Open access

Role of Inflammation and Angiogenic Growth Factors in Malignant Mesothelioma

Written By

Loredana Albonici, Camilla Palumbo and Vittorio Manzari

Submitted: 16 September 2011 Published: 19 December 2012

DOI: 10.5772/47951

From the Edited Volume

Malignant Mesothelioma

Edited by Carmen Belli and Santosh Anand

Chapter metrics overview

1,876 Chapter Downloads

View Full Metrics

1. Introduction

Malignant mesothelioma (MM) is a highly aggressive tumor which arises from the mesothelial cell lining of the serosal surfaces, most cases (>90%) being of pleural origin (Attanoos & Gibbs, 1997; Robinson & Lake, 2005). The pathogenesis of MM has been mainly associated with previous asbestos exposure (Berman & Crump, 2008), with a latency period of up to 40 years, although other agents such as Simian virus 40 (SV40) or genetic susceptibility factors have been linked to the development of this tumor (Carbone et al., 2002; Pisick & Salgia, 2005). Indeed, human mesothelial cells are highly susceptible to SV40-mediated transformation in vitro and SV40 DNA sequences and large T antigen (Tag) have been detected in human MM cells (Bocchetta et al., 2000; Carbone et al., 2012; Gazdar et al., 2003).

MM is largely unresponsive to conventional chemotherapy or radiotherapy and, despite its low metastatic efficiency, it is highly invasive to surrounding tissues so that its extensive growth leads to the failure of the organs underlying the serosal membranes (Astoul, 1999). In fact, the primary cause of fatality in MM is related to the propensity of the tumor cells to invade locally, even though MM metastasis are more common after surgery and, at the autopsy, metastatic diffusion is observed in 50% of patients (Astoul, 1999). At present, the median survival from diagnosis of MM is less than two years (Palumbo et al., 2008).

The mesothelium is not just a passive protective surface, but a highly dynamic membrane (Mutsaers, 2004). It consists of a single layer of elongated, flattened, squamous-like cells of mesodermal origin, characterized by dual epithelial/mesenchymal features. Cuboidal mesothelial cells can also be found at various locations in physiological conditions. Further, mesothelial cells can adopt a cuboidal morphology, which reflects a metabolically activated state, after injury or stimulation of the serosal surface (Mutsaers, 2004). Indeed, mesothelial cells are sentinel cells that can sense and respond to a variety of signals within their microenvironment. They participate in serosal inflammation by secreting both pro- and anti-inflammatory as well as immunomodulatory mediators. Besides, these cells can act as antigen presenting cells for T lymphocytes (Hausmann et al., 2000), regulate tissue repair, control fibrin deposition and breakdown, and modulate adhesion, growth and dissemination of tumor cells metastasizing to the serosal membranes (Mutsaers, 2002). In particular, in response to different types of stimuli, including cytokines and asbestos fibers, mesothelial cells have been reported to release prostaglandins, chemokines, reactive oxygen and nitrogen species and growth factors which represent key effectors in the modulation of inflammatory reactions that occur in response to pleural injury (Fleury-Feith et al., 2003; Mutsaers, 2002).

Advertisement

2. Asbestos-induced carcinogenesis as an inflammation-driven process

The association between exposure to asbestos fibers and development of lung cancer and mesothelioma is well established in both humans and animals models (Greillier & Astoul, 2008; Huang et al., 2011; Mossman & Churg, 1998; Yarborough, 2007). A variety of mediators, either generated directly from asbestos fibers or elaborated intracellularly or extracellularly by cells exposed to asbestos, are implicated in the initiation and promotion of mesothelial cell transformation.

The mechanisms underlying asbestos-induced carcinogenesis involve mutagenic and non-mutagenic pathways, the latter including inflammation, enhanced mitogenesis, cell signaling alterations, and cytotoxic apoptosis/necrosis. Neither of these two mechanisms alone fully accounts for the complex biological abnormalities produced by asbestos fibers, even though in MM asbestos appears to act as a complete carcinogen (Dong et al., 1994; Huang et al., 2011). Still, the chronic inflammatory response induced by asbestos inhalation seems to play a critical role in mesothelial cell transformation.

Asbestos exposure induces an inflammatory reaction with a large component of mononuclear phagocytes (Antony et al., 1993; Branchaud et al., 1993; Carbone et al., 2012; Choe et al., 1997). Upon differentiation into macrophages, these cells phagocytize asbestos fibers and, in response, release numerous cytokines and reactive oxygen species with mutagenic properties (Robledo & Mossman, 1999). Thus, many of the pathological consequences occurring in the lung following exposure to asbestos fibers are believed to arise from an inflammatory cascade involving both autocrine and paracrine events (Hillegass et al., 2010). Persistent pulmonary inflammation is observed in animal models of asbestosis that can be correlated with fibroproliferative responses (Mossman & Churg, 1998).

Experimental models, as well as in vitro studies, have shown that mesothelial cells are particularly susceptible to the cytotoxic effects of asbestos (Baldys et al., 2007; BéruBé et al., 1996; Broaddus et al., 1996). Asbestos does not induce transformation of primary human mesothelial cells in vitro, instead, it is very cytotoxic to this cell type, causing extensive cell death. This finding raised an apparent paradoxical issue of how asbestos causes MM if human mesothelial cells exposed to this mineral die (Liu et al., 2000). This apparent paradox is reconciled by the current hypothesis that the chronic inflammation induced by asbestos leads to the persistent activation of the nuclear factor kappa B (NF-κB) transcription factor, which in turn mediates the activation of prosurvival genes and prevents apoptosis of the damaged mesothelial cells (Mantovani et al., 2008; Micheau & Tschopp, 2003; Philip et al., 2004). This allows mesothelial cells with asbestos-induced DNA damage to survive and divide rather than die and, if sufficient genetic damage accumulates, to eventually develop into a MM (Miura et al., 2006; Nymark, 2007). In fact, apoptosis is an important mechanism by which cells with DNA damage are eliminated without eliciting an inflammatory response (Ullrich et al., 2008; Yoshida et al., 2010). However, failure of apoptosis in cells with unrepaired DNA and chromosomal damage after chronic exposure to asbestos may lead to permanent genetic alterations and trigger the development of a clone of cancerous cells (Roos & Kaina, 2006; Wu, 2006). Consistently, MM cells are found to be apoptosis-resistant as compared to primary cultured mesothelial cells (Fennel & Rudd, 2004; Villanova et al., 2008).

2.1. Tumor Necrosis Factor-α and other pro-inflammatory cytokines

Tumor Necrosis Factor-α (TNF-α) is probably the most studied candidate for initiating inflammatory and fibrotic events linked to lung diseases such as asbestosis. Asbestos fibers cause the accumulation of macrophages in the pleura and lung. When these macrophages encounter asbestos, they release TNF-α. At the same time, asbestos induces the secretion of TNF-α and the expression of TNF-α receptor I (TNF-RI) in mesothelial cells (Yang et al., 2006). Remarkably, treatment of mesothelial cells with TNF-α significantly reduced asbestos cytotoxicity. Indeed, TNF-α activates NF-κB, which in turn promotes mesothelial cell survival and resistance to the cytotoxic effects of asbestos. Thus, TNF-α signaling through NF-κB-dependent mechanisms increases the percentage of mesothelial cells that survive asbestos exposure, thereby increasing the pool of asbestos-damaged cells susceptible to malignant transformation (Haegens et al., 2007; Janssen-Heininger et al., 1999; Yang et al., 2006).

It has been reported that rats receiving a single intratracheal instillation of fibrogenic chrysotile asbestos developed lung chronic inflammatory reactions characterized by the accumulation of alveolar macrophages producing elevated levels of both Interleukin (IL)-1 and IL-6 (Lemaire & Ouellet, 1996). An increased production and/or release of these cytokines triggers inflammatory cell recruitment, thus amplifying and sustaining local inflammation. It has also been demonstrated that crocidolite asbestos and TNF-α can stimulate a dose-dependent increase in IL-6 expression and secretion from cultured, transformed and normal, human alveolar type II epithelial cells that is dependent upon intracellular redox potential (Simeonova et al., 1997). Interestingly, although MM cells appear to express low levels of IL-6 receptor (IL-6R), IL-6 can act as a growth factor for these cells through a trans-signaling mechanism involving the interaction of macromolecular complexes of IL-6 and soluble IL-6R (sIL-6R) with the transmembrane glycoprotein gp130 expressed on the surface of MM cells (Adachi et al., 2006; Rose-John et al., 2007). High levels of both IL-6 and sIL-6R are typical of several chronic inflammatory conditions (Rose-John et al., 2007).

Thus, inflammatory cytokines such as TNF-α and IL-6 appear to play a dual role in MM pathogenesis: they induce and sustain pleural inflammation and at the same time can act as survival or mitogenic factors for normal and transformed mesothelial cells, respectively.

2.2. Reactive Oxygen and Nitrogen Species (ROS/RNS)

The mechanisms of injury and disease development caused by asbestos fibers are presumed to be related to their greater fibrogenic and carcinogenic properties in comparison to other minerals. Asbestos–induced mutagenicity is mediated through both direct and indirect pathways. Asbestos fibers may induce mutagenicity and genotoxicity directly through physical interaction with the mitotic machinery after being phagocytized by the target cells, or indirectly as a result of DNA and chromosome damage caused by asbestos-induced reactive oxygen (ROS) and nitrogen species (RNS) (Kamp & Weitzman, 1999; Shukla et al., 2003a, 2003b). ROS and RNS can be generated primarily by asbestos fibers or secondarily through fiber-induced inflammation (Aust et al., 2011; Gulumian, 2005; Hoidal, 2001). Free radicals generated from asbestos fibers plus the direct damage induced by the fibers are linked to cell signaling, inflammation, and a plethora of other responses (mutagenesis, proliferation, etc.) associated with the pathogenesis of asbestos-associated diseases (Heinz et al., 2010; Manning et al., 2002; Shukla et al., 2003a, 2003b).

Several evidences indicate that a main factor in determining the surface and biological reactivity of different types of asbestos fibers is their ability to participate in redox reactions that generate free radicals (Kamp & Weitzman, 1999; Shukla et al., 2003a). Although the nature of the free radical-generating surface sites on asbestos fibers is not yet clear, asbestos fibers have an intrinsic redox activity and contain ferrous iron, which catalyzes reactions generating active oxygen intermediates on the fiber surface. Within the tissues several asbestos fiber types can produce reactive oxygen free radicals from hydrogen peroxide, a common product of intermediary tissue metabolism. Epidemiological studies have identified crocidolite as one of the most potent forms of asbestos associated with the induction of MM (Heintz et al., 2010). Crocidolite has a greater surface-area and a higher ferrous iron content compared to other fiber types such as chrysotile, and it is more biologically active in the generation of free radicals (Toyokuni, 2009). However, the ability of asbestos fibers to elicit these effects is not related to total iron content, suggesting the presence of specific iron active sites at the fibers’ surface (Shukla et al., 2003a).

Cells exposed to asbestos have also been reported to produce a higher amount of nitric oxide (NO). In this regard, it has been reported that in human mesothelial cells crocidolite increases the expression of the inducible NO synthase (NOS) isoform (iNOS), the activity of the constitutive endothelial NOS (eNOS), and the synthesis of NO via NF-κB and Akt activation (Riganti et al., 2007). Thus, the asbestos-induced upregulation of iNOS or NO in the lungs, as well as the induction of inflammation by fibers, may contribute along with ROS, to the pathogenesis of lung and pleural injury (Hussain et al., 2003; Tanaka et al., 1998). Indeed, ROS and RNS can cause breakage of DNA, lipid peroxidation, release of inflammatory cytokines such as TNF-α, and the modification of cellular proteins including phosphatases involved in cell signaling cascades (Gossart et al., 1996; Hussain et al., 2003), so that their increased synthesis by various cell types may have multiple roles in cellular events critical to the establishment of lung and pleural inflammation and uncontrolled cell proliferation.

