You are on page 1of 20

Review

Therapeutic
For reprint orders, please contact reprints@future-science.com
Delivery
Treatment of intermediate stage
hepatocellular carcinoma: a review of
intrahepatic doxorubicin drug-delivery
systems

The biopharmaceutical properties of doxorubicin delivered via two drug-delivery Ilse R Dubbelboer, Elsa
systems (DDSs) for the palliative treatment of unresectable hepatocellular carcinoma Lilienberg, Emelie Ahnfelt,
were reviewed with relation to the associated liver and tumor (patho)physiology. Erik Sjögren, Niklas Axén &
These two DDSs, doxorubicin emulsified with Lipiodol® and doxorubicin loaded into Hans Lennernäs*
Department of Pharmacy, Uppsala
DC Bead® are different regarding tumor delivery, release rate, local bioavailability, if University; Box 580, SE-751 23 Uppsala,
and how they can be given repeatedly, biodegradability, length of embolization and Sweden
safety profile. There have been few direct head-to-head comparisons of these DDSs, *Author for correspondence:
and in-depth investigations into their in vitro and in vivo performance is warranted. Tel.: +46 18 471 4317
Fax: +46 18 471 4223
hans.lennernas@farmaci.uu.se
The global incidence of liver cancer is increas- To date, several DDSs delivering various
ing and this type of cancer has a poor progno- chemotherapeutic drug(s) and pharmaceu-
sis and is ranked as the third most common tical excipients have been developed and
lethal cancer form [1,2] . The recommended clinically evaluated for TACE therapy [5] .
treatment for hepatocellular carcinoma A review of these options of TACE DDSs
(HCC), a primary liver cancer, is dependent is warranted in that it would form the basis
on the stage of the disease [3] . For unresect- for optimizing tumor-targeted therapy for
able, intermediate stage HCC, transarterial HCC treatment, and subsequent develop-
chemoembolization (TACE) is recom- ment of novel, tumor-targeted DDSs and
mended. TACE involves the delivery of the dosing strategies. Two common DDSs used
cytostatic agent(s) in a drug-delivery system for TACE therapy for intermediate stage
(DDS) to the tumor via the hepatic artery (HA) HCC involve cytostatic agents emulsified
[3] . In contrast to normal liver tissue, which in Lipiodol® (LIP) or loaded into drug-
has a dual blood supply from the HA and the eluting beads. LIP is an iodized poppy seed
portal vein [4] , HCC is typically mainly vascu- oil derivative visible on x-ray, and after
larized by the HA. Local drug administration administration, the contrast is preferentially
by the TACE DDS is expected to increase the retained in tumor tissue [6,7] . The combina-
specificity of the tumor response while reduc- tion of the cytostatic agent emulsified in
ing the frequency of side effects and morbidity LIP can be administered with or without
compared with systemic dosed treatments [4] . additional embolizing materials, generat-
Embolization, caused by the DDS, obstructs ing complete or partial embolization of the
the blood flow in the tumor, induces hypoxia, targeted tumor-feeding vessel(s) [3,8] . DC
and results in increased drug concentrations Bead® (DCB) is one of several commercially
and prolonged residence times in the tumor- available drug-eluting embolizing bead sys-
target area [4] . Patients with untreated HCC tems [9] . Positively charged chemotherapeu-
that has not invaded the portal vein or spread tic drugs can be loaded into DCB for sub-
extrahepatically have an expected median sur- sequent local release [9] . The beads have a
vival of approximately 16 months. With cur- double function, serving as both a DDS and
rent palliative TACE strategies, the expected an embolic material, and result in complete,
median survival is prolonged by approximately nonreversible embolization in the targeted
part of
4 months [3] . vessels [9,10] .

10.4155/TDE.14.11 © 2014 IR Dubbelboer, E Lilienberg, Therapeutic Delivery (2014) 5(4), 447–466 ISSN 2041-5990 447
E Ahnfelt, E Sjögren, N Axén & H Lennernäs
Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

Keyterms DOX
DOX is the only cytostatic agent that is used in both
Hepatocellular carcinoma: Has a global incidence of LIP emulsion and DCB DDSs and is the dominating
6% and is the third most common cause of cancer related
death. The primary risk factor is liver cirrhosis, primarily cytotoxic agent for intermediate-stage HCC. Its physi-
caused by hepatitis B virus and C virus, or by other diseases cochemical properties are presented in Table 1. Chemi-
such as alcoholic liver disease, nonalcoholic steatohepatitis, cally, DOX is an amphiphilic active pharmaceutical
hemochromatosis, primary biliary cirrhosis and autoimmune
ingredient often used as its HCl salt in pharmaceuti-
hepatitis.
cal formulations. It consists of a daunosamine group
Lipiodol® : Iodinated and ethylated poppy seed oil, (sugar component) attached to an a glycone by a glyco-
consisting of ethyl linoleate, ethyl oleate, ethyl palmitate
and ethyl stearate, that contains 37–39% iodine.
sidic bond (Figure 1) [15] . DOXHCl is sparingly water
soluble (10 mg/ml at unspecified pH) and DOX parti-
DC Bead® : Polyvinyl alcohol-based hydrogel bead system tions between octanol and water in a pH-dependent
that can load doxorubicin or irinotecan and is available in
different sizes ranging from 70 up to 700 μm.
manner, in accordance with its three pKa values and
alkaline functions (logD7.4 = 2.4; Figure 2 and Table 1).
Transarterial chemoembolization: An interventional The polar surface area of 222.9 Å 2 is relatively large
radiology procedure that involves the injection of
embolizing drug-delivery systems containing a cytostatic when considering membrane transport properties [16] .
agent into the vicinity of the tumors via the femoral Crystalline DOX is hygroscopic and DOX forms an
and hepatic artery. The injection may be followed by an orange-red solution in water [17] .
additional infusion of an embolizing agent to impede the DOX is an anthracycline, antibiotic, antineoplastic
blood flow locally.
drug, which is indicated for multiple forms of cancer
LIPDOX: Drug-delivery system consisting of an aqueous such as bone sarcomas, Hodgkin’s disease, and ovary,
solution of doxorubicin emulsified with Lipiodol.
breast, and lung cancer [23] . In addition, DOX is used
DCBDOX: Drug-delivery system with doxorubicin loaded off-label to treat intermediate HCC. The pharmaco-
into DC Bead. logical effects of DOX appear to be mediated by at
least three antitumor mechanisms: reversible binding
These two DDSs were chosen for review as they to topoisomerase I and II, intercalation to DNA base
are both clinically used to deliver the chemotherapeu- pairs, and free-radical generation, which causes DNA
tic drug doxorubicin (DOX), a potent agent against damage [15] . The concentration of DOX required for
HCC. Recent clinical reports suggest that DOX deliv- 50% growth inhibition (IC50) in vitro is both time-
ered via these two DDSs has a similar antitumor effect, and cell-line-dependent [24] . IC50-values decreased
but different adverse event profiles [11–14] . LIP delivery from 3.8–0.029 μM in human HCC Hep-G2 cells
of DOX was associated with higher incidence of DOX- and from 8.8–1.13 μM in human HCC Hep3B cells
related side effects than DCB delivery (such as alope- over 36–72 h [25–27] . For 13 different HCC cell lines,
cia and myelosuppression), probably related to higher the IC50values from 72 h incubations ranged from
systemic exposure of DOX and its active metabolite 0.024–6.0 μM [27–29] .
doxorubicinol (DOXol). However, the total number When DOX is administered as an intravenous (i.v.)
of serious adverse events following the two treatments infusion, doses ranging from 60–75 mg/m2 body
did not significantly differ [11] . Another study showed surface area (BSA) or 1.2–2.4 mg/kg are repeatedly
that TACE with drug-eluting beads is associated with administered every 3 or 4 weeks [23] . Systemic exposure
a 6.6-times higher risk of liver/biliary events (i.e., to DOX and its active metabolite DOXol increases the
dilated bile duct, portal vein narrowing, portal venous risk of severe side effects such as cardiomyopathy and
thrombosis and biloma/liver infarct) compared with myelosuppression [23] . Suggested risk factors for cardio-
TACE and LIP as a DDS [13] . It is obvious that sev- myopathy include: cumulative DOX doses exceeding
eral important pharmaceutical and biopharmaceuti- 500 mg/m2 BSA and high peak blood concentrations
cal properties of these DDSs differ, with significant of DOX [30,31] .
implications for the treatment benefit. The objective In vitro studies in Caco-2 cell lines indicate that
of this review was to examine the pharmaceutical the intestinal epithelium is poorly permeable to DOX
and biopharmaceutical properties of the two selected (0.1 × 10 -6 cm2 /s), which is in accordance with its rela-
DDSs. The general characteristics of DOX solutions tively high polar surface area [32,33] . Like many other
emulsified in LIP (LIPDOX) and DOX loaded into drugs, DOX is transported across cell membranes by
DCBs (DCBDOX) are described in detail, and fac- both passive diffusion and carrier-mediated (CM)
tors influencing the in vitro release, in vivo release and transport [34] . It has recently been reported that the
liver pharmacokinetics (PK) of DOX are thoroughly solute-carrier influx transporter SLC22A16 is involved
discussed. in the influx of DOX into cells [35] . Several CM efflux

448 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

Table 1. The physicochemical properties of doxorubicin.


Parameter Value Ref.
Molecular weight (g/mol) 543.53 [21]

Polar surface area (Å ) 2


222.89 [21]

pKa 7.34, 8.46, 9.46 [22]

LogD    
pH 4.8 -0.91 [19]

pH 5.8 -0.45 [19]

pH 6.8 0.18 [19]

pH 7.2 0.71 [19]

pH 7.5 2.42 ± 0.08 [20]

Solubility† (mg/ml) 10 [21]

Hydrogen bond donor 6 [21]

Hydrogen bond acceptor 12 [21]



Solubility is presented for the HCI salt, at an unknown pH.
LogD: Logarithm of the distribution coefficient; pKa: Acid dissociation constant.

