millirods, beta-lapachone release kinetics in vivo showed a burst release of approximately 0.5 mg within 12 hours and a subsequently sustained release of the drug ( approximately 0.4 mg/kg/d) comparable with that observed in vitro. Antitumor efficacy studies showed significant tumor growth inhibition by beta-lapachone millirods compared with controls (P < 0.0001; n = 10 per group). Kaplan-Meier survival curves showed that tumor-bearing mice treated with beta-lapachone millirods survived nearly 2-fold longer than controls, without observable systemic toxicity. Intratumoral delivery of beta-lapachone using polymer millirods showed the promising therapeutic potential for human prostate tumors.
Article
To evaluate the use of 5-fluorouracil (5-FU)-laden polymer implants as an adjunct to radiofrequency (RF) ablation for tumor treatment. All animal studies were performed in compliance with the Case Western Reserve University Institutional Animal Care and Use Committee guidelines. Three studies were performed to investigate (a) in vitro dissolution of 5-FU-laden polymer implants in saline and bovine serum, (b) tissue distribution of 5-FU and its metabolite, 5-fluorouridine (5-FUrd), in the ablated liver tissue of rats (n = 4), and (c) efficacy of combined approach (n = 4) compared with that of ablation alone (n = 6) for VX2 liver tumor model in rabbits. Characterization of 5-FU release in vitro and distribution of 5-FU in rat liver tissue were analyzed by using high performance liquid chromatography; in vivo efficacy was assessed by using computed tomography and pathologic examination. Results of the in vitro dissolution study showed that a 75% release of 5-FU occurred in 2 days when exposed to bovine serum and in 9 days when exposed to phosphate-buffered saline. In the ablated rat liver, the 5-FU level was higher at the center and lower at the periphery of the tissue both at 24 hours (41.0 mg per kilogram tissue vs 15.0 mg per kilogram tissue, respectively) and at 48 hours (8.0 mg per kilogram tissue vs 2.0 mg per kilogram tissue, respectively). The 5-FUrd concentration was twofold higher peripherally than centrally and was higher at 48 hours than at 24 hours. In rabbits, local delivery of 5-FU immediately after RF ablation provided a significant (P < .05) reduction in tumor size compared with ablation alone (1.80 cm3 +/- 0.28 [standard error] vs 3.53 cm3 +/- 0.52, respectively; P = .034) and a more than 20-fold reduction in tumor size compared with the control (1.80 cm3 +/- 0.28 vs 41.95 cm3 +/- 11.58, respectively; P = .018). Combined treatment by using 5-FU polymer implants and RF ablation shows uniform sustained release of 5-FU for 48 hours at least 8 mm from the edge of the ablation zone and appears to be successful at controlling the growth of an experimental tumor in rabbits appreciably better than does ablation alone.
Article
Full-text available
Previously, biodegradable polymer implants (polymer millirods) to release chemotherapeutic agents directly into tumors have been developed. The purpose of this study is to evaluate local drug distribution from these implants in liver tumors treated with radiofrequency (RF) ablation and determine if the implants provide a therapeutic improvement over RF ablation alone. Cylindrical implants were fabricated using 65% poly(D,L-lactide-co-glycolide) (PLGA), 21.5% NaCl, and 13.5% doxorubicin. Control or drug-containing millirods were implanted inside VX2 liver tumors (11 mm diameter) in rabbits after RF ablation. Therapeutic efficacy was assessed 4 and 8 days after treatment using tumor size, histology, and fluorescence measurement of drug distribution. Tumors in both test groups recurred at the boundary of the ablated region. Therapeutic doxorubicin concentrations were found in more than 80% of the ablated area, but concentrations declined rapidly at the boundary between normal and ablated tissue. This region was characterized by a developing fibrous capsule with resolving inflammation, which restricted drug transport out of the ablated zone. The intratumoral doxorubicin implants delivered high concentrations of drug within the ablated region but only limited amounts outside the ablation zone. Future studies will focus on overcoming the fibrotic transport barrier and enhancing drug delivery to the periphery of the ablation region to prevent tumor progression.
Article
Full-text available
To find effective chemoembolization mixtures, we tested combinations of carboplatin with the embolizates Spherex and Gelfoam in comparison to a therapy with NaCl-solution, a treatment with the cytostatic drug only, and a therapy with each of the embolizates alone. The experiments were carried out using as a model the VX2 tumor in the liver of male chinchilla rabbits (five for each group). Carboplatin was revealed by the 3-(4,5-dimethylthiazole-2)-yl-2,5-diphenyltetrazolium bromide test to be a potent cytostatic drug for VX2 rabbit tumor cells. We used magnetic resonance imaging to examine the tumor volume and signal intensity enhancement up to 15 min after Gd-DTPA administration within the tumor and liver before and after the different therapies. These parameters allowed us to evaluate tumor growth and vitality as well as liver injury for the different therapy types. The results found by magnetic resonance imaging corresponded very well to those obtained by histological analysis of the tumors. The chemoembolization therapies were significantly more efficient than the other therapies, as indicated by the reduction of signal intensity enhancement after contrast agent administration within the tumor and by the histologically determined necrotic fraction after therapy. In addition, we found a significant decrease of the tumor volume and no significant live injury for a therapy with Carboplat and Gelfoam.
Article
Origins in imaging, procedural emphasis, and dependence on innovation characterize interventional radiology, which will continue as the field of image-guided minimally invasive therapies. A steady supply of innovators will be needed. Current workforce shortages demand that this problem be addressed and in an ongoing fashion. Interventional radiology’s major identity problem will require multiple corrective measures, including a name change. Diagnostic radiologists must fully embrace the concept of the dedicated interventionalist. Interspecialty turf battles will continue, especially with cardiologists and vascular surgeons. To advance the discipline, interventional radiologists must remain involved in cutting-edge therapies such as endograft repair of aortic aneurysms and carotid stent placement. As the population ages, interventionalists will experience a shift toward a greater emphasis on cancer treatment. Political agendas and public pressure will improve access to care and result in managed health car...
Article
A simple procedure is described for the determination of platinum in tumours after cisplatin therapy. Tumours were digested in 65% nitric acid by incubation at 37°C for 2 days and platinum analysed under optimum conditions by electrothermal atomic absorption spectrometry with Zeeman background correction. The reproducibility of measurements in general was better than ±2%. The calibration graph was linear from 30.0 up to 1000 μg l−1 of platinum, while the limit of detection (3σ) was found to be 3.0 μg l−1 (sample volume 20 μl). Aqueous standard solutions and the standard addition method were applied in the calibration procedure. Under the recommended analytical conditions, the sample matrix did not influence the determination of platinum significantly. In 72% of samples analysed the differences between results obtained by the two calibration procedures did not exceed ±5%.
Article
This clinical trial was performed to evaluate the safety and feasibility of interactive MR-guided radiofrequency (RF) interstitial thermal ablation (ITA) performed entirely within the MR imager. RF-ITA was performed on 11 intra-abdominal metastatic tumors during 13 sessions. The RF electrode was placed under MR guidance on a .2-T system using rapid fast imaging with steady state precession (FISP) and true FISP images. A custom 17-gauge electrode was used and was modified in four sessions to allow circulation of iced saline for cooling during ablation. Tissue necrosis monitoring and electrode repositioning were based on rapid T2-weighted and short-inversion-time inversion recovery (STIR) sequences. Morbidity and toxicity were assessed by clinical and imaging criteria. The region of tissue destruction was visible in all 11 tumors treated, as confirmed on subsequent contrast-enhanced images. No significant morbidity was noted, and patient discomfort was minimal. In conclusion, interactive MR-guided RF-ITA is feasible on a clinical .2-T C-arm system with supplemental interventional accessories with only minor patient morbidity. The ability to completely ablate tumors with RF-ITA depends on tumor size and vascularity.
