Back to Journals » Cancer Management and Research » Volume 17

Progress in Mechanistic Research and the Use of Traditional Chinese Medicine in Treating Malignant Pleural Effusion

Authors Gong F, Cheng Y, Li Y, Fan Q, Qu R, Fan T, Lv Y, Dai J 

Received 20 April 2025

Accepted for publication 30 June 2025

Published 15 July 2025 Volume 2025:17 Pages 1377—1388

DOI https://doi.org/10.2147/CMAR.S529467

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Lu-Zhe Sun



Fengying Gong,1,2 Yunshui Cheng,1 Yongchun Li,1 Qin Fan,3 Rongmei Qu,4 Tingyu Fan,4 Ying Lv,1 Jingxing Dai4

1Department of Traditional Chinese Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, People’s Republic of China; 2The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, People’s Republic of China; 3School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People’s Republic of China; 4Guangdong Provincial Key Laboratory of Medical Biomechanics and Guangdong Engineering Research Center for Translation of Medical 3D Printing Application and National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, People’s Republic of China

Correspondence: Jingxing Dai, Email [email protected]; [email protected] Ying Lv, Email [email protected]

Abstract: Malignant pleural effusion (MPE) is characterized by the accumulation of fluid in the chest cavity, secondary to metastasis from a primary pleural tumor or malignant neoplasms originating from other anatomical sites. MPE is associated with a poor prognosis. Consequently, timely and effective prevention and management of MPE are critical. In Western medicine, the treatment of MPE primarily involves procedures such as surgical puncture for drainage, pleural fixation, chemotherapy, and targeted therapy. In contrast, traditional Chinese medicine (TCM) offers therapeutic modalities including oral decoctions, thoracic perfusion with herbal injections, topical applications of medicinal pastes, and acupoint therapies. The TCM approaches have demonstrated satisfactory clinical outcomes. Advances in the study of TCM for managing MPE in lung tumors are expected to yield a wealth of therapeutic strategies, facilitating the development of more optimized clinical treatments.

Keywords: malignant pleural effusion, mechanism, prevention and management, traditional Chinese medicine, lung tumors

Introduction

Pleural effusion (PE) refers to the abnormal accumulation of fluid in the pleural cavity. Potential etiologies encompass cardiac insufficiency, malignant tumors, tuberculous pleurisy, pneumonia, liver cirrhosis, and nephrotic syndrome, among others. Based on the characteristics of pleural effusion, it can be categorized into exudative, transudative (serous or hemorrhagic), hemothorax, empyema, and chylothorax. A critical challenge in the differential diagnosis of pleural effusion is the early identification and differentiation between benign and malignant types. Early intervention for benign pleural effusion can lead to successful resolution and minimize the risk of complications. Meanwhile, early management of malignant pleural effusion (MPE) can enhance the quality of life and improve survival rates in patients with advanced malignancies.1,2

The objective of this paper is to present a thorough and up-to-date overview of the pathogenesis of MPE and the use of traditional Chinese medicine (TCM) for the treatment and management of MPE. We expect that this review will help clinicians personalize healthcare for patients with MPE and improve the survival and quality of life of patients with cancer.

Profile of Malignant Pleural Effusion

Malignant pleural effusion occurs because of a primary pleural malignancy or metastasis of a malignant tumor to the pleura from another site. The diagnosis of MPE can be confirmed by detecting malignant tumor cells in a pleural effusion sample or pleural biopsy tissue.3 Almost all malignant tumors can cause MPE as a complication with disease progression.4 The incidence of MPE associated with lung cancer (35%), breast cancer (23%), and lymphoma (10%) has been reported to exceed 60%.5 Median patient survival in such cases is only 3–12 months.6 Clinical management of MPE involves several aspects, including local and systemic therapies such as thoracentesis, chest tube drainage, pleural fixation, chemotherapy, and targeted therapy.7 However, these remedies have limited effectiveness.

Physiologic Significance of Pleural Effusion

The normal chest cavity contains approximately 0.26 mL/kg of liquid, which acts as a lubricant in the chest space to reduce the friction from the wall to the guts during respiration and maintain negative pressure to avoid lung collapse. The volume of pleural fluid depends on hydrostatic pressure, mesothelial cell activity, and lymphatic drainage.8,9 In a healthy body, luminal fluid secretion and absorption are always maintained in a state of equilibrium. Fluid accumulation occurs if fluid production exceeds the ability of the lymphatic vessels to absorb it, due to factors such as increased production, decreased absorption, or both.10 Consequently, when the amount of pleural fluid produced is greater than the amount absorbed, the fluid accumulates in the chest cavity.

Pathogenesis and Molecular Mechanisms of MPE

The etiology of MPE has not been fully elucidated to date. One common mechanism leading to the occurrence of MPE is the blockage of the nodes of the mural pleura and the resultant lymphatic obstruction caused by the growth and proliferation of tumor cells into the thoracic cage.11 Primary or metastatic lesions of the pleura and inflammatory stimuli can lead to increased capillary permeability and abnormal angiogenesis, causing the occurrence of PEs.7 A large number of cytokines and chemokines have been implicated in the formation of malignant effusions, including vascular endothelial growth factor (VEGF),12 interleukin (IL)-1β,13 transforming growth factor (TGF)-β1,14 IL-6, IL-8, and IL-10.15 MPE also contains a host of immunologic cells and cytokines with immunosuppressive properties, which contribute to tumor immune escape and promote pleural tumor growth and MPE formation.16

Direct Invasion or Hematogenous Metastasis

The pleura may be penetrated by lymphatic spread and/or invasion of tissue lesions in the diaphragm, pericardium, chest wall, and other adjacent organs. Autopsy data show that tumor cells disperse into the pleura predominantly via blood circulation and first invade the visceral pleura. As a result, tumor cells may spread in an adherent manner or as detached cancer cells floating in fluid secondary to pleural spread.17 Tumor cells adhere to the pleural mesh, bypass the pleural immunity mechanism, and infiltrate pleural tissue to access nourishment and growth factors upon reaching the pleural wall layer. In patients with MPE, impaired macrophage and lymphocyte function, along with the production of pro-inflammatory factors and pro-tumor mediators, create complex interactions between cancer cells and their hosts. These processes contribute to an immune-suppressing microenvironment within the pleura. Even though they are not attached to pleural structures, floating tumor cells can generate lesions elsewhere in the pleural cavity, indicating that malignant tumors can enter the pleura through direct invasion or hematogenous metastasis and disrupt the pleural barrier function to cause PE.16

Involvement of Lymphatic Vessels

Lymphatic vessels constitute an important system involved in fluid reflux. Fluid arrives in the cavity through the pleural wall capillaries, and since the pressure slope for the motion of fluid in the plenum is almost zero, peritoneal effluent is primarily aspirated by lymph vessels through the lymphatic micropores of the mural pleura.11 Obstruction of lymphatic drainage in the mural pleura is the primary mechanism underlying the formation of MPE. This obstruction occurs through a combination of compression by the lung cancer mass and metastasis through the lymphatic channels, thus causing MPE.18

Role of Angiogenesis in MPE

The typical clinical symptom of MPE is compression of lung tissue, and the consequent respiratory and circulatory dysfunction seriously affect patients’ quality of life and can even be life-threatening.17 Angiogenesis and vascular remodeling are key pathophysiological stages in the creation of MPE.16 Several investigations have revealed that a hyper-permeable pleural wall or internal vasculature and the tumor vasculature play pivotal roles in the pathobiology of MPE.7,17,19 Pleural tumor cells secrete a diverse array of vasoactive mediators, including angiopoietin and/or VEGF, leading to excessive angiogenesis and invasion of the thoracic cavity. The vascular hyperpermeability associated with pleural malignancy and the anti-inflammatory host reaction induced by tumor cells in the cavity have been linked to the etiology of MPE.20,21

Cytokines Released by Tumor Cells

MPE is a high-protein fluid that accumulates in the presence of cytokines and growth factors with pro-inflammatory, anticancer, and angiogenic characteristics (eg, VEGF) and suppressor factors (eg, IL-10).15,22 This indicates that MPE may create a nutritious microenvironment that favors the growth of tumors while simultaneously inhibiting the immune system’s anti-tumor activity.

Role of Angiogenic Factors in MPE

Angiogenic factors are known to influence the kinetic basis of PE development since they can also induce leakages in the vasculature. Angiogenic cytokines such as basic fibroblast growth factor (bFGF), VEGF, and angiopoietin are likely initiators of MPE, since they have been correlated with the onset of neovascularization, vascular permeability, and bleeding, in the process of inflammation and in tumor progression.23

i) Vascular endothelial growth factor: VEGF is an essential angiogenesis-stimulating factor. Tumors invading the chest wall secrete vast quantities of VEGF, which increases the permeability of blood vessels.24 VEGF levels in patients with MPE have been shown to be significantly higher than those in patients with other diseases such as congestive heart failure or pulmonary tuberculosis.25–28 In one study investigating 79 cases of unilateral PE,29 the VEGF levels were clearly greater in malignant tumors than in benign tumors. In another study, VEGF expression in MPE was 77-fold greater than that in benign PE.30

Furthermore, MPE with elevated pleural VEGF levels has been shown to be associated with significantly lower survival rates than MPE with normal VEGF levels.31,32 The effects of VEGF are primarily mediated by VEGF receptors 1 and 2 (VEGFR-1 and VEGFR-2, respectively) [19]. VEGF is recruited by its receptor and activates downstream signals including protein kinase C and mitogen-activated protein kinase (MAPK), which triggers vascular endothelial proliferation and differentiation, stimulates endothelial apoptosis, and eventually leads to endothelial apertures and loss of barrier cohesion, promoting the initiation of tumor growth and progression of MPE.33,34 Neuropilin 1 (NRP1) co-operates with endothelin and VEGFR-2 in regulating VEGF signal transduction and endothelial cell germination.35 VEGF elicits phosphorylation of adhesion and dense junction components and increases vascular permeability, leading to increased vascular permeability.36

ii) Basic fibroblast growth factor: bFGF is a potent angiogenic factor that promotes endothelial cell growth in vitro and angiogenesis in vivo. bFGF is a known angiogenic factor37 and a mitogenic factor in a variety of cells, including fibroblasts, smooth muscle cells, and endothelial cells. Tumor size and bFGF levels are negatively correlated. bFGF levels in pleural fluid samples from patients with extensive tumor involvement of the pleural surface have been shown to be significantly lower than those from patients with limited involvement of the pleural surface, who showed significantly higher levels of bFGF and underwent successful pleurectomy.38 Moreover, bFGF expression is significantly higher in benign pleural effusions than in MPE, and this difference was particularly evident in malignant pleural mesothelioma and benign inflammatory pleural disease.39

iii) Heparinase: Heparinase is an endoglycosidase that degrades the extracellular matrix (ECM) and cell surface heparan sulfate, playing an important role in tumor metastasis and angiogenesis.40 Heparinase-mediated cleavage of heparan sulfate leads to catabolism of ECM and promotes cellular dissemination associated with tumor metastasis, angiogenesis, and inflammation.41,42 Heparinase is upregulated in almost all human tumors.43 Heparinase levels are associated with all steps of tumor formation, including tumor initiation, growth, metastasis, and chemotherapy resistance.44

Inflammation and Malignant Pleural Fluid

MPE manifests as vascular leakage, leading to pleural space effusion. Its etiology involves not only lymphatic obstruction caused by the tumor but also fulminant inflammation and neovascularization.21 Chronic inflammation promotes malignancy development, progression, metastatic spread, and treatment resistance.7,45 Eosinophils counts are increased in MPE, and these cells promote tumor cell migration and metastasis formation by secreting C-C motif chemokine ligand 6 (CCL6). Furthermore, the inhibition of C-C chemokine receptor 1 (CCR1; a receptor for CCL6), which serves as a receptor for CCL6, has been shown to reduce tumor cell migration and metastasis.46 Additionally, Kirsten rat sarcoma viral oncogene homolog (KRAS) mutant cancer cells act as molecular determinants that increase propensity of pleural metastatic tumor cells to develop MPE.47 Nevertheless, the C-C motif chemokine ligand 2 (CCL2) released by pleural tumors plays a pivotal role in the development of MPE by promoting angiogenesis and vascular permeability as well as facilitating the migration of bone marrow-derived cells, including monocytes and mast cells, from the bone marrow to the metastatic environment within the pleura.6,21,48

Nuclear factor-κB (NF-κB) activity in tumor cells is critical for MPE formation, drives pro-inflammatory gene expression, and promotes pleural tumor cell survival.49–51 Mutant cancer cell KRAS is the molecular culprit in MPE formation. KRAS can establish host-to-tumor signaling circuits by promoting IKKα-mediated tumor cell responsiveness to host IL-1β, ultimately leading to inflammatory MPE development and drug resistance.4 Tumor necrosis factor (TNF)-α is found within the microenvironment of human tumors, including MPE. The MPE model shows host- and tumor-originated TNF-α. Moreover, TNF-α neutralization can remarkably depress tumor proliferation, effusion formation, vascular hyperpermeability, TNF-α and VEGF expression, and angiogenesis, resulting in improved survival.50,51 For the diagnosis of MPE, the levels of TNF-α and interferon-γ in the pleural fluid are of great practical value.2

The mesothelial cells are the major structural axis of inflammation.52 In response to stimulation by hardeners (eg, talc), mesothelial cells secrete a number of mediators, including IL-8 and monocyte chemotactic protein-1 (MCP-1), VEGF, platelet-derived growth factor (PDGF), bFGF, and TGF-β.53 These mediators are key players in several inflammatory pathways. IL-8 belongs to the chemokine cytokine family and is associated with the inflammatory process. IL-8 production is notably increased in response to inflammatory stimulation,54,55 thereby inducing an influx of neutrophils into the pleural space.56 One study has suggested that IL-8 may be engaged in coagulation events that may result from talc pleurodesis, resulting in early death in patients with MPE.57 MCP-1 generated by tumor cells is an intrinsic determinant of their ability to provoke MPE formation. MCP-1 appears to have angiogenic and inflammatory properties. Recent studies suggest that MCP-1 may be an effect mediator of NF-κB involved in the inflammatory cascade response in the pathogenesis of MPE.58 Increased proliferation of tumor cells, evasion of programmed death of tumor cells, sustained formation of neointima, enhanced vascular leakage, and increased macrophage recruitment are possible mechanisms by which MCP-1 promotes MPE.20,59

Immune Cells and MPE

Pleural effusion induced by tumor cells establishes its own immune microenvironment during the progression of formation. The tumor microenvironment can be metabolically reprogrammed by immune cells,60 which reduces immune activation signals and downregulates antigen recognition and presentation. This subsequently induces immunosuppressive effects within MPE environment,61 thereby facilitating the growth of disseminated tumor cells and promoting immune escape in MPE. Various immune cell populations within tumor microenvironment of malignant pleural effusion, including T lymphocyte subsets, tumor-associated macrophages, dendritic cells, natural killer cells, myeloid-derived suppressor cells, and B lymphocytes are subject to immunosuppressive effects mediated by MPE. Consequently, this facilitates the unrestricted growth of tumor cells in MPE that have evaded immune surveillance.62 Complex interactions between immune cells are essential in the pathogenesis of MPE. Single-cell analysis of multiple immunophenotypes of MPE have shown that immune cells in MPE are enriched for transcriptional signatures of macrophages, dendritic cells, B cells, and functional T cells.63 Th17 cells and their cytokines are important elements in many diseases, especially MPE. MPE is mediated by the activation of Th17 cells and their interactions with other immune cells. In addition, Th17 cells and their pro-differentiation cytokines have been proposed to be recruited into the development of MPE by mediating tumor-promoting and anti-tumor functions, and modulating the production and differentiation of Th17 cells in MPE.64

As the main immune cells in the cancer microenvironment, tumor-associated macrophages represent the major inflammatory cells in MPE, and the M2 macrophage number can reflect poor prognoses in these patients.65 Tumor-associated macrophages represent the predominant inflammatory cell population in MPE, with M2 polarization being induced by signaling through the colony-stimulating factor 1 (CSF1)/CSF1 receptor (CSF1R) axis. CSF1R+ macrophages facilitate the onset of PE by increasing vessel perfusion, destabilizing the vasculature, and/or fostering an immunosuppressive environment.65 Macrophages in malignant ascites and MPE can transform into fibroblastic-like elements.66 The results of single-cell RNA-sequencing analyses shed light on the immune-related milieu of MPE with small cell lung cancer (SCLC). B cells and T cells exhibit varying degrees of expression of tumor-associated genes pertinent to SCLC during the differentiation process of MPE. Furthermore, immune cells are primarily involved in the communication with malignant cells by means of major histocompatibility complex II (MHCII), IL-16, lectin, and amyloid precursor protein pathways.67

Tumor-Associated Fibroblasts and MPE

The presence of MPE is indicative of advanced cancer and is associated with a poor prognosis that develops because of complex orchestrated interactions among tumor cells, inflammatory cells, as well as the vasculature. Studies of the cancer and stromal cell secretome have provided valuable insights into the interactions of cells in the malignant environment as well as the crucial role of critical target effectors in the creation of premetastatic sites at remote metastatic locations.68 Cancer-associated fibroblasts represent crucial stromal constituents in primary and diffuse pleural lesions.69 During tumorigenesis, the imbalance of cell homeostasis and the high heterogeneity of cancer-associated fibroblasts have major effects on the protein pool secreted by malignant tumor cells.70

Adenocarcinoma (AdC), which is often accompanied by PE, is the most prevalent subtype of carcinoma. Lumican is a sulfated kitin that belongs to the SLRP group of ECM proteins and is expressed in various forms in a wide range of organs and tissues, including skin, corneas, bones, arteries, kidneys, and lungs. Alterations in lumican expression are thought to be associated with the spread of cancer.71,72 The study by Cappellesso et al,73 showed that in AdC surgical specimens, lumican expression was lower within cancer cells and higher and more diffuse throughout the matrix surrounding the neoplastic tumor. Western blot analysis indicated that the high fluorescein values of AdC PEs may have been released by fibroblasts surrounding the tumor. PDGF levels in PEs from lung adenocarcinoma are significantly greater than that those in PEs associated with SCLC and non-malignant conditions, and this finding correlates with fibroblast proliferation and pro-activity.74

Hypoxia-Inducible Factor and MPE

Repeated MPE caused by NSCLC is difficult to cure with regular therapy. In an extensive MPE cohort study, patients with recurrent MPE showed significantly increased Claudin-4 (CLDN4) expression in PE cells and poorer overall survival.75 CLDN4 expression is associated with hypoxia-inducible factor 1-α (HIF1-α) and VEGF-A. The HIF1-α (P) and CD133/HIF1-α (S) ratios have been shown to be markers for the assessment of lymph node metastasis in patients with MPE.76

Water Channel Protein and MPE

Increased vascular permeability in the chest wall and an imbalance in the production and absorption of pleural liquid in patients with lung cancer may be the principal cause of the emergence of MPE. Several studies have implicated aquaporin-1 (AQP1) in this pathological mechanism.77,78 Aquaporins (AQPs) are a family of transmembrane-bound proteins that facilitate transmembrane and transseptal permeability across cellular membranes, playing a crucial role in water transport and metabolic processes in vivo.79 Among the six aquaporins identified in the lungs, AQP1 has been most comprehensively studied.80–82 AQP1 is exposed on the sural surface of the visual and parietal pleura. It plays a critical role in pulmonary edema and severe lung damage, since AQP1 deficiency leads to impaired fluid exchange between pulmonary capillaries and interstitial tissue. PE is mainly absorbed by the lymph vessels in the upper parietal pleura. The fundamental function of AQP1 is regulation of fluid homeostasis within the thoracic cavity. Research involving AQP1-knockout mice demonstrated that the time required for osmotic pressure in the pleural cavity to achieve equilibrium was significantly longer than that in wild-type mice. In vitro investigations have indicated that inhibition of AQP1 can influence the osmotic pressure associated with pleural fluid transport.77,78

These studies underscore the critical role of AQP1 in maintaining pleural fluid homeostasis. Furthermore, AQP1 has been implicated in tumorigenesis, with evidence indicating that this protein is significantly overexpressed in various malignancies, including lung cancer.83,84 Inhibition of AQP1 has been demonstrated to impede the invasion and progression of various tumors, while quantitative transcriptome analysis of AQPs showed that increased transcript ratios of AQP1, AQP3, AQP5, and AQP9 were associated with poor survival.85,86 Increased levels of AQP1 mRNA as well as VEGF expression have also been shown to be correlated with elevated MPE volumes.87

Advancements in the Application of TCM for the Management of MPE

PE is not simply a byproduct of malignancy, but also an energetic driver of tumor growth, necessitating changes in the treatment paradigm of MPE to aggressively control PE formation. These changes are vital to lengthen patients’ survival and enhance their quality of life. At present, the management of MPE in Western medicine is primarily based on symptomatic treatment and palliative care, which can still achieve some efficacy in the early stages. For large or rapidly recurring PEs, only puncture aspiration is possible, but repeated maneuvers can cause disturbances in water and electrolyte balance and protein loss and may also increase the danger of pleural infection.

Combinations of Chinese and Western medicine have been widely recognized for treating MPE. TCM treatment can boost the immune system, alleviate complaints, and increase the quality of life with fewer adverse reactions, making the treatment more acceptable to patients. This approach is the preferred option for patients who cannot tolerate chemotherapy.88–90 Numerous clinical trials have shown favorable efficacy of this approach.91,92

1) Formulas: Formulas are prescriptions for combinations of plants/minerals used by TCM practitioners based on their clinical experience to enhance therapeutic efficacy and minimize adverse effects. TCM practitioners use formulas containing multiple herbs to harmonize their effects and restore the patient’s “balance”.93

i) Gansuigancao Tang: Gansuigancao Tang (GSGCT) is a TCM soup that consists of two herbs, Gansui and Gancao, and is uniquely effective in the treatment of edema. Among the two constituent herbs, Gansui promotes urinary excretion and is prescribed in cases of swelling, abdominal ascites, and asthma. Gansui has also shown diuretic effects and has been reported to drain MPE (P < 0.01).94 Gansui has a therapeutic effect on MPE, and combining it with licorice can diminish its efficacy and even produce severe toxicity in certain proportions. Gansui:Gancao at a ratio of 2:1–1:1 is considered a dose-toxicity-efficacy inflection point. At ratios within this point, GSGCT has a curative therapeutic effect on MPE with minimal toxicity. However, at ratios higher than this point, GSGCT can cause liver and cardiac toxicity, which is detrimental to the efficacy of the treatment.

ii) Xiaoshui decoction: Xiaoshui decoction (XSD) is a herbal compound adapted from the classic formula for the treatment of “Hanging Drinks” in The Essentials of the Golden Poverty-Tinglidazaoxiefei Decoction. XSD is often utilized in MPE management. XSD has shown efficacy in cleaning the lungs and removing phlegm, facilitating horizontal wheezing, and reducing the retention of fluid. One study95 showed that XSD effectively inhibited the evolution of MPE through decreased pleural leakage and angle formation and reprogramming the tumor-associated macrophage phenotype by inducing autophagy.

iii) Shizao tang: Shizao tang (SZT), which is used for the treatment of typhoid fever, is composed of Coriander, Glycyrrhiza glabra, Dacryocybe, and Jujubae. SZT is prescribed for the healing of edema, ascites, pleural effusions, and other disorders where fluid balance is impaired. Euphorbia kansui and Euphorbia pekinensis have been found to have diuretic and laxative effects on improving water retention in rats with malignant ascites.96 The results of network pharmacological analysis suggested that SZT limited MPE by modulating the VEGF-phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway and altering vascular permeability and through modulation of the PI3K-Akt/MAPK/NF-κB pathway by upregulating AQP8 protein production. In animal studies, SZT/VSZT significantly reduced PE, increased urinary output, decreased inflammatory cytokine expression, and elevated AQP8 levels (P < 0.05).97

iv) Ginseng and Astragalus Decoction: Ginseng and Astragalus Decoction (GAD) has been reported to help replenish Qi and Yang.98 GAD was recorded to be used in Ge Hong’s Elbow Preparedness Formula of the Eastern Jin Dynasty, and subsequently appeared in classic formulas for the treatment of water disorders, including Baoyuan Tang, Buzhongyiqi Tang and Guizhi Tang. Recent studies in modern pharmacology have shown that astragalus and ginseng have anti-tumor and immunological actions. One study utilized network pharmacology and molecular-docking technology to explore the potential compounds and activities of GAD for the treatment of MPE. The target core network included 22 key compounds, 26 main targets, and 16 signal paths of GAD. Molecular-docking analyses revealed six partial docking targets to ginsenoside RH2, kaempferol, mangostin, fructone A, and quercetin. Multiple C-components of GAD targeting signaling pathways are useful for treating MPE.99

Purification

  • i) Kanglaite injection: The major component of Kanglaite injection (KLT) is Coix seed oil, which is emulsified by modern technology. KLT, which is recognized as a broad-spectrum anticancer agent, has been employed in China for the treatment of malignant tumors.100 One study101 reported that KLT administered at a dosage of 200 mL/day for 21 consecutive days resulted in a remarkable increase of 55.9% in the mean body weight of patients with advanced lung cancer in comparison with their pre-treatment weight. KLT has been shown to increase anti-tumor activity in clinical trials, improving patients’ quality of life and reducing side effects.102,103 KLT combined with conventional chemotherapeutic agents has shown superior efficacy and safety in controlling MPE in comparison with intrapleural injection of conventional chemotherapeutic agents alone.92
  • ii) Aidi injection: Aidi injection consists of xanthoxylin, ginsenoside, astragaloside, and artemisinin saponin E.104 As an important adjuvant for chemotherapy, Aidi injection is frequently used in conjunction with gemcitabine and cisplatin to manage MPE in NSCLC. Research has demonstrated that the clinical effectiveness of Aidi injection alone administered into the pleural cavity is comparable to that of cisplatin. The combination of Aidi injection and cisplatin has been shown to substantially enhance patients’ complete remission rates and quality of life while demonstrating a reduction in pleural fixation breakdown, disease evolution, and secondary events. For moderate-to-severe bruises, the combination of Aidi injection and cisplatin has been shown to significantly enhance clinical symptoms as well as improve Karnofsky performance status scores by ≥50 or the expected survival duration by ≥3 months. Evidence indicates that Aidi injection can perform a major clinical function in the management of MPE.105
  • iii) Elemene injection: β-Elemene extracted from a species of ginger is processed into elemene injection (EI),106 which has been widely used as an adjuvant anti-tumor drug in numerous countries.107 The use of EI alone has been found to significantly improve the objective response rate (ORR) in MPE and reduce the rate of bladder cancer recurrence, while also resulting in fewer adverse reactions. EI is emerging as an efficacious proprietary drug to treat MPE with non-inferior efficacy and less toxicity than conventional drugs.108
  • External Medicines

    Over the past few years, TCM-based external medicines have played an essential part in MPE treatment due to their distinctive benefits.88 Glauberite is a commonly used Chinese medicine, and its main component is sodium sulfate (Na2SO4·10H2O). Glauberite can osmotically soak up the effusion to help remove edema. When applied externally to the chest, glauberite can create a local hyperosmotic state and stimulate the absorption of excess effusion through osmotic pressure. Mirabilite is used to treat many clinical conditions, including inflammation and edema; it has also been used for wound healing and intestinal preparation before microscopic examination.109 Rhubarb has been shown to clear heat and detoxify inflammation.110 Rhubarb has the potential to cleanse the intestines, alleviate internal abdominal pressure, mitigate endotoxin-induced intestinal mucosal damage, decrease pancreatic amylase secretion, resolve inflammatory edema, and reduce serum levels of IL-6.111 The synergistic action of mirabilite and rhubarb can relieve congestion and reduce swelling. Topical application of glauberite and rhubarb, in conjunction with thoracic perfusion of cisplatin, has been shown to be an effective approach for treating MPE. The underlying mechanism of this combined treatment may involve the reduction of capillary permeability, enhancement of microcirculation barriers, facilitation of a hypertonic state, and osmotic suction of fluid in the chest pleura, thereby promoting resolution of MPE.112

    Conclusions

    The presence of MPE often indicates severe disease, with the rapid accumulation of PE mechanically restricting lung expansion and causing considerable damage to the patient’s respiratory and circulatory systems. In Western medicine, symptomatic management and palliative care are frequently employed for the treatment of MPE. Enhancing symptom management, improving quality of life, and extending survival are the primary goals of MPE treatment. TCM has demonstrated distinctive advantages in achieving these goals. The combination of TCM and Western medicine represents a notable trend in advancing MPE treatment. The integration of local medicine with internal administration of TCM has demonstrated a notable auxiliary therapeutic effect. Treatment protocols should prioritize individualized approaches tailored to the patient characteristics and stages of illness.

    Currently, the research level of TCM regarding MPE requires enhancement. The diagnostic and therapeutic criteria, research methodologies, and statistical analyses of results require further standardization to bolster the credibility of these scientific results (Figure 1).

    Figure 1 A Mermaid mind map representation of the mechanistic research and traditional Chinese medicine (TCM) in the treatment of malignant pleural effusion (MPE).

    Abbreviations

    AQP1, aquaporin-1; bFGF, basic fibroblast growth factor; CLDN4, claudin-4; CSF1R, CSF1 receptor; KLT, kanglaite injection; MAPK, mitogen-activated protein kinase; MCP-1, monocyte chemotactic protein-1; MPE, malignant pleural effusion; NRP1, neuropilin 1; NSCLC, non-small cell lung cancer; ORR, objective response rate; PDGF, platelet-derived growth factor; PE, pleural effusion; TCM, traditional Chinese medicine; TGF-β1, transforming growth factor-β1; TGF-β, transforming growth factor β; Th17, T helper cell 17; VEGF, vascular endothelial growth factor; VSZT, toxic members in shizaotang processed with vinegar.

    Data Sharing Statement

    No additional data are available.

    Ethics Approval

    The study did not require ethical review board approval or informed consent.

    Use of AI

    The authors declare that they have not use AI-generated work in this manuscript.

    Funding

    i Guangdong Basic and Applied Basic Research Foundation (No. 2023A1515012338). ii Construction Project for Renowned Traditional Chinese Medicine (TCM) Inheritance Studios in Guangdong Province 2024 - Ying Lv’s Renowned TCM Inheritance Studio (Guangdong TCM Office Document [2023] No. 108). iii President Foundation of Nanfang Hospital, Southern Medical University (No. 2023CR034).

    Disclosure

    All authors declare that they have no competing interests.

    References

    1. Zhang X, Duan H, Yu Y, et al. Differential diagnosis between benign and malignant pleural effusion with dual-energy spectral CT. PLoS One. 2018;13(4):e0193714. doi:10.1371/journal.pone.0193714

    2. Li M, Wang H, Wang X, et al. Diagnostic accuracy of tumor necrosis factor-alpha, interferon-gamma, interlukine-10 and adenosine deaminase 2 in differential diagnosis between tuberculous pleural effusion and malignant pleural effusion. J Cardiothorac Surg. 2014;9:118. doi:10.1186/1749-8090-9-118

    3. Bashour SI, Mankidy BJ, Lazarus DR. Update on the diagnosis and management of malignant pleural effusions. Respir Med. 2022;196:106802. doi:10.1016/j.rmed.2022.106802

    4. Marazioti A, Lilis I, Vreka M, et al. Myeloid-derived interleukin-1β drives oncogenic KRAS-NF-κΒ addiction in malignant pleural effusion. Nat Commun. 2018;9(1):672. doi:10.1038/s41467-018-03051-z

    5. Pardessus Otero A, Rafecas-Codern A, Porcel JM, et al. Malignant pleural effusion: a multidisciplinary approach. Open Respir Arch. 2024;6(4):100349. doi:10.1016/j.opresp.2024.100349

    6. Rintoul RC, Ritchie AJ, Edwards JG, et al. Efficacy and cost of video-assisted thoracoscopic partial pleurectomy versus talc pleurodesis in patients with malignant pleural mesothelioma (MesoVATS): an open-label, randomised, controlled trial. Lancet. 2014;384(9948):1118–1127. doi:10.1016/S0140-6736(14)60418-9

    7. Spella M, Giannou AD, Stathopoulos GT. Switching off malignant pleural effusion formation-fantasy or future? J Thorac Dis. 2015;7(6):1009–1020. doi:10.3978/j.issn.2072-1439.2015.05.20

    8. Finley DJ, Rusch VW. Anatomy of the Pleura. Thorac Surg Clin. 2011;21(2):157–163. doi:10.1016/j.thorsurg.2010.12.001

    9. Ruan Z, Zhuo X, Xu C. Diagnosis, treatment, and prognosis of stage IB non-small cell lung cancer with visceral pleural invasion. Front Oncol. 2024;13:1310471. doi:10.3389/fonc.2023.1310471

    10. Wahidi MM, Reddy C, Yarmus L, et al. Randomized trial of pleural fluid drainage frequency in patients with malignant pleural effusions. The ASAP trial. Am J Respir Crit Care Med. 2017;195(8):1050–1057. doi:10.1164/rccm.201607-1404OC

    11. Scallan JP, Zawieja SD, Castorena Gonzalez JA, Davis MJ. Lymphatic pumping: mechanics, mechanisms and malfunction. J Physiol. 2016;594(20):5749–5768. doi:10.1113/JP272088

    12. Maa HC, Chao TT, Wang CY, et al. VEGF-D as a marker in the aid of malignant metastatic pleural effusion diagnosis. Appl Immunohistochem Mol Morphol. 2015;23(3):209–214. doi:10.1097/PAI.0000000000000079

    13. Giannou AD, Marazioti A, Spella M, et al. Mast cells mediate malignant pleural effusion formation. J Clin Invest. 2015;125(6):2317–2334. doi:10.1172/JCI79840

    14. Li L, Yang L, Wang L, et al. Impaired T cell function in malignant pleural effusion is caused by TGF-beta derived predominantly from macrophages. Int, J Cancer. 2016;139(10):2261–2269. doi:10.1002/ijc.30289

    15. Klimatsidas M, Anastasiadis K, Foroulis C, et al. Elevated levels of anti inflammatory IL-10 and pro inflammatory IL-17 in malignant pleural effusions. J Cardiothorac Surg. 2012;7:104. doi:10.1186/1749-8090-7-104

    16. Murthy P, Ekeke CN, Russell KL, et al. Making cold malignant pleural effusions hot: driving novel immunotherapies. Oncoimmunology. 2019;8(4):e1554969. doi:10.1080/2162402X.2018.1554969

    17. Psallidas I, Kalomenidis I, Porcel JM, Robinson BW, Stathopoulos GT. Malignant pleural effusion: from bench to bedside. Eur Respir Rev. 2016;25(140):189–198. doi:10.1183/16000617.0019-2016

    18. Gonnelli F, Hassan W, Bonifazi M, et al. Malignant pleural effusion: current understanding and therapeutic approach. Respir Res. 2024;25(1):47. doi:10.1186/s12931-024-02684-7

    19. Yano S, Shinohara H, Herbst RS, et al. Production of experimental malignant pleural effusions is dependent on invasion of the pleura and expression of vascular endothelial growth factor/vascular permeability factor by human lung cancer cells. Am J Pathol. 2000;157(6):1893–1903. doi:10.1016/S0002-9440(10)64828-6

    20. Dipper A, Jones HE, Bhatnagar R, et al. Interventions for the management of malignant pleural effusions: an updated network meta-analysis. Eur Respir Rev. 2021;30(160):210025. doi:10.1183/16000617.0025-2021

    21. Stathopoulos GT, Kalomenidis I. Malignant pleural effusion: tumor-host interactions unleashed. Am J Respir Crit Care Med. 2012;186(6):487–492. doi:10.1164/rccm.201203-0465PP

    22. Chen Y, Yang W, Whang-Peng J, Kuo BI, Perng R. Elevation of lnterleukin-10 Levels in Malignant Pleural Effusion. Chest. 1996;110(2):433–436. doi:10.1378/chest.110.2.433

    23. Lending G, El Ghani YA, Kaykov E, et al. Hemorrhagic malignant pleural effusion: diagnosis, survival rate, and response to talc pleurodesis. Indian J Surg Oncol. 2021;12(1):54–60. doi:10.1007/s13193-020-01099-2

    24. Nie K, Zhang Z, You Y, et al. 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. 2020;11(1):8–14. doi:10.1111/1759-7714.13238

    25. Hooper CE, Elvers KT, Welsh GI, Millar AB, Maskell, Maskell NA. NA. VEGF and sVEGFR-1 in malignant pleural effusions: association with survival and pleurodesis outcomes. Lung Cancer. 2012;77(2):443–449. doi:10.1016/j.lungcan.2012.03.006

    26. Hsu LH, Soong TC, Chu NM, et al. The inflammatory cytokine profile of patients with malignant pleural effusion treated with pleurodesis. J Clin Med. 2020;9(12):4010. doi:10.3390/jcm9124010

    27. Sabang RL, Gandhiraj D, Fanucchi M, Epelbaum O. Role of bevacizumab in the management of the patient with malignant pleural effusion: more questions than answers. Expert Rev Respir Med. 2018;12(2):87–94. doi:10.1080/17476348.2018.1417042

    28. Sack U, Hoffmann M, Zhao XJ, et al. Vascular endothelial growth factor in pleural effusions of different origin. Eur Respir J. 2005;25(4):600–604. doi:10.1183/09031936.05.00037004

    29. Fiorelli A, Vicidomini G, Di Domenico M, et al. Vascular endothelial growth factor in pleural fluid for differential diagnosis of benign and malignant origin and its clinical applications. Interact Cardiovasc Thorac Surg. 2011;12(3):420–424. doi:10.1510/icvts.2010.250357

    30. Lieser EA, Croghan GA, Nevala WK, et al. Up-regulation of pro-angiogenic factors and establishment of tolerance in malignant pleural effusions. Lung Cancer. 2013;82(1):63–68. doi:10.1016/j.lungcan.2013.07.007

    31. Gkiozos I, Tsagouli S, Charpidou A, et al. 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. 2015;35(2):1129–1137.

    32. Zhang Y, Yu LK, Lu GJ, et al. Prognostic values of VEGF and endostatin with malignant pleural effusions in patients with lung cancer. Asian Pac J Cancer Prev. 2014;15(19):8435–8440. doi:10.7314/APJCP.2014.15.19.8435

    33. He D, Ding R, Wen Q, Chen L. Novel therapies for malignant pleural effusion: anti‑angiogenic therapy and immunotherapy (Review). Int J Oncol. 2021;58(3):359–370. doi:10.3892/ijo.2021.5174

    34. Sharma S, Ehrlich M, Zhang M, Blobe GC, Henis YI. NRP1 interacts with endoglin and VEGFR2 to modulate VEGF signaling and endothelial cell sprouting. Commun Biol. 2024;7(1):112. doi:10.1038/s42003-024-05798-2

    35. Wang L, Liu W, Broussy S, Han B, Fang H. Recent advances of anti-angiogenic inhibitors targeting VEGF/VEGFR axis. Front Pharmacol. 2024;14:1307860. doi:10.3389/fphar.2023.1307860

    36. Zachary I. VEGF signalling: integration and multi-tasking in endothelial cell biology. Biochem Soc Trans. 2003;31(Pt 6):1171–1177. doi:10.1042/bst0311171

    37. Ardizzone A, Bova V, Casili G, et al. Role of basic fibroblast growth factor in cancer: biological activity, targeted therapies, and prognostic value. Cells. 2023;12(7):1002. doi:10.3390/cells12071002

    38. Antony VB, Nasreen N, Mohammed KA, et al. Talc pleurodesis: basic fibroblast growth factor mediates pleural fibrosis. Chest. 2004;126(5):1522–1528. doi:10.1378/chest.126.5.1522

    39. Strizzi L, Vianale G, Catalano A, et al. Basic fibroblast growth factor in mesothelioma pleural effusions: correlation with patient survival and angiogenesis. Int J Oncol. 2001;18(5):1093–1098. doi:10.3892/ijo.18.5.1093

    40. Barash U, Lapidot M, Zohar Y, et al. Involvement of heparanase in the pathogenesis of mesothelioma: basic aspects and clinical applications. J Natl Cancer Inst. 2018;110(10):1102–1114. doi:10.1093/jnci/djy032

    41. Barash U, Cohen-Kaplan V, Dowek I, et al. Proteoglycans in health and disease: new concepts for heparanase function in tumor progression and metastasis. FEBS J. 2010;277(19):3890–3903. doi:10.1111/j.1742-4658.2010.07799.x

    42. Vlodavsky I, Beckhove P, Lerner I, et al. Significance of heparanase in cancer and inflammation. Cancer Microenvironment. 2012;5(2):115–132. doi:10.1007/s12307-011-0082-7

    43. Liu Y-H, Wu L-H, Fan W-J, et al. A tellurium-based small compound ameliorates tumor metastasis by downregulating heparanase expression. J Cancer. 2024;15(16):5308–5317. doi:10.7150/jca.96001

    44. Li L, Barash U, Ilan N, et al. A new synthesized dicarboxylated oxy-heparin efficiently attenuates tumor growth and metastasis. Cells. 2024;13(3):211. doi:10.3390/cells13030211

    45. Shalapour S, Karin M. Immunity, inflammation, and cancer: an eternal fight between good and evil. J Clin Invest. 2015;125(9):3347–3355. doi:10.1172/JCI80007

    46. Li F, Du X, Lan F, et al. Eosinophilic inflammation promotes CCL6-dependent metastatic tumor growth. Sci Adv. 2021;7(22):eabb5943. doi:10.1126/sciadv.abb5943

    47. Agalioti T, Giannou AD, Krontira AC, et al. Mutant KRAS promotes malignant pleural effusion formation. Nat Commun. 2017;8:15205. doi:10.1038/ncomms15205

    48. Burgers JA, Kunst PW, Koolen MG, et al. Pleural drainage and pleurodesis: implementation of guidelines in four hospitals. Eur Respir J. 2008;32(5):1321–1327. doi:10.1183/09031936.00165607

    49. Psallidas I, Karabela SP, Moschos C, et al. Specific effects of bortezomib against experimental malignant pleural effusion: a preclinical study. Mol Cancer. 2010;9:56. doi:10.1186/1476-4598-9-56

    50. Stathopoulos GT, Kollintza A, Moschos C, et al. Tumor necrosis factor-alpha promotes malignant pleural effusion. Cancer Res. 2007;67(20):9825–9834. doi:10.1158/0008-5472.CAN-07-1064

    51. Stathopoulos GT, Zhu Z, Everhart MB, et al. Nuclear factor-κb affects tumor progression in a mouse model of malignant pleural effusion. Am J Respir Cell Mol Biol. 2006;34(2):142–150. doi:10.1165/rcmb.2005-0130OC

    52. Mierzejewski M, Korczynski P, Krenke R, Janssen JP. Chemical pleurodesis - a review of mechanisms involved in pleural space obliteration. Respir Res. 2019;20(1):247. doi:10.1186/s12931-019-1204-x

    53. Schwarz Y, Star A, Friedberg J. Role of talc modulation on cytokine activation in cancer patients undergoing pleurodesis. Pulm Med. 2012;2012(2012):1–6. doi:10.1155/2012/806183

    54. Koch AE, Polverini PJ, Kunkel SL, et al. Interleukin-8 as a macrophage-derived mediator of angiogenesis. Science. 1992;258(5089):1798–1801. doi:10.1126/science.1281554

    55. Yoshida S, Ono M, Shono T, et al. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol. 1997;17(7):4015–4023. doi:10.1128/MCB.17.7.4015

    56. Zhao J, Liu B, Liu N, et al. The role of angiogenesis in malignant pleural effusion: from basic research to clinical application. Am J Cancer Res. 2022;12(11):4879–4891.

    57. Montes-Worboys A, Rodriguez-Portal JA, Arellano-Orden E, Digon-Pereiras J, Rodriguez-Panadero F. Interleukin-8 activates coagulation and correlates with survival after talc pleurodesis. Eur Respir J. 2010;35(1):160–166. doi:10.1183/09031936.00146808

    58. Stathopoulos GT, Psallidas I, Moustaki A, et al. A central role for tumor-derived monocyte chemoattractant protein-1 in malignant pleural effusion. J Natl Cancer Inst. 2008;100(20):1464–1476. doi:10.1093/jnci/djn325

    59. Dipper A, Bhatnagar R, Maskell N. Management of malignant pleural effusions. Curr Opin Pulm Med. 2020;26(4):341–345. doi:10.1097/MCP.0000000000000685

    60. Jiang W, Chan CK, Weissman IL, Kim BYS, Hahn SM. Immune Priming of the Tumor Microenvironment by Radiation. Trends Cancer. 2016;2(11):638–645. doi:10.1016/j.trecan.2016.09.007

    61. Li Y, Wan YY, Zhu B. Immune cell metabolism in tumor microenvironment. 2017;1011:163–196. doi:10.1007/978-94-024-1170-6_5

    62. Ge S, Zhao Y, Liang J, et al. Immune modulation in malignant pleural effusion: from microenvironment to therapeutic implications. Cancer Cell Int. 2024;24(1):105. doi:10.1186/s12935-024-03211-w

    63. Huang ZY, Shao MM, Zhang JC, et al. Single-cell analysis of diverse immune phenotypes in malignant pleural effusion. Nat Commun. 2021;12(1):6690. doi:10.1038/s41467-021-27026-9

    64. Niu Y, Zhou Q. Th17 cells and their related cytokines: vital players in progression of malignant pleural effusion. Cell Mol Life Sci. 2022;79(4):194. doi:10.1007/s00018-022-04227-z

    65. Kosti CN, Vaitsi PC, Pappas AG, et al. CSF1/CSF1R signaling mediates malignant pleural effusion formation. JCI Insight. 2022;7(6):e155300. doi:10.1172/jci.insight.155300

    66. Ito M, Nakano M, Ariyama H, et al. Macrophages are primed to transdifferentiate into fibroblasts in malignant ascites and pleural effusions. Cancer Lett. 2022;532:215597. doi:10.1016/j.canlet.2022.215597

    67. Wang S, An J, Hu X, et al. Single-cell RNA sequencing reveals immune microenvironment of small cell lung cancer-associated malignant pleural effusion. Thorac Cancer. 2024;15(1):98–103. doi:10.1111/1759-7714.15145

    68. Schaaij-Visser TB, de Wit M, Lam SW, Jimenez CR. The cancer secretome, current status and opportunities in the lung, breast and colorectal cancer context. Biochim Biophys Acta. 2013;1834(11):2242–2258. doi:10.1016/j.bbapap.2013.01.029

    69. Watabe S, Kikuchi Y, Morita S, et al. Clinicopathological significance of microRNA-21 in extracellular vesicles of pleural lavage fluid of lung adenocarcinoma and its functions inducing the mesothelial to mesenchymal transition. Cancer Med. 2020;9(8):2879–2890. doi:10.1002/cam4.2928

    70. Liberato T, Pessotti DS, Fukushima I, et al. Signatures of protein expression revealed by secretome analyses of cancer associated fibroblasts and melanoma cell lines. J Proteomics. 2018;174:1–8. doi:10.1016/j.jprot.2017.12.013

    71. Nikitovic D, Papoutsidakis A, Karamanos NK, Tzanakakis GN. Lumican affects tumor cell functions, tumor–ECM interactions, angiogenesis and inflammatory response. Matrix Biol. 2014;35:206–214. doi:10.1016/j.matbio.2013.09.003

    72. Schaefer L, Tredup C, Gubbiotti MA, Iozzo RV. Proteoglycan neofunctions: regulation of inflammation and autophagy in cancer biology. FEBS J. 2017;284(1):10–26. doi:10.1111/febs.13963

    73. Cappellesso R, Millioni R, Arrigoni G, et al. Lumican is overexpressed in lung adenocarcinoma pleural effusions. PLoS One. 2015;10(5):e0126458. doi:10.1371/journal.pone.0126458

    74. Safi A, Sadmi M, Martinet N, et al. Presence of elevated levels of platelet-derived growth factor (PDGF) in lung adenocarcinoma pleural effusions. Chest. 1992;102(1):204–207. doi:10.1378/chest.102.1.204

    75. Zhang X, Wang X, Wen Y, et al. Single-cell transcriptomics reveal metastatic CLDN4 + cancer cells underlying the recurrence of malignant pleural effusion in patients with advanced non-small-cell lung cancer. Clin Transl Med. 2024;14(4):e1649. doi:10.1002/ctm2.1649

    76. Ozmen ZC, Kupeli M. Clinical importance of serum and pleural fluid prominin-1 and hypoxia-inducible factor-1α concentration in the evaluation of lymph node involvement in patients with malignant pleural effusion. Biochem Med. 2023;33(3):295–304. doi:10.11613/BM.2023.030701

    77. Borok Z, Verkman AS. Lung edema clearance: 20 years of progress: invited review: role of aquaporin water channels in fluid transport in lung and airways. J Appl Physiol. 2002;93(6):2199–2206. doi:10.1152/japplphysiol.01171.2001

    78. Song Y, Yang B, Matthay MA, Ma T, Verkman AS. Role of aquaporin water channels in pleural fluid dynamics. Am J Physiol Cell Physiol. 2000;279(6):C1744–50. doi:10.1152/ajpcell.2000.279.6.C1744

    79. Calamita G. Advances in Aquaporins. Cells. 2023;12(2):303. doi:10.3390/cells12020303

    80. Alfieri L, Montana A, Frisoni P, D’Errico S, Neri M. Application of aquaporins as markers in forensic pathology: a systematic review of the literature. Int J Mol Sci. 2024;25(5):2664. doi:10.3390/ijms25052664

    81. Benga G. Birth of water channel proteins-the aquaporins. Cell Biol Int. 2003;27(9):701–709. doi:10.1016/S1065-6995(03)00171-9

    82. Keskinidou C, Vassiliou AG, Dimopoulou I, Kotanidou A, Orfanos SE. Mechanistic understanding of lung inflammation: recent advances and emerging techniques. J Inflamm Res. 2022;15:3501–3546. doi:10.2147/JIR.S282695

    83. Imredi E, Liszkay G, Kenessey I, et al. Aquaporin-1 protein expression of the primary tumor may predict cerebral progression of cutaneous melanoma. Pathol Oncol Res. 2020;26(1):405–410. doi:10.1007/s12253-018-0513-6

    84. Pulford E, McEvoy J, Hocking A, et al. the effect of aquaporin 1-inhibition on vasculogenic mimicry in malignant mesothelioma. Int J Mol Sci. 2017;18(11):2293. doi:10.3390/ijms18112293

    85. Chow PH, Bowen J, Yool AJ. Combined systematic review and transcriptomic analyses of mammalian aquaporin classes 1 to 10 as biomarkers and prognostic indicators in diverse cancers. Cancers. 2020;12(7):1911. doi:10.3390/cancers12071911

    86. Ma B, Xiang Y, Li T, Yu HM, Li XJ. Inhibitory effect of topiramate on Lewis lung carcinoma metastasis and its relation with AQP1 water channel. Acta Pharmacol Sin. 2004;25(1):54–60.

    87. Zhang JX, Xie CM, Zhu ZW, Huang HY, Zeng ZL. Potential role of AQP1 and VEGF in the development of malignant pleural effusion in mice. Med Oncol. 2012;29(2):656–662. doi:10.1007/s12032-011-9960-6

    88. Lin Z, Jiang M, Gao L, Zhang H. The clinical efficacy of traditional Chinese medicine in the treatment of malignant pleural effusion. Medicine. 2020;99(39):e22403. doi:10.1097/MD.0000000000022403

    89. Nolte S, Liegl G, Petersen MA, et al. General population normative data for the EORTC QLQ-C30 health-related quality of life questionnaire based on 15,386 persons across 13 European countries, Canada and the Unites States. Eur J Cancer. 2019;107:153–163. doi:10.1016/j.ejca.2018.11.024

    90. Zhong XG, Zheng FJ, Li YH, et al. Specific link between lung and large intestine: a new perspective on neuropeptide secretion in lung with herbal laxative stimulation. Evid Based Complement Alternat Med. 2013;2013:547837. doi:10.1155/2013/547837

    91. Feize W, Meng L, Yanni L, et al. A randomized controlled study to observe the efficacy of external treatment with a traditional Chinese medicine herbal ointment on malignant plural effusion: outcome report and design review. Integr Cancer Ther. 2017;16(4):473–478. doi:10.1177/1534735416660193

    92. Fuhong D, Xiang G, Haiying L, et al. Evaluation of efficacy and safety for Brucea javanica oil emulsion in the control of the malignant pleural effusions via thoracic perfusion. BMC Cancer. 2018;18(1):411. doi:10.1186/s12885-018-4328-3

    93. Tian P. Where West meets East. Nature. 2011;480:S84–S86. doi:10.1038/480S84a

    94. Shen J, Wang J, Shang E, et al. The dosage-toxicity-efficacy relationship of kansui and licorice in malignant pleural effusion rats based on factor analysis. J Ethnopharmacol. 2016;186:251–256. doi:10.1016/j.jep.2016.04.012

    95. Jin Z, Shen C, Zhang H, et al. Chinese medicine Xiaoshui decoction inhibits malignant pleural effusion in mice and mediates tumor-associated macrophage polarization by activating autophagy. J Ethnopharmacol. 2020;249:112412. doi:10.1016/j.jep.2019.112412

    96. Lou JW, Cao LL, Zhang Q, et al. The toxicity and efficacy evaluation of different fractions of Kansui fry-baked with vinegar on Walker-256 tumor-bearing malignant ascites effusion rats and normal rats. J Ethnopharmacol. 2018;219:257–268. doi:10.1016/j.jep.2018.03.010

    97. Zhang Q, Li ZL, Xu JD, et al. Toxicity reduction and water expelling effect preservation of Shizaotang after its toxic members processing with vinegar on rats with malignant pleural effusions. J Ethnopharmacol. 2021;268:113583. doi:10.1016/j.jep.2020.113583

    98. Ru J, Li P, Wang J, et al. TCMSP: a database of systems pharmacology for drug discovery from herbal medicines. J Cheminform. 2014;6:13. doi:10.1186/1758-2946-6-13

    99. Gong F, Qu R, Yang Y, et al. The mechanism of ginseng and astragalus decoction in the treatment of malignant pleural effusion based on network pharmacology and molecular docking technology. Evid Based Complement Alternat Med. 2022;2022:7731402. doi:10.1155/2022/7731402

    100. Liu J, Liu X, Ma J, Li K, Xu C. The clinical efficacy and safety of kanglaite adjuvant therapy in the treatment of advanced hepatocellular carcinoma: a PRISMA-compliant meta-analysis. Biosci Rep. 2019;39(11). doi:10.1042/BSR20193319

    101. Wu Y, Zhang J, Hong Y, Wang X. Effects of Kanglaite injection on serum miRNA-21 in patients with advanced lung cancer. Med Sci Monit. 2018;24:2901–2906. doi:10.12659/MSM.909719

    102. Huang X, Wang J, Lin W, et al. Kanglaite injection plus platinum-based chemotherapy for stage III/IV non-small cell lung cancer: a meta-analysis of 27 RCTs. Phytomedicine. 2020;67:153154. doi:10.1016/j.phymed.2019.153154

    103. Li J, Li H, Zhu G, et al. Efficacy and safety of Kanglaite injection combined with first-line platinum-based chemotherapy in patients with advanced NSCLC: a systematic review and meta-analysis of 32 RCTs. Ann Palliat Med. 2020;9(4):1518. doi:10.21037/apm-20-616

    104. Wang CQ, Zheng XT, Chen XF, et al. the optimal adjuvant strategy of aidi injection with gemcitabine and cisplatin in advanced non-small cell lung cancer: a meta-analysis of 70 randomized controlled trials. Front Pharmacol. 2021;12:582447. doi:10.3389/fphar.2021.582447

    105. Wang C, Xu J, Jiang H, et al. The evidence framework of traditional Chinese medicine injection (Aidi injection) in controlling malignant pleural effusion: a clustered systematic review and meta-analysis. Phytomedicine. 2023;115:154847. doi:10.1016/j.phymed.2023.154847

    106. Zhai B, Zeng Y, Zeng Z, et al. Drug delivery systems for elemene, its main active ingredient beta-elemene, and its derivatives in cancer therapy. Int J Nanomed. 2018;13:6279–6296. doi:10.2147/IJN.S174527

    107. Wang X, Liu Z, Sui X, et al. Elemene injection as adjunctive treatment to platinum-based chemotherapy in patients with stage III/IV non-small cell lung cancer: a meta-analysis following the PRISMA guidelines. Phytomedicine. 2019;59:152787. doi:10.1016/j.phymed.2018.12.010

    108. Chen J, Chen S, Luo H, et al. The complementary and alternative roles of elemene injection in cancer: an umbrella review. Pharmacol Res. 2023;198:107007. doi:10.1016/j.phrs.2023.107007

    109. Tao L, Fu J, Wang F, et al. The application of mirabilite in traditional Chinese medicine and its chemical constituents, processing methods, pharmacology, toxicology and clinical research. Front Pharmacol. 2023;14:1293097. doi:10.3389/fphar.2023.1293097

    110. Liudvytska O, Bandyszewska M, Skirecki T, et al. Anti-inflammatory and antioxidant actions of extracts from Rheum rhaponticum and Rheum rhabarbarum in human blood plasma and cells in vitro. Biomed Pharmacother. 2023;165:115111. doi:10.1016/j.biopha.2023.115111

    111. Gao Q, Liang N. Integrated traditional Chinese medicine improves acute pancreatitis via the downregulation of PRSS1 and SPINK1. Exp Ther Med. 2015;9(3):947–954. doi:10.3892/etm.2015.2191

    112. Zhang H, Jiang M, Gao L, Lin Z. The clinical efficacy of external application of mirabilite and rhubarb combined with intrathoracic chemotherapy in treating malignant pleural effusion: a prospective, randomized, controlled clinical trial. Medicine. 2021;100(7):e24758. doi:10.1097/MD.0000000000024758

    Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.