1,189
Views
0
CrossRef citations to date
0
Altmetric
Review

Unveiling the hidden players: exploring the role of gut mycobiome in cancer development and treatment dynamics

, & ORCID Icon
Article: 2328868 | Received 01 Feb 2024, Accepted 06 Mar 2024, Published online: 14 Mar 2024

ABSTRACT

The role of gut fungal species in tumor-related processes remains largely unexplored, with most studies still focusing on fungal infections. This review examines the accumulating evidence suggesting the involvement of commensal and pathogenic fungi in cancer biological process, including oncogenesis, progression, and treatment response. Mechanisms explored include fungal influence on host immunity, secretion of bioactive toxins/metabolites, interaction with bacterial commensals, and migration to other tissues in certain types of cancers. Attempts to utilize fungal molecular signatures for cancer diagnosis and fungal-derived products for treatment are discussed. A few studies highlight fungi’s impact on the responsiveness and sensitivity to chemotherapy, radiotherapy, immunotherapy, and fecal microbiota transplant. Given the limited understanding and techniques in fungal research, the studies on gut fungi are still facing great challenges, despite having great potentials.

This article is part of the following collections:
Gut Microbiota in Cancer Development and Treatment

1. Introduction

While the gut microbiome comprises bacteria, fungi, and viruses, research on diseases has predominantly centered on understanding the interaction between the host and gut bacteria. This emphasis arises from the fact that the intestinal bacteriome constitutes more than 99.9% of the overall gut microbiome.Citation1 Fungi, constituting the remaining 0.1%, however, also play a crucial role in normal and pathological mechanisms, akin to bacteria. Fungi establish early colonization in the intestine during birth, breastfeeding, or through ingestion via food, respiratory tracts, or skin-mouth contact. Food is another significant fungal source. The gut mycobiota is mainly composed of Ascomycota, Basidiomycota, Chytridiomycota, and Zygomycota. Prominent commensal fungi species in the human gut include Candida spp., Saccharomyces spp., and Malassezia spp.Citation2 Normally nonpathogenic, these fungi can become serious threats when the host’s homeostasis is disrupted, particularly in cases of a suppressed immune system or reduced bacterial population. Fungi exhibit morphological adaptability, capable of transitioning between unicellular and multicellular forms, particularly yeast and hyphae, respectively, which influences their pathogenicity. Hyphae typically exhibit higher invasiveness, while yeasts are usually non-intrusive and often linked to mutualistic relationships.Citation3–5 Fungi also demonstrate the ability to generate both harmful and beneficial compounds. Utilizing toxic metabolites and proteases, fungi can switch between various morphological states, aiding in adhesion, host barrier penetration, and subversion of the host immune response. Recent studies affirm that specific gut bacteria possess the ability to trigger tumorigenesis in genetically susceptible murine models.

Certain types of tumor, especially colorectal cancer (CRC), have been extensively studied, with emphasis given to Enterotoxigenic Bacteroides Fragilis, Escherichia coli, and Fusobacterium nucleatum as key components in their pathogenesis.Citation6 Limited research has focused on the tumorigenesis of intestinal fungi. Nevertheless, existing studies suggest a potential connection between diverse cancer types and various fungal species.

2. Common pathogenic mechanisms

Over the decades, studies have been majorly focused on the infections and septicemia caused by gut fungi. However, there are still studies implicating the possible common mechanisms by which gut fungi are able to induce host immune and microbiome dysregulation, and subsequently leading to oncogenesis. These may include the fungi–host recognition and immune regulation, biofilm formation, bacterial–fungal interactions, and toxins and metabolites produced by them ().

Figure 1. The common mechanisms of fungi that are involved in triggering pathogenesis, especially in oncogenesis. PRRs, pattern recognition receptors; CLRs, C-type lectin receptors; CARD9, Caspase Recruitment Domain-containing protein9; STAT1, signal transducer and activator of transcription 1; Th17, T helper 17 cell; IL-6, interleukin 6; IL-23, interleukin 23; TNF-α, tumor necrosis factor α; IL-12, interleukin 12; IFN-γ, interferon γ; IL-1β, interleukin 1β; MDSCs, myeloid-derived suppressor cells; TCL, cytotoxic T cell; ACH, acetaldehyde; MAPK, mitogen-activated protein kinases; EGFR, epithelial growth factor receptor; AhR, aryl hydrocarbon receptor; Hsp27, heat shock protein 27; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor.

Figure 1. The common mechanisms of fungi that are involved in triggering pathogenesis, especially in oncogenesis. PRRs, pattern recognition receptors; CLRs, C-type lectin receptors; CARD9, Caspase Recruitment Domain-containing protein9; STAT1, signal transducer and activator of transcription 1; Th17, T helper 17 cell; IL-6, interleukin 6; IL-23, interleukin 23; TNF-α, tumor necrosis factor α; IL-12, interleukin 12; IFN-γ, interferon γ; IL-1β, interleukin 1β; MDSCs, myeloid-derived suppressor cells; TCL, cytotoxic T cell; ACH, acetaldehyde; MAPK, mitogen-activated protein kinases; EGFR, epithelial growth factor receptor; AhR, aryl hydrocarbon receptor; Hsp27, heat shock protein 27; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor.

2.1. Fungi-host recognition and inflammatory signaling pathway

C-type lectin receptors (CLRs), typically Dectin-1, Dectin-2, Dectin-3, and Mincle, are subtypes of pattern recognition receptors (PRRs), primarily expressed on myeloid cell surfaces including surfaces of macrophages, neutrophils, and dendritic cells. These CLRs detect fungal cell wall components like β-glucans and α-mannans, engage in sensing fungal infections and activating downstream signaling pathways, triggering inflammation and immune responses.Citation7 This ligand-receptor binding activates multiple intracellular signaling pathways, including adaptor molecules, kinases, and transcription factors. The Caspase Recruitment Domain-containing protein9 (CARD9) is an essential adaptor protein restrictedly expressed in myeloid cells and is critical in initiating inflammatory caspase in response to Candida, Malassezia, and Aspergillus. CLR requires CARD9 for subsequent innate immunity activation.Citation8–11

Dectin-1 signaling triggers the segmentation of pro-IL-1β, resulting in the ultimate production of IL-1β through activation of NLRP3 and caspase-1 or caspase-8 inflammasome pathways.Citation12–14 The NLRP3 inflammasome is potentiated by diverse activators and recognizing patterns, including damage-associated molecular patterns (DAMP), and pathogen-associated molecular patterns (PAMP), which are targets of fungi, bacteria, and viruses.Citation15 Subsequently, downstream immune response is triggered by these activators in the different circumstances, including fungal invasions and infections.

NLRP3 inflammasome activation is found to be associated with two stepwise signals. Signal 1 initiates the immune recognition response such as recognition by Dectin-1 mentioned above, followed by activation of nuclear factor-κB (NF-κB), driving the production of pro-IL-1β. The signal 2 involves subsequent activation of caspase-1, resulting in the maturation of IL-1β.Citation16,Citation17

Dectin-3, also known as CLED4D or CLECSF8, operates as a PRR in various myeloid cells. It recognizes multiple fungi, including C. albicans and Cryptococcus, by detecting α-mannans on their surfaces. A prior study conducted by Zhu et al. revealed that Dectin-3 forms a heterodimeric complex with Dectin-2 to detect α-mannans.Citation18

Recognition of fungi such as C. albicans activates pathways like MAPK, STAT1, STAT3, NF-κB, and IRF3, leading to the production of mediators such as interleukins (IL-1, IL-6, IL-12, and IL-23), tumor necrosis factor-α (TNF-α), and interferon gamma (IFN-γ).Citation19–22 These responses contribute to both innate and adaptive immunity.Citation23 The inflammatory activation pathways and interferons further promote the regulation and alteration of the host immune response, playing a crucial role in the development of both malignant and benign conditions. Malassezia can elicit cooperative recognition by Mincle and Dectin-2 through distinct ligands that cause skin inflammation.Citation24

The CLRs, accompanied by the subsequent downstream activation of variable signaling pathways, actively participate in the regulation of immune responses, and thus affecting disease progression.

2.2. Immune cell activation

Myeloid-derived suppressor cells (MDSCs), composed of granulocytic MDSCs (G-MDSCs) and monocytic MDSCs (GM-MDSCs), constituting a heterogeneous group of immature myeloid cells. Their role involves promoting immune suppression and aiding in tumor development. Fungal dysbiosis leads to an increase in the release of MDSCs, thus contributing to tumorigenesis. In CRC patients, a positive correlation occurs between the abundance of MDSCs at tumor sites and the fungal burden. Several transcriptional factors, including STAT3 and IRF8, have been implicated in the expansion and differentiation of MDSC.Citation25–29

Th17 cells, an antigen-specific subset of CD4+ T cells, contribute to coordinating immune responses against extracellular bacteria and fungi. Th17 cells, initially activated by C. albicans, broaden their response to target other fungi through cross-reactivity. Intestinal inflammation expands C. albicans-specific and cross-reactive Th17 cells.Citation30 Dectin-SyK-CARD9 signaling pathways can stimulate the secretion of interferons and cytokines, as well as the differentiation of T lymphocytes into IL-17-producing CD4+ T cells.Citation31,Citation32 Pro-inflammatory cytokine IL-17 secreted by Th17 cells is regarded as a contributor to multiple autoimmune diseases including autoimmune thyroid disease,Citation33 atopic dermatitis,Citation34 IBD,Citation35 etc. Studies have explored its role in tumor development in mice, revealing that indole’s carcinogenic effect involves interfering the Th17 cell differentiation and IL-17 production under AhR regulation.Citation36 Additionally, IL-23, a cytokine crucial for expanding and sustaining Th17 cells, is vital for inducing T-cell mediated colitis in murine models.Citation37 Moreover, it has been observed to foster angiogenesis and contribute to tumor growth.Citation37–39

MDSCs and Th17 cells significantly promote immune suppression and tumor development. Fungal dysbiosis induces increased MDSC release and Th17 differentiation, contributing to tumor progression. Th17 cells, initially activated by C. albicans, amplified their response to target fungi and contributes to inflammation. The stimulation of stimulate Th17 differentiation and IL-17 production is assisted by the Dectin-SyK-CARD9 pathways, as implicated in autoimmune diseases and tumor development.

2.3. Bacterial–fungal interactions

Bacteria and fungi can interact synergistically, mutually, or antagonistically, shaping the microbiome’s unique features in different human body parts and health states. Bacterial species are often found accompanied with fungi in the formation of polymicrobial biofilms, such as Streptococcus mutans, P. aeruginosa, and S. aureus .Citation40 Infections originating from biofilms that involve both bacteria and fungi pose a significantly greater challenge for treatment compared to those involving a single species. Addressing these infections requires intricate multi-drug treatment approaches.Citation41 The host-bacterial-fungal interactions can affect the survival, morphogenesis, and invasiveness of fungi.Citation42 Bacteria can inhibit the growth of C. albicans through the secretion of molecules, metabolites, and factors that function directly on fungi or indirectly through host response.Citation43–46 For example, in murine models, Bacteroides thetaiotaomicron upregulates the transcription factor HIF-1α and anti-microbial peptide LL-37, initiating host innate immune response to inhibit C. albicans colonization.Citation44 Notably, it is also said that B. thetaiotaomicron-processed mucins enhance C. albicans growth more than unprocessed mucins, and thus shaping its spatial distribution.Citation47 Short-chain fatty acids (SCFAs), produced by bacterial metabolization and fermentation of non-digestible dietary fibers, typically Lactobacillus, are found to hinder hyphal morphogenesis and promote T cell response to C. albicans in mice, and finally inhibit biofilm formation and prevent invasion.Citation45,Citation48

Bacteria can exploit the microenvironment established by fungi for their growth. The hypoxic microenvironment generated by C. albicans in its biofilm is conducive to the growth of anaerobic bacteria such as Clostridium perfringens and Bacteroides fragilis .Citation40 B. thetaiotaomicron is known to possess enzymes specialized in breaking down α-mannan, a typical cell wall component of yeasts.Citation49 The growth of anaerobic Bacteroides species such as B. vulgatus and B. fragilis are also benefited from metabolizing fungal components in vitro.Citation50 Several bacteria, including Streptococcal species, Fusobacterium nucleatum, and Helicobacter pylori, have been documented to exhibit synergistic relationships with C. albicans, amplifying their virulence.Citation51

A study revealed that C. albicans influence the recolonization of cecal microbiota in mice treated with antibiotics. This resulted in an elevation of Enterococcus faecalis and a decrease in probiotic Lactobacillus strains.Citation52 A subsequent investigation demonstrated that intestinal C. albicans in antibiotic-treated mice elicit altered expression of specific genes regarding inflammation and host response, resulting in a shift in bacterial diversity while not inducing gut inflammation, supporting the role of C. albicans in helping with the recovery of bacterial community.Citation53

The dysbiosis of either of bacteria or fungi can cause the dysbiosis of the other. The administration of antibiotics can induce fungal dysbiosis and the overgrowth of C. albicans .Citation54 Jiang et al. demonstrated that diverse fungal species can effectively substitute for enteric bacteria in addressing infectious and inflammatory disorders, including colitis resulting from the elimination of commensal bacteria.Citation55 Van Tilburg Bernardes et al. illustrated that fungal colonization leads to significant alterations in the ecology of the bacterial microbiome. Furthermore, the co-colonization of bacteria and fungi is associated with an elevated level of colonic inflammation.Citation56–58

In all, bacteria and fungi interact in various ways, forming polymicrobial biofilms and influencing each other’s growth and virulence. Bacteria inhibit C. albicans through diverse mechanisms, while fungal biofilms can create favorable environments for anaerobic bacteria. Dysbiosis in one can disrupt the other, with fungi potentially substituting for bacteria in addressing inflammatory disorders. This interplay significantly alters microbiome ecology and can impact the course and severity of pathogenic status such as colonic inflammation.

2.4. Invasiveness and biofilm formation

Biofilms are closely packed communities of cells that attach to biotic and abiotic surfaces, including the intestine.Citation59,Citation60 C. albicans biofilms exhibit a highly organized structure, incorporating yeast-forming cells, pseudohyphae, and hyphae enveloped by an extracellular matrix. These biofilms participate in the survival of C. albicans. C. albicans biofilms are typically formed following three stages: yeast cell adsorption and adherence, hyphae development with microcolony formation and extracellular matrix (ECM) production, and maturation with cell dispersal for potential dissemination to secondary infection sites.Citation61,Citation62 The formation process may be influenced by several genes: TEC1, BCR1, ALS3, HWP1, and EFH1 .Citation63–68 The ECM is composed of self-produced extracellular biopolymers within the biofilm. Macromolecules, such as protein, lipid, and various polysaccharides including α-mannans and β-1,6-glucans,Citation69 as well as plentiful of host cells such as neutrophils exist, indicating host engagement in the formation and maintenance of these biofilms.Citation70

Quorum sensing factor farnesol, initially identified in high-density cultures, serves as a crucial inhibitor of hyphal formation and participates in the dispersal of biofilm cells of C. albicans .Citation71 Farnesol hinders the maturation of dendritic cells, and thus causing failure in inducing proper T cell response and innate immunity against C. albicans .Citation72 Candida biofilms offer enhanced protection against the immune system and antifungal therapy compared to planktonic cells. Factors like hyphal morphology, ECM, farnesol, resistance-associated gene expression, fungal-bacteria interactions, persister cells, and biofilm extracellular vesicles all contribute to this mechanism.Citation61,Citation73,Citation74

C. albicans possess a strong propensity for invasion through the epithelial cells in their hyphae form, an important factor in their pathogenesis.Citation75,Citation76 Enzymes like enolase and phytase possess the ability to anchor to the membrane, contribute to hyphal growth, host fibronectin, and laminin binding, epithelial tissue damage, and nutrient acquisition.Citation77,Citation78 Although much have been concerned about Candida biofilm and infection, the relationship between fungal biofilm and carcinogenesis has seldomly been reported, except that in oral cancer.Citation79 The specific features of biofilm, especially its resistance to host immune system, and strong capacity of invasion and dispersion may play a role in the tumorigenesis process.

Fungal species other than C. albicans are also capable of forming biofilms and causing biofilm-related infection. Some Malassezia species such as Malassezia pachydermatis and Malassezia furfur are also found to be able to form biofilms in vitro, and these biofilms also possess the ability of pathogenesis and drug resistance.Citation80,Citation81 Aspergillus ,Citation82 Cryptococcus, Citation83 and many other fungi also possess the ability to form biofilms and act differently on the pathological process.

In other words, biofilms are crucial for the survival of fungi, especially C. albicans, exhibiting structured layers composed of yeast, pseudohyphae, and hyphae within an extracellular matrix. Factors including farnesol, ECM, and fungal-bacteria interactions enhance C. albicans biofilm resistance against immunity and antifungal therapy. The invasiveness of the hyphae form of C. albicans contributes to its pathogenesis. While rarely reported, fungal biofilms may impact carcinogenesis, exemplified in oral cancer.

2.5. Toxins and metabolites

2.5.1. Candidalysin

Candidalysin is a cytolytic toxin produced by C. albicans that enables its capacity to damage and evade through host epithelium, and is encoded by the hypha-specific ECE1 gene.Citation84 Candidalysin activates the MAPK pathway, triggering host immune responses such as heat shock protein (Hsp) activation, IL-6 release, and neutrophil recruitment.Citation85 However, the detailed signaling caspase involved is still unknown. A recent study highlighted the significance of EGFR activation in candidalysin-induced MAPK signaling. This activation leads to the induction of the c-Fos transcription factor, ultimately triggering neutrophil recruitment through G-CSF and GM-CSF. The process relies on the release of EGFR ligands and calcium influx.Citation86 While S. A. Nikou et al. mentioned another mechanism of candidalysin in oral epithelial cells that involves the activation of EGFR and c-Fos independent of MAPK.Citation87 Rogiers et al. revealed the pivotal role of candidalysin in eliciting C. albicans to induce NLRP3 inflammasome responses, leading to the maturation and secretion of IL-1β in macrophages.Citation88 Additionally, C. albicans mutants that are unable to transit from yeast to hyphae exhibited compromised TCRαβ+ cell proliferation and diminished IL-17a expression in oral epithelial cells. This suggests a potential role of candidalysin in engaging in the recognition and induction of innate immunity of fungi as mentioned above that involves IL-1β and IL-17.Citation89

2.5.2. Acetaldehyde

C. albicans, as along with some Candia species other than albicans such as C. glabrata and C. tropicalis, are able to produce acetaldehyde (ACH) in vitro.Citation90 ACH is a genotoxic compound involved in ethanol metabolism, disrupting DNA repair processes and inducing oxidative stress. Dysbiosis can increase local ACH production by opportunistic pathogens, forming a causal link with oral and gastrointestinal carcinogenesis. High ethanol-derived ACH-producing Candida were more prevalent in oral cancer than non-oral cancer patients.Citation79

2.5.3. Indole

Indoles are alkalic molecules transformed by tryptophan, including indirubin, indolo (3,2-b) carbazole, and FICZ. They can be produced by Malassezia metabolism.Citation91,Citation92 Indole compounds are robust ligands and agonists of the aryl hydrogen receptor (AhR), and they are associated with skin carcinogenesis.Citation93 Abundant AhR, malassezin, and indoles can be observed in M. furfur yeast strains causing skin infections, and is related to disrupted TLR-associated dendritic cell maturation.Citation92 Kynurenine, a key oncometabolite in the tryptophan pathway linked to numerous diseases, serves as a ligand activating AhR in cells. Blocking kynurenine has proven to inhibit the growth of CRC cells in colon organoids.Citation94

Mycotoxins such as aflatoxin patulin and Xaralenone, which are produced by exogenous fungi are also associated with carcinogenesis, despite largely unexplored mechanisms.Citation95–97 Aspergillus species are able to produce aflatoxin that is a well-studied carcinogenic mycotoxin for hepatocellular carcinoma (HCC) development, and we will mention it in detail later.

3. Specific fungi in oncogenesis

Similar to bacteria, specific gut fungal genus, and species display an augmentation in the intestine of specific tumor patients, and they may contribute to tumor progression through different mechanisms (). They are suspected to be a cause even if the tumor does not originate along the digestive tract, and the possibility of migration helps to explain this phenomenon.

Figure 2. Fungi and their roles in specific tumors. Shown are the various mechanisms through which major gut fungi species are able to influence the specific cancers, including the immune responses, cytotoxins and metabolites, and bacterial–fungal interactions

Figure 2. Fungi and their roles in specific tumors. Shown are the various mechanisms through which major gut fungi species are able to influence the specific cancers, including the immune responses, cytotoxins and metabolites, and bacterial–fungal interactions

3.1. Candida

Candida is commonly manifested as a commensal component of the human gastrointestinal tract. However, it can become pathogenic due to various host-related factors such as gut microbiome disruption, immunocompromised status, and significant metabolic changes. While candidemia is the most direct manifestation, Candida is also associated with multiple types of cancer. Its role in oral cancer is the most extensively explored,Citation98 with reported but inconclusive evidence of its involvement in other cancers including colorectal cancer, neck and cervical cancer.

3.1.1. Candida Albicans

  • C. albicans, in normal health status, presents as harmless native commensal microbiota colonizing mucosa at multiple sites, including mouth, skin, the gastrointestinal tract, vagina, and vascular system.Citation64,Citation65,Citation68 However, it also acts as an opportunistic pathogen capable of causing severe and potentially fatal bloodstream infections, particularly in both immunocompromised and immunocompetent individuals. C. albicans can hematogenously disseminate, reaching multiple organs and potentially leading to serious complications. C. albicans displays significantly increased pseudohyphae morphogenesis, virulence factor secretion, and biofilm formation compared to certain non-albicans Candida species such as C. auris, C. tropicalis, C. parapsilosis, C. haemulonii, and C. glabrata, observed in vitro studies and in murine models.Citation99,Citation100

  • C. albicans can potentially drive cancer progression through multiple mechanisms, such as the generation of carcinogenic byproducts, initiation of chronic inflammation, alteration of the immune microenvironment, activation of oncogenic signaling pathways, and interactions with bacteria. Here, we summarize the possible relationship between cancers and C. albicans in or derived from the gut.

3.1.1.1. Esophageal cancer

Esophageal cancer (EC) has been found to be associated with Candida infection, of which C. albicans are the most relevant.Citation101 It is more prevalent in patients with EC than in those with esophagitis (27% compared to 15%), Candida infection may be correlated to the development of EC.Citation102 On average, patients with fungal infections are 7 years younger than those with EC, suggesting fungi as potential triggers for EC initiation and development.Citation103

Chronic esophageal candidiasis is primarily observed in immunocompromised conditions, including chronic mucocutaneous candidiasis (CMC), which is a common clinical symptom of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). APECED patients demonstrate an impairment of negative selection of autoreactive T cells resulting in autoimmunity, rendering them vulnerable to chronic bacterial and fungal infections and esophageal squamous cell carcinoma (ESCC), as evidenced by multiple case reports.Citation104–106 Patients with C. albicans may experience differing degrees of T-lymphocyte dysfunction, potentially increasing their susceptibility to esophageal cancer, proven by the fact that fungal depletion and administration, respectively, inhibits and accelerates ESCC development.Citation107 Delsing and Koo suggested a relationship between STAT1 function deficiency, autoreactive CD4+ T cells, and chronic fungal infection. Koo et al. documented a family with CMC and a confirmed gain-of-function STAT1 mutation. The patients, who experienced CMC in childhood, presented severe oral and esophageal candidiasis and were at risk of developing oral cancer and/or ESCC.Citation106 Delsing et al. expressed concern about nitrosamine production, and defective T-lymphocyte function against oncogenesis along with the STAT1 mutation in the development of esophageal carcinoma.Citation105 Another potential mechanism could be the local production of ACH by C. albicans, given the significant microbial composition similarity between the esophageal mucosa and the oral cavity.Citation108,Citation109

3.1.1.2. Gastric cancer

H. pylori has long been acknowledged as the most important risk factor for gastric cancer (GC). However, whether other microbials take their part in the development of GC remains largely unexplored.

