Next Article in Journal
It Takes More than Two to Tango: Complex, Hierarchal, and Membrane-Modulated Interactions in the Regulation of Receptor Tyrosine Kinases
Next Article in Special Issue
Fibrosis in Mesothelioma: Potential Role of Lysyl Oxidases
Previous Article in Journal
The Prognostic Role of Intratumoral Stromal Content in Lobular Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Incidence, Survival Analysis and Future Perspective of Primary Peritoneal Mesothelioma (PPM): A Population-Based Study from SEER Database

1
Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
2
Department of Surgery, San Joaquin General Hospital, French Camp, CA 95231, USA
3
Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
4
Department of Surgery, Brandon Regional Hospital, Brandon, FL 33511, USA
5
Department of Medicine, Bolan Medical College, Quetta 87300, Pakistan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(4), 942; https://doi.org/10.3390/cancers14040942
Submission received: 13 January 2022 / Revised: 3 February 2022 / Accepted: 9 February 2022 / Published: 14 February 2022
(This article belongs to the Special Issue Translational Therapies for Mesothelioma)

Abstract

:

Simple Summary

Mesotheliomas arising from the lining of the mesothelial cells of the peritoneum are rare. However, they are strongly linked to asbestos exposure, similar to the relatively well-known pleural mesotheliomas. It is slightly more common in men than in women, with the majority of cases seen in Caucasians older than 50 years. Distant spread of tumor, size > 4 cm, and negative lymph node status were observed in our study among the patients with evaluable data. Optimal outcomes are achieved when patients are treated in specialized centers with surgical debulking followed by heated chemotherapy administered intraoperatively. Systemic chemotherapy and radiation are options for the selected patient groups. Patients with poorly differentiated large tumors (>4 cm), Caucasian race, and distant spread of disease outside the abdominal cavity have worse prognosis.

Abstract

Background: Primary peritoneal mesothelioma (PPM) is a rare and aggressive tumor arising from the visceral and parietal peritoneum. The diagnosis and treatment of PPM are often delayed because of non-specific clinical presentation, and the prognosis is worse. The current study investigated the demographic, clinical, and pathological factors affecting patient prognosis and survival in PPM. Methods: Demographic and clinical data of 1998 patients with PPM were extracted from the Surveillance Epidemiology and End Results (SEER) database (1975–2016). The chi-square test, paired t-test, and multivariate analysis were used to analyze the data. Results: The majority of PPM patients were male (56.2%, p < 0.005) and Caucasian (90.4%, p < 0.005, with a mean age of diagnosis was 69 ± 13 years. The grading, histological, and tumor size information were classified as “Unknown” in most of the cases, but when available, poorly differentiated tumors (8.7%), malignant mesothelioma, not otherwise specified (63.4%) and tumors > 4 cm in size (8%), respectively, were most common, p < 0.005. Chemotherapy was administered to 50.6% of patients, followed by resection (29.2%) and radiation (1.5%), p < 0.001. The cohort of PPM had a five-year overall survival of 20.3% (±1.1), compared to 43.5% (±5.9), 25.9% (± 8.4), and 18.7% (±1.6) for those with surgery, radiation, or chemotherapy alone, respectively. Poor differentiation (OR = 4.2, CI = 3.3–4.9), tumor size > 4 cm (OR = 3.9, CI = 3.2–4.5), Caucasian race (OR = 2.9, CI = 2.6–4.4), and distant SEER stage (OR = 2.5, CI = 1.1–3.2) were all linked with increased mortality (p < 0.001). Conclusion: An extremely rare and aggressive peritoneal tumor, PPM may be difficult to identify at the time of diagnosis. Radiation therapy likely to have a limited function in the treatment of this condition, with surgery and chemotherapy being the primary choices. All PPM patients should be enrolled in a nationwide registry to improve our understanding of the pathogenesis and identify factors affecting survival.

1. Introduction

Malignant mesothelioma (MM) is an aggressive and lethal disease. It affects pleural and peritoneal membranes and is associated with asbestos exposure [1,2]. PPM comprises 30% of all mesotheliomas, second to pleural mesothelioma [3,4]. Approximately 85% of all mesotheliomas are associated with asbestos exposure in males [5].
Due to nonspecific clinical features and an indolent course, diagnosis is often delayed; if an adequate tumor specimen is not available, such as that obtained by ascitic fluid sampling, it may be mistaken for other benign or malignant abdominal processes [6].
Suspected cases were mostly advanced. PPM presents with ascites, significant weight loss, fatigue, anorexia, palpable abdominal mass, and signs of intestinal obstruction [7]. Likewise, PPM has different variants, and the precise diagnosis of PPM with the subtype can only be made with tissue biopsy using special stains [8]. Although no specific imaging has been beneficial in diagnosing PPM, abdominal imaging, particularly multidetector computed tomography (MDCT), helps in further evaluation and extent of the tumor [9,10].
Additionally, no specific guidelines exist for PPM management; however, cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) are recommended in most eligible patients [11]. The role of systemic chemotherapy is limited to patients who are not good candidates for cytoreductive surgery and HIPEC [11]. Even with optimum treatment, the overall survival of PPM is dismal [11]. So far, most data regarding PPM are based on a few case reports and case series. To better understand this rare entity, we conducted an updated population-based outcome study using data from the SEER database.

2. Materials and Methods

The Surveillance, Epidemiology, and End Result (SEER) database, SEER Stat program version 8.0.4 (https://seer.cancer.gov/seerstat/, accessed on 12 January 2022), was used to retrieve the data for the current study (1975–2016), and individuals with histologically proven PPM were recognized and imported to IBM SPSS®v20.2 (IBM Corp, Armonk, NY, USA) for further analysis. Both anatomical codes (C48.1, C48.2) and histological codes (9050/3, 9051/3, 9052/3, 9053/3, and 9055/3) were used to abstract the data. Significant variables included age, gender, race, tumor stage, and type of treatment received.
Moreover, patients without histological confirmation and those diagnosed with in situ cancer were excluded. The current study looked at overall survival, mortality, and 1-, 2-, and 5-year cancer survival. For categorical variables, the chi-square test was utilized, while for continuous variables, the paired t-test and ANOVA were used. Statistical significance was defined at p < 0.05 for multivariable analysis.

3. Results

Data from 1998 patients were extracted. Patient’s tumor and survival characteristics were observed in the entire cohort.

3.1. Demographical Characteristics of Entire Cohort

With an overall mean age at diagnosis of 69 ± 13 years, PPM patients were more likely to be male (n = 1122; 56.2%), followed by women (n = 876; 43.8%), with a male: female (M: F) ratio of 1:3; p < 0.005. Caucasians were the predominant race affected by PPM (n = 1806; 90.4%), followed by African Americans (n = 101; 5%), and others (n = 85; 4.3%); p < 0.005. Racial data were not available (n = 6; 0.3%); Table 1.

3.2. Tumor Characteristics (Histological and Grading)

Grading information and histological information were not available for most patients (n = 1649; 82.5%). When available, poorly differentiated tumors (Grade 3) (n = 173; 49.6%) were the most common grade, followed by well-differentiated (Grade 1) (n = 106; 5.3%), and moderately differentiated (Grade 2) (n = 37; 1.9%), p < 0.005.
Similarly, the histological type was not specified in most of the cases (n = 1266; 63.4%), but when the information was available, most PPMs were of epithelioid type (n = 616; 30.8%), followed by biphasic PPM (n = 67; 3.3%), fibrous PPM (n = 48; 2.4%), and multicystic PPM (n = 1; 0.05%), p < 0.005; Table 2.

3.3. Tumor Size and Extent Characteristics

Data regarding tumor size were not available for most of the patients (n = 1785; 89.3%). When available, most of PPMs were >4 cm in size (n = 160; 8%), followed by 2–4 cm in size (n = 32; 1.60%), and <2 cm in size (n = 21, 1.05%), p < 0.005 (Table 3)
Similarly, most of the data regarding the extent of the disease were classified as unknown (n = 889; 44.50%), whereas when specific information was provided, most of the PPM presented with distant spread (n = 591; 29.60%), followed by those with regional spread (n = 182, 9.10%), and localized extent (n = 166; 8.30%), p < 0.001. Most patients with PPM had lymph node-negative status (n = 1431; 71.60%), followed an unknown status (n = 445; 22.30%), and lymph node-positive status (n = 122, 6.10%), p < 0.001 (Table 3).

3.4. Treatment Characteristics

In those patients who received any type of treatment, the most frequent regimen used was chemotherapy (n = 1010; 50.6%), followed by surgical resection (n = 584; 29.2%), and radiation therapy (n = 29; 1.5%), and the treatment data of (n = 375; 18.7%) patients were missing; p < 0.001. The chemotherapy group received both HIPEC and systemic chemotherapy regimens (Table 4).

3.5. Survival Characteristics

For the entire cohort, the five-year survival rate was found to be 20.3% (±1.1), whereas those who underwent surgery alone, radiotherapy alone, and chemotherapy alone had five-year survival rates of 43% (±5.9), 25.9% (±8.4), and 18.7% (±1.6), p < 0.037) (Table 5).

3.6. Multivariable Analysis

Multivariable analysis revealed that poor differentiation (OR = 4.2, CI = 3.3–4.9), tumor size > 4 cm (OR = 3.9, CI = 3.2–4.5), Caucasian race (OR = 2.9, CI = 2.6–4.4), and distant SEER stage (OR = 2.5, CI = 1.1–3.2) were all linked with increased mortality (p < 0.001) (Table 6).

4. Discussion

The World Health Organization (WHO) classifies mesothelioma into three histologic subtypes; epithelioid, sarcomatoid, and biphasic. The epithelioid type is further divided into papillary, tubulopapillary, acinar, adenomatoid, and solid types. The epithelioid type resembles normal mesothelial cells with papillary and tubolopapillary architectures with minimal cellular atypia. The sarcomatoid subtype comprises of spindle cells with malignant osteoid and, chondroid elements. The biphasic subtype of mesothelioma contains both epithelioid and sarcomatoid components [10]. Along with tumor node metastasis (TNM), the staging peritoneal cancer index (PCI) is a measure of disease spread. PCI was scored in 13 abdominal regions for tumor size and distribution. PCI for peritoneal spread of the disease is evaluated using either laparotomy or computed tomography. A high PCI score is associated with a worse prognosis [12]. However, the subclassification of epithelioid mesothelioma and data on the peritoneal cancer index are not available in the SEER registry.
Peritoneal mesotheliomas are fatal neoplasms, with a median survival of 6–12 months [13]. Due to variability and vague presentation, a deeper characterization of the disease is indispensable to advance our understanding. In addition to describing epidemiological characteristics, we investigated contributing factors, such as pathological and clinical factors, that impact the prognosis and survival of patients with PPM through this population-based study from 1975 to 2016 using the SEER database.
PPM primarily affects male patients in their seventh decade of life. To better analyze the age-adjusted incidence rate of PPM in both men and women, Moolgavkar et al. conducted a SEER database study from 1973–2005 [14]. They reported an age-adjusted PPM rate in men to be 1.2 per million person-years and 0.8 per million person-years in women [14]. Consistent with these findings, we report that 56.2% of patients diagnosed with PPM were men, with an average age of 69 years at the time of diagnosis. Moreover, similar to our extensive database study, age at diagnosis has been shown to be a predictor of survival in previous studies, with patients older than 65 years having poor median overall survival [15].
Although a few non-asbestos-related mesothelioma cases have been reported, prolonged asbestos exposure has been linked to PPM development [8,16,17]. Asbestos toxicity generates reactive oxygen species via oxidative stress, causing genomic instability and DNA damage [18]. The molecular changes in PPM have not been well established; however, in 40–70% of PPM patients, loss of 9p, including cyclin-dependent kinase inhibitor 2A (CDKN2A), or 22q, including neurofibromatosis type 2 (NF2), has been established [19]. Studies have shown that the epithelioid subtype confers a more favorable overall survival (OS) compared to sarcomatoid and biphasic subtypes [20].

4.1. Molecular Profiling and Future Personalized Approach to Therapy

The molecular pathogenesis of PPM is poorly understood. Inactivation of BAP1 is frequent in PPM, seen in up to 79% of cases [21]. Some evidence shows that BAP1 mutations are associated with improved survival and may serve as predictive biomarkers for immunotherapy [21,22]. Other genetic alterations identified included cyclin-dependent kinase inhibitor 2A (CDKN2A) deletions (29%), neurofibromin 2 (NF2) (35%), and anaplastic lymphoma kinase (ALK) gene rearrangements (13%) [19,21,22]. Interestingly, ALK-rearranged PPM does not feature BAP1 or NF2 alterations and may be a potential target for ALK-targeted tyrosine kinase inhibitors [19].
Moreover, owing to the insidious nature of the disease, it is difficult to reliably suspect PPM clinically at the initial stages [23]. When the disease progresses to an advanced stage, the most common clinical presentation of PPM includes increased abdominal girth, abdominal pain, nausea, weight loss, and bowel obstruction [7]. Similar to clinical presentation, suspecting PPM on abdominal imaging in isolation is exceptionally challenging. Abdominal compute tomography (CT) findings are nonspecific, ranging from peritoneum-based masses to ascites with associated peritoneal thickening and scalloping of adjacent abdominal organs. Unlike pleural mesotheliomas, calcified peritoneal plaques are rare [24]. Nonetheless, abdominal CT has traditionally been used more frequently than any other imaging modality for disease extent determination in patients with PPM [9].
Ultimately, the gold standard method for PPM diagnosis is tissue biopsy using immunohistochemical staining [25]. The initial panel usually contains two mesothelial markers (cytokeratin 5/6, calretinin, Wilms tumor 1 (WT-1), and D2-40) and two epithelial markers (MOC-31 and claudin-4). After confirmation of the mesothelial lineage, BAP1 loss, CDKN2A homozygous deletion, and MTAP loss were the most specific markers for the diagnosis of malignant mesothelioma. 5-hmC loss and increased EZH2 expression are novel markers for the diagnosis of malignant mesothelioma but are not widely used yet [26,27,28].
The type of therapy employed is determined by the patient clinical status and spread of the disease [29]. Cytoreductive surgery with HIPEC remains the gold standard for suitable candidates, and for those who cannot tolerate surgical resection, systemic chemotherapy alone can be considered [11,29,30]. However, the long-term benefits of systemic chemotherapy alone are not well understood. To the best of our knowledge, no uniformly accepted guidelines are available for radical resection in PPM patients; however, surgical resection is considered in most PPM patients, with no extraperitoneal spread [31]. Likewise, adjuvant radiation therapy is unlikely to provide survival benefits in PPM patients [31,32]. PPM develops in the anatomical region amidst several vital organs, and radiation therapy may add to additional organ damage in these patients [33]. Our study showed that patients receiving surgical intervention had a more favorable five-year survival rate than those receiving either chemotherapy or radiation therapy alone; however, no statistical significance was observed after analyzing the data.
Optimal cytoreductive surgery (CRS) followed by HIPEC is the gold standard for fitting patients without extraperitoenal spread. For patients who are not candidates for CRS/HIPEC, debulking surgery is not routinely performed and systemic chemotherapy is preferred. Pemetrexed in combination with cisplatin, carboplatin, or gemcitabine is preferred for systemic use. No targeted agents or immunotherapies have been specifically approved for PPM treatment. Based on preclinical models, pharmacological inhibition of the PI3K-PTEN-AKT-mTOR pathway has been tested in phase I/II trials [34]. Agents targeting novel molecular pathways and targets, such as mesothelin, vascular endothelial growth factor, histone deacetylase, focal adhesion kinase, and anaplastic lymphoma kinase, are being actively explored [34]. Although immune checkpoint inhibitors are approved for pleural mesothelioma, trials with these agents do not include patients with PPM. Many clinicians use checkpoint inhibitors off-label for MPM, especially PD-1/PDL-1 antibodies, based on responses in single-arm phase I/II trials. A randomized phase IIb trial with tremelimumab, a CTLA4 antibody (DETERMINE), in PPM was negative [35]. While whole abdominal radiation is part of the traditional treatment paradigm for diffuse MPM, radiation has been vanishingly rare in modern times due to its increased toxicity and dubious survival benefit, as evident in our study. Owing to the lack of specific genomic data and treatment details, we were unable to discuss the use of novel or experimental agents in our group of patients.

4.2. Limitations

Despite these findings, our study has limitations that are applicable to most database-based studies. Information regarding the timing of chemotherapy relative to surgical resection (adjuvant vs. neoadjuvant), type of chemotherapy (HIPEC vs. systemic chemotherapy), specific agents used for each modality, extent and nature of surgical resection, and radiotherapy dosing schedule were not provided in the SEER database, limiting our interpretation of the results. The outcome data of our study is divided into subgroups of patients who received each modality of treatment, but we do not know whether each treatment modality was used alone or in combination. Some of the critical clinical factors, such as socioeconomic factors, performance status, comorbidities, sub-classification of epithelioid mesothelioma, peritoneal cancer index, mitotic tumor index, and other associated pathologies that might affect the interpretation of the results, are not coded correctly in the SEER database. Finally, the side-effects and complications of surgery, chemotherapy, and radiation therapy were not available in the SEER database. Despite these limitations, our study attempts to adequately describe the clinical and demographic aspects of PPM patients.
Ongoing clinical trials on primary peritoneal mesothelioma (PPM) from National Institute of Health (NIH), United States (Table 7).

5. Conclusions

PPM is an aggressive malignancy of the peritoneal surface, where tumor size, Caucasian race, and advanced SEER stage of the disease correlated with poor survival in our study. Surgical resection and HIPEC offers optimum management, with systemic chemotherapy being an option for nonsurgical candidates, and radiation therapy has a limited role in treating patients with PPM. To the best of our knowledge, the current cohort is the most extensive database study of this rare entity. Although the disease is rare; we were able to use a national registry to obtain a substantial number of cases. With the advent of novel agents, further analysis is required to account for changes in the prognosis of various histopathologic subtypes. We strongly suggest that an international registry enrolling all patients with PPM should be introduced to better understand this rare disease.

Author Contributions

Conceptualization, A.W., J.K. and A.U.; literature search, L.V.Z., A.M., B.T. and N.N.; writing—original draft preparation, A.W., N.R.K. and A.U.; writing—review and editing, M.S.P., A.W., A.U. and N.R.K.; Supervision, N.A.K., S.M., F.D.C., N.M. and J.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

All data is publicly available and no IRB required.

Informed Consent Statement

Patient consent was waived due to this article is from SEER database, which is publicly available deidentified patients data from National Cancer Institute (NCI), USA.

Data Availability Statement

All data is publicly available. The data of this manuscript was presented at Society of American Gastrointestinal and Endoscopic Surgeons (SAGES), Los Vegas, Nevada, 31 August–3 September 2021.

Acknowledgments

We acknowledge Zarmina Bibi and Abdul Tahir for their efforts in organizing tables for this project.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pouliquen, D.L.; Kopecka, J. Malignant Mesothelioma. Cancers 2021, 13, 3447. [Google Scholar] [CrossRef] [PubMed]
  2. Cunha, P.; Luz, Z.; Seves, I.; Sousa, C.; Ribeiro, L.S.; Marques, C.; Oliveira, M. Mesotelioma peritoneal maligno: Dificuldades diagnósticas e terapêuticas. Acta Med. Port. 2002, 15, 383–386. [Google Scholar] [PubMed]
  3. Gonzaga, E.S.R.; Rojas, P.G.; Vanar, V. Malignant Peritoneal Mesothelioma Mimicking Recurrent Diverticulitis. Cureus 2019, 11, e3906. [Google Scholar] [CrossRef] [Green Version]
  4. Mirarabshahii, P.; Pillai, K.; Chua, T.C.; Pourgholami, M.H.; Morris, D.L. Diffuse malignant peritoneal mesothelioma—An update on treatment. Cancer Treat. Rev. 2012, 38, 605–612. [Google Scholar] [CrossRef]
  5. Dipper, A.; Maskell, N.; Bibby, A. Ancillary Diagnostic Investigations in Malignant Pleural Mesothelioma. Cancers 2021, 13, 3291. [Google Scholar] [CrossRef]
  6. Asensio, J.A.; Goldblatt, P.; Thomford, N.R. Primary Malignant Peritoneal Mesothelioma: A report of seven cases and a review of the literature. Arch. Surg. 1990, 125, 1477–1481. [Google Scholar] [CrossRef]
  7. García-Fadrique, A.; Mehta, A.; Mohamed, F.; Dayal, S.; Cecil, T.; Moran, B.J. Clinical presentation, diagnosis, classification and management of peritoneal mesothelioma: A review. J. Gastrointest. Oncol. 2017, 8, 915–924. [Google Scholar] [CrossRef] [Green Version]
  8. Valeria, A. Pathologic diagnosis of malignant mesothelioma: Chronological prospect and advent of recommendations and guidelines. Ann. Dell’istituto Super. Sanità 2015, 51, 52–59. [Google Scholar] [CrossRef]
  9. Krishnamurthy, S.; Balasubramaniam, R. Role of Imaging in Peritoneal Surface Malignancies. Indian J. Surg. Oncol. 2016, 7, 441–452. [Google Scholar] [CrossRef] [Green Version]
  10. Kim, J.; Bhagwandin, S.; Labow, D.M. Malignant peritoneal mesothelioma: A review. Ann. Transl. Med. 2017, 5, 236. [Google Scholar] [CrossRef] [Green Version]
  11. Levý, M.; Boublíková, L.; Büchler, T.; Šimša, J. Treatment of Malignant Peritoneal Mesothelioma. Klin. Onkol. 2019, 32, 333–337. [Google Scholar] [CrossRef] [PubMed]
  12. Salo, S.A.S.; Lantto, E.; Robinson, E.; Myllärniemi, M.; Laaksonen, S.; Salo, J.A.; Rantanen, T.; Ilonen, I. Prognostic role of radiological peritoneal cancer index in malignant peritoneal mesothelioma: National cohort study. Sci. Rep. 2020, 10, 13257. [Google Scholar] [CrossRef] [PubMed]
  13. Le Stang, N.; Bouvier, V.; Glehen, O.; Villeneuve, L.; Galateau-Sallé, F.; Clin, B. Incidence and survival of peritoneal malignant mesothelioma between 1989 and 2015: A population-based study. Cancer Epidemiol. 2019, 60, 106–111. [Google Scholar] [CrossRef] [PubMed]
  14. Moolgavkar, S.H.; Meza, R.; Turim, J. Pleural and peritoneal mesotheliomas in SEER: Age effects and temporal trends, 1973–2005. Cancer Causes Control 2009, 20, 935–944. [Google Scholar] [CrossRef]
  15. Feldman, A.L.; Libutti, S.K.; Pingpank, J.F.; Bartlett, D.L.; Beresnev, T.H.; Mavroukakis, S.M.; Steinberg, S.M.; Liewehr, D.J.; Kleiner, D.E.; Alexander, H.R. Analysis of Factors Associated with Outcome in Patients with Malignant Peritoneal Mesothelioma Undergoing Surgical Debulking and Intraperitoneal Chemotherapy. J. Clin. Oncol. 2003, 21, 4560–4567. [Google Scholar] [CrossRef]
  16. Peterson, J.T., Jr.; Greenberg, S.D.; Buffler, P.A. Non-asbestos-related malignant mesothelioma. A review. Cancer 1984, 54, 951–960. [Google Scholar] [CrossRef]
  17. Musk, A.W.; de Klerk, N.; Reid, A.; Hui, J.; Franklin, P.; Brims, F. Asbestos-related diseases. Int. J. Tuberc. Lung Dis. 2020, 24, 562–567. [Google Scholar] [CrossRef]
  18. Neri, M.; Ugolini, D.; Boccia, S.; Canessa, P.A.; Cesario, A.; Leoncini, G.; Mutti, L.; Bonassi, S. Chemoprevention of asbestos-linked cancers: A systematic review. Anticancer Res. 2012, 32, 1005–1013. [Google Scholar]
  19. Hung, Y.P.; Dong, F.; Watkins, J.C.; Nardi, V.; Bueno, R.; Cin, P.D.; Godleski, J.J.; Crum, C.P.; Chirieac, L.R. Identification of ALK Rearrangements in Malignant Peritoneal Mesothelioma. JAMA Oncol. 2018, 4, 235–238. [Google Scholar] [CrossRef]
  20. Baratti, D.; Kusamura, S.; Martinetti, A.; Seregni, E.; Oliva, D.G.; Laterza, B.; Deraco, M. Circulating CA125 in Patients with Peritoneal Mesothelioma Treated with Cytoreductive Surgery and Intraperitoneal Hyperthermic Perfusion. Ann. Surg. Oncol. 2006, 14, 500–508. [Google Scholar] [CrossRef]
  21. Singhi, A.D.; Krasinskas, A.M.; A Choudry, H.; Bartlett, D.L.; Pingpank, J.F.; Zeh, H.J.; Luvison, A.; Fuhrer, K.; Bahary, N.; Seethala, R.R.; et al. The prognostic significance of BAP1, NF2, and CDKN2A in malignant peritoneal mesothelioma. Mod. Pathol. 2016, 29, 14–24. [Google Scholar] [CrossRef] [PubMed]
  22. Shrestha, R.; Nabavi, N.; Lin, Y.-Y.; Mo, F.; Anderson, S.; Volik, S.; Adomat, H.H.; Lin, D.; Xue, H.; Dong, X.; et al. BAP1 haploinsufficiency predicts a distinct immunogenic class of malignant peritoneal mesothelioma. Genome Med. 2019, 11, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hesdorffer, M.E.; Chabot, J.; DeRosa, C.; Taub, R. Peritoneal Mesothelioma. Curr. Treat. Options Oncol. 2008, 9, 180–190. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, Y.; Gunawardane, D.; Moffat, D.; Carter, C. Metastatic primary peritoneal malignant mesothelioma mimicking primary diffuse gastric carcinoma: A rare encounter in gastric biopsy. Pathology 2017, 49, 317–319. [Google Scholar] [CrossRef]
  25. Stahel, R.A.; Weder, W.; Felley-Bosco, E.; Petrausch, U.; Curioni-Fontecedro, A.; Schmitt-Opitz, I.; Peters, S. Searching for targets for the systemic therapy of mesothelioma. Ann. Oncol. 2015, 26, 1649–1660. [Google Scholar] [CrossRef]
  26. Chapel, D.B.; Schulte, J.J.; Husain, A.N.; Krausz, T. Application of immunohistochemistry in diagnosis and management of malignant mesothelioma. Transl. Lung Cancer Res. 2020, 9 (Suppl. S1), S3–S27. [Google Scholar] [CrossRef]
  27. Joseph, N.M.; Chen, Y.-Y.; Nasr, A.; Yeh, I.; Talevich, E.; Onodera, C.; Bastian, B.; Rabban, J.T.; Garg, K.; Zaloudek, C.; et al. Genomic profiling of malignant peritoneal mesothelioma reveals recurrent alterations in epigenetic regulatory genes BAP1, SETD2, and DDX3X. Mod. Pathol. 2017, 30, 246–254. [Google Scholar] [CrossRef]
  28. Averbach, A.M.; Sugarbaker, P.H. Peritoneal mesothelioma: Treatment approach based on natural history. Cancer Treat. Res. 1996, 81, 193–211. [Google Scholar] [CrossRef]
  29. Yıldırım, Ö.A.; Aksel, B.; Demirci, U. Multimodality Treatment for Peritoneal Mesothelioma: A Single Center Experience. Acta Oncol. Turc. 2020, 53, 511–515. [Google Scholar] [CrossRef]
  30. Salo, S.A.; Ilonen, I.; Laaksonen, S.; Myllärniemi, M.; Salo, J.A.; Rantanen, T. Malignant Peritoneal Mesothelioma: Treatment Options and Survival. Anticancer Res. 2019, 39, 839–845. [Google Scholar] [CrossRef]
  31. Cao, S.; Jingyan, C.; Cao, J.; Shen, J.; Hu, J.; Che, D.; Pan, B.; Zhang, J.; He, X.; Ding, D.; et al. Advances in malignant peritoneal mesothelioma. Int. J. Colorectal Dis. 2015, 30, 1–10. [Google Scholar] [CrossRef] [PubMed]
  32. Gordon, W.; Antman, K.H.; Greenberger, J.S.; Weichselbaum, R.R.; Chaffey, J.T. Radiation therapy in the management of patients with mesothelioma. Int. J. Radiat. Oncol. 1982, 8, 19–25. [Google Scholar] [CrossRef]
  33. Yano, M.; Ikeda, Y.; Kato, T.; Sakaki, M.; Sato, S.; Yabuno, A.; Kozawa, E.; Yasuda, M. A case of peritoneal malignant mesothelioma following radiation therapy for cervical cancer. Mol. Clin. Oncol. 2018, 8, 302–305. [Google Scholar] [CrossRef] [PubMed]
  34. Boussios, S.; Moschetta, M.; Karathanasi, A.; Tsiouris, A.K.; Kanellos, F.S.; Tatsi, K.; Katsanos, K.H.; Christodoulou, D.K. Malignant peritoneal mesothelioma: Clinical aspects, and therapeutic perspectives. Ann. Gastroenterol. 2018, 31, 659–669. [Google Scholar] [CrossRef]
  35. Maio, M.; Scherpereel, A.; Calabrò, L.; Aerts, J.; Perez, S.C.; Bearz, A.; Nackaerts, K.; Fennell, D.A.; Kowalski, D.; Tsao, A.S.; et al. Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): A multicentre, international, randomised, double-blind, placebo-controlled phase 2b trial. Lancet Oncol. 2017, 18, 1261–1273. [Google Scholar] [CrossRef]
Table 1. Demographic Profiles of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
Table 1. Demographic Profiles of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
VariableFrequency (n)Percentage (%)p-Value
Total1998
Age (Y)<5037919<0.005
>50161981
GenderMale112256.2
%
<0.005
Female87643.8
%
RaceCaucasians180690.4<0.005
African Americans1015
Others854.3
Unknown60.3
Abbreviations: n = number; Y = years; % = percentage.
Table 2. Tumor Characteristics of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
Table 2. Tumor Characteristics of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
VariableFrequency (n)Percentage (%)p-Value
Total1998
Grade of DifferentiationWell differentiated: Grade 11065.3<0.005
Moderately differentiated: Grade 2371.9
Poorly differentiated: Grade 31738.7
Undifferentiated: Anaplastic: Grade 4331.7
Unknown164982.4
Histological VariantMesothelioma, malignant NOS126663.4<0.005
Fibrous Mesothelioma, malignant482.4
Epithelioid Mesothelioma, malignant61630.8
Biphasic, Mesothelioma, malignant673.3
Multicystic Mesothelioma Malignant10.05
Abbreviations: n = number; % = percentage; NOS = not otherwise specified.
Table 3. Tumor stage, size, and lymph node characteristics of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
Table 3. Tumor stage, size, and lymph node characteristics of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
VariableFrequency (n)Percentage (%)p-Value
Total1998
StageLocalized1668.30<0.001
Regional1829.10
Distant59129.60
Unstaged1708.50
Unknown88944.50
Tumor SizeUnknown178589.30<0.005
<2 cm211.05
2–4 cm321.60
>4 cm1608
Lymph Nose StatusNodes Negative143171.60<0.001
Nodes positive1226.10
Unknown44522.30
Abbreviations: n = number; cm = centimeters; % = percentage.
Table 4. Treatment of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
Table 4. Treatment of 1998 Patients with Primary Peritoneal Mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
VariableFrequency (n)Percentage (%)p-Value
Chemotherapy only101056.4<0.001
Surgery alone58429.2
No treatment data37518.7
Radiation only291.5
Abbreviations: n= number; % = percentage.
Table 5. Overall and Treatment Associated Survival Analysis of 1998 Patients with Primary Peritoneal mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
Table 5. Overall and Treatment Associated Survival Analysis of 1998 Patients with Primary Peritoneal mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
VariablePercentage (%)
Overall Survival
1 year46.5% ± 1.3
2 year34.2% ± 1.3
3 year26.7% ± 1.2
4 year22.7% ± 1.2
5 year20.3% ± 1.1
Any Surgery
1 year73.5% ± 6.9
2 year62.2% ± 9.7
3 year54.5% ± 9.8
4 year49.7% ± 8.4
5 year43% ± 5.9
Any Chemotherapy
1 year52.8% ± 1.9
2 year38% ± 1.9
3 year27.1% ± 1.8
4 year21.5% ± 1.7
5 year18.7% ± 1.6
Any Radiation
1 year59.3% ± 9.5
2 year44.4% ± 9.6
3 year29.6% ± 8.8
4 year25.9% ± 8.4
5 year25.9% ± 8.4
Abbreviations: % = percentage.
Table 6. Multivariable analysis of factors influencing mortality in patients with Primary Peritoneal mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
Table 6. Multivariable analysis of factors influencing mortality in patients with Primary Peritoneal mesothelioma from the Surveillance, Epidemiology, and End Results (SEER) database, 1975–2016.
VariablesOdds Ratio (95% C.I.)p-Value
Poorly differentiated4.2 (3.3–4.9,)<0.001
Tumor size > 4 cm3.9 (3.2–4.5)
Caucasian race2.9 (2.6–4.4)
Distant SEER stage2.5 (1.1–3.2)
Abbreviations: % = percentage.
Table 7. Selected ongoing treatment trials in primary peritoneal mesothelioma. Source: Clinicaltrials.gov, accessed 1 February 2022.
Table 7. Selected ongoing treatment trials in primary peritoneal mesothelioma. Source: Clinicaltrials.gov, accessed 1 February 2022.
Trial NumberStudy TitleStudy TypeInterventionPrimary OutcomeStatus
NCT03875144 (MESOTIP)Treatment of malignant peritoneal mesotheliomaPhase 2, randomized, open labelPressurized intraperitoneal aerosol chemotherapy of cisplatin + doxorubicin vs. systemic cisplatin + pemetrexedOSRecruiting
NCT05041062A study of immunotherapy drugs nivolumab and ipilimumab in patients with resectable malignant peritoneal mesotheliomaPhase 2, open label, single armIpilumumab and nivolumabMajor pathologic response rateNot recruiting
NCT05001880Chemotherapy with or without immunotherapy for peritoneal mesotheliomaPhase 2 RandomizedAtezolizumab + bevacizumab + carboplatin + pemetrexed followed by CRS and HIPEC or atezolizumab and bevacizumab vs bevacizumab + carboplatin + pemetrexed followed by CRS and HIPEC or atezolizumab with or without bevacizumabResponse rateRecruiting
NCT04462809 (TALAMESO)Efficacy of a maintenance treatment with talazoparib following first line platinum-based chemotherapy in malignant mesotheliomaPhase 2, multiple cohortsTalazoparib maintenance for two years after surgery and chemotherapyNon-progression proportionNot recruiting
NCT04847063Individualized response assessment to heated intraperitoneal chemotherapy for the treatment of peritoneal carcinomatosis from ovarian, colorectal, appendiceal, or peritoneal mesothelioma histologiesPhase 1, open labelHIPEC with intraperitoneal oxaliplatin and IV 5-FU vs. HIPEC with intraperitoneal mitomycin C vs. HIPEC with intraperitoneal cisplatin and doxorubicin, in addition to IV sodium thiosulfate vs HIPEC with intraperitoneal cisplatin and mitomycin C, in addition to IV sodium thiosulfateCorrelation between ex vivo simulated HIPEC and in vivo HIPEC with respect to two measures of treatment: percent necrosis and Ki-67Recruiting
NCT00996385Velcade and eloxatin for patients with malignant pleural or peritoneal mesotheliomaPhase 2, open labelBortezomib + oxaliplatinObjective tumor response rateUnknown
NCT00024271Surgery, chemotherapy, and radiation therapy in treating patients with peritoneal cancerPhase 2Surgery + intraperitoneal chemotherapy with doxorubicin, cisplatin & gemcitabine and intraperitoneal interferron gamma + radiationN/ANot recruiting
NCT02399371Pembrolizumab in treating patients with malignant mesotheliomaPhase 2PembrolizumabAbility of PD-L1 to predict responseNot recruiting
NCT02535312Methoxyamine, cisplatin, and pemetrexed disodium in treating patients with advanced solid tumors or mesothelioma that cannot be removed by surgery or mesothelioma that is refractory to pemetrexed disodium and cisplatin or carboplatinPhase 1/2Methoxyamine + pemetrexed disodium + cisplatin vs. methoxyamine + pemetrexed disodiumDose-limiting toxicity, response rateNot recruiting
NCT03054298CAR T-cells in mesothelin expressing cancersPhase 1Lentiviral transduced human CART-cells against mesothelin with or without lymphodepletionTreatment-related adverse eventsRecruiting
NCT03907852Phase 1/2 trial of gavo-cel (TC-210) in patients with advanced mesothelin-expressing cancerPhase 1/2Lymphodepletion followed by gavo-cel (CAR-T cells against mesothelin)Recommended phase 2 dose and efficacyRecruiting
NCT04000906 (Nab-PIPAC)PIPAC with nab-paclitaxel and cisplatin in peritoneal carcinomatosisPhase 1bPIPAC administration of nab-paclitaxel and cisplatinMaximum tolerated doseRecruiting
Abbreviations; OS, overall survival; CRS, cytoreductive surgery; HIPEC, hyperthermic intraperitoneal chemotherapy; vs., versus; IV, intravenous; 5-FU, 5-fluorouracil; PD-L1, programmed death-ligand 1; N/A, not available; CAR-T, chimeric antigen receptor modified T; gavo-cel, gavocabtagene autoleucel; PIPAC, pressurized intraperitoneal aerosol chemotherapy.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ullah, A.; Waheed, A.; Khan, J.; Mishra, A.; Tareen, B.; Nama, N.; Karki, N.R.; Panezai, M.S.; Zarate, L.V.; White, J.; et al. Incidence, Survival Analysis and Future Perspective of Primary Peritoneal Mesothelioma (PPM): A Population-Based Study from SEER Database. Cancers 2022, 14, 942. https://doi.org/10.3390/cancers14040942

AMA Style

Ullah A, Waheed A, Khan J, Mishra A, Tareen B, Nama N, Karki NR, Panezai MS, Zarate LV, White J, et al. Incidence, Survival Analysis and Future Perspective of Primary Peritoneal Mesothelioma (PPM): A Population-Based Study from SEER Database. Cancers. 2022; 14(4):942. https://doi.org/10.3390/cancers14040942

Chicago/Turabian Style

Ullah, Asad, Abdul Waheed, Jaffar Khan, Ankita Mishra, Bisma Tareen, Noor Nama, Nabin Raj Karki, Muhammad Saleem Panezai, Luis Velasquez Zarate, Joseph White, and et al. 2022. "Incidence, Survival Analysis and Future Perspective of Primary Peritoneal Mesothelioma (PPM): A Population-Based Study from SEER Database" Cancers 14, no. 4: 942. https://doi.org/10.3390/cancers14040942

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

Article Metrics

Back to TopTop