Skip to main content
Journal of Clinical Oncology logoLink to Journal of Clinical Oncology
. 2009 Sep 8;27(31):5287–5297. doi: 10.1200/JCO.2009.23.5556

Matrix Metalloproteinases As Novel Biomarkers and Potential Therapeutic Targets in Human Cancer

Roopali Roy 1, Jiang Yang 1, Marsha A Moses 1,
PMCID: PMC2773480  PMID: 19738110

Abstract

The matrix metalloproteinase (MMP) family of enzymes is comprised of critically important extracellular matrix remodeling proteases whose activity has been implicated in a number of key normal and pathologic processes. The latter include tumor growth, progression, and metastasis as well as the dysregulated angiogenesis that is associated with these events. As a result, these proteases have come to represent important therapeutic and diagnostic targets for the treatment and detection of human cancers. In this review, we summarize the literature that establishes these enzymes as important clinical targets, discuss the complexity surrounding their choice as such, and chronicle the development strategies and outcomes of their clinical testing to date. The status of the MMP inhibitors currently in US Food and Drug Administration approved clinical trials is presented and reviewed. We also discuss the more recent and successful targeting of this enzyme family as diagnostic and prognostic predictors of human cancer, its status, and its stage. This analysis includes a wide variety of human cancers and a number of human sample types including tissue, plasma, serum, and urine.

INTRODUCTION

Matrix metalloproteases (MMPs) are a multigene family of zinc-dependent endopeptidases that share a similar structure and which collectively, have the capacity to degrade virtually every component of the extracellular matrix (ECM). The basic domain structure of MMP family members is provided in Figure 1. MMP activity is inhibited specifically and reversibly by a group of structurally related, endogenous inhibitors known as tissue inhibitors of metalloproteases (TIMPs). To date, four TIMPs have been identified: TIMP-1, -2, -3, and -4.13 The role of MMPs and TIMPs in tumor growth, metastasis, and angiogenesis has been widely investigated. We refer the reader to a number of comprehensive reviews on this topic47 as well as for a review of the general biochemistry of the MMP family.2,8,9 Based on their substrate specificity, MMPs have been divided into distinct subclasses: collagenases, gelatinases, stromelysins, and matrilysins. However, MMPs exhibit considerable promiscuity with respect to their substrates, leading to considerable redundancy in biologic functions as discussed below.

Fig 1.

Fig 1.

Basic domain structure of matrix metalloprotease (MMP) and a disintegrin and metalloprotease (ADAM) family members. The characteristic domain structure of MMPs includes the signal peptide domain, which guides the enzyme into the rough endoplasmic reticulum during synthesis, the propeptide domain, which sustains the latency of these enzymes until it is removed or disrupted, the catalytic domain, which houses the highly conserved Zn2+ binding region and is responsible for enzyme activity, the hemopexin domain, which determines the substrate specificity of MMPs, and a small hinge region, which enables the hemopexin region to present substrate to the active core of the catalytic domain. The subfamily of membrane-type MMPs (MT-MMPs) possesses an additional transmembrane domain and an intracellular domain. MMPs are produced in a latent form and most are activated by extracellular proteolytic cleavage of the propeptide. MT-MMPs also contain a cleavage site for furin proteases, providing the basis for furin-dependent activation of latent MT-MMPs before secretion. ADAMs are multidomain proteins composed of propeptide, metalloprotease, disintegrin-like, cysteine-rich, and epidermal growth factor–like domains. Membrane-anchored ADAMs contain a transmembrane and cytoplasmic domain. ADAMTSs have at least one thrombospondin type I sequence repeat motif. EGF, epidermal growth factor.

A related family of enzymes, the a disintegrin and metalloprotease (ADAMs), include integral membrane and secreted glycoproteins comprised of two subgroups: the membrane-anchored ADAMs1012 and the secreted ADAMTSs (Fig 1).13 Like MMPs, some ADAM family members have a zinc binding consensus sequence at their catalytic site and display proteolytic activity. ADAMs are multifunctional enzymes involved in ectodomain shedding, regulation of growth factor availability, and in cell-cell/matrix interactions in both normal and pathologic states.1012 Unlike the MMPs, a role for ADAMs in tumorigenesis has only now begun to be explored.

FUNCTIONAL ROLES OF MMPS IN CANCER

Proteolysis of ECM

In most organs, the principle components of the ECM are collagens and numerous other proteins including laminin, entactin, and proteoglycans that make up the basement membrane. Tumor cells overexpress proteases and/or induce expression of these enzymes in neighboring stromal cells in order to degrade the basement membrane and invade the surrounding tissue. Several MMPs have been implicated in the ECM degradation associated with tumor growth and angiogenesis. This proteolytic activity is also required for a cancer cell to invade a nearby blood vessel (intravasation) and then extravasate at a distant location and invade the distant tissue in order to seed a new metastatic site (Fig 2).

Fig 2.

Fig 2.

Multiple functions of matrix metalloproteases (MMPs) in cancer progression. (counterclockwise) MMPs degrade components of extracellular matrix (ECM), facilitating angiogenesis, tumor cell invasion, and metastasis. MMPs modulate the interactions between tumor cells by cleaving E-cadherin, and between tumor cells and ECM by processing integrins, which also enhances the invasiveness of tumor cells. MMPs also process and activate signaling molecules, including growth factors and cytokines, making these factors more accessible to target cells by either liberating them from the ECM (eg, vascular endothelial growth factor [VEGF] and basic fibroblast growth factor [FGF]) and inhibitory complexes (eg, transforming growth factor-β), or by shedding them from cell surface (eg, heparin-binding epidermal growth factor).

Modulation of Cell Adhesion, Migration, and Epithelial to Mesenchymal Transition

ECM degradation products display unique biologic properties that can trigger a variety of cellular signals. For example, cleavage of collagen IV and laminin-5 generates cryptic peptides that can in turn promote migration of tumor cells.14,15 MMP substrates include non-ECM molecules, ranging from growth factor precursors and cell surface adhesion molecules to angiogenic inhibitor precursors (Fig 2). E-cadherin is cleaved by MMP-3, MMP-7, and ADAM10,16,17 leading to the release of soluble E-cadherin and the disruption of cell-cell interactions leading to disruption of cell adhesion and increase in migration. In addition, several integrins can serve as substrates for MMPs (Fig 2). For example, MT1-MMP can process pro-αv, -α5, -α3,18 a process that can contribute to αvβ5-mediated signaling and migration in breast tumor cells.19

MMPs have also been implicated in the epithelial to mesenchymal transition (EMT), a hallmark of cancer progression to metastasis.20 During EMT, tumor cells acquire migratory characteristics and more readily invade into surrounding tissues and metastasize to secondary sites. Activation of growth factors and cleavage of adhesion molecules are some of the proposed mechanisms underlying MMP-induced EMT. Proteolytic activation of latent transforming growth factor–β has been shown to be essential during MMP-28–induced EMT.21 MMP-3–induced EMT has been shown to be the result of E-cadherin cleavage22 and increased expression of an alternatively spliced form of Rac1b.23

Processing of Cytokines and Receptors

Recent studies point to an emerging role for MMPs in modulating aspects of immunity and inflammation during tumorigenesis.24 Cytokine signaling is an integral aspect of inflammation. A variety of cytokines, cytokine receptors, and chemokines have been found to undergo MMP-mediated cleavage. For example, MMP-7 and/or ADAM17 activity is required for release of the proangiogenic inflammatory cytokine tumor necrosis factor (TNF) -α from its membrane-bound form. 2527 In breast cancer, MMP-9 expression is upregulated in tumor-associated stromal cells including neutrophils, macrophages, and lymphocytes28 and may play a role in tumor-associated inflammation.

Processing of Growth Factors and Receptors

Several members of the MMP and ADAM family can regulate cellular proliferation by modulating the bioavailability of growth factors or cell-surface receptors (Fig 2). For example, the bioavailability of insulin-like growth factors (IGFs) is mainly regulated by IGF binding proteins (IGFBP). MMP-1, -2, -3, and ADAM12 cleave IGFBP-3,2931 while IGFBP-1 is a substrate for MMP-11.32 Ligands for several growth factor receptors are processed by MMP/ADAM family members as well. Chief among them are the epidermal growth factor (EGF) receptor ligands: heparin-binding EGF (HB-EGF), amphiregulin, betacellulin, heregulin, and epiregulin. Normally, signaling via the EGF receptor (EGFR) pathway is tightly controlled. In cancer, as a consequence of increased shedding of active EGFR ligands and induction of constitutively active EGFR kinases, signaling through these pathways is upregulated, resulting in uncontrolled proliferation, migration, and survival of cancer cells. MMP-3, -7, ADAM17, and ADAM12 can proteolytically release HB-EGF allowing it to transactivate the EGFR3335 and in turn promote tumor growth and angiogenesis.36 ADAM17 overexpression in breast cancer cells can lead to increased proliferation, migratory, and invasive capacity of cancer cells,37 properties that can be attributed in part, to the increased shedding of basement membrane transforming growth factor–α and amphiregulin in these cells.38,39

MMPs have been shown to promote angiogenesis through their release of angiogenic factors stored in the ECM such as vascular endothelial growth factor (VEGF)40,41 and basic fibroblast growth factor (bFGF; Fig 2).42 Stroma-derived MMP-9 can facilitate the liberation of ECM-sequestered VEGF during tumor angiogenesis.41 Similarly, cleavage of perlecan by MMP-1 and MMP-3 releases active bFGF.42

Role in Tumor-Associated Angiogenesis

Angiogenesis is the formation of new blood vessels from a preexisting one. It is widely believed that a tightly controlled balance of pro- and antiangiogenic molecules regulates angiogenesis in tumors. MMPs play complex and sometimes conflicting roles in regulating angiogenesis. Remodeling of the ECM during angiogenesis is accomplished largely through the activity of MMPs.4,8,43 Angiogenic mitogens, such as bFGF and VEGF, can stimulate the production of MMPs by capillary endothelial cells.44,45 Studies have also demonstrated that MMPs are involved in the angiogenic switch, one of the earliest stages of tumor growth and progression. In a model of tumor progression which reliably recapitulates this switch, MMP-2 was shown to play an important role in the development of the angiogenic phenotype.46 It has also been shown that MMP-9 can be a regulator of the angiogenic switch in a pancreatic tumor model,41 further confirming the pro-angiogenic role of MMPs. These findings strongly suggest that MMP activity is critical, not only to the initiation of angiogenesis, but to the maintenance of the growing vascular bed, which in turn supports tumor growth and metastasis. MMP activity can, however, result in the production of negative regulators of angiogenesis as well. For example, MMPs have now been shown to cleave the parent molecules plaminogen and collagen XVIII into the cryptic, endogenous angiogenesis inhibitors, angiostatin47,48 and endostatin,49 respectively. These data underline the importance of MMPs as both positive and negative regulators of angiogenesis and cancer.

