You are on page 1of 7

Methods 55 (2011) 246–252

Contents lists available at ScienceDirect

Methods
journal homepage: www.elsevier.com/locate/ymeth

Iodine-131-Lipiodol therapy in hepatic tumours


Hojjat Ahmadzadehfar a,⇑, Amir Sabet a, Kai Wilhelm b, Hans Jürgen Biersack a, Jörn Risse c
a
Department of Nuclear Medicine, University Hospital Bonn, Germany
b
Department of Radiology, University Hospital Bonn, Germany
c
Radiology and Nuclear Medicine Institute, Bad Honnef, Germany

a r t i c l e i n f o a b s t r a c t

Article history: The incidence of hepatocellular carcinoma (HCC) is worldwide sharply on the rise and patients with
Available online 12 June 2011 advanced disease carry a poor prognosis. HCC is the sixth most common cancer and the third leading
cause of cancer associated deaths in the world. Intra-arterially administered 131I-Lipiodol is selectively
Keywords: retained by hepatocellular carcinomas, and has been used as a vehicle for delivery of therapeutic agents
Iodine-131 Lipiodol to these tumours. In this review we focus on the therapeutic indications, usefulness and methods of treat-
Liver cancer ment with 131-Iodine Lipiodol.
Transarterial interventions
The effectiveness of 131I-Lipiodol treatment is proven both in the treatment of HCC with portal throm-
Portal vein thrombosis
Hepatocellular carcinoma
bosis and also as an adjuvant to surgery after the resection of HCCs. It is at least as effective as chemo-
Cancer therapy embolization and is tolerated much better. Severe liver dysfunction represents theoretic contraindication
for radioembolization as well as for TACE. In such cases 131I-Lipiodol is an alternative therapy option
especially in tumours smaller than 6 cm.
Ó 2011 Elsevier Inc. All rights reserved.

1. Introduction 2. Treatment options of HCC

The incidence of hepatocellular carcinoma (HCC) is worldwide Despite a variety of treatment strategies for HCC patients, the
sharply on the rise and patients with advanced disease carry a poor only available curative options are liver transplantation, partial li-
prognosis. HCC is the sixth most common cancer and the third ver resection and locoregional ablative treatment, being feasible
leading cause of cancer associated deaths in the world [1]. In the only in a small minority of the patients. In general, the choice of
United States, the incidence of HCC is increasing faster than any treatment is dependent not only on the extent of the HCC but also
other malignancy [2,3]. on the severity of underlying hepatic dysfunction [10]. Surgery can
Chronic viral hepatitis B, C and D, alcohol, metabolic liver dis- be associated with substantial difficulty in patients with underly-
eases such as hemochromatosis and a-1-antitrypsin deficiency as ing cirrhosis and peri-operative mortality is as high as 30% to
well as non-alcoholic fatty liver disease may cause chronic liver in- 50% in patients with Child-Pugh class B or C. In the contrary, pa-
jury, regeneration, and cirrhosis [4]. Chronic hepatitis B (CHB) and tients with Child-Pugh class A show a surgical mortality of only
chronic hepatitis C (CHC) are recognised as the major risk factors of 5% to 10% [10,11].
HCC, with risk being greater in the presence of a co-infection [5,6]. For selected patients with HCC, confined to the liver but not
It is now widely accepted that a dysplastic nodule commonly amenable to resection or transplantation, both systemic and loco-
found in cirrhotic livers may slowly degenerate to low-grade HCC regional therapies can be considered. These include
[7,8]. Low-grade (well-differentiated) HCC tends to develop round,
well-defined, encapsulated masses. Over time, focal areas within a 1. Percutaneous interventions (ethanol injection, radiofrequency
low-grade HCC can degrade to poorly differentiated tumours infil- thermal ablation),
trating the liver and invading the venous or arterial network, 2. Transarterial interventions
which causes a more aggressive spread of the disease [9]. (a) Embolization (TAE), chemoperfusion (TAC), or chemoembo-
lization (TACE),
(b) Radioembolization (RE) or radioactive agent perfusion such
as 131Iodine-Lipiodol or 188RE-Lipiodol.
3. External radiation therapy and
4. Chemotherapy, including gene and immune therapy.
⇑ Corresponding author. Address: Department of Nuclear Medicine, University
Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany. Fax: +49 228 287
9019858. Until the recent approval of sorafenib (Nexavar, Bayer Health-
E-mail address: hojjat.ahmadzadehfar@ukb.uni-bonn.de (H. Ahmadzadehfar). Care Pharmaceuticals Inc,Wayne, NJ) by the US Food and Drug

1046-2023/$ - see front matter Ó 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.ymeth.2011.05.003
H. Ahmadzadehfar et al. / Methods 55 (2011) 246–252 247

