Skip to main content
Integrative Cancer Therapies logoLink to Integrative Cancer Therapies
. 2017 Nov 2;17(2):200–209. doi: 10.1177/1534735417736861

A Preclinical Evaluation of the Antitumor Activities of Edible and Medicinal Mushrooms: A Molecular Insight

Thomson Patrick Joseph 1, Warren Chanda 1, Arshad Ahmed Padhiar 1, Samana Batool 1, Shao LiQun 1, MinTao Zhong 1, Min Huang 1,
PMCID: PMC6041903  PMID: 29094602

Abstract

Cancer is the leading cause of morbidity and mortality around the globe. For certain types of cancer, chemotherapy drugs have been extensively used for treatment. However, severe side effects and the development of resistance are the drawbacks of these agents. Therefore, development of new agents with no or minimal side effects is of utmost importance. In this regard, natural compounds are well recognized as drugs in several human ailments, including cancer. One class of fungi, “mushrooms,” contains numerous compounds that exhibit interesting biological activities, including antitumor activity. Many researchers, including our own group, are focusing on the anticancer potential of different mushrooms and the underlying molecular mechanism behind their action. The aim of this review is to discuss PI3K/AKT, Wnt-CTNNB1, and NF-κB signaling pathways, the occurrence of genetic alterations in them, the association of these aberrations with different human cancers and how different nodes of these pathways are targeted by various substances of mushroom origin. We have given evidence to propose the therapeutic attributes and possible mode of molecular actions of various mushroom-originated compounds. However, anticancer effects were typically demonstrated in in vitro and in vivo models and very limited number of studies have been conducted in the human population. It is our belief that this review will help the research community in designing concrete preclinical and clinical studies to test the anticancer potential of mushroom-originated compounds on different cancers harboring particular genetic alteration(s).

Keywords: cancer, signaling pathway, genetic alterations, mushroom, molecular insight

Introduction

Globally, the morbidity and mortality rate because of cancer is increasing constantly. In one estimate, approximately 1 688 780 new cases and 600 920 cancer deaths are expected to occur in 2017 in the United States alone.1 The modern era of cancer chemotherapy began in 1942; since then chemotherapeutic agents have been used extensively as single agents,2 in combination,3 and in conjunction with radiotherapy.4-6 However, development of resistance to chemotherapy7-9 and molecular-targeted therapies,10-13 and severe side effects14 are the major pitfalls of these agents. Therefore, life scientists are focusing more aggressively on the development of novel agents exhibiting antitumor and antimetastatic potential with no or minimal side effects.

The chemical compound or a substance produced by a living organism, found in nature is termed a “natural product.” Historically, chemical substances derived from animals, plants, and microorganisms have been found to have anticancer potential.15,16 Later, many compounds have been isolated from natural resources and among them, antitumor antibiotics are of great importance, including those isolated from various species of Streptomyces.17-20 Keeping in mind the role and contribution of natural products in drug discovery, one cannot ignore the therapeutic potential and contribution of edible and medicinal mushrooms in the field of medicine. Out of 14 000 different species of mushrooms, approximately 700 exhibit medicinal properties.21 Many of them have long been used in traditional Chinese medicine (TCM) and now interest has increased in the extraction of secondary metabolites for the discovery of novel drugs or lead compounds. Series of bioactive components22-24 have been isolated from mushrooms having antioxidant, antitumor, antiviral, antimicrobial and immunomodulating activities. Despite the discovery of many antitumor components of mushrooms, still, there is a need to increase the range of available compounds, find more effective and less toxic molecules and to understand deeply single and/or multiple targets, so as to battle cancer more precisely.

In this review, we have given evidence that the compounds of mushroom (Basidiomycota, Ascomycota) origin have antitumor and antimetastatic activity in in vitro and in vivo models, with a particular focus on the signaling pathways they target. Our first goal is to briefly portray a picture of genetic alterations associated with PI3K/AKT, Wnt-CTNNB1, andNF-κB signaling pathways and their role in the initiation and progression of cancer. Our second goal is to emphasize the antitumor potential of mushroom origin compounds in a signaling pathway-specific manner. It is our hope that appreciating the role of these mushroom originated compounds will help in targeting cancer in alteration-specific and cancer type–specific manner.

The Anticancer Role of Mushrooms: Mechanistic Approach

In Asian countries, medicinal use of mushrooms has a long history; however, in recent decades, their use has also increased in other parts of the world. Many mushrooms are in clinical trials, and a few are used in clinics (Table 1). The tremendous therapeutic potential of edible and medicinal mushrooms is because of the lectin, arginine, ergosterol, β-glucan, and other bioactive substances in mushrooms. Some of the bioactive substances of mushroom origin, with experimental model type and possible molecular mechanisms are listed in Table 2. However, there are a few shortcomings in the already existing investigations, including limited studies on the interaction of mushroom components with different nodes of the signal transduction pathways. Few studies show the antitumor effects in vivo models and many lack the IC50 value of the agent under consideration. In order to target cancer more precisely, it is necessary to have a good understanding of both the molecular mechanisms underlying the development and progression of cancer and of the molecular targets of mushroom compounds at the same time. Furthermore, in this review, we discuss in some detail PI3K/AKT, Wnt-CTNNB1, and NF-κB signaling pathways, the occurrence of genetic alterations in them, the association of these genetic alterations in human cancers and how different nodes of these pathways are targeted by mushroom origin compounds.

Table 1.

Mushrooms in Clinical Trials and in the Clinics.

Mushroom Clinical Trial Description
Identifier Investigator First Received Last Updated Cancer/Condition Phase Status
Maitake NCT02603016 Shunchang Jiao 2015 2015 Lung neoplasms Phase 1 Ongoing
Maitake NCT02603016 Shunchang Jiao 2015 2015 Breast carcinoma Phase 2 Ongoing
Reishi NCT02486796 Christina Shannon 2015 2016 Breast cancer, quality of life Phases 1 and 2 Ongoing
Agaricus blazei murill NCT00970021 Jon-Magnus Tangen 2009 2014 Multiple myeloma Phase 2 Completed
Coriolus versicolor NCT00680667 Carolyn Torkelson 2008 2012 Breast cancer Phase 1 Completed
Yunzhi NCT00647075 Pere Gascon 2008 2010 Breast cancer Phase 4 Ongoing
White button NCT00709020 Shiuan Chen 2008 2015 Breast cancer, cancer survivor Phase 1 Completed
White button NCT00779168 W. Twardowski 2008 2016 Prostate cancer Phase 1 Ongoing
Shiitake NCT00269555 Robert Hackman 2005 2010 Prostate cancer Not mentioned Completed
Mushroom Clinical Description
Bioactive Substance Commercial Name of Product Mode of Action Route of Administration
Coriolus versicolor Protein bound polysaccharide PSK, Krestin Immunostimulant, inhibit tumor growth Oral
Coriolus versicolor Polysaccharide peptide PSP Immunostimulant, inhibit tumor growth Oral
Grifola frondosa Polysaccharide Grifolan Antiviral, antitumor, balance immune system Oral
Lentinula edodes Polysaccharide Lentinan Immunostimulant Intravenously
Schizophyllum commune Polysaccharide SPG, Sonifilan Immunostimulant Intratumoral

Table 2.

Effect of Mushroom’s Bioactive Substances on Different Cancer Types: In Vitro and In Vivo Experiments.a

Class Mushroom Species/Reference Bioactive Substance Experimental Study
Concentration Used IC50 Target/Mechanism
Cancer Type Model Type
Edible mushrooms Albatrellus confluens 25 Grifolin Osteosarcoma U2OS, MG63 cell line 0, 5, 10, 25, 50, or 100 μM NG Activation of caspase 3,9, cleavage of PARP, ⊗AKT, FOXO↓, GSK3↓
Albatrellus confluens 26 Neogrifolin Osteosarcoma U2OS, MG63 cell line 0, 5, 10, 25, 50, or 100 μM NG Activation of caspase 3,9, cleavage of PARP, ⊗AKT, FOXO↓, GSK3↓, IAP↓
Auricularia auricula 27 Polysaccharide Liver cancer HepG2, Bel-7404 0, 0.25, 0.5, and 1.0 mg/mL NG p27kip↑, cyclinD1/CDK4↓, cyclin E/CDK2↓, ⊗AKT at Thr 308 and Ser 473, pPTEN↑,Bcl-2 activation, Apoptosis, cCaspase 3 and 9↑
Grifola frondosa 28 Polysaccharide Liver cancer Hep3B/Nude mice 0, 250, 500, and 1000 µg/mL, 10, 20 and 50 mg/kg NG PI3K ↓, cJun ↑, S phase arrest, apoptosis
Grifola frondosa 29 Protein-bound polysaccharide Breast cancer MCF7 cell line 0, 18, 36, 91, 183, 367 mg/mL NG Apoptosis, BAK-1 ↑, cytochrome c transcript ↑
Lentinus crinitus 30 Panepoxydone (PP) Breast cancer MDA-MB-453, MCF-7, MDA-MB-468 and MDA-MB-231 cell lines 2.5, 5, and 10 μM/2, 4, and 8 μM/3, 6, and 12 μM/7.5,15, and 30 μM 4, 5, 6, and 15 mM Bax ↑, cPARP↑. Bcl-2↓, survivin↓. cyclin D1↓, caspase 3↓ FOXO1 ↓,⊗ IκBα
Lentinula edodes 31 *Protein (Latcripin-1) Lung cancer A549 cell line 7.5, 15, and 30 µg/mL NG Apoptosis
Lentinula edodes 32 *Protein (Latcripin-3) Lung cancer A549 cell line 7.5, 15, and 30 µg/mL NG Apoptosis/S phase arrest
Lentinula edodes 33 *Protein (Latcripin-13) Lung cancer A549 cell line 6.25, 12.5, 25, 50, 100 and 200 µg/mL NG Apoptosis
Lentinula edodes 34 *Protein (Latcripin-15) Lung cancer A549 cell line 12.5, 25, 50, 100, and 200 µg/mL NG Apoptosis
Lentinus edodes 35 Ploysaccharide Hepatocarcinoma of mouse H22 cell line 0, 5, 50, and 500 µg/mL NG Apoptosis, G2/M Phase arrest
Pleurotus ferulae 36 Terpenoids Melanoma/Gastric cancer B16F10,BGC823, GES-1 cell line/C57BL/6 female mice 0, 0.4, 0.8, and 1.6 mg/mL NG Apoptosis, G0/G1 arrest, MMP ↓, caspase 3↑
Pleurotus ostreatus 37 Polysaccharide Sarcoma Sarcoma 180 cells /male BALB/c mice 100, 200, and 400 µg/mL and mg/kg NG TNF- and iNOS↑, phosphorylation of p65, IκB↓
Termitomyces clypeatus 38 Sugar entities Brain, breast, acute myeloid leukemia, lung, ovary, retinoblastoma U373MG, MDA-MB-468, HepG2, HL-60, A549, U937, OAW-42 and Y-79/Male Swiss albino mice 0.01, 0.1, 1, 10, and 100 µg/mL; 200, 400 mg/kg NG Antioxidant/anticancer
Medicinal mushrooms Amauroderma rude 39 Ergosterol Breast cancer MDA-MB-231, SK-BR-3, MDA-MB-468, MCF-7, 4T1 0, 50, 100, 150, 200, 250 and 300 µg/mL NG Growth inhibition and apoptosis. Fas↑, FasL↑, BimL↑, BimS ↑
Antrodia camphorata 40 Ploysaccharide (ACE) Hepatocellular carcinoma HepG2 cell line 2.65, 5.3, 10.6, and 21.2 µg/mL NG Apoptosis
Antrodia camphorata 41 Antroquinonol Pancreatic carcinoma PANC-1 and AsPC-1 cells 0.3, 1, 3, 10, and 30 μM 18.6 and 20.2 µM ⊗AKT at Ser 473, ⊗mTOR at Ser 2448
Antrodia camphorata 42 Antroquinonol Colon cancer HCT15, HCT-116 and LoVo cells 0, 2.5, 5, 10, 20, 40, and 80 μM 34.8, >80, and 17.9 µM PI3K/AKT/β-catenin signaling ↓
Antrodia camphorata 43 4-Acetylantroquinonol B Colorectal cancer DLD-1, HCT-116, SW-480, RKO, HT-29 0, 10, 20, 30, 40, 50, and 60 μM 11.3, 34.6, 38.1, 22.3, 39.2 µM Lgr5/Wnt/β-catenin, JAK–STAT↓
Cordyceps militaris 44 Cordycepin NRK-52E cell line 0, 0.5, 1, 2, and 5 µg/mL NG ↓NF-κB
Fomes fomentarius 45 Ploysaccharide (MFKF-AP1β) Lung cancer A549 cell line 25, 50, and 100 µg/mL NG Apoptosis
Grifola frondosa 46 Sulfated polysaccharide Liver cancer HepG2 0, 10, 20, 40, 60, 80, and 100 µg/mL 61 µg/mL Apoptosis, S phase arrest, notch1 ↓, IκB-α degradation, FLIP ↓, Caspase 3,8 ↑
Ganoderma lucidum 27 Polysaccharide Liver cancer HepG2, Bel-7404 0, 0.25, 0.5, and 1.0 mg/mL NG p27kip↑, cyclinD1/CDK4↓, cyclin E/CDK2↓, ⊗AKT at Thr 308 and Ser 473, pPTEN↑, Bcl-2 activation, apoptosis, caspase 3 and 9↑
Inonotus obliquus 47 Ergosterol peroxide Colorectal cancer HCT116, HT-29, SW620, DLD-1 CRC cell lines 0, 5, 10, and 20 µg/mL NG β-catenin pathway ↓
Phellinus linteus 48 Protein-bound polysaccharide Colon cancer SW480 cell line 125, 250, 500, and 1000 μg/mL NG Wnt/β-catenin Pathway ↓, Cyclin D1↓, TCF/LEF↓
Phellinus linteus 49 Hispolon Human hepatoma cells SK-Hep1 cells 4.6, 11.5, 22.8, 34.3, 45.7, 114, and 228 μM NG MMP2↓, MMP9↓, uPA ↓, ⊗ ERK1/2, ⊗PI3K/AKT, ⊗FAK
Phellinus linteus 27 Polysaccharide Liver cancer HepG2, Bel-7404 0, 0.25, 0.5, and 1.0 mg/mL NG p27kip↑, cyclinD1/CDK4↓, cyclin E/CDK2↓, ⊗AKT at Thr 308 and Ser 473, pPTEN↑, Bcl-2 activation, apoptosis, caspase 3 and 9↑
Pleurotus pulmonarius 50 Protein-bound polysaccharide Liver cancer Huh7, Hep3B, SMMC-7721, HepG2 0,25,50,100, 200, or 400 µg/mL NG VEGF-induced PI3K/AKT signaling↓
a

*recombinant protein; ↑ upregulation; ↓ downregulation; ⊗ phosphorylation inhibition; NG not given.

Targeting the PI3K/AKT Signaling Pathway in Cancer

Phosphoinositide 3-kinases (PI3Ks) are a key signal transduction system that on activation cause the activation of AKT and other downstream pathways.51 This signaling pathway has been reported to have a role in different cellular processes, including cell survival, proliferation, and differentiation.52,53 Several studies have declared that many of the components of this pathway are altered, including activation of growth factor receptor, PIK3CA or AKT amplification or mutation, and PTEN loss of function in common human cancers.54-56

Several lines of evidence indicate that mushroom compounds are capable enough of showing antitumor and antimetastatic activity by targeting various nodes of the PI3K/AKT pathway. In 2010, a group49 showed the dose-dependent inhibitory effect of “Hispolon” (Phellinus linteus) on invasion and motility of a highly metastatic liver cancer cell line (SK-Hep1), suggesting its role as an antimetastatic agent. According to their findings, this phenolic compound results in the down-regulation of MMP2, MMP9, and uPa. Together with this it also causes the phosphorylation inhibition of the ERK1/2, PI3K/AKT, and FAK pathways. In harmony with this study, the antiproliferative activity of proteoglycan (P1) from Phellinus linteus was demonstrated in multiple human cancer cells and in BALB/c-nu/nu mice bearing HT-29 cells.57 As far as the molecular mechanism is concerned, P1 resulted in a notable decrease in AKT, Reg IV, EGFR, and plasmatic PGE2 concentrations measured by ELISA. Similarly, Xu et al50 demonstrated the suppression of PI3K/AKT signaling in liver cancer cells by a polysaccharide-protein complex isolated from Pleurotus pilmonarius (PP). Their results indicated that PP not only significantly reduced in vitro cancer cell proliferation and invasion but also inhibited tumor growth in xenograft BALB/c nude mice when administered orally and intraperitoneally. As per their findings, these inhibitory effects are because of the inhibition of the autocrine VEGF-induced PI3K/AKT signaling pathway. In addition to this, when PP was used together with cisplatin, it resulted in the enhancement of liver cancer cell sensitization to cisplatin. It has been demonstrated before, that on treatment with cisplatin, BAD is phosphorylated at Ser 136 via AKT and this phosphorylation is needed for cell viability. When this AKT node was inhibited in ovarian cancer cells, they become sensitive to cisplatin.58 Most probably, the same mechanism might be involved in the case of liver cancer cells: On inhibition of PI3K/AKT signaling by PP, they become more sensitive to cisplatin.

Antroquinonol, a derivative of natural ubiquinone, has been reported to inhibit the phosphorylation of Akt and mTOR at Ser473 and Ser2448, respectively in human pancreatic carcinoma cells.41 This compound also resulted in the induction of cross-talk between apoptosis, autophagy, and senescence, explained by the upregulation of p21Waf1/Cip1 and K-ras.

In another study, polysaccharide-induced apoptosis was demonstrated in human hepatoma cells, and polysaccharide was extracted from 3 medicinal mushrooms P linteus (PL), Ganoderma lucidum (GL), and Auricularia auricula (AA).27 Three of the polysaccharides resulted in enhanced expression of p27Kip in HepG2 cells and, on the other hand, suppressed the activity of cyclin D1/CDK4 and/or cyclin E/CDK2. AKT, one of the key nodes of PI3K/AKT pathway, once phosphorylated and activated results in the activation of downstream genes, that is, GSK3, FOXOs, thereby regulating key cellular processes involved in protein synthesis, cell survival, proliferation, and metabolism. However, both phosphorylation events (at Thr308 and Ser473) are required for full activation of AKT. Surprisingly, polysaccharides from these medicinal mushrooms suppressed AKT activity through the inhibition of AKT phosphorylation at Thr308 and/or Ser473. In addition, they also resulted in the activation of mitochondrial-mediated apoptosis and G1 and/or S phase arrest in HepG2 cells. After a few years, the same research group, in one of their recent studies59 on proteomic analysis of HepG2 cells treated with mushroom polysaccharide, revealed the differential expression of 59 proteins identified through MALDI-TOF-MS, where DJ-1 and 14-3-3 were the key proteins identified as a result of Protein-Protein interaction analysis. DJ-1 is a novel oncogene product that has been reported to negatively regulate the PTEN activity.60 Therefore, lower expression of DJ-1 can decrease the phosphorylation of AKT, whereas a higher expression of DJ-1 can increase AKT phosphorylation and cell survival.61 Interestingly, in this study, DJ- 1 protein was down-regulated in the treated HepG2 cells. One of the recent studies demonstrated the effect of antroquinonol (ANQ) on cancer stem cell-like properties in colon cancer.42 The authors demonstrated the interaction of ANQ with PI3K, identified by molecular docking, and further showed the suppression of PI3K/AKT/β-catenin signaling in colon cancer, which is crucial for regulating the expression of pluripotent genes.

The PI3K/AKT pathway has already been reported as one of the signaling pathways that are involved in acquiring chemotherapeutic drug resistance in cancer.62 The aforementioned studies suggest that compounds from mushrooms could lead to the restoration of drug sensitivity. Second, these compounds can not only be used as a single agent but may also be useful as adjuvant therapeutic agents in the treatment of cancer.

Wnt/β-Catenin Pathway as a Therapeutic Target in Cancer

Wnts are the secreted glycoproteins that are involved in several critical cellular processes.63 They regulate many of the signaling pathways by β-catenin dependent or independent mechanisms.64 The occurrence of a high frequency of aberrations in the Wnt signaling pathway in many different cancers highlights the importance of this pathway to carcinogenesis. APC, CTNNB1, AXIN1, FAM123B, and TCF7L2 are the key nodes of Wnt signaling that harbor somatic mutations associated with some of the common human cancers.64 The development and progression of sporadic colorectal cancer (CRC) and familial adenomatous polyposis (FAP)-associated disease are believed to be because of the mutations in APC (adenomatous polyposis coli) gene.65 In addition to this, two inactivating mutations often occur in the APC gene, one leading to allelic loss and other resulting in a truncated protein.66,67

Most mutations noted in the Wnt pathway results in the hyperactivation of this signaling pathway. Although mutations observed in genes that cause the inactivation of Wnt-CTNNB1 signaling has been identified in other disorders but not in cancer.64 Depending on the stage and type of cancer, Wnt-CTNNB1 signaling pathway can either promote or inhibit the tumor initiation, growth, metastasis and drug resistance. Herein, because of the limited scope of this review, we will only address the oncogenic role of Wnt-CTNNB1 signaling and its inhibition by different mushroom originated compounds.

As mentioned before mutated APC gene is common in colon cancer, so aberrant activation of Wnt-CTNNB1 signaling pathway results in its progression. Many researchers have attemptd to target cancer using mushroom components; one such study48 conducted on colon cancer in 2011 highlighted the antitumor property of polysaccharides extracted from P linteus (PL) mushroom. Researchers showed that this polysaccharide is capable of suppressing tumor growth, invasion and angiogenesis by down-regulating certain downstream genes (cyclin D1, TCF/LEF) of the Wnt signaling pathway in SW480 human colon cancer cells. In vivo anticancer potential of this mushroom extract was determined by the heterotrophic mouse model. A recent study done on colorectal cancer cell lines47 highlights the anti-proliferative activity of ergosterol peroxide isolated from Chaga mushroom (Inonotus obliquus). According to the findings, ergosterol peroxide inhibited the nuclear β-catenin level, which resulted in the down-regulation of downstream genes. In another recent study,43 investigators claimed much better effect of 4-acetylantroquinonol (4-AAQB), closely related to a well-known antroquinonol, in suppressing colorectal cancer and cancer stem-like phenotype. As per their findings, 4-AAQB negatively regulates the genes responsible for the oncogenic effect and the signal transduction pathways that are involved in stemness.

Targeting Wnt- CTNNB-1 signaling by natural compounds is not only limited to colon cancer: the antitumor activity of the mushroom component against human meningioma cells has also been reported.68 Ganoderic acid A and ganoderic Acid DM suppress the expression of Wnt 5α/β and β-catenin and also cause the phosphorylation of GSK3β, which results in the proteaosomal degradation of β-catenin in the cytoplasm. On reduced β–catenin activity expression of β-catenin target genes (c-myc, cyclin D1, and VEGF) also reduces, thus giving anti-tumorigenic effect in meningioma cells. Das et al have also included normal human neuron cells in their study and fortunately, all the compounds, including ganoderic acid A and ganoderic acid DM have no toxic effects on normal cells. These compounds might, therefore, be potential candidates for the pharmaceutical treatment of human meningiomas.

Hyperactivation of the Wnt-CTNNB1 pathway because of the mutations in APC and AXIN1 limits the potential molecular targets for pathway modulation because factors acting upstream of the destruction complex are no longer necessary for the activation of a pathway. So, the prime focus of researchers is disrupting the interaction of CTNNB1 with TCF7L2, thus inhibiting the CTNNB1 transcription. In the above-given examples, on treatment with mushroom compounds, reduction in the CTNNB1 level and downstream genes is observed, indicating that mushroom extracts can be used to inhibit the CTNNB1-mediated gene transcription. However, targeting the Wnt-CTNNB1 pathway directly is not simple because of the redundancy of many components of the pathway, so further studies on the interaction of mushroom compounds and nodes of this pathway are needed to overcome the challenges in targeting Wnt-CTNNB1 signaling.

Targeting the NF-κB Signaling Pathway in Cancer

The NF-κB signaling pathway is a signal transduction pathway that on activation controls the expression of hundreds of genes. In mammals, the NF-κB family consists of five related transcription factors, p50, p52, RelA (p65), c-Rel, and RelB.69 Activation of the NF-κB transcription factor may take place via the classical (canonical) pathway or through the alternative (noncanonical) pathway, where the common regulatory step is the activation of IκB kinase consisting of catalytic and regulatory subunits.70,71 NF-κB is well known for its role in immunity, inflammation, and oncogenesis.72,73 As far as the alteration in this signaling pathway is concerned, mutations in the core signaling components have been reported to have associations with human diseases.74

Numerous lines of evidence highlight the antitumor effect of edible and medicinal mushrooms through targeting the NF-κB signaling pathway. Cordyceps militaris is one of the traditional medicinal mushrooms, whose major bioactive component cordycepin (3-deoxyadenosine) has been reported to have both pro- and anti-apoptotic activity. In 2012, Kadomatsu et al44 investigated the effect of cordycepin on cell survival on exposure to TNF-α. They found that, on treatment with a sub-lethal dose of cordycepin, cells become sensitive to TNF-α mediated apoptosis, which takes place via suppression of pro-survival NF-κB. As far as the detailed molecular mechanism is concerned, mTORC1 activation by eIF2α was responsible for the suppression of NF-κB, a key pro-survival component in TNF-α signaling.

In another study, the molecular mechanism behind the antitumor effect of sulfated polysaccharide obtained from Grifola frondosa (S-GFB) on HepG2 was explored.46 The results indicated a typical change in cell morphology, S phase arrest, notch1 expression inhibition, degradation of IκB-α, translocation of NF-κB from the cytoplasm to nucleus and activation of caspase-3 and 8. In short, S-GFB resulted in apoptosis of HepG2 cells through the notch1/NF-B/p65-mediated caspase pathway.

Panepoxydone (PP), isolated from Lentinus crinitus was found to be a natural NF-κB inhibitor.75 Genetic alterations do exist in NF-κB signaling, and breast cancer is one of those cancers in which NF-κB is overexpressed, suggesting it as a therapeutic target. Arora et al isolated PP from L crinitus and reported it as an inhibitor of NF-κB. The antitumor effect of PP was reported in MCF-7 and triple negative breast cancer cell lines. According to their investigation, PP resulted in significant cytotoxicity, increased apoptosis, decreased invasion and migration in all tested cell lines. PP upregulated Bax and cleaved PARP, inhibited the phosphorylation of NF-κB with cytoplasmic accumulation and on other hand downregulated Bcl-2, survivin, cyclin D1, caspase-3, and FOXM1.30

Kong et al in 2014 37 demonstrated the immunostimulating effect of polysaccharide (WPOP-N1) isolated from Pleurotus ostreatus, a famous edible mushroom. They highlighted the antitumor activity of (WPOP-N1) in an in vivo model and also suggested its role in the activation of macrophages through NF-κB signaling pathway.

Conclusion

From the evidence presented in this review, we can anticipate the molecular mechanism of different mushroom originated compounds, where they exhibit anticancer potential through modulating different nodes of PI3K/Akt, Wnt-CTNNB1, and NF-κB signaling pathways and thus affecting a variety of cellular processes, for example, by induction of apoptosis, cell cycle arrest, increasing the antioxidant capacity of host, and interfering with tumor angiogenesis. Because a plethora of genes/proteins associated with signal transduction pathways are deregulated during the development and progression of cancer, they may thus serve as targets to different components of mushrooms depending on their location. As indicated, some of the mushroom compounds target a single node of a signaling pathway and some have multiple targets in the same and/or different signaling pathway. This property highlights the vertical or horizontal blockade of the signal transduction pathways in cancer by these compounds. In addition, studies presented here also highlight the role of mushroom components in restoring drug sensitivity and their implications as single and adjuvant therapeutic agents. However, a choice of which mushroom compounds to select for the treatment of cancer depends on multiple factors and needs further investigation.

Extensive studies have demonstrated the anticancer potential of different mushrooms in preclinical models however, they still have significant limitations. In many of the studies, the anticancer potential of mushroom extracts has been demonstrated instead of specific active components, and where the action of specific active component(s) originated from mushroom has been reported, it lacks the interaction of mushroom components with the reported nodes (proteins/enzymes) of the signaling pathways. Most of the investigators have not determined the IC50 values of the bioactive component used against cancer, and many preclinical studies are in their initial phases. Future studies are needed to explore the structure of the bioactive components, their interaction with different nodes of signaling pathways, and their preclinical effects on more complicated animal models that can closely mimic the human tumors in terms of genetic profile and tumor microenvironment.

This review provides the possible molecular insight of the action of mushroom origin compounds and it would be helpful in designing anticancer arsenals from mushroom for combating cancer.

Acknowledgments

We thank Richardson Patrick Joseph for editorial assistance and Sadia Kanwal and Abdullah Faqeer Mohammad for critically reading the manuscript.

Footnotes

Authors’ Note: Thomson Patrick Joseph and Warren Chanda contributed equally to the paper.

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by Chinese Government Scholarship (CSC No.: 2014GXY960).

References

  • 1. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin. 2017;67:7-30. [DOI] [PubMed] [Google Scholar]
  • 2. Saxena A, Schneider BJ, Christos PJ, Audibert LF, Cagney JM, Scheff RJ. Treatment of recurrent and platinum-refractory stage IV non-small cell lung cancer with nanoparticle albumin-bound paclitaxel (nab-paclitaxel) as a single agent. Med Oncol. 2016;33:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Li F, Zhao C, Wang L. Molecular-targeted agents combination therapy for cancer: developments and potentials. Int J Cancer. 2014;134:1257-1269. [DOI] [PubMed] [Google Scholar]
  • 4. Ozsaran Z, Yalman D, Yürüt V, et al. Radiochemotherapy for patients with locally advanced cervical cancer: early results. Eur J Gynaecol Oncol. 2003;24:191-194. [PubMed] [Google Scholar]
  • 5. Liao Z, Cox JD, Komaki R. Radiochemotherapy of esophageal cancer. J Thorac Oncol. 2007;2:553-568. [DOI] [PubMed] [Google Scholar]
  • 6. Schiffmann L, Wedermann N, Gock M, et al. Intensified neoadjuvant radiochemotherapy for rectal cancer enhances surgical complications. BMC Surg. 2013;13:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Thomas H, Coley HM. Overcoming multidrug resistance in cancer: an update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control.2003;10:159-165. [DOI] [PubMed] [Google Scholar]
  • 8. Chen S, Dai Y, Harada H, Dent P, Grant S. Mcl-1 down-regulation potentiates ABT-737 lethality by cooperatively inducing Bak activation and Bax translocation. Cancer Res. 2007;67:782-791. [DOI] [PubMed] [Google Scholar]
  • 9. Kirschner K, Melton DW. Multiple roles of the ERCC1-XPF endonuclease in DNA repair and resistance to anticancer drugs. Anticancer Res. 2010;30:3223-3232. [PubMed] [Google Scholar]
  • 10. Shervington A, Lu C. Expression of multidrug resistance genes in normal and cancer stem cells. Cancer Invest. 2008;26:535-542. [DOI] [PubMed] [Google Scholar]
  • 11. Chen Y, Takita J, Choi YL, et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature. 2008;455:971-974. [DOI] [PubMed] [Google Scholar]
  • 12. Camidge DR, Bang YJ, Kwak EL, et al. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol. 2012;13:1011-1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Recupero D, Daniele L, Marchiò C, et al. Spontaneous and pronase-induced HER2 truncation increases the trastuzumab binding capacity of breast cancer tissues and cell lines. J Pathol. 2013;229:390-399. [DOI] [PubMed] [Google Scholar]
  • 14. Brenner GM, Stevens CW. Antineoplastic drugs. In: Pharmacology. 3rd ed. Philadelphia, PA: Saunders/Elseiver; 2010:493-511. [Google Scholar]
  • 15. Harvey AL. Natural products in drug discovery. Drug Discov Today. 2008;13:894-901. [DOI] [PubMed] [Google Scholar]
  • 16. Dias DA, Urban S, Roessner U. A historical overview of natural products in drug discovery. Metabolites. 2012;2:303-336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Arcamone F. Anthracyclines. In: Cragg GM, Kingston DGI, Newman DJ, eds. Anticancer Agents From Natural Products. Boca Raton, FL: CRC Press; 2012:383-405. [Google Scholar]
  • 18. Hecht SM. Bleomycin group antitumor agents. In: Cragg GM, Kingston DGI, Newman DJ, eds. Antitcancer Agents From Natural Products. 2nd ed. Boca Raton, FL: CRC Press; 2012:451-478. [Google Scholar]
  • 19. Remers W. The mitomycins. In: Cragg GM, Kingston DGI, Newman DJ, eds. Antitcancer Agents From Natural Products. 1st ed Boca Raton, FL: CRC Press; 2005:475-497. [Google Scholar]
  • 20. Hamann PU, Upeslacis J, Borders DB. Enediynes. In: Cragg GM, Kingston DGI, Newman DJ, eds. Antitcancer Agents From Natural Products. 2nd ed. Boca Raton, FL: CRC Press; 2012:575-619. [Google Scholar]
  • 21. Wasser SP. Current findings, future trends, and unsolved problems in studies of medicinal mushrooms. Appl Microbiol Biotechnol. 2011;89:1323-1332. [DOI] [PubMed] [Google Scholar]
  • 22. Quang DN, Hashimoto T, Asakawa Y. Inedible mushrooms: a good source of biologically active substances. Chem Rec. 2006;6:79-99. [DOI] [PubMed] [Google Scholar]
  • 23. Ferreira IC, Vaz JA, Vasconcelos MH, Martins A. Compounds from wild mushrooms with antitumor potential. Anticancer Agents Med Chem. 2010;10:424-436. [DOI] [PubMed] [Google Scholar]
  • 24. Wasser SP. Medicinal mushroom science: current perspectives, advances, evidences, and challenges. Biomed J. 2014;37:345-356. [DOI] [PubMed] [Google Scholar]
  • 25. Jin S, Pang RP, Shen JN, Huang G, Wang J, Zhou JG. Grifolin induces apoptosis via inhibition of PI3K/AKT signalling pathway in human osteosarcoma cells. Apoptosis. 2007;12:1317-1326. [DOI] [PubMed] [Google Scholar]
  • 26. Chen Y, Peng GF, Han XZ, Wang W, Zhang GQ, Li X. Apoptosis prediction via inhibition of AKT signaling pathway by neogrifolin. Int J Clin Exp Pathol. 2015;8:1154-1164. [PMC free article] [PubMed] [Google Scholar]
  • 27. 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. [DOI] [PubMed] [Google Scholar]
  • 28. Lin CH, Chang CY, Lee KR, et al. Cold-water extracts of Grifola frondosa and its purified active fraction inhibit hepatocellular carcinoma in vitro and in vivo. Exp Biol Med (Maywood). 2016;241:1374-1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Soares R, Meireles M, Rocha A, et al. Maitake (D fraction) mushroom extract induces apoptosis in breast cancer cells by BAK-1 gene activation. J Med Food. 2011;14:563-572. [DOI] [PubMed] [Google Scholar]
  • 30. Arora R, Yates C, Gary BD, et al. Panepoxydone targets NF-κB and FOXM1 to inhibit proliferation, induce apoptosis and reverse epithelial to mesenchymal transition in breast cancer. PLoS One. 2014;9:e98370. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 31. Liu B, Zhong M, Lun Y, et al. A novel apoptosis correlated molecule: expression and characterization of protein Latcripin-1 from Lentinula edodes C(91-3). Int J Mol Sci. 2012;13:6246-6265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Ann XH, Lun YZ, Zhang W, et al. Expression and characterization of protein Latcripin-3, an antioxidant and antitumor molecule from Lentinula edodes C91-3. Asian Pac J Cancer Prev. 2014;15:5055-5061. [DOI] [PubMed] [Google Scholar]
  • 33. Wang J, Zhong M, Liu B, et al. Expression and functional analysis of novel molecule—Latcripin-13 domain from Lentinula edodes C91-3 produced in prokaryotic expression system. Gene. 2015;555:469-475. [DOI] [PubMed] [Google Scholar]
  • 34. Tian L, Wang X, Li X, et al. In vitro antitumor activity of Latcripin-15 regulator of chromosome condensation 1 domain protein. Oncol Lett. 2016;12:3153-3160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. You RX, Liu JY, Li SJ, Wang L, Wang KP, Zhang Y. Alkali-soluble polysaccharide, isolated from Lentinus edodes, induces apoptosis and G2/M cell cycle arrest in H22 cells through microtubule depolymerization. Phytother Res. 2014;28:1837-1845. [DOI] [PubMed] [Google Scholar]
  • 36. Wang W, Chen K, Liu Q, et al. Suppression of tumor growth by Pleurotus ferulae ethanol extract through induction of cell apoptosis, and inhibition of cell proliferation and migration. PLoS One. 2014;9:e102673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kong F, Li FE, He Z, et al. Anti-tumor and macrophage activation induced by alkali-extracted polysaccharide from Pleurotus ostreatus. Int J Biol Macromol. 2014;69:561-566. [DOI] [PubMed] [Google Scholar]
  • 38. Mondal A, Banerjee D, Majumder R, Maity TK, Khowala S. Evaluation of in vitro antioxidant, anticancer and in vivo antitumour activity of Termitomyces clypeatus MTCC 5091. Pharm Biol. 2016;54:2536-2546. [DOI] [PubMed] [Google Scholar]
  • 39. Li X, Wu Q, Xie Y, et al. Ergosterol purified from medicinal mushroom Amauroderma rude inhibits cancer growth in vitro and in vivo by up-regulating multiple tumor suppressors. Oncotarget. 2015;6:17832-17846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Chang JS, Kuo HP, Chang KL, Kong ZL. Apoptosis of hepatocellular carcinoma cells induced by nanoencapsulated polysaccharides extracted from antrodia camphorata. PLoS One. 2015;10:e0136782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Yu CC, Chiang PC, Lu PH, et al. Antroquinonol, a natural ubiquinone derivative, induces a cross talk between apoptosis, autophagy and senescence in human pancreatic carcinoma cells. J Nutr Biochem. 2012;23:900-907. [DOI] [PubMed] [Google Scholar]
  • 42. Lin HC, Lin MH, Liao JH, et al. 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. [DOI] [PubMed] [Google Scholar]
  • 43. Chang TC, Yeh CT, Adebayo BO, et al. 4-Acetylantroquinonol B inhibits colorectal cancer tumorigenesis and suppresses cancer stem-like phenotype. Toxicol Appl Pharmacol. 2015;288:258-268. [DOI] [PubMed] [Google Scholar]
  • 44. Kadomatsu M, Nakajima S, Kato H, et al. Cordycepin as a sensitizer to tumour necrosis factor (TNF)- α-induced apoptosis through eukaryotic translation initiation factor 2α (eIF2 α)- and mammalian target of rapamycin complex 1 (mTORC1)-mediated inhibition of nuclear factor (NF)-κB. Clin Exp Immunol. 2012;168:325-332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Kim SH, Jakhar R, Kang SC. Apoptotic properties of polysaccharide isolated from fruiting bodies of medicinal mushroom Fomes fomentarius in human lung carcinoma cell line. Saudi J Biol Sci. 2015;22:484-490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Wang CL, Meng M, Liu SB, Wang LR, Hou LH, Cao XH. A chemically sulfated polysaccharide from Grifola frondos induces HepG2 cell apoptosis by notch1-NF-κB pathway. Carbohydr Polym. 2013;95:282-287. [DOI] [PubMed] [Google Scholar]
  • 47. Kang JH, Jang JE, Mishra SK, et al. 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. [DOI] [PubMed] [Google Scholar]
  • 48. Song KS, Li G, Kim JS, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Huang GJ, Yang CM, Chang YS, et al. 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. [DOI] [PubMed] [Google Scholar]
  • 50. Xu W, Huang JJ, Cheung PC. Extract of Pleurotus pulmonarius suppresses liver cancer development and progression through inhibition of VEGF-induced PI3K/AKT signaling pathway. PLoS One. 2012;7:e34406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 2009;8:627-644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Bader AG, Kang S, Zhao L, Vogt PK. Oncogenic PI3K deregulates transcription and translation. Nat Rev Cancer. 2005;5:921-929. [DOI] [PubMed] [Google Scholar]
  • 53. Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet. 2006;7:606-619. [DOI] [PubMed] [Google Scholar]
  • 54. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. [DOI] [PubMed] [Google Scholar]
  • 55. Wood LD, Parsons DW, Jones S, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318:1108-1113. [DOI] [PubMed] [Google Scholar]
  • 56. Ding L, Getz G, Wheeler DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069-1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Li YG, Ji DF, Zhong S, Zhu JX, Chen S, Hu GY. Anti-tumor effects of proteoglycan from Phellinus linteus by immunomodulating and inhibiting Reg IV/EGFR/Akt signaling pathway in colorectal carcinoma. Int J Biol Macromol. 2011;48:511-517. [DOI] [PubMed] [Google Scholar]
  • 58. Hayakawa J, Ohmichi M, Kurachi H, et al. Inhibition of BAD phosphorylation either at serine 112 via extracellular signal-regulated protein kinase cascade or at serine 136 via Akt cascade sensitizes human ovarian cancer cells to cisplatin. Cancer Res. 2000;60:5988-5994. [PubMed] [Google Scholar]
  • 59. Chai Y, Wang G, Fan L, Zhao M. A proteomic analysis of mushroom polysaccharide-treated HepG2 cells. Sci Rep. 2016;6:23565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Wang B, Qin H, Wang Y, et al. Effect of DJ-1 overexpression on the proliferation, apoptosis, invasion and migration of laryngeal squamous cell carcinoma SNU-46 cells through PI3K/AKT/mTOR. Oncol Rep. 2014;32:1108-1116. [DOI] [PubMed] [Google Scholar]
  • 61. Sitaram RT, Cairney CJ, Grabowski P, et al. The PTEN regulator DJ-1 is associated with hTERT expression in clear cell renal cell carcinoma. Int J Cancer. 2009;125:783-790. [DOI] [PubMed] [Google Scholar]
  • 62. McCubrey JA, Steelman LS, Kempf CR, et al. Therapeutic resistance resulting from mutations in Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR signaling pathways. J Cell Physiol. 2011;226:2762-2781. [DOI] [PubMed] [Google Scholar]
  • 63. Papkoff J, Brown AM, Varmus HE. The int-1 proto-oncogene products are glycoproteins that appear to enter the secretory pathway. Mol Cell Biol. 1987;7:3978-3984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Anastas JN, Moon RT. WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer. 2013;13:11-26. [DOI] [PubMed] [Google Scholar]
  • 65. Powell SM, Zilz N, Beazer-Barclay Y, et al. APC mutations occur early during colorectal tumorigenesis. Nature. 1992;359:235-237. [DOI] [PubMed] [Google Scholar]
  • 66. Lamlum H, Papadopoulou A, Ilyas M, et al. APC mutations are sufficient for the growth of early colorectal adenomas. Proc Natl Acad Sci U S A. 2000;97:2225-2228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Cheadle JP, Krawczak M, Thomas MW, et al. Different combinations of biallelic APC mutation confer different growth advantages in colorectal tumours. Cancer Res. 2002;62:363-366. [PubMed] [Google Scholar]
  • 68. Das A, Miller R, Lee P, et al. A novel component from citrus, ginger, and mushroom family exhibits antitumor activity on human meningioma cells through suppressing the Wnt/β-catenin signaling pathway. Tumour Biol. 2015;36:7027-7034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Hoffmann A, Natoli G, Ghosh G. Transcriptional regulation via the NF-κB signaling module. Oncogene. 2006;25:6706-6716. [DOI] [PubMed] [Google Scholar]
  • 70. Gilmore TD. Introduction to NF-κB: players, pathways, perspectives. Oncogene. 2006;25:6680-6684. [DOI] [PubMed] [Google Scholar]
  • 71. Scheidereit C. IkappaB kinase complexes: gateways to NF-κB activation and transcription. Oncogene. 2006;25:6685-6705. [DOI] [PubMed] [Google Scholar]
  • 72. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883-899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. DiDonato JA, Mercurio F, Karin M. NF-κB and the link between inflammation and cancer. Immunol Rev. 2012;246:379-400. [DOI] [PubMed] [Google Scholar]
  • 74. Courtois G, Gilmore TD. Mutations in the NF-κB signaling pathway: implications for human disease. Oncogene. 2006;25:6831-6843. [DOI] [PubMed] [Google Scholar]
  • 75. Erkel G, Wisser G, Anke T. Influence of the fungal NF-κB inhibitor panepoxydone on inflammatory gene expression in MonoMac6 cells. Int Immunopharmacol. 2007;7:612-624. [DOI] [PubMed] [Google Scholar]

Articles from Integrative Cancer Therapies are provided here courtesy of SAGE Publications

RESOURCES