MMPS AS BIOMARKERS OF CANCER

One of the more promising and exciting applications of MMPs in human cancers is as potential cancer biomarkers, both diagnostic and prognostic. Below we review their exploitation as tools for early detection, disease progression, and metastasis. We have specifically included those biomarkers for which validation studies have accompanied the preliminary discovery analyses (Table 1).5096

Table 1.

Candidate MMP and ADAM Biomarkers of Cancer

Type of Cancer and MMPs/ADAMs Detected in Tissue/Body Fluid Method of Analysis
Breast
    MMP-1350 Tissue IHC
    MMP-9, TIMP-151 Serum, tissue ELISA, IHC
    MMP-95254 Urine, serum, plasma, tissue Gelatin zymography, IHC
    ADAM1255 Urine Immunoblot
    ADAM1756 Tissue ELISA, immunoblot
    MMP-157 Tissue, nipple aspirates Gene analysis
Pancreas
    MMP-958 Pancreatic juice, serum ELISA, immunoblot
    MMP-259 Pancreatic juice, tissue Gelatin zymography
    MMP-760,61 Tissue, plasma IHC, RT-PCR, ELISA
    ADAM962 Tissue IHC
Lung
    MMP-9, TIMP-163,64 Serum, bronchial lavage ELISA
    MMP-765 Tissue IHC
    MMP-166,67 Tissue Gene analysis
Bladder
    MMP-968 Tissue IHC
    MMP-9, MMP-269,70 Urine Gelatin zymography
    MMP-971,72 Urine Gelatin zymography, immunoblot, ELISA
    MMP-9, telomerase73 Urine Gelatin zymography, cytology
Colorectal
    MMP-274,75 Tissue, plasma IHC, ELISA
    MMP-976 Tissue IHC
    MMP-2, MMP-977 Plasma ELISA
    MMP-778 Serum ELISA
    MMP-179 Tissue ELISA
    MMP-1380 Tissue Gelatin zymography
Ovarian
    MMP-981 Tissue Gelatin zymography
    MMP-9, MMP-1482 Tissue IHC
    MMP-282,83 Tissue IHC
    MMP-2, MMP-9, MMP-1484 Tissue IHC
    ADAM1785 Tissue RT-PCR, IHC
Prostate
    MMP-2, MMP-986,87 Plasma, tissue ELISA, IHC
    MMP-288 Tissue IHC
    MMP-970 Urine Gelatin zymography
    ADAM889 Tissue IHC
    ADAM990 Tissue IHC
Brain
    MMP-291,92 Tissue IHC, ELISA, gelatin zymography
    MMP-993,94 Tissue IHC, ELISA, gelatin zymography
    MMP-2, MMP-995,96 Tissue, cerebrospinal fluid, urine IHC, ELISA, gelatin zymography

Note: For each of the studies listed above, N ≥ 40, with the exception of brain tumor biomarker studies where larger sample sizes were not available.

Abbreviations: MMP, matrix metalloproteinase; ADAM, a disintegrin and metalloprotease; IHC, immunohistochemistry; ELISA, enzyme-linked immunosorbent assay; RT-PCR, reverse transcription polymerase chain reaction.

Breast Cancer

Evidence is emerging that members of the MMP and/or ADAM family can serve not only as potential markers for diagnosis and prognosis, early detection, and risk assessment, but also as indicators of tumor recurrence, metastatic spread, and response to primary and adjuvant therapy for breast cancer. MMP-9 levels in tumor tissue as well as serum, plasma, and urine are significantly elevated in patients with breast cancer.5153,71

We have previously reported that the detection of urinary ADAM12 in patients with breast cancer is predictive of disease status and stage and that ADAM12 protein levels in urine increase with progression of disease.55 Expression of a related enzyme, ADAM17, is significantly elevated in high-grade breast tumor tissue and correlates with shorter overall patient survival.56 Immunohistochemical (IHC) analysis of human breast biopsy materials have indicated that MMP-13 may be a useful prognostic indicator for invasive breast cancer. In this study, tumor-derived MMP-13, correlated with expression of Her2/neu and TIMP-1 and with aggressive tumor phenotypes and inversely correlated with overall patient survival.50

Recently, efforts have focused on the use of MMPs and ADAMs as potential biomarkers of early breast cancer. Studies from our laboratory indicate that urinary MMP-9 and ADAM12, in addition to being predictive markers for breast cancer, may also prove useful as noninvasive breast cancer risk assessment tools.97 Differential global gene expression analysis of tissue samples from patients with atypical ductal hyperplasia and a history of cancer versus patients with no history or who did not subsequently develop breast cancer indicated that MMP-1 mRNA and protein levels were upregulated in precancerous lesions.57 MMP-1 mRNA could also be detected in cells collected from nipple aspirates suggesting that MMP-1 might be useful as a diagnostic marker for screening atypical ductal hyperplasia to identify women with lesions that may eventually develop into cancer.57

Metastatic breast cancer involves the regional lymph nodes (LN) and liver or bone resulting in significant morbidity. Several independent studies have used circulating MMP-9 activity to predict metastatic spread of disease as well as to monitor patient response to primary and adjuvant therapy and to evaluate outcome.54,98 High levels of serum MMP-9 and TIMP-1 are associated with increased incidence of LN metastasis and decreased relapse-free and overall survival rates.51 MMPs may also be useful in predicting therapeutic efficacy. Plasma MMP-9 levels decrease after the surgical removal of primary breast tumors and a progressive decrease in plasma MMP-9 was observed in patients who responded well to adjuvant therapy.54 Importantly, in all patients who suffered a relapse of disease there was a gradual increase of plasma MMP-9 activity 1 to 8 months before the clinical diagnosis of recurrence.54 High MMP-14 mRNA expression (> 10% cells) in primary breast tumor tissue can also predict shorter overall survival.99

Pancreatic Cancer

Nowhere is the need more urgent for sensitive and specific biomarkers for early diagnosis and to screen high-risk patients than in pancreatic cancer. This disease is extremely difficult to diagnose in its early stages due to a lack of specific symptoms and the limitations of current diagnostic methods. Several studies have evaluated differentially expressed biomarkers for pancreatic cancer using tissue, blood, or pancreatic juice. Serum and tissue levels of MMP-9 are significantly higher in patients with pancreatic ductal adenocarcinoma than in patients with chronic pancreatitis and healthy controls.58 Active MMP-2 levels are upregulated in the pancreatic juice of patients with cancer (100%) as compared with patients with chronic pancreatitis (2%) or normal controls (0%).59 Similarly, plasma as well as tumor tissues from patients with pancreatic ductal adenocarcinoma have significantly elevated MMP-7 levels60 which may predict shortened survival of patients.61

Of the ADAMs studied to date, only ADAM9 has been analyzed in relation to pancreatic cancer. IHC analysis of ADAM9 in tumor tissue samples indicated that an increased expression of this protease correlates with poor tumor differentiation and shortened overall survival.62

Lung Cancer

Several studies have reported that plasma and/or serum levels of MMP-9 and TIMP-1 are elevated in patient with stage III or IV lung cancer when compared with those in patients with nonmalignant lung diseases.63,64 Retrospective studies of non–small-cell lung cancer (NSCLC) tissue found that MMP-7 expression was higher in squamous cell carcinomas than in adenocarcinomas and correlated with significantly lower overall survival in patients.65 In a similar study that included 159 patients with stage III and IV NSCLC, MMP-7 status correlated inversely with overall response to chemotherapy. These findings suggest that MMP-7 expression in NSCLC may be a significant prognostic factor and could be predictive of response to chemotherapy and outcome. MMP-1 overexpression has been reported in lung cancer cells100 and DNA variants of MMP-1 have been linked to lung cancer risk and susceptibility.66 Several single nucleotide polymorphisms within the MMP-1 gene have been shown to be significantly associated with the risk of early-onset lung cancer in particular for subgroups with high smoking intensity.67

Studies of ADAM-type proteases as biomarkers for lung cancer are rare. One exception is ADAM28, whose levels were found to be higher in lung carcinoma tissue compared with healthy lung tissue and correlated significantly with tumor size and LN metastasis.101

Bladder Cancer

As is expected, a majority of the biomarker studies in patients with bladder cancer have focused on urine. Studies from our group and others have shown that urinary MMP-2 and MMP-9 levels correlate with presence of bladder cancer as well as stage and grade of disease.6973 We have recently reported the identification of several MMP species in urine from patients with primary tumors in the bladder and prostate including MMP-2, MMP-9, MMP-9/neutrophil gelatinase-associated lipocalin complex and MMP-9 dimer.70 Each urinary MMP species was detected at significantly higher rates in urine from patients with cancer as compared with controls. The difference in detection of MMP species in the urine of the two types of cancers studied may serve as a tumor-specific fingerprint that can indicate both the presence of a tumor as well as its location.70 Increased levels of MMP-9 and MMP-2 in urine correlate with increased expression of these proteases in bladder tumor tissue as well.68 Urinary MMP-9 levels when combined with telomerase analysis of exfoliated cells from voided urine could also increase the sensitivity of cytology, a commonly used method for bladder cancer detection and monitoring.73 ADAM12 mRNA and protein were found to be upregulated in bladder cancer tissue and urinary ADAM12 levels were higher in patients with bladder cancer.102

