Next Article in Journal
Localization of Sesquiterpene Lactones Biosynthesis in Flowers of Arnica Taxa
Next Article in Special Issue
HPLC-PDA Method for Quantification of Bioactive Compounds in Crude Extract and Fractions of Aucklandia costus Falc. and Cytotoxicity Studies against Cancer Cells
Previous Article in Journal
Inhibition of Mitochondrial Antioxidant Defense and CDK4/6 in Mesothelioma
Previous Article in Special Issue
Anticancer Activity of Chalcones and Its Derivatives: Review and In Silico Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Bacterial and Fungal Peptides as a Promising Treatment to Defeat Lung Cancer Cells

by
Kamila Rybczyńska-Tkaczyk
1,
Anna Grenda
2,*,
Anna Jakubczyk
3 and
Paweł Krawczyk
2
1
Department of Environmental Microbiology, The University of Life Sciences, Leszczyńskiego Street 7, 20-069 Lublin, Poland
2
Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego Street 8, 20-954 Lublin, Poland
3
Department of Biochemistry and Food Chemistry, University of Life Sciences in Lublin, Skromna Street 8, 20-704 Lublin, Poland
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(11), 4381; https://doi.org/10.3390/molecules28114381
Submission received: 27 March 2023 / Revised: 22 May 2023 / Accepted: 25 May 2023 / Published: 27 May 2023
(This article belongs to the Special Issue Natural Products in Anticancer Activity)

Abstract

:
Despite the increasing availability of modern treatments, including personalized therapies, there is a strong need to search for new drugs that will be effective in the fight against cancer. The chemotherapeutics currently available to oncologists do not always yield satisfactory outcomes when used in systemic treatments, and patients experience burdensome side effects during their application. In the era of personalized therapies, doctors caring for non-small cell lung cancer (NSCLC) patients have been given a powerful weapon, namely molecularly targeted therapies and immunotherapies. They can be used when genetic variants of the disease qualifying for therapy are diagnosed. These therapies have contributed to the extension of the overall survival time in patients. Nevertheless, effective treatment may be hindered in the case of clonal selection of tumor cells with acquired resistance mutations. The state-of-the-art therapy currently used in NSCLC patients is immunotherapy targeting the immune checkpoints. Although it is effective, some patients have been observed to develop resistance to immunotherapy, but its cause is still unknown. Personalized therapies extend the lifespan and time to cancer progression in patients, but only those with a confirmed marker qualifying for the treatment (gene mutations/rearrangements or PD-L1 expression on tumor cells) can benefit from these therapies. They also cause less burdensome side effects than chemotherapy. The article is focused on compounds that can be used in oncology and produce as few side effects as possible. The search for compounds of natural origin, e.g., plants, bacteria, or fungi, exhibiting anticancer properties seems to be a good solution. This article is a literature review of research on compounds of natural origin that can potentially be used as part of NSCLC therapies.

1. Introduction

According to the Globocan database, the estimated global number of new cases of cancer in 2020 in both sexes and all ages was 19,292,789. The estimated mortality due to cancer worldwide in the same year was 9,958,133 [1]. It is estimated that the number of new cases of cancer will have increased to about 30 million and the death rate will have risen to about 16 million cases by 2040. These statistics indicate that, despite the increasing availability of modern therapies, including personalized therapies (small molecule therapy and immunotherapy), there is a strong need to search for new drugs that will be effective in the fight against cancer. Plant substances or plant-based chemotherapeutics have become one of the most important elements of the systemic treatment of cancer patients. For example, paclitaxel is the best-known plant-derived chemotherapeutic agent from Taxomyces andreanae. It is extracted from the inner bark of Taxus brevifolia and is used in the treatment of lung, ovarian, and breast cancer [2]. Microbes are the other natural sources of anticancer substances. Antibiotics derived from microorganisms exert the following effects: alkylation and thus damage to the DNA of cancer cells (melphalan), inhibition of DNA replication (mitomycin), or interruption of cell mitosis (doxorubicin). These drugs are widely used in systemic anticancer treatment. Currently, clinicians can use modern molecularly targeted therapies or immune checkpoint-targeting therapies. In the former type, specific gene alterations predisposing a patient to inclusion in treatment with inhibitors of kinases involved in cellular signal transduction pathways are searched for in BRAF (B-Raf Proto-Oncogene, Serine/Threonine Kinase), MEK (Mitogen-Activated Protein Kinase Kinase 1), RAS (RAS Proto-Oncogene, GTPase), EGFR (Epidermal Growth Factor Receptor), RET (RET Proto-Oncogene), HER2 (Erb-B2 Receptor Tyrosine Kinase 2), ROS1 (ROS Proto-Oncogene 1, Receptor Tyrosine Kinase), ALK (Anaplastic Lymphoma Receptor Tyrosine Kinase), and NTRK (Neurotrophic Receptor Tyrosine Kinase). Depending on the type of cancer, different percentages of patients are diagnosed with genetic variants. Activating BRAF mutations are observed in approximately 50% of melanoma patients [3]. In these patients, the identification of BRAF alterations, in particular, the V600E variant, allows the use of BRAF inhibitors: vemurafenib, dabrafenib, or encorafenib and MEK inhibitors: cobimetinib, tremetnib, or binimetinib [3]. Activating genetic variants in the EGFR gene, qualifying for treatment with EGFR inhibitors (erlotinib, gefitinib, afatinib, dacomitinib, and osimertinib) are observed in 10–15% of Caucasian patients and in at least 40% of Asian non-small cell lung cancer (NSCLC) patients [4]. In turn, ALK and ROS1 rearrangements are detected in approximately 4.5% and 1% of non-small cell lung cancer patients, respectively [5,6]. In the presence of ALK driver alterations, ALK tyrosine kinase inhibitors (crizotinib, ceritinib, alectinib, brigatinib, ensartinib, and lorlatinib) may be used. In turn, the crizotinib or entrectinib can be used in patients with ROS1-positive NSCLC. Molecularly targeted treatment has also shifted towards ‘tissue agnostic’ drug treatment [7]. The assumption is based on a model where it does not matter in which type of cancer a given genetic variant occurs but on the fact that the treatment targeted at the protein product resulting from a given mutation is effective. This approach is highly beneficial when genetic variants are detected in a very small percentage of patients with different types of cancer, as is the case of ROS1 or NTRK1/2/3 rearrangements.
These percentages indicate that not all cancer patients can be subjected to targeted therapy. Another therapeutic approach is the immune checkpoint-targeting immunotherapy. The use of monoclonal antibodies targeted the PD-1 (Programmed Cell Death 1), PD-L1 (Programmed Cell Death 1 Ligand 1), or CTLA4 (Cytotoxic T-Lymphocyte Associated Protein 4) molecules has revolutionized the treatment of oncological patients, bringing significant benefits, i.e., an extension of survival time or improvement of the quality of patient’s life related to the absence of side effects such as those occurring in chemotherapy treatment [8,9]. This does not mean that the therapy has no flaws. Side effects may occur, but they are different than those induced by TKI inhibitors or chemotherapy. In addition, resistance to immunotherapies may be observed, but its causes are not fully understood [10]. Several mechanisms of resistance to immunotherapies have been identified, such as the absence of tumor antigens, defective antigen presentation, modulation of critical cellular pathways, epigenetic changes, and changes in the tumor microenvironment. The elucidation of these mechanisms has contributed to the implementation of the chemoimmunotherapy strategy, where chemotherapy results in the presentation of neoantigens on cancer cells, making them more visible to the immune system recognizing them by blocking the PD1/PD-L1 or CTLA4/B.7 pathways. The estimated percentage of US patients with cancer who are eligible for checkpoint inhibitor drugs in 2018 was 43.63%. The percentage of patients estimated to respond to ICI in 2018 was 12.46% [11].
All this indicates that chemotherapy will not be replaced by modern therapies with kinase inhibitors or immunotherapy but will still be an important element of the strategy of treatment in oncological patients. It should also be mentioned that chemotherapy is important as an adjuvant or neoadjuvant therapy. According to the ACS (American Cancer Society), in the USA, 56% of women with stage III breast cancer undergo surgical removal of the breast followed by chemotherapy [11]. As reported by the ALA (American Lung Association), over 20% of lung cancer patients are treated surgically. In addition, chemotherapy in combination with immunotherapy is still the only option for small cell lung cancer. Moreover, according to the NCI (National Cancer Institute), on average, 64.5% of patients (age range 20–80 years old) with stage IIIB or IV non-small cell lung cancer in 1996–2018 received some chemotherapy regiments. All this indicates that chemotherapy is still needed, and the search for new natural sources of chemotherapeutics may contribute to improvement of the effectiveness of treatment of oncological patients, including those with lung cancer, which ranks second in terms of the overall incidence in the world and first in terms of the cause of cancer-related death. The search for plant, bacterial, or fungal compounds seems to be a challenge that can improve chemotherapy outcomes. There are some papers on this topic, but still in vitro, in vivo, and clinical research is required [12,13,14].

2. Anticancer Peptides (ACPs)

One of the potential innovative therapeutic methods in the fight against cancer cells is the use of anticancer peptides (ACPs). Their properties are determined by the amino acid composition and the addition of functional groups, which affects their conformation, charge, stability, bioavailability, more efficient tumor/tissue penetration than antibodies, and effectiveness. ACPs are usually composed of 10–60 amino acid residues that can inhibit the migration and proliferation of tumor cells or inhibit the formation of tumor vasculature. The advantage of ACPs is their lower ability to cause drug resistance. On the other hand, they can be hydrolyzed by proteases or cause cytotoxicity [15]. These compounds are either natural or modified with positive cationic moieties or functional groups to enhance their therapeutic efficiency and reduce side effects. Additionally, peptides can be modified to attack tumor cells and prevent tumor progression [16]. They can be used in a variety of ways to treat cancer. Anticancer peptides can be applied as drugs or can deliver drugs to cancer cells. They also simulate natural proteins to enhance or inhibit signal transduction and mediate therapeutic transport across the barrier [16,17]. Peptides with anticancer properties can be synthesized in various cells and conjugated with other compounds and subfields in various ways. Some peptides can cross the blood–brain barrier and affect the central nervous system. This property seems to be extremely important in terms of the appearance of metastases in the head area in the advanced stages of lung cancer. The advantage of bioactive peptides is that their production is simpler than the production of proteins and antibodies. In addition to their simplicity and ease of transport, peptides have a longer shelf life [17]. Cancer cells differ from healthy cells in their membrane. The anticancer effect of peptides is based on the induction of cancer cell apoptosis and necrosis through membrane lysis or the formation of pores in the membrane [18]. Cholesterol is a component of the cell membrane protecting healthy cells from the lytic action of various factors by changing the fluidity of the cell membrane. Cancer cells exhibit higher fluidity of the cell membrane than healthy cells. Their more abundant microvilli increase their surface area. In addition, healthy cells are electrically neutral, while cancer cells contain a negatively charged component on their surface, which leads to membrane destabilization, cytotoxicity, and cell lysis during interactions with small molecules, e.g., ACPs [19]. The interactions between peptides and the healthy cell membrane are hydrophobic, while electrostatic interactions occur between peptides and the membrane of tumor cells [20]. Currently, extensive research is being conducted on the use of peptides in the treatment and diagnosis of many cancers to develop peptide therapy. The basis of this therapy is the use of selective targeting of tumor-specific receptors. Peptides that target the tumor are more efficient in the penetration of tumor tissues than antibodies. Additionally, chemical modifications can increase their stability and pharmacokinetics. Peptides have already been applied in diagnostics, cancer imaging, and targeted drug delivery [17].

2.1. Classification and Mechanism of Action of ACPs

Anticancer peptides are characterized by different structures, modes of action, selectivity, and efficacy against specific cancer cells. There are many ACPs derived from different species and characterized by different properties; hence, these compounds can be classified in different ways. The most common classification based on their structure has distinguished four categories of ACPs: α-helical, β-pleated sheets, random coil, and cyclic [15].
It has been shown that the hydrophobic amino acid residues in the alpha structure enhance their cationic properties, while the amphipathic properties of these structures play an important role in cytotoxicity against cancer cells [21]. The formation of disulfide bridges is necessary to maintain the β-pleated sheet structure. It is worth noting that ACPs with an α-helical structure have higher antitumor properties than β-pleated peptides and higher toxicity to normal tissue [15].
Another classification is based on the mechanism of action: molecularly targeted peptides, which directly act on cancer cells via cytotoxic, antiproliferative, and apoptotic activities, ‘guiding missile’ peptides, or binding peptides, which are drug-binding peptides used for transporting drugs into cancer cell targets, and cell-stimulating peptides that indirectly influence other stimulating cells to kill cancer cells, e.g., via immunomodulatory activities and hormone receptors [22,23].

2.2. Effect of Peptides on Lung Cancer Cells

Many studies indicate the possibility of using peptides in the treatment of lung cancer due to their size and properties. These peptides can be obtained from food and non-food protein or through synthesis, as shown by results of in vivo or in vitro studies. It was noted that tryptophan-containing peptides exhibited increased cytotoxicity against non-small cell lung adenocarcinoma A549 cells, and their hemolytic activity was found to increase as well [24].
A protein obtained from Porphyra haitanesis was shown to be a natural source of anticancer peptides. To obtain bioactive peptides, the protein was hydrolyzed using trypsin, and then the peptides were purified with the use of chromatographic techniques. Three peptides showing antiproliferative properties against five cancer cell lines: MCF-7, HepG-2, SGC-7901, A549, and HT-29 were isolated and their IC50 (half maximal inhibitory concentration) values were in the range from 191.61 to 316.95 μg/mL [25]. Wang and Zhang described a new peptide YGFVMPRSGLWFR obtained from papain hydrolysates of Spirulina (Arthrospira) platensis proteins, which exerted high inhibition activity against A549 cancer cells with the IC50 value of 104.05 μg/mL [26].
However, the exact relationship between the structure and action of peptides has not been thoroughly clarified. It is known that the presence of certain amino acid residues enhances the anticancer efficacy of peptides. To elucidate the relationship between the structure and activity of peptides, their synthetic analogs are being studied. Thus, an analog of alyteserin-2a (ILGKLLSTAAGLLSNL.NH2), i.e., a peptide isolated from the skin secretion of the midwife toad Alytes obstetricians, was studied. In the analog, the amino acids on the hydrophobic side of the helix were replaced by L-tryptophan and the hydrophilic amino acids were substituted by one or more L-lysine or D-lysine residues. The results indicated that the tryptophan-containing peptides had up to 11-fold higher cytotoxic activity against non-small cell lung adenocarcinoma cells A549, and their hemolytic activity against human erythrocytes increased as well. The activity of the N15K analog on A549 cells (LC50 = 13 µM) increased sixfold relative to alliterin-2a and the therapeutic index (LC50 ratio for erythrocytes and tumor cells) increased twofold. In contrast, the insertion of the 11 D-lysine residue in the N15K analog produced a peptide which retained potency against A549 cells (LC50 = 15 µM), with a 13-fold higher therapeutic index relative to the native peptide. Therefore, increasing hydrophobicity while maintaining amphipathicity via the insertion of the L-tryptophan peptide in the structure yielded a peptide with increased antitumor activity, compared to the starting peptide [24].
Patil and Kunda studied cationic antimicrobial peptide D-LAK-120A (KKLALALAKKWLALAKKLALALAKK-NH2) as an anticancer factor in various NSCLC cell lines [27]. Their results indicated cytotoxicity in concentrations between 4.0 and 5.5 µM against A549, H358, H1975, and HCC827 cell lines. An increase in lactate dehydrogenase (LDH) activity and propidium iodide (PI) uptake across the compromised membrane was suggested as an inhibition pathway. The study also demonstrated that D-LAK-120A was an inductor of lung cancer apoptosis and cell arrest in the S phase (DNA synthesis) of the cell cycle. Moreover, this peptide was shown to be involved in the in vitro inhibition of single-cell proliferation and cancer cell migration. The 3D spheroid indicated tumor reduction, which suggests the potential use of D-LAK-120A as an anticancer substance for non-small-cell lung cancer treatment and a potential therapeutic agent [27].
Lunasin, which consists of 43–44 amino acid residues with nine consecutive aspartic acids at the C-terminus, is a soybean peptide with indicated anticancer activity. It shows both chemopreventive and chemotherapeutic activities. Noteworthy, treatment of NSCLC cells with this peptide was limited by cell-line anti-proliferative effects on anchorage-dependent growth; on the other hand, two normal bronchial epithelial cell lines were unaffected. The study conducted in a murine model indicated that 30 mg lunasin/kg body weight per day decreased the NSCLC H1299 tumor volume by 63.0% on day 32. Moreover, this peptide inhibited cell cycle progression at the G1/S phase interface in the NSCLC H661 cells but did not induce apoptosis [28]. It was also shown that protein hydrolysates obtained from Enteromorpha prolifera may be a source of a peptide with anticancer properties. A heptapeptide with the amino acid sequence GPLGAGP exhibited potent antiproliferative activity toward several human cancer cell lines. The results of the study indicated that the IC50 values for NCI-H460, HepG2, and A549 were 0.3686, 1.2564, and 0.9867 mg/mL, respectively. This peptide-induced cell apoptosis in a dose-dependent manner [29].
Peptides are also used to support the action of anticancer drugs. Nanoconjugates of alendronate sodium (potent inhibitor of farnesyl pyrophosphate synthase) with mastopran (potent anticancer polypeptide isolated from wasp venom) (ALS-MP) were tested as potential compounds to be used in the inhibition of lung cancer. The results showed a lower IC50 value (1.3 ± 0.34 μM) compared to ALS (IC50 37.6 ± 1.79 μM). Moreover, after treatment with the ALS-MP nanoconjugates, a higher percentage of cells in the G2-M phase was noted during cell cycle analysis [30]. Cisplatin is used as a standard and effective drug in lung cancer chemotherapy. To enhance the chemotherapeutic effects of cisplatin, the potential of a peptide from Lentinus squarrosulus (Mont.) used as an adjuvant was assessed. The results indicated that 24 h preincubation with 5 μg/mL of the peptide prior to treatment with 5 μM cisplatin significantly diminished % cell viability in various human lung cancer cells. However, it did not reduce the lifespan in the human dermal papilla and proximal renal cells. Flow cytometry indicated augmentation of cisplatin-induced apoptosis in the lung cancer cells. The peptide-pretreated lung cancer cells showed enhanced cisplatin-induced apoptosis and inhibition of colony formation. These data indicate the possibility of using a new combination therapy based on platinum compounds in the treatment of lung cancer [31].

3. Compounds of Bacterial Origin, Including Peptides, against Lung Cancer Cells

The anticancer activities of metabolites from many bacteria have been shown in recent years, but researchers are testing various niches that can be a source of bacterial anticancer substances. Examples of compounds of bacterial origin showing anticancer abilities against lung cancer cells are summarized in Table 1.
Silva et al. conducted their research on Deschampsia antarctica as a reservoir and source of microorganisms that produce metabolites with anticancer properties [32]. They performed next-generation sequencing with analysis of the 16S rRNA gene. They selected Streptomyces sp. CMAA 1527 and Streptomyces sp. CMAA 1653 as candidates for producing natural compounds with the potential to control the proliferation of breast (MCF-7), glioblastoma (U251), non-small cell lung (NCI-H460), and kidney (786-0) human cancer cell lines. The analyses indicated that Cinerubin B and actinomycin V were the predominant compounds identified in Streptomyces sp. CMAA 1527 and Streptomyces sp. CMAA 1653, respectively, which had anticancer properties [32]. Cinerubin B is an anthracycline antibiotic, but its importance as an anticancer agent has not yet been extensively studied. However, there are studies on actinomycin V which show cytotoxic activity and inhibition of proliferation, invasion, and migration of cancer cells, i.e., processes related to high expression of vimentin and E-cadherin [33,34]. As reported by Lin et al., actinomycin V caused up-regulation of p53, which suppressed the growth of lung cancer (A549) cells and induced cell cycle arrest and apoptosis. The cytotoxic activity of actinomycin V against the A549 cells with wild-type p53 was stronger than against NCI-H1299 cells (p53-deficient lung cancer cells). Actinomycin V decreased the expression of such M-phase related proteins as CdC2 (Cell Division Control Protein 2 Homolog), CdC25A (Cell Division Cycle 25A), and CCNB1 (Cyclin B1) arrested the cells in the G2/M phase, and subsequently triggered apoptosis by mediating the expression of the Bcl-2 family proteins (Bax and Bcl-2) [33]. Other results obtained by Lin et al. showed that actinomycin V treatment significantly down-regulated the levels of N-cadherin and vimentin expression, which promotes cellular proliferation, invasion, and migration of many cancer cells, including lung cancer lines [34]. Therefore, Actinomycin V may block the passage of EMT in cancer cells by reducing cell invasion and migration [34].
Sharma et al. studied the anticancer activity of enterocin from Enterococcus faecium 12a culture supernatant [35]. They found that enterocin 12a inhibited the growth of cell lines in a dose-dependent manner, and the 50% inhibitory concentration of this enterocin in lung cancer cell lines was 0.08 μg/mL [35]. The authors suggest that enterocin 12 should be tested in in vivo experiments as a potential anticancer agent. Arunmanee et al. tested the anticancer activity of colicin N [36]. Colicin N is a bacteriocin (peptide) produced by Escherichia coli that acts against other E. coli strains. It kills bacteria via the formation of pores in the cell membrane, leading to membrane depolarization and cell destruction. It can bind to receptors and induce formation of pores in the cell membrane. The authors found that treatment of human lung cancer H460, H292, and H23 cells with colicin N at 5–15 µM selectively caused cytotoxicity with no noticeable cell death in human dermal papilla cells [36]. In addition, in protein analysis, lung cancer cells cultured with colicin N (10–15 µM) for 12 h were found to activate extrinsic apoptosis [36]. This was evidenced by the reduction of c-FLIP (Caspase-Like Apoptosis Regulatory Protein) and caspase-8 as well as the altered expression of intrinsic apoptosis signaling proteins Bax (BCL2 Associated X, Apoptosis Regulator) and Mcl-1 (MCL1 Apoptosis Regulator, BCL2 Family Member), i.e., an anti-apoptosis protein overexpressed in NSCLC [36]. Furthermore, 5–15 µM colicin N downregulated the expression of activated AKT kinase (AKT Serine/Threonine Kinase 1) as well as integrins β1 and αV in human lung cancer cells. The authors conclude that colicin N exhibits selective anticancer activity associated with suppression of the integrin-modulated survival molecule. In a further study conducted by Arunmanee et al., it was shown that resurfacing of the receptor-binding domain of colicin N (solvent-exposed aspartic (D) and glutamic (E) acids of the receptor-binding domain substituted by lysine residues (K) as these substitutions, ColN+12) increased the net positive charge of the protein surface. ColN+12 was found to enhance its cytotoxic effect on human lung cancer cells [37]. The toxicity of ColN+12 was cancer selective. Human lung cancer cells H460 and H23 were sensitive to ColN, but human dermal papilla cells were not. ColN+12 also showed more potent toxicity against cancer cells than ColNWT. This confirmed that the polycationic resurfacing method improved the anticancer activity of ColN toward human lung cancer cells. The decrease in the percent cell viability determined was particularly evident in lung cancer cells exposed to 10 µM ColNWT and 5–10 µM ColN+12. As shown by the comparison of the use of the peptides at the same concentrations, the treatment with ColN+12 induced a greater reduction of viable cells than the ColNWT treatment in both H460 and H23 lung cancer cells. The determination of the mode of cell death demonstrated that the culture with either ColNWT (10 µM) or ColN+12 (5–10 µM) induced apoptosis. Interestingly, the authors indicate that, although the bactericidal activity of the ColN mutant was reduced, its cytotoxicity against human lung cancer cells was significantly increased, and selectivity for lung cancer cells was noted [37].
Nisin ZP is a polycyclic antimicrobial peptide produced by the Gram-positive bacterium Lactococcus lactis. Investigation results indicated a concentration-dependent decline in NSCLC cell viability with an IC50 value of 132.4 μM in A549 cells and 137 μM in H1299 cells upon nisin ZP treatment [38]. The results revealed that nisin ZP induced selective toxicity in lung cancer cells, compared to healthy cell lines [38]. Nisin ZP induced apoptosis and cell cycle arrest in the G0/G1 phase in non-small cell lung cancer cells, irrespective of the p53 expression. The inhibition of proliferation was caused via non-membranolytic pathways by mitochondrial membrane depolarization and enhanced generation of reactive oxygen species. Furthermore, nisin ZP was able to reduce the clonal proliferation and migration of tumor cells, indicating its effect in advanced metastatic non-small cell lung cancer [38]. Bacteriocins, which are mostly peptides produced by bacteria, are considered antimicrobial substances; given their ability to disintegrate the cell membrane and destroy cells, they are widely studied as potential anticancer substances. Laterosporulin10 isolated from Brevibacillus sp. strain SKDU10 demonstrated cytotoxic properties against lung cancer H1299 cells at a concentration below 10 μM [39]. It has also been observed that this peptide can induce apoptosis and necrosis [39]. Bacillus amyloliquefaciens BTSS3 produces glycine-rich bacteriocin BaCf3, i.e., a hydrophobic peptide with a characteristic property of AMPs acting on the cell wall [40]. The structure of BaCf3 obtained from TrRosetta had antiparallel β-sheets resembling laterosporulin [40]. In silico studies with an anticancer target have proved that bacteriocin BaCf3 is a potential anticancer drug candidate. Saidumohamed et al. also performed in vitro tests on a lung cancer cell line (A549) and found that the rate of proliferation of cells treated with BaCf3 was higher after 48 h incubation but lower than 50% after 72 h incubation, indicating that it is a slowly acting anticancer compound [40]. In addition, as demonstrated by in silico studies, BaCf3 interacts with GLUT1 (Glucose transporter 1), which is overexpressed in many types of cancer, including lung cancer, and with the kinase domain of the MET receptor (MET Proto-Oncogene, Receptor Tyrosine Kinase), whose overexpression is detected in 25–50% of NSCLC patients. All this indicates that BaCf3 is worth considering in further research as a potential drug against lung cancer.
The azurin protein produced by Pseudomonas aeruginosa has pro-apoptotic properties, and the p28 peptide itself, i.e., a derivative of azurin, located at the C-terminal part of the protein chain, has anticancer properties [41]. As indicated by Huang et al., both azurin and its peptide derivative can integrate functionally with many signaling pathways involved in carcinogenesis, e.g., by stabilizing the p53 protein or regulating the activity of tyrosine kinase signaling pathways [41]. Thus, they indicate that, as potential anticancer drugs, they do not induce resistance as molecularly targeted treatments, which must be constantly refined to target cells that have undergone clonal selection for the occurrence of genetic markers of resistance. It can also be proposed that they can be used as gene-agnostic drugs targeting tyrosine kinase domains present in many genes hyperactivated in cancer. In addition to p28, Garizo et al. indicated CT-p19LC as an inducer of lung cancer cell death [42]. They conducted research on line A549; the concentrations of both peptides from 10 to 100 µM exhibited cytotoxic activity against the cancer cells and a dose–response effect was evident [42]. Furthermore, the treatment with CT-p26 led to an approximately two- to seven-fold higher decrease in cell viability than the treatment with p28 [42].
Bacillus subtilis is a source of natural lipopeptides exerting pro-apoptotic and cell cycle inhibitory effects. Yin et al. proved that fengicin reduced the proliferation of lung cancer cells (95D) and showed that it inhibited tumor growth in in vivo studies [43]. Moreover, this lipopeptide was able to induce ROS production and calcium ion uptake as well as LDH release and loss of mitochondrial membrane potential [43]. The study evidenced that this compound induced apoptosis of the lung cancer cells and cell cycle arrest in the G0/G1 phase by reducing the expression of cyclin D1 and CDK4 (cyclin-dependent kinase 4). The ability to induce apoptosis was linked to the mitochondrial pathway. The researchers observed that fengicin increased the activity of caspase and the expression of the pro-apoptotic Bax protein (BCL2 Associated X, Apoptosis Regulator) and reduced the expression of the anti-apoptotic Bcl-2 protein (BCL2 Apoptosis Regulator) [40]. Bacillus subtilis is known to produce metabolites, e.g., fengicin, iturin, and surfactin, which are effective against lung cancer cells [44]. Shao et al. purified Bacillus subtilis NC16 metabolites and showed that metabolite extracts suppressed proliferation and migration but promoted apoptosis of NSCLC cell lines through the caspase-3/7 signaling pathway [45]. There is no information about the exact components of the purified extract that act in this way. It is necessary to identify and check each fraction of purified metabolites, as a mixture of metabolites comprising bacteriocins/peptides may act synergistically against lung cancer cells. Routhu et al. isolated an exopolymeric biosurfactant composed of lipoheptapeptides from Bacillus atrophaeus AKLSR [46]. The identified cyclic lipopolypeptide (CLP) variants forming one major polymeric lipopeptide were shown to cause lung cancer cell death but were not toxic to normal cells. It induced apoptosis, cell cycle arrest of A549 cells, ROS accumulation, nuclear fragmentation, and cell death [46].
Table 1. Examples of compounds of bacterial origin showing anticancer abilities against lung cancer cells.
Table 1. Examples of compounds of bacterial origin showing anticancer abilities against lung cancer cells.
Anti-NSCLC AgentType of SubstanceSourceAnticancer ActionCitation
Cinerubin Banthracycline antibioticStreptomyces sp. CMAA 1527to be studied[32]
Actinomycin VantibioticStreptomyces sp. CMAA 1653cytotoxicity, induction of apoptosis, and EMT transition blockade[32,33,34]
Enterocin 12abacteriocin Enterococcus faecium 12ainhibition of cancer cell growth[35]
Colicin NbacteriocinEscherichia colicytotoxicity and induction of apoptosis[36,37]
Nisin ZPpeptideLactococcus lactisinhibition of proliferation and induction of apoptosis[38]
Laterosporulin10bacteriocin/peptideBrevibacillus sp. strain SKDU10cytotoxicity, apoptosis induction, and necrotic death induction[39]
BaCf3bacteriocin/peptideBacillus amyloliquefaciensproliferation inhibition[40]
AzurinproteinPseudomonas aeruginosapro-apoptotic properties[41,42]
p28peptideAzurin from Pseudomonas aeruginosapro-apoptotic properties[41]
CT-p26peptideAzurin from Pseudomonas aeruginosacytotoxic activity, decrease in cell viability, pro-apoptotic properties[41]
FengicinlipopeptideBacillus subtilisproliferation and tumor growth inhibition, and induction of apoptosis[44]
Polymeric lipopeptides formed by cyclic lipopolypeptideslipopeptidesBacillus atrophaeus AKLSRinduction of apoptosis, cell cycle arrest, ROS accumulation, nuclear fragmentation, and cell death[46]

4. Secondary Metabolites Synthesized by Endophytes and Marine-Derived Fungi with Lung Cancer Treatment Potential

Fungi constitute a very large group of microorganisms synthesizing secondary metabolites with different anticancer properties, including their activity against lung cancer. Previous model studies on the possibility of the application of fungal metabolites in the treatment of cancer investigated edible mushrooms used in Chinese medicine [47,48]. In this case, the anticancer properties of fungal metabolites were tested using extracts (aqueous, ethanol, and methanol) or chemical compounds obtained from fungal cultures [47,48,49]. Usually, the cytotoxicity and antiproliferative properties of lung cancer cell lines [49] and the level of gene expression responsible for tumor growth were estimated in fungal extracts [48].
Fungi are a source of a wide range of bioactive secondary metabolites, but in terms of practical application, importance is assigned to bioactive metabolites synthesized by non-toxigenic fungi (marine-derived and endophytic). Table 2 shows anticancer activities of secondary metabolites from endophytic and marine-derived fungi used against lung cancer. Therefore, this chapter will discuss compounds synthesized by these fungi and their potential in the treatment of lung cancer. Metabolites synthesized by endophytic and marine-derived fungi vary in terms of chemical structure. They mainly include polyketides, pyrones, chromones, isocoumarins, xanthones, phenalenones, diphenyl ethers, terpenoids, meroterpenoids, sesquiterpenoids, diterpenoids, triterpenoids, steroids, macrolides, lactones, alkaloidsazaphilones, terphenyls, cytochalasans, anthracenones, polyketide-terpene hybrids, polyketide-nonribosomal peptide hybrids, and quinones [48,50,51]. Among non-toxigenic fungi, endophytic fungi, i.e., symbiotic fungi that colonize various tissues of their host, most often plants, deserve special attention. Endophytes are an excellent source of bioactive compounds with anticancer activity, including those against lung cancer [52,53,54,55,56,57,58]. In addition to endophytic fungi, bioactive metabolites with anticancer properties against lung cancer are synthesized by marine-derived fungi [59,60,61,62] (Table 1). Paclitaxel mentioned in the introduction, i.e., the best known and most extensively studied anticancer metabolite characterized in the 1990s, is synthesized by the endophytic fungus Taxomyces andreanae [56]. Currently known under the brand name Taxol®, it is widely used in anticancer treatment [63]. In 1999, the Food and Drug Administration (FDA) approved the use of Taxol® for the treatment of non–small cell lung cancer (NSCLC) [64]. This tubulin-binding agent is used for the treatment of small cell lung cancer (SCLC) as well [65].
One of the main mechanisms of action of anticancer fungal metabolites is the inhibition of the transcription factor, i.e., the NF kappa beta factor (nuclear factor kappa-light-chain-enhancer of activated B cells) [50]. NF-κB is a key player in inflammation, cancer development, and progression. Moreover, NF-κB stimulates cell proliferation, prevents apoptosis, and promotes tumor angiogenesis and metastasis [66].
The anticancer activity of fungal metabolites against lung cancer was usually tested in A549, Calu-3 [60] (adenocarcinoma lung cancer), HL251 (human lung cancer), 95-D (lung cancer cells), NCI-H187 (small cell lung cancer), H522-T1, NCI-H460, NCI-H1650, and NCI-H1975 (non-small cell lung cancer) human cell lines (Table 1). The fungal secondary metabolites applied to these cells exerted different effects, as shown by the wide range of IC50 values (from 0.003 to 100 and even higher) (Table 1). However, from the practical point of view and their potential use in the treatment of lung cancer, the most important agents are secondary metabolites synthesized by fungi for which the IC50 values are in the range of 0.002–3 µM depending on the type of cell line; these values are similar to the IC50 of the currently used anticancer drugs serving as positive controls in experiments, i.e., adriamycin, doxorubicin, epirubicin, and afatinib [56,67,68,69,70,71,72,73,74]. The experiments carried out so far on adenocarcinoma lung cancer cell line A549 have shown the highest cytotoxic activities of Alternarior 9-methyl ether (IC50 = 2.26 µM), Altertoxin II (IC50 = 1.15 µM), Versixantone L (IC50 = 1.60 µM), Asperterphenyllin G (IC50 = 0.40 µM), Prenylcandidusin G (IC50 = 2.80 µM), Prenylterphenyllin H (IC50 = 0.40 µM), Aspergillusone D (IC50 = 0.20 µM), Malformin E (IC50 = 2.42 µM), 21-epi-ophiobolin O (IC50 = 0.60 µM), Ophiobolin O (IC50 = 2.40 µM), 6-Formamide Chetomin (IC50 = 0.027 µM), Hispidulone B (IC50 = 2.71 µM), Sinopestalotiollide D (IC50 = 2.14 µM), 12′-hydroxyroridin E (IC50 = 2.08 µM), and Xanthocillins X (IC50 = 0.38 µM) synthesized by Alternaria sp. LV52, Aspergillus versicolor HDN1009, Aspergillus candidatus LDJ-5, Aspergillus clavatus L, Aspergillus tamari FR02, Aspergillus ustus 094102, Chaetomium sp. M336, Chaetosphaeronema hispidulum, Pestalotiopsis palmarum, Myrothecium roridum E-1069, and Penicillium chrysogenum CCTCC M 202001, respectively [69,75,76,77,78,79,80,81,82,83]. A previous study reported higher anticancer activity against lung cancer cells NCI-H460 exhibited by the following fungal secondary metabolites: (−)-(10E,15S)-4,6-dichloro-10(11)-dehydrocurvularin (IC50 = 1.45 µM), Chaunolidone A (IC50 = 0.09 µM), Beauvericin (IC50 = 1.41 µM), Bikaverin (IC50 = 0.43 µM), 3-epi-Waol A (IC50 = 1.00 µM), Epiroridin E (IC50 = 0.03 µM), Mytoxin B (IC50 = 0.07 µM), and Epiroridine acid (IC50 = 0.36 µM) synthesized by Alternaria sp. AST0039, Chaunopycnis sp. CMB-MF028, Fusarium oxysporum EPH2RAA, Fusarium oxysporum CECIS, Libertella blepharis F2644, and Myrthecim roridum A553 [61,79,84,85,86]. In the case of non-small cell lung cancer cell line NCI-H1975, secondary fungal metabolites Rhytidenone H (IC50 = 0.25 µM) and Rhytidenone F (IC50 = 1.17 µM) produced by Rhytidhysteron rufulum AS21B showed the highest anticancer activity [74] (Table 1). The most effective secondary fungal metabolites, i.e., (−)-(10E,15S)-4,6-dichloro-10(11)-dehydrocurvularin (Alternaria sp. AST0039), Chaunolidone A (Chaunopycnis sp. CMB-MF028), Beauvericin (Fusarium oxysporum EPH2RAA), Bikaverin (Fusarium oxysporum CECIS), 3-epi-Waol A (Libertella blepharis F2644), Epiroridin E, Mytoxin B, Epiroridine acid (Myrthecim roridum A553), Mycoleptodiscin B (Mycoleptodiscus spp. F0194), Brocazines F (Penicillium brocae MA-231), and Cytochalasin C, D, Q (Xylaria spp. NC1214), against non-small cell lung cancer HCI-H460 were characterized by the following IC50 values: 1.45, 0.09, 1.41, 0.43, 1.00, 0.003, 0.007, 0.36, 0.66, 0.89, 0.22, 1.06, and 1.51 µM, respectively [61,79,84,85,86,87,88,89]. Chinworrungsee et al., 2008 [90,91] indicated high cytotoxic activity of Brefeldin A, 8-deoxy-trichothecin, 7-hydroxytrichodermol (IC50 = 0.11; 1.48 and 1.73 µM) from strain KLAR 5 (Hypocreales) and Xanthoquinodin B9 (IC50 = 0.98 µM) from Chaetomium globosum 7s-1 against small cell lung cancer HCl-H187. Moreover, the cytotoxicity of Mycoleptodiscin B synthesized by Mycoleptodiscus spp. F0194 against non-small cell lung cancer H522-T1 was characterized by IC50 = 0.63 µM [87].
Table 2. Anticancer activities of secondary metabolites from endophytic and marine-derived fungi against lung cancer.
Table 2. Anticancer activities of secondary metabolites from endophytic and marine-derived fungi against lung cancer.
Fungal StrainsCompoundsIC50 Values
(µM)
Lung Cancer Cell LinesReferences
Alternaria sp. LV52Alternarior 9-methyl ether2.26A549[75]
Altertoxin II1.15
Aspergillus versicolor HDN1009Versixantone G17.80[62]
Versixantone H19.20
Versixantone L1.60
Versixantone M11.70
Aspergillus candidatus LDJ-5Asperterphenyllin G0.40[92]
Prenylcandidusin E19.10[93]
Prenylcandidusin G2.80
Prenylterphenyllin F10.20
Prenylterphenyllin G16.30
Prenylterphenyllin H0.40
Prenylterphenyllin I14.80
Prenylterphenyllin J7.60
Aspergillus clavatus LAspergillusone C41.90[81]
Aspergillusone D0.20
Aspergillus micronesiensis
MH938722
Cyschalasin B 16.79[94]
Aspergillus oryzae
KM999948
Oryzaein B 4.20[95]
Oryzaein A 6.50
Oryzaein C 6.80
Aspergillus tamarii
FR02
Malformin E 2.42[76]
Aspergillus ustus 09410221-epi-ophiobolin O0.60[77]
Ophiobolin O2.40
21-deoxyophiobolin K15.10
Ophiobolin Q33.80
Ophiobolin X;
21,21-O-dihydro6-epi-ophiobolin G
>50
Aspergillus sp. SCSIO41407Flavoglaucin22.20[96]
Aspergillus fumigates
2011041507–5
Alkaloids fumiquinazoline J 26.90[97]
Fumiquinazoline C 33.40
Trypacidin 31.00
Aspergillus versicolor F210Proversilin C15.00[98]
Proversilin E28.40
Proversilin A>40
Proversilin B>40
Proversilin D>40
Cordyceps taiiDeacetylcytochalasin C13.62[99]
Zygosporin D16.72
Cytochalasins 217.13
Cytochalasins 319.92
Cytochalasins 132.28
Chaetomium
globosum
kz-19
Penochalasin J14.90[70]
Phychaetoglobin C22.30
Phychaetoglobin D13.70
Chaetoglobosin C7.60
Chaetoglobosin E12.30
Chaetoglobosin G7.30
Chaetoglobosin V11.00
Chaetoglobosin J13.40
Chaetomium sp. M3366-Formamide Chetomin0.027[78]
Chaetosphaeronema
hispidulum
Hispidulone B2.71[83]
Emericella sp. TJ29Emeridone D11.33[100]
Pestalotiopsis palmarumSinopestalotiollide D2.14[82]
Sinopestalotiollide A31.29
Sinopestalotiollide C 36.13
Sinopestalotiollide B44.89
2H-pyran-2-one47.82
Phoma sp. SYSU-SK-7 Colletotric A37.73[101]
Colletotric A20.75
Hypocrea lixii R-18Cajanol20.50–32.80[102]
Eupenicillium sp. HJ002Penicilindole A5.50[72]
Penicilindole B18.60
Fusarium sp. 2ST2Fusarisetins E 8.70 [103]
Fusarisetins F 4.30
Fusarium oxysporum
GU250648
Beauvericin 10.40 [104]
Lasiodiplodia theobromae
ZJ-HQ1
Chloropreussomerin A 8.50 [68]
Chloropreussomerin B 8.90
Preussomerin A 40.20
Preussomerin D 6.60
Preussomerin F 7.70
Preussomerin G 6.20
Preussomerin H 9.40
Preussomerin K 5.40
Preussomerin M 36.10
Myrothecium roridum
E-1069
12′-hydroxyroridin E 2.08 [105]
Myrotoxin A 3.56
Mytoxin C 33.00
2′,3′-epoxymyrothecine A 36.45
Vertisporin 47.00
14′-hydroxymytoxin B 49.00
13′,14′-hydroxymytoxin B 53.00
Roridin E 55.00
Myrothecine A 95.00
Penicillium chrysogenum V11Penochalasin I16.13[67]
Penochalasin J35.93
Penochalasin K8.73[106]
Chaetoglobosin A6.56[67]
Chaetoglobosin C17.82
Chaetoglobosin E36.63
Chaetoglobosin F27.72
Penicillium chrysogenum
AD-1540
Chryxanthone A41.70[107]
Chryxanthone B20.40
Penicillium chrysogenum CCTCC M 2020019Xanthocillins X0.38[80]
Xanthocillins Y15.04
2-aminophenoxazin-3-one25.60
Chrysomamide;
N-[2-trans-(4-hydroxyphenyl)ethenyl]formamide;
42.87
N-acetylquestiomycin A52.61
Penicillium chrysogenumPenichryfurans A>100[108]
Penichryfurans B87.90
Penicillium polonicum TY12Polonidine A15.00 *[109]
Penicillium sp. sh18Isopenicin A37.06[110]
Isopenicin B>40
Isopenicin C>40
Preussia similisPreussilide C22.90[111]
Preussilide E 41.20
Preussilide D47.90
Preussilide A 60.30
Preussilide B70.30
Rhizopycnis vagum Nitaf22Rhizopycnin C25.50[112]
Trichoderma citrinovirideBislongiquinolide11.00[113]
Dihydrotrichodimerol33.00
Trichoderma reesei
HN-2016-018
24-hydroxy-trichodimerol5.10[114]
Mucor irregularis
QEN-189
Penitrem A8.40[115]
Penitrem C8.00
Penitrem F8.20
Rhizovarin A11.50
Rhizovarin B6.30
Rhizovarin E9.20
3b-hydroxy-4b-desoxypaxilline4.60
Dichotomomyces sp. L-8 (3R,6R)-bassiatin 14.54Calu-3[60]
Pestalotiopsis m. EF01Paclitaxel (=taxol)0.50HL251[116]
Alternaria sp. A744Alterperylenol5.47H460[117]
Altertoxin II9.67
6-epi-stemphytriol43.31
Isobenzofuranone A;
Indandione B;
Isosclerone;
2,4,8-trihydroxy-1-tetralone;
3,4-dihydro-3,4,8-trihydroxy-1[2H]-naphthalenone;
6-hydroxyisosclerone;
cis-4-hydroxyscytalone;
alternariol-4-methyl ether;
Dihydroalterperylenol;
alterperylenol
>100
Alternaria sp. AST0039(−)-(10E,15S)-4,6-dichloro-10(11)-dehydrocurvularin1.45[84]
(−)-(10E,15S)-6-chloro-10(11)-dehydrocurvularin3.57
Aspergillus sp. HN15-15DAspergisocoumrin A21.53[71]
Aspergillus oryzeaPaclitaxel (=taxol)50.00 *[118]
Aspergillus fumigates
2011041507–5
Alkaloids pyripyropene A 38.30[97]
Trypacidin 33.80
Bipolaris sorokiniana
A606
Cochlioquinone H15.40[119]
Cochlioquinone G26.90
Isocochlioquinone E31.10
Isocochlioquinone D42.60
Cerrena sp. A593Cerrenin D29.67[120]
Chaunopycnis sp.
CMB-MF028
Chaunolidone A0.09[61]
Chaetomium globosumGlobosumone A6.50[121]
Globosumone B24.80
Cytospora rhizophorae
A761
Cytorhizin B32.80[122]
Cytorhizin C54.70
Didymella sp.
CYSK-4
Ascomylactam A4.40[123]
Ascomylactam B13.00
Ascomylactam C4.40
Phomapyrrolidone C12.00
Pyrrolidone A28.00
Fusarium oxysporum EPH2RAABeauvericin1.41[85]
Fusarium oxysporum
CECIS
Bikaverin0.43
Libertella blepharis
F2644
3-epi-Waol A 1.00 [86]
Myrthecim roridum A553Epiroridin E 0.003 [79]
Mytoxin B 0.007
Epiroridine acid 0.36
Mycoleptodiscus spp. F0194Mycoleptodiscin B0.66[87]
Penicillium brocae
MA-231
Brocazines F0.89[88]
Pestalotiopsis flavidula2′-aminodechlorogeodoxin16.47[124]
2′-aminodechloromaldoxin18.63
Phyllosticta spinarumTauranin4.30[125]
strain PM0651480Ergoflavin4.00[126]
Xylaria spp. NC1214Cytochalasin C0.22[89]
Cytochalasin D1.06
Cytochalasin Q1.51
Cladosporium sp. OUCMDZ-3027-O-αD-ribosyl-5-hydroxy-2-propylchromone10.00HCI-H1975[73]
Aspergillus versicolor HDN1009Versixantone G9.80[62]
Versixantone H5.30
Versixantone M3.50
Versixantone N8.80[69]
Versixantone O8.50
Rhytidhysteron rufulum AS21BRhytidenone H0.25[74]
Rhytidenone F1.17
Rhytidenone G7.30
Rhytidenone E 10.24
Deoxypreussomerin B; Palmarumycin CP17;
1-oxo-1,4-dihydronapthalene-4-spiro-20-naptho[400-hydroxy-100,800-de] [10,30]-dioxin; Preussomerin EG4;
CJ-12,371;
4-O-methyl-CJ-12,371; Palmarumycin C5; Rhytidone A
>100.00
Pleosporales sp. Sigrf05Pleospyrone E6.26HCI-H1650[127]
Pleospyrone A15.10
Pleospyrone D29.60
Rhizopycnis v. Nitaf22TMC-2643.20[112]
Chaetomium globosum 7s-1Xanthoquinodin B90.98HCI-H187[91]
Eutypella sp. BCC 13199ent-4(15)-eudesmen-11-ol-1-one11.00[128]
Eutypellin A12.00
strain KLAR 5
Hypocreales
Brefeldin A 0.11[90]
8-deoxy-trichothecin1.48
7-hydroxytrichodermol1.73
Trichothecolone11.31
7-hydroxyscirpene27.76
Phomopsis spp. BCC 9789Oblongolide Z32.00 [129]
Xylaria spp. BCC 21097Eremophilanolide 17.20[130]
Eremophilanolide 23.80
Eremophilanolide 35.80
Xylaria cf. c. PK108Cytochalasin D5.95[131]
Ergosterol peroxide5.81
Mycoleptodiscus spp. F0194Mycoleptodiscin B0.63H522-T1[87]
Cordyceps taiiDeacetylcytochalasin C3.6795-D[99]
Zygosporin D4.04
Cytochalasins 320.69
Cytochalasins 123.67
Cytochalasins 226.03
* µg/mL.

5. Conclusions

The abundance of natural compounds in nature prompts research into their use in the treatment of cancers, including lung cancer. As natural compounds, peptides have great potential in this approach, and their effects on induction of apoptosis or inhibition of proliferation of cancer cells have been observed by many researchers. Peptides appear to be highly promising compounds for research into their use as anticancer drugs due to their structure, greater ease of production compared to full-length proteins, chemical properties leading to destruction of cancer cell membranes and ultimately entire cells, and the ability to penetrate the blood–brain barrier. Their abundance and ability to be modified by adding functional groups make them broadly applicable in oncological approaches, but more comprehensive research of individual peptides is required to determine their efficacy and possible toxic effects on healthy cells. Natural peptides may be adopted as targeted aptamer therapies, targeting specific cancer cell receptors. These issues leave a wide field for research, which may lead to the development of new targeted therapies based on naturally derived peptides. Moreover, the huge number of compounds produced by fungi generates the need to conduct studies in this field. As shown in this review, their IC50 values vary, and they may act in a dose-dependent manner. The papers cited here present in vitro investigations of model lung cancer cell lines. Future in vivo studies will be of great interest, as they may provide information about the toxicity of these compounds and their potential for implementation in clinical practice. This should be preceded by clinical trials determining the possibility of using these compounds alone or as supportive therapies for therapeutics that are currently registered for use in clinical practice.

Author Contributions

Conceptualization, writing—original draft preparation, writing—review and editing, visualization, supervision and project administration A.G., A.J. and K.R.-T., supervision P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. Cancer (IARC). T.I.A. for R. on Global Cancer Observatory. Available online: https://gco.iarc.fr/ (accessed on 29 December 2022).
  2. Croteau, R.; Ketchum, R.E.B.; Long, R.M.; Kaspera, R.; Wildung, M.R. Taxol Biosynthesis and Molecular Genetics. Phytochem. Rev. Proc. Phytochem. Soc. Eur. 2006, 5, 75–97. [Google Scholar] [CrossRef] [PubMed]
  3. Alqathama, A. BRAF in Malignant Melanoma Progression and Metastasis: Potentials and Challenges. Am. J. Cancer Res. 2020, 10, 1103–1114. [Google Scholar] [PubMed]
  4. Harrison, P.T.; Vyse, S.; Huang, P.H. Rare Epidermal Growth Factor Receptor (EGFR) Mutations in Non-Small Cell Lung Cancer. Semin. Cancer Biol. 2020, 61, 167–179. [Google Scholar] [CrossRef]
  5. Gendarme, S.; Bylicki, O.; Chouaid, C.; Guisier, F. ROS-1 Fusions in Non-Small-Cell Lung Cancer: Evidence to Date. Curr. Oncol. 2022, 29, 641–658. [Google Scholar] [CrossRef] [PubMed]
  6. Grenda, A.; Jarosz, B.; Krawczyk, P.; Kucharczyk, T.; Wojas-Krawczyk, K.; Reszka, K.; Krukowska, K.; Nicoś, M.; Pankowski, J.; Bryl, M.; et al. Discrepancies between ALK Protein Disruption and Occurrence of ALK Gene Rearrangement in Polish NSCLC Patients. J. Thorac. Dis. 2018, 10, 4994–5009. [Google Scholar] [CrossRef]
  7. Seligson, N.D.; Knepper, T.C.; Ragg, S.; Walko, C.M. Developing Drugs for Tissue-Agnostic Indications: A Paradigm Shift in Leveraging Cancer Biology for Precision Medicine. Clin. Pharmacol. Ther. 2021, 109, 334–342. [Google Scholar] [CrossRef]
  8. Kennedy, L.B.; Salama, A.K.S. A Review of Cancer Immunotherapy Toxicity. CA Cancer J. Clin. 2020, 70, 86–104. [Google Scholar] [CrossRef]
  9. Pan, C.; Liu, H.; Robins, E.; Song, W.; Liu, D.; Li, Z.; Zheng, L. Next-Generation Immuno-Oncology Agents: Current Momentum Shifts in Cancer Immunotherapy. J. Hematol. Oncol. 2020, 13, 29. [Google Scholar] [CrossRef]
  10. Frisone, D.; Friedlaender, A.; Addeo, A.; Tsantoulis, P. The Landscape of Immunotherapy Resistance in NSCLC. Front. Oncol. 2022, 12, 817548. [Google Scholar] [CrossRef]
  11. Haslam, A.; Prasad, V. Estimation of the Percentage of US Patients with Cancer Who Are Eligible for and Respond to Checkpoint Inhibitor Immunotherapy Drugs. JAMA Netw. Open 2019, 2, e192535. [Google Scholar] [CrossRef]
  12. Hashem, S.; Ali, T.A.; Akhtar, S.; Nisar, S.; Sageena, G.; Ali, S.; Al-Mannai, S.; Therachiyil, L.; Mir, R.; Elfaki, I.; et al. Targeting Cancer Signaling Pathways by Natural Products: Exploring Promising Anti-Cancer Agents. Biomed. Pharmacother. 2022, 150, 113054. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, Y.; Li, N.; Wang, T.-M.; Di, L. Natural Products with Activity against Lung Cancer: A Review Focusing on the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 10827. [Google Scholar] [CrossRef] [PubMed]
  14. Chanvorachote, P.; Chamni, S.; Ninsontia, C.; Phiboonchaiyanan, P.P. Potential Anti-Metastasis Natural Compounds for Lung Cancer. Anticancer Res. 2016, 36, 5707–5717. [Google Scholar] [CrossRef] [PubMed]
  15. Xie, M.; Liu, D.; Yang, Y. Anti-Cancer Peptides: Classification, Mechanism of Action, Reconstruction and Modification. Open Biol. 2020, 10, 200004. [Google Scholar] [CrossRef]
  16. Chiangjong, W.; Chutipongtanate, S.; Hongeng, S. Anticancer Peptide: Physicochemical Property, Functional Aspect and Trend in Clinical Application (Review). Int. J. Oncol. 2020, 57, 678–696. [Google Scholar] [CrossRef] [PubMed]
  17. Karami Fath, M.; Babakhaniyan, K.; Zokaei, M.; Yaghoubian, A.; Akbari, S.; Khorsandi, M.; Soofi, A.; Nabi-Afjadi, M.; Zalpoor, H.; Jalalifar, F.; et al. Anti-Cancer Peptide-Based Therapeutic Strategies in Solid Tumors. Cell. Mol. Biol. Lett. 2022, 27, 33. [Google Scholar] [CrossRef]
  18. Droin, N.; Hendra, J.-B.; Ducoroy, P.; Solary, E. Human Defensins as Cancer Biomarkers and Antitumour Molecules. J. Proteom. 2009, 72, 918–927. [Google Scholar] [CrossRef]
  19. Harris, F.; Dennison, S.R.; Singh, J.; Phoenix, D.A. On the Selectivity and Efficacy of Defense Peptides with Respect to Cancer Cells. Med. Res. Rev. 2013, 33, 190–234. [Google Scholar] [CrossRef]
  20. Li, G.; Huang, Y.; Feng, Q.; Chen, Y. Tryptophan as a Probe to Study the Anticancer Mechanism of Action and Specificity of α-Helical Anticancer Peptides. Molecules 2014, 19, 12224–12241. [Google Scholar] [CrossRef]
  21. Schaduangrat, N.; Nantasenamat, C.; Prachayasittikul, V.; Shoombuatong, W. ACPred: A Computational Tool for the Prediction and Analysis of Anticancer Peptides. Molecules 2019, 24, 1973. [Google Scholar] [CrossRef]
  22. Ray, T.; Kar, D.; Pal, A.; Mukherjee, S.; Das, C.; Pal, A. Molecular Targeting of Breast and Colon Cancer Cells by PAR1 Mediated Apoptosis through a Novel Pro-Apoptotic Peptide. Apoptosis Int. J. Program. Cell Death 2018, 23, 679–694. [Google Scholar] [CrossRef] [PubMed]
  23. Böhmová, E.; Machová, D.; Pechar, M.; Pola, R.; Venclíková, K.; Janoušková, O.; Etrych, T. Cell-Penetrating Peptides: A Useful Tool for the Delivery of Various Cargoes into Cells. Physiol. Res. 2018, 67, S267–S279. [Google Scholar] [CrossRef] [PubMed]
  24. Conlon, J.M.; Mechkarska, M.; Prajeep, M.; Arafat, K.; Zaric, M.; Lukic, M.L.; Attoub, S. Transformation of the Naturally Occurring Frog Skin Peptide, Alyteserin-2a into a Potent, Non-Toxic Anti-Cancer Agent. Amino Acids 2013, 44, 715–723. [Google Scholar] [CrossRef]
  25. Fan, X.; Bai, L.; Mao, X.; Zhang, X. Novel Peptides with Anti-Proliferation Activity from the Porphyra Haitanesis Hydrolysate. Process Biochem. 2017, 60, 98–107. [Google Scholar] [CrossRef]
  26. Wang, Z.; Zhang, X. Inhibitory Effects of Small Molecular Peptides from Spirulina (Arthrospira) Platensis on Cancer Cell Growth. Food Funct. 2016, 7, 781–788. [Google Scholar] [CrossRef]
  27. Patil, S.M.; Kunda, N.K. Anticancer Activity of D-LAK-120A, an Antimicrobial Peptide, in Non-Small Cell Lung Cancer (NSCLC). Biochimie 2022, 201, 7–17. [Google Scholar] [CrossRef] [PubMed]
  28. McConnell, E.J.; Devapatla, B.; Yaddanapudi, K.; Davis, K.R. The Soybean-Derived Peptide Lunasin Inhibits Non-Small Cell Lung Cancer Cell Proliferation by Suppressing Phosphorylation of the Retinoblastoma Protein. Oncotarget 2014, 6, 4649–4662. [Google Scholar] [CrossRef] [PubMed]
  29. Lin, X.; Dong, L.; Yan, Q.; Dong, Y.; Wang, L.; Wang, F. Preparation and Characterization of an Anticancer Peptide from Oriental Tonic Food Enteromorpha Prolifera. Foods 2022, 11, 3507. [Google Scholar] [CrossRef]
  30. Alhakamy, N.A.; Okbazghi, S.Z.; Alfaleh, M.A.; Abdulaal, W.H.; Bakhaidar, R.B.; Alselami, M.O.; Zahrani, M.A.; Alqarni, H.M.; Alghaith, A.F.; Alshehri, S.; et al. Wasp Venom Peptide Improves the Proapoptotic Activity of Alendronate Sodium in A549 Lung Cancer Cells. PLoS ONE 2022, 17, e0264093. [Google Scholar] [CrossRef]
  31. Khine, H.E.E.; Ecoy, G.A.U.; Roytrakul, S.; Phaonakrop, N.; Pornputtapong, N.; Prompetchara, E.; Chanvorachote, P.; Chaotham, C. Chemosensitizing Activity of Peptide from Lentinus squarrosulus (Mont.) on Cisplatin-Induced Apoptosis in Human Lung Cancer Cells. Sci. Rep. 2021, 11, 4060. [Google Scholar] [CrossRef]
  32. Silva, L.J.; Crevelin, E.J.; Souza, D.T.; Lacerda-Júnior, G.V.; de Oliveira, V.M.; Ruiz, A.L.T.G.; Rosa, L.H.; Moraes, L.A.B.; Melo, I.S. Actinobacteria from Antarctica as a Source for Anticancer Discovery. Sci. Rep. 2020, 10, 13870. [Google Scholar] [CrossRef]
  33. Lin, S.; Jia, F.; Zhang, C.; Liu, F.; Ma, J.; Han, Z.; Xie, W.; Li, X. Actinomycin V Suppresses Human Non-Small-Cell Lung Carcinoma A549 Cells by Inducing G2/M Phase Arrest and Apoptosis via the P53-Dependent Pathway. Mar. Drugs 2019, 17, 572. [Google Scholar] [CrossRef] [PubMed]
  34. Lin, S.; Zhang, C.; Liu, F.; Ma, J.; Jia, F.; Han, Z.; Xie, W.; Li, X. Actinomycin V Inhibits Migration and Invasion via Suppressing Snail/Slug-Mediated Epithelial-Mesenchymal Transition Progression in Human Breast Cancer MDA-MB-231 Cells In Vitro. Mar. Drugs 2019, 17, 305. [Google Scholar] [CrossRef] [PubMed]
  35. Sharma, P.; Kaur, S.; Chadha, B.S.; Kaur, R.; Kaur, M.; Kaur, S. Anticancer and Antimicrobial Potential of Enterocin 12a from Enterococcus Faecium. BMC Microbiol. 2021, 21, 39. [Google Scholar] [CrossRef] [PubMed]
  36. Arunmanee, W.; Ecoy, G.A.U.; Khine, H.E.E.; Duangkaew, M.; Prompetchara, E.; Chanvorachote, P.; Chaotham, C. Colicin N Mediates Apoptosis and Suppresses Integrin-Modulated Survival in Human Lung Cancer Cells. Molecules 2020, 25, 816. [Google Scholar] [CrossRef]
  37. Arunmanee, W.; Duangkaew, M.; Taweecheep, P.; Aphicho, K.; Lerdvorasap, P.; Pitchayakorn, J.; Intasuk, C.; Jiraratmetacon, R.; Syamsidi, A.; Chanvorachote, P.; et al. Resurfacing Receptor Binding Domain of Colicin N to Enhance Its Cytotoxic Effect on Human Lung Cancer Cells. Comput. Struct. Biotechnol. J. 2021, 19, 5225–5234. [Google Scholar] [CrossRef]
  38. Patil, S.M.; Kunda, N.K. Nisin ZP, an Antimicrobial Peptide, Induces Cell Death and Inhibits Non-Small Cell Lung Cancer (NSCLC) Progression in Vitro in 2D and 3D Cell Culture. Pharm. Res. 2022, 39, 2859–2870. [Google Scholar] [CrossRef]
  39. Baindara, P.; Gautam, A.; Raghava, G.P.S.; Korpole, S. Anticancer Properties of a Defensin like Class IId Bacteriocin Laterosporulin10. Sci. Rep. 2017, 7, 46541. [Google Scholar] [CrossRef]
  40. Saidumohamed, B.E.; Johny, T.K.; Raveendran, A.T.; Sheela, U.B.; Sreeranganathan, M.; Sasidharan, R.S.; Bhat, S.G. 3D Structure Elucidation and Appraisal of Mode of Action of a Bacteriocin BaCf3 with Anticancer Potential Produced by Marine Bacillus Amyloliquefaciens BTSS3. ReGEN Open 2022, 2, 45–56. [Google Scholar] [CrossRef]
  41. Huang, F.; Shu, Q.; Qin, Z.; Tian, J.; Su, Z.; Huang, Y.; Gao, M. Anticancer Actions of Azurin and Its Derived Peptide P28. Protein J. 2020, 39, 182–189. [Google Scholar] [CrossRef]
  42. Garizo, A.R.; Coelho, L.F.; Pinto, S.; Dias, T.P.; Fernandes, F.; Bernardes, N.; Fialho, A.M. The Azurin-Derived Peptide CT-P19LC Exhibits Membrane-Active Properties and Induces Cancer Cell Death. Biomedicines 2021, 9, 1194. [Google Scholar] [CrossRef] [PubMed]
  43. Yin, H.; Guo, C.; Wang, Y.; Liu, D.; Lv, Y.; Lv, F.; Lu, Z. Fengycin Inhibits the Growth of the Human Lung Cancer Cell Line 95D through Reactive Oxygen Species Production and Mitochondria-Dependent Apoptosis. Anticancer. Drugs 2013, 24, 587–598. [Google Scholar] [CrossRef]
  44. Dan, A.K.; Manna, A.; Ghosh, S.; Sikdar, S.; Sahu, R.; Parhi, P.K.; Parida, S. Molecular Mechanisms of the Lipopeptides from Bacillus Subtilis in the Apoptosis of Cancer Cells—A Review on Its Current Status in Different Cancer Cell Lines. Adv. Cancer Biol. Metastasis 2021, 3, 100019. [Google Scholar] [CrossRef]
  45. Shao, Y.; Wang, X.; Qiu, X.; Niu, L.; Ma, Z. Isolation and Purification of a New Bacillus Subtilis Strain from Deer Dung with Anti-Microbial and Anti-Cancer Activities. Curr. Med. Sci. 2021, 41, 832–840. [Google Scholar] [CrossRef] [PubMed]
  46. Routhu, S.R.; Chary, R.N.; Shaik, A.B.; Prabhakar, S.; Kumar, C.G.; Kamal, A. Induction of Apoptosis in Lung Carcinoma Cells by Antiproliferative Cyclic Lipopeptides from Marine Algicolous Isolate Bacillus Atrophaeus Strain AKLSR1. Process Biochem. 2019, 79, 142–154. [Google Scholar] [CrossRef]
  47. Song, F.; Liu, Y.; Kong, X.; Chang, W.; Song, G. MINI-REVIEW Progress on Understanding the Anticancer Mechanisms of Medicinal Mushroom: Inonotus Obliquus. Asian Pac. J. Cancer Prev. 2013, 14, 1571–1578. [Google Scholar] [CrossRef] [PubMed]
  48. Szychowski, K.A.; Rybczyńska-Tkaczyk, K.; Tobiasz, J.; Yelnytska-Stawasz, V.; Pomianek, T.; Gmiński, J. Biological and Anticancer Properties of Inonotus Obliquus Extracts. Process Biochem. 2018, 73, 180–187. [Google Scholar] [CrossRef]
  49. Sairi, A.M.M.; Ismail, S.I.; Sukor, A.; Rashid, N.M.N.; Saad, N.; Jamian, S.; Abdullah, S. Cytotoxicity and Anticancer Activity of Donkioporiella mellea on MRC5 (Normal Human Lung) and A549 (Human Lung Carcinoma) Cells Lines. Evid.-Based Complement. Altern. Med. 2020, 2020, 7415672. [Google Scholar] [CrossRef] [PubMed]
  50. Evidente, A.; Kornienko, A.; Cimmino, A.; Andolfi, A.; Lefranc, F.; Methieu, V.; Kiss, R. Fungal Metabolites with Anticancer Activity. Nat. Prod. Rep. 2014, 31, 617–627. [Google Scholar] [CrossRef]
  51. Yuan, S.; Gopal, J.V.; Ren, S.; Chen, L.; Liu, L.; Gao, Z. Anticancer Fungal Natural Products: Mechanisms of Action and Biosynthesis. Eur. J. Med. Chem. 2020, 202, 112502. [Google Scholar] [CrossRef]
  52. Kousar, R.; Naeem, M.; Jamaludin, M.I.; Arshad, A.; Shamsuri, A.N.; Ansari, N.; Akhtar, S.; Hazafa, A.; Uddin, J.; Khan, A.; et al. Exploring the Anticancer Activities of Novel Bioactive Compounds Derived from Endophytic Fungi: Mechanisms of Action, Current Challenges and Future Perspectives. Am. J. Cancer Res. 2022, 12, 2897–2919. [Google Scholar] [PubMed]
  53. Conrado, R.; Gomes, T.C.; Roque, G.S.C.; De Souza, A.O. Overview of Bioactive Fungal Secondary Metabolites: Cytotoxic and Antimicrobial Compounds. Antibiotics 2022, 11, 1604. [Google Scholar] [CrossRef] [PubMed]
  54. Luo, Y.; Luo, X.; Zhang, T.; Li, S.; Liu, S.; Ma, Y.; Wang, Z.; Jin, X.; Liu, J.; Wang, X. Anti-Tumor Secondary Metabolites Originating from Fungi in the South China Sea’s Mangrove Ecosystem. Bioengineering 2022, 9, 776. [Google Scholar] [CrossRef]
  55. Shevkar, C.; Pradhan, P.; Armarkar, A.; Pandey, K.; Kalia, K.; Paranagama, P.; Kate, A.S. Exploration of Potent Cytotoxic Molecules from Fungi in Recent Past to Discover Plausible Anticancer Scaffolds. Chem. Biodivers. 2022, 19, e202100976. [Google Scholar] [CrossRef]
  56. Prajapati, J.; Goswami, D.; Rawal, R.M. Endophytic Fungi: A Treasure Trove of Novel Anticancer Compounds. Curr. Res. Pharmacol. Drug Discov. 2021, 2, 100050. [Google Scholar] [CrossRef] [PubMed]
  57. El-Hawary, S.S.; Moawad, A.S.; Bahr, H.S.; Abdelmohsen, U.R.; Mohammed, R. Natural Product Diversity from the Endophytic Fungi of the GenusAspergillus. RSC Adv. 2020, 10, 22058–22079. [Google Scholar] [CrossRef]
  58. Hridoy, M.; Gorapi, M.Z.H.; Noor, S.; Chowdhury, N.S.; Rahman, M.M.; Muscari, I.; Masia, F.; Adorisio, S.; Delfino, D.V.; Mazid, M.A. Putative Anticancer Compounds from Plant-Derived Endophytic Fungi: A Review. Molecules 2022, 27, 296. [Google Scholar] [CrossRef]
  59. Noman, E.; Al-Shaibani, M.M.; Bakhrebah, M.A.; Almoheer, R.; Al-Sahari, M.; Al-Gheethi, A.; Radin Mohamed, R.M.S.; Almulaiky, Y.Q.; Abdulaal, W.H. Potential of Anti-Cancer Activity of Secondary Metabolic Products from Marine Fungi. J. Fungi 2021, 7, 436. [Google Scholar] [CrossRef]
  60. Huang, L.H.; Chen, Y.X.; Yu, J.C.; Yuan, J.; Li, H.J.; Ma, W.Z.; Watanapokasin, R.; Hu, K.C.; Niaz, S.I.; Yang, D.P.; et al. Secondary Metabolites from the Marine-Derived Fungus Dichotomomyces sp. L-8 and Their Cytotoxic Activity. Molecules 2017, 22, 444. [Google Scholar] [CrossRef]
  61. Shang, Z.; Li, L.; Espósito, B.P.; Salim, A.A.; Khalil, Z.G.; Quezada, M.; Bernhardt, P.V.; Capon, R.J. New PKS-NRPS Tetramic Acids and Pyridinone from an Australian Marine-Derived Fungus, Chaunopycnis sp. Org. Biomol. Chem. 2015, 13, 7795–7802. [Google Scholar] [CrossRef]
  62. Wu, G.; Qi, X.; Mo, X.; Yu, G.; Wang, Q.; Zhu, T.; Gu, Q.; Liu, M.; Li, J.; Li, D. Structure-Based Discovery of Cytotoxic Dimeric Tetrahydroxanthones as Potential Topoisomerase I Inhibitors from a Marine-Derived Fungus. Eur. J. Med. Chem. 2018, 148, 268–278. [Google Scholar] [CrossRef] [PubMed]
  63. Gallego-Jara, J.; Lozano-Terol, G.; Sola-Martínez, R.A.; Cánovas-Díaz, M.; de Diego Puente, T. A Compressive Review about Taxol®: History and Future Challenges. Molecules 2020, 25, 5986. [Google Scholar] [CrossRef] [PubMed]
  64. Weaver, B.A. How Taxol/Paclitaxel Kills Cancer Cells. Mol. Biol. Cell 2014, 25, 2677–2681. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, S.H.; Kim, M.J.; Kim, Y.J.; Chang, H.; Kim, J.W.; Lee, J.O.; Lee, K.W.; Kim, J.H.; Bang, S.M.; Lee, J.S. Paclitaxel as Third-Line Chemotherapy for Small Cell Lung Cancer Failing Both Etoposide-A Nd Camptothecin-Based Chemotherapy. Medicine 2017, 96, e8176. [Google Scholar] [CrossRef]
  66. Kaltschmidt, B.; Greiner, J.F.W.; Kaltschmidt, C. Subunit-Specific Role of NF-κ B in Cancer. Biomedicines 2018, 6, 44. [Google Scholar] [CrossRef]
  67. Huang, S.; Chen, H.; Li, W.; Zhu, X.; Ding, W.; Li, C. Bioactive Chaetoglobosins from the Mangrove Endophytic Fungus Penicillium Chrysogenum. Mar. Drugs 2016, 14, 172. [Google Scholar] [CrossRef]
  68. Chen, S.; Chen, D.; Cai, R.; Cui, H.; Long, Y.; Lu, Y.; Li, C.; She, Z. Cytotoxic and Antibacterial Preussomerins from the Mangrove Endophytic Fungus Lasiodiplodia Theobromae ZJ-HQ1. J. Nat. Prod. 2016, 79, 2397–2402. [Google Scholar] [CrossRef]
  69. Yu, G.; Wu, G.; Sun, Z.; Zhang, X.; Che, Q.; Gu, Q.; Zhu, T.; Li, D.; Zhang, G. Cytotoxic Tetrahydroxanthone Dimers from the Mangrove-Associated Fungus Aspergillus Versicolor HDN1009. Mar. Drugs 2018, 16, 335. [Google Scholar] [CrossRef]
  70. Li, T.; Wang, Y.; Li, L.; Tang, M.; Meng, Q.; Zhang, C.; Hua, E.; Pei, Y.; Sun, Y. New Cytotoxic Cytochalasans from a Plant-Associated Fungus Chaetomium Globosum Kz-19. Mar. Drugs 2021, 19, 438. [Google Scholar] [CrossRef]
  71. Wu, Y.; Chen, S.; Liu, H.; Huang, X.; Liu, Y.; Tao, Y.; She, Z. Cytotoxic Isocoumarin Derivatives from the Mangrove Endophytic Fungus Aspergillus sp. HN15-5D. Arch. Pharm. Res. 2019, 42, 326–331. [Google Scholar] [CrossRef]
  72. Zheng, C.J.; Bai, M.; Zhou, X.M.; Huang, G.L.; Shao, T.M.; Luo, Y.P.; Niu, Z.G.; Niu, Y.Y.; Chen, G.Y.; Han, C.R. Penicilindoles A-C, Cytotoxic Indole Diterpenes from the Mangrove-Derived Fungus Eupenicillium sp. HJ002. J. Nat. Prod. 2018, 81, 1045–1049. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, L.; Han, X.; Zhu, G.; Wang, Y.; Chairoungdua, A.; Piyachaturawat, P.; Zhu, W. Polyketides from the Endophytic Fungus Cladosporium sp. Isolated from the Mangrove Plant Excoecaria Agallocha. Front. Chem. 2018, 6, 344. [Google Scholar] [CrossRef] [PubMed]
  74. Siridechakorn, I.; Yue, Z.; Mittraphab, Y.; Lei, X.; Pudhom, K. Identification of Spirobisnaphthalene Derivatives with Anti-Tumor Activities from the Endophytic Fungus Rhytidhysteron Rufulum AS21B. Bioorg. Med. Chem. 2017, 25, 2878–2882. [Google Scholar] [CrossRef]
  75. Mahmoud, M.M.; Abdel-Razek, A.S.; Soliman, H.S.M.; Ponomareva, L.V.; Thorson, J.S.; Shaaban, K.A.; Shaaban, M. Diverse Polyketides from the Marine Endophytic Alternaria sp. LV52: Structure Determination and Cytotoxic Activities. Biotechnol. Rep. 2022, 33, e00628. [Google Scholar] [CrossRef] [PubMed]
  76. Ma, Y.M.; Liang, X.A.; Zhang, H.C.; Liu, R. Cytotoxic and Antibiotic Cyclic Pentapeptide from an Endophytic Aspergillus Tamarii of Ficus Carica. J. Agric. Food Chem. 2016, 64, 3789–3793. [Google Scholar] [CrossRef] [PubMed]
  77. Zhu, T.; Lu, Z.; Fan, J.; Wang, L.; Zhu, G.; Wang, Y.; Li, X.; Hong, K.; Piyachaturawat, P.; Chairoungdua, A.; et al. Ophiobolins from the Mangrove Fungus Aspergillus Ustus. J. Nat. Prod. 2018, 81, 2–9. [Google Scholar] [CrossRef]
  78. Yu, F.X.; Chen, Y.; Yang, Y.H.; Li, G.H.; Zhao, P.J. A New Epipolythiodioxopiperazine with Antibacterial and Cytotoxic Activities from the Endophytic Fungus Chaetomium sp. M336. Nat. Prod. Res. 2018, 32, 689–694. [Google Scholar] [CrossRef] [PubMed]
  79. Liu, H.X.; Liu, W.Z.; Chen, Y.C.; Sun, Z.H.; Tan, Y.Z.; Li, H.H.; Zhang, W.M. Cytotoxic Trichothecene Macrolides from the Endophyte Fungus Myrothecium Roridum. J. Asian Nat. Prod. Res. 2016, 18, 684–689. [Google Scholar] [CrossRef] [PubMed]
  80. Khan, I.; Zhang, H.; Liu, W.; Zhang, L.; Peng, F.; Chen, Y.; Zhang, Q.; Zhang, G.; Zhang, W.; Zhang, C. Identification and Bioactivity Evaluation of Secondary Metabolites from Antarctic-DerivedPenicillium ChrysogenumCCTCC M 2020019. RSC Adv. 2020, 10, 20738–20744. [Google Scholar] [CrossRef]
  81. Wang, J.; Bai, G.; Liu, Y.; Wang, H.; Li, Y.; Yin, W.; Wang, Y.; Lu, F. Cytotoxic Metabolites Produced by the Endophytic Fungus Aspergillus Clavatus. Chem. Lett. 2015, 44, 1148–1149. [Google Scholar] [CrossRef]
  82. Xiao, J.; Hu, J.Y.; Sun, H.D.; Zhao, X.; Zhong, W.T.; Duan, D.Z.; Wang, L.; Wang, X.L. Sinopestalotiollides A–D, Cytotoxic Diphenyl Ether Derivatives from Plant Endophytic Fungus Pestalotiopsis Palmarum. Bioorg. Med. Chem. Lett. 2018, 28, 515–518. [Google Scholar] [CrossRef]
  83. Zhang, X.; Tan, X.; Li, Y.; Wang, Y.; Yu, M.; Qing, J.; Sun, B.; Niu, S.; Ding, G. Hispidulones A and B, Two New Phenalenone Analogs from Desert Plant Endophytic Fungus Chaetosphaeronema Hispidulum. J. Antibiot. 2020, 73, 56–59. [Google Scholar] [CrossRef]
  84. Bashyal, B.P.; Wijeratne, E.M.K.; Tillotson, J.; Arnold, A.E.; Chapman, E.; Gunatilaka, A.A.L. Chlorinated Dehydrocurvularins and Alterperylenepoxide A from Alternaria sp. AST0039, a Fungal Endophyte of Astragalus Lentiginosus. J. Nat. Prod. 2017, 80, 427–433. [Google Scholar] [CrossRef] [PubMed]
  85. Zhan, J.; Burns, A.M.; Liu, M.X.; Faeth, S.H.; Gunatilaka, A.A.L. Search for Cell Motility and Angiogenesis Inhibitors with Potential Anticancer Activity: Beauvericin and Other Constituents of Two Endophytic Strains of Fusarium Oxysporum. J. Nat. Prod. 2007, 70, 227–232. [Google Scholar] [CrossRef] [PubMed]
  86. Adames, I.; Ortega, H.E.; Asai, Y.; Kato, M.; Nagaoka, K.; Tendyke, K.; Shen, Y.Y.; Cubilla-Ríos, L. 3-Epi-Waol A and Waol C: Polyketide-Derived γ-Lactones Isolated from the Endophytic Fungus Libertella Blepharis F2644. Tetrahedron Lett. 2015, 56, 252–255. [Google Scholar] [CrossRef]
  87. Ortega, H.E.; Graupner, P.R.; Asai, Y.; Tendyke, K.; Qiu, D.; Shen, Y.Y.; Rios, N.; Arnold, A.E.; Coley, P.D.; Kursar, T.A.; et al. Mycoleptodiscins A and B, Cytotoxic Alkaloids from the Endophytic Fungus Mycoleptodiscus sp. F0194. J. Nat. Prod. 2013, 76, 741–744. [Google Scholar] [CrossRef]
  88. Meng, L.H.; Li, X.M.; Lv, C.T.; Huang, C.G.; Wang, B.G. Brocazines A-F, Cytotoxic Bisthiodiketopiperazine Derivatives from Penicillium Brocae MA-231, an Endophytic Fungus Derived from the Marine Mangrove Plant Avicennia Marina. J. Nat. Prod. 2014, 77, 1921–1927. [Google Scholar] [CrossRef]
  89. Wei, H.; Xu, Y.; Espinosa-artiles, P.; Liu, M.X.; Luo, J.; Arnold, A.E.; Gunatilaka, A.A.L.; States, U.; States, U.; Biology, E.; et al. Sesquiterpenes and Other Constituents of Xylaria sp. NC1214, a Fungal Endophyte of the Moss Hypnum sp. Phytochemistry 2015, 118, 102–108. [Google Scholar] [CrossRef] [PubMed]
  90. Chinworrungsee, M.; Wiyakrutta, S.; Sriubolmas, N.; Chuailua, P.; Suksamrarn, A. Cytotoxic Activities of Trichothecenes Isolated from an Endophytic Fungus Belonging to Order Hypocreales. Arch. Pharm. Res. 2008, 31, 611–616. [Google Scholar] [CrossRef]
  91. Tantapakul, C.; Promgool, T.; Kanokmedhakul, K.; Soytong, K.; Song, J.; Hadsadee, S.; Jungsuttiwong, S.; Kanokmedhakul, S. Bioactive Xanthoquinodins and Epipolythiodioxopiperazines from Chaetomium globosum 7s-1, an Endophytic Fungus Isolated from Rhapis cochinchinensis (Lour.) Mart. Nat. Prod. Res. 2020, 34, 494–502. [Google Scholar] [CrossRef]
  92. Zhou, G.; Chen, X.; Zhang, X.; Che, Q.; Zhang, G.; Zhu, T.; Gu, Q.; Li, D. Prenylated P-Terphenyls from a Mangrove Endophytic Fungus, Aspergillus candidus LDJ-5. J. Nat. Prod. 2020, 83, 8–13. [Google Scholar] [CrossRef] [PubMed]
  93. Zhou, G.; Zhang, X.; Shah, M.; Che, Q.; Zhang, G.; Gu, Q.; Zhu, T.; Li, D. Polyhydroxy P-Terphenyls from a Mangrove Endophytic Fungus Aspergillus candidus LDJ-5. Mar. Drugs 2021, 19, 82. [Google Scholar] [CrossRef] [PubMed]
  94. Wu, Z.; Zhang, X.; Al Anbari, W.H.; Zhou, Q.; Zhou, P.; Zhang, M.; Zeng, F.; Chen, C.; Tong, Q.; Wang, J.; et al. Cysteine Residue Containing Merocytochalasans and 17,18-Seco-Aspochalasins from Aspergillus micronesiensis. J. Nat. Prod. 2019, 82, 2653–2658. [Google Scholar] [CrossRef] [PubMed]
  95. Zhou, M.; Zhou, K.; He, P.; Wang, K.M.; Zhu, R.Z.; Wang, Y.D.; Dong, W.; Li, G.P.; Yang, H.Y.; Ye, Y.Q.; et al. Antiviral and Cytotoxic Isocoumarin Derivatives from an Endophytic Fungus Aspergillus oryzae. Planta Med. 2016, 82, 414–417. [Google Scholar] [CrossRef]
  96. Cai, J.; Chen, C.; Tan, Y.; Chen, W.; Luo, X.; Luo, L.; Yang, B.; Liu, Y.; Zhou, X. Bioactive Polyketide and Diketopiperazine Derivatives from the Mangrove-Sediment-Derived Fungus Aspergillus sp. SCSIO41407. Molecules 2021, 26, 4851. [Google Scholar] [CrossRef]
  97. Xie, F.; Li, X.-B.; Zhou, J.-C.; Xu, Q.-Q.; Wang, X.-N.; Yuan, H.-Q.; Lou, H.-X. Secondary Metabolites from Aspergillus fumigatus,an Endophytic Fungus from the Liverwort Heteroscyphus tener (Steph.) Schiffn. Chem. Biodivers. 2015, 12, 1313–1321. [Google Scholar] [CrossRef]
  98. Li, H.; Zhang, R.; Cao, F.; Wang, J.; Hu, Z.; Zhang, Y. Proversilins A-E, Drimane-Type Sesquiterpenoids from the Endophytic Aspergillus Versicolor. J. Nat. Prod. 2020, 83, 2200–2206. [Google Scholar] [CrossRef]
  99. Li, X.G.; Pan, W.D.; Lou, H.Y.; Liu, R.M.; Xiao, J.H.; Zhong, J.J. New Cytochalasins from Medicinal Macrofungus Crodyceps Taii and Their Inhibitory Activities against Human Cancer Cells. Bioorg. Med. Chem. Lett. 2015, 25, 1823–1826. [Google Scholar] [CrossRef]
  100. Li, Q.; Chen, C.; Cheng, L.; Wei, M.; Dai, C.; He, Y.; Gong, J.; Zhu, R.; Li, X.N.; Liu, J.; et al. Emeridones A-F, a Series of 3,5-Demethylorsellinic Acid-Based Meroterpenoids with Rearranged Skeletons from an Endophytic Fungus Emericella sp. TJ29. J. Org. Chem. 2019, 84, 1534–1541. [Google Scholar] [CrossRef]
  101. Chen, Y.; Yang, W.; Zou, G.; Chen, S.; Pang, J.; She, Z. Bioactive Polyketides from the Mangrove Endophytic Fungi Phoma Sp. SYSU-SK-7. Fitoterapia 2019, 139, 104369. [Google Scholar] [CrossRef]
  102. Zhao, J.; Li, C.; Wang, W.; Zhao, C.; Luo, M.; Mu, F.; Fu, Y.; Zu, Y.; Yao, M. Hypocrea lixii, Novel Endophytic Fungi Producing Anticancer Agent Cajanol, Isolated from Pigeon Pea (Cajanus cajan [L.] Millsp.). J. Appl. Microbiol. 2013, 115, 102–113. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, Y.; Wang, G.; Yuan, Y.; Zou, G.; Yang, W.; Tan, Q.; Kang, W.; She, Z. Metabolites with Cytotoxic Activities from the Mangrove Endophytic Fungus Fusarium sp. 2ST2. Front. Chem. 2022, 10, 842405. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, Q.X.; Li, S.F.; Zhao, F.; Dai, H.Q.; Bao, L.; Ding, R.; Gao, H.; Zhang, L.X.; Wen, H.A.; Liu, H.W. Chemical Constituents from Endophytic Fungus Fusarium Oxysporum. Fitoterapia 2011, 82, 777–781. [Google Scholar] [CrossRef] [PubMed]
  105. Lin, T.; Wang, G.; Zhou, Y.; Zeng, D.; Liu, X.; Ding, R.; Jiang, X.; Zhu, D.; Shan, W.; Chen, H. Structure Elucidation and Biological Activity of Two New Trichothecenes from an Endophyte, Myrothecium Roridum. J. Agric. Food Chem. 2014, 62, 5993–6000. [Google Scholar] [CrossRef] [PubMed]
  106. Zhu, X.; Zhou, D.; Liang, F.; Wu, Z.; She, Z.; Li, C. Penochalasin K, a New Unusual Chaetoglobosin from the Mangrove Endophytic Fungus Penicillium Chrysogenum V11 and Its Effective Semi-Synthesis. Fitoterapia 2017, 123, 23–28. [Google Scholar] [CrossRef]
  107. Zhao, D.L.; Yuan, X.L.; Du, Y.M.; Zhang, Z.F.; Zhang, P. Benzophenone Derivatives from an Algal-Endophytic Isolate of Penicillium Chrysogenum and Their Cytotoxicity. Molecules 2018, 23, 3378. [Google Scholar] [CrossRef]
  108. Chen, J.; Huo, L.N.; Gao, Y.; Zhang, Y.L.; Chen, Y. Two New N-Acetyl-ᴅ-Glucosamine Derivatives from the Medical Algae-Derived Endophytic Fungus Penicillium Chrysogenum. Nat. Prod. Res. 2022, 36, 3988–3991. [Google Scholar] [CrossRef]
  109. Cai, X.Y.; Wang, J.P.; Shu, Y.; Hu, J.T.; Sun, C.T.; Cai, L.; Ding, Z.T. A New Cytotoxic Indole Alkaloid from the Fungus Penicillium Polonicum TY12. Nat. Prod. Res. 2022, 36, 2270–2276. [Google Scholar] [CrossRef]
  110. Tang, J.W.; Kong, L.M.; Zu, W.Y.; Hu, K.; Li, X.N.; Yan, B.C.; Wang, W.G.; Sun, H.D.; Li, Y.; Puno, P.T. Isopenicins A-C: Two Types of Antitumor Meroterpenoids from the Plant Endophytic Fungus Penicillium sp. Sh18. Org. Lett. 2019, 21, 771–775. [Google Scholar] [CrossRef]
  111. Noumeur, S.R.; Helaly, S.E.; Jansen, R.; Gereke, M.; Stradal, T.E.B.; Harzallah, D.; Stadler, M. Preussilides A-F, Bicyclic Polyketides from the Endophytic Fungus Preussia Similis with Antiproliferative Activity. J. Nat. Prod. 2017, 80, 1531–1540. [Google Scholar] [CrossRef]
  112. Lai, D.; Wang, A.; Cao, Y.; Zhou, K.; Mao, Z.; Dong, X.; Tian, J.; Xu, D.; Dai, J.; Peng, Y.; et al. Bioactive Dibenzo-α-Pyrone Derivatives from the Endophytic Fungus Rhizopycnis Vagum Nitaf22. J. Nat. Prod. 2016, 79, 2022–2031. [Google Scholar] [CrossRef] [PubMed]
  113. Balde, E.S.; Andolfi, A.; Bruyère, C.; Cimmino, A.; Lamoral-Theys, D.; Vurro, M.; Van Damme, M.; Altomare, C.; Mathieu, V.; Kiss, R.; et al. Investigations of Fungal Secondary Metabolites with Potential Anticancer Activity. J. Nat. Prod. 2010, 73, 969–971. [Google Scholar] [CrossRef]
  114. Rehman, S.U.; Yang, L.J.; Zhang, Y.H.; Wu, J.S.; Shi, T.; Haider, W.; Shao, C.L.; Wang, C.Y. Sorbicillinoid Derivatives From Sponge-Derived Fungus Trichoderma Reesei (HN-2016-018). Front. Microbiol. 2020, 11, 1334. [Google Scholar] [CrossRef]
  115. Gao, S.S.; Li, X.M.; Williams, K.; Proksch, P.; Ji, N.Y.; Wang, B.G. Rhizovarins A-F, Indole-Diterpenes from the Mangrove-Derived Endophytic Fungus Mucor Irregularis QEN-189. J. Nat. Prod. 2016, 79, 2066–2074. [Google Scholar] [CrossRef] [PubMed]
  116. Kumaran, R.S.; Kim, H.J.; Hur, B.K. Taxol Promising Fungal Endophyte, Pestalotiopsis Species Isolated from Taxus Cuspidata. J. Biosci. Bioeng. 2010, 110, 541–546. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, Y.; Liu, H.X.; Chen, Y.C.; Sun, Z.H.; Li, H.H.; Li, S.N.; Yan, M.L.; Zhang, W.M. Two New Metabolites from the Endophytic Fungus Alternaria sp. A744 Derived from Morinda officinalis. Molecules 2017, 22, 765. [Google Scholar] [CrossRef]
  118. Suresh, G.; Kokila, D.; Suresh, T.C.; Kumaran, S.; Velmurugan, P.; Vedhanayakisri, K.A.; Sivakumar, S.; Ravi, A.V. Mycosynthesis of Anticancer Drug Taxol by Aspergillus Oryzae, an Endophyte of Tarenna Asiatica, Characterization, and Its Activity against a Human Lung Cancer Cell Line. Biocatal. Agric. Biotechnol. 2020, 24, 101525. [Google Scholar] [CrossRef]
  119. Wang, M.; Sun, Z.H.; Chen, Y.C.; Liu, H.X.; Li, H.H.; Tan, G.H.; Li, S.N.; Guo, X.L.; Zhang, W.M. Cytotoxic Cochlioquinone Derivatives from the Endophytic Fungus Bipolaris Sorokiniana Derived from Pogostemon Cablin. Fitoterapia 2016, 110, 77–82. [Google Scholar] [CrossRef] [PubMed]
  120. Liu, H.X.; Tan, H.B.; Chen, Y.C.; Li, S.N.; Li, H.H.; Zhang, W.M. Cytotoxic Triquinane-Type Sesquiterpenoids from the Endophytic Fungus Cerrena sp. A593. Nat. Prod. Res. 2020, 34, 2430–2436. [Google Scholar] [CrossRef]
  121. Bashyal, B.P.; Wijeratne, E.M.K.; Faeth, S.H.; Gunatilaka, A.A.L. Globosumones A-C, Cytotoxic Orsellinic Acid Esters from the Sonoran Desert Endophytic Fungus Chaetomium globosum. J. Nat. Prod. 2005, 68, 724–728. [Google Scholar] [CrossRef]
  122. Liu, H.; Tan, H.; Chen, Y.; Guo, X.; Wang, W.; Guo, H.; Liu, Z.; Zhang, W. Cytorhizins A-D, Four Highly Structure-Combined Benzophenones from the Endophytic Fungus Cytospora Rhizophorae. Org. Lett. 2019, 21, 1063–1067. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, Y.; Liu, Z.; Huang, Y.; Liu, L.; He, J.; Wang, L.; Yuan, J.; She, Z. Ascomylactams A-C, Cytotoxic 12- or 13-Membered-Ring Macrocyclic Alkaloids Isolated from the Mangrove Endophytic Fungus Didymella sp. CYSK-4, and Structure Revisions of Phomapyrrolidones A and C. J. Nat. Prod. 2019, 82, 1752–1758. [Google Scholar] [CrossRef] [PubMed]
  124. Rao, L.; You, Y.X.; Su, Y.; Liu, Y.; He, Q.; Fan, Y.; Hu, F.; Xu, Y.K.; Zhang, C.R. Two Spiroketal Derivatives with an Unprecedented Amino Group and Their Cytotoxicity Evaluation from the Endophytic Fungus Pestalotiopsis Flavidula. Fitoterapia 2019, 135, 5–8. [Google Scholar] [CrossRef] [PubMed]
  125. Wijeratne, E.M.K.; Paranagama, P.A.; Marron, M.T.; Gunatilaka, M.K.; Arnold, A.E.; Gunatilaka, A.A.L. Sesquiterpene Quinones and Related Metabolites from Phyllosticta Spinarum, a Fungal Strain Endophytic in Platycladus Orientalis of the Sonoran Desert. J. Nat. Prod. 2008, 71, 218–222. [Google Scholar] [CrossRef]
  126. Deshmukh, S.K.; Mishra, P.D.; Kulkarni-Almeida, A.; Verekar, S.; Sahoo, M.R.; Periyasamy, G.; Goswami, H.; Khanna, A.; Balakrishnan, A.; Vishwakarma, R. Anti-Inflammatory and Anticancer Activity of Ergoflavin Isolated from an Endophytic Fungus. Chem. Biodivers. 2009, 6, 784–789. [Google Scholar] [CrossRef]
  127. Lai, D.; Mao, Z.; Zhou, Z.; Zhao, S.; Xue, M.; Dai, J.; Zhou, L.; Li, D. New Chlamydosporol Derivatives from the Endophytic Fungus Pleosporales Sp. Sigrf05 and Their Cytotoxic and Antimicrobial Activities. Sci. Rep. 2020, 10, 8193. [Google Scholar] [CrossRef]
  128. Isaka, M.; Palasarn, S.; Lapanun, S.; Chanthaket, R.; Boonyuen, N.; Lumyong, S. γ-Lactones and Ent-Eudesmane Sesquiterpenes from the Endophytic Fungus Eutypella sp. BCC 13199. J. Nat. Prod. 2009, 72, 1720–1722. [Google Scholar] [CrossRef]
  129. Bunyapaiboonsri, T.; Yoiprommarat, S.; Srikitikulchai, P.; Srichomthong, K.; Lumyong, S. Oblongolides from the Endophytic Fungus Phomopsis Sp. BCC 9789. J. Nat. Prod. 2010, 73, 55–59. [Google Scholar] [CrossRef]
  130. Isaka, M.; Chinthanom, P.; Boonruangprapa, T.; Rungjindamai, N.; Pinruan, U. Eremophilane-Type Sesquiterpenes from the Fungus Xylaria Sp. BCC 21097. J. Nat. Prod. 2010, 73, 683–687. [Google Scholar] [CrossRef]
  131. Sawadsitang, S.; Mongkolthanaruk, W.; Suwannasai, N.; Sodngam, S. Antimalarial and Cytotoxic Constituents of Xylaria Cf. Cubensis PK108. Nat. Prod. Res. 2015, 29, 2033–2036. [Google Scholar] [CrossRef]
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

Rybczyńska-Tkaczyk, K.; Grenda, A.; Jakubczyk, A.; Krawczyk, P. Natural Bacterial and Fungal Peptides as a Promising Treatment to Defeat Lung Cancer Cells. Molecules 2023, 28, 4381. https://doi.org/10.3390/molecules28114381

AMA Style

Rybczyńska-Tkaczyk K, Grenda A, Jakubczyk A, Krawczyk P. Natural Bacterial and Fungal Peptides as a Promising Treatment to Defeat Lung Cancer Cells. Molecules. 2023; 28(11):4381. https://doi.org/10.3390/molecules28114381

Chicago/Turabian Style

Rybczyńska-Tkaczyk, Kamila, Anna Grenda, Anna Jakubczyk, and Paweł Krawczyk. 2023. "Natural Bacterial and Fungal Peptides as a Promising Treatment to Defeat Lung Cancer Cells" Molecules 28, no. 11: 4381. https://doi.org/10.3390/molecules28114381

Article Metrics

Back to TopTop