In 2014, Sano et al. showed that infection caused by orally or intravenously administrated C. albicans leads to chronic hyperplastic candidiasis and ultimately induces oncogenic processes in the stomach in diabetic rat models.Citation110 Zhong et al. later substantiated the hypothesis by revealing the notable fungal dysbiosis in the intratumor biopsy samples, characterized by a significant increase of C. albicans abundance and a minor increase in other species such as Fusicolla acetilerea and Arcopilus aureus .Citation111 This enrichment of C. albicans is also shown in a pan-cancer analysis.Citation112 Furthermore, a case report mentions the possibility that genetic mutations and C. albicans infection corporately induce GC development, both leading to defective production of IL-17.Citation113 Intravacuolar H. pylori, found within Candida cytosols in mice, indicates endosymbiosis function of C. albicans. This protective environment within C. albicans vacuoles may participate in promoting the virulence and survival of H. pylori, potentially contributing to tumor development.Citation114

Contrarily, Zhang et al.‘s in silico study found no difference in Candida species abundance between tumor lesions and surrounding normal tissues in 61 GC patients. Instead, a significant enrichment in the Solicoccozyma was observed.Citation115 Furthermore, a prospective cohort study conducted by Ndegwa et al. revealed no associated risk of Candida-related oral lesions with the development of GC.Citation116

3.1.1.3. Colorectal cancer

Until now, there have been no consistent perspectives on the role of C. albicans in CRC development. Patients with inflammatory bowel disease are always regarded as a high-risk population of CRC. Prior studies on inflammatory bowel disease have identified fungal community dysbiosis, manifested with an elevated abundance of intestinal Candida species and Malassezia species.Citation117–119 There are studies showing no increase in albicans abundance neither in CRC nor in adenoma patients in their intratumor biopsy samples.Citation120,Citation121 Gao et al. even reported the considerable decrease in the number of Candida species in CRC.Citation120 Furthermore, Jin et al.'s research revealed that, despite the fact that high-fat diet is associated with dysbiosis and alteration of multiple bacterial and fungal species, the abundance of gut Candida was reduced other than increased in mice that developed CRC.Citation122

Anti-fungi immunity, especially for C. albicans, sensed via Dectin-Syk-CARD9 pathway, is found to be a protective factor for colitis-associated cancer (CAC). In Card9−/− mice, fungal dysbiosis was observed, leading to increased STAT3 activation-an essential transcription factor for MDSCs and a significant driver for CRC tumorigenesis.Citation26,Citation123 In Zhu’s study, the elevated C. albicans load in Dectin-3−/− mice load triggers glycolysis in macrophages and IL-7 secretion. The IL-22 production followed by IL-7 production in innate lymphoid cells are implemented through the AHR and STAT3 experiments.Citation124 Loss of Dectin-3 also leads to decreased inflammasomes and IL-18 activation, leading to impaired CD8+ T cell function, decreasing epithelial barrier restitution and IFN-γ production in mice.Citation125

Although distinct relationship has not been discovered, bacterial–fungal interactions can be highly suspected in the tumorigenesis process. Sovran et al. proposed a bacterial–fungal interaction affecting DSS-induced colitis in mice. Antibiotic treatment altered susceptibility and the impact of fungi on colitis, with C. albicans exacerbating colitis in untreated settings.Citation57

3.1.1.4. Liver cancer

Recently, the possible link between liver disease and C. albicans has been taken into consideration, and mechanisms explored include translocation of β-glucan,Citation126 Th17 cells,Citation127 and candidalysinCitation128 derived from or secreted by intestinal albicans, as well as the activation of Dectin-1 in Kupffer cells, and subsequent increase in IL-1β expression.Citation129 Nevertheless, still little is known about whether gut fungi count in HCC, although some bacterial species are found to be correlated.Citation130

The gut mycobiome showed decreased diversity in HCC patients among various studies.Citation131,Citation132 Despite this, C. albicans has undergone an enrichment in the gut of HCC patients. In tumor-bearing mice, C. albicans was observed to promote HCC by reprogramming NLRP6-dependent cancer cell metabolism.Citation131 C. albicans was found to be significantly more abundant in late stage HCC.Citation132 Usami et al. suggested a likelihood between intestinal Candida and serum fatty acid metabolism in HCC. They found strong positive correlations (coefficients 0.81 and 0.88) between serum EPA (eicosapentaenoic acid), EPA/AA (arachidonic acid) ratio, and fecal Candida in the liver cirrhosis (LC) group of 46 HCC patients.Citation133

As a commensal microbe, C. albicans exhibits higher virulence compared to other Candida species, contributing to cancer progression via carcinogenic byproducts, chronic inflammation, and altered immune microenvironment. In esophageal cancer, C. albicans infection correlates with younger onset and autoimmune conditions; gastric cancer may be influenced by fungal dysbiosis and the protective effect of C. albicans on H. pylori, potentially promoting oncogenesis; in colorectal cancer, the role of C. albicans are most studied with the involvement of Dectin-Syk-CARD9 pathway, with bacterial–fungal interactions suspected; liver cancer studies suggest C. albicans enrichment in HCC patients, impacting metabolism and disease progression. Also, C. albicans colonized in the mucosa other than the gut can influence site-specific tumors with various mechanisms. C. albicans in the oral cavity is related to oral pre-cancer lesions and can directly induce oral cancer through IL-17 activation, biofilm formation, oncogenic metabolites, and mycotoxin production, such as candidalysin, nitrosamine, and ACH.Citation76,Citation134 Interestingly, oral albicans have been found to be correlated with blood malignancy in several cohorts.Citation135,Citation136

3.1.2. C. tropicalis

C. tropicalis was found to be closely related to several stages in the pathological formation of colon diseases, including ulcerative colitis, formation of adenoma, oncogenesis process of CAC, and resistance to chemotherapy.Citation121,Citation137–139 Until now, the majority of the observed mechanical processes are associated with Dectin-Syk-CARD9 pathway. C. tropicalis is specifically increased in Dectin-3−/− and Card9−/− CAC mice, and antifungal treatment ameliorated CAC in the previous study.Citation26,Citation125 Deficiency of Dectin-3 impairs phagocytic and fungicidal abilities of macrophages in C. tropicalis and thus promotes colitis.Citation125 The increase of C. tropicalis is demonstrated to enhance the accumulation of MDSCs in Card9−/− mice.Citation26 However, further research indicated that C. tropicalis promotes tumorigenesis through influencing aerobic glycolysis by the Dectin-3-Syk-PKM2-HIF-1α glycolysis signaling axis, which involves MDSCs.Citation138 Toxic products such as NADPH oxidase 2 and reactive oxygen species (ROS) are produced and lead to T cell dysregulation.Citation138 Their further investigation revealed that the C. tropicalis facilitates the NLRP3 inflammasome production through the Dectin-3-glycolysis pathway, and finally leads to the increase in IL-1β production on MDSCs. The wholistic process involves activation of several transcription factors and signaling molecules, including STAT1, pro-caspase-1, mitochondrial ROS, and finally inducing IL-1β production. The elevation of these participants to immune pathways may suggest another target for CRC treatment.Citation17 A new potential mechanism is asserted by Qu et al. that C. tropicalis can interfere with tumor PD-1 expression by increasing tumor cell autophagy and thus promote CRC progression.Citation140

3.2. Malassezia

To our recent knowledge, Malassezia is composed of 18 species, most of which colonize the skin, but can also colonize other mucosal surfaces such as human gut, as well as abiotic surface that form biofilms.Citation141 Malassezia genus has been reported to be the second most abundant genus among all human stools in proportions from 2 to 4%, while M. restricta (more than 80%) and M. globosa (36%) account for the most prevalent species.Citation142,Citation143

3.2.1. CRC and liver cancer

While Malassezia has been extensively studied as an important contributor to the tumorigenesis of skin and breast cancer due to its skin colonization,Citation144 there is relatively limited research on its impact on cancers resulting from gut colonization.

An increased abundance of Malassezia spp. and fungal dysbiosis was found in IBD patients,Citation117 as well as in CRC patients.Citation145,Citation146 Interestingly, one study also mentioned an increased abundance of Malassezia species in HCC patients compared with cirrhosis and healthy controls.Citation132

Unfortunately, few studies have been dedicated to the underlying mechanisms of gut Malassezia in gastrointestinal oncogenesis so far, and mountainous research needs to be done to clarify the exact relationship. A possible mechanism may lie in the metabolism of tryptophan into indole that is associated with kynurenine production.Citation94

3.2.2. Pancreatic cancer

While there is extensive discussion on the role of bacteria, such as Fusobacterium sp., Enterobacteriaceae, and H. pylori, in pancreatic cancer tumorigenesis, limited efforts have been paid on the involvement of fungi in this oncogenic process.Citation147 Research on tumorigenesis in KC mice (p48cre;LSL-KrasG12D), a model expressing oncogenic Kras in pancreatic progenitor cells for slow progressive pancreatic ductal adenocarcinoma (PDA), demonstrated significant fungal dysbiosis in the pancreas, with Malassezia spp. being the predominant genus.Citation148 Unlike the gut or normal pancreas, both mouse and human PDA mycobiomes featured increased Malassezia species. Notably, Malassezia in the pancreas tissue is shown to have migrated from the gut.Citation148

Dectin-1 is not normally expressed in human pancreatic cells.Citation149 However, it has been identified in association with inflammation caused by Malassezia folliculitis .Citation14 Dectin-1 functions, particularly in fungal infections, by initiating the Syk-CARD9-NFκB pathway and activating the downstream caspase-8-ASC complex, ultimately leading to the production of IL-1β.Citation12,Citation150,Citation151 Alam et al. discovered that this Dectin-1 pathway contributes to the pathogenic pathway of Malassezia through activation of cytokines IL-33 from PDA cells, a downstream target of KrasG12D in tumor-bearing mice. Synergistically, biochemical factors like ROS can stimulate and amplify IL-33 secretion in collaboration with the mycobiome.Citation149 This IL-33 secretion may participate in triggering a type 2 immune response, hastening the pancreatic cancer progression along with reducing survival.Citation152

Aykut et al. proposed that Kras oncogenic murine models exhibit fungal dysbiosis, exacerbating tumor progression through the activation of the C3 complement cascade via recognition and ligation with mannose on cell walls of Malassezia through the mannose-binding lectin. The complement system then participates in the corruption of body immunity, expansion, and apoptosis of tumor cells and therapeutic resistance.Citation147,Citation148

In a comparative cohort study examining 16 pancreatic cancer patients who achieved long-term survival (at least 4 years) post-pancreatectomy without recurrence, with eight patients after pancreatectomy and chemical therapy as the control group, we found no significant difference in Malassezia abundance in their fecal samples, indicating that Malassezia may not influence the prognosis.Citation153

Increased Malassezia spp. abundance has been observed in IBD, CRC, and HCC patients, suggesting its potential involvement in tumorigenesis. Mechanisms may involve tryptophan metabolism and IL-33-mediated inflammation through the complement cascade activation. However, the clinical importance of Malassezia has not been proven yet, awaiting further studies.

3.3. Aspergillus

3.3.1. Colorectal cancer

An in silico multi-cohort analysis revealed that A. rambellii and other five fungal species were enriched in CRC patients compared with patients with adenoma or normal controls. The enrichment of A. rambellii was validated both in vivo and in vitro, with another interesting finding that interactions occur among the enriched microbes. The enriched fungi showed significant co-current features, and the A. rambellii also exhibited a correlation with Fusobacterium nucleatum and P. micra (z score −5.95 and −5.07 respectively).Citation154 A. rambellii administration promoted tumor growth, weight, and proliferation both in vitro and in a xenograft mice model. They further announced that the panel combing both fungi and bacteria better distinguishes CRC patients from health controls and adenoma patients than pure fungal or bacterial panels.Citation154 A. rambellii and other two species were among the top 3 fungal species that were significantly enriched in CRC according to Gao et al. Other Aspergillus species, that is A. ochraceoroseus and A. flavus, were also found to have increased in abundance in CRC.Citation120

3.3.2. Liver cancer

Aflatoxins have been one of the earliest discovered and studied mycotoxins worldwide, with countless studies reporting their potential pathogenic and carcinogenic effects majorly focusing on two species: A. flavus and A. parasiticus .Citation132 The majority of aflatoxins contributing to liver cirrhosis and HCC are of exogenous origin, particularly in developing countries. Aflatoxins are most commonly absorbed through ingestion and rapidly cleared from blood. AFB1 has been experimentally shown to possess the capability to damage intestinal epithelial cells, leading to the disruption of intestinal structure.Citation155 This leads to an influx of immune cells, which may exacerbate tissue damage and impair the absorption of nutrients, particularly vitamins A and E.Citation156 Upon absorption, AFB1 is mainly distributed in the liver, while lesser amounts are found disseminating into kidneys and mesenteric vein. Metabolized by the CYP450 system, AFB1 forms DNA adducts, which is its major mechanism for tumorigenesis. AFB1 also induces ROS and inhibits protein, RNA, and DNA synthesis, ultimately leading to apoptosis in microglia via NF-kB activation in oxidative stress.Citation96

A. rambellii, A. flavus and A. parasiticus exhibit enrichment in tumors, indicating the potential role of Aspergillus in tumor development. AFB1 produced by A. flavus and A. parasiticus significantly contributes to HCC development by damaging intestinal epithelial cells, impairing nutrient absorption, and inducing oxidative stress, leading to apoptosis and tumorigenesis.

3.4. Other fungi

Some fungi species other than those mentioned above are reported to show differences in abundance in cancer patients, despite the fact that they only take up a little account of the gut fungal composition. Chang et al. mentioned that apart from C. albicans, Torulopsis glabrata, Torulopsis tomata, C. tropicalis, C. krusei, and C. parepsilosishave have also been isolated from esophageal samples.Citation103 Considering GI tumors with intratumor lesions, C. albicans, C. tropicalis, C. dubliniensis, and other Candida species could all be distinguished with an increase in abundance, with interactions with certain bacterial and fungal species, such as Lactobacillus gasseri and S. cerevisiae .Citation112

Multiple fungi species, although not directly implicated in tumor progression, are potentially associated with exacerbating diseases that have a high likelihood of progressing into cancer. Phoma, another opportunistic pathogen, is found to cause lung massCitation36 and subcutaneous mycosis.Citation121 C. metapsilosis M2006B demonstrated the ability to alleviate experimental colitis in a large-scale human intestinal fungal cultivation and functional analysis.Citation117

4. Fungi as biomarkers

Several studies have demonstrated the possibility of utilizing fungi as biomarkers for tumor screening and diagnosis due to the significant difference in fungal composition compared with the healthy control.Citation146,Citation157

An important parameter is the Basidiomycota: Ascomycota ratio (B/A ratio), a significant increase in B/A ratio is found concerning CRC and ulcerative colitis,Citation146,Citation158 although this can be controversial among different studies.Citation145 Models incorporating diverse microbiomes have been proposed for predicting diseases like colon adenoma and CRC. They exhibit varying compositions, resulting in differences in accuracy that can be estimated by the area under the receiver-operating characteristic curve (AUROC). However, there are studies showing no difference in fungal composition in CAC.Citation159 Decreased diversity of fungi indicating fungal dysbiosis is found in polyp patients, supporting the role of fungi in chronic tumorigenesis process.Citation121,Citation145 Luan observed a correlation between malignancy potential of adenoma, and disease stage with changes in fungal composition, highlighting specific fungal species composition at each stage.Citation121 Gao et al. reported a non-significant increase in M. restricta, with Leucoagaricus_sp_SymCcos and fungal_sp_ARF18 undergoing depletion in adenomas.Citation120 In the study conducted by Cocker et al., 14 fungal biomarkers achieved an AUROC of 0.93 that effectively distinguished CRC from controls. However, these fungal markers only achieved AUROCs of 0.63 in classifying adenoma subjects from controls.Citation146 Another study discovered that 16 multi-domain markers including 11 bacterial, four fungal, and one archaeal feature achieved good performance in diagnosing patients with early-stage CRC with an AUROC of 0.78.Citation157 Gao et al. regarded Taxomyces andreanae, A. rambellii, Lachancea_waltii, fungal_sp_ARF18, and Phanerochaete chrysosporium as the top five most important biomarkers with the AUROC of 0.926.Citation154

Some other cancers were also evaluated for the potential of utilizing gut fungi as tumor biomarkers, including GC and HCC.Citation111,Citation115,Citation132

Numerous studies have agreed on the potential that fungal-bacterial interplay influences the tumor development. Cocker et al. announced a significant increase in amount and detrimentality, but also more antagonistic bacterial–fungal interplay in healthy patients compared with CRC patients (p < .00001), very similar with the discovery by Liu et al., as well as our previous research, demonstrates that an addition of bacterial biomarkers to the fungal marker can further increase the AUROC of our random forest model in predicting the effect of immune checkpoint inhibitors.Citation146,Citation157,Citation160

Two large pan-cancer studies manifested a significant relationship between intra-tumor fungal ecologies and the tumor type and the fungal–fungal interaction or fungal–bacterial interaction as potential indicator of prognosis, such as the increased Candida-to-Saccharomyces ratio in late-stage metastatic CRC, and specific fungal–bacterial-immune clusters.Citation112,Citation161

5. Influence on tumor treatment

5.1. Chemotherapy

Researchers have discovered the effect of gut microbiota, especially gram-positive bacteria, in reducing the pathogenic Th17 cell response and rescuing cyclophosphamide resistance.Citation162,Citation163 New strategies have been discovered targeting gut microbiota to enhance the efficacy of chemotherapy, including bacterial derivatives and phage-guided nanotechnology.Citation164,Citation165

Few studies have been dedicated to the association of the fungal community with the efficiency and resistance of tumor chemotherapy. C. tropicalis is associated with CRC and is shown to promote chemoresistance via lactate production and MLH1 inhibition. Inhibiting lactate mitigates the resistance via the GPR81-cAMP-PKA-CREB pathway, regulating MMR protein expression.Citation139

5.2. Fecal microbiota transplantation

Fecal microbiota transplantation (FMT) is a transformative method that effectively alters recipients’ gut microbiota by transferring a diverse consortium of host-adapted microbial species, encompassing bacteria, bacteriophages, fungi, and their metabolites.Citation166 In recent years, FMT has been taken into clinical trials of multiple diseases, both intra-intestinal and extra-­intestinal, including recurrent C. difficile infection (CDI), IBD, graft­-versus-­host-­disease (GVHD), irritable bowel syndrome, obesity, and diabetes.Citation158,Citation167–170

FMT is able to reverse the reduction in fungi taxa and increase in C. albicans in recipients with CDI and ulcerative colitis.Citation158,Citation167 Overloading C. albicans in donor stool correlates with reduced FMT efficacy in CDI patients.Citation167 Meanwhile, recipients with relatively high abundance of gut C. albicans respond better to FMT than controls in the condition of ulcerative colitis, and the mechanism may involve the beneficial condition for bacterial diversity recovery.Citation158 It is also found to increase the abundance of certain fungi such as Aspergillus in CDICitation167 and Schizosaccharomyces pombe in ulcerative colitisCitation171 after FMT.

The application of FMT has not extended to the treatment of malignancies until now, except for some attempts at improving the efficacy of immunotherapies. Wang et al. presented two case series of immune checkpoint inhibitor-associated colitis successfully treated with FMT, restoring gut microbiota balance, substantially decreasing cytotoxic T cells, accompanying the concomitant increase of regulatory T cells.Citation172 Yet, since only two patients were treated and no control groups were involved in the study, more valid evidence should be given to prove its efficiency and safety.Citation172

The long-time safety of FMT has not been proven, although studied for years. The known possible adverse events are only limited to mild side effects such as abdominal cramping, bloating, and sore throat, with one death discovered attributed to FMT.Citation173–175 More clinical trials and cohorts should be done to explore all the possible adverse effects to ensure its safety.

5.3. Fungal- derived products

Bacterial-associated products, including prebiotics, probiotics, and synbiotics, are the most commonly acknowledged microbial adjustment products that may work in tumor prevention and treatment.Citation176 Meanwhile, fungal-derived products are also worth mentioning. Citrinin is a mycotoxin derived from polyketides, and it is produced by various fungal strains, including those belonging to the genera Monascus, Aspergillus, and Penicillium. It has been studied for a long time for its potential antineoplastic effect by inducing cell death, reducing DNA repair capacity, and inducing cytotoxic and genotoxic effects.Citation177,Citation178 β-glucan administration, whether oral or systemic, has shown promising antineoplastic effects in multiple malignancies including breast cancer, colon cancer, and melanoma, exhibiting benefits in metastasis treatment. The mechanism might involve the induction of tumoricidal peripheral blood monocytes, macrophages, and NK cells, as well as pro-inflammatory cytokines/chemokines including IFN-γ, TNF-a, CXCL9, and CXCL10, and enhanced IRF1 and PD-L1 expression.Citation179–181 Marine-derived Aspergillus is capable of producing metabolites that are potentially tumoricidal. Asperphenin A inhibits CRC growth by triggering microtubule disassembly and cell cycle arrest, and also has anti-proliferative effect in multiple tumors.Citation182 Preussin displayed cytotoxic and anti-proliferative activities in breast cancer cell lines.Citation183 β-Galactosidases, known as one of a kind of prebiotics, function as lysosomal exoglycosidases involved in glycoconjugate metabolism. This enzyme can also be isolated from Aspergillus Terreus to effectively promote the proliferation of MCF-7 breast cancer cells in vitro.Citation184

5.4. Radiotherapy

Whether microbiome affects radiotherapy has not been clearly comprehended until now. Commensal microbiomes may differently influence the response to radiotherapy. Depletion of fungi is beneficial for radiotherapy whereas antibiotic-mediated bacterial depletion reduces responsiveness. The immune responses included in these effects are orchestrated by Dectin-1.Citation185

5.5. Immunotherapy

Immunotherapy is revolutionizing tumor treatment. Strategies have been proposed to augment the potency of drugs and immune generation. Response to PD-1 blockade treatment was observed to have a positive correlation with increasing abundance of Akkermansia after FMT in murine models, followed by increased recruitment of CCR9+CXCR3+CD4+ T cells.Citation186 FMT also shows positive results in counteracting tumor microenvironment and rescuing anti-PD-1 sensitivity in clinical trials of melanoma.Citation187 Β-glucan and its nanoparticles are demonstrated to manipulate the tumor microenvironment, thereby augmenting the response to immunotherapy.Citation188 Our recent study also demonstrated the gut bacterial-fungi interplay and the possibility of fungi as biomarker for efficacy of immunotherapy.Citation160

Overall, the usage of fungi in the tumor treatment is almost a brand-new field. Recent studies majorly focus on the potential causal relationships of the fungi with the effectiveness, efficacy, and resistance of tumor treatment. Some specific fungi species and fungal products are mentioned, including C. tropicalis that promotes chemoresistance in CRC, some fungal-derived products like citrinin and β-glucan exhibiting antineoplastic effects, and Aspergillus metabolites that show promise in CRC treatment. But much more about fungi and cancer treatment awaits to be found, especially in the field of FMT and immunotherapy, which are the revolutionizing medical techniques and therapies for disease treatment.

Conclusion

Although fungi and cancer have been widely studied separately for years, the cancer mycobiome field remains at its starting-line. The techniques and database are far from united and integrated, and there are large amounts of unexplored gut fungi beyond our knowledge. The sample type and size vary greatly between studies and significant amounts of unclassified taxa were mentioned. These limitations have greatly disturbed scientists to find the actual causal relationship between cancer development and fungal changes. Nonetheless, recent studies still help with the discovery of the carcinogenic power of pathogenic fungi, functioning through immune activation, metabolites, toxin production, bacterial–fungal interaction, or biofilm formation. Although the validity, feasibility, and security of the present strategies and investigation methods involving gut mycobiome need to be further addressed, the application of gut fungi to the screening, prevention, and treatment of cancers is a promising field well-worth dedication.

Acknowledgments

This work was supported by grants from the National Natural Science Foundation of China (82073115); the National Key Research and Development Plan of the Ministry of Science and Technology (2022YFE0125300); the Shanghai Municipal Education Commission-Gaofeng Clinical Medicine Grant (no. 20152512).

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Additional information

Funding

The work was supported by the National Natural Science Foundation of China [82073115].

