Open Access

Influence of tumor mycobiome on cancer pathogenesis (Review)

  • Authors:
    • Weipeng Liu
    • Zongrui Li
    • Xiaopeng Li
    • Haiyang Cao
    • He Jiang
    • Qingbin Niu
    • Baoguang Hu
  • View Affiliations

  • Published online on: November 2, 2023     https://doi.org/10.3892/ol.2023.14128
  • Article Number: 541
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer tissues harbor a large microbiome. There is growing evidence that the tumor microbiome is significantly correlated with the prognosis of cancer patients, but the exact underlying mechanisms have remained elusive. Although the tumor mycobiome is less abundant than the biome of bacteria, it is prevalent in most cancers in humans. The present review describes in detail the impact of the tumor mycobiome on cancer pathogenesis. The tumor mycobiome promotes tumor progression and metastasis by affecting the human immune system, maintaining a pro‑inflammatory environment, producing aflatoxins, attenuating cell adhesion mechanisms and fungal‑bacterial interactions. Furthermore, the tumor mycobiome likewise has great potential for cancer prevention, diagnosis and treatment.

Introduction

Cancer is not only a major public health problem worldwide, but also one of the leading causes of human mortality. Host-microbe immune interactions profoundly influence cancer development, progression and treatment outcomes (111). Fungi and bacteria co-colonize the mammalian skin epithelium, respiratory tract, gastrointestinal tract and reproductive organs, forming a complex ecosystem of microbe-microbe and host-microbe interactions that significantly impact human health (1221). Although only ~0.1% of microbial DNA is present in the gut (22), fungal infections cause more than 1.5 million deaths worldwide each year (23).

There is growing evidence linking the human microbiome (bacteria, fungi and viruses) to cancer and cancer outcomes (24,25). In recent years, several bacteria have been observed to be associated with cancer development and progression. Helicobacter pylori infection is the strongest risk factor for the development of malignant tumors in the stomach, and epidemiologic studies have determined that the attributable risk of gastric cancer due to Helicobacter pylori is ~75% (26). At the same time, in the lower gastrointestinal tract, genotoxic Escherichia coli, Bacteroides fragilis, Streptococcus bovis and Fusobacterium nucleatum are associated with the pathogenesis of colorectal cancer (27). Among these cancer-associated bacteria, they can modulate host immunity and cause chronic inflammation, which is thought to have oncogenic effects. Recent reports have also shown that bacterial DNA circulating in the blood of cancer patients can be used as a predictive biomarker for tumors (28,29) and intracellular bacteria have been found in numerous tumor types (30).

In recent times, scientists have been exploring the link between cancer and bacteria and viruses, but few studies have focused on the relationship between fungi and cancer. Although the human fungal biome is less abundant than the bacterial group, it can still significantly affect human health (3133). Previous studies have demonstrated that fungal DNA is present in most human cancer tissues. However, the role and impact of the tumor mycobiome on cancer pathogenesis remain largely unknown.

Presence of tumor mycobiome within the tumor tissue

Narunsky-Haziza et al (34) statistically characterized the tumor mycobiome in 17,401 tissue, blood and plasma samples from four independent cohorts of 35 cancer types using internal transcribed spacer sequencing and whole-genome sequencing methods. Statistics revealed that 5 fungi, including Malasseziomycetes, Saccharomycetes, Diothideomycetes, Sordariomycetes and Candida, were significantly enriched in all types of cancer (lung, breast, melanoma, osteosarcoma, ovarian, gastric, colorectal and head and neck tumors). By contrast, certain specific fungi (Microbotryomycetes, Wallemiomycetes, Agaricomycetes, Tremellomycetes) were only found in specific cancer sites, so the tumor mycobiome is prevalent in most cancers in humans (35).

Differences in fungal biome composition in cancer patients compared to healthy controls

Fungal biome imbalance was found in patients with gastric cancer. The fungal biome characteristics of patients with gastric cancer differed significantly from controls, with reduced diversity and enrichment of Candida and Alternaria in gastric cancer tissues (36). Colorectal fungi were altered in patients with colorectal cancer compared to normal subjects. Patients with colorectal cancer had increased Malasseziomycetes, decreased Saccharomycetes and a distorted ratio of Basidiomycota to Ascomycota (37). The distribution of the fungal genera Aspergillus, Malassezia, Rhodotorula, Pseudogymnoascus, Kwoniella, Talaromyces, Debaryomyces, Moniliophthora, Pneumocystis and Nosemia was altered in colorectal cancer, as verified in independent Chinese and European cohorts (38).

The tumor mycobiome is less abundant than the bacterial one, and both have a symbiotic relationship in tumors

Analysis of the tumor mycobiome in various body parts showed a maximum of 1 fungal cell per 10,000 tumor cells (39). In The Cancer Genome Atlas (TCGA) primary tumors, the average relative abundance of bacteria and fungi was 96 and 4%, respectively, which confirms the lower abundance of fungi compared to bacteria (34). It has been found that there is a symbiotic rather than competitive ecological interaction between fungal and bacterial biomes in the tumor microenvironment (35). However, this differs from the manifestation of alternating fungal and bacterial populations in the gastrointestinal tract (9,40).