proteins, including ATP-binding cassette (ABC) ered a minor route of elimination as only 3–4% of an
transporters such as ABCB1, ABCC1, ABCC2 and i.v. DOX dose (60 mg/m2) was excreted within 5 days
ABCG2, and Ral binding proteins such as RALBP1, of administration in one study [43] .
have been associated with DOX transport [36] .
The pKa values of DOX indicate that a maximum Lip
50% of the DOX dose is ionized in the physiological The fate of LIP in the hepatic vascular system
pH range. The level of ionization controls the lipophilic LIP (also known as Ethiodol®) is an iodinated and
partitioning (Log Doctanol:buffer) of DOX in both patho- ethylated poppy seed oil that consists of ethyl linole-
logical and normal physiological pH ranges (Figure 2) ate (70% w/w), ethyl oleate (15% w/w), ethyl palmi-
[19,20] . The pH of intracellular fluid is reported to be tate (10% w/w), ethyl stearate (5% w/w), and contains
higher than that of extracellular fluid in some tumors, 37–39% iodine (400 mg/ml) [44,45] . Following injec-
while the opposite is the case in healthy tissue [18] . tion into the HA, LIP is accumulated in tumor tis-
Thus, once DOX has been distributed into the cells in sue for at least 3 months, as lipid layers (nonglobular)
the tumor tissue, it may bind more extensively to lipid extracellularly and as droplets (globular) intracellularly
structures than it would in healthy tissue as the ionized (Figure 3) [7,46] . This preferential localization in tumor
fraction is greater [15] . tissue initiated the pharmaceutical development of an
DOX is reported to be extensively bound to the intrahepatic DDS based on LIP for the treatment of
tissue compartment (e.g., to liver, kidneys, heart and unresectable HCC [6–7,46–47] . Other important prop-
lungs), which is reflected in its large volume of distri- erties of LIP are a density of 1.280 g/cm3 (at 15°C)
bution (22 l/kg) [37,38] . DOX is distributed to healthy and a viscosity of 25 mPa/s (at 37°C), properties that
tissues at a higher extent than to tumor tissue, this is are thought to influence the in vivo performance of the
most likely explained by the tumor vasculature, for DDS. The density of LIP is an important factor for
example irregular or static blood flow [39] . The PK
of DOX are best described by a three-compartment H
O H
model, with a terminal half-life of 35 h [23,37] . DOX is O H
metabolized into six metabolites; active DOXol is the O O

most abundant of these [40] . The biotransformation to O


H
DOXol occurs mainly in hepatocytes, but also extrahe- O O O O O
H
patically, via the cytosolic enzymes aldoketo-reductase H
H H H
and carbonyl-reductase [41] . The biliary elimination H O
route dominates for both DOX and DOXol. After an N
H H
i.v. dose of DOX, 25% of the dose is excreted into the
bile as the parent drug and 25% as metabolites (mainly
DOXol) within 7 days [42] . Renal excretion is consid- Figure 1. Doxorubicin.

future science group www.future-science.com 449


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

by the perfusing blood. There was a higher content


pHt,e pHt,i of oil in the liver tissue, slower clearance (removal)
3 of oil from the liver, increased embolizing effects (as
blood flow was reduced), and more water droplets in
2 the oil phase after a w/o emulsion than after an oil-
in-water (o/w) emulsion infusion [8,52] . When a w/o
LogD(oct/buffer)

1
emulsion was infused into the HA in tumor-bearing
rats and mice, the oil phase was preferentially located
in the liver tumor vessels [52] . The accumulation of
0 linoleic acid (main ingredient in LIP) to hypervas-
5 6 7 8
cular VX2 tumors in rabbits is shown in Figure 3
-1 [6] . The oil phase of a w/o emulsion contained more
pH pHh,i pHh,e
water droplets when administered to hypervascular
tumors than to hypovascular tumors, possibly due to
Figure 2. Experimentally determined distribution higher vessel density in hypervascular tumors [52] . A
coefficient (logarithm) (LogD; octanol/buffer) values higher HA blood flow has been shown to increase the
for doxorubicin. The intracellular pH (pHt,i, dark gray percentage of the LIP dose distributed to the tumor
area) and extracellular pH (pHt,e, light gray area) of
[53] . The size of the oil droplet has also been corre-
tumor tissue, and intracellular pH (pHh,i, dotted area)
and extracellular pH (pHh,e, striped area) of healthy lated to the extent of intra-tumor distribution;oil
tissue are shown [18] . droplets less than 1 μm in diameter were distributed
Data taken from [19,20] . homogeneously in the vascular lumen of the tumor,
while droplets larger than 15 μm were distributed
the stability of the emulsion and the viscosity of LIP unevenly [54] . Droplets smaller than 0.1 μm appear
is about five-times higher than that of human blood to pass through the hepatic sinusoidal fenestrations
(6.1 mP/s in men, 4.5 mPa/s in women), which may in healthy tissue and are expected to behave simi-
contribute to the chemoembolization effect [48,49] . larly in tumor sinusoids [54] . Taken together, these
The available data indicate that the vascular and findings suggest the importance of implementing in
tumor distribution of LIP in vivo are affected by a vivo studies and in vitro–in vivo performances early
dynamic interplay between the size of the oil drop- in the development of pharmaceutical formulations,
lets, the size of the vasculature, and the physiological especially for parenterals.
hydrodynamics. After intrahepatic infusion of LIP into
healthy animals (mice, rats, rabbits and pigs), either The fate of LIP in hepatic extracellular
as a pure oil or emulsified with water, distribution of & intracellular tissue
LIP into the hepatic-vascular system resulted in tem- Having passed the vascular barrier, the lipids accu-
porary embolization, local hypoxia and, if the vessels mulate on the surfaces of the tumor cells, as observed
were fully embolized, atrophy of the sinusoids [50,51] . both in vitro and in vivo [7,55–56] . Based on in vitro
LIP is distributed into the sinusoids and is also shunted studies, membrane-bound droplets of LIP are
to the portal venules, where it accumulates and slows thought to enter the cell via pinocytosis (fluid endo-
the sinusoidal blood flow [50–52] . If the LIP droplets are cytosis) [45,55–58] . After resection of HCC tumors in
smaller than the vessel diameter (25–50 μm in rabbits patients treated with a LIP-cytostatic agent emulsion,
and humans) the droplets can pass through the sinu- LIP was found in both the vascular lumen and the
soids [50] . Kan et al. demonstrated that the embolizing endothelial cells of the tumors [45] . Necrosis in these
effect of LIP is reversible and that the time to complete resected liver tumors was thought to have occurred
restoration of the sinusoidal blood flow is dose-depen- as a result of drug activity and/or of hypoxia related
dent in the clinical dose range of 0.1–0.4 ml/kg. It has to embolization. In the same clinical study, resected
also been suggested that it is possible to control the in untreated tumor lesions were perfused through the
vivo droplet size of LIP by adjusting the injection rate HA with LIP ex vivo and fixed in formalin within
into the HA: the slower the injection into the HA, the 15 min. After fixation, droplets of LIP were observed
smaller the generated droplets [51] . in the vascular lumen, in endothelial cells lining the
When injected as a water-in-oil (w/o) emulsion, LIP tumor vessels, and in tumor cells, suggesting that
was observed in the portal venules and sinusoids as a LIP uptake into both tumor tissue and cells occurs
series of oil droplets containing smaller water drop- rapidly (<15 min) [45] . The distribution to, and accu-
lets [52] . The water droplets in the emulsion eventu- mulation of, LIP in tumors (Figure 3) can partly be
ally separated from the oil phase and were swept away explained by the enhanced vascular permeability

450 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

and retention (EPR) effect [59] . The EPR effect is a


500
well-known consequence of abnormal vascularization

Radioactivity (dpm/g × 10-3)


and increased vascular permeability in solid tumors 400 15 min
in combination with impaired lymphatic clearance.
3 days
Reduced lipid metabolism in the diseased liver, seen 300
7 days
as an increase in free fatty acids and a decrease in
(apo-)lipoproteins, probably contributes to further 200
tissue accumulation of LIP [60] . In addition, phago-
cytosis by Kupffer cells is down-regulated in HCC 100
tissue, which slows the elimination of lipids [61,62] .
Although these mechanisms contribute strongly to 0

or

or
or

h
le

ac
the tumor-selective accumulation of LIP, additional

Bi
m
m

om
Tu

tu
tu

to
o
processes may also exist.

St
tt

nt
en

ta
ac

is
rd
dj
Release of DOX from LIPDOX in vitro

ra

ve
ve

Li
The release of DOX from LIPDOX in vitro is affected

Li
by factors such as the partitioning of DOX between the
Figure 3. Distribution of 14C after injection of 0.2
LIP and aqueous phases, the type and stability of the ml [14C]-linoleic acid into the hepatic artery in the
LIPDOX emulsion, and the chosen in vitro test method. hypervascular VX2 tumor in rabbits. Tissues expressing
Despite extensive use of LIPDOX, the partition- less than 5 dpm/g × 10 −3 radioactivity, for example lung,
ing coefficient of DOX between LIP and the aqueous kidney and heart are not depicted.
Data taken from [6] .
phase has not yet been established. DOX is relatively
lipophilic at neutral pH (logD7.4 = 2.4), but its distribu- phase will be the rate-limiting step for in vitro release
tion between the oil and aqueous phase is pH-depen- [63,64] . In conclusion, DOX is released more slowly
dent (Figure 2) . The dependency of DOX distribution from LIPDOXw/o emulsions than from LIPDOXo/w
in LIPDOX emulsions on pH is an important factor emulsions in vitro, in accordance with the observed
that needs to be considered in vitro, since it may affect poor permeation of DOX across lipid bilayers [65] .
drug release in vivo. The release of DOX from LIPDOXw/o emulsions is
The emulsion type affects the DOX release and is also crucially influenced by the stability of the emul-
determined by the ratio between the aqueous and the sion, since the inner-aqueous phase (containing most
LIP phases. This ratio can be adjusted to form dif- of the DOX) will be in direct contact with the in vitro
ferent types of emulsions with different distribution release medium if the emulsion breaks, releasing the
and release properties. Kan et al. have reported that DOX instantly. The thermodynamical stability of
aqueous:LIP ratios of 1:2–4 generated LIPDOXw/o LIPDOX emulsions can be improved by the addition
emulsions with a water droplet diameter range of of densifiers (substances that increase the density of the
2–3 μm, and that 2–4:1 aqueous:LIP ratios formed aqueous phase) and emulsifiers [67] . When the density
LIPDOXo/w emulsions, all made with an extempora- of the aqueous phase was increased to bring it closer
neous syringe mixing technique [52] . The oil droplet to that of LIP (1.28 g/cm3), the stability period for
diameters in the LIPDOXo/w emulsions were in the LIPDOXw/o emulsions was increased from 3–24 h [69] .
range of 5–300 μm (mean around 7 μm). As the larger Various densifiers, such as 60% diatrizoate sodium
oil droplets contained smaller water droplets of unspec- meglumine (Urografin®), iohexol (Omnipaque
ified size, the emulsion was classified as water-in-oil-in- 300™), or iopamidol, have been used in LIPDOX for-
water (w/o/w) [52] . The impact of the emulsion type mulations [52,63,67,69] . Various emulsifiers with different
on in vitro release of DOX is visualized in Figure 4 and degrees of hydrophilic–lipophilic balance (HLB) have
Table 2 : DOX is released faster from LIPDOXo/w emul- also been tested for their ability to improve LIPDOX
sions than from LIPDOXw/o emulsions. These data emulsion stability. Hydrogenated castor oil 60 (HCO-
strongly indicate that DOX is preferentially distrib- 60, HLB 14–15) and Pluronic F68 (HLB 29) have
uted to the aqueous phase of the emulsion, as adding been used for this purpose [63,66–67,70] . As shown in
a LIPDOXo/w emulsion to the aqueous release medium Figure 4, release of DOX from LIPDOXw/o emulsions
resulted in the immediate release of DOX in vitro. If stabilized with emulsifiers and/or densifiers was slower
the DOX had partitioned into the LIP, it would be in vitro than that from the unstabilized LIPDOXw/o
expected to be released more slowly. Similarly, when emulsion. In vitro release of DOX from the only
a LIPDOXw/o emulsion is added to an aqueous-release LIPDOXw/o/w emulsion included in Figure 4 (Table 2)
medium, the diffusion of DOX through the lipid outer was faster than that from the LIPDOXw/o emulsions,