Finally, in mesothelial and lung epithelial cells asbestos fibers, as opposed to nonpathogenic minerals, cause a persistent induction of the redox-sensitive transcription factors NF-κB and Activator Protein-1 (AP-1), which is accompanied by chronic alterations in gene expression (Heintz et al., 1993; Janssen et al., 1995). As mentioned above, the aberrant activation of the NF-κB pathway is regarded as a critical event for mesothelial cell transformation (Toyooka et al., 2008).

2.3. Transcription factors

2.3.1. NF-κB

NF-κB proteins are dimeric transcription factors composed of five different subunits, namely p65 (RelA), RelB, c-Rel, NF-κB1 p50 and NF-κB2 p52, which regulate gene expression events that impact on cell survival and differentiation. Moreover, since activation of NF-κB is critical in up-regulating the expression of many genes linked to proliferation, apoptosis resistance, and chemokine/cytokine production, this is undoubtedly a critical transcription factor in inflammatory responses occurring in target cells of asbestos-related diseases (Janssen et al., 1995, 1997).

In unstimulated cells, the NF-κB transcription dimers are retained in the cytoplasm in an inactive state through the interaction with a family of inhibitors called IκBs (Inhibitors of κB) or with the p50 and p52 precursor proteins, p105 and p100, respectively (Hayden & Ghosh, 2008; Scheidereit, 2006). Indeed, p50 and p52 are translated as precursors proteins containing an IκB-like C-terminal portion (Sun, 2011).

Two different NF-κB-activation pathways exist: the classical and the alternative NF-κB pathway. The classical NF-κB pathway is initiated by signals elicited by diverse receptors, including TNF receptors type 1/2, Toll/IL-1 receptor, T-cell and B-cell receptors and EGF receptor, and also by cellular stresses and DNA damage (Hayden & Ghosh 2004; Le Page et al., 2005). These signals induce the activation of the IκB kinase (IKK) complex, which is composed by the catalytic subunits IKKα and IKKβ and by the regulatory subunit IKKγ/NEMO (Hayden & Ghosh, 2008; Scheidereit, 2006; Sun, 2011). The activated IKK complex phosphorylates IκB proteins, thereby triggering their proteasomal degradation. As a consequence, NF-κB dimers are released and can translocate into the nucleus. This pathway mainly leads to the activation of p50:RelA dimers (Sun, 2011). Conversely, the alternative NF-κB pathway predominantly targets activation of RelB:p52 complexes. This pathway relies on the inducible processing of p100 triggered by signaling from TNF receptor family members via the NF-κB-inducing kinase (NIK): NIK activates IKKα, which, in turn, phosphorylates p100 and triggers its processing to p52. This event results in the conversion of p100-inhibited NF-κB complexes into p52-containing NF-κB dimers, capable of translocating into the nucleus (Hayden & Ghosh, 2008; Scheidereit, 2006; Sun, 2011).

NF-κB-regulated genes have distinct requirements for NF-κB dimers. For instance, the NF-κB binding site of the IL-2 gene has been reported to bind preferentially c-Rel homodimers and p50:c-Rel, while that of the gene encoding IL-8 has been found to selectively bind Rel A (Hoffman et al., 2003, 2006). On the other hand, several genes are redundantly induced by more than one dimer (Hoffman et al., 2003, 2006; Saccani et al., 2003).

A number of studies have shown that nuclear retention and DNA binding of NF-κB protein complexes are increased following exposure of various cell types to a variety of extracellular stimuli that include oxidative stress (Bowie & O'Neill, 2000), hypoxia (Jung et al., 2003; Royds et al., 1998) and inflammatory cytokines (Mantovani et al., 2008). These observations are consistent with the hypothesis that persistent activation of NF-κB can contribute to the induction of multiple genes that are critical to the pathogenesis of asbestos-associated diseases, since oxidants, local hypoxia and inflammatory cytokines are all components involved in the effects induced by asbestos exposure.

It is noteworthy that among various carcinogenic and non-carcinogenic fibers studied for their effect on nuclear translocation of NF-κB, only carcinogenic fibers were found to cause a dose-dependent translocation of this transcription factor to the nucleus, and this effect was reported to be oxidative stress-dependent (Brown et al., 1999). In lung macrophages, the asbestos-induced expression and secretion of TNF-α are mediated by iron-catalyzed ROS products (Simeonova & Luster, 1995) through a process that involves NF-κB activation (Cheng et al., 1999). In rat alveolar type 2 cells, the crocidolite-induced activation of NF-κB as well as the expression of the macrophage inflammatory protein-2 (MIP-2) gene have also been shown to be dependent on mitochondrial-derived oxidative stress (Driscoll et al., 1998).

2.3.2. AP-1

AP-1 is a homo- or heterodimeric transcription factor composed by proteins encoded by the fos and jun early response proto-oncogenes. This family of proteins includes c-Fos, FosB, FosL1 (Fra-1), FosL2 (Fra-2), c-Jun, JunB and JunD (Milde-Langosch, 2005). Whereas Jun members are capable of forming homodimers able to bind DNA and regulate transcription, all Fos members must form heterodimers with Jun family members to bind DNA.

AP-1 is a redox-sensitive transcription factor typically associated with cell proliferation and tumor promotion (Eferl & Wagner, 2003). The first evidence showing that asbestos exerts regulatory effects linked to aberrant transcriptional responses, cell proliferation and cell transformation derives from studies in which asbestos fibers caused induction of c-fos and c-jun proto-oncogene mRNAs in pleural mesothelial cells and tracheo-bronchial epithelial cells in a dose–response fashion (Heintz et al., 1993).

The persistent induction of AP-1 by asbestos suggests a model of asbestos-induced carcinogenesis involving chronic stimulation of cell proliferation through activation of early response genes (Schonthaler et al., 2011). Of note, early response genes are a set of genes whose transcription is rapidly induced in response to growth factors. Furthermore, AP-1 activity is induced by growth factors, pro-inflammatory cytokines and genotoxic stress (Jochum et al., 2001; Shaulian & Karin, 2002). These stimuli activate mitogen-activated protein kinase (MAPK) cascades through the phosphorylation of distinct substrates such as ERK, JNK and p38 MAPK (Chang & Karin, 2001). Indeed, the MAPK signal transduction pathway uses AP-1 as a converging point not only to regulate the expression of various genes but also to autoregulate AP-1 gene transcription (Reuter et al., 2010).

Several genes, which play very important roles in injury, repair, and differentiation, contain binding site(s) for AP-1 in their promoter and/or enhancer regions (Chang & Karin, 2001). These genes include extracellular matrix metalloproteinases (MMPs), antioxidant enzymes, growth factors and their receptors, differentiation markers, cytokines, chemokines and other transcription factors (Shaulian & Karin, 2001).

2.3.3. Nuclear Factor of Activated T Cells (NFAT)

The Nuclear Factor of Activated T cells (NFAT) family of transcription factors consists of five proteins that are evolutionarily related to the Rel/NF-κB family. NFAT can be present in both the cytoplasm and the nucleus. In the cytoplasm NFAT is in a highly phosphorylated, inactive state. Cell stimuli leading to the elevation of intracellular Ca2+ levels induce the activation of the phosphatase PP2B/Calcineurin which dephosphorylates NFAT. This results in its nuclear relocalization and transcriptional activation. Interestingly, NFAT family members can act synergistically with AP-1 on composite DNA elements which contain adjacent NFAT and AP-1 binding sites (Macián et al., 2001). A functional cooperation has also been reported to occur between NFAT and NF-κB (Jash et al., 2012).

Initially, NFAT was identified in lymphocytes and was reported to be expressed in activated but not resting T cells (Macián et al., 2005; Shaw et al., 1988). NFAT regulates not only T cell activation and differentiation but also the function of other immune cells, including dendritic cells (DCs), B cells and megakaryocytes. In addition, NFAT has crucial roles in numerous developmental programs in vertebrates.

Dysregulation of NFAT signalling is now known to be associated with malignant transformation and the development of cancer (Mancini & Toker, 2009; Müller & Rao, 2010). The observation that NFAT can be activated by asbestos-induced oxidative stress suggests that this transcription factor may play multiple roles in asbestos-induced inflammation and carcinogenesis (Li et al., 2002). Indeed, NFAT mediates the expression of several inflammatory cytokines, including TNF-α, and is involved in cell transformation, proliferation, invasive migration, tumor cell survival and tumor angiogenesis (Mancini & Toker, 2009).

Advertisement

3. Multifaceted role of angiogenic growth factors in MM

Angiogenesis is a common feature of solid tumors. Indeed, the development of a clinically observable tumor requires the neoformation of a vascular network sufficient to sustain tumor growth (Ribatti et al., 2007). Tumor angiogenesis is stimulated by the secretion of angiogenic molecules which induce endothelial cells from nearby vessels to switch from a quiescent to an activated state. Further, upon the stimulation of angiogenic growth factors, activated endothelial cells disrupt the extracellular matrix, proliferate and migrate (Ribatti et al., 2007). Angiogenic growth factors include, among the others, Vascular Endothelial Growth Factor (VEGF), Placenta Growth Factor (PlGF), Platelet-Derived Growth Factor (PDGF) and acidic and basic Fibroblast Growth Factors (FGF-1 and -2, respectively). VEGF is regarded as the most important player in angiogenesis (Ono, 2008).

The link between angiogenesis and tumor progression is provided by the negative prognostic value of intratumoral microvascular density (IMD) (Folkman, 2006; Kerbel, 2008). In MM the IMD has an independent prognostic value (Kumar-Singh et al., 1997). MM demonstrates a higher IMD than colon and breast tumors and, consistently, presents with minimal central necrosis despite its huge size (Gasparini & Harris, 1995; Kumar-Singh et al., 1997).

On the other hand, the involvement of angiogenic growth factors in MM goes beyond the stimulation of angiogenesis. Indeed, as discussed below, MM cells express receptors for several angiogenic factors which, accordingly, can directly modulate MM cell behavior.

3.1. Angiogenic growth factors of the VEGF family

The human VEGF family consists of five members: VEGF (VEGF-A), VEGF-B, VEGF-C, VEGF-D and PlGF. These growth factors are secreted as dimers and their biological effects are mediated by binding to three tyrosine kinase receptors, i.e. VEGF-R1/Flt-1, VEGF-R2/KDR (whose murine homologue is known as Flk-1) and VEGF-R3/Flt-4, and two non-enzymatic co-receptors known as neuropilin-1 and -2 (Ferrara et al., 2003; Koch et al., 2011; Roskoski, 2007).

3.1.1. VEGF

VEGF is regarded as the major mediator of tumor angiogenesis. It is expressed in the majority of cancers and has a central role in tumor growth and metastasis. In fact, this growth factor is essential for the mobilization of bone-marrow-derived endothelial precursors in neovascularization (Asahara et al., 1999), and stimulates vascular endothelial cells mobility, proliferation and survival (Waltenberger et al., 1994).

High levels of VEGF are present both in malignant and non-malignant pleural effusions leading to increased vascular permeability. On the other hand, VEGF levels in serum or pleural effusions of MM patients are higher than those found in patients with non-malignant pleuritis or lung cancer involving malignant pleural effusions. Further, in MM patients elevated serum or pleural effusion levels of VEGF correlate with a worse prognosis and may also contribute to increase resistance to chemotherapy (Hirayama et al., 2011; Yasumitsu et al., 2010; Zebrowski et al., 1999). In fact, VEGF status has proved to be of value in predicting the effectiveness of radiotherapy and chemotherapy on different cancers (Choi et al., 2008; Kumar et al., 2009; Toi et al., 2001).

In addition to its role in tumor vascularization, VEGF can directly affect the behavior of cancer cells in an autocrine or paracrine manner. Indeed, many tumor cell types express VEGF receptors. VEGF has been found to promote the growth of transformed cell lines in vitro (Masood et al., 2001) and to act as a survival factor for tumor cells by enhancing the expression of the antiapoptotic factors bcl-2 (Harmey & Bouchier-Hayes, 2002) and survivin (Kanwar et al., 2011). In this context, MM cells have been shown to express high amounts of VEGF, VEGF receptors and co-receptors both in vitro and in vivo, and VEGF has been demonstrated to act as an autocrine growth factor for this tumor cell type (Albonici et al., 2009; Ohta et al., 1999; Pompeo et al., 2009; Strizzi et al., 2001a).

VEGF-R1 participates in cell migration; it has an important role in monocyte chemotaxis and promotes recruitment of circulating endothelial precursor cells from bone marrow (Hattori et al., 2002). Its expression is increased in various tumors, correlates with disease progression and can predict poor prognosis, metastasis and recurrent disease in humans (Dawson et al., 2009; Fischer et al., 2008; Kerber et al., 2008). This receptor is also expressed by MM cells in vitro and in vivo, where it appears to mediate proliferative and cell survival responses (Albonici et al., 2009; Strizzi et al., 2001a).

VEGF-R2 is the main mediator of VEGF-stimulated endothelial cell migration, proliferation, survival and enhanced vascular permeability (Olsson et al., 2006; Shibuya, 2006). VEGF-R2 expression is induced in conjunction with active angiogenesis, such as during the reparative process, and in pathological conditions associated with neovascularization, such as cancer (Plate et al., 1993). VEGF-R2 is overexpressed in MM cells and specimens, and VEGF-R2 silencing by small intefering RNA has been shown to induce cell death in MM or immortalized mesotelial cells in vitro (Albonici et al., 2009; Catalano et al., 2009; Pompeo et al., 2009; Strizzi et al., 2001a). Interestingly, it has been reported that in MM cells this receptor can be activated also via the semaphorin-6D receptor Plexin-A1, triggering a prosurvival program that promotes anchorage-independent growth through a NF-κB-dependent pathway (Catalano et al., 2009). Remarkably, the expression of plexin-A1 is induced by asbestos fibers and overexpression of plexin-A1 in non-malignant mesothelial cells inhibits cell death after asbestos exposure, thus suggesting a role for this receptor not only in MM promotion and progression but also in asbestos-induced mesothelial carcinogenesis (Catalano et al., 2009).

In vitro studies have shown that transfection of normal mesothelial cells with SV40 Tag potently increases VEGF protein and mRNA levels (Cacciotti et al., 2002) as well as mesothelial cell proliferation (Catalano et al., 2002). These data indicate that VEGF regulation by SV40 transforming proteins can also represent a key event in MM onset and progression.

3.1.2. PlGF

PlGF, originally identified in the placenta during the early embryonic development (Khaliq et al., 1996; Maglione et al., 1991), is expressed in several other organs including the heart, lung, thyroid, skeletal muscle and adipose tissue (Persico et al., 1999) but not normal mesothelium (Albonici et al., 2009).

Although the role exerted by PlGF in tumor growth is controversial yet, PlGF can stimulate vessel growth and maturation directly by affecting endothelial and mural cells, as well as indirectly by recruiting pro-angiogenic cell types (Barillari et al., 1998; Carmeliet, 2003). It also promotes the recruitment and maturation of angiogenesis-competent myeloid progenitors to growing sprouts and collateral vessels (Hattori et al., 2002; Luttun et al., 2002; Rafii et al., 2003). Further, PlGF is able to protect endothelial cells from apoptosis, in a similar manner as VEGF, by inducing the expression of antiapoptotic genes such as survivin (Adini et al., 2002).

Under pathological conditions, PlGF abundance is elevated in various cell types and tissues, including vascular endothelial cells, and many different tumor cells (Albonici et al., 2009; Cao et al., 1996; Fischer et al., 2007; Oura et al., 2003). PlGF expression is switched on in hyperplastic/reactive mesothelium and in MM cells (Albonici et al., 2009). Moreover, in MM as well as in different types of cancer, including melanoma, gastric, colorectal and breast carcinomas, PlGF plasma levels and intratumoral expression have been found to correlate with tumor stage, vascularity, recurrence, metastasis and survival (Chen et al., 2004; Marcellini et al., 2006; Parr et al. 2005; Pompeo et al.; 2009; Wei et al., 2005).

In vitro studies have shown that administration of recombinant PlGF to MM cells triggers the activation of Akt but does not elicit a significant stimulation of cell growth. Conversely, the administration of PlGF-neutralizing antibodies causes a significant reduction of MM cell viability, demonstrating the PlGF acts as a survival factor for MM cells (Albonici et al., 2009).

PlGF binds VEGF-R1 and the co-receptors neuropilin-1 and -2, but, unlike VEGF, it does not bind VEGF-R2. Accordingly, it can act independently of VEGF in cells which primarily express VEGF-R1 (Fischer et al., 2007). Worthy of note, even though VEGF and PlGF both bind VEGF-R1, PlGF was reported to stimulate the phosphorylation of specific VEGF-R1 tyrosine residues and the expression of distinct downstream target genes as compared to VEGF (Autiero et al., 2003). On the other hand, PlGF can also sustain VEGF activity through different mechanisms involving both VEGF-R1 and VEGF-R2. One of these mechanisms relies on the formation of PlGF:VEGF heterodimers. Indeed, PlGF:VEGF heterodimers have been isolated from cells producing both factors and shown to bind VEGF-R1:VEGF-R2 receptor complexes, thus inducing receptor cross-talk and activation of VEGF-R2, the major mediator of VEGF activities (Autiero et al., 2003; Cao et al., 1996). In addition, the activation of VEGF-R1 by PlGF homodimers may induce the intermolecular transphosphorylation and activation of VEGF-R2 (Carmeliet et al., 2001).

It is noteworthy that in vivo anti-PlGF treatment was reported to inhibit tumor growth without affecting healthy vessels, thus reducing tumor infiltration by angiogenic macrophages and severe tumor hypoxia, and preventing the switch on of the angiogenic rescue program leading to the enhanced release different angiogenic factors responsible for resistance to VEGF receptors inhibitors (Fischer et al., 2007).

3.2. PDGF

PDGFs comprise a family of dimeric growth factors structurally and functionally related to VEGFs (Andrae et al., 2008). PDGF homodimers are formed by four different chains, i.e. PDGF-A, PDGF-B, PDGF-C and PDGF-D. In addition, PDGF-A and –B chains can form the heterodimeric PDGF-AB. The biological effects of PDGF are mediated by two tyrosine kinase receptors, namely the PDGF receptor alpha (PDGFRα), which binds PDGF-A, -B, and –C chains, and the PDGF receptor beta (PDGFR), which binds PDGF-B and –D. Accordingly, upon ligand binding different receptor dimers may form depending on ligand configuration and the pattern of receptor expression. Cellular responses to PDGF signaling include stimulation of cell growth, differentiation, migration and inhibition of apoptosis (Andrae et al., 2008).

An increased PDGF activity has been linked with tumors, vascular and fibrotic diseases (Andrae et al., 2008). Autocrine PDGF signaling leading to enhanced proliferation of tumor cells occurs in several types of cancer (Ostman, 2004). In addition, PDGF secretion by cancer cells and activated endothelial cells promotes the formation of both fibrous and vascular tumor stroma. In particular, PDGF-BB participates in tumor angiogenesis by stimulating endothelial cell motility and pericyte recruitment to neoformed vessels, thus leading to vessel stabilization, tumor cell survival and growth. Instead, both PDGF-AA and PDGF-BB appear involved in tumor recruitment of PDGFR-positive fibroblasts which, in turn, can be activated by PDGFs to produce VEGF and other tumor-promoting growth factors (Andrae et al., 2008; Cao et al., 2008; Homsi & Daud, 2007).

Either high PDGF-AB serum levels or a strong expression of PDGFR signaling effectors in MM tissues have been associated with a lower survival in MM patients (Filiberti et al., 2005; Kothmaier et al., 2008). In fact, several evidence support a role for PDGF in MM promotion and progression through both autocrine and paracrine mechanisms.

While PDGFRα expression levels are lower in MM than in normal mesothelial cells, PDGFRβ, PDGF-A and PDGF-B are overexpressed in MM cells as compared to their non-transformed counterparts (Langerak et al., 1996a, 1996b; Metheny-Barlow et al., 2001). Functional studies have shown that transduction of MM cells with a hammerhead ribozyme against PDGFRβ mRNA reduced both PDGFRβ expression and MM cell proliferation, demonstrating the involvement of a PDGF-BB autocrine loop in MM cell growth (Dorai et al., 1994). Conversely, the role of PDGF-A in MM cell proliferation is controversial. Indeed, the transfection of MM cells with antisense oligonucleotides to PDGF-A has been reported to either inhibit or stimulate MM cell growth in vitro (Garlepp & Leong, 1995; Metheny-Barlow et al., 2001). On the other hand, PDGF-A appears to play an important role in sustaining MM cell growth in vivo through paracrine mechanisms. Indeed, PDGF-A overexpression in MM cells inoculated in nude mice was found to increase tumor incidence, tumor growth rate and to decrease the latency period to tumor formation (Metheny-Barlow et al., 2001). In this regard, it has been suggested that PDGF-A participates in a malignant cytokine network through which MM cells instigates tumor-associated fibroblasts to produce growth factors, such as hepatocyte growth factor (HGF), with tumor-promoting activities (Li et al., 2011).

3.3. FGF

The FGF family encompasses 22 structurally related ligands in mammals. The effects of most FGF family members, including FGF-1 and -2, are mediated by binding to a family of tyrosine kinase receptors designated FGF receptors (FGFR1 to FGFR5), whereas a smaller number of FGF isoforms does not bind FGFRs but interacts with voltage-gated sodium channels (Knights & Cook, 2010).

FGFs regulate cell proliferation, differentiation, survival, wound healing and angiogenesis. In cancer, FGF signaling is frequently de-regulated, resulting in mitogenic, anti-apoptotic and angiogenic responses (Knights & Cook, 2010). FGF-1 and -2, but also other less-studied FGF isoforms, exert pro-angiogenic effects by modulating proliferation and migration of endothelial cells and by stimulating the production of proteases (Lieu et al., 2011; Saylor et al., 2012). Worthy of note, it has been demonstrated that FGF-2 can synergize with both VEGF and PDGF-BB in stimulating neovascularization, this synergism relying on multiple mechanisms. For instance, FGF-2 promotes hypoxia-induced VEGF release by cancer cells and the expression of both VEGF and VEGFRs in endothelial cells, whereas VEGF, in turn, upregulates the expression of FGF-2 (Lieu et al., 2011; Saylor et al., 2012). Moreover, FGF-2 upregulates PDGFRs expression and increases the responsiveness to PDGF-BB in endothelial cells, whereas PDGF-BB enhances FGFR1 expression and FGF-2 responsiveness in vascular smooth muscle cells (Cao et al., 2008; Liu et al., 2011). Remarkably, FGFs are thought to play a critical role in the resistance to anti-VEGF therapy (Lieu et al., 2011; Saylor et al, 2005). Besides, both FGF-1 and -2 may also be involved in tumor cell growth through cell-autonomous, autocrine mechanisms (Kumar-Singh et al., 1999).

FGF-1 and -2 are expressed in the majority of MMs in vivo and high levels of FGF-2 in tumor tissues, serum or pleural effusions are associated with a worse prognosis in MM patients (Davidson et al., 2004; Kumar-Singh et al., 1999; Strizzi et al., 2001b). Furthermore, the combined expression levels of FGF-1, FGF-2, VEGF and Transforming Growth Factor beta (TGFβ) in MM tissues correlates with both IMD and a poorer prognosis (Kumar-Singh et al., 1999). In addition to their role in tumor angiogenesis, FGFs act as autocrine growth factors for MM cells. Indeed, MM cells express FGFs and FGF receptors and the transfection with short interfering RNAs to FGF-1 and FGF-2 reduces MM cell proliferation (Kumar-Singh et al., 1999; Liu & Klominek, 2003; Stapelberg et al., 2005). It has also been reported that treatment of MM cells with exogenous FGF-2 stimulates the secretion of matrix metalloproteinases involved in tumor invasion and angiogenesis (Liu & Klominek, 2003).

Advertisement

4. Cross-talk between inflammation and angiogenic growth factors

Experimental and epidemiological evidences indicate that chronic inflammation is associated with most, if not all, tumors and supports their progression (Coussens & Werb 2002; Mantovani et al., 2008; Mantovani et al., 2010; Porta et al., 2009). Chronic inflammation appears to have a versatile function in tumor onset and progression. Indeed, as discussed above, a long-lasting inflammation can contribute to cancer initiation through the production ROS and RNS with DNA-damaging properties. On the other hand, it can also participate in cancer promotion and progression by increasing the availability of mediators (growth factors, cytokines, chemokines, prostaglandins) which contribute to the growth of initiated cells and to neoangiogenesis (Mantovani, 2010). Besides, once a tumor is established, cancer cells promote a constant influx of myelomonocytic cells that express inflammatory mediators supporting pro-tumoral functions. In this regard, myelomonocytic cells are key orchestrators of cancer-related inflammatory processes supporting proliferation and survival of malignant cells, subversion of adaptive immune responses, stromal remodeling and angiogenesis (David Dong et al., 2009; Loges et al., 2009; Porta et al., 2009).

Tissue infiltration by macrophages is a dramatic and common feature of inflammation, angiogenesis and cancer (Pollard, 2004; Sica, 2010). High densities of tumor-infiltrating macrophages are associated with poor survival in patients with MM (Burt et al., 2011). In fact, the recruitment and infiltration of macrophages in the tumor microenvironment can activate them to support the malignant progression of cancer cells. These macrophages are called tumor-associated macrophages (TAMs) (Lawrence, 2011; Sica, 2010). Cancer cells co-cultured with macrophages and incubated with inflammatory cytokines are synergistically stimulated to produce various angiogenesis-related factors (Izzi et al., 2009; Ono, 2008). This inflammatory angiogenesis is mediated, in part, by activation of NF-κB and AP-1 (Angelo & Kurzrock, 2007; Huang et al., 2000; Ono, 2008). In fact, treatment of both vascular endothelial cells and cancer cells with IL-1α/β, TNF-α and ROS in vitro results in a marked induction of VEGF and FGF-2, through the transcriptional activation of NF-κB, Specificity protein 1 (Sp-1), AP-1 and hypoxia response elements.

In addition to macrophages, other tumor-infiltrating immune cells including T cells, B cells, natural killer cells and neutrophils can release cytokines, such as IL-1α/β, TNF-α and IL-6, able to sustain the synthesis of angiogenic growth factors (Angelo & Kurzrock, 2007). As for, IL-6, this pro-inflammatory cytokine has been reported to play a critical role in the stimulation of VEGF synthesis by different cell types, including MM cells (Adachi et al., 2006; Angelo & Kurzrock, 2007). Of note, MMs usually produce high levels of IL-6 but express low levels of IL-6R, so that the presence of sIL-6Rs, which may be provided by inflammatory cells recruited to the tumor region, is essential for the IL-6-dependent stimulation of VEGF expression by MM cells (Adachi et al., 2006). Inflammation can also induce the expression of receptors for angiogenic growth factors. In this regard, the expression of PDGFRs is known to be induced by inflammatory cytokines such as TNF-α and IL-1 (Andrae et al., 2008). Besides, inflammatory cells themselves can directly release angiogenic factors such as VEGF, PlGF, FGF-2 and PDGF, among many others, which exert mitogenic and migratory effects on surrounding cells (Sica 2010, Ono 2008). Inflammatory cells recruited in the tumor microenvironment can also produce matrix metalloproteinases which promote the formation of new vessels by degrading the basement membrane and by releasing angiogenic growth factors, such as VEGF, PlGF-2 and FGF-2, stored in the extracellular matrix (Barillari et al.,1998; Cao et al., 2008; Lieu et al., 2011).

The high amount of chemokines/cytokines, growth factors, proteolytic enzymes, proteoglycans, lipid mediators and prostaglandins which is typically found in the tumor microenvironment sustains and exacerbates both inflammation and angiogenesis (Costa et al., 2007; Lin & Karin, 2007; Ono, 2008). In this context, the cross-talk between inflammation and angiogenesis is further corroborated by the evidence that, if on one hand inflammatory mediators have significant effects on angiogenesis, on the other hand angiogenic factors can effectively promote inflammation. As a matter of fact, in addition to their angiogenic role, VEGF and PlGF appear to act as direct proinflammatory mediators in the pathogenesis of different inflammatory conditions (Angelo & Kurzrock, 2007; Yoo et al., 2008). In this regard, VEGF was found to increase the production of TNF-α and IL-6 by human peripheral blood mononuclear cells and macrophages (Yoo et al., 2008). Moreover, VEGF stimulates monocyte recruitment to tumor areas (Barleon et al., 1996). An additional link between inflammatory and angiogenic growth factors has been provided with the demonstration that in myelomonocytic cells TNF-α is upregulated by PlGF in a NFAT1-dependent manner and, in turn, contributes to PlGF-induced myelomonocytic cell recruitment (Ding et al., 2010). PlGF can also contribute to inflammation by acting as survival factor for monocytes and macrophages (Adini et al., 2002).

Advertisement

5. Cooperation between asbestos and angiogenic growth factors in MM onset and progression

As reported above, asbestos stimulates the expression of c-fos and c-jun mRNA in mesothelial cells in a dose-dependent fashion (Heintz et al., 1993; Ramos-Nino et al., 2002). One of the mechanisms by which VEGF and PlGF elicit biological responses is the induction of Fos-B and c-Fos expression in endothelial cells and monocytes (Holmes & Zachary, 2004). The coexistence of different stimuli, such as asbestos fibers and angiogenic growth factors, concurring to the activation of early response genes might lead to the persistent induction of AP-1 in mesothelial cells and to the chronic stimulation of mesothelial cell proliferation, thus favoring cell transformation.

Further, asbestos and angiogenic growth factors can cooperate in inducing an immunosuppressive tumor microenvironement. Indeed, asbestos has been found to possess immunosuppressive properties. For example, chrysotile fibers have been shown to depress the in vitro proliferation of phytohemagglutinin-stimulated peripheral blood lymphocytes and to suppress natural killer activity. Moreover, asbestos significantly reduces the generation and activity of lymphokine-activated killer (LAK) cells, which are immune effectors with a strong lytic activity against MM cells (Manning et al., 1991; Valle et al., 1998).

Immunosuppressive properties have been reported for angiogenic growth factors as well (Ohm et al., 2001; Ziogas et al., 2012). Impaired antigen-presenting function in DCs as a result of abnormal differentiation is an important mechanism of tumor escape from immune control. It has been demonstrated that VEGF can inhibit the maturation of DCs induced by lipopolysaccharide (Takahashi et al., 2004). VEGF can also affect the ability of hematopoetic progenitor cells (HPCs) to differentiate into functional DCs during the early stages of hematopoiesis in vivo (Gabrilovich et al., 1996; Oyama et al., 1998). In this regard, it has been shown that VEGF binds to specific receptors on the surface of HPCs and this binding appears to involve VEGF-R1. Interestingly, the number of binding sites available for VEGF decreased with DC maturation and correlated with decreased levels of VEGF-R1 mRNA expression in the late-stage cells (Gabrilovich et al., 1996). PlGF was also found to inhibit the activation and maturation of human DCs effectively and rapidly through the NF-κB pathway (Lin et al., 2007). The results of this study further indicate that by modulating the function of DCs, PlGF can down-regulate T helper immune responses (Lin et al., 2007). In addition, both VEGF and PlGF are also involved in the recruitment of macrophages with immunosuppressive, tumor-promoting roles to the tumor stroma.

On the whole, these findings suggest mechanisms by which tumor-derived soluble factors such as VEGF or PlGF may synergize with asbestos to down-regulate immune responses to MM antigens.

Advertisement

6. Conclusions

Collectively, the reported findings demonstrate that a complex network involving asbestos, inflammation and angiogenic factors upregulation is involved in the pathogenesis of MM. In particular, the abnormal expression of angiogenic factors appears to play multiple roles in MM: it stimulates tumor neovascularization, increases pleural effusion formation by increasing vascular permeability, supports autocrine tumor cell growth and finally, in synergism with asbestos fibers, can sustain inflammation and bias host immune responses. Accordingly, the upregulation of angiogenic growth factors appears to be a crucial event in mesothelial cell transformation and MM progression.

Given the involvement of multiple angiogenic growth growth factors in the formation of tumor vessels, in tumor inflammation and MM cell growth and survival, the therapeutic development of antiangiogenic agents for the treatment of this tumor should be aimed at blocking multiple growth factor signaling pathways and their complex interactive network (Cao et al., 2008; Ikuta et al., 2009; Homsi & Daud, 2007; Lieu et al., 2011).

References

  1. 1. AdachiY.AokiC.Yoshio-HoshinoN.TakayamaK.CurielD. T.NishimotoN.Interleukin-6 induces both cell growth and VEGF production in malignant mesotheliomas. Int. J. Cancer, 1196September 2006130313110020-7136
  2. 2. AdiniA.KornagaT.FiroozbakhtF.BenjaminL. E.2002Placenta growth factor is a survival factor for human endothelial cells and macrophages. Cancer Res., 6210May 2002), 274927520008-5472
  3. 3. AlboniciL.DoldoE.PalumboC.OrlandiA.BeiR.PompeoE.MineoT. C.ModestiA.ManzariV.2009Placenta growth factor is a survival factor for human malignant mesotelioma cells. Int. J. Immunopathol. Pharmacol., 222April-June 2009), 3894010394-6320
  4. 4. AndraeJ.GalliniR.BetsholtzC.2008Role of platelet-derived growth factors in physiology and medicine. Genes Dev., 2210May 2008), 127613120890-9369
  5. 5. AngeloL. S.KurzrockR.2007Vascular endothelial growth factor and its relationship to inflammatory mediators. Clin. Cancer Res., 1310May 2007), 282528301078-0432
  6. 6. 0022-17671215172167223Antony, V. B., Godbey, S. W., Kunkel, S. L., Hott, J. W., Hartman, D. L., Burdick, M. D. & Strieter, R. M. (1993). Recruitment of inflammatory cells to the pleural space. Chemotactic cytokines, IL-8, and monocyte chemotactic peptide-1 in human pleural fluids. J. Immunol., Vol. 151, No. 12, (December 1993), pp. 7216-7223, ISSN 0022-1767
  7. 7. AsaharaT.TakahashiT.MasudaH.KalkaC.ChenD.IwaguroH.InaiY.SilverM.IsnerJ. M.1999VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J., 1814July 1999), 396439720261-4189
  8. 8. AstoulP.1999Pleural mesothelioma. Curr. Opin. Pulm. Med., 54July 1999), 2592681070-5287
  9. 9. AttanoosR. L.GibbsA. R.1997Pathology of malignant mesothelioma. Histopathology, 305May 1997), 4034180309-0167
  10. 10. AustA. E.CookP. M.DodsonR. F.2011Morphological and chemical mechanisms of elongated mineral particle toxicities. J. Toxicol. Environ. Health. B Crit. Rev., 141-440751093-7404
  11. 11. AutieroM.WaltenbergerJ.CommuniD.KranzA.MoonsL.LambrechtsD.KrollJ.PlaisanceS.De MolM.BonoF.KlicheS.FellbrichG.Ballmer-HoferK.MaglioneD.Mayr-BeyrleU.DewerchinM.DombrowskiS.StanimirovicD.Van HummelenP.DehioC.HicklinD. J.PersicoG.HerbertJ. M.CommuniD.ShibuyaM.CollenD.ConwayE. M.CarmelietP.2003Role of PlGF in the intra- and intermolecular cross-talk between the VEGF receptors Flt-1 and Flk-1. Nat. Med., 97July 2003), 9369431078-8956
  12. 12. BaldysA.PandeP.MoslehT.ParkS. H.AustA. E.2007Apoptosis induced by crocidolite asbestos in human lung epithelial cells involves inactivation of Akt and MAPK pathways. Apoptosis, 122February 2007), 4334471360-8185
  13. 13. BarillariG.AlboniciL.FranzeseO.ModestiA.LiberatiF.BarillariP.EnsoliB.ManzariV.SanteusanioG.1998The basic residues of placenta growth factor type 2 retrieve sequestered angiogenic factors into soluble form. Implication for tumor angiogenesis. Am. J. Pathol., 1525May 1998), 116111660002-9440
  14. 14. BarleonB.SozzaniS.ZhouD.WeichH. A.MantovaniA.MarméD.1996Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood, 878April 1996), 333633430006-4971
  15. 15. BermanD. W.CrumpK. S.2008A meta-analysis of asbestos-related cancer risk that addresses fiber size and mineral type. Crit. Rev. Toxicol., 38Suppl. 1, 49731040-8444
  16. 16. BéruBé. K. A.QuinlanT. R.FungH.MagaeJ.VacekP.TaatjesD. J.MossmanB. T.1996Apoptosis is observed in mesothelial cells after exposure to crocidolite asbestos. Am. J. Respir. Cell. Mol. Biol., 151July 1996), 1411471044-1549
  17. 17. BocchettaM.Di RestaI.PowersA.FrescoR.TosoliniA.TestaJ. R.PassH. I.RizzoP.CarboneM.Human mesothelial cells are unusually susceptible to simian virus 40-mediated transformation and asbestos cocarcinogenicity. Proc. Natl. Acad. Sci. USA, 9718August 200010214102190027-8424
  18. 18. BowieA.O’NeillL. A.2000Oxidative stress and nuclear factor-kappaB activation: a reassessment of the evidence in the light of recent discoveries. Biochem. Pharmacol., 591January 2000), 13230006-2952
  19. 19. BranchaudR. M.GarantL. J.KaneA. B.1993Pathogenesis of mesothelial reactions to asbestos fibers. Monocyte recruitment and macrophage activation. Pathobiology, 613-41541631015-2008
  20. 20. BroaddusV. C.YangL.ScavoL. M.ErnstJ. D.BoylanA. M.1996Asbestos induces apoptosis of human and rabbit pleural mesothelial cells via reactive oxygen species. J. Clin. Invest., 989November 1996), 205020590021-9738
  21. 21. BrownD.BeswickP.DonaldsonK.1999Induction of nuclear translocation of NF-κB in epithelial cells by respirable mineral fibres. J. Pathol., 1892October 1999), 2582640022-3417
  22. 22. BurtB. M.RodigS. J.tillemanT. R.ElbardissiA. W.BuenoR.SugarbakerD. J.Circulating and tumor-infiltrating myeloid cells predict survival in human pleural mesothelioma. Cancer, 11722November 2011523452440000-8543X
  23. 23. CacciottiP.StrizziL.VianaleG.IaccheriL.LibenerR.PortaC.TognonM.GaudinoG.MuttiL.2002The presence of simian-virus 40 sequences in mesothelioma and mesothelial cells is associated with high levels of vascular growth factor. Am. J. Respir. Cell. Mol. Biol., 262February 2002), 1891931044-1549
  24. 24. CaoY.CaoR.HedlundE. M.2008R Regulation of tumor angiogenesis and metastasis by FGF and PDGF signaling pathways. J Mol. Med. (Berl.), 867July 2008), 7857890946-2716
  25. 25. CaoY.ChenH.ZhouL.ChiangM. K.Anand-ApteB.WeatherbeeJ. A.WangY.FangF.FlanaganJ. G.TsangM. L.1996Heterodimers of placenta growth factor/vascular endothelial growth factor. Endothelial activity, tumor cell expression, and high affinity binding to Flk-1/KDR. J. Biol. Chem., 2716February 1996), 315431620021-9258
  26. 26. CarboneM.KratzkeR. A.TestaJ. R.2002The pathogenesis of mesothelioma. Semin. Oncol., 291February 2002), 2170093-7754
  27. 27. CarboneM.LyB. H.DodsonR. F.PaganoI.MorrisP. T.DoganU. A.GazdarA. F.PassH. I.YangH.2012Malignant mesothelioma: Facts, myths and hypotheses. J. Cell. Physiol., 2271January 2012), 44580021-9541
  28. 28. CarmelietP.2003Angiogenesis in health and disease. Nat. Med., 96June 2003), 6536601078-8956
  29. 29. CarmelietP.MoonsL.LuttunA.VincentiV.CompernolleV.De MolM.WuY.BonoF.DevyL.BeckH.ScholzD.AckerT.Di PalmaT.DewerchinM.NoelA.StalmansI.BarraA.BlacherS.VandendriesscheT.PontenA.ErikssonU.PlateK. H.FoidartJ. M.SchaperW.Charnock-JonesD. S.HicklinD. J.HerbertJ. M.CollenD.PersicoM. G.Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nat. Med., 75May 20015755831078-8956
  30. 30. CatalanoA.LazzariniR.Di NuzzoS.OrciariS.ProcopioA.2009The plexin-A1 receptor activates vascular endothelial growth factor-receptor 2 and nuclear factor-kappaB to mediate survival and anchorage-independent growth of malignant mesothelioma cells. Cancer Res., 694February 2009), 148514930008-5472
  31. 31. CatalanoA.RomanoM.MartinottiS.ProcopioA.2002Enhanced expression of vascular endothelial growth factor (VEGF) plays a critical role in the tumor progression potential induced by simian virus 40 large T antigen. Oncogene, 2118April 2002), 289629000950-9232
  32. 32. ChangL.KarinM.2001Mammalian MAP kinase signalling cascades. Nature, 4106824March 2001), 37400028-0836
  33. 33. ChenC. N.HsiehF. J.ChengY. M.ChengW. F.SuY. N.ChangK. J.LeeP. H.2004The significance of placenta growth factor in angiogenesis and clinical outcome of human gastric cancer. Cancer Lett., 2131September 2004), 73820304-3835
  34. 34. ChengN.ShiX.YeJ.CastranovaV.ChenF.LeonardS. S.VallyathanV.RojanasakulY.1999Role of transcription factor NF-kappaB in asbestos-induced TNFalpha response from macrophages. Exp. Mol. Pathol., 663August 1999), 2012100014-4800
  35. 35. ChoeN.TanakaS.XiaW.HemenwayD. R.RoggliV. L.KaganE.Pleural macrophage recruitment and activation in asbestos-induced pleural injury. Environ. Health Perspect., 105Suppl. 5, (September 1997125712600091-6765
  36. 36. ChoiC. H.SongS. Y.ChoiJ. J.ParkY. A.KangH.KimT. J.LeeJ. W.KimB. G.LeeJ. H.BaeD. S.2008Prognostic significance of VEGF expression in patients with bulky cervical carcinoma undergoing neoadjuvant chemotherapy. BMC Cancer, 8October 2008), 2951471-2407
  37. 37. CostaC.IncioJ.SoaresR.2007Angiogenesis and chronic inflammation: cause or consequence? Angiogenesis, 1031491660969-6970
  38. 38. CoussensL. M.WerbZ.2002Inflammation and cancer. Nature, 4206917December 2002), 8608670028-0836
  39. 39. DavidDong. Z. M.AplinA. C.NicosiaR. F.2009Regulation of angiogenesis by macrophages, dendritic cells, and circulating myelomonocytic cells. Curr. Pharm. Des., 1543653791381-6128
  40. 40. DavidsonB.VintmanL.ZchariaE.BedrossianC.BernerA.NielsenS.IlanN.VlodavskyI.ReichR.2004Heparanase and basic fibroblast growth factor are co-expressed in malignant mesothelioma. Clin. Exp. Metastasis, 2154694760262-0898
  41. 41. DawsonM. R.DudaD. G.FukumuraD.JainR. K.2009VEGFR1-activity-independent metastasis formation. Nature, 4617262September 2009), E4E50028-0836
  42. 42. DingY.HuangY.SongN.GaoX.YuanS.WangX.CaiH.FuY.LuoY.2010NFAT1 mediates placental growth factor-induced myelomonocytic cell recruitment via the induction of TNF-alpha. J. Immunol., 1845March 2010), 259326010022-1767
  43. 43. DongH. Y.BuardA.RenierA.LevyF.Saint-EtienneL.JaurandM. C.1994Role of oxygen derivatives in the cytotoxicity and DNA damage produced by asbestos on rat pleural mesothelial cells in vitro. Carcinogenesis, 156June 1994), 125112550143-3334
  44. 44. DoraiT.KobayashiH.HollandJ. F.OnhumaT.1994Modulation of platelet-derived growth factor-beta mRNA expression and cell growth in a human mesothelioma cell line by a hammerhead ribozyme. Mol. Pharmacol., 463September 1994, 4374440002-6895X
  45. 45. DriscollK.CarterJ.HowardB.HassenbeinD.JanssenY.MossmanB. T.1998Crocidolite activates NF-κB and MIP-2 gene expression in rat alveolar epithelial cells. Role of mitochondrial-derived oxidants. Environ. Health Perspect., 106Suppl. 5, (October 1998), 117111740091-6765
  46. 46. EferlR.WagnerE. F.2003AP1: a double-edged sword in tumorigenesis. Nature Rev. Cancer, 311November 2003), 8598680147-4175X
  47. 47. FennellD. A.RuddR. M.2004Defective core-apoptosis signaling in diffuse malignant pleural mesothelioma: opportunities for effective drug development. Lancet Oncol., 56June 2004), 3543621470-2045
  48. 48. FerraraN.GerberH. P.Le CouterJ.2003The biology of VEGF and its receptors. Nat. Med., 96June 2003), 6696761078-8956
  49. 49. FilibertiR.MarroniP.NeriM.ArdizzoniA.BettaP. G.CafferataM. A.CanessaP. A.PuntoniR.IvaldiG. P.PaganuzziM.2005Serum PDGF-AB in pleural mesotelioma. Tumour Biol., 265September-October 2005), 2212261010-4283
  50. 50. FischerC.JonckxB.MazzoneM.ZacchignaS.LogesS.PattariniL.ChorianopoulosE.LiesenborghsL.KockM.De MolM.AutieroM.WynsS.PlaisanceS.MoonsL.van RooijenN.GiaccaM.StassenJ. M.DewerchinM.CollenD.CarmelietP.Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell, 1313November 20074634750092-8674
  51. 51. FischerC.MazzoneM.JonckxB.CarmelietP.2008FLT1 and its ligands VEGFB and PlGF: drug targets for anti-angiogenic therapy? Nat. Rev. Cancer., 812December 2008), 9429560147-4175X
  52. 52. Fleury-FeithJ.PilatteY.JaurandM. C.2003Cells in the pleural cavity, In: Textbook of pleural diseases, Light, R. W. & Lee, Y. C. G., 1734Arnold Publishers, 978-0-34080-794-1London.
  53. 53. FolkmanJ.2006Angiogenesis. Annu. Rev. Med., 571180066-4219
  54. 54. GabrilovichD. I.ChenH. L.GirgisK. R.CunninghamH. T.MenyG. M.NadafS.KavanaughD.CarboneD. P.1996Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med., 210October 1996), 109611031078-8956
  55. 55. GarleppM. J.LeongC. C.1995Biological and immunological aspects of malignant mesothelioma. Eur. Respir. J., 84April 1995), 6436500903-1936
  56. 56. GaspariniG.HarrisA. L.1995Clinical importance of the determination of tumor angiogenesis in breast carcinoma: much more than a new prognostic tool. J. Clin. Oncol., 133March 1995), 7657820073-2183X
  57. 57. GazdarA. F.CarboneM.2003Molecular pathogenesis of malignant mesothelioma and its relationship to simian virus 40. Clin. Lung Cancer, 53November 2003), 1771811525-7304
  58. 58. GossartS.CambonC.OrfilaC.SéquélasM. H.LepertJ. C.RamiJ.CarrèP.PipyB.1996Reactive oxygen intermediated as regulators of TNF-alpha production in rat lung inflammation induced by silica. J. Immunol., 1564February 1996), 154015480022-1767
  59. 59. GreillierL.AstoulP.2008Mesothelioma and asbestos-related pleural diseases. Respiration, 7611150025-7931
  60. 60. GulumianM.2005An update on the detoxification processes for silica particles and asbestos fibers: successess and limitations. J. Toxicol. Environ. Health. B Crit. Rev., 86November-December 2005), 4534831093-7404
  61. 61. HaegensA.BarrettT. F.GellJ.ShuklaA.MacphersonM.VacekP.PoynterM. E.ButnorK. J.Janssen-HeiningerY. M.SteeleC.MossmanB. T.2007Airway epithelial NF-kappaB activation modulates asbestos-induced inflammation and mucin production in vivo. J. Immunol., 1783February 2007), 180018080022-1767
  62. 62. HarmeyJ. H.Bouchier-HayesD.2002Vascular endothelial growth factor (VEGF), a survival factor for tumour cells: implications for anti-angiogenic therapy. Bioessays, 243March 2002), 2802830265-9247
  63. 63. HattoriK.HeissigB.WuY.DiasS.TejadaR.FerrisB.HicklinD. J.ZhuZ.BohlenP.WitteL.HendrikxJ.HackettN. R.CrystalR. G.MooreM. A.WerbZ.LydenD.RafiiS.2002Placental growth factor reconstitutes hematopoiesis by recruiting VEGFR1(+) stem cells from bone-marrow microenvironment. Nat. Med., 88August 2002), 8418491078-8956
  64. 64. HausmannM. J.RogachevB.WeilerM.ChaimovitzC.DouvdevaniA.2000Accessory role of human peritoneal mesothelial cells in antigen presentation and T-cell growth. Kidney Int., 572February 2000), 4764860085-2538
  65. 65. HaydenM. S.GoshS.2004Signaling to NF-κB. Genes Dev., 1818Spetember 2004), 219522240890-9369
  66. 66. HaydenM. S.GhoshS.2008Shared principles in NF-kappaB signaling. Cell, 1323February 2008), 3443620092-8674
  67. 67. HeintzN. H.JanssenY. M.MossmanB. T.1993Persistent induction of c-fos and c-jun expression by asbestos. Proc. Natl. Acad. Sci. USA, 908April 1993), 329933030027-8424
  68. 68. HeintzN. H.Janssen-HeiningerY. M.MossmanB. T.2010Asbestos, lung cancers, and mesotheliomas: from molecular approaches to targeting tumor survival pathways. Am. J. Respir. Cell Mol. Biol., 422February 2010), 1331391044-1549
  69. 69. HillegassJ. M.ShuklaA.LathropS. A.MacPherson. M. B.BeuschelS. L.ButnorK. J.TestaJ. R.PassH. I.CarboneM.SteeleC.MossmanB. T.2010Inflammation precedes the development of human malignant mesotheliomas in a SCID mouse xenograft model. Ann. N. Y. Acad. Sci., 1203August 2010), 7140077-8923
  70. 70. HirayamaN.TabataC.TabataR.MaedaR.YasumitsuA.YamadaS.KuribayashiK.FukuokaK.NakanoT.2011Pleural effusion VEGF levels as a prognostic factor of malignant pleural mesothelioma. Respir. Med., 1051January 2011), 1371420954-6111
  71. 71. HoffmannA.NatoliG.BaltimoreD.2003Genetic analysis of NF-kappaB/Rel transcription factor defines functional specificities. EMBO J., 2220October 2003), 553055390261-4189
  72. 72. HoffmannA.NatoliG.GhoshG.2006Transcriptional regulation via the NF-kappaB signaling module. Oncogene, 2551October 2006), 670667160950-9232
  73. 73. HoidalJ. R.2001Reactive oxygen species and cell signaling. Am. J. Respir. Cell Mol. Biol., 256December 2001), 6616631044-1549
  74. 74. HolmesD. I.ZacharyI.2004Placental growth factor induces FosB and c-Fos gene expression via Flt-1 receptors. FEBS Lett., 5571-3January 2004), 93980014-5793
  75. 75. HomsiJ.DaudA. I.2007Spectrum of activity and mechanism of action of VEGF/PDGF inhibitors. Cancer Control, 143July 2007), 2852941073-2748
  76. 76. HuangS.RobinsonJ. B.DeguzmanA.BucanaC. D.FidlerI. J.2000Blockade of nuclear factor-kappaB signaling inhibits angiogenesis and tumorigenicity of human ovarian cancer cells by suppressing expression of vascular endothelial growth factor and interleukin 8. Cancer Res., 6019October 2000), 533455390008-5472
  77. 77. HuangS. X.JaurandM. C.KampD. W.WhysnerJ.HeiT. K.2011Role of mutagenicity in asbestos fiber-induced carcinogenicity and other diseases. J. Toxicol. Environ. Health B. Crit. Rev., 141-41792451093-7404
  78. 78. HussainS. P.HofsethL. J.HarrisC. C.2003Radical causes of cancer. Nat. Rev. Cancer., 34April 2003), 2762850147-4175X
  79. 79. IkutaK.YanoS.TrungV. T.HanibuchiM.GotoH.LiQ.WangW.YamadaT.OginoH.KakiuchiS.UeharaH.SekidoY.UenakaT.NishiokaY.SoneS.2009E7080, a multi-tyrosine kinase inhibitor, suppresses the progression of malignant pleural mesothelioma with different proangiogenic cytokine production profiles. Clin. Cancer Res., 1523December 2009), 722972371078-0432
  80. 80. IzziV.ChiurchiùV.D’AquilioF.PalumboC.TresoldiI.ModestiA.BaldiniP. M.2009Differential effects of malignant mesothelioma cells on THP-1 monocytes and macrophages. Int. J. Oncol., 342February 2009), 5435501019-6439
  81. 81. Janssen-HeiningerY. M.MacaraI.MossmanB. T.1999Cooperativity between oxidants and tumor necrosis factor in the activation of nuclear factor (NF)-kappaB: requirement of Ras/mitogen-activated protein kinases in the activation of NF-kappaB by oxidants. Am. J. Respir. Cell. Mol. Biol., 205May 1999), 9429521044-1549
  82. 82. JanssenY. M.BarchowskyA.TreadwellM.DriscollK. E.MossmanB. T.1995Asbestos induces nuclear factor-κB (NF-κB) DNA-binding activity and NF-κB-dependent gene expression in tracheal epithelial cells. Proc. Natl. Acad. Sci. USA, 9218August 1995), 845884620027-8424
  83. 83. JanssenY. M.DriscollK. E.HowardB.QuinlanT. R.TreadwellM.BarchowskyA.MossmanB. T.1997Asbestos causes translocation of p65 protein and increases NF-kappa B DNA binding activity in rat lung epithelial and pleural mesothelial cells. Am. J. Pathol., 1512August 1997), 3894010002-9440
  84. 84. JashA.SahooA.KimG. C.ChaeC. S.HwangJ. S.KimJ. E.ImS. H.2012Nuclear factor of activated T cells 1 (NFAT1) induced permissive chromatin modification facilitates nuclear Factor-κB (NF-κB) mediated interleukin-9 (IL-9) transactivation. J. Biol. Chem., (March 2012), Epub ahead of print, 0108-33511083351X
  85. 85. JochumW.PasseguéE.WagnerE. F.2001AP-1 in mouse development and tumorigenesis. Oncogene, 2019April 2001), 240124120950-9232
  86. 86. JungY. J.IsaacsJ. S.LeeS.TrepelJ.NeckersL.2003IL-1beta-mediated up-regulation of HIF-1alpha via an NF-kappaB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis. FASEB J., 1714November 2003), 211521170892-6638
  87. 87. KampD. W.WeitzmanS. A.1999The molecular basis of asbestos induced lung injury. Thorax, 547July 1999), 6386520040-6376
  88. 88. KanwarJ. R.KamalapuramS. K.KanwarR. K.2011Targeting surviving in cancer: the cell-signalling perspective. Drug Discov. Today, 1611-12June 2011), 4854941359-6446
  89. 89. KerbelR. S.2008Tumor angiogenesis. N. Engl. J. Med., 35819May 2008), 203920490028-4793
  90. 90. KerberM.ReissY.WickersheimA.JugoldM.KiesslingF.HeilM.TchaikovskiV.WaltenbergerJ.ShibuyaM.PlateK. H.MacheinM. R.(20082008Flt-1 signaling in macrophages promotes glioma growth in vivo. Cancer Res., 6818September 2008), 734273510008-5472
  91. 91. KhaliqA.LiX. F.ShamsM.SisiP.AcevedoC. A.WhittleM. J.WeichH.AhmedA.1996Localization of placenta growth factor (PlGF) in human term placenta. Growth Factors, 133-42432500897-7194
  92. 92. KnightsV.CookS. J.2010De-regulated FGF receptors as therapeutic targets in cancer. Pharmacol. Ther., 1251January 2010), 1051170163-7258
  93. 93. KochS.TuguesS.LiX.GualandiL.Claesson-WelshL.2011Signal transduction by vascular endothelial growth factor receptors. Biochem J., 4372July 2011), 1691830000-0264
  94. 94. KothmaierH.QuehenbergerF.HalbewedlI.MorbiniP.DemiragF.ZerenH.CominC. E.MurerB.CagleP. T.AttanoosR.GibbsA. R.Galateau-SalleF.PopperH. H.2008EGFR and PDGFR differentially promote growth in malignant epithelioid mesothelioma of short and long term survivors. Thorax, 634April 2008), 3453510040-6376
  95. 95. KumarS.GuleriaR.SinghV.BhartiA. C.MohanA.DasB. C.2009Efficacy of plasma vascular endothelial growth factor in monitoring first-line chemotherapy in patients with advanced non-small cell lung cancer. BMC Cancer, 9December 2009), 4211471-2407
  96. 96. Kumar-SinghS.VermuelenP. B.WeylerJ.SegersK.WejnB.Van DaeleA.DirixL. Y.Van OosteromA. T.Van MarkE.1997Evalutation of tumor angiogenesis as a prognostic marker in malignant mesothelioma. J. Pathol., 1822June 1997), 2112160022-3417
  97. 97. Kumar-SinghS.WeylerJ.MartinM. J.VermeulenP. B.Van MarckE.1999Angiogenic cytokines in mesothelioma: a study of VEGF, FGF-1 and-2, and TGF beta expression. J. Pathol., 1891September 1999), 72820022-3417
  98. 98. LangerakA. W.van der Linden-vanBeurden. C.A. W., van der Linden-van Beurden, C. A. & Versnel, M. A. (1996aRegulation of differential expression of platelet-derived growth factor alpha- and beta-receptor mRNA in normal and malignant human mesothelial cell lines. Biochim. Biophys. Acta., 13051-2February 1996), 63700006-3002
  99. 99. LangerakA. W.De LaatP. A.Van Der Linden VanBeurden. C.A., Van Der Linden-Van Beurden, C. A., Delahaye, M., Van Der Kwast, T. H., Hoogsteden, H. C., Benner, R. & Versnel, M. A. (1996bExpression of platelet-derived growth factor (PDGF) and PDGF receptors in human malignant mesothelioma in vitro and in vivo. J. Pathol., 1782February 1996), 1511600022-3417
  100. 100. LawrenceT.2011Macrophages and NF-κB in cancer. Curr. Top. Microbiol. Immunol., 3491711840007-0217X
  101. 101. LemaireI.OuelletS.1996Distinctive profile of alveolar macrophage-derived cytokine release induced by fibrogenic and nonfibrogenic mineral dusts. J. Toxicol. Environ. Health., 475April 1996), 4654780098-4108
  102. 102. Le PageC.KoumakpayiI. H.LessardL.Mes-MassonA. M.SaadF.2005EGFR and Her-2 regulate the constitutive activation of NF-kappaB in PC-3 prostate cancer cells. Prostate, 652October 2005), 1301400270-4137
  103. 103. LiJ.HuangB.ShiX.CastranovaV.VallyathanV.HuangC.2002Involvement of hydrogen peroxide in asbestos-induced NFAT activation. Mol. Cell. Biochem, 234-2351-2May-June 2002), pp. 161-168, 0300-8177
  104. 104. LiQ.WangW.YamadaT.MatsumotoK.SakaiK.BandoY.UeharaH.NishiokaY.SoneS.IwakiriS.ItoiK.UtsugiT.YasumotoK.YanoS.2011Pleural mesothelioma instigates tumor-associated fibroblasts to promote progression via a malignant cytokine network. Am. J. Pathol., 1793September 2011), 148314930002-9440
  105. 105. LieuC.HeymachJ.OvermanM.TranH.KopetzS.2011Beyond VEGF: inhibition of the fibroblast growth factor pathway and antiangiogenesis. Clin. Cancer Res., 1719October 2011), 613061391078-0432
  106. 106. LinY. L.LiangY. C.ChiangB. L.2007Placental growth factor down-regulates type 1 T helper immune response by modulating the function of dendritic cells. J. Leukoc. Biol., 826December 2007), 147314800741-5400
  107. 107. LinW. W.KarinM.2007A cytokine-mediated link between inmate immunity, inflammation, and cancer. J. Clin. Invest., 1175May 2007), 117511820021-9738
  108. 108. LiuW.ErnstJ. D.BroaddusV. C.2000Phagocytosis of crocidolite asbestos induces oxidative stress, DNA damage, and apoptosis in mesothelial cells. Am. J. Respir. Cell. Mol. Biol., 233September 2000), 3713781044-1549
  109. 109. LiuZ.KlominekJ.2003Regulation of matrix metalloprotease activity in malignant mesothelioma cell lines by growth factors. Thorax, 583March 2003), 1982030040-6376
  110. 110. LogesS.SchmidtT.CarmelietP.2009Antimyeloangiogenic" therapy for cancer by inhibiting PlGF. Clin. Cancer Res., 1511June 2009), 364836531078-0432
  111. 111. LuttunA.TjwaM.CarmelietP.2002Placenta growth factor (PlGF) and its receptor Flt-1 (VEGFR-1): novel therapeutic targets for angiogenic disorders. Ann. N. Y. Acad. Sci., 979December 2002), 80930077-8923
  112. 112. MacianF.2005NFAT proteins: key regulators of T-cell development and function. Nature Rev. Immunol., 56June 2005), 4724841474-1733
  113. 113. MaciánF.López-RodríguezC.RaoA.2001Partners in transcription: NFAT and AP-1. Oncogene, 2019April 2001), 247624890950-9232
  114. 114. MaglioneD.GuerrieroV.VigliettoG.Delli-BoviP.PersicoM. G.1991Isolation of a human placenta cDNA coding for a protein related to the vascular permeability factor. Proc. Natl. Acad. Sci. USA, 8820October 1991), 926792710027-8424
  115. 115. ManciniM.TokerA.2009NFAT proteins: emerging roles in cancer progression. Nature Rev. Cancer., 911November 2009), 8108200147-4175X
  116. 116. ManningC. B.VallyathanV.MossmanB. T.2002Diseases caused by asbestos: mechanisms of injury and disease development. Int. Immunopharmacol., 22-3February 2002), 1912001567-5769
  117. 117. ManningL. S.DavisM. R.RobinsonB. W.1991Asbestos fibres inhibit the in vitro activity of lymphokine-activated killer (LAK) cells from healthy individuals and patients with malignant mesothelioma. Clin. Exp. Immunol., 831January 1991), 85910009-9104
  118. 118. MantovaniA.2010Molecular pathways linking inflammation and cancer. Curr. Mol. Med., 104June 2010), 3693731566-5240
  119. 119. MantovaniA.AllavenaP.SicaA.BalkwillF.2008Cancer-related inflammation. Nature, 4547203July 2008), 4364440028-0836
  120. 120. MarcelliniM.De LucaN.RiccioniT.CiucciA.OrecchiaA.LacalP. M.RuffiniF.PesceM.CianfaraniF.ZambrunoG.OrlandiA.FaillaM. C.2006Increased melanoma growth and metastasis spreading in mice overexpressing placenta growth factor. Am. J. Pathol., 1692August 2006), 6436540002-9440
  121. 121. MasoodR.CaiJ.ZhengT.SmithD. L.HintonD. R.GillP. S.2001Vascular endothelial growth factor (VEGF) is an autocrine growth factor for VEGF receptor-positive human tumors. Blood, 986September 2001), 190419130006-4971
  122. 122. Metheny-BarlowL. J.FlynnB.van GijsselH. E.MarrogiA.GerwinB. I.2001Paradoxical effects of platelet-derived growth factor-A overexpression in malignant mesothelioma. Antiproliferative effects in vitro and tumorigenic stimulation in vivo. Am. J. Respir. Cell Mol. Biol., 246June 2001), 6947021044-1549
  123. 123. MicheauO.TschoppJ.2003Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell, 1142July 2003), 1811900092-8674
  124. 124. Milde-LangoschK.2005The Fos family of transcription factors and their role in tumourigenesis. Eur. J. Cancer., 4116November 2005), 244924610014-2964
  125. 125. MiuraY.NishimuraY.KatsuyamaH.MaedaM.HayashiH.DongM.HyodohF.TomitaM.MatsuoY.UesakaA.KuribayashiK.NakanoT.KishimotoT.OtsukiT.2006Involvement of IL-10 and Bcl-2 in resistance against an asbestos-induced apoptosis of T cells. Apoptosis, 1110October 2006), 182518351360-8185
  126. 126. MossmanB. T.ChurgA.1998Mechanisms in the pathogenesis of asbestosis and silicosis. Am. J. Respir. Crit. Care Med., 1575May 1998), 166616800107-3449X
  127. 127. MüllerM. R.RaoA.2010NFAT, immunity and cancer: a transcription factor comes of age. Nat. Rev. Immunol., 109September 2010), 6456561474-1733
  128. 128. MutsaersS. E.2002Mesothelial cells: Their structure, function and role in serosal repair. Respirology, 73September 2002), 1711911323-7799
  129. 129. MutsaersS. E.2004The mesothelial cell. Int. J. Biochem. Cell Biol., 361January 2004), 9161357-2725
  130. 130. NymarkP.LindholmP. M.KorpelaM. V.LahtiL.RuosaariS.KaskiS.HollménJ.AnttilaS.KinnulaV. L.KnuutilaS.2007Gene expression profiles in asbestos-exposed epithelial and mesothelial lung cell lines. BMC Genomics, 8March 2007), 621471-2164
  131. 131. OhmJ. E.CarboneD. P.2001VEGF as a mediator of tumor-associated immunodeficiency. Immunol. Res., 232-32632720025-7277X
  132. 132. OhtaY.ShridharV.BrightR. K.KalemkerianG. P.DuW.CarboneM.WatanabeY.PassH. I.1999VEGF and VEGF type C play an important role in angiogenesis and lymphangiogenesis in human malignant mesothelioma tumours. Br. J. Cancer., 811September 1999), 54610007-0920
  133. 133. OlssonA. K.DimbergA.KreugerJ.Claesson-WelshL.2006VEGF receptor signaling- in control of vascular function. Nat. Rev. Mol. Cell. Biol., 75May 2006), 3593711471-0072
  134. 134. OnoM.2008Molecular links between tumor angiogenesis and inflammation: inflammatory stimuli of macrophages and cancer cells as targets for therapeutic strategy. Cancer Sci., 998August 2008), 150115061347-9032
  135. 135. 1359-6101415275286Ostman, A. (2004). PDGF receptors-mediators of autocrine tumor growth and regulators of tumor vasculature and stroma. Cytokine growth Factor Rev., Vol. 15, No. 4, (August 2004), pp. 275-286, ISSN 1359-6101
  136. 136. Oura, H., Bertoncini, J., Velasco, P., Brown, L. F., Carmeliet, P. & Detmar, M. (2003). A critical role of placenta growth factor in the induction of inflammation and edema formation. Blood, Vol. 101, No. 2, (January 2003), pp. 560-567, ISSN 0006-4971
  137. 137. OyamaT.RanS.IshidaT.NadafS.KerrL.CarboneD. P.GabrilovichD. I.1998Vascular endothelial growth factor affects dendritic cell maturation through the inhibition of nuclear factor-kappa B activation in hemopoietic progenitor cells. J Immunol., 1603February 1998), 122412320022-1767
  138. 138. PalumboC.BeiR.ProcopioA.ModestiA.2008Molecular targets and targeted therapies for malignant mesothelioma. Curr. Med. Chem., 1598558670929-8673
  139. 139. ParrC.WatkinsG.BoultonM.CaiJ.JiangW. G.2005Placenta growth factor is over-expressed and has prognostic value in human breast cancer. Eur. J. Cancer, 4118December 2005), 281928270014-2964
  140. 140. PersicoM. G.VincentiV.Di PalmaT.1999Structure, expression and receptor-binding properties of placenta growth factor (PlGF). Curr. Top. Microbiol. Immunol., 23731400007-0217X
  141. 141. PhilipM.RowleyD. A.SchreiberH.2004Inflammation as a tumor promoter in cancer induction. Semin. Cancer Biol., 146December 2004), 4334390104-4579X
  142. 142. PisickE.SalgiaR.2005Molecular biology of malignant mesothelioma: a review. Hematol. Oncol. Clin. North Am., 196December 2005), 99710230889-8588
  143. 143. PlateK. H.BreierG.MillauerB.UllrichA.RisauW.1993Up-regulation of vascular endothelial growth factor and its cognate receptors in a rat glioma model of tumor angiogenesis. Cancer Res., 5323December 1993), 582258270008-5472
  144. 144. PollardJ. W.2004Tumor-educated macrophages promote tumor progression and metastasis. Nat. Rev. Cancer, 41January 2004), 71780000-1474X
  145. 145. PompeoE.AlboniciL.DoldoE.OrlandiA.ManzariV.ModestiA.MineoT. C.2009Placenta growth factor expression has prognostic value in malignant pleural mesothelioma. Ann. Thorac. Surg., 8824264310003-4975
  146. 146. PortaC.LarghiP.RimoldiM.TotaroM. G.AllavenaP.MantovaniA.SicaA.2009Cellular and molecular pathways linking inflammation and cancer. Immunobiology, 2149-107617770171-2985
  147. 147. RafiiS.AvecillaS.ShmelkovS.ShidoK.TejadaR.MooreM. A.HeissigB.HattoriK.2003Angiogenic factors reconstitute hematopoiesis by recruiting stem cells from bone marrow microenvironment. Ann. N. Y. Acad. Sci., 996May 2003), 49600077-8923
  148. 148. Ramos-NinosM.TimblinC.MossmanB. T.2002Mesothelial cell transformation requires increased AP-1 binding activity and ERK-dependent Fra-1 expression. Cancer Res., 6221November 2002), 606560690008-5472
  149. 149. ReuterS.GuptaS. C.ChaturvediM. M.AggarwalB. B.Oxidativestress.inflammationcancerhow.arethey.linked?Free.RadicBiol. Med., 4911December 2010160316160891-5849
  150. 150. RibattiD.NicoB.CrivellatoE.RoccaroA. M.VaccaA.2007The history of the angiogenic switch concept. Leukemia, 211January 2007), 44520887-6924
  151. 151. RigantiC.OrecchiaS.SilvagnoF.PescarmonaG.BettaP. G.GazzanoE.AldieriE.GhigoD.BosiaA.2007Asbestos induces nitric oxide synthesis in mesothelioma cells via Rho signaling inhibition. Am. J. Respir. Cell. Mol. Biol., 366June 2007), 7467561044-1549
  152. 152. RobinsonB. W.LakeR. A.2005Advances in malignant mesothelioma. N. Engl. J. Med., 35315October 2005), 159116030028-4793
  153. 153. RobledoR.MossmanB.1999Cellular and molecular mechanisms of asbestos-induced fibrosis. J. Cell. Physiol., 1802August 1999), 1581660021-9541
  154. 154. RoosW. P.KainaB.2006DNA damage-induced cell death by apoptosis. Trends Mol. Med., 129September 2006), 4404501471-4914
  155. 155. Rose-JohnS.WaetzigG. H.SchellerJ.GrötzingerJ.SeegertD.2007The IL-6/sIL-6R complex as a novel target for therapeutic approaches. Expert Opin. Ther. Targets, 115May 2007), 6136241472-8222
  156. 156. RoskoskiR.Jr 2007Vascular endothelial growth factor (VEGF) signaling in tumor progression. Crit. Rev. Oncol. Hematol., 623June 2007), 1792131040-8428
  157. 157. RoydsJ. A.DowerS. K.QwarnstromE. E.LewisC. E.1998Response of tumour cells to hypoxia: role of p53 and NFkB. Mol. Pathol., 512April 1998), 55611366-8714
  158. 158. SaccaniS.PantanoS.NatoliG.2003Modulation of NF-kappaB activity by Exchange of dimers. Mol. Cell, 116June 2003), 156315741097-2765
  159. 159. SaylorP. J.EscudierB.MichaelsonM. D.2012Importance of Fibroblast Growth Factor Receptor in Neovascularization and Tumor Escape from Antiangiogenic Therapy. Clin. Genitourin. Cancer, (February 2012), Epub ahead of print, 1558-767315587673
  160. 160. ScheidereitC.2006IkappaB kinase complexes: gateways to NF-kappaB activation and transcription. Oncogene, 2551October 2006), 668567050950-9232
  161. 161. ShaulianE.KarinM.2001AP-1 in cell proliferation and survival. Oncogene, 2019April 2001), 239024000950-9232
  162. 162. ShaulianE.KarinM.2002AP-1 as a regulator of cell life and death. Nat. Cell Biol., 45May 2002), E131E1361465-7392
  163. 163. ShawJ. P.UtzP. J.DurandD. B.TooleJ. J.EmmelE. A.CrabtreeG. R.1988Identification of a putative regulator of early T cell activation genes. Science, 2414862July 1988), 2022050036-8075
  164. 164. ShibuyaM.2006Differential roles of vascular endothelial growth factor receptor-1 and receptor-2 in angiogenesis. J. Biochem. Mol. Biol., 395September 2006), 4694781225-8687
  165. 165. SchonthalerH. B.Guinea-ViniegraJ.WagnerE. F.2011Targeting inflammation by modulating the Jun/AP-1 pathway. Ann. Rheum, Dis., 70Suppl. 1, (March 2011), i109i1120003-4967
  166. 166. ShuklaA.GulumianM.HeiT. K.KampD.RahmanQ.MossmanB. T.2003aMultiple roles of oxidants in the pathogenesis of asbestos-induced diseases. Free Radical Biol. Med., 349May 2003), 111711290891-5849
  167. 167. ShuklaA.JungM.SternM.FukagawaN. K.TaatjesD. J.SawyerD.Van Houte,nB.MossmanB. T.2003bAsbestos induces mitochondrial DNA damage and dysfunction linked to the development of apoptosis. Am. J. Physiol. Lung Cell. Mol. Physiol., 2855November 2003), L1018L10251040-0605
  168. 168. SicaA.2010Role of tumour-associated macrophages in cancer-related inflammation. Exp. Oncol., 323September 2010), 1531581812-9269
  169. 169. SimeonovaP.LusterM.1995Iron and reactive oxygen species in the asbestos-induced tumor necrosis factor- response from alveolar macrophages. Am. J. Respir. Cell. Mol. Biol., 126June 1995), 6766831044-1549
  170. 170. SimeonovaP. P.ToriumiW.KommineniC.ErkanM.MunsonA. E.RomW. N.LusterM. I.1997Molecular regulation of IL-6 activation by asbestos in lung epithelial cells: role of reactive oxygen species. J. Immunol., 1598October 1997), 392139280022-1767
  171. 171. StapelbergM.GellertN.SwettenhamE.TomasettiM.WittingP. K.ProcopioA.NeuzilJ.2005Alpha-tocopheryl succinate inhibits malignant mesothelioma by disrupting the fibroblast growth factor autocrine loop: mechanism and the role of oxidative stress. J. Biol. Chem., 28027July 2005), 25369253760021-9258
  172. 172. StrizziL.CatalanoA.VianaleG.OrecchiaS.CasaliniA.TassiG.PuntoniR.MuttiL.ProcopioA.2001aVascular endothelial growth factor is an autocrine growth factor in human malignant mesothelioma. J. Pathol., 1934April 2001), 4684750022-3417
  173. 173. StrizziL.VianaleG.CatalanoA.MuraroR.MuttiL.ProcopioA.2001bBasic fibroblast growth factor in mesothelioma pleural effusions: correlation with patient survival and angiogenesis. Int. J. Oncol., 185May 2001), 109310981019-6439
  174. 174. SunS. C.2011Non-canonical NF-κB signaling pathway. Cell Res., 211January 2011), 71851001-0602
  175. 175. TakahashiA.KonoK.IchiharaF.SugaiH.FujiH.MatsumotoY.(20042004Vascular endothelial growth factor inhibits maturation of dendritic cells induced by lipopolysaccharide, but not by proinflammatory cytokines. Cancer Immunol., Immunother., 536June 2004), 5435500340-7004
  176. 176. TanakaS.ChoeN.HemenwayD. R.ZhuS.MatalonS.KaganE.1998Asbestos inhalation induces reactive nitrogen species and nitrotyrosine formation in the lungs and pleura of the rat. J. Clin. Invest., 1022July 1998), 4454540021-9738
  177. 177. ToiM.MatsumotoT.BandoH.2001Vascular endothelial growth factor: its prognostic, predictive, and therapeutic implications. Lancet Oncol., 211November 2001), 6676731470-2045
  178. 178. ToyokuniS.2009Mechanisms of asbestos-induced carcinogenesis. Nagoya J. Med. Sci., 711-2February 2009), 1100027-7622
  179. 179. ToyookaS.KishimotoT.DateH.2008Advances in the molecular biology of malignant mesothelioma. Acta Med., Okayama, 621February 2008), 170038-6300X
  180. 180. UllrichE.BonmortM.MignotG.KroemerG.ZitvogelL.2008Tumor stress, cell death and the ensuing immune response. Cell Death Differ., 151January 2008), 21281350-9047
  181. 181. ValleM. T.CastagnetoB.ProcopioA.CarboneM.GiordanoA.MuttiL.1998Immunobiology and immune defense mechanisms of mesothelioma cells. Monaldi Arch. Chest Dis., 532April 1998), 2192271122-0643
  182. 182. VillanovaF.ProcopioA.RippoM. R.2008Malignant mesothelioma resistance to apoptosis: recent discoveries and their implication for effective therapeutic strategies. Curr. Med. Chem., 1576316410929-8673
  183. 183. WaltenbergerJ.Claesson-WelshL.SiegbahnA.ShibuyaM.HeldinC. H.1994Different signal transduction properties of KDR and Flt1, two receptors for vascular endothelial growth factor. J. Biol. Chem., 26943October 1994), 26988269950021-9258
  184. 184. WeiS. C.TsaoP. N.YuS. C.ShunC. T.Tsai-WuJ. J.WuC. H.SuY. N.HsiehF. J.WongJ. M.2005Placenta growth factor expression is correlated with survival of patients with colorectal cancer. Gut, 545May 2005), 6666720017-5749
  185. 185. WuW. S.2006The signaling mechanism of ROS in tumor progression. Cancer Metastasis Rev., 254December 2006), 6957050167-7659
  186. 186. YangH.BocchettaM.KroczynskaB.ElmishadA. G.ChenY.LiuZ.BubiciC.MossmanB. T.PassH. I.TestaJ. R.FranzosoG.CarboneM.2006TNF-alpha inhibits asbestos-induced cytotoxicity via a NF-kappaB-dependent pathway, a possible mechanism for asbestos-induced oncogenesis. Proc. Natl. Acad. Sci. USA., 10327July 2006), 10397104020027-8424
  187. 187. YarboroughC. M.2007The risk of mesothelioma from exposure to chrysotile asbestos. Curr. Opin. Pulm. Med., 134July 2007), 3343381070-5287
  188. 188. YasumitsuA.TabataC.TabataR.HirayamaN.MurakamiA.YamadaS.TeradaT.IidaS.TamuraK.FukuokaK.KuribayashiK.NakanoT.2010Clinical significance of serum vascular endothelial growth factor in malignant pleural mesothelioma. J. Thorac. Oncol., 54April 2010), 4794831556-0864
  189. 189. YooS. A.KwokS. K.KimW. U.2008Proinflammatory role of vascular endothelial growth factor in the pathogenesis of rheumatoid arthritid: prospect for therapeutic intervention. Mediators Inflamm., 2008Article ID 129873, 0962-9351
  190. 190. YoshidaK.MikiY.2010The cell death machinery governed by the p53 tumor suppressor in response to DNA damage. Cancer Sci., 1014April 2010), 8318351347-9032
  191. 191. ZebrowskiB. K.YanoS.LiuW.ShaheenR. M.HicklinD. J.PutnamJ. B.EllisL. M.1999Vascular endothelial growth factor levels and induction of permeability in malignant pleural effusions. Clin. Cancer Res., 511November 1999), 336433681078-0432
  192. 192. ZiogasA. C.GavalasN. G.TsiatasM.TsitsilonisO.PolitiE.TerposE.RodolakisA.VlahosG.ThomakosN.HaidopoulosD.AntsaklisA.DimopoulosM. A.BamiasA.2012VEGF directly suppresses activation of T cells from ovarian cancer patients and healthy individuals via VEGF receptor Type 2. Int. J. Cancer., 1304February 2012), 8578640020-7136

Written By

Loredana Albonici, Camilla Palumbo and Vittorio Manzari

Submitted: 16 September 2011 Published: 19 December 2012