The tumor mycobiome promotes cancer progression and metastasis

The tumor mycobiome not only resides in tumors but also promotes tumor progression and metastasis and spreads systemically by affecting the human immune system, maintaining a pro-inflammatory environment, producing aflatoxins, attenuating cell adhesion mechanisms and fungal-bacterial interactions (Fig. 1).

The tumor mycobiome affects the surveillance of cancer by the human immune system

As numerous cancer patients are immunosuppressed, they are more susceptible to fungal infections, which may further aggravate their condition (41). It has been demonstrated that fungal-driven pancreatic cancer occurs through complement cascade activation and IL-33 secretion (35). Aykut et al (31) have shown that Malassezia can secrete hydrolases to release host lipids and activate the C3 complement mannose-binding lectin pathway to promote an immunosuppressive tumor environment in pancreatic cancer. The tumor mycobiome activates dectin-1-mediated Src-Syk-caspase recruitment domain family member 9 (CARD9) signaling in the pancreas, leading to IL-33 secretion and tumor growth, and thus, this may be the mechanism by which the tumor mycobiome promotes pancreatic cancer progression (42).

The Colorectal Cancer Risk Factor Assessment report indicated that the human body has an inadequate immune response to fungi, such as inflammatory bowel disease (43) and ulcerative colitis (12). Antifungal treatment has been reported to exacerbate colitis and colorectal cancer, while colonic fungi enhance azoxymethane/dextran sodium sulfate-induced inflammatory vesicle activation in colitis (44).

The tumor mycobiome maintains a pro-inflammatory environment and promotes cancer progression

There are numerous hypotheses about tumor pathogenesis, among which the theory of an inflammatory mechanism is a widely accepted hypothesis. Inflammation is usually the basis for resistance to harmful stimuli, accelerating wound recovery and maintaining normal tissue function, and its role involves endothelial cells, immune cells and inflammatory factors (45).

Self-limiting acute inflammation benefits the healing process (46). However, when it gets out of control, it may develop into chronic inflammation that induces tissue lesions and predisposes to cancer (47), including tumorigenesis, progression and metastasis (48). Only a small percentage of cancers are attributed to germ cell lineage mutations, while 90% of cancers are associated with somatic mutations and environmental hazards, and the latter is always associated with chronic inflammation or infection (49). Epidemiological surveys have shown that inflammation is strongly associated with the development of ~20% of cancers (50). Available evidence suggests that hypoxia-associated inflammatory cytokines or chemokines, such as IL-1, IL-6 and TNF, are significantly elevated in the tumor microenvironment (51). Cancer patients may benefit from anti-inflammatory drugs, such as TNF blockers and non-steroidal anti-inflammatory drugs (52,53).

Malassezia

In cancer patients, higher levels of Malassezia are associated with unfavorable prognosis (54). Malassezia also exhibits various pro-inflammatory biological properties, such as disruption of the epithelial barrier, enrichment of inflammatory factors and degradation of the extracellular matrix, all of which can promote tumor formation and malignant progression (54). Malassezia can activate NLR family pyrin domain containing 3 inflammasome via Dectin2/caspase recruitment domain family member 9 signaling and accelerate IL-1β production to exacerbate inflammation (55). Zhang et al (56) also demonstrated that Malassezia could produce nanovesicles rich in allergens or proteins, which may trigger and maintain inflammation by activating the NF-κB pathway and upregulating IL-6 production in the immune microenvironment (Fig. 2).

In addition, other mechanisms may be involved in Malassezia-associated inflammatory cancer transformation processes, such as DNA lesion accumulation and imbalance of oncogenes and anti-oncogenes. Inflammatory cells may induce DNA damage by releasing cytotoxic reactive oxygen species (57). A persistent inflammatory state may lead to increased and accumulated DNA damage in cells, which may promote genetic mutations, generate genomic instability and ultimately produce oncogenic effects (58).

Candida

Candida is functionally associated with a variety of cancers. Studies have shown that gastrointestinal cancers have different relative abundances of Candida and Saccharomyce, so gastrointestinal cancers can be classified as Candida- and Saccharomyce-associated tumors (35).

Candida-dominant tumors are associated with enhanced expression of IL-1 pro-inflammatory immune pathways and increased neutrophils, a major inflammatory cytokine that plays a crucial role in carcinogenesis and tumor progression (35). Candida increases inflammation, which promotes Candida colonization, thereby maintaining a pro-inflammatory environment, leading to a vicious cycle that persists (35). Therefore, prevention and management of Candida infection and associated inflammation may help to block this destructive inflammatory state in cancer and may be a reasonable combination therapy option during cancer treatment.

