Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2008 Dec 1.
Published in final edited form as: Semin Oncol. 2007 Dec;34(6):555–565. doi: 10.1053/j.seminoncol.2007.09.009

Angiogenesis in Melanoma

Ganapati H Mahabeleshwar 1, Tatiana V Byzova 1,
PMCID: PMC2365306  NIHMSID: NIHMS36506  PMID: 18083379

Summary

The process of angiogenesis is crucial for progression and metastasis of the majority of solid tumors including melanomas. The purpose of this review is to summarize existing knowledge of the mechanisms of angiogenesis in melanoma as well as current anti-angiogenic therapeutic strategies and their targets. Here, we have focused primarily on the role of key growth factors which are secreted by melanoma cells and known to trigger angiogenic responses, and their receptors as expressed on both endothelial and melanoma cells. Many of these growth factors function in synergy with receptors for extracellular matrix, integrins and matrix metalloproteinases. All these systems of molecules are activated during major stages of angiogenesis such as endothelial migration, proliferation and reorganization of surrounding extracellular matrix. The blockade of these molecules and their downstream pathways leads to inhibition of melanoma vascularization. Thus, these classes of molecules are essential for melanoma angiogenesis and, therefore, might serve as promising targets for therapeutic intervention. Many recently developed compounds targeting key pathways in angiogenesis are in their final stages of clinical trials.

Keywords: Melanoma, Angiogenesis, Extracellular matrix, Growth factors, Integrins, Matrix metalloproteinase

Introduction

Angiogenesis, the process of formation of neovasculature from pre-existing blood vessels, is widely considered as an essential process to ensure the supply of nutrients and oxygen to rapidly growing tumors as well as to provide a route for tumor cell metastasis1. Angiogenesis is a distinct feature of several human cutaneous melanomas and was first described by Warren and Shubik following transplantation of human melanoma tumor tissue into a hamster cheek pouch2. These studies were later confirmed by Hubler and Wolf 3. Rapid angiogenesis of cutaneous melanomas dramatically enhances the risk of lethality and contributes to the progression of the most common type of cancer in young adults.

Like any other cutaneous neoplasia, melanomas follow discrete sequential transformation processes in which the nevus attains dysplastic radial growth phase (Figure.1). This is followed by a vertical growth phase46, The vertical growth phase requires high angiogenic activity, which, in turn, contributes to melanoma cell metastasis. However, the most critical step in metastasis is the spreading of melanomas to lymphatic vessels surrounding the tumor tissue. Following lymph node invasion, melanoma cells metastasize to the lung, liver, cerebrum, and other sites. Exposure to ultraviolet radiation is known to cause genetic changes in skin, which modulate the cutaneous immune response and increase production of several growth factors 79. This causes uncontrolled proliferation of melanocytes, which, in turn, dramatically increases the consumption of oxygen and nutrients eventually leading to cell starvation and hypoxia10,11. To fulfill this increasing demand additional vasculature needs to be developed. Thus, to increase the blood supply, the tissue begins to produce a spectrum of growth factors which trigger the process of angiogenesis.

Figure 1. Malignant progression of melanoma.

Figure 1

Aberrant proliferation of normal melanocytes leads to formation of benign naevus. The rapidly proliferating transformed melanocytes form dysplastic naevi, which are asymmetric with irregular borders. This is followed by a rapid radial growth phase, which is characterized by intraepidermal growth. The last step in benign melanoma growth is the vertical growth phase. It is also called the dermal invasion phase. Malignant melanomas develop vasculature very rapidly providing a route for further metastasis.

Studies using human melanoma xenograft models in nude mice indicated that melanoma tumor cells serve as a source of several growth factors including but not limited to vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), two powerful stimulators of rapid angiogenesis12. Despite recent advances in our understanding of the molecular mechanisms of tumor growth/metastasis and the availability of modern technology to detect and treat various human cancers, our knowledge about the mechanisms underlying cutaneous tumor angiogenesis and metastasis remains rudimentary.

In this review we have focused on the role of cytokines and growth factors secreted by melanoma cells in tumor angiogenesis. These growth factors are involved in both autocrine and paracrine regulation of melanoma progression. Importantly, growth factors and their receptors function in intimate relationship with extracellular matrix proteins, their receptors, integrins, and matrix metalloproteinases. These classes of molecules are essential for melanoma angiogenesis and, therefore, serve as promising targets for therapeutic intervention. Most of the relatively recently developed compounds are at the final stages of clinical trials. Thus, this review is intended to summarize the basic biological knowledge regarding melanoma tumor angiogenesis as well as current clinical approaches to treat late stages of malignant melanomas.

Growth factor dependent melanoma tumor growth and angiogenesis

Angiogenesis in melanoma is stimulated by a variety of growth factors such as VEGF, basic FGF, acidic FGF, platelet derived growth factor (PDGF), and transforming growth factors α and β (TGF-α and β) are among the others (Figure 2). Angiogenesis serves as a turning point in melanoma tumor growth and metastasis.

Figure 2. Cell surface receptor mimicry between malignant melanoma cells and endothelial cells.

Figure 2

Malignant melanoma cells express VEGFR-2, VEGFR-1 and co-receptor neuropilin-1/2 which are commonly not expressed on most of the cancer cells. Therefore, VEGF is able to induce similar intracellular signaling responses in both endothelial and melanoma cells. Placental growth factor can independently bind to VEGFR-1 and neuropilin-1/2 to induce intracellular signaling. PIGF also form heterodimeric complexes with VEGF and interact with VEGFR-2. Other receptors expressed on both melanoma and endothelial cells include uPA receptor, CXCR-1/2 and FGFR-1. Therefore, there is a spectrum of growth factors/chemokines which are able to induce similar signaling responses in both cell types.

VEGF and FGF

Transformed melanocytes are known to produce high amounts of bFGF as well as VEGF during conversion of the dysplastic naevus to the vertical growth phase. This extensive growth factor production continues until new blood vessel formation is complete. Recent reports indicate that basal expression of VEGF by transformed melanocytes plays a more important role in promoting angiogenesis in melanomas than hypoxia-induced VEGF up-regulation1315. At the same time, bFGF regulates endothelial cell proliferation and angiogenesis by both autocrine and paracrine mechanisms. Since bFGF is devoid of the classic signal peptide for secretion, tumor cells release this factor by exocytosis from endoplasmic reticulum. Significant amounts of bFGF were found to be associated with extracellular matrix as well as with basement membrane of the newly formed blood vessels in human melanomas16. Digestion of extracellular matrix by matrix metalloproteinases of melanoma or endothelial origin promotes release of matrix bound bFGF, which, in turn, stimulates endothelial cell proliferation and vascular tube formation in melanomas17,18. Therefore drugs targeting these molecules are currently under various stages of clinical trial (Table 1).

Table 1.

