adaptation of energy metabolism in breast cancer brain ... · nates breast cancer spreading to the...

16
Adaptation of Energy Metabolism in Breast Cancer Brain Metastases Emily I. Chen, 1,2 Johannes Hewel, 1 Joseph S. Krueger, 2 Claire Tiraby, 1 Martin R. Weber, 2 Anastasia Kralli, 1 Katja Becker, 3 John R. Yates III, 1 and Brunhilde Felding-Habermann 2 Departments of 1 Cell Biology and 2 Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California and 3 Nutritional Biochemistry, Interdisciplinary Research Center, Justus-Liebig University Giessen, Giessen, Germany Abstract Brain metastases are among the most feared complications in breast cancer, as no therapy exists that prevents or elimi- nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties that allow breast cancer cell growth within the brain tissue. To provide information in this direction, we established a human breast cancer cell model for brain metastasis based on circulating tumor cells from a breast cancer patient and variants of these cells derived from bone or brain lesions in immunodeficient mice. The brain-derived cells showed an increased potential for brain metastasis in vivo and exhibited a unique protein expression profile identified by large-scale proteomic analysis. This protein profile is consistent with either a selection of predisposed cells or bioenergetic adaptation of the tumor cells to the unique energy metabolism of the brain. Increased expression of enzymes involved in glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation pathways suggests that the brain metastatic cells derive energy from glucose oxidation. The cells further showed enhanced activation of the pentose phosphate pathway and the glutathione system, which can minimize production of reactive oxygen species resulting from an enhanced oxidative metabolism. These changes promoted resistance of brain metastatic cells to drugs that affect the cellular redox balance. Importantly, the metabolic alterations are associated with strongly enhanced tumor cell survival and proliferation in the brain microenvironment. Thus, our data support the hypothesis that predisposition or adaptation of the tumor cell energy metabolism is a key element in breast cancer brain metastasis, and raise the possibility of targeting the functional differentiation in breast cancer brain lesions as a novel therapeutic strategy. [Cancer Res 2007;67(4):1472–86] Introduction Brain metastases are the most feared complication in breast cancer. Nearly 20% of patients with advanced breast cancer are eventually diagnosed with brain lesions, making breast tumors the main source of metastatic brain disease in women (1–3). The incidence of brain metastases increases as patients live longer in response to improved cancer therapy, but no current regimen significantly affects breast cancer brain metastases. Palliative treatment extends survival in most cases for only a few weeks or months, but often with severe impact on the quality of life (3–5). Therefore, it is imperative to gain a better understanding of the nature and functionality of breast cancer cells that cause brain metastases for development of effective regimens to prevent and control this stage of the disease. In the past, much attention focused on invasive properties of metastatic cancer cells and their ability to attract new blood vessels (6–8). However, intrinsic mechanisms allowing metastatic tumor cells to survive and proliferate in preferred target organs are poorly understood. Thus, the goal of this study was to analyze protein expression profiles of tumor cells from breast cancer brain metastases and gain insight into cellular properties that promote tumor cell survival in the unique brain microenviron- ment. Metastatic breast cancer cells colonize the brain mainly from the bloodstream (9). We therefore isolated circulating tumor cells from a stage IV breast cancer patient, reintroduced the cells into the bloodstream of immunodeficient mice, and recovered tumor cells from the brain, or long bone for comparison. To define determinants of breast cancer cell growth in the brain, we examined the protein expression profiles of the parental cell line and its brain or bone homing variants by multidimensional proteomic analysis, MudPIT (10). More than 300 proteins were found uniquely regulated in the brain metastatic cells. Most of these proteins are involved in cellular metabolism and cell stress response. Our proteomic results, transcriptional validation, and cell function analyses in vitro indicate that brain metastatic cells use enhanced mitochondrial respiratory pathways for energy production and antioxidant defense mechanisms. The metabolic changes identified in the brain metastatic cells may reflect a predisposition or adaptation of the tumor cells to the brain microenvironment where a constant high energy demand is met almost entirely by glucose oxidation (11–14). Our results provide new evidence that brain metastatic breast cancer cells use energy metabolism pathways that are distinct from anaerobic glycolysis, which is predominant in most cancer cells in oxygen- poor tumor microenvironments. Furthermore, the redox state of brain metastatic breast cancer cells may provide a link between their energy metabolism and gene regulation. Our results show the significance of understanding the metabolic state of metastatic cells and support the rationale of targeting tumor energy metabolism as a potential therapy for breast cancer brain metastasis. Materials and Methods Real-time PCR primers. Real-time PCR was done with the following gene-specific primers: Requests for reprints: Emily I. Chen, E-mail: [email protected]; or Brunhilde Felding-Habermann, Department of Molecular and Experimental Medicine, Scripps Research Institute, MEM 175, La Jolla, CA 92037. Phone: 858-784-2021; Fax: 858-784- 2030; E-mail: [email protected]. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-06-3137 Cancer Res 2007; 67: (4). February 15, 2007 1472 www.aacrjournals.org Research Article Research. on October 15, 2020. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 02-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

Adaptation of Energy Metabolism in Breast Cancer

Brain Metastases

Emily I. Chen,1,2Johannes Hewel,

1Joseph S. Krueger,

2Claire Tiraby,

1Martin R. Weber,

2

Anastasia Kralli,1Katja Becker,

3John R. Yates III,

1and Brunhilde Felding-Habermann

2

Departments of 1Cell Biology and 2Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California and3Nutritional Biochemistry, Interdisciplinary Research Center, Justus-Liebig University Giessen, Giessen, Germany

Abstract

Brain metastases are among the most feared complicationsin breast cancer, as no therapy exists that prevents or elimi-nates breast cancer spreading to the brain. New therapeuticstrategies depend on specific knowledge of tumor cellproperties that allow breast cancer cell growth within thebrain tissue. To provide information in this direction, weestablished a human breast cancer cell model for brainmetastasis based on circulating tumor cells from a breastcancer patient and variants of these cells derived from boneor brain lesions in immunodeficient mice. The brain-derivedcells showed an increased potential for brain metastasisin vivo and exhibited a unique protein expression profileidentified by large-scale proteomic analysis. This proteinprofile is consistent with either a selection of predisposedcells or bioenergetic adaptation of the tumor cells to theunique energy metabolism of the brain. Increased expressionof enzymes involved in glycolysis, tricarboxylic acid cycle, andoxidative phosphorylation pathways suggests that the brainmetastatic cells derive energy from glucose oxidation. Thecells further showed enhanced activation of the pentosephosphate pathway and the glutathione system, which canminimize production of reactive oxygen species resultingfrom an enhanced oxidative metabolism. These changespromoted resistance of brain metastatic cells to drugs thataffect the cellular redox balance. Importantly, the metabolicalterations are associated with strongly enhanced tumor cellsurvival and proliferation in the brain microenvironment.Thus, our data support the hypothesis that predispositionor adaptation of the tumor cell energy metabolism is a keyelement in breast cancer brain metastasis, and raise thepossibility of targeting the functional differentiation in breastcancer brain lesions as a novel therapeutic strategy. [CancerRes 2007;67(4):1472–86]

Introduction

Brain metastases are the most feared complication in breastcancer. Nearly 20% of patients with advanced breast cancer areeventually diagnosed with brain lesions, making breast tumorsthe main source of metastatic brain disease in women (1–3). Theincidence of brain metastases increases as patients live longer inresponse to improved cancer therapy, but no current regimen

significantly affects breast cancer brain metastases. Palliativetreatment extends survival in most cases for only a few weeks ormonths, but often with severe impact on the quality of life (3–5).Therefore, it is imperative to gain a better understanding of thenature and functionality of breast cancer cells that cause brainmetastases for development of effective regimens to prevent andcontrol this stage of the disease.In the past, much attention focused on invasive properties of

metastatic cancer cells and their ability to attract new bloodvessels (6–8). However, intrinsic mechanisms allowing metastatictumor cells to survive and proliferate in preferred target organsare poorly understood. Thus, the goal of this study was to analyzeprotein expression profiles of tumor cells from breast cancerbrain metastases and gain insight into cellular properties thatpromote tumor cell survival in the unique brain microenviron-ment. Metastatic breast cancer cells colonize the brain mainlyfrom the bloodstream (9). We therefore isolated circulating tumorcells from a stage IV breast cancer patient, reintroduced the cellsinto the bloodstream of immunodeficient mice, and recoveredtumor cells from the brain, or long bone for comparison. Todefine determinants of breast cancer cell growth in the brain, weexamined the protein expression profiles of the parental cell lineand its brain or bone homing variants by multidimensionalproteomic analysis, MudPIT (10). More than 300 proteins werefound uniquely regulated in the brain metastatic cells. Most ofthese proteins are involved in cellular metabolism and cell stressresponse. Our proteomic results, transcriptional validation, andcell function analyses in vitro indicate that brain metastatic cellsuse enhanced mitochondrial respiratory pathways for energyproduction and antioxidant defense mechanisms. The metabolicchanges identified in the brain metastatic cells may reflect apredisposition or adaptation of the tumor cells to the brainmicroenvironment where a constant high energy demand is metalmost entirely by glucose oxidation (11–14). Our results providenew evidence that brain metastatic breast cancer cells useenergy metabolism pathways that are distinct from anaerobicglycolysis, which is predominant in most cancer cells in oxygen-poor tumor microenvironments. Furthermore, the redox state ofbrain metastatic breast cancer cells may provide a link betweentheir energy metabolism and gene regulation. Our results showthe significance of understanding the metabolic state ofmetastatic cells and support the rationale of targeting tumorenergy metabolism as a potential therapy for breast cancer brainmetastasis.

Materials and Methods

Real-time PCR primers. Real-time PCR was done with the following

gene-specific primers:

Requests for reprints: Emily I. Chen, E-mail: [email protected]; or BrunhildeFelding-Habermann, Department of Molecular and Experimental Medicine, ScrippsResearch Institute, MEM 175, La Jolla, CA 92037. Phone: 858-784-2021; Fax: 858-784-2030; E-mail: [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-06-3137

Cancer Res 2007; 67: (4). February 15, 2007 1472 www.aacrjournals.org

Research Article

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

Breast cancer cell model. BCM2 Parent cells were established

from blood of a stage IV breast cancer patient with widespread metastasis

and cultured in EMEM with 10% fetal bovine serum (FBS) after isola-tion with antiepithelial immunomagnetic beads (monoclonal antibody

BerEP4; Dynal, Lake Success, NY). BCM2 parent cells (2.5 � 105) were i.v.

injected into 5- to 6-week-old female severe combined immunodeficient

(SCID) mice (C.B17/lcTac-Prkdc scid) and tumor cells were recovered fromthe brain (BCM2 BrainG1) or femur (BCM2 Bone) 6 weeks later. BCM2

BrainG2 cells were from a brain lesion after i.v. injection of BCM2 BrainG1

cells. For in vivo tracking, the cell lines were transduced with fireflyluciferase (F-luc) in a lentiviral vector system with cytomegalovirus

promoter, injected i.v. into female SCID mice and followed by repeated

noninvasive bioluminescence imaging with an IVIS 200 system (Xenogen,

Alameda, CA). For ex vivo organ imaging, mice were injected i.p. with100 mg/kg D-luciferin 5 min before necropsy and the excised organs were

incubated with 100 Ag/mL D-luciferin (5 min). Bioluminescence signal was

quantified as photons per second per square centimeter in defined regions

of interest using Living Image software (Xenogen). Where appropriate,signal was normalized to the bioluminescence expression of each cell

variant. Background luminescence in vivo was 1 � 105 to 2 � 105 photons/s.

To measure tumor cell growth in the brain directly, 1 � 104 tumor cells in

2 AL were implanted into the forebrain of female SCID mice (2 mm lateral,1 mm anterior to bregma, 3-mm depth from dura). All animal work was in

accordance with The Scripps Research Institute Animal Resources

(Association for Assessment and Accreditation of Laboratory Animal Careaccredited).

Protein fractionation and preparation. BCM2 Parent, BCM2 Bone,

and BCM2 BrainG1 cells were seeded at the same density and harvested

at f80% confluency. Whole-cell protein extracts were prepared with theTotalProteinExtraction kit (BioChain, Hayward, CA) and protein concen-

tration was determined by bicinchoninic acid (BCA) assay (Pierce,

Rockford, IL). For each cell line, 1 mg of total lysate was resuspended

in starting buffer following the buffer exchange method of the Proteo-meLab PF2D kit from Beckman Coulter (Fullerton, CA). Cell lysates

were resolved based on their isoelectric point (pI) using the method of

Beckman Coulter (15) and fractions collected in intervals of 0.3 pH unitusing the chromatofocusing separation of the ProteomeLab PF2D system.

Eleven fractions were generated for each cell line, and each fraction

was precipitated with trichloroacetic acid/acetone before in-solution

digest with trypsin. Protein pellets from each fraction were resuspendedin trypsin digestion buffer (50 mmol/L ammonium bicarbonate + 0.1%

Rapigest; Waters Corp., Milford, MA) and digested with trypsin overnight

at 37jC.Multidimensional chromatography and tandem mass spectrometry.

Peptide mixtures were resolved by strong cation exchange liquid chroma-

tography upstream of reverse-phase liquid chromatography as described

(16). Eluted peptides were electrosprayed directly into an LTQ ion trap

mass spectrometer equipped with a nano-liquid chromatography elec-trospray ionization source (ThermoFinnigan, San Jose, CA). Full mass

spectra were recorded over a 400 to 1,600 m/z range, followed by three

tandem mass spectrometry (MS/MS) events sequentially generated in adata-dependent manner on the first, second, and third most intense

ions selected from the full MS spectrum (at 35% collision energy). Mass

spectrometer scan functions and high-performance liquid chromatography

solvent gradients were controlled by the Xcalibur data system (Thermo-

Finnigan).

Interpretation of MS/MS data sets. SEQUEST (17) was used to matchMS/MS spectra. The validity of peptide/spectrum matches was assessed

using SEQUEST-defined parameters, the cross-correlation score (XCorr) and

normalized difference in cross-correlation scores (DCn).Distribution of XCorr and DCn values for (a) direct and (b) decoy

database hits was obtained, and the two subsets were separated by

quadratic discriminant analysis. Full separation of the direct and decoy

subsets is not generally possible. Therefore, the discriminant score wasset such that a false-positive rate of 5% was determined based on the

number of accepted decoy database peptides. This procedure was

independently done on data subsets for charge states +1, +2, and +3.

In addition, spectra were only retained if they had a DCn of at least 0.08and minimum XCorr of 1.8 for +1, 2.5 for +2, and 3.5 for +3 spectra. In

addition, the minimum sequence length was seven amino acid residues.

DTASelect (18) was used to select and sort peptide/spectrum matches

passing this set of criteria. The human protein database used wasdownloaded from the International Protein Index (IPI) version 3.12 in

November 2005 and reverse protein sequences of the IPI database were

used as the decoy database. Peptide hits from multiple runs were compared

using CONTRAST (18). Proteins were considered detected if they wereidentified by at least half tryptic status and more than two peptides. A

semiquantitative comparison of protein expression was obtained based on

the ratio of spectra counts (the number of spectra collected per protein;refs. 16, 19).

Western blot analysis. Whole-cell protein extracts were prepared with

the TotalProteinExtraction kit (BioChain) and protein concentrations

determined by BCA assay (Pierce). Total cell lysates (50 Ag) were used tocompare protein expression. Immunoblotting assays were carried out by

standard procedures using AMP-activated protein kinase (AMPK)-a and

phospho-AMPK-a(Thr172) antibodies from Cell Signaling Technology

(Beverly, MA); anti–h-actin was used for equal loading control (Sigma, St.Louis, MO). Bands were detected using horseradish peroxidase–labeled

secondary antibodies and enhanced chemiluminescence (Amersham

Pharmacia, Piscataway, NJ).Determination of cellular ATP. Cellular ATP levels were measured in

96-well plates with the CellTiter Glo luminescence ATP assay (Promega)

according to the manufacturer’s instructions. Cells (2.5 � 104 per well)

in growth medium containing 10% FBS were seeded in quadruplicatesand the assays repeated thrice with similar results. The cells in this

assay were not genetically tagged with luciferase. ATP-dependent lumi-

nescence was related to an ATP standard curve ranging from 10 nmol/L

to 1 Amol/L.Real-time quantitative PCR. Changes in mRNA expression of identified

metabolic enzymes were examined by real-time PCR after reverse

transcription of 400-ng RNA from each sample with SuperScript II reverse

transcriptase (Invitrogen Life Technologies, Inc., Carlsbad, CA). cDNAwas diluted 20-fold before PCR amplification using a 2720 Thermal Cycler

and SYBR Green mix (Applied Biosystems, Foster City, CA). A typical

protocol involved 10-min denaturation at 95jC, 40 cycles with denaturationat 95jC for 20 s, annealing at 60jC for 20 s, and extension at 72jC for 20 s,

and final elongation at 72jC for 5 min. Melting curve analysis verified that

all primers yielded a single PCR product. Priming specificity for human

Gene name Forward Reverse

MDH2 GCAGCCACTTTCACTTCTC ACTCCAGCCGGAATAACTACTPI1 CACTGAGAAGGTTGTTTTCG TAAATGATACGGGTGCTCTG

PCK2 ATCCACATCTGTGATGGAAC CGTCTTGCTCTCTACTCGTG

ERRa TTCTCATCGCTGTCGCTGTCT CAGCCGCCGCACTAGTTGPGC-1a CTGGAGAGCCCCTGTGAGAGT GTGGGCTTGTACGGTGGTGT

PGC-1b GTTTCACCTCCAGCCTCAGAG CCAGGCAGGCCTCAGATCTA

IDH3A ATTGATCGGAGGTCTCGGTGT CAGGAGGGCTGTGGGATTC

ACO2 CCCGAGGTGAAGAATGTCATC GAAGCCCGTTGTACCAGCPGLS TGAGGACTACGCCAAGAAG AGTTGCCACAAAGATGACAG

ACAA2 ACAGACAATGCAGGTAGACG GCCAGTGGTGTGAAGTTATG

GAPDH GAAGGTGAAGGTCGGAGTC GAAGATGGTGATGGGATTTC

Molecular Analysis of Breast Cancer Brain Metastases

www.aacrjournals.org 1473 Cancer Res 2007; 67: (4). February 15, 2007

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

genes was established with mouse brain cDNA. Glyceraldehyde-3-phosphatedehydrogenase (GAPDH) primers were used for mRNA normalization and

quantitative real-time PCR was done twice in triplicates for each gene.

Cytotoxicity assay with bortezomib and 2-deoxy-D-glucose. Cells

(1 � 104 per well in 96-well plates) were seeded in growth medium andallowed to attach overnight before three gentle washes with PBS and

addition of bortezomib or 2-deoxy-D-glucose (2DG). Bortezomib (VEL-

CADE, Millenium Pharmaceuticals, Inc., Cambridge, MA) was reconstituted

in DMSO and 2DG in water. Bortezomib dilutions from 40 Amol/L to0.04 Amol/L were made with growth medium containing 2% FBS, and 2DG

ranging from 20 mmol/L to 0.31 mmol/L with growth medium containing

2% dialyzed FBS (Invitrogen). Drug-treated cells were compared with cells

treated with the respective diluent medium. Cytotoxic effects of each drugwere assessed after 24 h using the Cell Counting-8 viability assay (Dojindo

Molecular Technologies, Inc., Gaithersburg, MD). Each experiment was

repeated twice.Determination of total glutathione. Total glutathione was deter-

mined in cell lysates as described (20). Cell pellets were resuspended in

2 volumes of PBS, lysed by freezing and thawing in liquid nitrogen,

sonicated twice for 10 s, and centrifuged at 11,800 � g at 4jC for 10 min.Ten microliters of supernatant were analyzed in 0.5-mL phosphate buffer

[143 mmol/L sodium phosphate, 6 mmol/L EDTA (pH 7.5) containing

0.6 mmol/L 5,5¶-dithiobis(2-nitrobenzoic acid), 0.5 units/mL glutathione

reductase, and 0.3 mmol/L NADPH]. Glutathione reductase was producedrecombinantly as described (21). Reduction of 5,5¶-dithiobis(2-nitrobenzoicacid) was measured at 25jC by light absorbance at 412 nm. Glutathione

contents were determined against a calibration curve and corrected fortotal protein content. Protein concentration of cell extracts was determined

by the Bradford method.

Results and Discussion

Establishing a tumor cell model for breast cancer brainmetastasis. It is generally accepted that metastatic breast cancercells reach the brain primarily from the bloodstream (4). Wetherefore reasoned that blood-borne tumor cells could serve as asource for brain homing breast cancer cells and as a control againstwhich cells derived from brain metastases can be compared withdefined determinants of metastatic growth in the brain. We thusisolated circulating tumor cells from a stage IV breast cancerpatient, established the tumor cells in culture, reintroduced theminto the bloodstream of immunodeficient mice by tail-veininjection, and recovered tumor cells that had colonized the brain,or long bone for comparison. The cell line representing thecirculating tumor cells is named BCM2 Parent (22), and theirderivatives from bone lesion or brain lesions BCM2 Bone or BCM2BrainG1, respectively. To enrich for a brain metastatic phenotype,BCM2 BrainG1 cells were subjected to an additional round ofin vivo selection and the resulting cells were named BCM2 BrainG2.To establish the brain homing properties of these cells, they weregenetically tagged with F-luc and their fate and distributionfollowed in female SCID mice after tail-vein injection bynoninvasive bioluminescence imaging. BCM2 Parent cells colo-nized the brain and all other major target organs of breast cancermetastasis as seen in the clinic (Fig. 1A). A similar spectrum oforgan colonization was observed for the in vivo selected cellvariants derived from bone (BCM2 Bone) or brain metastases(BCM2 BrainG2), indicating a pluripotential of these cells oninjection into the tail vein. Despite the fact that this route favorslung colonization, because the pulmonary vasculature is the firstcapillary bed that the cells encounter, the incidence of brainmetastases in mice injected with the brain lesion-derived cells(83%) was significantly higher than that seen for BCM2 Parent(22%) or bone-derived cells (20%; Fig. 1B). Furthermore, tumor

burden in the brain caused by the brain metastatic cells was >36-fold larger than that induced by the parental cells, and >17-foldlarger than that caused by the bone metastatic cells. Measurementswere based on total photon flux in the excised brains (Fig. 1Cand D). Thus, BCM2 Parent cells represent a circulating breastcancer cell population that can efficiently colonize the brain fromthe bloodstream. Importantly, brain homing descendants of thesecells show an increased propensity to survive and proliferate withinthe brain microenvironment. These results indicate a stable changein the functional phenotype of brain metastatic breast cancercells and provide the basis for a molecular characterization of thisspecialized cell type.

Establishing a molecular profile of brain metastatic breastcancer cells. To define determinants of metastatic growth ofbreast cancer cells in the brain, we examined the proteinexpression profiles of the parental cell line and its brain or bonehoming descendants by shotgun proteomic analyses. Total proteinlysates were fractionated by chromatofocusing using the PF2DProteomeLab System (Beckman Coulter) and the fractionsanalyzed by multidimensional protein identification technology,MudPIT, to establish comprehensive protein profiles of BCM2Parent, BCM2 BrainG1, and BCM2 Bone cells. More than 300identified proteins were found z2-fold up-regulated or down-regulated in the brain metastatic cells compared with the parentalcells and bone metastatic variant. Functional classification ofthe proteins found differentially expressed in the brain metastaticcells revealed that 50% belong to the functional category of cellmetabolism. Proteins involved in cell stress response comprisethe second largest category (8%). Given the strong predominanceof alterations in the metabolic protein profile of the brainmetastatic cells, we focused our attention on proteins involved incell metabolism and identified 63 proteins, whose differentialexpression particularly marks the brain-derived cells. Theseproteins were further clustered into three subcategories: glucoseoxidation, fatty acid oxidation, and cellular redox-active proteins(Table 1).

Brain metastatic cells show enhanced mitochondrial respi-ratory pathways for energy production. Combining our large-scale expression data and knowledge of known metabolic net-works, we were able to construct a metabolic profile of brainmetastatic cells based on the differentially expressed proteins listedin Table 1. Proteins up-regulated in the brain metastatic cellsindicate three major changes in the energy metabolism of thesecells: enhanced glycolysis coupled to increased activity of thetricarboxylic acid (TCA) cycle, increased h-oxidation of fatty acids,and an elevated pentose phosphate pathway. Major up-regulatedproteins indicating these changes in the context of their energymetabolism pathways are highlighted in Fig. 2.

Oxidative metabolism. Instead of generating energy throughanaerobic glycolysis, which is often used by fast proliferat-ing cancer cells including BCM2 Parent cells, BCM2 brain-derivedcells seem to use primarily aerobic glycolysis, coupled to the TCAcycle and oxidative phosphorylation, to generate energy for cellgrowth. We found several lines of evidence supporting this hypo-thesis. First, we detected considerable up-regulation of proteinsinvolved in glycolysis, TCA cycle (marked in gray italics in Fig. 2),and oxidative phosphorylation based on our proteomic analyses(Table 1). Second, we found corresponding increase in thetranscription of genes encoding proteins that are involved inoxidative energy metabolism. Quantitative real-time PCR analysesconfirmed a trend in gene expression changes consistent with

Cancer Research

Cancer Res 2007; 67: (4). February 15, 2007 1474 www.aacrjournals.org

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

our proteomic data and revealed a specific up-regulation ofproteins involved in glycolysis and oxidative metabolism in thebrain metastatic cells (Fig. 3). The up-regulated proteins includethe glycolytic enzyme triose phosphate isomerase (TPI) andphosphoenolpyruvate carboxylase (PCK2), which can enhanceTCA flux via cataplerosis. Also up-regulated were the TCA cycleenzymes, aconitate hydratase (ACO2), isocitric dehydrogenase(IDH3A), and mitochondrial malate dehydrogenase (MDH2;Fig. 3A). Furthermore, we detected increased expression of

transcriptional regulators that promote oxidative metabolism,including members of the poly(ADP-ribose) polymerase-g coac-tivator-1 (PGC-1) family of transcriptional coactivators, PGC-1aand PGC-1h (23), and the nuclear receptor ERRa (Fig. 3A). ERRais a downstream effector of PGC-1a in the regulation of mito-chondrial energy metabolism and enables PGC-1a to induce targetgenes such as IDH3A (24). The coordinated up-regulation of allthree regulators, PGC-1a, PGC-1h, and ERRa, suggests thatthe brain metastatic cells have undergone a transcriptional switch

Figure 1. Human tumor cell model forbreast cancer brain metastasis. Metastaticprofiles of BCM2 cell variants. BCM2Parent cells were established from a bloodsample of a stage IV breast cancer patient.Bone and brain metastatic variants ofthese cells, designated as BCM2 Boneand BCM2 BrainG2, respectively, werederived from metastatic bone or brainlesions in female SCID mice after injectingBCM2 Parent cells into the tail vein.A, widespread metastatic activity in femaleSCID mice measured by noninvasivebioluminescence imaging 56 d aftertail-vein injection of 2.5 � 105

F-luc–tagged tumor cells. Threerepresentative mice are shown for eachexperimental group (n = 15). B, increasedincidence of brain metastases in miceinjected with the in vivo selected brainmetastatic cells (BCM2 BrainG2). *,P < 0.0001, Fisher’s exact test. Brainmetastasis was measured by ex vivobioluminescence imaging of the excisedbrains 8 wk after tail-vein injection of2.5 � 105 BCM2 Parent cells (22%incidence; n = 18), BCM2 BrainG2 cells(83% incidence; n = 23), or BCM2Bone cells (20% incidence; n = 15).C, metastatic burden in the brain inducedby BCM2 BrainG2 cells is >36-fold largerthan that induced by BCM2 Parent cellsand >17-fold larger than that by BCM2Bone cells. Total metastatic load wasmeasured ex vivo in the excised brains56 d after tail-vein injection of the tumorcells. Brain tumor burden was determinedby photon flux (photons/s/cm2) in definedregions of interest covering the entire brainand normalized to the bioluminescenceactivity of each cell line. D, representativeimages of excised brains from each groupshown on an identical signal scale toillustrate the differences in metastaticburden.

Molecular Analysis of Breast Cancer Brain Metastases

www.aacrjournals.org 1475 Cancer Res 2007; 67: (4). February 15, 2007

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

Table 1. Proteins identified by proteomic analysis as differentially expressed in brain metastatic breast cancer cells

Locus Description L MW pI P

SeqCount

P

SpecCount

P

SeqCov

Energy pathway

IPI00019383 Galactokinase 392 42,272 6.5 � � �

IPI00294380 Phosphoenolpyruvate carboxykinase,

mitochondrial precursor

640 70,637 7.6 � � �

IPI00465028 Triose phosphateisomerase 1 variant

249 26,713 7.4 13 79 57

IPI00017895 Glycero l-3-phosphate dehydrogenase,

mitochondrial precursor

727 80,815 7.4 � � �

IPI00383237 Pyruvate kinase M2 530 57,781 7.7 � � �IPI00549725 Phosphoglycerate mutase 1 266 30,048 7 4 24 19.2

IPI00025366 Citrate synthase, mitochondrial

precursor

466 51,712 8.3 3 5 6.2

IPI00017855 Aconitate hydratase, mitochondrial precursor 780 85,425 7.6 4 11 17.7IPI00305166 Succinate dehydrogenase [ubiquinone]

flavoprotein subunit, mitochondrial precursor

664 72,692 7.4 � � �

IPI00296053 Fumarate hydratase, mitochondrial precursor 510 54,637 8.8 11 35 20

IPI00011107 Isocitrate dehydrogenase [NADP],

mitochondrial precursor

452 50,909 8.7 2 7 7.3

IPI00291006 Malate dehydrogenase, mitochondrial precursor 338 35,531 8.7 2 4 12.1IPI00219217 L-lactate dehydrogenase B chain (LDH-H) 333 36,507 6.1 3 6 12.9

IPI00176698 Cytochrome c 105 11,966 9.5 5 10 40

IPI00021793 Cytochrome c oxidase polypeptide VIa-liver,

mitochondrial precursor; COX6A1

109 12,155 9.3 � � �

IPI00006579 Cytochrome c oxidase subunit IV isoform 1,mitochondrial precursor

169 19,577 9.5 3 12 20.1

IPI00008398 Cytochrome P450 26B1 512 57,513 8.5 2 2 8.4

IPI00010810 Electron transfer flavoprotein a-subunit,mitochondrial precursor

333 35,080 8.4 � � �

IPI00513827 Hypothetical protein DKFZp686M24262 454 50,271 7.8 � � �IPI00032297 Isovaleryl CoA dehydrogenase 426 46,568 8 1 17 4IPI00031109 Mimitin, mitochondrial precursor 169 19,856 9 � � �

IPI00619898 NAD(P)H menadione oxidoreductase 1,

dioxin-inducible isoform c

236 26,365 8.8 5 22 21.6

IPI00419266 NADH dehydrogenase (ubiquinone) 1asubcomplex, 6, 14 kDa

154 17,871 10.1 2 2 17.5

IPI00026964 Ubiquinol-cytochrome c reductase iron-sulfursubunit, mitochondrial precursor

274 29,652 8.3 � � �

IPI00011217 NADH-ubiquinone oxidoreductase 18 kDa

subunit, mitochondrial precursor

175 20,108 10.3 3 11 10.3

IPI00025796 NADH-ubiquinone oxidoreductase 30 kDa

subunit, mitochondrial precursor

264 30,242 7.5 � � �

IPI00028520 NADH-ubiquinone oxidoreductase 51 kDa

subunit, mitochondrial precursor

464 50,817 8.2 � � �

IPI00255052 NADH-ubiquinone oxidoreductase B22 subunit 178 21,700 8.4 � � �

IPI00654562 Cytochrome c oxidase polypeptide VIb 115 13,317 5.5 � � �IPI00013847 Ubiquinol-cytochrome-c reductase complex core

protein I, mitochondrial precursor

480 52,619 6.4 2 2 4

IPI00305383 Ubiquinol-cytochrome-c reductase complex core

protein 2, mitochondrial precursor

453 48,443 8.6 3 11 8.2

(Continued on the following page)

Cancer Research

Cancer Res 2007; 67: (4). February 15, 2007 1476 www.aacrjournals.org

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

Table 1. Proteins identified by proteomic analysis as differentially expressed in brain metastatic breast cancer cells (Cont’d)

Bo

SeqCount

Bo

SpecCount

Bo

SeqCov

Br

SeqCount

Br

SpecCount

Br

SeqCov

Cellular component Biological process

2 2 6.9 12 40 32.7 cytoplasm galactose metabolism;

metabolism� � � 6 15 11.7 mitochondrion gluconeogenesis

15 77 50.6 35 339 65.9 none gluconeogenesis;glycolysis

� � � 3 9 5.5 mitochondrion glucose catabolism

� � � 2 3 6 none glycolysis2 6 11.3 7 76 25.9 cytosol glycolysis� 1 � 5 14 12.2 mitochondrion tricarboxylic acid cycle

4 11 7.6 22 75 33.7 mitochondrion tricarboxylic acid cycle� � � 2 13 4.4 mitochondrion tricarboxylic acid cycle

5 17 17.8 12 72 24.9 mitochondrion; TCAcycle enzyme complex

tricarboxylic acid cycle

� � � 7 23 18.8 mitochondrion tricarboxylic acid cycle

� � � 8 40 26.3 mitochondrial matrix tricarboxylic acid cycle� � � 4 16 15.6 cytoplasm L-lactate dehydrogenase activity;

oxidoreductase activity

tricarboxylic acid cycle;

anaerobic glycolysis� � � � � � none electron transport� � � 5 7 24.8 mitochondrial membrane;

inner membrane

electron transport

� � � � � � mitochondrion;inner membrane

electron transport

� � � � � � endoplasmic reticulum;

microsome; membrane

electron transport

2 11 6 25 87 58.3 mitochondrial matrix electron transport

� � � 3 4 12.8 none electron transport

2 8 4 3 4 10.8 mitochondrial matrix electron transport� � � 2 6 16.6 mitochondrion; mitochondrial

inner membrane

electron transport

� � � 2 5 11.9 none electron transport

� � � � � � mitochondrion; mitochondrial

inner membrane

electron transport

� � � 2 2 9.9 mitochondrion;respiratory chain

complex III (sensu Eukarya)

electron transport

� � � � � � membrane fraction;

mitochondrion

‘‘mitochondrial electron transport,

NADH to ubiquinone’’� � � 3 5 17.4 membrane fraction;

mitochondrion

mitochondrial electron transport,

NADH to ubiquinone� � � 5 6 14.4 mitochondrion; mitochondrial

inner membrane

mitochondrial electron transport,

NADH to ubiquinone� � � 2 5 14 mitochondrion; mitochondrial

inner membrane

mitochondrial electron transport,

NADH to ubiquinone� � � 2 3 25.2 mitochondrion electron transport; metabolism� � � 3 5 14 mitochondrion;

inner membrane

electron transport; oxidative

phosphorylation� � � 8 24 20.3 mitochondrial electron

transport chain

electron transport; oxidative

phosphorylation

(Continued on the following page)

Molecular Analysis of Breast Cancer Brain Metastases

www.aacrjournals.org 1477 Cancer Res 2007; 67: (4). February 15, 2007

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

Table 1. Proteins identified by proteomic analysis as differentially expressed in brain metastatic breast cancer cells (Cont’d)

Locus Description L MW pI P

SeqCount

P

SpecCount

P

SeqCov

IPI00550882 Pyrroline-5-carboxylate reductase 1 319 33,361 7.6 � � �IPI00025252 Protein disulfide-isomerase A3 precursor 505 56,782 6.4 2 13 5

IPI00021785 Cytochrome c oxidase polypeptide Vb,

mitochondrial precursor

129 13,696 8.8 � � �

IPI00012069 NAD(P)H dehydrogenase [quinone] 1 274 30,868 8.9 5 22 18.6

IPI00219381 NADH-ubiquinone oxidoreductase B8 subunit 98 10,790 9.6 2 5 31.6

IPI00029561 NADH-ubiquinone oxidoreductase 42 kDasubunit, mitochondrial precursor

355 40,751 8.5 � � �

IPI00027776 Ferrochelatase, mitochondrial precursor 423 47,862 8.7 � � �

IPI00553153 Hypothetical protein DKFZp564G0422 107 12,405 9.6 2 12 15.9

IPI00419255 ATP6V1F protein 119 13,370 5.5 � � �

IPI00303476 ATP synthase h chain, mitochondrial precursor 529 56,560 5.4 � � �

IPI00029133 ATP synthase B chain, mitochondrial precursor 256 28,909 9.4 2 9 12.5

IPI00218848 ATP synthase e chain, mitochondrial 68 7,802 9.4 4 27 55.9

IPI00003856 Vacuolar ATP synthase subunit E 226 26,145 8 2 6 6.2

IPI00642733 NADH:ubiquinone oxidoreductase 206 22,143 9.9 2 5 9.7

Fatty acid h-oxidationIPI00298406 3-Hydroxyacyl-CoA dehydrogenase, isoform 2 390 42,123 9.3 2 7 7.2

IPI00001539 3-ketoacyl-CoA thiolase, mitochondrial 397 41,924 8.1 � � �IPI00005040 Acyl-CoA dehydrogenase, medium-chain

specific, mitochondrial precursor421 46,588 8.4 � � �

IPI00333838 Cytosolic acyl CoA thioester hydrolase, inducible 421 46,277 7.3 � � �IPI00011416 D3,5-y2,4-dienoyl-CoA isomerase,

mitochondrial precursor328 35,994 7.1 � � �

IPI00024993 Enoyl-CoA hydratase, mitochondrial precursor 290 31,387 8.1 � � �IPI00017726 3-hydroxyacyl-CoA dehydrogenase type-2 261 26,923 7.8 � � �

IPI00298202 Peroxisomal acyl-CoA thioester hydrolase 1 319 35,914 7.6 � � �

IPI00294398 Short chain 3-hydroxyacyl-CoA dehydrogenase,

mitochondrial precursor

314 34,278 8.9 2 7 8.9

IPI00010415 Splice Isoform 1 of Cytosolic acyl

CoA thioester hydrolase

380 41,796 8.5 3 30 11.3

IPI00220906 Splice Isoform 1 of Peroxisomal acyl-CoA

thioester hydrolase 2a

483 53,257 8.7 � � �

IPI00031522 Trifunctional enzyme a subunit,

mitochondrial precursor

763 83,000 9 5 18 10.7

IPI00022793 Trifunctional enzyme h subunit,

mitochondrial precursor

475 51,396 9.4 3 6 7.6

Cell redox homeostasisIPI00412561 Glutaredoxin family protein 379 42,170 9.2 � � �IPI00333763 Glutaredoxin-related protein C14orf87 157 16,628 6.8 � � �

(Continued on the following page)

Cancer Research

Cancer Res 2007; 67: (4). February 15, 2007 1478 www.aacrjournals.org

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

Table 1. Proteins identified by proteomic analysis as differentially expressed in brain metastatic breast cancer cells (Cont’d)

Bo

SeqCount

Bo

SpecCount

Bo

SeqCov

Br

SeqCount

Br

SpecCount

Br

SeqCov

Cellular component Biological process

� � � 2 2 8.8 none electron transport� � � 2 2 5.3 endoplasmic reticulum electron transport;

protein-nucleus import;

1 12 14 4 31 32.6 mitochondrial membrane;

inner membrane

electron transport

� � � 2 5 10.2 cytoplasm electron transport;

xenobiotic metabolism� � � � � � membrane fraction;

mitochondrionenergy pathways

� � � 3 4 17.2 membrane fraction;

mitochondrion

energy pathways

� � � 3 3 9.2 mitochondrion energy pathways

� � � 2 2 11.2 mitochondrion energy pathways� � � 4 10 46.2 proton-transporting

two-sector ATPase complexATP synthesis coupled

proton transport ATP

synthase complex

2 6 5.5 2 2 5.1 mitochondrion;proton-transporting

ATP synthesis coupled protontransport; proton transport

� � � � � � mitochondrial matrix ATP synthesis coupled proton

transport; proton transportATPase complex

� � � � � � mitochondrion; proton-

transporting two-sector

ATP synthesis coupled proton

transport; proton transport� � � � � � plasma membrane; proton-

transporting ATPase

complex

ATP synthesis coupled

proton transport;

proton transport

2 2 16 � � � none mitochondrial electron transport,

NADH to ubiquinone

2 2 8.2 8 33 22.8 none fatty acid metabolism� � � 6 11 14.4 mitochondrion lipid metabolism� � � 3 4 13.8 mitochondrial matrix fatty acid h-oxidation

� � � 8 10 24 none lipid metabolism� � � 3 3 11 mitochondrion; peroxisome fatty acid h-oxidation

� � � 2 4 11.7 mitochondrion fatty acid h-oxidation� � � 3 5 26.4 mitochondrion;

plasma membrane

lipid metabolism

� � � 2 5 11 peroxisome lipid metabolism; acyl-CoAmetabolism

2 2 10.2 8 33 28.3 mitochondrion fatty acid metabolism

5 16 13.9 7 60 22.6 cytoplasm lipid metabolism

� � � 8 10 20.9 peroxisome lipid metabolism;acyl-CoA metabolism

� � � 10 48 19 mitochondrion fatty acid metabolism

� � � 9 35 19.4 mitochondrial membrane fatty acid h-oxidation

� � � 5 9 11.3 none cell redox homeostasis� � � 3 11 28 mitochondrion cell redox homeostasis

(Continued on the following page)

Molecular Analysis of Breast Cancer Brain Metastases

www.aacrjournals.org 1479 Cancer Res 2007; 67: (4). February 15, 2007

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

that supports expression of oxidative metabolism pathways.Interestingly, PGC-1a, but not PGC-1h or ERRa, was elevatedin the bone metastatic cells, suggesting that PGC-1a expressionalone was not sufficient to activate the expression of down-stream targets such as ACO2 and IDH3A. ERRa is also a regulatorof other mitochondrial energy transduction pathways, includingfatty acid oxidation and oxidative phosphorylation (24–26).Consistent with this role, our proteomic results showed up-regulation of several enzymes involved in fatty acid oxidation,as well as components of the electron transfer chain (Table 1).Similar to the enzymes in the TCA cycle, we found increased mRNAlevels for fatty acid oxidation genes, such as h-ketothiolase(ACAA2) in the brain metastatic cells (Fig. 3B). To evaluate anin vivo relevance of our gene and protein expression resultsobtained with in vitro cultured BCM2 cell variants, we propa-gated the brain homing metastatic cells in the mouse brainand analyzed gene expression directly in the brain lesions.Consistent with the results in Fig. 3A to C , several of the genesup-regulated in the cultured brain metastatic cells were found atsimilarly elevated levels in the brain lesions (Fig. 3D). This findingindicates that the changes in gene regulation seen in the brainhoming cells can be maintained when the cells are expandedculture.In addition to enzymes and transcriptional regulators, our

proteomic analyses identified at least one signaling proteinimportant for energy homeostasis, the AMPK, up-regulated inbrain metastatic cells (Table 1). AMPK is a key regulator and hasbeen implicated in the control of PGC-1a in muscle (27). Wefound increased levels of AMPK protein expression and activation,based on Thr172 phosphorylation, in the brain metastatic cells(Fig. 4A). This result supports a possible functional link betweenAMPK, PGC-1a expression, and the altered energy metabolismin the brain homing cells. Numerous studies have establishedAMPK as a key regulator of energy homeostasis within the cell.On activation, AMPK switches off ATP consuming biosyntheticpathways (e.g., fatty acid synthesis) and turns on ATP generat-

ing metabolic pathways (e.g., fatty acid oxidation and glycolysis)to preserve ATP levels for cell survival (28, 29). The role offatty oxidation in the altered energy metabolism of brainmetastatic breast cancer cells is less clear and may have beeninfluenced by the culture conditions to which the cells wereexposed. However, because one of the metabolites of h-ketothiolase is acetyl-CoA, a substrate for the TCA cycle, it ispossible that fatty acid oxidation is switched on by AMPK atan increased rate in the brain metastatic cells to further sup-port the enhanced mitochondrial respiratory chain pathwaysfor energy production. In addition to acute effects of AMPKactivation, long-term effects can influence the overall regulationof energy metabolism, including increased mitochondrial enzymecontent and mitochondrial biogenesis (27, 28, 30, 31). Thus, AMPKcan contribute to the maintenance of high ATP levels, whichmay stay remarkably stable for high energy–dependent mole-cular activities (32). Consistent with increased glycolysis and oxi-dative phosphorylation in the brain metastatic cells, indicatedby our proteomic and transcriptional analyses, we found elevatedlevels of cellular ATP in these cells (Fig. 4B). It is possible thatlong-term effects promoted by constitutive increase in AMPKprotein and activation support adaptation of the brain meta-static cells to energy pathways predominant in the brain andcontribute to the growth advantage of tumor cells in the brainmicroenvironment.

Pentose phosphate pathway. Besides enhanced mitochondrialrespiratory chain pathways, we also detected an up-regulation inthe brain metastatic cells of enzymes involved in the oxidativephase of the pentose phosphate pathway (Figs. 2 and 3B). Thepentose phosphate pathway serves to generate NADPH andpentose ( five-carbon) sugars. This pathway consists of two distinctphases: oxidative and nonoxidative. The oxidative phase generatesNADPH and is one of three major ways in which the body createsreducing molecules to prevent oxidative stress (33). We found twokey enzymes of the oxidative phase up-regulated in the brainmetastatic cells: glucose-6-phosphate dehydrogenase, a NADPH

Table 1. Proteins identified by proteomic analysis as differentially expressed in brain metastatic breast cancer cells (Cont’d)

Locus Description L MW pI P

SeqCount

P

SpecCount

P

SeqCov

IPI00219757 Glutathione S-transferase P 209 23,225 5.6 3 6 14.4

IPI00016862 Glutathione reductase, mitochondrial precursor 522 56,257 8.5 � � �

IPI00465436 Catalase 526 59,625 7.4 4 5 10.3

IPI00289800 Glucose-6-phosphate dehydrogenase 515 59,257 6.8 7 14 17.3IPI00029997 6-phosphogluconolactonase 258 27,547 6.1 � � �

NOTE: Protein expression profiles of BCM2 Parent cells and their bone- or brain lesion–derived variants, BCM2 Bone and BCM2 BrainG1, determined by

shotgun proteomic analysis using multidimensional protein identification technology, MudPIT. List of proteins found z2-fold up-regulated or down-regulated in the brain metastatic cells compared with the parental and bone metastatic cells. The shown proteins represent the main functional

categories that were found differentially expressed in the brain metastatic cells.

Abbreviations: SeqCount, sequence count: number of nonredundant peptides used to identify this protein; SpecCount, spectra count: sum of all peptides

used to identify this protein; SeqCov, sequence coverage: percent of amino acid sequence covered by the identified peptides; �, no peptide identificationfor the listed protein; P, BCM2 Parent; Bo, BCM2 Bone; Br, BCM2 BrainG1.

Cancer Research

Cancer Res 2007; 67: (4). February 15, 2007 1480 www.aacrjournals.org

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

producing enzyme, and 6-phosphogluconolactonase (Table 1).Oxidative stress within cells is controlled primarily by the actionof the tripeptide glutathione (GSH), which can reduce peroxides tomaintain a reducing milieu within the cell. Regeneration of reducedglutathione by flavoenzyme glutathione reductase uses NADPH asa source of reducing equivalents (34). Consistent with the knownmetabolic network connecting the pentose phosphate pathway andthe glutathione system, we found an up-regulation of glutathione-dependent enzymes in the brain metastatic cells. Prominentexamples are glutathione reductase and glutathione S-transferaseP. In addition, catalase, another important antioxidant enzyme,was found up-regulated (Table 1; Fig. 2). We believe that theNADPH-producing phase of the pentose phosphate pathway, incombination with the induction of antioxidant enzymes, is crucialin the brain metastatic cells to support the detoxification ofoxidative stress. Our results suggest that the brain metastatic cellshave an enhanced mitochondrial respiratory metabolism, which

may lead to an increased production of reactive oxygen species. Wetherefore hypothesize that enhanced antioxidative defense mech-anisms are crucial in the cells to maintain reduced GSH and tominimize oxidative stress caused by reactive oxygen species in thebrain microenvironment.

Brain metastatic cells show reduced susceptibility to 2DG-induced cytotoxicity. It is well established that many cancercells exhibit increased rates of glycolysis and reduced rates ofrespiration (35, 36). Fast proliferating cancer cells adopt anaerobicglycolysis to support rapid growth, and this renders the cellssensitive to glucose deprivation (37, 38). Glucose deprivationcauses oxidative stress in cancer cells, and a synthetic glucoseanalogue, 2DG, mimics the effect of glucose deprivation. 2DG hasbeen used to target cancer cells by direct inhibition of glycolysis.Recently, 2DG has been shown to cause selective cytotoxicity intumor tissue by mechanisms similar to those triggered by glucosedeprivation (39, 40). To further confirm the metabolic differences

Figure 2. Schematic representation ofenergy pathways altered in brainmetastatic breast cancer cells based onproteomic identification of differentiallyexpressed proteins. Schematic outline ofoxidative energy pathways, highlightingproteins in gray italics that were foundup-regulated in the brain metastatic cells.The first pathway includes glucosecatabolism by glycolysis and the TCAcycle. The second pathway includes fattyacid h-oxidation, which generates acetylCoA, a substrate for the TCA cycle.The third pathway includes the pentosephosphate pathway, which providesreducing power (NADPH) and can regulatecellular redox homeostasis.

Table 1. Proteins identified by proteomic analysis as differentially expressed in brain metastatic breast cancer cells (Cont’d)

Bo

SeqCount

Bo

SpecCount

Bo

SeqCov

Br

SeqCount

Br

SpecCount

Br

SeqCov

Cellular component Biological process

� � � 6 22 27.8 none glutathione transferase activity;

transferase activity centralnervous system development;

metabolism� � � 7 25 20.3 mitochondrion cell redox homeostasis;

glutathione metabolism� � � 9 11 22.6 peroxisome cell redox homeostasis;

response to oxidative stress

2 3 6.6 11 28 30.7 cellular component unknown Hexose Monophosphate shunt� � � 2 4 10.9 none Hexose Monophosphate shunt

Molecular Analysis of Breast Cancer Brain Metastases

www.aacrjournals.org 1481 Cancer Res 2007; 67: (4). February 15, 2007

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 11: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

between the originally circulating parental breast cancer cells andtheir bone- or brain lesion–derived metastatic variants, we studiedthe effect of 2DG on the growth rate of these cells. We found thatthe brain metastatic cells, BCM2 BrainG1 and BrainG2, were 2-foldless sensitive to 2DG treatment than the parental and bonemetastatic breast cancer cells (Fig. 5A). Pyruvate, a metabolite ofglucose provided in the culture medium, may allow the cells tobypass the requirement for glucose and can direct the energyproduction via oxidative metabolism. It is also possible that theenhanced antioxidant capacity seen in the brain metastatic cellscontributes to the partial resistance of these cells to 2DG.Ample evidence suggests that glucose deprivation is sufficient toinduce cytotoxicity in cancer cells through metabolic oxidativestress (41, 42). Glucose deprivation can lead to a lack of NADPHand pyruvate and, consequently, to an impairment of thehydroperoxide metabolism (42). Thus, the enhanced pentosephosphate pathway and glutathione system detected in the brainmetastatic cells could help the cells to quickly correct anydisturbance of the cellular pro-oxidant and antioxidant balance,whereas the parental and bone metastatic breast cancer cellsapparently lack this flexibility.

Brain metastatic cells show enhanced resistance to oxida-tive damage related to the cellular glutathione status. Tofurther explore the hypothesis that an enhanced redox system inthe brain metastatic cells can provide protection against oxidative

stress, we studied the stress response of the parental breast cancercells and their bone or brain metastatic variants to oxidativedamage induced by bortezomib. Bortezomib, or PS341, is a specificinhibitor of proteasome activity that targets the protein degrada-tion system. The ubiquitin-proteasome system is responsible forintracellular proteolysis, particularly the degradation of short-livedor oxidized proteins (43). Ample evidence suggests that proteasomeinhibition induces mitochondrial dysfunction, increases generationof reactive oxygen species, elevates RNA and DNA oxidation, andpromotes protein oxidation (44). We exposed the parental breastcancer cells, as well as their bone- and brain-derived metastaticvariants, to increasing concentrations of bortezomib and foundthat the brain metastatic cells (BCM2 BrainG1 and BrainG2) were>60-fold less sensitive to this treatment than the parental cells(Fig. 5B). Furthermore, we measured the cellular content of totalglutathione as an indicator of the cellular redox state under normalculture conditions and in the presence of bortezomib. Undernormal growth conditions, which include 10% FBS in the medium,BCM2 Parent cells had the highest cellular glutathione levels.The difference in glutathione between the parental and brainmetastatic cells under normal culture conditions might be relatedto the slower in vitro growth rate of BCM2 BrainG1 and BrainG2cells, which we consistently observed (data not shown). However,in the presence of bortezomib, the glutathione levels in the brainmetastatic cells notably increased (22% in BCM2 BrainG1 cells and

Figure 3. Differential expression of genesinvolved in energy metabolism in brainmetastatic cells. mRNA expression ofgenes involved in energy metabolism wasmeasured by real-time PCR. mRNAexpression in BCM2 Bone, BrainG1, andBrainG2 cells was normalized to mRNAexpression measured in BCM2 Parentcells. A, enzymes involved in glycolytic(TPI) or TCA cycle (PCK2, IDH3A,MDH2, and ACO2). B, enzymes involvedin fatty acid oxidation (ACAA2) or thepentose phosphate pathway (PGLS).C, transcriptional regulators of oxidativemetabolism programs (PGC-1a, PGC-1b,and ERRa). D, genes found up-regulatedin the cultured brain metastatic cells werealso expressed at similarly elevated levelsin brain lesions caused by intracraniallyinjected BCM2 BrainG2 cells.

Cancer Research

Cancer Res 2007; 67: (4). February 15, 2007 1482 www.aacrjournals.org

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 12: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

13% in BrainG2 cells) whereas GSH decreased (�13%) in theparental cells (Fig. 5C). This finding is consistent with the reducedsensitivity of the brain metastatic cells to this drug and the highersensitivity of BCM2 Parent cells, where glutathione is depletedin response to bortezomib (Fig. 5B). This depletion likely reflectsenhanced oxidation of glutathione to GSSG, followed by GSSGexport. Together, these results support our hypothesis that brainmetastatic cells can up-regulate and rapidly adapt their cellularantioxidant defense to protect against oxidative stress. Up-regulation of NADPH production coupled with enhanced glutathi-one reductase activity allows for the detoxification of higherreactive oxygen species and GSSG fluxes, thus maintaining highGSH levels. The observed changes in energy metabolism andcellular stress response may thus confer a growth advantage tobrain metastatic breast cancer cells, allowing them to thrive inthe microenvironment of the brain tissue.

Brain metastatic breast cancer cells have a growth advant-age in the brain. Our experimental metastasis data and pro-teomic analyses indicate that the brain metastatic cells, selectedin vivo for their ability to establish brain metastases, possess a

phenotype distinct from the parental circulating tumor cellsand their bone metastatic counterparts. The protein expres-sion profile of the brain metastatic cells and its functionalvalidation imply a predisposition or bioenergetic adaptationof the tumor cells to the energy metabolism of the brain, confer-ring an advantage for tumor cell survival and proliferation in thebrain microenvironment. We found that i.v. injection of thebrain lesion–derived breast cancer cells leads to a high incidenceof brain metastasis significantly surpassing the parental cells andthe bone metastatic variant (Fig. 1). However, this approachincludes the possibility that the formation of brain metastasesis a result of enhanced tumor cell survival in the bloodstreamor an ability to cross the blood-brain barrier efficiently. Toeliminate these factors and directly assess tumor cell survivaland growth in the brain, we implanted the F-luc–tagged BCM2BrainG2 versus BCM2 Bone cells into the forebrain of experi-mental mice by stereotactic injection. Tumor cell growth wasfollowed by repeated noninvasive bioluminescence imaging overa period of 21 days. Based on the percent increase in photonflux over the signal of the original implant, we measured a sig-nificant enhancement in tumor cell growth for the brainmetastatic cells compared with their bone-derived counterparts(Fig. 6A). After 21 days, the overall tumor burden in the braincaused by the brain metastatic cells was cumulatively f100-foldlarger than that measured for the bone metastatic cells (Fig. 6B).Furthermore, we found that the brain metastatic cells spreadto other areas of the central nervous system and frequentlyextended to the spine (Fig. 6C), similar to the clinical situationoften seen in breast cancer patients with brain metastases. Thus,breast cancer cells from brain lesions can acquire a stable change infunctional phenotype that confers a growth advantage in the centralnervous system. Our proteomic data and cell function analysessuggest that this phenotype involves an adaptation in energymetabolism that promotes tumor cell growth in the brain.An important question arising from this and other studies is

whether tumor cell clones with a potential for seeding brainmetastases already exist within certain primary tumors, or if thedevelopment of brain metastases is primarily based on the evolutionof a tumor cell phenotype that supports brain colonization andexpansion within the unique microenvironment of the brain. Anumber of studies suggest that gene expression signatures, indicatinga metastatic potential or poor prognosis, can exist within primarytumors and be preserved throughout metastatic progression (45–49).Many of those genes are directly involved in cell proliferation,indicate the epithelial cell type from which the primary tumorderived, or contribute to overall cell motility and invasion. Inaddition, expression profiles associated with organ-specific breastcancer metastasis to bone or lung have been reported and comprisegenes that are likely critical for tumor cell entry into those targetorgan microenvironments (50, 51). Notably, these and other studiesalso show that gene expression pattern, indicative of metastaticpotential and target organ tropism, can be preserved after in vivoselected tumor cells are expanded in tissue culture (52).Our study is based on originally circulating breast cancer cells that

represent an advanced state of disease progression, as the cells werederived from a patient with active metastatic disease, years after theprimary tumor had been removed. Strikingly, the majority ofproteins, which we found differentially expressed in the brainmetastatic variant of the originally circulating tumor cells, isinvolved in cell metabolism and may reflect either a predispositionor an adaptation of the tumor cells to the specific conditions in the

Figure 4. Expression and activation of AMPK in brain metastatic cells. A, AMPKprotein expression in BCM2 Parent, BCM2 Bone, BCM2 BrainG1, and BCM2BrainG2 cells analyzed by Western blot analysis. Activation of AMPK byphosphorylation was examined with the same sample set using specificantibodies against the phosphorylated form of AMPK [pAMPK(T172) ].h-Actin antibody was used to show equal loading of proteins in each lane.B, measurements of ATP pool levels in BCM2 Parent, Bone, and BrainG2 cells.Cellular ATP measured in 2.5 � 104 cells per well in 96-well plates and growthmedium containing 10% FBS based on a luminescence ATP assay. Eachanalysis was done in quadruplicate and repeated twice. Columns, averagevalues of ATP; bars, SD. Brain metastatic cells produced significantly higherATP levels than the parental and bone metastatic cells. *, P = 0.0005, F = 10.2(one-way ANOVA followed by Tukey’s pairwise comparison). The cells used inthis assay were not tagged with luciferase.

Molecular Analysis of Breast Cancer Brain Metastases

www.aacrjournals.org 1483 Cancer Res 2007; 67: (4). February 15, 2007

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 13: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

brain tissue. In this context, it is important to emphasize that brainmetastases have an exceptional long latency, with a median latentinterval of 2 to 3 years after initial breast cancer diagnosis (53).Metastases to bone and other target organs generally appear muchearlier and also occur with a significantly higher frequency (54). Ourdata are compatible with the concept that a subset of tumor cellsevolves during malignant progression, perhaps from a preexistingcancer cell population, which is capable of gaining entry to theprotected environment of the central nervous system. Importantly,these tumor cells can apparently differentiate along pathways thatallow them to adapt and ultimately thrive in the brain microenvi-ronment in response to signals from the brain tissue.

Acknowledgments

Received 8/30/2006; revised 11/11/2006; accepted 12/7/2006.Grant support: NIH training grant T32 HL 07695 and later by the NIAID sub-

contract grant UCSD/MCB0237059 (E.I. Chen); grant U19 AI063603-02 (J. Hewel); SusanG. Komen grant PDF0403205 (J.S. Krueger); the Deutsche Forschungsgemeinschaft(BE 1540-9/1; to K. Becker). Additional support came from NIH grant P41RR11823(J.R. Yates III); NCI grants CA095458 and CA112287, and CBCRP grants 10YB-020 and11IB-0077 (B. Felding-Habermann); NIH grant DK064951 (A. Kralli); and the Sam andRose Stein Endowment Fund.

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.The lentiviral expression system for breast cancer cell tagging with Firefly

luciferase was developed and provided by Drs. Bruce Torbett and Mario Tschan ofTSRI and the statistical analyses were supported by Dr. Jim Koziol of TSRI.

Figure 5. Differential response of brain metastatic cellsto 2-DG and bortezomib. The cytotoxic effects of 2DGand bortezomib on BCM2 Parent, BCM2 Bone, BCM2BrainG1, and BCM2 BrainG2 cells were measured andcompared. The cells were exposed to serial dilutions of2DG (0–20 mmol/L; A) or bortezomib (0–40 Amol/L; B) for24 h before measuring cytotoxicity. The experiments weredone in triplicate and repeated twice. Points, averagevalues of two experiments; bars, SD. Comparing BCM2Parent, Bone, BrainG1, and BrainG2 cells, IC50 values for2DG were 0.97, 1.29, 3.12, and 3.12 mmol/L, respectively.Comparing the cells in the same order, IC50 values forbortezomib were 0.625, 10, J40, and J40 Amol/L.C, levels of glutathione. Total glutathione (GSH) wasmeasured in BCM2 Parent, Bone, BrainG1, and BrainG2cells after a 24-h exposure to 25 nmol/L bortezomib ordrug-free growth medium. Concentration of GSH from eachcell line was normalized to total protein concentration.Percent change in GSH level is shown for each cell line,comparing exposure to bortezomib versus normal growthmedium (BCM2 Parent �13%, BCM2 Bone +7%, BCM2BrainG1 +22%, BCM2 BrainG2 +13%). GSH productionin BCM2 BrainG1 and BrainG2 cells in response tobortezomib is significantly higher than in BCM2 Parentcells. P < 0.0001, F = 19.2 (one-way ANOVA followedby Tukey’s pairwise comparison).

Cancer Research

Cancer Res 2007; 67: (4). February 15, 2007 1484 www.aacrjournals.org

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 14: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

Figure 6. Growth advantage of brainmetastatic breast cancer cells in the brain.For direct measurements of tumor cellgrowth rates within the brain tissue,1 � 104 F-luc–tagged BCM2 BrainG2versus BCM2 Bone cells were implantedinto the forebrain of 6-wk-old female SCIDmice by stereotactic injection at 2 mmlateral, 1 mm anterior to bregma, and 3-mmdepth from dura. A, growth rate of brainimplanted tumor cells measured overa period of 21 d by noninvasivebioluminescence imaging. The percentincrease in tumor signal was significantlyhigher in BCM2 BrainG2 cells than inBCM2 Bone cells. P = 0.0005, t = 4.26(two-sample t test comparing meanincrease in photon flux with group sizes ofn = 10). Each symbol represents anindividual animal. Red bar, median valuefor each group. B, ex vivo imaging ofexcised brains 21 d after tumor cellimplantation illustrates enhanced overalltumor burden and intracranial spreading ofBCM2 BrainG2 cells compared with BCM2Bone cells. C, metastatic extension fromthe site of implantation in the brain to thespine was seen only in mice injected withBCM2 BrainG2 cells. Two representativemice are shown from each groupcomparing BCM2 BrainG2 to BCM2 Bone.

References1. Kirsch DG, Loeffler JS. Brain metastases in patientswith breast cancer: new horizons. Clin Breast Cancer2005;6:115–24.

2. Weil RJ, Palmieri DC, Bronder JL, Stark AM, Steeg PS.Breast cancer metastasis to the central nervoussystem. Am J Pathol 2005;167:913–20.

3. Klos KJ, O’Neill BP. Brain metastases. Neurologist2004;10:31–46.

4. Lassman AB, DeAngelis LM. Brain metastases. NeurolClin 2003;21:1–23, vii.

5. Chang EL, Lo S. Diagnosis and management ofcentral nervous system metastases from breast cancer.Oncologist 2003;8:398–410.

6. Fidler IJ. The organ microenvironment and cancermetastasis. Differentiation 2002;70:498–505.

7. Folkman J. Angiogenesis in cancer, vascular, rheu-matoid and other disease. Nat Med 1995;1:27–31.

8. Hanahan D, Weinberg RA. The hallmarks of cancer.Cell 2000;100:57–70.

9. Ohno S, Ishida M, Kataoka A, Murakami S. Brainmetastasis of breast cancer. Breast Cancer 2004;11:27–9.

10. Washburn MP, Wolters D, Yates JR III. Large-scaleanalysis of the yeast proteome by multidimensionalprotein identification technology. Nat Biotechnol 2001;19:242–7.

11. Raichle ME, Mintun MA. Brain work and brainimaging. Annu Rev Neurosci 2006;29:449–76.

12. Magistretti PJ, Pellerin L, Rothman DL, Shulman RG.Energy on demand. Science 1999;283:496–7.

13. Garcia-Espinosa MA, Rodrigues TB, Sierra A, et al.Cerebral glucose metabolism and the glutamine cycleas detected by in vivo and in vitro 13C NMRspectroscopy. Neurochem Int 2004;45:297–303.

14. Rao J, Oz G, Seaquist ER. Regulation of cerebral glu-cose metabolism. Minerva Endocrinol 2006;31:149–58.

15. Lubman DM, Kachman MT, Wang H, et al. Two-dimensional liquid separations-mass mapping ofproteins from human cancer cell lysates. J ChromatogrB Analyt Technol Biomed Life Sci 2002;782:183–96.

16. Chen EI, Florens L, Axelrod FT, et al. Maspin altersthe carcinoma proteome. FASEB J 2005;19:1123–4.

17. Yates JR III, Eng JK, McCormack AL, Schieltz D.Method to correlate tandem mass spectra of modifiedpeptides to amino acid sequences in the proteindatabase. Anal Chem 1995;67:1426–36.

18. Tabb DL, McDonald WH, Yates JR III. DTASelectand Contrast: tools for assembling and comparingprotein identifications from shotgun proteomics. JProteome Res 2002;1:21–6.

19. Liu H, Sadygov RG, Yates JR III. A model for randomsampling and estimation of relative protein abundancein shotgun proteomics. Anal Chem 2004;76:4193–201.

20. Becker K, Gui M, Traxler A, Kirsten C, Schirmer RH.Redox processes in malaria and other parasiticdiseases. Determination of intracellular glutathione.Histochemistry 1994;102:389–95.

21. Nordhoff A, Bucheler US, Werner D, Schirmer RH.Folding of the four domains and dimerization areimpaired by the Gly446!Glu exchange in humanglutathione reductase. Implications for the design ofantiparasitic drugs. Biochemistry 1993;32:4060–6.

22. Rolli M, Fransvea E, Pilch J, Saven A, Felding-Habermann B. Activated integrin avh3 cooperates withmetalloproteinase MMP-9 in regulating migration ofmetastatic breast cancer cells. Proc Natl Acad Sci U S A2003;100:9482–7.

23. Lin J, Handschin C, Spiegelman BM. Metaboliccontrol through the PGC-1 family of transcriptioncoactivators. Cell Metab 2005;1:361–70.

24. Schreiber SN, Emter R, Hock MB, et al. Theestrogen-related receptor a (ERRa) functions in PPARgcoactivator 1a (PGC-1a)-induced mitochondrial bio-genesis. Proc Natl Acad Sci U S A 2004;101:6472–7.

25. Vega RB, Huss JM, Kelly DP. The coactivator PGC-1cooperates with peroxisome proliferator-activatedreceptor a in transcriptional control of nuclear genesencoding mitochondrial fatty acid oxidation enzymes.Mol Cell Biol 2000;20:1868–76.

26. Huss JM, Torra IP, Staels B, Giguere V, Kelly DP.Estrogen-related receptor a directs peroxisome pro-

liferator-activated receptor a signaling in the tran-scriptional control of energy metabolism in cardiacand skeletal muscle. Mol Cell Biol 2004;24:9079–91.

27. Zong H, Ren JM, Young LH, et al. AMP kinase isrequired for mitochondrial biogenesis in skeletalmuscle in response to chronic energy deprivation.Proc Natl Acad Sci U S A 2002;99:15983–7.

28. Carling D. The AMP-activated protein kinasecascade-a unifying system for energy control. TrendsBiochem Sci 2004;29:18–24.

29. Xue B, Kahn BB. AMPK integrates nutrient andhormonal signals to regulate food intake and energybalance through effects in the hypothalamus andperipheral tissues. J Physiol 2006;574:73–83.

30. Fryer LG, Carling D. AMP-activated protein kinaseand the metabolic syndrome. Biochem Soc Trans 2005;33:362–6.

31. Luo Z, Saha AK, Xiang X, Ruderman NB. AMPK, themetabolic syndrome and cancer. Trends Pharmacol Sci2005;26:69–76.

32. Neumann D, Schlattner U, Wallimann T. A molecularapproach to the concerted action of kinases involved inenergy homoeostasis. Biochem Soc Trans 2003;31:169–74.

33. Horecker BL. Alternative pathways of carbohydratemetabolism in relation to evolutionary development.Comp Biochem Physiol 1962;4:363–9.

34. Holmgren A. Thioredoxin and glutaredoxin systems.J Biol Chem 1989;264:13963–6.

35. Warburg O. On the origin of cancer cells. Science1956;123:309–14.

36. Weber G. Enzymology of cancer cells ( first of twoparts). N Engl J Med 1977;296:486–92.

37. Weber G. Enzymology of cancer cells (second of twoparts). N Engl J Med 1977;296:541–51.

38. Lee YJ, Galoforo SS, Berns CM, et al. Glucosedeprivation-induced cytotoxicity in drug resistant humanbreast carcinoma MCF-7/ADR cells: role of c-myc andbcl-2 in apoptotic cell death. J Cell Sci 1997;110:681–6.

39. Laszlo J, Humphreys SR, Goldin A. Effects of glucoseanalogues (2-deoxy-D-glucose, 2-deoxy-D-galactose) onexperimental tumors. J Natl Cancer Inst 1960;24:267–81.

Molecular Analysis of Breast Cancer Brain Metastases

www.aacrjournals.org 1485 Cancer Res 2007; 67: (4). February 15, 2007

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 15: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

40. Lin X, Zhang F, Bradbury CM, et al. 2-Deoxy-D-glucose-induced cytotoxicity and radiosensitization intumor cells is mediated via disruptions in thiolmetabolism. Cancer Res 2003;63:3413–7.

41. Blackburn RV, Spitz DR, Liu X, et al. Metabolicoxidative stress activates signal transduction and geneexpression during glucose deprivation in human tumorcells. Free Radic Biol Med 1999;26:419–30.

42. Spitz DR, Sim JE, Ridnour LA, Galoforo SS, Lee YJ.Glucose deprivation-induced oxidative stress in humantumor cells. A fundamental defect in metabolism? AnnN Y Acad Sci 2000;899:349–62.

43. Miller J, Gordon C. The regulation of proteasomedegradation by multi-ubiquitin chain binding proteins.FEBS Lett 2005;579:3224–30.

44. Ding Q, Dimayuga E, Keller JN. Proteasomeregulation of oxidative stress in aging and age-relateddiseases of the CNS. Antioxid Redox Signal 2006;8:163–72.

45. Weigelt B, Glas AM, Wessels LF, et al. Geneexpression profiles of primary breast tumors main-tained in distant metastases. Proc Natl Acad Sci U S A2003;100:15901–5.

46. Weigelt B, Hu Z, He X, et al. Molecular portraits and70-gene prognosis signature are preserved throughoutthe metastatic process of breast cancer. Cancer Res2005;65:9155–8.

47. Sorlie T, Tibshirani R, Parker J, et al. Repeatedobservation of breast tumor subtypes in independentgene expression data sets. Proc Natl Acad Sci U S A2003;100:8418–23.

48. Sorlie T, Perou CM, Tibshirani R, et al. Geneexpression patterns of breast carcinomas distinguishtumor subclasses with clinical implications. Proc NatlAcad Sci U S A 2001;98:10869–74.

49. Van’t Veer LJ, Dai H, van de Vijver MJ, et al. Geneexpression profiling predicts clinical outcome of breastcancer. Nature 2002;415:530–6.

50. Minn AJ, Kang Y, Serganova I, et al. Distinct organ-specific metastatic potential of individual breastcancer cells and primary tumors. J Clin Invest 2005;115:44–55.

51. Montel V, Huang TY, Mose E, Pestonjamasp K, TarinD. Expression profiling of primary tumors and matchedlymphatic and lung metastases in a xenogeneic breastcancer model. Am J Pathol 2005;166:1565–79.

52. Ross DT, Scherf U, Eisen MB, et al. Systematicvariation in gene expression patterns in human cancercell lines. Nat Genet 2000;24:227–35.

53. Bendell JC, Domchek SM, Burstein HJ, et al. Centralnervous system metastases in women who receivetrastuzumab-based therapy for metastatic breastcarcinoma. Cancer 2003;97:2972–7.

54. Pantel K, Muller V, Auer M, et al. Detection andclinical implications of early systemic tumor celldissemination in breast cancer. Clin Cancer Res 2003;9:6326–34.

Cancer Research

Cancer Res 2007; 67: (4). February 15, 2007 1486 www.aacrjournals.org

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 16: Adaptation of Energy Metabolism in Breast Cancer Brain ... · nates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties

2007;67:1472-1486. Cancer Res   Emily I. Chen, Johannes Hewel, Joseph S. Krueger, et al.   MetastasesAdaptation of Energy Metabolism in Breast Cancer Brain

  Updated version

  http://cancerres.aacrjournals.org/content/67/4/1472

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/4/1472.full#ref-list-1

This article cites 54 articles, 16 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/4/1472.full#related-urls

This article has been cited by 27 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://cancerres.aacrjournals.org/content/67/4/1472To request permission to re-use all or part of this article, use this link

Research. on October 15, 2020. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from