Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 7, 2014; 20(37): 13246-13257
Published online Oct 7, 2014. doi: 10.3748/wjg.v20.i37.13246
Impact of proteolytic enzymes in colorectal cancer development and progression
László Herszényi, Zsolt Tulassay, 2nd Department of Medicine, Semmelweis University, H-1088 Budapest, Hungary
Loránd Barabás, Gábor István, 2nd Department of Surgery, Semmelweis University, H-1024 Budapest, Hungary
István Hritz, 1st Department of Medicine, Fejér Megyei Szent György University Teaching Hospital, H-8000 Székesfehérvár, Hungary
Author contributions: Herszényi L, Barabás L, Hritz I, István G and Tulassay Z contributed equally to this work; Herszényi L, Barabás L and Hritz I wrote the paper; István G contributed to the revision of the manuscript; Tulassay Z critically revised the manuscript for important intellectual content.
Correspondence to: László Herszényi, MD, DSc, 2nd Department of Medicine, Semmelweis University, Szentkirályi str. 46, H-1088 Budapest, Hungary. herszenyi.laszlo@med.semmelweis-univ.hu
Telephone: +36-1-2660926  Fax: +36-1-2660816
Received: October 26, 2013
Revised: January 26, 2014
Accepted: May 23, 2014
Published online: October 7, 2014

Abstract

Tumor invasion and metastasis is a highly complicated, multi-step phenomenon. In the complex event of tumor progression, tumor cells interact with basement membrane and extracellular matrix components. Proteolytic enzymes (proteinases) are involved in the degradation of extracellular matrix, but also in cancer invasion and metastasis. The four categories of proteinases (cysteine-, serine-, aspartic-, and metalloproteinases) are named and classified according to the essential catalytic component in their active site. We and others have shown that proteolytic enzymes play a major role not only in colorectal cancer (CRC) invasion and metastasis, but also in malignant transformation of precancerous lesions into cancer. Tissue and serum-plasma antigen concentrations of proteinases might be of great value in identifying patients with poor prognosis in CRC. Our results, in concordance with others indicate the potential tumor marker impact of proteinases for the early diagnosis of CRC. In addition, proteinases may also serve as potential target molecules for therapeutic agents.

Key Words: Proteinase, Cathepsin, Plasminogen activator, Matrix metalloproteinase, Colorectal cancer, Adenoma, Invasion, Metastasis, Biomarker, Prognosis

Core tip: Tumor invasion and metastasis is a highly complex phenomenon. Proteolytic enzymes (proteinases) are involved in the degradation of extracellular matrix, in colorectal cancer (CRC) invasion and metastasis, as well as in the malignant transformation of colorectal adenomas. Tissue and serum-plasma antigen concentrations of proteinases are strong prognostic factors in CRC and may have tumor marker impact for early diagnosis. Proteolytic enzymes may serve as potential target molecules for CRC therapy. Their use in combination with established chemotherapeutic strategies might have the potential to become a valuable oncological treatment modality.



MECHANISMS OF TUMOR PROGRESSION

Tumor invasion and metastasis is a highly complicated, multi-step phenomenon. The multistep metastatic process requires the actions of several genes and involves the initial transforming event (i.e., oncogene activation), proliferation of transformed cells and the ability of tumor cells to avoid destruction by immune-mechanisms. Furthermore, it comprises of nutrition supply to the tumor mass by the release of tumor angiogenesis factors, cancer cell local invasion and destruction of extracellular matrix (ECM) components, epithelial mesenchymal transition (EMT), shedding from primary tumor, intravasation, arrest, extravasation and colonization at a preferential site resulting in the formation of a secondary tumor (distant metastases)[1-9].

Chronic and persistent inflammation can predispose to carcinogenesis and contribute to cancer development. Cancer-associated inflammation includes infiltrating leucocytes, cytokines, chemokines, growth factors and matrix-degrading enzymes. Inflammatory conditions can initiate oncogenic transformation and epigenetic and genetic changes in malignant cells. In essence, two interrelated pathways connect inflammation and cancer: (1) genetic alterations (including chromosomal amplification, activation of oncogenes and inactivation of tumor-suppressor genes) leading to neoplastic transformation; and (2) presence of tumor-infiltrating leukocytes that are prime regulators of cancer-related inflammation. The integration of these two pathways activates transcription factors and finally creates a tumor-associated inflammatory milieu[10-19].

BASEMENT MEMBRANE, EXTRACELLULAR MATRIX AND CANCER CELLS

All normal or pre-invasive tumor epithelia are physically segregated from vascular structures within the stroma by the basement membrane (BM). The BM consists of laminins, type IV collagen and surrounding epithelial cells. The tumor stroma is comprised of ECM, non-malignant cells, and the signalling molecules they produce. During tumor invasion and metastasis, tumor cells are interacting with the BM and the ECM. The disruption of the BM and the ECM is an essential pre-requisite for cancer cell invasion and metastasis. Interaction of tumor cells with the BM and ECM comprises of three steps: attachment, matrix dissolution and migration. The first step is tumor cell attachment to the matrix. The attachment is mediated by tumor cell surface receptors, when tumor cells bind to BM surface. This process involves specific glycoproteins such as fibronectin, type IV collagen and laminin. In the second step tumor cells directly secrete degradative enzymes or induce the host to produce proteolytic enzymes to degrade ECM. The matrix lysis takes place in a highly localized region close to the tumor cell surface. During the third step (migration), cancer cells are propelled across the BM and stroma through the zone of matrix proteolysis. Invasion of ECM is accomplished by reverberation of these three steps[3,6,9,20-27].

GENERAL ROLES OF PROTEOLYTIC ENZYMES IN CANCERS

The four categories of proteinases (cysteine-, serine-, aspartic-, and metalloproteinases) are named and classified according to the essential catalytic component (usually an amino acid) in their active site[3,28]. Table 1 summarises the four categories and some general properties of each group. Proteolytic enzymes play a major role in the breakdown and reconstitution of ECM in a variety of physiological and pathological processes, such as protein turnover, tissue remodeling, wound repair, angiogenesis, destructive diseases, inflammatory disorders as well as tumor invasion and metastasis (Table 2). Tumor cells have been shown to produce and release several proteolytic enzymes, which are thought to be involved in tumor invasion and metastasis. Proteolytic enzymes are also frequently produced by surrounding stromal cells, including fibroblasts and inflammatory cells. It has been proposed that these proteolytic enzymes cause degradation or disruption of BM and ECM components allowing the in situ cancer cells to migrate into the adjacent stroma or to disseminate to distant organ. It is commonly accepted that progression from in situ to invasive or metastatic cancer is caused by proteolytic enzymes (proteinases) produced by tumor cells that increase linearly in concentration with tumor progression[3,6,9,29-31].

Table 1 Categories and main properties of proteolytic enzymes.
PropertyCysteine proteasesSerine proteasesAspartic proteasesMetallo proteases
Active siteCysteineSerineAspartic acidZinc
Optimum pH7-93-72-45-9
LocationLysosomesIntra-extracellularLysosomesIntra-extracellular
ExamplesCathepsinsElastasePepsinogen I-IICollagenase
B, L, C, HTrypsinCathepsins D, EStromelysin
PA: uPA, tPAGelatinaseMatrilysin
InhibitorsCystatinAntitrypsinUnknownTIMP
Antithrombin III
PAI
Table 2 General roles of proteolytic enzymes in cancers.
Degradation or disruption of basement membrane and extracellular matrix
Produce components which allow the in situ cancer cells to disseminate to distant organ
Formation of a complex microenvironment that promotes malignant transformation
Activation of growth factors, adhesion molecules
Suppression of tumor cell apoptosis
Destruction of chemokine gradients
Modulation of antitumor immune reactions
Dual and complex role in angiogenesis

The impact of proteolytic enzymes in tumor progression is much more complex than that derived from their direct degradative action on BM and ECM components. Now they are known to have functions that extend far outside matrix remodeling. Proteolytic processing of bioactive molecules by proteinases contributes to the formation of a complex microenvironment that promotes malignant transformation. Proteolytic enzymes can contribute to tumor growth either directly or indirectly via growth factors such as transforming growth factor-β (TGF-β), basic fibroblast growth factor (bFGF) or, insulin-like growth factor (IGF). Proteinases also act on other non-matrix substrates (e.g., chemokines, adhesion molecules, apoptotic mediators, angiogenic factors) that yield the critical cellular responses that are essential for tumor growth and progression. Proteolytic enzymes are also associated with a variety of escape mechanisms that tumor cells develop to avoid immune response including chemokine cleavage and regulation of chemokine mobilization. Tumor cells produce chemokines, cytokines, and the extracellular matrix metalloprotease inducer (EMMPRIN), which in turn activates tumor-cell invasion. During angiogenesis, proteolytic enzymes can have both a pro-angiogenic impact, by releasing matrix-bound pro-angiogenic factors such as TGF-β, bFGF, triggering the angiogenic switch during carcinogenesis and facilitating vascular remodeling and neovascularization at distant sites during metastasis, and also may have anti-angiogenic role, by cleaving the ECM components into anti-angiogenic factors[32-36].

