Open access peer-reviewed chapter

Surgical Management of Malignant Pleural Mesothelioma: From the Past to the Future

Written By

Alice Bellini, Beatrice Aramini and Franco Stella

Submitted: 30 November 2021 Reviewed: 14 February 2022 Published: 20 June 2022

DOI: 10.5772/intechopen.103686

From the Edited Volume

Mesothelioma - Diagnostics, Treatment and Basic Research

Edited by Ilze Strumfa

Chapter metrics overview

115 Chapter Downloads

View Full Metrics

Abstract

Malignant pleural mesothelioma (MPM) is an aggressive malignancy with a poor prognosis, principally caused by a prior asbestos exposure. Up to the present, multimodality protocols including surgery with chemotherapy (CT) and/or radiotherapy (RT) represent the therapeutic gold standard for selected patients (epithelial and early-stage MPM). In this context, the aim of surgery is to accomplish the macroscopic complete resection (MCR). There are two main surgical options to obtain MCR—extrapleural pneumonectomy (EPP) and pleurectomy/decortication (PD). The superiority of one surgical approach over the other is still discussed. To date, the decision to carry out one or the other in a multimodal setting is established on surgeons’ preference more than on strong scientific evidence. Due to the high morbidity, both surgical techniques should be achieved in tertiary referral centres. In summary, surgery, CT, and RT have failed as single modality therapies with no effects on patients survival. This aspect may be justified by the lack of randomized trials. Thus, novel therapeutic strategies, such as multimodality treatment and targeted agents, seem to prolong the survival and the quality of life. The aim of this chapter is to provide a complete overview of the current surgical approaches to MPM, discussing within the frameworks of pre-operative diagnostic evaluation and multimodality oncological treatments.

Keywords

  • malignant pleural mesothelioma
  • extrapleural pneumonectomy
  • pleurectomy/decortication
  • multimodality treatment
  • chemotherapy
  • radiotherapy
  • target therapy

1. Introduction

MPM is a rare tumor that has become a world health issue due to its poor prognosis and its increasing incidence, largely due to prior asbestos exposure (the latency is about 20–40 years). Median overall survival (OS) is approximately 1 year in patients with MPM, and the 5-year OS rate is about 10% [1]. In recent years, there has been a notable advancement in the comprehension of MPM pathogenesis, leading to new promising drugs and therapeutic schemes [2, 3]. Particularly, recent trials including innovative drugs, such as targeted therapies or immunotherapies, have encouraged MPM patients [4]. Optimal treatment strategy in MPM has not yet been well established, consequently, current guidelines from the British Thoracic Society (BTS) [5], the American Society of Clinical Oncology (ASCO) [6], the National Comprehensive Cancer Network (NCCN) [1] and the European Society for Medical Oncology (ESMO) [7] have examined similar studies but reached different conclusions. Newly, a task force composed of the European Respiratory Society (ERS), the European Society of Thoracic Surgeons (ESTS), the European Association for Cardio-Thoracic Surgery (EACTS) and the European Society for Radiotherapy and Oncology (ESTRO) [8] proposed updated and practical guidelines on routine management of MPM, after a systematic review of the 2009–2018 literature, including new promising therapies and strategies. Up to the present, therapeutic strategies for MPM are still discussed; therefore, with a lack of a homogeneous consensus on this theme, physicians preferred to evaluate every single patient in a multidisciplinary team to adopt the best treatment based on the performance status of the patient and the stage of the tumor.

The current Eighth Edition of tumor, nodes, metastasis (TNM) classification for MPM [1] is reported in Tables 1 and 2. As stated in the aforementioned guidelines, at early stages (disease confined to the pleural envelope, without N2 lymph node involvement) with favorable histology (epithelial), a surgical approach with curative intent in a multimodal protocol appears to be indicated to enhance survival and quality of life. Instead, in advanced stages with distant spread palliative or supportive care must be preferred.

TPrimary tumor
TXPrimary tumor can not bed assessed
T0No evidence of primary tumor
T1Tumor limited to the ipsilateral pleural with or without the involvement of:
  • Visceral pleura

  • Mediastinal pleura

  • Diaphragmatic pleura

T2Tumor involving each of the ipsilateral pleural surfaces (parietal, mediastinal, diaphragmatic, and visceral pleura) with at least one of the following features:
  • Involvement of diaphragmatic muscle

  • Extension of tumor from visceral pleura into the underlying pulmonary parenchyma

T3Locally advanced but potentially resectable tumor.
Tumor involving all ipsilateral pleural surfaces (parietal, mediastinal, diaphragmatic, and visceral pleura), with at least one of the following features:
  • Involvement of the endothoracic fascia

  • Extension into the mediastinal fat

  • Solitary, completely resectable focus of tumor extending into the soft tissues of the chest wall

  • Nontransmural involvement of the pericardium

T4Locally advanced technically unresectable tumor.
Tumor involving all ipsilateral pleural surfaces (parietal, mediastinal, diaphragmatic, and visceral pleura), with at least one of the following features:
  • Diffuse extension or multifocal masses of tumor in the chest wall, with or without associated rib destruction

  • Direct transdiaphragmatic extension of the tumor to the peritoneum

  • Direct transdiaphragmatic extension of the tumor to the contralateral pleura

  • Direct transdiaphragmatic extension of the tumor to mediastinal organs

  • Direct transdiaphragmatic extension of the tumor to the spine

  • Tumor extending through to the internal surface of the pericardium with or without a pericardial effusion; or involving the myocardium

NRegional lymph nodes
NXRegional lymph nodes can not be assessed
N0No regional lymph nodes metastases
N1Metastases in the ipsilateral bronchopulmonary, hilar, or mediastinal (including the internal mammary, peridiaphragmatic, pericardial fat pad, or intercostal) lymph nodes
N2Metastases in the contralateral mediastinal, ipsilateral, or contralateral supraclavicular lymph nodes
MDistant metastasis
M0No distant metastasis
M1Distant metastasis present

Table 1.

Definitions for TNM for MPM (according to the current eighth edition).

TNM: tumor nodes metastasis; MPM: malignant pleural mesothelioma.

StageTNM
IAT1N0M0
IBT2–T3N0M0
IIT1–T2N1M0
IIIAT3N1M0
IIIBT1–T3
T4
N2
Any N
M0
M0
IVAny TAny NM1

Table 2.

Prognostic group for MPM (according to the current eighth edition).

MPM: malignant pleural mesothelioma; TNM: tumor nodes metastasis.

Because of the diffuse growth pattern and the lack of surgical margins, microscopic complete resection is theoretically impossible. Thus, a MCR should be the aim of the surgery, even though the optimal cytoreductive procedure is still controversial [9, 10]. There are two main surgical options to obtain MCR—EPP and PD; the superiority of one technique over the other is still debated [11]. Due to the high morbidity, both surgical techniques should be achieved only in tertiary referral centres with a wide experience in thoracic surgery [12]. Generally, surgery achieves only cytoreduction, hence it must be associated with induction CT (iCT) or adjuvant CT (aCT) with or without adjuvant RT (aRT) to achieve better outcomes in terms of survival and control of the disease.

The best combination of these different therapeutic approaches is still a matter of debate [13, 14]. Hence, the aim of this up-to-dated literature review is to provide a complete overview of the current surgical approaches to MPM, discussing within the frameworks of pre-operative diagnostic evaluation and multimodality oncological treatments.

Advertisement

2. Surgery for MPM

2.1 The importance of the MCR in the surgery for MPM

The MPM is characterized by a singular growth along the pleural surface, representing a challenge for its surgical resection to provide a microscopic free margin (R0 resection) avoiding its direct manipulation. Hence, the best surgical result is a MCR with microscopic positive margins (R1 resection) [9, 10, 15, 16]. For this reason, MCR came to be the main principle of surgery for MPM, based on retrospective data showing longer survival for MCR when compared to R2 resection [15, 17, 18]. As claimed by the literature, 30% of the patients addressed to surgery is found unresectable in the operating room [15]. Basically, it is important to improve the preoperative identification of an unresectable disease to prevent a futile explorative thoracotomy (ET), promote enrolment to medical therapies, and avoid expensive and not necessary costs [19].

2.2 The pre-operative role of computed tomography (CT) and spirometry

Across the literature, the most common factor precluding MCR is the diffuse chest wall invasion (DCWI), which is frequently associated with the contraction of the ipsilateral hemithorax in the CT scan [15, 20]. Growing up, MPM conducts to a restrictive syndrome and reduces the thoracic cage expansion and the diaphragmatic contraction, leading to a respiratory pump failure, as a necessary consequence [21, 22].

Recently, few authors analyzed the thoracic cage volume (TCV), the aerated lung volumes and the pleural thickness according to Response Evaluation Criteria in Solid Tumors (RECIST) modified criteria (the radiological parameters that correlated with the contracted hemithorax) and the pulmonary functions tests (PFTs), particularly the total lung capacity (TLC) (an indicator of the restrictive syndrome), as possible preoperative predictors of unresectability. Particularly, Burt et al. created a novel three-dimensional radiographic metric of the TCV, based on a fully manual segmentation, and demonstrated that a 5% decrease in TCV compared with the contralateral side was significantly associated with unresectability due to DCWI [15], while Bellini and co-workers used two methods already codified in the literature: the semi-automated segmentation of the aerated lung volumes [23] and the RECIST modified criteria measuring pleural thickness (the sum of the two maximum tumor thicknesses, perpendicular to the chest wall or mediastinum, measured at three levels, was reported as the disease burden) [24]. The Italian group found the TLC and the disease burden as independent predictors of unresectability in the multivariable analysis, with an optimal cut-off value of <77.5% and >120.5 mm, respectively; whereas the aerated lung volumes were significantly associated with ET only in the univariable analysis, probably due to the strong correlation with the disease burden. The PFTs seems to be an additional tool to better improve the preoperative identification of MPM disease not amenable to MCR [20], besides to be indicators of cytoreductive efficacy of iCT, as previously demonstrated by Marulli and collaborators [21, 22]. Moreover, both lung volumes and pleural thickness according to RECIST modified criteria play an important and consolidated prognostic role in MPM survival [23, 24, 25, 26, 27, 28]. In particular, the pleural thickness has been recently reported as a useful prognostic indicator of MPM—there is joint approval of the Eighth Edition of the tumor, node and metastasis (TNM) staging system and a recommendation to prospectively evaluate the importance of tumor volume or an approximation by tumor thickness [27, 28, 29, 30].

2.3 Surgical indications

Early stages MPM (stage I to IIIA according to Eighth Edition TNM staging system) in patients with epithelial subtype and good performance status represents the best indication for surgery. Conversely, absolute contraindications are patients with sarcomatoid or sarcomatoid-predominant histology, pN2 disease (according to Eighth Edition TNM staging system) and/or stage IV, unless in the context of clinical trials [1, 8, 30, 31]. Despite the overall poor prognosis of biphasic histology, according to recent multicentre analyses, a multimodal approach, including cancer-directed surgery, seems to be associated with improved long-term results in very selected patients with biphasic MPM [31, 32], mostly in patients with a lower proportion of sarcomatoid disease [33]. The ipsilateral nodal disease is not an absolute contraindication for surgery, in fact, the pattern of lymphatic drainage of the pleura does not follow the same pathway as for the lung parenchyma; mediastinal nodes may be the initial site of metastases before the lung parenchyma is involved. The International Association for the Study of Lung Cancer (IASLC) staging project recently reported no survival difference between traditional pN1 and pN2. Therefore, clinical and pathological N1 and N2 are combined into a single N1 category including all ipsilateral, intrathoracic nodal metastases, conversely, contralateral or all extrathoracic nodal metastases (N2 category) represent an absolute contraindication for surgery [34]. Before radical surgery, it is recommended to have a diagnosis not only based on cytology, because of the high risk of diagnostic error, but also on tissue confirmation by pleural biopsy (by either video-assisted thoracoscopy (VATS) or by mini-thoracotomy in the presence of fused pleural space, minimizing the number and size of incisions due to the risk of recurrence in the port-sites) to confirm the presence of microscopic subpleural fat tissue invasion and to allow for adequate immunohistochemical analysis [30].

2.4 Surgical procedures with curative intent: EPP vs. PD, which one to choose?

To obtain MCR there are two main surgical options with curative intent—EPP and PD. Both often allows to obtain only cytoreduction, hence surgical resection must be incorporated in multimodality regimens which include CT and/or RT in the neoadjuvant or adjuvant setting, to achieve better outcomes in term of survival and control of the disease [12, 13]. The EPP is a well-standardized procedure, based on the en bloc resection of the parietal and visceral pleura, ipsilateral lung, pericardium, and hemidiaphragm [35]; it has been deemed for many decades the best technique to achieve MCR with its survival benefits [9]. Conversely, PD is a lung-sparing approach, first reported in 1975 [36] and not yet homogenized in all centres: its description has changed according to the surgical technique, curative intent, and clinical indications [37]. Originally, it was suggested as a cytoreductive substitute in patients with a reduced cardiorespiratory reserve, which cannot tolerate the resection of the entire lung. In 2011, the International Mesothelioma Interest Group (IMIG) and the IASLC recommended that surgical procedures for MPM should be classified into three categories—(1) extended PD (EPD), (2) PD, and (3) partial pleurectomy [38], while mediastinal node sampling should be performed with a goal to obtain at least three nodal stations [1].

Both EPP and EPD required diaphragm and pericardial resections and reconstructions. Due to the lack of consistent guidelines, different materials (alloplastic and autologous) and techniques are available according to surgeons’ preferences, with the aim to maximize the strength of the patch and to decrease the complications rate. The most frequent complications after diaphragmatic and pericardial reconstructions are the patch dehiscence with abdominal herniation (mostly in the left side), inferior vena cava (IVC) stenosis, cardiac herniation, cardiac tamponade and infection [39].

The most popular material for diaphragmatic reconstruction is the 2 mm-thick expanded polytetrafluroethylene (e-PTFE), often in its dual mesh formulation (with the 2 different surfaces both reduce the adhesion of abdominal organs and facilitate the proliferation of cells in the thoracic side.), fixed with interrupted non-absorbable stitches across the ribs. The use of a synthetic alloplastic material on one hand permits an improvement in resistance, but on the other hand, it is characterized by a non-insignificant risk of infection (2.4%), while the herniation risk oscillates from 3.8–12%, in particular, the left posterior mediastinum represents the area with the highest incidence of patch dehiscence [39]. To reduce the risk of gastric herniation, it may help leave a small rim (maximus 2 cm) of autologous diaphragm next to the aortic arch and oesophagal hiatus to anchor the patch. On the right side, the herniation of the abdominal organs is less common because of the presence of the liver. However, surgeons should pay attention to preventing the IVC stenosis from leaving a short rim of diaphragmatic tissue or fixing the diaphragmatic mesh to the pericardium edge or the pericardium patch [39].

Similar to a diaphragmatic replacement, synthetic patches are preferred for the repair of the pericardium: among the non-permeable group, the most used is the 0.1 mm-thick e-PTFE, while in the permeable group the polyester and polypropylene prosthesis. The patch is generally sutured with interrupted non-absorbable stitches beginning from the deeper posterior part, while in the inferior side it might be fixed to the diaphragmatic mesh increasing the pericardial space. In fact, the purpose of the pericardial reconstruction is to allow a normal cardiac function, preventing tamponade or diastolic dysfunction. It could be helpful both the fenestration of the mesh or its anchorage leaving unfixed it’s superior part for the regular outflow of the pericardial fluid in the directions of the pleural space [39].

EPP involves en bloc resection of the visceral and parietal pleura, lung and, if necessary, ipsilateral hemidiaphragm, and pericardium (Figure 1). The lung removal allows administrating a higher dose of RT with no risk of radiation pneumonitis, improving the local control of the disease. This procedure was first employed in 1976 [40], becoming the treatment of choice for potentially resectable MPM. In 1999, Sugarbaker and colleagues reported a 5-year OS rate of 46% and a low mortality rate for patients affected by an early stage epithelial MPM, underwent EPP in a multimodal regimen [12]. Subsequently, there have been different series demonstrating a similar trend with a OS of 20–24 months [35, 41]. One decade ago, in a European survey composed of 802 thoracic surgeons, EPP was considered more efficacious than PD and the supplementation with aCT or other associations of multimodal treatments were deemed to enhance the possibility of cure [42]. Nevertheless, its survival advantages, EPP is charged by some disadvantages: it is a debilitating surgical procedure, associated with a morbidity rate of almost 50% and a mortality rate of 5% even in tertiary referral centres, with high expertise in the surgical management of MPM [41].

Figure 1.

(a and b) En bloc resection of the lung, parietal, and visceral pleural with diaphragm and pericardium after extrapleural pneumonectomy.

In particular, it is associated with a reduction in quality of life, a worsening of postoperative cardiorespiratory function, and difficulties in administration, tolerance, and compliance of adjuvant therapy. A single centre trial (Surgery for Mesothelioma After Radiation Therapy, SMART) embraced a novel protocol, consisting in EPP after intensity-modulate radiation therapy (IMRT) (a short hemi-thoracic high dose technique), with good early and long-term results [43]. However, the employment of EPP as part of the multimodal treatment of MPM has been recently debated after the Mesothelioma and Radical Surgery (MARS)-1 trial reports. This wide randomized trial, comparing EPP with no surgery in terms of survival and quality of life, concluded that “EPP within trimodal therapy offers no benefit and possibly harms patient” [44]. Anyhow, these results were controversial because survival was not the primary outcome of the study, the sample size was small, and the surgical mortality was higher than expected—this trial, in fact, faced several problems in the enrolment of patients with few cases treated by few centres with a not acceptable high mortality rate in the EPP arm that finally conditioned the survival results [45].

PD involves the total resection of both the parietal and visceral pleura, while the lung is spared (Figure 2).

Figure 2.

(A and B) Pleurectomy/decortication (PD). (C) Pathological specimen (visceral and parietal pleura) after PD.

As claimed by the IMIG classification, it is categorized in [38]:

  • Extended PD: the parietal and visceral pleurectomy associated with the resection of the pericardium and/or diaphragm;

  • PD: the parietal and visceral pleurectomy without the removal of the diaphragm or pericardium;

  • Partial pleurectomy (PP): the partial removal of the parietal and/or visceral pleura.

The first employment of pleurectomy for MPM was in 1975 by Martin and collaborators, who reported a median OS of 16 months in a series of 14 patients [36], extended the year later with 33 MPM patients with a median OS of 21 months [46].

Since then, several non-randomized studies have demonstrated the feasibility and safety of PD with various multimodality schemes involving induction and adjuvant treatments [37, 47, 48]. The preservation of the ipsilateral lung is the main advantage of PD compared to EPP, in fact, it allows a surgical treatment even in patients with a marginal cardiopulmonary reserve, making more feasible adjuvant therapies. The efficacy and radicality of PD in advanced MPM are controversial, however, data from literature are divergent [49]. Almost one decade ago, in an editorial Raja Flores [50] underlined the general trend of thoracic surgeons moving from EPP to PD, due to the lack of solid evidence about a significant survival difference between the aforementioned two surgical procedures [51]. According to the author, the main goal of surgery is the removal of as much tumor as possible preventing pneumonectomy with a consequent reduction in perioperative morbidity and mortality.

On the basis of the currently available data the equation tips in favor of PD rather than EPP. The MARS-2 trial [48], a phase III study of 328 patients with resectable MPM of any sub-type, recently completed the recruitment and, in a little over 2 years, will address the question of whether PD adds any survival benefit to systemic CT alone. While awaiting the results of MARS-2, we are justified in offering surgery as part of multimodality treatment to those with the best prognostic factors, ie, epithelioid with no clinical evidence of nodal disease [30]. The correct surgical strategy must be planned with the intention to accomplish MCR opting for the less invasive technique, basically, surgeons should enter the operating room with the intention to perform PD, except in case of extensive lung invasion. With the lack of randomized controlled trials comparing the two intended to treat techniques, it is still debatable which one provides better long-term outcomes. This is the reason why until now there is not a unique therapeutic approach for MPM and physicians base their decision according to their expertise, the performance status of the patient and the characteristic of the neoplasm. Anyhow, it is important that the sick person and his relatives acquire satisfactory information about both the disease and the available treatments [52].

Among the major complications after both surgical techniques, haemothorax is one of the most frequent, mostly after EPP (1–20.6%) than PD (0–4%). In fact, the extensive pleurectomy and the creation of a post-pneumonectomy cavity increase the risk of bleeding. Surgeons should meticulously verify the hemostasis at the end of the procedure in presence of a normal blood pressure, often using tissue sealants, argon-beam coagulation or oxidized regenerated cellulose products.

Empyema is a frequent complication after surgery for MPM (EPP 1.5–29.7%, PD 4–6.8%), often consequently the development of a bronchopleural fistula (BPF) after EPP (1–12%) in debilitated patients, because of the neoadjuvant treatments and the surgical procedure itself. To prevent the development of such life-threatening complication, it is mandatory keeping the bronchial stump as short as possible, to avoid blind-end secretion retention, and prevent excessive devascularisation of the bronchus. To date, there is no evidence that the preventive use of bronchial stump coverage decreases the rate of BPF after EPP. Late empyema could also occur, several weeks after the intervention, in this case often not associated with BPF.

The peculiar and commonest complication after PD is the prolonged air leakage (3.5–57%), consequently the peeling of the visceral pleura with the underlined lung damage. This kind of complication could also cause an empyema due to ascending infection through the longtime chest drain. Surgeons should carefully reduce the post-operative air leaks, mending the lung with stapling device, sutures and sealant. Most of the time conservative management is a correct strategy with the removal of the active suction as soon as possible and the preservation of the chest tube until the resolution of the air leaks with a satisfactory lung expansion, which may take even 2–3 weeks. The post-operative management after PD is not well standardized among the centres—to prevent bleeding and air leakage, some centres prefer to keep patients on mechanical ventilation with positive and expiratory pressure (up to 48 h) to maintain the maximal lung inflation, which aids both the parietal hemostasis by compression and the closure of the parenchymal wounds; other centres prefer to reduce positive pressure ventilation to minimize the air leaks by extubating the patient as soon as possible [37].

2.5 Extracorporeal life support (ECLS) for life-threatening complications in MPM’s surgery: is it worthwhile?

In thoracic surgery, the use of ECLS in the postoperative period is augmented in the last decades [53]. Up to now, the unique absolute contraindication to extracorporeal membrane oxygenation (ECMO) is a pre-existing state incompatible with healing, such as an end-stage tumor [54]. Literature provides only a few reports on ECLS as a bridge to support oncological patients affected by complications of their illness [55], its therapy [56, 57] or cardiac arrest [58]. MPM is a locally aggressive tumor, with a very poor prognosis. The multimodality therapies including surgery offered to selected cases (early stages with epithelial histology) are often characterized by major and/or minor perioperative morbidities [13]. According to Burt et al., the EPP is burdened by a significantly higher rate of acute distress respiratory syndrome (ARDS) (8.4 vs. 0.8%) and 30-day mortality (10.5 vs. 3.1%), compared to the PD [52, 59]. Anyhow, in the case of perioperative complications after surgery for MPM the employment of ECLS represents an ethical dilemma due to the fatal nature of this malignancy.

Fica and collaborators mentioned the use of a single-site veno-venous ECMO to support ventilation in an early post-pneumonectomy broncho-pleural fistula [57], while Bellini and collaborators successfully used the veno-arterial (V-A) ECMO as cardiac support in two MPM patients (66%), conversely, the only case (33%) of V-A ECMO implanted primarily for respiratory support in pneumonia-associated ARDS of the residual lung had a negative outcome [60]. Similar results were reported in the literature; according to Gow and collaborators, patients with a better pulmonary reserve and cardiac indication for ECLS are the better oncological candidates for ECMO [61].

The disease process itself and/or the employed treatments lead patients with thoracic cancer to have less pulmonary reserve compared to adults generally demanding ECLS. Secondary infections and bleeding are the major problems for the use of ECLS for oncological patients [61], both potentially life-threatening. In this scenario, on one hand, we have potentially reversible complications not responsive to conventional therapies, while on the other hand frail and immunosuppressed patients with poor prognosis and at risk to develop life-threatening ECMO-related drawbacks.

In accordance with the aforementioned recent literature, in case of a potentially reversible condition especially if heart-related, ECLS could be used as a stopgap device until common therapies work, in very selected MPM patients, permitting the recovery and the completion of the multimodal protocol [60, 61].

2.6 Palliative surgery

The MesoVATS trial is an open-label randomized controlled trial conducted in 12 centres in the United Kingdom, that compared PP by VATS versus talc pleurodesis in patients with MPM [62]. There were no differences between groups in the OS at 1 year nor at 6 months of follow-up. Furthermore, the benefits of VATS-PP (better quality of life, less short-term pleural effusion) do not balance the inconveniences (surgical complications and longer hospital stay leading to more costs). Guidelines strongly recommend talc poudrage via thoracoscopy to control a recurrent MPM effusion as the first choice to achieve pleurodesis in patients with expanded lungs, while weakly suggest, with a low grade of recommendation, palliative VATS-PP to obtain pleural effusion control in symptomatic patients fit enough to undergo surgery who cannot benefit from (or after the failure of) chemical pleurodesis or indwelling catheter [8].

2.7 Surgery for MPM relapse

Recurrence of MPM after multimodality treatment is a common problem. Nevertheless, there has been no established therapy for relapse to date. Major studies about the treatment of recurrent MPM are reported in Tables 3 and 4. Over the literature, MPM with distant spread (associated or not with local relapse) is the most frequent pattern of recurrence, mostly in the EPP group, while the PD group showed a higher local-only failure rate [63, 64, 65, 66, 67, 68, 69, 70, 71]. A poor prognosis for recurrent MPM after multimodality treatment has been reported in the literature, with a median post-recurrence survival (PRS) after EPP ranging from 3 to 6.5 months [64, 65, 66, 72]. Newly, comfortable PRS were described after PD by Nakamura et al. and Kai and collaborators (14.4 and 20 months, respectively) [65, 67].

AuthorSurgery, NMultimodality, NRelapse, N (%)Pattern of recurrence, %
Kostron, 2016 [63]EPP, 136Bimodal, 47
Trimodal, 59
106 (77.9)L 24.3
D 19.9
L + D 33.8
Takuwa, 2017 [64]EPP, 59Bimodal, 27
Trimodal, 12
39 (66.1)NR
Kai, 2018 [65]EPP, 29
PD, 15
Bimodal, 26
Trimodal, 18
32 (72.7)L 18.2
D 27.3
L + D 27.3
Soldera, 2019 [66]EPP, 93Bimodal 43
Trimodal 10
53 (57.0)L 5.4
D 38.7
L + D 12.9
Nakamura, 2020 [67]PD, 90Bimodal, 9057 (63.3)L 43
D 6.7
L + D 13.3
Politi, 2010 [68]EPP, 8NR8 (100)L 50
D 50
Okamoto, 2013 [69]EPP, 10NR8 (80)L 40
D 40
Burt, 2012 [70]EPP, 32
PD, 15
NR47 (100)L 100
Bellini, 2021 [71]EPP, 49
PD, 45
Bimodal, 18
Trimodal, 76
94 (100)L, 28.7
D, 28.7
L + D, 42.6

Table 3.

Major studies about the treatment of recurrent MPM: Multimodality regimen and pattern of failure.

MPM: malignant pleural mesothelioma; EPP: extrapleural pneumonectomy; PD: pleurectomy/decortication; NR: not reported; L: local; D: distant; L + D: local+distant.

AuthorMedian DFS (m)Relapse treatment, N (%)Median PRS (m)Median OS (m)
Kostron, 2016 [63]9None, 28 (26.4)
Surgery, 16 (15.1)
Medical treatment, 73 (68.9)
722a
Takuwa, 2017 [64]11.6None, 12 (30.7)
Medical treatment, 27 (69.2)
6.522
Kai, 2018 [65]Overall, 14b
EPP, 13b
PD, 21b
Medical treatment, 17 (53.1)Overall, 5
EPP, 3
PD, 20
Overall, 22b
EPP, 17b
PD, 34b
Soldera, 2019 [66]NRNone, 27 (50.9)
Medical treatment, 15 (28.3)
NR, 11 (20.8)
4.8NR
Nakamura, 2020 [67]19Surgery, 3 (5.3)
Medical treatment, 40 (70.2)
Best supportive care, 14 (24.5)
14.457b
Politi, 2010 [68]NRSurgery, 8 (100)14.5NR
Okamoto, 2013 [69]15.4Surgery, 2 (25)
Medical treatment, 6 (75)
17.849.6
Burt, 2012 [70]16.1Surgery, 47 (100)Epithelial, 20.4
Biphasic, 7.0
44.9
Bellini, 2021 [71]Overall, 14
EPP, 20
PD, 11
None, 13 (13.8)
Surgery, 13 (13.8)
Medical treatment, 68 (72.3)
Overall, 12
EPP, 14
PD, 8
Overall, 33
EPP, 38
PD, 23

Table 4.

Major studies about the treatment of recurrent MPM: Oncological outcomes.

MPM: malignant pleural mesothelioma; EPP: extrapleural pneumonectomy; PD: pleurectomy/decortication; NR: not reported; L: local; D: distant; L + D: local+distant; DFS: disease-free survival; PRS: post recurrence survival; OS: overall survival, calculated from the date of surgery, except (a) from the first cycle of neoadjuvant chemotherapy; (b) from the date of pleural biopsy.

Conversely, Bellini and collaborators recently noted that the type of surgical resection did not affect the PRS (14 and 8 months in the EPP and PD group, respectively) if patients are fit enough to receive post-recurrence treatments [71]. Across the literature, the post-recurrence treatment is the main predictor of better PRS [63, 65, 67], in particular, Bellini and co-authors found tailored medical therapies as the best strategy to face relapse, even in the case of local failure [71], in contrast with satisfactory PRS after redoing surgery, which was reported by Kostron et al. [63]. The Italian group cautiously hypothesized that the early local-only failure may likely reflect a less radical local resection that could benefit from timely systemic therapies, rather than redo surgery that is rarely radical in most of the cases [71]. Moreover, several authors reported a long disease-free survival (DFS) (≥12 months) as significantly associated with good survival [61, 67, 71], probably reflecting a slower tumor growth speed associated with a less aggressive recurrent disease. Furthermore, epithelial histology [65, 71] and local recurrence [71] resulted as a favorable prognostic factor for PRS, the latter may be due to a less deleterious effect on performance status and, consequently, on survival compared with distant spread [71]. In conclusion, in patients presenting with recurrence of MPM after an MCR procedure, radical surgery to resect the recurrent tumor could have a role in the improvement of survival in selected patients [73].

Advertisement

3. Multimodality treatment for MPM

3.1 Bimodal and trimodal therapy in MPM

The microscopic complete resection represents an unattainable goal for surgery alone in MPM disease. For this reason, in the last decades, the surgical approach is mainly used as cytoreduction with improved survival, but with the necessity at least to combine with a bimodal/trimodal treatment, although the income of the novel strategies as immunotherapy, are suggesting a multimodal approach [40, 62, 74, 75]. The debate regarding the possible surgical approach as the closest to the radicality for MPM is still opened [76, 77]. In particular, the believers for EPP as the most oncologically correct approach, strongly support the theory that a near-complete surgical resection associated with chemo-or high-dose aRT may be the best treatment for earlier stages [78]. Recent studies have shown improved survival with EPP associated with neoadjuvant or adjuvant chemoradiotherapy [79], in highly-selected patients, although this type of surgery is very aggressive and invasive and not far from postoperative complications with significant morbidity (25%) and mortality (4–15%) [5]. In 2011 the MARS-1 trial compared patients treated with EPP and patients without, defining this surgery as not effective for the high morbidity and 30-day mortality. Particularly, it was a feasibility multicentre randomized controlled trial carried on between October 2005 and November 2008 in 12 English Hospitals. It included 112 patients aged 18 years or older affected by MPM and fit enough to undergo trimodal treatment. In the pre-randomization registration phase, all patients underwent induction platinum-based CT, followed by a clinical revaluation. The main reasons for not proceeding to randomization were a progression of the disease (33 patients), inoperability (five patients), and patient choice (19 patients). Finally, 50 patients were randomly assigned (1:1): 24 to EPP followed by radical RT and 26 to no EPP. The EPP was completed in 16 patients (in five patients it was not started and in three it was abandoned). The clinical outcomes evaluated were the proportion of patients of the EPP group who completed the trimodal treatment; perioperative mortality; quality of life; OS; and disease-free survival. Of the 16 surgical treated patients, there were tw perioperative deaths, while eight completed the trimodal protocol receiving the radical RT. Serious adverse events were higher in the EPP group (n = 10) than in the no EPP group (n = 2). The median OS for the EPP and the no EPP group were 14.4 months and 19.5 months, respectively. The median DFS for the EPP and the no EPP group were 7.6 months and 9 months, respectively. There was a statistically significant difference in the survival outcomes, while a trend toward a lower quality of life in the EPP group was reported.

Following the MARS-1 trial, the scientific community and surgeons focused their attention on PD, which is for sure less invasive than EPP, but not less effective than EPP [80, 81]. In particular, in the last decades, some retrospective studies and systematic reviews described a comparable survival between the two procedures, but with less morbidity and mortality for the patients treated with PD [51, 74, 82, 83, 84, 85] with the association of improved quality of life [84, 85, 86], although there are also research groups showing no difference in morbidity and mortality [87, 88, 89]. For the fact that data are still extremely different [90], the most focused expert for MPM suggests that a multidisciplinary approach and randomized controlled trials need to be set to further define the best surgery in MPM [42, 90, 91, 92, 93, 94]. In particular, the MARS-2 trial is trying to set if PD plus iCT may offer an improvement of the survival than the only CT [48, 91, 95]. Regarding the most updated recommendation, several scientific societies use a trimodal approach (surgery, CT and RT) for MPM) [1, 6, 7, 96]. This has been also confirmed by the BTS and the European Respiratory Society for clinical trials [5, 97]; although the timing for this therapy is still unknown as well as also the sequences of each treatment [97]. However, even in the case of the trimodal approach, the long-term survival of these patients remains still poor and only 5% survive at 5 years [98]. The association of pemetrexed (folate antimetabolite) plus B12 and folic acid supplementation [1, 5, 6, 7, 97] is an association with the most standard platinum-based therapy, cisplatin, typically, or carboplatin [99, 100], seems to improve the survival by 2.8 months compared with single drug treatment, probably for the fact that B12 and folic acid supplementation reduce the toxicity, especially in elderly patients [101]. In a recent large phase III trial, MAPS, even the use of bevacizumab, a vascular endothelial growth factor (VEGF) inhibitor in combination with pemetrexed/cisplatin is strongly recommended, which compares pemetrexed/cisplatin alone [102]. In particular, this trial showed improved survival of 2.7 months in unresectable MPM [102]. The ASCO has suggested vinorelbine as second-line treatment, although the NCCN suggested immunotherapy and CT as second-line treatment [1]. In particular, the second-line pemetrexed seems to have some effect against tumor, reducing the tumor progression, as described in a phase III trial [103]; for vinorelbine, the recent randomized controlled trials showed improved survival in patients treated with this drug [104] with good control of symptoms. There are other treatments as PD with intraoperative intracavitary hyperthermic CT, although more studies need to support and analyze concerning the long-term results [105, 106, 107, 108, 109, 110, 111, 112, 113]. The majority of studies that evaluate the trimodal approach are retrospective reviews [114, 115, 116, 117, 118, 119]. Since 2007, a multicentric clinical trial demonstrated that iCT plus EPP is feasible [41] showing an OS of 23 months compared with patients not treated with surgery with a survival of 19.8 months [41]. An Italian study published similar results [120]. The combination with neoadjuvant cisplatin/pemetrexed treatment has been discussed since 2009 in a multicentric phase II clinical trial in which patients in stage I-III MPM underwent 4 cycles of cisplatin/pemetrexed. Of these patients, who showed a good response to this medical treatment has been then underwent EPP followed by adjuvant hemithoracic radiation [121] with a median survival of 29.1 months with a 2-year survival of 61.2% [122]. These data let the researcher to conclude that a trimodal therapy may be effective and beneficial [122]. In 2010 the European Organization for Research and Treatment of Cancer (EORTC) published a similar multicentre phase II study [121], in which 65% of the patients underwent EPP 65% plus aRT showed a good result for the 42% of patients, although the neoadjuvant therapy associated with EPP, and adjuvant therapy have been shown to be challenging for the poor long term results [41, 116, 117, 118, 119]. With regards to the less invasive surgical treatment in MPM, represented by PD, few studies considered the use of trimodal therapy with PD. In particular, in 2012, a non-randomized prospective trial compared EPP to PD in trimodal approach [82] comparing 3 cycles of either cisplatin/gemcitabine or cisplatin/pemetrexed before EPP and aRT to PD with hyperthermic pleural lavage with povidone-iodine and aCT [82]. However, the median survival was 12.8 months for EPP patients, although a better survival of 23 months has been noted for the second group of patients. These results demonstrated that PD is a more feasible approach with better outcomes for MPM patients underwent trimodal therapy [82]. The role of adjuvant or neoadjuvant radiation therapy is not yet been clarified. In particular, the classic hemithoracic radiation of the entire pleural cavity after EPP is not a problem [123], although the lungs and the other organs cannot be spared from the radiation [124]. However, recent retrospective studies have shown an increased local recurrence in patients treated with adjuvant IMRT following EPP [125]. Recently, the SMART trial concluded the analysis about the role of neoadjuvant IMRT in T1–3N0 MPM followed by EPP, associated with aCT (cisplatin and an anti-folate) in case of mediastinal lymph nodes involvement, achieving encouraging survival results. The results were satisfying in consideration of a median OS of 42.8 months for the epithelial subtype, compared with 18 months for the biphasic one. The authors postulated that a probable mechanism for the distant spread is the spillage of the tumor cells into the coelomic cavities during EPP, hence the IMRT immediately before surgery could inactive these cells making them non-viable, preventing distant seeding with better survival outcomes. Probably the epithelial subtype is more sensible to the action of IMRT. Possible confounder factors for these promising survival results may be as follow: firstly, the criteria for inclusion in the trial were strict leading to a possible selection bias, secondly, the presence of BAP1 mutations (associated with better OS regardless of therapy) was not routinely investigated, so the potential inclusion of these patients in the trial could confound. [43]. In 2016 a group from Memorial Sloan Kettering Cancer Centre (MSKCC) showed the feasibility of IMRT and iCT and PD in IMPRINT phase II trial [126]; however, the same researcher’s group published a retrospective study comparing PD trimodal therapy with conventional RT to hemithoracic IMRT [126] with better survival for the patients treated with IMRT [127]. In US both ASCO and NCCN- guidelines strongly support the multimodality treatment for stages I-III epithelioid MPM [128]. The upcoming MARS-2 results may be very helpful to clarify the feasibility and benefits of the multimodality treatment [128]. Table 5 summarizes the main studies commented on in this paragraph.

Author, year, type of studyType of surgery, NMultimodal treatment, NMedian OS (m)
Spaggiari L, 2014, multicentre retrospective [79]EPP, 518iCT, 271
aRT, 213
aCT, 43
aRT+aCT, 117
18
Fahrner R, 2012, single centre retrospective [98]EPP, 21Port-site RT, 18
iCT, 19:
platinum+gemcitabine, 15
platinum+pemetrexed, 4
aRT, 16
707 days
Katirtzoglou N, 2010, multicentre phase II[100]Unresectable, 62CT with carboplatin+pemetrexed14
Zalcman G, 2016, multicentre randomized controlled open-label phase III [102]Unresectable, 448Cisplatin+bevacizumab, 223
Cisplatin only, 225
With bevacizumab: 18.8
Cisplatin only: 16.1
Muers MF, 2008, multicentre randomized [104]Unresectable, 409ASC alone (treatment could include steroids, analgesics drugs, bronchodilatators, palliative radiotherapy), 136
ASC plus mitomycin+vinblastine+cisplatin, 137
ASC plus vinorelbine, 136
ASC alone: 7.6
ASC plus CT: 8.5
Burt BM, 2018, single centre phase I [107]EPP, 59
PD, 41
Debulking, 4
HIOC with gemcitabine added to cisplatin, 104
aCT, 45
aRT, 10
All 20.3
EPP 17.7
PD 38.8
Opitz I,. 2020, single centre phase I [109]PD, 12Intracavitary cisplatin-fibrin, 1221
Ried M,. 2013, single centre prospective observational [111]PD, 10HIOC with cisplatin, 10NR
Sugarbaker DJ, 2013, single centre retrospective [113]EPP, 74 (53 HIOC group)
PD, 29 (19 HIOC group)
HIOC with cisplatin, 72
No HIOC, 31
HIOC: 35.3
No HIOC: 22.8
Rusch VW, 2001, single centre phase II [114]EPP, 62
PD, 5
aRT, 57 (54 EPP; 3 PD)EPP: 17
Early stages 33.8
Advanced stages 10
de Perrot M, 2009, single centre retrospective [116]EPP, 45
Unresectable, 15
iCT:
Cisplatin+vinorelbine, 26
Cisplatin+pemetrexed, 24
Cisplatin+raltitrexed, 6
Cisplatin+gemcitabine, 4
aRT, 30
Trimodality completed 59
Thieke C, 2015, single centre retrospective [118]EPP, 62iCT, 62
cisplatin+pemetrexed, 30
carboplatin+pemetrexed, 23
cisplatin+gemcitabine, 9
aRT (IMRT), 62
20.4
Rea F, 2007, single centre prospective [120]EPP, 17iCT with carboplatin+gemcitabine, 21
aRT, 15
All 25.5
EPP, 27.5
Van Schil PE, 2009, multicentre phase II [121]EPP, 42iCT with cisplatin+pemetrexed, 55
aRT, 37
All 18.4
Trimodality completed 33
Krug LM, 2009, multicentre phase II [122]EPP, 54iCT, 77
aRT, 40
All 16.8
Trimodality completed 29.1
Cho BC, 2021, single centre phase IIEPP, 96
Epithelial, 83
Biphasic, 8
Unkown, 5
iRT (IMRT), 9624.4
Epithelial: 42.8
Biphasic:18
Rimner A, 2016, multicentre phase II [126]PD, 8
Partial pleurectomy, 13
Unresectable, 11
iCT, 45:
Cisplatin+pemetrexed, 26
Carboplatin+pemetrexed, 18
Combination, 4
aRT (IMPRINT), 27
Trimodality completed 23.7
Shaikh F, 2016, single centre retrospective [127]PD, 209aRT:
conventional, 131
IMPRINT, 78
Chemotherapy, 85 (15 conventional, 70 IMPRINT)
Conventional,12.3
IMPRINT, 20.2

Table 5.

Summary of the main studies commented in Section 3.1.

OS: overall survival; EPP: extrapleural pneumonectomy; iCT: induction chemotherapy; aRT: adjuvant radiotherapy; aCT: adjuvant chemotherapy; PD: pleurectomy/decortication; ASC: active symptom control; HIOC: heated intraoperative chemotherapy; NR: not reported; iRT: induction radiotherapy; IMPRINT: intensity-modulated pleural radiotherapy; IMRT: intensity-modulated radiotherapy.

3.2 New target therapies in MPM

Although the scientific community is studying genes and proteins which may be used to set new treatments against MPM, the targets for mesothelioma have not been clearly yet identified. Besides the fact that the common MPM treatments including surgery, CT and radiation therapies show a poor OS from 9 to 17 months from the diagnosis [120, 129, 130], new emerging treatments are setting from the scientific community to improve the quality of life and the survival. The most used approach in this field is immunotherapy which seems to play an important role in MPM for the connections and reactions with the patient immunity, driving immunoregulatory mechanisms with a direct good response [131, 132]. In particular, in patients with MPM showing a high infiltrate of cytotoxic CD8+ T associated with programmed death-ligand 1 (PDL-1) expression, the use of pembrolizumab and nivolumab, or nivolumab with the cytotoxic T lymphocyte antigen 4 (CTLA-4) antibody ipilimumab showed very encouraging results [133, 134].

Several trials have been published showing the use of immunotherapy in advanced MPM alone or with standard CT. In particular, pembrolizumab seems to improve by 22%, the response rate compared with gemcitabine or vinorelbine which showed a 6% of response [135]. However, other immunotherapies have been analyzed in the last decades, for example, in MAPS2 open-label, phase 2 trial that has been studied in 21 French hospitals aiming to set nivolumab alone or combined with ipilimumab. This study showed a 12-weeks disease control in 44% of patients who received only nivolumab, compared with the 50% who underwent nivolumab plus ipilimumab [130]. In January of this year, the Checkmate 743 study was published in Lancet, which reported the superiority of the combination of nivolumab (3 mg/kg every 2 weeks) + ipilimumab (1 mg/kg every 6 weeks) over the above standard. With a significant advantage in the entire study population, particularly marked in the sarcomatoid subtype [136]. For patients progressing to the first line, however, no scheme has been shown to improve survival; however, these patients can be offered gemcitabine, vinorelbine or rechallenge with pemetrexed mono-CT, with unfortunately marginal benefits. Nivolumab, on the other hand, in a phase 3 study (CONFIRM) conducted against placebo in a heavily pretreated population, has shown efficacy in improving both progression-free survival and OS, but at the moment the drug is not approved in Italy. this indication [137].

From preclinical evidence, the role of angiogenesis in the development of this disease and the potential efficacy of inhibiting this mechanism appears to be relevant, although in lines subsequent to the first some antiangiogenic drugs have already been tested without success [138]. In particular, there is a strong rationale for angiogenesis inhibition in mesothelioma. VEGF plays an important role as an autocrine growth factor and strong mitogen in mesothelioma. Furthermore, the abnormal tumor vasculature increases the interstitial pressure and hypoxia, which may hinder the effect of the anticancer drugs against mesothelioma cells. Ramucirumab is a fully humanized monoclonal antibody directed against the VEGF receptor 2 (VEGFR-2), currently reimbursed in Italy for use in clinical practice in gastric cancer and tested in numerous other solid tumors [139]. VEGFR-2 is expressed not only in 90% of mesothelioma cells but also on the surface of macrophages present in the tumor microenvironment, considered to be responsible for the resistance to chemo- and immunotherapeutic treatments [140]. Inhibiting this receptor seems to allow the switch from a hypoxic and treatment-resistant environment to a more sensitive and immuno-permissive tumor milieu. In this context, the RAMES study was born, a multicentre, phase 2, randomized and double-blind trial [141]. In the study, which involved 26 Italian centres, patients with Eastern Cooperative Oncology Group performance status (ECOG PS) 0–2 and diagnosed with progressive MPM during or after a first-line with platinum and pemetrexed were enrolled. Patients were randomized to a second line with gemcitabine 1000 mg/m2 on days 1 and 8 of every 21 plus placebo (in the control group) or ramucirumab (an anti-VEGFR2 antibody) at 10 mg/m2 on day 1 of each cycle up to progression or severe toxicity. Central randomization was performed according to a minimization algorithm, using the following stratification factors: ECOG PS, age, histology and time to first-line progression (>/<6 months) [141]. The primary endpoint of the study was OS. Secondary aims were progression-free survival, objective response rate, disease control rate, drug safety, patient quality of life and the possible presence of predictive markers. It was planned to enroll 156 patients to obtain a power of 80% assuming a benefit of the experimental treatment of 13% at 1 year compared to the standard arm. From December 2016 to July 2018, 165 patients were enrolled, of whom 161 were correctly assigned and received treatment, 83 in the placebo arm and 81 in the experimental arm. The median age was 69, with about 40% over seventy in both arms. 74% of patients were male and 99% had an ECOG PS of 0–1 [141]. The database was closed in March 2020 and, after a median follow-up of 21.9 months, the observed OS was higher in the experimental arm (Hazard Ratio 0.71; p = 0.028). Specifically, in the ramucirumab arm, the median survival was 13.8 months versus only 7.5 months in the placebo arm and the one-year probability of survival improved from 33 to 56% with the addition of ramucirumab [141]. Progression-free survival was also higher in the experimental arm (median 6.4 versus 3.3 months), but without reaching statistical significance (p = 0.082). Disease control was achieved in 73% of patients treated in the ramucirumab group versus 52% in the placebo arm [141]. A post hoc analysis showed a duration of response of 8.4 months in the experimental arm versus 5.4 months in the standard arm [141]. The pre-specified analysis of the subgroups shows that the survival advantage was independent of the histological subtype and the time of progression of the tumor to the first line [141]. No unexpected toxicities occurred. Grade 3–4 adverse events were recorded in 44% of patients treated with gemcitabine + ramucirumab compared to 30% in the gemcitabine + placebo arm. In particular, the most frequent severe toxicities were neutropenia (20% and 12% in the experimental and standard arms, respectively), arterial hypertension (6% with ramucirumab, 0 with placebo) and fatigue (5% and 4% respectively) [141]. The authors conclude that the association between ramucirumab and gemcitabine significantly improved the OS of patients with progressive MPM following standard first-line CT, with a favorable safety profile. It is clear, however, that in light of the new standard of treatment with the immunotherapy brace nivolumab + ipilimumab, the scheme proposed by the RAMES study [141] is part of a therapeutic context that has changed from the one that had seen the start of the study: in particular, the scheme gemcitabine—ramucirumab has not been tested in patients who have received the combination of the 2 immunotherapies. In addition, the treatment landscape of pleural mesothelioma may still change, pending the results of randomized trials, following interesting phase 2 data for first-line chemo-immunotherapy (NCT02899195 and NCT04334759). Taking this into account, ramucirumab, given its action not only on cancer cells but also on the immune infiltrate and tumor microenvironment, could continue to play a role in patients progressing to chemo-immunotherapy, but further studies will need to be conducted.

Another important drug is the CTLA-4 inhibitor tremelimumab which has been studied in a randomized phase II trial (DETERMINE) in 571 patients with a median survival of 7.7 months compared with patients who underwent placebo with 7.3 months of survival: although its safety profile was consistent, tremelimumab did not significantly improve OS [142].

3.3 T-cell therapies

Adoptive T-cell therapy seems to be promising in MPM and this has been highlighted in a recent phase I trial studying the chimeric antigen receptor (CAR) T-cell therapy targeting mesothelin in MPM patients. Of 18 patients treated with a single dose of CD28-costimulated MSLN CAR-T cells plus the I-caspase-9 safety gene treated intrapleurally showed that in 14 patients, 2 had complete remission, 5 partial, and 4 patients showed the stability of the disease [143]. In the context of safety, no CAR-T cell-related toxicities have been noted. The most promising approach seems to be represented by the combination between CAR-T cells and anti- programmed death-1 (PD-1) therapy [144], although these considerations need to be further investigated.

3.4 Vaccines

One of the most considered new therapies against MPM is vaccine therapy. In particular, the Wilms tumor-1 (WT1) protein in MPM is highly expressed and it is considered a future target for the setting of a cancer vaccine. A recent phase II randomized trial evaluated a WT1 analogue peptide vaccine associated with immunologic adjuvants, showing an improved survival at 1 year (45%) compared with the control group with a 33% of survival rate. However, the OS for vaccinated patients was 22.8 months compared with patients not treated (18.3 months) [145].

Other new target agents as dendritic cells (DC) therapy started to be recently considered to investigate the anti-tumor immune response [146, 147].

3.5 Other investigational therapies

Besides the vaccines and the T-cells therapies which are coming up for the better control of MPM, the oncolytic viruses and other targets as some vascular endothelial growth factors receptors (VEGFR), the platelet-derived growth factor receptor (PDGFR), and the fibroblast growth factor receptor (FGFR) tyrosine-kinase inhibitor, are under consideration. Some phase I/II clinical trials are running at the moment to evaluate the safety of the biological effects [148, 149, 150, 151, 152].

Advertisement

4. Conclusions

In conclusion, the optimal treatment of MPM is still a matter of debate. Surgery, CT, and RT have failed as single modality therapies with no effects on patients’ survival. It is evident that optimal multidisciplinary care is fundamental in the management of MPM patients, as the best results are obtained when patients manage to undergo more complex treatment protocols, often consisting in trimodality approaches. New prospective studies are needed to provide high-quality evidence on the field. While it is reasonable to assume that surgery will remain a central component in the multimodality treatment of MPM, a great development is expected from novel treatment modalities such as targeted therapies, T-cell therapy, vaccines and other investigational therapies, leading to the possibility of prolonged control of the disease, increased survival rates, and better quality of life.

References

  1. 1. National Comprehensive Cancer Network (NCCN). Malignant Pleural Mesothelioma Guidelines Version 2.2021—February 16, 2021 [Internet]. 2021. Available from: https://www.nccn.org/professionals/physician_gls/pdf/mpm.pdf [Accessed: November 01, 2021]
  2. 2. Bueno R, Stawiski EW, Goldstein LD. Comprehensive genomic analysis of malignant pleural mesothelioma identifies recurrent mutations, gene fusions and splicing alterations. Nature Genetics. 2016;48:407-416. DOI: 0.1038/ng.3520
  3. 3. Yap TA, Aerts JG, Popat S. Novel insights into mesothelioma biology and implications for therapy. Nature Reviews Cancer. 2017;17:475-488. DOI: 10.1038/nrc.2017.42
  4. 4. Scherpereel A, Wallyn F, Albelda S. Novel medical therapies for malignant pleural mesothelioma: A review. The Lancet Oncology. 2018;19:e161-e172. DOI: 10.1016/S1470-2045(18)30100-1
  5. 5. Woolhouse I, Bishop L, Darlison L. British Thoracic Society Guideline for the investigation and management of malignant pleural mesothelioma. Thorax. 2018;73(Suppl.1):i1-i30. DOI: 10.1136/thoraxjnl-2017-211321
  6. 6. Kindler HL, Ismaila N, Armato SG 3rd. Treatment of malignant pleural mesothelioma: American Society of Clinical Oncology clinical practice guideline. Journal of Clinical Oncology. 2018;36:1343-1373. DOI: 10.1200/JCO.2017.76.6394
  7. 7. Baas P, Fennell D, Kerr KM. Malignant pleural mesothelioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of Oncology. 2015;26(Suppl. 5):v31-v39. DOI: 10.1093/annonc/mdv199
  8. 8. Scherpereel A, Opitz I, Berghmans T. ERS/ESTS/EACTS/ESTRO guidelines for the management of malignant pleural mesothelioma. The European Respiratory Journal. 2020;55:1900953. DOI: 10.1183/13993003.00953-2019
  9. 9. Sugarbaker DJ. Macroscopic complete resection: The goal of primary surgery in multimodality therapy for pleural mesothelioma. Journal of Thoracic Oncology. 2006;1:175-176. DOI: 10.1016/S1556-0864(15)31536-7
  10. 10. Treasure T. What is the best approach for surgery of malignant pleural mesothelioma? It is to put our efforts into obtaining trustworthy evidence for practice. The Journal of Thoracic and Cardiovascular Surgery. 2016;151:307-309. DOI: 10.1016/j.jtcvs.2015.09.086
  11. 11. Rusch V, Baldini EH, Bueono R. The role of cytoreduction in the treatment of malignant pleural mesothelioma: Meeting summary of the International Mesothelioma Interest Group Congress, September 11–14, 2012, Boston, MA. Journal of Thoracic Cardiovascular Surgery. 2013;145:909-910. DOI: 10.1016/j.jtcvs.2013.01.039
  12. 12. Sugarbaker DJ, Flores RM, Jaklitsch MT. Resection margins, extrapleural nodal status and cell type determine post-operative long term survival in trimodality therapy of malignant pleural mesothelioma: Results in 183 patients. The Journal of Thoracic and Cardiovascular Surgery. 1999;117:54-63. DOI: 10.1016/s0022-5223(99)70469-1
  13. 13. Marulli G, Faccioli E, Bellini A. Induction chemotherapy vs post-operative adjuvant therapy for malignant pleural mesothelioma. Expert Review of Respiratory Medicine. 2017;11(8):649-660. DOI: 10.1080/17476348.2017.1338951
  14. 14. Shohdy K, Abdel-Rahman O. The timing of chemotherapy in the management plan for medically operable early-stage malignant pleural mesothelioma. Expert Review of Respiratory Medicine. 2019;13(6):579-584. DOI: 10.1080/17476348.2019.1607300
  15. 15. Burt MB, Lee HS, Raghuram AC. Preoperative prediction of unresectability in malignant pleural mesothelioma. The Journal of Thoracic and Cardiovascular Surgery. 2020;159:2512-2520. DOI: 10.1016/j.jtcvs.2019.11.035
  16. 16. Marulli G, Breda C, Fontana P. Pleurectomy-decortication in malignant pleural mesothelioma: Are different surgical techniques associated with different outcomes? Results from a multicentre study. European Journal of Cardio-Thoracic Surgery. 2017;52:63-69. DOI: 10.1093/ejcts/ezx079
  17. 17. Rusch VW, Giroux D, Kennedy C. Initial analysis of the international association for the study of lung cancer mesothelioma database. Journal of Thoracic Oncology. 2012;7:1631-1639. DOI: 10.1097/JTO.0b013e31826915f1
  18. 18. Lang-Lazdunski L, Bille A, Papa S. Pleurectomy/decortication, hyperthermic pleural lavage with povidone-iodine, prophylactic radiotherapy and systemic chemotherapy in patients with malignant pleural mesothelioma: A 10-year experience. The Journal of Thoracic and Cardiovascular Surgery. 2015;149:558-566. DOI: 10.1016/j.jtcvs.2014.10.041
  19. 19. Giles AE, Kidane B. Commentary: Know your enemy–understanding futility in the battle against malignant pleural mesothelioma. The Journal of Thoracic and Cardiovascular Surgery. 2020;159:2523-2524. DOI: 10.1016/j.jtcvs.2020.01.034
  20. 20. Bellini A, Dell’Amore A, Giraudo C. Computed tomography and spirometry can predict unresectability in malignant pleural mesothelioma. Journal of Clinical Medicine. 2021;10:4407. DOI: 10.3390/jcm10194407
  21. 21. Marulli G, Rea F, Nicotra S. Effect of induction chemotherapy on lung function and exercise capacity in patients affected by malignant pleural mesothelioma. European Journal of Cardio-Thoracic Surgery. 2010;37:1464-1469. DOI: 10.1016/j.ejcts.2010.01.008
  22. 22. Marulli G, Di Chiara F, Braccioni F. Changes in pulmonary function tests predict radiological response to chemotherapy in malignant pleural mesothelioma. European Journal of Cardio-Thoracic Surgery. 2013;44:104-110. DOI: 10.1093/ejcts/ezs624
  23. 23. Sensakovic WF, Armato SG III, Straus C. Computerized segmentation and measurement of malignant pleural mesothelioma. Medical Physics. 2011;38:238-244. DOI: 10.1118/1.3525836
  24. 24. Armato SG III, Nowak AK. Revised modified RECIST criteria for the assessment of response in malignant pleural mesothelioma (version 1.1). Journal of Thoracic Oncology. 2018;13:1012-1021. DOI: 10.1016/j.jtho.2018.04.034
  25. 25. Armato SG III, Sensakovic WF. Automated lung segmentation for thoracic CT: Impact on computer-aided diagnosis. Academic Radiology. 2004;11:1011-1021. DOI: 10.1016/j.acra.2004.06.005
  26. 26. Labby EZ, Armato SG III, Dignam JJ. Lung volume measurements as surrogate marker for patient response in malingant pleural mesothelioma. Journal of Thoracic Oncology. 2013;8:478-486. DOI: 10.1097/JTO.0b013e31828354c8
  27. 27. Hashimoto M, Takeuchi J, Teruhisa T. Pleural thickness after neoadjuvant chemotherapy is a prognostic factor in malignant pleural mesothelioma. The Journal of Thoracic and Cardiovascular Surgery. 2019;157:404-413. DOI: 10.1016/j.jtcvs.2018.09.106
  28. 28. De Perrot M. Commentary: Is thoracic cage volume a newparameter for clinical staging inmesothelioma? The Journal of Thoracic and Cardiovascular Surgery. 2020;159:2520-2521. DOI: 10.1016/j.jtcvs.2019.12.048
  29. 29. Nowak AK, Chansky K, Rice DC. The IASLC Mesothelioma Staging Project: Proposals for revisions of the T descriptors in the forthcoming eighth editions of the TNM classification for mesothelioma. Journal of Thoracic Oncology. 2016;11:2089-2099. DOI: 10.1016/j.jtho.2016.08.147
  30. 30. Waller DA, Opitz I, Bueno R. Divided by an ocean of water but united in an ocean of uncertainty: A transatlantic review of mesothelioma surgery guidelines. The Journal of Thoracic and Cardiovascular Surgery. 2021;161:1922-1925. DOI: 10.1093/ejcts/ezaa363
  31. 31. Kim S, Bull DA, Garland L. Is there a role for cancer-directed surgery in early-stage sarcomatoid or biphasic mesothelioma? The Annals of Thoracic Surgery. 2019;107:194-201. DOI: 10.1016/j.athoracsur.2018.07.081
  32. 32. Lococo F, Torricelli F, Lang-Lazdunski L. Survival results in biphasic malignant pleural mesothelioma patients: A multicentric analysis. The Journal of Thoracic and Cardiovascular Surgery. 2020;159(4):1584-1593. DOI: 10.1016/j.jtcvs.2019.08.027
  33. 33. Harling L, Kolokotroni SM, Nair A. Differential survival characteristics of sarcomatoid subtype in biphasic pleural mesothelioma. The Annals of Thoracic Surgery. 2019;107(3):929-935. DOI: 10.1016/j.athoracsur.2018.09.019
  34. 34. Rice D, Chansky K, Nowak A. The IASLC Mesothelioma Staging Project: Proposals for revisions of the N descriptors in the forthcoming eighth edition of the TNM classification for pleural mesothelioma. Journal of Thoracic Oncology. 2016;11:2100-2111. DOI: 10.1016/j.jtho.2016.09.121
  35. 35. Sugarbaker DJ, Jaklitsch MT, Bueno R. Prevention, early detection, and management of complications after 328 consecutive extrapleural pneumonectomies. The Journal of Thoracic and Cardiovascular Surgery. 2004;128:138-146. DOI: 10.1016/j.jtcvs.2004.02.021
  36. 36. Martini N, Bains MS, Beattie EJ Jr. Indications for pleurectomy in malignant effusion. Cancer. 1975;35:734-738. DOI: 10.1002/1097-0142(197503)35:3<734::aid-cncr2820350328>3.0.co;2-n
  37. 37. Cao C, Tian DH, Pataky KA. Systematic review of pleurectomy in the treatment of malignant pleural mesothelioma. Lung Cancer. 2013;81:39-27. DOI: 10.1016/j.lungcan.2013.04.024
  38. 38. Rice D, Rusch V, Pass H. Recommendation for uniform definitions of surgical techniques for malignant pleural mesothelioma. A consensus report of the International Association for the Study of Lung Cancer International Staging Committee and the International Mesothelioma Interest Group. Journal of Thoracic Oncology. 2011;6:1304-1312. DOI: 10.1097/JTO.0b013e3182208e3f
  39. 39. Bertoglio P, Garelli E, Brandolini J. Surgical management and reconstruction of diaphragm, pericardium and chest wall in mesothelioma surgery: A review. Journal of Clinical Medicine. 2021;10:2330. DOI: 10.3390/jcm10112330
  40. 40. Butchart EG, Ashcroft T, Barnsley WC. Pleuropneumonectomy in the management of diffuse pleural mesothelioma of the pleura. Experience with 29 patients. Thorax. 1976;31:15-24. DOI: 10.1136/thx.31.1.15
  41. 41. Weder W, Stahel RA, Bernhard J. Multicenter trial of neoadjuvant chemotherapy followed by extrapleural pneumonectomy in malignant pleural mesothelioma. Annals of Oncology. 2007;18:1196-1202. DOI: 10.1093/annonc/mdm093
  42. 42. Treasure T, Internullo E, Fiorentino F. A survey of opinions and beliefs concerning surgery for malignant pleural mesothelioma amongst 802 members of the European Association for Cardio Thoracic Surgery (EACTS), the European Society of Thoracic Surgeons (ESTS) and the society of Thoracic Surgeons (STS). Interactive Cardiovascular and Thoracic Surgery. 2011;12:341-346. DOI: 10.1510/icvts.2010.251488
  43. 43. Cho BC, Donahoe L, Bradbury PD. Surgery for malignant pleural mesothelioma after radiotherapy (SMART): Final results from a single-centre, phase 2 trial. The Lancet Oncology. 2021;22:190-197. DOI: 10.1016/S1470-2045(20)30606-9
  44. 44. Treasure T, Lang-lazdunski L, Waller D. Extrapleural pneumonectomy versus no extrapleural pneumonectomy for patients with malignant pleural mesothelioma: Clinical outcomes of the Mesothelioma and Radical Surgery (MARS) randomized feasibility study. The Lancet Oncology. 2011;12:763-772. DOI: 10.1016/S1470-2045(11)70149-8
  45. 45. Weder W, Stahel RA, Baas P. The MARS feasibility trial: Conclusions not supported by data. The Lancet Oncology. 2011;12:1093-1094. DOI: 10.1016/S1470-2045(11)70307-2
  46. 46. Wanebo HJ, Martini N, Melamed MR. Pleural mesothelioma. Cancer. 1976;38:2481-2488. DOI: 10.1002/1097-0142(197612)38:6<2481::aid-cncr2820380637>3.0.co;2-9
  47. 47. Teh E, Fiorentino F, Tan C. A systematic review of lung sparing extirpative surgery for pleural mesothelioma. Journal of the Royal Society of Medicine. 2011;104:69-80. DOI: 10.1258/jrsm.2010.100345
  48. 48. Lim E, Darlison L, Edwards J. Mesothelioma and Radical Surgery 2 (MARS 2): Protocol for a multicentre randomised trial comparing (extended) pleurectomy decortication versus no (extended) pleurectomy decortication for patients with malignant pleural mesothelioma. BMJ Open. 2020;10:e038892
  49. 49. Friedberg JS, Culligan MJ, Mick R. Radical pleurectomy and intraoperative photodynamic therapy for malignant pleural mesothelioma. The Annals of Thoracic Surgery. 2012;93:1658-1667. DOI: 10.1016/j.athoracsur.2012.02.009
  50. 50. Flores RM. The mesothelioma surgery shift. The Journal of Thoracic and Cardiovascular Surgery. 2016;151:485-486. DOI: 10.1016/j.jtcvs.2015.10.054
  51. 51. Flores RM, Pass HI, Seshan VE. Extrapleural pneumonectomy versus pleurectomy/decortication in the surgical management of malignant pleural mesothelioma results in 663 patients. The Journal of Thoracic and Cardiovascular Surgery. 2008;135:620-626. DOI: 10.1016/j.jtcvs.2007.10.054
  52. 52. Faccioli E, Bellini A, Mammana M. Extrapleural pneumonectomies for pleural mesothelioma. Expert Review of Respiratory Medicine. 2020;14(1):67-79. DOI: 10.1080/17476348.2020.1688147
  53. 53. Reeb J, Olland A, Massard G. Extracorporeal life support in thoracic surgery. European Journal of Cardio-Thoracic Surgery. 2018;53(03):489-494. DOI: 10.1093/ejcts/ezx477
  54. 54. Extracorporeal Life Support Organization. ELSO Guidelines for Cardiopulmonary Extracorporeal Life Support, Version 1.4, August 2017 Ann Arbor, MI, USA [Internet]. 2017. Available from: https://www.elso.org [Accessed: November 1, 2021]
  55. 55. Tathineni P, Pandya M, Chaar B. The utility of ECMO in patients with hematologic malignancies: A literature review. Cureus. 2020;12:e9118. DOI: 10.7759/cureus.9118
  56. 56. Dünser M, HasibederW RM. Successful therapy of severe pneumonia-associated ARDS after pneumonectomy with ECMO and steroids. The Annals of Thoracic Surgery. 2004;78(01):335-337. DOI: 10.1016/S0003-4975(03)01264-5
  57. 57. Fica M, Suarez F, Aparicio R. Single site venovenous ECMO as an alternative to invasive ventilation in post-pneumonectomy fistula with acute respiratory failure. European Journal of Cardio-Thoracic Surgery. 2012;41:950-952. DOI: 10.1093/ejcts/ezr103
  58. 58. Hadem J, Schroder F, Winkler T. One day from dyspnea to death: Unsuccessful application of ECMO in toxoplasma myocarditis following bone marrow transplantation. Clinical Research in Cardiology. 2006;95:477-481. DOI: 10.1007/s00392-006-0402-y
  59. 59. Burt BM, Cameron RB, Mollberg NM. Malignant pleural mesothelioma and the Society of Thoracic Surgeons Database: An analysis of surgical morbidity and mortality. The Journal of Thoracic and Cardiovascular Surgery. 2014;148(01):30-35. DOI: 10.1016/j.jtcvs.2014.03.011
  60. 60. Bellini A, Dell’Amore A, Ferrigno P. ECLS for life-threatening complications in mesothelioma’s surgery: Is it worthwhile? The Thoracic and Cardiovascular Surgeon. 2021;69(6):548-550. DOI: 10.1055/s-0041-1724037
  61. 61. Gow KW, Lao OB, Leong T. Extracorporeal life support for adults with malignancy and respiratory or cardiac failure: The Extracorporeal Life Support experience. American Journal of Surgery. 2010;199(05):669-675. DOI: 10.1016/j.amjsurg.2010.01.018
  62. 62. Rintoul RC, Ritchie AJ, Edwards JG. Efficacy and cost of video-assisted thoracoscopic partial pleurectomy versus talc pleurodesis in patients with malignant pleural mesothelioma (MesoVATS): An open-label, randomised, controlled trial. Lancet. 2014;384:1118-1127. DOI: 10.1016/S0140-6736(14)60418-9
  63. 63. Kostron A, Friess M, Crameri O. Relapse pattern and second-line treatment following multimodality treatment for malignant pleural mesothelioma. European Journal of Cardio-Thoracic Surgery. 2016;49:1516-1523. DOI: 10.1093/ejcts/ezv398
  64. 64. Takuwa T, Hashimoto M, Matsumoto S. Post-recurrence chemotherapy for mesothelioma patients undergoing extrapleural pneumonectomy. International Journal of Clinical Oncology. 2017;22:857-864. DOI: 10.1007/s10147-017-1126-x
  65. 65. Kai Y, Tsutani Y, Tsubokawa N. Prolonged post-recurrence survival following pleurectomy/decortication for malignant pleural mesothelioma. Oncology Letters. 2019;17:3607-3614. DOI: 10.3892/ol.2019.9979
  66. 66. Soldera SV, Kavanagh J, Pintilie M. Systemic therapy use and outcomes after relapse from preoperative radiation and extrapleural pneumonectomy for malignant pleural mesothelioma. The Oncologist. 2019;24:e510-e517. DOI: 10.1634/theoncologist.2018-0501
  67. 67. Nakamura A, Takuwa T, Hashimoto M. Clinical outcomes with recurrence after pleurectomy/decortication for malignant pleural mesothelioma. The Annals of Thoracic Surgery. 2020;109:1537-1543. DOI: 10.1016/j.athoracsur.2019.11.048
  68. 68. Politi L, Borzellino G. Second surgery for recurrence of malignant pleural mesothelioma after extrapleural pneumonectomy. The Annals of Thoracic Surgery. 2010;89:207-211. DOI: 10.1016/j.athoracsur.2009.09.028
  69. 69. Okamoto T, Yano T, Haro A. Treatment for recurrence after extrapleural pneumonectomy for malignant pleural mesothelioma: A single institution experience. Thorac Cancer. 2013;4(1):66-70. DOI: 10.1111/j.1759-7714.2012.00138.x
  70. 70. Burt BM, Ali SO, DaSilva MC. Clinical indications and results after chest wall resection for recurrent mesothelioma. The Journal of Thoracic and Cardiovascular Surgery. 2013;146(6):1373-1379. DOI: 10.1016/j.jtcvs.2013.07.012
  71. 71. Bellini A, Dell’Amore A, Terzi S. Relapse patterns and tailored treatments strategies for malignant pleural mesothelioma recurrence after multimodality therapy. Journal of Clinical Medicine. 2021;10(5):1134. DOI: 10.3390/jcm10051134
  72. 72. Baldini EH, Recht A, Strauss GM. Patterns of failure after trimodality therapy for malignant pleural mesothelioma. The Annals of Thoracic Surgery. 1997;63:334-338. DOI: 10.1016/s0003-4975(96)01228-3
  73. 73. Elsayed HH, Hassaballa AS, Ahmed TA. Recurrence of mesothelioma after a macroscopic complete resection procedure: Is a second radical surgery justified? Interactive Cardiovascular and Thoracic Surgery. 2021;32:761-763. DOI: 10.1093/icvts/ivaa338
  74. 74. Cao C, Tian D, Park J. A systematic review and meta-analysis of surgical treatments for malignant pleural mesothelioma. Lung Cancer. 2014;83:240-245. DOI: 10.1016/j.lungcan.2013.11.026
  75. 75. Van Schil PE, Opitz I, Weder W. Multimodal management of malignant pleural mesothelioma: Where are we today? The European Respiratory Journal. Sep 2014;44(3):754-764. DOI: 10.1183/09031936.00207213
  76. 76. Lee DS, Carollo A, Alpert N. VATS pleurectomy decortication is a reasonable alternative for higher risk patients in the management of malignant pleural mesothelioma: An analysis of short-term outcomes. Cancers (Basel). 2021;13(5):1068. DOI: 10.3390/cancers13051068
  77. 77. Opitz I, Weder W. Pleural mesothelioma: Is the surgeon still there? Annals of Oncology. 2018;29(8):1710-1717. DOI: 10.1093/annonc/mdy195
  78. 78. Nelson DB, Rice DC, Mitchell KG. Defining the role of adjuvant radiotherapy for malignant pleural mesothelioma: A propensity-matched landmark analysis of the National Cancer Database. Journal of Thoracic Disease. 2019;11(4):1269-1278. DOI: 10.21037/jtd.2019.04.27
  79. 79. Spaggiari L, Marulli G, Bovolato P. Extrapleural pneumonectomy for malignant mesothelioma: An Italian multicenter retrospective study. The Annals of Thoracic Surgery. 2014;97:1859-1865. DOI: 10.1016/j.athoracsur.2014.01.050
  80. 80. Sugarbaker DJ, Wolf AS. Surgery for malignant pleural mesothelioma. Expert Review of Respiratory Medicine. 2010;4:363-372. DOI: 10.1586/ers.10.35
  81. 81. Flores RM. Pleurectomy decortication for mesothelioma: The procedure of choice when possible. The Journal of Thoracic and Cardiovascular Surgery. 2016;151:310-312. DOI: 10.1016/j.jtcvs.2015.10.036
  82. 82. Lang-Lazdunski L, Bille A, Lal R. Pleurectomy/decortication is superior to extrapleural pneumonectomy in the multimodality management of patients with malignant pleural mesothelioma. Journal of Thoracic Oncology. 2012;7:737-743. DOI: 10.1097/JTO.0b013e31824ab6c5
  83. 83. Batirel HF, Metintas M, Caglar HB. Adoption of pleurectomy and decortication for malignant mesothelioma leads to similar survival as extrapleural pneumonectomy. The Journal of Thoracic and Cardiovascular Surgery. 2016;151:478-484. DOI: 10.1016/j.jtcvs.2015.09.121
  84. 84. Rena O, Casadio C. Extrapleural pneumonectomy for early stage malignant pleural mesothelioma: A harmful procedure. Lung Cancer. 2012;77:151-155. DOI: 10.1016/j.lungcan.2011.12.009
  85. 85. Magouliotis DE, Tasiopoulou VS, Athanassiadi K. Updated meta-analysis of survival after extrapleural pneumonectomy versus pleurectomy/decortication in mesothelioma. General Thoracic and Cardiovascular Surgery. 2019;67:312-320. DOI: 10.1007/s11748-018-1027-6
  86. 86. Vigneswaran WT, Kircheva DY, Rodrigues AE. Influence of pleurectomy and decortication in health-related quality of life among patients with malignant pleural mesothelioma. World Journal of Surgery. 2018;42:1036-1045. DOI: 10.1007/s00268-017-4264-4
  87. 87. Papaspyros S. Surgical management of malignant pleural mesothelioma: Impact of surgery on survival and quality of life—relation to chemotherapy, radiotherapy, and alternative therapies. ISRN Surgery. 2014;2014:817203. DOI: 10.1155/2014/817203
  88. 88. Kostron A, Friess M, Inci I. Propensity matched comparison of extrapleural pneumonectomy and pleurectomy/decortication for mesothelioma patients. Interactive Cardiovascular and Thoracic Surgery. 2017;24:740-746. DOI: 10.1093/icvts/ivw422
  89. 89. Sharkey AJ, Tenconi S. Nakas A The effects of an intentional transition from extrapleural pneumonectomy to extended pleurectomy/decortication. European Journal of Cardio-Thoracic Surgery. 2016;49:1632-1641. DOI: 10.1093/ejcts/ezv403
  90. 90. Bueno R, Opitz I, IASCL Mesothelioma Taskforce. Surgery in malignant pleural mesothelioma. Journal of Thoracic Oncology. 2018;13:1638-1654. DOI: 10.1016/j.jtho.2018.08.001
  91. 91. Datta A, Smith R, Fiorentino F. Surgery in the treatment of malignant pleural mesothelioma: Recruitment into trials should be the default position. Thorax. 2014;69:194-197. DOI: 10.1136/thoraxjnl-2013-203846
  92. 92. Treasure T, Sedrakyan A. Pleural mesothelioma: Little evidence, still time to do trials. Lancet. 2004;364:1183-1185. DOI: 10.1016/S0140-6736(04)17108-0
  93. 93. Fournel L, Janet-Vendroux A, Canny-Hamelin E. Malignant pleural mesothelioma: The role of surgery. Revue de Pneumologie Clinique. 2018;74:351-358. DOI: 10.1016/j.pneumo.2018.09.006
  94. 94. Treasure T, Macbeth F. Fifteen years in the evaluation of extrapleural pneumonectomy: Lessons to be learned. The Journal of Thoracic and Cardiovascular Surgery. 2015;149:1382-1383. DOI: 10.1016/j.jtcvs.2014.10.083
  95. 95. Hiddinga BI, van Meerbeeck JP. Surgery in mesothelioma-where do we go after MARS? Journal of Thoracic Oncology. 2013;8:525-529. DOI: 10.1097/JTO.0b013e31828353d7
  96. 96. van Zandwijk N, Clarke C, Henderson D. Guidelines for the diagnosis and treatment of malignant pleural mesothelioma. Journal of Thoracic Disease. 2013;5:E254. DOI: 10.3978/j.issn.2072-1439.2013.11.28
  97. 97. Scherpereel A, Astoul P, Baas P. Guidelines of the European Respiratory Society and the European Society of Thoracic Surgeons for the management of malignant pleural mesothelioma. The European Respiratory Journal. 2010;35:479-495. DOI: 10.1183/09031936.00063109
  98. 98. Fahrner R, Ochsenbein A, Schmid RA. Long term survival after trimodal therapy in malignant pleural mesothelioma. Swiss Medical Weekly. 2012;142:w13686. DOI: 10.4414/smw.2012.13686
  99. 99. Ceresoli GL, Castagneto B, Zucali PA. Pemetrexed plus carboplatin in elderly patients with malignant pleural mesothelioma: Combined analysis of two phase II trials. British Journal of Cancer. 2008;99:51-56. DOI: 10.1038/sj.bjc.6604442
  100. 100. Katirtzoglou N, Gkiozos I, Makrilia N. Carboplatin plus pemetrexed as first-line treatment of patients with malignant pleural mesothelioma: A Phase II Study. Clinical Lung Cancer. 2010;11:30-35. DOI: 10.3816/CLC.2010.n.005
  101. 101. Ceresoli GL, Zucali PA, Favaretto AG. Phase II Study of pemetrexed plus carboplatin in malignant pleural mesothelioma. Journal of Clinical Oncology. 2006;24:1443-1448. DOI: 10.1200/JCO.2005.04.3190
  102. 102. Zalcman G, Mazieres J, Margery J. Bevacizumab for newly diagnosed pleural mesothelioma in the Mesothelioma Avastin Cisplatin Pemetrexed Study (MAPS): A randomised, controlled, open-label, phase 3 trial. Lancet. 2016;387:1405-1414. DOI: 10.1016/S0140-6736(15)01238-6
  103. 103. Jassem J, Ramlau R, Santoro A. Phase III trial of pemetrexed plus best supportive care compared with best supportive care in previously treated patients with advanced malignant pleural mesothelioma. Journal of Clinical Oncology. 2008;26:1698-1704. DOI: 10.1200/JCO.2006.09.9887
  104. 104. Muers MF, Stephens RJ, Fisher P. Active symptom control with or without chemotherapy in the treatment of patients with malignant pleural mesothelioma (MS01): A multicentre randomised trial. Lancet. 2008;371:1685-1694. DOI: 10.1016/S0140-6736(08)60727-8
  105. 105. Richards WG, Zellos L, Bueno R. Phase I to II study of pleurectomy/decortication and intraoperative intracavitary hyperthermic cisplatin lavage for mesothelioma. Journal of Clinical Oncology. 2006;24:1561-1567. DOI: 10.1200/JCO.2005.04.6813
  106. 106. Gomez D, Tsao AS. Local and systemic therapies for malignant pleural mesothelioma. Current Treatment Options in Oncology. 2014;15:683-699. DOI: 10.1007/s11864-014-0314-4
  107. 107. Burt BM, Richards WG, Lee HS. A Phase I Trial of surgical resection and intraoperative hyperthermic cisplatin and gemcitabine for pleural mesothelioma. Journal of Thoracic Oncology. 2018;13:1400-1409. DOI: 10.1016/j.jtho.2018.04.032
  108. 108. Bertoglio P, Aprile V, Ambrogi MC. The role of intracavitary therapies in the treatment of malignant pleural mesothelioma. Journal of Thoracic Disease. 2018;10:S293-S297. DOI: 10.21037/jtd.2017.10.165
  109. 109. Opitz I, Lauk O, Meerang M. Intracavitary cisplatin-fibrin chemotherapy after surgery for malignant pleural mesothelioma: A phase I trial. The Journal of Thoracic and Cardiovascular Surgery. 2020;159:330-340. DOI: 10.1016/j.jtcvs.2019.07.073
  110. 110. Okabe K. Intraoperative intracavitary hyperthermic chemotherapy for malignant pleural mesothelioma. Annals of Translational Medicine. 2017;5:233. DOI: 10.21037/atm.2017.03.78
  111. 111. Ried M, Potzger T, Braune N. Local and systemic exposure of cisplatin during hyperthermic intrathoracic chemotherapy perfusion after pleurectomy and decortication for treatment of pleural malignancies. Journal of Surgical Oncology. 2013;107:735-740. DOI: 10.1002/jso.23321
  112. 112. Ishibashi H, Kobayashi M, Takasaki C. Interim results of pleurectomy/decortication and intraoperative intrapleural hyperthermic cisplatin perfusion for patients with malignant pleural mesothelioma intolerable to extrapleural pneumonectomy. General Thoracic and Cardiovascular Surgery. 2015;63:395-400. DOI: 10.1007/s11748-015-0535-x
  113. 113. Sugarbaker DJ, Gill RR, Yeap BY. Hyperthermic intraoperative pleural cisplatin chemotherapy extends interval to recurrence and survival among low-risk patients with malignant pleural mesothelioma undergoing surgical macroscopic complete resection. The Journal of Thoracic and Cardiovascular Surgery. 2013;145:955-963. DOI: 10.1016/j.jtcvs.2012.12.037
  114. 114. Rusch VW, Rosenzweig K, Venkatraman E. A phase II trial of surgical resection and adjuvant high-dose hemithoracic radiation for malignant pleural mesothelioma. The Journal of Thoracic and Cardiovascular Surgery. 2001;122:788-795. DOI: 10.1067/mtc.2001.116560
  115. 115. Buduhan G, Menon S, Aye R. Trimodality therapy for malignant pleural mesothelioma. The Annals of Thoracic Surgery. 2009;88:870-875. DOI: 10.1016/j.athoracsur.2009.05.036
  116. 116. de Perrot M, Feld R, Cho BC. Trimodality therapy with induction chemotherapy followed by extrapleural pneumonectomy and adjuvant high-dose hemithoracic radiation for malignant pleural mesothelioma. Journal of Clinical Oncology. 2009;27:1413-1418. DOI: 10.1200/JCO.2008.17.5604
  117. 117. Kapeles M, Gensheimer MF, Mart DA. Trimodality treatment of malignant pleural mesothelioma: An institutional review. American Journal of Clinical Oncology. 2018;41:30-35. DOI: 10.1097/COC.0000000000000225
  118. 118. Thieke C, Nicolay NH, Sterzing F. Long-term results in malignant pleural mesothelioma treated with neoadjuvant chemotherapy, extrapleural pneumonectomy and intensity-modulated radiotherapy. Radiation Oncology. 2015;10:267. DOI: 10.1186/s13014-015-0575-5
  119. 119. Mencoboni M, Filiberti RA, Taveggia P. Clinical features and treatment outcome of malignant pleural mesothelioma. Oncology Research and Treatment. 2017;40:364-369. DOI: 10.1159/000464410
  120. 120. Rea F, Marulli G, Bortolotti L. Induction chemotherapy, extrapleural pneumonectomy (EPP) and adjuvant hemi-thoracic radiation in malignant pleural mesothelioma (MPM): Feasibility and results. Lung Cancer. 2007;57:89-95. DOI: 10.1016/j.lungcan.2007.02.004
  121. 121. Van Schil PE, Baas P, Gaafar R. Trimodality therapy for malignant pleural mesothelioma: Results from an EORTC phase II multicentre trial. The European Respiratory Journal. 2010;36:1362-1369. DOI: 10.1183/09031936.00039510
  122. 122. Krug LM, Pass HI, Rusch VW. Multicenter phase II trial of neoadjuvant pemetrexed plus cisplatin followed by extrapleural pneumonectomy and radiation for malignant pleural mesothelioma. Journal of Clinical Oncology. 2009;27:3007-2013. DOI: 10.1200/JCO.2008.20.3943
  123. 123. Gomez DR, Rimner A, Simone CB 2nd. The use of radiation therapy for the treatment of malignant pleural mesothelioma: Expert Opinion from the National Cancer Institute Thoracic Malignancy Steering Committee, International Association for the Study of Lung Cancer, and Mesothelioma Applied Research Foundation. Journal of Thoracic Oncology. 2019;14:1172-1183. DOI: 10.1016/j.jtho.2019.03.030
  124. 124. Rice DC, Stevens CW, Correa AM. Outcomes after extrapleural pneumonectomy and intensity-modulated radiation therapy for malignant pleural mesothelioma. The Annals of Thoracic Surgery. 2007;84:1685-1692. DOI: 10.1016/j.athoracsur.2007.04.076
  125. 125. Gomez DR, Hong DS, Allen PK. Patterns of failure, toxicity, and survival after extrapleural pneumonectomy and hemithoracic intensity-modulated radiation therapy for malignant pleural mesothelioma. Journal of Thoracic Oncology. 2013;8:238-245. DOI: 10.1097/JTO.0b013e31827740f0
  126. 126. Rimner A, Zauderer MG, Gomez DR. Phase II study of hemithoracic intensity-modulated pleural radiation therapy (IMPRINT) as part of lung-sparing multimodality therapy in patients with malignant pleural mesothelioma. Journal of Clinical Oncology. 2016;34:2761. DOI: 10.1200/JCO.2016.67.2675
  127. 127. Shaikh F, Zauderer MG, von Reibnitz D. Improved outcomes with modern lung-sparing trimodality therapy in patients with malignant pleural mesothelioma. Journal of Thoracic Oncology. 2017;12:993-1000. DOI: 10.1016/j.jtho.2017.02.026
  128. 128. Espinoza-Mercado F, Borgella JD, Berz D. Disparities in compliance with national guidelines for the treatment of malignant pleural mesothelioma. The Annals of Thoracic Surgery. 2019;108:889-896. DOI: 10.1016/j.athoracsur.2019.03.052
  129. 129. Ahamad A, Stevens CW, Smythe WR. Promising early local control of malignant pleural mesothelioma following postoperative intensity modulated radiotherapy (IMRT) to the chest. Cancer Journal. 2003;9:476-484. DOI: 10.1097/00130404-200311000-00008
  130. 130. Vogelzang NJ, Rusthoven JJ, Symanowski J. Phase III study of pemetrexed in combination with cisplatin versus cisplatin alone in patients with malignant pleural mesothelioma. Journal of Clinical Oncology. 2003;21:2636-2644. DOI: 10.1200/JCO.2003.11.136
  131. 131. Sandin LC, Eriksson F, Ellmark P. Local CTLA4 blockade effectively restrains experimental pancreatic adenocarcinoma growth in vivo. Oncoimmunology. 2014;3:e27614. DOI: 10.4161/onci.27614
  132. 132. Fransen MF, van der Sluis TC, Ossendorp F. Controlled local delivery of CTLA-4 blocking antibody induces CD8+ T-cell-dependent tumor eradication and decreases risk of toxic side effects. Clinical Cancer Research. 2013;19:5381-5389. DOI: 10.1158/1078-0432.CCR-12-0781
  133. 133. Popat S, Curioni-Fontecedro A, Polydoropoulou V. A multicentre randomized phase III trial comparing pembrolizumab (P) vs single agent chemotherapy (CT) for advanced pre-treated malignant pleural mesothelioma (MPM): Results from the European Thoracic Oncology Platform (ETOP 9-15) PROMISE-meso trial. Annals of Oncology. 2020;31(12):1734-1745. DOI: 10.1016/j.annonc.2020.09.009
  134. 134. Scherpereel A, Mazieres J, Greillier L. Nivolumab or nivolumab plus ipilimumab in patients with relapsed malignant pleural mesothelioma (IFCT-1501 MAPS2): A multicentre, open-label, randomised, non-comparative, phase 2 trial. The Lancet Oncology. 2019;20:239-253. DOI: 10.1016/S1470-2045(18)30765-4
  135. 135. Menis J, Pasello G, Remon J. Immunotherapy in malignant pleural mesothelioma: A review of literature data. Transl Lung Cancer Res. 2021;10(6):2988-3000. DOI: 10.21037/tlcr-20-673
  136. 136. Baas P, Scherpereel A, Nowak AK. First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): A multicentre, randomised, open-label, phase 3 trial. Lancet. 2021;397(10272):375-386. DOI: 10.1016/S0140-6736(20)32714-8
  137. 137. CheckpOiNt Blockade For Inhibition of Relapsed Mesothelioma (CONFIRM) [Internet]. 2017. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03063450 [Accessed: November 01, 2021]
  138. 138. Nowak AK, Brosseau S, Cook A. Antiangiogeneic strategies in mesothelioma. Frontier in Oncologia. 2020;10:126. DOI: 10.3389/fonc.2020.00126
  139. 139. Aprile G, Rijavec E, Fontanella C. Ramucirumab: Preclinical research and clinical development. Oncotargets and Therapy. 2014;7:1997-2006. DOI: 10.2147/OTT.S61132
  140. 140. Désage AL, Karpathiou G, Peoc’h M. The immune microenvironment of malignant pleural mesothelioma: A literature review. Cancers (Basel). 2021;13(13):3205. DOI: 10.3390/cancers13133205
  141. 141. Pinto C, Zucali PA, Pagano M. Gemcitabine with or without ramucirumab as second-line treatment for malignant pleural mesothelioma (RAMES): A randomised, double-blind, placebo-controlled, phase 2 trial. The Lancet Oncology. 2021;22(10):1438-1447. DOI: 10.1016/S1470-2045(21)00404-6
  142. 142. Maio M, Scherpereel A, Calabro L. Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): A multicentre, international, randomised, double-blind, placebo-controlled phase 2b trial. The Lancet Oncology. 2017;18:1261-1273. DOI: 10.1016/S1470-2045(17)30446-1
  143. 143. Adusumilli PS, Zauderer MG, Rusch VW. Regional delivery of mesothelin-targeted CAR T cells for pleural cancers: Safety and preliminary efficacy in combination with anti-PD-1 agent. Journal of Clinical Oncology. 2019;37:2511. DOI: 10.1200/JCO.2019.37.15_suppl.2511
  144. 144. Chong EA, Svoboda J, Dwivedy NS. Sequential anti-CD19 directed chimeric antigen receptor modified T-cell therapy (CART19) and PD-1 blockade with pembrolizumab in patients with relapsed or refractory B-cell non-Hodgkin Lymphomas. Blood. 2018;132:4198. DOI: 10.3389/fonc.2019.00767
  145. 145. Zauderer MG, Tsao AS, Dao T. A Randomized Phase II Trial of Adjuvant Galinpepimut-S, WT-1 analogue peptide vaccine, after multimodality therapy for patients with malignant pleural mesothelioma. Clinical Cancer Research. 2017;23:7483-7489. DOI: 10.1158/1078-0432.CCR-17-2169
  146. 146. Gabrilovich DI, Ciernik IF, Carbone DP. Dendritic cells in antitumor immune responses. I. Defective antigen presentation in tumor-bearing hosts. Cellular Immunology. 1996;170:101-110. DOI: 10.1006/cimm.1996.0139
  147. 147. Enk AH, Jonuleit H, Saloga J. Dendritic cells as mediators of tumor-induced tolerance in metastatic melanoma. International Journal of Cancer. 1997;73:309-316. DOI: 10.1002/(sici)1097-0215(19971104)73:3<309::aid-ijc1>3.0.co;2-3
  148. 148. Muggerud AA, Hallett M, Johnsen H. Molecular diversity in ductal carcinoma in situ (DCIS) and early invasive breast cancer. Molecular Oncology. 2010;4:357-368. DOI: 10.1016/j.molonc.2010.06.007
  149. 149. Grosso F, Steele N, Novello S. Nintedanib plus pemetrexed/cisplatin in patients with malignant pleural mesothelioma: Phase II results from the Randomized, Placebo-Controlled LUME-Meso Trial. Journal of Clinical Oncology. 2017;35:3591-3600. DOI: 10.1200/JCO.2017.72.9012
  150. 150. Scagliotti GV, Gaafar R, Nowak AK. Nintedanib in combination with pemetrexed and cisplatin for chemotherapy-naive patients with advanced malignant pleural mesothelioma (LUME-Meso): A double-blind, randomised, placebo-controlled phase 3 trial. The Lancet Respiratory Medicine. 2019;7:569-580. DOI: 10.1016/S2213-2600(19)30139-0
  151. 151. Kelly WK, O’Connor OA, Krug LM. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. Journal of Clinical Oncology. 2005;23:3923-3931. DOI: 10.1200/JCO.2005.14.167
  152. 152. Krug LM, Kindler HL, Calvert H. Vorinostat in patients with advanced malignant pleural mesothelioma who have progressed on previous chemotherapy (VANTAGE-014): A phase 3, double-blind, randomised, placebo-controlled trial. The Lancet Oncology. 2015;16:447-456. DOI: 10.1016/S1470-2045(15)70056-2

Written By

Alice Bellini, Beatrice Aramini and Franco Stella

Submitted: 30 November 2021 Reviewed: 14 February 2022 Published: 20 June 2022