Next Article in Journal
Ovarian Accumulation of Nanoemulsions: Impact of Mice Age and Particle Size
Next Article in Special Issue
Inhibition of Cell Proliferation and Metastasis by Scutellarein Regulating PI3K/Akt/NF-κB Signaling through PTEN Activation in Hepatocellular Carcinoma
Previous Article in Journal
Are Changes in the Percentage of Specific Leukocyte Subpopulations Associated with Endogenous DNA Damage Levels in Testicular Cancer Patients?
Previous Article in Special Issue
4-Acetyl-Antroquinonol B Improves the Sensitization of Cetuximab on Both Kras Mutant and Wild Type Colorectal Cancer by Modulating the Expression of Ras/Raf/miR-193a-3p Signaling Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phytochemicals in Malignant Pleural Mesothelioma Treatment—Review on the Current Trends of Therapies

by
Malgorzata Chmielewska-Kassassir
and
Lucyna A. Wozniak
*
Department of Structural Biology, Medical University of Lodz, Zeligowskiego 7/9 Str., 90-752 Lodz, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(15), 8279; https://doi.org/10.3390/ijms22158279
Submission received: 5 July 2021 / Revised: 24 July 2021 / Accepted: 28 July 2021 / Published: 31 July 2021

Abstract

:
Malignant pleural mesothelioma (MPM) is a rare but highly aggressive tumor of pleura arising in response to asbestos fibers exposure. MPM is frequently diagnosed in the advanced stage of the disease and causes poor prognostic outcomes. From the clinical perspective, MPM is resistant to conventional treatment, thus challenging the therapeutic options. There is still demand for improvement and sensitization of MPM cells to therapy in light of intensive clinical studies on chemotherapeutic drugs, including immuno-modulatory and targeted therapies. One way is looking for natural sources, whole plants, and extracts whose ingredients, especially polyphenols, have potential anticancer properties. This comprehensive review summarizes the current studies on natural compounds and plant extracts in developing new treatment strategies for MPM.

1. Introduction

Malignant pleural mesothelioma (MPM) is a relatively rare tumor of pleura arising in approximately 80% of cases due to prolonged occupational (rarely environmental or domestic) exposure to asbestos fibers in the past. Each year, the incidence rate of MPM increases significantly, and according to the European Society for Medical Oncology (ESMO), only within the next 20 years, the newly reported incidents of patients with MPM will be doubled, with a fivefold higher incidence in case of men [1,2]. In many European countries, the incidences of mesothelioma result from a prolonged latency period between asbestos exposure (starting in the 1970s) and cancer diagnosis [3]. Worryingly, this particular cancer type is highly aggressive and insensitive to conventional treatment, including radiotherapy, resection (in justified cases), and primary chemotherapy. Chemotherapy with antifolic analog is the first-line standard therapy—pemetrexed and cytostatic platinum, with the median overall survival reaching approximately one and a half. New targeted strategies and immunotherapies, especially immune checkpoint inhibitors, are under intensive investigation for patients who are not candidates for cisplatin-based therapy.
Additionally, due to the delayed (as a result of latent character of the disease lasting even 30 years) and very often ambiguous diagnosis (misdiagnosed with small-cell lung cancer; SCLC), the course of the illness and, in consequence, prognostic outcomes for MPM-bearing patients are significantly reduced. In the face of the dark nature of MPM, apart from improving the accurate and early diagnosis, it seems reasonable to search for novel therapeutic strategies, including natural compounds that may help manage MPM tumors. This review sheds light on the current state of research on phytochemicals investigated in MPM therapy.

2. Novel Targeted Therapies against Malignant Pleural Mesothelioma Cells

The prolonged inhalation of asbestos fibers leads to chronic inflammation within the pleura and, as a consequence, facilitates carcinogenic processes. Asbestos fibers mechanically damage the integrity of mesothelial tissue, leading to a local inflammatory response with observed overproduction of free radicals (reactive oxygen species, ROS, and reactive nitrogen species, RNS) and pro-inflammatory cytokines [4,5]. At the beginning of the inflammatory process, the tumor necrosis factor-alpha (TNF-α) and its downstream nuclear factor kappa B (NF-κB) signaling pathway play a critical role. Upon mechanistic injury, mesothelial cells secrete high-mobility group bax 1 protein (HMGBP1), which stimulates macrophages recruitment in the surrounding space of the damaged mesothelial cells and releases excessive amounts of TNF-α mediator [6]. TNF-α acts directly with its receptor TNF-R1 expressed on the surface of human mesothelial (HM) cells [7]. In turn, this interaction stimulates HM cells into further autocrine release of TNF-α, which finally activates and accelerates NF-κB downstream signaling, mainly pro-survival gene transcription by the p65 subunit of NF-κB, skipping an apoptosis process of the injured cells [8]. Consequently, HM cells intensively divide rather than dying, causing mistakes in the uncontrolled division that accumulates DNA synthesis, leading to malignant mesothelial cell transformation (Figure 1).
Several alterations in genes and molecular pathways related to mesothelial cell proliferation, susceptibility to apoptosis, and angiogenic processes are observed under MPM carcinogenesis [9]. The most frequent in MPM pathogenesis are genetic mutations in tumor suppressor genes (including neurofibromin 2 (NF2, merlin), cyclin-dependent kinase inhibitor 2A (CDKN2A), which shares a locus for both p16/INK4 and p14/ARF, and BRCA-associated protein-1 (BAP-1). The observed genetic alternations in these genes play a significant role in MPM development due to their direct involvement in controlling cell shape, adhesion, and cell growth [10]. Some additional alternations involve Wnt/β-catenin signaling pathways, as well as epigenetic silencing of metabolic pathway-related genes, including arginine succinate synthase (ASS1) [11]. High expression of angiogenic growth factors (e.g., vascular endothelial growth factor, VEGF) as well as their surface receptors (e.g., VEGFR, EGFR (epidermal growth factor receptor) or MET (hepatocyte growth factor receptor) and other glycoproteins (mesothelin) is a predominant determinant of intracellular signaling pathways in carcinogenic MPM cells [12]. These molecular factors and their downstream molecular pathways are currently intensively investigated and comprise the first line of interest in targeted MPM treatment strategy (Figure 2). However, until now, no significant effects have been observed in the case of anti-angiogenic drugs (in either VEGF/VEGFR or PDGF/PDGFR (platelet-derived growth factor/PDGF receptor targets)) or pro-apoptotic therapies including histones deacetylases inhibitors (HDAC) and PI3K/AKT/mTOR pathway (phosphatidylinositol-3-kinase and alpha serine/threonine-protein kinase) [13,14]. Both targeted therapies, including anti-mesothelin, anti-MET, and anti-Hsp90 (Heat shock protein-90) antibodies, and immunotherapies with predominant immune checkpoint inhibitors (anti-CTLA-4, anti-PD-1, and anti-PD-L1) are still under clinical trials due to lack of direct recommendations of such treatment to MPM patients.
Further detailed studies are required to correlate patients’ characteristics and expression status of target molecules with the implementation of a particular therapy. The observed discrepancy between clinical trials and documented unresponsiveness of mesothelioma to most targeted agents can be explained by, first of all, the non-randomized type of studies (unselected based on unique biomarker patients) as well as the analysis of various endpoints (response rate, progression-free rate vs. overall survival data). Second, the resistance of malignant pleural mesothelioma cells to apoptosis-triggering signals can be highly individual; thus, the development of selective biomarkers of response is urgently needed. Moreover, unrelated directly to apoptotic pathways, new approaches have recently been extensively studied, including metabolite deprivation (e.g., arginine starvation of malignant mesothelioma cells) or enhancement of immune system response (including administration of oncolytic viruses and therapeutic peptide or dendritic cells vaccines) [13].
Some MPM subtypes are characterized by epigenetically lost or reduced expression of argininosuccinate synthetase-1 (ASS1), known as a rate-limiting enzyme involved in the endogenous synthesis of L-arginine in the urea cycle. Recent studies have demonstrated that the growth and viability of these so-called ASS1-negative cancer cells are highly dependent on the L-arginine level [15]. This L-arginine auxotrophic phenomenon of ASS1-negative tumors, including melanoma, hepatocellular carcinoma, and bladder cancer, has led to the development of a new therapeutic strategy using the arginine deiminase enzyme (in pegylated form; ADI-PEG20), responsible for the deprivation of L-arginine presented in the environment of the tumor cells and lethality of MPM cancer [16,17]. A Phase 1 TRAP trial confirmed positive outcomes in ASS1-deficient patients under treatment with ADI-PEG20 alone or combined with cisplatin plus pemetrexed [18,19]. The arginine starvation treatment strategy resulted in encouraging results. The randomized, double-blinded Phase 2/3 clinical trial, NCT02709512, is currently being conducted to evaluate the effectiveness of ADI-PEG20 in combinatory treatment with first-line standard therapeutics (cisplatin/pemetrexed) in patients with biphasic (mixed) or sarcomatoid subtypes of MPM. The ASS1-related targeted therapy is the first of the triplet chemotherapy combination (ADIPEMCIS) studies that causes (based on Phase 1 results) effective plasma arginine deprivation in recruited MPM patients, with similar good prognostic outcomes reflected in both the median progression-free survival (PFS) value and median overall response (OS) rate, 5.6 and 10.1 months, respectively.
Today, the ongoing predominant clinical trials are focused on immunotherapy since the immune system and immune cells play a critical role in the pathogenesis of malignant pleural mesothelioma. While the development of MPM is a consequence of mechanical damage of epithelioid cells with the concomitant chronic inflammatory process, lasting a long time in the surrounding environment of pleura, several critical immune checkpoint targets are under investigation [13,14]. The most promising targets receiving research attention are cytotoxic T lymphocyte-associated protein (CTLA-4) and a programmed cell death protein-1 (PD-1) expressed on regulatory (CD4 ‘helper’) T cells, and a programmed cell death protein ligand-1 (PD-L1) highly expressed by MPM cells. The targeted strategy against these molecules aims to restore CD8 ‘killer’ T lymphocytes’ activity and proper anti-tumor immune response [20]. However, despite the positive preliminary results, neither anti-CTLA-4 nor anti-PD-L1 provides successful benefits in a single treatment strategy; thus, further validation of clinical outcomes is still required in combinatory studies of a larger randomized trial. The most hopeful seems to be the anti-PD-1 immunotherapy in unresectable MPM cases [21].
Some encouraging results have also been seen in utilizing chimeric monoclonal anti-mesothelin antibody (amatuximab) in the clinical trial. The overexpression of mesothelin, observed mainly in epithelial cancers, seems to be related to tumorigenesis and metastasis. The preclinical studies revealed that amatuximab induced antibody-dependent cellular cytotoxicity of mesothelin-expressing tumor cells in in vitro studies and significantly reduced mesothelin-positive tumors in the xenograft model. The Phase 2 clinical study demonstrated that amatuximab treatment was well-tolerated by patients with unresectable MPM, and its synergy with chemotherapeutics, cisplatin, and pemetrexed, improved the median overall survival. However, it did not influence the progression-free survival rate [22].
Moreover, in another study, Hassan et al. firmly pointed out the potential induction of innate and adaptive response, mainly CD4+- and CD8+-specific T-cells, to non-virulent Listeria monocytogenes bacterium-expressing mesothelin surface glycoprotein (CRS-207 construct) [23]. A Phase 2 study performed on patients with an advanced stage of mesothelioma has shown that CRS-207 was well-tolerated, with the observed reduction of tumor size. The CRS-207 therapy alone and before the standard chemotherapy rearranges the number of circulating immune cells, providing an effector response of modified tumor microenvironment to tumor cells.
In summary, currently, there are several widespread targets in studies of mechanism and MPM therapy, as collected in Figure 3. They focus on inflammation reduction (IL-6 and IL-8), immune system failure (by blocking CTLA-4 and PD-1 receptors on unresponsive T-cells), invasion and metastasis (by anti-mesothelin as well as anti-CD13 expressed on tumor endothelium), but also angiogenesis (via an anti-VEGF approach) as well as direct cell death (by HDAC inhibitors, which induce tumor-cell-selective expression of pro-apoptotic genes). The miRNAs [24] and ASS-1 remain obvious targets for genomic alterations, whereas changes in cellular energetics, oxidative stress, or processes of replicative immortality still remain new and are not well-investigated and incorporated into the trials. Therefore, further in-depth research on possible targets is still ongoing, in both in vitro and in vivo models.

3. Polyphenols and Natural Compounds in Cancer Treatment

Numerous plant-derived dietary supplements have been used for years as a natural support of the body in handling inflammatory processes, treating various metabolic diseases, or improving the immune system. The extracts obtained from natural sources, including green tea, grape seeds, and borage seeds, are characterized by a rich polyphenol composition and well-documented antioxidant properties [26].
Polyphenols are secondary metabolites that belong to the phenolic family of naturally occurring compounds present in plants’ roots, leaves, and seeds. Chemically, they consist of phenol structures divided into several subclasses depending on the number of phenolic rings and the arrangement of substituent groups (hydroxyl, alkyl, and glycosyl groups) that bind to these rings. In general, polyphenols are classified into flavonoids (consisting of flavonols, flavones, flavanols, flavanones, isoflavones, and anthocyanidins) and non-flavonoid compounds with phenolic acids, stilbenes, lignans, and other polyphenols. The main representatives of these phytochemicals are listed in Table 1.
In the light of the revealed significant antioxidant activity and protective role in the whole human body of polyphenols and other natural compounds, phytochemicals are increasingly considered dietary supplements in anticancer therapy [27]. Recent studies demonstrated that polyphenol-rich plant extracts abolish common side effects, such as cardiotoxicity and nephrotoxicity, associated with standard chemotherapy in tumor-bearing patients [28]. For instance, Zhang et al. summarized in detail the beneficial results of penta-O-galloyl-β-D-glucose (PGG), a gallotannin that is a penta-gallic acids ester of glucose (Figure 4), not only in diabetes treatment but particularly in anticancer therapies including prostate, lung, and breast cancers. PGG can reveal antitumor potential through several mechanisms, including anti-angiogenic and metastatic properties (through the VEGF inhibition and direct diminishing the activity of matrix metalloproteinase-9, MMP-9), antiproliferative actions (through S-phase perturbation and immediate G0/G1 growth phase arrest), and the anti-inflammatory and antioxidant role in normal, healthy cells (by scavenging reactive oxygen species (ROS) in a dose-dependent manner) [29]. It seems that several key carcinogenic molecular players are targets for not only PGG antitumor activity but also other phenolic derivatives. These include nuclear factor-kappaB (NF-κB) and tumor suppressor genes (P53 and retinoblastoma, Rb) [30].
Interestingly, PGG may also reverse drug resistance in treated malignant cells by directly inhibiting P-glycoprotein, the cell membrane protein responsible for rejecting therapeutic drugs out of cancer cells [31]. Furthermore, Ryu et al. demonstrated for the first time that PGG decreased the cytotoxicity effect of cisplatin in renal cells by reducing reactive oxygen species generation [32]. Additionally, the increased sensitization of Caki-2 renal cancer cells to the antitumor activity of cisplatin in the presence of PGG was observed, which strongly indicates a positive role of PGG supplementation strategy.
Recently, there have been many reports on the beneficial anticancer properties of polyphenolic compounds. The molecular mechanisms of various valuable phenolic compounds are still under deep investigation in either in vitro or in vivo models [27,29,33]. Numerous reports have shown either the modulatory effect or direct interaction of phenolic compounds with the particular oncogenes, affecting the major oncogenic signal transduction pathways involved in carcinogenesis and cancer progression. Moreover, several studies revealed the potential antitumor action of mixtures of selected polyphenolic compounds. The most commonly used combined treatment strategies with curcumin, flavonoid epigallocatechin gallate (EGCG), or resveratrol show good prognostic results in inhibiting tumor cell proliferation and arresting cancer cell growth. The promising effects of polyphenol combinations or natural extracts, in low doses, have also been seen in several cancer types, including lung cancer, breast cancer, prostate cancer, and leukemia cells [34,35,36,37]. In particular, polyphenolic compounds can be successfully administered in conjugation with standard chemotherapeutics overcoming the drug resistance phenomenon [27].

4. Polyphenols in Malignant Pleural Mesothelioma Therapy

Polyphenols are a diverse group of natural compounds that may act on the cells either in an antioxidative or pro-oxidative way, depending on the concentration, cell type, and operating conditions. Their biological pro-oxidative properties are essential for cancer treatment since the excessive generation of ROS leads to the breakdown of DNA, inhibiting cell growth and death. The cancer cells are more susceptible to ROS than normal ones [38]. ROS mediate apoptosis, necrosis, and autophagy processes that are desirable targets in anticancer therapies. Additionally, ROS contribute to the increased cytotoxicity of chemotherapeutic agents in combined treatment strategies with polyphenol compounds.
Curcumin, widely studied for its activity [39,40], was one of the first compounds reported to cause mesothelioma cell death, partially by direct stimulation of cell apoptosis [41]. The MPM cell growth suppression induced by curcumin took place via direct stimulation of Bax proteins and caspase-9, both engaged in initiating the apoptotic pathway. Moreover, Wang et al. revealed for the first time that curcumin was also responsible for the induction of expression of novel transducing molecules, including XIAP-associated factor-1 (XAF1) and CARP1/CCAR1 proteins, responsible for signaling to apoptosis. Finally, they demonstrated that curcumin treatment stimulated the expression of tumor suppressor protein SULF1, which could influence multiple tyrosine kinase-signaling pathways, making SULF1 a potential molecular target for MPM and other cancer treatment options. Polyphenols, including curcumin, can modulate immune cells involved in tumor development and control inflammatory processes that engage cytokines and further ROS generation [42]. Miller et al. observed that in MPM cells, curcumin enhanced activation of caspase-1, an interleukin-conversing enzyme (ICE), physiologically responsible for maturing pro-inflammatory cytokines interleukin-1β and -18. However, the authors also found a higher concentration of pro-IL-1β, which evidenced an anti-inflammatory effect of curcumin on MPM cells, leading to lytic programmed cells death [43].
Moreover, curcumin-mediated apoptosis was related to the increased Bax protein, p53 expansion, activation of pro-apoptotic caspase-9, and accumulation of cells at the Sub-G1 phase [44]. Beyond the influence on the cell cycle, curcumin treatment led to phosphorylation of ERK1/2 and p38 mitogen-activated protein kinase (MAPK), blockage of NF-κB nuclear translocation, and JNK pathway inhibition. In the Masueli et al. study, treatment of the murine model of MPM with curcumin reduced tumor development and increased the median survival of an experimental animal. Additionally, the curcumin combined with another natural compound, piperine (commercially available as C3 complex® and Bioperine®), reduced the tumorigenic properties of mesothelioma cells, including cell proliferation, colonization, invasion, and reduced angiogenesis in a mesothelioma xenograft mouse model [45]. Under the daily intraperitoneal administration of C3 complex/Bioperine®, a significant delay in the growth of tumor cells was observed and a decrease in ectopic cell proliferation and the number of vessels and increased apoptotic index were revealed. Pouliguen et al. demonstrated the inhibitory effect of curcumin on mesothelioma cells’ motility and further tumor metastasis and liver colonization [46]. From the clinical point of view, the most significant advantage of curcumin is its biosafety and immediate interaction [47]. Thus, the direct intrapleural (local) administration is still further investigated as a future alternative for patients with pleural cavity tumors, including MPM [48].
Epigallocatechin-3-gallate (EGCG) is another compound extensively investigated against mesothelioma cells [49]. There are three concomitant reports that complement each other. In all cases, EGCG treatment has induced mesothelioma cell death. The studies on the mechanism of EGCG action indicated the pro-oxidative properties of this polyphenol at higher therapeutic doses. EGCG induced the formation of reactive oxygen species, including hydrogen peroxide and superoxide, outside of the cells while impairing mitochondrial membrane potential [50,51]. This flavanol decreased the MPM proliferation and led to mitochondria apoptosis only in tumor-changed cells. EGCG has also been successfully combined in reducing MPM cell viability with ascorbic acid and gemcitabine, the latter being the cytostatic antimetabolite, an analog of pyrimidine 2′-deoxycytidine [52]. The combined treatment of EGCG with gemcitabine led to non-inflammatory apoptosis of REN MPM cells by downregulation of the p65 subunit of NF-κB, restoring programmed cells death [53]. EGCG has also been shown to modulate the constitutive for tumor cells unfold protein process response (UPR) by accumulating molecular chaperone GRP78 in the endoplasmic reticulum (ER). The increased GRP78 protein in ER promotes the activation of pro-apoptotic caspases 3 and 8, together with an increased expression of transcription factors directly related to stress-responsive genes, including cyclin AMP-dependent transcription factor (ATF-4) and X-box-binding protein 1 (XBP1) [54]. These observations indicated the reasonable possibility of the therapeutic use of EGCG in MPM treatment.
The beneficial effect of resveratrol (3,5,4′-trihydroxystilbene) with clofarabine or cisplatin as chemotherapeutics was observed in the in vitro model mesothelioma MSTO-H211 cells [55,56,57]. In the presence of a low dose of the chemotherapeutic agent (clofarabine or cisplatin), resveratrol caused inhibition of MPM cell growth and initiation of apoptotic processes. Upon the ROS-mediated inflammatory process, activation of the Nrf2 transcription factor (nuclear factor E2-related factor 2) is observed. Under the physiological state, Nrf2 is ubiquitously sequestered by Keap-1 (Kelch-like ECH-associated protein). However, when MPM arises, Keap-1 undergoes a conformational change, releasing Nrf2, which translocates to the nucleus and activates the transcription of anti-inflammatory genes [58]. It has been established that the increased Nrf2 level created is favorable for the MPM immune-resistant environment. Lee YJ et al. reported that the observed restoration of MPM cell sensitivity to clofarabine in the presence of resveratrol was accompanied by downregulation of Nrf2 [55]. Resveratrol contributed to overcoming the chemoresistance of MPM cells to clofarabine via a p53-dependent apoptotic pathway with significant accumulation of tumor cells in the G1 phase and increased caspase-3/7 activity [56].
On the other hand, the synergistic activity of resveratrol and cisplatin led to increased ROS production and induced apoptosis through promoted mitochondria oxidative damage events [57]. The most significant role in oncogenesis is transcription factor Specificity Protein 1 (Sp1). That protein is abundantly expressed in different tumor cells and controls the transcription of oncogenes and tumor suppressors and genes involved in cancer proliferation, differentiation, apoptosis, genetic material damage responses, and so on [59]. Thus, this particular signaling factor seems to be a promising target for anticancer management. Lee KA et al. demonstrated that resveratrol induced growth arrest of MSTO-H211 cells at the Sub-G1 phase in vitro study and directly suppressed Sp1 protein level and expression of Sp1 regulatory genes cyclin D1, p21, p27, surviving, and myeloid cell leukemia (Mcl-1) [60]. Concomitantly, Lee YJ et al. confirmed that resveratrol combined with clofarabine reduced the nuclear accumulation of Sp1 and decreased the levels of p-Akt, Sp1, c-Met, cyclin D1, and p21 in MSTO-H211 cells upon the synergistic antiproliferative [56]. Finally, resveratrol alone reduced MSTO-H211 tumor growth in a BALB/c athymic (nu+/nu+) mice animal model via the inhibition of Sp1 expression. The authors observed a decreased tumor volume and activation of pro-apoptotic signals transduction [60]. The Mcl-1 protein seems to be a direct downstream target, responsible for the observed effect of treatment of malignant mesothelioma cells with resveratrol and clofarabine, locking the apoptotic pathway. The downregulation of Mcl-1 protein after administrating the resveratrol and clofarabine formulation restored the growth-inhibitory effect and induced the caspase-3 activation pathway [61].
Analogously, quercetin, another plant flavonoid, revealed a potential inhibitory effect on human MSTO-H211 cell viability. In the study pf Chae JI et al., quercetin increased the population of Sub-G1 cells via interaction with the Sp1 transcription factor, significantly suppressing its expression at protein and mRNA levels [62]. Quercetin caused downregulation of expression of the Sp1-related genes (cyclin D1, Mcl-1, and survivin) and activated pro-apoptotic caspase-3 signals, triggering apoptotic cell death. Quercetin, in combination with cisplatin, revealed a positive mode of action. In the in vitro study on SCP212 and SCP111 cells, quercetin reduced MPM cell proliferation and activated caspase-9 and caspase-3, leading to apoptosis [63]. Additionally, the combinatory dose of quercetin and cisplatin was more effective in MPM treatment than the individual agents. Demiroglu-Zergeroglu et al. confirmed in another study that quercetin alone and in combination with cisplatin modulated the expression of genes involved in cell-cycle progression, mainly cyclins and cyclin-related molecular pathways, with parallel upregulated expression of MAPK-related genes [64]. Quercetin and cisplatin triggered S-phase cell cycle arrest via increased cyclin-dependent kinase inhibitor genes (CDI) and forced cell death by activating p38 and JNK pathways.
Epidermal growth factor receptor (EGFR) is one of the main targets in mesothelioma treatment strategies due to its overexpression reaching 75% in MPM patients. Until now, gallic acid was one of the phenolic acids that showed EGFR-dependent pro-apoptotic and antiproliferative activity in malignant pleural mesothelioma cells [65]. GA induced transient cancer cells growth arrest in the G1 phase and mitochondrial dysfunction, leading to apoptosis. The mechanism underlying its biological activity was tightly related to p38 MAPK inactivation. After GA treatment, the reduced viability of mesothelioma cells resulted in a down-regulated cyclin D and Bcl-2 gene expression and simultaneous upregulated expression of p21, genes directly involved in cell cycle progression and apoptotic signals.
Cafestol and kahweol, two diterpenes present in the beans of coffee Arabica, are among the natural compounds also investigated for MPM treatment [66]. These bioactive compounds induced MPM cell apoptosis again via downregulation of the Sp1 protein level and downstream Sp1 gene-related regulatory genes, including cyclin D, Mcl1, and survivin. Moreover, the single use of either kahweol or cafestol chemical led to increased pro-apoptotic signals in MSTO-H211 cells via the regulation of Bcl-2-related proteins (upregulation of Bax, downregulation of Bcl-xL), and activation of Bid, caspase-3, and PARP, respectively. The combinatory dose of kahweol and cafestol has a chemopreventive effect on an animal model, where a mixture of these two compounds inhibited carcinogen-activated hepatic cytochrome P450 (CYP450) and sulfotransferase (SULT) [67].
Lee KA et al. [68] demonstrated that hesperidin, a bioflavonoid from citrus fruits, significantly reduced the Sp1 at protein and mRNA levels. Furthermore, this flavanone glycoside suppressed the viability of MSTO-H211, leading to the accumulation of cells at the Sub-G1 phase, and induced an apoptotic pathway by the cleavage of Bid, caspase-3, and PARP, as well as upregulation of Bax and reduction of Bcl-xL protein levels. Analogously, Chae JI et al. described that honokiol, an active component of the Magnolia genus, used in traditional Chinese medicine, similarly inhibited MPM cell growth to the above-discussed diterpenes from coffee, quercetin, and citrus-derived hesperidin via downregulation of Sp1 expression and its target transcription genes [69]. Like other above-discussed natural compounds, honokiol also led to mesothelioma cell growth arrest in the Sub-G1 phase and induced apoptosis-signaling cascade events, with observed Bax activation caspase-3 and PARP with parallel reduction of anti-apoptotic Bid and Bcl-xL proteins. The equal contribution of the Sp1 transcription factor was documented for licochalcone A (LicA), a natural compound extracted from the roots of licorice species Glycyrrhiza ssp. LicA is a well-known anti-inflammatory agent that protects sensitive and injured skin and serves as a beneficial component of dermo-cosmetics [70]. Kim HK et al. demonstrated that the isolated and purified LicA regulated the growth and proliferation of the investigated MPM cell lines and arrested the cell cycle at the Sub-G1 phase [71]. Furthermore, LicA down-regulated the expression of Sp1 and consequently its downstream genes, as cyclin D1, Mcl1, and survivin. Finally, as others mentioned, LicA induced the intrinsic mitochondrial apoptotic pathway by modulating pro-apoptotic and anti-apoptotic proteins of the Bcl-2 family. The simplified scheme of molecular pathways modulated by natural compounds is present in Figure 5.
Withaferin A (WA), isolated from the Withania somnifera, an Indian medicinal plant, is among the compounds studied against MPM. This steroidal lactone exerts potential anti-inflammatory, anti-angiogenic, and anti-metastatic properties that target multiple molecular pathways, including NF-κB, PI3K/AKT/mTOR signaling pathways, and the generation of ROS, mitochondrial dysfunction, and proteasome activity inhibition [72]. Several molecular alternations were observed in both in vivo murine allografts and in vitro patient-derived MPM models after WA treatment. WA caused tumor growth inhibition, involving direct apoptosis, the increase in pro-apoptotic proteins (p38 stress-activated caspase-3, elevated Bax, PARP cleavage, stimulated expression of CARP1/CCAR1), and blockage in the chymotryptic activity of proteasome [73].
Given the recent miRNA targeting MPM therapy, it is impressively attractive that phytochemicals seem to be reasonable miRNA regulatory tools. A broad range of natural compounds has been tested for unique miRNA regulatory properties, including curcumin, EGCG, resveratrol, quercetin, and others [74]. So far, only ursolic acid, presented in leaves, fruits, flowers, and berries, has a documented role in epithelial mesothelioma miRNA expression [75]. In the light of altered gene expression patterns observed after the administration of particular polyphenols and documented in other various cancer types’ miRNA modulatory potential, dietary phytochemicals appeared to be a promising additional option for MPM patients.
The discussed above studies confirm the potential anticancer activity of polyphenols and other natural compounds that can directly induce apoptosis of mesothelioma cells and increase MPM cells’ sensitivity to chemotherapy. The summary of the current results of anticancer treatment strategies for malignant pleural mesothelioma, with natural compounds, is presented in Table 2.
Besides the intensively conducted research on the single or combined treatment of particular polyphenolic compounds, several reports demonstrate the antitumor effects of complex mixtures of natural extracts obtained from parts of plants or isolated from secondary post-production wastes.
The first pioneering work on the nutrient mixture composed of green tea extract, vitamin C, amino acids (L-arginine, L-lysine, and proline) and microelements, manganese, and copper was reported by Roomie et al. They revealed a potential therapeutic application of well-balanced supplementation against the MPM cells [76]. The authors proved the inhibitory effect of this composition on the MMPs activity and the level of their secretion by mesothelial cells in a dose-dependent manner (below 500 µg/mL), at the same time ensuring the strengthening of the extracellular matrix components and affecting the ability of malignant mesothelial cells to invade the matrix and expand. Moreover, Roomie’s team highlighted the safety issue of the tested preparation, indicating the lack of side effects on organs and serum enzymes in animal models compared to chemotherapy agents [77]. Likewise, the previously discussed example, Pulito et al., revealed encouraging results in both in vitro and in vivo models of malignant pleural mesothelioma cells treated with artichoke leaf (Cynara scolymus) extract [78]. Treatment with the artichoke leaf extract, enriched in phenolic compounds, strongly affected MPM cell growth by inducing apoptosis and impaired the migration and invasion of mesothelioma cells in the first 12 h after administration. Furthermore, by introducing the extract into the diet, these authors observed significantly reduced MPM tumor size in mouse xenografts, this affected the engraftment of the MPM cell line after C. scolymus extract treatment. Recently, we reported that a mixture of polyphenols obtained from Oenothera paradoxa seeds (evening primrose extract, EPE) revealed anti-invasive effects against MPM cells [79]. Evening primrose (Oenothera paradoxa Hudziok), belonging to the Oenothera species, is used in Europe for edible oil production. It is well-established that the oil obtained from Oenothera species seeds is rich in γ-linolenic acid and exhibits anti-diabetic and anti-inflammatory effects. Nowadays, it is widely used in hypercholesterolemia and obesity treatment and the therapy of atopic eczema, premenstrual syndrome, and multiple sclerosis [80]. Several studies on the determination of valuable compounds of defatted EPE seeds demonstrated significantly high polyphenols content comparable to polyphenols presented in extracts obtained from green tea or grapefruit seeds [81]. Pharmacological studies have revealed that the extracts obtained from O. biennis and O. paradoxa have biomedical properties, demonstrating the antioxidant and iron (II) chelating activity [82,83]. Analysis of the individual components of Oenothera paradoxa seeds extracts led to the identification of biologically active compounds, including gallic acid and PGG, which, apart from anti-inflammatory and antioxidant properties [84,85], displayed a selective pro-apoptotic activity against certain types of tumors [29,86]. The influence of EPE from evening primrose post-industrial seeds on cancer proliferation has been studied before [87,88,89]. Depending on the extraction method, these preparations vary in the total amount of polyphenols and composition; nevertheless, the presence of the main polyphenol compounds, including gallic acid, epicatechin, PGG, and procyanidins, make the extract valuable for therapeutic purposes. So far, research conducted on human melanoma, colon cancer, and Ehrlich ascites cells confirmed that both EPE and its main compound PGG could induce apoptosis in these cells in a pro-oxidative manner [90,91,92]. On the other hand, Lewandowska et al. have shown potential anti-metastatic properties of EPE against prostate and breast cancer types, namely by inhibiting the type IV collagenases (MMP-2 and MMP-9) responsible for cell migration, invasiveness, and metastasis [93]. Our latest study [79] revealed that EPE possesses the anti-metastatic specificity toward invasive MPM cancer cell types (MSTO-H211 and JU77). The extract, containing ellagic acid, gallic acid, (+)-catechin, PGG, and procyanidin B2, arrested invasive cells in the G2/M phase, negatively correlated with the migration of MPM cells, and inhibited the secretion of matrix metalloproteinase-7 (MMP-7), the latter factor directly engaged in the invasion process and metastasis. Considering that MPM is a cancer with one of the worst prognoses, which increases rapidly in the advanced stage, this property of EPE makes it valuable in inhibiting the progression and worsening of this malignancy. It was reported previously by Jaszewska et al. that the O. paradoxa extract containing PGG and procyanidins potentiates the cytotoxicity of the chemotherapeutic agent vincristine in human metastatic melanoma cells (HTB-140) [94]. The authors confirmed the extract’s potency in more invasive cells than hepatoma cells (HepG2), indicating the potential pro-oxidative activity of its components on tumor cells that bear elevated intracellular levels of the reduced form glutathione (GSH). Following this premise, we evaluated the activity of individual constituents of EPE preparation. We did not observe the potency of a single use of phenolic compounds (unpublished data), raising the rationale for the extensive use of natural extracts. Our ongoing results provide evidence for further in-depth analysis of the molecular mechanisms of EPE in MPM tumor-growth suppression. In the light of recent reports, it seems that the polyphenol-rich extracts obtained from Oenothera sp. have a dual biological effect, acting as an antioxidant and a protective agent toward normal cells and revealing anticancer activity toward MPM cells, with concomitant support of the standard chemotherapy.
Regarding the significant limitations to the successful treatment of malignant pleural mesothelioma, including severe side effects and chemoresistance, phytochemicals seem to be a reasonable approach to alleviating the aggressiveness of standard doses of chemotherapeutic agents while sensitizing cancer cells to treatment and protecting healthy tissues and organs. The involvement of natural extracts in the MPM treatment regimen might improve the tolerance and efficacy of the therapy.

5. Conclusions

Malignant pleural mesothelioma therapy is still an emerging issue due to the ineffective treatment options, especially in late-diagnosed advanced-stage patients. The main frontline of the MPM treatment strategy is surgery, curative intents, or palliative chemotherapy. New therapeutic approaches involving targeted strategies and immunotherapies and novel treatment options, considering combinatory treatment and involvement of natural compounds and plants-origin extracts, are still needed. Herein, we discussed the best with today’s knowledge of the research studies that consider targeted therapies and immunotherapies in MPM tumor treatment, focusing on the natural compounds’ phytotherapy. The beneficial effects of polyphenols and plant-derived extracts, including Oenothera paradoxa preparation, on the treatment of MPM neoplasm, presented in this work, provide a new opportunity to engage pure nature compositions in inhibiting MPM cancer progress. We take a step further to influence this malignancy with the in-depth coverage of the molecular and cellular pathways involved in polyphenols activity. We hope that, as in the case of other solid tumors, including melanoma and non-small cell lung carcinoma, MPM patients will soon gain benefits from long-lasting disease control and a better prognosis for the future.

Author Contributions

Conceptualization, M.C.-K.; resources, M.C.-K.; data curation, M.C.-K.; writing—original draft preparation, M.C.-K.; writing—review and editing, M.C.-K. and L.A.W.; visualization, M.C.-K.; supervision, L.A.W.; project administration, M.C.-K.; funding acquisition, M.C.-K. All authors have read and agreed to the published version of the manuscript.

Funding

This review study was supported by National Science Centre, Poland, grant no. 2015/19/N/NZ3/01497.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stahel, R.A.; Weder, W.; Lievens, Y.; Felip, E. Malignant pleural mesothelioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. On behalf of the ESMO Guidelines Working Group. Ann. Oncol. 2010, 21 (Suppl. 5), v126–v128. [Google Scholar] [CrossRef] [PubMed]
  2. Parkin, D.M.; Bray, F.; Ferlay, J.; Pisani, P. Global cancer statistics, 2002. CA Cancer J. Clin. 2005, 55, 74–108. [Google Scholar] [CrossRef] [PubMed]
  3. Kameda, T.; Takahashi, K.; Kim, R.; Jiang, Y.; Movahed, M.; Park, E.K.; Rantanen, J. Asbestos: Use, bans and disease burden in Europe. Bull. World Health Organ 2014, 92, 790–797. [Google Scholar] [CrossRef] [PubMed]
  4. Acencio, M.M.; Soares, B.; Marchi, E.; Silva, C.S.; Teixeira, L.R.; Broaddus, V.C. Inflammatory cytokines contribute to asbestos-induced injury of mesothelial cells. Lung 2015, 193, 831–837. [Google Scholar] [CrossRef] [PubMed]
  5. Carbone, M.; Yang, H. Molecular pathways: Targeting mechanisms of asbestos and erionite carcinogenesis in mesothelioma. Clin. Cancer Res. 2012, 18, 598–604. [Google Scholar] [CrossRef] [Green Version]
  6. Yang, H.; Rivera, Z.; Jube, S.; Nasu, M.; Bertino, P.; Goparaju, C.; Franzoso, G.; Lotze, M.T.; Krausz, T.; Pass, H.I.; et al. Programmed necrosis induced by asbestos in human mesothelial cells causes high-mobility group box 1 protein release and resultant inflammation. Proc. Nat. Acad. Sci. USA 2010, 107, 12611–12616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Zucali, P.A.; Ceresoli, G.L.; de Vincenzo, F.; Simonelli, M.; Lorenzi, E.; Gianoncelli, L.; Santoro, A. Advances in the biology of malignant pleural mesotheliomamesothalioma. Cancer Treat. Rev. 2011, 37, 543–558. [Google Scholar] [CrossRef]
  8. Yang, H.; Bocchetta, M.; Kroczynska, B.; Elmishad, A.G.; Chen, Y.; Liu, Z.; Bubici, C.; Mossman, B.T.; Pass, H.I.; Testa, J.R.; et al. TNF-alpha inhibits asbestos-induced cytotoxicity via a NF-kappa-B-dependent pathway, a possible mechanism for asbestos-induced oncogenesis. Proc. Natl. Acad. Sci. USA 2006, 103, 10397–10402. [Google Scholar] [CrossRef] [Green Version]
  9. Jean, D.; Daubriac, J.; Le Pimpec-Barthes, F.; Galateau-Salle, F.; Jaurand, M.C. Molecular changes in mesothelioma with an impact on prognosis and treatment. Arch. Pathol. Lab. Med. 2012, 136, 277–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Xu, J.; Kadariya, Y.; Cheung, M.; Pei, J.; Talarchek, J.; Sementino, E.; Tan, Y.; Menges, C.W.; Cai, K.Q.; Litwin, S.; et al. Germline mutation of BAP1 accelerates development of asbestos-induced malignant mesothelioma. Cancer Res. 2014, 74, 4388–4397. [Google Scholar] [CrossRef] [Green Version]
  11. Szlosarek, P.W.; Klabatsa, A.; Pallaska, A.; Sheaff, M.; Smith, P.; Crook, T.; Grimshaw, M.J.; Steele, J.P.; Rudd, R.M.; Balkwill, F.R.; et al. In Vivo loss of expression of argininosuccinate synthetase in malignant pleural mesothelioma is a biomarker for susceptibility to arginine depletion. Clin. Cancer Res. 2006, 12, 7126–7131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chew, S.H.; Toyokuni, S. Malignant mesothelioma as an oxidative stress-induced cancer: An update. Free Radic. Biol. Med. 2015, 86, 166–178. [Google Scholar] [CrossRef] [PubMed]
  13. Scherpereel, A.; Wallyn, F.; Albelda, S.M.; Munck, C. Novel therapies for malignant pleural mesothelioma. Lancet Oncol. 2018, 19, e161–e172. [Google Scholar] [CrossRef]
  14. Bonelli, M.A.; Fumarola, C.; La Monica, S.; Alfieri, R. New therapeutic strategies in malignant pleural mesothelioma. Biochem. Pharmacol. 2017, 123, 8–18. [Google Scholar] [CrossRef]
  15. Delage, B.; Fennell, D.A.; Nicholson, L.; McNeish, L.; Lemoine, N.R.; Crook, T.; Szlosarek, P.W. Arginine deprivation and argininosuccinate synthetase expression in the treatment of cancer. Int. J. Cancer 2010, 126, 2762–2772. [Google Scholar] [CrossRef]
  16. Ensor, C.M.; Holtsberg, F.W.; Bomalaski, J.S.; Clark, M.A. Pegylated arginine deiminase (ADI-SS PEG20,000mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo. Cancer Res. 2002, 62, 5443–5450. [Google Scholar]
  17. Allen, M.; Luong, P.; Hudson, C.; Leyton, J.; Delage, B.; Ghazaly, E.; Cutts, R.; Yuan, M.; Syed, N.; Lo Nigro, C.; et al. Prognostic and therapeutic impact of argininosuccinate synthase-1 control in bladder cancer as monitored longituf=dinally by PET imaging. Cancer Res. 2014, 74, 896–907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Szlosarek, P.W.; Steele, J.P.; Nolan, L.; Gilligan, D.; Taylor, P.; Spicer, J.; Lind, M.; Mitra, S.; Shamash, J.; Phillips, M.M.; et al. Arginine deprivation with pegylated arginine deiminase in patients with argininosuccinate synthetase 1–deficient malignant pleural mesothelioma: A randomized clinical trial. JAMA Oncol. 2017, 3, 58–66. [Google Scholar] [CrossRef]
  19. Phillips, M.M.; Szyszko, T.; Hall, P.; Cook, G.J.R.; Khadeir, R.; Steele, J.P.C.; Spicer, J.F.; Feng, X.; Hategan, M.; Rashid, S.; et al. Expansion study of ADI-PEG 20, pemetrexed and cisplatin in patients with ASS1-deficient malignant pleural mesothelioma (TRAP). Proc. Am. Soc. Clin. Oncol. 2017, 35 (Suppl. 15), 8553. [Google Scholar] [CrossRef]
  20. Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell 2015, 27, 450–461. [Google Scholar] [CrossRef] [Green Version]
  21. Hotta, K.; Fujimoto, N. Current evidence and future perspectives of immune-checkpoint inhibitors in unresectable malignant pleural mesothelioma. J. Immunother Cancer 2020, 8, e000461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hassan, R.; Kindler, H.L.; Jahan, T.; Bazhenova, L.; Reck, M.; Thomas, A.; Pastan, I.; Parno, J.; O’Shannessy, D.J.; Fatato, P.; et al. Phase II clinical trial of amatuximab, a chimeric anti-mesothelin antibody with pemetrexed and cisplatincisplatin in advanced unresectable pleural mesothelioma. Clin. Cancer Res. 2014, 20, 5927–5936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hassan, R.; Alley, E.; Kindler, H.; Antonia, S.; Jahan, T.; Honarmand, S.; Nair, N.; Whiting, C.C.; Enstrom, A.; Lemmens, E.; et al. Clinical Response of Live-Attenuated, Listeria monocytogenes Expressing Mesothelin (CRS-207) with Chemotherapy in Patients with Malignant Pleural Mesothelioma. Clin. Cancer Res. 2019, 25, 5787–5798. [Google Scholar] [CrossRef]
  24. Reid, G.; Johnson, T.G.; van Zandwijk, N. Manipulating microRNAs for the Treatment of Malignant Pleural Mesothelioma: Past, Present and Future. Front. Oncol. 2020, 10, 105. [Google Scholar] [CrossRef] [PubMed]
  25. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  26. Bukowiecka-Matusiak, M.; Turek, I.A.; Woźniak, L.A. Natural Phenolic Antioxidants and Their Synthetic Derivatives. In Systems Biology of FreeRadicals and Antioxidants; Laher, I., Ed.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 4047–4061. [Google Scholar]
  27. Fantini, M.; Benvenuto, M.; Masuelli, L.; Frajese, G.V.; Tresoldi, I.; Modesti, A.; Bei, R. In Vitro and in vivo antitumoral effects of combinations of polyphenols, or polyphenols and anticancer drugs: Perspectives on cancer treatment. Int. J. Mol. Sci. 2015, 16, 9236–9282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Piasek, A.; Bartoszek, A.; Namieśnik, J. Phytochemicals that counteract the cardiotoxic side effects of cancer chemotherapy. Postepy Hig. Med. Dosw. 2009, 63, 142–158. [Google Scholar]
  29. Zhang, J.; Li, L.; Kim, S.H.; Hagerman, A.E.; Lü, J. Anti-cancer, anti-diabetic and other pharmacologic and biological activities of penta-galloyl-glucose. Pharm. Res. 2009, 26, 2066–2080. [Google Scholar] [CrossRef] [Green Version]
  30. Oh, G.S.; Pae, H.O.; Oh, H.; Hong, S.G.; Kim, I.K.; Chai, K.Y.; Yun, Y.G.; Kwon, T.O.; Chung, H.T. In Vitro antiproliferative effect of 1,2,3,4,6-penta-O-galloyl-beta-D-glucose on the human hepatocellular carcinoma cell line, SK-HEP-1 cells. Cancer Lett. 2001, 174, 17–24. [Google Scholar] [CrossRef]
  31. Kitagawa, S.; Nabekura, T.; Nakamura, Y.; Takahashi, T.; Kashiwada, Y. Inhibition of P-glycoprotein function by tannic acid and pentagalloylglucose. J. Pharm. Pharmacol. 2007, 59, 965–969. [Google Scholar] [CrossRef]
  32. Ryu, H.G.; Jeong, S.J.; Kwon, H.Y.; Lee, H.J.; Lee, E.O.; Lee, M.H.; Choi, S.H.; Ahn, K.S.; Kim, S.H. Penta-O-galloyl-β-D-glucose attenuates cisplatin-induced nephrotoxicity via reactive oxygen species reduction in renal epithelial cells and enhances antitumor activity in Caki-2 renal cancer cells. Toxicol. In Vitro 2012, 26, 206–214. [Google Scholar] [CrossRef] [PubMed]
  33. Ceci, C.; Lacal, P.M.; Tentori, L.; de Martino, M.G.; Miano, R.; Graziani, G. Experimental Evidence of the Antitumor, Antimetastatic and Antiangiogenic Activity of Ellagic Acid. Nutrients 2018, 10, 1756. [Google Scholar] [CrossRef] [Green Version]
  34. Zhou, D.H.; Wang, X.; Yang, M.; Shi, X.; Huang, W.; Feng, Q. Combination of low concentration of (-)-epigallocatechin gallate (EGCG) and curcumin strongly suppresses the growth of non-small cell lung cancer in vitro and in vivo through causing cell cycle arrest. Int. J. Mol. Sci. 2013, 14, 12023–12036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Somers-Edgar, T.J.; Scandlyn, M.J.; Stuart, E.C.; Le Nedelec, M.J.; Valentine, S.P.; Rosengren, R.J. The combination of epigallocatechin gallate and curcumin suppresses ER alpha-breast cancer cell growth in vitro and in vivo. Int. J. Cancer 2008, 122, 1966–1971. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, P.; Wang, B.; Chung, S.; Wu, Y.; Henning, S.M.; Vadgama, J.V. Increased chemopreventive effect by combining arctigenin, green tea polyphenol and curcumin in prostate and breast cancer cells. RSC Adv. 2014, 4, 35242–35250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mertens-Talcott, S.U.; Percival, S.S. Ellagic acid and quercetin interact synergistically with resveratrol in the induction of apoptosis and cause transient cell cycle arrest in human leukemia cells. Cancer Lett. 2005, 218, 141–151. [Google Scholar] [CrossRef] [PubMed]
  38. León-Gonzáles, A.J.; Auger, C.; Schini-Kerth, V.B. Pro-oxidant activity of polyphenols and its implication on cancer chemoprevention and chemotherapy. Biochem. Pharmacol. 2015, 98, 371–380. [Google Scholar] [CrossRef]
  39. Wojcik, M.; Krawczyk, M.; Wojcik, P.; Cypryk, K.; Wozniak, L.A. Molecular Mechanisms Underlying Curcumin-Mediated Therapeutic Effects in Type 2 Diabetes and Cancer. Oxidative Med. Cell. Longev. 2018, 2018, 1–14. [Google Scholar] [CrossRef] [Green Version]
  40. Baldi, A.; De Luca, A.; Maiorano, P.; D’Angelo, C.; Giordano, A. Curcumin as an anticancer agent in malignant mesothelioma: A review. Int. J. Mol. Sci. 2020, 21, 1839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Wang, Y.; Rishi, A.K.; Wu, W.; Polin, L.; Sharma, S.; Levi, E.; Albelda, S.; Pass, H.I.; Wali, A. Curcumin suppresses the growth of mesothelioma cells in vitro and in vivo, in part, by stimulating apoptosis. Mol. Cell Biochem. 2011, 357, 83–94. [Google Scholar] [CrossRef] [Green Version]
  42. Ghiringhelli, F.; Rebe, C.; Hichami, A.; Delmas, D. Immunomodulation and anti-inflammatory roles of polyphenols as anticancer agents. Anticancer Agents Med. Chem. 2012, 12, 852–873. [Google Scholar] [CrossRef]
  43. Miller, J.M.; Thompson, J.K.; MacPherson, M.B.; Beuschel, S.L.; Westbom, C.M.; Sayan, M.; Shukla, A. Curcumin: A double hit on malignant mesothelioma. Cancer Prev. Res. 2014, 7, 330–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Masuelli, L.; Benvenuto, M.; di Stefano, E.; Mattera, R.; Fantini, M.; de Feudis, G.; de Smaele, E.; Tresoldi, I.; Giganti, M.G.; Modesti, A.; et al. Curcumin blocks autophagy and activates apoptosis of malignant mesothelioma cell lines and increases the survival of mice intraperitoneally transplanted with a malignant mesothelioma cell line. Oncotarget 2017, 8, 34405–34422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Di Meo, F.; Filosa, S.; Madonna, M.; Giello, G.; Di Pardo, A.; Maglione, V.; Baldi, A.; Crispi, S. Curcumin C3 complex/Bioperine has antineoplastic activity in mesothelioma: An in vitro and in vivo analysis. J. Exp. Clin. Cancer Res. 2019, 38, 360–371. [Google Scholar] [CrossRef] [PubMed]
  46. Pouliquen, D.L.; Boissard, A.; Henry, C.; Blandin, S.; Richomme, P.; Coqueret, O.; Guette, C. Curcumin Treatment Identifies Therapeutic Targets within Biomarkers of Liver Colonization by Highly Invasive Mesothelioma Cells-Potential Links with Sarcomas. Cancers 2020, 12, 3384. [Google Scholar] [CrossRef]
  47. Pouliquen, D.L.; Nawrocki-Raby, B.; Nader, J.; Blandin, S.; Robard, M.; Birembaut, P.; Grégoire, M. Evaluation of intracavitary administration of curcumin for the treatment of sarcomatoid mesothelioma. Oncotarget 2017, 8, 57552–57573. [Google Scholar] [CrossRef] [Green Version]
  48. Hocking, A.; Tommasi, S.; Sordillo, P.; Klebe, S. The safety and exploration of the pharmacokinetics of intrapleural liposomal curcumin. Int. J. Nanomed. 2020, 15, 943–952. [Google Scholar] [CrossRef]
  49. Valenti, D.; de Bari, L.; Manente, G.A.; Rossi, L.; Mutti, L.; Moro, L.; Vacca, R.A. Negative modulation of mitochondrial oxidative phosphorylation by epigallocatechin-3 gallate leads to growth arrest and apoptosis in human malignant pleural mesothelioma cells. Biochim. Biophys. Acta 2013, 1832, 2085–2096. [Google Scholar] [CrossRef]
  50. Ranzato, E.; Martinotti, S.; Magnelli, V.; Murer, B.; Biffo, S.; Mutti, L.; Burlando, B. Epigallocatechin-3-gallate induces mesothelioma cell death via H2O2 -dependent T-type Ca2+ channel opening. J. Cell Mol. Med. 2012, 16, 2667–2678. [Google Scholar] [CrossRef]
  51. Satoh, M.; Takemura, Y.; Hamada, H.; Sekido, Y.; Kubota, S. EGCG induces human mesothelioma cell death by inducing reactive oxygen species and autophagy. Cancer Cell Int. 2013, 13, 19–27. [Google Scholar] [CrossRef] [Green Version]
  52. Martinotti, S.; Ranzato, E.; Burlando, B. In Vitro screening of synergistic ascorbate-drug combinations for the treatment of malignant mesothelioma. Toxicol. In Vitro 2011, 25, 1568–1574. [Google Scholar] [CrossRef]
  53. Martinotti, S.; Ranzato, E.; Parodi, M.; Vitale, M.; Burlando, B. Combination of ascorbate/epigallocatechin-3-gallate/gemcitabine synergistically induces cell cycle deregulation and apoptosis in mesothelioma cells. Toxicol. Appl. Pharmacol. 2014, 274, 35–41. [Google Scholar] [CrossRef] [PubMed]
  54. Martinotti, S.; Ranzato, E.; Burlando, B. (−)- Epigallocatechin-3-gallate induces GRP78 accumulation in the ER and shifts mesothelioma constitutive UPR into pro-apoptotic ER stress. J. Cell. Physiol. 2018, 233, 7082–7090. [Google Scholar] [CrossRef]
  55. Lee, Y.J.; Im, J.H.; Lee, D.M.; Park, J.S.; Won, S.Y.; Cho, M.K.; Nam, H.S.; Lee, Y.J.; Lee, S.H. Synergistic inhibition of mesothelioma cell growth by the combination of clofarabine and resveratrol involves Nrf2 downregulation. BMB Rep. 2012, 45, 647–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Lee, Y.J.; Lee, Y.J.; Im, J.H.; Won, S.Y.; Kim, Y.B.; Cho, M.K.; Nam, H.S.; Choi, Y.J.; Lee, S.H. Synergistic anticancer effects of resveratrol and chemotherapeutic agent clofarabine against human malignant mesothelioma MSTO-211H cells. Food Chem. Toxicol. 2013, 52, 61–68. [Google Scholar] [CrossRef] [PubMed]
  57. Lee, Y.J.; Lee, G.J.; Yi, S.S.; Heo, S.H.; Park, C.R.; Nam, H.S.; Cho, M.K.; Lee, S.H. Cisplatin and resveratrol induce apoptosis and autophagy following oxidative stress in malignant mesothelioma cells. Food Chem. Toxicol. 2016, 97, 96–107. [Google Scholar] [CrossRef]
  58. Motohashi, H.; Yamamoto, M. Nrf2-Keap1 defines a physiologically important stress response mechanism. Trends Mol. Med. 2004, 10, 549–557. [Google Scholar] [CrossRef]
  59. Beishline, K.; Azizkhan-Clifford, J. Sp1 and the ‘hallmarks of cancer’. FEBS J. 2015, 282, 224–258. [Google Scholar] [CrossRef] [PubMed]
  60. Lee, K.A.; Lee, Y.J.; Ban, J.O.; Lee, Y.J.; Lee, S.H.; Cho, M.K.; Nam, H.S.; Hong, J.T.; Shim, J.H. The flavonoid resveratrol suppresses growth of human malignant pleural mesothelioma cells through direct inhibition of specificity protein 1. Int. J. Mol. Med. 2012, 30, 21–27. [Google Scholar]
  61. Lee, Y.J.; Lee, Y.J.; Lee, S.H. Resveratrol and clofarabine induces a preferential apoptosis-activating effect on malignant mesothelioma cells by Mcl-1 down-regulation and caspase-3 activation. BMB Rep. 2015, 48, 166–171. [Google Scholar] [CrossRef]
  62. Chae, J.I.; Cho, J.H.; Lee, K.A.; Choi, N.J.; Seo, K.S.; Kim, S.B.; Lee, S.H.; Shim, J.H. Role of transcription factor Sp1 in the quercetin-mediated inhibitory effect on human malignant pleural mesothelioma. Int. J. Mol. Med. 2012, 30, 835–841. [Google Scholar] [CrossRef] [Green Version]
  63. Demiroglu-Zergeroglu, A.; Basara-Cigerim, B.; Kilic, E.; Yanikkaya-Demirel, G. The investigation of effects of quercetin and its combination with cisplatinCisplatin on malignant mesothelioma cells in vitro. J. Biomed. Biotechnol. 2010, 2010, 851589. [Google Scholar] [CrossRef]
  64. Demiroglu-Zergeroglu, A.; Ergene, E.; Ayvali, N.; Kuete, V.; Sivas, H. Quercetin and Cisplatin combined treatment altered cell cycle and mitogen-activated protein kinase expressions in malignant mesothelioma cells. BMC Complement Altern. Med. 2016, 16, 281. [Google Scholar] [CrossRef] [Green Version]
  65. Demiroglu-Zergeroglu, A.; Candemir, G.; Turhanlar, E.; Sagir, F.; Ayvali, N. EGFR-dependent signalling reduced and p38 dependent apoptosis required by Gallic acid in Malignant Mesothelioma cells. Biomed. Pharmacother. 2016, 84, 2000–2007. [Google Scholar] [CrossRef] [PubMed]
  66. Lee, K.A.; Chae, J.I.; Shim, J.H. Natural diterpenes from coffee, cafestol and kahweol induce apoptosis through regulation of specificity protein 1 expression in human malignant pleural mesothelioma. J. Biomed. Sci. 2012, 19, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Huber, W.W.; Rossmanith, W.; Grusch, M.; Haslinger, E.; Prustomersky, S.; Peter-Vörösmarty, B.; Parzefall, W.; Scharf, G.; Schulte-Hermann, R. Effects of coffee and its chemopreventive components kahweol and cafestol on cytochrome P450 and sulfotransferase in rat liver. Food Chem. Toxicol. 2008, 46, 1230–1238. [Google Scholar] [CrossRef] [PubMed]
  68. Lee, K.A.; Lee, S.H.; Lee, Y.J.; Baeg, S.M.; Shim, J.H. Hesperidin induces apoptosis by inhibiting Sp1 and its regulatory protein in MSTO-211H cells. Biomol. Ther. 2012, 20, 273–279. [Google Scholar] [CrossRef] [Green Version]
  69. Chae, J.I.; Jeon, Y.J.; Shim, J.H. Downregulation of Sp1 is involved in honokiol-induced cell cycle arrest and apoptosis in human malignant pleural mesothelioma cells. Oncol. Rep. 2013, 29, 2318–2324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Kolbe, L.; Immeyer, J.; Batzer, J.; Wensorra, U.; tom Dieck, K.; Mundt, C.; Wolber, R.; Stäb, F.; Schönrock, U.; Ceilley, R.I.; et al. Anti-inflammatory efficacy of Licochalcone A: Correlation of clinical potency and in vitro effects. Arch. Dermatol. Res. 2006, 298, 23–30. [Google Scholar] [CrossRef]
  71. Kim, K.H.; Yoon, G.; Cho, J.J.; Cho, J.H.; Cho, Y.S.; Chae, J.I.; Shim, J.H. Licochalcone A induces apoptosis in malignant pleural mesothelioma through downregulation of Sp1 and subsequent activation of mitochondria-related apoptotic pathway. Int. J. Oncol. 2015, 46, 1385–1392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chirumamilla, C.; Pérez-Novo, C.; Van Ostade, X.; Vanden Berghe, W. Molecular insights into cancer therapeutic effects of the dietary medicinal phytochemical withaferin A. Proc. Nutr. Soc. 2017, 76, 96–105. [Google Scholar] [CrossRef] [PubMed]
  73. Yang, H.; Wang, Y.; Cheryan, V.T.; Wu, W.; Cui, C.Q.; Polin, L.A.; Pass, H.I.; Dou, Q.P.; Rishi, A.K.; Wali, A. Withaferin A inhibits the proteasome activity in mesothelioma in vitro and in vivo. PLoS ONE 2012, 7, e41214. [Google Scholar] [CrossRef] [PubMed]
  74. Sayeed, M.A.; Bracci, M.; Lucarini, G.; Lazzarini, R.; Di Primio, R.; Santarelli, L. Regulation of microRNA using promising dietary phytochemicals: Possible preventive and treatment option of malignant mesothelioma. Biomed. Pharmacother. 2017, 94, 1197–1224. [Google Scholar] [CrossRef] [PubMed]
  75. Sohn, E.J.; Won, G.; Lee, J.; Yoon, S.S.; Lee, I.; Kim, H.J.; Kim, S.H. Blockage of epithelial to mesenchymal transition and upregulation of let 7b are critically involved in ursolic acid induced apoptosis in malignant mesothelioma cell. Int. J. Biol. Sci. 2016, 12, 1279–1288. [Google Scholar] [CrossRef] [Green Version]
  76. Roomi, M.W.; Ivanov, V.; Kalinovsky, T.; Niedzwiecki, A.; Rath, M. Inhibition of malignant mesothelioma cell matrix metalloproteinase production and invasion by a novel nutrient mixture. Exp. Lung Res. 2006, 32, 69–79. [Google Scholar] [CrossRef]
  77. Roomi, M.W.; Ivanov, V.; Netke, S.P.; Niedzwiecki, A.; Rath, M. Serum markers of the liver, heart, and kidney and lipid profile and histopathology in ODS rats treated with nutrient synergy. J. Am. Coll. Nutr. 2003, 22, 477. [Google Scholar]
  78. Pulito, C.; Mori, F.; Sacconi, A.; Casadei, L.; Ferraiuolo, M.; Valerio, M.C.; Santoro, R.; Geoman, F.; Maidecchi, A.; Mattoli, L.; et al. Cynara scolymus affects malignant pleural mesothelioma by promoting apoptosis and restraining invasion. Oncotarget 2015, 6, 18134–18150. [Google Scholar] [CrossRef] [Green Version]
  79. Chmielewska-Kassassir, M.; Sobierajska, K.; Ciszewski, W.M.; Bukowiecka-Matusiak, M.; Szczesna, D.; Burzynska-Pedziwiatr, I.; Wiczkowski, W.; Wagner, W.; Wozniak, L.A. Polyphenol Extract from Evening Primrose (Oenothera paradoxa) Inhibits Invasion Properties of Human Malignant Pleural Mesothelioma Cells. Biomolecules 2020, 10, 1574. [Google Scholar] [CrossRef]
  80. Kiss, A.K.; Derwińska, M.; Dawidowska, A.; Naruszewicz, M. Novel biological properties of Oenothera paradoxa defatted seed extracts: Effect on metallopeptidase activity. J. Agric. Food Chem. 2008, 56, 7845–7852. [Google Scholar] [CrossRef]
  81. Birch, A.E.; Fenner, G.P.; Watkins, R.; Boyd, L.C. Antioxidant properties of evening primrose seed extracts. J. Agric. Food Chem. 2001, 49, 4502–4507. [Google Scholar] [CrossRef]
  82. Wettasinghe, M.; Shahidi, F. Evening primrose meal: A source of natural antioxidants and scavenger of hydrogen peroxide and oxygen-derived free radicals. J. Agric. Food Chem. 1999, 47, 1801–1812. [Google Scholar] [CrossRef] [PubMed]
  83. Wettasinghe, M.; Shahidi, F. Iron (II) chelation activity of extracts of borage and evening primrose meals. Food Res. Int. 2002, 35, 65–71. [Google Scholar] [CrossRef]
  84. Kiss, A.; Filipek, A.; Czerwińska, M.; Naruszewicz, M. Oenothera paradoxa defatted seeds extract, and its bioactive component penta-O-gallolyl-β-D-glucose decreased production of reactive oxygen species and inhibited release of leukotriene B4, interleukin-8, elastase and myeloperoxidase, in human neutrophils. J. Agric. Food Chem. 2010, 58, 9960–9966. [Google Scholar]
  85. Burzynska-Pedziwiatr, I.; Bukowiecka-Matusiak, M.; Wojcik, M.; Machala, W.; Bienkiewicz, M.; Spolnik, G.; Danikiewicz, W.; Wozniak, L.A. Dual stimulus-dependent effect of Oenothera paradoxa extract on the respiratory burst in human leukocytes: Suppressing for Escherichia coli and phorbol myristate acetate and stimulating for formyl-methionyl-leucyl-phenylalanine. Oxid. Med. Cell Longev. 2014, 2014, 764367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Pellegrina, C.D.; Padovani, G.; Mainente, F.; Zoccatelli, G.; Bissoli, G.; Mosconi, S.; Veneri, G.; Peruffo, A.; Andrighetto, G.; Rizzi, C.; et al. Anti-tumor potential of a gallic acid-containing phenolic fraction from Oenothera biennis. Cancer Lett. 2005, 226, 17–25. [Google Scholar] [CrossRef] [PubMed]
  87. Kiss, A.K.; Kaplon-Cieslicka, A.; Filipiak, K.J.; Opolski, G.; Naruszewicz, M. Ex Vivo effects of an Oenothera paradoxa extract on the reactive oxygen species generation and neutral endopeptidase activity in neutrophils from patients after acute myocardial infarction. Phytother. Res. 2011, 26, 482–487. [Google Scholar] [CrossRef] [PubMed]
  88. Granica, S.; Czerwińska, M.E.; Piwowarski, J.P.; Ziaja, M.; Kiss, A.K. Chemical composition, antioxidative and anti-inflammatory activity of extracts prepared from aerial parts of Oenothera biennis L. and Oenothera paradoxa hudziok obtained after seeds cultivation. J. Agric. Food Chem. 2013, 61, 801–810. [Google Scholar] [CrossRef] [PubMed]
  89. Sałaga, M.; Lewandowska, U.; Sosnowska, D.; Zakrzewski, P.K.; Cygankiewicz, A.I.; Piechota-Polańczyk, A.; Sobczak, M.; Mosinska, P.; Chen, C.; Krajewska, W.M.; et al. Polyphenol extract from evening primrose pomace alleviates experimental colitis after intracolonic and oral administration in mice. Naunyn Schmiedeberg’s Arch. Pharmacol. 2014, 387, 1069–1078. [Google Scholar] [CrossRef] [Green Version]
  90. Jaszewska, E.; Kośmider, A.; Kiss, A.K.; Naruszewicz, M. Pro-oxidative and pro-apoptotic action of defatted seeds of Oenothera paradoxa on human skin melanoma cells. J. Agric. Food Chem. 2009, 57, 8282–8289. [Google Scholar] [CrossRef]
  91. Gorlach, S.; Wagner, W.; Podsedek, A.; Sosnowska, D.; Dastych, J.; Koziołkiewicz, M. Polyphenols from evening primrose (Oenothera paradoxa) defatted seeds induce apoptosis in human colon cancer Caco-2 cells. J. Agric. Food Chem. 2011, 59, 6985–6997. [Google Scholar] [CrossRef] [PubMed]
  92. Arimura, T.; Kojima-Yuasa, A.; Tatsumi, Y.; Kennedy, D.O.; Matsui-Yuasa, I. Involvement of polyamines in evening primrose extract-induced apoptosis in Ehrlich ascites tumor cells. Amino Acids 2005, 28, 21–27. [Google Scholar] [CrossRef] [PubMed]
  93. Lewandowska, U.; Owczarek, K.; Szewczyk, K.; Podsędek, A.; Koziołkiewicz, M.; Hrabec, M. Influence of polyphenol extract from evening primrose (Oenothera paradoxa) seeds on human prostate and breast cancer cell lines. Postepy Hig. Med. Dosw. 2014, 68, 110–118. [Google Scholar] [CrossRef] [PubMed]
  94. Jaszewska, E.; Kosmider, A.; Kiss, A.K.; Naruszewicz, M. Oenothera paradoxa defatted seeds extract containing pentagalloylglucose and procyanidins potentiates the cytotoxicity of vincristine. J. Physiol. Pharm. 2010, 61, 637–643. [Google Scholar]
Figure 1. Pathogenesis of human mesothelial cells upon asbestos fibers contact. (TNF-α, tumor necrosis factor-alpha (grey dots); TNFR, tumor necrosis factor-alpha receptor (Y symbol); NF-κB, nuclear factor kappa B; ROS, reactive oxygen species; RNS, reactive nitrogen species; EGFR, epidermal growth factor receptor).
Figure 1. Pathogenesis of human mesothelial cells upon asbestos fibers contact. (TNF-α, tumor necrosis factor-alpha (grey dots); TNFR, tumor necrosis factor-alpha receptor (Y symbol); NF-κB, nuclear factor kappa B; ROS, reactive oxygen species; RNS, reactive nitrogen species; EGFR, epidermal growth factor receptor).
Ijms 22 08279 g001
Figure 2. Molecular pathways involved in the pathogenesis of MPM—potential targets for therapy. (EGFR, epidermal growth factor receptor; IGFR, insulin-like growth factor receptor; VEGFR, vascular endothelial growth factor receptor; MET, hepatocyte growth factor receptor; PDL-1, programmed cell death protein ligand-1; CTLA-4, cytotoxic T lymphocyte-associated protein; PD-1, programmed cell death protein-1; Hsp90, heat shock protein-90; P, phosphorylated form of signaling molecules).
Figure 2. Molecular pathways involved in the pathogenesis of MPM—potential targets for therapy. (EGFR, epidermal growth factor receptor; IGFR, insulin-like growth factor receptor; VEGFR, vascular endothelial growth factor receptor; MET, hepatocyte growth factor receptor; PDL-1, programmed cell death protein ligand-1; CTLA-4, cytotoxic T lymphocyte-associated protein; PD-1, programmed cell death protein-1; Hsp90, heat shock protein-90; P, phosphorylated form of signaling molecules).
Ijms 22 08279 g002
Figure 3. Mapping targets in the MPM therapy (based on Hanahan D, Weinberg RA. 2011 [25]).
Figure 3. Mapping targets in the MPM therapy (based on Hanahan D, Weinberg RA. 2011 [25]).
Ijms 22 08279 g003
Figure 4. Penta-O-galloyl-β-D-glucose polyphenol (PGG).
Figure 4. Penta-O-galloyl-β-D-glucose polyphenol (PGG).
Ijms 22 08279 g004
Figure 5. Molecular targets of natural compounds in malignant pleural mesothelioma treatment. (Cyt c, cytochrome c (black dots); P, phosphorylated form of signaling molecules).
Figure 5. Molecular targets of natural compounds in malignant pleural mesothelioma treatment. (Cyt c, cytochrome c (black dots); P, phosphorylated form of signaling molecules).
Ijms 22 08279 g005
Table 1. Major classes of polyphenols and their representatives.
Table 1. Major classes of polyphenols and their representatives.
FlavonoidsFlavonolsFlavonesFlavanols
quercetin
Ijms 22 08279 i001
luteolin
Ijms 22 08279 i002
catechin
Ijms 22 08279 i003
epigallocatechin-3-gallate (EGCG)
AnthocyanidinsIsoflavonesFlavanones
cyanidin
Ijms 22 08279 i004
genistein
Ijms 22 08279 i005
hesperidin
Ijms 22 08279 i006
Phenolic acidsgallic acid
Ijms 22 08279 i007
caffeic acid
Ijms 22 08279 i008
protocatechuic acid
Ijms 22 08279 i009
Stilbenesresveratrol
Ijms 22 08279 i010
Lignanspinoresinol
Ijms 22 08279 i011
Other polyphenolscurcumin
Ijms 22 08279 i012
Table 2. Studies on polyphenolic compounds in the treatment of malignant pleural mesothelioma.
Table 2. Studies on polyphenolic compounds in the treatment of malignant pleural mesothelioma.
CompoundMolecular Target EffectEffectReference
Curcumin↑ Bax proteins and caspase-9 expression;
Induction of XAF1, CARP1/CCAR1, SULF1
Stimulation of pro-apoptotic factors;
induction of novel apoptosis signaling transducers;
[41]

[44]
reduction of tumorigenic properties of MPM cells in an animal model[43]
Epigallocatechin-3-gallate
(+ascorbate and gemcitabine)
↑ ROS generation Impairment of mitochondrial membrane potential;[51]
↓ NF-κB activation (↓ p65 subunit)restoring programmed cell death;[53]
pro-apoptotic ER stress (UPR shift and GRP78 accumulation in ER)induction of pro-apoptotic signals[54]
Resveratrol (+cisplatin or +clofarabine)↑ROS generation in mitochondria
↓ Sp1 protein level
↓ expression of Sp1 regulatory proteins: p21, p27, cyclin D1, Mcl1, and survivin
↑ caspase-3 activation↓ anti-apoptotic Bcl-xL
↑ pro-apoptotic Bax
Activation of the apoptotic pathway;
MSTO-H211 cells sensitization to the antitumor effect of clofarabine
and cisplatin
[55,56,57]
[60,61]

Quercetin
(+ cisplatin)
↓ Sp1 protein and mRNA level
↑ caspase-3 signaling
↓ anti-apoptotic Bcl-xL
↑ pro-apoptotic Bax
S-phase arrest;
activation of the apoptotic pathway;
sensitization to the antitumor effect of cisplatin
[62,63,64]

Gallic acid
↓ expression of cyclin D and Bcl-2 genes;
altered expression of p21
G1 phase arrest;
mitochondria dysfunction;
apoptosis
[65]

Cafestol

Ijms 22 08279 i013
Kahweol
Ijms 22 08279 i014
↓ Sp1 protein level
↓ expression of Sp1 regulatory proteins: cyclin D1, Mcl1, and survivin
↑ caspase-3 signaling
↓ anti-apoptotic Bcl-xL
↑ pro-apoptotic Bax, Bid
Growth arrest of MSTO-H211 cellsat Sub-G1 phase;
activation of apoptotic signals
[66]

Hesperidin
Ijms 22 08279 i015
↓ Sp1 mRNA and protein level
↓ expression of Sp1 regulatory proteins: cyclin D1, Mcl1, and survivin
↑ caspase-3 signaling
↓ anti-apoptotic
Bcl-xL
↑ pro-apoptotic Bax, Bid
Growth arrest of MSTO-H211 cellsat Sub-G1 phase;
activation of apoptotic signals
[68]

Honokiol

Ijms 22 08279 i016
↓ Sp1 mRNA and protein level
↓ expression of Sp1 regulatory proteins: cyclin D1, Mcl1, and survivin
↑ caspase-3 signaling
↓ anti-apoptotic Bcl-xL
↑ pro-apoptotic Bax, Bid
Proliferation inhibition;
growth arrest of MSTO-H211 cellsat Sub-G1 phase;
activation of apoptotic signals
[69]

Licochalcone A
Ijms 22 08279 i017
↓ Sp1 mRNA and protein level
↓ expression of Sp1 regulatory proteins: cyclin D1, Mcl1, and survivin
↑ caspase-3 signaling
↓ anti-apoptotic Bcl-xL
↑ pro-apoptotic Bax, Bid
Growth arrest of MSTO-H211 and NCI-H28 cells
at Sub-G1 phase;
depolarization of mitochondria membrane;
activation of apoptotic signals
[71]
Withaferin A

Ijms 22 08279 i018
↑ p38 stress-activated caspase-3, ↑ Bax protein, PARP cleavage, induction of CARP1/CCAR1 Increase of pro-apoptotic proteins;
blockage in the chymotryptic
activity of proteasome
[73]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chmielewska-Kassassir, M.; Wozniak, L.A. Phytochemicals in Malignant Pleural Mesothelioma Treatment—Review on the Current Trends of Therapies. Int. J. Mol. Sci. 2021, 22, 8279. https://doi.org/10.3390/ijms22158279

AMA Style

Chmielewska-Kassassir M, Wozniak LA. Phytochemicals in Malignant Pleural Mesothelioma Treatment—Review on the Current Trends of Therapies. International Journal of Molecular Sciences. 2021; 22(15):8279. https://doi.org/10.3390/ijms22158279

Chicago/Turabian Style

Chmielewska-Kassassir, Malgorzata, and Lucyna A. Wozniak. 2021. "Phytochemicals in Malignant Pleural Mesothelioma Treatment—Review on the Current Trends of Therapies" International Journal of Molecular Sciences 22, no. 15: 8279. https://doi.org/10.3390/ijms22158279

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

Article Metrics

Back to TopTop