Of note, there are interactions between Candida and different bacteria in gastric cancer. Candida was observed to be positively correlated with Lactobacillus and negatively correlated with Helicobacter pylori (35).

The tumor mycobiome produces aflatoxins that promote cancer progression

Colorectal cancer is the third most common cancer type worldwide, with >500,000 related deaths per year (59,60). The contribution of the intestinal flora to colorectal cancer progression has been widely recognized (6163). Intestinal fungi constitute a significant component of the human intestinal flora, but their role in colorectal cancer has remained elusive (64). Several studies have confirmed a correlation between intestinal fungi and colon cancer (38,44,6567).

Lin et al (64) conducted a meta-analysis using shotgun metagenomics pooling 1,329 metagenomes from 8 cohorts (454 colorectal cancers, 350 adenomas and 525 healthy subjects) to evaluate the lesser bias of the gut fungal biome on colorectal cancer. Statistical analysis of the intestinal fungal biome composition showed that Aspergillus rambellii was identified as the most abundant fungal species. Seven of the eight cohorts showed a consistent association of Aspergillus rambellii with colorectal cancer. Further studies showed that Aspergillus rambellii promoted colorectal cancer cell growth in vitro and tumor growth in xenograft mice (64).

Aspergillus rambellii has been shown to have the ability to produce multiple aflatoxins (e.g., aflatoxin B, aflatoxin G and the aflatoxin precursor sterigmatocystin) (6870). The association of Aspergillus spp. of fungi with cancer has been frequently reported (71). Aflatoxins are toxins of fungal origin classified as carcinogens and mutagens, exemplified by their powerful liver cancer-promoting effects (62). For instance, long-term consumption of foods containing aflatoxins was determined to be associated with a significantly increased risk of liver cancer (72). Because aflatoxins can damage macrophages and dendritic cells by activating Toll-like receptors, they can induce immune dysregulation and promote tumor progression (73,74) (Fig. 3).

The tumor mycobiome attenuates cell adhesion mechanisms and promotes cancer metastasis

In colon cancer, Candida not only predicts disease but is also associated with diminished cell adhesion mechanisms and tumor metastasis (35). Loss of epithelial barrier function and increased tight junction permeability are standard features of lower gastrointestinal cancers (75) and are high-risk factors for tumor metastasis (76). Malassezia can promote cancer metastasis by disrupting the epithelial barrier (54). Dohlman et al (39) found that tumor and blood samples from the same patient had highly similar fungal DNA, suggesting that an increased abundance of Candida in advanced metastatic gastrointestinal tumors directly or indirectly leads to genetic dysregulation of cell adhesion, resulting in a weakened epithelial barrier and translocation of fungal DNA from the primary tumor site into the bloodstream, promoting tumor metastasis.

Tumor fungal-bacterial biome interactions promote cancer progression

Through extensive genomic analysis, tumor fungal-bacterial biome interactions can promote colorectal carcinogenesis through the upregulation of D-arginine and D-ornithine metabolic pathways and stimulation of the butanoate metabolic pathway (77). Liu et al (77) demonstrated that two marker genes, oraS and oraE, in the D-arginine and D-ornithine metabolic pathways were upregulated in colorectal cancer. The butanoate metabolic pathway, which is strongly activated in colorectal cancer but less studied, has also been identified (78). Tumor fungal-bacterial biome interactions promote colorectal cancer progression through upregulation of bdhA and bdhB gene expression in the butanoate metabolic pathway (77). Therefore, butanoate in the butanoate metabolic pathway is crucial in supporting the tumor microenvironment (79). Tumor fungal-bacterial biome interactions are being explored as an effective means of maintaining homeostasis in the gut.

The tumor mycobiome may be used as a marker for cancer diagnosis

Numerous studies have indicated the potential of bacteria as biomarkers for the diagnosis of colorectal cancer (64). Wirbel et al (80) and Thomas et al (81) performed meta-analyses to identify several bacteria enriched in colorectal cancer with utility as diagnostic biomarkers for colorectal cancer.

By contrast, with the study of the tumor microbiome, fungi, in addition to bacteria, may be used as biomarkers for the noninvasive diagnosis of tumor patients. In TCGA cohorts, fungal biome richness varied significantly among cancers (35). For instance, Candida, Saccharomyces cerevisiae and Cyberlindnera jadinii are highly abundant in the gastrointestinal tumor fungal organism community and Blastomyces and Malassezia are highly abundant in lung cancer and breast cancer, respectively (39).

A further study performed qualitative and quantitative analyses of 20 different fungal DNAs released into the bloodstream and the results suggested that they may be used to distinguish patients with cancer from healthy individuals, even in early-stage disease (35). This suggests that the tumor fungal biome has utility in cancer diagnosis.

