Next Article in Journal
Active Substances from the Micro-Immunotherapy Medicine 2LC1® Show In Vitro Anti-Cancer Properties in Colon, Prostate, and Breast Cancer Models and Immune-Enhancing Capabilities in Human Macrophages
Previous Article in Journal
Regulation of Immune-Related Gene Expression by Salinity-Induced HPI Axis in Large Yellow Croaker, Larimichthys crocea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms of Tumor Progression and Novel Therapeutic and Diagnostic Strategies in Mesothelioma

1
Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
2
Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(9), 4299; https://doi.org/10.3390/ijms26094299
Submission received: 28 March 2025 / Revised: 27 April 2025 / Accepted: 29 April 2025 / Published: 1 May 2025
(This article belongs to the Special Issue Translational Oncology: From Molecular Basis to Therapy)

Abstract

Mesothelioma is characterized by the inactivation of tumor suppressor genes, with frequent mutations in neurofibromin 2 (NF2), BRCA1-associated protein 1 (BAP1), and cyclin-dependent kinase inhibitor 2A (CDKN2A). These mutations lead to disruptions in the Hippo signaling pathway and histone methylation, thereby promoting tumor growth. NF2 mutations result in Merlin deficiency, leading to uncontrolled cell proliferation, whereas BAP1 mutations impair chromatin remodeling and hinder DNA damage repair. Emerging molecular targets in mesothelioma include mesothelin (MSLN), oxytocin receptor (OXTR), protein arginine methyltransferase (PRMT5), and carbohydrate sulfotransferase 4 (CHST4). MSLN-based therapies, such as antibody–drug conjugates and immunotoxins, have shown efficacy in clinical trials. OXTR, upregulated in mesothelioma, is correlated with poor prognosis and represents a novel therapeutic target. PRMT5 inhibition is being explored in tumors with MTAP deletions, commonly co-occurring with CDKN2A loss. CHST4 expression is associated with improved prognosis, potentially influencing tumor immunity. Immune checkpoint inhibitors targeting PD-1/PD-L1 have shown promise in some cases; however, resistance mechanisms remain a challenge. Advances in multi-omics approaches have improved our understanding of mesothelioma pathogenesis. Future research will aim to identify novel therapeutic targets and personalized treatment strategies, particularly in the context of epigenetic therapy and combination immunotherapy.

1. Introduction

Mesothelioma is a universally lethal and highly aggressive cancer that mainly affects the pleura; it is increasing in incidence worldwide and has a well-established causal relationship with asbestos exposure [1,2,3]. Despite improvements in our understanding of its complex pathobiology, including extensive alterations in genetics, epigenetics, the tumor microenvironment, and immunobiology, platinum–antifolate combination chemotherapy (cisplatin and pemetrexed) remains the treatment of choice. This regimen, which is used both for unresectable cases and as part of multimodal therapy for resectable lesions, has shown a tumor response rate of approximately 45.5%, progression-free survival (PFS) of 6.1 months, and overall survival (OS) of between 8.0 and 13.3 months, with a very poor 5-year survival rate of only 5–10% [4,5]. Surgical intervention, as evidenced by the MARS 2 trial, has not conferred a considerable survival benefit and is associated with a higher incidence of grade ≥ 3 adverse events than chemotherapy alone [6]. Considering these limitations, the recent exponential growth in mesothelioma research has prompted the development of novel therapeutic strategies. Extensive genomic analyses have revealed frequent inactivation of key tumor suppressors, such as neurofibromin 2 (NF2), BRCA1-associated protein 1 (BAP1), and cyclin-dependent kinase inhibitor 2A (CDKN2A), through various mechanisms, thereby providing new targets for treatment [7]. Among emerging therapies, immunotherapy has shown particular promise; phase III trials have reported improved survival with agents such as nivolumab and ipilimumab compared with conventional chemotherapy, and neoadjuvant immunotherapy trials have further suggested potential benefits [8,9,10,11,12]. With the expansion of immunotherapeutic options, the identification of reliable biomarkers for predicting prognosis and treatment response will become critical for the optimization of future therapeutic strategies. In this review, we explore the latest advancements in our understanding of mesothelioma genetics, epigenetics, the tumor microenvironment, and immunobiology. We examine how these findings translate into clinical applications and highlight emerging therapeutic strategies under development.

2. Frequent Inactivation of Tumor Suppressor Genes in Mesothelioma

2.1. Genetic Landscape and Clinical Implications of Interpatient Heterogeneity

Chronic inflammation plays a pivotal role in the development of mesothelioma caused by asbestos and other mineral fibers. At the sites where these fibers are deposited, mesothelial cells often undergo programmed necrosis, which leads to the release of high mobility group box 1 (HMGB1). HMGB1 recruits macrophages and activates inflammasome pathways, thereby triggering the release of inflammatory cytokines and the activation of NF-κB signaling. This proinflammatory environment facilitates mesothelial cell survival, proliferation, and accumulation of mutations, ultimately contributing to the development of mesothelioma [13].
Extensive studies of the genetic landscape of mesothelioma have reported that the most frequent mutations involve the inactivation of tumor suppressor genes, which are critical regulators of cell growth and division [14,15,16]. These mutations occur through various mechanisms, including single nucleotide variants, copy number losses, gene fusions, and splicing alterations. The most commonly inactivated tumor suppressor genes in mesothelioma are NF2, BAP1, and CDKN2A. Comprehensive analyses of the mesothelioma genome revealed that key genetic alterations often disrupt pathways involved in the Hippo signaling pathway and histone methylation, both of which are essential in regulating cell proliferation, growth, and survival [16].
Despite the relatively low incidence of point mutations in genes directly associated with cancer, mesothelioma is characterized by substantial global inactivation of tumor suppressor genes via CpG methylation, a process that silences genes by adding a methyl group to cytosine nucleotides in DNA [17]. This widespread methylation serves as a key feature that distinguishes between mesothelioma and normal pleural cells. Understanding these genetic abnormalities is crucial for developing targeted therapies that inhibit tumor progression and improve patient outcomes.
One of the defining characteristics of mesothelioma is its high level of morphological and genetic heterogeneity across patients, which is reflected in its various histological subtypes. The most common subtypes are epithelioid and sarcomatoid, with the latter typically associated with poor prognosis [18]. The epithelioid subtype is generally less aggressive, whereas the sarcomatoid subtype often presents with a more invasive and rapidly progressing form of the disease. The biphasic subtype, which contains epithelioid and sarcomatoid components, also tends to have a poorer prognosis than purely epithelioid tumors [19].
Recent genomic studies have highlighted distinct genetic alterations between these subtypes, which may explain the differences in their clinical behavior [16]. For example, mutations in TP53, a gene frequently mutated across numerous cancers, are relatively rare in epithelioid mesotheliomas but are common in the biphasic and sarcomatoid subtypes, albeit still at a frequency of <10%. This suggests that TP53 plays a role in the increased aggressiveness observed in these subtypes [16]. Despite significant advancements in our understanding of the genomic differences between mesothelioma subtypes, there remains a critical gap in knowledge regarding which mutations act as clonal or subclonal drivers of the disease.

2.2. NF2 Inactivation and Hippo Signaling Pathway

The NF2 gene, located on chromosome 22q, is frequently mutated or deleted in mesothelioma [14,20]. Merlin, which is encoded by the NF2 gene, exerts its tumor-suppressive effects by controlling the expression of oncogenic genes through the activation of Hippo signaling (Figure 1). Merlin regulates contact inhibition and controls organ size by inhibiting the oncogenic transcriptional coactivators yes-associated protein 1 (YAP1) and transcriptional coactivator with PDZ-binding motif (TAZ). YAP1 encodes a transcription factor that regulates many tumorigenic genes, including survivin (also known as BIRC5), cellular inhibitor of apoptosis 1 (cIAP1; also known as BIRC2), KI67, and MYC [21]. NF2 loss results in YAP1 activation, thereby driving tumorigenesis [22]. In addition, YAP1 amplification is commonly observed in primary mesothelioma tumors [23], further supporting the role of Hippo signaling pathway disruption in mesothelioma development. Furthermore, studies have demonstrated the frequent loss of large tumor suppressor kinase 1 (LATS1) and large tumor suppressor kinase 2 (LATS2), kinases that negatively regulate YAP1, contributing to the hyperactivation of the Hippo signaling pathway [24,25]. In relation to LATS2 and YAP, AJUBA, a binding partner of LATS2, negatively regulates YAP activity through the LATS family. AJUBA activation inhibits mesothelioma cell proliferation [26]. Together, NF2 loss, YAP1 amplification, and LATS1/2 inactivation highlight the central role of this pathway in mesothelioma. Mice harboring heterozygous Nf2 mutations exhibit increased susceptibility to asbestos-induced mesothelioma than mice with wild-type Nf2 [27]. In these established tumors, mutational loss of the remaining wild-type Nf2 allele has been reported, suggesting that asbestos directly promotes Nf2 mutation [28]. Recently, VT3989, a TEAD inhibitor targeting the Hippo-YAP pathway, was well tolerated and demonstrated promising antitumor activity, including partial responses, in patients with mesothelioma and NF2-mutant tumors. The clinical benefit rate in mesothelioma was 57%, which support further development [29].
Preclinical studies have demonstrated that mesothelioma cells lacking NF2 are sensitive to inhibitors of focal adhesion kinase (FAK), which disrupts the interaction of FAK with SRC and the p85 regulatory subunit of PI3K [30]. Merlin, the protein encoded by NF2, typically inhibits FAK by preventing its phosphorylation; thus, when NF2 is lost, FAK activity increases, leading to enhanced cell invasiveness. The reintroduction of NF2 into these cells reverses this invasive behavior, suggesting that FAK targeting is a potential therapeutic strategy for NF2-deficient mesotheliomas [31,32]. In phase I studies, the FAK inhibitor GSK2256098 was shown to improve PFS in patients with mesothelioma who had low Merlin expression [33]. However, the COMMAND trial, a phase II trial of the FAK inhibitor defactinib, recruited 344 patients with mesothelioma, and low levels of Merlin did not improve disease outcomes [34].

2.3. BAP1 Inactivation and Therapeutic Potential

Approximately 65% of mesothelioma cases involve the inactivation of BAP1, a tumor suppressor gene located on chromosome 3p21.1 [35,36]. While germline mutations in BAP1 are rare, they markedly increase the risk of developing mesothelioma and other cancers. BAP1 functions as a deubiquitinase, a protein that removes ubiquitin molecules from other proteins, and is involved in processes such as DNA double-strand break repair and epigenetic regulation [37,38]. BAP1 loss disrupts these processes, making cells more vulnerable to DNA damage and other cancer-promoting factors. In mesothelioma, BAP1 inactivation is associated with alterations in the polycomb repressive complex 2 (PRC2) pathway, which regulates histone methylation, a key process in gene expression control (Figure 2). This disruption suggests that mesotheliomas with BAP1 mutations are sensitive to inhibitors targeting EZH2, a component of the PRC2 complex [39]. A phase II clinical trial has yielded promising results for tazemetostat, an EZH2 inhibitor, in mesothelioma patients with BAP1 inactivation, offering hope for targeted therapies in this patient subgroup. Further refinement of biomarkers for tazemetostat activity could help identify a subset of tumors most likely to derive prolonged benefit or shrinkage from this therapy [40].

2.4. CDKN2A Inactivation in Mesothelioma

The CDKN2A gene, located on chromosome 9p21, is the most frequently inactivated tumor suppressor gene in mesothelioma, with homozygous deletions occurring in 50–100% of cases [41,42]. This gene plays a pivotal role in regulating the cell cycle, and its loss is associated with more aggressive disease and reduced patient survival, particularly in cases of nonepithelioid mesothelioma [43,44]. In asbestos-induced mesotheliomas, CDKN2A mutations often occur after biallelic inactivation of NF2, another key tumor suppressor gene [45]. CDKN2A can also be epigenetically inactivated by methylation in approximately 19% of cases [46,47]. CDKN2A encodes two proteins, INK4A (p16) and ARF (p14 in humans), through alternative reading frames, a process by which different proteins are produced from the same gene (Figure 3) [48]. INK4A acts as an inhibitor of cyclin-dependent kinases CDK4 and CDK6, proteins involved in driving the cell cycle from the G1 to the S phase. INK4A loss removes this checkpoint, leading to unchecked cell proliferation [49]. Meanwhile, ARF interacts with the MDM2 protein, promoting its degradation and thus activating p53, a major tumor suppressor involved in the prevention of abnormal cell growth [50,51,52]. ARF loss results in increased MDM2 levels which, in turn, represses p53 activity, further contributing to tumorigenesis [53]. In addition, a fusion transcript involving YY1 and EWSR1 may indirectly suppress p53 by stabilizing the ARF–MDM2 interaction [54]. Efforts are underway to treat patients with p16-deficient mesothelioma with the CDK4/6 inhibitor abemaciclib. The MiST2 study was a single-arm, open-label, phase II clinical trial conducted at two centers in the UK. This trial reported disease control in 54% of patients within 12 weeks and is being investigated further in randomized trials as a targeted stratified therapy [55].

3. Other Targets in Mesothelioma

3.1. Mesothelin

Mesothelin (MSLN) is a 40-kDa glycoprotein, originally believed to be produced solely by mesothelioma cells; however, later investigations revealed its expression in numerous normal human tissues [56,57]. Approximately 77% of patients with epithelial mesothelioma tested positive for MSLN, a finding not observed in sarcomatoid tumors [58,59]. To measure MSLN levels, several blood assays that detect soluble MSLN-related peptides have been developed, among which MESOMARK [60] and the N-ERC/MSLN test [61] are the most prominent. Although these biomarkers alone are insufficient for diagnosing mesothelioma, researchers are optimistic that combining MSLN assays with tests for additional markers, such as calretinin, could enhance the detection accuracy [62].
Given the high expression of MSLN in epithelial mesothelioma, a range of innovative treatment strategies are currently being evaluated in various phases of clinical trials. These emerging therapies include the anti-MSLN immunotoxin SS1P, the monoclonal antibody amatuximab (MORAb-009) [63], and the antibody–drug conjugate anetumab ravtansine (BAY 94-9343) [64]. After a phase I study established the safety of SS1P, its combination with pemetrexed and cisplatin was tested in 24 chemotherapy-naïve patients with unrespectable mesothelioma, which led to an objective response rate (ORR) of 77% [65]. Amatuximab, a mouse–human chimeric monoclonal antibody that selectively binds to MSLN, mainly blocks the interaction between MSLN and the antigen CA125/MUC16 [66]. In a phase II trial, 89 patients with unresectable mesothelioma received a regimen combining amatuximab with pemetrexed and cisplatin. Although this approach did not improve the PFS of patients compared with controls, it yielded promising outcomes, with a median OS of 14.8 months and an ORR of 39% [63]. Anetumab ravtansine (BAY 94-9343) is a novel, highly potent, and selective antibody–drug conjugate composed of a human anti-MSLN antibody associated with the maytansinoid tubulin inhibitor DM4 through a disulfide-containing linker, with an average of three lysyl conjugations [67]. In vivo studies have shown that anetumab ravtansine specifically localizes to MSLN-positive tumors and effectively inhibits tumor growth in subcutaneous and orthotopic xenograft models [68]. Moreover, it can exert a bystander effect on adjacent tumor cells that do not express MSLN. Promising results were reported in a study of 148 patients with mesothelioma, ovarian cancer, and other malignancies, in which anetumab ravtansine targeted MSLN-expressing tumors [69]. Following these outcomes, a randomized trial comparing the combination of anetumab ravtansine and pembrolizumab with pembrolizumab alone was conducted; however, no significant differences were observed in response rates or progression-free survival between the two groups. Notably, high levels of soluble MSLN—which can neutralize anti-MSLN antibodies—were associated with poorer survival in patients receiving anetumab ravtansine [70].

3.2. Oxytocin Receptor

The oxytocin receptor (OXTR), a G-protein-coupled receptor, was recently significantly upregulated in mesothelioma cell lines compared with other cancer types. Cases with elevated OXTR expression have poor clinical outcomes [71]. Typically, OXTR is mainly expressed in tissues such as the mammary glands and uterine myometrium at the end of pregnancy, where it facilitates milk release and uterine contractions [72]. OXTR is also present in the central nervous system, influencing various behaviors [73]. Notably, OXTR expression surges in the uterine myometrium toward the end of pregnancy, although oxytocin secretion remains relatively stable [72]. It has been demonstrated that OXTR expression is regulated by interleukin (IL)-6 and IL-1β, but the addition of these cytokines to mesothelioma cell lines with low OXTR expression did not markedly increase OXTR mRNA levels [74]. Analysis of mesothelioma cell lines and The Cancer Genome Atlas (TCGA) data revealed a significant correlation between OXTR mRNA expression and NF2 gene inactivation. The experimental results further confirmed that NF2 knockdown increased OXTR expression, suggesting that NF2 negatively regulates OXTR transcription [71]. Reduction of OXTR expression notably inhibited the proliferation of mesothelioma cell lines with high OXTR levels by disrupting the tumor cell cycle. Furthermore, OXTR antagonists reduced mesothelioma cell growth, and oral administration of the OXTR antagonist cligosiban hindered mesothelioma tumor progression. These findings point to the role of OXTR in mesothelioma cell proliferation and its potential as a therapeutic target.

3.3. PRMT5

The protein arginine methyltransferase 5 (PRMT5) is a methyltransferase enzyme that works in conjunction with its cofactor methylosome protein 50 and uses S-adenosyl methionine (SAM) as a methyl donor to symmetrically dimethylate arginine residues on target proteins [75,76]. This posttranslational modification plays a pivotal role in several cellular processes, such as RNA splicing, transcription, and translation. PRMT5 has been identified as a synthetic lethal target in cancers with a homozygous deletion of the MTAP gene, which is commonly co-deleted with the tumor suppressor gene CDKN2A [77]. MTAP gene deletions are frequently observed in various cancers, including mesothelioma [16,78]. Typically, MTAP catalyzes the breakdown of 5′-deoxy-5′-methylthioadenosine (MTA), a metabolite that competes with SAM for binding to PRMT5 and acts as a moderate inhibitor of the ability of PRMT5 to add symmetric dimethylarginine (SDMA) modifications to its protein targets. MTAP loss leads to MTA accumulation, making MTAP-deficient cancer cells susceptible to PRMT5 inhibition [79]. PRMT5 inhibitors exerted promising effects on in vitro models of mesothelioma with MTAP deletion, as its inhibition selectively inhibited tumor cell proliferation and downregulated genes related to cell cycle and epithelial–mesenchymal transition [80]. MRTX1719, developed using a structure-based drug design, is a small-molecule inhibitor that targets the PRMT5/MTA complex. The drug is currently being evaluated in a phase I/II clinical trial (NCT05245500) for its safety and effectiveness against advanced solid tumors with MTAP deletions. Early findings from this trial have indicated six confirmed objective responses, suggesting that MRTX1719 can effectively inhibit SDMA production at well-tolerated doses, leading to substantial tumor shrinkage [81].

3.4. CHST4

Recently, the MARS 2 trial demonstrated that surgery did not offer all patients significant survival advantages compared with chemotherapy alone, although this trial has been criticized in various aspects [6]. Consequently, there is a growing demand for biomarkers that can predict postoperative outcomes and help tailor therapeutic strategies for each patient with mesothelioma. Through an extensive survival analysis of the TCGA mesothelioma dataset, the carbohydrate sulfotransferase 4 (CHST4) gene emerged as a promising indicator of favorable OS in patients with mesothelioma [82]. In this study, enrichment analysis of genes associated with favorable prognosis, including CHST4, revealed immune-related ontological terms, whereas analysis of genes associated with unfavorable prognosis highlighted cell-cycle-related terms. Notably, CHST4 mRNA expression in mesothelioma demonstrated a significant correlation with immune–gene signatures representing tumor-infiltrating lymphocytes [83]. As a sulfotransferase expressed in high endothelial venules [84], CHST4 plays a pivotal role in the immune system by facilitating the homing of lymphocytes to lymphoid organs (Figure 4) [85]. It is hypothesized that CHST4 contributes to tumor immunity against malignant tumors. Further functional analysis of this gene may advance the development of immunotherapeutic strategies.
To explore the relationship between CHST4 expression and patient outcomes, researchers examined CHST4 protein levels via immunohistochemistry in 23 surgical samples from patients with mesothelioma who underwent complete macroscopic resection. The researchers quantified CHST4 expression using a scoring system that incorporated the percentage of stained cells and staining intensity. The analysis revealed that patients with higher CHST4 scores experienced significantly better postoperative outcomes, with a median OS of 107.8 months compared with 38.0 months in those with lower scores [82]. Although pleural thickness is now reflected in the ninth edition of the TNM classification [86] and has been reported as a risk factor for postoperative complications [87], it was not correlated with the intensity of CHST4 expression, suggesting that CHST4 may serve as an independent prognostic marker.

4. Effect of Immunotherapy on Mesothelioma

4.1. Tumor Microenvironment in Mesothelioma

In mesothelioma, the tumor microenvironment (TME) is a key factor influencing tumor development, immune suppression, and treatment resistance [88]. Chronic inflammation caused by asbestos exposure shapes a distinctive TME populated by immunosuppressive elements, such as tumor-associated macrophages (TAMs), regulatory T cells, cancer-associated fibroblasts, and dysfunctional T cells. Among these, M2-like TAMs are particularly prevalent and promote tumor progression through the release of inhibitory cytokines, such as IL-10 and TGF-β, correlating with unfavorable clinical outcomes [89,90]. The immunosuppressive landscape is further reinforced by the adenosine signaling pathway, which dampens immune cell activity and supports TAM proliferation [91]. Notably, the composition and immune features of the TME vary across different mesothelioma histological subtypes. Nonepithelioid subtypes, such as sarcomatoid and biphasic mesothelioma, are generally associated with a more immunosuppressive microenvironment, characterized by high expression of immune checkpoint molecules, including PD-L1, PD-L2, TIM-3, and CTLA-4 [92]. Overall, the interplay of stromal and immune cells in the TME significantly contributes to the aggressiveness and treatment resistance of mesothelioma, highlighting the importance of developing treatment strategies that particularly target the TME [93].

4.2. Chemoimmunotherapy

Recent advances in immune checkpoint inhibition, particularly targeting molecules such as cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PD-1), and programmed death ligand 1 (PD-L1), have proven successful in enhancing immune responses and T cell function across various malignancies [94]. Based on encouraging outcomes in patients with relapsed disease [8], the combination therapy of nivolumab and ipilimumab, which specifically target PD-1 and CTLA4, respectively, has shown superior efficacy compared to standard chemotherapy as a first-line treatment (median survival: 18.1 vs. 14.1 months), particularly among patients with nonepithelioid mesothelioma (CheckMate 743) [11]. Consequently, frontline treatment with ipilimumab and nivolumab is strongly recommended for patients with biphasic or sarcomatoid mesothelioma. This predefined interim analysis supported the US FDA’s approval of this combination therapy in 2020, marking it as the first FDA-approved systemic treatment for mesothelioma since 2004 [9]. The impact of this regimen on older or frailer patients as compared with clinical trial participants remains an area of interest.
Table 1 presents recent clinical trials of first-line and salvage immunotherapy conducted for mesothelioma. To date, many phase III trials on maintenance therapy—long-term treatment aimed at delaying relapse after induction chemotherapy—have shown considerable improvements in OS [34,95,96]. However, in certain cases, rechallenging with platinum–pemetrexed chemotherapy has shown potential benefits [97]. In addition, the combination of immune checkpoint inhibitors (ICIs) and chemotherapy has exerted a synergistic effect and is currently recognized as a standard of care for non-small cell lung cancer [98]. KEYNOTE-028, pembrolizumab therapy for previously treated patients with PD-L1-positive, also showed well-tolerated findings [99]. Consequently, randomized trials are currently evaluating the efficacy of PD-1 or PD-L1 inhibitors in combination with chemotherapy for mesothelioma, with promising results reported in the Canadian Cancer Trials Group (NCT02784171) [100]. In addition, phase II trials, such as PrECOG0505 (NCT02899195) [101] and DREAM [102], have laid the groundwork for the DREAM3R trial, an international phase III study that compared standard cisplatin–pemetrexed therapy with and without durvalumab, a PD-L1 inhibitor [103].
Vascular endothelial growth factor (VEGF) plays a key role in mesothelioma by promoting tumor angiogenesis and immune suppression. It supports tumor growth and aids the tumor in evading immune responses by recruiting regulatory immune cells and increasing PD-L1 expression. VEGF targeting may inhibit tumor progression and immune evasion [104]. Findings from the Mesothelioma Avastin Cisplatin–Pemetrexed Study (MAPS) indicate that antiangiogenic agents enhance immune cell differentiation and activity [105], providing the rationale for the phase III BEAT-meso trial (NCT03762018), which is assessing the addition of the PD-L1 inhibitor atezolizumab to bevacizumab and standard chemotherapy (ABC) [106]. In this study, a notable improvement in median PFS for ABC was observed, which led to a numerical increase in median OS, although it was not statistically significant. In the prespecified histology-based analysis, ABC showed superior OS and PFS in nonepithelioid cases compared with BC.
Among recent salvage immunotherapy trials, the Japanese MERIT trial holds historical importance as the first to obtain regulatory approval for a salvage checkpoint inhibitor [107]. In contrast, the DETERMINE trial, which investigated tremelimumab monotherapy, revealed that single-agent CTLA-4 blockade showed no benefit for mesothelioma [95]. Similarly, the PROMISE-Meso trial failed to demonstrate a survival advantage for pembrolizumab over investigator-selected chemotherapy (gemcitabine or vinorelbine) in PD-L1–positive patients [96]. However, the CONFIRM trial demonstrated that nivolumab markedly improved PFS and OS compared with placebo [108]. The administration of adenovirus-mediated interferon alfa-2b therapy is currently under evaluation in the phase III INFINITE trial, following promising phase II data showing a disease control rate of 88% [109]. This trial involves previously treated patients randomly assigned to receive intrapleural adenovirus therapy followed by either celecoxib and gemcitabine or celecoxib and gemcitabine alone, with treatment continuing until disease progression or treatment termination due to toxicity. These findings suggest that a subset of mesothelioma patients derives substantial benefits from immunotherapy. Until predictive biomarkers are further refined, all mesothelioma patients are recommended to receive an immunotherapy regimen at some stage of their treatment.

4.3. Cellular Therapy

Immune system modulation provides potential for more sustained disease control compared with chemotherapy and results in fewer toxic side effects. Researchers have investigated innovative approaches to altering immune system behavior. One such strategy involves genetically engineered T cells, known as chimeric antigen receptor T (CAR-T) cells, designed to specifically target MSLN—an antigen found mainly, although not exclusively, in mesothelioma cells [110,111,112]. A phase I clinical trial that investigated the intrapleural administration of these CAR-T cells in combination with an ICI to 18 patients with mesothelioma reported a median OS of 23.9 months, which is an exciting finding in such an early-phase study [113].

5. Conclusions

Recent advances in mesothelioma treatment—ranging from innovative medical devices to novel immunotherapy combinations—have injected renewed momentum into this field. However, despite these promising developments, improvements in patient survival remain modest, owing to the limited number of large-scale randomized trials and considerable interpatient heterogeneity. Accurate identification of histological subtypes is paramount for tailoring frontline systemic therapy, whereas ongoing clinical investigations continue to explore expanded options for patients with unresectable diseases. Notably, challenges persist in patients refractory to current immunotherapeutic approaches. To overcome these hurdles, future research must prioritize well-controlled, multicenter trials, the adoption of individualized treatment strategies, and the development of master protocols that facilitate global collaboration. Furthermore, the increasing incidence of mesothelioma in developing countries highlights the urgent need for effective, accessible, and scalable treatment solutions. Ultimately, a multidisciplinary approach that integrates molecular insights with clinical innovation is necessary to translate these advances into meaningful improvements in patient outcomes.

Author Contributions

Conceptualization, T.K., and I.T.; methodology, T.K., and I.T.; validation, T.K., I.T., H.H., and S.O.; formal analysis, T.K., I.T., H.H., and S.O.; data curation, Y.I., Y.N., H.T., H.W., Y.K (Yuta Kawasumi), Y.K. (Yuka Kadomatsu), K.N., H.U., S.N., and T.M.; writing—original draft preparation, T.K.; writing—review and editing, T.K., and I.T.; supervision, T.F.C.-Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from Aichi Cancer Research Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zhai, Z.; Ruan, J.; Zheng, Y.; Xiang, D.; Li, N.; Hu, J.; Shen, J.; Deng, Y.; Yao, J.; Zhao, P.; et al. Assessment of Global Trends in the Diagnosis of Mesothelioma From 1990 to 2017. JAMA Netw. Open 2021, 4, e2120360. [Google Scholar] [CrossRef]
  2. Huang, J.; Chan, S.C.; Pang, W.S.; Chow, S.H.; Lok, V.; Zhang, L.; Lin, X.; Lucero-Prisno, D.E., 3rd; Xu, W.; Zheng, Z.J.; et al. Global Incidence, Risk Factors, and Temporal Trends of Mesothelioma: A Population-Based Study. J. Thorac. Oncol. 2023, 18, 792–802. [Google Scholar] [CrossRef]
  3. Milano, M.T.; Zhang, H. Malignant pleural mesothelioma: A population-based study of survival. J. Thorac. Oncol. 2010, 5, 1841–1848. [Google Scholar] [CrossRef]
  4. Vogelzang, N.J.; Rusthoven, J.J.; Symanowski, J.; Denham, C.; Kaukel, E.; Ruffie, P.; Gatzemeier, U.; Boyer, M.; Emri, S.; Manegold, C.; 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, 2636–2644. [Google Scholar] [CrossRef] [PubMed]
  5. Yap, T.A.; Aerts, J.G.; Popat, S.; Fennell, D.A. Novel insights into mesothelioma biology and implications for therapy. Nat. Rev. Cancer 2017, 17, 475–488. [Google Scholar] [CrossRef]
  6. Lim, E.; Waller, D.; Lau, K.; Steele, J.; Pope, A.; Ali, C.; Bilancia, R.; Keni, M.; Popat, S.; O’Brien, M.; et al. Extended pleurectomy decortication and chemotherapy versus chemotherapy alone for pleural mesothelioma (MARS 2): A phase 3 randomised controlled trial. Lancet Respir. Med. 2024, 12, 457–466. [Google Scholar] [CrossRef] [PubMed]
  7. Sato, T.; Sekido, Y. NF2/Merlin Inactivation and Potential Therapeutic Targets in Mesothelioma. Int. J. Mol. Sci. 2018, 19, 988. [Google Scholar] [CrossRef] [PubMed]
  8. Scherpereel, A.; Mazieres, J.; Greillier, L.; Lantuejoul, S.; Do, P.; Bylicki, O.; Monnet, I.; Corre, R.; Audigier-Valette, C.; Locatelli-Sanchez, M.; et al. Nivolumab or nivolumab plus ipilimumab in patients with relapsed malignant pleural mesothelioma (IFCT-1501 MAPS2): A multicentre, open-label, randomised, non-comparative, phase 2 trial. Lancet Oncol. 2019, 20, 239–253. [Google Scholar] [CrossRef]
  9. Baas, P.; Scherpereel, A.; Nowak, A.K.; Fujimoto, N.; Peters, S.; Tsao, A.S.; Mansfield, A.S.; Popat, S.; Jahan, T.; Antonia, S.; et al. First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): A multicentre, randomised, open-label, phase 3 trial. Lancet 2021, 397, 375–386. [Google Scholar] [CrossRef]
  10. Tsao, A.S.; Lindwasser, O.W.; Adjei, A.A.; Adusumilli, P.S.; Beyers, M.L.; Blumenthal, G.M.; Bueno, R.; Burt, B.M.; Carbone, M.; Dahlberg, S.E.; et al. Current and Future Management of Malignant Mesothelioma: A Consensus Report from the National Cancer Institute Thoracic Malignancy Steering Committee, International Association for the Study of Lung Cancer, and Mesothelioma Applied Research Foundation. J. Thorac. Oncol. 2018, 13, 1655–1667. [Google Scholar] [CrossRef]
  11. Peters, S.; Scherpereel, A.; Cornelissen, R.; Oulkhouir, Y.; Greillier, L.; Kaplan, M.A.; Talbot, T.; Monnet, I.; Hiret, S.; Baas, P.; et al. First-line nivolumab plus ipilimumab versus chemotherapy in patients with unresectable malignant pleural mesothelioma: 3-year outcomes from CheckMate 743. Ann. Oncol. 2022, 33, 488–499. [Google Scholar] [CrossRef] [PubMed]
  12. Bylicki, O.; Guisier, F.; Scherpereel, A.; Daniel, C.; Swalduz, A.; Grolleau, E.; Bernardi, M.; Hominal, S.; Prevost, J.B.; Pamart, G.; et al. Real-World efficacy and safety of combination nivolumab plus ipilimumab for Untreated, Unresectable, pleural Mesothelioma: The Meso-Immune (GFPC 04-2021) trial. Lung Cancer 2024, 194, 107866. [Google Scholar] [CrossRef]
  13. Gaudino, G.; Xue, J.; Yang, H. How asbestos and other fibers cause mesothelioma. Transl. Lung Cancer Res. 2020, 9, S39–S46. [Google Scholar] [CrossRef] [PubMed]
  14. Guo, G.; Chmielecki, J.; Goparaju, C.; Heguy, A.; Dolgalev, I.; Carbone, M.; Seepo, S.; Meyerson, M.; Pass, H.I. Whole-exome sequencing reveals frequent genetic alterations in BAP1, NF2, CDKN2A, and CUL1 in malignant pleural mesothelioma. Cancer Res. 2015, 75, 264–269. [Google Scholar] [CrossRef] [PubMed]
  15. Lo Iacono, M.; Monica, V.; Righi, L.; Grosso, F.; Libener, R.; Vatrano, S.; Bironzo, P.; Novello, S.; Musmeci, L.; Volante, M.; et al. Targeted next-generation sequencing of cancer genes in advanced stage malignant pleural mesothelioma: A retrospective study. J. Thorac. Oncol. 2015, 10, 492–499. [Google Scholar] [CrossRef]
  16. Bueno, R.; Stawiski, E.W.; Goldstein, L.D.; Durinck, S.; De Rienzo, A.; Modrusan, Z.; Gnad, F.; Nguyen, T.T.; Jaiswal, B.S.; Chirieac, L.R.; et al. Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nat. Genet. 2016, 48, 407–416. [Google Scholar] [CrossRef]
  17. Christensen, B.C.; Houseman, E.A.; Poage, G.M.; Godleski, J.J.; Bueno, R.; Sugarbaker, D.J.; Wiencke, J.K.; Nelson, H.H.; Marsit, C.J.; Kelsey, K.T. Integrated profiling reveals a global correlation between epigenetic and genetic alterations in mesothelioma. Cancer Res. 2010, 70, 5686–5694. [Google Scholar] [CrossRef]
  18. Fennell, D.A.; Parmar, A.; Shamash, J.; Evans, M.T.; Sheaff, M.T.; Sylvester, R.; Dhaliwal, K.; Gower, N.; Steele, J.; Rudd, R. Statistical validation of the EORTC prognostic model for malignant pleural mesothelioma based on three consecutive phase II trials. J. Clin. Oncol. 2005, 23, 184–189. [Google Scholar] [CrossRef]
  19. Muruganandan, S.; Alfonso, H.; Franklin, P.; Shilkin, K.; Segal, A.; Olsen, N.; Reid, A.; de Klerk, N.; Musk, A.B.; Brims, F. Comparison of outcomes following a cytological or histological diagnosis of malignant mesothelioma. Br. J. Cancer. 2017, 116, 703–708. [Google Scholar] [CrossRef]
  20. Sekido, Y. Targeting the Hippo Pathway Is a New Potential Therapeutic Modality for Malignant Mesothelioma. Cancers 2018, 10, 90. [Google Scholar] [CrossRef]
  21. Yu, F.X.; Zhao, B.; Guan, K.L. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell 2015, 163, 811–828. [Google Scholar] [CrossRef] [PubMed]
  22. Mizuno, T.; Murakami, H.; Fujii, M.; Ishiguro, F.; Tanaka, I.; Kondo, Y.; Akatsuka, S.; Toyokuni, S.; Yokoi, K.; Osada, H.; et al. YAP induces malignant mesothelioma cell proliferation by upregulating transcription of cell cycle-promoting genes. Oncogene 2012, 31, 5117–5122. [Google Scholar] [CrossRef] [PubMed]
  23. Wu, J.; Minikes, A.M.; Gao, M.; Bian, H.; Li, Y.; Stockwell, B.R.; Chen, Z.N.; Jiang, X. Intercellular interaction dictates cancer cell ferroptosis via NF2-YAP signalling. Nature 2019, 572, 402–406. [Google Scholar] [CrossRef]
  24. Murakami, H.; Mizuno, T.; Taniguchi, T.; Fujii, M.; Ishiguro, F.; Fukui, T.; Akatsuka, S.; Horio, Y.; Hida, T.; Kondo, Y.; et al. LATS2 is a tumor suppressor gene of malignant mesothelioma. Cancer Res. 2011, 71, 873–883. [Google Scholar] [CrossRef] [PubMed]
  25. Miyanaga, A.; Masuda, M.; Tsuta, K.; Kawasaki, K.; Nakamura, Y.; Sakuma, T.; Asamura, H.; Gemma, A.; Yamada, T. Hippo pathway gene mutations in malignant mesothelioma: Revealed by RNA and targeted exon sequencing. J. Thorac. Oncol. 2015, 10, 844–851. [Google Scholar] [CrossRef]
  26. Tanaka, I.; Osada, H.; Fujii, M.; Fukatsu, A.; Hida, T.; Horio, Y.; Kondo, Y.; Sato, A.; Hasegawa, Y.; Tsujimura, T.; et al. LIM-domain protein AJUBA suppresses malignant mesothelioma cell proliferation via Hippo signaling cascade. Oncogene 2015, 34, 73–83. [Google Scholar] [CrossRef]
  27. Altomare, D.A.; Vaslet, C.A.; Skele, K.L.; De Rienzo, A.; Devarajan, K.; Jhanwar, S.C.; McClatchey, A.I.; Kane, A.B.; Testa, J.R. A mouse model recapitulating molecular features of human mesothelioma. Cancer Res. 2005, 65, 8090–8095. [Google Scholar] [CrossRef]
  28. Fleury-Feith, J.; Lecomte, C.; Renier, A.; Matrat, M.; Kheuang, L.; Abramowski, V.; Levy, F.; Janin, A.; Giovannini, M.; Jaurand, M.C. Hemizygosity of Nf2 is associated with increased susceptibility to asbestos-induced peritoneal tumours. Oncogene 2003, 22, 3799–3805. [Google Scholar] [CrossRef]
  29. Yap, T.A.; Kwiatkowski, D.J.; Desai, J.; Dangogo-Jack, I.; Millward, M.; Kindler, H.L.; Tolcher, A.W.; Frentzas, S.; Thurston, A.W.; Post, L.; et al. First-in-class, first-in-human phase 1 trial of VT3989, an inhibitor of yes-associated protein (YAP)/transcriptional enhancer activator domain (TEAD), in patients (pts) with advanced solid tumors enriched for malignant mesothelioma and other tumors with neurofibromatosis 2 (NF2) mutations. Cancer Res. 2023, 83 (Suppl. 8), CT006. [Google Scholar]
  30. Provenzano, L.; Ryan, Y.; Hilton, D.A.; Lyons-Rimmer, J.; Dave, F.; Maze, E.A.; Adams, C.L.; Rigby-Jones, R.; Ammoun, S.; Hanemann, C.O. Cellular prion protein (PrP(C)) in the development of Merlin-deficient tumours. Oncogene 2017, 36, 6132–6142. [Google Scholar] [CrossRef]
  31. Kato, T.; Sato, T.; Yokoi, K.; Sekido, Y. E-cadherin expression is correlated with focal adhesion kinase inhibitor resistance in Merlin-negative malignant mesothelioma cells. Oncogene 2017, 36, 5522–5531. [Google Scholar] [CrossRef]
  32. Shapiro, I.M.; Kolev, V.N.; Vidal, C.M.; Kadariya, Y.; Ring, J.E.; Wright, Q.; Weaver, D.T.; Menges, C.; Padval, M.; McClatchey, A.I.; et al. Merlin deficiency predicts FAK inhibitor sensitivity: A synthetic lethal relationship. Sci. Transl. Med. 2014, 6, 237ra268. [Google Scholar] [CrossRef] [PubMed]
  33. Soria, J.C.; Gan, H.K.; Blagden, S.P.; Plummer, R.; Arkenau, H.T.; Ranson, M.; Evans, T.R.; Zalcman, G.; Bahleda, R.; Hollebecque, A.; et al. A phase I, pharmacokinetic and pharmacodynamic study of GSK2256098, a focal adhesion kinase inhibitor, in patients with advanced solid tumors. Ann. Oncol. 2016, 27, 2268–2274. [Google Scholar] [CrossRef] [PubMed]
  34. Fennell, D.A.; Baas, P.; Taylor, P.; Nowak, A.K.; Gilligan, D.; Nakano, T.; Pachter, J.A.; Weaver, D.T.; Scherpereel, A.; Pavlakis, N.; et al. Maintenance Defactinib Versus Placebo After First-Line Chemotherapy in Patients With Merlin-Stratified Pleural Mesothelioma: COMMAND-A Double-Blind, Randomized, Phase II Study. J. Clin. Oncol. 2019, 37, 790–798. [Google Scholar] [CrossRef] [PubMed]
  35. Dey, A.; Seshasayee, D.; Noubade, R.; French, D.M.; Liu, J.; Chaurushiya, M.S.; Kirkpatrick, D.S.; Pham, V.C.; Lill, J.R.; Bakalarski, C.E.; et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science 2012, 337, 1541–1546. [Google Scholar] [CrossRef]
  36. Kadariya, Y.; Cheung, M.; Xu, J.; Pei, J.; Sementino, E.; Menges, C.W.; Cai, K.Q.; Rauscher, F.J.; Klein-Szanto, A.J.; Testa, J.R. Bap1 Is a Bona Fide Tumor Suppressor: Genetic Evidence from Mouse Models Carrying Heterozygous Germline Bap1 Mutations. Cancer Res. 2016, 76, 2836–2844. [Google Scholar] [CrossRef]
  37. Ismail, I.H.; Davidson, R.; Gagne, J.P.; Xu, Z.Z.; Poirier, G.G.; Hendzel, M.J. Germline mutations in BAP1 impair its function in DNA double-strand break repair. Cancer Res. 2014, 74, 4282–4294. [Google Scholar] [CrossRef]
  38. Yu, H.; Pak, H.; Hammond-Martel, I.; Ghram, M.; Rodrigue, A.; Daou, S.; Barbour, H.; Corbeil, L.; Hebert, J.; Drobetsky, E.; et al. Tumor suppressor and deubiquitinase BAP1 promotes DNA double-strand break repair. Proc. Natl. Acad. Sci. USA 2014, 111, 285–290. [Google Scholar] [CrossRef]
  39. Kemp, C.D.; Rao, M.; Xi, S.; Inchauste, S.; Mani, H.; Fetsch, P.; Filie, A.; Zhang, M.; Hong, J.A.; Walker, R.L.; et al. Polycomb repressor complex-2 is a novel target for mesothelioma therapy. Clin. Cancer Res. 2012, 18, 77–90. [Google Scholar] [CrossRef]
  40. Zauderer, M.G.; Szlosarek, P.W.; Le Moulec, S.; Popat, S.; Taylor, P.; Planchard, D.; Scherpereel, A.; Koczywas, M.; Forster, M.; Cameron, R.B.; et al. EZH2 inhibitor tazemetostat in patients with relapsed or refractory, BAP1-inactivated malignant pleural mesothelioma: A multicentre, open-label, phase 2 study. Lancet Oncol. 2022, 23, 758–767. [Google Scholar] [CrossRef]
  41. Marshall, K.; Jackson, S.; Jones, J.; Holme, J.; Lyons, J.; Barrett, E.; Taylor, P.; Bishop, P.; Hodgson, C.; Green, M.; et al. Homozygous deletion of CDKN2A in malignant mesothelioma: Diagnostic utility, patient characteristics and survival in a UK mesothelioma centre. Lung Cancer 2020, 150, 195–200. [Google Scholar] [CrossRef] [PubMed]
  42. Vandenhoeck, J.; van Meerbeeck, J.P.; Fransen, E.; Raskin, J.; Van Camp, G.; Op de Beeck, K.; Lamote, K. DNA Methylation as a Diagnostic Biomarker for Malignant Mesothelioma: A Systematic Review and Meta-Analysis. J. Thorac. Oncol. 2021, 16, 1461–1478. [Google Scholar] [CrossRef]
  43. Hida, T.; Matsumoto, S.; Hamasaki, M.; Kawahara, K.; Tsujimura, T.; Hiroshima, K.; Kamei, T.; Taguchi, K.; Iwasaki, A.; Oda, Y.; et al. Deletion status of p16 in effusion smear preparation correlates with that of underlying malignant pleural mesothelioma tissue. Cancer Sci. 2015, 106, 1635–1641. [Google Scholar] [CrossRef]
  44. Jennings, C.J.; Murer, B.; O’Grady, A.; Hearn, L.M.; Harvey, B.J.; Kay, E.W.; Thomas, W. Differential p16/INK4A cyclin-dependent kinase inhibitor expression correlates with chemotherapy efficacy in a cohort of 88 malignant pleural mesothelioma patients. Br. J. Cancer 2015, 113, 69–75. [Google Scholar] [CrossRef] [PubMed]
  45. Jongsma, J.; van Montfort, E.; Vooijs, M.; Zevenhoven, J.; Krimpenfort, P.; van der Valk, M.; van de Vijver, M.; Berns, A. A conditional mouse model for malignant mesothelioma. Cancer Cell 2008, 13, 261–271. [Google Scholar] [CrossRef] [PubMed]
  46. Wong, L.; Zhou, J.; Anderson, D.; Kratzke, R.A. Inactivation of p16INK4a expression in malignant mesothelioma by methylation. Lung Cancer 2002, 38, 131–136. [Google Scholar] [CrossRef]
  47. Singhi, A.D.; Krasinskas, A.M.; Choudry, H.A.; Bartlett, D.L.; Pingpank, J.F.; Zeh, H.J.; Luvison, A.; Fuhrer, K.; Bahary, N.; Seethala, R.R.; et al. The prognostic significance of BAP1, NF2, and CDKN2A in malignant peritoneal mesothelioma. Mod. Pathol. 2016, 29, 14–24. [Google Scholar] [CrossRef]
  48. Chernova, T.; Murphy, F.A.; Galavotti, S.; Sun, X.M.; Powley, I.R.; Grosso, S.; Schinwald, A.; Zacarias-Cabeza, J.; Dudek, K.M.; Dinsdale, D.; et al. Long-Fiber Carbon Nanotubes Replicate Asbestos-Induced Mesothelioma with Disruption of the Tumor Suppressor Gene Cdkn2a (Ink4a/Arf). Curr. Biol. 2017, 27, 3302–3314. [Google Scholar] [CrossRef]
  49. Frizelle, S.P.; Kratzke, M.G.; Carreon, R.R.; Engel, S.C.; Youngquist, L.; Klein, M.A.; Fourre, L.; Shekels, L.L.; Kratzke, R.A. Inhibition of both mesothelioma cell growth and Cdk4 activity following treatment with a TATp16INK4a peptide. Anticancer Res. 2008, 28, 1–7. [Google Scholar]
  50. Nguyen, T.T.T.; Shingyoji, M.; Hanazono, M.; Zhong, B.; Morinaga, T.; Tada, Y.; Shimada, H.; Hiroshima, K.; Tagawa, M. An MDM2 inhibitor achieves synergistic cytotoxic effects with adenoviruses lacking E1B55kDa gene on mesothelioma with the wild-type p53 through augmenting NFI expression. Cell Death Dis. 2021, 12, 663. [Google Scholar] [CrossRef]
  51. Singh, A.; Bhattacharyya, N.; Srivastava, A.; Pruett, N.; Ripley, R.T.; Schrump, D.S.; Hoang, C.D. MicroRNA-215-5p Treatment Suppresses Mesothelioma Progression via the MDM2-p53-Signaling Axis. Mol. Ther. 2019, 27, 1665–1680. [Google Scholar] [CrossRef] [PubMed]
  52. Ou, W.B.; Lu, M.; Eilers, G.; Li, H.; Ding, J.; Meng, X.; Wu, Y.; He, Q.; Sheng, Q.; Zhou, H.M.; et al. Co-targeting of FAK and MDM2 triggers additive anti-proliferative effects in mesothelioma via a coordinated reactivation of p53. Br. J. Cancer 2016, 115, 1253–1263. [Google Scholar] [CrossRef] [PubMed]
  53. Walter, R.F.; Mairinger, F.D.; Ting, S.; Vollbrecht, C.; Mairinger, T.; Theegarten, D.; Christoph, D.C.; Schmid, K.W.; Wohlschlaeger, J. MDM2 is an important prognostic and predictive factor for platin-pemetrexed therapy in malignant pleural mesotheliomas and deregulation of P14/ARF (encoded by CDKN2A) seems to contribute to an MDM2-driven inactivation of P53. Br. J. Cancer 2015, 112, 883–890. [Google Scholar] [CrossRef] [PubMed]
  54. Sui, G.; Affar el, B.; Shi, Y.; Brignone, C.; Wall, N.R.; Yin, P.; Donohoe, M.; Luke, M.P.; Calvo, D.; Grossman, S.R.; et al. Yin Yang 1 is a negative regulator of p53. Cell 2004, 117, 859–872. [Google Scholar] [CrossRef]
  55. Fennell, D.A.; King, A.; Mohammed, S.; Greystoke, A.; Anthony, S.; Poile, C.; Nusrat, N.; Scotland, M.; Bhundia, V.; Branson, A.; et al. Abemaciclib in patients with p16ink4A-deficient mesothelioma (MiST2): A single-arm, open-label, phase 2 trial. Lancet Oncol. 2022, 23, 374–381. [Google Scholar] [CrossRef]
  56. Servais, E.L.; Colovos, C.; Rodriguez, L.; Bograd, A.J.; Nitadori, J.; Sima, C.; Rusch, V.W.; Sadelain, M.; Adusumilli, P.S. 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, 2478–2489. [Google Scholar] [CrossRef]
  57. Pastan, I.; Hassan, R. Discovery of mesothelin and exploiting it as a target for immunotherapy. Cancer Res. 2014, 74, 2907–2912. [Google Scholar] [CrossRef]
  58. Kushitani, K.; Takeshima, Y.; Amatya, V.J.; Furonaka, O.; Sakatani, A.; Inai, K. Immunohistochemical marker panels for distinguishing between epithelioid mesothelioma and lung adenocarcinoma. Pathol. Int. 2007, 57, 190–199. [Google Scholar] [CrossRef]
  59. Pantazopoulos, I.; Boura, P.; Xanthos, T.; Syrigos, K. Effectiveness of mesothelin family proteins and osteopontin for malignant mesothelioma. Eur. Respir. J. 2013, 41, 706–715. [Google Scholar] [CrossRef]
  60. Hollevoet, K.; Reitsma, J.B.; Creaney, J.; Grigoriu, B.D.; Robinson, B.W.; Scherpereel, A.; Cristaudo, A.; Pass, H.I.; Nackaerts, K.; Rodriguez Portal, J.A.; et al. Serum mesothelin for diagnosing malignant pleural mesothelioma: An individual patient data meta-analysis. J. Clin. Oncol. 2012, 30, 1541–1549. [Google Scholar] [CrossRef]
  61. Panou, V.; Vyberg, M.; Weinreich, U.M.; Meristoudis, C.; Falkmer, U.G.; Roe, O.D. The established and future biomarkers of malignant pleural mesothelioma. Cancer Treat. Rev. 2015, 41, 486–495. [Google Scholar] [CrossRef] [PubMed]
  62. Johnen, G.; Burek, K.; Raiko, I.; Wichert, K.; Pesch, B.; Weber, D.G.; Lehnert, M.; Casjens, S.; Hagemeyer, O.; Taeger, D.; et al. Prediagnostic detection of mesothelioma by circulating calretinin and mesothelin—A case-control comparison nested into a prospective cohort of asbestos-exposed workers. Sci. Rep. 2018, 8, 14321. [Google Scholar] [CrossRef] [PubMed]
  63. Hassan, R.; Kindler, H.L.; Jahan, T.; Bazhenova, L.; Reck, M.; Thomas, A.; Pastan, I.; Parno, J.; O’Shannessy, D.J.; Fatato, P.; 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, 5927–5936. [Google Scholar] [CrossRef] [PubMed]
  64. Kindler, H.L.; Novello, S.; Bearz, A.; Ceresoli, G.L.; Aerts, J.; Spicer, J.; Taylor, P.; Nackaerts, K.; Greystoke, A.; Jennens, R.; et al. Anetumab ravtansine versus vinorelbine in patients with relapsed, mesothelin-positive malignant pleural mesothelioma (ARCS-M): A randomised, open-label phase 2 trial. Lancet Oncol. 2022, 23, 540–552. [Google Scholar] [CrossRef]
  65. Hassan, R.; Sharon, E.; Thomas, A.; Zhang, J.; Ling, A.; Miettinen, M.; Kreitman, R.J.; Steinberg, S.M.; Hollevoet, K.; Pastan, I. 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, 3311–3319. [Google Scholar] [CrossRef]
  66. Baldo, P.; Cecco, S. Amatuximab and novel agents targeting mesothelin for solid tumors. Onco Targets Ther. 2017, 10, 5337–5353. [Google Scholar] [CrossRef]
  67. Golfier, S.; Kopitz, C.; Kahnert, A.; Heisler, I.; Schatz, C.A.; Stelte-Ludwig, B.; Mayer-Bartschmid, A.; Unterschemmann, K.; Bruder, S.; Linden, L.; 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, 1537–1548. [Google Scholar] [CrossRef] [PubMed]
  68. Hassan, R.; Thomas, A.; Alewine, C.; Le, D.T.; Jaffee, E.M.; Pastan, I. Mesothelin Immunotherapy for Cancer: Ready for Prime Time? J. Clin. Oncol. 2016, 34, 4171–4179. [Google Scholar] [CrossRef]
  69. Hassan, R.; Blumenschein, G.R., Jr.; Moore, K.N.; Santin, A.D.; Kindler, H.L.; Nemunaitis, J.J.; Seward, S.M.; Thomas, A.; Kim, S.K.; Rajagopalan, P.; et al. First-in-Human, Multicenter, Phase I Dose-Escalation and Expansion Study of Anti-Mesothelin Antibody-Drug Conjugate Anetumab Ravtansine in Advanced or Metastatic Solid Tumors. J. Clin. Oncol. 2020, 38, 1824–1835. [Google Scholar] [CrossRef]
  70. Mansfield, A.S.; Vivien Yin, J.; Bradbury, P.; Kwiatkowski, D.J.; Patel, S.; Bazhenova, L.A.; Forde, P.; Lou, Y.; Dizona, P.; Villaruz, L.C.; et al. Randomized trial of anetumab ravtansine and pembrolizumab compared to pembrolizumab for mesothelioma. Lung Cancer 2024, 195, 107928. [Google Scholar] [CrossRef]
  71. Kodama, Y.; Tanaka, I.; Sato, T.; Hori, K.; Gen, S.; Morise, M.; Matsubara, D.; Sato, M.; Sekido, Y.; Hashimoto, N. Oxytocin receptor is a promising therapeutic target of malignant mesothelioma. Cancer Sci. 2021, 112, 3520–3532. [Google Scholar] [CrossRef]
  72. Jurek, B.; Neumann, I.D. The Oxytocin Receptor: From Intracellular Signaling to Behavior. Physiol. Rev. 2018, 98, 1805–1908. [Google Scholar] [CrossRef]
  73. Horie, K.; Inoue, K.; Suzuki, S.; Adachi, S.; Yada, S.; Hirayama, T.; Hidema, S.; Young, L.J.; Nishimori, K. Oxytocin receptor knockout prairie voles generated by CRISPR/Cas9 editing show reduced preference for social novelty and exaggerated repetitive behaviors. Horm. Behav. 2019, 111, 60–69. [Google Scholar] [CrossRef] [PubMed]
  74. Schmid, B.; Wong, S.; Mitchell, B.F. Transcriptional regulation of oxytocin receptor by interleukin-1beta and interleukin-6. Endocrinology 2001, 142, 1380–1385. [Google Scholar] [CrossRef] [PubMed]
  75. Marjon, K.; Cameron, M.J.; Quang, P.; Clasquin, M.F.; Mandley, E.; Kunii, K.; McVay, M.; Choe, S.; Kernytsky, A.; Gross, S.; et al. MTAP Deletions in Cancer Create Vulnerability to Targeting of the MAT2A/PRMT5/RIOK1 Axis. Cell Rep. 2016, 15, 574–587. [Google Scholar] [CrossRef] [PubMed]
  76. Guccione, E.; Richard, S. The regulation, functions and clinical relevance of arginine methylation. Nat. Rev. Mol. Cell Biol. 2019, 20, 642–657. [Google Scholar] [CrossRef]
  77. Mavrakis, K.J.; McDonald, E.R., 3rd; Schlabach, M.R.; Billy, E.; Hoffman, G.R.; deWeck, A.; Ruddy, D.A.; Venkatesan, K.; Yu, J.; McAllister, G.; et al. Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5. Science 2016, 351, 1208–1213. [Google Scholar] [CrossRef]
  78. Hmeljak, J.; Sanchez-Vega, F.; Hoadley, K.A.; Shih, J.; Stewart, C.; Heiman, D.; Tarpey, P.; Danilova, L.; Drill, E.; Gibb, E.A.; et al. Integrative Molecular Characterization of Malignant Pleural Mesothelioma. Cancer Discov. 2018, 8, 1548–1565. [Google Scholar] [CrossRef]
  79. Kryukov, G.V.; Wilson, F.H.; Ruth, J.R.; Paulk, J.; Tsherniak, A.; Marlow, S.E.; Vazquez, F.; Weir, B.A.; Fitzgerald, M.E.; Tanaka, M.; et al. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science 2016, 351, 1214–1218. [Google Scholar] [CrossRef]
  80. Barbarino, M.; Cesari, D.; Bottaro, M.; Luzzi, L.; Namagerdi, A.; Bertolino, F.M.; Bellan, C.; Proietti, F.; Somma, P.; Micheli, M.; et al. PRMT5 silencing selectively affects MTAP-deleted mesothelioma: In vitro evidence of a novel promising approach. J. Cell Mol. Med. 2020, 24, 5565–5577. [Google Scholar] [CrossRef]
  81. Engstrom, L.D.; Aranda, R.; Waters, L.; Moya, K.; Bowcut, V.; Vegar, L.; Trinh, D.; Hebbert, A.; Smith, C.R.; Kulyk, S.; et al. MRTX1719 Is an MTA-Cooperative PRMT5 Inhibitor That Exhibits Synthetic Lethality in Preclinical Models and Patients with MTAP-Deleted Cancer. Cancer Discov. 2023, 13, 2412–2431. [Google Scholar] [CrossRef] [PubMed]
  82. Okado, S.; Kato, T.; Hanamatsu, Y.; Emoto, R.; Imamura, Y.; Watanabe, H.; Kawasumi, Y.; Kadomatsu, Y.; Ueno, H.; Nakamura, S.; et al. CHST4 Gene as a Potential Predictor of Clinical Outcome in Malignant Pleural Mesothelioma. Int. J. Mol. Sci. 2024, 25, 2270. [Google Scholar] [CrossRef]
  83. Bindea, G.; Mlecnik, B.; Tosolini, M.; Kirilovsky, A.; Waldner, M.; Obenauf, A.C.; Angell, H.; Fredriksen, T.; Lafontaine, L.; Berger, A.; et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 2013, 39, 782–795. [Google Scholar] [CrossRef] [PubMed]
  84. Veerman, K.; Tardiveau, C.; Martins, F.; Coudert, J.; Girard, J.P. Single-Cell Analysis Reveals Heterogeneity of High Endothelial Venules and Different Regulation of Genes Controlling Lymphocyte Entry to Lymph Nodes. Cell Rep. 2019, 26, 3116–3131. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, F.; Zhang, H.; Qiu, G.; Li, Z.; Wang, Y. The LINC00452/miR-204/CHST4 Axis Regulating Thymic Tregs Might Be Involved in the Progression of Thymoma-Associated Myasthenia Gravis. Front. Neurol. 2022, 13, 828970. [Google Scholar] [CrossRef]
  86. Nowak, A.K.; Giroux, D.J.; Eisele, M.; Rosenthal, A.; Bille, A.; Gill, R.R.; Kindler, H.L.; Pass, H.I.; Rice, D.; Ripley, R.T.; et al. The International Association for the Study of Lung Cancer Pleural Mesothelioma Staging Project: Proposal for Revision of the TNM Stage Groupings in the Forthcoming (Ninth) Edition of the TNM Classification for Pleural Mesothelioma. J. Thorac. Oncol. 2024, 19, 1339–1351. [Google Scholar] [CrossRef]
  87. Ito, T.; Nakamura, S.; Kadomatsu, Y.; Ueno, H.; Kato, T.; Ozeki, N.; Fukumoto, K.; Chen-Yoshikawa, T.F. Impact of Pleural Thickness on Occurrence of Postoperative Complications in Patients with Malignant Pleural Mesothelioma. Ann. Surg. Oncol. 2023, 30, 1574–1583. [Google Scholar] [CrossRef]
  88. Cersosimo, F.; Barbarino, M.; Lonardi, S.; Vermi, W.; Giordano, A.; Bellan, C.; Giurisato, E. Mesothelioma Malignancy and the Microenvironment: Molecular Mechanisms. Cancers 2021, 13, 5664. [Google Scholar] [CrossRef]
  89. Izzi, V.; Chiurchiu, V.; D’Aquilio, F.; Palumbo, C.; Tresoldi, I.; Modesti, A.; Baldini, P.M. Differential effects of malignant mesothelioma cells on THP-1 monocytes and macrophages. Int. J. Oncol. 2009, 34, 543–550. [Google Scholar] [CrossRef]
  90. Chu, G.J.; van Zandwijk, N.; Rasko, J.E.J. The Immune Microenvironment in Mesothelioma: Mechanisms of Resistance to Immunotherapy. Front. Oncol. 2019, 9, 1366. [Google Scholar] [CrossRef]
  91. Hasko, G.; Pacher, P. Regulation of macrophage function by adenosine. Arter. Thromb. Vasc. Biol. 2012, 32, 865–869. [Google Scholar] [CrossRef] [PubMed]
  92. Blum, Y.; Meiller, C.; Quetel, L.; Elarouci, N.; Ayadi, M.; Tashtanbaeva, D.; Armenoult, L.; Montagne, F.; Tranchant, R.; Renier, A.; et al. Dissecting heterogeneity in malignant pleural mesothelioma through histo-molecular gradients for clinical applications. Nat. Commun. 2019, 10, 1333. [Google Scholar] [CrossRef]
  93. Ujiie, H.; Kadota, K.; Nitadori, J.I.; Aerts, J.G.; Woo, K.M.; Sima, C.S.; Travis, W.D.; Jones, D.R.; Krug, L.M.; Adusumilli, P.S. The tumoral and stromal immune microenvironment in malignant pleural mesothelioma: A comprehensive analysis reveals prognostic immune markers. Oncoimmunology 2015, 4, e1009285. [Google Scholar] [CrossRef] [PubMed]
  94. Lievense, L.A.; Sterman, D.H.; Cornelissen, R.; Aerts, J.G. Checkpoint Blockade in Lung Cancer and Mesothelioma. Am. J. Respir. Crit. Care Med. 2017, 196, 274–282. [Google Scholar] [CrossRef]
  95. Maio, M.; Scherpereel, A.; Calabro, L.; Aerts, J.; Perez, S.C.; Bearz, A.; Nackaerts, K.; Fennell, D.A.; Kowalski, D.; Tsao, A.S.; et al. Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): A multicentre, international, randomised, double-blind, placebo-controlled phase 2b trial. Lancet Oncol. 2017, 18, 1261–1273. [Google Scholar] [CrossRef]
  96. Popat, S.; Curioni-Fontecedro, A.; Dafni, U.; Shah, R.; O’Brien, M.; Pope, A.; Fisher, P.; Spicer, J.; Roy, A.; Gilligan, D.; et al. A multicentre randomised phase III trial comparing pembrolizumab versus single-agent chemotherapy for advanced pre-treated malignant pleural mesothelioma: The European Thoracic Oncology Platform (ETOP 9-15) PROMISE-meso trial. Ann. Oncol. 2020, 31, 1734–1745. [Google Scholar] [CrossRef]
  97. Ceresoli, G.L.; Zucali, P.A.; De Vincenzo, F.; Gianoncelli, L.; Simonelli, M.; Lorenzi, E.; Ripa, C.; Giordano, L.; Santoro, A. Retreatment with pemetrexed-based chemotherapy in patients with malignant pleural mesothelioma. Lung Cancer 2011, 72, 73–77. [Google Scholar] [CrossRef] [PubMed]
  98. Gandhi, L.; Rodriguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef]
  99. Alley, E.W.; Lopez, J.; Santoro, A.; Morosky, A.; Saraf, S.; Piperdi, B.; van Brummelen, E. Clinical safety and activity of pembrolizumab in patients with malignant pleural mesothelioma (KEYNOTE-028): Preliminary results from a non-randomised, open-label, phase 1b trial. Lancet Oncol. 2017, 18, 623–630. [Google Scholar] [CrossRef]
  100. Chu, Q.; Perrone, F.; Greillier, L.; Tu, W.; Piccirillo, M.C.; Grosso, F.; Lo Russo, G.; Florescu, M.; Mencoboni, M.; Morabito, A.; et al. Pembrolizumab plus chemotherapy versus chemotherapy in untreated advanced pleural mesothelioma in Canada, Italy, and France: A phase 3, open-label, randomised controlled trial. Lancet 2023, 402, 2295–2306. [Google Scholar] [CrossRef]
  101. Forde, P.M.; Anagnostou, V.; Sun, Z.; Dahlberg, S.E.; Kindler, H.L.; Niknafs, N.; Purcell, T.; Santana-Davila, R.; Dudek, A.Z.; Borghaei, H.; et al. Durvalumab with platinum-pemetrexed for unresectable pleural mesothelioma: Survival, genomic and immunologic analyses from the phase 2 PrE0505 trial. Nat. Med. 2021, 27, 1910–1920. [Google Scholar] [CrossRef]
  102. Nowak, A.K.; Lesterhuis, W.J.; Kok, P.S.; Brown, C.; Hughes, B.G.; Karikios, D.J.; John, T.; Kao, S.C.; Leslie, C.; Cook, A.M.; et al. Durvalumab with first-line chemotherapy in previously untreated malignant pleural mesothelioma (DREAM): A multicentre, single-arm, phase 2 trial with a safety run-in. Lancet Oncol. 2020, 21, 1213–1223. [Google Scholar] [CrossRef] [PubMed]
  103. Kok, P.S.; Forde, P.M.; Hughes, B.; Sun, Z.; Brown, C.; Ramalingam, S.; Cook, A.; Lesterhuis, W.J.; Yip, S.; O’Byrne, K.; et al. Protocol of DREAM3R: DuRvalumab with chEmotherapy as first-line treAtment in advanced pleural Mesothelioma-a phase 3 randomised trial. BMJ Open 2022, 12, e057663. [Google Scholar] [CrossRef] [PubMed]
  104. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef]
  105. Zalcman, G.; Mazieres, J.; Margery, J.; Greillier, L.; Audigier-Valette, C.; Moro-Sibilot, D.; Molinier, O.; Corre, R.; Monnet, I.; Gounant, V.; et al. Bevacizumab for newly diagnosed pleural mesothelioma in the Mesothelioma Avastin Cisplatin Pemetrexed Study (MAPS): A randomised, controlled, open-label, phase 3 trial. Lancet 2016, 387, 1405–1414. [Google Scholar] [CrossRef] [PubMed]
  106. Felip, E.; Popat, S.; Dafni, U.; Ribi, K.; Pope, A.; Cedres, S.; Shah, R.; de Marinis, F.; Cove Smith, L.; Bernabe, R.; et al. A randomised phase III study of bevacizumab and carboplatin-pemetrexed chemotherapy with or without atezolizumab as first-line treatment for advanced pleural mesothelioma: Results of the ETOP 13-18 BEAT-meso trial. Ann. Oncol. 2025, 36, 548–560. [Google Scholar] [CrossRef]
  107. Okada, M.; Kijima, T.; Aoe, K.; Kato, T.; Fujimoto, N.; Nakagawa, K.; Takeda, Y.; Hida, T.; Kanai, K.; Imamura, F.; et al. Clinical Efficacy and Safety of Nivolumab: Results of a Multicenter, Open-label, Single-arm, Japanese Phase II study in Malignant Pleural Mesothelioma (MERIT). Clin. Cancer Res. 2019, 25, 5485–5492. [Google Scholar] [CrossRef]
  108. Fennell, D.A.; Ewings, S.; Ottensmeier, C.; Califano, R.; Hanna, G.G.; Hill, K.; Danson, S.; Steele, N.; Nye, M.; Johnson, L.; et al. Nivolumab versus placebo in patients with relapsed malignant mesothelioma (CONFIRM): A multicentre, double-blind, randomised, phase 3 trial. Lancet Oncol. 2021, 22, 1530–1540. [Google Scholar] [CrossRef]
  109. Sterman, D.H.; Alley, E.; Stevenson, J.P.; Friedberg, J.; Metzger, S.; Recio, A.; Moon, E.K.; Haas, A.R.; Vachani, A.; Katz, S.I.; et al. Pilot and Feasibility Trial Evaluating Immuno-Gene Therapy of Malignant Mesothelioma Using Intrapleural Delivery of Adenovirus-IFNalpha Combined with Chemotherapy. Clin. Cancer Res. 2016, 22, 3791–3800. [Google Scholar] [CrossRef]
  110. Fesnak, A.D.; June, C.H.; Levine, B.L. Engineered T cells: The promise and challenges of cancer immunotherapy. Nat. Rev. Cancer 2016, 16, 566–581. [Google Scholar] [CrossRef]
  111. Maus, M.V.; Haas, A.R.; Beatty, G.L.; Albelda, S.M.; Levine, B.L.; Liu, X.; Zhao, Y.; Kalos, M.; June, C.H. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol. Res. 2013, 1, 26–31. [Google Scholar] [CrossRef] [PubMed]
  112. Beatty, G.L.; Haas, A.R.; Maus, M.V.; Torigian, D.A.; Soulen, M.C.; Plesa, G.; Chew, A.; Zhao, Y.; Levine, B.L.; Albelda, S.M.; et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol. Res. 2014, 2, 112–120. [Google Scholar] [CrossRef] [PubMed]
  113. Adusumilli, P.S.; Zauderer, M.G.; Riviere, I.; Solomon, S.B.; Rusch, V.W.; O’Cearbhaill, R.E.; Zhu, A.; Cheema, W.; Chintala, N.K.; Halton, E.; et al. A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti-PD-1 Agent Pembrolizumab. Cancer Discov. 2021, 11, 2748–2763. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A schematic illustration depicting the dysregulation of the NF2/Merlin-Hippo signaling cascade in mesothelioma cells. Extracellular signals are transmitted through cell–cell interactions (via cadherins), cell–matrix interactions (via integrins), and growth factor receptors, such as receptor tyrosine kinases, influencing the tumor-suppressive function of Merlin. When Merlin is in an active, underphosphorylated state, it modulates the Hippo signaling cascade and inhibits the activity of YAP1/TAZ transcriptional coactivators. However, in mesothelioma cells, frequent alterations in Merlin (the NF2 gene product) and key Hippo signaling pathway components, including LATS1/2, lead to YAP1/TAZ activation (underphosphorylation). This, in turn, promotes the expression of multiple pro-oncogenic genes.
Figure 1. A schematic illustration depicting the dysregulation of the NF2/Merlin-Hippo signaling cascade in mesothelioma cells. Extracellular signals are transmitted through cell–cell interactions (via cadherins), cell–matrix interactions (via integrins), and growth factor receptors, such as receptor tyrosine kinases, influencing the tumor-suppressive function of Merlin. When Merlin is in an active, underphosphorylated state, it modulates the Hippo signaling cascade and inhibits the activity of YAP1/TAZ transcriptional coactivators. However, in mesothelioma cells, frequent alterations in Merlin (the NF2 gene product) and key Hippo signaling pathway components, including LATS1/2, lead to YAP1/TAZ activation (underphosphorylation). This, in turn, promotes the expression of multiple pro-oncogenic genes.
Ijms 26 04299 g001
Figure 2. Transcriptional regulation machinery comprising BAP1, PRC1, and PRC2. BAP1 is a major deubiquitinase that removes H2AK119Ub, a histone marker deposited by the PRC1 complex. BAP1 loss leads to H2AK119Ub accumulation, which in turn promotes recruitment of the PRC2 complex including EZH2, resulting in elevated H3K27me3 levels and transcriptional repression.
Figure 2. Transcriptional regulation machinery comprising BAP1, PRC1, and PRC2. BAP1 is a major deubiquitinase that removes H2AK119Ub, a histone marker deposited by the PRC1 complex. BAP1 loss leads to H2AK119Ub accumulation, which in turn promotes recruitment of the PRC2 complex including EZH2, resulting in elevated H3K27me3 levels and transcriptional repression.
Ijms 26 04299 g002
Figure 3. The CDKN2A gene produces two distinct tumor suppressor proteins through alternative reading frames, namely, p16INK4A (P16), derived from the alpha transcript, and p14ARF (P14), derived from the beta transcript. These proteins play pivotal roles in halting cell cycle progression, particularly during the G1 to S phase transition.
Figure 3. The CDKN2A gene produces two distinct tumor suppressor proteins through alternative reading frames, namely, p16INK4A (P16), derived from the alpha transcript, and p14ARF (P14), derived from the beta transcript. These proteins play pivotal roles in halting cell cycle progression, particularly during the G1 to S phase transition.
Ijms 26 04299 g003
Figure 4. Role of CHST4 in lymphocyte homing and mesothelioma. Lymphocytes begin their interaction with high endothelial venules by binding L-selectin to its ligand, leading to integrin-mediated adhesion and transendothelial migration. Sulfation by CHST4 plays a pivotal role in the enhancement of lymphocyte homing and the chemokine-induced activation step of this sequential adhesion process. In mesothelioma, CHST4 mRNA expression exhibited a substantial correlation with immune–gene signatures representing tumor-infiltrating lymphocytes. CHST4 is hypothesized to contribute to tumor immunity against mesothelioma via lymphocyte infiltration.
Figure 4. Role of CHST4 in lymphocyte homing and mesothelioma. Lymphocytes begin their interaction with high endothelial venules by binding L-selectin to its ligand, leading to integrin-mediated adhesion and transendothelial migration. Sulfation by CHST4 plays a pivotal role in the enhancement of lymphocyte homing and the chemokine-induced activation step of this sequential adhesion process. In mesothelioma, CHST4 mRNA expression exhibited a substantial correlation with immune–gene signatures representing tumor-infiltrating lymphocytes. CHST4 is hypothesized to contribute to tumor immunity against mesothelioma via lymphocyte infiltration.
Ijms 26 04299 g004
Table 1. Recent clinical trials of first-line and salvage immunotherapy conducted in mesothelioma.
Table 1. Recent clinical trials of first-line and salvage immunotherapy conducted in mesothelioma.
TrialIDTreatmentStatus/Result
DREAM3RNCT04334759Randomized phase III trial of durvalumab with chemotherapy as first-line treatmentActive but not recruiting
MAPSNCT00651456Randomized phase III trial of bevacizumab plus standard chemotherapy as first-line treatmentPositive
BEAT-meso trialNCT03762018Randomized phase III trial comparing atezolizumab plus bevacizumab and standard chemotherapy with bevacizumab and standard chemotherapy as first-line treatmentNegative
PROMISE-MesoNCT02991482Randomized phase III trial comparing pembrolizumab with standard chemotherapy for pretreated mesotheliomaNegative
CONFIRMNCT03063450Placebo controlled phase III trial to evaluate the efficacy of nivolumab in relapsed mesotheliomaPositive
INFINITE NCT03710876Randomized phase III trial of intrapleural administration of adenovirus-delivered interferon alfa-2b in combination with celecoxib and gemcitabine for pretreated mesotheliomaActive but
not recruiting
CheckMate 743NCT0289929Randomized phase III trial of nivolumab plus ipilimumab vs. pemetrexed and platinum as first-line treatmentPositive
KEYNOTE-028NCT02054806Single-arm phase Ib trial of pembrolizumab for previous treated mesotheliomaOS 18 months
DREAMACTRN
12616001170415
Single-arm phase II trial of durvalumab in combination with cisplatin and pemetrexed as first-line treatmentOS 18.4 months
PrECOG0505NCT02899195 Sigle-arm phase II trial of durvalumab in combination with cisplatin and pemetrexed as the first-line treatmentOS 20.5 months
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kato, T.; Tanaka, I.; Huang, H.; Okado, S.; Imamura, Y.; Nomata, Y.; Takenaka, H.; Watanabe, H.; Kawasumi, Y.; Nakanishi, K.; et al. Molecular Mechanisms of Tumor Progression and Novel Therapeutic and Diagnostic Strategies in Mesothelioma. Int. J. Mol. Sci. 2025, 26, 4299. https://doi.org/10.3390/ijms26094299

AMA Style

Kato T, Tanaka I, Huang H, Okado S, Imamura Y, Nomata Y, Takenaka H, Watanabe H, Kawasumi Y, Nakanishi K, et al. Molecular Mechanisms of Tumor Progression and Novel Therapeutic and Diagnostic Strategies in Mesothelioma. International Journal of Molecular Sciences. 2025; 26(9):4299. https://doi.org/10.3390/ijms26094299

Chicago/Turabian Style

Kato, Taketo, Ichidai Tanaka, Heng Huang, Shoji Okado, Yoshito Imamura, Yuji Nomata, Hirofumi Takenaka, Hiroki Watanabe, Yuta Kawasumi, Keita Nakanishi, and et al. 2025. "Molecular Mechanisms of Tumor Progression and Novel Therapeutic and Diagnostic Strategies in Mesothelioma" International Journal of Molecular Sciences 26, no. 9: 4299. https://doi.org/10.3390/ijms26094299

APA Style

Kato, T., Tanaka, I., Huang, H., Okado, S., Imamura, Y., Nomata, Y., Takenaka, H., Watanabe, H., Kawasumi, Y., Nakanishi, K., Kadomatsu, Y., Ueno, H., Nakamura, S., Mizuno, T., & Chen-Yoshikawa, T. F. (2025). Molecular Mechanisms of Tumor Progression and Novel Therapeutic and Diagnostic Strategies in Mesothelioma. International Journal of Molecular Sciences, 26(9), 4299. https://doi.org/10.3390/ijms26094299

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop