Novel therapies for malignant pleural effusion: Anti‑angiogenic therapy and immunotherapy (Review)

  • Authors:
    • Dan He
    • Ruilin Ding
    • Qinglian Wen
    • Longxia Chen
  • View Affiliations

  • Published online on: January 22, 2021     https://doi.org/10.3892/ijo.2021.5174
  • Pages: 359-370
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Patients with a variety of malignancies can develop malignant pleural effusion (MPE). MPE can cause significant symptoms and result in a marked decrease in quality of life and a poor prognosis. MPE is primarily considered as an immune and vascular manifestation of pleural metastases. In the present review, the existing evidence supporting the applicability of anti‑angiogenic therapy and immunotherapy for the treatment of MPE was summarized. Patients with MPE have benefited from anti‑angiogenic agents, including bevacizumab and endostar; however, no relevant prospective phase III trial has, thus far, specifically analyzed the benefit of anti‑angiogenic therapy in MPE. Immunotherapy for MPE may be sufficient to turn a dire clinical situation into a therapeutic advantage. Similar to anti‑angiogenic therapy, more clinical data on the efficiency and safety of immunotherapy for controlling MPE are urgently required. The combined use of anti‑angiogenic therapy and immunotherapy may be a promising strategy for MPE, which requires to be further understood.

1. Introduction

Tumor cells that have metastasized to the pleural space may result in malignant pleural effusion (MPE), which can develop in patients with various types of tumor, including breast, lung and hematological tumors (1,2). Dyspnea, chest pain and coughing are common symptoms of MPE (1). A significant decrease in quality of life (QoL) and a poor prognosis can be observed in patients with MPE (1).

Currently, the popular approaches for MPE management include pleuroscopy with subsequent chemical pleurodesis and thoracostomy (2). However, these treatment methods only provide symptomatic relief, with poor and unsatisfactory results (2). Furthermore, adverse effects such as chest pain, fever and dyspnea are often observed (3).

At present, the pathogenesis of MPE is not fully understood, but it is associated with impaired pleural fluid drainage (4). When metastatic cancer infiltrates the thoracic lymph nodes and pleura, the normal cycle of fluid secretion and absorption is interrupted, and the fluid is finally collected (4,5). MPE is the build-up of fluid in the pleural space, which contains immune cells, cancer cells and proteins (5). Cytokines and chemokines, including interleukin (IL)-10 (6), IL-6 (5), transforming growth factor (TGF)p (7) and vascular endothelial growth factor (VEGF) (8), are abundant in MPE. These factors serve an important role in MPE formation and can be used as therapeutic targets that enable MPE treatment. Among them, VEGF, which can prompt the formation of new blood vessels, is a key mediator of MPE pathogenesis (9). Several therapeutic strategies for MPE have focused on this protein (4,8,9). Furthermore, the pleural space in MPE is regarded as a tumor-tolerogenic milieu, which has a complex connection with immunosuppressive factors (10). However, this tumor-tolerogenic milieu can be reversed by immunotherapy, which has the potential to stimulate tumor-specific immune responses in the pleural space (10). Therefore, immunotherapy has been an area of special interest for MPE treatment (10).

MPE is primarily considered as an immune and vascular manifestation of pleural-metastasized cancer (11,12). Therefore, in the present review, the existing evidence supporting the applicability of anti-angiogenic treatment and immunotherapy for the treatment of MPE was summarized, and the implications of the recent developments in MPE treatment were highlighted.

2. Anti-angiogenic treatment for malignant pleural effusion (MPE)

Role of VEGF in MPE

Large amounts of VEGF can be produced as tumor cells invade into the pleura, resulting in the acceleration of vascular permeability (13). The pleural fluid VEGF levels in MPE are significantly higher than those in effusions of benign disease, such as congestive heart failure and tuberculosis (2,8,14). In 2011, Fiorelli et al (9) studied 79 patients with unilateral PE. The levels of VEGF were demonstrated to be much higher in malignant than in benign exudates (9). In addition, in a study by Lieser et al (15), a 77-fold higher VEGF expression was observed in MPE compared with that in benign PEs. VEGF has therefore been suggested to be a diagnostic marker for MPE (16). Furthermore, patients with MPE with a high pleural VEGF level have been reported to have a significantly shorter survival than those with normal VEGF level (17,18).

The effects of VEGF are mainly mediated by endothelial cell receptors VEGF receptor-1 (VEGFR-1) and VEGFR-2 (19). VEGF binds to its receptors and induces downstream signaling, such as that of protein kinase C and mitogen-activated protein kinase; this process can induce vascular endothelium differentiation and proliferation, stimulate capillary sprouting and finally produce endothelial fenestrations and loss of junctional integrity, contributing to tumor growth and MPE development (4). Thus, the inhibition of VEGF activity with VEGF inhibitors is regarded as a promising approach for improving the management of MPE.

Bevacizumab treatment for MPE

Bevacizumab (Avastin®; Roche), the first humanized monoclonal antibody against VEGF, has been used to treat several types of tumor, including lung cancer and gynecological cancers (20-22). It can directly inhibit the proliferation and migration of vascular endothelial cells, promote the apoptosis of endothelial cells and suppress VEGF-induced neoangiogenesis and vascular permeability (23). Several studies have specifically researched the efficacy and safety of bevacizumab for the management of MPE (Table I). Among them, 6 are prospective studies (13,24-28), including two phase II trials conducted by Japanese groups (24,25). Unfortunately, there is still a lack of evidence from large phase 111 trials to confirm the effect of bevacizumab in the management of MPE.

Table I

Efficacy of anti-angiogenic therapy for MPE.

Table I

Efficacy of anti-angiogenic therapy for MPE.

Authors, yearStudy design (phase)Antiangiogenic agentN AdministrationrouteTreatmentMPE control, n/total n (%)Long-term efficacyRef.
Usui et al, 2016P (II)Bevacizumab28IVBevacizumab + carboplatin26/28 (92.9)amPFS: 8.2 months
mOS: 18.6 months
(24)
Tamiya et al, 2013P (II)Bevacizumab23IVBevacizumab + carboplatin + paclitaxel21/23 (91.3)bmPFS: 7.1 months
mOS: 11.7 months
(25)
Qi et al, 2016PBevacizumab24IPi) Paclitaxel;
ii) paclitaxel + bevacizumab
i) 5/10 (50.0)c;
ii) 11/14 (78.6)c
1 year survival rate:
i) 20.8%
ii) 45.8%
(26)
Du et al, 2013PBevacizumab70IPi) Cisplatin;
ii) cisplatin + bevacizumab
i) 17/34 (50.0)c
ii) 30/36 (83.3)c
mPFS: i) 4.5 months
ii) 5.3 months
(27)
Wang et al, 2018PBevacizumab33IPPaclitaxel + bevacizumab25/33 (75.8%)cmPFS: 8.4 months
mOS: 22.2 months
(28)
Nie et al, 2020PBevacizumab43IP/IVi) IV bevacizumab;
ii) IP bevacizumab
i) 14/21 (66.7)c
ii) 16/20 (80.0)c
NR(13)
Masago et al, 2015RBevacizumab21IVBevacizumab + chemotherapy15/21 (71.4)cNR(30)
Tao et al, 2018RBevacizumab21IVBevacizumab + chemotherapy(90.0 at 12 months)dmOS: 25.8 months(3)
Kitamura et al, 2013RBevacizumab13IVBevacizumab + chemotherapy12/13 (92.3)bmPFS without re accumulation of MPE: 312 days(29)
Jiang et al, 2017RBevacizumab86IVi) Bevacizumab + EGFR TKI;
ii) bevacizumab + chemotherapy
i) 42/47 (89.4)c; ii) 25/39 (64.1)cmPFS: i) 6.3 months; ii) 4.8 months(35)
Song et al, 2018RBevacizumab45IPi) Bevacizumab + pemetrexed;
ii) bevacizumab + cisplatin
i) 19/22 (86.4)c; ii) 13/23 (56.5)cmPFS: i) 5.4 months;
ii) 4.0 months
mOS: i) 10.5 months;
ii) 10.3 months
(33)
Jiang et al, 2016RBevacizumab43eIPi) Bevacizumab + cisplatin;
ii) cisplatin
i) 16/20 (80.0)c;
ii) 11/23 (47.8)c
mPFS: i) 5.4 months;
ii) 4.0 months
mOS: i) 10.5 months;
ii) 10.3 months
(32)
Wang et al, 2005PEndostar128IPi) Endostar + pemetrexed + cisplatin;
ii) pemetrexed + cisplatin
i) 62/66 (93.9)b;
ii) 49/62 (79.0)b
1-year survival rate:
i) 78.79%
ii) 74.19%
(37)
Zhao et al, 2014PEndostar45eIPi) Endostar + cisplatin;
ii) cisplatin
i) 18/23 (78.3)c;
ii) 9/22 (40.9)c
NR(39)
Matsumorii et al, 2006P (II)Vandetanib20OralVandetanib-mOS: 10.6 months(41)
Mulder et al, 2014P (II)Cediranib12eOrali) Cediranib administrated on day 1
ii) cediranib administrated on day 29
-mOS: i) 73 days;
ii) 24 days
(44)

a MPE control defined as not needing pleurodesis at 8 weeks of treatment;

b MPE control defined as no re-accumulation of MPE for ≥8 weeks after the start of treatment;

c MPE control defined as complete response and partial response/incomplete relief;

d MPE control defined as the percentage of patients who did not experience re-accumulation of pleural fluid for 4 weeks from the time of the catheter being removed.

e Including patients with symptomatic malignant ascites and MPE. MPE, malignant pleural effusion; mOS, median overall survival; mPFS, median progression free survival; P, prospective; R, retrospective; IV, intravenous; IP, intraperitoneal; EFGR-TKI, epidermal growth factor receptor tyrosine kinase inhibitor; NR, not reported.

Retrospective studies

The first retrospective study to investigate the efficacy of bevacizumab plus chemotherapy for patients with MPE was conducted by Kitamura et al (29). The aforementioned study analyzed data from 13 patients with MPE caused by non-small cell lung cancer (NSCLC) who received bevacizumab (15 mg/kg, intravenously) plus chemotherapy as first- or second-line treatment (29). As expected, an MPE control lasting >8 weeks was achieved in 12/13 patients (92.3%) (29). Similar results were obtained in other studies (3,30), which analyzed the records of patients treated for NSCLC-associated MPE, who consequently received bevacizumab (15 or 7.5 mg/kg, intravenously) plus chemotherapy. In one study, a total of 15/21 patients were responders, and a response rate of 71.4% was reported (30), while an MPE response rate of 81.0% was observed in the other study (3).

The delivery of bevacizumab directly to the pleural space is an alternative for MPE treatment. The intrapleural administration of bevacizumab for MPE treatment has been suggested in previous studies. Chen et al (31) demonstrated that the intrapleural infusion of bevacizumab was effective in controlling MPE without apparent toxicity. Jiang et al (32) came to a similar conclusion, with bevacizumab significantly improving the response rate and QoL of patients with MPE without notable adverse events (AEs). Song et al (33) demonstrated that, in patients with MPE treated with an intrapleural infusion of bevacizumab (200 mg) combined with pemetrexed (BP group) or cisplatin (BD group), the response rates in the BP and BD groups were 56.52 and 86.36%, respectively, and the overall survival (OS) time for both groups was both >10 months. These results suggested that the intrapleural infusion of bevacizumab combined with chemotherapy may be effective for patients with MPE (33).

Prospective studies

Two phase II trials analyzing the efficacy of bevacizumab in treating MPE were conducted on Japanese patients (24,25). The first study enrolled 23 patients with NSCLC with MPE who were treated with bevacizumab (15 mg/kg, intravenously) and carboplatin-paclitaxel (25). The primary endpoint of this study was an overall response rate (ORR) of 60.8% (25). The disease control rate (DCR), median progression-free survival (PFS) time and median OS time were 87.0%, 7.1 and 11.7 months, respectively (25). Furthermore, the addition of bevacizumab into the treatment protocol increased the MPE control rate from 78.3 to 91.3% (25). Correspondingly, the serum VEGF levels were decreased from 513.6 to 25.1 pg/ml following bevacizumab treatment (25). The second study focused on the treatment of bevacizumab with carboplatin-pemetrexed (24). A total of 28 patients with NSCLC-associated MPE were treated with carboplatin and pemetrexed with bevacizumab (15 mg/kg, intravenously) every 3 weeks (24). The control rate of MPE without pleurodesis at 8 weeks after treatment was defined as the primary endpoint (24). A total of 26/28 (92.8%) patients reached the primary endpoint (24). The median PFS, OS and median pleurodesis-free survival times were 8.2, 18.6 and 13.9 months, respectively (24). A high VEGF (≥100 pg/ml) level in the plasma, indicating a poor prognosis, was also observed (24).

Three Chinese prospective studies investigated the efficacy of intrapleural bevacizumab administration in patients with MPE (26-28). Wang et al (28) enrolled 33 patients with NSCLC with MPE who all received paclitaxel and bevacizumab (5 mg/kg, intrapleurally) once every 3 weeks for 12 consecutive weeks. The total response rate of this study reached 77% (28). The median OS and median PFS times were 22.2 and 8.4 months, respectively (28). Du et al (27) directly compared the efficacy of combined intrapleural therapy with bevacizumab (300 mg) and cisplatin versus cisplatin alone in controling MPE. A total of 70 patients with NSCLC with MPE were included, with 35 patients per group (27). The results revealed that the addition of bevacizumab improved the ORR from 50 to 83.3% (27). Additionally, patients in the bevacizumab + cisplatin group exhibited a higher QoL benefit and a higher decrease in pleural VEGF levels (27). Another group of 24 patients with NSCLC was enrolled in a similar study by Qi et al (26). A total of 14 patients in that study received intrapleural infusion of paclitaxel and bevacizumab (5 mg/kg), and the remaining 10 received paclitaxel alone (26). The results revealed that combination therapy significantly decreased the MPE levels, with an overall efficacy rate of 78.6% (a 29% increase compared with paclitaxel alone) (26). The addition of bevacizumab also improved the 1-year survival rate from 20.8 to 45.8% (26).

In 2017, Zongwen et al (34) performed a meta-analysis that included 11 randomized controlled trials (RCTs) with a total of 769 patients with lung cancer, to investigate the efficacy and safety of bevacizumab in controling MPE. All the studies in the meta-analysis were conducted by Chinese groups and most of them (10/11) were published in Chinese (34). This meta-analysis provided further evidence to support the administration of bevacizumab via intrapleural injection in patients with MPE (34). As compared with platinum alone, the addition of bevacizumab significantly increased the ORR (P=0.003), decreased the incidence of chest pain (P<0.001) and relieved dyspnea in patients (P=0.002) (34).

Intrapleural or intravenous infusion

The aforementioned studies indicated that the combination of bevacizumab and chemotherapy may be effective for controlling MPE, although the sample sizes of these studies were small. For MPE management, bevacizumab can be administered intravenously or intrapleurally. However, the optimal administration route of bevacizumab has not yet been defined. Theoretically, compared with intravenous infusion, intrapleural infusion has some advantages, including the site-specific concentration of therapeutic agents with lower overall doses (10). Furthermore, the closed nature of the pleural space makes it an ideal site for intrapleural infusion (10). A recent randomized clinical study compared the efficiency and safety of intrapleural (7.5 mg/kg) and intravenous (7.5 mg/kg) infusion of bevacizumab in the management of MPE (13). Hypertension, epistaxis and proteinuria are common AEs associated with bevacizumab, which occurred more often in the intravenous group compared with in the intrapleural one (13). A higher ORR and a longer median duration of response were also observed in the intrapleural group, but the difference between the two groups was not statistically significant (13). The reason for this may be that the sample size of the study was too small (n=43). Therefore, large studies are required to confirm these results.

Future directions and ongoing trials

Currently, to the best of our knowledge, there are no prospective phase III studies specifically focusing on the benefit of bevacizumab combined with chemotherapy in MPE, which is a major issue. In addition, most of the aforementioned studies, whether prospective or retrospective, were conducted by Chinese and Japanese groups, which may lead to geographical and ethnical differences in the results. Therefore, data from other ethnic groups are required. Furthermore, there is a lack of data concerning rare tumor-driver mutations in patients with MPE caused by NSCLC. Jiang et al (35) retrospectively investigated 86 patients with NSCLC with MPE who had developed acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) therapy. In the aforementioned study, patients who were treated with bevacizumab and EGFR-TKIs had a longer PFS and higher curative efficacy rate for MPE, indicating that this combination therapy may be a valuable treatment option for patients with MPE (EGFR mutation-positive) caused by NSCLC (35).

Treatment of MPE with other anti-angiogenic drugs Recombinant human endostatin

Endostatin is an endogenous inhibitor of angiogenesis, which can interfere with the pro-angiogenic effects of growth factors and inhibit angiogenesis in a wide range of tumors, including lung, gastric and colon cancer (36-39). Similarly to VEGF, endostatin is also a potential prognostic factor for MPE (18,36). To enhance the solubility and stability of endostatin, recombinant human endostatin was engineered (a 9-amino acid sequence was added to the N-terminal of the protein) and termed endostar (16). Based on the results of a phase III study (37), endostar was listed as a first-line drug for the treatment of NSCLC in Chinese patients.

For patients with MPE, endostar was effective when administered via intrapleural infusion (37-39). A Chinese group evaluated the efficacy of endostar combined with cisplatin/pemetrexed chemotherapy for elderly patients with MPE (38). A total of 128 patients with lung adenocarcinoma with MPE were randomly assigned to receive chemotherapy plus endostar (treatment group) or chemotherapy alone (control group) (38). Compared with chemotherapy alone, endostar plus chemotherapy significantly improved MPE control rates (93.94 vs. 79.03%; P=0.013) and decreased recurrence rates (9.68 vs. 30.61%; P=0.005) with tolerable side effects (38). Similar results were obtained in a smaller RCT that included 45 patients with MPE or ascites (39). Furthermore, Biaoxue et al (40) conducted a meta-analysis based on 13 RCTs that included 1,066 patients with MPE, demonstrating that the addition of endostar to chemotherapeutic agents can significantly improve ORR and DCR, indicating that endostar is effective in treating MPE (40). However, one of the limitations of the aforementioned meta-analysis was that all included studies were from China. To the best of our knowledge, no data are available from outside China. Therefore, although endostar appears to be promising in controlling MPE, it still requires further investigation before it can be recommended for clinical application.

Anti-angiogenic TKIs

In addition to VEGF, other pro-angiogenic factors, such as TGF, platelet derived growth factor (PDGF) and fibroblast growth factor, have been associated with the development of PEs (4). Anti-angiogenic TKIs, which block the kinase activity of receptors, have been explored for the treatment of MPE. In in vitro and murine models of MPE, anti-angiogenic TKIs, such as lenvatinib, vatalanib and nintedanib, have been reported to control MPE (41,42).

Data from clincal trials evaluating the efficacy of EGFR-TKIs in patients with MPE are limited, with only two phase II trials (43,44). The first study enrolled 20 patients with NSCLC to evaluate the efficacy of vandetanib (VEGFR- and EGFR-TKI) for MPE control (43). The patients received oral vandetanib at a dose of 300 mg once a day for a maximum of 10 weeks, and time to pleurodesis was the primary endpoint (43). The results revealed that vandetanib was well tolerated, but it did not significantly decrease time to pleurodesis (43). The second study enrolled 12 patients with malignant ascites or MPE to assess the palliative value of cediranib (VEGF-TKI) (44). The primary endpoint was puncture-free survival, which was defined as the time from the start of the study to the time that paracentesis or thoracentesis were first needed, or the time of death (44). As expected, cediranib treatment significantly increased puncture-free survival with an acceptable toxicity profile (44). This phase II trial was the first study to show the palliative effects of oral VEGFR-TKI in patients with malignant effusions (44). Although sorafenib (RAF-, PDGFR- and VEGFR-TKI) has been reported to decrease MPE in one patient with advanced thyroid carcinoma (45), no further evidence has been acquired to support this treatment strategy.

The clinical trials on TKIs in this context are limited, and more data are urgently required. The other important issue is that most anti-angiogenic TKIs are administrated intraorally. Therefore, whether safe doses of anti-angiogenic TKIs comprise therapeutic concentrations for MPE through oral administration needs to be further investigated.

Another anti-VEGF antibody

In addition to bevacizumab, ramucirumab is another anti-VEGF antibody used in a clinical setting. The PLEURAM study is an ongoing phase II trial evaluating the efficacy and safety of ramucirumab in controling MPE (46). The study plans to enroll 15 patients with NSCLC and ramucirumab (10 mg/kg) combined with docetaxel (60 mg/m2) will be administered to each patient every 3 weeks (46). The MPE control rate at 8 weeks will be the primary endpoint (46).

3. Immunotherapy for MPE

Research on MPE control has al so shed light on immunotherapy. Given the potential for stimulating tumor-specific immune responses in the pleural space, intrapleural immunotherapy has been an area of notable interest for MPE treatment (47). Cytokines, which can be used as potent immunostimulatory agents to counter tumor-mediated immune tolerance and T-cell exhaustion, have long been investigated for the treatment of MPE. Adoptive therapy with chimeric antigen receptor (CAR) T cells or tumor-infiltrating lymphocytes (TILs), most often using the patients' own immune cells to treat their cancer, are also being examined for use in MPE treatment. Other treatments for MPE, such as immunogene therapy and oncolytic virotherapy, are currently underdeveloped.

Cytokine-based immunotherapy
1L-2 treatment

IL-2, which is produced primarily by activated CD4+ and CD8+ T cells, may act as a growth factor for all T-cell subsets (48), which may change a non-inflamed tumor into an inflamed tumor, thereby increasing the sensitivity of that tumor to further immune attack (49). IL-2 has been used to control MPE through intrapleural infusion for a long time. As early as in 1993, a group from France performed a phase I study to determine the safety and efficacy of intrapleural recombinant IL-2 infusion in 22 patients with MPE (50). The results revealed that 10/22 patients achieved responses during the course of the treatment, including l case of complete remission (CR) and 9 of partial remission (PRs) (50). Subsequently, other phase I/II studies on the intrapleural infusion of recombinant IL-2 for patients with MPE had an ORR of 21.7-22.0% (51,52), except for a phase I study conducted by Suzuki et al (53) in 1993, which had a total response rate of 100.0% (11/11 patients). The toxicity of IL-2 was found to be dose-dependent and the most common AE was fever; transient abnormal renal function, eosinophilia and flu-like syndrome were also observed (51-53). A meta-analysis of 18 Chinese clinical trials demonstrated that the thoracic injection of IL-2 and cisplatin led to a higher ORR, DCR and QoL than cisplatin alone in patients with MPE (54). Furthermore, Hu et al (55) revealed that IL-2 administration decreased the expression levels of programmed cell death protein 1 (PD-1), increased those of granzyme B and interferon (IFN)γ and enhanced the proliferation of CD8+ T cells in MPE. These results indicated that the exhaustion phenotype of CD8+ T cells, which contributes to tumor immune escape and metastasis, may be reversed by IL-2 treatment. Therefore, Hu et al (55) provided new evidence supporting the use of IL-2 in MPE.

To the best of our knowledge, no studies have been performed in the past 3 years on the role of IL-2 in treating MPE, particularly outside China. Although IL-2 treatment is not a mainstay of MPE treatment, IL-2 should be reexamined in this setting for novel combination therapy. For example, IL-2 may be used in a rationally designed combination therapy with immune checkpoint inhibitors (ICIs), due to its role in PD-1 expression (55). Other interleukins, such as IL-10 (56), IL-17 (57) and IL-27 (58), may also have the potential to inhibit the development of MPE, but they require further exploration.

Tumor necrosis factor-α (TNF-α) treatment

Similar to IL-2, TNF-a has been studied for its potential role in MPE management. In a study by Li et al (59), 102 patients with lung cancer with MPE received a single dose of recombinant human TNF-a (rhu-TNF) following maximum drainage of the pleural cavity. The results revealed that intrapleural infusion of rhu-TNF sufficiently controlled MPE with a response rate of 81.37% and that the AEs of this treatment were well tolerated (59). The data demonstrated that the short-term efficacy of TNF-a treatment was non-inferior to that of anti-angiogenic treatment, such as bevacizumab (59). However, the study was limited in that it was a retrospective trial.

IFN treatment

IFNs are a family of cytokine mediators. Type I IFNs, such as IFN-a and IFN-p, are known to stimulate the immune system and inhibit tumor cell proliferation (60). Due to their role in the immune system, IFNs are being investigated and used in various respiratory disorders, including MPE (60). In 1993, Goldman et al (61) evaluated the safety and efficacy of intrapleural IFN-a2b in patients with MPE. A total of 14/20 (70%) evaluable patients exhibited responses lasting for a median of 6 months, including 8 cases of CR and 6 of PR (61). The response rate of IFN-α2b treatment in that study was encouraging. However, a prospective randomized trial published in 2004 revealed that standard bleomycin chemotherapy was more effective than IFN-a2b in MPE, with patients in the bleomycin group exhibiting a higher response rate and longer survival (62). Since then, studies on IFN-a2b in MPE treatment have been rare.

IFN-β is another type I IFN that has been investigated for MPE treatment. In total, 10 patients with malignant pleural mesothelioma (MPM) or MPE were enrolled in a phase I study to evaluate the safety and feasibility of a single-dose intrapleural IFN-β gene transfer using an adenoviral vector expressing IFN-β (Ad.IFN-β) (63). Intrapleural Ad.IFN-β was generally well tolerated, with 7/10 patients responding to this treatment method (63), which indicated that intrapleural Ad.IFN-β may be a potentially useful approach for the treatment of MPE. A follow-up phase I trial was conducted 3 years later, and 7 patients with MPE and 10 with MPM were enrolled to receive 2 doses of intrapleural Ad.IFN (64). After the first dose, the pleural IFN-β levels were significantly elevated; however, the elevated levels were not sustained, falling to <1 ng/ml after the second dose (64). These unsatisfactory results may have been associated with the rapid development of neutralizing antibodies against the adenoviral vector after the second dose (64).

Intrapleural immunogene therapy

The aforementioned Ad.IFN-β treatment is an example of using intrapleural immunogene therapy to treat MPE (63,64). For intrapleural immunogene therapy, viral vectors are often used, functioning as an 'in-situ vaccination' (10). Based on the expression of the coxsackie-adenovirus receptor on the tumor cell surface, adenovirus-based viral vectors can selectively infect tumor cells (10). After reaching the pleural space, Ad.IFN efficiently transfects tumor cells. As a result, large concentrations of IFN are produced, serving a role in immunity stimulation and tumor inhibition (65).

Another example of the use of intrapleural immunogene therapy for the treatment of MPE is from a phase I study conducted by Aggarwal et al (66). A total of 19 patients with MPE caused by MPM, NSCLC and breast cancer were enrolled to evaluate the tolerability and efficacy of intrapleural gene-mediated cytotoxic immunotherapy (GMCI) (66). GMCI is an immune strategy that consists of two steps: First, an adenovirus-mediated herpes simplex virus thymidine kinase gene (Ad.V-tk) is administered via intrapleural delivery; next, the anti-herpetic drug valacyclovir is administrated the day after Ad.V-tk infusion (66). Following intrapleural injection, Ad.V-tk efficiently transfected tumor cells to express the thymidine kinase gene, resulting in the production of a large amount of nucleotide analogs, which can interrupt normal DNA replication and trigger tumor cell death, consequently releasing tumor neoantigens (66). It has been revealed that tumor neoantigens from dying tumor cells can also induce antitumor immune responses (10). The results of Aggarwal et al (66) demonstrated that GMCI was safe and well tolerated in patients with MPE, with an encouraging efficacy (DCR of 71%).

ICIs

Currently, ICI treatment is the mainstay of anticancer immunotherapy. Agents that target cytotoxic T lymphocyte-associated protein 4 (CTLA-4), such as ipilimumab, and PD-1, such as nivolumab, are two major classes of ICIs (67). PD-1, a receptor on the surface of T cells, binds to its ligand (PD-L1 or PD-L2) to decrease T-cell activity, causing apoptosis in cytotoxic T cells (68). CTLA-4 is a key negative regulator of T-cell responses, which can restrict the antitumor immune response (69). ICIs can discrupt these two immunosuppressive signaling pathways, resulting in improved survival outcomes for patients with solid tumors, such as NSCLC, melanoma and colorectal cancer (70-72).

However, there is currently almost no available data on the response of patients with MPE to ICIs. The efficacy of ICIs in MPE remains to be determined by clinical trials. Recently, two retrospective studies suggested that the presence of MPE in patients is a negative predictor of anti-PD-1 antibody efficacy (73,74). However, these results did not suggest that ICIs are not valid for MPE treatment. In a study by Grosu et al (75), a high concordance in PD-L1 expression was identified between histological specimens and matched pleural fluid from patients with NSCLC, suggesting that, if the primary tumor is sensitive to anti-PD-1 treatment, MPE may also be affected by this treatment. The role of the PD-L1/PD-1 pathway in MPE development has also been explored previously (76). In patients with lung cancer with MPE, Prado-Garcia et al (76) found that tumor-responding CD8+ T cells were not completely differentiated into effector cells, which were negative regulated by PD-L1, so that the PD-L1/PD-1 pathway could promote the dysfunction of tumor-responding T cells from MPE.

Oncolytic virotherapy

As a promising therapeutic modality for the treatment of cancer, oncolytic virotherapy has attracted more attention in recent years. Oncolytic virotherapy infects malignant tissues with tumor-specific viruses, causing the lytic destruction of solid tumors (77). Tumor cell lysis caused by viral infection can induce antitumor immune responses (78). A total of 13 patients with MPM were enrolled in a phase I/IIa trial to receive intrapleural oncolytic herpes simplex virus 1716; the systemic immune responses observed further indicated that an intrapleural oncolytic virus could induce antitumor immune responses, which may serve a role in MPE treatment (79). In a previous preclinical study, oncolytic virotherapy using a tumor-specific vaccinia virus represented a novel and promising treatment modality for the treatment of MPE in tumor mouse models (77). However, clinical evidence on this remains limited. Certain phase I/II clinical trials of oncolytic virotherapy in MPE are ongoing (Table II). NCT01766739 is an ongoing phase I study on the intrapleural administration of GL-ONC1, a genetically modified vaccinia virus, in patients with MPE, aiming to assess the safety and efficacy of this treatment method. NCT03597009 is an ongoing phase I/II trial to evaluate the feasibility of adminstering oncolytic virus talimogene laherparepvec (via intrapleural perfusion) and nivolumab (intravenously) in patients with MPE; it is the first clinical trial to combine oncolytic virotherapy and ICIs in MPE treatment.

Table II

Major ongoing clinical trials of anti angiogenic therapy and immunotherapy in the treatment of MPE.

Table II

Major ongoing clinical trials of anti angiogenic therapy and immunotherapy in the treatment of MPE.

NumberPhaseTumor typeCountryStatusPlanned number of patientsDrugPrimary outcomes
NCT02942043IINSCLCChinaRecruiting87BevacizumabObjective response rate
NCT02054078IIAny tumorChinaNR1831. Bevacizumab
2. Pulvis talci
Changes in chest drainage
NCT02005120IINSCLCChinaNR201. Bevacizumab
2. Recombinant human endostatin
Objective response rate
jRCTs07119001IINSCLCJapanNR15RamucirumabThe pleural effusion control rate at eight week
NCT04123886IAny tumorChinaRecruiting12Recombinant Human TRAIL-Trimer Fusion Protein (SCB-313)Dose limiting toxicity
NCT03869697IAny tumorAustraliaRecruiting16Recombinant Human TRAIL-Trimer Fusion Protein (SCB-313)Dose limiting toxicity
NCT03736122I/IIAny tumorAustraliaNot yet recruiting58BSG-001 (an immune-modulator that primarily exerts its effect via Toll-like receptor)1. Safety and tolerability
2. Change in fluid volume
NCT03597009I/IIMetastatic cancerUSARecruiting241. Talimogene laherparepvec
2. Nivolumab
Phase 1: Treatment related adverse events
Phase II: Resolution of MPE
NCT02429726IIAny tumorChinaNR90Recombinant adenoviral human p53 geneObjective response rate
NCT01997190IAny tumorUSAActive, not recruiting19AdV-tkSafety
NCT01766739ILung cancerUSAActive, not recruiting18GL-ONC1 (a genetically modified vaccinia virus)Maximum tolerated dose
NCT02414269I/IIMalignant pleural diseaseUSARecruiting179Mesothelin-targeted T cellsPhase I: Safety
Phase II: Clinical benefit rate
NCT03054298I Mesothelin-expressing cancerUSAActive, not recruiting-Lentiviral transduced huCART-meso cellsSafety

[i] MPE, malignant pleural effusion; NSCLC, non-small cell lung cancer; TRAIL, TNF-related apoptosis-inducing ligand; AdV-tk, adenovirus-mediated herpes simplex virus thymidine kinase; huCART cells, humanized chimeric antigen receptor-modified T cells; NR, not reported.

Dendritic cell (DC) vaccination

In the immune system, DCs are the dominant antigen-presenting cells (80). DC-based immunotherapy is a promising cancer treatment method in various types of cancer, including MPE (81). A previous study enrolled 8 patients with late-stage lung cancer who were then injected with autologous DCs generated by culturing adherent mononuclear cells from MPE (81). No grade II/III toxicity was observed (81). Following DC vaccination, an increase in T-cell responses against tumor antigens was observed in 6/8 patients (81). Another 5 patients with MPE or malignant ascites, who were resistant to standard chemotherapy, were treated with combined immunotherapy using monocyte-derived DCs, activated lymphocytes and low-dose OK-432 (a streptococcal preparation) (82). Effusion production was decreased in all of the patients and the mean OS time was >9 months (82). Furthermore, the presence of inflammatory DCs (infDCs) in patients with NSCLC with MPE has been recently observed (83). infDCs represent a distinct human DC subset that can induce T helper 1 cell differentiation in the presence of Toll-like receptor agonists (83). This promising finding may provide a new approach for MPE treatment.

CAR - cell treatment

CAR T-cell treatment is another research hotspot in cancer immunotherapy. This technique involves CAR T-cell receptors being specifically engineered to eradicate tumors by recognizing surface proteins expressed on tumor cells (84). In previous studies, CAR T-cell treatment has had substantial clinical success in treating patients with hematological malignancies (85-87). Currently, a growing number of clinical trials of CAR T-cell treatment have focused on solid tumors, targeting surface proteins, including carcinoembryonic antigen, mesothelin, fibroblast activation protein (FAP) and human epidermal growth factor receptor 2 (HER2) (84,88-90). However, to the best of our knowledge, no clinical trial has specifically targeted MPE, despite the local application of CAR T-cell treatment being an attractive approach. A phase I study of intrapleural CAR T cells directed against FAP in patients with MPM with MPE has been completed, but the results have not yet been published (NCT01722149). NCT02414269 and NCT03054298 are two ongoing trials on intrapleural and/or systemic mesothelin CAR T-cell delivery for patients with MPM (Table II). As patients with MPM often present with PE (91), the aforementioned studies will also provide a foundation for controlling MPE via CAR T-cell delivery. FAP and mesothelin are tumor-specific antigens (TSAs) expressed on the surface of MPM cells (92). In order to investigate the efficacy of CAR T-cell delivery for MPE secondary to NSCLC or breast cancer, other targeted TSAs, such as EGFR and HER2, should be investigated.

TIL treatment

TILs are cell clusters with an antigen effect resulting from tumorigenesis (93). T lymphocytes, B lymphocytes and natural killer lymphocytes are the main components of TILs (93). Among them, CD8+ T lymphocytes mainly exert an anticancer activity (94). In a retrospective study, 27 patients with MPE and ascites were treated with either cisplatin (60 mg) or TILs (100 ml) (95). Compared with patients who received cisplatin, patients who received TILs had a higher ORR (83.33 vs. 33.33%) and DCR (71.43 vs. 28.57%), without severe adverse effects (95). Therefore, TILs may represent a promising treatment method for MPE, and should be investigated further.

4. Conclusion and future research direction

The significant progress that has been made in targeted cancer treatment and immunotherapy over the last decade has rendered the identification of novel treatment methods to treat, rather than palliate, MPE. Since angiogenesis serves a key role in MPE development, attention for MPE treatment has inevitably been focused on anti-angiogenic treatments. Patients with MPE have benefited from antiangiogenic agents, including bevacizumab and endostar. However, no relevant prospective phase III trial has, thus far, specifically analyzed the benefit of anti-angiogenic therapies in MPE. In addition, the majority of clinical studies that have focused on this field are from East Asia, which indicates that this treatment strategy for MPE has not yet attracted worldwide attention. The reasons may due to the following aspects: Firstly, bevacizumab and endostar are more often used in East Asia; secondly, East Asia has a large number of patients with MPE that need anti-angiogenic therapy. More clinical data on anti-angiogenic therapies for MPE control are warranted.

The advent of effective immunotherapy with ICIs, as well as adoptive cell therapies for lung cancer and other malignancies, has led to a renewed examination of local and systemic immunotherapies for patients with MPE. Prior strategies, such as cytokine-based immunotherapy, have been successfully used in MPE treatment. However, these strategies have not been as effective as expected. Since MPE has an immunosuppressive microenvironment, strategies involving the activation of the adaptive immune response and inhibition of tumor immune escape mechanisms may serve a role in MPE control. Therefore, treatment with ICIs, CAR T cells, immunogene therapy or oncolytic viruses may be sufficient to turn a dire clinical situation into a therapeutic advantage. Similar to anti-angiogenic therapy, more clinical data on the efficacy and safety of immunotherapy for MPE control are urgently required.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

DH and RD made substantial contributions to the conception and design of the study, assessed the authenticity of the data and wrote the manuscript. QW and LC contributed to the study design and assisted in the literature search for this review article. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Clive AO, Kahan BC, Hooper CE, Bhatnagar R, Morley AJ, Zahan-Evans N, Bintcliffe OJ, Boshuizen RC, Fysh ETH, Tobin CL, et al: Predicting survival in malignant pleural effusion: Development and validation of the LENT prognostic score. Thorax. 69:1098–1104. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Sabang RL, Gandhiraj D, Fanucchi M and Epelbaum O: Role of bevacizumab in the management of the patient with malignant pleural effusion: More questions than answers. Expert Rev Respir Med. 12:87–94. 2018. View Article : Google Scholar

3 

Tao H, Meng Q, Li M, Shi L, Tang J and Liu Z: Outcomes of bevacizumab combined with chemotherapy in lung adenocarcinoma-induced malignant pleural effusion. Thorac Cancer. 9:298–304. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Marquez-Medina D and Popat S: Closing faucets: The role of anti-angiogenic therapies in malignant pleural diseases. Clin Transl Oncol. 18:760–768. 2016. View Article : Google Scholar

5 

Donnenberg AD, Luketich JD, Dhupar R and Donnenberg VS: Treatment of malignant pleural effusions: The case for localized immunotherapy. J Immunother Cancer. 7:110. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Chen YM, Yang WK, Whang-Peng J, Kuo BI and Perng RP: Elevation of interleukin-10 levels in malignant pleural effusion. Chest. 110:433–436. 1996. View Article : Google Scholar : PubMed/NCBI

7 

Thomas R, Cheah HM, Creaney J, Turlach BA and Lee YCG: Longitudinal Measurement of Pleural Fluid Biochemistry and Cytokines in Malignant Pleural Effusions. Chest. 149:1494–1500. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Hooper CE, Elvers KT, Welsh GI, Millar AB and Maskell NA: VEGF and sVEGFR-1 in malignant pleural effusions: Association with survival and pleurodesis outcomes. Lung Cancer. 77:443–449. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Fiorelli A, Vicidomini G, Di Domenico M, Napolitano F, Messina G, Morgillo F, Ciardiello F and Santini M: Vascular endothelial growth factor in pleural fluid for differential diagnosis of benign and malignant origin and its clinical applications. Interact Cardiovasc Thorac Surg. 12:420–424. 2011. View Article : Google Scholar

10 

Murthy V, Katzman D and Sterman DH: Intrapleural immunotherapy: An update on emerging treatment strategies for pleural malignancy. Clin Respir J. 13:272–279. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Stathopoulos GT and Kalomenidis I: Malignant pleural effusion: Tumor-host interactions unleashed. Am J Respir Crit Care Med. 186:487–492. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Giannou AD, Marazioti A, Spella M, Kanellakis NI, Apostolopoulou H, Psallidas I, Prijovich ZM, Vreka M, Zazara DE, Lilis I, et al: Mast cells mediate malignant pleural effusion formation. J Clin Invest. 125:2317–2334. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Nie K, Zhang Z, You Y, Zhuang X, Zhang C and Ji Y: A randomized clinical study to compare intrapleural infusion with intravenous infusion of bevacizumab in the management of malignant pleural effusion in patients with non-small-cell lung cancer. Thorac Cancer. 11:8–14. 2020. View Article : Google Scholar

14 

Sack U, Hoffmann M, Zhao XJ, Chan KS, Hui DSC, Gosse H, Engelmann L, Schauer J, Emmrich F and Hoheisel G: Vascular endothelial growth factor in pleural effusions of different origin. Eur Respir J. 25:600–604. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Lieser EAT, Croghan GA, Nevala WK, Bradshaw MJ, Markovic SN and Mansfield AS: Up-regulation of pro-angiogenic factors and establishment of tolerance in malignant pleural effusions. Lung Cancer. 82:63–68. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Chen Y, Mathy NW and Lu H: The role of VEGF in the diagnosis and treatment of malignant pleural effusion in patients with non small cell lung cancer (Review). Mol Med Rep. 17:8019–8030. 2018.PubMed/NCBI

17 

Gkiozos I, Tsagouli S, Charpidou A, Grapsa D, Kainis E, Gratziou C and Syrigos K: Levels of vascular endothelial growth factor in serum and pleural fluid are independent predictors of survival in advanced non-small cell lung cancer: Results of a prospective study. Anticancer Res. 35:1129–1137. 2015.PubMed/NCBI

18 

Zhang Y, Yu L-K, Lu G-J, Xia N, Xie H-Y, Hu W, Hao K-K, Xu C-H and Qian Q: Prognostic values of VEGF and endostatin with malignant pleural effusions in patients with lung cancer. Asian Pac J Cancer Prev. 15:8435–8440. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Peach CJ, Mignone VW, Arruda MA, Alcobia DC, Hill SJ, Kilpatrick LE and Woolard J: Molecular Pharmacology of VEGF-A Isoforms: Binding and Signalling at VEGFR2. Int J Mol Sci. 19:12642018. View Article : Google Scholar :

20 

Chellappan DK, Leng KH, Jia LJ, Aziz NABA, Hoong WC, Qian YC, Ling FY, Wei GS, Ying T, Chellian J, et al: The role of bevacizumab on tumour angiogenesis and in the management of gynaecological cancers: A review. Biomed Pharmacother. 102:1127–1144. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Assoun S, Brosseau S, Steinmetz C, Gounant V and Zalcman G: Bevacizumab in advanced lung cancer: State of the art. Future Oncol. 13:2515–2535. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Chen DS and Hurwitz H: Combinations of Bevacizumab With Cancer Immunotherapy. Cancer J. 24:193–204. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Bradshaw M, Mansfield A and Peikert T: The role of vascular endothelial growth factor in the pathogenesis, diagnosis and treatment of malignant pleural effusion. Curr Oncol Rep. 15:207–216. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Usui K, Sugawara S, Nishitsuji M, Fujita Y, Inoue A, Mouri A, Watanabe H, Sakai H, Kinoshita I, Ohhara Y, et al North East Japan Study Group: A phase II study of bevacizumab with carboplatin-pemetrexed in non-squamous non-small cell lung carcinoma patients with malignant pleural effusions: North East Japan Study Group Trial NEJ013A. Lung Cancer. 99:131–136. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Tamiya M, Tamiya A, Yamadori T, Nakao K, Asami K, Yasue T, Otsuka T, Shiroyama T, Morishita N, Suzuki H, et al: Phase2 study of bevacizumab with carboplatin-paclitaxel for non-small cell lung cancer with malignant pleural effusion. Med Oncol. 30:676. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Qi N, Li F, Li X, Kang H, Zhao H and Du N: Combination use of paclitaxel and avastin enhances treatment effect for the NSCLC patients with malignant pleural effusion. Medicine (Baltimore). 95:e5392. 2016. View Article : Google Scholar

27 

Du N, Li X, Li F, Zhao H, Fan Z, Ma J, Fu Y and Kang H: Intrapleural combination therapy with bevacizumab and cisplatin for non-small cell lung cancer mediated malignant pleural effusion. Oncol Rep. 29:2332–2340. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Wang Z, Zheng Y and Fang Z: The clinical efficacy and safety of paclitaxel combined with avastin for NSCLC patients diagnosed with malignant pleural effusion. Rev Assoc Med Bras (1992). 64:230–233. 2018. View Article : Google Scholar

29 

Kitamura K, Kubota K, Ando M, Takahashi S, Nishijima N, Sugano T, Toyokawa M, Miwa K, Kosaihira S, Noro R, et al: Bevacizumab plus chemotherapy for advanced non-squamous non-small-cell lung cancer with malignant pleural effusion. Cancer Chemother Pharmacol. 71:457–461. 2013. View Article : Google Scholar

30 

Masago K, Fujimoto D, Fujita S, Hata A, Kaji R, Ohtsuka K, Okuda C, Takeshita J and Katakami N: Response to bevacizumab combination chemotherapy of malignant pleural effusions associated with non-squamous non-small-cell lung cancer. Mol Clin Oncol. 3:415–419. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Chen D, Song X, Shi F, Zhu H, Wang H, Zhang N, Zhang Y, Kong L and Yu J: Greater efficacy of intracavitary infusion of bevacizumab compared to traditional local treatments for patients with malignant cavity serous effusion. Oncotarget. 8:35262–35271. 2017. View Article : Google Scholar :

32 

Jiang L, Li P, Gong Z, Hu B, Ma J, Wang J, Chu H, Zhang L, Sun P and Chen J: Effective Treatment for Malignant Pleural Effusion and Ascites with Combined Therapy of Bevacizumab and Cisplatin. Anticancer Res. 36:1313–1318. 2016.PubMed/NCBI

33 

Song X, Chen D, Guo J, Kong L, Wang H and Wang Z: Better efficacy of intrapleural infusion of bevacizumab with pemetrexed for malignant pleural effusion mediated from nonsquamous non-small cell lung cancer. OncoTargets Ther. 11:8421–8426. 2018. View Article : Google Scholar

34 

Zongwen S, Song K, Cong Z, Tian F and Yan Z: Evaluation of efficacy and safety for bevacizumab in treating malignant pleural effusions caused by lung cancer through intrapleural injection. Oncotarget. 8:113318–113330. 2017. View Article : Google Scholar

35 

Jiang T, Li A, Su C, Li X, Zhao C, Ren S, Zhou C and Zhang J: Addition of bevacizumab for malignant pleural effusion as the manifestation of acquired EGFR-TKI resistance in NSCLC patients. Oncotarget. 8:62648–62657. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Tian P, Shen Y, Feng M, Zhu J, Song H, Wan C, Chen L and Wen F: Diagnostic accuracy of endostatin for malignant pleural effusion: A clinical study and meta-analysis. Postgrad Med. 127:529–534. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Wang J, Sun Y, Liu Y, Yu Q, Zhang Y, Li K, Zhu Y, Zhou Q, Hou M, Guan Z, et al: Results of randomized, multicenter, double-blind phase III trial of rh-endostatin (YH-16) in treatment of advanced non-small cell lung cancer patients. Zhongguo Fei Ai Za Zhi. 8:283–290. 2005.In Chinese. PubMed/NCBI

38 

Jie Wang X, Miao K, Luo Y, Li R, Shou T, Wang P and Li X: Randomized controlled trial of endostar combined with cisplatin/pemetrexed chemotherapy for elderly patients with advanced malignant pleural effusion of lung adenocarcinoma. J BUON. 23:92–97. 2018.PubMed/NCBI

39 

Zhao WY, Chen DY, Chen JH and Ji ZN: Effects of intracavitary administration of Endostar combined with cisplatin in malignant pleural effusion and ascites. Cell Biochem Biophys. 70:623–628. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Biaoxue R, Xiguang C, Hua L, Wenlong G and Shuanying Y: Thoracic perfusion of recombinant human endostatin (Endostar) combined with chemotherapeutic agents versus chemotherapeutic agents alone for treating malignant pleural effusions: A systematic evaluation and meta-analysis. BMC Cancer. 16:888. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Matsumori Y, Yano S, Goto H, Nakataki E, Wedge SR, Ryan AJ and Sone S: ZD6474, an inhibitor of vascular endothelial growth factor receptor tyrosine kinase, inhibits growth of experimental lung metastasis and production of malignant pleural effusions in a non-small cell lung cancer model. Oncol Res. 16:15–26. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Ikuta K, Yano S, Trung VT, Hanibuchi M, Goto H, Li Q, Wang W, Yamada T, Ogino H, Kakiuchi S, et al: E7080, a multi-tyrosine kinase inhibitor, suppresses the progression of malignant pleural mesothelioma with different proangiogenic cytokine production profiles. Clin Cancer Res. 15:7229–7237. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Massarelli E, Onn A, Marom EM, Alden CM, Liu DD, Tran HT, Mino B, Wistuba II, Faiz SA, Bashoura L, et al: Vandetanib and indwelling pleural catheter for non-small-cell lung cancer with recurrent malignant pleural effusion. Clin Lung Cancer. 15:379–386. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Mulder SF, Boers-Sonderen MJ, van der Heijden HFM, Vissers KCP, Punt CJA and van Herpen CML: A phase II study of cediranib as palliative treatment in patients with symptomatic malignant ascites or pleural effusion. Target Oncol. 9:331–338. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Liu M, Shen Y, Ruan M, Li M and Chen L: Notable decrease of malignant pleural effusion after treatment with sorafenib in radioiodine-refractory follicular thyroid carcinoma. Thyroid. 24:1179–1183. 2014. View Article : Google Scholar : PubMed/NCBI

46 

Takemoto S, Fukuda M, Yamaguchi H, Ikeda T, Akagi K, Tomono H, Umeyama Y, Dotsu Y, Taniguchi H, Gyotoku H, et al: Phase II study of ramucirumab and docetaxel for previously treated non-small cell lung cancer patients with malignant pleural effusion: Protocol of PLEURAM study. Thorac Cancer. 11:389–393. 2020. View Article : Google Scholar

47 

Murthy P, Ekeke CN, Russell KL, Butler SC, Wang Y, Luketich JD, Soloff AC, Dhupar R and Lotze MT: Making cold malignant pleural effusions hot: Driving novel immunotherapies. OncoImmunology. 8:e1554969. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Ross SH and Cantrell DA: Signaling and Function of Interleukin-2 in T Lymphocytes. Annu Rev Immunol. 36:411–433. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Minor DR, Moores SP and Chan JK: Prolonged survival after intraperitoneal interleukin-2 immunotherapy for recurrent ovarian cancer. Gynecol Oncol Rep. 22:43–44. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Astoul P, Viallat JR, Laurent JC, Brandely M and Boutin C: Intrapleural recombinant IL-2 in passive immunotherapy for malignant pleural effusion. Chest. 103:209–213. 1993. View Article : Google Scholar : PubMed/NCBI

51 

Castagneto B, Zai S, Mutti L, Lazzaro A, Ridolfi R, Piccolini E, Ardizzoni A, Fumagalli L, Valsuani G and Botta M: Palliative and therapeutic activity of IL-2 immunotherapy in unresectable malignant pleural mesothelioma with pleural effusion: Results of a phase II study on 31 consecutive patients. Lung Cancer. 31:303–310. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Viallat JR, Boutin C, Rey F, Astoul P, Farisse P and Brandely M: Intrapleural immunotherapy with escalating doses of interleukin-2 in metastatic pleural effusions. Cancer. 71:4067–4071. 1993. View Article : Google Scholar : PubMed/NCBI

53 

Suzuki H, Abo S, Kitamura M, Hashimoto M and Izumi K: The intrapleural administration of recombinant interleukin-2 (rIL-2) to patients with malignant pleural effusion: Clinical trials. Surg Today. 23:1053–1059. 1993. View Article : Google Scholar : PubMed/NCBI

54 

Han L, Jiang Q, Yao W, Fu T and Zeng Q: Thoracic injection of low-dose interleukin-2 as an adjuvant therapy improves the control of the malignant pleural effusions: A systematic review and meta-analysis base on Chinese patients. BMC Cancer. 18:725. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Hu CY, Zhang YH, Wang T, Chen L, Gong ZH, Wan YS, Li QJ, Li YS and Zhu B: Interleukin-2 reverses CD8(+) T cell exhaustion in clinical malignant pleural effusion of lung cancer. Clin Exp Immunol. 186:106–114. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Wu X-Z, Zhai K, Yi F-S, Wang Z, Wang W, Wang Y, Pei X-B, Shi X-Y, Xu L-L and Shi HZ: IL-10 promotes malignant pleural effusion in mice by regulating TH 1- and TH 17-cell differentiation and migration. Eur J Immunol. 49:653–665. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Lu Y, Lin H, Zhai K, Wang X, Zhou Q and Shi H: Interleukin-17 inhibits development of malignant pleural effusion via interleukin-9-dependent mechanism. Sci China Life Sci. 59:1297–1304. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Li S, You W-J, Zhang J-C, Zhou Q and Shi H-Z: Immune Regulation of Interleukin-27 in Malignant Pleural Effusion. Chin Med J (Engl). 128:1932–1941. 2015. View Article : Google Scholar

59 

Li Q, Sun W, Yuan D, Lv T, Yin J, Cao E, Xiao X and Song Y: Efficacy and safety of recombinant human tumor necrosis factor application for the treatment of malignant pleural effusion caused by lung cancer. Thorac Cancer. 7:136–139. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Antoniou KM, Ferdoutsis E and Bouros D: Interferons and their application in the diseases of the lung. Chest. 123:209–216. 2003. View Article : Google Scholar : PubMed/NCBI

61 

Goldman CA, Skinnider LF and Maksymiuk AW: Interferon instillation for malignant pleural effusions. Ann Oncol. 4:141–145. 1993. View Article : Google Scholar : PubMed/NCBI

62 

Sartori S, Tassinari D, Ceccotti P, Tombesi P, Nielsen I, Trevisani L and Abbasciano V: Prospective randomized trial of intrapleural bleomycin versus interferon alfa-2b via ultrasound-guided small-bore chest tube in the palliative treatment of malignant pleural effusions. J Clin Oncol. 22:1228–1233. 2004. View Article : Google Scholar : PubMed/NCBI

63 

Sterman DH, Recio A, Carroll RG, Gillespie CT, Haas A, Vachani A, Kapoor V, Sun J, Hodinka R, Brown JL, et al: A phase I clinical trial of single-dose intrapleural IFN-beta gene transfer for malignant pleural mesothelioma and metastatic pleural effusions: High rate of antitumor immune responses. Clin Cancer Res. 13:4456–4466. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Sterman DH, Recio A, Haas AR, Vachani A, Katz SI, Gillespie CT, Cheng G, Sun J, Moon E, Pereira L, et al: A phase I trial of repeated intrapleural adenoviral-mediated interferon-beta gene transfer for mesothelioma and metastatic pleural effusions. Mol Ther. 18:852–860. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Sterman DH, Alley E, Stevenson JP, Friedberg J, Metzger S, Recio A, Moon EK, Haas AR, Vachani A, Katz SI, et al: Pilot and Feasibility Trial Evaluating Immuno-Gene Therapy of Malignant Mesothelioma Using Intrapleural Delivery of Adenovirus-IFNa Combined with Chemotherapy. Clin Cancer Res. 22:3791–3800. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Aggarwal C, Haas AR, Metzger S, Aguilar LK, Aguilar-Cordova E, Manzanera AG, Gômez-Hernandez G, Katz SI, Alley EW, Evans TL, et al: Phase I Study of Intrapleural Gene-Mediated Cytotoxic Immunotherapy in Patients with Malignant Pleural Effusion. Mol Ther. 26:1198–1205. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Hwang WL, Pike LRG, Royce TJ, Mahal BA and Loeffler JS: Safety of combining radiotherapy with immune-checkpoint inhibition. Nat Rev Clin Oncol. 15:477–494. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Lehrer EJ, McGee HM, Peterson JL, Vallow L, Ruiz-Garcia H, Zaorsky NG, Sharma S and Trifiletti DM: Stereotactic Radiosurgery and Immune Checkpoint Inhibitors in the Management of Brain Metastases. Int J Mol Sci. 19:30542018. View Article : Google Scholar :

69 

Kamath SD and Kumthekar PU: Immune Checkpoint Inhibitors for the Treatment of Central Nervous System (CNS) Metastatic Disease. Front Oncol. 8:414. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Brahmer JR, Tykodi SS, Chow LQM, Hwu W-J, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 366:2455–2465. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Govindan R, Szczesna A, Ahn M-J, Schneider C-P, Gonzalez Mella PF, Barlesi F, Han B, Ganea DE, Von Pawel J, Vladimirov V, et al: Phase III Trial of Ipilimumab Combined With Paclitaxel and Carboplatin in Advanced Squamous Non-Small-Cell Lung Cancer. J Clin Oncol. 35:3449–3457. 2017. View Article : Google Scholar : PubMed/NCBI

72 

Topalian SL, Drake CG and Pardoll DM: Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell. 27:450–461. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Shibaki R, Murakami S, Shinno Y, Matsumoto Y, Goto Y, Kanda S, Horinouchi H, Fujiwara Y, Motoi N, Yamamoto N, et al: Malignant pleural effusion as a predictor of the efficacy of anti-PD-1 antibody in patients with non-small cell lung cancer. Thorac Cancer. 10:815–822. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Kawachi H, Tamiya M, Tamiya A, Ishii S, Hirano K, Matsumoto H, Fukuda Y, Yokoyama T, Kominami R, Fujimoto D, et al: Association between metastatic sites and first-line pembrolizumab treatment outcome for advanced non-small cell lung cancer with high PD-L1 expression: A retrospective multicenter cohort study. Invest New Drugs. 38:211–218. 2020. View Article : Google Scholar

75 

Grosu HB, Arriola A, Stewart J, Ma J, Bassett R, Hernandez M, Ost D and Roy-Chowdhuri S: PD-L1 detection in histology specimens and matched pleural fluid cell blocks of patients with NSCLC. Respirology. 24:1198–1203. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Prado-Garcia H, Romero-Garcia S, Puerto-Aquino A and Rumbo-Nava U: The PD-L1/PD-1 pathway promotes dysfunction, but not 'exhaustion', in tumor-responding T cells from pleural effusions in lung cancer patients. Cancer Immunol Immunother. 66:765–776. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Weibel S, Hofmann E, Basse-Luesebrink TC, Donat U, Seubert C, Adelfinger M, Gnamlin P, Kober C, Frentzen A, Gentschev I, et al: Treatment of malignant effusion by oncolytic virotherapy in an experimental subcutaneous xenograft model of lung cancer. J Transl Med. 11:106. 2013. View Article : Google Scholar : PubMed/NCBI

78 

Andtbacka RHI, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, et al: Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J Clin Oncol. 33:2780–2788. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Danson SJ, Conner J, Edwards JG, Blyth KG, Fisher PM, Muthana M, Salawu A, Taylor F, Hodgkinson E, Joyce P, et al: Oncolytic herpesvirus therapy for mesothelioma - A phase I/IIa trial of intrapleural administration of HSV1716. Lung Cancer. 150:145–151. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Reardon DA and Mitchell DA: The development of dendritic cell vaccine-based immunotherapies for glioblastoma. Semin Immunopathol. 39:225–239. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Chang G-C, Lan H-C, Juang S-H, Wu Y-C, Lee H-C, Hung Y-M, Yang H-Y, Whang-Peng J and Liu K-J: A pilot clinical trial of vaccination with dendritic cells pulsed with autologous tumor cells derived from malignant pleural effusion in patients with late-stage lung carcinoma. Cancer. 103:763–771. 2005. View Article : Google Scholar : PubMed/NCBI

82 

Morisaki T, Matsumoto K, Kuroki H, Kubo M, Baba E, Onishi H, Tasaki A, Nakamura M, Inaba S and Katano M: Combined immunotherapy with intracavital injection of activated lymphocytes, monocyte-derived dendritic cells and low-dose OK-432 in patients with malignant effusion. Anticancer Res. 23:4459–4465. 2003.PubMed/NCBI

83 

Gu F-F, Wu J-J, Liu Y-Y, Hu Y, Liang J-Y, Zhang K, Li M, Wang Y, Zhang Y-A and Liu L: Human inflammatory dendritic cells in malignant pleural effusions induce Th1 cell differentiation. Cancer Immunol Immunother. 69:779–788. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Akhavan D, Alizadeh D, Wang D, Weist MR, Shepphird JK and Brown CE: CAR T cells for brain tumors: Lessons learned and road ahead. Immunol Rev. 290:60–84. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, et al: Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 371:1507–1517. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, Boesteanu AC, Wang Y, O'Connor RS, Hwang WT, et al: Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 24:563–571. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Heng G, Jia J, Li S, Fu G, Wang M, Qin D, Li Y, Pei L, Tian X, Zhang J, et al: Sustained Therapeutic Efficacy of Humanized Anti-CD19 Chimeric Antigen Receptor T Cells in Relapsed/Refractory Acute Lymphoblastic Leukemia. Clin Cancer Res. 26:1606–1615. 2020. View Article : Google Scholar

88 

Junghans RP, Ma Q, Rathore R, Gomes EM, Bais AJ, Lo AS, Abedi M, Davies RA, Cabral HJ, Al-Homsi AS, et al: Phase I Trial of Anti-PSMA Designer CAR-T Cells in Prostate Cancer: Possible Role for Interacting Interleukin 2-T Cell Pharmacodynamics as a Determinant of Clinical Response. Prostate. 76:1257–1270. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, Liu E, Dakhova O, Ashoori A, Corder A, et al: Human Epidermal Growth Factor Receptor 2 (HER2) -Specific Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of HER2-Positive Sarcoma. J Clin Oncol. 33:1688–1696. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Beatty GL, O'Hara MH, Lacey SF, Torigian DA, Nazimuddin F, Chen F, Kulikovskaya IM, Soulen MC, McGarvey M, Nelson AM, et al: Activity of Mesothelin-Specific Chimeric Antigen Receptor T Cells Against Pancreatic Carcinoma Metastases in a Phase 1 Trial. Gastroenterology. 155:29–32. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Scherpereel A, Grigoriu BD, Noppen M, Gey T, Chahine B, Baldacci S, Trauet J, Copin MC, Dessaint JP, Porte H, et al: Defect in recruiting effector memory CD8+ T-cells in malignant pleural effusions compared to normal pleural fluid. BMC Cancer. 13:3242013. View Article : Google Scholar : PubMed/NCBI

92 

Nicolini F, Bocchini M, Bronte G, Delmonte A, Guidoboni M, Crino L and Mazza M: Malignant Pleural Mesothelioma: State-of-the-Art on Current Therapies and Promises for the Future. Front Oncol. 9:15192020. View Article : Google Scholar : PubMed/NCBI

93 

Maibach F, Sadozai H, Seyed Jafari SM, Hunger RE and Schenk M: Tumor-Infiltrating Lymphocytes and Their Prognostic Value in Cutaneous Melanoma. Front Immunol. 11:21052020. View Article : Google Scholar : PubMed/NCBI

94 

Wouters MCA and Nelson BH: Prognostic Significance of Tumor-Infiltrating B Cells and Plasma Cells in Human Cancer. Clin Cancer Res. 24:6125–6135. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Chu H, Du F, Gong Z, Lian P, Wang Z, Li P, Hu B, Chi C and Chen J: Better Clinical Efficiency of TILs for Malignant Pleural Effusion and Ascites than Cisplatin Through Intrapleural and Intraperitoneal Infusion. Anticancer Res. 37:4587–4591. 2017.PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 58 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He D, Ding R, Wen Q and Chen L: Novel therapies for malignant pleural effusion: Anti‑angiogenic therapy and immunotherapy (Review). Int J Oncol 58: 359-370, 2021
APA
He, D., Ding, R., Wen, Q., & Chen, L. (2021). Novel therapies for malignant pleural effusion: Anti‑angiogenic therapy and immunotherapy (Review). International Journal of Oncology, 58, 359-370. https://doi.org/10.3892/ijo.2021.5174
MLA
He, D., Ding, R., Wen, Q., Chen, L."Novel therapies for malignant pleural effusion: Anti‑angiogenic therapy and immunotherapy (Review)". International Journal of Oncology 58.3 (2021): 359-370.
Chicago
He, D., Ding, R., Wen, Q., Chen, L."Novel therapies for malignant pleural effusion: Anti‑angiogenic therapy and immunotherapy (Review)". International Journal of Oncology 58, no. 3 (2021): 359-370. https://doi.org/10.3892/ijo.2021.5174