future science group www.future-science.com 451


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

probably because of impaired emulsion stability and The release-test method used is also known to
subsequent partitioning of DOX to the outer aque- influence the release of DOX from LIPDOX in vitro.
ous phase. In general, w/o/w emulsions are unstable Parameters affecting in vitro release include the agi-
and immediate release from LIPDOXw/o/w emulsions tation speed, the type and rate of flow, and the type
has been reported [67,71] . However, some published of membrane chosen [73] . The sample-and-separate,
sustained-release profiles indicate that LIPDOXw/o/w free-flowing, and dialysis membrane-based methods
emulsions can be stabilized [66] . are commonly used [63,66–67] .
A third factor influencing the release of DOX from Thus, for prolonged stability and optimal modi-
LIPDOX is the mixing procedure, which influences fied release of DOX from LIPDOX, the emulsion
both the stability and the type of emulsion formed. should be of the w/o type, have an aqueous:LIP ratio
Two main mixing procedures have been described: of at least 1:2, be stabilized up to 24 h by the addi-
homogenization by ultrasonic bath or homogenizer tion of densifiers and/or emulsifiers, and be prepared
and an extemporaneous pumping technique where by vigorous mixing prior to administration. Proposed
two connected syringes (one containing an aqueous in vitro DOX release mechanisms from LIPDOX are
solution of DOX and one containing LIP) are used shown in Figure 5A .
to emulsify the two phases by pumping the syringes It has been suggested that lipids may act as perme-
until an emulsion is formed [52,67] . The preparation ability enhancers by mechanisms such as increasing
technique chosen will probably influence both the in membrane fluidity, inhibiting carrier-mediated efflux
vitro and the in vivo release of DOX from the emul- transporters, increasing solubility, and/or decreas-
sion, as the intensity of mixing influences the droplet ing enzymatic hydrolysis both in vitro and in vivo
size of the inner phase, irrespective of whether the [74–76] . If so, the intracellular uptake of DOX might
inner phase is aqueous or LIP. Homogenization cre- be affected by the presence of LIP. However, most in
ated emulsions with inner droplet sizes of 10–40 μm, vitro models of cellular uptake are directly correlated
and the pumping technique created emulsions with with prolonged contact time between the formula-
inner droplet sizes of 30–120 μm in one study [8] . tion and the cells. As a consequence, it is difficult
Emulsification via ultrasound, a less used option, to accurately predict the in vivo outcomes and the
generated smaller droplets and more stable emulsions relevant mechanisms involved in tumoral uptake of
than homogenization created emulsions [72] . DOX from LIPDOX formulations based only on

100
LIPDOX I

80 LIPDOX II
Dose released (%)

LIPDOX III
60
LIPDOX IV

40 LIPDOX V

LIPDOX VI
20
LIPDOX VII

0
0 2 4 6 8 20 40 60 80

Time (h)

Figure 4. The in vitro release profiles for aqueous doxorubicin from Lipiodol® emulsions. Details of the
composition of the emulsions are provided in Table 2. Results represented by circles were released using the sample-
and-separate release method, those represented by squares were released using the T-apparatus method, and those
represented by triangles were released using dialysis membrane-based methods. Closed symbols, open symbols, and
framed closed symbols represent water-in-oil, oil-in-water and water-in-oil-in-water emulsions, respectively.
LIPDOX: Lipiodol emulsions.
Data taken from [63,66–68] .

452 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

Table 2. The composition of the aqueous doxorubicin from Lipiodol® emulsions reviewed in this
paper and the in vitro method used to study the release of doxorubicin.
Formulation Type of Concentration of Water:oil Emulsifiers Densifiers In vitro Ref.
emulsion DOX in aqueous (:water) (v/v) method
phase (mg/ml)
LIPDOX I w/o 2.6 1:3.33 – – DMB [66]

LIPDOX II NA 5.0 1:1 – – T-app [68]

LIPDOX III w/o 2.6 1:3.33 HCO-60 – DMB [66]

LIPDOX IV w/o 5.0 1:5 HCO-10 – S-S [63]

LIPDOX V w/o 4.0 1:4 HCO-60 Iopamiro DMB [67]

LIPDOX VI w/o/w 0.88 1:3.33:8.33 Pluronic F-68 – DMB [66]

LIPDOX VII o/w 5.0 1:4 †


Kaytwo Urografin ®
S-S [63]
The emulsions were composed of aqueous doxorubicin solution, Lipiodol®, emulsifiers, and/or densifiers. The in vitro release studies were
performed with NaCl 0.9% [66], distilled water [63], or phosphate-buffered saline [67,68].
Kaytwo (10 mg) not included in ratio water:oil.
DMB: Dialysismembrane-based; DOX: Doxorubicin; HCO: Hydrogenated castor oil; Kaytwo: Syrup containing vitamin K; LIPDOX: Aqueous
doxorubicin solutions emulsified in Lipiodol; NA: Not available; o/w: Oil-in-water; S-S: Sample-and-separate; T-app: T-apparatus;
w/o: Water-in-oil; w/o/w: Water-in-oil-in-water.

in vitro models [77] . Instead, a combination of rel- model, which suggests that LIP droplets containing
evant in vivo models and theoretical physiologically DOX might accumulate in the tumor. These data
based PK/pharmacodynamics models are required are supported by the higher DOX concentrations in
to better understand and predict the clinical perfor- the tumor than in the surrounding liver tissue after
mance of a DDS. intra-hepatic LIPDOXw/o infusions in both VX2 rab-
bits and HCC patients [69,79] . Lower DOX concentra-
In vivo behavior of LIPDOX in the tions in the liver tissue surrounding the tumor than
hepatobiliary system in the tumor itself are indicative of a more tumor-
When LIPDOX is administered in vivo, the droplet directed anticancer effect and fewer systemic side
sizes of both the inner and outer phases of the emul- effects. Even though co-administration of DOX with
sion need to be considered. In the blood, the w/o LIP (unknown composition) increased tumor DOX
emulsion droplet size should be smaller than 20 μm concentrations, the intra-tumor distribution of the
to enable passage through the sinusoids and facilitate iodine component of LIP did not predict the intra-
distribution within the tumor [51,54] . It is important tumor distribution of DOX [80,81] . DOX was mostly
that the aqueous phase of LIPDOX is readily dis- distributed only around the tumor blood vessels 4 h
persed in the oil phase to withstand emulsion break- after intra-arterial administration [82] . Drug penetra-
age during administration [78] . However, the in vivo tion was enhanced with LIP, but adjacent avascular
droplet size could potentially be reduced by a slower regions were never reached with DOX [82] .
infusion speed as suggested by Kan et al. [51] . There is strong in vitro and in vivo evidence indi-
In an in vivo comparison of two LIPDOXw/o emul- cating that the formulation properties of LIPDOX
sions, one stabilized with HCO-60 and iopamiro have an impact on liver disposition and presum-
and prepared using homogenization, and one not ably on the benefit:safety ratio. Surprisingly often,
stabilized and prepared using the syringe-pumping however, this crucial information on the formula-
technique, in healthy mongrel dogs [67] , the stabilized tion is not fully described and summarized in clini-
emulsion had a longer elimination half-life than the cal reports [12,53,83–86] . When it is reported, ratios
nonstabilized emulsion. This correlates well with the of aqueous:LIP appear to be in the range of 4:1 to
slower release from stabilized LIPDOXw/o emulsions 1:3.33, but are mostly 1:1 [87–92] . As suggested by the
in vitro. In another in vivo study, in VX2 carcinoma- in vitro data (Figure 4) , only aqueous:LIP ratios of
bearing rabbits, the plasma AUC 0-120min for DOX 1:2–4 will result in w/o emulsions with sustained in
was three-times lower for a stabilized LIPDOXw/o vitro release of DOX. The volume of infused LIP var-
emulsion than for a stabilized LIPDOXo/w emulsion ies widely, with literature reports of 2–20 ml. Because
and, after 24 h, intra-tumoral DOX concentrations of the many unknown factors in the preparation of
were twice as high for LIPDOXw/o [63] . This could clinically used LIPDOX emulsions, the published
be explained by the EPR effect in the rabbit tumor plasma PK profiles can provide only limited infor-

future science group www.future-science.com 453


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

mation on the effectiveness of the emulsion. Hence, ous solution of DOX is added to DCB directly after
a direct pharmaceutical comparison based on pub- removing the buffer solution. As diffusion and ion
lished LIPDOX data is virtually impossible, since exchange proceed, the positively charged primary
information on the crucial formulation properties amine of DOX forms an ionic bond with the nega-
with respect to its performance at the site of action tively charged sulfonate groups, thus replacing the
is not available. This apparent lack of consistency in sodium ions [68,99] .
the clinical use of LIP-based DDSs strongly indicates The DCB have a smooth shell containing stabi-
a need for development of a more standardized DDS. lizer (cellulose acetatebutyrate) and therefore lack
In summary, to reach high DOX concentrations ionic bonding sites [95,98–99] . In the region underlying
in the tumor and low peak-plasma concentrations, the shell, DOX is evenly distributed throughout the
LIPDOXw/o emulsions should be stabilized and have DCB at lower concentrations (up to 0.05 mg DOX/
a small inner and outer droplet size (<20 μm) to ml DCB) [98] . However, at higher concentrations
enable efficient distribution through the permeable, (from 5 mg DOX/ml DCB), two distinct regions are
leaky vascular system of the tumor. Furthermore, distinguishable with fluorescent and transmission
LIPDOX formulations used in both nonclinical and microscopy: a bright red outer layer and a black core
clinical settings should be described in greater detail region [98,100] . According to the microscopy images
to allow more meaningful comparisons between in Biondi et al., the size of the core region appears to
studies. increase and the outer layer to decrease with increas-
ing DOX concentrations [98] . While distribution
DCBs throughout the outer layer was uniform at higher
DCB (Biocompatibles Ltd, Farnham, UK) is a poly- concentrations in this study, the distribution and
vinyl alcohol (PVA)-based hydrogel bead system into concentration of DOX in the black-core region could
which DOX (DCBDOX) or irinotecan can be loaded. not be determined [98] . It is likely that DOX not only
The beads are available in several size ranges: 70–150 forms ionic bonds with the sulfonate groups, but
μm, 100–300 μm, 300–500 μm, and 500–700 μm also self-aggregates at high concentrations, which
[93,94] . DCB700–900μm and DCB900–1200μm are no lon- quenches the fluorescent signal [98] . Self-aggrega-
ger available, but have been extensively investigated tion is caused by the hydrophobic surface of DOX
and are therefore included in this review. Few stud- and is increased with increasing concentrations of
ies have investigated DCB70–150μm because it has been DOX [101] .
on the market for only a short while. DCB is manu- Ion exchange in combination with aggregate for-
factured by polymerization of methacyloyl-modified mation is thought to form a more hydrophobic envi-
PVA macromers suspended in an oil phase [95,96] . ronment, causing water displacement out from the
The macromers are synthesized by reacting PVA with DCB, and subsequent increased microsphere density
N-acrylamido-aminoacetaldehyde, followed by copo- and shrinkage of the bead size [98,99] . The diameter of
lymerization with 2-acrylamido-2-methylpropane the DCB700–900μm can be reduced by as much as 35%
sulfonate by free radicals [96] . with increasing DOX concentrations, compared with
DCB is biocompatible, nonbiodegradable and the bead size in the standard sodium buffer solution
mechanically flexible. Hence, in the transarterial [95] . DCB size is virtually unchanged (3% decrease)
treatment of HCC, it provides permanent vascu- with increasing concentrations of a saline solution,
lar embolization, limiting the number of doses that suggesting that all negative sulfonate groups are neu-
can be applied to the tumor through the same blood tralized by sodium ions from the standard sodium
vessels. buffer solution, and no self-aggregation of sodium
occurs [68,95,98–99] . The large difference in the reduc-
Loading & distribution of DOX in DCB tion of bead diameter (Na + vs DOX+) is most likely
The loading and release of DOX into and from DCB due to DOX creating a more hydrophobic intra-
has been described as an ion-exchange mechanism. particle environment and thus being more efficient
However, the diffusion of the ions is considered the at replacing water. The displacement of water could
rate-limiting step, as ion exchange is considered to also explain the increased resistance of DCB700–900μm
be instantaneous [68,97–99] . DCB is provided in a to compressive forces after loading DOX [68] .
sodium phosphate buffer of physiological pH, where The time to load DCB with DOX increases
the sodium ions are bound to the sulfonate groups roughly exponentially with the amount of DOX in
within the DCB. Prior to DCB loading with DOX, the solution (Table 3) . This has been explained by the
the excess buffer solution is removed with a needle, increasing hydrophobicity and reduced water content
taking care not to remove any DCB and an aque- within the polymer with increasing DOX concentra-

