International Immunopharmacology 1 Ž2001. 1651–1667 www.elsevier.comrlocaterintimp
Review
Inflammation, carcinogenesis and cancer F.A. Fitzpatrick Huntsman Cancer Institute, 2000 Circle of Hope, UniÕersity of Utah, Salt Lake City, UT 84112-5550, USA Received 9 May 2001; accepted 17 May 2001
Abstract To fulfill their role in host-defense, granulocytes secrete chemically reactive oxidants, radicals, and electrophilic mediators. While this is an effective way to eradicate pathogenic microbes or parasites, it inevitably exposes epithelium and connective tissue to certain endogenous genotoxic agents. In ordinary circumstances, cells have adequate mechanisms to reduce the genotoxic burden imposed by these agents to a negligible level. However, inflammation persisting for a decade eventually elevates the risk of cancer sufficiently that it is discernible in case control epidemiological studies. Advances in our understanding of tumor suppressors and inflammatory mediators offer an opportunity to assess the molecular and cellular models used to guide laboratory investigations of this phenomenon. Disappointing results from recent clinical trials with anti-oxidant interventions raise questions about the risks from specific endogenous agents such as hydrogen peroxide and oxy radicals. Simultaneously, the results from the anti-oxidant trials draw attention to an alternate hypothesis, favoring epigenetic inactivation of key tumor suppressors, such as p53, and the consequent liability this places on genomic integrity. q 2001 Elsevier Science B.V. All rights reserved. Keywords: Inflammation; Carcinogenesis; Cancer
1. Introduction It is timely to review the association between persistent inflammation, carcinogenesis and cancer. Section 2 discusses the epidemiological data supporting such an association. Section 3 depicts a conventional molecular and cellular model for the increased
AbbreÕiations: COX, cyclooxygenase; NSAID, non-steroidal anti-inflammatory drug; PG, prostaglandin E-mail address:
[email protected] ŽF.A. Fitzpatrick..
risk of cancer, secondary to persistent inflammation. Section 3 also discusses the utility and limitations of this current model. For prominent electrophilic mediators of inflammation Žhydrogen peroxide and oxy radicals, nitric oxide, malondialdehyde, 4-hydroxynonenal, and eicosanoids., previously overlooked epigenetic mechanisms may contribute greatly to the disruption of genomic integrity. Section 4 discusses the role of cyclooxygenase ŽCOX. isoenzymes in cancer, distinct from their role in inflammation. Section 4 also discusses the benefits and limitations of non-steroidal anti-inflammatory drugs ŽNSAIDs. in cancer prevention, and the case for assessing alternative anti-inflammatory agents for their benefitsrrisks in cancer prevention.
1567-5769r01r$ - see front matter q 2001 Elsevier Science B.V. All rights reserved. PII: S 1 5 6 7 - 5 7 6 9 Ž 0 1 . 0 0 1 0 2 - 3
1652
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
2. Persistent inflammation and cancer risk: epidemiological evidence 2.1. UlceratiÕe colitis and colon cancer Inflammation, occurring episodically but persistently over a decade, is a risk factor for several types of cancer ŽTable 1.. Ulcerative colitis typifies this premise w1–7x. In a large study, involving over 3000 patients with chronic ulcerative colitis, investigators observed an incidence of colon cancer 5.7-fold higher than expected w2x. This increased risk of cancer correlated with Ži. the severity of disease, ranging from 1.7-fold for patients with ulcerative proctitis to 14.8-fold for patients with total colonic inflammation Žpancolitis.; and Žii. the duration of the disease. Colitis persisting for 35–40 years carries an absolute risk of colon cancer ranging from 30% to 40% w2,4x. Additionally, colon cancer associated with ulcerative colitis has the worst prognosis and lowest 5-year survival rate Ž- 40%., among three high-risk groups, including familial adenomatous polyposis, and hereditary non-polyposis colorectal cancer w6x. Importantly, ulcerative colitis does not increase the risk of other types of cancer w7x, consistent with the hypothesis that neoplasia results in part from persistent, local inflammation ŽFig. 1.. Microsatellite instability occurs in the mucosa of 50% of patients who have no dysplasia, suggesting that cumulative disruption of genomic integrity accompanies inflammation and precedes transformation w8x.
Management of colitis with the anti-inflammatory agent sulfasalazine can reduce the incidence of colon cancer w9,10x. The anti-inflammatory mechanism Žsic. of sulfasalazine is either uncertain, non-specific, pleiotropic or all three depending on one’s preference. As expected in an inflammatory environment, COX-2 is highly expressed in ulcerative colitis w11,12x. However, selective COX-2 inhibitors exert little or no anti-inflammatory effect in animal models of inflammatory bowel disease. It is debatable whether they will confer any anti-neoplastic benefit in humans w12–14x based on their suppression of colitis. 2.2. Lung inflammation and lung cancer Localized, chronic inflammatory disorders appear to increase the risk for cancer in several other organs, including the lung, pancreas, esophagus, and skin. Analysis of cancer incidence in asthmatic patients in five separate epidemiological studies is intriguing. A case-control study, involving 78,000 patients with asthma, showed significant excess risk of lung cancer in both men and women, consistent with a contribution from local, chronic inflammation w15x. A second study on 31,000 patients reported a significantly increased risk of death from lung cancer in men with asthma, after adjustment for cigarette smoking habits Žhazard ratio 3.19. w16x. A third study on 64,000 patients with asthma reported a reduced risk of most cancer types with the exception of lung
Table 1 Association of inflammation with cancer Inflammatory condition
Oncogenic consequences
References
Ulcerative colitis) 8 years Asthma
≠ Risk of colon cancer ≠ Risk of lung cancer x Risk of several cancers, except lung cancer ≠ x Uncertain association with risk of ovarian cancer ≠ Risk of ovarian cancer ≠ Risk of bladder cancer ≠ Risk of pancreatic cancer Association with penile cancer ≠ Risk of dysplasia ≠ Risk of lung, liver, skin cancer ≠ Risk of verrucous carcinoma
w1–10x w15–19x
Pelvic inflammatory disease Ovarian epithelial inflammation Eosinophilic cystitis Pancreatitis Foreskin inflammation and phimosis Barret’s esophagous Sarcoidosis Ulcerative lichen planus Žinflammatory mucocutaneous disorder.
w36,37x w31–33,35x w26,27x w38–41,43x w28x w44,45x w25x w29,30x
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
Fig. 1. Localized inflammation correlates with risk for localized cancer. Ulcerative colitis.
cancer and endocrine cancer w17x, suggesting that asthma confers a protective effect on certain organs, but not the respiratory tract. Finally, two population based, case-control studies involving lifetime nonsmoking women participants both affirmed that asthma elevates the risk for lung cancer w18,19x. Can one reconcile a AprotectiveB effect of asthma against cancer in certain organs with its detrimental effect on the lung? Yes, if one accepts that asthmatics abstain or smoke infrequently w20x. In this context, the elevated risk of lung cancer observed in asthmatic patients is even more striking. Local inflammation then becomes one plausible source of the heightened risk of lung cancer, considering that asthmatic patients often endured persistent lung inflammation prior to the introduction of treatment guidelines recognizing asthma as an inflammatory disorder of the airway w21–23x. It is important to acknowledge an alternative hypothesis that inhaled corticoids used in asthma management might create a localized immune deficiency in the lung, and this local immune deficiency increases the risk of cancer. However, this latter hypothesis is not fully consistent with models of particle-induced lung cancer which favor inflammation as an endogenous source of mutations w24x. 2.3. Inflammation and cancers of the oÕary, pancreas, bladder, skin and esophagus Other representative associations between inflammation, carcinogenesis, and cancer include lung and
1653
liver cancer following sarcoidosis w25x; bladder cancer in humans with eosinophilic or irritant-induced cystitis w26,27x, skin cancer w28x, verrucous carcinoma w29,30x ovarian cancer w31–36x and Barret’s esophagus w44,45x. The relationship between the number of lifetime ovarian cycles and the risk for ovarian cancer is partly endocrinological, but the granulocytic inflammation that accompanies ovulation may also contribute w31,32x. The number of ovarian cycles correlates quantitatively with expression of a mutated p53 tumor suppressor in ovarian cancer w33x and it is notable that patients with chronic endometriosis w34x or pelvic inflammatory disease may also have a significantly elevated risk of ovarian cancer w36x. The latter association is debatable since larger case control studies fail to confirm any association between pelvic inflammation and ovarian cancer w37x. Case-controlled studies indicate that chronic pancreatitis is a risk factor for pancreatic cancer w38–40x and mutated alleles of p53 occur in resected, morphologically normal tissue from 10% of patients with chronic pancreatitis w41x, consistent with the molecular genetics of pancreatic cancer progression w42x. However, one population-based study suggests that the risk of pancreatic cancer for patients with chronic pancreatitis declines with time, in contrast to associations between inflammation and cancer in other organ systems w43x. The difficulty of early detection, the rapid course, and the high morbidity of this oncological disorder complicate epidemiological experiments seeking a relationship between pancreatitis and pancreatic cancer. 2.4. Effect of non-steroidal anti-inflammatory drugs on cancer risk With rare exception w49x, over 20 separate investigations during the past 12 years affirm that regular consumption of aspirin or NSAIDs lowers the incidence, or mortality, of colon cancer w50–76x. However, one should interpret these results prudently. The results do support the notion that cyclooxygenase enzyme, a source of eicosanoid mediators, is a participant in the molecular progression of colon cancer w77–82x. However, NSAIDs are not front line therapy for patients with inflammatory bowel disease. In fact, some investigations suggest that their
1654
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
use is contra-indicated because they aggravate inflammatory bowel disease w14,83x; other investigations are inconclusive w84x. Thus, the beneficial effects of NSAIDs in colon cancer do not pertain directly to the risk associated with or due to persistent colitis. We discuss the benefits of NSAIDs and the association between cyclooxygenase and cancer in more detail in Section 4 of this review.
3. Molecular and cellular determinants of inflammation-associated cancer 3.1. The conÕentional model The epidemiological data in Section 2 strongly support the conclusion that persistent inflammation raises the risk of cancer within the involved organ, but not distal organs. How does this happen? As part of their normal host-defense function, granulocytes secrete chemically reactive radicals and electrophilic mediators ŽFig. 2.. Thus, inflammation inevitably exposes proximal epithelial and stromal cells to substances with mutagenic potential in vitro w85–89x. Note that activated neutrophils can transform cell lines to variants with neoplastic traits w87,89x. Granulocytes and lymphocytes generate at least four separate types of products that are genotoxic or mutagenic: Ži. hydrogen peroxide and oxy radicals w90–100x; Žii. nitric oxide w101–109x; Žiii. malondialdehyde w110–118x; and Živ. 4-hydroxy-2nonenal w119–122x. The latter two are prominent by-products of lipid peroxidation w123x. Oxy radicals derived from H 2 O 2 can act directly, causing DNA
strand breaks that introduce oncogenic mutations w123,124x, or indirectly by modulating gene transcription w125,126x, and suppressing genomic repair pathways w127,128x. Table 2 summarizes an appreciable collection of data suggesting that oxygen and nitrogen radicals, acting as endogenous mutagens, account for some of the elevated risk of cancer associated with persistent inflammation. In view of this abundant evidence, it might surprise readers to learn that the radical scavenging anti-oxidant, bcarotene, does not reduce the risk of cancer in apparently healthy subjects w129–133x. In high risk populations, it may actually have increased the risk w132x. Physicians’ Health Study II is currently testing vitamin E, in combination with vitamin A and vitamin C, as anti-oxidants w134x. Nevertheless, the failure of b-carotene to confer any benefit in several large, prospective clinical trials should prompt debate about the underlying hypothesis, as well as the trial design. Accordingly, Collins w129x has provided a thoughtful critique of the hypothesis that oxidative DNA damage is a significant risk factor for cancer. His analysis of the literature shows that steady-state estimates of 8-oxo-deoxyguanosine Ž8-oxo-dG. in DNA have declined steadily as analytical methodologies improved. Current levels of 8-oxo-dG composition are sufficiently small to suggest that constitutive antioxidant defenses Žreduced glutathione, catalase, superoxide dismutase, GSH peroxidase. and DNA repair processes can minimize the risk of cancer from endogenous oxidants. If so, what molecules other than oxy radicals might account for the elevated risk of cancer during inflammation? How might they create risks?
Fig. 2. Model for the cellular and molecular basis of inflammation-related cancer risk.
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
1655
Table 2 Events associated with disruption of genomic integrity
H 2 O2 Ø Nitric oxide ŽNO . malondialdehyde 4-OH-2-nonenal Eicosanoids
Modification of nucleosides and nucleotides
Modification or damage of DNA
w91,95x
w91–95x w101–106x w112,113x w120,121x w136x
w110,111x w119,120x w135x
Collins suggests that alkylation of DNA poses a more serious risk to genomic integrity than does 8-oxo-dG. In this context, malondialdehyde, 4-hydroxy-nonenal and certain electrophilic eicsoanoids warrant attention. We propose that these agents increase cancer risks from two complementary processes: Ži. direct reaction with nucleophiles on DNA; and Žii. impairment of genomic sentinels, typified by the p53 transcription factor. So far, investigators have neglected the latter process as a risk. 3.2. Epigenetic inactiÕation of tumor suppressor p53: sentinel of genomic integrity Investigations on chemical carcinogenesis often proceed via the following experimental sequence. First, one demonstrates that a chemical reaction between a mutagen and DNA can occur under favorable conditions in vitro. Then one develops ultra-sensitive analysis methods for determining if it does occur in vitro or in vivo. Next, one applies these methods to determine if any occurrence in vivo has oncological significance. This approach is systematic and well-aligned with the hypothesis that loss of genomic integrity causes cancer in a two phase process involving induction and promotion w139x. However, in practice, it focuses primarily on direct chemical modification of DNA as the problem and it neglects epigenetic mechanisms. Furthermore, it positions the investigation of pathological significance at the end of the process. It is possible to consider epigenetic, as well as genetic processes, and to place the pathological significance at the beginning of the investigation by adapting a different approach. This approach relies on the premise that any endogenous agents that disrupt genomic integrity should activate
p53 alteration
w108,109x
w137x
Mutation or transformation of cells w96–100x w101,102,107x w114–118x w122x w138x
a response by the p53 tumor suppressor. Results with this approach have been informative and surprising w108,137x. Cells harboring a wild-type p53 tumor suppressor gene normally maintain a low steady-state concentration of the p53 protein via its post-translational regulation w140–145x. When genomic damage occurs, cells slow their degradation of p53, and accumulate it as a nuclear homo-tetramer to transactivate genes that arrest the cell cycle Žp21WA F1rCIP1 .; repair DNA damage ŽGADD 45.; autoregulate its degradation ŽMDM-2.; or propagate apoptosis ŽBAX.. These responses enable p53 to function as a genomic sentinel w146–149x. Impairment of the p53 gene or protein has grave cellular and medical consequences w150–153x. This impairment can occur by two prominent mechanisms: Ži. mutation or loss of a p53 allele w154–156x; Žii. binding of ubiquitin ligase oncoproteins Že.g. mdm-2, E6. to p53 w157–161x. Estimates suggest that 40–60% of tumors harbor an allelic variant of p53. The vast collection of p53 nucleotide polymorphisms collapses into two classes of mutant p53 proteins. AContactB mutants Žclass I. retain a wildtype conformation but they have altered residues in direct contact with DNA Žamino acid residues 112– 141, 236–251 and 271–286.. AConformationalB mutants Žclass II. have alterations in residues that maintain p53 conformational integrity Žamino acid residues 163–195.. AContactB and AconformationalB mutations each disrupt the p53:mdm-2 auto-regulatory cycle, allowing cells to accumulate appreciable levels of mutant p53 protein Žp53 mut . under basal condition w153,159x. A high basal level of p53 mut and a failure to accumulate more p53 mut in response to genomic damage are distinctive traits of cells harbor-
1656
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
ing a mutant p53 gene. In such cells the basal degradation rate of p53 mut is much slower than normal ŽFig. 3.. In certain tumors, viral oncoproteins Že.g. human papilloma virus E6. or mdm-2 impair p53 by binding directly to it; facilitating its conjugation with ubiquitin, and hastening its degradation by the 26S proteasome w144,145x. Very low basal levels of p53 and failure to accumulate p53 in response to genomic damage are distinctive traits of cells harboring oncoproteins with ubiquitin ligase activity. In such cells, the basal degradation rate of p53 is much faster than normal ŽFig. 3.. Distinct from somatic mutation or sequestration by ubiquitin ligases, p53 is redox sensitive. Oxidizing agents Ždiamide, pyrrolidine dithiocarbamate, Ø H 2 O 2 . and nitric oxide ŽNO . donors ŽSNAP or NAP. disrupt the conformation and sequence-specific DNA binding of isolated p53 w108,162,164x. Notably, this requires an appreciable excess of oxidizing agent over isolated p53 w108,162,164x and the redox sensitivities of isolated p53 versus cellular p53 do not always correlate well w165x. Some investigators report that H 2 O 2 impairs DNA binding and transcription by both isolated and cellular p53 w164x; others report that H 2 O 2 activates cellular p53 transcription w166x and nuclear translocation w167,168x. Recently, Wu and Momand w168x and we w137x established that intact cells exposed to 25 m M dithiocarbamate; or to 20 m M prostaglandin ŽPG. A 1 and
PGA 2 accumulate p53 protein that is conformationally, and functionally deranged. These results suggest that distinctive conditions, separable from somatic mutation or association with ubiquitin ligases, can impair cellular p53 ŽFig. 4.. We stress that our model should also apply to other electrophiles. For example, paradoxical effects of benzo w g xchrysene 11,12-dihydrodiol 13,14 epoxide on p53 and p21 expression in MCF-7 cells fit into the framework we propose w163x. Thus, exposure of cells to various endogenous or xenobiotic electrophiles may inactivate p53 by a mechanism analogous to the one described here. Sporadic, or episodic, impairment of p53 via redox or alkylating agents might favor the emergence of cells with a mutator phenotype w169,170x, inaugurating carcinogenesis. This integrated hypothesis offers a framework to investigate why persistent inflammation increases the risk for cancer localized to the site of inflammation. Inflammation bathes epithelial and mesenchymal tissue with exudate containing Ø NO and PGE 2 which dehydrates readily into PGA 2 w171x. Inflammation does not necessarily expose cells Ø to sufficient NO or PGA 2 , individually, to duplicate the results reported by Calmels et al. w108x and Moos et al. w137x. However, inflammatory exudate contains a blend of electrophiles typified by a , b unsaturated aldehydes derived from eicosanoid biosynthesis or lipid peroxidation Žmalondialdehyde, 4-OH-nonenal. w123x; a , b unsaturated ketones derived from
Fig. 3. Dysfunctional p53: response to genomic damage.
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
1657
Fig. 4. Epigenetic inactivation of p53 by inflammatory mediators: response to genomic damage.
eicosanoid metabolism Ž15-keto-PGF2 a , 15-KetoPGE 2 , 5-, 12-, and 15-oxo-eicosatetraenoic acid.; and a , b unsaturated ketones derived from albumin dehydrating PGD 2 Ž D12-PGJ2 and 15-deoxy-D12PGJ2 . w172x. Thus, inflammation might expose cells to a mixture of electrophiles in quantities sufficient to impair transactivation by p53. Accordingly, the link between inflammation and cancer will be clarified by discerning the structure–activity relationships and mechanisms that enable electrophilic mediators of inflammation to impair transactivation, conformation and phosphorylation of p53 and other redox sensitive transcription factors involved in genomic surveillance and repair. 3.3. Mechanistic and biological implications Knudson’s hypothesis asserts that tumor formation requires the mutation or deletion of both copies of a tumor suppressor gene w46x. Tumor suppressor p53 conforms to this hypothesis in most cases. For instance, a point mutation in one p53 allele, paired with a loss-of-heterozygosity in the other p53 allele are abundant in tumors. To accommodate Knudsen’s
hypothesis in subsets of tumors with an intact p53 allele paired with a mutant allele, Fearon and Vogelstein w47x proposed the Adominant negativeB effect—most experimental data support this proposal. However, a Adominant negativeB effect does not accommodate Knudsen’s hypothesis with tumors harboring a wild-type allele paired with deleted allele, nor does it explain the tumor incidence and survival rates among p53 Žyry ., p53 Žqry . and p53 Žqrq . mice w48x. Our results may provide a way to accommodate Knudsen’s hypothesis with the special case of p53 Žqry . tissue. Loss of heterozygosity represents one of the AhitsB and sporadic inactivation of the wild-type p53, by the process described by Calmels et al. w108x or Moos et al. w137x, represents the AsecondB hit. Instead of a gene dosage effect, we speculate that the incidence of tumorigenesis is a function of the frequency and duration of the epigenetic impairment of p53 by endogenous or exogenous electrophiles. In other words, if electrophiles impair p53 in cells heterozygous for p53, it would correspond essentially to the Asecond hitB of Knudsen’s hypothesis and it would offer a mechanism other than the dominant negative activity of the mutant to account for oncogenesis.
1658
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
4. Cyclooxygenase (COX) isoenzymes and cancer 4.1. COX-2 expression and function in cancer Numerous investigations have established the medical importance of the COX-1 and COX-2 isoenzymes w173–184x. Experimental and clinical data fit the notion that disease-restricted expression of the COX-2 gene helps to govern their respective roles in physiological and pathological processes w185x. Restricted expression of COX-2 in neoplasms can occur, distinct from inflammation. Inflammation is neither an intermediate step nor a risk factor for inherited syndromes like Familial Adenomatous Polyposis. Furthermore, several types of neoplastic and pre-neoplastic tissue overexpress COX-2, compared to adjacent non-involved tissue ŽTable 3.. In some of these tissues, COX-2 expression appears causal, not incidental, and its expression is not associated with any inflammatory process. The Min mouse model of intestinal polyposis, in particular, shows that expression of COX-2 facilitates progression to neoplasia w186–189x. For instance, homologous disruption of the COX-2 gene diminishes polyp formation in Min mice by ; 80% w188, 189x. In colon cancer, or related animal models such as the Min mouse, COX-2 exerts an oncogenic effect by modifying, i.e. aggravating, the pathological consequences of mutations in the adenomatous polyposis coli Ž APC . tumor suppressor gene. Once COX-2 protein accumulates, following induction or stabiliza-
tion of COX-2 mRNA, it catalyzes the formation of prostaglandins and reactive by-products that may accelerate the carcinogenesis process. COX-2 catalysis can aggravate carcinogenesis via at least three mechanisms, each of which has some experimental support. First, accumulation of arachidonic acid favors apoptosis, and depletion of arachidonic acid favors cells survival w190–193x. Accordingly, COX-2 catalysis depletes an apoptotic signal by lowering the intracellular level of free arachidonic acid. The second, more conventional mechanism, is that prostaglandins Že.g. PGE 2 . formed by COX-2 catalysis modulate cellular processes that govern cell growth and differentiation. For instance, PGE 2 inhibits apoptosis by inducing the Bcl-2 protooncogene w194x and enhances angiogenesis and adhesion by modulating integrin expression w195,196x. PGE 2 ordinarily elevates intracellular cyclic AMP, which suppresses apoptosis in the gastrointestinal tract w197x. Accordingly, COX-2 catalysis generates products with several biochemical effects converging at suppression of apoptosis. A third mechanism involves the effects of malondialdehyde, an endogenous mutagen w110– 116x, generated coordinately with the PG endoperoxides, PGG2 and PGH 2 . Exogenously applied malondialdehyde is not carcinogenic in SENCAR mice w117x; however, this result is consistent with the fact that it exists predominately as an anion that cannot traverse the plasma membrane very readily w123x. This trait makes it difficult to compare the consequences of endogenous production of malondialde-
Table 3 Disease-restricted expression of COX-2 in oncology Organ system
Observation
Incidence
Colon carcinoma w219x Colon adenocarcinoma w220x Breast tumor w220x Lung tumor w220x Gastric tumor w221x Head and neck squamous cell carcinoma ŽHNSCC. w222x Pancreatic cancer w223x
COX-2 mRNA at site of pathologly) the non-involved site COX-2 protein at site of pathology) non-involved site COX-2 protein at site of pathology) non-involved site COX-2 protein at site of pathology) non-involved site COX-2 protein at site of pathology) non-involved site Mean COX-2 mRNA in HNSCC 150-fold) normal volunteers
5r5 subjects 5r7 subjects 11r20 subjects 18r20 subjects 73r104 subjects
Mean COX-2 mRNA in pancreatic cancer 60-fold) adjacent non-involved tissue. COX-2 protein at site of pathology) non-involved site COX-2 levels ) in tumors with larger sizes and deeper invasion
9r10
Colon carcinoma w224x Cervical cancer w225x
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
hyde at a porous nuclear membrane with the consequences of its exogenous administration exterior to the plasma membrane barrier w117,118x. Presumably, overexpression of COX-2 derives from somatic mutations that persistently activate signaling pathways Že.g. ras, various growth factor receptor tyrosine kinase pathways. w198,199x or from factors that regulate COX-2 mRNA stability w200,201x in some neoplastic tissue. As one might expect, COX-2 overexpression is not observed in all tumors. For instance, in 13–14% of colorectal adenomas, the methylation of CpG islands near exon 1 silences COX-2 expression w202x. This implies that adenomas lacking a functional COX-2 gene, due to its epigenetic silencing, should not respond to treatment with COX-2 specific inhibitors. Likewise, according to Vogelstein’s model of colon cancer progression w203,204x, these adenomas should remain in a pre-neoplastic state and resist transformation into adenocarcinoma longer than adenomas with COX-2. This can be tested experimentally. 4.2. Non-steroidal anti-inflammatory drugs (NSAIDs) and cancer preÕention As noted in Section 2.4, administration of various NS AIDs retards development of malignant colon tumors, or causes pre-malignant adenomas to regress, thereby lowering the incidence and mortality of colon cancer w48–76x. In other types of solid tumors, especially those that do not have precursor lesions, preliminary investigations suggest that NSAIDs do not always confer such remarkable benefits w205–209x. At present, there are no explicit guidelines for the use of NSAIDs as preventive agents in oncology w210x. When these guidelines are established, it will be important to treat them as guidelines, not dogma —we still have much to learn about the preventive mechanisms of NSAIDs in cancer. For examples, inhibition of COX catalysis appears sufficient, but not necessary, for benefit, if one considers the results with exisulind and the R-enantiomer of flurbiprofen ŽE-7869., two atypical NSAID analogs w211x. Recent pre-clinical studies by its manufacturer suggest that Exisulind Žsulindac sulfone. induces apoptosis via inhibition of cyclic GMP-phosphodiesterase. Certain precancerous and cancerous cells may overexpress this enzyme, accounting for some selective toxicity
1659
of exisulind. Nominally, exisulind acts independently of COX-1 or COX-2 inhibition w212x; however, investigations with human tumors transplanted to rodents challenge this assertion w213x. Data with the R-enantiomer of flurbiprofen are equally provocative, and promising. Flurbiprofen occurs as a racemic mixture of R- and S-enantiomers like other NSAIDs in the 2-arylpropionic acid family. The S-enantiomers of NSAIDs, including flurbiprofen, are typically 100-fold more potent as inhibitors of COX enzymes, compared to the R-enantiomers. Clearance of the enantiomers is also stereoselective in both humans and rats. Importantly, metabolic inversion of R-flurbiprofen ŽCOX inactive. into S-flurbiprofen ŽCOX inhibitor. was not detected after oral administration of R-flurbiprofen to humans w214x. These latter data strengthen the argument that pharmacological effects of R-flurbiprofen in humans are not attributable to inhibition of COX isoenzymes. Animal experiments affirm that this agent might confer benefit in human disease w215–218x. In the Min mouse, a model of familial adenamatous polyposis, R-flurbiprofen prolongs survival when administered chronically; in the mouse TRAMP model of human prostate cancer, R-flurbiprofen lowered the incidence of metastasis in animal treated with a high fat diet. In these studies w215–218x, investigators took great pains to assure that results were not due to metabolic inversion of R-flurbiprofen ŽCOX inactive. to S-flurbiprofen ŽCOX inhibitor.. Nevertheless, there always remains a formal possibility that this occurs in rodent models.
5. Conclusion Persistent inflammation is a significant risk factor for several types of cancer. This phenomenon seems closely related to granulocytic secretion of genotoxic electrophiles—low exposures over a period of years elevate the risk for an oncogenic mutation. An implication of this model is that anti-inflammatory agents that reduce the granulocytic contribution to genomic instability will confer medical benefit in individuals at risk for cancer from their inflammatory disorder. Oddly, benefits from anti-inflammatory agents that actually reduce granulocytic inflammation are the exception, not the rule. The preventive benefit con-
1660
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
ferred by NSAIDs in colon polyps originates from their effects on cylooxygenase in the adenomatous tissue, or surrounding tissue, not from their effects on granulocytes. The recent failure of b-carotene anti-oxidant trials as a cancer prevention method accentuates the gap between the hypotheses driving these interventions and their successful implementation. It is difficult to abandon these hypotheses, leaving investigators with chemical factors other than electrophiles, such as the cytokines described by Nowicki et al. w226x, Hudson et al. w227x and Lin et al. w228x, as alternative explanations.
w8x
w9x
w10x
w11x
Acknowledgements w12x
The Huntsman Cancer Foundation, the National Cancer Institute ŽP30 CA 42014., and the National Institute of Allergy and Infectious Diseases RO1 AI 26730 provided funding. FAF is the Dee Glenn and Ida W. Smith Chair of Cancer Research. We thank Dr. Ernest Hawk, MD MPH from the Division of Cancer Prevention at the NCI for providing current, comprehensive information on NSAIDs and colorectal cancer prevention.
w13x
w14x
w15x
w16x
References w17x w1x Morson BC. Precancer and cancer in inflammatory bowel disease. Pathology 1985;17:173–80. w2x Ekbom A, Helmnick C, Zack M, Adami HO. Ulcerative colitis and colorectal cancer, a population based study. N Engl J Med 1990;323:1228–33. w3x Choi PM, Zelig MP. Similarity of colorectal cancer in Crohn’s disease and ulcerative colitis: implications for carcinogenesis and prevention. Gut 1994;35:950–4. w4x Lashner BA. Colorectal cancer in ulcerative colitis patients: survival curves and surveillance. Cleveland Clin J Med 1994;61:272–5. w5x Snapper SB, Syngal S, Friedman LS. Ulcerative colitis and colon cancer: more controversy than clarity. Dig Dis 1998; 16:81–7. w6x Aarnio M, Mustonen H, Mecklin JP, Jarvinen HJ. Prognosis of colorectal cancer varies in different high risk conditions. Ann Med 1998;30:75–80. w7x Ekbom A, Helmick CG, Zack M, Holmberg L, Adami HO.
w18x
w19x
w20x w21x w22x w23x w24x
Survival and causes of deaths in patients with inflammatory bowel disease: a population based study. Gastroenterology 1992;103:954–60. Brentnall TA, Crispin DA, Bronner MP, Cherian SP, Hueffed M, Rabinovitch PS, et al. Microsatellite instability in non-neoplastic mucosa from patients with chronic ulcerative colitis. Cancer Res 1996;56:1237–40. Moody GA, Jayanthi V, Probert CS, Mac Kay H, Mayberry JF. Long term therapy with sulphasalazine protects against colorectal cancer in ulcerative colitis; a retrospective study of colorectal cancer risk and compliance with treatment in Leicestershire. Eur J Gastroenterol Hepatol 1996;8:1179–83. Eaden J, Abrams K, Ekbom A, Jackson E, Mayberry J. Colorectal cancer prevention in ulcerative colitis: a casecontrol study. Aliment Pharmacol Ther 2000;14Ž2.:145–53. Singer II, Kawka DW, Schloemann S, Tessner T, Riehl T, Stenson WF. Cyclooxygenase 2 is induced in colonic epithelial cells in inflammatory bowel disease. Gastroenterology 1998;115:297–306. Agoff SN, Brentnall TA, Crispin DA, Taylor SL, Raaka S, Haggitt RC, et al. The role of cyclooxygenase 2 in ulcerative colitis-associated neoplasia. Am J Pathol 2000;157Ž3.: 737–45. Lesch CA, Kraus ER, Sanchez B, Gilbertsen R, Guglietta A. Lack of beneficial effect of COX-2 inhibitors in an experimental model of colitis. Exp Clin Pharmacol 1999; 21:99–104. Felder JB, Korelitz BI, Rajapakse R, Schwarz S, Horatagis AP, Gleim G. Effects of nonsteroidal antiinflammatory drugs on inflammatory bowel disease: a case-control study. Am J Gastroenterol 2000;95Ž8.:1949–54. Vesterinen E, Pukkala E, Timonen T, Aromaa A. Cancer incidence among 78,000 asthma patients. Int J Epidemiol 1993;22:976–82. Huovinen E, Kaprio J, Vesterinen E, Koskenvuo M. Mortality of adults with asthma: a prospective cohort study. Thorax 1997;52:49–54. Kallen B, Gunnarskog J, Conradson TB. Cancer risk in asthmatic subjects selected from hospital discharge registry. Eur Respir J 1993;6:694–7. Alavanja MC, Brownson RC, Boice JD, Hock E. Preexisting lung disease and lung cancer among nonsmoking women. Am J Epidemiol 1992;136Ž6.:623–32. Sep. 15. Wu AH, Fontham ET, Reynolds P, Greenberg RS, Buffler P, Liff J, et al. Previous lung disease and risk of lung cancer among lifetime nonsmoking women in the United States. Am J Epidemiol 1995;141Ž11.:1023–32. Ford RM. Primary lung cancer and asthma. Ann Allergy 1978;40:240–2. Calverly P. Asthma. Postgrad Med J 1996;72:12–8. Barnes P. Pathophysiology of asthma. Br J Clin Pharmacol 1996;42:3–10. Keeley D, Rees J. New guidelines in asthma management. Br Med J 1997;314:315–6. Borm PJ, Driscoll K. Particles, inflammation and respiratory tract carcinogenesis. Toxicol Lett 1996;88:109–13.
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667 w25x Askling J, Grunewald J, Eklund A, Hillerdal G, Ekbom A. Increased risk for cancer following sarcoidosis. Am J Respir Crit Care Med 1999;160Ž5 Pt. 1.:1668–72. w26x Rosin MP, Anwar WA, Ward AJ. Inflammation, chromosomal instability, and cancer: the schistosomiasis model. Cancer Res 1994;54Ž7 Suppl..:1929S–33S. w27x Burin GJ, Gibb HJ, Hill RN. Human bladder cancer: evidence for a potential irritation-induced mechanism. Food Chem Toxicol 1995;33:785–95. w28x Persky L. Epidemiology of cancer of the penis. Recent Results Cancer Res 1977;60:97–109. w29x Carlson JA, Ambros R, Malfetano J, Ross J, Grabowski R, Lamb P, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective: implications for chronic inflammation and sclerosis in the development of neoplasia. Hum Pathol 1998;29:932–48. w30x Mayron R, Grimwood RE, Siegle RJ, Camisa C. Verrucous carcinoma arising in ulcerative lichen planus of the soles. J Dermatol Surg Oncol 1988;14:547–51. w31x Ness RB, Cottreau C. Possible role of ovarian epithelial inflammation in ovarian cancer. J Natl Cancer Inst 1999; 91Ž17.:1459–67. w32x Ness RB, Grisso JA, Cottreau C, Klapper J, Vergona R, Wheeler JE, et al. Factors related to inflammation of the ovarian epithelium and risk of ovarian cancer. Epidemiology 2000;11Ž2.:111–7. w33x Schildkraut JM, Bastos E, Berchuk A. Relationship between lifetime ovarian cycles and overexpression of mutant p53 in epithelial ovarian cancer. J Natl Cancer Inst 1997;89:932–8. w34x Baker TR, Piver MS. Etiology, biology and epidemiology of ovarian cancer. Semin Surg Oncol 1994;10:242–8. w35x Brinton LA, Gridley G, Persson I, Baron J, Bergquist A. Cancer risk after a hospital discharge diagnosis of endometriosis. Am J Obstet Gynecol 1997;176:572–9. w36x Risch HA, Howe GR. Pelvic inflammatory disease and the risk of epithelial ovarian cancer. Cancer Epidemiol, Biomarkers Prev 1995;4:447–51. w37x Parazzini F, La Vecchia C, Negri E, Moroni S, dal Pino D, Fedele L. Pelvic inflammatory disease and risk of ovarian cancer. Cancer Epidemiol, Biomarkers Prev 1996;5Ž8.: 667–9. w38x Bansal P, Sonnenberg A. Pancreatitis is a risk factor for pancreatic cancer. Gastroenterology 1995;109:247–51. w39x Cylwik B, Nowak HF, Puchalski Z, Barczyk J. Epithelial anomalies in chronic pancreatitis as a risk factor of pancreatic cancer. Hepatogastroenterology 1998;45:528–32. w40x Kern SE. Molecular genetic alterations in ductal pancreatic adenocarcinomas. Med Clin North Am 2000;84:691–5. w41x Gansauge S, Schmid RM, Muller J, Adler G, Mattfeldt T, Berger HG. Genetic alterations in chronic pancreatitis: evidence for early occurrence of p53 but not K-ras mutations. Br J Surg 1998;85:337–40. w42x Howe JR, Conlon KC. The molecular genetics of pancreatic cancer. Surg Oncol 1997;6:1–18. w43x Karlson BM, Ekbom A, Josefsson S, McLaughlin JK, Fraumeni JF, Nyren O. The risk of pancreatic cancer following
w44x
w45x
w46x w47x w48x
w49x
w50x
w51x
w52x
w53x w54x
w55x
w56x
w57x
w58x
w59x
w60x
w61x
1661
pancreatitis: an association due to confounding? Gastroenterology 1997;113:587–92. Oberg S, Clark GW, DeMeester TR. Barrett’s esophagus; update of pathophysiology and management. Hepatogastroenterology 1998;45:1348–56. Morgan G, Vainio H. Barrett’s oesophagus, oesophageal cancer and colon cancer: an explanation of the association and cancer chemopreventive potential of non-steroidalantiinflammatory drugs. Eur J Cancer Prev 1998;7:195–9. Knudson AG. Mutation and cancer: Statistical study of retinoblastoma. Proc Natl Acad Sci U S A 1971;68:820–3. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990;61:759–67. Venkatachalam S, Shi YP, Jones SN, Vogel H, Bradley A, Pinkel D, et al. Retention of wild-type p53 in tumors from p53 heterozygous mice: reduction of p53 dosage can promote cancer formation. EMBO J 1998;17:4657–67. Paganini-Hill A, Chao A, Ross RK, Henderson BE. Aspirin use and chronic disease: a cohort study of the elderly. BMJ 1989;299:1247–50. Kune GA, Kune SA, Watson LF. Colorectal cancer risk, chronic illnesses, operations and medications: case control results from the Melbourne Colorectal Cancer Study. Cancer Res 1988;48:4399–404. Rosenberg L, Palmer JR, Zauber AG, Warshauer ME, Stolley PD, Shapiro S. A hypothesis: nonsteroidal anti-inflammatory drugs reduce the incidence of large-bowel cancer. J Natl Cancer Inst 1991;83Ž5.:355–8. Gridley G, McLaughlin JK, Ekbom A, Klareskog L, Adami HO, Hacker DG, et al. Incidence of cancer among patients with rheumatoid arthritis. J Natl Cancer Inst 1993;85:301–7. Suh O, Mettlin C, Petrelli NJ. Aspirin use, cancer, and polyps of the large bowel. Cancer 1993;72Ž4.:1171–7. Gann PH, Manson JE, Glynn RJ, et al. Low-dose aspirin and incidence of colorectal tumors in a randomized trial. J Natl Cancer Inst 1993;85:1220–4. Thun MJ, Namboodiri MM, Calle EE, Flanders DW, Heath Jr. CW. Aspirin use and risk of fatal cancer. Cancer Res 1993;53:1322–7. Greenberg ER, Baron JA, Freeman Jr. DH, et al. Reduced risk of large-bowel adenomas among aspirin users. J Natl Cancer Inst 1993;85:912–6. Schreinermachers DM, Everson RB. Aspirin use and lung, colon and breast cancer incidence in a prospective study. Epidemiology 1994;5:138–46. Giovannucci E, Rimm EG, Stampfer MJ, Colditz GA, Ascherio A, Willett WC. Aspirin use and the risk of colorectalcancer and adenoma in male health professionals. Ann Intern Med 1994;121:241–6. Giovannucci E, Egan KM, Hunter DJ, et al. Asprin and the risk of colorectal cancer in women. N Engl J Med 1995; 333:609–14. Muller AD, Sonnenberg A, Wasserman IH. Diseases preceding colon cancer. A case-control study among veterans. Dig Dis Sci 1994;39Ž11.:2480–4. Peleg II, Maibach HT, Brown SH, Wilcox CM. Aspirin and nonsteroidal anti-inflammatory drug use and the risk of
1662
w62x
w63x
w64x
w65x
w66x
w67x
w68x
w69x
w70x
w71x
w72x
w73x
w74x
w75x
w76x
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667 subsequent colorectal cancer. Arch Intern Med 1994; 154Ž4.:394–9. Martinez ME, McPherson RS, Levin B, Annegers JF. Aspirin and other nonsteroidal anti-inflammatory drugs and risk of colorectal adenomatous polyps among endoscoped individuals. Cancer Epidemiol, Biomarkers Prev 1995;4Ž7.: 703–7. Bansal P, Sonnenberg A. Risk factors of colorectal cancer in inflammatory bowel disease. Am J Gastroenterol 1996; 91:44–8. Peleg II, Lubin MF, Cotsonis GA, Clark WS, Wilcox CM. Long-term use of nonsteroidal antiinflammatory drugs and other chemopreventors and risk of subsequent colorectal neoplasia. Dig Dis Sci 1996;41Ž7.:1319–26. La Vecchia C, Negri E, Franceschi S, Conti E, Montella M, Giacosa A, et al. Aspirin and colorectal cancer. Br J Cancer 1997;76Ž5.:675–7. Rosenberg L, Louik C, Shapiro S. Nonsteroidal antiinflammatory drug use and reduced risk of large bowel carcinoma. Cancer 1998;82Ž12.:2326–33. Sandler RS, Galanko JC, Murray SC, Helm JF, Woosley JT. Aspirin and nonsteroidal anti-inflammatory agents and risk for colorectal adenomas. Gastroenterology 1998;114: 441–7. Sturmer T, Glynn RJ, Lee IM, et al. Aspirin use and colorectal cancer: post-trial follow-up data from the Physicians’ Health Study. Ann Intern Med 1998;128:713–20. Smalley W, Ray WA, Dougherty J, et al. Use of nonsteroidal anti-inflammatory drugs and incidence of colorectal cancer. Arch Intern Med 1999;159:161–6. Bucher C, Jordan P, Nickeleit V, Torhorst J, Mihatsch MJ. Relative risk of malignant tumors in analgesic abusers; effects of long-term intake of aspirin. Clin Nephrol 1999; 51Ž2.:67–72. Collet JP, Sharpe C, Belzile E, Boivin JF, Hanley J, Abenhaim L. Colorectal cancer prevention by non-steroidal antiinflammatory drugs: effects of dosage and timing. Br J Cancer 1999;81Ž1.:62–8. Garcia Rodriguez LA, Huerta-Alvarez C. Reduced incidence of colorectal adenoma among long-term users of nonsteroidal antiinflammatory drugs: a pooled analysis of published studies and a new population-based study. Epidemiology 2000;11Ž4.:376–81. Garcia-Rodriguez LA, Huerta-Alvarez C. Reduced risk of colorectal cancer among long-term users of aspirin and non aspirin nonsteroidal antiinflammatory drugs. Epidemiology 2001;12Ž1.:88–93. Dannenberg AJ, Zakim D. Chemoprevention of colorectal cancer through inhibition of cyclooxygenase-2. Semin Oncol 1999;26Ž5.:499–504. Coogan PF, Rosenberg L, Louik C, Zauber AG, Stolley PD, Strom BL, et al. NSAIDs and risk of colorectal cancer according to presence or absence of family history of the disease. Cancer Causes Control 2000;11Ž3.:249–55. Breuer-Katschinski B, Nemes K, Rump B, Leiendecker B, Marr A, Breuer N, et al. Long-term use of nonsteroidal antiinflammatory drugs and the risk of colorectal adenomas.
w77x
w78x
w79x
w80x w81x w82x w83x
w84x
w85x
w86x
w87x
w88x
w89x
w90x w91x
w92x
w93x
The Colorectal Adenoma Study Group. Digestion 2000; 61Ž2.:129–34. Williams CS, Tsujii M, Reese J, Dey SK, DuBois RN. Host cyclooxygenase-2 modulates carcinoma growth. J Clin Invest 2000;105Ž11.:1589–94. Sheng H, Shao J, Morrow JD, Beauchamp RD, DuBois RN. Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res 1998; 58Ž2.:362–6. Langenbach R, Loftin CD, Lee C, Tiano H. Cyclooxygenase-deficient mice. A summary of their characteristics and susceptibilities to inflammation and carcinogenesis. Ann N Y Acad Sci 1999;889:52–61. Taketo MM. Cyclooxygenase-2 inhibitors in tumorigenesis Žpart I.. J Natl Cancer Inst 1998;90Ž20.:1529–36. Taketo MM. Cyclooxygenase-2 inhibitors in tumorigenesis ŽPart II.. J Natl Cancer Inst 1998;90Ž21.:1609–20. Majerus PW. Prostaglandins: critical roles in pregnancy and colon cancer. Curr Biol 1998;8Ž3.:R87–9. Felder JB, Korelitz BI, Rajapakse R, Schwarz S, Horatagis AP, Gleim G. Effects of nonsteroidal antiinflammatory drugs on inflammatory bowel disease: a case-control study. Am J Gastroenterol 2000;95Ž8.:1949–54. Bonner GF, Walczak M, Kitchen L, Bayona M. Tolerance of nonsteroidal antiinflammatory drugs in patients with inflammatory bowel disease. Am J Gastroenterol 2000; 95Ž8.:1946–8. Lewis JG, Adams DO. Inflammation, oxidative DNA damage, and carcinogenesis. Environ Health Perspect 1987;76: 19–27. Jackson JH, Gajewski E, Schraufstatter IU, Hyslop PA, Fuciarelli AF, Cochrane CG, et al. Damage to the bases in DNA induced by stimulated human neutrophils. J Clin Invest 1989;84Ž5.:1644–9. Weitzman SA, Weitberg AB, Clark EP, Stossel TP. Phagocytes as carcinogens: malignant transformation produced by human neutrophils. Science 1985;227Ž4691.:1231–3. Weitzman SA, Gordon LI. Inflammation and cancer: role of phagocyte-generated oxidants in carcinogenesis. Blood 1990;76Ž4.:655–63. Tamatani T, Turk P, Weitzman S, Oyasu R. Tumorigenic conversion of a rat urothelial cell line by human polymorphonuclear leukocytes activated by lipopolysaccharide. Jpn J Cancer Res 1999;90Ž8.:829–36. Cerutti PA. Oxy-radicals and cancer. Lancet 1994; 344Ž8926.:862–3. Kasai H, Crain PF, Kuchino Y, Nishimura S, Ootsuyama A, Tanooka H. Formation of 8-hydroxyguanine moiety in cellular DNA by agents producing oxygen radicals and evidence for its repair. Carcinogenesis 1986;7Ž11.:1849–51. Olson MJ. DNA strand breaks induced by hydrogen peroxide in isolated rat hepatocytes. J Toxicol Environ Health 1988;23Ž3.:407–23. Muehlematter D, Larsson R, Cerutti P. Active oxygen induced DNA strand breakage and poly ADP-ribosylation in promotable and non-promotable JB6 mouse epidermal cells. Carcinogenesis 1988;9Ž2.:239–45.
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667 w94x Beehler BC, Przybyszewski J, Box HB, Kulesz-Martin MF. Formation of 8-hydroxydeoxyguanosine within DNA of mouse keratinocytes exposed in culture to UVB and H2O2. Carcinogenesis 1992;13Ž11.:2003–7. w95x Lloyd DR, Carmichael PL, Phillips DH. Comparison of the X formation of 8-hydroxy-2 -deoxyguanosine and single- and double-strand breaks in DNA mediated by fenton reactions. Chem Res Toxicol 1998;11Ž5.:420–7. w96x Hirota N, Yokoyama T. Enhancing effect of hydrogen peroxide upon duodenal and upper jejunal carcinogenesis in rats. Gann 1981;72Ž5.:811–2. w97x Radosevich CA, Weitzman SA. Hydrogen peroxide induces squamous metaplasia in a hamster tracheal organ explant culture model. Carcinogenesis 1989;10Ž10.:1943–6. w98x Moraes EC, Keyse SM, Tyrrell RM. Mutagenesis by hydrogen peroxide treatment of mammalian cells: a molecular analysis. Carcinogenesis 1990;11Ž2.:283–93. w99x Huang RP, Peng A, Hossain MZ, Fan Y, Jagdale A, Boynton AL. Tumor promotion by hydrogen peroxide in rat liver epithelial cells. Carcinogenesis 1999;20Ž3.:485–92. w100x Lin X, Ramamurthi K, Mishima M, Kondo A, Howell SB. p53 interacts with the DNA mismatch repair system to modulate the cytotoxicity and mutagenicity of hydrogen peroxide. Mol Pharmacol 2000;58Ž6.:1222–9. w101x Wink DA, Kasprzak KS, Maragos CM, Elespuru RK, Misra M, Dunams TM, et al. DNA deaminating ability and genotoxicity of nitric oxide and its progenitors. Science 1991; 254Ž5034.:1001–3. w102x Nguyen T, Brunson D, Crespi CL, Penman BW, Wishnok JS, Tannenbaum SR. DNA damage and mutation in human cells exposed to nitric oxide in vitro. Proc Natl Acad Sci U S A 1992;89Ž7.:3030–4. w103x Grishko VI, Druzhyna N, LeDoux SP, Wilson GL. Nitric oxide-induced damage to mtDNA and its subsequent repair. Nucleic Acids Res 1999;27Ž22.:4510–6. w104x Burney S, Caulfield JL, Niles JC, Wishnok JS, Tannenbaum SR. The chemistry of DNA damage from nitric oxide and peroxynitrite. Mutat Res 1999;424Ž1–2.:37–49. w105x Caulfield JL, Wishnok JS, Tannenbaum SR. Nitric oxide-induced deamination of cytosine and guanine in deoxynucleosides and oligonucleotides. J Biol Chem 1998;273Ž21.: 12689–95. w106x deRojas-Walker T, Tamir S, Ji H, Wishnok JS, Tannenbaum SR. Nitric oxide induces oxidative damage in addition to deamination in macrophage DNA. Chem Res Toxicol 1995;8Ž3.:473–7. w107x Felley-Bosco E. Role of nitric oxide in genotoxicity: implication for carcinogenesis. Cancer Metastasis Rev 1998; 17Ž1.:25–37. w108x Calmels S, Hainaut P, Ohshima H. Nitric oxide induces conformational and functional modifications of wild-type p53 tumor suppressor protein. Cancer Res 1997;57Ž16.: 3365–9. w109x Chazotte-Aubert L, Pluquet O, Hainaut P, Ohshima H. Nitric oxide prevents gamma-radiation-induced cell cycle arrest by impairing p53 function in MCF-7 cells. Biochem Biophys Res Commun 2001;281Ž3.:766–71.
1663
w110x Basu AK, O’Hara SM, Valladier P, Stone K, Mols O, Marnett LJ. Identification of adducts formed by reaction of guanine nucleosides with malondialdehyde and structurally related aldehydes. Chem Res Toxicol 1988;1Ž1.:53–9. w111x Stone K, Uzieblo A, Marnett LJ. Studies of the reaction of malondialdehyde with cytosine nucleosides. Chem Res Toxicol 1990;3Ž5.:467–72. w112x Chaudhary AK, Nokubo M, Reddy GR, Yeola SN, Morrow JD, Blair IA, et al. Detection of endogenous malondialdehyde–deoxyguanosine adducts in human liver. Science 1994;265Ž5178.:1580–2. w113x Marnett LJ. Lipid peroxidation-DNA damage by malondialdehyde. Mutat Res 1999;424:83–95. w114x Mukai FH, Goldstein BD. Mutagenicity of malonaldehyde, a decomposition product of peroxidized polyunsaturated fatty acids. Science 1976;191:868–9. w115x Basu AK, Marnett LJ. Unequivocal demonstration that malondialdehyde is a mutagen. Carcinogenesis 1983;4: 331–3. w116x Benamira M, Johnson K, Chaudhary A, Bruner K, Tibbetts C, Marnett LJ. Induction of mutations by replication of malondialdehyde-modified M13 DNA in Escherichia coli: determination of the extent of DNA modification, genetic requirements for mutagenesis, and types of mutations induced. Carcinogenesis 1995;16:93–9. w117x Fischer SM, Ogle S, Marnett LJ, Nesnow S, Slaga TJ. The lack of initiating andror promoting activity of sodium malondialdehyde on SENCAR mouse skin. Cancer Lett 1983;19:61–6. w118x Ji C, Rouzer CA, Marnett LJ, Pietenpol JA. Induction of cell cycle arrest by the endogenous product of lipid peroxidation, malondialdehyde. Carcinogenesis 1998;19: 1275–83. w119x Douki T, Ames BN. An HPLC-EC assay for 1,N2-propano X adducts of 2 -deoxyguanosine with 4-hydroxynonenal and other alpha,beta-unsaturated aldehydes. Chem Res Toxicol 1994;7:511–8. w120x Lee SH, Rindgen D, Bible RH, Hajdu E, Blair IA. CharacX terization of 2 -deoxyadenosine adducts derived from 4oxo-2-nonenal, a novel product of lipid peroxidation. Chem Res Toxicol 2000;13:565–74. w121x Chung FL, Nath RG, Ocando J, Nishikawa A, Zhang L. Deoxyguanosine adducts of t-4-hydroxy-2-nonenal are endogenous DNA lesions in rodents and humans: detection and potential sources. Cancer Res 2000;60:1507–11. w122x Nishikawa A, Furukawa F, Kasahara K, Ikezaki S, Itoh T, Suzuki T, et al. Trans-4-hydroxy-2-nonenal, an aldehydic lipid peroxidation product, lacks genotoxicity in lacI transgenic mice. Cancer Lett 2000;148:81–6. w123x Burcham PC. Genotoxic lipid peroxidation products: their DNA damaging properties and role in formation of endogenous DNA adducts. Mutagenesis 1998;13:287–305. w124x Feig DI, Reid TM, Loeb A. Reactive oxygen species in tumorigenesis. Cancer Res 1994;54:1890S–4S. w125x Du MQ, Carmichael PL, Philips DH. Induction of activating mutations in the human c-Ha-ras-1 proto-oncogene by oxygen free radicals. Mol Carcinog 1994;11:170–5.
1664
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
w126x Cerda S, Weitzman SA. Influence of oxygen radical injury on DNA methylation. Mutat Res 1997;386:141–52. w127x Ghosh R, Mitchell DL. Effect of oxidative DNA damage in promoter elements on transcription factor binding. Nucleic Acids Res 1999;27:3213–8. w128x Hu JJ, Dubin N, Kurland D, Ma BL, Roush GC. The effects of hydrogen peroxide on DNA repair activities. Mutat Res 1995;336:193–201. w129x Collins AR. Oxidative DNA damage, antioxidants, and cancer. Bioessays 1999;21:238–46. w130x Hennekens CH, Buring JE, Manson JE, Stampfer M, Rosner B, Cook NR, et al. Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular disease. N Engl J Med 1996; 334:1145–9. w131x Lee IM, Cook NR, Manson JE, Buring JE, Hennekens CH. Beta-carotene supplementation and incidence of cancer and cardiovascular disease: the Women’s Health Study. J Natl Cancer Inst 1999;91:2102–6. w132x Frieling UM, Schaumberg DA, Kupper TS, Muntwyler J, Hennekens CH. A randomized, 12-year primary-prevention trial of beta carotene supplementation for nonmelanoma skin cancer in the physician’s health study. Arch Dermatol 2000;136:179–84. w133x Omenn GS, Goodman GE, Thornquist MD, Balmes J, Cullen MR, Glass A, et al. Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N Engl J Med 1996;334:1150–5. w134x Christen WG, Gaziano JM, Hennekens CH. Design of Physicians’ Health Study II—a randomized trial of betacarotene, vitamins E and C, and multivitamins, in prevention of cancer, cardiovascular disease, and eye disease, and review of results of completed trials. Ann Epidemiol 2000; 10:125–34. w135x Reiber DC, Murphy RC. Covalent binding of LTAŽ4. to nucleosides and nucleotides. Arch Biochem Biophys 2000; 379:119–26. w136x Brambilla G, Martelli A, Marinari UM. Is lipid peroxidation associated with DNA damage? Mutat Res 1989;214Ž1.: 123–7. w137x Moos PJ, Edes K, Fitzpatrick FA. Inactivation of the wildtype p53 tumor supprerssor by electrophilic prostaglandins. Proc Natl Acad Sci U S A 2000;97-16:9215–20. w138x Zhang JR, Sevanian A. The genotoxic effects of arachidonic acid in V79 cells are mediated by peroxidation products. Toxicol Appl Pharmacol 1993;121Ž2.:193–202. w139x Miller EC, Miller JA. Searches for ultimate chemical carcinogens and their reactions with cellular macromolecules. Cancer 1981;47:2327–45. w140x Oren M, Maltzman W, Levine AJ. Post-translational regulation of the 54 K cellular tumor antigen in normal and transformed cells. Mol Cell Biol 1981;1:101–10. w141x Momand J, Zambetti GP, Olson DC, George D, Levine AJ. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 1992;69:1237–45.
w142x Wu XW, Bayle JH, Olson D, Levine AJ. The p53 mdm-2 autoregulatory feedback loop. Gene Dev 1993;7:1126–32. w143x Kubbutat MHG, Jones SN, Vousden KH. Regulation of p53 stability by Mdm2. Nature 1997;387:299–303. w144x Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature 1997;387:296–9. w145x Rodriguez MS, Desterro JM, Lain S, Lane DP, Hay RT. Multiple C-terminal lysine residues target p53 for ubiquitin-proteasome-mediated degradation. Mol Cell Biol 2000; 20:8458–67. w146x Lane DP. P53, guardian of the genome. Nature 1992;358: 15–6. w147x Yu J, Zhang L, Hwang PM, Rago C, Kinzler KW, Vogelstein B. Identification and classification of p53-regulated genes. Proc Natl Acad Sci U S A 1999;96:14517–22. w148x Prives C, Hall PA. The p53 pathway. J Pathol 1999;187: 112–26. w149x Ljungman M. Dial 9-1-1 for p53: mechanisms of p53 activation by cellular stress. Neoplasia 2000;2:209–26. w150x Goh HS, Yao J, Smith DR. p53 point mutation and survival in colorectal cancer patients. Cancer Res 1995;55:5217–21. w151x Peller S. Clinical implications of p53: effect on prognosis, tumor progression and chemotherapy response. Semin Cancer Biol 1998;8:379–87. w152x van Oijen MGC, Slootweg PJ. Gain of function mutations in the tumor suppressor gene p53. Clin Cancer Res 2000;6: 2138–45. w153x O’Connor PM, Jackman J, Bae I, Myers TG, Fan S, Mutoh M, et al. Characterization of the p53 tumor suppressor pathway in cell lines of the National cancer Institute anticancer drug screen and correlations with the growth inhibitory potency of 123 anticancer agents. Cancer Res 1997;57:4285–300. w154x Baker SJ, Fearon ER, Nigro JM, Hamilton SR, Preisinger AC, Jessup JM, et al. Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas. Science 1989; 244:217–21. w155x Hainaut P, Hollstein M. p53 and human cancer: the first ten thousand mutations. Adv Cancer Res 2000;77:81–137. w156x Soussi T, Dehouche K, Beroud C. p53 website and analysis of p53 gene mutations in human cancer: forging a link between epidemiology and carcinogenesis. Hum Mutat 2000;15:105–13. w157x Reich NC, Levine AJ. Specific interaction of the SV40 T antigen–cellular p53 protein complex with SV40 DNA. Virology 1982;117:286–90. w158x Vousden K. Interactions of human papillomavirus transforming proteins with the products of tumor suppressor genes. FASEB J 1993;7:872–9. w159x Oliner JD, Kinzler KW, Meltzer PS, Georges DL, Vogelstein B. Amplification of a gene encoding a p53 associated protein in human sarcomas. Nature 1992;358:80–3. w160x Maki CG, Huibregtse JM, Howley PM. In vivo ubiquitination and proteasome-mediated degradation of p53. Cancer Res 1996;56:2649–54. w161x Kubbutat MH, Vousden KH. Keeping an old friend under
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
w162x
w163x
w164x
w165x w166x
w167x
w168x
w169x w170x w171x
w172x
w173x
w174x
w175x
w176x
w177x
control: regulation of p53 stability. Mol Md Today 1998;4: 250–6. Hainaut P, Milner J. A structural role for metal ions in the Awild-typeB conformation of the tumor suppressor protein p53. Cancer Res 1993;53Ž8.:1739–42. Khan QA, Agarwal R, Seidel A, Frank H, Vousden KH, Dipple A. DNA adduct levels associated with p53 induction and delay of MCF-7 cells in S phase after exposure to benzowgxchrysene dihydrodiol epoxide enantiomers. Mol Carcinog 1998;23:115–20. Parks D, Bolinger R, Mann K. Redox state regulates binding of p53 to sequence specific DNA but not to non-specific or mismatched DNA. Nucleic Acids Res 1997;25:1289–95. Jayaaman L, Prives C. Covalent and non-covalent modifiers of the p53 protein. Cell Mol Life Sci 1999;55:76–87. Vile GF, Rothwell LA, Kettle AJ. Initiation of rapid, p53dependent growth arrest in cultured human skin fibroblasts by reactive chlorine species. Arch Biochem Biophys 2000; 377:122–8. Martinez JD, Pennington ME, Craven MT, Warters RL, Cress AE. Free radicals generated by ionizing radiation signal nuclear translocation of p53. Cell Growth Differ 1997;8:941–9. Wu HH, Momand J. Pyrrolidine dithiocarbamate prevents p53 activation and promotes p53 cysteine residue oxidation. J Biol Chem 1998;273:18898–905. Loeb KR, Loeb LA. Significance of multiple mutations in cancer. Carcinogenesis 2000;21:379–85. Meuth M. Patterns of mutation in cancer cells. Cancer Surv 1996;28:33–46. Fitzpatrick FA, Wynalda MA. Albumin–lipid interactions: prostaglandin stability as a probe for characterizing binding sites on vertebrate albumins. Biochemistry 1981;20:6129– 34. Fitzpatrick FA, Wynalda MA. Albumin catalyzed metabolism of prostaglandin D2: products formed in vitro. J Biol Chem 1983;258:11713–8. Lin AH, Bienkowski MJ, Gorman RR. Regulation of prostaglandin H synthase mRNA levels and prostaglandin biosynthesis by platelet-derived growth factor. J Biol Chem 1989;264:17379–83. Bailey JM, Muza B, Hla T, Salata K. Restoration of prostacyclin synthase in vascular smooth muscle cells after aspirin treatment: regulation by epidermal growth factor. J Lipid Res 1985;26:54–61. Bartolini G, Orlandi M, Chiricolo M, Minghetti L, Guerrini F, Fidan M, et al. Regulation of thromboxane A2 biosynthesis in platelet-free human monocytes and the possible role of polypeptide growth factorŽs. in the induction of cyclooxygenase system. Biochim Biophys Acta 1986;876:486– 93. O’Neill LA, Barrett ML, Lewis GP. Induction of cyclooxygenase by interleukin-1 in rheumatoid synovial cells. FEBS Lett 1987;212:35–9. Frasier-Scott K, Hatzakis H, Seong D, Jones CM, Wu KK. Influence of natural and recombinant interleukin-2 on endothelial cell arachidonate metabolism. Induction of de
w178x
w179x
w180x
w181x
w182x
w183x
w184x
w185x
w186x w187x w188x
w189x
w190x
w191x
w192x
1665
novo synthesis of prostaglandin H synthase. J Clin Invest 1988;82:1877–83. Raz A, Wyche A, Siegel N, Needleman P. Regulation of fibroblast cyclooxygenase synthesis by interleukin-1. J Biol Chem 1988;263:3022–8. Raz A, Wyche A, Needleman P. Temporal and pharmacological division of fibroblast cyclooxygenase expression into transcriptional and translational phases. Proc Natl Acad Sci U S A 1989;86:1657–61. Fu JY, Masferrer JL, Seibert K, Raz A, Needleman P. The induction and suppression of prostaglandin H2 synthase Žcyclooxygenase. in human monocytes. J Biol Chem 1990; 265:16737–40. Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Herschman HR. TIS10, a phorbol ester tumor promoter-inducible mRNA from Swiss 3T3 cells, encodes a novel prostaglandin synthasercyclooxygenase homologue. J Biol Chem 1991;266:12866–72. Jones DA, Carlton DP, McIntyre TM, Zimmerman GA, Prescott SM. Molecular cloning of human prostaglandin endoperoxide synthase type II and demonstration of expression in response to cytokines. J Biol Chem 1993;268: 9049–54. Ristimaki A, Garfinkel S, Wessendorf J, Maciag T, Hla T. Induction of cyclooxygenase-2 by interleukin-1 alpha, evidence for post-transcriptional regulation. J Biol Chem 1994;269:11769–75. Smith WL, Marnett LJ. Prostaglandin endoperoxide synthase: structure and catalysis. Biochim Biophys Acta 1991; 1083:1–17. Lipsky PE, Abramson SB, Breedveld FC, Brook P, Burmester R, Buttgereit F, et al. Analysis of the effect of COX-2 specific inhibitors and recommendations for their use in clinical practice. J Rheumatol 2000;27:1338–40. Taketo MM. Cyclooxygenase-2 inhibitors in tumorigenesis Žpart I.. J Natl Cancer Inst 1998;90:1529–36. Taketo MM. Cyclooxygenase-2 inhibitors in tumorigenesis ŽPart II.. J Natl Cancer Inst 1998;90:1609–20. Oshima M, Dinchuk JE, Kargman SL, Oshima H, Hancock B, Kwong E, et al. Suppression of intestinal polyposis in Apc delta716 knockout mice by inhibition of cyclooxygenase 2 ŽCOX-2.. Cell 1996;87:803–9. Chulada PC, Thompson MB, Mahler JF, Doyle CM, Gaul BW, Lee C, et al. Genetic disruption of Ptgs-1, as well as Ptgs-2, reduces intestinal tumorigenesis in Min mice. Cancer Res 2000;60:4705–8. Chan TA, Morin PJ, Vogelstein B, Kinzler KW. Mechanisms underlying nonsteroidal antiinflammatory drug-mediated apoptosis. Proc Natl Acad Sci U S A 1998;95Ž2.:681–6. Surette ME, Fonteh AN, Bernatchez C, Chilton FH. Perturbations in the control of cellular arachidonic acid levels block cell growth and induce apoptosis in HL-60 cells. Carcinogenesis 1999;20:757–63. Longo WE, Grossmann EM, Erickson B, Panesar N, Mazuski JE, Kaminski DL. The effect of phospholipase A2 inhibitors on proliferation and apoptosis of murine intestinal cells. Surg Res 1999;84:51–6.
1666
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667
w193x Cao Y, Pearman AT, Zimmerman GA, McIntyre TM, Prescott SM. Intracellular unesterified arachidonic acid signals apoptosis. Proc Natl Acad Sci U S A 2000;97Ž21.: 11280–5. w194x Sheng H, Shao J, Morrow JD, Beauchamp RD, DuBois RN. Modulation of apoptosis and Bcl-2 expression by prostaglandin E2 in human colon cancer cells. Cancer Res 1998; 58:362–6. w195x Tsujii M, DuBois RN. Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2. Cell 1995;83:493–501. w196x Tsujii M, Kawano S, Tsuji S, Sawaoka H, Hori M, DuBois RN. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell 1998;93:705–16. Published erratum appears in Cell 94, following 271, 1998. w197x Robert A. Prostaglandins: effects on the gastrointestinal tract. Clin Physiol Biochem 1984;2Ž2–3.:61–9. w198x Guan Z, Buckman SY, Pentland AP, Templeton DJ, Morrison AR. Induction of cyclooxygenase-2 by the activated MEKK1–SEK1rMKK4–p38 mitogen-activated protein kinase pathway. J Biol Chem 1998;273Ž21.:12901–8. w199x Moos PJ, Muskardin DT, Fitzpatrick FA. Effect of taxol and taxotere on gene expression in macrophages: induction of the prostaglandin H synthase-2 isoenzyme. J Immunol 1999;162Ž1.:467–73. w200x Dean JL, Brook M, Clark AR, Saklatvala J. p38 mitogenactivated protein kinase regulates cyclooxygenase-2 mRNA stability and transcription in lipopolysaccharide-treated human monocytes. J Biol Chem 1999;274Ž1.:264–9. w201x Dixon DA, Kaplan CD, McIntyre TM, Zimmerman GA, Prescott SM. Post-transcriptional control of cyclooxyX genase-2 gene expression. The role of the 3 -untranslated Ž . region. J Biol Chem 2000;275 16 :11750–7. w202x Toyota M, Shen L, Ohe-Toyota M, Hamilton SR, Sinicrope FA, Issa JP. Aberrant methylation of the cyclooxygenase-2 CpG island in colorectal tumors. Cancer Res 2000;60: 4044–8. w203x Kinzler KW, Vogelstein BW. Lessons from hereditary colorectal cancer. Cell 1996;87:159–70. w204x Prescott SM, White RL. Self-promotion? Intimate connections between APC and prostaglandin H synthase-2. Cell 1996;87:783–6. w205x Harris RE, Namboodiri KK, Farrar WB. Non-steroidal antiinflammatory drugs and breast cancer. Epidemiology 1996; 7:203–5. w206x Egan KM, Stampfer MJ, Giovannucci E, Rosner BA, Colditz GA. Prospective study of regular aspirin use and the risk of breast cancer. J Natl Cancer Inst 1996;88:988–93. w207x Langman MJ, Cheng KK, Gilman EA, Lancashire RJ. Effect of anti-inflammatory drugs on overall risk of common cancer: case-control study in general practice research database. Br Med J 2000;320:1642–6. w208x Tavani A, Gallus S, La Vecchia C, Conti E, Montella M, Franceschi S. Aspirin and ovarian cancer: an Italian casecontrol study. Ann Oncol 2000;11:1171–3. w209x Nelson JE, Harris RE. Inverse association of prostate cancer and non-steroidal anti-inflammatory drugs ŽNSAIDs.: results of a case-control study. Oncol Rep 2000;7:169–70.
w210x Sjodahl R. Extent, mode, and dose dependence of anticancer effects. Am J Med 2001;110Ž1 Suppl. 1.:S66–9. w211x Marx J. Anti-inflammatories inhibit cancer growth—but how? Science 2001;291:5504. w212x Piazza GA, Alberts DS, Hixson LJ, Paranka NS, Li H, Finn T, et al. Sulindac sulfone inhibits azoxymethane-induced colon carcinogenesis in rats without reducing prostaglandin levels. Cancer Res 1997;57:2909–15. w213x Williams CS, Goldman AP, Sheng H, Morrow JD, DuBois RN. Sulindac sulfide, but not sulindac sulfone, inhibits colorectal cancer growth. Neoplasia 1999;1:170–6. w214x Jamali F, Berry BW, Tehrani MR, Russell AS. Stereoselective pharmacokinetics of flurbiprofen in humans and rats. J Pharm Sci 1988;77:666–9. w215x McCracken JD, Wechter WJ, Liu Y, Chase RL, Kantoci D, Murray Jr. ED, et al. Antiproliferative effects of the enantiomers of flurbiprofen. J Clin Pharmacol 1996;36:540–5. w216x Wechter WJ, Kantoci D, Murray Jr. ED, Quiggle DD, Leipold DD, Gibson KM, et al. R-flurbiprofen chemoprevention and treatment of intestinal adenomas in the APCŽMin.rqmouse model: implications for prophylaxis and treatment of colon cancer. Cancer Res 1997;57:4316– 24. w217x Wechter WJ, Murray Jr. ED, Kantoci D, Quiggle DD, Leipold DD, Gibson KM, et al. Treatment and survival study in the C57BLr6J-APCŽMin.rqŽMin. mouse with R-flurbiprofen. Life Sci 2000;66:745–53. w218x Wechter WJ, Leipold DD, Murray Jr. ED, Quiggle D, McCracken JD, Barrios RS, et al. E-7869 Ž R-flurbiprofen. inhibits progression of prostate cancer in the TRAMP mouse. Cancer Res 2000;60:2203–8. w219x Kutchera W, Jones DA, Matsunami N, Groden J, McIntyre TM, Zimmerman GA, et al. Prostaglandin H synthase 2 is expressed abnormally in human colon cancer: evidence for a transcriptional effect. Proc Natl Acad Sci U S A 1996;93: 4816–20. w220x Soslow RA, Dannenberg AJ, Rush D, Woerner BM, Khan KN, Masferrer J, et al. COX-2 is expressed in human pulmonary, colonic, and mammary tumors. Cancer 2000; 89:2637–45. w221x Lim HY, Joo HJ, Choi JH, Yi JW, Yang MS, Cho DY, et al. Increased expression of cyclooxygenase-2 protein in human gastric carcinoma. Clin Cancer Res 2000;6:519–25. w222x Chan G, Boyle JO, Yang EK, Zhang F, Sacks PG, Shah JP, et al. Cyclooxygenase-2 expression is up-regulated in squamous cell carcinoma of the head and neck. Cancer Res 1999;59:991–4. w223x Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, et al. Cyclooxygenase-2 expression is up-regulated in human pancreatic cancer. Cancer Res 1999;59: 987–90. w224x Fujita T, Matsui M, Takaku K, Uetake H, Ichikawa W, Taketo MM, et al. Size- and invasion-dependent increase in cyclooxygenase 2 levels in human colorectal carcinomas. Cancer Res 1998;58:4823–6. w225x Kulkarni S, Rader JS, Zhang F, Liapis H, Koki AT, Masferrer JL, et al. Cyclooxygenase-2 is overexpressed in human cervical cancer. Clin Cancer Res 2001;7:429–34.
F.A. Fitzpatrickr International Immunopharmacology 1 (2001) 1651–1667 w226x Nowicki A, Szenajch J, Ostrowska G, Wojtowicz A, Wojtowicz K, Kruszewski AA, et al. Impaired tumor growth in colony-stimulating factor 1 ŽCSF-1.-deficient, macrophagedeficient oprop mouse: evidence for a role of CSF-1-dependent macrophages in formation of tumor stroma. Int J Cancer 1996;65:112–9.
1667
w227x Hudson JD, Shoaibi MA, Maestro R, Carnero A, Hannon GJ, Beach DH. A proinflammatory cytokine inhibits p53 tumor suppressor activity. J Exp Med 1999;190:1375–82. w228x Lin EY, Nguyen AV, Russell RG, Pollard JW. Colonystimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med 2001;193:727–40.