Next Article in Journal
tRNA-Derived Fragments as Biomarkers in Bladder Cancer
Previous Article in Journal
Th2 Cells Are Associated with Tumor Recurrence Following Radiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Multimodal Treatment of Pleural Mesothelioma with Cytoreductive Surgery and Hyperthermic Intrathoracic Chemotherapy: Impact of Additive Chemotherapy

1
Department of Thoracic Surgery, Thoraxklinik, University Hospital Heidelberg, 69126 Heidelberg, Germany
2
German Center for Lung Research (DZL), 69120 Heidelberg, Germany
3
Division of Thoracic Surgery, Ludwig-Maximilians-University Munich, Asklepios Lung Clinic Gauting, 82131 Gauting, Germany
4
Center for Clinical Studies, University Hospital Regensburg, 93053 Regensburg, Germany
5
Department of Thoracic Surgery, University Hospital Freiburg, 79106 Freiburg im Breisgau, Germany
6
Department of Thoracic Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
7
Department of Thoracic Surgery, Barmherzige Brüder Hospital Regensburg, 93049 Regensburg, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2024, 16(8), 1587; https://doi.org/10.3390/cancers16081587
Submission received: 25 March 2024 / Revised: 11 April 2024 / Accepted: 16 April 2024 / Published: 20 April 2024
(This article belongs to the Section Clinical Research of Cancer)

Abstract

:

Simple Summary

Cytoreductive surgery combined with intraoperative hyperthermic chemoperfusion (HITOC) within a multimodal treatment approach has a positive impact on the survival of highly selected patients with epithelioid pleural mesothelioma. The addition of chemotherapy significantly affects the interval to tumor recurrence.

Abstract

Cytoreductive surgery (CRS) combined with hyperthermic intrathoracic chemoperfusion (HITOC) is a promising treatment strategy for pleural mesothelioma (PM). The aim of this study was to evaluate the impacts of this multimodal approach in combination with systemic treatment on disease-free survival (DFS) and overall survival (OS). In this retrospective multicenter study, clinical data from patients after CRS and HITOC for PM at four high-volume thoracic surgery departments in Germany were analyzed. A total of 260 patients with MPM (220 epithelioid, 40 non-epithelioid) underwent CRS and HITOC as part of a multimodal treatment approach. HITOC was administered with cisplatin alone (58.5%) or cisplatin and doxorubicin (41.5%). In addition, 52.1% of patients received neoadjuvant and/or adjuvant chemotherapy. The median follow-up was 48 months (IQR = 38 to 58 months). In-hospital mortality was 3.5%. Both the resection status (macroscopic complete vs. incomplete resection) and histologic subtype (epithelioid vs. non-epithelioid) had significant impacts on DFS and OS. In addition, adjuvant chemotherapy (neoadjuvant/adjuvant) significantly increased DFS (p = 0.003). CRS and HITOC within a multimodal treatment approach had positive impacts on the survival of patients with epithelioid PM after macroscopic complete resection. The addition of chemotherapy significantly prolonged the time to tumor recurrence or progression.

Graphical Abstract

1. Introduction

A rare and highly aggressive malignancy, malignant pleural mesothelioma (PM), is located in the pleural cavity and is primarily associated with asbestos exposure [1]. The widespread use of asbestos over the past decades coupled with a long latency period to disease onset are responsible for a continuously relevant incidence of PM [2]. Although immunotherapy has recently been introduced as a first-line treatment, breakthrough improvements in overall survival are still lacking [3,4]. In addition, there is a lack of data on multimodal treatment concepts that include cytoreductive surgery combined with intracavitary therapies and chemotherapy.
Cytoreductive surgery (CRS), defined as pleurectomy/decortication (PD) or extended pleurectomy/decortication (ePD) with resection of diaphragmatic or pericardial structures or extrapleural pneumonectomy (EPP), within a multimodal treatment approach allows the prolongation of overall survival. However, local tumor recurrence often occurs within a short period of time due to microscopic tumor deposits remaining in the chest, as there is no safety margin and, at best, macroscopic complete resection (MCR) can be achieved by CRS [5,6]. Recent studies have demonstrated a clear advantage of lung-sparing cytoreduction within a multimodal treatment approach over EPP in terms of overall survival [7]. The introduction of hyperthermic intrathoracic chemotherapy (HITOC) after CRS aims to improve local tumor control and prolong recurrence-free and overall survival [8]. In addition, the effect of adjuvant chemotherapy within a multimodal treatment approach may have an impact on residual tumor cells.
Due to the rarity of PM and delayed diagnosis at advanced stages, there is still no standardized multimodal treatment approach for localized disease [9,10]. Few studies have been published on multimodal treatment with CRS, HITOC, and neoadjuvant/adjuvant chemotherapy in patients with PM [7,8,11,12,13]. We postulated the study hypothesis that patients with PM may particularly benefit from complete multimodal therapy including tumor resection and intracavitary and systemic chemotherapy.
The aim of this retrospective multicenter study was to evaluate the impacts of CRS and HITOC within a multimodal treatment concept for patients with PM on disease-free survival (DFS) and overall survival (OS) and, in particular, to assess additional chemotherapy and relevant prognostic factors.

2. Patients and Methods

2.1. Patient Selection

In this retrospective multicenter study, clinical outcomes of 260 patients with a primary diagnosis of epithelioid or non-epithelioid PM from the German HITOC study were analyzed (Figure 1). Between 2008 and 2019, four high-volume departments of thoracic surgery in Germany collected clinical data on pre-, peri-, and postoperative variables according to a common protocol [14]. The study was funded by the German Research Association (DFG, RI2905/3-1). The trial was registered in the German Registry of Clinical Studies (DRKS-ID: DRKS00015012). The approval of the Ethics Committee of the University of Regensburg (No. 18-1119-104) and of the ethics committees of the respective participating centers were obtained. All patients underwent elective CRS and HITOC within one surgery. The surgical procedures and HITOC have been conducted as described before [12,15]. The existing database was supplemented by the additional collection of 64 clinical and follow-up data, which were updated until November 2021. One patient from the entire study population (n = 261) was excluded because he was initially treated for local recurrence of PM.

2.2. Definition of Variables

The histological subtype was categorized in epithelioid vs. non-epithelioid (biphasic and sarcomatoid). UICC stage was categorized as stage I ≥ II according to the 8th UICC tumor classification. Additive chemotherapy was defined as either induction chemotherapy and/or adjuvant chemotherapy. The beginning of primary therapy was defined as the date of surgery if no induction chemotherapy was applied. If induction chemotherapy was applied, beginning of primary therapy was set as the date of the first application of induction chemotherapy. HITOC was performed for 60–90 min at 42 °C with a flow rate of approximately 1000 mL per minute, depending on the standard operating procedure of the participating centers. The concentration for intrathoracic perfusion with cisplatin was categorized as a low (≤125 mg/m2 BSA = body surface area) vs. high (>125 mg/m2 BSA) dose. Overall survival (OS) was defined as the time from the beginning of the primary therapy until death from any cause. Disease-free survival (DFS) was defined as the time from the beginning of the primary therapy until the first objective tumor recurrence/progression or death from any cause, which ever occurred first. Recurrence or progression were defined as documented intrathoracic ipsilateral and/or contralateral tumor detection and distant metastases by cytology/histology and/or imaging. Patients received additive chemotherapy or follow-up following CRS and HITOC depending on the tumor stage, resection status, and the recommendation of the interdisciplinary tumor board at each of the four study centers.
Primary objective of the study was to analyze the OS rates of patients with PM who underwent CRS and HITOC within a multimodal treatment regimen. Secondary endpoints included DFS and the identification of prognostic factors.

2.3. Statistical Analysis

Descriptive analyses were performed for demographic, treatment, postoperative, and follow-up data using frequencies (n), percentages (%), means (m), standard deviations (SDs), medians (med), and interquartile ranges (IQRs). Median follow-up time was calculated using the reverse Kaplan–Meier method. OS and DFS were analyzed by the Kaplan–Meier estimator for the total patient cohort. Moreover, univariable analyses (log-rank tests) were used to compare OS and DFS between the cisplatin dosage, chemotherapeutical agent, resection status, histological subtype, UICC (Union Internationale Contre le Cancer Classification) stage, postoperative acute kidney injury, and additive chemotherapy. The estimates for the probability of surviving were presented graphically in Kaplan–Meier survival curves. Additionally, multivariable Cox proportional hazard regression models (enter method) were calculated. For the endpoints OS and DFS, the abovementioned clinical parameters (cisplatin dosage, chemotherapeutic agent, resection status, histological subtype, UICC stage, postoperative acute kidney injury, and additive chemotherapy), as well as the confounding variable time between the initial diagnosis and beginning of primary therapy, were included. Hazard ratios (HRs) with corresponding 95% CIs are presented. All statistical analyses were conducted using the software package SPSS (version 26 or higher). The significance level was set at ptwo-sided ≤ 0.050.

3. Results

The HITOC database contains a total of 350 patients. In total, 260 patients met the prespecified inclusion and exclusion criteria and were included in the present analyses. The mean age of the patient cohort was 65.5 ± 9.0 years, and 81.2% were male. Good performance status with ECOG (Performance Status Scale of the Eastern Cooperative Oncology Group) ≤ 1 was documented in 95.4% of the patients. Regarding tumor histology, 84.6% had the epithelioid tumor subtype, 13.5% had the biphasic subtype, and 1.9% had the sarcomatoid tumor subtype. According to tumor stage, 48.1% had stage I disease, 15.0% had stage II disease, 34.6% had stage III disease, and 2.3% had stage IV disease. Detailed patient characteristics are shown in Table 1.

3.1. Surgical Approach

CRS by lung-sparing PD was performed in 60 patients (23.1%), while ePD was performed in 193 patients (74.2%), including partial resection of the diaphragm in 173 patients (66.5%) and partial resection of the pericardium in 118 patients (45.4%). Partial resection of the chest wall was performed in 25 patients (9.6%) due to local tumor infiltration. Only 2.7% of patients underwent EPP. Macroscopic complete resection (MCR) was achieved in 222 patients (85.4%) and only 38 patients (14.6%) had macroscopic incomplete resection (R2) due to tumor infiltration into non-resectable structures. MCR was defined as the resection of all of the visible tumor with a residual tumor volume of less than 1 cm3, as defined by Sugarbaker and colleagues [6,16]. The median blood loss during surgery was 800 mL (IQR = 500, 1200). In total, 145 (55.8%) patients received a transfusion of erythrocytes, platelets, or fresh frozen plasma during surgery. Detailed characteristics are shown in Table 2.
Intrathoracic chemotherapy (HITOC) consisted of cisplatin as a single dose (58.5%) or in combination with doxorubicin (41.5%) and was administered with low-dose cisplatin (≤125 mg/m2 BSA; 73.2%) or high-dose cisplatin (>125 mg/m2 BSA; 26.8%) according to the capabilities of the participating center.

3.2. Postoperative Data

Detailed postoperative data are shown in Table 3. Total postoperative complications were documented in 142 patients (54.5%), while Clavien–Dindo grade I and II complications were documented in 24.6%. Complications requiring surgical intervention (Clavien–Dindo IIIb) occurred in 37 patients (14.2%) due to prolonged air leak and coagulo- or chylothorax. Grade IV complications occurred in 1.6% and in-hospital mortality was 3.5%. The median intensive care unit (ICU) stay was 2 days (IQR = 1 to 4 days), while the median hospital stay was 20 days (IQR = 15 to 29 days).

3.3. Multimodal Treatment and Follow-Up

A total of 136 patients (52.3%) received additive chemotherapy. Within this patient cohort, 50 patients received neoadjuvant chemotherapy, while 73 patients received adjuvant chemotherapy, and 13 patients received both neoadjuvant and adjuvant chemotherapy. Median follow-up, defined as the time from the start of primary therapy to the end of follow-up, was 48 months (95% CI = 38.4, 57.6). During follow-up, 157 patients (80.5%) had locoregional tumor recurrence (in the case of MCR) or tumor progression (when macroscopic incomplete resection (R2) was documented). Distant metastases with or without locoregional recurrence or progression occurred in 32 patients (14.5%). Tumor recurrence or progression was treated with chemotherapy in 117 patients (60.0%) and radiotherapy in 30 patients (15.4%). Details of additive treatment and follow-up are shown in Table 4.

3.4. Survival Analysis

Median OS was 27 months (95% CI = 21.9, 32.1, n = 258), and OS rates for 1, 3, and 5 years were 82%, 40%, and 22%, respectively (Figure 2A). Median DFS was 13 months (95% CI = 11.0, 15.0, n = 259) and rates for 1, 3, and 5 years were 52%, 15%, and 5%, respectively (Figure 2B). Median survival was significantly (p < 0.001) improved in patients with the epithelioid subtype (31 months, 95% CI = 25.9, 36.1) compared to the non-epithelioid (14 months, 95% CI = 10.8, 17.2) subtype (Figure 3A). The epithelioid subtype also had a positive impact on DFS (15 months (95% CI = 13.1, 16.9) vs. 8 months (95% CI = 7.2, 8.8); p < 0.001; Figure 3B). Patients with MCR had significantly longer OS (31 months (95% CI = 26.1, 35.9) vs. 15 months (95% CI = 13.1, 16.9); p < 0.001) and DFS (14 months (95% CI = 12.1, 15.9) vs. 8 months (95% CI = 5.8, 10.2); p = 0.008) compared to patients with an R2 resection status.
In addition, the addition of chemotherapy significantly improved the median OS (32 months (95% CI = 26.8, 37.2) vs. 21 months (95% CI = 13.9, 24.1); p = 0.020; Figure 3C), as well as DFS (15 months (95% CI = 12.6, 17.4) vs. 9 months (95% CI = 7.3, 10.7); p < 0.001; Figure 3D). UICC stage I vs. ≥II had a significant impact on OS (32 months (95% CI = 27.4, 36.6) vs. 19 months (95% CI = 13.9, 24.2); p = 0.028) but not on DFS (14 months (95% CI = 10.5, 17.5) vs. 11 months (95% CI = 9.2, 12.8); p = 0.272). The cisplatin dose (high versus low dose) had no significant impact on OS (39 months (95% CI = 16.1, 61.9) vs. 25 months (95% CI = 18.9, 31.2); p = 0.103) or DFS (11 months (95% CI = 8.8, 13.2) vs. 13 months (95% CI = 10.9, 15.1); p = 0.703).
Using Cox regression for the multivariable analysis (Table 5), the resection status with MCR and histologic subtype with epithelioid histology retained their significant impacts on OS (p-values < 0.001), as well as DFS (p = 0.010 and p < 0.001). In addition, additive chemotherapy still had a significant impact on DFS (p = 0.003).

4. Discussion

This multicenter study confirmed that patients with operable PM can benefit from CRS and HITOC as part of a multimodal therapy concept regarding disease-free survival and overall survival. In particular, a macroscopic complete resection status and epithelioid subtype were identified as relevant prognostic factors. Median survival of up to 31 months was demonstrated in these patients. The longest OS (median survival of 39 months) was observed in patients receiving high-dose cisplatin at 175 mg/m2 BSA, but this parameter did not reach statistical significance compared to low-dose cisplatin. The addition of chemotherapy also appears to have a positive impact on DFS. Thus, multimodal therapy including CRS in combination with HITOC and additional chemotherapy offers an opportunity to significantly improve the survival of selected patients with PM.
Systemic treatment with immunotherapy has become the standard of care for PM patients with locoregional disease that infiltrates surrounding structures [3]. However, immunotherapy has not shown a breakthrough in improving OS compared to chemotherapy specifically for the subset of epithelioid PM [3]. Therefore, a multimodal therapeutic approach is warranted for patients with early-stage disease to improve overall survival. Complete tumor resection and disease control should be accompanied by a good postoperative quality of life for the mostly elderly patients [6,15,17]. Although Canadian colleagues achieved attractive survival rates with a trimodal concept of radiotherapy, EPP and adjuvant chemotherapy in cases of nodal involvement, this concept showed a high morbidity with grade 3 and 4 complications in almost 50% of the cases [18]. In recent years, lung-sparing CRS in combination with HITOC has become a promising treatment concept for localized PM [7,16,18,19]. The preservation of the lung parenchyma provides the patient with a good cardiopulmonary reserve, which is particularly relevant for further systemic therapies within the multimodal therapy concept, as well as for the treatment of tumor recurrence [15,17,20,21]. Therefore, CRS has become the standard surgical technique in many thoracic surgical centers [7,9,11,16,22]. However, the ideal multimodal treatment approach has not yet been identified. In addition, the effects of additive chemotherapy together with CRS and HITOC need to be further investigated [23].
As a relevant outcome parameter for survival, the completion of multimodal therapy has a significant impact on DFS and OS [9,10]. Perioperative morbidity, as well as side effects of neoadjuvant systemic treatment, strongly influence further therapeutic management and the completion of multimodal treatment. In recent years, a multicenter, randomized phase II trial (EORTC 1205) investigated the feasibility of PD or ePD with neoadjuvant or adjuvant chemotherapy [24]. The primary endpoint was successful completion of multimodal treatment, followed by endpoints related to surgical quality, morbidity, and mortality. Initial results from this trial are expected at the end of the year. Our analysis did not show a difference in neoadjuvant or adjuvant systemic treatment. This may be due to the small number of patients receiving neoadjuvant therapy in our cohort.
CRS showed characteristic postoperative morbidity due to the large wound surface of the parietal and visceral pleura. In our study, surgical intervention was required in 14.2% of patients due to prolonged air leak, coagulothorax or cylothorax. In-hospital mortality was 3.5%. As other studies have shown, morbidity and mortality are comparable for this extensive cytoreductive procedure [11,19]. In addition, PD is a defensible cytoreductive surgical procedure within the multimodal therapy of PM compared to known studies including EPP in the multimodal approach [25,26,27]. Although no randomized trials can be performed to evaluate the benefit of HITOC, at least this additional procedure does not show a higher intraoperative complication rate. Our study population has comparable perioperative morbidity and mortality to other centers without HITOC [4,11,19]. Therefore, in our opinion, HITOC can be used as an additional procedure in the fight against PM [7,28].
Based on our retrospective data analysis, the interval from the end of multimodal treatment to tumor recurrence or progression was significantly improved, even after the multivariable analysis, especially in patients receiving additive chemotherapy. It is possible that adjuvant chemotherapy, usually administered postoperatively, can reach residual tumor cells through the large, well-vascularized wound surface. The results of the EORTC trial may provide recommendations on the timing of multimodal therapy. Nevertheless, our results clearly support the inclusion of additive chemotherapy in a multimodal treatment approach. Some centers in our multicenter analysis have not included additive chemotherapy in their standardized treatment regimens in recent years. As a result, we may have reduced the selection bias and the impact of a postoperative decreased general condition on the completion rate of multimodal treatment with additive chemotherapy. This may support our conclusion that additive chemotherapy has a positive impact on disease progression. In accordance with the latest recommendations of the WHO 2021 classification, future analyses may show whether there is an impact on therapeutic decisions and the treatment response when epithelioid mesothelioma is classified as a low-grade or high-grade tumor subtype [29].
The study has some relevant limitations due to its retrospective nature and the analysis of PM patients treated at different institutions over a long period of time. The described cohorts also include the clinical data during the implementation of CRS and HITOC in the different institutions. Based on this, the decision making and the definition of selection criteria for surgical candidates may have influenced the data on perioperative morbidity, MCR, DFS, and OS. Especially due to the recent discussions about multimodal therapy including surgery in the MARS-2 trial, critical patient selection is essential for the best possible survival of the individual patient. However, in summary, carefully selected PM patients could be treated with CRS, HITOC, and additive chemotherapy with low morbidity and promising survival.
Based on the significant impact of additive chemotherapy within this multimodal treatment approach, the NICITA trial (clinicaltrials.gov: NCT04177953), a multicenter trial in Germany of adjuvant chemoimmunotherapy versus chemotherapy alone after CRS for epithelioid PM, will hopefully provide promising results on the impact of immunotherapy within the multimodal treatment of the epithelioid subtype after CRS [30]. In addition, a detailed analysis of PM tumor tissue may help to further understand the tumor biology and develop effective treatment strategies.

5. Conclusions

Cytoreductive surgery and HITOC within a multimodal treatment approach have a positive impact on the survival of patients with PM, especially the epithelioid tumor subtype and after macroscopic complete tumor resection. Furthermore, the addition of chemotherapy significantly affects the interval to tumor recurrence compared to CRS and HITOC alone.

Author Contributions

Conceptualization: M.R., H.-S.H. and M.E.E.; methodology: M.R., M.E.E., L.V.K., J.Z., T.M., K.M., G.H. and M.K.; software: G.H., K.M. and M.K.; data acquisition: L.V.K., J.Z., M.E.E., J.K., M.R., T.M., M.H., B.P., H.-S.H., R.A.H. and S.S.; statistical analysis: K.M. and M.K.; writing—original draft preparation: L.V.K. and J.Z.; writing—reviewing & editing: M.E.E., M.R., L.V.K., K.M. and H.W.; data visualization, K.M.; funding acquisition: M.R. All authors have read and agreed to the published version of the manuscript.

Funding

This study was founded by the German Research Association (Deutsche Forschungsgemeinschaft (DFG), Funding number RI2905/3-1), Kennedyallee 40, 53170 Bonn, Germany.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the University of Regensburg (No. 18-1119-104) and of the ethics committees of the respective participating centers.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available on request due to privacy and ethical restrictions.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tedesco, J.; Jaradeh, M.; Vigneswaran, W.T. Malignant Pleural Mesothelioma: Current Understanding of the Immune Microenvironment and Treatments of a Rare Disease. Cancers 2022, 14, 4415. [Google Scholar] [CrossRef] [PubMed]
  2. Roe, O.D.; Stella, G.M. Malignant pleural mesothelioma: History, controversy and future of a manmade epidemic. Eur. Respir. Rev. 2015, 24, 115–131. [Google Scholar] [CrossRef] [PubMed]
  3. Baas, P.; Scherpereel, A.; Nowak, A.K.; Fujimoto, N.; Peters, S.; Tsao, A.S.; Mansfield, A.S.; Popat, S.; Jahan, T.; Antonia, S.; et al. First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): A multicentre, randomised, open-label, phase 3 trial. Lancet 2021, 397, 375–386. [Google Scholar] [CrossRef] [PubMed]
  4. Taioli, E.; Wolf, A.S.; Flores, R.M. Meta-analysis of survival after pleurectomy decortication versus extrapleural pneumonectomy in mesothelioma. Ann. Thorac. Surg. 2015, 99, 472–480. [Google Scholar] [CrossRef] [PubMed]
  5. Baldini, E.H.; Recht, A.; Strauss, G.M.; DeCamp, M.M., Jr.; Swanson, S.J.; Liptay, M.J.; Mentzer, S.J.; Sugarbaker, D.J. Patterns of failure after trimodality therapy for malignant pleural mesothelioma. Ann. Thorac. Surg. 1997, 63, 334–338. [Google Scholar] [CrossRef] [PubMed]
  6. Sugarbaker, D.J. Macroscopic complete resection: The goal of primary surgery in multimodality therapy for pleural mesothelioma. J. Thorac. Oncol. 2006, 1, 175–176. [Google Scholar] [CrossRef] [PubMed]
  7. Klotz, L.V.; Hoffmann, H.; Shah, R.; Eichhorn, F.; Gruenewald, C.; Bulut, E.L.; Griffo, R.; Muley, T.; Christopoulos, P.; Baum, P.; et al. Multimodal therapy of epithelioid pleural mesothelioma: Improved survival by changing the surgical treatment approach. Transl. Lung Cancer Res. 2022, 11, 2230–2242. [Google Scholar] [CrossRef] [PubMed]
  8. Sugarbaker, D.J.; Gill, R.R.; Yeap, B.Y.; Wolf, A.S.; DaSilva, M.C.; Baldini, E.H.; Bueno, R.; Richards, W.G. Hyperthermic intraoperative pleural cisplatin chemotherapy extends interval to recurrence and survival among low-risk patients with malignant pleural mesothelioma undergoing surgical macroscopic complete resection. J. Thorac. Cardiovasc. Surg. 2013, 145, 955–963. [Google Scholar] [CrossRef] [PubMed]
  9. Scherpereel, A.; Opitz, I.; Berghmans, T.; Psallidas, I.; Glatzer, M.; Rigau, D.; Astoul, P.; Bolukbas, S.; Boyd, J.; Coolen, J.; et al. ERS/ESTS/EACTS/ESTRO guidelines for the management of malignant pleural mesothelioma. Eur. Respir. J. 2020, 55, 1900953. [Google Scholar] [CrossRef]
  10. Kindler, H.L.; Ismaila, N.; Armato, S.G., 3rd; Bueno, R.; Hesdorffer, M.; Jahan, T.; Jones, C.M.; Miettinen, M.; Pass, H.; Rimner, A.; et al. Treatment of Malignant Pleural Mesothelioma: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2018, 36, 1343–1373. [Google Scholar] [CrossRef]
  11. Cao, C.; Tian, D.; Park, J.; Allan, J.; Pataky, K.A.; Yan, T.D. A systematic review and meta-analysis of surgical treatments for malignant pleural mesothelioma. Lung Cancer 2014, 83, 240–245. [Google Scholar] [CrossRef] [PubMed]
  12. Ried, M.; Kovacs, J.; Markowiak, T.; Muller, K.; Huppertz, G.; Koller, M.; Winter, H.; Klotz, L.V.; Hatz, R.; Zimmermann, J.; et al. Hyperthermic Intrathoracic Chemotherapy (HITOC) after Cytoreductive Surgery for Pleural Malignancies—A Retrospective, Multicentre Study. Cancers 2021, 13, 4580. [Google Scholar] [CrossRef] [PubMed]
  13. Verma, V.; Ahern, C.A.; Berlind, C.G.; Lindsay, W.D.; Grover, S.; Friedberg, J.S.; Simone, C.B., 2nd. Treatment of malignant pleural mesothelioma with chemotherapy preceding versus after surgical resection. J. Thorac. Cardiovasc. Surg. 2019, 157, 758–766.e1. [Google Scholar] [CrossRef] [PubMed]
  14. Markowiak, T.; Koller, M.; Zeman, F.; Huppertz, G.; Hofmann, H.S.; Ried, M.; Group, H.S. Protocol of a retrospective, multicentre observational study on hyperthermic intrathoracic chemotherapy in Germany. BMJ Open 2020, 10, e041511. [Google Scholar] [CrossRef]
  15. Klotz, L.V.; Gruenewald, C.; Bulut, E.L.; Eichhorn, F.; Thomas, M.; Shah, R.; Kriegsmann, M.; Schmidt, W.; Kofler, O.; Winter, H.; et al. Cytoreductive Thoracic Surgery Combined with Hyperthermic Chemoperfusion for Pleural Malignancies: A Single-Center Experience. Respiration 2021, 100, 1165–1173. [Google Scholar] [CrossRef]
  16. Bueno, R.; Opitz, I.; Taskforce, I.M. Surgery in Malignant Pleural Mesothelioma. J. Thorac. Oncol. 2018, 13, 1638–1654. [Google Scholar] [CrossRef]
  17. Schwartz, R.M.; Watson, A.; Wolf, A.; Flores, R.; Taioli, E. The impact of surgical approach on quality of life for pleural malignant mesothelioma. Ann. Transl. Med. 2017, 5, 230. [Google Scholar] [CrossRef] [PubMed]
  18. Cho, B.C.J.; Donahoe, L.; Bradbury, P.A.; Leighl, N.; Keshavjee, S.; Hope, A.; Pal, P.; Cabanero, M.; Czarnecka, K.; McRae, K.; et al. Surgery for malignant pleural mesothelioma after radiotherapy (SMART): Final results from a single-centre, phase 2 trial. Lancet Oncol. 2021, 22, 190–197. [Google Scholar] [CrossRef] [PubMed]
  19. Lapidot, M.; Gill, R.R.; Mazzola, E.; Freyaldenhoven, S.; Swanson, S.J.; Jaklitsch, M.T.; Sugarbaker, D.J.; Bueno, R. Pleurectomy Decortication in the Treatment of Malignant Pleural Mesothelioma: Encouraging Results and Novel Prognostic Implications Based on Experience in 355 Consecutive Patients. Ann. Surg. 2022, 275, 1212–1220. [Google Scholar] [CrossRef]
  20. Ambrogi, M.C.; Bertoglio, P.; Aprile, V.; Chella, A.; Korasidis, S.; Fontanini, G.; Fanucchi, O.; Lucchi, M.; Mussi, A. Diaphragm and lung-preserving surgery with hyperthermic chemotherapy for malignant pleural mesothelioma: A 10-year experience. J. Thorac. Cardiovasc. Surg. 2017, 155, 1857–1866.e2. [Google Scholar] [CrossRef]
  21. Sharkey, A.J.; Bilancia, R.; Tenconi, S.; Nakas, A.; Waller, D.A. Extended pleurectomy decortication for malignant pleural mesothelioma in the elderly: The need for an inclusive yet selective approach. Interact. Cardiovasc. Thorac. Surg. 2017, 25, 696–702. [Google Scholar] [CrossRef] [PubMed]
  22. Ripley, R.T. Extended Pleurectomy and Decortication for Malignant Pleural Mesothelioma. Thorac. Surg. Clin. 2020, 30, 451–460. [Google Scholar] [CrossRef] [PubMed]
  23. Sharkey, A.J.; O’Byrne, K.J.; Nakas, A.; Tenconi, S.; Fennell, D.A.; Waller, D.A. How does the timing of chemotherapy affect outcome following radical surgery for malignant pleural mesothelioma? Lung Cancer 2016, 100, 5–13. [Google Scholar] [CrossRef] [PubMed]
  24. Raskin, J.; Surmont, V.; Cornelissen, R.; Baas, P.; van Schil, P.E.Y.; van Meerbeeck, J.P. A randomized phase II study of pleurectomy/decortication preceded or followed by (neo-)adjuvant chemotherapy in patients with early stage malignant pleural mesothelioma (EORTC 1205). Transl. Lung Cancer Res. 2018, 7, 593–598. [Google Scholar] [CrossRef] [PubMed]
  25. Stahel, R.A.; Riesterer, O.; Xyrafas, A.; Opitz, I.; Beyeler, M.; Ochsenbein, A.; Fruh, M.; Cathomas, R.; Nackaerts, K.; Peters, S.; et al. Neoadjuvant chemotherapy and extrapleural pneumonectomy of malignant pleural mesothelioma with or without hemithoracic radiotherapy (SAKK 17/04): A randomised, international, multicentre phase 2 trial. Lancet Oncol. 2015, 16, 1651–1658. [Google Scholar] [CrossRef] [PubMed]
  26. Waller, D.A.; Dawson, A.G. Randomized controlled trials in malignant pleural mesothelioma surgery-mistakes made and lessons learned. Ann. Transl. Med. 2017, 5, 240. [Google Scholar] [CrossRef] [PubMed]
  27. Treasure, T.; Lang-Lazdunski, L.; Waller, D.; Bliss, J.M.; Tan, C.; Entwisle, J.; Snee, M.; O’Brien, M.; Thomas, G.; Senan, S.; et al. Extra-pleural pneumonectomy versus no extra-pleural pneumonectomy for patients with malignant pleural mesothelioma: Clinical outcomes of the Mesothelioma and Radical Surgery (MARS) randomised feasibility study. Lancet Oncol. 2011, 12, 763–772. [Google Scholar] [CrossRef] [PubMed]
  28. Zhao, Z.Y.; Zhao, S.S.; Ren, M.; Liu, Z.L.; Li, Z.; Yang, L. Effect of hyperthermic intrathoracic chemotherapy on the malignant pleural mesothelioma: A systematic review and meta-analysis. Oncotarget 2017, 8, 100640–100647. [Google Scholar] [CrossRef]
  29. Sauter, J.L.; Dacic, S.; Galateau-Salle, F.; Attanoos, R.L.; Butnor, K.J.; Churg, A.; Husain, A.N.; Kadota, K.; Khoor, A.; Nicholson, A.G.; et al. The 2021 WHO Classification of Tumors of the Pleura: Advances Since the 2015 Classification. J. Thorac. Oncol. 2022, 17, 608–622. [Google Scholar] [CrossRef]
  30. Shah, R.; Klotz, L.V.; Chung, I.; Feißt, M.; Schneider, M.A.; Riedel, J.; Bischoff, H.; Eichhorn, M.E.; Thomas, M. A Phase II Trial of Nivolumab With Chemotherapy Followed by Maintenance Nivolumab in Patients with Pleural Mesothelioma After Surgery: The NICITA Study Protocol. Clin. Lung Cancer 2020, 22, 142–146. [Google Scholar] [CrossRef]
Figure 1. CONSORT diagram of the identification and selection of eligible patients treated with CRS and HITOC for pleural mesothelioma.
Figure 1. CONSORT diagram of the identification and selection of eligible patients treated with CRS and HITOC for pleural mesothelioma.
Cancers 16 01587 g001
Figure 2. Overall and disease-free survival. (A) OS and (B) DFS for the whole study cohort of PM patients.
Figure 2. Overall and disease-free survival. (A) OS and (B) DFS for the whole study cohort of PM patients.
Cancers 16 01587 g002
Figure 3. Impacts of the histologic subtype and additive chemotherapy on disease-free and overall survival. (A) OS and (B) DFS distributed by histologic tumor subtype (epithelioid versus non-epithelioid). (C) OS and (D) DFS distributed by additive chemotherapy versus no additive chemotherapy.
Figure 3. Impacts of the histologic subtype and additive chemotherapy on disease-free and overall survival. (A) OS and (B) DFS distributed by histologic tumor subtype (epithelioid versus non-epithelioid). (C) OS and (D) DFS distributed by additive chemotherapy versus no additive chemotherapy.
Cancers 16 01587 g003
Table 1. Patient characteristics.
Table 1. Patient characteristics.
Clinical Parametersn = 260
Sex (n, %)
female49 (18.8)
male211 (81.2)
Age (years) (mean ± SD)65.5 ± 9.0
BMI (m2/kg) (mean ± SD)26.3 ± 3.9
ECOG (n, %)
0159 (61.2)
189 (34.2)
21 (0.4)
missing11 (4.2)
Asbestos exposure (n, %)
no56 (21.5)
yes171 (65.8)
missing/unknown33 (12.7)
Side (n, %)
right156 (60.0)
left104 (40.0)
MPM histology (n, %)
epithelioid220 (84.6)
biphasic35 (13.5)
sarcomatoid5 (1.9)
MPM tumor stage (UICC) (n, %)
IA15 (5.8)
IB110 (42.3)
II39 (15.0)
III90 (34.6)
IV6 (2.3)
Clinical data of 260 patients with PM, including histopathological results. BMI—body mass index.
Table 2. Surgical approach.
Table 2. Surgical approach.
Intraoperative Parametersn = 260
Resection method (n, %)
P/D60 (23.1)
eP/D193 (74.2)
EPP7 (2.7)
Resection of diaphragm (n, %)
no87 (54.6)
yes173 (66.5)
Replacement of diaphragm (n, %)
no193 (74.2)
yes67 (25.8)
Resection of pericardium (n, %)
no142 (54.6)
yes118 (45.4)
Replacement of pericardium (n, %)
no199 (76.5)
yes61 (23.5)
Resection of chest wall (n, %)
no235 (90.4)
yes25 (9.6)
Resection status
MCR222 (85.4)
R238 (14.6)
Blood loss (median, IQR) *800 (500, 1200)
Transfusion (n, %)
no112 (43.1)
yes145 (55.8)
missing3 (1.2)
Chemotherapy substances (n, %)
cisplatin alone152 (58.5)
cisplatin plus doxorubicin108 (41.5)
Cisplatin dosage (n, %)
low dose (≤125 mg/m2 BSA)191 (73.2)
high dose (>125 mg/m2 BSA)70 (26.8)
Intraoperative complication
no252 (96.9)
yes8 (3.1)
* Blood loss was documented in 200 out of 260 (76.9%) patients. The number of transfusions was computed for 257 of 260 (98.8%) patients. Detailed data on the extent of resection during CRS and HITOC.
Table 3. Postoperative data.
Table 3. Postoperative data.
Postoperative Parametersn = 260
Postoperative complications (n, %)
no118 (45.4)
yes/Clavien–Dindo classification142 (54.6)
I12 (4.6)
II52 (20.0)
IIIa28 (10.8)
IIIb37 (14.2)
IVa1 (0.4)
IVb3 (1.2)
V9 (3.5)
Revision (n, %)
no224 (86.2)
yes/revision due to36 (13.8)
coagulothorax9 (3.5)
pleural empyema3 (1.2)
chylothorax5 (1.9)
parenchymal fistula/bronchial fistula8 (3.1)
other11 (4.2)
Prolonged ventilation > 24 h (n, %)
no251 (96.5)
yes9 (3.5)
Extubation in the OR (n, %)
no21 (8.1)
yes239 (91.9)
ICU stay [days] (median, IQR)2 (1, 4)
Hospital stay [days] (median, IQR)20 (15, 29)
In-hospital mortality (n, %)9 (3.5)
Overview on postoperative complications and surgical revisions together with data on hospitalization. ICU—intensive care unit, OR—operating room.
Table 4. Multimodal treatment and follow-up.
Table 4. Multimodal treatment and follow-up.
Postoperative Coursen = 260
Neoadjuvant chemotherapy (n, %)
no197 (75.8)
yes63 (24.2)
Adjuvant chemotherapy (n, %)
no171 (65.8)
yes86 (33.1)
missing3 (1.2)
Additive chemotherapy (n, %)
no121 (46.5)
yes136 (52.3)
missing3 (1.2)
Adjuvant radiotherapy (n, %)
no199 (76.5)
yes58 (22.3)
missing3 (1.2)
Recurrence (MCR, n = 222) (n, %)
no52 (23.4)
yes170 (76.6)
Progression (R2, n = 38) (n, %)
no13 (34.2)
yes38 (65.8)
Location of recurrence/progression (n = 195) (n, %)
locoregional157 (80.5)
distant metastasis15 (5.8)
locoregional and distant metastasis17 (8.7)
missing6 (3.1)
Site of recurrence/progression (n = 195) (n, %)
ipsilateral174 (89.2)
contralateral6 (3.1)
missing15 (5.8)
Therapy for recurrence/progression (n = 195) (n, %)
no11 (5.6)
unknown16 (8.2)
yes/type168 (86.2)
surgery19 (9.7)
chemotherapy117 (60.0)
radiotherapy30 (15.4)
supportive care14 (7.2)
other3 (1.5)
Time between the start of primary therapy until the end of follow-up in months *
(reverse Kaplan–Meier OS) (median, 95% CI)
48 (38, 58)
* A total of 258 of 260 (99.2%) patients; two patients were excluded from analyses as either the date of start of primary therapy or the date of death was missing. Both patients died. Additive treatment within a multimodal treatment concept including follow-up data.
Table 5. Cox regression analysis of OS and DFS.
Table 5. Cox regression analysis of OS and DFS.
Overall SurvivalDisease-Free Survival
Sig.HR95% CISig.HR95% CI
cisplatin dosage0.3500.810.531.250.8311.040.741.45
chemotherapeutic agent0.1141.350.931.950.5561.100.801.53
resection status<0.0012.251.453.490.0101.721.142.60
histological subtype<0.0012.271.503.41<0.0012.431.633.61
UICC stage0.5441.110.791.570.9861.000.741.34
postoperative AKI0.8971.030.641.650.5151.140.771.70
additive chemotherapy0.1840.790.561.120.0030.640.470.85
time between the initial diagnosis and start of primary therapy0.7121.010.971.050.7721.000.981.04
N = 255 *; n = 162 dead, n = 93 censoredN = 256 **; n = 218 recurrence/progression or dead, n = 38 censored
* Five out of 260 (1.9%) patients were excluded due to missing information about additive chemotherapy (n = 3) or time points (n = 2). ** Four out of 260 (1.5%) patients were excluded due to a missing date of the start of primary therapy (n = 1) or missing information about additive chemotherapy (n = 3). For the one patient with a missing date of death, the date of recurrence/progression was available. Thus, the patient was included in recurrence/progression-free survival analysis. Reference categories: low-dose cisplatin, cisplatin alone, resection status R1, epithelioid MPM, UICC stage I, no postoperative acute kidney insufficiency (AKI), and no additive chemotherapy.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Klotz, L.V.; Zimmermann, J.; Müller, K.; Kovács, J.; Hassan, M.; Koller, M.; Schmid, S.; Huppertz, G.; Markowiak, T.; Passlick, B.; et al. Multimodal Treatment of Pleural Mesothelioma with Cytoreductive Surgery and Hyperthermic Intrathoracic Chemotherapy: Impact of Additive Chemotherapy. Cancers 2024, 16, 1587. https://doi.org/10.3390/cancers16081587

AMA Style

Klotz LV, Zimmermann J, Müller K, Kovács J, Hassan M, Koller M, Schmid S, Huppertz G, Markowiak T, Passlick B, et al. Multimodal Treatment of Pleural Mesothelioma with Cytoreductive Surgery and Hyperthermic Intrathoracic Chemotherapy: Impact of Additive Chemotherapy. Cancers. 2024; 16(8):1587. https://doi.org/10.3390/cancers16081587

Chicago/Turabian Style

Klotz, Laura V., Julia Zimmermann, Karolina Müller, Julia Kovács, Mohamed Hassan, Michael Koller, Severin Schmid, Gunnar Huppertz, Till Markowiak, Bernward Passlick, and et al. 2024. "Multimodal Treatment of Pleural Mesothelioma with Cytoreductive Surgery and Hyperthermic Intrathoracic Chemotherapy: Impact of Additive Chemotherapy" Cancers 16, no. 8: 1587. https://doi.org/10.3390/cancers16081587

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

Article Metrics

Back to TopTop