References

  • Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: an integrative view. Cell. 2012;148(6):1258–23. doi:10.1016/j.cell.2012.01.035.
  • Wang ZK, Yang YS, Stefka AT, Sun G, Peng LH. Review article: fungal microbiota and digestive diseases. Aliment Pharmacol Ther. 2014;39(8):751–766. doi:10.1111/apt.12665.
  • Gauthier GM. Fungal Dimorphism and Virulence: Molecular Mechanisms for Temperature Adaptation, Immune Evasion, and In Vivo Survival. Mediators Inflamm. 2017;2017:1–8. doi:10.1155/2017/8491383.
  • Witchley JN, Penumetcha P, Abon NV, Woolford CA, Mitchell AP, Noble SM. Candida albicans morphogenesis programs control the balance between gut commensalism and invasive infection. Cell Host & Microbe. 2019;25(3):432–443.e6. doi:10.1016/j.chom.2019.02.008.
  • Gow NA, van de Veerdonk FL, Brown AJ, Netea MG. Candida albicans morphogenesis and host defence: discriminating invasion from colonization. Nat Rev Microbiol. 2011;10(2):112–122. doi:10.1038/nrmicro2711.
  • Wong SH, Yu J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16(11):690–704. doi:10.1038/s41575-019-0209-8.
  • Hardison SE, Brown GD. C-type lectin receptors orchestrate antifungal immunity. Nat Immunol. 2012;13(9):817–822. doi:10.1038/ni.2369.
  • Lamas B, Richard ML, Leducq V, Pham HP, Michel ML, Da Costa G, Bridonneau C, Jegou S, Hoffmann TW, Natividad JM. et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med. 2016;22(6):598–605. doi:10.1038/nm.4102.
  • Hara H, Ishihara C, Takeuchi A, Imanishi T, Xue L, Morris SW, Inui M, Takai T, Shibuya A, Saijo S. et al. The adaptor protein CARD9 is essential for the activation of myeloid cells through ITAM-associated and Toll-like receptors. Nat Immunol. 2007;8(6):619–629. doi:10.1038/ni1466.
  • Doron I, Leonardi I, Li XV, Fiers WD, Semon A, Bialt-DeCelie M, Migaud M, Gao IH, Lin WY, Kusakabe T. et al. Human gut mycobiota tune immunity via CARD9-dependent induction of anti-fungal IgG antibodies. Cell. 2021;184(4):1017–1031.e14. doi:10.1016/j.cell.2021.01.016.
  • Gross O, Gewies A, Finger K, Schafer M, Sparwasser T, Peschel C, Forster I, Ruland J. Card9 controls a non-TLR signalling pathway for innate anti-fungal immunity. Nature. 2006;442(7103):651–656. doi:10.1038/nature04926.
  • Gringhuis SI, Kaptein TM, Wevers BA, Theelen B, van der Vlist M, Boekhout T, Geijtenbeek TB. Dectin-1 is an extracellular pathogen sensor for the induction and processing of IL-1β via a noncanonical caspase-8 inflammasome. Nat Immunol. 2012;13(3):246–254. doi:10.1038/ni.2222.
  • Ketelut-Carneiro N, Ghosh S, Levitz SM, Fitzgerald KA, da Silva JS. A dectin-1-caspase-8 pathway licenses canonical caspase-1 inflammasome activation and interleukin-1β release in response to a pathogenic fungus. J Infect Dis. 2018;217(2):329–339. doi:10.1093/infdis/jix568.
  • Liang N, Yang YP, Li W, Wu YY, Zhang ZW, Luo Y, Fan YM. Overexpression of NLRP 3, NLRC 4 and AIM 2 inflammasomes and their priming-associated molecules (TLR 2, TLR 4, dectin-1, dectin-2 and NF κB) in Malassezia folliculitis. Mycoses. 2018;61(2):111–118. doi:10.1111/myc.12711.
  • Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–489. doi:10.1038/s41577-019-0165-0.
  • Jo EK, Kim JK, Shin DM, Sasakawa C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol Immunol. 2016;13(2):148–159. doi:10.1038/cmi.2015.95.
  • Zhang Z, Chen Y, Yin Y, Chen Y, Chen Q, Bing Z, Zheng Y, Hou Y, Shen S, Chen Y. et al. Candida tropicalis induces NLRP3 inflammasome activation via glycogen metabolism-dependent glycolysis and JAK-STAT1 signaling pathway in myeloid-derived suppressor cells to promote colorectal carcinogenesis. Int Immunopharmacol. 2022;113:109430. doi:10.1016/j.intimp.2022.109430.
  • Zhu LL, Zhao XQ, Jiang C, You Y, Chen XP, Jiang YY, Jia XM, Lin X. C-type lectin receptors Dectin-3 and Dectin-2 form a heterodimeric pattern-recognition receptor for host defense against fungal infection. Immunity. 2013;39(2):324–334. doi:10.1016/j.immuni.2013.05.017.
  • Talapko J, Mestrovic T, Dmitrovic B, Juzbasic M, Matijevic T, Bekic S, Eric S, Flam J, Belic D, Petek Eric A. et al. A putative role of Candida albicans in promoting cancer development: a Current state of evidence and proposed mechanisms. Microorganisms. 2023;11(6):1476. doi:10.3390/microorganisms11061476.
  • Aparicio-Fernandez L, Antoran A, Areitio M, Rodriguez-Erenaga O, Martin-Souto L, Buldain I, Marquez J, Benedicto A, Arteta B, Pellon A. et al. Candida albicans increases the aerobic glycolysis and activates MAPK–dependent inflammatory response of liver sinusoidal endothelial cells. Microbes Infect. 2024;105305:105305. doi:10.1016/j.micinf.2024.105305.
  • Zheng NX, Wang Y, Hu DD, Yan L, Jiang YY. The role of pattern recognition receptors in the innate recognition of Candida albicans. Virulence. 2015;6(4):347–361. doi:10.1080/21505594.2015.1014270.
  • Luisa Gil M, Murciano C, Yanez A, Gozalbo D. Role of Toll-like receptors in systemic Candida albicans infections. Front Biosci. 2016;21(2):278–302. doi:10.2741/4388.
  • Netea MG, Joosten LA, van der Meer JW, Kullberg BJ, van de Veerdonk FL. Immune defence against Candida fungal infections. Nat Rev Immunol. 2015;15(10):630–642. doi:10.1038/nri3897.
  • Ishikawa T, Itoh F, Yoshida S, Saijo S, Matsuzawa T, Gonoi T, Saito T, Okawa Y, Shibata N, Miyamoto T. et al. Identification of distinct ligands for the C-type lectin receptors mincle and dectin-2 in the pathogenic fungus malassezia. Cell Host Microbe. 2013;13(4):477–488. doi:10.1016/j.chom.2013.03.008.
  • Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32(1):19–25. doi:10.1016/j.it.2010.10.002.
  • Wang T, Fan C, Yao A, Xu X, Zheng G, You Y, Jiang C, Zhao X, Hou Y, Hung MC. et al. The adaptor protein CARD9 protects against colon cancer by Restricting Mycobiota-Mediated Expansion of Myeloid-Derived Suppressor Cells. Immunity. 2018;49(3):504–514.e4. doi:10.1016/j.immuni.2018.08.018.
  • Weber R, Riester Z, Huser L, Sticht C, Siebenmorgen A, Groth C, Hu X, Altevogt P, Utikal JS, Umansky V. IL-6 regulates CCR5 expression and immunosuppressive capacity of MDSC in murine melanoma. J Immunother Cancer. 2020;8(2) doi:10.1136/jitc-2020-000949.
  • Wang Y, Liu H, Zhang Z, Bian D, Shao K, Wang S, Ding Y. G-MDSC-derived exosomes mediate the differentiation of M-MDSC into M2 macrophages promoting colitis-to-cancer transition. J Immunother Cancer. 2023;11(6)doi:10.1136/jitc-2022-006166.
  • Waight JD, Netherby C, Hensen ML, Miller A, Hu Q, Liu S, Bogner PN, Farren MR, Lee KP, Liu K. et al. Myeloid-derived suppressor cell development is regulated by a STAT/IRF-8 axis. J Clin Invest. 2013;123(10):4464–4478. doi:10.1172/JCI68189.
  • Bacher P, Hohnstein T, Beerbaum E, Rocker M, Blango MG, Kaufmann S, Rohmel J, Eschenhagen P, Grehn C, Seidel K. et al. Human Anti-fungal Th17 Immunity and Pathology Rely on Cross-Reactivity against Candida albicans. Cell. 2019;176(6):1340–1355.e15. doi:10.1016/j.cell.2019.01.041.
  • LeibundGut-Landmann S, Gross O, Robinson MJ, Osorio F, Slack EC, Tsoni SV, Schweighoffer E, Tybulewicz V, Brown GD, Ruland J. et al. Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17. Nat Immunol. 2007;8(6):630–638. doi:10.1038/ni1460.
  • Sparber F, De Gregorio C, Steckholzer S, Ferreira FM, Dolowschiak T, Ruchti F, Kirchner FR, Mertens S, Prinz I, Joller N. et al. The skin commensal yeast malassezia triggers a type 17 response that coordinates anti-fungal immunity and exacerbates skin inflammation. Cell Host & Microbe. 2019;25(3):389–403.e6. doi:10.1016/j.chom.2019.02.002.
  • Shao S, Yu X, Shen L. Autoimmune thyroid diseases and Th17/Treg lymphocytes. Life Sci. 2018;192:160–165. doi:10.1016/j.lfs.2017.11.026.
  • Noda S, Suarez-Farinas M, Ungar B, Kim SJ, de Guzman Strong C, Xu H, Peng X, Estrada YD, Nakajima S, Honda T. et al. The Asian atopic dermatitis phenotype combines features of atopic dermatitis and psoriasis with increased TH17 polarization. J Allergy Clin Immunol. 2015;136(5):1254–1264. doi:10.1016/j.jaci.2015.08.015.
  • Rovedatti L, Kudo T, Biancheri P, Sarra M, Knowles CH, Rampton DS, Corazza GR, Monteleone G, Di Sabatino A, Macdonald TT. Differential regulation of interleukin 17 and interferon production in inflammatory bowel disease. Gut. 2009;58(12):1629–1636. doi:10.1136/gut.2009.182170.
  • Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E, Caccamo M, Oukka M, Weiner HL. Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature. 2008;453(7191):65–71. doi:10.1038/nature06880.
  • Kumamoto CA. Inflammation and gastrointestinal Candida colonization. Curr Opin Microbiol. 2011;14(4):386–391. doi:10.1016/j.mib.2011.07.015.
  • Moschen AR, Tilg H, Raine T. IL-12, IL-23 and IL-17 in IBD: immunobiology and therapeutic targeting. Nat Rev Gastroenterol Hepatol. 2019;16(3):185–196. doi:10.1038/s41575-018-0084-8.
  • Yan J, Smyth MJ, Teng MWL. Interleukin (IL)-12 and IL-23 and their conflicting roles in cancer. Cold Spring Harb Perspect Biol. 2018;10(7):a028530. doi:10.1101/cshperspect.a028530.
  • MacAlpine J, Robbins N, Cowen LE. Bacterial-fungal interactions and their impact on microbial pathogenesis. Mol Ecol. 2022;32(10):2565–2581. doi:10.1111/mec.16411.
  • Montelongo-Jauregui D, Saville SP, Lopez-Ribot JL, Lorenz M. Contributions of Candida albicans Dimorphism, adhesive interactions, and extracellular matrix to the formation of dual-species biofilms with streptococcus gordonii. mBio. 2019;10(3). doi:10.1128/mBio.01179-19.
  • Peleg AY, Hogan DA, Mylonakis E. Medically important bacterial–fungal interactions. Nat Rev Microbiol. 2010;8(5):340–349. doi:10.1038/nrmicro2313.
  • Garcia C, Tebbji F, Daigneault M, Liu NN, Kohler JR, Allen-Vercoe E, Sellam A, Mitchell AP. The human gut microbial metabolome modulates fungal growth via the TOR signaling pathway. mSphere. 2017;2(6). doi:10.1128/mSphere.00555-17.
  • Fan D, Coughlin LA, Neubauer MM, Kim J, Kim MS, Zhan X, Simms-Waldrip TR, Xie Y, Hooper LV, Koh AY. Activation of HIF-1α and LL-37 by commensal bacteria inhibits Candida albicans colonization. Nat Med. 2015;21(7):808–814. doi:10.1038/nm.3871.
  • Guinan J, Wang S, Hazbun TR, Yadav H, Thangamani S. Antibiotic-induced decreases in the levels of microbial-derived short-chain fatty acids correlate with increased gastrointestinal colonization of Candida albicans. Sci Rep. 2019;9(1):8872. doi:10.1038/s41598-019-45467-7.
  • Cabral DJ, Penumutchu S, Norris C, Morones-Ramirez JR, Belenky P. Microbial competition between Escherichia coli and Candida albicans reveals a soluble fungicidal factor. Microb Cell. 2018;5(5):249–255. doi:10.15698/mic2018.05.631.
  • Eckstein MT, Moreno-Velasquez SD, Perez JC. Gut bacteria shape intestinal microhabitats occupied by the fungus Candida albicans. Curr Biol. 2020;30(23):4799–4807.e4. doi:10.1016/j.cub.2020.09.027.
  • Zeise KD, Woods RJ, Huffnagle GB. Interplay between Candida albicans and lactic acid bacteria in the gastrointestinal tract: impact on colonization resistance, microbial carriage, opportunistic infection, and Host immunity. Clin Microbiol Rev. 2021;34(4):e0032320. doi:10.1128/CMR.00323-20.
  • Cuskin F, Lowe EC, Temple MJ, Zhu Y, Cameron E, Pudlo NA, Porter NT, Urs K, Thompson AJ, Cartmell A. et al. Human gut bacteroidetes can utilize yeast mannan through a selfish mechanism. Nature. 2015;517(7533):165–169. doi:10.1038/nature13995.
  • Valentine M, Benade E, Mouton M, Khan W, Botha A. Binary interactions between the yeast Candida albicans and two gut-associated bacteroides species. Microb Pathog. 2019;135:103619. doi:10.1016/j.micpath.2019.103619.
  • Perez JC. The interplay between gut bacteria and the yeast Candida albicans. Gut Microbes. 2021;13(1):1979877. doi:10.1080/19490976.2021.1979877.
  • Mason KL, Erb Downward JR, Mason KD, Falkowski NR, Eaton KA, Kao JY, Young VB, Huffnagle GB, Deepe GS. Candida albicans and bacterial microbiota interactions in the cecum during recolonization following broad-spectrum antibiotic therapy. Infect Immun. 2012;80(10):3371–3380. doi:10.1128/IAI.00449-12.
  • Erb Downward JR, Falkowski NR, Mason KL, Muraglia R, Huffnagle GB. Modulation of post-antibiotic bacterial community reassembly and host response by Candida albicans. Sci Rep. 2013;3(1):2191. doi:10.1038/srep02191.
  • 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. 2020;8(1):133. doi:10.1186/s40168-020-00899-6.
  • Jiang TT, Shao TY, Ang WXG, Kinder JM, Turner LH, Pham G, Whitt J, Alenghat T, Way SS. Commensal Fungi Recapitulate the Protective Benefits of Intestinal Bacteria. Cell Host Microbe. 2017;22(6):809–816.e4. doi:10.1016/j.chom.2017.10.013.
  • van Tilburg Bernardes E, Pettersen VK, Gutierrez MW, Laforest-Lapointe I, Jendzjowsky NG, Cavin JB, Vicentini FA, Keenan CM, Ramay HR, Samara J. et al. Intestinal fungi are causally implicated in microbiome assembly and immune development in mice. Nat Commun. 2020;11(1):2577. doi:10.1038/s41467-020-16431-1.
  • Sovran B, Planchais J, Jegou S, Straube M, Lamas B, Natividad JM, Agus A, Dupraz L, Glodt J, Da Costa G. et al. Enterobacteriaceae are essential for the modulation of colitis severity by fungi. Microbiome. 2018;6(1):152. doi:10.1186/s40168-018-0538-9.
  • Jang YJ, Kim WK, Han DH, Lee K, Ko G. Lactobacillus fermentum species ameliorate dextran sulfate sodium-induced colitis by regulating the immune response and altering gut microbiota. Gut Microbes. 2019;10(6):696–711. doi:10.1080/19490976.2019.1589281.
  • Lohse MB, Gulati M, Johnson AD, Nobile CJ. Development and regulation of single- and multi-species Candida albicans biofilms. Nat Rev Microbiol. 2018;16(1):19–31. doi:10.1038/nrmicro.2017.107.
  • Hasan F, Xess I, Wang X, Jain N, Fries BC. Biofilm formation in clinical Candida isolates and its association with virulence. Microbes Infect. 2009;11(8–9):753–761. doi:10.1016/j.micinf.2009.04.018.
  • Ponde NO, Lortal L, Ramage G, Naglik JR, Richardson JP. Candida albicans biofilms and polymicrobial interactions. Crit Rev Microbiol. 2021;47(1):91–111. doi:10.1080/1040841X.2020.1843400.
  • Blankenship JR, Mitchell AP. How to build a biofilm: a fungal perspective. Curr Opin Microbiol. 2006;9(6):588–594. doi:10.1016/j.mib.2006.10.003.
  • Nobile CJ, Andes DR, Nett JE, Smith FJ, Yue F, Phan QT, Edwards JE, Filler SG, Mitchell AP, Johnson A. Critical role of Bcr1-dependent adhesins in C. albicans biofilm formation in vitro and in vivo. PLoS Pathog. 2006;2(7):e63. doi:10.1371/journal.ppat.0020063.
  • Harriott MM, Lilly EA, Rodriguez TE, Fidel PL, Noverr MC. Candida albicans forms biofilms on the vaginal mucosa. Microbiol. 2010;156(12):3635–3644. doi:10.1099/mic.0.039354-0.
  • White SJ, Rosenbach A, Lephart P, Nguyen D, Benjamin A, Tzipori S, Whiteway M, Mecsas J, Kumamoto CA, Cormack BP. Self-regulation of Candida albicans population size during GI colonization. PLoS Pathog. 2007;3(12):e184. doi:10.1371/journal.ppat.0030184.
  • Nobile CJ, Schneider HA, Nett JE, Sheppard DC, Filler SG, Andes DR, Mitchell AP. Complementary adhesin function in C. albicans biofilm formation. Curr Biol. 2008;18(14):1017–1024. doi:10.1016/j.cub.2008.06.034.
  • Nett JE, Marchillo K, Spiegel CA, Andes DR. Development and validation of an in vivo Candida albicans biofilm denture model. Infect Immun. 2010;78(9):3650–3659. doi:10.1128/IAI.00480-10.
  • Dwivedi P, Thompson A, Xie Z, Kashleva H, Ganguly S, Mitchell AP, Dongari-Bagtzoglou A, Davis D. Role of Bcr1-activated genes Hwp1 and Hyr1 in Candida albicans oral mucosal biofilms and neutrophil evasion. PLoS One. 2011;6(1):e16218. doi:10.1371/journal.pone.0016218.
  • Andes D, Nett J, Oschel P, Albrecht R, Marchillo K, Pitula A. Development and characterization of an in vivo central venous catheter Candida albicans biofilm model. Infect Immun. 2004;72(10):6023–6031. doi:10.1128/IAI.72.10.6023-6031.2004.
  • Zarnowski R, Westler WM, Lacmbouh GA, Marita JM, Bothe JR, Bernhardt J, Lounes-Hadj Sahraoui A, Fontaine J, Sanchez H, Hatfield RD. et al. Novel entries in a fungal biofilm matrix encyclopedia. mBio. 2014;5(4):e01333–01314. doi:10.1128/mBio.01333-14.
  • Hornby JM, Jensen EC, Lisec AD, Tasto JJ, Jahnke B, Shoemaker R, Dussault P, Nickerson KW. Quorum sensing in the dimorphic fungus Candida albicans is mediated by farnesol. Appl Environ Microbiol. 2001;67(7):2982–2992. doi:10.1128/AEM.67.7.2982-2992.2001.
  • Leonhardt I, Spielberg S, Weber M, Albrecht-Eckardt D, Blass M, Claus R, Barz D, Scherlach K, Hertweck C, Loffler J. et al. The fungal quorum-sensing molecule farnesol activates innate immune cells but suppresses cellular adaptive immunity. mBio. 2015;6(2):e00143. doi:10.1128/mBio.00143-15.
  • Fan F, Liu Y, Liu Y, Lv R, Sun W, Ding W, Cai Y, Li W, Liu X, Qu W. Candida albicans biofilms: antifungal resistance, immune evasion, and emerging therapeutic strategies. Int J Antimicrob Agents. 2022;60(5–6):106673. doi:10.1016/j.ijantimicag.2022.106673.
  • Ramage G, Rajendran R, Sherry L, Williams C. Fungal biofilm resistance. Iran J Pediatr Hematol Oncol. 2012;2012:1–14. doi:10.1155/2012/528521.
  • Lachat J, Pascault A, Thibaut D, Le Borgne R, Verbavatz JM, Weiner A. Trans-cellular tunnels induced by the fungal pathogen Candida albicans facilitate invasion through successive epithelial cells without host damage. Nat Commun. 2022;13(1):3781. doi:10.1038/s41467-022-31237-z.
  • Wang X, Zhang W, Wu W, Wu S, Young A, Yan Z. Is Candida albicans a contributor to cancer? A critical review based on the current evidence. Microbiol Res. 2023;272:127370. doi:10.1016/j.micres.2023.127370.
  • Tsang PW, Fong WP, Samaranayake LP, Sturtevant J. Candida albicans orf19.3727 encodes phytase activity and is essential for human tissue damage. PLoS One. 2017;12(12):e0189219. doi:10.1371/journal.pone.0189219.
  • Reyna-Beltran E, Iranzo M, Calderon-Gonzalez KG, Mondragon-Flores R, Labra-Barrios ML, Mormeneo S, Luna-Arias JP. The Candida albicans ENO1 gene encodes a transglutaminase involved in growth, cell division, morphogenesis, and osmotic protection. J Biol Chem. 2018;293(12):4304–4323. doi:10.1074/jbc.M117.810440.
  • Alnuaimi AD, Ramdzan AN, Wiesenfeld D, O’Brien-Simpson NM, Kolev SD, Reynolds EC, McCullough MJ. Candida virulence and ethanol-derived acetaldehyde production in oral cancer and non-cancer subjects. Oral Dis. 2016;22(8):805–814. doi:10.1111/odi.12565.
  • Conkova E, Proskovcova M, Vaczi P, Malinovska Z. In Vitro Biofilm Formation by Malassezia pachydermatis Isolates and Its Susceptibility to Azole Antifungals. J Fungi (Basel). 2022;8(11):1209. doi:10.3390/jof8111209.
  • Angiolella L, Leone C, Rojas F, Mussin J, de Los Angeles Sosa M, Giusiano G. Biofilm, adherence, and hydrophobicity as virulence factors in Malassezia furfur. Med Mycol. 2018;56(1):110–116. doi:10.1093/mmy/myx014.
  • Morelli KA, Kerkaert JD, Cramer RA, Xue C. Aspergillus fumigatus biofilms: toward understanding how growth as a multicellular network increases antifungal resistance and disease progression. PLoS Pathog. 2021;17(8):e1009794. doi:10.1371/journal.ppat.1009794.
  • Walsh TJ, Schlegel R, Moody MM, Costerton JW, Salcman M. Ventriculoatrial shunt infection due to Cryptococcus neoformans: an ultrastructural and quantitative microbiological study. Neurosurgery. 1986;18(3):373–375. doi:10.1097/00006123-198603000-00025.
  • Moyes DL, Wilson D, Richardson JP, Mogavero S, Tang SX, Wernecke J, Hofs S, Gratacap RL, Robbins J, Runglall M. et al. Candidalysin is a fungal peptide toxin critical for mucosal infection. Nature. 2016;532(7597):64–68. doi:10.1038/nature17625.
  • Moyes DL, Runglall M, Murciano C, Shen C, Nayar D, Thavaraj S, Kohli A, Islam A, Mora-Montes H, Challacombe SJ. et al. A biphasic innate immune MAPK response discriminates between the yeast and hyphal forms of Candida albicans in epithelial cells. Cell Host Microbe. 2010;8(3):225–235. doi:10.1016/j.chom.2010.08.002.
  • Ho J, Yang X, Nikou SA, Kichik N, Donkin A, Ponde NO, Richardson JP, Gratacap RL, Archambault LS, Zwirner CP. et al. Candidalysin activates innate epithelial immune responses via epidermal growth factor receptor. Nat Commun. 2019;10(1):2297. doi:10.1038/s41467-019-09915-2.
  • Nikou SA, Zhou C, Griffiths JS, Kotowicz NK, Coleman BM, Green MJ, Moyes DL, Gaffen SL, Naglik JR, Parker PJ. The Candida albicans toxin candidalysin mediates distinct epithelial inflammatory responses through p38 and EGFR-ERK pathways. Sci Signal. 2022;15(728):eabj6915. doi:10.1126/scisignal.abj6915.
  • Rogiers O, Frising UC, Kucharikova S, Jabra-Rizk MA, van Loo G, Van Dijck P, Wullaert A, Lorenz M. Candidalysin crucially contributes to Nlrp3 inflammasome activation by Candida albicans hyphae. mBio. 2019;10(1). doi:10.1128/mBio.02221-18.
  • Verma AH, Richardson JP, Zhou C, Coleman BM, Moyes DL, Ho J, Huppler AR, Ramani K, McGeachy MJ, Mufazalov IA. et al. Oral epithelial cells orchestrate innate type 17 responses to Candida albicans through the virulence factor candidalysin. Sci Immunol. 2017;2(17). doi:10.1126/sciimmunol.aam8834.
  • Nieminen MT, Uittamo J, Salaspuro M, Rautemaa R. Acetaldehyde production from ethanol and glucose by non-Candida albicans yeasts in vitro. Oral Oncol. 2009;45(12):e245–e248. doi:10.1016/j.oraloncology.2009.08.002.
  • Gaitanis G, Magiatis P, Hantschke M, Bassukas ID, Velegraki A. The malassezia genus in skin and systemic diseases. Clin Microbiol Rev. 2012;25(1):106–141. doi:10.1128/CMR.00021-11.
  • Vlachos C, Schulte BM, Magiatis P, Adema GJ, Gaitanis G. Malassezia-derived indoles activate the aryl hydrocarbon receptor and inhibit Toll-like receptor-induced maturation in monocyte-derived dendritic cells. Br J Dermatol. 2012;167(3):496–505. doi:10.1111/j.1365-2133.2012.11014.x.
  • Pollet M, Shaik S, Mescher M, Frauenstein K, Tigges J, Braun SA, Sondenheimer K, Kaveh M, Bruhs A, Meller S. et al. The AHR represses nucleotide excision repair and apoptosis and contributes to UV-induced skin carcinogenesis. Cell Death Differ. 2018;25:1823–1836. doi:10.1038/s41418-018-0160-1.
  • Venkateswaran N, Lafita-Navarro MC, Hao YH, Kilgore JA, Perez-Castro L, Braverman J, Borenstein-Auerbach N, Kim M, Lesner NP, Mishra P. et al. MYC promotes tryptophan uptake and metabolism by the kynurenine pathway in colon cancer. Genes Dev. 2019;33(17–18):1236–1251. doi:10.1101/gad.327056.119.
  • Saxena N, Ansari KM, Kumar R, Chaudhari BP, Dwivedi PD, Das M. Role of mitogen activated protein kinases in skin tumorigenicity of patulin. Toxicol Appl Pharmacol. 2011;257(2):264–271. doi:10.1016/j.taap.2011.09.012.
  • Yagudayev E, Ray SD. Aflatoxin. In: Wexler P. editor. Encyclopedia of Toxicology. Fourth. Academic Press; 2024. pp. 193–200. doi:10.1016/B978-0-12-824315-2.00179-2.
  • Lo EKK, Wang X, Lee PK, Wong HC, Lee JC, Gomez-Gallego C, Zhao D, El-Nezami H, Li J. Mechanistic insights into zearalenone-accelerated colorectal cancer in mice using integrative multi-omics approaches. Comput Struct Biotechnol J. 2023;21:1785–1796. doi:10.1016/j.csbj.2023.02.048.
  • Sanjaya PR, Gokul S, Gururaj Patil B, Raju R. Candida in oral pre-cancer and oral cancer. Med Hypotheses. 2011;77(6):1125–1128. doi:10.1016/j.mehy.2011.09.018.
  • Silva S, Negri M, Henriques M, Oliveira R, Williams DW, Azeredo J. Candida glabrata, Candida parapsilosis and Candida tropicalis: biology, epidemiology, pathogenicity and antifungal resistance. FEMS Microbiol Rev. 2012;36(2):288–305. doi:10.1111/j.1574-6976.2011.00278.x.
  • Fakhim H, Vaezi A, Dannaoui E, Chowdhary A, Nasiry D, Faeli L, Meis JF, Badali H. Comparative virulence of Candida auris with Candida haemulonii, Candida glabrata and Candida albicans in a murine model. Mycoses. 2018;61(6):377–382. doi:10.1111/myc.12754.
  • Talukdar A, Barman R, Sarma A, Krishnatreya M, Sharma JD, Kataki AC. Fungal profile and antifungal susceptibility pattern of candidiasis in esophageal cancer patients. J Cancer Res Ther. 2020;16(8):S209–S212. doi:10.4103/jcrt.JCRT_581_18.
  • Scott BB, Jenkins D. Gastro-oesophageal candidiasis. Gut. 1982;23(2):137–139. doi:10.1136/gut.23.2.137.
  • Chang F, Syrjanen S, Wang L, Syrjanen K. Infectious agents in the etiology of esophageal cancer. Gastroenterology. 1992;103(4). doi:10.1016/0016-5085(92)91526-A.
  • Domingues-Ferreira M, Grumach AS, Duarte AJ, De Moraes-Vasconcelos D. Esophageal cancer associated with chronic mucocutaneous candidiasis. Could chronic candidiasis lead to esophageal cancer? Med Mycol. 2009;47(2):201–205. doi:10.1080/13693780802342545.
  • Delsing CE, Bleeker-Rovers CP, van de Veerdonk FL, Tol J, van der Meer JW, Kullberg BJ, Netea MG. Association of esophageal candidiasis and squamous cell carcinoma. Med Mycol Case Rep. 2012;1(1):5–8. doi:10.1016/j.mmcr.2012.02.003.
  • Koo S, Kejariwal D, Al-Shehri T, Dhar A, Lilic D. Oesophageal candidiasis and squamous cell cancer in patients with gain-of-function STAT1 gene mutation. United European Gastroenterol J. 2017;5(5):625–631. doi:10.1177/2050640616684404.
  • 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. 2017;21(4):478–493.e7. doi:10.1016/j.chom.2017.03.006.
  • Ohashi S, Miyamoto S, Kikuchi O, Goto T, Amanuma Y, Muto M. Recent Advances From Basic and Clinical Studies of Esophageal Squamous Cell Carcinoma. Gastroenterology. 2015;149(7):1700–1715. doi:10.1053/j.gastro.2015.08.054.
  • Nieminen MT, Salaspuro M. Local Acetaldehyde—An Essential Role in Alcohol-Related Upper Gastrointestinal Tract Carcinogenesis. Cancers Basel. 2018;10(1):11. doi:10.3390/cancers10010011.
  • Sano T, Ozaki K, Terayama Y, Kodama Y, Matsuura T. A novel diabetic murine model of Candida albicans-induced mucosal inflammation and proliferation. J Diabetes Res. 2014;2014:1–6. doi:10.1155/2014/509325.
  • Zhong M, Xiong Y, Zhao J, Gao Z, Ma J, Wu Z, Song Y, Hong X. Candida albicans disorder is associated with gastric carcinogenesis. Theranostics. 2021;11(10):4945–4956. doi:10.7150/thno.55209.
  • Dohlman AB, Klug J, Mesko M, Gao IH, Lipkin SM, Shen X, Iliev ID. A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell. 2022;185(20):3807–3822.e12. doi:10.1016/j.cell.2022.09.015.
  • Vogelaar IP, Ligtenberg MJ, van der Post RS, de Voer RM, Kets CM, Jansen TJ, Jacobs L, Schreibelt G, International Gastric Cancer Genetics G, de Vries IJ. et al. Recurrent candidiasis and early-onset gastric cancer in a patient with a genetically defined partial MYD88 defect. Fam Cancer. 2016;15(2):289–296. doi:10.1007/s10689-015-9859-z.
  • Hiengrach P, Panpetch W, Chindamporn A, Leelahavanichkul A. Helicobacter pylori, protected from antibiotics and stresses inside Candida albicans vacuoles, cause gastritis in mice. Int J Mol Sci. 2022;23(15):8568. doi:10.3390/ijms23158568.
  • Zhang Z, Feng H, Qiu Y, Xu Z, Xie Q, Ding W, Liu H, Li G. Dysbiosis of gastric mucosal fungal microbiota in the gastric cancer microenvironment. J Immunol Res. 2022;2022:1–14. doi:10.1155/2022/6011632.
  • Ndegwa N, Ploner A, Liu Z, Roosaar A, Axell T, Ye W. Association between poor oral health and gastric cancer: a prospective cohort study. Int J Cancer. 2018;143(9):2281–2288. doi:10.1002/ijc.31614.
  • Huo X, Li D, Wu F, Li S, Qiao Y, Wang C, Wang Y, Zhou C, Sun L, Luan Z. et al. Cultivated human intestinal fungus Candida metapsilosis M2006B attenuates colitis by secreting acyclic sesquiterpenoids as FXR agonists. Gut. 2022;71(11):2205–2217. doi:10.1136/gutjnl-2021-325413.
  • Limon JJ, Tang J, Li D, Wolf AJ, Michelsen KS, Funari V, Gargus M, Nguyen C, Sharma P, Maymi VI. et al. Malassezia is associated with Crohn’s disease and exacerbates colitis in mouse models. Cell Host Microbe. 2019;25(3):377–388.e6. doi:10.1016/j.chom.2019.01.007.
  • 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. 2017;66(6):1039–1048. doi:10.1136/gutjnl-2015-310746.
  • Gao R, Xia K, Wu M, Zhong H, Sun J, Zhu Y, Huang L, Wu X, Yin L, Yang R. et al. Alterations of gut mycobiota profiles in adenoma and colorectal cancer. Front Cell Infect Microbiol. 2022;12:839435. doi:10.3389/fcimb.2022.839435.
  • 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. 2015;5(1):7980. doi:10.1038/srep07980.
  • Jin H, Zhang C. High fat high calories diet (HFD) increase gut susceptibility to Carcinogens by altering the gut microbial community. J Cancer. 2020;11(14):4091–4098. doi:10.7150/jca.43561.
  • Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, Scheller J, Rose-John S, Cheroutre H, Eckmann L. et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 2009;15(2):103–113. doi:10.1016/j.ccr.2009.01.001.
  • Zhu Y, Shi T, Lu X, Xu Z, Qu J, Zhang Z, Shi G, Shen S, Hou Y, Chen Y. et al. Fungal-induced glycolysis in macrophages promotes colon cancer by enhancing innate lymphoid cell secretion of IL-22. EMBO J. 2021;40(11):e105320. doi:10.15252/embj.2020105320.
  • Malik A, Sharma D, Malireddi RKS, Guy CS, Chang TC, Olsen SR, Neale G, Vogel P, Kanneganti TD. SYK-CARD9 signaling axis promotes gut fungi-mediated inflammasome activation to restrict colitis and colon cancer. Immunity. 2018;49(3):515–530 e515. doi:10.1016/j.immuni.2018.08.024.
  • Yang AM, Inamine T, Hochrath K, Chen P, Wang L, Llorente C, Bluemel S, Hartmann P, Xu J, Koyama Y. et al. Intestinal fungi contribute to development of alcoholic liver disease. J Clin Invest. 2017;127:2829–2841. doi:10.1172/JCI90562.
  • Zeng S, Rosati E, Saggau C, Messner B, Chu H, Duan Y, Hartmann P, Wang Y, Ma S, Huang WJM. et al. Candida albicans-specific Th17 cell-mediated response contributes to alcohol-associated liver disease. Cell Host Microbe. 2023;31(3):389–404.e7. doi:10.1016/j.chom.2023.02.001.
  • Chu H, Duan Y, Lang S, Jiang L, Wang Y, Llorente C, Liu J, Mogavero S, Bosques-Padilla F, Abraldes JG. et al. The Candida albicans exotoxin candidalysin promotes alcohol-associated liver disease. J Hepatol. 2020;72(3):391–400. doi:10.1016/j.jhep.2019.09.029.
  • Furuya S, Cichocki JA, Konganti K, Dreval K, Uehara T, Katou Y, Fukushima H, Kono H, Pogribny IP, Argemi J. et al. Histopathological and molecular signatures of a mouse Model of acute-on-chronic alcoholic liver injury demonstrate concordance with human alcoholic hepatitis. Toxicol Sci. 2019;170(2):427–437. doi:10.1093/toxsci/kfy292.
  • Ren Z, Li A, Jiang J, Zhou L, Yu Z, Lu H, Xie H, Chen X, Shao L, Zhang R. et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut. 2019;68(6):1014–1023. doi:10.1136/gutjnl-2017-315084.
  • Liu Z, Li Y, Li C, Lei G, Zhou L, Chen X, Jia X, Lu Y. Intestinal Candida albicans promotes hepatocarcinogenesis by up-regulating NLRP6. Front Microbiol. 2022;13:812771. doi:10.3389/fmicb.2022.812771.
  • Zhang L, Chen C, Chai D, Li C, Qiu Z, Kuang T, Liu L, Deng W, Wang W. Characterization of the intestinal fungal microbiome in patients with hepatocellular carcinoma. J Transl Med. 2023;21(1):126. doi:10.1186/s12967-023-03940-y.
  • Usami M, Miyoshi M, Kanbara Y, Aoyama M, Sakaki H, Shuno K, Hirata K, Takahashi M, Ueno K, Hamada Y. et al. Analysis of fecal microbiota, organic acids and plasma lipids in hepatic cancer patients with or without liver cirrhosis. Clin Nutr. 2013;32(3):444–451. doi:10.1016/j.clnu.2012.09.010.
  • Ramirez-Garcia A, Rementeria A, Aguirre-Urizar JM, Moragues MD, Antoran A, Pellon A, Abad-Diaz-de-Cerio A, Hernando FL. Candida albicans and cancer: can this yeast induce cancer development or progression? Crit Rev Microbiol. 2016;42(2):181–193. doi:10.3109/1040841X.2014.913004.
  • Nasri E, Vaezi A, Falahatinejad M, Rizi MH, Sharifi M, Sadeghi S, Ataei B, Mirhendi H, Fakhim H. Species distribution and susceptibility profiles of oral candidiasis in hematological malignancy and solid tumor patients. Braz J Microbiol. 2023;54(1):143–149. doi:10.1007/s42770-022-00863-6.
  • Robinson S, Peterson CB, Sahasrabhojane P, Ajami NJ, Shelburne SA, Kontoyiannis DP, Galloway-Pena JR, Perlin DS. Observational cohort study of oral mycobiome and Interkingdom Interactions over the course of induction therapy for leukemia. mSphere. 2020;5(2). doi:10.1128/mSphere.00048-20.
  • Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, Brown J, Becker CA, Fleshner PR, Dubinsky M. et al. Interactions between commensal fungi and the C-type lectin receptor dectin-1 influence colitis. Science. 2012;336(6086):1314–1317. doi:10.1126/science.1221789.
  • Zhang Z, Zheng Y, Chen Y, Yin Y, Chen Y, Chen Q, Hou Y, Shen S, Lv M, Wang T. Gut fungi enhances immunosuppressive function of myeloid-derived suppressor cells by activating PKM2-dependent glycolysis to promote colorectal tumorigenesis. Exp Hematol Oncol. 2022;11(1):88. doi:10.1186/s40164-022-00334-6.
  • Qu J, Sun Z, Peng C, Li D, Yan W, Xu Z, Hou Y, Shen S, Chen P, Wang T. C. tropicalis promotes chemotherapy resistance in colon cancer through increasing lactate production to regulate the mismatch repair system. Int J Biol Sci. 2021;17(11):2756–2769. doi:10.7150/ijbs.59262.
  • Qu J, Chen Q, Bing Z, Shen S, Hou Y, Lv M, Wang T. C. tropicalis promotes CRC by down-regulating tumor cell-intrinsic PD-1 receptor via autophagy. J Cancer. 2023;14(10):1794–1808. doi:10.7150/jca.79664.
  • Ianiri G, LeibundGut-Landmann S, Dawson TL Jr. Malassezia: A Commensal, Pathogen, and Mutualist of Human and Animal Skin. Annu Rev Microbiol. 2022;76(1):757–782. doi:10.1146/annurev-micro-040820-010114.
  • Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC, Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA. et al. The gut mycobiome of the human microbiome project healthy cohort. Microbiome. 2017;5(1):153. doi:10.1186/s40168-017-0373-4.
  • Grice EA, Kong HH, Conlan S, Deming CB, Davis J, Young AC, Program NCS, Bouffard GG, Blakesley RW, Murray PR. et al. Topographical and temporal diversity of the human skin microbiome. Science. 2009;324(5931):1190–1192. doi:10.1126/science.1171700.
  • Sato Y, Fujimura T, Tanita K, Chunbing L, Matsushita S, Fujisawa Y, Otsuka A, Yamamoto Y, Hidaka T, Aiba S. Malassezia -derived aryl hydrocarbon receptor ligands enhance the CCL20/Th17/soluble CD163 pathogenic axis in extra-mammary Paget’s disease. Exp Dermatol. 2019;28(8):933–939. doi:10.1111/exd.13944.
  • Gao R, Kong C, Li H, Huang L, Qu X, Qin N, Qin H. Dysbiosis signature of mycobiota in colon polyp and colorectal cancer. Eur J Clin Microbiol Infect Dis. 2017;36(12):2457–2468. doi:10.1007/s10096-017-3085-6.
  • Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, Chan FKL, Sung JJY, Yu J. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 2019;68(4):654–662. doi:10.1136/gutjnl-2018-317178.
  • Stasiewicz M, Kwasniewski M, Karpinski TM. Microbial associations with pancreatic cancer: a new frontier in biomarkers. Cancers Basel. 2021;13(15):3784. doi:10.3390/cancers13153784.
  • 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. 2019;574(7777):264–267. doi:10.1038/s41586-019-1608-2.
  • 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. 2022;40(2):153–167.e11. doi:10.1016/j.ccell.2022.01.003.
  • Wolf AJ, Limon JJ, Nguyen C, Prince A, Castro A, Underhill DM. Malassezia spp. induce inflammatory cytokines and activate NLRP3 inflammasomes in phagocytes. J Leukoc Biol. 2021;109:161–172. doi:10.1002/JLB.2MA0820-259R.
  • Kistowska M, Fenini G, Jankovic D, Feldmeyer L, Kerl K, Bosshard P, Contassot E, French LE. Malassezia yeasts activate the NLRP3 inflammasome in antigen-presenting cells via Syk-kinase signalling. Exp Dermatol. 2014;23(12):884–889. doi:10.1111/exd.12552.
  • De Monte L, Reni M, Tassi E, Clavenna D, Papa I, Recalde H, Braga M, Di Carlo V, Doglioni C, Protti MP. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208(3):469–478. doi:10.1084/jem.20101876.
  • Kharofa J, Haslam D, Wilkinson R, Weiss A, Patel S, Wang K, Esslinger H, Olowokure O, Sohal D, Wilson G. et al. Analysis of the fecal metagenome in long-term survivors of pancreas cancer. Cancer. 2023;129(13):1986–1994. doi:10.1002/cncr.34748.
  • 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. 2022;163(4):908–921. doi:10.1053/j.gastro.2022.06.038.
  • Li B, Jiang T, Liu H, Miao Z, Fang D, Zheng L, Zhao J. Andrographolide protects chondrocytes from oxidative stress injury by activation of the Keap1–Nrf2–are signaling pathway. J Cell Physiol. 2018;234(1):561–571. doi:10.1002/jcp.26769.
  • Fang J, Zhu P, Yang Z, Peng X, Zuo Z, Cui H, Ouyang P, Shu G, Chen Z, Huang C. et al. Selenium Ameliorates AFB1−Induced Excess Apoptosis in Chicken Splenocytes Through Death Receptor and Endoplasmic Reticulum Pathways. Biol Trace Elem Res. 2019;187(1):273–280. doi:10.1007/s12011-018-1361-7.
  • 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. 2022;7(2):238–250. doi:10.1038/s41564-021-01030-7.
  • 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. 2020;27(5):823–829.e3. doi:10.1016/j.chom.2020.03.006.
  • Richard ML, Liguori G, Lamas B, Brandi G, da Costa G, Hoffmann TW, Pierluigi Di Simone M, Calabrese C, Poggioli G, Langella P. et al. Mucosa-associated microbiota dysbiosis in colitis associated cancer. Gut Microbes. 2018;9(2):131–142. doi:10.1080/19490976.2017.1379637.
  • Huang X, Hu M, Sun T, Li J, Zhou Y, Yan Y, Xuan B, Wang J, Xiong H, Ji L. et al. Multi-kingdom gut microbiota analyses define bacterial-fungal interplay and microbial markers of pan-cancer immunotherapy across cohorts. Cell Host Microbe. 2023;31(11):1930–1943.e4. doi:10.1016/j.chom.2023.10.005.
  • Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, Gavert N, Stajich JE, Amit G, Gonzalez A. et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185(20):3789–3806.e17. doi:10.1016/j.cell.2022.09.005.
  • Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971–976. doi:10.1126/science.1240537.
  • Daillere R, Vetizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, Duong CPM, Flament C, Lepage P, Roberti MP. et al. Enterococcus hirae and barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45(4):931–943. doi:10.1016/j.immuni.2016.09.009.
  • Zheng DW, Dong X, Pan P, Chen KW, Fan JX, Cheng SX, Zhang XZ. Phage-guided modulation of the gut microbiota of mouse models of colorectal cancer augments their responses to chemotherapy. Nat Biomed Eng. 2019;3(9):717–728. doi:10.1038/s41551-019-0423-2.
  • Cao Z, Liu J. Bacteria and bacterial derivatives as drug carriers for cancer therapy. J Control Release. 2020;326:396–407. doi:10.1016/j.jconrel.2020.07.009.
  • Lam S, Bai X, Shkoporov AN, Park H, Wu X, Lan P, Zuo T. Roles of the gut virome and mycobiome in faecal microbiota transplantation. Lancet Gastroenterol Hepatol. 2022;7(5):472–484. doi:10.1016/S2468-1253(21)00303-4.
  • 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. 2018;9(1):3663. doi:10.1038/s41467-018-06103-6.
  • Zhang F, Zuo T, Yeoh YK, Cheng FWT, Liu Q, Tang W, Cheung KCY, Yang K, Cheung CP, Mo CC. et al. Longitudinal dynamics of gut bacteriome, mycobiome and virome after fecal microbiota transplantation in graft-versus-host disease. Nat Commun. 2021;12(1):65. doi:10.1038/s41467-020-20240-x.
  • Halkjaer SI, Christensen AH, Lo BZS, Browne PD, Gunther S, Hansen LH, Petersen AM. Faecal microbiota transplantation alters gut microbiota in patients with irritable bowel syndrome: results from a randomised, double-blind placebo-controlled study. Gut. 2018;67(12):2107–2115. doi:10.1136/gutjnl-2018-316434.
  • Rasmussen TS, Mentzel CMJ, Kot W, Castro-Mejia JL, Zuffa S, Swann JR, Hansen LH, Vogensen FK, Hansen AK, Nielsen DS. Faecal virome transplantation decreases symptoms of type 2 diabetes and obesity in a murine model. Gut. 2020;69(12).doi:10.1136/gutjnl-2019-320005.
  • Chen Q, Fan Y, Zhang B, Yan C, Chen Z, Wang L, Hu Y, Huang Q, Su J, Ren J. et al. Specific fungi associated with response to capsulized fecal microbiota transplantation in patients with active ulcerative colitis. Front Cell Infect Microbiol. 2022;12:1086885. doi:10.3389/fcimb.2022.1086885.
  • Wang Y, Wiesnoski DH, Helmink BA, Gopalakrishnan V, Choi K, DuPont HL, Jiang ZD, Abu-Sbeih H, Sanchez CA, Chang CC. et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat Med. 2018;24(12):1804–1808. doi:10.1038/s41591-018-0238-9.
  • Youngster I, Russell GH, Pindar C, Ziv-Baran T, Sauk J, Hohmann EL. Oral, capsulized, frozen fecal microbiota transplantation for relapsing Clostridium difficile infection. JAMA. 2014;312(17):1772–1778. doi:10.1001/jama.2014.13875.
  • Angelberger S, Reinisch W, Makristathis A, Lichtenberger C, Dejaco C, Papay P, Novacek G, Trauner M, Loy A, Berry D. Temporal bacterial community dynamics vary among ulcerative colitis patients after fecal microbiota transplantation. Am J Gastroenterol. 2013;108(10):1620–1630. doi:10.1038/ajg.2013.257.
  • Kelly CR, Ihunnah C, Fischer M, Khoruts A, Surawicz C, Afzali A, Aroniadis O, Barto A, Borody T, Giovanelli A. et al. Fecal microbiota transplant for treatment of Clostridium difficile infection in immunocompromised patients. Am J Gastroenterol. 2014;109(7):1065–1071. doi:10.1038/ajg.2014.133.
  • Kazmierczak-Siedlecka K, Daca A, Fic M, van de Wetering T, Folwarski M, Makarewicz W. Therapeutic methods of gut microbiota modification in colorectal cancer management – fecal microbiota transplantation, prebiotics, probiotics, and synbiotics. Gut Microbes. 2020;11(6):1518–1530. doi:10.1080/19490976.2020.1764309.
  • de Menezes APM, Aguiar RPS, Santos JVO, Sarkar C, Islam MT, Braga AL, Hasan MM, da Silva FCC, Sharifi-Rad J, Dey A. et al. Citrinin as a potential anti-cancer therapy: a comprehensive review. Chem Biol Interact. 2023;381:110561. doi:10.1016/j.cbi.2023.110561.
  • de Oliveira Filho JWG, Andrade T, de Lima RMT, Dos Reis AC, Silva DHS, Santos JVO, de Menezes APM, da Mata AMO, Dias ACS, de Alencar M. et al. Citrinin against breast cancer: A cytogenotoxicological study. Phytother Res. 2021;35(1):504–516. doi:10.1002/ptr.6830.
  • Demir G, Klein HO, Mandel-Molinas N, Tuzuner N. Beta glucan induces proliferation and activation of monocytes in peripheral blood of patients with advanced breast cancer. Int Immunopharmacol. 2007;7(1):113–116. doi:10.1016/j.intimp.2006.08.011.
  • Yoon TJ, Kim TJ, Lee H, Shin KS, Yun YP, Moon WK, Kim DW, Lee KH. Anti-tumor metastatic activity of β-glucan purified from mutated Saccharomyces cerevisiae. Int Immunopharmacol. 2008;8(1):36–42. doi:10.1016/j.intimp.2007.10.005.
  • Zhang M, Chun L, Sandoval V, Graor H, Myers J, Nthale J, Rauhe P, Senders Z, Choong K, Huang AY. et al. Systemic administration of β-glucan of 200 kDa modulates melanoma microenvironment and suppresses metastatic cancer. Oncoimmunology. 2018;7(2):e1387347. doi:10.1080/2162402X.2017.1387347.
  • Bae SY, Liao L, Park SH, Kim WK, Shin J, Lee SK. Antitumor activity of asperphenin A, a lipopeptidyl benzophenone from marine-derived Aspergillus sp. Fungus, by inhibiting tubulin polymerization in colon cancer Cells. Mar Drugs. 2020;18(2):110. doi:10.3390/md18020110.
  • Malhao F, Ramos AA, Buttachon S, Dethoup T, Kijjoa A, Rocha E. Cytotoxic and antiproliferative effects of Preussin, a hydroxypyrrolidine derivative from the Marine sponge-associated fungus aspergillus candidus KUFA 0062, in a panel of breast cancer cell lines and using 2D and 3D cultures. Mar Drugs. 2019;17(8):448. doi:10.3390/md17080448.
  • Vidya B, Palaniswamy M, Angayarkanni J, Ayub Nawaz K, Thandeeswaran M, Krishna Chaithanya K, Tekluu B, Muthusamy K, Gopalakrishnan VK. Purification and characterization of beta-galactosidase from newly isolated aspergillus terreus (KUBCF1306) and evaluating its efficacy on breast cancer cell line (MCF-7). Bioorg Chem. 2020;94:103442. doi:10.1016/j.bioorg.2019.103442.
  • 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. 2021;39(9):1202–1213.e6. doi:10.1016/j.ccell.2021.07.002.
  • Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP. et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi:10.1126/science.aan3706.
  • 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. 2021;371(6529):595–602. doi:10.1126/science.abf3363.
  • Zhang M, Kim JA, Huang AY. Optimizing Tumor Microenvironment for Cancer Immunotherapy: beta-Glucan-Based Nanoparticles. Front Immunol. 2018;9:341. doi:10.3389/fimmu.2018.00341.