In the study conducted by Lin et al (64), the average areas under the receiver operating characteristic curves (AUC) of pure bacterial biomarkers was only 73%, while the combination of fungal and bacterial mixed biomarkers showed a significant improvement in diagnostic performance with an average AUC of 83% and an increase in the relative change in AUC of 1.44–10.60% (64). This suggests that the combination of fungal and bacterial biomarkers is more accurate than the combination of pure bacterial species in differentiating patients with colorectal cancer from healthy individuals, thus highlighting the potential use of the tumor mycobiome in clinical diagnostic applications.

In the study of colorectal cancer conducted by Liu et al (77), a comprehensive analysis of different national microbiomes was performed using colorectal cancer metagenomic datasets of 8 different cohorts. They found that fungi, archaea and viruses were able to distinguish patients with colorectal cancer from healthy controls in multiple geographic cohorts (77). Coker et al (38) successfully distinguished 184 patients with colorectal cancer from 204 healthy controls by detecting fungal biomes in the stool.

Candida is transcriptionally active in gastrointestinal tumors (35). Enrichment of tumor-associated Candida DNA was found to be significantly associated with reduced survival in patients with gastrointestinal tumors due to the association of Candida with gene expression for cytosolic DNA sensing, Toll-like receptor signaling and Nod-like receptor signaling in gastric cancer (39). This not only suggests that Candida increases the severity of gastrointestinal tumors but also that Candida may be a promising biomarker for predicting disease outcomes.

The tumor mycobiome has potential preventive or therapeutic value for cancer

The use of antimicrobial agents that target known pathogenic microorganisms effectively prevents the onset and progression of the disease. The use of targeted antifungal agents is helpful in the prevention or treatment of gastrointestinal cancers (37). In a mouse model of human pancreatic ductal adenocarcinoma, Malassezia infiltrates and accelerates the progression of human pancreatic ductal adenocarcinoma, a condition that can be reversed by antifungal treatment (31). Antifungal treatment targeting Malassezia resulted in a 40% reduction in the incidence of pancreatic cancer in mice (31). In a mouse model of esophageal cancer, the oral fungus Dictyostelium significantly increased the severity of esophageal squamous cell carcinoma, a condition that could be reversed by antifungal treatment (82).

In addition, the use of fungal probiotics can prevent and treat gastrointestinal cancers. Probiotics are microorganisms that improve health when consumed in the correct amounts. The most common probiotics are bacteria that have been shown to inhibit the proliferation of pathogenic intestinal microorganisms and to prevent carcinogenic inflammation in the esophagus, stomach, pancreas and colorectum (8385). By contrast, fungi can also be ingested as probiotics and have been reported to be used to alleviate gastrointestinal cancers (37). The Helicobacter pylori eradication rate improved when Saccharomyces boulardii was combined with Lactobacillus gasseri, Lactobacillus reuteri, Lactobacillus acidophilus, Streptococcus faecalis, Bacillus subtilis and Bifidobacterium (86).

The use of fungal probiotics in treating gastrointestinal cancers is of great importance. Because of their characteristic cellular structure, they can survive in the unfavorable environment of the gastrointestinal tract (87,88). As more research on the efficacy and safety of fungal probiotics is conducted, they may directly or indirectly modulate the tumor microbiome to prevent and treat gastrointestinal cancers (37).

With the above summary and analysis, the fungal biome that targets and attacks tumors is likely to be a good way to treat cancer.

Conclusion and future perspective

This review provides a comprehensive summary of the impact of the tumor mycobiome on cancer pathogenesis. The tumor mycobiome promotes tumor progression and metastasis by affecting the human immune system, maintaining a pro-inflammatory environment, producing aflatoxins, attenuating cellular adhesion mechanisms and fungal-bacterial interactions. Furthermore, the tumor mycobiome also has tremendous potential for cancer prevention, diagnosis and treatment.

Although studies on the effect of the tumor mycobiome on cancer pathogenesis have become more frequent, the results are not uniform because of the differences between different populations and inconsistent standards for metagenomic data generation and processing. The future development of standardized and low-cost sequencing technologies and pipeline analysis methods to improve the quality of data collection and analytical processing, and the initiation of longitudinal studies with large sample sizes in different populations to clarify the specific mechanistic relationships, will be crucial for research in this field.

Acknowledgements

Not applicable.

Funding

This study was supported by a grant from the Natural Science Foundation of Shandong (grant no. ZR2017LH050).

Availability of data and materials

Not applicable.

Authors' contributions

WL wrote the manuscript, searched the literature and prepared the figures. ZL was involved in the design of the study and revised the manuscript. BH provided ideas to improve the article, modified the figures and revised the manuscript. XL, HC, HJ and QN performed the literature search and selected relevant articles. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al: Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 371:595–602. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Dzutsev A, Badger JH, Perez-Chanona E, Roy S, Salcedo R, Smith CK and Trinchieri G: Microbes and cancer. Annu Rev Immunol. 35:199–228. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Finlay BB, Goldszmid R, Honda K, Trinchieri G, Wargo J and Zitvogel L: Can we harness the microbiota to enhance the efficacy of cancer immunotherapy? Nat Rev Immunol. 20:522–528. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Garrett WS: The gut microbiota and colon cancer. Science. 364:1133–1135. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Grivennikov SI, Greten FR and Karin M: Immunity, inflammation, and cancer. Cell. 140:883–899. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al: Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 342:967–970. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al: Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 359:91–97. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Sharma P, Hu-Lieskovan S, Wargo JA and Ribas A: Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 168:707–723. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Shiao SL, Kershaw KM, Limon JJ, You S, Yoon J, Ko EY, Guarnerio J, Potdar AA, McGovern DPB, Bose S, et al: Commensal bacteria and fungi differentially regulate tumor responses to radiation therapy. Cancer cell. 39:1202–1213.e6. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Spencer CN, McQuade JL, Gopalakrishnan V, McCulloch JA, Vetizou M, Cogdill AP, Khan MAW, Zhang X, White MG, Peterson CB, et al: Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science. 374:1632–1640. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Tanoue T, Morita S, Plichta DR, Skelly AN, Suda W, Sugiura Y, Narushima S, Vlamakis H, Motoo I, Sugita K, et al: A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 565:600–605. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Sokol H, Leducq V, Aschard H, Pham HP, Jegou S, Landman C, Cohen D, Liguori G, Bourrier A, Nion-Larmurier I, et al: Fungal microbiota dysbiosis in IBD. Gut. 66:1039–1048. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Findley K, Oh J, Yang J, Conlan S, Deming C, Meyer JA, Schoenfeld D, Nomicos E, Park M; NIH Intramural Sequencing Center Comparative Sequencing Program, ; et al: Topographic diversity of fungal and bacterial communities in human skin. Nature. 498:367–370. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Hoarau G, Mukherjee PK, Gower-Rousseau C, Hager C, Chandra J, Retuerto MA, Neut C, Vermeire S, Clemente J, Colombel JF, et al: Bacteriome and mycobiome interactions underscore microbial dysbiosis in familial Crohn's disease. mBio. 7:e01250–16. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Leonardi I, Paramsothy S, Doron I, Semon A, Kaakoush NO, Clemente JC, Faith JJ, Borody TJ, Mitchell HM, Colombel JF, et al: Fungal trans-kingdom dynamics linked to responsiveness to fecal microbiota transplantation (FMT) therapy in ulcerative colitis. Cell Host Microbe. 27:823–829.e3. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Doron I, Mesko M, Li XV, Kusakabe T, Leonardi I, Shaw DG, Fiers WD, Lin WY, Bialt-DeCelie M, Román E, et al: Mycobiota-induced IgA antibodies regulate fungal commensalism in the gut and are dysregulated in Crohn's disease. Nat Microbiol. 6:1493–1504. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Lewis JD, Chen EZ, Baldassano RN, Otley AR, Griffiths AM, Lee D, Bittinger K, Bailey A, Friedman ES, Hoffmann C, et al: Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn's disease. Cell Host Microbe. 18:489–500. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Liguori G, Lamas B, Richard ML, Brandi G, da Costa G, Hoffmann TW, Di Simone MP, Calabrese C, Poggioli G, Langella P, et al: Fungal dysbiosis in mucosa-associated microbiota of Crohn's disease patients. J Crohns Colitis. 10:296–305. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Tipton L, Müller CL, Kurtz ZD, Huang L, Kleerup E, Morris A, Bonneau R and Ghedin E: Fungi stabilize connectivity in the lung and skin microbial ecosystems. Microbiome. 6:122018. View Article : Google Scholar : PubMed/NCBI

20 

Zhai B, Ola M, Rolling T, Tosini NL, Joshowitz S, Littmann ER, Amoretti LA, Fontana E, Wright RJ, Miranda E, et al: High-resolution mycobiota analysis reveals dynamic intestinal translocation preceding invasive candidiasis. Nat Med. 26:59–64. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Zuo T, Wong SH, Cheung CP, Lam K, Lui R, Cheung K, Zhang F, Tang W, Ching JYL, Wu JCY, et al: Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat Commun. 9:36632018. View Article : Google Scholar : PubMed/NCBI

22 

Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al: A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 464:59–65. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Brown GD, Denning DW, Gow NA, Levitz SM, Netea MG and White TC: Hidden killers: Human fungal infections. Sci Transl Med. 4:165rv132012. View Article : Google Scholar : PubMed/NCBI

24 

Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V and Wargo JA: The microbiome, cancer, and cancer therapy. Nat Med. 25:377–388. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Vogtmann E and Goedert JJ: Epidemiologic studies of the human microbiome and cancer. Br J Cancer. 114:237–242. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Polk DB and Peek RM Jr: Helicobacter pylori: Gastric cancer and beyond. Nat Rev Cancer. 10:403–414. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA and Knight R: The microbiome and human cancer. Science. 371:eabc45522021. View Article : Google Scholar : PubMed/NCBI

28 

Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, Wandro S, Kosciolek T, Janssen S, Metcalf J, Song SJ, et al: Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature. 579:567–574. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Dohlman AB, Arguijo Mendoza D, Ding S, Gao M, Dressman H, Iliev ID, Lipkin SM and Shen X: The cancer microbiome atlas: A pan-cancer comparative analysis to distinguish tissue-resident microbiota from contaminants. Cell Host Microbe. 29:281–298.e5. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, Rotter-Maskowitz A, Weiser R, Mallel G, Gigi E, et al: The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 368:973–980. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Aykut B, Pushalkar S, Chen R, Li Q, Abengozar R, Kim JI, Shadaloey SA, Wu D, Preiss P, Verma N, et al: The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature. 574:264–267. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Elaskandrany M, Patel R, Patel M, Miller G, Saxena D and Saxena A: Fungi, host immune response, and tumorigenesis. Am J Physiol Gastrointest Liver Physiol. 321:G213–G222. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Iliev ID and Leonardi I: Fungal dysbiosis: Immunity and interactions at mucosal barriers. Nat Rev Immunol. 17:635–646. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, Gavert N, Stajich JE, Amit G, González A, et al: Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 185:3789–3806.e17. 2022. View Article : Google Scholar : PubMed/NCBI

35 

Li X and Saxena D: The tumor mycobiome: A paradigm shift in cancer pathogenesis. Cell. 185:3648–3651. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Zhong M, Xiong Y, Zhao J, Gao Z, Ma J, Wu Z, Song Y and Hong X: Candida albicans disorder is associated with gastric carcinogenesis. Theranostics. 11:4945–4956. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Coker OO: Non-bacteria microbiome (virus, fungi, and archaea) in gastrointestinal cancer. J Gastroenterol Hepatol. 37:256–262. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, Chan FKL, Sung JJY and Yu J: Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 68:654–662. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Dohlman AB, Klug J, Mesko M, Gao IH, Lipkin SM, Shen X and Iliev ID: A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell. 185:3807–3822.e12. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Seelbinder B, Chen J, Brunke S, Vazquez-Uribe R, Santhaman R, Meyer AC, de Oliveira Lino FS, Chan KF, Loos D, Imamovic L, et al: Antibiotics create a shift from mutualism to competition in human gut communities with a longer-lasting impact on fungi than bacteria. Microbiome. 8:1332020. View Article : Google Scholar : PubMed/NCBI

41 

Viscoli C, Castagnola E and Machetti M: Antifungal treatment in patients with cancer. J Intern Med Suppl. 740:89–94. 1997. View Article : Google Scholar : PubMed/NCBI

42 

Alam A, Levanduski E, Denz P, Villavicencio HS, Bhatta M, Alhorebi L, Zhang Y, Gomez EC, Morreale B, Senchanthisai S, et al: Fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell. 40:153–167.e11. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Qiu X, Zhang F, Yang X, Wu N, Jiang W, Li X, Li X and Liu Y: Changes in the composition of intestinal fungi and their role in mice with dextran sulfate sodium-induced colitis. Sci Rep. 5:104162015. View Article : Google Scholar : PubMed/NCBI

44 

Malik A, Sharma D, Malireddi RKS, Guy CS, Chang TC, Olsen SR, Neale G, Vogel P and Kanneganti TD: SYK-CARD9 signaling axis promotes gut fungi-mediated inflammasome activation to restrict colitis and colon cancer. Immunity. 49:515–530.e5. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Medzhitov R: Origin and physiological roles of inflammation. Nature. 454:428–435. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Bannenberg GL, Chiang N, Ariel A, Arita M, Tjonahen E, Gotlinger KH, Hong S and Serhan CN: Molecular circuits of resolution: Formation and actions of resolvins and protectins. J Immunol. 174:4345–4355. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Gordon S: Phagocytosis: An immunobiologic process. Immunity. 44:463–475. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Bishehsari F, Engen PA, Preite NZ, Tuncil YE, Naqib A, Shaikh M, Rossi M, Wilber S, Green SJ, Hamaker BR, et al: Dietary fiber treatment corrects the composition of gut microbiota, promotes SCFA production, and suppresses colon carcinogenesis. Genes (Basel). 9:1022018. View Article : Google Scholar : PubMed/NCBI

49 

Singh N, Baby D, Rajguru JP, Patil PB, Thakkannavar SS and Pujari VB: Inflammation and cancer. Ann Afr Med. 18:121–126. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Naylor MS, Stamp GW, Foulkes WD, Eccles D and Balkwill FR: Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression. J Clin Invest. 91:2194–2206. 1993. View Article : Google Scholar : PubMed/NCBI

52 

Haghnegahdar H, Du J, Wang D, Strieter RM, Burdick MD, Nanney LB, Cardwell N, Luan J, Shattuck-Brandt R and Richmond A: The tumorigenic and angiogenic effects of MGSA/GRO proteins in melanoma. J Leukoc Biol. 67:53–62. 2000. View Article : Google Scholar : PubMed/NCBI

