ArticlePDF Available

Yao M, Kargman S, Lam EC, Kelly CR, Zheng Y, Luk P, Kwong E, Evans JF, Wolfe MMInhibition of cyclooxygenase-2 by rofecoxib attenuates the growth and metastatic potential of colorectal carcinoma in mice. Cancer Res 63: 586-592

Authors:

Abstract and Figures

A large number of epidemiological studies have shown that regular use of aspirinor other nonsteroidal anti-inflammatory drugs (NSAIDs) results in a 40-50% reduced risk of colorectal cancer (CRC). Furthermore, NSAIDs cause the regression of preexisting adenomas in patients with familial adenomatous polyposis and significantly inhibit tumor growth in animal models of CRC. To establish a CRC liver metastasis model, we implanted mouse colon tumor MC-26 cells into the splenic subcapsule of BALB/c mice, after which mice were given either standard chow or chow containing the cyclooxygenase (COX)-2-specific inhibitor rofecoxib, alone or in combination with the standard antineoplastic agents, 5-fluoruracil or irinotecan. After 14 days, mice that were given rofecoxib or irinotecan, but not 5-fluoruracil, had significantly smaller primary tumors and fewer metastases. Rofecoxib, at clinical anti-inflammatory plasma concentrations, enhanced the effects of both antineoplastic agents when used in combination. Biochemical analyses of the primary splenic tumor in rofecoxib-treated mice showed no alteration in COX-1 expression, but significant decreases in the expression of the tumor-promoting proteins COX-2, cyclin D1, cytosolic beta-catenin, matrix metalloproteinases-2 and -9, and vascular endothelial cell- derived growth factor. Rofecoxib also decreased growth-enhancing prostaglandin E(2) and tumor-suppressive interleukin-10, whereas antineoplastic interleukin-12 was increased. Two separate survival studies were performed. When mice were fed chow containing 0.01% rofecoxib beginning on day 0 after tumor cell implantation, which achieved clinical anti-inflammatory plasma concentrations, survival time was significantly longer compared with mice given control chow. After 30 days, mortality in the control group was 90%, whereas only one mouse (5%) treated with rofecoxib had died after 30 days. In the second survival study, all of the mice were initially fed with regular chow after tumor cell implantation. On day 7, mice were randomly divided into three dietary groups: control chow, low-dose (0.01%) rofecoxib chow, and high-dose (0.025%) rofecoxib chow. After 28 days, mortality was 100%, 20%, and 10% in control, low-, and high-dose rofecoxib fed groups, respectively. These studies demonstrate that rofecoxib decreases the growth and metastatic potential of CRC in mice through multiple mechanisms. These studies in mice also provide important information that supports the benefit of COX-2 inhibition, not only in the prevention of CRC, but also potentially in the treatment of this common malignancy. Clinical trials will be necessary to assess the utility of COX-2 inhibitors as adjuvant therapy for early-stage disease and as potential agents, either alone or in combination, with more established drugs, for the treatment of refractory CRC.
Content may be subject to copyright.
2003;63:586-592. Cancer Res
Min Yao, Stacia Kargman, Eugene C. Lam, et al.
Mice
Growth and Metastatic Potential of Colorectal Carcinoma in
Inhibition of Cyclooxygenase-2 by Rofecoxib Attenuates the
Updated version
http://cancerres.aacrjournals.org/content/63/3/586
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/63/3/586.full.html#ref-list-1
This article cites by 34 articles, 14 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/63/3/586.full.html#related-urls
This article has been cited by 26 HighWire-hosted articles. Access the articles at:
E-mail alerts
related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
[CANCER RESEARCH 63, 586–592, February 1, 2003]
Inhibition of Cyclooxygenase-2 by Rofecoxib Attenuates the Growth and Metastatic
Potential of Colorectal Carcinoma in Mice
Min Yao,
1
Stacia Kargman,
1
Eugene C. Lam, Colleen R. Kelly, Yaxin Zheng, Pauline Luk, Elizabeth Kwong,
Jilly F. Evans, and M. Michael Wolfe
2
Section of Gastroenterology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts 02118 [M. Y., E. C. L., C. R. K., Y. Z., M. M. W.]; Merck
Frosst Centre for Therapeutic Research, Pointe Claire, Dorval, Quebec, H9H 3L1 Canada [S. K., P. L., E. K.]; and Merck Research Laboratories, West Point, Pennsylvania
19486 [J. F. E.]
ABSTRACT
A large number of epidemiological studies have shown that regular use of
aspirin or other nonsteroidal anti-inflammatory drugs (NSAIDs) results in a
40–50% reduced risk of colorectal cancer (CRC). Furthermore, NSAIDs
cause the regression of preexisting adenomas in patients with familial ade-
nomatous polyposis and significantly inhibit tumor growth in animal models
of CRC. To establish a CRC liver metastasis model, we implanted mouse
colon tumor MC-26 cells into the splenic subcapsule of BALB/c mice, after
which micewere giveneither standardchow orchow containingthe cyclooxy-
genase (COX)-2-specific inhibitor rofecoxib, alone or in combination with the
standard antineoplastic agents, 5-fluoruracil or irinotecan. After 14 days,
mice that were given rofecoxib or irinotecan, but not 5-fluoruracil, had
significantly smaller primary tumors and fewer metastases. Rofecoxib, at
clinical anti-inflammatory plasma concentrations, enhanced the effects of
both antineoplastic agents when used in combination. Biochemicalanalyses of
the primary splenic tumor in rofecoxib-treated mice showed no alteration in
COX-1 expression, but significant decreases in the expression of the tumor-
promoting proteins COX-2, cyclin D1, cytosolic
-catenin, matrix metallo-
proteinases-2 and -9, and vascular endothelial cell- derived growth factor.
Rofecoxib also decreased growth-enhancing prostaglandin E
2
and tumor-
suppressive interleukin-10, whereas antineoplastic interleukin-12 was in-
creased. Two separate survival studies were performed. When mice were fed
chow containing 0.01% rofecoxib beginning on day 0 after tumor cell im-
plantation, which achieved clinical anti-inflammatory plasma concentrations,
survival time was significantly longer compared with mice given control
chow. After 30 days, mortality in the control group was 90%, whereas only
one mouse (5%) treated with rofecoxib had died after 30 days. In the second
survival study, all of the mice were initially fed with regular chow after tumor
cell implantation. On day 7, mice were randomly divided into three dietary
groups: control chow, low-dose (0.01%) rofecoxib chow, and high-dose
(0.025%) rofecoxib chow. After 28 days, mortality was 100%, 20%, and 10%
in control, low-, and high-dose rofecoxib fed groups, respectively. These
studies demonstrate that rofecoxib decreases the growth and metastatic po-
tential of CRC in mice through multiple mechanisms. These studies in mice
also provide important information that supports the benefit of COX-2
inhibition, not only in the prevention of CRC, but also potentially in the
treatment of this common malignancy. Clinical trials will be necessary to
assess the utility of COX-2 inhibitors as adjuvant therapy for early-stage
disease and as potential agents, either alone or in combination, with more
established drugs, for the treatment of refractory CRC.
INTRODUCTION
CRC
3
is second only to lung cancer as a cause of death from
malignant disease in the United States (1), with one-half of patients
incurable at their presentation (2). Nearly 130,000 new cases of CRC
were diagnosed in the United States in 1999, resulting in 56,600
associated deaths (3). Approximately 6% of Americans will develop
CRC during their lifetime, and 2.6% will die from this disease (4).
Chemotherapeutic modalities to treat refractory CRC, although asso-
ciated with significant toxicity, have provided only minimal benefit in
improving survival (5, 6). A large number of epidemiological studies
have shown that regular use of aspirin or other NSAIDs results in a
40–50% reduced risk of CRC (7, 8). Furthermore, NSAIDs cause the
regression of preexisting adenomas in patients with familial adenom-
atous polyposis (9) and significantly inhibit tumor growth in animal
models of CRC (8, 10).
The mechanism of inhibition of COX activity by aspirin and
NSAIDs was first described by Vane in 1971 (11). This observation
led to the hypothesis that both the toxicity and efficacy of NSAIDs are
mediated through the inhibition of COX-mediated prostaglandin syn-
thesis (12). In the early 1990s, several groups reported the discovery
of two COX isoforms, a constitutively expressed isoform, COX-1, and
a strongly inducible form, COX-2, which is involved in growth
proliferation and the inflammatory response (13, 14). Despite 60%
amino acid identity, the two COX isoenzymes are encoded by distinct
genes and differ significantly with regard to tissue-specific distribu-
tion (12). COX-1 appears to function as a physiological regulatory
enzyme in most tissues, including the gastric mucosa, the kidney, and
platelets, whereas COX-2 is nearly undetectable in most (but not all)
tissues under normal physiological conditions (12). COX-2 mRNA
expression and protein was found to be enhanced in human colorectal
adenomas and adenocarcinomas, leading to the hypothesis that selec-
tive COX-2 inhibition might thereby reduce the development of CRC
(15–17). Specific COX-2 inhibition, either by targeted knockout of
the COX-2 gene or by pharmacological intervention, has been shown
to effectively decrease the growth of murine intestinal adenomas (17–
19). In a rat model of chemical-induced CRC, the COX-2 selective
inhibitor celecoxib suppressed the formation of azoxymethane-
induced aberrant crypt foci (20). Celecoxib also inhibited the inci-
dence and multiplicity of colon tumors by 93 and 97%, respectively,
and diminished the overall rat colon tumor burden by 87% (21). A
recent human study demonstrated that selective COX-2 inhibition
with celecoxib in 77 individuals with familial adenomatous polyposis
decreased the mean number of colonic polyps and the polyp burden by
28.0 and 30.7%, respectively (9).
No published studies have reported the benefits of specific COX-2
inhibitors in treating metastatic cancer. However, one clinical study
involving the use of the potent nonselective COX inhibitor indo-
methacin demonstrated that the treatment of end-stage metastatic
patients (mainly CRC and liver, pancreas, and gastric primary can-
cers) resulted in increased survival (250–510 days), less pain, and
diminished use of other analgesics (22, 23).
In the present study, we sought to determine whether rofecoxib, a
specific inhibitor of COX-2, could reduce primary tumor growth and
liver metastatic potential of mouse colon tumors in vivo. We used
BALB/c mice, in which MC-26 cells, a transplantable mouse CRC
Received 4/30/02; accepted 11/27/02.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance with
18 U.S.C. Section 1734 solely to indicate this fact.
1
M. Y. and S. K. contributed equally to this work.
2
To whom requests for reprints should be addressed, at Boston Medical Center,
Section of Gastroenterology, 650 Albany Street, Boston, MA 02118. Phone: (617)
638-8330; Fax: (617) 638-7785; E-mail: michael.wolfe@bmc.org.
3
The abbreviations used are: CRC, colorectal cancer; NSAID, nonsteroidal anti-
inflammatory drug; COX, cyclooxygenase; 5-FU, 5-fluoruracil; LV, leucovorin; MMP,
matrix metalloproteinase; VEGF, vascular endothelial growth factor; IL, interleukin;
HPLC, high-performance liquid chromatography; PGE
2
, prostaglandin E
2
; CPT-11, iri
-
notecan.
586
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
cell line expressing COX-2 protein, were implanted surgically into the
splenic capsule. Mice were then administered either standard chow or
chow containing the COX-2-specific inhibitor rofecoxib alone, or in
combination with standard antineoplastic agents. Our initial hypoth-
esis was that rofecoxib alone would have minimal effect but would
have additive or synergistic effects with the standard antineoplastic
agents. Our studies demonstrate that COX-2 inhibition by rofecoxib
alone decreases the growth and liver metastatic potential of CRC in
mice, in addition to augmenting the antineoplastic properties of the
standard chemotherapeutic agents.
MATERIALS AND METHODS
Preparation of Rofecoxib and Other Antineoplastic Agents
We purchased 5-FU and LV from Sigma Chemical Co. (St. Louis, MO).
The working solution was prepared by adding 5-FU and LV to 0.9% NaCl
adjusted to pH 8.6 with 0.01 M NaOH, after which the solution was sonicated
for 5 min and then filtered through a 0.22
m filter. We obtained CPT-11 from
Pharmacia (Kalamazoo, MI) and prepared the working solution by diluting 20
mg/ml of the agent into a 2 mg/ml concentration with 5% glucose. 5-FU, LV,
and CPT-11 were injected i.p. at doses of 30 mg/kg, 50 mg/kg, and 30 mg/kg,
respectively. We prepared 0.01% rofecoxib chow, equivalent to a dose of 20
mg/kg/day, for use in these experiments.
Cell Line
We obtained MC-26 cells, a transplantable mouse colon cancer cell line
(24), from Dr. K. K. Tanabe Massachusetts General Hospital, (Boston, MA)
and maintained in DMEM (Life Technologies, Inc., Gaithersburg, MD), sup-
plemented with 10% FBS and antibiotics (100 units/ml penicillin and 100
g/ml streptomycin), at 37°C in a humidified atmosphere of 95% air/5% CO
2
.
Animal Preparation, Treatment, and Surveillance
We purchased 6-to-10-week-old male BALB/c mice from Taconic (Ger-
mantown, NY). We harvested MC-26 cells from subconfluent cultures using
trypsin-EDTA (Life Technologies, Inc.), followed by centrifugation at 300 g
for 15 min at room temperature. We then resuspended cells in serum-free
DMEM or HBSS (Life Technologies, Inc.), and the cell number was adjusted
to a final concentration of 200,000 cells/ml. A 1-cm incision was made in the
mice under i.p. anesthesia with pentobarbital (Abbott Laboratories, North
Chicago, IL) at 65 mg/kg body weight. Using a 27-gauge needle and a 1-ml
syringe, we injected 100
l of tumor cell suspension (2 10
4
) into the
subsplenic capsule of mice. All of the animal studies were conducted using a
protocol approved by the Institutional Animal Care and Use Committee at
Boston University Medical Center.
On day 0, after complete recovery from surgery, we randomly divided mice
into eight groups: Group 1, control (n 20), received control chow plus 0.9%
NaCl or 5% glucose i.p. injection on days 4, 5, 7, 8, 11, and 12; Group 2,
rofecoxib alone (n 20), received rofecoxib 0.01% chow, and 0.9% NaCl or
5% glucose i.p. injection on days 4, 5, 7, 8, 11, and 12; Group 3, 5-FU/LV
alone (n 11), received control chow plus 5-FU 30 mg/kg and LV 50 mg/kg
on days 5, 7, and 12. Injection of 5-FU was performed 1 h after LV injection.
Five % glucose i.p. injection was given on days 4, 8, and 11; Group 4, CPT-11
alone (n 11), received control chow plus CPT-11 30 mg/kg on days 4, 8, and
11, and 0.9% NaCl i.p. injection on days 5, 7, and 12; Group 5, rofecoxib/5-
FU/LV (n 11), received rofecoxib chow plus 5-FU 30 mg/kg and LV 50
mg/kg on days 5, 7 and 12, and 5% glucose i.p. injection on days 4, 8, and 11.
Group 6, rofecoxib/CPT-11 (n 11), received rofecoxib chow plus CPT-11 30
mg/kg on days 4, 8, and 11, and 0.9% NaCl i.p. injection on days 5, 7, and 12;
Group 7, 5-FU/LV/CPT-11 (n 11), received control chow plus 5-FU 30
mg/kg and LV 50 mg/kg on days 5, 7, and 12, and CPT-11 30 mg/kg on days
4, 8, and 11; Group 8, rofecoxib/5-FU/LV/CPT-11 (n 11); received rofe-
coxib chow plus 5-FU 30 mg/kg and LV 50 mg/kg on days 5, 7, and 12, and
CPT-11 30 mg/kg on days 4, 8, and 11.
On day 14, blood samples were obtained, after which all of the mice were
sacrificed. During the dissection, primary (splenic) tumor size was determined
by weighing and then measuring the longest and shortest diameters of the
tumor. Tumor volume (mm
3
) was calculated using the standard formula: tumor
volume (shortest diameter)
2
(longest diameter) 0.5. We fixed a portion
of the tumor tissue in 10% formalin for subsequent histological examination
and then flash-froze the remaining tissue in liquid nitrogen and stored it at
70°C. Metastatic nodules in the liver were detected macroscopically and
confirmed microscopically, and the percentage of mice with hepatic metastasis
was recorded.
In a separate experiment, to identify the effect of rofecoxib on liver weight,
five mice without tumor cell injection were fed 0.01% rofecoxib chow, and
another five mice without tumor cell injection were fed control chow for 14
days. All of the mice in both groups were sacrificed on day 14, and liver
weights were measured.
Survival Studies
Two separate experiments were performed to assess survival.
Protocol 1. On day 0, after complete recovery from surgery, mice were
randomly divided into two groups (n 20 in each group): a control group, in
which mice were fed with regular chow; and a rofecoxib group, in which mice
were fed with rofecoxib 0.01% chow (20 mg/kg mouse weight, which
achieved a plasma concentration of 0.26
g/ml). This survival study was
terminated on day 30.
Protocol 2. On day 0, after complete recovery from surgery, all of the mice
were fed regular chow. On day 7, mice were randomly divided into three
groups (n 20 in each group): a control group, which received regular chow;
a low-dose rofecoxib group, which received chow containing rofecoxib 0.01%
(which achieved a plasma concentration of 0.26
g/ml); and a high-dose
rofecoxib group, which received chow containing rofecoxib 0.025% (50
mg/kg mouse weight, which achieved a plasma concentration of 0.55
g/ml).
The survival study was terminated when mortality in the control group reached
100%.
Preparation of Microsomal Membranes from Splenic Tumor Tissue
Splenic tumor tissue was excised, immediately frozen in liquid N
2
, and
stored at 70°C. Frozen tissues were thawed in ice-cold homogenization
buffer [50 m
M potassium phosphate (pH 7.1), containing 0.1 M NaCl, 2 mM
EDTA, 0.4 mM phenylmethylsulfonyl fluoride, 60 g/ml soybean trypsin inhib-
itor, 2 g/ml leupeptin, 2 g/ml aprotinin, and 2 g/ml pepstatin, all from Sigma
Chemical Co.]. Tissues were disrupted twice, for 10 s each, on ice using a
tissue tearer (IKA Labortechnik, Staufen, Germany). Samples were homoge-
nized by sonication at 4°C using a Cole Parmer 4710 series ultrasonic homog-
enizer (Cole Parmer Instrument Co., Chicago, IL). Debris was removed by
centrifugation at 1,000 g for 15 min at 4°C, and the resultant supernatants
were subjected to centrifugation at 100,000 g for 30 min at 4°C. Membrane
fractions were resuspended in homogenization buffer and then sonicated to
obtain a homogeneous membrane suspension. Protein concentrations were
determined for each sample using a protein assay kit (Bio-Rad, Mississauga,
Ontario, Canada), and equal gel loading was determined by Coomassie Blue
staining (Sigma).
SDS-PAGE and Immunoblot Analysis
Microsomal and cytosolic fractions were mixed with 0.5 volume of SDS
sample buffer [20 m
M Tris-HCl (pH 6.8), containing 0.4% (w/v) SDS, 4%
glycerol, 0.24 M
-mercaptoethanol, and 0.5% bromphenol blue], boiled for 5
min and analyzed by SDS-PAGE on 9 10 cm precast 420% Tris-glycine
acrylamide gels (NOVEX, San Diego, CA), according to the method of
Laemmli (25). Proteins were electrophoretically transferred to nitrocellulose
membranes, as described previously (26). Primary antisera to COX-1 (MF241;
Merck-Frosst, Montreal, Canada), COX-2 (MF243; Merck-Frosst),
-catenin
(Sigma), cyclin D1 (BD Transduction), MMP-2 (Oncogene), MMP-9 (Onco-
gene), and VEGF (Santa Cruz Biotechnology, Santa Cruz, CA) were used at
final dilutions of 1:3,000; 1:7,500; 1:5,000; 1:1,000; 1:1,000; 1:1,000, and
1:200, respectively, according to the manufacturersinstructions. The second-
ary horseradish peroxidase-linked goat antirabbit or goat antimouse IgG anti-
bodies (Santa Cruz) were used at dilutions of 1:2,000 to 1:6,000. Immunode-
tection was performed using enhanced chemiluminescence according to the
manufacturers instructions (Amersham, Elk Grove Village, IL). Protein bands
were visualized using a Fuji LAS-1000 plus Luminescent Image Analyzer
587
COX-2 INHIBITION BY ROFECOXIB IN THE TREATMENT OF CRC
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
(Fuji Photo Film Company, Tokyo, Japan) and quantitated using Fuji Film
Image Gauge version 3.122. The volumes of absorbance corresponding to the
purified COX isoform (Cayman), a purified M
r
46,000 tagged fusion protein
corresponding to NH
2
-terminal amino acids 1147 of human VEGF or
-cate
-
nin (a thioredoxin fusion protein; a kind gift of Astrid Kral, MRL, West Point,
PA) proteins were used to calculate the quantity of COX or VEGF protein in
splenic tumor samples. Immunoblot analyses of the above proteins were
performed using the same nitrocellulose membrane by stripping the membrane
with a buffered solution containing 62.5 m
M Tris-HCl, 2% SDS, and 100 nM
2-mercaptoenthenol.
ELISA and Measurement of Plasma Rofecoxib Levels
The concentrations of IL-10, IL-12, and PGE
2
in splenic tumor cytosolic
fractions were determined by quantitative ELISA, according to the manufac-
turers directions. IL-10 and IL-12 immunoassay kits were purchased from
Biosource International (Camillaro, CA), and PGE
2
kits were purchased from
Assay Designs, Inc. (Ann Arbor, MI).
Plasma rofecoxib levels were measured by HPLC. A set of four rofecoxib
standards, ranging from 0.1 to 5.0
g/ml (R
2
0.9999), were prepared in
acetonitrile. We added 100
l of blank mouse plasma to each 100-
l aliquot
of the standards and then mixed the standards on an automatic vortex mixer for
10 min. The sample was centrifuged at 14,000 rpm for 15 min, and the
supernatant was transferred to a HPLC vial for injection. Sample preparation
was identical to the standard preparation procedure with the exception of a
100-
l aliquot of acetonitrile added to 100
l of plasma sample. An HP1090
system equipped with an UV detector was fitted with an Eclipse XDB-C18
Rapid 74Resolution (4.6 75 mm, 3.5
m) analytical column. We used a flow
rate of 1.0 ml/min with a solvent ratio of 65% aqueous (0.1% trifluoracetic acid
in water) to 35% organic (0.1% trifluoracetic acid in acetonitrile), an oven
temperature of 40°C, and an injection volume of 25
l. Rofecoxib was
detected at a wavelength of 275 nm.
Statistical Analysis
Using SAS 8.0, we performed one-way ANOVA to compare tumor volume
and liver weight and densitometric values of Western blot bands among the
different animal groups, followed by Tukeys procedure for paired compari-
sons. We used the Fisher Exact
2
test for categorical data such as the
incidence rate of liver metastasis. We performed survival analysis using the
Kaplan-Meier method and log-rank tests. We assigned statistical significance
if P 0.05.
RESULTS
Plasma Rofecoxib Concentrations. Rofecoxib could be detected
in the plasma of all of the groups treated with rofecoxib alone or in
combination with standard antineoplastic agents. Plasma drug con-
centrations in mice fed with 0.01% rofecoxib chow were similar to
trough levels achieved at steady-state with once daily 25-mg dosing in
a 70-kg human (27), and no significant differences were detected
among the various groups (Fig. 1).
Effect of Rofecoxib Treatment on the Expression of COX-1,
COX-2, Cyclin D1,
-Catenin, MMP-2, MMP-9, and VEGF. We
examined the effect of rofecoxib treatment on the expression of mouse
splenic tumor proteins involved in proliferation, cell cycle regulation,
angiogenesis, and metastasis. Splenic tumor samples from rofecoxib-
treated or control groups were assessed for expression of COX-1,
COX-2,
-catenin, cyclin D1, MMP-2, MMP-9, and VEGF proteins
by specific immunoblot analyses. Mice treated with rofecoxib had a
statistically significant reduction in splenic tumor levels of COX-2,
cyclin D1,
-catenin, MMP-2, and VEGF protein, in comparison with
mice treated with control chow (Fig. 2, B–E, G). Mice treated with
rofecoxib showed a trend toward reduction of MMP-9 protein expres-
sion; however, this reduction did not reach statistical significance
(Fig. 2F).
Effect of Rofecoxib Treatment on the Expression of PGE
2
,
IL-10, and IL-12. Splenic tumor samples from rofecoxib-treated and
control mice were assessed for PGE
2
, IL-10, and IL-12 concentrations
by quantitative ELISA, as described above. The concentration of
growth-proliferative, immunosuppressive PGE
2
was reduced in the
splenic tumors of mice treated with rofecoxib in comparison with
control animals (Fig. 3A). This reduction in PGE
2
is consistent with
the reduction in the COX-2 enzyme in the tumors of rofecoxib-treated
animals (Fig. 2B). The tumor promoting IL-10 cytokine was signifi-
cantly reduced in tumor samples of rofecoxib-treated animals as
compared with controls (Fig. 3B). In contrast, levels of the potent
proinflammatory tumor suppressive cytokine IL-12 were markedly
increased in rofecoxib-treated splenic tumors as compared with con-
trol mouse splenic tumors (Fig. 3C).
Effect of Rofecoxib Treatment on Tumor Growth and Metas-
tasis and on the Survival of Mice with Implanted Splenic CRC
Cells. Rofecoxib alone, or in combination with standard antineoplas-
tic agents, significantly inhibited in situ colon cancer growth and the
incidence of spontaneous liver metastasis (Fig. 4). Primary splenic
tumor volume was significantly decreased in all of the groups, al-
though to a lesser extent in those mice treated with 5-FU/LV (Fig.
5A). Moreover, rofecoxib potentiated the effects of both 5-FU/LV and
CPT-11 on tumor volume (Fig. 5A). Liver weight (Fig. 5B) and the
incidence of liver metastasis (Fig. 5C) were not significantly affected
in mice treated with 5-FU/LV alone. In contrast, both rofecoxib and
CPT-11 decreased liver weight and metastasis significantly, and the
effects of rofecoxib and CPT-11 were additive (Figs. 5, B and C). We
did not detect metastases in any of mice treated with both rofecoxib
and CPT-11 (Fig. 5C). Liver weight and metastasis were diminished
in mice treated with a combination of rofecoxib and 5-FU/LV com-
pared with the latter alone but were increased compared with mice
treated with rofecoxib alone.
In the initial survival study, in which mice were fed with 0.01%
rofecoxib chow on day 0, the survival time in mice treated with
rofecoxib chow was significantly longer than in mice given control
chow. After 30 days, mortality in the control group was 90%, whereas
only one mouse (5%) treated with rofecoxib had died after 30 days
(P 0.00001; Fig. 6A). In the second survival study, mice were fed
with regular chow on recovery from surgery, and on day 7, they were
randomly divided into three groups. All of the mice fed with control
chow died by day 28, and mortality rates were 100, 20, and 10%,
respectively, in the control, 0.01 and 0.025% rofecoxib groups
(P 0.0001 for both treatment groups; Fig. 6B).
Fig. 1. Plasma rofecoxib concentrations in different groups of mice receiving rofe-
coxib-containing chow. Blood was obtained from mice at day 14, and after centrifugation
and separation, plasma rofecoxib levels were measured by HPLC, as described in
Materials and Methods.Values represent the mean SE of nine samples in each group.
588
COX-2 INHIBITION BY ROFECOXIB IN THE TREATMENT OF CRC
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
DISCUSSION
A large body of experimental evidence supports the premise that
COX-2 expression may contribute to tumorigenesis and that COX-2
inhibitors might be useful in the prevention of intestinal polyposis and
colon cancer (9, 22, 23). Increased levels of COX-2 mRNA and
COX-2 protein have been found in colonic tumors and polyps, in
comparison with normal colonic epithelium (1517). Several mecha-
nisms have been supported for why COX-2 expression in neoplastic
tissues enhances tumor growth. There is evidence for amplification of
tumor cell proliferation by COX-2-produced PGE
2
and inhibition of
tumor cell apoptosis, enhancement of stromal cell angiogenesis, and
decreased immune surveillance of tumor cells. Recently, Dormond et
al. (28) demonstrated an important functional link among COX-2,
integrin
V
3, and the small GTPases Cdc42 and Rac. Using human
umbilical vein endothelial cells, they found that both NS-398 and
indomethacin, but not the specific COX-1 inhibitor SC-560, sup-
pressed the activity of integrin
V
3 (an adhesion receptor critically
involved in mediating tumor angiogenesis) and prevented activation
of Cdc42 and Rac, which are critically involved in regulating cell
migration after integrin engagement (28).
COX-2-produced prostaglandins also increase the metastatic poten-
tial of human CRC cells by enhancing proteolysis of the basement
membrane (15). Tsujii et al. (15) programmed Caco-2 cells to con-
stitutively express COX-2, which increased invasiveness compared
with control cells. This phenotypic change was associated with an
increase in the activation of MMP-2, as determined using gelatin
zymography, and an increase in MMP-1 mRNA. Both effects were
reversed by the nonspecific NSAID metabolite sulindac sulfide (15).
Tomozawa et al. (10) injected MC-26 cells into the tail vein of
BALB/c mice and demonstrated that the i.p. injection of JTE-522, a
selective COX-2 inhibitor, decreased the number of lung metastases.
The authors concluded that selective COX-2 inhibition might be
effective in decreasing the hematogenous metastasis of CRC (10).
Using a cell invasion/migration assay that employs Matrigel-coated
Fig. 2. Effect of rofecoxib treatment on mouse
splenic tumor COX-1 and -2, cyclin D1,
-catenin,
MMP-2 and -9, and VEGF expression. Splenic
tumors from rofecoxib-treated and control mice
were homogenized, separated by SDS-PAGE,
transferred to nitrocellulose, and blotted with
COX-1, COX-2, cyclin D1,
-catenin, MMP-2 and
-9, and VEGF antisera, with detection by enhanced
chemiluminescence. The amounts of COX-1,
COX-2,
-catenin, and VEGF were determined by
image quantification using a charge-coupled device
camera as indicated in Materials and Methods.
The area of absorbance for known quantities of
COX-1, COX-2,
-catenin, and VEGF proteins
were used to assess approximate amounts of these
proteins in splenic tumor samples. Values represent
the mean SE for nine animals in each treatment
group for COX-1 (A), COX-2 (B), cytoplasmic
-catenin (D), and cytoplasmic VEGF (G), and for
five animals in each group for Cyclin D1 (C) and
MMP (E and F) proteins. To the left of each bar
graph pair, a representative immunoblot analysis
from five animals in each treatment group for each
protein examined, as well as a representative im-
munoblot of
-actin. O.D., absorbance.
589
COX-2 INHIBITION BY ROFECOXIB IN THE TREATMENT OF CRC
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
chambers, we have recently reported that the COX-2 inhibitor NS-398
decreased MC-26 cell migration by 60%, indicating that COX-2
inhibition may decrease the metastatic potential of CRC, at least in
part, by inhibiting cellular invasion (29).
As stated previously, CRC usually presents at an advanced stage
and is the second leading cause of cancer deaths in the United States.
Up to 50% of patients will die of their disease within 5 years from
diagnosis (4). Whereas the 5-year survival of early-stage CRC (Dukes
A or B) is relatively high, only 40% of those individuals with
advanced stages (Dukes C and D) will be alive 5 years after the initial
diagnosis (4, 6). In a recent study by Saltz et al. (5), of 683 patients
with refractory CRC, a combination of CPT-11 and 5-FU/LV im-
proved survival by only 2.2 months over either agent alone, from a
median of 12.6 months to 14.8 months. Although the addition of
CPT-11 to 5-FU/LV did not compromise the quality of life of partic-
ipants in this study, subsequent studies (30, 31) have reported signif-
icant toxicity associated with the use of this intense parenteral drug
regimen. In contrast, as p.o. administered drugs, COX-2 inhibitors are
convenient and relatively well-tolerated by most individuals and have
been reported to incite fewer upper gastrointestinal-related events,
such as ulcer-associated hemorrhage (32, 33).
In the present study, we examined the effect of rofecoxib treatment
on the growth and metastatic potential of colorectal carcinoma in a
mouse model of colorectal metastases. The rofecoxib plasma concen-
trations in drug-treated mice were equivalent to the trough levels in a
human 25-mg anti-inflammatory dose. Remarkably, this relatively
low dose of rofecoxib not only decreased the size of the primary CRC
tumor but also highly significantly decreased the rate and extent of
liver metastasis. A striking difference in mortality was also evident in
rofecoxib-treated mice when compared with control mice. We also
sought to determine molecular mechanisms that might account for the
beneficial effects of rofecoxib observed in this in vivo study by
examining factors shown in various in vitro models to be affected by
COX-2 overexpression or inhibition, such as cell proliferation, me-
tastasis, angiogenesis, and immune modulation. Immunoblot analysis
of the primary tumors demonstrated decreases in the protein expres-
sion of COX-2,
-catenin, cyclin D1, VEGF, MMP-2, MMP-9, and
IL-10. Interestingly, a significant increase in IL-12 protein was de-
tected in the primary tumors of mice treated with rofecoxib. Recent
studies (3436) have suggested that these two important cytokines
play an important role in modulating tumor activity and that COX-2
Fig. 3. Effect of rofecoxib treatment on mouse splenic tumor PGE
2
(A), IL-10 (B), and
IL-12 (C) expression. Splenic tumors from rofecoxib-treated and control animals were
homogenized, as described in Materials and Methods. The concentrations of splenic
tumor PGE2, IL-10, and IL-12 were determined by quantitative ELISA. Values represent
mean SE for nine animals in each treatment group for PGE
2
and IL-12 and for four
animals in each treatment group for IL-10.
Fig. 4. Primary tumor and hepatic metastasis at
day 14 in a representative control mouse (left) and
in one mouse treated with rofecoxib (right).
590
COX-2 INHIBITION BY ROFECOXIB IN THE TREATMENT OF CRC
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
may alter their balance in vivo. IL-10 has been found to be tumor
promoting, whereas IL-12 appears to function as a tumor-suppressing
cytokine (36). Huang et al. (34) found that PGE
2
production by
non-small cell lung cancer was dependent on COX-2 expression and
that an increase in PGE
2
was associated with the induction of IL-10
and the suppression of IL-12. COX-2 inhibition reversed the balance
of these cytokines in the lung cancer microenvironment (34). More
recent studies using a murine Lewis lung carcinoma model have
corroborated these observations (36).
The prolonged survival of rofecoxib-treated animals in our mouse
model of CRC metastasis is most likely the result of multiple mech-
anisms, including antiproliferative and antiangiogenic effects and
enhanced immune surveillance. Reductions in primary tumor size and
metastatic potential are associated with decreases not only in COX-2
expression and PGE
2
synthesis but also in the lowering of various
angiogenic factors and a restoration of the balance between cytokines
that favors tumor suppression. These studies in mice provide impor-
tant information that support the benefit of COX-2 inhibition not only
in the prevention on CRC but also in the treatment of this common
malignancy. Clinical trials will be necessary to assess the utility of
COX-2 inhibitors as adjuvant therapy for early-stage disease and as
potential agents, either alone or in combination with more established
drugs, for the treatment of refractory CRC.
ACKNOWLEDGMENTS
We are grateful to Merck and Co., USA, for a Vioxx Medical School Grant
and to Dr. Ian Rodger for his thoughtful suggestions during the course of these
studies and in the preparation of the manuscript.
REFERENCES
1. Winawer, S. J., Fletcher, R. H., Miller, L., Godlee, F., Stolar, M. H., Mulrow, C. D.,
Woolf, S. H., Glick, S. N., Ganiats, T. G., Bond, J. H., Rosen, L., Zapka, J. G., Olsen,
S. J., Giardiello, F. M., Sisk, J. E., Van Antwerp, R., Brown-Davis, C., Marciniak,
D. A., and Mayer, R. J. Colorectal cancer screening: clinical guidelines and rationale.
Gastroenterology, 112: 594642, 1997.
2. Moertel, C. G., Fleming, T. R., Macdonald, J. S., Haller, D. G., Laurie, J. A.,
Goodman, P. J., Ungerleider, J. S., Emerson, W. A., Tormey, D. C., Glick, J. H., et
Fig. 6. Thirty-day survival in control mice and in mice treated with rofecoxib. A, two
groups of mice (n 20 in each group) were treated with regular mouse chow or
rofecoxib-containing chow on day 0 after tumor cell implantation, and survival was
assessed for a period of 30 days. B, on day 0, after complete recovery from surgery, all
of the mice were fed regular chow. On day 7, mice were randomly divided into three
groups (n 20 in each group): a control group (regular chow), 0.01% rofecoxib chow,
and 0.025% rofecoxib chow. Survival analysis was done using the Kaplan-Meier method
and log-rank tests.
Fig. 5. Primary splenic tumor volume (A), liver
weight (B), and percentage of mice with metastasis
or free of metastasis (C). Tumor volume and weight
are expressed as mean SE. B, L1, the liver weight
in mice without tumor cell injection fed rofecoxib
chow; L2, the liver weight in mice without tumor
cell injection fed control chow. , P 0.05, ⴱⴱ,
P 0.01 versus control.
591
COX-2 INHIBITION BY ROFECOXIB IN THE TREATMENT OF CRC
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
al. Levamisole and fluorouracil for adjuvant therapy of resected colon carcinoma.
N. Engl. J. Med., 322: 352358, 1990.
3. Ladabaum, U., Chopra, C. L., Huang, G., Scheiman, J. M., Chernew, M. E., and
Fendrick, A. M. Aspirin as an adjunct to screening for prevention of sporadic
colorectal cancer. A cost-effectiveness analysis. Ann. Intern. Med., 135: 769781,
2001.
4. Schroy, P. C., III. A Companion to Sleisenger and Fordtran Gastrointestinal Disease,
2000.
5. Saltz, L. B., Cox, J. V., Blanke, C., Rosen, L. S., Fehrenbacher, L., Moore, M. J.,
Maroun, J. A., Ackland, S. P., Locker, P. K., Pirotta, N., Elfring, G. L., and Miller,
L. L. Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer.
Irinotecan Study Group. N. Engl. J. Med., 343: 905914, 2000.
6. Netzer, P., Domek, M., Pai, R., Halter, F., and Tarnawski, A. Inhibition of human
colon cancer cell growth by antisense oligodeoxynucleotides targeted at basic fibro-
blast growth factor. Aliment. Pharmacol. Ther., 15: 16731679, 2001.
7. Tsujii, M., Kawano, S., Tsuji, S., Sawaoka, H., Hori, M., and DuBois, R. N.
Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell, 93:
705716, 1998.
8. Smalley, W. E., and DuBois, R. N. Colorectal cancer and nonsteroidal anti-inflam-
matory drugs. Adv. Pharmacol., 39: 120, 1997.
9. Steinbach, G., Lynch, P. M., Phillips, R. K., Wallace, M. H., Hawk, E., Gordon, G. B.,
Wakabayashi, N., Saunders, B., Shen, Y., Fujimura, T., Su, L. K., and Levin, B. The
effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis.
N. Engl. J. Med., 342: 19461952, 2000.
10. Tomozawa, S., Nagawa, H., Tsuno, N., Hatano, K., Osada, T., Kitayama, J., Sunami,
E., Nita, M. E., Ishihara, S., Yano, H., Tsuruo, T., Shibata, Y., and Muto, T. Inhibition
of haematogenous metastasis of colon cancer in mice by a selective COX-2 inhibitor,
JTE-522. Br. J. Cancer, 81: 12741279, 1999.
11. Vane, J. R. Inhibition of prostaglandin synthesis as a mechanism of action for
aspirin-like drugs. Nat. New Biol., 231: 232235, 1971.
12. Wolfe, M. M., Lichtenstein, D. R., and Singh, G. Gastrointestinal toxicity of nonste-
roidal anti-inflammatory drugs. N. Engl. J. Med., 340: 18881899, 1999.
13. Masferrer, J. L., Seibert, K., Zweifel, B., and Needleman, P. Endogenous glucocor-
ticoids regulate an inducible cyclooxygenase enzyme. Proc. Natl. Acad. Sci. USA, 89:
39173921, 1992.
14. Kujubu, D. A., Fletcher, B. S., Varnum, B. C., Lim, R. W., and Herschman, H. R.
TIS10, a phorbol ester tumor promoter-inducible mRNA from Swiss 3T3 cells,
encodes a novel prostaglandin synthase/cyclooxygenase homologue. J. Biol. Chem.,
266: 1286612872, 1991.
15. Tsujii, M., Kawano, S., and DuBois, R. N. Cyclooxygenase-2 expression in human
colon cancer cells increases metastatic potential. Proc. Natl. Acad. Sci. USA, 94:
33363340, 1997.
16. Kargman, S. L., ONeill, G. P., Vickers, P. J., Evans, J. F., Mancini, J. A., and Jothy,
S. Expression of prostaglandin G/H synthase-1 and -2 protein in human colon cancer.
Cancer Res., 55: 25562559, 1995.
17. Oshima, M., Dinchuk, J. E., Kargman, S. L., Oshima, H., Hancock, B., Kwong, E.,
Trzaskos, J. M., Evans, J. F., and Taketo, M. M. Suppression of intestinal polyposis
in Apc
716 knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell, 87:
803809, 1996.
18. Reddy, B. S., Rao, C. V., and Seibert, K. Evaluation of cyclooxygenase-2 inhibitor for
potential chemopreventive properties in colon carcinogenesis. Cancer Res., 56:
45664569, 1996.
19. Oshima, M., Murai (Hata), N., Kargman, S., Aguello, M., Luk, P., Kwong, E., Taketo,
M. M., and Evans, J. F. Chemoprevention of intestinal polyposis in the Apc
716
mouse by rofecoxib, a specific cyclooxygenase-2 inhibitor. Cancer Res., 61: 1733
1740, 2001.
20. Yoshimi, N., Kawabata, K., Hara, A., Matsunaga, K., Yamada, Y., and Mori, H.
Inhibitory effect of NS-398, a selective cyclooxygenase-2 inhibitor, on azoxymethane-
induced aberrant crypt foci in colon carcinogenesis of F344 rats. Jpn. J. Cancer Res., 88:
10441051, 1997.
21. Kawamori, T., Rao, C. V., Seibert, K., and Reddy, B. S. Chemopreventive activity of
celecoxib, a specific cyclooxygenase-2 inhibitor, against colon carcinogenesis. Can-
cer Res., 58: 409412, 1998.
22. Lundholm, K., Gelin, J., Hyltander, A., Lonnroth, C., Sandstrom, R., Svaninger, G.,
Korner, U., Gulich, M., Karrefors, I., Norli, B., et al. Anti-inflammatory treatment
may prolong survival in undernourished patients with metastatic solid tumors. Cancer
Res., 54: 56025606, 1994.
23. Taketo, M. M. Cyclooxygenase-2 inhibitors in tumorigenesis (part I). J. Natl. Cancer
Inst. (Bethesda), 90: 15291536, 1998.
24. Singh, P., Walker, J. P., Townsend, C. M., Jr., and Thompson, J. C. Role of gastrin
and gastrin receptors on the growth of a transplantable mouse colon carcinoma
(MC-26) in BALB/c mice. Cancer Res., 46: 16121616, 1986.
25. Laemmli, U. K. Cleavage of structural proteins during the assembly of the head of
bacteriophage T4. Nature (Lond.), 227: 680685, 1970.
26. Towbin, H., Staehelin, T., and Gordon, J. Electrophoretic transfer of proteins from
polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc.
Natl. Acad. Sci. USA, 76: 43504354, 1979.
27. Porras, A., Ko, A., Larson, P., Agrawal, N., Woolf, E., Ebel, D., Lassiter, K., Gertz,
B., and Schwartz, J. Single and multiple pharmacokinetics [PK] of rofecoxib [R] in
healthy subjects. Clin. Pharm. Ther., 67: 137, 2000.
28. Dormond, O., Foletti, A., Paroz, C., and Ruegg, C. NSAIDs inhibit
V
3 integrin-
mediated and Cdc42/Rac-dependent endothelial-cell spreading, migration and angio-
genesis. Nat. Med., 7: 10411047, 2001.
29. Yao, M., Lam, E. C., Kelly, C. R., and Wolfe, M. M. Cyclooxygenase-2 inhibition
reduces the proliferative and invasive properties of colorectal adenocarcinoma cells.
Gastroenterology, 122: A-240, 2002.
30. Van Cutsem, E., Douillard, J. Y., and Kohne, C. H. Toxicity of irinotecan in patients
with colorectal cancer. N. Engl. J. Med., 345: 13511352, 2001.
31. Sargent, D. J., Niedzwiecki, D., OConnell, M. J., and Schilsky, R. L. Recommen-
dation for caution with irinotecan, fluorouracil, and leucovorin for colorectal cancer.
N. Engl. J. Med., 345: 144145; discussion 146, 2001.
32. Silverstein, F. E., Faich, G., Goldstein, J. L., Simon, L. S., Pincus, T., Whelton, A.,
Makuch, R., Eisen, G., Agrawal, N. M., Stenson, W. F., Burr, A. M., Zhao, W. W.,
Kent, J. D., Lefkowith, J. B., Verburg, K. M., and Geis, G. S. Gastrointestinal toxicity
with celecoxib vs nonsteroidal anti-inflammatory drugs for osteoarthritis and rheu-
matoid arthritis: the CLASS study: a randomized controlled trial. Celecoxib Long-
term Arthritis Safety Study. JAMA, 284: 12471255, 2000.
33. Bombardier, C., Laine, L., Reicin, A., Shapiro, D., Burgos-Vargas, R., Davis, B.,
Day, R., Ferraz, M. B., Hawkey, C. J., Hochberg, M. C., Kvien, T. K., and Schnitzer,
T. J. Comparison of upper gastrointestinal toxicity of rofecoxib and naproxen in
patients with rheumatoid arthritis. VIGOR Study Group. N. Engl. J. Med., 343:
15201528, 2 p following 1528, 2000.
34. Huang, M., Stolina, M., Sharma, S., Mao, J. T., Zhu, L., Miller, P. W., Wollman, J.,
Herschman, H., and Dubinett, S. M. Non-small cell lung cancer cyclooxygenase-2-
dependent regulation of cytokine balance in lymphocytes and macrophages: up-
regulation of interleukin 10 and down-regulation of interleukin 12 production. Cancer
Res., 58: 12081216, 1998.
35. Sharma, S., Stolina, M., Lin, Y., Gardner, B., Miller, P. W., Kronenberg, M., and
Dubinett, S. M. T cell-derived IL-10 promotes lung cancer growth by suppressing
both T cell and APC function. J. Immunol., 163: 50205028, 1999.
36. Stolina, M., Sharma, S., Lin, Y., Dohadwala, M., Gardner, B., Luo, J., Zhu, L.,
Kronenberg, M., Miller, P. W., Portanova, J., Lee, J. C., and Dubinett, S. M. Specific
inhibition of cyclooxygenase 2 restores antitumor reactivity by altering the balance of
IL-10 and IL-12 synthesis. J. Immunol., 164: 361370, 2000.
592
COX-2 INHIBITION BY ROFECOXIB IN THE TREATMENT OF CRC
Research.
on May 31, 2013. © 2003 American Association for Cancercancerres.aacrjournals.org Downloaded from
... Cancer metastasis is a complex series of events, including migration of cancer cells from their primary tumor, survival in circulation, attachment to the metastatic site, and proliferation as micrometastases (4,5). During the metastasis, cyclooxygenase-2 (COX-2), a key enzyme in the conversion of arachidonic acid to prostaglandins and other eicosanoids, played an important role in promoting a more metastatic phenotype in colorectal tumor cells, while COX-2 inhibition in vivo can attenuate the metastatic potential of colorectal tumors in both humans and mice (6,7). Although COX-2 is also highly expressed and correlated with angiogenesis, the contribution of COX-2 in the regulation of prostate cancer metastasis remains unclear (8). ...
Article
Full-text available
Pyruvate kinase M2 (PKM2) is a key enzyme of glycolysis, which is highly expressed in many tumor cells, and has emerged as an important player in tumor progression and metastasis. However, the functional roles of PKM2 in tumor metastasis remain elusive. Here we showed that PKM2 promoted prostate cancer metastasis via extracellular-regulated protein kinase (ERK)-cyclooxygenase (COX-2) signaling. Based on public databases, we found that PKM2 expression was upregulated in prostate cancer and positively associated with tumor metastasis. Further analysis showed that PKM2 promoted prostate cancer cell migration/invasion and epithelial-mesenchymal transition (EMT) through upregulation of COX-2. Mechanistically, PKM2 interacted with ERK1/2 and regulated its phosphorylation, leading to phosphorylation of transcription factor c-Jun, downstream of ERK1/2, to activate COX-2 transcription by IP and ChIP assay, while inhibition of COX-2 significantly reversed the promotion effect of PKM2 on tumor metastasis in vivo. Taken together, our results suggest that a novel of PKM2-ERK1/2-c-Jun-COX-2 axis is a potential target in controlling prostate cancer metastasis.
... Up to 85% of colorectal adenocarcinoma patients were found to have high COX-2 expression. COX-2 affected the production of prostaglandins and also promoted the expression of MMP-2, MMP-9, and VEGF in colorectal cancer patients (14). Moreover, COX-2 was found to be significantly upregulated in gastric cancer, skin cancer, prostate cancer, and breast cancer tissues compared to normal tissues or adjacent tissues (15). ...
Article
Full-text available
Objective: Cyclooxygenase-2 (COX-2) is a critical enzyme associated with inflammation and tumorigenesis. Although several studies have compared the expression of COX-2 in endometrial cancer tissues and normal tissues, the results have been inconsistent thus far. This study aims to conduct a meta-analysis to elucidate the role of COX-2 in the determination of the risk, prognosis, and clinical features of endometrial cancer. Methods: We retrieved the suitable studies on the association between COX-2 expression and endometrial cancer from PubMed, EMBASE, and Web of Science databases that were published between 1999 and September 31st, 2019. The hazard ratio (HR) and 95% confidence intervals (CIs) were retrieved to assess the relationship between COX-2 expression and the prognosis of endometrial cancer. The odds ratio (OR) and 95% CIs were calculated to evaluate the correlation between COX-2 expression and the risk and clinical features of endometrial cancer. Results: To investigate the association between COX-2 expression and the susceptibility, clinical features, and prognosis of endometrial cancer, we performed a meta-analysis on data from selected studies that collectively involved 273 normal individuals and 1,376 patients with endometrial cancer. Overall, the pooled analysis indicated that COX-2 expression was significantly associated with susceptibility (Caucasians, OR = 3.94, 95% CI = 2.17–7.17, P < 0.05; Asians, OR = 20.51, 95% CI = 8.54–49.26, P < 0.05), cancer stage (OR = 3.01, 95% CI = 1.95–4.67, P < 0.05), myometrial invasion (OR = 1.59, 95% CI = 1.17–2.15, P < 0.05), lymph node metastasis (OR = 1.63, 95% CI = 1.18–2.26, P < 0.05), and prognosis (OR = 2.91, 95% CI = 1.17–4.66, P < 0.05) in endometrial cancer. Conclusions: Our findings suggested that COX-2 overexpression is significantly associated with poor prognosis and advanced clinical features in endometrial cancer. Therefore, COX-2 may function as an effective prognostic biomarker and a potential therapeutic target for endometrial cancer.
... COX-2 overexpression can affect adhesion properties of intestinal cells [72] and promote matrix metalloproteinase activity and thus cancer invasion [73]. Inhibition of COX-2 can antagonize colorectal metastasis in human [74] and in mouse [75] models. c-Met, hepatocyte growth factor receptor, may be transactivated by PGE2 via EGFR in case of colorectal cancer [76] which is linked with loss of cell-cell contact and invasion [77]. ...
Article
Full-text available
Colorectal cancer is the third common cancer in this world, accounting for more than 1 million cases each year. However, detailed etiology and mechanism of colorectal cancer have not been fully understood. For example, cyclooxygenase-2 (COX-2) and its product prostaglandin E2 (PGE2) have been closely linked to its occurrence, progression and prognosis. However, the mechanisms on how COX-2 and PGE2-mediate the pathogenesis of colorectal cancer are obscure. In this review, we have summarized recent advances in studies of pathogenesis and control in colorectal cancer to assist further advances in the research for the cure of the cancer. In addition, the knowledge gained may also guide the audiences for reduction of the risk and control of this deadly disease.
... There is evidence that COX-2 plays a crucial role in various aspects of carcinogenesis and is overexpressed in many types of malignant tumors, such as colorectal, prostate, breast lung, OS, and chondrosarcoma (20)(21)(22)(23)(24)(25)(26)(27)(28). Many NSAIDs, including indomethacin, have been shown to be toxic to certain malignancies and clinical findings suggest that it is a useful therapeutic agent (29)(30)(31)(32)(33)(34)(35). In addition, results from a meta-analysis have shown that NSAID usage significantly reduced the risk of metastasis development in prostate and breast cancer patients (36). ...
Article
Recurrence or metastasis remains the major cause of poor prognosis and mortality in Osteosarcoma patients. Therefore, development of more effective therapeutic approaches is required. We showed that indomethacin, significantly induces apoptosis in MNNG/HOS cell line, which was confirmed by morphological changes, increased Annexin-V + cells and nuclear fragmentation. Apoptosis was accompanied by increased cleavage of caspase-3 and PARP, suggesting activation of caspase-dependent cell death. Indomethacin significantly decreased the expression of β-catenin, a key player in tumor metastasis. These results indicate that indomethacin may have the potential to be used as neoadjuvant or adjuvant treatment; however, additional studies are required.
... Administration of COX-2 inhibitors decreases the survival, migration and invasion of endometriotic cells as a result of decreased production of PGE2 [127,134]. Additionally, COX-2-associated migration and invasion are decreased when COX-2 is inhibited in endometriotic cells, and are mediated by matrix metalloproteinase (MMP)-2 and MMP-9 in humans [135]. In addition, there is an interesting observation that COX-2 inhibitors produce more detrimental effects on invasion compared with migration in endometriotic cells; however, the underlying molecular mechanisms of these selective effects are unknown [21]. ...
Article
Full-text available
Endometriosis (EMS) is the most common gynecological disease in women of reproductive age, and it is associated with chronic pelvic pain, dyspareunia and infertility. As a consequence of genetic, immune and environmental factors, endometriotic lesions have high cyclooxygenase (COX)-2 and COX-2-derived prostaglandin E2 (PGE2) biosynthesis compared with the normal endometrium. The transcription of the PTGS2 gene for COX-2 is associated with multiple intracellular signals, which converge to cause the activation of mitogen-activated protein kinases (MAPKs). COX-2 expression can be regulated by several factors, such as estrogen, hypoxia, proinflammatory cytokines, environmental pollutants, metabolites and metabolic enzymes, and platelets. High concentrations of COX-2 lead to high cell proliferation, a low level of apoptosis, high invasion, angiogenesis, EMS-related pain and infertility. COX-2-derived PGE2 performs a crucial function in EMS development by binding to EP2 and EP4 receptors. These basic findings have contributed to COX-2-targeted treatment in EMS, including COX-2 inhibitors, hormone drugs and glycyrrhizin. In this review, we summarize the most recent basic research in detail and provide a short summary of COX-2-targeted treatment.
... Another possibility is that β-catenin mutation might be an independent risk factor for HCC in addition to cirrhosis. (143). Given the side effect profile associated with Cox-2 inhibitors, development of similar compounds which lack these side effects is underway. ...
Article
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide due to poor prognosis and limited therapeutic options. Point mutations affecting phosphorylation sites such as serine-45 in Beta-catenin gene are evident in around 30% of HCC. We developed a transgenic mouse that expresses Ser45 mutated Beta-catenin (TG) in hepatocytes. While this mutation did not induce spontaneous tumorigenesis, it promoted diethylnitrosamine (DEN)-induced HCC through cyclin-D1 overexpression and other factors.The Wnt/Beta-catenin signaling is important in stem cell self-renewal. The adult progenitor cell of the liver, or oval cells that emanate from atypical ductular proliferation (ADP), maybe involved in liver regeneration and/or hepatocarcinogenesis and can be observed after exposure to DDC diet, which induces hepatic and biliary injury. When challenged with chronic-DDC diet, Beta-catenin transgene led to a cellular disparity in the form of increased appearance of atypical hepatocytes (positive for ductular marker A6), which was associated with better resolution of intrahepatic cholestasis. We also utilized DDC diet in conditional Beta-catenin knockout mice (KO) that lacked Beta-catenin in hepatocytes and cholangiocytes. ADP was blunted after short-term DDC feeding in KO mice; however, long-term feeding resulted in gradual increase in ADP, hepatic fibrosis and HCC. Interestingly, the KO livers begin to exhibit periportal Beta-catenin-positive hepatocytes, which eventually populate the entire livers over the course of this process. Finally, we explored targeting of the Wnt pathway with pegylated interferon-alpha2A (Peg-IFN). We found that Peg-IFN decreased Beta-catenin activity in mouse liver and several human hepatoma cell lines. The mechanism seemed to be at least partly due to upregulation of a nuclear export factor, RanBP3.Thus, this study characterizes an animal model utilizing Beta-catenin mutation, which is evident in HCC patients. DEN-exposure in these animals led to HCC development, thus providing a valuable tool to study mechanisms of hepatocarcinogenesis and providing a model to test therapeutic inhibition of Beta-catenin by agents such as peg-IFN and others. Our studies also provide evidence that Wnt activation may resolve intrahepatic cholestasis. Finally, we show that chronic damage to the liver in KO led to appearance of Beta-catenin-positive hepatocytes, which continued to proliferate and in the face of continued injury and fibrosis, led to development of HCC, which is also relevant clinically.
... The function of the ratelimiting enzyme prostaglandin-endoperoxide synthase (also known as cyclooxygenase or COX) is involved in the oxidative transformation of arachidonic acid into various prostaglandin compounds [42]. In the last 10 years, many studies have demonstrated a relationship between COX-2 overexpression, tumor growth, and increasing VEGF expression for angiogenesis Sang Hun Lee, et al: VEGF pathway in tumor angiogenesis in many solid tumors, including gastric [43], colon [44], prostate [45], breast [46], and pancreatic [47] cancers. Moreover, many in vivo research showed the VEGF expression by COX-2 in many cell lines [41]; however, COX-2 inhibitors do not affect VEGF expression in all tumors, indicating that this effect may be tumor-dependent. ...
Article
Full-text available
The shaping of new blood vessels is a significant event in cancer growth and metastasis. Therefore, the molecular system of cancer angiogenesis has garnered considerable interest in cancer research. The vascular endothelial growth factor (VEGF) and VEGF receptor pathway are recognized as the key regulators of the angiogenic process. Activation of the VEGF/VEGF-receptor pathway initiates signaling cascades that promote endothelial cell growth, migration, and differentiation. Recently, VEGF was shown to play a role in the recruitment of bone marrow-derived endothelial progenitor cells to neovascularization sites. The role of VEGF in promoting tumor angiogenesis and the occurrence of human cancers has led to the rational design and development of agents that selectively target this pathway. Moreover, these anti-VEGF/VEGF receptor agents show therapeutic potential by inhibition of angiogenesis and tumor growth in preclinical models. In this review, we summarize the role of the VEGF pathway during tumor angiogenesis.
Article
Full-text available
Background: The link between inflammation and cancer has been confirmed by the use of anti-inflammatory therapies in cancer prevention and treatment. 5-aminosalicylic acid (5-ASA) was shown to decrease the growth and survival of colorectal cancer (CRC) cells. Studies also revealed that metformin induced apoptosis in several cancer cell lines. Methods: We investigated the combinatory effect of 5-ASA and metformin on HCT-116 and Caco-2 CRC cell lines. Apoptotic markers were determined using western blotting. Expression of pro-inflammatory cytokines was determined by RT-PCR. Inflammatory transcription factors and metastatic markers were measured by ELISA. Results: Metformin enhanced CRC cell death induced by 5-ASA through significant increase in oxidative stress and activation of apoptotic machinery. Moreover, metformin enhanced the anti-inflammatory effect of 5-ASA by decreasing the gene expression of IL-1β, IL-6, COX-2 and TNF-α and its receptors; TNF-R1 and TNF-R2. Significant inhibition of activation of NF-κB and STAT3 transcription factors, and their downstream targets was also observed. Metformin also enhanced the inhibitory effect of 5-ASA on MMP-2 and MMP-9 enzyme activity, indicating a decrease in metastasis. Conclusion: The current data demonstrate that metformin potentiates the antitumor effect of 5-ASA on CRC cells suggesting their potential use as an adjuvant treatment in CRC.
Chapter
Inflammation plays an important role in the development and progression of many forms of cancer, including colorectal cancer (CRC). Several lines of evidence support a major role for an inflammatory background in CRC: (1) inflammatory bowel disease patients have a higher risk of developing CRC; (2) regular use of non-steroidal anti-inflammatory drugs (e.g., aspirin) prevents CRC development in cancer-free patients and promotes regression of established cancers; and (3) even CRCs that do not develop under inflammatory conditions are strongly infiltrated with multiple types of pro-tumorigenic immune cells. In this chapter, the association between chronic inflammation and CRC is reviewed and the major molecular mechanisms leading to tumor development are summarized. In addition, the contributions of distinct immune cell populations for cancer progression are discussed. Finally, the implications of these associations for cancer prevention and treatment are highlighted.
Chapter
Genetic studies in species such as Xenopus, Drosophila, and Caenorhabditis have lent themselves quite well to further our understanding of the molecular basis of human diseases. A classical example is the identification and characterization of the Wnt/β-catenin pathway that is crucial in normal development including embryogenesis, organogenesis, and epithelial-mesenchymal transition and at the same time its deregulation is implicated in disorders such as cancers (reviewed in [1–3]). This pathway has remained conserved through the evolutionary process. In Drosophila, the role of Wnt or Wingless (Wg) was initially identified in normal wing development, however, it was later recognized for multiple functions such as inducing segment polarity and anterior–posterior patterning that were imperative for a viable embryo [4–6]. As the importance of Wnt emerged, several key components of this pathway were identified. The discovery of armadillo (or β-catenin) added a significant player to this orchestra and although circumstantial evidence suggesting such a relationship existed earlier it was a few years later that β-catenin was positively identified as a central component of the canonical Wg pathway [5, 7–9]. These studies led to the emergence of a model system for cell adhesion and signal transduction [10]. This was also the beginning of the understanding of the Wnt/β-catenin pathway and its role in complex cellular processes such as cell–cell adhesion, mitogenesis, motogenesis, and morphogenesis in the vertebrates.
Article
Full-text available
Cyclooxygenase-2 (COX-2), the enzyme at the rate-limiting step of prostanoid production, has been found to be overexpressed in human lung cancer. To evaluate lung tumor COX-2 modulation of antitumor immunity, we studied the antitumor effect of specific genetic or pharmacological inhibition of COX-2 in a murine Lewis lung carcinoma (3LL) model. Inhibition of COX-2 led to marked lymphocytic infiltration of the tumor and reduced tumor growth. Treatment of mice with anti-PGE2 mAb replicated the growth reduction seen in tumor-bearing mice treated with COX-2 inhibitors. COX-2 inhibition was accompanied by a significant decrement in IL-10 and a concomitant restoration of IL-12 production by APCs. Because the COX-2 metabolite PGE2 is a potent inducer of IL-10, it was hypothesized that COX-2 inhibition led to antitumor responses by down-regulating production of this potent immunosuppressive cytokine. In support of this concept, transfer of IL-10 transgenic T lymphocytes that overexpress IL-10 under control of the IL-2 promoter reversed the COX-2 inhibitor-induced antitumor response. We conclude that abrogation of COX-2 expression promotes antitumor reactivity by restoring the balance of IL-10 and IL-12 in vivo.
Article
Full-text available
Tumor-derived prostaglandin E2 (PGE2) modifies cytokine balance and inhibits host immunity. We hypothesized that a high level of PGE2 production by lung tumor cells is dependent on tumor cyclooxygenase (COX)-2 expression. We found that PGE2 production by A549 non-small cell lung cancer (NSCLC) cells was elevated up to 50-fold in response to interleukin (IL)-1beta. Reversal of IL-1beta-induced PGE2 production in A549 cells was achieved by specific pharmacological or antisense oligonucleotide inhibition of COX-2 activity or expression. In contrast, specific COX-1 inhibition was not effective. Consistent with these findings, IL-1beta induced COX-2 mRNA expression and protein production in A549 cells. Specific inhibition of COX-2 abrogated the capacity of IL-1beta-stimulated A549 cells to induce IL-10 in lymphocytes and macrophages. Furthermore, specific inhibition of A549 COX-2 reversed the tumor-derived PGE2-dependent inhibition of macrophage IL-12 production when whole blood was cultured in tumor supernatants. Our results indicate that lung tumor-derived PGE2 plays a pivotal role in promoting lymphocyte and macrophage IL-10 induction while simultaneously inhibiting macrophage IL-12 production. Immunohistochemistry of human NSCLC tissues obtained from lung cancer resection specimens revealed cytoplasmic staining for COX-2 within tumor cells. This is the first description of functional COX-2 expression by NSCLC cells and the definition of a pathway whereby tumor COX-2 expression and a high level of PGE2 production mediate profound alteration in cytokine balance in the lung cancer microenvironment.
Article
Full-text available
Cyclooxygenase-2 (COX-2), a key enzyme in arachidonic acid metabolism, is overexpressed in many cancers. Inhibition of COX-2 by nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the risk of cancer development in humans and suppresses tumor growth in animal models. The anti-cancer effect of NSAIDs seems to involve suppression of tumor angiogenesis, but the underlying mechanism is not completely understood. Integrin alpha V beta 3 is an adhesion receptor critically involved in mediating tumor angiogenesis. Here we show that inhibition of endothelial-cell COX-2 by NSAIDs suppresses alpha V beta 3-dependent activation of the small GTPases Cdc42 and Rac, resulting in inhibition of endothelial-cell spreading and migration in vitro and suppression of fibroblast growth factor-2-induced angiogenesis in vivo. These results establish a novel functional link between COX-2, integrin alpha V beta 3 and Cdc42-/Rac-dependent endothelial-cell migration. Moreover, they provide a rationale to the understanding of the anti-angiogenic activity of NSAIDs.
Article
Full-text available
A method has been devised for the electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets. The method results in quantitative transfer of ribosomal proteins from gels containing urea. For sodium dodecyl sulfate gels, the original band pattern was obtained with no loss of resolution, but the transfer was not quantitative. The method allows detection of proteins by autoradiography and is simpler than conventional procedures. The immobilized proteins were detectable by immunological procedures. All additional binding capacity on the nitrocellulose was blocked with excess protein; then a specific antibody was bound and, finally, a second antibody directed against the first antibody. The second antibody was either radioactively labeled or conjugated to fluorescein or to peroxidase. The specific protein was then detected by either autoradiography, under UV light, or by the peroxidase reaction product, respectively. In the latter case, as little as 100 pg of protein was clearly detectable. It is anticipated that the procedure will be applicable to analysis of a wide variety of proteins with specific reactions or ligands.
Article
Full-text available
TIS10 is a primary response gene whose cDNA was cloned as a result of its rapid, superinducible expression in Swiss 3T3 cells in response to 12-O-Tetradecanoylphorbol-13-acetate. The 5'-untranslated region of the 3.9-kilobase TIS10 message contains only 124 nucleotides, whereas the 3'-untranslated region is almost 2 kilobases in length. Within this long 3' region, there are multiple repeats of the sequence ATTTA, a sequence often present in rapidly degraded mRNA species. Primer extension revealed that the TIS10 cDNA begins 16 base pairs downstream of the transcription start site for the TIS10 gene. The TIS10 cDNA encodes a predicted protein of 604 amino acids. A computer search identified striking similarities between the predicted TIS10 protein product and the murine, sheep, and human prostaglandin synthase/cyclooxygenase proteins. The TIS10 protein has many of the same conserved amino acids that are thought to be important for cyclooxygenase function. TIS10 mRNA is undetectable by Northern analysis in quiescent 3T3 cells. The TIS10 gene is rapidly and transiently induced by forskolin and serum, as well as by 12-O-tetradecanoylphorbol-13-acetate, in Swiss 3T3 cells. These agents elicit far more dramatic changes in TIS10 mRNA levels than in cyclooxygenase mRNA levels. The expression of the TIS10 gene appears to be highly cell type-restricted in cultured cell lines; of 12 cell lines tested under superinducing conditions, only the rodent embryonic Swiss 3T3 and Rat1 cell lines expressed TIS10 gene.
Article
Recent epidemiologic studies have shown a 40-50% reduction in mortality from colorectal cancer in individuals who take nonsteroidal antiinflammatory drugs on a regular basis compared with those not taking these agents. One property shared by all of these drugs is their ability to inhibit cyclooxygenase (COX), a key enzyme in the conversion of arachidonic acid to prostaglandins. Two isoforms of COX have been characterized, COX-1 and COX-2. COX-2 is expressed at high levels in intestinal tumors in humans and rodents. Human colon cancer cells (Caco-2) were permanently transfected with a COX-2 expression vector or the identical vector lacking the COX-2 insert. The Caco-2 cells, which constitutively expressed COX-2, acquired increased invasiveness compared with the parental Caco-2 cells or the vector transfected control cells. Biochemical changes associated with this phenotypic change included activation of metalloproteinase-2 and increased RNA levels for the membrane-type metalloproteinase. Increased invasiveness and prostaglandin production were reversed by treatment with sulindac sulfide, a known COX inhibitor. These studies demonstrate that constitutive expression of COX-2 can lead to phenotypic changes that alter the metastatic potential of colorectal cancer cells.
Article
The effect of endogenous glucocorticoids on the expression of the cyclooxygenase enzyme was studied by contrasting cyclooxygenase expression and prostanoid synthesis in adrenalectomized and sham-adrenalectomized mice with or without the concurrent administration of endotoxin. Peritoneal macrophages obtained from adrenalectomized mice showed a 2- to 3-fold induction in cyclooxygenase synthesis and activity when compared to sham controls. Intravenous injection of a sublethal dose of endotoxin (5 micrograms/kg) further stimulated cyclooxygenase synthesis, resulting in a 4-fold increase in prostaglandin production. Similar cyclooxygenase induction can be achieved in macrophages obtained from normal mice but only after high doses of endotoxin (2.5 mg/kg) that are 100% lethal to adrenalectomized mice. Restoration of glucocorticoids in adrenalectomized animals with dexamethasone completely inhibited the elevated cyclooxygenase and protected these animals from endotoxin-induced death. In contrast, no signs of cyclooxygenase induction were observed in the kidneys of the adrenalectomized mice, even when treated with endotoxin. Dexamethasone did not affect the constitutive cyclooxygenase activity and prostaglandin production present in normal and adrenalectomized kidneys. These data indicate the existence of a constitutive cyclooxygenase that is normally present in most cells and tissues and is unaffected by steroids and of an inducible cyclooxygenase that is expressed only in the context of inflammation by proinflammatory cells, like macrophages, and that is under glucocorticoid regulation. Under normal physiological conditions glucocorticoids maintain tonic inhibition of inducible cyclooxygenase expression. Depletion of glucocorticoids or the presence of an inflammatory stimulus such as endotoxin causes rapid induction of this enzyme, resulting in an exacerbated inflammatory response that is often lethal.
Article
Twelve hundred ninety-six patients with resected colon cancer that either was locally invasive (Stage B2) or had regional nodal involvement (Stage C) were randomly assigned to observation or to treatment for one year with levamisole combined with fluorouracil. Patients with Stage C disease could also be randomly assigned to treatment with levamisole alone. The median follow-up time at this writing is 3 years (range, 2 to 5 1/2). Among the patients with Stage C disease, therapy with levamisole plus fluorouracil reduced the risk of cancer recurrence by 41 percent (P less than 0.0001). The overall death rate was reduced by 33 percent (P approximately 0.006). Treatment with levamisole alone had no detectable effect. The results in the patients with Stage B2 disease were equivocal and too preliminary to allow firm conclusions. Toxic effects of levamisole alone were infrequent, usually consisting of mild nausea with occasional dermatitis or leukopenia, and those of levamisole plus fluorouracil were essentially the same as those of fluorouracil alone--i.e., nausea, vomiting, stomatitis, diarrhea, dermatitis, and leukopenia. These reactions were usually not severe and did not greatly impede patients' compliance with their regimen. We conclude that adjuvant therapy with levamisole and fluorouracil should be standard treatment for Stage C colon carcinoma. Since most patients in our study were treated by community oncologists, this approach should be readily adaptable to conventional medical practice.