Cysteine and serine proteinases

It has been observed that cysteine proteinases [cathepsin B (CATB) and cathepsin L (CATL)], play a crucial role in the destruction of various elements of the cell-surrounding ECM. The serine proteinase urokinase-type plasminogen inhibitor (uPA) is also involved in many protein degrading processes. uPA seems to promote invasion through a plasmin mediated degradation of ECM proteins. Active uPA catalyzes the conversion from plasminogen to plasmin, which is a potent activator of several metalloproteinase proenzymes, such as prostromelysin, procollagenase, and progelatinase B. Beyond its direct proteolytic capacity, CATB has also been shown to activate the prourokinase-type plasminogen activator (pro-uPA). The tissue-type plasminogen activator (TPA) is a key enzyme in the fibrinolytic cascade. Plasminogen activators (PA) are controlled by plasminogen activator inhibitors, which are members of the serine proteinase inhibitors (serpin) family. The PA inhibitor type-1 (PAI-1) under normal physiologic conditions inhibits both uPA and TPA. The exact role of PAI-1 in tumor biology is complex: PAI-1 may represent a specific protein of transformed malignant tissue; may protect cancer tissue against the proteolytic degradation triggered by the tumor on surrounding normal tissue; and finally, PAI-1 has a role in angiogenesis playing a part in tumor spread[37-45].

Cathepsins and components of the plasminogen activator and inhibitor system have been demonstrated in various malignant tissues, e.g., breast cancer[46-48], lung cancer[49,50], head and neck cancer[51], ovarian cancer[52] or gastric cancer[53-56] and might therefore be useful as a diagnostic tool.

With respect to the gastrointestinal (GI) tract, we have previously shown that cysteine and serine proteinases are widely distributed in GI tissues, being implicated in processes of GI tissue remodelling, angiogenesis, wound healing, inflammation, may have a role not only in the process of esophageal or gastric cancer invasion, but also in the progression of GI precancerous lesions into cancer[57-61]. Several studies, along with our own, have also pointed to the prognostic value of cysteine and serine proteinases for survival, for instance, in gastric cancer[56,58,59,62,63], pancreatic cancer[64], or hepatocellular carcinoma[65].

Matrix metalloproteinases

Matrix metalloproteinases (MMPs) family consists of 26 members of homologous zinc-dependent endopeptidases. The expression of MMPs is induced by a variety of external stimuli such as cytokines and growth factors, including interleukins, interferons, vascular endothelial growth factor, fibroblast growth factor (FGF), tumor necrosis factor-alpha (TNF-α) or TNF-β, and EMMPRIN. MMPs play an important role in the degradation of ECM components, are crucial for tumor growth, invasion and metastasis. MMPs are synthesized as inactive zymogens, which are then activated by either other MMPs or by serine proteinases. Based on substrate specificities and sequence homology, MMPs can be classified as gelatinases, collagenases, stromelysins and matrilysins. MMPs are responsible for cleaving all of the major ECM proteins[66-74].

We and others have shown that MMPs, particularly type IV collagenase MMP-9 (gelatinase B), are essential in the process of tumor invasion and metastasis, as well as in the remodeling and inflammatory processes in IBD[75-82].

MMPs have also been considered as potential diagnostic and prognostic biomarkers in many types and stages of GI cancer[83-85].

Tissue inhibitors of matrix metalloproteinases

MMPs activity is specifically inhibited by natural inhibitors, called tissue inhibitors of metalloproteinases (TIMPs). Currently, four different TIMPs have been characterized in humans (TIMP-1, -2, -3 and -4). The balanced interaction of MMPs with TIMPs regulates ECM homeostasis. The imbalance between MMPs and TIMPs is an essential step in the development of malignancies. TIMPs might display a dual influence on tumor progression: either beneficial by inhibiting MMPs and impairing angiogenesis or harmful by facilitating cancer cell generation, tumor growth and metastasis. The sensitive balance between MMPs and TIMPs is essential for many physiological processes in the gut[6,86-91].

We have recently demonstrated that not only MMPs but also TIMPs may contribute to the inflammatory and remodeling processes in IBD and serum TIMP-1 might be useful as additional biomarker in the assessment of IBD activity[82].

IMPACT OF PROTEOLYTIC ENZYMES IN COLORECTAL CANCER

colorectal cancer (CRC) is the third most common malignant neoplasm worldwide. CRC is the second most common newly diagnosed cancer and the second most common cause of cancer death in the European Union (EU). Despite the advances in screening, diagnosis, and treatment, the overall long-term outcome of curatively resected patients has not significantly changed in the last decades, the five-year survival rate being approximately 60%. More than a half of CRCs are still diagnosed only when the disease involves regional or distant organs, and these patients are candidates for systemic chemotherapy. The prognosis of CRC is determined primarily by TNM staging and pathomorphological parameters. Furthermore, therapeutic strategies for chemotherapy are based on traditional prognostic systems. For risk stratification, it would be useful for clinicians to have new and more efficient preoperative tumor markers and prognostic indicators available, for instance to better identify patients who need adjuvant or neoadjuvant treatment[92-97].

We review hereinafter the prognostic value and potential tumor marker impact of a number of proteolytic enzymes. We also discuss proteinases as potential target molecules for therapeutic agents Table 3 summarises the clinical significance of proteolytic enzymes in CRC.

Table 3 Clinical significance of proteolytic enzymes in colorectal cancer.
Role in colorectal tumor biology, in the process of tumor invasion and metastasis
Role in malignant transformation of colorectal precancerous conditions and lesions
Potential diagnostic tool
New and independent prognostic factors
Potential tumor marker impact for early diagnosis
Potential target molecules for therapeutic agents
Tissue expression of cysteine and serine proteinases in CRC

Cathepsins or components of the plasminogen activator and inhibitor system have been demonstrated in colorectal cancer tissue and might therefore be useful as diagnostic tools[98-100]. Some of the studies also showed that cathepsins or some of PAs also have a prognostic significance for patients with CRC[98-106]. In a former study we have demonstrated the simultaneous up-regulation of both cysteine and serine proteinases in CRC confirming their role in colorectal tumor biology and particularly in the process of tumor invasion and metastasis. We have also shown that CATL, uPA and PAI-1 have a major prognostic impact in patients with CRC[107].

Serum and plasma concentrations of cysteine and serine proteinases in CRC

Given the lack in the literature of a comparison of the behavior of cysteine proteinase CATB and serine proteinase uPA in the same experimental setting in different GI tumors, in a previous study we have surveyed the possible clinical impact of serum CATB and plasma UPA antigen in CRC, gastric cancer, hepatocellular carcinoma, pancreatic cancer, and colorectal adenomas[61]. We have demonstrated that preoperative serum CATB and plasma uPA antigen concentrations were significantly higher in GI tract cancers overall than those found in control non-cancer patients, thus confirming the relevance of cysteine and serine protease-dependent mechanisms in GI cancers. We have also further confirmed that serum CATB and uPA plasma levels were elevated in patients with CRC[108-111]. In addition, we have shown that serum CATB and plasma uPA did show a significant increase in advanced CRC stage. We have also demonstrated for the first time that antigen levels of CATB and uPA were significantly higher in blood samples of patients with colorectal adenomas as compared to controls. Furthermore, we have found significantly higher CATB and uPA antigen levels in patients with tubulovillous adenomas with high-grade dysplasia (HGD) compared to those with tubular adenomas with low-grade dysplasia (LGD). Thus taken together, the data from blood samples with previous results obtained in colorectal tissues confirm that a concomitant activation of serine (CATB) and uPA may be involved in the progression from premalignant colorectal adenomas into CRC[112,113].

Some other studies have also suggested the potential impact of cysteine or serine proteases as tumor markers in CRC[108-111]. Given the lack in the literature for a comparison of the tumor marker utility and possible prognostic relevance of cathepsins (CATB, CATL) and the uPA/PAI-1 system in the same experimental setting, we have surveyed the behavior of CATB, CATL, uPA, PAI-1 in CRC and compared it with the commonly used gastrointestinal tumor markers CEA and CA 19-9, and then evaluated any correlation between these parameters and the clinico-pathological staging of CRC[114]. In this study, we have demonstrated the potential tumor marker utility and prognostic relevance of cysteine and serine proteinases for patients with CRC. We have also shown the concomitant activation of these systems in CRC. At the time of clinical presentation, proteinases were more sensitive indicators of diagnosis than the most commonly used markers CEA and CA 19-9. When proteinases, CEA and CA 19-9 were used as single markers, we found that their sensitivity was more indicative of CRC than CEA or CA 19-9. The simultaneous determination of several markers led to a higher sensitivity in our group of CRC patients: PAI-1 combined with CATB or uPA was superior compared to the combinations of all other markers. In addition, the sensitivity of CEA or CA 19-9 in combination with a proteinase antigen level was more indicative for CRC than CEA or CA 19-9 alone. In this study increased serum or plasma proteinase concentrations significantly correlated with advanced tumor stage. In a univariate survival analysis high serum CATB, CATL and plasma PAI-1 antigen levels identified patients with shorter survival and those who were at higher risk of death. In addition, PAI-1 and CATB were proved as independent predictor variables in a multivariate statistical analysis. We also confirmed that cysteine and serine proteinases were significantly higher in blood samples of patients with colorectal adenomas compared to controls, suggesting that these proteinases may be involved in the malignant transformation of colorectal adenomas[114].