Article
Radiofrequency (RF)-induced tissue coagulation represents a new approach for the thermal destruction of tumors within the liver. The purpose of the current study was to 1) assess technique safety; 2) determine the extent and evolution of induced cellular damage; and 3) correlate the observed pathologic effects with radiologic studies. Twenty-three tumors measuring ≤ 8 cm (19 colorectal metastases and 4 hepatomas) in 22 patients were treated with RF (range, 500–1550 milliamperes) using internally cooled electrodes. All treated tumors were resected to allow pathologic analysis. Eleven tumors were treated intraoperatively under ultrasonographic guidance and excised immediately. Twelve tumors were treated percutaneously using ultrasound or computed tomography (CT) guidance and subsequently were excised 3–7 days after ablation. Contrast-enhanced CT (n = 12) and magnetic resonance imaging (MRI) (n = 2) were performed after ablation of all percutaneously treated patients. Tumors treated intraoperatively did not demonstrate definitive coagulative necrosis. However, pathologic abnormalities suggestive of tissue injury were observed with hematoxylin and eosin staining, and absent cytosolic and mitochondrial enzyme activity suggested irreversible cellular damage. In contrast, specimens removed > 3 days after ablation showed definite, contiguous coagulative necrosis without intervening areas of viable tumor. CT and MRI scans demonstrated circumscribed hypodense, nonenhancing regions surrounding the electrode tract as early as 15 minutes after ablation. These corresponded within 2 mm to measurements of coagulation at pathology. RF ablation is a minimally invasive and safe approach to the treatment of tumors in the liver. Tumors treated with RF energy do not immediately demonstrate coagulative necrosis, but do show evidence of irreversible cellular damage. The extent of tumor necrosis correlates closely with findings at contrast-enhanced imaging. Cancer 2000;88:2452–63.
Article
A compression-heat molding procedure was developed to fabricate poly(D,L-lactide-co-glycolide) (PLGA) controlled release drug delivery devices for the local treatment of tumors. The drug delivery devices were designed in the shape of a cylindrical millirod (1.6-mm diameter, 10-mm length), which allows them to be implanted by a modified 14-gauge tissue biopsy needle into tumor tissues via image-guided interventional procedures. In this study, the prototype trypan blue-containing PLGA millirods were fabricated under a compression pressure of 4.6 x 10(6) Pa and different fabrication temperatures for 2 h. The scanning electron microscopy results showed complete polymer annealing for millirods fabricated at 80 and 90 degrees C, while the cross sections of the 60 and 70 degrees C millirods showed incompletely annealed PLGA microspheres and trypan blue powders. The density, flexural modulus, and release properties of the PLGA millirods were also characterized and compared. The average values of the density and flexural modulus of the millirods increased with an increase in fabrication temperature. The flexural modulus values of most PLGA millirods were above 1 x 10(8) Pa, which provides sufficient stiffness for implantation within the tumor tissue. In addition, a Delta c(p) method was developed to determine the loading density of trypan blue in the PLGA millirods by differential scanning calorimetry. Results from the Delta c(p) measurement showed that trypan blue was homogeneously distributed in the millirod. Release studies in phosphate-buffered saline showed that the release rate decreased for the millirods fabricated at higher temperatures. The times for the release of 50% trypan blue were 5, 25, 25, and 25 h for millirods fabricated at 60, 70, 80, and 90 degrees C, respectively. Millirods fabricated at 90 degrees C had the most reproducible release profiles. The results from this study established compression--heat molding as an effective method to fabricate controlled release PLGA millirods with sufficient mechanical strength and reproducible release profiles for local cancer therapy.
Article
Carboplatin shares some of the therapeutic advantages of cisplatin, but without a significant incidence of the dose-limiting neurotoxicity and nephrotoxicity which is experienced with cisplatin. However, its use is associated with dose-limiting bone marrow suppression. Carboplatin is present in the blood as 3 distinct species. These are total platinum and 2 unbound species, carboplatin itself and a decarboxylated platinum-containing degradation product. The 2 main methods used to assay the unbound species are flameless atomic absorption spectrophotometry and high performance liquid chromatography. The first of these methods assays both unbound platinum species, the second is specific for carboplatin. Both unbound species have similar pharmacokinetic profiles for the first 12 hours post-dose. Carboplatin appears to have a linear pharmacokinetic profile over the doses used clinically and does not interact significantly with drugs that are used commonly in combination chemotherapy. The pharmacokinetics of carboplatin are adequately described by an open 2-compartment model with elimination from the central compartment. Its clearance is proportional to the glomerular filtration rate and the volume of distribution of the central compartment appears to correlate with extracellular fluid volume. The elimination half-life varies with renal function and is typically between 2 and 6 hours in patients with a normal glomerular filtration rate and may be as long as 18 hours in patients with impaired renal function. Relationships between systemic exposure to carboplatin, described as the area under the concentration-time curve (AUC), and both toxicity and response have been described. For toxicity the strongest evidence exists for a relationship between AUC and thrombocytopenia. To a lesser extent the relationship between AUC and neutropenia has also been described. Patients already treated with platinum analogues have been shown to develop a greater degree of myelosuppression from any given AUC. In addition, some evidence suggests a relationship between the shape of the concentration-time curve and myelotoxicity, where constant infusions appear less likely to cause myelosuppression on a mg/m2 dose administration basis. The relationship between AUC and response rate is not as clear, this may be related to the lack of studies describing both the dose and AUC of carboplatin. There appears to be a more clearly defined AUC-response relationship for ovarian cancer than for other malignancies, with an AUC of between 5 and 7 mg/ml · min being associated with the maximal response rate [located at the plateau on an AUC-response curve]. However, new data suggest that higher AUCs may lead to greater response rates. Data from testicular cancer also strongly supports an AUC-response relationship with an increased number of treatment failures with carboplatin AUCs < 5 to 6 mg/ml · min. Given the AUC-effect relationships described above a number of studies have been performed to develop models to describe the relationship between both dose and AUC and dose and platelet nadir. In adults, perhaps the most common method is that of Calvert which describes the relationship between dose and AUC. Paediatric formulas have also been described. More recently a number of limited sampling strategies have been proposed as well as Bayesian dose individualisation techniques.
Article
The purpose of this study was to determine the suitability of MRI to accurately detect radiofrequency (RF) thermoablative lesions created under MR guidance. In vivo RF lesions were created in the livers of six New Zealand White rabbits using a 2-mm-diameter titanium alloy RF electrode with a 20-mm exposed tip and a 50-W RF generator. This was performed using a 0.2T clinical C-arm MR imager for guidance and monitoring. Each animal was sacrificed and gross evaluation was performed. Histologic correlation was performed on the first two animals. The MR-compatible RF electrode was easily identified on rapid gradient-echo images used to guide electrode placement. A single lesion was created in each rabbit liver. Lesions ranged from approximately 10 to 17 mm in diameter (mean, 13.5 mm). T2-weighted and short T1 inversion recovery (STIR) images demonstrated lesions ranging in diameter from 12 to 18 mm (mean, 14.6 mm). Lesion dimensions determined from images closely correlated with those determined at gross examination with the discrepancy never exceeding 2 mm, for an r2 value of .87. MRI performed at the time of MR-guided RF ablation accurately demonstrated created lesions. This modality may provide a new option for the treatment of local and regional neoplastic disease.
Article
This clinical trial was performed to evaluate the safety and feasibility of interactive MR-guided radiofrequency (RF) interstitial thermal ablation (ITA) performed entirely within the MR imager. RF-ITA was performed on 11 intra-abdominal metastatic tumors during 13 sessions. The RF electrode was placed under MR guidance on a .2-T system using rapid fast imaging with steady state precession (FISP) and true FISP images. A custom 17-gauge electrode was used and was modified in four sessions to allow circulation of iced saline for cooling during ablation. Tissue necrosis monitoring and electrode repositioning were based on rapid T2-weighted and short-inversion-time inversion recovery (STIR) sequences. Morbidity and toxicity were assessed by clinical and imaging criteria. The region of tissue destruction was visible in all 11 tumors treated, as confirmed on subsequent contrast-enhanced images. No significant morbidity was noted, and patient discomfort was minimal. In conclusion, interactive MR-guided RF-ITA is feasible on a clinical .2-T C-arm system with supplemental interventional accessories with only minor patient morbidity. The ability to completely ablate tumors with RF-ITA depends on tumor size and vascularity.