Top Banner
Differential cellular and molecular effects of bortezomib, a proteasome inhibitor, in human breast cancer cells Jordi Codony-Servat, 1 Maria A. Tapia, 1 Marta Bosch, 1 Cristina Oliva, 1 Josep Domingo-Domenech, 1 Begon ˜ a Mellado, 1 Mark Rolfe, 2 Jeffrey S. Ross, 2,3 Pere Gascon, 1 Ana Rovira, 1 and Joan Albanell 1 1 Laboratory of Experimental Oncology, Medical Oncology Department, Institut d’Investigacions Biomediques August Pi i Sunyer, Hospital Clinic i Provincial de Barcelona, Barcelona, Spain; 2 Millennium Pharmaceuticals, Inc., Cambridge, Massachusetts; and 3 Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York Abstract The cellular and molecular effects of the proteasome inhibitor bortezomib on breast cancer cells are as yet poorly characterized. Here, in a panel of six breast cancer cell lines, bortezomib reduced viability in a concentration- dependent, time-dependent, and cell line – dependent manner. Proteasome activity was relatively high in two of the three more resistant cell lines. No relationship was observed between bortezomib effects on cell viability and expression/phosphorylation of HER-2, epidermal growth factor receptor (EGFR), AKT, or extracellular signal- regulated kinase 1/2 (ERK1/2). Molecular effects of bortezomib were further studied in SK-BR-3 and BT-474 cells because they share expression of EGFR and over- expression of HER-2 while, in contrast, SK-BR-3 cells were 200-fold more sensitive to this agent. Proteasome activity was inhibited to a similar extent in the two cell lines, and known proteasome substrates accumulated similarly. In SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration of bortezomib. In contrast, phosphorylation of Raf/mitogen-activated protein kinase kinase 1/2 (MEK 1/2)/ERK1/2 increased by bortezomib. In BT-474 cells, the effects were much less pronounced. Treatment of SK- BR-3 cells with bortezomib combined with pharmacologic inhibitors of EGFR, phosphatidylinositol 3V -kinase, or MEK resulted in modest or no enhancement of the effects on cell viability. Collectively, these results show that borte- zomib has differential cellular and molecular effects in human breast cancer cells. The bortezomib-observed effects on signaling transduction molecules might be relevant to help to design mechanistic-based combination treatments. [Mol Cancer Ther 2006;5(3):665 – 75] Introduction The proteasome plays a pivotal role in the cellular housekeeping by eliminating mutant, misfolded, and damaged proteins. Moreover, the proteasome is involved in the targeted elimination of regulatory proteins, such as transcription factors, signaling molecules, and cell cycle inhibitors (1). The inhibition of the proteasome results in the abnormal accumulation of many intracellular proteins, thereby disrupting cellular homeostasis. Thus, cells under- go cell cycle arrest or programmed cell death (2). The observation that tumor cells are more sensitive to protea- some inhibition than normal cells led to propose the proteasome as a novel target for cancer treatment (3). Bortezomib (Velcade, formerly known as PS-341) is a selective and reversible proteasome inhibitor that results in a wide range of molecular sequels, including stabiliza- tion of cell cycle regulatory proteins, inhibition of nuclear factor-nB (NF-nB) activation, induction of apoptosis, and override of Bcl-2 resistance and antiangiogenesis (3, 4). More importantly, bortezomib has a wide range of antitumor activity and increases the activity of multiple chemotherapeutic agents (5). Bortezomib inhibits 20S pro- teasome activity in whole cells with a K i of f7 nmol/L, consistent with its mean IC 50 (7 nmol/L) in the National Cancer Institute panel of 60 cell lines (4). Preclinical studies suggested that twice-weekly regimens resulting in protea- some inhibition, as measured in blood, approaching but not exceeding 80% would be optimal (6). Early clinical trials of bortezomib incorporated a pharmacodynamic ex vivo assay developed to study the degree and kinetics of proteasome inhibition achieved in patients as a key element to select the dose for patients (7 – 9). Bortezomib is approved in many countries for the treatment of chemorefractory multiple myeloma patients (10, 11) and is in further clinical development in multiple tumor types, including breast cancer (12 – 14). Preliminary reports of trials using bortezomib in breast cancer patients have shown lack of significant clinical activity against this disease (15, 16). The potential clinical development of bortezomib in breast cancer would need to be based on combination therapeutic strategies with agents such as docetaxel or the anti-HER-2 antibody trastuzumab (17 – 20). To fully explore bortezomib-based combinations, it is of importance to gain further knowledge in the molecular effects of bortezomib in breast cancer. In the present study, Received 5/10/05; revised 12/14/05; accepted 1/12/06. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Note: J. Codony-Servat and M.A. Tapia contributed equally to this work. Requests for reprints: Joan Albanell, Medical Oncology Department, Hospital del Mar, Passeig Maritim, 25-29 08003 Barcelona, Spain. Phone: 34-93-248-3137; Fax: 34-93-248-3366. E-mail: [email protected] Copyright C 2006 American Association for Cancer Research. doi:10.1158/1535-7163.MCT-05-0147 665 Mol Cancer Ther 2006;5(3). March 2006 on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from
13

Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

Jun 06, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

Differential cellular and molecular effects of bortezomib, aproteasome inhibitor, in human breast cancer cells

Jordi Codony-Servat,1 Maria A. Tapia,1

Marta Bosch,1 Cristina Oliva,1

Josep Domingo-Domenech,1 Begona Mellado,1

Mark Rolfe,2 Jeffrey S. Ross,2,3 Pere Gascon,1

Ana Rovira,1 and Joan Albanell1

1Laboratory of Experimental Oncology, Medical OncologyDepartment, Institut d’Investigacions Biomediques August Pi iSunyer, Hospital Clinic i Provincial de Barcelona, Barcelona,Spain; 2Millennium Pharmaceuticals, Inc., Cambridge,Massachusetts; and 3Department of Pathology and LaboratoryMedicine, Albany Medical College, Albany, New York

AbstractThe cellular and molecular effects of the proteasomeinhibitor bortezomib on breast cancer cells are as yetpoorly characterized. Here, in a panel of six breast cancercell lines, bortezomib reduced viability in a concentration-dependent, time-dependent, and cell line–dependentmanner. Proteasome activity was relatively high in twoof the three more resistant cell lines. No relationship wasobserved between bortezomib effects on cell viability andexpression/phosphorylation of HER-2, epidermal growthfactor receptor (EGFR), AKT, or extracellular signal-regulated kinase 1/2 (ERK1/2). Molecular effects ofbortezomib were further studied in SK-BR-3 and BT-474cells because they share expression of EGFR and over-expression of HER-2 while, in contrast, SK-BR-3 cells were200-fold more sensitive to this agent. Proteasome activitywas inhibited to a similar extent in the two cell lines, andknown proteasome substrates accumulated similarly. InSK-BR-3 cells, a marked inhibition of EGFR, HER-2, andAKT phosphorylation was observed at a clinically relevantconcentration of bortezomib. In contrast, phosphorylationof Raf/mitogen-activated protein kinase kinase 1/2 (MEK1/2)/ERK1/2 increased by bortezomib. In BT-474 cells,the effects were much less pronounced. Treatment of SK-BR-3 cells with bortezomib combined with pharmacologicinhibitors of EGFR, phosphatidylinositol 3V-kinase, or MEKresulted in modest or no enhancement of the effects on

cell viability. Collectively, these results show that borte-zomib has differential cellular and molecular effects inhuman breast cancer cells. The bortezomib-observedeffects on signaling transduction molecules might berelevant to help to design mechanistic-based combinationtreatments. [Mol Cancer Ther 2006;5(3):665–75]

IntroductionThe proteasome plays a pivotal role in the cellularhousekeeping by eliminating mutant, misfolded, anddamaged proteins. Moreover, the proteasome is involvedin the targeted elimination of regulatory proteins, such astranscription factors, signaling molecules, and cell cycleinhibitors (1). The inhibition of the proteasome results inthe abnormal accumulation of many intracellular proteins,thereby disrupting cellular homeostasis. Thus, cells under-go cell cycle arrest or programmed cell death (2). Theobservation that tumor cells are more sensitive to protea-some inhibition than normal cells led to propose theproteasome as a novel target for cancer treatment (3).Bortezomib (Velcade, formerly known as PS-341) is aselective and reversible proteasome inhibitor that resultsin a wide range of molecular sequels, including stabiliza-tion of cell cycle regulatory proteins, inhibition of nuclearfactor-nB (NF-nB) activation, induction of apoptosis, andoverride of Bcl-2 resistance and antiangiogenesis (3, 4).More importantly, bortezomib has a wide range ofantitumor activity and increases the activity of multiplechemotherapeutic agents (5). Bortezomib inhibits 20S pro-teasome activity in whole cells with a K i of f7 nmol/L,consistent with its mean IC50 (7 nmol/L) in the NationalCancer Institute panel of 60 cell lines (4). Preclinical studiessuggested that twice-weekly regimens resulting in protea-some inhibition, as measured in blood, approaching but notexceeding 80% would be optimal (6). Early clinical trials ofbortezomib incorporated a pharmacodynamic ex vivo assaydeveloped to study the degree and kinetics of proteasomeinhibition achieved in patients as a key element to select thedose for patients (7–9).Bortezomib is approved in many countries for the

treatment of chemorefractory multiple myeloma patients(10, 11) and is in further clinical development in multipletumor types, including breast cancer (12–14). Preliminaryreports of trials using bortezomib in breast cancer patientshave shown lack of significant clinical activity againstthis disease (15, 16). The potential clinical developmentof bortezomib in breast cancer would need to be based oncombination therapeutic strategies with agents such asdocetaxel or the anti-HER-2 antibody trastuzumab (17–20).To fully explore bortezomib-based combinations, it is ofimportance to gain further knowledge in the moleculareffects of bortezomib in breast cancer. In the present study,

Received 5/10/05; revised 12/14/05; accepted 1/12/06.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely toindicate this fact.

Note: J. Codony-Servat and M.A. Tapia contributed equally to this work.

Requests for reprints: Joan Albanell, Medical Oncology Department,Hospital del Mar, Passeig Maritim, 25-29 08003 Barcelona, Spain. Phone:34-93-248-3137; Fax: 34-93-248-3366. E-mail: [email protected]

Copyright C 2006 American Association for Cancer Research.

doi:10.1158/1535-7163.MCT-05-0147

665

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 2: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

we aimed to characterize the effects of bortezomib in apanel of human breast cancer cell lines on viability, cellcycle, and apoptosis. Considering the important role of theepidermal growth factor receptor (EGFR) and HER-2 andtheir signaling transduction pathways in breast cancer, theeffects of bortezomib on these pathways were also invest-igated. We also sought to explore the effects of cotreatmentwith bortezomib and selected pharmacologic agents thattarget these pathways.

Materials andMethodsCell Lines and ReagentsHuman breast cancer cell lines were purchased from

the American Type Culture Collection (Rockville, MD)and cultured at 37jC with 5% CO2 in a humidifiedincubator in DMEM/F12 (MDA-MD-231, MDA-MD-453,MDA-MD-468, BT-474, and SK-BR-3) or DMEM (MCF-7)supplemented with 2 mmol/L L-glutamine and 10% fetalbovine serum. For BT-474, insulin (0.01 Ag/mL) wasadded. Media and supplements were obtained from LifeTechnologies (Gaithersburg, MD). Recombinant humantumor necrosis factor-a (TNF-a) and EGF were purchasedfrom R&D Systems (Minneapolis, MN). Chemical inhib-itors for the EGFR tyrosine kinase (AG1478) and mitogen-activated protein kinase kinase [MEK (PD-098059)] andphosphatidylinositol 3V-kinase [PI3K (LY294002)] werefrom Calbiochem (San Diego, CA). All of them weredissolved as recommended by the manufacturers, ali-quoted, and stored at �20jC. Bortezomib was kindlyprovided by Millennium Pharmaceuticals (Cambridge,MA). MG-132 was purchased from Calbiochem. In bothcases, 10 mmol/L aliquots of drug in DMSO were storedat �20jC, thawed, and diluted just before use. Allchemicals not specified below were purchased fromSigma (St. Louis, MO).

3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazo-lium BromideAssayThe effects of bortezomib on the proliferation and

viability of breast cancer cells were studied by use of thecolorimetric method provided by the MTS-CellTiter 96Aqueous Non-Radioactive Cell Proliferation Assay kit(Promega, Madison, WI). 3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium(MTS) assay is a modification from 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay anddepends on two solutions: a tetrazolium compound (MTS)and an electron coupling reagent (phenazine methosulfate).MTS is bioreduced by cells into a formazan product. Theconversion of MTS into aqueous, soluble formazan isaccomplished by dehydrogenase enzymes found in cellsthat are metabolically active. The quantity of formazanproduct as measured by the amount of 490-nm absorbanceis directly proportional to the number of living cells inculture (21). We did the MTS assay in 96-well, flat-bottomed plates (Nunc, Naperville, IL). Approximately1 � 104 cells were seeded in 100 AL drug-free media andincubated for 24 hours before drug treatment; 100 AL of

various 2� drug concentration (1� final concentration)were added for times from 24 to 72 hours. Forty microlitersof MTS/phenazine methosulfate solution were added tothe wells, and the cells were further incubated for 2 to 4hours. The amount of soluble formazan produced, bycellular reduction of the MTS, was measured by theabsorbance on a microplate spectrophotometer (MolecularDynamics, Sunnyvale, CA) at 490 nm (test wavelength) and690 nm (reference wavelength). The percentage of surviv-ing cells was estimated by dividing the A490 nm � A690 nm oftreated cells by the A490 nm � A690 nm of control cells.Approximate IC50 values were determined from the dose-response curve. Data were derived from at least threeindependent experiments (in quatriplicate).

Flow CytometryCells (2 � 106) were seeded in 100-mm plates, allowed to

attach overnight, and then treated as indicated for times upto 72 hours. At each time point, cells (floater and adherent)were collected, counted, and washed twice with cold PBS.For cell cycle analysis based on DNA content, cells (1 �106/mL) were fixed with 70% ethanol in PBS at �20jCfor 2 to 3 days, centrifuged, and stained with propidiumiodide (Sigma; 20 Ag/mL in PBS with 0.1% Triton X-100) inthe presence of RNase A (0.2 mg/mL; Sigma) at roomtemperature for 30 minutes in the dark. The cells were thenanalyzed with FACScan and Cell-Quest software (BectonDickinson, Mountain View, CA). For apoptosis analysis,phosphatidylserine externalization was measured using theAnnexin V/FITC Apoptosis Detection kit (Roche, Indian-apolis, IN). Cells were labeled with Annexin V/FITC andcounterstained with propidium iodide according to themanufacturer’s protocol. Dual-variable flow cytometricanalysis was done to determine the percentage of apoptoticcells (Annexin V alone–positive cells), necrotic cells(propidium iodide–positive cells), or viable cells (stainingnegative for Annexin V and propidium iodide). All assayswere done on two separate occasions.

ProteasomeActivityAssayCell lysates were prepared, and the fluorogenic peptide

substrate, Suc-Leu-Leu-Val-Tyr-AMC (for the proteasomalchymotrypsin-like activity), was used according to theprocedures described by the 20S Proteasome ActivityAssay kit (Chemicon, Temecula, CA). In brief, control ordrug-treated cells were broken in a lysis buffer [150mmol/L NaCl, 20 mmol/L Tris (pH 7.2), 1% Triton X-100, 1 mmol/L DTT] without protease inhibitors. Total celllysate (50 Ag) was incubated with 20 Amol/L of fluorogenicsubstrate Suc-Leu-Leu-Val-Tyr-AMC at 45jC in 100 ALof assay buffer [25 mmol/L HEPES (pH 7.5), 0.5 mmol/Lof 0.05% NP40, and 0.001% SDS]. Free AMC liberatedby the substrate hydrolysis was quantified for 90 minutesat 1-minute intervals on a microtiter plate fluorometrer(FLUOstar Optima; BMG Labtech, Durham, NC; excitation,355 nm; emission, 460 nm). Preliminary experimentswith control cells indicated that reaction rates were linearfor at least 2 hours. A fluorescence standard curve withknown dilutions of Suc-Leu-Leu-Val-Tyr-AMC was gener-ated. The data were plotted as arbitrary fluorescence units

Bortezomib and Breast Cancer666

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 3: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

versus time, and we obtained the slope of a line fit to thedata using an appropriate linear regression program.Proteasome activity values (% control) were derived bydividing the slope obtained in the presence of bortezomibby the slope obtained in its absence �100. For specificactivity calculations, we used the formula: activity (pmol/min) = slope (arbitrary fluorescence units/min) � conver-sion factor (Ag AMC/arbitrary fluorescence unit) � assayvolume (AL).ImmunofluorescenceAssayBriefly, cells were seeded in 35-mm tissue culture plates

on sterile glass coverslips until subconfluent. After appro-priate treatments, cells were washed with PBS and fixedwith methanol for 1 hour at �20jC. After blocking with 1%(w/v) bovine serum albumin in PBS for 30 minutes at 37jC,cells were incubated with anti-NF-nB p65 antibody (1:180dilution in PBS plus 1% bovine serum albumin) for 2 hoursat 37jC. Controls included buffer alone or nonspecificpurified rabbit immunoglobulin G. Then cells were washedwith PBS and incubated with the secondary antibody Alexa546–coupled goat anti-rabbit IgG (1:1,000 dilution in PBSplus 1% bovine serum albumin) for an additional hour. Theslides were further washed with PBS and then mounted inMowiol (Calbiochem) for fluorescent microscopic exami-nation. Fluorescence confocal and phase images wereacquired using a Leica TCS SL laser scanning confocalspectral microscope (Leica Microsystems, HeilderbergGmbH, Mannheim, Germany). Image assembly and treat-ment were done using the Image Processing Leica ConfocalSoftware.

Western Blot AnalysisFor Western blot assays, cells were cultures in six-well

plates and left untreated or treated as indicated in eachexperiment. Cells were lysed in ice-cold NP40 buffer [1%NP40, 50 mmol/L Tris-HCl (pH 7.4), 150 mmol/L NaCl,5 mmol/L EDTA containing 5 mmol/L NaF, 2 mmol/LNa3VO4, 1 mmol/L phenylmethylsulfonyl fluoride, 5 Ag/mL leupeptine, and 5 Ag/mL aprotinine]. After incubatingfor 30 minutes at 4jC, the samples were centrifuged, andthe supernatant was kept as the NP40-soluble fraction.Unless otherwise specified, Western blot assays were doneusing the abovementioned conditions. In selected experi-ments, the pellet resuspended in SDS buffer [2% SDS, 80mmol/L Tris (pH 6.8), 100 mmol/L DTT, and 10% glycerol]and sonicated constituted the NP40-insoluble fraction, asreported by others (22). Cell extracts (10 Ag/lane) wereseparated by SDS-PAGE and transferred to polyvinylidenedifluoride membranes (Millipore, Bedford, MA). Westernblotting was carried out according to a standard procedureusing horseradish peroxidase–conjugated secondary anti-bodies (Santa Cruz Biotechnology, Santa Cruz, CA). Thefollowing primary antibodies were used: antibodies tophospho-ERK (p-ERK; Thr202/Tyr204), ERK, p-MEK1/2(Ser217/Ser221), MEK1/2, phospho-c-Jun NH2-terminal ki-nase (p-JNK; Thr183/Tyr185), JNK, p-Raf (Ser259), p-AKT(Ser473), AKT, p-EGFR (Tyr1068), EGFR, PI3K (p85), PTEN,and p27 were obtained from Cell Signaling Technology(Beverly, MA). Antibodies to p21 (sc-6246) and MKP-1

(sc-370) were obtained from Santa Cruz Biotechnology.Rabbit polyclonal anti-p-HER-2 (Y1248) was bought fromUpstate Biotechnology (Lake Placid, NY). Mouse mono-clonal anti-HER-2 was from BioGenex (San Ramon, CA).Immunoblotting with h-tubulin mouse monoclonal anti-body (Sigma) was done to confirm equal protein loading.Target proteins were visualized after enhanced chemi-luminescence treatment (Amersham, Piscataway, NJ) ofmembranes and subsequent exposure to X-OMAT X-rayfilm (Sigma).

Statistical AnalysisStatistical analysis was carried out with SPSS version 11.0

(SPSS, Inc, Chicago, IL). To analyze correlations, theunpaired t test was used to determine the statisticaldifferences. Statistical tests were conducted at the two-sided 0.05 level of significance.

ResultsDifferential Effects of Bortezomib on Viability in

Breast Carcinoma Cell LinesThe antiproliferative effects of bortezomib were

assessed by MTT in a panel of six human breast cancercell lines cultured with increasing concentrations ofbortezomib for periods of 24, 48, or 72 hours. Bortezomibcaused a time-dependent and dose-dependent reductionin cell viability in all tested cell lines. An evidentinhibition in cell viability was observed as early as 24hours in some cell lines (data not shown), and a IC50

was achieved after 72 hours of incubation in all of them(Fig. 1A). Bortezomib IC50 F SD varied widely among celllines: 5 F 1.4 nmol/L for SK-BR-3, 5 F 2.4 nmol/L forMDA-MB-468, 7 F 2.7 nmol/L for MDA-MB-231, 100 F8.4 nmol/L for MCF-7, 100 F 12.1 nmol/L for MDA-MB-453, and 1,000 F 14.4 nmol/L for BT-474 cells. Thisdistinction may be of clinical importance because concen-trations of 10 nmol/L are consistent with the one thatresults in proteasome inhibition at therapeutic doses ofbortezomib (4).Searching for potential molecular correlates of the

heterogeneous antiproliferative response to bortezomib,we assayed the basal proteasome activity of each cell line.Protein extracts were prepared under similar cultureconditions used for the MTT assays. Proteasome activitywas consistently higher in BT-474 and MCF-7 cellscompared with the other four cell lines. To assess specificproteasome activity, we did a standard calibration curvewith AMC substrate, and proteasome activity in BT-474and MCF-7 cells was 1.5- to 2-fold higher than in the othercells (Fig. 1B). Thus, a higher proteasome activity wasfound in two of the three cell lines more resistant to thegrowth-inhibitory effect of bortezomib. We also character-ized the expression of EGFR/HER-2 receptors and associ-ated signaling molecules that are commonly dysregulatedin breast cancer (Fig. 1C). No evident relationships wereobserved between total or phosphorylated protein levels ofHER-2, EGFR, ERK1/2, AKT, JNK, or MKP-1 and bortezo-mib IC50s in the studied cell lines.

Molecular Cancer Therapeutics 667

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 4: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

We further studied bortezomib effects on SK-BR-3 andBT-474 cells because they share expression of EGFR andoverexpression of HER-2, whereas their sensitivity tobortezomibwasmarkedly different (Fig. 1A). In cells treatedwith 10 nmol/L bortezomib, effects on cell viability weremarkedlymore pronounced in SK-BR-3 cells comparedwithBT-474 cells (Fig. 2A). The differences on cell viability amongthe two cell lines were statistically significant (P < 0.05, twosided) at all the time points tested. We also assessed byfluorescence-activated cell sorting the effects of bortezomibon cell cycle and on apoptosis. Apoptosis was clearlyinduced in SK-BR-3 cells at 10 nmol/L of bortezomib,whereas effects on BT-474 cells were very slight (Table 1).High concentration and exposure to bortezomib for at least24 hours were required to observe apoptosis in BT-474 cells.On the other hand, bortezomib effects on the cell cycle weremore pronounced in BT-474 cells. At a 10 nmol/L concent-ration, bortezomib induced a moderate cell cycle arrest andcell accumulation in G2-M phase in BT-474 cells (Table 2).The difference in the assays used (MTT and fluorescence-activated cell sorting) and the important role of duration anddose on the effects of bortezomib on the resulting effect oncell proliferation and apoptosis contributed to the fact thatthe magnitude of the effects observed in MTT assays (acomposite effect on cell activation, proliferation, and death)were more pronounced than the proapoptotic effectsobserved by fluorescence-activated cell sorting analysis.

Effects of Bortezomib on Proteasome Activity and onProteasomeSubstratesinSensitiveandResistantCell LinesWe next compared the effects of bortezomib on protea-

some activity in SK-BR-3 versus BT-474 cell lines. Cells

were exposed to various concentrations of bortezomib for2 hours, a time point used to measure bortezomib-inducedproteasome inhibition in clinical studies (9). In both celllines, relative proteasome activity inhibition was similar(Fig. 2B). At 10 nmol/L, relative proteasome activityinhibition was consistent with the level of inhibitionachieved in patients. To study the durability of proteasomeactivity in both cell lines, cells were exposed to bortezomib10 nmol/L for 72 hours. Relative proteasome activityinhibition at this time point was 85.1 F 8.7% in SK-BR-3cells and 59.5 F 28% in BT-474 cells.To test whether the proteasome inhibition induced by

bortezomib resulted in accumulation of well-known pro-teasome substrates, we examined the effects on p21 and p27and, indirectly, on the transcription factor NF-nB. Assaysfor p27 and p21 protein levels were done at 24 hours ofbortezomib exposure. Increased levels of p27 protein weredetected in both SK-BR-3 and BT-474 cells treated atconcentrations of z10 nmol/L (Fig. 2C). Like p27, p21levels were also substantially increased after 24 hours oftreatment in BT-474 cells (Fig. 2C). We were not able todetect basal p21 in SK-BR-3 cells, and no p21 accumulationwas detected following bortezomib exposure. Levels of p21(in BT-474) and p27 (in both cell lines) proteins accumu-lated as early as 2 hours (data not shown) and remainedelevated up to 24 hours of bortezomib exposure.NF-nB/Rel transcription factors is another system

regulated by the proteasome because proteasome inhibi-tion prevents InB degradation and therefore abrogatesNF-nB/p65 nuclear translocation. To test whether borte-zomib exhibited a similar effect on NF-nB in both SK-BR-3

Figure 1. A, sensitivities of breast cancer cells to bortezomib. Cells were seeded into 96-well tissue culture plates (1 � 104 per well) and allowed toattach overnight. Bortezomib (0.01–104 nmol/L) was then added, and cells were allowed to grow for an additional 72 h. Cell viability was assayed usingCellTiter 96 AQueous Non-Radioactive Cell Proliferation Assay kit and expressed as relative to untreated controls cells. Viability for BT-474 (o), SK-BR-3(D), MDA-MB-453 (E), MDA-MB-231(!), MCF-7(.), and MDA-MB-468 (n). Representative of three experiments. Points, means of at least four replicatewells; bars, SD. The final concentration of DMSO never exceeded 0.1%, and this concentration showed no effect on the proliferation of all tested cell lines(data not shown). B, proteasome basal-specific activity in breast cancer cells. Cell extracts (50 Ag) were incubated for 1 h at 37jC with 20 Amol/Lfluorogenic peptide substrate in the absence of bortezomib for the proteasomal chymotrypsin-like activity followed by measurement of free AMC groups.Columns, mean (n =3); bars, SD. C, expression levels of signal transduction molecules in breast cancer cells. Cells lysates were generated, and the levelsof indicated proteins were detected by Western blotting. Simultaneous immunoblotting of tubulin was used as an internal control for equivalent proteinloading. Assays were done twice. Representative results.

Bortezomib and Breast Cancer668

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 5: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

and BT-474 cells, we examined by immunofluorescencethe cellular localization of the NF-nB/p65 subunit in cellsstimulated with TNF-a (a classic activator of NF-nB) withor without bortezomib pretreatment (Fig. 2D). Cells wereincubated with bortezomib for 90 minutes, and thenTNF-a was added for three additional hours. A similardose-dependent inhibition by bortezomib of TNF-a in-duced NF-nB/p65 nuclear translocation was seen in thetwo cell lines.These observations indicated that the effects of borte-

zomib-induced proteasome inhibition on known sub-strates of the proteasome were achieved at similarconcentrations of bortezomib in both cell lines, despite

that viability was affected to a much greater extent inSK-BR-3 cells. These data supported the view that theeffects of bortezomib on cell growth were unrelated (ornot fully related) to differences on proteasome activity inthe two cell lines.

Bortezomib Decreased the Phosphorylation of HER-2,EGFR, and AKTBecause the HER receptor system and its signaling

transduction pathways play an important role in bothSK-BR-3 (bortezomib sensitive) and BT-474 (bortezomibresistant) cells, we investigated the effects of bortezomib onselected protein kinases and phosphatases of this pathway.We initially did concentration-dependent experiments,

Table 1. Percentage of apoptosis induced by bortezomib in SK-BR-3 and BT-474 cells

Bortezomib (nmol/L) SK-BR-3 BT-474

15 h 24 h 48 h 15 h 24 h 48 h

0 4.59 F 0.28 6.33 F 0.15 6.09 F 0.11 0.99 F 0.21 0.98 F 3.2 2.06 F 0.431 5.29 F 1.16 5.26 F 0.21 6.12 F 1.50 0.81 F 0.50 0.83 F 3.2 1.71 F 0.75

10 9.16 F 0.14 17.3 F 0.88 25.1 F 0.96 1.01 F 0.29 1.73 F 3.2 7.55 F 3.21100 9.27 F 1.14 23.5 F 1.96 25.9 F 0.89 1.72 F 0.80 2.71 F 3.2 14.1 F 0.21

NOTE: Results represent means of three experiments F SD.

Figure 2. A, effects of bortezomib on viability of SK-BR-3 and BT-474 cells. Cells were treated with 10 nmol/L bortezomib. At 24, 48, and 72 h, cellviability was measured. Statistical analysis was carried out using Student’s t test with P < 0.05 as significant. B, concentration-dependent inhibition ofproteasome activity by bortezomib in SK-BR-3 and BT-474 cells. Cells were cultured with or without bortezomib at indicated concentrations for 2 h beforemeasurement of proteasome activity; control cells were exposed to an equivalent DMSO. C, accumulation of proteasomal degradation– related proteinsp21Cip1/Waf1 and p27Kip1 by bortezomib in SK-BR-3 and BT-474 cells. Cells were exposed for 24 h to increase concentrations of bortezomib (1, 10, or 100nmol/L). Lysates were prepared and assayed for p21 and p27 protein levels by Western blot; one representative experiment of three. D, nucleartranslocation of NF-nB/p65 in response to treatment with TNF-a is prevented by bortezomib. Cells were left untreated or incubated with bortezomib (1, 10,or 100 nmol/L) for 90 min and then stimulated with 10 ng/mL TNF-a. Four hours after stimulation with TNF-a, cells were fixed, stained with anti-p65antibody, and analyzed using confocal microscopy. a, control cells; b, 10 nmol/L bortezomib; c, TNF-a; d, TNF-a + 1 nmol/L bortezomib; e, TNF-a + 10nmol/L bortezomib; f, TNF-a + 100 nmol/L Bortezomib. Two independent experiments with the same results were done.

Molecular Cancer Therapeutics 669

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 6: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

exposing cells to bortezomib for 24 hours. At concen-trations of z10 nmol/L, total and phosphorylated HER-2protein receptor modestly decreased in SK-BR-3 cells. InBT-474 cells, bortezomib reduced slightly HER-2 phos-phorylation, and effects on total HER-2 receptor wereminor (Fig. 3A). Total levels of EGFR remained essentiallyunchanged in both cell lines during bortezomib exposure.However, in SK-BR-3 cells a marked inhibition of EGFRphosphorylation occurred at bortezomib concentrations ofz10 nmol/L. These effects were less evident in BT-474cells.To assay whether receptor protein changes observed in

cells treated with bortezomib for 24 hours were related toa shift in their subcellular compartments, which mayresult in their presence in detergent-insoluble fractions,we studied receptor protein levels in detergent-solubleand insoluble fractions, following a methodology previ-ously reported by others (22). Bortezomib minimallyattenuated EGFR levels, and accumulation in the deter-gent-insoluble fraction was undetected. In contrast, treat-ment with bortezomib resulted in an evident decline ofHER-2 protein in the detergent (NP40)–soluble fraction,with a concomitant increase in the detergent-insolublefraction (Fig. 3C).We also studied whether bortezomib was able to prevent

ligand-induced EGFR phosphorylation. To this end, weexposed SK-BR-3 and BT-474 cells to EGF in the presence orabsence of bortezomib (Fig. 3D). EGF addition, as expected,induced a marked increase in the phosphorylation of theEGFR and decreased the levels of total EGFR. The decreasein the levels of EGFR is in agreement with the ability of EGFto efficiently induce the degradation of the receptor (23).Pretreatment of cells with bortezomib did not preventEGF-induced receptor phosphorylation and only slightlyprevented total EGFR down-modulation. Under these ex-perimental conditions, we also analyzed the NP40-insolublefractions. EGF addition resulted in down-modulation oftotal EGFR levels without any detected increased in theNP40-insoluble fraction. However, bortezomib pretreat-ment (2 hours) prevented partially EGF-induced down-modulation of total EGFR (Fig. 3D), and EGFR accumulatedin the NP40-insoluble fraction (data not shown).We also examined the effects of bortezomib on the cell

survival PI3K/AKT pathway, which is regulated byHER-2 and EGFR. Total levels of the p85 subunit ofPI3K and the levels of PTEN were unaffected by

bortezomib. A marked suppression of p-AKT was notedat 24 hours in SK-BR-3 cells at bortezomib concentrationsof z10 nmol/L. Inhibition of AKT phosphorylation wasless pronounced in BT-474 cells. Total level of AKTprotein slightly decreased at a concentration of bortezo-mib of 100 nmol/L (Fig. 3A).Time course experiments were then done (Fig. 3B).

HER-2 phosphorylation decreased only at 24 hours oftreatment. Inhibition of EGFR phosphorylation was ob-served at 4 hours (no effects were seen at earlier timepoints; data not shown) and remained inhibited for up to 24hours (latest time point analyzed) in SK-BR-3 cells. AKTphosphorylation was transiently increased in SK-BR-3 cells,but a marked decrease in AKT phosphorylation was notedat 24 hours. Effects in BT-474 cells were less pronounced(Fig. 3B).To study whether proteasome inhibitors other than

bortezomib were more toxic to SK-BR-3 cells than BT-474cells, we assayed cell viability by MTT and the effects onHER-2, EGFR, and AKT in cells treated with the protea-some inhibitor MG-132 (24). In these experiments, SK-BR-3cell viability (IC50 = 0.28 F 0.05 Amol/L) was reduced to agreater extent than BT-474 cell viability (IC50 = 1.5 F 0.08Amol/L; P < 0.05; data not shown). Similarly, the reductionin phosphorylation of HER-2, EGFR, and AKT induced byMG-132 was more pronounced in SK-BR-3 cells comparedwith BT-474 cells (Fig. 3E).

Bortezomib Activated the Raf/MEK/ERK1/2 Pathwayand AffectedMKP1/JNKTo study the effects of bortezomib on the Raf/MEK/

ERK1/2 pathway, which is also regulated by HER-2 andEGFR, we did a series of Western blotting assays under thesame culture conditions as indicated above. At 24 hours ofexposure to bortezomib (z10 nmol/L), there was anincreased phosphorylation of Raf, MEK1/2, and ERK1/2(Fig. 4A), which were more pronounced in SK-BR-3 cellsthan in BT-474 cells. In time course experiments, bortezo-mib-induced phosphorylation of ERK1/2 started 4 hoursafter exposure (Fig. 4B). We also analyzed bortezomibeffects on JNK and MKP-1. The specific protein phospha-tases MKP-1 (or CL100), a proteasome substrate, accumu-lated in both cell lines at concentrations of z10 nmol/L(Fig. 4A), and this was seen at 4 hours of exposure (Fig. 4B).Similar concentration-dependent and time-dependenteffects were seen on p-JNK, without changes in total JNKprotein levels (Fig. 4B).

Table 2. Bortezomib induces accumulation of BT-474 cells in G2

Time (h) G2 (%)

Basal 1 nmol/L Bortezomib 10 nmol/L Bortezomib 100 nmol/L Bortezomib

15 11.2 F 0.71 14.9 F 0.54 28.6 F 1.81 24.7 F 0.8424 11.4 F 0.92 10.2 F 0.13 23.1 F 2.51 27.7 F 1.3248 12.6 F 1.81 12.7 F 0.92 20.6 F 1.02 33.2 F 1.82

NOTE: Results represent means of three experiments F SD.

Bortezomib and Breast Cancer670

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 7: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

Effects on Cell Viability of Combined Exposure ofBortezomib and Pharmacologic Inhibitors of HER Sig-naling PathwaysWe next did a series of MTT assays to study potential

interactions on cell viability between bortezomib andpharmacologic inhibitors of selected signaling moleculesthat were altered during bortezomib treatment. Theseexperiments were done in SK-BR-3 cells due to theirsensitivity to clinically relevant concentrations of bortezo-

mib. The EGFR tyrosine kinase inhibitor AG 1478 reducedSK-BR-3 viability by f50% at 10 Amol/L following 72hours of exposure. A combined treatment with AG1478and bortezomib (both agents used at their estimated IC50s)resulted in an additional 8 F 5% decrease in cell viabilitycompared with each agent used alone. Similar results wereobserved with the PI3K inhibitor LY294002 (used at 40Amol/L, a concentration that resulted in inhibition of AKTphosphorylation; data not shown). Combined treatment

Figure 3. A, Bortezomib dose-dependent effects in EGFR/HER-2 receptors and PI3K/AKT pathway in SK-BR-3 and BT-474 cells. The indicated breastcancer cell lines were exposed for 24 h to increased concentrations of bortezomib (1, 10, or 100 nmol/L). Equal amounts of protein from the cell lysateswere resolved by SDS-PAGE and immunoblotted with a series of antibodies [p-HER-2, total HER-2, p-EGFR, total EGFR, PI3K (p85), PTEN, p-AKT (Ser473),total AKT, tubulin; from top to bottom ]. To detect p-EGFR, long exposures of the film were required. Bortezomib concentration values (above each bottomlane ). Representative of three separate experiments. B, bortezomib time-dependent effects in EGFR/HER-2 receptors and PI3K/AKT pathway in SK-BR-3and BT-474 cells. Exponentially growing SK-BR-3 and BT-474 cells (0 h) were treated with 10 nmol/L of bortezomib for the indicated hours, and thenrepresentative molecules were assayed by Western blot. C, bortezomib effects on EGFR and HER-2 in detergent NP40-insoluble fraction. Followingtreatment of SK-BR-3 at the indicated concentration of bortezomib for 24 h, cells were lysed in 1% NP40 buffer. Insoluble precipitates were resolubilizedwith SDS buffer (2% SDS). Equal amounts of protein were analyzed by Western blotting with anti-EGFR (A ) or anti-HER-2 antibodies (B). Tubulin levelsserved as the control for equal protein loading. NP40 soluble (S ) and insoluble (I ) fractions. Representative of two independent experiments. D, bortezomibeffects on EGF-induced receptor levels and phosphorylation. Cells were serum starved for 24 h and pretreated with the indicated concentrations ofbortezomib or vehicle for 2 h. Cells were challenged for 15 min with EGF (100 ng/mL) or left untreated and lysed. Phosphorylated and total EGFR wereassayed by Western blot. Tubulin is shown as loading control. One of two independent experiments with similar results. E, concentration-dependenteffects of MG-132 in EGFR/HER-2 receptors and AKT in SK-BR-3 and BT-474 cells. Cells were cultured in the absence or presence of varyingconcentrations of MG-132 for 24 h, and protein extracts of cells were analyzed by Western blotting using antibodies as indicated. Tubulin was used as aloading control. Representative of two independent experiments.

Molecular Cancer Therapeutics 671

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 8: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

with both agents further reduced by 10 F 4% cell viabilitywhen compared with LY294002 alone. Finally, exposure ofSK-BR-3 cells to a specific inhibitor of MEK, PD98059 (usedat 40 Amol/L, a concentration that resulted in inhibition ofERK1/2 phosphorylation; data not shown), resulted inincreased cell viability compared with untreated cells. Thisobservation suggested that the hyperactivation state ofRaf/MEK/ERK pathway in bortezomib-treated cells maycontribute to growth inhibition and apoptosis induced bybortezomib in SK-BR-3 cells. Combined treatment withPD98059 did not appreciably increase the effects ofbortezomib on cell viability (data not shown).

DiscussionThe work presented here provides novel information oncellular and molecular effects of bortezomib in breastcancer cells. Bortezomib reduced cell viability in aconcentration-dependent and time-dependent manner inbreast cancer cell lines. Bortezomib’s effects showedmarked heterogeneity in drug responsiveness, rangingfrom highly resistant (IC50 z 100 nmol/L: MCF-7, MDA-MB-453, and BT-474) to highly sensitive (IC50 < 10 nmol/L:MDA-MB-468, MDA-MB-231, and SK-BR-3). Heteroge-neous responses to bortezomib have been also recentlyreported in other tumor types such as prostate cancer (25).

A series of studies indicate that human tumors com-monly have high proteasome expression levels (26). Morerecently, an analysis of transcriptional profiles from f200solid tumors indicated that mRNAs encoding proteasomesubunits were highly coregulated in these cancers (27).These in vivo studies are also supported by findings intumor cell lines. For instance, MCF-7 human breast cancercells express elevated levels of one of the highly conservedproteasome subunits (28). In the present study, basalproteasome activity was 1.5- to 2-fold higher in thebortezomib-resistant MCF-7 and BT-474 cells comparedwith the other cell lines assayed. Although we cannot ruleout that these differences may play a role in the differentialsensitivity to bortezomib, a number of observations suggestthat additional factors play a role in bortezomib effects inbreast cancer cells. For instance, MDA-MB-453 cells had abasal proteasome activity similar to that of sensitive cells,whereas their IC50 was similar to the resistant MCF-7 cells.In addition, the degree of proteasome inhibition achievedduring bortezomib exposure was similar in the tested celllines, as reported in other models (18). Although relativeproteasome inhibition by bortezomib measured at 2 hourswas identical in SK-BR-3 (sensitive) and BT-474 (resistant)cells, the finding of a higher degree of proteasomeinhibition at 72 hours of bortezomib exposure in SK-BR-3cells raises the possibility that a quicker recovery fromproteasome inhibition might contribute to the resistanceof the BT-474 cells to bortezomib compared with SK-BR-3cells.Known proteasome substrates accumulated at a concen-

tration of z10 nmol/L in cells with either relatively high orlow basal proteasome activity. Bortezomib exposure inSK-BR-3 and BT-474 cells was associated with p27 accu-mulation and, in BT-474 cells, with p21 accumulationaccompanied by induction of cell cycle arrest. p27 is amultifunctional protein, which, in addition to its cell cycleregulatory role, is a putative tumor suppressor (a proa-poptotic protein; refs. 29, 30). In certain cancer cell linestreated with bortezomib, stabilization of p27 was observedin the induction of apoptosis (31, 32). In addition, bypretreating cells with bortezomib, the TNF-a-induced NF-nB/p65 translocation to the nucleus was attenuated to asimilar extent in both cell lines. The inhibition of NF-nB bybortezomib is an effect well reported by others (33).One of the best-characterized and clinically relevant

signaling transduction systems in human breast cancer isthe HER tyrosine kinase receptor system (34, 35). The HERreceptor family is composed of four members [HER1 (alsoknown as EGFR), HER-2, HER3, and HER4]. Signaling bythe HER receptor tyrosine kinase family occurs throughseveral downstream pathways, including the Raf/MEK/ERK1/2 and the PI3K/AKT pathways to promote cellproliferation and to inhibit apoptosis (36, 37). However, wedid not observe any relationship between bortezomibeffects on viability and expression/phosphorylation ofHER-2, EGFR, AKT, or ERK1/2. We focused further studiesin SK-BR-3 and BT-474 cell lines because both expresssimilar levels of EGFR and HER-2 and respond similarly to

Figure 4. Bortezomib dose-dependent (A) and time-dependent (B)effects on the Raf/MEK/ERK1/2 and JNK pathways in SK-BR-3 andBT-474 cells. A, the indicated breast cancer cell lines were exposed for24 h to increased concentrations of bortezomib (1, 10, or 100 nmol/L).Equal amounts of protein from the cell lysates were resolved by SDS-PAGEand immunoblotted with a series of antibodies (p-Raf1, p-MEK1/2, totalMEK1/2, p-ERK1/2, total ERK1/2, MKP-1, p-JNK and JNK; from top tobottom ). Antibodies to the phosphorylated form have been used to monitorthe activation state of the enzyme in cell lysates. The level of tubulin wasused as a protein-loading reference control. Bortezomib concentrationvalues (above each bottom lane ). Representative of three separateexperiments. B, exponentially growing SK-BR-3 and BT-474 cells (0 h)were treated with 10 nmol/L of bortezomib for the indicated hours, and thenrepresentative molecules were assayed by Western blot.

Bortezomib and Breast Cancer672

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 9: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

anti-HER-2 (i.e., trastuzumab) or anti-EGFR agents (38, 39),whereas they exhibited markedly different responses tobortezomib. Apoptosis was quickly and strongly inducedin SK-BR-3 cells when exposed to a clinically relevant con-centration of bortezomib, whereas higher concentrationsand a prolonged exposure to bortezomib were required toinduce apoptosis in BT-474 cells. The relative resistance ofBT-474 cells to bortezomib is in agreement with previousreports (40). The finding of similar results with anothercommonly used proteasome inhibitor (MG-132) supportsthe notion that the observed differential sensitivity of thesetwo cell lines was not unique to bortezomib.Bortezomib resulted in a marked inhibition of EGFR

phosphorylation at 4 hours and remained inhibited for upto 24 hours (latest time point analyzed) in SK-BR-3 cells. Inadditional experiments, bortezomib did not affect EGF-induced receptor phosphorylation. Notably, EGF additioninduced EGFR degradation, and this was modestlyprevented by pretreatment with bortezomib (with aconcomitant increase of receptor levels in NP40-insolublefraction). This finding suggests that an intact proteasomefunction is required for the pathway of EGF-induceddegradation of the receptor. Further experiments areneeded to delineate the possibility that EGFR was degradedby lysosomal proteases in addition to the ubiquitin-dependent proteasomal pathway.HER-2 phosphorylation decreased only at 24 hours of

treatment. The levels of total HER-2 also decreased duringbortezomib exposure. The observation of down-modula-tion of HER-2 protein in HER-2-overexpressing cellsexposed to bortezomib was intriguing, in light of theirregulation by the proteasome. However, it should be notedthat it is still unclear to what extent proteasomal processingis involved in down-regulation of receptors. With regard toHER-2, the down-modulation of protein levels observed intreated cells in our experiments is in keeping with findingsby others that showed that proteasome inhibitors (MG-132and bortezomib) repressed HER-2 promoter activity,destabilized mature cytoplasmic HER-2 transcripts, andinduced a decline in HER-2 mRNA and protein levels inHER-2-overexpressing breast cancer cells (MDA-MB-453,SK-BR-3, and BT-474; ref. 40). Our results also showed thatbortezomib treatment reduced the levels of HER-2 in thedetergent (NP40)–soluble fraction. Moreover, we showedthat this was associated to HER-2 recruitment in NP40-insoluble fraction in response to bortezomib. Studies ofHER-2 ubiquitination and intracellular trafficking would berequired to further characterize this finding (23). It istempting to speculate that bortezomib caused HER-2 toaccumulate as ubiquitinated proteins in aggresomes, whichmight then be found in the detergent-insoluble pelletfraction of cell lysates because bortezomib has beenrecently reported to affect ubiquitination (41). However,the down-modulation of HER-2 observed in the solublefraction of cell extracts may explain, at least in part, thecooperative antitumor effects reported by others betweenbortezomib and the anti-HER-2 antibody trastuzumab (thatdown-modulates HER-2 levels; refs. 42, 43) or bortezomib-

and hsp90-targeting agents (44) or HDAC inhibitors (40).With regard to the EGFR, at 24 hours of exposure, weobserved a slight decrease in EGFR levels and noaccumulation of receptor in the detergent insolublefraction. This finding may be related to a limited role ofthe proteasome in EGFR degradation in the absence ofligand activation. Supporting this view, in cells exposed tocycloheximide, to facilitate studies of EGFR degradation byinhibiting protein synthesis, there was no evidence ofconstitutive decrease of total EGFR levels for the timepoints studied (up to 7 hours; ref. 23). Additional experi-ments pointed that the EGFR as such was not a directproteasomal target in the absence of ligand-inducedactivation. These data provide possible explanations toour results of lack of EGFR accumulation during bortezo-mib exposure under our experimental conditions. Bortezo-mib treatment resulted in an early increase of AKTphosphorylation followed by a strong blockade of AKTphosphorylation. This was not associated to parallelchanges in the total AKT or PI3K/p85 or PTEN proteinlevels. Taken together, it is tempting to speculate that theinhibition of AKT phosphorylation is associated to theupstream inhibition of EGFR and HER-2. The degree of theeffects observed on phosphorylation of AKT in SK-BR-3cells was much less evident in BT-474 cells.In contrast to the inhibition of AKT phosphorylation,

bortezomib treatment resulted in an enhanced phosphor-ylation of the Raf/MEK/ERK1/2 pathway. ERK1/2 isactivated by phosphorylation on Tyr185 and Thr183 by adual specificity kinase MEK, which can be activated byRaf-1. Raf-1 activation can be achieved by the GTP-boundactivated form of Ras. An increased phosphorylation ofERK1/2 occurred at 4 hours of bortezomib exposure andremained sustained throughout the experiment. Moreover,these effects were associated with stimulation of MEK1/2and Raf phosphorylation, thus showing that upstreamcomponents of the ERK signaling cascade participate in thisprocess. As observed for EGFR, HER-2, and AKT, themagnitude of the effects on Raf/MEK/ERK1/2 weregreater in SK-BR-3 cells than in BT-474 cells. Phosphory-lation of JNK, a common mediator of bortezomib cell kill,was also observed (45). MKP-1 levels were increasedfollowing bortezomib treatment, in agreement with resultsrecently reported by Small et al. (45). MKP-1 belongs to afamily of inducible nuclear dual-specificity phosphatasesexerting catalytic activity to phosphotyrosine-containingand phosphothreonine-containing proteins. However, itseems not to be a direct link between the increase in MKP-1and the decline in p-HER-2, p-EGFR, and p-AKT afterbortezomib treatment, because in our time-response stud-ies, MKP-1 increase preceded the decline in the phosphor-ylation of HER-2, EGFR, and AKT. This view is furthersupported by the fact that these molecules are not knownsubstrates for MKP-1. MKP-1 is known to inactivate ERK1/2 (among other ERKs); however, in our assays, we couldnot find an association between MKP-1 accumulationand ERK1/2 phosphorylation. Proteasome inhibitor–mediated induction of MKP-1 has been reported to have

Molecular Cancer Therapeutics 673

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 10: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

an antiapoptotic role. We observed that MKP-1 was para-doxically induced more strongly by bortezomib in SK-BR-3bortezomib-sensitive cells than in the BT-474 bortezomib-resistant cells. The potential role of MKP-1 accumulationinduced by bortezomib in the sensitivity of SK-BR-3 andBT-474 cells to this agent remains to be characterized.Notably, in other cell lines tested, such as MCF-7 and

MDA-MB-231, ERK1/2 phosphorylation decreased duringbortezomib exposure (data not shown). This later observa-tion is in agreement with the findings observed with othernonclinical proteasome inhibitors in MDA-MB231 cells (46)or in other tumor systems (47). The reasons behind theopposite effects on ERK1/2 between the HER-2 over-expressing cells (i.e., ERK1/2 activation in SK-BR-3 and BT-474) and the non–HER-2-overexpressing cells (i.e., ERK1/2inhibition in MDA-MB-231 and MCF-7) are yet uncharac-terized. In general, Raf/MEK/ERK1/2 pathway activationis coupled with cell proliferation and survival (48),although a prolonged activation may exert a proapoptoticinfluence depending upon the cellular context (36, 49).Here, exposure of SK-BR-3 cells to PD98059, a specificinhibitor of MEK, resulted in increased cell viability. Thus,pharmacologic blockade of MEK and the resulting inhibi-tion of ERK1/2 phosphorylation were possibly associatedto the release of proapoptotic stimuli to the cells. Thisobservation suggests, indirectly, that the hyperactivationstate of the Raf/MEK/ERK1/2 pathway observed inbortezomib-treated SK-BR-3 cells may contribute to growthinhibition and apoptosis in SK-BR-3 cells. Interestingly,combined treatments with bortezomib and PD98059 inthese cells resulted in an antitumor effect similar to thatachieved with bortezomib alone. Collectively, these resultsshow that bortezomib has distinct and opposite effects onthe phosphorylation of EGFR, HER-2, and AKT (suppres-sion) and of the Raf/MEK/ERK1/2 pathway (activation) inHER-2-overexpressing breast cancer cells. Such effects weremore pronounced in SK-BR-3 than in BT-474 cells. Whethersuch molecular effects play a role in the response tobortezomib in breast cancer cells or reflects differingdegrees of residual proteasome activity is yet unknown.In conclusion, the data presented here provide novel

evidence for differential cellular and molecular effects ofbortezomib in a panel of human breast cancer cells. Theobserved molecular effects of bortezomib on importantsignaling transduction molecules regulated by the HERreceptor system may be relevant to help to designmechanistic-based combination treatments against breastcancer. Future studies on other proteasome inhibitors thatact on similar and different parts of the proteasomepathway might be important for delineating the role ofthis pathway in mediating anticarcinogenic responses. Theresults presented here show differential responses that aredependent on cell context, and their interaction with otheragents were also variable. These results illustrate thecomplex mechanism of bortezomib action. Mechanisticstudies will be needed to assess the role of the proteinsaffected by bortezomib reported here in the antitumoreffects of this agent.

References

1. Zwickl P, Voges D, Baumeister W. The proteasome: a macromolecularassembly designed for controlled proteolysis. Philos Trans R Soc Lond BBiol Sci 1999;354:1501–11.

2. Ciechanover A. The ubiquitin-proteasome pathway: on protein deathand cell life. EMBO J 1998;17:7151–60.

3. Adams J. The proteasome: a suitable antineoplastic target. Nat RevCancer 2004;4:349–60.

4. Adams J, Palombella V, Sausville A, et al. Proteasome inhibitors: anovel class of potent and effective antitumor agents. Cancer Res 1999;59:2615–22.

5. Ross JS, Schenkein DP, Pietrusko R, et al. Targeted therapies forcancer 2004. Am J Clin Pathol 2004;122:598–609.

6. Papandreou C, Daliani D, Millikan RE, et al. Phase I study of intravenous(I.V.) proteasome inhibitor PS-341 in patients (pts) with advancedmalignancies. Proc Am Soc Clin Oncol 2001;20:86a.

7. Lightcap ES, McCormack TA, Pien CS, Chau V, Adams J, Elliott PJ.Proteasome inhibition measurements: clinical application. Clin Chem2000;46:673–83.

8. Cortes J, Thomas D, Koller C, et al. Phase I study of bortezomib inrefractory or relapsed acute leukemias. Clin Cancer Res 2004;10:3371–6.

9. Papandreou CN, Daliani DD, Nix D, et al. Phase I trial of the proteasomeinhibitor bortezomib in patients with advanced solid tumors withobservations in androgen-independent prostate cancer. J Clin Oncol2004;22:2108–21.

10. Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study ofbortezomib in relapsed, refractory myeloma. N Engl J Med 2003;348:2609–17.

11. Bross PF, Kane R, Farrell AT, et al. Approval summary for bortezomibfor injection in the treatment of multiple myeloma. Clin Cancer Res 2004;10:3954–64.

12. Rajkumar SV, Richardson PG, Hideshima T, Anderson KC. Protea-some inhibition as a novel therapeutic target in human cancer. J Clin Oncol2005;23:630–9.

13. Orlowski RZ, Dees EC. The role of the ubiquitination-proteasomepathway in breast cancer: applying drugs that affect the ubiquitin-proteasome pathway to the therapy of breast cancer. Breast Cancer Res2003;5:1–7.

14. Cardoso F, Ross JS, Picart MJ, Sotiriou C, Durbecq V. Targeting theubiquitin-proteasome pathway in breast cancer. Clin Breast Cancer 2004;5:148–57.

15. Brown J, Von Roenn J, O’Regan RM, et al. A phase II study of theproteasome inhibitor PS-341 in patients (pts) with metastatic breastcancer (MBC). J Clin Oncol (Post-Meeting Edition) 2004;22:abstract 546.

16. Cristofanilli M, Gonzalez-Angulo AM, Nealy KN, et al. A phase II trialof PS-341 in metastatic breast cancer (MBC). J Clin Oncol (Post-MeetingEdition) 2004;22:abstract 3102.

17. Pierson-Mullany LK, Skildum A, Faivre E, Lange CA. Cross-talkbetween growth factor and progesterone receptor signaling pathways:implications for breast cancer cell growth. Breast Dis 2003;18:21–31.

18. Nawrocki ST, Sweeney-Gotsch B, Takamori R, McConkey DJ. Theproteasome inhibitor bortezomib enhances the activity of docetaxel inorthotopic human pancreatic tumor xenografts. Mol Cancer Ther 2004;3:59–70.

19. Albanell J, Baselga J, Guix M, et al. A phase I dose-escalation andpharmacodynamic study of the proteasome inhibitor bortezomib (VELCA-DETM) with docetaxel in anthracycline-pretreated advanced breast cancer[abstract 63]. Proc Am So Clin Oncol 2003;22:16.

20. Durbecq V, Laes J-F, Lagneaux L, et al. Involvement of NFkB and P27pathways in the synergistic apoptosis induction by the proteasomeinhibitor bortezomib (B) (VELCADE2) and trastuzumab (T) (HerceptinR)in HER-2 overexpressing breast cancer cell lines. Proc Am Assoc CancerRes 2004;45:157.

21. Goodwin CJ, Holt SJ, Downes S, Marshall NJ. Microculturetetrazolium assays: a comparison between two new tetrazolium salts,XTT and MTS. J Immunol Methods 1995;179:95–103.

22. Nimmanapalli R, Fuino L, Bali P, et al. Histone deacetylase inhibitorLAQ824 both lowers expression and promotes proteasomal degradation ofBcr-Abl and induces apoptosis of imatinib mesylate-sensitive or -refractorychronic myelogenous leukemia-blast crisis cells. Cancer Res 2003;63:5126–35.

Bortezomib and Breast Cancer674

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 11: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

23. Longva KE, Blystad FD, Stang E, Larsen AM, Johannessen LE,Madshus IH. Ubiquitination and proteasomal activity is required fortransport of the EGF receptor to inner membranes of multivesicularbodies. J Cell Biol 2002;156:843–54.

24. Yoshida T, Shiraishi T, Nakata S, et al. Proteasome inhibitor MG132induces death receptor 5 through CCAAT/enhancer-binding proteinhomologous protein. Cancer Res 2005;65:5662–7.

25. Williams S, Pettaway C, Song R, Papandreou C, Logothetis C,McConkey DJ. Differential effects of the proteasome inhibitor bortezomibon apoptosis and angiogenesis in human prostate tumor xenografts. MolCancer Ther 2003;2:835–43.

26. Kanayama H, Tanaka K, Aki M, et al. Changes in expressions ofproteasome and ubiquitin genes in human renal cancer cells. Cancer Res1991;51:6677–85.

27. Mulligan G, D’Cruz C, Palermo A, et al. Elevated expression of theproteasome in cancer. Proc Am Soc Clin Oncol 2002;21:424a.

28. Ren S, Smith MJ, Louro ID, et al. The p44S10 locus, encoding asubunit of the proteasome regulatory particle, is amplified duringprogression of cutaneous malignant melanoma. Oncogene 2000;19:1419–27.

29. Lloyd RV, Erickson LA, Jin L, et al. p27kip1: a multifunctional cyclin-dependent kinase inhibitor with prognostic significance in human cancers.Am J Pathol 1999;154:313–23.

30. St Croix B, Florenes VA, Rak JW, et al. Impact of the cyclin-dependent kinase inhibitor p27Kip1 on resistance of tumor cells toanticancer agents. Nat Med 1996;2:1204–10.

31. Wu Y, Luo H, Kanaan N, Wu J. The proteasome controls theexpression of a proliferation-associated nuclear antigen Ki-67. J CellBiochem 2000;76:596–604.

32. Adams J, Behnke M, Chen S, et al. Potent and selective inhibitors ofthe proteasome: dipeptidyl boronic acids. Bioorg Med Chem Lett 1998;8:333–8.

33. An J, Sun Y, Fisher M, Rettig MB. Maximal apoptosis of renal cellcarcinoma by the proteasome inhibitor bortezomib is nuclear factor-kappaB dependent. Mol Cancer Ther 2004;3:727–36.

34. Albanell J, Baselga J. The ErbB receptors as targets for breast cancertherapy. J Mammary Gland Biol Neoplasia 1999;4:337–51.

35. Yarden Y. Biology of HER-2 and its importance in breast cancer.Oncology 2001;61:1–13.

36. Marshall CJ. Specificity of receptor tyrosine kinase signaling:transient versus sustained extracellular signal-regulated kinase activation.Cell 1995;80:179–85.

37. Thompson JE, Thompson CB. Putting the rap on AKT. J Clin Oncol2004;22:4217–26.

38. Lewis GD, Figari I, Fendly B, et al. Differential responses of humantumor cell lines to anti-p185HER-2 monoclonal antibodies. CancerImmunol Immunother 1993;37:255–63.

39. Anido J, Matar P, Albanell J, et al. ZD1839, a specific epidermalgrowth factor receptor (EGFR) tyrosine kinase inhibitor, induces theformation of inactive EGFR/HER-2 and EGFR/HER3 heterodimers andprevents heregulin signaling in HER-2-overexpressing breast cancer cells.Clin Cancer Res 2003;9:1274–83.

40. Scott G, Marx C, Amend C, Benz C. Proteasome and histonedeacetylase inhibitors repress ErbB2 transcript levels and additively inhibitbreast cancer growth. Proc Am Soc Clin Oncol 2004;45:abstract 2447.

41. Xu Q, Farah M, Webster JM, Wojcikiewicz RJ. Bortezomib rapidlysuppresses ubiquitin thiolesterification to ubiquitin-conjugating enzymesand inhibits ubiquitination of histones and type I inositol 1,4,5-tri-sphosphate receptor. Mol Cancer Ther 2004;3:1263–9.

42. Sliwkowski MX, Lofgren JA, Lewis GD, Hotaling TE, Fendly BM, FoxJ. Nonclinical studies addressing the mechanism of action of trastuzumab(HerceptinR). Semin Oncol 1999;26:60–70.

43. Albanell J, Codony J, Rovira A, Mellado B, Gascon P. Mechanism ofaction of anti-HER-2 monoclonal antibodies: scientific update on trastu-zumab and 2C4. Adv Exp Med Biol 2003;532:253–68.

44. Mimnaugh EG, Xu W, Vos M, et al. Simultaneous inhibition of hsp 90and the proteasome promotes protein ubiquitination, causes endoplasmicreticulum-derived cytosolic vacuolization, and enhances antitumor activ-ity. Mol Cancer Ther 2004;3:551–66.

45. Small GW, Shi YY, Edmund NA, Somasundaram S, Moore DT,Orlowski RZ. Evidence that mitogen-activated protein kinase phosphatase-1 induction by proteasome inhibitors plays an antiapoptotic role. MolPharmacol 2004;66:1478–90.

46. Orlowski RZ, Small GW, Shi YY. Evidence that inhibition of p44/42mitogen-activated protein kinase signaling is a factor in proteasomeinhibitor-mediated apoptosis. J Biol Chem 2002;277:27864–71.

47. Yu C, Rahmani M, Conrad D, Subler M, Dent P, Grant S. Theproteasome inhibitor bortezomib interacts synergistically with histonedeacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive andresistant to STI571. Blood 2003;102:3765–74.

48. Albanell J, Codony-Servat J, Rojo F, et al. Activated extracellularsignal-regulated kinases: association with epidermal growth factorreceptor/transforming growth factor alpha expression in head and necksquamous carcinoma and inhibition by anti-epidermal growth factorreceptor treatments. Cancer Res 2001;61:6500–10.

49. Chen JR, Plotkin LI, Aguirre JI, et al. Transient versus sustainedphosphorylation and nuclear accumulation of ERKs underlie anti-versuspro-apoptotic effects of estrogens. J Biol Chem 2005;280:4632–8.

Molecular Cancer Therapeutics 675

Mol Cancer Ther 2006;5(3). March 2006

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Page 12: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

Article on bortezomib andbreast cancer

In the article on bortezomib and breast cancer in theMarch 2006 (1) issue, the grant support footnote wasomitted. The appropriate grant support information isbelow.

Grant support: SAF 2003-08181 (Spanish Science andTechnology Ministry, MCYT), GEN2003-20243-C08-08 andAsociacion Espanola Contra el Cancer (AECC)/Catalunacontra el Cancer 2002; "Premi Fi de Residencia 2003-2004"research grant from the Hospital Clınic of Barcelona,Spain (J. Domingo-Domenech); fellowship from la Funda-cion Cientıfica de la Asociacion Espanola contra elCancer (AECC; to A. Rovira); and Fundacio Cellex,Barcelona (generous grant to the Laboratory of ExperimentalOncology).

Reference

1. Codony-Servat J, Tapia MA, Bosch M, et al. Differential cellular andmolecular effects of bortezomib, a proteasome inhibitor, in human breastcancer cells. Mol Cancer Ther 2006;5:665–75.

Copyright C 2006 American Association for Cancer Research.

doi:10.1158/1535-7163.MCT-5-5-COR1

Correction

1383

Mol Cancer Ther 2006;5(5). May 2006

Page 13: Differential cellular and molecular effects of bortezomib ... · SK-BR-3 cells, a marked inhibition of EGFR, HER-2, and AKT phosphorylation was observed at a clinically relevant concentration

2006;5:665-675. Mol Cancer Ther   Jordi Codony-Servat, Maria A. Tapia, Marta Bosch, et al.   proteasome inhibitor, in human breast cancer cellsDifferential cellular and molecular effects of bortezomib, a

  Updated version

  http://mct.aacrjournals.org/content/5/3/665

Access the most recent version of this article at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/5/3/665.full#ref-list-1

This article cites 49 articles, 23 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/5/3/665.full#related-urls

This article has been cited by 20 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://mct.aacrjournals.org/content/5/3/665To request permission to re-use all or part of this article, use this link

on June 13, 2020. © 2006 American Association for Cancer Research. mct.aacrjournals.org Downloaded from