Rosiglitazone impairs proliferation of human adrenocortical cancer: preclinical study in a xenograft...

9
AUTHOR COPY ONLY Rosiglitazone impairs proliferation of human adrenocortical cancer: preclinical study in a xenograft mouse model Michaela Luconi 1 , Monica Mangoni 1 , Stefania Gelmini 1 , Giada Poli 1 , Gabriella Nesi 2 , Michela Francalanci 1 , Nicola Pratesi 1 , Giulia Cantini 1 , Adriana Lombardi 1 , Monica Pepi 2 , Tonino Ercolino 1 , Mario Serio 1 , Claudio Orlando 1 and Massimo Mannelli 1 Departments of 1 Clinical Physiopathology and 2 Human Pathology and Oncology, DENOthe Center of Excellence for Research, Transfer and High Education, University of Florence, Viale Pieraccini 6, Florence 50139, Italy (Correspondence should be addressed to M Luconi; Email: [email protected]fi.it) Abstract Adrenocortical carcinoma (ACC) is a rare aggressive tumor with a poor prognosis. The lack of a specific and effective medical treatment is due to the poor knowledge of the mechanisms underlying tumor growth. Research on potential drugs able to specifically interfere with tumor proliferation is essential to develop more efficacious therapies. We evaluated for the first time the in vivo effect of rosiglitazone (RGZ), an anti-diabetic drug with in vitro anti-tumor properties, on ACC proliferation in a xenograft model obtained by s.c. injection of human ACC H295R cells in athymic mice. When the tumor size reached 5 mm, animals were allocated to 5 mg/kg RGZ- or water-treated groups. Tumor volume was measured twice a week. A significant reduction of tumor growth in RGZ versus control (control) group was observed and was already maximal following 17 day treatment (1KT/CZ75.4% (43.7–93.8%)). After 31 days of treatment, mice were killed and tumor analyzed. Tumor histological evaluation revealed characteristics of invasiveness, richness in small vessels and mitotic figures in control group, while RGZ group tumors presented non infiltrating borders, few vessels, and many apoptotic bodies. Tumor immunohistochemistry showed that Ki-67 was reduced in RGZ versus control group. Quantitative real-time RT-PCR demonstrated a significant reduction in the expression of angiogenic (VEGF), vascular (CD31), proliferation (BMI-1), and anti-apoptotic (Bcl-2) genes in RGZ versus control group tumors. The same inhibitory effects were confirmed in in vitro RGZ-treated H295R. Our findings support and expand the role of RGZ in controlling ACC proliferation and angiogenesis in vivo and in vitro. Endocrine-Related Cancer (2010) 17 169–177 Introduction Adrenocortical carcinoma (ACC) is a rare (1:4!10 6 annual estimated incidence) and aggressive endocrine tumor with a poor prognosis, generally characterized by a limited response to radio/chemotherapy (Allolio & Fassnacht 2006). At present, early diagnosis followed by total surgical tumor resection is the only valuable option for ACC cure. Prognosis depends on the tumor stage at surgery – mean survival rate at 5 years is 16–38% (Allolio & Fassnacht 2006); however, metastatic disease reduces survival to !10% (Fassnacht et al. 2009). The lack of a specific and effective medical regimen is due to the poor knowledge of the mechanisms underlying malignant tumor transformation and progression. Mitotane is the only pharmacological adjuvant treatment available for ACC in advanced disease (Luton et al. 1990, Terzolo et al. 2007), and its combination with other chemotherapic drugs has led to variable results (Khan et al. 2000, Berruti et al. 2005). However, its effects are mainly due to its adrenocortical cytotoxicity. Thus, the development of new efficacious and less toxic drugs, specific for ACC, to be eventually combined with mitotane (Barlaskar et al. 2009) are required. The peroxisome proliferator-activated receptor (PPAR)-g is a ligand-activated transcription factor, Endocrine-Related Cancer (2010) 17 169–177 Endocrine-Related Cancer (2010) 17 169–177 1351–0088/10/017–169 q 2010 Society for Endocrinology Printed in Great Britain DOI: 10.1677/ERC-09-0170 Online version via http://www.endocrinology-journals.org

Transcript of Rosiglitazone impairs proliferation of human adrenocortical cancer: preclinical study in a xenograft...

AUTHOR COPY ONLYEndocrine-Related Cancer (2010) 17 169–177

Rosiglitazone impairs proliferation ofhuman adrenocortical cancer: preclinicalstudy in a xenograft mouse model

Michaela Luconi1, Monica Mangoni1, Stefania Gelmini1, Giada Poli1,Gabriella Nesi 2, Michela Francalanci1, Nicola Pratesi1, Giulia Cantini1,Adriana Lombardi1, Monica Pepi 2, Tonino Ercolino1, Mario Serio1,Claudio Orlando1 and Massimo Mannelli1

Departments of 1Clinical Physiopathology and 2Human Pathology and Oncology, DENOthe Center of Excellence for Research,

Transfer and High Education, University of Florence, Viale Pieraccini 6, Florence 50139, Italy

(Correspondence should be addressed to M Luconi; Email: [email protected])

