You are on page 1of 12

Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.

CAPR-11-0023

Cancer
Prevention
Research Article Research

Rapamycin Partially Mimics the Anticancer Effects of Calorie


Restriction in a Murine Model of Pancreatic Cancer
Laura M. Lashinger1,2, Lauren M. Malone2, Graham W. Brown2, Elizabeth A. Daniels2, Jason A. Goldberg2,
Glen Otto3, Susan M. Fischer1, and Stephen D. Hursting1,2

Abstract
Etiologic factors for pancreatic cancer, the 4th deadliest malignant neoplasm in the United States,
include obesity and abnormal glucose metabolism. Calorie restriction (CR) and rapamycin each affect
energy metabolism and cell survival pathways via inhibition of mammalian target of rapamycin (mTOR)
signaling. By using a Panc02 murine pancreatic cancer cell transplant model in 45 male C57BL/6 mice,
we tested the hypothesis that rapamycin mimics the effects of CR on pancreatic tumor growth. A
chronic regimen of CR, relative to an ad libitum-fed control diet, produced global metabolic effects such
as reduced body weight (20.6  1.6 g vs. 29.3  2.3 g; P < 0.0001), improved glucose responsiveness,
and decreased circulating levels of insulin-like growth factor (IGF)-1 (126  8 ng/mL vs. 199  11 ng/mL;
P 0.0006) and leptin (1.14  0.2 ng/mL vs. 5.05  1.2 ng/mL; P 0.01). In contrast, rapamycin
treatment (2.5 mg/kg intraperitoneal every other day, initiated in mice following 20 weeks of ad libitum
control diet consumption), relative to control diet, produced no significant change in body weight, IGF-1
or leptin levels, but decreased glucose responsiveness. Pancreatic tumor volume was significantly reduced
in the CR group (221  107 mm3; P < 0.001) and, to a lesser extent, the rapamycin group (374  206 mm3;
P 0.04) relative to controls (550  147 mm3), and this differential inhibition correlated with expression
of the proliferation marker Ki-67. Both CR and rapamycin decreased phosphorylation of mTOR, p70/S6K,
and S6 ribosomal protein, but only CR decreased phosphorylation of Akt, GSK-3b, extracellular signal
regulated kinase/mitogen-activated protein kinase, and STAT3TYR705. These findings suggest that rapamy-
cin partially mimics the anticancer effects of CR on tumor growth in a murine model of pancreatic cancer.
Cancer Prev Res; 4(7); 104151. 2011 AACR.

Introduction creatic cancer prevention and control. Calorie restriction


(CR) and the drug rapamycin are dietary and pharmaco-
Effective prevention and treatment strategies are urgently logic interventions, respectively, that act on pathways
needed for pancreatic cancer, the 4th leading cause of related to energy metabolism and inhibit various tumor
cancer-related death in both men and women in the United types (6, 7). To our knowledge, the effects of these inter-
States (1). Only 10% to 15% of pancreatic cancer patients ventions in pancreatic cancer have not been previously
have localized disease amenable to curative resection, and compared.
the overall 5-year survival rate in affected patients is less CR, typically involving a 20% to 40% reduction in total
than 5% (1, 2). Interrelated etiologic factors in pancreatic energy intake but isonutrient for vitamins, minerals, fatty
cancer include obesity, abnormal glucose metabolism, and acids, and amino acids relative to an ad libitum-fed control
positive energy balance (defined as caloric intake exceeding regimen, prevents or reverses obesity, improves insulin
energy expenditure; refs. 35). Components of energy sensitivity, and inhibits the development and/or progres-
metabolism pathways thus may be useful targets for pan- sion of many types of cancer (7). In animal models, the
anticancer effects of CR are strongly associated with reduc-
tions in circulating levels of the nutrient-responsive mito-
Authors' Affiliations: 1Department of Molecular Carcinogenesis, Univer- gen insulinlike growth factor (IGF)-1 (8, 9). For example,
sity of Texas M.D. Anderson Cancer Center, Smithville; 2Departments of CR suppresses the development and pathological severity
Nutritional Sciences and 3Molecular Genetics and Microbiology, Univer- of murine COX-2driven pancreatitis and pancreatic can-
sity of Texas at Austin, Austin, Texas
cer through a reduction of circulating IGF-1 levels (10).
Corresponding Author: Stephen D. Hursting, Department of Nutritional
Sciences, Dell Pediatric Research Institute, University of Texas at Austin, Epidemiologic evidence shows that (i) pancreatic cancer
1400 Barbara Jordan Blvd., Mail Code R1800, Austin, TX 78723. Phone: patients, relative to healthy controls, have higher levels of
512-475-3020; Fax: 512-475-4945. E-mail: shursting@mail.utexas.edu serum IGF-1 and pancreatic expression of the IGF-1 recep-
doi: 10.1158/1940-6207.CAPR-11-0023 tor (IGF-1R), and (ii) pancreatic cancer patients with ele-
2011 American Association for Cancer Research. vated circulating IGF-1, relative to patients with normal

www.aacrjournals.org 1041

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Lashinger et al.

levels, have a worse prognosis (11, 12). IGF-1 exerts its Diets; #D03020702) consumed in daily aliquots to provide
effects on cellular growth and metabolism, at least partially 70% of the energy and 100% of all nutrients (except
through activation of the phosphatidylinositol 3-kinase carbohydrates) relative to the control group, n 18. Food
(PI3K)/Akt/mammalian target of rapamycin (mTOR) path- intake and body weights were recorded weekly until
way (13, 14). Panc02 tumors became palpable (27 weeks of study).
Despite the substantial evidence highlighting the bene- At 16 weeks of study, 5 mice from each diet group were
ficial effects of CR, maintaining this lifestyle in humans is fasted for 12 hours then anesthetized by CO2 inhalation.
extremely challenging (7). This has prompted an intense They then underwent cardiac puncture for blood collection
search for CR mimetics, which are pharmacologic agents and subsequently were killed by cervical dislocation. After
that possess anticancer effects similar to CR without coagulating at room temperature (RT) for 30 minutes,
restricting energy intake (7). One putative CR mimetic is blood samples were centrifuged at 9,300 g for 5 minutes.
rapamycin (7). Unlike CR, rapamycin and its analogs affect Serum was separated, then snap-frozen and stored at
neither body weight nor energy balance (15). However, 80 C until assayed for hormones. Tissues were collected
rapamycin and CR each suppress signaling through mTOR and flash-frozen for subsequent molecular and biochem-
and extend lifespan in both invertebrates and mammalian ical studies.
species (7, 1618). The lifespan extension in mammals is At 20 weeks of study, the remaining mice in the control
primarily due to reduced tumor development (17). mTOR group (n 22) were randomized to receive (every other
acts as a sensor that integrates growth factor signals, nutri- day) an intraperitoneal (i.p.) injection of vehicle (0.1%
ent availability, and energy status with translational control dimethyl sulfoxide in 0.9% saline; n 11) or 2.5 mg/kg
of new proteins (19). Pharmacologic inhibition of mTOR rapamycin (n 11). The remaining mice in the CR group
by rapamycin halts DNA synthesis and proliferation of (n 13) also began receiving vehicle via i.p. injection every
pancreatic cancer cells in vitro through regulation of ribo- other day. This dose was chosen based on reports in the
somal S6 kinase and other downstream targets (20, 21). literature showing effective tumor inhibition with rapamy-
Rapamycin treatment in various xenograft models of pan- cin between 1 to 10 mg/kg daily or every other day, with the
creatic cancer in immunodeficient mice inhibits tumor lower doses showing comparable antitumor effects without
burden by impeding cell proliferation and angiogenesis the toxicity of the higher doses (15, 28). For our study, a
while promoting apoptosis (22, 23). In addition, recent moderate dose was selected because of the extended treat-
studies using murine prostatic, lung, and mammary cancer ment period.
models support a role for rapamycin and its analogs in
cancer chemoprevention (2427). Tumor cell injection and tumor monitoring
In the present study, we tested the hypothesis that Mouse pancreatic cancer cells (Panc02, generously pro-
rapamycin mimics the effects of CR on pancreatic tumor vided by Dr. K. Hance and Dr. J. Schlom, NCI; ref. 29) were
growth using a Panc02 pancreatic tumor cell transplant cultured under an atmosphere of 5% CO2 in a 37 C
model. We found that (i) chronic CR decreases pancreatic incubator with McCoys media (HyClone) supplemented
tumor growth in association with a reduction in prosurvi- with 10% fetal bovine serum (HyClone), penicillin/strep-
val signaling; (ii) rapamycin also reduces tumor growth tomycin, glutamine, nonessential amino acids, sodium
through targeted inhibition of the mTOR pathway; and (iii) pyruvate, and HEPES. Cells were trypsinized, washed in
rapamycin-induced mTOR inhibition, although effective, Hanks Buffered Saline Solution (HBSS), centrifuged, and
does not completely mimic the effects of CR on tumor resuspended in HBSS for injection. Karyotyping and species
growth inhibition, metabolism, or cell signaling in a pan- identification of Panc02 cells were verified by the Mole-
creatic cancer cell transplant model. cular Cytogenetics Core Facility at The U.T. M.D. Anderson
Cancer Center (Houston, TX).
Materials and Methods At 22 weeks of study, mice were s.c. injected into the right
flank with 1  106 Panc02 cells. One mouse from the
Mice control and CR groups each and 2 mice from the control
The Institutional Animal Care and Use Committee of the plus rapamycin (CONRapa) group died before tumor
University of Texas, Austin, TX, approved all mouse experi- development within 2 weeks of Panc02 injection, and were
ments. Male C57BL/6 mice were received from Jackson thus censored from subsequent analyses. Once palpable,
Laboratories (Bar Harbor, ME) between 4 to 6 weeks of age, tumors were measured with calipers weekly until approxi-
singly housed in a semibarrier facility at the Animal mately half of tumors from any group were 1.5 cm in
Resource Center, University of Texas at Austin, and fed a diameter. Tumor volume was approximated using the
chow diet for a 2-week acclimation period before study formula for an ellipsoid (4/3pr12r2). At study termination,
initiation. all remaining mice were fasted for 12 hours then anesthe-
tized by CO2 inhalation. They then underwent cardiac
Interventions puncture for blood collection and subsequently were killed
Mice were randomized to receive 1 of 2 diets for 30 by cervical dislocation. Blood samples were processed as
weeks: (i) control diet (Research Diets, Inc.; #D12450B) detailed earlier. Pancreatic tumors were harvested and
consumed ad libitum, n 27; or (ii) CR diet (Research either snap-frozen in liquid nitrogen and stored at

1042 Cancer Prev Res; 4(7) July 2011 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Rapamycin and Calorie Restriction Inhibit Pancreatic Cancer

80 C, or fixed with 10% neutral buffered formalin over- p-STAT3Ser727 (Cell Signaling; 1:50, 4 C overnight);
night and then switched to 70% ethanol, paraffin STAT3 (Cell Signaling; 1:100; 1 hour RT); p-mTORSer2448
embedded, and used for histologic and immunohisto- (Cell Signaling; 1:50, 4 C overnight); mTOR (Cell Signal-
chemical analyses as described before. ing; 1:100; 1 hour RT); p-S6 ribosomal protein(Ser235/236)
(Cell Signaling; 1:50, 1 hour RT); S6 (Cell Signaling;
Glucose tolerance test 1:100; 4 C overnight); and cyclin D1 (Santa Cruz Bio-
A glucose tolerance test (GTT) was done, as previously technology; 1:500, 2 hours RT). Slides were washed twice
described (30), on randomly selected mice from the con- in PBS, incubated with horseradish peroxidase (HRP)
trol and CR groups at 14 weeks of study (n 12 per group). labeled a-rabbit secondary antibody (Dako) for 30 min-
At 22 weeks of study, following 2 weeks of rapamycin or utes at RT at a concentration of 1:200 with the following
vehicle treatment and before Panc02 cell injection, a sec- exceptions: (a) Ki-67 [rabbit anti-rat immunoglobulin
ond GTT was done on a subset of 10 mice from each diet/ (Ig) G; 1:200, 30 minutes RT; Vector]; (b) p-STAT3 and
drug group. p-mTOR (rabbit HRP-polymer; 30 minutes RT; Biocare);
and (c) cyclin D1 (rabbit anti-mouse F(ab)0 ; 1:250, 15
Energy balancerelated serum hormones minutes RT; Accurate Chemical). Slides were then washed
Serum IGF-1 was measured using radioimmunoassay 5 times with PBS. Diaminobenzidine was used to develop
according to manufacturers instructions (DSL-2900 Kit, the antibody staining followed with a hematoxylin coun-
DSL/Beckman Coulter Laboratories, Webster, TX). Serum terstain. Images were captured using a light microscope
insulin and leptin were measured using Lincoplex bead equipped with a digital camera (Leica Camera, Inc.).
based assays (Millipore Corporation) on a BioRad Bioplex Immunohistochemically stained tumor sections were
analyzer (BioRad) according to manufacturers directions. assigned a score based on the following criteria: 1 mild
Analyses were conducted on samples obtained at 16 weeks, (majority of tumor section exhibited weakly positive
before Panc02 cell injection (to assess diet-induced changes staining pattern or less than 50% of cells in field stained
in the absence of potentially confounding tumor effects on an equivalent intensity to control tumors), 2 moderate
circulating hormone levels; n 5 per group) and at study (majority of tumor section exhibited moderate-to-dark
termination (to exclude the possibility that rapamycin brown stain or 50% to 75% of cells in field stained an
affected circulating levels within the control group; control, equivalent intensity to control tumors), or 3 marked
n 6; rapamycin-treated control, n 3). (majority of tumor section exhibited dark brown stain or
greater than 75% of cells in field stained an equivalent
Histopathology and immunohistochemistry intensity to control tumors). The quality of staining
Formalin-fixed pancreatic tumors were embedded in determined whether the intensity of stain or percentage
paraffin and then cut into 4-mm-thick sections and pro- of stained cells in field was quantified. For example, the
cessed for either hematoxylin and eosin (H&E) or immu- antibodies against p-IGF1-R and p-ERK produced weak
nohistochemical staining at the Histology Core but consistent signal intensity; therefore, the percentage
Laboratory at The U.T. M.D. Anderson Cancer Center, of stained cells in field was assessed. Sample size was
Science Park Research Division (Smithville, TX). Antibo- determined based on the quality of tissue fixation/stain-
dies used for immunohistochemistry were optimized by ing. More tumors were used to analyze phosphorylated
core personnel using both positive and negative controls IGF-1R, Akt, GSK-3b, ERK, STAT3, mTOR, and S6 ribo-
that were repeated with each analysis. Slides were depar- somal protein antibodies, and total cyclin D1 antibody
affinized and hydrated sequentially in ethanol to water. (for each antibody: control, n 4; rapamycin-treated
Antigen retrieval required microwaving slides for 10 control, n 7; CR, n 5) than Ki-67 (n 3/group)
minutes with 10 mmol/L citrate buffer. Endogenous because the latter yielded a more easily quantifiable
peroxidase activity was quenched with 3% hydrogen nuclear staining pattern. Images were captured at 40
peroxide for 10 minutes. Nonspecific binding was magnification (with the exception of cyclin D1 which was
blocked with Biocare blocking reagent (Biocare Medical) captured at 20) in 3 to 4 fields in a tumor section from
for 30 minutes at RT, then sections were incubated with each tumor analyzed.
primary antibody diluted in blocking buffer. The follow-
ing primary antibodies (source, dilution, and incubation Western blotting
conditions presented parenthetically) were used: Ki-67 Pancreatic tumors were homogenized and lysed in radio-
(Dako; 1:200, 4 C overnight); phospho (p)-IGF-1RTyr1131 immunoprecipitation assay buffer (Sigma) with protease
and IGF-1R (Cell Signaling; 1:50, 4 C overnight); inhibitor tablet (Roche Applied Sciences) and phosphatase
p-AktSer473 (Santa Cruz Biotechnology; 1:50, 1 hour inhibitor cocktails I and II (Sigma). Protein lysates (5080
RT); Akt (Cell Signaling; 1:100; 4 C overnight) p-GSK- mg) were resolved by SDS-PAGE using 6%, 10%, or 12%
3bSer9 (Santa Cruz Biotechnology; 1:50, 1 hour RT); GSK- gels, transferred to polyvinylidene difluoride membranes
3b (Millipore; 1:200; 1 hour RT); p-extracellular signal (Bio-Rad), and blocked using LI-COR Blocking Buffer
regulated kinase (ERK)Thr202/Tyr204 (Cell Signaling; 1:100; (LI-COR Biotechnologies). Membranes were incubated
1 hour RT); ERK (Cell Signaling; 1:25; 1 hour RT); overnight at 4 C with primary antibody (from Cell Signal-
p-STAT3Tyr705 (Cell Signaling; 1:50; 4 C overnight); ing unless otherwise stated) diluted in blocking buffer and

www.aacrjournals.org Cancer Prev Res; 4(7) July 2011 1043

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Lashinger et al.

specific for: p-AktSer473 (1:500); Akt (1:1,000); actin bolus injection in CR mice peaked at 15 minutes and
(1:1,000); p-GSK-3bSer9 (Santa Cruz, 1:1,000); GSK-3b averaged 332  78 mg/dL, whereas the control group
(Millipore, 1:1,000); p-ERKThr202/Tyr204 (1:1,000); ERK peaked at 30 minutes and averaged 510  73 mg/dL (n
(1:1,000); p-mTORSer2448 (1:500); mTOR (1:1,000); 12 per group). After the peaks were achieved, blood
p-p70/S6KThr389 (1:500); p70/S6K (1:1,000); and myosin glucose concentrations remained consistently lower in the
IIa (1:1,000). Actin was used as a loading control for all CR mice (P < 0.0001 for between-group comparison at the
antibodies except mTOR (for which myosin IIa was used 120-minute measurement). Comparable diet-dependent
because the 6% polyacrylamide gel used to resolve mTOR effects on glucose tolerance were noted at 22 weeks of
did not retain actin). After 3 washes (5 minutes each) in study, at which time the CR and control mice had been
0.1% Tween-20/PBS (PBS-T), membranes were incubated receiving, for 2 weeks, vehicle by i.p. injection every other
for 1 hour at RT in species-specific secondary antibody (LI- day (Fig. 1E; n 10 each).
COR Biotechnologies) diluted in LI-COR blocking buffer In a subset of mice fed for 16 weeks and then bled (n 5
(1:5,000). Following 3 washes in PBS-T, membranes were per group), the CR mice had significantly lower serum
scanned using the Odyssey infrared fluorescent imaging levels of IGF-1 (126  8 ng/mL; P 0.0006) and leptin
system. Densitometry was performed using LI-COR Soft- (1.14  0.17 ng/mL; P 0.01) than control mice (199 
ware (LI-COR Biotechnologies). Relative expression of 11 ng/mL and 5.05  1.18 ng/mL, respectively; Fig. 1D).
phosphorylated proteins was calculated by using 3 tumors No between-group difference in serum insulin levels was
per group. detected (P 0.15).

Statistical analyses Effects of rapamycin on body weight, glucose


Data are presented as mean  SD, except serum hormone tolerance, and serum hormones
levels and Ki-67 which are presented as mean  SEM. Beginning at 20 weeks of study, the control mice
Statistical analyses were conducted using SPSS (Apache received their assigned diet plus i.p. injections every other
Software Foundation). Temporal differences between day of either 2.5 mg/kg rapamycin [for CONRapa
groups with respect to body weight and caloric intake were group, n 9] or vehicle (CON, n 10). The CR group
assessed using repeated measures analysis; final measure- (n 12) also received vehicle. Moderate reduction of
ments were compared using independent t tests. Pretumor caloric intake in the control group was noted between
serum hormone levels between the control and CR groups weeks 22 and 24 relative to the CONRapa group
were compared using independent t tests. Pairwise com- (Fig. 1A). However, this decrease did not approach sta-
parisons of glucose tolerance and tumor burden as a tistical significance, nor did it significantly affect body
function of time and treatment group were performed weight (Fig. 1B). This modest differential between the 2
using a linear mixed effects model. Final tumor volume groups subsided by week 25. Between weeks 20 to 27 of
measurements were compared using independent t tests. study, body weights remained stable within each diet/
Differences between groups in immunohistochemical drug group and at 27 weeks were comparable between the
staining of (i) Ki-67 were determined by 1-way ANOVA control mice (29.3  2.3 g) and rapamycin-treated mice
followed by Tukeys post hoc test of significance and (ii) all (32.8  2.1 g; Fig. 1A). As previously mentioned, the CR
other antibodies were determined by Fisher exact test. mice were leaner than the others.
Differences in Western blot densitometry between each The average peak glucose levels in the rapamycin-treated
test group (rapamycin-treated controls and CR), relative mice (530  130 mg/dL) occurred 60 minutes after the
to the control group, were compared by independent t tests. glucose bolus was administered, whereas control mice
Results were considered significant if P < 0.05. receiving vehicle peaked at 30 minutes and only reached,
on average, 441  80 mg/dL (Fig. 1E). Moreover, blood
Results glucose levels at the final time point (120 minutes after
glucose bolus) were significantly higher in rapamycin-treated
Effects of CR on body weight, glucose tolerance, and mice (462  169 mg/dL; P 0.02) relative to the control plus
serum hormone levels vehicle group (309  87 mg/dL). Consistent with the GTT
Male C57BL/6 mice were fed either a control diet known results at 14 weeks, CR mice, as compared with the others,
to result in an overweight phenotype or a 30% CR diet (7) exhibited the lowest levels of glucose at all points including
for 30 weeks (including 22 weeks of diet before Panc02 cell the final assessment (151  24 mg/dL; P < 0.0001).
injection). Relative to controls, the CR mice had signifi- To exclude the possibility that rapamycin impacts circu-
cantly reduced body weights beginning as early as 3 weeks lating energy balancerelated hormones relative to the con-
on study (P < 0.01; Fig. 1A and B). Diet-dependent effects trol plus vehicle group, serum collected at study termination
on body weight continued throughout the study and at 27 was assessed for levels of IGF-1, insulin, and leptin (Fig 1F).
weeks, mean body weights were 20.6  1.6 g in CR mice No statistical differences (all P > 0.15) were detected
(n 12; P < 0.0001) and 29.3  2.3 g in controls (n 10). between rapamycin-treated mice (n 3) and controls
The CR group, relative to control, displayed enhanced (n 6), respectively, in circulating levels of IGF-1 (176 
glucose tolerance as assessed by GTT. At 14 weeks of study 14 vs. 147  21 ng/mL), insulin (1.9  0.3 vs. 1.2 
(Fig. 1C), blood glucose concentrations following glucose 0.3 ng/mL), or leptin (2.0  0.6 vs. 1.9  0.2 ng/mL).

1044 Cancer Prev Res; 4(7) July 2011 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Rapamycin and Calorie Restriction Inhibit Pancreatic Cancer

GTT #2/
Tumor
injection
A Rapa Tumor B
GTT #1 initiated palpable

100 40

Average body weight (g)


Average caloric intake

80 30
(kcal)

60
* 20 *
40
CON
CON + Rapa 10 CON
20 CON + Rapa
CR
CR
0 0
0 4 8 12 16 20 24 28 0 4 8 12 16 20 24 28
Weeks on diet Weeks on diet
C D Serum hormone levels prior to tumor injection (16 wk)
14 wk GTT
Blood glucose (mg/dL)

600 CON 250 0.5 7

Leptin levels (ng/mL)


Insulin levels (ng/mL)
IGF-1 levels (ng/mL)

500 CR 6
200 0.4
400 5
150 0.3
300 * 4
3
200 100 0.2

100 * 50 0.1
2
1 *
0 0 0.0 0
0 15 30 60 120 CON CR CON CR CON CR
Time after glucose injection
(min)
E F Serum hormone levels in control groups at study termination
22 Week GTT (2 wk after rapa initiated)
Blood glucose (mg/dL)

600 200 2.5


Insulin levels (ng/mL)

Leptin levels (ng/mL)


IGF-1 levels (ng/mL)

500 b
CON 150 2.0
400 CON + Rapa 2
a 1.5
300 CR 100
1.0
200 1
c 50
0.5
100
0 0 0.0 0
0 15 30 60 120 CON CON + Rapa CON CON + Rapa CON CON + Rapa

Time after glucose injection


(min)

Figure 1. Effects of CR versus rapamycin on food intake, body weight, glucose tolerance, and serum hormone levels. A and B, C57BL/6 male mice fed
a CR diet (n 12) for 27 weeks consumed fewer calories and weighed significantly less than mice fed a control diet (CON; n 10). Within the mice
receiving control diet, a 10-week rapamycin treatment (CONRapa; n 9), initiated at 20 weeks of study, had no significant effect on caloric intake or body
weight relative to the control mice administered vehicle (n 10). C, GTT performed on a subset of mice fed either a control or CR diet for 14 weeks
(n 12/group). D, pretumor serum levels of IGF-1, insulin, and leptin in a subset of mice fed either a control or CR diet for 16 weeks (n 5/group). E, GTT
performed at 22 weeks of study (immediately prior to Panc02 cell injection) on mice fed CR or control diet and administered vehicle or rapamycin beginning
at week 20 of the study (n 10/group). F, posttumor serum levels (at study termination) of IGF-1, insulin, and leptin in mice receiving the control diet vehicle
(CON, n 6) versus the control diet rapamycin (CONRapa; n 3). A to C and E, error bars represent SD. D and E, error bars represent SEM. A to D,
significant differences (P < 0.05) are denoted with *. E, different letters denote significant differences. F, no values were significantly different.

Effects of CR and rapamycin on Panc02 tumor growth slower than in control mice receiving vehicle. CR exerted a
and histology more dramatic inhibitory effect than rapamycin as evi-
To compare the anticancer effects of CR and rapamycin denced by a significant difference between their tumor
interventions, we injected mice from each diet/drug group growth (P 0.005; Fig. 2A). Final tumor volumes from
with Panc02 cells at week 22 and monitored tumor growth rapamycin-treated mice (374  206 mm3; P 0.04) and
during the next 8 weeks. Tumors in rapamycin-treated mice CR mice (221  107 mm3; P < 0.0001) were significantly
(P 0.009) and CR mice (P < 0.0001) grew significantly smaller, on average, than controls (550  147 mm3). The

www.aacrjournals.org Cancer Prev Res; 4(7) July 2011 1045

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Lashinger et al.

A B

Tumor volume (mm3)


700 CON
600 CON + Rapa a Tumor incidence
500 CR
Week 5 Week 6 Week 7 Week 8
400 b
CON 30% 89% 100% 100%
300 CON + Rapa 22% 33% 88% 100%
c
200 CR 0% 17% 66% 100%
100
0
5 6 7 8
Weeks after injection
C CON CON + Rapa CR
D

Ki-67-positive cells/field
300
a
Ki67
200
b

100 c

0
CON CON + Rapa CR
H&E

Figure 2. Effect of CR versus rapamycin treatment on tumor growth and histology. A, weekly tumor measurements were taken using calipers beginning
when the first tumor became palapable (5 weeks) after Panc02 cells were s.c. injected into C57BL/6 mice receiving the control diet plus vehicle (CON, n 10),
control diet plus rapamycin (CON Rapa, n 9), or CR diet plus vehicle (n 12). Error bars represent SD. B, percentage of mice in each diet/drug
group with palpable tumor at weeks 5, 6, 7, and 8 after Panc02 cell injection. C, immunohistochemical staining for Ki-67 expression and H&E staining
of tumors. Scale bars for Ki-67, 100 mm and H&E, 400 mm. Arrows indicate adipocytes. D, quantification of Ki-67positive cells from representative
images captured at 40 magnification. Data are expressed as mean  SEM number of Ki-67positive cells/field in tumors (n 3). Values with different
letters are significantly different at P < 0.05.

CR and rapamycin interventions each delayed tumor onset, (P 0.01), mTOR (P 0.02), S6 ribosomal protein (P
with only 17% and 33% of mice, respectively, having 0.004), GSK-3b (P 0.06), IGF-1R (P 0.11), and
palpable tumors 6 weeks after Panc02 cell injection com- cyclin D1 (P 0.11; Fig. 3A and B). Rapamycin produced
pared to 89% of the control group (Fig. 2B). Only at study a more selective signaling profile than CR in which only
termination, did all mice in the CR and control diet plus p-STAT3Ser727 (P 0.02) and mTOR pathway components
rapamycin groups have detectable tumors. were inhibited relative to control, including p-mTOR (P
The influence of CR or rapamycin treatment on cell 0.02), p-S6 ribosomal protein (P 0.006; Fig. 3A and B),
proliferation was assessed in tumor tissues by immunohis- but not p-IGF-1R (P 1), p-Akt (P 0.3), p-GSK-3b (P
tochemical staining against Ki-67 (Fig. 2C and D). CR 1), p-ERK (P 0.54), or p-STAT3TYR705 (P 0.24). No
(74.8  9.9 positive cells/field; n 3; P 0.001) and appreciable changes to total protein expression were noted
rapamycin treatment (141.9  11.0 positive cells/field; (data not shown).
n 3; P 0.013) each significantly reduced cell prolifera- Immunoblotting of various signaling intermediates con-
tion (based on Ki-67 immunopositivity) relative to con- firmed the dampening of other survival signals in CR
trols (231.7  21.3 positive cells/field; n 3). CR tumors (Fig. 4A). CR (n 3) significantly reduced phos-
produced a more substantial reduction in proliferation phorylation of Akt (P 0.006), GSK-3b (P 0.04), ERK
(P 0.04) relative to rapamycin treatment. (P 0.004), mTOR (P 0.05), and p70/S6K (P 0.05)
The effect of CR and rapamycin intervention on tumor relative to the control diet which, despite an occasionally
histology was qualitatively assessed using H&E staining variable expression pattern, had higher levels of phos-
(Fig. 2C). There was a considerable presence of adipocytes phorylated proteins when averaged over 3 tumors
in the control tumors that were not seen in the CR tumors. (Fig. 4B). Rapamycin potently and consistently reduced
Unlike CR, rapamycin treatment did not lessen the level of phosphorylation of mTOR (P 0.005) and p70/S6K (P
adipocyte infiltration associated with the control diet. 0.02) when compared with control tumors, and decreased
phosphorylation of p70/S6K more robustly than did CR
Effects of CR and rapamycin on signaling (P 0.008; Fig. 4B). Despite the potent inhibitory effects of
intermediates rapamycin on mTOR signaling intermediates, levels of
Based on immunohistochemical analysis of signaling phosphorylated Akt, GSK-3b, and ERK were similar to
intermediates, CR tumors relative to control tumors dis- control tumors (Fig. 4A). Neither CR nor rapamycin had
played reduced expression of phosphorylated Akt (P a significant effect on total protein expression of Akt,
0.03), ERK (P 0.05), STAT3Tyr705 (P 0.02), STAT3Ser727 mTOR, p70/S6K, GSK-3b, or ERK (Fig. 4A).

1046 Cancer Prev Res; 4(7) July 2011 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Rapamycin and Calorie Restriction Inhibit Pancreatic Cancer

A CON CON + Rapa CR


B

p-IGF1R

pIGF1R pAkt
a a a a a b
p-Akt 100 pIGF1R 100 pAkt staining

percentage

percentage
75 staining score: 75 score:

Score
3 3

Score
50 2 50 2
25 1 25 1
0 0
CON CON + Rapa CR CON CON + Rapa CR
p-GSK3 pGSK3 pERK
a a a P = 0.06 100 a a b
100 pERK staining
pGSK3 staining

percentage
percentage
75 score:
75 score:
3

Score
Score
3 50
50
2 2
25 1 25 1
0 0
p-ERK CON CON + Rapa CR CON CON + Rapa CR

pSTATTYR705 pSTATSER727
a a b 100 a b b
100 pSTATTYR705 pSTATSER727

percentage
percentage

75 staining score: 75 staining score:

Score
Score

3 3
50 50 2
2
p-STAT3Tyr705 25 1 25 1
0 0
CON CON + Rapa CR CON CON + Rapa CR

pmTOR pS6
100
a b b a b b
pmTOR 100 pS6 staining
percentage

staining score: score:

percentage
75 75
Score

p-STAT3Ser727 3 3

Score
50 2 50 2
25 1 1
25
0 0
CON CON + Rapa CR
CON CON + Rapa CR

Cyclin D1
100
a a a
p-mTOR Cyclin D1
percentage

75 staining score:
Score

3
50 2
25 1
0
CON CON + Rapa CR

p-S6

Cyclin D1

Figure 3. Effect of CR and rapamycin on signaling pathways in pancreatic tumors by immunohistochemical analysis. A, immunohistochemical analyses
of phosphorylated IGF1R, Akt, GSK-3b, ERK, STAT3, mTOR, and S6 ribosomal protein as well as total protein expression of cyclin D1. Scale bars,
100 mm except cyclin D1, 200 mm. B, quanitification of antibody staining patterns. Tumors were assigned a score that represented the majority of the
section: 1 (mild white), 2 (moderate gray), or 3 (marked black). The bar graph represents the percentage of tumors in that group associated with the
corresponding score assignment (CON, n 4; CONRapa, n 7; CR, n 5). Values with different letters are significantly different at P < 0.05.

Discussion anticancer effects of a chronic CR dietary regimen versus an


acute pharmacological intervention, rapamycin, which
CR is effective at inhibiting cancer growth in many share several downstream signaling targets but have not
model systems (7). Although we found a strong anticancer been directly compared in a pancreatic cancer model. We
effect of CR in a COX-2driven transgenic model of pan- found in C57BL/6 mice that CR (relative to a control diet
creatic neoplasia (10), we sought to establish and under- fed ad libitum that results in overweight mice) reduced
stand the growth prohibitive effects of CR on transplanted Panc02 tumor burden. Furthermore, we established that
pancreatic tumors. In addition, we wanted to compare the exposure to rapamycin (at a dose of 2.5 mg/kg i.p. every

www.aacrjournals.org Cancer Prev Res; 4(7) July 2011 1047

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Lashinger et al.

A B
0.5

Relative phosphorylation
CON CON + Rapa CR CON
CON + Rapa
CR
0.4
p-Akt
0.3
Akt
0.2
*
Actin 0.1
* * Figure 4. Effect of CR and
0.0 rapamycin on signaling pathways
p-GSK3 pAkt pGSK3 pERK
in pancreatic tumors by
a immunoblot analysis. A,
GSK3 5 immunoblotting analyses of
b

phosphorylation
Relative mTOR
4 phosphorylated and total protein
b
Actin expression of Akt, GSK-3b, ERK,
3 mTOR, and p70/S6K. Data shown
2 are representative blots from at
p-Erk least 3 tumors for each treatment.
1 B, relative phosphorylation of
Erk p-Akt, p-GSK-3b, p-ERK, p-
0
CON CON + Rapa CR mTOR, and p-p70/S6K proteins,
Actin quantified by densitometry using
LI-COR Odyssey software. Data
0.4 a represent mean of 3 tumors/
p-mTOR
group; error bars represent SD.
Relative p70/S6K
phosphorylation

0.3 Significance denoted by * or


mTOR
different letters (P < 0.05).
Myosin 0.2 c
b
0.1
p-p70/S6k
0.0
p70/S6K CON CON + Rapa CR

Actin

other day, which effectively inhibited mTOR without toxi- tance, reduces pancreatic cancer risk in type II diabetics by
city) also significantly suppressed Panc02 tumor growth, 62% (32), and also prevents development of carcinogen-
although to a lesser extent than CR. Central to both inter- induced pancreatic lesions enhanced by a high-fat diet in
ventions is an ability to dampen signaling through the hamsters (33), suggesting improved glucose metabolism
mTOR pathway, a highly conserved protein at the crux of and/or mTOR inhibition may contribute to decreased
intracellular energy sensing and external growth factor pancreatic cancer development.
signaling. However, the unique ability of CR to alter multi- In addition to improved responsiveness to insulin in the
ple signaling pathways involved in proliferation and sur- CR group, there was a significant decrease in serum levels of
vival was more influential on pancreatic tumor growth IGF-1. This is important given the connection between
than the selective targeting of mTOR with rapamycin. pancreatic cancer risk and serum levels of IGF-1 in human
Our finding that chronic CR (30 weeks), relative to subjects (11, 12), and the implication that reduced IGF-1
control diet, inhibited Panc02 tumor growth in association underlies many of the anticancer effects of CR (8, 10). The
with improved insulin sensitivity and reduced circulating protective effects of reduced IGF-1 signaling in a skin carci-
IGF-1 and leptin, is consistent with established links nogenesis model have been associated with an abrogation
between altered metabolism and pancreatic cancer. Speci- in mTOR signaling (9). The current study recapitulates this
fically, there is a 2-fold higher risk for developing pancrea- connection between CR, reduced circulating IGF-1, and
tic cancer in the context of altered glucose metabolism, subsequently diminished Akt/mTOR signaling. It also
such as that occurring in diabetes (4, 31). Although fasting shows that CR dampened other components of prosurvival
levels of insulin were moderately but not statistically signaling such as GSK-3b, ERK, and STAT3. Although this is
decreased in the CR group (P 0.08), the functional not a comprehensive assessment of proliferation and survi-
sensitivity of CR mice to insulin was dramatically improved val signaling pathways, these findings suggest that the
relative to the control group, as indicated by GTT. The ability to suppress multiple pathways underlies the potent
antidiabetic biguanide metformin improves insulin resis- and broad-acting anticancer effects of CR.

1048 Cancer Prev Res; 4(7) July 2011 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Rapamycin and Calorie Restriction Inhibit Pancreatic Cancer

Our studies also aimed to establish and understand with a known diminution of an S6K-mediated feedback
the pancreatic cancer therapeutic potential of rapamy- inhibition that can result in Akt activation in response to
cin. Recently, several reports have offered evidence that mTOR inhibitors (4144). However, rapamycin reduced
mTOR inhibitors might have cancer preventive activity, phosphorylation of an mTOR-sensitive residue (serine
at least in part by mimicking some effects of CR (2427). 727) on p-STAT3 that is necessary for full transcriptional
For example, low-dose rapamycin treatment of HER-2/ activation of STAT3 (45). It is plausible that the transcrip-
neu mice significantly diminished tumor formation, tional activity of STAT3 (which we did not assess in our
with one third of mice having no detectable tumors at system) is dampened by rapamycin when compared with
sacrifice (26). In a mouse prostate model expressing control tumors, but not CR tumors, especially consider-
human AKT1, the rapamycin analog, RAD001, reversed ing CR resulted in reduced phosphorylation at both the
prostatic intraepithelial neoplasia by enhancing apop- TYR705 and SER727 sites.
tosis and abrogating expression of HIF-1a genes (25). A histological difference between tumors from CR and
Another rapamycin analog, CCI-779, abrogates the pro- rapamycin-treated mice was also noted. Adipocytes were
gression of lung adenomas that are induced by activating consistently present in the tumors from mice receiving
K-RAS mutations (2527, 34). Sensitivity to mTOR the control diet, regardless of whether they received
inhibition is enhanced in tumors that rely on PI3K/ rapamycin or vehicle. In contrast, CR clearly reduced
Akt signaling such as those with a deficiency in the adipocyte infiltration into tumors. Zhang and colleagues
phosphatase and tensin homolog tumor suppressor (46) showed that stromal and endothelial cells from
(3537). Excessive activation of the PI3K/AKT pathway adipose tissue migrate to tumors and promote tumor
may be largely responsible for the detrimental effects of growth, and endogenous white adipose tissue
obesity (38). Thus, it seems plausible that rapamycin (enhanced in an overweight or obese state) was the
treatment could overcome the growth-promoting effects source of the adipose progenitors infiltrating the tumors
of excessive energy intake. (46). Furthermore, studies show that coadministration
Our findings show that treatment with rapamycin cir- of tumor cells with mouse adipose cells promote tumor
cumvents some of the protumorigenic effects of the calorie- take and growth (47, 48). Further contributing to poten-
dense control diet. This effect is seen in our rapamycin- tial growth-promoting effects of local adipocytes is the
treated mice despite insulin resistance, elevated serum release of soluble adipokines/cytokines (49), one of
levels of IGF-1 and leptin, and other metabolic parameters which (leptin) was elevated in the serum of mice in
typically associated with enhanced tumor growth. Rapa- our study receiving the high-calorie control diet, irre-
mycin treatment actually resulted in a worsened sensitivity spective of treatment with rapamycin or vehicle. As
to a glucose bolus than the control group, despite having recently suggested by Subbaramaiah and colleagues,
no effect on caloric intake and body weights. The alteration multiple energy balancerelated signals contribute to
in glucose homeostasis in response to rapamycin is con- inflammatory cross talk between macrophages, adipo-
sistent with findings in the Psammomys obesus diabetic rat cytes, and epithelial tumor cells (50, 51).
model in which insulin resistance is enhanced by rapamy- Taken together, our findings show that CR had a
cin because of increased b cell apoptosis, decreased b cell stronger inhibitory effect on Panc02 tumor growth than
mass, and reduced glucose-induced insulin biosynthesis did rapamycin. CR and rapamycin decreased phosphor-
and secretion from islet cells (39). Moreover, chronic ylation of mTOR pathway components, but only CR
rapamycin treatment stimulates hepatic gluconeogenesis reduced circulating IGF-1 and leptin levels and phosphor-
(40), which could further disrupt glucose homeostasis. ylation of Akt, GSK-3b, ERK/mitogen-activated protein
These metabolic outcomes of mTOR inhibition potentially kinase, and STAT3bTYR705. Thus, treatment with rapamy-
enhance glucose bioavailability to cancer cells, possibly cin, which specifically inhibited mTOR and significantly
offsetting some of the growth-inhibitory effects of rapamy- decreased Panc02 tumor growth relative to control diet,
cin. Despite these metabolic alterations, including high only partially mimics (at the dose used) the more potent
levels of circulating mitogens, mTOR signaling was sub- effects of CR on multiple progrowth signals and pancrea-
stantially inhibited by rapamycin (even to a greater extent tic tumor growth inhibition. This observation that
than by CR), resulting in pancreatic tumor growth suppres- CR, relative to rapamycin, is a more potent suppressor
sion. This further supports the mTOR pathway as an of pancreatic tumor growth and is more promiscuous in
important target for pancreatic cancer prevention and terms of targeting multiple signaling pathways suggests
control. that combination approaches (such as an mTOR inhibi-
In addition to disparate responses to insulin and glu- tor plus a lifestyle or pharmacologic intervention that
cose between the CR and rapamycin interventions, sig- targets other key pathways) will be most effective for the
naling patterns were also divergent. Although both prevention and control of pancreatic cancer. Future stu-
interventions diminished mTOR signaling, rapamycin dies are being designed to verify the hypothesis that a
did not inhibit other survival signals such as ERK, GSK- synergistic effect is exerted on tumor inhibition when the
3b, and STAT3TYR705. Akt phosphorylation in the tumors broader acting effects of CR are combined with low-to-
from the rapamycin-treated group was generally higher intermediate doses of pharmacologic mTOR inhibitors
than in tumors from the control group. This is consistent such as rapamycin.

www.aacrjournals.org Cancer Prev Res; 4(7) July 2011 1049

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Lashinger et al.

Disclosure of Potential Conflicts of Interest Grant Support

No potential conflicts of interest were disclosed. R01 CA135386 (S. Hursting and S. Fischer); R25T CA57730; and T32
CA135386 (L. Lashinger).
The costs of publication of this article were defrayed in part by the
Acknowledgments payment of page charges. This article must therefore be hereby marked
advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate
We thank Dr. J. DiGiovanni and Dr. P. Dennis for assistance with this fact.
rapamycin dose selection, Dr. K. Hance and Dr. J. Schlom for generously
providing the Panc02 cells, and Dr. D. Kusewitt for assistance with quanti- Received January 14, 2011; revised March 15, 2011; accepted May 6,
fication of immunohistochemical staining patterns. 2011; published OnlineFirst May 18, 2011.

References
1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 19. Petroulakis E, Mamane Y, Le Bacquer O, Shahbazian D, Sonenberg N.
2009. CA Cancer J Clin 2009;59:22549. mTOR signaling: implications for cancer and anticancer therapy. Br J
2. Lillemoe KD, Yeo CJ, Cameron JL. Pancreatic cancer: state-of-the-art Cancer 2006;94:1959.
care. CA Cancer J Clin 2000;50:24168. 20. Shah SA, Potter MW, Ricciardi R, Perugini RA, Callery MP. FRAP-
3. Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, p70s6K signaling is required for pancreatic cancer cell proliferation. J
obesity, and mortality from cancer in a prospectively studied cohort of Surg Res 2001;97:12330.
U.S. adults. N Engl J Med 2003;348:162538. 21. Grewe M, Gansauge F, Schmid RM, Adler G, Seufferlein T. Regulation
4. Gapstur SM, Gann PH, Lowe W, Liu K, Colangelo L, Dyer A. Abnormal of cell growth and cyclin D1 expression by the constitutively active
glucose metabolism and pancreatic cancer mortality. JAMA 2000; FRAP-p70s6K pathway in human pancreatic cancer cells. Cancer Res
283:25528. 1999;59:35817.
5. Silverman DT, Swanson CA, Gridley G, Wacholder S, Greenberg RS, 22. Stephan S, Datta K, Wang E, Li J, Brekken RA, Parangi S, et al. Effect
Brown LM, et al. Dietary and nutritional factors and pancreatic cancer: of rapamycin alone and in combination with antiangiogenesis therapy
a case-control study based on direct interviews. J Natl Cancer Inst in an orthotopic model of human pancreatic cancer. Clin Cancer Res
1998;90:17109. 2004;10:69937000.
6. Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer 23. Wang Y, Zhao Q, Ma S, Yang F, Gong Y, Ke C. Sirolimus inhibits
therapy. Nat Rev Cancer 2004;4:33548. human pancreatic carcinoma cell proliferation by a mechanism linked
7. Hursting SD, Smith SM, Lashinger LM, Harvey AE, Perkins SN. to the targeting of mTOR/HIF-1 alpha/VEGF signaling. IUBMB Life
Calories and carcinogenesis: lessons learned from 30 years of calorie 2007;59:71721.
restriction research. Carcinogenesis 2010;31:839. 24. Namba R, Young LJ, Abbey CK, Kim L, Damonte P, Borowsky AD,
8. Dunn SE, Kari FW, French J, Leininger JR, Travlos G, Wilson R, et al. et al. Rapamycin inhibits growth of premalignant and malignant
Dietary restriction reduces insulin-like growth factor I levels, which mammary lesions in a mouse model of ductal carcinoma in situ. Clin
modulates apoptosis, cell proliferation, and tumor progression in p53- Cancer Res 2006;12:261321.
deficient mice. Cancer Res 1997;57:466772. 25. Majumder PK, Febbo PG, Bikoff R, Berger R, Xue Q, McMahon LM,
9. Moore T, Carbajal S, Beltran L, Perkins SN, Yakar S, Leroith D, et al. et al. mTOR inhibition reverses Akt-dependent prostate intraepithelial
Reduced susceptibility to two-stage skin carcinogenesis in mice with neoplasia through regulation of apoptotic and HIF-1-dependent path-
low circulating insulin-like growth factor I levels. Cancer Res 2008;68: ways. Nat Med 2004;10:594601.
36808. 26. Liu M, Howes A, Lesperance J, Stallcup WB, Hauser CA, Kadoya K,
10. Lashinger LM, Malone LM, McArthur MJ, Goldberg JA, Daniels et al. Antitumor activity of rapamycin in a transgenic mouse model of
EA, Pavone A, et al. Genetic reduction of insulin-like growth ErbB2-dependent human breast cancer. Cancer Res 2005;65:
factor-1 mimics the anticancer effects of calorie restriction on 532536.
cyclooxygenase-2driven pancreatic neoplasia. Cancer Prev Res 27. Wislez M, Spencer ML, Izzo JG, Juroske DM, Balhara K, Cody DD,
2011;4:103040. et al. Inhibition of mammalian target of rapamycin reverses alveolar
11. Karna E, Surazynski A, Orlowski K, Laszkiewicz J, Puchalski Z, Nawrat epithelial neoplasia induced by oncogenic K-ras. Cancer Res 2005;
P, et al. Serum and tissue level of insulin-like growth factor-I (IGF-I) 65:322635.
and IGF-I binding proteins as an index of pancreatitis and pancreatic 28. Granville CA, Warfel N, Tsurutani J, Hollander MC, Robertson M, Fox
cancer. Int J Exp Pathol 2002;83:23945. SD, et al. Identification of a highly effective rapamycin schedule that
12. Lin Y, Tamakoshi A, Kikuchi S, Yagyu K, Obata Y, Ishibashi T, et al. markedly reduces the size, multiplicity, and phenotypic progression of
Serum insulin-like growth factor-I, insulin-like growth factor binding tobacco carcinogen-induced murine lung tumors. Clin Cancer Res
protein-3, and the risk of pancreatic cancer death. Int J Cancer 2007;13:22819.
2004;110:5848. 29. Corbett TH, Roberts BJ, Leopold WR, Peckham JC, Wilkoff LJ,
13. Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling Griswold DP, Jr, et al. Induction and chemotherapeutic response of
pathways: insights into insulin action. Nat Rev 2006;7:8596. two transplantable ductal adenocarcinomas of the pancreas in
14. Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. C57BL/6 mice. Cancer Res 1984;44:71726.
Nat Rev Cancer 2008;8:91528. 30. De Angel RE, Berrigan D, Nunez NP, Hursting SD, Perkins SN. Dietary
15. Wu Q, Kiguchi K, Kawamoto T, Ajiki T, Traag J, Carbajal S, et al. calcium source influences body composition, glucose metabolism
Therapeutic effect of rapamycin on gallbladder cancer in a transgenic and hormone levels in a mouse model of postmenopausal obesity. In
mouse model. Cancer Res 2007;67:3794800. Vivo 2009;23:52735.
16. Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S. Regulation 31. Giovannucci E, Michaud D. The role of obesity and related metabolic
of lifespan in drosophila by modulation of genes in the TOR signaling disturbances in cancers of the colon, prostate, and pancreas. Gastro-
pathway. Curr Biol 2004;14:88590. enterology 2007;132:220825.
17. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, 32. Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidia-
et al. Rapamycin fed late in life extends lifespan in genetically hetero- betic therapies affect risk of pancreatic cancer. Gastroenterology
geneous mice. Nature 2009;460:3925. 2009;137:4828.
18. Vellai T, Takacs-Vellai K, Zhang Y, Kovacs AL, Orosz L, Muller F. 33. Schneider MB, Matsuzaki H, Haorah J, Ulrich A, Standop J, Ding XZ,
Genetics: influence of TOR kinase on lifespan in C. elegans. Nature et al. Prevention of pancreatic cancer induction in hamsters by
2003;426:620. metformin. Gastroenterology 2001;120:126370.

1050 Cancer Prev Res; 4(7) July 2011 Cancer Prevention Research

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Rapamycin and Calorie Restriction Inhibit Pancreatic Cancer

34. Namba R, Maglione JE, Davis RR, Baron CA, Liu S, Carmack CE, et al. 43. Tremblay F, Brule S, Hee Um S, Li Y, Masuda K, Roden M, et al.
Heterogeneity of mammary lesions represent molecular differences. Identification of IRS-1 Ser-1101 as a target of S6K1 in nutrient- and
BMC Cancer 2006;6:275. obesity-induced insulin resistance. Proc Natl Acad Sci U S A
35. Neshat MS, Mellinghoff IK, Tran C, Stiles B, Thomas G, Petersen R, 2007;104:1405661.
et al. Enhanced sensitivity of PTEN-deficient tumors to inhibition of 44. Ozes ON, Akca H, Mayo LD, Gustin JA, Maehama T, Dixon JE, et al. A
FRAP/mTOR. Proc Natl Acad Sci U S A 2001;98:103149. phosphatidylinositol 3-kinase/Akt/mTOR pathway mediates and
36. Podsypanina K, Lee RT, Politis C, Hennessy I, Crane A, Puc J, et al. An PTEN antagonizes tumor necrosis factor inhibition of insulin signaling
inhibitor of mTOR reduces neoplasia and normalizes p70/S6 kinase through insulin receptor substrate-1. Proc Natl Acad Sci U S A
activity in Pten/- mice. Proc Natl Acad Sci U S A 2001;98:103205. 2001;98:46405.
37. Yu K, Toral-Barza L, Discafani C, Zhang WG, Skotnicki J, Frost P, et al. 45. Yokogami K, Wakisaka S, Avruch J, Reeves SA. Serine phosphoryla-
mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR tion and maximal activation of STAT3 during CNTF signaling is
inhibitor, in preclinical models of breast cancer. Endocr Relat Cancer mediated by the rapamycin target mTOR. Curr Biol 2000;10:4750.
2001;8:24958. 46. Zhang Y, Daquinag A, Traktuev DO, Amaya-Manzanares F, Simmons
38. Moore T, Beltran L, Carbajal S, Strom S, Traag J, Hursting SD, et al. PJ, March KL, et al. White adipose tissue cells are recruited by
Dietary energy balance modulates signaling through the Akt/mam- experimental tumors and promote cancer progression in mouse
malian target of rapamycin pathways in multiple epithelial tissues. models. Cancer Res 2009;69:525966.
Cancer Prev Res 2008;1:6576. 47. Galie M, Konstantinidou G, Peroni D, Scambi I, Marchini C, Lisi V, et al.
39. Fraenkel M, Ketzinel-Gilad M, Ariav Y, Pappo O, Karaca M, Castel J, Mesenchymal stem cells share molecular signature with mesenchy-
et al. mTOR inhibition by rapamycin prevents beta-cell adaptation to mal tumor cells and favor early tumor growth in syngeneic mice.
hyperglycemia and exacerbates the metabolic state in type 2 dia- Oncogene 2008;27:254251.
betes. Diabetes 2008;57:94557. 48. Muehlberg FL, Song YH, Krohn A, Pinilla SP, Droll LH, Leng X, et al.
40. Houde VP, Brule S, Festuccia WT, Blanchard PG, Bellmann K, Tissue-resident stem cells promote breast cancer growth and metas-
Deshaies Y, et al. Chronic rapamycin treatment causes glucose tasis. Carcinogenesis 2009;30:58997.
intolerance and hyperlipidemia by upregulating hepatic gluconeogen- 49. Trayhurn P, Wood IS. Signalling role of adipose tissue: adipokines
esis and impairing lipid deposition in adipose tissue. Diabetes and inflammation in obesity. Biochem Soc Trans 2005;33(Pt 5):
2010;59:133848. 107881.
41. Zhang J, Gao Z, Yin J, Quon MJ, Ye J. S6K directly phosphorylates 50. Subbaramaiah K, Howe LR, Bhardwaj P, Du B, Gravaghi C, Yantiss
IRS-1 on Ser-270 to promote insulin resistance in response to TNF- RK, et al. Obesity is associated with inflammation and elevated
(alpha) signaling through IKK2. J Biol Chem 2008;283:3537582. aromatase expression in the mouse mammary gland. Cancer Prev
42. Harrington LS, Findlay GM, Gray A, Tolkacheva T, Wigfield S, Rebholz Res 2011;4:32946.
H, et al. The TSC12 tumor suppressor controls insulin-PI3K signaling 51. Hursting SD. Inflammatory talk: linking obesity, NF-{kappa}B, and
via regulation of IRS proteins. J Cell Biol 2004;166:21323. aromatase. Cancer Prev Res 2011;4:2857.

www.aacrjournals.org Cancer Prev Res; 4(7) July 2011 1051

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.
Published OnlineFirst May 18, 2011; DOI: 10.1158/1940-6207.CAPR-11-0023

Rapamycin Partially Mimics the Anticancer Effects of Calorie


Restriction in a Murine Model of Pancreatic Cancer
Laura M. Lashinger, Lauren M. Malone, Graham W. Brown, et al.

Cancer Prev Res 2011;4:1041-1051. Published OnlineFirst May 18, 2011.

Updated version Access the most recent version of this article at:
doi:10.1158/1940-6207.CAPR-11-0023

Cited articles This article cites 51 articles, 28 of which you can access for free at:
http://cancerpreventionresearch.aacrjournals.org/content/4/7/1041.full.html#ref-list-1

Citing articles This article has been cited by 13 HighWire-hosted articles. Access the articles at:
http://cancerpreventionresearch.aacrjournals.org/content/4/7/1041.full.html#related-urls

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, contact the AACR Publications
Department at permissions@aacr.org.

Downloaded from cancerpreventionresearch.aacrjournals.org on June 3, 2016. 2011 American Association


for Cancer Research.

You might also like