454 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

A Release medium B Release medium


Unstirred Liquid
water layer film layer

Oil
DC Bead®
phase

Water
phase
D D Deff 1
[ion+] [ion+] [ion+]

[HSO3-]

D Log P D Log P D D Deff


[DOX] [DOX] [DOX] [DOX] [DOX+] [DOX+] [DOX+] 2

Figure 5. Proposed in vitro release mechanisms. (A) Lipiodol® -DOX emulsion and (B) DOX loaded into DC Bead®
(DCBDOX). Release from both Lipiodol-DOX and DCBDOX is controlled by D both within the formulation and
in the unstirred water layer/liquid film layer that forms at the border between the formulation and the release
medium. For Lipiodol-DOX, the partition coefficient between the two phases will affect the release of DOX.
Diffusion within the bulk of DCBDOX is hampered to some degree by DOX–bead interactions, so it is more useful
to describe the bulk transport as the effective diffusion. In addition, the diffusion of counterions is crucial for the
release of DOX from DCBDOX, since ion exchange is needed to trigger its release.
D: Diffusion; Deff: Effective diffusion; DOX: Doxorubicin; LogP: Partition coefficient.

tions in the beads, which reduces the diffusion rate of In conclusion, the time for loading DOX into DCB
DOX through DCB [68,95,100] . The loading time was is dependent on the initial bead size and the concen-
also proportional to the decrease in DCB surface area tration of the DOX loading solution. The maximum
with increasing bead sizes [68,95] . drug load for DCBDOX is determined not only by
For sufficiently long loading times, the maxi- the number of sulfonate binding sites, but also by the
mum drug load for DCB is approximately 40 mg self-aggregation of DOX within the polymer matrix,
of DOX/ml of beads, irrespective of the bead size. which in its turn contributes to the burst release.
This strongly suggests a correlation with the number DOX distributes homogeneously within DCB at low
of binding sites available in DCB [68,95] . It was esti- concentrations. At high concentrations, self-quench-
mated that one single DCB100–300μm contains about ing of DOX interferes with the analysis of DOX in
2.54 × 1014 ionic binding sites, which corresponds the inner region of the DCB and no conclusion on
to 1.40 × 1019 ionic binding sites/ml of DCB100–300μm the distribution of DOX in this region can be drawn.
[98,99] . The maximum loadable amount of DOX, 40 Future studies need to investigate how pH, tempera-
mg per ml DCB, corresponds to 1.31 × 1025 mole- ture and agitation affect distribution, loading time
cules, which is more than the number of free bind- and maximum DOX loading in the extemporaneous
ing sites, however this can be explained by the self- preparation of DCBDOX prior to dosing.
aggregation of DOX [98] . Associated with the excess
loading, an initial burst release (unknown amount) Release of DOX from DCBDOX in vitro
has been reported [68] . The burst release is thought The in vitro release of DOX from DCBDOX is
to be the result of a larger concentration gradient dependent on a number of factors including the
between the DCBDOX and the release medium due bead size, the ionic strength of the release medium,
to a higher concentration of DOX in the surround- the amount of DOX loaded, and the in vitro release
ing thin layer, promoting diffusion. The maximum method. The fraction released after 22 h from smaller
drug load can be reduced by increased ionic strengths beads (DCBDOX100–300μm) was threefold higher than
in the loading solution and by left-over supernatant from larger beads (DCBDOX700–900μm) in one study,
(residuals from the excess buffer solution) in the vial, most likely because of the shorter diffusion path-
which indicates competitive binding of DOX and way within the smaller beads and a larger total-
the counter ion [95] . It is therefore important that surface area [68] . A tenfold increase in ionic strength
only deionized water should be used to prepare the increased the fraction released in vitro over 24 h by
DOX-loading solution. a factor of three, from DCBDOX900–1200μm loaded

future science group www.future-science.com 455


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

Table 3. Time to load DC Bead® with 99% of the loading concentration of doxorubicin in relation to
bead size.
Bead size (μm) Loading concentration of DOX Time to load 99% of loading Ref.
(mg/ml DCB) concentration of DOX (min)
100–300 25.0 20 [68]

300–500 25.0 60 [68]

500–700 5.0 20 [68]

500–700 10.0 45 [68]

500–700 25.0 120 [100]

500–700 25.0 90 [68]

500–700 37.5 360 [68]

500–700 45.0 1440 [68]


DOX: Doxorubicin.

with 25 mg/ml DOX [102] . The influence of differ- T-apparatus for DCBDOX500–700μm loaded with vari-
ent loading doses on the in vitro release of DOX is ous doses (Figure 6A & 6B) [102] . In a sample-and-sepa-
complex and is shown in Figure 6. The maximum rate method, DCB100–300μm released DOX completely
fraction released after 24 h was only about 13% in a within 4 to 24 h, dependent on the loading dose

A 15 B 2.0
6.25 mg/ml 6.25 mg/ml

12.5 mg/ml 12.5 mg/ml


Amount released (mg)
Fraction released (%)

1.5
10 25 mg/ml 25 mg/ml

37.5 mg/ml 37.5 mg/ml


1.0

5
0.5

0 0.0
0 6 12 18 24 0 6 12 18 24
Time (h) Time (h)

C D 15
100 5 mg/ml
10 mg/ml
Amount released (mg)
Fraction released (%)

25 mg/ml
10

50

5 mg/ml 5
10 mg/ml
25 mg/ml
0 0
0 6 12 18 24 0 2 4 6 8
Time (h) Time (h)

Figure 6. The fraction of doxorubicin released and amount released during 24 and 8h after different loading
doses of doxorubicin into DC Bead®. (A & B) Release of doxorubicin from DC Bead doxorubicin500-700μm in phospate
buffered saline in a T-apparatus and (C & D) from DC Bead doxorubicin100–300μm using a phosphate buffered solution
and a sample-and-separate method. Data in (D) were recalculated from (C) assuming that 1 ml doxorubicin loaded
DC Bead was added to the release medium.
Data taken from [99,102] .

456 Therapeutic Delivery (2014) 5(4) future science group


Table 4. Summary of the influence of DC Bead®, loaded or unloaded with doxorubicin, on hepatic tumor tissue in vivo in humans and hepatic
nontumor tissue in humans and pigs.
Species DCB® Loading (mg Admin. Time post- Subjects Concentrations Proportion of all examined
size (μm) DOX/ml beads) dose treatment (n) cases with specified tissue
(days) lesion surrounding the DCB (%) † 

future science group


            DCB Tissue Non-NT NT CN IFT Viable liver Viable tumor
(mg/ml) (μM) ‡,§  (μM) §,¶  (μM) §  parenchyma
Human # 100–300 37.5 98.3 ± 24.4 8 h 1 – 5.00 ± NA NA 0 NA NA NA
mg DOX 3.45
(75–
150 mg)††
Human # 100–300 37.5 98.3 ± 24.4 9–14 3 – 2.1 ± 2.00 ± 2.30 ± 37 51 8 2
mg DOX 1.70 1.45 1.75
(75–
150 mg)††
Human # 100–300 37.5 98.3 ± 24.4 32–36 2 – 0.65 ± 0.50 ± 0.90 ± 40 60 0 0
mg DOX 0.50 0.40 0.55
(75–
150 mg)††
Pig‡‡ 100–300 0.0 2 ml DCB 28 3 – – – – 0 0 100 –
Pig‡‡ 100–300 0.0 2 ml DCB 90 2 – – – – 0 33 67 –
Pig‡‡ 100–300 37.5 103 mg 28 3 22.5 1.05 0.55 ± 1.45 ± 58 41 1 –
DOX§§ 0.80 1.10
Pig‡‡ 100–300 37.5 103 mg 90 2 4 0.75 0.60 ± 2.30 ± 12 81 7 –
DOX§§ 0.90 1.50
Pig‡‡ 700–900 37.5 103 mg 28 3 15 2.05 1.40 ± 4.80 ± 19 81 0 –
DOX§§ 1.05 2.00
Pig‡‡ 700–900 37.5 103 mg 90 2 7.4 1.25 1.25 ± NA 0 100 0 –
DOX§§ 1.05

Results obtained with histology hemateineosin saffron.

Every 20 μm from surface of bead to 600 μm away from bead, independent on tissue lesion.
§
Above or in IC50 range (0.024–6.0 μM; 72 h incubation; 50% cell death) [27–29].

Data taken from [109].
#
Inflammatory fibrotic tissue, viable liver parenchyma and viable tumor.
††
Mean ± standard deviation (minimum–maximum).
‡‡

www.future-science.com
Data taken from [108].
§§
Mean dose for both treated groups combined.
Admin.: Administered; CN: Coagulative necrosis; DCB: DC Bead®; DOX: Doxorubicin; IFT: Inflammatory fibrotic tissue; NA: Not available; NT: Necrotic tissue.
Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems 

457
Review
Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

1000
100

Plasma concentration DOX (ng/ml)

Plasma concentration DOX (ng/ml)


Rabbit 100–300 11.25 mg Human 500–700 25 mg
Pig 100–300 50 mg Human 500–700 75 mg

100 Pig 100–300 127.5 mg Human 500–700 100 mg


Pig 700–900 78.7 mg Human 500–700 128 mg

10

10

1 1
0 2 4 6 0 6 12 18 24
Time (h) Time (h)

Figure 7. Plasma concentration-time curves showing the in vivo doxorubicin release from DC Bead® for different
species. (A) The 6 h plasma DOX concentration–time curves showing release from DC Bead® DOX100–300μm (closed
symbols) and DC Bead® DOX 700–900μm (open symbols) after a range of doses in pigs (circles) and rabbits (triangles).
(B) The plasma DOX concentration-–time curves showing release from DC Bead® DOX500–700μm in humans over 24 h.
The lower limit of quantification was 1 ng/ml for Varela et al. and Poon et al., and was unspecified by Lewis et al.
DOX: Doxorubicin.
Data taken from [12,96,104–105,107] .

(Figure 6C–D) . In both DCB100–300μm and DCB500–700μm ticles. One suggests that the rate-limiting process is
the relative release rate (%/h) was faster with lower diffusion across the film layer (aqueous boundary
concentrations (Figure 6A & 6C) . The absolute release layer), while the other proposes that it is diffusion
rate (mg/h), however, is higher with higher loading within the particle (Figure 5B) [97,99,103] . Biondi et al.
doses in DCB500–700μm, while it is similar for differ- proposed that film diffusion was the rate-limiting step
ent loading doses in DCB100–300μm (Figure 6B & D) . for DCBDOX [99] . However, the change in DCB size,
This indicated that the difference in absolute-release total surface area and volume during the release of
rate (mg/h) might be related to the difference in bead DOX, and the ionic strength of the release medium,
size, assuming that the ranking order of release rate were not considered when these conclusions were
will not be affected by the different release methods. made. Thus, to achieve a mechanistic understanding
This observation could be explained by the greater of this highly dynamic DDS, more work is needed in
shrinkage of the bigger DCBs when loaded with a the development of new in vitro methods and data
higher dose, thus supporting the previously described analysis models.
bead size-release rate relationship [68] . It is also
important to consider that these data were regener- In vivo release of & tissue exposure to DOX
ated using PlotDigitizer (Free Software Foundation from DCBDOX
Inc., MA, USA) and the observations could well have The in vivo performance of DCBDOX has been
been affected by this process. investigated in both nonclinical models and patients
Very different release rates have been reported [12,96,104–109] . A study investigating liver tissue from a
using different in vitro test methods [68,99,102] . Lewis single transplanted HCC patient, resected 8 h after
et al. reported 17% release of DOX in the T-appara- treatment with a transarterial infusion of DCBDOX100–
tus, whereas Biondi et al. reported complete release 300μm
(Table 4) , found that DCB was distributed both
(100%) using a sample-and-separate method [68,99] . within and outside of the tumor, with a range of 13
Both studies were performed with 25 mg/ml loaded mm from the tumor surface [109] . In pig liver and kid-
DCBDOX100–300μm in PBS over 24 h, but with differ- ney, smaller DCBDOX penetrated further into the
ent volumes of DCB and media and different release tissue from the injection site than larger beads [108,110] .
methods. As shown by Jordan et al., altering condi- For instance, DCBDOX700–900μm occluded large ves-
tions such as flow rate and hydrodynamics will affect sels in portal spaces whereas DCBDOX100–300μm were
the release rate [100] . located in the arterial branches closer to the hepatic
There is a need to improve our understanding lobules and sinusoids. Penetration of DCB deep into
of the mechanism(s) behind the DOX release from the tumor tissue is desirable, but the risk of passing
DCBDOX and its rate-limiting steps. Two theoreti- through the tumor tissue increased with decreasing
cal models have been proposed for ion-exchange par- DCB size. So far, the optimal bead size and proper-

458 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

LIPDOX DCBDOX

Blood ECF Hepatocyte ECF Blood

Ion+

DCBDOX

Stasis

Blood Blood
flow flow

LIP LIPDOX w/o emulsion DOX

Figure 8. Proposed mechanisms of in vivo delivery using LIPDOX, (to the left) or DCBDOX, (to the right). LIPDOX
is administered into the liver segment containing the tumor and reaches the tumor feeding vessels via the blood.
In the blood, the LIPDOX is either separated into unloaded LIP and free DOX, after which DOX is transferred into
the extracellular fluid, or delivered through the endothelia into the ECF. In the ECF, LIPDOX releases DOX into the
ECF or into the hepatocytes, or it may be pinocytosed into the hepatocytes where it releases DOX. DCBDOX is
administered adjacent to the tumor via a tumor-feeding vessel. DC Bead® subsequently embolizes the vessel and
slowly releases DOX into the blood in exchange for cations from the blood. The released DOX enters the ECF and
eventually reaches the intracellular spaces in the hepatocytes.
DCBDOX: Doxurubicin loaded into DC Beads® ; DOX: Doxurubicin; ECF: Extracellular fluid; LIPDOX: Aqueous
doxorubicin solution emulsified in Lipiodol® ; w/o: Water in oil.

ties of DCB for tumor-targeted treatment of HCC are It can also be speculated that the local embolization
unknown. retards diffusion of counter ions required for ionic
Plasma exposure to DOX after administration of exchange, thus decreasing the release rate of DOX
various bead sizes and doses of DCBDOX to rabbits, from DCBDOX.
pigs and humans is depicted in Figure 7. Increased Liver concentrations of DOX after DCBDOX
plasma exposure to DOX was directly correlated with administration have been measured in both HCC
higher doses and smaller DCB sizes in vivo, which is patients and healthy pigs (Table 4) . Concentrations
in agreement with the higher in vitro release data for of DOX in DCBDOX decreased over time in pigs; a
these beads (Figures 6 & 7) . The systemic concentra- faster decrease was seen for DCBDOX100–300μm than
tions of DOX after DCBDOX injection (various sizes for DCBDOX700–900μm [108] , which is in good agree-
and doses) were below the limit of quantification, or ment with the higher in vitro release rate seen for this
very low, after 2 and 7 days in pigs and humans, respec- smaller particle size. The tissue concentrations, mea-
tively (Figure 7) [12,104,107] . Plausible explanations for sured up to 0.6 mm from the DCB, tended to decrease
this include a slow release rate from DCBDOX and/ both over distance from DCB and over time for both
or high local tissue uptake and accumulation of DOX. species, irrespective of DCB size (from 8 h to 32–36

future science group www.future-science.com 459


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

Table 5. Summary of the comparison of the Lipiodol® and DC Bead® drug-delivery systems.
Parameters LIPDOX DCBDOX
Active pharmaceutical ingredient DOX DOX
Vehicle LIP DCB
Type of formulation Emulsion, Dispersion of DOX-loaded beads in aqueous
preferably w/o solution
with higher LIP
volume than
aqueous DOX
volume
Biodegradable Yes, but LIP can No
be accumulated
in tumor tissue
for up to 3
months
Loading mechanism Emulsification Ion-exchange mechanism, where DOX binds to
of aqueous DOX anions in the bead
solution in LIP
Common drug load Variable 37.5 mg DOX/ml DCB
Maximum dose (per treatment) 50 mg 150 mg
Release mechanism Diffusion Ion-exchange and diffusion
Release rate Dependent on Dependent on drug load and bead size
type of emulsion
Technique used for Via image- Via image-guided insertion of catheter into hepatic
administration guided insertion artery via femoral vein
of catheter into
hepatic artery via
femoral vein
Site of administration Whole-liver, Tumor-feeding artery†
lobar, segmental
or subsegmental
Embolization after administration Yes, partial and Yes, full and permanent
temporary‡
In vivo distribution Dependent Dependent on site of injection
on tumor size
and tumor
vasculature
LIPDOX is an emulsion containing an aqueous doxorubicin solution and DCBDOX are embolic drug-eluting beads loaded with DOX.

Often described as superselective in clinical research papers.

Embolizing agents, for example a gelatin sponge, can be inserted after administration of the emulsion. These agents cause full permanent
or temporaryembolization.
DCB: DC Bead®; DCBDOX: Doxorubicin loaded into DC Beads; DOX: Doxorubicin; LIP: Lipiodol®; LIPDOX: Aqueous doxorubicin solution
emulsified in Lipiodol; w/o: Water in oil.

days in HCC patients and from 28–90 days in pigs) bly because the DOX tissue concentrations were higher
[108,109] . However, the concentrations remained in or closer to the DCB and consequently generated more
above the cytotoxic range (IC50 0.024–6.0 μM). In necrotic tissue. A further explanation to the observa-
agreement with the faster in vitro release from smaller tion might be that necrotic tissue cannot eliminate
beads, DCBDOX100–300μm was correlated with lower DOX as effectively as nonnecrotic tissue can.
tissue concentrations at both time points (28 and 90 The tissue surrounding DCBDOX was histologi-
days) than DCBDOX700–900μm in healthy pigs [108] . cally classified as necrotic, inflammatory-fibrotic,
Necrotic tissue had significantly higher concentrations healthy liver parenchyma, or healthy tumor (Table 4) .
of DOX than nonnecrotic tissue in both species, possi- It appeared that the necrosis caused by DCBDOX in

460 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

healthy pig liver was reversible, while in HCC liver istered via the same route, and used to treat the same
patients the necrosis remained unaltered [108,109] . indication. Although both can be loaded with DOX, the
Although differences in the measurement time points recommended doses for DCBDOX and LIPDOX are
may have been responsible for this trend, the presence 150 mg and 50 mg, respectively [88,104] . The loading of
of cirrhosis in the human tissue could have had a major DOX into DCB prior to administration is considered to
effect on the difference in outcome [111] . In pig liver, be more standardized and reproducible than the prepara-
unloaded DCB100–300μm did not cause necrosis, but the tion of LIPDOX, which can vary greatly between clini-
percentage of inflammatory-fibrotic tissue tended to cal/manufacturing sites [87–91] . Although both the DDSs
increase over time, most likely due to the nondegrad- are administered as a hepatic TACE infusion, the cathe-
able nature of this DDS. This suggests that while embo- ters are placed with different tissue and tumor specificity.
lization in itself does not cause cell death, it does acti- It is recommended that DCBDOX should be adminis-
vate an immune response [112,113] . DCBDOX100–300μm tered via the tumor-feeding artery, whereas LIPDOX can
tended to cause more necrosis in healthy pig liver than also be administered to (sub)segmental or lobular areas,
DCBDOX700–900μm [108] , which is again in agreement or to the whole liver (Figure 8). The liver distribution of
with the faster release of DOX from smaller beads. DCBDOX is dependent on the site of injection, and the
However, the smaller beads were still surrounded by size of both beads and microvessels. As these nonbiode-
some viable liver parenchyma, while the larger beads gradable particles accumulate, the DCBDOX system
had no viable liver parenchyma in their vicinity. In the provides complete and permanent embolization. Because
clinical group, the amount of viable liver parenchyma instantaneous and irreversible embolization occurs, the
and viable tumor tissue seemed to decrease over time, vessels used to administer DCBDOX can only be used
suggesting a long-term effect of DCBDOX [109] . once,although the tumor can be treated repeatedly by
Up to three months after treatment, DCBDOX using other vessels [9,10] .
was associated with local tissue damage and necro- In contrast, repeated administration of LIPDOX
sis in both healthy and tumor tissue [108,109] . Smaller through the same vessel is possible, as LIPDOX causes
DCBDOX have higher plasma exposure and lower tis- only temporary embolization [88] . LIPDOX distribu-
sue exposure than larger DCBDOX, which correlates tion is affected by the tumor and droplet size as well as
with the faster release rate of DOX. There is, however, the vasculature in and around the tumor. These factors
a need for a better description of the distribution of will influence both the local and systemic concentra-
DOX in the vicinity of the particles and of DCB in tion–time profiles of DOX and its active metabolite(s).
relation to the effects in vivo, and for a definition of Both DDSs deliver DOX to the proximity of the
the optimal properties of the particles [107,109–110] . tumor, but penetration of DOX into the tumor is still
diffusion dependent [82,109] .
Comparison of the formulations The in vivo release profiles for DOX from both LIP-
The pharmaceutical differences between these DDSs (the DOX and DCBDOX have been compared in patients
liquid LIP and the solid DCB) are numerous (Table 5). with HCC [12] . A higher plasma exposure (AUC) was
However, they can both be loaded with DOX, admin- observed with LIPDOX (composition unknown)

Table 6. Summary of the factors affecting the release rate of doxorubicin from drug-delivery
systems comprising an aqueous solution of doxorubicin emulsified with Lipiodol® and doxorubicin
loaded into drug-eluting beads.
Parameters LIPDOX DCBDOX
  In vitro  In vivo  In vitro  In vivo 
Ions Unknown Unknown Y Y
Flow rate Y Y Y Unknown
Membrane used in vitro Y NA Unknown NA
Size of bead NA NA Y Y
Drug load Unknown Unknown Y Y
Type of emulsion Y Y NA NA
Droplet size Unknown Unknown NA NA
Stability of emulsion Y Y NA NA
DCBDOX: Doxorubicin loaded into DC Beads ; DOX: Doxorubicin; LIPDOX: Lipiodol ; NA: Not applicable; Y: Yes.
® ®

future science group www.future-science.com 461


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

than with DCBDOX500-700μm (drug load unknown), of the cellular targets, improvements to catheter tech-
although the doses administered were 50–75 mg and nology, and increased demands from the population.
<150 mg, respectively. A direct comparison of the Although the local DDSs discussed in this paper may
formulations was also performed in healthy pigs and have improved the palliative treatment of intermediate-
in a VX2 rabbit model. The higher systemic plasma stage HCC, this article shows that there is a need for sig-
and hepatic intracellular exposure seen with LIPDOX nificant improvements to release and robustness aspects
compared with DCBDOX suggested faster release of the DDSs, with subsequent improvements to anti-
from LIPDOX [96,105] . The factors affecting the in vitro cancer effects, side effects and associated morbidity. For
and in vivo release of DOX from both DDSs have been instance, there is an ongoing debate regarding the clini-
summarized in Table 6. cal efficacy of, and benefits from, transarterial intrahe-
This review has demonstrated that LIPDOX patic delivery of these formulations [14,114–116] . It seems
releases DOX faster than DCBDOX in vivo, resulting likely that, in the foreseeable future, more targeted
in higher local and systemic exposure to both DOX DDSs will be developed in multidisciplinary projects.
and its active metabolite(s). The local liver tissue and More specific targeting of malignant cells using multi-
tumor tissue concentration–time profiles of DOX have functional nanoparticles could well provide a strategy to
not been well characterized for either of the DDSs. better anticancer effects and lower morbidity associated
However, in vivo release from DCBDOX is thought with this treatment approach for HCC [117] .
to be highly dependent on the intrahepatic environ-
ment of freely diffusible cationic ions. Therefore, it is Financial & competing interests disclosure
still unclear which of the formulations better targets Financial support to E Ahnfelt was provided by the Swed-
the cancer cells and provides the most positive clinical ish Research Council (521-2011-3773). The authors have no
benefit:safety ratio. other relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial
Future perspective conflict with the subject matter or materials discussed in the
As cancer is an increasing health problem in an increas- manuscript. This includes employment, consultancies, hono-
ingly elderly population there is a need to develop novel raria, stock ownership or options, expert t-estimony, grants
multifunctional-drug delivery principles to destroy or patents received or pending, or royalties. No writing
cancer cells while retaining a minimal effect on non- assistance was utilized in the production of this manuscript.
malignant cells. The local administration of established
substances as part of innovative modified-release DDSs Open Access
has evolved considerably over the last decade as a con- This work is licensed under the Creative Commons Attribu-
sequence of multidisciplinary joint ventures. Develop- tion-NonCommercial 3.0 Unported License. To view a copy
ment has been largely driven by novel imaging tools of this license, visit http://creativecommons.org/licenses/by-
becoming clinically standard, increased knowledge nc-nd/3.0/

Executive summary
Lipiodol® –doxorubicin emulsion
• The optimal aqueous doxorubicin solutions emulsified in Lipiodol (LIPDOX); emulsion is water-in-oil,
thermodynamically stabilized, and prepared by a homogenization technique that will create sufficiently small
(<20 μm) water droplets in the oil phase that efficiently penetrates into tumor tissue.
• Clinical and nonclinical studies should describe the composition as well as the preparation technique for
LIPDOX formulations, to allow comparisons of clinical study results.
Doxorubicin-compatible beads
• Release rate of doxorubicin (DOX) from DOX loaded into DC Bead® (DCBDOX) is dependent on at least bead
size, loading dose and composition and diffusivity of ions in the bead surroundings.
• The in vitro release characteristics of DOX from both investigated drug-delivery systems (DDSs), depend
strongly on the used in vitro release test method and it is therefore necessary to establish the most suitable
method for these DDSs so as to be able to compare in vitro with in vivo situations.
Comparison of the formulations
• LIPDOX and DCBDOX are both mainly passively delivered to hepatocellular carcinoma tissue rather than
nontumor tissue, but act in very different ways.
• LIPDOX releases DOX faster than DCBDOX in vivo, resulting in higher local and systemic exposure to both DOX
and its active metabolite(s).
• More research comparing the in vitro and in vivo performance of these DDSs, in particular regarding their
tumor and systemic exposure-effect relationship, is needed.

462 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

References
1 Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin delivery systems. J. Pharm. Pharmacol. 65(2), 157–170
DM. Estimates of worldwide burden of cancer in 2008: (2013).
GLOBOCAN 2008. Int. J. Cancer. 127(12), 2893–2917 16 Winiwarter S, Bonham NM, Ax F, Hallberg A, Lennernas
(2010). H, Karlen A. Correlation of human jejunal permeability (in
2 El-Serag HB. Hepatocellular carcinoma. New England J. vivo) of drugs with experimentally and theoretically derived
Med. 365(12), 1118–1127 (2011). parameters. A multivariate data analysis approach. J. Med.
3 European Association for the Study of the Liver, European Chem. 41(25), 4939–4949 (1998).
Organisation for Research Treatment of Cancer. EASL- 17 Moffat AC. Clarke’s Analysis of Drugs and Poisons: In
EORTC clinical practice guidelines: management of Pharmaceuticals, Body Fluids and postMortem Material (3rd).
hepatocellular carcinoma. J. Hepatol. 56(4), 908–943 Pharmaceutical Press, London, UK. (2004).
(2012). 18 Cardone RA, Casavola V, Reshkin SJ. The role of disturbed
4 Carr BI. Hepatocellular carcinoma: Diagnosis and treatment. pH dynamics and the Na+/H+ exchanger in metastasis.
Carr BI (Ed.). Humana Press, Philidelphia, PA, USA Nat. Rev. Cancer 5(10), 786–795 (2005).
(2010). 19 Formariz TP, Sarmento VH, Silva-Junior AA, Scarpa MV,
5 Tam KY, Leung KC, Wang YX. Chemoembolization agents Santilli CV, Oliveira AG. Doxorubicin biocompatible O/W
for cancer treatment. Eur. J Pharm. Sci. 44(1–2), 1–10 microemulsion stabilized by mixed surfactant containing
(2011). soya phosphatidylcholine. Colloids Surf. B, Biointerfaces
6 Iwai K, Maeda H, Konno T. Use of oily contrast medium 51(1), 54–61 (2006).
for selective drug targeting to tumor: enhanced therapeutic 20 Rivory LP, Avent KM, Pond SM. Effects of lipophilicity
effect and x-ray image. Cancer Res. 44(5), 2115–2121 and protein binding on the hepatocellular uptake and
(1984). hepatic disposition of two anthracyclines, doxorubicin and
7 Park C, Choi SI, Kim H, Yoo HS, Lee YB. Distribution of iododoxorubicin. Cancer Chemother. Pharmacol. 38(5),
Lipiodol in hepatocellular carcinoma. Liver 10(2), 72–78 439–445 (1996).
(1990). 21 Benet LZ, Broccatelli F, Oprea TI. BDDCS applied to over
8 De Baere T, Denys A, Briquet R, Chevallier P, Dufaux J, 900 drugs. AAPS J. 13(4), 519–547 (2011).
Roche A. Modification of arterial and portal hemodynamics 22 US National Library Of Medicine. TOXNET Hazardous
after injection of iodized oils and different emulsions of Substances Data Bank. http://toxnet.nlm.nih.gov/cgi-bin/
iodized oils in the hepatic artery: an experimental study. J. sis/htmlgen?HSDB
Vasc. Interv. Radiol. 9(2), 305–310 (1998). 23 The complete drug reference. www.medicinescomplete.com/
9 Lewis AL, Holden RR. DC Bead embolic drug-eluting 24 Levasseur LM, Slocum HK, Rustum YM, Greco WR.
bead: clinical application in the locoregional treatment of Modeling of the time-dependency of in vitro drug
tumours. Expert Opin. Drug Deliv. 8(2), 153–169 (2011). cytotoxicity and resistance. Cancer Res. 58(24), 5749–5761
10 Malagari K, Chatzimichael K, Alexopoulou E et al. (1998)
Transarterial chemoembolization of unresectable 25 Lee TK, Lau TC, Ng IO. Doxorubicin-induced apoptosis
hepatocellular carcinoma with drug eluting beads: results and chemosensitivity in hepatoma cell lines. Cancer
of an open-label study of 62 patients. Cardiovasc. Interv. Chemother. Pharmacol. 49(1), 78–86 (2002).
Radiol. 31(2), 269–280 (2008).
26 Hu QD, Chen W, Yan TL et al. NSC 74859 enhances
11 Lammer J, Malagari K, Vogl T et al. Prospective doxorubicin cytotoxicity via inhibition of epithelial-
randomized study of doxorubicin-eluting-bead embolization mesenchymal transition in hepatocellular carcinoma cells.
in the treatment of hepatocellular carcinoma: results of the Cancer Lett. 325(2), 207–213 (2012).
PRECISION V study. Cardiovasc. Interv. Radiol. 33(1),
27 Chang AY, Wang M. In vitro growth inhibition
41–52 (2010).
of chemotherapy and molecular targeted agents in
12 Varela M, Real MI, Burrel M et al. Chemoembolization of hepatocellular carcinoma. Anti Cancer Drugs 24(3), 251–259
hepatocellular carcinoma with drug eluting beads: efficacy (2013).
and doxorubicin pharmacokinetics. J. Hepatol. 46(3),
28 Kim JH, Chung JB, Park IS et al. Combined use of
474–481 (2007).
tamoxifen, cyclosporin A, and verapamil for modulating
13 Guiu B, Deschamps F, Aho S et al. Liver/biliary injuries multidrug resistance in human hepatocellular carcinoma
following chemoembolisation of endocrine tumours and cell lines. Yonsei Med. J. 34(1), 35–44 (1993).
hepatocellular carcinoma: Lipiodol versus drug-eluting
29 Yuan S, Zhang X, Lu L, Xu C, Yang W, Ding J. Anticancer
beads. J. Hepatol. 56(3), 609–617 (2012).
activity of methoxymorpholinyl doxorubicin (PNU 152243)
14 Huang K, Zhou Q, Wang R, Cheng D, Ma Y. on human hepatocellular carcinoma. Anti Cancer Drugs
Doxorubicin-eluting bead versus conventional transarterial 15(6), 641–646 (2004).
chemoembolization for the treatment of HCC: a meta-
30 Jain D. Cardiotoxicity of doxorubicin and other
analysis. J. Gastroenterol. Hepatol. (2013).
anthracycline derivatives. J. Nucl. Cardiol. 7(1), 53–62
15 Tacar O, Sriamornsak P, Dass CR. Doxorubicin: an update (2000).
on anticancer molecular action, toxicity and novel drug

future science group www.future-science.com 463


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

31 Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. 47 Wolfsohn JM, Morrissey EJ. On the value of lipiodol as an
Anthracyclines: molecular advances and pharmacologic aid in neurologic localization: a report based on six cases.
developments in antitumor activity and cardiotoxicity. West J. Med. 26(1), 55–59 (1927).
Pharmacol. Rev. 56(2), 185–229 (2004). 48 Vazquez BY. Blood pressure and blood viscosity are not
32 Kim JE, Cho HJ, Kim JS et al. The limited intestinal correlated in normal healthy subjects. Vasc. Health Risk
absorption via paracellular pathway is responsible for the Manag. 8, 1–6 (2012).
low oral bioavailability of doxorubicin. Xenobiotica 43(7), 49 Guerbert. Lipiodol®. http://www.guerbet.hk/index.
579–591 (2013). php?id=4465
33 Neuhoff S, Artursson P, Zamora I, Ungell AL. Impact of 50 Kan Z, Sato M, Ivancev K et al. Distribution and effect
extracellular protein binding on passive and active drug of iodized poppyseed oil in the liver after hepatic artery
transport across Caco-2 cells. Pharm. Res. 23(2), 350–359 embolization: experimental study in several animal species.
(2006). Radiology 186(3), 861–866 (1993)
34 Sugano K, Kansy M, Artursson P et al. Coexistence of 51 Kan Z, Wallace S. Sinusoidal embolization: impact of
passive and carrier-mediated processes in drug transport. iodized oil on hepatic microcirculation. J. Vasc. Inter.
Nat. Rev. Drug Discov. 9(8), 597–614 (2010). Radiol. 5(6), 881–886 (1994).
35 Okabe M, Unno M, Harigae H et al. Characterization of 52 Kan Z, Wright K, Wallace S. Ethiodized oil emulsions in
the organic cation transporter SLC22A16: a doxorubicin hepatic microcirculation: in vivo microscopy in animal
importer. Biochem. Biophys. Res. Comm. 333(3), 754–762 models. Academic Radiol. 4(4), 275–282 (1997).
(2005).
53 Yang L, Zhang XM, Zhou XP et al. Correlation between
36 Thorn CF, Oshiro C, Marsh S et al. Doxorubicin pathways: tumor perfusion and lipiodol deposition in hepatocellular
pharmacodynamics and adverse effects. Pharmacogenet. carcinoma after transarterial chemoembolization. J. Vasc.
Genomics 21(7), 440–446 (2011). Inter. Radiol. 21(12), 1841–1846 (2010).
37 Robert J, Bui NB, Vrignaud P. Pharmacokinetics of 54 Bhattacharya S, Novell JR, Winslet MC, Hobbs KE.
doxorubicin in sarcoma patients. Eur. J. Clinical Pharmacol. Iodized oil in the treatment of hepatocellular carcinoma.
31(6), 695–699 (1987). Br. J. Surgery 81(11), 1563–1571 (1994).
38 Mross K, Maessen P, Van Der Vijgh WJ, Gall H, Boven 55 Kim H, Park C, Lee YB, Yoo HS, Park JH, Lee WY. Effects
E, Pinedo HM. Pharmacokinetics and metabolism of of iodinated fatty acid ester on human hepatocellular
epidoxorubicin and doxorubicin in humans. J. Clinical Oncol. carcinoma cells. Yonsei Medical J. 32(1), 16–23 (1991).
6(3), 517–526 (1988).
56 Chou FI, Fang KC, Chung C et al. Lipiodol uptake and
39 Patel KJ, Tredan O, Tannock IF. Distribution of the retention by human hepatoma cells. Nucl. Med. Biol. 22(3),
anticancer drugs doxorubicin, mitoxantrone and topotecan 379–386 (1995).
in tumors and normal tissues. Cancer Chemother. Pharmacol.
57 Al-Mufti R A, Pedley RB, Marshall D et al. In vitro
72(1), 127–138 (2013).
assessment of Lipiodol-targeted radiotherapy for liver
40 Speth Pa, Van Hoesel QG, Haanen C. Clinical and colorectal cancer cell lines. Br. J. Cancer 79(11–12),
pharmacokinetics of doxorubicin. Clinical Pharmacokinet. 1665–1671 (1999).
15(1), 15–31 (1988).
58 Towu E, Al-Mufti R, Winslet M. Uptake of lipiodol-
41 Kassner N, Huse K, Martin HJ et al. Carbonyl reductase 1 cytotoxics conjugates by hepatocellular carcinoma cells. J.
is a predominant doxorubicin reductase in the human liver. Pediatr. Surg. 39(2), 203–206; discussion 203–206 (2004).
Drug Metab. Dispos. 36(10), 2113–2120 (2008).
59 Maeda H, Sawa T, Konno T. Mechanism of tumor-targeted
42 Danesi R, Fogli S, Gennari A, Conte P, Del Tacca M. delivery of macromolecular drugs, including the EPR
Pharmacokinetic-pharmacodynamic relationships of the effect in solid tumor and clinical overview of the prototype
anthracycline anticancer drugs. Clin. Pharmacokinet. 41(6), polymeric drug SMANCS. J. Control. Release 74(1–3),
431–444 (2002). 47–61 (2001).
43 Benjamin RS, Wiernik PH, Bachur NR. Adriamycin 60 Jiang J, Nilsson-Ehle P, Xu N. Influence of liver cancer on
chemotherapy – efficacy, safety, and pharmacologic basis of lipid and lipoprotein metabolism. Lipids Health Dis. 5, 4
an intermittent single high-dosage schedule. Cancer 33(1), (2006).
19–27 (1974).
61 Hippalgaonkar K, Majumdar S, Kansara V. Injectable lipid
44 Untoro J, Schultink W, West CE, Gross R, Hautvast JG. emulsions-advancements, opportunities and challenges.
Efficacy of oral iodized peanut oil is greater than that of AAPS PharmSciTech 11(4), 1526–1540 (2010).
iodized poppy seed oil among Indonesian schoolchildren.
62 Liu K, He X, Lei XZ et al. Pathomorphological study on
Am. J. Clin. Nutr. 84(5), 1208–1214 (2006).
location and distribution of Kupffer cells in hepatocellular
45 Bhattacharya S, Dhillon AP, Winslet MC et al. Human liver carcinoma. World J. Gastroenterol. 9(9), 1946–1949
cancer cells and endothelial cells incorporate iodised oil. Br. (2003).
J. Cancer 73(7), 877–881 (1996).
63 Yamashita YT, Fukushima S, Nishida M, Nakano M.
46 Yumoto Y, Jinno K, Tokuyama K et al. Hepatocellular Experimental study of hepatic artery administration of
carcinoma detected by iodized oil. Radiology 154(1), 19–24 various formulations of lipiodol with anticancer agents.
(1985). Kumamoto Med. J. 40(2), 53–65 (1987).

464 Therapeutic Delivery (2014) 5(4) future science group


Treatment of intermediate stage hepatocellular carcinoma: a review of intrahepatic doxorubicin drug-delivery systems  Review

64 Bjerregaard S, Soderberg I, Vermehren C, Frokjaer S. 80 Gaba RC, Baumgarten S, Omene BO et al. Ethiodized oil
Formulation and evaluation of release and swelling uptake does not predict doxorubicin drug delivery after
mechanism of a water-in-oil emulsion using factorial design. chemoembolization in VX2 liver tumors. J. Vascular Int.
Int. J. Pharm. 193(1), 1–11 (1999). Radiol. 23(2), 265–273 (2012).
65 Meng F, Xu W. Drug permeability prediction using PMF 81 Baumgarten S, Gaba RC, Van Breemen RB. Confirmation
method. J. Mol. Model. 19(3), 991–997 (2013). of drug delivery after liver chemoembolization: direct tissue
66 Lin SY, Wu WH, Lui WY. In vitro release, pharmacokinetic doxorubicin measurement by UHPLC-MS-MS. Biomed.
and tissue distribution studies of doxorubicin hydrochloride Chromatogr. 26(12), 1529–1533 (2012).
(Adriamycin HCl) encapsulated in lipiodolized w/o 82 Liang B, Xiong F, Wu H et al. Effect of transcatheter
emulsions and w/o/w multiple emulsions. Die Pharmazie intraarterial therapies on the distribution of Doxorubicin
47(6), 439–443 (1992). in liver cancer in a rabbit model. PloS ONE 8(10), e76388
67 Yi SW, Kim YH, Kwon IC et al. Stable lipiodolized emulsions (2013).
for hepatoma targeting and treatment by transcatheter arterial 83 Mabed M, Esmaeel M, El-Khodary T, Awad M, Amer
chemoembolization. J. Control. Release 50(1–3), 135–143 T. A randomized controlled trial of transcatheter arterial
(1998). chemoembolization with lipiodol, doxorubicin and cisplatin
68 Lewis AL, Gonzalez MV, Lloyd AW et al. DC bead: in vitro versus intravenous doxorubicin for patients with unresectable
characterization of a drug-delivery device for transarterial hepatocellular carcinoma. Eur J. Cancer Care (Engl.) 18(5),
chemoembolization. J. Vasc. Interv. Radiol. 17(2 Pt 1), 492–499 (2009).
335–342 (2006). 84 Sacco R, Bargellini I, Bertini M et al. Conventional versus
69 Nakamura H, Hashimoto T, Oi H, Sawada S. Transcatheter doxorubicin-eluting bead transarterial chemoembolization
oily chemoembolization of hepatocellular carcinoma. for hepatocellular carcinoma. J. Vasc. Interv. Radiol. 22(11),
Radiology 170(3 Pt 1), 783–786 (1989). 1545–1552 (2011).

70 Aungst BJ, Nguyen NH, Rogers NJ et al. Amphiphilic 85 Song MJ, Park CH, Kim JD et al. Drug-eluting bead
vehicles improve the oral bioavailability of a poorly soluble loaded with doxorubicin versus conventional Lipiodol-
HIV protease inhibitor at high doses. Int. J. Pharm. 156(1), based transarterial chemoembolization in the treatment
79–88 (1997). of hepatocellular carcinoma: a case-control study of Asian
patients. Eur. J. Gastroenterol. Hepatol. 23(6), 521–527
71 Benichou A, Aserin A, Garti N. Double emulsions stabilized
(2011).
with hybrids of natural polymers for entrapment and slow
release of active matters. Adv. Colloid Interface Sci. 108–109, 86 Petruzzi NJ, Frangos AJ, Fenkel JM et al. Single-center
29–41 (2004). comparison of three chemoembolization regimens for
hepatocellular carcinoma. J. Vasc. Interv. Radiol. (2012).
72 Abismail B, Canselier JP, Wilhelm AM, Delmas H, Gourdon
C. Emulsification by ultrasound: drop size distribution and 87 Dhanasekaran R, Kooby DA, Staley CA, Kauh JS, Khanna
stability. Ultrasonics Sonochem. 6(1–2), 75–83 (1999). V, Kim HS. Comparison of conventional transarterial
chemoembolization (TACE) and chemoembolization with
73 Larsen C, Larsen SW, Jensen H, Yaghmur A, Ostergaard J.
doxorubicin drug eluting beads (DEB) for unresectable
Role of in vitro release models in formulation development
hepatocelluar carcinoma (HCC). J. Surg. Oncol. 101(6),
and quality control of parenteral depots. Expert Opin. Drug
476–480 (2010).
Deliv. 6(12), 1283–1295 (2009).
88 Noren A, Urdzik J, Duraj F, Barbier CE, Karlson BM,
74 Constantinides PP, Wasan KM. Lipid formulation strategies
Haglund U. Longterm follow-up after transarterial
for enhancing intestinal transport and absorption of
chemotherapy for hepatocellular carcinoma in a Scandinavian
P-glycoprotein (P-gp) substrate drugs: in vitro/in vivo case
centre. HPB (Oxford) 12(9), 637–643 (2010).
studies. J. Pharm. Sci. 96(2), 235–248 (2007).
89 Vogl TJ, Lammer J, Lencioni R et al. Liver, gastrointestinal,
75 Kong X, Ge H, Chen L et al. Gamma-linolenic acid
and cardiac toxicity in intermediate hepatocellular carcinoma
modulates the response of multidrug-resistant K562 leukemic
treated with PRECISION TACE with drug-eluting beads:
cells to anticancer drugs. Toxicol. In Vitro 23(4), 634–639
results from the PRECISION V randomized trial. Am. J.
(2009).
Roentgenol. 197(4), W562–W570 (2011).
76 Das UN, Madhavi N, Sravan Kumar G, Padma M,
90 Van Malenstein H, Maleux G, Vandecaveye V et al.
Sangeetha P. Can tumour cell drug resistance be reversed
A randomized Phase II study of drug-eluting beads
by essential fatty acids and their metabolites? Prostaglandins
versus transarterial chemoembolization for unresectable
Leukot. Essent. Fatty Acids 58(1), 39–54 (1998).
hepatocellular carcinoma. Onkologie 34(7), 368–376 (2011).
77 Kwon IK, Lee SC, Han B, Park K. Analysis on the current
91 Boulin M, Ciboulet A, Guiu B et al. Randomised controlled
status of targeted drug delivery to tumors. J. Control. Release
trial of lipiodol transarterial chemoembolisation with
164(2), 108–114 (2012).
or without amiodarone for unresectable hepatocellular
78 Konno T. Targeting cancer chemotherapeutic agents by use of carcinoma. Dig. Liver Dis. 43(11), 905–911 (2011).
lipiodol contrast medium. Cancer 66(9), 1897–1903 (1990).
92 Johnson PJ, Kalayci C, Dobbs N et al. Pharmacokinetics
79 Furuta T, Kanematsu T, Kakizoe S, Sugimachi K. Selective and toxicity of intraarterial adriamycin for hepatocellular
effect of doxorubicin suspended in lipiodol on VX2 carcinoma: effect of coadministration of lipiodol. J. Hepatol.
carcinoma in rabbits. J. Surg. Oncol. 39(4), 229–234 (1988). 13(1), 120–127 (1991).

future science group www.future-science.com 465


Review  Dubbelboer, Lilienberg, Ahnfelt, Sjögren, Axén & Lennernäs

93 Biocompatibles UK Ltd. Biocompatibles products. http:// following intrahepatic delivery of different dosage forms.
www.biocompatibles.com/products Mol. Pharm. 11(1), 131–144 (2014).
94 Terumo Europe NV embolotherapy (DC Bead® and DC 106 Verret V, Namur J, Ghegediban SH et al. Toxicity of
Bead® M1). www.terumo-europe.com/endovascular/ doxorubicin on pig liver after chemoembolization with
embolotherapy.php doxorubicin-loaded microspheres: a pilot DNA-microarrays
95 Lewis AL, Gonzalez MV, Leppard SW et al. Doxorubicin and histology study. Cardiovasc. Interv. Radiol. 36(1),
eluting beads – 1: effects of drug loading on bead 204–212 (2013).
characteristics and drug distribution. J. Mater. Sci. 18(9), 107 Lewis AL, Taylor RR, Hall B, Gonzalez MV, Willis
1691–1699 (2007) SL, Stratford PW. Pharmacokinetic and safety study of
96 Hong K, Khwaja A, Liapi E, Torbenson MS, Georgiades doxorubicin-eluting beads in a porcine model of hepatic
CS, Geschwind JF. New intra-arterial drug delivery system arterial embolization. J. Vasc. Interv. Radiol. 17(8),
for the treatment of liver cancer: preclinical assessment 1335–1343 (2006).
in a rabbit model of liver cancer. Clin. Cancer Res. 12(8), 108 Namur J, Wassef M, Millot JM, Lewis AL, Manfait M,
2563–2567 (2006). Laurent A. Drug-eluting beads for liver embolization:
97 Boyd GE, Schubert J, Adamson AW. The exchange concentration of doxorubicin in tissue and in beads in a pig
adsorption of ions from aqueous solutions by organic model. J. Vasc. Interv. Radiol. 21(2), 259–267 (2010).
zeolites; ion-exchange equilibria. J. Am. Chem. Soc. 69(11), 109 Namur J, Citron SJ, Sellers MT et al. Embolization
2818–2829 (1947). of hepatocellular carcinoma with drug-eluting beads:
98 Biondi M, Fusco S, Lewis AL, Netti PA. New insights into doxorubicin tissue concentration and distribution in patient
the mechanisms of the interactions between doxorubicin and liver explants. J. Hepatol. 55(6), 1332–1338 (2011).
the ion-exchange hydrogel DC Bead for use in transarterial 110 Dreher MR, Sharma KV, Woods DL et al. Radiopaque drug-
chemoembolization (TACE). J. Biomater. Sci. Polym. Ed. eluting beads for transcatheter embolotherapy: experimental
23(1–4), 333–354 (2012). study of drug penetration and coverage in swine. J. Vasc.
99 Biondi M, Fusco S, Lewis AL, Netti PA. Investigation of Interv. Radiol. (2011).
the mechanisms governing doxorubicin and irinotecan 111 Carr BI, Nagalla S. Medical Therapy of HCC. In:
release from drug-eluting beads: mathematical modeling and Hepatocellular Carcinoma: Diagnosis and Treatment. Carr BI
experimental verification. J. Mater. Sci. 24(10), 2359–23570. (Ed.). Humana Press, New York, USA, 530–568 (2010).
(2013). 112 Vinchon-Petit S, Jarnet D, Michalak S, Lewis A, Benoit JP,
100 Jordan O, Denys A, De Baere T, Boulens N, Doelker E. Menei P. Local implantation of doxorubicin drug eluting
Comparative study of chemoembolization loadable beads: beads in rat glioma. Int. J. Pharm. 402(1–2), 184–189
in vitro drug release and physical properties of DC Bead and (2010).
hepasphere loaded with doxorubicin and irinotecan. J. Vasc. 113 Stampfl S, Stampfl U, Bellemann N et al.
Interv. Radiol. 21(7), 1084–1090 (2010). Immunohistochemical characterization of specific
101 Li X, Hirsh DJ, Cabral-Lilly D et al. Doxorubicin physical inflammatory tissue reactions following embolization with
state in solution and inside liposomes loaded via a pH four different spherical agents in the minipig kidney model.
gradient. Biochim. Biophys. Acta 1415(1), 23–40 (1998). J. Vasc. Interv. Radiol. 20(7), 936–945 (2009).
102 Gonzalez MV, Tang Y, Phillips GJ et al. Doxorubicin 114 Oliveri RS, Wetterslev J, Gluud C. Transarterial (chemo)
eluting beads-2: methods for evaluating drug elution and embolisation for unresectable hepatocellular carcinoma.
in-vitro:in-vivo correlation. J. Mater. Sci. 19(2), 767–775 Cochrane Database Syst. Rev. (3), CD004787 (2011).
(2008). 115 Ray CE Jr, Haskal ZJ, Geschwind JF, Funaki BS. The use
103 Jeong SH, Berhane NH, Haghighi K, Park K. Drug release of transarterial chemoembolization in the treatment of
properties of polymer coated ion-exchange resin complexes: unresectable hepatocellular carcinoma: a response to the
experimental and theoretical evaluation. J. Pharm. Sci. Cochrane Collaboration review of 2011. J. Vasc. Interv.
96(3), 618–632 (2007). Radiol. 22(12), 1693–1696 (2011).
104 Poon RT, Tso WK, Pang RW et al. A Phase I/II trial of 116 Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma.
chemoembolization for hepatocellular carcinoma using a Lancet 379(9822), 1245–1255 (2012).
novel intra-arterial drug-eluting bead. Clin. Gastroenterol. 117 Shi J, Votruba AR, Farokhzad OC, Langer R.
Hepatol. 5(9), 1100–1108 (2007). Nanotechnology in drug delivery and tissue engineering:
105 Lilienberg E, Ebeling Barbier C, Nyman R et al. from discovery to applications. Nano Lett. 10(9), 3223–3230
Investigation of hepatobiliary disposition of Doxorubicin (2010).

466 Therapeutic Delivery (2014) 5(4) future science group

You might also like