Next Article in Journal
Combined PARP and Dual Topoisomerase Inhibition Potentiates Genome Instability and Cell Death in Ovarian Cancer
Next Article in Special Issue
The Role of NF-κB in Endometrial Diseases in Humans and Animals: A Review
Previous Article in Journal
New Insights on Heme Uptake in Leishmania spp.
Previous Article in Special Issue
Potential Application of Leelamine as a Novel Regulator of Chemokine-Induced Epithelial-to-Mesenchymal Transition in Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Uses of Mushrooms in Cancer Treatment and Their Anticancer Mechanisms

Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Korea
Int. J. Mol. Sci. 2022, 23(18), 10502; https://doi.org/10.3390/ijms231810502
Submission received: 1 August 2022 / Revised: 1 September 2022 / Accepted: 7 September 2022 / Published: 10 September 2022
(This article belongs to the Special Issue Role of NF-κB in Carcinogenesis and Its Therapeutic Regulation)

Abstract

:
Cancer is the leading cause of mortality worldwide. Various chemotherapeutic drugs have been extensively used for cancer treatment. However, current anticancer drugs cause severe side effects and induce resistance. Therefore, the development of novel and effective anticancer agents with minimal or no side effects is important. Notably, natural compounds have been highlighted as anticancer drugs. Among them, many researchers have focused on mushrooms that have biological activities, including antitumor activity. The aim of this review is to discuss the anticancer potential of different mushrooms and the underlying molecular mechanisms. We provide information regarding the current clinical status and possible modes of molecular actions of various mushrooms and mushroom-derived compounds. This review will help researchers and clinicians in designing evidence-based preclinical and clinical studies to test the anticancer potential of mushrooms and their active compounds in different types of cancers.

1. Introduction

Cancer is one of the leading causes of deaths worldwide, and accounted for nearly 10 million deaths in 2020 according to WHO [1]. Accurate cancer diagnosis is important for effective treatment because each type of cancer requires a specific regimen. Cancer treatments are diverse, including surgery, anticancer drug treatment, radiotherapy, and systemic therapy (chemotherapy and targeted biological therapies). However, the current anticancer drugs available in the market are not target-specific, leading to the development of drug resistance, and even causing several side effects in clinical chemotherapy [2]. Therefore, it is important to develop novel and effective anticancer agents with low toxicity. In this regard, natural compounds have been highlighted as anticancer drugs. Mushrooms have been used in traditional medicines in East Asia due to their immunomodulatory, anticancer, and anti-inflammatory activities [2]. Among 14,000 different species of mushrooms, approximately 700 species have been reported to exhibit medicinal properties [3]. Recently, many studies have revealed the biological activities and the mechanisms of actions of mushroom compounds [2,3,4,5]. Some mushrooms and their active compounds possess anticancer properties. Polysaccharides isolated from Phellinus linteus (PLP) suppressed tumor growth and pulmonary metastasis through stimulating the immune response, not directly toxic to cancer cells [4]. Triterpenoids from Ganoderma lucidum showed anticancer properties [5]. β-D-glucans from Ganoderma lucidum exhibited anticancer effect by inhibiting cancer cells, protecting normal cells against free radicals, and reducing normal cell damage [2]. Their potential use as adjuncts in cancer therapy or as anticancer agents has emerged. Numerous clinical trials are in progress to assess the benefits of medicinal mushroom extracts in chemotherapy [2].
In this review, the clinical use and anticancer mechanisms of mushrooms are described. Our goal is to provide information pertaining to the potential therapeutic use of mushroom extracts and their active compounds against various cancers by elucidating the underlying targeted signaling pathways. Furthermore, these mushroom-derived compounds with anticancer activities can be exploited as novel anticancer agents.

2. Uses of Mushrooms in Cancer Therapy

Many groups have reported that mushrooms possess anticancer activities and minimize undesirable side effects such as nausea, bone marrow suppression, anemia, and insomnia, and lower drug resistance after chemotherapy and radiation therapy [6] (Table 1 and Table S1).
In the RCT conducted by Tsai et al., advanced adenocarcinoma patients treated with Antrodia cinnamomea alongside chemotherapy developed less severe gastrointestinal symptoms, such as abdominal pain and diarrhea, than those in the placebo group [7]. Twardowski et al. reported that Agaricus blazei Murill decreased prostate-specific antigen (PSA) levels and regulated recurrent prostate cancer by decreasing immunosuppressive factor [8]. Ahn et al. reported that patients with gynecological cancers receiving chemotherapy showed fewer side effects, such as loss of appetite, alopecia, and general weakness, when the therapy was accompanied by Agaricus blazei Murill compared to those in the placebo group [9]. Hetland et al. demonstrated that there was an increased number of plasmacytoid dendritic cells (pDC) and T regulatory cells (Tregs) in the blood; increased serum levels of IL-1Ra (receptor antagonist), IL-5, and IL-7; and an increased level of immunoglobulin genes, killer immunoglobulin receptor (KIR) genes, and human leukocyte (HLA) genes in the bone marrow in Agaricus blazei mushroom extract (AndoSanTM)-treated myeloma patients [10]. Loss of appetite decreased over time in patients that underwent six cycles of chemotherapy accompanied with Agaricus sylvaticus, while most patients in the placebo group suffered from loss of appetite and gastrointestinal symptoms, such as diarrhea, constipation, nausea, and vomiting [11]. In advanced lung cancer, 3–84% of patients receiving Ganoderma lucidum exhibited significantly improved cancer-related symptoms (e.g., fever, cough, weakness, sweating, and insomnia) compared to the placebo group (11–43%) [12]. In the randomized controlled trial (RCT) conducted by Zhao, breast cancer patients treated with Ganoderma lucidum showed less cancer-related fatigue than patients undergoing endocrine therapy [13]. In a phase I/II trial of breast cancer survivors, Grifola frondosa extract acted as an immunomodulator by increasing the production of IL-2, IL-10, TNF-α, and IFN-γ by subsets of T cells [14]. Hackman et al., reported that Lentinula endodes treatment alone was ineffective in treating prostate cancer patients [15]. The anticancer activity of a semisynthetic derivative of illudin S from Omphalotus illudens is due to the alkylation of DNA, RNA, and proteins. However, its use in the clinic is limited due to its strong retinal toxicity and narrow therapeutic index [16]. In a phase I trial conducted by Torkelson, Trametes versicolor enhanced the immune status in immunocompromised breast cancer patients [17]. In an RCT performed by Chay et al. for advanced hepatocellular carcinoma (HCC), patients treated with Trametes versicolor had a longer median overall survival (OS) and median progression-free survival compared to the placebo group [18]. The immunostimulatory effect and direct toxicity to cancer cells exhibited by Trametes versicolor polysaccharides implies that they can be applied as more than an adjuvant therapy [19].

3. Anticancer Compounds from Medicinal Mushrooms

The bioactive compounds found in mushrooms include polysaccharides, proteins, fats, phenolics, alkaloids, ergosterol, selenium, folate, enzymes, and organic acids. The anticancer components in mushrooms are antroquinonol, cordycepin, hispolon, lectin, krestin, polysaccharide, sulfated polysaccharide, lentinan, and Maitake D Fraction [20]. Many mushrooms are currently under clinical trials, and only a few are available for clinical use [2]. Polysaccharides are the most potent mushroom compounds with antitumor and immunomodulatory properties. Among polysaccharides, β-glucan consists of a backbone of glucose residues linked by β-(1→3)-glycosidic bonds, frequently with attached side-chain glucose residues joined by β-(1→6) linkages [21]. β-Glucan stimulates the immune system as a non-self molecule by inducing the production of cytokines that activate phagocytes and leukocytes [20]. Lentinan and lectins from Lentinula edodes have shown cytotoxic effects on breast cancer cells [22]. Lentinan from Lentinula edodes (also called Pyogo in Korea), schizophyllan (also called SPG, sonifilan, sizofiran, and sizofilan) from Schizophyllum commune, and PSK (also called krestin) from Trametes versicolor have been approved as prescription anticancer drugs in Japan [23]. Mushroom polysaccharides stimulate natural killer cells, T cells, B cells, and macrophages, leading to an increased immune response [24]. Cordycepin, also known as 3-deoxyadenosine, is a major anticancer compound in Cordyceps species. It exerts an apoptotic effect via dysregulated polyadenylation, and causes the termination of DNA or RNA elongation by binding to the site where nucleic acids are to be bound [25]. Hispolon, an active polyphenol compound, has been reported to exert potent antineoplastic properties and enhance the cytotoxicity of chemotherapeutic agents [26]. These findings suggest that some mushrooms may act synergistically in combination with commercial anticancer drugs as effective tools for treating drug-resistant cancers [6].

4. Anticancer Mechanisms of Medicinal Mushrooms

In Asian countries, medicinal use of mushrooms has been prevalent for a long time; however, in recent decades, their use for treating a number of diseases, including cancers, has increased in other parts of the world. The tremendous therapeutic potential of edible and medicinal mushrooms is attributed to the bioactive substances present in mushrooms. To increase the therapeutic success rates against cancer, it is important to understand the molecular mechanisms underlying cancer development and progression and the molecular targets of mushroom-derived bioactive compounds. In this section, we discuss how mushrooms help overcome multidrug resistance (MDR) and target signaling pathways, such as PI3K/AKT, Wnt-CTNNB1, and MAPK, during cancer treatment (Table 2).

4.1. Overcoming Pgp-Mediated MDR Using Mushrooms

Drug resistance is a major obstacle in chemotherapy. Endogenous or acquired drug resistance represents the simultaneous development of resistance in tumor cells to drugs that are not mechanically or structurally related. This phenomenon is known as multidrug resistance (MDR). Resistance to anticancer drugs is one of the main factors of treatment failure, resulting in high morbidity. The overexpression of efflux pumps, which leads to multidrug resistance, is an important issue that needs to be resolved.
A major form of MDR is the overexpression of ATP-binding cassette (ABC) transporters and P-glycoprotein (Pgp), a 170 KDa transmembrane glycoprotein product encoded by the MDR1 gene. The mechanism of drug resistance in Pgp-expressing tumor cells involves an increase in the extracellular transport of various chemotherapeutic agents, which diminishes cellular accumulation, and thus, decreases drug efficacy [31,44,45]. Anticancer agents with Pgp-mediated MDR include paclitaxel (TAX), doxorubicin (DOX), actinomycin D, vinblastine, and etoposide, whereas Pgp does not affect the cytotoxicity of certain other anticancer drugs, such as 5-fluorouracil, cisplatin, and carboplatin [46]. To confirm the drug resistance reversal activity of Basidiomycete mushroom extracts collected in Korea, the cytotoxic activity of paclitaxel (TAX), a well-known Pgp-related anticancer drug, on Pgp-positive and -negative human cancer cells in the presence or absence of the tested mushroom extract was compared to that in the presence of verapamil (VER), a well-known MDR reversal agent. Cantharellus cibarius (M02) and Russula emetica (M12) increased the cytotoxic activity of TAX by blocking Pgp-mediated drug efflux in Pgp-positive HCT15 and MES-SA/dX5 cancer cells, but not in Pgp-negative A549 and MES-SA cancer cells. Cantharellus cibarius and Russula emetica also increased the cytotoxicity of doxorubicin, another Pgp-associated anticancer drug, against MES-SA/DX5 cells [31]. Ganoderma species induced apoptosis in drug-sensitive (H69) and multidrug-resistant (VPA) human small-cell lung cancer (SCLC) cells that were resistant to etoposide and doxorubicin [47]. Ganoderma lucidum polysaccharide (PLP) inhibits the constitutive activation of NF-κB, decreasing the expression of PGP in cancer cells [48]. Zhankuic acids A–C isolated from Taiwanofungus camphoratus were found to exert inhibitory effects against Pgp, which reversed drug resistance against doxorubicin, vincritine, and paclitaxel in human cancer cells [43] (Figure 1).

4.2. Overcoming Tumor Resistance to Inhibit Immune Checkpoint Interactions, the PD-1 Pathway, and CTLA-4/CD80, Using Mushrooms

In recent years, immune checkpoint blockade (ICB) therapy has caused a paradigm shift in cancer immunotherapy; it primarily inhibits various checkpoints that control host T cell activity by regulating the immune checkpoint interactions, PD-1/PD-L1 and CTLA-4/CD80. Programmed cell death-1 (PD-1) (CD279) is an inhibitory receptor, which is expressed in activated CD8+ T cells (as well as on B cells and natural killer cells) and leads to reduced innate and adaptive immune responses [49]. Particularly, PD-1 is highly expressed in tumor-specific T cells, which has prompted researchers to examine whether the inhibition of PD-1 suppresses cancer aggression by promoting an effective immune response [50,51]. The binding of PD-1 to the PD-L1 ligand allows cancer cells to evade the host immune response. In addition, PD-L1 induction protects cancer cells from T-cell-mediated destruction [52]. PD-1/PD-L1-checkpoint-blocking antibodies have been focused on as a powerful ICB therapy for cancer patients. However, patients with cancer quickly develop resistance to immunotherapy. β-Glucan from medicinal mushrooms, which acts as an immune adjuvant, has been found to stimulate innate and adaptive immune responses. It has been reported that administration of whole glucan particle (WGP) β-glucan along with PD-1/PD-L1-checkpoint-blocking antibodies leads to increased recruitment of immune-associated cells, improves the regulation of the balance between T cell activation and immune tolerance, and delays tumor progression [53]. This combination therapy was also found to improve progression-free survival in patients with advanced cancer who had previously discontinued anti-PD-1/PD-L1 therapy because of disease progression. These findings suggest that β-glucan can be used as an immune adjuvant to reverse anti-PD-1/PD-L1 resistance by regulating the immune system [54]. Ganoderma lucidum and its bioactive compounds reduced PD-1 protein level in cultured GM00130 and GM02248 human B-lymphocytes, thus, preventing and treating cancer [33] (Figure 2).
The binding between CD80 on antigen-presenting cells (APCs) and CD28 on naive T cells results in T cell activation in the lymph nodes, which elevates the immune response and kills cancer cells. However, the interaction of CTLA-4 on naive T cells with CD80 on cancer cells produces an inhibitory signal for T cell activation, leading to the inhibition of T cell activation and suppression of the immune response [55]. Inonotus obliquus blocks CTLA-4/CD80 interaction and increases T cell activity so that cancer cells cannot escape the immune response [36] (Figure 2).

4.3. Targeting the PI3K/AKT Signaling Pathway in Cancer Using Mushrooms

The PI3K/AKT pathway is involved in the acquisition of chemotherapeutic drug resistance. The activation of phosphatidylinositol 3-kinase (PI3K) signaling leads to VEGF production, reduces tumor CD8+ T cell infiltration, and induces subsequent resistance to PD-1 blockade therapy. Currently, to avoid primary resistance to the PD-1/PD-L1 blockade, clinical treatment regimens that combine kinase inhibitor therapy with an immune checkpoint blockade are in use to enhance the response rates [52]. Activated phosphoinositide 3-kinase (PI3K) is a key signaling molecule that affects cell survival, proliferation, and differentiation by triggering the sequential activation of AKT and other downstream pathways [2,56]. When many components of this pathway are altered, it leads to the development of various cancers in humans [2].
Several research groups have demonstrated that mushroom-derived compounds can exert antitumor and antimetastatic effects by affecting various molecules in the PI3K/AKT pathway. For example, hispolon derived from Phellinus linteus inhibited the invasion and motility of a highly metastatic liver cancer cell line (SK-Hep1) by downregulating MMP2, MMP9, and uPa, and inhibiting the activation of the ERK1/2, PI3K/AKT, and FAK pathways [38]. Proteoglycan (P1) from Phellinus linteus showed antiproliferative activity in multiple human cancer cells by inducing a notable decrease in AKT, Reg IV, EGFR, and plasma PGE2 concentrations [57]. A polysaccharide–protein complex isolated from Pleurotus pilmonarius (PP) suppressed PI3K/AKT signaling in liver cancer cells [2]. Additionally, when PP was used in combination with cisplatin, the sensitization of liver cancer cells to cisplatin was improved. The phosphorylation of BAD at Ser 136 via AKT is required for cell viability. When this AKT node is suppressed in ovarian cancer cells, they become sensitive to cisplatin. Inhibition of PI3K/AKT signaling by PP made the cells more sensitive to cisplatin [2]. Ganoderic acid from Ganoderma lucidum suppressed human glioblastoma by inducing apoptosis and autophagy via the inactivation of the PI3K/AKT signaling pathway [58].

4.4. Targeting the Wnt/β-Catenin Pathway in Cancer Using Mushrooms

A high rate of abnormality in the Wnt signaling pathway has been observed in many cancers. APC, CTNNB1, AXIN1, FAM123B, and TCF7L2 are the key molecules in Wnt signaling pathway that may undergo somatic mutations related to common human cancers, including colon cancer [59]. The onset and progression of sporadic colon cancer (CRC) and familial adenomatous polyposis (FAP)-associated diseases are believed to be caused by mutations in the adenomatous polyposis coli (APC) gene [2]. Depending on the stage and type of cancer, the Wnt-CTNNB1 signaling pathway can either promote or inhibit tumor initiation, growth, metastasis, and drug resistance [2]. An inverse correlation has been reported between β-catenin/Wnt activation in cancer and the degree of CD8+ T cell infiltration in a mouse model of melanoma [60]. Increased β-catenin/Wnt activity also correlated with diminished CD103+ dendritic cell infiltration due to reduced levels of CCL4, a chemokine responsible for attracting them. PD-1 blockade therapy was ineffective in melanoma tumors with β-catenin/Wnt activation, whereas this treatment worked well in tumors without β-catenin/Wnt mutations [52,61]. Wnt inhibition increased tumor T cell infiltration and inhibited tumor proliferation and migration by enhancing PD-1 antibody treatment and upregulating the expression of PD-L1 in mice with glioblastoma (GBM) [62].
Several groups have reported the anti-oncogenic activities of different mushroom-derived compounds via Wnt–CTNNB1 signaling. For instance, 4-acetylantroquinonol and antroquinonol from Antrodia camphorata was discovered to inhibit colon cancer by suppressing the Wnt/β-catenin pathway [28,29]. Antrodia camphorata grown on germinated brown rice suppressed human colon cancer cell proliferation by upregulating G0/G1 phase arrest and apoptosis and reducing β-catenin signaling [40]. Researchers have shown that Phellinus linteus can inhibit tumor growth, invasion, and angiogenesis by downregulating genes (cyclin D1 and TCF/LEF) of the Wnt signaling pathway in SW480 human colon cancer cells as well as in vivo. Phellinus linteus grown on germinated brown rice attenuated the levels of NF-κB, β-catenin, and mitogen-activated protein kinase (MAPK) proteins [41]. Ergosterol peroxide and 4-acetylantroquinonol from Inonotus obliquus inhibited nuclear β-catenin in colon cancer cells [2]. When the level of β-catenin activity was reduced, the expression of β-catenin target genes (c-myc, cyclin D1, and VEGF) was also decreased, thus exerting anticancer effects on meningioma cells [63]. Therefore, these compounds may be potential candidates for pharmaceutical treatment of human meningiomas.

4.5. Targeting the MAPK Pathway in Cancer Using Mushrooms

Other mutations associated with T cell exclusion and subsequent resistance to PD-1/PD-L1 blockade usually occur within the mitogen-activated protein kinase (MAPK) signaling cascade. Constitutive oncogenic signaling activated through this pathway leads to the production of immunosuppressive cytokines, viz., vascular endothelial growth factor (VEGF) and interleukin 8 (IL-8), which inhibit T cell recruitment to the cancer tissue as well as their activity [52]. Mutations within the MAPK cascade are common in melanomas, and inhibition of this cascade is known to improve CD8+ T cell infiltration within cancers and sensitize them to PD-1 blockade therapy [52]. This result strongly suggests that a combination therapy involving multikinase inhibition with PD-1 blockade can be used in cancers with such mutations.
Platinum-based anticancer drugs have been shown to upregulate PD-L1 expression through the MAPK pathway in gastric cancer cells. A β-glucan from Lentinula edodes, viz., lentinan, suppressed cisplatin- or oxaliplatin-induced PD-L1 expression, suggesting that a combination of lentinan and platinum-based chemotherapy can recover the chemosensitivity of cells [64]. In addition, the MAPK pathway plays a pivotal role in oncogenesis, as several oncoproteins upstream of MAPK cascade, including ErbB-2, Scr RTKs, Ras, and Raf, are mutated into activated forms of enzymes [65]. The triterpene-enriched fraction, WEES-G6, from Ganoderma lucidum inhibited Huh-7 human hepatoma cell growth [66,67]. Yang et al. showed that Antrodia camphorata markedly inhibited the MAPK signaling pathway, thereby suppressing the invasion/migration of highly metastatic MDA-MB-231 cells [26,68]. Furthermore, G. lucidum triterpene extract (GLT) suppressed the phosphorylation of p38 MAPK, leading to antophagy in colon cancer cells [69]. Co-treatment using the extract of Phellinus linteus grown on germinated brown rice (PBR) and cetuximab reduced MAPK signaling by decreasing KRAS expression. PBR enhanced the sensitivity of KRAS-mutated colon cancer cells to cetuximab [40].

4.6. Targeting the NF-κB Pathway in Cancer Using Mushrooms

The nuclear transcription factor κB (NF-κB) is one of the factors responsible for cellular chemoresistance, and controls a myriad of gene expressions, including antigen receptors on immune cells, adhesion molecules, proinflammatory cytokines, and chemoattractants for inflammatory cells [70]. NF-κB is associated with neoplastic development, including insensitivity to growth inhibitory signals, avoidance of apoptosis, metastasis and sustained angiogenesis, and chronic inflammation [71].
Numerous studies highlight the antitumor effect of mushrooms through targeting the NF-κB signaling pathway. Antroquinonol (AQ) and 4-acetylantroquinonol B (4-AAQB) from Antrodia Camphorata exhibited inhibitory effects on NF-κB signaling in MCF-7 breast cancer cells [72]. Kadomatsu et al. found that treatment with cordycepin, a major compound of Cordyceps militaris, became sensitive to TNF-α-mediated apoptosis, which suppresses pro-survival NF-κB. Ho’s group reported that Ganoderma suppressed metastasis in highly invasive breast and prostate cancer cells by blocking constitutively active AP-1 and NF-κB signaling [73]. Treatment with sulfated polysaccharide obtained from Grifola frondosa (S-GFB) resulted in apoptosis of HepG2 cells through the induction of S phase arrest, inhibiting notch1 expression, degradation of IκB-α, translocation of NF-κB from the cytoplasm to the nucleus, and the activation of caspase 3 and 8 [35]. Phellinus linteus was shown to produce caffeic acid phenethyl ester (CAPE), which specifically inhibits NF-κB binding to DNA [74,75].

5. Regulating Immune Function in Cancer Using Medicinal Mushrooms

Despite the increasing success of existing personalized cancer treatments, recurrence and metastasis are common, depending on the type and stage of the disease [11]. Many studies have reported the beneficial effects of medicinal mushrooms, which particularly enhance the quality of life and reduce the side effects of conventional chemotherapy. In addition, their positive effects on anticancer activity and immune regulation have been reported. Several mechanisms have been suggested for the antiproliferative and immunomodulatory effects of medicinal mushrooms [2,20,76,77]. Mushroom polysaccharides stimulate dormant natural killer cells, T cells, B cells, and macrophage-dependent immune responses [24].
Mushroom-derived compounds activate immune cells to induce either cell-mediated or direct cytotoxicity in cancer cells by binding to pathogen recognition receptors. Compounds, such as lentinan, increase the proliferation of cytotoxic T lymphocytes and macrophages and induce nonspecific immune responses [78]. Pleurotus tuber and Pleurotus rhinoceros extracts were shown to promote the activation of lymphocytes and NK cells and increase macrophage proliferation, T helper cell number, and CD4/CD8 ratio and population, conferring anticancer effects [9,11,20,79]. Natural killer cells act as the key cells in innate immunity by attacking major histocompatibility class I-negative target cells that can evade immune surveillance of cytotoxic T cells. The activity of natural killer cells in patients with gynecological cancer undergoing chemotherapy was significantly enhanced when co-treated with Agaricus blazei Murill for 3 to 6 weeks as compared to that in the placebo group [9]. Leukopenia results in cachexia and metabolic changes in cancer patients and increases the risk of infection [80]. In patients with multiple myeloma treated with Agaricus blazei Murill, the immune status was much better in terms of maintaining the population of white blood cells and immunoglobins and led to fewer infections [81]. The mushroom’s main component, β-glucan, exerts hematopoietic effects and increases bone marrow regeneration in vitro [82]. β-Glucan also significantly increased the population of DCs (CD11c+/CD8+) and macrophages (CD11b+/F4-80+) and decreased the population of regulatory T cells and myeloid-derived suppressor cells (MDSC)s, resulting in an enhanced immune response [54]. Ganoderma lucidum supplementation resulted in a more stable disease state in the lung cancer study population than in the control group [83]. In addition, there was a significant increase in CD3 percentage, natural killer cell activity, and lymphocyte mitogenic reactivity against concanavalin A in lung cancer patients [11]. Cordyceps militaris fermented with Pediococcus pentosaceus (GRC-ON89A) treatment was reported to aid the recovery of immune activity in high-dose cyclophosphamide (a chemotherapeutic drug)-treated mice by increasing the phagocytic activity of mouse peritoneal macrophages and stimulating NO production in macrophages. GRC-ON89A reduced the toxicity of anticancer agents through the recovery of the immune system [84].
Thus, mushroom-derived compounds induce innate and adaptive immunity by enhancing immune surveillance against cancer by affecting monocytes, macrophages, NK cells, and B cells, and by activating immune organs [85,86], which leads to cancer cell apoptosis, cell cycle arrest, and prevention of angiogenesis and metastasis [20]. Consumption of mushroom compounds also boosts the secretion of antitumor cytokines by CTLs and activation of immune organs, thereby eliminating cancer cells and strengthening the weakened immune system [87].

6. Prebiotic Properties of Medicinal Mushrooms in Cancer

Several groups have reported that medicinal mushrooms can act as prebiotics, and thus enhance the growth of beneficial microbiota. Prebiotics can affect the human intestinal microbial population and suppress various diseases such as diabetes, obesity, and cancer [77]. The important sources of prebiotics in mushrooms are polysaccharides, such as chitin, hemicellulose, β- and α-glucans, mannans, xylans, and galactans, which can suppress the proliferation of pathogens by increasing the growth of probiotics in the gut [6,88]. A poor intestinal microbiota composition can lead to the development of cancer [88]. It is possible that prebiotic effects medicinal mushrooms could enhance quality of life (QOL) during and after cancer therapy.

7. Conclusions

This review demonstrates the potential use of mushrooms and their anticancer mechanisms in cancer treatment. Mushroom-derived bioactive compounds activate and/or regulate the immune system by affecting the maturation, differentiation, and proliferation of immune cells, thereby inhibiting cancer cell metastasis and growth. It is very important to understand the underlying mechanisms of action of the anticancer compounds derived from mushrooms to suppress cancer and improve the quality of life of cancer patients. Mushrooms show anticancer potential by regulating a single molecule of a specific signaling pathway, or by having multiple targets in the same or different signaling pathway(s), including the PI3K/Akt, Wnt/β-catenin, and MAPK pathways. In addition, several studies have highlighted the effect of mushroom-derived components as single and adjuvant therapeutic agents in reversing MDR by targeting Pgp, PD-1/PD-L1, and CTLA-4/CD80 interactions. In addition, the prebiotic effects of medicinal mushrooms could enhance quality of life (QOL) during and after cancer therapy by recovering the intestinal microbiota.
However, only a few clinical studies of a small number of mushrooms demonstrate the positive effects of medicinal mushrooms, including reductions in the adverse effects of conventional therapies, as well as antitumor activity and immunomodulation. Therefore, more clinical research on mushrooms with anticancer potential needs to be conducted, especially by employing high-quality methodology, larger sample sizes, standard mushroom preparations, and long-term follow-ups. In addition, future studies should investigate the preventive aspects of medicinal mushrooms in reducing the rate of cancer occurrence by being a part of a healthy diet and lifestyle. High-quality clinical studies are needed to identify the potential of medicinal mushrooms in cancer treatment.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms231810502/s1.

Funding

This work was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT, No. NRF-2021R1F1A1048156).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 29 August 2022).
  2. Joseph, T.P.; Chanda, W.; Padhiar, A.A.; Batool, S.; LiQun, S.; Zhong, M.; Huang, M. A preclinical evaluation of the antitumor activities of edible and medicinal mushrooms: A molecular insight. Integr. Cancer Ther. 2018, 17, 200–209. [Google Scholar] [CrossRef] [PubMed]
  3. Han, S.B.; Lee, C.W.; Jeon, Y.J.; Hong, N.D.; Yoo, I.D.; Yang, K.-H.; Kim, H.M. The inhibitory effect of polysaccharides isolated from Phellinus linteus on tumor growth and metastasis. Immunopharmacology 1999, 41, 157–164. [Google Scholar] [CrossRef]
  4. Wu, G.-S.; Guo, J.-J.; Bao, J.-L.; Li, X.-W.; Chen, X.-P.; Lu, J.-J.; Wang, Y.-T. Anti-cancer properties of triterpenoids isolated from Ganoderma lucidum—A review. Expert Opin. Investig. Drugs 2013, 22, 981–992. [Google Scholar] [CrossRef]
  5. Boh, B.; Berovic, M.; Zhang, J.; Zhi-Bin, L. Ganoderma lucidum and its pharmaceutically active compounds. Biotechnol. Annu. Rev. 2007, 13, 265–301. [Google Scholar] [PubMed]
  6. Jayachandran, M.; Xiao, J.; Xu, B. A Critical Review on Health Promoting Benefits of Edible Mushrooms through Gut Microbiota. Int. J. Mol. Sci. 2017, 18, 1934. [Google Scholar] [CrossRef] [PubMed]
  7. Tsai, M.-Y.; Hung, Y.-C.; Chen, Y.-H.; Chen, Y.-H.; Huang, Y.-C.; Kao, C.-W.; Su, Y.-L.; Chiu, H.-H.E.; Rau, K.-M. A preliminary randomised controlled study of short-term Antrodia cinnamomea treatment combined with chemotherapy for patients with advanced cancer. BMC Complement. Altern. Med. 2016, 16, 322. [Google Scholar] [CrossRef] [PubMed]
  8. Twardowski, P.; Kanaya, N.; Frankel, P.; Synold, T.; Ruel, C.; Pal, S.K.; Junqueira, M.; Prajapati, M.; Moore, T.; Tryon, P.; et al. A phase I trial of mushroom powder in patients with biochemically recurrent prostate cancer: Roles of cytokines and myeloid-derived suppressor cells for Agaricus bisporus–induced prostate-specific antigen responses. Cancer 2015, 121, 2942–2950. [Google Scholar] [CrossRef] [PubMed]
  9. Ahn, W.S.; Kim, D.J.; Chae, G.T.; Lee, J.M.; Bae, S.M.; Sin, J.I.; Kim, Y.W.; Namkoong, S.E.; Lee, I.P. Natural killer cell activity and quality of life were improved by consumption of a mushroom extract, Agaricus blazei Murill Kyowa, in gynecological cancer patients undergoing chemotherapy. Int. J. Gynecol. Cancer 2004, 14, 589–594. [Google Scholar] [CrossRef]
  10. Hetland, G.; Tangen, J.-M.; Mahmood, F.; Mirlashari, M.R.; Nissen-Meyer, L.S.H.; Nentwich, I.; Therkelsen, S.P.; Tjønnfjord, G.E.; Johnson, E. Antitumor, anti-inflammatory and antiallergic effects of Agaricus blazei mushroom extract and the related medicinal basidiomycetes mushrooms, Hericium erinaceus and Grifola frondosa: A review of preclinical and clinical studies. Nutrients 2020, 12, 1339. [Google Scholar] [CrossRef]
  11. Jeitler, M.; Michalsen, A.; Frings, D.; Hubner, M.; Fischer, M.; Koppold-Liebscher, D.A.; Murthy, V.; Kessler, C.S. Significance of Medicinal Mushrooms in Integrative Oncology: A Narrative Review. Front. Pharmacol. 2020, 11, 580656. [Google Scholar] [CrossRef]
  12. Gao, Y.; Dai, X.; Chen, G.; Ye, J.; Zhou, S. A randomized, placebo-controlled, multicenter study of Ganoderma lucidum (W.Curt.:Fr.) Lloyd (Aphyllophoromycetideae) polysaccharides (Ganopoly®) in patients with advanced lung cancer. Int. J. Med. Mushrooms 2003, 5, 14. [Google Scholar] [CrossRef]
  13. Zhao, H.; Zhang, Q.; Zhao, L.; Huang, X.; Wang, J.; Kang, X. Spore Powder of Ganoderma lucidum Improves Cancer-Related Fatigue in Breast Cancer Patients Undergoing Endocrine Therapy: A Pilot Clinical Trial. Evid. Based Complement. Altern. Med. 2012, 2012, 809614. [Google Scholar] [CrossRef] [PubMed]
  14. Deng, G.; Lin, H.; Seidman, A.; Fornier, M.; D’Andrea, G.; Wesa, K.; Yeung, S.; Cunningham-Rundles, S.; Vickers, A.J.; Cassileth, B. A phase I/II trial of a polysaccharide extract from Grifola frondosa (Maitake mushroom) in breast cancer patients: Immunological effects. J. Cancer Res. Clin. Oncol. 2009, 135, 1215–1221. [Google Scholar] [CrossRef] [PubMed]
  15. White, R.W.d.; Hackman, R.M.; Soares, S.E.; Beckett, L.A.; Sun, B. Effects of a mushroom mycelium extract on the treatment of prostate cancer. Urology 2002, 60, 640–644. [Google Scholar] [CrossRef]
  16. Seiden, M.V.; Gordon, A.N.; Bodurka, D.C.; Matulonis, U.A.; Penson, R.T.; Reed, E.; Alberts, D.S.; Weems, G.; Cullen, M.; McGuire, W.P., III. A phase II study of irofulven in women with recurrent and heavily pretreated ovarian cancer. Gynecol. Oncol. 2006, 101, 55–61. [Google Scholar] [CrossRef]
  17. Torkelson, C.J.; Sweet, E.; Martzen, M.R.; Sasagawa, M.; Wenner, C.A.; Gay, J.; Putiri, A.; Standish, L.J. Phase 1 clinical trial of Trametes versicolor in women with breast cancer. ISRN Oncol. 2012, 2012, 251632. [Google Scholar] [CrossRef]
  18. Chay, W.Y.; Tham, C.K.; Toh, H.C.; Lim, H.Y.; Tan, C.K.; Lim, C.; Wang, W.W.; Choo, S.P. Coriolus versicolor (Yunzhi) Use as Therapy in Advanced Hepatocellular Carcinoma Patients with Poor Liver Function or Who Are Unfit for Standard Therapy. J. Altern. Complement. Med. 2017, 23, 648–652. [Google Scholar] [CrossRef]
  19. Habtemariam, S. Trametes versicolor (Synn. Coriolus versicolor) polysaccharides in cancer therapy: Targets and efficacy. Biomedicines 2020, 8, 135. [Google Scholar] [CrossRef]
  20. Ayeka, P.A. Potential of Mushroom Compounds as Immunomodulators in Cancer Immunotherapy: A Review. Evid. Based Complement. Altern. Med. 2018, 2018, 7271509. [Google Scholar] [CrossRef]
  21. Chen, J.; Seviour, R. Medicinal importance of fungal beta-(1→3), (1→6)-glucans. Mycol. Res. 2007, 111 Pt 6, 635–652. [Google Scholar] [CrossRef]
  22. Israilides, C.; Kletsas, D.; Arapoglou, D.; Philippoussis, A.; Pratsinis, H.; Ebringerova, A.; Hribalova, V.; Harding, S.E. In Vitro cytostatic and immunomodulatory properties of the medicinal mushroom Lentinula edodes. Phytomedicine 2008, 15, 512–519. [Google Scholar] [CrossRef] [PubMed]
  23. Mizuno, T. The Extraction and Development of Antitumor-Active Polysaccharides from Medicinal Mushrooms in Japan (Review). Int. J. Med. Mushrooms 1999, 1, 9–29. [Google Scholar] [CrossRef]
  24. Wasser, S.P. Medicinal Mushrooms in Human Clinical Studies. Part I. Anticancer, Oncoimmunological, and Immunomodulatory Activities: A Review. Int. J. Med. Mushrooms 2017, 19, 279–317. [Google Scholar] [CrossRef] [PubMed]
  25. Yoon, S.Y.; Park, S.J.; Park, Y.J. The Anticancer Properties of Cordycepin and Their Underlying Mechanisms. Int. J. Mol. Sci. 2018, 19, 3027. [Google Scholar] [CrossRef]
  26. Patel, S.; Goyal, A. Recent developments in mushrooms as anti-cancer therapeutics: A review. 3 Biotech 2012, 2, 1–15. [Google Scholar] [CrossRef]
  27. Chang, T.C.; Yeh, C.T.; Adebayo, B.O.; Lin, Y.C.; Deng, L.; Rao, Y.K.; Huang, C.C.; Lee, W.H.; Wu, A.T.; Hsiao, M.; et al. 4-Acetylantroquinonol B inhibits colorectal cancer tumorigenesis and suppresses cancer stem-like phenotype. Toxicol. Appl. Pharmacol. 2015, 288, 258–268. [Google Scholar] [CrossRef]
  28. Lin, H.C.; Lin, M.H.; Liao, J.H.; Wu, T.H.; Lee, T.H.; Mi, F.L.; Wu, C.H.; Chen, K.C.; Cheng, C.H.; Lin, C.W. Antroquinonol, a Ubiquinone Derivative from the Mushroom Antrodia camphorata, Inhibits Colon Cancer Stem Cell-like Properties: Insights into the Molecular Mechanism and Inhibitory Targets. J. Agric. Food Chem. 2017, 65, 51–59. [Google Scholar] [CrossRef]
  29. Park, D.K.; Lim, Y.H.; Park, H.-J. Antrodia camphorata grown on germinated brown rice inhibits HT-29 human colon carcinoma proliferation through inducing G0/G1 phase arrest and apoptosis by targeting the β-catenin signaling. J. Med. Food 2013, 16, 681–691. [Google Scholar] [CrossRef]
  30. Park, D.K.; Park, H.-J. Anti-proliferative properties of ethyl acetate extract of Phellinus linteus grown on ginseng (Panax ginseng) on HT-29 human colon carcinoma cells through inducing apoptosis. Food Sci. Biotechnol. 2012, 21, 683–690. [Google Scholar] [CrossRef]
  31. Choi, C.W.; Yoon, J.-W.; Yon, G.H.; Kim, Y.S.; Ryu, S.Y.; Seok, S.-J.; Kang, S.; Kim, Y.H. Multidrug resistance reversal activity of methanol extracts from basidiomycete mushrooms in cancer cells. Nat. Prod. Sci. 2012, 18, 239–243. [Google Scholar]
  32. Kadomatsu, M.; Nakajima, S.; Kato, H.; Gu, L.; Chi, Y.; Yao, J.; Kitamura, M. Cordycepin as a sensitizer to tumour necrosis factor (TNF)-α-induced apoptosis through eukaryotic translation initiation factor 2α (eIF2α)- and mammalian target of rapamycin complex 1 (mTORC1)-mediated inhibition of nuclear factor (NF)-κB. Clin. Exp. Immunol. 2012, 168, 325–332. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, G.; Wang, L.; Zhou, J.; Xu, X. The Possible Role of PD-1 Protein in Ganoderma lucidum-Mediated Immunomodulation and Cancer Treatment. Integr. Cancer Ther. 2019, 18, 1534735419880275. [Google Scholar] [CrossRef] [PubMed]
  34. OuYang, F.; Wang, G.; Guo, W.; Zhang, Y.; Xiang, W.; Zhao, M. AKT signalling and mitochondrial pathways are involved in mushroom polysaccharide-induced apoptosis and G1 or S phase arrest in human hepatoma cells. Food Chem. 2013, 138, 2130–2139. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, C.L.; Meng, M.; Liu, S.B.; Wang, L.R.; Hou, L.H.; Cao, X.H. A chemically sulfated polysaccharide from Grifola frondos induces HepG2 cell apoptosis by notch1–NF-κB pathway. Carbohydr. Polym. 2013, 95, 282–287. [Google Scholar] [CrossRef] [PubMed]
  36. Kim, T.I.; Choi, J.-G.; Kim, J.H.; Li, W.; Chung, H.-S. Blocking Effect of Chaga Mushroom (Inonotus oliquus) Extract for Immune Checkpoint CTLA-4/CD80 Interaction. Appl. Sci. 2020, 10, 5774. [Google Scholar] [CrossRef]
  37. Kang, J.H.; Jang, J.E.; Mishra, S.K.; Lee, H.J.; Nho, C.W.; Shin, D.; Jin, M.; Kim, M.K.; Choi, C.; Oh, S.H. Ergosterol peroxide from Chaga mushroom (Inonotus obliquus) exhibits anti-cancer activity by down-regulation of the β-catenin pathway in colorectal cancer. J. Ethnopharmacol. 2015, 173, 303–312. [Google Scholar] [CrossRef]
  38. Huang, G.J.; Yang, C.M.; Chang, Y.S.; Amagaya, S.; Wang, H.C.; Hou, W.C.; Huang, S.S.; Hu, M.L. Hispolon suppresses SK-Hep1 human hepatoma cell metastasis by inhibiting matrix metalloproteinase-2/9 and urokinase-plasminogen activator through the PI3K/Akt and ERK signaling pathways. J. Agric. Food Chem. 2010, 58, 9468–9475. [Google Scholar] [CrossRef]
  39. Song, K.S.; Li, G.; Kim, J.S.; Jing, K.; Kim, T.D.; Kim, J.P.; Seo, S.B.; Yoo, J.K.; Park, H.D.; Hwang, B.D.; et al. Protein-bound polysaccharide from Phellinus linteus inhibits tumor growth, invasion, and angiogenesis and alters Wnt/β-catenin in SW480 human colon cancer cells. BMC Cancer 2011, 11, 307. [Google Scholar] [CrossRef]
  40. Park, H.J.; Park, J.B.; Lee, S.J.; Song, M. Phellinus linteus Grown on Germinated Brown Rice Increases Cetuximab Sensitivity of KRAS-Mutated Colon Cancer. Int. J. Mol. Sci. 2017, 18, 1746. [Google Scholar] [CrossRef]
  41. Park, H.-J. Phellinus linteus grown on germinated brown rice Suppress metastasis and Induce Apoptosis of Colon Cancer Cells by suppressing NF-κB and Wnt/β-catenin Signaling Pathways. J. Funct. Foods 2015, 14, 289–298. [Google Scholar] [CrossRef]
  42. Park, H.-J.; Han, E.s.; Park, D.K. The ethyl acetate extract of PGP (Phellinus linteus grown on Panax ginseng) suppresses B16F10 melanoma cell proliferation through inducing cellular differentiation and apoptosis. J. Ethnopharmacol. 2010, 132, 115–121. [Google Scholar] [CrossRef] [PubMed]
  43. Teng, Y.N.; Wang, Y.H.; Wu, T.S.; Hung, H.Y.; Hung, C.C. Zhankuic Acids A, B and C from Taiwanofungus Camphoratus Act as Cytotoxicity Enhancers by Regulating P-Glycoprotein in Multi-Drug Resistant Cancer Cells. Biomolecules 2019, 9, 759. [Google Scholar] [CrossRef] [PubMed]
  44. Ramesh, V.; Santosh, K.; Anand, T.D.; Shanmugaiah, V.; Kotamraju, S.; Karunakaran, C.; Rajendran, A. Novel Bioactive Wild Medicinal Mushroom--Xylaria sp. R006 (Ascomycetes) against Multidrug Resistant Human Bacterial Pathogens and Human Cancer Cell Lines. Int. J. Med. Mushrooms 2015, 17, 1005–1017. [Google Scholar] [CrossRef]
  45. Lum, B.L.; Gosland, M.P.; Kaubisch, S.; Sikic, B.I. Molecular targets in oncology: Implications of the multidrug resistance gene. Pharmacotherapy 1993, 13, 88–109. [Google Scholar] [PubMed]
  46. Ueda, K.; Cardarelli, C.; Gottesman, M.M.; Pastan, I. Expression of a full-length cDNA for the human "MDR1" gene confers resistance to colchicine, doxorubicin, and vinblastine. Proc. Natl. Acad. Sci. USA 1987, 84, 3004–3008. [Google Scholar] [CrossRef]
  47. Sadava, D.; Still, D.W.; Mudry, R.R.; Kane, S.E. Effect of Ganoderma on drug-sensitive and multidrug-resistant small-cell lung carcinoma cells. Cancer Lett. 2009, 277, 182–189. [Google Scholar] [CrossRef]
  48. Sohretoglu, D.; Huang, S. Ganoderma lucidum polysaccharides as an anti-cancer agent. Anti-Cancer Agents Med. Chem. (Former. Curr. Med. Chem. -Anti-Cancer Agents) 2018, 18, 667–674. [Google Scholar] [CrossRef]
  49. Grabie, N.; Gotsman, I.; DaCosta, R.; Pang, H.; Stavrakis, G.; Butte, M.J.; Keir, M.E.; Freeman, G.J.; Sharpe, A.H.; Lichtman, A.H. Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart. Circulation 2007, 116, 2062–2071. [Google Scholar] [CrossRef]
  50. Meng, Q.; Yang, P.; Li, B.; Zhou, H.; Huang, X.; Zhu, L.; Ren, Y.; Kijlstra, A. CD4+PD-1+ T cells acting as regulatory cells during the induction of anterior chamber-associated immune deviation. Investig. Ophthalmol. Vis. Sci. 2006, 47, 4444–4452. [Google Scholar] [CrossRef]
  51. Maier, H.; Isogawa, M.; Freeman, G.J.; Chisari, F.V. PD-1:PD-L1 interactions contribute to the functional suppression of virus-specific CD8+ T lymphocytes in the liver. J. Immunol. 2007, 178, 2714–2720. [Google Scholar] [CrossRef]
  52. Nowicki, T.S.; Hu-Lieskovan, S.; Ribas, A. Mechanisms of Resistance to PD-1 and PD-L1 Blockade. Cancer J. 2018, 24, 47–53. [Google Scholar] [CrossRef] [PubMed]
  53. Ding, J.; Ning, Y.; Bai, Y.; Xu, X.; Sun, X.; Qi, C. β-Glucan induces autophagy in dendritic cells and influences T-cell differentiation. Med. Microbiol. Immunol. 2019, 208, 39–48. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, M.; Bai, Y.; Pei, J.; Li, D.; Pu, X.; Zhu, W.; Xia, L.; Qi, C.; Jiang, H.; Ning, Y. beta-Glucan Combined With PD-1/PD-L1 Checkpoint Blockade for Immunotherapy in Patients With Advanced Cancer. Front. Pharmacol. 2022, 13, 887457. [Google Scholar] [CrossRef] [PubMed]
  55. Li, W.; Kim, T.I.; Kim, J.H.; Chung, H.S. Immune Checkpoint PD-1/PD-L1 CTLA-4/CD80 are Blocked by Rhus verniciflua Stokes and its Active Compounds. Molecules 2019, 24, 4062. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [PubMed]
  57. Nowakowski, P.; Markiewicz-Zukowska, R.; Bielecka, J.; Mielcarek, K.; Grabia, M.; Socha, K. Treasures from the forest: Evaluation of mushroom extracts as anti-cancer agents. Biomed. Pharmacother. 2021, 143, 112106. [Google Scholar] [CrossRef]
  58. Cheng, Y.; Xie, P. Ganoderic acid A holds promising cytotoxicity on human glioblastoma mediated by incurring apoptosis and autophagy and inactivating PI3K/AKT signaling pathway. J. Biochem. Mol. Toxicol. 2019, 33, e22392. [Google Scholar] [CrossRef]
  59. Anastas, J.N.; Moon, R.T. WNT signalling pathways as therapeutic targets in cancer. Nat. Rev. Cancer 2013, 13, 11–26. [Google Scholar] [CrossRef]
  60. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
  61. Sweis, R.F.; Spranger, S.; Bao, R.; Paner, G.P.; Stadler, W.M.; Steinberg, G.; Gajewski, T.F. Molecular Drivers of the Non-T-cell-Inflamed Tumor Microenvironment in Urothelial Bladder Cancer. Cancer Immunol. Res. 2016, 4, 563–568. [Google Scholar] [CrossRef]
  62. Zhang, H.; Bi, Y.; Wei, Y.; Liu, J.; Kuerban, K.; Ye, L. Blocking Wnt/β-catenin Signal Amplifies Anti-PD-1 Therapeutic Efficacy by Inhibiting Tumor Growth, Migration, and Promoting Immune Infiltration in GlioblastomasTargeting Wnt/β-catenin Signal and PD-1 in GBM. Mol. Cancer Ther. 2021, 20, 1305–1315. [Google Scholar] [CrossRef] [PubMed]
  63. Das, A.; Alshareef, M.; Henderson, F., Jr.; Santos, J.L.M.; Vandergrift, W.A., 3rd; Lindhorst, S.M.; Varma, A.K.; Infinger, L.; Patel, S.J.; Cachia, D. Ganoderic acid A/DM-induced NDRG2 over-expression suppresses high-grade meningioma growth. Clin. Transl. Oncol. 2020, 22, 1138–1145. [Google Scholar] [CrossRef]
  64. Ina, H.; Yoneda, M.; Mitsuro, K.; Kodera, Y.; Kabeya, M.; Yuasa, S.; Kataoka, T.; Furuta, R.; Ina, K. Lentinan, A Shiitake Mushroom ÃÂ2-Glucan, Stimulates Tumor-Specific Adaptive Immunity through PD-L1 Down-Regulation in Gastric Cancer Cells. Med. Chem. 2016, 6, 710–714. [Google Scholar] [CrossRef]
  65. Hilger, R.A.; Scheulen, M.E.; Strumberg, D. The Ras-Raf-MEK-ERK pathway in the treatment of cancer. Onkologie 2002, 25, 511–518. [Google Scholar] [CrossRef] [PubMed]
  66. Lin, S.B.; Li, C.H.; Lee, S.S.; Kan, L.S. Triterpene-enriched extracts from Ganoderma lucidum inhibit growth of hepatoma cells via suppressing protein kinase C, activating mitogen-activated protein kinases and G2-phase cell cycle arrest. Life Sci. 2003, 72, 2381–2390. [Google Scholar] [CrossRef]
  67. Zaidman, B.-Z.; Yassin, M.; Mahajna, J.; Wasser, S.P. Medicinal mushroom modulators of molecular targets as cancer therapeutics. Appl. Microbiol. Biotechnol. 2005, 67, 453–468. [Google Scholar] [CrossRef] [PubMed]
  68. Yang, H.L.; Kuo, Y.H.; Tsai, C.T.; Huang, Y.T.; Chen, S.C.; Chang, H.W.; Lin, E.; Lin, W.H.; Hseu, Y.C. Anti-metastatic activities of Antrodia camphorata against human breast cancer cells mediated through suppression of the MAPK signaling pathway. Food Chem. Toxicol. 2011, 49, 290–298. [Google Scholar] [CrossRef]
  69. Thyagarajan, A.; Jedinak, A.; Nguyen, H.; Terry, C.; Baldridge, L.A.; Jiang, J.; Sliva, D. Triterpenes from Ganoderma Lucidum induce autophagy in colon cancer through the inhibition of p38 mitogen-activated kinase (p38 MAPK). Nutr. Cancer 2010, 62, 630–640. [Google Scholar] [CrossRef]
  70. Petrova, R.D.; Reznick, A.Z.; Wasser, S.P.; Denchev, C.M.; Nevo, E.; Mahajna, J. Fungal metabolites modulating NF-κB activity: An approach to cancer therapy and chemoprevention. Oncol. Rep. 2008, 19, 299–308. [Google Scholar] [CrossRef]
  71. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef]
  72. Lin, T.-C.; Germagian, A.; Liu, Z. The NF-κB Signaling and Wnt/β-catenin Signaling in MCF-7 Breast Cancer Cells in Response to Bioactive Components from Mushroom Antrodia Camphorata. Am. J. Chin. Med. 2021, 49, 199–215. [Google Scholar] [CrossRef] [PubMed]
  73. Sliva, D.; Labarrere, C.; Slivova, V.; Sedlak, M.; Lloyd Jr, F.P.; Ho, N.W. Ganoderma lucidum suppresses motility of highly invasive breast and prostate cancer cells. Biochem. Biophys. Res. Commun. 2002, 298, 603–612. [Google Scholar] [CrossRef]
  74. Mattila, P.; Könkö, K.; Eurola, M.; Pihlava, J.-M.; Astola, J.; Vahteristo, L.; Hietaniemi, V.; Kumpulainen, J.; Valtonen, M.; Piironen, V. Contents of vitamins, mineral elements, and some phenolic compounds in cultivated mushrooms. J. Agric. Food Chem. 2001, 49, 2343–2348. [Google Scholar] [CrossRef] [PubMed]
  75. Nakamura, T.; Akiyama, Y.; Matsugo, S.; Uzuka, Y.; Shibata, K.; Kawagishi, H. Purification of caffeic acid as an antioxidant from submerged culture mycelia of Phellinus linteus (Berk. et Curt.) Teng (Aphyllophoromycetideae). Int. J. Med. Mushrooms 2003. [Google Scholar] [CrossRef]
  76. Aras, A.; Khalid, S.; Jabeen, S.; Farooqi, A.A.; Xu, B. Regulation of cancer cell signaling pathways by mushrooms and their bioactive molecules: Overview of the journey from benchtop to clinical trials. Food Chem. Toxicol. 2018, 119, 206–214. [Google Scholar] [CrossRef] [PubMed]
  77. Rossi, P.; Difrancia, R.; Quagliariello, V.; Savino, E.; Tralongo, P.; Randazzo, C.L.; Berretta, M. B-glucans from Grifola frondosa and Ganoderma lucidum in breast cancer: An example of complementary and integrative medicine. Oncotarget 2018, 9, 24837–24856. [Google Scholar] [CrossRef] [PubMed]
  78. Wasser, S.P.; Weis, A.L. Medicinal properties of substances occurring in higher basidiomycetes mushrooms: Current perspectives. Int. J. Med. Mushrooms 1999, 1, 31–62. [Google Scholar] [CrossRef]
  79. Liu, C.; Choi, M.W.; Xue, X.; Cheung, P.C.K. Immunomodulatory Effect of Structurally Characterized Mushroom Sclerotial Polysaccharides Isolated from Polyporus rhinocerus on Bone Marrow Dendritic Cells. J. Agric. Food Chem. 2019, 67, 12137–12143. [Google Scholar] [CrossRef]
  80. Wasser, S.P.; Weis, A.L. Therapeutic effects of substances occurring in higher Basidiomycetes mushrooms: A modern perspective. Crit. Rev. Immunol. 1999, 19, 65–96. [Google Scholar]
  81. Tangen, J.M.; Tierens, A.; Caers, J.; Binsfeld, M.; Olstad, O.K.; Troseid, A.M.; Wang, J.; Tjonnfjord, G.E.; Hetland, G. Immunomodulatory effects of the Agaricus blazei Murrill-based mushroom extract AndoSan in patients with multiple myeloma undergoing high dose chemotherapy and autologous stem cell transplantation: A randomized, double blinded clinical study. Bio. Res. Int. 2015, 2015, 718539. [Google Scholar] [CrossRef]
  82. Sorimachi, K.; Akimoto, K.; Ikehara, Y.; Inafuku, K.; Okubo, A.; Yamazaki, S. Secretion of TNF-alpha, IL-8 and nitric oxide by macrophages activated with Agaricus blazei Murill fractions in vitro. Cell Struct. Funct. 2001, 26, 103–108. [Google Scholar] [CrossRef] [PubMed]
  83. Tang, W.; Gao, Y.; Chen, G.; Gao, H.; Dai, X.; Ye, J.; Chan, E.; Huang, M.; Zhou, S. A randomized, double-blind and placebo-controlled study of a Ganoderma lucidum polysaccharide extract in neurasthenia. J. Med. Food 2005, 8, 53–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kwon, H.-K.; Jo, W.-R.; Park, H.-J. Immune-enhancing activity of C. militaris fermented with Pediococcus pentosaceus (GRC-ON89A) in CY-induced immunosuppressed model. BMC Complement. Altern. Med. 2018, 18, 75. [Google Scholar] [CrossRef] [PubMed]
  85. Goodridge, H.S.; Wolf, A.J.; Underhill, D.M. Beta-glucan recognition by the innate immune system. Immunol. Rev. 2009, 230, 38–50. [Google Scholar] [CrossRef]
  86. Kumagai, Y.; Akira, S. Identification and functions of pattern-recognition receptors. J. Allergy Clin. Immunol. 2010, 125, 985–992. [Google Scholar] [CrossRef] [PubMed]
  87. Boh, B. Ganoderma lucidum: A potential for biotechnological production of anti-cancer and immunomodulatory drugs. Recent Pat. Anti-Cancer Drug Discov. 2013, 8, 255–287. [Google Scholar] [CrossRef]
  88. Friedman, M. Mushroom Polysaccharides: Chemistry and Antiobesity, Antidiabetes, Anticancer, and Antibiotic Properties in Cells, Rodents, and Humans. Foods 2016, 5, 80. [Google Scholar] [CrossRef] [Green Version]
Figure 1. An illustration of the anticancer mechanisms of mushrooms by overcoming Pgp-mediated MDR. Structure of zhankuic acids A, B, and C from T. camphoratus adapted from Zhankuic Acids A, B and C from Taiwanofungus Camphoratus Act as Cytotoxicity Enhancers by Regulating P-Glycoprotein in Multi-Drug Resistant Cancer Cells. Biomolecules, 2019. 9(12).
Figure 1. An illustration of the anticancer mechanisms of mushrooms by overcoming Pgp-mediated MDR. Structure of zhankuic acids A, B, and C from T. camphoratus adapted from Zhankuic Acids A, B and C from Taiwanofungus Camphoratus Act as Cytotoxicity Enhancers by Regulating P-Glycoprotein in Multi-Drug Resistant Cancer Cells. Biomolecules, 2019. 9(12).
Ijms 23 10502 g001
Figure 2. The illustration of the anticancer mechanisms of mushrooms by inhibiting the PD-1 pathway and CTLA-4/CD80 pathway.
Figure 2. The illustration of the anticancer mechanisms of mushrooms by inhibiting the PD-1 pathway and CTLA-4/CD80 pathway.
Ijms 23 10502 g002
Table 1. Ongoing clinical trials for use of mushrooms in cancer treatment.
Table 1. Ongoing clinical trials for use of mushrooms in cancer treatment.
MushroomCancerPhaseStudy StatusActive Compound/sIdentifierInvestigator
Agaricus bisporusBreast cancer, cancer survivorsPhase 1CompletedPolysaccharides, lectinNCT00709020Shiuan Chen
Agaricus bisporusProstate cancerPhase 1bcompletedPolysaccharides, lectinNCT00779168W. Twardowski
Agaricus blazei Murill (AndoSan)Multiple myelomaPhase 2CompletedAgaricus polysaccharidesNCT00970021Jon-Magnus Tangen
Lentinula edodesProstate cancerNot mentionedCompletedGenistein Combined Polysaccharide (GCP)NCT00269555Robert Hackman
Lentinula edodeHepatocellular Carcinoma and Hepatitis B and C InfectionNot mentionedCompletedArabinoxylan extracted from rice bran treated enzymatically with extract from Lentinula edodeNCT01018381Mai Hong Bang
Grifola frondosaLung Neoplasms and Breast CarcinomaPhase 1CompletedPolysaccharidesNCT02603016Shunchang Jiao
Omphalotus illudensThyroid CancerPhase 2CompletedA semisynthetic derivative of illudin SNCT00124527Eisai Inc.
Omphalotus illudensRecurrent or Metastatic Gastric CancerPhase 2CompletedA semisynthetic derivative of illudin SNCT00062257Winnie Yeo
Omphalotus illudensRecurrent or Persistent Epithelial Ovarian CancerPhase 2CompletedA semisynthetic derivative of illudin SNCT00019552Gisele A. Sarosy
Trametes versicolorBreast cancerPhase 1CompletedKrestin, PSK, PSPNCT00680667Carolyn Torkelson
Table 2. Anticancer mechanisms of mushrooms and their bioactive substances.
Table 2. Anticancer mechanisms of mushrooms and their bioactive substances.
Mushroom Species/ReferenceBioactive SubstanceExperimental StudyTarget/Mechanism
Antrodia camphorata [27]4-Acetylantroquinonol BColorectal cancerDLD-1, HCT-116, SW-480, RKO, HT-29Lgr5/Wnt/β-catenin, JAK–STAT↓
Antrodia camphorata [21]Polysaccharide (ACE)Hepatocellular carcinomaHepG2 cell lineApoptosis
Antrodia camphorata [9] AntroquinonolPancreatic carcinomaPANC-1 and AsPC-1 cellsAKT at p-Ser 473↓
mTOR at p-Ser 2448↓
Antrodia camphorata [28]AntroquinonolColon cancerHCT15, HCT-116 and LoVo cellsPI3K/AKT/β-catenin signaling↓
Antrodia camphorata grown on germinated brown rice (CBR) [29]AdenosineMelanoma MITF and TRP-1↑, p53↑
Antrodia camphorata grown on germinated brown rice (CBR) [30] Colon cancer β-catenin pathway↓
Cantharellus cibarius [31] Drug resistance in Pgp-expressing tumor cells↓
Cordyceps militaris [32]Cordycepin NRK-52E cell lineNF-κB↓
Ganoderma lucidum [33] PD-1 protein↓
Ganoderma lucidum [34] PolysaccharideLiver cancerHepG2, Bel-7404p27kip↑, cyclinD1/CDK4↓, cyclin E/CDK2↓, AKT at p-Thr 308 and p-Ser 473↓, pPTEN↑, Bcl-2 activation, apoptosis, caspase 3 and 9↑
Grifola frondose [35]Sulfated polysaccharideLiver cancerHepG2Apoptosis, S phase arrest, NOTCH1↓, IκB-α degradation, FLIP↓, Caspase 3 and 8↑
Inonotus obliquus [36]Lanosterol, terpenoid CTLA-4/CD80 interaction↓
Activation of T cells↑
Inonotus obliquus [37]Ergosterol peroxideColorectal cancerHCT116, HT-29, SW620, DLD-1 CRC cell linesβ-catenin pathway↓
Phellinus linteus [38]HispolonHuman hepatoma cellsSK-Hep1 cellsMMP2↓, MMP9↓, uPA↓, p-ERK1/2, p-PI3K/AKT↓, p-FAK↓
Phellinus linteus [39]Protein-bound polysaccharideColon cancerSW480 cellsWnt/β-catenin Pathway↓, Cyclin D1↓, TCF/LEF↓
Phellinus linteus [34]PolysaccharideLiver cancerHepG2, Bel-7404p27kip↑, cyclinD1/CDK4↓, cyclin E/CDK2↓, AKT at p-Thr 308 and p-Ser 473↓, pPTEN↑, Bcl-2 activation, apoptosis, caspase 3 and 9↑
Phellinus linteus grown on germinated brown rice (PBR) [40]Not determinedKRAS-mutated colon cancer MAPK pathway↓
Phellinus linteus grown on germinated brown rice (PBR) [41]γ-Aminobutyric Acid and β-glucancolon cancer metastasized to the lung NF-κB, β-catenin, MAPK pathway↓, MMP2 and 9 activities↓
Phellinus linteus grown on Panax ginseng (PGP) [42]Rd, Rg1, Re, Rb2, and Rg3melanoma Caspase 8 and 9, p53 and p21 ↑
Phellinus linteus grown on Panax ginseng (PGP) [30]Not determinedColon cancer Caspase 8 and 9↑
Russula emetic [31] Drug resistance in Pgp-expressing tumor cells↓
Taiwanofungus camphoratus [43]Zhankuic acids A–C Drug resistance in Pgp-expressing tumor cells↓
Note: ↑, upregulation; ↓, downregulation; p, phosphorylation; NG, not given.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Park, H.-J. Current Uses of Mushrooms in Cancer Treatment and Their Anticancer Mechanisms. Int. J. Mol. Sci. 2022, 23, 10502. https://doi.org/10.3390/ijms231810502

AMA Style

Park H-J. Current Uses of Mushrooms in Cancer Treatment and Their Anticancer Mechanisms. International Journal of Molecular Sciences. 2022; 23(18):10502. https://doi.org/10.3390/ijms231810502

Chicago/Turabian Style

Park, Hye-Jin. 2022. "Current Uses of Mushrooms in Cancer Treatment and Their Anticancer Mechanisms" International Journal of Molecular Sciences 23, no. 18: 10502. https://doi.org/10.3390/ijms231810502

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