Major growth factors expressed in malignant melanoma and clinical significance

Molecule Functions Clinical Trial Reference
VEGF/VEGFR Enhance EC proliferation, migration and angiogenesis Phase 2/3 [14], [15]
FGF/FGFR Enhance expression of VEGF and uPA and prevent EC apoptosis Phase 1 [17], [18]
PIGF Recruit bone marrow-derived hematopoietic precursors site of angiogenesis None [19], [20]
IL-8 Induce tumor neovascularization and tumor cell proliferation Phase 2 [24], [25]
uPA Increase tumor vasculature and migration and metastasis of tumor cells Phase 1 [30], [31]

PIGF

Another important stimulator of melanoma angiogenesis is placental growth factor (PIGF). PIGF exists in several isoforms that are generated by alternative splicing of the same gene product and it exhibits distinct heparin-binding properties. PIGF-1 and -2 are expressed by melanoma cells and are known to bind to neuropilin-1 and -2 receptors expressed on endothelial cells19. In addition, PIGF acts through binding to VEGF receptor-1. Also, PIGF forms heterodimers with VEGF and, as a result, is able to bind to VEGF receptor-2 on endothelial cells (Figure 2). PIGF enhances melanoma tumor angiogenesis not only by acting on pre-existing endothelial cells, but also by mobilization and recruitment of VEGFR-1 positive hematopoietic precursors from bone marrow20. Further, PIGF is known to contribute to angiogenesis by acting on VEGFR-1-expressing smooth muscle cells/pericytes, thereby enhancing blood vessel maturation. PIGF secreted by both primary and metastatic melanomas induces tumor cell proliferation, indicating PIGF might contribute to melanoma tumor growth through an autocrine mechanism. Several studies also indicated that PIGF mediated neovascularization resulted in rapid vessel branching and formation of large and stable blood vessels in various in vivo models21.

IL-8 and TGF-1

Several growth factors and their receptors are implicated in the pathogenesis of a variety of human melanomas since their upregulation is associated with melanoma progression. Among these is interleukin-8 (IL-8). Its expression was found to be very minimal in normal epidermis and benign melanocytic lesions. However, it is dramatically increased in a majority of cutaneous melanomas examined. In a clinical study analysis IL-8 serum levels in melanoma patients are significantly elevated compared to healthy individuals22. Further, serum IL-8 levels correlate with advanced disease stage as well as with overall survival of melanoma patients. A recent study by Liu et al23 indicated that transforming growth factor-1 (TGF-1) is able to enhance expression of IL-8 in human melanoma cells and promote angiogenesis in several mouse xenograft models. Several studies conclusively demonstrated that while tumor cell-derived IL-8 directly supports the growth of melanomas, IL-8 of endothelial origin further promotes melanoma cell migration. Additionally, tumor-derived IL-8 is able to induce endothelial cell migration (Figure 2).

IL-8 is also known to modulate vascular permeability. It functions through activation of G-protein coupled receptors on endothelial cells, leading to enhanced actin stress fiber formation, which, in turn, results in cell retraction and gap formation between endothelial cells Experimental evidence from several laboratories indicated that over-expression of IL-8 in poorly vascularized and nonmetastatic melanoma cells resulted in enhanced angiogenesis, rapid tumor growth and increased metastatic potential of melanomas24,25.

Plasminogen system

Besides a number of growth factors discussed above, there are other important players in melanoma which might eventually serve as therapeutic targets. Urokinase plasminogen activator and its receptor (uPA/uPAR) have been demonstrated to play a crucial role in several stages of melanoma tumor progression including melanoma cell migration, invasion and metastasis26. At the same time, uPA secreted from melanoma tumor cells is able to regulate endothelial cell functions including migration and the organization of endothelial cells into tube-like structures. Analysis of biopsy specimens of skin lesions indicated that uPA expression highly correlated with disease progression in a majority of patients. Delbaldo et al27 demonstrated that uPA and plasminogen activator inhibitor type 1 mRNAs accumulate in atypical naevocytes and in melanoma cells, but not in benign naevocytes. Further, these observations suggest that up-regulation of the uPA gene is an early event of melanocyte transformation and that unbalanced enzyme activity is associated with the malignant phenotype. Studies from Hearing et al28 indicated that mouse melanoma cells express the single chain form of uPA on their cell surface and that these cells are capable of plasminogen-dependent extracellular matrix degradation. Further, melanoma cells that had been treated with anti-uPA antibody showed significantly decreased metastatic activity compared to controls. Studies from Min et al29 demonstrated that molecules which prevent uPA binding to its receptor are able to inhibit bFGF-induced neovascularization in vivo and mouse melanoma tumor growth in syngeneic mice. Additional studies also demonstrated inhibition of melanoma tumor cell metastasis by uPA inhibitors in similar models. Taken together, these studies suggest that uPA/uPAR system is required for melanoma tumor angiogenesis and growth, as well as metastasis (Figure 2). Therefore, antagonists of the uPA/uPAR system might be utilized as potential inhibitors of tumor progression through several mechanisms30,31.

Integrin signaling in melanoma tumor progression and angiogenesis

Integrins are heterodimeric transmembrane glycoproteins consisting of α and β subunits. Their large ectodomain is required for ligand binding, while a short cytoplasmic domain transfers signals from the extracellular environment to the cytoplasm via a short transmembrane domain (Figure 3). Integrins mediate cellular processes such as migration, invasion, proliferation and metastasis in a variety of human cancers including melanomas32. Even though integrins are classically described as cell adhesion molecules, recent advances in integrin biology indicate that they play a very significant role in signal transduction, gene expression, cellular proliferation and regulation of apoptosis or anoikis of normal as well as transformed cells (Figure 3). Beyond this, integrins also play very crucial roles in embryogenesis, inflammation, immunity, tissue differentiation, regulation of cell growth, hemostasis and angiogenesis33.

Figure 3. Regulation of integrin signaling by ECM proteins and growth factor receptors.

Figure 3

Integrins and their ligand expression are up-regulated in most of the human melanomas. Ligand binding to the extracellular domain of specific integrin leads to initiation of signaling events required for a broad spectrum of processes such as cellular proliferation, survival, and actin cytoskeleton regulation. Integrin interactions with its ligands are the prerequisites for cellular adhesion, invasion and migration. Growth factors, via receptors expressed on endothelial cells or melanoma cells, are able to regulate integrin function activity, i.e. its affinity and avidity of interactions with ECM proteins. The process of functional crosstalk between growth factor receptors and integrins is regulated by several intracellular signaling molecules which are able to shuffle between growth factor receptors and integrin cytoplasmic domains.

Integrins αvβ3 and αvβ5 are known to upregulate on endothelial cells during tumor neovascularization34. Several αvβ3 integrin ligands including vitronectin, osteopontin and bone sialoprotein are known to modulate both VEGF as well as FGF-2-induced tumor angiogenesis in mouse models35. Studies from several groups indicate that αvβ3 integrin plays a very crucial role in the progression of cutaneous melanomas from the benign radial growth phase to the metastatic vertical growth phase36. Studies utilizing αvβ3 integrin inhibitory peptides, antibodies or small molecular compounds indicate that αvβ3 integrin indeed is required for tumor endothelial cell survival, therefore its inhibition leads to the regression of tumor vasculature37. These studies further indicate that the expression of αv, α2, α3 and α4 integrin subunits are elevated in malignant melanoma tumor cells compared to normal melanocytes. Similarly, laminin receptor α6β4 integrin expression is significantly reduced in melanoma cells compared to benign cells38. In contrast, clinical samples of human malignant tissue show a significant increase in α6β4 expression39. Therefore, the role of α6β4 integrin in melanoma tumor growth and metastasis is controversial. Among the beta subunits, expression of β1 and β3 are more frequently elevated in several tumor types, including melanomas (Table 2).

Table 2.

