15
Cell Metabolism Article Blocking Lipid Synthesis Overcomes Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni, 1,5, * Jonathan Cimino, 1,2,5 Silvia Blacher, 1 Irina Primac, 1 Alice Truong, 1 Gabriel Mazzucchelli, 2 Alexandra Paye, 1 David Calligaris, 2 Delphine Debois, 2 Pascal De Tullio, 3 Bernard Mari, 4 Edwin De Pauw, 2 and Agnes Noel 1 1 Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium 2 Mass Spectrometry Laboratory, GIGA-R, Department of Chemistry, University of Liege, 4000 Liege, Belgium 3 Laboratory of Drug Research Center, University of Liege, 4000 Liege, Belgium 4 UMR-7275 CNRS, University of Nice Sophia-Antipolis, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France 5 Co-first author *Correspondence: [email protected] http://dx.doi.org/10.1016/j.cmet.2014.05.022 SUMMARY The molecular mechanisms responsible for the failure of antiangiogenic therapies and how tu- mors adapt to these therapies are unclear. Here, we applied transcriptomic, proteomic, and metabolomic approaches to preclinical models and provide evi- dence for tumor adaptation to vascular endothelial growth factor blockade through a metabolic shift toward carbohydrate and lipid metabolism in tumors. During sunitinib or sorafenib treatment, tumor growth was inhibited and tumors were hypoxic and glycolytic. In sharp contrast, treatment withdrawal led to tumor regrowth, angiogenesis restoration, moderate lactate production, and enhanced lipid synthesis. This metabolic shift was associated with a drastic increase in metastatic dissemination. Interestingly, pharmacological lipogenesis inhibition with orlistat or fatty acid synthase downregulation with shRNA inhibited tumor regrowth and metasta- ses after sunitinib treatment withdrawal. Our data shed light on metabolic alterations that result in can- cer adaptation to antiangiogenic treatments and identify key molecules involved in lipid metabolism as putative therapeutic targets. INTRODUCTION Antiangiogenic agents currently used in clinic target the vascular endothelial growth factor (VEGF) signaling pathway via an anti- VEGF antibody (bevacizumab, Avastin) or small-molecule re- ceptor tyrosine kinase inhibitors (RTKIs). Anti-VEGF antibodies lead to increased overall survival or progression-free survival in patients with metastatic colorectal cancer, non-small cell lung cancer, or breast cancer when administered in combination with conventional chemotherapeutic regimens (Jain et al., 2006). Sunitinib (Sutent, Pfizer), a multi-RTKI that targets VEGF recep- tors, platelet-derived growth factor (PDGF) receptor, and cKIT, also provides clinical benefit to patients with renal cell carci- noma or advanced gastrointestinal stromal tumors (GISTs) (Adams and Leggas, 2007; van der Veldt et al., 2008). Sorafenib (Nexavar, Bayer and Onyx), another antiangiogenic RTKI that also displays inhibitory activity toward Raf kinase, has been approved for the treatment of renal cell carcinoma and liver can- cer (Wu et al., 2008). Overall, the survival benefits of antiangio- genic drugs have been relatively modest, and surprisingly, most cancer patients stop responding or do not respond at all to antiangiogenic therapy (Ebos and Kerbel, 2011). Alarmingly, preclinical studies have reported increased tumor growth and metastatic formation after the withdrawal of treatment with VEGF receptor inhibitors, including sunitinib (Ebos et al., 2009a; Pa ` ez-Ribes et al., 2009), a monoclonal antibody against VEGFR2 (DC101), or a polyclonal antibody against VEGF (Sen- nino et al., 2012). This tumor evasion of antiangiogenic treatment appears to be dose dependent and tumor model dependent (Singh et al., 2012; Welti et al., 2012). Whether tumor regrowth can occur when therapy is stopped is an important clinical issue, and several studies have raised pertinent questions about how best to use antiangiogenic drugs (Bruce et al., 2014; Griffioen et al., 2012). A deeper understanding of tumor responses to anti- angiogenic therapy and the mechanisms by which resistance is acquired is urgently needed. The poor response to anti-VEGF monotherapy is likely due to the occurrence of multiple adaptive processes as a result of a multitude of resistance mechanisms that occur in both tumor cells and the tumor microenvironment (Sounni and Noel, 2013). VEGF blockade can be compensated by an increased production of alternative angiogenic factors (Ebos et al., 2009b). Other mechanisms include the recruitment of vascular progenitors, HIF-1a-induced autophagy, and cancer stem cell pool expansion (Ebos et al., 2009b; Giuliano and Page ` s, 2013; Sounni and Noel, 2013). In addition, cancer cells differ from normal cells regarding how their metabolic pathways are used to fuel cellular growth and survival. Metabolic adapta- tion in cancer is supported by clinical studies that have linked altered whole-body metabolism to cancer development and progression and poor treatment outcomes (Vander Heiden, 2011). Whether a metabolic switch contributes to the poor response of tumors to antiangiogenic treatment has not been determined. 280 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc.

Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

  • Upload
    ngominh

  • View
    222

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Article

Blocking Lipid Synthesis OvercomesTumor Regrowth and Metastasisafter Antiangiogenic Therapy WithdrawalNor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia Blacher,1 Irina Primac,1 Alice Truong,1 Gabriel Mazzucchelli,2

Alexandra Paye,1 David Calligaris,2 DelphineDebois,2 Pascal De Tullio,3 BernardMari,4 Edwin DePauw,2 andAgnesNoel11Laboratory of Tumor and Developmental Biology, GIGA-CANCER, University of Liege, 4000 Liege, Belgium2Mass Spectrometry Laboratory, GIGA-R, Department of Chemistry, University of Liege, 4000 Liege, Belgium3Laboratory of Drug Research Center, University of Liege, 4000 Liege, Belgium4UMR-7275 CNRS, University of Nice Sophia-Antipolis, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France5Co-first author

*Correspondence: [email protected]://dx.doi.org/10.1016/j.cmet.2014.05.022

SUMMARY

The molecular mechanisms responsible for thefailure of antiangiogenic therapies and how tu-mors adapt to these therapies are unclear. Here, weapplied transcriptomic, proteomic, andmetabolomicapproaches to preclinical models and provide evi-dence for tumor adaptation to vascular endothelialgrowth factor blockade through a metabolic shifttoward carbohydrate and lipidmetabolism in tumors.During sunitinib or sorafenib treatment, tumorgrowth was inhibited and tumors were hypoxic andglycolytic. In sharp contrast, treatment withdrawalled to tumor regrowth, angiogenesis restoration,moderate lactate production, and enhanced lipidsynthesis. This metabolic shift was associatedwith a drastic increase in metastatic dissemination.Interestingly, pharmacological lipogenesis inhibitionwith orlistat or fatty acid synthase downregulationwith shRNA inhibited tumor regrowth and metasta-ses after sunitinib treatment withdrawal. Our datashed light on metabolic alterations that result in can-cer adaptation to antiangiogenic treatments andidentify key molecules involved in lipid metabolismas putative therapeutic targets.

INTRODUCTION

Antiangiogenic agents currently used in clinic target the vascular

endothelial growth factor (VEGF) signaling pathway via an anti-

VEGF antibody (bevacizumab, Avastin) or small-molecule re-

ceptor tyrosine kinase inhibitors (RTKIs). Anti-VEGF antibodies

lead to increased overall survival or progression-free survival

in patients with metastatic colorectal cancer, non-small cell

lung cancer, or breast cancer when administered in combination

with conventional chemotherapeutic regimens (Jain et al., 2006).

Sunitinib (Sutent, Pfizer), a multi-RTKI that targets VEGF recep-

tors, platelet-derived growth factor (PDGF) receptor, and cKIT,

280 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc

also provides clinical benefit to patients with renal cell carci-

noma or advanced gastrointestinal stromal tumors (GISTs)

(Adams and Leggas, 2007; van der Veldt et al., 2008). Sorafenib

(Nexavar, Bayer and Onyx), another antiangiogenic RTKI that

also displays inhibitory activity toward Raf kinase, has been

approved for the treatment of renal cell carcinoma and liver can-

cer (Wu et al., 2008). Overall, the survival benefits of antiangio-

genic drugs have been relatively modest, and surprisingly,

most cancer patients stop responding or do not respond at all

to antiangiogenic therapy (Ebos and Kerbel, 2011). Alarmingly,

preclinical studies have reported increased tumor growth

and metastatic formation after the withdrawal of treatment

with VEGF receptor inhibitors, including sunitinib (Ebos et al.,

2009a; Paez-Ribes et al., 2009), a monoclonal antibody against

VEGFR2 (DC101), or a polyclonal antibody against VEGF (Sen-

nino et al., 2012). This tumor evasion of antiangiogenic treatment

appears to be dose dependent and tumor model dependent

(Singh et al., 2012; Welti et al., 2012). Whether tumor regrowth

can occur when therapy is stopped is an important clinical issue,

and several studies have raised pertinent questions about how

best to use antiangiogenic drugs (Bruce et al., 2014; Griffioen

et al., 2012). A deeper understanding of tumor responses to anti-

angiogenic therapy and the mechanisms by which resistance is

acquired is urgently needed. The poor response to anti-VEGF

monotherapy is likely due to the occurrence of multiple adaptive

processes as a result of a multitude of resistance mechanisms

that occur in both tumor cells and the tumor microenvironment

(Sounni and Noel, 2013). VEGF blockade can be compensated

by an increased production of alternative angiogenic factors

(Ebos et al., 2009b). Other mechanisms include the recruitment

of vascular progenitors, HIF-1a-induced autophagy, and cancer

stem cell pool expansion (Ebos et al., 2009b; Giuliano and

Pages, 2013; Sounni and Noel, 2013). In addition, cancer cells

differ from normal cells regarding how their metabolic pathways

are used to fuel cellular growth and survival. Metabolic adapta-

tion in cancer is supported by clinical studies that have linked

altered whole-body metabolism to cancer development and

progression and poor treatment outcomes (Vander Heiden,

2011). Whether a metabolic switch contributes to the poor

response of tumors to antiangiogenic treatment has not been

determined.

.

Page 2: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

(legend on next page)

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc. 281

Page 3: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

Here, using transcriptomic, proteomic, and metabolomic ap-

proaches, we provide evidence for a metabolic shift of tumors

to de novo lipogenesis following RTKI treatment withdrawal,

which is associated with rapid tumor regrowth and accelerated

metastatic dissemination. The functional implications of lipo-

genesis and fatty acid synthase (FASN) in this process

are supported by the blockade of tumor relapse through the

inhibition or downregulation of FASN with orlistat or shRNA,

respectively. Our data support the innovative concept of using

metabolic pathway inhibitors to sensitize tumors to antiangio-

genic drugs.

RESULTS

Withdrawal of Angiogenesis Inhibitor TreatmentAccelerates Tumor Growth and MetastasisWe applied several treatment regimens to a metastatic model of

human breast cancer involving MDA-MB-231 xenografts. Long-

term treatment with sunitinib at a therapeutic dose of 40 mg/kg

for 30 days resulted in a drastic inhibition of primary tumor

growth and metastasis formation (Figure 1A). By contrast,

short-term pretreatment with sunitinib 9 days prior to subcu-

taneous (s.c.) injection of tumor cells affected neither tumor

growth nor metastasis (Figures S1A and S1B, available online).

We next evaluated the effects of long-term treatment with suni-

tinib followed by treatment withdrawal (W-sunitinib) (Figures 1B

and 1C). As expected, whereas primary tumors responded

effectively to sunitinib during the treatment period, treatment

withdrawal resulted in accelerated tumor regrowth (Figures

1B). Indeed, sunitinib treatment resulted in tumor growth inhibi-

tion, as characterized by a stabilization of tumor volumes to

approximately 50–70 mm3 during the entire treatment period.

After sunitinib withdrawal tumors regrew rapidly, reaching a

volume of 300 mm3 5-fold faster than vehicle-treated tumors

(7 days for W-sunitinib tumors versus 35 days for control tu-

mors) (Figure 1E). Moreover, sunitinib withdrawal resulted in

increased metastasis in organs of mice sacrificed when tumors

reached the volume of the tumors of the vehicle group

(400 mm3) (Figure 1B, lower panel). A drastic increase in metas-

tases was also seen by bioluminescence and immunohisto-

chemical (IHC) staining of organs (lung, brain, and lymph nodes)

(Figures 1B and 1C). Similar results were obtained after the

withdrawal of treatment with sorafenib (W-sorafenib), another

RTKI approved for antiangiogenic therapy (Strumberg, 2005)

(Figures 1D and 1E). This drastic acceleration of tumor regrowth

and metastatic dissemination suggests a selection of aggres-

Figure 1. Long-Term Sunitinib Treatment Increases a Tumor Regrowth

during the Treatment

MDA-MB-231 LUC+ cells were subcutaneously injected in RAG1�/� mice.

(A) Evolution of tumor volumes in mice treated daily with vehicle or sunitinib

quantification of bioluminescence in organs (lower).

(B) Evolution of tumor volumes in mice treated with vehicle or sunitinib and follo

bioluminescence (images in bottom panel) and quantified (graph in middle panel

(C) Representative immunohistochemical images visualizing micrometastases b

Scale bar, 100 mm.

(D) Evolution of tumor volumes in mice treated daily with vehicle or sorafenib (50 m

line) (top) and quantification of bioluminescence in organs (bottom).

(E) Representative table showing a shorter latency period to reach a volume of 3

Data are presented as mean ± SD for tumor growth and mean ± SEM for biolum

282 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc

sive tumor cells or an adaptation of the tumors to RTK inhibitor

withdrawal.

The Evaluation of Cancer Hallmarks in Primary TumorsReveals a Metabolic Shift after AntiangiogenesisTreatment WithdrawalTo determine if histopathological features of primary tumors

were altered in response to antiangiogenesis treatment cessa-

tion, IHC analyses of key hallmarks of cancer progression (angio-

genesis, inflammation, matrix, and metabolism) were conducted

at the end of the assay when the vehicle and W-sunitinib tumors

both reached a volume of 400 mm3. We did not observe a differ-

ence in CD31 staining, a marker of angiogenesis, between the

two tumor types. Computerized quantification of vessel density

and distribution from the border to the center of the tumors re-

vealed a similar pattern in both experimental groups (Figure S2A).

Thus, angiogenesis was fully restored after sunitinib withdrawal.

However, the density and distribution of vessels positive for

a-smooth muscle actin (a-SMA) were twice as high as those in

the W-sunitinib group, indicating an increase in vessel matura-

tion after sunitinib withdrawal (Figure S2B). No difference in tu-

mor lymphangiogenesis and inflammatory infiltrates was seen

(Figures S2C and S2D). The analysis of matrix deposition by

saffron staining did not reveal any difference in the collagen den-

sity and distribution between W-sunitinib and vehicle tumors

(Figure S2E).

Recent preclinical studies have identified several mechanisms

of tumor adaptation to antiangiogenic agents, including the

compensatory production of alternative proangiogenic factors

(Ebos et al., 2009b; Sounni and Noel, 2013). As an initial attempt

to identify putative alternative factors, we analyzed the mRNA

expression levels of major pro-lymph/angiogenic and pro-

invasive molecules in W-sunitinib and control tumors (Fig-

ure S2F). Notably, VEGF-A, VEGF-C, HIF-1a, PAI-1, MMP-11,

and MMP-13 were expressed at similar levels. We then applied

global transcriptomic and proteomic analyses to compareW-su-

nitinib and vehicle tumors. Gene profiling using whole-genome

mouse microarrays indicated that sunitinib withdrawal induced

the modulation of around a hundred genes (Supplemental

Information, microarray_W-sunitinib). Interestingly, one of the

main relevant networks identified by ingenuity pathway analysis

(IPA) was related to carbohydrate metabolism and, notably, lipid

metabolism/regulation (Figure 2A).