Abstract

Adrenocortical carcinoma (ACC) is a rare aggressive tumor with a poor prognosis. The lack of aspecific and effective medical treatment is due to the poor knowledge of the mechanismsunderlying tumor growth. Research on potential drugs able to specifically interfere with tumorproliferation is essential to develop more efficacious therapies. We evaluated for the first time thein vivo effect of rosiglitazone (RGZ), an anti-diabetic drug with in vitro anti-tumor properties, onACC proliferation in a xenograft model obtained by s.c. injection of human ACC H295R cells inathymic mice. When the tumor size reached 5 mm, animals were allocated to 5 mg/kg RGZ- orwater-treated groups. Tumor volume was measured twice a week. A significant reduction of tumorgrowth in RGZ versus control (control) group was observed and was already maximal following17 day treatment (1KT/CZ75.4% (43.7–93.8%)). After 31 days of treatment, mice were killed andtumor analyzed. Tumor histological evaluation revealed characteristics of invasiveness, richnessin small vessels and mitotic figures in control group, while RGZ group tumors presented noninfiltrating borders, few vessels, and many apoptotic bodies. Tumor immunohistochemistryshowed that Ki-67 was reduced in RGZ versus control group. Quantitative real-time RT-PCRdemonstrated a significant reduction in the expression of angiogenic (VEGF), vascular (CD31),proliferation (BMI-1), and anti-apoptotic (Bcl-2) genes in RGZ versus control group tumors. Thesame inhibitory effects were confirmed in in vitro RGZ-treated H295R. Our findings support andexpand the role of RGZ in controlling ACC proliferation and angiogenesis in vivo and in vitro.

Endocrine-Related Cancer (2010) 17 169–177

Introduction

Adrenocortical carcinoma (ACC) is a rare (1:4!106

annual estimated incidence) and aggressive endocrine

tumor with a poor prognosis, generally characterized

by a limited response to radio/chemotherapy (Allolio

& Fassnacht 2006). At present, early diagnosis

followed by total surgical tumor resection is the only

valuable option for ACC cure. Prognosis depends

on the tumor stage at surgery – mean survival rate at

5 years is 16–38% (Allolio & Fassnacht 2006);

however, metastatic disease reduces survival to

!10% (Fassnacht et al. 2009). The lack of a specific

and effective medical regimen is due to the poor

Endocrine-Related Cancer (2010) 17 169–177

1351–0088/10/017–169 q 2010 Society for Endocrinology Printed in Great

knowledge of the mechanisms underlying malignant

tumor transformation and progression. Mitotane is

the only pharmacological adjuvant treatment available

for ACC in advanced disease (Luton et al. 1990,

Terzolo et al. 2007), and its combination with other

chemotherapic drugs has led to variable results (Khan

et al. 2000, Berruti et al. 2005). However, its effects

are mainly due to its adrenocortical cytotoxicity. Thus,

the development of new efficacious and less toxic

drugs, specific for ACC, to be eventually combined

with mitotane (Barlaskar et al. 2009) are required.

The peroxisome proliferator-activated receptor

(PPAR)-g is a ligand-activated transcription factor,

Britain

DOI: 10.1677/ERC-09-0170

Online version via http://www.endocrinology-journals.org

AUTHOR COPY ONLYM Luconi et al.: Rosiglitazone anti-proliferative effects in ACC

its thiazolidinedione (TZD) ligands, used in type 2

diabetes therapy, have been shown to exert additional

anticancer effects in several solid tumors (Panigrahy

et al. 2005). Although the antitumor action of TZDs

(due to their anti-proliferative, anti-angiogenic, and

anti-inflammatory effects) have been well documented

in in vitro and animal studies, the results which have

emerged from the few clinical trials are still quite

controversial (Burton et al. 2008). Very few studies have

been conducted on TZD effects in adrenal cancer. In

particular, rosiglitazone (RGZ), the TZD with the highest

affinity for the receptor, has been demonstrated to

interfere with cancer growth by blocking cell prolifer-

ation/migration and to induce cell differentiation/

apoptosis in the ACC cell line H295R (Betz

et al. 2005, Ferruzzi et al. 2005, Cantini et al. 2008).

In this study, we investigated the effects of RGZ

in a human ACC xenograft model as obtained by s.c.

H295R injection into athymic mice. In particular, we

evaluated RGZ inhibition on tumor growth in vivo and

evidenced some specific target genes involved in the

interfering effects of RGZ both in vivo and in vitro.

Materials and methods

Materials

Media and sera for cell cultures were from Lonza (Milan,

Italy) and tissue plasticware from Corning (Milan, Italy).

Other reagents were obtained from Sigma–Aldrich,

except where differently indicated. RGZ was obtained

from Alexis Biochemicals (San Diego, CA, USA).

Cell cultures

The human ACC cell line H295R was obtained from

the American Type Culture Collection (Manassas, VA,

USA). H295R cells were cultured in 10% fetal bovine

serum (FBS) (Euroclone, Celbio, Milan, Italy), 2 mM

L-glutamine, 100 U/ml penicillin, and 100 mg/ml

streptomycin, DMEM/F-12 medium enriched with a

mixture of insulin/transferrin/selenium (Sigma–

Aldrich) at 37 8C in a humidified 5% CO2 atmosphere.

Subconfluent cells were treated for 24 h with 20 mM

RGZ, following overnight starvation.

Xenograft model

A total of 33 female athymic CD1 nude mice

(6–8-week-old, Charles River Laboratories, Italy)

were inoculated subcutaneously with H295R cell

suspension (7!106 cells/100 ml) in two independently

performed experiments. Data obtained in the two

experiments were pooled. Subcutaneous tumor growth

170

was daily monitored, and when solid tumors reached a

5 mm mean diameter (9 days after H295R inoculation),

the animals were assigned to be orally treated with RGZ

(5 mg/kg in 100 ml, 6 days/week) or vehicle, according

with randomization decided before s.c. injection of

H295R. Tumor volume (mm3) was the mean of values

estimated by two independent investigators from two

dimensional tumor measurements by the formula:

length!width2/2. After 31 days of oral treatment

(in accordance with RGZ treatment interval of 4 week

already described in literature (Heaney et al. 2002,

Dai et al. 2008)), mice were killed by cervical

dislocation, and tumors were collected and split for

mRNA or histological/immunohistochemical analyses.

Drug anti-tumor activity was evaluated as the

tumor growth inhibition percentage calculated by

the 1Ktreated/control tumor volume ratios (1KT/C)

(Teicher 1998, Johnson et al. 2001, Hollingshead

2008). Data were expressed as the median with the

minimum–maximum value interval.

Drug tolerability was assessed in tumor-bearing

mice in terms of: a) lethal toxicity, i.e. any death

in treated mice occurring before any death in

control mice; b) body weight loss percentage

Z100K(body weight on day x/body weight on day

1!100), where x represents a day after or during the

treatment period (Teicher 1998, Johnson et al. 2001,

Hollingshead 2008).

Animal studies were performed in compliance with

an institutionally approved protocol and with the

National Institutes of Health Guide for the Care and

Use of Laboratory Animals.

Tumor measurements, as well as tumor histologic/

immunohistochemical and gene expression analyses,

were performed by independent investigators working

in blind conditions.

Histologic and immunohistochemical

examination of xenografts

Tissues were fixed in 10% buffered formalin and

paraffin-embedded. Five-micrometer sections were

hematoxylin and eosin (H/E) stained for histologic

evaluation, or used for immunohistochemistry. Apop-

totic body (ABI) and mitotic (MI) indices were

obtained by counting 1000 cells in H/E-stained

sections. Apoptotic bodies were recognized as shrunk

cells with compact or segregated, sharply delineated

masses of chromatin in nodular, ring-like or bead-like

patterns. If present, the cytoplasm was deeply

eosinophilic. As a result of cellular shrinkage,

apoptotic cells were frequently surrounded by a clear

halo. Apoptotic bodies also consisted of dense

www.endocrinology-journals.org

AUTHOR COPY ONLYEndocrine-Related Cancer (2010) 17 169–177

extracellular or intracellular chromatin fragments, with

or without associated cytoplasm. Mitotic figures were

identified by morphologic features of metaphase,

anaphase, or telophase.

Immunohistochemical analysis with mouse anti-

human Ki-67 monoclonal MIB1 antibody (Dako,

Glostrup, Denmark) was performed with the Ventana

Benchmark XT system (Ventana Medical Systems,

Tucson, AZ, USA). Nuclei were hematoxylin-

counterstained. Ki-67 positive nuclei were counted

on 1000 tumor cells. Negative controls were performed

by omitting the primary antibody.

Figure 1 RGZ reduces tumor volume in ACC xenografts.Growth curves of xenograft ACC. Values represent meanGS.E.M. of measured tumor volume over time in control group(filled squares, nZ16) and in RGZ-treated group (filled circles,nZ17). Data have been pooled from mice treated in twoindependent experiments. P!0.001 between control and RGZgroups for all points starting from day 3. *P!0.005 and**P!0.001 versus day 0.

RNA isolation and quantitative real time RT-PCR

Xenograft tumors, snap frozen in liquid nitrogen, and

pelleted H295R cells were processed for RNA

extraction. Total mRNA isolation and quantitative

real time RT-PCR were performed as detailed else-

where (Lombardi et al. 2008) using specific primers/

probes (Assay on Demand; Applied Biosystems,

Warrington, UK). The amount of target mRNA was

given by 2KDDCt calculation, following normalization to

an endogenous reference gene (18S, conserved in the

mouse and humans, for xenograft tumor analysis;

glyceraldehyde-3-phosphate dehydrogenase (GAPDH)

for H295R analysis) and relative to a calibrator

(Stratagene, La Jolla, CA, USA).

Statistical analysis

The statistical analysis was performed using SPSS

12.0 (SPSS 12.0; SPSS Inc). One-way ANOVA

followed by Dunnett’s post hoc test was applied for

multiple comparisons, whereas Student’s t-test was

used for comparisons of two classes of data. A P value

!0.05 was considered significant.

Results

RGZ treatment interferes with tumor growth

in the xenograft model of ACC

All H295R injected mice (nZ33 from two independent

experiments) developed a detectable tumor (100%

overall tumor take rate), confirming the aggressiveness

of ACC-derived cells. Tumors for each group of

treatment, control and RGZ, were followed and

measured during the 31 days of treatment. Tumor

volume was calculated for each mouse at each time

point. Figure 1 shows the tumor growth rate in the two

treatment groups. Data were plotted as mean tumor

volume (Fig. 1) in each group over time. RGZ

administration resulted in a reduced increase in tumor

www.endocrinology-journals.org

volume, which was already statistically significant

after 3 days of treatment (Fig. 1). After 3 days of

treatment, RGZ inhibitory effect was already relevant

(1KT/CZ45.3% (15.5–74.4%)), reached a maximum

after 17 days (1KT/CZ75.4% (43.7–93.8%)) and

remained constant up to the end of the treatment (1KT/

CZ74.5% (25.3–94.1%)). Interestingly, the tumor

volume increased compared to the initial volume at

any time point in the control animals, and such an

increase became statistically significant starting from

day 17. Conversely, in the RGZ-treated mice, there

was an initial reduction in the tumor size, followed by a

slow increase in the long term, which was statistically

significant only from day 27 (Fig. 1). Moreover, the

differences in the tumor volume increase versus day 0

were statistically significant at any time point between

control- and RGZ-treated groups. The drug was well

tolerated without lethal toxicity and body weight loss

during treatment (data not shown).

The xenograft tumors obtained by s.c. injection

of H295R (Fig. 2) were histologically similar to the

adrenocortical tumor from which H295R cells were

derived confirming previous observations (Logie

et al. 2000).

In the control group, the tumors displayed a solid,

diffuse architecture (Fig. 2A) and consisted of rather

small, uniform cells with coarse chromatin and

prominent nucleoli. A mixture of large caliber vessels

and a disorganized network of small-caliber vessels

were seen (Fig. 2C). A remarkable number of mitotic

171

AUTHOR COPY ONLY

Figure 2 RGZ effects on ACC xenograft cyto-architecture. Hematoxylin/eosin staining of tumors excised from mice treated for31 days with vehicle (A, C, and E: control group) or with 5 mg/kg RGZ (B, D, and F: RGZ group). Magnifications: A and B (10!); Cand D (20!); E and F (40!). Arrows and arrowheads indicate mitotic and apoptotic figures respectively.

M Luconi et al.: Rosiglitazone anti-proliferative effects in ACC

figures were present, often with atypical forms

(Fig. 2E). Conversely, in the RGZ group, the tumors

showed well-demarcated borders (Fig. 2B), a lower

degree of vascularization (Fig. 2D), a greater number

of apoptotic bodies and a lower MI (Fig. 2F). Table 1

shows the mean percentage of MI and ABI detected in

H/E-stained tumor sections in control and RGZ groups.

RGZ-treated group showed a statistically significant

decrease in the percentage of mitosis (P!0.05, nZ10

animals for each group of treatment obtained in two

independent experiments) and a statistically significant

increase in the percentage of apoptotic figures

(P!0.05, nZ10 animals for each group of treatment

obtained in two independent experiments) as compared

to control group.

Immunohistochemical analysis of tumor sections

from xenografts revealed that RGZ treatment was

associated with a reduction in Ki-67 staining in cell

172

nuclei (Fig. 3B) as compared with control (Fig. 3A),

suggesting an inhibitory effect of RGZ on tumor

proliferation. RGZ-treated group showed a statisti-

cally significant decrease in the percentage of Ki-67

labeling index as compared with control group

(P!0.001, nZ10 animals for each group of treatment

obtained in two independent experiments), Table 1.

Molecular analysis of the xenograft tumors was

performed on total mRNA extracted from control

and RGZ-treated animals.

Similarly to data obtained in H295R cells, PPARgwas expressed in all xenografts and not affected by

RGZ treatment, as detected by quantitative real-time

RT-PCR (PPARg versus 18S expressionGS.E.M. in

control: 3.4G1.8 and RGZ: 3.1G1.3 groups, nZ8).

Tumor cell proliferation was reduced by 31 days

RGZ treatment as demonstrated by the statistically

significant decrease in the proliferation marker

www.endocrinology-journals.org

AUTHOR COPY ONLYTable 1 Rosiglitazone (RGZ) stimulates apoptosis and

interferes with proliferation in xenograft adrenocortical carci-

noma (ACC). Apoptotic body (ABI) andmitotic (MI) indices were

expressed as mean percentageGS.E.M. of positive cells in

hematoxylin and eosin-stained tumor sections from control and

RGZ groups. The Ki-67 labeling index was expressed as the

meanGS.E.M. percentage of Ki-67 positive nuclei in tumor

sections from control and RGZ groups, subjected to immuno-

histochemical analysis with anti-Ki-67 antibody

ABI MI Ki-67 labeling

Control 4.1G0.4 1.7G0.8 66.4G2.4

RGZ 6.0G0.7* 1.2G0.6* 50.3G1.4†

*P!0.05 and †P!0.001 versus control, nZ10 mice in eachgroup from two independent experiments. RGZ treatmentresulted in a 46% increase in ABI as well as in a 30 and 25%decrease inMI andKi-67 respectively compared to control group.

Endocrine-Related Cancer (2010) 17 169–177

BMI-1 (Fig. 4A). RGZ not only affected tumor

proliferation but also the angiogenic process support-

ing tumorigenesis. In fact, both the angiogenic factor

VEGF and the endothelial marker CD31 were highly

expressed in the untreated control tumors, while their

expression was significantly reduced in tumors from

RGZ-treated group (Fig. 4A). Confirming the histo-

logical features, TaqMan analysis demonstrated a

significant reduction in the expression of the anti-

apoptotic marker Bcl-2 in the RGZ-treated versus the

untreated control group (Fig. 4A). Finally, in tumors

from both groups we measured expression of seladin-1/

DHCR24, the gene encoding 3-b-hydroxysterol

D-24-reductase (DHCR24), which converts desmos-

terol into cholesterol (Greeve et al. 2000). In fact,

seladin-1 mRNA levels have been demonstrated to be

significantly lower in adrenocortical cancer and in the

derived primary cell cultures compared to adenoma

Figure 3 RGZ interferes with cell proliferation in xenografttumor. Ki-67 immunohistochemistry in tumor sections frommicetreated for 31 days with vehicle (A, control group) or with5mg/kg RGZ (B, RGZ group). Magnifications: A and B (40!).

www.endocrinology-journals.org

and normal adrenal and their derived primary cell

cultures (Luciani et al. 2004). At variance with the

decrease found in all the above described genes,

seladin-1 mRNA levels were slightly, although not

significantly, up-regulated in the RGZ-treated group

(Fig. 4A).

RGZ effects on expression of adrenal

tumorigenesis genes in in vitro H295R cultures

confirm results obtained in the mouse

xenograft model

To confirm that the effects observed on tumor growth

following mouse treatment with RGZ were also due to

the direct effect exerted by RGZ on H295R and not

only on the mouse tissue tumor micro-environment, we

treated H295R with RGZ in vitro and analyzed the

expression of the adrenal tumorigenesis genes

Figure 4 RGZ modulation of gene expression in xenografttumor and H295R. Gene expression was evaluated byquantitative real-time RT-PCR in tissue extracts of tumors frommice treated for 31 day treatment with vehicle (control group) orwith 5 mg/kg RGZ (RGZ group) (A, nZ13) or in H295R cellsin vitro-treated with vehicle (control group) or with 20 mM RGZ(RGZ group) for 24 h (B, nZ4). Data represent meanGS.E.M.fold change of gene expression in RGZ versus control taken as 1(dot line). The percentage of decrease (K) or increase (C) ingene expression following RGZ treatment is indicated.*P!0.05, **P!0.01, 8P!0.005, #P!0.001 versus respectivecontrol.

173

AUTHOR COPY ONLYM Luconi et al.: Rosiglitazone anti-proliferative effects in ACC

previously evaluated on xenograft tumor samples.

H295R cells were treated with 20 mM RGZ for 24 h,

and total mRNA was extracted and subjected to

quantitative real-time RT-PCR analysis with the

same primers used above. This RGZ concentration

has already been used for in vitro experiments in H295

and H295R cells (Betz et al. 2005, Ferruzzi et al. 2005)

and has been calculated to be the IC50 for RGZ

inhibition of H295R cell proliferation (Cantini et al.

2008). Expression of the proliferation, angiogenic and

anti-apoptotic markers, BMI-1, VEGF and Bcl-2

respectively, was significantly downregulated, while

expression of seladin-1 was significantly increased by

RGZ cell treatment (Fig. 4B), confirming the results

obtained in the xenograft model.

Discussion

This paper reports, for the first time, the inhibitory

effect exerted in vivo by RGZ on adrenocortical

cancer growth in a xenograft model of human ACC

obtained by s.c. injection of H295R in athymic CD1

mice. In particular, we demonstrate that 4 week oral

administration of 5 mg/kg per day RGZ signi-

ficantly inhibits tumor growth (1KT/CZ74.5%) and

vascularization by reducing cell proliferation (signi-

ficant reduction of MI and Ki-67 staining in RGZ

versus control group) and stimulating apoptosis

(significant increase in ABI in RGZ versus control

group). These histological findings have been

confirmed at molecular levels by the ability of RGZ

to reduce the expression of angiogenic/vascular

markers such as VEGF and CD31, of proliferation

markers such as BMI-1 and of the anti-apoptotic

marker, Bcl-2. The in vivo effects of RGZ in mouse

xenografts have been confirmed by analogous findings

obtained in vitro in RGZ-treated H295R.

Previous studies demonstrated that TZD inhibits

in vitro proliferation of the human ACC cell model,

H295R. In particular, RGZ reduces H295R cell growth

in vitro with a calculated IC50 of 20 mM (Cantini et al.

2008), by interfering with the insulin-like growth

factor receptor -1 axis and inducing cell differentiation

and apoptosis (Betz et al. 2005, Ferruzzi et al. 2005,

Cantini et al. 2008). RGZ anti-neoplastic effects have

been observed in several types of cancer cells and in

xenograft tumors (Keshamouni et al. 2004, Yoshizumi

et al. 2004, Annicotte et al. 2006, Dai et al. 2008),

although it is not yet clear whether RGZ action is

mediated by PPARg. Of interest, we found no

alterations in PPARg expression in xenograft tumors

following RGZ treatment.

174

Our results indicate that RGZ anti-proliferative

effects involve a downregulation of angiogenic and

an upregulation of apoptotic genes both in vivo and

in vitro, resulting in a significant reduction in

vascularization and proliferation and an increase in

apoptosis in RGZ-treated xenograft tumors. Gene

expression inhibition by RGZ, as fold increase over

controls, seems more evident in xenograft tumors than

in in vitro treated cells, suggesting that RGZ effect is

not only acting on H295R but also on the murine

vascular component of the tumor. Indeed, all primers

used, except for CD31 which is mouse-specific, can

detect the expression of both human and mouse

genes. Interestingly, RGZ inhibitory effects on gene

expression are specific, since expression of seladin-1 is

not inhibited but is even stimulated by RGZ treatment,

in particular in in vitro experiments. Since seladin-1 is

involved in the synthesis of cholesterol, the precursor

of steroid hormones produced by the adrenal cortex,

and since it is expressed at lower levels in malignant

ACC than in normal adrenal gland (Luciani et al.

2004), it may be hypothesized that its increased

expression in xenograft tumor and in H295R following

RGZ, possibly associates with induced differentiation

and reduced malignancy. The reduced expression of

the polycomb gene Bmi-1 in RGZ-treated xenograft

ACC not only confirms inhibition of proliferation, but

also suggests that RGZ interferes with malignancy.

In fact, in both lymphomas (van Kemenade et al.

2001) and breast cancer (Datta et al. 2007), BMI-1

emerged to be associated not only with proliferation

markers such as Ki-67, but also with the degree of

malignancy, thus representing a possible prognostic

factor (Silva et al. 2007). This is the first time that

BMI-1 expression has been studied in ACC. Further

investigations are required to define the correlation

between BMI-1 expression and ACC differentiation

and malignancy.

Although in preclinical studies TZD showed anti-

proliferative, pro-apoptotic, and differentiating effects,

their use in clinical trials planned for the treatment of

limited cohorts of patients affected by different types of

solid tumors gave controversial and not encouraging

results (Demetri et al. 1999, Debrock et al. 2003, Vogt

et al. 2003, Reichle et al. 2004, Smith et al. 2004, Yee

et al. 2007, Read et al. 2008). Indeed, only a few

studies found TZD to exert some positive therapeutic

effect, particularly if combined with chemotherapic

and angiostatic drugs (Vogt et al. 2003, Reichle et al.

2004). This is of particular notice, since RGZ has been

demonstrated to inhibit primary tumor growth and

metastasis in different xenograft tumors through

suppression of angiogenesis (Panigrahy et al. 2002).

www.endocrinology-journals.org

AUTHOR COPY ONLYEndocrine-Related Cancer (2010) 17 169–177

Also in our ACC model, RGZ, at even lower doses,

reduced the vessel number as well as the expression of

endothelial and angiogenic markers. However, all the

clinical studies so far reported have enrolled a limited

number of patients and are too heterogeneous in terms

of type and stage of tumors, patient’s age and sex and

type of treatment (TZD doses, combined therapy,

duration) to allow a solid analysis of TZD anticancer

effects. Interestingly, Monami’s meta-analysis

(Monami et al. 2008) found a significantly lower

incidence of malignancies in RGZ-treated diabetic

patients, suggesting a protective effect of RGZ

versus specific cancers, such as gastro-intestinal and

lung cancer.

The different RGZ effects observed in clinical and

pre-clinical studies may be due to several factors, such

as the duration of treatment and the doses of RGZ. The

8 mg/day oral administration of RGZ to diabetic

patients, resulting in 1.3 mM RGZ circulating level, is

the maximal dose reached in cancer studies and is far

lower than the doses exerting anti-neoplastic effect

in vitro (20–50 mM) and in xenograft tumors

(50–150 mg/kg per day, Heaney et al. 2002). Thus, in

an attempt to evaluate its true potential anticancer

effects, it might be useful to increase RGZ doses to

those which have been found efficacious in preclinical

studies. Concerns about the potential cardiovascular

risks (Nissen & Wolski 2007) of using RGZ at doses

higher than 8 mg/day should be inconsistent since, at

variance with diabetic subjects, ACC patients seldom

suffer from cardiovascular or renal complications.

Moreover, an overall well-tolerated RGZ adminis-

tration was reported in patients with Cushing’s disease

(Pecori Giraldi et al. 2006) or solid tumors (Read et al.

2008) at doses (12 and 16 mg/die for up to 8 months)

higher than those used in diabetes treatment. Of note,

we obtained a 70% inhibition of xenograft tumor

growth with a RGZ dose 30 time lower than those

currently used in xenograft studies (Heaney et al.

2002), without observing body weight loss or toxicity

during the treatment. This result is notable, since a

tumor inhibition percentage (1KT/C) R60% has been

defined for drug anticancer efficacy (Teicher 1998).

However, pharmacokinetic studies in the animals

should be set up in order to evaluate which are the

RGZ serum levels achieved in orally treated xenograft

mice, which result in tumor growth inhibition in vivo.

Before hypothesizing a possible therapeutic use of

RGZ and other TZD in ACC treatment, pharmacoki-

netic studies are mandatory to establish whether oral

doses capable of resulting in around 20 mM circulating

concentrations possess toxic effects in vivo in humans.

www.endocrinology-journals.org

In conclusion, this is the first study demonstrating

that RGZ effectively inhibits tumor growth in a human

xenograft ACC model. These findings suggest a

potential role for RGZ as a novel and promising

adjuvant therapy after surgical removal of ACC, alone

or in combination with mitotane to obtain more

efficacious antitumor results. Further studies are

needed to test the effect of a combined treatment of

RGZ and mitotane in vitro and in vivo.

Declaration of interest

The authors declare that there is no conflict of interest that

would prejudice the impartiality of this scientific work.

Funding

This work was supported by Tuscany REgional Study Of

Rosiglitazone (TRESOR), Istituto Toscano Tumori (ITT),

and Cassa di Risparmio di Firenze (CRF).

Acknowledgements

We thank Prof. Andrea Galli (University of Florence) for his

helpful suggestions.

References

Allolio B & Fassnacht M 2006 Clinical review: adreno-

cortical carcinoma: clinical update. Journal of Clinical

Endocrinology and Metabolism 91 2027–2037.

Annicotte JS, Iankova I, Miard S, Fritz V, Sarruf D, Abella A,

Berthe ML, Noel D, Pillon A, Iborra F et al. 2006

Peroxisome proliferator-activated receptor gamma

regulates E-cadherin expression and inhibits growth

and invasion of prostate cancer. Molecular and Cellular

Biology 26 7561–7574.

Barlaskar FM, Spalding AC, Heaton JH, Kuick R, Kim AC,

Thomas DG, Giordano TJ, Ben-Josef E & Hammer GD

2009 Preclinical targeting of the type I insulin-like

growth factor receptor in adrenocortical carcinoma.

Journal of Clinical Endocrinology and Metabolism

94 204–212.

Berruti A, Terzolo M, Sperone P, Pia A, Casa SD, Gross DJ,

Carnaghi C, Casali P, Porpiglia F, Mantero F et al. 2005

Etoposide, doxorubicin and cisplatin plus mitotane in the

treatment of advanced adrenocortical carcinoma: a large

prospective phase II trial. Endocrine-Related Cancer

12 657–666.

Betz MJ, Shapiro I, Fassnacht M, Hahner S, Reincke M,

Beuschlein F & for the German and Austrian Adrenal

Network 2005 Peroxisome proliferator-activated

receptor-gamma agonists suppress adrenocortical

tumor cell proliferation and induce differentiation.

Journal of Clinical Endocrinology and Metabolism

90 3886–3896.

175

AUTHOR COPY ONLYM Luconi et al.: Rosiglitazone anti-proliferative effects in ACC

Burton JD, Goldenberg DM & Blumenthal RD 2008

Potential of peroxisome proliferator-activated receptor

gamma antagonist compounds as therapeutic agents for

a wide range of cancer types. PPAR Research 2008

494161.

Cantini G, Lombardi A, Piscitelli E, Poli G, Ceni E,

Marchiani S, Ercolino T, Galli A, Serio M, Mannelli M

et al. 2008 Rosiglitazone inhibits adrenocortical cancer

cell proliferation by interfering with the IGF-IR intra-

cellular signaling. PPAR Research 2008 904041.

Dai Y, Qiao L, Chan KW, Zou B, Ma J, Lan HY, Gu Q, Li Z,

Wang Y, Wong BL et al. 2008 Loss of XIAP sensitizes

rosiglitazone-induced growth inhibition of colon

cancer in vivo. International Journal of Cancer 122

2858–2863.

Datta S, Hoenerhoff MJ, Bommi P, Sainger R, Guo WJ,

Dimri M, Band H, Band V, Green JE & Dimri GP 2007

Bmi-1 cooperates with H-Ras to transform human

mammari epithelial cells via dysregulation of multiple

growth-regulatory pathways. Cancer Research 67

10286–10295.

Debrock G, Vanhentenrijk V, Sciot R, Debiec-Rychter M,

Oyen R & Van Oosterom A 2003 A phase II trial with

rosiglitazone in liposarcoma patients. British Journal of

Cancer 89 1409–1412.

Demetri GD, Fletcher CD, Mueller E, Sarraf P, Naujoks R,

Campbell N, Spiegelman BM & Singer S 1999 Induction

of solid tumor differentiation by the peroxisome pro-

liferator-activated receptor-gamma ligand troglitazone in

patients with liposarcoma. PNAS 96 3951–3956.

Fassnacht M, Johanssen S, Quinkler M, Bucsky P,

Willenberg HS, Beuschlein F, Terzolo M, Mueller HH,

Hahner S & Allolio B 2009 Limited prognostic value of

the 2004 International Union Against Cancer staging

classification for adrenocortical carcinoma: proposal for a

Revised TNM Classification. Cancer 115 243–250.

Ferruzzi P, Ceni E, Tarocchi M, Grappone C, Milani S, Galli

A, Fiorelli G, Serio M & Mannelli M 2005 Thiazolidi-

nediones inhibit growth and invasiveness of the human

adrenocortical cancer cell line H295R. Journal of Clinical

Endocrinology and Metabolism 90 1332–1339.

Greeve I, Hermans-Borgmeyer I, Brellinger C, Kasper D,

Gomez-Isla T, Behl C, Levkau B & Nitsch RM 2000

The human DIMINUTO/DWARF1 homolog seladin-1

confers resistance to Alzheimer’s disease-associated

neurodegeneration and oxidative stress. Journal of

Neuroscience 20 7345–7352.

Heaney AP, Fernando M, Yong WH & Melmed S 2002

Functional PPAR-gamma receptor is a novel therapeutic

target for ACTH-secreting pituitary adenomas. Nature

Medicine 8 1281–1287.

Hollingshead MG 2008 Antitumor efficacy testing in rodents.

Journal of the National Cancer Institute 100 1500–1510.

Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti

JM, Schepartz S, Kalyandrug S, Christian M, Arbuck S,

Hollingshead M et al. 2001 Relationships between drug

176

activity in NCI preclinical in vitro and in vivo models and

early clinical trials. British Journal of Cancer 84

1424–1431.

van Kemenade FJ, Raaphorst FM, Blokzijl T, Fieret E,

Hamer KM, Satijn DP, Otte AP & Meijer CJ 2001

Coexpression of BMI-1 and EZH2 polycomb-group

proteins is associated with cycling cells and degree of

malignancy in B-cell non-Hodgkin lymphoma. Blood 97

3896–3901.

Keshamouni VG, Reddy RC, Arenberg DA, Joel B,

Thannickal VJ, Kalemkerian GP & Standiford TJ 2004

Peroxisome proliferator-activated receptor-gamma

activation inhibits tumor progression in non-small-cell

lung cancer. Oncogene 23 100–108.

Khan TS, Imam H, Juhlin C, Skogseid B, Grondal S, Tibblin

S, Wilander E, Oberg K & Eriksson B 2000 Streptozocin

and o,p’DDD in the treatment of adrenocortical cancer

patients: long-term survival in its adjuvant use. Annals

of Oncology 11 1281–1287.

Logie A, Boudou P, Boccon-Gibod L, Baudin E, Vassal G,

Schlumberger M, Le Bouc Y & Gicquel C 2000

Establishment and characterization of a human adreno-

cortical carcinoma xenograft model. Endocrinology 141

3165–3171.

Lombardi A, Cantini G, Piscitelli E, Gelmini S, Francalanci

M, Mello T, Ceni E, Varano G, Forti G, Rotondi M et al.

2008 A new mechanism involving ERK contributes to

rosiglitazone inhibition of tumor necrosis factor-alpha and

interferon-gamma inflammatory effects in human endo-

thelial cells. Arteriosclerosis, Thrombosis, and Vascular

Biology 28 718–724.

Luciani P, Ferruzzi P, Arnaldi G, Crescioli C, Benvenuti S,

Nesi G, Valeri A, Greeve I, Serio M, Mannelli M et al.

2004 Expression of the novel adrenocorticotropin-

responsive gene selective Alzheimer’s disease indicator-1

in the normal adrenal cortex and in adrenocortical

adenomas and carcinomas. Journal of Clinical

Endocrinology and Metabolism 89 1332–1339.

Luton JP, Cerdas S, Billaud L, Thomas G, Guilhaume B,

Bertagna X, Laudat MH, Louvel A, Chapuis Y, Blondeau P

et al. 1990 Clinical features of adrenocortical carcinoma,

prognostic factors, and the effect of mitotane therapy.

New England Journal of Medicine 322 1195–1201.

Monami M, Lamanna C, Marchionni N & Mannucci E

2008 Rosiglitazone and risk of cancer: a meta-analysis

of randomized clinical trials. Diabetes Care 31

1455–1460.

Nissen SE & Wolski K 2007 Effect of rosiglitazone on the

risk of myocardial infarction and death from cardio-

vascular causes. New England Journal of Medicine 356

2457–2471.

Panigrahy D, Singer S, Shen LQ, Butterfield CE,

Freedman DA, Chen EJ, Moses MA, Kilroy S,

Duensing S, Fletcher C et al. 2002 PPARgamma

ligands inhibit primary tumor growth and metastasis

by inhibiting angiogenesis. Journal of Clinical

Investigation 110 923–932.

www.endocrinology-journals.org

AUTHOR COPY ONLYEndocrine-Related Cancer (2010) 17 169–177

Panigrahy D, Huang S, Kieran MW & Kaipainen A 2005

PPARgamma as a therapeutic target for tumor angiogenesis

and metastasis. Cancer Biology & Therapy 4 687–693.

Pecori Giraldi F, Scaroni C, Arvat E, Martin M, Giordano R,

Albiger N, Leao AA, Picu A, Mantero F & Cavagnini F

2006 Effect of protracted treatment with rosiglitazone, a

PPARgamma agonist, in patients with Cushing’s disease.

Clinical Endocrinology 64 219–224.

Read WL, Baggstrom MQ, Fracasso PM & Govindan R 2008

A phase I study of bexarotene and rosiglitazone in patients

with refractory cancers. Chemotherapy 54 236–241.

Reichle A, Bross K, Vogt T, Bataille F, Wild P, Berand A,

Krause SW & Andreesen R 2004 Pioglitazone and

rofecoxib combined with angiostatically scheduled

trofosfamide in the treatment of far-advanced melanoma

and soft tissue sarcoma. Cancer 101 2247–2256.

Silva J, Garcıa V, Garcıa JM, Pena C, Domınguez G, Dıaz R,

Lorenzo Y, Hurtado A, Sanchez A & Bonilla F 2007

Circulating Bmi-1 mRNA as a possible prognostic factor

for advanced breast cancer patients. Breast Cancer

Research 9 R55.

Smith MR, Manola J, Kaufman DS, George D, Oh WK,

Mueller E, Slovin S, Spiegelman B, Small E & Kantoff

PW 2004 Rosiglitazone versus placebo for men with

prostate carcinoma and a rising serum prostate-specific

antigen level after radical prostatectomy and/or radiation

therapy. Cancer 101 1569–1574.

www.endocrinology-journals.org

Teicher BA 1998 Preclinical models for high-dose therapy.

In Anticancer Drug Development Guide: Preclinical

Screening, Clinical Trials and Approval, pp 145–182.

Ed B Teicher. Totowa (NJ): Humana Press, Inc.

Terzolo M, Angeli A, Fassnacht M, Daffara F, Tauchmanova

L, Conton PA, Rossetto R, Buci L, Sperone P,

Grossrubatscher E et al. 2007 Adjuvant mitotane

treatment for adrenocortical carcinoma. New England

Journal of Medicine 356 2372–2380.

Vogt T, Hafner C, Bross K, Bataille F, Jauch KW, Berand A,

Landthaler M, Andreesen R & Reichle A 2003

Antiangiogenetic therapy with pioglitazone, rofecoxib,

and metronomic trofosfamide in patients with advanced

malignant vascular tumors. Cancer 98 2251–2256.

Yee LD, Williams N, Wen P, Young DC, Lester J, Johnson

MV, Farrar WB, Walker MJ, Povoski SP, Suster S et al.

2007 Pilot study of rosiglitazone therapy in women with

breast cancer: effects of short-term therapy on tumor

tissue and serum markers. Clinical Cancer Research 13

246–252.

Yoshizumi T, Ohta T, Ninomiya I, Terada I, Fushida S,

Fujimura T, Nishimura G, Shimizu K, Yi S & Miwa K

2004 Thiazolidinedione, a peroxisome proliferator-

activated receptor-gamma ligand, inhibits growth and

metastasis of HT-29 human colon cancer cells through

differentiation-promoting effects. International

Journal of Oncology 25 631–639.

177