Tissue MMPs and TIMPs in CRC

The behavior of MMPs and TIMPs in CRC has recently been extensively reviewed by our own group[115]. Several studies have shown that the expression of several MMPs and TIMPs are enhanced in CRC. In an immunohistochemical study we have demonstrated that tissue expression of one particular MMP, MMP-9 was significantly higher in moderately (G2) and poorly (G3) differentiated tumors than in well differentiated (G1) cancers, as well as in advanced Dukes stages compared with Dukes stage A[116]. Some recent studies have confirmed that tissue MMP-9 can be considered as an independent prognostic marker in CRC[117-120]. In addition, it has been demonstrated that not only MMP-9, but also tissue expressions of other MMPs and TIMPs have strong prognostic impact in CRC[121-126].

MMPs and TIMPs also play a role in malignant transformation from colorectal adenomas to CRC. Our group in concordance with others has shown that tissue expression of MMP-9, MMP-2, TIMP-1 and TIMP-2 were significantly higher in advanced versus non-advanced adenomas, suggesting that MMPs and TIMPs might be markers for early colorectal carcinogenesis. The ability of MMPs and TIMPs to distinguish adenomas with HGD from adenomas without HGD may be of clinical value in predicting additional cancer risk for an individual patient[116,127-130].

Serum and plasma MMPs and TIMPs in CRC

The diagnostic, prognostic and tumor marker impact of serum-plasma MMPs and TIMPs in CRC has been extensively studied. We have demonstrated that serum antigen concentrations of MMP-2, MMP-7, MMP-9 as well as TIMP-1 and TIMP-2 were significantly higher in patients with CRC than in healthy controls. All examined parameters were significantly higher in patients with CRC than in patients with adenomas. Higher antigen concentrations of MMPs and TIMPs significantly correlated with preoperative tumor stage. The data from blood samples confirmed previous results of tissue expressions concluding that MMPs and their inhibitors TIMP-1 and TIMP-2 play an important role not only in CRC invasion and metastasis, but they are also activated in premalignant colorectal adenomas[82].

Several recent studies confirmed that high preoperative serum or plasma MMP-2, MMP-9 and mainly TIMP-1 antigen levels are strong predictive factors for poor prognosis in patients with CRC[131-137].

The potential tumor marker role of MMPs and TIMPs has also been extensively studied. It has been demonstrated that MMP-9 and TIMP-1 have significant potential as biomarkers in CRC. Diagnostic sensitivity of MMP-9 and TIMP-1 was consistently higher as compared with the conventional biomarkers (CEA or CA 19-9)[131,138-140].

It has also been proposed that TIMPs can predict individual response to chemotherapy and could be considered as an additional tool for monitoring chemotherapy in CRC[141-144].

One of the greatest challenges in CRC management is to predict the outcome of each patient so that we can determine who will really benefit from intensified adjuvant therapy. The classical TNM staging system relied heavily on the exact extent of cancer at the time of diagnosis and is greatly predictive in stage I and stage IV tumors. However, it is less informative for patients including stage II and stage III CRC. After curative surgery, stage III CRC patients experience 50% chance of developing recurrence. It is well documented that the overall survival rate of stage III CRC could clearly benefit from adjuvant chemotherapy. In contrast, the role of adjuvant chemotherapy for stage II CRC is still controversial, despite the 20% recurrence in this group. We and others have suggested that proteolytic enzymes could constitute useful independent markers in addition to the TNM staging system for the intermediate groups including stage II and stage III CRC. Proteolytic enzymes may help to identify patients who are more likely to have disease relapse and high risk of death, thus those who are potential candidates to receive aggressive adjuvant chemotherapy[107,119,145-147].

On the other hand, taken into consideration that proteolytic enzymes may have a crucial role not only in the invasive process of CRC, but also in the progression of precancerous conditions and lesions into cancer, quantification of proteinases might be useful to identify patients at higher risk for progression to cancer, who could be subjected to a more strict follow-up protocol.

PROTEOLYTIC ENZYMES AS POTENTIAL TARGET FOR CANCER THERAPY

The accumulated evidence strongly supports the concept of the use of cysteine proteinases (cathepsins) and serine proteinases (uPA-PAI system) as targets for cancer therapy. It has been suggested that cathepsin inhibition represents a potential therapeutic strategy for the treatment of cancer. Cathepsins inhibition seems to reduce invasion and metastasis, but there is concern that selective cathepsin inhibition induces compensatory activity by other cathepsins. The combination of cathepsin inhibition with conventional chemotherapy seems to be more effective and has yielded more consistent clinical results. Future research should be focused on the exact mechanisms and clinical effects of this combination treatment[148-151].

The multifunctionality of the uPA-uPAR-PAI-1 system in cancer spotlighted serine proteinases to become potential targets for anticancer treatment. In addition, uPA system is also increasingly being recognized as a candidate target for gene therapy in cancer[152-159]. Several strategies, including the use of ribozymes, DNAzyme, antisense oligodeoxynucleotides, uPA inhibitors, soluble uPAR, catalytically inactive uPA fragments, the interactions of uPAR with integrins and transmembrane receptors, synthetic peptides and synthetic hybrids are under study, as they all interfere with the activity of uPA or uPAR in tumor cells. Many clinical studies are ongoing and some uPA-related compounds have reached Phase II clinical trials.

Several therapeutic MMP inhibitors (MMPIs) have also been developed to target MMPs. Various natural compounds have been identified as inhibiting MMPs. In addition, several generations of synthetic MMPIs were tested in phase III clinical trials in humans, including peptidomimetics, non-peptidomimetic inhibitors or tetracycline derivates, targeting MMPs in the extracellular space. Other strategies of MMP inhibition involve small interfering and antisense RNA technology[160-165]. In contrast to their promising effect in preclinical models, most of these agents unfortunately failed in clinical trials, thus they are yet not available for routine use. The use of broad-spectrum MMPIs may lead to undesired clinical consequences as a result of the wide range of MMPs that are inhibited. In addition, toxic side effects, such as musculoskeletal syndrome, have limited drug efficiency.

The ADAMs is also a family of potential new targets for cancer therapy. ADAMs (a disintegrin and metalloproteinase) are members of a zinc-dependent family of matrix metalloproteinases. Preclinical findings suggest that selective ADAM inhibitors might be novel anticancer agents. ADAMs inhibitors may be particularly useful in treating cancers that depend on HER or TNF-α-mediated signalling[166-169].

One of the major challenges for the future is the development of monoclonal antibodies or inhibitors that are specific for certain MMPs, showing no cross-reaction with other MMPs with improved pharmacokinetic properties and selectivity. In addition, their use in combination with established chemotherapeutic strategies might have the potential to become valuable oncological treatment modalities[161,170-176].

CONCLUSION

Proteolytic enzymes play a sophisticated role in cancer development and progression due to their abilities to degrade various substrates. However, the role of proteolytic enzymes in tumor progression is much more complex than that derived from their direct degradative action on BM and ECM components. Proteinases may have a crucial role not only in the invasive process of CRC, but also in the progression of precancerous conditions and lesions to CRC. Proteolytic enzymes could constitute effective independent prognostic markers additive to TNM staging system in CRC. Their determination might be useful to identify patients at higher risk for progression to cancer, who could be subjected to a more strict endoscopic follow-up protocol.

It has recently been demonstrated in experimental settings that a newly developed near-infrared bioactivable probe (MMPSense) that reports the activity of a broad array of MMP isoforms detects both polypoid and nonpolypoid early colorectal adenomas with a high specificity[177]. Future studies will focus on the use of such fluorescent probes combined with colonoscopy to identify neoplastic lesions based on their molecular “fingerprint” (i.e., proteinase enzymatic activity) rather than solely on their morphologic properties. The ability to detect nonpolypoid lesions using this fluorescent probe alone or in combination with other molecular probes may offer new future perspectives for colorectal screening. In addition, it might also serve as a potential strategy for the pharmacodynamic monitoring of targeted therapy. The pharmacological targeting of CRC by the development of a new generation of effective and selective proteinase inhibitors is another emerging area of research.

Footnotes

P- Reviewer: Altomare DF, Cai SJ, Lam AK, Morris DL, Wei SC S- Editor: Ma YJ L- Editor: A E- Editor: Wang CH

References
1.  Hart IR, Saini A. Biology of tumour metastasis. Lancet. 1992;339:1453-1457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 235]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
2.  Nigam AK, Pignatelli M, Boulos PB. Current concepts in metastasis. Gut. 1994;35:996-1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 17]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
3.  Herszényi L, Plebani M, Carraro P, De Paoli M, Roveroni G, Cardin R, Foschia F, Tulassay Z, Naccarato R, Farinati F. Proteases in gastrointestinal neoplastic diseases. Clin Chim Acta. 2000;291:171-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
4.  Hoon DS, Ferris R, Tanaka R, Chong KK, Alix-Panabières C, Pantel K. Molecular mechanisms of metastasis. J Surg Oncol. 2011;103:508-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
5.  Witte MH, Dellinger MT, McDonald DM, Nathanson SD, Boccardo FM, Campisi CC, Sleeman JP, Gershenwald JE. Lymphangiogenesis and hemangiogenesis: potential targets for therapy. J Surg Oncol. 2011;103:489-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
6.  Herszényi L, Lakatos G, Hritz I, Varga MZ, Cierny G, Tulassay Z. The role of inflammation and proteinases in tumor progression. Dig Dis. 2012;30:249-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
7.  Kawada K, Hasegawa S, Murakami T, Itatani Y, Hosogi H, Sonoshita M, Kitamura T, Fujishita T, Iwamoto M, Matsumoto T. Molecular mechanisms of liver metastasis. Int J Clin Oncol. 2011;16:464-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
8.  Said N, Theodorescu D. Permissive role of endothelin receptors in tumor metastasis. Life Sci. 2012;91:522-527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Man YG, Stojadinovic A, Mason J, Avital I, Bilchik A, Bruecher B, Protic M, Nissan A, Izadjoo M, Zhang X. Tumor-infiltrating immune cells promoting tumor invasion and metastasis: existing theories. J Cancer. 2013;4:84-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 117]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
10.  Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6912]  [Cited by in F6Publishing: 7713]  [Article Influence: 482.1]  [Reference Citation Analysis (0)]
11.  Mantovani A, Garlanda C, Allavena P. Molecular pathways and targets in cancer-related inflammation. Ann Med. 2010;42:161-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 141]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
12.  Maletzki C, Emmrich J. Inflammation and immunity in the tumor environment. Dig Dis. 2010;28:574-578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
13.  Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30:1073-1081.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1802]  [Cited by in F6Publishing: 1953]  [Article Influence: 130.2]  [Reference Citation Analysis (0)]
14.  Hold GL, El-Omar EM. Genetic aspects of inflammation and cancer. Biochem J. 2008;410:225-235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
15.  Scheede-Bergdahl C, Watt HL, Trutschnigg B, Kilgour RD, Haggarty A, Lucar E, Vigano A. Is IL-6 the best pro-inflammatory biomarker of clinical outcomes of cancer cachexia? Clin Nutr. 2012;31:85-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 72]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
16.  Dennis KL, Blatner NR, Gounari F, Khazaie K. Current status of interleukin-10 and regulatory T-cells in cancer. Curr Opin Oncol. 2013;25:637-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 189]  [Article Influence: 18.9]  [Reference Citation Analysis (0)]
17.  Zheng M, Jiang J, Tang YL, Liang XH. Oncogene and non-oncogene addiction in inflammation-associated cancers. Future Oncol. 2013;9:561-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
18.  Monteleone G, Pallone F, Stolfi C. The dual role of inflammation in colon carcinogenesis. Int J Mol Sci. 2012;13:11071-11084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
19.  Marusawa H, Jenkins BJ. Inflammation and gastrointestinal cancer: an overview. Cancer Lett. 2014;345:153-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
20.  Yurchenco PD, Schittny JC. Molecular architecture of basement membranes. FASEB J. 1990;4:1577-1590.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  DeClerck YA, Mercurio AM, Stack MS, Chapman HA, Zutter MM, Muschel RJ, Raz A, Matrisian LM, Sloane BF, Noel A. Proteases, extracellular matrix, and cancer: a workshop of the path B study section. Am J Pathol. 2004;164:1131-1139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 163]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
22.  Cavallo-Medved D, Rudy D, Blum G, Bogyo M, Caglic D, Sloane BF. Live-cell imaging demonstrates extracellular matrix degradation in association with active cathepsin B in caveolae of endothelial cells during tube formation. Exp Cell Res. 2009;315:1234-1246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 88]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
23.  Liotta LA, Kohn EC. The microenvironment of the tumour-host interface. Nature. 2001;411:375-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1724]  [Cited by in F6Publishing: 1678]  [Article Influence: 73.0]  [Reference Citation Analysis (0)]
24.  Geho DH, Bandle RW, Clair T, Liotta LA. Physiological mechanisms of tumor-cell invasion and migration. Physiology (Bethesda). 2005;20:194-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 100]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
25.  Cirri P, Chiarugi P. Cancer-associated-fibroblasts and tumour cells: a diabolic liaison driving cancer progression. Cancer Metastasis Rev. 2012;31:195-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 366]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
26.  Sleeman JP. The metastatic niche and stromal progression. Cancer Metastasis Rev. 2012;31:429-440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 146]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
27.  Catalano V, Turdo A, Di Franco S, Dieli F, Todaro M, Stassi G. Tumor and its microenvironment: a synergistic interplay. Semin Cancer Biol. 2013;23:522-532.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Polgár L. Common feature of the four types of protease mechanism. Biol Chem Hoppe Seyler. 1990;371 Suppl:327-331.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Mignatti P, Rifkin DB. Biology and biochemistry of proteinases in tumor invasion. Physiol Rev. 1993;73:161-195.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Liotta LA, Stetler-Stevenson WG. Tumor invasion and metastasis: an imbalance of positive and negative regulation. Cancer Res. 1991;51:5054s-5059s.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Sun J. Matrix metalloproteinases and tissue inhibitor of metalloproteinases are essential for the inflammatory response in cancer cells. J Signal Transduct. 2010;2010:985132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 54]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
32.  Folgueras AR, Pendás AM, Sánchez LM, López-Otín C. Matrix metalloproteinases in cancer: from new functions to improved inhibition strategies. Int J Dev Biol. 2004;48:411-424.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Yoon SO, Park SJ, Yun CH, Chung AS. Roles of matrix metalloproteinases in tumor metastasis and angiogenesis. J Biochem Mol Biol. 2003;36:128-137.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Martin MD, Matrisian LM. The other side of MMPs: protective roles in tumor progression. Cancer Metastasis Rev. 2007;26:717-724.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Rmali KA, Puntis MC, Jiang WG. Tumour-associated angiogenesis in human colorectal cancer. Colorectal Dis. 2007;9:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Rudmik LR, Magliocco AM. Molecular mechanisms of hepatic metastasis in colorectal cancer. J Surg Oncol. 2005;92:347-359.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Premzl A, Turk V, Kos J. Intracellular proteolytic activity of cathepsin B is associated with capillary-like tube formation by endothelial cells in vitro. J Cell Biochem. 2006;97:1230-1240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
38.  Sloane BF. Cathepsin B and cystatins: evidence for a role in cancer progression. Semin Cancer Biol. 1990;1:137-152.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Murphy G, Atkinson S, Ward R, Gavrilovic J, Reynolds JJ. The role of plasminogen activators in the regulation of connective tissue metalloproteinases. Ann N Y Acad Sci. 1992;667:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 179]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
40.  Danø K, Behrendt N, Høyer-Hansen G, Johnsen M, Lund LR, Ploug M, Rømer J. Plasminogen activation and cancer. Thromb Haemost. 2005;93:676-681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
41.  Krueger S, Kalinski T, Wolf H, Kellner U, Roessner A. Interactions between human colon carcinoma cells, fibroblasts and monocytic cells in coculture--regulation of cathepsin B expression and invasiveness. Cancer Lett. 2005;223:313-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
42.  Cavallo-Medved D, Mai J, Dosescu J, Sameni M, Sloane BF. Caveolin-1 mediates the expression and localization of cathepsin B, pro-urokinase plasminogen activator and their cell-surface receptors in human colorectal carcinoma cells. J Cell Sci. 2005;118:1493-1503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 97]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
