Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Oct 1.
Published in final edited form as: Expert Rev Respir Med. 2015 Aug 26;9(5):633–654. doi: 10.1586/17476348.2015.1081066

Latest developments in our understanding of the pathogenesis of mesothelioma and the design of targeted therapies

Angela Bononi 1, Andrea Napolitano 1,2, Harvey I Pass 3, Haining Yang 1, Michele Carbone 1,*
PMCID: PMC4887271  NIHMSID: NIHMS775266  PMID: 26308799

Abstract

Malignant mesothelioma is an aggressive cancer whose pathogenesis is causally linked to occupational exposure to asbestos. Familial clusters of mesotheliomas have been observed in settings of genetic predisposition. Mesothelioma incidence is anticipated to increase worldwide in the next two decades. Novel treatments are needed, as current treatment modalities may improve the quality of life, but have shown modest effects in improving overall survival. Increasing knowledge on the molecular characteristics of mesothelioma has led to the development of novel potential therapeutic strategies, including: (i) molecular targeted approaches, i.e. the inhibiton of vascular endothelial growth factor with Bevacizumab; (ii) immunotherapy with chimeric monoclonal antibody, immunotoxin, antibody drug conjugate, vaccine and viruses; (iii) inhibition of asbestos-induced inflammation, i.e. aspirin inhibition of HMGB1 activity may decrease or delay mesothelioma onset and/or growth. We elaborate on the rationale behind new therapeutic strategies, and summarize available preclinical and clinical results, as well as efforts still ongoing.

Keywords: Mesothelioma, Carcinogenesis, Polyclonal tumors, Asbestos, BAP1, HMGB1, Aspirin, Molecular therapy, Immunotherapy

Introduction

Malignant mesothelioma (MM) is a rare tumor arising from the mesothelial cells lining the pleural and peritoneal cavities, or less commonly from the pericardium and the tunica vaginalis of the testis [1].

In the US and Europe up to 80% of MMs occur in the pleura and are defined as malignant pleural mesothelioma (MPM), since asbestos, after inhalation in the lungs, reaches the pleura via the lymphatic system. The association of MPM with asbestos exposure is well established [2]; further dissemination of asbestos to the peritoneum may occur when exposure is high [3]. The mean age at diagnosis is 74 years old, and since the main source of asbestos exposure is occupational in origin, the male to female sex ratio is 4:1–8:1 (highest in areas where asbestos exposure is high). MPM prognosis remains very poor, with a median survival of 6–12 months, and a 5-year survival <5% [1]. MPM is resistant to chemotherapy, although the combination of pemetrexed and cisplatin led to an overall survival benefit of about 11 weeks [4]; presently this is the most commonly used chemotherapy.

Peritoneal mesotheliomas, instead, are less common, representing about 20% of all MMs in the US, and are rarely associated with asbestos exposure, partially explaining why they are less frequent than MPM [1]. Peritoneal mesotheliomas often occur in a younger age group and the male to female sex ratio is close to 1. All of these findings are expected in MMs that are not related to occupational exposure. In peritoneal mesotheliomas, cytoreductive therapy together with hyperthermic intraperitoneal chemotherapy can significantly improve prognosis [57]. Thus, peritoneal mesotheliomas often present different pathogenesis and clinical challenges than MPM.

In this review, we choose to focus on MPM. The increase in MPM incidence has been associated with widespread use of asbestos in the past century. The latency period of asbestos-associated MPM is on average 30–60 years [8], which accounts, at least in part, for the fact that MPM incidence is still raising worldwide despite working bans on the use of asbestos in the early 1990s [9]. Moreover, many economical emerging countries have not (yet?) prohibited asbestos usage; therefore, in these countries an even higher number of MPM patients is expected in the future [10]. Asbestos refers to a family of six mineral fibers that were used commercially in the ‘70s, that are classified into two subgroups: (i) the amphiboles, a group of rod-like fibers including amosite (brown asbestos), crocidolite (blue asbestos), anthophyllite, actinolite and tremolite; and (ii) the serpentine group, consisting of chrysotile (white asbestos) [11]. The mechanisms responsible for the genesis of MPM as a result of asbestos exposure are being elucidated. What has emerged so far, is that the inhalation of long and thin asbestos fibers induces a chronic inflammatory response at sites of fibers deposition that over time may lead to malignant cell transformation. Three main contributing mechanisms have been proposed. (i) Mesothelial cells and macrophages exposed to asbestos fibers generate reactive oxygen species (ROS) and reactive nitrogen species, which lead to DNA damage [12,13]. (ii) Asbestos fibers absorb a variety of proteins and chemicals, which may result in the accumulation of hazardous molecules including carcinogens [14]. (iii) Asbestos-exposed mesothelial cells and macrophages release a variety of cytokines and growth factors, including high-mobility group box 1 (HMGB1) and tumor-necrosis factor-α (TNF-α) [1517], which induce inflammation and facilitate malignant transformation of mesothelial cells that have accumulated DNA damage [18].

In the US and Europe, exposure to asbestos is the main, but not the exclusive, risk factor connected with MM. The fact that fewer than 5% of asbestos miners who mined asbestos for more than 10 years developed MM [1], indicates that –fortunately- most people exposed to high amounts of asbestos do not develop MM. Additional well-established risk factors for MM include exposure to the naturally occurring asbestos-like mineral fibers, such as erionite, antigorite, etc., chest irradiation, and germline BRCA1-associated protein 1 (BAP1) mutations [1,1924].

In this review we briefly summarize established therapies, and then we focus on the current knowledge of the molecular pathogenesis of MM, and the rationale behind investigating novel targeted approaches. We discuss three main areas: (i) molecular therapies, (ii) immunotherapy, (iii) targeting asbestos-induced inflammation. For each topic we review available results on agents under clinical investigations, and ongoing efforts to improve MPM treatment.

Established therapies

Treating MPM patients remains a challenge. Current approaches are: chemotherapy, and multimodal treatment including surgical resection combined with chemotherapy and/or radiotherapy, photodynamic therapy (PDT), and hyperthermic perfusion of the pleura followed by resection [25]. Residual microscopic disease persist even after the most complete surgical resection, and therefore resection is often associated with a local adjuvant treatment in order to kill residual tumor cells [25]. Pleurectomy is currently the surgery of choice as extrpeural pneumectomy has higher morbidity without showing significant survival advantages [26,27]. Although MM in general is sensitive to radiation, the overall efficacy of radiation therapy is limited because irradiation to a widespread tumor area with a high radiation dose causes severe adverse effects, including pneumonitis, myocarditis, and myelopathy due to spinal cord toxicity. Radiation therapy is therefore mostly used for palliative purposes or in combination with surgery. Among local adjuvant treatments to resection, PDT is a light-based cancer treatment. The effect of PDT requires the interaction of three components: a photosensitizer, oxygen, and light with the specific wavelength activating the photosensitizer. None of these are individually toxic, but when combined they induce a tumoricidal photochemical reaction [28,29]. The first, and so far only, phase randomized III clinical trial conducted by Dr. Pass and colleagues at the NCI, found no differences in survival by randomizing patients to surgery with intraoperative PDT versus surgery without PDT (14.4 versus 14.1 months survival, respectively) [30]. It has been suggested that the technique of intrapleural PDT has improved through the years of practice, leading to a better tolerance of this treatment [28,29]. Two recent studies suggested that PDT offers an improved survival even in patients with locally advanced stages of MPM [3133]. The hypothesis that PDT improves survival in MM patients needs to be confirmed by a new randomized clinical trial.

Surgery in combination with radiation and chemotherapy can be used for healthy patients with early stage disease, but most patients have unresectable disease at the time of diagnosis and are often treated only with palliative chemotherapy [34]. Cisplatin and pemetrexed (an antifolate) combination chemotherapy is the current standard first-line therapy approved by the US Food and Drug Administration (FDA) for patients with advanced and unresectable MPM. Two Phase III trials have demonstrated improved survival of about 11 weeks with an antifolate plus cisplatin over treatment with cisplatin alone [4,35]. However, most MPM patients do not respond to this or other therapies, and even those MPM that responds to therapy, rapidly become resistant to it. MPM patients have therefore a limited life expectancy usually associated to poor quality of life [36]. Second-line therapy is currently not well defined [37].

Our recent finding that MM are polyclonal tumors, i.e., formed by the coalescence of different independent subclones, may account for a high degree of intratumoral heterogeneity and contribute to the emergence of drug-resistant subpopulations [38]. The polyclonal origin of MM [38] highlights the need to attack simultaneously several different molecular targets in order to eliminate the different clones, as each clone may carry its own distinct set of molecular alterations.

Molecular genetics and molecular therapies

Gremline and somatic BAP1 mutations: opening of new frontiers

In 2001 we discovered that, in some Turkish families, an extremely high susceptibility to develop MM was transmitted in an autosomal dominant fashion [19]. Our findings were initially received with some skepticism, as some researchers did not believe that genetics could play a role in MM [39]. Only recently we identified germline mutations in BAP1, a tumor suppressor gene located on chromosome 3p21.3, as the cause of the “BAP1 cancer syndrome” [40]. This cancer syndrome is characterized by the presence of benign atypical melanocytic lesions [41,42], known as melanocytic BAP1-mutated atypical intradermal tumors (MBAITs) [40], a very high incidence of both pleural and peritoneal MMs as well as uveal melanomas (UVMs). Carriers of germline BAP1 mutations also have an elevated risk of developing several other malignancies, such as cutaneous melanoma, clear cell renal cell carcinoma, intrahepatic cholangiocarcinoma, basal cell carcinoma, etc. [24,4346]. All known carriers of germline BAP1 mutations have -so far- developed one or more malignancy by age 55 [47].

In addition to germline mutations, we recently conclusively demonstrated that the majority (63.6%) of sporadic MMs contain somatic BAP1 mutations/inactivation [48]. Our findings confirmed previous data on BAP1 inactivation in epithelioid-type MM [49], and are supported by two recent next generation sequencing (NGS) studies of the MPM genome [50,51]. These two NGS studies revealed that various inactivating mutations occur randomly and are rarely shared among MPM biopsies, with the exception of BAP1 that was found mutated in 41% [50] and 58% [51] of MPMs, respectively (peritoneal MM were not studied), pointing at BAP1 as the putative driver mutation for a significant number of MMs.

It has been suggested that somatic loss of BAP1 is associated to a slightly longer survival [52,53], a finding probably related to the fact that most somatic mutations are detected in epithelial MM that have a better prognosis than biphasic and sarcomatoid MM.

When MM occur in a setting of germline BAP1 mutations, their prognosis is significantly better with survivals of 5–10 or more years [54]. We are investigating how germline BAP1 mutations on one hand promote MM development, while on the other hand they are associated to reduce disease aggressiveness.

BAP1 is a member of the ubiquitin C-terminal hydrolase (UCH) subfamily of deubiquitinating enzymes (DUBs). Among UCH family members, BAP1 is unique because of its long C-terminal tail, which contains two nuclear localization signals [55]. It has been postulated that to function as a tumor suppressor BAP1 must maintain both nuclear localization and deubiquitinating activity [56]. In the nucleus, BAP1 is present in multiprotein complexes and has been functionally implicated in the regulation of various biologic processes including: cell cycle, cellular differentiation, gluconeogenesis, chromatin remodeling, gene transcription, and DNA repair [47]. Currently, BAP1 functions as a tumor suppressor have been ascribed to (i) BAP1 deubiquitination of histone H2A, leading to transcriptional activation of genes that regulate cell growth [57]; (ii) BAP1 functions as a transcriptional coregulator by interaction to host cell factor-1 (HCF1), Ying Yang 1 (YY1), and E2F1, to induce transcription of genes involved in cell cycle regulation [5861]; and (iii) BAP1 contribution to DNA repair [62,63] (FIGURE 1).

Figure 1. Molecular alterations involved in the development of MM and possible strategies for therapeutic intervention.

Figure 1

(a) Key genetic alterations. The BAP1 gene encodes BRCA1 associated protein-1 (BAP1), which plays a role in the regulation of gene transcription, chromatin remodeling and DNA repair, by forming multi-protein complexes with several nuclear proteins, including host cell factor-1 (HCF1), Ying Yang 1 (YY1) and histone H2A. The NF2 gene encodes the protein merlin, which acts as an upstream regulator of the mammalian target of rapamycin (mTOR), focal adhesion kinase (FAK) and Hippo pathways. The merlin-Hippo signaling inactivation leads to constitutive Yes-associated protein (YAP) activation. The CDKN2A/ARF gene encodes p16INK4a and p14ARF, respectively. The p16INK4a protein activates the retinoblastoma protein (pRb) pathway by inhibiting the cyclin-dependent kinase (CDK)-mediate hyperphosphorylation of pRb. The p14ARF protein mediates p53 stabilization by promoting the degradation of the human ortholog of mouse double minute 2 (MDM2). (b) Receptor tyrosine kinases (RTKs) are frequently activated in MM. Inhibition of RTKs or their ligands with specific antibodies or small molecules has been tested as targeted approach in MM. Strategies for therapeutic intervention are summarized in this and in the following figures, accordingly to results obtained in past and ongoing clinical trials, or to possible future directions. The color code defines drugs that have not been tested (blue), drugs that gave positive results and are under further evaluation (green), and drugs that gave negative results (red). For further detail refer to the relative section in the main text.

Given the involvement of BAP1 in chromatin remodeling and its ability to deubiquitinate H2A, it is possible that BAP1 mutations confer an enhanced sensitivity to ‘epigenetic’ modulators. Epigenetic regulation of tumor suppressor genes through chromatin condensation and decondensation has emerged as an important mechanism that leads to tumorigenesis. The balance between the acetylated and deacetylated forms of histone proteins is regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HATs increase acetylation promoting greater chromatin accessibility for gene expression. HDAC inhibitors alter the wrapping of the DNA around histones, modify the access of transcription factors and consequently impact the expression of various genes. The effect of 4 different HDAC inhibitors, valproic acid, trichostatin A, LBH-589, and suberoylanilide hydroxamic acid (vorinostat), was analyzed in primary UVM cells and in UVM cell lines. These compounds were able to reverse the H2A hyperubiquitination caused by BAP1 loss, and induced morphologic differentiation, cell-cycle exit, and a shift to a differentiated, melanocytic gene expression profile in cultured UVM cells. Valproic acid inhibited the growth of UVM tumors in vivo [64]. In vitro data for the role of HDAC inhibition in MM showed increased apoptosis in MM cell lines after treatment with HDAC inhibitors, either alone or in combination with chemotherapy [6570]. Complete suppression in a mouse xenograft model of a human MM line was observed after a combination of valproic acid and chemotherapy [70]. Recently, BAP1 loss was shown to alter sensitivity to HDAC inhibitors in MPM cells through regulating the transcription of HDAC2. However, it was also found that established MM cell lines with low endogenous HDAC2 were resistant to HDAC inhibition [71]. These findings suggested that HDAC inhibitors might be effective in the adjuvant therapy of patients with BAP1 mutated MM. Vorinostat is approved by FDA for the treatment of cutaneous T-cell lymphoma. In a Phase I trial, four (30%) of 13 patients with MPM that received vorinostat had a stabilization of their disease lasting more than 4 months; in addition, two unconfirmed partial responses were observed [72]. However, in a recently completed Phase III trial (VANTAGE 014) including 660 pre-treated advanced MPM patients, vorinostat given as a second-line or third-line therapy did not improve overall survival. Therefore, was not recommended as a therapy in MPM patients [73]. Another small phase II trial (conducted on thirteen patients) with the HDAC inhibitor, belinostat, also produced negative results [74] (FIGURE 1).

The molecular mechanism of BAP1 as tumor suppressor has not been fully elucidated yet, and there is much hope for possible therapies targeting BAP1 functions or capable to restore its activity.

