The present study was designed to determine whether: (a) chronic administration of dietary celecoxib (Celebrex), a potent nonsteroidal anti-inflammatory drug, which targets the cyclooxygenase-2 (COX-2) enzyme, negatively impacts host immunity; and (b) celecoxib can be coupled with a poxvirus-based vaccine to impact tumor burden in a murine tumor model of spontaneous adenomatous polyposis coli. Naive mice fed the celecoxib-supplemented diets developed eosinophilia with lowered plasma prostaglandin E2 levels and reduced COX-2 mRNA expression levels in their splenic T cells. Responses of splenic T, B, and natural killer cells to broad-based and antigen-specific stimuli were, for the most part, unchanged in those mice as well as COX-2 knockout mice; exceptions included: (a) reduced IFN-γ production by concanavalin A- or antigen-stimulated T cells; and (b) heightened lipopolysaccharide response of naive B cells from mice fed a diet supplemented with 1000 ppm of celecoxib. When transgenic mice that express the human carcinoembryonic antigen (CEA) gene (CEA transgenic) were bred with mice bearing a mutation in the ApcΔ850 gene (multiple intestinal neoplasia mice), the progeny (CEA transgenic/multiple intestinal neoplasia) spontaneously develop multiple intestinal neoplasms that overexpress CEA and COX-2. Beginning at 30 days of age, the administration of a diversified prime/boost recombinant CEA-poxvirus-based vaccine regimen or celecoxib (1000 ppm)-supplemented diet reduced the number of intestinal neoplasms by 54% and 65%, respectively. Combining the CEA-based vaccine with the celecoxib-supplemented diet reduced tumor burden by 95% and significantly improved overall long-term survival. Both tumor reduction and improved overall survival were achieved without any evidence of autoimmunity directed at CEA-expressing or other normal tissues. Celecoxib is prescribed for the treatment of familial adenomatous polyposis in humans, and the CEA-based vaccines have been well tolerated and capable of eliciting anti-CEA host immune responses in early clinical studies. The results suggest that the administration of a recombinant poxvirus-based vaccine is compatible with celecoxib, and this combined chemoimmuno-based approach might lead to an additive therapeutic antitumor benefit not only in patients diagnosed with familial adenomatous polyposis but, perhaps, in other preventive settings in which COX-2 overexpression is associated with progression from premalignancy to neoplasia.

Both chemotherapeutic and immunological-based approaches have been independently explored as potential strategies for the intervention of colorectal cancer. One chemotherapeutic/prevention approach that has proven successful in both experimental models and the treatment of familial adenomatous polyposis (FAP) in humans is the selective targeting of the cyclooxygenase (COX) pathway. Two COX isoenzymes have been identified: COX-1 is expressed in most tissues and necessary for healthy mucosa, kidneys, and platelets (1), and COX-2, which is virtually undetectable in most tissues, but induced in response to inflammation, cytokines, growth factors, and other stimuli (2, 3). Both COX isoenzymes convert arachidonic acid to prostaglandin H2, which serves as the substrate for a number of prostaglandin synthetases. Additionally, COX overexpression has been linked with resistance to apoptosis and tumor growth promotion (4). A regular intake of nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin, ibuprofen, and sulindac, inhibits both COX enzymes with an associated reduction of cancer risk (5, 6, 7, 8, 9). Subsequent findings reported that prolonged suppression of COX-1 activity caused unwanted side effects (10), which underscored the need to develop selective COX-2 inhibitors. COX-2 mRNA and protein are overexpressed in neoplastic epithelial cells (11, 12, 13), and COX-2 interruption by pharmacological agents or gene knockout reduced tumor development in a variety of experimental murine models (14, 15, 16). Celecoxib, one of the most studied COX-2 inhibitors, reduced the multiplicity and size of intestinal tumors (primarily adenomas) in the multiple intestinal neoplasia (MIN) mouse model (17). In a subsequent clinical study, oral celecoxib (Celebrex) administration reduced the number of colorectal polyps in patients with FAP significantly (18).

A separate investigational path has involved the generation and evaluation of recombinant cancer vaccines directed against carcinoembryonic antigen (CEA), a Mr 180,000–200,000 glycoprotein expressed by normal human colonic mucosa. CEA overexpression by a high percentage of human colorectal cancer, colonic polyps (19), and other adenocarcinomas (20, 21) provided an opportunity for the immune system to generate anti-CEA host immune responses and thereby served as the rationale to develop CEA-based vaccines. Using preclinical mouse models expressing the complete human CEA gene as a transgene (22, 23), several different CEA-directed vaccines were reported to overcome CEA immune tolerance by inducing anti-CEA-specific immunity which, in turn, correlated with the regression of CEA-expressing tumors (24, 25, 26, 27, 28, 29). Subsequent clinical studies have demonstrated the generation of anti-CEA host T-cell immune responses after vaccination with recombinant poxviruses expressing CEA, three costimulatory molecules (B7.1, ICAM-1, and LFA-3, designated TRICOM), and granulocyte-macrophage colony-stimulating factor (GM-CSF; Refs. 30, 31, 32). Phase I clinical trials have shown evidence of some reductions in serum levels of tumor markers and antitumor responses, as well as prolonged survival after vaccination. Studies are under way to determine whether those CEA-specific immune responses are indeed associated with the improved clinical outcome of vaccinated patients.

The present study was designed to investigate whether COX-2 and CEA can be simultaneously targeted in a combined chemoimmuno-based approach to cancer prevention and/or therapy. Such an approach, perhaps, might lead one to argue that combining a potent anti-inflammatory agent, such as celecoxib, with a proinflammatory vaccine might be counterproductive. Conflicting data exist as to whether a reduction of COX-2 levels in T cells is associated with functional changes (33, 34). To address those concerns, initial studies were undertaken to determine whether chronic exposure to celecoxib, a strong COX-2 inhibitor, might alter innate and/or adaptive host immune responses. Next, celecoxib and the CEA-based vaccine were combined in the CEA transgenic (CEA.Tg)/MIN mouse model in which mice spontaneously develop numerous intestinal tumors that overexpress CEA and COX-2. Results clearly show that host immunity remains, for the most part, unchanged in mice fed celecoxib-supplemented diets, and treatment of CEA.Tg/MIN mice with celecoxib combined with a CEA-based vaccine significantly reduced tumor multiplicity and prolonged survival. The findings demonstrate that CEA and COX-2 can be simultaneously targeted with a cancer vaccine and a COX-2 inhibitor in a combined chemoimmuno-based approach to cancer prevention/treatment.

Mice and Celecoxib-Supplemented Diets.

Six-8-week-old female C57BL/6 (B6; H-2b) mice were purchased from Taconic Farms (Germantown, NY). Mice expressing the gene for human CEA [CEA.Tg, Line 2682, C57BL/6 (H-2b), heterozygous] were obtained from John Thompson (University of Freiburg, Freiburg, Germany). MIN (C57BL/6J-ApcMIN/+) mice were purchased from The Jackson Laboratory (Bar Harbor, ME) and both colonies were continuously back-crossed with C57BL/6 mice. CEA.Tg/MIN mice were derived and screened for CEA and MIN expression as reported previously (29). Briefly, all CEA.Tg/MIN mice were derived by breeding female CEA.Tg mice with male MIN mice. Fecal and blood samples were taken from the CEA.Tg x MIN F1 offspring at weaning, and the presence of the CEA transgene and the Apc mutation identified by fecal CEA protein levels was detected using a solid-phase, double-determinant, anti-CEA ELISA kit (AMDL, Inc. Tustin, CA), and allele-specific PCR analysis of DNA isolated from the blood, respectively. All of the genotypes were rechecked at the completion of the study. COX-2 +/+ and COX-2 knockout (KO; B6;129P2-Ptgs2tml; Ref. 35) mice were generously provided by Dr. Robert Langenbach (National Institute of Environmental Health Sciences, Research Triangle Park, NC). Animal care was in compliance with recommendations of the Guide for Care and Use of Laboratory Animals, National Research Council.

Mice received Purina Certified Rodent Chow #5002 supplemented with 500, 1000, or 1500 ppm of celecoxib (Research Diets, Inc., New Brunswick, NJ) beginning at weaning.

Vaccines, Adjuvants, and Injection Schema.

Vaccines used were: (a) β-galactosidase (β-gal) protein (100 μg; Prozyme, Inc., San Leandro, CA) emulsified in incomplete Freund’s adjuvant; and (b) recombinant poxviruses [vaccinia (rV-) and fowlpox (rF-)] engineered to express the genes encoding bacterial LacZ or human CEA, and three murine costimulatory molecules, B7.1, ICAM-1, and LFA-3 (designated TRICOM); those vaccines are termed rV-, rF-LacZ-, rV-, or rF-CEA-TRICOM. Control vaccines contained the three costimulatory molecules alone and were designated either rV- or rF-TRICOM. Details of the construction and production of the recombinant vaccinia (36) and avipox (fowlpox) viruses (37) have been published. A description of the construction of the recombinant avipox (fowlpox) virus expressing murine GM-CSF (rF-GM-CSF) has been reported (25). All of the vaccines were administered s.c. in 100 μl HBSS at the base of the tail. Recombinant vaccinia-based vaccines, rV-CEA-TRICOM or rV-TRICOM, were the primary vaccines and administered at a dose of 108 plaque-forming units in combination with 107 plaque-forming units of rF-GM-CSF. Booster vaccinations were also administered s.c. in 100 μl containing 108 plaque-forming units of either rF-CEA-TRICOM or rF-TRICOM combined with 107 plaque-forming units of rF-GM-CSF. A group of mice received injections of the vehicle (HBSS) alone and designated the vehicle control.

Plasma Bicyclo-Prostaglandin E2 (PGE2) Assay.

To analyze celecoxib activity in mice fed the control diet or diets supplemented with 500 ppm, 1000 ppm, or 1500 ppm celecoxib, plasma PGE2 levels were measured. Because PGE2 is rapidly converted to 13,14-dihydro-15-keto-PGE2in vivo, the stable end metabolite bicyclo-PGE2 was measured as a surrogate marker for PGE2. Plasma from mice on the four diets was separated from whole blood using Vacutainer PST Gel lithium heparin tubes (Becton-Dickinson, Inc., Franklin Lakes, NJ). Approximately 3–5 ml of blood from 5 mice per group was collected into the Vacutainer PST Gel lithium heparin tubes, and the plasma was separated by centrifugation at 1000 rpm for 5 min at 4°C. Bicyclo-PGE2 levels were measured using a PGE2 metabolite EIA kit (Cayman Chemical, Inc., Ann Arbor, MI) according to manufacturer’s directions.

COX-2 mRNA Expression.

T cells were purified using Dynabeads B220 (Dynal, A.S., Oslo, Norway) from spleens isolated from mice fed either a control diet or a diet supplemented with 1500 ppm celecoxib. Five million T cells were incubated in complete medium in the presence or absence of 2 μg/ml concanavalin A (Con A) for 24 h. Total RNA was isolated from those cells using the RNAeasy RNA isolation kit (Qiagen, Inc., Valencia, CA). cDNA synthesis and COX-2-specific PCR amplification was performed by using Clontech Titanium One Tube reverse transcription-PCR (Clontech, Palo Alto, CA) with specific COX-2 primers. Briefly, 50 ng of RNA was reverse transcribed and amplified into COX-2-specific 500 bp PCR product by using the sense primer 5′GGA ACA TGG ACT CAC TCA 3′ and the antisense primer 5′ TAG GCT GTG GAT CTT GCA 3′. A 540-bp murine β-actin-specific PCR product was used as control and amplified by using a sense primer 5′-GTG GGC CGC TCT AGG CAC CAA-3′ and an antisense primer 5′-CTC TTT GAT GTC ACG CAC GAT TTC- 3′. DNA amplification was performed using a Perkin-Elmer Gene-AMP PCR System 9600 thermal cycler (Perkin-Elmer, Inc., Boston, MA). The amplification protocol consisted of the following denaturation, annealing, and elongation cycles: 50°C for 1 h, 94°C for 5 min, followed by 40 cycles at 94°C for 30 s, 57°C for 30 s, 68°C for 1 min, and 68°C for 2 min. Products were separated by gel electrophoresis on a 1.5–2.0% agarose gel, visualized under UV light using ethidium bromide, and quantified with normalization for β-actin expression using a Kodak 3.0 documentation and analysis system.

Serum Anti-β-gal and CEA Antibody Titers.

β-gal and CEA antibodies were measured by ELISA. Microtiter plates were sensitized overnight at 4°C with 100 ng/well β-gal CEA (International Enzymes, Fallbrook, CA), or ovalbumin (Sigma Chemicals, St. Louis, MO). Wells were blocked with DPBS containing 5% BSA, followed by a 1-h incubation of diluted mouse serum (1:10 to 1:31,250). Antibodies bound to the wells were detected with horseradish peroxidase-conjugated goat antimouse IgG (Kirkegaard & Perry Labs., Inc., Gaithersburg, MD) using an ELISA microplate autoreader at A490nm. Positive controls for β-gal and CEA were a commercially available mouse anti-β-galactosidase monoclonal antibody [IgG2a(κ), Promega Corp., Madison, WI] and a murine IgG2a anti-CEA monoclonal antibody, COL-1 (38). Antibody titers were determined as the reciprocal of the serum dilution that results in an A490 nm of 1.0.

Fluorescence-Activated Cell Sorter Analyses.

Splenocytes were prepared and analyzed using either single or double staining consisting of a FITC-conjugated antibody alone or combined with a phycoerythrin-conjugated antibody (25). FITC-conjugated antibodies used were antimouse CD3e (clone 145–2C11), antimouse CD4 (clone RM4–5), and antimouse NK1.1 (clone PK136). Phycoerythrin-labeled antibodies used were antimouse CD19 (clone 1D3), antimouse CD8a (clone 53–6.7), antimouse Ly-6G (Gr-1; clone RB6–8C5), and antimouse CD25 (clone 3C7; PharMingen, Inc., San Diego, CA). All of the samples also contained 1 μg of the unlabeled 2.2G2 antibody (CD16) to block Fc receptors. Data were gathered from 10,000 cells using a live gate.

Lymphoproliferation.

Mouse splenocytes were enriched for either B or T cells by magnetic murine pan T (Thy1.1) or B (B220) Dynabeads (Dynal, A.S.), and FACS analysis showed that the resulting cell population was >95% CD19+ or CD3+, respectively. Isolated B or T lymphocytes were resuspended in RPMI 1640 containing 15 mm HEPES (pH 7.4), 10% heat-inactivated fetal bovine serum, 2 mml-glutamine, 0.1 mm nonessential amino acids, 1 mm sodium pyruvate, 50 units/ml gentamicin, and 50 μm β-mercaptoethanol. B cells were incubated in the presence of 10.0–0.3 μg lipopolysaccharide/ml, whereas isolated T cells were incubated in the presence of 2.0–0.1 μg Con A/ml. After 2–3 days in culture, the cells were pulsed with [3H]thymidine (1 μCi/well; Amersham Corp., Arlington Heights, IL) and harvested 24 h later. Lymphoproliferative assays were also used to measure CEA or β-gal-specific CD4 T-cell responses. Purified splenic T cells from immunized mice against β-gal or CEA were coincubated with splenocytes from naive, syngeneic B6 mice as outlined above. CEA, β-gal, or ovalbumin (50–3.125 μg/ml) was added in the appropriate well of flat-bottomed, 96-well plates. Cells were pulsed with [3H]thymidine after 5 days, harvested 24 h later, and counted by liquid scintillation spectroscopy.

Cytokine Production Assays.

T cells were isolated from naive B6, COX +/+, COX KO mice, or B6 mice vaccinated with either β-gal or recombinant poxviruses expressing LacZ or CEA as described previously (25). T cells were incubated in flat-bottomed, 96-well plates in the presence of 5 × 106 irradiated syngeneic splenocytes and 10, 1, or 0.1 μg/ml of CEA526–533 (EAQNTTYL; Db-epitope; Ref. 39), β-gal96–103 (DAPIYTNV; Kb-epitope; Ref. 40), or a control peptide, Flu H3N2 influenza A virus nucleoprotein epitope (NP366–374; which is also a H-2Db epitope Flu NP366–374; Ref. 41). Supernatants were harvested 48 h later, and IFN-γ levels measured using an ELISA assay (Endogen, Inc., Cambridge, MA).

Tumor Scoring, Histopathology, and Immunohistochemical Staining.

CEA.Tg/MIN mice were sacrificed by CO2 inhalation and the entire gastrointestinal GI tract was removed. The intestine and colon were isolated by cutting at the point just distal to the gastric/duodenal border (pyloric sphincter) and the rectum at the anus. The intestine was cut into four sections, corresponding to the duodenum, proximal, and distal jejunum, and the ileum, and placed on dampened filter paper with Dulbecco’s PBS. The colon was also excised and similarly prepared. Using microdissecting scissors, each segment was opened longitudinally, and the mucosal surface was rinsed free of content with Dulbecco’s PBS. Using a dissecting microscope (×10 magnification), each segment was scored (blinded to the scorer) for the presence of tumors. The smallest scorable gross tumor was ∼1 mm, and tumors were divided according to size <2 mm, 2–5 mm, and >5 mm. The sum of the number of intestinal and colonic tumors was the measure of total GI tumor burden.

For histopathological evaluation, intestine and colon tissues were fixed in 10% neutral buffered formalin, embedded in paraffin blocks, and processed by routine histological methods for H&E staining. The largest and smallest tumors from each gut segment were examined microscopically. Proliferative epithelial lesions had microscopic morphology typical of that described for MIN mice (42, 43). Lesions were classified as intestinal intraepithelial neoplasms (dysplasia and carcinomas in situ) if they did not involve the full thickness of the mucosa and did not compress adjacent tissue. Adenomas involved the full thickness of the mucosa, and compressed adjacent tissue and adenocarcinomas invaded the muscularis mucosae. The majority of gross lesions were either intraepithelial neoplasms or adenomas.

Long-Term Survival Studies.

CEA.Tg/MIN mice received two diets beginning at weaning, control and celecoxib (1000 ppm). Mice on either diet were placed into three treatment groups, CEA-based vaccine, non-CEA-based vaccine, and vehicle control. The mice remained on the appropriate diet for the entire study, and the rV-based primary vaccine was administered by 30 days of age and with subsequent monthly boosts with the rF-based vaccine for the duration of the study. Any mouse whose weight fell for 4 consecutive weeks and whose hematocrit level was ≤25 was sacrificed, age recorded, and the GI tract examined for tumor burdens.

Statistical Methods.

Survival estimates among treatment groups were generated using Kaplan-Meier survival analyses. Differences in survivor functions between treatment groups were assessed using the log-rank test. Two-way ANOVA was used to assess polyp number by treatment group and polyp size, and the interaction of treatment by polyp size category. Linear regression analyses were used to examine the independent association between treatment groups and tumor multiplicity. All of the analyses were conducted using STATA software (version 1.0; Stata Corp., College Station, TX), and statistical significance was accepted at the P < 0.05 level.

Celecoxib-Supplemented Diets.

At weaning (21–28 days of age), groups of B6 mice were placed on a standard mouse chow diet (control diet) or that same chow supplemented with 500, 1000, or 1500 ppm celecoxib, and their general health status was closely monitored. Two in vivo indicators of chronic NSAID administration, eosinophilia (>0.24 eosinophils/103 cells/μl; Ref. 44), and a drop in plasma PGE2 levels (45), appeared in mice fed the celecoxib-supplemented diets. After >2 months, eosinophilia was present in mice fed diets supplemented with 1000 and 1500 ppm celecoxib. Plasma levels of bicyclo-PGE2, a stable metabolite of PGE2, were reduced by 11.0%, 54.3%, and 63.9% in mice fed the 500, 1000, or 1500 ppm celecoxib-supplemented diets, respectively (normal plasma bicyclo-PGE2 levels = ∼1.0 ng/ml). Despite those changes, individual weight gain and complete blood count/differential counts were similar for mice in each of the four diet groups (data not shown).

Mice from each diet group were sacrificed, and their spleens were isolated and analyzed for any macroscopic and/or cellular phenotypic changes associated with the celecoxib-supplemented diets. No significant differences were observed for either individual spleen weights (72–77 mg) or total splenocyte number (∼100 million). Furthermore, there were no significant alterations in the percentages of CD19+ (55–60%), CD3+ (31–33%), natural killer (NK) cells (1.9–2.5%), GR-1+ granulocytes/neutrophils (9–11%), CD4/CD25+ (9–11%) splenocytes, or CD4:CD8 ratios among the groups of mice fed either the control or the celecoxib-supplement diets. No changes were found in the number of dendritic cells (CD11c+/I-Ab+) in any of the diet groups. Several studies have reported that COX-2 is transcriptionally up-regulated after activation of isolated human T cells in vitro, which was subsequently shown to be blocked by the addition of COX-2 inhibitors (33, 46). To analyze COX-2 expression in murine T cells, splenic T cells were purified from mice fed either the control diet or the 1500 ppm celecoxib-supplemented diet and incubated for 24 h in medium alone or medium containing 2 μg Con A/ml (Fig. 1). Reverse transcription-PCR-based analyses of the total RNA from splenic T cells from mice fed the control diet revealed very low COX-2 mRNA levels in unstimulated T cells, which were increased ∼9-fold by Con A stimulation (Fig. 1, bottom panel, relative units). In contrast, COX-2 mRNA levels were undetectable in either resting or Con A-stimulated T cells isolated from mice fed the 1500 ppm celecoxib-supplemented diet (Fig. 1).

Celecoxib Effects on Innate Splenic T, B, and NK Cell Functions.

In separate groups of mice, splenic T, B, and NK cells were purified from mice fed the control or celecoxib-supplemented diets to determine whether dietary exposure to celecoxib might alter their responses to broad-based stimuli. No differences were observed with respect to splenic (a) T-cell proliferative responses to Con A or anti-CD3; or (b) NK cytolysis of YAC-1 cells among the groups of mice fed either the control or celecoxib-supplemented diets (Table 1). In contrast, splenic B cells from mice fed the 1000 ppm celecoxib-supplemented diets had significantly higher proliferative responses to lipopolysaccharide (P < 0.05) than B cells from mice fed the control diet, or diets supplemented with 500 ppm or 1500 ppm celecoxib (Table 1). Splenic T cells from mice fed the celecoxib-supplemented diets did produce lower IFN-γ levels after in vitro stimulation with 1 μg Con A/ml, but no such reduction was observed with 2 μg Con A/ml (Table 1).

Antigen-Specific Host Immunity in Mice Fed the Celecoxib-Supplemented Diets and COX-2 KO Mice.

Mice fed the celecoxib-supplemented diets or COX-2 KO mice were vaccinated against β-gal protein using the whole β-gal protein in adjuvant or recombinant poxviruses expressing LacZ to examine whether reduction of COX-2 by dietary celecoxib or genetic ablation of the COX-2 gene alters host immune responses to different vaccine types. Strong anti-β-gal serum IgG antibody titers were found in all groups of mice vaccinated with β-gal protein in adjuvant, with significantly higher titers in mice fed the 1500 ppm celecoxib-supplemented diet (Fig. 2,A; P < 0.05 versus titers from mice fed the control, 500 ppm, or 1000 ppm celecoxib-supplemented diets). For example, serum anti-β-gal IgG titers in those mice were ∼31,000 [1/SD = 1.0 abs (@ A490 nm)]; 5–10-fold higher than the titers measured in mice fed either the control, or 500 ppm or 1,000 ppm celecoxib-supplemented diets. Splenic T cells from those mice were tested for their ability to generate a CD4-specific proliferative recall response to β-gal protein; similar β-gal-specific lymphoproliferative responses were found in all four groups of mice (Fig. 2 B). The measurement of IFN-γ and interleukin 4 levels in splenic T-cell supernatants after in vitro stimulation with exogenous addition of β-gal protein indicated a TH1-like response (IFN-γ > interleukin 4). Comparison of the relative levels of IFN-γ and interleukin 4 produced by splenic T cells from mice fed the control diet, or diets supplemented with 500 ppm, 1000 ppm, or 1500 ppm celecoxib revealed no significant differences (data not shown).

Another approach to examine what role COX-2 may play in host immunity used COX-2 KO mice. Similar to mice fed the celecoxib-supplemented diets, eosinophilia was found in most COX-2 KO mice as well as a slight, but reproducible decrease (5–10%) in plasma levels of bicyclo-PGE2 levels. Other investigators have reported eosinophilia in bronchoalveolar lavages of COX-2 KO mice after airway response to an allergen (47). No differences in spleen weights, total splenocyte counts, and B, T, and NK cell counts were apparent in the COX-2 KO mice when compared with those measured in the COX-2 +/+ mice (data not shown). COX-2 KO and COX-2 +/+ mice were subsequently vaccinated with a bacterial LacZ gene engineered into recombinant poxviruses that expressed three murine costimulatory molecules (rV/rF-LacZ-TRICOM). Both COX-2 +/+ and COX-2 KO mice developed equivalent serum anti-β-gal IgG titers (Fig. 3,A). β-Gal-specific lymphoproliferative responses were slightly, but not significantly, lower in the COX-2 KO mice (Fig. 3,B). However, IFN-γ production, an indicator of CD8-specific T-cell activation (40), was significantly (P < 0.05) lower as measured in the supernatants from those splenic T-cell cultures from immune COX-2 KO mice when compared with COX-2 +/+ mice (Fig. 3 C).

Antitumor Responses in CEA.Tg/MIN Mice by Combining Dietary Celecoxib (1000 ppm) with a CEA-Based Vaccine.

The results, thus far, indicated that the administration of an anti-inflammatory agent, such as celecoxib, with a proinflammatory recombinant poxvirus-based vaccine might be compatible in an experimental antitumor model. The CEA.Tg/MIN murine model was chosen to test that hypothesis. CEA.Tg/MIN mice were placed on either the control or celecoxib (1000 ppm)-supplemented diets beginning at 30 days of age. At that time, CEA.Tg/MIN mice also received the primary vaccination comprised of the vaccinia-based vaccine (rV-) with (rV-CEA-TRICOM) or without (rV-TRICOM) the CEA gene. Mice were administered the appropriate monthly boosts with either of the rF-based vaccines (i.e., rF-CEA-TRICOM or rF-TRICOM). Like the MIN mice, CEA.Tg/MIN mice (29) developed adult-onset anemia accompanied by severe, progressive weight loss, overt changes that are tightly linked with intestinal tumor burden and serve as good indicators of treatment response. Average weight gain of CEA.Tg/MIN mice fed the control diet and administered either the vehicle alone or the non-CEA-based vaccine was 5–6 g/mouse, with severe anemia developing in all of the mice by 150–160 days of age (Table 2). Total number of intestinal tumors in those two groups of CEA.Tg/MIN mice was 38.8 ± 3.2 (Fig. 4,A) and 37.8 ± 2.1 (Fig. 4,C). As reported previously, CEA.Tg/MIN mice that received the CEA-based vaccine gained more weight (8.5 ± 0.5 g; P < 0.05 versus vehicle or non-CEA-based vaccine-treated CEA.Tg/MIN) and maintained normal hematocrit levels (Table 2); these changes correlated with a significantly lower intestinal tumor burden (21.1 ± 2.6; P < 0.05 versus vehicle or non-CEA-based vaccine-treated CEA.Tg/MIN; Fig. 4,B). Adding celecoxib to the diet alone significantly increased the average weight gain with accompanying normal hematocrit levels of the CEA.Tg/MIN mice, whether or not they were vaccinated (Table 2). CEA.Tg/MIN mice on the celecoxib-supplemented diet alone had lower numbers of intestinal tumors (13.4 ± 2.0; Fig. 4,D; P < 0.001 versus mice fed the control diet and administered the vehicle alone; Fig. 4,A). No additional reduction in tumor multiplicity was achieved with the administration of the non-CEA-based vaccine (P = 0.78; Fig. 4,F). Combining the celecoxib-supplemented diet with the administration of the CEA-based vaccine resulted in the highest average weight gain (Table 2); this also correlated with a substantial reduction in the average number of intestinal tumors to 2.1 ± 0.6 (P < 0.001 versus vehicle control-treated mice, CEA-based vaccine, and control diet and celecoxib-treated mice; Fig. 4 E). Six of those 16 CEA.Tg/MIN mice receiving the combined treatment regimen remained tumor-free. Importantly, the effects of the CEA-based vaccine and celecoxib treatment appear to be additive as the linear regression coefficient of the combined treatment, i.e., CEA vaccine and celecoxib (β = −12.18), approximate the added effect of each treatment alone, i.e., CEA-based vaccine (β = −8.46) and celecoxib treatment (β = −5.93).

Individual tumor sizes were measured for all of the CEA.Tg/MIN mice in the treatment groups and divided as small (<2 mm), medium (2–5 mm), or large (>5 mm), and the percentage of total number of intestinal tumors in those three size categories was compared for each treatment group (Fig. 5). For CEA.Tg/MIN mice fed the control diet (Fig. 5,A) and administered the vehicle alone, 12.9%, 65.0%, and 22.2% of the tumors fell into the three size categories. Administration of the CEA-based vaccine slightly reduced the percentage of medium-sized (2–5 mm) tumors to 51.1%, but had no effect on the percentage of larger (>5 mm) tumors. The most dramatic reductions in tumor size were found in CEA.Tg/MIN mice fed the celecoxib-supplemented diets whether or not a vaccine was given (Fig. 5,B). In both the celecoxib-supplemented diet alone, and combined with either the non-CEA or CEA-based vaccine, 61–66% of the tumors were small tumors (<2 mm; P < 0.001 versus matched treatment groups of mice fed the control diet). Whereas the administration of the CEA-based vaccine did significantly decrease tumor multiplicity in the CEA.Tg/MIN mice (Fig. 4,E), the sizes of the tumors that did develop were no smaller when compared with the other mice fed the celecoxib-supplemented diet (Fig. 5 B).

CEA.Tg/MIN mice fed either the control or celecoxib-supplemented diets with or without vaccination were followed with periodic hematocrit readings (Fig. 6) and in a long-term (18 month) survival study (Fig. 7). The disease course in the CEA.Tg/MIN mice fed the control diet and treated with the vehicle alone included the onset of anemia by 90–110 days of age (Fig. 6,A) with progressive weight loss that required their sacrifice by 6 months of age (Fig. 7). Administration of the vector-control, non-CEA-based vaccine had little effect on overall survival (Fig. 7). As reported previously (29), administration of the CEA-based vaccine delayed the onset of anemia (Fig. 6,C) and significantly improved overall survival (P < 0.001 versus either vehicle control or non-CEA vaccine treated mice); 55% of CEA.Tg/MIN mice were alive at 13 months (Fig. 7). The celecoxib-supplemented diet additionally delayed the onset of anemia (Fig. 6, D–F) and prolonged survival (Fig. 7) in all groups of CEA.Tg/MIN mice whether or not they received a vaccine (P < 0.001 versus matched treatment groups of mice fed the control diet). CEA.Tg/MIN mice fed either the celecoxib-supplemented diet alone or combined with the control vaccine eventually developed anemia (Fig. 6, D and E) with progressive weight loss beginning at 11 months of age. By 18 months of age, overall survival in those two treatment groups of CEA.Tg/MIN mice was 40–60% of those mice. The most dramatic improvement in general health status (estimated by hematocrit levels) occurred in those CEA.Tg/MIN mice that were vaccinated monthly with the CEA-based vaccine and fed the celecoxib-supplemented diet. Hematocrit levels remained normal (Fig. 6 F) and at 18 months of age 100% (12 of 12) of those CEA.Tg/MIN mice were alive, a significant improvement in overall survival (χ2 = 25.27; P < 0.001 versus CEA-based vaccine alone, and χ2 = 7.76; P = 0.0054 versus celecoxib diet alone).

Anti-CEA Immunity.

Of interest was to examine whether there were any differences in CEA-specific host immunity in CEA.Tg/MIN mice vaccinated with the CEA-based vaccine and fed either the control or celecoxib-supplemented diets. Three age-matched (145–150 days of age) CEA.Tg/MIN mice from each group that had received four vaccinations (1 primary and 3 boosters), and had similar weight gains and hematocrit levels were analyzed for CEA-specific host immunity. Anti-CEA serum IgG titers and CD4-proliferative responses were significantly lower (P < 0.05) in the vaccinated CEA.Tg/MIN mice fed the control when compared with the mice fed the celecoxib-supplemented diet (Table 3). CD8-mediated, peptide-pulsed cytotoxicity was also lower in the CEA.Tg/MIN mice fed the control diet and administered the CEA-based vaccine, but the difference was not significant (P = 0.09). Intestinal tumor burden was significantly higher (P < 0.001) in the CEA.Tg/MIN mice fed the control diet and administered the CEA-based vaccine than in those mice fed the celecoxib-supplemented diet. No detectable CEA-specific immune responses were found in CEA.Tg/MIN that were fed either of the diets and vaccinated with the non-CEA-based vaccine or the vehicle control.

Histopathology.

Tables 4 and 5 summarize the histopathological analyses of the CEA-expressing (tongue, trachea, esophagus, stomach, intestine, caecum, and colon) and CEA-negative tissues. No macroscopic or microscopic abnormalities were found in the tongue, trachea, esophagus, and stomach of 4 individual CEA.Tg/MIN mice fed the celecoxib-supplemented diet and administered 5–6 injections of the CEA-based vaccine. Where indicated, adenomas and intraepithelial neoplasms were identified in the intestine and colon. In those GI tissues free of neoplasms, histopathological analyses found normal tissues with an occasional area of gut-associated lymphoid tissue hyperplasia. Gut-associated lymphoid tissue was also noted in CEA.Tg/MIN mice that were administered either the CEA-based vaccine or celecoxib as single agents. In a single CEA.Tg/MIN mouse treated with the CEA-based vaccine and the celecoxib, and found to be tumor-free, the entire GI tract was judged to be within normal histological limits.

In most tumor-bearing mice, enlarged spleens and hepatic extramedullary hematopoiesis were evident due to increased extramedullary hematopoiesis associated with anemia (Table 5). Subacute liver inflammation, the presence of a lipogenic pigment in the adrenals, lung lymphocytic infiltrate, and melanosis of a heart valve were also noted. Because those abnormalities were found in all of the treatment groups, their appearance did not seem to be associated with the administration of either the celecoxib-supplemented diet or the CEA-based vaccine.

This study was designed: (a) to examine whether the chronic administration of an anti-inflammatory NSAID (celecoxib) might impair the ability of the murine-adaptive immune system to respond to broad-based stimuli as well as to generate host immune responses to different antigens; and (b) to evaluate whether targeting CEA and COX-2 in the CEA.Tg/MIN mouse model with a CEA-based cancer vaccine and celecoxib may be efficacious as a chemoimmuno-based approach to tumor therapy. Previous results have reported that COX-2 mRNA and protein levels in human T cells rose after activation (33). However, after the in vitro addition of celecoxib, several events associated with T-cell activation, such as CD25/CD71 expression, cytokine production, and proliferation, were reduced (33, 46), suggesting that celecoxib and other COX-selective NSAIDs may down-regulate T-cell activation resulting in immune suppression. Upon additional examination, however, it was apparent that the in vitro celecoxib levels needed to inhibit T-cell activation were supra-physiological (10–100 μm) and, in a more recent study, down-regulation of COX-2 mRNA by the in vitro addition of COX-2 antisense oligonucleotides did not impede T-cell activation (34).

In the present study, B6 mice were fed diets supplemented with 500 ppm, 1000 ppm, or 1500 ppm celecoxib, levels shown previously to reduce intestinal tumor multiplicity and size in MIN mice (17). Blood celecoxib levels in the mice fed those diets have been reported to be 0.09, 0.26, and 1.80 μg/ml, respectively (17), comparable with the 0.5–2.0 μm concentrations in patients diagnosed with FAP and treated with 200 or 400 mg celecoxib twice/day (19). Mice fed the 1000-ppm and 1500-ppm celecoxib diets developed eosinophilia, which often coincides with chronic NSAIDs administration. Reduction in plasma PGE2 levels, as measured by plasma bicyclo-PGE2 levels, was also present in all groups of mice fed the celecoxib-supplemented diets, another indication of the in vivo bioactivity of celecoxib. Thus, the present study provides a physiologically relevant setting to examine celecoxib effects on COX-2 expression in T cells and the ability of the host to mount antigen-specific immune responses during chronic celecoxib administration.

The findings that in vivo celecoxib administration completely suppressed COX-2 mRNA levels in resting and Con A-stimulated splenic T cells are in accord with those from previous studies reporting suppression of COX-2 mRNA and protein levels after an acute in vitro exposure of human T cells to celecoxib (33). Moreover, the findings are also consistent with the hypothesis that COX-2 expression is regulated by a positive feedback loop, and any disruption within the pathway would be expected to suppress COX-2 expression levels (48). Despite the virtual absence of COX-2 mRNA, those splenic T cells responded normally to broad-based proliferative stimuli (i.e., Con A and anti-CD3; Table 1). Interestingly, splenic B cells from mice fed the 1000-ppm celecoxib diet had a heightened (P < 0.05) in vitro response to lipopolysaccharide, which was not seen for splenic B cells from mice fed either the 500- or 1500-ppm celecoxib diets. No apparent differences in NK-mediated cytolysis were observed in mice fed the celecoxib-supplemented diets. In other experiments, B6 mice fed the celecoxib-supplemented diets as well as COX-2 KO mice responded normally in mounting host immune responses after vaccination with either a whole protein (β-gal in incomplete Freund’s adjuvant) or recombinant poxviruses engineered to express the LacZ gene and three costimulatory molecules (rV-/rF-LacZ-TRICOM). In fact, mice fed the 1500-ppm celecoxib diet and vaccinated with the β-gal protein-based vaccine generated more robust anti-β-gal serum IgG titers than mice fed the control, or 500 or 1000 ppm celecoxib diets (Fig. 2 A).

At this point, the data suggested that disruption of the COX-2 isoenzyme by dietary celecoxib or by genetic ablation of the COX-2 gene resulted in some alterations in host immunity. On the one hand, IFN-γ production by T cells was reduced after (a) in vitro Con A stimulation of naive T cells from celecoxib-treated mice (Table 1); and (b) β-gal peptide-specific immune T cells from COX-2 KO mice vaccinated with the recombinant poxviruses (Fig. 3 C). In contrast, more robust anti-β-gal serum titers were found in mice fed the 1500-ppm celecoxib-supplemented diet and vaccinated with the β-gal protein in adjuvant. However, those increases were not observed in COX-2 KO mice, indicating that other celecoxib-sensitive pathways, such as Akt activation (49), may play a role. Whereas the initial data suggest that dietary celecoxib may mediate a modest shift from a TH1 to a TH2 response, preliminary evidence found no significant changes in the cytokine profiles from antigen-stimulated T cells from mice vaccinated with β-gal in adjuvant. Finally, in an experimental lung tumor model, abrogation of COX-2 expression, which reduced PGE2 levels, restored the balance between TH1 and TH2 cytokines (i.e., interleukin-10 and interleukin-12) resulting in normal dendritic cell function and more effective antitumor responses (50, 51). Those observations underscore the complexity of the interactions among COX enzymes, prostaglandin production, and host immunity, as well as the need to design future mechanistic studies focused on those questions.

Our immediate interests were to investigate the compatibility of the combined use of celecoxib and a tumor vaccine in an experimental mouse tumor model. The CEA.Tg/MIN spontaneous intestinal tumor model was chosen for those studies because (a) CEA.Tg/MIN mice develop multiple spontaneous GI tumors (MIN genotypes), which overexpress CEA (29) and COX-2 (17), and (b) treatment with either the CEA-based vaccine or dietary celecoxib alone was known to significantly reduce tumor multiplicity along the GI tract (17, 29). For the CEA-based vaccine to induce CEA-based antitumor host immunity, immune tolerance, due to the constitutive CEA along the GI tract, must be overcome. If the reduced IFN-γ production by Con A or β-gal peptide-stimulated splenic T cells from naive or immune mice might be linked with a diminution in antitumor immunity, that linkage might be best tested in a setting that requires the immune system to initially overcome immune tolerance against a weak, self-antigen. As reported previously (17, 29), administration of either the CEA-based vaccine or dietary celecoxib as single agents reduced the number of intestinal tumors in CEA.Tg/MIN mice by 54% and 65%, respectively. Combining the CEA-based vaccine with dietary celecoxib (1000 ppm) led to an additional reduction in intestinal tumor multiplicity, 95% reduction when compared with untreated CEA.Tg/MIN mice. In fact, 6 of 16 CEA.Tg/MIN mice administered monthly injections of the CEA-based vaccine and dietary celecoxib were tumor-free at 150–160 days of age. Another cohort of CEA.Tg/MIN mice that was administered the combined treatment regimen continued their weight gain with normal hematocrit levels resulting in a significant improvement in survival. At 18 months of age, 100% (12 of 12) of CEA.Tg/MIN mice that continued to receive monthly booster vaccinations of rF-CEA-TRICOM combined with rF-GM-CSF, as well as being maintained on the 1000-ppm celecoxib-supplemented diet, remained alive.

The antitumor mechanisms of the combined treatment of CEA.Tg/MIN mice with the CEA-based vaccine and dietary celecoxib are currently under study. The combined treatments began at 30 days of age, when the intestinal neoplasms found in most MIN mice are aberrant crypt foci (ACFMin), which are believed to be preneoplastic lesions that later form adenomas (52). Whether COX-2 is expressed by the ACFMin is unknown. In ApcΔ716 mice, however, COX-2 expression was found in polyp stromal cells and polyps >1 mm, not in polyps <1 mm, in diameter (53). If a similar COX-2 tissue expression profile exists in the CEA.Tg/MIN mice, then the target of celecoxib seems to be the polyp stroma, not the epithelial cells. Despite the dietary supplementation of celecoxib, which reduces tumor multiplicity, intestinal tumors do emerge and are attacked by CEA-specific immune cells. That combined intervention successfully reduced tumor multiplicity by 95%. Table 3 provides a direct comparison of the relevant strengths of the CEA-specific immune response in CEA.Tg/MIN mice fed either the control or celecoxib-supplemented diet and vaccinated four times with the CEA-based vaccine. Interestingly, in the CEA.Tg/MIN mice fed the celecoxib-supplemented diet and administered the CEA-based vaccine, serum anti-IgG and CD4-proliferative responses were significantly higher than in those CEA immune mice fed the control diet. Whereas the increase in serum anti-CEA IgG levels agreed with previous findings, the increase in CEA-specific lymphoproliferative responses seemed to contradict previous results. One possible explanation is that the presence of intestinal tumors in CEA.Tg/MIN mice fed the control diet and vaccinated with the CEA-based vaccine might dampen CEA-specific host immunity. Tumor-associated suppression of host immunity is well established and considered one of many tumor escape mechanisms (54). In this study, no direct evidence is presented that a CEA-specific immune response generated by the CEA-based vaccines combined with the antitumor properties of celecoxib resulted in the additive reduction of tumor multiplicity in CEA.Tg/MIN mice. However, in previous studies, that same vaccine regimen reduced the growth of CEA-expressing, transplantable s.c. tumors in those same CEA.Tg mice. Furthermore, antibody-based in vivo depletion studies revealed a requirement for CD4 and CD8 T cells, as well as NK cells to elicit the antitumor effects of the vaccine (55). Subsequent studies will investigate the temporal relationships between CEA and COX-2 expression in the tumor tissue. In particular, the intestinal tumors that form in CEA.Tg/MIN mice after the administration of the CEA-based vaccine alone were fewer in number, but the same size as those in the unvaccinated mice. Additional study will address whether the administration of the CEA-based vaccine results in an outgrowth of intestinal tumors with altered CEA and/or class I antigen expression, which may impact overall tumor growth rate. Furthermore, studies will investigate whether administration of the CEA-based vaccine alters COX-2 expression or other celecoxib-modulated biological actions (i.e., blocking Akt activation, antiangiogenesis, prostaglandin production, and so forth).

Those results provide another example of a CEA-based vaccination protocol that generates substantial antitumor immunity with little or no evidence of autoreactive T cells resulting in autoimmune pathology. Histological analyses of CEA-expressing normal tissues of the gastrointestinal tract in CEA.Tg/MIN mice that received 5–6 vaccinations revealed normal tissue architecture. As reported previously (29), the most prevalent pathological finding was splenic and hepatic extramedullary hematopoiesis, a compensatory response to developing anemia. The reasons(s) that the CEA-based vaccine elicits strong antitumor responses without accompanying autoimmune involvement remains an intriguing observation. Some possible explanations that offer avenues for future studies are: (a) a selective susceptibility of tumor tissue to immune attack due to a combination of the disruption of tissue architecture and CEA overexpression; and (b) a “braking” action of tolerizing antigen-presenting cells and/or regulatory T cells on autoreactive T cells (56).

Finally, the data provide a strong argument for future experimental as well as clinical efforts to combine cancer vaccines with NSAID targeting of COX expression. A recent study reported that COX-2 inhibition enhanced the efficacy of an adenovirus-based intratumoral therapy (57). The most immediate population that may benefit from a combined CEA-based vaccine with celecoxib is patients diagnosed with FAP. Celecoxib (Celebrex) is being prescribed for FAP patients based on a 30% reduction in polyp burden (19). In several early clinical studies, administration of the CEA-based vaccine has been well tolerated and able to generate anti-CEA host immune responses (30, 31, 32). One question that remains is whether CEA overexpression is present in the polyps of the FAP patients. CEA levels, as measured by quantitative analyses, were 2–6-fold higher in tubulovillous adenomas and hyperplastic polyps when compared with CEA levels in normal mucosa from healthy donors (18). A larger study is needed to not only examine CEA expression in colorectal polyps, but also to determine the tissue expression of COX-2. A high percentage of other carcinomas, including colorectal (FAP, hereditary nonpolyposis colorectal carcinoma, and sporadic), gastric, pancreas, breast (invasive and ductal carcinoma in situ), non-small cell lung, cervical, and head and neck (58, 59, 60, 61, 62, 63) overexpress both CEA and COX-2. Experimental studies are needed to determine the breadth of combining a cancer vaccine with targeting a COX isoenzyme. Whereas the present results encourage the use of this combined treatment to inhibit the progression from premalignancy to tumor, the effectiveness of treating overt cancers with such a combined chemoimmuno-based approach requires additional research.

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.

Requests for reprints: John Greiner, Laboratory of Tumor Immunology and Biology, Building 10, Room 8B09, MSC 1750, Bethesda, MD 20892.

Fig. 1.

Celecoxib effects on T cell cyclooxygenase (COX)-2 mRNA expression levels. Splenic T cells were purified from mice fed either the control diet (no celecoxib) or the 1500-ppm celecoxib-supplemented diet and incubated in the absence or presence of 2.0 μg/ml concanavalin A (Con A) for 24 h. Total RNA was isolated and the reverse transcription-PCR amplification using primer pairs for COX-2 and β-actin was carried out as described in “Materials and Methods.” Gel images were taken, and COX-2 mRNA transcript levels were quantified with normalization for β-actin expression as shown in the bottom panel. Data are from a single experiment that was repeated with similar results.

Fig. 1.

Celecoxib effects on T cell cyclooxygenase (COX)-2 mRNA expression levels. Splenic T cells were purified from mice fed either the control diet (no celecoxib) or the 1500-ppm celecoxib-supplemented diet and incubated in the absence or presence of 2.0 μg/ml concanavalin A (Con A) for 24 h. Total RNA was isolated and the reverse transcription-PCR amplification using primer pairs for COX-2 and β-actin was carried out as described in “Materials and Methods.” Gel images were taken, and COX-2 mRNA transcript levels were quantified with normalization for β-actin expression as shown in the bottom panel. Data are from a single experiment that was repeated with similar results.

Close modal
Fig. 2.

Adaptive host immune responses in B6 mice (5/group) fed either a control diet or diets supplemented with celecoxib and vaccinated twice with β-gal in incomplete Freund’s adjuvant at monthly intervals. Host immune responses were measured as described in “Materials and Methods” 1 month after the second vaccination. A, anti-β-gal serum IgG titers from mice fed the control diet (○), or diets supplemented with 500 ppm (▴), 1000 ppm (▪), or 1500 ppm (•) celecoxib. ▵ represent absorbance associated with incubation in the presence of normal mouse serum. Data represent the mean of triplicate determinations of individual serum samples from 3–5 mice/group and from two separate experiments; bars, ±SE. B, CD4-proliferative responses were measured using purified splenic CD4 T cells. Solid lines (—) represent β-gal-specific lymphoproliferative responses, whereas the dashed lines (---) represent lymphoproliferative responses to ovalbumin, a control antigen. B6 mice were fed a control diet (○), or diets supplemented with 500 ppm (▴), 1000 ppm (▪), or 1500 ppm (•) celecoxib. Data are the mean of triplicate determinations from a representative experiment that was repeated with similar results; bars, ±SE.

Fig. 2.

Adaptive host immune responses in B6 mice (5/group) fed either a control diet or diets supplemented with celecoxib and vaccinated twice with β-gal in incomplete Freund’s adjuvant at monthly intervals. Host immune responses were measured as described in “Materials and Methods” 1 month after the second vaccination. A, anti-β-gal serum IgG titers from mice fed the control diet (○), or diets supplemented with 500 ppm (▴), 1000 ppm (▪), or 1500 ppm (•) celecoxib. ▵ represent absorbance associated with incubation in the presence of normal mouse serum. Data represent the mean of triplicate determinations of individual serum samples from 3–5 mice/group and from two separate experiments; bars, ±SE. B, CD4-proliferative responses were measured using purified splenic CD4 T cells. Solid lines (—) represent β-gal-specific lymphoproliferative responses, whereas the dashed lines (---) represent lymphoproliferative responses to ovalbumin, a control antigen. B6 mice were fed a control diet (○), or diets supplemented with 500 ppm (▴), 1000 ppm (▪), or 1500 ppm (•) celecoxib. Data are the mean of triplicate determinations from a representative experiment that was repeated with similar results; bars, ±SE.

Close modal
Fig. 3.

Comparison of the adaptive immune responses of cyclooxygenase (COX)-2 knockout (KO) and COX-2 +/+ mice (3–5/group) given a primary vaccination of vaccinia-LacZ-TRICOM followed a month later with a booster vaccination of fowlpox-LacZ-TRICOM. Immune responses were measured 1 month after the booster vaccination. A, anti-β-gal serum IgG responses in COX-2 KO (▴) and COX-2 +/+ (•). Data are the mean of triplicate determinations from a representative experiment that was repeated with similar results; bars, ±SE. B, lymphoproliferative responses to soluble β-gal (circles) and ovalbumin (triangles), a control antigen, using purified T cells from COX-2 KO (closed symbols) and COX-2 +/+ (open symbols) mice. Data are the mean from triplicate wells of a representative experiment that was performed 2–3 times with similar results; bars, ±SE. C, IFN-γ production by splenic T cells from COX-2 KO (▴) and COX-2 +/+ (•) mice. Data are the mean from triplicate wells from a representative experiment that was repeated with similar results; bars, ±SE. ∗, P < 0.05 (versus COX-2 +/+ group).

Fig. 3.

Comparison of the adaptive immune responses of cyclooxygenase (COX)-2 knockout (KO) and COX-2 +/+ mice (3–5/group) given a primary vaccination of vaccinia-LacZ-TRICOM followed a month later with a booster vaccination of fowlpox-LacZ-TRICOM. Immune responses were measured 1 month after the booster vaccination. A, anti-β-gal serum IgG responses in COX-2 KO (▴) and COX-2 +/+ (•). Data are the mean of triplicate determinations from a representative experiment that was repeated with similar results; bars, ±SE. B, lymphoproliferative responses to soluble β-gal (circles) and ovalbumin (triangles), a control antigen, using purified T cells from COX-2 KO (closed symbols) and COX-2 +/+ (open symbols) mice. Data are the mean from triplicate wells of a representative experiment that was performed 2–3 times with similar results; bars, ±SE. C, IFN-γ production by splenic T cells from COX-2 KO (▴) and COX-2 +/+ (•) mice. Data are the mean from triplicate wells from a representative experiment that was repeated with similar results; bars, ±SE. ∗, P < 0.05 (versus COX-2 +/+ group).

Close modal
Fig. 4.

Total number of intestinal tumors in CEA.Tg/MIN mice that were fed either a control diet (A–C) or a celecoxib-supplemented (1000 ppm) diet (D–F). Mice also received either the vehicle alone (A and D), the CEA-based vaccine (B and E), or the non-CEA-based vaccine (C and F). • represent individual mice and horizontal lines (—) the mean number of tumors for each treatment group.

Fig. 4.

Total number of intestinal tumors in CEA.Tg/MIN mice that were fed either a control diet (A–C) or a celecoxib-supplemented (1000 ppm) diet (D–F). Mice also received either the vehicle alone (A and D), the CEA-based vaccine (B and E), or the non-CEA-based vaccine (C and F). • represent individual mice and horizontal lines (—) the mean number of tumors for each treatment group.

Close modal
Fig. 5.

Comparison of tumor sizes from CEA.Tg/MIN mice illustrated in Fig. 3. A and B show the percentages (Y axis) of total tumor number from mice fed a control diet or a celecoxib-supplemented diet (1000 ppm), respectively, and administered the vehicle alone (none), the non-CEA-based vaccine, or the CEA-based vaccine (X axis). Within each bar are shown the percentages of tumors that were <2 mm (▪), 2–5 mm (□), and >5 mm (). Number of mice (n) and total number of tumors measured for each group were: Control Diet, vehicle (n = 10; 373 tumors), non-CEA-based vaccine (n = 5; 168 tumors), CEA-based vaccine (n = 13; 273 tumors); Celecoxib Diet, vehicle (n = 12; 161 tumors), non-CEA-based vaccine (n = 6; 56 tumors), CEA-based vaccine (n = 16; 36 tumors).

Fig. 5.

Comparison of tumor sizes from CEA.Tg/MIN mice illustrated in Fig. 3. A and B show the percentages (Y axis) of total tumor number from mice fed a control diet or a celecoxib-supplemented diet (1000 ppm), respectively, and administered the vehicle alone (none), the non-CEA-based vaccine, or the CEA-based vaccine (X axis). Within each bar are shown the percentages of tumors that were <2 mm (▪), 2–5 mm (□), and >5 mm (). Number of mice (n) and total number of tumors measured for each group were: Control Diet, vehicle (n = 10; 373 tumors), non-CEA-based vaccine (n = 5; 168 tumors), CEA-based vaccine (n = 13; 273 tumors); Celecoxib Diet, vehicle (n = 12; 161 tumors), non-CEA-based vaccine (n = 6; 56 tumors), CEA-based vaccine (n = 16; 36 tumors).

Close modal
Fig. 6.

Hematocrit levels from individual CEA.Tg/MIN mice fed either the control diet (A–C) or the celecoxib-supplemented (1000 ppm) diet (D–F) and administered the vehicle alone (A and D), the non-CEA-based vaccine (B and E), or the CEA-based vaccine (C and F). Hematocrit levels were measured every 4–6 weeks, and the solid lines (—) represent individual mice. The dashed lines (---) identify a hematocrit level of 36, below which the mice are considered anemic.

Fig. 6.

Hematocrit levels from individual CEA.Tg/MIN mice fed either the control diet (A–C) or the celecoxib-supplemented (1000 ppm) diet (D–F) and administered the vehicle alone (A and D), the non-CEA-based vaccine (B and E), or the CEA-based vaccine (C and F). Hematocrit levels were measured every 4–6 weeks, and the solid lines (—) represent individual mice. The dashed lines (---) identify a hematocrit level of 36, below which the mice are considered anemic.

Close modal
Fig. 7.

Long-term survival of CEA.Tg/MIN mice fed either the control or celecoxib-supplemented (1000 ppm) diets ± the indicated vaccine. CEA.Tg/MIN mice fed the control diet were injected with the vehicle alone (n = 10, ----), the CEA-based vaccine (n = 11, ○), or the non-CEA-based vaccine (n = 6, ▵). Other groups of CEA.Tg/MIN mice were fed the celecoxib-supplemented diet (1000 ppm) and also injected with the vehicle alone (n = 10, ▪), the CEA-based vaccine (n = 12, •), or the non-CEA-based vaccine (n = 7, ▴). Vaccines were administered as outlined in “Materials and Methods.”

Fig. 7.

Long-term survival of CEA.Tg/MIN mice fed either the control or celecoxib-supplemented (1000 ppm) diets ± the indicated vaccine. CEA.Tg/MIN mice fed the control diet were injected with the vehicle alone (n = 10, ----), the CEA-based vaccine (n = 11, ○), or the non-CEA-based vaccine (n = 6, ▵). Other groups of CEA.Tg/MIN mice were fed the celecoxib-supplemented diet (1000 ppm) and also injected with the vehicle alone (n = 10, ▪), the CEA-based vaccine (n = 12, •), or the non-CEA-based vaccine (n = 7, ▴). Vaccines were administered as outlined in “Materials and Methods.”

Close modal
Table 1

Comparison of T, B, and NKa cellular responses in mice fed either the control or celecoxib-supplemented diets

SplenocyteParameterStimuliConcentrationControl dietCelecoxib-supplemented diets
+ 500 ppm+ 1000 ppm+ 1500 ppm
T cell Proliferation (cpm/72 h) Con A 1.00 μg/ml 201151 ± 11372 231104 ± 21575 206773 ± 17573 194860 ± 14752 
   0.5 μg/ml 172246 ± 9790 152383 ± 11904 149750 ± 11424 154431 ± 8789 
   0.25 μg/ml 71564 ± 6903 62962 ± 3304 58949 ± 6330 58005 ± 3408 
  Anti-CD3 31,250 T cells 277058 ± 8927 231113 ± 18109 281430 ± 13888 262538 ± 14464 
        
 IFN-γ production (ng/48 h) Con A 2.0 μg/ml 19.0 ± 2.0 17.0 ± 1.5 15.3 ± 2.2 16.2 ± 1.7 
   1.0 μg/ml 17.5 ± 1.8 10.7 ± 2.2b 7.7 ± 0.6b 6.4 ± 0.5b 
B cell Proliferation (cpm/72 h) LPS 5.00 μg/ml 12687 ± 344 11575 ± 1120 15976 ± 659c 11410 ± 752 
   1.25 μg/ml 6554 ± 555 6295 ± 618 8097 ± 692c 6436 ± 794 
   0.31 μg/ml 4487 ± 225 4396 ± 282 5691 ± 1249c 3993 ± 243 
        
NK cell YAC-1 lysis (%) E:T 200:1 14.3 ± 0.5 13.1 ± 0.6 17.4 ± 0.6 16.3 ± 0.5 
   100:1 8.6 ± 1.1 9.8 ± 1.2 9.8 ± 0.4 8.2 ± 0.5 
   50:1 9.3 ± 0.8 8.8 ± 0.3 8.6 ± 0.2 7.8 ± 0.2 
   25:1 5.9 ± 0.2 5.7 ± 0.3 4.5 ± 0.4 4.0 ± 0.3 
SplenocyteParameterStimuliConcentrationControl dietCelecoxib-supplemented diets
+ 500 ppm+ 1000 ppm+ 1500 ppm
T cell Proliferation (cpm/72 h) Con A 1.00 μg/ml 201151 ± 11372 231104 ± 21575 206773 ± 17573 194860 ± 14752 
   0.5 μg/ml 172246 ± 9790 152383 ± 11904 149750 ± 11424 154431 ± 8789 
   0.25 μg/ml 71564 ± 6903 62962 ± 3304 58949 ± 6330 58005 ± 3408 
  Anti-CD3 31,250 T cells 277058 ± 8927 231113 ± 18109 281430 ± 13888 262538 ± 14464 
        
 IFN-γ production (ng/48 h) Con A 2.0 μg/ml 19.0 ± 2.0 17.0 ± 1.5 15.3 ± 2.2 16.2 ± 1.7 
   1.0 μg/ml 17.5 ± 1.8 10.7 ± 2.2b 7.7 ± 0.6b 6.4 ± 0.5b 
B cell Proliferation (cpm/72 h) LPS 5.00 μg/ml 12687 ± 344 11575 ± 1120 15976 ± 659c 11410 ± 752 
   1.25 μg/ml 6554 ± 555 6295 ± 618 8097 ± 692c 6436 ± 794 
   0.31 μg/ml 4487 ± 225 4396 ± 282 5691 ± 1249c 3993 ± 243 
        
NK cell YAC-1 lysis (%) E:T 200:1 14.3 ± 0.5 13.1 ± 0.6 17.4 ± 0.6 16.3 ± 0.5 
   100:1 8.6 ± 1.1 9.8 ± 1.2 9.8 ± 0.4 8.2 ± 0.5 
   50:1 9.3 ± 0.8 8.8 ± 0.3 8.6 ± 0.2 7.8 ± 0.2 
   25:1 5.9 ± 0.2 5.7 ± 0.3 4.5 ± 0.4 4.0 ± 0.3 
a

NK, natural killer; Con A, concanavalin A; LPS, lipopolysaccharide.

b

P < 0.05 (versus IFN-γ produced by Con A-stimulated T cells from mice fed the control diet).

c

P < 0.05 (splenic B-cell proliferative response to LPS from mice fed the control diet and 500 or 1500 ppm celecoxib-supplemented diets).

Table 2

Average weight gain and hematocrit levels in CEA.Tg/MIN mice (150–160 days old) that received the CEA vaccine + celecoxib-supplemented diet

VaccineDiet# miceΔ body weightaHematocrit levels (%)
AverageRange
None Control 10 5.3 ± 0.6 28.2 ± 3.5 21.0–32.5 
CEA-based  15 8.5 ± 0.5b 41.0 ± 5.5b 34.5–50.0 
Non-CEA-based  5.6 ± 0.4 29.5 ± 2.2 25.5–34.0 
      
None Celecoxib 14 8.6 ± 0.8 45.0 ± 2.0b 38.5–49.5 
CEA-based  14 11.6 ± 0.8c 48.5 ± 3.5b 44.5–51.0 
Non-CEA-based  13 8.2 ± 0.4 46.5 ± 2.5b 47.5–52.0 
VaccineDiet# miceΔ body weightaHematocrit levels (%)
AverageRange
None Control 10 5.3 ± 0.6 28.2 ± 3.5 21.0–32.5 
CEA-based  15 8.5 ± 0.5b 41.0 ± 5.5b 34.5–50.0 
Non-CEA-based  5.6 ± 0.4 29.5 ± 2.2 25.5–34.0 
      
None Celecoxib 14 8.6 ± 0.8 45.0 ± 2.0b 38.5–49.5 
CEA-based  14 11.6 ± 0.8c 48.5 ± 3.5b 44.5–51.0 
Non-CEA-based  13 8.2 ± 0.4 46.5 ± 2.5b 47.5–52.0 
a

Maximum body weight gained; mean (g) ± SE.

b

P < 0.05 (versus CEA.Tg/MIN mice fed the control diet and administered the non-CEA-based vaccine or the vehicle alone).

c

P < 0.05 (versus CEA.Tg/MIN mice fed the celecoxib-supplemented diet combined with the vehicle alone or the non-CEA-based vaccine).

Table 3

Comparison of CEA-specific immune response in CEA.Tg/MIN mice fed either the control or celecoxib-supplemented diet and vaccinated with the CEA-based vaccine

Vaccine/DietMouse #ΔBWa (g)Hematocrit (%)CEA-specific immune responses
Serum IgG titersbCD4 proliferation (cpm)cCD8 peptide-pulsed cytolysis (%)dTumor burden
CEA/Control 6.7 43 3530 ± 225 2563 ± 230  19 
 6.3 41 2190 ± 195 5175 ± 92  15 
 7.1 45 3820 ± 305 3775 ± 112  14 
 Average 6.7 ± 0.2 43 ± 1 3180 ± 202 3837 ± 175 9.0 ± 1.1 16 ± 2 
CEA/Celecoxib 6.1 51 6070 ± 413 6970 ± 238  
 8.3 52 6700 ± 102 14050 ± 764  
 7.7 51 7520 ± 222 12220 ± 556  
 Average 7.4 ± 0.7 51 ± 1e 6763 ± 430e 11080 ± 612e 15.6 ± 2.3 0.3 ± 0.3e 
Vaccine/DietMouse #ΔBWa (g)Hematocrit (%)CEA-specific immune responses
Serum IgG titersbCD4 proliferation (cpm)cCD8 peptide-pulsed cytolysis (%)dTumor burden
CEA/Control 6.7 43 3530 ± 225 2563 ± 230  19 
 6.3 41 2190 ± 195 5175 ± 92  15 
 7.1 45 3820 ± 305 3775 ± 112  14 
 Average 6.7 ± 0.2 43 ± 1 3180 ± 202 3837 ± 175 9.0 ± 1.1 16 ± 2 
CEA/Celecoxib 6.1 51 6070 ± 413 6970 ± 238  
 8.3 52 6700 ± 102 14050 ± 764  
 7.7 51 7520 ± 222 12220 ± 556  
 Average 7.4 ± 0.7 51 ± 1e 6763 ± 430e 11080 ± 612e 15.6 ± 2.3 0.3 ± 0.3e 
a

BW, body weight.

b

Antibody titers presented as mean ± SE and calculated as the 1/serum dilution = A490nm of 0.5.

c

CD4-proliferative responses to soluble CEA (50 μg/ml) are presented as the mean ± SE as outlined in “Materials and Methods.”

d

Splenocytes from the 3 mice/group were pooled and grown for 1 week in the presence of the CEA526–533 (EAQNTTYL) peptide. Cytolytic activity was assessed as described in “Materials and Methods.” MC32A cells were pulsed with 1.0 μg/ml of either the CEA526–533 peptide or with a control peptide VSVN. Data are presented as the mean ± SE from triplicate well using an E:T ratio of 37.5:1.

e

P < 0.05 (versus CEA immune response from mice given the CEA-based vaccine and the control diet).

Table 4

Histopathology of CEA-expressing tissues in CEA.Tg/MIN mice administered the CEA-based vaccine and the celecoxib-supplemented diet

CEA.Tg/MIN Mouse
#1#2#3#4
Age (days) 231 188 231 184 
# Vaccinations 
Tumor burdenb 
Tongue Normal Normal Normal Normal 
Trachea Normal Normal Normal Normal 
Esophagus Normal Normal Normal Normal 
Stomach Normal Normal Normal Normal 
Intestine     
 Duodenum Hyperplasia, GALTa Hyperplasia, GALT Normal Normal 
 Prox. jejunum Normal Hyperplasia, GALT Intraepithelial neoplasia (3) Normal 
 Distal jejunum Normal Adenoma (8) Intraepithelial neoplasia (1) Normal 
 Ileum Normal Hyperplasia, GALT Normal Normal 
Caecum Normal Normal Normal Normal 
Colon Adenoma (1) Normal Normal Normal 
CEA.Tg/MIN Mouse
#1#2#3#4
Age (days) 231 188 231 184 
# Vaccinations 
Tumor burdenb 
Tongue Normal Normal Normal Normal 
Trachea Normal Normal Normal Normal 
Esophagus Normal Normal Normal Normal 
Stomach Normal Normal Normal Normal 
Intestine     
 Duodenum Hyperplasia, GALTa Hyperplasia, GALT Normal Normal 
 Prox. jejunum Normal Hyperplasia, GALT Intraepithelial neoplasia (3) Normal 
 Distal jejunum Normal Adenoma (8) Intraepithelial neoplasia (1) Normal 
 Ileum Normal Hyperplasia, GALT Normal Normal 
Caecum Normal Normal Normal Normal 
Colon Adenoma (1) Normal Normal Normal 
a

GALT, gut-associated lymphoid tissues (Peyer’s patches).

b

Tumor burden represents the total number of neoplasms (intraepithelial neoplasia or adenoma) found in the intestine and colon at time of necropsy.

Table 5

Histopathology of CEA-negative tissues in CEA.Tg/MIN mice

Other tissues that were found within normal histological limits included brain, thyroid, parathyroid, pituitary, urinary bladder, eye, bone (femur), spinal cord, vertebra, mammary gland, uterus, seminal vesicle, epididymis, testis, kidney, pancreas, thymus, and gall bladder. No hematopoietic neoplasms were found.

TissuePathologyControl dietCelecoxib diet
No vaccine (n = 3)CEA-based (n = 6)No vaccine (n = 3)Non-CEA-based (n = 4)CEA-based (n = 5)
Spleen Extramedullary hematopoiesis 3/3 6/6 3/3 4/4 2/5 
Liver Subacute inflammation 1/3 2/6 3/3 3/4 3/5 
 Extramedullary hematopoiesis 2/3 4/6 3/3 4/4 none 
Adrenals Lipogenic pigment 3/3 3/6 3/3 4/4 4/5 
Lung Lymphocytic infiltrate 3/3 1/6 2/3 2/4 4/5 
Heart Melanosis 2/3 2/3 2/3 3/4 1/5 
TissuePathologyControl dietCelecoxib diet
No vaccine (n = 3)CEA-based (n = 6)No vaccine (n = 3)Non-CEA-based (n = 4)CEA-based (n = 5)
Spleen Extramedullary hematopoiesis 3/3 6/6 3/3 4/4 2/5 
Liver Subacute inflammation 1/3 2/6 3/3 3/4 3/5 
 Extramedullary hematopoiesis 2/3 4/6 3/3 4/4 none 
Adrenals Lipogenic pigment 3/3 3/6 3/3 4/4 4/5 
Lung Lymphocytic infiltrate 3/3 1/6 2/3 2/4 4/5 
Heart Melanosis 2/3 2/3 2/3 3/4 1/5 

We thank Garland Davis and Donald Hill for excellent technical assistance. We also thank Debra Weingarten for editorial assistance on this manuscript.

1
Smith WL, Garavito RM, DeWitt DL. Prostaglandin endoperoxide H synthetase (cyclooxygenases)-1 and −2.
J Biol Chem
,
271
:
33157
-60,  
1996
.
2
Masferrer JL, Seibert K, Zweifel B, Needleman P. Endogenous glucocorticoids regulate an inducible cyclooxygenase enzyme.
Proc Natl Acad Sci USA
,
89
:
3917
-21,  
1992
.
3
Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Herschman HR. TIS10, a phorbal ester tumor promoter-inducible mRNA from Swiss 3T3 cells, encodes a novel prostaglandin synthetase/cyclooxygenase homologue.
J Biol Chem
,
266
:
12866
-72,  
1991
.
4
Souza RF, Shewmake K, Beer DG, Cryer B, Spechler SJ. Selective inhibition of cyclooxygenase-2 suppresses growth and induces apoptosis in human esophageal adenocarcinoma cells.
Cancer Res
,
60
:
5767
-72,  
2000
.
5
Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs.
Nat New Biol
,
231
:
232
-5,  
1971
.
6
Smalley WE, DuBois RN. Colorectal cancer and nonsteroidal anti-inflammatory drugs.
Adv Pharmacol
,
39
:
1
-20,  
1997
.
7
Sandler RS, Halabi S, Baron JA, et al A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer.
N Eng J Med
,
348
:
883
-90,  
2003
.
8
Baron JA, Cole BF, Sandler RS, et al randomized trial of aspirin to prevent colorectal adenomas.
N Eng J Med
,
348
:
891
-9,  
2003
.
9
Cruz-Correa M, Hylind LM, Romans KE, Booker SV, Giardiello FM. Long-term treatment with sulindac in familial adenomatous polyposis: a prospective cohort study.
Gastroenterology
,
122
:
641
-5,  
2002
.
10
Wolfe MM, Lichtenstein DR, Singh G. Gastrointestinal toxicity of nonsteroidal anti-inflammatory drugs.
N Eng J Med
,
340
:
1888
-99,  
1999
.
11
Koki AT, Leahy KM, Masferrer JL. Potential utility of COX-2 inhibitors in chemoprevention and chemotherapy.
Expert Opin Invest Drugs
,
8
:
1623
-38,  
1999
.
12
Fosslien E. Biochemistry of cyclooxygenase (COX)-2 inhibitors and molecular pathology of COX-2 in neoplasia.
Crit Rev Clin Lab Sci
,
37
:
431
-502,  
2000
.
13
Masferrer JL, Leahy KM, Koki AT, et al Antiangiogenesis and antitumor activities of cyclooxygenase-2 inhibitors.
Cancer Res
,
60
:
1306
-11,  
2000
.
14
Oshima M, Dinchuk JE, Kargman SL, et al Suppression of intestinal polyposis in Apc 716 knockout mice by inhibition of cyclooxygenase 2 (COX-2).
Cell
,
87
:
803
-9,  
1996
.
15
Reddy BS, Hirose Y, Lubet R, et al Chemoprevention of colon cancer by specific cyclooxygenase-2 inhibitor celecoxib, administered during different stages of carcinogenesis.
Cancer Res
,
60
:
293
-7,  
2000
.
16
Oshima M, Murai N, Kargman S, et al Chemoprevention of intestinal polyposis in the Apc716 mouse by rofecoxib, a specific cyclooxygenase-2 inhibitor.
Cancer Res
,
61
:
1733
-40,  
2001
.
17
Jacoby RF, Seibert K, Cole CE, Kelloff G, Lubet RA. The cyclooxygenase-2 inhibitor celecoxib is a preventative and therapeutic agent in the Min mouse model of adenomatous polyposis.
Cancer Res
,
60
:
5040
-4,  
2000
.
18
Steinbach G, Lynch PM, Phillips RK, et al The effect of celecoxib, a cyclooxygenase-2 inhibitor in familial adenomatous polyposis.
N Engl J Med
,
342
:
1946
-52,  
2000
.
19
Guadagni F, Roselli M, Cosimelli M, et al Quantitative analysis of CEA expression in colorectal adenocarcinoma and serum: Lack of correlation.
Int J Cancer
,
72
:
949
-54,  
1997
.
20
Gold P, Freedman SO. Demonstration of tumor-specific antigens in human colonic carcinomata by immunological tolerance and absorption techniques.
J Exp Med
,
121
:
439
-62,  
1965
.
21
Thompson JA, Grunert F, Zimmermann W. Carcinoembryonic antigen gene family: molecular biology and clinical perspectives.
J Lab Clin Anal
,
5
:
344
-66,  
1991
.
22
Eades-Perner A-M, van der Putten H, Hirth A, et al Mice transgenic for the human carcinoembryonic antigen gene maintain its spatiotemporal expression pattern.
Cancer Res
,
54
:
4169
-76,  
1994
.
23
Clarke P, Mann J, Simpson JF, Rickard-Dickson K, Primus FJ. Mice transgenic for human carcinoembryonic antigen as a model for immunotherapy.
Cancer Res
,
58
:
1469
-77,  
1998
.
24
Kass E, Schlom J, Thompson J, Guadagni F, Graziano P, Greiner J. Induction of protective host immunity to carcinoembryonic antigen (CEA), a self antigen in CEA transgenic mice, by immunizing with a recombinant vaccinia-CEA virus.
Cancer Res
,
59
:
676
-83,  
1999
.
25
Kass E, Panicali DL, Mazzara G, Schlom J, Greiner JW. Granulocyte/macrophage-colony stimulating factor produced by recombinant avian poxviruses enriches the regional lymph nodes with antigen-presenting cells and acts as an immunoadjuvant.
Cancer Res
,
61
:
206
-14,  
2001
.
26
Mizobata S, Tompkins K, Simpson JF, Shyr Y, Primus FP. Induction of cytotoxic T cells and their antitumor activity in mice transgenic for carcinoembryonic antigen.
Cancer Immunol Immunother
,
49
:
285
-95,  
2000
.
27
Niethammer AG, Primus FJ, Xiang R, et al An oral DNA vaccine against human carcinoembryonic antigen (CEA) prevents growth and dissemination of Lewis lung carcinoma in CEA transgenic mice.
Vaccine
,
20
:
421
-9,  
2001
.
28
Xiang R, Primus FJ, Ruehlmann JM, et al A dual-function DNA vaccine encoding carcinoembryonic antigen and CD40 ligand trimer induces T cell-mediated protective immunity against colon cancer in carcinoembryonic antigen-transgenic mice.
J Immunol
,
167
:
4560
-5,  
2001
.
29
Greiner JW, Zeytin H, Anver MR, Schlom J. Vaccine-based therapy directed against carcinoembryonic antigen (CEA) demonstrates antitumor activity on spontaneous intestinal tumors in the absence of autoimmunity.
Cancer Res
,
62
:
6944
-52,  
2002
.
30
Marshall JL, Hawkins MJ, Tsang KY, et al Phase I study in cancer patients of a replication-defective avipox recombinant vaccine that expresses human carcinoembryonic antigen.
J Clin Oncol
,
17
:
332
-7,  
1999
.
31
von Mehren M, Arlen P, Tsang KY, et al Pilot study of a dual gene recombinant avipox vaccine containing both carcinoembryonic antigen (CEA) and B7.1 transgenes in patients with recurrent CEA-expressing adenocarcinomas.
Clin Cancer Res
,
6
:
2219
-28,  
2000
.
32
Marshall JL, Hoyer RJ, Toomey MA, et al Phase I study in cancer patients of a diversified prime and boost vaccination protocol using recombinant vaccinia virus and recombinant nonreplicating avipox virus to elicit anti-carcinoembryonic antigen immune responses.
J Clin Oncol
,
18
:
3964
-73,  
2000
.
33
Iniguez MA, Punzon C, Fresno M. Induction of cyclooxygenase-2 on activated T lymphocytes: Regulation of T cell activation by cyclooxygenase-2 inhibitors.
J Immunol
,
163
:
111
-9,  
1999
.
34
Rivero M, Santiago B, Galindo M, Brehmer MT, Pablos JL. Cyclooxygenase-2 inhibition lacks immunomodulatory effects on T cells.
Clin. and Exp Rheumatol
,
20
:
379
-85,  
2002
.
35
Langenbach R, Loftin C, Lee C, Tiano H. Cyclooxygenase knockout mice. Models for elucidating isoform-specific functions.
Biochem Pharmacol
,
58
:
1237
-46,  
1999
.
36
Hodge JW, Sabzevari H, Yafal AG, Gritz L, Lorenz MG, Schlom J. A triad of costimulatory molecules synergize to amplify T-cell activation.
Cancer Res
,
59
:
5800
-7,  
1999
.
37
Grosenbach DW, Barrientos JC, Schlom J, Hodge JW. Synergy of vaccine strategies to amplify antigen-specific immune responses and antitumor effects.
Cancer Res
,
61
:
4497
-505,  
2001
.
38
Muraro R, Wunderlich D, Thor A, et al Definition of monoclonal antibodies of a repertoire of epitopes on carcinoembryonic antigen differentially expressed in human colon carcinoma versus normal adult tissues.
Cancer Res
,
45
:
5769
-80,  
1985
.
39
Schmitz J, Reali E, Hodge JW, et al Identification of an interferon–inducible carcinoembryonic antigen (CEA) CD8+ T-cell epitope which mediates tumor killing in CEA transgenic mice.
Cancer Res
,
62
:
5058
-64,  
2002
.
40
Overwijk WW, Surman DR, Tsung K, Restifo NP. Identification of a Kb-restricted CTL epitope of beta-galactosidase: potential use in development of immunization protocols for “self” antigens.
Methods: A Comparison to Methods in Enzymol
,
12
:
117
-23,  
1997
.
41
Flynn KJ, Belz GT, Altman JD, Ahmed R, Woodland DL, Doherty PD. Virus-specific CD8+ T cells in primary and secondary influenza pneumonia.
Immunity
,
8
:
683
-91,  
1998
.
42
Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse.
Science
,
247
:
322
-4,  
1990
.
43
Su L-K, Kinzler KW, Vogelstein B, et al Multiple intestinal neoplasia caused by a mutation in the murine homologue of the APC gene.
Science
,
256
:
668
-70,  
1992
.
44
Goodwin SD, Glenny RW. Nonsteroidal anti-inflammatory drug-associated pulmonary infiltrates with eosinophilia. Review of the literature and Food and Drug Administration Adverse Drug Reaction reports.
Arch Intern Med
,
152
:
1521
-4,  
1992
.
45
Takahashi Y, Roman C, Chemtob S, Tse MM, Lin E, Heymann MA, Clyman RI. Cyclooxygenase-2 inhibitors constrict the fetal lamb ductus arteriosus both in vitro and in vivo.
Am J Physiol Regulatory Integrative Comp Physiol
,
278
:
R1496
-505,  
2000
.
46
Iniguez MA, Matinez-Martinez S, Punzon C, Redondo JM, Fresno M. An essential role of the nuclear factor of activated T cells in the regulation of the expression of the cyclooxygenase-2 gene in human T lymphocytes.
J Biol Chem
,
275
:
23627
-35,  
2000
.
47
Carey MA, Germolec DR, Bradbury A, et al Accentuated T helper type 2 airway response after allergen challenge in cyclooxygenase-1 −/− but not cyclooxygenase-2 −/− mice.
Am J Respir Crit Care Med
,
167
:
1509
-15,  
2003
.
48
Sonoshita M, Takaku K, Sasaki N, et al Acceleration of intestinal polyposis through prostaglandin receptor EP2 in Apc(Delta 716) knockout mice.
Nat Med
,
7
:
1048
-51,  
2001
.
49
Hsu AL, Ching T-T, Wang DS, Song X, Rangnekar VM, Chen CS. The cyclooxygenase-2 inhibitor celecoxib induces apoptosis by blocking Akt activation in human prostate cancer cells independently of Bcl-2.
J Biol Chem
,
275
:
11397
-403,  
2000
.
50
Stolina M, Sharma S, Lin Y, et al Specific inhibition of cyclooxygenase 2 restores the balance of IL-10 and IL-12 synthesis.
J Immunol
,
164
:
361
-70,  
2000
.
51
Sharma S, Stolina M, Yang SC, et al Tumor cyclooxygenase 2-dependent suppression of dendritic cell function.
Clin Cancer Res
,
9
:
961
-8,  
2003
.
52
Paulsen JE, Namork E, Steffensen I-L, Eide TJ, Alexander J. Identification and quantification of aberrant crypt foci in the colon of Min mice - A murine model of familial adenomatous polyposis.
Scand J Gastroenterol
,
35
:
534
-9,  
2000
.
53
Takeda H, Sonoshita M, Oshima H, et al Cooperation of cyclooxygenase 1 and cyclooxygenase 2 in intestinal polyposis.
Cancer Res
,
63
:
4872
-7,  
2003
.
54
Finke J, Ferrone S, Frey A, Mufson A, Ocha A. Where have all the T cells gone? Mechanisms of immune evasion by tumor.
Immunol Today
,
20
:
158
-60,  
1999
.
55
Hodge JW, Grosenbach DW, Aarts WM, Poole DJ, Schlom J. Vaccine therapy of established tumors in the absence of autoimmunity.
Clin Cancer Res
,
9
:
1837
-49,  
2003
.
56
Gilboa E. The risk of autoimmunity associated with tumor immunotherapy.
Nature Immun
,
2
:
789
-92,  
2001
.
57
DeLong P, Tanaka T, Kruklitis R, et al Use of cyclooxygenase-2 inhibition to enhance the efficacy of immunotherapy.
Cancer Res
,
63
:
7845
-52,  
2003
.
58
Sheehan KM, Sheahan K, O’Donoghue DP, et al The relationship between cyclooxygenase-2 expression and colorectal cancer.
J Am Med Assoc
,
282
:
1254
-7,  
1999
.
59
Saukkonen K, Nieminen O, van Rees B, et al Expression of cyclooxygenase-2 in dysplasia of the stomach and in intestinal-type gastric adenocarcinoma.
Clin Cancer Res
,
7
:
1923
-31,  
2001
.
60
Tucker ON, Dannenberg AJ, Yang EK, et al Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer.
Cancer Res
,
59
:
987
-90,  
1999
.
61
Achiwa H, Yatabe Y, Hida T, et al Prognostic significance of elevated cyclooxygenase 2 expression in primary, resected lung carcinoma.
Clin Cancer Res
,
5
:
1001
-5,  
1999
.
62
Kulkarni S, Rader JS, Zhang F, et al Cyclooxygenase-2 is overexpressed in human cervical cancer.
Clin Cancer Res
,
7
:
429
-34,  
2001
.
63
Chan G, Boyle JO, Yang EK, et al Cyclooxygesase-2 expression is up-regulated in squamous cell carcinoma of the head and neck.
Cancer Res
,
59
:
991
-4,  
1999
.