For proteomic analysis, total tumor extracts were analyzed by

mass spectrometry. Again, amarkedmetabolic shift appeared in

W-sunitinib tumors, as reflected by the modulation of enzymes

and Metastatic Dissemination after Treatment Withdrawal, but Not

(40 mg/kg) (D-sunitinib) for 30 days (as indicated by dashed line) (top) and

wed by sunitinib withdrawal (W-sunitinib) (top). Metastases were detected by

) after sunitinib withdrawal.

y immunostaining for human Ki67 in organs of the W-sunitinib-treated group.

g/kg) followed by sorafenib withdrawal (W-sorafenib) (as indicated by dashed

00 or 400 mm3 in W-sunitinib and W-sorafenib groups.

inescence quantification in organs. p % 0.05.

.

Page 4: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Figure 2. Sunitinib Treatment Withdrawal Results in a Cancer Metabolic Shift Identified by Transcriptomics, Proteomics, and Metabolomics

Analyses

(A) Representative table of the relevant mouse genes analyzed by RNA microarray of primary W-sunitinib tumors compared with vehicle-treated tumors. The

transcriptional profiling showed an upregulation of genes associated with carbohydrate and lipid metabolism. The diagram represents an ingenuity network

representation of the main upregulated (red) and downregulated (green) gene network.

(B) Proteomic analysis of primary tumors by mass spectrometry for human and mouse proteins. Relative protein quantity variations upon sunitinib treatment and

withdrawal in primary tumors are indicated as fold change obtained from the ratio of vehicle/W-sunitinib. *p % 0.05; **p % 0.01. Sunitinib treatment withdrawal

(legend continued on next page)

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc. 283

Page 5: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

associated with the citric acid cycle (TCA), lactate regulation,

and lipid transporters (Figure 2B). Intriguingly, many glycolytic

enzymes were downregulated in W-sunitinib tumors compared

with control tumors. These modifications were similar when

considering human and mouse proteins, suggesting a metabolic

adaptation in both host and tumor compartments (Figure 2B).

Thus, the metabolic perturbations observed by proteomic and

transcriptomic analyses appeared to boost fatty acid, pyruvate,

and amino acid metabolisms, whereas glucose metabolism and

cellular carbohydrate catabolism were downregulated by suniti-

nib withdrawal (Figure 2C). Furthermore, the identification of

metabolic end products in tumor extracts by nuclear magnetic

resonance (NMR) (Figure 2D) and scores plot analysis revealed

a clear difference in metabolites between vehicle and W-suniti-

nib tumors. The main discriminants identified through this me-

tabolomics approach correspond to lactate and lipids.

The mRNA levels of several genes in the metabolic network

were validated by RT-PCR analysis. Upon sunitinib withdrawal,

we observed increased mRNA levels of FASN, a key enzyme

in de novo lipogenesis that condenses malonyl coenzyme A

(malonyl-CoA) and acetyl-CoA into a long-chain fatty acid (Fig-

ure 3A). The expression of acetyl-CoA carboxylase (ACC-a),

the enzyme that transforms acetyl-CoA into malonyl-CoA, was

also enhanced. Similar modulations were detected using spe-

cific human and mouse primers (Figure S3A), suggesting that

de novo lipogenesis is triggered in both the tumor microenviron-

ment and cancer cells. The shift to lipid metabolism was further

supported by increased expression of adiponectin and perilipin

mRNAs. Western blot analysis of FASN and ACC-a confirmed

their increased levels in W-sunitinib tumors, while no difference

was seen in ACC-a phosphorylation status (Figure 3B). The pro-

duction of PPARg, a factor regulating lipid beta-oxidation, was

not affected by sunitinib withdrawal (Figure 3B). These data

underline a metabolic reprogramming toward de novo lipogen-

esis to sustain cancer growth and invasion. Furthermore,

increased FASN production and lipid droplet amounts inW-suni-

tinib tumors is supported by IHC stainings using antibodies

against FASN and perilipin, a marker of adipocytes (Figures 3C

and 3D). A computerized quantification performed on whole-

tumor sections confirmed the increased abundance of lipid

droplets throughout the tumors (Figure 3D). Enhanced deposi-

tion of collagen type VI produced mainly by adipocytes was

also detected (Figure 3E). Thus, W-sunitinib tumors were char-

acterized by substantial adipocyte infiltration and/or differentia-

tion. Notably, FASN, ACC-a, and perilipin modulations at mRNA

and protein levels were also increased in W-sorafenib tumors

(Figures 3F–3H).

leads to an upregulation of lipid metabolism, lactate regulation, and TCA enzym

regulation (blue).

(C) Schematic diagram summarizing the bioinformatics analysis of the proteomi

withdrawal: (1) glycolysis (17%), (2) cellular carbohydrate catabolic process (17%

monosaccharide metabolic process (16%), (6) cellular macromolecule catabolic

rivative metabolic process (4%), (9) glutathione metabolic process (4%). The right

metabolism (14%), (3) pyruvate metabolism (14%), (4) pentose phosphate pathwa

methionine metabolism (10%), (7) arginine and proline metabolism (10%), (8) a

D-glutamine and D-glutamate metabolism (5%). Proteomic profiling with mass sp

from W-sunitinib and three tumors from vehicle-treated mice. Changes in transc

(D)Metabolic analysis by NMRof primary tumors treatedwith vehicle (V1-9) or afte

the principal component analysis (PCA) model. Main discriminant metabolites ar

284 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc

Cancer Cell Metabolism Is Differentially Altered duringand after the Withdrawal of Treatment withAngiogenesis InhibitorsThe adaptation of tumormetabolism to lipogenesis inW-sunitinib

tumors prompted us to analyze the metabolic profile of tumors

during a period of sustained treatment with sunitinib (D-sunitinib)

(Figure S3B). Lipogenesis was decreased rather than increased

during sunitinib treatment. The expression of perilipin and

FASN in mice was drastically reduced in D-sunitinib tumors,

and no difference was found in the expression levels of human

FASN and ACC-a. As assessed by NMR, an enhancement of

lactate production was detected and was higher in D-sunitinib

than in W-sunitinib tumor extracts (5-fold versus 1.7-fold as

compared to control tumors) (Figure 4A). Carbonic anhydrase-

IX (CA-IX) staining revealed increased hypoxia during sunitinib

treatment (Figure 4B), but not after its withdrawal (Figure S4A).

Interestingly, the expression of monocarboxylate transporter 1

of lactate (MCT1) was enhanced in both D-sunitinib and W-suni-

tinib tumors, whereas MCT4 expression was induced only in

D-sunitinib tumors (Figures 4C–4E). Taken together, these obser-

vations suggest that during sunitinib treatment, tumors become

more glycolytic. By contrast, after treatment cessation, tumors

may be fueled by lipid metabolism and increased TCA activity.

Accordingly, the levels of twomajor regulators of pyruvate dehy-

drogenase (PDH) activity, pyruvate dehydrogenase kinases 1/2

(PDK1 and PDK2), were increased in D-sunitinib tumors (Fig-

ure 4F), indicating a reduction/inhibition of pyruvate oxidative

decarboxylation, which fuels the TCAcycle in D-sunitinib tumors.

In sharp contrast, upon sunitinib withdrawal, glycolysis was

reduced,PDK1andPDK2expressionwasdecreased (Figure4G),

and TCA activity was increased in W-sunitinib (Figures 4B and

4C). Consistently, PDK1 protein levels were decreased in W-su-

nitinib tumors (Figure S4B). Accordingly, the in vitro treatment of

MDA-MB-231 cells with sunitinib resulted in increased lactate

production in cell media and extracts and correlated with a

marked increase in glucose uptake by cells (Figure 5A). Similar

results were generated in breast (BT-549) and colorectal (HCT-

116) cancer cells treated with sunitinib or sorafenib (Figures 5A

and 5B). MCT4, PDK1, and PDK2 expression levels were

also induced after treatment with both RTKIs (Figures 5C and

5D). Notably, MCT1 is not produced by MDA-MB-231 cells

under basal conditions; its expression is induced upon RTKI

treatment. We next extended our investigation to stromal cells

(Figures 5E–5G). Lactate production and glucose uptake were

enhanced >1.5-fold in endothelial cells (HUVECs) (Figures 5E

and 5F). This correlates with increased mRNA levels of PDK1

and PDK2 expression, whereas the expression of MCT4,

es (orange, gray, and red, respectively) and to a downregulation of glycolysis

c data. The left circle corresponds to downregulated pathways after sunitinib

), (3) glucosemetabolic process (16%), (4) hexose metabolic process (16%), (5)

process (6%), (7) cellular protein metabolic process (4%), (8) amino acid de-

circle corresponds to upregulated pathways: (1) TCA cycle (19%), (2) fatty acid

y (9%), (5) alanine, aspartate, and glutamate metabolism (9%), (6) cysteine and

dipocytokine signaling pathway (5%), (9) tyrosine metabolism (5%), and (10)

ectrometry was performed on total protein extracts from three different tumors

riptomic and proteomic data are obtained in 100% tumor samples.

r sunitinib withdrawal (W-S1-9). Score plots are shown for PC1 versus PC2 from

e shown below.

.

Page 6: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

(legend on next page)

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc. 285

Page 7: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

MCT1, or FASN was not affected (Figure 5G). A similar effect on

lactate production and glucose consumption was observed

upon 48 hr of RTKI treatment of cancer-associated fibroblasts

(CAFs) isolated from MMTV-PyMT mice (Figures 5E and 5F).

Angiogenesis Inhibitor Treatment Withdrawal InducesTumorRegrowth and aMetabolic Shift in SeveralModelsof CancerRAG1�/� mice bearing human colorectal HT-29 tumors of

70 mm3 were treated with sunitinib (40 mg/kg/day) for 30 days

(Figure 6A). Again, tumor growth was inhibited during the period

of sunitinib treatment, and accelerated tumor regrowth was

observed after treatment cessation. The tumor volumes

increased from 70 mm3 to 800–900 mm3 within 10 days after su-

nitinib withdrawal. Control tumors required twice as much time

to reach a similar volume (Figure 6A). Metastasis formation

was also drastically increased (right panel). Moreover, lactate

quantification revealed a 4-fold increase of this metabolite in

W-sunitinib tumors (Figure 6B). FASN density on tumor sections

was again increased inW-sunitinib HT-29 tumors comparedwith

vehicle-treated ones (Figure 6C). Lactate production and

glucose uptake increased significantly in vitro after RTKI treat-

ments (Figures 6D), recapitulating the response observed in

the other cell lines tested (Figure 5).

An oncogenic mouse model of mammary carcinoma was also

used. MMTV-PyMT mice (55 days old) bearing mammary tu-

mors of 50–75 mm3 were treated with sunitinib (n = 7) or vehicle

(n = 6) for 21 days (from day 55 to 76) followed by sunitinib with-

drawal for 17 days (Figure 6E). Sunitinib administration inhibited

tumor development during the treatment period, whereas suni-

tinib withdrawal was associated with a faster tumor growth and

rapid apparition of more numerous tumors when compared to

vehicle-treated mice. Indeed, treatment withdrawal led to the

apparition of tumors >5 mm within 12 days, while it took almost

88 days to reach this volume for fewer tumors in vehicle-treated

animals (Figure 6E). Lung metastasis was also drastically

increased after suninitib withdrawal, as revealed by hematoxylin

and eosin (H&E) staining of lung sections and computerized

quantification of metastasis size on whole-lung sections (Fig-

ure 6F). Importantly, an upregulation of lipid metabolism-asso-

ciated genes, including perilipin, adiponectin, and FASN, was

revealed by RT-PCR analyses (Figure S5A). The increase in

FASN production was further validated by IHC staining on tu-

mor sections (Figure 6G). The modulation of lactate production

and glucose consumption in cancer cells isolated from MMTV-

PyMT tumors was also confirmed in vitro upon RTKI treat-

ments (Figure S5B). Taken together, these data validate our

Figure 3. Cancer Adaptation to Sunitinib or Sorafenib Treatment Withdr

(A) RT-PCR analysis of mouse (m) and/or human (h) mRNA levels of fatty acid sy

W-sunitinib compared to vehicle-treated tumors (1–5; five individual tumor sample

levels normalized to ribosomal 28S.

(B) Western analysis of phosphorylated ACC (S79), total ACC, FASN, and PPAR

(C–E) Immunohistological images of primary tumors stained with antibody to FA

density and distance distribution from tumor edge to the center in whole-tumor

100 mm.

(F) RT-PCR analysis of mouse (m) and/or human (h) mRNA levels of FASN, adipon

(1–5; five individual tumor samples are shown). The graphs on the right correspo

(G and H) Immunostaining on tumor sections of FASN (G) and perilipin (H). The g

*p % 0.05; **p % 0.01; ***p % 0.005.

286 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc

concept of tumor metabolic reprogramming during and after

RTKI administration.

The Adaptation to Angiogenesis Inhibitor TreatmentWithdrawal Is Reversed by FASN Blockade in SeveralCancer ModelsOur findings indicate ametabolic adaptation of tumors to antian-

giogenic therapy that differs during and after treatment with su-

nitinib (Figure 6H). Glycolysis was induced during the treatment,

and lipid metabolism was associated with a more aggressive

phenotype after antiangiogenic therapy withdrawal. We hypoth-

esized that interfering with lipid metabolism via FASN inhibition

after sunitinib treatment might overcome this tumor adaptation

and block the associated metastatic dissemination. Our working

hypothesis was first evaluated by using orlistat, a pharmacolog-

ical inhibitor of FASN. Mice were separated in four experimental

groups: (1) vehicles for sunitinib and orlistat (Vehicle S/O); (2)

orlistat and the vehicle for sunitinib (Orlistat + vehicle S); (3) suni-

tinib as a single agent and the vehicle for orlistat (W-sunitinib +

VehicleO); and (4) orlistat after sunitinibwithdrawal (W-sunitinib +

orlistat).

Treatment with orlistat significantly reduced tumor growth at

an early stage of MDA-MB-231 tumor development (Figure 7A).

However, this treatment had no effect at later stages, and by the

end of the treatment tumors reached the volume of vehicle-

treated tumors. As expected, treatment with sunitinib inhibited

tumor growth during the treatment period, followed by a rapid

regrowth after sunitinib withdrawal. Interestingly, orlistat treat-

ment following sunitinib withdrawal reduced tumor regrowth

and inhibited metastasis formation (Figure 7B). Since mice

administered with orlistat displayed a reduction in body weight

of 5%–10% after 21 days of treatment (Figure 7A), we checked

whether orlistat could affect the whole-body metabolism by

applying a metabolomic approach to mice sera. The global

metabolic profiles of mice sera were indistinguishable between

the experimental groups (Figure 7C). Orlistat treatment of W-su-

nitinib tumors reduced lactate production and normalized its

level to vehicle- and orlistat-treated tumors (Figure 7D). Comput-

erized quantification of IHC stainings revealed a strong inhibition

of FASN and perilipin production upon orlistat treatment (Fig-

ure 7E). As expected, CA-IX stainings were similar in the four

experimental groups (Figure 7E).

We next extended our investigation to four additional models,

including colorectal HT29 xenografts, syngeneic models of

4T1 tumors heterotopically (s.c.) or orthotopically (in the mam-

mary fat pad, MFP) implanted in Balb/c mice, and Lewis lung

carcinoma (LLC) s.c. injected into C57BL6 mice (Figure S6).

awal Is Strongly Linked to aMetabolic Shift toward LipidMetabolism

nthase (FASN), adiponectin, perilipin, and acetyl-CoA carboxylase (ACC-a) in

s are shown). The graphs on the right correspond to the quantification of mRNA

g. HSC70 and actin are used as loading controls.

SN (C), perilipin (D), and collagen type VI (E). Computerized quantification of

sections are shown below the images (n = 15 tumors per group). Scale bar,

ectin, perilipin, and ACC-a in W-sorafenib compared to vehicle-treated tumors

nd to the quantification of mRNA levels normalized to ribosomal 28S.

raphs correspond to quantification performed as described above.

.

Page 8: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Figure 4. Cancer Metabolism Shifts fromHypoxia andGlycolysis during Sunitinib Treatment to Normoxia and a Reduction of Glycolysis after

Sunitinib Withdrawal

(A) Lactate production in tumors was measured by NMR after treatment for 30 days with sunitinib (D-sunitinib) and after withdrawal (W-sunitinib).

(B and C) Images of immunohistochemical stainings with carbonic anhydrase (CA)-IX (B) and MCT1 (C) antibodies and their quantifications (graph) in D-sunitinib

tumors. The graphs represent a computerized quantification in whole-scanned tumor sections of each staining. Results are presented as staining density (top

graphs) and as distribution from the tumor edge to the center (bottom graphs). n = 12 tumors per group. Scale bar, 100 mm.

(D–G) RT-PCR analysis of expression levels of lactate transporters MCT1 and MCT4 in tumors during (D) and after (E) sunitinib treatment withdrawal. RT-PCR

analysis of mRNA expression levels of PDK1 and PDK2 in tumors during (F) and after (G) sunitinib treatment withdrawal. n = 5 per group. *p% 0.05; **p% 0.01;

***p % 0.005.

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

Interestingly, in all these models, sunitinib withdrawal was

associated with increased tumor regrowth and metastasis,

which can be completely reversed by the subsequent treatment

with orlistat.

To achieve a specific FASN inhibition in the tumor compart-

ment, MDA-MB-231 cells were stably transfected with control

shRNA (shCTR) or shRNA targeting FASN (shFASN) (Figures

7F–7I). Although the downregulation of FASN in untreated mice

(shFASN vehicle) delayed the onset of tumor development

in vivo, at the end of the assay these tumors reached volumes

similar to those of vehicle-treated tumors. Interestingly, the tu-

mor regrowth observed after sunitinib withdrawal did not occur

Cel

in FASN downregulated tumors (shFASN W-sunitinib), and the

metastatic dissemination was completely abolished (Figures

7G–7I). Overall, these data clearly demonstrate that FASN activ-

ity in cancer cells is a rate-limiting event for themalignant pheno-

type observed after drug withdrawal. These experiments clearly

show that interfering with tumor FASN is sufficient to overcome

the tumor aggressiveness induced by drug withdrawal.

DISCUSSION

Herein, we provide mechanistic insights into how tumors adapt

their metabolism to VEGF-based antiangiogenic treatment.

l Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc. 287

Page 9: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

(legend on next page)

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

288 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc.

Page 10: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

Global transcriptomic, proteomic approaches, as well as histo-

chemical, biochemical, and NMR analyses, revealed a shift to-

ward lipid metabolism and increased TCA activity to fuel cancer

cells after TKI treatment withdrawal (Figure 6H). Our data ob-

tained in five experimental models highlight the usefulness

of FASN inhibition to reverse this malignant adaptation and the

associated acceleratedmetastatic dissemination. These innova-

tive results have clinical implication for the improvement of anti-

angiogenic therapies and provide mechanistic insights into the

intriguing recent clinical data revealing that RTKI treatment break

can be associated with tumor regrowth (Bruce et al., 2014).

In line with previous studies (Ebos et al., 2009a; Paez-Ribes

et al., 2009), sunitinib treatment in preclinical xenograft models

led to an adaptive-evasive response after treatment withdrawal,

but not during treatment administration (Figures 1 and 6H). During

sunitinib treatment of human MDA-MB-231 and HT-29 xeno-

grafts, tumors shrank and metastases were inhibited, whereas

treatmentwithdrawal accelerated tumor regrowth andmetastatic

dissemination to different organs. Similar effects were observed

with a transgenic model of MMTV-PyMT mice and syngeneic

mousemodelsof 4T1andLLC tumors.Histological analysisofpri-

mary tumors after sunitinib treatment and withdrawal show a

restoration of blood vascularization and an increased density of

a-SMApositive cells, butnodifference in inflammatory cell infiltra-

tion and lymphangiogenesis. These phenomena were assessed

by a robust computerized quantification applied to whole-tumor

sections fromat least 15primary tumors per condition (FigureS2).

Importantly, a metabolic shift to de novo lipogenesis after su-

nitinib treatment cessation was indicated by the upregulation of

key enzymes and cytokines involved in lipid metabolism and

cancer progression. Notably, both transcriptomic and proteomic

approaches highlighted the same perturbed pathway, which is

associated with lipid and carbohydrate metabolism. Among

the genes validated by RT-PCR analysis, FASN, perilipin, and

adiponectin were significantly upregulated after sunitinib with-

drawal. Perilipin immunostaining of tumor sections confirmed

the increased presence of lipid droplets in adipocytes inside

W-sunitinib tumors. Adipocyte accumulation is also supported

by enhanced deposition of type VI collagen mainly produced

by these cells (Nakajima et al., 2002).

The strength of our study is its depiction of cancer metabolism

during and after RTKI administration (Figure 6H).Wedemonstrate

that tumors adopt a glycolytic phenotype during RTKI treatment,

whereas after drug withdrawal, tumors shift to lipid metabolism

and TCA activity. RTKIs can have a direct effect on the meta-

bolism of cancer cells and stromal cells, as demonstrated by

the in vitro study (Figure 5). The glycolytic phenotype of D-suniti-

nib tumors during RTKI treatment was supported by an increase

in lactate production, lactate transporter (MCT1 and MCT4)

Figure 5. Sunitinib and Sorafenib Induce the Warburg Effect in Cancer

(A) Lactate production was measured by NMR in vitro in medium, and cell ext

sunitinib (5 mM) for 24 or 48 hr. Glucose uptake was evaluated by measuring the

(B) Following the same experimental design in (A), cancer cells were incubated w

(C andD) RT-PCR analysis ofmRNA expression levels of MCT1,MCT4, FASN, PD

or sorafenib (D).

(E and F) Lactate production was measured by NMR in medium from HUVECs a

(G) RT-PCR analysis of mRNA expression levels of MCT1, MCT4, FASN, PDK1,

Data are presented as mean ± SD. *p % 0.05; **p % 0.01.

Cel

expression, PDK expression, and a hypoxic microenvironment

resulting from angiogenesis inhibition. These features reflect the

useof anaerobicglycolysis tomeet thebioenergetic needsof pro-

liferation and/or survival, known as the Warburg effect (Feron,

2009). Hypoxia is known to play a critical role in reprogramming

cancermetabolism towardanaerobicglycolysis (i.e., theWarburg

effect) through the induction of PDK and lactate dehydrogenase

(LDH) expression and increased lactate production (Kim et al.,

2006). Indeed, lactate was recently identified as a major energy

source in tumors; the lactate produced by hypoxic tumor cells

may diffuse and be taken up by oxygenated tumor cells (Son-

veaux et al., 2008). In vitro treatment with sunitinib and sorafenib

of fourdifferent cancer cell lines in addition tocancer cells isolated

from MMTV-PyMT induced a rapid and drastic increase in

glucose uptake and lactate production. A significant induction

of PDK1 and PDK2 was also observed in vitro. RTKI- mediated

glycolytic effects were also observed in CAFs and HUVECs, indi-

cating a similar response of stromal cells to therapy. This is in line

with the key role of glycolysis reported in endothelial cells during

angiogenesis (Schoors et al., 2014). Thesedata demonstrate that,

beyond their antiangiogenic effects, RTKIs can directly affect the

metabolism of both cancer and stromal cells.

After RTKI withdrawal (Figure 6H), W-sunitinib tumors dis-

played a metabolic shift toward (1) a decrease in glycolytic en-

zymes, (2) an increase in lipid metabolism and transporter levels,

and (3) an increase in TCA enzyme levels. A decrease in perilipin-

positive cells and low FASN and ACC-amRNA expression levels

were observed during sunitinib administration. Thus, the meta-

bolic shift toward lipogenesis is specific to sunitinib withdrawal

rather than to a direct effect of the RTKI during the treatment.

The moderate increase in lactate levels observed after with-

drawal is likely due to tumor revascularization, which reduces

hypoxia. This finding is consistent with the decrease in PDK1

and PDK2 expression, which also supports increased TCA activ-

ity supplied by pyruvate. In proliferating cells, citrate provided

by increased TCA activity is exported from the mitochondria to

the cytoplasm, where citrate can be converted to the lipogenic

precursors acetyl-CoA and oxaloacetate by ATP citrate lyase

(Feron, 2009). The reduction in glycolysis in the W-sunitinib

tumors and the increase in citrate cycle activity may result in

increased lipids synthesis to sustain tumor proliferation. While

no obvious relative increase in beta-oxydation markers (PPARg

and phoshorylated ACC) was seen in W-sunitinib tumors, the

increased de novo lipid synthesis is likely used as building blocks

for tumor rebound following treatment withdrawal.

The link between therapeutic sensitivity and dysregulated can-

cer cell metabolism is rapidly being dissected (reviewed in Butler

et al., 2013). Lipogenic enzymes, and particularly FASN, have

become an attractive target for therapeutic intervention (Currie

and Stromal Cells In Vitro

racts from MDA-MB-231, BT-549, and HCT-116 cancer cells incubated with

glucose levels in medium.

ith 5 mM sorafenib for 24 and 48 hr.

K1, and PDK2 inMDA-MB-231 cells incubated for 24 and 48 hr with sunitinib (C)

nd CAFs incubated with sunitinib (E) or sorafenib (F).

and PDK2 in HUVECs incubated for 24 and 48 hr with sunitinib.

l Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc. 289

Page 11: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

(legend on next page)

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

290 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc.

Page 12: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

et al., 2013). The pharmacological inhibition of FASNwith orlistat

completely reversed tumor regrowth and metastatic dissemina-

tion induced by sunitinib withdrawal in five experimental models

(Figures 7 and S6). The treatment of mice with orlistat at the indi-

cated concentration and period did not show a significant differ-

ence in the globalmetabolic profile of sera. This indicates that the

effects observed could not be ascribed to a systemic impact on

thewhole-bodymetabolism, but rather to a shift occurring specif-

ically in the tumors. However, we cannot exclude differences in

some specific metabolites in sera of mice treated with orlistat.

It is worth noting that homeostasis of the blood metabolism is a

highly regulated mechanism and that only urine analysis may

reveal more precisely metabolic changes. In addition, the 5%–

10% weight loss caused by orlistat is likely not high enough to

significantly affect the global metabolic profile of sera. Although

the role of FASN in initial cancer growth is well documented,

our study is the first to provide its implication in malignant adap-

tation to antiangiogenic therapy removal. The key role played

by FASN in the tumor compartment is supported by the use of

shRNA technology in MDA-MB-231 tumors. Importantly, shRNA

FASN completely abolished the tumor regrowth and metastatic

dissemination induced by sunitinib withdrawal in vivo. These ex-

periments clearly show that interfering with tumor FASN is suffi-

cient to overcome the tumor aggressiveness induced by drug

withdrawal, and those results have potential clinical implications.

By providing a detailed metabolic profile of tumor adaptation

during and after RTKI treatment, our study shed light on recent

alarming clinical data reporting that RTKI treatment break could

boost cancer malignancy.

EXPERIMENTAL PROCEDURES

Cell Culture

Human cells from breast cancer (MDA-MB-231 expressing or not expressing

luciferase [LUC+]), BT-549 cells, human colorectal cancer HCT-116 andHT-29

Figure 6. Colorectal Cancer Xenograft and Transgenic MouseModel of

of Breast Cancer Xenografts

(A–D) RAG1�/�mice were injected subcutaneously with HT-29 LUC+ cells and trea

tumor volumes showing an inhibition of tumor growth during sunitinib administratio

Metastasis detection by bioluminescence in organs of vehicle- andW-sunitinib-tre

level measurement by NMR in HT-29 tumors. (C) Representative immunohistoc

100 mm. Quantification of FASN positive cell density and distribution from the bord

same procedure as described in Figures 5A and 5B, lactate production and glu

sorafenib treatment for 24 and 48 hr.

(E–G) MMTV-PyMT mice bearing mammary tumors (50 mm3) were treated with ve

corresponds to the tumor volume evolution. The table indicates the number of tum

its withdrawal (W-sunitinib). (F) H&E staining of lung sections showing metastase

whole-lung sections. Scale bar, 1mm. (G) Representative immunohistochemical im

and stained for FASN (left). Scale bar, 100 mm. Quantification of FASN positive c

(n = 36 tumors for W-sunitinib and n = 31 for vehicle group). Data are presented

(H) Schematic representation of the tumor metabolism shift during and after treatm

tumors), RTKIs can have direct effects on the metabolic process of stromal and

hypoxia. Hypoxic tumors shrink and become hypoxic, leading to an increase in gl

anaerobic glycolysis to sustain their energy needs for survival. Pyruvate dehy

D-sunitinib tumors, which results in the inhibition of pyruvate dehydrogenase (PDH

acid (TCA) cycle and to the accumulation of lactate. Cancer cells overexpress

overexpress MCT4 only in hypoxic conditions imposed by antiangiogenic therap

overexpress MCT1 for the import of lactate and produce high levels of LDH th

W-sunitinib tumors, citrate cycle enzymes are upregulated, and PDK1 and PDK

metabolism, a process associated with the acquisition of a malignant phenotype a

synthesis after sunitinib withdrawal, the acquisition of the malignant phenotype c

Cel

LUC+, mouse mammary carcinoma 4T1-LUC+, mouse Lewis lung carcinoma

LLC-LUC+ cells expressing luciferase, HUVECs, PyMT mammary carcinoma

epithelial cancer cells, and CAFs isolated from MMTV- PyMT mice were

used. Cell culture and transfection of MDA-MB-231 LUC+ cells with shRNA

against FASN are described in the Supplemental Experimental Procedures.

In Vivo Experiments

Preclinical Models

For human xenografts, MDA-MB-231 LUC+ or HT-29 LUC+ subconfluent cells

were trypsinized, resuspended in serum-free medium (5 3 106 cells/ml), and

mixed with an equal volume of cold Matrigel according to a previous report

(Sounni et al., 2002). Cell suspension (106 cells/400 ml) was injected s.c. into

RAG1�/� immunodeficient mice in both flanks. For the syngeneic model,

4T1-LUC+ cell (5 104) suspension in 100 ml Dulbecco’s modified Eagle’s me-

dium (DMEM) was mixed with 100 ml Matrigel and subcutaneously (heterotyp-

ic) injected in Balb/c mice. For orthotopic injections in the MFP, 8-week-old

(n = 8) Balb/c female mice were anesthetized, and 50 ml of cell suspension

(5 3 104 cells) was injected in the mammary gland after a small incision in

the skin (0.5 cm). For the syngeneic model of Lewis lung carcinoma (LLC),

cell (5 3 104) suspensions in 100 ml DMEM were mixed with 100 ml Matrigel

and subcutaneously (heterotypic) implanted in 8-week-old C57BL6 mice. Tu-

mor growth was assessed by measuring the length and width of tumors every

3–4 days. Tumor volumes were estimated as previously described (length 3

width2 3 0.4) (Noel et al., 1993). For in vivo imaging, mice were injected intra-

peritoneally with 75 mg/kg D-luciferin (Caliper Life Sciences), anesthetized

with isofluorane, and then imaged. Animals were imaged once a week, and or-

gans were imaged at sacrifice. Imaging was performed using a Lumina II IVIS

(in vivo imaging system) instrument (Caliper Life Sciences) with biolumines-

cence quantification using Living Image software (Caliper Life Sciences) ac-

cording to manufacturer’s recommendations. All animal procedures with small

animals were performed according to the Federation of European Laboratory

Animal Sciences Associations (FELASA) and local ethical committee at Univer-

sity of Liege (Belgium). Animals were housed within the accredited animal

facility of GIGA located at the CHU Sart Tilman (University of Liege, Belgium).

Drug administration and experimental groups are described in detail in the

Supplemental Experimental Procedures.

Primary Cell Isolation and Culture

Extraction and specific isolation of CAFs and PyMT epithelial cancer cells are

described in the Supplemental Experimental Procedures.

Mammary Carcinoma Recapitulate the Tumor Adaptation Response

ted daily with suninitib (40mg/kg) for 30 days (from day 7 to 37). (A) Evolution of

n and a rapid tumor regrowth after sunitinib treatment withdrawal (W-sunitinib).

ated animals (right). Data are presented asmean ± SEM (p% 0.01). (B) Lactate

hemical images of primary HT-29 tumors stained for FASN (left). Scale bar,

er to the center of the tumors (right) (n = 12 tumors per group). (D) Following the

cose uptake of HT-29 cells were evaluated in vitro in response to sunitinib or

hicle (n = 6) or sunitinib (n = 7) from day 55 to day 76 (after birth). (E) The graph

ors (T) of different sizes at the end of sunitinib treatment (D-sunitinib) and after

s. The graph corresponds to computerized quantification of metastasis size on

ages of mammary tumors fromMMTV-PyMTmice treated or not with sunitinib

ell density and distribution from the border to the center of the tumors (right)

as mean ± SD. *p % 0.05; **p % 0.01; ***p % 0.001.

ent with angiogenesis inhibitor (RTKIs). Left: during drug treatment (D-sunitinib

cancer cells in an autonomous manner or indirect effects through induction of

ucose consumption as revealed by lactate formation. Tumors preferentially use

drogenase kinase-1 and -2 (PDK1 and PDK2) expressions are increased in

) and the consequent blockade of aerobic glucose oxidation in the tricarboxylic

both MCT1 for lactate uptake and MCT4 for lactate export and stromal cells

y. Right: after drug withdrawal (W-sunitinib tumors), vascularized tumors still

at convert lactate to pyruvate and vice versa. While glycolysis is inhibited in

2 expressions are increased. In W-sunitinib tumors, cells adapt toward lipid

fter sunitinib treatment withdrawal. Our working model is that by blocking lipid

an be blocked, restoring the beneficial effects of sunitinib.

l Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc. 291

Page 13: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

(legend on next page)

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

292 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc.

Page 14: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

RNA Extraction, Semiquantitative RT-PCR, andMicroarray Analyses

See the Supplemental Experimental Procedures for details.

Proteomic Analysis

Preparation of tumor extracts for western blot and label-free mass spec-

trometry quantification are described in the Supplemental Experimental

Procedures.

Nuclear Magnetic Resonance Analyses

Sample preparation and NMR measurements are described in detail in the

Supplemental Experimental Procedures.

Immunohistochemistry

Detailed procedures for IHC and computerized quantification are described in

the Supplemental Experimental Procedures.

Statistical Analysis

Mann-Whitney test, one-way ANOVA test, and t test were used as described

in the Supplemental Experimental Procedures. The error bars represented in

each figure aremean ± SDormean ± SEM, and respective analysis is indicated

in the figure legends. p < 0.05 was considered statistically significant.

ACCESSION NUMBERS

The NCBI GEO accession number for the microarray results reported in this

paper is GSE50795.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Supplemental Experimental Procedures,

six figures, and one table and can be found with this article online at http://

dx.doi.org/10.1016/j.cmet.2014.05.022.

AUTHOR CONTRIBUTIONS

N.E.S. and J.C. contributed equally to this work. N.E.S. and J.C. performed the

experiments and analyzed the data. N.E.S. and A.N. designed the study and

wrote the manuscript. The other authors of this research paper also directly

participated in the planning, execution, or analysis of the study. They also

read and approved the final version of the submitted manuscript.

ACKNOWLEDGMENTS

This work was supported by grants from the Action de Recherche Concertee

(ARC) (Universite de Liege), the Fonds de la Recherche Scientifique-FNRS

(F.R.S.-FNRS), the Foundation against Cancer (foundation of public interest),

the Centre Anticancereux pres l’Universite de Liege, the Fonds Leon Fredericq

(University of Liege), the Direction Generale Operationnelle de l’Economie, de

l’Emploi et de la Recherche from the S.P.W. (RegionWallonne), the Interuniver-

sity Attraction Poles Programme - Belgian Science Policy (all from Belgium).

Figure 7. The Blockade of FASNReversed the Tumor Adaptation and Ac

Withdrawal

(A–E) Orlistat was used to inhibit FASN activity. (A) Evolution of tumor volume cur

xenografts and treated as described in the Experimental Procedures: (1) mice tr

sunitinib (Orlistat + Vehicle S), (3) mice treated with sunitinib and vehicle for orlista

sunitinib and followed with orlistat after sunitinib withdrawal (W-sunitinib + Orlist

corresponds to the initiation of treatment with orlistat. The graph below corresp

organs. (C) Metabolic analysis by NMR of mice sera showing identical metabolom

(E) Immunohistochemical stainings for FASN, perilipin, and carbonic anhydrase (C

groups. Scale bar, 100 mm. The graphs represent a computerized quantification

(F–I) Downregulation of FASN by shRNA inMDA-MB-231 cells. (F) Western blot an

nontargeting shRNA (sh CTR) or shRNA directed against FASN (shFASN). (G) Evo

for 30 days and followed by its withdrawal. (H) Images illustrating tumor sizes at th

organs.

Data are presented as mean ± SD for tumor growth; statistical analysis with dash

For bioluminescence quantification, data are presented as mean ± SEM; p % 0.

Cel

We acknowledge technology platforms of the GIGA (University of Liege) (ani-

mal, proteomic, and bioinformatics facilities) and Nice-Sophia Antipolis Func-

tional Genomics Platform. We acknowledge the technical support of Guy

Roland, Patricia Gavitelli, Emilie Feyereisen, Isabelle Dasoul, Nathalie Lefin,

and Marie Dehuy. J.C. and A.P. are supported by a F.R.S.-FNRS-Televie.

I.P. is supported by the Marie Curie Actions of the EU’s Seventh FP7/2007-

2013/ under REA grant CAFFEIN. The authors declare no conflict of interest.

Received: September 30, 2013

Revised: April 1, 2014

Accepted: May 15, 2014

Published: July 10, 2014

REFERENCES

Adams, V.R., and Leggas, M. (2007). Sunitinib malate for the treatment of met-

astatic renal cell carcinoma and gastrointestinal stromal tumors. Clin. Ther. 29,

1338–1353.

Bruce, J.Y., Kolesar, J.M., Hammers, H., Stein, M.N., Carmichael, L., Eickhoff,

J., Johnston, S.A., Binger, K.A., Heideman, J.L., Perlman, S.B., et al. (2014). A

phase I pharmacodynamic trial of sequential sunitinib with bevacizumab in

patients with renal cell carcinoma and other advanced solid malignancies.

Cancer Chemother. Pharmacol. 73, 485–493.

Butler, E.B., Zhao, Y., Munoz-Pinedo, C., Lu, J., and Tan, M. (2013). Stalling

the engine of resistance: targeting cancer metabolism to overcome therapeu-

tic resistance. Cancer Res. 73, 2709–2717.

Currie, E., Schulze, A., Zechner, R., Walther, T.C., and Farese, R.V., Jr. (2013).

Cellular fatty acid metabolism and cancer. Cell Metab. 18, 153–161.

Ebos, J.M., and Kerbel, R.S. (2011). Antiangiogenic therapy: impact on inva-

sion, disease progression, and metastasis. Nat Rev Clin Oncol 8, 210–221.

Ebos, J.M., Lee, C.R., Cruz-Munoz, W., Bjarnason, G.A., Christensen, J.G.,

and Kerbel, R.S. (2009a). Accelerated metastasis after short-term treatment

with a potent inhibitor of tumor angiogenesis. Cancer Cell 15, 232–239.

Ebos, J.M., Lee, C.R., and Kerbel, R.S. (2009b). Tumor and host-mediated

pathways of resistance and disease progression in response to antiangiogenic

therapy. Clin. Cancer Res. 15, 5020–5025.

Feron, O. (2009). Pyruvate into lactate and back: from the Warburg effect

to symbiotic energy fuel exchange in cancer cells. Radiother. Oncol. 92,

329–333.

Giuliano, S., and Pages, G. (2013). Mechanisms of resistance to anti-angio-

genesis therapies. Biochimie 95, 1110–1119.

Griffioen, A.W., Mans, L.A., de Graaf, A.M., Nowak-Sliwinska, P., de Hoog,

C.L., de Jong, T.A., Vyth-Dreese, F.A., van Beijnum, J.R., Bex, A., and

Jonasch, E. (2012). Rapid angiogenesis onset after discontinuation of sunitinib

treatment of renal cell carcinoma patients. Clin. Cancer Res. 18, 3961–3971.

Jain, R.K., Duda, D.G., Clark, J.W., and Loeffler, J.S. (2006). Lessons from

phase III clinical trials on anti-VEGF therapy for cancer. Nat. Clin. Pract.

Oncol. 3, 24–40.

quisition of theMalignant Phenotype Induced by Sunitinib Treatment

ves in four experimental groups of RAG1�/� mice bearing MDA-MB-231 LUC+

eated with vehicles (Vehicle S/O), (2) mice treated with orlistat and vehicle for

t after sunitinib withdrawal (W-sunitinib + Vehicle O), and (4) mice treated with

at). The dashed line indicates the period of sunitinib treatment, and the arrow

onds to mice weight measurements. (B) Quantification of bioluminescence in

e in the four experiment groups. (D) Lactate levels measured by NMR in tumors.

A)-IX and their quantifications (graph) in tumor sections from four experimental

in whole-scanned tumor sections.

alysis of FASN production inMDA-MB-231 LUC+ cells stably transfectedwith a

lution of tumor volumes in RAG1�/� mice treated with sunitinib (40 mg/kg/day)

e end of the experiment. (I) Quantification of metastasis by bioluminescence in

ed line compares vehicle to orlistat group; *p% 0.05; **p% 0.01; ***p% 0.001.

05.

l Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc. 293

Page 15: Blocking Lipid Synthesis Overcomes Tumor Regrowth and ... · Tumor Regrowth and Metastasis after Antiangiogenic Therapy Withdrawal Nor Eddine Sounni,1,5,* Jonathan Cimino,1,2,5 Silvia

Cell Metabolism

Cancer Metabolism Shift and Antiangiogenic Drugs

Kim, J.W., Tchernyshyov, I., Semenza, G.L., and Dang, C.V. (2006). HIF-1-

mediated expression of pyruvate dehydrogenase kinase: a metabolic switch

required for cellular adaptation to hypoxia. Cell Metab. 3, 177–185.

Nakajima, I., Muroya, S., Tanabe, R., and Chikuni, K. (2002). Extracellular ma-

trix development during differentiation into adipocytes with a unique increase

in type V and VI collagen. Biol. Cell 94, 197–203.

Noel, A., De Pauw-Gillet, M.C., Purnell, G., Nusgens, B., Lapiere, C.M., and

Foidart, J.M. (1993). Enhancement of tumorigenicity of humanbreast adenocar-

cinomacells in nudemicebymatrigel andfibroblasts.Br. J.Cancer68, 909–915.

Paez-Ribes, M., Allen, E., Hudock, J., Takeda, T., Okuyama, H., Vinals, F.,

Inoue, M., Bergers, G., Hanahan, D., and Casanovas, O. (2009).

Antiangiogenic therapy elicits malignant progression of tumors to increased

local invasion and distant metastasis. Cancer Cell 15, 220–231.

Schoors, S., De Bock, K., Cantelmo, A.R., Georgiadou, M., Ghesquiere, B.,

Cauwenberghs, S., Kuchnio, A., Wong, B.W., Quaegebeur, A., Goveia, J.,

et al. (2014). Partial and transient reduction of glycolysis by PFKFB3 blockade

reduces pathological angiogenesis. Cell Metab. 19, 37–48.

Sennino, B., Ishiguro-Oonuma, T., Wei, Y., Naylor, R.M., Williamson, C.W.,

Bhagwandin, V., Tabruyn, S.P., You, W.K., Chapman, H.A., Christensen,

J.G., et al. (2012). Suppression of tumor invasion andmetastasis by concurrent

inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors.

Cancer Discov 2, 270–287.

Singh, M., Couto, S.S., Forrest, W.F., Lima, A., Cheng, J.H., Molina, R., Long,

J.E., Hamilton, P., McNutt, A., Kasman, I., et al. (2012). Anti-VEGF antibody

therapy does not promote metastasis in genetically engineered mouse tumour

models. J. Pathol. 227, 417–430.

294 Cell Metabolism 20, 280–294, August 5, 2014 ª2014 Elsevier Inc

Sonveaux, P., Vegran, F., Schroeder, T., Wergin, M.C., Verrax, J., Rabbani,

Z.N., De Saedeleer, C.J., Kennedy, K.M., Diepart, C., Jordan, B.F., et al.

(2008). Targeting lactate-fueled respiration selectively kills hypoxic tumor cells

in mice. J. Clin. Invest. 118, 3930–3942.

Sounni, N.E., and Noel, A. (2013). Targeting the tumor microenvironment for

cancer therapy. Clin. Chem. 59, 85–93.

Sounni, N.E., Devy, L., Hajitou, A., Frankenne, F., Munaut, C., Gilles, C.,

Deroanne, C., Thompson, E.W., Foidart, J.M., and Noel, A. (2002).

MT1-MMP expression promotes tumor growth and angiogenesis through an

up-regulation of vascular endothelial growth factor expression. FASEB J. 16,

555–564.

Strumberg, D. (2005). Preclinical and clinical development of the oral

multikinase inhibitor sorafenib in cancer treatment. Drugs Today (Barc) 41,

773–784.

van der Veldt, A.A., Meijerink, M.R., van den Eertwegh, A.J., Bex, A., de

Gast, G., Haanen, J.B., and Boven, E. (2008). Sunitinib for treatment of

advanced renal cell cancer: primary tumor response. Clin. Cancer Res. 14,

2431–2436.

Vander Heiden, M.G. (2011). Targeting cancer metabolism: a therapeutic win-

dow opens. Nat. Rev. Drug Discov. 10, 671–684.

Welti, J.C., Powles, T., Foo,S.,Gourlaouen,M., Preece,N., Foster, J., Frentzas,

S., Bird, D., Sharpe, K., van Weverwijk, A., et al. (2012). Contrasting effects

of sunitinib within in vivo models of metastasis. Angiogenesis 15, 623–641.

Wu, S., Chen, J.J., Kudelka, A., Lu, J., and Zhu, X. (2008). Incidence and risk of

hypertension with sorafenib in patients with cancer: a systematic review and

meta-analysis. Lancet Oncol. 9, 117–123.

.