Colorectal Cancer

MMP-2 and MMP-9 have been studied as potential prognostic biomarkers of colorectal cancer. A study of patients with Dukes' stages A to D colorectal cancer found that high levels of MMP-2, but not MMP-1, -7, and -13, in the malignant epithelium and stroma were associated with decreased survival.74 Elevated plasma MMP-2 levels have also been shown to correlate with lymph node metastasis.75 Enhanced MMP-9 staining in primary tumors was found to be an independent marker of poor prognosis in a study with T3-T4 node-negative patients.76 Plasma MMP-2 and MMP-9 levels were significantly elevated in patients with colorectal cancer and those with adenomatous polyps, and significant reduction in both were observed after tumor resections, suggesting their potential as markers for therapeutic efficacy.77 These MMPs may not be prognostic markers for tumor recurrence, however, since plasma proMMP-2 and -9 activities did not correlate with disease relapse after surgery.103

In addition to the gelatinases, serum MMP-7 levels were reported to predict decreased survival in patients with advanced colorectal cancer.78 A study of paired colorectal tumor and normal mucosal tissues revealed the significant correlation between MMP-1 levels and pathology (ie, Dukes' stage, tumor depth, and lymphatic invasion).79 MMP-13 activity was significantly elevated in tumor samples and the tumor to normal tissue ratio of MMP-13 activity significantly correlated with poor survival.80

Ovarian Cancer

MMP-2, -9, and -14 are among the most studied MMPs as biomarkers for ovarian cancer. MMP-9 activity in tissue extracts was significantly increased in advanced ovarian cancers (International Federation of Gynecology and Obstetrics stage III) compared with benign tumors and was found to be an independent prognosticator of poor survival.81 In another study of invasive epithelial ovarian cancer, high stromal expressions of MMP-9 and -14 were significantly correlated with cancer progression and were independent prognostic markers.82 Correlation with ovarian cancer progression has also been reported for MMP-282,104 and elevated levels of MMP-2 in cancer cells of peritoneal implants were associated with a significant risk of death in stage III ovarian carcinomas.83

Tissue MMPs have also been shown to distinguish different histotypes of ovarian cancer, which is a significant finding given that different histotypes have different prognoses.105 A recent study showed that more than 90% of clear-cell carcinomas expressed moderate to high levels of MMP-2 or MMP-14, compared with 30% to 55% of the other ovarian cancer histotypes (serous, endometroid, and mucinous), whereas MMP-9 was expressed more widely in other histotypes.84 Importantly, the cellular source of MMPs must be considered when evaluating MMPs as ovarian cancer biomarkers. For example, strong MMP-9 levels in cancer cells were associated with longer survival whereas strong stromal MMP-9 was associated with shorter survival, suggesting a dual role for MMP-9 during ovarian cancer progression.106

Among the ADAM family members, ADAM17 expression was significantly increased in both early and advanced ovarian cancer tissues and correlated with the expression of HB-EGF.85

Prostate Cancer

MMP-2, -9, -15, and -26 expression in tissue or serum have been positively correlated with Gleason score in prostate cancer.107109 Among these MMPs, the activities of plasma MMP-2 and -9 increased significantly in metastatic prostate cancer.86 Furthermore, overexpression of MMP-2 in cancer tissue was associated with shorter disease-free survival in a study with T3N0-2M0 patients.88 Analysis of MMP-2 and -9 levels in radical prostatectomy specimens revealed these two as significant predictors of cancer recurrence.87 These two enzymes may also be markers of therapeutic efficacy, since both the levels and activities of plasma MMP-2 and -9 decreased significantly in metastatic patients after therapy.86 In addition, increased urinary MMP-9 activity has been shown to distinguish between prostate and other types of cancer (eg, bladder cancer).70 MMPs can also be combined with other markers to increase their predictive capability. For example, the mRNA ratio of gelatinases (MMP-2 and MMP-9) to E-cadherin in biopsy samples independently predicted prostate cancer stage.110

ADAM8 levels in prostate cancer tissue have been significantly correlated with higher tumor status, positive nodal status, and higher Gleason scores.89 Higher ADAM9 levels were associated with shortened prostate-specific antigen relapse-free survival.90

Brain Tumors

Elevated tissue levels of MMP-2 and MMP-9 have been reported in aggressive brain tumors.9194 Positive MMP-2 expression in tissue was also associated with shorter survival in patients with malignant brain tumors.92 Both latent and activated forms of MMP-2 and MMP-9 have been detected in the cerebrospinal fluid of patients with brain tumors.95 In studies of primary glial tumors and other CNS tumors, we have recently shown that detection of MMP-2, MMP-9, MMP-9/neutrophil gelatinase-associated lipocalin complex, and/or VEGF in the urine predicted disease status and therapeutic efficiency of patients with brain cancer.96,111 Importantly, these studies showed that the upregulation of MMP-2 and -9 in the source tumor tissue was also reflected in CSF as well as in urine of these patients.

It is important to note that the measurement of MMPs in body fluids, in particular serum or plasma, can be influenced by the type of fluid and method of collection and storage. For example, basal MMP-9 levels in serum/plasma can be influenced by the use of EDTA or heparin,112 a problem that can be alleviated by using sodium citrate instead.113 Another issue to be considered is that of sample storage. For example, it has been reported that plasma MMP-9 is unstable and degrades rapidly even when stored at −80°C.114,115 This problem can be alleviated by storing samples in liquid nitrogen and analyzing them shortly after collection.

The context in which these or any other biomarkers are utilized in the clinic is a critical consideration. It has been suggested that, rather than being used as screening tools, MMPs might best be used to provide useful clinical information as part of a longitudinal assessment of a patient's disease progression and therapeutic efficacy with the patient himself/herself serving as an internal control.

MMPS AS THERAPEUTIC TARGETS

Given the important roles that MMPs play in tumor growth, metastasis, and the dysregulated angiogenesis that drives them, there has been significant attention paid to the development of clinically useful antagonists of this enzyme family. There are a number of matrix metalloproteinase inhibitors (MMPIs) that are currently being tested in all three phases of clinical trials against a variety of human cancers (Table 2).116119 The promise of this therapeutic approach has yet to be realized and the academic, pharmaceutical, and biotechnology arenas continue to debate the potential issues underlying the lack of therapeutic success in cancer treatment. Although certainly not in the majority, there have been some promising results from some clinical trials. For example, Neovastat administration resulted in a significantly longer median survival time in patients with refractory renal cell carcinoma in a phase II trial.120 However, in many cases, when there appeared to be positive results, the reports indicated that although biomarker levels decreased in the course of the treatment, positive clinical correlates were not necessarily observed.

Table 2.

MMP Inhibitors Currently in Clinical Trials for Cancer

Drug by Phase Type of Cancer Type of Drug Target MMP
BMS-27591
    II Prostate Nonhydroxamate MMP-1, 2, 8, 9, 14
    III Non–small-cell lung Nonpeptidomimetic
COL-3
    I Advanced solid tumors Chemically modified tetracycline MMP-2, 9
    II Kaposi's sarcoma
Dalteparin (trials completed)*
    II Glioblastoma Low molecular weight synthetic heparin MMP-9
    III Advanced cancers (breast, lung, colon, prostate)
Disulfiram
    I Melanoma, solid tumors, non–small-cell lung Tetraethyliuram disulfide MMP-2, 9
    II/III
Genistein
    II Breast, kidney, melanoma, prostate, bladder, pancreatic, breast cancer prevention Soy isoflavone MMP-2, 9
INCB7839
    I/II Breast Selective sheddase inhibitor ADAM10, 17
Marimastat
    III Breast Hydroxamate peptidomimetic MMP-1, 2, 3, 7, 9, 12
Neovastat (AE941; trials completed)
    II Multiple myeloma Shark cartilage extract MMP-2, 9, 12
    III Non–small-cell lung, kidney, breast, colorectal
PCK 3145 (trials completed)
    I Prostate Prostate secretory protein MMP-9
94-derived synthetic peptide
Prinomastat (AG3340)
    II Glioblastoma, non–small-cell lung, prostate Hydroxamate MMP-2, 9, 13, 14
    III Nonpeptidomimetic

Abbreviations: MMP, matrix metalloproteinase; ADAM, a disintegrin and metalloprotease.

*

Completed trials in glioblastoma116 and advanced cancers.117

Completed trials in non-small cell lung cancer118 and breast and colon cancer.119

Trials have been completed but results are unpublished.

Current Design of MMPIs

Peptidomimetic MMPIs are pseudopeptide derivatives that mimic the structure of collagen at the MMP cleavage site.121 These substrate-based MMPIs are usually broad spectrum and block MMP activity by occupying the substrate-binding site and chelating the zinc with, in most cases, a hydroxamic acid functional group. The earliest generation of these inhibitors, including batimastat (BB-94), had low water solubility and was therefore not orally available. The next generation of hydroxamate-based inhibitors, such as marimastat (BB-2516), was designed to be orally available, however, they were commonly associated with musculoskeletal syndrome, probably due to their off-target effects on non-MMP metalloproteinases.122

To improve specificity, the current knowledge of the three-dimentional conformation of the enzyme active site has been incorporated into the design of MMPIs. These structure-based inhibitors include tanomastat (BAY 12-9566; Bayer Corporation, West Haven, CT), prinomastat (AG3340; Agouron Pharmaceuticals, La Jolla, CA), and BMS-275291 Bristol-Myers-Squibb, New York, NY. Some of these inhibitors also contain hydroxamic acid group to chelate zinc, such as prinomastat. Musculoskeletal toxicity has also been reported in clinical trials with prinomastat and BMS-275291.123,124 These MMPIs, while not as broad spectrum as the substrate-based MMPIs, still target several enzymes with similar potency. New structure-based inhibitors targeting a single MMP have been described for MMP-12 and -13.125,126

Another group of MMPIs are the chemically modified tetracyclines (CMTs), which do not possess antibiotic activities.127,128 CMTs may inhibit MMPs by binding to the key metal ions, such as zinc and calcium, or by regulating MMP transcription.129 CMTs used as MMPIs include metastat (COL-3), minocycline, and doxycycline.

Novel mechanism-based MMPIs have also been reported.130 One of the first of such inhibitors, SB-3CT, was designed to be highly selective for gelatinases. It covalently binds to the active site of MMP-2 and restructures the enzyme back to its proenzyme state.131 It reduced liver metastasis and increased survival in an aggressive mouse model of T-cell lymphoma.132 A new variant of this inhibitor class further improves the specificity by exclusively targeting MMP-2.133

Several small molecule inhibitors targeted specifically to the ADAM family of enzymes have also been recently evaluated.134136 Of these, INCB7839 (Incyte Corporation, Wilmington, DE), a sheddase inhibitor, is currently in phase II clinical trials against breast carcinoma and several other solid tumors. Such inhibitors could prove to be particularly useful in targeting tumors dependant on EGFR signaling either as single agents or in a synergistic manner with currently approved tyrosine kinase inhibitors. Notably, unlike the first generation MMPIs, INCB7839 did not induce musculoskeletal adverse effects in initial animal studies.136

Far less attention has been given to the issues limiting the use of the TIMPs in the clinical setting, with the key limitations being the difficulty in the large scale production of biologically active protein and optimization of drug delivery. Just as is the case with their cognate enzymes, the TIMPs have also now been shown to be multifunctional proteins with activities independent of their MMP inhibitory ones.4,137,138 Interestingly, there may be significant clinical potential in exploiting these non-MMP activities in that this strategy might permit circumvention of the limitations associated with the targeting of MMP inhibition alone.

Challenges of Anti-MMP Therapy

The road to clinical use of MMPIs has not been straightforward. The reader is referred to a number of recent reviews for an extensive analysis of the design, development, and complexities that have slowed and limited the creation and mobilization of MMPIs into the clinic.139,140 Several reasons might explain the unfavorable clinical outcomes in some of the MMPI clinical trials in various cancer types. Firstly, adverse effects, including musculoskeletal syndrome, have limited the maximum-tolerated dose of the early generation of MMPIs, thereby limiting drug efficacy. Secondly, the best window for MMPI treatment may have been missed in patients recruited in the trials who are often at the most advanced, metastatic stage of cancer. In addition, the nonspecific nature of the inhibitors has negatively impacted the therapeutic efficacy of these inhibitors due, at least in part, to the wide range of MMPs and physiological events affected.

As is the case for many anticancer drugs, MMPIs may actually be more effective when administered at earlier stages of cancer progression rather than when a patient is suffering from end stage disease having experienced failure with all other conventional therapies. Therapy at an earlier stage in disease progression might also afford the opportunity to use lower doses of drug thereby perhaps limiting the toxicity that has so hampered these trials. Interestingly, Hanahan et al141 demonstrated that MMPIs (in this case the broad spectrum, hydroxamate-based BB-94) would be most useful when tested in the very early stages of tumor progression, in the RIP1-Tag2 model of pancreatic carcinogenesis, such as at the time of the angiogenic switch.

As discussed earlier, MMPs can be multifaceted during cancer progression. Several MMPs have been shown to be protective against cancer. Tumor progression in various animal models has been inversely correlated with the expression status of MMP-3, -8, and -12.142144 In human studies, MMP-12 overexpression in the tumor has also been associated with more positive prognosis.145 Importantly, the same MMP may play an opposite role at different stages of cancer progression. For example, a study with a transgenic mouse model of invasive squamous cell carcinoma suggested that MMP-9 stimulates proliferation during the early stages of cancer but restricts further malignant progression at later stages.146 For many of these MMPs, it remains unclear exactly which effect, positive or negative, they are exerting and these effects can vary as a function of different cancer types and different stages of disease.147

The mechanisms underlying the putative protective activities of MMPs are not fully understood. One potential mechanism may be the processing of antiangiogenic factors (cryptic angiogenic inhibitors) from inactive parental proteins.4749,148,149 Another possible mechanism is the participation of MMPs in the host defense against a tumor143,150 by modulating the activities of chemokines and cytokines.151

Another major challenge in developing MMP-targeted therapy is the accurate evaluation of MMPI efficacy in vivo. To overcome this hurdle, a MMP-2-sensitive imaging probe has been studied which contains a MMP-2 peptide substrate and quenched near-infrared fluorochromes.152 Fluorescence is detected when the peptide is cleaved by active MMP-2. A significant reduction in probe fluorescence in implanted tumors was detected in live mice treated with MMPI prinomastat. These noninvasive imaging techniques make it possible to evaluate the therapeutic efficacy of MMPIs in patients and may also be used to monitor MMP activity at different stages of cancer progression. Taken together, such techniques provide insight into the functions of MMPs in cancer and may lead to effective MMP-targeting therapeutics.

Acknowledgment

The authors gratefully acknowledge Kristin Johnson for graphics assistance and Susan Connors for her contributions. Due to space constraints, we have in some cases been forced to cite review articles as opposed to the original articles.

Glossary Terms

Angiogenesis:

The process involved in the generation of new blood vessels. While this is a normal process that naturally occurs and is controlled by “on” and “off” switches, blocking tumor angiogenesis (antiangiogenesis) disrupts the blood supply to tumors, thereby preventing tumor growth.

Biomarker:

A functional biochemical or molecular indicator of a biologic or disease process that has predictive, diagnostic, and/or prognostic utility.

MMP (matrix metalloprotease [metalloproteinases]):

MMPs belong to a family of enzymes (zinc-dependent endoproteinases) that are involved in the degradation of the extracellular matrix. MMPs are involved in both normal and pathologic tissue remodeling, where their selective proteolysis is now appreciated to help regulate cell growth, angiogenesis, and invasiveness.

Prognostic (prognostic marker):

A marker that predicts the prognosis of a patient (eg, the likelihood of relapse, progression, and/or death) independent of future treatment effects. A factor can be both prognostic and predictive.

ECM (extracellular matrix):

Components that are extracellular and composed of secreted fibrous proteins (eg, collagen) and gel-like polysaccharides (eg, glycosaminoglycans) binding cells and tissues together. In addition, the ECM contains adhesion proteins (eg, fibronectin and laminin) that link components of the matrix both to one another and to attached cells. Depending on the tissue, the composition of the ECM differs. For example, collagen is the major component of ECM; elastin fibers contain cross-linked elastin; and integrins are cell surface receptors responsible for the attachment of cells to the ECM.

ADAMs (a disintegrin and metalloprotease):

A family of integral membrane and secreted glycoproteins. ADAMs are multifunctional enzymes involved in cell-surface remodeling, ectodomain shedding, regulation of growth factor availability, ECM degradation, and mediation of cell-cell and cell-matrix interactions in both normal development as well as in pathological states.

MMPIs (matrix metalloproteinase inhibitor):

Natural proteins or synthesized compounds that inhibit the activity of one or more MMPs.

Footnotes

Supported by Grants No. R01 CA118764 and P01 CA45548 from the National Institutes of Health, and by the Ellison Foundation.

Terms in blue are defined in the glossary, found at the end of this article and online at www.jco.org.

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Although all authors completed the disclosure declaration, the following author(s) indicated a financial or other interest that is relevant to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for more information about ASCO's conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of Potential Conflicts of Interest section in Information for Contributors.

Employment or Leadership Position: None Consultant or Advisory Role: Roopali Roy, Predictive Biosciences, Inc. (C); Marsha A. Moses, Predictive Biosciences, Inc. (C) Stock Ownership: Marsha A. Moses, Predictive Biosciences, Inc. Honoraria: None Research Funding: None Expert Testimony: None Other Remuneration: None

AUTHOR CONTRIBUTIONS

Conception and design: Roopali Roy, Jiang Yang, Marsha A. Moses

Financial support: Marsha A. Moses

Manuscript writing: Roopali Roy, Jiang Yang, Marsha A. Moses

Final approval of manuscript: Roopali Roy, Jiang Yang, Marsha A. Moses

REFERENCES

  • 1.Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 2006;69:562–573. doi: 10.1016/j.cardiores.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 2.Murphy G, Nagase H. Progress in matrix metalloproteinase research. Mol Aspects Med. 2008;29:290–308. doi: 10.1016/j.mam.2008.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Stetler-Stevenson WG. Tissue inhibitors of metalloproteinases in cell signaling: Metalloproteinase-independent biological activities. Sci Signal. 2008;1:re6. doi: 10.1126/scisignal.127re6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Harper J, Moses MA. Molecular regulation of tumor angiogenesis: Mechanisms and therapeutic implications. EXS. 2006:223–268. doi: 10.1007/3-7643-7378-4_10. [DOI] [PubMed] [Google Scholar]
  • 5.Roy R, Zhang B, Moses MA. Making the cut: Protease-mediated regulation of angiogenesis. Exp Cell Res. 2006;312:608–622. doi: 10.1016/j.yexcr.2005.11.022. [DOI] [PubMed] [Google Scholar]
  • 6.Cruz-Munoz W, Khokha R. The role of tissue inhibitors of metalloproteinases in tumorigenesis and metastasis. Crit Rev Clin Lab Sci. 2008;45:291–338. doi: 10.1080/10408360801973244. [DOI] [PubMed] [Google Scholar]
  • 7.van Hinsbergh VW, Koolwijk P. Endothelial sprouting and angiogenesis: Matrix metalloproteinases in the lead. Cardiovasc Res. 2008;78:203–212. doi: 10.1093/cvr/cvm102. [DOI] [PubMed] [Google Scholar]
  • 8.Matrisian LM, Wright J, Newell K, et al. Matrix-degrading metalloproteinases in tumor progression. Princess Takamatsu Symp. 1994;24:152–161. [PubMed] [Google Scholar]
  • 9.Bode W, Maskos K. Structural studies on MMPs and TIMPs. Methods Mol Biol. 2001;151:45–77. [PubMed] [Google Scholar]
  • 10.Stone AL, Kroeger M, Sang QX. Structure-function analysis of the ADAM family of disintegrin-like and metalloproteinase-containing proteins. J Protein Chem. 1999;18:447–465. doi: 10.1023/a:1020692710029. [DOI] [PubMed] [Google Scholar]
  • 11.Edwards DR, Handsley MM, Pennington CJ. The ADAM metalloproteinases. Mol Aspects Med. 2008;29:258–289. doi: 10.1016/j.mam.2008.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Murphy G. The ADAMs: Signaling scissors in the tumour microenvironment. Nat Rev Cancer. 2008;8:929–941. doi: 10.1038/nrc2459. [DOI] [PubMed] [Google Scholar]
  • 13.Apte SS. A disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motifs: The ADAMTS family. Int J Biochem Cell Biol. 2004;36:981–985. doi: 10.1016/j.biocel.2004.01.014. [DOI] [PubMed] [Google Scholar]
  • 14.Xu J, Rodriguez D, Petitclerc E, et al. Proteolytic exposure of a cryptic site within collagen type IV is required for angiogenesis and tumor growth in vivo. J Cell Biol. 2001;154:1069–1079. doi: 10.1083/jcb.200103111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Koshikawa N, Giannelli G, Cirulli V, et al. Role of cell surface metalloprotease MT1-MMP in epithelial cell migration over laminin-5. J Cell Biol. 2000;148:615–624. doi: 10.1083/jcb.148.3.615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Noe V, Fingleton B, Jacobs K, et al. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 2001;114:111–118. doi: 10.1242/jcs.114.1.111. [DOI] [PubMed] [Google Scholar]
  • 17.Maretzky T, Reiss K, Ludwig A, et al. ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and beta-catenin translocation. Proc Natl Acad Sci U S A. 2005;102:9182–9187. doi: 10.1073/pnas.0500918102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ratnikov BI, Rozanov DV, Postnova TI, et al. An alternative processing of integrin alpha(v) subunit in tumor cells by membrane type-1 matrix metalloproteinase. J Biol Chem. 2002;277:7377–7385. doi: 10.1074/jbc.M109580200. [DOI] [PubMed] [Google Scholar]
  • 19.Baciu PC, Suleiman EA, Deryugina EI, et al. Membrane type-1 matrix metalloproteinase (MT1-MMP) processing of pro-alphav integrin regulates cross-talk between alphavbeta3 and alpha2beta1 integrins in breast carcinoma cells. Exp Cell Res. 2003;291:167–175. doi: 10.1016/s0014-4827(03)00387-2. [DOI] [PubMed] [Google Scholar]
  • 20.Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442–454. doi: 10.1038/nrc822. [DOI] [PubMed] [Google Scholar]
  • 21.Illman SA, Lehti K, Keski-Oja J, et al. Epilysin (MMP-28) induces TGF-beta mediated epithelial to mesenchymal transition in lung carcinoma cells. J Cell Sci. 2006;119:3856–3865. doi: 10.1242/jcs.03157. [DOI] [PubMed] [Google Scholar]
  • 22.Lochter A, Galosy S, Muschler J, et al. Matrix metalloproteinase stromelysin-1 triggers a cascade of molecular alterations that leads to stable epithelial-to-mesenchymal conversion and a premalignant phenotype in mammary epithelial cells. J Cell Biol. 1997;139:1861–1872. doi: 10.1083/jcb.139.7.1861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Radisky DC, Levy DD, Littlepage LE, et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature. 2005;436:123–127. doi: 10.1038/nature03688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Noel A, Jost M, Maquoi E. Matrix metalloproteinases at cancer tumor-host interface. Semin Cell Dev Biol. 2008;19:52–60. doi: 10.1016/j.semcdb.2007.05.011. [DOI] [PubMed] [Google Scholar]
  • 25.Chandler S, Cossins J, Lury J, et al. Macrophage metalloelastase degrades matrix and myelin proteins and processes a tumour necrosis factor-alpha fusion protein. Biochem Biophys Res Commun. 1996;228:421–429. doi: 10.1006/bbrc.1996.1677. [DOI] [PubMed] [Google Scholar]
  • 26.Haro H, Crawford HC, Fingleton B, et al. Matrix metalloproteinase-7-dependent release of tumor necrosis factor-alpha in a model of herniated disc resorption. J Clin Invest. 2000;105:143–150. doi: 10.1172/JCI7091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Black RA, Rauch CT, Kozlosky CJ, et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature. 1997;385:729–733. doi: 10.1038/385729a0. [DOI] [PubMed] [Google Scholar]
  • 28.Benaud C, Dickson RB, Thompson EW. Roles of the matrix metalloproteinases in mammary gland development and cancer. Breast Cancer Res Treat. 1998;50:97–116. doi: 10.1023/a:1006061115909. [DOI] [PubMed] [Google Scholar]
  • 29.Thrailkill KM, Quarles LD, Nagase H, et al. Characterization of insulin-like growth factor-binding protein 5-degrading proteases produced throughout murine osteoblast differentiation. Endocrinology. 1995;136:3527–3533. doi: 10.1210/endo.136.8.7543045. [DOI] [PubMed] [Google Scholar]
  • 30.Fowlkes JL, Enghild JJ, Suzuki K, et al. Matrix metalloproteinases degrade insulin-like growth factor-binding protein-3 in dermal fibroblast cultures. J Biol Chem. 1994;269:25742–25746. [PubMed] [Google Scholar]
  • 31.Loechel F, Wewer UM. Activation of ADAM 12 protease by copper. FEBS Lett. 2001;506:65–68. doi: 10.1016/s0014-5793(01)02873-3. [DOI] [PubMed] [Google Scholar]
  • 32.Manes S, Mira E, Barbacid MM, et al. Identification of insulin-like growth factor-binding protein-1 as a potential physiological substrate for human stromelysin-3. J Biol Chem. 1997;272:25706–25712. doi: 10.1074/jbc.272.41.25706. [DOI] [PubMed] [Google Scholar]
  • 33.Suzuki M, Raab G, Moses MA, et al. Matrix metalloproteinase-3 releases active heparin-binding EGF-like growth factor by cleavage at a specific juxtamembrane site. J Biol Chem. 1997;272:31730–31737. doi: 10.1074/jbc.272.50.31730. [DOI] [PubMed] [Google Scholar]
  • 34.Schafer B, Marg B, Gschwind A, et al. Distinct ADAM metalloproteinases regulate G protein-coupled receptor-induced cell proliferation and survival. J Biol Chem. 2004;279:47929–47938. doi: 10.1074/jbc.M400129200. [DOI] [PubMed] [Google Scholar]
  • 35.Sahin U, Weskamp G, Kelly K, et al. Distinct roles for ADAM10 and ADAM17 in ectodomain shedding of six EGFR ligands. J Cell Biol. 2004;164:769–779. doi: 10.1083/jcb.200307137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ongusaha PP, Kwak JC, Zwible AJ, et al. HB-EGF is a potent inducer of tumor growth and angiogenesis. Cancer Res. 2004;64:5283–5290. doi: 10.1158/0008-5472.CAN-04-0925. [DOI] [PubMed] [Google Scholar]
  • 37.McGowan PM, Ryan BM, Hill AD, et al. ADAM-17 expression in breast cancer correlates with variables of tumor progression. Clin Cancer Res. 2007;13:2335–2343. doi: 10.1158/1078-0432.CCR-06-2092. [DOI] [PubMed] [Google Scholar]
  • 38.Blobel CP. ADAMs: Key components in EGFR signaling and development. Nat Rev Mol Cell Biol. 2005;6:32–43. doi: 10.1038/nrm1548. [DOI] [PubMed] [Google Scholar]
  • 39.Kenny PA, Bissell MJ. Targeting TACE-dependent EGFR ligand shedding in breast cancer. J Clin Invest. 2007;117:337–345. doi: 10.1172/JCI29518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Houck KA, Leung DW, Rowland AM, et al. Dual regulation of vascular endothelial growth factor bioavailability by genetic and proteolytic mechanisms. J Biol Chem. 1992;267:26031–26037. [PubMed] [Google Scholar]
  • 41.Bergers G, Brekken R, McMahon G, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol. 2000;2:737–744. doi: 10.1038/35036374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Whitelock JM, Murdoch AD, Iozzo RV, et al. The degradation of human endothelial cell-derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin, and heparanases. J Biol Chem. 1996;271:10079–10086. doi: 10.1074/jbc.271.17.10079. [DOI] [PubMed] [Google Scholar]
  • 43.Birkedal-Hansen H. Proteolytic remodeling of extracellular matrix. Curr Opin Cell Biol. 1995;7:728–735. doi: 10.1016/0955-0674(95)80116-2. [DOI] [PubMed] [Google Scholar]
  • 44.Unemori EN, Ferrara N, Bauer EA, et al. Vascular endothelial growth factor induces interstitial collagenase expression in human endothelial cells. J Cell Physiol. 1992;153:557–562. doi: 10.1002/jcp.1041530317. [DOI] [PubMed] [Google Scholar]
  • 45.Lamoreaux WJ, Fitzgerald ME, Reiner A, et al. Vascular endothelial growth factor increases release of gelatinase A and decreases release of tissue inhibitor of metalloproteinases by microvascular endothelial cells in vitro. Microvasc Res. 1998;55:29–42. doi: 10.1006/mvre.1997.2056. [DOI] [PubMed] [Google Scholar]
  • 46.Fang J, Shing Y, Wiederschain D, et al. Matrix metalloproteinase-2 is required for the switch to the angiogenic phenotype in a tumor model. Proc Natl Acad Sci U S A. 2000;97:3884–3889. doi: 10.1073/pnas.97.8.3884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.O'Reilly MS, Wiederschain D, Stetler-Stevenson WG, et al. Regulation of angiostatin production by matrix metalloproteinase-2 in a model of concomitant resistance. J Biol Chem. 1999;274:29568–29571. doi: 10.1074/jbc.274.41.29568. [DOI] [PubMed] [Google Scholar]
  • 48.Dong Z, Kumar R, Yang X, et al. Macrophage-derived metalloelastase is responsible for the generation of angiostatin in Lewis lung carcinoma. Cell. 1997;88:801–810. doi: 10.1016/s0092-8674(00)81926-1. [DOI] [PubMed] [Google Scholar]
  • 49.Wen W, Moses MA, Wiederschain D, et al. The generation of endostatin is mediated by elastase. Cancer Res. 1999;59:6052–6056. [PubMed] [Google Scholar]
  • 50.Zhang B, Cao X, Liu Y, et al. Tumor-derived matrix metalloproteinase-13 (MMP-13) correlates with poor prognoses of invasive breast cancer. BMC Cancer. 2008;83:1–10. doi: 10.1186/1471-2407-8-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wu ZS, Wu Q, Yang JH, et al. Prognostic significance of MMP-9 and TIMP-1 serum and tissue expression in breast cancer. Int J Cancer. 2008;122:2050–2056. doi: 10.1002/ijc.23337. [DOI] [PubMed] [Google Scholar]
  • 52.Fernandez CA, Yan L, Louis G, et al. The matrix metalloproteinase-9/neutrophil gelatinase-associated lipocalin complex plays a role in breast tumor growth and is present in the urine of breast cancer patients. Clin Cancer Res. 2005;11:5390–5395. doi: 10.1158/1078-0432.CCR-04-2391. [DOI] [PubMed] [Google Scholar]
  • 53.La Rocca G, Pucci-Minafra I, Marrazzo A, et al. Zymographic detection and clinical correlations of MMP-2 and MMP-9 in breast cancer sera. Br J Cancer. 2004;90:1414–1421. doi: 10.1038/sj.bjc.6601725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ranuncolo SM, Armanasco E, Cresta C, et al. Plasma MMP-9 (92 kDa-MMP) activity is useful in the follow-up and in the assessment of prognosis in breast cancer patients. Int J Cancer. 2003;106:745–751. doi: 10.1002/ijc.11288. [DOI] [PubMed] [Google Scholar]
  • 55.Roy R, Wewer UM, Zurakowski D, et al. ADAM 12 cleaves extracellular matrix proteins and correlates with cancer status and stage. J Biol Chem. 2004;279:51323–51330. doi: 10.1074/jbc.M409565200. [DOI] [PubMed] [Google Scholar]
  • 56.McGowan PM, McKiernan E, Bolster F, et al. ADAM-17 predicts adverse outcome in patients with breast cancer. Ann Oncol. 2008;19:1075–1081. doi: 10.1093/annonc/mdm609. [DOI] [PubMed] [Google Scholar]
  • 57.Poola I, DeWitty RL, Marshalleck JJ, et al. Identification of MMP-1 as a putative breast cancer predictive marker by global gene expression analysis. Nat Med. 2005;11:481–483. doi: 10.1038/nm1243. [DOI] [PubMed] [Google Scholar]
  • 58.Tian M, Cui YZ, Song GH, et al. Proteomic analysis identifies MMP-9, DJ-1 and A1BG as overexpressed proteins in pancreatic juice from pancreatic ductal adenocarcinoma patients. BMC Cancer. 2008;241:1–11. doi: 10.1186/1471-2407-8-241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Yokoyama M, Ochi K, Ichimura M, et al. Matrix metalloproteinase-2 in pancreatic juice for diagnosis of pancreatic cancer. Pancreas. 2002;24:344–347. doi: 10.1097/00006676-200205000-00004. [DOI] [PubMed] [Google Scholar]
  • 60.Kuhlmann KF, van Till JW, Boermeester MA, et al. Evaluation of matrix metalloproteinase 7 in plasma and pancreatic juice as a biomarker for pancreatic cancer. Cancer Epidemiol Biomarkers Prev. 2007;16:886–891. doi: 10.1158/1055-9965.EPI-06-0779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Jones LE, Humphreys MJ, Campbell F, et al. Comprehensive analysis of matrix metalloproteinase and tissue inhibitor expression in pancreatic cancer: Increased expression of matrix metalloproteinase-7 predicts poor survival. Clin Cancer Res. 2004;10:2832–2845. doi: 10.1158/1078-0432.ccr-1157-03. [DOI] [PubMed] [Google Scholar]
  • 62.Grutzmann R, Luttges J, Sipos B, et al. ADAM9 expression in pancreatic cancer is associated with tumour type and is a prognostic factor in ductal adenocarcinoma. Br J Cancer. 2004;90:1053–1058. doi: 10.1038/sj.bjc.6601645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Jumper C, Cobos E, Lox C. Determination of the serum matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) in patients with either advanced small-cell lung cancer or non-small-cell lung cancer prior to treatment. Respir Med. 2004;98:173–177. doi: 10.1016/j.rmed.2003.08.014. [DOI] [PubMed] [Google Scholar]
  • 64.Koc M, Ediger D, Budak F, et al. Matrix metalloproteinase-9 (MMP-9) elevated in serum but not in bronchial lavage fluid in patients with lung cancer. Tumori. 2006;92:149–154. doi: 10.1177/030089160609200211. [DOI] [PubMed] [Google Scholar]
  • 65.Liu D, Nakano J, Ishikawa S, et al. Overexpression of matrix metalloproteinase-7 (MMP-7) correlates with tumor proliferation, and a poor prognosis in non-small cell lung cancer. Lung Cancer. 2007;58:384–391. doi: 10.1016/j.lungcan.2007.07.005. [DOI] [PubMed] [Google Scholar]
  • 66.Su L, Zhou W, Park S, et al. Matrix metalloproteinase-1 promoter polymorphism and lung cancer risk. Cancer Epidemiol Biomarkers Prev. 2005;14:567–570. doi: 10.1158/1055-9965.EPI-04-0482. [DOI] [PubMed] [Google Scholar]
  • 67.Sauter W, Rosenberger A, Beckmann L, et al. Matrix metalloproteinase 1 (MMP1) is associated with early-onset lung cancer. Cancer Epidemiol Biomarkers Prev. 2008;17:1127–1135. doi: 10.1158/1055-9965.EPI-07-2840. [DOI] [PubMed] [Google Scholar]
  • 68.Papathoma AS, Petraki C, Grigorakis A, et al. Prognostic significance of matrix metalloproteinases 2 and 9 in bladder cancer. Anticancer Res. 2000;20:2009–2013. [PubMed] [Google Scholar]
  • 69.Gerhards S, Jung K, Koenig F, et al. Excretion of matrix metalloproteinases 2 and 9 in urine is associated with a high stage and grade of bladder carcinoma. Urology. 2001;57:675–679. doi: 10.1016/s0090-4295(00)01087-6. [DOI] [PubMed] [Google Scholar]
  • 70.Roy R, Louis G, Loughlin KR, et al. Tumor-specific urinary matrix metalloproteinase fingerprinting: Identification of high molecular weight urinary matrix metalloproteinase species. Clin Cancer Res. 2008;14:6610–6617. doi: 10.1158/1078-0432.CCR-08-1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Moses MA, Wiederschain D, Loughlin KR, et al. Increased incidence of matrix metalloproteinases in urine of cancer patients. Cancer Res. 1998;58:1395–1399. [PubMed] [Google Scholar]
  • 72.Sier CF, Casetta G, Verheijen JH, et al. Enhanced urinary gelatinase activities (matrix metalloproteinases 2 and 9) are associated with early-stage bladder carcinoma: A comparison with clinically used tumor markers. Clin Cancer Res. 2000;6:2333–2340. [PubMed] [Google Scholar]
  • 73.Eissa S, Swellam M, el-Mosallamy H, et al. Diagnostic value of urinary molecular markers in bladder cancer. Anticancer Res. 2003;23:4347–4355. [PubMed] [Google Scholar]
  • 74.Hilska M, Roberts PJ, Collan YU, et al. 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. doi: 10.1002/ijc.22747. [DOI] [PubMed] [Google Scholar]
  • 75.Langenskiold M, Holmdahl L, Falk P, et al. Increased plasma MMP-2 protein expression in lymph node-positive patients with colorectal cancer. Int J Colorectal Dis. 2005;20:245–252. doi: 10.1007/s00384-004-0667-4. [DOI] [PubMed] [Google Scholar]
  • 76.Cho YB, Lee WY, Song SY, et al. Matrix metalloproteinase-9 activity is associated with poor prognosis in T3-T4 node-negative colorectal cancer. Hum Pathol. 2007;38:1603–1610. doi: 10.1016/j.humpath.2007.03.018. [DOI] [PubMed] [Google Scholar]
  • 77.Tutton MG, George ML, Eccles SA, et al. Use of plasma MMP-2 and MMP-9 levels as a surrogate for tumour expression in colorectal cancer patients. Int J Cancer. 2003;107:541–550. doi: 10.1002/ijc.11436. [DOI] [PubMed] [Google Scholar]
  • 78.Maurel J, Nadal C, Garcia-Albeniz X, et al. Serum matrix metalloproteinase 7 levels identifies poor prognosis advanced colorectal cancer patients. Int J Cancer. 2007;121:1066–1071. doi: 10.1002/ijc.22799. [DOI] [PubMed] [Google Scholar]
  • 79.Baker EA, Leaper DJ. The plasminogen activator and matrix metalloproteinase systems in colorectal cancer: Relationship to tumour pathology. Eur J Cancer. 2003;39:981–988. doi: 10.1016/s0959-8049(03)00065-0. [DOI] [PubMed] [Google Scholar]
  • 80.Leeman MF, McKay JA, Murray GI. Matrix metalloproteinase 13 activity is associated with poor prognosis in colorectal cancer. J Clin Pathol. 2002;55:758–762. doi: 10.1136/jcp.55.10.758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lengyel E, Schmalfeldt B, Konik E, et al. Expression of latent matrix metalloproteinase 9 (MMP-9) predicts survival in advanced ovarian cancer. Gynecol Oncol. 2001;82:291–298. doi: 10.1006/gyno.2001.6243. [DOI] [PubMed] [Google Scholar]
  • 82.Kamat AA, Fletcher M, Gruman LM, et al. The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin Cancer Res. 2006;12:1707–1714. doi: 10.1158/1078-0432.CCR-05-2338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Perigny M, Bairati I, Harvey I, et al. Role of immunohistochemical overexpression of matrix metalloproteinases MMP-2 and MMP-11 in the prognosis of death by ovarian cancer. Am J Clin Pathol. 2008;129:226–231. doi: 10.1309/49LA9XCBGWJ8F2KM. [DOI] [PubMed] [Google Scholar]
  • 84.Adley BP, Gleason KJ, Yang XJ, et al. Expression of membrane type 1 matrix metalloproteinase (MMP-14) in epithelial ovarian cancer: High level expression in clear cell carcinoma. Gynecol Oncol. 2009;112:319–324. doi: 10.1016/j.ygyno.2008.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tanaka Y, Miyamoto S, Suzuki SO, et al. Clinical significance of heparin-binding epidermal growth factor-like growth factor and a disintegrin and metalloprotease 17 expression in human ovarian cancer. Clin Cancer Res. 2005;11:4783–4792. doi: 10.1158/1078-0432.CCR-04-1426. [DOI] [PubMed] [Google Scholar]
  • 86.Morgia G, Falsaperla M, Malaponte G, et al. Matrix metalloproteinases as diagnostic (MMP-13) and prognostic (MMP-2, MMP-9) markers of prostate cancer. Urol Res. 2005;33:44–50. doi: 10.1007/s00240-004-0440-8. [DOI] [PubMed] [Google Scholar]
  • 87.Miyake H, Muramaki M, Kurahashi T, et al. Expression of potential molecular markers in prostate cancer: Correlation with clinicopathological outcomes in patients undergoing radical prostatectomy. Urol Oncol. 2008 Oct 9; doi: 10.1016/j.urolonc.2008.08.001. epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 88.Trudel D, Fradet Y, Meyer F, et al. Membrane-type-1 matrix metalloproteinase, matrix metalloproteinase 2, and tissue inhibitor of matrix proteinase 2 in prostate cancer: Identification of patients with poor prognosis by immunohistochemistry. Hum Pathol. 2008;39:731–739. doi: 10.1016/j.humpath.2007.09.021. [DOI] [PubMed] [Google Scholar]
  • 89.Fritzsche FR, Jung M, Xu C, et al. ADAM8 expression in prostate cancer is associated with parameters of unfavorable prognosis. Virchows Arch. 2006;449:628–636. doi: 10.1007/s00428-006-0315-1. [DOI] [PubMed] [Google Scholar]
  • 90.Fritzsche FR, Jung M, Tolle A, et al. ADAM9 expression is a significant and independent prognostic marker of PSA relapse in prostate cancer. Eur Urol. 2008;54:1097–1106. doi: 10.1016/j.eururo.2007.11.034. [DOI] [PubMed] [Google Scholar]
  • 91.Sawaya RE, Yamamoto M, Gokaslan ZL, et al. Expression and localization of 72 kDa type IV collagenase (MMP-2) in human malignant gliomas in vivo. Clin Exp Metastasis. 1996;14:35–42. doi: 10.1007/BF00157684. [DOI] [PubMed] [Google Scholar]
  • 92.Jaalinoja J, Herva R, Korpela M, et al. Matrix metalloproteinase 2 (MMP-2) immunoreactive protein is associated with poor grade and survival in brain neoplasms. J Neurooncol. 2000;46:81–90. doi: 10.1023/a:1006421112839. [DOI] [PubMed] [Google Scholar]
  • 93.Rao JS, Yamamoto M, Mohaman S, et al. Expression and localization of 92 kDa type IV collagenase/gelatinase B (MMP-9) in human gliomas. Clin Exp Metastasis. 1996;14:12–18. doi: 10.1007/BF00157681. [DOI] [PubMed] [Google Scholar]
  • 94.Choe G, Park JK, Jouben-Steele L, et al. Active matrix metalloproteinase 9 expression is associated with primary glioblastoma subtype. Clin Cancer Res. 2002;8:2894–2901. [PubMed] [Google Scholar]
  • 95.Friedberg MH, Glantz MJ, Klempner MS, et al. Specific matrix metalloproteinase profiles in the cerebrospinal fluid correlated with the presence of malignant astrocytomas, brain metastases, and carcinomatous meningitis. Cancer. 1998;82:923–930. doi: 10.1002/(sici)1097-0142(19980301)82:5<923::aid-cncr18>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 96.Smith ER, Zurakowski D, Saad A, et al. Urinary biomarkers predict brain tumor presence and response to therapy. Clin Cancer Res. 2008;14:2378–2386. doi: 10.1158/1078-0432.CCR-07-1253. [DOI] [PubMed] [Google Scholar]
  • 97.Pories SE, Zurakowski D, Roy R, et al. Urinary metalloproteinases: Noninvasive biomarkers for breast cancer risk assessment. Cancer Epidemiol Biomarkers Prev. 2008;17:1034–1042. doi: 10.1158/1055-9965.EPI-07-0365. [DOI] [PubMed] [Google Scholar]
  • 98.Zucker S, Hymowitz M, Conner C, et al. Measurement of matrix metalloproteinases and tissue inhibitors of metalloproteinases in blood and tissues: Clinical and experimental applications. Ann N Y Acad Sci. 1999;878:212–227. doi: 10.1111/j.1749-6632.1999.tb07687.x. [DOI] [PubMed] [Google Scholar]
  • 99.Tetu B, Brisson J, Wang CS, et al. The influence of MMP-14, TIMP-2 and MMP-2 expression on breast cancer prognosis. Breast Cancer Res. 2006;R28:1–9. doi: 10.1186/bcr1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Schutz A, Schneidenbach D, Aust G, et al. Differential expression and activity status of MMP-1, MMP-2 and MMP-9 in tumor and stromal cells of squamous cell carcinomas of the lung. Tumour Biol. 2002;23:179–184. doi: 10.1159/000064034. [DOI] [PubMed] [Google Scholar]
  • 101.Ohtsuka T, Shiomi T, Shimoda M, et al. ADAM28 is overexpressed in human non-small cell lung carcinomas and correlates with cell proliferation and lymph node metastasis. Int J Cancer. 2006;118:263–273. doi: 10.1002/ijc.21324. [DOI] [PubMed] [Google Scholar]
  • 102.Frohlich C, Albrechtsen R, Dyrskjot L, et al. Molecular profiling of ADAM12 in human bladder cancer. Clin Cancer Res. 2006;12:7359–7368. doi: 10.1158/1078-0432.CCR-06-1066. [DOI] [PubMed] [Google Scholar]
  • 103.Waas ET, Wobbes T, Lomme RM, et al. Plasma gelatinase activity does not reflect disease activity after operation for colorectal cancer. Oncology. 2005;68:256–262. doi: 10.1159/000086960. [DOI] [PubMed] [Google Scholar]
  • 104.Schmalfeldt B, Prechtel D, Harting K, et al. Increased expression of matrix metalloproteinases (MMP)-2, MMP-9, and the urokinase-type plasminogen activator is associated with progression from benign to advanced ovarian cancer. Clin Cancer Res. 2001;7:2396–2404. [PubMed] [Google Scholar]
  • 105.Omura GA, Brady MF, Homesley HD, et al. Long-term follow-up and prognostic factor analysis in advanced ovarian carcinoma: The Gynecologic Oncology Group experience. J Clin Oncol. 1991;9:1138–1150. doi: 10.1200/JCO.1991.9.7.1138. [DOI] [PubMed] [Google Scholar]
  • 106.Sillanpaa S, Anttila M, Voutilainen K, et al. Prognostic significance of matrix metalloproteinase-9 (MMP-9) in epithelial ovarian cancer. Gynecol Oncol. 2007;104:296–303. doi: 10.1016/j.ygyno.2006.09.004. [DOI] [PubMed] [Google Scholar]
  • 107.Wood M, Fudge K, Mohler JL, et al. In situ hybridization studies of metalloproteinases 2 and 9 and TIMP-1 and TIMP-2 expression in human prostate cancer. Clin Exp Metastasis. 1997;15:246–258. doi: 10.1023/a:1018421431388. [DOI] [PubMed] [Google Scholar]
  • 108.Sauer CG, Kappeler A, Spath M, et al. Expression and activity of matrix metalloproteinases-2 and -9 in serum, core needle biopsies and tissue specimens of prostate cancer patients. Virchows Arch. 2004;444:518–526. doi: 10.1007/s00428-004-1016-2. [DOI] [PubMed] [Google Scholar]
  • 109.Riddick AC, Shukla CJ, Pennington CJ, et al. Identification of degradome components associated with prostate cancer progression by expression analysis of human prostatic tissues. Br J Cancer. 2005;92:2171–2180. doi: 10.1038/sj.bjc.6602630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kuniyasu H, Ukai R, Johnston D, et al. The relative mRNA expression levels of matrix metalloproteinase to E-cadherin in prostate biopsy specimens distinguishes organ-confined from advanced prostate cancer at radical prostatectomy. Clin Cancer Res. 2003;9:2185–2194. [PubMed] [Google Scholar]
  • 111.Smith ER, Manfredi M, Scott RM, et al. A recurrent craniopharyngioma illustrates the potential usefulness of urinary matrix metalloproteinases as noninvasive biomarkers: Case report. Neurosurgery. 2007;60:E1148–E1149. doi: 10.1227/01.NEU.0000255464.37634.3C. discussion E1149, 2007. [DOI] [PubMed] [Google Scholar]
  • 112.Jung K, Laube C, Lein M, et al. Kind of sample as preanalytical determinant of matrix metalloproteinase 2 and 9 and tissue inhibitor of metalloproteinase 2 in blood. Clin Chem. 1998;44:1060–1062. [PubMed] [Google Scholar]
  • 113.Makowski GS, Ramsby ML. Use of citrate to minimize neutrophil matrix metalloproteinase-9 in human plasma. Anal Biochem. 2003;322:283–286. doi: 10.1016/j.ab.2003.07.030. [DOI] [PubMed] [Google Scholar]
  • 114.Rouy D, Ernens I, Jeanty C, et al. Plasma storage at −80 degrees C does not protect matrix metalloproteinase-9 from degradation. Anal Biochem. 2005;338:294–298. doi: 10.1016/j.ab.2004.10.052. [DOI] [PubMed] [Google Scholar]
  • 115.Zucker S, Cao J. Measurement of matrix metalloproteinases in serum of patients with melanoma: Snarled in technical pitfalls. Clin Cancer Res. 2005;11:5069–5070. doi: 10.1158/1078-0432.CCR-05-0774. [DOI] [PubMed] [Google Scholar]
  • 116.Robins HI, O'Neill A, Gilbert M, et al. Effect of dalteparin and radiation on survival and thromboembolic events in glioblastoma multiforme: A phase II ECOG trial. Cancer Chemother Pharmacol. 2008;62:227–233. doi: 10.1007/s00280-007-0596-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sideras K, Schaefer PL, Okuno SH, et al. Low-molecularity-weight heparin in patients with advanced cancer: A phase III clinical trial. Mayo Clin Proc. 2006;81:758–767. doi: 10.4065/81.6.758. [DOI] [PubMed] [Google Scholar]
  • 118.Loprinzi CL, Levitt R, Barton DL, et al. Evaluation of shark cartilage in patients with advanced cancer: A North Central Cancer Treatment Group trial. Cancer. 2005;104:176–182. doi: 10.1002/cncr.21107. [DOI] [PubMed] [Google Scholar]
  • 119.Lu C, Lee JJ, Komaki R, et al. A phase III study of AE-941 with induction chemotherapy (IC) and concomitant chemoradiotherapy (CRT) for stage III non-small cell lung cancer (NSCLC) (NCI T99-0046, RTOG 02-70, MDA 99-303) J Clin Oncol. 25(suppl 18S):391S. abstr 7527. [Google Scholar]
  • 120.Batist G, Patenaude F, Champagne P, et al. Neovastat (AE-941) in refractory renal cell carcinoma patients: Report of a phase II trial with two dose levels. Ann Oncol. 2002;13:1259–1263. doi: 10.1093/annonc/mdf195. [DOI] [PubMed] [Google Scholar]
  • 121.Betz M, Huxley P, Davies SJ, et al. 1.8-A crystal structure of the catalytic domain of human neutrophil collagenase (matrix metalloproteinase-8) complexed with a peptidomimetic hydroxamate primed-side inhibitor with a distinct selectivity profile. Eur J Biochem. 1997;247:356–363. doi: 10.1111/j.1432-1033.1997.00356.x. [DOI] [PubMed] [Google Scholar]
  • 122.Saghatelian A, Jessani N, Joseph A, et al. Activity-based probes for the proteomic profiling of metalloproteases. Proc Natl Acad Sci U S A. 2004;101:10000–10005. doi: 10.1073/pnas.0402784101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hidalgo M, Eckhardt SG. Development of matrix metalloproteinase inhibitors in cancer therapy. J Natl Cancer Inst. 2001;93:178–193. doi: 10.1093/jnci/93.3.178. [DOI] [PubMed] [Google Scholar]
  • 124.Miller KD, Saphner TJ, Waterhouse DM, et al. A randomized phase II feasibility trial of BMS-275291 in patients with early stage breast cancer. Clin Cancer Res. 2004;10:1971–1975. doi: 10.1158/1078-0432.ccr-03-0968. [DOI] [PubMed] [Google Scholar]
  • 125.Dublanchet AC, Ducrot P, Andrianjara C, et al. Structure-based design and synthesis of novel non-zinc chelating MMP-12 inhibitors. Bioorg Med Chem Lett. 2005;15:3787–3790. doi: 10.1016/j.bmcl.2005.05.079. [DOI] [PubMed] [Google Scholar]
  • 126.Chen JM, Nelson FC, Levin JI, et al. Structure-based design of a novel, potent, and selective inhibitor for MMP-13 utilizing NMR spectroscopy and computer-aided molecular design. J Am Chem Soc. 2000;122:9648–9654. [Google Scholar]
  • 127.Golub LM, Ramamurthy NS, McNamara TF, et al. Tetracyclines inhibit connective tissue breakdown: New therapeutic implications for an old family of drugs. Crit Rev Oral Biol Med. 1991;2:297–321. doi: 10.1177/10454411910020030201. [DOI] [PubMed] [Google Scholar]
  • 128.Acharya MR, Venitz J, Figg WD, et al. Chemically modified tetracyclines as inhibitors of matrix metalloproteinases. Drug Resist Updat. 2004;7:195–208. doi: 10.1016/j.drup.2004.04.002. [DOI] [PubMed] [Google Scholar]
  • 129.Sapadin AN, Fleischmajer R. Tetracyclines: Nonantibiotic properties and their clinical implications. J Am Acad Dermatol. 2006;54:258–265. doi: 10.1016/j.jaad.2005.10.004. [DOI] [PubMed] [Google Scholar]
  • 130.Brown S, Bernardo MM, Li Z, et al. Potent and selective mechanism-based inhibition of gelatinases. J Am Chem Soc. 2000;122:6799–6800. [Google Scholar]
  • 131.Kleifeld O, Kotra LP, Gervasi DC, et al. X-ray absorption studies of human matrix metalloproteinase-2 (MMP-2) bound to a highly selective mechanism-based inhibitor: Comparison with the latent and active forms of the enzyme. J Biol Chem. 2001;276:17125–17131. doi: 10.1074/jbc.M011604200. [DOI] [PubMed] [Google Scholar]
  • 132.Kruger A, Arlt MJ, Gerg M, et al. Antimetastatic activity of a novel mechanism-based gelatinase inhibitor. Cancer Res. 2005;65:3523–3526. doi: 10.1158/0008-5472.CAN-04-3570. [DOI] [PubMed] [Google Scholar]
  • 133.Ikejiri M, Bernardo MM, Meroueh SO, et al. Design, synthesis, and evaluation of a mechanism-based inhibitor for gelatinase A. J Org Chem. 2005;70:5709–5712. doi: 10.1021/jo050339+. [DOI] [PubMed] [Google Scholar]
  • 134.Moss ML, Bartsch JW. Therapeutic benefits from targeting of ADAM family members. Biochemistry. 2004;43:7227–7235. doi: 10.1021/bi049677f. [DOI] [PubMed] [Google Scholar]
  • 135.Liu X, Fridman JS, Wang Q, et al. Selective inhibition of ADAM metalloproteases blocks HER-2 extracellular domain (ECD) cleavage and potentiates the anti-tumor effects of trastuzumab. Cancer Biol Ther. 2006;5:648–656. doi: 10.4161/cbt.5.6.2707. [DOI] [PubMed] [Google Scholar]
  • 136.Fridman JS, Caulder E, Hansbury M, et al. Selective inhibition of ADAM metalloproteases as a novel approach for modulating ErbB pathways in cancer. Clin Cancer Res. 2007;13:1892–1902. doi: 10.1158/1078-0432.CCR-06-2116. [DOI] [PubMed] [Google Scholar]
  • 137.Fernandez CA, Butterfield C, Jackson G, et al. Structural and functional uncoupling of the enzymatic and angiogenic inhibitory activities of tissue inhibitor of metalloproteinase-2 (TIMP-2): Loop 6 is a novel angiogenesis inhibitor. J Biol Chem. 2003;278:40989–40995. doi: 10.1074/jbc.M306176200. [DOI] [PubMed] [Google Scholar]
  • 138.Seo DW, Li H, Guedez L, et al. TIMP-2 mediated inhibition of angiogenesis: An MMP-independent mechanism. Cell. 2003;114:171–180. doi: 10.1016/s0092-8674(03)00551-8. [DOI] [PubMed] [Google Scholar]
  • 139.Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: Trials and tribulations. Science. 2002;295:2387–2392. doi: 10.1126/science.1067100. [DOI] [PubMed] [Google Scholar]
  • 140.Fingleton B. MMPs as therapeutic targets–still a viable option? Semin Cell Dev Biol. 2008;19:61–68. doi: 10.1016/j.semcdb.2007.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Bergers G, Javaherian K, Lo KM, et al. Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science. 1999;284:808–812. doi: 10.1126/science.284.5415.808. [DOI] [PubMed] [Google Scholar]
  • 142.Witty JP, Lempka T, Coffey RJ, Jr, et al. Decreased tumor formation in 7,12-dimethylbenzanthracene-treated stromelysin-1 transgenic mice is associated with alterations in mammary epithelial cell apoptosis. Cancer Res. 1995;55:1401–1406. [PubMed] [Google Scholar]
  • 143.Balbin M, Fueyo A, Tester AM, et al. Loss of collagenase-2 confers increased skin tumor susceptibility to male mice. Nat Genet. 2003;35:252–257. doi: 10.1038/ng1249. [DOI] [PubMed] [Google Scholar]
  • 144.Acuff HB, Sinnamon M, Fingleton B, et al. Analysis of host- and tumor-derived proteinases using a custom dual species microarray reveals a protective role for stromal matrix metalloproteinase-12 in non-small cell lung cancer. Cancer Res. 2006;66:7968–7975. doi: 10.1158/0008-5472.CAN-05-4279. [DOI] [PubMed] [Google Scholar]
  • 145.Yang W, Arii S, Gorrin-Rivas MJ, et al. Human macrophage metalloelastase gene expression in colorectal carcinoma and its clinicopathologic significance. Cancer. 2001;91:1277–1283. [PubMed] [Google Scholar]
  • 146.Coussens LM, Tinkle CL, Hanahan D, et al. MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell. 2000;103:481–490. doi: 10.1016/s0092-8674(00)00139-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Overall CM, Kleifeld O. Tumour microenvironment - opinion: Validating matrix metalloproteinases as drug targets and anti-targets for cancer therapy. Nat Rev Cancer. 2006;6:227–239. doi: 10.1038/nrc1821. [DOI] [PubMed] [Google Scholar]
  • 148.Cornelius LA, Nehring LC, Harding E, et al. Matrix metalloproteinases generate angiostatin: Effects on neovascularization. J Immunol. 1998;161:6845–6852. [PubMed] [Google Scholar]
  • 149.Ferreras M, Felbor U, Lenhard T, et al. Generation and degradation of human endostatin proteins by various proteinases. FEBS Lett. 2000;486:247–251. doi: 10.1016/s0014-5793(00)02249-3. [DOI] [PubMed] [Google Scholar]
  • 150.McCawley LJ, Crawford HC, King LE, Jr, et al. A protective role for matrix metalloproteinase-3 in squamous cell carcinoma. Cancer Res. 2004;64:6965–6972. doi: 10.1158/0008-5472.CAN-04-0910. [DOI] [PubMed] [Google Scholar]
  • 151.Parks WC, Wilson CL, Lopez-Boado YS. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol. 2004;4:617–629. doi: 10.1038/nri1418. [DOI] [PubMed] [Google Scholar]
  • 152.Bremer C, Tung CH, Weissleder R. In vivo molecular target assessment of matrix metalloproteinase inhibition. Nat Med. 2001;7:743–748. doi: 10.1038/89126. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Clinical Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES