9

Click here to load reader

Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Embed Size (px)

Citation preview

Page 1: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Peripheral benzodiazepine receptor ligands induce apoptosis and cellcycle arrest in human hepatocellular carcinoma cells and enhance

chemosensitivity to paclitaxel, docetaxel, doxorubicinand the Bcl-2 inhibitor HA14-1

Andreas P. Sutter1, Kerstin Maaser1, Patricia Grabowski1, Gesine Bradacs1, Kirsten Vormbrock1,Michael Hopfner1, Antje Krahn1, Bernhard Heine2, Harald Stein2, Rajan Somasundaram1,

Detlef Schuppan3, Martin Zeitz1, Hans Scherubl1,*

1Medical Clinic I, Charite-Universitatsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany2Institute of Pathology, Charite-Universitatsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany

3Department of Internal Medicine, Friedrich Alexander University, Erlangen, Germany

Background/Aims: Hepatocellular carcinoma (HCC) is one of the most common causes of cancer deaths worldwide.

Thus, novel therapies are urgently needed. A promising approach is the use of peripheral benzodiazepine receptor

(PBR) ligands which inhibit the proliferation of various tumors.

Methods: PBR expression both in human HCC cell lines and in tumor specimens of HCC patients was analyzed by

RT-PCR and immunostaining. To evaluate PBR ligands for the treatment of HCC, we tested their effects on human

HCC cells.

Results: PBR was localized to the mitochondria both of HCC cell lines and tumor tissues of HCC patients. In contrast,

normal liver did not express PBR. PBR ligands inhibited the proliferation of HCC cell lines by inducing apoptosis andcell cycle arrest. Apoptosis was characterized by a breakdown of the mitochondrial membrane potential, caspase-3

activation and nuclear degradation. Furthermore, pro-apoptotic Bax was overexpressed while anti-apoptotic Bcl-2 and

Bcl-XL were suppressed. Cell cycle was arrested both at the G1/S- and G2/M-checkpoints. Synergistic anti-neoplastic

effects were obtained by a combination of PBR ligands with cytostatic drugs (paclitaxel, docetaxel, doxorubicin), or

with an experimental Bcl-2 inhibitor.

Conclusions: This is the first report on the induction of apoptosis and cell cycle arrest by PBR ligands in HCC cells.

Moreover, PBR ligands sensitized HCC cells to taxans and doxorubicin.

q 2004 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.

Keywords: Bax; Bcl-2; Chemoresistance; Chemotherapy; Peripheral benzodiazepine receptor; Apoptosis; Cell cycle

0168-8278/$30.00 q 2004 European Association for the Study of the Liver. Pub

doi:10.1016/j.jhep.2004.07.015

Received 11 February 2004; received in revised form 21 June 2004;

accepted 12 July 2004; available online 29 July 2004

* Corresponding author. Tel.: C49-30-8445-3534; fax: C49-30-8445-

4481.

E-mail address: [email protected] (H. Scherubl).

Abbreviations: FGIN-1-27, N,N-di-n-hexyl-2-(4-fluorophenyl)indole-3-

acetamide; HCC, hepatocellular carcinoma; PBR, peripheral benzo-

diazepine receptor; PK 11195, 1-(2-chlorophenyl)-N-methyl-N-(1-methyl-

propyl)-3-isoquinolinecarboxamide; PTP, permeability transition pore.

1. Introduction

Hepatocellular carcinoma (HCC) is the fifth most

common malignancy in the world and is estimated to

cause half a million deaths annually. The incidence of HCC

is dramatically increasing in the USA, Europe and Asia,

most probably due to the increasing prevalence of hepatitis

C [1,2]. Unfortunately, treatment of HCC is unsatisfactory.

Curative ablation or resection of HCC, or liver transplan-

tation can be achieved only in a minority of patients. Local

tumor destruction, chemoembolisation or systemic

Journal of Hepatology 41 (2004) 799–807

www.elsevier.com/locate/jhep

lished by Elsevier B.V. All rights reserved.

Page 2: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Table 1

Clinicopathological characteristics of the patients

Patients (n) 32

Gender

Male 20

Female 12

Age (years)

Average 59.8

Range 16–79

Grading (of the HCC)

G1 12

G2 12

G3 7

N/A 1

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807800

chemotherapy are the remaining treatment options in

advanced HCC. However, overall survival is poor [3].

Therefore, innovative treatment approaches are urgently

needed.

Venturini et al. were the first to show an overexpression

of the peripheral benzodiazepine receptor (PBR) in human

hepatocellular carcinoma cells; they suggested the use of

PBR ligands as an innovative treatment of HCC [4]. PBR

ligands have been shown to inhibit the proliferation of

esophageal, colorectal and breast cancer cells by induction

of apoptosis and cell cycle arrest [5–7]. Moreover, PBR

ligands have been shown to overcome Bcl-2-mediated

chemoresistance in lung cancer [8].

The PBR was initially discovered as a binding site for

benzodiazepines in peripheral tissues. It is structurally and

pharmacologically distinct from the central-type benzo-

diazepine binding site which is associated with the GABAA

receptor [9]. PBR is mainly localized in the outer

mitochondrial membrane [5,6] but has also been detected

in the plasma membrane [10] and nucleus [11]. PBR is

overexpressed in tumors of the liver, colon, and breast

[11–13], thus offering the possibility of a tumor-targeted

therapy [14].

PBR has not yet been studied functionally in hepatocel-

lular carcinoma. Here we report on the mitochondrial

expression of PBR in hepatocellular carcinoma. We

demonstrate the anti-proliferative and pro-apoptotic actions

of PBR ligands in HCC cells. Moreover, we provide new

insight into PBR-ligand-mediated apoptosis and evidence

for a synergistic anti-neoplastic action of PBR ligands and

cytostatics.

2. Materials and methods

2.1. Cell lines and drugs

The human HCC cell lines Huh-7 [15] and HepG2 [16] were cultured inRPMI1640 medium containing 10% fetal bovine serum and 100 U/mlpenicillin and streptomycin. Primary human keratinocytes [17] were grownin Keratinocyte SFM (Gibco, Paisley, United Kingdom). Cells were kept ina humidified atmosphere (5% CO2) at 37 8C.

Cells were incubated with culture medium containing FGIN-1-27, PK11195 (Tocris, Bristol, United Kingdom), or FGIN-1-52 [18]. Forcombination treatment, cells were incubated simultaneously with PBRligands and one of the following agents: Paclitaxel (Sigma), docetaxel(Fluka Chemie, Buchs, Switzerland), doxorubicin (Sigma), or HA14-1(Alexis, Grunberg, Germany).

2.2. Analysis of peripheral benzodiazepine

receptor expression

Semi-quantitative analysis of PBR mRNA expression was carried outby RT-PCR with the number of cycles at which the band intensity increasedlinearly with the amount of mRNA used. Total RNA was extracted withRNAClean (Hybaid, London, United Kingdom) and subsequently digestedwith DNAse I (Gibco, Karlsruhe, Germany). Oligo-dT-primers and theSuperScript Preamplification-Kit (Gibco) were used for cDNA synthesis.PCR reactions were performed as previously described [5,19]. The identityof the PCR product as being a fragment of PBR was confirmed by

sequencing (Invitek Sequencing Service GmbH, Berlin, Germany). PBRgene expression was standardized to the expression of the housekeepinggene b-actin.

For analysis of PBR protein expression, cells were immunostained asdescribed [5]. In brief, samples were fixed and permeabilized andsubsequently incubated with a polyclonal anti-PBR antibody (1:50, R&Dsystems, Wiesbaden, Germany), or isotypic control rabbit IgG1 (DAKO,Hamburg, Germany). Cells were then incubated with a secondary FITC-labeled goat-anti-rabbit IgG antibody (BD Pharmingen, Heidelberg,Germany). Fluorescence was detected by flow cytometry on a FACSCa-libur (Becton Dickinson, Heidelberg, Germany) and analyzed usingCellQuest software. PBR expression was further studied immunohisto-chemically both in normal liver and in HCCs of 32 patients whohad undergone surgery for HCC. For detailed clinicopathological datasee Table 1. Microsections were deparaffinized, rehydrated, and automati-cally stained, as described [5]. In brief, sections were stained with the anti-PBR antibody 8D7 [20]. After washing, samples were incubated with asecondary anti-mouse antibody and subsequently with the alkalinephosphatase-anti-alkaline phosphatase (APAAP) complex (DAKO). Stain-ing was detected using the ‘fast-red system’ (DAKO), and samples werecounterstained in Mayer’s haematoxylin. The subcellular localization ofPBR in HCC cells was analyzed by immunofluorescence microscopy, aspreviously described [5]. In brief, cells were stained with the mitochondrialdye CMTMRos, fixed, permeabilized, and subsequently incubated with aprimary anti-PBR polyclonal antibody (R&D systems). Thereafter, cellswere incubated with a secondary FITC-labeled goat-anti-rabbit antibody.Imaging was performed using the inverted confocal microscope LSM 510with a 63!/1.2 W Korr objective (Zeiss, Oberkochen, Germany).

2.3. Radioligand binding assays

Cells were homogenized in ice-cold PBS using a small potter-typemechanical homogenizer. Protein concentration was determined using theBradford assay (BioRad, Hercules, CA). Total [3H]PK 11195 (specificactivity 83.5 Ci/mmol; NEN Life Science Products) binding wasdetermined using serial dilutions of the labeled ligand (0.5–15 nM). Non-specific binding was determined by adding 13.3 mM unlabeled PK 11195.Each sample contained 50–100 mg protein in PBS in a final volume of300 ml. Samples were incubated at 4 8C overnight, and each was transferredby vacuum filtration onto 25 mm Whatman filters using a Brandel harvester(Brandel, Gaithersburg, MD). Bound [3H]PK 11195 was quantified byliquid scintillation spectrometry. Dissociation constants (KD) and thenumber of binding sites (Bmax) were determined by Curve-Fit (Prismversion 3.0; Graph Pad Software Inc., San Diego, CA).

2.4. Determination of cell number

Cell number was evaluated by crystal violet staining, as described [21].In brief, cells in 96-well plates were fixed with 1% glutaraldehyde. Thencells were stained with 0.1% crystal violet. The unbound dye was removedby washing with water. Bound crystal violet was solubilized with 0.2%

Page 3: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Fig. 1. Expression of PBR in hepatocellular carcinoma (HCC) cells. (A)

PBR mRNA expression in Huh-7 (lane 1) and HepG2 cells (lane 2)

detected by RT-PCR. The expression of the housekeeping gene b-actin

(amplicon: 822 bp) in Huh-7 (lane 3) and HepG2 cells (lane 4) was

analyzed for standardization. (B) PBR protein expression in HCC cells

was shown by flow cytometry. Huh-7 (left panel) and HepG2 cells (right

panel) were stained with a polyclonal anti-PBR antibody with (gray

area) or without (gray line) previous membrane permeabilization.

Black line: isotypic control.

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807 801

Triton-X-100. Light extinction which increases linerarly with the cellnumber was analyzed at 570 nm using an ELISA-Reader.

2.5. Detection of apoptosis

Changes in DJM were assessed as described [21]. In brief, cells werestained with 5,5 0,6,6 0-tetrachloro-1,10,3,3 0-tetraethyl-benzimidazolylcarbo-cyanine iodide (JC-1, 1 mg/ml, Molecular Probes, Eugene, OR) for 15 minat 37 8C in the dark, prior to analysis by flow cytometry.

Caspase-3 activity was assessed as described [22]. The activity ofcaspase-3 was calculated from the cleavage of the fluorogenic substrateDEVD-AMC (Calbiochem-Novabiochem, Bad Soden, Germany).

The proportion of apoptotic cells was determined by quantifying thepercentage of sub-G1 (hypodiploid) cells after flow cytometric analysis ofpropidium iodide-stained isolated nuclei.

2.6. Western blotting

Western blotting was performed as described [19]. Blots were blockedin 1% non-fat dry milk for 30 min, and then incubated at 4 8C overnightwith anti-human Bcl-2 (1:200, Novo Castra Laboratories, Newcastle uponTyne, United Kingdom), Bcl-XL (1:200, Santa Cruz Biotechnology, CA),Bax (1:1000, Santa Cruz) or b-actin (1:5000, Sigma). Band intensities wereanalyzed densitometrically using TINA software (raytest Isotopenmessge-rate, Straubenhardt, Germany).

2.7. Cell cycle analysis

Cell cycle analysis was performed by the method of Vindelov andChristensen [23]. Cells were trypsinized, washed, and the nuclei wereisolated using CycleTest PLUS DNA Reagent Kit (Becton Dickinson).DNA was stained with propidium iodide according to the manufacturers’instructions. The DNA content of the nuclei was detected by flow cytometryand analyzed using CellFit software (Becton Dickinson).

2.8. Statistical analysis

If not stated otherwise, means of four independent experimentsGSEMare shown. Individual drug treatment and patient groups were compared bythe unpaired, two-tailed Mann-Whitney U-test. Dichotomized variableswere compared using the chi square test. P values were considered to besignificant at !0.05.

Fig. 2. Localization of PBR in HCC tissues and cells. (A, B)

Immunohistochemical detection of PBR (red) in HCC tissue and in

adjacent non-neoplastic tissue (NO). (A) BarZ200 mm; (B) BarZ100 mM. (C–E) For immunocytochemistry, PBR was immunostained

with a polyclonal antibody (C), and mitochondria were marked with

CMTMRos (D). Superposition of both fluorescence images resulted in a

bright yellow color (E), indicating a co-localization of PBR and

mitochondria in Huh-7 cells. BarZ10 mM. (For interpretation of the

references to color in this figure legend, the reader is referred to the

web version of this article).

3. Results

3.1. Expression and subcellular localization of PBR

in hepatocellular carcinoma cells

Both Huh-7 and HepG2 cells expressed mRNA tran-

scripts of PBR (Fig. 1A). Expression of PBR protein was

detected in permeabilized Huh-7 and HepG2 cells, whereas

in non-permeabilized cells, no PBR-specific fluorescence

was observed, indicating an intracellular localization of PBR

(Fig. 1B). The expression levels both of PBR transcript and

protein did not notably differ between the two cell lines. In

11 of 32 analyzed HCC patients, PBR was expressed in HCC

tissues, whereas the respective non-neoplastic liver tissues of

the same patients were negative for PBR. In this small group

of patients PBR expression did neither correlate with age,

gender, histopathological grading, nor with underlying liver

disease (also see Table 1). In all 11 PBR-positive HCCs, the

specific PBR staining was observed to be unevenly

distributed within the cytoplasm. No specific PBR staining

was observed in the plasma membrane nor in the cell nuclei,

suggesting that the PBR is located in the mitochondria of

HCC cells (Fig. 2A and B). The mitochondrial localization

Page 4: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Fig. 3. Anti-proliferative effects of PBR ligands. The PBR ligands

FGIN-1-27 and PK 11195 induced a time- and dose-dependent growth

inhibition in Huh-7 (A) and HepG2 (B) cells. Growth inhibition was

significant for 10–100 mM FGIN-1-27, and for 25–100 mM PK 11195

(P!0.05). Means of four independent experiments are shown. (C)

PBR- and tumor cell specificity of the growth inhibitory effects. In

contrast to PBR ligands, FGIN-1-52 did not inhibit the growth of Huh-7

(white columns) or HepG2 cells (hatched columns; Fig. 2C, left panel).

Non-malignant human primary keratinocytes remained nearly unaf-

fected by incubation with 50 mM FGIN-1-27 (white columns) or 75 mM

PK 11195 (black columns), in contrast to their inhibitory actions in

Huh-7 and HepG2 cells (Fig. 2C, right panel).

Table 2

Growth inhibition induced by the PBR ligands FGIN-1-27 and PK 11195, th

inhibitor HA14-1 in Huh-7 and HepG2 cells

Drug Huh-7

IC10 IC50 IC90

FGIN-1-27 4!103G1!103 2!104G3!103 O1!105

PK 11195 1!104G2!103 4.3!104G4!103 8.9!104G5

Paclitaxel 1.7G0.9 7.1G1 O25

Docetaxel 2.1G1.1 7.9G2 O25

Doxorubicin 140G60 652G57 O1!103

HA14-1 8!103G3!103 2.5!104G3!103 4.3!104G2

The IC10, IC50, and IC90 values (nM) were calculated from interpolations of the d

experiments.

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807802

of PBR was further confirmed for Huh-7 cells

by simultaneously staining mitochondria and PBR

(Fig. 2C–E). Green fluorescence of PBR was detected within

the cytoplasm, but neither in the cell membrane nor in the

nucleus (C). The staining pattern was comparable to the red

one obtained by staining of mitochondria (D). Superposition

of both images resulted in a yellow color, indicating a

co-localization of PBR and mitochondria (E).

3.2. [3H]PK 11195 binding properties of PBR in

hepatocellular carcinoma cells

PBR expressed in liver cancer cell lines binds with high

affinity [3H]PK 11195 (Huh-7: KDZ0.42G0.02 nM,

HepG2: KDZ1.04G0.36 nM), in good agreement with a

previous study in liver cancer tissues [24]. Saturation

isotherm analysis revealed that [3H]PK 11195 binding

capacity (Bmax) amounted to 234.05G18.69 fmol/mg in

Huh-7 and to 97.47G23.90 fmol/mg in HepG2 cells.

3.3. Inhibition of cell proliferation by PBR ligands

The PBR ligands FGIN-1-27 and PK 11195 (10–

100 mM) dose-dependently inhibited the proliferation of

Huh-7 (Fig. 3A) and HepG2 cells (Fig. 3B). After 96 h of

incubation with FGIN-1-27 or PK 11195, a maximal

decrease in cell growth by 80–100% was observed. The

IC10, IC50 and IC90 values are given in Table 2. While PK

11195 (100 mM) decreased original cell numbers after 96 h,

FGIN-1-27 was predominantly cytostatic.

Despite the indoleacetamide structure of FGIN-1-52

being similar to FGIN-1-27, it displays almost no affinity

to PBR [18,25]. Accordingly, FGIN-1-52 did not inhibit

the proliferation of HCC cells, even at concentrations as

high as 100 mM (Fig. 3C, left panel), suggesting PBR

specificity of growth inhibition. Next, we analyzed the

tumor specificity of growth inhibition using primary

human keratinocytes as a model of non-malignant

epithelial tissue. FGIN-1-27 or PK 11195 (10–75 mM)

did not inhibit the growth of PBR-expressing [5]

keratinocytes (Fig. 3C, right panel).

e cytostatic drugs paclitaxel, docetaxel and doxorubicin, and the Bcl-2

HepG2

IC10 IC50 IC90

5!103G1!103 2!104G2!103 O1!105

!103 2.9!104G3!103 5.5!104G3!103 9!104G6!103

1G0.1 4.5G0.9 O25

1G0.3 6.5G1 O25

38G10 457G100 O 1!103

!103 2.2!104G2!103 2.8!104G1!103 4!104G3!103

ose–response relationships. Data are derived from at least four independent

Page 5: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Fig. 4. Mitochondrial alterations and apoptosis induction by PBR

ligands. Huh-7 (black symbols) and HepG2 cells (open symbols) cells

were incubated with 10–100 mM FGIN-1-27 (squares) or PK 11195

(circles). FGIN-1-27 and PK 11195 dose-dependently decreased the

DJM after 24 h of incubation (A), induced caspase-3 activation after

48 h of incubation (B), and increased the proportion of apoptotic cells

measured as subdiploidy after 96 h of incubation (C). Data are shown

as meansGSEM of 4 independent experiments. *Statistical significance

(P!0.05) compared to untreated controls.

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807 803

3.4. PBR ligands induced DJM disruption and apoptosis

After 24 h, FGIN-1-27 and PK 11195 dose-dependently

induced a significant decrease of the mitochondrial

membrane potential (DJM) in HCC cells (Fig. 4A).

Furthermore, PBR ligands induced an increase in caspase-

3 activity after 48 h (Fig. 4B). They also increased the

incidence of nuclear apoptosis after 96 h of treatment (Fig.

4C). While a modest but significant apoptotic peak was

observed after treatment with FGIN-1-27, PK 11195

induced a stronger increase of apoptotic cells. This could

explain the cytotoxic effects of PK 11195 but not of FGIN-

1-27 after 96 h (Fig. 3).

3.5. Modulation of Bcl-2, Bcl-XL and Bax expression

by PBR ligands

As mitochondrial alterations occurred during PBR

ligand-induced apoptosis, the expression levels of mito-

chondrial pro-(Bax) and anti-apoptotic (Bcl-2 and Bcl-XL)

proteins were analyzed after 96 h of treatment. In HepG2

cells, Bcl-2 and Bcl-XL expression were downregulated,

whereas Bax expression was increased (Fig. 5). Due to these

alterations, the Bax/Bcl-XL ratio increased upon PBR ligand

treatment. Similarly, this ratio was dose-dependently

elevated by either FGIN-1-27 (up to 1.5-fold) or PK

11195 (up to 8.5-fold) in Huh-7 cells.

3.6. PBR ligands induce cell cycle arrest in the G0/G1

and G2/M phase

Treatment with PBR ligands (24 h) dose-dependently

arrested Huh-7 cells in the G0/G1-phase of the cell cycle

(Fig. 6A). In HepG2 cells, both a dose-dependent G0/G1

arrest and, at high concentrations (100 mM), a G2/M arrest

was observed (Fig. 6B).

Fig. 5. PBR ligands modulate the expression of pro- and anti-apoptotic

proteins. Modulation of protein expression in HepG2 cells was assessed

by Western blot analysis after 96 h of treatment with PBR ligands. The

anti-apoptotic proteins Bcl-XL and Bcl-2 were dose-dependently

downregulated by PBR ligands, whereas the pro-apoptotic protein

Bax was upregulated. A representative result out of three independent

experiments is shown.

Page 6: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Fig. 6. Induction of cell cycle arrest in the G0/G1 and G2/M phases by

PBR ligands. After 24 h of incubation of Huh-7 (A) and HepG2 cells (B)

with PBR ligands, cells accumulated in the G0/G1 phase (white

columns) of the cell cycle, while the proportion of cells in the S phase

(hatched columns) decreased. Means of four independent experiments

are shown. When compared to control, significant differences of the

proportion of cells in the G0/G1 phase of the cell cycle were observed in

response to FGIN-1-27 and PK 11195 from 50–100 mM (P!0.05).

Additionally, a significant (P!0.05) arrest in the G2/M phase (black

columns) was observed in HepG2 cells after treatment with FGIN-1-27

or PK 11195 (100 mM).

Fig. 7. Synergistic growth inhibition of HCC cells by cytostatic drugs or

the Bcl-2 inhibitor HA14-1 when combined with the PBR ligand FGIN-

1-27. Combination treatment with sub-IC50 concentrations of cytostatic

agents (paclitaxel, docetaxel, doxorubicin) or the Bcl-2 inhibitor

HA14-1 and the PBR-ligand FGIN-1-27 for 96 h led to synergistic

growth inhibitory effects in Huh-7 (A) and HepG2 cells (B). Black bars

indicate the values of the calculated additive growth inhibition. Data

are given as percentage of untreated controls (meansGSEM of at least

3 independent experiments).

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807804

3.7. PBR ligands chemosensitize HCC cells to treatment

with paclitaxel, docetaxel, doxorubicin, or a Bcl-2 inhibitor

Next, we investigated whether the combination of PBR

ligands with either cytostatic drugs or the Bcl-2 inhibitor

HA14-1 was superior to the anti-proliferative treatment with

single agents. First, the IC10, IC50 and IC90 values of

the taxans paclitaxel and docetaxel, the intercalating agent

doxorubicin, and HA14-1 were determined after 96 h of

treatment (Table 2). The IC50 values of taxans were the

lowest (low nanomolar range), followed by doxorubicin

(high nanomolar range), and HA14-1 (micromolar range).

For combination treatments, individual drugs were used

at two different concentrations between their respective IC10

and IC50 values. The combination of either cytostatic drug

or HA14-1 with FGIN-1-27 acted in a synergistic or additive

way (Fig. 7). Similar results were obtained with PK 11195.

Paclitaxel or docetaxel or doxorubicin at concentrations

inducing 10–50% growth inhibition on its own caused

growth inhibition by 50–90% when each of them was

combined with one of the PBR ligands. In Huh-7 cells, the

most effective synergism (20% observed versus 53%

calculated growth) was seen for paclitaxel plus PK 11195

(10 mM), whereas in HepG2 cells PK 11195 (10 mM) most

dramatically enhanced doxorubicin-induced growth inhi-

bition (28% observed versus 69% calculated growth). In

contrast to those synergisms, the combination of either PBR

ligand with HA14-1 proved to be less than additive in Huh-7

cells.

Based on the synergism between PBR ligands and

paclitaxel, we next investigated whether this combination

led to synergistic induction of apoptosis and/or cell cycle

arrest in Huh-7 cells. PBR ligands (at 50 mM) or paclitaxel

(at 2.5 nM) by themselves induced only a modest apoptotic

response after 96 h. However, the combination of PBR

ligands and paclitaxel led to a dramatic increase of apoptotic

cells compared to treatment with either agent alone (Fig. 8).

Page 7: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

Fig. 8. Synergistic apoptotic effects of a combination of PBR ligands

and paclitaxel in Huh-7 cells. The combination of either the PBR ligand

FGIN-1-27 or PK 11195 with paclitaxel led to a synergistic increase of

hypodiploid Huh-7 cells after 96 h of treatment. The percentage of sub-

G1 apoptotic cells is noted on each histogram. Representative data

from three independent experiments are shown.

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807 805

4. Discussion

In this study we show that the growth of human

hepatocellular carcinoma (HCC) cells is inhibited by PBR

ligands. In addition, PBR ligands sensitized HCC cells to

cytostatic agents.

As the overexpression of mitochondrial PBR in HCC

[12] has recently been discussed controversially [26], we

evaluated PBR expression in HCCs of 32 patients. No PBR

expression was observed in normal liver. Yet, PBR was

found to be expressed in 11 out of 32 HCCs. Thus, one third

of HCC patients may respond to a PBR ligand-based

therapy. In contrast to our findings, PBR expression had

been described in earlier reports [12,26] to occur even in

normal liver. The reasons for this discrepancy are unclear.

In esophageal, colorectal and breast cancer, PBR protein is

expressed both in tumoral and normal tissues. It is

overexpressed in 33% (esophageal) or 88% (colorectal) of

the tumor tissues [5,6]. In breast cancer, PBR expression

correlates with the tumor stage [26].

The PBR ligands FGIN-1-27 and PK 11195 induced both

a dose- and time-dependent growth inhibition and apoptosis

in the PBR-expressing HCC cell lines HepG2 and Huh-7,

suggesting that PBR ligands may antagonize the mitogenic

and anti-apoptotic intrinsic functions ascribed to PBR

[27,28]. The fact that the anti-proliferative effects of PBR

ligands were observed in different cancer types [5,6,29]

suggests that PBR ligands interfere with a common

signaling pathway. As the anti-proliferative effects of PBR

ligands were found in tumor cells but not in non-malignant

keratinocytes (Fig. 3) or hepatocytes [30,31], PBR ligands

appear to be tumor-selective agents [5,32]. The reason why

PBR ligands induced growth inhibition of tumor cells but

not of keratinocytes is yet unknown.

Furthermore, we show that PBR ligands sensitize HCC

cells to cytostatic drugs. The addition of PBR ligands led to

a synergistic anti-neoplastic action with either paclitaxel, or

docetaxel, or doxorubicin. Thus, the dose effectiveness of

doxorubicin was doubled, the one of paclitaxel or docetaxel

was increased five-fold. The synergistic enhancement of

paclitaxel-induced growth inhibition of Huh-7 cells was

shown to be due to a dramatic enhancement of apoptosis.

These findings argue for combination therapies of doxor-

ubicin or taxans with PBR ligands. By inhibiting Bcl-2

expression, PBR ligands may also allow to overcome Bcl-2-

mediated chemoresistance in HCC [8,33].

We further showed that a cell cycle arrest contributed to

the anti-proliferative effects of PBR ligands. The proportion

of HCC cells in the G0/G1 phase significantly increased

upon treatment with PBR ligands. In addition, a G2/M arrest

was observed in HepG2 cells. Thus, our results suggest that

PBR ligands can act both at the G1/S and G2/M checkpoints

in HCC cells, which has previously been found for breast

cancer cells [7]. In colorectal and esophageal cancer cells,

however, only a G1/S arrest was found [5,6]. The exact

mechanisms by which PBR ligands interfere with cell cycle

regulation are still under investigation. Recently, we

identified the p38MAPK pathway as a key regulator of

PBR-ligand-mediated G1/S arrest and apoptosis in esopha-

geal cancer cells [19].

Moreover, we observed a significant rise of apoptotic cells

after treatment with PBR ligands. Mitochondrial alterations

are well-known initiating events in the process of apoptosis

[34]. PBR was found to be localized in the mitochondria of

all PBR-expressing HCCs studied. Within the mitochondrial

membrane, PBR has been suggested to contribute to the

formation of the permeability transition pore [35]. The

opening of this pore, leading to DJM breakdown, is

considered to play a critical role in the initiation of apoptosis

[36]. Similar to our findings in colorectal and esophageal

cancer cells [5,6], our present results suggest that a drop in

DJM and the activation of caspase-3 are involved in PBR

ligand-induced apoptosis of HCC cells. The permeability

transition pore is known to interact with proteins of the Bcl-2

family [37,38]. PBR expression correlates positively with

Bcl-XL and negatively with Bax expression during apopto-

sis, suggesting an interaction between Bcl-2 proteins and

PBR [30,39]. We herein demonstrate the induction of Bax

and the suppression of Bcl-2 and Bcl-XL by PBR ligands.

The PBR ligand-induced suppression of Bcl-2 may

sensitize Bcl-2-overexpressing cells to chemotherapy [8,

40,41]. Another strategy against Bcl-2-mediated

Page 8: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807806

(chemo)resistance is the administration of the Bcl-2

inhibitor HA14-1, a useful parental compound for the

future development of clinically applicable agents [42]. The

down-regulation of Bcl-2 by PBR ligands combined with

its inhibition by HA14-1 proved to inhibit the growth of

HepG2 cells synergistically. Thus, we conclude that Bcl-2

is an attractive target for innovative treatment strategies of

hepatocellular carcinoma which can be effectively com-

bined with PBR ligands.

Although PK 11195 binding affinity in HCC cells was

determined to be in the low nanomolar range, the fact that

FGIN-1-52 [18] did not have any anti-proliferative effects

on HCC cells suggests that the effect of FGIN-1-27 was

PBR-specific. PBR-specificity of pro-apoptotic and anti-

mitotic signaling was previously addressed in esophageal

and colorectal carcinoma cells [5,6]. The quantitative

discrepancy between the micromolar ligand concentrations

necessary to induce apoptosis and cell cycle arrest and the

nanomolar binding affinities of PBR ligands has been

extensively discussed before [5,6].

Liver toxicity of alpidem, another PBR ligand, in

patients has been suggested to be due to its interaction

with mitochondrial permeability transition [43]. More-

over, the inhibitory actions of PBR ligands on tetra-

pyrrole transport may lead to long-term liver cytotoxicity

[44]. However, PBR ligands have been safely adminis-

tered in in vivo studies showing no short-term toxicity

and being well-tolerated [8,45]. In line with these results,

no acute toxicity on rat hepatocytes was found [30].

Nevertheless, long-term effects of the PBR ligands

applied in this study should be addressed in future

studies, potentially requiring the development of ligands

lacking this property.

In conclusion, in this report we demonstrate PBR-ligand-

mediated growth inhibition of HCC cells by induction of

apoptosis and cell cycle arrest. PBR ligands qualify for the

development of novel tumor-specific therapies. Further-

more, we suggest the chemosensitization of HCC cells by

PBR ligands as a new approach for enhancing the

susceptibility of HCC cells to chemotherapy.

Acknowledgements

We are indebted to Pierre Carayon who generously

provided us with the anti-PBR antibody 8D7. We thank

Bastian Gerst for expert technical assistance and Yelda

Oezdem for preparing the immunohistochemical slides.

We are indebted to Carola Muller and Holger Seltmann for

kindly providing us with primary human keratinocytes. This

study was supported by grants of the Deutsche Krebshilfe

and Berliner Krebsgesellschaft. Gesine Bradacs was

supported by a scholarship from the DFG, Graduiertenkol-

leg 276/3, ‘signal transduction and recognition’.

References

[1] El Serag HB, Davila JA, Petersen NJ, McGlynn KA. The continuing

increase in the incidence of hepatocellular carcinoma in the United

States: an update. Ann Intern Med 2003;139:817–823.

[2] McGlynn KA, Tsao L, Hsing AW, Devesa SS, Fraumeni Jr JF.

International trends and patterns of primary liver cancer. Int J Cancer

2001;94:290–296.

[3] El Serag HB, Mason AC, Key C. Trends in survival of patients with

hepatocellular carcinoma between 1977 and 1996 in the United States.

Hepatology 2001;33:62–65.

[4] Venturini I, Zeneroli ML, Corsi L, Baraldi C, Ferrarese C, Pecora N,

et al. Diazepam binding inhibitor and total cholesterol plasma levels

in cirrhosis and hepatocellular carcinoma. Regul Pept 1998;74:

31–34.

[5] Sutter AP, Maaser K, Hopfner M, Barthel B, Grabowski P, Faiss S,

et al. Specific ligands of the peripheral benzodiazepine receptor

induce apoptosis and cell cycle arrest in human esophageal cancer

cells. Int J Cancer 2002;102:318–327.

[6] Maaser K, Hopfner M, Jansen A, Weisinger G, Gavish M,

Kozikowski AP, et al. Specific ligands of the peripheral benzo-

diazepine receptor induce apoptosis and cell cycle arrest in human

colorectal cancer cells. Br J Cancer 2001;85:1771–1780.

[7] Carmel I, Fares FA, Leschiner S, Scherubl H, Weisinger G, Gavish M.

Peripheral-type benzodiazepine receptors in the regulation of

proliferation of MCF-7 human breast carcinoma cell line. Biochem

Pharmacol 1999;58:273–278.

[8] Decaudin D, Castedo M, Nemati F, Beurdeley-Thomas A, De

Pinieux G, Caron A, et al. Peripheral benzodiazepine receptor ligands

reverse apoptosis resistance of cancer cells in vitro and in vivo.

Cancer Res 2002;62:1388–1393.

[9] Braestrup C, Squires RF. Specific benzodiazepine receptors in rat

brain characterized by high-affinity (3H)diazepam binding. Proc Natl

Acad Sci USA 1977;74:3805–3809.

[10] Garnier M, Boujrad N, Oke BO, Brown AS, Riond J, Ferrara P, et al.

Diazepam binding inhibitor is a paracrine/autocrine regulator of

Leydig cell proliferation and steroidogenesis: action via peripheral-

type benzodiazepine receptor and independent mechanisms. Endo-

crinology 1993;132:444–458.

[11] Hardwick M, Fertikh D, Culty M, Li H, Vidic B, Papadopoulos V.

Peripheral-type benzodiazepine receptor (PBR) in human breast

cancer: correlation of breast cancer cell aggressive phenotype with

PBR expression, nuclear localization, and PBR-mediated cell

proliferation and nuclear transport of cholesterol. Cancer Res 1999;

59:831–842.

[12] Venturini I, Alho H, Podkletnova I, Corsi L, Rybnikova E, Pellicci R,

et al. Increased expression of peripheral benzodiazepine receptors and

diazepam binding inhibitor in human tumors sited in the liver. Life Sci

1999;65:2223–2231.

[13] Maaser K, Grabowski P, Sutter AP, Hopfner M, Foss HD, Stein H,

et al. Overexpression of the peripheral benzodiazepine receptor is a

relevant prognostic factor in stage III colorectal cancer. Clin Cancer

Res 2002;8:3205–3209.

[14] Guo P, Ma J, Li S, Guo Z, Adams AL, Gallo JM. Targeted delivery of

a peripheral benzodiazepine receptor ligand-gemcitabine conjugate to

brain tumors in a xenograft model. Cancer Chemother Pharmacol

2001;48:169–176.

[15] Nakabayashi H, Taketa K, Miyano K, Yamane T, Sato J. Growth of

human hepatoma cells lines with differentiated functions in

chemically defined medium. Cancer Res 1982;42:3858–3863.

[16] Aden DP, Fogel A, Plotkin S, Damjanov I, Knowles BB. Controlled

synthesis of HBsAg in a differentiated human liver carcinoma-derived

cell line. Nature 1979;282:615–616.

[17] Stadler R, Muller R, Orfanos CE. Effect of recombinant alpha

A-interferon on DNA synthesis and differentiation of human

keratinocytes in vitro. Br J Dermatol 1986;114:273–277.

Page 9: Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin

A.P. Sutter et al. / Journal of Hepatology 41 (2004) 799–807 807

[18] Kozikowski AP, Ma D, Brewer J, Sun S, Costa E, Romeo E, et al.

Chemistry, binding affinities, and behavioral properties of a new class

of antineophobic mitochondrial DBI receptor complex (mDRC)

ligands. J Med Chem 1993;36:2908–2920.

[19] Sutter AP, Maaser K, Barthel B, Scherubl H. Ligands of the peripheral

benzodiazepine receptor induce apoptosis and cell cycle arrest in

oesophageal cancer cells: involvement of the p38MAPK signalling

pathway. Br J Cancer 2003;89:564–572.

[20] Carayon P, Portier M, Dussossoy D, Bord A, Petitpretre G, Canat X,

et al. Involvement of peripheral benzodiazepine receptors in the

protection of hematopoietic cells against oxygen radical damage.

Blood 1996;87:3170–3178.

[21] Hopfner M, Sutter AP, Beck NI, Barthel B, Maaser K, Jockers-

Scherubl MC, et al. Meta-iodobenzylguanidine induces growth

inhibition and apoptosis of neuroendocrine gastrointestinal tumor

cells. Int J Cancer 2002;101:210–216.

[22] Maaser K, Hopfner M, Kap H, Sutter AP, Barthel B, von Lampe B,

et al. Extracellular nucleotides inhibit growth of human oesophageal

cancer cells via P2Y(2)-receptors. Br J Cancer 2002;86:636–644.

[23] Vindelov L, Christensen IJ. An integrated set of methods for routine

flow cytometric DNA analysis. Methods Cell Biol 1990;33:127–137.

[24] Venturini I, Zeneroli ML, Corsi L, Avallone R, Farina F, Alho H, et al.

Up-regulation of peripheral benzodiazepine receptor system in

hepatocellular carcinoma. Life Sci 1998;63:1269–1280.

[25] Wang JK, Taniguchi T, Spector S. Structural requirements for the

binding of benzodiazepines to their peripheral-type sites. Mol

Pharmacol 1984;25:349–351.

[26] Han Z, Slack RS, Li W, Papadopoulos V. Expression of peripheral

benzodiazepine receptor (PBR) in human tumors: relationship to

breast, colorectal, and prostate tumor progression. J Recept Signal

Transduct Res 2003;23:225–238.

[27] Stoebner PE, Carayon P, Casellas P, Portier M, Lavabre-Bertrand T,

Cuq P, et al. Transient protection by peripheral benzodiazepine

receptors during the early events of ultraviolet light-induced

apoptosis. Cell Death Differ 2001;8:747–753.

[28] Amri H, Li H, Culty M, Gaillard JL, Teper G, Papadopoulos V. The

peripheral-type benzodiazepine receptor and adrenal steroidogenesis.

Curr Opin Endocrinol Diabetes 1999;6:179–184.

[29] Wang JK, Morgan JI, Spector S. Benzodiazepines that bind at

peripheral sites inhibit cell proliferation. Proc Natl Acad Sci USA

1984;81:753–756.

[30] Fischer R, Schmitt M, Bode JG, Haussinger D. Expression of the

peripheral-type benzodiazepine receptor and apoptosis induction in

hepatic stellate cells. Gastroenterology 2001;120:1212–1226.

[31] Pastorino JG, Simbula G, Gilfor E, Hoek JB, Farber JL. Proto-

porphyrin IX, an endogenous ligand of the peripheral benzodiazepine

receptor, potentiates induction of the mitochondrial permeability

transition and the killing of cultured hepatocytes by rotenone. J Biol

Chem 1994;269:31041–31046.

[32] Xia W, Spector S, Hardy L, Zhao S, Saluk A, Alemane L, et al. Tumor

selective G2/M cell cycle arrest and apoptosis of epithelial and

hematological malignancies by BBL22, a benzazepine. Proc Natl

Acad Sci USA 2000;97:7494–7499.

[33] Takahashi M, Saito H, Okuyama T, Miyashita T, Kosuga M,

Sumisa F, et al. Overexpression of Bcl-2 protects human hepatoma

cells from Fas-antibody-mediated apoptosis. J Hepatol 1999;31:

315–322.

[34] Marzo I, Brenner C, Zamzami N, Susin SA, Beutner G, Brdiczka D,

et al. The permeability transition pore complex: a target for apoptosis

regulation by caspases and bcl-2-related proteins. J Exp Med 1998;

187:1261–1271.

[35] Zorov DB. Mitochondrial damage as a source of diseases and aging: a

strategy of how to fight these. Biochim Biophys Acta 1996;1275:

10–15.

[36] Zamzami N, Susin SA, Marchetti P, Hirsch T, Gomez-Monterrey I,

Castedo M, et al. Mitochondrial control of nuclear apoptosis. J Exp

Med 1996;183:1533–1544.

[37] Narita M, Shimizu S, Ito T, Chittenden T, Lutz RJ, Matsuda H, et al.

Bax interacts with the permeability transition pore to induce

permeability transition and cytochrome c release in isolated

mitochondria. Proc Natl Acad Sci USA 1998;95:14681–14686.

[38] Marzo I, Brenner C, Zamzami N, Susin SA, Beutner G, Brdiczka D,

et al. The permeability transition pore complex: a target for apoptosis

regulation by caspases and bcl-2-related proteins. J Exp Med 1998;

187:1261–1271.

[39] Papadopoulos V, Dharmarajan AM, Li H, Culty M, Lemay M,

Sridaran R. Mitochondrial peripheral-type benzodiazepine receptor

expression. Correlation with gonadotropin-releasing hormone

(GnRH) agonist-induced apoptosis in the corpus luteum. Biochem

Pharmacol 1999;58:1389–1393.

[40] Hirsch T, Decaudin D, Susin SA, Marchetti P, Larochette N,

Resche-Rigon M, et al. PK11195, a ligand of the mitochondrial

benzodiazepine receptor, facilitates the induction of apoptosis and

reverses Bcl-2-mediated cytoprotection. Exp Cell Res 1998;241:

426–434.

[41] Ravagnan L, Marzo I, Costantini P, Susin SA, Zamzami N, Petit PX,

et al. Lonidamine triggers apoptosis via a direct, Bcl-2-inhibited effect

on the mitochondrial permeability transition pore. Oncogene 1999;18:

2537–2546.

[42] Wang JL, Liu D, Zhang ZJ, Shan S, Han X, Srinivasula SM, et al.

Structure-based discovery of an organic compound that binds Bcl-2

protein and induces apoptosis of tumor cells. Proc Natl Acad Sci USA

2000;97:7124–7129.

[43] Berson A, Descatoire V, Sutton A, Fau D, Maulny B, Vadrot N, et al.

Toxicity of alpidem, a peripheral benzodiazepine receptor ligand, but

not zolpidem, in rat hepatocytes: role of mitochondrial permeability

transition and metabolic activation. J Pharmacol Exp Ther 2001;299:

793–800.

[44] Wendler G, Lindemann P, Lacapere JJ, Papadopoulos V. Proto-

porphyrin IX, binding and transport by recombinant mouse PBR.

Biochem Biophys Res Commun 2003;311:847–852.

[45] Walter RB, Raden BW, Cronk MR, Bernstein ID, Appelbaum FR,

Banker DE. The peripheral benzodiazepine receptor ligand PK11195

overcomes different resistance mechanisms to sensitize AML cells to

gemtuzumab ozogamicin. Blood 2004;103:4276–4284.