Administration (FDA) no systemic chemotherapeutics showed sig- 5 cm exhibit a high retention (of type 3 or 4 according to the crite-
nificant increases in survival in patients with HCC [14,15]. This lim- ria of Maki et al. [21]) whereas 74% of the HCCs larger than 10 cm
itation of systemic treatments resulted in the utility of exhibit only a weak retention of less than 50% (of type 1 or 2
interventional radiology techniques in the treatment of HCC. according to the criteria of Maki) [22,23]. Moreover, the clearance
Although nonresectional locoregional therapies are not curative, of Lipiodol from the tumour lasts longer than from the normal liver
they lead to tumour destruction whilst preserving nontumourous or from the lungs, possibly because of a lack of macrophagic cells in
liver parenchyma and may serve as a bridge to a more definitive the tumour [17]. 131I-Lipiodol is eliminated mainly through the uri-
therapy like liver transplantation or as a salvage treatment for nary tract, reaching 30–50% of the injected activity at day 8, but
post-resection recurrence [12,13]. also to a small extent by way of the faeces (<3% at day 5) [18].
In this review we focus only on the therapeutic indications, use- The required therapy activity for the intended tumour dose can
fulness and methods of treatment with 131-Iodine Lipiodol (131I- be calculated for a given tumour mass according to its size. For in-
Lipiodol). Its rationale comes from the anatomical and physiologi- stance producing 120 Gy in a tumour with a diameter of 1 cm re-
cal aspects of HCC being exploited for the delivery of the therapeu- quires less than 3 MBq I-131 whilst the same dose in a tumour
tic agent. The liver has a dual blood supply: the hepatic artery and with a five-fold larger diameter requires hundredfold more activity
the portal vein. Observations on vascular supply to hepatic malig- [24].
nancies have demonstrated that metastatic hepatic tumours The radionuclide is supplied in 2 ml solution for injection and
>3 mm derive in contrast to healthy hepatic cells 80–100% of their stored in a 4 ml cone shaped glass vial. The specific activity at cal-
blood supply from the arterial rather than the portal hepatic circu- ibration date is 1.1 GBq/ml and its shelf life from this time is 3 days
lation [16]. [25,26].
The conjunction of lipiodol with TACE, also called TOCA (tran-
sarterial oily chemoembolization), has also been used allowing
131
3. Basic characteristics of I-Lipiodol the delivery of locally concentrated therapy due to the selective
retention of the agent in the tumours for an extended period [10].
131
I is a beta emitting radionuclide with a physical half life of
8.04 days. The maximum and mean beta particle energies are
0.61 MeV and 0.192 MeV respectively. Additionally, 131I emits a 4. Basics of interventional nuclear medicine therapies of HCC
principal gamma photon of 364 keV (81% abundance). The Beta with I131-Lipiodol
radiation of 131I is responsible for its therapeutic effects whilst
gamma radiation makes the distribution of the radiopharmaceuti- Radiation is tumouricidal if sufficient doses can be delivered
cal visible. selectively to the tumour without damaging adjacent normal tis-
Lipiodol is a mixture of iodised esters of poppy seed oil fatty sue [27]. Normal hepatocytes have a lower tolerance to the effects
acids used as a contrast medium for the detection of HCC which re- of radiation than neoplastic tissue. The dose required to destroy so-
mains in these tumours for a longer period compared to normal li- lid tumours, estimated at P70 Gy, is far greater than the liver tol-
ver or in other tissues. Isotopic exchange is used to label iodine- erance dose of 35 Gy when delivered to the whole liver in 1.8 Gy/d
rich Lipiodol with I131 (LipiocisÒ, CIS Bio International/ member fractions [28]. If the whole liver is exposed to external-beam radi-
of IBA group). LipiocisÒ is licensed in France for the treatment of ation at a mean radiation dose of 43 Gy, more than 50% of patients
patients with HCC and portal vein thrombosis (PVT) [17]. It was develop liver dysfunction [29]. Conformal and stereotactic radia-
developed in the 1990s and was the first radiolabelled vector to tion therapy techniques can be used to deliver much higher radia-
be used in this setting. According to biodistribution data, more tion doses in cases with focal involvement [30]; however, since
than 75% of the 131I-Lipiodol stays following the arterial adminis- primary neoplasms are often multifocal and irregular in shape
tration in the liver and the remainder reaches the lungs [18]. Other and potentially replace large parts of the liver volume, only a small
tissues, including the thyroid gland, receive very little radiation. minority of patients are optimal candidates for such therapies [31].
An in vitro assessment by Al-Mutfi et al. [19] showed an unspe- Internal radiation aims at improving the selectivity of the radiation
cific uptake of Lipiodol in non-malignant endothelial cells apart by targeting the radioisotopes to the tumour cells. To achieve this
from its concentration in cancer cell lines. Lipiodol alone does targeting, the therapeutic radiopharmaceuticals such as 90Y
not appear to have any cytotoxic effect against any of the studied microspheres or 131I-Lipiodol may be administered via the hepatic
cell lines. However, 131I-Lipiodol is highly and only selectively artery [32].
cytotoxic for the cancer cell lines. Inability of tumourous cells un- Based on studies considering biodistribution of intraarterial
131
like non-malignant ones to expel Lipiodol, revealed by quantifica- I-Lipiodol in humans [18,33,34], radiation doses for every mCi
tion methods using computer-assisted image analysis, may further (37 MBq) of administered activity were estimated to be 31 cGy
enhance the cytotoxic effect of 131I-Lipiodol in cancer cells. There is for the normal liver, 22 cGy for the lungs, and 239 cGy for a tumour
no separation of the 131I from Lipiodol during the uptake process with a greatest diameter of 4 cm using the formula:
by the cancer cells, as Lipiodol is a naturally iodinated oil com-
A0 A0
pound with an iodine content of 38–40%. 131I-Lipiodol shows cyto- DoseðcGyÞ ¼ 73:8  E  Te  þ 0:0346  C  g 
toxic effects against the cancer cell lines even in doses as small as a
M M
quarter of the radioactivity of the 131I alone [19]. where E is the average beta energy (MeV), Te is the effective half-life
The tumour/ non tumour uptake ratio is about 15–20:1 and in- (days), G is the specific gamma ray constant (cGy cm2/mCi hr), Ao is
creases with time, whilst the effective half life within the HCC tis- the initial activity (lCi), M is the mass (g), and g is the geometric
sue is also longer than that in healthy tissues (almost 6 days versus factor (cm).
4 days). Generally, more than 10% of the injected Lipiodol remains
within the tumour [20]. The retention of 131I-Lipiodol in HCC tu-
mours after intra-arterial infusion is related not only to emboliza- 5. Absolute contraindications
tion in the micro vessels of the tumour, but also to the entry into
the interstitium and the tumour cells themselves. This retention 1 – Pregnancy and breastfeeding, 2 – life expectancy less than
can be assessed by tomodensitometric examination and inversely 1 month, 3 – hepatic encephalopathy, 4 – tumour Stage OKUDA
relates to the tumour size. Indeed, 88% of the HCCs smaller than III [25,26].
248 H. Ahmadzadehfar et al. / Methods 55 (2011) 246–252

6. Relative contraindications

1 – High extrahepatic tumour burden, 2 – acute or severe


chronic pulmonary disease, 3 – The presence of contraindications
for hepatic artery catheterisation [25,26].