43.  Sprengers ED, Kluft C. Plasminogen activator inhibitors. Blood. 1987;69:381-387.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Cubellis MV, Wun TC, Blasi F. Receptor-mediated internalization and degradation of urokinase is caused by its specific inhibitor PAI-1. EMBO J. 1990;9:1079-1085.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Hildenbrand R, Allgayer H, Marx A, Stroebel P. Modulators of the urokinase-type plasminogen activation system for cancer. Expert Opin Investig Drugs. 2010;19:641-652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
46.  Harbeck N, Alt U, Berger U, Krüger A, Thomssen C, Jänicke F, Höfler H, Kates RE, Schmitt M. Prognostic impact of proteolytic factors (urokinase-type plasminogen activator, plasminogen activator inhibitor 1, and cathepsins B, D, and L) in primary breast cancer reflects effects of adjuvant systemic therapy. Clin Cancer Res. 2001;7:2757-2764.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Manders P, Tjan-Heijnen VC, Span PN, Grebenchtchikov N, Foekens JA, Beex LV, Sweep CG. Predictive impact of urokinase-type plasminogen activator: plasminogen activator inhibitor type-1 complex on the efficacy of adjuvant systemic therapy in primary breast cancer. Cancer Res. 2004;64:659-664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
48.  Harbeck N, Schmitt M, Paepke S, Allgayer H, Kates RE. Tumor-associated proteolytic factors uPA and PAI-1: critical appraisal of their clinical relevance in breast cancer and their integration into decision-support algorithms. Crit Rev Clin Lab Sci. 2007;44:179-201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
49.  Werle B, Kotzsch M, Lah TT, Kos J, Gabrijelcic-Geiger D, Spiess E, Schirren J, Ebert W, Fiehn W, Luther T. Cathepsin B, plasminogenactivator-inhibitor (PAI-1) and plasminogenactivator-receptor (uPAR) are prognostic factors for patients with non-small cell lung cancer. Anticancer Res. 2004;24:4147-4161.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Offersen BV, Pfeiffer P, Andreasen P, Overgaard J. Urokinase plasminogen activator and plasminogen activator inhibitor type-1 in nonsmall-cell lung cancer: relation to prognosis and angiogenesis. Lung Cancer. 2007;56:43-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 44]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
51.  Speleman L, Kerrebijn JD, Look MP, Meeuwis CA, Foekens JA, Berns EM. Prognostic value of plasminogen activator inhibitor-1 in head and neck squamous cell carcinoma. Head Neck. 2007;29:341-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
52.  Dorn J, Harbeck N, Kates R, Magdolen V, Grass L, Soosaipillai A, Schmalfeldt B, Diamandis EP, Schmitt M. Disease processes may be reflected by correlations among tissue kallikrein proteases but not with proteolytic factors uPA and PAI-1 in primary ovarian carcinoma. Biol Chem. 2006;387:1121-1128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
53.  Sier CF, Verspaget HW, Griffioen G, Ganesh S, Vloedgraven HJ, Lamers CB. Plasminogen activators in normal tissue and carcinomas of the human oesophagus and stomach. Gut. 1993;34:80-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 51]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
54.  Iwamoto J, Mizokami Y, Takahashi K, Nakajima K, Ohtsubo T, Miura S, Narasaka T, Takeyama H, Omata T, Shimokobe K. Expressions of urokinase-type plasminogen activator, its receptor and plasminogen activator inhibitor-1 in gastric cancer cells and effects of Helicobacter pylori. Scand J Gastroenterol. 2005;40:783-793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
55.  Sakakibara T, Hibi K, Koike M, Fujiwara M, Kodera Y, Ito K, Nakao A. Plasminogen activator inhibitor-1 as a potential marker for the malignancy of gastric cancer. Cancer Sci. 2006;97:395-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
56.  Beyer BC, Heiss MM, Simon EH, Gruetzner KU, Babic R, Jauch KW, Schildberg FW, Allgayer H. Urokinase system expression in gastric carcinoma: prognostic impact in an independent patient series and first evidence of predictive value in preoperative biopsy and intestinal metaplasia specimens. Cancer. 2006;106:1026-1035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 50]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
57.  Herszènyi L, Plebani M, Carraro P, De Paoli M, Cardin R, Di Mario F, Kusstatscher S, Naccarato R, Farinati F. Impaired fibrinolysis and increased protease levels in gastric and duodenal mucosa of patients with active duodenal ulcer. Am J Gastroenterol. 1997;92:843-847.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Plebani M, Herszènyi L, Cardin R, Roveroni G, Carraro P, Paoli MD, Rugge M, Grigioni WF, Nitti D, Naccarato R. Cysteine and serine proteases in gastric cancer. Cancer. 1995;76:367-375.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Plebani M, Herszènyi L, Carraro P, De Paoli M, Roveroni G, Cardin R, Tulassay Z, Naccarato R, Farinati F. Urokinase-type plasminogen activator receptor in gastric cancer: tissue expression and prognostic role. Clin Exp Metastasis. 1997;15:418-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
60.  Farinati F, Herszényi L, Plebani M, Carraro P, De Paoli M, Cardin R, Roveroni G, Rugge M, Nitti D, Grigioni WF. Increased levels of cathepsin B and L, urokinase-type plasminogen activator and its inhibitor type-1 as an early event in gastric carcinogenesis. Carcinogenesis. 1996;17:2581-2587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
61.  Herszényi L, István G, Cardin R, De Paoli M, Plebani M, Tulassay Z, Farinati F. Serum cathepsin B and plasma urokinase-type plasminogen activator levels in gastrointestinal tract cancers. Eur J Cancer Prev. 2008;17:438-445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
62.  Nekarda H, Schmitt M, Ulm K, Wenninger A, Vogelsang H, Becker K, Roder JD, Fink U, Siewert JR. Prognostic impact of urokinase-type plasminogen activator and its inhibitor PAI-1 in completely resected gastric cancer. Cancer Res. 1994;54:2900-2907.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Ganesh S, Sier CF, Heerding MM, van Krieken JH, Griffioen G, Welvaart K, van de Velde CJ, Verheijen JH, Lamers CB, Verspaget HW. Prognostic value of the plasminogen activation system in patients with gastric carcinoma. Cancer. 1996;77:1035-1043.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Niedergethmann M, Hildenbrand R, Wolf G, Verbeke CS, Richter A, Post S. Angiogenesis and cathepsin expression are prognostic factors in pancreatic adenocarcinoma after curative resection. Int J Pancreatol. 2000;28:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Zheng Q, Tang ZY, Xue Q, Shi DR, Song HY, Tang HB. Invasion and metastasis of hepatocellular carcinoma in relation to urokinase-type plasminogen activator, its receptor and inhibitor. J Cancer Res Clin Oncol. 2000;126:641-646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 85]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
66.  van Kempen LC, de Visser KE, Coussens LM. Inflammation, proteases and cancer. Eur J Cancer. 2006;42:728-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 133]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
67.  Affara NI, Andreu P, Coussens LM. Delineating protease functions during cancer development. Methods Mol Biol. 2009;539:1-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
68.  Noël A, Jost M, Maquoi E. Matrix metalloproteinases at cancer tumor-host interface. Semin Cell Dev Biol. 2008;19:52-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 184]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
69.  Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 2010;141:52-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3192]  [Cited by in F6Publishing: 3489]  [Article Influence: 249.2]  [Reference Citation Analysis (0)]
70.  Page-McCaw A, Ewald AJ, Werb Z. Matrix metalloproteinases and the regulation of tissue remodelling. Nat Rev Mol Cell Biol. 2007;8:221-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1975]  [Cited by in F6Publishing: 2046]  [Article Influence: 120.4]  [Reference Citation Analysis (0)]
71.  Hayden DM, Forsyth C, Keshavarzian A. The role of matrix metalloproteinases in intestinal epithelial wound healing during normal and inflammatory states. J Surg Res. 2011;168:315-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
72.  Puthenedam M, Wu F, Shetye A, Michaels A, Rhee KJ, Kwon JH. Matrilysin-1 (MMP7) cleaves galectin-3 and inhibits wound healing in intestinal epithelial cells. Inflamm Bowel Dis. 2011;17:260-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
73.  Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol. 2001;17:463-516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2922]  [Cited by in F6Publishing: 2818]  [Article Influence: 122.5]  [Reference Citation Analysis (0)]
74.  Buergy D, Fuchs T, Kambakamba P, Mudduluru G, Maurer G, Post S, Tang Y, Nakada MT, Yan L, Allgayer H. Prognostic impact of extracellular matrix metalloprotease inducer: immunohistochemical analyses of colorectal tumors and immunocytochemical screening of disseminated tumor cells in bone marrow from patients with gastrointestinal cancer. Cancer. 2009;115:4667-4678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
75.  Herszenyi L, Hritz I, Pregun I, Sipos F, Juhasz M, Molnar B, Tulassay Z. Alterations of glutathione S-transferase and matrix metalloproteinase-9 expressions are early events in esophageal carcinogenesis. World J Gastroenterol. 2007;13:676-682.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Mäkitalo L, Rintamäki H, Tervahartiala T, Sorsa T, Kolho KL. Serum MMPs 7-9 and their inhibitors during glucocorticoid and anti-TNF-α therapy in pediatric inflammatory bowel disease. Scand J Gastroenterol. 2012;47:785-794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
77.  Kirkegaard T, Hansen A, Bruun E, Brynskov J. Expression and localisation of matrix metalloproteinases and their natural inhibitors in fistulae of patients with Crohn’s disease. Gut. 2004;53:701-709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 151]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
78.  Stallmach A, Chan CC, Ecker KW, Feifel G, Herbst H, Schuppan D, Zeitz M. Comparable expression of matrix metalloproteinases 1 and 2 in pouchitis and ulcerative colitis. Gut. 2000;47:415-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 78]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
79.  von Lampe B, Barthel B, Coupland SE, Riecken EO, Rosewicz S. Differential expression of matrix metalloproteinases and their tissue inhibitors in colon mucosa of patients with inflammatory bowel disease. Gut. 2000;47:63-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 283]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
80.  Ravi A, Garg P, Sitaraman SV. Matrix metalloproteinases in inflammatory bowel disease: boon or a bane? Inflamm Bowel Dis. 2007;13:97-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 99]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
81.  Lakatos G, Sipos F, Miheller P, Hritz I, Varga MZ, Juhász M, Molnár B, Tulassay Z, Herszényi L. The behavior of matrix metalloproteinase-9 in lymphocytic colitis, collagenous colitis and ulcerative colitis. Pathol Oncol Res. 2012;18:85-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
82.  Lakatos G, Hritz I, Varga MZ, Juhász M, Miheller P, Cierny G, Tulassay Z, Herszényi L. The impact of matrix metalloproteinases and their tissue inhibitors in inflammatory bowel diseases. Dig Dis. 2012;30:289-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
83.  Roy R, Yang J, Moses MA. Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer. J Clin Oncol. 2009;27:5287-5297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 588]  [Article Influence: 39.2]  [Reference Citation Analysis (0)]
84.  Yeh YC, Sheu BS, Cheng HC, Wang YL, Yang HB, Wu JJ. Elevated serum matrix metalloproteinase-3 and -7 in H. pylori-related gastric cancer can be biomarkers correlating with a poor survival. Dig Dis Sci. 2010;55:1649-1657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 54]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
85.  Singh N, Das P, Datta Gupta S, Sahni P, Pandey RM, Gupta S, Chauhan SS, Saraya A. Prognostic significance of extracellular matrix degrading enzymes-cathepsin L and matrix metalloproteases-2 [MMP-2] in human pancreatic cancer. Cancer Invest. 2013;31:461-471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
86.  Hornebeck W, Lambert E, Petitfrère E, Bernard P. Beneficial and detrimental influences of tissue inhibitor of metalloproteinase-1 (TIMP-1) in tumor progression. Biochimie. 2005;87:377-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
87.  Schrötzlmair F, Kopitz C, Halbgewachs B, Lu F, Algül H, Brünner N, Gänsbacher B, Krüger A. Tissue inhibitor of metalloproteinases-1-induced scattered liver metastasis is mediated by host-derived urokinase-type plasminogen activator. J Cell Mol Med. 2010;14:2760-2770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
88.  Schelter F, Halbgewachs B, Bäumler P, Neu C, Görlach A, Schrötzlmair F, Krüger A. Tissue inhibitor of metalloproteinases-1-induced scattered liver metastasis is mediated by hypoxia-inducible factor-1α. Clin Exp Metastasis. 2011;28:91-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
89.  Medina C, Radomski MW. Role of matrix metalloproteinases in intestinal inflammation. J Pharmacol Exp Ther. 2006;318:933-938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 155]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
90.  Wiercinska-Drapalo A, Jaroszewicz J, Flisiak R, Prokopowicz D. Plasma matrix metalloproteinase-1 and tissue inhibitor of metalloproteinase-1 as biomarkers of ulcerative colitis activity. World J Gastroenterol. 2003;9:2843-2845.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Meijer MJ, Mieremet-Ooms MA, van Hogezand RA, Lamers CB, Hommes DW, Verspaget HW. Role of matrix metalloproteinase, tissue inhibitor of metalloproteinase and tumor necrosis factor-alpha single nucleotide gene polymorphisms in inflammatory bowel disease. World J Gastroenterol. 2007;13:2960-2966.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127:2893-2917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11128]  [Cited by in F6Publishing: 11614]  [Article Influence: 893.4]  [Reference Citation Analysis (4)]
93.  Ferlay J, Parkin DM, Steliarova-Foucher E. Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 2010;46:765-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1579]  [Cited by in F6Publishing: 1596]  [Article Influence: 114.0]  [Reference Citation Analysis (0)]
94.  Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, Smith T, Cooper D, Gansler T, Lerro C, Fedewa S. Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin. 2012;62:220-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1973]  [Cited by in F6Publishing: 2026]  [Article Influence: 168.8]  [Reference Citation Analysis (1)]
95.  Moghimi-Dehkordi B, Safaee A. An overview of colorectal cancer survival rates and prognosis in Asia. World J Gastrointest Oncol. 2012;4:71-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 102]  [Cited by in F6Publishing: 123]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
96.  Wittmann T, Stockbrugger R, Herszényi L, Jonkers D, Molnár B, Saurin JC, Regula J, Malesci A, Laghi L, Pintér T. New European initiatives in colorectal cancer screening: Budapest Declaration. Official appeal during the Hungarian Presidency of the Council of the European Union under the Auspices of the United European Gastroenterology Federation, the European Association for Gastroenterology and Endoscopy and the Hungarian Society of Gastroenterology. Dig Dis. 2012;30:320-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
97.  Soerjomataram I, Lortet-Tieulent J, Parkin DM, Ferlay J, Mathers C, Forman D, Bray F. Global burden of cancer in 2008: a systematic analysis of disability-adjusted life-years in 12 world regions. Lancet. 2012;380:1840-1850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 407]  [Cited by in F6Publishing: 409]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
98.  Sier CF, Vloedgraven HJ, Ganesh S, Griffioen G, Quax PH, Verheijen JH, Dooijewaard G, Welvaart K, van de Velde CJ, Lamers CB. Inactive urokinase and increased levels of its inhibitor type 1 in colorectal cancer liver metastasis. Gastroenterology. 1994;107:1449-1456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
99.  Ganesh S, Sier CF, Heerding MM, van Krieken JH, Griffioen G, Welvaart K, van de Velde CJ, Verheijen JH, Lamers CB, Verspaget HW. Contribution of plasminogen activators and their inhibitors to the survival prognosis of patients with Dukes’ stage B and C colorectal cancer. Br J Cancer. 1997;75:1793-1801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
100.  Montemurro P, Conese M, Altomare DF, Memeo V, Colucci M, Semeraro N. Blood and tissue fibrinolytic profiles in patients with colorectal carcinoma. Int J Clin Lab Res. 1995;25:195-200.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Adenis A, Huet G, Zerimech F, Hecquet B, Balduyck M, Peyrat JP. Cathepsin B, L, and D activities in colorectal carcinomas: relationship with clinico-pathological parameters. Cancer Lett. 1995;96:267-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 58]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
102.  Talieri M, Papadopoulou S, Scorilas A, Xynopoulos D, Arnogianaki N, Plataniotis G, Yotis J, Agnanti N. Cathepsin B and cathepsin D expression in the progression of colorectal adenoma to carcinoma. Cancer Lett. 2004;205:97-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 59]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
103.  Troy AM, Sheahan K, Mulcahy HE, Duffy MJ, Hyland JM, O’Donoghue DP. Expression of Cathepsin B and L antigen and activity is associated with early colorectal cancer progression. Eur J Cancer. 2004;40:1610-1616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 45]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
104.  Kim TD, Song KS, Li G, Choi H, Park HD, Lim K, Hwang BD, Yoon WH. Activity and expression of urokinase-type plasminogen activator and matrix metalloproteinases in human colorectal cancer. BMC Cancer. 2006;6:211.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Märkl B, Renk I, Oruzio DV, Jähnig H, Schenkirsch G, Schöler C, Ehret W, Arnholdt HM, Anthuber M, Spatz H. Tumour budding, uPA and PAI-1 are associated with aggressive behaviour in colon cancer. J Surg Oncol. 2010;102:235-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
106.  Halamkova J, Kiss I, Pavlovsky Z, Tomasek J, Jarkovsky J, Cech Z, Tucek S, Hanakova L, Moulis M, Zavrelova J. Clinical significance of the plasminogen activator system in relation to grade of tumor and treatment response in colorectal carcinoma patients. Neoplasma. 2011;58:377-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
107.  Herszènyi L, Plebani M, Carraro P, De Paoli M, Roveroni G, Cardin R, Tulassay Z, Naccarato R, Farinati F. The role of cysteine and serine proteases in colorectal carcinoma. Cancer. 1999;86:1135-1142.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Hirano T, Manabe T, Takeuchi S. Serum cathepsin B levels and urinary excretion of cathepsin B in the cancer patients with remote metastasis. Cancer Lett. 1993;70:41-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
109.  Kos J, Nielsen HJ, Krasovec M, Christensen IJ, Cimerman N, Stephens RW, Brünner N. Prognostic values of cathepsin B and carcinoembryonic antigen in sera of patients with colorectal cancer. Clin Cancer Res. 1998;4:1511-1516.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Sebzda T, Saleh Y, Gburek J, Warwas M, Andrzejak R, Siewinski M, Rudnicki J. Total and lipid-bound plasma sialic acid as diagnostic markers in colorectal cancer patients: correlation with cathepsin B expression in progression to Dukes stage. J Exp Ther Oncol. 2006;5:223-229.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Huber K, Kirchheimer JC, Sedlmayer A, Bell C, Ermler D, Binder BR. Clinical value of determination of urokinase-type plasminogen activator antigen in plasma for detection of colorectal cancer: comparison with circulating tumor-associated antigens CA 19-9 and carcinoembryonic antigen. Cancer Res. 1993;53:1788-1793.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Shuja S, Sheahan K, Murnane MJ. Cysteine endopeptidase activity levels in normal human tissues, colorectal adenomas and carcinomas. Int J Cancer. 1991;49:341-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
113.  de Bruin PA, Griffioen G, Verspaget HW, Verheijen JH, Lamers CB. Plasminogen activators and tumor development in the human colon: activity levels in normal mucosa, adenomatous polyps, and adenocarcinomas. Cancer Res. 1987;47:4654-4657.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Herszényi L, Farinati F, Cardin R, István G, Molnár LD, Hritz I, De Paoli M, Plebani M, Tulassay Z. Tumor marker utility and prognostic relevance of cathepsin B, cathepsin L, urokinase-type plasminogen activator, plasminogen activator inhibitor type-1, CEA and CA 19-9 in colorectal cancer. BMC Cancer. 2008;8:194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 69]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
115.  Herszényi L, Hritz I, Lakatos G, Varga MZ, Tulassay Z. The behavior of matrix metalloproteinases and their inhibitors in colorectal cancer. Int J Mol Sci. 2012;13:13240-13263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 109]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
116.  Herszényi L, Sipos F, Galamb O, Solymosi N, Hritz I, Miheller P, Berczi L, Molnár B, Tulassay Z. Matrix metalloproteinase-9 expression in the normal mucosa-adenoma-dysplasia-adenocarcinoma sequence of the colon. Pathol Oncol Res. 2008;14:31-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
117.  Jensen SA, Vainer B, Bartels A, Brünner N, Sørensen JB. Expression of matrix metalloproteinase 9 (MMP-9) and tissue inhibitor of metalloproteinases 1 (TIMP-1) by colorectal cancer cells and adjacent stroma cells--associations with histopathology and patients outcome. Eur J Cancer. 2010;46:3233-3242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
118.  Bendardaf R, Buhmeida A, Hilska M, Laato M, Syrjänen S, Syrjänen K, Collan Y, Pyrhönen S. MMP-9 (gelatinase B) expression is associated with disease-free survival and disease-specific survival in colorectal cancer patients. Cancer Invest. 2010;28:38-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 59]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
119.  Buhmeida A, Bendardaf R, Hilska M, Collan Y, Laato M, Syrjänen S, Syrjänen K, Pyrhönen S. Prognostic significance of matrix metalloproteinase-9 (MMP-9) in stage II colorectal carcinoma. J Gastrointest Cancer. 2009;40:91-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
120.  Langers AM, Verspaget HW, Hawinkels LJ, Kubben FJ, van Duijn W, van der Reijden JJ, Hardwick JC, Hommes DW, Sier CF. MMP-2 and MMP-9 in normal mucosa are independently associated with outcome of colorectal cancer patients. Br J Cancer. 2012;106:1495-1498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
121.  Hilska M, Roberts PJ, Collan YU, Laine VJ, Kössi J, Hirsimäki P, Rahkonen O, Laato M. Prognostic significance of matrix metalloproteinases-1, -2, -7 and -13 and tissue inhibitors of metalloproteinases-1, -2, -3 and -4 in colorectal cancer. Int J Cancer. 2007;121:714-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 115]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
122.  Møller Sørensen N, Vejgaard Sørensen I, Ørnbjerg Würtz S, Schrohl AS, Dowell B, Davis G, Jarle Christensen I, Nielsen HJ, Brünner N. Biology and potential clinical implications of tissue inhibitor of metalloproteinases-1 in colorectal cancer treatment. Scand J Gastroenterol. 2008;43:774-786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
123.  Kim YW, Ko YT, Kim NK, Chung HC, Min BS, Lee KY, Park JP, Kim H. A comparative study of protein expression in primary colorectal cancer and synchronous hepatic metastases: the significance of matrix metalloproteinase-1 expression as a predictor of liver metastasis. Scand J Gastroenterol. 2010;45:217-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
124.  Chu D, Zhao Z, Zhou Y, Li Y, Li J, Zheng J, Zhao Q, Wang W. Matrix metalloproteinase-9 is associated with relapse and prognosis of patients with colorectal cancer. Ann Surg Oncol. 2012;19:318-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
125.  Yang B, Gao J, Rao Z, Shen Q. Clinicopathological significance and prognostic value of MMP-13 expression in colorectal cancer. Scand J Clin Lab Invest. 2012;72:501-505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
126.  Yang B, Gao J, Rao Z, Shen Q. Clinicopathological and prognostic significance of α5β1-integrin and MMP-14 expressions in colorectal cancer. Neoplasma. 2013;60:254-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
127.  Liabakk NB, Talbot I, Smith RA, Wilkinson K, Balkwill F. Matrix metalloprotease 2 (MMP-2) and matrix metalloprotease 9 (MMP-9) type IV collagenases in colorectal cancer. Cancer Res. 1996;56:190-196.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Tomita T, Iwata K. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in colonic adenomas-adenocarcinomas. Dis Colon Rectum. 1996;39:1255-1264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 42]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
129.  Gimeno-García AZ, Santana-Rodríguez A, Jiménez A, Parra-Blanco A, Nicolás-Pérez D, Paz-Cabrera C, Díaz-González F, Medina C, Díaz-Flores L, Quintero E. Up-regulation of gelatinases in the colorectal adenoma-carcinoma sequence. Eur J Cancer. 2006;42:3246-3252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
130.  Murnane MJ, Cai J, Shuja S, McAneny D, Willett JB. Active matrix metalloproteinase-2 activity discriminates colonic mucosa, adenomas with and without high-grade dysplasia, and cancers. Hum Pathol. 2011;42:688-701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
131.  Mroczko B, Groblewska M, Okulczyk B, Kedra B, Szmitkowski M. The diagnostic value of matrix metalloproteinase 9 (MMP-9) and tissue inhibitor of matrix metalloproteinases 1 (TIMP-1) determination in the sera of colorectal adenoma and cancer patients. Int J Colorectal Dis. 2010;25:1177-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
132.  Dragutinović VV, Radonjić NV, Petronijević ND, Tatić SB, Dimitrijević IB, Radovanović NS, Krivokapić ZV. Matrix metalloproteinase-2 (MMP-2) and -9 (MMP-9) in preoperative serum as independent prognostic markers in patients with colorectal cancer. Mol Cell Biochem. 2011;355:173-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
133.  Nielsen HJ, Christensen IJ, Brünner N. A novel prognostic index in colorectal cancer defined by serum carcinoembryonic antigen and plasma tissue inhibitor of metalloproteinases-1. Scand J Gastroenterol. 2010;45:200-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
134.  Birgisson H, Nielsen HJ, Christensen IJ, Glimelius B, Brünner N. Preoperative plasma TIMP-1 is an independent prognostic indicator in patients with primary colorectal cancer: a prospective validation study. Eur J Cancer. 2010;46:3323-3331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
135.  Min BS, Kim NK, Jeong HC, Chung HC. High levels of serum VEGF and TIMP-1 are correlated with colon cancer liver metastasis and intrahepatic recurrence after liver resection. Oncol Lett. 2012;4:123-130.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Lee JH, Choi JW, Kim YS. Plasma or serum TIMP-1 is a predictor of survival outcomes in colorectal cancer: a meta-analysis. J Gastrointestin Liver Dis. 2011;20:287-291.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Hadler-Olsen E, Winberg JO, Uhlin-Hansen L. Matrix metalloproteinases in cancer: their value as diagnostic and prognostic markers and therapeutic targets. Tumour Biol. 2013;34:2041-2051.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 269]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
138.  Emara M, Cheung PY, Grabowski K, Sawicki G, Wozniak M. Serum levels of matrix metalloproteinase-2 and -9 and conventional tumor markers (CEA and CA 19-9) in patients with colorectal and gastric cancers. Clin Chem Lab Med. 2009;47:993-1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
139.  Nielsen HJ, Brünner N, Jorgensen LN, Olsen J, Rahr HB, Thygesen K, Hoyer U, Laurberg S, Stieber P, Blankenstein MA. Plasma TIMP-1 and CEA in detection of primary colorectal cancer: a prospective, population based study of 4509 high-risk individuals. Scand J Gastroenterol. 2011;46:60-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
140.  Wilson S, Damery S, Stocken DD, Dowswell G, Holder R, Ward ST, Redman V, Wakelam MJ, James J, Hobbs FD. Serum matrix metalloproteinase 9 and colorectal neoplasia: a community-based evaluation of a potential diagnostic test. Br J Cancer. 2012;106:1431-1438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
141.  Sørensen NM, Byström P, Christensen IJ, Berglund A, Nielsen HJ, Brünner N, Glimelius B. TIMP-1 is significantly associated with objective response and survival in metastatic colorectal cancer patients receiving combination of irinotecan, 5-fluorouracil, and folinic acid. Clin Cancer Res. 2007;13:4117-4122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 59]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
142.  Aldulaymi B, Christensen IJ, Sölétormos G, Jess P, Nielsen SE, Brünner N, Nielsen HJ. Changes in soluble CEA and TIMP-1 levels during adjuvant chemotherapy for stage III colon cancer. Anticancer Res. 2010;30:233-237.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Ramer R, Eichele K, Hinz B. Upregulation of tissue inhibitor of matrix metalloproteinases-1 confers the anti-invasive action of cisplatin on human cancer cells. Oncogene. 2007;26:5822-5827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
144.  Watanabe T, Kobunai T, Yamamoto Y, Matsuda K, Ishihara S, Nozawa K, Iinuma H, Ikeuchi H. Gene expression of vascular endothelial growth factor A, thymidylate synthase, and tissue inhibitor of metalloproteinase 3 in prediction of response to bevacizumab treatment in colorectal cancer patients. Dis Colon Rectum. 2011;54:1026-1035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
145.  Wu T, Li Y, Liu X, Lu J, He X, Wang Q, Li J, Du X. Identification of high-risk stage II and stage III colorectal cancer by analysis of MMP-21 expression. J Surg Oncol. 2011;104:787-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
146.  Biasi F, Guina T, Maina M, Nano M, Falcone A, Aroasio E, Saracco GM, Papotti M, Leonarduzzi G, Poli G. Progressive increase of matrix metalloprotease-9 and interleukin-8 serum levels during carcinogenic process in human colorectal tract. PLoS One. 2012;7:e41839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
147.  Langenskiöld M, Ivarsson ML, Holmdahl L, Falk P, Kåbjörn-Gustafsson C, Angenete E. Intestinal mucosal MMP-1 - a prognostic factor in colon cancer. Scand J Gastroenterol. 2013;48:563-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
148.  Palermo C, Joyce JA. Cysteine cathepsin proteases as pharmacological targets in cancer. Trends Pharmacol Sci. 2008;29:22-28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 227]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
149.  Reinheckel T, Peters C, Krüger A, Turk B, Vasiljeva O. Differential Impact of Cysteine Cathepsins on Genetic Mouse Models of De novo Carcinogenesis: Cathepsin B as Emerging Therapeutic Target. Front Pharmacol. 2012;3:133.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Lankelma JM, Voorend DM, Barwari T, Koetsveld J, Van der Spek AH, De Porto AP, Van Rooijen G, Van Noorden CJ. Cathepsin L, target in cancer treatment? Life Sci. 2010;86:225-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 117]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
151.  Maynadier M, Vezenkov LL, Amblard M, Martin V, Gandreuil C, Vaillant O, Gary-Bobo M, Basile I, Hernandez JF, Garcia M. Dipeptide mimic oligomer transporter mediates intracellular delivery of Cathepsin D inhibitors: a potential target for cancer therapy. J Control Release. 2013;171:251-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
152.  Swedberg JE, Harris JM. Natural and engineered plasmin inhibitors: applications and design strategies. Chembiochem. 2012;13:336-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
153.  Carriero MV, Stoppelli MP. The urokinase-type plasminogen activator and the generation of inhibitors of urokinase activity and signaling. Curr Pharm Des. 2011;17:1944-1961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
154.  Schmitt M, Harbeck N, Brünner N, Jänicke F, Meisner C, Mühlenweg B, Jansen H, Dorn J, Nitz U, Kantelhardt EJ. Cancer therapy trials employing level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn. 2011;11:617-634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 74]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
155.  Lund IK, Illemann M, Thurison T, Christensen IJ, Høyer-Hansen G. uPAR as anti-cancer target: evaluation of biomarker potential, histological localization, and antibody-based therapy. Curr Drug Targets. 2011;12:1744-1760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 61]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
156.  Mekkawy AH, Morris DL, Pourgholami MH. Urokinase plasminogen activator system as a potential target for cancer therapy. Future Oncol. 2009;5:1487-1499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
157.  Ngo JC, Jiang L, Lin Z, Yuan C, Chen Z, Zhang X, Yu H, Wang J, Lin L, Huang M. Structural basis for therapeutic intervention of uPA/uPAR system. Curr Drug Targets. 2011;12:1729-1743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
158.  Dass K, Ahmad A, Azmi AS, Sarkar SH, Sarkar FH. Evolving role of uPA/uPAR system in human cancers. Cancer Treat Rev. 2008;34:122-136.  [PubMed]  [DOI]  [Cited in This Article: ]
159.  Pillay V, Dass CR, Choong PF. The urokinase plasminogen activator receptor as a gene therapy target for cancer. Trends Biotechnol. 2007;25:33-39.  [PubMed]  [DOI]  [Cited in This Article: ]
160.  Mannello F, Tonti G, Papa S. Matrix metalloproteinase inhibitors as anticancer therapeutics. Curr Cancer Drug Targets. 2005;5:285-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 114]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
161.  Gialeli C, Theocharis AD, Karamanos NK. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J. 2011;278:16-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1046]  [Cited by in F6Publishing: 1137]  [Article Influence: 81.2]  [Reference Citation Analysis (0)]
162.  Tu G, Xu W, Huang H, Li S. Progress in the development of matrix metalloproteinase inhibitors. Curr Med Chem. 2008;15:1388-1395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
163.  Zucker S, Cao J, Chen WT. Critical appraisal of the use of matrix metalloproteinase inhibitors in cancer treatment. Oncogene. 2000;19:6642-6650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 405]  [Cited by in F6Publishing: 423]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
164.  Fingleton B. MMPs as therapeutic targets--still a viable option? Semin Cell Dev Biol. 2008;19:61-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 207]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
165.  Li X, Wu JF. Recent developments in patent anti-cancer agents targeting the matrix metalloproteinases (MMPs). Recent Pat Anticancer Drug Discov. 2010;5:109-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
166.  Duffy MJ, McKiernan E, O’Donovan N, McGowan PM. Role of ADAMs in cancer formation and progression. Clin Cancer Res. 2009;15:1140-1144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 167]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
167.  Duffy MJ, Mullooly M, O’Donovan N, Sukor S, Crown J, Pierce A, McGowan PM. The ADAMs family of proteases: new biomarkers and therapeutic targets for cancer? Clin Proteomics. 2011;8:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 144]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
168.  Merchant NB, Voskresensky I, Rogers CM, Lafleur B, Dempsey PJ, Graves-Deal R, Revetta F, Foutch AC, Rothenberg ML, Washington MK. TACE/ADAM-17: a component of the epidermal growth factor receptor axis and a promising therapeutic target in colorectal cancer. Clin Cancer Res. 2008;14:1182-1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 74]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
169.  Kveiborg M, Jacobsen J, Lee MH, Nagase H, Wewer UM, Murphy G. Selective inhibition of ADAM12 catalytic activity through engineering of tissue inhibitor of metalloproteinase 2 (TIMP-2). Biochem J. 2010;430:79-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
170.  Konstantinopoulos PA, Karamouzis MV, Papatsoris AG, Papavassiliou AG. Matrix metalloproteinase inhibitors as anticancer agents. Int J Biochem Cell Biol. 2008;40:1156-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
171.  Batra J, Robinson J, Mehner C, Hockla A, Miller E, Radisky DC, Radisky ES. PEGylation extends circulation half-life while preserving in vitro and in vivo activity of tissue inhibitor of metalloproteinases-1 (TIMP-1). PLoS One. 2012;7:e50028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
172.  He M, Wang L, Pu J, Yang Q, Li G, Hao J. Proliferin-related protein overexpression in SGC-7901 gastric cancer cells inhibits in vitro cell growth and tumorigenesis in nude mice. Oncol Rep. 2013;29:2243-2248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
173.  Bai J, Mei P, Zhang C, Chen F, Li C, Pan Z, Liu H, Zheng J. BRG1 is a prognostic marker and potential therapeutic target in human breast cancer. PLoS One. 2013;8:e59772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 77]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
174.  Hadler-Olsen E, Fadnes B, Sylte I, Uhlin-Hansen L, Winberg JO. Regulation of matrix metalloproteinase activity in health and disease. FEBS J. 2011;278:28-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 267]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
175.  Dufour A, Overall CM. Missing the target: matrix metalloproteinase antitargets in inflammation and cancer. Trends Pharmacol Sci. 2013;34:233-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 269]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
176.  Alcantara MB, Dass CR. Regulation of MT1-MMP and MMP-2 by the serpin PEDF: a promising new target for metastatic cancer. Cell Physiol Biochem. 2013;31:487-494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
177.  Clapper ML, Hensley HH, Chang WC, Devarajan K, Nguyen MT, Cooper HS. Detection of colorectal adenomas using a bioactivatable probe specific for matrix metalloproteinase activity. Neoplasia. 2011;13:685-691.  [PubMed]  [DOI]  [Cited in This Article: ]