Next Article in Journal
Therapeutic Targeting of TLR4 for Inflammation, Infection, and Cancer: A Perspective for Disaccharide Lipid A Mimetics
Next Article in Special Issue
Isolation of the Lanostane Triterpenes Pholiols L–S from Pholiota populnea and Evaluation of Their Antiproliferative and Cytotoxic Activities
Previous Article in Journal
Chemical and Biological Investigations of Allium scorodoprasum L. Flower Extracts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

In Vitro Efficacy of Extracts and Isolated Bioactive Compounds from Ascomycota Fungi in the Treatment of Colorectal Cancer: A Systematic Review

by
Cristina Luque
1,2,3,†,
Ana Cepero
1,2,3,†,
Gloria Perazzoli
1,3,
Cristina Mesas
1,2,3,
Francisco Quiñonero
1,2,3,
Laura Cabeza
1,2,3,
Jose Prados
1,2,3,* and
Consolación Melguizo
1,2,3
1
Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
2
Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
3
Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18014 Granada, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2023, 16(1), 22; https://doi.org/10.3390/ph16010022
Submission received: 16 November 2022 / Revised: 15 December 2022 / Accepted: 18 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Natural Terpenoids for Drug Candidates)

Abstract

:
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Despite the advances and success of current treatments (e.g., chemotherapy), there are multiple serious side effects which require the development of new treatment strategies. In recent years, fungi have gained considerable attention as a source of extracts and bioactive compounds with antitumor capabilities because of their antimicrobial and antioxidant properties and even their anti-inflammatory and antiviral activities. In the present review, a systematic search of the existing literature in four electronic databases was carried out in which the antitumor activity against CRC cells of Ascomycota fungi extracts or compounds was tested. The systematical research in the four databases resulted in a total of 883 articles. After applying exclusion and inclusion criteria, a total of 75 articles were finally studied. The order Eurotiales was the most studied (46% of the articles), and the ethyl acetate extraction was the most used method (49% of the papers). Penicillium extracts and gliotoxin and acetylgliotoxin G bioactive compounds showed the highest cytotoxic activity. This review also focuses on the action mechanisms of the extracts and bioactive compounds of fungi against CRC, which were mediated by apoptosis induction and the arrest of the cell cycle, which induces a notable reduction in the CRC cell proliferation capacity, and by the reduction in cell migration that limits their ability to produce metastasis. Thus, the ability of fungi to induce the death of cancer cells through different mechanisms may be the basis for the development of new therapies that improve the current results, especially in the more advanced stages of the CCR.

1. Introduction

Colorectal cancer (CRC) is the third most common cancer type worldwide and the second deadliest malignancy for both sexes combined. In 2020, it was estimated that 935,000 deaths secondary to CRC occurred, and 1.9 million new cases were diagnosed. Specifically, the American Cancer Society had predicted that, in the United States in 2022, more than 100,000 people would be diagnosed with this type of cancer and approximately 52,000 deaths would be caused by CRC. It is known that CRC is associated with a high socioeconomic status, which explains its high incidence in European countries [1,2]. A clear correlation has been demonstrated between the development of the disease and environmental, hereditary, and lifestyle factors, including obesity, a sedentary lifestyle, smoking, processed or red meat, and alcohol. Nevertheless, certain preventive factors have also been identified, such as physical activity or a healthy diet (e.g., fruit, vegetables, fish, and garlic) [3].
The treatment of choice for CRC includes surgery (in the case of resectable tumours) and chemotherapy, such as 5-fluorouracil, oxaliplatin, irinotecan, and capecitabine, which can be used in mono-therapy or in combination with other drugs. However, chemotherapy causes multiple and serious secondary effects, such as high toxicity to healthy cells or cancer drug resistance. New biological drugs have been developed, such as monoclonal antibodies against epidermal growth factor receptor (EGFR) (cetuximab or panitumumab) or vascular endothelial growth factor (VEGF) (bevacizumab or ramucirumab), which also showed serious limitations both related to specific side effects (i.e., diarrhea, ocular-skin toxicity, etc) and their low efficacy in some types of CC [3,4]. Thus, it is necessary to develop new CRC therapeutic strategies [5,6]. Since the early ages, plants and fungi have aroused the interest of the scientific community as sources of bioactive compounds with antitumor capacities and, therefore, with a promising future as potential drugs [7,8,9].
In this context, fungi represent a relevant resource for isolating bioactive compounds, including polysaccharides, terpenes, terpenoids, proteins, amino acids, nucleosides, and phenols, among others, with antitumor activity and different mechanisms of action, from cell cycle arrest to the suppression of angiogenesis, and a metastatic capacity or induction of cell apoptosis [10]. In fact, Ascomycota and Basidiomycota are the two most researched fungal phyla in oncology [11,12]. Ascomycota is the largest fungal taxonomic group with around 6600 genera classified in approximately 130 orders [13]. Some of these genera, such as Aspergillus, Penicillium, Fusarium, or Cordyceps, are known to have antimicrobial and antioxidant properties and some additional characteristics, such as anti-inflammatory (Cordyceps and Aspergillus) or antiviral (Penicillium) characteristics. Likewise, many of the genera of the phylum Ascomycota showed significant activity against numerous types of tumors, such as leukemia, breast, lung, colon, or liver cancer [12,14,15,16]. Specifically, exopolysaccharides isolated from fungi, such as Cordyceps sinensis or Ganoderma lucidum, suppressed autophagolysosome formation in CRC culture cells. Furthermore, polysaccharides isolated from G. lucidum protected non-tumor colon cells from the accumulation of reactive oxygen species and potentiated the effects of 5-fluorouracil, decreasing tumor size and increasing survival in mouse models [17,18].
The aim of this systematic review was to analyze the most recent literature regarding the application of the fungal phylum Ascomycota for the CRC treatment using functional extracts or bioactive compounds with clearly identified antitumor activity, and gathers information on the processes through which they cause tumor cell death.

2. Results and Discussion

The systematic search in the four electronic databases resulted in a total of 883 articles (Figure 1A). After removing duplicates (n = 335) and articles that did not match the topic or language (n = 419), 129 articles were selected for full-text analysis, of which 16 were eliminated because the full text was not available. Thus, 113 articles were carefully analyzed and, after removing those that did not meet the inclusion criteria (n = 43) or did not comply with the minimum required quality (n = 12), 74 articles were finally obtained, to which one study was added by searching the references of the previous articles. Therefore, the present systematic review comprised a total of 75 articles. This systematic review analyzed a total of 13 orders of the phylum Ascomycota, and 41 articles studied different genera of the order Eurotiales (Aspergillus, Penicillium, Byssochlamys, Talaromyces, and Neosartorya). A total of eight genera of the order Hypocreales (Beauveria, Bionectria, Cordyceps, Engyodontium, Fusarium, Metarhizium, Trichoderma, and Myrothecium) were investigated in 16 articles. The genera of the orders Pleosporales (Alternaria, Bipolaris, Curvularia, Drechslera, Paradendryphiella, Phoma, and Setophoma) and Sordariales (Chaetomium, Trichlocladium, and Scytalidium) were studied in eight articles each. Other genera, such as Pezizales, Capnodiales, Incertae sedis, Diaporthales, or Leotiales, among others, were studied marginally. Finally, the most commonly used extraction method was ethyl acetate, followed by methanol, although many other approaches, such as methanol or water, were also applied (Figure 1B). Most publications assayed a functional extract or bioactive compound dissolved in DMSO against HCT-116 and HT-29, the most commonly used CRC cells lines. The MTT assay was the method of choice to determine antiproliferative activity on cells.
A total of 151 bioactive compounds have been isolated from the different orders of Ascomycota, tested in CRC cell lines (Supplementary Table S1), and analyzed to determine antitumor action mechanisms. As shown in Figure 2, the most studied pathways are those of the Eurotiales and Hypocreales orders.

2.1. Order Eurotiales

As shown in Table 1, 41 articles on the order Eurotiales were analyzed, showing that ethyl acetate was the most frequent extraction method (26 articles), followed by methanol [19,20,21] and ethanol [22,23,24]. Mohamed [25] employed sonication, centrifugation, and lyophilization. In addition, seven articles used differential extraction between a liquid medium and mycelia, most commonly employing ethyl acetate and methanol, respectively [26,27,28,29,30,31,32].

2.1.1. Genus Aspergillus

Twenty-four articles analyzed species of the genus Aspergillus, of which eight articles tested functional extracts on CRC cell lines. Ethyl acetate extracts (five articles) showed IC50 values between 42.75 and 185.9 μg/mL [33,36,58]. Moreover, Ali et al. [36] reported that ethyl acetate extracts from nine different Aspergillus species induced death of 50.1 to 69.1% of HCT-116 cells. A similar extract obtained by Artasasta et al. [33] was reported to cause a significant reduction in the viability of WiDr cells. Asfour et al. [32] also used methanol as a mycelium extraction method, obtaining IC50 values between 15–100 μg/mL in HCT-116 cells, while Alasmary et al. [23] obtained an ethanolic extract with higher antitumor activity (IC50 125 μg/mL) in the same cell line. Furthermore, Abd El-Hady et al. [19] tested a sequential extract of ethyl acetate, methanol, and dichloromethane (100 µg/mL) that induced significant cytotoxicity (15.8%) in the same cell line. Finally, a crude extract obtained by sonication, centrifugation, and lyophilization showed an IC50 value of 9.84 µg/mL in CaCo-2 cells [25]. Functional extract fractions (three articles) were tested on the HCT-116 cell line, with IC50 values between 5.28–193.64 µg/mL [24,33] and 15.8–88% cytotoxicity [19]. Interestingly, most of the extracts obtained from the genus Aspergillus were also tested on other cancer cell lines of liver, larynx, cervix, and breast [23,32,36,58], in which, in general, a higher cytotoxic effect was noted compared to CRC cells. Only the functional extracts obtained by Ashour et al. [24] reduced the IC50 to a greater extent in CRC (more than half the IC50) than in other tumor cells, such as those derived from liver and breast cancer.
On the other hand, 49 bioactive compounds from the genus Aspergillus were described in a total of 14 articles. For example, malformin C was effective in MC-38 and HCT-116 cell lines (IC50 0.27 and 0.18 µM, respectively), with similar results being obtained in the murine pancreatic cancer cell line PanO2 and in the human lung adenocarcinoma cell line H1975. This bioactive compound induced G2/M phase arrest, DNA damage, apoptosis, autophagy, and necrosis [40]. Two of the most promising compounds in relation to the treatment of CRC were gliotoxin and acetylgliotoxin G, which showed very low IC50 values (0.41 and 1.06 µg/mL, respectively) against HCT-116 cells [44]. In fact, gliotoxin was also reported to have antitumor efficacy in chondrosarcoma, cervix, and glioblastoma cells [59,60]. In addition, asperphenin A showed greater activity in CRC cells than in breast cancer cells (IC50 0.84 vs. 6.48 µM, respectively), inducing G2/M cell cycle arrest by the inhibition of tubulin polymerization, induction of apoptosis, and production of reactive oxygen species (ROS). In addition, asperphenin demonstrated a synergistic effect in combination with irinotecan and paclitaxel [20]. Other bioactive compounds, such as clavatustide B, inhibited the G1/S phase, while acetylaranotin, acetylapoaranotin, and deoxyapoaranotin induced apoptosis mediated by caspases 3, 9, and 8 [28,31,42]. Finally, isolated compounds from the genus aspergillus, such as asperphenin A, malformin C, or acetylapoaranotin have succeeded in taking a further step toward in vivo murine research, although more studies are needed [20,38,42].

2.1.2. Genus Penicillium

The most relevant studies in the genus Penicillium used the ethyl acetate extraction method (five out of fourteen) [22,32] or methanol and ethanol methods (two out of fourteen) [49,54,56] to develop functional extracts that showed IC50 values between 0.2 and 102 µg/mL in CRC cells. Canturk et al. [56] and Dikmen et al. [38] showed that ethyl acetate extracts reduced the invasiveness of cancer cells by decreasing cell migration and the expression of genes related to angiogenesis and metastasis. In addition, a total of 48 bioactive compounds from different species of the genus Penicillium (nine of fourteen articles), including arenicolin A, isopenicin A, penipacids A, and norverrucosidin, were detected, showing the lowest IC50 values against HCT-116 (7.3 μg/mL), SW-180 (0.74 μg/mL), RKO (8.4 μg/mL), and HCT-116 cells (5.7 μg/mL), respectively. Furthermore, isopenicin A induced apoptosis and modulated proteins involved in cell cycle progression from G2 to M [21,29,30,52,53,55,57]. The anti-tumor activity of some of the extracts and bioactive compounds from the genus Penicillium were tested against breast, cervix, and liver cancer cells, obtaining similar results [21,32,52,53,55,57].

2.1.3. Genera Neosartorya, Byssochlamys and Talaromyces

Only two studies on the genus Neosartorya developed ethyl acetate extracts (IC50 139 µg/mL in HCT-116 cells) [45,47], while the other three articles reported the isolation of 18 bioactive compounds, such as gliotoxin and acetylgliotoxin, both of which are active against RKO cells (IC50 1.24 μM). Moreover, reduced gliotoxin showed high toxicity in HCT-116 cells (IC50 0.89 μM) [46,48], inducing apoptosis and ROS production [61]. On the other hand, the genera Talaromyces and Byssochlamys were studied by Castro-Carvalho et al. [45] and Khiralla et al. [39], respectively. The latter obtained an acetate extract with IC50 values of 56.3 and 30.4 µg/mL in HT-29 and HCT-116 CRC cell lines, respectively. Specifically, Byssochlamys extracts showed a significantly weaker antiproliferative effect on CRC cells compared to the breast cancer cell line MCF-7 (IC50 1.51 µg/mL).

2.2. Order Hypocreales

2.2.1. Genera Cordyceps, Fusarium and Trichoderma

As shown in Table 2, the studies on the order Hypocreales (16 articles) used a wide variety of extraction methods, although methanol and ethyl acetate were the most common. Four articles focused on the genus Cordyceps, obtaining methanol extracts (two articles) that showed IC50 values between 72.57 and 250 μg/mL against HCT-116, SW-480, and HCT-15, reducing both cell migration and cytoplasmic β-catenin [62,63]. An ethanol extract induced cell morphological changes and G2/M cell cycle arrest [64], and a butanol extract from Cordyceps militaris (sprouted soybean) induced a strong inhibition of HT-29 cell proliferation (56%) and G2/M phase arrest by blocking cyclin B1 and the expression of Cdc25c [65]. Lee et al. [64] tested this ethanol extract in a xenograft mouse model and found a significant inhibition of tumor growth and a reduction in mouse mortality. On the other hand, the genus Fusarium was studied in four articles, showing active functional extracts against CaCo-2, HCT-116, and HCT-8 cells (IC50 between 0.3779–98.68 μg/mL) [25,66,67,68]. In one article, standard camptothecin and camptothecin crude extract were isolated and tested against CaCo-2 cells, resulting in IC50 values of 2.41 and 0.291 μM, respectively [68]. This compound has been used for the development of a conjugate, CT-2106, that has been studied in a clinical trial in combination with 5-fluorouracil and folic acid (NCT00291785), whose results had not been reported yet. In addition, camptothecin is the precursor of irinotecan, an antitumor drug that, in combination with other anticancer agents, has been widely used in clinical trials and its clinical use is well accepted [69]. Finally, the genus Trichoderma was analyzed in four articles, obtaining functional extracts (IC50 between 11–100 μg/mL), fractions (IC50 between 7.3 and 14.9 μg/mL) [24,66,70], or bioactive compounds, such as trichodermaloid A and B (IC50 9.3 and 8.6 μM in the SW-620 cell line, respectively) [71]. All of these findings are consistent with those obtained in other forms of tumors (breast, lung, liver, and cervix cancers, among others).

2.2.2. Other Genera

The genera Beauveria, Bionectria, Engyodontium, Metarhizium, and Myrothecium were analyzed in one article each. 1-Hydroxy-10-methoxy-dibenz[b, e]oxepin-6,11-dione, chrysazin (IC50 > 30 μM), and globosuxanthone A (IC50 10.7 μM) were isolated from the genus Beauveria and tested on HCT-15 cells [72]. Beauvericin, another compound from the genus Beauveria, has been used for in vivo assays in BALB/c and CB-17/SCID mice, decreasing tumor volumes and increasing necrotic areas of tumors, becoming a potentially interesting drug for the treatment of CRC [76]. Exopolysaccharides isolated from the genus Bionectria (0.45 mg/mL) significantly reduced HT-29 cell viability (15.42%) [73]. In addition, functional extracts from the genera Engyodontium and Myrothecium showed IC50 values of 2.5 μg/mL and 380 ng/mL in HCT-116 cells, respectively. Specifically, Myrothecium extract showed higher cytotoxic activity in breast MCF-7 cells (IC50 107 ng/mL) and lower in the liver cell line HepG2 (IC50 711 ng/mL) [49,73]. Finally, destruxins A, B, and E from the genus Metarhizium were tested in CaCo-2 and HCT-116 cells, showing IC50 values between 0.04 and 10 μM. However, they were also active against the KB-3-1 cell line derived from the epidermal carcinoma and A549 lung cancer cells. Furthermore, destruxin E induced ROS production and activated apoptotic caspases, even before mitochondrial membrane depolarization. The three destruxins reduced cell migration and angiogenesis, induced G0/G1 cell cycle arrest in the CaCo-2 cell line, and interfered with the MAPK and/or PI3K/Akt signaling pathways [74].

2.3. Orders Pleosporales and Sordariales

As shown in Table 3, the main studies of the order Pleosporales focused on the genus Alternaria (three out of eight articles) to obtain methanol extracts that were tested on HCT-116 and SW-480 cells (IC50 5.39 and 12.37 μg/mL, respectively) [39,77]. In addition, the compound (6aR, 6bS, 7S)-3, 6a, 7, 10-tetra-hydroxy-4, 9-dioxo-4, 6a, 6b, 7, 8, 9-hexahydroperylene was isolated from the extract of the genus Alternaria with an IC50 value of 1.78 μmol/L in HCT-8 cells [78]. Pleosporales spp. were used to obtain an ethyl acetate extract that led to IC50 values of 69.4 μg/mL in HT-29 cells and 36.7 μg/mL in HCT-116 cells, while its cytotoxic activity in the MCF-7 breast cancer line was even half of that of the latter CRC line [39]. An aqueous extract and an organic residue obtained with dichloromethane were tested in HCT-116 cells, obtaining IC50 values between 12 and 100 μg/mL, respectively [66]. In addition, the genera Bipolaris, Phoma, Drechslera, and Curvularia were studied by obtaining functional extracts, which evidenced IC50 values ranging from 18.97 to 202.5 µg/mL against HCT-116, HT-29, and HCT-8 cells, with similar antiproliferative activity in breast cancer cell lines [23,36,70]. From the Drechslera genus, di-2-ethylhexyl phthalate was isolated (IC50 9.5 ± 0.4 µg/mL in HCT-116 cell line). Another compound, (3R, 6R) hyalodendrin, was isolated from the genus Paradendryphiella (IC50 between 48.0 ± 9.3 nM and 163.7 ± 11.0 nM). Finally, seven bioactive compounds were isolated from the Setophoma genus and tested on SW-620 cells, with IC50 values between 0.21 (penicillixanthone A) and 19.12 μM (secalonic acid E) [23,79,80].
On the other hand, the order Sordariales (eight articles) included the study of the genus Chaetomium (six articles) (Table 4). The analysis of ethanol extracts against HCT-8, HCT-116, and HT-29 cells was reported with IC50 values ranging from 1.2 to 152.8 µg/mL [39,49,66] and twelve bioactive compounds. Specifically, Chaetocochins C and J resulted in the lowest IC50 values: 0.63 and 0.56 μM in SW-480 and HCT-116 cells, respectively [81,82,83]. Furthermore, Trichocladinols D-H, E, F, and G were isolated from the genus Trichlocladium (one article), showing IC50 values between 41.7 and 56.6 μM against the HCT-116 and SW-480 cells lines [84]. Finally, a total of 11 bioactive compounds were isolated from the genus Scytalidium in one study, including 5’-formyl-2’-hydroxyl-4’-methoxy-(E,E)-sorbophenone (IC50 0.5 μM) and 5 ‘-formyl -2′-hydroxy-4′-methoxy-(E)-4-hexenophenone, which showed the best results (IC50 2.5 μM) against SW-620 cells [85].

2.4. Minoritary Orders

As shown in Table 5, the antitumor activity of the order Capnodiales (three articles) was studied using the genera Cladosporium (functional extracts) and Zasmidium (bioactive compound). In fact, 8,8′-Bijuglone showed an IC50 value of 45 µg/mL in the HCT-116 cell line. Functional extracts and bioactive compounds from Cladosporium were tested on both CRC and breast cancer cells with a significant differential effect [39,86,87]. Taxol was one of the compounds, which, due to its potent antitumor effect, has not only been tested in several clinical trials but has also come to be used in clinics against CRC [88]. In addition, the genera Sclerotinia and Lachnum (order Helotiales) were processed to obtain the exopolysaccharide LEP-2b and derivates from the genus Lachnum, which showed high antitumor activity (e.g., IC50 of LEP-2b, 85.78 μg/mL) in the CT-26 cell line, among other tumor cells [89,90,91].
Studies on the order Diaportales (two articles) showed methanol and ethyl acetate extracts with IC50 values ranging from 5.63 to 24.47 µg/mL in SW-480 and HCT-116 cells lines [77], and the isolation of dicerandrol A and B with significant antitumor activity in HCT-116 CRC cells with IC50 values of 2.64 and 3.94 μM, respectively [92]. All of them were also highly effective against cell lines of other cancer types, such as breast, lung, and liver. The order Pezizales (two articles) was studied by Liu et al. [93] and Tang et al. [94]. The latter showed polysaccharides from the genus Morchella with high CRC cell cytotoxicity (IC50 between 1.229 and 2.827 mg/mL in CaCo-2 cells). This finding was supported by results in the hepatocellular cancer line HepG2. Similarly, four different compounds were isolated from the order Xylariales, highlighting 5-methylmellein and daldinone F, which showed significant antitumor activity (IC50 of 2 and 9.59 μM) in SW-480 and HCT-116 cells, respectively. Moreover, 5-Methylmellein showed activity against prostate and breast cancer cells. Indeed, it was encapsulated in nanoparticles, increasing the IC50 to <0.5 µg/mL, and inducing apoptosis, ROS production, and the loss of the mitochondrial membrane potential [95,96]. Finally, other orders, such as Boliniales, Incertae sedis, Leotiales, and Venturiales were studied using ethyl acetate extracts or bioactive compounds, such as xylarenone D, greensporone C, and O-desmethyl greensporone C, which were effective against CRC cells (IC50 1.5, 7.5 and 13.8 μM, respectively), among other cancer types (melanoma, glioblastoma, and leukemia) [34,39,49,97].
Table 5. Antitumor activity of the extracts or isolated compounds from Minoritary orders in CRC cancer cell lines.
Table 5. Antitumor activity of the extracts or isolated compounds from Minoritary orders in CRC cancer cell lines.
OrderGenusIsolated fromExtractionIsolated CompoundsCell Line/Administration/Cytotoxicity AssayCompound and IC50 or Cell Death (%)Mechanism of ActionReference
CapnodialesZasmidiumFoliageEthyl acetate (EtOAc)8,8′-BijugloneHCT-116
DMSO
MTT
8,8′-Bijuglone: 45 μg/mL-[87]
Capnodiales
Incertae sedis
Cladosporium
Hansfordia
C. procera
Vernonia amygdalina
EtOAc-HT-29 and HCT-116
-
MTT
C. cladosporioides extracts 1 (HT-29: 77.7 μg/mL; HCT-116: 45.6 μg/mL), 2 (HT-29 and HCT-116: >100 μg/mL); H. sinuosae extract: HT-29 (47.6 μg/mL), HCT-116 (>100 μg/mL) -[39]
CapnodialesCladosporiumMedicinal plantsSodium bicarbonateTaxolHCT-15
Methanol
MTT
Taxol: 3.5 μM-[86]
HelotialesSclerotiniaContaminated soybean seedEthanol-HCT-8
-
MTT
Fractions ethyl acetate (48.03 μg/mL), F3 (250.50 μg/mL)-[91]
HelotialesSclerotiniaSoybeansTrituration and boiled-CCD-18Co and HT-29
Alone
Electronic counter
Aqueous extract: CCD-18Co (11%), HT-29 (58%)Aqueous extract induces ROS and extrinsic pathway[90]
HelotialesLachnum-EthanolExopolysaccharide LEP-2bCT-26
DMSO
MTT
LEP-2b: 8816.27 μg/mL; PLEP-2b: 85.78 μg/mL; SLEP_2b: 154.52 μg/mL-[89]
DiaporthalesPhomopsisAcanthus ilicifoliusMethanolPhomolactonexanthone A (1), B (2) and C (3)
Dicerandrol A (4), B (5) and C (6)
Deacetylphomoxanthone B (7)
Penexanthone A (8)
HCT-116
Alone
MTT
(1) and (2): >50 μM; (3): 44.06 μM; (4): 2.64 μM; (5): 3.94 μM; (6): 42.63 μM; (7): 7.12 μM; (8): 6.92 μM-[92]
DiaporthalesPhomopsisMiquelia dentataMethanol
EtOAc
-SW-480 and HCT-116
-
Hoechst 33342
Methanol extract: HCT-116 (24.47 μg/mL), SW-480 (14.45 μg/mL)
Ethyl acetate extract: HCT-116 (5.63 μg/mL), SW-480 (23.5 μg/mL)
-[77]
PezizalesMorchella-Hot waterPolysaccharides CaCo-2
Alone
Methylene Blue Assay
PMEP: 1.840 mg/mL; Ac-PMEP 1 (2.094 mg/mL), 2 (2.827 mg/mL), 3 (1.229 mg/mL)-[94]
PezizalesMorchella-Pulsed electric field-HT-29
Alone
MTT
M2 fraction: 54.29%M2 fraction induces apoptosis[93]
XylarialesXylariaAegle marmelosEtOAc5-methylmelleinHCT-116
Alone
SRB
5-methylmellein: 2.0 μg/mL;
5-methylmellein nanoparticle: <0.5 μg/mL
5-methylmellein nanoparticle induces apoptosis, ROS and mitochondrial membrane potential loss[95]
XylarialesDaldiniaTenodera aridifoliaEtOAcDaldinone F (1)
Nodulisporin G (2)
Dalmanol C (3)
SW-480 and HCT-116
Alone
MTT
(1): SW-480 (9.59 μM), HCT-116 (>20 μM); (2) and (3): SW-480 and HCT-116 (>20 μM)-[96]
BolinialesCamaropsAlibertia macrophyllaEtOAcXylarenone C and DHCT-8
Alone
MTT
Xylarenones C (1.9 μg/mL),
D (1.5 μg/mL)
Xylarenone D shows weak AChE inhibitory activity[34]
LeotialesHalenosporaWood Greensporone A (1), C (2)
Dechlorogreensporone A (3), D (4)
O-Desmethylgreensporone C (5)
HT-29
DMSO
CellTiter 96 Aqueous One Solution Cell Proliferation Assay
(1) and (3): >20 μM; (2): 7.5 μM; (4): 25.4 μM; (5): 13.8 μM-[97]
VenturialesOchroconis-EtOAc-HCT-116
DMSO
SRB
Ochroconis sp. extract: 70.5 μg/mL-[49]
PMEP: polysaccharides extracted from Morchella angusticepes Peck; Ac-PMEP: acetylated derivatives of PMEP; SRB: sulforhodamine B; MTT: 3-(4,5-dimethytlthiazol-2-yl)-2,5-diphenyltetrazolium bromide; IC50: half maximal inhibitory concentration; DMSO: dimethyl sulfoxide; ROS: reactive oxygen species; AChE: acetylcholinesterase; PLEP: phosphorylated polysaccharide; SLEP: sulfated polysaccharide.

3. Materials and Methods

A complete method was thoroughly organized with the collection of data and the steps of analysis, including the protocol registration (https://doi.org/10.17605/OSF.IO/X5KTD accessed on 10 November 2022).

3.1. Study Eligibility

Since the purpose of this review was to compile the most recent and representative knowledge of the antitumor capacities against colorectal cancer of bioactive compounds isolated from Ascomycota fungi or their functional extracts, a bibliometric analysis was carried out. A period of 10 years was established, considering older results obsolete (Burton–Kebler index for obsolescence) and including more than half of the actual disponible papers [98].

3.2. Inclusion Criteria

Research articles published in English from January 2011 to October 2021 in which extracts, or compounds isolated from Ascomycota fungi, had their antitumor activity on CRC cell lines tested were included in this systematic review. The research articles had been published in peer reviewed journals with the full text accessible.

3.3. Exclusion Criteria

Papers in which any colon cancer cell line was not used, or the bioactive compound or extract tested had no antitumor activity, were excluded. Furthermore, publications in which the bioactive compound was synthesized/purchased, or the extraction methodology was not specified, were also excluded. Finally, studies that did not exceed the minimum requirements of an in vitro study or with a low quality of the study, were excluded from the present review.

3.4. Data Sources

Four electronic databases were used to perform the systematic review: MedLars Online International Literature, via PubMed, SCOPUS, Web of Science Core Collection, and the Cochrane Library Plus. Firstly, the following Medical Subject Headings (MeSH) were defined to use as descriptors in Pubmed: “Colorectal Neoplasms”, “Fungi”, “Ascomycota”, and “Aspergillus”. The final equation was ((“Colorectal Neoplasms”[MeSH Terms] OR ((“colon”[Title/Abstract] OR “rectal”[Title/Abstract] OR “colorectal”[Title/Abstract] OR “colonic”[Title/Abstract]) AND (“cancer*”[Title/Abstract] OR “tumor*”[Title/Abstract] OR “tumour*”[Title/Abstract] OR “neoplasm*”[Title/Abstract] OR “carcinoma*”[Title/Abstract] OR “adenocarcinoma*”[Title/Abstract]))) AND (“Fungi”[MeSH Terms] OR “fung*”[Title/Abstract] OR “Ascomycota”[MeSH Terms] OR “Ascomycota”[Title/Abstract] OR “Ascomycetes”[Title/Abstract] OR “Aspergillus”[MeSH Terms] OR “Aspergillus”[Title/Abstract] OR “Chaetomium”[Title/Abstract] OR “Cordyceps”[Title/Abstract] OR “Neosartorya”[Title/Abstract]) AND (“Metabolite”[Title/Abstract] OR “extract*”[Title/Abstract] OR “bioactive compound*”[Title/Abstract])). The equation was adapted for the other three databases. The final list of studies included was completed by a manual search from the references of the selected publications.

3.5. Study Selection

Two of the authors (C.L. and A.C.) performed the bibliographic search, screened the abstract of the resulting publications, and selected the adequate ones for a fully-text review. Editorials, conference papers, bibliographic and meta-analysis reviews, book chapters, epidemiological studies, and case reports were excluded. In the following stage of the selection process, the same authors analyzed and included or excluded the full-text articles. Because the aim of this study was to review the current data available relating to in vitro publications, in vivo and clinical trials were manually excluded.

3.6. Data Extraction

Once the list of the articles included in the study was obtained, the same authors independently evaluated and extracted data from the selected studies according to the Cohen kappa statistical test for agreements (more than 0.8) [99]. Any discrepancy was resolved by a consensus between C.L. and A.C. or two more authors (J.P and C.M), if necessary. All of the selected articles were analyzed for quality using a specific questionnaire for in vitro studies with a first part in which the minimum requirements of an in vitro study were determined (score > 6), and a second part in which the quality of the study was analyzed (0–6 = low; 7–14 = good; 15–20 = excellent) based on materials and methods, results, and conclusions. Publications were classified according to the order of the studied fungi and the extracted data are condensed in Table 1, Table 2, Table 3, Table 4 and Table 5. To facilitate the interpretation of the selected studies, reference genera of fungi studied, where the fungi were isolated from, the extraction method, isolated compounds, the cell line used, cytotoxicity activity, and the mechanism of action.

4. Conclusions

This systematic review focused on in vitro studies on the antitumor activity of extracts and compounds isolated from fungi of the phylum Ascomycota. Of all of the genera analyzed in the literature, Penicillium, Fusarium, and Chaetomium produced the extracts with the greatest antitumor activity in CRC. A wide variety of bioactive compounds have been isolated from different genera of the phylum, some of which are particularly interesting given their high anticancer activity against this tumor. Although current results are very promising, more research is needed on genera that have been less studied. It is also important to move towards in vivo studies and/or clinical trials of the extracts and/or bioactive compounds with the aim that they could be used as a therapy against CRC in the future.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph16010022/s1.

Author Contributions

Conceptualization, J.P. and C.M. (Consolación Melguizo); methodology, C.L., A.C., C.M. (Cristina Mesas) and G.P.; software, F.Q.; validation, F.Q., G.P. and L.C.; formal analysis, C.L.; investigation, C.L., C.M. (Cristina Mesas) and A.C.; data curation, C.L.; writing—original draft preparation, C.L., A.C., G.P. and C.M. (Cristina Mesas); writing—review and editing, L.C. and C.M. (Cristina Mesas); visualization, C.M. (Consolación Melguizo); supervision, F.Q. and L.C.; funding acquisition, J.P. and C.M. (Consolación Melguizo). All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by the Project P20_00540 (Proyectos I+D+i Junta de Andalucía 2020), PYC20 RE 035, and P18-TP-1420 (Junta de Andalucía) and Innbio INB-009 (Granada University and ibs. GRANADA). C.L. and A.C. acknowledges the P-FIS (2020) and FPU (2019) grant from the Instituto de Salud Carlos III and Ministerio de Educación, Ciencia y Deporte y Competitividad (Spain).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

We thank the Instrumentation Scientific Center (CIC) from the University of Granada for technical assistance.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

CRCColorectal cancer
EGFREpidermal growth factor receptor
VEGFVascular endothelial growth factor
DMSODimethyl sulfoxide
MTT3-(4,5-dimethytlthiazol-2-yl)-2,5-diphenyltetrazolium bromide
Asp. AAsperphenins A
Asp. BAsperphenins B
SRBSulforhodamine B
ROSReactive oxygen species
TXLPaclitaxel
EtOAcEthyl acetate
CH2Cl2 extractSequential ethyl acetate, methanol, and dichloromethane extract
DoxDoxorubicin
CCK8Cell counting kit-8
WST-14-[3-(4-Iodo-phenyl)-2-(4-nitrophenyl)-2H-5 tetrazolio]-1,3-benzene disulphonate
RTCA-DPReal-Time Cell Analysis System
EAEEthyl acetate extract
MTS((3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)) assay
MrnaMessenger RNA
IC50Half maximal inhibitory concentration
GSCCordyceps militaris cultivated on germinated soybeans
EEEthanol extract
HCT-116/HT-29 oxaOxaliplatin resistant cell
HCT-116/HT-29 5FU5-fluorouracil resistant cell
HCT-116/HT-29 SN-38SN-38 resistant cell
CK01Pleosporales sp. Extract
SN-387-Ethyl-10-hydroxycamptothecin
AMPKAMP-activated protein kinase
PARPPoly (ADP-ribose) polymerase
PMEPPolysaccharides extracted from Morchella angusticepes Peck
Ac-PMEPAcetylated derivatives of PMEP
AChEAcetylcholinesterase
PLEPPhosphorylated polysaccharide
SLEPSulfated polysaccharide

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  3. Recio-Boiles, A.; Cagir, B. Colon Cancer. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  4. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef] [PubMed]
  5. Hervieu, C.; Christou, N.; Battu, S.; Mathonnet, M. The Role of Cancer Stem Cells in Colorectal Cancer: From the Basics to Novel Clinical Trials. Cancers 2021, 13, 1092. [Google Scholar] [CrossRef] [PubMed]
  6. Sang, R.; Stratton, B.; Engel, A.; Deng, W. Liposome technologies towards colorectal cancer therapeutics. Acta Biomater. 2021, 127, 24–40. [Google Scholar] [CrossRef]
  7. Redondo-Blanco, S.; Fernández, J.; Gutiérrez-del-Río, I.; Villar, C.J.; Lombó, F. New Insights toward Colorectal Cancer Chemotherapy Using Natural Bioactive Compounds. Front. Pharm. 2017, 8, 109. [Google Scholar] [CrossRef] [Green Version]
  8. Zhang, M.; Zhang, Y.; Zhang, L.; Tian, Q. Mushroom polysaccharide lentinan for treating different types of cancers: A review of 12 years clinical studies in China. Prog. Mol. Biol. Transl. Sci. 2019, 163, 297–328. [Google Scholar]
  9. Mesas, C.; Fuel, M.; Martínez, R.; Prados, J.; Melguizo, C.; Porres, J.M. In vitro evidence of the antitumor capacity of Solanaceae and Cucurbitaceae in colon cancer: A systematic review. Crit. Rev. Food Sci. Nutr. 2021, 62, 1–22. [Google Scholar]
  10. Dai, R.; Liu, M.; Nik Nabil, W.N.; Xi, Z.; Xu, H. Mycomedicine: A Unique Class of Natural Products with Potent Anti-tumour Bioactivities. Molecules 2021, 26, 1113. [Google Scholar] [CrossRef]
  11. Contreras-Ochoa, C.O.; Maza-Lopez, J.; Mendoza de Gives, P.; Aguilar-Marcelino, L.; Mojica-Cardoso, C.; Dimas-González, J.; Fernández-Coto, D.L.; Reyna-Figueroa, J.; López-Arellano, M.E.; Lagunas-Martínez, A. Cell death induction by mycelium extracts from Pleurotus spp. on cervical cancer cell lines. Nat. Prod. Res. 2022, 36, 6091–6609. [Google Scholar] [CrossRef]
  12. El-Bondkly, E.A.M.; El-Bondkly, A.A.M.; El-Bondkly, A.A.M. Marine endophytic fungal metabolites: A whole new world of pharmaceutical therapy exploration. Heliyon 2021, 7, e06362. [Google Scholar] [CrossRef] [PubMed]
  13. Wijayawardene, N.N.; Hyde, K.D.; Lumbsch, H.T.; Liu, J.K.; Maharachchikumbura, S.S.N.; Ekanayaka, A.H.; Tian, Q.; Phookamsak, R. Outline of Ascomycota: 2017. Fungal Divers. 2018, 88, 167–263. [Google Scholar] [CrossRef]
  14. Rupa, E.J.; Li, J.F.; Arif, M.H.; Yaxi, H.; Puja, A.M.; Chan, A.J.; Hoang, V.; Kaliraj, L.; Yang, D.C.; Kang, S.C. Cordyceps militaris Fungus Extracts-Mediated Nanoemulsion for Improvement Antioxidant, Antimicrobial, and Anti-Inflammatory Activities. Molecules 2020, 25, 5733. [Google Scholar] [CrossRef] [PubMed]
  15. Ukwatta, K.M.; Lawrence, J.L.; Wijayarathne, C.D. Antimicrobial, anti-cancer, anti-filarial and anti-inflammatory activities of Cowabenzophenone A extracted from the endophytic fungus Aspergillus terreus isolated from a mangrove plant Bruguiera gymnorrhyza. Mycology 2020, 11, 297–305. [Google Scholar] [CrossRef] [Green Version]
  16. El-Gendy, M.M.A.A.; Awad, M.F.; El-Shenawy, F.S.; El-Bondkly, A.M.A. Production, purification, characterization, antioxidant and antiproliferative activities of extracellular L-asparaginase produced by Fusarium equiseti AHMF4. Saudi J. Biol. Sci. 2021, 28, 2540–2548. [Google Scholar] [CrossRef]
  17. Opattova, A.; Horak, J.; Vodenkova, S.; Kostovcikova, K.; Cumova, A.; Macinga, P.; Galanova, N.; Rejhova, A.; Vodickova, L.; Kozics, K.; et al. Ganoderma Lucidum induces oxidative DNA damage and enhances the effect of 5-Fluorouracil in colorectal cancer in vitro and in vivo. Mutat. Res. Gen Tox. Environ. 2019, 845, 403065. [Google Scholar] [CrossRef]
  18. Qi, W.; Zhou, X.; Wang, J.; Zhang, K.; Zhou, Y.; Chen, S.; Nie, S.; Xie, M. Cordyceps sinensis polysaccharide inhibits colon cancer cells growth by inducing apoptosis and autophagy flux blockage via mTOR signaling. Carbohydr. Polym. 2020, 237, 116113. [Google Scholar] [CrossRef]
  19. Abd El-Hady, F.K.; Shaker, K.H.; Souleman, A.M.A.; Fayad, W.; Abdel-Aziz, M.S.; Hamed, A.A.; Iodice, C.; Tommonaro, G. Comparative Correlation Between Chemical Composition and Cytotoxic Potential of the Coral-Associated Fungus Aspergillus sp. 2C1-EGY Against Human Colon Cancer Cells. Curr. Microbiol. 2017, 74, 1294–1300. [Google Scholar] [CrossRef]
  20. Bae, S.Y.; Liao, L.; Park, S.H.; Kim, W.K.; Shin, J.; Lee, S.K. Antitumor Activity of Asperphenin, A., a Lipopeptidyl Benzophenone from Marine-Derived Aspergillus sp. Fungus, by Inhibiting Tubulin Polymerization in Colon Cancer Cells. Mar. Drugs 2020, 18, 110. [Google Scholar] [CrossRef] [Green Version]
  21. Perlatti, B.; Lan, N.; Earp, C.E.; AghaAmiri, S.; Vargas, S.H.; Azhdarinia, A.; Bills, G.F.; Gloer, J.B. Arenicolins: C-Glycosylated Depsides from Penicillium arenicola. J. Nat. Prod. 2020, 83, 668–674. [Google Scholar] [CrossRef]
  22. Shoeb, M.; Thoo-Lin, P.K.; Nahar, N. Anti-colon cancer activity of endophytic fungal strains from Terminalia chebula Rezt. Bangladesh. J. Pharm. 2012, 7, 47–49. [Google Scholar] [CrossRef] [Green Version]
  23. Alasmary, F.A.S.; Awaad, A.S.; Kamal, M.; Alqasoumi, S.I.; Zain, M.E. Antitumor activity of extract and isolated compounds from Drechslera rostrata and Eurotium tonophilum. Saudi Pharm. J. 2018, 26, 279–285. [Google Scholar] [CrossRef] [PubMed]
  24. Ashour, M.; Abdelwahab, M. Antimicrobial and Cytotoxic Activities of Plant-Derived Endophytic Metabolites from Northern Border Region, KSA. Indo Am. 2018, 5, 1122–1132. [Google Scholar]
  25. Mohamed, H.F. Molecular analysis and anticancer properties of two identified isolates, Fusarium solani and Emericella nidulans isolated from Wady El-Natron soil in Egypt against Caco-2 (ATCC) cell line. Asian Pac. J. Trop. Biomed. 2012, 2, 863–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Montenegro, T.G.C.; Rodrigues, F.A.R.; Jimenez, P.C.; Angelim, A.L.; Melo, V.M.M.; Rodrigues Filho, E.; de Oliveira, M.; Costa-Lotufo, L. Cytotoxic Activity of Fungal Strains Isolated from the Ascidian Eudistoma vannamei. Chem. Biodivers. 2012, 9, 2203–2209. [Google Scholar] [CrossRef]
  27. Xiao, L.; Liu, H.; Wu, N.; Liu, M.; Wei, J.; Zhang, Y.; Lin, X. Characterization of the high cytochalasin E and rosellichalasin producing-Aspergillus sp. nov. F1 isolated from marine solar saltern in China. World J. Microbiol. Biotechnol. 2013, 29, 11–17. [Google Scholar] [CrossRef]
  28. Ye, P.; Shen, L.; Jiang, W.; Ye, Y.; Chen, C.T.A.; Wu, X.; Wu, B. Zn-Driven Discovery of a Hydrothermal Vent Fungal Metabolite Clavatustide, C, and an Experimental Study of the Anti-Cancer Mechanism of Clavatustide B. Mar. Drugs 2014, 12, 3203–3217. [Google Scholar] [CrossRef] [Green Version]
  29. Xu, X.; Zhang, X.; Nong, X.; Wang, J.; Qi, S. Brevianamides and Mycophenolic Acid Derivatives from the Deep-Sea-Derived Fungus Penicillium brevicompactum DFFSCS025. Mar. Drugs 2017, 15, 43. [Google Scholar] [CrossRef] [Green Version]
  30. Zhang, Y.; Feng, Y.; Kramer, M.; Essmann, F.; Grond, S. A New Acetylenic Compound and Other Bioactive Metabolites from a Shark Gill-derived Penicillium Strain. Rec. Nat. Prod. 2017, 11, 31–36. [Google Scholar]
  31. Phainuphong, P.; Rukachaisirikul, V.; Phongpaichit, S.; Sakayaroj, J.; Kanjanasirirat, P.; Borwornpinyo, S.; Akrimajirachoote, N.; Yimnual, C.; Muanprasat, C. Depsides and depsidones from the soil-derived fungus Aspergillus unguis PSU-RSPG204. Tetrahedron 2018, 74, 5691–5699. [Google Scholar] [CrossRef]
  32. Asfour, H.Z.; Awan, Z.A.; Bagalagel, A.A.; Elfaky, M.A.; Abdelhameed, R.F.A.; Elhady, S.S. Large-Scale Production of Bioactive Terrein by Aspergillus terreus Strain S020 Isolated from the Saudi Coast of the Red Sea. Biomolecules 2019, 9, 480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Artasasta, M.; Yanwirasti, T.M.; Djamaan, A.; Handayani, D. Cytotoxic and antibacterial activities of marine sponge-derived fungus Aspergillus nomius NC06. Rasayan. J. Chem. 2019, 12, 1463–1469. [Google Scholar] [CrossRef]
  34. Mandelare, P.E.; Adpressa, D.A.; Kaweesa, E.N.; Zakharov, L.N.; Loesgen, S. Coculture of Two Developmental Stages of a Marine-Derived Aspergillus alliaceus Results in the Production of the Cytotoxic Bianthrone Allianthrone A. J. Nat. Prod. 2018, 81, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
  35. Yin, J.; Zhang, C.; Huang, J.; Zhang, J.; Liu, D.; Huang, J.; Proksch, P.; Lin, W. Violaceimides A–E, sulfur-containing metabolites from a sponge-associated fungus Aspergillus violaceus. Tetrahedron. Lett. 2018, 59, 3157–3160. [Google Scholar] [CrossRef]
  36. Ali, M.; AbdElfattah, A.; Khalil, N.; Sayed, M. Biological Activities of the Alkaloid Quinazoline Extracted from Aspergillus nomius. Egypt J. Bot. 2017, 57, 565–582. [Google Scholar] [CrossRef]
  37. Artasasta, M.A.; Yanwirasti, D.A.; Handayani, S. Cytotoxic activity screening of ethyl acetate fungal extracts derived from the marine sponge Neopetrosia chaliniformis AR-01. J. Appl. Pharm. Sci. 2017, 7, 174–178. [Google Scholar]
  38. Dikmen, M.; Canturk, Z.; Artagan, O.; Ozturk, N. Antioxidant, Antiproliferative and Apoptotic Effects of Secondary Metabolites of Halotolerant Aspergillus terreus on Colon Adenocarcinoma Caco-2 Cells. Int. J. Pharm. 2017, 13, 227–236. [Google Scholar] [CrossRef] [Green Version]
  39. Khiralla, A.; Mohamed, I.E.; Tzanova, T.; Schohn, H.; Slezack-Deschaumes, S.; Hehn, A.; André, P.; Carre, G.; Spina, R.; Lobstein, A.; et al. Endophytic fungi associated with Sudanese medicinal plants show cytotoxic and antibiotic potential. FEMS Microbiol. Lett. 2016, 363, fnw089. [Google Scholar] [CrossRef]
  40. Wang, J.; Jiang, Z.; Lam, W.; Gullen, E.A.; Yu, Z.; Wei, Y.; Wang, L.; Zeiss, C.; Beck, A.; Cheng, E.; et al. Study of malformin C, a Fungal Source Cyclic Pentapeptide, as an Anti-Cancer Drug. PLoS ONE 2015, 10, e0140069. [Google Scholar] [CrossRef]
  41. Gu, W.; Qiao, C. Furandiones from an endophytic Aspergillus terreus residing in Malus halliana. Chem. Pharm. Bull. 2012, 60, 1474–1477. [Google Scholar] [CrossRef] [Green Version]
  42. Choi, E.J.; Park, J.S.; Kim, Y.J.; Jung, J.H.; Lee, J.K.; Kwon, H.C.; Yang, H.O. Apoptosis-inducing effect of diketopiperazine disulfides produced by Aspergillus sp. KMD 901 isolated from marine sediment on HCT116 colon cancer cell lines. J. Appl. Microbiol. 2011, 110, 304–313. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, Y.X.; Xu, M.Y.; Li, H.J.; Zeng, K.J.; Ma, W.Z.; Tian, G.B.; Xu, J.; Yang, D.; Lan, W. Diverse Secondary Metabolites from the Marine-Derived Fungus Dichotomomyces cejpii F31-1. Mar. Drugs 2017, 15, 339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Rodrigues, B.S.F.; Sahm, B.D.B.; Jimenez, P.C.; Pinto, F.C.L.; Mafezoli, J.; Mattos, M.C.; Rodrigues-Filho, E.; Pfenning, L.H.; Abreu, L.M.; Costa-Lotufo, L.; et al. Bioprospection of Cytotoxic Compounds in Fungal Strains Recovered from Sediments of the Brazilian Coast. Chem. Biodivers. 2015, 12, 432–442. [Google Scholar] [CrossRef] [PubMed]
  45. Castro-Carvalho, B.; Ramos, A.A.; Prata-Sena, M.; Malhão, F.; Moreira, M.; Gargiulo, D.; Dethoup, T.; Buttachon, S.; Kijjoa, A.; Rocha, E. Marine-derived Fungi Extracts Enhance the Cytotoxic Activity of Doxorubicin in Nonsmall Cell Lung Cancer Cells A459. Pharm. Res. 2017, 9, S92–S928. [Google Scholar]
  46. Prata-Sena, M.; Ramos, A.A.; Buttachon, S.; Castro-Carvalho, B.; Marques, P.; Dethoup, T.; Kijjoa, A.; Rocha, E. Cytotoxic activity of Secondary Metabolites from Marine-derived Fungus Neosartorya siamensis in Human Cancer Cells. Phytother. Res. 2016, 30, 1862–1871. [Google Scholar] [CrossRef]
  47. Ramos, A.A.; Castro-Carvalho, B.; Prata-Sena, M.; Dethoup, T.; Buttachon, S.; Kijjoa, A.; Rocha, E. Crude Extracts of Marine-derived and Soil Fungi of the Genus Neosartorya Exhibit Selective Anticancer Activity by Inducing Cell Death in Colon, Breast and Skin Cancer Cell Lines. Pharm. Res. 2016, 8, 8–15. [Google Scholar]
  48. Liang, W.L.; Le, X.; Li, H.J.; Yang, X.L.; Chen, J.X.; Xu, J.; Liu, H.; Wang, L.; Wang, K.; Hu, K.; et al. Exploring the Chemodiversity and Biological Activities of the Secondary Metabolites from the Marine Fungus Neosartorya pseudofischeri. Mar. Drugs 2014, 12, 5657–5676. [Google Scholar] [CrossRef]
  49. Lim, S.M.; Agatonovic-Kustrin, S.; Lim, F.T.; Ramasamy, K. High-performance thin layer chromatography-based phytochemical and bioactivity characterisation of anticancer endophytic fungal extracts derived from marine plants. J. Pharm. Biomed. Anal. 2021, 193, 113702. [Google Scholar] [CrossRef]
  50. Chen, L.; Fan, D.M.; Tang, J.W.; An, T.; Li, X.; Kong, L.M.; Puno, P.; Li, Y. Discovery of isopenicin A, a meroterpenoid as a novel inhibitor of tubulin polymerization. Biochem. Biophys. Res. Commun. 2020, 525, 303–307. [Google Scholar] [CrossRef]
  51. Vinale, F.; Salvatore, M.M.; Nicoletti, R.; Staropoli, A.; Manganiello, G.; Venneri, T.; Borrelli, F.; DellaGreca, M.; Salvatore, F.; Andolfi, A. Identification of the Main Metabolites of a Marine-Derived Strain of Penicillium brevicompactum Using LC and GC MS Techniques. Metabolites 2020, 10, 55. [Google Scholar] [CrossRef] [Green Version]
  52. Abou Zied, A.M.; Abd El-Zaher, E.H.; Elsharquawy, H.A.; Hammad, T. Exopolysaccharides Production and Characterization from Marine-Derived Penicillium commune KP942881.1 with Some Medical Potential Applications. Egypt. J. Bot. 2017, 57, 17–30. [Google Scholar] [CrossRef]
  53. Ali, T.; Inagaki, M.; Chai, H.; Wieboldt, T.; Rapplye, C.; Rakotondraibe, L.H. Halogenated Compounds from Directed Fermentation of Penicillium concentricum, an Endophytic Fungus of the Liverwort Trichocolea tomentella. J. Nat. Prod. 2017, 80, 1397–1403. [Google Scholar] [CrossRef] [PubMed]
  54. Dikmen, M.; Canturk, Z.; Tilki, E.K.; Engur, S. Evaluation of Antiangiogenic and Antimetastatic Effects of Penicillium chrysogenum Secondary Metabolites. Indian J. Pharm. Sci. 2017, 79, 49–57. [Google Scholar] [CrossRef]
  55. Liu, Z.G.; Bao, L.; Liu, H.W.; Ren, J.W.; Wang, W.Z.; Wang, L.; Li, W.; Yin, W. Chemical diversity from the Tibetan Plateau fungi Penicillium kongii and P. brasilianum. Mycology 2018, 9, 10–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Canturk, Z.; Artagan, Ö.; Dıkmen, M. Anticancer effects of secondary metabolites of Penicillium chrysogenum var. chrysogenum on colon adenocarcinoma cells. Fresenius Environ. Bull. 2016, 25, 6190–6197. [Google Scholar]
  57. Li, C.S.; Li, X.M.; Gao, S.S.; Lu, Y.H.; Wang, B.G. Cytotoxic Anthranilic Acid Derivatives from Deep Sea Sediment-Derived Fungus Penicillium paneum SD-44. Mar. Drugs 2013, 11, 3068–3076. [Google Scholar] [CrossRef] [PubMed]
  58. Kuriakose, G.C.; Lakshmanan, M.D.; Bp, A.; Rs, H.K.; Th, A.K.; Ananthaswamy, K.; C, J. Extract of Penicillium sclerotiorum an endophytic fungus isolated from Cassia fistula L. induces cell cycle arrest leading to apoptosis through mitochondrial membrane depolarization in human cervical cancer cells. Biomed. Pharm. 2018, 105, 1062–1071. [Google Scholar] [CrossRef]
  59. Nguyen, V.T.; Lee, J.S.; Qian, Z.J.; Li, Y.X.; Kim, K.N.; Heo, S.J.; Heo, S.J.; Jeon, Y.J.; Park, W.S.; Choi, I.W.; et al. Gliotoxin Isolated from Marine Fungus Aspergillus sp. Induces Apoptosis of Human Cervical Cancer and Chondrosarcoma Cells. Mar. Drugs 2013, 12, 69–87. [Google Scholar] [CrossRef]
  60. Tang, W.; Liu, Z.L.; Mai, X.Y.; Qi, X.; Li, D.H.; Gu, Q.Q. Identification of Gliotoxin isolated from marine fungus as a new pyruvate kinase M2 inhibitor. Biochem. Biophys. Res. Commun. 2020, 528, 594–600. [Google Scholar] [CrossRef]
  61. Chen, J.; Lou, Q.; He, L.; Wen, C.; Lin, M.; Zhu, Z.; Wang, F.; Huang, L.; Lan, W.; Iwamoto, A.; et al. Reduced-gliotoxin induces ROS-mediated anoikis in human colorectal cancer cells. Int. J. Oncol. 2018, 52, 1023–1032. [Google Scholar] [CrossRef] [Green Version]
  62. Reis, F.S.; Barros, L.; Calhelha, R.C.; Cirić, A.; van Griensven, L.J.L.D.; Soković, M.; Ferreira, I. The methanolic extract of Cordyceps militaris (L.) Link fruiting body shows antioxidant, antibacterial, antifungal and antihuman tumor cell lines properties. Food Chem. Toxicol. 2013, 62, 91–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Šeklić, D.S.; Stanković, M.S.; Milutinović, M.G.; Topuzović, M.D.; Štajn, A.Š.; Marković, S.D. Cytotoxic, antimigratory, pro-and antioxidative activities of extracts from medicinal mushrooms on colon cancer cell lines. Arch. Biol. Sci. 2016, 68, 93–105. [Google Scholar] [CrossRef] [Green Version]
  64. Lee, H.H.; Lee, S.; Lee, K.; Shin, Y.S.; Kang, H.; Cho, H. Anti-cancer effect of Cordyceps militaris in human colorectal carcinoma RKO cells via cell cycle arrest and mitochondrial apoptosis. DARU J. Pharm. Sci. 2015, 23, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Mollah, M.L.; Park, D.K.; Park, H.J. Cordyceps militaris Grown on Germinated Soybean Induces G2/M Cell Cycle Arrest through Downregulation of Cyclin B1 and Cdc25c in Human Colon Cancer HT-29 Cells. Evid. Based Complement. Alternat. Med. 2012, 2012, e249217. [Google Scholar] [CrossRef] [Green Version]
  66. Katoch, M.; Singh-Ranger, G.; Sharma, S.; Gupta, N.; Sangwan, P.L.; Saxena, A.K. Cytotoxic and antimicrobial activities of endophytic fungi isolated from Bacopa monnieri (L.) Pennell (Scrophulariaceae). BMC Complement. Altern. Med. 2014, 14, 52. [Google Scholar] [CrossRef] [Green Version]
  67. Mohammed, A.E.; Sonbol, H.; Alwakeel, S.S.; Alotaibi, M.O.; Alotaibi, S.; Alothman, N.; Suliman, R.S.; Ahmedah, H.T.; Ali, R. Investigation of biological activity of soil fungal extracts and LC/MS-QTOF based metabolite profiling. Sci. Rep. 2021, 11, 4760. [Google Scholar] [CrossRef]
  68. Venugopalan, A.; Potunuru, U.R.; Dixit, M.; Srivastava, S. Reprint of: Effect of fermentation parameters, elicitors and precursors on camptothecin production from the endophyte Fusarium solani. Bioresour. Technol. 2016, 213, 311–318. [Google Scholar] [CrossRef]
  69. Riera, P.; Páez, D. Elucidating the role of pharmacogenetics in irinotecan efficacy and adverse events in metastatic colorectal cancer patients. Expert. Opin. Drug Metab. Toxicol. 2021, 17, 1157–1163. [Google Scholar] [CrossRef]
  70. Setyowati, E.P.; Pratiwi, S.U.T.; Purwantini, I. In-vitro cytotoxicity and apoptosis mechanism of ethyl acetate extract from Trichoderma reesei strain TV221 associated with marine sponge: Stylissa flabelliformis. J. Appl. Pharm. Sci. 2018, 8, 151–157. [Google Scholar]
  71. Cui, J.; Shang, R.Y.; Sun, M.; Li, Y.X.; Liu, H.Y.; Lin, H.W.; Jiao, W.H. Trichodermaloids A–C, Cadinane Sesquiterpenes from a Marine Sponge Symbiotic Trichoderma sp. SM16 Fungus. Chem. Biodivers. 2020, 17, e2000036. [Google Scholar] [CrossRef]
  72. Yamazaki, H.; Rotinsulu, H.; Kaneko, T.; Murakami, K.; Fujiwara, H.; Ukai, K.; Namikoshi, M. A New Dibenz[b,e]oxepine Derivative, 1-Hydroxy-10-methoxy-dibenz[b,e]oxepin-6,11-dione, from a Marine-Derived Fungus, Beauveria bassiana TPU942. Mar. Drugs 2012, 10, 2691–2697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Li, Y.; Guo, S.; Zhu, H. Statistical optimization of culture medium for production of exopolysaccharide from endophytic fungus Bionectria ochroleuca and its antitumor effect in vitro. EXCLI J. 2016, 15, 211–220. [Google Scholar] [PubMed]
  74. Dornetshuber-Fleiss, R.; Heffeter, P.; Mohr, T.; Hazemi, P.; Kryeziu, K.; Seger, C.; Berger, W.; Lemmens-Gruber, R. Destruxins: Fungal-derived cyclohexadepsipeptides with multifaceted anticancer and antiangiogenic activities. Biochem. Pharm. 2013, 86, 361–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. El-Khawaga, M.A.; El-Aziz, M.M.A.; Hegazi, G. Identification and bioactive potential of endophytic fungi isolated from Calotropis procera (Ait.) R. Br. Life Sci. J. 2013, 10, 2804–2814. [Google Scholar]
  76. Heilos, D.; Rodríguez-Carrasco, Y.; Englinger, B.; Timelthaler, G.; Van Schoonhoven, S.; Sulyok, M.; Boecker, S.; Süssmuth, R.; Heffeter, P.; Lemmens-Gruber, R.; et al. The Natural Fungal Metabolite Beauvericin Exerts Anticancer Activity In Vivo: A Pre-Clinical Pilot Study. Toxins 2017, 9, 258. [Google Scholar] [CrossRef] [Green Version]
  77. Shweta, S.; Gurumurthy, B.R.; Ravikanth, G.; Ramanan, U.S.; Shivanna, M.B. Endophytic fungi from Miquelia dentata Bedd., produce the anti-cancer alkaloid, camptothecine. Phytomedicine 2013, 20, 337–342. [Google Scholar] [CrossRef]
  78. Fang, Z.F.; Yu, S.S.; Zhou, W.Q.; Chen, X.G.; Ma, S.G.; Li, Y.; Qu, J. A new isocoumarin from metabolites of the endophytic fungus Alternaria tenuissima (Nees & T. Nees: Fr.) Wiltshire. Chin. Chem. Lett. 2012, 23, 317–320. [Google Scholar]
  79. El-Elimat, T.; Figueroa, M.; Raja, H.A.; Graf, T.N.; Swanson, S.M.; Falkinham, J.O.; Wani, M.C.; Pearce, C.J.; Oberlies, N.H. Biosynthetically Distinct Cytotoxic Polyketides from Setophoma terrestris. Eur. J. Org. Chem. 2015, 2015, 109–121. [Google Scholar] [CrossRef] [Green Version]
  80. Dezaire, A.; Marchand, C.H.; Vallet, M.; Ferrand, N.; Chaouch, S.; Mouray, E.; Larsen, A.K.; Sabbah, M.; Lemaire, S.D.; Prado, S.; et al. Secondary Metabolites from the Culture of the Marine-derived Fungus Paradendryphiella salina PC 362H and Evaluation of the Anticancer Activity of Its Metabolite Hyalodendrin. Mar. Drugs 2020, 18, 191. [Google Scholar] [CrossRef] [Green Version]
  81. Li, H.; Xiao, J.; Gao, Y.Q.; Tang, J.; Zhang, A.L.; Gao, J.M. Chaetoglobosins from Chaetomium globosum, an Endophytic Fungus in Ginkgo biloba, and Their Phytotoxic and Cytotoxic Activities. J. Agric. Food Chem. 2014, 62, 3734–3741. [Google Scholar] [CrossRef]
  82. Wang, F.; Jiang, J.; Hu, S.; Ma, H.; Zhu, H.; Tong, Q.; Cheng, L.; Hao, X.; Zhang, G.; Zhang, Y. Secondary metabolites from endophytic fungus Chaetomium sp. induce colon cancer cell apoptotic death. Fitoterapia 2017, 121, 86–93. [Google Scholar] [CrossRef] [PubMed]
  83. Hu, S.; Yin, J.; Yan, S.; Hu, P.; Huang, J.; Zhang, G.; Wang, F.; Tong, Q.; Zhang, Y. Chaetocochin J, an epipolythiodioxopiperazine alkaloid, induces apoptosis and autophagy in colorectal cancer via AMPK and PI3K/AKT/mTOR pathways. Bioorganic. Chem. 2021, 109, 104693. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, S.; Ren, F.; Niu, S.; Liu, X.; Che, Y. Dioxatricyclic and Oxabicyclic Polyketides from Trichocladium opacum. J. Nat. Prod. 2014, 77, 9–14. [Google Scholar] [CrossRef] [PubMed]
  85. El-Elimat, T.; Raja, H.; Figueroa, M.; Swanson, S.M.; Falkinham, J.O., III; Lucas, D.M.; Grever, M.R.; Wani, M.C.; Pearce, C.J.; Oberlies, N.H. Sorbicillinoid analogs with cytotoxic and selective anti-Aspergillus activities from Scytalidium album. J. Antibiot. 2015, 68, 191–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Gokul Raj, K.; Manikandan, R.; Arulvasu, C.; Pandi, M. Anti-proliferative effect of fungal taxol extracted from Cladosporium oxysporum against human pathogenic bacteria and human colon cancer cell line HCT 15. Spectrochim. Acta A Mol. Biomol. Spectrosc. 2015, 138, 667–674. [Google Scholar] [CrossRef] [PubMed]
  87. González-Montiel, G.A.; Kaweesa, E.N.; Feau, N.; Hamelin, R.C.; Stone, J.K.; Loesgen, S. Chemical, Bioactivity, and Biosynthetic Screening of Epiphytic Fungus Zasmidium pseudotsugae. Molecules 2020, 25, 2358. [Google Scholar] [CrossRef]
  88. Yang, M.H.; Baek, S.H.; Ha, I.J.; Um, J.Y.; Ahn, K.S. Brassinin enhances the anticancer actions of paclitaxel by targeting multiple signaling pathways in colorectal cancer cells. Phytother. Res. 2021, 35, 3875–3885. [Google Scholar] [CrossRef]
  89. He, Y.; Ye, M.; Jing, L.; Du, Z.; Surhio, M.M.; Xu, H.; Li, J. Preparation, characterization and bioactivities of derivatives of an exopolysaccharide from Lachnum. Carbohydr. Polym. 2015, 117, 788–796. [Google Scholar] [CrossRef]
  90. Azevedo, L.; Chagas-Paula, D.A.; Kim, H.; Roque, A.C.M.; Días, K.S.T.; Machado, J.C.; Azevedo, M.G.; Mertens-Talcott, S.U. White mold (Sclerotinia sclerotirum), friend or foe: Cytotoxic and mutagenic activities in vitro and in vivo. Food Res. Int. 2016, 80, 27–35. [Google Scholar] [CrossRef]
  91. Pressete, C.G.; Giannini, L.S.V.; de Paula, D.A.C.; do Carmo, M.A.V.; Assis, D.M.; Santos, M.F.C.; Machado, J.C.; Marques, M.J.; Soares, M.G.; Azevedo, L. Sclerotinia Sclerotiorum (White Mold): Cytotoxic, Mutagenic, and Antimalarial Effects In Vivo and In Vitro. J. Food Sci. 2019, 84, 3866–3875. [Google Scholar] [CrossRef]
  92. Ding, B.; Yuan, J.; Huang, X.; Wen, W.; Zhu, X.; Liu, Y.; Li, H.; Lu, Y.; He, L.; Tan, H.; et al. New Dimeric Members of the Phomoxanthone Family: Phomolactonexanthones, A, B and Deacetylphomoxanthone C Isolated from the Fungus Phomopsis sp. Mar. Drugs 2013, 11, 4961–4972. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, C.; Sun, Y.; Mao, Q.; Guo, X.; Li, P.; Liu, Y.; Xu, N. Characteristics and Antitumor Activity of Morchella esculenta Polysaccharide Extracted by Pulsed Electric Field. Int. J. Mol. Sci. 2016, 17, 986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tang, Y.; Chen, J.; Li, F.; Yang, Y.; Wu, S.; Ming, J. Antioxidant and Antiproliferative Activities of Modified Polysaccharides Originally Isolated from Morchella Angusticepes Peck. J. Food Sci. 2019, 84, 448–456. [Google Scholar] [CrossRef] [PubMed]
  95. Arora, D.; Kumar, A.; Gupta, P.; Chashoo, G.; Jaglan, S. Preparation, characterization and cytotoxic evaluation of bovine serum albumin nanoparticles encapsulating 5-methylmellein: A secondary metabolite isolated from Xylaria psidii. Bioorg. Med. Chem. Lett. 2017, 27, 5126–5130. [Google Scholar] [CrossRef]
  96. Wang, G.; Fan, J.Y.; Zhang, W.J.; Hua, C.P.; Chen, C.J.; Yan, W.; Ge, H.M.; Jiao, R.H.; Tan, R.X. Polyketides from Mantis-Associated Fungus Daldinia eschscholzii IFB-TL01. Chem. Biodivers. 2015, 12, 1349–1355. [Google Scholar] [CrossRef]
  97. Gubiani, J.R.; Nogueira, C.R.; Pereira, M.D.P.; Young, M.C.M.; Ferreira, P.M.P.; de Moraes, M.O.; Pessoa, C.; Bolzani, V.S.; Araujo, A.R. Rearranged Sesquiterpenes and Branched Polyketides Produced by the Endophyte Camarops sp. Phytochem. Lett. 2016, 17, 251–257. [Google Scholar] [CrossRef] [Green Version]
  98. Száva-Kováts, E. Unfounded attribution of the “half-life” index-number of literature obsolescence to Burton and Kebler: A literature science study. J. Am. Soc. Inf. Sci. 2002, 53, 1098–1105. [Google Scholar] [CrossRef]
  99. Cohen, J. Weighted kappa: Nominal scale agreement with provision for scaled disagreement or partial credit. Psychol. Bull. 1968, 70, 213–220. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of the eligible studies included in this systematic review (A). Graphical representation of the method of extraction used to obtain the extract that was analyzed against tumor cells (B).
Figure 1. Flow diagram of the eligible studies included in this systematic review (A). Graphical representation of the method of extraction used to obtain the extract that was analyzed against tumor cells (B).
Pharmaceuticals 16 00022 g001
Figure 2. Mechanisms of action through which the different fungi of Ascomycota perform their effect as against CRC.
Figure 2. Mechanisms of action through which the different fungi of Ascomycota perform their effect as against CRC.
Pharmaceuticals 16 00022 g002
Table 1. Antitumor activity of the extracts or isolated compounds from Eurotiales order in CRC cancer cell lines.
Table 1. Antitumor activity of the extracts or isolated compounds from Eurotiales order in CRC cancer cell lines.
GenusIsolated fromExtractionIsolated CompoundsCell Line/Administration/Cytotoxicity AssayCompound and IC50 or Cell Death (%)Mechanism of ActionReference
AspergillusSubmerged decaying woodMethanol/
Dichloromethane
Asperphenins A (Asp. A)
Asperphenins B (Asp. B)
RKO
Alone and in combination with irinotecan and TXL
SRB
Asp. A: 0.84 µM
Asp. B: 1.26 µM
Asp. A + Irinotecan at 1.25, 2.5, 5 and 10 µM (0.652, 0.811, 0.756, 0.694 and 0.652 μg/mL, respectively)
Asp. A + TXL at 0.16, 0.8, 4 and 20µM (1.673, 1.925, 0.742 and 1.185 μg/mL, respectively)
Asp. A inhibits tubulin polymerization, generates ROS and induce G2/M arrest and apoptosis
Asp. A and Irinotecan show synergism
[20]
AspergillusNeopetrosia chaliniformisEthyl acetate (EtOAc)-HCT-116
-
MTT
Fractions I (193.64 μg/mL), II (5.28 μg/mL), III (15.82 μg/mL), IV
(10.27 μg/mL), V (45.57 μg/mL)
-[33]
Aspergillus
Penicillium
Stylissa carteri
Sediment
Hyrtios erectus
Marine sediment
Liquid medium:
EtOAc
Mycelia:
Methanol
-HCT-116
DMSO
SRB
Penicillium Mycelia (M): between 15.00 and 92.60 μg/mL; Broth (B): between 74.20 and >100 μg/mL
Aspergillus M: between 48.00 and 97.00 μg/mL; B: between 42.75 and 90.00 μg/mL
-[32]
AspergillusBruguiera gymnorrhyzaEtOAc-HCT-116
Alone
MTT
EtOAc extract (EAE): 10.1 μM-[15]
AspergillusAchillea fragrantissimaEthanol-HCT-116
DMSO
MTT
Subfractions N. Hexane (76 μg/mL), EtOAc (26.3 μg/mL), Butanol (89.1 μg/mL)-[24]
AspergillusMarine AlgaEtOAcAllianthrone A-CHCT-116
DMSO
MTT
Allianthrone A, B and C: >20 μM-[34]
Aspergillus-Liquid medium:
Methanol
Mycelia:
EtOAc
Aspergiside B (1)
Aspergisidone (2)
Emeguisin A (3)
Folipastatin (4)
Aspergillusidone C (5)
Unguinol (6)
2-Chlorounguinol (7)
2,4-Dichlorounguinol (8)
Nidulin (9)
HCT-116
-
MTT
(1): 3.98%; (2): 20.19%; (3): 23.5 µM; (4): 53.69%; (5): 7.84%; (6): 21.59%; (7): 4.92%; (8): 52.49%; (9): 3.58%-[31]
AspergillusSpongeEtOAcViolaceimide A-EHCT-8
-
MTT
Violaceimides A (1.5 μM), B (2.51 μM), C, D (>20 μM), E (>100 μM)-[35]
AspergillusSinularia sp.EtOAc
Methanol
Dichloromethane
-HCT-116
DMSO
MTT
CH2Cl2 extract: 15.8%
Fractions 1 (29%), 2 (38%), 2c (88%), 2d (85%)
-[19]
AspergillusSoilEtOAc-HCT-116
DMSO
SRB
A. niger (69.1%), A. nomius (68.2%), A. terreus (63.7%), A. fumigatus (60.3%), A. flavus (55.7%), A. candidus (55.1%), A. stellifer (51.9%), A. oryzae (50.7%), A. violaceus (50.1%)-[36]
AspergillusNeopetrosia chaliniformisEtOAc-WiDr
-
MTT
EAE of NC01 (87.89%), NC02 (102.43%), NC03 (70.98%), A. nomius (29.69%), NC07 (85.96%), NC08 (4.48%), NC09 (53.96%)EAE of NC01 induces apoptosis[37]
AspergillusLakeEtOAc-CaCo-2
DMSO
WST-1 and RTCA
EAE: 185.9 μg/mL-[38]
Aspergillus
Byssochlamys
Calotropis procera
Catharanthus roseus
Euphorbia prostrata
Vernonia amygdalina
EtOAc-HT-29 and HCT-116
-
MTT
A. terreus 1: HT-29 and HCT-116: >100 μg/mL
A. terreus 2: HT-29: >100 μg/mL, HCT-116: 30.7 μg/mL
Byssochlamys: HT-29: 56.3 μg/mL, HCT-116: 30.4 μg/mL
-[39]
AspergillusSand soilN-butyl alcoholMalformin CMC-38 and HCT-116
-
Methylene blue
Malformin C: 0.27 and 0.18 µM (MC-38 and HCT-116, respectively)Malformin C induces G2/M arrest, DNA damage, apoptosis, autophapy and necrosis[40]
AspergillusXenograpsus testudinatusLiquid medium:
EtOAc
Mycelia:
Methanol
Clavatustide BSW-480
-
CCK-8
Clavatustide B: 37%Clavatustide B inhibits G1/S phase cell cycle transit[28]
AspergillusSolar salternLiquid medium:
EtOAc
Mycelia:
Ethanol
Ergosterol (1)
Rosellichalasin (2)
Cytochalasin E (3)
RKO
DMSO
MTT
(1): 3.3 μM; (2): 62.3 μM; (3): 37.3 μM-[27]
AspergillusMalus hallianaEtOAcAsperterone B and CSW-1116
-
MTT
Asperterones B (57.5 µM), C (1.0 µM)-[41]
AspergillusEudistoma vannameiLiquid medium:
EtOAc
Mycelia:
Methanol
Isocoumarin (R)-mellein (1)
Penicillic acid (2)
cis-4-hydroxymellein (3)
trans-4-hydroxymellein (4)
HCT-8
DMSO
MTT
(1), (3) and (4): >25 μg/mL; (2): 8.76 μg/ml-[26]
AspergillusMarine sedimentEtOAcAcetylaranotin (1)
Acetylapoaranotin (2)
Deoxyapoaranotin (3)
HCT-116
-
MTT
(1): 21.2 μmol/L; (2): 13.8 μmol/L; (3): 52 μmol/LAll compounds induce caspases 3-, 9- and 8-dependent apoptosis[42]
Dichotomomyces
(Aspergillus)
Lobophytum crassumEtOAcPityriacitrinHCT-116
Alone
SRB
Pityriacitrin: 35.1 µM-[43]
Dichotomomyces
(Aspergillus)
Marine sedimentEtOAcBis(dethio)bis(methylsulfanyl)gliotoxin (1)
6-acetylbis(dethio)bis(methylsulfanyl)gliotoxin (2)
Acetylgliotoxin G (3)
Gliotoxin (4)
Acetylgliotoxin (5)
Fiscalin B (6)
HCT-116
DMSO
MTT
(1): 23.56 μg/mL; (2): 35.97 μg/mL; (3): 1.06 μg/mL; (4): 0.41 μg/mL; (5): >50 μg/mL; (6): 33.51 μg/mL-[44]
Emericella
Aspergillus)
SoilSonication, centrifugation
and lyophilization
-CaCo-2
-
MTT
Crude extract: 9.84 μg/mL-[25]
Eurotium
(Aspergillus)
-Ethanol1,8-Dihydroxy-3-methoxy-6-methyl- anthraquinoneHCT-116
DMSO
MTT
Eurotium extract: 125.0 μg/mL
1,8-Dihydroxy-3-methoxy-6-methyl- anthraquinone: 18.6 μg/mL
-[23]
Neosartorya
Aspergillus
Talaromyces
Aka coralliphaga
Porites lutea
Coastal forest soil
Rhabdermia sp.
Chondrilla australiensis
Clathria reianwardii
EtOAc-HCT-116 and HT29
Alone or combined with Dox
MTT
--[45]
NeosartoryaRumphella sp.EtOAcChevalone C (1)
Nortryptoquivaline (2)
Tryptoquivaline H (3)
Fiscalin A (4)
epi-fiscalin A (5) and C (6)
epi-neofiscalin A (7)
HCT-116
DMSO
MTT
(1): 153 μM; (2): 114 μM; (3): 202 μM; (4): 123 μM; (5): 277 μM, (6): 86 μM; (7): 203 μM-[46]
NeosartoryaAka coralliphaga
Coastal forest soil
Porites lutea
EtOAc-HCT-116 and HT-29
DMSO
MTT
Extracts 1 (HCT-116 and HT-29: >200 μg/mL), 2 (HCT-116: 139 μg/mL, HT-29: 200 μg/mL), 3 (HCT-116: 189 μg/mL, HT29: 196 μg/mL)-[47]
NeosartoryaAcanthaster planciEtOAc1,2,3,4-Tetrahydro-2-methyl-3-methylene-1,4-dioxopyrazino [1,2-a]índole (1)
1,2,3,4-Tetrahydro-2-methyl-1,3,4-trioxopyrazino [1,2-a]índole (2)
Gliotoxin (3)
Acetylgliotoxin (4)
Reduced gliotoxin (5)
6-Acetylbis(methylthio)gliotoxin (6)
Bisdethiobis(methylthio)gliotoxin (7)
Didehydrobisdethiobis(methylthio)gliotoxin (8)
Bis-N-norgliovictin (9)
HCT-116 and RKO
DMSO
MTS
(1): HCT-116: 10.34 μM, RKO: 33.56 μM; (2), (6), (8) and (9): HCT-116 and RKO: >50 μM; (3) and (4): HCT-116: 1.24 μM, RKO: 0.80 μM; (5): HCT-116: 0.89 μM, RKO: 1.24 μM; (7): HCT-116: 8.59 μM, RKO: 10.32 μM -[48]
PenicilliumSonneratia sp.EtOAc-HCT-116
DMSO
SRB
Eupenicillium sp.: 13.9 μg/mL
P. decumbens: 0.2 μg/mL
-[49]
PenicilliumIsodonEtOAcIsopenicin ASW-480
DMSO
MTT
Isopenicin A: 8.33 μmol/LIsopenicin A regulates cycle progression from G2 to M and induces apoptosis[50]
Penicillium-MethanolArenicolin AHCT-116
-
CellTiter-Glo
Arenicolin A: 7.3 μM-[21]
PenicilliumAnemonia sulcataEtOAccis-bis(methylthio)silvatinCaCo-2 and HCT-116
DMSO
MTT
cis-bis(methylthio)silvatin: HCT-116 29.29 µM, CaCo-2 35.31 µM-[51]
PenicilliumMarine water-ExopolysaccharidesCaCo-2
-
SRB
Exopolysaccharides: 3.21 mg/mL-[52]
PenicilliumTrichocolea tomentellaEtOAcEpoxydon (1)
3,6,8-trihydroxy-1-methylxanthone (2)
Gentisyl alcohol (3)
(R,S)-1-phenyl-1,2-ethanediol (4)
Dehydrodechlorogriseofulvin (5)
Dechlorogriseofulvin (6)
Griseofulvin (7)
Ethylene glycol benzoate (8)
Alternariol (9)
Griseoxanthone C (10)
Drimiopsin H (11)
Griseophenone B (12) and C (13)
HT-29
-
-
(1): 14.1 μM; (2), (4), (5), (6), (7), (8), (9), (10), (11), (12) and (13): >20 μM; (3): 6.4 μM-[53]
PenicilliumLakeEtOAc-CaCo-2
DMSO
WST-1 and RTCA-DP
EAE: 55.2 μg/mLEAE decreases angiogenesis and metastasis gene expression, cell migration and invasively[54]
PenicilliumPlant leafEtOAcKongiiline A (1), B (2)
Pebrolide (3)
1-deoxypebrolide (4)
Asperphenamate (5)
Asperphenamate B (6), C (7)
N-benzoyl-phenylalaninol (8)
Orsellinic acid (9)
Mycophenolic acid (10)
5,7-dihydroxy-4-methylphthalide (11)
HCT-116
DMSO
MTT
(1), (2), (3), (4), (8), (9) and (11): 100 μM; (5): 88.16 μM; (6): 77.68 μM; (7): 91.72 μM; (10): 36.92 μM-[55]
PenicilliumMarine sedimentLiquid medium:
Methanol
Mycelia:
80% acetone/
H2O
(–)-Brevianamide CHCT-116
DMSO
SRB
(–)-Brevianamide C: 15.6 µM-[29]
PenicilliumIsurus oxyrinchusLiquid medium:
EtOAc
Mycelia:
Methanol
Fructigenine A (1)
Verrucosidin (2)
Norverrucosidin (3)
HCT-116
-
Crystal violet
(1): 40.5 μg/mL; (2): 30.8 μg/mL; (3): 5.7 μg/ml-[30]
Penicillium-EtOAc-CaCo-2
DMSO
WST-1 and RTCA-DP
EAE: 102 μg/mLEAE induces apoptosis[56]
PenicilliumMarine sedimentAcetonePenipacids A and ERKO
-
MTT
Penipacids A (8.4 μM), E (9.7 μM)-[57]
PenicilliumTerminalia chebula RetzEtOAc
Ethanol
-CaCo-2
DMSO
MTT
EAE of IR-4 (55 μg/mL), IR-6 (44 μg/mL), IR-7 (67 μg/mL)
Ethanol extract of IR-6: 71 μg/mL
-[22]
SRB: sulforhodamine B; ROS: reactive oxygen species; TXL: paclitaxel; CH2Cl2 extract: sequential ethyl acetate, methanol, and dichloromethane extract; Dox: doxorubicin; CCK8: cell counting kit-8; WST-1: 4-[3-(4-Iodo-phenyl)-2-(4-nitrophenyl)-2H-5 tetrazolio]-1,3-benzene disulphonate; RTCA-DP: real-time cell analysis system; EAE: ethyl acetate extract; MTS: ((3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)) assay; mRNA: messenger RNA; MTT: 3-(4,5-dimethytlthiazol-2-yl)-2,5-diphenyltetrazolium bromide; IC50: half maximal inhibitory concentration; DMSO: dimethyl sulfoxide.
Table 2. Antitumor activity of the extracts or isolated compounds from Hypocreales order in CRC cancer cell lines.
Table 2. Antitumor activity of the extracts or isolated compounds from Hypocreales order in CRC cancer cell lines.
GenusIsolated fromExtractionIsolated CompoundsCell Line/Administration/Cytotoxicity AssayCompound and IC50 or Cell Death (%)Mechanism of ActionReference
BeauveriaMarine spongeAcetone1-Hydroxy-10-methoxy-dibenz[b, e]oxepin-6,11-dione (1)
Chrysazin (2)
Globosuxanthone A (3)
HCT-15
-
MTT
(1) and (2): >30 μM; (3): 10.7 μM-[72]
BionectriaPsidium guajavaEthanolExopolysaccharidesHT-29
Alone
MTT
Exopolysaccharides (84.58%)-[73]
Cordyceps-Methanol-HCT-116 and SW-480
DMSO
MTT
Extract: HCT-116: >250 μg/mL; SW-480: 178.70 μg/mLExtract reduces cell migration and cytoplasmic β-catenin[63]
Cordyceps-Ethanol-RKO
Distilled water
CCK-8
-Extract induces cell morphological changes, G2/M phase arrest and apoptosis[64]
Cordyceps-Methanol:water (80:20 v/v)-HCT-15
-
SRB
Extract: 72.57 μg/mL-[62]
Cordyceps-Methanol
Buthanol
-HT-29
Alone
EZ-CyTox kit
GSC (46.56%)
BuOH extract of C. militaris (36.23%)
GSC induces cell morphological changes and G2/M phase arrest, blocking the cyclin B1 and Cdc25c protein[65]
EngyodontiumTerminalia sp.Ethyl acetate (EtOAc)-HCT-116
DMSO
SRB
EtOAc extract (EAE): 2.5 μg/mL-[49]
FusariumSoilEtOAc-HCT-8 and HCT-116
DMSO
CellTiter-Glo assay
EAE: HCT-8: 0.3779 μg/mL, HCT-116: 15.86 μg/mL-[67]
Fusarium--CamptothecinCaCo-2
DMSO:Methanol (1:50)
Alamar blue assay
Standard camptothecin: 2.41 μM; Crude camptothecin extract: 0.591 μM-[68]
FusariumSoilSonication, centrifugation
and lyophilization
-CaCo-2
-
MTT
Crude extract: 6.24 μg/mL-[25]
Metarhizium-Liquid medium:
Filtration
Acetonitrile and sodium chloride
Destruxin A (1), B (2) and E (3)CaCo-2 and HCT-116
DMSO
MTT
(1): CaCo-2 (2.18 μM), HCT-116 (2.06 μM); (2): CaCo-2 (1.34 μM), HCT-116 (3.22 μM); (3): CaCo-2 (0.05 μM), HCT-116 (0.04 μM)(3) activates apoptotic caspases and induces ROS
All destruxins induce G0/G1 phase arrest in CaCo-2 cells, reduce cell migration, have antiangiogenic activity and interfered with the MAPK and/or PI3K/Akt signaling pathways
[74]
MyrotheciumCalotropis proceraLiquid medium: Chloroform and methanol (2:1 v/v)-HCT-116
DMSO
Crystal violet staining
Extract: 380 ng/mL-[75]
TrichodermaDysidea sp.EtOAcTrichodermaloid A (1), B (2) and C (3)
Aspergilloid G (4)
Rhinomilisin E (5) and G (6)
SW-620
DMSO
MTT
(1): 9.3 μM; (2): 8.6 μM; (3): 12.7 μM; (4) and (6): >32 μM; (5): 22.7 μM-[71]
TrichodermaPolygonum aviculareEthanol-HCT-116
DMSO
MTT
Fractions F2 (14.9 μg/mL), F4 (7.3 μg/mL), F5 (7.61 μg/mL)-[24]
TrichodermaStylissa
flabelliformis
EtOAc-WiDr
-
MTT
EAE: 88.88 μg/mLEAE induces apoptosis[70]
Trichoderma
Fusarium
Bacopa monnieriMethanol-HCT-116
DMSO
MTT
B1, B20, BX1: O and A (>100 μg/mL); T. aureoviride: O (11 μg/mL), A (>100 μg/mL); Fusarium sp. 6241: 5 μg/mL; F. oxysporum: O (22 μg/mL), A (98.68 μg/mL)-[66]
CCK8: cell counting kit-8; BuOH: buthanol extract; GSC: cordyceps militaris cultivated on germinated soybeans; SRB: sulforhodamine B; MTT: 3-(4,5-dimethytlthiazol-2-yl)-2,5-diphenyltetrazolium bromide; IC50: half maximal inhibitory concentration; DMSO: dimethyl sulfoxide; ROS: reactive oxygen species; O: organic residue; A: water extract; EAE: ethyl acetate extract; EE: ethanol extract.
Table 3. Antitumor activity of the extracts or isolated compounds from Pleosporales order in CRC cancer cell lines.
Table 3. Antitumor activity of the extracts or isolated compounds from Pleosporales order in CRC cancer cell lines.
GenusIsolated fromExtractionIsolated CompoundsCell Line/
/Administration
Cytotoxicity Assay
Compound and IC50 or Cell Death (%)Reference
AlternariaMiquelia dentataMethanol
Ethyl acetate (EtOAc)
-SW-480 and HCT-116
-
Hoechst 33342
Methanol extract: HCT-116 (5.39 μg/mL), SW-480 (12.37 μg/mL); EAE: HCT-116 (6.59 μg/mL), SW-480 (7.2 μg/mL) [77]
AlternariaErythrophleum fordiiEthanol(6aR, 6bS, 7S)-3, 6a, 7, 10-tetra-hydroxy-4, 9-dioxo-4, 6a, 6b, 7, 8, 9-hexahydroperylene (1)HCT-8
DMSO
MTT
(1): 1.78 μmol/L[78]
BipolarisSoilEtOAc-HCT-8 and HCT-116
DMSO
CellTiter-Glo assay
Extract: HCT-8 (202.5 μg/mL), HCT-116 (18.97 μg/mL)[67]
Drechslera-Ethanoldi-2-ethylhexyl phthalateHCT-116
DMSO
MTT
Drechslera extract: 104.0; di-2-ethylhexyl phthalate: 9.5 [23]
ParadendryphiellaPomacea canaliculataEtOAc(3R, 6R) hyalodendrinSW-48, DLD-1, LS513, LOVO, RKO, LS174T, SW-480 HT-29 and HCT-116
DMSO
MTT
(3R, 6R) hyalodendrin: SW-48 (149.0 nM), DLD-1 (40.0 nM), HT-29 (58.0 nM), HT-29 5FU (146.8 nM), HT-29 oxa (141.8 nM), HT-29 SN-38 (93.8 nM), HCT-116 (48.0 nM), HCT-116 5FU (72.0 nM), HCT-116 oxa (25.7 nM), HCT-116 SN-38 (43.8 nM), LS513 (78.0 nM), LOVO (73.4 nM), RKO (74.3 nM), LS174T (158.0 nM), SW-480 (163.7 nM)[80]
Phoma
Curvularia
Pleosporales sp.
Alternaria
Catharanthus roseus
Euphorbia prostrata
Calotropis procera
EtOAc-HT-29 and HCT-116
-
MTT
C. aeria extract: HT29 (74.5 μg/mL), HCT-116 (53.9 μg/mL); Pleosporales sp. extract: HT29 (69.4 μg/mL), HCT-116 (36.7 μg/mL); P. multirostrata extract: HT29 and HCT-116 (>100 μg/mL); C. australiensis extracts 1: HT29 (54.3 μg/mL), HCT-116 (25.6 μg/mL); 2: HT29 (>100 μg/mL), HCT-116 (59.7 μg/mL); A. alternata extract: HT29 (>100 μg/mL), HCT-116 (52.5 μg/mL); Alternaria sp. extract: HT29 (28.4 μg/mL), HCT-116 (29.1 μg/mL)[39]
Pleosporales sp.Bacopa monnieriDichloromethane-HCT-116
DMSO
MTT
CK01: O (12 μg/mL), A (>100 μg/mL)[66]
SetophomaLeaf litter Methanol/
Chloroform (1:1)
Secalonic acid A (1), E (2) and G (3)
Penicillixanthone A (4) and B (5)
Blennolide J (6)
Hypothemycin (7)
SW-620
DMSO
CellTiter 96 AQueous One Solution Cell Proliferation Assay
(1): 0.41 μM; (2): 19.12 μM; (3): 3.67 μM; (4): 0.21 μM); (5): 5.55 μM; (6): 6.14 μM; (7): 2.14 μM[79]
HCT-116/HT-29 oxa: oxaliplatin resistant cell; HCT-116/HT-29 5FU: 5-fluorouracil resistant cell; HCT-116/HT-29 SN-38: SN-38 resistant cell; MTT: 3-(4,5-dimethytlthiazol-2-yl)-2,5-diphenyltetrazolium bromide; IC50: half maximal inhibitory concentration; DMSO: dimethyl sulfoxide; CK01: Pleosporales sp. extract; O: organic residue; A: water extract; SN-38: 7-Ethyl-10-hydroxycamptothecin; EAE: ethyl acetate extract.
Table 4. Antitumor activity of the extracts or isolated compounds from Sordariales order in CRC cancer cell lines.
Table 4. Antitumor activity of the extracts or isolated compounds from Sordariales order in CRC cancer cell lines.
GenusIsolated fromExtractionIsolated CompoundsCell Line/
/Administration Cytotoxicity Assay
Compound and IC50 or Cell Death (%)Mechanism of ActionReference
Chaetomium-Liquid medium:
Ethyl acetate (EtOAc)
Mycelia:
Methanol
Chaetocochin (Ch.) J RKO, SW-480 and HCT-116
-
CCK8
Ch. J: RKO (0.56 μM), SW-480 (0.61 μM), HCT-116 (0.65 μM)Ch. J. induces apoptosis, autophagy and activates AMPK and PI3K/AKT/mTOR signaling pathway[83]
Chaetomium-EtOAc-HCT-116
DMSO
SRB
C. globosum extract: 1.2 μg/ml-[49]
ChaetomiumSoilEtOAc-HCT-8 and HCT-116
DMSO
CellTiter-Glo assay
Ethyl acetate extract (EAE): HCT-8 (8.744 μg/mL), HCT-116 (152.8 μg/mL)-[67]
ChaetomiumCymbidium goeringiiLiquid medium:
EtOAc
Mycelia:
Methanol
Ch. A and CSW-480
Alone
MTT
Ch. A: 15.21 μM
Ch. C: 0.63 μM
Ch. C. induces G2/M arrest, apoptosis, activation of the caspase 3 and PARP degradation, increased Bax and decreased Bcl-2 level[82]
ChaetomiumTrigonella foenum-graecumEtOAc-HT-29 and HCT-116
-
MTT
C. globosum extract: HT-29 (>100 μg/mL), HCT-116 (75.2 μg/mL) -[39]
ChaetomiumGinkgo bilobaMethanolChaetoglobosin A (1), G (2), V (3), Vb (4), E (5), F (6), Fex (7), Fa (8)
20-dihydrochaetoglobosin A (9)
HCT-116
Alone
SRB
(1): 3.15 μM; (2): 65.6 μM; (3): 29.5 μM; (4): 18.4 μM; (5): >100 μM; (6): 17.8 μM; (7): 17.8 μM; (8): 5.85 μM; (9): 8.44 μM-[81]
TrichlocladiumSoilEtOAcTrichocladinols E (1), F (2) and G (3)SW-480 and HCT-116
DMSO
MTS
(1): SW-480 (54.9 μM), HCT-116 (48.8 μM); (2): SW-480 (51.9 μM), HCT-116 (56.6 μM); (3): SW-480 (43.6 μM), HCT-116 (41.7 μM)-[84]
Scytalidium-Methanol/Chloroform (1:1)[5’-formyl-2’-hydroxyl-4’-methoxy-(E,E)-sorbophenone (1)
Scalbucillin B (2)
1-(2’-hydroxy-4’-methoxy-5’-methylphenyl)-2,4-E,E-hexadien-1-one (3)
5′-formyl-2′-hydroxy-4′-methoxy-(E)-4-hexenophenone (4)
SW-620
DMSO
CellTiter 96® AQueous One Solution Cell Proliferation Assay
(1): 0.5 μM; (2): 16 μM; (3): 15.1 μM; (4): 2.5 μM-[85]
SRB: sulforhodamine B; CCK8: cell counting kit-8; MTS: ((3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)) assay; MTT: 3-(4,5-dimethytlthiazol-2-yl)-2,5-diphenyltetrazolium bromide; IC50: half maximal inhibitory concentration; DMSO: dimethyl sulfoxide; AMPK: AMP-activated protein kinase; PARP: poly (ADP-ribose) polymerase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Luque, C.; Cepero, A.; Perazzoli, G.; Mesas, C.; Quiñonero, F.; Cabeza, L.; Prados, J.; Melguizo, C. In Vitro Efficacy of Extracts and Isolated Bioactive Compounds from Ascomycota Fungi in the Treatment of Colorectal Cancer: A Systematic Review. Pharmaceuticals 2023, 16, 22. https://doi.org/10.3390/ph16010022

AMA Style

Luque C, Cepero A, Perazzoli G, Mesas C, Quiñonero F, Cabeza L, Prados J, Melguizo C. In Vitro Efficacy of Extracts and Isolated Bioactive Compounds from Ascomycota Fungi in the Treatment of Colorectal Cancer: A Systematic Review. Pharmaceuticals. 2023; 16(1):22. https://doi.org/10.3390/ph16010022

Chicago/Turabian Style

Luque, Cristina, Ana Cepero, Gloria Perazzoli, Cristina Mesas, Francisco Quiñonero, Laura Cabeza, Jose Prados, and Consolación Melguizo. 2023. "In Vitro Efficacy of Extracts and Isolated Bioactive Compounds from Ascomycota Fungi in the Treatment of Colorectal Cancer: A Systematic Review" Pharmaceuticals 16, no. 1: 22. https://doi.org/10.3390/ph16010022

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