Major integrin receptors expressed on endothelial/malignant melanoma cells and clinical significance

Integrin Receptor Functions Expression Clinical Trial Reference
αvβ3 Enhance tumor vasculature and tumor growth EC, MM Phase 2 [36]
αvβ5 Similar to αvβ3 integrin induce tumor growth and angiogenesis EC Phase 2 [34]
α5β1 Essential for metastasis of melanoma cells EC, MM Phase 2 [41]
α6β4 Required for vascular maturation, associated with aggressive behavior of tumor cells EC, MM None [38]
α2β1 Essential for endothelial and tumor cell interaction with basal lamina EC, MM Phase 1 [45]
α3β1 Required for malignant progression of melanoma cells MM None [46]
α4β1 Required for melanoma cell metastasis EC, MM Phase 1/2 [47]

Other major functions assigned to integrins expressed by melanoma cells are to support ECM anchorage-dependent tumor cell proliferation and anchorage-independent invasion. Integrins are also crucial in tumor angiogenesis and tumor cell metastasis40. Expression analysis at different stages of melanoma progression have indicated that the levels of β3 and β1 integrins promote melanoma transition from the radial growth phase to the vertical growth phase41. This phenomenon is further connected with rapid tumor neovascularization as well as tumor cell metastasis. Another interesting phenomenon observed in melanoma patients is the enhanced expression of integrin ligands. One such αvβ3 integrin ligand is osteopontin42. Constitutive expression of osteopontin in dermal cells leads to sustained anti-apoptotic signaling in melanocytes and a high rate of proliferation. A recent study utilizing numerous clinical samples of melanomas in various clinical grades indicated that the expression of osteopontin is positively correlated with an invasive phenotype43. Further clinical studies have indicated that serum osteopontin levels are significantly higher in patients with benign or metastatic tumors compared to healthy individuals44. Therefore, serum osteopontin levels could potentially be a prognostic marker for various human cancers.

Studies from various laboratories indicate that expression levels of α2β1 and α5β1 are higher during the vertical growth phase of melanoma compared to the radial growth phase or malignant transformation4547. Melanoma cells during the vertical growth phase bind to collagen type-1 through α2β1 and α5β1 integrins (Table 2). This stimulates the expression of MMP-1 and -2, which are crucial for collagen fibril degradation in the dermis which in turn facilitates vertical spreading of melanomas. The accumulated, denatured collagen acts as the αvβ3 integrin ligand as the denaturation process exposes RGD sequences48. The binding of denatured collagen to αvβ3 integrin further stimulates MMP-2 expression in these cells, increasing their invasive potential. One of the major photolytic degradation products due to over-expression of MMPs in dermis is fibronectin. The fibronectin receptor α5β1 is expressed abundantly in most melanomas studied. Over- expression of α5β1 in mouse melanoma cells leads to enhanced expression of MMP-2 and MMP-749. To summarize, activation of integrins leads to enhanced expression of MMPs, which in turn degrade ECM components thereby generating a series of integrin ligands. These integrin ligands generated by photolytic degradation bind to the invasive melanomas and activate the signaling cascades required for malignant transformation.

As integrins are known to positively regulate angiogenesis, tumor growth and metastasis, several inhibitors of integrins are currently under clinical trials50. Most clinical trials are focused on inhibitors of the αvβ3 integrin complex or αv integrin alone37. These include cilengitide, ATN-161, CNTO-95 and vitaxin (the last two are humanized monoclonal antibodies). These compounds specifically mask ligand binding sites and promote the internalization of targeted integrins. Peptide integrin inhibitors currently under clinical trials include cilengitide and ATN-161. Cilengitide is a cyclic RGD peptide that specifically inhibits αvβ3 and αvβ5 integrin function. In preclinical studies, cilengitide substantially reduced tumor growth in a mouse melanoma xenograft model. Even though cilengitide has been in clinical trials for some time, the outcome of this trial has yet be published34. Similarly, the peptide integrin inhibitor ATN-161 is still under phase 1 clinical trials and data indicates that it exhibits anti-angiogenic and anti-metastatic activities51. Results of phase 1 clinical trials of vitaxin indicated that it stabilized disease and reduced the risk of metastases. However, results of phase 2 clinical trials indicated a very modest response and were not very encouraging. Therefore, in recent phase 2 clinical trials, vitaxin was administrated in combination with a standard chemotherapeutic compound (dacarbazine). Beyond this, several integrin inhibitors including small molecule compounds are currently in the preclinical phase of development. E7820, an aromatic sulfonamide derivative known to inhibit α2 integrin, entered its phase 1 clinical trial in early 200450. Similarly, volociximab, a humanized monoclonal antibody specifically targeting α5β1 integrin is also currently in phase 2 clinical trial. Phase 1 clinical studies aimed to determine the optimal concentration did not find any dose limiting toxicity of this antibody. Overall, integrin inhibitor clinical trials are encouraging and several small molecule compounds have successfully completed preclinical studies.

Role of matrix metalloproteinases in melanoma angiogenesis

Matrix metalloproteinases (MMPs) are the major class of proteases that play crucial roles in tissue remodeling during embryonic development, bone resorption, wound healing and angiogenesis. Their catalytic activity is regulated partially by tissue inhibitors of matrix metalloproteinases and it has been demonstrated by several research groups that MMPs produced by actively proliferating tumor cells facilitate angiogenesis, tumor growth and metastasis52. As MMPs are actively involved in efficient matrix degradation, MMP expression and catalytic activity are tightly regulated, at the stages of transcription, post-translational extracellular activation, and by suppression by its inhibitors53. Several studies indicated that the basal level of MMP production in benign or normal melanocytes is typically low and expression of MMPs is highly correlated with disease progression. MMP activation is accomplished by removal of the N-terminal propeptide domain through exogenous or autocatalytic cleavage54. Previous studies demonstrated that serine proteases such as plasmin activate most of the MMPs through this mechanism. MMP-2, which is abundantly expressed in early stages of malignant transformation, is known to achieve activation in a membrane-associated manner in endothelial cells and melanoma tumor cells. Further, cell surface associated membrane-type matrix metalloproteinase (MT1-MMP) is also known to activate MMP-2 through this mechanism55. To date the most extensively studied MMPs in melanomas are MMP-2 and MMP-9. It has been demonstrated by several groups that the expression and activation of these enzymes has been correlated to the invasive and metastatic phenotypes of melanomas56. Previous reports indicated that MMP-2 and MMP-9 are constitutively expressed in malignant melanomas and their expression is highly associated with melanoma atypia and dedifferentiation in melanocytic lesions57.

Currently, cell surface associations of secreted MMPs through post translational modification have developed wide interest in the scientific community. It has been previously demonstrated that MMP-2/TIMP-2 associated with the cell surface in melanomas exhibits enhanced catalytic activity against its substrates compared to MMPs in secreted phase. Malignant melanoma cells are known to express a number of MMPs, including MMP-1, -2, -9, -13, and -14, as well as inhibitors of MMPs such as TIMP-1, -2 and -356. A recently published study from Kerkela et al clearly demonstrates a specific distribution of MMPs within cutaneous squamous cell carcinomas57. Another recent clinical study also indicated that increased MMP-2 expression in melanomas was highly correlated with metastasis. Further, increases in expression of MMPs were shown to highly correlate with low survival rates in patients with malignant melanoma tumors58. It is also very important to note that not only expression of MMPs, but also their functional activity, is required for malignant tumor progression. Genetic overexpression of MT1-MMP in melanoma cells induced activation of MMP-2 and this activation is crucial for extracellular matrix degradation when localized on the leading edge of invasive carcinomas. In a clinical study of human melanoma lesions consisting of different stages of tumor progression it was found that MMP-2 and MT1-MMP positive tumor cells were often restricted to the interface between the tumor stroma and the invasive part of the tumor57. Surprisingly, expression of MMPs is not restricted to tumor cells but is also found abundantly in stromal cells. This clearly indicates a major contribution of host-derived proteases to melanoma tumor progression.

One of the major MMPs found to be expressed in human melanoma is MMP-159. A series of studies has also indicated that MMP-1 expression is highly associated with malignant melanoma progression. In vitro studies indicated that degradation of collagen types I and IV and tumor cell invasion through Matrigel required MMP-1 expression (Table 3). Other than MMP-1 and -2, the major MMP expressed in melanoma tumor cells is MMP-9 which is also referred to as gelatinase B60. MMP-9 expression in melanoma tumor cells was found exclusively during the horizontal growth phase but not during the vertical growth phase. This clearly suggests that expression of MMP-9 is an early event in melanoma progression. Studies using a mouse model indicated that MMP-9 expression was only detected in advanced stages of disease, not in early melanocyte lesions61. Further, melanomas expressing constitutively high levels of MMP-9 exhibited increased lung colonization in experimental lung metastasis models. These advancements in understanding of MMP-9 biology indicate that MMPs expressed either by neoplastic or stromal cells are important in the metastasis of melanomas62.

Table 3.

Major MMPs expressed in malignant melanoma and clinical significance

MMP Functions Clinical Trial Reference
MMP-1 Collagen type I and IV degradation and melanoma cell invasion Phase 2/3 [59]
MMP-2 Melanoma cell invasion and angiogenesis Phase 2/3 [63]
MMP-3 Promotes tumor growth and angiogenesis Phase 2/3 [64]
MMP-7 Growth, invasion, metastasis of tumor cells and angiogenesis Phase 1/2 [65]
MMP-9 Tumor cell invasion, metastasis and angiogenesis Phase 1 to 3 [62]
MMP-10 Malignant melanoma tumor cell invasion and metastasis None [66]
MMP-13 Expression correlates with tumor progression and aggressive tumor behavior None [67]
MMP-14 Melanoma tumor cell invasion and angiogenesis None [68]

Several studies using either model cell lines or animals have demonstrated that the balance between MMPs and their inhibitors finally determines melanoma tumor progression6368. To date, tissue inhibitors of matrix metalloproteinases (TIMPs) are extensively studied as they are natural inhibitors of MMPs and hence could be potential therapeutic targets. Numerous conclusive studies demonstrate that overexpression of (TIMP) -1,-2 and -3 significantly reduces melanoma tumor cell invasion, migration, tumor growth and metastasis69. Further, several studies have indicated that TIMPs significantly reduce tumor neovascularization in the several tumor models studied. Even though TIMPs are known to inhibit tumor cell metastasis in several experimental animal models, in human melanoma cells TIMP expression significantly enhances tumor cell proliferation70. Therefore, the role of TIMPs in melanoma tumor growth remains controversial.

As MMPs are known to play very crucial roles during the processes of tumor progression, several inhibitors specifically targeting MMPs are currently undergoing clinical trials. In the early '90s MMP inhibitors generated great enthusiasm among several research groups wishing to take them to clinical trials. Preclinical trials of MMP inhibitors were very promising, showing minimum side effects compared to other drugs available at the time71. Several current inhibitors, which have been tested in preclinical and clinical trials, are broad category MMP inhibitors. Pharmacological inhibitors such as batimastat and its analog marimastat, which interfere with the catalytic site of the MMPs, were the first inhibitors studied in detail. A recent review by Coussens et al discusses the status of several MMP inhibitors in clinical trials72. The first clinical trial of MMP inhibitors started in 1997 with marimastat and prinomastat. Phase 1 and 2 clinical trials were mostly focused on the optimal biological dose of MMP inhibitor rather than clinical outcome. Phase 2 and 3 clinical trials include three major strategies: (1) the clinical outcomes of MMP inhibitors were directly compared with standard chemotherapy. (2) MMP inhibitors were used in combination with standard chemotherapy. (3) Effects of MMP inhibitors were directly compared with placebo, which was administered in those patients who had no clinical evidence of disease after standard chemotherapy71. Therefore, the results of the MMP inhibitor clinical trials were very conclusive. Most of the MMP inhibitors tested in clinical trials were not very promising due to the lack of positive outcomes and the appearance of substantial drug side effects, which were not observed in preclinical studies. Therefore, most of the MMP inhibitor clinical trials were terminated following phase 3 clinical trials71. This failure of early MMP inhibitor clinical trials substantially suppressed the initiation of new clinical trials of MMP inhibitors targeting cancer.

Anti-angiogenic therapies in melanoma

Tumor vasculature is essential for a tumor's growth, progression and metastasis to distant sites. According to the original theory of Judah Folkman, the destruction of tumor vasculature must initiate the process of tumor suppression and death73. Based on the results of the most recently published studies, inhibition of angiogenesis might make tumors highly susceptible to radiation and chemotherapy74. Most importantly, the process of angiogenesis generally is not required for the normal physiology of normal adult organisms, with the exception of the female reproductive cycle or recovery from an injury. Therefore, minimal side effects are anticipated from the inhibition of this process. Based on preclinical data, one might predict the side effects will be restricted to impaired wound healing, a process known to be dependent on angiogenesis. Taken together, the predicted specificity and effectiveness of the approach initiated an interest in angiogenesis as a therapeutic target.

It is not surprising that numerous anti-angiogenic agents are currently in clinical trials or in development7578. Table 4 shows several classes of anti-angiogenic compounds divided into groups based on their molecular specificity. The list of molecular targets for anti-angiogenesis therapy includes: 1) Components or fragments of extracellular matrix and metalloproteinase inhibitors79,80; 2) Angiogenic growth factors and their receptors (expressed both on tumor as well as on endothelial cells). This category includes monoclonal antibodies and soluble forms of receptors designed to bind and neutralize growth factors, small molecules designed to inhibit growth factor-receptor interaction or tyrosine kinase activity of receptors8183. In addition, we included oligosaccharide-based inhibitors of growth factor release from extracellular matrix into this category. 3) A class of compounds which targets cellular receptors for extracellular matrix, integrins expressed on both melanoma and endothelial cells84. There are monoclonal antibodies, peptides and small molecule inhibitors in this category. All of the drugs presented in Table 4 have demonstrated significant inhibition of tumor angiogenesis and tumor growth in preclinical studies. Many experimental models included melanoma85.

Table 4.

Major angiogenic molecules targeted for cancer therapy, clinical approaches and outcome.

Category Molecular Target Class Name Preclinical studies Clinical trials status in melanoma or other cancers Some observed side effects Refs

Angiogenic growth factors and its receptors VEGF Monoclonal antibody Bevacizumab Very potent inhibitor of melanomas in several in vivo models Phase II (response observed) Minimal toxicity, delayed wound healing 75
Tyrosine kinase receptors Small molecule Sorafenib Inhibitor of various tumors in vivo and in vitro Phase II ( minimal effect as a single agent) Well tolerated 76
VEGF Soluble VEGF receptor VEGF-trap Potent inhibitor in various in vivo models Phase I/II Same as above 77
VEGF receptor-1 Small molecule Semaxinib (SU-5416) Effective in vivo Phase II (partial response observed) Deep vein thrombosis 78
VEGF receptors Small molecule PTK/ZK Effective in vivo (including melanomas) Phase I/II Well tolerated 81
Heparanase inhibitor (VEGF and FGF) Mixture of oligosaccharides PI-88 Inhibitor of VEGF/FGF release and tumor growth and invasion in vivo Phase I (partial response, recommended for phase II) Well tolerated, thrombocytopenia 82
VEGF receptor-2 Monoclonal antibody DC101 Very potent in vivo -- -- 83

Receptors for extracellular matrix, integrins Integrin αvβ3 Monoclonal antibody Vitaxin (MEDI-522) Potent in vivo inhibitor of tumor growth and metastasis Phase I (recommended for phase II) Limited toxicity (MI) 37
αv integrins Monoclonal antibody CNTO95 Same Phase I/II Well tolerated 84
alphavbeta3/alphav beta5 Small molecule Cilengitide (EMD 121974) Same Phase I/II Well tolerated 34

Components of extracellular matrix and proteases MMPs Small molecule Batimastat Inhibits melanoma growth and lung metastasis in mice Phase I/II, limited activity Poor solubility 79
MMPs Small molecule Marimastat Inhibits melanoma growth in vivo Phase I/I Muscle pain and stiffness 80
Extracellular matrix Fragment of matrix Endostatin Potent angiogenesis inhibitor in vitro and in vivo Phase I Minimal toxicity

Complex mechanism of action Angiogenesis inhibitors and immunomodulators Small molecule Thalidomide and lenalidomide Effective in vivo Phase II Neuropathy, skin rash 78
Cyclooxygenase-2 Small molecule Celecoxib Effective in vivo Phase I/II (shows response) Well tolerated 85

At present, it is quite intriguing to follow the results of clinical studies. Not all of the trials have been reported and discussed in detail in the literature. As a result, the overall picture of the efficacy of various groups of anti-angiogenic compounds is constantly changing, making any sort of generalization challenging. An interest in anti-angiogenic therapy dramatically increased after the US Food and Drug Administration (FDA) approved bevacizumab (Avastin) for several forms of cancers86. Inhibition of angiogenesis by bevacizumab appears to be a promising and effective strategy for solid tumors. It has been successfully used alone as well as in combination with chemotherapy treatments in several types of cancers86. Bevacizumab is relatively well tolerated; however, some observed side effects were surprising and not predicted based on the known mechanism of action for this antibody. Thus, it appears that additional preclinical studies will be required to fully address the effects of bevacizumab and its underlying mechanism on normal functional characteristics of vascular and hemostatic systems. A PUBMED search using the combination of key words "bevacizumab and melanoma" in the clinical trials category resulted in "0" hits. However, based on information from the NIH website "www.ClinicalTrials.gov" it appears that at least seven studies of bevacizumab as monotherapy or in combination with other compounds (carboplatin, paclitaxel, sorafenib, dacarbazine, interferon-alpha, imatinib were listed) in melanoma are currently recruiting patients. Results of bevacizumab clinical trials in melanoma are listed as "to be published" on http://www.centerwatch.com/patient/trialresults/druglst_Avastin_bevacizumab.html Since bevacizumab was the first FDA approved anti-angiogenic compound, it will most likely serve as a gold standard for other therapies which are currently in development. However, it is clear that drugs designed to target the same pathway, or even the same molecule, are not anticipated to display similar efficacies or similar profiles of side effects in melanoma or other cancer patients.

In sum, the process of angiogenesis in melanoma is crucial for tumor development and metastasis and undoubtedly requires further detailed investigation of its underlying mechanisms. Since this process involves a synergistic action of several classes of molecules and signaling pathways, there are numerous possibilities for the development of non-overlapping therapeutic strategies. Several angiogenic compounds are already in clinical trials for melanoma and available preliminary results are encouraging.

Acknowledgements

We acknowledge financial support from the US National Institutes of Health (HL071625 and HL073311 to TVB) and American Heart Association (0625271B to GHM).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Folkman J. Angiogenesis. Annu Rev Med. 2006;57:1–18. doi: 10.1146/annurev.med.57.121304.131306. [DOI] [PubMed] [Google Scholar]
  • 2.Warren BA, Shubik P. The growth of the blood supply to melanoma transplants in the hamster cheek pouch. Lab Invest. 1966;15(2):464–478. [PubMed] [Google Scholar]
  • 3.Hubler WR, Jr, Wolf JE., Jr Melanoma. Tumor angiogenesis and human neoplasia. Cancer. 1976;38(1):187–192. doi: 10.1002/1097-0142(197607)38:1<187::aid-cncr2820380129>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
  • 4.Elder DE, Van Belle P, Elenitasas R, Halpern A, Guerry D. Neoplastic progression and prognosis in melanoma. Semin Cutan Med Surg. 1996;15:336–348. doi: 10.1016/s1085-5629(96)80047-2. [DOI] [PubMed] [Google Scholar]
  • 5.Meier F, Satyamoorthy K, Nesbit M, Hsu MY, Schittek B, Garbe C, Herlyn M. Molecular events in melanoma development and progression. Front Biosci. 1998;3:D1005–D1010. doi: 10.2741/a341. [DOI] [PubMed] [Google Scholar]
  • 6.Tuthill RJ, Reed RI. Failure of Senescence in the Dysplasia-Melanoma Sequence: Demonstration using a Tissue Microarray and a Revised Paradigm for Melanoma. Semin Oncology. 2007 doi: 10.1053/j.seminoncol.2007.09.014. [DOI] [PubMed] [Google Scholar]
  • 7.Jhappan C, Noonan FP, Merlino G. Ultraviolet radiation and cutaneous malignant melanoma. Oncogene. 2003;22:3099–3112. doi: 10.1038/sj.onc.1206450. [DOI] [PubMed] [Google Scholar]
  • 8.Lin JY, Fisher DE. Melanocyte biology and skin pigmentation. Nature. 2007;445:843–850. doi: 10.1038/nature05660. [DOI] [PubMed] [Google Scholar]
  • 9.Cooper K, et al. Semin Oncology. 2007 Jul 22; [Google Scholar]
  • 10.Michaylira CZ, Nakagawa H. Hypoxic miroenvirornment as a cradle for melanoma development and progression. Cancer Biol Ther. 2006;5:476–479. doi: 10.4161/cbt.5.5.2749. [DOI] [PubMed] [Google Scholar]
  • 11.Hendrix MJ, Seftor EA, Seftor RE, Kasemeier-Kulesa J, Kulesa PM, Postovit LM. Reprogramming metastatic tumor cells with embryonic microenvironments. Nat Rev Cancer. 2007;7:246–255. doi: 10.1038/nrc2108. [DOI] [PubMed] [Google Scholar]
  • 12.Wanebo HJ, Argiris A, Bergsland E, et al. Targeting growth factors and angiogenesis; using small molecules in malignancy. Cancer Metastasis Rev. 2006;25(2):279–292. doi: 10.1007/s10555-006-8508-2. [DOI] [PubMed] [Google Scholar]
  • 13.Gille J. Antiangiogenic cancer therapies get their act together: current developments and future prospects of growth factor- and growth factor receptor-targeted approaches. Exp Dermatol. 2006;15(3):175–186. doi: 10.1111/j.1600-0625.2006.00400.x. [DOI] [PubMed] [Google Scholar]
  • 14.Sparano JA, Gray R, Giantonio B, et al. Evaluating antiangiogenesis agents in the clinic: the Eastern Cooperative Oncology Group Portfolio of Clinical Trials. Clin Cancer Res. 2004;10(4):1206–1211. doi: 10.1158/1078-0432.ccr-03-0238. [DOI] [PubMed] [Google Scholar]
  • 15.Motl S. Bevacizumab in combination chemotherapy for colorectal and other cancers. Am J Health Syst Pharm. 2005;62(10):1021–1032. doi: 10.1093/ajhp/62.10.1021. [DOI] [PubMed] [Google Scholar]
  • 16.Vacca A, Ria R, Ribatti D, et al. [Angiogenesis and tumor progression in melanoma] Recenti Prog Med. 2000;91(11):581–587. [PubMed] [Google Scholar]
  • 17.Plum SM, Holaday JW, Ruiz A, et al. Administration of a liposomal FGF-2 peptide vaccine leads to abrogation of FGF-2-mediated angiogenesis and tumor development. Vaccine. 2000;19(9–10):1294–1303. doi: 10.1016/s0264-410x(00)00210-3. [DOI] [PubMed] [Google Scholar]
  • 18.Sessa C, Vigano L, Grasselli G, et al. Phase I clinical and pharmacological evaluation of the multi-tyrosine kinase inhibitor SU006668 by chronic oral dosing. Eur J Cancer. 2006;42(2):171–178. doi: 10.1016/j.ejca.2005.09.033. [DOI] [PubMed] [Google Scholar]
  • 19.Odorisio T, Cianfarani F, Failla CM, et al. The placenta growth factor in skin angiogenesis. J Dermatol Sci. 2006;41(1):11–19. doi: 10.1016/j.jdermsci.2005.08.008. [DOI] [PubMed] [Google Scholar]
  • 20.Donnini S, Machein MR, Plate KH, et al. Expression and localization of placenta growth factor and PIGF receptors in human meningiomas. J Pathol. 1999;189(1):66–71. doi: 10.1002/(SICI)1096-9896(199909)189:1<66::AID-PATH390>3.0.CO;2-X. [DOI] [PubMed] [Google Scholar]
  • 21.Luttun A, Autiero M, Tjwa M, et al. Genetic dissection of tumor angiogenesis: are PIGF and VEGFR-1 novel anti-cancer targets? Biochim Biophys Acta. 2004;1654(1):79–94. doi: 10.1016/j.bbcan.2003.09.002. [DOI] [PubMed] [Google Scholar]
  • 22.Bar-Eli M. Role of interleukin-8 in tumor growth and metastasis of human melanoma. Pathobiology. 1999;67(1):12–18. doi: 10.1159/000028045. [DOI] [PubMed] [Google Scholar]
  • 23.Liu G, Zhang F, Lee J, et al. Selective induction of interleukin-8 expression in metastatic melanoma cells by transforming growth factor-beta 1. Cytokine. 2005;31(3):241–249. doi: 10.1016/j.cyto.2005.03.008. [DOI] [PubMed] [Google Scholar]
  • 24.Mahler DA, Huang S, Tabrizi M, et al. Efficacy and safety of a monoclonal antibody recognizing interleukin-8 in COPD: a pilot study. Chest. 2004;126(3):926–934. doi: 10.1378/chest.126.3.926. [DOI] [PubMed] [Google Scholar]
  • 25.Melnikova VO, Bar-Eli M. Bioimmunotherapy for melanoma using fully human antibodies targeting MCAM/MUC18 and IL-8. Pigment Cell Res. 2006;19(5):395–405. doi: 10.1111/j.1600-0749.2006.00331.x. [DOI] [PubMed] [Google Scholar]
  • 26.Mueller BM. Different roles for plasminogen activators and metalloproteinases in melanoma metastasis. Curr Top Microbiol Immunol. 1996;213(Pt 1):65–80. doi: 10.1007/978-3-642-61107-0_5. [DOI] [PubMed] [Google Scholar]
  • 27.Delbaldo C, Masouye I, Saurat JH, et al. Plasminogen activation in melanocytic neoplasia. Cancer Res. 1994;54(16):4547–4552. [PubMed] [Google Scholar]
  • 28.Hearing VJ, Law LW, Corti A, et al. Modulation of metastatic potential by cell surface urokinase of murine melanoma cells. Cancer Res. 1988;48(5):1270–1278. [PubMed] [Google Scholar]
  • 29.Min HY, Doyle LV, Vitt CR, et al. Urokinase receptor antagonists inhibit angiogenesis and primary tumor growth in syngeneic mice. Cancer Res. 1996;56(10):2428–2433. [PubMed] [Google Scholar]
  • 30.Berkenblit A, Matulonis UA, Kroener JF, et al. A6, a urokinase plasminogen activator (uPA)-derived peptide in patients with advanced gynecologic cancer: a phase I trial. Gynecol Oncol. 2005;99(1):50–57. doi: 10.1016/j.ygyno.2005.05.023. [DOI] [PubMed] [Google Scholar]
  • 31.Muehlenweg B, Sperl S, Magdolen V, et al. Interference with the urokinase plasminogen activator system: a promising therapy concept for solid tumours. Expert Opin Biol Ther. 2001;1(4):683–691. doi: 10.1517/14712598.1.4.683. [DOI] [PubMed] [Google Scholar]
  • 32.Kuphal S, Bauer R, Bosserhoff AK. Integrin signaling in malignant melanoma. Cancer Metastasis Rev. 2005;24(2):195–222. doi: 10.1007/s10555-005-1572-1. [DOI] [PubMed] [Google Scholar]
  • 33.Hynes RO. The emergence of integrins: a personal and historical perspective. Matrix Biol. 2004;23(6):333–340. doi: 10.1016/j.matbio.2004.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Eskens FA, Dumez H, Hoekstra R, et al. Phase I and pharmacokinetic study of continuous twice weekly intravenous administration of Cilengitide (EMD 121974), a novel inhibitor of the integrins alphavbeta3 and alphavbeta5 in patients with advanced solid tumours. Eur J Cancer. 2003;39(7):917–926. doi: 10.1016/s0959-8049(03)00057-1. [DOI] [PubMed] [Google Scholar]
  • 35.Stupack DG, Cheresh DA. Integrins and angiogenesis. Curr Top Dev Biol. 2004;64:207–238. doi: 10.1016/S0070-2153(04)64009-9. [DOI] [PubMed] [Google Scholar]
  • 36.Seftor RE. Role of the beta3 integrin subunit in human primary melanoma progression: multifunctional activities associated with alpha(v)beta3 integrin expression. Am J Pathol. 1998;153(5):1347–1351. doi: 10.1016/s0002-9440(10)65719-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Posey JA, Khazaeli MB, DelGrosso A, et al. A pilot trial of Vitaxin, a humanized anti-vitronectin receptor (anti alpha v beta 3) antibody in patients with metastatic cancer. Cancer Biother Radiopharm. 2001;16(2):125–132. doi: 10.1089/108497801300189218. [DOI] [PubMed] [Google Scholar]
  • 38.Nikolopoulos SN, Blaikie P, Yoshioka T, et al. Integrin beta4 signaling promotes tumor angiogenesis. Cancer Cell. 2004;6(5):471–483. doi: 10.1016/j.ccr.2004.09.029. [DOI] [PubMed] [Google Scholar]
  • 39.Ziober BL, Chen YQ, Ramos DM, et al. Expression of the alpha7beta1 laminin receptor suppresses melanoma growth and metastatic potential. Cell Growth Differ. 1999;10(7):479–490. [PubMed] [Google Scholar]
  • 40.Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 2004;5(10):816–826. doi: 10.1038/nrm1490. [DOI] [PubMed] [Google Scholar]
  • 41.Ramakrishnan V, Bhaskar V, Law DA, et al. Preclinical evaluation of an anti-alpha5beta1 integrin antibody as a novel anti-angiogenic agent. J Exp Ther Oncol. 2006;5(4):273–286. [PubMed] [Google Scholar]
  • 42.Brown LF, Papadopoulos-Sergiou A, Berse B, et al. Osteopontin expression and distribution in human carcinomas. Am J Pathol. 1994;145(3):610–623. [PMC free article] [PubMed] [Google Scholar]
  • 43.Geissinger E, Weisser C, Fischer P, et al. Autocrine stimulation by osteopontin contributes to antiapoptotic signalling of melanocytes in dermal collagen. Cancer Res. 2002;62(16):4820–4828. [PubMed] [Google Scholar]
  • 44.Reiniger IW, Wolf A, Welge-Lussen U, et al. Osteopontin as a serologic marker for metastatic uveal melanoma: results of a pilot study. Am J Ophthalmol. 2007;143(4):705–707. doi: 10.1016/j.ajo.2006.11.040. [DOI] [PubMed] [Google Scholar]
  • 45.Semba T, Funahashi Y, Ono N, et al. An angiogenesis inhibitor E7820 shows broad-spectrum tumor growth inhibition in a xenograft model: possible value of integrin alpha2 on platelets as a biological marker. Clin Cancer Res. 2004;10(4):1430–1438. doi: 10.1158/1078-0432.ccr-0109-03. [DOI] [PubMed] [Google Scholar]
  • 46.Pochec E, Litynska A, Amoresano A, et al. Glycosylation profile of integrin alpha 3 beta 1 changes with melanoma progression. Biochim Biophys Acta. 2003;1643(1–3):113–123. doi: 10.1016/j.bbamcr.2003.10.004. [DOI] [PubMed] [Google Scholar]
  • 47.Miller DH, Khan OA, Sheremata WA, et al. A controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med. 2003;348(1):15–23. doi: 10.1056/NEJMoa020696. [DOI] [PubMed] [Google Scholar]
  • 48.Felding-Habermann B, Fransvea E, O'Toole TE, et al. Involvement of tumor cell integrin alpha v beta 3 in hematogenous metastasis of human melanoma cells. Clin Exp Metastasis. 2002;19(5):427–436. doi: 10.1023/a:1016377114119. [DOI] [PubMed] [Google Scholar]
  • 49.Seftor RE, Seftor EA, Stetler-Stevenson WG, et al. The 72 kDa type IV collagenase is modulated via differential expression of alpha v beta 3 and alpha 5 beta 1 integrins during human melanoma cell invasion. Cancer Res. 1993;53(14):3411–3415. [PubMed] [Google Scholar]
  • 50.Tucker GC. Integrins: molecular targets in cancer therapy. Curr Oncol Rep. 2006;8(2):96–103. doi: 10.1007/s11912-006-0043-3. [DOI] [PubMed] [Google Scholar]
  • 51.Cianfrocca ME, Kimmel KA, Gallo J, et al. Phase 1 trial of the antiangiogenic peptide ATN-161 (Ac-PHSCN-NH(2)), a beta integrin antagonist, in patients with solid tumours. Br J Cancer. 2006;94(11):1621–1626. doi: 10.1038/sj.bjc.6603171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Overall CM, Lopez-Otin C. Strategies for MMP inhibition in cancer: innovations for the post-trial era. Nat Rev Cancer. 2002;2(9):657–672. doi: 10.1038/nrc884. [DOI] [PubMed] [Google Scholar]
  • 53.Chirco R, Liu XW, Jung KK, et al. Novel functions of TIMPs in cell signaling. Cancer Metastasis Rev. 2006;25(1):99–113. doi: 10.1007/s10555-006-7893-x. [DOI] [PubMed] [Google Scholar]
  • 54.Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 2006;69(3):562–573. doi: 10.1016/j.cardiores.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 55.Chen WT, Wang JY. Specialized surface protrusions of invasive cells, invadopodia and lamellipodia, have differential MT1-MMP, MMP-2, and TIMP-2 localization. Ann N Y Acad Sci. 1999;878:361–371. doi: 10.1111/j.1749-6632.1999.tb07695.x. [DOI] [PubMed] [Google Scholar]
  • 56.Hofmann UB, Westphal JR, Van Muijen GN, et al. Matrix metalloproteinases in human melanoma. J Invest Dermatol. 2000;115(3):337–344. doi: 10.1046/j.1523-1747.2000.00068.x. [DOI] [PubMed] [Google Scholar]
  • 57.Kerkela E, Saarialho-Kere U. Matrix metalloproteinases in tumor progression: focus on basal and squamous cell skin cancer. Exp Dermatol. 2003;12(2):109–125. doi: 10.1034/j.1600-0625.2003.120201.x. [DOI] [PubMed] [Google Scholar]
  • 58.Nikkola J, Vihinen P, Vuoristo MS, et al. High serum levels of matrix metalloproteinase-9 and matrix metalloproteinase-1 are associated with rapid progression in patients with metastatic melanoma. Clin Cancer Res. 2005;11(14):5158–5166. doi: 10.1158/1078-0432.CCR-04-2478. [DOI] [PubMed] [Google Scholar]
  • 59.Durko M, Navab R, Shibata HR, et al. Suppression of basement membrane type IV collagen degradation and cell invasion in human melanoma cells expressing an antisense RNA for MMP-1. Biochim Biophys Acta. 1997;1356(3):271–280. doi: 10.1016/s0167-4889(97)00004-9. [DOI] [PubMed] [Google Scholar]
  • 60.Cotignola J, Reva B, Mitra N, et al. Matrix Metalloproteinase-9 (MMP-9) polymorphisms in patients with cutaneous malignant melanoma. BMC Med Genet. 2007;8:10. doi: 10.1186/1471-2350-8-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Schnaeker EM, Ossig R, Ludwig T, et al. Microtubule-dependent matrix metalloproteinase-2/matrix metalloproteinase-9 exocytosis: prerequisite in human melanoma cell invasion. Cancer Res. 2004;64(24):8924–8931. doi: 10.1158/0008-5472.CAN-04-0324. [DOI] [PubMed] [Google Scholar]
  • 62.Wojtowicz-Praga S, Torri J, Johnson M, et al. Phase I trial of Marimastat, a novel matrix metalloproteinase inhibitor, administered orally to patients with advanced lung cancer. J Clin Oncol. 1998;16(6):2150–2156. doi: 10.1200/JCO.1998.16.6.2150. [DOI] [PubMed] [Google Scholar]
  • 63.Rudek MA, Figg WD, Dyer V, et al. Phase I clinical trial of oral COL-3, a matrix metalloproteinase inhibitor, in patients with refractory metastatic cancer. J Clin Oncol. 2001;19(2):584–592. doi: 10.1200/JCO.2001.19.2.584. [DOI] [PubMed] [Google Scholar]
  • 64.Jin X, Yagi M, Akiyama N, et al. Matriptase activates stromelysin (MMP-3) and promotes tumor growth and angiogenesis. Cancer Sci. 2006;97(12):1327–1334. doi: 10.1111/j.1349-7006.2006.00328.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wojtowicz-Praga S, Low J, Marshall J, et al. Phase I trial of a novel matrix metalloproteinase inhibitor batimastat (BB-94) in patients with advanced cancer. Invest New Drugs. 1996;14(2):193–202. doi: 10.1007/BF00210790. [DOI] [PubMed] [Google Scholar]
  • 66.Bodey B, Bodey B, Jr, Groger AM, et al. Invasion and metastasis: the expression and significance of matrix metalloproteinases in carcinomas of the lung. In Vivo. 2001;15(2):175–180. [PubMed] [Google Scholar]
  • 67.Airola K, Karonen T, Vaalamo M, et al. Expression of collagenases-1 and -3 and their inhibitors TIMP-1 and -3 correlates with the level of invasion in malignant melanomas. Br J Cancer. 1999;80(5–6):733–743. doi: 10.1038/sj.bjc.6690417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hofmann UB, Westphal JR, Zendman AJ, et al. Expression and activation of matrix metalloproteinase-2 (MMP-2) and its co-localization with membrane-type 1 matrix metalloproteinase (MT1-MMP) correlate with melanoma progression. J Pathol. 2000;191(3):245–256. doi: 10.1002/1096-9896(2000)9999:9999<::AID-PATH632>3.0.CO;2-#. [DOI] [PubMed] [Google Scholar]
  • 69.Anand-Apte B, Bao L, Smith R, et al. A review of tissue inhibitor of metalloproteinases-3 (TIMP-3) and experimental analysis of its effect on primary tumor growth. Biochem Cell Biol. 1996;74(6):853–862. doi: 10.1139/o96-090. [DOI] [PubMed] [Google Scholar]
  • 70.Bloomston M, Shafii A, Zervos E, et al. TIMP-1 antisense gene transfection attenuates the invasive potential of pancreatic cancer cells in vitro and inhibits tumor growth in vivo. Am J Surg. 2005;189(6):675–679. doi: 10.1016/j.amjsurg.2005.03.008. [DOI] [PubMed] [Google Scholar]
  • 71.Pavlaki M, Zucker S. Matrix metalloproteinase inhibitors (MMPIs): the beginning of phase I or the termination of phase III clinical trials. Cancer Metastasis Rev. 2003;22(2–3):177–203. doi: 10.1023/a:1023047431869. [DOI] [PubMed] [Google Scholar]
  • 72.Coussens LM, Fingleton B, Matrisian LM. Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science. 2002;295(5564):2387–2392. doi: 10.1126/science.1067100. [DOI] [PubMed] [Google Scholar]
  • 73.Folkman J. Tumor angiogensis: role in regulation of tumor growth. Symp Soc Dev Biol. 1974;30(0):43–52. [PubMed] [Google Scholar]
  • 74.Jain RK, Duda DG, Clark JW, et al. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Pract Oncol. 2006;3(1):24–40. doi: 10.1038/ncponc0403. [DOI] [PubMed] [Google Scholar]
  • 75.Shih T, Lindley C. Bevacizumab: an angiogenesis inhibitor for the treatment of solid malignancies. Clin Ther. 2006;28(11):1779–1802. doi: 10.1016/j.clinthera.2006.11.015. [DOI] [PubMed] [Google Scholar]
  • 76.Eisen T, Ahmad T, Flaherty KT, et al. Sorafenib in advanced melanoma: a Phase II randomised discontinuation trial analysis. Br J Cancer. 2006;95(5):581–586. doi: 10.1038/sj.bjc.6603291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rudge JS, Thurston G, Davis S, et al. VEGF trap as a novel antiangiogenic treatment currently in clinical trials for cancer and eye diseases, and VelociGene- based discovery of the next generation of angiogenesis targets. Cold Spring Harb Symp Quant Biol. 2005;70:411–418. doi: 10.1101/sqb.2005.70.052. [DOI] [PubMed] [Google Scholar]
  • 78.Mita MM, Rowinsky EK, Forero L, et al. A phase II, pharmacokinetic, and biologic study of semaxanib and thalidomide in patients with metastatic melanoma. Cancer Chemother Pharmacol. 2007;59(2):165–174. doi: 10.1007/s00280-006-0255-0. [DOI] [PubMed] [Google Scholar]
  • 79.Rasmussen HS, McCann PP. Matrix metalloproteinase inhibition as a novel anticancer strategy: a review with special focus on batimastat and marimastat. Pharmacol Ther. 1997;75(1):69–75. doi: 10.1016/s0163-7258(97)00023-5. [DOI] [PubMed] [Google Scholar]
  • 80.Quirt I, Bodurth A, Lohmann R, et al. Phase II study of marimastat (BB-2516) in malignant melanoma: a clinical and tumor biopsy study of the National Cancer Institute of Canada Clinical Trials Group. Invest New Drugs. 2002;20(4):431–437. doi: 10.1023/a:1020625423524. [DOI] [PubMed] [Google Scholar]
  • 81.Lee L, Sharma S, Morgan B, et al. Biomarkers for assessment of pharmacologic activity for a vascular endothelial growth factor (VEGF) receptor inhibitor, PTK787/ZK 222584 (PTK/ZK): translation of biological activity in a mouse melanoma metastasis model to phase I studies in patients with advanced colorectal cancer with liver metastases. Cancer Chemother Pharmacol. 2006;57(6):761–771. doi: 10.1007/s00280-005-0120-6. [DOI] [PubMed] [Google Scholar]
  • 82.Basche M, Gustafson DL, Holden SN, et al. A phase I biological and pharmacologic study of the heparanase inhibitor PI-88 in patients with advanced solid tumors. Clin Cancer Res. 2006;12(18):5471–5480. doi: 10.1158/1078-0432.CCR-05-2423. [DOI] [PubMed] [Google Scholar]
  • 83.Miller DW, Vosseler S, Mirancea N, et al. Rapid vessel regression, protease inhibition, and stromal normalization upon short-term vascular endothelial growth factor receptor 2 inhibition in skin carcinoma heterotransplants. Am J Pathol. 2005;167(5):1389–1403. doi: 10.1016/S0002-9440(10)61226-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Trikha M, Zhou Z, Nemeth JA, et al. CNTO 95, a fully human monoclonal antibody that inhibits alphav integrins, has antitumor and antiangiogenic activity in vivo. Int J Cancer. 2004;110(3):326–335. doi: 10.1002/ijc.20116. [DOI] [PubMed] [Google Scholar]
  • 85.Gogas H, Polyzos A, Stavrinidis I, et al. Temozolomide in combination with celecoxib in patients with advanced melanoma. A phase II study of the Hellenic Cooperative Oncology Group. Ann Oncol. 2006;17(12):1835–1841. doi: 10.1093/annonc/mdl311. [DOI] [PubMed] [Google Scholar]
  • 86.Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med. 2004;10(2):145–147. doi: 10.1038/nm988. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES