Next Article in Journal
Both Matricaria chamomilla and Metformin Extract Improved the Function and Histological Structure of Thyroid Gland in Polycystic Ovary Syndrome Rats through Antioxidant Mechanism
Next Article in Special Issue
Phytochemicals and Gastrointestinal Cancer: Cellular Mechanisms and Effects to Change Cancer Progression
Previous Article in Journal
Distinct Peculiarities of In Planta Synthesis of Isoprenoid and Aromatic Cytokinins
Previous Article in Special Issue
Combination Therapy with Vitamin C Could Eradicate Cancer Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anticancer Potential of Lichens’ Secondary Metabolites

1
Department of Pharmacology, Faculty of Medicine, P.J. Šafárik University in Košice, 040 01 Košice, Slovakia
2
Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
3
Department of Medical Biology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
4
Department of Experimental Carcinogenesis, Biomedical Centre Martin, Division of Oncology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
5
Weill Cornell Medicine in Qatar, Qatar Foundation-Education City, Doha 24144, Qatar
6
Department of Medical Biology, Faculty of Medicine, P.J. Šafárik University in Košice, 040 01 Košice, Slovakia
*
Author to whom correspondence should be addressed.
Biomolecules 2020, 10(1), 87; https://doi.org/10.3390/biom10010087
Submission received: 25 November 2019 / Revised: 31 December 2019 / Accepted: 3 January 2020 / Published: 5 January 2020

Abstract

:
Lichens produce different classes of phenolic compounds, including anthraquinones, xanthones, dibenzofuranes, depsides and depsidones. Many of them have revealed effective biological activities such as antioxidant, antiviral, antibiotics, antifungal, and anticancer. Although no clinical study has been conducted yet, there are number of in vitro and in vivo studies demonstrating anticancer effects of lichen metabolites. The main goal of our work was to review most recent published papers dealing with anticancer activities of secondary metabolites of lichens and point out to their perspective clinical use in cancer management.

1. Introduction

Despite noticeable progress in the anticancer therapy, cancer disease remains one of the major health problems worldwide [1,2]. Plant natural substances represent the rich source of active molecules that can find many applications in the field of biology, pharmacy, and medicine including oncology. Carcinogenesis is related to an imbalance between proliferation and apoptosis, inappropriate activity of epigenetic and transcription factors, suppression of anti-oxidative defense mechanisms and increase in angiogenesis, that leads to the consequent raising of invasiveness and formation of metastases. All these mechanisms of carcinogenesis have been well-documented as direct molecular targets for plant secondary metabolites—phytochemicals [3,4,5,6,7,8,9]. Based on many preclinical and clinical studies, phytochemicals demonstrate excellent potential how to improve the clinical state in cancer patients. The proper administration of plant natural substances may represent a significant approach how to complete the conventional management of the disease in terms of either chemoprevention or treatment. Numerous biomolecules derived from plants are also capable of synergizing with chemo- and radiotherapy. Such a combination can potentially lead to the increase of therapeutic effects and/or can reduce the side effects because lower doses of conventional therapeutics are needed [10,11,12,13,14,15].
Lichens are symbiotic organisms producing various secondary metabolites. The considerable chemical diversity of lichen secondary metabolites makes them a powerful natural source of pharmaceutical reagents that may be used in the medical practice [16]. Secondary metabolites present in lichens include aliphatic, cycloaliphatic, aromatic, and terpenic compounds, which demonstrate significant biological and pharmacological effects including those of anti-inflammatory, antiviral, antibacterial, analgesic, antipyretic, anti-proliferative, and cytotoxic [17]. These compounds have demonstrated also antineoplastic activities in preclinical research in which revealed significant effects against cancer cells [18,19,20,21,22].
The aim of this paper is to provide up-to-date knowledge about anticancer effects of lichen extracts or their secondary metabolites on different signal pathways involved in cancer and/or carcinogenesis. We discuss here the potential clinical utility of lichen secondary metabolites or natural mixtures of phytochemicals regarding the prevention and therapy of cancer. Our data were received from the English-language biomedical literature by use of “lichens” or “secondary metabolites” or “cancer” or “cell lines” and “animal models” keyword or medical subject headings (MeSH) term for searches in the PubMed bibliographic database. We have used the most recent scientific papers from the years 2015–2019.

2. Molecular Mechanisms of Lichen Anticancer Potential

Lichens as well as lichen secondary metabolites, also known as lichen acids, exert important abilities in the protection against carcinogenesis [23], which is due to their antioxidant, cytotoxic, pro-apoptotic, anti-proliferative, anti-migrative, anti-invasive, and overall anti-tumorigenic abilities [21,23,24,25].
Antioxidant action of most lichens is associated with the phenolic compounds, which exhibit high free radical scavenging ability [21,23]. Lichens prevent mutagenesis and/or carcinogenesis by inhibiting oxidation of cellular macromolecules [24]. Consequently, the protective effects of lichens or their metabolites against oxidative damage can be determined through the monitoring of oxidative stress markers such as superoxide dismutase (SOD) or malodialdehyde (MDA) [26].
The cytotoxic activity of lichens was observed in different cancer cell lines and we have noticed that the cytotoxic abilities of lichens in cancer cells are higher than in non-cancer cells [21,23]. First of all, the strong cytotoxic capacity of lichens against cancer cells is mediated through the processes including apoptosis, necrosis or autophagy together with the cell cycle arrest at G2/M, S, or G0/G1 phases [27]. Lichens act also as regulators of the cell cycle through the various mechanisms, such as those associated with cyclin-dependent kinases (CDK4, CDK6) or cyclin D1 [28].
In regard to programmed cell death, lichens act as activators of apoptosis in various cancer cells [21,26] through the modulation of gene expression of products related to apoptosis such as caspases, p53, p38, or anti-/pro-apoptotic proteins of Bcl-2 family [29]. Induction of apoptosis by lichens might be associated also with an increase of cleaved PARP, a stress response protein repairing damaged DNA and regulating chromatin structure [30], with inactivation of the mammalian target of rapamycin (mTOR) or activation of c-Jun N-terminal kinase (JNK) signaling [27]. Anti-proliferative effects of lichens can be modulated through the regulation of other signaling pathways such as ERK1/2 and AKT [31] or proliferation protein marker Ki-67 [32]. Interestingly, anticancer potential of lichens can also be maintained by modulation of pathways associated with the cancer invasiveness such as c-Met, which acts as mesenchymal epithelial transition factor regulating PI3K/Akt/mTOR, Paxillin/Rac-1, and STATs signaling cascades [33]. Despite regulation of STAT3 activity, [20,34] lichens target β-catenin or its downstream effectors that consequently lead to the modulation of Wnt/β-catenin target genes including the cell cycle regulating genes (c-myc, cyclin D1) as well as genes associated with cell migration (MMP7), apoptosis (BIRC5) or other regulators such as Axin2 [20,26]. Additionally, regulation of c-Jun and c-fos, members of the AP-1 family of critical regulators of gene expression and reduction of KITENIN-mediated AP-1 activity are also associated with anticancer mechanisms of lichens [20].
Indeed, anti-invasive and anti-migrative potential of lichens is related to the modulation of various signaling molecules such as members of Ras superfamily of small GTPases (RhoA, Rac1, Cdc42, and KITENIN) which play a significant role in tumor development and progression. In this regard, metastasis-related genes including CAPN1, CDC42, CFL1, IGF1, or WASF1 as well as epithelial–mesenchymal markers (Twist, Snail, Snug) might also be targets of lichens. On the other hand, anticancer effect of lichen realized through the angiogenesis inhibitory activities are related to the suppression of endothelial tube formation [35] or vascular endothelial growth factor receptor (VEGFR)-2-mediated Akt and extracellular signal-regulated kinase (ERK) signaling [29]. Current results showed that anticancer effects of lichens are also associated with the modulation of inflammatory responses via TNF-α, IL-1β, IL-6, and TGF-β1 [36] and with targeting of microRNA molecules [37].
Overview of cancer-associated molecular mechanisms modulated by lichens is shown in Figure 1. Table 1 shows secondary metabolites of lichens, their origin, chemical structure and also their beneficial activities including anticancer one.

3. Anti-Neoplastic Effect of Lichens in Preclinical Research

3.1. In Vitro Evaluation of Anticancer Efficacy of Isolated Lichen Compounds

Pro-apoptotic abilities of usnic acid (UA), a secondary lichen metabolite isolated from Usnea diffracta, was evaluated in human colorectal adenocarcinoma (CaCo2), human rhabdomyosarcoma (RD), human cervical carcinoma (Hep2C), human hepatocellular carcinoma (HepG2), mouse fibrosarcoma (Wehi), as well as in nonmalignant African green monkey kidney (Vero) and mouse subcutaneous connective tissue (L929) cell lines. Both natural and commercial UA had similar cytotoxicity on cancer cells but no significant effect in non-malignant L929 and Vero cells. Indeed, UA increased the expression of Bax and decreased the expression of Bcl-2 and p53 genes in cancer but not in non-malignant cells [29]. Additionally, the concentration of UA at 8 μM, inhibited SCF-mediated migration of human colorectal cancer (HCT116, LS174 c-KIT+) cells. UA induced an inhibition of mTOR activity in HCT116 cells thereby activated suppression of protein kinase C alpha type (PKC-A) and induced an autophagy followed by the degradation and transcriptional inhibition of mast/stem cell growth factor receptor Kit (c-KIT) as well as to an increase in LDH release [68]. Another study of UA demonstrated its ability to inhibit the proliferation of human gastric carcinoma (BGC823, SGC7901) cells through the G0/G1 and G2/M cell cycle arrest, respectively while BGC823 cells were more sensitive. Both apoptosis and autophagy of BGC823 and SGC7901 cells with increased ratio of Bax/Bcl-2 and increased activity of caspase-3 and PARP cleavage after UA therapy were identified [69]. Anti-proliferative effects of UA were also evaluated in human breast cancer cell lines in which 67, 15, and 8 miRNAs in MDA-MB-231, BT-474, and MCF-7 cells, respectively after UA therapy were detected. Indeed, enrichment analysis revealed different groups of miRNA with targets as follows: Apoptosis, Hedgehog, TGF-β, and MAPK pathways identified as prominent [37]. Pro-apoptotic abilities of UA were demonstrated through the G0/G1 and G2/M cell cycle arrest in both hepatocellular carcinoma HepG2 and SNU-449 cells [27]. Similarly, an application of UA in lung cancer cells (A549) led to the decrease of cells, induced apoptosis and inhibited cell proliferation, which was accompanied by G0/G1 cell cycle arrest, decreased expression of CDK4, CDK6, and cyclin D1 and increased expression of CDK inhibitor (CDKI) p21/cip1 protein. Consequently, the apoptotic effect of UA co-occurred with enhanced PARP cleavage [28]. The anticancer effects of UA isolated from Cladonia arbuscula were demonstrated through the formation of autophagosome as a consequence of H+ shuttling in mitochondria and lysosomes in human breast cancer (MCF-7, T47D) as well as in human pancreatic cancer cell lines (Capan-2) with the decrease in ATP, activation of AMP-kinase and the detection of cellular stress markers [70]. Similarly, UA induced an apoptosis of MCF-7 cells through the generation of ROS and mitochondrial/caspase pathway. On the contrary, N-acetylcysteine blocked ROS generation, reduced apoptosis mediated by c-Jun-N-terminal kinase, caused a loss of mitochondrial membrane potential, released the cytochrome-c and activated caspases [71]. In another study, UA isolated from several lichens (Alectoria samentosa, Flavocetraria nivalis, Alectoria ochroleuca, and Usnea florida) showed the significant inhibitory potential against migration and invasion of human lung cancer (A549, H460, H1650, and H1975) cells. It was also associated with the inhibition of A549 cells motility. Anti-invasive efficacy of UA in H1650 and H1975 cells was demonstrated by the decline in β-catenin-mediated TOPFLASH and KITENIN-mediated AP-1 activity. Additionally, synthetic derivatives of UA exerted anti-proliferative activity in human prostate cancer (PC-3), human epithelial carcinoma (HeLa), and MCF-7 cells. All active derivatives arrested cells at G0/G1 phase and decreased the fraction of HeLa cells in S and G2/M phases. Derivatives 2a and 2b arrested also MCF-7 cells at G0/G1 phase and induced the apoptosis of these cells. Derivatives 2a and 2b also activated strong cytoplasmic vacuolization, which was associated with dynein-dependent endocytosis. This mechanism has not yet been demonstrated in UA and it is the first time reported in synthetic derivatives of UA [72]. The potasium usnate (PU), which was prepared to enhance the solubility of UA, demonstrated cytotoxic activity in each tested colorectal cancer cells (human HCT116, DLD1, SW480, HT29, SW620, Caco2, COLO320, and mouse CT26) with lower IC50 values than UA except of SW480 and CT26 cells. Potasium usnate showed stronger inhibitory effects on the invasion of Caco2 and HCT116 cells compared to UA. Importantly, PU downregulated the epithelial–mesenchymal markers (EMT) including Twist, Snail, and Snug and decreased the expression of metastasis-related genes CAPN1, CDC42, CFL1, IGF1, WASF1, and WASL in Caco2 cells [34]. An anticancer efficacy of UA was evaluated together with diffractaic acid (DA) and lobaric acid (LA) in human glioblastoma multiforme (U87MG-GBM) and rat cerebral cortex cells (PRCC). Lobaric acid exerted highly toxic effects demonstrated by the drop of cell viability to 35.09% in PRCC and 30.47% in GBM cells, while DA and UA exerted greater total antioxidant capacity (37.74 and 37.34 mmol Trolox equivalent/L, respectively) in PRCC cells when compared with other compounds [73]. Cancer inhibitory effect of UA and atranorin (ATR) was also evaluated in human melanoma (HTB-140) and human prostate cancer (DU-145, PC-3) cell lines. Interestingly, both UA and ATR inhibited cancer cells proliferation, migration and actin organization while their effects on apoptosis were less significant [74]. An impact of UA isolated from Cladonia foliacea Wild and vulpinic acid (VA) isolated from Letharia vulpina Hue on proliferation and viability was evaluated in HepG2, mouse neuroblastoma NS2OY and human umbilical vein endothelial (HUVEC) cells. Although UA was more cytotoxic against all cell lines, it had higher anti-proliferative effects in HepG2 cells. On the other hand, VA inhibited the proliferation of NS2OY cells more effectively. Interestingly, the cytotoxic effects of both metabolites against HUVEC were only mild. Moreover, both UA as well as VA exhibited anti-angiogenic abilities evaluated by the endothelial tube formation assay [75]. Vulpinic acid also decreased viability and induced apoptosis of human breast cancer cells (MCF-7, MDA-MB-231, BT-474, SK-BR-3) compared to human non-malignant breast epithelial cells (MCF-12A). An evaluation of apoptosis-related genes showed that the expression of p53 after VA therapy was almost six times higher in SK-BR-3 cells than in MCF-12A cells [47]. Similarly, an apoptotic activity of VA was evaluated in vitro in CaCo2, HepG2, Hep2C, RD, Wehi as well as in normal Vero and mouse L929 cells. Vulpinic acid inhibited growth of all tested cell lines in a time and dose-dependent manner and a higher efficacy was found in CaCo2 cells. Vulpinic acid also exhibited significant cytotoxic effects on all tested cancer cells. On the other hand, it did not exert any significant cytotoxicity of on normal L929 and Vero cells, but interestingly, all mRNA, Bax protein levels and p53 were more significantly increased in cancer compared to normal cells. In addition, mRNA and Bcl-2 protein levels showed 7 fold decrease in HepG2 and CaCo2 cells and 5–6 fold decrease in Hep2C, RD and Wehi cells [76].
Similarly, natural compound ATR, isolated from lichens, was tested against mouse breast cancer (4T1) cells. ATR reduced the clonogenic potential of 4T1 cells compared to normal mammal non-malignant epithelial (NMuMG) cells, in which the clonogenic ability remained unaffected. BrdU incorporation assay did not confirm the anti-proliferative effect of ATR in 4T1 cells. On the contrary, ATR induced caspase-3 activity, PARP cleavage and depletion of Bcl-xL in 4T1, but not in NmuMG cells [45]. Atranorin, isolated from Stereocaulon caespitosum also inhibited the growth of human hepatocellular carcinoma (SK-Hep1, Huh-7, SNU-182) cell lines when used in concentration higher than 10 μg/mL. Atranorin arrested SK-Hep1 cells at G2/M phase, induced cell death at 24 h time point and suppressed migration and invasiveness of Sk-Hep1 and Huh-7 cells [77].
However, only high concentrations of ATR and gyrophoric acid (GA) had similar effect on human melanoma A375 cells, physodic acid (PA) induced apoptosis in A375 cells by mechanism probably involving the downregulation of HSP70 [17]. In this regard, Emsen et al. analyzed the effect of PA together with olivetoric acid (OA) and psoromic acid (PSA) on U87MG and rat PRCC cells and found a positive correlation between the cytotoxicity associated with the three tested metabolites and their concentrations, lactate dehydrogenase (LDH) activity, and oxidative damage of DNA [43].
Furthermore, Parmotrema dilatatum, Parmotrema lichexanthonicum, Usnea subcavata, Ramalina sp., Dirinaria aspera, Cladina confusa and their secondary metabolites were tested on melanoma cancer (UACC-62), murine melanoma (B16-10), and human fibroblast (NIH/3T3) cells. Protocetraric acid (PrA), norstictic (NA), and PSA (depsidones) acids together with divaricatic (DiA) and perlatolic (PeA) (depsides) acids showed a strong cytotoxic effect on UACC-62 cells and reached higher selectivity for melanoma cells compared to 3T3 normal cells. In this regard, NA and DiA was also the most effective against B16-F10. Protocetraric acid proved to be the best candidate for in vivo studies of melanoma since it showed the highest selectivity index against UACC-62 cells [78].
Paluszczak et al. evaluated effects of lichen-derived compounds on Wnt signaling in colorectal cancer (HCT116, DLD-1) and immortalized keratinocyte (HaCaT) cell lines. Caperatic acid (CA) isolated from Platismatia glauca downregulated β-catenin-regulated expression of Axin2 gene in both colorectal cancer cell lines, but lecanoric acid (LeA), obtained from Hypocenomyce scalaris decreased the expression of Axin2 in HCT116 cells just moderately. On the contrary, CA and PA (isolated from Hypogymnia physodes), downregulated the expression of MMP7 and survivin in HaCat cells in a concentration-dependent manner. Furthermore, CA inhibited the migration of both colorectal carcinomas by 20%, while PA did not have any effect on the cell migration. Caperatic acid showed also the strongest cytotoxic effects on cancer cells at the concentration of 100 μM while other metabolites in this concentration demonstrated just moderate efficacy [26].
Barbatic acid (BA), isolated from acetone extract of Usnea longissima, exhibited strong cytotoxic activity against HeLa, A549, MCF-7, and DU-145 cells. Barbatic acid arrested A549 cells in G0/G1 phase with 71% of cell accumulation at the concentration of 1 μM and the same concentration induced the apoptosis of A549 cells with increased caspase-3 activity, PARP cleavage, annexin V staining, and chromatin condensation [38].
In addition, anti-angiogenic and anti-migratory efficacy of lichen-derived small molecule barbatolic acid (BrA) isolated from acetone extract of Bryoria capillaris was evaluated on T-47D, HUVEC, and cisplatin-resistant BRCA2-mutated human breast TNM stage IV adenocarcinoma cells (HCC1428). Sub-cytotoxic concentrations (25–100 µM) of BrA dose-dependently inhibited both endothelial tube formation as well as migration determined by the scratch wound healing assay. In fact, the effect of BrA on the migration of cancer T-47D and HCC1428 cells was more effective than on normal HUVEC cells [35].
Hypostictic acid (HA), extracted from Pseudoparmelia sphaerospora, exerted significant anti-proliferative efficacy against chronic myelogenous leucaemia (K562), murine melanoma (B16-F10), and renal cancer (786-0) cell lines. Salazanic acid (SA) obtained from Parmotrema cetratum, showed in vitro anti-proliferative effects also in K562 and B16-F10 as well as in colon cancer (HT-29) cell lines [79]. In this regard, SA and protocetraric acid (PrA) are major phenolic compounds in lichens including Parmelia caperata, Parmelia saxatilis and Parmelia sulcate. Manojlovic et al. evaluated an impact of the above-mentioned compounds against human melanoma FemX and LS174 cell lines [80].
Retigeric acid B (RA-B), a pentacyclic triterpenic acid isolated from Lobaria kurokawae induced apoptosis in PC-3 and DU145 cells through the inhibition of IκBα and p65 (subunit of NF-κB) phosphorylations. In addition, microarray analysis revealed alterations in the expression of genes associated with cellular processes including apoptosis, invasion, and proliferation after RA-B application [81].
Protolichesterinic acid (PLA) isolated from Centraria islandica exerted anticancer efficiacy in SK-BR-3 and T47D cell lines while anti-proliferative effects were also observed in SK-BR-3 cells. Indeed, this metabolite increased the expression of fatty acid synthase and decreased the expression of HER2 in SK-BR-3 cells. Additionally, downregulation of ERK1/2 and AKT signaling was observed probably due to reduced HER2. There was also demonstrated some synergistic effect of PLA and lapatinib in SK-BR-3 cells [31]. Similarly, PLA affected HeLa, K562, and human neuroblastoma cell lines (SH-SY5Y). Interestingly, PLA combined with doxorubicin exerted synergistic cytotoxicicity in HeLa cells but not on SH-SY5Y and K562 cells. The mechanism of synergistic effects might be associated with the apoptosis induced by both PLA as well as doxorubicin, which can induce caspases -3, -8, and -9 activities. Bim expression, which mediates cytochrome-c release might be increased in an additive manner by both PLA and doxorubicin. Protolichesterinic acid also seems to behave as a competitive inhibitor of fatty acid synthase [82]. Bessadóttir et al. also tested effects of PLA (from Cetraria islandica) in human multiple myeloma (RPMI 8226), (U266) and human pancreatic carcinoma (AsPC-1). Moreover, PLA inhibited the proliferation of RPMI 8226, U266, and AsPC-1 cells with very low IC50 values. It induced the cell cycle arrest of pancreatic AsPC-1 cells in G1 phase and the apoptosis of RPMI 8226 and U266 cells but not the apoptosis of pancreatic AsPC-1 cells. Indeed, the pro-apoptotic activity of PLA was cancer cell line-dependent while high concentrations of PLA inhibited the production of 5- and 12-HETE but only in pancreatic not in myeloma cells [83].
Ramalin (RAM) a metabolite isolated from Ramalina terebrata inhibited at high concentrations (50 and 100 μg/mL) the proliferation of HCT116 cells. Ramalin induced G2/M cell cycle arrest through the upregulation of TP53 and p21 and downregulation of cyclin B1 and CDK1. The highest concentration of RAM (100 μg/mL) induced significant number of apoptotic cells. Furthermore, wound healing, invasion, and migration of HCT116 cells were also dose-dependently suppressed after the treatment with RAM [84].
Physciosporin (PHY) isolated from Pseudocyphellaria granulata, was evaluated in colorectal cancer cell lines (CT26, Caco2, HCT116, DLD1, SW620). While toxic concentrations of PHY induced apoptosis of tested cancer cells, non-toxic ones inhibited migration, invasion, and colony formation of colon cancer cells in concentration-dependent manner. Physciosporin down-regulated downstream transcription factors and/or target genes of EMT, KITENIN, β-catenin and reduced actin-based cell motility [22]. Another PHY, isolated from Pseudocyphellaria coriacea demonstrated anticancer effects in A549, H1650, and H1975 through inhibition of migration and invasion of human lung cancer cells through the downregulation of N-cadherin, KITENIN-mediated AP-1 activity, Cdc42, and Rac1. Interestingly, the metastasis suppressor gene KAI1 was also upregulated by PHY [85].
Interestingly metabolites isolated from acetone extract of Dirinaria consimilis: antarvediside A (antarA), antarvediside B (antarB), sekikaic acid (SeA), ATR, divaricatic acid (DiA), 2′-O-methyl DiA revealed anticancer potential. In this regard, antarB at the concentration of 30 μg/mL inhibited the growth of HeLa and MCF-7 stronger than the doxorubicin at the concentration of 10 μg/mL. Only metabolites antarB and 2′-O-methyl DiA inhibited the proliferation of A549 cells. Importantly, all metabolites had lower toxicity against human mammary epithelial normal NHME cells in comparison to cancer cells [86]. Table 2 shows an overview of anticancer effects of above-mentioned isolated lichen secondary metabolites.

3.2. Combined Studies of Lichen Extracts and Isolated Lichen Compounds In Vitro

Notably, several authors studied anticancer potential of lichen extracts in combination with isolated lichen compounds. An acetone extract of Flavocetraria cucullata or its subcomponent UA exerted a selective cytotoxicity on cancer cells by inducing apoptosis at lethal concentrations. Sub-lethal concentrations of this extract and UA inhibited tumorigenesis and motility of cancer cells, suppressed epithelial–mesenchymal transition (EMT) and inhibited Akt phosphorylation. Interestingly, the anticancer activity of the extract was more potent than that of UA [87].
Another study analyzed anticancer properties of acetone extract of Toninia candida and Usnea barbata as well as their major compounds nortictic acid (NA) and UA against FemX and LS174 cells using tetrazolium assay. The results of the study proved the anticancer activity of NA, UA, and lichen extracts in both cell lines. Importantly, UA demonstrated the strongest cytotoxic effect while both NA and UA induced apoptosis in FemX and LS174 cells [54]. Moreover, anticancer and antioxidant availabilities of supercritical CO2 extract of old man’s beard (Usnea barbata) (SCE) compared to extracts obtained through conventional methods (Soxhlet extracts and macerate) were evaluated in mouse melanoma (B16), rat glioma (C6), and HaCaT cells. SCE exerted the highest cytotoxic effect on both B16 and C6 cells. The cytotoxic effect of lichen extracts correlated well with the content of UA (the most abundant compound) and ROS production. The cytotoxic effects of extracts were evaluated for apoptosis and autophagy processes via monitoring of cells for cell cycle phases and formation of acidic cytoplasmic vesicles. Consequently, both, SCE and UA induced apoptosis and/or autophagy in B16 and C6 cells. Indeed only very low toxicities of all tested extracts were demonstrated against normal HaCaT cells [88].
Furthermore, acetone extracts of lichens including Evernia prunastri and Pseudoevernia furfuraceae and their major secondary metabolites, mainly PA, demonstrated anticancer activity in FemX and LS174 cells using MTT test. Physodic acid exerted the best cytotoxic effect against both cell lines. Additionally, authors also observed a reduction of FemX and LS174 cells in the S and G2/M phase. On the other hand, there was an increase in sub-G1 population of cells after the treatment with mentioned above extracts and metabolites [89]. Similarly, PA (isolated from Hypogymnia physodes) demonstrated the most significant cytotoxic effect on MCF-7 cells followed by T47D and MDA-MB-231 cells. The viability of MCF-10A cells was not changed even at concentration of PA higher than 100 μM [24].
Ethanol extract of Usnea strigosa as well as its compound NA also showed anticancer effects in MDA-MB-231, MDA-MB-468, MCF-7, T-47D, BT-474, and SK-BR-3 cells. NA exerted anti-proliferative effects in all six breast cancer cell lines with the best inhibition demonstrated in MDA-MB-231 and MDA-MB-468 cells. Nortictic acid inhibited the migration of MDA-MB-468 cells and invasion of MDA-MB-231 cells through the basement membrane. On the other hand, they did not show any significant cytotoxic effects on human non-tumorigenic MCF-10A cells. This acid suppressed also c-Met, STAT3, paxillin/Rac-1and FAK phosphorylation in MDA-MB-231 cells [33]. Moreover, acetone extract of Melanelia subaurifera and Melanelia fuliginosa and their compounds LeA and 2′-O-methyl anziaic acid (2′-O-MA) suppressed HeLa, A549, and LS174 cells. Indeed, LeA and 2′-O-MA metabolites had lower cytotoxic effects compared to both extracts, while both extracts as well as metabolites did not exerted any cytotoxicity on normal MRC5 cells [25].
Ether extract of Cladonia salzmannii showed cytotoxic effects on human acute promyelocytic leukemia (HL-60), human laryngeal carcinoma (HEP-2), human lung mucoepidermoid carcinoma (NCI-H292), and murine macrophage (RAW-264.7) cells. In this regard, acetone extract revealed cytotoxic effects on MCF-7 and NCI-H292 cells while purified BA exerted cytotoxicity on HEP-2, MCF-7 and RAW-264.7 cells [90]. Barbatic acid a secondary metabolite of Cladia aggregate, was applied against HEp-2, human squamous cell lung cancer (NCI-H292), and human nasopharyngeal squamous cell carcinoma (KB). Lichen extract as well as the isolated compound showed the cytotoxicity against all tested cancer cell lines while the most sensitive ones were HE-p2 cells [91].
Acetone extract of Everniastrum vexans inhibited the migration of A549 cells (10 μg/mL). Atranorin identified as an active secondary metabolite of the extract showed the cytotoxic effect on A549 cells at concentrations higher than 5 μg/mL. Atranorin inhibited TOPFLASH activity mediated by β-catenin reducing nuclear import of β-catenin and suppressing the expression of c-jun/AP-1 target genes. Moreover, ATR decreased the mRNA expression of KITENIN and increased the expression of KAI1 mRNA. It also decreased the level of GTP-Cdc42, GTP-RhoA and STAT proteins [20].
In addition, acetone extract of the endolichenic fungus EL002332 isolated from Endocarpon pusillum exerted selective cytotoxicity on human gastric cancer AGS and mouse colon cancer CT26 cell lines. It also showed synergistic effects on human gastric cell lines (AGS, TMK-1) in combination with docetaxel chemotherapy. Active compound isolated from acetone extract (myC) had even higher anticancer effect on AGS cells than the crude extract. In fact, myC induced apoptosis through the activation of caspase activity and regulating the expression of Bcl-2 family proteins [92].
Table 3 summarizes combined studies of lichen extracts and isolated lichen compounds evaluating their anticancer effectiveness.

3.3. Determination of Lichens’ Extracts Effect Against Cancer Cells

In addition, beneficial anticancer activity is associated also with lichen extracts. As revealed by Kosanic et al., extracts of Umbilicaria crustulosa, Umbilicaria cylindrica, and Umbilicaria polyphylla showed strong anti-neoplastic effects against FemX and LS174 cell lines evaluated using MTT assay [93]. Similarly, methanol extract of Lasallia pustulata and extracts from Parmelia caperata, Parmelia sulcate, and Parmelia saxatilis showed strong cytotoxic activity against FemX and LS174 [94,95]. Another study evaluated the anticancer effect of 70% methanolic extract of Parmotrema reticulatum against MCF-7 and A549 cancer cells and normal fibroblast WI-38 cells. Analyzed data documented the strong cytotoxic activity of this extract on MCF-7 cells, while low cytotoxicity on A549 and WI-38 cells. Moreover, the extract induced S and G2 cell cycle arrest of MCF-7 cells associated with decreased expression of B1, Cdk2, Cdc25C, and increased expression of p53 and p21. An increased ratio of Bax/Bcl-2 and higher caspase activity were detected in MCF-7 cells leading to PARP cleavage and apoptosis [96]. Another study demonstrated anticancer activity of methanol extract of Cetraria islandica on FemX and LS174 cells [97]. In addition, Kosanic et al. studied biological activity of Parmelia arseneana and Acarospora fuscata acetone extracts against FemX, LS174, A549, and human chronic myeologenous leucaemia cells K562. Their results revealed strong anticancer effects of the P. Arseneana extract in all tested cell lines [98]. All extracts of unique trans-Himalayan lichens demonstrated a cytotoxic effect against HepG2 and human colon cancer (RKO) cells. Moreover, methanol extract of Lobothallia alphoplaca and Melanelia disjuncta exhibited high cytotoxicity against both cancer cell lines [99]. Additionally, treatment of A549, PC-3, Hep3B, and C6 cell lines with methanol extracts of Parmelia sulcata Taylor and Usnea filipendula Stirt led to the cytotoxic, genotoxic and apoptosis-inducing effects in all tested cell lines [100]. Another in vitro experiment of methanol extract of Hypogymnia physodes (low concentrations) indicated anticancer and/or apoptosis-inducing effects on MCF-7 and MDA-MB-231 cells and the genotoxic (high concentrations) impact on human lymphocytes [101]. Moreover, methanol extracts of Cladonia rangiformis and Cladonia convolute exerted a significant anti-proliferative effect and these extracts induced also significant apoptosis of MCF-7 cells demonstrated by TUNEL assay [102]. Additionally, acetone extract of Xanthoria parietina represents a unique mixture of the secondary metabolites with anticancer abilities. X. parietina extract inhibited the proliferation of MCF-7 and MDA-MB-231 cells with the maximum effect observed at the concentration of 1.5–3 mg/mL. This extract induced the G1 cell cycle inhibition with the upregulation of p27 and p16 and downregulation of cyclin D1 and cyclin A. Interestingly, therapy of MCF-7 and MDA-MB-231 cells with X. parietina decreased the expression of anti-apoptotic Bcl-2 and increased the expression of pro-apoptotic TRAIL and pBAD proteins [103]. In addition, a metanol extract of Parmelia sulcata had a cytotoxic effect on both MCF-7 and MDA-MB-231 cells. The apoptosis induced in MCF-7 and MDA-MB-231 cells by P. sulcata extract was rather caspase-independent due to the lack of changes in caspase-3 activation or in PARP cleavage [104]. Recently, researchers evaluated crude extracts of 17 lichen species against Human Burkitt’s lymphoma (Raji). Xanthoparmelia chlorochroa and Tuckermannopsis ciliaris extracts induced apoptosis (dose-dependent), accumulation of cells in the GO/G1 stage and increase of p53 protein [105]. Acetone extracts of Parmotrema gardneri, Pannaria sp., and Canoparmelia aptata applied to human gastric adenocarcinoma (AGS) and A549 cancer cells as well as normal Canine Madin–Darby kidney cells (MDCK) exerted anticancer potential. On the contrary, C. aptata extract had just weak cytotoxicity on AGS and A549 cells [106]. Additionally, Cladonia rangiformis and Cladonia convolute represent fruticose lichens with antimicrobial and cytotoxic effects. The extracts from both lichens showed strong cytotoxic impact on MCF-7 cells [107].
Furthermore, Caloplaca pusilla grown on G-LBM medium decreased cell viability and induced apoptosis of MCF-7, PC-3 and HeLa cells in concentration-dependent manner. Mycelia of Xanthoria parietina grown on PDA and G-LBM decreased cell the viability of MCF-7 and HeLa cell lines [108]. Also, methanol extract of Cladonia pocillum increased the apoptosis in MCF-7 cells in concentration-dependent manner [109]. Extract of Pleurosticta acetabulum exerted strong cytotoxic effect on HT-29 cells. This extract also inhibited the proliferation of HT-29 cells through the presence of cytochalasin E and induced apoptosis [110]. Furthermore, polysaccharide from Umbilicaria esculenta decreased the viability of human melanoma A875 and A375 cells but did not exhibit any cytotoxic effect on HUVEC cells. Both, Annexin-V positive as well as TUNEL positive A875 cells were induced after this therapy in concentration-dependent manner. In this regard, apoptosis of A875 but not HUVEC cells was induced by polysaccharide of U. esculenta (as a result of ROS generation) followed by increased expression of caspase-3 and -9 [111]. An evaluation of biological activities of five lichen species of Cladonia genus revealed the highest cytotoxic effect of Cladonia foliacea extract on A549 and human colon cancer (LS174) cells. Moreover, the highest cytotoxic effect on HeLa cells was observed after the treatment with the extract of Cladonia furcata [112]. In addition, high concentration of Candelariella vitellina extract reduced the proliferation (Ki-67) and induced apoptosis and necrosis of Caco-2 cells. The extract of C. vitelline decreased Bcl-2 but increased Bax and CASP3 protein levels, so the ratio of Bax/Bcl-2 increased [32]. Anticancer potential of methanol extracts of lichens was evaluated in human lung (H1299, A549) and breast (MDA-MB-231, MCF-7) cancer cell lines. The extract of Usnea intermedia exerted the strongest anti-proliferative effects especially observed in H1299 and MDA-MB-231 cells. The extract of U. intermedia also induced apoptosis confirmed via the phosphatidylserine translocation, increased caspase 3/7 activity, loss of mitochondrial membrane potential and the formation of pyknotic nuclei [113]. Isolates from Nemania serpens and Nemania aenea var. aureolatum were the most active compounds against human colorectal cancer (HT-29, HCT116) and human prostate cancer (PC-3, DU145) cell lines. These isolates induced apoptosis characterized by activated caspase 3, 8, PARP cleavage and chromatin fragmentation [114]. In addition, Nugraha et al. evaluated anticancer activities of nine lichens of East Java Indonesia, of which Physcia cf. Milegrana showed the most significant cytoxicity, but only on HeLa cells. Other metabolites exhibited very low cytotoxicity either in cancer or normal African green monkey kidney (Vero) cells [115]. Table 4 shows an overview of anticancer effects of above-mentioned lichen extracts in various cancer cell lines.

4. Anticancer Effects of Lichens in Animal Models

Anticancer activities of naturally occurring plant compounds and extracts derived from lichens are supported by numerous studies using animal xenograft/allograft models. There are several studies evaluating the anticancer potential of UA in vivo. Usnic acid inhibited angiogenesis in both chick embryo chorioallantoic membrane as well as in VEGF-induced mouse corneal angiogenesis model. Moreover, UA inhibited the growth of human Bcap-37 BC cells inoculated into C57BL/6 female nude mice and suppressed the angiogenesis in tumor tissue. The inhibition of angiogenesis was evaluated by anti-CD31 and showed the reduction in integrated optical density of tumor blood vessels in the UA-treatment group. In this regard, in vivo study was supported by the parallel in vitro evaluation, where UA reduced proliferation, migration, and tube formation of HUVEC cells, blocked VEGFR2 mediated ERK1/2 and AKT signaling and induced apoptosis via decrease in Bcl-x1 and survivin levels and through the elevation of caspase 3 activity and PARP cleavage [116]. Furthermore, UA inhibited tumor growth of human MCF-7 breast cancer-bearing mice dose-dependently through the generation of ROS and JNK stimulation associated with mitochondrial/caspase pathway leading to apoptosis of cancer cells. Indeed, UA was well tolerated and did not show any toxic effects in animals [71]. This metabolite reduced also toxicity of bleomycin therapy in Kunming mice with inoculated mouse H22 hepatocellular carcinoma cells and the combination of these molecules was more effective against H22 cancer compared to single bleomycine. Indeed, the combination of UA and bleomycin arrested tumor cells at G0/G1 phase and induced the apoptosis via caspase-3 and -8 activation (cleavage) probably as a consequence of transcription regulation of p53/p21/cyclin pathway. Furthermore, UA decreased the level of MDA, hydroxyproline, TNF-α, IL-1β, IL-6, and TGF-β1 and its combination with bleomycin increased the level of SOD in lung tissues of H22-bearing mice probably through the downregulation of p-Smad2/3 and the upregulation of Smad7 proteins [36]. Furthermore, benzylidene analogue of UA demonstrated anticancer activity in vivo on two MDA-MB-231 and MCF-7 xenograft mouse breast cancer models. Both UA as well as its benzylidene analogue induced autophagy and the inhibition of mTOR signaling pathway accompanied by significantly decreased level of mTOR downstream effectors p-S6K and p-4E-BP1 in treatment groups of both models [117]. In the mouse model of human gastric BGC823 carcinoma, UA decreased the volume and weight of tumors without any weight loss of animals. Moreover, evaluating the Bax/Bcl-2 ratio, UA exhibited stronger pro-apoptotic activity, when compared to 5-FU. Although 5-FU had the same effect on tumor volume and weight, it was associated with significant animal weight loss [69]. In another experiment, Nguyen et al. [87] tested anticancer activities of Flavocetraria cucullata acetone extract, UA, and LiA on human A549 lung cancer cells using xenograft Balb/c mouse model [87]. The highest tumor free survival number was found in F. cucullata pretreated group (tumor free in six out of eight mouse) compared to DMSO group (zero out of eight), UA group (four out of eight) or LiA group (two out of eight) [87].
Interestingly, PU demonstrated better bioavailability in the tumor, liver, and plasma compared to UA in CT26-Fluc syngeneic mouse tumor model. Potassium usnate inhibited the growth of colorectal cancer cells and inhibited liver metastasis probably through the reduction of EMT markers such Twist, Snail, and Slug and the metastasis-related genes CAPN1, CDC42, CFL1, IGF1, WASF1, and WASL in this model. In addition, PU did not have any hepatotoxic effect in the mouse liver metastasis model [34]. Interestingly, ethyl acetate extract of Usnea longissima prevented esophagogastric adenocarcinoma induced by oral administration of N-methyl-N-nitro-N-nitrosoguanidin in Albino Wistar male rats. U. longissima extract demonstrated prominent anticancer effect and selectivity to cancer tissue in animals at concentrations of 50 and 100 mg/kg without any toxic effects. This extract did not reveal any lethal effect even when administered in high concentrations (500, 1000, and 2000 mg/kg) [118].
Study from our laboratory showed significant anticancer effects of ATR in mouse 4T1 breast cancer allograft model in BALB/c mice. Atranorin significantly increased survival time of tumor-bearing animals, reduced the tumor volume and had rather direct proapoptotic than anti-proliferative effect on tumor cells. In addition, ATR protected livers of tumor-bearing mice against oxidative stress [45]. In another study, ATR reduced tumor volume and weight and diminished Ki-67 marker of proliferation in Lewis lung carcinoma xenograft model in C57BL/6 mice. Moreover, ATR decreased the expression of KITENIN, CD44, STAT, and cyclin-D1 genes in both in vitro as well as in in vivo conditions [20].
There are also several more lichen extracts or metabolites that were evaluated in cancer animal models. In this regard, Poornima et al. evaluated the extract of Rocella montagnei against Dalton’s lymphoma ascites cells, that were inoculated into Albino Wistar rats and resulted in suppression of tumor growth. Briefly about experiment workflow: Thirteen days after inoculation, cancer fluid was aspirated from the peritoneal cavity of rats and consequently injected into another group of animals. Analyzed data demonstrated, that lichen extract reduced the volume of tumors and the effect was comparable to standard therapy with Vincristine [119]. In another study, the anticancer effects of endolichenic fungus EL002332 (Endocarpon pusillum) extract was evaluated in mouse colorectal CT26 cancer cells that were inoculated into BALB/c syngeneic mice. TMK1 cells were injected into the abdominal cavity of BALB/c mice to establish intraperitoneal xenografts. Both tumor score and tumor volume were significantly reduced in skin and intraperitoneal tumor-bearing animals after EL002332 crude extract [92]. El-Garawani et al. tested anticancer activities of Candelariella vitelline extract on Ehrlich ascites carcinoma cells that were injected and consequently transferred every 5 days to new Swiss albino mice by inoculation. Both C. vitelline extract (150 mg/kg) and 5-FU (20 mg/kg) reduced the tumor volume by 80% and 69.8%, respectively. Both therapies decreased tumor cell invasion, mitotic activity and increased the formation of apoptotic bodies evaluated using H&E staining and further processed by immunohistochemistry. Notably, mRNA expression of Bax and caspase 3 was decreased and the elevated level of Bcl-2 was detected in solid Ehrlich carcinoma tissue in the treated group [32]. Lichen-derived molecule PHY, isolated from Pseudocyphellaria granulate, inhibited the growth of CT26 xenograft in BALB/c mice. The authors observed reduced tumor volume and weight by 55% and 75%, respectively vs control animals after the treatment. In addition, PHY did not reduce the animal body weight [22].
Several lichen-derived acids demonstrated promissing therapeutic efficacy in cancer models in vivo. The study aims of Martins et al. were to perform in vivo evaluations of the anticancer potential of BA isolated from the lichen Cladia aggregata. Sarcoma-180 cells were inoculated in the right axillary region of female albino Swiss mice. Results showed that BA did not affect the proliferation of tumor cells, however, it significantly reduced tumor weight. Barbatic acid also demonstrated low toxicity rate in animals. Experimental data in vivo showed a tight cross-connection between the application of secondary metabolite of lichens and antineoplastic events in tumor cells, probably through the activation of the apoptotic cascade leading to cell death [91]. Hypostictic acid and SA, isolated from Pseudoparmelia sphaerospora and Parmotrema cetratum respectively, showed anti-tumor effects in the murine melanoma model using B16-F10 cell line. Authors found that SA reduced tumor volume by 88% and HA by 72% compared to controls. Interestingly, both acids demonstrated high cancer selectivity associated with low animal toxicity [79]. Karagoz et al. found the anticancer effect of DA on Ehrlich ascites carcinoma cells (EAC) inoculated into Balb/C mice. Indeed, histopathological and hematological analyses demonstrated that lower concentrations of DA have protective effects in various organs when compared to higher ones [120]. Similarly, in another mice xenograft model, NA reduced both tumor volume and weight of human MDA-MB-231/GFP cells when compared to untreated controls. Western blot analysis of tumor tissue lysates revealed significant inhibition of cell signaling pathway linked with c-Met phosphorylation in treated animals [33]. Table 5 shows an overview of in vivo studies evaluating anticancer effects of extracts or isolated secondary metabolites of lichens.

5. Conclusions and Future Directions

Lichens represent a rich source of bioactive molecules that have great potential of their clinical utility in cancer disease. The modern methodologic biomedicinal approaches that facilitate an isolation and characterization of lichen metabolites and consequently define their very complex biological cellular effects, provide suitable conditions for the acceleration of the research which includes the future clinical testing and following use of lichen-derived anticancer drugs in medical practice [121]. In this paper, we have comprehensively reviewed most recent scientific preclinical data of this topic. We have described significant anticancer efficacy of isolated lichen compounds, lichen extracts, or isolated compounds in combination with lichen extracts in cancer cell lines or animal cancer models. Despite above mentioned optimistic scenario about perspective of lichens extracts or isolated lichen molecules as anticancer substances, we did not find any relevant clinical research that provide insight into the anticancer activity of lichen species in humans so far. Therefore, further studies are still needed to determine the potential clinical application and clarify their beneficial effects in cancer patients or risk individuals [9,122]. However, regarding clinical utility of lichens’ secondary metabolites in cancer disease, there are logic limitations that must be addressed to clinical oncologists/researchers. Most of malignant tumors represent very dynamic structures with numerous cell lines that differ in genotypes and phenotypes. There can be observed in such malignancies, a highly variable sensitivity to therapeutics and some of cell lines develop resistance to the treatment, including lichen-derived molecules or extracts [8,14]. The concept using of the drug combination of lichen-derived molecules (or their natural coctails present in extracts) with the conventional therapy to target a wider range of signaling pathways in cancer cells seems to be substantially beneficial compared to using of single anticancer drug (e.g., in the delay of drug resistance, prolonging the progression-free and overall survival in patients or prolonging the cancer latency during long-term chemoprevention in high-risk individuals) [123].
The future preclinical and clinical research focused on isolated lichen metabolites or extracts influencing the carcinogenesis should be directed toward the several important issues: (1) Clarifying the molecular targets and signaling pathways involved in anticancer activity. (2) Determining of an effective (and non-toxic) doses in humans. (3) Assessing a combined effect of several lichen metabolites or extracts targeting several relevant genetic, epigenetic, and immunomodulatory pathways. (4) Investigating epigenetic mechanisms such as modifications of methylation status in gene promoters, posttranslation histone modifications, and expression on miRNAs spectrum. (5) Assessing of the cancer stem cells survival, regarding the relapse, multidrug resistance, or re-sensitizing cancer cells towards standard chemotherapy. (6) Considering the improved bioavailability of lichen metabolites by utilizing, for example, nanoparticles carriers. (7) The detection of new spectrum of lichen-derived bioactive molecules with proved anticancer activities in the form of isolated molecules or extracts, that can potentially provide progressive therapeutic approaches for clinical practice. (8) Chemical structure of most lichen molecules is simple which prompt their easy synthesis. In this regard, many of these synthetic substances may be applied as precursors to fit specific mechanisms of anticancer action, increase stability, and decrease undesirable side effects in the body, that could lead to their improved anticancer activities and provide reasonable clinical use. (10) Later, it could be better understanding of the target mechanisms associated with the individual characteristics with the aim to develop personalized medications from lichens.

Author Contributions

Z.S. and P.S. were responsible for the conception. Manuscript was drafted by Z.S., A.L., M.S., and P.K. and critically revised by D.B. and P.S. All authors have read and agreed to the published version of the manuscript.

Funding

The present study was supported by the Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic (Bratislava, Slovak Republic; grant no. VEGA 1/0536/19).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Mathers, C.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int. J. Cancer 2019, 144, 1941–1953. [Google Scholar] [CrossRef] [Green Version]
  2. Patafio, F.M.; Brooks, S.C.; Wei, X.; Peng, Y.; Biagi, J.; Booth, C.M. Research output and the public health burden of cancer: Is there any relationship? Curr. Oncol. 2016, 23, 75. [Google Scholar] [CrossRef] [Green Version]
  3. Solár, P.; Ferenc, P.; Koval’, J.; Mikeš, J.; Solárová, Z.; Hrčková, G.; Fulton, B.L.; Fedoročko, P. Photoactivated Hypericin Induces Downregulation of HER2 Gene Expression. Radiat. Res. 2011, 175, 51–56. [Google Scholar] [CrossRef]
  4. Abotaleb, M.; Samuel, S.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Büsselberg, D. Flavonoids in Cancer and Apoptosis. Cancers 2018, 11, 28. [Google Scholar] [CrossRef] [Green Version]
  5. Kubatka, P.; Uramova, S.; Kello, M.; Kajo, K.; Samec, M.; Jasek, K.; Vybohova, D.; Liskova, A.; Mojzis, J.; Adamkov, M.; et al. Anticancer Activities of Thymus vulgaris L. in Experimental Breast Carcinoma in Vivo and in Vitro. IJMS 2019, 20, 1749. [Google Scholar] [CrossRef] [Green Version]
  6. Kapinova, A.; Kubatka, P.; Golubnitschaja, O.; Kello, M.; Zubor, P.; Solar, P.; Pec, M. Dietary phytochemicals in breast cancer research: Anticancer effects and potential utility for effective chemoprevention. Environ. Health Prev. Med. 2018, 23, 36. [Google Scholar] [CrossRef]
  7. Samec, M.; Liskova, A.; Kubatka, P.; Uramova, S.; Zubor, P.; Samuel, S.M.; Zulli, A.; Pec, M.; Bielik, T.; Biringer, K.; et al. The role of dietary phytochemicals in the carcinogenesis via the modulation of miRNA expression. J. Cancer Res. Clin. Oncol. 2019, 145, 1665–1679. [Google Scholar] [CrossRef]
  8. Liskova, A.; Kubatka, P.; Samec, M.; Zubor, P.; Mlyncek, M.; Bielik, T.; Samuel, S.M.; Zulli, A.; Kwon, T.K.; Büsselberg, D. Dietary Phytochemicals Targeting Cancer Stem Cells. Molecules 2019, 24, 899. [Google Scholar] [CrossRef] [Green Version]
  9. Uramova, S.; Kubatka, P.; Dankova, Z.; Kapinova, A.; Zolakova, B.; Samec, M.; Zubor, P.; Zulli, A.; Valentova, V.; Kwon, T.K.; et al. Plant natural modulators in breast cancer prevention: Status quo and future perspectives reinforced by predictive, preventive, and personalized medical approach. EPMA J. 2018, 9, 403–419. [Google Scholar] [CrossRef]
  10. Chen, V.; Staub, R.E.; Fong, S.; Tagliaferri, M.; Cohen, I.; Shtivelman, E. Bezielle Selectively Targets Mitochondria of Cancer Cells to Inhibit Glycolysis and OXPHOS. PLoS ONE 2012, 7, e30300. [Google Scholar] [CrossRef] [Green Version]
  11. Perez, A.T.; Arun, B.; Tripathy, D.; Tagliaferri, M.A.; Shaw, H.S.; Kimmick, G.G.; Cohen, I.; Shtivelman, E.; Caygill, K.A.; Grady, D.; et al. A phase 1B dose escalation trial of Scutellaria barbata (BZL101) for patients with metastatic breast cancer. Breast Cancer Res. Treat. 2010, 120, 111–118. [Google Scholar] [CrossRef] [PubMed]
  12. Rugo, H.; Shtivelman, E.; Perez, A.; Vogel, C.; Franco, S.; Tan Chiu, E.; Melisko, M.; Tagliaferri, M.; Cohen, I.; Shoemaker, M.; et al. Phase I trial and antitumor effects of BZL101 for patients with advanced breast cancer. Breast Cancer Res. Treat. 2007, 105, 17–28. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, G.; Wang, Y.; Zhang, Y.; Wan, X.; Li, J.; Liu, K.; Wang, F.; Liu, Q.; Yang, C.; Yu, P.; et al. Anti-Cancer Activities of Tea Epigallocatechin-3-Gallate in Breast Cancer Patients under Radiotherapy. CMM 2012, 12, 163–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kapinova, A.; Stefanicka, P.; Kubatka, P.; Zubor, P.; Uramova, S.; Kello, M.; Mojzis, J.; Blahutova, D.; Qaradakhi, T.; Zulli, A.; et al. Are plant-based functional foods better choice against cancer than single phytochemicals? A critical review of current breast cancer research. Biomed. Pharmacother. 2017, 96, 1465–1477. [Google Scholar] [CrossRef]
  15. Jasek, K.; Kubatka, P.; Samec, M.; Liskova, A.; Smejkal, K.; Vybohova, D.; Bugos, O.; Biskupska-Bodova, K.; Bielik, T.; Zubor, P.; et al. DNA Methylation Status in Cancer Disease: Modulations by Plant-Derived Natural Compounds and Dietary Interventions. Biomolecules 2019, 9, 289. [Google Scholar] [CrossRef] [Green Version]
  16. Stanojković, T. Investigations of Lichen Secondary Metabolites with Potential Anticancer Activity. In Lichen Secondary Metabolites: Bioactive Properties and Pharmaceutical Potential; Ranković, B., Ed.; Springer International Publishing: Cham, Switzerland, 2015; pp. 127–146. ISBN 978-3-319-13374-4. [Google Scholar]
  17. Cardile, V.; Graziano, A.C.E.; Avola, R.; Piovano, M.; Russo, A. Potential anticancer activity of lichen secondary metabolite physodic acid. Chem. Biol. Interact. 2017, 263, 36–45. [Google Scholar] [CrossRef]
  18. White, P.; Oliveira, R.; Oliveira, A.; Serafini, M.; Araújo, A.; Gelain, D.; Moreira, J.; Almeida, J.; Quintans, J.; Quintans-Junior, L.; et al. Antioxidant Activity and Mechanisms of Action of Natural Compounds Isolated from Lichens: A Systematic Review. Molecules 2014, 19, 14496–14527. [Google Scholar] [CrossRef]
  19. Yang, Y.; Nguyen, T.T.; Jeong, M.-H.; Crişan, F.; Yu, Y.H.; Ha, H.-H.; Choi, K.H.; Jeong, H.G.; Jeong, T.C.; Lee, K.Y.; et al. Inhibitory Activity of (+)-Usnic Acid against Non-Small Cell Lung Cancer Cell Motility. PLoS ONE 2016, 11, e0146575. [Google Scholar] [CrossRef]
  20. Zhou, R.; Yang, Y.; Park, S.-Y.; Nguyen, T.T.; Seo, Y.-W.; Lee, K.H.; Lee, J.H.; Kim, K.K.; Hur, J.-S.; Kim, H. The lichen secondary metabolite atranorin suppresses lung cancer cell motility and tumorigenesis. Sci. Rep. 2017, 7, 8136. [Google Scholar] [CrossRef] [Green Version]
  21. Nguyen, T.T.H.; Dinh, M.H.; Chi, H.T.; Wang, S.-L.; Nguyen, Q.; Tran, T.D.; Nguyen, A.D. Antioxidant and cytotoxic activity of lichens collected from Bidoup Nui Ba National Park, Vietnam. Res. Chem. Intermed. 2019, 45, 33–49. [Google Scholar] [CrossRef]
  22. Taş, İ.; Han, J.; Park, S.-Y.; Yang, Y.; Zhou, R.; Gamage, C.D.B.; Van Nguyen, T.; Lee, J.-Y.; Choi, Y.J.; Yu, Y.H.; et al. Physciosporin suppresses the proliferation, motility and tumourigenesis of colorectal cancer cells. Phytomedicine 2019, 56, 10–20. [Google Scholar] [CrossRef]
  23. Zambare, V.P.; Christopher, L.P. Biopharmaceutical potential of lichens. Pharm. Biol. 2012, 50, 778–798. [Google Scholar] [CrossRef]
  24. Studzińska-Sroka, E.; Piotrowska, H.; Kucińska, M.; Murias, M.; Bylka, W. Cytotoxic activity of physodic acid and acetone extract from Hypogymnia physodes against breast cancer cell lines. Pharm. Biol. 2016, 54, 2480–2485. [Google Scholar] [CrossRef] [Green Version]
  25. Ristić, S.; Ranković, B.; Kosanić, M.; Stanojković, T.; Stamenković, S.; Vasiljević, P.; Manojlović, I.; Manojlović, N. Phytochemical study and antioxidant, antimicrobial and anticancer activities of Melanelia subaurifera and Melanelia fuliginosa lichens. J. Food Sci. Technol. 2016, 53, 2804–2816. [Google Scholar] [CrossRef] [Green Version]
  26. Paluszczak, J.; Kleszcz, R.; Studzińska-Sroka, E.; Krajka-Kuźniak, V. Lichen-derived caperatic acid and physodic acid inhibit Wnt signaling in colorectal cancer cells. Mol. Cell. Biochem. 2018, 441, 109–124. [Google Scholar] [CrossRef] [Green Version]
  27. Yurdacan, B.; Egeli, U.; Eskiler, G.G.; Eryilmaz, I.; Cecener, G.; Tunca, B. The role of usnic acid-induced apoptosis and autophagy in hepatocellular carcinoma. Hum. Exp. Toxicol. 2019, 38, 201–215. [Google Scholar] [CrossRef]
  28. Singh, N.; Nambiar, D.; Kale, R.K.; Singh, R.P. Usnic Acid Inhibits Growth and Induces Cell Cycle Arrest and Apoptosis in Human Lung Carcinoma A549 Cells. Nutr. Cancer 2013, 65, 36–43. [Google Scholar] [CrossRef]
  29. Dinçsoy, A.B.; Duman, D.C. Changes in apoptosis-related gene expression profiles in cancer cell lines exposed to usnic acid lichen secondary metabolite. Turk. J. Biol. 2017, 41, 484–493. [Google Scholar] [CrossRef] [Green Version]
  30. Hong, J.-M.; Suh, S.-S.; Kim, T.; Kim, J.; Han, S.; Youn, U.; Yim, J.; Kim, I.-C. Anti-Cancer Activity of Lobaric Acid and Lobarstin Extracted from the Antarctic Lichen Stereocaulon alpnum. Molecules 2018, 23, 658. [Google Scholar] [CrossRef] [Green Version]
  31. Bessadóttir, M.; Skúladóttir, E.Á.; Gowan, S.; Eccles, S.; Ómarsdóttir, S.; Ögmundsdóttir, H.M. Effects of anti-proliferative lichen metabolite, protolichesterinic acid on fatty acid synthase, cell signalling and drug response in breast cancer cells. Phytomedicine 2014, 21, 1717–1724. [Google Scholar] [CrossRef]
  32. El-Garawani, I.M.; Elkhateeb, W.A.; Zaghlol, G.M.; Almeer, R.S.; Ahmed, E.F.; Rateb, M.E.; Abdel Moneim, A.E. Candelariella vitellina extract triggers in vitro and in vivo cell death through induction of apoptosis: A novel anticancer agent. Food Chem. Toxicol. 2019, 127, 110–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ebrahim, H.Y.; Elsayed, H.E.; Mohyeldin, M.M.; Akl, M.R.; Bhattacharjee, J.; Egbert, S.; El Sayed, K.A. Norstictic Acid Inhibits Breast Cancer Cell Proliferation, Migration, Invasion, and In Vivo Invasive Growth Through Targeting C-Met: Norstictic Acid as Breast Cancer and c-Met Inhibitor. Phytother. Res. 2016, 30, 557–566. [Google Scholar] [CrossRef] [Green Version]
  34. Yang, Y.; Bae, W.K.; Lee, J.-Y.; Choi, Y.J.; Lee, K.H.; Park, M.-S.; Yu, Y.H.; Park, S.-Y.; Zhou, R.; Taş, İ.; et al. Potassium usnate, a water-soluble usnic acid salt, shows enhanced bioavailability and inhibits invasion and metastasis in colorectal cancer. Sci. Rep. 2018, 8, 16234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Varol, M. Anti-breast cancer and anti-angiogenic potential of a lichen-derived small-molecule: Barbatolic acid. Cytotechnology 2018, 70, 1565–1573. [Google Scholar] [CrossRef]
  36. Su, Z.-Q.; Liu, Y.-H.; Guo, H.-Z.; Sun, C.-Y.; Xie, J.-H.; Li, Y.-C.; Chen, J.-N.; Lai, X.-P.; Su, Z.-R.; Chen, H.-M. Effect-enhancing and toxicity-reducing activity of usnic acid in ascitic tumor-bearing mice treated with bleomycin. Int. Immunopharmacol. 2017, 46, 146–155. [Google Scholar] [CrossRef]
  37. Kiliac, N.; Islakoglu, Y.O.; Buyuk, A.; Gur-Dedeoglu, B.; Cansaran-Duman, D. Determination of Usnic Acid Responsive miRNAs in Breast Cancer Cell Lines. ACAMC 2018, 18, 1463–1472. [Google Scholar]
  38. Reddy, S.D.; Siva, B.; Kumar, K.; Babu, V.S.P.; Sravanthi, V.; Boustie, J.; Nayak, V.L.; Tiwari, A.K.; Rao, C.V.; Sridhar, B.; et al. Comprehensive Analysis of Secondary Metabolites in Usnea longissima (Lichenized Ascomycetes, Parmeliaceae) Using UPLC-ESI-QTOF-MS/MS and Pro-Apoptotic Activity of Barbatic Acid. Molecules 2019, 24, 2270. [Google Scholar] [CrossRef] [Green Version]
  39. Sepulveda, B.; Chamy, M.C.; Piovano, M.; Areche, C. LICHENS: MIGHT BE CONSIDERED AS A SOURCE OF GASTROPROTECTIVE MOLECULES? J. Chil. Chem. Soc. 2013, 58, 1750–1752. [Google Scholar] [CrossRef] [Green Version]
  40. Demir, L.; Toğar, B.; Türkez, H.; Sozio, P.; Aslan, A.; Stefano, A.D.; Demir, L.; Toğar, B.; Türkez, H.; Sozio, P.; et al. The investigation of cytogenetic and oxidative effects of diffractaic acid on human lymphocyte cultures. Braz. Arch. Biol. Technol. 2015, 58, 75–81. [Google Scholar] [CrossRef] [Green Version]
  41. Morita, H.; Tsuchiya, T.; Kishibe, K.; Noya, S.; Shiro, M.; Hirasawa, Y. Antimitotic activity of lobaric acid and a new benzofuran, sakisacaulon A from Stereocaulon sasakii. Bioorg. Med. Chem. Lett. 2009, 19, 3679–3681. [Google Scholar] [CrossRef]
  42. Kim, T.K.; Kim, J.E.; Youn, U.J.; Han, S.J.; Kim, I.-C.; Cho, C.-G.; Yim, J.H. Total Syntheses of Lobaric Acid and Its Derivatives from the Antarctic Lichen Stereocaulon alpinum. J. Nat. Prod. 2018, 81, 1460–1467. [Google Scholar] [CrossRef]
  43. Emsen, B.; Aslan, A.; Togar, B.; Turkez, H. In Vitro antitumor activities of the lichen compounds olivetoric, physodic and psoromic acid in rat neuron and glioblastoma cells. Pharm. Biol. 2016, 54, 1748–1762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Studzinska-Sroka, E.; Galanty, A.; Bylka, W. Atranorin—An Interesting Lichen Secondary Metabolite. Mini Rev. Med. Chem. 2017, 17, 1633–1645. [Google Scholar] [CrossRef] [PubMed]
  45. Solár, P.; Hrčková, G.; Koptašíková, L.; Velebný, S.; Solárová, Z.; Bačkor, M. Murine breast carcinoma 4T1 cells are more sensitive to atranorin than normal epithelial NMuMG cells in vitro: Anticancer and hepatoprotective effects of atranorin in vivo. Chem. Biol. Interact. 2016, 250, 27–37. [Google Scholar] [CrossRef] [PubMed]
  46. Sahin, E.; Dabagoglu Psav, S.; Avan, I.; Candan, M.; Sahinturk, V.; Koparal, A. Vulpinic acid, a lichen metabolite, emerges as a potential drug candidate in the therapy of oxidative stress–related diseases, such as atherosclerosis. Hum. Exp. Toxicol. 2019, 38, 675–684. [Google Scholar] [CrossRef]
  47. Kılıç, N.; Aras, S.; Cansaran-Duman, D. Determination of Vulpinic Acid Effect on Apoptosis and mRNA Expression Levels in Breast Cancer Cell Lines. ACAMC 2019, 18, 2032–2041. [Google Scholar] [CrossRef]
  48. Türk, H.; Yılmaz, M.; Tay, T.; Türk, A.Ö.; Kıvanç, M. Antimicrobial Activity of Extracts of Chemical Races of the Lichen Pseudevernia furfuracea and their Physodic Acid, Chloroatranorin, Atranorin, and Olivetoric Acid Constituents. Z. Für Nat. C 2006, 61, 499–507. [Google Scholar] [CrossRef]
  49. Stojanović, I.Z.; Najman, S.; Jovanović, O.; Petrović, G.; Najdanović, J.; Vasiljević, P.; Smelcerović, A. Effects of depsidones from Hypogymnia physodes on HeLa cell viability and growth. Folia Biol. (Praha) 2014, 60, 89–94. [Google Scholar]
  50. Emsen, B.; Turkez, H.; Togar, B.; Aslan, A. Evaluation of antioxidant and cytotoxic effects of olivetoric and physodic acid in cultured human amnion fibroblasts. Hum. Exp. Toxicol. 2017, 36, 376–385. [Google Scholar] [CrossRef]
  51. Behera, B.C.; Mahadik, N.; Morey, M. Antioxidative and cardiovascular-protective activities of metabolite usnic acid and psoromic acid produced by lichen species Usnea complanata under submerged fermentation. Pharm. Biol. 2012, 50, 968–979. [Google Scholar] [CrossRef] [Green Version]
  52. Nishanth, K.S.; Sreerag, R.S.; Deepa, I.; Mohandas, C.; Nambisan, B. Protocetraric acid: An excellent broad spectrum compound from the lichen Usnea albopunctata against medically important microbes. Nat. Prod. Res. 2015, 29, 574–577. [Google Scholar] [CrossRef] [PubMed]
  53. Carlos, I.Z.; Carli, C.B.A.; Maia, D.C.G.; Benzatti, F.P.; Lopes, F.C.M.; Roese, F.M.; Watanabe, M.; Micheletti, A.C.; dos Santos, L.C.; Vilegas, W.; et al. Immunostimulatory effects of the phenolic compounds from lichens on nitric oxide and hydrogen peroxide production. Rev. Bras. De Farmacogn. 2009, 19, 847–852. [Google Scholar] [CrossRef]
  54. Ranković, B.; Kosanić, M.; Stanojković, T.; Vasiljević, P.; Manojlović, N. Biological Activities of Toninia candida and Usnea barbata Together with Their Norstictic Acid and Usnic Acid Constituents. IJMS 2012, 13, 14707–14722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ranković, B. Lichen Secondary Metabolites: Bioactive Properties and Pharmaceutical Potential; Springer International Publishing: Cham, Switzerland, 2019; ISBN 978-3-030-16814-8. [Google Scholar]
  56. Oettl, S.K.; Gerstmeier, J.; Khan, S.Y.; Wiechmann, K.; Bauer, J.; Atanasov, A.G.; Malainer, C.; Awad, E.M.; Uhrin, P.; Heiss, E.H.; et al. Imbricaric Acid and Perlatolic Acid: Multi-Targeting Anti-Inflammatory Depsides from Cetrelia monachorum. PLoS ONE 2013, 8, e76929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Reddy, R.G.; Veeraval, L.; Maitra, S.; Chollet-Krugler, M.; Tomasi, S.; Dévéhat, F.L.-L.; Boustie, J.; Chakravarty, S. Lichen-derived compounds show potential for central nervous system therapeutics. Phytomedicine 2016, 23, 1527–1534. [Google Scholar] [CrossRef]
  58. Çobanoğlu, G. Evaluation of the Antimicrobial Properties of Some Lichens. South West. J. Hortic. 2010, 6, 153–158. [Google Scholar]
  59. Luo, H.; Yamamoto, Y.; Kim, J.A.; Jung, J.S.; Koh, Y.J.; Hur, J.-S. Lecanoric acid, a secondary lichen substance with antioxidant properties from Umbilicaria antarctica in maritime Antarctica (King George Island). Polar. Biol. 2009, 32, 1033–1040. [Google Scholar] [CrossRef]
  60. Martins, M.C.B.; Lima, M.J.G.D.; Silva, F.P.; Azevedo-Ximenes, E.; Silva, N.H.D.; Pereira, E.C. Cladia aggregata (lichen) from Brazilian northeast: Chemical characterization and antimicrobial activity. Braz. Arch. Biol. Technol. 2010, 53, 115–122. [Google Scholar] [CrossRef] [Green Version]
  61. Verma, N.; Behera, B.C.; Sonone, A.; Makhija, U. Cell Aggregates Derived from Natural Lichen Thallus Fragments: Antioxidant Activities of Lichen Metabolites Developed In Vitro. Nat. Prod. Commun. 2008, 3, 1911–1918. [Google Scholar] [CrossRef] [Green Version]
  62. Honda, N.K.; Pavan, F.R.; Coelho, R.G.; de Andrade Leite, S.R.; Micheletti, A.C.; Lopes, T.I.B.; Misutsu, M.Y.; Beatriz, A.; Brum, R.L.; Leite, C.Q.F. Antimycobacterial activity of lichen substances. Phytomedicine 2010, 17, 328–332. [Google Scholar] [CrossRef]
  63. Manojlovic, N.T.; Vasiljevic, P.J.; Maskovic, P.Z.; Juskovic, M.; Bogdanovic-Dusanovic, G. Chemical Composition, Antioxidant, and Antimicrobial Activities of Lichen Umbilicaria cylindrica (L.) Delise (Umbilicariaceae). Evid. Based Complement. Altern. Med. 2012, 2012, 452431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sun, L.; Sun, S.; Cheng, A.; Wu, X.; Zhang, Y.; Lou, H. In vitro activities of retigeric acid B alone and in combination with azole antifungal agents against Candida albicans. Antimicrob. Agents Chemother. 2009, 53, 1586–1591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Robertsdottir, A.R. Icelandic Herbs and Their Medicinal Uses; North Atlantic Books: Berkeley, CA, USA, 2016; ISBN 978-1-62317-023-3. [Google Scholar]
  66. Paudel, B.; Bhattarai, H.D.; Koh, H.Y.; Lee, S.G.; Han, S.J.; Lee, H.K.; Oh, H.; Shin, H.W.; Yim, J.H. Ramalin, a novel nontoxic antioxidant compound from the Antarctic lichen Ramalina terebrata. Phytomedicine 2011, 18, 1285–1290. [Google Scholar] [CrossRef] [PubMed]
  67. Paudel, B.; Bhattarai, H.D.; Lee, J.S.; Hong, S.G.; Shin, H.W.; Yim, J.H. Antibacterial potential of Antarctic lichens against human pathogenic Gram-positive bacteria. Phytother. Res. 2008, 22, 1269–1271. [Google Scholar] [CrossRef] [PubMed]
  68. Wu, W.; Hou, B.; Tang, C.; Liu, F.; Yang, J.; Pan, T.; Si, K.; Lu, D.; Wang, X.; Wang, J.; et al. (+)-Usnic Acid Inhibits Migration of c-KIT Positive Cells in Human Colorectal Cancer. Evid. Based Complement. Altern. Med. 2018, 2018, 1–13. [Google Scholar] [CrossRef]
  69. Geng, X.; Zhang, X.; Zhou, B.; Zhang, C.; Tu, J.; Chen, X.; Wang, J.; Gao, H.; Qin, G.; Pan, W. Usnic Acid Induces Cycle Arrest, Apoptosis, and Autophagy in Gastric Cancer Cells In Vitro and In Vivo. Med. Sci. Monit. 2018, 24, 556–566. [Google Scholar] [CrossRef] [Green Version]
  70. Bessadottir, M.; Egilsson, M.; Einarsdottir, E.; Magnusdottir, I.H.; Ogmundsdottir, M.H.; Omarsdottir, S.; Ogmundsdottir, H.M. Proton-Shuttling Lichen Compound Usnic Acid Affects Mitochondrial and Lysosomal Function in Cancer Cells. PLoS ONE 2012, 7, e51296. [Google Scholar] [CrossRef]
  71. Zuo, S.; Wang, L.; Zhang, Y.; Zhao, D.; Li, Q.; Shao, D.; Fang, X. Usnic acid induces apoptosis via an ROS-dependent mitochondrial pathway in human breast cancer cells in vitro and in vivo. RSC Adv. 2015, 5, 153–162. [Google Scholar] [CrossRef]
  72. Pyrczak-Felczykowska, A.; Narlawar, R.; Pawlik, A.; Guzow-Krzemińska, B.; Artymiuk, D.; Hać, A.; Ryś, K.; Rendina, L.M.; Reekie, T.A.; Herman-Antosiewicz, A.; et al. Synthesis of Usnic Acid Derivatives and Evaluation of Their Antiproliferative Activity against Cancer Cells. J. Nat. Prod. 2019, 82, 1768–1778. [Google Scholar] [CrossRef]
  73. Emsen, B.; Aslan, A.; Turkez, H.; Joughi, A.; Kaya, A. The anti-cancer efficacies of diffractaic, lobaric, and usnic acid: In vitro inhibition of glioma. J. Cancer Res. Ther. 2018, 14, 941. [Google Scholar] [CrossRef]
  74. Galanty, A.; Koczurkiewicz, P.; Wnuk, D.; Paw, M.; Karnas, E.; Podolak, I.; Węgrzyn, M.; Borusiewicz, M.; Madeja, Z.; Czyż, J.; et al. Usnic acid and atranorin exert selective cytostatic and anti-invasive effects on human prostate and melanoma cancer cells. Toxicol. Vitr. 2017, 40, 161–169. [Google Scholar] [CrossRef] [PubMed]
  75. Koparal, A.T. Anti-angiogenic and antiproliferative properties of the lichen substances (-)—Usnic acid and vulpinic acid. Z. Nat. C J. Biosci. 2015, 70, 159–164. [Google Scholar] [CrossRef] [PubMed]
  76. Kılıc, N.; Derici, M.K.; Buyuk, I.; Aydın, S.S.; Aras, S.; Duman, D.C. Evaluation of in vitro Anticancer Activity of Vulpinic Acid and its Apoptotic Potential Using Gene Expression and Protein Analysis. IJPER 2018, 52, 626–634. [Google Scholar] [CrossRef] [Green Version]
  77. Jeon, Y.-J.; Kim, S.; Kim, J.H.; Youn, U.J.; Suh, S.-S. The Comprehensive Roles of ATRANORIN, A Secondary Metabolite from the Antarctic Lichen Stereocaulon caespitosum, in HCC Tumorigenesis. Molecules 2019, 24, 1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Brandão LF, G.; Alcantara, G.B.; Matos MD, F.C.; Bogo, D.; dos Santos Freitas, D.; Oyama, N.M.; Honda, N.K. Cytotoxic Evaluation of Phenolic Compounds from Lichens against Melanoma Cells. Chem. Pharm. Bull. 2013, 61, 176–183. [Google Scholar] [PubMed] [Green Version]
  79. Alexandrino, C.A.F.; Honda, N.K.; Matos, M.D.F.C.; Portugal, L.C.; Souza, P.R.B.D.; Perdomo, R.T.; Guimarães, R.D.C.A.; Kadri, M.C.T.; Silva, M.C.B.L.; Bogo, D. Antitumor effect of depsidones from lichens on tumor cell lines and experimental murine melanoma. Rev. Bras. De Farmacogn. 2019, 29, 449–456. [Google Scholar] [CrossRef]
  80. Manojlović, N.; Ranković, B.; Kosanić, M.; Vasiljević, P.; Stanojković, T. Chemical composition of three Parmelia lichens and antioxidant, antimicrobial and cytotoxic activities of some their major metabolites. Phytomedicine 2012, 19, 1166–1172. [Google Scholar] [CrossRef]
  81. Liu, Y.-Q.; Hu, X.-Y.; Lu, T.; Cheng, Y.-N.; Young, C.Y.F.; Yuan, H.-Q.; Lou, H.-X. Retigeric Acid B Exhibits Antitumor Activity through Suppression of Nuclear Factor-κB Signaling in Prostate Cancer Cells in Vitro and in Vivo. PLoS ONE 2012, 7, e38000. [Google Scholar] [CrossRef]
  82. Brisdelli, F.; Perilli, M.; Sellitri, D.; Bellio, P.; Bozzi, A.; Amicosante, G.; Nicoletti, M.; Piovano, M.; Celenza, G. Protolichesterinic acid enhances doxorubicin-induced apoptosis in HeLa cells in vitro. Life Sci. 2016, 158, 89–97. [Google Scholar] [CrossRef]
  83. Bessadóttir, M.; Eiríksson, F.F.; Becker, S.; Ögmundsdóttir, M.H.; Ómarsdóttir, S.; Thorsteinsdóttir, M.; Ögmundsdóttir, H.M. Anti-proliferative and pro-apoptotic effects of lichen-derived compound protolichesterinic acid are not mediated by its lipoxygenase-inhibitory activity. Prostaglandins Leukot. Essent. Fat. Acids 2015, 98, 39–47. [Google Scholar] [CrossRef]
  84. Suh, S.-S.; Kim, T.; Kim, J.; Hong, J.-M.; Nguyen, T.; Han, S.; Youn, U.; Yim, J.; Kim, I.-C. Anticancer Activity of Ramalin, a Secondary Metabolite from the Antarctic Lichen Ramalina terebrata, against Colorectal Cancer Cells. Molecules 2017, 22, 1361. [Google Scholar] [CrossRef] [PubMed]
  85. Yang, Y.; Park, S.-Y.; Nguyen, T.T.; Yu, Y.H.; Nguyen, T.V.; Sun, E.G.; Udeni, J.; Jeong, M.-H.; Pereira, I.; Moon, C.; et al. Lichen Secondary Metabolite, Physciosporin, Inhibits Lung Cancer Cell Motility. PLoS ONE 2015, 10, e0137889. [Google Scholar] [CrossRef] [PubMed]
  86. Tatipamula, V.B.; Vedula, G.S.; Sastry, A.V.S. Antarvedisides A-B from Manglicolous Lichen Dirinaria consimilis (Stirton) and their Pharmacological Profile. Asian J. Chem. 2019, 31, 805–812. [Google Scholar] [CrossRef]
  87. Nguyen, T.T.; Yoon, S.; Yang, Y.; Lee, H.-B.; Oh, S.; Jeong, M.-H.; Kim, J.-J.; Yee, S.-T.; Crişan, F.; Moon, C.; et al. Lichen Secondary Metabolites in Flavocetraria cucullata Exhibit Anti-Cancer Effects on Human Cancer Cells through the Induction of Apoptosis and Suppression of Tumorigenic Potentials. PLoS ONE 2014, 9, e111575. [Google Scholar] [CrossRef] [PubMed]
  88. Zugic, A.; Jeremic, I.; Isakovic, A.; Arsic, I.; Savic, S.; Tadic, V. Evaluation of Anticancer and Antioxidant Activity of a Commercially Available CO2 Supercritical Extract of Old Man’s Beard (Usnea barbata). PLoS ONE 2016, 11, e0146342. [Google Scholar] [CrossRef] [Green Version]
  89. Kosanić, M.; Manojlović, N.; Janković, S.; Stanojković, T.; Ranković, B. Evernia prunastri and Pseudoevernia furfuraceae lichens and their major metabolites as antioxidant, antimicrobial and anticancer agents. Food Chem. Toxicol. 2013, 53, 112–118. [Google Scholar] [CrossRef]
  90. Gonçalves, J.P.; Martins, M.C.B.; Buril, M.D.L.L.; Aguiar, J.D.S.; Da Silva, T.G.; Dos Santos Souza, T.G.; Santos, N.P.D.S.; Chagas, C.A.; Pereira, E.C.; Falcão, E.P.D.S.; et al. Antineoplastic Activity and Genotoxicity of Organic Extracts and Barbatic Acid Isolated from the Lichen Cladonia salzmannii Nyl. Int. Arch. Med. 2018, 11, 53. [Google Scholar] [CrossRef]
  91. Barroso Martins, M.C.; Alves Rocha, T.; Santos Silva, T.D.; Pacífico Cavalcanti-Neto, M.; Pereira da Silva Santos, N.; Gonçalves da Silva, T.; Amanajás Aguiar-Junior, F.C.; da Silva Falcão, E.P.; Pereira, E.C.; da Silva, N.H. In Vitro And In Vivo Antineoplastic Activity Of Barbatic Acid. Int. Arch. Med. 2016, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
  92. Yang, Y.; Bae, W.K.; Nam, S.-J.; Jeong, M.-H.; Zhou, R.; Park, S.-Y.; Taş, İ.; Hwang, Y.-H.; Park, M.-S.; Chung, I.J.; et al. Acetonic extracts of the endolichenic fungus EL002332 isolated from Endocarpon pusillum exhibits anticancer activity in human gastric cancer cells. Phytomedicine 2018, 40, 106–115. [Google Scholar] [CrossRef]
  93. Kosanić, M.; Ranković, B.; Stanojković, T. Antioxidant, Antimicrobial, and Anticancer Activity of 3 Umbilicaria Species. J. Food Sci. 2012, 77, T20–T25. [Google Scholar] [CrossRef]
  94. Kosanić, M.; Ranković, B.; Stanojković, T.; Stošić, I.; Grujičić, D.; Milošević-Djordjević, O. Lasallia pustulata lichen as possible natural antigenotoxic, antioxidant, antimicrobial and anticancer agent. Cytotechnology 2016, 68, 999–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kosanić, M.M.; Ranković, B.R.; Stanojković, T.P. Antioxidant, antimicrobial and anticancer activities of three Parmelia species. J. Sci. Food Agric. 2012, 92, 1909–1916. [Google Scholar] [CrossRef] [PubMed]
  96. Ghate, N.B.; Chaudhuri, D.; Sarkar, R.; Sajem, A.L.; Panja, S.; Rout, J.; Mandal, N. An Antioxidant Extract of Tropical Lichen, Parmotrema reticulatum, Induces Cell Cycle Arrest and Apoptosis in Breast Carcinoma Cell Line MCF-7. PLoS ONE 2013, 8, e82293. [Google Scholar] [CrossRef] [PubMed]
  97. Grujičić, D.; Stošić, I.; Kosanić, M.; Stanojković, T.; Ranković, B.; Milošević-Djordjević, O. Evaluation of in vitro antioxidant, antimicrobial, genotoxic and anticancer activities of lichen Cetraria islandica. Cytotechnology 2014, 66, 803–813. [Google Scholar] [CrossRef]
  98. Kosanic, M.; Rankovic, B.; Stanojkovic, T.; Vasiljevic, P.; Manojlovic, N. Biological activities and chemical composition of lichens from Serbia. EXCLI J. 2014, 13, 1226–1238. [Google Scholar]
  99. Kumar, J.; Dhar, P.; Tayade, A.B.; Gupta, D.; Chaurasia, O.P.; Upreti, D.K.; Arora, R.; Srivastava, R.B. Antioxidant Capacities, Phenolic Profile and Cytotoxic Effects of Saxicolous Lichens from Trans-Himalayan Cold Desert of Ladakh. PLoS ONE 2014, 9, e98696. [Google Scholar] [CrossRef]
  100. Ari, F.; Aztopal, N.; Oran, S.; Bozdemir, S.; Celikler, S.; Ozturk, S.; Ulukaya, E. Parmelia sulcata Taylor and Usnea filipendula Stirt induce apoptosis-like cell death and DNA damage in cancer cells. Cell Prolif. 2014, 47, 457–464. [Google Scholar] [CrossRef]
  101. Ari, F.; Celikler, S.; Oran, S.; Balikci, N.; Ozturk, S.; Ozel, M.Z.; Ozyurt, D.; Ulukaya, E. Genotoxic, cytotoxic, and apoptotic effects of Hypogymnia physodes (L.) Nyl. on breast cancer cells: GENOTOXIC, CYTOTOXIC, AND APOPTOTIC EFFECTS OF HYPOGYMNIA. Environ. Toxicol. 2014, 29, 804–813. [Google Scholar] [CrossRef]
  102. Coskun, Z.M.; Ersoz, M.; Acikgoz, B.; Karalti, I.; Cobanoglu, G.; Sesal, C. Anti-Proliferative and Apoptotic Effects of Methanolic Extracts from Different Cladonia Species on Human Breast Cancer Cells. Folia Biol. (Praha) 2015, 61, 97–103. [Google Scholar]
  103. Basile, A.; Rigano, D.; Loppi, S.; Di Santi, A.; Nebbioso, A.; Sorbo, S.; Conte, B.; Paoli, L.; De Ruberto, F.; Molinari, A.; et al. Antiproliferative, Antibacterial and Antifungal Activity of the Lichen Xanthoria parietina and Its Secondary Metabolite Parietin. IJMS 2015, 16, 7861–7875. [Google Scholar] [CrossRef]
  104. Ari, F.; Ulukaya, E.; Oran, S.; Celikler, S.; Ozturk, S.; Ozel, M.Z. Promising anticancer activity of a lichen, Parmelia sulcata Taylor, against breast cancer cell lines and genotoxic effect on human lymphocytes. Cytotechnology 2015, 67, 531–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Shrestha, G.; El-Naggar, A.M.; Clair, L.L.S.; O’Neill, K.L. Anticancer Activities of Selected Species of North American Lichen Extracts: ANTICANCER ACTIVITIES OF LICHEN. Phytother. Res. 2015, 29, 100–107. [Google Scholar] [CrossRef] [PubMed]
  106. De Jesus, E. Antibacterial, antioxidant, and cytotoxic activities of the corticolous lichens Canoparmelia aptata, Pannaria sp., and Parmotrema gardneri collected from Mt. Banahaw, Quezon, Philippines. CREAM 2016, 6, 173–183. [Google Scholar] [CrossRef]
  107. Açikgöz, B.; Karalti, I.; Ersöz, M.; Coşkun, Z.M.; Cobanoğlu, G.; Sesal, C. Screening of antimicrobial activity and cytotoxic effects of two Cladonia species. Z. Naturforsch. C. J. Biosci. 2013, 68, 191–197. [Google Scholar] [CrossRef]
  108. Felczykowska, A.; Pastuszak-Skrzypczak, A.; Pawlik, A.; Bogucka, K.; Herman-Antosiewicz, A.; Guzow-Krzemińska, B. Antibacterial and anticancer activities of acetone extracts from in vitro cultured lichen-forming fungi. BMC Complement. Altern. Med. 2017, 17, 300. [Google Scholar] [CrossRef]
  109. Ersoz, M.; Coskun, Z.M.; Acikgoz, B.; Karalti, I.; Cobanoglu, G.; Cesal, C. In vitro evaluation of cytotoxic, anti-proliferative, anti-oxidant, apoptotic, and anti-microbial activities of Cladonia pocillum. Cell. Mol. Biol. (Noisy-Le-Grand) 2017, 63, 69. [Google Scholar] [CrossRef]
  110. Delebassée, S.; Mambu, L.; Pinault, E.; Champavier, Y.; Liagre, B.; Millot, M. Cytochalasin E in the lichen Pleurosticta acetabulum. Anti-proliferative activity against human HT-29 colorectal cancer cells and quantitative variability. Fitoterapia 2017, 121, 146–151. [Google Scholar] [CrossRef]
  111. Sun, Y.; Li, J.; Zhang, Y.; Tu, Y.; Huang, C.; Tao, J.; Yang, M.; Yang, L. The Polysaccharide Extracted from Umbilicaria esculenta Inhibits Proliferation of Melanoma Cells through ROS-Activated Mitochondrial Apoptosis Pathway. Biol. Pharm. Bull. 2018, 41, 57–64. [Google Scholar] [CrossRef] [Green Version]
  112. Kosanić, M. EXTRACTS OF FIVE CLADONIA LICHENS AS SOURCES OF BIOLOGICALLY ACTIVE COMPOUNDS. Farmacia 2018, 66, 644–651. [Google Scholar] [CrossRef]
  113. Ozturk, S.; Erkisa, M.; Oran, S.; Ulukaya, E.; Celikler, S.; Ari, F. Lichens exerts an anti-proliferative effect on human breast and lung cancer cells through induction of apoptosis. Drug Chem. Toxicol. 2019, 1–9. [Google Scholar] [CrossRef]
  114. Lagarde, A.; Millot, M.; Pinon, A.; Liagre, B.; Girardot, M.; Imbert, C.; Ouk, T.S.; Jargeat, P.; Mambu, L. Antiproliferative and antibiofilm potentials of endolichenic fungi associated with the lichen Nephroma laevigatum. J. Appl. Microbiol. 2019, 126, 1044–1058. [Google Scholar] [CrossRef] [PubMed]
  115. Nugraha, A.S.; Pratoko, D.K.; Damayanti, Y.D.; Lestari, N.D.; Laksono, T.A.; Addy, H.S.; Untari, L.F.; Kusumawardani, B.; Wangchuk, P. Antibacterial and Anticancer Activities of Nine Lichens of Indonesian Java Island. J. Biol. Act. Prod. Nat. 2019, 9, 39–46. [Google Scholar] [CrossRef]
  116. Song, Y.; Dai, F.; Zhai, D.; Dong, Y.; Zhang, J.; Lu, B.; Luo, J.; Liu, M.; Yi, Z. Usnic acid inhibits breast tumor angiogenesis and growth by suppressing VEGFR2-mediated AKT and ERK1/2 signaling pathways. Angiogenesis 2012, 15, 421–432. [Google Scholar] [CrossRef] [PubMed]
  117. Ebrahim, H.Y.; Akl, M.R.; Elsayed, H.E.; Hill, R.A.; El Sayed, K.A. Usnic Acid Benzylidene Analogues as Potent Mechanistic Target of Rapamycin Inhibitors for the Control of Breast Malignancies. J. Nat. Prod. 2017, 80, 932–952. [Google Scholar] [CrossRef] [PubMed]
  118. Mammadov, R.; Suleyman, B.; Altuner, D.; Demirci, E.; Cetin, N.; Yilmaz, A.; Baykal, H.; Alpcan, H.; Turumtay, E.A.; Suleyman, H. Effect of ethyl acetate extract of usnea longissima on esophagogastric adenocarcinoma in rats. Acta Cir. Bras. 2019, 34, 3. [Google Scholar] [CrossRef] [PubMed]
  119. Poornima, S. Evaluation of Anti-Cancer Properties of Lichens Using <i>Albino wistar</i> Rats as an Animal Model. CRJ 2016, 4, 84. [Google Scholar]
  120. Karagoz, I.D.; Ozaslan, M.; Guler, I.; Uyar, C.; Yalim, T.; Kazanci, U.; Aslan, A.; Cakir, A. In vivo Antitumoral Effect of Diffractaic Acid from Lichen Metabolites on Swiss Albino Mice with Ehrlich Ascites Carcinoma: An Experimental Study. Int. J. Pharmacol. 2014, 10, 307–314. [Google Scholar]
  121. Shukla, V.; Joshi, G.P.; Rawat, M.S.M. Lichens as a potential natural source of bioactive compounds: A review. Phytochem. Rev. 2010, 9, 303–314. [Google Scholar] [CrossRef]
  122. Golubnitschaja, O.; Baban, B.; Boniolo, G.; Wang, W.; Bubnov, R.; Kapalla, M.; Krapfenbauer, K.; Mozaffari, M.S.; Costigliola, V. Medicine in the early twenty-first century: Paradigm and anticipation—EPMA position paper 2016. EPMA J. 2016, 7, 23. [Google Scholar] [CrossRef] [Green Version]
  123. Abotaleb, M.; Kubatka, P.; Caprnda, M.; Varghese, E.; Zolakova, B.; Zubor, P.; Opatrilova, R.; Kruzliak, P.; Stefanicka, P.; Büsselberg, D. Chemotherapeutic agents for the treatment of metastatic breast cancer: An update. Biomed. Pharmacother. 2018, 101, 458–477. [Google Scholar] [CrossRef]
Figure 1. The role of lichens in the modulation of molecular mechanisms associated with cancer.
Figure 1. The role of lichens in the modulation of molecular mechanisms associated with cancer.
Biomolecules 10 00087 g001
Table 1. Secondary metabolites of lichens, their origin, chemical structure and activities.
Table 1. Secondary metabolites of lichens, their origin, chemical structure and activities.
OriginChemical StructureActivitiesReferences
Usnic acid (UA)
Biomolecules 10 00087 i001
Usnea diffracta, Cladonia arbuscula,2,6-diacetyl-7,9-dihydroxy-8,9b-dimethyl-1,3antimicrobial, antiprotozoal, antiviral, antiproliferate, anti-inflammatory, analgesic, antipyretic [28,29]
CAS ID (125-46-2)
Alectoria samentosa,
Flavocetraria nivalis,
Alectoria ochroleuca,
Usnea florida
Diffractaic acid (DA)
Biomolecules 10 00087 i002
Usnea longissimi, Usnea subcavata,4-[(2,4-dimethoxy-3,6-dimethylbenzoyl)oxy]-2-hydroxy-3,6-dimethylbenzoic acidantioxidant, gastroprotective, analgesic, antiviral, [38,39,40]
Protousnea magellanicaCAS ID (436-32-8)
Lobaric acid (LA)
Biomolecules 10 00087 i003
Stereocaulon alpinum, Cladonia sp., Stereocaulon sasakii3-hydroxy-9-methoxy-6-oxo-7-(1-oxopentyl)-1-pentyl-2-benzo[b][1,4]benzodioxepincarboxylic acidantibacterial, antioxidant, antimitotic[41,42,43]
CAS ID (522-53-2)
Atranorin (ATR)
Biomolecules 10 00087 i004
Parmelia sulcate, Parmotrema stuppeum, Stereocaulon alpinum, Physcia aipolia3-hydroxy-4-methoxycarbonyl-2,5-dimethylphenylantimicrobial, antiprotozoal, antiviral, antifungal, antioxidant[44,45]
CAS ID (479-20-9)
Vulpinic acid (VA)
Biomolecules 10 00087 i005
Letharia vulpinamethyl (2E)-2-(3-hydroxy-5-oxo-4-phenylfuran-2-ylidene)-2-phenylacetateantiproliferative, antimicrobial, antiangiogenic, [46,47]
CAS ID (73622-57-8)
Physodic acid (PA)
Biomolecules 10 00087 i006
Hypogymnia physodes3,9-dihydroxy-6-oxo-7-(2-oxoheptyl)-1-pentylbenzo[b][1,4]benzodioxepine-2-carboxylic acidantimicrobial, antioxidant, immunoprotective[48,49]
CAS ID (84-24-2)
Olivetoric acid (OA)
Biomolecules 10 00087 i007
Pseudevernia furfuracea4-[2,4-dihydroxy-6-(2-oxoheptyl)benzoyl]oxy-2-hydroxy-6-pentylbenzoic acidantimicrobial, antioxidant[48,50]
CAS ID (491-72-5)
Psoromic acid (PSA)
Biomolecules 10 00087 i008
Usnea camplanata10-formyl-9-hydroxy-3-methoxy-4,7-dimethyl-6-oxobenzo[b][1,4]benzodioxepine-1-carboxylic acidcardioprotective[51]
CAS ID (7299-11-8)
Protocetraric acid (PrA)
Biomolecules 10 00087 i009
Parmelia caperata, Usnea albopunctata, Parmelia saxatilis, Parmelia sulcate10-formyl-3,9-dihydroxy-4-(hydroxymethyl)-1,7-dimethyl-6-oxobenzo[b][1,4]benzodioxepine-2-carboxylic acidantimicrobial, immunostimulatory [52,53]
CAS ID (489-51-0)
Norstictic acid (NA)
Biomolecules 10 00087 i010
Toninia candida1,3-Dihydro-1,4,10-trihydroxy-5,8-dimethyl-3,7-dioxo-7H-isobenzofuro(4,5-b)(1,4)benzodioxepin-11-carboxaldehydeantioxidant, antibacterial[54]
CAS ID (571-67-5)
Divaricatic acid (DiA)
Biomolecules 10 00087 i011
Evernia mesomorpha2-hydroxy-4-[(2-hydroxy-4-methoxy-6-propylbenzoyl)oxy]-6-propylbenzoic acidantimicrobial, antioxidant[55]
CAS ID (491-62-3)
Perlatolic acid (PeA)
Biomolecules 10 00087 i012
Cetrelia monachorum2-Hydroxy-4-((2-hydroxy-4-methoxy-6-pentylbenzoyl)oxy)-6-pentylbenzoic acidanti-inflammatory, anti-neurodegenerative[56,57]
CAS ID (529-47-5)
Caperatic acid (CA)
Biomolecules 10 00087 i013
Platismatia glauca2-hydroxy-2-(2-methoxy-2-oxoethyl)-3-tetradecylbutanedioic acidfungitoxic[58]
CAS ID (29227-64-3)
Lecanoric acid (LeA)
Biomolecules 10 00087 i014
Usnea subvacata Motyka, Parmotrema stuppuem, Parmotrema tinctorum and Parmotrema grayana4-(2,4-dihydroxy-6-methylbenzoyl)oxy-2-hydroxy-6-methylbenzoic acidantioxidant[59]
CAS ID (480-56-8)
Barbatic acid (BA)
Biomolecules 10 00087 i015
Usnea longissima2-hydroxy-4-(2-hydroxy-4-methoxy-3,6-dimethylbenzoyl)oxy-3,6-dimethylbenzoic acidantioxidant, antimicrobial[60,61]
CAS ID (17636-16-7)
Barbatolic acid (BrA)
Biomolecules 10 00087 i016
Bryoria capillaris3-formyl-6-[(3-formyl-2,4-dihydroxy-6-methylbenzoyl)oxymethyl]-2,4-dihydroxybenzoic acidantimicrobial [35]
CAS ID (529-50-0)
Lobastin (LOB)
Biomolecules 10 00087 i017
Stereocaulon alpnum3-[[(1Z)-1-butylidene-6-methoxy-3-oxo-2-benzofuran-4-yl]oxy]-4,6-dihydroxy-2-pentylbenzoic acidantibacterial, antioxidant[30]
Hypostictic acid (HA)
Biomolecules 10 00087 i018
Pseudoparmelia sphaerospora(1,4-dihydroxy-10-methoxy-5,8,11-trimethyl-1H-benzo[e]furo[3′,4′:3,4]benzo[b][1,4]dioxepine-3,7-dione)antimicrobial [62]
Salazinic acid (SA)
Biomolecules 10 00087 i019
Parmelia caperata5,13,17-trihydroxy-12-(hydroxymethyl)-7-methyl-9,15-dioxo-2,10,16-trioxatetracyclo[9.7.0.03,8.014,18]octadeca-1(11),3(8),4,6,12,14(18)-hexaene-4-carbaldehydeantibacterial, antifungal, antioxidant, antiviral[55,63]
CAS ID (521-39-1)
Retigeric acid B (RA-B)
Biomolecules 10 00087 i020
Lobaria kurokawae(3R,3aR,5aR,5bR,7aR,8S,9R,10R,11aR,13aS,13bR)-9,10-dihydroxy-3a,5a,8,13a-tetramethyl-3-propan-2-yl-1,2,3,4,5,5b,6,7,7a,9,10,11,13,13b-tetradecahydrocyclopenta[a]chrysene-8,11a-dicarboxylic acidantifungal[64]
CAS ID (38327-77-4)
Protolichesterinic acid (PLA)
Biomolecules 10 00087 i021
Cornicularia aculeate, Centraria islandica4-methylidene-5-oxo-2-tridecyloxolane-3-carboxylic acidantibacterial, anti-inflammatory [65]
CAS ID (1448-96-0)
Ramalin (RAM)
Biomolecules 10 00087 i022
Ramalina terebrataγ-glutamyl-N′-(2-hydroxyphenyl)hydrazideantioxidant, antibacterial[66,67]
Physciosporin (PHY)
Biomolecules 10 00087 i023
Pseudocyphellaria granulate, Pseudocyphellaria coriaceamethyl 8-chloro-10-formyl-3,9-dihydroxy-1,4,7-trimethyl-6-oxobenzo[b][1,4]benzodioxepine-2-carboxylateantiproliferative[20]
Sekikaic acid (SeA)
Biomolecules 10 00087 i024
Cladonia sp., Ramalina roesleri2-hydroxy-3-(2-hydroxy-4-methoxy-6-propylbenzoyl)oxy-4-methoxy-6-propylbenzoic acidantioxidant, antibacterial, antidiabetic[18]
CAS ID (607-11-4)
Abbreviations: ATR, atranorin; BA, barbatic acid; BrA, barbatolic acid; CA, caperatic acid; DA, diffractaic acid; DiA, divaricatic acid; GA, gyrophoric acid; HA, hypostictic acid; LA, lobaric acid; LeA, lecanoric acid; Li, lichexanthone; LOB, lobastin; NA, nortictic acid; OA, olivetoric acid; PA, physodic acid; PeA, perlatolic acid; PLA, protolichesterinicc acid; PrA, protocetraric acid; PSA, psoromic acid; PU, potassium usnate; RA-B, retigeric acid B; RAM, ramalin; SA, salazanic acid; SeA, sekikaic acid; UA, usnic acid; VA, vulpinic acid.
Table 2. Anticancer potential of isolated lichen compounds.
Table 2. Anticancer potential of isolated lichen compounds.
Lichen Acid/SpeciesCell LinesEffectsReference
UACaCo2, HepG2, Hep2C, RD, Wehi,↑ cytotoxicity of CaCo2 (IC50 7.05 μM), HepG2 (IC50 15.4 μM), Hep2C (IC50 21.8 μM), RD (IC50 22.9 μM), Wehi (IC50 15.8 μM)[29]
↑ Bax ↓ Bcl-2 ↓ p53
HCT116, LS174↓ SCF-induced proliferation and migration of HCT116 and LS174 (c-KIT+)[68]
↑ autophagy of HCT116 (via ↓ mTOR)
↓ level of phosphorylated PKC-A, c-KIT of HCT116
BGC823, SGC7901↓ proliferation + G0/G1 and G2/M arrest of BGC823 (IC50 236.55 μM) and SGC7901 (IC50618.82 μM)[69]
→ apoptosis, ↑ autophagy
↑ Bax/Bcl-2 ratio
↑ caspase-3, ↑ PARP
MCF-7, MDA-MB-231, BT-474MDA-MB-231: 67 UA-responsive miRNAs[37]
BT-474: 15 UA-responsive miRNAs
MCF-7: 8 UA-responsive miRNAs
HepG2, SNU-449→ apoptosis and autophagy[27]
G0/G1, G2/M arrest
A549→ apoptosis[28]
↓ cell number
↓ proliferation
↓ expression CDK4, CDK6, cyclin D1
↑ expression of p21/cip1 protein
T47D and MCF-7Formation of autophagosome (H+ shuttling in mitochondria and lysosomes)[70]
Capan-2
MCF-7→ apoptosis[71]
A549, H460, H1650 and H1975↓ motility of A549[19]
↓ invasion of H1650 and H1975 (↓β-catenin-mediated TOPFLASH and KITENIN-mediated AP-1 activity)
↓ expression of CD44, c-myc and Cyclin D1 in all cell lines
↓ GTP-Rac1 and RhoA
Synthetic derivatives of UAMCF-7, PC-3, HeLaDerivatives 2a, 2b:[72]
↓ proliferation of PC-3, MCF-7 (IC50 value 3 μM), HeLa (IC50 1 μM)
G0/G1 arrest + → apoptosis of MCF-7
Activation of cytoplasmic vacuolisation
All active derivatives:
G0/G1 arrest + ↓ fraction in S and G2/M phase of HeLa
PUHCT116, DLD1, SW480, HT29, SW620, Caco2, COLO320, CT26↑ cytotoxicity (lower IC50 than UA, except of SW480 and CT26 cells)[34]
↓ invasion of Caco2 and HCT116
↓ Caco2 motility (↓CAPN1, CDC42, CFL1, IGF1, WASF1, WASL)
DAU87MG-GBM, PRCCLA: ↑cytotoxicity of GBM and PRCC (IC50 of LA, DA and UA 9.08, 122.26, 132.69 mg/L in PRCC and 5.77, 35.67 and 41.55 mg/L in U87MG)[73]
LA
UA
UAHTB-140, DU-145, PC-3proliferation, ↓ migration, ↓ actin organization[74]
ATR
UAHepG2, HUVEC, NS2OYUA: ↑ cytotoxicity[75]
VA↓ proliferation of NS2OY after VA- treatment
↑ antiangiogenic effect
VAMCF-7, MDA-MB-231, BT-474, SK-BR-3, MCF-12 A↑ cytotoxicity[47]
apoptosis
↑ P53 in SK-BR-3 (vs MCF-12A)
CaCo2, HepG2 and Hep2C, RD, Wehi, L929, Vero↑ cytotoxicity of CaCo2 (IC50 13.7 μM), HepG2 (IC50 23.8 μM), Hep2C (IC50 25.3 μM), RD (IC50 34.4 μM), Wehi (IC50 38.6 μM)[76]
↓ growth (HepG2, CaCo2, Hep2C, RD, Wehi)
↑ Bax and p53 (HepG2, CaCo2, Hep2C, RD, Wehi)
↓ Bcl-2 (HepG2, CaCo2, Hep2C, RD, Wehi)
ATR4T1, NMuMG↓ clonogenic potential of 4T1 cells; → caspase-3, PARP cleavage, depletion of Bcl xL (4T1)[45]
ATRSK-Hep1, Huh-7, SNU-182↓ cancer cells growth (concentration, >10 μg/mL[77]
↑ necrotic cell death, ↓ migration and invasion (Sk-Hep1, Huh-7)
G2/M arrest (SK-Hep1)
ATRA375PA (concentration, 6.25–50 μM): ↓ A375, ↑ apoptosis[17]
GAATR and GA (high concentrations): ↓ A375
OAPRCC↑ cytotoxicity[43]
PAU87MG
PSA
ATRUACC-62↑ cytotoxicity (PrA, NA, PsA, DiA, PeA against UACC-62)[78]
PrANIH/3T3↑ selectivity of PrA, NA, PsA, DiA, PeA for UACC-62
UAB16-F10↑ effectivity of NA and DIA against B16-F10
DA
Li
NA
PeA
DiA
PSA
PAHCT116, DLD-1, HaCaTCA: strongest cytotoxic (concentration, 100 μM)[26]
CACA (HCT116, DLD-1): ↓ β-catenin regulated expression of Axin2, ↓ migration
LeACA (HCT116): ↓ Axin2
CA + PA (HaCat): ↓ MMP7, ↓ survivin
BAHeLa, A549, MCF-7, DU-145, HEK293↓ HeLa (IC50 3.2 μg/mL), A549 (IC50 1.8, 3.2 μg/mL), MCF-7 (IC50 3.2 μg/mL), DU-145 (IC50 9.0 μg/mL)[38]
BA (concentration, 1 μM):
G0/G1 arrest, ↑ apoptosis, ↑ caspase-3 activity, PARP cleavage, annexin V staining and chromatin condensation (A549)
BrAT-47D, HCC1428, HUVECsendothelial tube formation[35]
migration
LAHeLa, HCT116viability (IC50 50 μM)[30]
LOB↓ proliferation
→ G2/M arrest
→ apoptosis (↑ Annexin V-positivity and PARP cleavage,
↓ Bcl-2)
HA, SAB16-F10, 786-0, HT-29, K562HA (K562, B16-F10, 786-0): ↓ proliferation[79]
SA (K562, HT-29, B16-F10): ↓ proliferation
SA, PAFemX, LS174↑ cytotoxicity[80]
RA-BPC-3, DU145→ apoptosis[81]
↓ expression of Bcl-2, Bcl-XL, cyclin D1, and survivin
PLASK-BR-3, T-47D↓ proliferation of SK-BR-3[31]
↑ expression of fatty acid synthase
↓ expression of HER2
↓ ERK1/2 and AKT signalling
HeLa, SH-SY5Y, K562PLA with doxorubicin: synergic cytotoxic effect (HeLa)[82]
RPMI 8226 and U266↓ proliferation[83]
AsPC-1→ cell arrest of AsPC-1
→ apoptosis (RPMI 8226, U266)
↑ cytotoxicity of U266 (IC50 3.5μg/mL), AsPC-1 (IC50 3.5μg/mL), RPMI 8226 (IC501.8 μg/mL)
RAMHCT116↓ proliferation (concentration, 50 - 100 μg/mL)[84]
↑ G2/M arrest (via ↑ TP53, ↑ p21, ↓ cyclin B1, ↓ CDK1)
→ apoptotic cells (concentration, 100 μg/mL)
↓ wound healing, invasion, migration
PHYCaco2, DLD1, HCT116, SW620, CT26↑ cytotoxicity of CT26 (IC50 11.5 μg/mL), SW620 (IC50 12.6 μg/mL), Caco2 (IC50 13.3 μg/mL), HCT116 (IC50 19.8 μg/mL) and DLD1 (IC50 24.9 μg/mL)[22]
apoptosis (PHY at toxic concentrations)
↓ migration, invasion, colony formation (PHY at non-toxic doses)
↓ downstream transcription factors and/or target genes of EM
↓ KITENIN, ↓ β-catenin
↓ actin-based cell motility
A549, H1650, H1975↓ migration[85]
↓ invasion
antarAMCF-7, HeLa, A549, NHMEAntarB (concentration, 30 μg/mL): stronger growth inhibition (HeLa, MCF-7) vs doxorubicin (concentration, 10 μg/mL)[86]
antarBAntarB and 2′-O-methyl DiA: ↓ proliferation of A549 (IC50 values of 22.5 and 27.5 μg/mL, respectively)
SeAAll metabolites: ↓ toxicity against NHME vs cancer cells
ATR
DiA
2′-O-methyl DiA
Explanatory notes: ↑ increase; ↓ decrease; → induction; + plus/and. Abbreviations: 2′-O-methyl DiA, 2′-O-methyl divaricatic acid; antarA, antarvediside A; antarB, antarvediside B; ATR, atranorin; BA, barbatic acid; BrA, barbatolic acid; CA, caperatic acid; DA, diffractaic acid; DiA, divaricatic acid; GA, gyrophoric acid; HA, hypostictic acid; LA, lobaric acid; LeA, lecanoric acid; Li, lichexanthone; LOB, lobastin; NA, nortictic acid; OA, olivetoric acid; PA, physodic acid; PeA, perlatolic acid; PLA, protolichesterinicc acid; PrA, protocetraric acid; PSA, psoromic acid; PU, potassium usnate; RA-B, retigeric acid B; RAM, ramalin; SA, salazanic acid; SeA, sekikaic acid; UA, usnic acid; VA, vulpinic acid.
Table 3. An overview of anticancer efficacy of isolated lichen compounds in combination with lichen extracts.
Table 3. An overview of anticancer efficacy of isolated lichen compounds in combination with lichen extracts.
Lichen Acid/SpeciesCell LinesEffectsReference
UAHT29, AGS, A549, CWR22Rv-1↑ selective cytotoxicity (acetone extract and UA)[87]
Extract of Flavocetraria cucullata↓ tumorigenesis and motility
↓ EMT and Akt phosphorylation
↑ anticancer activity of extract vs. UA
Extract of Toninia candida,FemX, LS174↑ cytotoxicity[54]
Extract of Usnea barbata→ apoptosis after UA treatment
NA, UA
SCEB16, C6, HaCaT↑ cytotoxicity of B16 (IC50 31.21 μg/mL) and C6 (IC50 43.40 μg/mL)[88]
↑ apoptosis and/or autophagy in B16 and C6
Low toxicity against HaCaT
Acetone extract of Evernia prunastriFemX, LS174PA: ↑ cytotoxicity LS 174 and FemX[89]
Acetone extracts of Pseudoevernia furfuraceae↓ FemX and LS174: S and G2/M arrest
PA
PAMCF-7, T47D, MDA-MB-231, MCF-10APA: ↑ cytotoxicity of MCF-7 (IC50 72.4 μg/mL), T47D (IC50 75.4 IC50 μg/mL), MDA-MB-231 (IC50 93.9 μg/mL)[24]
Acetone extract of Hypogymnia physodes
Ethanol extract of Usnea strigosaMD-MB-231, MDA-MB-468, MCF-7, T-47D, BT-474, SK-BR-3, MCF-10AU. strigosa extracts: ↑ cytotoxicity of MD-MB-231 (IC50 3.7 μg/mL) MDA-MB-468 (IC50 4.5 μg/mL), MCF-7 IC50 6.4 μg/mL), T-47D (IC50 9.6 μg/mL), BT-474 (IC50 7.9 μg/mL), SK-BR-3 IC50 7.5 μg/mL)[33]
NANA: MD-MB-231(IC50 14.9 μg/mL), MDA-MB-468 (IC50 17.3 μg/mL)
↓ proliferation
↓ migration of MDA-MB-468
↓ invasion of MDA-MB-231
↓ c-Met, STAT3, paxillin/Rac-1and FAK phosphorylation in MDA-MB-231
Acetone extracts of Melanelia subaurifera and Melanelia fuliginosaHeLa, A549, LS174, MRC5Melanelia subaurifera extract: ↑ cytotoxicity of HeLa (IC50 9.88 μg/mL) A549 (IC50 31.25 μg/mL), LS174 (IC50 31.64 μg/mL);[25]
LeAMelanelia fuliginosa extract: HeLa (IC50 45.24 μg/mL) A549 (IC50 125.276 μg/mL), LS174 (IC50 142.87 μg/mL);
2′-O-MA
Extract of Cladonia salzmanniiRAW 264.7, NCI-H292, HEp-2, MCF-7, HL-60Ether extract: cytotoxicity of HL-60 (IC50 3.59 μg/mL), HEP-2 (IC50 26.75 μg/mL), NCI-H292 (IC50 29.91 μg/mL), RAW-264.7 (IC50 36.54 μg/mL)[90]
Acetone extract: cytotoxicity of MCF-7 (7.55 μg/mL) and NCI-H292 (16.60 μg/mL)
BACytotoxicity of HEP-2 (IC50 15.79 μg/mL), MCF-7 (IC50 18.28 μg/mL), RAW-264.7 (IC50 20.79 μg/mL)
BAHEp-2, NCI-H292, KBCytotoxicity of HEp-2 (IC50 6.25 μg/mL)[91]
Acetone extracts ofA549migration (concentration, 10 μg/mL)[20]
Everniastrum vexans
ATR↑ cytotoxicity (concentration, >5 μg/mL)
↓ β-catenin-mediated TOPFLASH activity (via ↓ nuclear import of β-catenin, ↓ c-jun/AP-1)
↓ mRNA expression of KITENIN
↑ KAI1 mRNA
↓ GTP-Cdc42, GTP-RhoA, STAT proteins
Extracts of endolichenic fungus EL002332AGS, TMK-1, CT26↑ cytotoxicity (on AGS and CT26)[92]
EL002332 + docetaxel: synergistic effects (on AGS and TMK-1)
myCapoptosis (caspase activation, Bcl-2 family regulation)
Explanatory notes: ↑ increase; ↓ decrease; → induction; + plus/and Abbreviations: 2′-O-MA, 2′-O-methyl anziaic acid; ATR, atranorin; BA, barbatic acid; LeA, lecanoric acid; NA, nortictic acid; PA, physodic acid; SCE, supercritical CO2 extract of old´s man beard; UA, usnic acid.
Table 4. Anticancer potential of lichen extracts.
Table 4. Anticancer potential of lichen extracts.
Lichen Acid/speciesCell LinesEffectsReference
Extract of Umbilicaria crustulosaFemX, LS174↑ cytotoxicity (all tested extracts)[93]
Extract of Umbilicaria cylindrica
Extract of Umbilicaria polyphylla
Methanol extract of Lasallia pustulataFemX, LS174↑ cytotoxicity: FemX (IC50 46.66 μg/mL); LS174 (IC50 71.71 μg/mL)[94]
Extract of Parmelia caperataFemX, LS174↑ cytotoxicity (all tested extracts)[95]
Extract of Parmelia sulcata
Extract of Parmelia saxatilis
Extract of Parmotrema reticulatumMCF-7, A549, WI-38↑ cytotoxicity[96]
→ cell cycle arrest
Methanol extract of Cetraria islandicaFemX, LS174↑ cytotoxicity: FemX (IC50 22.68 μg/mL); LS174 (IC50 33.74 μg/mL)[97]
Acetone extract of Parmelia arseneanaFemX, LS174, A549, K562↑ cytotoxicity (IC50 11.61–47.06 μg/mL)[98]
Water extracts of Dermatocarpon vellereum, Umbilicaria vellea, Xanthoria elegans and Melanelia disjunctaHepG2, RKO↑ cytotoxicity (all extracts, mainly L. alphoplaca and M. disjuncta)[99]
Methanol extracts of Melanelia disjuncta, Lobothallia alphoplaca and Xanthoparmelia stenophylla
Methanol extracts of Parmelia sulcata Taylor and Usnea filipendula StirtA549, PC-3, Hep3B↑ cytotoxicity (IC50 32.9–98.5 μg/mL)[100]
Rat glioma C6
→ genotoxicity
→ apoptotosis
Methanol extract of Hypogymnia physodesMCF-7, MDA-MB-231↑ anticancer and/or apoptosis-inducing (low concentration) effect[101]
↑ genotoxicity (high concentration)
Methanol extracts of Cladonia rangiformis and Cladonia convoluteMCF-7→ apoptosis[102]
↓ proliferation
↑ cytoxicity
Acetone extract of Xanthoria parietinaMCF-7, MDA-MB-231↓ proliferation[103]
↓ cell cycle
↑ apoptosis
Metanol extract of Parmelia sulcataMCF-7, MDA-MB-231↑ cytotoxicity MCF-7 (IC5039.1 μg/mL); MDA-MB-231 (IC50 16.5 μg/mL)[104]
→ apoptosis
Extracts of Xanthoparmelia chlorochroa and Tuckermannopsis ciliarisHuman Burkitt’s lymphoma (Raji)→ apoptosis[105]
→ cell arrest
↑ p53 expression
Acetone extracts of AGS, A549, MDCKP. gardneri: ↑ cytotoxicity of AGS (IC50 39.1 μg/mL), A549 (IC50 20.24 μg/mL), MDCK (IC50 66.35 μg/mL); Canoparmelia aptata: AGS (IC50 167.9 μg/mL), A549 (IC50 200 μg/mL)[106]
Parmotrema gardneri, Pannaria sp., and Canoparmelia aptata
Extract of Cladonia rangiformisMCF-7↑ cytotoxicity[107]
Extract of Cladonia convoluta
Caloplaca pusilla (on G-LBM medium)HeLa, MCF-7, PC-3↓ viability of MCF-7 (IC50 7.29 μg/mL), PC-3 (IC50 7.96 μg/mL), HeLa (IC50 6.57 μg/mL)[108]
→ apoptosis
Xanthoria parietina (on PDA and G-LBM)↓ cancer cell viability of MCF-7, HeLa (IC50 about 8 μg/mL)
Methanol extract of Cladonia pocillumonMCF-7apoptosis (concentration-dependent)[109]
Acetone extract of Pleurosticta acetabulumHT-29↑ cytotoxicity (IC50 after 48 h, 6 μg/mL) [110]
(cytochalasin E) ↓ proliferation
→ apoptosis
Polysaccharide from Umbilicaria esculentaA875, A375, HUVEC↑ cytotoxicity of A875 and A375 [111]
↑ Annexin-V positive and TUNEL positive A875
→ apoptosis of A875 (ROS generation followed by ↑ caspase-3 and -9)
Acetone extracts of Cladonia furcata and Cladonia foliacea HeLaExtract of C. foliacea: ↑ cytotoxicity of A549 (IC50 13.58 μg/mL), LS174 (IC50 28.98 μg/mL)[112]
Human lung carcinoma A549Extract of C. furcata: ↑ cytotoxicity of HeLa (IC50 11.69 μg/mL)
Human colon carcinoma LS174
Extract of Candelariella vitellineCaco-2↓ proliferation (Ki-67) [32]
→ apoptosis, ↑ necrosis (Caco-2, IC50 125 μg/mL)
↓ Bcl-2
↑ Bax, ↑CASP3 protein level
↑ Bax/Bcl-2 ratio
Methanol extract of Usnea intermediaA549, H1299 MCF7, MDA-MB-231↓ proliferation of H1299 (IC50 10.2 μg/mL) and MDA-MB-231 (IC50 3.0 μg/mL) [113]
→ apoptosis (phophatidylserine translocation, ↑ caspase 3/7 activity, loss of mitochondrial membrane potential, formation of pyknotic nuclei)
Nemania serpens and Nemania aenea var. aureolatum (isolates of endolichenic fungi associated with the lichen Nephroma laevigatum)HT-29, HCT116, PC-3 and DU145↑ anticancer efficacy (IC50 13–39 μg/mL)[114]
→ apoptosis (activated caspase 3, 8, PARP cleavage, chromatin fragmentation)
Physcia cf. Milegrana HeLa, Vero↑ cytotoxicity of HeLa (IC50 137 μg/mL)[115]
Explanatory notes: ↑ increase; ↓ decrease; → induction.
Table 5. In vivo studies dealing with anticancer effects of extracts or isolated secondary metabolites of lichens.
Table 5. In vivo studies dealing with anticancer effects of extracts or isolated secondary metabolites of lichens.
Lichen Metabolites/EXTRACSModelEffectsReferences
UABcap-37 cells inoculated s.c. into C57BL/6 female nude mice; chick embryo chorioallantoic membrane assay; mouse corneal angiogenesis model↓ angiogenesis and VEGFR2 mediated ERK1/2 and AKT signaling; ↓ Bcap-37 cells growth; ↓ proliferation, migration, and tube formation and ↑ apoptosis of HUVEC cells[116]
Human breast cancer MCF-7 cells inoculated s.c. into Balb/c nude mouse↓ tumor growth in dose dependent manner; any toxic effect in animals[71]
H22 cells inoculated into male Kunming mice↓ toxicity of bleomycin therapy; ↑ efficacy of combined therapy vs bleomycine alone-arrested tumor cells in G0/G1; ↑ caspase-3 and -8; ↓ levels of MDA, hydroxyproline, TNF-α, IL-1β, IL-6 and TGF-β1 and ↑ levels of SOD; ↓ p-Smad2/3; ↑ Smad7 proteins[36]
UA and its benzylidene analogueHuman breast cancer MDA-MB-231 and MCF-7 cells inoculated into athymic nude mice↑ anticancer activity on both xenograft models; ↑ autophagy; ↓ mTOR signaling[117]
UAHuman gastric carcinoma BGC823 cells inoculated s.c. into the flank of female BALB/C nude mice↓ tumor volume and weight; ↑ tumor ratio of Bax/Bcl-2 compared to 5-FU[69]
Flavocetraria cucullata extract, UA (F. cucullata), LiA (F. cucullata)Human lung cancer A549 cells injected s.c. into the flank region of Balb/c nude mousetumor free survival number: F. cucullata group ˃ UA group ˃ LiA group.[87]
UA and PUMouse colorectal cancer CT26-Fluc cells inoculated by intrasplenic injection of male BALB/c mice↓ tumor growth in orthotopic liver metastasis model; ↓ levels of EMT; PU without hepatotoxic effect in liver metastasis model[34]
Ethyl acetate extract of Usnea longissimiGastric and esophageal adenocarcinomas of Albino Wistar male rats induced by oral N-methyl-N-nitro-N-nitrosoguanidin administration↓ tumor formation; extract concentrations of 50 and 100 mg/kg demonstrated selectivity to cancer tissue and low toxicity profile in animals[118]
ATRMouse breast carcinoma 4T1 cells inoculated s.c. into BALB/c mice↑ survival time of tumor-bearing animals; ↓ tumor volume; ↑ apoptosis; ↓ oxidative stress in livers of tumor-bearing mice[45]
Mouse Lewis lung carcinoma cells inoculated s.c. into the flanks of C57BL/6 mice↓ tumor volume and weight; ↓ Ki-67; ↓ KITENIN, CD44, STAT, and cyclin-D1[20]
Extract of Rocella montagneiDalton’s lymphoma ascites cells inoculated into Albino Wistar rats and consequent cancer fluid aspiration from rat peritoneal cavity injected into new animals↓ tumor volume; effect comparable to Vincristine[119]
Endolichenic fungus EL002332 (Endocarpon pusillum)Mouse colorectal cancer CT26 cells inoculated s.c. into BALB/c syngeneic mice; TMK1 cells injected into the abdominal cavity of BALB/c mice (intraperitoneal xenografts)↓ tumor score and tumor volume in skin and intraperitoneal tumor-bearing animals[92]
Extract of Candelariella vitellineEhrlich ascites carcinoma cells were injected i.p. and consequently transferred every 5 days into new female Swiss albino mice↓ tumor volume; ↓ tumor cell invasion and mitotic activity; ↑ formation of apoptotic bodies; ↑ ratio of Bax/Bcl-2 on both mRNA and protein levels[32]
Physciosporin (Pseudocyphellaria granulata)Mouse colorectal cancer cells CT26 implanted s.c. into male BALB/c mice↓ tumor volume and weight; without changes in body weight of animals[22]
BA (Cladia aggregate)Sarcoma-180 cells inoculated in the right axillary region of female albino Swiss mice↓ tumor weight; ↑ apoptosis (supposed mechanism)[91]
HA (Pseudoparmelia sphaerospora)Murine melanoma B16-F10 inoculated s.c. into male BALB/c mice↓ tumor volume in both acids; high cancer selectivity and low toxicity in both acids[79]
SA (Parmotrema cetratum)
DA (Usnea longissima)Ehrlich ascites carcinoma (EAC) cells inoculated i.p. to Balb/C male miceanticancer effect on EAC cells; protective activity on different mouse organs[120]
NA (Usnea strigosa)Human breast cancer MDA-MB-231/GFP cells inoculated into female nude mice↓ tumor volume and weight; ↓ c-Met phosphorylation[33]
Explanatory notes: ↑ increase; ↓ decrease; → induction; Abbreviations: ATR, atranorin; BA, barbatic acid; DA, diffractaic acid; EMT, epithelial–mesenchymal transition; HA, hypostictic acid; LiA, lichesterinic acid; NA, nortictic acid; PU, potassium usnate; SA, salazanic acid; UA, usnic acid.

Share and Cite

MDPI and ACS Style

Solárová, Z.; Liskova, A.; Samec, M.; Kubatka, P.; Büsselberg, D.; Solár, P. Anticancer Potential of Lichens’ Secondary Metabolites. Biomolecules 2020, 10, 87. https://doi.org/10.3390/biom10010087

AMA Style

Solárová Z, Liskova A, Samec M, Kubatka P, Büsselberg D, Solár P. Anticancer Potential of Lichens’ Secondary Metabolites. Biomolecules. 2020; 10(1):87. https://doi.org/10.3390/biom10010087

Chicago/Turabian Style

Solárová, Zuzana, Alena Liskova, Marek Samec, Peter Kubatka, Dietrich Büsselberg, and Peter Solár. 2020. "Anticancer Potential of Lichens’ Secondary Metabolites" Biomolecules 10, no. 1: 87. https://doi.org/10.3390/biom10010087

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