7. Appropriate imaging

The work-up includes a three-phase contrast computed tomog-


raphy (CT) and gadolinium-enhanced magnetic resonance imaging
(MRI) of the liver for the assessment of tumour and non-tumour
volume, portal vein patency, and the extent of extra-hepatic dis-
ease. Although FDG-PET is suitable for tumours showing high glu-
cose metabolism such as colorectal carcinoma, melanoma, head
and neck, and breast cancers, malignancies such as HCC or neuro-
endocrine tumours, except for their aggressive types show no or a
very low-grade FDG uptake[35].
The suboptimal sensitivity of FDG-PET for the detection of HCC,
ranging between 50% and 70% makes it an unsatisfactory imaging
choice for the pre- and post-treatment evaluation in this group of
patients; however, it adds valuable prognostic information (meta-
bolic grading) as patients with a negative FDG-PET show a better
prognosis than those with high FDG uptake. Choline PET/CT may
also play a role in imaging of specific types of HCC in the near fu-
ture [35–38]. Fig. 1. Quantitative whole body scintigraphy obtained 7 days post therapeutic in a
52-year-old man showing significant activity in the HCCs and only a faint lung
uptake.
8. Intervention technique

Treatment with 131I-Lipiodol serves as an example for multidis- radiation exposure to their environment becomes low enough to
ciplinary management including the patient selection and care, the be discharged (e.g. in Germany <1 mSv/year) [40].
detailed anatomical study and dose calculation, the post-interven- Dexamethasone and tamoxifen could increase the tumoural up-
tional treatment including timing and selection of systemic treat- take of Lipiodol and could be combined with 131I-Lipiodol to im-
ment, adequate follow-up of the patient as well as the prove the therapy effectiveness [41].
management of possible complications. Prior to the therapy, pa- Post therapeutic quantitative whole body imaging (Fig. 1) per-
tients should receive both written and verbal information about formed one week later is recommended to confirm the distribution
the procedure and the palliative nature of the treatment [26]. of 131I-Lipiodol [26]. CT of the liver could be used for dosimetry,
The renal functional status should be adequate for the use of con- considering the visibility of Lipiodol in CT as a contrast agent. Now-
trast agents during the pre-therapeutic angiogram. Haemodialysis adays a better quantification of the radioactive distribution and
patients may be treated with 131I-Lipiodol, however, dialysis has to dosimetry could be achieved by performing a hybrid SPECT/CT of
be planned and timed before and after the intervention. Patients the liver (Fig. 2).
should be given a peri-interventional drug regimen with slow For certain patients, in particular those presenting with a rela-
intravenous infusion of 24 mg dexamethasone and 8 mg ondanset- tively large tumour several injections in 2–3 months interval may
rone during the whole procedure and intraarterial injection of be necessary to obtain an effective response.
50 mg pethidine immediately before application of the radiophar- As Garin et al. stated, nuclear medicine specialist receives an
maceutical. After blocking the thyroid with perchlorate, diagnostic average dose between 140 and 443 lSv at the level of the fingers
angiography of the liver vessels should be performed with special and about 17 lSv at the thorax. For the radiologists, the average
attention to collateral arteries to other organs. Occasionally, com- doses received are 215 lSv and 15 lSv at the level of the fingers
puted tomography with arterioportography (CT-AP) via the cathe- and at the thorax respectively. The authors showed that the admin-
ter could be added, partly with the injection of small amounts of istration of high therapeutic activities of 131I-Lipiodol can be car-
‘‘cold’’ Lipiodol (GuerbetÒ). In patients with ‘‘normal’’ hepatic arte- ried out with a dose much lower than the European regulatory
rial anatomy the catheter can be positioned in the main hepatic ar- limit of 500 mSv at the level of the fingers for the exposed person-
tery for nonselective injection of 131I-Lipiodol. In cases with nel [42].
multiple nodules or variable arterial blood supply, the liver lobe
containing the largest tumour mass is treated in the first session
and the remaining masses in the following ones. 9. Follow-up
131
I-Lipiodol (2.22 GBq in 2 ml/ampulla) should be prepared in
an appropriately ventilated cabinet in the department of Nuclear Definitions of the appropriate length of the follow-up and the
Medicine to avoid radioiodine aerosol inhalation. Injecion should time points required for technical success are not well established
take at least some minutes to ensure good tolerance by the patient. and post treatment follow-up schedules vary depending on the
The procedure and special radiation protection measures have treatment plan of each patient. For the evaluation of response,
been readily published in detail [39]. The applied activity should abdominal imaging should be performed 4 weeks and 2–3 months
be determined as the difference between activity in the last recep- after each therapy [26]. Follow-up should be performed with the
tacle before injection and the remaining activity of last receptacle same modality used pre-therapeutic such as MRI, 3 phase CT,
and the catheter-system after flushing with saline. After the proce- 18F-FDG, 11C-/18F-Cholin or 11C-/18F-Acetate PET/CT. AFP should
dure the patients are transferred to the station and isolated until be tested for patients with initially increased AFP.
H. Ahmadzadehfar et al. / Methods 55 (2011) 246–252 249

Fig. 2. SPECT/CT of the liver could be used for a better quantification of the radioactive distribution as well as dosimetry simultaneously. Note the distribution of 131I-Lipiodol
in the HCCs showed with arrows.

10. Dosimetry fairly frequently consist of moderate and temporary fever (29%),
moderate and temporary disturbances of the biological liver test
Becker et al. performed a dosimetric evaluation using SPECT/CT (20%) and hepatic pain on injection (12.5%).
[43], applied to a series of 41 patients who received one or more Moderate and reversible leukopenia (7%) and serious interstitial
injections of 131I-Lipiodol for treatment of HCC using a standard pneumopathies (2%) are observed more rarely [25,26,48]. Intersti-
activity of 2220 MBq. They found a mean tumoural absorbed dose tial pneumonia has been reported in an estimated prevalence of
of 248 ± 176 Gy for the first treatment, compared with 15.5 cases per 1000 treated patients with a median respiratory
152 ± 122 Gy for the second treatment, and pointed out a correla- symptoms delay of 30 days [49].
tion between the tumoural absorbed dose, calculated in tomo- Some authors have expressed their concern about hypothyroid-
graphic mode, and morphological response to first treatment ism despite the very low thyroid uptake [50]. Using potassium io-
alone. These results pleaded in favour of increasing the adminis- dide premedication can significantly decrease thyroid iodide
tered activity at the first treatment. A tumoural absorbed dose of uptake and consequently probability of hypothyroidism [51].
280 Gy was found to represent an effective threshold absorbed
dose. whilst 84% of the patients above this value were responders, 12. Therapeutic indications for 131
I-Lipiodol
no one appeared to respond at lower absorbed doses [43]. Tumour
response adversely correlates with tumour size. High response 12.1. Palliative treatment of inoperable primary hepatocellular
rates have been found in tumour nodes with diameters below carcinoma
6 cm in which an absorbed dose of up to 288 Gy has been calcu-
lated [44–46]. However, it must be considered that a reduction An improved survival rate is readily documented for those trea-
in tumour mass does not necessarily correlate with improved sur- ted with 131I-Lipiodol, comparing to those receiving only medical
vival [20]. support [52]. The treatment is more effective for small, solitary
and well encapsulated tumours and the response rate decreases
with increasing tumour size. Its main limitation is its ineffective-
131
11. Safety of I-Lipiodol therapy ness in large (>5 cm) tumours. However, transarterial therapy with
131
I-Lipiodol is superior to systemic therapy in tumours up to 5 cm
A phase I study [47] and a multi centric phase II trial [46] could in diameter [53].
not demonstrate any specific toxic effects of 131I-Lipiodol. The The data on therapy efficacy have remained non-uniform. A de-
main limiting factor was the long patient isolation required for crease in tumour size has been reported in about 50% to 60% of pa-
the purpose of radioprotection. Consequently, a fixed injected tients; two early studies with seven and nine patients showed
activity of 2.2 GBq is proposed as a good compromise to achieve response in all tumours [44,47]. In larger studies classifying ther-
the desired efficacy whilst reducing the hospital stay to 1 week. apy success according to the WHO criteria, the response rate de-
Serious adverse effects are very rare. Undesirable effects observed creases to about 30% [54]. This is in agreement with our own
250 H. Ahmadzadehfar et al. / Methods 55 (2011) 246–252

data [55]. Reported response rates in different studies are summa- palliative treatment of HCC in the presence of portal vein thrombo-
rised in Table 1; only reports with more than ten patients, data on sis [52]. However it should be noted that radioembolization (RE)
changes in tumour size and follow-up studies of at least one with 90Y-Microspheres has proved to be a feasible alternative
month, are considered. treatment for HCC patients with PVT as discussed in following
131
I-Lipiodol is also comparatively favourable to other regional section.
therapy procedures for palliative treatment of HCC. A multicentre
randomised trial by Raoul et al. [56] showed that 131I-Lipiodol 12.1.2. 131I-Lipiodol versus radioembolization with 90Y-microspheres
therapy (n = 73) was associated with a better patient tolerance 90Y radioembolization (RE), or selective internal radiation ther-
and fewer vascular complications than TACE (n = 69), although apy (SIRT), is a promising catheter-based liver-directed modality
no survival advantage was demonstrated [56]. approved by the Food and Drug Administration for patients with
In a published study from our group 38 courses of intra-arterial primary and metastatic liver cancer [35]. In a meta-analysis of 14
131
I-Lipiodol therapy with a total activity up to 6.7 GBq were per- published articles Venti et al. showed almost 80% any response
formed in 18 patients with HCC (6 with PVT) [40]. Tumour volume (AR = [CR + PR + SD]) for a total of 325 patients with HCC. Accord-
decreased in 20/32 index nodules (63%) after the first course. Re- ing to this meta-analysis, treatment with resin microspheres was
peated therapy frequently resulted in further tumour reduction. associated with a significantly higher proportion of AR than with
Partial response was seen in 11 nodules whilst four nodules glass microsphere (0.89 vs 0.78 [P < 0.02]) [66].
showed a minor response. Stable disease was found in 12 patients Although RE is currently the preferred method for treating HCC
and progressive disease in 5. Significant response was associated compared to 131I-Lipiodol, its embolic nature makes severe liver
with pre-therapeutic nodule volume up to 150 ml (diameter of dysfunction a theoretic contraindication for this technique, as in
6.6 cm). Survival rate after 3, 6, 9, 12, 24 and 36 months was TACE. In addition, techniques using 90Y-labelled products tend to
78%, 61%, 50%, 39%, 17%, and 6% respectively. Matched-pairs anal- cost up to 10 times more than therapy with 131I-Lipiodol [67],
ysis of survival revealed 131I-Lipiodol to be superior to medical affecting the therapeutic approach especially in the countries with
treatment. The most important side effect was a pancreatitis-like a poor insurance system.
syndrome whilst overall tolerance was very acceptable. PVT is not an absolute contraindication for RE any more.
The median survival in patients with unresectable HCC under- Although resin microspheres could pose the patient at risk for sig-
going 131I-Lipiodol therapy has been reported between 7 and nificant liver dysfunction based on the embolic treatment effect,
27 months [61–64]. Chua et al. reported the factors associated with therasphere has been shown to be safe even when the portal vein
survival in a newly published study including the AJCC stage (1–2 has been invaded by tumour [27]. However, recent studies describe
vs 3–4), Barcelona Clinic Liver Cancer stage (A–B vs C), Cancer of safe performance of RE with resin microspheres even in PVT. There
the Liver Italian Programme score (<2 vs >2), maximum tumour is no prospective study which compares these two methods (RE vs
131
size (<4 vs >4 cm), extrahepatic disease, previous surgery and re- I-Lipiodol) in patients with PVT in HCC considering response
sponse to treatment (favourable vs unfavourable) [64]. Considering rate, time-to-progression, and survival.
response rate, AJCC stage 1–2 and Cancer of the Liver Italian Pro-
gramme score <2 were found to be independent predictors for a 12.1.3. 131I-Lipiodol-therapy versus TAC and TACE
131
favourable response in this study [64]. I-Lipiodol therapy in patients with cirrhosis and HCC has an
efficacy similar to that of TACE/TAE therapy [56,59,67]. However
131
12.1.1. Palliative treatment of HCC with Portal vein thrombosis (PVT) I-Lipiodol appears to be of more benefit in patients with ad-
Although the PVT prohibits any attempt of TACE, 131I-Lipiodol vanced disease, for example, in those with Okuda stage III disease
has proved to be an effective therapy option in patients with or BCLC stage D as well as in patients with PVT [67]. PVT patients
HCC and PVT [65]. Raoul et al. compared 131I-Lipiodol therapy receiving 131I-Lipiodol lived on average almost a year longer than
and best supportive care in patients with HCC and PVT. The sur- those treated with TACE or TAE. Moreover, 131I-Lipiodol is much
vival rates at 3, 6, and 9 mo was 71%, 48%, and 7% for the treated better tolerated than chemoembolization, both in terms of clini-
group; and 10%, 0% and 0% for the group receiving only supportive cally expressed side effects and arteriographic findings [56].
care. The authors concluded that 131I-Lipiodol is a safe and effective
12.2. Performing 131I-Lipiodol as an adjuvant or neo-adjuvant therapy

Early intrahepatic recurrence is common after curative resec-


Table 1 tion of hepatocellular carcinoma, frequently due to microscopic
131
Efficacy of I-Lipiodol therapy in patients with unresectable HCC. metastatic disease or less commonly, metachronous multicentric
Reference Number of evaluable CR PR MR SD PD AFP carcinoma within the liver remnant not detected before or during
patients decrease resection by conventional imaging [68] and has led to attempts at
Yoo [57] 24 0 3 5 8 8 13/16 adjuvant or neoadjuvant therapies.
Raoul [46] 30 0 18 4 6 2 22/29
Leung [58] 22 1 3 – 12 6 13/25 12.2.1. 131I-Lipiodol as an adjuvant therapy
Bhattacharya 22 0 7 – 8 7 2/22 It has been thought that when the postsurgical liver starts to
[59]
Raoul [56] 25 1 15 4 3 2 20/40
regenerate, small microscopic daughter tumours can be stimulated
De Baere [54] 23 0 3 – 12 8 – to grow. If these were pre-cleared the chances of a recurrence
Risse [55] 13 0 2 3 4 4 – would be lowered. Thus,131I-Lipiodol has been used in an adjuvant
Rindani [60] 12 0 6 – 5 1 5/7 setting after curative surgical resection [69]. A single dose of
Borbath [61] 19 1 0 – 7 11 2/24
1.85 GBq 131I-Lipiodol administered 6 weeks after curative resec-
Risse [40] 17 0 4 1 7 5 –
Boucher [62] 40 1 18 – 19 2 tion of hepatocellular carcinoma could significantly decrease the
rate of recurrence and increased disease-free and overall survival
CR: complete response, PR: partial response (tumour reduction >50%), MR: minimal
in a study by Lau et al. [68]. In their recent published paper the
response (tumour reduction between 25 and 50%, AFP alpha-fetoprotein, SD: stable
disease (tumour size change between 25% and +25%), PD: progressive disease
same authors confirmed their results after a longer follow up
(tumour increase >25%). [70]. These findings were also confirmed by Boucher et al. admin-
istering 2.4 GBq 131I-Lipiodol 8–12 weeks after surgery [71]. NG
H. Ahmadzadehfar et al. / Methods 55 (2011) 246–252 251

et al. [72] also reported a good overall survival with 1.7 GBq 131I- [3] S.F. Altekruse, K.A. McGlynn, M.E. Reichman, J. Clin. Oncol. 27 (2009) 1485–
1491.
Lipiodol 1–8 months after surgery. It seems that there is no clini-
[4] H.E. Blum, H.C. Spangenberg, Arch Iran Med 10 (2007) 361–371.
cally significant adverse effect of adjuvant therapy by intra-arte- [5] M. Chiaramonte, T. Stroffolini, A. Vian, M.A. Stazi, A. Floreani, U. Lorenzoni, S.
rial131I-Lipiodol after curative liver resection for HCC [73]. These Lobello, F. Farinati, R. Naccarato, Cancer 85 (1999) 2132–2137.
results were also confirmed in a recent published study from Chua [6] L. Benvegnu, G. Fattovich, F. Noventa, F. Tremolada, L. Chemello, A. Cecchetto,
A. Alberti, Cancer 74 (1994) 2442–2448.
et al. [74]. It is necessary to perform more randomised controlled [7] F. Kondo, Hepatol Int 3 (2009) 283–293.
trials with more patients to confirm the role of 131I-Lipiodol as [8] H.B. El-Serag, K.L. Rudolph, Gastroenterology 132 (2007) 2557–2576.
an adjuvant treatment especially considering the timing and the [9] C.R. Davis, Cancer Control 17 (2010) 87–99.
[10] M.B. Thomas, D. Jaffe, M.M. Choti, J. Belghiti, S. Curley, Y. Fong, G. Gores, R.
administered dose. Kerlan, P. Merle, B. O’Neil, R. Poon, L. Schwartz, J. Tepper, F. Yao, D. Haller, M.
Mooney, A. Venook, J. Clin. Oncol. 28 (2010) 3994–4005.
12.2.2. Neo-adjuvant 131I-Lipiodol administration before liver [11] T.S. Helling, C.E. Woodall 3rd, J Gastrointest Surg 11 (2007) 76–81.
[12] J.T. Heckman, M.B. Devera, J.W. Marsh, P. Fontes, N.B. Amesur, S.E. Holloway,
transplantation/resection M. Nalesnik, D.A. Geller, J.L. Steel, T.C. Gamblin, Ann. Surg. Oncol. 15 (2008)
In a study by Raoul et al. curative resection of hepatocellular 3169–3177.
carcinoma or liver transplantation was preceded by 2 injections [13] S.M. Lin, C.J. Lin, C.C. Lin, C.W. Hsu, Y.C. Chen, Gut 54 (2005) 1151–1156.
[14] M.S. Copur, N. Engl. J. Med. 359 (2008) 2498 (author reply 98–99).
of 2.4 GBq 131I-Lipiodol (5 weeks preoperative) to reduce intrahe- [15] J.M. Llovet, S. Ricci, V. Mazzaferro, P. Hilgard, E. Gane, J.F. Blanc, A.C. de
patic recurrence. The procedure was well tolerated and associated Oliveira, A. Santoro, J.L. Raoul, A. Forner, M. Schwartz, C. Porta, S. Zeuzem, L.
with an objective tumour response in 56% and a complete histolog- Bolondi, T.F. Greten, P.R. Galle, J.F. Seitz, I. Borbath, D. Haussinger, T. Giannaris,
M. Shan, M. Moscovici, D. Voliotis, J. Bruix, N. Engl. J. Med. 359 (2008) 378–
ical response in 24% of 34 treated patients. An objective tumour re-
390.
sponse or necrosis of more than 90% of the tumour mass was [16] W.M. Lien, N.B. Ackerman, Surgery 68 (1970) 334–340.
observed in three-quarters of these patients. Brans et al. treated [17] J.L. Raoul, E. Boucher, Y. Rolland, E. Garin, Nat Rev Gastroenterol Hepatol 7
(2010) 41–49.
10 HCC patients with almost 1.8 GBq 131I-Lipiodol in a pilot study
[18] J.L. Raoul, P. Bourguet, J.F. Bretagne, R. Duvauferrier, S. Coornaert, P. Darnault,
followed by liver transplantation within 1–9 months [75]. The evi- A. Ramee, J.Y. Herry, J. Gastard, Radiology 168 (1988) 541–545.
dence of an anti-tumoural effect of 131I-Lipiodol was observed in [19] R.A. Al-Mufti, R.B. Pedley, D. Marshall, R.H. Begent, A. Hilson, M.C. Winslet, K.E.
50% of patients. However, performing randomised comparisons be- Hobbs, Br. J. Cancer 79 (1999) 1665–1671.
[20] J.H. Risse, C. Menzel, F. Grunwald, H. Strunk, H.J. Biersack, H. Palmedo, Rom J
tween surgery alone versus surgery after intra-arterial injection of Gastroenterol 13 (2004) 119–124.
131
I-labelled Lipiodol in patients with large but technically resect- [21] S. Maki, T. Konno, H. Maeda, Cancer 56 (1985) 751–757.
able tumours is needed. [22] B. Brans, K. Bacher, V. Vandevyver, P. Vanlangenhove, P. Smeets, H. Thierens,
R.A. Dierckx, L. Defreyne, Nucl. Med. Commun. 24 (2003) 391–396.
[23] E. Garin, P. Bourguet, in: H.J. Biersack, L.M. Freeman (Eds.), Clinical Nuclear
12.3. Therapy of liver metastases Medicine, Springer-Verlag, Berlin Heidelberg, 2007, pp. 491–508.
[24] H.S. Yoo, C.H. Park, J.H. Suh, J.T. Lee, D.I. Kim, B.S. Kim, M.T. Madsen, Cancer
Chemother Pharmacol 23 (1989) S54–S58.
The few studies carried out in this field are disappointing [25] Guidelines for 131I-ethiodised oil (Lipiodol) therapy. Eur J Nucl Med Mol
[18,47,76]. Biodistribution studies show that the ‘‘metastasis/ Imaging 2003;30:BP20–22.
non-tumoural liver’’ fixation ratios are not as high as that for [26] F. Giammarile. in: 23rd Annual Congress of the European Association of
Nuclear Medicine, 2010, Vienna, pp. 40–41.
HCC [18,77], probably due to a preferential fixing of 131I-Lipiodol
[27] R. Salem, R. Lewandowski, C. Roberts, J. Goin, K. Thurston, M. Abouljoud, A.
at the periphery of the metastases [23,76]. The decrease in re- Courtney, J. Vasc. Interv. Radiol. 15 (2004) 335–345.
sponse rates with increasing tumour size [57,78] is also observed [28] A. Kennedy, S. Nag, R. Salem, R. Murthy, A.J. McEwan, C. Nutting, A. Benson 3rd,
J. Espat, J.I. Bilbao, R.A. Sharma, J.P. Thomas, D. Coldwell, Int. J. Radiat. Oncol.
in liver metastases. This might be attributed to two factors: (1).
Biol. Phys. 68 (2007) 13–23.
A high percentage of large tumours have necrotic centres that take [29] L.A. Dawson, D. Normolle, J.M. Balter, C.J. McGinn, T.S. Lawrence, R.K. Ten
up the radiopharmaceutical poorly and the beta radiation from 131I Haken, Int. J. Radiat. Oncol. Biol. Phys. 53 (2002) 810–821.
can only penetrate up to 2.4 mm of soft tissue. Thus, Viable tumour [30] C. Greco, G. Catalano, A. Di Grazia, R. Orecchia, Tumori 90 (2004) 73–79.
[31] R. Murthy, R. Nunez, J. Szklaruk, W. Erwin, D.C. Madoff, S. Gupta, K. Ahrar, M.J.
cells in necrotic areas may not receive adequate radiation doses. Wallace, A. Cohen, D.M. Coldwell, A.S. Kennedy, M.E. Hicks, Radiographics 25
(2). The activity of 131I-Lipiodol used was insufficient to deliver (Suppl 1) (2005) S41–S55.
the required tumouricidal radiation doses [32]. [32] S. Ho, W.Y. Lau, T.W. Leung, P.J. Johnson, Cancer 83 (1998) 1894–1907.
[33] M. Nakajo, H. Kobayashi, K. Shimabukuro, K. Shirono, H. Sakata, M. Taguchi, N.
In a study by Bretagne et al. [47] no objective response was ob- Uchiyama, T. Sonoda, S. Shinohara, J. Nucl. Med. 29 (1988) 1066–1077.
served amongst 8 patients presenting with hepatic metastases [34] M.T. Madsen, C.H. Park, M.L. Thakur, J. Nucl. Med. 29 (1988) 1038–1044.
treated by 131I-Lipiodol, even if a clear and sustained analgesic ef- [35] H. Ahmadzadehfar, H.J. Biersack, S. Ezziddin, Semin. Nucl. Med. 40 (2010) 105–
121.
fect was found in 3 patients with pain. [36] S.A. Kwee, T.R. DeGrado, J.N. Talbot, F. Gutman, M.N. Coel, Semin. Nucl. Med.
37 (2007) 420–428.
13. Conclusion [37] J.N. Talbot, F. Gutman, L. Fartoux, J.D. Grange, N. Ganne, K. Kerrou, D. Grahek, F.
Montravers, R. Poupon, O. Rosmorduc, Eur J Nucl Med Mol Imaging 33 (2006)
1285–1289.
The effectiveness of 131I-Lipiodol treatment is proven both in [38] Y. Yamamoto, Y. Nishiyama, R. Kameyama, K. Okano, H. Kashiwagi, A. Deguchi,
the treatment of HCC with portal thrombosis and as an adjuvant M. Kaji, M. Ohkawa, J. Nucl. Med. 49 (2008) 1245–1248.
[39] J.H. Risse, C. Ponath, H. Palmedo, C. Menzel, F. Grunwald, H.J. Biersack, Eur. J.
to surgery after the resection of HCCs. It is at least as effective as Nucl. Med. 28 (2001) 914–918.
chemoembolization and is tolerated much better. Severe liver dys- [40] J.H. Risse, C. Rabe, D. Pauleit, K. Reichmann, C. Menzel, F. Grunwald, H. Strunk,
function represents theoretic contraindication for radioemboliza- H.J. Biersack, H. Palmedo, Nuklearmedizin 45 (2006) 185–192.
[41] S. Becker, V. Ardisson, N. Lepareur, O. Sergent, S. Bayat, N. Noiret, F. Gaboriau,
tion as well as for TACE. In such cases 131I-Lipiodol is an B. Clement, E. Boucher, J.L. Raoul, E. Garin, Nucl. Med. Biol. 37 (2010) 777–784.
alternative therapy option especially in tumours smaller than [42] E. Garin, S. Laffont, Y. Rolland, D. Olivie, J. Lecloirec, J.Y. Herry, E. Boucher, J.L.
6 cm. In addition, techniques using 90Y-labelled products tend to Raoul, P. Bourguet, Nucl. Med. Commun. 24 (2003) 671–678.
[43] S. Becker, S. Laffont, F. Vitry, Y. Rolland, J. Lecloirec, E. Boucher, J.L. Raoul, J.Y.
cost up to 10 times more than treatment with 131I-Lipiodol, which Herry, P. Bourguet, E. Garin, Nucl. Med. Commun. 29 (2008) 815–825.
has a very important impact on therapeutic approach in all coun- [44] H. Kobayashi, H. Hidaka, Y. Kajiya, P. Tanoue, H. Inoue, K. Ikeda, M. Nakajo, S.
tries with resource-constrained health care systems. Shinohara, Acta Radiol. Diagn. (Stockh) 27 (1986) 139–147.
[45] W.Y. Lui, R.S. Liu, J.H. Chiang, J.G. Lo, K.H. Lai, K.L. King, H.C. Cheng, Y.Y. Wei,
C.W. Chi, F.K. P’Eng, et al., Zhonghua Yi Xue Za Zhi (Taipei) 46 (1990) 125–133.
References [46] J.I. Raoul, J.F. Bretagne, J.P. Caucanas, E.A. Pariente, J. Boyer, J.C. Paris, H. Michel,
P. Bourguet, G. Victor, F. Therain, et al., Cancer 69 (1992) 346–352.
[1] D.M. Parkin, C.S. Muir, IARC Sci. Publ. (1992) 45–173. [47] J.F. Bretagne, J.L. Raoul, P. Bourguet, R. Duvauferrier, Y. Deugnier, R. Faroux, A.
[2] H.B. El-Serag, A.C. Mason, C. Key, Hepatology 33 (2001) 62–65. Ramee, J.Y. Herry, J. Gastard, Radiology 168 (1988) 547–550.
252 H. Ahmadzadehfar et al. / Methods 55 (2011) 246–252

[48] B. Lambert, C. Van de Wiele, Eur J Nucl Med Mol Imaging 32 (2005) 980–989. [64] T.C. Chua, F. Chu, S.P. Butler, R.J. Quinn, D. Glenn, W. Liauw, D.L. Morris, Cancer
[49] S. Jouneau, E. Vauleon, S. Caulet-Maugendre, E. Polard, A.C. Volatron, C. 116 (2010) 4069–4077.
Meunier, P. Tattevin, D. Montani, E. Garin, J.L. Raoul, P. Delaval, Chest [65] J.L. Raoul, E. Boucher, V. Roland, E. Garin, Q J Nucl Med Mol Imaging 53 (2009)
(2010). 348–355.
[50] M. Toubeau, C. Touzery, A. Berriolo-Riedinger, A. Cochet, F. Brunotte, L. [66] M.A. Vente, M. Wondergem, I. van der Tweel, M.A. van den Bosch, B.A.
Bedenne, J.P. Cercueil, D. Krause, Eur. J. Nucl. Med. 28 (2001) 669–670. Zonnenberg, M.G. Lam, A.D. van Het Schip, J.F. Nijsen, Eur. Radiol. 19 (2009)
[51] K. Bacher, B. Brans, M. Monsieurs, F. De Winter, R.A. Dierckx, H. Thierens, Eur J 951–959.
Nucl Med Mol Imaging 29 (2002) 1311–1316. [67] L. Marelli, V. Shusang, J.R. Buscombe, E. Cholongitas, R. Stigliano, N. Davies, J.
[52] J.L. Raoul, D. Guyader, J.F. Bretagne, R. Duvauferrier, P. Bourguet, D. Bekhechi, Tibballs, D. Patch, T. Meyer, A.K. Burroughs, J. Nucl. Med. 50 (2009) 871–877.
Y.M. Deugnier, M. Gosselin, J. Nucl. Med. 35 (1994) 1782–1787. [68] W.Y. Lau, T.W. Leung, S.K. Ho, M. Chan, D. Machin, J. Lau, A.T. Chan, W. Yeo, T.S.
[53] W.Y. Lau, E.C. Lai, Hepatobiliary Pancreat Dis Int 7 (2008) 237–257. Mok, S.C. Yu, N.W. Leung, P.J. Johnson, Lancet 353 (1999) 797–801.
[54] T. de Baere, P. Taourel, J.M. Tubiana, V. Kuoch, M. Ducreux, J. Lumbroso, A.J. [69] J.R. Buscombe, Nucl. Med. Commun. 23 (2002) 837–841.
Roche, Radiology 212 (1999) 665–668. [70] W.Y. Lau, E.C. Lai, T.W. Leung, S.C. Yu, Ann. Surg. 247 (2008) 43–48.
[55] J.H. Risse, F. Grunwald, W. Kersjes, H. Strunk, W.H. Caselmann, H. Palmedo, H. [71] E. Boucher, G. Bouguen, E. Garin, A. Guillygomarch, K. Boudjema, J.L. Raoul, J.
Bender, H.J. Biersack, Cancer Biother Radiopharm 15 (2000) 65–70. Nucl. Med. 49 (2008) 362–366.
[56] J.L. Raoul, D. Guyader, J.F. Bretagne, J.F. Heautot, R. Duvauferrier, P. Bourguet, D. [72] K.M. Ng, R. Niu, T.D. Yan, J. Zhao, E. McKay, F.C. Chu, D.L. Morris, HPB (Oxford)
Bekhechi, Y.M. Deugnier, M. Gosselin, Hepatology 26 (1997) 1156–1161. 10 (2008) 388–395.
[57] H.S. Yoo, J.T. Lee, K.W. Kim, B.S. Kim, H.J. Choi, K.S. Lee, C.I. Park, C.Y. Park, J.H. [73] C. Partensky, G. Sassolas, L. Henry, P. Paliard, G.J. Maddern, Arch. Surg. 135
Suh, J.J. Loh, Cancer 68 (1991) 1878–1884. (2000) 1298–1300.
[58] W.T. Leung, W.Y. Lau, S. Ho, M. Chan, N. Leung, J. Lin, K.C. Ho, C. Metreweli, P.J. [74] T.C. Chua, A. Saxena, F. Chu, S.P. Butler, R.J. Quinn, D. Glenn, D.L. Morris, Int. J.
Johnson, A.K. Li, J. Nucl. Med. 35 (1994) 1313–1318. Clin. Oncol. (2010).
[59] S. Bhattacharya, J.R. Novell, G.M. Dusheiko, A.J. Hilson, R. Dick, K.E. Hobbs, [75] B. Brans, F. De Winter, L. Defreyne, R. Troisi, P. Vanlangenhove, H. Van
Cancer 76 (1995) 2202–2210. Vlierberghe, B. Lambert, M. Praet, B. de Hemptinne, R.A. Dierckx, Cancer
[60] R.B. Rindani, T.J. Hugh, J. Roche, P.J. Roach, R.C. Smith, ANZ J Surg 72 (2002) Biother. Radiopharm. 16 (2001) 333–338.
210–214. [76] S. Perring, R. Hind, J. Fleming, S. Birch, V. Batty, I. Taylor, Nucl. Med. Commun.
[61] I. Borbath, R. Lhommel, L. Bittich, P. Goffette, L. Annet, B.E. Van Beers, M.A. 15 (1994) 34–38.
Bonny, S. Pauwels, Y. Horsmans, Eur. J. Gastroenterol. Hepatol. 17 (2005) 905– [77] R.E. Hind, M. Loizidou, S. Perring, J. Fleming, V. Batty, S. Birch, I. Taylor, Br. J.
910. Surg. 79 (1992) 952–954.
[62] E. Boucher, E. Garin, A. Guylligomarc’h, D. Olivie, K. Boudjema, J.L. Raoul, [78] H.S. Yoo, C.H. Park, J.T. Lee, K.W. Kim, C.S. Yoon, J.H. Suh, C.Y. Park, B.S. Kim, H.J.
Radiother. Oncol. 82 (2007) 76–82. Choi, K.S. Lee, et al., Cancer Chemother. Pharmacol. 33 (Suppl) (1994) S128–
[63] H.A. Kanhere, L.N. Leopardi, L. Fischer, M.I. Kitchener, G.J. Maddern, ANZ J Surg S133.
78 (2008) 371–376.

You might also like