Review Open Access
Copyright ©2012 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastrointest Surg. Mar 27, 2012; 4(3): 62-72
Published online Mar 27, 2012. doi: 10.4240/wjgs.v4.i3.62
Inflammation and cancer
Noemí Eiró, Francisco J Vizoso, Research Unit, Fundación Hospital de Jove, 33290 Gijón, Asturias, Spain
Author contributions: Both authors made substantial contributions to the conception and design, acquisition, analysis and interpretation of data, drafting the article and revising it critically for important intellectual content; and both gave final approval of the version to be published.
Correspondence to: Francisco J Vizoso, MD, PhD, Head, Research Unit, Fundación Hospital de Jove, 33290 Gijón, Asturias, Spain. investigacion@hospitaldejove.com
Telephone: +34-985-320050 Fax: +34-985-315710
Received: October 20, 2010
Revised: November 17, 2011
Accepted: November 25, 2011
Published online: March 27, 2012

Abstract

There is evidence supporting the hypothesis that inflammation participates in providing conditions that lead to cancer. An unresolved inflammation due to any failure in the precise control of the immune response can continue to perturb the cellular microenvironment, thereby leading to alterations in cancer-related genes and posttranslational modification in crucial cellular proteins involved in the cell cycle, DNA repair and apoptosis. In addition, there are data indicating that inflammatory cells and immunomodulatory mediators present in the tumor microenvironment influence tumor progression and metastasis. Historically, tumor-infiltrating leukocytes have been considered to be manifestations of an intrinsic defence mechanism against developing tumors. However, increasing evidence indicates that leukocyte infiltration can promote tumor phenotypes, such as angiogenesis, growth and invasion. This may be due to inflammatory cells that probably can influence cancer promotion by secreting cytokines, growth factors, chemokines and proteases, which stimulate proliferation and invasiveness of cancer cells. Consequently, events and molecules implicated in this cross talk between the tumor microenvironment and inflammatory process may emerge as attractive targets in anticancer therapeutic interventions with significant clinical impact.

Key Words: Inflammation, Cancer, Cytokines, Proliferation, Cancer progression, Metastasis



INTRODUCTION

That continuous irritation over long periods of time can lead to cancer was described in Ayurveda (means the science of long life), written as far back as 5000 years ago. Aulus Cornelius Celsus, a Roman medic in the first century, defined inflammation as: “rubor” (redness), “tumor” (swelling), “calor” (heat) and “dolor” (pain). Rodolf Virchow postulated that microinflammation that results from irritation leads to the development of most chronic diseases, including cancer[1]. Although this concept has long been suspected, only recently experimental and clinical studies have confirmed this hypothesis, which is now a generally accepted paradigm[2,3]. It is estimated that underlying infections and inflammatory reactions are linked to 15%-20% of all cancer deaths.

CHRONIC INFLAMMATION AS A PREDISPOSITION TO CANCER

The first evidence that non-cancerous cells might affect the formation and growth of tumors derives from the field of inflammation. Today, there is evidence supporting the hypothesis that inflammation participates in providing conditions that lead to cancer. It is estimated that underlying infections and inflammatory reactions are linked to 25% of all cancer cases. In many epidemiological studies, the role of chronic inflammation in the carcinogenesis process was examined through studies of pro-inflammatory and anti-inflammatory cytokines along with other factors, including viral infections and genetic markers that take part in the inflammatory response[4].

There are well known associations between inflammatory processes and cancer, such as bowel disease (Crohn’s disease and especially ulcerative colitis) and colorectal cancer[5,6], viral hepatitis B and C or alcoholic liver cirrhosis and hepatocarcinoma[7], chronic reflux esophagitis resulting in Barrett’s esophagus and esophageal carcinoma[6], cervical infection by human papillomavirus and cervical cancer, prostatitis and prostate cancer, pancreatitis and pancreatic cancer, or gastric infection from Helicobacter pylori, which increases gastric cancer risk by 75%[8,9].

Tissue injury, whether physical, chemical or infectious, triggers a sequence of events that constitutes the inflammatory response. Inflammation is an important mechanism that can eliminate the agent responsible for the injury and initiate tissue repair by launching a well-coordinated immune response. The inflammatory mechanism involves both innate and adaptive immunity, which is characterized by coordinated blood borne delivery to injured tissues of cells and soluble mediators. After the elimination of the invading pathogen and wound healing, inflammation subsides. However, an unresolved inflammation on account of any failure in the precise control of the immune response can continue to perturb the cellular microenvironment, thereby leading to alterations in cancer-related genes and posttranslational modification in key cell signaling proteins involved in cell cycle, DNA repair and apoptosis. In fact, it is of note that mononuclear inflammatory cells (MICs) are often present at the very early stage of tumor development, in close association with areas of hyperplasia and atypia[10,11]. These findings support the concept that MICs themselves are a driving force that contributes to tumor initiation and/or initial tumoral progression. In addition to macrophages, mast cells and neutrophils can also support tumoral development by leading to upregulation of non-specific pro-inflammatory cytokines, such as interferon-γ, tumor necrosis factor (TNF), interleukin (IL)-1α/β or IL-6[12,13]. Likewise, activated nuclear factor-κB (NF-κB) transcription factor is one of the main links between inflammation and tumorogenesis and may be key to allowing both preneoplastic and malignant cells to escape from apoptosis[14,15]. Therefore, all of these factors may act as initiators and promoters of carcinogenesis by directly increasing the proliferation of epithelial cells. Table 1 shows studies reporting the relationship between overexpression of molecular components from inflammation pathway and gastrointestinal carcinogenesis.

Table 1 Overexpression of inflammatory factors in gastrointestinal carcinogenesis.
FactorsGastrointestinal cancers
NF-κBDifferential expression and constitutive activation was correlated with severity of oral lesions in the course of oral cancer development[112]
Elevated activated nuclear factor-κB-regulated cytokines were found in oral lichen planus patients and in the saliva of patients with oral squamous cell carcinoma[113,114]
Associated with hepatocarcinogenesis induced by viral hepatitis B or viral hepatitis C infection[115,116]
COX-2Associated with pathogenesis in mucositis[117]
Associated in Barrett's metaplasia with a change in the local inflammatory reaction[118]
Important overexpression in the pathogenesis of cholangiocarcinomas[119]
Contribute to suppression of local immune responses and enhancement of metastatic potential in human hepatocellular carcinoma[120]
Predictive marker in the early stages of hepatocarcinogenesis[121]
Related to the histological grade of intraductal papillary-mucinous tumor of the pancreas[122]
Gene expression was elevated in human colorectal cancer compared to normal mucosa[123]
Associated with prognosis and intestinal pathways in gastric carcinogenesis[124,125]
STAT-3Associated with the tumorigenesis of colorectal carcinoma[126]
Associated with cell survival in gastric cancer[127]
Associated with the development and proliferation of colorectal cancer[128]
IL-6Associated with the development of apoptosis resistance Barrett's esophagus[129]
Associated with the growth and proliferation in human colorectal cancer[130,131]
NOSCorrelated with Barrett's-associated neoplastic progression[132]
5-LOXAssociated with the development of pancreatic cancer[133]

On the other hand, it is noteworthy that increased toll-like receptors (TLRs) expression has been described in different human tumors[16-24]. It is an interesting finding because TLRs are considered a link between innate (non-specific) and adaptive (specific) immunity and contribute to the immune system’s capacity to efficiently combat pathogens[17]. As molecular sensors, TLRs detect pathogen-derived products and couple to different adapter proteins that trigger specific signaling pathways, such as the IL-1 receptor-associated kinase family and serine/threonine-protein kinase (TBK-1). These adapters initiate pathways leading to the activation of their respective transcription factors, NF-κB and interferon regulatory factor 3 (IRF3). Both NF-κB and IRF3 induce the release of various immune and inflammatory cytokines, such as TNF and IL6, which proved to be excellent targets for inflammatory diseases[25]. TLR-deficient mice were found to be protected from or develop less inducible tumors in experimental models[16,24]. In addition, for example, components of bacteria and viruses have been identified within pathological specimens of men with prostate cancer. There is evidence that the presence of pathogens in the urinary system may contribute to the malignant transformation of prostate epithelia through the activation of TLRs[19,26,27]. Therefore, all of this evidence indicates that biological signals elicited from TLR-activated tumor cells might also be a molecular link between inflammation and cancer.

TUMOR MICROENVIRONMENT AND ITS CONTRIBUTION TO TUMOR PROGRESSION TOWARDS METASTASIS

Tumors are composed not only of cancer cells but also of other cell types constituting the stroma. These stromal cells include cancer associated fibroblasts, endothelial cells, pericytes and variable representation of leukocytes. Leukocytes can account for as much 50% of the total tumor mass in invasive breast carcinomas. Initially, tumor cells and cells of the tumor microenvironment respond to tumor hypoxia and necrosis, secondary to excessive tumor cell proliferation, by releasing a number of growth factors and cytokines that are chemoattractive for monocytes and macrophages. These latter factors include colony stimulating factor (CSF)-1, granulocyte-monocyte-CSF, transforming growth factor (TGF)-β and chemokines[13]. In addition, macrophage-tumor cell interaction leads to the release of macrophage-derived cytokines, chemokines and growth/motility factors, such as IL-8 and fibroblast growth factor, which in turn promote the recruitment of additional inflammatory cells[28].

Historically, tumor-infiltrating leukocytes have been considered to be manifestations of an intrinsic defence mechanism against developing tumors[29,30]. The presence of leukocytes in tumors was subsequently interpreted as an aborted attempt of the immune system to reject the tumor. However, increasing evidence indicates that leukocyte infiltration can promote tumor phenotypes, such as angiogenesis, growth and invasion[31,32]. This may be due to inflammatory cells that can probably influence cancer promotion by secreting cytokines, growth factors, chemokines and proteases, which stimulate proliferation and invasiveness of cancer cells[28,33,34]. These factors reported as released by recruited MICs, included TNF, vascular endothelial growth factor (VEGF)- A and -C, hepatocyte growth factor (HGF), epidermal growth factor (EGF) family members, basic fibroblast growth factor, platelet-derived growth factor and chemokines, such as chemokine ligand 12 (CXCL12) and IL-8[4,13,35]. In addition, it is also remarkable that MICs bring in much of the cyclooxygenase-2 (COX-2) present in the tumor environment. It is because COX-2 expression and prostaglandins production within the tumor environment stimulate tumor cell proliferation, survival and motility, but also tumor angiogenesis[36].

CLINICAL RELEVANCE OF INFLAMMATORY COMPONENTS IN TUMOR PROGRESSION

Accumulating clinical data for solid tumors shows a correlation between high-density leukocytic infiltration into tumors and poor outcome of patients with malignancies of different origins, such as breast[37,38], bladder[39], rectum[40], endometrium[41], melanomas[42], gliomas[43] or leiomyosarcomas[44]. In addition, deficient monocyte recruitment at tumor sites in mice lacking CSF-1 expression was shown to attenuate late-stage progression and metastasis formation, suggesting that monocytes contribute to tumor progression[45]. Nevertheless, the presence of inflammatory cells can be an indicator of favorable prognosis in some tumor types, as for example, the presence of macrophages in colorectal cancer[46,47], gastric[48] or ovarian carcinomas[49]. These latter data suggest that, at least in some situations, inflammatory cells may be able to eliminate tumor cells just as they can destroy normal cells. On the other hand, one of the reasons why the prognostic significance of the lymphoid infiltrate at the tumor site remains controversial is perhaps because the evaluation criteria for tumor infiltrates are not sufficiently standardized to yield reliable and reproducible results in different institutions. Leukocyte infiltrate includes a variable representation of leukocytes, including macrophages, neutrophils, mast cells and T and B lymphocytes[31,50]. In addition, there is evidence indicating that different types of leukocyte infiltration occur in different carcinomas and that probably these are induced by different abilities to control tumor growth according to the tumor dissemination[51]. Therefore, inflammatory cells and immunomodulatory mediators present in the tumor microenvironment polarize host immune response toward specific phenotypes impacting tumor progression.

Macrophages are often the most abundant immune cell population in the tumor microenvironment. Recruitment of monocyte precursors circulating in the blood leads to their differentiation into tumor-associated macrophages. It has been reported that, once recruited into tumors, macrophages can assume two different phenotypes: M1 or M2, based on environmental stimuli and each expressing specialized functional properties[52]. The M1 phenotype is associated with inflammation and microbial killing activity, whereas the M2 phenotype is associated with activities which are predominant and key events in cancer, including inhibition of T helper 1 adaptive immunity by immunosuppressive mediators [TGFβ, IL-10 or prostaglandin E2 (PGE2)], production of growth and survival factors (EGF, IL-6 and CXCL8), secretion of angiogenic factors (VEGF, TGFα or PGE2), production of matrix metalloproteases (MMPs) which degrade extracellular matrix, and chemokines able to recruit more inflammatory cells (CCL17, CCL18 or CCL22)[33,52,53].

In this sense, we recently identified a phenotype of MICs at the intratumor stroma of 40% of breast carcinomas, which is associated with the development of distant metastasis. These MICs were characterized by overexpression of metalloproteases (MMP)-7, 9, 11, 13 and 14, and tissue inhibitors of metalloproteases (TIMP)-1 and 2[54,55]. This may be because MMPs play an essential role in the degradation of the stromal connective tissue and basement membrane components, which are key elements in tumor invasion and metastasis. MMPs are also able to impact in vivo on tumor cell behavior as a consequence of their capacity to cleave growth factors, cell surface receptors, cell adhesion molecules and chemokines/cytokines[56-58]. Furthermore, by cleaving proapoptotic factors, MMPs produce a more aggressive phenotype via generation of apoptotic resistant cells[56]. MMPs also regulate cancer-related angiogenesis, positively through their ability to mobilize or activate proangiogenic factors[59], and negatively via generation of angiogenesis inhibitors, such as angiostatin and endostatin, cleaved from large protein precursors[60]. In addition, inflammatory cells from tumor stroma play a role in tumor angiogenesis by releasing other factors, such as VEGF, HGF or IL-8, which are able to stimulate and activate endothelial cells. On the other hand, it is now accepted that TIMPs are multifactorial proteins also involved in the induction of proliferation and the inhibition of apoptosis[61,62].

It is noteworthy that many of these molecules which have been identified as playing a critical role in inflammation are regulated by NF-κB. This is a transcription factor that is ubiquitous to all cell types and present in the cytoplasm in its resting stage. There is evidence pointing to the role of NF-κB in tumoral progression. Thus, NF-κB has also been linked with the survival of cancer stem cells[63]. NF-κB regulates the expression of most antiapoptotic gene products associated with the survival of tumors [bcl2-like 1 (bcl-xl), B-cell lymphoma 2 (bcl-2), X-linked inhibitor of apoptosis protein, cellular FLICE-inhibitory protein, inhibitor of apoptosis (IAP)-1 and IAP-2 and surviving], as well as gene products linked with proliferation of tumors (cyclin D1, c-myc and COX-2). In addition, recent data supports a role of the NF-κB-regulated inflammatory network in the progression, diagnosis, prognosis, recurrence and treatment of cancer in patients[64]. Table 2 shows several studies reporting the relationship between NF-κB and/or related molecules with poor prognosis in gastrointestinal tumors.

Table 2 Different factors associated with poor prognosis in gastrointestinal tumors.
FactorsGastrointestinal cancers
NF-κBAn independent prognostic indicator of poor outcome in patients with esophageal adenocarcinoma[134]
High expression of activated nuclear factor-κB indicates poor patient survival in pancreatic cancer[135]
Activation in hepatocellular carcinoma was implicated in a poor patient outcome[136]
Associated with a shorter overall survival rate and prognosis biomarker in gastric cancer[137-139]
Nuclear factor-κB positivity after radiotherapy was linked with worse clinical outcome in rectal cancer[140]
COX-2The most important predictor of poor survival in oropharyngeal squamous cell carcinoma[141]
Correlated with tumor progression and an unfavorable prognostic factor in esophageal carcinomas [142-146]
Prognostic factor after surgical resection in patients affected by cancer of ampulla of vater[147]
Associated with liver metastasis and poor survival in primary colorectal cancer[148-152]
Associated with invasion, metastasis and implicated a poor prognosis in gastric carcinoma[153]
Linked to an increased risk of hematogenous metastatic spread in rectal carcinoma[154]
CXCR-4Associated with poor clinical outcome in esophageal cancer patients[155]
Associated with lymph node metastasis and early distant relapse in colorectal cancer[156,157]
VEGFAn independent prognostic factor for patients with nasopharyngeal carcinoma[158]
Associated with prognosis in squamous cell carcinoma of the esophagus[159]
VEGF-C and VEGF-D expression was associated with lymphatic metastasis and prognosis in patients with pancreatic adenocarcinoma and induced lymphangiogenesis[160]
Associated with prognosis in patients with hepatocellular carcinoma[161]
Associated with prognosis in colorectal cancer patients[162,163]
Associated with angiogenesis and metastasis in gastric cancer[164]
Associated with lymph node metastasis and progression of ampullary carcinoma[165]
IL-6Interleukin-1β and Interleukin-6 expression was associated with the growth and progression of human gastric carcinoma[166]
Associated with progression and poor prognosis of colorectal carcinoma[167,168]
IL-8Associated with the tumor progression and hepatic metastasis in patients with colorectal cancer[169-171]
Associated with metastatic potential, angiogenesis and cell proliferation in human hepatocellular carcinoma[172]
Associated with prognosis in human gastric carcinoma[173]
MMP-9Correlated with the metastasis of lymph node in gastric cancer[174]
Associated with tumor cell differentiation, vessel permeation, lymph node metastasis in esophageal squamous cell carcinoma[175]
NOSAssociated with angiogenesis of hepatocellular carcinoma[176]
Correlated with the progression in gastric cancer patients[177]

On the other hand, as mentioned above, elevated TLRs expression has been described in different human tumors[16-24]. It is noteworthy because cancer cells activated by TLR signals may release cytokines and chemokines that in turn may recruit immune cells and stimulate them to release further cytokines and chemokines. This process results in a cytokine profile that is associated with immune tolerance, cancer progression and propagation of the tumor microenvironment[22]. In fact, recent studies by our group revealed that breast carcinomas with high TLR3 expression by tumor cell or with high TLR4 expression by MICs were significantly associated with higher probability of metastasis[65]. Likewise, we found that samples of prostate carcinomas with recurrence exhibited a significant increase in the mRNA levels of TLR3, TLR4 and TLR9. In addition, the tumors that showed high level expression of TLR3 or TLR9 were significantly associated with higher probability of biochemical recurrence[66]. However, we also found that breast tumors with high TLR9 expression by fibroblast-like cells were associated with a low metastasis rate. Although the biological significance of TLR9 expression by stromal fibroblast-like cells is currently unknown, there are data pointing to a protective role against tumor progression. Indeed, it was demonstrated that stimulation of TLR9 activates human plasmacytoid dendritic cells and B cells and this induces potent innate immune responses in preclinical tumor models as well as in patients[67]. The increasing interest in using bindings of nucleic acid-sensing TLR9 as a pharmacological intervention in various diseases is thus understandable. All of these data suggest that further studies of the expression of TLRs in malignant tumors may help to better understand the process that links inflammation and cancer, as well as to assess the biological and clinical importance of the interaction between tumor and stroma.

IMMUNOTHERAPY AND ANTI-INFLAMMATORY THERAPY IN CANCER

It is considered that the main mechanism of tumor immunity is due to an antitumoral T cell response[68]. This antitumor response can be due to the direct killing of tumor cells by CD8 cytotoxic T lymphocytes which recognize major histocompatibility complex class I and other antigens expressed on the surface of tumor cells. However, it is generally assumed that during growth, tumors develop strategies to evade or limit the effects of the host’s immune responses[69]. In most cases, the adaptive immune response against tumor cells is very weak and largely inefficient, since the tumor itself and the surrounding microenvironment tumor induce immunosuppression by the down-regulation of CD8 cytotoxic T lymphocytes response[68]. It has also been suggested that increasing immune activity or immunotherapy will exacerbate the rate of immune escape and select for a tumor sub-population, which will be resistant to immunotherapy. In addition, the secretion of immunosuppressive cytokines and chemokines into the tumor microenvironment, such as TGF-β, IL-6 and IL-10, which can interfere with multiple steps and pathways in the generation of an effective immune response, has been described in many types of tumors[68]. Nevertheless, it is noteworthy that an alternative approach was recently described in which the expression of new, and thereby potent, antigens are induced in tumor cells by inhibiting nonsense-mediated messenger RNA decay (NMD)[70,71]. It has been demonstrated that small interfering RNA-mediated inhibition of NMD in tumor cells led to the expression of new antigenic determinants and their immune-mediated rejection[72]. Therefore, it would be of interest to determine whether the NMD-induced antigens are cross-reactive among different tumors in future studies, and if so, to identify the dominant antigens induced by NMD inhibition.

On the other hand, it is clear that several inflammatory markers are expressed in various cancers and mediate their progression. Consequently, agents which suppress these inflammatory markers or the pathways activated by them have a potential for prevention and treatment of cancer. Some of these agents are being tested, such as steroids (dexamethasone and prednisolone)[73,74], TNF inhibitors (humira, remicade, enbrel and thalidomide)[75-77], proteasome inhibitors (velcade)[78] and NF-κB inhibitors (curcumin)[79]. It is also of note that most nutraceuticals derived from fruits, vegetables, legumes or spices have been shown to suppress NF-κB activation pathways, thus leading to suppression of various inflammatory markers. COX-2 has also been proposed as a therapeutic target for cancer prevention and treatment [COX-2 inhibitors (COXIB), such as aspirin and celecoxib][80]. The appearance of cardiovascular complications induced by potent COXIBs has dampened enthusiasm and hampered the widespread use of COXIBs for cancer chemoprevention. TLRs may also represent a good therapeutic target in cancer. In this sense, there are studies that show a variable antineoplastic effect caused by a blockade of TLR3[81,82]. However, the authors suggested that TLR3 plays a role in inhibiting the cell cycle and inducing apoptosis[20]. In addition, the use of TLR3 agonists has been successful in prostate immune-based therapies[83,84]. The implications for therapeutic interventions using TLRs as targets could justify numerous creative strategies. Further study of this association is warranted and therapeutic strategies to boost or block these pathways may be relevant.

Mesenchymal stem cells (MSCs) represent a promising tool for new clinical concepts in supporting cellular therapy. Although bone marrow (BM) has been the main source for the isolation of multipotent MSCs, adipose tissue is another source of this cell[85], but the harvest of BM and adipose tissue is a highly invasive procedure. Therefore, alternative sources from which to isolate MSCs are subject to intensive investigation. One alternative source is umbilical cord blood, which can be obtained by a less invasive method, without harm to the mother or infant[86]. Other sources of MSCs were identified in a variety of other human adult tissues, including placenta[87], scalp tissue[88] and myometrium[89].

MSCs appear to suppress inflammation through secretion of anti-inflammatory cytokines, such as IL-10[90], TGF-β[91], soluble human leukocyte antigen-G[92] and IL-1 receptor antagonist[93], and expression of immune regulatory enzymes, such as cycloxygenase[94] and indolamine 2,3 deoxygenase. MSCs were able to suppress T-lymphocyte activation and proliferation in vitro[95-97]. The mechanisms are probably mediated by both direct cell-cell contacts and soluble factors. As regards the effect of MSC on B-lymphocyte function, MSC inhibit immunoglobulin production and arrest B-lymphocytes in the G0/G1 phase of the cell cycle. MSCs have been demonstrated to interfere with dendritic cell differentiation, maturation and function[98-101]. Based on these properties, MSCs are being used in regenerative medicine, for the treatment of autoimmune diseases[102,103], graft versus host disease[104-109] and the tropism of MSCs for human gliomas can also be exploited to therapeutic advantage[110]. These cells can be a new alternative in cancer studies; in fact, recently an inhibiting effect of the MSC on the proliferation of the tumor was described[111].

It seems clear that an understanding of how tumor cells control and benefit from host inflammation responses can open the way towards the identification of therapeutic strategies targeting the molecular mechanisms that underlie relevant tumor-host interactions.

CONCLUSION

Chronic and persistent inflammation contributes to cancer development and even predisposes to carcinogenesis. In addition, cellular and molecular components from tumor-associated inflammation may affect neoplastic progression. Events and molecules implicated in this crosstalk between the tumor and inflammatory microenvironment may emerge as attractive targets in anticancer therapeutic intervention with significant clinical impact. Thus, anti-inflammatory agents should be explored for both prevention and treatment of cancer. Their true potential will be recognized only through well-controlled clinical trials.

Footnotes

Peer reviewer: Shouji Shimoyama, MD, PhD, President, Gastrointestinal Unit, Settlement Clinic, 4-20-7, Towa, Adachi-ku, Tokyo 120-0003, Japan

S- Editor Wang JL L- Editor Roemmele A E- Editor Xiong L

References
1.  Heidland A, Klassen A, Rutkowski P, Bahner U. The contribution of Rudolf Virchow to the concept of inflammation: what is still of importance? J Nephrol. 2006;19 Suppl 10:S102-S109.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell. 2005;7:211-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1284]  [Cited by in F6Publishing: 1267]  [Article Influence: 66.7]  [Reference Citation Analysis (0)]
3.  Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5245]  [Cited by in F6Publishing: 5418]  [Article Influence: 235.6]  [Reference Citation Analysis (0)]
4.  Lorusso G, Rüegg C. The tumor microenvironment and its contribution to tumor evolution toward metastasis. Histochem Cell Biol. 2008;130:1091-1103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 343]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
5.  Eaden J, Abrams K, Ekbom A, Jackson E, Mayberry J. Colorectal cancer prevention in ulcerative colitis: a case-control study. Aliment Pharmacol Ther. 2000;14:145-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 405]  [Cited by in F6Publishing: 430]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
6.  van der Woude CJ, Kleibeuker JH, Jansen PL, Moshage H. Chronic inflammation, apoptosis and (pre-)malignant lesions in the gastro-intestinal tract. Apoptosis. 2004;9:123-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
7.  Matsuzaki K, Murata M, Yoshida K, Sekimoto G, Uemura Y, Sakaida N, Kaibori M, Kamiyama Y, Nishizawa M, Fujisawa J. Chronic inflammation associated with hepatitis C virus infection perturbs hepatic transforming growth factor beta signaling, promoting cirrhosis and hepatocellular carcinoma. Hepatology. 2007;46:48-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 228]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
8.  Hussain SP, Amstad P, Raja K, Ambs S, Nagashima M, Bennett WP, Shields PG, Ham AJ, Swenberg JA, Marrogi AJ. Increased p53 mutation load in noncancerous colon tissue from ulcerative colitis: a cancer-prone chronic inflammatory disease. Cancer Res. 2000;60:3333-3337.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Kuper H, Adami HO, Trichopoulos D. Infections as a major preventable cause of human cancer. J Intern Med. 2000;248:171-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 505]  [Cited by in F6Publishing: 487]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
10.  Mantovani A, Schioppa T, Porta C, Allavena P, Sica A. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev. 2006;25:315-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 604]  [Cited by in F6Publishing: 588]  [Article Influence: 34.6]  [Reference Citation Analysis (0)]
11.  Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71-78.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Aggarwal BB, Shishodia S, Sandur SK, Pandey MK, Sethi G. Inflammation and cancer: how hot is the link? Biochem Pharmacol. 2006;72:1605-1621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 954]  [Cited by in F6Publishing: 903]  [Article Influence: 50.2]  [Reference Citation Analysis (0)]
13.  Robinson SC, Coussens LM. Soluble mediators of inflammation during tumor development. Adv Cancer Res. 2005;93:159-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 88]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
14.  Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006;441:431-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2609]  [Cited by in F6Publishing: 2679]  [Article Influence: 148.8]  [Reference Citation Analysis (0)]
15.  Naugler WE, Karin M. NF-kappaB and cancer-identifying targets and mechanisms. Curr Opin Genet Dev. 2008;18:19-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 454]  [Cited by in F6Publishing: 471]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
16.  Fukata M, Chen A, Vamadevan AS, Cohen J, Breglio K, Krishnareddy S, Hsu D, Xu R, Harpaz N, Dannenberg AJ. Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology. 2007;133:1869-1881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 449]  [Cited by in F6Publishing: 474]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
17.  Kawai T, Akira S. TLR signaling. Cell Death Differ. 2006;13:816-825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1429]  [Cited by in F6Publishing: 1476]  [Article Influence: 82.0]  [Reference Citation Analysis (0)]
18.  Kelly MG, Alvero AB, Chen R, Silasi DA, Abrahams VM, Chan S, Visintin I, Rutherford T, Mor G. TLR-4 signaling promotes tumor growth and paclitaxel chemoresistance in ovarian cancer. Cancer Res. 2006;66:3859-3868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 369]  [Cited by in F6Publishing: 394]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
19.  Kundu SD, Lee C, Billips BK, Habermacher GM, Zhang Q, Liu V, Wong LY, Klumpp DJ, Thumbikat P. The toll-like receptor pathway: a novel mechanism of infection-induced carcinogenesis of prostate epithelial cells. Prostate. 2008;68:223-229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 110]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
20.  Paone A, Starace D, Galli R, Padula F, De Cesaris P, Filippini A, Ziparo E, Riccioli A. Toll-like receptor 3 triggers apoptosis of human prostate cancer cells through a PKC-alpha-dependent mechanism. Carcinogenesis. 2008;29:1334-1342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 129]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
21.  Salaun B, Coste I, Rissoan MC, Lebecque SJ, Renno T. TLR3 can directly trigger apoptosis in human cancer cells. J Immunol. 2006;176:4894-4901.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Sato Y, Goto Y, Narita N, Hoon DS. Cancer Cells Expressing Toll-like Receptors and the Tumor Microenvironment. Cancer Microenviron. 2009;2 Suppl 1:205-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 234]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
23.  Stark JR, Wiklund F, Grönberg H, Schumacher F, Sinnott JA, Stampfer MJ, Mucci LA, Kraft P. Toll-like receptor signaling pathway variants and prostate cancer mortality. Cancer Epidemiol Biomarkers Prev. 2009;18:1859-1863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
24.  Swann JB, Vesely MD, Silva A, Sharkey J, Akira S, Schreiber RD, Smyth MJ. Demonstration of inflammation-induced cancer and cancer immunoediting during primary tumorigenesis. Proc Natl Acad Sci U S A. 2008;105:652-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 241]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
25.  O'Neill LA, Bryant CE, Doyle SL. Therapeutic targeting of Toll-like receptors for infectious and inflammatory diseases and cancer. Pharmacol Rev. 2009;61:177-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 318]  [Cited by in F6Publishing: 335]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
26.  Nickel JC, Moon T. Chronic bacterial prostatitis: an evolving clinical enigma. Urology. 2005;66:2-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
27.  Tanner MA, Shoskes D, Shahed A, Pace NR. Prevalence of corynebacterial 16S rRNA sequences in patients with bacterial and "nonbacterial" prostatitis. J Clin Microbiol. 1999;37:1863-1870.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Balkwill F. Cancer and the chemokine network. Nat Rev Cancer. 2004;4:540-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1747]  [Cited by in F6Publishing: 1735]  [Article Influence: 86.8]  [Reference Citation Analysis (0)]
29.  Johnson JP, Riethmüller G, Schirrmacher V. Tumor immunology: Paul Ehrlich's heritage. Immunol Today. 1989;10:S35-S37.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Lin EY, Pollard JW. Role of infiltrated leucocytes in tumour growth and spread. Br J Cancer. 2004;90:2053-2058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 186]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
31.  Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860-867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10123]  [Cited by in F6Publishing: 10557]  [Article Influence: 479.9]  [Reference Citation Analysis (0)]
32.  Daniel D, Chiu C, Giraudo E, Inoue M, Mizzen LA, Chu NR, Hanahan D. CD4+ T cell-mediated antigen-specific immunotherapy in a mouse model of cervical cancer. Cancer Res. 2005;65:2018-2025.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
33.  Le Bitoux MA, Stamenkovic I. Tumor-host interactions: the role of inflammation. Histochem Cell Biol. 2008;130:1079-1090.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 80]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
34.  Sica A, Bronte V. Altered macrophage differentiation and immune dysfunction in tumor development. J Clin Invest. 2007;117:1155-1166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 898]  [Cited by in F6Publishing: 943]  [Article Influence: 55.5]  [Reference Citation Analysis (0)]
35.  Benelli R, Lorusso G, Albini A, Noonan DM. Cytokines and chemokines as regulators of angiogenesis in health and disease. Curr Pharm Des. 2006;12:3101-3115.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Prescott SM, Fitzpatrick FA. Cyclooxygenase-2 and carcinogenesis. Biochim Biophys Acta. 2000;1470:M69-M78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 144]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
37.  Pollard JW. Macrophages define the invasive microenvironment in breast cancer. J Leukoc Biol. 2008;84:623-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 324]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
38.  Shabo I, Stål O, Olsson H, Doré S, Svanvik J. Breast cancer expression of CD163, a macrophage scavenger receptor, is related to early distant recurrence and reduced patient survival. Int J Cancer. 2008;123:780-786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 166]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
39.  Hanada T, Nakagawa M, Emoto A, Nomura T, Nasu N, Nomura Y. Prognostic value of tumor-associated macrophage count in human bladder cancer. Int J Urol. 2000;7:263-269.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Shabo I, Olsson H, Sun XF, Svanvik J. Expression of the macrophage antigen CD163 in rectal cancer cells is associated with early local recurrence and reduced survival time. Int J Cancer. 2009;125:1826-1831.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 103]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
41.  Salvesen HB, Akslen LA. Significance of tumour-associated macrophages, vascular endothelial growth factor and thrombospondin-1 expression for tumour angiogenesis and prognosis in endometrial carcinomas. Int J Cancer. 1999;84:538-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
42.  Jensen TO, Schmidt H, Møller HJ, Høyer M, Maniecki MB, Sjoegren P, Christensen IJ, Steiniche T. Macrophage markers in serum and tumor have prognostic impact in American Joint Committee on Cancer stage I/II melanoma. J Clin Oncol. 2009;27:3330-3337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 220]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
43.  Nishie A, Ono M, Shono T, Fukushi J, Otsubo M, Onoue H, Ito Y, Inamura T, Ikezaki K, Fukui M. Macrophage infiltration and heme oxygenase-1 expression correlate with angiogenesis in human gliomas. Clin Cancer Res. 1999;5:1107-1113.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Lee CH, Espinosa I, Vrijaldenhoven S, Subramanian S, Montgomery KD, Zhu S, Marinelli RJ, Peterse JL, Poulin N, Nielsen TO. Prognostic significance of macrophage infiltration in leiomyosarcomas. Clin Cancer Res. 2008;14:1423-1430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 135]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
45.  Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med. 2001;193:727-740.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Forssell J, Oberg A, Henriksson ML, Stenling R, Jung A, Palmqvist R. High macrophage infiltration along the tumor front correlates with improved survival in colon cancer. Clin Cancer Res. 2007;13:1472-1479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 373]  [Cited by in F6Publishing: 393]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
47.  Zhou Q, Peng RQ, Wu XJ, Xia Q, Hou JH, Ding Y, Zhou QM, Zhang X, Pang ZZ, Wan DS. The density of macrophages in the invasive front is inversely correlated to liver metastasis in colon cancer. J Transl Med. 2010;8:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 149]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
48.  Ohno S, Inagawa H, Dhar DK, Fujii T, Ueda S, Tachibana M, Suzuki N, Inoue M, Soma G, Nagasue N. The degree of macrophage infiltration into the cancer cell nest is a significant predictor of survival in gastric cancer patients. Anticancer Res. 2003;23:5015-5022.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Tanaka Y, Kobayashi H, Suzuki M, Kanayama N, Suzuki M, Terao T. Upregulation of bikunin in tumor-infiltrating macrophages as a factor of favorable prognosis in ovarian cancer. Gynecol Oncol. 2004;94:725-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
50.  Lin EY, Pollard JW. Macrophages: modulators of breast cancer progression. Novartis Found Symp. 2004;256:158-68; discussion 168-72, 259-69.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Canevari S, Pupa SM, Ménard S. 1975-1995 revised anti-cancer serological response: biological significance and clinical implications. Ann Oncol. 1996;7:227-232.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Allavena P, Sica A, Garlanda C, Mantovani A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol Rev. 2008;222:155-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 438]  [Cited by in F6Publishing: 449]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
53.  Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23:549-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3655]  [Cited by in F6Publishing: 3755]  [Article Influence: 170.7]  [Reference Citation Analysis (0)]
54.  González LO, Pidal I, Junquera S, Corte MD, Vázquez J, Rodríguez JC, Lamelas ML, Merino AM, García-Muñiz JL, Vizoso FJ. Overexpression of matrix metalloproteinases and their inhibitors in mononuclear inflammatory cells in breast cancer correlates with metastasis-relapse. Br J Cancer. 2007;97:957-963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 68]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
55.  Vizoso FJ, González LO, Corte MD, Rodríguez JC, Vázquez J, Lamelas ML, Junquera S, Merino AM, García-Muñiz JL. Study of matrix metalloproteinases and their inhibitors in breast cancer. Br J Cancer. 2007;96:903-911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 176]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
56.  Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2:161-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4447]  [Cited by in F6Publishing: 4393]  [Article Influence: 199.7]  [Reference Citation Analysis (0)]
57.  Mañes S, Llorente M, Lacalle RA, Gómez-Moutón C, Kremer L, Mira E, Martínez-A C. The matrix metalloproteinase-9 regulates the insulin-like growth factor-triggered autocrine response in DU-145 carcinoma cells. J Biol Chem. 1999;274:6935-6945.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Noë V, Fingleton B, Jacobs K, Crawford HC, Vermeulen S, Steelant W, Bruyneel E, Matrisian LM, Mareel M. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 2001;114:111-118.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Stetler-Stevenson WG. Matrix metalloproteinases in angiogenesis: a moving target for therapeutic intervention. J Clin Invest. 1999;103:1237-1241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 593]  [Cited by in F6Publishing: 590]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
60.  Cornelius LA, Nehring LC, Harding E, Bolanowski M, Welgus HG, Kobayashi DK, Pierce RA, Shapiro SD. Matrix metalloproteinases generate angiostatin: effects on neovascularization. J Immunol. 1998;161:6845-6852.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Jiang Y, Goldberg ID, Shi YE. Complex roles of tissue inhibitors of metalloproteinases in cancer. Oncogene. 2002;21:2245-2252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 370]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
62.  Würtz SØ, Schrohl AS, Sørensen NM, Lademann U, Christensen IJ, Mouridsen H, Brünner N. Tissue inhibitor of metalloproteinases-1 in breast cancer. Endocr Relat Cancer. 2005;12:215-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 93]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
63.  Guzman ML, Rossi RM, Neelakantan S, Li X, Corbett CA, Hassane DC, Becker MW, Bennett JM, Sullivan E, Lachowicz JL. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood. 2007;110:4427-4435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 301]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
64.  Aggarwal BB, Gehlot P. Inflammation and cancer: how friendly is the relationship for cancer patients? Curr Opin Pharmacol. 2009;9:351-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 265]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
65.  González-Reyes S, Marín L, González L, González LO, del Casar JM, Lamelas ML, González-Quintana JM, Vizoso FJ. Study of TLR3, TLR4 and TLR9 in breast carcinomas and their association with metastasis. BMC Cancer. 2010;10:665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 166]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
66.  González-Reyes S, Fernández JM, González LO, Aguirre A, Suárez A, González JM, Escaff S, Vizoso FJ. Study of TLR3, TLR4, and TLR9 in prostate carcinomas and their association with biochemical recurrence. Cancer Immunol Immunother. 2011;60:217-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 99]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
67.  Flores-Reséndiz D, Castellanos-Juárez E, Benítez-Bribiesca L. [Proteases in cancer progression]. Gac Med Mex. 2009;145:131-142.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Mantovani A, Romero P, Palucka AK, Marincola FM. Tumour immunity: effector response to tumour and role of the microenvironment. Lancet. 2008;371:771-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 372]  [Cited by in F6Publishing: 393]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
69.  Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by tumors. Adv Immunol. 2006;90:51-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 488]  [Cited by in F6Publishing: 490]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
70.  Maquat LE. Nonsense-mediated mRNA decay: splicing, translation and mRNP dynamics. Nat Rev Mol Cell Biol. 2004;5:89-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 866]  [Cited by in F6Publishing: 899]  [Article Influence: 45.0]  [Reference Citation Analysis (0)]
71.  Mühlemann O, Eberle AB, Stalder L, Zamudio Orozco R. Recognition and elimination of nonsense mRNA. Biochim Biophys Acta. 2008;1779:538-549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
72.  Pastor F, Kolonias D, Giangrande PH, Gilboa E. Induction of tumour immunity by targeted inhibition of nonsense-mediated mRNA decay. Nature. 2010;465:227-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 193]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
73.  Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, Oudard S, Théodore C, James ND, Turesson I. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351:1502-1512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4284]  [Cited by in F6Publishing: 4185]  [Article Influence: 209.3]  [Reference Citation Analysis (0)]
74.  Venkitaraman R, Thomas K, Huddart RA, Horwich A, Dearnaley DP, Parker CC. Efficacy of low-dose dexamethasone in castration-refractory prostate cancer. BJU Int. 2008;101:440-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 36]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
75.  Adams GP, Weiner LM. Monoclonal antibody therapy of cancer. Nat Biotechnol. 2005;23:1147-1157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 822]  [Cited by in F6Publishing: 794]  [Article Influence: 41.8]  [Reference Citation Analysis (0)]
76.  Eisen T, Boshoff C, Mak I, Sapunar F, Vaughan MM, Pyle L, Johnston SR, Ahern R, Smith IE, Gore ME. Continuous low dose Thalidomide: a phase II study in advanced melanoma, renal cell, ovarian and breast cancer. Br J Cancer. 2000;82:812-817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 260]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
77.  Madhusudan S, Foster M, Muthuramalingam SR, Braybrooke JP, Wilner S, Kaur K, Han C, Hoare S, Balkwill F, Talbot DC. A phase II study of etanercept (Enbrel), a tumor necrosis factor alpha inhibitor in patients with metastatic breast cancer. Clin Cancer Res. 2004;10:6528-6534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 150]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
78.  Orlowski RZ, Kuhn DJ. Proteasome inhibitors in cancer therapy: lessons from the first decade. Clin Cancer Res. 2008;14:1649-1657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 422]  [Cited by in F6Publishing: 433]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
79.  Singh S, Khar A. Biological effects of curcumin and its role in cancer chemoprevention and therapy. Anticancer Agents Med Chem. 2006;6:259-270.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Xu XC. COX-2 inhibitors in cancer treatment and prevention, a recent development. Anticancer Drugs. 2002;13:127-137.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Khan AL, Richardson S, Drew J, Larsen F, Campbell M, Heys SD, Ah-See AK, Eremin O. Polyadenylic-polyuridylic acid enhances the natural cell-mediated cytotoxicity in patients with breast cancer undergoing mastectomy. Surgery. 1995;118:531-538.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Lacour F, Lacour J, Spira A, Michelson M, Petit JY, Delage G, Contesso G, Merlin-Nahon E, Sarrazin D, Viguier J. [Adjuvant immunotherapy using polyadenylic acid and polyuridylic acid (Poly A., Poly U.) in operable carcinoma of the breast (author's transl)]. Chirurgie. 1980;106:737-743.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Chin AI, Miyahira AK, Covarrubias A, Teague J, Guo B, Dempsey PW, Cheng G. Toll-like receptor 3-mediated suppression of TRAMP prostate cancer shows the critical role of type I interferons in tumor immune surveillance. Cancer Res. 2010;70:2595-2603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 81]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
84.  Nicodemus CF, Wang L, Lucas J, Varghese B, Berek JS. Toll-like receptor-3 as a target to enhance bioactivity of cancer immunotherapy. Am J Obstet Gynecol. 2010;202:608.e1-608.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
85.  Dicker A, Le Blanc K, Aström G, van Harmelen V, Götherström C, Blomqvist L, Arner P, Rydén M. Functional studies of mesenchymal stem cells derived from adult human adipose tissue. Exp Cell Res. 2005;308:283-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 217]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
86.  Kern S, Eichler H, Stoeve J, Klüter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells. 2006;24:1294-1301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2248]  [Cited by in F6Publishing: 2215]  [Article Influence: 123.1]  [Reference Citation Analysis (0)]
87.  In 't Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE, Kanhai HH. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22:1338-1345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 824]  [Cited by in F6Publishing: 790]  [Article Influence: 41.6]  [Reference Citation Analysis (0)]
88.  Shih DT, Lee DC, Chen SC, Tsai RY, Huang CT, Tsai CC, Shen EY, Chiu WT. Isolation and characterization of neurogenic mesenchymal stem cells in human scalp tissue. Stem Cells. 2005;23:1012-1020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 184]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
89.  Gálvez BG, Martín NS, Salama-Cohen P, Lazcano JJ, Coronado MJ, Lamelas ML, Alvarez-Barrientes A, Eiró N, Vizoso F, Rodríguez C. An adult myometrial pluripotential precursor that promotes healing of damaged muscular tissues. In Vivo. 2010;24:431-441.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Nasef A, Chapel A, Mazurier C, Bouchet S, Lopez M, Mathieu N, Sensebé L, Zhang Y, Gorin NC, Thierry D. Identification of IL-10 and TGF-beta transcripts involved in the inhibition of T-lymphocyte proliferation during cell contact with human mesenchymal stem cells. Gene Expr. 2007;13:217-226.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Ryan JM, Barry F, Murphy JM, Mahon BP. Interferon-gamma does not break, but promotes the immunosuppressive capacity of adult human mesenchymal stem cells. Clin Exp Immunol. 2007;149:353-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 461]  [Cited by in F6Publishing: 466]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
92.  Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, Borg C, Saas P, Tiberghien P, Rouas-Freiss N. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells. 2008;26:212-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 762]  [Cited by in F6Publishing: 802]  [Article Influence: 47.2]  [Reference Citation Analysis (0)]
93.  Ortiz LA, Dutreil M, Fattman C, Pandey AC, Torres G, Go K, Phinney DG. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci U S A. 2007;104:11002-11007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 762]  [Cited by in F6Publishing: 751]  [Article Influence: 44.2]  [Reference Citation Analysis (0)]
94.  English K, Barry FP, Field-Corbett CP, Mahon BP. IFN-gamma and TNF-alpha differentially regulate immunomodulation by murine mesenchymal stem cells. Immunol Lett. 2007;110:91-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 314]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
95.  Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, Hardy W, Devine S, Ucker D, Deans R. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol. 2002;30:42-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1730]  [Cited by in F6Publishing: 1613]  [Article Influence: 73.3]  [Reference Citation Analysis (0)]
96.  Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, Gianni AM. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood. 2002;99:3838-3843.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Tse WT, Pendleton JD, Beyer WM, Egalka MC, Guinan EC. Suppression of allogeneic T-cell proliferation by human marrow stromal cells: implications in transplantation. Transplantation. 2003;75:389-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1141]  [Cited by in F6Publishing: 1067]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
98.  Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD, Mao N. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 2005;105:4120-4126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 963]  [Cited by in F6Publishing: 954]  [Article Influence: 50.2]  [Reference Citation Analysis (0)]
99.  Li YP, Paczesny S, Lauret E, Poirault S, Bordigoni P, Mekhloufi F, Hequet O, Bertrand Y, Ou-Yang JP, Stoltz JF. Human mesenchymal stem cells license adult CD34+ hemopoietic progenitor cells to differentiate into regulatory dendritic cells through activation of the Notch pathway. J Immunol. 2008;180:1598-1608.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE. Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells. J Immunol. 2006;177:2080-2087.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Ramasamy R, Fazekasova H, Lam EW, Soeiro I, Lombardi G, Dazzi F. Mesenchymal stem cells inhibit dendritic cell differentiation and function by preventing entry into the cell cycle. Transplantation. 2007;83:71-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
102.  Augello A, Tasso R, Negrini SM, Cancedda R, Pennesi G. Cell therapy using allogeneic bone marrow mesenchymal stem cells prevents tissue damage in collagen-induced arthritis. Arthritis Rheum. 2007;56:1175-1186.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Parekkadan B, Tilles AW, Yarmush ML. Bone marrow-derived mesenchymal stem cells ameliorate autoimmune enteropathy independently of regulatory T cells. Stem Cells. 2008;26:1913-1919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 124]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
104.  Ball L, Bredius R, Lankester A, Schweizer J, van den Heuvel-Eibrink M, Escher H, Fibbe W, Egeler M. Third party mesenchymal stromal cell infusions fail to induce tissue repair despite successful control of severe grade IV acute graft-versus-host disease in a child with juvenile myelo-monocytic leukemia. Leukemia. 2008;22:1256-1257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
105.  Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, Lanino E, Sundberg B, Bernardo ME, Remberger M. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371:1579-1586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2047]  [Cited by in F6Publishing: 1967]  [Article Influence: 122.9]  [Reference Citation Analysis (0)]
106.  Le Blanc K, Rasmusson I, Sundberg B, Götherström C, Hassan M, Uzunel M, Ringdén O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363:1439-1441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2044]  [Cited by in F6Publishing: 1970]  [Article Influence: 98.5]  [Reference Citation Analysis (0)]
107.  Müller I, Kordowich S, Holzwarth C, Isensee G, Lang P, Neunhoeffer F, Dominici M, Greil J, Handgretinger R. Application of multipotent mesenchymal stromal cells in pediatric patients following allogeneic stem cell transplantation. Blood Cells Mol Dis. 2008;40:25-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 152]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
108.  Ning H, Yang F, Jiang M, Hu L, Feng K, Zhang J, Yu Z, Li B, Xu C, Li Y. The correlation between cotransplantation of mesenchymal stem cells and higher recurrence rate in hematologic malignancy patients: outcome of a pilot clinical study. Leukemia. 2008;22:593-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 259]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
109.  Ringdén O, Uzunel M, Rasmusson I, Remberger M, Sundberg B, Lönnies H, Marschall HU, Dlugosz A, Szakos A, Hassan Z. Mesenchymal stem cells for treatment of therapy-resistant graft-versus-host disease. Transplantation. 2006;81:1390-1397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
110.  Nakamizo A, Marini F, Amano T, Khan A, Studeny M, Gumin J, Chen J, Hentschel S, Vecil G, Dembinski J. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005;65:3307-3318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 780]  [Cited by in F6Publishing: 791]  [Article Influence: 41.6]  [Reference Citation Analysis (0)]
111.  Zhu Y, Sun Z, Han Q, Liao L, Wang J, Bian C, Li J, Yan X, Liu Y, Shao C. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia. 2009;23:925-933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 239]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
112.  Mishra A, Bharti AC, Varghese P, Saluja D, Das BC. Differential expression and activation of NF-kappaB family proteins during oral carcinogenesis: Role of high risk human papillomavirus infection. Int J Cancer. 2006;119:2840-2850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 105]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
113.  Rhodus NL, Cheng B, Myers S, Bowles W, Ho V, Ondrey F. A comparison of the pro-inflammatory, NF-kappaB-dependent cytokines: TNF-alpha, IL-1-alpha, IL-6, and IL-8 in different oral fluids from oral lichen planus patients. Clin Immunol. 2005;114:278-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 121]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
114.  Rhodus NL, Ho V, Miller CS, Myers S, Ondrey F. NF-kappaB dependent cytokine levels in saliva of patients with oral preneoplastic lesions and oral squamous cell carcinoma. Cancer Detect Prev. 2005;29:42-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 146]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
115.  O'Neil BH, Bůzková P, Farrah H, Kashatus D, Sanoff H, Goldberg RM, Baldwin AS, Funkhouser WK. Expression of nuclear factor-kappaB family proteins in hepatocellular carcinomas. Oncology. 2007;72:97-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
116.  Tai DI, Tsai SL, Chang YH, Huang SN, Chen TC, Chang KS, Liaw YF. Constitutive activation of nuclear factor kappaB in hepatocellular carcinoma. Cancer. 2000;89:2274-2281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
117.  Logan RM, Gibson RJ, Sonis ST, Keefe DM. Nuclear factor-kappaB (NF-kappaB) and cyclooxygenase-2 (COX-2) expression in the oral mucosa following cancer chemotherapy. Oral Oncol. 2007;43:395-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 109]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
118.  Ling FC, Baldus SE, Khochfar J, Xi H, Neiss S, Brabender J, Metzger R, Drebber U, Dienes HP, Bollschweiler E. Association of COX-2 expression with corresponding active and chronic inflammatory reactions in Barrett's metaplasia and progression to cancer. Histopathology. 2007;50:203-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
119.  Karamitopoulou E, Tornillo L, Zlobec I, Cioccari L, Carafa V, Borner M, Schaffner T, Brunner T, Diamantis I, Zimmermann A. Clinical significance of cell cycle- and apoptosis-related markers in biliary tract cancer: a tissue microarray-based approach revealing a distinctive immunophenotype for intrahepatic and extrahepatic cholangiocarcinomas. Am J Clin Pathol. 2008;130:780-786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 29]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
120.  Gao YW, Chen YX, Wang ZM, Zhou LD, Li XY, Li LX, Luo QZ, Tian W, Fu CY, Zhou JH. Correlation between expression of cyclooxygenase-2 and the presence of CD4+ infiltrating T-lymphocyte in human primary hepatocellular carcinoma. Hepatogastroenterology. 2008;55:345-350.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  El-Bassiouny AE, Zoheiry MM, Nosseir MM, El-Ahwany EG, Ibrahim RA, El-Bassiouni NE. Expression of cyclooxygenase-2 and transforming growth factor-beta1 in HCV-induced chronic liver disease and hepatocellular carcinoma. MedGenMed. 2007;9:45.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Niijima M, Yamaguchi T, Ishihara T, Hara T, Kato K, Kondo F, Saisho H. Immunohistochemical analysis and in situ hybridization of cyclooxygenase-2 expression in intraductal papillary-mucinous tumors of the pancreas. Cancer. 2002;94:1565-1573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
123.  Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S, DuBois RN. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology. 1994;107:1183-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 184]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
124.  Joo YE, Oh WT, Rew JS, Park CS, Choi SK, Kim SJ. Cyclooxygenase-2 expression is associated with well-differentiated and intestinal-type pathways in gastric carcinogenesis. Digestion. 2002;66:222-229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
125.  Yamagata R, Shimoyama T, Fukuda S, Yoshimura T, Tanaka M, Munakata A. Cyclooxygenase-2 expression is increased in early intestinal-type gastric cancer and gastric mucosa with intestinal metaplasia. Eur J Gastroenterol Hepatol. 2002;14:359-363.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Ma XT, Wang S, Ye YJ, Du RY, Cui ZR, Somsouk M. Constitutive activation of Stat3 signaling pathway in human colorectal carcinoma. World J Gastroenterol. 2004;10:1569-1573.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Kanda N, Seno H, Konda Y, Marusawa H, Kanai M, Nakajima T, Kawashima T, Nanakin A, Sawabu T, Uenoyama Y. STAT3 is constitutively activated and supports cell survival in association with survivin expression in gastric cancer cells. Oncogene. 2004;23:4921-4929.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 241]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
128.  Zhang H, Wang S, Zhang YC, Ye YJ, Cui ZR, Fang WG. [Correlation between Stat3 signal transduction pathway and expression of cyclooxygenase-2 in colorectal cancer cells]. Zhonghua Yi Xue Za Zhi. 2005;85:2899-2904.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Dvorakova K, Payne CM, Ramsey L, Holubec H, Sampliner R, Dominguez J, Dvorak B, Bernstein H, Bernstein C, Prasad A. Increased expression and secretion of interleukin-6 in patients with Barrett's esophagus. Clin Cancer Res. 2004;10:2020-2028.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Kinoshita T, Ito H, Miki C. Serum interleukin-6 level reflects the tumor proliferative activity in patients with colorectal carcinoma. Cancer. 1999;85:2526-2531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
131.  Komoda H, Tanaka Y, Honda M, Matsuo Y, Hazama K, Takao T. Interleukin-6 levels in colorectal cancer tissues. World J Surg. 1998;22:895-898.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Wilson KT, Fu S, Ramanujam KS, Meltzer SJ. Increased expression of inducible nitric oxide synthase and cyclooxygenase-2 in Barrett's esophagus and associated adenocarcinomas. Cancer Res. 1998;58:2929-2934.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Hennig R, Grippo P, Ding XZ, Rao SM, Buchler MW, Friess H, Talamonti MS, Bell RH, Adrian TE. 5-Lipoxygenase, a marker for early pancreatic intraepithelial neoplastic lesions. Cancer Res. 2005;65:6011-6016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 68]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
134.  Izzo JG, Malhotra U, Wu TT, Luthra R, Correa AM, Swisher SG, Hofstetter W, Chao KS, Hung MC, Ajani JA. Clinical biology of esophageal adenocarcinoma after surgery is influenced by nuclear factor-kappaB expression. Cancer Epidemiol Biomarkers Prev. 2007;16:1200-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
135.  Weichert W, Boehm M, Gekeler V, Bahra M, Langrehr J, Neuhaus P, Denkert C, Imre G, Weller C, Hofmann HP. High expression of RelA/p65 is associated with activation of nuclear factor-kappaB-dependent signaling in pancreatic cancer and marks a patient population with poor prognosis. Br J Cancer. 2007;97:523-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 117]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
136.  Asakawa M, Kono H, Amemiya H, Matsuda M, Suzuki T, Maki A, Fujii H. Role of interleukin-18 and its receptor in hepatocellular carcinoma associated with hepatitis C virus infection. Int J Cancer. 2006;118:564-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
137.  Levidou G, Korkolopoulou P, Nikiteas N, Tzanakis N, Thymara I, Saetta AA, Tsigris C, Rallis G, Vlasis K, Patsouris E. Expression of nuclear factor kappaB in human gastric carcinoma: relationship with I kappaB a and prognostic significance. Virchows Arch. 2007;450:519-527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
138.  Wu L, Pu Z, Feng J, Li G, Zheng Z, Shen W. The ubiquitin-proteasome pathway and enhanced activity of NF-kappaB in gastric carcinoma. J Surg Oncol. 2008;97:439-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
139.  Yamanaka N, Sasaki N, Tasaki A, Nakashima H, Kubo M, Morisaki T, Noshiro H, Yao T, Tsuneyoshi M, Tanaka M. Nuclear factor-kappaB p65 is a prognostic indicator in gastric carcinoma. Anticancer Res. 2004;24:1071-1075.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Voboril R, Voborilova J, Rychterova V, Jirasek T, Dvorak J. Dissociated invasively growing cancer cells with NF-kappaB/p65 positivity after radiotherapy: a new marker for worse clinical outcome in rectal cancer? Preliminary data. Clin Exp Metastasis. 2008;25:491-496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
141.  Chang BW, Kim DH, Kowalski DP, Burleson JA, Son YH, Wilson LD, Haffty BG. Prognostic significance of cyclooxygenase-2 in oropharyngeal squamous cell carcinoma. Clin Cancer Res. 2004;10:1678-1684.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Alici S, Ugras S, Bayram I, Izmirli M. Prognostic factors and COX-2 expression in advanced stage esophageal squamous cell carcinoma. Adv Ther. 2006;23:672-679.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  France M, Drew PA, Dodd T, Watson DI. Cyclo-oxygenase-2 expression in esophageal adenocarcinoma as a determinant of clinical outcome following esophagectomy. Dis Esophagus. 2004;17:136-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
144.  Liu B, Ren Z, Shi Y, Guan C, Pan Z, Zong Z. Activation of signal transducers and activators of transcription 3 and overexpression of its target gene CyclinD1 in laryngeal carcinomas. Laryngoscope. 2008;118:1976-1980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
145.  Yang GZ, Li L, Ding HY, Zhou JS. Cyclooxygenase-2 is over-expressed in Chinese esophageal squamous cell carcinoma, and correlated with NF-kappaB: an immunohistochemical study. Exp Mol Pathol. 2005;79:214-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
146.  Yoshikawa R, Fujiwara Y, Koishi K, Kojima S, Matsumoto T, Yanagi H, Yamamura T, Hashimoto-Tamaoki T, Nishigami T, Tsujimura T. Cyclooxygenase-2 expression after preoperative chemoradiotherapy correlates with more frequent esophageal cancer recurrence. World J Gastroenterol. 2007;13:2283-2288.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Santini D, Vincenzi B, Tonini G, Scarpa S, Vasaturo F, Malacrino C, Vecchio F, Borzomati D, Valeri S, Coppola R. Cyclooxygenase-2 overexpression is associated with a poor outcome in resected ampullary cancer patients. Clin Cancer Res. 2005;11:3784-3789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
148.  Konno H, Baba M, Shoji T, Ohta M, Suzuki S, Nakamura S. Cyclooxygenase-2 expression correlates with uPAR levels and is responsible for poor prognosis of colorectal cancer. Clin Exp Metastasis. 2002;19:527-534.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Masunaga R, Kohno H, Dhar DK, Ohno S, Shibakita M, Kinugasa S, Yoshimura H, Tachibana M, Kubota H, Nagasue N. Cyclooxygenase-2 expression correlates with tumor neovascularization and prognosis in human colorectal carcinoma patients. Clin Cancer Res. 2000;6:4064-4068.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Nakamoto RH, Uetake H, Iida S, Kolev YV, Soumaoro LT, Takagi Y, Yasuno M, Sugihara K. Correlations between cyclooxygenase-2 expression and angiogenic factors in primary tumors and liver metastases in colorectal cancer. Jpn J Clin Oncol. 2007;37:679-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
151.  Ogino S, Kirkner GJ, Nosho K, Irahara N, Kure S, Shima K, Hazra A, Chan AT, Dehari R, Giovannucci EL. Cyclooxygenase-2 expression is an independent predictor of poor prognosis in colon cancer. Clin Cancer Res. 2008;14:8221-8227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 160]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
152.  Soumaoro LT, Uetake H, Higuchi T, Takagi Y, Enomoto M, Sugihara K. Cyclooxygenase-2 expression: a significant prognostic indicator for patients with colorectal cancer. Clin Cancer Res. 2004;10:8465-8471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 202]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
153.  Shi H, Xu JM, Hu NZ, Xie HJ. Prognostic significance of expression of cyclooxygenase-2 and vascular endothelial growth factor in human gastric carcinoma. World J Gastroenterol. 2003;9:1421-1426.  [PubMed]  [DOI]  [Cited in This Article: ]
154.  Petersen S, Haroske G, Hellmich G, Ludwig K, Petersen C, Eicheler W. COX-2 expression in rectal carcinoma: immunohistochemical pattern and clinical outcome. Anticancer Res. 2002;22:1225-1230.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Kaifi JT, Yekebas EF, Schurr P, Obonyo D, Wachowiak R, Busch P, Heinecke A, Pantel K, Izbicki JR. Tumor-cell homing to lymph nodes and bone marrow and CXCR4 expression in esophageal cancer. J Natl Cancer Inst. 2005;97:1840-1847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 169]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
156.  Ottaiano A, Franco R, Aiello Talamanca A, Liguori G, Tatangelo F, Delrio P, Nasti G, Barletta E, Facchini G, Daniele B. Overexpression of both CXC chemokine receptor 4 and vascular endothelial growth factor proteins predicts early distant relapse in stage II-III colorectal cancer patients. Clin Cancer Res. 2006;12:2795-2803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 136]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
157.  Yoshitake N, Fukui H, Yamagishi H, Sekikawa A, Fujii S, Tomita S, Ichikawa K, Imura J, Hiraishi H, Fujimori T. Expression of SDF-1 alpha and nuclear CXCR4 predicts lymph node metastasis in colorectal cancer. Br J Cancer. 2008;98:1682-1689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 108]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
158.  Li YH, Hu CF, Shao Q, Huang MY, Hou JH, Xie D, Zeng YX, Shao JY. Elevated expressions of survivin and VEGF protein are strong independent predictors of survival in advanced nasopharyngeal carcinoma. J Transl Med. 2008;6:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 95]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
159.  Kimura S, Kitadai Y, Tanaka S, Kuwai T, Hihara J, Yoshida K, Toge T, Chayama K. Expression of hypoxia-inducible factor (HIF)-1alpha is associated with vascular endothelial growth factor expression and tumour angiogenesis in human oesophageal squamous cell carcinoma. Eur J Cancer. 2004;40:1904-1912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
160.  Zhang B, Zhao WH, Zhou WY, Yu WS, Yu JM, Li S. Expression of vascular endothelial growth factors-C and -D correlate with evidence of lymphangiogenesis and angiogenesis in pancreatic adenocarcinoma. Cancer Detect Prev. 2007;31:436-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
161.  Jia JB, Zhuang PY, Sun HC, Zhang JB, Zhang W, Zhu XD, Xiong YQ, Xu HX, Tang ZY. Protein expression profiling of vascular endothelial growth factor and its receptors identifies subclasses of hepatocellular carcinoma and predicts survival. J Cancer Res Clin Oncol. 2009;135:847-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
162.  Dassoulas K, Gazouli M, Rizos S, Theodoropoulos G, Christoni Z, Nikiteas N, Karakitsos P. Common polymorphisms in the vascular endothelial growth factor gene and colorectal cancer development, prognosis, and survival. Mol Carcinog. 2009;48:563-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 71]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
163.  Kim JG, Chae YS, Sohn SK, Cho YY, Moon JH, Park JY, Jeon SW, Lee IT, Choi GS, Jun SH. Vascular endothelial growth factor gene polymorphisms associated with prognosis for patients with colorectal cancer. Clin Cancer Res. 2008;14:62-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 100]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
164.  Tang H, Wang J, Bai F, Zhai H, Gao J, Hong L, Xie H, Zhang F, Lan M, Yao W. Positive correlation of osteopontin, cyclooxygenase-2 and vascular endothelial growth factor in gastric cancer. Cancer Invest. 2008;26:60-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
165.  Chen XL, Wang LC, Zhang WG, Chen XY, Sun ZM. [Correlations of S100A4 and MMP9 expressions to infiltration, metastasis and prognosis of non-small cell lung cancer]. Nan Fang Yi Ke Da Xue Xue Bao. 2008;28:1254-1258.  [PubMed]  [DOI]  [Cited in This Article: ]
166.  Kai H, Kitadai Y, Kodama M, Cho S, Kuroda T, Ito M, Tanaka S, Ohmoto Y, Chayama K. Involvement of proinflammatory cytokines IL-1beta and IL-6 in progression of human gastric carcinoma. Anticancer Res. 2005;25:709-713.  [PubMed]  [DOI]  [Cited in This Article: ]
167.  Chung YC, Chaen YL, Hsu CP. Clinical significance of tissue expression of interleukin-6 in colorectal carcinoma. Anticancer Res. 2006;26:3905-3911.  [PubMed]  [DOI]  [Cited in This Article: ]
168.  Ito H, Miki C. Profile of circulating levels of interleukin-1 receptor antagonist and interleukin-6 in colorectal cancer patients. Scand J Gastroenterol. 1999;34:1139-1143.  [PubMed]  [DOI]  [Cited in This Article: ]
169.  Haraguchi M, Komuta K, Akashi A, Matsuzaki S, Furui J, Kanematsu T. Elevated IL-8 levels in the drainage vein of resectable Dukes' C colorectal cancer indicate high risk for developing hepatic metastasis. Oncol Rep. 2002;9:159-165.  [PubMed]  [DOI]  [Cited in This Article: ]
170.  Rubie C, Frick VO, Pfeil S, Wagner M, Kollmar O, Kopp B, Graber S, Rau BM, Schilling MK. Correlation of IL-8 with induction, progression and metastatic potential of colorectal cancer. World J Gastroenterol. 2007;13:4996-5002.  [PubMed]  [DOI]  [Cited in This Article: ]
171.  Terada H, Urano T, Konno H. Association of interleukin-8 and plasminogen activator system in the progression of colorectal cancer. Eur Surg Res. 2005;37:166-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 36]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
172.  Kubo F, Ueno S, Hiwatashi K, Sakoda M, Kawaida K, Nuruki K, Aikou T. Interleukin 8 in human hepatocellular carcinoma correlates with cancer cell invasion of vessels but not with tumor angiogenesis. Ann Surg Oncol. 2005;12:800-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 58]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
173.  Kido S, Kitadai Y, Hattori N, Haruma K, Kido T, Ohta M, Tanaka S, Yoshihara M, Sumii K, Ohmoto Y. Interleukin 8 and vascular endothelial growth factor -- prognostic factors in human gastric carcinomas? Eur J Cancer. 2001;37:1482-1487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 111]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
174.  Hu ZL, Wen JF, Shen M, Liu Y. [Expressions of TGIF, MMP9 and VEGF proteins and their clinicopathological relationship in gastric cancer]. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2006;31:70-74.  [PubMed]  [DOI]  [Cited in This Article: ]
175.  Gu ZD, Li JY, Li M, Gu J, Shi XT, Ke Y, Chen KN. Matrix metalloproteinases expression correlates with survival in patients with esophageal squamous cell carcinoma. Am J Gastroenterol. 2005;100:1835-1843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 70]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
176.  Wang L, Tang Z, Sun H. [Nitric oxide synthase and vascular endothelial growth factor expression in hepatocellular carcinoma and their relation to angiogenesis]. Zhonghua Zhong Liu Za Zhi. 2000;22:301-303.  [PubMed]  [DOI]  [Cited in This Article: ]
177.  Rajnakova A, Moochhala S, Goh PM, Ngoi S. Expression of nitric oxide synthase, cyclooxygenase, and p53 in different stages of human gastric cancer. Cancer Lett. 2001;172:177-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]