Skip to main content

EDITORIAL article

Front. Toxicol., 03 January 2025
Sec. Immunotoxicology
This article is part of the Research Topic Asbestos and disease genomics: is mesothelioma a genomic paradigm? View all 5 articles

Editorial: Asbestos and disease genomics: is mesothelioma a genomic paradigm?

  • 1Université de Paris, Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Functional Genomics of Solid Tumors, Paris, France
  • 2Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
  • 3Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland

1 Introduction

A paradigm is a set of assumptions that creates a viewpoint of the world. However, as summarized by Carlo Rovelli (Carlo Rovelli, 2016), “scientific knowledge is the process of continuously modifying and improving our conception of the world, selectively and constantly questioning the assumptions on which it is based.” In this Editorial, we aim to challenge the current assumptions surrounding the concept of mesothelioma as a genomic disease through a comprehensive review of our understanding of disease pathogenesis and integration of recent findings reported in this Research Topic (Leinardi et al.; Sekido and Sato; Farahmand et al.; Fisher et al.).

The recognition of asbestos fibers as lung and pleural carcinogens has been long established from epidemiological data and is supported by experimental studies. The clear link between fiber inhalation and cancer raised important questions on the carcinogenic mechanism of action of asbestos fibers in exposed populations. Concomitantly, experimental studies of the pathological consequences of exposure to asbestos unraveled a role for specific features of fibers in disease pathogenesis. The integration of this knowledge is expressed in the fiber pathogenicity paradigm (FPP), which directly links the dimensions (diameter and length) and biopersistence of the fiber, in addition to dose, to mesothelioma hazard (Van Dorn, 2017; Rose, 2022). Advancing this work led to the formulation of a concept of “elongated mineral particles (EMPs)” to classify fiber types that share similar features with asbestos (Institute of Medicine, 2009). This concept triggered further studies to investigate the potential pathological potency of EMPs in relation to the asbestos hazard, especially at the thoracic level, and more recently flagged concerns over potential increasing exposure to novel engineered materials as a result of advances in (nano)technological developments of manufactured fibers (Nel, 2023).

2 Brief history

2.1 Discovery of asbestos-related diseases

Asbestos mining on an industrial scale started at the end of the 1800s, with the development of mesothelioma in exposed miners first reported 50 years later (Wagner et al., 1960). The use of asbestos has been regulated in many countries since 1995; however, several developed and developing countries continue to mine, export, and use asbestos in high volumes (Frank, 2020), with approximately 1,200,000 metric tons of asbestos used worldwide in 2021 (USGS, 2022 https://pubs.usgs.gov/publication/mcs2022). Over time, the risk of exposure identified at the workplace has been extended to using asbestos-containing material and during efforts to remove asbestos from existing structures (Gottesfeld, 2024). Secondary exposure scenarios have become an increasing cause for concern as many asbestos-containing buildings constructed in the mid-twentieth century are reaching their end of life and will require significant repair, reconstruction, or demolition in the near future. These existing and emerging threats have the potential to increase asbestos exposure in more diverse populations and continue the risk for the development of asbestos-related diseases (Alpert et al., 2020; Singh and Frank, 2023; Metintas et al., 2024).

Our historical experience with asbestos has left us with a heavy burden of disease. Lessons, however, have been learned that are relevant to both the scientific context, including an increased understanding of the mechanism underpinning fiber toxicity, and from an ethical perspective. Recognition of the long latency period from initial exposure to disease development and the lack of compensatory mechanisms within the body to neutralize pathogenic fibers highlights the need to address the humanitarian concern of ongoing exposure and anticipate health risks before developing new industrial procedures utilizing EMPs and during the design of novel fiber-like materials.

2.2 The remarkable characteristics of asbestos fibers related to toxicity

Experimental studies have revealed that fiber morphology and physico-chemical properties modulate the biological effects of asbestos fibers, emphasizing the role of fiber dimensions, especially length (Stanton et al., 1981). Additionally, in vitro acellular systems have been used to quantify the fiber dissolution rate and confirm the role of fiber durability in the biopersistence of pathogenic fibers. Epidemiological studies have substantiated these findings, especially on the relevance of fiber dimensions. From this research, it can be summarized that size, chemistry, and surface reactivity are basic parameters that govern toxicity. They are involved in biological responses such as fiber uptake and phagocytosis, interactions with biological molecules, genetic alterations, inflammation, immunity, translocation processes, and biopersistence (Sayan and Mossman, 2015; Nagai and Toyokuni, 2010; Donaldson et al., 2010; Kuroda, 2021; Huang et al., 2011). Further research has added several fiber parameters that are involved in the list of toxicological effects, such as rigidity. This information has been used to develop models to predict a mesothelioma potency hazard based on fiber dimensions (Nel, 2023; Wylie and Korchevskiy, 2023) or, with further refinement, the “fiber potential toxicity/pathogenicity index (FPTI),” which includes 18 parameters associated with an adverse effect in the pathological process (Gualtieri, 2018; Wylie and Korchevskiy, 2023).

The FPP is now being applied to assess the carcinogenic potential of fibrous particles such as glass and refractory fibers, carbon nanotubes (CNTs), metallic fibers, and new manufactured particles (high aspect ratio nanomaterials, or HARNs) (Nel, 2023; Murphy et al., 2021; Kane et al., 2018).

The FPP has left us with the legacy of continuing research into asbestos toxicity mechanisms and studying EMPs and HARNs to avoid health damage.

3 Mesothelial cell response to asbestos and mesothelioma characteristics

3.1 Early pleural responses to asbestos fibers

Experimental research has demonstrated a translocation of inhaled asbestos fibers toward the pleural space, although the mechanism is not fully understood (Miserocchi et al., 2008). The retention of asbestos fibers in the pleural cavity seems partly related to the size of stomata or pores through which pleural fluid drains to the lymphatic system (from 0.8 µm in mice to 10 µm in humans) (Schinwald et al., 2012). Subsequent accumulation of long fibers in the pleural cavity leads to direct irritation of the mesothelial layer, frustrated phagocytosis of pleural macrophages, and inflammation. Direct instillation of CNTs and other high-aspect ratio nanomaterials (HARN) into the pleural space have demonstrated a similar pathogenicity and mechanism of action to asbestos fibers in terms of production of oxidative stress, inflammation, and genotoxicity (Donaldson et al., 2013; Yoshida, 2019; Nagai and Toyokuni, 2010).

The mesothelial cell response to asbestos fibers was studied in cell culture models, including an SV40-immortalized, non-tumorigenic human mesothelial cell line. Normal mesothelial cells internalize the fibers, and chrysotile fibers were found in phagolysosomes with a lysosome degranulation (Nagai and Toyokuni, 2012). Inflammatory factors shown to be released in vitro by mesothelial cells may propagate a chronic inflammatory environment in vivo, subjecting mesothelial cells to ongoing oxidative stress, which may eventually result in cell transformation (Sayan and Mossman, 2015). In this Research Topic, Leinardi et al. provide a comprehensive review of the role inflammatory components released from cells after cell death can contribute to chronic disease development in the context of silica exposure. While silica induces disease by the release of pro-inflammatory damage-associated molecular patterns, including HMGB1 from macrophages, asbestos carcinogenesis can be promoted by the release of HMGB1 directly from mesothelial cells (Suarez et al., 2023).

To examine early changes along the mesothelium in response to fibers, a transcriptomic kinetic analysis of mesothelial cells exposed by injection of fibers into the pleural cavity of C57BL/6 mice was carried out at timepoints between 1 week up to 20 months after exposure (Chernova et al., 2017). Samples consisted of long and straight CNTs, short CNTs, and long (carcinogenic) and short (lower pathogenicity) amosite asbestos fibers (Chernova et al., 2017). Inflammatory lesions studied from 1 week to 6 months after injection were similar in mice exposed to both long samples in terms of cell components in the pleural cavity and expression of inflammatory response genes, whereas gene expression from mice exposed to short fibers and controls clustered together. Inflammatory response pathways and activation of kinases involved in pro-oncogenic pathways were identified in early lesions with dysregulation maintained through to tumor development. The status of the Cdkn2a gene (encoding two proteins, p16Ink4a and p19ARF), an ortholog of the key tumor suppressor genes (TSGs) in human mesothelioma CDKN2A (encoding P16INK4a and P14ARF), was examined in inflammatory lesions at 1-year post injection before tumors developed and in tumors induced by exposure to long CNT and amosite asbestos (Chernova et al., 2017). The authors reported silencing of Cdkn2a (Ink4a/Arf) by hypermethylation and co-deletion of the proteins in the fiber-induced inflammatory lesions that increased in tumors, which also acquired a p19/Arf deletion. This shows that epigenetic changes are present early in an inflammatory, pre-tumoral stage and emphasizes the similarities with human pleural mesothelioma (PM) (Chernova et al., 2017).

Transcriptome analyses of asbestos-induced inflamed tissue were investigated in heterozygous Nƒ2+/−C57Bl6 mice intraperitoneally exposed to crocidolite fibers for 12 weeks and assessed up to 33 weeks after the last exposure (Rehrauer et al., 2018). They revealed a decreased level of Nƒ2 expression and activation of Yap/Taz localized in the cell nucleus in inflamed mesothelium, which increased in tumors, indicating deregulation of the Hippo pathway in these mice (Rehrauer et al., 2018). Although conducted in a genetically modified mouse model to increase mesothelioma susceptibility, this study highlights the potential role that dysregulation of the Hippo pathway due to Nf2 mutation plays in the progression of mesothelioma. Although Nf2 loss is regularly identified in mesothelioma tumors reviewed in the article by Sekido and Sato as part of this Research Topic, in human disease, the NF2 mutation appears to be a late event, indicating that genomic damage of NF2 may not be a direct asbestos effect but a result of the chronic inflammatory and oxidative environment.

3.2 Histopathology and molecular genetic characterization of PM

The characteristics of human PM are continuously evolving and concern several fields of cell and tumor biology, from histological classification, genetic, epigenetic, and chromosomal status, state of regulatory pathways, and cell-to-cell interactions with the immunological microenvironment.

The recent histological classification of PM retains the three main histologic subtypes: epithelioid, biphasic, and sarcomatoid, with biphasic including epithelioid and sarcomatoid elements. Mesothelioma subtypes show a variety of architectures, cellular aspects, and stroma, and their prognosis is different, with a worse survival for the sarcomatoid subtype than the epithelioid (Sauter et al., 2022; Husain et al., 2024). There was no epidemiological evidence of an association between the histological classification of mesothelioma and exposure to a given type of asbestos fibers (Vorster et al., 2022; Franklin et al., 2016). However, in a genetically engineered conditional mouse model, where mostly sarcomatoid mesothelioma develops spontaneously after co-deletion of Nf2, p53, and Cdkn2a in mesothelial cells, asbestos exposure accelerates the onset of mostly epithelioid tumors (Farahmand et al.). One notable feature of the response to asbestos exposure in this genetically modified mouse model is the increased recruitment of macrophages observed as a precursor to mesothelioma development. This raises the possibility that the tumor microenvironment and epigenetic events downstream of asbestos exposure provide cues favoring proliferation when compared to the tumor developing in the absence of asbestos. This is also supported by the observation that different methylation levels of CpG sites were detected within tumors and were reflective of intratumor heterogeneity of the histological subtype. DNA methylation was preferentially located in CpG islands for the sarcomatoid subtype while mainly located in non-CpG islands for the areas with high epithelioid histology, suggesting that histo-molecular gradients are linked to epigenetic regulation (Blum et al., 2019).

While there are limited studies showing the effects of asbestos fibers on the regulation of gene expression after short-term exposure of mesothelial cells to asbestos fibers, there is currently a large body of data on the pathological and biomolecular characteristics of PM. They are usually investigated long after the onset of the tumors, which are biopsied a long time after the beginning of exposure, possibly several decades.

Investigations of the molecular landscape of mesothelioma began in the 2000s with the development of methodologies for large-scale analytical methods that provide high-throughput analysis of biological data. The genetic and epigenetic modifications of the tumors were studied using multi-omic approaches, such as next-generation sequencing and microarrays. As observed in histology, PM is a heterogeneous tumor, with a rather low number of somatic gene mutations compared to other cancers, but with a high number and types of chromosomal rearrangements, copy number alterations, genome duplication, and mutations in a number of key genes, most of them being TSGs (CDKN2A, BAP1, NF2, SETD2, LATS2, and TP53) and a mutation in the TERT promoter (Bueno et al., 2016; Meiller et al., 2021; Febres-Aldana et al., 2024; Creaney et al., 2022; Nair et al., 2023; Mangiante et al., 2023). At a lower rate, mutations were detected in genes of the SWI/SNF family (ARID1A, ARID2, and SMARCA4) and genes related to histone methylation (KMT2D and SETD2) were mutually exclusive (Quetel et al., 2020; Febres-Aldana et al., 2024).

Transcriptomic analyses revealed that the heterogeneous molecular profiles of PM could be identified allowing a molecular classification of pleural PM. This refined classification identified several subgroups characterized by different molecular profiles and gene alterations that distinguish the epithelioid from the sarcomatoid phenotype and were linked to the patients’ survival, with a better outcome for epithelioid molecular profile than sarcomatoid (Bueno et al., 2016; Blum et al., 2019). Investigation of the intra-tumor heterogeneity showed that, in reality, tumors from individual patients are composed of a combination of epithelioid-like and sarcomatoid-like components (defined by E/S score) (Blum et al., 2019; Alcala et al., 2019). There was a significant enrichment of BAP1 and SETD2 mutations in tumors of the highest E score, of TERT_prom, NF2, and TP53 alterations in tumors with the highest S score, and LATS2 was more frequently altered in nonepithelioid PM and positively associated with the S score (Quetel et al., 2020; Blum et al., 2019).

A genetic predisposition was suggested in families of mesothelioma cases, and a high incidence of mesothelioma related to BAP1 tumor predisposition syndrome multifunctional gene (BRCA1-associated protein, BAP-1 gene) was discovered (Testa et al., 2011). Other studies reported a significant proportion of frequency of germline mutations and most pathogenic variants in DNA repair and TSGs (Panou et al., 2018; Belcaid et al., 2023).

Additional evidence for the influence of genetic predisposition in mesothelioma pathogenesis is suggested by experimental models, such as the Cross Collaborative MexTAg mouse model, where 72 different genetic backgrounds were tested, resulting in the identification of genetic variants predictive of different disease latency (Fisher et al.).

Knowing that asbestos induces inflammation and chromosome damage, including chromosome missegregation in mesothelial and other mammalian cells, research was performed to detect mutation signatures in PM (Huang et al., 2011). Recently, an analysis of clinical genomic profiling of patients with PM identified near-haploidization in an aggressive biphasic subtype that occurred in younger patients without asbestos exposure (Yang et al., 2024). Losses in chromosomes 14q have been reported in a few studies; for example, recurrent loss in 14q11.2-q21 was found in asbestos-exposed patients compared to patients not exposed, losses in chromosome 14q were similarly identified in fiber-induced murine mesothelioma (Björkqvist et al., 1999; Jean et al., 2011). While DNA oxidation was reported in asbestos-exposed animals, no reactive oxygen species (ROS)-induced mutation signature was reported in human PM. Recently, data on PM were reinvestigated with a new statistical analysis, signature variability analysis (SVA), which considers the heterogeneity of the tumor mutations and the variability of mutations within and across tumors (Morrison et al., 2023). While there was no difference in copy number alteration and single base substitution signatures between exposed and unexposed patients, tumors from patients exposed to asbestos have more within-sample signature diversity and less across-sample heterogeneity than those from unexposed patients, suggesting that SVA could be used to generate a footprint of asbestos exposure. Interestingly, analysis of biopsies taken at distinct anatomical sites revealed intra-tumor heterogeneity (Meiller et al., 2021; Zhang et al., 2021). In this study, NGS performed on key mesothelioma genes showed heterogenous variants, especially NF2, which appears to be a late event.

A recent meta-analysis of DNA methylation in PM investigated 53 studies for DNA methylation of genes in mesothelioma in a total of 97 genes, including microRNAs (miRNA) analyzed at CpG methylation sites (Vandenhoeck et al., 2021). In this study, methylation was more frequent in mesotheliomas of the epithelioid subtype. The number of aberrantly methylated genes was also positively linked to asbestos body counts in the lung, which is a signature of asbestos exposure. The genes most significantly hypermethylated in mesothelioma in comparison with normal tissue are CDH1, ESR1, miR-34b/c, PGR, RARb, SFRP1, and WIF1, and one, APC is hypomethylated.

A promoter hypermethylation of the cell cycle control-associated genes CDKN2A, CDKN2B, RASSF1, CCND2, APC, and HPPBP1 was found in asbestos-exposed patients with a high asbestos body burden after control of confounding factors (Christensen et al., 2008). Asbestos body counts were also positively linked to methylation at CpG sites (Christensen et al., 2009).

Dysregulation of both miRNA and long non-coding RNAs (LncRNAs) has been identified in mesothelioma tumors (Abd-Elmawla et al., 2023; Xu et al., 2023). Many miRNAs target epithelial or mesenchymal markers, and their expression is dependent on the E/S score (Blum et al., 2019). LncRNA, such as NEAT1, PCT6, HOTAIR, and GAS5, were identified as potential biomarkers.

RNA-editing patterns of PM have been studied in humans according to the E/S scores of the tumors in untranslated regions (UTR) of transcripts and in introns. Results showed that PM with a high E-score had RNA frequency editing at the 3′UTR and in introns in PM with a high S score (Felley-Bosco et al., 2023). Then, the regions vary with the EMT, consistent with the epigenetic regulation of EMT. In asbestos-exposed Nf2+/− mice, an RNA-editing signature, mediated by adenosine-deaminase-acting-on dsRNA (ADAR), was found in inflamed tissues 33 weeks after exposure, with a higher number of RNA-editing events in tumors (Rehrauer et al., 2018).

Research on the PM microenvironment aims to identify the different cell components, including immune cells, with the goal of reactivating the immune defense. Single-cell transcriptomics identified a sarcomatoid-enriched phenotype associated with fetal-like endothelial cells, CXCL9/10/11+ macrophages, and cytotoxic, regulatory, and exhausted T lymphocytes (Giotti B, et al., 2024), in line with bulk-RNA studies (Alcala et al., 2019; Mangiante et al., 2023). Detailed analysis of tumor cell populations will permit novel immunotherapy to increase mesothelioma cell susceptibility to death via immune cell activity.

4 Conclusion

Research on PM concerning the role and mechanism of action of asbestos fibers has demonstrated the role of asbestos as a major etiological factor and its multifactorial mechanism of action.

Asbestos specificities are related to the mechanism of fiber–cell interaction (phagocytosis) and genomic damage. One of the key mechanisms of cancer involves gene mutations, which are not at a high level in PM, but chromosomal damage is significant. As PM is an infrequent cancer among the whole population, largely dependent on asbestos exposure in diverse populations, it is likely that DNA repair polymorphism plays a significant role in PM induction. Additionally, a background of persistent inflammation can act at different levels (ROS production, increased proliferation of pre-malignant cells, immunosuppression) to elicit the neoplastic progression and modify the tumor microenvironment. The different tumor microenvironments according to PM histology and the E and S components of the tumor are remarkable, in line with tumoral/EMT evolution, posing a significant challenge for effective therapeutic intervention.

While mesothelioma genomics provided us with footprints on the link between established disease and asbestos, early imprints are poorly identified. Investigations of the early effects of asbestos on mesothelial cells and pleura have shown activation of inflammatory pathways and apoptosis and permitted identifying early genetic and epigenetic impacts at the onset of exposure (Sayan and Mossman, 2015; Huang et al., 2011; Chernova et al., 2017; Rehrauer et al., 2018). Studies of asbestos-exposed mice heterozygous on key TSGs confirmed the importance of these genes in the neoplastic progression of mesothelial cells under asbestos exposure [reviewed in Blanquart et al. (2020) and Testa and Berns (2020)]. Although NF2 is frequently altered, its alteration is a late event (reviewed in Sekido and Sato), and the application of novel technologies will reveal cell environment cues driving that alteration.

Presently, we may propose that PM appears as a double paradigm, toxicologic for approaches to further research on EMPs and genomic for asbestos diseases due to some specific molecular changes. Nevertheless, progress must be made to distinguish the asbestos mechanism of action from the neoplastic progression of mesothelial cells.

Author contributions

M-CJ: writing–original draft. FM: writing–review and editing. EF-B: writing–review and editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abd-Elmawla, M. A., Abdel Mageed, S. S., AL-Noshokaty, T. M., Elballal, M. S., Abulsoud, A. I., Elshaer, S. S., et al. (2023). Melodic maestros: unraveling the role of miRNAs in the diagnosis, progression, and drug resistance of malignant pleural mesothelioma. Pathol. - Res. Pract. 250, 154817. doi:10.1016/j.prp.2023.154817

PubMed Abstract | CrossRef Full Text | Google Scholar

Alcala, N., Mangiante, L., Le-Stang, N., Gustafson, C. E., Boyault, S., Damiola, F., et al. (2019). Redefining malignant pleural mesothelioma types as a continuum uncovers immune-vascular interactions. EBioMedicine 48, 191–202. doi:10.1016/j.ebiom.2019.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Alpert, N., Gerwen, M. V., and Taioli, E. (2020). Epidemiology of mesothelioma in the 21st century in Europe and the United States, 40 years after restricted/banned asbestos use. Lung Cancer Res. 9, S28–S38. doi:10.21037/tlcr.2019.11.11

CrossRef Full Text | Google Scholar

Belcaid, L., Bertelsen, B., Wadt, K., Tuxen, I., Spanggaard, I., Højgaard, M., et al. (2023). New pathogenic germline variants identified in mesothelioma. Lung Cancer 179, 107172. doi:10.1016/j.lungcan.2023.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Björkqvist, A.-M., Wolf, M., Nordling, S., Tammilehto, L., Knuuttila, A., Kere, J., et al. (1999). Deletions at 14q in malignant mesothelioma detected by microsatellite marker analysis. Br. J. Cancer 81, 1111–1115. doi:10.1038/sj.bjc.6690816

PubMed Abstract | CrossRef Full Text | Google Scholar

Blanquart, C., Jaurand, M.-C., and Jean, D. (2020). The biology of malignant mesothelioma and the relevance of preclinical models. Front. Oncol. 10, 388. doi:10.3389/fonc.2020.00388

PubMed Abstract | CrossRef Full Text | Google Scholar

Blum, Y., Meiller, C., Quetel, L., Elarouci, N., Ayadi, M., Tashtanbaeva, D., et al. (2019). Dissecting heterogeneity in malignant pleural mesothelioma through histo-molecular gradients for clinical applications. Nat. Commun. 10, 1333. doi:10.1038/s41467-019-09307-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Bueno, R., Stawiski, E. W., Goldstein, L. D., Durinck, S., De Rienzo, A., Modrusan, Z., et al. (2016). Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nat. Genet. 48, 407–416. doi:10.1038/ng.3520

PubMed Abstract | CrossRef Full Text | Google Scholar

Carlo Rovelli, (2016). Anaximander and the nature of science.

Google Scholar

Chernova, T., Murphy, F. A., Galavotti, S., Sun, X. M., Powley, I. R., Grosso, S., et al. (2017). Long-fiber carbon nanotubes replicate asbestos-induced mesothelioma with disruption of the tumor suppressor gene Cdkn2a (Ink4a/Arf). Curr. Biol. 27, 3302–3314 e6. doi:10.1016/j.cub.2017.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Christensen, B. C., Godleski, J. J., Marsit, C. J., Houseman, E. A., Lopez-Fagundo, C. Y., Longacker, J. L., et al. (2008). Asbestos exposure predicts cell cycle control gene promoter methylation in pleural mesothelioma. Carcinogenesis 29, 1555–1559. doi:10.1093/carcin/bgn059

PubMed Abstract | CrossRef Full Text | Google Scholar

Christensen, B. C., Houseman, E. A., Godleski, J. J., Marsit, C. J., Longacker, J. L., Roelofs, C. R., et al. (2009). Epigenetic profiles distinguish pleural mesothelioma from normal pleura and predict lung asbestos burden and clinical outcome. Cancer Res. 69, 227–234. doi:10.1158/0008-5472.CAN-08-2586

PubMed Abstract | CrossRef Full Text | Google Scholar

Creaney, J., Patch, A.-M., Addala, V., Sneddon, S. A., Nones, K., Dick, I. M., et al. (2022). Comprehensive genomic and tumour immune profiling reveals potential therapeutic targets in malignant pleural mesothelioma. Genome Med. 14, 58. doi:10.1186/s13073-022-01060-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Donaldson, K., Murphy, F. A., Duffin, R., and Poland, C. A. (2010). Asbestos, carbon nanotubes and the pleural mesothelium: a review of the hypothesis regarding the role of long fibre retention in the parietal pleura. Inflamm. mesothelioma. doi:10.1186/1743-8977-7-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Donaldson, K., Poland, C. A., Murphy, F. A., MacFarlane, M., Chernova, T., and Schinwald, A. (2013). Pulmonary toxicity of carbon nanotubes and asbestos — similarities and differences. Adv. Drug Deliv. Rev. 65, 2078–2086. doi:10.1016/j.addr.2013.07.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Febres-Aldana, C. A., Chang, J. C., Jungbluth, A. A., Adusumilli, P. S., Bodd, F. M., Frosina, D., et al. (2024). Comparison of immunohistochemistry, next-generation sequencing and fluorescence in situ hybridization for detection of MTAP loss in pleural mesothelioma. Mod. Pathol. 37, 100420. doi:10.1016/j.modpat.2023.100420

PubMed Abstract | CrossRef Full Text | Google Scholar

Felley-Bosco, E., Qi, W., Jean, D., Meiller, C., and Rehrauer, H. (2023). The pattern of RNA editing changes in pleural mesothelioma upon epithelial-mesenchymal transition. Int. J. Mol. Sci. 24, 2874. doi:10.3390/ijms24032874

PubMed Abstract | CrossRef Full Text | Google Scholar

Frank, A. L. (2020). Global use of asbestos - legitimate and illegitimate issues. J. Occup. Med. Toxicol. 15, 16. doi:10.1186/s12995-020-00267-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Franklin, P., Alfonso, H., Reid, A., Olsen, N., Shilkin, K. B., Brims, F., et al. (2016). Asbestos exposure and histological subtype of malignant mesothelioma. Occup. Environ. Med. oemed-2016-103721 73, 749–752. doi:10.1136/oemed-2016-103721

PubMed Abstract | CrossRef Full Text | Google Scholar

Giotti, B., Dolasia, K., Zhao, W., Cai, P., Sweeney, R., Merritt, E., et al. (2024). Single-cell view of tumor microenvironment gradients in pleural mesothelioma. Cancer Discov. 14, 2262–2278. doi:10.1158/2159-8290.CD-23-0017

PubMed Abstract | CrossRef Full Text | Google Scholar

Gottesfeld, P. (2024). Exposure hazards from continuing use and removal of asbestos cement products. Ann. Work Expo. Health 68, 8–18. doi:10.1093/annweh/wxad066

PubMed Abstract | CrossRef Full Text | Google Scholar

Gualtieri, A. F. (2018). Towards a quantitative model to predict the toxicity/pathogenicity potential of mineral fibers. Toxicol. Appl. Pharmacol. 361, 89–98. doi:10.1016/j.taap.2018.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, S. X. L., Jaurand, M.-C., Kamp, D. W., Whysner, J., and Hei, T. K. (2011). Role of mutagenicity in asbestos fiber-induced carcinogenicity and other diseases. J. Toxicol. Environ. Health Part B 14, 179–245. doi:10.1080/10937404.2011.556051

PubMed Abstract | CrossRef Full Text | Google Scholar

Husain, A. N., Chapel, D. B., Attanoos, R., Beasley, M. B., Brcic, L., Butnor, K., et al. (2024). Guidelines for pathologic diagnosis of mesothelioma: 2023 update of the consensus statement from the international mesothelioma interest group. Arch. Pathol. Lab. Med. 148, 1251–1271. doi:10.5858/arpa.2023-0304-RA

PubMed Abstract | CrossRef Full Text | Google Scholar

Institute of Medicine (US), National Research Council (US), Committee for the Review of the NIOSH Research Roadmap on Asbestos Fibers and Other Elongate Mineral Particles (2009). A review of the NIOSH roadmap for research on asbestos fibers and other elongate mineral particles. Washington, D.C: National Academies Press. doi:10.17226/12697

CrossRef Full Text | Google Scholar

Jean, D., Thomas, E., Manié, E., Renier, A., de Reynies, A., Lecomte, C., et al. (2011). Syntenic relationships between genomic profiles of fiber-induced murine and human malignant mesothelioma. Am. J. Pathol. 178, 881–894. doi:10.1016/j.ajpath.2010.10.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Kane, A. B., Hurt, R. H., and Gao, H. (2018). The asbestos-carbon nanotube analogy: an update. Toxicol. Appl. Pharmacol. 361, 68–80. doi:10.1016/j.taap.2018.06.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuroda, A. (2021). Recent progress and perspectives on the mechanisms underlying Asbestos toxicity. Genes Environ. 43, 46. doi:10.1186/s41021-021-00215-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Mangiante, L., Alcala, N., Sexton-Oates, A., Di Genova, A., Gonzalez-Perez, A., Khandekar, A., et al. (2023). Multiomic analysis of malignant pleural mesothelioma identifies molecular axes and specialized tumor profiles driving intertumor heterogeneity. Nat. Genet. 55, 607–618. doi:10.1038/s41588-023-01321-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Meiller, C., Montagne, F., Hirsch, T. Z., Caruso, S., de Wolf, J., Bayard, Q., et al. (2021). Multi-site tumor sampling highlights molecular intra-tumor heterogeneity in malignant pleural mesothelioma. Genome Med. 13, 113. doi:10.1186/s13073-021-00931-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Metintas, M., Ak, G., and Metintas, S. (2024). Environmental asbestos exposure and lung cancer. Lung Cancer 194, 107850. doi:10.1016/j.lungcan.2024.107850

PubMed Abstract | CrossRef Full Text | Google Scholar

Miserocchi, G., Sancini, G., Mantegazza, F., and Chiappino, G. (2008). Translocation pathways for inhaled asbestos fibers. Env. Health 7, 4. doi:10.1186/1476-069X-7-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Morrison, M. L., Mangé, L., Senkin, S., Rosenberg, N. A., Foll, M., Fernandez-Cuesta, L., et al. (2023). Variability of mutational signatures is a footprint of carcinogens. doi:10.1101/2023.11.23.23298821

CrossRef Full Text | Google Scholar

Murphy, F., Dekkers, S., Braakhuis, H., Ma-Hock, L., Johnston, H., Janer, G., et al. (2021). An integrated approach to testing and assessment of high aspect ratio nanomaterials and its application for grouping based on a common mesothelioma hazard. NanoImpact 22, 100314. doi:10.1016/j.impact.2021.100314

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagai, H., and Toyokuni, S. (2010). Biopersistent fiber-induced inflammation and carcinogenesis: lessons learned from asbestos toward safety of fibrous nanomaterials. Arch. Biochem. Biophys. 502, 1–7. doi:10.1016/j.abb.2010.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagai, H., and Toyokuni, S. (2012). Differences and similarities between carbon nanotubes and asbestos fibers during mesothelial carcinogenesis: shedding light on fiber entry mechanism. Cancer Sci. 103, 1378–1390. doi:10.1111/j.1349-7006.2012.02326.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Nair, N. U., Jiang, Q., Wei, J. S., Misra, V. A., Morrow, B., Kesserwan, C., et al. (2023). Genomic and transcriptomic analyses identify a prognostic gene signature and predict response to therapy in pleural and peritoneal mesothelioma. Cell Rep. Med. 4, 100938. doi:10.1016/j.xcrm.2023.100938

PubMed Abstract | CrossRef Full Text | Google Scholar

Nel, A. (2023). Carbon nanotube pathogenicity conforms to a unified theory for mesothelioma causation by elongate materials and fibers. Environ. Res. 230, 114580. doi:10.1016/j.envres.2022.114580

PubMed Abstract | CrossRef Full Text | Google Scholar

Panou, V., Gadiraju, M., Wolin, A., Weipert, C. M., Skarda, E., Husain, A. N., et al. (2018). Frequency of germline mutations in cancer susceptibility genes in malignant mesothelioma. J. Clin. Oncol. 36, 2863–2871. doi:10.1200/jco.2018.78.5204

PubMed Abstract | CrossRef Full Text | Google Scholar

Quetel, L., Meiller, C., Assié, J. B., Blum, Y., Imbeaud, S., Montagne, F., et al. (2020). Genetic alterations of malignant pleural mesothelioma: association with tumor heterogeneity and overall survival. Mol. Oncol. 14, 1207–1223. doi:10.1002/1878-0261.12651

PubMed Abstract | CrossRef Full Text | Google Scholar

Rehrauer, H., Wu, L., Blum, W., Pecze, L., Henzi, T., Serre-Beinier, V., et al. (2018). How asbestos drives the tissue towards tumors: YAP activation, macrophage and mesothelial precursor recruitment, RNA editing, and somatic mutations. Oncogene 37, 2645–2659. doi:10.1038/s41388-018-0153-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Rose, A. (2022). Asbestos – the last modernist object. Edinburgh: University Press. doi:10.1515/9781474482448

CrossRef Full Text | Google Scholar

Sauter, J. L., Dacic, S., Galateau-Salle, F., Attanoos, R. L., Butnor, K. J., Churg, A., et al. (2022). The 2021 WHO classification of tumors of the pleura: advances since the 2015 classification. J. Thorac. Oncol. 17, 608–622. doi:10.1016/j.jtho.2021.12.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Sayan, M., and Mossman, B. T. (2015). The NLRP3 inflammasome in pathogenic particle and fibre-associated lung inflammation and diseases. Part. Fibre Toxicol. 13, 51. doi:10.1186/s12989-016-0162-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Schinwald, A., Murphy, F. A., Prina-Mello, A., Poland, C. A., Byrne, F., Movia, D., et al. (2012). The threshold length for fiber-induced acute pleural inflammation: shedding light on the early events in asbestos-induced mesothelioma. Toxicol. Sci. 128, 461–470. doi:10.1093/toxsci/kfs171

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, R., and Frank, A. L. (2023). Does the presence of asbestos-containing materials in buildings post-construction and before demolition have an impact on the exposure to occupants in non-occupational settings? Cureus 15, e37305. doi:10.7759/cureus.37305

PubMed Abstract | CrossRef Full Text | Google Scholar

Stanton, M. F., Layard, M., Tegeris, A., Miller, E., May, M., Morgan, E., et al. (1981). Relation of particle dimension to carcinogenicity in amphibole asbestoses and other fibrous minerals. J. Natl. Cancer Inst. 67, 965–975.

PubMed Abstract | Google Scholar

Suarez, J. S., Novelli, F., Goto, K., Ehara, M., Steele, M., Kim, J.-H., et al. (2023). HMGB1 released by mesothelial cells drives the development of asbestos-induced mesothelioma. Proc. Natl. Acad. Sci. 120, e2307999120. doi:10.1073/pnas.2307999120

PubMed Abstract | CrossRef Full Text | Google Scholar

Testa, J. R., and Berns, A. (2020). Preclinical models of malignant mesothelioma. Front. Oncol. 10, 101. doi:10.3389/fonc.2020.00101

PubMed Abstract | CrossRef Full Text | Google Scholar

Testa, J. R., Cheung, M., Pei, J., Below, J. E., Tan, Y., Sementino, E., et al. (2011). Germline BAP1 mutations predispose to malignant mesothelioma. Nat. Genet. 43, 1022–1025. doi:10.1038/ng.912

PubMed Abstract | CrossRef Full Text | Google Scholar

USGS (2022). Mineral commodity summaries. Reston, VA. doi:10.3133/mcs2022

CrossRef Full Text | Google Scholar

Vandenhoeck, J., Van Meerbeeck, J. P., Fransen, E., Raskin, J., Van Camp, G., Op De Beeck, K., et al. (2021). DNA methylation as a diagnostic biomarker for malignant mesothelioma: a systematic review and meta-analysis. J. Thorac. Oncol. 16, 1461–1478. doi:10.1016/j.jtho.2021.05.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Dorn, A. (2017). Libby: the long legacy of a public health disaster. Lancet Respir. Med. 5, 174–175. doi:10.1016/S2213-2600(17)30048-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Vorster, T., Mthombeni, J., teWaterNaude, J., and Phillips, J. I. (2022). The association between the histological subtypes of mesothelioma and asbestos exposure characteristics. Int. J. Environ. Res. Public. Health 19, 14520. doi:10.3390/ijerph192114520

PubMed Abstract | CrossRef Full Text | Google Scholar

Wagner, J. C., Sleggs, C. A., and Marchand, P. (1960). Diffuse pleural mesothelioma and asbestos exposure in the north western cape province. Br. J. Ind. Med. 17, 260–271. doi:10.1136/oem.17.4.260

PubMed Abstract | CrossRef Full Text | Google Scholar

Wylie, A. G., and Korchevskiy, A. A. (2023). Dimensions of elongate mineral particles and cancer: a review. Environ. Res. 230, 114688. doi:10.1016/j.envres.2022.114688

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, X., Li, H., Xie, M., Zhou, Z., Wang, D., and Mao, W. (2023). LncRNAs and related molecular basis in malignant pleural mesothelioma: challenges and potential. Crit. Rev. Oncol. Hematol. 186, 104012. doi:10.1016/j.critrevonc.2023.104012

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, S. R., Jayakumaran, G., Benhamida, J., Febres-Aldana, C. A., Fanaroff, R., Chang, J., et al. (2024). Diffuse pleural mesotheliomas with genomic near-haploidization: a newly recognized subset with distinct clinical, histologic, and molecular features. Clin. Cancer Res. 30, 2780–2789. doi:10.1158/1078-0432.CCR-24-0085

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshida, G. J. (2019). Beyond Stanton and Pott hypothesis; carbon nanotubes-induced malignant mesothelioma as a disease of gene loss. J. Occup. Health 61, 203–205. doi:10.1002/1348-9585.12019

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, M., Luo, J. L., Sun, Q., Harber, J., Dawson, A. G., Nakas, A., et al. (2021). Clonal architecture in mesothelioma is prognostic and shapes the tumour microenvironment. Nat. Commun. 12, 1751. doi:10.1038/s41467-021-21798-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: asbestos, elongated mineral particles, mesothelioma, multi-omics-approaches, social impact, fiber pathogenicity paradigm

Citation: Jaurand M-C, Murphy F and Felley-Bosco E (2025) Editorial: Asbestos and disease genomics: is mesothelioma a genomic paradigm?. Front. Toxicol. 6:1536344. doi: 10.3389/ftox.2024.1536344

Received: 28 November 2024; Accepted: 05 December 2024;
Published: 03 January 2025.

Edited and reviewed by:

François Huaux, Université Catholique de Louvain, Belgium

Copyright © 2025 Jaurand, Murphy and Felley-Bosco. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Marie-Claude Jaurand, bWFyaWUtY2xhdWRlLmphdXJhbmRAaW5zZXJtLmZy

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.