in vivo efficacy of griseofulvin against multiple myeloma

4
Leukemia Research 35 (2011) 1070–1073 Contents lists available at ScienceDirect Leukemia Research jou rnal h omepa g e: www.elsevier.com/locate/leukres In vivo efficacy of griseofulvin against multiple myeloma Young Kim a , Petra Alpmann a , Sabine Blaum-Feder a , Simon Krämer a , Tomoyuki Endo b,c , Desheng Lu b , Dennis Carson b , Ingo G.H. Schmidt-Wolf a,a Department of Internal Medicine III, Center for Integrated Oncology, University of Bonn, Bonn, Germany b University of California San Diego, Moores Cancer Center, La Jolla, CA, USA c Department of Internal Medicine II, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan a r t i c l e i n f o Article history: Received 1 October 2010 Received in revised form 8 October 2010 Accepted 11 October 2010 Available online 26 November 2010 Keywords: Myeloma Lymphoma Wnt Beta catenin Griseofulvin Apoptosis a b s t r a c t We recently confirmed that ciclopirox olamine inhibits Wnt/beta catenin signalling in myeloma. Griseo- fulvin (GF) has similar chemical features as compared to ciclopirox olamine. In this study the anti-tumor effect of GF was investigated. GF demonstrated a major apoptotic activity in various human and murine myeloma and lymphoma cell lines as well as in human primary cells. In vivo, tumor growth as well as overall survival were significantly reduced in mice treated with GF as compared to untreated mice. In conclusion, our results reveal a significant selective induction of apoptosis by GF and suggest a significant in vivo effect against myeloma. © 2010 Elsevier Ltd. All rights reserved. 1. Introduction Major progress has been achieved in the treatment of multi- ple myeloma by the introduction of novel agents like thalidomide, lenalidomide and bortezomib. Nevertheless, multiple myeloma remains an incurable disease. In newly diagnosed patients, the combination of lenalidomide and dexamethasone has a response rate of 91%. Several groups have shown that the Wnt/beta catenin pathway plays an important role in the regulation of cell proliferation, dif- ferentiation and apoptosis [1–3]. Aberrant activation of the Wnt signaling pathway has major oncogenic effects [4–7]. In the canon- ical Wnt pathway, the secreted Wnt proteins bind to a receptor complex, consisting of a member of the Frizzled (Fzd) family and the low-density lipoprotein-receptor-related proteins (LRP) 5 or LRP6. Subsequently, the cytoplasmic adaptor protein disheveled (Dvl) is phosphorylated and inhibits glycogen synthase kinase (GSK)-3 activity through its association with axin. Unphospho- rylated -catenin accumulates in the cytoplasm and translocates into the nucleus, where it interacts with T cell (TCF) and lymphoid- enhancing (LEF) factors to activate transcription of Wnt target Corresponding author at: Center for Integrated Oncology (CIO), Department of Internal Medicine III, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany. Tel.: +49 228 287 15507; fax: +49 228 287 15849. E-mail address: [email protected] (I.G.H. Schmidt-Wolf). genes [4,5,8]. In addition, it has been demonstrated that the Wnt pathway is activated in lymphoma. Thus, the Wnt/beta catenin signaling molecules are attractive candidates for development of targeted approaches in lymphoma treatment. In a more recent work, our group confirmed that the antifungal agent ciclopirox olamine (CIC) inhibits Wnt/beta catenin signaling [9]. Ciclopirox olamine (CIC) is a synthetic antifungal agent used topically for the treatment of yeast infections in humans and is degraded by glucoronidation [10]. It serves as a chelator of polyva- lent metal cations (e.g. Fe 3+ and Al 3+ ) resulting in the inhibition of metal depending enzymes, occurring in the metabolism of the cell. Furthermore, it blocks the cell cycle near the G1/S phase boundary [11]. Griseofulvin (GF), an ethanolamine salt of the hydroxamic acid derivative piroctone, is a pyridone derivate as is CIC [12]. It is used as an antifungal drug. In this study, we investigated the effect of griseofulvin (GF) on multiple myeloma and lymphoma cells in vitro and in vivo in a murine myeloma model. 2. Methods 2.1. Cell lines and culture conditions The lymphoma cell lines LAM-53, SU-DHL-4, Daudi and Raji, as well as the myeloma cell lines OPM-2, RPMI-8226 and U-266 (all obtained from DSMZ, Collec- tion of Microorganisms and Cell Culture, Braunschweig, Germany) were cultured in RPMI-1640 Medium consisting of 10% heat-inactivated fetal calf serum (FCS, Invit- 0145-2126/$ see front matter © 2010 Elsevier Ltd. All rights reserved. doi:10.1016/j.leukres.2010.10.008

Upload: young-kim

Post on 26-Jun-2016

218 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: In vivo efficacy of griseofulvin against multiple myeloma

I

YTa

b

c

a

ARRAA

KMLWBGA

1

plrcr

pfsictL((rie

IG

0d

Leukemia Research 35 (2011) 1070– 1073

Contents lists available at ScienceDirect

Leukemia Research

jou rna l h omepa g e: www.elsev ier .com/ locate / leukres

n vivo efficacy of griseofulvin against multiple myeloma

oung Kima, Petra Alpmanna, Sabine Blaum-Federa, Simon Krämera,omoyuki Endob,c, Desheng Lub, Dennis Carsonb, Ingo G.H. Schmidt-Wolfa,∗

Department of Internal Medicine III, Center for Integrated Oncology, University of Bonn, Bonn, GermanyUniversity of California San Diego, Moores Cancer Center, La Jolla, CA, USADepartment of Internal Medicine II, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan

r t i c l e i n f o

rticle history:eceived 1 October 2010eceived in revised form 8 October 2010ccepted 11 October 2010vailable online 26 November 2010

a b s t r a c t

We recently confirmed that ciclopirox olamine inhibits Wnt/beta catenin signalling in myeloma. Griseo-fulvin (GF) has similar chemical features as compared to ciclopirox olamine. In this study the anti-tumoreffect of GF was investigated. GF demonstrated a major apoptotic activity in various human and murinemyeloma and lymphoma cell lines as well as in human primary cells. In vivo, tumor growth as well asoverall survival were significantly reduced in mice treated with GF as compared to untreated mice. Inconclusion, our results reveal a significant selective induction of apoptosis by GF and suggest a significant

eywords:yeloma

ymphomant

eta cateninriseofulvin

in vivo effect against myeloma.© 2010 Elsevier Ltd. All rights reserved.

poptosis

. Introduction

Major progress has been achieved in the treatment of multi-le myeloma by the introduction of novel agents like thalidomide,

enalidomide and bortezomib. Nevertheless, multiple myelomaemains an incurable disease. In newly diagnosed patients, theombination of lenalidomide and dexamethasone has a responseate of 91%.

Several groups have shown that the Wnt/beta catenin pathwaylays an important role in the regulation of cell proliferation, dif-erentiation and apoptosis [1–3]. Aberrant activation of the Wntignaling pathway has major oncogenic effects [4–7]. In the canon-cal Wnt pathway, the secreted Wnt proteins bind to a receptoromplex, consisting of a member of the Frizzled (Fzd) family andhe low-density lipoprotein-receptor-related proteins (LRP) 5 orRP6. Subsequently, the cytoplasmic adaptor protein disheveledDvl) is phosphorylated and inhibits glycogen synthase kinaseGSK)-3� activity through its association with axin. Unphospho-

ylated �-catenin accumulates in the cytoplasm and translocatesnto the nucleus, where it interacts with T cell (TCF) and lymphoid-nhancing (LEF) factors to activate transcription of Wnt target

∗ Corresponding author at: Center for Integrated Oncology (CIO), Department ofnternal Medicine III, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105 Bonn,ermany. Tel.: +49 228 287 15507; fax: +49 228 287 15849.

E-mail address: [email protected] (I.G.H. Schmidt-Wolf).

145-2126/$ – see front matter © 2010 Elsevier Ltd. All rights reserved.oi:10.1016/j.leukres.2010.10.008

genes [4,5,8]. In addition, it has been demonstrated that the Wntpathway is activated in lymphoma. Thus, the Wnt/beta cateninsignaling molecules are attractive candidates for development oftargeted approaches in lymphoma treatment.

In a more recent work, our group confirmed that the antifungalagent ciclopirox olamine (CIC) inhibits Wnt/beta catenin signaling[9]. Ciclopirox olamine (CIC) is a synthetic antifungal agent usedtopically for the treatment of yeast infections in humans and isdegraded by glucoronidation [10]. It serves as a chelator of polyva-lent metal cations (e.g. Fe3+ and Al3+) resulting in the inhibition ofmetal depending enzymes, occurring in the metabolism of the cell.Furthermore, it blocks the cell cycle near the G1/S phase boundary[11].

Griseofulvin (GF), an ethanolamine salt of the hydroxamic acidderivative piroctone, is a pyridone derivate as is CIC [12]. It is usedas an antifungal drug.

In this study, we investigated the effect of griseofulvin (GF) onmultiple myeloma and lymphoma cells in vitro and in vivo in amurine myeloma model.

2. Methods

2.1. Cell lines and culture conditions

The lymphoma cell lines LAM-53, SU-DHL-4, Daudi and Raji, as well as themyeloma cell lines OPM-2, RPMI-8226 and U-266 (all obtained from DSMZ, Collec-tion of Microorganisms and Cell Culture, Braunschweig, Germany) were cultured inRPMI-1640 Medium consisting of 10% heat-inactivated fetal calf serum (FCS, Invit-

Page 2: In vivo efficacy of griseofulvin against multiple myeloma

esearch 35 (2011) 1070– 1073 1071

rf3

d1sch

2

spwg

2

t(wf

2

Dad(ia

2

bw3iaDse

r

2

sBPcwiKp

2

cw

2

W

2

mi

Table 1The effect of griseofulvin, thalidomide,and lenalidomide on myeloma (OPM-2, U266,RPMI 8226 and KMS18) and lymphoma (LAM53, SU-DHL-4 and Raji) cell lines wasassayed. PBMC and PBL derived from healthy individuals were used as controls.1 × 105 cells were cultured with each compound using various concentrations for 3days. Then cell viability was measured by DIOC6 staining by flow cytometry and IC50 rates were determined. Results represent data from 2 to 4 separate experimentseach.

Cell line IC50 ofgriseofulvin

IC50 ofthalidomide

IC50 oflenalidomide

OPM-2 ND 53 �M 13 �MU266 18 �M 300 �M 34 �MRPMI 8226 26 �M >400 �M 2 �MRaji 33 �M >400 �M >1000 �MOCI- Ly8 LAM 53 30 �M >400 �M >1000 �MSU-DHL 4 22 �M >400 �M >1000 �MMPC 11 41 �M ND ND

Similar results were obtained for human lymphoma cell linesRaji (33 �M), OCI-Ly-LAM-53 (30 �M) and SU-DHL 4 (22 �M)(Table 1).

Y. Kim et al. / Leukemia R

ogen, Karlsruhe, Germany), 2.5% 1 M HEPES, and 1x penicillin/streptomycin (allrom PAA Laboratories GmbH, Cölbe, Germany). Cells were cultured at a density of.3 × 105 cells ml−1 and incubated at 37 ◦C with 5% CO2 and 95% humidity.

MPC11 (DMSZ, Braunschweig, Germany) is a murine plasmocytoma cell lineerived from the Balb/c strain expressing IgG2b. Cells were cultured in RPMI640 medium (PAA Laboratories GmbH, Austria) supplemented with 5% fetal calferum (FCS), 2 mM glutamine (both from PAA, Cölbe, Germany), 100 U/ml peni-illin/100 U/ml streptomycin (both from Seromed, Jülich, Germany) at 37 ◦C in aumidified 5% CO2 atmosphere.

.2. Human samples

Peripheral blood mononuclear cells (PBMC) and PBL were isolated from bloodamples of healthy volunteers by Ficoll density gradient centrifugation (Lympho-rep, Nycomed, Oslo, Norway). In addition, bone marrow samples from patientsith myeloma were obtained. Ethics approval had been obtained according to the

uidelines of our institution.

.3. Drugs and chemical reagents

In our experiments the following drugs were used: Thalidomide (Grünen-hal Pharma GmbH, Aachen, Germany), lenalidomide was obtained from CelgeneMunich, Germany), and griseofulvin (PO) from Spinnrad, Bonn, Germany. All drugsere tested at various concentrations for 24–72 h. In addition, in this study griseo-

ulvin (GF, Spinnrad, Bonn, Germany) was applied orally.

.4. DiOC6 and PI-staining

1 × 105 cells were cultured in 3 ml medium in 6-well plates. GF was dissolved inMSO, and added in optimized concentrations between 10 �M (CIC) and 30 �M (EA)lone or in combination with the therapeutic agents at various concentrations for 3ays. The apoptosis assay was performed with 3′ ,3-dihexyloxacarbocyanine iodideDiOC6) detecting mitochondrial membrane potential in viable cells, and propidiumodide (PI) which binds to DNA in necrotic cells, measured by a fluorescence-ctivated cell sorter (FACS).

.5. Fluorescence-activated cell analysis

For FACS analysis, 500 �l staining solution containing 80 nM DiOC6 in FACSuffer, consisting of deficient RPMI-medium with 0.5% bovine serum albumin (BSA),as mixed with equal volumes of the cell sample in a glass tube and incubated at

7 ◦C for 15 min. After a washing step with PBS/BSA 1% the cells were re-suspendedn 500 �l PBS/BSA 1%. After addition of 5 �l PI-solution (100 �g/ml) the cells werenalyzed by FACS. Using this assay, viable cells reveal high fluorescence intensity foriOC6 and a low expression for PI. In contrary, apoptotic cells show a low expres-

ion for DiOC6 and also a low expression for PI (2). Finally, necrotic cells show a lowxpression for DiOC6 and a high expression for PI.

A mean IC50 value in myeloma cells was determined using the mean of the IC50

esults determined in OPM-2, U266 and RPMI 8226 cells.

.6. Isolation of peripheral blood mononuclear cells (PBMCs)

PBMCs were isolated from blood of healthy donors by Ficoll-Hypaque den-ity gradient centrifugation. Blood from buffy coats was mixed 1:2 with PBS/1%SA (both PAA, Cölbe, Germany) and used for a ficoll gradient (LymphoPrep,AA, Cölbe, Germany). After the centrifugation at 800×g for 30 min, the leuko-yte layer was removed and transferred to new tubes. Subsequently, these cellsere washed 3 times with PBS/1% BSA and re-suspended in fresh medium, consist-

ng of RPMI medium (PAA, Cölbe, Germany) with 10% fetal calf serum (Invitrogen,arlsruhe, Germany), 2.5% hepes buffer solution (PAA, Cölbe, Germany), and 1%enicillin/streptomycin (PAA, Cölbe, Germany).

.7. Bone marrow samples

Bone marrow samples from patients with myeloma were taken after informedonsent, single cell suspensions generated and cells ficolled. Cells were incubatedith or without GF for 3 days and measured for viability.

.8. Western blot

We analyzed the effect of GF on the Wnt/beta catenin pathway by Western blot.estern blot was performed as described recently [13].

.9. Animal studies

All animal experiments were done at least in duplicate with groups of six BALB/cice (Charles River, Sulzfeld, Germany). 5 × 105 MCP11 murine myeloma cells were

njected per mouse subcutaneously into Balb/c mice. Mice were treated orally with

PBMC 180 �M >400 �M >400 �MPBL 80 �M >400 �M >200 �M

450 �g/day PO. Overall survival and tumor growth were measured. Tumor vol-ume was calculated as follows: volume = length × width2 × 0.52. Animals were killedwhen tumor volume reached 2000 mm3.

2.10. Statistical analysis

For statistical analysis, the numbers of the results comprising the relative via-bility are expressed as the mean ± standard error of the mean (SEM). Differentsample sizes (n) were chosen for different cell lines. Student’s t-test was used forstatistical analysis. A p value below 0.05 was considered significant. Statistical sur-vival analyses were performed with the software GraphPad InStat, Version 3.0.0(GraphPad Software, San Diego, CA), applied the Mann-Whitney test (non-paired,non-parametric).

3. Results

3.1. Effect of GF in vitro on viability of various cell lines

GF significantly decreased the viability of all myeloma cell linesin vitro (Table 1 and Figs. 1–4). Even small dosages of griseoful-vin were toxic, so the IC50 could be reached with less than 50 �Mof griseofulvin for the human myeloma cell lines U-266 (18 �M),RPMI-8226 (26 �M) and murine MPC-11 (41 �M).

Fig. 1. Flow cytometric analysis of griseofulvin in U266 cells. 1 × 105 cells werecultured with various concentrations of griseofulvin for 3 days. Then cell viabilitywas measured by DIOC6 staining in flow cytometry. Results represent data from twoseparate experiments. Data are shown as mean ± SD.

Page 3: In vivo efficacy of griseofulvin against multiple myeloma

1072 Y. Kim et al. / Leukemia Researc

Fig. 2. Flow cytometric analysis of griseofulvin in RPMI 8226 cells. 1 × 105 cells werecultured with various concentrations of griseofulvin for 3 days. Then cell viabilitywas measured by DIOC6 staining in flow cytometry. Results represent data from twoseparate experiments. Data are shown as mean ± SD.

Fig. 3. Flow cytometric analysis of griseofulvin in peripheral blood lymphocytes(PBL). 1 × 105 cells were cultured with various concentrations of griseofulvin for 3days. Then cell viability was measured by DIOC6 staining in flow cytometry. Resultsrepresent data from two separate experiments. Data are shown as mean ± SD.

Fig. 4. Flow cytometric analysis of griseofulvin in murine MPC11 myeloma cells.1 × 105 cells were cultured with various concentrations of griseofulvin for 3 days.Then cell viability was measured by DIOC6 staining in flow cytometry. Results rep-resent data from two separate experiments. Data are shown as mean ± SD.

h 35 (2011) 1070– 1073

In contrast, in PBMCs (180 �M), which were used as control cells,griseofulvin was less efficient (Table 1).

Cytotoxic activity was shown to be via induction of apoptosis(data not shown).

3.2. Effect of DMSO on viability of cell lines

In contrast to PBMC and lymphoma cell lines, the myelomacell line OPM-2 was highly sensitive towards DMSO and showeda decrease of relative viability to 66.8 ± 2.3% (data not shown).

3.3. Effect of thalidomide, lenalidomide and GF on viability ofmyeloma cell lines

All three compounds significantly (p < 0.05) decreased the via-bility of myeloma cell lines in vitro (Table 1, Figs. 1–4). A mean IC50value in myeloma cells was determined: lenalidomide (16.3 �M)and GF (22 �M) were more effective than thalidomide (>251 �M;Table 1).

In contrast the effect on normal PBL was marginal in GF andlenalidomide. Here, no effect was found for thalidomide (Table 1).

3.4. Effect of thalidomide, lenalidomide and GF on viability oflymphoma cell lines

Only GF significantly (p < 0.05) decreased the viability of lym-phoma cell lines in vitro (Table 1). In contrast, lenalidomide andthalidomide showed no effect (Table 1).

3.5. Effect of GF on the Wnt/beta catenin pathway in myelomacells

The effect of GF on �-catenin expression in myeloma cells wastested. MPC11 cells were found to be �-catenin positive (data notshown).

3.6. Effect of GF on viability of myeloma primary cells

Preliminary data suggest that GF inhibits the growth of primarymyeloma cells derived from bone marrow from patients with mul-tiple myeloma (data not shown).

3.7. Effect of GF on myeloma cells in vivo

All animals in the control group developed a tumor larger than2000 mm3 and therefore had to be killed by day 22 (Fig. 5).

GF was administered by gavage with a daily dosage of 450 �g peranimal in the treatment group. After the MPC11 wildtype cells were

Fig. 5. In vivo effect of griseofulvin (PO) in MPC11 bearing mice. PO was adminis-tered orally by gavage at a concentration of 450 �g/day/mouse. Tumor growth inmice is presented. Six animals were used in each group. The observation time was60 days.

Page 4: In vivo efficacy of griseofulvin against multiple myeloma

Y. Kim et al. / Leukemia Researc

Fig. 6. In vivo effect of griseofulvin (PO) in MPC11 bearing mice. PO was adminis-tm6

asot

o

4

rmwoWsotCdmwatdittbpb

mlvtsa

uoWbl

[

[

[

[

[

[

[

cytic leikemia cells by inhibition of the Wnt/beta-catenin pathway. PloS One

ered orally by gavage at a concentration of 450 �g/day/mouse. Overall survival ofice is presented. Six animals were used in each group. The observation time was

0 days.

dministered subcutaneously at the back of the animals on day 8,olid tumor nodules formed after a few days in 5 of 6 animals. Theverall survival time was significantly (p < 0.05) longer as comparedo the control group (Fig. 6).

With respect to toxicity of GF administration no side effects werebserved at the applied dose.

. Discussion

The Wnt signaling pathway has been shown to play a criticalole in the early phases of B lymphocyte development. Multipleyeloma (MM) cells, but not cells from healthy donors and patientsith monoclonal gammopathy of undetermined significance or

ther plasma cell dyscrasias involving bone marrow express thent-signaling antagonist DKK1 [14]. It has been reported that

ecretion of DKK1 by MM cells likely contributes to the formationf osteolytic lesions in this disease by inhibiting Wnt signaling:he latter being essential for osteoblast differentiation and survival.hanges of DKK1 expression in MM cells could be traced throughisturbances in the JNK signaling cascade which is differentiallyodulated by oxidative stress and interactions between MM cellsith osteoclasts, in vitro. Despite its role as a tumor suppressor

nd mediator of apoptosis in other cell types, including osteoblasts,he data indicate that DKK1 – a stress-responsive gene in MM –oes not mediate apoptotic signaling, is not activated by TP53, and

ts forced over expression could not inhibit cell growth or sensi-ize MM cells towards apoptosis, following treatment with eitherhalidomide or lenalidomide. Therefore, specific strategies may beeneficial to modulate persistent activation of the JNK pathway inreventing disease progression and treating myeloma associatedone disease by inhibiting DKK1 expression [14].

Many hypotheses have been proposed to explain the molecularechanism of thalidomide’s teratogenicity, in particular regarding

imb defects. Most experimental evidence in vivo has been pro-ided for a model suggesting the generation of oxidative stress byhalidomide with subsequent down regulation of both Wnt and Akturvival pathways. In addition, transcription factors Tbx5 and Sall4re involved in thalidomide induced molecular pathology [15].

Since thalidomide down regulates the Wnt pathway and issed for treatment of myeloma patients, testing the combination

f thalidomide and Wnt inhibitors became of particular interest.e have recently demonstrated a synergistic effect of the com-

ination of thalidomide, PO, CIC and EA in myeloma cells. In theymphoma cell lines SU-DHL4 and LAM-53 the Wnt inhibitors CIC

[

h 35 (2011) 1070– 1073 1073

and EA induce apoptosis through down regulation of beta catenin,an important molecule within the Wnt pathway [16,17].

In this study we could show that the daily oral administration ofGF could lead to apoptosis of myeloma and lymphoma cells in vitroand in vivo. Combined with our previous results, our data suggestthat GF, like EA and CIC may inhibit Wnt/beta catenin signaling inmyeloma cell lines and thus significantly prolong the overall sur-vival time in vivo, without deteriorating the general condition ofthe animals. These results might lead to new treatment optionsin patients with multiple myeloma and should be followed-up infurther studies for the benefit of patients with myeloma.

Conflict of interest statement

All authors declare no conflict of interest.

Acknowledgements

We kindly acknowledge the support of T. Kipps, La Jolla, P.Brossart, Bonn, Germany and Axel Glasmacher, Bonn. This studywas kindly supported in part by the Deutsche Krebshilfe, Bonn,Germany.

Contributions: YK, PA, SBF and SK performed the experiments.ISW, TE, DL and DC designed the study and revised the manuscript.ISW wrote the paper.

References

[1] Cadigan KM, Liu YI. Wnt signaling: complexity at the surface. J Cell Sci2006;119:395–406.

[2] Miller JR, Hocking AM, Brown JD, Moon RT. Mechanism and function of sig-nal transduction by the Wnt/beta catenin and Wnt/Ca2+ pathways. Oncogene1999;18(55):7860–72.

[3] You Z, Saims D, Chen S, Zhang Z, Guttridge DC, Guan KL, MacDougald OA,Brown AM, Evan G, Kitajewski J, Wang CY. Wnt signaling promotes oncogenictransformation by inhibiting c-Myc-induced apoptosis. J Cell Biol 2002;157:429–40.

[4] Clevers H. Wnt/beta-catenin signaling in development and disease. Cell2006;127:469–80.

[5] Moon RT, Kohn AD, De Ferrari GV, Kaykas A. WNT and beta-catenin signalling:diseases and therapies. Nat Rev Genet 2004;5:691–701.

[6] Nusse R. Wnt signaling in disease and in development. Cell Res 2005;15:28–32.[7] Polakis P. Wnt signaling and cancer. Genes Dev 2000;14:1837–51.[8] Willert K, Jones KA. Wnt signaling: is the party in the nucleus? Genes Dev

2006;20:1394–404.[9] Lu D, Zhao Y, Tawatao R, Cottam HB, Sen M, Leoni LM, Kipps TJ, Corr M, Carson

DA. Activation of the Wnt signaling pathway in chronic lymphocytic leukemia.Proc Natl Acad Sci 2004;101:3118–23.

10] Aizawa S, Ookawa K, Kudo T, Asano J, Hayakari M, Tsuchida S. Characterizationof cell death induced by ethacrynic acid in a human colon cancer cell line DLD-1and suppression by N-acetyl-l-cysteine. Cancer Sci 2002;94:886–93.

11] Tew KD, Bomber AM, Hoffman SJ. Ethacrynic acid and piriprost as enhancersof cytotoxicity in drug resistant and sensitive cell lines. Cancer Res1988;48:3622–5.

12] Estrela JM, Ortega A, Obrador E. Glutathione in cancer biology and therapy. CritRev Clin Lab Sci 2006;43:143–81.

13] McCaughan FM, Brown AL, Harrison DJ. The effect of inhibition of glutathioneS-transferase P on the growth of the Jurkat human T cell line. J Pathol1994;172:357–62.

14] Nagourney RA, Messenger JC, Kern DH, Weisenthal LM. Enhancement of anthra-cycline and alkylator cytotoxicity by ethacrynic acid in primary cultures ofhuman tissues. Cancer Chemother Pharmacol 1990;26:318–22.

15] Hoffman BD, Hanauske-Abel HM, Flint A, Lalande M. A new class of reversiblecell cycle inhibitors. Cytometry 1991;12:26–32.

16] Lu D, Liu JX, Endo T, Zhou H, Yao S, Willert K, Schmidt-Wolf IGH, Kipps TJ,Carson DA. Ethacrynic acid exhibitis selective cytotoxicity to chronic Lympho-

2009;4(12):e8294.17] Schmidt M, Sievers E, Endo T, Lu D, Carson DA, Schmidt-Wolf IGH. Target-

ing Wnt pathway in lymphoma and myeloma cells. Br J Haematol 2009;144:796–8.