53 

Thun MJ, Namboodiri MM, Calle EE, Flanders WD and Heath CW Jr: Aspirin use and risk of fatal cancer. Cancer Res. 53:1322–1327. 1993.PubMed/NCBI

54 

Yang Q, Ouyang J, Pi D, Feng L and Yang J: Malassezia in inflammatory bowel disease: Accomplice of evoking tumorigenesis. Front Immunol. 13:8464692022. View Article : Google Scholar : PubMed/NCBI

55 

Wolf AJ, Limon JJ, Nguyen C, Prince A, Castro A and Underhill DM: Malassezia spp. induce inflammatory cytokines and activate NLRP3 inflammasomes in phagocytes. J Leukoc Biol. 109:161–172. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Zhang YJ, Han Y, Sun YZ, Jiang HH, Liu M, Qi RQ and Gao XH: Extracellular vesicles derived from Malassezia furfur stimulate IL-6 production in keratinocytes as demonstrated in in vitro and in vivo models. J Dermatol Sci. 93:168–175. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Berti M and Vindigni A: Replication stress: Getting back on track. Nat Struct Mol Biol. 23:103–109. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Kawanishi S, Ohnishi S, Ma N, Hiraku Y and Murata M: Crosstalk between DNA damage and inflammation in the multiple steps of carcinogenesis. Int J Mol Sci. 18:18082017. View Article : Google Scholar : PubMed/NCBI

59 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Lin Y, Wang G, Yu J and Sung JJY: Artificial intelligence and metagenomics in intestinal diseases. J Gastroenterol Hepatol. 36:841–847. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Wong SH and Yu J: Gut microbiota in colorectal cancer: Mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 16:690–704. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Perrone G and Gallo A: Aspergillus species and their associated mycotoxins. Methods Mol Biol. 1542:33–49. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Dai Z, Coker OO, Nakatsu G, Wu WKK, Zhao L, Chen Z, Chan FKL, Kristiansen K, Sung JJY, Wong SH and Yu J: Multi-cohort analysis of colorectal cancer metagenome identified altered bacteria across populations and universal bacterial markers. Microbiome. 6:702018. View Article : Google Scholar : PubMed/NCBI

64 

Lin Y, Lau HC, Liu Y, Kang X, Wang Y, Ting NL, Kwong TN, Han J, Liu W, Liu C, et al: Altered mycobiota signatures and enriched pathogenic Aspergillus rambellii are associated with colorectal cancer based on multicohort fecal metagenomic analyses. Gastroenterology. 163:908–921. 2022. View Article : Google Scholar : PubMed/NCBI

65 

Luan C, Xie L, Yang X, Miao H, Lv N, Zhang R, Xiao X, Hu Y, Liu Y, Wu N, et al: Dysbiosis of fungal microbiota in the intestinal mucosa of patients with colorectal adenomas. Sci Rep. 5:79802015. View Article : Google Scholar : PubMed/NCBI

66 

Gao R, Kong C, Li H, Huang L, Qu X, Qin N and Qin H: Dysbiosis signature of mycobiota in colon polyp and colorectal cancer. Eur J Clin Microbiol Infect Dis. 36:2457–2468. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Richard ML, Liguori G, Lamas B, Brandi G, da Costa G, Hoffmann TW, Pierluigi Di Simone M, Calabrese C, Poggioli G, et al: Mucosa-associated microbiota dysbiosis in colitis associated cancer. Gut Microbes. 9:131–142. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Cary JW, Ehrlich KC, Beltz SB, Harris-Coward P and Klich MA: Characterization of the Aspergillus ochraceoroseus aflatoxin/sterigmatocystin biosynthetic gene cluster. Mycologia. 101:352–362. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Frisvad JC, Skouboe P and Samson RA: Taxonomic comparison of three different groups of aflatoxin producers and a new efficient producer of aflatoxin B1, sterigmatocystin and 3-O-methylsterigmatocystin, Aspergillus rambellii sp. nov. Syst Appl Microbiol. 28:442–453. 2005. View Article : Google Scholar : PubMed/NCBI

70 

Navale V, Vamkudoth KR, Ajmera S and Dhuri V: Aspergillus derived mycotoxins in food and the environment: Prevalence, detection, and toxicity. Toxicol Rep. 8:1008–1030. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Uka V, Cary JW, Lebar MD, Puel O, De Saeger S and Diana Di Mavungu J: Chemical repertoire and biosynthetic machinery of the Aspergillus flavus secondary metabolome: A review. Compr Rev Food Sci Food Saf. 19:2797–2842. 2020. View Article : Google Scholar : PubMed/NCBI

72 

McCullough AK and Lloyd RS: Mechanisms underlying aflatoxin-associated mutagenesis-implications in carcinogenesis. DNA Repair (Amst). 77:76–86. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Bianco G, Russo R, Marzocco S, Velotto S, Autore G and Severino L: Modulation of macrophage activity by aflatoxins B1 and B2 and their metabolites aflatoxins M1 and M2. Toxicon. 59:644–650. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Mohammadi A, Mehrzad J, Mahmoudi M and Schneider M: Environmentally relevant level of aflatoxin B1 dysregulates human dendritic cells through signaling on key toll-like receptors. Int J Toxicol. 33:175–186. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Soler AP, Miller RD, Laughlin KV, Carp NZ, Klurfeld DM and Mullin JM: Increased tight junctional permeability is associated with the development of colon cancer. Carcinogenesis. 20:1425–1431. 1999. View Article : Google Scholar : PubMed/NCBI

76 

Martin TA and Jiang WG: Loss of tight junction barrier function and its role in cancer metastasis. Biochim Biophys Acta. 1788:872–891. 2009. View Article : Google Scholar : PubMed/NCBI

77 

Liu NN, Jiao N, Tan JC, Wang Z, Wu D, Wang AJ, Chen J, Tao L, Zhou C, Fang W, et al: Multi-kingdom microbiota analyses identify bacterial-fungal interactions and biomarkers of colorectal cancer across cohorts. Nat Microbiol. 7:238–250. 2022. View Article : Google Scholar : PubMed/NCBI

78 

Gmeiner WH, Hellmann GM and Shen P: Tissue-dependent and -independent gene expression changes in metastatic colon cancer. Oncol Rep. 19:245–251. 2008.PubMed/NCBI

79 

Tjalsma H, Boleij A, Marchesi JR and Dutilh BE: A bacterial driver-passenger model for colorectal cancer: Beyond the usual suspects. Nat Rev Microbiol. 10:575–582. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Wirbel J, Pyl PT, Kartal E, Zych K, Kashani A, Milanese A, Fleck JS, Voigt AY, Palleja A, Ponnudurai R, et al: Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med. 25:679–689. 2019. View Article : Google Scholar : PubMed/NCBI

81 

Thomas AM, Manghi P, Asnicar F, Pasolli E, Armanini F, Zolfo M, Beghini F, Manara S, Karcher N, Pozzi C, et al: Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat Med. 25:667–678. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Zhu F, Willette-Brown J, Song NY, Lomada D, Song Y, Xue L, Gray Z, Zhao Z, Davis SR, Sun Z, et al: Autoreactive T cells and chronic fungal infection drive esophageal carcinogenesis. Cell Host Microbe. 21:478–493.e7. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Azad MAK, Sarker M, Li T and Yin J: Probiotic species in the modulation of gut microbiota: An overview. Biomed Res Int. 2018:94786302018. View Article : Google Scholar : PubMed/NCBI

84 

Mozaffari Namin B, Daryani NE, Mirshafiey A, Yazdi MKS and Dallal MMS: Effect of probiotics on the expression of Barrett's oesophagus biomarkers. J Med Microbiol. 64:348–354. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Rosania R, Minenna MF, Giorgio F, Facciorusso A, De Francesco V, Hassan C, Panella C and Ierardi E: Probiotic multistrain treatment may eradicate Helicobacter pylori from the stomach of dyspeptics: A placebo-controlled pilot study. Inflamm Allergy Drug Targets. 11:244–249. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Zhu R, Chen K, Zheng YY, Zhang HW, Wang JS, Xia YJ, Dai WQ, Wang F, Shen M, Cheng P, et al: Meta-analysis of the efficacy of probiotics in Helicobacter pylori eradication therapy. World J Gastroenterol. 20:18013–18021. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Kumar V, Yadav AN, Verma P, Sangwan P, Saxena A, Kumar K and Singh B: β-Propeller phytases: Diversity, catalytic attributes, current developments and potential biotechnological applications. Int J Biol Macromol. 98:595–609. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Lipke PN and Ovalle R: Cell wall architecture in yeast: New structure and new challenges. J Bacteriol. 180:3735–3740. 1998. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2023
Volume 26 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu W, Li Z, Li X, Cao H, Jiang H, Niu Q and Hu B: Influence of tumor mycobiome on cancer pathogenesis (Review). Oncol Lett 26: 541, 2023
APA
Liu, W., Li, Z., Li, X., Cao, H., Jiang, H., Niu, Q., & Hu, B. (2023). Influence of tumor mycobiome on cancer pathogenesis (Review). Oncology Letters, 26, 541. https://doi.org/10.3892/ol.2023.14128
MLA
Liu, W., Li, Z., Li, X., Cao, H., Jiang, H., Niu, Q., Hu, B."Influence of tumor mycobiome on cancer pathogenesis (Review)". Oncology Letters 26.6 (2023): 541.
Chicago
Liu, W., Li, Z., Li, X., Cao, H., Jiang, H., Niu, Q., Hu, B."Influence of tumor mycobiome on cancer pathogenesis (Review)". Oncology Letters 26, no. 6 (2023): 541. https://doi.org/10.3892/ol.2023.14128