To date, we found that none of the MM patients carrying germline BAP1 mutations were professionally exposed to asbestos. A possible explanation is that germline BAP1 mutations alone can cause MM and many other tumor types. Moreover, BAP1 mutations might influence the asbestos-induced inflammatory response that is linked to asbestos carcinogenesis, thereby increasing the risk of developing MPM after minimal exposure. Indeed, in a recent study we observed that BAP1+/− mice [75] had a significantly higher incidence of MM after exposure to very low doses of asbestos fibers that rarely induced MM in wild-type mice [76]. This was correlated with significant alterations of the inflammatory response. We observed significantly higher levels of pro-tumorigenic alternatively polarized M2 macrophages, lower levels of anti-tumoral classically activated M1 macrophages, and therefore a significantly higher M2/M1 ratio in asbestos-exposed BAP1+/− mice [76], that is associated with a worse prognosis [77]. BAP1+/− mice also showed lower levels of several chemokines and cytokines, in particular monocyte chemoattractant protein-1 (MCP-1), leukemia inhibitory factor (LIF), and keratinocyte-derived chemokine (KC) [76]. Thus, since germline BAP1 mutations lead to an altered immune response following deposition of asbestos in tissues, interfering with this immune response might help prevent or delay MM in individuals carrying BAP1 mutations. We found that BAP1+/− mice exposed to low doses of asbestos developed MM at a similar rate as wild-type mice exposed to ten times higher doses [76]. Therefore these findings support the hypothesis that germline BAP1 heterozygosity increases susceptibility to the carcinogenic effects of low dose of asbestos.

In summary, appropriate genetic counseling and clinical management is important for individuals who carry germline BAP1 mutation. Reducing exposure to even minimal sources of carcinogenic fibers, and preventive therapies that are capable to modulate asbestos-induced inflammation, such as aspirin, might prove helpful (see below).

Loss of the tumor suppressor gene NF2, encoding Merlin

The role of NF2 in MM pathogenesis was initially supported by data showing that asbestos-treated NF2+/− mice exhibit a markedly accelerated MM tumor formation compared to wild-type littermates [78]. Biallelic inactivation of NF2 was observed in all asbestos-induced MMs from NF2+/− mice, and in 50% of MMs from wild-type mice. The NF2 gene located on chromosome 22q12 encodes the tumor suppressor merlin, whose activity is tightly regulated by its phosphorylation status [79]. Merlin can interact with various proteins, and in this way modulates multiple signal transduction cascades, including mTOR, focal adhesion kinase (FAK) and Hippo signaling pathways (FIGURE 1).

Loss of Merlin activates mTOR signaling pathway

Merlin mediates cell proliferation through inhibition of mTOR, in an AKT-independent manner [80]. Loss of merlin causes activation of mTOR signaling in MM cells [81], therefore in merlin-silenced tumors there is an upregulation of mitogenic signaling and increased cell proliferation. As expected, merlin-negative MM cells were more sensitive to the mTOR inhibitor rapamycin, compared to merlin-positive cells [81]. This observation identifies mTOR as a therapeutic target in the large fraction of MMs that carry NF2 mutations, and provided a rationale for studying mTOR inhibitors in MM. Unfortunately, the oral mTOR inhibitor everolimus (RAD001) had limited clinical activity when tested in a phase II trial S0722 (NCT 00770120) as second- and third-line treatment agent in unselected pre-treated MPM patients [82]. Another phase II trial tested everolimus in MPM patients with NF2 loss, as a biomarker to predict sensitivity (NCT 01024946), but also in these patients everolimus showed limited clinical activity. It was concluded that additional studies of single-agent everolimus in advanced MPM were not warranted [82].

An aspect to take into account is that mTOR inhibition alone produces compensatory upregulation of PI3KCA, and thereby allows restoration of the downstream AKT signaling [83]. To address this mechanism of mTOR resistance, GDC-0980, a potent and selective oral dual inhibitor of class I PI3K and mTOR were tested. GDC-0980 demonstrated broad activity in various xenograft cancer models, including MPM [84], but pulmonary toxicity of this class of agents limits their application in a clinical setting. Another dual PI3K/mTOR inhibitor, LY3023414 is being tested in a phase I trial.

However, in spite of these so far disappointing results, the role of PI3K/AKT/mTOR survival pathway upregulation in MM is being further evaluated in clinical trials (NCT01655225, NCT01991938).

Loss of Merlin and FAK inhibitor sensitivity

In normal cells, Merlin negatively regulates FAK, a cytoplasmic protein kinase that integrates signals from growth factor receptors and integrins, to control cell adhesion, migration and invasion [85]. FAK expression and/or activity are up-regulated in a wide range of malignancies [86]. In NF2-null MM cells, stable overexpression of FAK increased their invasiveness, which was decreased significantly when merlin expression was restored [87]. This finding suggested another possible targeted approach based on patient’s selection by merlin status.

The oral FAK inhibitor GSK2256098 is being tested in MM patients alone (NCT01138033) and in combination with MEK inhibitors (NCT01938443). Promising preliminary results for the monotherapy were reported at the 24th EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics, with average time before the disease progressed doubled in patients with inactive merlin (Abstract #610). Another ongoing phase I trial is testing the other FAK inhibitor defactinib (VS-6063) in MM (NCT01870609).

Loss of Merlin and disruption of the Hippo signaling pathway

Mutation or inactivation of the NF2 gene also causes the disruption of the Hippo signaling pathway in MM. The Hippo signaling pathway is a regulator of organ size, development, differentiation, and tissue regeneration, by restricting cell growth, regulating cell division and promoting apoptosis [88]. Merlin is an upstream regulator of the Hippo signaling cascade, which controls the transcriptional co-activator Yes-associated protein (YAP). The merlin-Hippo signaling inactivation leads to constitutive YAP activation [89]. YAP activation in MMs was observed after occasional gene amplification of chromosome 11q22, which is the locus of the YAP gene [90]. YAP activation induces transcription of multiple cancer-promoting genes, including cell cycle promoting genes like cyclin D1, forkhead box M1 and connective tissue growth factor [91].

The direct inhibition of YAP activity is another possible approach for therapeutic intervention in MM. The screening of a Johns Hopkins Drug Library identified three compounds related to porphyrin that could inhibit the transcriptional activity of YAP in vitro [92]. One of these, verteporfin, is in clinical use in photocoagulation therapy for macular degeneration, and it was moderately effective at blocking mouse YAP1-overexpression- or loss of NF2-driven hepatic tumorigenesis. Another possible approach is targeting molecules involved in the NF2/Hippo pathway. Hedgehog signaling has a role in maintaining YAP protein stability, and is activated in mesothelioma. Treatment of mesothelioma xenografts with the hedgehog antagonist HhAntag led to a decrease of the tumor volume accompanied by a decrease in Ki-67 labeling index [93]. These data suggest that a further investigation of these compounds in MM may be warranted.

Deficiency in the CDKN2A /ARF locus

Molecular genetic analysis has revealed several key genetic alterations in MM. Previously, cyclin-dependent kinase inhibitor 2A (CDKN2A)/alternative reading frame (ARF) and neurofibromatosis type 2 (NF2) were considered the most commonly mutated tumor suppressor genes in MM, as they are found mutated in 30% to 50% and 35% to 40% human MM biopsies, respectively [79,94,95].

The CDKN2A/ARF is a tumor suppressor gene located at chromosome 9p21.3 [96]. CDKN2A encodes p16INK4a, whereas ARF encodes p14ARF. p16INK4a maintains the retinoblastoma protein (pRb) in its active hypophosphorylated form, by inhibiting the cyclin-dependent kinase (CDK)-mediate hyperphosphorylation that leads to pRb inactivation; loss of p16INK4a results in inactivation of pRb and, consequently, failure of cell cycle arrest. The p14ARF protein promotes degradation of the human ortholog of mouse double minute 2 (MDM2), leading to stabilization of p53. The homozygous deletion of CDKN2A/ARF leads to loss of function of both p53 and pRb tumor suppressors, with a consequent major impact on the cell cycle (FIGURE 1). Studies in genetically engineered mouse showed that both CDKN2A/ARF gene products suppress asbestos carcinogenicity. ARF inactivation appears to contribute to MM pathogenesis and genomic instability, and the inactivation of both genes cooperates to accelerate asbestos-induced tumorigenesis in mice [97].

Only a limited number of MM biopsies contain TP53 mutations, the tumor suppressor gene that encodes p53. However, since genetic defects in p16INK4a/p14ARF are very common, and lead to loss of function of both p53 and Rb, the defective p53 pathways is a potential target for MM gene therapy [98]. Direct restoration of p16INK4a using gene therapy has also been tested and it has shown some promising activity in preclinical models, but is still far from clinical development [99,100].

Receptor Tyrosine Kinase Inhibitors

Receptor tyrosine kinases (RTKs) constitute a large family of receptors that regulate the cell cycle and are often activated in MM [101]. Activation of these receptors triggers biochemical cascades that lead to the transduction of abnormal cell growth signals. This is a crucial event in cancer initiation and progression, and inhibition of RTKs or of their ligands with specific antibodies or small molecules has been proven to be an effective and safe targeted approach in several malignancies [102].

Mesothelioma secretes pro-angiogenic factors, platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF), both of which are also associated with cell proliferation and pleural effusion. Inhibition of angiogenesis was shown to produce antitumor responses and decrease pleural effusion [103].

High levels of VEGF were found in the pleural effusions of MM patients and were associated with a worse patient survival [104]. Bevacizumab is a humanized monoclonal antibody against VEGF approved for use in several cancers, including metastatic colorectal and renal cancer, and non-small cell lung cancer [105]. Addition of bevacizumab to the standard of care failed to increase survival of MM patients in three independent phase II clinical trials [106108]. However, at the most recent meeting of the American Society of Clinical Oncology, results from a randomized phase III trial (IFCT-GFPC-0701 MAPS) were presented: in this study, when bevacizumab was added to the standard of care (pemetrexed + cisplatin), patients experienced a significant longer median survival (18.82 months vs. 16.07 months, p = 0.0127) (2015 ASCO Meeting, Abstract #7500). These results might translate into addition of bevacizumab as part of the first line treatment for MPM. A specific RTK inhibitor targeting all three isoforms of VEGF receptors (VEGFRs) 1–3, cediranib, also showed only modest activity in unselected MM patients [109,110]. Moreover, other RTK inhibitors targeting VEGFRs and multiple other receptors, such as sunitinib, sorafenib and valatanib, also showed very limited activity in MM [111116].

An autocrine growth stimulatory effect of PDGF via PDGF receptors (PDGFRs) plays a role in the pathogenesis of MM [117]. High serum levels of PDGF were found in MPM patients and associated with poor prognosis [118]. Expression patterns of PDGF alpha- and beta-receptors (PDGFRα and PDGFRβ) differ between normal and malignant mesothelial cell lines. Normal mesothelial cells predominantly express PDGFRα subunit and less PDGFRβ mRNA and protein, whereas most MM cell lines produce PDGFRβ mRNA and protein [119121]. Therefore, two PDGFRβ inhibitors, imatinib and dasatinib, were tested in clinical trials; however, their use did not result in improved survival in MM patients [122125].

Erlotinib and gefitinib, inhibitors of epithelial growth factor receptor (EGFR), also failed in clinical trails, despite high expression of EGFR in MM specimens [126,127]. Negative results were obtained also using erlotinib in combination with bevacizumab after platinum-based chemotherapy [128].

Several fibroblast growth factors (FGFs) and FGF receptors (FGFRs) are expressed in MM tissue and cell lines, and a correlation of FGF2 levels with tumor aggressiveness and reduced patient survival has been observed [129,130]. Down-regulation of FGF2 was shown to suppress proliferation of MM cells but did not influence nonmalignant cells [131]. Moreover, inhibition of FGFR1 impairs MM cell growth and migration in vitro and in vivo, and potentiates the effect of chemotherapeutic drug or ionizing irradiation [130]. Thus, inhibition of FGF signals appears encouraging and may warrant further evaluation of FGFR targeting strategies as potential anti-MM therapies.

Also the hepatocyte growth factor (HGF) and the receptor c-Met are activated in MM. The HGF/c-Met pathway has a crucial role in tumor invasion and metastasis, and it has been demonstrated that inhibition of this pathway suppresses tumor infiltration into neighboring tissues [132]. An inhibitor for the c-Met kinase is under investigation for clinical efficacy. Also preclinical studies to inhibit the HGF/c-Met pathways were conducted for MM [84,133].

It appears to us worthwhile to note that in several of the mentioned clinical trials, a small percentages of patients (usually ~1–5%), did experience partial benefits from the therapy with RTK inhibitors, highlighting the need to identify predictive biomarkers to select likely responders (FIGURE 1).

Arginosuccinate synthetase-1 (ASS1) deficiency: synthetic lethal approaches using the arginine depletor ADI-PEG20

Arginine is an amino acid that modulates a diverse array of metabolic pathways. Reduced expression of arginosuccinate synthetase-1 (ASS1), the rate-limiting enzyme for arginine biosynthesis, is observed in 63% of MPM. ASS1-deficient MPM cells are auxotrophic for arginine (i.e., unable to synthesize it) and consequently susceptible to arginine deprivation, a novel antimetabolite strategy [134].

A randomized phase II study examined the efficacy and safety of the arginine-lowering agent pegylated arginine deiminase (ADI-PEG20) in patients with ASS1-deficient MPM; compared to best supportive care, ADI-PEG20 almost doubled progression-free survival and was generally safe. The main grade 3–4 toxicities were neutropenia, fatigue and anaphylactic reactions (7%) [NCT01279967]. Therefore ADI-PEG20 should be further examined either alone or in combination with selected therapies.

Deregulation of microRNAs

Deregulation of microRNAs (miRNAs) expression is generating strong interest in cancer research [135]. Specific miRNA expression profiles have been found in MM biopsies compared to normal tissue [136]. miRNAs have been proposed as possible diagnostic tools [136], prognostic markers [137], and treatment option for MM [138]. To overcome the difficulties of directly delivering miRNA mimics, minicells composed of achromosomal bacterial cells and targeted by specific antibodies have been developed. EGFR-targeted minicells have been used to restore miR16 and induce growth arrest in mesothelioma xenografts [138]. Minicells can be given safely to patients with advanced cancer [139], and a clinical trial has started in MM (ACTRN12614001248651).

Other molecular alterations

The proteasome inhibitor bortezomib showed significant preclinical activity, mainly via inhibition of NF-κB activity [140]. However, in a recent phase II study, bortezomib failed to show the hypothesized efficacy as a first-line treatment in combination with cisplatin [141]. There are no current registered trials investigating the use of bortezomib in MM. Inhibition of NF-κB activity is also one of the activities of ranpirnase, a ribonuclease enzyme with activity against MM cells [142]. Ranpirnase has been tested in MM patients in combination with doxorubicin (NCT00003034). However, it did not meet statistical significance for the primary endpoint of survival in MM in a confirmatory phase IIIb clinical trial according to the manufacturer’s annual report [143]. Other less-investigated targets for MM therapy include the Ras/MAPK pathway [144,145]; ii) calretinin [146]; iii) Wnt2 [147]; iv) HSP90 (Heat shock protein S90) [148]. More experimental data are required to determine whether these molecules may be realistic therapeutic target.

In summary, so far, molecular therapies have not positively influenced the average survival of MM patients. Therefore, and considering the intertumor and intratumor genetic heterogeneity of MMs, the challenge ahead of us is to identify the subset of patients who will respond to a given type of therapy. Various studies have proposed that future treatment strategies should not be based on monotherapy (i.e., targeting one gene), but should comprise multimodal treatment targeting 2 or more genes/pathways. The problem is that such combined molecular therapies have been shown some promising results in mice but are, so far, quite toxic, and this toxicity precludes their use in clinic [149].

Immunotherapy

Immune checkpoint inhibitors

Avoiding immune destruction has been proposed as one of the new hallmarks of cancer [150]. One way to evade immune destruction is by expression of endogenous immune checkpoints that normally terminate immune responses after antigen activation (FIGURE 2). This state of tumor-induced immunological anergy is associated to up-regulation in tumor-infiltrating T cells of immune checkpoint molecules, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and programmed cell death protein 1 (PD-1) [151]. Alternatively tumors can block immune activation by upregulating PD-1 ligands. Like many others tumors, MM express high levels of the immunosuppressive PD-1 ligand 1 (PD-L1) [152,153]. Monoclonal antibodies against CTLA4 (tremelimumab, ipilimumab), PD-1 (nivolumab, pembrolizumab), and PD-L1 (avelumab, MPDL3280A) can reactivate the immune response against cancer cells, and have shown promising clinical results in melanoma and some other cancer types [151].

Figure 2. Anti-tumor immune response and mechanisms of MM immune escape with possible strategies for immunotherapeutic interventions.

Figure 2

Malignant mesothelioma (MM) cells express mesothelin and other tumor specific antigens. During the anti-tumor immune response (displayed on the left), antigen-presenting cells (APCs) display tumor antigens represented by major histocompatibility complexes (MHCs) to naïve T cells, inducing T cell activation and subsequent immune destruction of cancer cells via T cell receptor (TCR). However, tumors can activate several immune escape (displayed on the right) mechanisms to evade immune destruction. Molecules like transforming growth factor β (TGF-β) can block T cell function. T cell function can be also abrogated by activation of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) on activated T cells. CTLA-4 binding to B7, and the interaction of PD-1 ligand 1 (PD-L1) with PD-1, are crucial to inhibit immune response. MM cells can also block immune activation by upregulating PD-L1. Potential targets and strategies for MM immunotherapy are summarized in the figure. Color code: drugs that have not been tested (blue), drugs that gave positive results and are under further evaluation (green), and drugs that gave negative results (red).

The CTLA4 inhibitor, tremelimumab, at a dose of 15 mg/kg once every 12 weeks, showed clinical activity in 38% of advanced MM patients in a phase II study [154]. An intensified regiment, i.e., 10 mg/kg once every 4 weeks for six doses, followed by 10 mg/kg every 12 weeks, showed a good safety profile, and clinical and immunological activity in patients with advanced MM, with more than 40% of the patients achieving disease control with a median duration of response of almost 11 months [155]. The same intensified schedule is now being investigated in an ongoing randomized, double-blind, placebo-controlled, phase 2b study (NCT01843374). Other clinical trials with PD-1 inhibitor pembrolizumab (NCT02399371) and PD-L1 inhibitor avelumab (NCT01772004) are currently ongoing. Recently, results from the KEYNOTE-028 trial showed that pembrolizumab is well tolerated and improves clinical outcomes for patients with MM; response rate to pembrolizumab was 28%, and another 48% of patients achieved stable disease. Compared with second-line chemotherapy, the response rate achieved with this immune checkpoint inhibitor is considerably higher [156].

Overall, immune checkpoint inhibitors (FIGURE 2) represent one of the most promising class of new molecules for cancer therapy, and results from the trials in MM are eagerly expected.

Anti-transforming growth factor-β

Transforming growth factor β (TGF-β) is a pleiotropic cytokine and a potent growth inhibitor in normal conditions. However, advanced tumors frequently do not respond to TGF-β, and often produce large amounts of this cytokine. In several malignancies, TGF-β promotes remodeling of the microenvironment to support tumor growth, facilitate metastases and attenuates host antitumor immune responses. Elevated TGF-β plasma levels correlate with advanced tumor stage, metastases, and poor survival. GC1008 (fresolimumab), a humanized monoclonal antibody that neutralizes all three human isoforms of TGF-β (FIGURE 2), has been tested in a phase I trial in 13 patients with MPM. No objective response was seen; however, three patients had stable disease at 3 months and five patients developed an enhanced immune response to MM [157].

Mesothelin-targeting agents

Mesothelin is expressed at low levels in most normal tissues, and it is expressed at higher levels by many solid tumors including MM, pancreatic cancer, ovarian cancer, and non-small cell lung cancer [158]. For this reason, mesothelin has been considered a potential therapeutical target in all of these malignancies. The biologic function of mesothelin overexpression in MM is unknown, but preclinical data suggest that it promotes MM cell invasion and matrix metalloproteinase secretion [159]. Other studies supported the hypothesis that mesothelin plays a role in cell adhesion by showing that it is the receptor for cancer antigen-125 (CA-125, also known as mucin 16 or MUC16), and the interaction between mesothelin and CA-125 leads to heterotypic adhesion [160,161].

Mesothelin-targeting agents (FIGURE 2) can be divided into three categories: (i) anticancer antibodies/antibodies-drug conjugates; (ii) mesothelin-targeting vaccines; (iii) mesothelin-targeting recombinant T cells.

Antibodies and antibody-conjugates

Amatuximab (MORAb-009) is a humanized antibody that showed strong pre-clinical activity in combination with chemotherapy against mesothelin-expressing tumors, via inhibition of the interaction of mesothelin with CA-125 [162,163]. In a phase I clinical trial, amatuximab showed an acceptable toxicity profile and promising activity, with 11/24 (46%) patients showing sustained stable disease [164]. A phase II trial of amatuximab in combination with cisplatinum and pemetrexed showed that for treated patients, compared to historical controls, the progression-free survival was not significantly different. However, the median overall survival of 14.8 months with a third of patients alive and 5 continuing to receive amatuximab at the time of analysis was superior to historical controls [165]. A phase III study is planned to further investigate these findings.

Antibodies against specific molecules have been recently engineered to carry cytotoxic drugs specifically to cancer sites [166]. SS1P is an immunotoxin consisting of the variable fragment of amatuximab linked to the cytotoxic bacterial toxin Pseudomonas exotoxin A that showed strong preclinical activity against mesothelin-expressing cancers [167,168]. Two phase I trials of SS1P monotherapy with different treatment schedules showed modest activity and minor responses [169,170]. In another phase I trial, SS1P was co-administered with pemetrexed and cisplatin in chemonaïve patients, and resulted in partial responses in 77% of the patients [171]. Decrease in serum mesothelin, megakaryocyte potentiating factor, and CA-125 correlated with tumor responses [171]. The relatively modest results obtained in these trials were attributed to the formation of neutralizing antibodies against SS1P after the first cycle of treatment. Induction of an immunosuppressive state with pentostatin and cyclophosphamide effectively prevented the formation of anti-SS1P antibodies, allowed the infusion of more cycles of SS1P therapy and resulted in durable partial responses in 30% of the patients [172]. This combination is currently being explored in a phase I/II trial (NCT01362790). To overcome the limitations due to SS1P immunogenicity, a low-immunogenic engineered version (RG7787) has been developed and tested preclinically in mesothelin-expressing malignancies [173,174]. Interestingly, a recent RNA interference whole-genome screen identified components of the endoplasmic reticulum/Golgi as potential targets for chemical intervention that could increase SS1P killing of cancer cells [175], supporting rationally designed combination therapies of immunotoxins and chemical drugs. Similar to SS1P, αMSLN-MMAE is a novel antibody-drug conjugate formed by a humanized anti-mesothelin antibody and microtubule-disrupting agent monomethyl auristatin E, which showed significant preclinical activity [176] that is being translated in a phase I clinical trial.

BAY 94–9343 (ametuman ravtansine) is a novel, potent, antibody-drug conjugate consisting of a human anti-mesothelin antibody conjugated to the maytansinoid tubulin inhibitor DM4 via a disulfide-containing linker [177]. Preclinical evidences showed superiority to standard chemotherapy, and have led to clinical testing in an ongoing phase I trial (NCT01439152).

Also, Bristol-Myers Squibb has developed a mesothelin-targeted antibody drug conjugate (BMS-986148) that will be tested in a phase I trial (NCT02341625).

Mesothelin-targeted immunoactivators represent a novel class of emerging compounds. A mesothelin-targeted interleukin-12 conjugate showed some promising preclinical efficacy, and represented the first reported immunocytokine for mesothelin-positive tumors [178]. Moreover, a mesothelin-targeted Mycobacterium tuberculosis heat shock protein 70 promoted T cells activity against mesothelin-expressing cancers [179].

Mesothelin-targeting vaccines

Stimulating MM patients’ immunity against mesothelin is an additional line of investigation. CRS-207 is a genetically modified Listeria monocytogenes attenuated vaccine expressing mesothelin, which stimulates activation of T cells against mesothelin-expressing cancer cells [180]. An ongoing Phase I trial is evaluating the combination of CRS-207 with chemotherapy in MM patients (NCT01675765). In pancreatic cancer, priming of the immune response with GVAX (granulocyte-macrophage colony-stimulating factor-secreting allogeneic pancreatic tumor cells) followed by CRS-207 treatment resulted in extension of survival, supporting the hypothesis that CRS-207 can synergize with other immune-stimulating approaches with minimal toxicity [181]. Combination of GVAX/CRS-207 with PD-1 inhibitor nivolumab is being tested in pancreatic cancer (NCT02243371) and, if successful, could be studied also in MM.

Other vectors are also being considered besides Listeria. Recently, preclinical evidences of anti-cancer activity in a pancreatic model have been shown for a mesothelin vaccine based on human adenovirus 40 [182]. Further development of mesothelin-targeting vaccines is expected in the coming years.

Mesothelin-targeting recombinant T cells

Redirecting T lymphocyte antigen specificity by gene transfer can provide large numbers of tumor-reactive T lymphocytes for adoptive immunotherapy. After gene transfer, autologous T cells express a chimeric antigen receptor (CAR), which enables the T cell to destroy target cells. CAR T cells are engineered cells expressing artificial receptors used to graft the specificity of monoclonal antibodies onto T cells. Mesothelin-targeted CAR T cells (CARTmeso) were pioneered and improved at the University of Pennsylvania, and have shown very robust pre-clinical activity, both in vitro and in vivo [183187]. CARTmeso can be however reversibly inactivated within the solid tumor microenvironment, and inhibition of the PD-1 pathway could augment human CAR T cell function [188]. Also, the route of administration of CARTmeso might influence the efficacy of therapy [189]. Phase I studies are currently ongoing (NCT02159716, NCT01583686).

Other immunotherapies

Besides immune checkpoint inhibitors and mesothelin-targeting agents, several other approaches are currently being tested in phase I clinical trials after promising pre-clinical results.

Wilms tumor protein 1 (WT1) is transcription factor highly expressed in MM and ovarian cancers that has gained researchers’ attention as a potential immunotherapeutic target. WT1 peptides are immunogenic and induce T-cell responses against MM cell lines [190]. In fact, WT1 peptide vaccines have been developed [190,191] which showed immune responses in patients with MM [192]. Phase I clinical trials have been recently completed or are still ongoing to test the safety of WT1-based vaccines (NCT00398138, NCT01265433, NCT01890980). Similar to mesothelin, CAR T cells against WT1 have been tested for anti-cancer activity [193,194]. A phase I clinical trial is ongoing (NCT02408016).

A novel target for engineered CAR T cells is fibroblast activation protein (FAP), a protein expressed by cancer-associated fibroblast of most solid tumors [195]. In addition, MM cells also express FAP, making FAP even more attractive as a target [196], and preclinical evidences strongly support FAP’s potential role in MM therapy [197]. A phase I clinical trials with FAP-CAR T cells is currently recruiting patients (NCT01722149).

Preclinical eradication of MM was achieved with a PD-1 based DNA vaccine, which simultaneously exploits the immune stimulating functions of the vaccination and those of PD-1 inhibition [198].

Vaccination with MM tumor cell lysate is another strategy being used to induce an antitumoral response. A phase I trial is testing an autologous tumor cell vaccine with an adjuvant (ISCOMATRIX) and celecoxib to augment antigen presentation (NCT01258868). Another phase I trial is evaluating an allogeneic tumor cell vaccine (K526-GM) in combination with cyclophosphamide and celecoxib (NCT01143545).

Other possible targets under investigation are survivin and membrane chondroitin sulphate proteoglycan 4 (CSPG4). Survivin is member of the inhibitory apoptotic proteins (IAPs) family and plays an important role in the control of apoptosis, cell division and cell migration/metastasis. Survivin is an attractive candidate for cancer immunotherapy since it is abundantly expressed in most human cancers, including MM, and it is largely absent in normal adult tissues. A vaccine based on Fowlpox vector expressing survivin was found to successfully trigger an immune response in a MM mouse model [199]. The response resulted in delayed tumor growth and improved survival, indicating that the vaccine could serve as the basis for the development of clinically relevant MM immune-based treatments and/or prevention strategies [199]. Recently, highly immunogenic vaccines against human survivin have been developed and tested preclinically [200], confirming their potential role in human MM therapy.

CSPG4, which has been successfully targeted in melanoma and breast cancer, is highly expressed in MM, but not in normal mesothelium. Treatment with CSPG4 mAb TP41.2 reduced MM cells adhesion, motility, migration, invasiveness and growth in soft agar. It also inhibited MM growth in vivo and significantly increased the survival of xenografted SCID mice, which suggest that CSPG4 mAb-based immunotherapy may represent a novel approach for the treatment of MM [201].

Finally, other immunotherapeutic approaches investigated for MM involved the use of an IL4-toxin conjugate [202], gene therapy, and oncolytic viral therapy. These therapies, whose results have been significant in preclinical models but have not been successfully translated to the clinics, have been extensively reviewed elsewhere [98,203].

Oncolytic viruses

Viruses are strong stimulants to the immune system by the activation of the innate as well as the adaptive responses. Oncolytic viruses can induce host cell death and antigen release. Intrapleural administration of viral vectors is technically feasible, and has several advantages in MPM treatments because MPM develops in a closed cavity and is localized within the cavity in its early stage. Besides direct lysis, oncolytic viruses also stimulate an anti-cancer immune response. Combination of oncolytic virotherapy and immunotherapy is under investigation [204,205].

Significant evidences of preclinical activity have been obtained in MM in particular for measles virus (MV) [206,207]. MV-NIS is an attenuated MV expressing the human thyroidal sodium iodide symporter (NIS), which allows radiologic detection of the virus distribution and combination with radiometabolic therapy with iodine [206]. A phase I trial with MV-NIS is currently ongoing [NCT01503177]. Also adenovirus [208], retrovirus [209], and herpes simplex virus [210] have been examined for the therapeutic effects in preclinical settings, and are progressing to early human experimentation [NCT01721018, NCT01212367].

Targeting asbestos-induced inflammation in both MM treatment and prevention

Inflammation is the hallmark of asbestos deposition in tissue and contributes to asbestos carcinogenesis [211]. Novel strategies that interfere with asbestos- and other carcinogenic mineral fibers- mediated inflammation might prevent or delay the onset of MM in high-risk cohorts, including genetically predisposed individuals, and/or inhibit tumor growth. HMGB1, NOD-like receptor family pyrin domain containing 3 (NALP3) inflammasome, TNF-α, and Interleukin-1β (IL-1β), can all serve as potential targets for inhibiting chronic inflammation induced by asbestos. Early inflammatory reactions triggered by asbestos are NLRP3-dependent, but NLRP3 is not critical in the chronic development of asbestos-induced MM; indeed, after the administration of asbestos, acute IL-1β production and recruitment of immune cells into peritoneal cavity were shown to be significantly decreased in the NLRP3-deficient mice, but NLRP3-deficient mice still displayed a similar incidence of MM and survival times as wild-type mice [212].

Asbestos induces necrotic cell death of mesothelial cells and therefore causes the release of HMGB1 into the extracellular space [15,16]. HMGB1 is a damage-associated molecular pattern (DAMP) and a key mediator of inflammation [213]. In healthy cells, HMGB1 is found primarily in the nucleus, where it stabilizes chromatin and plays multiple roles in DNA transcription, replication, and recombination. During programmed cell necrosis, HMGB1 translocates from the nucleus to the cytosol and is then passively released to extracellular space, where it binds several pro-inflammatory molecules and triggers the inflammatory responses that distinguish this type of cell death from apoptosis. Secreted HMGB1 stimulates receptor for advanced glycation end products (RAGE), Toll-like receptor 2 (TLR2), and TLR4, expressed on neighboring inflammatory cells, mostly macrophages. HMGB1 induces the secretion of TNF-α by macrophages and activation of nuclear factor κB (NF-κB) [214]. Activation of NF-κB promotes cell proliferation and inhibits cell death, leading to enhanced survival of human mesothelial cells that have accumulated DNA alterations following asbestos exposure, thus facilitating their malignant transformation [18] (FIGURE 3). Moreover, we found that most MM cells actively secrete high levels of HMGB1, and that HMGB1 plays a critical role in MM growth and progression. Therefore, targeting HMGB1 may help to prevent or even treat MM. Therapies that seek to block HMGB1 signaling have been investigated; specific molecules that target the activities of HMGB1 are: anti-HMGB1 and anti-RAGE antibodies, HMGB1 antagonist Box A, and ethyl pyruvate that inhibits HMGB1 secretion [214].

Figure 3. Targeting asbestos-induced inflammation to prevent malignant transformation of mesothelial cells.

Figure 3

Asbestos is very cytotoxic to human primary mesothelial (HM) cells and causes DNA damage, as well as extensive necrotic cell death leading to the release of high-mobility group box 1 (HMGB1) into the extra cellular space. Secreted HMGB1 stimulates receptor for advanced glycation end products (RAGE), Toll-like receptor 2 (TLR2), and TLR4, expressed on neighboring inflammatory cells, mostly macrophages. HMGB1 release elicits macrophage accumulation and triggers the inflammatory response, with secretion of Interleukin-1β (IL-1β) and especially tumor-necrosis factor-α (TNF-α). HMGB1 and TNF-α cooperate to promote the activation of nuclear factor κB (NF-κB) pathway, which increases HM survival after asbestos exposure. This allows HM with asbestos-induced DNA damage to divide rather than die and, if key genetic alterations accumulate, to eventually develop into malignant mesothelioma (MM). HMGB1 is also highly expressed and secreted by MM cells, establishing an autocrine circuit that further influences their proliferation and survival. Potential targets and strategies for MM therapy that have been proposed based on recent studies are summarized in the figure. Color code: drugs that have not been tested (blue), drugs that gave positive results and are under further evaluation (green), and drugs that gave negative results (red).

Since chronic inflammation has been associated with an increased risk of developing numerous types of cancers, many anti-inflammatory agents have been studied. For example, celecoxib, a nonsteroidal anti-inflammatory drug targeting COX-2, was shown to have antitumor properties in MM and it was able to inhibit the in vitro and in vivo tumorigenic potential of MM cells [215]. Aspirin is a widely used anti-inflammatory drug. It has been found that daily treatment with aspirin for ≥5 years can reduce tumor burden in colon cancer and other common malignancies [216]. We recently discovered that aspirin and its metabolite salicylic acid inhibit HMGB1 and reduce MM cell migration and growth in vitro and in vivo, providing a novel rationale to the known anti-cancer effects of aspirin [217]. Also the HMGB1 specific inhibitor BoxA [218], markedly reduced MM growth in xenograft mice and significantly improved survival of treated animals, in a way not additive to aspirin, consistent with our hypothesis that both act via inhibition of HMGB1 activity [217]. Our data [217] suggest that aspirin treatment, and treatment with specific anti-HMGB1 antagonists, might be beneficial to MM patients. Although to the best of our knowledge the possible therapeutic effects of aspirin in MM have not been studied, we found some evidence supporting a role of aspirin in preventing or possibly delaying the growth, and thus the diagnosis, of MM. Specifically, the Physician’s Health Study suggests a possible association between aspirin use and reduced MM incidence (Anonymous (1989) Final report on the aspirin component of the ongoing Physicians’ Health Study. Steering Committee of the Physicians’ Health Study Research Group. The New England journal of medicine 321(3):129–135.). In this study, 22,067 physicians were followed for 24 years with 17 reported cases of MM. An intention to treat analysis comparing aspirin (325 mg on alternate days) and placebo treatment revealed a relative risk of 0.7 (0.3–1.9, 95% confidence interval), indicating a 30% reduction in MM risk. Although the small number of MM cases resulted in insufficient power for a statistically significant analysis, these research findings together with our recent data suggest that aspirin may have potential as an effective drug for MM.

Finally, specific FDA-approved reagents that inhibit molecules involved in asbestos-induced inflammation are available: anakinra, an IL-1β receptor antagonist; infliximab, a chimeric human-mouse anti-TNF-α; etanercept, a soluble TNF receptor fusion protein; glyburide, that inhibits the NALP3 inflammasome [219]. All these molecules should be further investigated for MM treatment (FIGURE 3).

Moreover, preliminary evidence from our laboratory suggests that serum levels of HMGB1 might increase following asbestos exposure and MM [16,220,221]. Therefore, in addition to the therapeutic potential of abrogating HMGB1 functions [217], HMGB1 is a possible biomarker for asbestos exposure and early detection of MM [15,16]. Since early detection of MM improves overall patient survival, the availability of biomarkers for early detection of MM would greatly facilitate the follow up of individuals at high-risk for developing MM. Several others MM biomarkers have been investigated, including mesothelin [222,223], osteopontin [224] and fibulin-3 [225]. Availability of reliable biomarkers is extremely important in MM, because given the long latency between initial exposure to mineral fibers and the development of MM [1], there is a large window of time that provide the opportunity not only for early detection, but also for prevention of disease.

Some chemopreventative approaches have been tested in the past to reduce MM incidence. However, most of these studies were conducted when the molecular pathogenesis of MM was mostly unknown, and were based on the efficacy of chemopreventive agents in reducing incidence and mortality of other malignancies, particularly colorectal cancer. Antioxidant vitamins (e.g., isoforms of vitamin A and E) showed contradictory results in reducing MM incidence, with early reports of efficacy [226] not confirmed after a longer follow-up [227]. Recently, it has been reported that antioxidant vitamins do not reduce MM incidence in a murine model of asbestos-induced MM [228]. More recently, prevention of MM with anti-inflammatory drugs (NSAIDS and COX-2 inhibitors) has been also proposed, however evidences from both animal models and a human cohort do not support this hypothesis [229]. Similarly, statins, which like some NSAIDS have been shown to reduce the chances of developing colorectal cancer, do not alter the incidence of MM in asbestos-exposed mice or humans [230]. Other experimental approaches involving iron reduction also showed limited efficacy in animal models [231]. So far, chemoprevention of MM has been as desirable as elusive. With increased understanding of MM pathogenesis, targeted strategies aimed to prevent MM are being developed. For example, we are currently testing the hypothesis that HMGB1 inhibition with aspirin might reduce or delay the onset of MM.

Expert commentary

Despite progress in several other solid tumor types, MM is still associated with a very poor overall survival, and limited treatment options with no targeted therapies.

So far, all attempts to treat MM using gene therapy have failed. The polyclonal origin of MM [38] and the lack of driver mutations, with the possible exception of BAP1 [48,50,51], may be largely responsible for this failure. Accordingly, a small subset of patients (~5%) enrolled in clinical trials responded to genetic therapy; these might be the same patients in which most tumor cells harbored the targeted gene alteration.

Combination therapies are often proposed as the way to address the failure of current genetic therapies to treat cancer patients. However, combination therapies have caused serious toxic effects, and their toxicity limited clinical use [149]. An apparent exception was presented this year at the ASCO meeting: when bevacizumab was added to the standard of care (pemetrexed + cisplatin), patients experienced a significant longer median survival (18.82 months vs. 16.07 months, p = 0.0127). These results might translate into addition of bevacizumab as part of the first line treatment for MPM.

BAP1 is the most promising driver gene in MM pathogenesis, because more than 60% of MM biopsies contain BAP1 mutations/inactivation [48,49] and germline BAP1 mutations cause a cancer syndrome with high incidence of MM [24,40]. Future studies will address whether targeting BAP1 will represent the possible exception to the failure, to this day, of genetic therapy in MM treatment. The study of pathways directly or indirectly affected by the BAP1 loss of function might be a promising strategy to develop targeted therapies in a large set of BAP1 mutant MM patients.

Given the disappointing results obtained so far in molecular therapy in MM, an increasing number of investigators are shifting their efforts to immunotherapies. Immune based approaches can target the inflammatory response caused by asbestos deposits that over years leads to MM, or might target MM tumor cells or the tumor cell stroma.

Finally, the lack of chemoprevention trials for individuals at risk of MM because of exposure to carcinogenic mineral fibers, and/or BAP1 germline mutations, highlights the urgent need for research in this field: we will test the hypothesis that aspirin by targeting HMGB1 and influencing the chronic inflammatory process that drives mineral fiber carcinogenesis might, similarly to what observed in colon cancer, reduce or delay the onset of MM.

Five-year view

MM incidence is still increasing worldwide and it is not expected to decrease in the next two decades. MPM is commonly associated with asbestos deposition in tissues, which causes a chronic inflammatory reaction and contributes to carcinogenesis. Because the latency period from initial asbestos exposure to disease progression is decades long, therapies aimed at decreasing chronic inflammation, such as aspirin intake [232], could prevent or delay carcinogenesis in exposed individuals and lead to a substantial decrease in MM mortality.

Also genetic testing for BAP1 mutations in exposed cohorts will help identify genetically susceptible individuals who have the highest risk of developing MM.

Key issues.

  • MM is a very aggressive tumor that arises from mesothelial cells of the pleural, pericardial, and peritoneal cavities. Patients are usually diagnosed at advanced stages and the prognosis is very poor, with a median survival of 6–12 months and a 5-year survival of <5% [211].

  • MM incidence it is still increasing worldwide and it is not expected to fall off in the next two decades [1,10].

  • Most MPM patients have unresectable disease and are treated with palliative combination chemotherapy (cisplatin and pemetrexed is the first-line treatment approved by the FDA). However, MPM is largely unresponsive to conventional therapy and the minority of patients that initially respond to therapy eventually become resistant [36].

  • MPM is often associated with asbestos exposure. Asbestos carcinogenesis is linked to chronic inflammation that may lead to malignant mesothelial cell transformation after decades long latency [15,18,211].

  • Novel strategies that interfere with asbestos-mediated inflammation might prevent or delay the onset of MM. HMGB1 is a candidate target for anti-inflammatory therapy, and a potential marker of exposure to carcinogenic mineral fibers [16]. Aspirin is an FDA-approved candidate drug that targets HMGB1 and this drug taken in daily amounts of 85–375 mg has conclusively shown the ability to decrease the incidence of colon cancer and of other inflammation-related malignancies. Similarly, individuals exposed to asbestos and at risk of developing MM may benefit from daily doses of aspirin [232]. Also reactivation of the immune system against cancer cells via immune checkpoint inhibitors has shown significant clinical results.

  • People have different genetic susceptibilities to asbestos and to MM development. Germline BAP1 mutations are present in families with a high incidence of MM [24]. BAP1 somatic mutations have also been found in >60% of sporadic MM, pointing at BAP1 as the most commonly mutated gene in this malignancy [48]. Genetic testing for BAP1 mutations should help in the identification of genetically susceptible individuals who have the highest risk of developing MM.

  • Germline BAP1 mutations predispose to the tumorigenic effect of asbestos, possibly because of a deregulated inflammatory response. Care of individuals carrying germline BAP1 mutations should consider reducing exposure of to even minimal sources of carcinogenic fibers and a preventive therapy to target the inflammatory response [233].

  • The polyclonal origin of MM complicates attempts to develop molecular therapies [38]. Future treatment strategies should not be based on monotherapy, but should comprise multisite and multimodal treatment, if such treatment can be developed without the present toxic effects.

Acknowledgments

This work was supported by NCI 1R01 CA198138-01, NCI R01 CA106567, NCI P01 CA114047 and NCI P30 CA071789 received by M. Carbone, by NCI R01 CA160715-0A, DOD CA120355, and The Riviera United 4-a Cure received by H.Y, by the V-Foundation to M .Carbone and H. Yang and by the University of Hawai’i Foundation, which received donations to support mesothelioma research from Honeywell International Inc., to M. Carbone.. The University of Hawaii has filed for patents on HMGB1 and mesothelioma, on which M. Carbone, H. Yang and H.I Pass are the inventors. M. Carbone has pending patent applications on BAP1 and provides consultation for mesothelioma expertise and diagnosis.

Footnotes

Financial and competing interests disclosure

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

References

* = of interest

** = of considerable interest

  • 1.Carbone M, Ly BH, Dodson RF, et al. Malignant mesothelioma: facts, myths, and hypotheses. Journal of cellular physiology. 2012;227(1):44–58. doi: 10.1002/jcp.22724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wagner JC, Sleggs CA, Marchand P. Diffuse pleural mesothelioma and asbestos exposure in the North Western Cape Province. Br J Ind Med. 1960;17:260–271. doi: 10.1136/oem.17.4.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mensi C, Bonzini M, Macchione M, Sieno C, Riboldi L, Pesatori AC. Differences among peritoneal and pleural mesothelioma: data from the Lombardy Region Mesothelioma Register (Italy) Med Lav. 2011;102(5):409–416. [PubMed] [Google Scholar]
  • 4.Vogelzang NJ, Rusthoven JJ, Symanowski J, et al. Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. J Clin Oncol. 2003;21(14):2636–2644. doi: 10.1200/JCO.2003.11.136. [DOI] [PubMed] [Google Scholar]
  • 5.Lee M, Alexander HR, Burke A. Diffuse mesothelioma of the peritoneum: a pathological study of 64 tumours treated with cytoreductive therapy. Pathology. 2013;45(5):464–473. doi: 10.1097/PAT.0b013e3283631cce. [DOI] [PubMed] [Google Scholar]
  • 6.Liu S, Staats P, Lee M, Alexander HR, Burke AP. Diffuse mesothelioma of the peritoneum: correlation between histological and clinical parameters and survival in 73 patients. Pathology. 2014;46(7):604–609. doi: 10.1097/PAT.0000000000000181. [DOI] [PubMed] [Google Scholar]
  • 7.Ihemelandu C, Bijelic L, Sugarbaker PH. Iterative cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for recurrent or progressive diffuse malignant peritoneal mesothelioma: clinicopathologic characteristics and survival outcome. Ann Surg Oncol. 2015;22(5):1680–1685. doi: 10.1245/s10434-014-3977-y. [DOI] [PubMed] [Google Scholar]
  • 8.Lanphear BP, Buncher CR. Latent period for malignant mesothelioma of occupational origin. J Occup Med. 1992;34(7):718–721. [PubMed] [Google Scholar]
  • 9.Linton A, Vardy J, Clarke S, van Zandwijk N. The ticking time-bomb of asbestos: its insidious role in the development of malignant mesothelioma. Crit Rev Oncol Hematol. 2012;84(2):200–212. doi: 10.1016/j.critrevonc.2012.03.001. [DOI] [PubMed] [Google Scholar]
  • 10.Burki T. Health experts concerned over India’s asbestos industry. Lancet. 2010;375(9715):626–627. doi: 10.1016/s0140-6736(10)60251-6. [DOI] [PubMed] [Google Scholar]
  • 11.Baumann F, Ambrosi JP, Carbone M. Asbestos is not just asbestos: an unrecognised health hazard. Lancet Oncol. 2013;14(7):576–578. doi: 10.1016/S1470-2045(13)70257-2. [DOI] [PubMed] [Google Scholar]
  • 12.Choe N, Tanaka S, Kagan E. Asbestos fibers and interleukin-1 upregulate the formation of reactive nitrogen species in rat pleural mesothelial cells. Am J Respir Cell Mol Biol. 1998;19(2):226–236. doi: 10.1165/ajrcmb.19.2.3111. [DOI] [PubMed] [Google Scholar]
  • 13.Huang SX, Jaurand MC, Kamp DW, Whysner J, Hei TK. Role of mutagenicity in asbestos fiber-induced carcinogenicity and other diseases. J Toxicol Environ Health B Crit Rev. 2011;14(1–4):179–245. doi: 10.1080/10937404.2011.556051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sekido Y. Molecular pathogenesis of malignant mesothelioma. Carcinogenesis. 2013;34(7):1413–1419. doi: 10.1093/carcin/bgt166. [DOI] [PubMed] [Google Scholar]
  • 15.Yang H, Rivera Z, Jube S, et al. Programmed necrosis induced by asbestos in human mesothelial cells causes high-mobility group box 1 protein release and resultant inflammation. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(28):12611–12616. doi: 10.1073/pnas.1006542107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Jube S, Rivera ZS, Bianchi ME, et al. Cancer cell secretion of the DAMP protein HMGB1 supports progression in malignant mesothelioma. Cancer Res. 2012;72(13):3290–3301. doi: 10.1158/0008-5472.CAN-11-3481. ** High-mobility group box 1 (HMGB1) inhibition in vivo reduces the growth of malignant mesothelioma xenografts and extends survival.
  • 17.Qi F, Okimoto G, Jube S, et al. Continuous exposure to chrysotile asbestos can cause transformation of human mesothelial cells via HMGB1 and TNF-alpha signaling. The American journal of pathology. 2013;183(5):1654–1666. doi: 10.1016/j.ajpath.2013.07.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yang H, Bocchetta M, Kroczynska B, et al. TNF-alpha inhibits asbestos-induced cytotoxicity via a NF-kappaB-dependent pathway, a possible mechanism for asbestos-induced oncogenesis. Proc Natl Acad Sci U S A. 2006;103(27):10397–10402. doi: 10.1073/pnas.0604008103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Roushdy-Hammady I, Siegel J, Emri S, Testa JR, Carbone M. Genetic-susceptibility factor and malignant mesothelioma in the Cappadocian region of Turkey. Lancet. 2001;357(9254):444–445. doi: 10.1016/S0140-6736(00)04013-7. [DOI] [PubMed] [Google Scholar]
  • 20.Dogan AU, Baris YI, Dogan M, et al. Genetic predisposition to fiber carcinogenesis causes a mesothelioma epidemic in Turkey. Cancer research. 2006;66(10):5063–5068. doi: 10.1158/0008-5472.CAN-05-4642. [DOI] [PubMed] [Google Scholar]
  • 21.Baumann F, Maurizot P, Mangeas M, Ambrosi JP, Douwes J, Robineau B. Pleural mesothelioma in New Caledonia: associations with environmental risk factors. Environ Health Perspect. 2011;119(5):695–700. doi: 10.1289/ehp.1002862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Carbone M, Baris YI, Bertino P, et al. Erionite exposure in North Dakota and Turkish villages with mesothelioma. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(33):13618–13623. doi: 10.1073/pnas.1105887108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Baumann F, Buck BJ, Metcalf RV, McLaurin BT, Merkler DJ, Carbone M. The Presence of Asbestos in the Natural Environment is Likely Related to Mesothelioma in Young Individuals and Women from Southern Nevada. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2015;10(5):731–737. doi: 10.1097/JTO.0000000000000506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Testa JR, Cheung M, Pei J, et al. Germline BAP1 mutations predispose to malignant mesothelioma. Nature genetics. 2011;43(10):1022–1025. doi: 10.1038/ng.912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zervos MD, Bizekis C, Pass HI. Malignant mesothelioma 2008. Curr Opin Pulm Med. 2008;14(4):303–309. doi: 10.1097/MCP.0b013e328302851d. [DOI] [PubMed] [Google Scholar]
  • 26.Flores RM, Pass HI, Seshan VE, et al. Extrapleural pneumonectomy versus pleurectomy/decortication in the surgical management of malignant pleural mesothelioma: results in 663 patients. J Thorac Cardiovasc Surg. 2008;135(3):620–626. doi: 10.1016/j.jtcvs.2007.10.054. 626 e621–623. [DOI] [PubMed] [Google Scholar]
  • 27.Flores RM. Surgical options in malignant pleural mesothelioma: extrapleural pneumonectomy or pleurectomy/decortication. Semin Thorac Cardiovasc Surg. 2009;21(2):149–153. doi: 10.1053/j.semtcvs.2009.06.008. [DOI] [PubMed] [Google Scholar]
  • 28.Friedberg JS. Radical pleurectomy and photodynamic therapy for malignant pleural mesothelioma. Ann Cardiothorac Surg. 2012;1(4):472–480. doi: 10.3978/j.issn.2225-319X.2012.11.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhu TC, Kim MM, Jacques SL, et al. Real-time treatment light dose guidance of Pleural PDT: an update. Proc SPIE Int Soc Opt Eng. 2015:9308. doi: 10.1117/12.2080110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pass HI, Temeck BK, Kranda K, et al. Phase III randomized trial of surgery with or without intraoperative photodynamic therapy and postoperative immunochemotherapy for malignant pleural mesothelioma. Ann Surg Oncol. 1997;4(8):628–633. doi: 10.1007/BF02303746. [DOI] [PubMed] [Google Scholar]
  • 31.Friedberg JS, Mick R, Culligan M, et al. Photodynamic therapy and the evolution of a lung-sparing surgical treatment for mesothelioma. Ann Thorac Surg. 2011;91(6):1738–1745. doi: 10.1016/j.athoracsur.2011.02.062. [DOI] [PubMed] [Google Scholar]
  • 32.Friedberg JS, Culligan MJ, Mick R, et al. Radical pleurectomy and intraoperative photodynamic therapy for malignant pleural mesothelioma. Ann Thorac Surg. 2012;93(5):1658–1665. doi: 10.1016/j.athoracsur.2012.02.009. discussion 1665–1657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Munck C, Mordon SR, Scherpereel A, Porte H, Dhalluin X, Betrouni N. Intrapleural Photodynamic Therapy for Mesothelioma: What Place and Which Future? Ann Thorac Surg. 2015;99(6):2237–2245. doi: 10.1016/j.athoracsur.2014.12.077. [DOI] [PubMed] [Google Scholar]
  • 34.Flores RM. Induction chemotherapy, extrapleural pneumonectomy, and radiotherapy in the treatment of malignant pleural mesothelioma: the Memorial Sloan-Kettering experience. Lung Cancer. 2005;49(Suppl 1):S71–S74. doi: 10.1016/j.lungcan.2005.03.015. [DOI] [PubMed] [Google Scholar]
  • 35.van Meerbeeck JP, Gaafar R, Manegold C, et al. Randomized phase III study of cisplatin with or without raltitrexed in patients with malignant pleural mesothelioma: an intergroup study of the European Organisation for Research and Treatment of Cancer Lung Cancer Group and the National Cancer Institute of Canada. J Clin Oncol. 2005;23(28):6881–6889. doi: 10.1200/JCO.20005.14.589. [DOI] [PubMed] [Google Scholar]
  • 36.Fennell DA, Gaudino G, O’Byrne KJ, Mutti L, van Meerbeeck J. Advances in the systemic therapy of malignant pleural mesothelioma. Nat Clin Pract Oncol. 2008;5(3):136–147. doi: 10.1038/ncponc1039. [DOI] [PubMed] [Google Scholar]
  • 37.Ceresoli GL, Zucali PA, Gianoncelli L, Lorenzi E, Santoro A. Second-line treatment for malignant pleural mesothelioma. Cancer Treat Rev. 2010;36(1):24–32. doi: 10.1016/j.ctrv.2009.09.003. [DOI] [PubMed] [Google Scholar]
  • 38. Comertpay S, Pastorino S, Tanji M, et al. Evaluation of clonal origin of malignant mesothelioma. J Transl Med. 2014;12:301. doi: 10.1186/s12967-014-0301-3. ** Recent finding that malignant mesothelioma originate as polyclonal tumors contribute to explain the occurrence of drug-resistance and complicates the attempt to develop target therapies; therefore different approaches such as targeting chronic inflammation and/or tumor escape from immune surveillance appear to be most promising for therapeutic intervention.
  • 39.Saracci R, Simonato L. Familial malignant mesothelioma. Lancet. 2001;358(9295):1813–1814. doi: 10.1016/S0140-6736(01)06816-7. [DOI] [PubMed] [Google Scholar]
  • 40. Carbone M, Ferris LK, Baumann F, et al. BAP1 cancer syndrome: malignant mesothelioma, uveal and cutaneous melanoma, and MBAITs. J Transl Med. 2012;10:179. doi: 10.1186/1479-5876-10-179. ** Identification of germline mutations in the BRCA-associated protein 1 ( BAP1 ) tumor suppressor gene as the cause of the “BAP1 cancer syndrome” that predispose to malignant mesothelioma and other cancers.
  • 41.Wiesner T, Obenauf AC, Murali R, et al. Germline mutations in BAP1 predispose to melanocytic tumors. Nature genetics. 2011;43(10):1018–1021. doi: 10.1038/ng.910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Piris A, Mihm MC, Jr, Hoang MP. BAP1 and BRAFV600E expression in benign and malignant melanocytic proliferations. Hum Pathol. 2015;46(2):239–245. doi: 10.1016/j.humpath.2014.10.015. [DOI] [PubMed] [Google Scholar]
  • 43.Abdel-Rahman MH, Pilarski R, Cebulla CM, et al. Germline BAP1 mutation predisposes to uveal melanoma, lung adenocarcinoma, meningioma, and other cancers. J Med Genet. 2011;48(12):856–859. doi: 10.1136/jmedgenet-2011-100156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Popova T, Hebert L, Jacquemin V, et al. Germline BAP1 mutations predispose to renal cell carcinomas. Am J Hum Genet. 2013;92(6):974–980. doi: 10.1016/j.ajhg.2013.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Jiao Y, Pawlik TM, Anders RA, et al. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nature genetics. 2013;45(12):1470–1473. doi: 10.1038/ng.2813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wadt KA, Aoude LG, Johansson P, et al. A recurrent germline BAP1 mutation and extension of the BAP1 tumor predisposition spectrum to include basal cell carcinoma. Clinical genetics. 2014 doi: 10.1111/cge.12501. [DOI] [PubMed] [Google Scholar]
  • 47.Carbone M, Yang H, Pass HI, Krausz T, Testa JR, Gaudino G. BAP1 and cancer. Nature reviews. Cancer. 2013;13(3):153–159. doi: 10.1038/nrc3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Nasu M, Emi M, Pastorino S, et al. High Incidence of Somatic BAP1 Alterations in Sporadic Malignant Mesothelioma. J Thorac Oncol. 2015;10(4):565–576. doi: 10.1097/JTO.0000000000000471. * BAP1 is mutated in about 60% of sporadic malignant mesotheliomas making it the most commonly mutated gene in this malignancy.
  • 49.Yoshikawa Y, Sato A, Tsujimura T, et al. Frequent inactivation of the BAP1 gene in epithelioid-type malignant mesothelioma. Cancer Sci. 2012;103(5):868–874. doi: 10.1111/j.1349-7006.2012.02223.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Guo G, Chmielecki J, Goparaju C, et al. Whole-exome sequencing reveals frequent genetic alterations in BAP1, NF2, CDKN2A, and CUL1 in malignant pleural mesothelioma. Cancer Res. 2015;75(2):264–269. doi: 10.1158/0008-5472.CAN-14-1008. [DOI] [PubMed] [Google Scholar]
  • 51.Lo Iacono M, Monica V, Righi L, et al. Targeted next-generation sequencing of cancer genes in advanced stage malignant pleural mesothelioma: a retrospective study. J Thorac Oncol. 2015;10(3):492–499. doi: 10.1097/JTO.0000000000000436. [DOI] [PubMed] [Google Scholar]
  • 52.Arzt L, Quehenberger F, Halbwedl I, Mairinger T, Popper HH. BAP1 protein is a progression factor in malignant pleural mesothelioma. Pathol Oncol Res. 2014;20(1):145–151. doi: 10.1007/s12253-013-9677-2. [DOI] [PubMed] [Google Scholar]
  • 53.Farzin M, Toon CW, Clarkson A, et al. Loss of expression of BAP1 predicts longer survival in mesothelioma. Pathology. 2015;47(4):302–307. doi: 10.1097/PAT.0000000000000250. [DOI] [PubMed] [Google Scholar]
  • 54.Baumann F, Flores E, Napolitano A, et al. Mesothelioma patients with germline BAP1 mutations have 7-fold improved long-term survival. Carcinogenesis. 2015;36(1):76–81. doi: 10.1093/carcin/bgu227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jensen DE, Proctor M, Marquis ST, et al. BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene. 1998;16(9):1097–1112. doi: 10.1038/sj.onc.1201861. [DOI] [PubMed] [Google Scholar]
  • 56.Ventii KH, Devi NS, Friedrich KL, et al. BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer research. 2008;68(17):6953–6962. doi: 10.1158/0008-5472.CAN-08-0365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Scheuermann JC, de Ayala Alonso AG, Oktaba K, et al. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature. 2010;465(7295):243–247. doi: 10.1038/nature08966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Misaghi S, Ottosen S, Izrael-Tomasevic A, et al. Association of C-terminal ubiquitin hydrolase BRCA1-associated protein 1 with cell cycle regulator host cell factor 1. Molecular and cellular biology. 2009;29(8):2181–2192. doi: 10.1128/MCB.01517-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Machida YJ, Machida Y, Vashisht AA, Wohlschlegel JA, Dutta A. The deubiquitinating enzyme BAP1 regulates cell growth via interaction with HCF-1. J Biol Chem. 2009;284(49):34179–34188. doi: 10.1074/jbc.M109.046755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Eletr ZM, Wilkinson KD. An emerging model for BAP1’s role in regulating cell cycle progression. Cell Biochem Biophys. 2011;60(1–2):3–11. doi: 10.1007/s12013-011-9184-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Yu H, Mashtalir N, Daou S, et al. The ubiquitin carboxyl hydrolase BAP1 forms a ternary complex with YY1 and HCF-1 and is a critical regulator of gene expression. Mol Cell Biol. 2010;30(21):5071–5085. doi: 10.1128/MCB.00396-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ismail IH, Davidson R, Gagne JP, Xu ZZ, Poirier GG, Hendzel MJ. Germline mutations in BAP1 impair its function in DNA double-strand break repair. Cancer Res. 2014;74(16):4282–4294. doi: 10.1158/0008-5472.CAN-13-3109. [DOI] [PubMed] [Google Scholar]
  • 63.Yu H, Pak H, Hammond-Martel I, et al. Tumor suppressor and deubiquitinase BAP1 promotes DNA double-strand break repair. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(1):285–290. doi: 10.1073/pnas.1309085110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Landreville S, Agapova OA, Matatall KA, et al. Histone deacetylase inhibitors induce growth arrest and differentiation in uveal melanoma. Clin Cancer Res. 2012;18(2):408–416. doi: 10.1158/1078-0432.CCR-11-0946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006;5(9):769–784. doi: 10.1038/nrd2133. [DOI] [PubMed] [Google Scholar]
  • 66.Cao XX, Mohuiddin I, Ece F, McConkey DJ, Smythe WR. Histone deacetylase inhibitor downregulation of bcl-xl gene expression leads to apoptotic cell death in mesothelioma. Am J Respir Cell Mol Biol. 2001;25(5):562–568. doi: 10.1165/ajrcmb.25.5.4539. [DOI] [PubMed] [Google Scholar]
  • 67.Neuzil J, Swettenham E, Gellert N. Sensitization of mesothelioma to TRAIL apoptosis by inhibition of histone deacetylase: role of Bcl-xL down-regulation. Biochem Biophys Res Commun. 2004;314(1):186–191. doi: 10.1016/j.bbrc.2003.12.074. [DOI] [PubMed] [Google Scholar]
  • 68.Nguyen DM, Schrump WD, Chen GA, et al. Abrogation of p21 expression by flavopiridol enhances depsipeptide-mediated apoptosis in malignant pleural mesothelioma cells. Clin Cancer Res. 2004;10(5):1813–1825. doi: 10.1158/1078-0432.ccr-0901-3. [DOI] [PubMed] [Google Scholar]
  • 69.Symanowski J, Vogelzang N, Zawel L, Atadja P, Pass H, Sharma S. A histone deacetylase inhibitor LBH589 downregulates XIAP in mesothelioma cell lines which is likely responsible for increased apoptosis with TRAIL. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2009;4(2):149–160. doi: 10.1097/JTO.0b013e318194f991. [DOI] [PubMed] [Google Scholar]
  • 70.Vandermeers F, Hubert P, Delvenne P, et al. Valproate, in combination with pemetrexed and cisplatin, provides additional efficacy to the treatment of malignant mesothelioma. Clin Cancer Res. 2009;15(8):2818–2828. doi: 10.1158/1078-0432.CCR-08-1579. [DOI] [PubMed] [Google Scholar]
  • 71.Sacco JJ, Kenyani J, Butt Z, et al. Loss of the deubiquitylase BAP1 alters class I histone deacetylase expression and sensitivity of mesothelioma cells to HDAC inhibitors. Oncotarget. 2015 doi: 10.18632/oncotarget.3765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kelly WK, O’Connor OA, Krug LM, et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol. 2005;23(17):3923–3931. doi: 10.1200/JCO.2005.14.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Krug LM, Kindler HL, Calvert H, et al. Vorinostat in patients with advanced malignant pleural mesothelioma who have progressed on previous chemotherapy (VANTAGE-014): a phase 3, double-blind, randomised, placebo-controlled trial. Lancet Oncol. 2015;16(4):447–456. doi: 10.1016/S1470-2045(15)70056-2. * Negative results of the VANTAGE-014 study raise awareness about the relevance and predictively of preclinical outcomes.
  • 74.Ramalingam SS, Belani CP, Ruel C, et al. Phase II study of belinostat (PXD101), a histone deacetylase inhibitor, for second line therapy of advanced malignant pleural mesothelioma. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2009;4(1):97–101. doi: 10.1097/JTO.0b013e318191520c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Dey A, Seshasayee D, Noubade R, et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science. 2012;337(6101):1541–1546. doi: 10.1126/science.1221711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Napolitano A, Pellegrini L, Dey A, et al. Minimal asbestos exposure in germline BAP1 heterozygous mice is associated with deregulated inflammatory response and increased risk of mesothelioma. Oncogene. 2015 doi: 10.1038/onc.2015.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cornelissen R, Lievense LA, Maat AP, et al. Ratio of intratumoral macrophage phenotypes is a prognostic factor in epithelioid malignant pleural mesothelioma. PloS one. 2014;9(9):e106742. doi: 10.1371/journal.pone.0106742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Altomare DA, Vaslet CA, Skele KL, et al. A mouse model recapitulating molecular features of human mesothelioma. Cancer research. 2005;65(18):8090–8095. doi: 10.1158/0008-5472.CAN-05-2312. [DOI] [PubMed] [Google Scholar]
  • 79.Bianchi AB, Mitsunaga SI, Cheng JQ, et al. High frequency of inactivating mutations in the neurofibromatosis type 2 gene (NF2) in primary malignant mesotheliomas. Proc Natl Acad Sci U S A. 1995;92(24):10854–10858. doi: 10.1073/pnas.92.24.10854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ladanyi M, Zauderer MG, Krug LM, et al. New strategies in pleural mesothelioma: BAP1 and NF2 as novel targets for therapeutic development and risk assessment. Clin Cancer Res. 2012;18(17):4485–4490. doi: 10.1158/1078-0432.CCR-11-2375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lopez-Lago MA, Okada T, Murillo MM, Socci N, Giancotti FG. Loss of the tumor suppressor gene NF2, encoding merlin, constitutively activates integrin-dependent mTORC1 signaling. Molecular and cellular biology. 2009;29(15):4235–4249. doi: 10.1128/MCB.01578-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Ou SH, Moon J, Garland LL, et al. SWOG S0722: phase II study of mTOR inhibitor everolimus (RAD001) in advanced malignant pleural mesothelioma (MPM) Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2015;10(2):387–391. doi: 10.1097/JTO.0000000000000360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Carracedo A, Ma L, Teruya-Feldstein J, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K–dependent feedback loop in human cancer. J Clin Invest. 2008;118(9):3065–3074. doi: 10.1172/JCI34739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kanteti R, Dhanasingh I, Kawada I, et al. MET and PI3K/mTOR as a potential combinatorial therapeutic target in malignant pleural mesothelioma. PLoS One. 2014;9(9):e105919. doi: 10.1371/journal.pone.0105919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zhou S, Liu L, Li H, et al. Multipoint targeting of the PI3K/mTOR pathway in mesothelioma. British journal of cancer. 2014;110(10):2479–2488. doi: 10.1038/bjc.2014.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC. The role of focal-adhesion kinase in cancer - a new therapeutic opportunity. Nature reviews. Cancer. 2005;5(7):505–515. doi: 10.1038/nrc1647. [DOI] [PubMed] [Google Scholar]
  • 87. Poulikakos PI, Xiao GH, Gallagher R, Jablonski S, Jhanwar SC, Testa JR. Re-expression of the tumor suppressor NF2/merlin inhibits invasiveness in mesothelioma cells and negatively regulates FAK. Oncogene. 2006;25(44):5960–5968. doi: 10.1038/sj.onc.1209587. * Correlation between merlin inactivation and upregulation of FAK activity in malignant mesothelioma pathogenesis set the rationale for promising clinical trials using FAK inhibitors.
  • 88.Pan D. The hippo signaling pathway in development and cancer. Dev Cell. 2010;19(4):491–505. doi: 10.1016/j.devcel.2010.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Striedinger K, VandenBerg SR, Baia GS, McDermott MW, Gutmann DH, Lal A. The neurofibromatosis 2 tumor suppressor gene product, merlin, regulates human meningioma cell growth by signaling through YAP. Neoplasia. 2008;10(11):1204–1212. doi: 10.1593/neo.08642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Yokoyama T, Osada H, Murakami H, et al. YAP1 is involved in mesothelioma development and negatively regulated by Merlin through phosphorylation. Carcinogenesis. 2008;29(11):2139–2146. doi: 10.1093/carcin/bgn200. [DOI] [PubMed] [Google Scholar]
  • 91.Mizuno T, Murakami H, Fujii M, et al. YAP induces malignant mesothelioma cell proliferation by upregulating transcription of cell cycle-promoting genes. Oncogene. 2012;31(49):5117–5122. doi: 10.1038/onc.2012.5. [DOI] [PubMed] [Google Scholar]
  • 92.Liu-Chittenden Y, Huang B, Shim JS, et al. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev. 2012;26(12):1300–1305. doi: 10.1101/gad.192856.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Shi Y, Moura U, Opitz I, et al. Role of hedgehog signaling in malignant pleural mesothelioma. Clin Cancer Res. 2012;18(17):4646–4656. doi: 10.1158/1078-0432.CCR-12-0599. [DOI] [PubMed] [Google Scholar]
  • 94.Hirao T, Bueno R, Chen CJ, Gordon GJ, Heilig E, Kelsey KT. Alterations of the p16(INK4) locus in human malignant mesothelial tumors. Carcinogenesis. 2002;23(7):1127–1130. doi: 10.1093/carcin/23.7.1127. [DOI] [PubMed] [Google Scholar]
  • 95.Sekido Y, Pass HI, Bader S, et al. Neurofibromatosis type 2 (NF2) gene is somatically mutated in mesothelioma but not in lung cancer. Cancer research. 1995;55(6):1227–1231. [PubMed] [Google Scholar]
  • 96.Musti M, Kettunen E, Dragonieri S, et al. Cytogenetic and molecular genetic changes in malignant mesothelioma. Cancer Genet Cytogenet. 2006;170(1):9–15. doi: 10.1016/j.cancergencyto.2006.04.011. [DOI] [PubMed] [Google Scholar]
  • 97.Altomare DA, Menges CW, Xu J, et al. Losses of both products of the Cdkn2a/Arf locus contribute to asbestos-induced mesothelioma development and cooperate to accelerate tumorigenesis. PloS one. 2011;6(4):e18828. doi: 10.1371/journal.pone.0018828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Tada Y, Shimada H, Hiroshima K, Tagawa M. A potential therapeutic strategy for malignant mesothelioma with gene medicine. Biomed Res Int. 2013;2013:572609. doi: 10.1155/2013/572609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Frizelle SP, Grim J, Zhou J, et al. Re-expression of p16INK4a in mesothelioma cells results in cell cycle arrest, cell death, tumor suppression and tumor regression. Oncogene. 1998;16(24):3087–3095. doi: 10.1038/sj.onc.1201870. [DOI] [PubMed] [Google Scholar]
  • 100.Frizelle SP, Kratzke MG, Carreon RR, et al. Inhibition of both mesothelioma cell growth and Cdk4 activity following treatment with a TATp16INK4a peptide. Anticancer research. 2008;28(1A):1–7. [PubMed] [Google Scholar]
  • 101.Brevet M, Shimizu S, Bott MJ, et al. Coactivation of receptor tyrosine kinases in malignant mesothelioma as a rationale for combination targeted therapy. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2011;6(5):864–874. doi: 10.1097/jto.0b013e318215a07d. [DOI] [PubMed] [Google Scholar]
  • 102.Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nature reviews. Cancer. 2009;9(1):28–39. doi: 10.1038/nrc2559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Yano S, Li Q, Wang W, et al. Antiangiogenic therapies for malignant pleural mesothelioma. Front Biosci (Landmark Ed) 2011;16:740–748. doi: 10.2741/3716. [DOI] [PubMed] [Google Scholar]
  • 104.Strizzi L, Catalano A, Vianale G, et al. Vascular endothelial growth factor is an autocrine growth factor in human malignant mesothelioma. J Pathol. 2001;193(4):468–475. doi: 10.1002/path.824. [DOI] [PubMed] [Google Scholar]
  • 105.Muhsin M, Graham J, Kirkpatrick P. Bevacizumab. Nat Rev Drug Discov. 2004;3(12):995–996. doi: 10.1038/nrd1601. [DOI] [PubMed] [Google Scholar]
  • 106.Ceresoli GL, Zucali PA, Mencoboni M, et al. Phase II study of pemetrexed and carboplatin plus bevacizumab as first-line therapy in malignant pleural mesothelioma. British journal of cancer. 2013;109(3):552–558. doi: 10.1038/bjc.2013.368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Dowell JE, Dunphy FR, Taub RN, et al. A multicenter phase II study of cisplatin, pemetrexed, and bevacizumab in patients with advanced malignant mesothelioma. Lung Cancer. 2012;77(3):567–571. doi: 10.1016/j.lungcan.2012.05.111. [DOI] [PubMed] [Google Scholar]
  • 108.Kindler HL, Karrison TG, Gandara DR, et al. Multicenter, double-blind, placebo-controlled, randomized phase II trial of gemcitabine/cisplatin plus bevacizumab or placebo in patients with malignant mesothelioma. J Clin Oncol. 2012;30(20):2509–2515. doi: 10.1200/JCO.2011.41.5869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Campbell NP, Kunnavakkam R, Leighl N, et al. Cediranib in patients with malignant mesothelioma: a phase II trial of the University of Chicago Phase II Consortium. Lung Cancer. 2012;78(1):76–80. doi: 10.1016/j.lungcan.2012.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Garland LL, Chansky K, Wozniak AJ, et al. Phase II study of cediranib in patients with malignant pleural mesothelioma: SWOG S0509. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2011;6(11):1938–1945. doi: 10.1097/JTO.0b013e318229586e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Laurie SA, Gupta A, Chu Q, et al. Brief report: a phase II study of sunitinib in malignant pleural mesothelioma. the NCIC Clinical Trials Group. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2011;6(11):1950–1954. doi: 10.1097/JTO.0b013e3182333df5. [DOI] [PubMed] [Google Scholar]
  • 112.Nowak AK, Millward MJ, Creaney J, et al. A phase II study of intermittent sunitinib malate as second-line therapy in progressive malignant pleural mesothelioma. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2012;7(9):1449–1456. doi: 10.1097/JTO.0b013e31825f22ee. [DOI] [PubMed] [Google Scholar]
  • 113.Camidge DR, Blais N, Jonker DJ, et al. Sunitinib combined with pemetrexed and cisplatin: results of a phase I dose-escalation and pharmacokinetic study in patients with advanced solid malignancies, with an expanded cohort in non-small cell lung cancer and mesothelioma. Cancer chemotherapy and pharmacology. 2013;71(2):307–319. doi: 10.1007/s00280-012-2008-6. [DOI] [PubMed] [Google Scholar]
  • 114.Dubey S, Janne PA, Krug L, et al. A phase II study of sorafenib in malignant mesothelioma: results of Cancer and Leukemia Group B 30307. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2010;5(10):1655–1661. doi: 10.1097/JTO.0b013e3181ec18db. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Papa S, Popat S, Shah R, et al. Phase 2 study of sorafenib in malignant mesothelioma previously treated with platinum-containing chemotherapy. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2013;8(6):783–787. doi: 10.1097/JTO.0b013e31828c2b26. [DOI] [PubMed] [Google Scholar]
  • 116.Jahan T, Gu L, Kratzke R, et al. Vatalanib in malignant mesothelioma: a phase II trial by the Cancer and Leukemia Group B (CALGB 30107) Lung Cancer. 2012;76(3):393–396. doi: 10.1016/j.lungcan.2011.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Honda M, Kanno T, Fujita Y, Gotoh A, Nakano T, Nishizaki T. Mesothelioma cell proliferation through autocrine activation of PDGF-betabeta receptor. Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology. 2012;29(5–6):667–674. doi: 10.1159/000176386. [DOI] [PubMed] [Google Scholar]
  • 118.Filiberti R, Marroni P, Neri M, et al. Serum PDGF-AB in pleural mesothelioma. Tumour Biol. 2005;26(5):221–226. doi: 10.1159/000087376. [DOI] [PubMed] [Google Scholar]
  • 119.Versnel MA, Claesson-Welsh L, Hammacher A, et al. Human malignant mesothelioma cell lines express PDGF beta-receptors whereas cultured normal mesothelial cells express predominantly PDGF alpha-receptors. Oncogene. 1991;6(11):2005–2011. [PubMed] [Google Scholar]
  • 120.Langerak AW, van der Linden-van Beurden CA, Versnel MA. Regulation of differential expression of platelet-derived growth factor alpha- and beta-receptor mRNA in normal and malignant human mesothelial cell lines. Biochimica et biophysica acta. 1996;1305(1–2):63–70. doi: 10.1016/0167-4781(95)00196-4. [DOI] [PubMed] [Google Scholar]
  • 121.Langerak AW, De Laat PA, Van Der Linden-Van Beurden CA, et al. Expression of platelet-derived growth factor (PDGF) and PDGF receptors in human malignant mesothelioma in vitro and in vivo. J Pathol. 1996;178(2):151–160. doi: 10.1002/(SICI)1096-9896(199602)178:2<151::AID-PATH425>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
  • 122.Porta C, Mutti L, Tassi G. Negative results of an Italian Group for Mesothelioma (G.I.Me.) pilot study of single-agent imatinib mesylate in malignant pleural mesothelioma. Cancer chemotherapy and pharmacology. 2007;59(1):149–150. doi: 10.1007/s00280-006-0243-4. [DOI] [PubMed] [Google Scholar]
  • 123.Mathy A, Baas P, Dalesio O, van Zandwijk N. Limited efficacy of imatinib mesylate in malignant mesothelioma: a phase II trial. Lung Cancer. 2005;50(1):83–86. doi: 10.1016/j.lungcan.2005.04.010. [DOI] [PubMed] [Google Scholar]
  • 124.Tsao AS, Harun N, Lee JJ, et al. Phase I trial of cisplatin, pemetrexed, and imatinib mesylate in chemonaive patients with unresectable malignant pleural mesothelioma. Clin Lung Cancer. 2014;15(3):197–201. doi: 10.1016/j.cllc.2013.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Dudek AZ, Pang H, Kratzke RA, et al. Phase II study of dasatinib in patients with previously treated malignant mesothelioma (cancer and leukemia group B 30601): a brief report. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2012;7(4):755–759. doi: 10.1097/JTO.0b013e318248242c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Garland LL, Rankin C, Gandara DR, et al. Phase II study of erlotinib in patients with malignant pleural mesothelioma: a Southwest Oncology Group Study. J Clin Oncol. 2007;25(17):2406–2413. doi: 10.1200/JCO.2006.09.7634. [DOI] [PubMed] [Google Scholar]
  • 127.Govindan R, Kratzke RA, Herndon JE, 2nd, et al. Gefitinib in patients with malignant mesothelioma: a phase II study by the Cancer and Leukemia Group B. Clin Cancer Res. 2005;11(6):2300–2304. doi: 10.1158/1078-0432.CCR-04-1940. [DOI] [PubMed] [Google Scholar]
  • 128.Jackman DM, Kindler HL, Yeap BY, et al. Erlotinib plus bevacizumab in previously treated patients with malignant pleural mesothelioma. Cancer. 2008;113(4):808–814. doi: 10.1002/cncr.23617. [DOI] [PubMed] [Google Scholar]
  • 129.Strizzi L, Vianale G, Catalano A, Muraro R, Mutti L, Procopio A. Basic fibroblast growth factor in mesothelioma pleural effusions: correlation with patient survival and angiogenesis. Int J Oncol. 2001;18(5):1093–1098. doi: 10.3892/ijo.18.5.1093. [DOI] [PubMed] [Google Scholar]
  • 130.Schelch K, Hoda MA, Klikovits T, et al. Fibroblast growth factor receptor inhibition is active against mesothelioma and synergizes with radio- and chemotherapy. Am J Respir Crit Care Med. 2014;190(7):763–772. doi: 10.1164/rccm.201404-0658OC. [DOI] [PubMed] [Google Scholar]
  • 131.Stapelberg M, Gellert N, Swettenham E, et al. Alpha-tocopheryl succinate inhibits malignant mesothelioma by disrupting the fibroblast growth factor autocrine loop: mechanism and the role of oxidative stress. The Journal of biological chemistry. 2005;280(27):25369–25376. doi: 10.1074/jbc.M414498200. [DOI] [PubMed] [Google Scholar]
  • 132.Matsumoto K, Nakamura T. NK4 (HGF-antagonist/angiogenesis inhibitor) in cancer biology and therapeutics. Cancer Sci. 2003;94(4):321–327. doi: 10.1111/j.1349-7006.2003.tb01440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Leon LG, Gemelli M, Sciarrillo R, Avan A, Funel N, Giovannetti E. Synergistic activity of the c-Met and tubulin inhibitor tivantinib (ARQ197) with pemetrexed in mesothelioma cells. Curr Drug Targets. 2014;15(14):1331–1340. doi: 10.2174/1389450116666141205160924. [DOI] [PubMed] [Google Scholar]
  • 134.Szlosarek PW, Klabatsa A, Pallaska A, et al. In vivo loss of expression of argininosuccinate synthetase in malignant pleural mesothelioma is a biomarker for susceptibility to arginine depletion. Clin Cancer Res. 2006;12(23):7126–7131. doi: 10.1158/1078-0432.CCR-06-1101. [DOI] [PubMed] [Google Scholar]
  • 135.Hata A, Lieberman J. Dysregulation of microRNA biogenesis and gene silencing in cancer. Science signaling. 2015;8(368):re3. doi: 10.1126/scisignal.2005825. [DOI] [PubMed] [Google Scholar]
  • 136.Guled M, Lahti L, Lindholm PM, et al. CDKN2A, NF2, and JUN are dysregulated among other genes by miRNAs in malignant mesothelioma -A miRNA microarray analysis. Genes Chromosomes Cancer. 2009;48(7):615–623. doi: 10.1002/gcc.20669. [DOI] [PubMed] [Google Scholar]
  • 137.Busacca S, Germano S, De Cecco L, et al. MicroRNA signature of malignant mesothelioma with potential diagnostic and prognostic implications. Am J Respir Cell Mol Biol. 2010;42(3):312–319. doi: 10.1165/rcmb.2009-0060OC. [DOI] [PubMed] [Google Scholar]
  • 138.Reid G, Pel ME, Kirschner MB, et al. Restoring expression of miR-16: a novel approach to therapy for malignant pleural mesothelioma. Ann Oncol. 2013;24(12):3128–3135. doi: 10.1093/annonc/mdt412. [DOI] [PubMed] [Google Scholar]
  • 139.MacDiarmid JA, Brahmbhatt H. Minicells: versatile vectors for targeted drug or si/shRNA cancer therapy. Curr Opin Biotechnol. 2011;22(6):909–916. doi: 10.1016/j.copbio.2011.04.008. [DOI] [PubMed] [Google Scholar]
  • 140.Sartore-Bianchi A, Gasparri F, Galvani A, et al. Bortezomib inhibits nuclear factor-kappaB dependent survival and has potent in vivo activity in mesothelioma. Clin Cancer Res. 2007;13(19):5942–5951. doi: 10.1158/1078-0432.CCR-07-0536. [DOI] [PubMed] [Google Scholar]
  • 141.O’Brien ME, Gaafar RM, Popat S, et al. Phase II study of first-line bortezomib and cisplatin in malignant pleural mesothelioma and prospective validation of progression free survival rate as a primary end-point for mesothelioma clinical trials (European Organisation for Research and Treatment of Cancer 08052) Eur J Cancer. 2013;49(13):2815–2822. doi: 10.1016/j.ejca.2013.05.008. [DOI] [PubMed] [Google Scholar]
  • 142.Nasu M, Carbone M, Gaudino G, et al. Ranpirnase Interferes with NF-kappaB Pathway and MMP9 Activity, Inhibiting Malignant Mesothelioma Cell Invasiveness and Xenograft Growth. Genes Cancer. 2011;2(5):576–584. doi: 10.1177/1947601911412375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Corporation A. Annual Report 2009. (Ed.^(Eds) 2009 [Google Scholar]
  • 144.de Melo M, Gerbase MW, Curran J, Pache JC. Phosphorylated extracellular signal-regulated kinases are significantly increased in malignant mesothelioma. J Histochem Cytochem. 2006;54(8):855–861. doi: 10.1369/jhc.5A6807.2006. [DOI] [PubMed] [Google Scholar]
  • 145.Zanella CL, Posada J, Tritton TR, Mossman BT. Asbestos causes stimulation of the extracellular signal-regulated kinase 1 mitogen-activated protein kinase cascade after phosphorylation of the epidermal growth factor receptor. Cancer research. 1996;56(23):5334–5338. [PubMed] [Google Scholar]
  • 146.Blum W, Schwaller B. Calretinin is essential for mesothelioma cell growth/survival in vitro: a potential new target for malignant mesothelioma therapy? Int J Cancer. 2013;133(9):2077–2088. doi: 10.1002/ijc.28218. [DOI] [PubMed] [Google Scholar]
  • 147.Mazieres J, You L, He B, et al. Wnt2 as a new therapeutic target in malignant pleural mesothelioma. Int J Cancer. 2005;117(2):326–332. doi: 10.1002/ijc.21160. [DOI] [PubMed] [Google Scholar]
  • 148.Okamoto J, Mikami I, Tominaga Y, et al. Inhibition of Hsp90 leads to cell cycle arrest and apoptosis in human malignant pleural mesothelioma. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2008;3(10):1089–1095. doi: 10.1097/JTO.0b013e3181839693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Dy GK, Adjei AA. Understanding, recognizing, and managing toxicities of targeted anticancer therapies. CA Cancer J Clin. 2013;63(4):249–279. doi: 10.3322/caac.21184. [DOI] [PubMed] [Google Scholar]
  • 150.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 151.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Mansfield AS, Roden AC, Peikert T, et al. B7-H1 expression in malignant pleural mesothelioma is associated with sarcomatoid histology and poor prognosis. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2014;9(7):1036–1040. doi: 10.1097/JTO.0000000000000177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Cedres S, Ponce-Aix S, Zugazagoitia J, et al. Analysis of expression of programmed cell death 1 ligand 1 (PD-L1) in malignant pleural mesothelioma (MPM) PloS one. 2015;10(3):e0121071. doi: 10.1371/journal.pone.0121071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Calabro L, Morra A, Fonsatti E, et al. Tremelimumab for patients with chemotherapy-resistant advanced malignant mesothelioma: an open-label, single-arm, phase 2 trial. Lancet Oncol. 2013;14(11):1104–1111. doi: 10.1016/S1470-2045(13)70381-4. [DOI] [PubMed] [Google Scholar]
  • 155. Calabro L, Morra A, Fonsatti E, et al. Efficacy and safety of an intensified schedule of tremelimumab for chemotherapy-resistant malignant mesothelioma: an open-label, single-arm, phase 2 study. Lancet Respir Med. 2015;3(4):301–309. doi: 10.1016/S2213-2600(15)00092-2. ** The CTLA-4 inhibitor, tremelimumab, showed clinical and immunological activity in patients with advanced malignant mesothelioma, and a good safety profile.
  • 156.A Potential Immune Therapy for Mesothelioma. Cancer discovery. 2015 doi: 10.1158/2159-8290.CD-NB2015-067. [DOI] [PubMed] [Google Scholar]
  • 157.Stevenson JP, Kindler HL, Papasavvas E, et al. Immunological effects of the TGFbeta-blocking antibody GC1008 in malignant pleural mesothelioma patients. Oncoimmunology. 2013;2(8):e26218. doi: 10.4161/onci.26218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Pastan I, Hassan R. Discovery of mesothelin and exploiting it as a target for immunotherapy. Cancer research. 2014;74(11):2907–2912. doi: 10.1158/0008-5472.CAN-14-0337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Servais EL, Colovos C, Rodriguez L, et al. Mesothelin overexpression promotes mesothelioma cell invasion and MMP-9 secretion in an orthotopic mouse model and in epithelioid pleural mesothelioma patients. Clin Cancer Res. 2012;18(9):2478–2489. doi: 10.1158/1078-0432.CCR-11-2614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Rump A, Morikawa Y, Tanaka M, et al. Binding of ovarian cancer antigen CA125/MUC16 to mesothelin mediates cell adhesion. The Journal of biological chemistry. 2004;279(10):9190–9198. doi: 10.1074/jbc.M312372200. [DOI] [PubMed] [Google Scholar]
  • 161.Gubbels JA, Belisle J, Onda M, et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer. 2006;5(1):50. doi: 10.1186/1476-4598-5-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Hassan R, Ebel W, Routhier EL, et al. Preclinical evaluation of MORAb-009, a chimeric antibody targeting tumor-associated mesothelin. Cancer Immun. 2007;7:20. [PMC free article] [PubMed] [Google Scholar]
  • 163.Hassan R, Schweizer C, Lu KF, et al. Inhibition of mesothelin-CA-125 interaction in patients with mesothelioma by the anti-mesothelin monoclonal antibody MORAb-009: Implications for cancer therapy. Lung Cancer. 2010;68(3):455–459. doi: 10.1016/j.lungcan.2009.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hassan R, Cohen SJ, Phillips M, et al. Phase I clinical trial of the chimeric anti-mesothelin monoclonal antibody MORAb-009 in patients with mesothelin-expressing cancers. Clin Cancer Res. 2010;16(24):6132–6138. doi: 10.1158/1078-0432.CCR-10-2275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Hassan R, Kindler HL, Jahan T, et al. Phase II clinical trial of amatuximab, a chimeric antimesothelin antibody with pemetrexed and cisplatin in advanced unresectable pleural mesothelioma. Clin Cancer Res. 2014;20(23):5927–5936. doi: 10.1158/1078-0432.CCR-14-0804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Zolot RS, Basu S, Million RP. Antibody-drug conjugates. Nat Rev Drug Discov. 2013;12(4):259–260. doi: 10.1038/nrd3980. [DOI] [PubMed] [Google Scholar]
  • 167.Hassan R, Lerner MR, Benbrook D, et al. Antitumor activity of SS(dsFv)PE38 and SS1(dsFv)PE38, recombinant antimesothelin immunotoxins against human gynecologic cancers grown in organotypic culture in vitro. Clin Cancer Res. 2002;8(11):3520–3526. [PubMed] [Google Scholar]
  • 168.Li Q, Verschraegen CF, Mendoza J, Hassan R. Cytotoxic activity of the recombinant anti-mesothelin immunotoxin, SS1(dsFv)PE38, towards tumor cell lines established from ascites of patients with peritoneal mesotheliomas. Anticancer research. 2004;24(3a):1327–1335. [PubMed] [Google Scholar]
  • 169.Hassan R, Bullock S, Premkumar A, et al. Phase I study of SS1P, a recombinant anti-mesothelin immunotoxin given as a bolus I.V. infusion to patients with mesothelin-expressing mesothelioma, ovarian, and pancreatic cancers. Clin Cancer Res. 2007;13(17):5144–5149. doi: 10.1158/1078-0432.CCR-07-0869. [DOI] [PubMed] [Google Scholar]
  • 170.Kreitman RJ, Hassan R, Fitzgerald DJ, Pastan I. Phase I trial of continuous infusion anti-mesothelin recombinant immunotoxin SS1P. Clin Cancer Res. 2009;15(16):5274–5279. doi: 10.1158/1078-0432.CCR-09-0062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Hassan R, Sharon E, Thomas A, et al. Phase 1 study of the antimesothelin immunotoxin SS1P in combination with pemetrexed and cisplatin for front-line therapy of pleural mesothelioma and correlation of tumor response with serum mesothelin, megakaryocyte potentiating factor, and cancer antigen 125. Cancer. 2014;120(21):3311–3319. doi: 10.1002/cncr.28875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Hassan R, Miller AC, Sharon E, et al. Major cancer regressions in mesothelioma after treatment with an anti-mesothelin immunotoxin and immune suppression. Sci Transl Med. 2013;5(208):208ra147. doi: 10.1126/scitranslmed.3006941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Hollevoet K, Mason-Osann E, Liu XF, Imhof-Jung S, Niederfellner G, Pastan I. In vitro and in vivo activity of the low-immunogenic antimesothelin immunotoxin RG7787 in pancreatic cancer. Mol Cancer Ther. 2014;13(8):2040–2049. doi: 10.1158/1535-7163.MCT-14-0089-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Alewine C, Xiang L, Yamori T, Niederfellner G, Bosslet K, Pastan I. Efficacy of RG7787, a next-generation mesothelin-targeted immunotoxin, against triple-negative breast and gastric cancers. Mol Cancer Ther. 2014;13(11):2653–2661. doi: 10.1158/1535-7163.MCT-14-0132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Pasetto M, Antignani A, Ormanoglu P, et al. Whole-genome RNAi screen highlights components of the endoplasmic reticulum/Golgi as a source of resistance to immunotoxin-mediated cytotoxicity. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(10):E1135–E1142. doi: 10.1073/pnas.1501958112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Scales SJ, Gupta N, Pacheco G, et al. An antimesothelin-monomethyl auristatin e conjugate with potent antitumor activity in ovarian, pancreatic, and mesothelioma models. Mol Cancer Ther. 2014;13(11):2630–2640. doi: 10.1158/1535-7163.MCT-14-0487-T. [DOI] [PubMed] [Google Scholar]
  • 177.Golfier S, Kopitz C, Kahnert A, et al. Anetumab ravtansine: a novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther. 2014;13(6):1537–1548. doi: 10.1158/1535-7163.MCT-13-0926. [DOI] [PubMed] [Google Scholar]
  • 178.Kim H, Gao W, Ho M. Novel immunocytokine IL12-SS1 (Fv) inhibits mesothelioma tumor growth in nude mice. PloS one. 2013;8(11):e81919. doi: 10.1371/journal.pone.0081919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Yuan J, Kashiwagi S, Reeves P, et al. A novel mycobacterial Hsp70-containing fusion protein targeting mesothelin augments antitumor immunity and prolongs survival in murine models of ovarian cancer and mesothelioma. Journal of hematology & oncology. 2014;7:15. doi: 10.1186/1756-8722-7-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Le DT, Brockstedt DG, Nir-Paz R, et al. A live-attenuated Listeria vaccine (ANZ-100) and a live-attenuated Listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: phase I studies of safety and immune induction. Clin Cancer Res. 2012;18(3):858–868. doi: 10.1158/1078-0432.CCR-11-2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Le DT, Wang-Gillam A, Picozzi V, et al. Safety and Survival With GVAX Pancreas Prime and Listeria Monocytogenes-Expressing Mesothelin (CRS-207) Boost Vaccines for Metastatic Pancreatic Cancer. J Clin Oncol. 2015;33(12):1325–1333. doi: 10.1200/JCO.2014.57.4244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Yamasaki S, Miura Y, Davydova J, Vickers SM, Yamamoto M. Intravenous genetic mesothelin vaccine based on human adenovirus 40 inhibits growth and metastasis of pancreatic cancer. Int J Cancer. 2013;133(1):88–97. doi: 10.1002/ijc.27983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Zhao Y, Moon E, Carpenito C, et al. Multiple injections of electroporated autologous T cells expressing a chimeric antigen receptor mediate regression of human disseminated tumor. Cancer research. 2010;70(22):9053–9061. doi: 10.1158/0008-5472.CAN-10-2880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Moon EK, Carpenito C, Sun J, et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin Cancer Res. 2011;17(14):4719–4730. doi: 10.1158/1078-0432.CCR-11-0351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Lanitis E, Poussin M, Hagemann IS, et al. Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptor. Mol Ther. 2012;20(3):633–643. doi: 10.1038/mt.2011.256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Riese MJ, Wang LC, Moon EK, et al. Enhanced effector responses in activated CD8+ T cells deficient in diacylglycerol kinases. Cancer research. 2013;73(12):3566–3577. doi: 10.1158/0008-5472.CAN-12-3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Beatty GL, Haas AR, Maus MV, et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res. 2014;2(2):112–120. doi: 10.1158/2326-6066.CIR-13-0170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Moon EK, Wang LC, Dolfi DV, et al. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin Cancer Res. 2014;20(16):4262–4273. doi: 10.1158/1078-0432.CCR-13-2627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Adusumilli PS, Cherkassky L, Villena-Vargas J, et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med. 2014;6(261):261ra151. doi: 10.1126/scitranslmed.3010162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.May RJ, Dao T, Pinilla-Ibarz J, et al. Peptide epitopes from the Wilms’ tumor 1 oncoprotein stimulate CD4+ and CD8+ T cells that recognize and kill human malignant mesothelioma tumor cells. Clin Cancer Res. 2007;13(15 Pt 1):4547–4555. doi: 10.1158/1078-0432.CCR-07-0708. [DOI] [PubMed] [Google Scholar]
  • 191.Oka Y, Tsuboi A, Taguchi T, et al. Induction of WT1 (Wilms’ tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(38):13885–13890. doi: 10.1073/pnas.0405884101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Krug LM, Dao T, Brown AB, et al. WT1 peptide vaccinations induce CD4 and CD8 T cell immune responses in patients with mesothelioma and non-small cell lung cancer. Cancer Immunol Immunother. 2010;59(10):1467–1479. doi: 10.1007/s00262-010-0871-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Tamanaka T, Oka Y, Fujiki F, et al. Recognition of a natural WT1 epitope by a modified WT1 peptide-specific T-cell receptor. Anticancer research. 2012;32(12):5201–5209. [PubMed] [Google Scholar]
  • 194.Asai H, Fujiwara H, An J, et al. Co-introduced functional CCR2 potentiates in vivo anti-lung cancer functionality mediated by T cells double gene-modified to express WT1-specific T-cell receptor. PloS one. 2013;8(2):e56820. doi: 10.1371/journal.pone.0056820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Kakarla S, Chow KK, Mata M, et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol Ther. 2013;21(8):1611–1620. doi: 10.1038/mt.2013.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Schuberth PC, Hagedorn C, Jensen SM, et al. Treatment of malignant pleural mesothelioma by fibroblast activation protein-specific re-directed T cells. J Transl Med. 2013;11:187. doi: 10.1186/1479-5876-11-187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Wang LC, Lo A, Scholler J, et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe toxicity. Cancer Immunol Res. 2014;2(2):154–166. doi: 10.1158/2326-6066.CIR-13-0027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Tan Z, Zhou J, Cheung AK, et al. Vaccine-elicited CD8+ T cells cure mesothelioma by overcoming tumor-induced immunosuppressive environment. Cancer research. 2014;74(21):6010–6021. doi: 10.1158/0008-5472.CAN-14-0473. [DOI] [PubMed] [Google Scholar]
  • 199.Bertino P, Panigada M, Soprana E, et al. Fowlpox-based survivin vaccination for malignant mesothelioma therapy. Int J Cancer. 2013;133(3):612–623. doi: 10.1002/ijc.28048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Hoffmann PR, Panigada M, Soprana E, et al. Preclinical development of HIvax: human survivin Highly Immunogenic vaccines. Hum Vaccin Immunother. 2015 doi: 10.1080/21645515.2015.1050572. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Rivera Z, Ferrone S, Wang X, et al. CSPG4 as a target of antibody-based immunotherapy for malignant mesothelioma. Clin Cancer Res. 2012;18(19):5352–5363. doi: 10.1158/1078-0432.CCR-12-0628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Beseth BD, Cameron RB, Leland P, et al. Interleukin-4 receptor cytotoxin as therapy for human malignant pleural mesothelioma xenografts. Ann Thorac Surg. 2004;78(2):436–443. doi: 10.1016/j.athoracsur.2004.03.010. discussion 436–443. [DOI] [PubMed] [Google Scholar]
  • 203.Tagawa M, Tada Y, Shimada H, Hiroshima K. Gene therapy for malignant mesothelioma: current prospects and challenges. Cancer Gene Ther. 2013;20(3):150–156. doi: 10.1038/cgt.2013.1. [DOI] [PubMed] [Google Scholar]
  • 204.Zamarin D, Holmgaard RB, Subudhi SK, et al. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci Transl Med. 2014;6(226):226ra232. doi: 10.1126/scitranslmed.3008095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Quetglas JI, Labiano S, Aznar MA, et al. Virotherapy with a Semliki Forest Virus-Based Vector Encoding IL12 Synergizes with PD-1/PD-L1 Blockade. Cancer Immunol Res. 2015;3(5):449–454. doi: 10.1158/2326-6066.CIR-14-0216. [DOI] [PubMed] [Google Scholar]
  • 206.Li H, Peng KW, Dingli D, Kratzke RA, Russell SJ. Oncolytic measles viruses encoding interferon beta and the thyroidal sodium iodide symporter gene for mesothelioma virotherapy. Cancer Gene Ther. 2010;17(8):550–558. doi: 10.1038/cgt.2010.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Gauvrit A, Brandler S, Sapede-Peroz C, Boisgerault N, Tangy F, Gregoire M. Measles virus induces oncolysis of mesothelioma cells and allows dendritic cells to cross-prime tumor-specific CD8 response. Cancer Res. 2008;68(12):4882–4892. doi: 10.1158/0008-5472.CAN-07-6265. [DOI] [PubMed] [Google Scholar]
  • 208.Pesonen S, Diaconu I, Kangasniemi L, et al. Oncolytic immunotherapy of advanced solid tumors with a CD40L–expressing replicating adenovirus: assessment of safety and immunologic responses in patients. Cancer research. 2012;72(7):1621–1631. doi: 10.1158/0008-5472.CAN-11-3001. [DOI] [PubMed] [Google Scholar]
  • 209.Kawasaki Y, Tamamoto A, Takagi-Kimura M, et al. Replication-competent retrovirus vector-mediated prodrug activator gene therapy in experimental models of human malignant mesothelioma. Cancer Gene Ther. 2011;18(8):571–578. doi: 10.1038/cgt.2011.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Adusumilli PS, Stiles BM, Chan MK, et al. Imaging and therapy of malignant pleural mesothelioma using replication-competent herpes simplex viruses. J Gene Med. 2006;8(5):603–615. doi: 10.1002/jgm.877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Carbone M, Yang H. Molecular pathways: targeting mechanisms of asbestos and erionite carcinogenesis in mesothelioma. Clin Cancer Res. 2012;18(3):598–604. doi: 10.1158/1078-0432.CCR-11-2259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Chow MT, Tschopp J, Moller A, Smyth MJ. NLRP3 promotes inflammation-induced skin cancer but is dispensable for asbestos-induced mesothelioma. Immunol Cell Biol. 2012;90(10):983–986. doi: 10.1038/icb.2012.46. [DOI] [PubMed] [Google Scholar]
  • 213.Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81(1):1–5. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
  • 214.Ellerman JE, Brown CK, de Vera M, et al. Masquerader: high mobility group box-1 and cancer. Clin Cancer Res. 2007;13(10):2836–2848. doi: 10.1158/1078-0432.CCR-06-1953. [DOI] [PubMed] [Google Scholar]
  • 215.Catalano A, Graciotti L, Rinaldi L, et al. Preclinical evaluation of the nonsteroidal anti-inflammatory agent celecoxib on malignant mesothelioma chemoprevention. Int J Cancer. 2004;109(3):322–328. doi: 10.1002/ijc.11710. [DOI] [PubMed] [Google Scholar]
  • 216.Rothwell PM, Fowkes FG, Belch JF, Ogawa H, Warlow CP, Meade TW. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet. 2011;377(9759):31–41. doi: 10.1016/S0140-6736(10)62110-1. [DOI] [PubMed] [Google Scholar]
  • 217.Yang H, Pellegrini L, Napolitano A, et al. Aspirin delays mesothelioma growth by inhibiting HMGB1-mediated tumor progression. Cell death & disease. 2015;6:e1786. doi: 10.1038/cddis.2015.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Sitia G, Iannacone M, Muller S, Bianchi ME, Guidotti LG. Treatment with HMGB1 inhibitors diminishes CTL-induced liver disease in HBV transgenic mice. J Leukoc Biol. 2007;81(1):100–107. doi: 10.1189/jlb.0306173. [DOI] [PubMed] [Google Scholar]
  • 219.Lamkanfi M, Mueller JL, Vitari AC, et al. Glyburide inhibits the Cryopyrin/Nalp3 inflammasome. The Journal of cell biology. 2009;187(1):61–70. doi: 10.1083/jcb.200903124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Tabata C, Shibata E, Tabata R, et al. Serum HMGB1 as a prognostic marker for malignant pleural mesothelioma. BMC Cancer. 2013;13:205. doi: 10.1186/1471-2407-13-205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Tabata C, Kanemura S, Tabata R, et al. Serum HMGB1 as a diagnostic marker for malignant peritoneal mesothelioma. J Clin Gastroenterol. 2013;47(8):684–688. doi: 10.1097/MCG.0b013e318297fa65. [DOI] [PubMed] [Google Scholar]
  • 222.Schneider J, Hoffmann H, Dienemann H, Herth FJ, Meister M, Muley T. Diagnostic and prognostic value of soluble mesothelin-related proteins in patients with malignant pleural mesothelioma in comparison with benign asbestosis and lung cancer. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2008;3(11):1317–1324. doi: 10.1097/JTO.0b013e318187491c. [DOI] [PubMed] [Google Scholar]
  • 223.Hollevoet K, Reitsma JB, Creaney J, et al. Serum mesothelin for diagnosing malignant pleural mesothelioma: an individual patient data meta-analysis. J Clin Oncol. 2012;30(13):1541–1549. doi: 10.1200/JCO.2011.39.6671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Pass HI, Lott D, Lonardo F, et al. Asbestos exposure, pleural mesothelioma, and serum osteopontin levels. N Engl J Med. 2005;353(15):1564–1573. doi: 10.1056/NEJMoa051185. [DOI] [PubMed] [Google Scholar]
  • 225.Pass HI, Levin SM, Harbut MR, et al. Fibulin-3 as a blood and effusion biomarker for pleural mesothelioma. N Engl J Med. 2012;367(15):1417–1427. doi: 10.1056/NEJMoa1115050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.de Klerk NH, Musk AW, Ambrosini GL, et al. Vitamin A and cancer prevention II: comparison of the effects of retinol and beta-carotene. Int J Cancer. 1998;75(3):362–367. doi: 10.1002/(sici)1097-0215(19980130)75:3<362::aid-ijc6>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  • 227.Alfonso HS, Reid A, de Klerk NH, et al. Retinol supplementation and mesothelioma incidence in workers earlier exposed to blue asbestos (Crocidolite) at Wittenoom, Western Australia. Eur J Cancer Prev. 2010;19(5):355–359. doi: 10.1097/CEJ.0b013e32833c1bcb. [DOI] [PubMed] [Google Scholar]
  • 228.Robinson C, Woo S, Walsh A, Nowak AK, Lake RA. The antioxidants vitamins A and E and selenium do not reduce the incidence of asbestos-induced disease in a mouse model of mesothelioma. Nutr Cancer. 2012;64(2):315–322. doi: 10.1080/01635581.2012.649100. [DOI] [PubMed] [Google Scholar]
  • 229.Robinson C, Alfonso H, Woo S, et al. Effect of NSAIDS and COX-2 inhibitors on the incidence and severity of asbestos-induced malignant mesothelioma: evidence from an animal model and a human cohort. Lung Cancer. 2014;86(1):29–34. doi: 10.1016/j.lungcan.2014.08.005. [DOI] [PubMed] [Google Scholar]
  • 230.Robinson C, Alfonso H, Woo S, et al. Statins do not alter the incidence of mesothelioma in asbestos exposed mice or humans. PloS one. 2014;9(8):e103025. doi: 10.1371/journal.pone.0103025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Nagai H, Okazaki Y, Chew SH, Misawa N, Yasui H, Toyokuni S. Deferasirox induces mesenchymal-epithelial transition in crocidolite-induced mesothelial carcinogenesis in rats. Cancer Prev Res (Phila) 2013;6(11):1222–1230. doi: 10.1158/1940-6207.CAPR-13-0244. [DOI] [PubMed] [Google Scholar]
  • 232.Yang H, Pellegrini L, Napolitano A, et al. Aspirin delays mesothelioma growth by inhibiting HMGB1-mediated tumor progression. Cell Death and Disease. 2015 doi: 10.1038/cddis.2015.153. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Napolitano A, Pellegrini L, Dey A, et al. Minimal asbestos exposure in germline BAP1 heterozygous mice is associated with deregulated inflammatory response and increased risk of mesothelioma. Oncogene. 2015 doi: 10.1038/onc.2015.243. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES