Synopsis
Malignant pleural mesothelioma (MPM) is a rare, aggressive malignancy of the pleural lining associated with asbestos exposure in >80% of cases. It is characterized by molecular heterogeneity both between patients and within individual tumors. Next-generation sequencing technology and novel computational techniques have resulted in a greater understanding of the epigenetic, genetic, and transcriptomic hallmarks of MPM. This chapter will review these features and discuss the implications of advances in MPM molecular biology in clinical practice.
Keywords: mesothelioma, molecular, genetic, gene expression, epigenetic
6.1. Introduction
Malignant pleural mesothelioma (MPM) is a rare but aggressive cancer associated with asbestos exposure in >80% of cases1. It is almost uniformly lethal, and while decreasing use of asbestos has led to a plateau of incidence in Western countries, a long latency period after exposure combined with continued global asbestos use make MPM an ongoing area of concern2. MPM is classified into three histologic subtypes: epithelioid, sarcomatoid, and biphasic. Epithelioid histology confers the most favorable prognosis and sarcomatoid the least, and a greater proportion of epithelial differentiation in biphasic tumors correlates with longer survival3,4.
At the molecular level, MPM is a highly heterogeneous disease both between patients and within individual tumors5–8. Intratumor heterogeneity can be further conceptualized as a combination of longitudinal (change over time) and spatial (among samples of the same tumor) heterogeneity7. The broad molecular variation seen in MPM and its microenvironment poses a significant challenge in diagnosis, prognostication, and treatment of this devastating disease. While advances in molecular oncology have led to effective novel therapeutics for several solid organ cancers, first line medical therapy for MPM in the form of cytotoxic combination cisplatin/pemetrexed-based chemotherapy has remained unchanged for decades9,10. The application of surgery and hemithoracic radiation in multimodal approaches prolongs survival only in a subset of patients, and the ability to accurately predict patient response to any form of treatment is limited9,11.
Next-generation sequencing (NGS) technology and novel computational techniques combined with international collaborative efforts have resulted in greater understanding the molecular basis of MPM. This chapter will describe current molecular mechanisms behind MPM tumorigenesis, review the epigenetic, genetic, and transcriptomic hallmarks of MPM, and discuss the implications of advances in MPM molecular biology in clinical practice.
6.2. Current Evidence & Research
6.2.1. Tumorigenesis
MPM arises from malignant transformation of the mesothelial cell monolayer on the surface of the parietal pleura. Approximately 70–90% of cases are associated with exposure to asbestos fibers12,13. The majority of research into asbestos as a cause of MPM relies on self-report of exposure, and quantitative data on the relation of asbestos exposure to mesothelioma risk are rare14.
Asbestos, unlike chemical carcinogens, exerts its effects over a long period which is consistent with the 10–40-year latency period between estimated exposure and MPM diagnosis15. There is debate over the specific mechanisms through which asbestos causes mesothelioma6. Implicated pathways include generation of reactive oxygen species (ROS), direct cytotoxicity, kinase-mediated signaling, chronic inflammation, and cytokine and growth factor dysregulation6,13,16. A small subset of MPM tumors exhibit widespread loss of heterozygosity which may be consistent with spindle damage induced directly by asbestos fibers as well2. It is also likely that these mechanisms overlap and that no single pathway can be identified as a sole sufficient cause of malignant transformation.
Reactive Oxygen Species
Asbestos fibers generate ROS both directly and indirectly (e.g. through immune-mediated inflammation). These in turn lead to epigenetic and somatic genetic changes in mesothelial cells17. At the epigenetic level, increased ROS cause altered DNA methylation17. In vitro treatment of Met5A cells with asbestos, for example, resulted in significant methylation changes in CpG islands located in the promoter regions of genes involved in migration/cell adhesion. However, no correlation between changes in methylation and expression of these genes was observed except for a significant inverse correlation with DKK1, whose protein is an antagonist of the Wnt/β-catenin signaling pathway18. Interestingly, in the absence of asbestos, in vitro treatment of MPM cell lines with exemestane, an aromatase inhibitor which generates ROS, had an antiproliferative effect19.
Downstream Effects of Environmental Stress: Mutagenesis and Failure of DNA Repair
MPM has a relatively low rate of somatic mutation compared to other solid cancers5. However, whether through ROS or other molecular mechanisms, asbestos fibers are clearly mutagenic at the chromosomal and gene levels in both in vitro and in vivo models, leading to tumorigenesis16,20. These damage patterns are consistent with known frequent alterations in DNA repair genes5. In contrast, asbestos-induced pleural thickening and plaques is produced by changes in gene regulation secondary to inflammation and ROS without mutagenesis16,21,22. This suggests that mutagenesis is a key step in development of asbestos-induced MPM. However, whether there is a threshold of asbestos exposure below which cancer does not develop remains controversial and likely depends on patient-specific genetic factors16,23.
Although chronic inflammation is implicated in MPM development, the role of inflammation-related genes in development of MPM following asbestos exposure is still controversial. Crovella and colleagues23 investigated the role of 93 genetic variants in 12 genes encoding inflammasome and iron metabolism proteins in relation to the number of asbestos bodies (ABs), considered a hallmark of asbestos exposure, in 81 patients who died of MPM. Although there was no association between the number of ABs and most of the selected genes, the frequency of the single nucleotide polymorphism (SNP) rs12150220 A/T (17p13.2) in the NLRP1 gene correlated with a significantly lower number of ABs, suggesting that NLRP1 inflammasome may contribute in the development of lung ABs. A subsequent analysis by the same group found no association between polymorphisms in NLRP1 or NLRP3 and susceptibility to MPM in asbestos-exposed individuals24.
Non-Asbestos Causes of MPM
Despite the myriad of pathways through which asbestos can cause MPM, the risk of developing MPM among high-risk individuals with industrial asbestos exposure is only ~5%25. Other factors have been associated with mesothelioma: non-asbestos mineral fibers (e.g. erionite, fluoro-edenite, carbon nanotubes), therapeutic radiation, chronic pleural inflammation, and (in rare cases) germline genetic mutations12. Apart from germline mutations, a definitive molecular signature to differentiate these etiologies has not yet been developed.
6.2.2. Epigenetics
Chromosomal Losses
Aneuploidy, particularly chromosomal loss, is an epigenetic hallmark of MPM7,26–28. However, the copy number alteration (CNA) profiles of individual tumors are complex29. In one analysis of CNAs in 53 primary MPM tumor samples, 77% demonstrated a predominance of losses and 23% a predominance of gains30. The most common losses are in 1p, 3p14-p21, whole chromosome 4, 6q, 9p, and 22q7,26,27. None of these losses individually predominate in MPM. The frequencies of common losses include 9p21 (34%), 22q (32%), 4q31–32 (29%), 4p12–13 (25%), and 3p21 (16%)28. These regions contain some of the most commonly mutated genes in MPM, including BAP1 (3p), CDKN2A (9p), and NF2 (22q)7.
In addition to large chromosomal losses, focal losses have been described. For example, in the Guo et al series27 deletion of 9p21 containing CDKN2A/2B was identified. In The Cancer Genome Atlas (TCGA) analysis2, focal copy number deletions were found to affect canonical MPM tumor suppressor genes including CDKN2A (>50% of samples) and NF2 (>70%). Deletions of CDKN2A often involve the adjacent gene MTAP, which has been linked to increased sensitivity to pharmacologic inhibition2. Loss of CDKN2A was also associated with shorter overall survival2.
TCGA analysis also identified a rare MPM subtype in a small number of tumors exhibiting genomic near-haploidization, absent alteration in BAP1, PBRM1, or SETD2, and universal inactivation of SETDB1. Females were over-represented in this subtype (4F:1M), while histologic subtype showed no difference from the MPM cohort at large2.
Chromosomal Gains
Although less common than losses, gains in some MPM chromosomal regions have been described. In one comparative genomic hybridization (CGH) analysis of 26 MPM tumors, 7 (27%) were found to have recurrent gains in 17q, involving known cancer-related genes such as MAP3K3, SMARCD2, ERN1, and PRKCA31. Krismann and colleagues28 examined 90 MPM cases using CGH and DNA cytometry, finding common gains in 8q22–23 (18%), 1q23/1q32 (16%), 7p14–15 (14%), and 15q22–25 (14%). An analysis of 41 epithelioid MPM revealed relative gains in the regions encompassing KDM5A (12p13), DVL1 (1p36), and MYC (8q24) compared with peritoneal mesothelioma samples32.
Chromosomal Alterations by Histologic Subtype
In the Krismann cohort28, aneuploidy was significantly less frequent in sarcomatoid samples (75%) and significantly more frequent in epithelioid samples (88%), although absolute differences were small. Imbalances were detected by CGH in 84% of all samples with an average of 6.2 defects per sample. Losses of chromosomal regions were twice as frequent as gains consistent with observations in other studies30. Epithelioid MPM had distinct recurrent losses at several locations including 3p21 (33% vs. 16% in the whole cohort) and 17p12–pter (26%); sarcomatoid MPM had distinct recurrent losses at 7q31–qter (21%) and 15q (18%). Biphasic tumors demonstrated a CGH pattern consistent with a combination of the other two subtypes28.
Changes in Gene Regulation
Dysregulation of epigenetic control of tumor suppressor genes is also present in MPM, particularly hypermethylation33. In one high-throughput global screening analysis for aberrant DNA hypermethylation in 50 MPM specimens, an average of 6.3% of genes were found to be hypermethylated in MPM compared with 8.8% in lung adenocarcinoma34. Methylation patterns were distinct between the two tumors based on hierarchical cluster analysis, and three of the hypermethylated genes (TMEM30B, KAZALD1, and MAPK13) were unique to MPM suggesting a potential role for these genes as diagnostic markers34. Interestingly, four patients included in this study showed low levels of gene methylation and longer survival, suggesting that methylation may affect the progression of this disease. In addition, the number of methylated genes increased significantly in stages III and IV disease compared with stages I and II34. An analysis published in the same year by Christensen et al35 identified distinct epigenetic profiles between normal pleura and MPM. These data altogether suggest a unique epigenetic landscape in MPM compared with other forms of thoracic disease33–35.
Several key genes mutated in MPM are involved in epigenetic regulation. For example, in the Bueno cohort, 8% of tumors exhibited mutations in SETD2 which encodes a histone methyltransferase, often leading to loss of function. Mutations in the SETDB1 and SETD5 histone methyltransferase genes were also identified5. The downstream effects of these and other mutations in genes involved in epigenetic programming have not yet been fully elucidated. However, for example, ITGA7 is a known tumor suppressor gene that may be epigenetically regulated, and decreased expression of ITGA7 has been associated with decreased overall survival in MPM36. Tsou et al33 evaluated 52 MPM samples using the MethylLight technique for 28 methylation markers and found significant changes in methylation in the ESR1 (increased) and APC (decreased) loci, which are known to be involved in tumorigenesis. Similarly, tumors without DNA losses affecting DNMT1, a methyltransferase, exhibited higher average methylation indicating a significant change in the epigenetic landscape37.
Small non-coding RNAs (miRNAs) also participate in post-transcriptional regulation of gene expression; irreversible alterations in miRNA expression are associated with cancer development17. miR-126 in particular is known to play a crucial role in MPM pathogenesis, where it fails to act as an oncosuppressor by inhibition of the PI3K/AKT pathway. Treatment with exogenous miR-126 under these circumstances results in tumor suppression in vitro17. Over the last decade, the biological activity of many other miRNAs has been associated with MPM, including in the roles of tumor suppressor (miR-16–5p and miR-193a-3p) and cellular function (miR-182–5p, miR-183–5p, miR-24–3p) (reviewed in Reid et al, 2020)38.
6.2.3. DNA Mutation Signatures
Somatic Mutations
As previously described, MPM has a relatively low rate of protein-altering somatic point mutations compared to other solid cancers5. In a cohort of 74 MPM tumors, whole exome sequencing confirmed an overall rate of <2 nonsynonymous mutations per megabase in all but one sample2 and demonstrated that MPM had lower rate of protein-altering mutations than many other cancers except thyroid carcinoma and acute myeloid leukemia5.
In the Bueno series, targeted (n=103) and whole exome (n=99) sequencing of paired MPM tumors revealed an average of 24 +/− 11 protein-coding alterations per sample with no significant differences between molecular subtypes5. Quetel and colleagues demonstrated in 49 MPM primary cultures and in 35 frozen tumor specimens that mutations in MPM exhibit an enrichment in C>T transitions39. A recent review summarizing massively parallel sequencing studies has observed that genetic variations tend to cluster in the TP53/DNA repair pathway and the PI3K/AKT pathway29. Recent high-throughput analyses have identified recurrent mutations in several genes which underlie key features of MPM molecular biology.
Somatic Mutations – BAP1 and other Tumor Suppressors
The main recurrent genetic alterations in MPM have been identified in tumor suppressor genes. The most frequently mutated gene in most series is BRCA1-Assocated Protein 1 (BAP1), which is located in 3p21 (a region frequently lost in MPM) and altered in up to 60% of tumors2,5,26,30,40,41. BAP1 encodes a deubiquitinating enzyme involved in DNA repair, cell cycle, cellular differentiation, and DNA damage response42–44. BAP1 also promotes apoptosis in wild-type cells through deubiquitylation and stabilization of the IP3R3 channel45. Loss of nuclear BAP1 expression by immunohistochemistry (IHC) is currently used as a diagnostic marker in MPM. However, while loss of nuclear BAP1 staining can sometimes distinguish reactive versus neoplastic stroma particularly in biphasic tumors, BAP1 staining even within MPM is known to be heterogeneous46,47. There is also evidence that BAP1-mutant malignancies may be sensitive to epigenetically based therapies48. However, patient survival does not correlate with presence of BAP1 mutation itself2. In addition, MPM patients with germline BAP1 mutations have fewer chromosomal alterations than others2,49.
Beyond BAP1, frequently mutated tumor suppressor genes in MPM include CDKN2A, CDKN2B, NF2, and TP532,5,26,50. Seven additional significantly mutated genes, SETD2, ULK2, CFAP45, SETDB1, RYR2, DDX51, and DDX3X were identified in the Bueno cohort5. Mutations in TP53 were absent in epithelioid tumors. In this cohort, patients carrying TP53 mutation showed lower overall survival compared to patients with wild-type TP53 (P=0.0167). Another analysis of 49 MPM primary cell lines and 35 frozen tumors for 22 genes confirmed the high frequency of BAP1, NF2, CDKN2A/B, TP53, and SETD2 mutations in MPM39.
Another gene frequently mutated in MPM is LATS2, a member of the Hippo signaling pathway39. An analysis found alteration in LATS2 in 11% of 61 MPM primary cell lines51. Mutations in NF2 gene, another member of the Hippo pathway, were found to co-occur with LATS2 mutations in 8% of the cases. While other studies did not report a high rate of LATS2 mutation, large deletions of chromosome 13 where LATS2 resides may indicate potential loss of this gene and possible underestimation of the prevalence of LATS alterations51.
Germline Mutations
Germline mutations have been identified in up to 7–12% of patients with MPM52–54. Pleural site in general is less frequently associated with germline mutations than other primary mesothelioma sites52. Few studies have shown that germline mutation frequency increases with decreasing age at diagnosis52,54. In addition, patients with germline mutations are less likely to report asbestos exposure, more likely to report a second cancer diagnosis, and more likely to have epithelioid histology52,54.
Pathogenic germline variants in MPM are often involved in DNA damage repair and chromatin remodeling pathways, and BAP1 is the most frequently identified germline mutation52,54,55. Germline mutations in BAP1 are known to predispose families to mesothelioma56. BAP1 is also known to be frequently inactivated in cancers such as uveal melanoma, clear cell renal cancer, and cholangiocarcinoma57. Taken together, loss-of-function germline mutations in BAP1 constitute what is termed the familial BAP1 syndrome: MPM, uveal melanoma, cutaneous melanoma, and other dermatologic tumors, as well as renal cell carcinoma and meningioma58,59. MPM patients with germline BAP1 mutations almost always exhibit a second somatic BAP1 mutation leading to likely complete loss of function53. Germline BAP1 mutation is associated with less aggressive disease than sporadic MPM60.
Hassan and colleagues53 investigated the impact of inherited loss-of-function mutations on survival in mesothelioma following platinum-based chemotherapy. In a cohort of 385 MPM patients, they found significantly longer overall survival following platinum-based chemotherapy in patients with any germline mutation including BAP1 compared to patients without germline mutations (7.9y vs 2.4y, P=0.0012). The benefit was comparable across all the genes under investigation. Interestingly, the effect of genotype was significant for pleural, but not peritoneal mesothelioma. In addition, there was no difference in tumor histology or reported asbestos exposure between the germline mutant and control patients. Overall these results suggest that MPM patients with germline mutations in DNA repair and other tumor suppressor genes may benefit from platinum chemotherapy53. There is also evidence that the presence of germline mutations may predict sensitivity to PARP inhibition53,61.
Application of Gene Mutations to Diagnosis
MPM subtypes may be difficult to distinguish from benign pleural proliferation and from other tumors such as adenocarcinoma (for epithelioid MPM) and sarcoma (for sarcomatoid MPM)62. No single IHC stain is diagnostic, and agreement among expert pathologists classifying histologically biphasic MPM is moderate at best63. Homozygous deletion of CDKN2A by fluorescent in situ hybridization (FISH) can be useful in distinguishing benign florid stromal reaction from sarcomatoid components of biphasic MPM tumors64. Since chromosomal losses of CDKN2A often involve the adjacent gene MTAP, and IHC for MTAP correlates well with CDKN2A FISH, it has been suggested that IHC for MTAP may be clinically useful in diagnosis of MPM2,63.
6.2.4. Characterization of Gene Expression
Recent advances in gene expression profiling have allowed for the simultaneous analysis of thousands of genes. Gene expression data has been applied across major cancer types to identify novel subtypes, predict outcomes, and define heterogeneity and the need for personalized treatments65.
Some of the first molecular MPM classifications were generated in the early 2000s primarily using microarrays66–69. Microarray data has also been used to identify candidate tumor-associated genes. For example, an analysis of miRNA dysregulation implicated CDKN2A, NF2, JUN, HGF, and PDGF2A as frequently affected in mesothelioma70. A subsequent meta-analysis of several sets of microarray data defined a list of potential novel biomarkers for MPM including PTGS2, BIRC5, ASS1, JUNB, MCM2, AURKA, FGF2, MKI67, CAV1, SFRP1, CCNB1, CDK4, and MSLN71.
Several efforts have been made over the years to classify MPM tumors according to molecular characteristics. Gordon and colleagues used expression arrays to analyze 40 MPM tumors as well as normal pleura, normal lung, and MPM cell lines72. Unsupervised hierarchical clustering revealed two distinct groups of tumor samples that correlated loosely with tumor histology. Suraokar and colleagues used microarray and pathway analysis to define three molecular subgroups of MPM, which correlated only partially with histologic subtypes73.
Another analysis was published by de Reynies and colleagues in 201474. This group investigated microarray profiles of 67 MPM cell lines and generated two MPM subclasses (termed C1 and C2) partially related to histologic type and closely related to prognosis. These clusters were characterized by the differential expression of epithelial-to-mesenchymal (EMT) genes with C1 expressing an epithelial and C2 a mesenchymal phenotype. C1 was characterized by more frequent BAP1 and CDKN2A mutations, while C2 contained all of the sarcomatoid/desmoplastic samples among other subtypes of MPM. The authors created a predictor tool to discriminate samples between C1 and C2 using the expression levels of three genes: PPL, UPK3B, and TFPI. This tool was then used to validate the C1/C2 classification in 108 MPM tumor specimens with epithelioid and biphasic samples in both C1 and C2, and sarcomatoid samples only in C274.
In 2016, 211 MPM transcriptomes were characterized using unsupervised consensus clustering and four distinct molecular subtypes of MPM were identified: epithelioid, biphasic-E, biphasic-S, and sarcomatoid5. These associated to a degree with the spectrum from epithelioid to sarcomatoid histology. The 62% of histologically epithelioid samples classified into the biphasic-E, biphasic-S, or sarcomatoid clusters showed significantly lower overall survival than those in the epithelioid cluster indicating that epithelioid MPM can be distinguished into multiple different molecular groups. Differential expression analysis revealed that gene expression in the four clusters was related to a gradient of EMT, consistent with previous findings74. Further, a simple ratio of two genes, CLDN15 and VIM, was able to significantly differentiate the samples in the four clusters. Four (SETD2, TP53, NF2 and ULK2) of the most significantly mutated genes showed mutation rates significantly different between Cluster 1 and Clusters 2–475. Pathways implicated in this integrated analysis included histone methylation (consistent with previous findings e.g. Goto et al, 200934), Hippo, mTOR, RNA helicase and p53 signaling.
In 2018, TCGA performed integrated analysis of 74 MPM tumors including epigenetic, exomic, and transcriptomic profiles2. Integrative clustering performed using two separate algorithms (iCluster76 and PARADIGM77) identified four distinct subtypes of MPM in each. These were highly concordant, particularly with respect to the more extreme clusters 1 and 4. These two clusters correlated significantly with survival even when controlling for histologic subtype and deletion of CDKN2A78. Cluster 1 was enriched for epithelioid histology, while cluster 4 was enriched for sarcomatoid tumors similarly to the Bueno cohort5. Genes associated with epithelial-to-mesenchymal transition were again differentially expressed between clusters. In addition, each cluster was characterized by a distinct immune profile. In particular, Cluster 1 expressed the checkpoint inhibitor gene VISTA at high levels.
In an effort to deconvolute the signatures of epithelioid and sarcomatoid-like cell populations within bulk MPM samples, Blum and colleagues8 performed a meta-analysis using several publicly available datasets5,72,74,79,80. Initially, they used transcriptome data to classify 63 MPM samples into four distinct clusters (C1A, C1B, C2A, and C2B). Next, they compared the expression profile of each cluster with the previously published expression-based cluster data. They identified two highly correlated molecular groups among all datasets corresponding with the most extreme epithelioid and sarcomatoid subtypes. The intermediary tumors, however, did not form distinguishable clusters and therefore the authors suggest they reflect a continuum, or gradient, between epithelioid and sarcomatoid tumors. A panel of 150 common genes was used to generate two different scores, termed E-score and S-score, to determine the relative epithelioid-like and sarcomatoid-like molecular components present in individual tumors. Increased expression of UPK3B, MSLN, and CLDN15 was correlated with E-score and LOXL2 and VIM with the S-score. Pathway analysis revealed correlation of the S-score with EMT, TP53 signaling, cell cycle, angiogenesis, and immune checkpoints. The increasing sarcomatoid component identified by S-score was associated with worse outcomes in each series individually as well as in aggregate8.
Clinical Applications of Gene Expression
MPM can be challenging to diagnose. Pleural plaques are not diagnostic for mesothelioma, and as previously described the different MPM subtypes may be difficult to distinguish from other thoracic tumors on a histologic basis alone62. In addition, efforts to develop molecular predictors of clinical outcomes in MPM date back to the early 2000s corresponding with the rapid proliferation of novel and cost-effective sequencing technologies, but few are regularly employed in practice69,74,81–83.
The gene ratio-based method, developed by our laboratory, is able to overcome the difficulty of validating large gene signatures and offers improved clinical applicability75,84. Developed by comparing expression profiles between patients with different clinicopathologic parameters, these tests can then predict tumor characteristics or clinical outcomes based on a small number of genes66,75. With respect to diagnosis, Gordon and colleagues used 181 tissue samples to develop a six-gene three-ratio test to differentiate MPM from adenocarcinoma with 99% accuracy84. De Rienzo and colleagues used microarray data for 113 assorted MPM, non-MPM malignant, and benign samples develop a sequential combination of binary gene-expression ratio tests in frozen tissues to discern MPM from other thoracic cancers, as well as to distinguish epithelioid from sarcomatoid MPM85. Bruno and colleagues86 used NanoString technology to develop and validate a diagnostic tool employing 117 genes, of which 25 and 18 were up- and down-regulated in MPM respectively as compared with benign mesothelial hyperplasia. Designed to work with small quantities of RNA, this test could be performed on formalin-fixed paraffin-embedded (FFPE) specimens86.
Similar strategies have been applied to prognosis. For example, a 4-gene 3-ratio (TM4SF1/PKM2, TM4SF1/ARHGDIA, COBLL1/ARHGDIA) test was developed to predict treatment-related outcome independent of histology based on RT-PCR expression data83,84. While originally based on fresh-frozen tissue specimens, this score was later validated using FFPE tissue under Clinical Laboratory Improvement Amendments-approved guidelines in an independent multicenter cohort of MPM specimens85. It proved able to provide orthogonal risk information preoperatively and, postoperatively, predict overall survival when combined with histopathologic information.
In addition to gene ratio tests, expression-based molecular subtype74 and FAK protein expression87 have been shown to correlate with sensitivity to the targeted agents verteporfin and defactinib, respectively. However, these and other targeted agents have not succeeded in clinical trials and there are no current guidelines recommending their use20,29. Immunotherapy, while promising in several other cancer types, currently lacks biomarkers to predict efficacy in MPM as PD-L1 expression by IHC does not associate with treatment response88,89.
The Immune Microenvironment
Immunotherapy has expanded treatment options for tumors such as melanoma and non-small-cell lung cancer. Defining the immune microenvironment in MPM is an area of active investigation. In an early study, Burt et al demonstrated a significantly higher number of monocytes and tumor-infiltrating macrophages in non-epithelioid tumors90. This study also found a significant association between higher monocyte counts and shorter survival. The checkpoint ligand PD-L1 is expressed in almost 40% of MPM tumors by RNA-seq, with significantly higher expression in sarcomatoid tumors5. Expression of CTLA-4, another checkpoint molecule, was found in varying levels in 56% of MPM tumor samples by IHC, and higher in the epithelioid subtype91. In contrast, serum levels of soluble CTLA-4 were higher in patients with sarcomatoid disease as measured by ELISA91.
Expression of immune mediators can drive tumor biology. An analysis of 87 advanced-stage (III or IV) MPM tumors combining IHC for PD-L1 and NanoString analysis for 805 genes revealed PD-L1 expression in 16% of samples with significantly higher PD-L1 expression in sarcomatoid and biphasic samples92. Using hierarchical clustering by gene expression, these authors identified three subgroups of MPM: one with moderate T-cell effector gene expression but high B-cell gene expression (CD19, CD20); one with high PD-L1 expression and high T effector/T regulatory cell gene expression (including GZMA/GZMB, CXCL9, EOMES, FOXP3, ICOS, CTLA4); and one “immunologically ignorant” group with low expression of immune compartment-related genes but high stroma-related gene expression, including CTGF, DKK3, FN1, FAP, MMP2, and several genes encoding collagen subunits92. Taken together these results suggest heterogeneity in the interaction between MPM and the immune microenvironment that warrants further exploration.
6.3. Summary and Future Directions
MPM is a rare and aggressive cancer caused by asbestos exposure in the majority of cases. It is characterized by heterogeneity not only at the histologic but also the molecular level. Its hallmarks include widespread chromosomal loss, mutations in tumor suppressor genes such as BAP1, CDKN2A/2B, NF2, and TP53, and diverse transcriptomic phenotypes leading to several distinct molecular clusters. These clusters are defined at the extremes by epithelial and mesenchymal characteristics, with a histopathologic gradient stratifying the tumors in between. Multiple groups are working to develop predictive scores to classify individual tumors into these different subtypes, which have prognostic significance and may help guide choice of therapy.
Indeed, despite substantial advances in understanding the molecular biology of MPM, to date there have been relatively few changes in standard clinical practice based on these findings. MPM continues to present a diagnostic challenge and is often advanced at the time of detection. Histology remains the primary tool of prognostication in terms of overall survival and selection of therapeutic approach. Blunt, cytotoxic chemotherapy remains first line systemic treatment for a nuanced, recalcitrant, and biologically complex disease.
Ongoing work in the field of MPM molecular oncology will focus on deconvoluting the biological pathways involved in MPM tumorigenesis, growth, interaction with the tumor microenvironment, and response to therapy. Single-cell and single-nucleus transcriptomics have led to meaningful discoveries in several other cancers and offer the opportunity to define the contributions of individual tumor and immune/stromal cells to bulk tumor signatures. These techniques also provide a means to dissect intratumor heterogeneity and evaluate whether there are significant differences between malignant cells within an individual tumor, and how these might affect clinical outcomes.
Building on ever-expanding large datasets, deep learning and other advanced computational techniques are being used to integrate clinical, histopathologic and molecular data to refine diagnostic approaches and identify new prognostic biomarkers (Courtiol et al, 2019). New fields of study, such as proteomics and metabolomics, have yet to be incorporated into many of these analyses but show promise and merit further exploration (Sato et al 2018; Tomasetti et al, 2019). Finally, the development of unique molecular signatures for individual tumors will help guide treatment selection and identify approaches to meaningfully improve patient survival on an individualized basis.
Key Points:
Malignant pleural mesothelioma (MPM) is highly heterogeneous at the molecular level, leading to challenges in diagnosis, prognosis, and treatment.
MPM is associated with asbestos exposure in >80% of cases. Mechanisms of asbestos-associated tumorigenesis include reactive oxygen species (ROS), chronic inflammation, direct cytotoxicity, and cytokine and growth factor dysregulation.
Epigenetic hallmarks of MPM include widespread chromosomal loss and aberrant gene methylation, although these patterns are complex and variable.
MPM is characterized by the presence of fewer protein-altering somatic point-mutations compared with other cancers. Key mutated genes in MPM include tumor suppressors BAP1, NF2, CDKN2A/B, TP53, and SETD2.
Integrated multi-omic analyses identify up to four distinct clusters of MPM, with two extreme epithelioid-like and mesenchymal-like clusters separated by molecular gradient along the epithelial-to-mesenchymal transition spectrum.
Gene expression ratio tests and other molecular data can reliably improve diagnosis and prognostication in MPM.
Clinics Care Points.
More than 80% of MPM is caused by asbestos exposure.
MPM in patients with germline mutations is less aggressive and more chemotherapy-responsive than sporadic MPM, but these patients have a higher incidence of multiple other cancers.
Gene ratio tests can be useful in distinguishing MPM from other thoracic disease processes, as well as for predicting response to treatment and overall survival.
Transcriptomic and integrated multi-omic analyses can stratify MPM into distinct molecular clusters, which associate to a degree with histology and have independent implications for outcomes.
Biomarkers to identify candidates for targeted therapy or immunotherapy in MPM are currently lacking.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Disclosure Statement
The authors have nothing to disclose.
References
- 1.Broeckx G, Pauwels P. Malignant peritoneal mesothelioma: a review. Transl Lung Cancer Res 2018;7(5):537–42 doi: 10.21037/tlcr.2018.10.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hmeljak J, Sanchez-Vega F, Hoadley KA, et al. Integrative Molecular Characterization of Malignant Pleural Mesothelioma. Cancer Discov 2018;8(12):1548–65 doi: 10.1158/2159-8290.CD-18-0804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Tischoff I, Neid M, Neumann V, Tannapfel A. Pathohistological diagnosis and differential diagnosis. Recent Results Cancer Res 2011;189:57–78 doi: 10.1007/978-3-642-10862-4_5. [DOI] [PubMed] [Google Scholar]
- 4.Vigneswaran WT, Kircheva DY, Ananthanarayanan V, et al. Amount of Epithelioid Differentiation Is a Predictor of Survival in Malignant Pleural Mesothelioma. Ann Thorac Surg 2017;103(3):962–66 doi: 10.1016/j.athoracsur.2016.08.063. [DOI] [PubMed] [Google Scholar]
- 5.Bueno R, Stawiski EW, Goldstein LD, et al. Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nat Genet 2016;48(4):407–16 doi: 10.1038/ng.3520. [DOI] [PubMed] [Google Scholar]
- 6.Yap TA, Aerts JG, Popat S, Fennell DA. Novel insights into mesothelioma biology and implications for therapy. Nat Rev Cancer 2017;17(8):475–88 doi: 10.1038/nrc.2017.42. [DOI] [PubMed] [Google Scholar]
- 7.Oehl K, Vrugt B, Opitz I, Meerang M. Heterogeneity in Malignant Pleural Mesothelioma. Int J Mol Sci 2018;19(6) doi: 10.3390/ijms19061603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Blum Y, Meiller C, Quetel L, et al. Dissecting heterogeneity in malignant pleural mesothelioma through histo-molecular gradients for clinical applications. Nat Commun 2019;10(1):1333 doi: 10.1038/s41467-019-09307-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Network NCC. Malignant Pleural Mesothelioma, version 2.2015. Secondary Malignant Pleural Mesothelioma, version 2.2015 2015 http://www.hts.org.gr/assets/files/omades_ergasias/cancer/NCCN%20guidelines%20Mesothelicoma%202015.pdf.
- 10.Khan S, Gerber DE. Autoimmunity, checkpoint inhibitor therapy and immune-related adverse events: A review. Semin Cancer Biol 2019. doi: 10.1016/j.semcancer.2019.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gomez DR, Rimner A, Simone CB 2nd, et al. The Use of Radiation Therapy for the Treatment of Malignant Pleural Mesothelioma: Expert Opinion 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 2019;14(7):1172–83 doi: 10.1016/j.jtho.2019.03.030. [DOI] [PubMed] [Google Scholar]
- 12.Attanoos RL, Churg A, Galateau-Salle F, Gibbs AR, Roggli VL. Malignant Mesothelioma and Its Non-Asbestos Causes. Arch Pathol Lab Med 2018;142(6):753–60 doi: 10.5858/arpa.2017-0365-RA. [DOI] [PubMed] [Google Scholar]
- 13.Galani V, Varouktsi A, Papadatos SS, et al. The role of apoptosis defects in malignant mesothelioma pathogenesis with an impact on prognosis and treatment. Cancer Chemother Pharmacol 2019;84(2):241–53 doi: 10.1007/s00280-019-03878-3. [DOI] [PubMed] [Google Scholar]
- 14.Loomis D, Richardson DB, Elliott L. Quantitative relationships of exposure to chrysotile asbestos and mesothelioma mortality. Am J Ind Med 2019;62(6):471–77 doi: 10.1002/ajim.22985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sun HH, Vaynblat A, Pass HI. Diagnosis and prognosis-review of biomarkers for mesothelioma. Ann Transl Med 2017;5(11):244 doi: 10.21037/atm.2017.06.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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]
- 17.Tomasetti M, Gaetani S, Monaco F, Neuzil J, Santarelli L. Epigenetic Regulation of miRNA Expression in Malignant Mesothelioma: miRNAs as Biomarkers of Early Diagnosis and Therapy. Front Oncol 2019;9:1293 doi: 10.3389/fonc.2019.01293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Casalone E, Allione A, Viberti C, et al. DNA methylation profiling of asbestos-treated MeT5A cell line reveals novel pathways implicated in asbestos response. Arch Toxicol 2018;92(5):1785–95 doi: 10.1007/s00204-018-2179-y. [DOI] [PubMed] [Google Scholar]
- 19.Nuvoli B, Camera E, Mastrofrancesco A, Briganti S, Galati R. Modulation of reactive oxygen species via ERK and STAT3 dependent signalling are involved in the response of mesothelioma cells to exemestane. Free Radic Biol Med 2018;115:266–77 doi: 10.1016/j.freeradbiomed.2017.12.008. [DOI] [PubMed] [Google Scholar]
- 20.Zucali PA, Ceresoli GL, De Vincenzo F, et al. Advances in the biology of malignant pleural mesothelioma. Cancer Treat Rev 2011;37(7):543–58 doi: 10.1016/j.ctrv.2011.01.001. [DOI] [PubMed] [Google Scholar]
- 21.Pociask DA, Sime PJ, Brody AR. Asbestos-derived reactive oxygen species activate TGF-beta1. Lab Invest 2004;84(8):1013–23 doi: 10.1038/labinvest.3700109. [DOI] [PubMed] [Google Scholar]
- 22.Kamp DW, Weitzman SA. The molecular basis of asbestos induced lung injury. Thorax 1999;54(7):638–52 doi: 10.1136/thx.54.7.638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Crovella S, Moura RR, Cappellani S, et al. A genetic variant of NLRP1 gene is associated with asbestos body burden in patients with malignant pleural mesothelioma. J Toxicol Environ Health A 2018;81(5):98–105 doi: 10.1080/15287394.2017.1416911. [DOI] [PubMed] [Google Scholar]
- 24.Celsi F, Crovella S, Moura RR, et al. Pleural mesothelioma and lung cancer: the role of asbestos exposure and genetic variants in selected iron metabolism and inflammation genes. J Toxicol Environ Health A 2019;82(20):1088–102 doi: 10.1080/15287394.2019.1694612. [DOI] [PubMed] [Google Scholar]
- 25.Carbone M, Ly BH, Dodson RF, et al. Malignant mesothelioma: facts, myths, and hypotheses. J Cell Physiol 2012;227(1):44–58 doi: 10.1002/jcp.22724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jean D, Daubriac J, Le Pimpec-Barthes F, Galateau-Salle F, Jaurand MC. Molecular changes in mesothelioma with an impact on prognosis and treatment. Arch Pathol Lab Med 2012;136(3):277–93 doi: 10.5858/arpa.2011-0215-RA. [DOI] [PubMed] [Google Scholar]
- 27.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):264–9 doi: 10.1158/0008-5472.CAN-14-1008. [DOI] [PubMed] [Google Scholar]
- 28.Krismann M, Muller KM, Jaworska M, Johnen G. Molecular cytogenetic differences between histological subtypes of malignant mesotheliomas: DNA cytometry and comparative genomic hybridization of 90 cases. J Pathol 2002;197(3):363–71 doi: 10.1002/path.1128. [DOI] [PubMed] [Google Scholar]
- 29.Hylebos M, Van Camp G, van Meerbeeck JP, Op de Beeck K. The Genetic Landscape of Malignant Pleural Mesothelioma: Results from Massively Parallel Sequencing. J Thorac Oncol 2016;11(10):1615–26 doi: 10.1016/j.jtho.2016.05.020. [DOI] [PubMed] [Google Scholar]
- 30.Bott M, Brevet M, Taylor BS, et al. The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma. Nat Genet 2011;43(7):668–72 doi: 10.1038/ng.855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lindholm PM, Salmenkivi K, Vauhkonen H, et al. Gene copy number analysis in malignant pleural mesothelioma using oligonucleotide array CGH. Cytogenet Genome Res 2007;119(1–2):46–52 doi: 10.1159/000109618. [DOI] [PubMed] [Google Scholar]
- 32.Borczuk AC, Pei J, Taub RN, et al. Genome-wide analysis of abdominal and pleural malignant mesothelioma with DNA arrays reveals both common and distinct regions of copy number alteration. Cancer Biol Ther 2016;17(3):328–35 doi: 10.1080/15384047.2016.1145850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Tsou JA, Galler JS, Wali A, et al. DNA methylation profile of 28 potential marker loci in malignant mesothelioma. Lung Cancer 2007;58(2):220–30 doi: 10.1016/j.lungcan.2007.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Goto Y, Shinjo K, Kondo Y, et al. Epigenetic profiles distinguish malignant pleural mesothelioma from lung adenocarcinoma. Cancer Res 2009;69(23):9073–82 doi: 10.1158/0008-5472.CAN-09-1595. [DOI] [PubMed] [Google Scholar]
- 35.Christensen BC, Houseman EA, Godleski JJ, et al. Epigenetic profiles distinguish pleural mesothelioma from normal pleura and predict lung asbestos burden and clinical outcome. Cancer Res 2009;69(1):227–34 doi: 10.1158/0008-5472.CAN-08-2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Laszlo V, Hoda MA, Garay T, et al. Epigenetic down-regulation of integrin alpha7 increases migratory potential and confers poor prognosis in malignant pleural mesothelioma. J Pathol 2015;237(2):203–14 doi: 10.1002/path.4567. [DOI] [PubMed] [Google Scholar]
- 37.Christensen BC, Houseman EA, Poage GM, et al. Integrated profiling reveals a global correlation between epigenetic and genetic alterations in mesothelioma. Cancer Res 2010;70(14):5686–94 doi: 10.1158/0008-5472.CAN-10-0190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Reid G, Johnson TG, van Zandwijk N. Manipulating microRNAs for the Treatment of Malignant Pleural Mesothelioma: Past, Present and Future. Front Oncol 2020;10:105 doi: 10.3389/fonc.2020.00105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Quetel L, Tranchant R, Meiller C, et al. Abstract 112: Genetic alterations in molecular tumor subgroups of malignant pleural mesothelioma. Cancer Research 2016;76(14 Supplement):112–12 doi: 10.1158/1538-7445.Am2016-112. [DOI] [Google Scholar]
- 40.Cigognetti M, Lonardi S, Fisogni S, et al. BAP1 (BRCA1-associated protein 1) is a highly specific marker for differentiating mesothelioma from reactive mesothelial proliferations. Mod Pathol 2015;28(8):1043–57 doi: 10.1038/modpathol.2015.65. [DOI] [PubMed] [Google Scholar]
- 41.Ladanyi M, Robinson BW, Campbell PJ, Network ftTR. The TCGA malignant pleural mesothelioma (MPM) project: VISTA expression and delineation of a novel clinical-molecular subtype of MPM. Journal of Clinical Oncology 2018;36(15_suppl):8516–16 doi: 10.1200/JCO.2018.36.15_suppl.8516. [DOI] [Google Scholar]
- 42.Carbone M, Yang H, Pass HI, Krausz T, Testa JR, Gaudino G. BAP1 and cancer. Nat Rev Cancer 2013;13(3):153–9 doi: 10.1038/nrc3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yu H, Pak H, Hammond-Martel I, et al. Tumor suppressor and deubiquitinase BAP1 promotes DNA double-strand break repair. Proc Natl Acad Sci U S A 2014;111(1):285–90 doi: 10.1073/pnas.1309085110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.De Rienzo A, Archer MA, Yeap BY, et al. Gender-Specific Molecular and Clinical Features Underlie Malignant Pleural Mesothelioma. Cancer Res 2016;76(2):319–28 doi: 10.1158/0008-5472.CAN-15-0751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bononi A, Giorgi C, Patergnani S, et al. BAP1 regulates IP3R3-mediated Ca(2+) flux to mitochondria suppressing cell transformation. Nature 2017;546(7659):549–53 doi: 10.1038/nature22798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.McCambridge AJ, Napolitano A, Mansfield AS, et al. Progress in the Management of Malignant Pleural Mesothelioma in 2017. J Thorac Oncol 2018;13(5):606–23 doi: 10.1016/j.jtho.2018.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Righi L, Duregon E, Vatrano S, et al. BRCA1-Associated Protein 1 (BAP1) Immunohistochemical Expression as a Diagnostic Tool in Malignant Pleural Mesothelioma Classification: A Large Retrospective Study. J Thorac Oncol 2016;11(11):2006–17 doi: 10.1016/j.jtho.2016.06.020. [DOI] [PubMed] [Google Scholar]
- 48.LaFave LM, Beguelin W, Koche R, et al. Loss of BAP1 function leads to EZH2-dependent transformation. Nat Med 2015;21(11):1344–9 doi: 10.1038/nm.3947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sage AP, Martinez VD, Minatel BC, et al. Genomics and Epigenetics of Malignant Mesothelioma. High Throughput 2018;7(3) doi: 10.3390/ht7030020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Jean D, Jaurand M-C. Causes and pathophysiology of malignant pleural mesothelioma. Lung Cancer Management 2015;4(5):219–29 doi: 10.2217/lmt.15.21. [DOI] [Google Scholar]
- 51.Tranchant R, Quetel L, Tallet A, et al. Co-occurring Mutations of Tumor Suppressor Genes, LATS2 and NF2, in Malignant Pleural Mesothelioma. Clin Cancer Res 2017;23(12):3191–202 doi: 10.1158/1078-0432.CCR-16-1971. [DOI] [PubMed] [Google Scholar]
- 52.Panou V, Gadiraju M, Wolin A, et al. Frequency of Germline Mutations in Cancer Susceptibility Genes in Malignant Mesothelioma. J Clin Oncol 2018;36(28):2863–71 doi: 10.1200/JCO.2018.78.5204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hassan R, Morrow B, Thomas A, et al. Inherited predisposition to malignant mesothelioma and overall survival following platinum chemotherapy. Proc Natl Acad Sci U S A 2019;116(18):9008–13 doi: 10.1073/pnas.1821510116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Guo R, DuBoff M, Jayakumaran G, et al. Novel Germline Mutations in DNA Damage Repair in Patients with Malignant Pleural Mesotheliomas. J Thorac Oncol 2019. doi: 10.1016/j.jtho.2019.12.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Carbone M, Adusumilli PS, Alexander HR Jr., et al. Mesothelioma: Scientific clues for prevention, diagnosis, and therapy. CA Cancer J Clin 2019;69(5):402–29 doi: 10.3322/caac.21572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Testa JR, Cheung M, Pei J, et al. Germline BAP1 mutations predispose to malignant mesothelioma. Nat Genet 2011;43(10):1022–5 doi: 10.1038/ng.912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Luchini C, Veronese N, Yachida S, et al. Different prognostic roles of tumor suppressor gene BAP1 in cancer: A systematic review with meta-analysis. Genes Chromosomes Cancer 2016;55(10):741–9 doi: 10.1002/gcc.22381. [DOI] [PubMed] [Google Scholar]
- 58.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Walpole S, Pritchard AL, Cebulla CM, et al. Comprehensive Study of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families Worldwide. J Natl Cancer Inst 2018;110(12):1328–41 doi: 10.1093/jnci/djy171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Pulford E, Huilgol K, Moffat D, Henderson DW, Klebe S. Malignant Mesothelioma, BAP1 Immunohistochemistry, and VEGFA: Does BAP1 Have Potential for Early Diagnosis and Assessment of Prognosis? Dis Markers 2017;2017:1310478 doi: 10.1155/2017/1310478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Parrotta R, Okonska A, Ronner M, et al. A Novel BRCA1-Associated Protein-1 Isoform Affects Response of Mesothelioma Cells to Drugs Impairing BRCA1-Mediated DNA Repair. J Thorac Oncol 2017;12(8):1309–19 doi: 10.1016/j.jtho.2017.03.023. [DOI] [PubMed] [Google Scholar]
- 62.Ray M, Kindler HL. Malignant pleural mesothelioma: an update on biomarkers and treatment. Chest 2009;136(3):888–96 doi: 10.1378/chest.08-2665. [DOI] [PubMed] [Google Scholar]
- 63.Churg A, Nabeshima K, Ali G, Bruno R, Fernandez-Cuesta L, Galateau-Salle F. Highlights of the 14th international mesothelioma interest group meeting: Pathologic separation of benign from malignant mesothelial proliferations and histologic/molecular analysis of malignant mesothelioma subtypes. Lung Cancer 2018;124:95–101 doi: 10.1016/j.lungcan.2018.07.041. [DOI] [PubMed] [Google Scholar]
- 64.Galateau Salle F, Le Stang N, Nicholson AG, et al. New Insights on Diagnostic Reproducibility of Biphasic Mesotheliomas: A Multi-Institutional Evaluation by the International Mesothelioma Panel From the MESOPATH Reference Center. J Thorac Oncol 2018;13(8):1189–203 doi: 10.1016/j.jtho.2018.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Uhlen M, Zhang C, Lee S, et al. A pathology atlas of the human cancer transcriptome. Science 2017;357(6352) doi: 10.1126/science.aan2507. [DOI] [PubMed] [Google Scholar]
- 66.Gordon GJ. Transcriptional profiling of mesothelioma using microarrays. Lung Cancer 2005;49 Suppl 1:S99–S103 doi: 10.1016/j.lungcan.2005.03.018. [DOI] [PubMed] [Google Scholar]
- 67.Rihn BH, Mohr S, McDowell SA, et al. Differential gene expression in mesothelioma. FEBS Lett 2000;480(2–3):95–100 doi: 10.1016/s0014-5793(00)01913-x. [DOI] [PubMed] [Google Scholar]
- 68.Singhal S, Wiewrodt R, Malden LD, et al. Gene expression profiling of malignant mesothelioma. Clin Cancer Res 2003;9(8):3080–97 [PubMed] [Google Scholar]
- 69.Pass HI, Liu Z, Wali A, et al. Gene expression profiles predict survival and progression of pleural mesothelioma. Clin Cancer Res 2004;10(3):849–59 doi: 10.1158/1078-0432.ccr-0607-3. [DOI] [PubMed] [Google Scholar]
- 70.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):615–23 doi: 10.1002/gcc.20669. [DOI] [PubMed] [Google Scholar]
- 71.Melaiu O, Cristaudo A, Melissari E, et al. A review of transcriptome studies combined with data mining reveals novel potential markers of malignant pleural mesothelioma. Mutat Res 2012;750(2):132–40 doi: 10.1016/j.mrrev.2011.12.003. [DOI] [PubMed] [Google Scholar]
- 72.Gordon GJ, Rockwell GN, Jensen RV, et al. Identification of novel candidate oncogenes and tumor suppressors in malignant pleural mesothelioma using large-scale transcriptional profiling. Am J Pathol 2005;166(6):1827–40 doi: 10.1016/S0002-9440(10)62492-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Suraokar MB, Nunez MI, Diao L, et al. Expression profiling stratifies mesothelioma tumors and signifies deregulation of spindle checkpoint pathway and microtubule network with therapeutic implications. Ann Oncol 2014;25(6):1184–92 doi: 10.1093/annonc/mdu127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.de Reynies A, Jaurand MC, Renier A, et al. Molecular classification of malignant pleural mesothelioma: identification of a poor prognosis subgroup linked to the epithelial-to-mesenchymal transition. Clin Cancer Res 2014;20(5):1323–34 doi: 10.1158/1078-0432.CCR-13-2429. [DOI] [PubMed] [Google Scholar]
- 75.De Rienzo A, Richards W, Bueno R. Gene Signature of Malignant Pleural Mesothelioma, 2017:197–209. [Google Scholar]
- 76.Shen R, Olshen AB, Ladanyi M. Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis. Bioinformatics 2009;25(22):2906–12 doi: 10.1093/bioinformatics/btp543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Vaske CJ, Benz SC, Sanborn JZ, et al. Inference of patient-specific pathway activities from multi-dimensional cancer genomics data using PARADIGM. Bioinformatics 2010;26(12):i237–45 doi: 10.1093/bioinformatics/btq182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Dacic S, Kothmaier H, Land S, et al. Prognostic significance of p16/cdkn2a loss in pleural malignant mesotheliomas. Virchows Arch 2008;453(6):627–35 doi: 10.1007/s00428-008-0689-3. [DOI] [PubMed] [Google Scholar]
- 79.Lopez-Rios F, Chuai S, Flores R, et al. Global gene expression profiling of pleural mesotheliomas: overexpression of aurora kinases and P16/CDKN2A deletion as prognostic factors and critical evaluation of microarray-based prognostic prediction. Cancer Res 2006;66(6):2970–9 doi: 10.1158/0008-5472.CAN-05-3907. [DOI] [PubMed] [Google Scholar]
- 80.Center BITGDA. Analysis-ready standardized TCGA data from Broad GDAC Firehose stddata__2015_06_01run. In: Harvard BIoMa, ed., 2015. [Google Scholar]
- 81.Gordon GJ, Jensen RV, Hsiao LL, et al. Using gene expression ratios to predict outcome among patients with mesothelioma. J Natl Cancer Inst 2003;95(8):598–605 doi: 10.1093/jnci/95.8.598. [DOI] [PubMed] [Google Scholar]
- 82.Gordon GJ, Rockwell GN, Godfrey PA, et al. Validation of genomics-based prognostic tests in malignant pleural mesothelioma. Clin Cancer Res 2005;11(12):4406–14 doi: 10.1158/10780432.CCR-04-2181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Gordon GJ, Dong L, Yeap BY, et al. Four-gene expression ratio test for survival in patients undergoing surgery for mesothelioma. J Natl Cancer Inst 2009;101(9):678–86 doi: 10.1093/jnci/djp061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gordon GJ, Jensen RV, Hsiao LL, et al. Translation of microarray data into clinically relevant cancer diagnostic tests using gene expression ratios in lung cancer and mesothelioma. Cancer Res 2002;62(17):4963–7 [PubMed] [Google Scholar]
- 85.De Rienzo A, Cook RW, Wilkinson J, et al. Validation of a Gene Expression Test for Mesothelioma Prognosis in Formalin-Fixed Paraffin-Embedded Tissues. J Mol Diagn 2017;19(1):65–71 doi: 10.1016/j.jmoldx.2016.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bruno R, Ali G, Giannini R, et al. Malignant pleural mesothelioma and mesothelial hyperplasia: A new molecular tool for the differential diagnosis. Oncotarget 2017;8(2):2758–70 doi: 10.18632/oncotarget.13174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Tranchant R, Quetel L, Montagne F, et al. Assessment of signaling pathway inhibitors and identification of predictive biomarkers in malignant pleural mesothelioma. Lung Cancer 2018;126:15–24 doi: 10.1016/j.lungcan.2018.10.015. [DOI] [PubMed] [Google Scholar]
- 88.Nowak AK, McDonnell A, Cook A. Immune checkpoint inhibition for the treatment of mesothelioma. Expert Opin Biol Ther 2019;19(7):697–706 doi: 10.1080/14712598.2019.1606209. [DOI] [PubMed] [Google Scholar]
- 89.Alley EW, Lopez J, Santoro A, et al. 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(5):623–30 doi: 10.1016/S1470-2045(17)30169-9. [DOI] [PubMed] [Google Scholar]
- 90.Burt BM, Rodig SJ, Tilleman TR, Elbardissi AW, Bueno R, Sugarbaker DJ. Circulating and tumor-infiltrating myeloid cells predict survival in human pleural mesothelioma. Cancer 2011;117(22):5234–44 doi: 10.1002/cncr.26143. [DOI] [PubMed] [Google Scholar]
- 91.Roncella S, Laurent S, Fontana V, et al. CTLA-4 in mesothelioma patients: tissue expression, body fluid levels and possible relevance as a prognostic factor. Cancer Immunol Immunother 2016;65(8):909–17 doi: 10.1007/s00262-016-1844-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Patil NS, Righi L, Koeppen H, et al. Molecular and Histopathological Characterization of the Tumor Immune Microenvironment in Advanced Stage of Malignant Pleural Mesothelioma. J Thorac Oncol 2018;13(1):124–33 doi: 10.1016/j.jtho.2017.09.1968. [DOI] [PubMed] [Google Scholar]