Open access peer-reviewed chapter

Predictive and Prognosis Factors of Clinical Utility in Mesothelioma

Written By

Rodríguez-Cid Jeronimo Rafael and Flores-Mariñelarena Rodrigo Rafael

Submitted: 06 December 2019 Reviewed: 14 February 2020 Published: 03 May 2020

DOI: 10.5772/intechopen.91769

From the Edited Volume

Mesothelioma

Edited by Sonia Maciá

Chapter metrics overview

807 Chapter Downloads

View Full Metrics

Abstract

The constant research in therapeutics for mesothelioma has been improving their tumor response and overall survival, generating the need to propose markers that guide the doctor’s therapeutic approach in a more precise way. Recently, different predictive factors have been proposed, such as mesothelin-related peptides, fibulin-3, and osteopontin associated with an image giving information about the probability of tumor response to a therapeutic agent or a combination of agents. As is well known, the importance of prognostic markers of utility lies in providing prospective information on the evolution of the patient and thus their ability to guide therapeutic decisions. Although the clinical stage and histology are currently the most described prognostic factors, recent studies have shown interest in the expression of estrogen receptor beta and calretinin, among other promising factors. Given the heterogeneity of this broad field of research in mesothelioma, it is necessary to objectively present the prognostic and predictive factors of greater clinical utility.

Keywords

  • prognosis factors
  • predictive factors
  • response to treatment
  • clinical factors
  • histopathology factors
  • biological factors
  • clinical scores

1. Introduction

The prognosis of patients with mesothelioma is unfavorable, with a median survival of approximately 12 months from diagnosis [1, 2, 3, 4, 5]; this makes a clear need to improve the effectiveness of multimodality approaches and to define in a better way the subgroups’ prognosis [6, 7, 8, 9]. One way to achieve this objective is the use of prognostic and predictive factors; a prognostic factor provides prospective information on the evolution of the patient being able to guide therapeutic decisions, while a predictive factor gives us information on the probability of tumor response to a therapeutic agent.

The characteristics that a prognostic factor must meet are: (a) simple prediction method, (b) wide availability, (c) sensitivity, and (d) reproducibility in any clinical situation. The purpose of these markers is to help define the individual prognosis of clinical groups, select patients who may need other treatments, and assign the most effective treatments to improve survival and quality of life.

Although currently the therapeutic decisions are still based on the classic clinical and pathological prognostic factors already known, such as age, functional status, sex, chest pain, weight loss, thrombocytosis, leukocytosis, anemia, and histological type [3, 10], biological and genetic factors may soon be excellent options as prognostic and predictive factors.

Advertisement

2. Clinical factors

Multiple mesothelioma series have validated advanced TNM stage, age ≥ 50 years, male gender, poor performance status, weight loss, platelet counts ≥400,000, white blood cell counts ≥15.5, low hemoglobin level, low albumin levels, and high serum lactate dehydrogenase levels, among others, as poor predictive and prognosis factors [11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21].

TNM stage is one of the most studied prognosis factors describing a poor survival prognosis for those with advanced or metastatic stage, however, in the same stage of the disease, patients’ survival varies widely suggesting that TNM staging is not completely precise to predict a survival outcome [16]. Moreover, with the new changes applied since the release of the eighth edition of the TNM Classification for Lung and Pleural Tumors where all patients N0M0 malignant pleural mesothelioma as stage IA or IB, differing from the seventh edition classification, in which N0 also was listed within the classifications for stages II and III. These changes reclassified as stage I many patients who were formerly considered as stage II or III since some patients at stage IB experienced poorer prognosis than those at stage III [2223]. Identifying prognostic factors based on the new classification should help to identify the patients with a poor prognosis who may benefit from multimodality treatments. Additional to the TNM staging system, the true tumor volume was independently associated with overall survival and response to treatment; however, more studies need to be done to validate this variable [24, 25, 26, 27].

Previous studies have suggested that females with mesothelioma experience longer survival compared to males [6, 28, 29, 30, 31, 32, 33] with possible suggested explications like those they present at earlier stage [34], tumors with more favorable histology [30], different asbestos exposure responsible for a more indolent tumor biology [35], and a protective effect of circulating estrogen interacting with estrogen receptors present in their tumors, [32, 36, 37] however, only more indolent tumor biology associated to higher frequency of germline mutations in DNA repair genes [38, 39, 40, 41] and interaction of estrogens with estrogen receptor beta [36, 37, 42, 43], other theories still controversial [15].

Platelet count is a practical and easy blood test in clinical practice that has been studied for its role as a prognosis factor due to the interaction of platelets with tumor cells contributing to tumor progression, invasion, metastasis, and angiogenesis [44]. This interaction could be explained by five possible pathways: the first one refers to the release of growth factor by the platelets, including transforming growth factor β and fibroblast growth factor enhancing cancer cell proliferation [45]. Second, platelet membranes are rich in many adhesin molecules like selectins, integrins, immunoglobulin superfamily proteins, and leucine-rich glycoproteins stabilizing the cancer cell arrest in the vasculature, increasing potential of metastasis [46]. Third, platelets could mediate the invasive potential of cancer cells by the release of thromboxane A2, 12-hydroxyeicosatetranoic acid, and matrix metalloproteinases [47, 48, 49]. Fourth, platelets release a large number of pro-angiogenic mediators such as vascular endothelial growth factor and basic fibroblast growth factor influencing the tumor angiogenesis and consequently tumor growth [50, 51, 52]. Fifth, some studies have demonstrated that platelets facilitate the immune escape of cancer cells by surrounding tumor cells and protecting them from the cytotoxic effect of natural killer cells [53, 54]. Several studies concluded that thrombocytosis is correlated with worse overall survival in patients with mesothelioma, indicating that pretreatment could be an adequate and useful factor of prognosis [18].

Recently, many people have focused on the role of inflammation in cancer due to its contribution to tumor initiation and malignant progression. More specifically in mesothelioma, inflammation becomes relevant since most patients have a history of asbestos exposure, and this mineral can skewer cells and set off chemical reactions that lead to inflammation, DNA damage, and cell death [20]. Leukocyte blood count reflects a degree of the systemic inflammatory response in tumor patients, being a valuable and simple indicator [55]. Blood neutrophil-to-lymphocyte ratio is a systemic marker for inflammation closely related to the mortality rate and response to the treatment is useful as a predictive and prognostic factor, taking 3 as a dividing point [20, 56, 57, 58, 59, 60]. In the same way, serum c-protein can reflect an inflammatory environment; although its usefulness as a prognostic and predictive factor has been demonstrated in limited studies, more research is needed to validate its utility [61, 62, 63].

Malnutrition has been related to adverse outcomes in overall survival, quality of life, and increased mortality of malignant tumors [64, 65, 66]. Serum albumin level is a simple and objective indicator to evaluate malnutrition. Multiple studies have demonstrated hypoalbuminemia as an adverse independent prognostic factor for mesothelioma [19, 20, 67].

It is well known that cancer cells tend to employ alternate metabolic pathways, generating adenosine triphosphate through anaerobic glycolysis regulated by lactate dehydrogenase [68, 69]. Several studies assessed the value of high pretreatment lactate dehydrogenase levels for the prediction of a worse survival outcome in mesothelioma [10, 61, 62, 70, 71, 72, 73, 74, 75]. The association between high lactate dehydrogenase levels and poor prognosis on malignancies has tried to be explained in multiple ways. The first theory implies that the production of lactate acid could be up-regulated by lactate dehydrogenase, generating an acidic environment activating metalloproteases, macrophage-mediated angiogenesis and protecting mitochondria from oxidative stress, which induces resistance to hypoxia-induced apoptosis of tumor cells [76, 77, 78, 79, 80]. The second theory explains a strong correlation between elevated lactate dehydrogenase levels and an up-regulation of the hypoxia-inducible factor pathway resulting in a host immunological function attenuation, and enhanced tumor angiogenesis, which has an adverse impact on prognosis in malignant tumors [81]. Despite the great evidence of the utility of lactate dehydrogenase as a convenient and cost-effective indicator for predicting overall survival outcome, cut-off values of lactate dehydrogenase reported on the literature are inconsistent, and it is important to standardize the cut-off value in future studies.

Advertisement

3. Histopathology factors

Together with the TNM stage, the histological type is one of the strongest prognostic factors among patients with mesothelioma. However, with the support of immunohistochemistry markers, not only has diagnosis been improved, but also new markers have appeared for a more accurate prediction of response to treatment, overall survival, and developing better therapeutic approaches.

The most significant prognostic factor until now remains histology with a better prognosis for epithelioid type than sarcomatoid or biphasic type mesothelioma [10, 12, 82, 83]. In addition to histologic subtyping (with solid growth pattern being associated with a poor outcome), nuclear atypia, mitotic count, and the presence of necrosis were found to be independent prognostic factors in epithelioid malignant pleural mesothelioma [84, 85, 86].

Ki67 antigen is used for the assessment of growth fraction of cell populations, due to it being exclusively expressed in proliferating cells; cell cycle analysis showed that Ki67 is detectable in G1, G2, S, and mitosis phases but absent in quiescent cells [87, 88]. Despite most studies indicating that high expression of Ki67 leads to a poor prognosis, some malignancies showing high Ki67 levels actually show a better response to treatment, which could be explained by the fact that cells with high proliferation are susceptible to cytotoxic agents [89, 90, 91, 92, 93]. The detection of Ki67 is not a routine procedure for mesothelioma’s diagnosis and treatment; however, a group has suggested to consider it due to its utility as a possible prognostic marker in epithelioid mesothelioma with a better prognosis outcome in those with low expression levels [94, 95, 96, 97, 98].

Calretinin is a calcium-binding protein that has been established as a useful marker in distinguishing mesothelioma from adenocarcinomas with pleural metastases [99]; Additionally, interest in using higher calretinin scores as favorable prognostic factors has been growing, although further investigation is needed [100, 101, 102, 103, 104].

As mentioned above in the section of clinical factors, estrogen receptor beta expressed on mesothelial tumor cells has become a promising prognostic factor and a possible future therapeutic target [36, 37, 42, 43].

Advertisement

4. Biological factors

Several biomarkers are selectively elevated in patients with mesothelioma. However, further study and validation are required before they are recommended as routine predictive or prognosis factors and they should be adjunct to a radiological assessment. With considerable variation in response to treatment, the emergence of promising biomarkers that could select responders from non-responders at baseline or during treatment would guide to a better therapeutic approach, prevent patients from getting ineffective treatments, and improve cost-effectiveness.

The most researched biomarker until now is the mesothelin; soluble mesothelin is a circulating form of a membrane-bound glycoprotein highly expressed by mesothelial cells in mesothelioma (predominantly epithelioid type) and other malignancies [105]. Despite the controversial evidence reported in the literature [106, 107, 108, 109, 110, 111, 112, 113, 114], a meta-analysis conducted by Tian et al. [115] concluded that a high soluble mesothelin level may lead to a poor prognosis for patients with mesothelioma, it being appropriate to consider mesothelin level as an independent prognostic marker.

Human fibulin-3 is a secreted glycoprotein that plays an essential role in the regulation of cell proliferation and migration [116, 117]. Recent findings have documented altered levels on patients with mesothelioma, highlighting them as a novel biomarker for this malignancy; however, as most studies have been done with limited sample size [114, 118, 119, 120], and the results may not completely mirror the actual value of fibulin-3 for prognosis, further studies are needed for a more comprehensive prognostic role of human fibulin-3 in mesothelioma.

Osteopontin is a glycoprotein that mediates cell-matrix interactions with adverse outcomes for mesothelioma [98, 121, 122]; however, its utility is limited because of the significant variability in the cut-offs used between studies. In order to be validated in the future, a consensus approach is required for sampling and analysis [122].

CA 125 is a transmembrane glycoprotein that can be detected in the fallopian tube, endometrium, endocervix, and mesothelial surface of the peritoneum, pleura, and pericardium [98]. Some cases with non-gynecological cancer showed positive immunohistochemical staining for CA125 in tumor tissue and elevated CA 125 levels in serum [123, 124, 125]. The baseline levels of serum CA125 accompanied by the stage of the disease could be used as independent prognostic factors for patients with mesothelioma; the change in serum CA125 levels can predict overall survival and response to systemic treatments [126, 127, 128].

Advertisement

5. Clinical scores

The best-known clinical prognostic scoring systems for mesothelioma until now derive from the Cancer and Leukemia Group B (CALGB) and the European Organization for Research and Treatment of Cancer (EORTC), both scores have been widely used to better select patients who have a favorable prognosis and could tolerate and potentially benefit from a more aggressive combined modality treatment [3, 10].

The CALGB index was validated by examining the survival of a wide cohort dividing patients into six patient subgroups with different survival rates. The CALGB study considered extent pleural disease, lactate dehydrogenase >500 UI/L, poor performance status, platelets >400,000, non-epithelial histology, and >75 years as negative prognostic factors for survival. The most favorable characteristics were a performance status of 0, age < 49, and hemoglobin ≥14.6/μl [10].

The EORTC score has been validated in 523 patients included in 10 mesothelioma trials with the analysis suggesting that performance status >0, stage IV disease, and biphasic or sarcomatous histologies are associated with a worse outcome [129]. Additional reports confirmed that male sex, older age, and abnormal hematological values also give a poor prognosis [13, 130].

Despite both studies identifying performance status and histology as two main prognostic factors, these analyses included patients with heterogeneous tumor stages at diagnosis, the majority of whom underwent major surgery and whose treatment predated the use of pemetrexed as first-line treatment. Since the positioning of pemetrexed as a first-line treatment, no validated prognostic score has appeared, resulting in the need to generate new studies with the aforementioned scores [131].

Advertisement

6. Promising factors

Although there are multiple prognostic and predictive factors that are currently validated, many others have generated great interest for their potential as a therapeutic target in the future.

There is an increasing interest in the use of semi-quantitative 18F-FDG PET/CT parameters, like metabolic tumor volume and total lesion glycolysis to measure the metabolic activity in the entire tumor volume with great potential to predict response to treatment [119, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144]; however further investigation is needed in mesothelioma patients.

Despite the wide utility of the tissue biopsy, the invasive nature limits their application, especially when repeated biopsies are needed. Given the aforementioned, liquid biopsy has gained interest from oncologists and basic researchers [145]. Although liquid biopsy is still far from replacing tissue biopsy for mesothelioma, plasma and serum samples represent minimally invasive, low-risk, and easily obtained biological fluids that many studies have indicated as potentially interesting prognosis biomarkers as mentioned in the section “Biological factors” [146].

Nowadays, immunotherapy is gaining great relevance in cancer therapeutics. Soon, oncologists will routinely ask for programmed death-ligand 1 (PD-L1) status that has been correlated with better treatment response to anti-PD-L1 antibodies and overall survival outcomes [147, 148, 149, 150, 151]. However, different PD-L1 antibodies coupled with specific staining platforms and scoring criteria may be necessary since finding a suitable cut-off point remains a current challenge [151, 152].

A wide number of molecular prognostic markers for mesothelioma have been investigated. The number of tumor-infiltrating myeloid cells, c-MET expression, thymidylate synthase expression, among others, represent promising biomarkers associated with strong prognostic significance. c-MET is a tyrosine kinase receptor, its overexpression was associated with longer overall survival in patients with mesothelioma [98, 153]. Thymidylate synthase expression may predict pemetrexed efficacy, a certain correlation has also been found with overall survival and progression-free survival [154].

Dysregulated genes play a critical role in the development and progression of mesothelioma, making them future diagnosis and prognosis biomarkers [155]. Recently, Zhou et al. obtained an RNA-Seq count quantified by RSEM for RNA expression profiles of a large cohort of patients with mesothelioma according to The Cancer Genome Atlas guidelines. After a time-dependent receiver operated a characteristic curve to evaluate the prognostic performance of survival prediction, three genes (LSM6, GZMB, and HJURP) were found with a strong statistically significant prognostic association; this prognostic signature could be a clinically useful tool that in the future could be incorporated into a clinical sequencing program to individualize therapy [156].

Advertisement

7. Conclusion

Despite the wide variety of predictive and prognostic factors that exist, just a few are replicable worldwide. Furthermore, only pathological type and performance status are the grade-A recommendations of prognostic factors in pretreatment assessment, as well as the nodal stage, residual disease, and histology during treatment [16].

Although there is currently no validated prognostic approach, according to individual evidence, availability, and cost-benefit, it is recommended to pay special attention to the TNM classification, histological type, and serum CA125 in the decision for multimodal therapy. Despite the practicality of the prognostic scoring systems, further investigations are needed to validate the known scores or generated new ones that fit the new existing therapeutic modalities for mesothelioma.

In the near future, many other prognostic and predictive factors may be introduced in clinical practice making a selection of mesothelioma subgroups to improve the benefit achievable by currently available treatment strategies, and relentless efforts will have to be focused on designing innovative compounds selectively targeting the existing (or additional) markers to improve the grim prognosis of the disease.

Advertisement

Conflict of interest

Dr. Jeronimo Rafael Rodríguez-Cid has educational, investigational and advice relations with MSD, Bristol Myers, Roche, Takeda, Amgen, Abvie, Aztra Zeneca, Boehringer Ingelheim, Pfizer, Celgen, Novartis, and Bayer.

Dr. Rodrigo Rafael Flores-Mariñelarena have no conflicts of interest to declare.

Advertisement

Notes/Thanks/Other declarations

None to declare.

References

  1. 1. Ruffe PA. Pleural mesothelioma. Current Opinion in Oncology. 1991;3(2):328-334
  2. 2. de Pangher MV, Brollo A, Franceschi S, de Matthaeis M, Talamini R, Bianchi C. Prognostic factors of malignant mesothelioma of the pleura. Cancer. 1993;72(2):410-417
  3. 3. Curran D, Sahmoud T, Therasse P, et al. Prognostic factors in patients with pleural mesothelioma: The European Organization for Research and Treatment of Cancer experience. Journal of Clinical Oncology. 1998;16:145-152
  4. 4. Zellos L, Christiani DC. Epidemiology, biologic behavior, and natural history of mesothelioma. Thoracic Surgery Clinics. 2004;14:469-477
  5. 5. van Meerbeeck JP, Damhuis R. Facts, rumours and speculations about the mesothelioma epidemic. Respirology. 2011;16(7):1018-1019
  6. 6. Flores RM, Zakowski M, Venkatraman E, Krug L, Rosenzweig K, Dycoco J, et al. Prognostic factors in the treatment of malignant pleural mesothelioma at a large tertiary referral center. Journal of Thoracic Oncology. 2007;2(10):957-965
  7. 7. Sugarbaker DJ, Flores RM, Jaklitsch MT, et al. Resection margins, extrapleural nodal status, and cell type determine postoperative long-term survival in trimodality therapy of malignant pleural mesothelioma: Results in 183 patients. The Journal of Thoracic and Cardiovascular Surgery. 1999;117(1):54-63
  8. 8. Sterman DH, Treat J, Litzky LA, et al. Adenovirus-mediated herpes simplex virus with thymidine kinase/ ganciclovir gene therapy in patients with localized malignancy: Results of a phase I clinical trial in malignant mesothelioma. Human Gene Therapy. 1998;9(7):1083-1092
  9. 9. Takita H, Dougherthy TJ. Intracavitary photodynamic therapy for malignant pleural mesothelioma. Seminars in Surgical Oncology. 1995;11(5):368-371
  10. 10. Hendon JE, Green MR, Chahinian AP, et al. Factors predictive of survival among 337 patients with mesothelioma treated between 1984 and 1994 by the cancer and leukemia group B. Chest. 1998;113:723-731
  11. 11. Pass HI. Biomarkers and prognostic factors for mesothelioma. Annals of Cardiothoracic Surgery. 2012;1(4):449-456
  12. 12. Pass HI, Giroux D, Kennedy C, et al. IASLC staging committee and participating institutions. Supplementary prognostic variables for pleural mesothelioma: A report from the IASLC staging committee. Journal of Thoracic Oncology. 2014;9:856-864
  13. 13. Edwards JG, Abrams KR, Leverment JN, Spyt TJ, Waller DA, O’Byrne KJ. Prognostic factors for malignant mesothelioma in 142 patients: Validation of CALGB and EORTC prognostic scoring systems. Thorax. 2000;55(9):731-735
  14. 14. Steele JP, Rudd RM. Malignant mesothelioma: Predictors of prognosis and clinical trials. Thorax. 2000;55(9):725-726
  15. 15. Van Gerwen M, Alpert N, Wolf A, et al. Prognostic factors of survival in patients with malignant pleural mesothelioma: An analysis of the National Cancer Database. Carcinogenesis. 2019;40(4):529-536
  16. 16. van Zandwijk N, Clarke C, Henderson D, et al. Guidelines for the diagnosis and treatment of malignant pleural mesothelioma. Journal of Thoracic Disease. 2013;5(6):E254-E307
  17. 17. Nowak AK, Francis RJ, Phillips MJ, et al. A novel prognostic model for malignant mesothelioma incorporating quantitative FDG-PET imaging with clinical parameters. Clinical Cancer Research. 2010;16(8):2409-2417
  18. 18. Zhuo Y, Lin L, Zhang M. Pretreatment thrombocytosis as a significant prognostic factor in malignant mesothelioma: A meta-analysis. Platelets. 2016;28(6):560-566
  19. 19. Yao ZH, Tian GY, Yang SX, et al. Serum albumin as a significant prognostic factor in patients with malignant pleural mesothelioma. Tumour Biology. 2014;35:6839-6845
  20. 20. Yin W, Zheng G, Yang K, Song H, Liang Y. Analysis of prognostic factors of patients with malignant peritoneal mesothelioma. World Journal of Surgical Oncology. 2018;16(1)
  21. 21. Zhuo Y, Lin L, Wei S, Zhang M. Pretreatment elevated serum lactate dehydrogenase as a significant prognostic factor in malignant mesothelioma. Medicine. 2016;95(52):e5706
  22. 22. Abdel-Rahman O. Challenging a dogma; AJCC 8th staging system is not sufficient to predict outcomes of patients with malignant pleural mesothelioma. Lung Cancer. 2017;113:128-133
  23. 23. Berzenji L, Van Schil P, Carp L. The eighth TNM classification for malignant pleural mesothelioma. Translational Lung Cancer Research. 2018;7(5):543-549
  24. 24. Kawashima A, Libshitz HI. Malignant pleural mesothelioma: Manifestations in 50 cases. American Journal of Roentgenology. 1990;155:965-969
  25. 25. Heelan RT, Rusch VW, Begg CB, et al. Staging of malignant pleural mesothelioma: Comparison of CT and MR imaging. American Journal of Roentgenology. 1999;172:1039-1047
  26. 26. Armato SG, Ogarek JL, Starkey A, et al. Variability in mesothelioma tumor response classification. American Journal of Roentgenology. 2006;186:1000-1006
  27. 27. Gill RR, Richards WG, Yeap BY, et al. Epithelial malignant pleural mesothelioma after extrapleural pneumonectomy: Stratification of survival with CT-derived tumor volume. American Journal of Roentgenology. 2012;198(2):359-363
  28. 28. Flores RM, Riedel E, Donington JS, et al. Frequency of use and predictors of cancer-directed surgery in the management of malignant pleural mesothelioma in a community-based (surveillance, epidemiology, and end results [SEER]) population. Journal of Thoracic Oncology. 2010;5:1649-1654
  29. 29. Spirtas R, Conelly RR, Tucker MA. Survival patterns for malignant mesothelioma: The SEER experience. International Journal of Cancer. 1988;41:525-530
  30. 30. Milano MT, Zhang H. Malignant pleural mesothelioma: A population-based study of survival. Journal of Thoracic Oncology. 2010;5:1841-1848
  31. 31. Taioli E, Wolf AS, Camacho-Rivera M, Flores RM. Women with malignant pleural mesothelioma have a threefold better survival rate than men. The Annals of Thoracic Surgery. 2014;98:1020-1024
  32. 32. Wolf AS, Richards WG, Tilleman TR, et al. Characteristics of malignant pleural mesothelioma in women. The Annals of Thoracic Surgery. 2010;90:949-956
  33. 33. Rusch VW, Venkatraman ES. Important prognostic factors in patients with malignant pleural mesothelioma, managed surgically. The Annals of Thoracic Surgery. 1999;68:1799-1804
  34. 34. Yan TD, Popa E, Brun EA, Cerruto CA, Sugarbaker PH. Sex difference in diffuse malignant peritoneal mesothelioma. The British Journal of Surgery. 2006;93:1536-1542
  35. 35. Hillerdal G. Mesothelioma: Cases associated with non-occupational and low dose exposures. Occupational and Environmental Medicine. 1999;56:505-513
  36. 36. Pinton G, Brunelli E, Murer B, et al. Estrogen receptor-beta affects the prognosis of human malignant mesothelioma. Cancer Research. 2009;69:4598-4604
  37. 37. Rodríguez-Cid J, García-Acevedo O, Benjamin-Contreras J, et al. Expression of estrogen receptor beta (ERβ) and its prognostic value in pleural mesothelioma. Journal of Thoracic Disease. 2019;11(4):1456-1464
  38. 38. Hassan R, Morrow B, Walsh T, et al. Inherited predisposition to malignant mesothelioma (MM) due to mutations in DNA repair genes. Journal of Clinical Oncology. 2018;15:8504-8504
  39. 39. Panou V, Gadiraju M, Wolin A, et al. Frequency of germline mutations in cancer susceptibility genes in malignant mesothelioma. Journal of Clinical Oncology. 2018;36:2863-2871
  40. 40. Pastorino S, Yoshikawa Y, Pass H, et al. A subset of mesotheliomas with improved survival occurring in carriers of BAP1 and other germline mutations. Journal of Clinical Oncology. 2018;36(35):3485-3494
  41. 41. De Rienzo A, Archer MA, Yeap BY, et al. Gender-specific molecular and clinical features underlie malignant pleural mesothelioma. Cancer Research. 2016;76:319-328
  42. 42. Pinton G, Moro L. Expression and therapeutic significance of estrogen receptor beta in malignant pleural mesothelioma. Future Science OA. 2017;3:Fso175
  43. 43. Pinton G, Thomas W, Bellini P, et al. Estrogen receptor beta exerts tumor repressive functions in human malignant pleural mesothelioma via EGFR inactivation and affects response to gefitinib. PLoS One. 2010;5:e14110
  44. 44. Jain S, Harris J, Ware J. Platelets: Linking hemostasis and cancer. Arteriosclerosis, Thrombosis, and Vascular Biology. 2010;30(12):2362-2367
  45. 45. Honn KV, Tang DG, Crissman JD. Platelets and cancer metastasis: A causal relationship? Cancer and Metastasis Reviews. 1992;11(3-4):325-351
  46. 46. Li N. Platelets in cancer metastasis: To help the “villain” to do evil. International Journal of Cancer. 2016;138(9):2078-2087
  47. 47. Mezouar S, Mege D, Darbousset R, Farge D, Debourdeau P, Dignat-George F, et al. Involvement of platelet-derived microparticles in tumor progression and thrombosis. Seminars in Oncology. 2014;41(3):346-358
  48. 48. Rolli M, Fransvea E, Pilch J, Saven A, Felding-Habermann B. Activated integrin alphavbeta3 cooperates with metalloproteinase MMP-9 in regulating migration of metastatic breast cancer cells. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(16):9482-9487
  49. 49. Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Reviews. 2006;25(1):9-34
  50. 50. Sabrkhany S, Griffioen AW, Oude Egbrink MG. The role of blood platelets in tumor angiogenesis. Biochimica et Biophysica Acta. 2011;1815(2):189-196
  51. 51. Kisucka J, Butterfield CE, Duda DG, Eichenberger SC, Saffaripour S, Ware J, et al. Platelets and platelet adhesion support angiogenesis while preventing excessive hemorrhage. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(4):855-860
  52. 52. Massberg S, Konrad I, Schurzinger K, Lorenz M, Schneider S, Zohlnhoefer D, et al. Platelets secrete stromal cell-derived factor 1alpha and recruit bone marrow-derived progenitor cells to arterial thrombi in vivo. The Journal of Experimental Medicine. 2006;203(5):1221-1233
  53. 53. Borsig L, Wong R, Feramisco J, Nadeau DR, Varki NM, Varki A. Heparin and cancer revisited: Mechanistic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(6):3352-3357
  54. 54. Nieswandt B, Hafner M, Echtenacher B, Mannel DN. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Research. 1999;59(6):1295-1300
  55. 55. Proctor MJ, Morrison DS, Talwar D, et al. A comparison of inflammation-based prognostic scores in patients with cancer. A Glasgow inflammation outcome study. European Journal of Cancer. 2011;47:2633-2641
  56. 56. Urrejola GI, Bambs CE, Espinoza MA, et al. An elevated neutrophil/lymphocyte ratio is associated with poor prognosis in stage II resected colon cancer. Revista Médica de Chile. 2013;141:602-608
  57. 57. Ozdemir Y, Akin ML, Sucullu I, Balta AZ, Yucel E. Pretreatment neutrophil/ lymphocyte ratio as a prognostic aid in colorectal cancer. Asian Pacific Journal of Cancer Prevention. 2014;15:2647-2650
  58. 58. Azab B, Shah N, Radbel J, et al. Pretreatment neutrophil/lymphocyte ratio is superior to platelet/lymphocyte ratio as a predictor of long-term mortality in breast cancer patients. Medical Oncology. 2013;30:432-442
  59. 59. Szkandera J, Absenger G, Liegl-Atzwanger B, et al. Elevated preoperative neutrophil/lymphocyte ratio is associated with poor prognosis in soft-tissue sarcoma patients. British Journal of Cancer. 2013;108:1677-1683
  60. 60. Gondo T, Nakashima J, Ohno Y, et al. Prognostic value of neutrophil-to-lymphocyte ratio and establishment of novel preoperative risk stratification model in bladder cancer patients treated with radical cystectomy. Urology. 2012;79:1085-1091
  61. 61. Tanrikulu AC, Abakay A, Kaplan MA, et al. A clinical, radiographic and laboratory evaluation of prognostic factors in 363 patients with malignant pleural mesothelioma. Respiration. 2010;80:480-487
  62. 62. Nojiri S, Gemba K, Aoe K, et al. Survival and prognostic factors in malignant pleural mesothelioma: A retrospective study of 314 patients in the west part of Japan. Japanese Journal of Clinical Oncology. 2011;41:32-39
  63. 63. Ghanim B, Hoda MA, Winter MP, et al. Pretreatment serum C-reactive protein levels predict benefit from multimodality treatment including radical surgery in malignant pleural mesothelioma: A retrospective multicenter analysis. Annals of Surgery. 2012;256:357-362
  64. 64. Morgan TM, Tang D, Stratton KL, et al. Preoperative nutritional status is an important predictor of survival in patients undergoing surgery for renal cell carcinoma. European Urology. 2011;59:923-928
  65. 65. Nozoe T, Kohno M, Iguchi T, et al. The prognostic nutritional index can be a prognostic indicator in colorectal carcinoma. Surgery Today. 2012;42:532-535
  66. 66. Nozoe T, Kimura Y, Ishida M, Saeki H, Korenaga D, Sugimachi K. Correlation of pre-operative nutritional condition with post-operative complications in surgical treatment for oesophageal carcinoma. European Journal of Surgical Oncology. 2002;28:396-400
  67. 67. Murphy S, Probert G, Anderson J, et al. Malignant mesothelioma, hypoalbuminaemia and the effect of carboplatin/pemetrexed on survival. Clinical Oncology. 2013;25(12):713-718
  68. 68. Hsu PP, Sabatini DM. Cancer cell metabolism: Warburg and beyond. Cell. 2008;134:703-707
  69. 69. Serganova I, Rizwan A, Ni X, et al. Metabolic imaging: A link between lactate dehydrogenase A, lactate, and tumor phenotype. Clinical Cancer Research. 2011;17:6250-6261
  70. 70. Metintas M, Metintas S, Ucgun I, et al. Prognostic factors in diffuse malignant pleural mesothelioma: Effects of pretreatment clinical and laboratory characteristics. Respiratory Medicine. 2001;95:829-835
  71. 71. Ak G, Metintas S, Metintas M, et al. Prognostic factors according to the treatment schedule in malignant pleural mesothelioma. Journal of Thoracic Oncology. 2009;4:1425-1430
  72. 72. Suzuki H, Hirashima T, Kobayashi M, et al. Prognostic factors in malignant pleural mesothelioma: A retrospective study. Internal Medicine. 2012;51:707-710
  73. 73. Suzuki H, Asami K, Hirashima T, et al. Stratification of malignant pleural mesothelioma prognosis using recursive partitioning analysis. Lung. 2014;192:191-195
  74. 74. Abakay O, Tanrikulu AC, Palanci Y, et al. The value of inflammatory parameters in the prognosis of malignant mesothelioma. The Journal of International Medical Research. 2014;42:554-565
  75. 75. Kataoka Y, Yamamoto Y, Otsuki T, et al. A new prognostic index for overall survival in malignant pleural mesothelioma: The rPHS (regimen, PS, histology or stage) index. Japanese Journal of Clinical Oncology. 2015;45:562-568
  76. 76. Vaupel P, Kallinowski F, Okunieff P. Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: A review. Cancer Research. 1989;49:6449-6465
  77. 77. Stubbs M, McSheehy PM, Griffiths JR, et al. Causes and consequences of tumour acidity and implications for treatment. Molecular Medicine Today. 2000;6:15-19
  78. 78. Bonuccelli G, Tsirigos A, Whitaker-Menezes D, et al. Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle. 2010;9:3506-3514
  79. 79. Martinez-Outschoorn UE, Prisco M, Ertel A, et al. Ketones and lactate increase cancer cell “stemness,” driving recurrence, metastasis and poor clinical outcome in breast cancer: Achieving personalized medicine via Metabolo-Genomics. Cell Cycle. 2011;10:1271-1286
  80. 80. Nemoto S, Takeda K, Yu ZX, et al. Role for mitochondrial oxidants as regulators of cellular metabolism. Molecular and Cellular Biology. 2000;20:7311-7318
  81. 81. Kolev Y, Uetake H, Takagi Y, et al. Lactate dehydrogenase-5 (LDH-5) expression in human gastric cancer: Association with hypoxia-inducible factor (HIF-1alpha) pathway, angiogenic factors production and poor prognosis. Annals of Surgical Oncology. 2008;15:2336-2344
  82. 82. Rusch VW, Giroux D, Kennedy C, et al. Initial analysis of the International Association for the Study of Lung Cancer mesothelioma database. Journal of Thoracic Oncology. 2012;7:1631-1639
  83. 83. Davidson B. Prognostic factors in malignant pleural mesothelioma. Human Pathology. 2015;46:789-804
  84. 84. Kadota K, Suzuki K, Colovos C, et al. A nuclear grading system is a strong predictor of survival in epitheloid diffuse malignant pleural mesothelioma. Modern Pathology. 2012;25:260-271
  85. 85. Galateau-Salle F, Churg A, Roggli V. Tumours of the pleura. Mesothelial tumours. Diffuse malignant mesothelioma. Epithelioid Mesothelioma. In: Travis WD, Brambilla E, Burke AP, Marx A, Nicholson AG, editors. The World Health Organisation Classification of Tumours of the Lung, Pleura, Thymus and Heart. 7th ed. Lyon, France: International Agency of Research on Cancer; 2015. pp. 156-164
  86. 86. Habougit C, Thrombert-Paviot B, Karpathiou G, et al. Histopathologic features predict survival in diffuse pleural malignant mesothelioma on pleural biopsies. Virchows Archiv. 2017;470:639-646
  87. 87. Gerdes J, Lemke H, Baisch H, et al. Cell cycle analysis of a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody Ki-67. Journal of Immunology. 1984;133:1710-1715
  88. 88. Verheijen R, Kuijpers HJ, Schlingemann RO, et al. Ki-67 detects a nuclear matrix-associated proliferation-related antigen. I. Intracellular localization during interphase. Journal of Cell Science. 1989;92(pt 1):123-130
  89. 89. Dowsett M, Smith IE, Ebbs SR, et al. Prognostic value of Ki67 expression after short-term presurgical endocrine therapy for primary breast cancer. Journal of the National Cancer Institute. 2007;99:167-170
  90. 90. Baum M, Budzar AU, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: First results of the ATAC randomised trial. Lancet. 2002;359:2131-2139
  91. 91. Penault-Llorca F, Andre F, Sagan C, et al. Ki67 expression and docetaxel efficacy in patients with estrogen receptor-positive breast cancer. Journal of Clinical Oncology. 2009;27:2809-2815
  92. 92. Orth JD, Tang Y, Shi J, et al. Quantitative live imaging of cancer and normal cells treated with Kinesin-5 inhibitors indicates significant differences in phenotypic responses and cell fate. Molecular Cancer Therapeutics. 2008;7:3480-3489
  93. 93. Mitchison TJ. The proliferation rate paradox in antimitotic chemotherapy. Molecular Biology of the Cell. 2012;23:1-6
  94. 94. Hirano H, Fujisawa T, Maekawa K, et al. Malignant mesothelioma of the peritoneum: Case reports and immunohistochemical findings including Ki-67 expression. Medical Molecular Morphology. 2010;43:53-59
  95. 95. Pillai K, Pourgholami M, Chua T, Morris D. Prognostic significance of Ki67 expression in malignant peritoneal mesothelioma. American Journal of Clinical Oncology. 2015;38(4):388-394
  96. 96. Kusamura S, Torres Mesa PA, Cabras A, et al. The role of Ki-67 and pre-cytoreduction parameters in selecting diffuse malignant peritoneal mesothelioma (DMPM) patients for cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). Annals of Surgical Oncology. 2016;23(5):1468-1473
  97. 97. Ghanim B, Klikovits T, Hoda MA, et al. Ki67 index is an independent prognostic factor in epithelioid but not in non-epithelioid malignant pleural mesothelioma: A multicenter study. British Journal of Cancer. 2015;112(5):783-792
  98. 98. Vigneri P, Martorana F, Manzella L, Stella S. Biomarkers and prognostic factors for malignant pleural mesothelioma. Future Oncology. 2015;11(24s):29-33
  99. 99. Li D, Wang B, Long H, Wen F. Diagnostic accuracy of calretinin for malignant mesothelioma in serous effusions: A meta-analysis. Scientific Reports. 2015;5:9507
  100. 100. Yukio T, Inai K, Ishikawa Y, et al. The trial of differentiation grading of epithelioid mesothelioma with reference to its clinicopathological significance. In: Kyoto: International Mesothelioma Interest Group Meeting. 2010. Abstr P10-1
  101. 101. Kao SC, Klebe S, Henderson DW, et al. Low calretinin expression and high neutrophil-to-lymphocyte ratio are poor prognostic factors in patients with malignant mesothelioma undergoing extrapleural pneumonectomy. Journal of Thoracic Oncology. 2011;6:1923-1929
  102. 102. Kao SC, Pavlakis N, Harvie R, et al. High blood neutrophil-to- lymphocyte ratio is an indicator of poor prognosis in malignant mesothelioma patients undergoing systemic therapy. Clinical Cancer Research. 2010;16:5805-5813
  103. 103. Linton A, Pavlakis N, O’Connell R, et al. Factors associated with survival in a large series of patients with malignant pleural mesothelioma in New South Wales. British Journal of Cancer. 2014;111:1860-1869
  104. 104. Thapa B, Walkiewicz M, Murone C, et al. Calretinin but not caveolin-1 correlates with tumour histology and survival in malignant mesothelioma. Pathology. 2016;48(7):660-665
  105. 105. Robinson BW, Creaney J, Lake R, et al. Mesothelin-family proteins and diagnosis of mesothelioma. Lancet. 2003;362(9396):1612-1616
  106. 106. Franko A, Dolzan V, Kovac V, Arneric N, Dodic-Fikfak M. Soluble mesothelin-related peptides levels in patients with malignant mesothelioma. Disease Markers. 2012;32:123-131
  107. 107. Cristaudo A, Foddis R, Vivaldi A, et al. Clinical significance of serum mesothelin in patients with mesothelioma and lung cancer. Clinical Cancer Research. 2007;13:5076-5081
  108. 108. Grigoriu BD, Scherpereel A, Devos P, et al. Utility of osteopontin and serum mesothelin in malignant pleural mesothelioma diagnosis and prognosis assessment. Clinical Cancer Research. 2007;13:2928-2935
  109. 109. Creaney J, Francis RJ, Dick IM, et al. Serum soluble mesothelin concentrations in malignant pleural mesothelioma: Relationship to tumor volume, clinical stage and changes in tumor burden. Clinical Cancer Research. 2011;17:1181-1189
  110. 110. Dipalma N, Luisi V, Di Serio F, et al. Biomarkers in malignant mesothelioma: Diagnostic and prognostic role of soluble mesothelin-related peptide. The International Journal of Biological Markers. 2011;26:160-165
  111. 111. Schneider J, Hoffmann H, Dienemann H, Herth FJ, Meister M, Muley T. Diagnostic and prognostic value of soluble mesothelin-related proteins in patients with malignant pleural mesothelioma in comparison with benign asbestosis and lung cancer. Journal of Thoracic Oncology. 2008;3:1317-1324
  112. 112. Creaney J, Yeoman D, Naumoff LK, et al. Soluble mesothelin in effusions: A useful tool for the diagnosis of malignant mesothelioma. Thorax. 2007;62:569-576
  113. 113. Linch M, Gennatas S, Kazikin S, et al. A serum mesothelin level is a prognostic indicator for patients with malignant mesothelioma in routine clinical practice. BMC Cancer. 2014;14:674
  114. 114. Creaney J, Dick IM, Meniawy TM, et al. Comparison of Fibulin-3 and mesothelin as markers in malignant mesothelioma. Thorax. 2014;69:895-902
  115. 115. Tian L, Zeng R, Wang X, Shen C, Lai Y, Wang M, et al. Prognostic significance of soluble mesothelin in malignant pleural mesothelioma: A meta-analysis. Oncotarget. 2017;8(28)
  116. 116. Timpl R, Sasaki T, Kostka G, Chu ML. Fibulins: A versatile family of extracellular matrix proteins. Nature Reviews. Molecular Cell Biology. 2003;4:479-489
  117. 117. Kobayashi N, Kostka G, Garbe JH, et al. A comparative analysis of the bulin protein family. Biochemical characterization, binding interactions, and tissue localization. The Journal of Biological Chemistry. 2007;282:11805-11816
  118. 118. Kirschner MB, Pulford E, Hoda MA, et al. Fibulin-3 levels in malignant pleural mesothelioma are associated with prognosis but not diagnosis. British Journal of Cancer. 2015;113:963-969
  119. 119. Hooper CE, Lyburn ID, Searle J, Darby M, et al. The south west area mesothelioma and pemetrexed trial: A multicentre prospective observational study evaluating novel markers of chemotherapy response and prognostication. British Journal of Cancer. 2015;112:1175-1182
  120. 120. Pass HI, Levin SM, Harbut MR, et al. Fibulin-3 as a blood and effusion biomarker for pleural mesothelioma. The New England Journal of Medicine. 2012;367(15):1417-1427
  121. 121. Anborgh PH, Wilson SM, Tuck AB, et al. New dual monoclonal ELISA for measuring plasma osteopontin as a biomarker associated with survival in prostate cancer: Clinical validation and comparison of multiple ELISAs. Clinical Chemistry. 2009;55(5):895-903
  122. 122. Pass H, Goparaju C, Espin-Garcia O, Donington J, et al. Plasma biomarker enrichment of clinical prognostic indices in malignant pleural mesothelioma. Journal of Thoracic Oncology. 2016;11(6):900-909
  123. 123. Kawabat SE, Bast RC, Bhan AK, Welch WR, Knapp RC, Colvi RB. Tissue distribution of a coelomic epithelium related antigen recognized by the monoclonal antibody 0025. International Journal of Gynecology. 1984;2:275-285
  124. 124. Bast RC, Klug TL, John E, et al. A radioimmunoassay using a mono clonal antibody to monitor the course of epithelial ovarian cancer. The New England Journal of Medicine. 1983;309:883-887
  125. 125. Koclma IA, Nap M, Rodenburg CJ, Fleuren GJ. The value of tumour marker CA125 in surgical pathology. Histopathology. 1987;11:287-294
  126. 126. Berardi R, Fiordoliva I, De Lisa M, Ballatore Z, Caramanti M, Morgese F, et al. Clinical and pathologic predictors of clinical outcome of malignant pleural mesothelioma. Tumori. 2016;102(2):190-195
  127. 127. Duan HJ, Itoh N, Yamagami O, Katsuyama T, Shigematsu H. Diffuse malignant mesothelioma in a young woman with high serum level of CA125. Acta Pathologica Japonica. 1991;41:158-163
  128. 128. Simsek H, Kadayifci A, Okan E. Importance of serum CA 125 levels in malignant peritoneal mesothelioma. Tumor Biology. 1996;17(1):1-4
  129. 129. Francart J, Vaes E, Henrard S, et al. A prognostic index for progression-free survival in malignant mesothelioma with application to the design of phase II trials: A combined analysis of 10 EORTC trials. European Journal of Cancer. 2009;45(13):2304-2315
  130. 130. Blayney JK, Ceresoli GL, Castagneto B, et al. Response to chemotherapy is predictive in relation to longer overall survival in an individual patient combined-analysis with pleural mesothelioma. European Journal of Cancer. 2012;48:2983-2992
  131. 131. Billé A, Krug L, Woo K, Rusch V, Zauderer M. Contemporary analysis of prognostic factors in patients with unresectable malignant pleural mesothelioma. Journal of Thoracic Oncology. 2016;11(2):249-255
  132. 132. Larson SM, Erdi Y, Akhurst T, et al. Tumor treatment response based on visual and quantitative changes in global tumor glycolysis using PET-FDG imaging. The visual response score and the change in total lesion glycolysis. Clinical Positron Imaging. 1999;2(3):159-171
  133. 133. Ceresoli G, Chiti A, Zucali P, et al. Early evaluation in malignant pleural mesothelioma by positron emission tomography with [18F]fluorodeoxyglucose. Journal of Clinical Oncology. 2006;24:4587-4593
  134. 134. Francis R, Byrne M, van der Schaaf A, et al. Early prediction of response to chemotherapy and survival in malignant pleural mesothelioma using a novel semiautomated 3-dimensional volume-based analysis of serial 18F-FDG PET scans. Journal of Nuclear Medicine. 2007;48:1449-1458
  135. 135. Chung MK, Jeong HS, Park SG, et al. Metabolic tumor volume of 18Fluorodeoxyglucose positron emission tomography/computed tomography predicts short-term outcome to radiotherapy with or without chemotherapy in pharyngeal cancer. Clinical Cancer Research. 2009;15(18):5861-5868
  136. 136. Veit-Haibach P, Schaefer N, Steinert H, et al. Combined FDG-PET/CT in response evaluation of malignant pleural mesothelioma. Lung Cancer. 2010;67:311-317
  137. 137. Lee H, Hyun S, Lee K, et al. Volume-based parameter of 18F-FDG PET/CT in malignant pleural mesothelioma: Prediction of therapeutic response and prognostic implications. Annals of Surgical Oncology. 2010;17:2787-2794
  138. 138. Basu S, Saboury B, Torigian DA, et al. Current evidence base of FDG-PET/CT imaging in the clinical management of malignant pleural mesothelioma: Emerging significance of image segmentation and global disease assessment. Molecular Imaging and Biology. 2011;13(5):801-811
  139. 139. Genestreti G, Moretti A, Piciucchi S, et al. FDG PET/CT response evaluation in malignant pleural mesothelioma patients treated with talc pleurodesis and chemotherapy. Journal of Cancer. 2012;3:241-245
  140. 140. Schaefer N, Veit-Haibach P, Soyka J, Steinert H, Stahel R. Continued pemetrexed and platin-based chemotherapy in patients with malignant pleural mesothelioma (MPM): Value of 18F-FDGPET/CT. European Journal of Radiology. 2012;81:e19-e25
  141. 141. Klabatsa A, Chicklore S, Barrington S, Goh V, Lang-Lazdunski L, Cook C. The association of 18F-FDG PET/CT parameters with survival in malignant pleural mesothelioma. European Journal of Nuclear Medicine and Molecular Imaging. 2014;41:276-282
  142. 142. Marin-Oyaga VA, Salavati A, Houshmand S, et al. Feasibility and performance of an adaptive contrast-oriented FDG PET/CT quantification technique for global disease assessment of malignant pleural mesothelioma and a brief review of the literature. Hellenic Journal of Nuclear Medicine. 2015;18(1):11-18
  143. 143. Zucali P, Lopci E, Ceresoli G, et al. Prognostic and predictive role of [18F]fluorodeoxyglucose positron emission tomography (FDG-PET) in patients with unresectable malignant pleural mesothelioma (MPM) treated with up-front pemetrexed-based chemotherapy. Cancer Medicine. 2017;6(10):2287-2296
  144. 144. Niccoli-Asabella A, Di Palo A, Altini C, et al. 18F-FDG PET/CT in therapy response and in predicting responders or non-responders in malignant pleural mesothelioma patients, by using semi-quantitative mRECIST and EORTC criteria. Hellenic Journal of Nuclear Medicine. 2018;21(3):191-197
  145. 145. Luo W, Rao M, Qu J, Luo D. Applications of liquid biopsy in lung cancer—Diagnosis, prognosis prediction, and disease monitoring. American Journal of Translational Research. 2018;10(12):3911-3923
  146. 146. Cavallari I, Urso L, Sharova E, Pasello G, Ciminale V. Liquid biopsy in malignant pleural mesothelioma: State of the art, pitfalls, and perspectives. Frontiers in Oncology. 2019;9:7-10
  147. 147. Khanna S, Thomas A, Abate-Daga D, et al. Malignant mesothelioma effusions are infiltrated by CD3+ T cells highly expressing PD-L1 and the PD-L1+ tumor cells within these effusions are susceptible to ADCC by the anti-PD-L1 antibody avelumab. Journal of Thoracic Oncology. 2016;11(11):1993-2005
  148. 148. Combaz-Lair C, Galateau-Salle F, McLeer-Florin A, et al. Immune biomarkers PD-1/PD-L1 and TLR3 in malignant pleural mesotheliomas. Human Pathology. 2016;52:9-18
  149. 149. Cedres S, Ponce-Aix S, Zugazagoitia J, et al. Analysis of expression of programmed cell death 1 ligand 1 (PD-L1) in malignant pleural mesothelioma (MPM). PLoS One. 2015;10(3):e0121071
  150. 150. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. The New England Journal of Medicine. 2015;372(21):2018-2028
  151. 151. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immunecorrelates of anti-PD-1 antibody in cancer. The New England Journal of Medicine. 2012;366(26):2443-2454
  152. 152. Valmary-Degano S, Colpart P, Villeneuve L, et al. Immunohistochemical evaluation of two antibodies against PD-L1 and prognostic significance of PD-L1 expression in epithelioid peritoneal malignant mesothelioma: A RENAPE study. European Journal of Surgical Oncology. 2017;43(10):1915-1923
  153. 153. Levallet G, Vaisse-Lesteven M, Le Stang N, et al. Plasma cell membrane localization of c-MET predicts longer survival in patients with malignant mesothelioma: A series of 157 cases from the MESOPATH group. Journal of Thoracic Oncology. 2012;7(3):599-606
  154. 154. Zucali PA, Giovannetti E, Destro A, et al. Thymidylate synthase and excision repair cross-complementing group-1 as predictors of responsiveness in mesothelioma patients treated with pemetrexed/carboplatin. Clinical Cancer Research. 2011;17(8):2581-2590
  155. 155. Henderson DW, Reid G, Kao SC, van Zandwijk N, Klebe S. Challenges and controversies in the diagnosis of malignant mesothelioma: Part 2. Malignant mesothelioma subtypes, pleural synovial sarcoma, molecular and prognostic aspects of mesothelioma, BAP1, aquaporin-1 and microRNA. Journal of Clinical Pathology. 2013;66:854-861
  156. 156. Zhou J, Zhong H, Zhang J, Jin S, Roudi R, Ma H. Development and validation of a prognostic signature for malignant pleural mesothelioma. Frontiers in Oncology. 2019;9:6-9

Written By

Rodríguez-Cid Jeronimo Rafael and Flores-Mariñelarena Rodrigo Rafael

Submitted: 06 December 2019 Reviewed: 14 February 2020 Published: 03 May 2020